WO2022255399A1 - G9a inhibitor - Google Patents

G9a inhibitor Download PDF

Info

Publication number
WO2022255399A1
WO2022255399A1 PCT/JP2022/022270 JP2022022270W WO2022255399A1 WO 2022255399 A1 WO2022255399 A1 WO 2022255399A1 JP 2022022270 W JP2022022270 W JP 2022022270W WO 2022255399 A1 WO2022255399 A1 WO 2022255399A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
groups
alkyl group
alkyl
ring
Prior art date
Application number
PCT/JP2022/022270
Other languages
French (fr)
Japanese (ja)
Inventor
稔 吉田
文幸 白井
昭博 伊藤
翔平 高瀬
Original Assignee
国立研究開発法人理化学研究所
学校法人東京薬科大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立研究開発法人理化学研究所, 学校法人東京薬科大学 filed Critical 国立研究開発法人理化学研究所
Publication of WO2022255399A1 publication Critical patent/WO2022255399A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/42Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a derivative having histone methyltransferase G9a inhibitory activity useful as a drug, or a pharmacologically acceptable salt thereof, a pharmaceutical composition containing it, and its medical use.
  • G9a and GLP are the main enzymes that catalyze the mono- and dimethylation (H3K9me1 and H3K9me2) of the 9th lysine residue of histone H3. Also known as EHMT2 and EHMT1 (euchromatin histone-lysine N-methyltransferases 2 and 1).
  • H3K9me2 is an epigenetic mark involved in transcriptional repression. G9a and GLP are involved in epigenetic transcriptional repression through H3K9me2.
  • Non-Patent Documents 1-26 Non-Patent Documents 1-26.
  • Compounds having G9a inhibitory activity include BIX-01294 (Patent Document 1), quinazolines (Patent Document 2), 2-aminoindoles (Patent Document 3), heteroaryls (Patent Document 4), tricyclic compounds (Patent Document 5) and the like are known, but the structure is different from that of the compound of the present invention.
  • the present invention has been made in view of the above-mentioned problems of the prior art, and has excellent G9a inhibitory activity, for example, is useful for treating ⁇ -globin abnormalities such as sickle cell disease.
  • Compounds and pharmacologically acceptable salts thereof also useful for the treatment of proliferative diseases such as cancer, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, malaria, viral infections, myopathy, autism, etc. and G9a inhibitors and pharmaceutical compositions containing them.
  • a further object of the present invention is to provide a method for producing the compound and a pharmacologically acceptable salt thereof, and an intermediate compound useful for the production.
  • An object of the present invention is to provide a compound that has a G9a inhibitory action and excellent pharmacokinetic properties as a pharmaceutical.
  • R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, take the following structure A1a);
  • R 3 is a C 1 -C 6 alkyl group;
  • R 6 and R 7 are each independently a hydrogen atom, a C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group or a halogen atom;
  • the compound according to [2], or a pharmacologically acceptable salt thereof
  • R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, the following A1b), A1c), A1d) and A1e );
  • R 3 is a C 1 -C 6 alkyl group;
  • R 6 and R 7 are each independently a hydrogen atom, a C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group or a halogen atom;
  • the compound according to [2], or a pharmacologically acceptable salt thereof is a pharmacologically acceptable salt thereof.
  • [9] At least selected from a group of diseases consisting of proliferative diseases such as cancer, ⁇ -globin disorders, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, malaria, viral infections, myopathy, and autism
  • the compound (I) according to the present invention or a pharmacologically acceptable salt thereof is useful for proliferative diseases such as cancer, ⁇ -globin abnormalities, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, It is useful as a therapeutic or preventive agent for malaria, viral infections, myopathy, autism, and the like.
  • compound (I) or a pharmacologically acceptable salt thereof according to the present invention can be used in various pathological conditions (e.g., ⁇ -globin disorders such as sickle cell disease, gastric cancer, hepatocellular carcinoma, acute bone marrow disease, etc.).
  • pathological conditions e.g., ⁇ -globin disorders such as sickle cell disease, gastric cancer, hepatocellular carcinoma, acute bone marrow disease, etc.
  • Leukemia such as leukemia and chronic myelogenous leukemia, cervical cancer, neuroblastoma, glioma, pancreatic cancer, colorectal cancer, head and neck squamous cell cancer, breast cancer, lung cancer, ovarian cancer, melanoma, lung Fibrosis such as fibrosis and renal fibrosis, pain, neurodegenerative diseases such as Alzheimer's disease, Prader-Willi syndrome, malaria, foot-and-mouth disease, viral infections such as vesicular stomatitis, cardiomyopathy, myopathy, autism, etc.) It has high utility for treatment, prevention or suppression.
  • compound (I) or a pharmacologically acceptable salt thereof according to the present invention is highly useful for suppressing cancer metastasis and sex reassignment.
  • halogen atom as referred to herein means a fluorine atom, a chlorine atom, a bromine atom, or an iodine atom.
  • hydroxy C 1 -C 6 alkyl group as used herein means the above C 1 -C 6 alkyl group in which at least one hydrogen atom is substituted with a hydroxyl group.
  • Hydroxy C 1 -C 6 alkyl groups such as hydroxymethyl group, 1-hydroxyethyl group, 1-hydroxy-1,1-dimethylmethyl group, 2-hydroxyethyl group, 2-hydroxy-2-methylpropyl group, 3-hydroxypropyl group and the like.
  • hydroxy C 1 -C 3 alkyl group means a hydroxyalkyl group having 1 to 3 carbon atoms.
  • C 1 -C 6 alkoxy group as used herein means a straight or branched chain alkoxy group having 1 to 6 carbon atoms.
  • Examples of C 1 -C 6 alkoxy groups include methoxy, ethoxy, 1-propoxy, isopropoxy, isobutoxy, 1-butoxy, sec-butoxy, tert-butoxy, 1-pentyloxy, 1-hexyloxy group and the like.
  • halo C 1 -C 6 alkoxy group as used herein means a C 1 -C 6 alkoxy group in which at least one hydrogen atom is substituted with the same or different halogen atom.
  • halo C 1 -C 6 alkoxy groups include monofluoromethoxy, difluoromethoxy, trifluoromethoxy, 2-chloroethoxy, 2-fluoroethoxy, 2,2-difluoroethoxy, 1,1- difluoroethoxy group, 1,2-difluoroethoxy group, 1-chloro-2-fluoroethoxy group, 2,2,2-trifluoroethoxy group, 1,1,2,2,2-pentafluoroethoxy group, 2, 2,2-trichloroethoxy group, 3-fluoropropoxy group, 2-fluoropropoxy group, 1-fluoropropoxy group, 3,3-difluoropropoxy group, 2,2-difluoropropoxy group, 1,1-difluoropropoxy group,
  • C 1 -C 6 alkoxycarbonyl group as used herein means a carbonyl group to which a straight or branched chain alkoxy group having 1 to 6 carbon atoms is attached.
  • Examples of C 1 to C 6 alkoxycarbonyl groups include methoxycarbonyl, ethoxycarbonyl, 1-propoxycarbonyl, isopropoxycarbonyl, isobutoxycarbonyl, 1-butoxycarbonyl, sec-butoxycarbonyl, tert- butoxycarbonyl group, 1-pentyloxycarbonyl group, 1-hexyloxycarbonyl group and the like.
  • C 1 -C 6 alkylamino group as used herein means a linear or branched alkyl group in which 1 or 2 hydrogen atoms in the amino group have a total of 1 to 6 carbon atoms. It means a substituted amino group.
  • Examples of C 1 -C 6 alkylamino groups include methylamino group, ethylamino group, 1-propylamino group, isopropylamino group, 1-butylamino group, isobutylamino group, sec-butylamino group and tert-butylamino group.
  • C 1 -C 6 alkylaminocarbonyl group as used herein means a carbonyl group to which a linear or branched alkylamino group having a total of 1 to 6 carbon atoms is bonded.
  • Examples of C 1 -C 6 alkylaminocarbonyl groups include methylaminocarbonyl, ethylaminocarbonyl, 1-propylaminocarbonyl, isopropylaminocarbonyl, 1-butylaminocarbonyl, isobutylaminocarbonyl and sec-butyl.
  • aminocarbonyl group tert-butylaminocarbonyl group, 1-pentylaminocarbonyl group, isopentylaminocarbonyl group, neopentylaminocarbonyl group, 1-methylbutylaminocarbonyl group, 2-methylbutylaminocarbonyl group, 1,2- dimethylpropylaminocarbonyl group, 1-hexylaminocarbonyl group, isohexylaminocarbonyl group, dimethylaminocarbonyl group, diethylaminocarbonyl group, N-ethyl-N-methylaminocarbonyl group, N-ethyl-N-propylaminocarbonyl group, etc. is mentioned.
  • C 1 -C 6 acylamino group as used herein means that 1 or 2 hydrogen atoms of an amino group are substituted with a linear or branched acyl group having 1 to 6 carbon atoms. means an amino group.
  • the C 1 -C 6 acylamino group includes, for example, formylamino group, acetylamino group, 1-propanoylamino group, 1-butanoylamino group, 1-pentanoylamino group, hexanoylamino group and the like.
  • C 1 -C 6 alkylsulfanyl group as used herein means a group in which a straight or branched chain alkyl group having 1 to 6 carbon atoms is bonded to a sulfur atom.
  • Examples of C 1 -C 6 alkylsulfanyl groups include methylsulfanyl group, ethylsulfanyl group, 1-propylsulfanyl group, isopropylsulfanyl group, 1-butylsulfanyl group, isobutylsulfanyl group, sec-butylsulfanyl group and tert-butylsulfanyl group. and the like.
  • halo C 1 -C 6 alkylsulfanyl group as used herein means a C 1 -C 6 alkylsulfanyl group in which at least one hydrogen atom is substituted with the same or different halogen atom.
  • halo C 1 -C 6 alkylsulfanyl group for example, fluoromethylsulfanyl group, difluoromethylsulfanyl group, trifluoromethylsulfanyl group, 2-fluoroethylsulfanyl group, 2-chloroethylsulfanyl group, 2,2-difluoroethylsulfanyl group; group, 1,1-difluoroethylsulfanyl group, 1,2-difluoroethylsulfanyl group, 1-chloro-2-fluoroethylsulfanyl group, 2,2,2-trifluoroethylsulfanyl group, 1,1,2,2 , 2-pentafluoroethylsulfanyl group, 2,2,2-trichloroethylsulfanyl group, 3-fluoropropylsulfanyl group, 2-fluoropropylsulfanyl group, 1-fluor
  • a "5- to 10-membered heteroaryl group” as used herein includes 1 to 4 endocyclic heteroatoms independently selected from the group consisting of nitrogen, oxygen and sulfur atoms. means a to 10-membered monocyclic aromatic heterocyclic group or fused ring aromatic heterocyclic group, wherein the nitrogen and sulfur atoms are optionally oxidized (ie, N ⁇ O, SO or SO 2 ).
  • Examples of 5- to 10-membered heteroaryl groups include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzothiazolyl, benzo triazolyl group, benzisoxazolyl group, benzisothiazolyl group, benzimidazolinyl group, furanyl group, imidazolidinyl group, imidazolyl group, 1H-indazolyl group, imidazolopyridinyl group, indolenyl group, indolizinyl group, 3H-indolyl group, isobenzofuranyl group, isoindazolyl group, isoindolyl group, isoquinolinyl group, isothiazolyl group, isothiazolopyridinyl group, isoxazolyl group, isoxazolopyridiny
  • C 3 -C 10 cycloalkyl group as used herein means a monocyclic or bicyclic saturated alicyclic hydrocarbon group having 3 to 10 carbon atoms, and is a bridged type, spiro type can be Examples of C 3 -C 10 cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, spiroheptyl, spirooctyl and octahydropentalenyl groups. mentioned.
  • the C 3 -C 10 cycloalkyl group may also be fused with a further aromatic hydrocarbon ring group or 5-10 membered heteroaryl group, wherein the aromatic hydrocarbon ring group or 5-10 membered heteroaryl group is
  • the condensed C 3 -C 10 cycloalkyl group includes a dihydroindenyl group, a tetrahydronaphthyl group, and the like.
  • C 3 -C 4 cycloalkyl group means a cycloalkyl group having 3 to 4 carbon atoms.
  • the "3- to 10-membered heterocycloalkyl group” shown herein contains 1 to 4 endocyclic heteroatoms independently selected from the group consisting of nitrogen, oxygen and sulfur atoms. , monocyclic, bicyclic or tricyclic heterocycloalkyl groups having 3-10 membered rings, wherein the nitrogen and sulfur heteroatoms may be optionally oxidized (i.e., N ⁇ O, SO or SO 2 ), the nitrogen atom may be substituted or unsubstituted, may have 1 to 3 carbonyl groups, have 1 double bond in the ring, good too.
  • a 3- to 10-membered heterocycloalkyl group may also be bridged or spiro.
  • a 3-10 membered heterocycloalkyl group may be optionally fused with a further aromatic hydrocarbon ring group or a 5-10 membered heteroaryl group.
  • 3- to 10-membered heterocycloalkyl groups such as aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, azocanyl, dihydropyrrolyl, tetrahydropyridinyl, piperazinyl, morpholinyl, thiomorpholinyl, 1-oxidethiomorpholinyl group, 1,1-dioxidethiomorpholinyl group, oxazepinyl group, thiazepanyl group, 1-oxide-1,4-thiazepanyl group, 1,1-dioxide-1,4-thiazepanyl group , 1,4-diazepanyl group, 1,4-oxazocanyl group, 1,5-oxazocanyl group,
  • the term “bonded to each other to form a ring” refers to removing any one hydrogen atom from each of the two substituents forming the ring, and bonding the hydrogen-free sites to each other. means For example, when the methylene group has two substituents, if the two substituents forming the ring are a methyl group and a 1-hydroxyethyl group, is mentioned.
  • This embodiment relates to a compound represented by the following general formula (I) or a pharmacologically acceptable salt thereof.
  • Preferred compounds of the present embodiment include, for example, the following compounds.
  • compound (I) of the present embodiment or a pharmacologically acceptable salt thereof may exist as a hydrate or solvate. Any hydrate and solvate formed by the derivative represented by the general formula (I) or a salt thereof, including the preferred compounds specifically described above, are included in the scope of the present invention. be. Solvents that can form solvates include methanol, ethanol, isopropyl alcohol, acetone, ethyl acetate, dichloromethane, diisopropyl ether, and the like.
  • Compound (I) of the present embodiment can be a pharmacologically acceptable salt thereof, if necessary.
  • Pharmaceutically acceptable salts refer to salts with pharmaceutically acceptable non-toxic bases or acids (eg, inorganic or organic bases and inorganic or organic acids).
  • a pharmaceutically acceptable salt of compound (I) of the present embodiment is described in J. Chem. Pharm. Sci. 1977, 66, 1-19, and "Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002) or a method according thereto can be done.
  • diastereomeric mixtures can be separated into their respective diastereomers by commonly used methods such as chromatography and crystallization.
  • the respective diastereomers can also be made by using stereochemically uniform starting materials or by synthetic methods employing stereoselective reactions.
  • compound (I) of the present embodiment has geometric isomers such as cis isomers and trans isomers
  • the present invention includes all such geometric isomers.
  • Compound (I) of the present embodiment or a pharmacologically acceptable salt thereof may be a compound labeled with an isotope (eg, 3 H, 14 C, 35 S, etc.) or the like. Such compounds are also included in the present invention.
  • an isotope eg, 3 H, 14 C, 35 S, etc.
  • Compounds (2) to (10) in the formula may form salts, and examples of such salts include those similar to the salts of compound (I).
  • the compound obtained in each step can be used in the next reaction as a reaction solution or after being obtained as a crude product. It can be easily isolated and purified.
  • Synthetic route 1 Compound (I) (the compound represented by the following formula (2)) can be produced, for example, by the method shown in Synthetic Route 1, a method analogous thereto, or a method described in other literatures or a method analogous thereto. can.
  • Step 1-1 Compound (5) can be produced by amidating compound (3) with compound (4).
  • Compound (4) and the like are added in a solvent, and the reaction can be carried out at 0° C. to room temperature, optionally with heating under reflux.
  • a base such as triethylamine or N,N-diisopropylethylamine can be added.
  • Step 1-2 Compound (6) can be produced by removing the protecting group of compound (5).
  • reaction conditions when PG is a tert-butoxycarbonyl group, trifluoro Add an acid such as acetic acid, p-toluenesulfonic acid, hydrogen chloride, hydrobromic acid, sulfuric acid, boron trifluoride diethyl ether complex, boron tribromide, or aluminum chloride, and heat from -78°C to room temperature in some cases. It can be carried out at reflux.
  • PG is a benzyloxycarbonyl group
  • an acid such as acetic acid, trifluoroacetic acid, or 2,2,2-trifluoroethanol can be added as a reaction accelerator.
  • Synthetic route 2 The above compound (2) can be produced, for example, according to the method detailed in Synthetic Route 2 or a method analogous thereto, or the method described in other literatures or a method analogous thereto.
  • Step 2-1 Compound (9) can be produced by amidating compound (8) with compound (7).
  • the same method as in step 1-1 can be used.
  • Step 2-2 Compound (10) can be produced by hydrolyzing compound (9).
  • As the reaction conditions water, methanol, ethanol, 1-propanol, isopropyl alcohol, tetrahydrofuran, 1,4-dioxane, etc., or a water-containing mixed solvent thereof, lithium hydroxide, sodium hydroxide, potassium hydroxide, potassium carbonate, carbonate
  • an alkali metal salt such as sodium or cesium carbonate
  • the reaction can be carried out at 0° C. to reflux under basic conditions.
  • hydrogen chloride or the like in water, tetrahydrofuran, 1,4-dioxane, or a mixed solvent containing water thereof the reaction can be carried out under acidic conditions at 0° C. to heating under reflux.
  • Step 2-3 Compound (2) can be produced by amidating compound (10) with compound (4).
  • compound (4) and the like can be added, and the reaction can be carried out in the same manner as in step 1-1.
  • a protective group when required depending on the type of functional group, it can be carried out by appropriately combining introduction and elimination operations according to a conventional method.
  • introduction and elimination of protective groups see, for example, Theodra W. et al. Green & Peter G. M. Wuts, "Greene's Protective Groups in Organic Synthesis", fourth edition, Wiley-Interscience, 2006.
  • Intermediates used to produce compound (I) of the present embodiment may optionally be isolated and purified by solvent extraction, crystallization, recrystallization, chromatography, and isolation/purification means well known to those skilled in the art. It can be isolated and purified by graphics, preparative high-performance liquid chromatography, or the like.
  • intermediates may be used as crude products in the next reaction without isolation and purification, if necessary.
  • G9a inhibitory action in the present embodiment is the action of inhibiting G9a, which is a major enzyme involved in mono- and dimethylation (H3K9me1 and H3K9me2) at the 9th lysine residue of histone H3.
  • Compound (I) of the present embodiment exhibits potent inhibitory activity, for example, in a G9a inhibitory activity test.
  • compound (I) of the present embodiment or a pharmacologically acceptable salt thereof is useful for proliferative diseases such as cancer, ⁇ -globin abnormalities, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, , malaria, viral infections, myopathy, autism, etc., or as a preventive agent therefor.
  • a medicament containing compound (I) of the present embodiment as an active ingredient can be made into various dosage forms depending on the usage.
  • dosage forms include powders, granules, fine granules, dry syrups, tablets, capsules, injections, liquids, ointments, suppositories, patches, sublingual agents, and the like.
  • These medicaments can be configured as pharmaceutical compositions containing the compound (I) of the present embodiment as an active ingredient and pharmaceutically acceptable additives by a known method depending on the dosage form.
  • Additives contained in the pharmaceutical composition include excipients, disintegrants, binders, lubricants, diluents, buffers, tonicity agents, preservatives, wetting agents, emulsifiers, dispersants, stabilizers, agents, solubilizers, and the like.
  • the pharmaceutical composition can be prepared by appropriately mixing compound (I) of the present embodiment with an additive, or by diluting/dissolving compound (I) with an additive.
  • the pharmaceutical according to this embodiment can be administered systemically or locally, orally or parenterally (nasally, pulmonary, intravenously, intrarectally, subcutaneously, intramuscularly, transdermally, etc.).
  • 1 H-NMR means a spectrum measured by proton nuclear magnetic resonance spectroscopy.
  • CDCl 3 means chloroform-d
  • DMSO-D6 means dimethylsulfoxide-d 6
  • CD 3 OD means methanol-d 4 .
  • MS (ESI + ) and MS (ESI ⁇ ) are electrospray ionization methods
  • MS (FI + ) is field ionization methods
  • MS (FD + ) is field desorption ionization methods
  • MS (EI + ) is electron ionization methods
  • MS (CI + ) means mass spectral data measured by a chemical ionization method.
  • Room temperature means 1 to 30°C.
  • 5-fluoro-3-methyl-1H-indole-2-carboxylic acid (53.8 mg) was dissolved in dimethylformamide (4.0 mL), and 2-amino-4-cyclopropyl-N-(4 -((dimethylamino)methyl)benzyl)butanamide dihydrochloride (101 mg), 1-hydroxybenzotriazole monohydrate (64.0 mg), and N,N-diisopropylethylamine (170 ⁇ L, 0.976 mmol) were added. Ice cold. 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (80.2 mg) was added thereto, and the mixture was stirred overnight at room temperature.
  • Example 1-2 to 1-17 were obtained by the same method as in Example 1-1, the method described in Step 1-3, or a method analogous thereto. rice field.
  • ester (1.22 g) was dissolved in ethanol (20 mL), and 3 mL of 8N aqueous sodium hydroxide solution was added at room temperature. After stirring at room temperature for 15 hours, the mixture was neutralized with 2N hydrochloric acid and extracted twice with ethyl acetate. The combined organic layer was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the solvent was concentrated to give 4-cyclopropyl-2-(3-methyl-1H-indole-2-carboxamide)butanoic acid ( 1.0 g, 90%) was obtained.
  • Example 2-2 to 2-25 are obtained by the same method as in Example 2-1, the method described in Step 2-3, or a method analogous thereto. rice field.
  • Example 3-2 and 3-3 are obtained by the same method as in Example 3-1, the method described in Step 2-3, or a method analogous thereto. rice field.
  • tert-butyl (4-hydroxybenzyl) carbamate 515 mg
  • benzyl 4-hydroxypiperidine-1-carboxylate 595 mg
  • triphenylphosphine 726 mg
  • bis azodicarboxylate (2-Methoxyethyl) (648 mg)
  • Water and ethyl acetate were added to separate the layers, and the aqueous layer was extracted once with ethyl acetate.
  • the combined organic layer was washed twice with water and once with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
  • 3-methyl-1H-indole-2-carboxylic acid (58 mg) was dissolved in dimethylformamide (5.0 mL), and S-2-amino-4-cyclopropyl-N-(4-((dimethyl Amino)methyl)benzyl)butanamide dihydrochloride (100 mg) and N,N-diisopropylethylamine (0.156 mL) were added and ice-cooled.
  • 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate (136 mg) was added thereto, and the mixture was stirred overnight at room temperature.
  • G9a Inhibitory Activity Test The G9a inhibitory activity of the compounds was evaluated by measuring the G9a enzymatic activity by Amplified Luminescence Proximity Homogeneous Assay (ALPHA). First, recombinant human G9a protein (Active Motif, #31410) diluted with Tris buffer (50 mM Tris-HCl [pH 9.0], 50 mM NaCl, 0.01% Tween-20, 1 mM DTT) (final concentration 0 025 nM) and 0.5 ⁇ L of the test compound were added to a 384-well microplate (AlphaPlate-384 Shallow Well, PerkinElmer, #6008359), mixed by vortexing, and allowed to stand at room temperature for 10 minutes.
  • Tris buffer 50 mM Tris-HCl [pH 9.0], 50 mM NaCl, 0.01% Tween-20, 1 mM DTT
  • Tris buffer 50 mM Tris-HCl [pH 9.0],
  • AlphaLISA anti-H3K9me2 acceptor beads PerkinElmer,#AL117M
  • AlphaScreen streptavidin donor beads PerkinElmer,#6760002
  • epigenetic buffer PerkinElmer,AlphaLISA Epigenetics Buffer Kit #AL008C
  • the inhibitory rate of the compound is calculated based on the value of the control without the addition of the compound as 0% and the value of the control without the addition of the enzyme as 100%, and the G9a enzyme activity is inhibited to 50%.
  • concentration of compound (IC 50 value) required to achieve this was calculated. The results are shown below.
  • HUDEP-2 Globin Gene Expression Test The effect of the obtained compounds on the expression level of fetal globin gene in human erythroid cell line HUDEP-2 was evaluated by quantitative PCR.
  • HUDEP-2 cells were grown in Stemline II hematopoietic stem cell growth medium (Sigma) containing 1 ⁇ M dexamethasone (Sigma), 1 ⁇ g/ml doxycycline (Sigma), 50 ng/ml recombinant human SCF (R&D SYSTEMS), and 3 IU/mL epoetin alfa (Toho Yakuhin). cultured.
  • a cell solution was prepared from the cultured cells in a cell culture medium so that the concentration of the cultured cells was 1 ⁇ 10 5 cells/mL. Then, the cell solution was seeded in a 6-well plate (VIOLAMO) at 2 mL/well, and the test compound (DMSO solution) was added at 2 ⁇ L/well (DMSO final concentration 0.1%), 37° C., 5% CO 2 conditions. was cultured for 4 days. A DMSO solution was added at 2 ⁇ L/well as a control. The cultured cells were collected by centrifugation at 7500 rpm, 4° C. for 5 minutes, and RNA was extracted using Tissue Total RNA Mini Kit (Chiyoda Science).
  • cDNA was synthesized by reverse transcription at 37°C for 15 minutes and then at 50°C for 5 minutes.
  • cDNA was synthesized by reverse transcription at 37°C for 15 minutes and then at 50°C for 5 minutes.
  • cDNA was mixed with THUNDERBIRD SYBR qPCR (TOYOBO), and the CFX Connect Real-Time PCR Detection System (BIORAD) was used to detect the fetal ⁇ -globin gene, the adult ⁇ -globin gene, and the GAPDH gene as a reference. was measured.
  • BIORAD Real-Time PCR Detection System
  • P-gp P-glycoprotein
  • MDR1-MDCKII cells and MDCKII cells expressing P-glycoprotein (MDR1) adding the obtained compound to the apical side or the basolateral side, measuring the compound concentration on each side after incubation for 2 hours, Apparent membrane permeability coefficient (Papp) and flux ratio (PappBtA/PappAtB) of each cell were calculated to evaluate drug transport ability by P-glycoprotein.
  • the compounds according to the present invention are useful for proliferative diseases such as cancer, ⁇ -globin abnormalities, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, malaria, viral infections, myopathy, and autism due to G9a enzyme inhibitory activity. It is useful as a therapeutic or preventive agent for diseases and the like.

Abstract

The present invention pertains to a compound represented by general formula (I) or a pharmacologically acceptable salt thereof and a pharmaceutical use thereof, for example, a pharmaceutical composition that is to be used for preventing or treating at least one disease selected from the group of diseases consisting of proliferative diseases such as cancer, β-globin disorders, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, malaria, viral infections, myopathy and autism.

Description

G9a阻害剤G9a inhibitor
 本発明は、医薬品として有用なヒストンメチルトランスフェラーゼG9a阻害作用を有する誘導体、またはその薬理学的に許容される塩、それを含有する医薬組成物およびその医薬用途に関する。 The present invention relates to a derivative having histone methyltransferase G9a inhibitory activity useful as a drug, or a pharmacologically acceptable salt thereof, a pharmaceutical composition containing it, and its medical use.
 ヒストンのリジンメチル化は、S-アデノシルメチオニン(SAM)をメチル基ドナーとしてヒストンのリジン残基のεアミノ基にメチル基を付加する生化学反応である。ヒストンのリジンメチル化は、転写調節において重要な役割を担っており、細胞増殖や細胞分化を含む様々な生物学的プロセスにおいて重要である。ヒストンのリジンメチル化反応を触媒する酵素として、ヒストンメチル基転移酵素(リジンメチル基転移酵素とも言われている)が知られている(非特許文献1)。 Lysine methylation of histones is a biochemical reaction in which a methyl group is added to the ε-amino group of histone lysine residues using S-adenosylmethionine (SAM) as a methyl group donor. Histone lysine methylation plays an important role in transcriptional regulation and is important in a variety of biological processes, including cell proliferation and cell differentiation. Histone methyltransferase (also referred to as lysine methyltransferase) is known as an enzyme that catalyzes the lysine methylation reaction of histones (Non-Patent Document 1).
 G9aおよびGLP(G9a like protein)は、ヒストンH3の9番目のリジン残基のモノおよびジメチル化(H3K9me1およびH3K9me2)を触媒する主要酵素である。EHMT2およびEHMT1(ユークロマチンのヒストン-リジンN-メチルトランスフェラーゼ2および1)としても知られている。 G9a and GLP (G9a-like protein) are the main enzymes that catalyze the mono- and dimethylation (H3K9me1 and H3K9me2) of the 9th lysine residue of histone H3. Also known as EHMT2 and EHMT1 (euchromatin histone-lysine N-methyltransferases 2 and 1).
 H3K9me2は転写抑制に関わるエピジェネティックマークである。G9aおよびGLPはH3K9me2することにより、エピジェネティックな転写抑制に関与する。 H3K9me2 is an epigenetic mark involved in transcriptional repression. G9a and GLP are involved in epigenetic transcriptional repression through H3K9me2.
 よって、G9aの阻害は、H3K9me2による転写抑制を介した生物学的過程である細胞増殖や細胞分化などの制御に有用であると考えられる。G9a阻害剤が効果を示す可能性がある疾患としては、鎌状赤血球症などのβ-グロビン異常症、胃がん、肝細胞がん、急性骨髄性白血病や慢性骨髄性白血病などの白血病、子宮頸がん、神経芽腫、神経膠腫、膵がん、大腸がん、頭頸部扁平上皮がん、乳がん、肺がん、卵巣がん、メラノーマ、肺線維症や腎線維症などの線維症、疼痛、アルツハイマー病やFriedreich運動失調症などの神経変性疾患、プラダー・ウィリー症候群、マラリア、口蹄疫、水疱性口内炎などのウイルス感染症、心筋症、急性心筋梗塞症、ミオパチー、自閉症などが挙げられる。さらに、G9aの阻害は、がんの転移抑制にも有効である可能性が示唆されている。加えて、G9aの阻害は性転換にも有効であることが示されている(非特許文献1~26)。 Therefore, G9a inhibition is thought to be useful in regulating biological processes such as cell proliferation and cell differentiation mediated by transcriptional repression by H3K9me2. Diseases for which G9a inhibitors may be effective include β-globin disorders such as sickle cell disease, gastric cancer, hepatocellular carcinoma, leukemia such as acute myelogenous leukemia and chronic myelogenous leukemia, and cervical cancer. cancer, neuroblastoma, glioma, pancreatic cancer, colorectal cancer, head and neck squamous cell carcinoma, breast cancer, lung cancer, ovarian cancer, melanoma, fibrosis such as pulmonary fibrosis and renal fibrosis, pain, Alzheimer's disease neurodegenerative diseases such as disease and Friedreich's ataxia, viral infections such as Prader-Willi syndrome, malaria, foot-and-mouth disease, and bullous stomatitis, cardiomyopathy, acute myocardial infarction, myopathy, and autism. Furthermore, it has been suggested that inhibition of G9a may also be effective in suppressing cancer metastasis. In addition, inhibition of G9a has been shown to be effective in sex reversal (Non-Patent Documents 1-26).
 G9a阻害活性を有する化合物としては、BIX-01294(特許文献1)、キナゾリン類(特許文献2)、2-アミノインドール類(特許文献3)、ヘテロアリール類(特許文献4)、三環性化合物(特許文献5)などが知られているが、本発明化合物とは構造が異なる。 Compounds having G9a inhibitory activity include BIX-01294 (Patent Document 1), quinazolines (Patent Document 2), 2-aminoindoles (Patent Document 3), heteroaryls (Patent Document 4), tricyclic compounds (Patent Document 5) and the like are known, but the structure is different from that of the compound of the present invention.
国際公開2012/023285号WO2012/023285 国際公開2013/140148号WO2013/140148 US2015/0274660US2015/0274660 特表2019-513778号公報Japanese Patent Application Publication No. 2019-513778 特表2019-511472号公報Japanese Patent Publication No. 2019-511472
 本発明は、上記従来技術の有する課題に鑑みてなされたものであり、優れたG9a阻害活性を有し、例えば、鎌状赤血球症などのβ-グロビン異常症の治療に有用であり、その他がんなどの増殖性疾患、線維症、疼痛、神経変性疾患、プラダー・ウィリー症候群、マラリア、ウイルス感染症、ミオパチー、自閉症などの治療にも有用な化合物およびその薬理学的に許容される塩、並びに、それらを含有するG9a阻害剤および医薬組成物を提供することを目的とする。更には、前記化合物およびその薬理学的に許容される塩の製造方法、その製造に有用な中間体化合物を提供することを目的とする。 The present invention has been made in view of the above-mentioned problems of the prior art, and has excellent G9a inhibitory activity, for example, is useful for treating β-globin abnormalities such as sickle cell disease. Compounds and pharmacologically acceptable salts thereof also useful for the treatment of proliferative diseases such as cancer, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, malaria, viral infections, myopathy, autism, etc. and G9a inhibitors and pharmaceutical compositions containing them. A further object of the present invention is to provide a method for producing the compound and a pharmacologically acceptable salt thereof, and an intermediate compound useful for the production.
 現在、上述した種々の病態に対する予防および治療薬として、優れたG9a阻害作用を有し、十分に満足できる医薬品となり得る化合物は見出されていない。 At present, no compound has been found that has an excellent G9a inhibitory effect and can be a fully satisfactory pharmaceutical agent as a prophylactic and therapeutic agent for the various pathologies described above.
 本発明の目的は、G9a阻害作用を有し、医薬として体内動態においても優れた性質を有する化合物を提供することにある。 An object of the present invention is to provide a compound that has a G9a inhibitory action and excellent pharmacokinetic properties as a pharmaceutical.
 本発明者らは、酵素活性の最適化のみならず、G9a蛋白質とのX線複合体構造解析を行ってG9a阻害活性に必要な構造を特定するとともに、医薬としての体内動態に影響する各種パラメーターの最適化を鋭意検討した結果、下記一般式(I)で表される化合物(以下、化合物(I)という場合もある)、またはその薬理学的に許容される塩がG9a阻害作用を有し、医薬として十分に満足できる体内動態を有するために必要な、P-gp親和性の回避や高水溶性を維持することを見出し、本発明を完成した。
 即ち、本発明は以下のとおりである。
The present inventors have not only optimized the enzymatic activity, but also performed X-ray complex structural analysis with the G9a protein to identify the structure required for G9a inhibitory activity, and various parameters that affect pharmacokinetics as a drug. As a result of intensive studies on the optimization of the compound represented by the following general formula (I) (hereinafter sometimes referred to as compound (I)), or a pharmacologically acceptable salt thereof has a G9a inhibitory action , found that it avoids P-gp affinity and maintains high water solubility, which are necessary for having sufficiently satisfactory pharmacokinetics as a drug, and completed the present invention.
That is, the present invention is as follows.
[1]一般式(I):
Figure JPOXMLDOC01-appb-C000012
[式(I)中、RとRとは互いに結合して環を形成して、RとRが結合するピロール環とともに、以下のA1)の構造をとり、*が式(I)中の-CO-との結合位置を示し;
Figure JPOXMLDOC01-appb-C000013
3はC~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基またはヒドロキシC~Cアルキル基(該C~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基およびヒドロキシC~Cアルキル基はC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基およびヒドロキシC~Cアルキル基からなる群より選択される置換基で一つまたは複数置換されてもよく、置換基同士で互いに結合して環を形成してもよい)であり;
Xは炭素原子または4位、5位、6位、7位のいずれかの窒素原子を示し、それぞれインドールまたはアザインドール環類であり;
およびRはそれぞれ独立して、水素原子、ハロゲン原子、C~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基またはヒドロキシC~Cアルキル基(該C~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基およびヒドロキシC~Cアルキル基はC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基およびヒドロキシC~Cアルキル基からなる群より選択される置換基で一つまたは複数置換されてもよく、置換基同士で互いに結合して環を形成してもよい)であり;
は以下のB1)であり、*が式(I)中の-N-との結合位置を示し;
Figure JPOXMLDOC01-appb-C000014
およびRはそれぞれ独立して水素原子、C~Cアルキル基またはヒドロキシC~Cアルキル基であり;
およびRは互いに結合して環を形成してもよく;
およびRはRingAと結合して環を形成してもよく;
mは0または1であり;
RingAは、それぞれR10、R11およびR12で置換されてもよい、芳香族炭化水素環基、C~C10シクロアルキル基、5~10員ヘテロアリール基または3~10員ヘテロシクロアルキル基であり;
10およびR11はそれぞれ独立して水素原子、ハロゲン原子、水酸基、アミノ基、シアノ基、カルバモイル基(-CONH)、C~Cアルキル基、ハロC~Cアルキル基、ヒドロキシC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアルキルスルファニル基、ハロC~Cアルキルスルファニル基、C~Cアシル基、C~Cアルコキシカルボニル基、C~Cアルキルアミノカルボニル基またはC~Cアシルアミノ基であり;
12は-(CR1314-V-(CR1516-(W)-Qであり;
Vは結合、-O-または-NR17-であり;
Wは-NR18-であり;
13、R14、R15、R16、R17およびR18はそれぞれ独立して水素原子またはC~Cアルキル基であり;
qおよびrはそれぞれ独立して0~6の整数であり;
nは0または1であり;
Qは水素原子、水酸基、C~Cアルキル基、C~C10シクロアルキル基、5~10員ヘテロアリール基または3~10員ヘテロシクロアルキル基(該C~Cアルキルアミノ基、C~C10シクロアルキル基、5~10員ヘテロアリール基および3~10員ヘテロシクロアルキル基は、ハロゲン原子、C~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアルコキシカルボニル基、C~Cアルキルアミノカルボニル基およびヒドロキシC~Cアルキル基からなる群より選択される置換基で一つまたは複数置換されてもよい)であり;
10、R11およびR12は互いに結合して環を形成してもよい]
で表される化合物、またはその薬理学的に許容される塩。
[1] General formula (I):
Figure JPOXMLDOC01-appb-C000012
[In the formula (I), R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, the structure of A1) below is taken, and * is the formula (I ) indicates the bonding position with -CO- in;
Figure JPOXMLDOC01-appb-C000013
R 3 is C 1 -C 6 alkyl group, C 2 -C 6 alkenyl group, halo C 1 -C 6 alkyl group, C 1 -C 6 alkoxy group, C 1 -C 6 alkylamino group, C 1 -C 6 acyl group, C 1 -C 6 alkoxycarbonyl group, C 3 -C 10 cycloalkyl group or hydroxy C 1 -C 6 alkyl group (the C 1 -C 6 alkyl group, C 2 -C 6 alkenyl group, halo C 1 -C6 alkyl groups, C1 - C6 alkoxy groups, C1 - C6 alkylamino groups, C1 - C6 acyl groups, C1 - C6 alkoxycarbonyl groups, C3 - C10 cycloalkyl groups and hydroxy C 1 -C 6 alkyl group is a substituent selected from the group consisting of C 1 -C 6 alkyl group, C 1 -C 6 alkoxy group, C 1 -C 6 alkylamino group and hydroxy C 1 -C 6 alkyl group may be substituted one or more times, and the substituents may be bonded to each other to form a ring);
X represents a carbon atom or a nitrogen atom at any of the 4-, 5-, 6-, and 7-positions, and is an indole or azaindole ring, respectively;
R 6 and R 7 are each independently a hydrogen atom, a halogen atom, a C 1 -C 6 alkyl group, a C 2 -C 6 alkenyl group, a halo C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group, C 1 -C 6 alkylamino group, C 1 -C 6 acyl group, C 1 -C 6 alkoxycarbonyl group, C 3 -C 10 cycloalkyl group or hydroxy C 1 -C 6 alkyl group (the C 1 -C 6 Alkyl Group, C 2 -C 6 Alkenyl Group, Halo C 1 -C 6 Alkyl Group, C 1 -C 6 Alkoxy Group, C 1 -C 6 Alkylamino Group, C 1 -C 6 Acyl Group, C 1 -C 6 Alkoxycarbonyl groups, C 3 -C 10 cycloalkyl groups and hydroxy C 1 -C 6 alkyl groups are C 1 -C 6 alkyl groups, C 1 -C 6 alkoxy groups, C 1 -C 6 alkylamino groups and hydroxy C 1 may be substituted with one or more substituents selected from the group consisting of ~ C6 alkyl groups, and the substituents may be bonded to each other to form a ring);
R 4 is B1) below, * indicates the bonding position with -N- in formula (I);
Figure JPOXMLDOC01-appb-C000014
R 8 and R 9 are each independently a hydrogen atom, a C 1 -C 6 alkyl group or a hydroxy C 1 -C 6 alkyl group;
R 8 and R 9 may be joined together to form a ring;
R 8 and R 9 may combine with RingA to form a ring;
m is 0 or 1;
RingA is an aromatic hydrocarbon ring group, a C3 - C10 cycloalkyl group, a 5- to 10-membered heteroaryl group or a 3- to 10-membered heterocycloalkyl group, each of which may be substituted with R 10 , R 11 and R 12 is a group;
R 10 and R 11 each independently represent a hydrogen atom, a halogen atom, a hydroxyl group, an amino group, a cyano group, a carbamoyl group (—CONH 2 ), a C 1 -C 6 alkyl group, a halo C 1 -C 6 alkyl group, a hydroxy C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylamino, C 1 -C 6 alkylsulfanyl, halo C 1 -C 6 alkylsulfanyl, C 1 -C 6 acyl a C 1 -C 6 alkoxycarbonyl group, a C 1 -C 6 alkylaminocarbonyl group or a C 1 -C 6 acylamino group;
R 12 is -(CR 13 R 14 ) r -V-(CR 15 R 16 ) q -(W) n -Q;
V is a bond, -O- or -NR 17 -;
W is -NR 18 -;
R 13 , R 14 , R 15 , R 16 , R 17 and R 18 are each independently a hydrogen atom or a C 1 -C 6 alkyl group;
q and r are each independently an integer from 0 to 6;
n is 0 or 1;
Q is a hydrogen atom, a hydroxyl group, a C 1 to C 6 alkyl group, a C 3 to C 10 cycloalkyl group, a 5 to 10 membered heteroaryl group or a 3 to 10 membered heterocycloalkyl group (the C 1 to C 6 alkylamino group , C 3 to C 10 cycloalkyl groups, 5 to 10 membered heteroaryl groups and 3 to 10 membered heterocycloalkyl groups are halogen atoms, C 1 to C 6 alkyl groups, C 1 to C 6 alkoxy groups, C 1 to substituted with one or more substituents selected from the group consisting of C 6 alkylamino group, C 1 -C 6 alkoxycarbonyl group, C 1 -C 6 alkylaminocarbonyl group and hydroxy C 1 -C 6 alkyl group may be);
R 10 , R 11 and R 12 may combine with each other to form a ring]
A compound represented by or a pharmacologically acceptable salt thereof.
[2]式(I)中、RとRとが互いに結合して環を形成して、RとRが結合するピロール環とともに、以下のA1)の構造をとり;
Figure JPOXMLDOC01-appb-C000015
はC~Cアルキル基またはC~C10シクロアルキル基であり;
Xは炭素原子または4位、5位、6位、7位のいずれかの窒素原子を示し、それぞれインドールまたはアザインドール環類であり;
およびRはそれぞれ独立して、水素原子、C~Cアルキル基、C~Cアルコキシ基またはハロゲン原子である、
[1]に記載の化合物、またはその薬理学的に許容される塩。
[2] In formula (I), R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, take the following structure A1);
Figure JPOXMLDOC01-appb-C000015
R 3 is a C 1 -C 6 alkyl group or a C 3 -C 10 cycloalkyl group;
X represents a carbon atom or a nitrogen atom at any of the 4-, 5-, 6-, and 7-positions, and is an indole or azaindole ring, respectively;
R 6 and R 7 are each independently a hydrogen atom, a C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group or a halogen atom;
The compound according to [1], or a pharmacologically acceptable salt thereof.
[3]式(I)中、RとRとが互いに結合して環を形成して、RとRが結合するピロール環とともに、以下のA1a)の構造をとり;
Figure JPOXMLDOC01-appb-C000016
はC~Cアルキル基であり;
およびRはそれぞれ独立して、水素原子、C~Cアルキル基、C~Cアルコキシ基またはハロゲン原子である、
[2]に記載の化合物、またはその薬理学的に許容される塩。
[3] In formula (I), R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, take the following structure A1a);
Figure JPOXMLDOC01-appb-C000016
R 3 is a C 1 -C 6 alkyl group;
R 6 and R 7 are each independently a hydrogen atom, a C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group or a halogen atom;
The compound according to [2], or a pharmacologically acceptable salt thereof.
[4]式(I)中、RとRとが互いに結合して環を形成して、RとRが結合するピロール環とともに、以下のA1b)、A1c)、A1d)、A1e)のいずれかの構造をとり;
Figure JPOXMLDOC01-appb-C000017
はC~Cアルキル基であり;
およびRはそれぞれ独立して、水素原子、C~Cアルキル基、C~Cアルコキシ基またはハロゲン原子である、
[2]に記載の化合物、またはその薬理学的に許容される塩。
[4] In formula (I), R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, the following A1b), A1c), A1d) and A1e );
Figure JPOXMLDOC01-appb-C000017
R 3 is a C 1 -C 6 alkyl group;
R 6 and R 7 are each independently a hydrogen atom, a C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group or a halogen atom;
The compound according to [2], or a pharmacologically acceptable salt thereof.
[5]以下、
Figure JPOXMLDOC01-appb-T000018
Figure JPOXMLDOC01-appb-T000019
Figure JPOXMLDOC01-appb-I000020
Figure JPOXMLDOC01-appb-T000021
Figure JPOXMLDOC01-appb-I000022
から選択される化合物、またはその薬理学的に許容される塩。
[5] Below,
Figure JPOXMLDOC01-appb-T000018
Figure JPOXMLDOC01-appb-T000019
Figure JPOXMLDOC01-appb-I000020
Figure JPOXMLDOC01-appb-T000021
Figure JPOXMLDOC01-appb-I000022
or a pharmaceutically acceptable salt thereof.
[6][1]から[5]のいずれかに記載の化合物またはその薬理学的に許容される塩を有効成分として含有するG9a酵素阻害性組成物。
[7][1]から[5]のいずれかに記載の化合物またはその薬理学的に許容される塩を有効成分として含有する医薬組成物。
[8]がんなどの増殖性疾患、β-グロビン異常症、線維症、疼痛、神経変性疾患、プラダー・ウィリー症候群、マラリア、ウイルス感染症、ミオパチー、自閉症からなる疾病群から選ばれる少なくとも一種の疾病の予防または治療のための医薬を製造するための[1]から[5]のいずれかに記載の化合物またはその薬理学的に許容される塩の使用。
[9]がんなどの増殖性疾患、β-グロビン異常症、線維症、疼痛、神経変性疾患、プラダー・ウィリー症候群、マラリア、ウイルス感染症、ミオパチー、自閉症からなる疾病群から選ばれる少なくとも一種の疾病の予防または治療に用いられる、[1]から[5]のいずれかに記載の化合物またはその薬理学的に許容される塩および薬学的に許容される担体を含有する医薬組成物。
[6] A G9a enzyme inhibitory composition containing the compound of any one of [1] to [5] or a pharmacologically acceptable salt thereof as an active ingredient.
[7] A pharmaceutical composition containing the compound of any one of [1] to [5] or a pharmacologically acceptable salt thereof as an active ingredient.
[8] At least selected from a group of diseases consisting of proliferative diseases such as cancer, β-globin disorders, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, malaria, viral infections, myopathy, and autism Use of the compound or a pharmacologically acceptable salt thereof according to any one of [1] to [5] for manufacturing a medicament for the prevention or treatment of a kind of disease.
[9] At least selected from a group of diseases consisting of proliferative diseases such as cancer, β-globin disorders, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, malaria, viral infections, myopathy, and autism A pharmaceutical composition containing the compound or a pharmacologically acceptable salt thereof according to any one of [1] to [5] and a pharmaceutically acceptable carrier, which is used for the prevention or treatment of a kind of disease.
 化合物(I)、またはその薬理学的に許容される塩は、例えば、強力なG9a酵素阻害活性を示した。 Compound (I) or a pharmacologically acceptable salt thereof, for example, exhibited potent G9a enzyme inhibitory activity.
 よって、本発明に係る化合物(I)またはその薬理学的に許容される塩は、がんなどの増殖性疾患、β-グロビン異常症、線維症、疼痛、神経変性疾患、プラダー・ウィリー症候群、マラリア、ウイルス感染症、ミオパチー、自閉症等の治療薬またはその予防薬として有用である。 Therefore, the compound (I) according to the present invention or a pharmacologically acceptable salt thereof is useful for proliferative diseases such as cancer, β-globin abnormalities, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, It is useful as a therapeutic or preventive agent for malaria, viral infections, myopathy, autism, and the like.
 さらにまた、本発明に係る化合物(I)またはその薬理学的に許容される塩は、種々の病態(例えば、鎌状赤血球症などのβ-グロビン異常症、胃がん、肝細胞がん、急性骨髄性白血病や慢性骨髄性白血病などの白血病、子宮頸がん、神経芽腫、神経膠腫、膵がん、大腸がん、頭頸部扁平上皮がん、乳がん、肺がん、卵巣がん、メラノーマ、肺線維症や腎線維症などの線維症、疼痛、アルツハイマー病などの神経変性疾患、プラダー・ウィリー症候群、マラリア、口蹄疫、水疱性口内炎などのウイルス感染症、心筋症、ミオパチー、自閉症など)の治療、予防または抑制に対して高い有用性を有するものである。また、本発明に係る化合物(I)またはその薬理学的に許容される塩は、がんの転移抑制および性転換に関して高い有用性を有するものである。 Furthermore, compound (I) or a pharmacologically acceptable salt thereof according to the present invention can be used in various pathological conditions (e.g., β-globin disorders such as sickle cell disease, gastric cancer, hepatocellular carcinoma, acute bone marrow disease, etc.). Leukemia such as leukemia and chronic myelogenous leukemia, cervical cancer, neuroblastoma, glioma, pancreatic cancer, colorectal cancer, head and neck squamous cell cancer, breast cancer, lung cancer, ovarian cancer, melanoma, lung Fibrosis such as fibrosis and renal fibrosis, pain, neurodegenerative diseases such as Alzheimer's disease, Prader-Willi syndrome, malaria, foot-and-mouth disease, viral infections such as vesicular stomatitis, cardiomyopathy, myopathy, autism, etc.) It has high utility for treatment, prevention or suppression. In addition, compound (I) or a pharmacologically acceptable salt thereof according to the present invention is highly useful for suppressing cancer metastasis and sex reassignment.
 本明細書における用語について説明する。
 本明細書に示される「ハロゲン原子」とは、フッ素原子、塩素原子、臭素原子、またはヨウ素原子を意味する。
Terms used in this specification will be explained.
A "halogen atom" as referred to herein means a fluorine atom, a chlorine atom, a bromine atom, or an iodine atom.
 本明細書に示される「C~Cアルキル基」とは、炭素数1~6個の直鎖または分枝鎖の飽和炭化水素基を意味する。C~Cアルキル基として、例えば、メチル基、エチル基、1-プロピル基、イソプロピル基、1-ブチル基、イソブチル基、sec-ブチル基、tert-ブチル基、1-ペンチル基、イソペンチル基、ネオペンチル基、1-メチルブチル基、2-メチルブチル基、1,2-ジメチルプロピル基、1-ヘキシル基、イソヘキシル基等が挙げられる。また、「C~Cアルキル基」とは、炭素数1~3個の直鎖または分枝鎖の飽和炭化水素基を意味する。 The “C 1 -C 6 alkyl group” as used herein means a linear or branched saturated hydrocarbon group having 1 to 6 carbon atoms. Examples of C 1 -C 6 alkyl groups include methyl group, ethyl group, 1-propyl group, isopropyl group, 1-butyl group, isobutyl group, sec-butyl group, tert-butyl group, 1-pentyl group and isopentyl group. , neopentyl group, 1-methylbutyl group, 2-methylbutyl group, 1,2-dimethylpropyl group, 1-hexyl group, isohexyl group and the like. In addition, the “C 1 -C 3 alkyl group” means a linear or branched saturated hydrocarbon group having 1 to 3 carbon atoms.
 本明細書に示される「C~Cアルケニル基」とは、少なくとも1個の二重結合を有する、炭素数2~6個の直鎖または分岐鎖の不飽和炭化水素基を意味する。C~Cアルケニル基として、例えば、ビニル基、2-プロペニル基、1-プロペニル基、1-ブテン-1-イル基、1-ブテン-2-イル基、1-ブテン-3-イル基、2-ブテン-1-イル基、2-ブテン-2-イル基、1-ペンテン-1-イル基、1-ペンテン-2-イル基、1-ペンテン-3-イル基、2-ペンテン-1-イル基、2-ペンテン-2-イル基、2-ペンテン-3-イル基、1-ヘキセン-1-イル基、1-ヘキセン-2-イル基、1-ヘキセン-3-イル基、2-メチル-1-プロペン-1-イル基等が挙げられる。 As used herein, a "C 2 -C 6 alkenyl group" means a linear or branched unsaturated hydrocarbon group having 2 to 6 carbon atoms and having at least one double bond. C 2 -C 6 alkenyl groups such as vinyl group, 2-propenyl group, 1-propenyl group, 1-buten-1-yl group, 1-buten-2-yl group and 1-buten-3-yl group , 2-buten-1-yl group, 2-buten-2-yl group, 1-penten-1-yl group, 1-penten-2-yl group, 1-penten-3-yl group, 2-pentene- 1-yl group, 2-penten-2-yl group, 2-penten-3-yl group, 1-hexen-1-yl group, 1-hexen-2-yl group, 1-hexen-3-yl group, 2-methyl-1-propen-1-yl group and the like.
 本明細書に示される「C~Cアシル基」とは、炭素原子1~6個を有する直鎖または分岐鎖の脂肪族カルボン酸から誘導されるアシル基を意味する。例えば、ホルミル基、アセチル基、プロパノイル基、1-ブタノイル基、1-ペンタノイル基、1-ヘキサノイル基等が挙げられる。 As used herein, a "C 1 -C 6 acyl group" means an acyl group derived from a straight or branched chain aliphatic carboxylic acid having 1 to 6 carbon atoms. Examples include formyl group, acetyl group, propanoyl group, 1-butanoyl group, 1-pentanoyl group, 1-hexanoyl group and the like.
 本明細書に示される「ハロC~Cアルキル基」とは、少なくとも1つの水素原子が同種または異種のハロゲン原子で置換されたC~Cアルキル基を意味する。ハロC~Cアルキル基として、例えば、フルオロメチル基、ジフルオロメチル基、トリフルオロメチル基、2-フルオロエチル基、2-クロロエチル基、2,2-ジフルオロエチル基、1,1-ジフルオロエチル基、1,2-ジフルオロエチル基、1-クロロ-2-フルオロエチル基、2,2,2-トリフルオロエチル基、1,1,2,2,2-ペンタフルオロエチル基、2,2,2-トリクロロエチル基、3-フルオロプロピル基、2-フルオロプロピル基、1-フルオロプロピル基、3,3-ジフルオロプロピル基、2,2-ジフルオロプロピル基、1,1-ジフルオロプロピル基、4-フルオロブチル基、5-フルオロペンチル基、6-フルオロヘキシル基等が挙げられる。 The “halo C 1 -C 6 alkyl group” as used herein means a C 1 -C 6 alkyl group in which at least one hydrogen atom is substituted with the same or different halogen atom. Examples of halo C 1 -C 6 alkyl groups include fluoromethyl, difluoromethyl, trifluoromethyl, 2-fluoroethyl, 2-chloroethyl, 2,2-difluoroethyl and 1,1-difluoroethyl. group, 1,2-difluoroethyl group, 1-chloro-2-fluoroethyl group, 2,2,2-trifluoroethyl group, 1,1,2,2,2-pentafluoroethyl group, 2,2, 2-trichloroethyl group, 3-fluoropropyl group, 2-fluoropropyl group, 1-fluoropropyl group, 3,3-difluoropropyl group, 2,2-difluoropropyl group, 1,1-difluoropropyl group, 4- fluorobutyl group, 5-fluoropentyl group, 6-fluorohexyl group and the like.
 本明細書に示される「ヒドロキシC~Cアルキル基」とは、少なくとも1つの水素原子が水酸基で置換された上記C~Cアルキル基を意味する。ヒドロキシC~Cアルキル基として、例えば、ヒドロキシメチル基、1-ヒドロキシエチル基、1-ヒドロキシ-1,1-ジメチルメチル基、2-ヒドロキシエチル基、2-ヒドロキシ-2-メチルプロピル基、3-ヒドロキシプロピル基等が挙げられる。また、「ヒドロキシC~Cアルキル基」とは、炭素数1~3個のヒドロキシアルキル基を意味する。 The “hydroxy C 1 -C 6 alkyl group” as used herein means the above C 1 -C 6 alkyl group in which at least one hydrogen atom is substituted with a hydroxyl group. Hydroxy C 1 -C 6 alkyl groups such as hydroxymethyl group, 1-hydroxyethyl group, 1-hydroxy-1,1-dimethylmethyl group, 2-hydroxyethyl group, 2-hydroxy-2-methylpropyl group, 3-hydroxypropyl group and the like. Further, "hydroxy C 1 -C 3 alkyl group" means a hydroxyalkyl group having 1 to 3 carbon atoms.
 本明細書に示される「C~Cアルコキシ基」とは、炭素数1~6個の直鎖または分枝鎖のアルコキシ基を意味する。C~Cアルコキシ基として、例えば、メトキシ基、エトキシ基、1-プロポキシ基、イソプロポキシ基、イソブトキシ基、1-ブトキシ基、sec-ブトキシ基、tert-ブトキシ基、1-ペンチルオキシ基、1-ヘキシルオキシ基等が挙げられる。 The “C 1 -C 6 alkoxy group” as used herein means a straight or branched chain alkoxy group having 1 to 6 carbon atoms. Examples of C 1 -C 6 alkoxy groups include methoxy, ethoxy, 1-propoxy, isopropoxy, isobutoxy, 1-butoxy, sec-butoxy, tert-butoxy, 1-pentyloxy, 1-hexyloxy group and the like.
 本明細書に示される「ハロC~Cアルコキシ基」とは、少なくとも1つの水素原子が同種または異種のハロゲン原子で置換されたC~Cアルコキシ基を意味する。ハロC~Cアルコキシ基として、例えば、モノフルオロメトキシ基、ジフルオロメトキシ基、トリフルオロメトキシ基、2-クロロエトキシ基、2-フルオロエトキシ基、2,2-ジフルオロエトキシ基、1,1-ジフルオロエトキシ基、1,2-ジフルオロエトキシ基、1-クロロ-2-フルオロエトキシ基、2,2,2-トリフルオロエトキシ基、1,1,2,2,2-ペンタフルオロエトキシ基、2,2,2-トリクロロエトキシ基、3-フルオロプロポキシ基、2-フルオロプロポキシ基、1-フルオロプロポキシ基、3,3-ジフルオロプロポキシ基、2,2-ジフルオロプロポキシ基、1,1-ジフルオロプロポキシ基、4-フルオロブトキシ基、5-フルオロペンチルオキシ基、6-フルオロヘキシルオキシ基等が挙げられる。 The “halo C 1 -C 6 alkoxy group” as used herein means a C 1 -C 6 alkoxy group in which at least one hydrogen atom is substituted with the same or different halogen atom. Examples of halo C 1 -C 6 alkoxy groups include monofluoromethoxy, difluoromethoxy, trifluoromethoxy, 2-chloroethoxy, 2-fluoroethoxy, 2,2-difluoroethoxy, 1,1- difluoroethoxy group, 1,2-difluoroethoxy group, 1-chloro-2-fluoroethoxy group, 2,2,2-trifluoroethoxy group, 1,1,2,2,2-pentafluoroethoxy group, 2, 2,2-trichloroethoxy group, 3-fluoropropoxy group, 2-fluoropropoxy group, 1-fluoropropoxy group, 3,3-difluoropropoxy group, 2,2-difluoropropoxy group, 1,1-difluoropropoxy group, 4-fluorobutoxy group, 5-fluoropentyloxy group, 6-fluorohexyloxy group and the like.
 本明細書に示される「C~Cアルコキシカルボニル基」とは、炭素数1~6個の直鎖または分枝鎖のアルコキシ基が結合したカルボニル基を意味する。C~Cアルコキシカルボニル基として、例えば、メトキシカルボニル基、エトキシカルボニル基、1-プロポキシカルボニル基、イソプロポキシカルボニル基、イソブトキシカルボニル基、1-ブトキシカルボニル基、sec-ブトキシカルボニル基、tert-ブトキシカルボニル基、1-ペンチルオキシカルボニル基、1-ヘキシルオキシカルボニル基等が挙げられる。 The “C 1 -C 6 alkoxycarbonyl group” as used herein means a carbonyl group to which a straight or branched chain alkoxy group having 1 to 6 carbon atoms is attached. Examples of C 1 to C 6 alkoxycarbonyl groups include methoxycarbonyl, ethoxycarbonyl, 1-propoxycarbonyl, isopropoxycarbonyl, isobutoxycarbonyl, 1-butoxycarbonyl, sec-butoxycarbonyl, tert- butoxycarbonyl group, 1-pentyloxycarbonyl group, 1-hexyloxycarbonyl group and the like.
 本明細書に示される「C~Cアルキルアミノ基」とは、アミノ基の1個または2個の水素原子が、総炭素数1~6個の直鎖または分枝鎖のアルキル基で置換されたアミノ基を意味する。C~Cアルキルアミノ基として、例えば、メチルアミノ基、エチルアミノ基、1-プロピルアミノ基、イソプロピルアミノ基、1-ブチルアミノ基、イソブチルアミノ基、sec-ブチルアミノ基、tert-ブチルアミノ基、1-ペンチルアミノ基、イソペンチルアミノ基、ネオペンチルアミノ基、1-メチルブチルアミノ基、2-メチルブチルアミノ基、1,2-ジメチルプロピルアミノ基、1-ヘキシルアミノ基、イソヘキシルアミノ基、ジメチルアミノ基、ジエチルアミノ基、N-エチル-N-メチルアミノ基、N-エチル-N-プロピルアミノ基等が挙げられる。 The "C 1 -C 6 alkylamino group" as used herein means a linear or branched alkyl group in which 1 or 2 hydrogen atoms in the amino group have a total of 1 to 6 carbon atoms. It means a substituted amino group. Examples of C 1 -C 6 alkylamino groups include methylamino group, ethylamino group, 1-propylamino group, isopropylamino group, 1-butylamino group, isobutylamino group, sec-butylamino group and tert-butylamino group. group, 1-pentylamino group, isopentylamino group, neopentylamino group, 1-methylbutylamino group, 2-methylbutylamino group, 1,2-dimethylpropylamino group, 1-hexylamino group, isohexylamino group, dimethylamino group, diethylamino group, N-ethyl-N-methylamino group, N-ethyl-N-propylamino group and the like.
 本明細書に示される「C~Cアルキルアミノカルボニル基」とは、総炭素数1~6個の直鎖または分枝鎖のアルキルアミノ基が結合したカルボニル基を意味する。C~Cアルキルアミノカルボニル基として、例えば、メチルアミノカルボニル基、エチルアミノカルボニル基、1-プロピルアミノカルボニル基、イソプロピルアミノカルボニル基、1-ブチルアミノカルボニル基、イソブチルアミノカルボニル基、sec-ブチルアミノカルボニル基、tert-ブチルアミノカルボニル基、1-ペンチルアミノカルボニル基、イソペンチルアミノカルボニル基、ネオペンチルアミノカルボニル基、1-メチルブチルアミノカルボニル基、2-メチルブチルアミノカルボニル基、1,2-ジメチルプロピルアミノカルボニル基、1-ヘキシルアミノカルボニル基、イソヘキシルアミノカルボニル基、ジメチルアミノカルボニル基、ジエチルアミノカルボニル基、N-エチル-N-メチルアミノカルボニル基、N-エチル-N-プロピルアミノカルボニル基等が挙げられる。 The “C 1 -C 6 alkylaminocarbonyl group” as used herein means a carbonyl group to which a linear or branched alkylamino group having a total of 1 to 6 carbon atoms is bonded. Examples of C 1 -C 6 alkylaminocarbonyl groups include methylaminocarbonyl, ethylaminocarbonyl, 1-propylaminocarbonyl, isopropylaminocarbonyl, 1-butylaminocarbonyl, isobutylaminocarbonyl and sec-butyl. aminocarbonyl group, tert-butylaminocarbonyl group, 1-pentylaminocarbonyl group, isopentylaminocarbonyl group, neopentylaminocarbonyl group, 1-methylbutylaminocarbonyl group, 2-methylbutylaminocarbonyl group, 1,2- dimethylpropylaminocarbonyl group, 1-hexylaminocarbonyl group, isohexylaminocarbonyl group, dimethylaminocarbonyl group, diethylaminocarbonyl group, N-ethyl-N-methylaminocarbonyl group, N-ethyl-N-propylaminocarbonyl group, etc. is mentioned.
 本明細書に示される「C~Cアシルアミノ基」とは、アミノ基の1個または2個の水素原子が、炭素数1~6個の直鎖または分枝鎖のアシル基で置換されたアミノ基を意味する。C~Cアシルアミノ基として、例えば、ホルミルアミノ基、アセチルアミノ基、1-プロパノイルアミノ基、1-ブタノイルアミノ基、1-ペンタノイルアミノ基、ヘキサノイルアミノ基等が挙げられる。 A "C 1 -C 6 acylamino group" as used herein means that 1 or 2 hydrogen atoms of an amino group are substituted with a linear or branched acyl group having 1 to 6 carbon atoms. means an amino group. The C 1 -C 6 acylamino group includes, for example, formylamino group, acetylamino group, 1-propanoylamino group, 1-butanoylamino group, 1-pentanoylamino group, hexanoylamino group and the like.
 本明細書に示される「C~Cアルキルスルファニル基」とは、炭素数1~6個の直鎖または分岐鎖のアルキル基が硫黄原子に結合した基を意味する。C~Cアルキルスルファニル基として、例えば、メチルスルファニル基、エチルスルファニル基、1-プロピルスルファニル基、イソプロピルスルファニル基、1-ブチルスルファニル基、イソブチルスルファニル基、sec-ブチルスルファニル基、tert-ブチルスルファニル基等が挙げられる。 The “C 1 -C 6 alkylsulfanyl group” as used herein means a group in which a straight or branched chain alkyl group having 1 to 6 carbon atoms is bonded to a sulfur atom. Examples of C 1 -C 6 alkylsulfanyl groups include methylsulfanyl group, ethylsulfanyl group, 1-propylsulfanyl group, isopropylsulfanyl group, 1-butylsulfanyl group, isobutylsulfanyl group, sec-butylsulfanyl group and tert-butylsulfanyl group. and the like.
 本明細書に示される「ハロC~Cアルキルスルファニル基」とは、少なくとも1つの水素原子が同種または異種のハロゲン原子で置換されたC~Cアルキルスルファニル基を意味する。ハロC~Cアルキルスルファニル基として、例えば、フルオロメチルスルファニル基、ジフルオロメチルスルファニル基、トリフルオロメチルスルファニル基、2-フルオロエチルスルファニル基、2-クロロエチルスルファニル基、2,2-ジフルオロエチルスルファニル基、1,1-ジフルオロエチルスルファニル基、1,2-ジフルオロエチルスルファニル基、1-クロロ-2-フルオロエチルスルファニル基、2,2,2-トリフルオロエチルスルファニル基、1,1,2,2,2-ペンタフルオロエチルスルファニル基、2,2,2-トリクロロエチルスルファニル基、3-フルオロプロピルスルファニル基、2-フルオロプロピルスルファニル基、1-フルオロプロピルスルファニル基、3,3-ジフルオロプロピルスルファニル基、2,2-ジフルオロプロピルスルファニル基、1,1-ジフルオロプロピルスルファニル基、4-フルオロブチルスルファニル基、5-フルオロペンチルスルファニル基、6-フルオロヘキシルスルファニル基等が挙げられる。 The “halo C 1 -C 6 alkylsulfanyl group” as used herein means a C 1 -C 6 alkylsulfanyl group in which at least one hydrogen atom is substituted with the same or different halogen atom. halo C 1 -C 6 alkylsulfanyl group, for example, fluoromethylsulfanyl group, difluoromethylsulfanyl group, trifluoromethylsulfanyl group, 2-fluoroethylsulfanyl group, 2-chloroethylsulfanyl group, 2,2-difluoroethylsulfanyl group; group, 1,1-difluoroethylsulfanyl group, 1,2-difluoroethylsulfanyl group, 1-chloro-2-fluoroethylsulfanyl group, 2,2,2-trifluoroethylsulfanyl group, 1,1,2,2 , 2-pentafluoroethylsulfanyl group, 2,2,2-trichloroethylsulfanyl group, 3-fluoropropylsulfanyl group, 2-fluoropropylsulfanyl group, 1-fluoropropylsulfanyl group, 3,3-difluoropropylsulfanyl group, 2,2-difluoropropylsulfanyl group, 1,1-difluoropropylsulfanyl group, 4-fluorobutylsulfanyl group, 5-fluoropentylsulfanyl group, 6-fluorohexylsulfanyl group and the like.
 本明細書に示される「芳香族炭化水素環基」として、例えば、フェニル基、インデニル基、1-ナフチル基、2-ナフチル基、アズレニル基、ヘプタレニル基、ビフェニル基、インダセニル基、アセナフチル基、フルオレニル基、フェナレニル基、フェナントレニル基、アントラセニル基、ベンゾシクロオクテニル基等が挙げられる。 The "aromatic hydrocarbon ring group" shown in this specification includes, for example, phenyl group, indenyl group, 1-naphthyl group, 2-naphthyl group, azulenyl group, heptalenyl group, biphenyl group, indacenyl group, acenaphthyl group, fluorenyl group, phenalenyl group, phenanthrenyl group, anthracenyl group, benzocyclooctenyl group and the like.
 本明細書に示される「5~10員ヘテロアリール基」とは、独立して、窒素原子、酸素原子および硫黄原子からなる群から選択される1~4個の環内ヘテロ原子を含む、5~10員の単環芳香族複素環基または縮環芳香族複素環基を意味し、窒素および硫黄原子は所望により酸化されてもよい(すなわち、N→O、SOまたはSOである)。5~10員ヘテロアリール基の例として、以下に限定されないが、ベンゾイミダゾリル基、ベンゾフラニル基、ベンゾチオフラニル基、ベンゾチオフェニル基、ベンゾオキサゾリル基、ベンゾオキサゾリニル基、ベンゾチアゾリル基、ベンゾトリアゾリル基、ベンゾイソキサゾリル基、ベンゾイソチアゾリル基、ベンゾイミダゾリニル基、フラニル基、イミダゾリジニル基、イミダゾリル基、1H-インダゾリル基、イミダゾロピリジニル基、インドレニル基、インドリジニル基、3H-インドリル基、イソベンゾフラニル基、イソインダゾリル基、イソインドリル基、イソキノリニル基、イソチアゾリル基、イソチアゾロピリジニル基、イソキサゾリル基、イソキサゾロピリジニル基、ナフチリジニル基、1,2,3-オキサジアゾリル基、1,2,4-オキサジアゾリル基、1,2,5-オキサジアゾリル基、1,3,4-オキサジアゾリル基、オキサゾリジニル基、オキサゾリル基、オキサゾロピリジニル基、オキサゾリジニルペリミジニル基、オキシインドリル基、ピリミジニル基、ピラジニル基、ピラゾリジニル基、ピラゾリニル基、ピラゾロピリジニル基、ピラゾリル基、ピリダジニル基、ピリドオキサゾリル基、ピリドイミダゾリル基、ピリドチアゾリル基、ピリジニル基、ピロロピリジニル基、キナゾリニル基、キノリニル基、4H-キノリジニル基、キノキサリニル基、キヌクリジニル基、テトラゾリル基、6H-1,2,5-チアジアジニル基、1,2,3-チアジアゾリル基、1,2,4-チアジアゾリル基、1,2,5-チアジアゾリル基、1,3,4-チアジアゾリル基、チアントレニル基、チアゾリル基、チエニル基、チアゾロピリジニル基、チエノチアゾリル基、チエノオキサゾリル基、チエノイミダゾリル基、チオフェニル基、トリアジニル基、1,2,3-トリアゾリル基、1,2,4-トリアゾリル基、1,2,5-トリアゾリル基および1,3,4-トリアゾリル基等が挙げられる。また、上記のヘテロ環を含有する、縮合環およびスピロ環化合物も包含される。 A "5- to 10-membered heteroaryl group" as used herein includes 1 to 4 endocyclic heteroatoms independently selected from the group consisting of nitrogen, oxygen and sulfur atoms. means a to 10-membered monocyclic aromatic heterocyclic group or fused ring aromatic heterocyclic group, wherein the nitrogen and sulfur atoms are optionally oxidized (ie, N→O, SO or SO 2 ). Examples of 5- to 10-membered heteroaryl groups include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzothiazolyl, benzo triazolyl group, benzisoxazolyl group, benzisothiazolyl group, benzimidazolinyl group, furanyl group, imidazolidinyl group, imidazolyl group, 1H-indazolyl group, imidazolopyridinyl group, indolenyl group, indolizinyl group, 3H-indolyl group, isobenzofuranyl group, isoindazolyl group, isoindolyl group, isoquinolinyl group, isothiazolyl group, isothiazolopyridinyl group, isoxazolyl group, isoxazolopyridinyl group, naphthyridinyl group, 1,2,3- oxadiazolyl group, 1,2,4-oxadiazolyl group, 1,2,5-oxadiazolyl group, 1,3,4-oxadiazolyl group, oxazolidinyl group, oxazolyl group, oxazolopyridinyl group, oxazolidinylperimidinyl group, oxindolyl group, pyrimidinyl group, pyrazinyl group, pyrazolidinyl group, pyrazolinyl group, pyrazolopyridinyl group, pyrazolyl group, pyridazinyl group, pyridooxazolyl group, pyridoimidazolyl group, pyridothiazolyl group, pyridinyl group, pyrrolopyridinyl group, quinazolinyl group, quinolinyl group, 4H-quinolidinyl group, quinoxalinyl group, quinuclidinyl group, tetrazolyl group, 6H-1,2,5-thiadiazinyl group, 1,2,3-thiadiazolyl group, 1,2,4-thiadiazolyl group, 1,2,5-thiadiazolyl group, 1,3,4-thiadiazolyl group, thianthrenyl group, thiazolyl group, thienyl group, thiazolopyridinyl group, thienothiazolyl group, thienooxazolyl group, thienoimidazolyl group, thiophenyl triazinyl group, 1,2,3-triazolyl group, 1,2,4-triazolyl group, 1,2,5-triazolyl group and 1,3,4-triazolyl group. Also included are fused ring and spirocyclic compounds containing the above heterocycles.
 本明細書に示される「C~C10シクロアルキル基」とは、炭素数3~10個の単環式または二環式の飽和脂環式炭化水素基を意味し、架橋型、スピロ型であり得る。C~C10シクロアルキル基として、例えば、シクロプロピル基、シクロブチル基、シクロペンチル基、シクロヘキシル基、シクロへプチル基、シクロオクチル基、スピロヘプチル基、スピロオクチル基、オクタヒドロペンタレニル基等が挙げられる。また、C~C10シクロアルキル基は、さらなる芳香族炭化水素環基または5~10員ヘテロアリール基と縮合されていてもよく、芳香族炭化水素環基または5~10員ヘテロアリール基が縮合しているC~C10シクロアルキル基として、ジヒドロインデニル基、テトラヒドロナフチル基等が挙げられる。 The “C 3 -C 10 cycloalkyl group” as used herein means a monocyclic or bicyclic saturated alicyclic hydrocarbon group having 3 to 10 carbon atoms, and is a bridged type, spiro type can be Examples of C 3 -C 10 cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, spiroheptyl, spirooctyl and octahydropentalenyl groups. mentioned. The C 3 -C 10 cycloalkyl group may also be fused with a further aromatic hydrocarbon ring group or 5-10 membered heteroaryl group, wherein the aromatic hydrocarbon ring group or 5-10 membered heteroaryl group is The condensed C 3 -C 10 cycloalkyl group includes a dihydroindenyl group, a tetrahydronaphthyl group, and the like.
 また、「C~Cシクロアルキル基」とは、炭素数3~4個のシクロアルキル基を意味する。 Further, "C 3 -C 4 cycloalkyl group" means a cycloalkyl group having 3 to 4 carbon atoms.
 本明細書中に示される「3~10員ヘテロシクロアルキル基」とは、独立して、窒素原子、酸素原子および硫黄原子からなる群から選択される1~4個の環内ヘテロ原子を含む、単環式、二環式、または三環式の3~10員環を有するヘテロシクロアルキル基を意味し、窒素および硫黄ヘテロ原子は所望により酸化されてもよく(すなわち、N→O、SOまたはSOである)、窒素原子は置換されていても、置換されていなくてもよく、カルボニル基を1~3個有していてもよく、環内に二重結合を1個有してもよい。また3~10員ヘテロシクロアルキル基は、架橋型、スピロ型であり得る。3~10員ヘテロシクロアルキル基は、さらなる芳香族炭化水素環基または5~10員ヘテロアリール基と縮合されていてもよい。3~10員ヘテロシクロアルキル基として、例えば、アジリジニル基、アゼチジニル基、ピロリジニル基、ピペリジニル基、アゼパニル基、アゾカニル基、ジヒドロピロリル基、テトラヒドロピリジニル基、ピペラジニル基、モルホリニル基、チオモルホリニル基、1-オキシドチオモルホリニル基、1,1-ジオキシドチオモルホリニル基、オキサゼピニル基、チアゼパニル基、1-オキシド-1,4-チアゼパニル基、1,1-ジオキシド-1,4-チアゼパニル基、1,4-ジアゼパニル基、1,4-オキサゾカニル基、1,5-オキサゾカニル基、オキセタニル基、テトラヒドロフラニル基、テトラヒドロピラニル基、オクタヒドロシクロペンタ[c]ピロリル基、3-アザビシクロ[3.2.0]ヘプタニル基、3-アザビシクロ[3.1.0]ヘキサニル基、5-アザビシクロ[2.1.1]ヘキサニル基、2-アザビシクロ[2.1.1]ヘキサニル基、2-アザビシクロ[4.1.0]ヘプタニル基、3-アザビシクロ[4.1.0]ヘプタニル基、2-アザビシクロ[4.2.0]オクタニル基、3-アザビシクロ[4.2.0]オクタニル基、3-アザビシクロ[3.1.1]ヘプタニル基、2-アザビシクロ[2,2,1]ヘプタニル基、6-アザビシクロ[3.1.1]ヘプタニル基、8-アザビシクロ[3.2.1]オクタニル基、3-アザビシクロ[3.2.1]オクタニル基、6-アザビシクロ[3.2.1]オクタニル基、4-アザスピロ[2.4]ヘプタニル基、5-アザスピロ[2.4]ヘプタニル基、1-オキソ-5-アザスピロ[2.4]ヘプタニル基、5-アザスピロ[3.4]オクタニル基、6-アザスピロ[3.4]オクタニル基、2-オキソ-6-アザスピロ[3.4]オクタニル基、1-オキソ-6-アザスピロ[3.4]オクタニル基、4-アザスピロ[2.5]オクタニル基、5-アザスピロ[2.5]オクタニル基、6-アザスピロ[2.5]オクタニル基、1-オキサ-5-アザスピロ[2.5]オクタニル基、4-オキサ-7-アザスピロ[2.5]オクタニル基、1-オキサ-6-アザスピロ[2.5]オクタニル基、2,6-ジアザスピロ[3.4]オクタニル基等が挙げられる。 The "3- to 10-membered heterocycloalkyl group" shown herein contains 1 to 4 endocyclic heteroatoms independently selected from the group consisting of nitrogen, oxygen and sulfur atoms. , monocyclic, bicyclic or tricyclic heterocycloalkyl groups having 3-10 membered rings, wherein the nitrogen and sulfur heteroatoms may be optionally oxidized (i.e., N→O, SO or SO 2 ), the nitrogen atom may be substituted or unsubstituted, may have 1 to 3 carbonyl groups, have 1 double bond in the ring, good too. A 3- to 10-membered heterocycloalkyl group may also be bridged or spiro. A 3-10 membered heterocycloalkyl group may be optionally fused with a further aromatic hydrocarbon ring group or a 5-10 membered heteroaryl group. 3- to 10-membered heterocycloalkyl groups such as aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, azocanyl, dihydropyrrolyl, tetrahydropyridinyl, piperazinyl, morpholinyl, thiomorpholinyl, 1-oxidethiomorpholinyl group, 1,1-dioxidethiomorpholinyl group, oxazepinyl group, thiazepanyl group, 1-oxide-1,4-thiazepanyl group, 1,1-dioxide-1,4-thiazepanyl group , 1,4-diazepanyl group, 1,4-oxazocanyl group, 1,5-oxazocanyl group, oxetanyl group, tetrahydrofuranyl group, tetrahydropyranyl group, octahydrocyclopenta[c]pyrrolyl group, 3-azabicyclo[3. 2.0]heptanyl group, 3-azabicyclo[3.1.0]hexanyl group, 5-azabicyclo[2.1.1]hexanyl group, 2-azabicyclo[2.1.1]hexanyl group, 2-azabicyclo[ 4.1.0]heptanyl group, 3-azabicyclo[4.1.0]heptanyl group, 2-azabicyclo[4.2.0]octanyl group, 3-azabicyclo[4.2.0]octanyl group, 3- azabicyclo[3.1.1]heptanyl group, 2-azabicyclo[2,2,1]heptanyl group, 6-azabicyclo[3.1.1]heptanyl group, 8-azabicyclo[3.2.1]octanyl group, 3-azabicyclo[3.2.1]octanyl group, 6-azabicyclo[3.2.1]octanyl group, 4-azaspiro[2.4]heptanyl group, 5-azaspiro[2.4]heptanyl group, 1- oxo-5-azaspiro[2.4]heptanyl group, 5-azaspiro[3.4]octanyl group, 6-azaspiro[3.4]octanyl group, 2-oxo-6-azaspiro[3.4]octanyl group, 1-oxo-6-azaspiro[3.4]octanyl group, 4-azaspiro[2.5]octanyl group, 5-azaspiro[2.5]octanyl group, 6-azaspiro[2.5]octanyl group, 1- Oxa-5-azaspiro[2.5]octanyl group, 4-oxa-7-azaspiro[2.5]octanyl group, 1-oxa-6-azaspiro[2.5]octanyl group, 2,6-diazaspiro[3 .4] octanyl group and the like.
 本明細書に示される「互いに結合して環を形成」とは、環を形成する2つの置換基からさらに任意の水素原子をそれぞれ1個ずつ除いて、水素を除いた部位同士を結合させることを意味する。例えば、メチレン基に2つの置換基を有する場合、環を形成する2つの置換基がメチル基と1-ヒドロキシエチル基の場合は、
Figure JPOXMLDOC01-appb-C000023
が挙げられる。
As used herein, the term “bonded to each other to form a ring” refers to removing any one hydrogen atom from each of the two substituents forming the ring, and bonding the hydrogen-free sites to each other. means For example, when the methylene group has two substituents, if the two substituents forming the ring are a methyl group and a 1-hydroxyethyl group,
Figure JPOXMLDOC01-appb-C000023
is mentioned.
 以下、本実施形態をより詳細に説明する。
 以下において、一般式が有する官能基の定義については、すでに記載した定義を引用してその説明を省略することがある。引用している定義は、以下に記載する実施形態の説明中に記載した定義を指している。
 また、一般式が有する官能基についての定義に関し、特に言及しない限り、同一の符号で表される定義は、その符号を含む各一般式の間で共通する。
The present embodiment will be described in more detail below.
In the following description, the definitions of the functional groups in the general formulas may be omitted by citing the definitions already described. The definitions quoted refer to the definitions set forth in the description of the embodiments set forth below.
In addition, regarding definitions of functional groups in general formulas, definitions represented by the same reference numerals are common among general formulas including the reference unless otherwise specified.
 本実施形態は、下記一般式(I)で表される化合物またはその薬理学的に許容される塩に関する。
一般式(I):
Figure JPOXMLDOC01-appb-C000024
[式(I)中、RとRとは互いに結合して環を形成して、RとRが結合するピロール環とともに、以下のA1)の構造をとり、*が式(I)中の-CO-との結合位置を示し;
Figure JPOXMLDOC01-appb-C000025
3はC~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基またはヒドロキシC~Cアルキル基(該C~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基およびヒドロキシC~Cアルキル基はC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基およびヒドロキシC~Cアルキル基からなる群より選択される置換基で一つまたは複数置換されてもよく、置換基同士で互いに結合して環を形成してもよい)であり;
Xは炭素原子または4位、5位、6位、7位のいずれかの窒素原子を示し、それぞれインドールまたはアザインドール環類であり;
およびRはそれぞれ独立して、水素原子、ハロゲン原子、C~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基またはヒドロキシC~Cアルキル基(該C~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基およびヒドロキシC~Cアルキル基はC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基およびヒドロキシC~Cアルキル基からなる群より選択される置換基で一つまたは複数置換されてもよく、置換基同士で互いに結合して環を形成してもよい)であり;
は以下のB1)であり、*が式(I)中の-N-との結合位置を示し;
Figure JPOXMLDOC01-appb-C000026
およびRはそれぞれ独立して水素原子、C~Cアルキル基またはヒドロキシC~Cアルキル基であり;
およびRは互いに結合して環を形成してもよく;
およびRはRingAと結合して環を形成してもよく;
mは0または1であり;
RingAは、それぞれR10、R11およびR12で置換されてもよい、芳香族炭化水素環基、C~C10シクロアルキル基、5~10員ヘテロアリール基または3~10員ヘテロシクロアルキル基であり;
10およびR11はそれぞれ独立して水素原子、ハロゲン原子、水酸基、アミノ基、シアノ基、カルバモイル基(-CONH)、C~Cアルキル基、ハロC~Cアルキル基、ヒドロキシC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアルキルスルファニル基、ハロC~Cアルキルスルファニル基、C~Cアシル基、C~Cアルコキシカルボニル基、C~Cアルキルアミノカルボニル基またはC~Cアシルアミノ基であり;
12は-(CR1314-V-(CR1516-(W)-Qであり;
Vは結合、-O-または-NR17-であり;
Wは-NR18-であり;
13、R14、R15、R16、R17およびR18はそれぞれ独立して水素原子またはC~Cアルキル基であり;
qおよびrはそれぞれ独立して0~6の整数であり;
nは0または1であり;
Qは水素原子、水酸基、C~Cアルキル基、C~C10シクロアルキル基、5~10員ヘテロアリール基または3~10員ヘテロシクロアルキル基(該C~Cアルキルアミノ基、C~C10シクロアルキル基、5~10員ヘテロアリール基および3~10員ヘテロシクロアルキル基は、ハロゲン原子、C~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアルコキシカルボニル基、C~Cアルキルアミノカルボニル基およびヒドロキシC~Cアルキル基からなる群より選択される置換基で一つまたは複数置換されてもよい)であり;
10、R11およびR12は互いに結合して環を形成してもよい]
で表される化合物、またはその薬理学的に許容される塩。
This embodiment relates to a compound represented by the following general formula (I) or a pharmacologically acceptable salt thereof.
General formula (I):
Figure JPOXMLDOC01-appb-C000024
[In the formula (I), R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, the structure of A1) below is taken, and * is the formula (I ) indicates the bonding position with -CO- in;
Figure JPOXMLDOC01-appb-C000025
R 3 is C 1 -C 6 alkyl group, C 2 -C 6 alkenyl group, halo C 1 -C 6 alkyl group, C 1 -C 6 alkoxy group, C 1 -C 6 alkylamino group, C 1 -C 6 acyl group, C 1 -C 6 alkoxycarbonyl group, C 3 -C 10 cycloalkyl group or hydroxy C 1 -C 6 alkyl group (the C 1 -C 6 alkyl group, C 2 -C 6 alkenyl group, halo C 1 -C6 alkyl groups, C1 - C6 alkoxy groups, C1 - C6 alkylamino groups, C1 - C6 acyl groups, C1 - C6 alkoxycarbonyl groups, C3 - C10 cycloalkyl groups and hydroxy C 1 -C 6 alkyl group is a substituent selected from the group consisting of C 1 -C 6 alkyl group, C 1 -C 6 alkoxy group, C 1 -C 6 alkylamino group and hydroxy C 1 -C 6 alkyl group may be substituted one or more times, and the substituents may be bonded to each other to form a ring);
X represents a carbon atom or a nitrogen atom at any of the 4-, 5-, 6-, and 7-positions, and is an indole or azaindole ring, respectively;
R 6 and R 7 are each independently a hydrogen atom, a halogen atom, a C 1 -C 6 alkyl group, a C 2 -C 6 alkenyl group, a halo C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group, C 1 -C 6 alkylamino group, C 1 -C 6 acyl group, C 1 -C 6 alkoxycarbonyl group, C 3 -C 10 cycloalkyl group or hydroxy C 1 -C 6 alkyl group (the C 1 -C 6 Alkyl Group, C 2 -C 6 Alkenyl Group, Halo C 1 -C 6 Alkyl Group, C 1 -C 6 Alkoxy Group, C 1 -C 6 Alkylamino Group, C 1 -C 6 Acyl Group, C 1 -C 6 Alkoxycarbonyl groups, C 3 -C 10 cycloalkyl groups and hydroxy C 1 -C 6 alkyl groups are C 1 -C 6 alkyl groups, C 1 -C 6 alkoxy groups, C 1 -C 6 alkylamino groups and hydroxy C 1 may be substituted with one or more substituents selected from the group consisting of ~ C6 alkyl groups, and the substituents may be bonded to each other to form a ring);
R 4 is B1) below, * indicates the bonding position with -N- in formula (I);
Figure JPOXMLDOC01-appb-C000026
R 8 and R 9 are each independently a hydrogen atom, a C 1 -C 6 alkyl group or a hydroxy C 1 -C 6 alkyl group;
R 8 and R 9 may be joined together to form a ring;
R 8 and R 9 may combine with RingA to form a ring;
m is 0 or 1;
RingA is an aromatic hydrocarbon ring group, a C3 - C10 cycloalkyl group, a 5- to 10-membered heteroaryl group or a 3- to 10-membered heterocycloalkyl group, each of which may be substituted with R 10 , R 11 and R 12 is a group;
R 10 and R 11 each independently represent a hydrogen atom, a halogen atom, a hydroxyl group, an amino group, a cyano group, a carbamoyl group (—CONH 2 ), a C 1 -C 6 alkyl group, a halo C 1 -C 6 alkyl group, a hydroxy C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylamino, C 1 -C 6 alkylsulfanyl, halo C 1 -C 6 alkylsulfanyl, C 1 -C 6 acyl a C 1 -C 6 alkoxycarbonyl group, a C 1 -C 6 alkylaminocarbonyl group or a C 1 -C 6 acylamino group;
R 12 is -(CR 13 R 14 ) r -V-(CR 15 R 16 ) q -(W) n -Q;
V is a bond, -O- or -NR 17 -;
W is -NR 18 -;
R 13 , R 14 , R 15 , R 16 , R 17 and R 18 are each independently a hydrogen atom or a C 1 -C 6 alkyl group;
q and r are each independently an integer from 0 to 6;
n is 0 or 1;
Q is a hydrogen atom, a hydroxyl group, a C 1 to C 6 alkyl group, a C 3 to C 10 cycloalkyl group, a 5 to 10 membered heteroaryl group or a 3 to 10 membered heterocycloalkyl group (the C 1 to C 6 alkylamino group , C 3 to C 10 cycloalkyl groups, 5 to 10 membered heteroaryl groups and 3 to 10 membered heterocycloalkyl groups are halogen atoms, C 1 to C 6 alkyl groups, C 1 to C 6 alkoxy groups, C 1 to substituted with one or more substituents selected from the group consisting of C 6 alkylamino group, C 1 -C 6 alkoxycarbonyl group, C 1 -C 6 alkylaminocarbonyl group and hydroxy C 1 -C 6 alkyl group may be);
R 10 , R 11 and R 12 may combine with each other to form a ring]
A compound represented by or a pharmacologically acceptable salt thereof.
 本実施形態の好ましい化合物としては、例えば、以下の化合物が挙げられる。 Preferred compounds of the present embodiment include, for example, the following compounds.
Figure JPOXMLDOC01-appb-T000027
Figure JPOXMLDOC01-appb-T000027
Figure JPOXMLDOC01-appb-T000028
Figure JPOXMLDOC01-appb-I000029
Figure JPOXMLDOC01-appb-T000028
Figure JPOXMLDOC01-appb-I000029
Figure JPOXMLDOC01-appb-T000030
Figure JPOXMLDOC01-appb-I000031
Figure JPOXMLDOC01-appb-T000030
Figure JPOXMLDOC01-appb-I000031
 さらに本実施形態の化合物(I)、またはその薬理学的に許容される塩は、水和物または溶媒和物として存在することもある。上記に具体的に記載した好ましい化合物を含めて、前記一般式(I)で表される誘導体またはその塩が形成する任意の水和物および溶媒和物は、いずれも本発明の範囲に包含される。溶媒和物を形成し得る溶媒としては、メタノール、エタノール、イソプロピルアルコール、アセトン、酢酸エチル、ジクロロメタン、ジイソプロピルエーテル等が挙げられる。 Furthermore, compound (I) of the present embodiment or a pharmacologically acceptable salt thereof may exist as a hydrate or solvate. Any hydrate and solvate formed by the derivative represented by the general formula (I) or a salt thereof, including the preferred compounds specifically described above, are included in the scope of the present invention. be. Solvents that can form solvates include methanol, ethanol, isopropyl alcohol, acetone, ethyl acetate, dichloromethane, diisopropyl ether, and the like.
 本実施形態の化合物(I)は、必要に応じて、その薬理学的に許容される塩とすることができる。薬理学的に許容される塩とは、薬学上許容される非毒性塩基または酸(例えば無機または有機塩基および無機または有機酸)との塩を意味する。本実施形態の化合物(I)の薬学的に許容される塩は、第5版実験化学講座(日本化学会編、丸善株式会社刊)、J.Pharm.Sci.1977,66,1-19、および「Handbook of Pharmaceutical Salts:Properties,Selection,and Use」by Stahl and Wermuth(Wiley-VCH、Weinheim、Germany、2002)に記載の方法およびそれに準じた方法により製造することができる。 Compound (I) of the present embodiment can be a pharmacologically acceptable salt thereof, if necessary. Pharmaceutically acceptable salts refer to salts with pharmaceutically acceptable non-toxic bases or acids (eg, inorganic or organic bases and inorganic or organic acids). A pharmaceutically acceptable salt of compound (I) of the present embodiment is described in J. Chem. Pharm. Sci. 1977, 66, 1-19, and "Handbook of Pharmaceutical Salts: Properties, Selection, and Use" by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002) or a method according thereto can be done.
 薬学上許容される非毒性塩基から誘導される塩としては、ナトリウム塩、カリウム塩、カルシウム塩、マグネシウム塩等の無機塩基との塩、ピペリジン、モルホリン、ピロリジン、アルギニン、リジン等の有機塩基との塩が挙げることができる。 Salts derived from pharmaceutically acceptable non-toxic bases include salts with inorganic bases such as sodium salts, potassium salts, calcium salts and magnesium salts, and salts with organic bases such as piperidine, morpholine, pyrrolidine, arginine and lysine. Salt can be mentioned.
 薬学上許容される非毒性酸から誘導される塩としては、例えば、塩酸、臭化水素酸、硫酸、硝酸等の鉱酸との酸付加塩、ギ酸、酢酸、マレイン酸、フマル酸、コハク酸、乳酸、リンゴ酸、酒石酸、クエン酸、メタンスルホン酸、パラトルエンスルホン酸、サリチル酸、ステアリン酸、パルミチン酸等の有機酸との酸付加塩が挙げられる。 Salts derived from pharmaceutically acceptable non-toxic acids include, for example, acid addition salts with mineral acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, formic acid, acetic acid, maleic acid, fumaric acid, succinic acid. , lactic acid, malic acid, tartaric acid, citric acid, methanesulfonic acid, p-toluenesulfonic acid, salicylic acid, stearic acid and palmitic acid.
 本実施形態の化合物(I)、またはその薬理学的に許容される塩には、ラセミ体や光学活性体等の立体異性体も含まれる。 The compound (I) of the present embodiment or a pharmacologically acceptable salt thereof also includes stereoisomers such as racemates and optically active forms.
 本実施形態の化合物(I)が1つ以上の不斉炭素原子または硫黄原子を有する光学異性体である場合、本実施形態の化合物(I)は、各不斉炭素原子または硫黄原子における立体配置が、R配置またはS配置のいずれの立体配置であってもよい。また、本発明は単一のエナンチオマー、単一のジアステレオマー、エナンチオマーの混合物またはジアステレオマーの混合物のいずれも包含する。さらに、光学活性体の混合物において、等量の各光学異性体からなるラセミ体も本発明の範囲に含まれる。本実施形態の化合物(I)がラセミ体の固体または結晶である場合、ラセミ化合物、ラセミ混合物およびラセミ固溶体も本発明の範囲に含まれる。 When compound (I) of the present embodiment is an optical isomer having one or more asymmetric carbon atoms or sulfur atoms, compound (I) of the present embodiment has a steric configuration at each asymmetric carbon atom or sulfur atom. may be in either the R configuration or the S configuration. The invention also includes any single enantiomer, single diastereomer, mixture of enantiomers or mixture of diastereomers. Furthermore, in the mixture of optically active substances, a racemate consisting of equal amounts of each optical isomer is also included in the scope of the present invention. When compound (I) of this embodiment is a racemic solid or crystal, racemic compounds, racemic mixtures and racemic solid solutions are also included in the scope of the present invention.
 本実施形態の化合物(I)において、ジアステレオマー混合物は、クロマトグラフィーや結晶化などの慣用されている方法によって、それぞれのジアステレオマーに分離することができる。また、立体化学的に単一である出発物質を用いることにより、または立体選択的な反応を用いる合成方法によりそれぞれのジアステレオマーを作ることもできる。 In compound (I) of the present embodiment, diastereomeric mixtures can be separated into their respective diastereomers by commonly used methods such as chromatography and crystallization. The respective diastereomers can also be made by using stereochemically uniform starting materials or by synthetic methods employing stereoselective reactions.
 本実施形態の化合物(I)において、cis異性体およびtrans異性体等の幾何異性体が存在する場合、本発明はその幾何異性体のいずれも包含する。 When compound (I) of the present embodiment has geometric isomers such as cis isomers and trans isomers, the present invention includes all such geometric isomers.
 本実施形態の化合物(I)において、互変異性体が存在する場合、本発明はその互変異性体のいずれも包含する。 When compound (I) of the present embodiment has tautomers, the present invention includes all of the tautomers.
 本実施形態の化合物(I)、またはその薬理学的に許容される塩は、同位元素(例、H、14C、35S等)等で標識された化合物であってもよい。当該化合物も本発明に含まれる。 Compound (I) of the present embodiment or a pharmacologically acceptable salt thereof may be a compound labeled with an isotope (eg, 3 H, 14 C, 35 S, etc.) or the like. Such compounds are also included in the present invention.
 さらに、本実施形態の化合物(I)、またはその薬理学的に許容される塩は、HをH(D)に変換した重水素変換体であってもよい。当該化合物も本発明に含まれる。 Furthermore, compound (I) of the present embodiment or a pharmacologically acceptable salt thereof may be a deuterium conversion product in which 1 H is converted to 2 H (D). Such compounds are also included in the present invention.
本実施形態の化合物(I)の製造方法
 本実施形態の化合物(I)は、例えば、以下の合成経路1~2に詳述する方法もしくはそれに準じた方法、またはその他文献記載の方法もしくはそれらに準じた方法に従って製造することができる。
Method for producing compound (I) of the present embodiment, compound (I) of the present embodiment can be prepared, for example, by the method described in detail in the following synthetic pathways 1 to 2 or a method analogous thereto, or by other methods described in literature or by those methods. It can be manufactured according to the method according to.
 式中の化合物(2)~(10)は、塩を形成していてもよく、このような塩としては、例えば化合物(I)の塩と同様のものが挙げられる。また、各工程で得られた化合物は反応液のまま、または粗生成物として得た後に次反応に用いることもできるが、常法に従って反応混合物から再結晶、蒸留、クロマトグラフィーなどの分離手段により容易に単離、精製することができる。 Compounds (2) to (10) in the formula may form salts, and examples of such salts include those similar to the salts of compound (I). In addition, the compound obtained in each step can be used in the next reaction as a reaction solution or after being obtained as a crude product. It can be easily isolated and purified.
[合成経路1]
 化合物(I)(下記式(2)で表される化合物)は、例えば、合成経路1に示す方法、もしくはそれに準じた方法、またはその他文献記載の方法もしくはそれらに準じた方法に従って製造することができる。
[Synthetic route 1]
Compound (I) (the compound represented by the following formula (2)) can be produced, for example, by the method shown in Synthetic Route 1, a method analogous thereto, or a method described in other literatures or a method analogous thereto. can.
Figure JPOXMLDOC01-appb-C000032
(式中、PGは、tert-ブトキシカルボニル基、ベンジルオキシカルボニル基、9-フルオレニルメトキシカルボニル基、アリルオキシカルボニル基、アセチル基、ベンゾイル基またはtert-ブチルジメチルシリル基などの保護基を表し、R、R、RおよびRは前述したものと同義である。)
Figure JPOXMLDOC01-appb-C000032
(In the formula, PG represents a protective group such as a tert-butoxycarbonyl group, a benzyloxycarbonyl group, a 9-fluorenylmethoxycarbonyl group, an allyloxycarbonyl group, an acetyl group, a benzoyl group or a tert-butyldimethylsilyl group. , R 1 , R 2 , R 3 and R 4 are as defined above.)
工程1-1
 化合物(5)は、化合物(3)を、化合物(4)を用いてアミド化することにより製造することができる。
Step 1-1
Compound (5) can be produced by amidating compound (3) with compound (4).
 反応条件として、N,N-ジメチルホルムアミド、N,N-ジメチルアセトアミド、N-メチル-2-ピロリドン、酢酸エチル、ジクロロメタン、アセトニトリル、トルエン、ベンゼン、1,4-ジオキサン、テトラヒドロフラン等、またはそれらの混合溶媒中、化合物(4)等を添加し、0℃~室温、場合によっては加熱還流にて行うことができる。必要に応じて、トリエチルアミンやN,N-ジイソプロピルエチルアミン等の塩基を加えることもできる。また、必要に応じて、1-[ビス(ジメチルアミノ)メチレン]-1H-1,2,3-トリアゾロ[4,5-b]ピリジニウム-3-オキシドヘキサフルオロホスファート(HATU)、1-エチル-3-(3-ジメチルアミノプロピル)カルボジイミド(EDCI)、N,N’-ジシクロヘキシルカルボジイミド(DCC)、または4-(4,6-ジメトキシ-1,3,5-トリアジン-2-イル)-4-メチルモルホリニウムクロリド(DMT-MM)等の縮合剤や、N,N-ジメチル-4-アミノピリジン、ピリジン、1-ヒドロキシベンゾトリアゾール(HOBT)、1-ヒドロキシ-7-アザベンゾトリアゾール(HOAt)等の反応促進剤を添加することができる。 As reaction conditions, N,N-dimethylformamide, N,N-dimethylacetamide, N-methyl-2-pyrrolidone, ethyl acetate, dichloromethane, acetonitrile, toluene, benzene, 1,4-dioxane, tetrahydrofuran, etc., or a mixture thereof Compound (4) and the like are added in a solvent, and the reaction can be carried out at 0° C. to room temperature, optionally with heating under reflux. If necessary, a base such as triethylamine or N,N-diisopropylethylamine can be added. Also, if necessary, 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium-3-oxidehexafluorophosphate (HATU), 1-ethyl -3-(3-dimethylaminopropyl)carbodiimide (EDCI), N,N'-dicyclohexylcarbodiimide (DCC), or 4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4 -Condensing agents such as methylmorpholinium chloride (DMT-MM), N,N-dimethyl-4-aminopyridine, pyridine, 1-hydroxybenzotriazole (HOBT), 1-hydroxy-7-azabenzotriazole (HOAt ) can be added.
工程1-2
 化合物(6)は、化合物(5)の保護基を除去することにより製造することができる。 反応条件として、PGがtert-ブトキシカルボニル基の場合、ジクロロメタン、クロロホルム、1,4-ジオキサン、テトラヒドロフラン、トルエン、ベンゼン、メタノール、エタノール、酢酸エチル、または水等、またはそれらの混合溶媒中、トリフルオロ酢酸、パラトルエンスルホン酸、塩化水素、臭化水素酸、硫酸、三フッ化ホウ素ジエチルエーテル錯体、三臭化ホウ素、または塩化アルミニウム等の酸を添加し、-78℃~室温、場合によっては加熱還流にて行うことができる。
Step 1-2
Compound (6) can be produced by removing the protecting group of compound (5). As reaction conditions, when PG is a tert-butoxycarbonyl group, trifluoro Add an acid such as acetic acid, p-toluenesulfonic acid, hydrogen chloride, hydrobromic acid, sulfuric acid, boron trifluoride diethyl ether complex, boron tribromide, or aluminum chloride, and heat from -78°C to room temperature in some cases. It can be carried out at reflux.
 PGがベンジルオキシカルボニル基の場合、メタノール、エタノール、イソプロピルアルコール、1,4-ジオキサン、1,2-ジメトキシエタン、酢酸エチル、水、テトラヒドロフラン、tert-ブチルメチルエーテル、N,N-ジメチルホルムアミド、トルエン等、またはそれらの混合溶媒中、水素雰囲気下、パラジウムカーボン、ロジウムカーボン、白金カーボン、または酸化白金などの触媒を添加して0℃~加熱還流にて行うことができる。必要に応じて、酢酸、トリフルオロ酢酸、または2,2,2-トリフルオロエタノール等の酸を反応促進剤として添加することができる。または、メタノール、エタノール、イソプロピルアルコール、1,4-ジオキサン、1,2-ジメトキシエタン、酢酸エチル、水、テトラヒドロフラン、tert-ブチルメチルエーテル、N,N-ジメチルホルムアミド、トルエン等、またはそれらの混合溶媒中、トリフルオロ酢酸等の酸を添加して0℃~加熱還流にて行うことができる。 When PG is a benzyloxycarbonyl group, methanol, ethanol, isopropyl alcohol, 1,4-dioxane, 1,2-dimethoxyethane, ethyl acetate, water, tetrahydrofuran, tert-butyl methyl ether, N,N-dimethylformamide, toluene etc., or in a mixed solvent thereof, under a hydrogen atmosphere, by adding a catalyst such as palladium carbon, rhodium carbon, platinum carbon, or platinum oxide, and heating from 0° C. to reflux. If desired, an acid such as acetic acid, trifluoroacetic acid, or 2,2,2-trifluoroethanol can be added as a reaction accelerator. Alternatively, methanol, ethanol, isopropyl alcohol, 1,4-dioxane, 1,2-dimethoxyethane, ethyl acetate, water, tetrahydrofuran, tert-butyl methyl ether, N,N-dimethylformamide, toluene, or a mixed solvent thereof The reaction can be carried out by adding an acid such as trifluoroacetic acid and heating at 0° C. to reflux.
工程1-3
 化合物(2)は、化合物(6)を、化合物(7)を用いてアミド化することにより製造することができる。
 反応条件として、化合物(7)、またはそれらの塩、例えば、アニリン、ベンジルアミン等を添加し、工程1-1と同様の方法にて行うことができる。
Step 1-3
Compound (2) can be produced by amidating compound (6) with compound (7).
As reaction conditions, compound (7) or a salt thereof such as aniline, benzylamine or the like is added, and the reaction can be carried out in the same manner as in step 1-1.
[合成経路2]
 上述の化合物(2)は、例えば、合成経路2に詳述する方法もしくはそれに準じた方法、またはその他文献記載の方法もしくはそれらに準じた方法に従っても製造することができる。
[Synthetic route 2]
The above compound (2) can be produced, for example, according to the method detailed in Synthetic Route 2 or a method analogous thereto, or the method described in other literatures or a method analogous thereto.
Figure JPOXMLDOC01-appb-C000033
(式中、R20はC~Cアルキル基を表し、R、R、RおよびRは前述したものと同義である。)
Figure JPOXMLDOC01-appb-C000033
(In the formula, R 20 represents a C 1 -C 6 alkyl group, and R 1 , R 2 , R 3 and R 4 are as defined above.)
工程2-1
 化合物(9)は、化合物(8)を、化合物(7)を用いてアミド化することにより製造することができる。
 反応条件として、工程1-1と同様の方法にて行うことができる。
Step 2-1
Compound (9) can be produced by amidating compound (8) with compound (7).
As the reaction conditions, the same method as in step 1-1 can be used.
工程2-2
 化合物(10)は、化合物(9)を加水分解することにより製造することができる。
 反応条件として、水、メタノール、エタノール、1-プロパノール、イソプロピルアルコール、テトラヒドロフラン、1,4-ジオキサン等、またはそれらの含水混合溶媒中、水酸化リチウム、水酸化ナトリウム、水酸化カリウム、炭酸カリウム、炭酸ナトリウム、炭酸セシウム等のアルカリ金属塩を添加することで、塩基性条件下、0℃~加熱還流にて行うことができる。また、水、テトラヒドロフラン、1,4-ジオキサン、またはそれらの含水混合溶媒中、塩化水素等を添加することで、酸性条件下、0℃~加熱還流にて行うことができる。
Step 2-2
Compound (10) can be produced by hydrolyzing compound (9).
As the reaction conditions, water, methanol, ethanol, 1-propanol, isopropyl alcohol, tetrahydrofuran, 1,4-dioxane, etc., or a water-containing mixed solvent thereof, lithium hydroxide, sodium hydroxide, potassium hydroxide, potassium carbonate, carbonate By adding an alkali metal salt such as sodium or cesium carbonate, the reaction can be carried out at 0° C. to reflux under basic conditions. Further, by adding hydrogen chloride or the like in water, tetrahydrofuran, 1,4-dioxane, or a mixed solvent containing water thereof, the reaction can be carried out under acidic conditions at 0° C. to heating under reflux.
工程2-3
 化合物(2)は、化合物(10)を、化合物(4)を用いてアミド化することにより製造することができる。
 反応条件として、化合物(4)等を添加し、工程1-1と同様の方法にて行うことができる。
Step 2-3
Compound (2) can be produced by amidating compound (10) with compound (4).
As for the reaction conditions, compound (4) and the like can be added, and the reaction can be carried out in the same manner as in step 1-1.
 上記に示した合成経路は、本実施形態の化合物(I)を製造するための方法の例示であり、本実施形態の化合物(I)は、上記に示した方法もしくはそれに準じた方法、またはその他文献記載の方法もしくはそれらに準じた方法に従って製造することができる。これらの製造方法は、当業者の容易に理解され得るようなスキームへの様々な改変が可能である。 The synthetic route shown above is an example of a method for producing compound (I) of the present embodiment, and compound (I) of the present embodiment can be produced by the method shown above, a method analogous thereto, or other It can be produced according to the method described in the literature or a method based thereon. These production methods allow various modifications to the schemes that can be easily understood by those skilled in the art.
 また、官能基の種類により保護基が必要な場合は、常法に従って適宜導入および脱離の操作を組み合わせて実施することができる。保護基の種類、導入、脱離に関しては、例えば、Theodra W.Green&Peter G.M.Wuts著編、「Greene’s Protective Groups in Organic Synthesis」、fourth edition、Wiley-Interscience、2006年に記載の方法を挙げることができる。 In addition, when a protective group is required depending on the type of functional group, it can be carried out by appropriately combining introduction and elimination operations according to a conventional method. Regarding the type, introduction and elimination of protective groups, see, for example, Theodra W. et al. Green & Peter G. M. Wuts, "Greene's Protective Groups in Organic Synthesis", fourth edition, Wiley-Interscience, 2006.
 本実施形態の化合物(I)を製造するために使用される中間体は、必要に応じて、当該分野における当業者にとって周知の単離・精製手段である溶媒抽出、晶析、再結晶、クロマトグラフィー、分取高速液体クロマトグラフィー等により、単離・精製することができる。 Intermediates used to produce compound (I) of the present embodiment may optionally be isolated and purified by solvent extraction, crystallization, recrystallization, chromatography, and isolation/purification means well known to those skilled in the art. It can be isolated and purified by graphics, preparative high-performance liquid chromatography, or the like.
 また、中間体は、必要に応じて、単離・精製せず、粗生成物のまま次の反応に用いる場合もある。 In addition, intermediates may be used as crude products in the next reaction without isolation and purification, if necessary.
 本実施形態でいう「G9a阻害作用」とは、ヒストンH3の9番目のリジン残基でのモノおよびジメチル化(H3K9me1およびH3K9me2)に関わる主要酵素であるG9aを阻害する作用である。 The "G9a inhibitory action" in the present embodiment is the action of inhibiting G9a, which is a major enzyme involved in mono- and dimethylation (H3K9me1 and H3K9me2) at the 9th lysine residue of histone H3.
 本実施形態の化合物(I)、またはその薬理学的に許容される塩は、例えば、G9a阻害活性試験において、強力な阻害活性を示す。 Compound (I) of the present embodiment, or a pharmacologically acceptable salt thereof, exhibits potent inhibitory activity, for example, in a G9a inhibitory activity test.
 その結果、本実施形態の化合物(I)またはその薬理学的に許容される塩は、がんなどの増殖性疾患、β-グロビン異常症、線維症、疼痛、神経変性疾患、プラダー・ウィリー症候群、マラリア、ウイルス感染症、ミオパチー、自閉症等の治療薬またはその予防薬として有用であると理解できる。 As a result, compound (I) of the present embodiment or a pharmacologically acceptable salt thereof is useful for proliferative diseases such as cancer, β-globin abnormalities, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, , malaria, viral infections, myopathy, autism, etc., or as a preventive agent therefor.
 本実施形態の化合物(I)を有効成分として含有する医薬は、用法に応じ種々の剤形とすることができる。このような剤形としては、例えば、散剤、顆粒剤、細粒剤、ドライシロップ剤、錠剤、カプセル剤、注射剤、液剤、軟膏剤、坐剤、貼付剤、舌下剤等を挙げることができる。 A medicament containing compound (I) of the present embodiment as an active ingredient can be made into various dosage forms depending on the usage. Examples of such dosage forms include powders, granules, fine granules, dry syrups, tablets, capsules, injections, liquids, ointments, suppositories, patches, sublingual agents, and the like.
 これらの医薬は、その剤形に応じて公知の手法により、有効成分としての本実施形態の化合物(I)と、薬学的に許容される添加物とを含む医薬組成物として構成することができる。当該医薬組成物に含有される添加物としては、賦形剤、崩壊剤、結合剤、滑沢剤、希釈剤、緩衝剤、等張化剤、防腐剤、湿潤剤、乳化剤、分散剤、安定化剤、溶解補助剤等を挙げることができる。当該医薬組成物は、本実施形態の化合物(I)と添加物との適宜混合、または化合物(I)を添加物で希釈・溶解することにより調剤することができる。 These medicaments can be configured as pharmaceutical compositions containing the compound (I) of the present embodiment as an active ingredient and pharmaceutically acceptable additives by a known method depending on the dosage form. . Additives contained in the pharmaceutical composition include excipients, disintegrants, binders, lubricants, diluents, buffers, tonicity agents, preservatives, wetting agents, emulsifiers, dispersants, stabilizers, agents, solubilizers, and the like. The pharmaceutical composition can be prepared by appropriately mixing compound (I) of the present embodiment with an additive, or by diluting/dissolving compound (I) with an additive.
 本実施形態に係る医薬は、全身的または局所的に、経口または非経口(経鼻、経肺、静脈内、直腸内、皮下、筋肉、経皮等)により、投与することができる。 The pharmaceutical according to this embodiment can be administered systemically or locally, orally or parenterally (nasally, pulmonary, intravenously, intrarectally, subcutaneously, intramuscularly, transdermally, etc.).
 本明細書は、本願の優先権の基礎である特願2021-092001の明細書に記載される内容を包含する。 The present specification includes the contents described in the specification of Japanese Patent Application No. 2021-092001, which is the basis of the priority of this application.
 以下、試験例、実施例および参考例に基づいて本発明をより詳細に説明する。また、化合物(I)の製造に用いる原料化合物の中にも新規化合物が含まれているので、原料化合物の製造例についても参考例として説明する。本発明は、下記実施例に記載の化合物に限定されるものではなく、本発明の範囲を逸脱しない範囲で変化させてもよい。 The present invention will be described in more detail below based on Test Examples, Examples, and Reference Examples. In addition, since the raw material compounds used in the production of compound (I) also include novel compounds, production examples of the raw material compounds are also described as reference examples. The present invention is not limited to the compounds described in the Examples below and may be varied without departing from the scope of the invention.
 各参考例、各実施例、各表中で用いている記号のうち、H-NMRはプロトン核磁気共鳴分光法により測定したスペクトルを意味する。CDClはクロロホルム-d、DMSO-D6はジメチルスルホキシド-d、CDODはメタノール-dを意味する。MS(ESI)およびMS(ESI)はエレクトロスプレーイオン化法、MS(FI)は電界イオン化法、MS(FD)は電界脱離イオン化法、MS(EI)は電子イオン化法、MS(CI)は化学イオン化法により測定した質量分析スペクトルデータを意味する。室温とは、1~30℃を意味する。 Of the symbols used in each reference example, each example, and each table, 1 H-NMR means a spectrum measured by proton nuclear magnetic resonance spectroscopy. CDCl 3 means chloroform-d, DMSO-D6 means dimethylsulfoxide-d 6 , CD 3 OD means methanol-d 4 . MS (ESI + ) and MS (ESI ) are electrospray ionization methods, MS (FI + ) is field ionization methods, MS (FD + ) is field desorption ionization methods, MS (EI + ) is electron ionization methods, MS (CI + ) means mass spectral data measured by a chemical ionization method. Room temperature means 1 to 30°C.
<参考例1>
Figure JPOXMLDOC01-appb-C000034
<Reference example 1>
Figure JPOXMLDOC01-appb-C000034
 2-((tert-ブトキシカルボニル)アミノ)-4-シクロプロピルブタン酸エチルエステル(2.71g)に水/エタノール=1/1(20mL)と8M水酸化ナトリウム水溶液(4.38mL)を加えて40℃で1時間撹拌した。放冷後、濃塩酸(3.5mL)と飽和NaHCO水溶液(1.0mL)を加えてpH3に調整し、析出固体をろ取した。水で洗浄後、真空乾燥することで2.19g(90.1%)の2-((tert-ブトキシカルボニル)アミノ)-4-シクロプロピルブタン酸を取得した。 To 2-((tert-butoxycarbonyl)amino)-4-cyclopropylbutanoic acid ethyl ester (2.71 g) was added water/ethanol=1/1 (20 mL) and 8M aqueous sodium hydroxide solution (4.38 mL). Stir at 40° C. for 1 hour. After allowing to cool, concentrated hydrochloric acid (3.5 mL) and saturated NaHCO 3 aqueous solution (1.0 mL) were added to adjust the pH to 3, and the precipitated solid was collected by filtration. After washing with water, vacuum drying gave 2.19 g (90.1%) of 2-((tert-butoxycarbonyl)amino)-4-cyclopropylbutanoic acid.
 窒素雰囲気下、2-((tert-ブトキシカルボニル)アミノ)-4-シクロプロピルブタン酸(1.70g)をジメチルホルムアミド(28mL)に溶解し、4-(ジメチルアミノ)メチルベンジルアミン(1.83g)、1-ヒドロキシベンゾトリアゾール一水和物(1.61g)、N,N-ジイソプロピルエチルアミン(4.51mL)を添加し氷冷した。ここへ1-(3-ジメチルアミノプロピル)-3-エチルカルボジイミド塩酸塩(2.01g)を加え、室温で終夜攪拌した。反応液に飽和食塩水を加えて攪拌、分液した後に、水層を酢酸エチルで2回抽出した。有機層を水(10mL)で1回、飽和食塩水で1回洗浄し、硫酸ナトリウムで乾燥した。濃縮した残渣をシリカゲルカラム精製(Yamazen 2L+L;0-20%メタノールのジクロロメタン溶液)し、IPEで懸濁洗浄することで、tert-ブチル(4-シクロプロピル-1-((4-((ジメチルアミノ)メチル)ベンジル)アミノ)-1-オキソブタン-2-イル)カルバメート(2.11g,77.3%)を取得した。 Under a nitrogen atmosphere, 2-((tert-butoxycarbonyl)amino)-4-cyclopropylbutanoic acid (1.70 g) was dissolved in dimethylformamide (28 mL), and 4-(dimethylamino)methylbenzylamine (1.83 g ), 1-hydroxybenzotriazole monohydrate (1.61 g) and N,N-diisopropylethylamine (4.51 mL) were added and cooled with ice. 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (2.01 g) was added thereto, and the mixture was stirred overnight at room temperature. Saturated saline was added to the reaction solution, and the mixture was stirred and separated, and the aqueous layer was extracted twice with ethyl acetate. The organic layer was washed once with water (10 mL) and once with saturated brine, and dried over sodium sulfate. The concentrated residue was purified by silica gel column (Yamazen 2L+L; 0-20% methanol in dichloromethane solution), suspended and washed with IPE to give tert-butyl (4-cyclopropyl-1-((4-((dimethylamino )methyl)benzyl)amino)-1-oxobutan-2-yl)carbamate (2.11 g, 77.3%) was obtained.
 tert-ブチル(4-シクロプロピル-1-((4-((ジメチルアミノ)メチル)ベンジル)アミノ)-1-オキソブタン-2-イル)カルバメート(1.98g,5.08mmol)に2M塩酸/エタノール(25.4mL,50.8mmol HCl)を加えて40℃で2時間撹拌した。放冷後、反応溶液を減圧濃縮し、酢酸エチルで3回共沸した。固化した固体を酢酸エチルで懸濁洗浄することで、2-アミノ-4-シクロプロピル-N-(4-((ジメチルアミノ)メチル)ベンジル)ブタンアミド・2塩酸塩(2.00g,quant.)を取得した。 tert-butyl (4-cyclopropyl-1-((4-((dimethylamino)methyl)benzyl)amino)-1-oxobutan-2-yl)carbamate (1.98g, 5.08mmol) was added with 2M hydrochloric acid/ethanol (25.4 mL, 50.8 mmol HCl) was added and stirred at 40°C for 2 hours. After allowing to cool, the reaction solution was concentrated under reduced pressure and azeotroped three times with ethyl acetate. The solidified solid was suspended and washed with ethyl acetate to give 2-amino-4-cyclopropyl-N-(4-((dimethylamino)methyl)benzyl)butanamide dihydrochloride (2.00 g, quant.). obtained.
1H-NMR (DMSO-D6, 400MHz) δ: -0.06 - 0.01 (2H, m), 0.33 - 0.42 (2H, m), 0.61 - 0.70 (1H, m), 1.13 - 1.21 (2H, m), 1.81 - 1.89 (2H, m), 2.63 (3H, s), 2.64 (3H, s), 3.80 - 3.89 (1H, m), 4.24 (2H, d, J = 6.5 Hz), 4.36 (2H, d, J = 7.5 Hz), 7.36 (2H, d, J = 10.0 Hz), 7.56 (2H, d, J = 10.0 Hz), 8.36 (3H, d, J = 4.5 Hz), 9.26 (1H, t, J = 7.5 Hz), 11.19 (1H, br s).
MS (ESI+): 290 [M+H]+
1 H-NMR (DMSO-D6, 400MHz) δ: -0.06 - 0.01 (2H, m), 0.33 - 0.42 (2H, m), 0.61 - 0.70 (1H, m), 1.13 - 1.21 (2H, m), 1.81 - 1.89 (2H, m), 2.63 (3H, s), 2.64 (3H, s), 3.80 - 3.89 (1H, m), 4.24 (2H, d, J = 6.5 Hz), 4.36 (2H, d, J = 7.5 Hz), 7.36 (2H, d, J = 10.0 Hz), 7.56 (2H, d, J = 10.0 Hz), 8.36 (3H, d, J = 4.5 Hz), 9.26 (1H, t, J = 7.5 Hz), 11.19 (1H, br s).
MS (ESI + ): 290 [M+H] +
<実施例1-1>
Figure JPOXMLDOC01-appb-C000035
<Example 1-1>
Figure JPOXMLDOC01-appb-C000035
 窒素雰囲気下、5-フルオロ-3-メチル-1H-インドール-2-カルボン酸(53.8mg)をジメチルホルムアミド(4.0mL)に溶解し、2-アミノ-4-シクロプロピル-N-(4-((ジメチルアミノ)メチル)ベンジル)ブタンアミド・2塩酸塩(101mg)、1-ヒドロキシベンゾトリアゾール一水和物(64.0mg)、N,N-ジイソプロピルエチルアミン(170μL,0.976mmol)を添加し氷冷した。ここへ1-(3-ジメチルアミノプロピル)-3-エチルカルボジイミド塩酸塩(80.2mg)を加え、室温で終夜攪拌した。反応液に飽和食塩水(6mL)を加えて水層を酢酸エチル(6mL+3mL)で抽出した。有機層を水(3mL)、飽和食塩水(3mL)で洗浄し、硫酸ナトリウムで乾燥した。ろ過・減圧濃縮した残渣をシリカゲルカラム精製(Yamazen 2L+L;0-20%メタノールのジクロロメタン溶液)することで、N-(4-シクロプロピル-1-((4-((ジメチルアミノ)メチル)ベンジル)アミノ)-1-オキソブタン-2-イル)-5-フルオロ-3-メチル-1H-インドール-2-カルボキサミド(94.6mg,72.8%)を取得した。 Under a nitrogen atmosphere, 5-fluoro-3-methyl-1H-indole-2-carboxylic acid (53.8 mg) was dissolved in dimethylformamide (4.0 mL), and 2-amino-4-cyclopropyl-N-(4 -((dimethylamino)methyl)benzyl)butanamide dihydrochloride (101 mg), 1-hydroxybenzotriazole monohydrate (64.0 mg), and N,N-diisopropylethylamine (170 μL, 0.976 mmol) were added. Ice cold. 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (80.2 mg) was added thereto, and the mixture was stirred overnight at room temperature. Saturated saline (6 mL) was added to the reaction solution, and the aqueous layer was extracted with ethyl acetate (6 mL+3 mL). The organic layer was washed with water (3 mL) and saturated brine (3 mL) and dried over sodium sulfate. The residue obtained by filtration and concentration under reduced pressure was purified by silica gel column (Yamazen 2L+L; 0-20% methanol in dichloromethane solution) to give N-(4-cyclopropyl-1-((4-((dimethylamino)methyl)benzyl) Amino)-1-oxobutan-2-yl)-5-fluoro-3-methyl-1H-indole-2-carboxamide (94.6 mg, 72.8%) was obtained.
1H-NMR (DMSO-D6, 400MHz) δ: -0.05 - 0.01 (2H, m), 0.34 - 0.42 (2H, m), 0.64 - 0.74 (1H, m), 1.25 (2H, q, J = 7.5Hz), 1.74 - 1.96 (2H, m), 2.12 (6H, s), 2.48 (3H, s), 3.36 (2H, s), 4.27 (1H, dd, J = 15.2, 5.9 Hz), 4.33 (1H, dd, J = 15.2, 6.0 Hz), 4.54 (1H, td, J = 8.3, 5.3 Hz), 7.07 (1H, td, J = 9.2, 2.6 Hz), 7.22 (4H, s), 7.33 - 7.44 (2H, m), 7.93 (1H, d, J = 7.9 Hz), 8.59 (1H, t, J = 6.0 Hz), 11.42 (1H, br s).
MS (ESI+): 465 [M+H]+
1 H-NMR (DMSO-D6, 400MHz) δ: -0.05 - 0.01 (2H, m), 0.34 - 0.42 (2H, m), 0.64 - 0.74 (1H, m), 1.25 (2H, q, J = 7.5 Hz), 1.74 - 1.96 (2H, m), 2.12 (6H, s), 2.48 (3H, s), 3.36 (2H, s), 4.27 (1H, dd, J = 15.2, 5.9 Hz), 4.33 (1H , dd, J = 15.2, 6.0 Hz), 4.54 (1H, td, J = 8.3, 5.3 Hz), 7.07 (1H, td, J = 9.2, 2.6 Hz), 7.22 (4H, s), 7.33 - 7.44 ( 2H, m), 7.93 (1H, d, J = 7.9 Hz), 8.59 (1H, t, J = 6.0 Hz), 11.42 (1H, br s).
MS (ESI + ): 465 [M+H] +
 対応する出発物質および反応剤を用い、実施例1-1と同様の方法、工程1-3に記した方法、またはそれらに準じた方法により、以下の実施例1-2から1-17を得た。 Using the corresponding starting materials and reactants, the following Examples 1-2 to 1-17 were obtained by the same method as in Example 1-1, the method described in Step 1-3, or a method analogous thereto. rice field.
Figure JPOXMLDOC01-appb-T000036
Figure JPOXMLDOC01-appb-I000037
Figure JPOXMLDOC01-appb-I000038
Figure JPOXMLDOC01-appb-T000036
Figure JPOXMLDOC01-appb-I000037
Figure JPOXMLDOC01-appb-I000038
<参考例2>
Figure JPOXMLDOC01-appb-C000039
<Reference example 2>
Figure JPOXMLDOC01-appb-C000039
 窒素雰囲気下、3-メチル-1H-インドール-2-カルボン酸(1.1g)をジメチルホルムアミド(20mL)に溶解し、2-アミノ-4-シクロプロピルブタン酸エチル1塩酸塩(1.11g)、N,N-ジイソプロピルエチルアミン(1.18mL)を添加し氷冷した。ここへ1-[ビス(ジメチルアミノ)メチレン]-1H-1,2,3-トリアゾロ[4,5-b]ピリジニウム3-オキシドヘキサフルオロホスファート(2.62g)を加え、室温で15時間攪拌した。反応液に水、酢酸エチルを加えて攪拌した後、水層を分液して酢酸エチルで2回抽出した。合わせた有機層を水、飽和食塩水で洗浄し、硫酸ナトリウムで乾燥、ろ過後に濃縮した。残渣をシリカゲルカラム精製(ヘキサン-酢酸エチル)することで、4-シクロプロピル-2-(3-メチル-1H-インドール-2-カルボキサミド)ブタン酸エチル(1.27g,73%)を取得した。 Under a nitrogen atmosphere, 3-methyl-1H-indole-2-carboxylic acid (1.1 g) was dissolved in dimethylformamide (20 mL), ethyl 2-amino-4-cyclopropylbutanoate monohydrochloride (1.11 g). , N,N-diisopropylethylamine (1.18 mL) was added and cooled with ice. 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate (2.62 g) was added thereto and stirred at room temperature for 15 hours. did. Water and ethyl acetate were added to the reaction mixture and the mixture was stirred, and then the aqueous layer was separated and extracted twice with ethyl acetate. The combined organic layer was washed with water and saturated brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by silica gel column (hexane-ethyl acetate) to give ethyl 4-cyclopropyl-2-(3-methyl-1H-indole-2-carboxamide)butanoate (1.27 g, 73%).
1H-NMR (CHLOROFORM-D, 500 MHz) δ: -0.04 - 0.10 (2H, m), 0.44 (2H, d, J = 8.2 Hz), 0.65- 0.77 (1H, m), 1.20 - 1.43 (2H, m), 1.33 (3H, t, J = 7.2 Hz), 1.90 - 1.99 (1H, m), 2.08 - 2.19 (1H, m), 2.64 (3H, s), 4.20 - 4.33 (2H, m), 4.89 (1H, dt, J = 7.2, 7.0 Hz), 6.67 (1H, d, J = 7.7 Hz), 7.15 (1H, t, J = 7.7 Hz), 7.29 (1H, t, J = 7.6 Hz), 7.38 (1H, d, J = 8.3 Hz), 7.64 (1H, d, J = 8.3 Hz), 8.99 (1H, br s).
MS (ESI+): 329 [M+H]+
1 H-NMR (CHLOROFORM-D, 500 MHz) δ: -0.04 - 0.10 (2H, m), 0.44 (2H, d, J = 8.2 Hz), 0.65- 0.77 (1H, m), 1.20 - 1.43 (2H , m), 1.33 (3H, t, J = 7.2 Hz), 1.90 - 1.99 (1H, m), 2.08 - 2.19 (1H, m), 2.64 (3H, s), 4.20 - 4.33 (2H, m), 4.89 (1H, dt, J = 7.2, 7.0 Hz), 6.67 (1H, d, J = 7.7 Hz), 7.15 (1H, t, J = 7.7 Hz), 7.29 (1H, t, J = 7.6 Hz), 7.38 (1H, d, J = 8.3 Hz), 7.64 (1H, d, J = 8.3 Hz), 8.99 (1H, br s).
MS (ESI + ): 329 [M+H] +
Figure JPOXMLDOC01-appb-C000040
Figure JPOXMLDOC01-appb-C000040
 得られたエステル(1.22g)をエタノール(20mL)に溶解し、室温下で8N水酸化ナトリウム水溶液3mLを加えた。室温で15時間攪拌した後に、2N塩酸を加えて中和し、酢酸エチルで2回抽出した。合わせた有機層を飽和食塩水で洗浄した後、無水硫酸ナトリウムで乾燥、ろ過した後に溶媒を濃縮して4-シクロプロピル-2-(3-メチル-1H-インドール-2-カルボキサミド) ブタン酸(1.0g,90%)を取得した。 The obtained ester (1.22 g) was dissolved in ethanol (20 mL), and 3 mL of 8N aqueous sodium hydroxide solution was added at room temperature. After stirring at room temperature for 15 hours, the mixture was neutralized with 2N hydrochloric acid and extracted twice with ethyl acetate. The combined organic layer was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the solvent was concentrated to give 4-cyclopropyl-2-(3-methyl-1H-indole-2-carboxamide)butanoic acid ( 1.0 g, 90%) was obtained.
1H-NMR (CHLOROFORM-D, 500 MHz) δ: -0.09 - 0.08 (2H, m), 0.45 (2H, dd, J = 8.0, 1.6 Hz), 0.67 - 0.75 (1H, m), 1.28 - 1.46 (2H, m), 1.94 - 2.03 (1H, m), 2.16 - 2.24 (1H, m), 2.62 (3H, s), 4.89 (1H, q, J = 6.9 Hz), 6.56 (1H, d, J = 7.5 Hz), 7.14 (1H, t, J = 7.5 Hz),7.29 (1H, t, J = 7.5 Hz), 7.37 (1H, d, J = 8.0 Hz), 7.63 (1H, d, J = 8.2 Hz), 9.04 (1H, br s).
MS (ESI+): 301 [M+H]+
1 H-NMR (CHLOROFORM-D, 500 MHz) δ: -0.09 - 0.08 (2H, m), 0.45 (2H, dd, J = 8.0, 1.6 Hz), 0.67 - 0.75 (1H, m), 1.28 - 1.46 (2H, m), 1.94 - 2.03 (1H, m), 2.16 - 2.24 (1H, m), 2.62 (3H, s), 4.89 (1H, q, J = 6.9 Hz), 6.56 (1H, d, J = 7.5 Hz), 7.14 (1H, t, J = 7.5 Hz), 7.29 (1H, t, J = 7.5 Hz), 7.37 (1H, d, J = 8.0 Hz), 7.63 (1H, d, J = 8.2 Hz), 9.04 (1H, br s).
MS (ESI + ): 301 [M+H] +
<実施例2-1>
Figure JPOXMLDOC01-appb-C000041
<Example 2-1>
Figure JPOXMLDOC01-appb-C000041
 窒素雰囲気下、4-シクロプロピル-2-(3-メチル-1H-インドール-2-カルボキサミド)ブタン酸(50mg)、4-ジメチルアミノメチルベンジルアミン(47.4mg)およびN,N-ジイソプロピルエチルアミン(0.074mL)のジメチルホルムアミド溶液を氷冷した。ここへ1-[ビス(ジメチルアミノ)メチレン]-1H-1,2,3-トリアゾロ[4,5-b]ピリジニウム3-オキシドヘキサフルオロホスファート(82.3mg)を加え、室温で終夜攪拌した。反応液に水(5mL)を加えて水層を酢酸エチルで2回抽出した。有機層を水(10mL)で1回、飽和食塩水(10mL)で1回洗浄し、硫酸ナトリウムで乾燥した。ろ過・減圧濃縮した後に、残渣をシリカゲルカラム精製(Yamazen S;0-20%メタノールのジクロロメタン溶液)し、N-(4-シクロプロピル-1-((4-((ジメチルアミノ)メチル)ベンジル)アミノ)-1-オキソブタン-2-イル)-3-メチル-1H-インドール-2-カルボキサミド(43mg,58%)を取得した。 Under a nitrogen atmosphere, 4-cyclopropyl-2-(3-methyl-1H-indole-2-carboxamido)butanoic acid (50 mg), 4-dimethylaminomethylbenzylamine (47.4 mg) and N,N-diisopropylethylamine ( 0.074 mL) of dimethylformamide solution was ice-cooled. 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate (82.3 mg) was added thereto and stirred overnight at room temperature. . Water (5 mL) was added to the reaction mixture, and the aqueous layer was extracted twice with ethyl acetate. The organic layer was washed once with water (10 mL) and once with saturated brine (10 mL), and dried over sodium sulfate. After filtration and concentration under reduced pressure, the residue was purified by silica gel column (Yamazen S; 0-20% methanol in dichloromethane solution), N-(4-cyclopropyl-1-((4-((dimethylamino)methyl)benzyl) Amino)-1-oxobutan-2-yl)-3-methyl-1H-indole-2-carboxamide (43 mg, 58%) was obtained.
1H-NMR (CHLOROFORM-D, 500MHz) δ: -0.07 - 0.03 (2H, m), 0.38 - 0.42 (2H, m), 0.62 - 0.70 (1H, m), 1.21 - 1.40 (2H, m), 1.84 - 1.92 (1H, m), 2.07 - 2.15 (H, m), 2.22 (6H, s), 2.62 (3H, s), 3.38 (2H, s), 4.40 (1H, dd, J = 14.8, 5.6 Hz), 4.52 (1H, dd, J = 14.8, 6.0 Hz), 4.74 (1H, td, J = 7.5, 5.9 Hz), 6.60 (1H, t, J = 5.7 Hz), 6.92 (1H, d, J = 7.7 Hz), 7.14 (1H, ddd, J = 8.0, 6.8, 1.1 Hz), 7.19 - 7.31 (6H, m), 7.64 (1H, d, J = 7.9 Hz), 9.07 (1H, br s).
MS (ESI+): 447 [M+H]+
1 H-NMR (CHLOROFORM-D, 500MHz) δ: -0.07 - 0.03 (2H, m), 0.38 - 0.42 (2H, m), 0.62 - 0.70 (1H, m), 1.21 - 1.40 (2H, m), 1.84 - 1.92 (1H, m), 2.07 - 2.15 (H, m), 2.22 (6H, s), 2.62 (3H, s), 3.38 (2H, s), 4.40 (1H, dd, J = 14.8, 5.6 Hz), 4.52 (1H, dd, J = 14.8, 6.0 Hz), 4.74 (1H, td, J = 7.5, 5.9 Hz), 6.60 (1H, t, J = 5.7 Hz), 6.92 (1H, d, J = 7.7 Hz), 7.14 (1H, ddd, J = 8.0, 6.8, 1.1 Hz), 7.19 - 7.31 (6H, m), 7.64 (1H, d, J = 7.9 Hz), 9.07 (1H, br s).
MS (ESI + ): 447 [M+H] +
 対応する出発物質および反応剤を用い、実施例2-1と同様の方法、工程2-3に記した方法、またはそれらに準じた方法により、以下の実施例2-2から2-25を得た。 Using the corresponding starting materials and reactants, the following Examples 2-2 to 2-25 are obtained by the same method as in Example 2-1, the method described in Step 2-3, or a method analogous thereto. rice field.
Figure JPOXMLDOC01-appb-T000042
Figure JPOXMLDOC01-appb-I000043
Figure JPOXMLDOC01-appb-I000044
Figure JPOXMLDOC01-appb-I000045
Figure JPOXMLDOC01-appb-T000042
Figure JPOXMLDOC01-appb-I000043
Figure JPOXMLDOC01-appb-I000044
Figure JPOXMLDOC01-appb-I000045
<参考例3-1>
Figure JPOXMLDOC01-appb-C000046
<Reference example 3-1>
Figure JPOXMLDOC01-appb-C000046
 窒素雰囲気下、3-ホルミルベンゾニトリル(1.0g)、Boc-ピぺラジン(1.49g)および酢酸(0.53mL)のジクロロメタン溶液に、トリアセトキシ水素化ホウ素ナトリウム(2.42g)を室温で加えた。18時間室温で攪拌したのち、水20mLで希釈し、pHを9に調整したのちに、ジクロロメタンで3回抽出した。合わせた有機層を無水硫酸マグネシウムで乾燥後、ろ過、減圧濃縮した。残渣をカラムクロマトグラフィー(ヘキサン:酢酸エチル=10:0~7:3)で精製してtert-ブチル 4-(3-シアノベンジル)ピぺラジン-1-カルボキシレートを取得した。 Under a nitrogen atmosphere, sodium triacetoxyborohydride (2.42 g) was added to a dichloromethane solution of 3-formylbenzonitrile (1.0 g), Boc-piperazine (1.49 g) and acetic acid (0.53 mL) at room temperature. added with After stirring for 18 hours at room temperature, the mixture was diluted with 20 mL of water, adjusted to pH 9, and extracted three times with dichloromethane. The combined organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (hexane:ethyl acetate=10:0-7:3) to give tert-butyl 4-(3-cyanobenzyl)piperazine-1-carboxylate.
1H-NMR (CHLOROFORM-D, 500MHz) δ: 1.46 (9H, s), 2.35 - 2.41 (4H, m), 3.41 - 3.46 (4H, m), 3.53 (2H, s), 7.43 (1H, t, J = 7.7 Hz), 7.53 - 7.58 (2H, m), 7.66 (1H, s).
MS (FI+): 302 [M+H]+
1 H-NMR (CHLOROFORM-D, 500MHz) δ: 1.46 (9H, s), 2.35 - 2.41 (4H, m), 3.41 - 3.46 (4H, m), 3.53 (2H, s), 7.43 (1H, t , J = 7.7 Hz), 7.53 - 7.58 (2H, m), 7.66 (1H, s).
MS (FI + ): 302 [M+H] +
 窒素雰囲気下、tert-ブチル 4-(3-シアノベンジル)ピぺラジン-1-カルボキシレート(200mg)、塩化ニッケル(II)6水和物(15.8mg)のメタノール溶液(5mL)に、水素化ホウ素ナトリウム(126mg)を30分かけて添加した。反応液を終夜で攪拌したのちに、1N水酸化ナトリウム水溶液、クロロホルムを加えて攪拌した。水層を分液し、2回クロロホルムで抽出した。合わせた有機層を無水硫酸ナトリウムで乾燥後、ろ過、減圧濃縮した。残渣をカラムクロマトグラフィー(クロロホルム:メタノール=10:0~6:4)で精製してtert-ブチル 4-(3-(アミノメチル)ベンジル)ピぺラジン-1-カルボキシレートを得た。 Under a nitrogen atmosphere, hydrogen Sodium borohydride (126 mg) was added over 30 minutes. After the reaction mixture was stirred overnight, 1N sodium hydroxide aqueous solution and chloroform were added and stirred. The aqueous layer was separated and extracted twice with chloroform. The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (chloroform:methanol=10:0-6:4) to give tert-butyl 4-(3-(aminomethyl)benzyl)piperazine-1-carboxylate.
1H-NMR (CHLOROFORM-D, 500MHz) δ: 1.45 (9H, s), 2.32 - 2.44 (4H, m), 3.38 - 3.46 (4H, m), 3.50 (2H, s), 3.86 (2H, s), 7.17 - 7.24 (2H, m), 7.22 - 7.33 (2H, m).MS (ESI+): 306 [M+H]+ 1 H-NMR (CHLOROFORM-D, 500MHz) δ: 1.45 (9H, s), 2.32 - 2.44 (4H, m), 3.38 - 3.46 (4H, m), 3.50 (2H, s), 3.86 (2H, s ), 7.17 - 7.24 (2H, m), 7.22 - 7.33 (2H, m).MS (ESI + ): 306 [M+H] +
 対応する出発物質および反応剤を用い、参考例3-1と同様の方法、またはそれに準じた方法により、以下の参考例3-2を得た。 Using the corresponding starting materials and reactants, the following Reference Example 3-2 was obtained by the same method as Reference Example 3-1 or a method according thereto.
Figure JPOXMLDOC01-appb-T000047
Figure JPOXMLDOC01-appb-T000047
<実施例3-1>
Figure JPOXMLDOC01-appb-C000048
<Example 3-1>
Figure JPOXMLDOC01-appb-C000048
 参考例3-1で得られた出発物質を用いて、実施例2-1と同様の方法、工程2-3に記した方法、またはそれらに準じた方法により、tert-ブチル 4-(3-((4-シクロプロピル-2-(3-メチル-1H-インドール-2-カルボキサミド)ブタンアミド)メチル)ベンジル)ピぺラジン-1-カルボキシレートを取得した。 Using the starting material obtained in Reference Example 3-1, tert-butyl 4-(3- ((4-Cyclopropyl-2-(3-methyl-1H-indole-2-carboxamido)butanamido)methyl)benzyl)piperazine-1-carboxylate was obtained.
 このBoc保護体(44mg)のメタノール溶液に、4N塩酸/ジオキサン(1.5mL)を加えて室温で2時間撹拌した。反応溶液を減圧濃縮した。固化した残渣にクロロホルムと飽和炭酸水素ナトリウム溶液を加え、水層を分離して3回クロロホルムで抽出した。合わせた有機層を水、飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥後、ろ過・減圧濃縮することで、34mg(quant.)のN-(4-シクロプロピル-1-オキソ-((3-((ピぺラジン-1-イルメチル)ベンジル)アミノ)ブタン-2-イル)-3-メチル-1H-インドール-2-カルボキサミドを取得した。 4N hydrochloric acid/dioxane (1.5 mL) was added to the methanol solution of this Boc-protected form (44 mg), and the mixture was stirred at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure. Chloroform and a saturated sodium bicarbonate solution were added to the solidified residue, and the aqueous layer was separated and extracted with chloroform three times. The combined organic layer was washed with water and saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 34 mg (quant.) of N-(4-cyclopropyl-1-oxo-((3 -((Piperazin-1-ylmethyl)benzyl)amino)butan-2-yl)-3-methyl-1H-indole-2-carboxamide was obtained.
1H-NMR (CHLOROFORM-D, 500MHz) δ: -0.11 - -0.03 (2H, m), 0.31 - 0.38 (2H, m), 0.57 - 0.65 (1H, m), 1.19 - 1.35 (2H, m), 1.83 - 1.92(1H, m), 2.00 - 2.09 (1H, m),  2.23 - 2.33 (4H, m), 2.58 (3H, s), 2.75 - 2.81(4H, m), 3.34 (2H, d, J = 2.6 Hz), 4.38 (1H, dd, J = 15.0, 5.6 Hz), 4.47 (1H, dd, J = 15.0, 5.9 Hz), 4.79 - 4.86 (1H, m), 5.29 (1H, br s), 7.08 - 7.25 (7H, m), 7.35 - 7.43 (2H, m), 7.61 (1H, dd, J = 8.0, 1.1 Hz), 9.74 (1H, br s).
MS (ESI+): 488 [M+H]+
1 H-NMR (CHLOROFORM-D, 500MHz) δ: -0.11 - -0.03 (2H, m), 0.31 - 0.38 (2H, m), 0.57 - 0.65 (1H, m), 1.19 - 1.35 (2H, m) , 1.83 - 1.92 (1H, m), 2.00 - 2.09 (1H, m), 2.23 - 2.33 (4H, m), 2.58 (3H, s), 2.75 - 2.81 (4H, m), 3.34 (2H, d, J = 2.6 Hz), 4.38 (1H, dd, J = 15.0, 5.6 Hz), 4.47 (1H, dd, J = 15.0, 5.9 Hz), 4.79 - 4.86 (1H, m), 5.29 (1H, br s) , 7.08 - 7.25 (7H, m), 7.35 - 7.43 (2H, m), 7.61 (1H, dd, J = 8.0, 1.1 Hz), 9.74 (1H, br s).
MS (ESI + ): 488 [M+H] +
 対応する出発物質および反応剤を用い、実施例3-1と同様の方法、工程2-3に記した方法、またはそれらに準じた方法により、以下の実施例3-2、3-3を得た。 Using the corresponding starting materials and reactants, the following Examples 3-2 and 3-3 are obtained by the same method as in Example 3-1, the method described in Step 2-3, or a method analogous thereto. rice field.
Figure JPOXMLDOC01-appb-T000049
Figure JPOXMLDOC01-appb-T000049
<参考例4-1>
Figure JPOXMLDOC01-appb-C000050
<Reference example 4-1>
Figure JPOXMLDOC01-appb-C000050
 窒素雰囲気下、tert-ブチル(4-ヒドロキシベンジル)カルバメート(515mg)、ベンジル 4-ヒドロキシピペリジン-1-カルボキシレート(595mg)、トリフェニルホスフィン(726mg)のテトラヒドロフラン溶液(10mL)に、アゾジカルボン酸ビス(2-メトキシエチル)(648mg)を室温で加えて、20時間攪拌した。水、酢酸エチルを加え分液し、水層を1回酢酸エチルで抽出した。合わせた有機層を水で2回、飽和食塩水で1回洗浄し、無水硫酸ナトリウムで乾燥した後に、ろ過・減圧濃縮した。残渣をシリカゲルカラムクロマトグラフィー(酢酸エチル:ヘキサン=9:1~2:1)で精製し、ベンジル 4-(4-(((tert-ブトキシカルボニル)アミノ)メチル)フェノキシ)ピぺリジン-1-カルボキシレート(659mg、65%)を取得した。 Under a nitrogen atmosphere, tert-butyl (4-hydroxybenzyl) carbamate (515 mg), benzyl 4-hydroxypiperidine-1-carboxylate (595 mg), triphenylphosphine (726 mg) in tetrahydrofuran solution (10 mL), bis azodicarboxylate (2-Methoxyethyl) (648 mg) was added at room temperature and stirred for 20 hours. Water and ethyl acetate were added to separate the layers, and the aqueous layer was extracted once with ethyl acetate. The combined organic layer was washed twice with water and once with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (ethyl acetate:hexane=9:1-2:1), and benzyl 4-(4-(((tert-butoxycarbonyl)amino)methyl)phenoxy)piperidine-1- Carboxylate (659 mg, 65%) was obtained.
 得られたBoc保護体(138mg)のエタノール溶液(1mL)に、4M塩酸/ジオキサン(2mL)を加えて室温で2時間撹拌した。反応溶液を減圧濃縮し、固化したベンジル 4-(4-(アミノメチル)フェノキシ)ピぺリジン-1-カルボキシレート・塩酸塩(120mg,quant.)を取得した。
MS (ESI+):341 [M+H]+
4M hydrochloric acid/dioxane (2 mL) was added to an ethanol solution (1 mL) of the obtained Boc-protected compound (138 mg), and the mixture was stirred at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure to obtain solidified benzyl 4-(4-(aminomethyl)phenoxy)piperidine-1-carboxylate.hydrochloride (120 mg, quant.).
MS (ESI + ): 341 [M+H] +
 対応する出発物質および反応剤を用い、参考例4-1と同様の方法、またはそれに準じた方法により、以下の参考例4-2から4-3を得た。 Using the corresponding starting materials and reactants, the following Reference Examples 4-2 to 4-3 were obtained by the same method as Reference Example 4-1 or a method according thereto.
Figure JPOXMLDOC01-appb-T000051
Figure JPOXMLDOC01-appb-T000051
<実施例4-1>
Figure JPOXMLDOC01-appb-C000052
<Example 4-1>
Figure JPOXMLDOC01-appb-C000052
 参考例4-1で得られた出発物質を用いて、実施例2-1と同様の方法、工程2-3に記した方法、またはそれらに準じた方法により、ベンジル 4-(4-((4-シクロプロピル-2-(3-メチル-1H-インドール-2-カルボキサミド)ブタンアミド)メチル)ベンジル)ピぺラジン-1-カルボキシレート76mg(46%)を取得した。 Benzyl 4-(4-(( 76 mg (46%) of 4-cyclopropyl-2-(3-methyl-1H-indole-2-carboxamido)butanamido)methyl)benzyl)piperazine-1-carboxylate were obtained.
 得られたCbz体(76mg)および50%含水5%パラジウム炭素(20mg)のメタノール(5mL)溶液を、中圧還元容器に入れて水素雰囲気下に5時間、接触還元を行った。反応液をセライトでろ過した後に、減圧濃縮した。残渣をアミノシリカカラムクロマトグラフィー(クロロホルム:メタノール=10:0~9:1)で精製し、34mg(57%)のN-(4-シクロプロピル-1-((4-(ピぺリジン-4-イルオキシ)ベンジル)アミノ)-1-オキソブタン-2-イル)-3-メチル-1H-インドール-2-カルボキサミドを取得した。 A methanol (5 mL) solution of the obtained Cbz derivative (76 mg) and 50% hydrous 5% palladium carbon (20 mg) was placed in a medium-pressure reduction vessel and subjected to catalytic reduction under a hydrogen atmosphere for 5 hours. The reaction solution was filtered through celite and then concentrated under reduced pressure. The residue was purified by aminosilica column chromatography (chloroform:methanol=10:0 to 9:1) and 34 mg (57%) of N-(4-cyclopropyl-1-((4-(piperidine-4 -yloxy)benzyl)amino)-1-oxobutan-2-yl)-3-methyl-1H-indole-2-carboxamide is obtained.
1H-NMR (CHLOROFORM-D, 500MHz) δ: -0.05 - 0.05 (2H, m), 0.42 (2H, d, J = 7.7 Hz), 0.63 - 0.71 (1H, m), 1.22 - 1.40 (2H, m), 1.52 - 1.68 (3H, m), 1.84 - 1.92 (1H, m), 1.95 - 2.02 (2H, m), 2.08 - 2.16 (1H, m), 2.63 (3H, s), 2.67 - 2.74 (2H, m), 3.09 - 3.16 (2H, m), 4.30 - 4.40 (2H, m), 4.45 (1H, dd, J = 14.6, 6.0 Hz), 4.69 (1H, td, J = 6.8, 7.3 Hz), 6.33 (1H, t, J = 5.3 Hz), 6.79 (1H, d, J = 7.4 Hz), 6.86 (2H, d, J = 8.6 Hz), 7.15 (1H, t, J = 7.6 Hz), 7.18 (2H, d, J = 8.6 Hz), 7.29 (1H, t, J = 7.6 Hz), 7.36 (1H, d, J = 8.3 Hz), 7.64 (1H, d, J = 8.3 Hz), 8.94 (1H, br s).
MS (ESI+): 489 [M+H]+
1 H-NMR (CHLOROFORM-D, 500MHz) δ: -0.05 - 0.05 (2H, m), 0.42 (2H, d, J = 7.7 Hz), 0.63 - 0.71 (1H, m), 1.22 - 1.40 (2H, m), 1.52 - 1.68 (3H, m), 1.84 - 1.92 (1H, m), 1.95 - 2.02 (2H, m), 2.08 - 2.16 (1H, m), 2.63 (3H, s), 2.67 - 2.74 ( 2H, m), 3.09 - 3.16 (2H, m), 4.30 - 4.40 (2H, m), 4.45 (1H, dd, J = 14.6, 6.0 Hz), 4.69 (1H, td, J = 6.8, 7.3 Hz) , 6.33 (1H, t, J = 5.3 Hz), 6.79 (1H, d, J = 7.4 Hz), 6.86 (2H, d, J = 8.6 Hz), 7.15 (1H, t, J = 7.6 Hz), 7.18 (2H, d, J = 8.6 Hz), 7.29 (1H, t, J = 7.6 Hz), 7.36 (1H, d, J = 8.3 Hz), 7.64 (1H, d, J = 8.3 Hz), 8.94 (1H , br s).
MS (ESI + ): 489 [M+H] +
 対応する出発物質および反応剤を用い、実施例4-1と同様の方法、工程2-3に記した方法、またはそれらに準じた方法により、以下の実施例4-2から4-3を得た。 Using the corresponding starting materials and reactants, the following Examples 4-2 to 4-3 are obtained by the same method as in Example 4-1, the method described in Step 2-3, or a method analogous thereto. rice field.
Figure JPOXMLDOC01-appb-T000053
Figure JPOXMLDOC01-appb-T000053
<参考例5-1>
Figure JPOXMLDOC01-appb-C000054
<Reference example 5-1>
Figure JPOXMLDOC01-appb-C000054
 窒素雰囲気下、S-2-((tert-ブトキシカルボニル)アミノ)-4-シクロプロピルブタン酸(480mg)をジメチルホルムアミド(5mL)に溶解し、4-(ジメチルアミノ)メチルベンジルアミン(515mg)、N,N-ジイソプロピルエチルアミン(0.44mL)を添加し氷冷した。ここへ1-[ビス(ジメチルアミノ)メチレン]-1H-1,2,3-トリアゾロ[4,5-b]ピリジニウム3-オキシドヘキサフルオロホスファート(975mg)を加え、室温で終夜攪拌した。反応液に飽和食塩水(10mL)を加えて水層を酢酸エチル(10mL×2)で抽出した。有機層を水(10mL)で1回、飽和食塩水(10mL)で1回洗浄し、硫酸ナトリウムで乾燥した。濃縮した残渣をシリカゲルカラム精製(Yamazen M;0-20%メタノールのジクロロメタン溶液)し、濃縮することで、255mg(33.2%)のS-tert-ブチル(4-シクロプロピル-1-((4-((ジメチルアミノ)メチル)ベンジル)アミノ)-1-オキソブタ-2-ニル)カルバメートを取得した。 Under a nitrogen atmosphere, S-2-((tert-butoxycarbonyl)amino)-4-cyclopropylbutanoic acid (480 mg) was dissolved in dimethylformamide (5 mL), 4-(dimethylamino)methylbenzylamine (515 mg), N,N-diisopropylethylamine (0.44 mL) was added and ice-cooled. 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate (975 mg) was added thereto, and the mixture was stirred overnight at room temperature. Saturated saline (10 mL) was added to the reaction solution, and the aqueous layer was extracted with ethyl acetate (10 mL×2). The organic layer was washed once with water (10 mL) and once with saturated brine (10 mL), and dried over sodium sulfate. The concentrated residue was purified by silica gel column (Yamazen M; 0-20% methanol in dichloromethane) and concentrated to give 255 mg (33.2%) of S-tert-butyl (4-cyclopropyl-1-(( 4-((dimethylamino)methyl)benzyl)amino)-1-oxobut-2-yl)carbamate was obtained.
 tert-ブチル(4-シクロプロピル-1-((4-((ジメチルアミノ)メチル)ベンジル)アミノ)-1-オキソブタ-2-ニル)カルバメート(255mg)に4M塩酸/ジオキサン(8mL)を加えて室温で1時間撹拌した。放冷後、反応溶液を減圧濃縮し、酢酸エチルで3回共沸した。固化した固体を酢酸エチルで懸濁洗浄することで、2.00g(quant.)のS-2-アミノ-4-シクロプロピル-N-(4-((ジメチルアミノ)メチル)ベンジル)ブタンアミド・2塩酸塩を取得した。 4M hydrochloric acid/dioxane (8 mL) was added to tert-butyl (4-cyclopropyl-1-((4-((dimethylamino)methyl)benzyl)amino)-1-oxobut-2-yl)carbamate (255 mg). Stir at room temperature for 1 hour. After allowing to cool, the reaction solution was concentrated under reduced pressure and azeotroped three times with ethyl acetate. By suspending and washing the solidified solid with ethyl acetate, 2.00 g (quant.) of S-2-amino-4-cyclopropyl-N-(4-((dimethylamino)methyl)benzyl)butanamide・2 Hydrochloride salt was obtained.
1H-NMR (DMSO-D6, 500MHz) δ: -0.06 - 0.01 (2H, m), 0.33 - 0.42 (2H, m), 0.61 - 0.70 (1H, m), 1.13 - 1.21 (2H, m), 1.81 - 1.89 (2H, m), 2.63 (3H, s), 2.64 (3H, s), 3.80 - 3.89 (1H, m), 4.24 (2H, d, J = 6.5 Hz), 4.36 (2H, d, J = 7.5 Hz), 7.36 (2H, d, J = 10.0 Hz), 7.56 (2H, d, J = 10.0 Hz), 8.36 (3H, d, J = 4.5 Hz), 9.26 (1H, t, J = 7.5 Hz), 11.19 (1H, br s).
MS (ESI+): 290 [M+H]+
1 H-NMR (DMSO-D6, 500MHz) δ: -0.06 - 0.01 (2H, m), 0.33 - 0.42 (2H, m), 0.61 - 0.70 (1H, m), 1.13 - 1.21 (2H, m), 1.81 - 1.89 (2H, m), 2.63 (3H, s), 2.64 (3H, s), 3.80 - 3.89 (1H, m), 4.24 (2H, d, J = 6.5 Hz), 4.36 (2H, d, J = 7.5 Hz), 7.36 (2H, d, J = 10.0 Hz), 7.56 (2H, d, J = 10.0 Hz), 8.36 (3H, d, J = 4.5 Hz), 9.26 (1H, t, J = 7.5 Hz), 11.19 (1H, br s).
MS (ESI + ): 290 [M+H] +
 対応する出発物質および反応剤を用い、参考例5-1と同様の方法、工程1-1、2に記した方法、またはそれらに準じた方法により、以下の参考例5-2から5-18を得た。 Using the corresponding starting materials and reactants, the following Reference Examples 5-2 to 5-18 were prepared by the same method as in Reference Example 5-1, the method described in Steps 1-1 and 1-2, or a method analogous thereto. got
Figure JPOXMLDOC01-appb-T000055
Figure JPOXMLDOC01-appb-I000056
Figure JPOXMLDOC01-appb-T000055
Figure JPOXMLDOC01-appb-I000056
<実施例5-1>
Figure JPOXMLDOC01-appb-C000057
<Example 5-1>
Figure JPOXMLDOC01-appb-C000057
 窒素雰囲気下、3-メチル-1H-インドール-2-カルボン酸(58mg)をジメチルホルムアミド(5.0mL)に溶解し、S-2-アミノ-4-シクロプロピル-N-(4-((ジメチルアミノ)メチル)ベンジル)ブタンアミド 2塩酸塩(100mg)、N,N-ジイソプロピルエチルアミン(0.156mL)を添加し氷冷した。ここへ1-[ビス(ジメチルアミノ)メチレン]-1H-1,2,3-トリアゾロ[4,5-b]ピリジニウム 3-オキシドヘキサフルオロホスファート(136mg)を加え、室温で終夜攪拌した。反応液に飽和食塩水(5mL)を加えて水層を酢酸エチル(10mL×2)で抽出した。有機層を水(10mL)で1回、飽和食塩水(10mL)で1回洗浄し、硫酸ナトリウムで乾燥した。濃縮した残渣をシリカゲルカラム精製(Yamazen S;0-20%メタノールのジクロロメタン溶液)し、濃縮することで、41mg(33.2%)のS-N-(4-シクロプロピル-1-((4-((ジメチルアミノ)メチル)ベンジル)アミノ)-1-オキソブタン-2-イル)-3-メチル-1H-インドール-2-カルボキサミドを取得した。 Under a nitrogen atmosphere, 3-methyl-1H-indole-2-carboxylic acid (58 mg) was dissolved in dimethylformamide (5.0 mL), and S-2-amino-4-cyclopropyl-N-(4-((dimethyl Amino)methyl)benzyl)butanamide dihydrochloride (100 mg) and N,N-diisopropylethylamine (0.156 mL) were added and ice-cooled. 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate (136 mg) was added thereto, and the mixture was stirred overnight at room temperature. Saturated saline (5 mL) was added to the reaction solution, and the aqueous layer was extracted with ethyl acetate (10 mL×2). The organic layer was washed once with water (10 mL) and once with saturated brine (10 mL), and dried over sodium sulfate. The concentrated residue was purified by silica gel column (Yamazen S; 0-20% methanol in dichloromethane) and concentrated to give 41 mg (33.2%) of SN-(4-cyclopropyl-1-((4 -((dimethylamino)methyl)benzyl)amino)-1-oxobutan-2-yl)-3-methyl-1H-indole-2-carboxamide was obtained.
1H-NMR (CHLOROFORM-D, 500MHz) δ: -0.07 - 0.03 (2H, m), 0.38 - 0.42 (2H, m), 0.62 - 0.70 (1H, m), 1.21 - 1.40 (2H, m), 1.84 - 1.92 (1H, m), 2.07 - 2.15 (1H, m), 2.22 (6H, s), 2.62 (3H, s), 3.38 (2H, s), 4.40 (1H, dd, J = 14.8, 5.6 Hz), 4.52 (1H, dd, J = 14.8, 6.0 Hz), 4.74 (1H, td, J = 7.5, 5.9 Hz), 6.60 (1H, t, J = 5.7 Hz), 6.92 (1H, d, J = 7.7 Hz), 7.14 (1H, ddd, J = 8.0, 6.8, 1.1 Hz), 7.19 - 7.31 (6H, m), 7.64 (1H, d, J = 7.9 Hz), 9.07 (1H, br s).
MS (ESI+): 447 [M+H]+
1 H-NMR (CHLOROFORM-D, 500MHz) δ: -0.07 - 0.03 (2H, m), 0.38 - 0.42 (2H, m), 0.62 - 0.70 (1H, m), 1.21 - 1.40 (2H, m), 1.84 - 1.92 (1H, m), 2.07 - 2.15 (1H, m), 2.22 (6H, s), 2.62 (3H, s), 3.38 (2H, s), 4.40 (1H, dd, J = 14.8, 5.6 Hz), 4.52 (1H, dd, J = 14.8, 6.0 Hz), 4.74 (1H, td, J = 7.5, 5.9 Hz), 6.60 (1H, t, J = 5.7 Hz), 6.92 (1H, d, J = 7.7 Hz), 7.14 (1H, ddd, J = 8.0, 6.8, 1.1 Hz), 7.19 - 7.31 (6H, m), 7.64 (1H, d, J = 7.9 Hz), 9.07 (1H, br s).
MS (ESI + ): 447 [M+H] +
 対応する出発物質および反応剤を用い、実施例5-1と同様の方法、工程1-3に記した方法、またはそれらに準じた方法により、以下の実施例5-2から5-31を得た。 Using the corresponding starting materials and reactants, the following Examples 5-2 to 5-31 are obtained by the same method as in Example 5-1, the method described in Step 1-3, or a method analogous thereto. rice field.
Figure JPOXMLDOC01-appb-T000058
Figure JPOXMLDOC01-appb-I000059
Figure JPOXMLDOC01-appb-I000060
Figure JPOXMLDOC01-appb-I000061
Figure JPOXMLDOC01-appb-I000062
Figure JPOXMLDOC01-appb-T000058
Figure JPOXMLDOC01-appb-I000059
Figure JPOXMLDOC01-appb-I000060
Figure JPOXMLDOC01-appb-I000061
Figure JPOXMLDOC01-appb-I000062
<試験例1>
G9a阻害活性試験
 G9aの酵素活性をAmplified Luminescence Proximity Homogeneous Assay(ALPHA)で測定することにより、化合物のG9a阻害活性を評価した。先ず、Tris緩衝液(50mM Tris-HCl[pH 9.0],50mM NaCl,0.01% Tween-20,1mM DTT)で希釈したリコンビナントなヒトG9aタンパク質(Active Motif,#31410)(終濃度0.025nM)7.5μLと被験化合物0.5μLを384穴マイクロプレート(AlphaPlate-384 Shallow Well,PerkinElmer,#6008359)に添加し、ボルテックスして混和させた後に室温で10分静置した。ビオチン化ヒストンH3ペプチド(1-21)(AnaSpec,#61702)500nMとSAM(SIGMA,#A7007)150μMを1:1であらかじめ混ぜ、各ウェルに2μL添加し、ボルテックスで混和させた後、室温で1時間静置した。AlphaLISA anti-H3K9me2 acceptor beads(PerkinElmer,#AL117M)とAlphaScreen streptavidin donor beads(PerkinElmer,#6760002)をepigenetic buffer(PerkinElmer,AlphaLISA Epigenetics Buffer Kit #AL008C)で添加後の最終濃度が10μg/mlになるようにそれぞれ希釈し、遮光下で添加した後、室温で1時間遮光静置した。その後、EnSpire Alphaプレートリーダー(PerkinElmer,Waltham,MA,USA)を用いて測定した。被験化合物のG9a阻害活性は、化合物を添加しないコントロールの値を0%、酵素を添加しないコントロールの値を100%としたときの化合物の阻害率の割合を求め、G9a酵素活性を50%に抑制するのに必要な化合物濃度(IC50値)を算出した。結果を以下に示す。
 なお、表中、酵素阻害活性は、IC50(μM)=<5,>1=+;<1,>0.1=++;<0.1,>0.02=+++;<0.02=++++として表記した。
<Test Example 1>
G9a Inhibitory Activity Test The G9a inhibitory activity of the compounds was evaluated by measuring the G9a enzymatic activity by Amplified Luminescence Proximity Homogeneous Assay (ALPHA). First, recombinant human G9a protein (Active Motif, #31410) diluted with Tris buffer (50 mM Tris-HCl [pH 9.0], 50 mM NaCl, 0.01% Tween-20, 1 mM DTT) (final concentration 0 025 nM) and 0.5 μL of the test compound were added to a 384-well microplate (AlphaPlate-384 Shallow Well, PerkinElmer, #6008359), mixed by vortexing, and allowed to stand at room temperature for 10 minutes. 500 nM of biotinylated histone H3 peptide (1-21) (AnaSpec, #61702) and 150 μM of SAM (SIGMA, #A7007) were premixed 1:1, 2 μL was added to each well, mixed by vortexing, and left at room temperature. It was allowed to stand for 1 hour. AlphaLISA anti-H3K9me2 acceptor beads(PerkinElmer,#AL117M)とAlphaScreen streptavidin donor beads(PerkinElmer,#6760002)をepigenetic buffer(PerkinElmer,AlphaLISA Epigenetics Buffer Kit #AL008C)で添加後の最終濃度が10μg/mlになるようにEach solution was diluted, added in the dark, and then allowed to stand at room temperature for 1 hour. Measurements were then taken using an EnSpire Alpha plate reader (PerkinElmer, Waltham, Mass., USA). For the G9a inhibitory activity of the test compound, the inhibitory rate of the compound is calculated based on the value of the control without the addition of the compound as 0% and the value of the control without the addition of the enzyme as 100%, and the G9a enzyme activity is inhibited to 50%. The concentration of compound (IC 50 value) required to achieve this was calculated. The results are shown below.
In the table, the enzyme inhibitory activity is IC 50 (μM) = <5, >1 = +; <1, >0.1 = ++; <0.1, >0.02 = +++; <0.02 =++++.
Figure JPOXMLDOC01-appb-T000063
Figure JPOXMLDOC01-appb-I000064
Figure JPOXMLDOC01-appb-I000065
Figure JPOXMLDOC01-appb-T000063
Figure JPOXMLDOC01-appb-I000064
Figure JPOXMLDOC01-appb-I000065
<試験例2>
グロビン遺伝子発現試験
 得られた化合物のヒト赤芽球細胞株HUDEP―2に対する胎児型グロビン遺伝子発現量への効果を定量PCRにより評価した。HUDEP―2細胞は1μMデキサメタゾン(Sigma)、1μg/mlドキシサイクリン(Sigma)、50ng/mlリコンビナントヒトSCF(R&D SYSTEMS)、3IU/mLエポエチンアルファ(東邦薬品)を含むStemlineII造血幹細胞増殖培地(Sigma)で培養した。培養細胞を細胞培養用培地で1×10cells/mLになるように細胞液を調製した。次いで、細胞液を6穴プレート(VIOLAMO)に2mL/wellずつ播種し、試験化合物(DMSO溶液)を2μL/well(DMSO終濃度0.1%)添加し、37℃、5%CO条件下で4日間培養した。コントロールとしてDMSO溶液を2μL/well添加した。培養細胞を7500rpm、4℃、5分間の遠心にて回収し、Tissue Total RNA Mini Kit(チヨダサイエンス)を使用してRNAを抽出した。その後、ReverTra Ace qPCR RT Master Mix(TOYOBO)を用いて37℃、15分間の後、50℃、5分間の逆転写反応でcDNA合成した。希釈したcDNAを鋳型にしてTHUNDERBIRD SYBR qPCR(TOYOBO)と混合し、CFX Connect Real-Time PCR Detection System(BIORAD)を用いて、胎児型β-グロビン遺伝子、成人型β-グロビン遺伝子、リファレンスとしてGAPDH遺伝子を測定した。使用した遺伝子のプライマーは以下である。胎児型β-グロビン Forwaord:5’―TGGATGATCTCAAGGGCAC―3’;Reverse:5’―TCAGTGGTATCTGGAGGACA―3’、成人型β-グロビン Forwaord:5’―CAGTGCAGGCTGCCTATC―3’;Reverse:5’―ATACTTGTGGGCCAGGGCAT―3’、GAPDH Forwaord:5’―GCACCGTCAAGGCTGAGAAC―3’;Reverse:5’―TGGTGAAGACGCCAGTGGA―3’。遺伝子発現解析は0.1%DMSO溶液を添加したコントロールを1とし、ΔΔCt法にて比較した。
 なお、表中、発現量比は、DMSOのみ(1.0)に対してその比が<1.25=+;>1.25,<1.5=++;>1.5=+++として表記した。
<Test Example 2>
Globin Gene Expression Test The effect of the obtained compounds on the expression level of fetal globin gene in human erythroid cell line HUDEP-2 was evaluated by quantitative PCR. HUDEP-2 cells were grown in Stemline II hematopoietic stem cell growth medium (Sigma) containing 1 μM dexamethasone (Sigma), 1 μg/ml doxycycline (Sigma), 50 ng/ml recombinant human SCF (R&D SYSTEMS), and 3 IU/mL epoetin alfa (Toho Yakuhin). cultured. A cell solution was prepared from the cultured cells in a cell culture medium so that the concentration of the cultured cells was 1×10 5 cells/mL. Then, the cell solution was seeded in a 6-well plate (VIOLAMO) at 2 mL/well, and the test compound (DMSO solution) was added at 2 μL/well (DMSO final concentration 0.1%), 37° C., 5% CO 2 conditions. was cultured for 4 days. A DMSO solution was added at 2 μL/well as a control. The cultured cells were collected by centrifugation at 7500 rpm, 4° C. for 5 minutes, and RNA was extracted using Tissue Total RNA Mini Kit (Chiyoda Science). Thereafter, using ReverTra Ace qPCR RT Master Mix (TOYOBO), cDNA was synthesized by reverse transcription at 37°C for 15 minutes and then at 50°C for 5 minutes. Using the diluted cDNA as a template, it was mixed with THUNDERBIRD SYBR qPCR (TOYOBO), and the CFX Connect Real-Time PCR Detection System (BIORAD) was used to detect the fetal β-globin gene, the adult β-globin gene, and the GAPDH gene as a reference. was measured. The following gene primers were used. Fetal β-globin Forward: 5′-TGGATGATCTCAAGGGCAC-3′; Reverse: 5′-TCAGTGGTATCTGGAGGACA-3′, Adult β-globin Forward: 5′-CAGTGCAGGCTGCCTATC-3′; Reverse: 5′-ATACTTGTGGGCCAGGGCAT-3′, GAPDH Forward: 5'-GCACCGTCAAGGCTGAGAAC-3'; Reverse: 5'-TGGTGAAGACGCCAGTGGA-3'. Gene expression analysis was performed using the ΔΔCt method, with the control to which 0.1% DMSO solution was added as 1.
In the table, the expression level ratio is expressed as <1.25=+;>1.25, <1.5=++;>1.5=+++ relative to DMSO only (1.0). did.
Figure JPOXMLDOC01-appb-T000066
Figure JPOXMLDOC01-appb-T000066
<試験例3>
P-gp親和性試験
 以下の方法で、実施例で合成した化合物について、P-gp基質性を評価したところ良好であり、例えば、PCT/JP2020/043966に記載の化合物と比較して、より良好であった。
<Test Example 3>
P-gp affinity test The compounds synthesized in the examples were evaluated for P-gp substrate property by the following method, and were found to be good, for example, better than the compounds described in PCT/JP2020/043966. Met.
 得られた化合物がP-糖たんぱく質(P-gp)の基質となるか評価した。P-糖たんぱく質(MDR1)を発現したMDR1-MDCKII細胞およびMDCKII細胞を用い、得られた化合物をApical側またはBasolateral側に添加し、2時間インキュベーション後のそれぞれの側の化合物濃度を測定して、見かけの膜透過係数(Papp)、各細胞のFlux Ratio(PappBtоA/PappAtоB)を算出してP-糖たんぱく質による薬物輸送能を評価した。 We evaluated whether the resulting compound could be a substrate for P-glycoprotein (P-gp). Using MDR1-MDCKII cells and MDCKII cells expressing P-glycoprotein (MDR1), adding the obtained compound to the apical side or the basolateral side, measuring the compound concentration on each side after incubation for 2 hours, Apparent membrane permeability coefficient (Papp) and flux ratio (PappBtA/PappAtB) of each cell were calculated to evaluate drug transport ability by P-glycoprotein.
 本発明に係る化合物は、G9a酵素阻害活性によりがんなどの増殖性疾患、β-グロビン異常症、線維症、疼痛、神経変性疾患、プラダー・ウィリー症候群、マラリア、ウイルス感染症、ミオパチー、自閉症等の治療薬またはその予防薬として有用である。 The compounds according to the present invention are useful for proliferative diseases such as cancer, β-globin abnormalities, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, malaria, viral infections, myopathy, and autism due to G9a enzyme inhibitory activity. It is useful as a therapeutic or preventive agent for diseases and the like.
 本明細書中で引用した全ての刊行物、特許及び特許出願をそのまま参考として本明細書中にとり入れるものとする。 All publications, patents and patent applications cited in this specification shall be incorporated herein by reference.

Claims (9)

  1.  一般式(I):
    Figure JPOXMLDOC01-appb-C000001
    [式(I)中、RとRとは互いに結合して環を形成して、RとRが結合するピロール環とともに、以下のA1)の構造をとり、*が式(I)中の-CO-との結合位置を示し;
    Figure JPOXMLDOC01-appb-C000002
    3はC~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基またはヒドロキシC~Cアルキル基(該C~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基およびヒドロキシC~Cアルキル基はC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基およびヒドロキシC~Cアルキル基からなる群より選択される置換基で一つまたは複数置換されてもよく、置換基同士で互いに結合して環を形成してもよい)であり;
    Xは炭素原子または4位、5位、6位、7位のいずれかの窒素原子を示し、それぞれインドールまたはアザインドール環類であり;
    およびRはそれぞれ独立して、水素原子、ハロゲン原子、C~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基またはヒドロキシC~Cアルキル基(該C~Cアルキル基、C~Cアルケニル基、ハロC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアシル基、C~Cアルコキシカルボニル基、C~C10シクロアルキル基およびヒドロキシC~Cアルキル基はC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基およびヒドロキシC~Cアルキル基からなる群より選択される置換基で一つまたは複数置換されてもよく、置換基同士で互いに結合して環を形成してもよい)であり;
    は以下のB1)であり、*が式(I)中の-N-との結合位置を示し;
    Figure JPOXMLDOC01-appb-C000003
    およびRはそれぞれ独立して水素原子、C~Cアルキル基またはヒドロキシC~Cアルキル基であり;
    およびRは互いに結合して環を形成してもよく;
    およびRはRingAと結合して環を形成してもよく;
    mは0または1であり;
    RingAは、それぞれR10、R11およびR12で置換されてもよい、芳香族炭化水素環基、C~C10シクロアルキル基、5~10員ヘテロアリール基または3~10員ヘテロシクロアルキル基であり;
    10およびR11はそれぞれ独立して水素原子、ハロゲン原子、水酸基、アミノ基、シアノ基、カルバモイル基(-CONH)、C~Cアルキル基、ハロC~Cアルキル基、ヒドロキシC~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアルキルスルファニル基、ハロC~Cアルキルスルファニル基、C~Cアシル基、C~Cアルコキシカルボニル基、C~Cアルキルアミノカルボニル基またはC~Cアシルアミノ基であり;
    12は-(CR1314-V-(CR1516-(W)-Qであり;
    Vは結合、-O-または-NR17-であり;
    Wは-NR18-であり;
    13、R14、R15、R16、R17およびR18はそれぞれ独立して水素原子またはC~Cアルキル基であり;
    qおよびrはそれぞれ独立して0~6の整数であり;
    nは0または1であり;
    Qは水素原子、水酸基、C~Cアルキル基、C~C10シクロアルキル基、5~10員ヘテロアリール基または3~10員ヘテロシクロアルキル基(該C~Cアルキルアミノ基、C~C10シクロアルキル基、5~10員ヘテロアリール基および3~10員ヘテロシクロアルキル基は、ハロゲン原子、C~Cアルキル基、C~Cアルコキシ基、C~Cアルキルアミノ基、C~Cアルコキシカルボニル基、C~Cアルキルアミノカルボニル基およびヒドロキシC~Cアルキル基からなる群より選択される置換基で一つまたは複数置換されてもよい)であり;
    10、R11およびR12は互いに結合して環を形成してもよい]
    で表される化合物、またはその薬理学的に許容される塩。
    General formula (I):
    Figure JPOXMLDOC01-appb-C000001
    [In the formula (I), R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, the structure of A1) below is taken, and * is the formula (I ) indicates the bonding position with -CO- in;
    Figure JPOXMLDOC01-appb-C000002
    R 3 is C 1 -C 6 alkyl group, C 2 -C 6 alkenyl group, halo C 1 -C 6 alkyl group, C 1 -C 6 alkoxy group, C 1 -C 6 alkylamino group, C 1 -C 6 acyl group, C 1 -C 6 alkoxycarbonyl group, C 3 -C 10 cycloalkyl group or hydroxy C 1 -C 6 alkyl group (the C 1 -C 6 alkyl group, C 2 -C 6 alkenyl group, halo C 1 -C6 alkyl groups, C1 - C6 alkoxy groups, C1 - C6 alkylamino groups, C1 - C6 acyl groups, C1 - C6 alkoxycarbonyl groups, C3 - C10 cycloalkyl groups and hydroxy C 1 -C 6 alkyl group is a substituent selected from the group consisting of C 1 -C 6 alkyl group, C 1 -C 6 alkoxy group, C 1 -C 6 alkylamino group and hydroxy C 1 -C 6 alkyl group may be substituted one or more times, and the substituents may be bonded to each other to form a ring);
    X represents a carbon atom or a nitrogen atom at any of the 4-, 5-, 6-, and 7-positions, and is an indole or azaindole ring, respectively;
    R 6 and R 7 are each independently a hydrogen atom, a halogen atom, a C 1 -C 6 alkyl group, a C 2 -C 6 alkenyl group, a halo C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group, C 1 -C 6 alkylamino group, C 1 -C 6 acyl group, C 1 -C 6 alkoxycarbonyl group, C 3 -C 10 cycloalkyl group or hydroxy C 1 -C 6 alkyl group (the C 1 -C 6 Alkyl Group, C 2 -C 6 Alkenyl Group, Halo C 1 -C 6 Alkyl Group, C 1 -C 6 Alkoxy Group, C 1 -C 6 Alkylamino Group, C 1 -C 6 Acyl Group, C 1 -C 6 Alkoxycarbonyl groups, C 3 -C 10 cycloalkyl groups and hydroxy C 1 -C 6 alkyl groups are C 1 -C 6 alkyl groups, C 1 -C 6 alkoxy groups, C 1 -C 6 alkylamino groups and hydroxy C 1 may be substituted with one or more substituents selected from the group consisting of ~ C6 alkyl groups, and the substituents may be bonded to each other to form a ring);
    R 4 is B1) below, * indicates the bonding position with -N- in formula (I);
    Figure JPOXMLDOC01-appb-C000003
    R 8 and R 9 are each independently a hydrogen atom, a C 1 -C 6 alkyl group or a hydroxy C 1 -C 6 alkyl group;
    R 8 and R 9 may be joined together to form a ring;
    R 8 and R 9 may combine with RingA to form a ring;
    m is 0 or 1;
    RingA is an aromatic hydrocarbon ring group, a C3 - C10 cycloalkyl group, a 5- to 10-membered heteroaryl group or a 3- to 10-membered heterocycloalkyl group, each of which may be substituted with R 10 , R 11 and R 12 is a group;
    R 10 and R 11 each independently represent a hydrogen atom, a halogen atom, a hydroxyl group, an amino group, a cyano group, a carbamoyl group (—CONH 2 ), a C 1 -C 6 alkyl group, a halo C 1 -C 6 alkyl group, a hydroxy C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylamino, C 1 -C 6 alkylsulfanyl, halo C 1 -C 6 alkylsulfanyl, C 1 -C 6 acyl a C 1 -C 6 alkoxycarbonyl group, a C 1 -C 6 alkylaminocarbonyl group or a C 1 -C 6 acylamino group;
    R 12 is -(CR 13 R 14 ) r -V-(CR 15 R 16 ) q -(W) n -Q;
    V is a bond, -O- or -NR 17 -;
    W is -NR 18 -;
    R 13 , R 14 , R 15 , R 16 , R 17 and R 18 are each independently a hydrogen atom or a C 1 -C 6 alkyl group;
    q and r are each independently an integer from 0 to 6;
    n is 0 or 1;
    Q is a hydrogen atom, a hydroxyl group, a C 1 to C 6 alkyl group, a C 3 to C 10 cycloalkyl group, a 5 to 10 membered heteroaryl group or a 3 to 10 membered heterocycloalkyl group (the C 1 to C 6 alkylamino group , C 3 to C 10 cycloalkyl groups, 5 to 10 membered heteroaryl groups and 3 to 10 membered heterocycloalkyl groups are halogen atoms, C 1 to C 6 alkyl groups, C 1 to C 6 alkoxy groups, C 1 to substituted with one or more substituents selected from the group consisting of C 6 alkylamino group, C 1 -C 6 alkoxycarbonyl group, C 1 -C 6 alkylaminocarbonyl group and hydroxy C 1 -C 6 alkyl group may be);
    R 10 , R 11 and R 12 may combine with each other to form a ring]
    A compound represented by or a pharmacologically acceptable salt thereof.
  2.  式(I)中、RとRとが互いに結合して環を形成して、RとRが結合するピロール環とともに、以下のA1)の構造をとり;
    Figure JPOXMLDOC01-appb-C000004
    はC~Cアルキル基またはC~C10シクロアルキル基であり;
    Xは炭素原子または4位、5位、6位、7位のいずれかの窒素原子を示し、それぞれインドールまたはアザインドール環類であり;
    およびRはそれぞれ独立して、水素原子、C~Cアルキル基、C~Cアルコキシ基またはハロゲン原子である、
    請求項1に記載の化合物、またはその薬理学的に許容される塩。
    In formula (I), R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, take the following structure A1);
    Figure JPOXMLDOC01-appb-C000004
    R 3 is a C 1 -C 6 alkyl group or a C 3 -C 10 cycloalkyl group;
    X represents a carbon atom or a nitrogen atom at any of the 4-, 5-, 6-, and 7-positions, and is an indole or azaindole ring, respectively;
    R 6 and R 7 are each independently a hydrogen atom, a C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group or a halogen atom;
    A compound according to claim 1, or a pharmacologically acceptable salt thereof.
  3.  式(I)中、RとRとが互いに結合して環を形成して、RとRが結合するピロール環とともに、以下のA1a)の構造をとり;
    Figure JPOXMLDOC01-appb-C000005
    はC~Cアルキル基であり;
    およびRはそれぞれ独立して、水素原子、C~Cアルキル基、C~Cアルコキシ基またはハロゲン原子である、
    請求項2に記載の化合物、またはその薬理学的に許容される塩。
    In formula (I), R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, have the following structure A1a);
    Figure JPOXMLDOC01-appb-C000005
    R 3 is a C 1 -C 6 alkyl group;
    R 6 and R 7 are each independently a hydrogen atom, a C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group or a halogen atom;
    3. A compound according to claim 2, or a pharmacologically acceptable salt thereof.
  4.  式(I)中、RとRとが互いに結合して環を形成して、RとRが結合するピロール環とともに、以下のA1b)、A1c)、A1d)、A1e)のいずれかの構造をとり;
    Figure JPOXMLDOC01-appb-C000006
    はC~Cアルキル基であり;
    およびRはそれぞれ独立して、水素原子、C~Cアルキル基、C~Cアルコキシ基またはハロゲン原子である、
    請求項2に記載の化合物、またはその薬理学的に許容される塩。
    In formula (I), R 1 and R 2 are bonded to each other to form a ring, and together with the pyrrole ring to which R 1 and R 2 are bonded, any of the following A1b), A1c), A1d) and A1e) take the structure of;
    Figure JPOXMLDOC01-appb-C000006
    R 3 is a C 1 -C 6 alkyl group;
    R 6 and R 7 are each independently a hydrogen atom, a C 1 -C 6 alkyl group, a C 1 -C 6 alkoxy group or a halogen atom;
    3. A compound according to claim 2, or a pharmacologically acceptable salt thereof.
  5.  以下、
    Figure JPOXMLDOC01-appb-T000007
    Figure JPOXMLDOC01-appb-T000008
    Figure JPOXMLDOC01-appb-I000009
    Figure JPOXMLDOC01-appb-T000010
    Figure JPOXMLDOC01-appb-I000011
    から選択される化合物、またはその薬理学的に許容される塩。
    Less than,
    Figure JPOXMLDOC01-appb-T000007
    Figure JPOXMLDOC01-appb-T000008
    Figure JPOXMLDOC01-appb-I000009
    Figure JPOXMLDOC01-appb-T000010
    Figure JPOXMLDOC01-appb-I000011
    or a pharmaceutically acceptable salt thereof.
  6.  請求項1から5のいずれか1項に記載の化合物またはその薬理学的に許容される塩を有効成分として含有するG9a酵素阻害性組成物。 A G9a enzyme inhibitory composition containing the compound according to any one of claims 1 to 5 or a pharmacologically acceptable salt thereof as an active ingredient.
  7.  請求項1から5のいずれか1項に記載の化合物またはその薬理学的に許容される塩を有効成分として含有する医薬組成物。 A pharmaceutical composition containing the compound according to any one of claims 1 to 5 or a pharmacologically acceptable salt thereof as an active ingredient.
  8.  がんなどの増殖性疾患、β-グロビン異常症、線維症、疼痛、神経変性疾患、プラダー・ウィリー症候群、マラリア、ウイルス感染症、ミオパチー、自閉症からなる疾病群から選ばれる少なくとも一種の疾病の予防または治療のための医薬を製造するための請求項1から5のいずれか1項に記載の化合物またはその薬理学的に許容される塩の使用。 At least one disease selected from the group consisting of proliferative diseases such as cancer, β-globin disorders, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, malaria, viral infections, myopathy and autism Use of the compound according to any one of claims 1 to 5 or a pharmacologically acceptable salt thereof for manufacturing a medicament for the prevention or treatment of
  9.  がんなどの増殖性疾患、β-グロビン異常症、線維症、疼痛、神経変性疾患、プラダー・ウィリー症候群、マラリア、ウイルス感染症、ミオパチー、自閉症からなる疾病群から選ばれる少なくとも一種の疾病の予防または治療に用いられる、請求項1から5のいずれか1項に記載の化合物またはその薬理学的に許容される塩および薬学的に許容される担体を含有する医薬組成物。 At least one disease selected from the group consisting of proliferative diseases such as cancer, β-globin disorders, fibrosis, pain, neurodegenerative diseases, Prader-Willi syndrome, malaria, viral infections, myopathy and autism 6. A pharmaceutical composition containing the compound according to any one of claims 1 to 5 or a pharmacologically acceptable salt thereof and a pharmaceutically acceptable carrier, which is used for the prevention or treatment of .
PCT/JP2022/022270 2021-06-01 2022-06-01 G9a inhibitor WO2022255399A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2021092001 2021-06-01
JP2021-092001 2021-06-01

Publications (1)

Publication Number Publication Date
WO2022255399A1 true WO2022255399A1 (en) 2022-12-08

Family

ID=84323427

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2022/022270 WO2022255399A1 (en) 2021-06-01 2022-06-01 G9a inhibitor

Country Status (1)

Country Link
WO (1) WO2022255399A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2000501390A (en) * 1995-11-30 2000-02-08 ドクトル カルル トーマエ ゲゼルシャフト ミット ベシュレンクテル ハフツング Amino acid derivatives, pharmaceutical compositions containing these compounds and methods for their preparation
WO2001042208A1 (en) * 1999-12-08 2001-06-14 Teijin Limited Cycloamine ccr5 receptor antagonists
JP2011519845A (en) * 2008-04-29 2011-07-14 ヴァンティア リミテッド Aminopyridine derivatives
WO2014164867A1 (en) * 2013-03-11 2014-10-09 Imago Biosciences Kdm1a inhibitors for the treatment of disease
WO2021032933A1 (en) * 2019-08-21 2021-02-25 Kalvista Pharmaceuticals Limited Enzyme inhibitors
WO2021106988A1 (en) * 2019-11-27 2021-06-03 杏林製薬株式会社 G9a INHIBITOR
WO2021252644A1 (en) * 2020-06-09 2021-12-16 Pardes Biosciences, Inc. Inhibitors of cysteine proteases and methods of use thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2000501390A (en) * 1995-11-30 2000-02-08 ドクトル カルル トーマエ ゲゼルシャフト ミット ベシュレンクテル ハフツング Amino acid derivatives, pharmaceutical compositions containing these compounds and methods for their preparation
WO2001042208A1 (en) * 1999-12-08 2001-06-14 Teijin Limited Cycloamine ccr5 receptor antagonists
JP2011519845A (en) * 2008-04-29 2011-07-14 ヴァンティア リミテッド Aminopyridine derivatives
WO2014164867A1 (en) * 2013-03-11 2014-10-09 Imago Biosciences Kdm1a inhibitors for the treatment of disease
WO2021032933A1 (en) * 2019-08-21 2021-02-25 Kalvista Pharmaceuticals Limited Enzyme inhibitors
WO2021106988A1 (en) * 2019-11-27 2021-06-03 杏林製薬株式会社 G9a INHIBITOR
WO2021252644A1 (en) * 2020-06-09 2021-12-16 Pardes Biosciences, Inc. Inhibitors of cysteine proteases and methods of use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ANDRÉ BOLTJES; HUANG YIJUN; VAN DE VELDE ROB; RIJKEE LAURIE; WOLF SIGLINDE; GAUGLER JAMES; LESNIAK KATARZYNA; GUZIK KATARZYNA; HOL: "Fragment-Based Library Generation for the Discovery of a Peptidomimetic p53-Mdm4 Inhibitor", ACS COMBINATIONAL SCIENCE, vol. 16, no. 8, 11 August 2014 (2014-08-11), US , pages 393 - 396, XP055665962, ISSN: 2156-8952, DOI: 10.1021/co500026b *
KAHNBERG PIA, LUCKE ANDREW J., GLENN MATTHEW P., BOYLE GLEN M., TYNDALL JOEL D. A., PARSONS PETER G., FAIRLIE DAVID P.: "Design, Synthesis, Potency, and Cytoselectivity of Anticancer Agents Derived by Parallel Synthesis from α-Aminosuberic Acid", JOURNAL OF MEDICINAL CHEMISTRY, vol. 49, no. 26, 1 December 2006 (2006-12-01), US , pages 7611 - 7622, XP093009347, ISSN: 0022-2623, DOI: 10.1021/jm050214x *

Similar Documents

Publication Publication Date Title
EP1910333B1 (en) Piperidinyl-substituted isoquinolone derivatives as rho-kinase inhibitors
JP5600229B1 (en) Monocyclic pyridine derivative
WO2006019020A1 (en) Substituted ureas
US20100261743A1 (en) Novel seh inhibitors and their use
NZ521386A (en) Substituted beta-carbolines with lkB-kinase inhibiting activity
AU2015414743B2 (en) Process for the preparation of kinase inhibitors and intermediates thereof
MX2011001955A (en) Pyrrole compounds.
JP2010502617A (en) Hydantoin derivatives useful as antibacterial substances
WO2014134774A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
KR20110117194A (en) Novel ortho-aminoamides for the treatment of cancer
HUE033401T2 (en) Novel 5-fluorouracil derivative
EP3428159A1 (en) Crystalline forms of mesylate salt of pyridinyl amino pyrimidine derivative, preparation methods therefor, and applications thereof
ES2594252T3 (en) Quinolone compound and pharmaceutical composition
KR20180006450A (en) A urea derivative or a pharmacologically acceptable salt thereof
JP2010513395A (en) Compound showing a combination of cannabinoid-CB1 antagonism and acetylcholinesterase inhibition
EP0362759A1 (en) Pyridonecarboxylic acids
WO2020153434A1 (en) Pyrazole compound
WO2022255399A1 (en) G9a inhibitor
JP5017101B2 (en) Preparation of asymmetric tetrasubstituted carbon atom-containing compounds
WO2021106988A1 (en) G9a INHIBITOR
US20100311776A1 (en) Novel sEH Inhibitors and their Use
KR100497942B1 (en) Optically active quinoline carboxylic acid derivatives having 7-pyrrolidine substituentes causing optical activity and a process for the preparation thereof
US6825353B2 (en) Process for producing quinolonecarboxylic acids and intermediates thereof
CN111303128A (en) Polycyclic compound, preparation method and application thereof
EP0347851A1 (en) Quinolonecarboxylic acids

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: 22816145

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE