WO2016088813A1 - Novel diazabicyclo[2.2.2]octane derivative - Google Patents

Novel diazabicyclo[2.2.2]octane derivative Download PDF

Info

Publication number
WO2016088813A1
WO2016088813A1 PCT/JP2015/083926 JP2015083926W WO2016088813A1 WO 2016088813 A1 WO2016088813 A1 WO 2016088813A1 JP 2015083926 W JP2015083926 W JP 2015083926W WO 2016088813 A1 WO2016088813 A1 WO 2016088813A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
formula
acid
alkyl group
Prior art date
Application number
PCT/JP2015/083926
Other languages
French (fr)
Japanese (ja)
Inventor
大道 堀田
勲 櫻田
Original Assignee
持田製薬株式会社
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 持田製薬株式会社 filed Critical 持田製薬株式会社
Publication of WO2016088813A1 publication Critical patent/WO2016088813A1/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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4995Pyrazines or piperazines forming part of bridged ring systems
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • 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
    • C07D471/08Bridged systems

Definitions

  • the present invention relates to a compound having an orexin receptor antagonistic action, particularly a compound having a 2,5-diazabicyclo [2.2.2] octane structure represented by the following formula (I), or a compound thereof:
  • the present invention relates to a pharmaceutically acceptable salt, or a solvate thereof, and a pharmaceutical composition characterized by containing them as an active ingredient.
  • Orexin is a neuropeptide specifically expressed in neurons existing in the lateral hypothalamic area, OX-A consisting of 33 amino acids and 28 OX-B consisting of the following amino acids has been identified.
  • OX receptors which are specific receptors for these peptides, are OX1 receptor (hereinafter sometimes abbreviated as “OX1R”), and OX2 receptor (hereinafter, abbreviated as “OXR”).
  • OX1R OX1 receptor
  • OX2 receptor hereinafter, abbreviated as “OXR”.
  • Two subtypes have been reported (sometimes abbreviated as “OX2R”). All receptors are 7-transmembrane G protein-coupled receptors (GPCRs) expressed in the central nervous system, but the tissue distribution varies depending on the subtype and has various effects on various nerves. This suggests that orexin has complex physiological activities.
  • Insomnia insomnia symptoms
  • BZ benzodiazepines
  • BZ drugs including ultra-short-time BZ drugs zolpidem, zopiclone, etc.
  • institutions such as NIH
  • BZ drugs including ultra-short-time BZ drugs zolpidem, zopiclone, etc.
  • Non-Patent Document 3 OX knockout mice and OX2R knockout mice, inhibition of arousal is observed. From such knowledge, it is considered that the OXR antagonist is an excellent therapeutic agent for insomnia as a sleep induction agent that induces sleep by suppressing the arousal state.
  • OXR antagonists suvorexant (trade name: Bersomura (registered trademark)) and almorexant (ACT-0778573, clinical trial number NCT00608985) are known. These OXR antagonists are substances (dual orexin receptor antagonists (DORA)) that antagonize OX1R and OX2R to the same extent, and improvement of insomnia has been reported (Non-patent Documents 4 and 5). ).
  • DORA dual orexin receptor antagonists
  • an OX2R selective antagonist may be a therapeutic agent for insomnia that induces physiological sleep and reduces the risk of side effects.
  • OX2R selective antagonists MIN-202 (Minerva Neurosciences. Inc. (http://www.minerneunosciences.com)) and MK-1064 (Merck Sharp & Dohm Corp.) (non-patent documents) are currently available. It has been clinically developed as a treatment for insomnia.
  • Patent Document 1 discloses a compound having a cross-linked structure in the molecule as an OXR antagonist.
  • WO 2011/050198 pamphlet (Patent Document 2) and WO 2012/145581 pamphlet (Patent Document 3) include octahydropyrrolo [3,4-c] pyrrole in the molecule as an OXR regulator.
  • a compound having a structure is disclosed.
  • Patent Document 4 In WO 2011/050200 (Patent Document 4) and WO 2011/050202 (Patent Document 5), as an OXR regulator, 3,8-diaza-bicyclo [4. Compounds having 2.0] octane and 3,6-diazabicyclo [3.2.0] heptane structures are disclosed. However, the compounds disclosed therein are different in basic skeleton from the compounds having a diazabicyclo [2.2.2] octane structure, and the compounds having a diazabicyclo [2.2.2] octane structure are also disclosed. Nor.
  • Patent Document 6 a 5-substituted-2,5-diazabicyclo [2.2.2] octane-2-carbonyl structure is partially used as a potentiator of metabotropic glutamate receptors.
  • Patent Document 6 a 5-substituted-2,5-diazabicyclo [2.2.2] octane-2-carbonyl structure is partially used as a potentiator of metabotropic glutamate receptors.
  • Patent Document 7 discloses a partial 5-substituted-2,5-diazabicyclo [2.2.2] octane-2-carbonyl structure as an mTOR (mammalian target of rapamycin) inhibitor. Although a compound having a structure is disclosed, there is no disclosure of a specific compound of the present invention as shown below.
  • insomnia drugs including BZ drugs are available for pharmacotherapy of insomnia (insomnia symptoms), which is one of sleep disorders.
  • insomnia insomnia symptoms
  • the patient's treatment satisfaction is low due to side effects caused by the drug, and the development of a new insomnia drug having a better drug profile than that of existing drugs is required.
  • the present inventors have conducted extensive research to obtain an OXR antagonist that is highly safe and / or excellent in effectiveness, and as a result, is represented by the formula (I). That a compound having a 2,5-diazabicyclo [2.2.2] octane structure, or a pharmaceutically acceptable salt thereof, or a solvate thereof has an OXR antagonistic action. I found it.
  • the compound of the present invention has an OXR antagonistic action, and has an action of improving psychiatric and neurological disorders including sleep disorders such as insomnia.
  • the present invention relates to a compound having a 2,5-diazabicyclo [2.2.2] octane structure represented by the formula (I), or a pharmaceutically acceptable salt thereof, or a A solvate and a pharmaceutical composition characterized by containing them as active ingredients.
  • the compound of the present invention has an OXR antagonistic action, and has an action of improving psychological and neurological disorders including sleep disorders such as insomnia.
  • the pharmaceutical composition containing the compound of the present invention as an active ingredient can be administered orally, and OXR antagonists, diseases involving OXR, psychiatric and neurological disorders, particularly sleep disorders such as insomnia It is expected as a preventive and / or therapeutic agent.
  • the compound group of the present invention has at least one or more of such properties as good solubility, high metabolic stability, excellent oral absorption, and little hERG channel inhibitory action. Since it has characteristics, it is highly useful.
  • the present invention relates to a compound having a 2,5-diazabicyclo [2.2.2] octane structure represented by the following formula (I) shown in the following embodiments, or a pharmaceutically acceptable product thereof: Or a solvate thereof, and a pharmaceutical composition characterized by containing them as an active ingredient, and their pharmaceutical use, an OXR antagonist.
  • the present invention includes the following embodiments [1] to [14].
  • the first aspect of the present invention is The following formula (I): [In formula (I), m is an integer from 0 to 4; n is an integer of 0-4; ring A represents a C 6 ⁇ 14 aryl group or a 5-7 membered heteroaryl group, ; ring B represents a C 6 ⁇ 14 aryl group or a 5-7 membered heteroaryl group,; Q (the Q is diazabicyclo [2.2.2] amide bond at the adjacent position to couple the octane ring and ring B (o-position) to always replace: the substitution position of Q is also the same defined during each aspect described below), the C 3 ⁇ 8 cycloalkyl group, or -OR c or partial structural formula, (SF- Group represented by 1): Wherein (SF-1), ring C is C 6 ⁇ 14 aryl group, 5- to 7-membered heteroaryl group or 3
  • C 1-6 means that the number of constituent carbon atoms is 1 to 6, and unless otherwise specified, carbon atoms of a linear, branched or cyclic group Represents a number.
  • the chain group means “straight or branched chain having 1 to 6 carbon atoms”.
  • the cyclic group means “a cyclic group having 1 to 6 carbon atoms in the ring”.
  • the group containing a chain group and a cyclic group means “a group having 1 to 6 total carbon atoms”.
  • halogen atom examples include a fluorine atom, a chlorine atom, a bromine atom, and an iodine atom.
  • halogenated means having 1 to 5 of the above “halogen atoms” as a substituent. “Halogenation” is also referred to as “halogeno”.
  • examples of the “C 1-6 alkyl group” include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, Hexyl, isohexyl and the like can be mentioned.
  • halogenated C 1-6 alkyl group means a group in which the “C 1-6 alkyl group” is optionally substituted with 1 to 5 halogen atoms. Meaning, for example, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, 1,1,2,2-tetrafluoroethyl, pentafluoroethyl and the like.
  • hydroxyhalogenated C 1-6 alkyl group means that the above “halogenated C 1-6 alkyl group” is optionally substituted with 1 to 5 hydroxyl groups.
  • C 3 ⁇ 8 cycloalkyl group for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • the “aryl group” means “monocyclic aryl group”, “condensed aryl group (including bicyclic or tricyclic)” or “partially It may be a hydrogenated fused-ring aryl group.
  • the “partially hydrogenated condensed aryl group” means any hydrogen from a partially hydrogenated condensed ring in the above “condensed aryl group”. It means a monovalent group formed by removing an atom, and either a hydrogen atom in an aromatic ring part of a condensed ring or a hydrogen atom in a hydrogenated part may be removed.
  • C 6 ⁇ 14 aryl group for example, phenyl, 1-naphthyl, 2-naphthyl, 2-, 3-, 4-biphenyl anthryl, phenanthryl, acenaphthyl, Indanyl, indenyl, 1,2,3,4-tetrahydronaphthyl and the like can be mentioned.
  • the “heterocyclic group” means a 3 to 14-membered monocyclic or condensed ring containing 1 to 5 heteroatoms selected from a nitrogen atom, a sulfur atom and an oxygen atom. A monovalent group formed by removing any hydrogen atom from a cyclic ring.
  • examples of the “heterocyclic group” include “heteroaryl group” and “non-aromatic heterocyclic group”.
  • heteroaryl group means a 5- to 14-membered heteroaryl ring group containing 1 to 5 heteroatoms selected from a nitrogen atom, a sulfur atom, and an oxygen atom. means.
  • examples of the “heteroaryl group” include “monocyclic heteroaryl group”, “condensed heteroaryl group”, and “partially hydrogenated condensed ring”.
  • Formula heteroaryl group ".
  • the above “monocyclic heteroaryl group” is preferably one having 5 to 7 ring members (5 to 7 membered heteroaryl group), such as pyrrolyl, furyl, thienyl, Imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,3,4 Oxadiazolyl, furazanyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, 1,2,3-triazinyl, 1,2, 4-triazinyl, 1,3,5-triazinyl, 2H-1
  • condensed heteroaryl group means “heterocyclic group” and “aryl group”, or “heterocyclic group” and “monocyclic heteroaryl group”.
  • the above-mentioned “condensed heteroaryl group” is preferably one having 8 to 12 ring members, for example, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzothienyl, iso Benzothienyl, benzoxazolyl, 1,2-benzisoxazolyl, benzothiazolyl, 1,2-benzisothiazolyl, 1H-benzimidazolyl, 1H-indazolyl, 1H-benzotriazolyl, chromenyl, isochromenyl, quinolyl , Isoquinolyl, cinnolinyl, quinazolinyl, quinoxalinyl, phthalazinyl, benzoxazepinyl, benzoazepinyl, benzodiazepinyl, naphthyridinyl, purinyl, pteridinyl, carbazolyl
  • the “partially hydrogenated condensed heteroaryl group” means “heterocyclic group” and “aryl group”, or “heterocyclic group” and “heterocyclic group”.
  • a condensed ring formed by condensing an “aryl group” it means a monovalent group formed by removing any hydrogen atom from a partially hydrogenated condensed ring.
  • the arbitrary hydrogen atom is a hydrogen atom in any of the "heterocyclic group", “aryl group” and “heteroaryl group” in the condensed ring, or a hydrogen atom in the hydrogenated ring part.
  • quinoline is partially hydrogenated tetrahydroquinolyl
  • 5,6,7,8-tetrahydroquinolyl, 1,2,3,4-tetrahydroquinolyl and the like can be mentioned.
  • these groups can be, for example, 5,6,7,8-tetrahydroquinolyl-2-yl, -3-yl, -4-yl, -5-yl.
  • -6-yl, -7-yl, -8-yl and the like 1,2,3,4-tetrahydroquinolyl, for example, -1-yl, -2-yl, -3- Il, -4-yl, -5-yl, -6-yl, -7-yl, -8-yl and the like are exemplified.
  • the “partially hydrogenated condensed heteroaryl group” preferably has 8 to 12 ring members, such as indolinyl, 4, 5, 6, 7 -Tetrahydro-1H-indonyl, 2,3-dihydrobenzofuranyl, 4,5,6,7-tetrahydro-benzofuranyl, 2,3-dihydrobenzo [d] oxazolyl, 2,3-dihydrobenzo [d] thiazolyl, 4,5,6,7-tetrahydro-1H-indazolyl, chromanyl, 2H-chromenyl, 4H-chromenyl, isochromanyl, 1H-isochromenyl, 1,3-benzodioxolyl, 2,3-dihydrofuro [3,2-c ] Pyridyl etc. are mentioned.
  • the “monocyclic non-aromatic heterocyclic group (3- to 8-membered non-aromatic heterocyclic group)” is a hetero atom selected from an oxygen atom, a sulfur atom and a nitrogen atom. Represents a monovalent group formed by removing any hydrogen atom from a saturated or unsaturated heterocyclic ring, which is a 3- to 8-membered monocycle containing 1 to 4.
  • examples of the “monocyclic non-aromatic heterocyclic group (3- to 8-membered non-aromatic heterocyclic group)” include aziridinyl, azetidinyl, oxiranyl, thiranyl, oxetanyl, thietanyl, and the like.
  • the “—NR A R B group” means a group in which two hydrogen atoms on the nitrogen atom of the “amino group” are substituted with R A and R B. That is, “—NR A R B group” means that two hydrogen atoms on the nitrogen atom of “amino group” are each independently “hydrogen atom”, “C 1-6 alkyl group”, “halogenated C”.
  • n 0 to 2.
  • the ring A is preferably a group selected from a phenyl group, a pyridinyl group, a pyrimidinyl group, a pyrazinyl group, a thiazolyl group, or an oxazolyl group.
  • the ring A is more preferably ring A is a phenyl group, a pyridin-2-yl group, a pyrimidin-2-yl group, a pyrazin-2-yl group, a thiazole-2 -A group selected from an yl group and an oxazol-2-yl group.
  • the ring A is particularly preferably a pyridin-2-yl group or a pyrimidin-2-yl group.
  • the ring B is preferably a group selected from a phenyl group, a pyridinyl group, a pyridazinyl group, a furanyl group, a 1H-pyrazolyl group, and a thiazolyl group.
  • the ring B is more preferably the formula (B-1), the formula (B-2), the formula (B-3), the formula (B-4) or the formula (B -5) [in the formula (B-1), X 4 , X 5 , X 6 and X 7 each independently represents N or C—H] (provided that the formula (B— 1) to (B-5), the —C (O) — group and the Q group are not included in the ring B).
  • X 4 represents N or C-R 2a, where the R 2a represents a hydrogen atom, a halogen atom or -O-C 1 ⁇ 6 alkyl group,;
  • X 5 Represents C—R 2b , where R 2b is a hydrogen atom, a halogen atom, a C 1-6 alkyl group, or a 5- to 7-membered heteroaryl group (the 5- to 7-membered heteroaryl group is a halogen atom
  • X 1 represents N or C—R 2c , where R 2C represents a hydrogen atom, a halogen atom, a C atom, or a C 1-6 alkyl group (which may be substituted with 1 to 4 alkyl groups).
  • 1-6 alkyl group or -O-C 1 ⁇ represents an alkyl group
  • X 7 represents N or C-R 2d, wherein said R 2d represents a hydrogen atom or a halogen atom.
  • ring B is particularly preferably a group selected from formula (B-1-1), formula (B-1-2), or formula (B-3). (However, in formula (B-1-1), formula (B-1-2) and formula (B-3), the —C (O) — group and Q group are not included in ring B).
  • Q is —O-halogenated C 1-6 alkyl group (more specifically, 1,1,2,2-tetrafluoroethoxy group, and 2,2,2-trifluoroethoxy), and a group represented by the partial structural formula (SF-1):
  • ring C represents a pyridin-2-yl group, a 1H-pyrazol-1-yl group, a 2H-1,2,3-triazol-2-yl group, or 1,2,4.
  • p is an integer of 0 or 1
  • R 3 is a C 1-6 alkyl group (more specifically, R 3 is a methyl group) Represents).
  • R 1 is preferably a halogen atom, a cyano group, —R a1 , —OR a1 , —C (O) R a1 , —C (O) OR a1 , —SR a1 , —NR A R B group, 3- to 8-membered non-aromatic heterocyclic group, or oxo group (R A and R B in the —NR A R B group are each independently a hydrogen atom, or C 1- It represents an alkyl group; the R a1 is a C 1 ⁇ 6 alkyl group, a halogenated C 1 ⁇ 6 alkyl group, hydroxy halogenated C 1 ⁇ 6 alkyl group or an C 3 ⁇ 8 cycloalkyl group) .
  • R 1 a cyano group, C 1 ⁇ 6 alkyl group, C 3 ⁇ 8 cycloalkyl group, or -O-C 1 ⁇ 6 alkyl group, More specifically, R 1 is a cyano group, a methyl group, a cyclopropyl group, and a methoxy group.
  • R 2 is preferably a halogen atom, a cyano group, —R b1 , —OR b1 (where R b1 is a C 1-6 alkyl group, or a halogenated C 1 Represents a 6- alkyl group), a pyrazolyl group, or a pyridyl group (the pyrazolyl group and the pyridyl group may be substituted with 1 to 4 halogen atoms or a C 1-6 alkyl group).
  • R 2 is more preferably a halogen atom or a C 1-6 alkyl group, and more specifically, R 2 is fluorine and a methyl group.
  • the combination of the ring A and (R 1 ) m is preferably the following partial structural formula (A-1) or (A-2):
  • X 1 represents N or C—H
  • X 2 represents N or C—R 1a (wherein R 1a represents a hydrogen atom, a halogen atom, C 1-6 alkyl group, C 3 ⁇ 8 cycloalkyl group, -O-C 1 ⁇ 6 alkyl group, -S-C 1 ⁇ 6 alkyl group, -NR A R B group (said -NR A R in R B groups A and R B Each independently represents a hydrogen atom and a C 1-6 alkyl group), or a 3- to 8-membered non-aromatic heterocyclic group);
  • X 3 represents N or C—R 1b (the R 1b is a hydrogen atom, a cyano group, C 1 ⁇ 6 alkyl group, a hal
  • the combination of the ring A and (R 1 ) m is more preferably a formula (A-1) or a formula (A) which is a partial structural formula in the above embodiment [1-8].
  • X 1 represents N or C—H
  • X 2 represents N or C—R 1a (wherein R 1a represents a hydrogen atom, a fluorine atom, chlorine) An atom, a methyl group, a methoxy group, an ethoxy group, a (methoxy-d 3 ) group, a cyclopropyl group, a pyrrolidin-1-yl group, a methylthio group, or a dimethylamino group)
  • X 3 is N or C—R 1b (wherein R 1b represents a hydrogen atom, a cyano group, a methyl group, an ethyl group, a cyclopropyl group, an acetyl group, a methoxy group, a difluoromethoxy group, a trifluoromethyl group, 1-hydroxy-2,2,2- Trifluoroethyl group or methoxycarbonyl group
  • R 1c each independently represents a hydrogen atom, a flu
  • the combination of the ring A and (R 1 ) m is more preferably a 3,5-dimethylphenyl group, a 3-chloro-4-cyano-pyridin-2-yl group, 3-methyl-4- (trifluoromethyl) pyridin-2-yl group, 3-methoxy-4- (trifluoromethyl) pyridin-2-yl group, 4-acetylpyridin-2-yl group, 4-cyano- Pyridin-2-yl group, 4-cyano-3-cyclopropyl-pyridin-2-yl group, 4-cyano-3-methoxy-pyridin-2-yl group, 4-cyano-3-ethoxy-pyridine-2- Yl group, 4-cyano-3- (methoxy-d3) -pyridin-2-yl group, 4-cyano-6-methyl-pyridin-2-yl group, 4-cyano-3-fluoro-pyridin-2-yl Group 4-cyano
  • the combination of the ring A and (R 1 ) m is particularly preferably a 4-cyano-3-methoxy-pyridin-2-yl group, 4-cyano-3-cyclopropyl -Pyridin-2-yl group or 4,6-dimethylpyrimidin-2-yl group.
  • the combination of the rings B, Q and (R 2 ) n is preferably a partial structural formula of the formula (BQ-1), formula (BQ-2), formula (BQ -3), formula (BQ-4), or formula (BQ-5) (wherein, in formulas (BQ-1) to (BQ-5), the —C (O) — group represents ring B, Q and (R 2 ) Not included in the combination of n ):
  • X 4 represents N or C-R 2a (the R 2a represents a hydrogen atom, a halogen atom or -O-C 1 ⁇ 6 alkyl group,);
  • X 5 is C—R 2b (wherein R 2b is a hydrogen atom, a halogen atom, a C 1-6 alkyl group, or a 5- to 7-membered heteroaryl group (the 5- to 7-membered heteroaryl group is a halogen atom,
  • the combination of the rings B, Q and (R 2 ) n is more preferably a formula (BQ-1) which is a partial structural formula in the embodiment [1-9], Formula (BQ-2), Formula (BQ-3), Formula (BQ-4), or Formula (BQ-5)
  • X 4 represents N or C—R 2a
  • R 2a represents a hydrogen atom, a fluorine atom, a chlorine atom, a methoxy group, or an ethoxy group
  • X 5 represents C—R 2b (wherein R 2b represents a hydrogen atom, a fluorine atom, a methyl group
  • 1H— X 6 is N or C—R 2c (wherein R 2C is a hydrogen atom, a fluorine atom, a chlorine atom, a methyl atom), or a pyrazol-1-yl group or a 5-chloro-pyridin-3-yl
  • the combination of rings B, Q and (R 2 ) n is more preferably a 5-fluoro-3- (pyrimidin-2-yl) pyridin-2-yl group, 2,2′-bipyridin) -3-yl group, 2- (1H-1,2,3-triazol-1-yl) pyridin-3-yl group, 2- (1H-pyrazol-1-yl) pyridine- 3-yl group, 2- (1-methyl-1H-pyrazol-3-yl) pyridin-3-yl group, 2- (2H-1,2,3-triazol-2-yl) pyridin-3-yl group 2- (3-cyano-1H-pyrazol-1-yl) pyridin-3-yl group, 2- (3-methyl-1H-pyrazol-1-yl) pyridin-3-yl group, 2- (thiazol- 2-yl) pyridin-3-yl group, 2,5- (1H-
  • the combination of rings B, Q and (R 2 ) n is particularly preferably a 2- (2H-1,2,3-triazol-2-yl) phenyl group, 2 -Fluoro-6- (2H-1,2,3-triazol-2-yl) phenyl group, 5-methyl-2- (2H-1,2,3-triazol-2-yl) phenyl group, 2- ( 3-methyl-1,2,4-oxadiazol-5-yl) phenyl group, 2- (1H-pyrazol-1-yl) pyridin-3-yl group, 2- (2,2,2-trifluoro) An ethoxy) phenyl group or a 2- (1,1,2,2-tetrafluoroethoxy) phenyl group.
  • the combination of the ring B, Q and (R 2 ) n is preferably the following partial structural formula (BQ-6) or (BQ-7) (provided that In the formulas (BQ-6) and (BQ-7), the —C (O) — group is not included in the combination of ring B, Q and (R 2 ) n ):
  • [In formula (BQ-6) and formula (BQ-7), p and R 3 are the same as defined in the above embodiment [1-5];
  • X represents N or C—H;
  • R 2e represents a hydrogen atom, a cyano group, a C 1-6 alkyl group, or a halogenated C 1-6 alkyl group].
  • the combination of the rings B, Q and (R 2 ) n is more preferably a formula (BQ-6) which is a partial structural formula in the embodiment [1-10] or Formula (BQ-7) [In Formula (BQ-6) and Formula (BQ-7), p is an integer of 0 or 1; R 3 represents a cyano group; X is N or C—H. Y 2 represents S; R 2e represents a hydrogen atom, a cyano group, a methyl group, or a trifluoromethyl group.
  • the combination of rings B, Q and (R 2 ) n is more preferably a 3-methyl-1- (3-cyanophenyl) -1H-pyrazol-5-yl group 1- (pyridin-2-yl) -1H-pyrazol-5-yl group, 3-cyano-1- (pyridin-2-yl) -1H-pyrazol-5-yl group, 3-methyl-1- ( Pyridin-2-yl) -1H-pyrazol-5-yl group, 3- (trifluoromethyl) -1- (pyridin-2-yl) -1H-pyrazol-5-yl group, 1- (pyridazine-3- Yl) -1H-pyrazol-5-yl group or 1- (thiazol-2-yl) -1H-pyrazol-5-yl group.
  • the combination of rings B, Q and (R 2 ) n is particularly preferably a 1- (pyridin-2-yl) -1H-pyrazol-5-yl group.
  • the combination of the ring A and (R 1 ) m is preferably the same as defined in the embodiment [1-8]; the rings B, Q and (R 2 )
  • the combination of n is the same as defined in the above embodiment [1-9] or the above embodiment [1-10].
  • m is preferably an integer of 0 to 3.
  • m 2 is more preferable.
  • n is preferably an integer of 0 to 2.
  • n 0 or 1 is more preferable.
  • R 1 is preferably a halogen atom, a cyano group, —R a1 , —OR 1 , —C (O) R a1 , —C (O) OR a1 , —SR a1 , —NR A R B group, or 3- to 8-membered non-aromatic heterocyclic group (the above R a1 , R A , and R B are the same as defined in the above embodiment [1-6]) It is.
  • R 1 is more preferably the same as defined in the above embodiment [1-6-1].
  • R 2 is preferably a halogen atom, —R b1 , —OR b1 (wherein R b1 has the same definition as in the above embodiment [1-7]). ), A pyrazolyl group, or a pyridyl group (the pyrazolyl group and the pyridyl group may be substituted with 1 to 4 halogen atoms and a C 1-6 alkyl group).
  • R 2 represents more preferably a halogen atom or a C 1-6 alkyl group, and more specifically a fluorine and a methyl group.
  • the combination of the ring A ′ and (R 1 ) m is more preferably a combination of the ring A and the (R 1 ) m in the embodiment [1-8-1]. Same as definition.
  • the combination of the ring A ′ and (R 1 ) m is more preferably a combination of the ring A and the (R 1 ) m in the embodiment [1-8-2]. Same as definition.
  • the combination of the ring A ′ and (R 1 ) m is particularly preferably a combination of the ring A and the (R 1 ) m in the embodiment [1-8-3]. Same as definition.
  • the combination of the rings B ′, Q and (R 2 ) n is preferably a ring B, Q and (R 2 ) n in the embodiment [1-9].
  • the definition is the same as
  • n is more preferably a ring B, Q and (R) in the embodiment [1-9-1]. 2 ) Same definition as n .
  • n is more preferably a combination of the rings B, Q and (R) in the embodiment [1-9-2]. 2 ) Same definition as n .
  • the combination of the rings B ′, Q and (R 2 ) n is particularly preferably a ring B, Q and (R) in the above embodiment [1-9-3]. 2 ) Same definition as n .
  • m is preferably an integer of 0 to 3.
  • m 2 is more preferable.
  • Q is a group represented by the partial structural formula (SF-1): Wherein (SF-1), ring C is a phenyl group, pyridin-2-yl group, pyridazin-3-yl group or represents a group selected from thiazol-2-yl group,; p and R 3 are, It is the same as defined in the above embodiment [1-5-1].
  • Q is more preferably a pyridin-2-yl group.
  • R 1 is preferably a halogen atom, a cyano group, —R a2 , or —OR a2 (wherein R a2 is a C 1-6 alkyl group, and a halogenated group). Represents a group arbitrarily selected from a C 1-6 alkyl group.
  • R 1 is more preferably a cyano group, a C 1-6 alkyl group, or an —O—C 1-6 alkyl group, and more specifically, A cyano group, a methyl group, and a methoxy group;
  • R 2 is preferably a cyano group, a C 1-6 alkyl group, or a halogenated C 1-6 alkyl group.
  • the combination of the ring A ′ and (R 1 ) m is preferably the same as the definition of the ring A and (R 1 ) m in the embodiment [1-8]. It is.
  • the combination of the ring A ′ and (R 1 ) m is more preferably a combination of the ring A and the (R 1 ) m in the embodiment [1-8-1]. Same as definition.
  • the combination of the ring A ′ and (R 1 ) m is more preferably 4-cyano-pyridin-2-yl group, 4-trifluoromethyl-pyridine-2.
  • the combination of the ring A ′ and (R 1 ) m is particularly preferably a 4,6-dimethylpyrimidin-2-yl group.
  • the preferred embodiments can be arbitrarily formed.
  • the fourth aspect of the present invention is the compound of the above formula (I) of the above aspect [1], wherein the preferred compounds are the compounds listed below, or pharmaceutically acceptable salts thereof, or those: Solvates thereof, or optical isomers thereof.
  • the names of the compounds shown below are based on English names obtained according to the compound name naming program of ChemBioDraw (registered trademark) Ultra 14.0.0.117 (PerkinElmer Informations).
  • variable substituent R x in the following formula A can be substituted on any of the carbon atoms i, ii, iii or iv in the formula A
  • variable substituent R y in the following formula B can be substituted with the formula B It can be substituted with either carbon atom v or vi.
  • a fifth aspect of the present invention contains at least one of the compound represented by the above formula (I) or a pharmaceutically acceptable salt or solvate thereof as an active ingredient. It is a pharmaceutical composition characterized by these.
  • a sixth aspect of the present invention contains at least one of the compound represented by the above formula (I), a pharmaceutically acceptable salt thereof, or a solvate thereof as an active ingredient. It is a prophylactic and / or therapeutic agent for diseases involving OXR.
  • OXR is widely involved in biological functions. This suggests a potential role for OXR in the course of various diseases in humans or other species.
  • OXR OXR-associated disorders
  • sleep disorders such as insomnia, circadian rhythm sleep disorder, and sleep-related disorders
  • depression anxiety disorder, bipolar disorder, attention deficit hyperactivity disorder, autism, autism spectrum
  • Mental disorders such as disorders, drug dependence
  • neurodegenerative diseases such as Alzheimer's disease
  • memory disorders such as dementia
  • eating disorders such as bulimia.
  • An eighth aspect of the present invention contains at least one of the compound represented by the above formula (I), or a pharmaceutically acceptable salt or solvate thereof as an active ingredient.
  • At least one of the compound represented by the above formula (I), a pharmaceutically acceptable salt thereof or a solvate thereof is contained as an active ingredient, thereby preventing sleep disorder And / or a therapeutic agent, more preferably a prophylactic and / or therapeutic agent for insomnia.
  • insomnia include insomnia, circadian rhythm sleep disorder, and sleep-related complications. More specific examples of insomnia include primary insomnia, insomnia due to mental illness, insomnia due to physical disease, insomnia due to drugs, and the like. However, it is not limited to these.
  • a “mental disorder” specifically, depression, anxiety disorder, bipolar disorder, attention deficit hyperactivity disorder, autism, autism spectrum disorder, drug Addiction etc. are mentioned. However, it is not limited to these.
  • neurodegenerative diseases include Alzheimer's disease. However, it is not limited to these.
  • the “memory disorder” specifically includes dementia and the like. However, it is not limited to these.
  • treating disorders specifically includes bulimia and the like. However, it is not limited to these.
  • a ninth aspect of the present invention is an OXR antagonist comprising one or more of the compounds represented by the above formula (I), or a pharmaceutically acceptable salt or solvate thereof. .
  • a tenth aspect of the present invention is an OX2R selective antagonist comprising one or more of the compounds represented by the above formula (I), or a pharmaceutically acceptable salt or solvate thereof. It is.
  • Another aspect of the present invention is an OXR dual antagonist comprising one or more of the compounds represented by formula (I) above, or a pharmaceutically acceptable salt or solvate thereof.
  • the “OXR dual antagonist” means an orexin receptor antagonist having an orexin 1 receptor antagonistic action and an orexin 2 receptor antagonistic action.
  • An eleventh aspect of the present invention is the use of at least one pharmaceutical composition of a compound represented by the above formula (I), or a pharmaceutically acceptable salt or solvate thereof. is there.
  • a twelfth aspect of the present invention is the use of a compound represented by the above formula (I), or a pharmaceutically acceptable salt thereof or a solvate thereof as at least one orexin receptor antagonist. is there.
  • a thirteenth aspect of the present invention is a method for treating a disease selected from sleep disorder, mental disorder, neurodegenerative disorder, memory disorder and eating disorder, and is represented by the above formula (I) Administering at least one of a compound, or a pharmaceutically acceptable salt or solvate thereof, to a subject in need of treatment of the disease or condition.
  • a method for treating a sleep disorder wherein at least one of the compound represented by the above formula (I), or a pharmaceutically acceptable salt thereof or a solvate thereof is administered in the disease or condition.
  • a method comprising administering to a subject in need of treatment, more preferably a method of treating insomnia.
  • treatment refers to the progression of a “disease or condition” or one or more “diseases or conditions”. Means to mitigate, or suppress. Further, in the present specification, “treatment” refers to “disease” including preventing the onset of “disease or condition” or any symptoms related to “disease or condition” depending on the condition of the patient. Or prevention of “a condition” as well as reducing the severity of a “disease or condition” or any symptom thereof before onset. As used herein, “treating” is intended to include preventing and ameliorating the recurrence of a “disease or condition”.
  • the disease is insomnia, circadian rhythm sleep disorder, parasomnia, depression, anxiety disorder, bipolar disorder, attention deficit hyperactivity disorder, autism Or a prophylactic and / or therapeutic agent according to aspect [8] or [8-1] selected from the group consisting of: autism spectrum disorder, drug dependence, Alzheimer's disease, dementia, bulimia, or aspect [13] Or it is the method as described in [13-1].
  • the compound of the present invention is a compound having an IC 50 value for OXR of 1 ⁇ M or less when measured by a method in which orexin receptor antagonism is appropriately selected, for example, Pharmacological Experiment Example 1 (orexin receptor antagonism evaluation) described later. preferable. More preferred is a compound having an IC 50 value for the orexin receptor of 100 nM or less. Further, as a compound having a OX2R antagonism is preferably a compound an IC 50 value is less than 1 ⁇ M for OX2R, and more preferably not more than 100nM compound.
  • the compound having such an activity is a compound having an OXR antagonistic action, particularly an OX2R antagonistic action, and can be used as an OXR antagonist, particularly an OX2R antagonist, or a pharmaceutical composition.
  • diseases involving OXR sleep disorders (insomnia, circadian rhythm sleep disorders, sleep-related complications, etc.), mental disorders (depression, anxiety disorders, bipolar disorder, attention deficit hyperactivity disorder, autism , Autism spectrum disorders, drug addiction, etc.), neurodegenerative diseases (Alzheimer's disease etc.), memory disorders (dementia etc.) or eating disorders (eg bulimia), especially OX2R-related diseases, insomnia, etc. It can be used as a preventive and / or therapeutic agent for sleep disorders.
  • the compounds of the present invention include compounds having orexin 2 receptor antagonistic activity but not having orexining orexin 1 receptor antagonistic activity.
  • Such compounds include orexin 2 receptor. It can be used as a compound having a selective antagonism or an orexin 2 receptor selective antagonist.
  • a compound having an IC 50 value for orexin 1 receptor of 10 times or more, 20 times or more, 50 times or more, or 100 times or more of the IC 50 value for orexin 2 receptor can be mentioned.
  • a compound having an orexin 2 receptor selective antagonism is useful as a pharmaceutical composition having no side effects due to orexin 1 receptor antagonism.
  • diseases involving orexin 2 receptor sleep disorders (insomnia, circadian rhythm sleep disorders, sleep-related complications, etc.), mental disorders (depression, anxiety disorder, bipolar disorder, attention deficit hyperactivity disorder, Autism, autism spectrum disorder, drug addiction, etc.), neurodegenerative diseases (Alzheimer's disease, etc.), memory disorders (dementia, etc.) or eating disorders (eg, bulimia), especially sleep disorders such as insomnia It can be used as a preventive and / or therapeutic agent.
  • the compounds of the present invention include compounds having orexin 2 receptor antagonism as well as orexin 1 receptor antagonism, and such compounds are compounds having orexin receptor dual antagonism, It can be used as an orexin receptor dual antagonist or a pharmaceutical composition. Moreover, it can utilize as a preventive and / or therapeutic agent of the disease in which the above orexin receptor is involved.
  • the compound of the present invention may form an acid addition salt or a salt with a base depending on the type of substituent.
  • a salt is not particularly limited as long as it is a pharmaceutically acceptable salt.
  • metal salts, ammonium salts, salts with organic bases, salts with inorganic acids, salts with organic acids, basics, Or the salt with an acidic amino acid etc. are mentioned.
  • the metal salt include alkali metal salts such as lithium salt, sodium salt, potassium salt and cesium salt, alkaline earth metal salts such as calcium salt, magnesium salt and barium salt, and aluminum salt. (For example, besides a mono salt, a disodium salt and a dipotassium salt are also included).
  • the salt with an organic base include, for example, methylamine, ethylamine, t-butylamine, t-octylamine, diethylamine, trimethylamine, triethylamine, cyclohexylamine, dicyclohexylamine, dibenzylamine, ethanolamine, diethanolamine, triamine.
  • the salt with inorganic acid include salts with hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, sulfuric acid, phosphoric acid and the like.
  • the salt with an organic acid include, for example, formic acid, acetic acid, trifluoroacetic acid, propionic acid, butyric acid, valeric acid, enanthic acid, capric acid, myristic acid, palmitic acid, stearic acid, lactic acid, sorbic acid, Salts with aliphatic monocarboxylic acids such as mandelic acid, salts with aliphatic dicarboxylic acids such as oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, malic acid, tartaric acid, and aliphatic tricarboxylic acids such as citric acid Salts with acids, salts with aromatic monocarboxylic acids such as benzoic acid and salicylic acid, salts of aromatic dicarboxylic acids such as phthalic acid, cinnamic acid, glycolic acid, pyruvic acid, oxylic acid, salicylic acid, N-acetylcysteine, etc.
  • Salt with organic carboxylic acid salt with organic sulfonic acid such as methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, aspartic acid, gluta Acid addition salts with acidic amino acids such as phosphate and the like.
  • Preferable examples of salts with basic amino acids include, for example, salts with arginine, lysine, ornithine
  • preferable examples of salts with acidic amino acids include, for example, salts with aspartic acid, glutamic acid, and the like. Is mentioned. Of these, pharmaceutically acceptable salts are preferred.
  • an inorganic salt such as an alkali metal salt (eg, sodium salt, potassium salt), an alkaline earth metal salt (eg, calcium salt, magnesium salt, barium salt)
  • an inorganic acid such as hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, or acetic acid, phthalic acid, fumaric acid
  • examples thereof include salts with organic acids such as oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, methanesulfonic acid, and p-toluenesulfonic acid.
  • the salt is formed by mixing the compound of the present invention with a solution containing an appropriate amount of acid or base to form the desired salt, and then separated by filtration, or the mixed solvent is distilled off. Can be obtained.
  • the compound of the present invention or a salt thereof can form a solvate with a solvent such as water, ethanol or glycerol.
  • solvate means a molecular complex comprising a compound of the invention and one or more pharmaceutically acceptable solvent molecules (eg, water, ethanol, etc.).
  • solvent molecules eg, water, ethanol, etc.
  • hydrate When the solvent molecule is water, it is particularly called “hydrate”.
  • the description relating to the compound of the present invention includes the description relating to the salt, solvate, and solvate of the salt.
  • Prodrugs of the compounds of the present invention are also encompassed by the compounds of the present invention.
  • a “prodrug” is, for example, when a certain derivative of a compound of the invention, which may exhibit little or no desired pharmacological activity, is administered in or on the body, eg, hydrolysis. When converted to the compound of the present invention having the desired pharmacological activity by the like, the compound before administration is called “prodrug”.
  • the “prodrug” of the compound of the present invention can be prepared by, for example, converting an appropriate functional group present in the compound of the present invention into a method known in the literature, for example, Design of Products, H.P. It can be produced according to the method described in Bundgaard (Elsevier, 1985).
  • the compound of the present invention is substituted with a carboxy group (—COOH): the ester, ie, the hydrogen atom of the carboxy group (—COOH) is substituted with a “C 1-6 alkyl group” Compound.
  • the hydroxyl group (-OH) to a compound of the present invention is substituted: Part alkanoyloxy or ethers, i.e., a hydrogen atom is "C 2 ⁇ 7 alkanoyl group” in the hydroxyl group (-OH) or "C 2 compounds substituted with 1-7 alkanoyloxymethyl group ".
  • the compound of the present invention is a geometric isomer (geometric isomer), configurational isomer (configurational isomer), tautomer (tortomeric isomer), optical isomer (optical isomer), stereoisomer (diastereomer).
  • Positional isomers Regio isomers
  • rotational isomers Rotational isomers
  • both isomers and mixtures are also included in the compound of the present invention.
  • the optical isomer resolved from the racemate is also encompassed in the compound of the present invention.
  • the compound of the present invention has one or more asymmetric carbon atoms, two or more stereoisomers can exist. Further, the compounds of the present invention, if it contains "C 2 ⁇ 6 alkenyl group", geometric isomers (cis / trans or Z / E,) can be present. Also, tautomerism can occur when structural isomers can be interconverted by a low energy barrier. Examples of tautomerism include proton tautomerism in compounds having an imino, keto, or oxime group.
  • the formula (OC) includes both (1S, 4S) form (formula (OC-1)) and (1R, 4R) form (formula (OC-2)) isomers. It means that the body is included.
  • the compounds having * mark at the 1-position and 4-position of the formula (OC) have either steric configuration of the formula (OC-1) or the formula (OC-2). Meaning a compound.
  • the racemate when the compound of the present invention is an optically active substance, the racemate can be separated into a (+) isomer or a ( ⁇ ) isomer [D isomer or L isomer] by an ordinary optical resolution means.
  • each isomer is converted to a single isomer by a known synthesis method or separation method. It can be obtained as a compound.
  • optical resolution method include methods known per se, such as (1) fractional recrystallization method, (2) diastereomer method, (3) chiral column method and the like.
  • Fractionation recrystallization method After obtaining a crystalline diastereomer by ion-bonding an optical resolving agent to a racemate, it is separated by a fractional recrystallization method and, if desired, a neutralization step is performed. This is a method for obtaining a free optically pure compound.
  • the optical resolution agent include (+)-mandelic acid, ( ⁇ )-mandelic acid, (+)-tartaric acid, ( ⁇ )-tartaric acid, (+)-1-phenethylamine, ( ⁇ )-1-phenethylamine, Examples include cinchonine, ( ⁇ )-cinchonidine, brucine and the like.
  • Diastereomer method An optical resolution agent is covalently bonded (reacted) to a racemic mixture to obtain a mixture of diastereomers, which is then subjected to usual separation means (eg, fractional recrystallization, silica gel column chromatography). , HPLC (High Performance Liquid Chromatography, etc.) etc., and then optically pure by removing the optical resolving agent by chemical treatment such as hydrolysis reaction. This is a method for obtaining an optical isomer.
  • separation means eg, fractional recrystallization, silica gel column chromatography).
  • HPLC High Performance Liquid Chromatography, etc.
  • the compound of the present invention when the compound of the present invention has an intramolecular hydroxyl group or a primary or secondary amino group, the compound and an optically active organic acid (eg, MTPA [ ⁇ -methoxy- ⁇ - (trifluoromethyl) phenylacetic acid], (-)-Menthoxyacetic acid and the like) are subjected to a condensation reaction to obtain ester or amide diastereomers, respectively.
  • an amide or ester diastereomer can be obtained by subjecting the compound and an optically active amine or alcohol reagent to a condensation reaction. Each of the separated diastereomers is converted to an optical isomer of the original compound by subjecting it to an acid hydrolysis or basic hydrolysis reaction.
  • Chiral column method a method in which a racemate or a salt thereof is subjected to direct optical resolution by subjecting to a chromatography on a chiral column (optical isomer separation column).
  • a chiral column such as Daicel's CHIRAL series, water, various buffers (eg, phosphate buffer)
  • Optical isomers can be separated by developing using an organic solvent (eg, ethanol, methanol, isopropanol, acetonitrile, trifluoroacetic acid, diethylamine) alone or as a mixed solution.
  • the separation can be performed using a chiral column such as CP-Chirasil-DeX CB (manufactured by GL Sciences).
  • the compound of the present invention when it is an optically active substance, it can be synthesized by asymmetric synthesis in which one of optical isomers is selectively synthesized.
  • Optically active compounds are: (1) asymmetric synthesis reaction in which a racemic compound is enantioselectively reacted to lead to an optically active form, (2) diastereoselective from a naturally occurring optically active compound (sugar, amino acid, etc.). It can be synthesized by the synthesis method.
  • the compound of the present invention may be a crystal, and the compound of the present invention includes a single crystal form or a crystal form mixture.
  • the compound of the present invention may be a pharmaceutically acceptable cocrystal or cocrystal salt.
  • co-crystals or co-crystal salts are two or more unique at room temperature, each having different physical properties (eg structure, melting point, heat of fusion, hygroscopicity, solubility and stability). It means a crystalline substance composed of a simple solid.
  • the cocrystal or cocrystal salt can be produced according to a cocrystallization method known per se.
  • Compounds of the present invention include isotopes (eg, hydrogen isotopes, 2 H and 3 H, carbon isotopes, 11 C, 13 C, and 14 C, chlorine isotopes, 36 Cl, etc., fluorine Isotopes, 18 F, iodine isotopes, 123 I and 125 I, nitrogen isotopes, 13 N and 15 N, oxygen isotopes, 15 O, 17 O, and 18 O, phosphorus Also included are compounds labeled or substituted with isotopes, 32 P and the like, as well as sulfur isotopes, 35 S and the like.
  • isotopes eg, hydrogen isotopes, 2 H and 3 H, carbon isotopes, 11 C, 13 C, and 14 C, chlorine isotopes, 36 Cl, etc.
  • fluorine Isotopes, 18 F iodine isotopes, 123 I and 125 I
  • Compounds of the invention labeled or substituted with certain isotopes can be synthesized, for example, by Positron Emission Tomography; PET ) Can be used as a tracer (PET tracer) for use in medical diagnosis and the like.
  • Compounds of the invention labeled or substituted with certain isotopic labels are useful in drug and / or substrate tissue distribution studies.
  • 3 H and 14 C are useful for this research purpose because they are easy to label or displace and easy to detect.
  • the isotope-labeled compound of the present invention can be obtained by a common technique known to those skilled in the art or by a method similar to the synthesis method described in the Examples below.
  • the obtained isotope-labeled compound can be used for pharmacological experiments instead of the unlabeled compound.
  • a compound labeled or substituted with 2 H (or sometimes referred to as D or deuterium) (D compound, deuterated compound) is expected to have high stability and is useful as an active compound itself. is there.
  • a compound in which a hydrogen atom at a position to undergo metabolism is substituted with 2 H can be mentioned, and the metabolic reaction rate can be reduced with little influence on the properties of the compound.
  • a compound in which the position of irreversibly binding to a metabolic enzyme is substituted with 2 H can suppress the inhibition of the action of the metabolic enzyme, and can reduce the drug interaction at the time of combined use.
  • the isotope-labeled compound of the present invention can be obtained by a common technique known to those skilled in the art or by a method similar to the synthesis method described in the Examples below.
  • the obtained isotope-labeled compound can be used for pharmacological experiments instead of the unlabeled compound.
  • the definitions of Q, ring A, ring B, R 1 , R 2 , R 3 , m, n, p, and the like in each formula of the following production methods are the formulas ( It is the same as each definition of I).
  • the definition of M in the production method is a metal such as lithium, sodium and potassium unless otherwise specified.
  • the definition of X in the production method is a halogen atom or trifluoromethanesulfonyloxy (OTf) unless otherwise specified.
  • Definition of AR in the production process unless otherwise specified, a C 6 ⁇ 14 aryl group or 5- to 7-membered heteroaryl group.
  • B in the production method is boronic acid ester, boronic acid, trifluoroborate salt, boronic acid, or N-methyliminodiacetic acid ester unless otherwise specified.
  • P 1 and P 2 in the production method are a protecting group for a hydrogen atom or an imino group.
  • R alpha in the production process unless otherwise specified, is a C 1 ⁇ 6 alkyl group, C 6 ⁇ 14 aryl group or a benzyl group.
  • R beta in the production process unless otherwise specified, is a C 1 ⁇ 6 alkyl group or a C 3 ⁇ 8 cycloalkyl group.
  • R gamma in the manufacturing method, unless otherwise specified, C 3-8 cycloalkyl group, -O-C 1 ⁇ 6 alkyl group, -S-C 1 ⁇ 6 alkyl group, 3- to 8-membered non-aromatic A heterocyclic group, or a —NR A R B group;
  • each raw material compound used for the production of formula (I) may form a salt, and as such a salt, the same salts as those of the aforementioned formula (I) can be mentioned. Can be mentioned.
  • Each raw material compound used in the production of formula (I) can be used in the next reaction as a reaction solution or as a crude product, but can also be isolated from a reaction mixture according to a conventional method. It can be easily purified by known means, for example, separation means such as extraction, concentration, neutralization, filtration, distillation, recrystallization, chromatography and the like.
  • Examples of the solvent used for the recrystallization include water; alcohols such as methanol, ethanol, 2-propanol and butanol; ethers such as diethyl ether, tetrahydrofuran and 1,4-dioxane; n-hexane, cyclohexane and heptane.
  • Hydrocarbons such as benzene, toluene, xylene, etc .; amides such as N, N-dimethylformamide, N, N-dimethylacetamide, 1,3-dimethyl-2-imidazolidinone; chloroform , Halogenated hydrocarbons such as methylene chloride and 1,2-dichloroethane; nitriles such as acetonitrile; ketones such as acetone and diphenyl ketone; esters such as methyl acetate and ethyl acetate; sulfoxides such as dimethyl sulfoxide; , Trifluoroacetic acid, methanesulfur Phosphate, organic acids such as p- toluenesulfonic acid; and the like.
  • solvents can be used alone, or two or more kinds of solvents can be mixed at an appropriate ratio, for example, a ratio of 1: 1 to 1:10.
  • a ratio of 1: 1 to 1:10 can be used when the compound in a formula is marketed, a commercial item can also be used as it is, and what was manufactured by the method known per se or a method according to it can also be used.
  • substituent having the formula (I) is transformable functional group (e.g., carboxyl group, an amino group, a hydroxyl group, a carbonyl group, a mercapto group, -C (O) -O-C 1 ⁇ 6 alkyl group, -C ( O) -O-C 6 ⁇ 14 aryl group, -C (O) -O- benzyl group, a sulfo group, when containing a halogen atom, etc.), is converted by a method analogous to these functional groups per se known methods or in As a result, various compounds can be produced.
  • transformable functional group e.g., carboxyl group, an amino group, a hydroxyl group, a carbonyl group, a mercapto group, -C (O) -O-C 1 ⁇ 6 alkyl group, -C ( O) -O-C 6 ⁇ 14 aryl group, -C (O) -O- benzy
  • a “carboxy group” it can be converted by a reaction such as esterification, reduction, amidation, or a conversion reaction to an optionally protected amino group.
  • amino group it can be converted by a reaction such as amidation, sulfonylation, nitrosation, alkylation, arylation, imidation and the like.
  • hydroxyl group it can be converted by a reaction such as esterification, carbamoylation, sulfonylation, alkylation, arylation, oxidation, halogenation, and the like.
  • carbonyl group it can be converted by a reaction such as reduction, oxidation, iminization (including oximation and hydrazone formation), (thio) ketalization, alkylidene formation, thiocarbonylation and the like.
  • mercapto group it can be converted by a reaction such as alkylation or oxidation.
  • halogen atom In the case of a “halogen atom”, it can be converted by, for example, various nucleophilic substitution reactions, various coupling reactions, and the like.
  • a hydroxyl group an alcoholic hydroxyl group, a phenolic hydroxyl group, a heterocyclic hydroxyl group, etc.
  • an amino group as a substituent
  • Examples of the protective group for the hydroxyl group include, for example, C 1-6 alkyl groups (eg, methyl group, ethyl group, n-propyl group, isopropyl group, n-butyl group). group, and tert- butyl group); - C 1 ⁇ 6 alkyl -O-C 1 ⁇ 6 alkyl group (methoxymethyl group, methoxyethoxymethyl group, etc.); a tetrahydropyranyl group; C 7 ⁇ 20 aralkyl group (e.g.
  • Silyl group for example, trimethylsilyl group, triethylsilyl group, t-butyldimethylsilyl group, and t-butyldiphenylsilyl group); —C (O) —C 1- 6 alkyl group (e.g., acetyl group, ethylcarbonyl group, and pivaloyl group); - C (O) -C 7 ⁇ 20 aralkyl group (e.g., Ben Le carbonyl group); - C (O) -C 6 ⁇ 14 aryl group (e.g., benzoyl group); - C (O) -O -C 1 ⁇ 6 alkyl group (e.g., methoxycarbonyl group, ethoxycarbonyl group , and t- butoxycarbonyl group or the like); - C (O) -O -C 7 ⁇ 20 aralkyl group (e.g., a benzyl group
  • the protecting group of the carboxyl group for example, C 1 ⁇ 6 alkyl group (e.g., methyl group, ethyl group, n- propyl group, an isopropyl group, n- butyl group, and tert- butyl group); C 2 ⁇ 7 alkenyl group (vinyl group, and allyl group, etc.); C 6 ⁇ 14 aryl group (e.g., phenyl, etc.); C 7 ⁇ 20 aralkyl group (e.g., benzyl, and triphenylmethyl group); a silyl group (e.g. , Trimethylsilyl group, triethylsilyl group, t-butyldimethylsilyl group, t-butyldiphenylsilyl group, etc.).
  • C 1 ⁇ 6 alkyl group e.g., methyl group, ethyl group, n- propyl group, an isopropyl
  • Examples of the protecting group for the thiol group include C 1-6 alkyl groups (eg, methyl group, ethyl group, n-propyl group, isopropyl group, n-butyl group, tert-butyl group, etc.) ); C 7 ⁇ 20 aralkyl group (e.g., benzyl, and triphenylmethyl group and the like); - C (O) -C 1 ⁇ 6 alkyl group (e.g., acetyl group, ethylcarbonyl group, and pivaloyl group); -C (O) -C 6 ⁇ 14 aryl group (e.g., benzoyl group) and the like.
  • C 1-6 alkyl groups eg, methyl group, ethyl group, n-propyl group, isopropyl group, n-butyl group, tert-butyl group, etc.
  • Such protecting group introduction / removal method is appropriately carried out depending on the group to be protected or the type of protecting group.
  • Green, et al. Protective Groups in Organic Synthesis (Protective Groups in Organic). (Synthesis), 4th edition, 2007, John Wiley & Sons, can be performed by the method described in the book.
  • the deprotection methods of protecting groups e.g., -C (O) -C 1 ⁇ 6 alkyl group (e.g., acetyl group, ethylcarbonyl group, and pivaloyl group); - C (O) -O -C 1 ⁇ 6 alkyl group (e.g., methoxycarbonyl group, ethoxycarbonyl group, and t- butoxycarbonyl group or the like); - acyl type protecting groups such as C (O) -C 6 ⁇ 14 aryl group (e.g., benzoyl group, etc.),
  • a suitable base such as an alkali metal hydroxide such as lithium hydroxide, sodium hydroxide or potassium hydroxide.
  • -C 1 ⁇ 6 alkyl -O-C 1 ⁇ 6 alkyl group (methoxymethyl group, methoxyethoxymethyl group, etc.); a tetrahydropyranyl group; -C (O) -O-C 1 ⁇ 6 alkyl group (e.g., methoxy group, an ethoxycarbonyl group, and t- butoxycarbonyl group or the like); - C (O) -O -C 7 ⁇ 20 aralkyl group (e.g., benzyloxycarbonyl group, and para-methoxybenzyloxycarbonyl group, etc.); silyl groups Protecting groups such as trimethylsilyl group, triethylsilyl group, t-butyldimethylsilyl group, t-butyldiphenylsilyl group and the like are, for example, acetic acid, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, triflu
  • the silyl group can also be deprotected by using a suitable fluorine ion (F ⁇ ) generating reagent, for example, a reagent such as tetrabutylammonium fluoride or hydrogen fluoride.
  • a suitable fluorine ion (F ⁇ ) generating reagent for example, a reagent such as tetrabutylammonium fluoride or hydrogen fluoride.
  • C 7 ⁇ 20 aralkyl group e.g., benzyloxycarbonyl group, para-methoxybenzyloxycarbonyl group, and para-nitrobenzyloxycarbonyl group and the like
  • C 7 ⁇ 20 aralkyl group e.g., benzyl group And the like can be deprotected by hydrogenolysis using, for example, a palladium-carbon (Pd—C) catalyst.
  • the above C 7 ⁇ 20 aralkyl group (e.g., benzyl group) protecting groups are, for example, in liquid ammonia, can be deprotected by Birch reduction using metal sodium.
  • the triphenylmethyl group can be deprotected by using an appropriate acid, for example, an acid such as acetic acid, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, trifluoroacetic acid, trifluoromethanesulfonic acid or a combination thereof. . It can also be deprotected by liquid Birch reduction using metallic sodium or metallic lithium or hydrogenolysis using a palladium carbon catalyst.
  • an appropriate acid for example, an acid such as acetic acid, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, trifluoroacetic acid, trifluoromethanesulfonic acid or a combination thereof.
  • an appropriate acid for example, an acid such as acetic acid, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, trifluoroacetic acid, trifluoromethanesulfonic acid or a combination thereof.
  • It can also be deprotected by liquid Birch reduction using metallic sodium or metallic
  • deprotection can be achieved by using one-electron reduction using Na / anthracene or Na / naphthalene at low temperature or Birch reduction using metallic sodium or metallic lithium in liquid ammonia.
  • 2-nitrobenzenesulfonyl group and a 2,4-nitrobenzenesulfonyl group for example, the presence of a basic reagent such as potassium carbonate or triethylamine, reacting the thiol, Can be deprotected under mild conditions.
  • a basic reagent such as potassium carbonate or triethylamine
  • the protecting group deprotection method shown here is only one example.
  • Green et al. “Protective Groups in Organic Synthesis, 4th Edition, 2007. Deprotection is possible by applying the method described in the book of John Wiley & Sons or various published papers.
  • the reaction conditions in the production method described below are as follows unless otherwise specified.
  • the reaction temperature is in the range from ⁇ 78 ° C. to the temperature at which the solvent is refluxed. When the temperature is not described, it is room temperature (0 to 35 ° C.), and the reaction time is the time for which the reaction proceeds sufficiently. .
  • each step in the production method can be performed without solvent or by dissolving or suspending the raw material compound in a solvent not involved in an appropriate reaction before the reaction.
  • solvents that do not participate in the reaction include water; saturated hydrocarbon solvents such as cyclohexane and hexane; aromatic hydrocarbon solvents such as benzene, chlorobenzene, toluene, and xylene; methanol, ethanol, 1-propanol, 2 Alcohol solvents such as propanol, tert-butyl alcohol, 2-methoxyethanol; N, N-dimethylformamide, N, N-dimethylacetamide, hexamethylphosphoric triamide, 1,3-dimethyl-2-imidazolidinone Polar amide solvents such as: sulfoxide solvents such as dimethyl sulfoxide; nitrile solvents such as acetonitrile and propionitrile; diethyl ether, diisopropyl ether, diphenyl
  • the base (or deoxidizer) used in the method for producing the compound of the present invention include lithium hydroxide, sodium hydroxide, potassium hydroxide, magnesium hydroxide, lithium carbonate, sodium carbonate, potassium carbonate, Inorganic bases such as cesium carbonate, calcium carbonate, sodium bicarbonate; triethylamine, N, N-diisopropylethylamine (DIPEA), tributylamine, cyclohexyldimethylamine, pyridine, lutidine, 4-dimethylaminopyridine (DMAP), N, N- Dimethylaniline, N-methylpiperidine, N-methylpyrrolidine, N-methylmorpholine, 1,5-diazabicyclo [4.3.0] -5-nonene, 1,4-diazabicyclo [2.2.2] octane, 1 , 8-diazabicyclo [5.4.0] -7-undece
  • Organic bases such as imidazole; sodium methoxide, sodium e
  • inorganic acids such as hydrochloric acid, sulfuric acid, nitric acid, hydrobromic acid, phosphoric acid; acetic acid, trifluoroacetic acid, oxalic acid , Organic acids such as phthalic acid, fumaric acid, tartaric acid, maleic acid, citric acid, succinic acid, methanesulfonic acid, p-toluenesulfonic acid, 10-camphorsulfonic acid; boron trifluoride ether complex, zinc iodide, anhydrous Lewis acids such as aluminum chloride, anhydrous zinc chloride, and anhydrous iron chloride.
  • inorganic acids such as hydrochloric acid, sulfuric acid, nitric acid, hydrobromic acid, phosphoric acid
  • acetic acid trifluoroacetic acid, oxalic acid
  • Organic acids such as phthalic acid, fumaric acid, tartaric acid, maleic acid, citric acid, succinic acid, methanesulf
  • the salt of the formula (I) is prepared according to a method known per se.
  • an inorganic acid such as hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, etc.
  • organic acids such as formic acid, acetic acid, trifluoroacetic acid, phthalic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid
  • formula (I) is an acidic compound, ammonia, trimethylamine, triethylamine, pyridine, picoline, 2,6-lutidine, ethanolamine, diethanolamine, triethanolamine, cyclohexylamine, dicyclohexylamine, N, N -Diisopropylethylamine, N, N'-dibenzylethylenediamine, N, N-dialkyl
  • an organic base such as ruaniline or an inorganic base such as lithium carbonate, sodium carbonate, potassium carbonate, cesium carbonate, lithium hydroxide, sodium hydroxide
  • a Pd catalyst such as tris (dibenzylideneacetone) dipalladium (Pd 2 (dba) 3 ), and 4,5′-bis (diphenylphosphino)
  • phosphine reagents such as -9,9'-dimethylxanthene (XANTPHOS) and organic or inorganic bases such as triethylamine, N, N-diisopropylethylamine, potassium phosphate, potassium carbonate, cesium carbonate, toluene, xylene, N , N-dimethylformamide, N, N-dimethylacetamide, dimethoxyethane, acetonitrile (
  • the compound represented by the formula (AM-3) can be produced by reacting at a temperature at which the solvent is refluxed.
  • ⁇ Step 2> [Production Method A] Using a compound represented by the formula (AM-3) obtained in ⁇ Step 1>, a method known in the literature, for example, “Protective Groups in Organic Synthesis” 4ThEdition) 4th edition, 2007, John Wiley & Sons (John Wiley & Sons), in accordance with the method described in textbooks of green (Greene) et al ", in accordance with the type of the protecting group the protecting group P 2 By reacting by the method, a compound represented by the formula (AM-4) in which the P 2 group is deprotected can be produced.
  • ⁇ Step 3> [Production Method A] Using the amine of formula (AM-4) obtained in ⁇ Step 2> and the carboxylic acid of formula (CA), a method known in the literature, for example, “Experimental Chemistry Course 4th Edition 22 Organic Synthesis” In accordance with the method described in IV Acid / Amino Acid / Peptide, 191-309, 1992, Maruzen, etc., 1,3-dicyclohexylcarbodiimide (DCC), 1-ethyl-3- (3′-dimethylaminopropyl) ) Carbodiimide hydrochloride (EDCI), 1-hydroxybenzotriazole (HOBT), benzotriazole-1-iroxytris (dimethylamino) phosphonium hexafluorophosphate (BOP reagent), bis (2-oxo-3-oxazolidinyl) phosphinic Chloride (BOP-Cl), 2-chloro-1,3-dimethylimidazolinium
  • carboxylic acid of the formula (CA) is converted into a method known in the literature, for example, “Journal of the American Chemical Society, 109 (24), p7488-7494, 1987”.
  • a base such as triethylamine, N, N-diisopropylethylamine, N, N-dimethylaminopyridine, thionyl chloride, oxalyl chloride, phosphoryl chloride, sulfuryl chloride, trichloride.
  • halogenating agent such as phosphorus, phosphorus pentachloride or phosphorus tribromide and a solvent inert to the reaction such as dioxane, tetrahydrofuran, benzene, toluene, dichloromethane, 1,2-dichloroethane, chloroform, or a mixed solvent thereof.
  • a solvent inert such as dioxane, tetrahydrofuran, benzene, toluene, dichloromethane, 1,2-dichloroethane, chloroform, or a mixed solvent thereof.
  • ⁇ Step 2> [Production Method B] Using a compound of formula (AM-5) obtained in ⁇ Step 1>, a method known in the literature, for example, “Protective Groups in Organic Synthesis 4th Edition” 4th edition, 2007, according to the method described in the book of John Wiley & Sons, Greene et al., The protecting group P 1 is reacted by a method according to the type of the protecting group. Thus, a compound represented by the formula (AM-6) in which the P 1 group is deprotected can be produced.
  • ⁇ Step 2> [Method C] Compounds of ⁇ Step 1> In the resulting formula (IM-1), and halogenated C 6 ⁇ 14 aryl compound of formula (RG-1) (the compound document from commercially available compounds, or commercially available compound The compound is a compound that can be produced by a known production method), and a method known in the literature, for example, “Experimental Chemistry Course 5th edition 18 Synthesis of organic compounds VI —Organic synthesis using metals—327-352, 2004, Maruzen And palladium (II) acetate (Pd (OAc) 2 ) according to the methods described in Journal of Medicinal Chemistry, 48 (20), p 6326-6339, 2005.
  • trialkylborate such as trimethylborate and triisopropylborate was added, and the reaction was carried out at ⁇ 78 ° C. at room temperature, and then acid such as hydrochloric acid and sulfuric acid was added. The reaction is carried out at a temperature at which the solvent refluxes from -1) boronic acids can be produced.
  • a compound of formula (AR-2-1) and a compound of formula (AM-1) (formula (AR-2-1) and compound of formula (AM-1) are commercially available compounds, or known production from literature
  • the compound of formula (D-1) can be produced by carrying out a reaction according to [Production Method B] ⁇ Step 3> using a compound of (a compound that can be produced by the method).
  • the compound of formula (AR-2-2), and the compound of formula (AM-1) (formula (AR-2-2) and compound of formula (AM-1) are commercially available compounds, or prepared from commercially available compounds in the literature
  • the compound of formula (D-3) can be produced by carrying out a reaction according to [Production Method B] ⁇ Step 3>.
  • ⁇ Step 7> [Production Method D] Using the compound of the formula (D-3) obtained in ⁇ Step 6> and trifluoroacetic anhydride (TFAA), a method known in the literature, for example, “Patent of International Publication No. WO 2007/043777. In the presence of a base such as triethylamine, the reaction is carried out at a temperature at which the solvent is refluxed from 0 ° C. in the presence of a base such as triethylamine. Compounds can be produced.
  • TFAA trifluoroacetic anhydride
  • a compound of formula (E-1) (the compound is a commercially available compound or a compound that can be produced from a commercially available compound by a known production method in the literature), and a compound of formula (RG-2) or formula (RG-3)
  • the compound of formula (E-2) can be produced by carrying out a reaction according to [Production method C] ⁇ Step 4>.
  • the compound of the formula (CA-3) can be produced by performing the reaction at a temperature at which the solvent is refluxed from 0 ° C. using a solvent that does not participate in the reaction such as 4-dioxane and tetrahydrofuran, or a mixed solvent thereof.
  • reaction can be carried out at a temperature at which the solvent is refluxed from 0 ° C. to produce a compound of formula (CA-3).
  • a compound of the formula (G-1) (the compound is a commercially available compound or a compound that can be produced from a commercially available compound by a method known in the literature), and a hydrazine compound of the formula (G-2) (the compound is a commercially available compound) Or a compound that can be produced from a commercially available compound by a production method known in the literature), in the presence of acetic acid according to a method known in the literature, for example, the method described in “International Publication No. 2007/043777 pamphlet” or the like.
  • a solvent that does not participate in the reaction such as tetrahydrofuran, 2-methoxyethanol, or a mixed solvent thereof, at a temperature at which the solvent is refluxed from 0 ° C. to produce a compound of formula (G-3). it can.
  • LDA lithium diisopropylamide
  • J-1 the compound was a commercially available compound or It is a compound that can be produced from a commercially available compound by a known production method in the literature
  • the reaction solution is added with excess dry ice (solid piece) or excess carbon dioxide gas to carry out the reaction.
  • an acid such as (CA-5-3) It can be prepared of compounds.
  • Combination agent containing the compound of the present invention can be used in combination with other drugs or drugs by a general method used in the medical field.
  • Examples of the drug that can be combined or used in combination with the compound of the present invention include (A) a sleep disorder-related drug, (B) a therapeutic drug for a disease that easily causes sleep disorder, and the like.
  • Examples of the drug (A) include: (1) sleep inducers ((i) benzodiazepine-based sleep inducers [specifically, nitrazepam, estazolam, flurazepam hydrochloride, nimetazepam, flurazepam, haloxazolam, flunitrazepam, rilmazapine hydrochloride, Lormetazepam, triazolam, etc.], (ii) thienodiazepine sleep inducer [specifically, brotizolam, etc.], (iii) non-benzodiazepine sleep inducer [specifically, zolpidem, etc.], (iv) melatonin receptor operation Drugs (specifically, ramelteon, etc.), (v) cyclopyrrolone sleep inducers [specifically, zopiclone, etc.], (vi) orexin receptor antagonists [specifically, suvorexant, etc.], ( 2) Drugs prescribed for sleep apnea
  • Examples of the drug of (B) include (4) atypical antipsychotic drugs [specifically, olanzapine, quetiapine, clozapine, ziprasidone, risperidone, paliperidone, perospirone, blonanserin, lurasidone, aripiprazole, sertindole, amisulpride, Iloperidone, bifeprunox, asenapine, melperone, brexpiprazole, zotepine, etc.], (5) typical antipsychotics [specifically, chlorpromazine, pchlorperazine, perphenazine, levomepromazine, fluphenazine, thioridazine, propericazine, spiperone] , Moperon, haloperidol, timiperone, bromperidol, pimozide, fluropipamide, sulpiride, thioprid, s
  • Muscarinic M1 acetylcholine receptor activity modulator (21) Muscarinic M2 acetylcholine receptor activity modulator, (22) Adenosine receptor modulator, (23) Muscarinic M4 acetylcholine receptor activity regulator, (24 ) Muscarinic M5 acetylcholine receptor activity modulator, (25) adenosine receptor modulator, (26) glycine transporter 1 (GlyT1) inhibitor [specifically, ALX5407, SSR504734, etc.], (27) glutamate enhancer [Specifically, ampakine], (28) NMDA receptor inhibitor [specifically, memantine hydrochloride and the like], (29) metabolic glutamate receptor modulator (mGlu) [specifically, CDPPB, MPEP Etc.], (30) Anti-anxiety drugs ((i) benzodiazepine anxiolytic drugs [specifically Chlordiazepoxide, diazepam, oxazolam, medazep
  • Antidiabetic agent ((i) PPAR ⁇ agonist (agonist, inhibitor) [specifically, pioglitazone, rosiglitazone, troglitazone, siglitazone, darglitazone, englitazone, netoglitazone, etc.], (ii) insulin Secretion enhancer [(a) sulfonylurea (specifically, tolbutamide, acetohexamide, chlorpropamide, glibenclamide, gliclazide, glipizide, glimepiride, glipentide, glyquidone, glisolamide, tolazamide, etc.), (b) non-sulfonylurea Agents, etc.], (iii) fast-acting insulin secretagogues (specifically, nateglinide, mitiglinide, repaglinide, etc.), (iv) ⁇ -glucosidase inhibitors [specifically, acarbose, a
  • GPR40 agonists GPR40 agonists, (g) GPR119 agonists, (h) GPR120 agonists], (vi) hepatic gluconeogenesis inhibitors [specifically, glucagon antagonists, etc.], vii) biguanides [specifically, metformin, buformin, phenformin, etc.], (viii) insulin or insulin derivatives [specifically, insulin Phosphorus zinc suspension, insulin lispro, insulin aspart, regular insulin, NPH insulin, insulin glargine, insulin detemir, mixed insulin, etc.], (ix) ⁇ 2 antagonist [specifically, midaglyzol, isagridol, deliglidol, idazoxan , Efaloxane, etc.]),
  • anti-obesity drugs ((i) adrenergic ⁇ 3 receptor agonists [specifically, KRP-204, TRK-380 / TAC-301, etc.], (ii) CB-1 receptor antagonists [specifically Rimonabant, SR-147778, BAY-65-2520 etc.], (iii) Neuropeptide Y (NPY) receptor antagonist [specifically, S-2367 etc.], (iv) Antifeedant [monoamine] Reuptake inhibitors [specifically sibutramine, mazindol, etc.]], (v) lipase inhibitors [specifically, orlistat, cetiristat, etc.], (vi) peptide YY (PYY) receptor antagonists, etc.), (40) Antihyperlipidemic drugs such as cholesterol-lowering drugs ((i) ⁇ 3 fatty acids [specifically, ethyl icosapentate (EPA-E preparation, for example, product name: Epadale (registered trademark) ), Docos,
  • Antihypertensive agent (i) diuretic [specifically, trichlormethiazide, hydrochlorothiazide, mefluside, indapamide, methiclan, chlorthalidone, tripamide, furosemide, torasemide, bumetanide, ethacrynic acid, spironolactone, triamterene, eplerenone, etc.]
  • (Ii) Calcium receptor antagonists [specifically, amlodipine, felodipine, nicardipine, nifedipine, nimodipine, nitrendipine, nilvadipine, alanidipine, azelnidipine, manidipine, varnidipine, efonidipine, cilnidipine, benidipine, diltiazem etc.], (synten) Converting enzyme inhibitors [specifically, captopril, lisinopril,
  • Non-steroidal anti-inflammatory drugs [specifically, meloxicam, teoxicam, indomethacin, ibuprofen, celecoxib, rofecoxib, aspirin, indomethacin, etc.], (43) disease-modifying anti-rheumatic drugs, (44) anti-cytokine drugs [44] Specifically, TNF inhibitors, MAP kinase inhibitors], (45) steroid drugs [specifically, dexamethasone, hexestrol, cortisone acetate, etc.], (46) sex hormones or derivatives thereof [specifically, , Progesterone, estradiol, estradiol benzoate, etc.], (47) parathyroid hormone, (48) opioid agonist [specifically, morphine, pentazocine, tramadol], (49) pilin antipyretic analgesic [specifically, Sulpyrine], (50) non-pyrine antipyretic analgesics [specific Acetamin
  • the combination method using the drug is not limited to the above diseases, and the drug used in combination is not limited to the compounds exemplified above.
  • the compound of the present invention when used in combination with a drug used in combination, it may be a separate preparation or a combination. Moreover, in separate preparations, both can be taken simultaneously or can be administered at different times.
  • the compounds of the present invention can be administered either alone or in combination with a pharmaceutically acceptable carrier, either single or multiple doses.
  • suitable pharmaceutical carriers include inert solid diluents or fillers, sterile aqueous solution, and various organic solvents.
  • the pharmaceutical composition thereby formed can then be easily administered in various dosage forms such as tablets, powders, lozenges, liquid preparations, syrups, injection solutions and the like.
  • These pharmaceutical compositions can optionally contain additional ingredients such as flavoring agents, binders, excipients and the like.
  • the compounds of the present invention may be used for oral, buccal, nasal, parenteral (eg, intravenous, intramuscular, or subcutaneous), transdermal (eg, patch), or rectal administration, or by inhalation or insufflation.
  • parenteral eg, intravenous, intramuscular, or subcutaneous
  • transdermal eg, patch
  • rectal administration or by inhalation or insufflation.
  • the administration mode of the compound of the present invention and the concomitant drug is not particularly limited as long as the compound of the present invention and the concomitant drug are combined at the time of administration.
  • Such dosage forms include, for example, (1) administration of a single preparation obtained by simultaneously formulating the compound of the present invention and a concomitant drug, (2) Simultaneous administration by the same route of administration of two types of preparations obtained by separately formulating the compound of the present invention and a concomitant drug, (3) Administration of two types of preparations obtained by separately formulating the compound of the present invention and a concomitant drug at the same administration route with a time difference, (4) Simultaneous administration of two types of preparations obtained by separately formulating the compound of the present invention and the concomitant drug by different administration routes, (5) Administration of two types of preparations obtained by separately formulating the compound of the present invention and a concomitant drug at different time intervals in different administration routes (for example, administration in the
  • the concomitant drug and the compound of the present invention may be administered at the same time, but after administering the concomitant drug, the compound of the present invention may be administered.
  • a concomitant drug may be administered after administration of the compound of the invention.
  • the time difference varies depending on the active ingredient to be administered, dosage form, and administration method.
  • administering the concomitant drug first preferably within 1 minute to 3 days after administering the concomitant drug.
  • the concomitant drug is administered within 1 minute to 1 day, preferably within 10 minutes to 6 hours, more preferably within 15 minutes to 1 hour after the administration of the compound of the present invention. The method of doing is mentioned.
  • the daily dose as a concomitant drug varies depending on the administration subject, administration route, target disease, symptom, etc., for example, when orally administered to a patient with schizophrenia (adult, body weight about 60 kg), Usually, the dose is about 0.1 to about 20 mg / kg body weight, preferably about 0.2 to about 10 mg / kg body weight, more preferably about 0.5 to about 10 mg / kg body weight. It is desirable to administer once to several times (eg, 2, 3, 4 or 8 times). When the compound of the present invention is used in combination with a concomitant drug, the amount of each agent can be reduced within a safe range in consideration of the opposite effect of those agents.
  • the concomitant drug of the present invention has low toxicity.
  • the compound of the present invention, or (and) the above concomitant drug is mixed with a pharmacologically acceptable carrier according to a known method, for example, a pharmaceutical composition such as a tablet.
  • a pharmaceutical composition such as a tablet.
  • a pharmaceutical composition such as a tablet.
  • a pharmaceutical composition such as a tablet.
  • a pharmaceutical composition such as a tablet.
  • a pharmaceutical composition such as a tablet.
  • a pharmaceutical composition such as a tablet.
  • a pharmaceutical composition such as a tablet.
  • a pharmaceutical composition such as a tablet.
  • a pharmaceutical composition such as a tablet.
  • a pharmaceutical composition such as a tablet.
  • powders, granules, capsules (including soft capsules), liquids, injections, suppositories, sustained-release agents, and the like which can be oral or parenteral (Eg, topical, rectal, intravenous, etc.).
  • the same carriers as those used for the pharmaceutical composition of the present invention described above can be used.
  • the compounding ratio of the compound of the present invention and the concomitant drug in the concomitant drug of the present invention can be appropriately selected depending on the administration subject, administration route, disease and the like. Two or more of the above concomitant drugs may be used in combination at an appropriate ratio.
  • the dose of the concomitant drug can be appropriately selected based on the clinically used dose.
  • the compounding ratio of the compound of the present invention and the concomitant drug can be appropriately selected depending on the administration subject, administration route, target disease, symptom, combination and the like.
  • the administration subject is a human
  • 0.01 to 100 parts by mass of the concomitant drug may be used per 1 part by mass of the compound of the present invention.
  • the content of the compound of the present invention in the concomitant drug of the present invention varies depending on the form of the preparation, but is usually in the range of about 0.01 to 99.9% by mass, preferably about 0, based on the whole preparation. The range is from 1 to 50% by mass, and more preferably from about 0.5 to 20% by mass.
  • the content of the concomitant drug in the concomitant drug of the present invention varies depending on the form of the preparation, but is usually in the range of about 0.01 to 99.9% by mass, preferably about 0.1 to about It is in the range of 50% by weight, more preferably in the range of about 0.5 to about 20% by weight.
  • the content of an additive such as a carrier in the combination agent of the present invention varies depending on the form of the preparation, but is usually in the range of about 1 to 99.99% by mass, preferably about 10 to about 90% with respect to the whole preparation. It is the range of mass%.
  • the same content may be used when the compound of the present invention and the concomitant drug are formulated separately. As described above, since the dosage varies depending on various conditions, an amount smaller than the above dosage may be sufficient, and it may be necessary to administer beyond the range.
  • the medicament of the present invention is administered in the form of a pharmaceutical composition.
  • the pharmaceutical composition of the present invention only needs to contain at least one of the compounds represented by the formula (I) of the present invention, and is optionally combined with a pharmaceutically acceptable additive.
  • excipients eg; lactose, sucrose, mannitol, crystalline cellulose, silicic acid, corn starch, potato starch
  • binders eg; celluloses (hydroxypropylcellulose (HPC), hydroxypropylmethylcellulose (HPMC)), crystalline cellulose, saccharides (lactose, mannitol, sucrose, sorbitol, erythritol, xylitol), starches (corn starch, potato starch), pregelatinized starch, dextrin, polyvinylpyrrolidone (PVP), macrogol, polyvinyl Alcohol (PVA)), lubricants (eg; magnesium stearate, calcium stearate, talc, carboxymethylcellulose), disintegrants (eg; starches (corn starch, potato starch), carboxymethyl starch sodium, potassium Lumellose, carmellose calcium, croscarmellose sodium, crospovidone), coating agent (eg, cellulose
  • triethyl citrate, macrogol masking agents (e.g. titanium oxide), colorants, flavoring agents, preservatives (e.g. benzalkonium chloride, paraoxybenzoate), isotonic agents (e.g .; Glycerin, sodium chloride, calcium chloride, mannitol, glucose), pH adjuster (eg; buffer solution such as sodium hydroxide, potassium hydroxide, sodium carbonate, hydrochloric acid, sulfuric acid, phosphate buffer), stabilizer (eg; Sugar, sugar alcohol, xanthan gum), dispersant, antioxidant (eg Asco Binic acid, butylhydroxyanisole (BHA), propyl gallate, dl- ⁇ -tocopherol), buffer, preservative (eg, paraben, benzyl alcohol, benzalkonium chloride), fragrance (eg, vanillin, l-menthol) , Rose oil), solubilizers (eg, polyoxyethylene hydrogenated castor oil, polysorbate 80, polyethylene
  • Various dosage forms include, for example, tablets, capsules, granules, powders, pills, aerosols, inhalants, ointments, patches, suppositories, injections, troches, liquids, spirits, suspensions Agents, extracts, elixirs and the like.
  • the medicament of the present invention is, for example, oral, subcutaneous administration, intramuscular administration, intranasal administration, transdermal administration, intravenous administration, intraarterial administration, perineural administration, epidural administration, intradural administration. It can be administered to patients by intraventricular administration, rectal administration, inhalation and the like.
  • the compounds of the present invention can be formulated for parenteral administration by injection, including using conventional catheter techniques or infusion.
  • injectable formulations may be presented as unit dosage forms, for example, in ampoules or multi-dose containers, with the addition of preservatives.
  • These formulations can take the form of suspensions, solutions, or emulsions in oily or aqueous vehicles, and contain formulation agents such as suspending, stabilizing, and / or dispersing agents. be able to.
  • the active ingredient can be in powder form for reconstitution with a suitable vehicle, eg, sterile pyrogen-free water, before use.
  • the product solution is isolated and included in water (or other aqueous medium) in an amount sufficient to produce a solution of the strength required for oral or parenteral administration to the patient. It can be produced by dissolving the complex.
  • These compounds can be formulated into rapidly dispersed dosage forms (fddf) that are designed to release the active ingredient in the oral cavity. These formulations are often formulated using a matrix based on fast dissolving gelatin. These dosage forms are well known and can be used to deliver a wide range of drugs. Most rapid dispersion dosage forms utilize gelatin as a carrier or structure-forming agent. Gelatin is typically used to give a dosage form sufficient strength to prevent breakage when removed from the package, but once in the mouth, gelatin allows the dosage form to break down immediately. . Alternatively, various starches are used to achieve the same effect.
  • the compounds of the invention can also be formulated in rectal compositions such as suppositories or retention enemas, eg containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds of the invention may be administered in the form of a solution or suspension from a pump spray container which is squeezed or pumped by the patient, or a suitable propellant such as dichloromethane.
  • a suitable propellant such as dichloromethane.
  • the dosage unit can be determined by providing a valve that delivers a metered amount.
  • a pressurized container or nebulizer can contain a solution or suspension of the active compound.
  • Capsules and cartridges for use in an inhaler or insufflator should be formulated containing a mixed powder of a compound of the invention and a suitable powder base such as lactose or starch. Can do.
  • Aerosol formulations for treating the above-described conditions (eg, migraine) in the average adult preferably each metered dose or “puff” of the aerosol contains from about 20 mg to about 1000 mg of the compound of the invention. Is set as follows.
  • the total daily dose with an aerosol will be within the range of about 100 mg to about 10 mg.
  • Administration can be several times daily, for example 2, 3, 4 or 8 times, for example 1, 2 or 3 doses each time.
  • Proposed daily doses of the compounds of the invention administered orally, parenterally, rectally, or buccally to the average adult to treat the above conditions are, for example, units that can be administered 1 to 4 times daily From about 0.01 mg to about 2000 mg, preferably from about 0.1 mg to about 200 mg, of the active ingredient of formula (I) per dose.
  • OXR antagonistic activity of the test compounds OX2R and IC 50 values of OXlR (an IC 50 value is ++ with 100 nmol / L the following compounds, an IC 50 value is the compound of greater than 1000 nmol / L or less of the present invention 100 nmol / L + The compounds with IC 50 values greater than 1000 nmol / L are expressed as ⁇ ).
  • ADL-OXB is a peptide in which two amino acids of human orexin B are substituted, and has a 400 times higher affinity for OX2R than OX1R.
  • rats are cannulated and kept for at least 1 week as a recovery period.
  • 100 ng / 5 ⁇ L of angiotensin 2 (Peptide Institute) is administered intraventricularly as a preliminary test, and the amount of water consumed for 30 minutes after the administration is measured. Only rats that have reached 5 g of drinking water are used in this study. Rats are acclimated for at least 120 minutes in the measurement cage until this test.
  • either vehicle or test compound is administered and immediately returned to the measurement cage.
  • 30 to 120 minutes after administration of the test compound the rat is again taken out from the housing cage, and the solvent (saline) or ADL-OXB (3 nmol / 5 ⁇ L / rat) is administered into the ventricle and immediately returned to the measurement cage.
  • Spontaneous momentum is measured in a spontaneous momentum measurement chamber (Muromachi Kikai) equipped with an infrared sensor. Spontaneous exercise is counted every 10 minutes, and cumulative counts for 30, 60, 120 minutes after ADL-OXB administration are calculated for each treatment group. All data are expressed as mean and standard error of the mean.
  • comparison between the control group and ADL-OXB single administration group was performed using Student's t-test (significant difference at p ⁇ 0.05), and comparison between ADL-OXB single administration group and test compound administration group Uses Dunnett's test (significantly different at p ⁇ 0.05).
  • Pharmacological experiment example 3 Sleep EEG measurement test After purchasing male Sprague-Dawley rats and arriving at the breeding facility, the animals are used for experiments with an acclimatization period of at least one week. Animals are housed in a laboratory with controlled temperature and humidity under a 12 hour light / dark cycle, with free access to food and water. In order to confirm the effect on sleep, a sleep electroencephalogram test in rats is performed. Rats are subjected to EEG and EMG electrode implantation surgery for electroencephalogram (EEG) and electromyogram (EMG) measurement, and are kept for at least 1 week as a recovery period. After administration of vehicle or test compound, EEG and EMG signals are recorded for 6-12 hours.
  • EEG electroencephalogram
  • EMG electromyogram
  • the analysis is performed by using an automatic analysis software SleepSign (registered trademark) (Kissei Comtech) to analyze an electroencephalogram frequency and an electromyogram activity signal to classify one of three stages of arousal, REM sleep, and NREM sleep.
  • the cumulative time of each stage is calculated, and the sleep action of the test compound is confirmed from the decrease in the awakening time and the increase in the total sleep time (REM sleep + NREM sleep).
  • the percentage of REM sleep with respect to the total sleep time is calculated, and it is examined whether a physiological sleep pattern is shown by comparing with the result of the control group.
  • DMSO Precipitation Solubility (Kinetic Solubility) A 10 mM DMSO solution of the compound of the present invention is added to 50 mM phosphate buffer (pH 7.4) to a final concentration of 100 ⁇ M. The solution was incubated at 600 rpm with stirring at room temperature for 1.5 hours, then filtered through a filter plate (MultiScreen HTS- PCF filter plate (Merck Millipore)), and using a plate reader (Powerscan HT (Dainippon Pharmaceutical)). Then, the absorbance of the filtrate is measured at the maximum absorption wavelength.
  • a filter plate MultiScreen HTS- PCF filter plate (Merck Millipore)
  • Powerscan HT Powerscan HT (Dainippon Pharmaceutical)
  • the absorbance of each standard solution is measured using a DMSO solution to which a known concentration (1, 3, 10, 30, 100 ⁇ M) of the test compound is added as a calibration curve standard solution, and a calibration curve is created.
  • the solubility ( ⁇ M) of the compound is calculated from the absorbance values of the filtrate and standard solution.
  • Crystal solubility Thermodynamic Solubility
  • the compound of the present invention is added to a solvent (eg, water, buffer) so as to be 1 mg / mL. The solution is incubated with stirring at 1000 rpm for 24 hours at 25 ° C. or 37 ° C. and then filtered through a filter plate.
  • the filtrate is analyzed by HPLC, the peak is detected at the maximum absorption wavelength, and the peak area is measured.
  • a solution eg, DMSO solution, 1,4-dioxane solution
  • a known concentration of the test compound eg, 0.01, 0.03, 0.1, 0.3, 1, 3, 10 ⁇ g / mL
  • Methanol solution is used as a standard curve standard solution to measure each peak area, and the solubility ( ⁇ g / mL) of the compound is calculated from the peak area of the standard curve.
  • Metabolic stability test (1) Liver microsomal solution (human, rat, mouse, dog or monkey; XenoTech), NADPH generating solution ( ⁇ -NADP, Glucose-6-phosphate, G- 6-PDH (Y), water containing MgCl 2 ). The solution is incubated at 37 ° C. for 5, 10, 20 or 30 minutes and then quenched with acetonitrile. The reaction solution is centrifuged through a filter plate (MultiScreen HTS- HV plate (Merck Millipore)), and the test compound in the filtrate is measured using high performance liquid chromatogram / mass spectrometry.
  • a filter plate MultiScreen HTS- HV plate (Merck Millipore
  • the reaction solution is centrifuged through a filter plate (MultiScreenHTS-HV plate (Merck Millipore)), and the test compound in the filtrate is measured using high performance liquid chromatogram / mass spectrometry.
  • a sample with a reaction time of 0 minutes is measured as a control, and the residual rate (%) at each time point is calculated from the ratio of the hepatocyte reaction sample and the control.
  • the reaction time is plotted on the horizontal axis and the residual ratio is plotted on the vertical axis, and the clearance ( ⁇ L / min / mg protein) is calculated from the slope.
  • hERG Inhibition Test by Patch Clamp Method The effect on hERG (human ether-a-go related gene) channels is measured using a fully automatic patch clamp system (QPatch HT (Sophion Bioscience)).
  • QPatch HT fully automatic patch clamp system
  • the membrane potential is held at ⁇ 80 mV, followed by a depolarization pulse of ⁇ 50 mV, 0.02 seconds and 20 mV, 4.8 seconds. -50 mV, 5 seconds of repolarization pulse given once every 15 seconds.
  • the effect of the test compound on the hERG channel is confirmed by the tail current change induced by the repolarization pulse.
  • the measurement is performed at room temperature.
  • the hERG channel inhibition rate is calculated as the reduction rate (suppression rate) of the tail current 4 minutes after the addition relative to the tail current peak value before the addition of the test compound. By calculating this suppression rate, the possibility of inducing QT prolongation by drugs and subsequent fatal side effects (such as ventricular tachycardia and sudden death) is shown.
  • PK test The compound of the present invention was applied to animals (male Crl: CD (SD) rats of about 7 to 8 weeks of age, male beagle dogs of about 4 years of age, or male cynomolgus monkeys of about 6 years of age).
  • test compounds 0.001, 0.002, 0.005, 0.01, 0.02, 0.05, 0.1, 0.2, 0.5, 1, 2, 10 ⁇ g / (mL) is measured, the plasma concentration ( ⁇ g / mL) is calculated from the prepared calibration curve, and the maximum plasma concentration is defined as Cmax ( ⁇ g / mL).
  • Pharmacological experiment example 9 Calculation of various parameters in pharmacokinetics test Model-independent analysis of the time course of plasma concentration obtained by PK test (the above pharmacological experiment example 7) in rat, dog and monkey animal species Systemic clearance CLtot (L / kg / hr), distribution volume Vdss in steady state (L / kg), plasma concentration-time curve area AUC ( ⁇ g ⁇ hr / mL), half-life T1 / 2 (hr) Is calculated.
  • the bioavailability is calculated by comparing the AUC at the time of intravenous administration with the AUC at the time of oral administration.
  • Pharmacological experiment example 10 Prediction of pharmacokinetic parameters in humans Various parameters in animal pharmacokinetic tests, metabolic stability in in vitro tests, proteins obtained by the method described in pharmacological experiment examples 5, 7 or 8 above Using parameters such as binding rate, pharmacokinetic parameters in humans are predicted by methods known to those skilled in the art such as allometric scaling or IVIVE (in vitro / in vivo extrapolation).
  • Pharmacological experiment example 11 Safety test The compound of the present invention is orally administered to a mouse or rat once, and no deaths are observed, and no remarkable behavioral abnormality is observed. It is.
  • the compound of the present invention has an excellent orexin receptor antagonistic action. Furthermore, orexin antagonism is shown by an antagonism test against [Ala 11 , D-Leu 15 ] -orexin B-induced locomotor activity enhancement using rats, and sleep action is shown by a sleep electroencephalogram measurement test. In addition, no abnormality is observed in the safety test, indicating the low toxicity of the present invention. Furthermore, it is confirmed that the compound of the present invention is good in one point such as solubility, metabolic stability, pharmacokinetics, and avoidance of hERG channel inhibitory effect by conducting the above test.
  • the compounds of the present invention are orexin receptor antagonists, such as sleep disorders (insomnia, circadian rhythm sleep disorders, sleep-related complications, etc.), mental disorders (depression, anxiety disorders, bipolar disorders, attention deficits Prevention of diseases such as mobility disorders, autism, autism spectrum disorders, drug addiction, etc.), neurodegenerative diseases (such as Alzheimer's disease), memory disorders (such as dementia), and eating disorders (such as bulimia) And / or expected to be used in therapeutic agents.
  • the compounds of the present invention are expected to show promising preventive or therapeutic effects for various diseases shown below.
  • insomnia circadian rhythm sleep disorder
  • sleep-related comorbidities depression, anxiety disorder, bipolar disorder, attention deficit hyperactivity disorder, autism, autism spectrum disorder, drug addiction
  • Promising therapeutic effects can be expected for Alzheimer's disease, dementia, bulimia and the like.
  • JEOL JNM-ECX400 FT-NMR (JEOL) or JEOL JNM-ECX300 FT-NMR (JEOL) was used.
  • Liquid chromatography-mass spectrometry spectrum (LC-Mass) was measured by one of the following methods.
  • Root temperature in the examples and production examples represents the temperature in the laboratory, usually 20 ⁇ 15 ° C.
  • Step 2 Synthesis of 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane hydrochloride (intermediate 1) tert-butyl 5- (4,6 -Dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 1-1) (0.50 g) in 4N hydrochloric acid-dioxane solution (3.0 mL) ) And stirred at room temperature for 17 hours. The solvent was distilled off under reduced pressure to obtain the title compound (0.40 g) as a white solid.
  • Step 4> of tert-butyl 5- (4-cyano-3-methoxypyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 2-4) Synthesis 2-chloro-3-methoxyisonicotinonitrile (intermediate 2-3) (0.26 g), tert-butyl 2,5-diazabicyclo [2.2.2] octane-2-carboxylate (0.30 g) ), RuPhos (66 mg), and a mixed solution of cesium carbonate (0.69 g) in tetrahydrofuran (3.0 mL) were added with RuPhos Pd G2 (0.11 g) and stirred at 90 ° C.
  • Step 5 Synthesis of 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) -3-methoxyisonicotinonitrile (intermediate 2) tert-butyl 5- (4-cyano- 3-methoxypyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 2-4) (0.13 g) was added trifluoroacetic acid (1.0 mL). The mixture was further stirred at room temperature for 1 hour.
  • the solvent was evaporated under reduced pressure, 1N aqueous hydrochloric acid solution was added to the resulting residue, and the mixture was washed with an ethyl acetate-heptane mixed solvent (1: 1).
  • the aqueous layer was basified with 1N aqueous sodium hydroxide solution, sodium chloride was added, and the mixture was extracted 5 times with ethyl acetate.
  • the organic layer was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to obtain the title compound (21 mg) as a pale yellow gum.
  • Step 2> Synthesis of 1- (pyridin-2-yl) -1H-pyrazole-5-carboxylic acid (intermediate 4) Methyl 1- (pyridin-2-yl) -1H-pyrazole-5-carboxylate (intermediate) Lithium hydroxide monohydrate (0.11 g) was added to a THF (2.0 mL) -water (0.50 mL) mixed solution of Compound 3-1) (0.35 g), and the mixture was heated at 60 ° C. under a nitrogen atmosphere. Stir for 2 hours. 1N Hydrochloric acid aqueous solution was added to stop the reaction, and the mixture was extracted with ethyl acetate and washed with saturated brine.
  • Step 3> Synthesis of 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane optical isomer (intermediate 5-B) Step of Production Example 5 2 or a method analogous thereto, tert-butyl 5- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2] obtained in Step 1 of Production Example 5 was used. .2] The title compound (85 mg) was obtained as a pale yellow solid from the optical isomer of Octane-2-carboxylate (Intermediate 5-1-B) (0.13 g). UPLC: 219 [M + H] + (retention time 0.51 minutes)
  • Step 2 Synthesis of 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) -3-methoxyisonicotinonitrile optical isomer (intermediate 6-A)
  • tert-butyl 5- (4-cyano-3-methoxypyridin-2-yl) -2,5-diazabicyclo [O] obtained in Step 1 of Production Example 6 2.2.2
  • the title compound (40 mg) was obtained as an orange gum from the optical isomer of the octane-2-carboxylate (6-1-A) (60 mg).
  • Step 3 Synthesis of optical isomer of 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotinonitrile (intermediate 6-B)
  • tert-butyl 5- (4-cyano-3-methoxypyridin-2-yl) -2,5-diazabicyclo [O] obtained in Step 1 of Production Example 6 2.2.2
  • the title compound (41 mg) was obtained as an orange gum from the optical isomer of the octane-2-carboxylate (6-1-B) (58 mg).
  • tert-butyl 5- (4-cyano-3-cyclopropylpyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 7-2)
  • tert-butyl 5- (3-chloro-4-cyanopyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 7-1) (0.
  • tetrakis (triphenylphosphine) palladium (0.35 g) was added to a THF (2.5 mL) mixed solution of cyclopropyl zinc bromide (0.5 N THF solution) (9 mL), and the mixture was heated to 100 ° C. under a nitrogen atmosphere. And stirred for 15 hours. The reaction solution was returned to room temperature, saturated aqueous ammonium chloride solution was added, and the mixture was extracted with ethyl acetate.
  • Step 5 Synthesis of optical isomer of 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropylisonicotinonitrile (intermediate 7-B)
  • Production Example 5 Tert-Butyl 5- (4-cyano-3-cyclopropylpyridin-2-yl) -2,5- obtained in Step 3 of Production Example 7 in the same manner as in Step 2 above or a method analogous thereto.
  • the title compound (42 mg) was obtained as a yellow oil from the optical isomer of diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 7-3-B) (52 mg).
  • Example 1 (5- (4,6-Dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-yl) (5-methyl-2- (2H-1 , 2,3-Triazol-2-yl) phenyl) methanone tert-butyl 5- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2- Trifluoroacetic acid (0.88 mL) was added to a dichloromethane solution of carboxylate (intermediate 1-1) (0.11 g), and the reaction solution was stirred at room temperature for 2 hours.
  • Example 7 2- (5- (2- (2H-1,2,3-triazol-2-yl) benzoyl) -2,5-diazabicyclo [2.2.2] octane-2-yl)- Synthesis of 3-methoxyisonicotinonitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotinonitrile (intermediate 2) (11 mg), 2- ( To a mixed solution of 2H-1,2,3-triazol-2-yl) benzoic acid (9.5 mg) and HATU (21 mg) in DMF (1.0 mL) was added triethylamine (9.5 ⁇ L), and the mixture was stirred at room temperature for 64 hours.
  • Example 8 2- (5- (2- (1H-pyrazol-1-yl) nicotinoyl) -2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotino Synthesis of nitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotinonitrile (intermediate 2) (25 mg), 2- (1H-pyrazole-1- Il) 2,6-lutidine (36 ⁇ L) was added to a mixed solution of nicotinic acid (23 mg) and HATU (58 mg) in dichloromethane (1.0 mL), and the mixture was stirred at room temperature for 20 hours under a nitrogen atmosphere.
  • Example 11 (5- (4,6-Dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-yl) (1- (pyridin-2-yl)- Synthesis of 1H-pyrazol-5-yl) methanone optical isomer 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane optical isomer (intermediate 5 -B) (35 mg), 1- (pyridin-2-yl) -1H-pyrazole-5-carboxylic acid (Intermediate 4) (31 mg), in a mixed solution of DMF (1.0 mL), HATU (63 mg), diisopropyl Ethylamine (72 ⁇ L) was added and stirred at room temperature for 30 minutes.
  • Example 12 (5- (2- (2H-1,2,3-triazol-2-yl) benzoyl) -2,5-diazabicyclo [2.2.2] octane-2-yl)- Synthesis of 3-methoxyisonicotinonitrile optical isomer 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) -3 in the same manner as in Example 7 or a method analogous thereto The title compound (13 mg) was obtained as a colorless amorphous form from the optical isomer of 2-methoxyisonicotinonitrile (Intermediate 6-A) (14 mg). UPLC: 416 [M + H] + (retention time 1.02 minutes) 1 H-NMR was consistent with (Example 7).
  • Example 13 2- (5- (2- (2H-1,2,3-triazol-2-yl) benzoyl) -2,5-diazabicyclo [2.2.2] octane-2-yl)- Synthesis of 3-methoxyisonicotinonitrile optical isomer 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) -3 in the same manner as in Example 7 or a method analogous thereto The title compound (7.7 mg) was obtained as a colorless amorphous form from the optical isomer of 2-methoxyisonicotinonitrile (intermediate 6-B) (18 mg). UPLC: 416 [M + H] + (retention time 1.02 minutes) 1 H-NMR was consistent with (Example 7).
  • Example 14 (5- (2- (1H-pyrazol-1-yl) nicotinoyl) -2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotino
  • optical isomer of nitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotino by the same method as in Example 7 or a method analogous thereto
  • the title compound (6.2 mg) was obtained as a colorless amorphous form from the optical isomer of nitrile (Intermediate 6-A) (12 mg).
  • Example 15 2- (5- (2- (1H-pyrazol-1-yl) nicotinoyl) -2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotino Synthesis of optical isomer of nitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotino by the same method as in Example 7 or a method analogous thereto The title compound (9.6 mg) was obtained as a colorless amorphous form from the optical isomer of nitrile (Intermediate 6-B) (12 mg). UPLC: 416 [M + H] + (retention time 0.99, 1.01 min, rotamer) 1 H-NMR was consistent with (Example 8).
  • Example 16 2- (5- (2- (1H-pyrazol-1-yl) nicotinoyl) -2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropylisonicoti Synthesis of optical isomers of nononitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropylisonicotinonitrile optical isomer (intermediate 7-A) ( 22 mg), 2- (1H-pyrazol-1-yl) nicotinic acid (16 mg), and HATU (40 mg) in dichloromethane (2 mL) were added with 2,6-lutidine (20 ⁇ L), and a nitrogen atmosphere at room temperature was added.
  • Example 17 2- (5- (2- (1H-pyrazol-1-yl) nicotinoyl) -2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropylisonicoti Synthesis of optical isomers of nononitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropyliso-isolated by a method similar to or equivalent to that in Example 16. The title compound (26 mg) was obtained as a colorless amorphous form from the optical isomer of nicotinonitrile (Intermediate 7-B) (21 mg). UPLC: 426 [M + H] + (retention time 1.02, 1.06 min, rotamer)
  • Example 18 2- (5- (2- (2H-1,2,3-triazol-2-yl) benzoyl) -2,5-diazabicyclo [2.2.2] octane-2-yl)- Synthesis of optical isomers of 3-cyclopropylisonicotinonitrile 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) in the same manner as in Example 16 or a method analogous thereto The title compound (26 mg) was obtained as a colorless amorphous form from the optical isomer of -3-cyclopropylisonicotinonitrile (Intermediate 7-A) (22 mg).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Provided are: a compound having an antagonistic activity on an orexin receptor, a pharmaceutically acceptable salt of the compound, or a solvate of the compound or the pharmaceutically acceptable salt; a pharmaceutical composition characterized by containing the compound, the pharmaceutically acceptable salt or the solvate as an active ingredient; and a use of the compound or the pharmaceutical composition for medical purposes, specifically a use of the compound or the pharmaceutical composition as a prophylactic and/or therapeutic agent for diseases associated with an orexin receptor, such as sleep disorders, mental diseases, neurodegenerative diseases, memory impairments and eating disorders, particularly insomnia (an insomniac symptom) which is one of sleep disorders. Specifically provided is a compound represented by formula (I), a pharmaceutically acceptable salt of the compound, or a solvate of the compound or the pharmaceutically acceptable salt.

Description

新規ジアザビシクロ[2.2.2]オクタン誘導体Novel diazabicyclo [2.2.2] octane derivatives
 本発明は、オレキシン受容体拮抗作用を有する化合物、とりわけ、下記式(I)で表される2,5-ジアザビシクロ[2.2.2]オクタン構造を持つことを特徴とする化合物、またはそれらの製薬学的に許容される塩、またはそれらの溶媒和物、および、それらを有効成分として含有することを特徴とする医薬組成物に関する。 The present invention relates to a compound having an orexin receptor antagonistic action, particularly a compound having a 2,5-diazabicyclo [2.2.2] octane structure represented by the following formula (I), or a compound thereof: The present invention relates to a pharmaceutically acceptable salt, or a solvate thereof, and a pharmaceutical composition characterized by containing them as an active ingredient.
 オレキシン(以下、「OX」と略記する場合がある)は、視床下部外側野に存在する神経細胞に特異的に発現している神経ペプチドであり、33個のアミノ酸からなるOX-Aおよび28個のアミノ酸からなるOX-Bが同定されている。これらペプチドの特異的受容体であるOX受容体(以下、「OXR」と略記する場合がある)は、OX1受容体(以下、「OX1R」と略記する場合がある)、およびOX2受容体(以下、「OX2R」と略記する場合がある)の2つのサブタイプが報告されている。いずれの受容体も中枢神経系に発現する7回膜貫通型のGタンパク質共役型受容体(GPCR)であるが、サブタイプによって組織分布は異なり、様々な神経に多様な影響を与えていることから、オレキシンが複雑な生理活性を有することが示唆されている。 Orexin (hereinafter sometimes abbreviated as “OX”) is a neuropeptide specifically expressed in neurons existing in the lateral hypothalamic area, OX-A consisting of 33 amino acids and 28 OX-B consisting of the following amino acids has been identified. OX receptors (hereinafter sometimes abbreviated as “OXR”), which are specific receptors for these peptides, are OX1 receptor (hereinafter sometimes abbreviated as “OX1R”), and OX2 receptor (hereinafter, abbreviated as “OXR”). Two subtypes have been reported (sometimes abbreviated as “OX2R”). All receptors are 7-transmembrane G protein-coupled receptors (GPCRs) expressed in the central nervous system, but the tissue distribution varies depending on the subtype and has various effects on various nerves. This suggests that orexin has complex physiological activities.
 睡眠障害の1つとされる不眠症(不眠症状)は、日本人の5人に1人が罹患していると言われる疾患である。不眠症が長期に及ぶと、日中の日常生活や就労で支障をきたすだけでなく、精神的・身体的苦痛が生じる場合もある為、質的・量的にも十分な睡眠が確保できるような治療(薬物療法、または非薬物療法等)が必要となる。現在、不眠症の薬物療法で処方頻度の高い薬剤としては、ベンゾジアゼピン(以下、「BZ」と略記する場合がある)系薬等があるが、長期の使用では耐性や反跳性不眠、離脱症状、乱用等の問題が生じやすい傾向があり、欧米ではNIH等の機関から、BZ系薬剤(超短時間型BZ系薬剤であるゾルピデム、ゾピクロン等も含め)の、長期に及ぶ使用は避けるべきとの勧告がなされている。そこで、既存の睡眠薬の副作用・問題点を解決すべく、新しい作用機序を有する睡眠薬の開発が求められている。 Insomnia (insomnia symptoms), which is one of the sleep disorders, is a disease that is said to affect one out of five Japanese people. Long-term insomnia not only interferes with daily life and work during the day, but also may cause mental and physical distress, so that sufficient qualitative and quantitative sleep can be ensured. Treatment (such as drug therapy or non-drug therapy) is required. Currently, drugs with high prescription frequency for insomnia pharmacotherapy include benzodiazepines (hereinafter sometimes abbreviated as “BZ”), etc., but tolerance, rebound insomnia, withdrawal symptoms in long-term use , Abuse and other problems tend to occur. In Europe and the United States, long-term use of BZ drugs (including ultra-short-time BZ drugs zolpidem, zopiclone, etc.) from institutions such as NIH should be avoided. Recommendations have been made. Therefore, in order to solve the side effects and problems of existing hypnotic drugs, development of hypnotic drugs having a new mechanism of action is required.
 OXが同定された当初は、摂食行動の制御調節機能が注目されていたが、近年、OXと睡眠・覚醒の制御調節との関連が注目されている。ラット脳室内にOXを投与すると、OXRが活性化され、その結果、モノアミン/コリン作動性ニューロンが優位になり、自発運動量の亢進、覚醒時間の延長等の覚醒促進作用が認められる(非特許文献1及び非特許文献2)。また、OXノックアウトマウスやOX2Rノックアウトマウスでは、覚醒阻害が認められる(非特許文献3)。このような知見から、OXR拮抗剤は、覚醒状態を抑制することで睡眠を誘発する睡眠導入剤として、優れた不眠症治療薬になると考えられている。 At the beginning of the identification of OX, attention was paid to the control and regulation function of feeding behavior, but in recent years, the relationship between OX and the regulation and regulation of sleep / wakefulness has attracted attention. When OX is administered into the rat ventricle, OXR is activated, and as a result, monoamine / cholinergic neurons are dominant, and arousal promoting effects such as increased spontaneous momentum and prolonged awakening time are recognized (Non-Patent Document). 1 and Non-Patent Document 2). Moreover, in OX knockout mice and OX2R knockout mice, inhibition of arousal is observed (Non-patent Document 3). From such knowledge, it is considered that the OXR antagonist is an excellent therapeutic agent for insomnia as a sleep induction agent that induces sleep by suppressing the arousal state.
 OXR拮抗剤として、スボレキサント(商品名:ベルソムラ(登録商標))、およびアルモレキサント(ACT-078573、治験番号NCT00608985)が知られている。これらOXR拮抗剤はOX1R、およびOX2Rを同程度に拮抗する物質(デュアルオレキシン受容体拮抗剤(DORA))であり、不眠症状の改善が報告されている(非特許文献4、および非特許文献5)。 As OXR antagonists, suvorexant (trade name: Bersomura (registered trademark)) and almorexant (ACT-0778573, clinical trial number NCT00608985) are known. These OXR antagonists are substances (dual orexin receptor antagonists (DORA)) that antagonize OX1R and OX2R to the same extent, and improvement of insomnia has been reported (Non-patent Documents 4 and 5). ).
 スボレキサントは、既に日米で不眠症治療薬として製造販売承認が取得されている。しかし、其の使用上の注意の重要な基本的注意事項として、『本剤の影響が服用の翌朝以後に及び、眠気、注意力・集中力・反射運動能力等の低下が起こることがあるので、自動車の運転など危険を伴う機械の操作に従事させないように注意すること』(非特許文献6)とあり、その副作用が懸念される。 Suvorexant has already been approved for manufacture and sale as a treatment for insomnia in Japan and the United States. However, as an important basic precaution for its precautions, `` Because the effect of this drug extends after the morning after taking it, drowsiness, attention / concentration / reflex ability etc. may occur. ”Be careful not to engage in the operation of dangerous machinery such as driving a car” (Non-Patent Document 6), and there are concerns about its side effects.
 ノックアウトマウスを用いた実験結果から、OXによる覚醒作用やノンレム睡眠抑制作用は、主にOX2Rを介していることが報告されている(非特許文献7)。従って、OX2R選択的拮抗剤は、生理的な睡眠を誘導し、副作用リスクを低減させる不眠症治療薬になる可能性がある。OX2R選択的拮抗剤として、現在、MIN-202(Minerva Neurosciences.Inc.(http://www.minervaneurosciences.com))、およびMK-1064(Merck Sharp & Dohm Corp.)(非特許文献8)が不眠症治療薬として臨床開発されている。 From the results of experiments using knockout mice, it has been reported that the arousal action and non-REM sleep inhibitory action by OX are mainly mediated by OX2R (Non-patent Document 7). Therefore, an OX2R selective antagonist may be a therapeutic agent for insomnia that induces physiological sleep and reduces the risk of side effects. As OX2R selective antagonists, MIN-202 (Minerva Neurosciences. Inc. (http://www.minerneunosciences.com)) and MK-1064 (Merck Sharp & Dohm Corp.) (non-patent documents) are currently available. It has been clinically developed as a treatment for insomnia.
 国際公開第2008/008517号パンフレット(特許文献1)には、OXRアンタゴニストとして、分子内に架橋構造を有している化合物が開示されている。 International Publication No. 2008/008517 pamphlet (Patent Document 1) discloses a compound having a cross-linked structure in the molecule as an OXR antagonist.
 国際公開第2011/050198号パンフレット(特許文献2)、および国際公開第2012/145581号パンフレット(特許文献3)には、OXR調節因子として、分子内にオクタヒドロピロロ[3,4-c]ピロール構造を有している化合物が開示されている。 WO 2011/050198 pamphlet (Patent Document 2) and WO 2012/145581 pamphlet (Patent Document 3) include octahydropyrrolo [3,4-c] pyrrole in the molecule as an OXR regulator. A compound having a structure is disclosed.
 国際公開第2011/050200号パンフレット(特許文献4)、および国際公開第2011/050202号パンフレット(特許文献5)、には、OXR調節因子として、分子内に3,8-ジアザ-ビシクロ[4.2.0]オクタン、および3,6-ジアザビシクロ[3.2.0]ヘプタン構造を有している化合物が開示されている。しかしながら、これらに開示の化合物は、ジアザビシクロ[2.2.2]オクタン構造を持つ化合物とは基本骨格が異なっており、ジアザビシクロ[2.2.2]オクタン構造を持つ化合物については、開示も示唆もない。 In WO 2011/050200 (Patent Document 4) and WO 2011/050202 (Patent Document 5), as an OXR regulator, 3,8-diaza-bicyclo [4. Compounds having 2.0] octane and 3,6-diazabicyclo [3.2.0] heptane structures are disclosed. However, the compounds disclosed therein are different in basic skeleton from the compounds having a diazabicyclo [2.2.2] octane structure, and the compounds having a diazabicyclo [2.2.2] octane structure are also disclosed. Nor.
 国際公開第2011/034741号パンフレット(特許文献6)には、代謝型グルタミン酸受容体の増強剤として、5-置換-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボニル構造を部分構造として有する化合物が開示されているが、以下で示すような本発明の特定の化合物の開示はない。 In WO 2011/034741 (Patent Document 6), a 5-substituted-2,5-diazabicyclo [2.2.2] octane-2-carbonyl structure is partially used as a potentiator of metabotropic glutamate receptors. Although a compound having a structure is disclosed, there is no disclosure of a specific compound of the present invention as shown below.
 国際公開第2011/090935号パンフレット(特許文献7)には、mTOR(mammalian target of rapamycin)阻害剤として5-置換-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボニル構造を部分構造として有する化合物が開示されているが、以下で示すような本発明の特定の化合物の開示はない。 International Publication No. 2011/090935 (Patent Document 7) discloses a partial 5-substituted-2,5-diazabicyclo [2.2.2] octane-2-carbonyl structure as an mTOR (mammalian target of rapamycin) inhibitor. Although a compound having a structure is disclosed, there is no disclosure of a specific compound of the present invention as shown below.
 さて、医薬品開発においては、目的とする薬理活性のみでなく、吸収、分布、代謝、排泄等の各種の面で厳しいクライテリアを満たすことが要求される。例えば、薬物相互作用、脱感受性ないし耐性、経口投与時の消化管吸収、小腸内への移行速度、吸収速度と初回通過効果、臓器バリアー、蛋白結合、薬物代謝酵素の誘導や阻害、排泄経路や体内クリアランス、適用方法(適用部位、方法、目的)等において種々の検討課題が要求され、これらを満たすものはなかなか見出されない。しかしながら、医薬品における課題は常に生じるであろう。 Now, in drug development, it is required to satisfy severe criteria not only in the intended pharmacological activity but also in various aspects such as absorption, distribution, metabolism, and excretion. For example, drug interaction, desensitization or tolerance, digestive tract absorption after oral administration, transfer rate into the small intestine, absorption rate and first-pass effect, organ barrier, protein binding, induction and inhibition of drug metabolizing enzymes, excretion route and Various examination subjects are required in the body clearance, application method (application site, method, purpose) and the like, and it is difficult to find one that satisfies these. However, challenges in medicine will always arise.
国際公開第2008/008517号パンフレットInternational Publication No. 2008/008517 Pamphlet 国際公開第2011/050198号パンフレットInternational Publication No. 2011/050198 Pamphlet 国際公開第2012/145581号パンフレットInternational Publication No. 2012/145581 Pamphlet 国際公開第2011/050200号パンフレットInternational Publication No. 2011/050200 Pamphlet 国際公開第2011/050202号パンフレットInternational Publication No. 2011/050202 Pamphlet 国際公開第2011/034741号パンフレットInternational Publication No. 2011/034741 Pamphlet 国際公開第2011/090935号パンフレットInternational Publication No. 2011/090935 Pamphlet
 現在、睡眠障害の1つとされる不眠症(不眠症状)の薬物療法において、BZ系薬剤をはじめとする種々の不眠症治療薬が利用可能となっている。しかし一方で、薬剤に起因する副作用により、患者の治療満足度が低い状況であり、既存薬に比して、より良い薬剤プロファイルを有する新たな不眠症治療薬の開発が求められている。 Currently, various insomnia drugs including BZ drugs are available for pharmacotherapy of insomnia (insomnia symptoms), which is one of sleep disorders. On the other hand, however, the patient's treatment satisfaction is low due to side effects caused by the drug, and the development of a new insomnia drug having a better drug profile than that of existing drugs is required.
 OXR拮抗作用を有する化合物の報告例は複数あるが、上記の医薬品開発上の総合的課題は常にある。より具体的には、例えば、溶解性がよくないこと、代謝安定性が低く経口投与による全身曝露が困難であること、吸収性や持続性等の薬物動態が良好ではないこと、あるいは不整脈を起こす危険性があるhERG(human ether-a-go-go related gene)チャネルの阻害活性を示すこと、薬物代謝酵素(例えば、シトクロムP450等)の誘導あるいは阻害活性を示すことや高い蛋白結合率を示すなど、有用性や安全性の課題がある。これらの問題を可能な限り多く解決し、且つ有効性の高い化合物を見出すことが求められている。 There are multiple reports of compounds with OXR antagonistic activity, but there are always comprehensive issues in the above drug development. More specifically, for example, poor solubility, low metabolic stability, difficulty in systemic exposure by oral administration, poor pharmacokinetics such as absorption and persistence, or arrhythmia It exhibits hERG (human ether-a-go related gene) channel inhibitory activity, induces or inhibits drug metabolizing enzymes (eg, cytochrome P450), and exhibits a high protein binding rate. There are problems of usefulness and safety. There is a need to solve these problems as much as possible and find highly effective compounds.
 本発明者らは、上記の課題を解決すべく、安全性が高く、および/または有効性に優れたOXR拮抗剤を得るべく、鋭意研究を重ねてきた結果、式(I)で表される2,5-ジアザビシクロ[2.2.2]オクタン構造を持つことを特徴とする化合物、またはそれらの製薬学的に許容される塩、またはそれらの溶媒和物が、OXR拮抗作用を有することを見出した。本発明の化合物は、OXR拮抗作用を有し、不眠症等の睡眠障害を始めとする精神学的および神経学的障害の改善作用を有する。 In order to solve the above-mentioned problems, the present inventors have conducted extensive research to obtain an OXR antagonist that is highly safe and / or excellent in effectiveness, and as a result, is represented by the formula (I). That a compound having a 2,5-diazabicyclo [2.2.2] octane structure, or a pharmaceutically acceptable salt thereof, or a solvate thereof has an OXR antagonistic action. I found it. The compound of the present invention has an OXR antagonistic action, and has an action of improving psychiatric and neurological disorders including sleep disorders such as insomnia.
Figure JPOXMLDOC01-appb-C000003
 本発明は、式(I)で表される2,5-ジアザビシクロ[2.2.2]オクタン構造を持つことを特徴とする化合物、またはそれらの製薬学的に許容される塩、またはそれらの溶媒和物、および、それらを有効成分として含有することを特徴とする医薬組成物である。
Figure JPOXMLDOC01-appb-C000003
The present invention relates to a compound having a 2,5-diazabicyclo [2.2.2] octane structure represented by the formula (I), or a pharmaceutically acceptable salt thereof, or a A solvate and a pharmaceutical composition characterized by containing them as active ingredients.
 本発明の化合物は、OXR拮抗作用を有し、不眠症等の睡眠障害を始めとする精神学的および神経学的障害の改善作用を有している。また、本発明の化合物を有効成分として含有する医薬組成物は、経口投与可能であり、OXR拮抗剤や、OXRが関与する疾患、精神学的及び神経学的障害、とりわけ不眠症等の睡眠障害の予防、および/または治療剤として期待される。更に、本発明の化合物群は、溶解性が良好であること、代謝安定性が高いこと、優れた経口吸収性をもつこと、あるいはhERGチャネルの阻害作用が少ないこと、などの少なくとも一つ以上の特徴も有することから有用性が高い。 The compound of the present invention has an OXR antagonistic action, and has an action of improving psychological and neurological disorders including sleep disorders such as insomnia. In addition, the pharmaceutical composition containing the compound of the present invention as an active ingredient can be administered orally, and OXR antagonists, diseases involving OXR, psychiatric and neurological disorders, particularly sleep disorders such as insomnia It is expected as a preventive and / or therapeutic agent. Furthermore, the compound group of the present invention has at least one or more of such properties as good solubility, high metabolic stability, excellent oral absorption, and little hERG channel inhibitory action. Since it has characteristics, it is highly useful.
 本発明は、以下の態様に示される下記式(I)で表される2,5-ジアザビシクロ[2.2.2]オクタン構造を持つことを特徴とする化合物、またはそれらの製薬学的に許容される塩、またはそれらの溶媒和物、および、それらを有効成分として含有することを特徴とする医薬組成物、ならびにそれらの医薬用途、OXR拮抗剤である。 The present invention relates to a compound having a 2,5-diazabicyclo [2.2.2] octane structure represented by the following formula (I) shown in the following embodiments, or a pharmaceutically acceptable product thereof: Or a solvate thereof, and a pharmaceutical composition characterized by containing them as an active ingredient, and their pharmaceutical use, an OXR antagonist.
[本発明の態様]
 本発明は、以下の態様[1]~[14]を含む。
[1]本発明の第1の態様は、
下記式(I):
Figure JPOXMLDOC01-appb-C000004
[式(I)中、mは、0~4の整数であり;nは、0~4の整数であり;環Aは、C6~14アリール基、または5~7員ヘテロアリール基を表し;環Bは、C6~14アリール基、または5~7員ヘテロアリール基を表し; Q(該Qは、ジアザビシクロ[2.2.2]オクタン環と環Bを結合するアミド結合の隣接位(o-位)に必ず置換する:Qの置換位置は、後述の各態様中においても同定義とする)は、C3~8シクロアルキル基、または-ORc、または部分構造式(SF-1)で表される基:
Figure JPOXMLDOC01-appb-C000005
[式(SF-1)中、環Cは、C6~14アリール基、5~7員ヘテロアリール基、または3~8非芳香族複素環基を表し;pは、0~5の整数であり;R3は、各々独立に、ハロゲン原子、シアノ基、-Rd、または-ORdを表し、Rdは、C1~6アルキル基、ハロゲン化C1~6アルキル基、またはC3~8シクロアルキル基を表す]を表し;R1は、各々独立に、ハロゲン原子、シアノ基、-Ra、-ORa、-C(O)Ra、-C(O)ORa、-SRa、-SO2a、-NRAB基、3~8員非芳香族複素環基、またはオキソ基を表し; R2は、各々独立に、ハロゲン原子、シアノ基、-Rb、-ORb、または5~7員ヘテロアリール基表し、当該5~7員ヘテロアリール基は、1~5個のハロゲン原子、または水酸基で置換されていても良く;当該R1中の-NRAB基におけるRAおよびRBは、各々独立に、-Ra、-C(O)Ra、-C(O)ORa、または-SO2aを表し;当該R1および-NRAB基中におけるRaは、水素原子、C1~6アルキル基、ハロゲン化C1~6アルキル基、ヒドロキシC1~6アルキル基、ヒドロキシハロゲン化C1~6アルキル基、またはC3~8シクロアルキル基を表し;当該R2中のRbは、C1~6アルキル基、またはハロゲン化C1~6アルキル基を表し;当該Q中のRcは、C1~6アルキル基、ハロゲン化C1~6アルキル基、またはC6~14アリール基を表す]で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物である。
[Aspect of the Invention]
The present invention includes the following embodiments [1] to [14].
[1] The first aspect of the present invention is
The following formula (I):
Figure JPOXMLDOC01-appb-C000004
[In formula (I), m is an integer from 0 to 4; n is an integer of 0-4; ring A represents a C 6 ~ 14 aryl group or a 5-7 membered heteroaryl group, ; ring B represents a C 6 ~ 14 aryl group or a 5-7 membered heteroaryl group,; Q (the Q is diazabicyclo [2.2.2] amide bond at the adjacent position to couple the octane ring and ring B (o-position) to always replace: the substitution position of Q is also the same defined during each aspect described below), the C 3 ~ 8 cycloalkyl group, or -OR c or partial structural formula,, (SF- Group represented by 1):
Figure JPOXMLDOC01-appb-C000005
Wherein (SF-1), ring C is C 6 ~ 14 aryl group, 5- to 7-membered heteroaryl group or 3-8 represents a non-aromatic heterocyclic group,; p is an integer of 0 to 5 Yes; each R 3 independently represents a halogen atom, a cyano group, —R d , or —OR d , where R d is a C 1-6 alkyl group, a halogenated C 1-6 alkyl group, or C 3 And R 1 each independently represents a halogen atom, a cyano group, —R a , —OR a , —C (O) R a , —C (O) OR a , — SR a represents —SO 2 R a , —NR A R B group, 3- to 8-membered non-aromatic heterocyclic group, or oxo group; R 2 each independently represents a halogen atom, a cyano group, —R b represents -OR b or 5-7 membered heteroaryl group, the 5- to 7-membered heteroaryl group optionally substituted with one to five halogen atoms or hydroxyl, At best, the and R A in -NR A R B group in R 1 R B are each independently, -R a, -C (O) R a, -C (O) OR a , or -SO, 2 R a represents; R a in the R 1 and in -NR a R B group, a hydrogen atom, C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl group, hydroxy C 1 ~ 6 alkyl group, hydroxy halogenated C 1 ~ 6 alkyl group or an C 3 ~ 8 cycloalkyl group; R b in the R 2 represents a C 1 ~ 6 alkyl group or halogenated C 1 ~ 6 alkyl group; the Q R c in the, C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl group or C 6 ~ 14 compounds are represented by an aryl group, or a pharmaceutically acceptable salt thereof, or, The solvate.
 以下に、上記態様[1]の上記式(I)中の各基について具体的に説明する。
 本発明の化合物に関する説明において、例えば「C1~6」とは、構成炭素原子数が1から6であることを示し、特に断らない限り、直鎖、分枝鎖または環状の基の炭素原子数を表す。鎖状の基については「構成炭素原子数が1ないし6の直鎖または分枝鎖」を意味する。また、環状の基については「環の構成炭素員数が1ないし6の環状基」を意味する。鎖状の基と環状の基を含む基については「総炭素原子数が1ないし6の基」を意味する。
Below, each group in the said formula (I) of the said aspect [1] is demonstrated concretely.
In the description of the compounds of the present invention, for example, “C 1-6 ” means that the number of constituent carbon atoms is 1 to 6, and unless otherwise specified, carbon atoms of a linear, branched or cyclic group Represents a number. The chain group means “straight or branched chain having 1 to 6 carbon atoms”. Further, the cyclic group means “a cyclic group having 1 to 6 carbon atoms in the ring”. The group containing a chain group and a cyclic group means “a group having 1 to 6 total carbon atoms”.
 本明細書中、特に断りのない限り、「ハロゲン原子」としては、例えば、フッ素原子、塩素原子、臭素原子、およびヨウ素原子等が挙げられる。
 本明細書中、特に断りのない限り、「ハロゲン化」とは、置換基として1~5個の上記「ハロゲン原子」を有することを意味する。また、「ハロゲン化」は「ハロゲノ」と言い換えられる。
In the present specification, unless otherwise specified, examples of the “halogen atom” include a fluorine atom, a chlorine atom, a bromine atom, and an iodine atom.
In the present specification, unless otherwise specified, “halogenated” means having 1 to 5 of the above “halogen atoms” as a substituent. “Halogenation” is also referred to as “halogeno”.
 本明細書中、特に断りのない限り、「C1~6アルキル基」としては、例えば、メチル、エチル、プロピル、イソプロピル、ブチル、イソブチル、sec-ブチル、tert-ブチル、ペンチル、イソペンチル、ネオペンチル、ヘキシル、イソヘキシル等が挙げられる。 In the present specification, unless otherwise specified, examples of the “C 1-6 alkyl group” include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, Hexyl, isohexyl and the like can be mentioned.
 本明細書中、特に断りのない限り、「ハロゲン化C1~6アルキル基」とは、上記「C1~6アルキル基」が1~5個のハロゲン原子で任意に置換されている基を意味し、例えば、フルオロメチル、ジフルオロメチル、トリフルオロメチル、2,2,2-トリフルオロエチル、1,1,2,2-テトラフルオロエチル、ペンタフルオロエチル等が挙げられる。 In the present specification, unless otherwise specified, the “halogenated C 1-6 alkyl group” means a group in which the “C 1-6 alkyl group” is optionally substituted with 1 to 5 halogen atoms. Meaning, for example, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, 1,1,2,2-tetrafluoroethyl, pentafluoroethyl and the like.
 本明細書中、特に断りのない限り、「ヒドロキシハロゲン化C1~6アルキル基」とは、上記「ハロゲン化C1~6アルキル基」が1~5個の水酸基で任意に置換されている基を意味し、例えば、2,2,2-トリフルオロ-1-エタノール基等が挙げられる。 In the present specification, unless otherwise specified, the term “hydroxyhalogenated C 1-6 alkyl group” means that the above “halogenated C 1-6 alkyl group” is optionally substituted with 1 to 5 hydroxyl groups. Means a group such as 2,2,2-trifluoro-1-ethanol group.
 本明細書中、特に断りのない限り、「C3~8シクロアルキル基」としては、例えば、シクロプロピル、シクロブチル、シクロペンチル、シクロヘキシル、シクロヘプチルおよびシクロオクチル等が挙げられる。 Herein, unless otherwise specified, as the "C 3 ~ 8 cycloalkyl group", for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
 本明細書中、特に断りのない限り、「アリール基」は、「単環式アリール基」、「縮環式アリール基(2環式、または3環式が含まれる)」または「部分的に水素化された縮環式アリール基」であってよい。
 本明細書中、特に断りのない限り、「部分的に水素化された縮環式アリール基」とは、上記「縮環式アリール基」において、部分的水素化された縮合環から任意の水素原子を除いてできる1価の基を意味し、縮合環の芳香環部分の水素原子あるいは水素化された部分の水素原子のどちらが除かれてもよい。
 本明細書中、特に断りのない限り、「C6~14アリール基」としては、例えば、フェニル、1-ナフチル、2-ナフチル、2-,3-,4-ビフェニルアンスリル、フェナンスリル、アセナフチル、インダニル、インデニル、および1,2,3,4-テトラヒドロナフチル等が挙げられる。
In the present specification, unless otherwise specified, the “aryl group” means “monocyclic aryl group”, “condensed aryl group (including bicyclic or tricyclic)” or “partially It may be a hydrogenated fused-ring aryl group.
In the present specification, unless otherwise specified, the “partially hydrogenated condensed aryl group” means any hydrogen from a partially hydrogenated condensed ring in the above “condensed aryl group”. It means a monovalent group formed by removing an atom, and either a hydrogen atom in an aromatic ring part of a condensed ring or a hydrogen atom in a hydrogenated part may be removed.
Herein, unless otherwise specified, as the "C 6 ~ 14 aryl group", for example, phenyl, 1-naphthyl, 2-naphthyl, 2-, 3-, 4-biphenyl anthryl, phenanthryl, acenaphthyl, Indanyl, indenyl, 1,2,3,4-tetrahydronaphthyl and the like can be mentioned.
 本明細書中、特に断りのない限り、「複素環基」とは、窒素原子、硫黄原子および酸素原子から選ばれるヘテロ原子を1~5個含有する3~14員環の単環式もしくは縮環式の環から任意の水素原子を除いてできる1価の基を意味する。
 本明細書中、特に断りのない限り、「複素環基」としては、例えば、「ヘテロアリール基」および「非芳香族複素環基」等が挙げられる。
In the present specification, unless otherwise specified, the “heterocyclic group” means a 3 to 14-membered monocyclic or condensed ring containing 1 to 5 heteroatoms selected from a nitrogen atom, a sulfur atom and an oxygen atom. A monovalent group formed by removing any hydrogen atom from a cyclic ring.
In the present specification, unless otherwise specified, examples of the “heterocyclic group” include “heteroaryl group” and “non-aromatic heterocyclic group”.
 本明細書中、特に断りのない限り、上記「ヘテロアリール基」とは、窒素原子、硫黄原子、および酸素原子から選ばれるヘテロ原子を1~5個含有する5~14員ヘテロアリール環基を意味する。
 本明細書中、特に断りのない限り、上記「ヘテロアリール基」としては、例えば、「単環式ヘテロアリール基」、「縮環式ヘテロアリール基」、「部分的に水素化された縮環式ヘテロアリール基」が挙げられる。
In the present specification, unless otherwise specified, the above-mentioned “heteroaryl group” means a 5- to 14-membered heteroaryl ring group containing 1 to 5 heteroatoms selected from a nitrogen atom, a sulfur atom, and an oxygen atom. means.
In the present specification, unless otherwise specified, examples of the “heteroaryl group” include “monocyclic heteroaryl group”, “condensed heteroaryl group”, and “partially hydrogenated condensed ring”. Formula heteroaryl group ".
 本明細書中、特に断りのない限り、上記「単環式ヘテロアリール基」としては、環員数5~7のものが好ましく(5~7員ヘテロアリール基)、例えば、ピロリル、フリル、チエニル、イミダゾリル、ピラゾリル、オキサゾリル、イソキサゾリル、チアゾリル、イソチアゾリル、1,2,3-トリアゾリル、1,2,4-トリアゾリル、1,2,3-オキサジアゾリル、1,2,4-オキサジアゾリル、1,3,4-オキサジアゾリル、フラザニル、1,2,3-チアジアゾリル、1,2,4-チアジアゾリル、1,3,4-チアジアゾリル、テトラゾリル、ピリジル、ピリダジニル、ピリミジニル、ピラジニル、1,2,3-トリアジニル、1,2,4-トリアジニル、1,3,5-トリアジニル、2H-1,2,3-チアジアジニル、4H-1,2,4-チアジアジニル、6H-1,3,4-チアジアジニル、1,4-ジアゼピニル、1,4-オキサゼピニル等が挙げられる。 In the present specification, unless otherwise specified, the above “monocyclic heteroaryl group” is preferably one having 5 to 7 ring members (5 to 7 membered heteroaryl group), such as pyrrolyl, furyl, thienyl, Imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,3,4 Oxadiazolyl, furazanyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, 1,2,3-triazinyl, 1,2, 4-triazinyl, 1,3,5-triazinyl, 2H-1,2,3-thiadiazinyl, H-1,2,4-thiadiazinyl, 6H-1,3,4-thiadiazinyl, 1,4 diazepinyl, 1,4-oxazepinyl and the like.
 本明細書中、特に断りのない限り、上記「縮環式ヘテロアリール基」とは、「複素環基」と「アリール基」、もしくは、「複素環基」と「単環式ヘテロアリール基」が縮合して形成された縮合環から、任意の水素原子を除いてできる1価の基を意味し、当該任意の水素原子は縮合したいずれの環から除かれてもよい。 In the present specification, unless otherwise specified, the above-mentioned “condensed heteroaryl group” means “heterocyclic group” and “aryl group”, or “heterocyclic group” and “monocyclic heteroaryl group”. Means a monovalent group formed by removing an arbitrary hydrogen atom from a condensed ring formed by condensation, and the arbitrary hydrogen atom may be removed from any condensed ring.
 本明細書中、特に断りのない限り、上記「縮環式ヘテロアリール基」としては、環員数8~12のものが好ましく、例えば、インドリル、イソインドリル、ベンゾフラニル、イソベンゾフラニル、ベンゾチエニル、イソベンゾチエニル、ベンゾオキサゾリル、1,2-ベンゾイソキサゾリル、ベンゾチアゾリル、1,2-ベンゾイソチアゾリル、1H-ベンズイミダゾリル、1H-インダゾリル、1H-ベンゾトリアゾリル、クロメニル、イソクロメニル、キノリル、イソキノリル、シンノリニル、キナゾリニル、キノキサリニル、フタラジニル、ベンゾオキサゼピニル、ベンゾアゼピニル、ベンゾジアゼピニル、ナフチリジニル、プリニル、プテリジニル、カルバゾリル、カルボリニル、アクリジニル、フェノキサジニル、フェノチアジニル、フェナジニル、フェノキサチイニル、チアンスレニル、チアントレニル、フェナンスリジニル、フェナンスロリニル、インドリジニル、フロ[2,3-c]ピリジル、フロ[3,2-c]ピリジル、イミダゾ[1,5-a]ピラジニル、1H-ピラゾロ[3,4-c]ピリジル、1H-ピラゾロ[4,3-c]ピリジル、1H-ピラゾロ[4,3-b]ピリジル等が挙げられる。 In the present specification, unless otherwise specified, the above-mentioned “condensed heteroaryl group” is preferably one having 8 to 12 ring members, for example, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzothienyl, iso Benzothienyl, benzoxazolyl, 1,2-benzisoxazolyl, benzothiazolyl, 1,2-benzisothiazolyl, 1H-benzimidazolyl, 1H-indazolyl, 1H-benzotriazolyl, chromenyl, isochromenyl, quinolyl , Isoquinolyl, cinnolinyl, quinazolinyl, quinoxalinyl, phthalazinyl, benzoxazepinyl, benzoazepinyl, benzodiazepinyl, naphthyridinyl, purinyl, pteridinyl, carbazolyl, carbolinyl, acridinyl, phenoxazinyl, phenothiadi Phenazinyl, phenoxathiinyl, thianthrenyl, thianthenyl, phenanthridinyl, phenanthrolinyl, indolizinyl, furo [2,3-c] pyridyl, furo [3,2-c] pyridyl, imidazo [1,5 -A] pyrazinyl, 1H-pyrazolo [3,4-c] pyridyl, 1H-pyrazolo [4,3-c] pyridyl, 1H-pyrazolo [4,3-b] pyridyl and the like.
 本明細書中、特に断りのない限り、「部分的に水素化された縮環式ヘテロアリール基」とは、「複素環基」と「アリール基」、もしくは、「複素環基」と「ヘテロアリール基」が縮合して形成された縮合環において、部分的に水素化された縮合環から、任意の水素原子を除いてできる1価の基を意味する。当該任意の水素原子は、縮合環内における「複素環基」、「アリール基」および「ヘテロアリール基」の何れの環部の水素原子、あるいは水素化された環部の水素原子のどちらが除かれても良く、例えば、キノリンが部分的に水素化されたテトラヒドロキノリルであれば、5,6,7,8-テトラヒドロキノリルあるいは1,2,3,4-テトラヒドロキノリル等が挙げられる。これらの基は、任意の水素原子を除く位置により、例えば、5,6,7,8-テトラヒドロキノリルであれば、-2-イル、-3-イル、-4-イル、-5-イル、-6-イル、-7-イル、-8-イルなどが例示され、1,2,3,4-テトラヒドロキノリルであれば、例えば、-1-イル、-2-イル、-3-イル、-4-イル、-5-イル、-6-イル、-7-イル、-8-イルなどが例示される。 In this specification, unless otherwise specified, the “partially hydrogenated condensed heteroaryl group” means “heterocyclic group” and “aryl group”, or “heterocyclic group” and “heterocyclic group”. In a condensed ring formed by condensing an “aryl group”, it means a monovalent group formed by removing any hydrogen atom from a partially hydrogenated condensed ring. The arbitrary hydrogen atom is a hydrogen atom in any of the "heterocyclic group", "aryl group" and "heteroaryl group" in the condensed ring, or a hydrogen atom in the hydrogenated ring part. For example, if the quinoline is partially hydrogenated tetrahydroquinolyl, 5,6,7,8-tetrahydroquinolyl, 1,2,3,4-tetrahydroquinolyl and the like can be mentioned. Depending on the position of any hydrogen atom, these groups can be, for example, 5,6,7,8-tetrahydroquinolyl-2-yl, -3-yl, -4-yl, -5-yl. , -6-yl, -7-yl, -8-yl and the like, and 1,2,3,4-tetrahydroquinolyl, for example, -1-yl, -2-yl, -3- Il, -4-yl, -5-yl, -6-yl, -7-yl, -8-yl and the like are exemplified.
 本明細書中、特に断りのない限り、「部分的に水素化された縮環式ヘテロアリール基」としては、環員数8~12のものが好ましく、例えば、インドリニル、4,5,6,7-テトラヒドロ-1H-インドニル、2,3-ジヒドロベンゾフラニル、4,5,6,7-テトラヒドロ-ベンゾフラニル、2,3-ジヒドロベンゾ[d]オキサゾリル、2,3-ジヒドロベンゾ[d]チアゾリル、4,5,6,7-テトラヒドロ-1H-インダゾリル、クロマニル、2H-クロメニル、4H-クロメニル、イソクロマニル、1H-イソクロメニル、1,3-ベンゾジオキソリル、2,3-ジヒドロフロ[3,2-c]ピリジル等が挙げられる。 In the present specification, unless otherwise specified, the “partially hydrogenated condensed heteroaryl group” preferably has 8 to 12 ring members, such as indolinyl, 4, 5, 6, 7 -Tetrahydro-1H-indonyl, 2,3-dihydrobenzofuranyl, 4,5,6,7-tetrahydro-benzofuranyl, 2,3-dihydrobenzo [d] oxazolyl, 2,3-dihydrobenzo [d] thiazolyl, 4,5,6,7-tetrahydro-1H-indazolyl, chromanyl, 2H-chromenyl, 4H-chromenyl, isochromanyl, 1H-isochromenyl, 1,3-benzodioxolyl, 2,3-dihydrofuro [3,2-c ] Pyridyl etc. are mentioned.
 本明細書中、特に断りのない限り、「単環式非芳香族複素環基(3~8員非芳香族複素環基)」とは、酸素原子、硫黄原子および窒素原子から選ばれるヘテロ原子を1~4個含有する3~8員の単環である飽和もしくは不飽和の複素環から、任意の水素原子を除いてできる1価の基を意味する。 In this specification, unless otherwise specified, the “monocyclic non-aromatic heterocyclic group (3- to 8-membered non-aromatic heterocyclic group)” is a hetero atom selected from an oxygen atom, a sulfur atom and a nitrogen atom. Represents a monovalent group formed by removing any hydrogen atom from a saturated or unsaturated heterocyclic ring, which is a 3- to 8-membered monocycle containing 1 to 4.
 本明細書中、特に断りのない限り、「単環式非芳香族複素環基(3~8員非芳香族複素環基)」としては、例えば、アジリジニル、アゼチジニル、オキシラニル、チイラニル、オキセタニル、チエタニル、ピロリジニル、テトラヒドロフリル、ジヒドロフリル、チオラニル、ピラゾリニル、ピラゾリジニル、イミダゾリジニル、ピペリジニル、ジヒドロピラニル、テトラヒドロピラニル(オキサニル)、テトラヒドロチオピラニル、ピペラジニル、ジオキサニル、オキサゾリジニル、イソキサゾリニル、1,3-オキサゾリジニル、イソキサゾリジニル、チアゾリニル、イソチアゾリニル、1,3-チアゾリジニル、イソチアゾリジニル、オキサジアゾリニル、1,3,4-オキサジアゾリジニル、モルホリニル、チオモルホリニル、キヌクリジニル、アゼパニル、ジアゼピニル、オキセパニル等が挙げられる。 In the present specification, unless otherwise specified, examples of the “monocyclic non-aromatic heterocyclic group (3- to 8-membered non-aromatic heterocyclic group)” include aziridinyl, azetidinyl, oxiranyl, thiranyl, oxetanyl, thietanyl, and the like. , Pyrrolidinyl, tetrahydrofuryl, dihydrofuryl, thiolanyl, pyrazolinyl, pyrazolidinyl, imidazolidinyl, piperidinyl, dihydropyranyl, tetrahydropyranyl (oxanyl), tetrahydrothiopyranyl, piperazinyl, dioxanyl, oxazolidinyl, isoxazolinyl, 1,3-oxazolidinyl, iso Xazolidinyl, thiazolinyl, isothiazolinyl, 1,3-thiazolidinyl, isothiazolidinyl, oxadiazolinyl, 1,3,4-oxadiazolidinyl, morpholinyl, thiomorpholinyl, quinu Lysinyl, azepanyl, diazepinyl, oxepanyl, and the like.
 本明細書中、特に断りのない限り、「-NRAB基」とは、「アミノ基」の窒素原子上の二つの水素原子がRAおよびRBに置換された基を意味する。すなわち、「-NRAB基」とは、「アミノ基」の窒素原子上の二つの水素原子が、各々独立に、「水素原子」、「C1~6アルキル基」、「ハロゲン化C1~6アルキル基」、「C2~6アルケニル基」、「C2~6アルキニル基」、「-C(O)-C1~6アルキル基」、-C(O)O-C1~6アルキル基」、-SO2-C1~6アルキル基」、「-C(O)-ハロゲン化C1~6アルキル基」、-SO2-ハロゲン化C1~6アルキル基」等から任意に選ばれる基に置換されている基を意味する In the present specification, unless otherwise specified, the “—NR A R B group” means a group in which two hydrogen atoms on the nitrogen atom of the “amino group” are substituted with R A and R B. That is, “—NR A R B group” means that two hydrogen atoms on the nitrogen atom of “amino group” are each independently “hydrogen atom”, “C 1-6 alkyl group”, “halogenated C”. 1-6 alkyl group "," C 2 ~ 6 alkenyl group "," C 2 ~ 6 alkynyl group "," - C (O) -C 1 ~ 6 alkyl group ", -C (O) O-C 1 ~ 6 alkyl group ", -SO 2 -C 1 ~ 6 alkyl group", "- C (O) - halogenated C 1 ~ 6 alkyl group" -SO 2 - optionally halogenated C 1 ~ 6 alkyl group "such as Means a group substituted by a group selected by
[1-1]
 上記態様[1]の式(I)において、好ましくは、m=0~3である。
[1-1-1]
 上記態様[1]の式(I)において、より好ましくは、m=2である。
[1-1]
In the formula (I) of the above embodiment [1], preferably m = 0 to 3.
[1-1-1]
In the formula (I) of the above embodiment [1], m = 2 is more preferable.
[1-2]
 上記態様[1]の式(I)において、好ましくは、n=0~2である。
[1-2-1]
 上記態様[1]の式(I)において、より好ましくは、n=0、または1である。
[1-2]
In the formula (I) of the above embodiment [1], preferably n = 0 to 2.
[1-2-1]
In the formula (I) of the above embodiment [1], n = 0 or 1 is more preferable.
[1-3]
 上記態様[1]の式(I)において、環Aとして好ましくは、フェニル基、ピリジニル基、ピリミジニル基、ピラジニル基、チアゾリル基、またはオキサゾリル基から選ばれる基である。
[1-3-1]
 上記態様[1]の式(I)において、環Aとしてより好ましくは、環Aは、フェニル基、ピリジン-2-イル基、ピリミジン-2-イル基、ピラジン-2-イル基、チアゾール-2-イル基、またはオキサゾール-2-イル基から選ばれる基である。
[1-3-2]
 上記態様[1]の式(I)において、環Aとして特に好ましくは、ピリジン-2-イル基、またはピリミジン-2-イル基である。
[1-3]
In the formula (I) of the above embodiment [1], the ring A is preferably a group selected from a phenyl group, a pyridinyl group, a pyrimidinyl group, a pyrazinyl group, a thiazolyl group, or an oxazolyl group.
[1-3-1]
In the formula (I) of the above embodiment [1], the ring A is more preferably ring A is a phenyl group, a pyridin-2-yl group, a pyrimidin-2-yl group, a pyrazin-2-yl group, a thiazole-2 -A group selected from an yl group and an oxazol-2-yl group.
[1-3-2]
In the formula (I) of the above embodiment [1], the ring A is particularly preferably a pyridin-2-yl group or a pyrimidin-2-yl group.
[1-4]
 上記態様[1]の式(I)において、環Bとして好ましくは、フェニル基、ピリジニル基、ピリダジニル基、フラニル基、1H-ピラゾリル基、またはチアゾリル基から選ばれる基である。
[1-4-1]
 上記態様[1]の式(I)において、環Bとしてより好ましくは、式(B-1)、式(B-2)、式(B-3)、式(B-4)または式(B-5)[式(B-1)中、X4、X5、X6およびX7は、各々独立に、NまたはC-Hを表す]から選ばれる基である(但し、式(B-1)~(B-5)中、-C(O)-基及びQ基は環Bに含まない)。
Figure JPOXMLDOC01-appb-C000006
[式(B-1)中、X4は、NまたはC-R2aを表し、ここで当該R2aは、水素原子、ハロゲン原子、または-O-C1~6アルキル基を表し;X5は、C-R2bを表し、ここで当該R2bは、水素原子、ハロゲン原子、C1~6アルキル基、または5~7員ヘテロアリール基(当該5~7員ヘテロアリール基は、ハロゲン原子、またはC1~6アルキル基で1~4個置換されていても良く)を表し;X6は、NまたはC-R2cを表し、ここで当該R2Cは、水素原子、ハロゲン原子、C1~6アルキル基、または-O-C1~6アルキル基を表し;X7は、NまたはC-R2dを表し、ここで当該R2dは、水素原子、またはハロゲン原子を表す。]
[1-4]
In the formula (I) of the above embodiment [1], the ring B is preferably a group selected from a phenyl group, a pyridinyl group, a pyridazinyl group, a furanyl group, a 1H-pyrazolyl group, and a thiazolyl group.
[1-4-1]
In the formula (I) of the above embodiment [1], the ring B is more preferably the formula (B-1), the formula (B-2), the formula (B-3), the formula (B-4) or the formula (B -5) [in the formula (B-1), X 4 , X 5 , X 6 and X 7 each independently represents N or C—H] (provided that the formula (B— 1) to (B-5), the —C (O) — group and the Q group are not included in the ring B).
Figure JPOXMLDOC01-appb-C000006
Wherein (B-1), X 4 represents N or C-R 2a, where the R 2a represents a hydrogen atom, a halogen atom or -O-C 1 ~ 6 alkyl group,; X 5 Represents C—R 2b , where R 2b is a hydrogen atom, a halogen atom, a C 1-6 alkyl group, or a 5- to 7-membered heteroaryl group (the 5- to 7-membered heteroaryl group is a halogen atom X 1 represents N or C—R 2c , where R 2C represents a hydrogen atom, a halogen atom, a C atom, or a C 1-6 alkyl group (which may be substituted with 1 to 4 alkyl groups). 1-6 alkyl group or -O-C 1 ~ represents an alkyl group,; X 7 represents N or C-R 2d, wherein said R 2d represents a hydrogen atom or a halogen atom. ]
[1-4-2]
 上記態様[1]の式(I)において、環Bとして特に好ましくは、式(B-1-1)、式(B-1-2)、または式(B-3)から選ばれる基である(但し、式(B-1-1)、式(B-1-2)及び式(B-3)中、-C(O)-基及びQ基は環Bに含まない)。
Figure JPOXMLDOC01-appb-C000007
[1-4-2]
In formula (I) of the above embodiment [1], ring B is particularly preferably a group selected from formula (B-1-1), formula (B-1-2), or formula (B-3). (However, in formula (B-1-1), formula (B-1-2) and formula (B-3), the —C (O) — group and Q group are not included in ring B).
Figure JPOXMLDOC01-appb-C000007
[1-5]
 上記態様[1]の式(I)において、Qとして好ましくは、C3~8シクロアルキル基、-ORc1(当該Rc1は、C1~6アルキル基、ハロゲン化C1~6アルキル基、またはC6~14アリール基を表す)、または部分構造式(SF-1)で表される基:
Figure JPOXMLDOC01-appb-C000008
[式(SF-1)中、環Cは、フェニル基、ピリジル基、ピリミジニル基、ピリダジニル基、ピラゾリル基、トリアゾリル基、オキサジアゾリル基、チアゾリル基、オキサゾリル基、イソチアゾリル基、テトラゾリル基、ピロリジニル基、ピペラジニル基、またはモルホニル基から選ばれる基を表し;pは、0~2の整数であり;R3は、各々独立に、ハロゲン原子、シアノ基、-Rd1、または-ORd1(当該Rd1は、C1~6アルキル基、またはハロゲン化C1~6アルキル基を表す)を表す]である。
[1-5]
In the formula (I) of the above embodiment [1], preferably the Q, C 3 ~ 8 cycloalkyl group, -OR c1 (the R c1 is, C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl group, or C 6 ~ 14 aryl group), or a partial structural formula (SF-1) a group represented by:
Figure JPOXMLDOC01-appb-C000008
[In Formula (SF-1), Ring C is a phenyl group, pyridyl group, pyrimidinyl group, pyridazinyl group, pyrazolyl group, triazolyl group, oxadiazolyl group, thiazolyl group, oxazolyl group, isothiazolyl group, tetrazolyl group, pyrrolidinyl group, piperazinyl group A group or a group selected from morpholyl groups; p is an integer of 0 to 2; R 3 is independently a halogen atom, a cyano group, —R d1 , or —OR d1 (wherein R d1 is Represents a C 1-6 alkyl group or a halogenated C 1-6 alkyl group].
[1-5-1]
 上記態様[1]の式(I)において、Qとしてより好ましくは、C3~8シクロアルキル基、-ORc1(当該Rc1は、C1~6アルキル基、ハロゲン化C1~6アルキル基、またはC6~14アリール基を表す)、または部分構造式(SF-1)で表される基:
Figure JPOXMLDOC01-appb-C000009
[式(SF-1)中、環Cは、フェニル基、ピリジン-2-イル基、ピリミジン-2-イル基、ピリダジン-3-イル基、1H-ピラゾール-1-イル基、1-メチル-1H-ピラゾール-3-イル基、1H-ピラゾール-3-イル基、1H-ピラゾール-4-イル基、1H-1,2,3-トリアゾール-1-イル基、2H-1,2,3-トリアゾール-2-イル基、1,2,4-オキサジアゾール-5-イル基、チアゾール-2-イル基、チアゾール-4-イル基、チアゾール-5-イル基、オキサゾール-2-イル基、イソチアゾール-4-イル基、イソチアゾール-5-イル基、2H-テトラゾール-2-イル基、モルホリン-1-イル基、ピロリジン-1-イル基、ピロリジン-1-イル基、ピペラジン-1-イル基、またはモルホリン-1-イル基から選ばれる基を表し;pは、0または1の整数であり;R3は、各々独立に、ハロゲン原子、シアノ基、-C1~6アルキル基、または-O-C1~6アルキル基を表す]である。
[1-5-1]
In the formula (I) of the above embodiment [1], is more preferably Q, C 3 ~ 8 cycloalkyl group, -OR c1 (the R c1 is, C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl group , or C 6 ~ 14 aryl group), or a partial structural formula (SF-1) a group represented by:
Figure JPOXMLDOC01-appb-C000009
[In the formula (SF-1), ring C represents a phenyl group, a pyridin-2-yl group, a pyrimidin-2-yl group, a pyridazin-3-yl group, a 1H-pyrazol-1-yl group, 1-methyl- 1H-pyrazol-3-yl group, 1H-pyrazol-3-yl group, 1H-pyrazol-4-yl group, 1H-1,2,3-triazol-1-yl group, 2H-1,2,3- Triazol-2-yl group, 1,2,4-oxadiazol-5-yl group, thiazol-2-yl group, thiazol-4-yl group, thiazol-5-yl group, oxazol-2-yl group, Isothiazol-4-yl group, isothiazol-5-yl group, 2H-tetrazol-2-yl group, morpholin-1-yl group, pyrrolidin-1-yl group, pyrrolidin-1-yl group, piperazin-1- Il group or morpho It represents a group selected from the down-1-yl group; p is an integer of 0 or 1; R 3 is, each independently, a halogen atom, a cyano group, -C 1 ~ 6 alkyl group or -O-, Represents a C 1-6 alkyl group].
[1-5-2]
 上記態様[1]の式(I)において、Qとして更に好ましくは、-O-ハロゲン化C1~6アルキル基(より具体的には、1,1,2,2-テトラフルオロエトキシ基、および2,2,2-トリフルオロエトキシであり)、および部分構造式(SF-1)で表される基:
Figure JPOXMLDOC01-appb-C000010
[式(SF-1)中、環Cは、ピリジン-2-イル基、1H-ピラゾール-1-イル基、2H-1,2,3-トリアゾール-2-イル基、または1,2,4-オキサジアゾール-5-イル基から選ばれる基を表し;pは、0または1の整数であり;R3は、C1~6アルキル基(より具体的には、R3は、メチル基である)を表す]である。
[1-5-2]
In the formula (I) of the above embodiment [1], more preferably, Q is —O-halogenated C 1-6 alkyl group (more specifically, 1,1,2,2-tetrafluoroethoxy group, and 2,2,2-trifluoroethoxy), and a group represented by the partial structural formula (SF-1):
Figure JPOXMLDOC01-appb-C000010
[In the formula (SF-1), ring C represents a pyridin-2-yl group, a 1H-pyrazol-1-yl group, a 2H-1,2,3-triazol-2-yl group, or 1,2,4. Represents a group selected from -oxadiazol-5-yl group; p is an integer of 0 or 1; R 3 is a C 1-6 alkyl group (more specifically, R 3 is a methyl group) Represents).
[1-6]
 上記態様[1]の式(I)において、R1として好ましくは、ハロゲン原子、シアノ基、-Ra1、-ORa1、-C(O)Ra1、-C(O)ORa1、-SRa1、-NRAB基、3~8員非芳香族複素環基、またはオキソ基(該-NRAB基におけるRAおよびRBは、各々独立に、水素原子、またはC1~6アルキル基を表し;当該Ra1は、C1~6アルキル基、ハロゲン化C1~6アルキル基、ヒドロキシハロゲン化C1~6アルキル基、またはC3~8シクロアルキル基を表す)である。
[1-6-1]
 上記態様[1]の式(I)において、R1としてより好ましくは、シアノ基、C1~6アルキル基、C3~8シクロアルキル基、または-O-C1~6アルキル基を表し、より具体的には、R1は、シアノ基、メチル基、シクロプロピル基、およびメトキシ基である。
[1-6]
In the formula (I) of the above embodiment [1], R 1 is preferably a halogen atom, a cyano group, —R a1 , —OR a1 , —C (O) R a1 , —C (O) OR a1 , —SR a1 , —NR A R B group, 3- to 8-membered non-aromatic heterocyclic group, or oxo group (R A and R B in the —NR A R B group are each independently a hydrogen atom, or C 1- It represents an alkyl group; the R a1 is a C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl group, hydroxy halogenated C 1 ~ 6 alkyl group or an C 3 ~ 8 cycloalkyl group) .
[1-6-1]
In the formula (I) of the above embodiment [1], and more preferably as R 1, a cyano group, C 1 ~ 6 alkyl group, C 3 ~ 8 cycloalkyl group, or -O-C 1 ~ 6 alkyl group, More specifically, R 1 is a cyano group, a methyl group, a cyclopropyl group, and a methoxy group.
[1-7]
 上記態様[1]の式(I)において、R2として好ましくは、ハロゲン原子、シアノ基、-Rb1、-ORb1、(当該Rb1は、C1~6アルキル基、またはハロゲン化C1~6アルキル基を表す)、ピラゾリル基、またはピリジル基(当該、ピラゾリル基およびピリジル基は、ハロゲン原子、またはC1~6アルキル基で1~4個置換されていても良い)である。
[1-7-1]
 上記態様[1]の式(I)において、R2としてより好ましくは、ハロゲン原子、またはC1~6アルキル基を表し、より具体的には、R2は、フッ素、およびメチル基である。
[1-7]
In the formula (I) of the above embodiment [1], R 2 is preferably a halogen atom, a cyano group, —R b1 , —OR b1 (where R b1 is a C 1-6 alkyl group, or a halogenated C 1 Represents a 6- alkyl group), a pyrazolyl group, or a pyridyl group (the pyrazolyl group and the pyridyl group may be substituted with 1 to 4 halogen atoms or a C 1-6 alkyl group).
[1-7-1]
In the formula (I) of the above embodiment [1], R 2 is more preferably a halogen atom or a C 1-6 alkyl group, and more specifically, R 2 is fluorine and a methyl group.
[1-8]
 上記態様[1]の式(I)において、環Aおよび(R1)mの組み合わせとして好ましくは、以下の部分構造式である式(A-1)または式(A-2): 
Figure JPOXMLDOC01-appb-C000011
[式(A-1)中、X1は、NまたはC-Hを表し;X2は、NまたはC-R1aを表し(当該R1aは、水素原子、ハロゲン原子、C1~6アルキル基、C3~8シクロアルキル基、-O-C1~6アルキル基、-S-C1~6アルキル基、-NRAB基(当該-NRAB基におけるRAおよびRBは、各々独立に、水素原子、およびC1~6アルキル基を表す)、または3~8員非芳香族複素環基を表す);X3は、NまたはC-R1bを表し(当該R1bは、水素原子、シアノ基、C1~6アルキル基、ハロゲン化C1~6アルキル基、C3~8シクロアルキル基、-O-C1~6アルキル基、-O-ハロゲン化C1~6アルキル基、ヒドロキシハロゲン化C1~6アルキル基、-C(O)-C1~6アルキル基、または-C(O)O-C1~6アルキル基を表す);R1cは、各々独立に、水素原子、またはC1~6アルキル基を表し;式(A-2)中、Y1はO、S、またはN-R1dを表し、当該R1dは、各々独立に、水素原子、またはC1~6アルキル基を表す]である。
[1-8]
In the formula (I) of the above embodiment [1], the combination of the ring A and (R 1 ) m is preferably the following partial structural formula (A-1) or (A-2):
Figure JPOXMLDOC01-appb-C000011
[In the formula (A-1), X 1 represents N or C—H; X 2 represents N or C—R 1a (wherein R 1a represents a hydrogen atom, a halogen atom, C 1-6 alkyl group, C 3 ~ 8 cycloalkyl group, -O-C 1 ~ 6 alkyl group, -S-C 1 ~ 6 alkyl group, -NR A R B group (said -NR A R in R B groups A and R B Each independently represents a hydrogen atom and a C 1-6 alkyl group), or a 3- to 8-membered non-aromatic heterocyclic group); X 3 represents N or C—R 1b (the R 1b is a hydrogen atom, a cyano group, C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl group, C 3 ~ 8 cycloalkyl group, -O-C 1 ~ 6 alkyl group, -O- halogenated C 1 1-6 alkyl group, a hydroxy halogenated C 1 ~ 6 alkyl group, -C (O) -C 1 ~ 6 alkyl group, or -C (O) O-C 1 ~ 6 alkyl group,); R 1c Each independently represent a hydrogen atom or a C 1 ~ 6 alkyl group; wherein (A-2), Y 1 represents O, S or N-R 1d,, the R 1d are each independently, Represents a hydrogen atom or a C 1-6 alkyl group].
[1-8-1]
 上記態様[1]の式(I)において、環Aおよび(R1)mの組み合わせとしてより好ましくは、上記態様[1-8]中の部分構造式である式(A-1)または式(A-2)[式(A-1)中、X1は、NまたはC-Hを表し;X2は、NまたはC-R1aを表し(当該R1aは、水素原子、フッ素原子、塩素原子、メチル基、メトキシ基、エトキシ基、(メトキシ-d3)基、シクロプロピル基、ピロリジン-1-イル基、メチルチオ基、またはジメチルアミノ基を表す);X3は、NまたはC-R1bを表し(当該R1bは、水素原子、シアノ基、メチル基、エチル基、シクロプロピル基、アセチル基、メトキシ基、ジフルオロメトキシ基、トリフルオロメチル基、1-ヒドロキシ-2,2,2-トリフルオロエチル基、またはメトキシカルボニル基を表す);R1cは、各々独立に、水素原子、またはメチル基を表し:式(A-2)中、Y1はO、S、またはN-R1dを表し、当該R1dは、各々独立に、水素原子、またはメチル基を表す]である。
[1-8-1]
In the formula (I) of the above embodiment [1], the combination of the ring A and (R 1 ) m is more preferably a formula (A-1) or a formula (A) which is a partial structural formula in the above embodiment [1-8]. A-2) [in the formula (A-1), X 1 represents N or C—H; X 2 represents N or C—R 1a (wherein R 1a represents a hydrogen atom, a fluorine atom, chlorine) An atom, a methyl group, a methoxy group, an ethoxy group, a (methoxy-d 3 ) group, a cyclopropyl group, a pyrrolidin-1-yl group, a methylthio group, or a dimethylamino group); X 3 is N or C—R 1b (wherein R 1b represents a hydrogen atom, a cyano group, a methyl group, an ethyl group, a cyclopropyl group, an acetyl group, a methoxy group, a difluoromethoxy group, a trifluoromethyl group, 1-hydroxy-2,2,2- Trifluoroethyl group or methoxycarbonyl group R 1c each independently represents a hydrogen atom or a methyl group: In Formula (A-2), Y 1 represents O, S, or N—R 1d , and each R 1d represents each independently And represents a hydrogen atom or a methyl group.
[1-8-2]
 上記態様[1]の式(I)において、環Aおよび(R1)mの組み合わせとして更に好ましくは、3,5-ジメチルフェニル基、3-クロロ-4-シアノ-ピリジン-2-イル基、3-メチル-4-(トリフルオロメチル)ピリジン-2-イル基、3-メトキシ-4-(トリフルオロメチル)ピリジン-2-イル基、4-アセチルピリジン-2-イル基、4-シアノ-ピリジン-2-イル基、4-シアノ-3-シクロプロピル-ピリジン-2-イル基、4-シアノ-3-メトキシ-ピリジン-2-イル基、4-シアノ-3-エトキシ-ピリジン-2-イル基、4-シアノ-3-(メトキシ-d3)-ピリジン-2-イル基、4-シアノ-6-メチル-ピリジン-2-イル基、4-シアノ-3-フルオロ-ピリジン-2-イル基、4-シアノ-3-シクロプロピル-ピリジン-2-イル基、4-シアノ-3-(ピロリジン-1-イル)-ピリジン-2-イル基、4-シアノ-3-(メチルチオ)-ピリジン-2-イル基、4-シアノ-3-(ジメチルアミノ)-ピリジン-2-イル基、4-(ジフルオロメトキシ)ピリジン-2-イル基、4-(1-ヒドロキシ-2,2,2-トリフルオロエチル)-ピリジン-2-イル基、4-(トリフルオロメチル)ピリジン-2-イル基、4-(メトキシカルボニル)-ピリジン-2-イル基、6-メチル-4-(トリフルオロメチル)ピリジン-2-イル基、4,6-ジメチルピリジン-2-イル基、1,5-ジメチルピラジン-2(1H)-オン-3-イル基、4,5-ジメチルピリミジン-2-イル基、4,6-ジメチルピリミジン-2-イル基、4-エチル-6-メチルピリミジン-2-イル基、4,5,6-トリメチルピリミジン-2-イル基、4-シクロプロピル-6-メチルピリミジン-2-イル基、4-メトキシ-6-メチルピリミジン-2-イル基、および3-メトキシ-6-メチルピラジン-2-イル基、または4,5-ジメチルチアゾール-2-イル基から選ばれる基である。
[1-8-2]
In the formula (I) of the above embodiment [1], the combination of the ring A and (R 1 ) m is more preferably a 3,5-dimethylphenyl group, a 3-chloro-4-cyano-pyridin-2-yl group, 3-methyl-4- (trifluoromethyl) pyridin-2-yl group, 3-methoxy-4- (trifluoromethyl) pyridin-2-yl group, 4-acetylpyridin-2-yl group, 4-cyano- Pyridin-2-yl group, 4-cyano-3-cyclopropyl-pyridin-2-yl group, 4-cyano-3-methoxy-pyridin-2-yl group, 4-cyano-3-ethoxy-pyridine-2- Yl group, 4-cyano-3- (methoxy-d3) -pyridin-2-yl group, 4-cyano-6-methyl-pyridin-2-yl group, 4-cyano-3-fluoro-pyridin-2-yl Group 4-cyano-3- Cyclopropyl-pyridin-2-yl group, 4-cyano-3- (pyrrolidin-1-yl) -pyridin-2-yl group, 4-cyano-3- (methylthio) -pyridin-2-yl group, 4- Cyano-3- (dimethylamino) -pyridin-2-yl group, 4- (difluoromethoxy) pyridin-2-yl group, 4- (1-hydroxy-2,2,2-trifluoroethyl) -pyridine-2 -Yl group, 4- (trifluoromethyl) pyridin-2-yl group, 4- (methoxycarbonyl) -pyridin-2-yl group, 6-methyl-4- (trifluoromethyl) pyridin-2-yl group, 4,6-dimethylpyridin-2-yl group, 1,5-dimethylpyrazin-2 (1H) -one-3-yl group, 4,5-dimethylpyrimidin-2-yl group, 4,6-dimethylpyrimidine- 2-I Group, 4-ethyl-6-methylpyrimidin-2-yl group, 4,5,6-trimethylpyrimidin-2-yl group, 4-cyclopropyl-6-methylpyrimidin-2-yl group, 4-methoxy-6 -A group selected from a methylpyrimidin-2-yl group, a 3-methoxy-6-methylpyrazin-2-yl group, and a 4,5-dimethylthiazol-2-yl group.
[1-8-3]
 上記態様[1]の式(I)において、環Aおよび(R1)mの組み合わせとして特に好ましくは、4-シアノ-3-メトキシ-ピリジン-2-イル基、4-シアノ-3-シクロプロピル-ピリジン-2-イル基、または4,6-ジメチルピリミジン-2-イル基である。
[1-8-3]
In the formula (I) of the above embodiment [1], the combination of the ring A and (R 1 ) m is particularly preferably a 4-cyano-3-methoxy-pyridin-2-yl group, 4-cyano-3-cyclopropyl -Pyridin-2-yl group or 4,6-dimethylpyrimidin-2-yl group.
[1-9]
 上記態様[1]の式(I)において、環B、Qおよび(R2)nの組み合わせとして好ましくは、部分構造式である式(BQ-1)、式(BQ-2)、式(BQ-3)、式(BQ-4)、または式(BQ-5)(但し、式(BQ-1)~(BQ-5)中、-C(O)-基は環B、Qおよび(R2)nの組み合わせに含まない):
Figure JPOXMLDOC01-appb-C000012
[式(BQ-1)中、X4は、NまたはC-R2aを表し(当該R2aは、水素原子、ハロゲン原子、または-O-C1~6アルキル基を表す);X5は、C-R2bを表し(当該R2bは、水素原子、ハロゲン原子、C1~6アルキル基、または5~7員ヘテロアリール基(当該5~7員ヘテロアリール基は、ハロゲン原子、またはC1~6アルキル基で1~4個置換されていても良い)を表す);X6は、NまたはC-R2cを表し(当該R2Cは、水素原子、ハロゲン原子、C1~6アルキル基、または-O-C1~6アルキル基を表す);X7は、NまたはC-R2dを表し(当該R2dは、水素原子、またはハロゲン原子を表す);Q’は、ハロゲン原子、C3~8シクロアルキル基、-ORc1(当該Rc1は、C1~6アルキル基、ハロゲン化C1~6アルキル基、およびC6~14アリール基から任意に選ばれる基を表す)、または3~8員非芳香族複素環を表し:式(BQ-2)、式(BQ-3)、式(BQ-4)および式(BQ-5)中、pおよびR3は、上記態様[1-5]中の定義と同じであり;X4、X5,X6、およびX7は 上記式(BQ-1)中の定義と同じであり;Xは、NまたはC-Hを表し;Y2はO、S、N-HまたはN-CH3を表す]である。
[1-9]
In the formula (I) of the above embodiment [1], the combination of the rings B, Q and (R 2 ) n is preferably a partial structural formula of the formula (BQ-1), formula (BQ-2), formula (BQ -3), formula (BQ-4), or formula (BQ-5) (wherein, in formulas (BQ-1) to (BQ-5), the —C (O) — group represents ring B, Q and (R 2 ) Not included in the combination of n ):
Figure JPOXMLDOC01-appb-C000012
Wherein (BQ-1), X 4 represents N or C-R 2a (the R 2a represents a hydrogen atom, a halogen atom or -O-C 1 ~ 6 alkyl group,); X 5 is C—R 2b (wherein R 2b is a hydrogen atom, a halogen atom, a C 1-6 alkyl group, or a 5- to 7-membered heteroaryl group (the 5- to 7-membered heteroaryl group is a halogen atom, or C represents a may be one to four substituents) at 1-6 alkyl group); X 6 represents N or C-R 2c (the R 2C is hydrogen atom, halogen atom, C 1-6 alkyl X 7 represents N or C—R 2d (wherein R 2d represents a hydrogen atom or a halogen atom); Q ′ represents a halogen atom, or —O—C 1-6 alkyl group; , C 3 ~ 8 cycloalkyl group, -OR c1 (the R c1 is, C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl group, Oyo Fine C 6 ~ 14 represents a group selected from aryl group optionally), or 3-8 membered represents a non-aromatic heterocyclic ring: wherein (BQ-2), formula (BQ-3), formula (BQ-4) And in formula (BQ-5), p and R 3 are the same as defined in the above embodiment [1-5]; X 4 , X 5 , X 6 and X 7 are the same as those in formula (BQ-1) X represents N or C—H; Y 2 represents O, S, N—H or N—CH 3 ].
[1-9-1]
 上記態様[1]の式(I)において、環B、Qおよび(R2)nの組み合わせとしてより好ましくは、上記態様[1-9]中の部分構造式である式(BQ-1)、式(BQ-2)、式(BQ-3)、式(BQ-4)、または式(BQ-5)[上記式(BQ-1)中、X4は、NまたはC-R2a(当該R2aは、水素原子、フッ素原子、塩素原子、メトキシ基、またはエトキシ基を表す)であり;X5は、C-R2b(当該R2bは、水素原子、フッ素原子、メチル基、1H-ピラゾール-1-イル基、または5-クロロ-ピリジン-3-イル基を表す)であり;X6は、NまたはC-R2c(当該R2Cは、水素原子、フッ素原子、塩素原子、メチル基、またはメトキシ基を表す)であり;X7は、NまたはC-R2d(当該R2dは、水素原子、またはフッ素原子を表す)であり;Q’は、エトキシ基、フェノキシ基、トリフルオロメトキシ、1,1,2,2-テトラフルオロエトキシ基、シクロプロピル基、ピロリジン-1-イル基、またはモルホリン-1-イル基を表し:上記式(BQ-2)、式(BQ-3)、式(BQ-4)および式(BQ-5)中、pは、0または1の整数であり;R3は、上記態様[1-5]中の定義と同じであり;X4、X5,X6、およびX7は 態様[1-9-1]中の上記式(BQ-1)中の定義と同じであり;Xは、NまたはC-Hを表し;Y2はO、S、N-HまたはN-CH3を表す]である。
[1-9-1]
In the formula (I) of the embodiment [1], the combination of the rings B, Q and (R 2 ) n is more preferably a formula (BQ-1) which is a partial structural formula in the embodiment [1-9], Formula (BQ-2), Formula (BQ-3), Formula (BQ-4), or Formula (BQ-5) [In the above formula (BQ-1), X 4 represents N or C—R 2a R 2a represents a hydrogen atom, a fluorine atom, a chlorine atom, a methoxy group, or an ethoxy group; X 5 represents C—R 2b (wherein R 2b represents a hydrogen atom, a fluorine atom, a methyl group, 1H— X 6 is N or C—R 2c (wherein R 2C is a hydrogen atom, a fluorine atom, a chlorine atom, a methyl atom), or a pyrazol-1-yl group or a 5-chloro-pyridin-3-yl group; X 7 represents N or C—R 2d (wherein R 2d represents a hydrogen atom or fluorine) Q ′ represents an ethoxy group, phenoxy group, trifluoromethoxy, 1,1,2,2-tetrafluoroethoxy group, cyclopropyl group, pyrrolidin-1-yl group, or morpholine-1- In the above formula (BQ-2), formula (BQ-3), formula (BQ-4) and formula (BQ-5), p is an integer of 0 or 1; R 3 is X 4 , X 5 , X 6 and X 7 are the same as defined in the above formula (BQ-1) in the embodiment [1-9-1]. X represents N or C—H; Y 2 represents O, S, N—H or N—CH 3 ].
[1-9-2]
 上記態様[1]の式(I)において、環B、Qおよび(R2)nの組み合わせとして更に好ましくは、5-フルオロ-3-(ピリミジン-2-イル)ピリジン-2-イル基、(2,2’-ビピリジン)-3-イル基、2-(1H-1,2,3-トリアゾール-1-イル)ピリジン-3-イル基、2-(1H-ピラゾール-1-イル)ピリジン-3-イル基、2-(1-メチル-1H-ピラゾール-3-イル)ピリジン-3-イル基、2-(2H-1,2,3-トリアゾール-2-イル)ピリジン-3-イル基、2-(3-シアノ-1H-ピラゾール-1-イル)ピリジン-3-イル基、2-(3-メチル-1H-ピラゾール-1-イル)ピリジン-3-イル基、2-(チアゾール-2-イル)ピリジン-3-イル基、2,5-(1H-ピラゾール-1-イル)ピリジン-3-イル基、4-エトキシ-2-(1H-ピラゾール-1-イル)ピリジン-3-イル基、4-メトキシ-2-(1H-ピラゾール-1-イル)ピリジン-3-イル基、5’’-クロロ-[2,2’:5’,3’’-terピリジン]-3’-イル基、5-フルオロ-2-(1H-ピラゾール-1-イル)ピリジン-3-イル基、6-メトキシ-2-(1H-ピラゾール-1-イル)ピリジン-3-イル基、2-(2H-1,2,3-トリアゾール-2-イル)ピリジン-4-イル基、3-(1H-ピラゾール-1-イル)ピリダジン-4-イル基、[1,1’-ビフェニル]基、2-(1H-ピラゾール-1-イル)フェニル基、2-(2H-1,2,3-トリアゾール-2-イル)フェニル基、2-(2H-テトラゾール-2-イル)フェニル基、2-(3-メチル-1,2,4-オキサジアゾール-5-イル)フェニル基、2-(3-メチルイソチアゾール-5-イル)フェニル基、2-(4-シアノチアゾール-2-イル)フェニル基、2-(6-シアノピリジン-2-イル)フェニル基、2-(6-メチルピリジン-2-イル)フェニル基、2-(6-メトキシピリジン-2-イル)フェニル基、2-(イソチアゾール-4-イル)フェニル基、2-(オキサゾール-2-イル)フェニル基、2-(チアゾール-2-イル)フェニル基、2-(チアゾール-4-イル)フェニル基、2-(チアゾール-5-イル)フェニル基、2-(ピリジン-2-イル)フェニル基、2-(ピリミジン-2-イル)フェニル基、2-クロロ-6-(2H-1,2,3-トリアゾール-2-イル)フェニル基、2-フルオロ-6-(ピリミジン-2-イル)フェニル基、2-メトキシ-6-(2H-1,2,3-トリアゾール-2-イル)フェニル基、3’-(ジフルオロメトキシ)-[1,1’-ビフェニル]-2-イル基、3’-メトキシ-[1,1’-ビフェニル]-2-イル基、3-フルオロ-2-(2H-1,2,3-トリアゾール-2-イル)フェニル基、4’-フルオロ-[1,1’-ビフェニル]-2-イル基、4-クロロ-2-(2H-1,2,3-トリアゾール-2-イル)フェニル基、4-クロロ-2-(ピリミジン-2-イル)フェニル基、4-フルオロ-2-(2H-1,2,3-トリアゾール-2-イル)フェニル基、4-フルオロ-2-(ピリミジン-2-イル)フェニル基、4-メチル-2-(2H-1,2,3-トリアゾール-2-イル)フェニル基、4-メチル-2-(ピリミジン-2-イル)フェニル基、4-メトキシ-2-(1H-1,2,3-トリアゾール-1-イル)フェニル基、4-メトキシ-2-(2H-1,2,3-トリアゾール-2-イル)フェニル基、5-フルオロ-2-(2H-1,2,3-トリアゾール-2-イル)フェニル基、5-メチル-2-(2H-1,2,3-トリアゾール-2-イル)フェニル基、2-シクロプロピルフェニル基、2,6-ジエトキシフェニル基、2-(トリフルオロメトキシ)フェニル基、2-(1,1,2,2-テトラフルオロエトキシ)フェニル基、2-フェノキシフェニル基、2-(ピロリジン-1-イル)ピリジン-3-イル基、または2-モルホリノピリジン-3-イル基から選ばれる基である。
[1-9-2]
In the formula (I) of the above embodiment [1], the combination of rings B, Q and (R 2 ) n is more preferably a 5-fluoro-3- (pyrimidin-2-yl) pyridin-2-yl group, 2,2′-bipyridin) -3-yl group, 2- (1H-1,2,3-triazol-1-yl) pyridin-3-yl group, 2- (1H-pyrazol-1-yl) pyridine- 3-yl group, 2- (1-methyl-1H-pyrazol-3-yl) pyridin-3-yl group, 2- (2H-1,2,3-triazol-2-yl) pyridin-3-yl group 2- (3-cyano-1H-pyrazol-1-yl) pyridin-3-yl group, 2- (3-methyl-1H-pyrazol-1-yl) pyridin-3-yl group, 2- (thiazol- 2-yl) pyridin-3-yl group, 2,5- (1H-pyrazole 1-yl) pyridin-3-yl group, 4-ethoxy-2- (1H-pyrazol-1-yl) pyridin-3-yl group, 4-methoxy-2- (1H-pyrazol-1-yl) pyridine- 3-yl group, 5 ″ -chloro- [2,2 ′: 5 ′, 3 ″ -terpyridine] -3′-yl group, 5-fluoro-2- (1H-pyrazol-1-yl) pyridine -3-yl group, 6-methoxy-2- (1H-pyrazol-1-yl) pyridin-3-yl group, 2- (2H-1,2,3-triazol-2-yl) pyridin-4-yl Group, 3- (1H-pyrazol-1-yl) pyridazin-4-yl group, [1,1′-biphenyl] group, 2- (1H-pyrazol-1-yl) phenyl group, 2- (2H-1 , 2,3-Triazol-2-yl) phenyl group, 2- (2H-tetrazole-2- Yl) phenyl group, 2- (3-methyl-1,2,4-oxadiazol-5-yl) phenyl group, 2- (3-methylisothiazol-5-yl) phenyl group, 2- (4- Cyanothiazol-2-yl) phenyl group, 2- (6-cyanopyridin-2-yl) phenyl group, 2- (6-methylpyridin-2-yl) phenyl group, 2- (6-methoxypyridine-2- Yl) phenyl group, 2- (isothiazol-4-yl) phenyl group, 2- (oxazol-2-yl) phenyl group, 2- (thiazol-2-yl) phenyl group, 2- (thiazol-4-yl) ) Phenyl group, 2- (thiazol-5-yl) phenyl group, 2- (pyridin-2-yl) phenyl group, 2- (pyrimidin-2-yl) phenyl group, 2-chloro-6- (2H-1) , 2,3-Triazo Ru-2-yl) phenyl group, 2-fluoro-6- (pyrimidin-2-yl) phenyl group, 2-methoxy-6- (2H-1,2,3-triazol-2-yl) phenyl group, 3 '-(Difluoromethoxy)-[1,1'-biphenyl] -2-yl group, 3'-methoxy- [1,1'-biphenyl] -2-yl group, 3-fluoro-2- (2H-1 , 2,3-triazol-2-yl) phenyl group, 4′-fluoro- [1,1′-biphenyl] -2-yl group, 4-chloro-2- (2H-1,2,3-triazole- 2-yl) phenyl group, 4-chloro-2- (pyrimidin-2-yl) phenyl group, 4-fluoro-2- (2H-1,2,3-triazol-2-yl) phenyl group, 4-fluoro -2- (pyrimidin-2-yl) phenyl group, 4-methyl-2- 2H-1,2,3-triazol-2-yl) phenyl group, 4-methyl-2- (pyrimidin-2-yl) phenyl group, 4-methoxy-2- (1H-1,2,3-triazole- 1-yl) phenyl group, 4-methoxy-2- (2H-1,2,3-triazol-2-yl) phenyl group, 5-fluoro-2- (2H-1,2,3-triazole-2- Yl) phenyl group, 5-methyl-2- (2H-1,2,3-triazol-2-yl) phenyl group, 2-cyclopropylphenyl group, 2,6-diethoxyphenyl group, 2- (trifluoro Methoxy) phenyl group, 2- (1,1,2,2-tetrafluoroethoxy) phenyl group, 2-phenoxyphenyl group, 2- (pyrrolidin-1-yl) pyridin-3-yl group, or 2-morpholinopyridine -3 -A group selected from yl groups.
[1-9-3]
 上記態様[1]の式(I)において、環B、Qおよび(R2)nの組み合わせとして特に好ましくは、2-(2H-1,2,3-トリアゾール-2-イル)フェニル基、2-フルオロ-6-(2H-1,2,3-トリアゾール-2-イル)フェニル基、5-メチル-2-(2H-1,2,3-トリアゾール-2-イル)フェニル基、2-(3-メチル-1,2,4-オキサジアゾール-5-イル)フェニル基、2-(1H-ピラゾール-1-イル)ピリジン-3-イル基、2-(2,2,2-トリフルオロエトキシ)フェニル基、または2-(1,1,2,2-テトラフルオロエトキシ)フェニル基から選ばれる基である。
[1-9-3]
In the formula (I) of the above embodiment [1], the combination of rings B, Q and (R 2 ) n is particularly preferably a 2- (2H-1,2,3-triazol-2-yl) phenyl group, 2 -Fluoro-6- (2H-1,2,3-triazol-2-yl) phenyl group, 5-methyl-2- (2H-1,2,3-triazol-2-yl) phenyl group, 2- ( 3-methyl-1,2,4-oxadiazol-5-yl) phenyl group, 2- (1H-pyrazol-1-yl) pyridin-3-yl group, 2- (2,2,2-trifluoro) An ethoxy) phenyl group or a 2- (1,1,2,2-tetrafluoroethoxy) phenyl group.
[1-10]
 上記態様[1]の式(I)において、環B、Qおよび(R2)nの組み合わせとして好ましくは、以下の部分構造式である式(BQ-6)または式(BQ-7)(但し、式(BQ-6)及び式(BQ-7)中、-C(O)-基は環B、Qおよび(R2)nの組み合わせに含まない):
Figure JPOXMLDOC01-appb-C000013
[式(BQ-6)および式(BQ-7)中、pおよびR3は、上記態様[1-5]中の定義と同じであり;Xは、NまたはC-Hを表し;Y2は、O、S、N-H、またはN-CH3を表し;R2eは、水素原子、シアノ基、C1~6アルキル基、またはハロゲン化C1~6アルキル基を表す]である。
[1-10]
In the formula (I) of the above embodiment [1], the combination of the ring B, Q and (R 2 ) n is preferably the following partial structural formula (BQ-6) or (BQ-7) (provided that In the formulas (BQ-6) and (BQ-7), the —C (O) — group is not included in the combination of ring B, Q and (R 2 ) n ):
Figure JPOXMLDOC01-appb-C000013
[In formula (BQ-6) and formula (BQ-7), p and R 3 are the same as defined in the above embodiment [1-5]; X represents N or C—H; Y 2 Represents O, S, NH, or N—CH 3 ; R 2e represents a hydrogen atom, a cyano group, a C 1-6 alkyl group, or a halogenated C 1-6 alkyl group].
[1-10-1]
 上記態様[1]の式(I)において、環B、Qおよび(R2)nの組み合わせとしてより好ましくは、上記態様[1-10]中の部分構造式である式(BQ-6)または式(BQ-7)[式(BQ-6)および式(BQ-7)中、pは、0または1の整数であり;R3は、シアノ基を表し;Xは、NまたはC-Hを表し;Y2は、Sを表し;R2eは、水素原子、シアノ基、メチル基、またはトリフルオロメチル基を表す]である。
[1-10-1]
In the formula (I) of the embodiment [1], the combination of the rings B, Q and (R 2 ) n is more preferably a formula (BQ-6) which is a partial structural formula in the embodiment [1-10] or Formula (BQ-7) [In Formula (BQ-6) and Formula (BQ-7), p is an integer of 0 or 1; R 3 represents a cyano group; X is N or C—H. Y 2 represents S; R 2e represents a hydrogen atom, a cyano group, a methyl group, or a trifluoromethyl group.
[1-10-2]
 上記態様[1]の式(I)において、環B、Qおよび(R2)nの組み合わせとして更に好ましくは、3-メチル-1-(3-シアノフェニル)-1H-ピラゾール-5-イル基、1-(ピリジン-2-イル)-1H-ピラゾール-5-イル基、3-シアノ-1-(ピリジン-2-イル)-1H-ピラゾール-5-イル基、3-メチル-1-(ピリジン-2-イル)-1H-ピラゾール-5-イル基、3-(トリフルオロメチル)-1-(ピリジン-2-イル)-1H-ピラゾール-5-イル基、1-(ピリダジン-3-イル)-1H-ピラゾール-5-イル基、または1-(チアゾール-2-イル)-1H-ピラゾール-5-イル基から選ばれる基である。
[1-10-2]
In the formula (I) of the above embodiment [1], the combination of rings B, Q and (R 2 ) n is more preferably a 3-methyl-1- (3-cyanophenyl) -1H-pyrazol-5-yl group 1- (pyridin-2-yl) -1H-pyrazol-5-yl group, 3-cyano-1- (pyridin-2-yl) -1H-pyrazol-5-yl group, 3-methyl-1- ( Pyridin-2-yl) -1H-pyrazol-5-yl group, 3- (trifluoromethyl) -1- (pyridin-2-yl) -1H-pyrazol-5-yl group, 1- (pyridazine-3- Yl) -1H-pyrazol-5-yl group or 1- (thiazol-2-yl) -1H-pyrazol-5-yl group.
[1-10-3]
 上記態様[1]の式(I)において、環B、Qおよび(R2)nの組み合わせとして特に好ましくは、1-(ピリジン-2-イル)-1H-ピラゾール-5-イル基である。
[1-10-3]
In the formula (I) of the above embodiment [1], the combination of rings B, Q and (R 2 ) n is particularly preferably a 1- (pyridin-2-yl) -1H-pyrazol-5-yl group.
[1-11]
 上記態様[1]の式(I)において、好ましくは、mは、上記態様[1-1]中の定義と同じであり;nは、上記態様[1-2]中の定義と同じであり;環Aは、上記態様[1-3]中の定義と同じであり;環Bは、上記態様[1-4]中の定義と同じであり;Qは、上記態様[1-5]中の定義と同じであり;R1は、上記態様[1-6]中の定義と同じであり;R2は、上記態様[1-7]中の定義と同じである。
[1-11]
In the formula (I) of the above embodiment [1], preferably, m is the same as defined in the above embodiment [1-1]; n is the same as defined in the above embodiment [1-2]. Ring A is the same as defined in the above embodiment [1-3]; ring B is the same as defined in the above embodiment [1-4]; Q is defined in the above embodiment [1-5]. R 1 is the same as defined in the above embodiment [1-6]; R 2 is the same as defined in the above embodiment [1-7].
[1-11-1]
 上記態様[1]の式(I)において、より好ましくは、mは、上記態様[1-1-1]中の定義と同じであり;nは、上記態様[1-2-1]中の定義と同じであり;環Aは、上記態様[1-3-2]中の定義と同じであり;環Bは、上記態様[1-4-2]中の定義と同じであり;Qは、上記態様[1-5-2]中の定義と同じであり;R1は、上記態様[1-6-1]中の定義と同じであり;R2は、上記態様[1-7-1]中の定義と同じである。
[1-11-1]
In the formula (I) of the embodiment [1], more preferably, m is the same as defined in the embodiment [1-1-1]; n is the same as in the embodiment [1-2-1]. Ring A is the same as defined in the above embodiment [1-3-2]; ring B is the same as defined in the above embodiment [1-4-2]; R 1 is the same as defined in the above embodiment [1-6-1]; R 2 is defined as in the above embodiment [1-7-]. 1] Same definition as in [1].
[1-12]
上記態様[1]の式(I)において、好ましくは、環A、および(R1)mの組み合わせは、上記態様[1-8]中の定義と同じであり;環B、Qおよび(R2)nの組み合わせは、上記態様[1-9]中または上記態様[1-10]中の定義と同じである。
[1-12]
In the formula (I) of the embodiment [1], the combination of the ring A and (R 1 ) m is preferably the same as defined in the embodiment [1-8]; the rings B, Q and (R 2 ) The combination of n is the same as defined in the above embodiment [1-9] or the above embodiment [1-10].
[1-12-1]
上記態様[1]の式(I)において、好ましくは、環A、および(R1)mの組み合わせは、上記態様[1-8-3]中の定義と同じであり;環B、Qおよび(R2)nの組み合わせは、上記態様[1-9-3]中または上記態様[1-10-3]中の定義と同じである。
[1-12-1]
In the formula (I) of the above embodiment [1], preferably, the combination of the ring A and (R 1 ) m is the same as defined in the above embodiment [1-8-3]; The combination of (R 2 ) n is the same as defined in the above embodiment [1-9-3] or the above embodiment [1-10-3].
[2] 上記態様[1]の上記式(I)の化合物において、好ましい化合物は、下記式(I-a):
Figure JPOXMLDOC01-appb-C000014
[式(I-a)中、m、n、Q、R1、およびR2は上記態様[1]中の定義と同じであり;環A’中のX1、X2、およびX3は、各々独立して、NまたはC-Hを表し;環B’中のX4、X5、X6、およびX7は、各々独立して、NまたはC-Hを表す]で表される化合物である。また、本発明の好ましい態様として、製薬学的に許容される上記式(I-a)の化合物の塩またはそれらの溶媒和物が挙げられる。
[2] Among the compounds of the above formula (I) of the above embodiment [1], preferred compounds are those represented by the following formula (Ia):
Figure JPOXMLDOC01-appb-C000014
[In the formula (Ia), m, n, Q, R 1 and R 2 are the same as defined in the above embodiment [1]; X 1 , X 2 and X 3 in the ring A ′ are Each independently represents N or C—H; each of X 4 , X 5 , X 6 , and X 7 in ring B ′ independently represents N or C—H]. A compound. Further, as a preferred embodiment of the present invention, a pharmaceutically acceptable salt of the compound of the above formula (Ia) or a solvate thereof can be mentioned.
[2-1]
 上記態様[2]の式(I-a)において、好ましくは、m=0~3の整数である。
[2-1-1]
 上記態様[2]の式(I-a)において、より好ましくは、m=2である。
[2-1]
In the formula (Ia) of the above embodiment [2], m is preferably an integer of 0 to 3.
[2-1-1]
In the formula (Ia) of the above embodiment [2], m = 2 is more preferable.
[2-2]
 上記態様[2]の式(I-a)において、好ましくは、n=0~2の整数である。
[2-2-1]
 上記態様[2]の式(I-a)において、より好ましくは、n=0または1である。
[2-2]
In the formula (Ia) of the above embodiment [2], n is preferably an integer of 0 to 2.
[2-2-1]
In the formula (Ia) of the above embodiment [2], n = 0 or 1 is more preferable.
[2-3]
 上記態様[2]の式(I-a)において、環A’における、X1、X2、およびX3の組み合わせとして好ましくは、(X1=N、X2=X3=C-H)、(X1=X2=N、X3=C-H)、(X1=C-H、X2=X3=N)、または(X1=X2=X3=C-H)である。
[2-3]
In the formula (Ia) of the above embodiment [2], the combination of X 1 , X 2 and X 3 in the ring A ′ is preferably (X 1 = N, X 2 = X 3 = C—H) (X 1 = X 2 = N, X 3 = C—H), (X 1 = C—H, X 2 = X 3 = N), or (X 1 = X 2 = X 3 = C—H) It is.
[2-3-1]
 態様[2]の式(I-a)において、環A’における、X1、X2、およびX3の組み合わせとしてより好ましくは、(X1=N、X2=X3=C-H)、または(X1=X2=N、X3=C-H)である。
[2-3-1]
In the formula (Ia) of the embodiment [2], the combination of X 1 , X 2 and X 3 in the ring A ′ is more preferably (X 1 = N, X 2 = X 3 = C—H). Or (X 1 = X 2 = N, X 3 = C—H).
[2-4]
 態様[2]の式(I-a)において、環B’における、X4、X5、X6、およびX7の組み合わせとして好ましくは、(X4=N、X5=X6=X7=C-H)、(X4=X5=X6=C-H、X7=N)、(X4=X5=X7=C-H、X6=N)、(X4=X5=C-H、X6=X7=N)、または(X4=X5=X6=X7=C-H)である。
[2-4]
In the formula (Ia) of the embodiment [2], the combination of X 4 , X 5 , X 6 and X 7 in the ring B ′ is preferably (X 4 = N, X 5 = X 6 = X 7 = C-H), (X 4 = X 5 = X 6 = C-H, X 7 = N), (X 4 = X 5 = X 7 = C-H, X 6 = N), (X 4 = X 5 = C—H, X 6 = X 7 = N), or (X 4 = X 5 = X 6 = X 7 = C—H).
[2-4-1]
 上記態様[2]の式(I-a)において、環B’における、X4、X5、X6、およびX7の組み合わせとしてより好ましくは、(X4=N、X5=X6=X7=C-H)、(X4=X5=X6=C-H、X7=N)、または(X4=X5=X6=X7=C-H)である。
[2-4-1]
In the formula (Ia) of the above embodiment [2], the combination of X 4 , X 5 , X 6 and X 7 in ring B ′ is more preferably (X 4 = N, X 5 = X 6 = X 7 = C-H), a (X 4 = X 5 = X 6 = C-H, X 7 = N), or (X 4 = X 5 = X 6 = X 7 = C-H).
[2-5]
 上記態様[2]の式(I-a)において、Qとして好ましくは、C3~8シクロアルキル基、-ORc2(当該Rc2は、C1~6アルキル基、ハロゲン化C1~6アルキル基、およびC6~14アリール基から任意に選ばれる基を表し)、または部分構造式(SF-1)で表される基: 
Figure JPOXMLDOC01-appb-C000015
[式(SF-1)中、環C、p、R3は、上記態様[1-5]中の定義と同じである]である。
[2-5]
In the formula (I-a) of the above embodiment [2], preferably, C 3 ~ 8 cycloalkyl group, -OR c2 (the R c2 is, C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl as Q groups, and C 6 ~ 14 represent optionally a chosen group aryl group), or a group represented by the partial structural formula (SF-1):
Figure JPOXMLDOC01-appb-C000015
[In the formula (SF-1), rings C, p and R 3 are the same as defined in the above embodiment [1-5]].
[2-5-1]
 上記態様[2]の式(I-a)において、Qとしてより好ましくは、C3~8シクロアルキル基、-ORc2(当該Rc2は、C1~6アルキル基、ハロゲン化C1~6アルキル基、およびC6~14アリール基から任意に選ばれる基を表す)、または部分構造式(SF-1)で表される基:
Figure JPOXMLDOC01-appb-C000016
[式(SF-1)中、環Cは、フェニル基、ピリジン-2-イル基、ピリミジン-2-イル基、1H-ピラゾール-1-イル基、1-メチル-1H-ピラゾール-3-イル基、1H-ピラゾール-3-イル基、1H-ピラゾール-4-イル基、1H-1,2,3-トリアゾール-1-イル基、2H-1,2,3-トリアゾール-2-イル基、1,2,4-オキサジアゾール-5-イル基、チアゾール-2-イル基、チアゾール-4-イル基、オキサゾール-2-イル基、イソチアゾール-4-イル基、イソチアゾール-5-イル基、または2H-テトラゾール-2-イル基から選ばれる基を表し;pおよびR3は、上記態様[1-5-1]中の定義と同じである]である。
[2-5-1]
In the formula (I-a) of the above embodiment [2], is more preferably Q, C 3 ~ 8 cycloalkyl group, -OR c2 (the R c2 is, C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl groups, and C 6 ~ 14 represent the chosen group optionally aryl group), or a partial structural formula (SF-1) a group represented by:
Figure JPOXMLDOC01-appb-C000016
[In Formula (SF-1), Ring C is a phenyl group, a pyridin-2-yl group, a pyrimidin-2-yl group, a 1H-pyrazol-1-yl group, or a 1-methyl-1H-pyrazol-3-yl group. A group, 1H-pyrazol-3-yl group, 1H-pyrazol-4-yl group, 1H-1,2,3-triazol-1-yl group, 2H-1,2,3-triazol-2-yl group, 1,2,4-oxadiazol-5-yl group, thiazol-2-yl group, thiazol-4-yl group, oxazol-2-yl group, isothiazol-4-yl group, isothiazol-5-yl Or a group selected from 2H-tetrazol-2-yl group; p and R 3 are as defined in the above embodiment [1-5-1].
[2-5-2]
 上記態様[2]の式(I-a)において、Qとして更に好ましくは、上記態様[1-5-2]中の定義と同じである。
[2-5-2]
In the formula (Ia) of the above embodiment [2], Q is more preferably the same as defined in the above embodiment [1-5-2].
[2-6]
 上記態様[2]の式(I-a)において、R1として好ましくは、ハロゲン原子、シアノ基、-Ra1、-OR1、-C(O)Ra1、-C(O)ORa1、-SRa1、-NRAB基、または3~8員非芳香族複素環基(上記Ra1、RA、およびRBは、上記態様[1-6]中の定義と同じである)である。
[2-6]
In the formula (Ia) of the above embodiment [2], R 1 is preferably a halogen atom, a cyano group, —R a1 , —OR 1 , —C (O) R a1 , —C (O) OR a1 , —SR a1 , —NR A R B group, or 3- to 8-membered non-aromatic heterocyclic group (the above R a1 , R A , and R B are the same as defined in the above embodiment [1-6]) It is.
[2-6-1]
 上記態様[2]の式(I-a)において、R1としてより好ましくは、上記態様[1-6-1]中の定義と同じである。
[2-6-1]
In the formula (Ia) of the above embodiment [2], R 1 is more preferably the same as defined in the above embodiment [1-6-1].
[2-7]
 上記態様[2]の式(I-a)において、R2として好ましくは、ハロゲン原子、-Rb1、-ORb1(当該Rb1は、上記態様[1-7]中の定義と同じである)、ピラゾリル基、またはピリジル基(当該ピラゾリル基およびピリジル基は、ハロゲン原子、およびC1~6アルキル基で1~4個置換されていても良い)である。
[2-7]
In the formula (Ia) of the above embodiment [2], R 2 is preferably a halogen atom, —R b1 , —OR b1 (wherein R b1 has the same definition as in the above embodiment [1-7]). ), A pyrazolyl group, or a pyridyl group (the pyrazolyl group and the pyridyl group may be substituted with 1 to 4 halogen atoms and a C 1-6 alkyl group).
[2-7-1]
 上記態様[2]の式(I-a)において、R2としてより好ましくは、ハロゲン原子、またはC1~6アルキル基を表し、より具体的には、フッ素およびメチル基である。
[2-7-1]
In the formula (Ia) of the above embodiment [2], R 2 represents more preferably a halogen atom or a C 1-6 alkyl group, and more specifically a fluorine and a methyl group.
[2-8]
 上記態様[2]の式(I-a)において、環A’および(R1)mの組み合わせとして好ましくは、上記態様[1-8]中の環Aおよび(R1)mの定義と同じである。
[2-8]
In the formula (Ia) of the above embodiment [2], the combination of the ring A ′ and (R 1 ) m is preferably the same as the definition of the ring A and (R 1 ) m in the above embodiment [1-8]. It is.
[2-8-1]
 上記態様[2]の式(I-a)において、環A’および(R1)mの組み合わせとしてより好ましくは、上記態様[1-8-1]中の環Aおよび(R1)mの定義と同じである。
[2-8-1]
In the formula (Ia) of the embodiment [2], the combination of the ring A ′ and (R 1 ) m is more preferably a combination of the ring A and the (R 1 ) m in the embodiment [1-8-1]. Same as definition.
[2-8-2]
 上記態様[2]の式(I-a)において、環A’および(R1)mの組み合わせとして更に好ましくは、上記態様[1-8-2]中の環Aおよび(R1)mの定義と同じである。
[2-8-2]
In the formula (Ia) of the embodiment [2], the combination of the ring A ′ and (R 1 ) m is more preferably a combination of the ring A and the (R 1 ) m in the embodiment [1-8-2]. Same as definition.
[2-8-3]
 上記態様[2]の式(I-a)において、環A’および(R1)mの組み合わせとして特に好ましくは、上記態様[1-8-3]中の環Aおよび(R1)mの定義と同じである。
[2-8-3]
In the formula (Ia) of the embodiment [2], the combination of the ring A ′ and (R 1 ) m is particularly preferably a combination of the ring A and the (R 1 ) m in the embodiment [1-8-3]. Same as definition.
[2-9]
 上記態様[2]の式(I-a)において、環B’、Qおよび(R2)nの組み合わせとして好ましくは、上記態様[1-9]中の環B、Qおよび(R2)nの定義と同じである。
[2-9]
In the formula (Ia) of the embodiment [2], the combination of the rings B ′, Q and (R 2 ) n is preferably a ring B, Q and (R 2 ) n in the embodiment [1-9]. The definition is the same as
[2-9-1]
 上記態様[2]の式(I-a)において、環B’、Qおよび(R2)nの組み合わせとしてより好ましくは、上記態様[1-9-1]中の環B、Qおよび(R2)nの定義と同じである。
[2-9-1]
In the formula (Ia) of the embodiment [2], the combination of the rings B ′, Q and (R 2 ) n is more preferably a ring B, Q and (R) in the embodiment [1-9-1]. 2 ) Same definition as n .
[2-9-2]
 上記態様[2]の式(I-a)において、環B’、Qおよび(R2)nの組み合わせとして更に好ましくは、上記態様[1-9-2]中の環B、Qおよび(R2)nの定義と同じである。
[2-9-2]
In the formula (Ia) of the embodiment [2], the combination of the rings B ′, Q and (R 2 ) n is more preferably a combination of the rings B, Q and (R) in the embodiment [1-9-2]. 2 ) Same definition as n .
[2-9-3]
 上記態様[2]の式(I-a)において、環B’、Qおよび(R2)nの組み合わせとして特に好ましくは、上記態様[1-9-3]中の環B、Qおよび(R2)nの定義と同じである。
[2-9-3]
In the formula (Ia) of the above embodiment [2], the combination of the rings B ′, Q and (R 2 ) n is particularly preferably a ring B, Q and (R) in the above embodiment [1-9-3]. 2 ) Same definition as n .
[2-10]
 上記態様[2]の式(I-a)において、好ましくは、mは、上記態様[2-1]中の定義と同じであり;nは、上記態様[2-2]中の定義と同じであり;環A’は、上記態様[2-3]中の定義と同じであり;環Bは、上記態様[2-4]中の定義と同じであり;Qは、上記態様[2-5]中の定義と同じであり;R1は、上記態様[2-6]中の定義と同じであり;R2は、上記態様[2-7]中の定義と同じである。
[2-10]
In the formula (Ia) of the above embodiment [2], preferably, m is the same as defined in the above embodiment [2-1]; n is the same as defined in the above embodiment [2-2]. Ring A ′ is the same as defined in the above embodiment [2-3]; Ring B is the same as defined in the above embodiment [2-4]; Q is the above described embodiment [2-2]. 5] in the same definition as in; R 1 are as defined in the above aspect [2-6]; R 2 is the same as defined in the above aspect [2-7].
[2-10-1]
 上記態様[2]の式(I-a)において、より好ましくは、mは、上記態様[2-1-1]中の定義と同じであり;nは、上記態様[2-2-1]中の定義と同じであり;環Aは、上記態様[2-3-2]中の定義と同じであり;環Bは、上記態様[2-4-2]中の定義と同じであり;Qは、上記態様[2-5-2]中の定義と同じであり;R1は、上記態様[2-6-1]中の定義と同じであり;R2は、上記態様[2-7-1]中の定義と同じである。
[2-10-1]
In the formula (Ia) of the above embodiment [2], more preferably, m is the same as defined in the above embodiment [2-1-1]; n is the above embodiment [2-2-1]. Ring A is the same as defined in the above embodiment [2-3-2]; ring B is the same as defined in the above embodiment [2-4-2]; Q is the same as defined in the above embodiment [2-5-2]; R 1 is the same as defined in the above embodiment [2-6-1]; R 2 is defined in the above embodiment [2-5-2]. 7-1] The same definition as in [7-1].
[2-11]
 上記態様[2]の式(I-a)において、好ましくは、環A’および(R1)mの組み合わせは、上記態様[2-8]中の定義と同じであり;環B’、Qおよび(R2)nの組み合わせは、上記態様[2-9]中の定義と同じである。
[2-11]
In the formula (Ia) of the above embodiment [2], the combination of the ring A ′ and (R 1 ) m is preferably the same as defined in the above embodiment [2-8]; And the combination of (R 2 ) n is the same as defined in the above embodiment [2-9].
[2-11-1]
 上記態様[2]の式(I-a)において、より好ましくは、環A’および(R1)mの組み合わせは、上記態様[2-8-3]中の定義と同じであり;環B’、Qおよび(R2)nの組み合わせは、上記態様[2-9-3]中の定義と同じである。
[2-11-1]
In the formula (Ia) of the above embodiment [2], more preferably, the combination of the ring A ′ and (R 1 ) m is the same as defined in the above embodiment [2-8-3]; The combination of ', Q and (R 2 ) n is the same as defined in the above embodiment [2-9-3].
[3] 上記態様[1]の上記式(I)の化合物において、好ましい化合物は、下記式(I-b):
Figure JPOXMLDOC01-appb-C000017
[式(I-b)中、m、n、Q、R1、およびR2は上記態様[1]中の定義と同じであり;環A’中のX1、X2およびX3は、各々独立して、NまたはC-Hを表す]で表される化合物である。また、本発明の好ましい態様として、製薬学的に許容される上記式(I-b)の化合物の塩またはそれらの溶媒和物が挙げられる。
[3] Among the compounds of the above formula (I) of the above embodiment [1], preferred compounds are those represented by the following formula (Ib):
Figure JPOXMLDOC01-appb-C000017
[In the formula (Ib), m, n, Q, R 1 and R 2 are the same as defined in the above embodiment [1]; X 1 , X 2 and X 3 in the ring A ′ are Each independently represents N or C—H]. Further, as a preferred embodiment of the present invention, a pharmaceutically acceptable salt of the compound of the above formula (Ib) or a solvate thereof can be mentioned.
[3-1]
 上記態様[3]の式(I-b)において、好ましくは、m=0~3の整数である。
[3-1-1]
 上記態様[3]の式(I-b)において、より好ましくは、m=2である。
[3-1]
In the formula (Ib) of the above embodiment [3], m is preferably an integer of 0 to 3.
[3-1-1]
In the formula (Ib) of the above embodiment [3], m = 2 is more preferable.
[3-2]
 上記態様[3]の式(I-b)において、好ましくは、n=0、または1である。
[3-2-1]
 上記態様[3]の式(I-b)において、より好ましくは、n=0である。
[3-2]
In the formula (Ib) of the above embodiment [3], preferably n = 0 or 1.
[3-2-1]
In the formula (Ib) of the above embodiment [3], n = 0 is more preferable.
[3-3]
 上記態様[3]の式(I-b)において、環A’における、X1、X2、およびX3の組み合わせとして好ましくは、上記態様[2-3]中の定義と同じである。
[3-3]
In the formula (Ib) of the embodiment [3], the combination of X 1 , X 2 and X 3 in the ring A ′ is preferably the same as defined in the embodiment [2-3].
[3-3-1]
 上記態様[3]の式(I-b)において、環A’における、X1、X2、およびX3の組み合わせとしてより好ましくは、(X1=X2=N、X3=C-H)である。
[3-3-1]
In the formula (Ib) of the above embodiment [3], the combination of X 1 , X 2 and X 3 in the ring A ′ is more preferably (X 1 = X 2 = N, X 3 = C—H ).
[3-4]
 上記態様[3]の式(I-b)において、Qとして好ましくは、C3~8シクロアルキル基、または部分構造式(SF-1)で表される基: 
Figure JPOXMLDOC01-appb-C000018
[式(SF-1)中、環C、p、R3は、上記態様[1-5]中の定義と同じである]である。
[3-4]
In the formula (I-b) of the above embodiment [3], is preferably a Q, represented by C 3 ~ 8 cycloalkyl group or partial structural formula, (SF-1) group:
Figure JPOXMLDOC01-appb-C000018
[In the formula (SF-1), rings C, p and R 3 are the same as defined in the above embodiment [1-5]].
[3-4-1]
 上記態様[3]の式(I-b)において、Qとしてより好ましくは、部分構造式(SF-1)で表される基: 
Figure JPOXMLDOC01-appb-C000019
[式(SF-1)中、環Cは、フェニル基、ピリジン-2-イル基、ピリダジン-3-イル基、またはチアゾール-2-イル基から選ばれる基を表し;pおよびR3は、上記態様[1-5-1]中の定義と同じである]である。
[3-4-1]
In the formula (Ib) of the above embodiment [3], more preferably, Q is a group represented by the partial structural formula (SF-1):
Figure JPOXMLDOC01-appb-C000019
Wherein (SF-1), ring C is a phenyl group, pyridin-2-yl group, pyridazin-3-yl group or represents a group selected from thiazol-2-yl group,; p and R 3 are, It is the same as defined in the above embodiment [1-5-1].
[3-4-2]
 上記態様[3]の式(I-b)において、Qとして更に好ましくは、ピリジン-2-イル基である。
[3-4-2]
In the formula (Ib) of the above embodiment [3], Q is more preferably a pyridin-2-yl group.
[3-5]
 上記態様[3]の式(I-b)において、R1として好ましくは、ハロゲン原子、シアノ基、-Ra2、または-ORa2(当該Ra2は、C1~6アルキル基、およびハロゲン化C1~6アルキル基から任意に選ばれる基を表す)である。
[3-5]
In the formula (Ib) of the above embodiment [3], R 1 is preferably a halogen atom, a cyano group, —R a2 , or —OR a2 (wherein R a2 is a C 1-6 alkyl group, and a halogenated group). Represents a group arbitrarily selected from a C 1-6 alkyl group.
[3-5-1]
 上記態様[3]の式(I-b)において、R1としてより好ましくは、シアノ基、C1~6アルキル基、または-O-C1~6アルキル基を表し、より具体的には、シアノ基、メチル基、およびメトキシ基である。
[3-5-1]
In the formula (Ib) of the above embodiment [3], R 1 is more preferably a cyano group, a C 1-6 alkyl group, or an —O—C 1-6 alkyl group, and more specifically, A cyano group, a methyl group, and a methoxy group;
[3-6]
 上記態様[3]の式(I-b)において、R2として好ましくは、シアノ基、C1~6アルキル基、またはハロゲン化C1~6アルキル基である。
[3-6]
In the formula (Ib) of the above embodiment [3], R 2 is preferably a cyano group, a C 1-6 alkyl group, or a halogenated C 1-6 alkyl group.
[3-7]
 上記態様[3]の式(I-b)において、環A’および(R1)mの組み合わせとして好ましくは、上記態様[1-8]中の環Aおよび(R1)mの定義と同じである。
[3-7]
In the formula (Ib) of the embodiment [3], the combination of the ring A ′ and (R 1 ) m is preferably the same as the definition of the ring A and (R 1 ) m in the embodiment [1-8]. It is.
[3-7-1]
 上記態様[3]の式(I-b)において、環A’および(R1)mの組み合わせとしてより好ましくは、上記態様[1-8-1]中の環Aおよび(R1)mの定義と同じである。
[3-7-1]
In the formula (Ib) of the embodiment [3], the combination of the ring A ′ and (R 1 ) m is more preferably a combination of the ring A and the (R 1 ) m in the embodiment [1-8-1]. Same as definition.
[3-7-2]
 上記態様[3]の式(I-b)において、環A’および(R1)mの組み合わせとして更に好ましくは、4-シアノ-ピリジン-2-イル基、4-トリフルオロメチル-ピリジン-2-イル基、4-シアノ-3-メトキシ-ピリジン-2-イル基、4-シアノ-3-シクロプロピル-ピリジン-2-イル基、3-メトキシ-4-トリフルオロメチル-ピリジン-2-イル基、または4,6-ジメチルピリミジン-2-イル基から選ばれる基である。
[3-7-2]
In the formula (Ib) of the above embodiment [3], the combination of the ring A ′ and (R 1 ) m is more preferably 4-cyano-pyridin-2-yl group, 4-trifluoromethyl-pyridine-2. -Yl group, 4-cyano-3-methoxy-pyridin-2-yl group, 4-cyano-3-cyclopropyl-pyridin-2-yl group, 3-methoxy-4-trifluoromethyl-pyridin-2-yl Or a group selected from a 4,6-dimethylpyrimidin-2-yl group.
[3-7-3]
 上記態様[3]の式(I-b)において、環A’および(R1)mの組み合わせとして特に好ましくは、4,6-ジメチルピリミジン-2-イル基である。
[3-7-3]
In the formula (Ib) of the above embodiment [3], the combination of the ring A ′ and (R 1 ) m is particularly preferably a 4,6-dimethylpyrimidin-2-yl group.
[3-8]
 上記態様[3]の式(I-b)において、1H-ピラゾール環、Qおよび(R2)nの組み合わせとして好ましくは、上記態様[1-10]中の定義と同じである。
[3-8]
In the formula (Ib) of the above embodiment [3], the combination of 1H-pyrazole ring, Q and (R 2 ) n is preferably the same as defined in the above embodiment [1-10].
[3-8-1]
 上記態様[3]の式(I-b)において、1H-ピラゾール環、Qおよび(R2)nの組み合わせとしてより好ましくは、上記態様[1-10-1]中の定義と同じである。
[3-8-1]
In the formula (Ib) of the above embodiment [3], the combination of 1H-pyrazole ring, Q and (R 2 ) n is more preferably the same as the definition in the above embodiment [1-10-1].
[3-8-2]
 上記態様[3]の式(I-b)において、1H-ピラゾール環、Qおよび(R2)nの組み合わせとして更に好ましくは、上記態様[1-10-2]中の定義と同じである。
[3-8-2]
In the formula (Ib) of the above embodiment [3], the combination of 1H-pyrazole ring, Q and (R 2 ) n is more preferably the same as defined in the above embodiment [1-10-2].
[3-8-3]
 上記態様[3]の式(I-b)において、1H-ピラゾール環、Qおよび(R2)nの組み合わせとして特に好ましくは、上記態様[1-10-3]中の定義と同じである。
[3-8-3]
In the formula (Ib) of the above embodiment [3], the combination of 1H-pyrazole ring, Q and (R 2 ) n is particularly preferably the same as defined in the above embodiment [1-10-3].
[3-9]
 上記態様[3]の式(I-b)において、好ましくは、mは、上記態様[3-1]中の定義と同じであり;nは、上記態様[3-2]中の定義と同じであり;環A’は、上記態様[3-3]中の定義と同じであり;Qは、上記態様[3-4]中の定義と同じであり;R1は、上記態様[3-5]中の定義と同じであり;R2は、上記態様[3-6]中の定義と同じである。
[3-9]
In the formula (Ib) of the above embodiment [3], preferably, m is the same as defined in the above embodiment [3-1]; n is the same as defined in the above embodiment [3-2]. Ring A ′ is the same as defined in the above embodiment [3-3]; Q is the same as defined in the above embodiment [3-4]; R 1 is the above defined embodiment [3- 5] is the same as defined in R; R 2 is the same as defined in the above embodiment [3-6].
[3-9-1]
 上記態様[3]の式(I-b)において、より好ましくは、mは、上記態様[3-1-1]中の定義と同じであり;nは、上記態様[3-2-1]中の定義と同じであり;環Aは、上記態様[3-3-1]中の定義と同じであり;Qは、上記態様[3-4-2]中の定義と同じであり;R1は、上記態様[3-5-1]中の定義と同じであり;R2は、上記態様[3-6]中の定義と同じである。
[3-9-1]
In the formula (Ib) of the above embodiment [3], more preferably, m is the same as defined in the above embodiment [3-1-1]; n is the above embodiment [3-2-1]. Ring A is the same as defined in the above embodiment [3-3-1]; Q is the same as defined in the above embodiment [3-4-2]; R 1 is the same as defined in the above embodiment [3-5-1]; R 2 is the same as defined in the above embodiment [3-6].
[3-10]
 上記態様[3]の式(I-b)において、好ましくは、環A’および(R1)mの組み合わせは、上記態様[3-7]中の環Aおよび(R1)mの定義と同じであり;1H-ピラゾール環、Q、および(R2)nの組み合わせは、上記態様[3-8]中の定義と同じである。
[3-10]
In the formula (Ib) of the above embodiment [3], preferably the combination of the ring A ′ and (R 1 ) m is the same as the definition of the ring A and (R 1 ) m in the above embodiment [3-7]. The combination of 1H-pyrazole ring, Q, and (R 2 ) n is the same as defined in the above embodiment [3-8].
[3-10-1]
 上記態様[3]の式(I-b)において、より好ましくは、環A’および(R1)mの組み合わせは、上記態様[3-7-3]中の定義と同じであり;1H-ピラゾール環、Q、および(R2)nの組み合わせは、上記態様[3-8-3]中の定義と同じである。
[3-10-1]
In the formula (Ib) of the embodiment [3], more preferably, the combination of the ring A ′ and (R 1 ) m is the same as defined in the embodiment [3-7-3]; The combination of the pyrazole ring, Q, and (R 2 ) n is the same as defined in the above embodiment [3-8-3].
 以上、本発明の態様[1]~[3]までの各々及びその好ましい態様を、更には置換基の定義を適宜組み合わせることにより、上記態様[1]の上記式(I)で表される化合物の好ましい態様を任意に形成しうる。 As described above, each of the embodiments [1] to [3] of the present invention and preferred embodiments thereof, and further by appropriately combining the definitions of substituents, the compound represented by the above formula (I) of the above embodiment [1] The preferred embodiments can be arbitrarily formed.
[4]本発明の第4の態様は、上記態様[1]の上記式(I)の化合物において、好ましい化合物として、以下に列挙される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物、或いはそれらの光学異性体である。なお、以下に表される化合物の名称は、ChemBioDraw(登録商標) Ultra 14.0.0.117(PerkinElmer Informatics)の化合物名称命名プログラムに従って得られる英語名称に基づくものである。
Figure JPOXMLDOC01-appb-T000020
[4] The fourth aspect of the present invention is the compound of the above formula (I) of the above aspect [1], wherein the preferred compounds are the compounds listed below, or pharmaceutically acceptable salts thereof, or those: Solvates thereof, or optical isomers thereof. The names of the compounds shown below are based on English names obtained according to the compound name naming program of ChemBioDraw (registered trademark) Ultra 14.0.0.117 (PerkinElmer Informations).
Figure JPOXMLDOC01-appb-T000020
 本明細書中、特に断りのない限り、環状基に可変置換基が置換している場合、該可変置換基は環状基の特定の炭素原子に結合されていない事を意味する。例えば、下記式Aにおける可変置換基Rxは、該式Aにおける炭素原子i、ii、iiiまたはivの何れかに置換する事ができ、下記式Bにおける可変置換基Ryは、該式Bにおける炭素原子v、またはviの何れかに置換する事ができる事を意味する。
Figure JPOXMLDOC01-appb-C000021
In the present specification, unless otherwise specified, when a cyclic group is substituted with a variable substituent, it means that the variable substituent is not bonded to a specific carbon atom of the cyclic group. For example, the variable substituent R x in the following formula A can be substituted on any of the carbon atoms i, ii, iii or iv in the formula A, and the variable substituent R y in the following formula B can be substituted with the formula B It can be substituted with either carbon atom v or vi.
Figure JPOXMLDOC01-appb-C000021
[5]本発明の第5の態様は、上記式(I)で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の少なくとも1つを有効成分として含有することを特徴とする、医薬組成物である。 [5] A fifth aspect of the present invention contains at least one of the compound represented by the above formula (I) or a pharmaceutically acceptable salt or solvate thereof as an active ingredient. It is a pharmaceutical composition characterized by these.
[6]本発明の第6の態様は、上記式(I)で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の少なくとも1つを有効成分として含有することを特徴とする、OXRが関与する疾患の予防及び/または治療剤である。 [6] A sixth aspect of the present invention contains at least one of the compound represented by the above formula (I), a pharmaceutically acceptable salt thereof, or a solvate thereof as an active ingredient. It is a prophylactic and / or therapeutic agent for diseases involving OXR.
 OXRは、生物学的機能に広く関わっている。これは、ヒト、または他種での多様な疾患の経過におけるOXRの潜在的な役割を示唆している。 OXR is widely involved in biological functions. This suggests a potential role for OXR in the course of various diseases in humans or other species.
 OXRが関与する疾患として、不眠症、概日リズム睡眠障害、睡眠時随伴症等の睡眠障害;うつ病、不安障害、双極性障害、注意欠陥多動性障害、自閉症、自閉症スペクトラム障害、薬物依存症等の精神疾患;アルツハイマー病等の神経変性疾患;認知症等の記憶障害;および過食症等の摂食障害、等が挙げられる。 Diseases involving OXR include sleep disorders such as insomnia, circadian rhythm sleep disorder, and sleep-related disorders; depression, anxiety disorder, bipolar disorder, attention deficit hyperactivity disorder, autism, autism spectrum Mental disorders such as disorders, drug dependence; neurodegenerative diseases such as Alzheimer's disease; memory disorders such as dementia; and eating disorders such as bulimia.
[8]本発明の第8の態様は、上記式(I)で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の少なくとも1つを有効成分として含有することを特徴とする、睡眠障害、精神疾患、神経変性疾患、記憶障害または摂食障害の予防および/または治療剤である。 [8] An eighth aspect of the present invention contains at least one of the compound represented by the above formula (I), or a pharmaceutically acceptable salt or solvate thereof as an active ingredient. A prophylactic and / or therapeutic agent for sleep disorders, mental disorders, neurodegenerative diseases, memory disorders or eating disorders.
[8-1]
 好ましくは、上記式(I)で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の少なくとも1つを有効成分として含有することを特徴とする、睡眠障害の予防及び/または治療剤であり、より好ましくは、不眠症の予防および/または治療剤である。
[8-1]
Preferably, at least one of the compound represented by the above formula (I), a pharmaceutically acceptable salt thereof or a solvate thereof is contained as an active ingredient, thereby preventing sleep disorder And / or a therapeutic agent, more preferably a prophylactic and / or therapeutic agent for insomnia.
 本明細書中、特に断りのない限り、「睡眠障害」として、具体的には、不眠症、概日リズム睡眠障害、睡眠時随伴症等が挙げられる。不眠症として、より具体的には、原発性不眠症、精神疾患による不眠症、身体疾患による不眠症、薬物による不眠症等が挙げられる。但し、これらに限定されるものではない。 Unless otherwise specified, in this specification, specific examples of the “sleep disorder” include insomnia, circadian rhythm sleep disorder, and sleep-related complications. More specific examples of insomnia include primary insomnia, insomnia due to mental illness, insomnia due to physical disease, insomnia due to drugs, and the like. However, it is not limited to these.
 本明細書中、特に断りのない限り、「精神疾患」として、具体的には、うつ病、不安障害、双極性障害、注意欠陥多動性障害、自閉症、自閉症スペクトラム障害、薬物依存症等が挙げられる。但し、これらに限定されるものではない。 In this specification, unless otherwise specified, as a “mental disorder”, specifically, depression, anxiety disorder, bipolar disorder, attention deficit hyperactivity disorder, autism, autism spectrum disorder, drug Addiction etc. are mentioned. However, it is not limited to these.
 本明細書中、特に断りのない限り、「神経変性疾患」として、具体的には、アルツハイマー病等が挙げられる。但し、これらに限定されるものではない。 Unless otherwise specified, in this specification, specific examples of “neurodegenerative diseases” include Alzheimer's disease. However, it is not limited to these.
 本明細書中、特に断りのない限り、「記憶障害」として、具体的には、認知症等が挙げられる。但し、これらに限定されるものではない。 In the present specification, unless otherwise specified, the “memory disorder” specifically includes dementia and the like. However, it is not limited to these.
 本明細書中、特に断りのない限り、「摂食障害」として、具体的には、過食症等が挙げられる。但し、これらに限定されるものではない。 Unless otherwise specified, in this specification, “eating disorders” specifically includes bulimia and the like. However, it is not limited to these.
[9]本発明の第9の態様は、上記式(I)で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の1つ以上を含むOXR拮抗剤である。 [9] A ninth aspect of the present invention is an OXR antagonist comprising one or more of the compounds represented by the above formula (I), or a pharmaceutically acceptable salt or solvate thereof. .
[10]本発明の第10の態様は、上記式(I)で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の1つ以上を含むOX2R選択的拮抗剤である。
 本発明の別の態様は、上記式(I)で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の1つ以上を含むOXRdual拮抗剤である。ここで、「OXRdual拮抗剤」とは、オレキシン1受容体拮抗作用とオレキシン2受容体拮抗作用とを有するオレキシン受容体拮抗剤を意味する。
[10] A tenth aspect of the present invention is an OX2R selective antagonist comprising one or more of the compounds represented by the above formula (I), or a pharmaceutically acceptable salt or solvate thereof. It is.
Another aspect of the present invention is an OXR dual antagonist comprising one or more of the compounds represented by formula (I) above, or a pharmaceutically acceptable salt or solvate thereof. Here, the “OXR dual antagonist” means an orexin receptor antagonist having an orexin 1 receptor antagonistic action and an orexin 2 receptor antagonistic action.
[11]本発明の第11の態様は、上記式(I)で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の少なくとも1つの医薬組成物としての使用である。 [11] An eleventh aspect of the present invention is the use of at least one pharmaceutical composition of a compound represented by the above formula (I), or a pharmaceutically acceptable salt or solvate thereof. is there.
[12]本発明の第12の態様は、上記式(I)で表される化合物、または薬学的に許容できるその塩またはそれらの溶媒和物の少なくとも1つのオレキシン受容体拮抗剤としての使用である。 [12] A twelfth aspect of the present invention is the use of a compound represented by the above formula (I), or a pharmaceutically acceptable salt thereof or a solvate thereof as at least one orexin receptor antagonist. is there.
[13]本発明の第13の態様は、睡眠障害、精神疾患、神経変性疾患、記憶障害および摂食障害から選択される疾患を治療する方法であって、上記式(I)で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の少なくとも1つを上記疾患または状態の治療を必要とする対象に投与することを含む方法である。 [13] A thirteenth aspect of the present invention is a method for treating a disease selected from sleep disorder, mental disorder, neurodegenerative disorder, memory disorder and eating disorder, and is represented by the above formula (I) Administering at least one of a compound, or a pharmaceutically acceptable salt or solvate thereof, to a subject in need of treatment of the disease or condition.
[13-1]
 好ましくは、睡眠障害を治療する方法であって、上記式(I)で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の少なくとも1つを上記疾患または状態の治療を必要とする対象に投与することを含む方法であり、より好ましくは不眠症を治療する方法である。
[13-1]
Preferably, there is provided a method for treating a sleep disorder, wherein at least one of the compound represented by the above formula (I), or a pharmaceutically acceptable salt thereof or a solvate thereof is administered in the disease or condition. A method comprising administering to a subject in need of treatment, more preferably a method of treating insomnia.
 本明細書中、特に断りのない限り、「疾患または状態の治療」にあるような「治療」とは、「疾患または状態」の進行、または1つまたは複数の「疾患または状態」を回復させる、緩和する、または抑制することを意味する。また、本明細書中、「治療」は、患者の状態に応じて、「疾患または状態」の発症またはその「疾患または状態」に関連する任意の症状の発症を予防することを包含する「疾患または状態」の予防、ならびに発症前に「疾患または状態」またはその任意の症状の重症度を低減することも包含する。本明細書では、「治療する」はある「疾患または状態」の再発を予防するおよび改善することも含むものとする。 In this specification, unless stated otherwise, “treatment” as in “treatment of a disease or condition” refers to the progression of a “disease or condition” or one or more “diseases or conditions”. Means to mitigate, or suppress. Further, in the present specification, “treatment” refers to “disease” including preventing the onset of “disease or condition” or any symptoms related to “disease or condition” depending on the condition of the patient. Or prevention of “a condition” as well as reducing the severity of a “disease or condition” or any symptom thereof before onset. As used herein, “treating” is intended to include preventing and ameliorating the recurrence of a “disease or condition”.
[14]本発明の第14の態様は、上記疾患が、不眠症、概日リズム睡眠障害、睡眠時随伴症、うつ病、不安障害、双極性障害、注意欠陥多動性障害、自閉症、自閉症スペクトラム障害、薬物依存症、アルツハイマー病、認知症、過食症の群から選択される態様[8]または[8-1]に記載の予防および/または治療剤、または態様[13]または[13-1]に記載の方法である。 [14] In a fourteenth aspect of the present invention, the disease is insomnia, circadian rhythm sleep disorder, parasomnia, depression, anxiety disorder, bipolar disorder, attention deficit hyperactivity disorder, autism Or a prophylactic and / or therapeutic agent according to aspect [8] or [8-1] selected from the group consisting of: autism spectrum disorder, drug dependence, Alzheimer's disease, dementia, bulimia, or aspect [13] Or it is the method as described in [13-1].
 本発明の化合物は、オレキシン受容体拮抗作用を適宜選択した方法、例えば、後述の薬理実験例1(オレキシン受容体拮抗活性評価)で測定した場合、OXRに対するIC50値が1μM以下である化合物が好ましい。より好ましくは、オレキシン受容体に対するIC50値が100nM以下である化合物である。また、OX2R拮抗作用を有する化合物として、OX2Rに対するIC50値が1μM以下である化合物が好ましく、より好ましくは100nM以下である化合物である。このような活性を有する化合物は、OXR拮抗作用、とりわけOX2R拮抗作用を有する化合物であり、OXR拮抗剤、とりわけOX2R拮抗剤、または医薬組成物として、利用可能である。また、OXRが関与する疾患、睡眠障害(不眠症、概日リズム睡眠障害、睡眠時随伴症等)、精神疾患(うつ病、不安障害、双極性障害、注意欠陥多動性障害、自閉症、自閉症スペクトラム障害、薬物依存症等)、神経変性疾患(アルツハイマー病等)、記憶障害(認知症等)または摂食障害(過食症等)、とりわけOX2Rが関与する疾患、または不眠症等の睡眠障害の予防および/または治療剤として、利用可能である。 The compound of the present invention is a compound having an IC 50 value for OXR of 1 μM or less when measured by a method in which orexin receptor antagonism is appropriately selected, for example, Pharmacological Experiment Example 1 (orexin receptor antagonism evaluation) described later. preferable. More preferred is a compound having an IC 50 value for the orexin receptor of 100 nM or less. Further, as a compound having a OX2R antagonism is preferably a compound an IC 50 value is less than 1μM for OX2R, and more preferably not more than 100nM compound. The compound having such an activity is a compound having an OXR antagonistic action, particularly an OX2R antagonistic action, and can be used as an OXR antagonist, particularly an OX2R antagonist, or a pharmaceutical composition. In addition, diseases involving OXR, sleep disorders (insomnia, circadian rhythm sleep disorders, sleep-related complications, etc.), mental disorders (depression, anxiety disorders, bipolar disorder, attention deficit hyperactivity disorder, autism , Autism spectrum disorders, drug addiction, etc.), neurodegenerative diseases (Alzheimer's disease etc.), memory disorders (dementia etc.) or eating disorders (eg bulimia), especially OX2R-related diseases, insomnia, etc. It can be used as a preventive and / or therapeutic agent for sleep disorders.
 また、本発明の化合物は、オレキシン2受容体拮抗作用を有するが、オレキシン1受容体拮抗作用を有さないあるいは低い作用を有する化合物を包含しており、このような化合物は、オレキシン2受容体選択的な拮抗作用を有する化合物、またはオレキシン2受容体選択的拮抗剤として、利用可能である。例えば、オレキシン1受容体に対するIC50値が、オレキシン2受容体に対するIC50値の10倍以上、20倍以上、50倍以上または100倍以上である化合物が挙げられる。オレキシン2受容体選択的な拮抗作用を有する化合物は、オレキシン1受容体拮抗作用による副作用を有さない医薬組成物として有用である。また、オレキシン2受容体が関与する疾患、睡眠障害(不眠症、概日リズム睡眠障害、睡眠時随伴症等)、精神疾患(うつ病、不安障害、双極性障害、注意欠陥多動性障害、自閉症、自閉症スペクトラム障害、薬物依存症等)、神経変性疾患(アルツハイマー病等)、記憶障害(認知症等)または摂食障害(過食症等)、とりわけ不眠症等の睡眠障害の予防及び/または治療剤として、利用可能である。
 本発明の化合物は、オレキシン2受容体拮抗作用と共に、オレキシン1受容体拮抗作用を同程度に有する化合物を包含しており、このような化合物は、オレキシン受容体dual拮抗作用を有する化合物であり、オレキシン受容体dual拮抗剤または医薬組成物として、利用可能である。また、上述したオレキシン受容体が関与する疾患の予防及び/または治療剤として、利用可能である。
The compounds of the present invention include compounds having orexin 2 receptor antagonistic activity but not having orexining orexin 1 receptor antagonistic activity. Such compounds include orexin 2 receptor. It can be used as a compound having a selective antagonism or an orexin 2 receptor selective antagonist. For example, a compound having an IC 50 value for orexin 1 receptor of 10 times or more, 20 times or more, 50 times or more, or 100 times or more of the IC 50 value for orexin 2 receptor can be mentioned. A compound having an orexin 2 receptor selective antagonism is useful as a pharmaceutical composition having no side effects due to orexin 1 receptor antagonism. In addition, diseases involving orexin 2 receptor, sleep disorders (insomnia, circadian rhythm sleep disorders, sleep-related complications, etc.), mental disorders (depression, anxiety disorder, bipolar disorder, attention deficit hyperactivity disorder, Autism, autism spectrum disorder, drug addiction, etc.), neurodegenerative diseases (Alzheimer's disease, etc.), memory disorders (dementia, etc.) or eating disorders (eg, bulimia), especially sleep disorders such as insomnia It can be used as a preventive and / or therapeutic agent.
The compounds of the present invention include compounds having orexin 2 receptor antagonism as well as orexin 1 receptor antagonism, and such compounds are compounds having orexin receptor dual antagonism, It can be used as an orexin receptor dual antagonist or a pharmaceutical composition. Moreover, it can utilize as a preventive and / or therapeutic agent of the disease in which the above orexin receptor is involved.
 本明細書中、特に断りのない限り、「式(I)の化合物」、「式(I)で表される化合物」等と記載している場合、「式(I-a)の化合物」、「式(I-b)の化合物」、「式(I-c)の化合物」の下位概念にあたる化合物にも言及するものとする。 In the present specification, unless otherwise specified, when “a compound of formula (I)”, “a compound represented by formula (I)” or the like is described, “a compound of formula (Ia)”, Reference is also made to compounds that are subordinate concepts of “compound of formula (Ib)” and “compound of formula (Ic)”.
 以上の全ての態様において、「化合物」の文言を用いるとき、「その製薬学的に許容される塩または当該化合物及び当該塩の溶媒和物」についても言及するものとする。 In all of the above embodiments, when the term “compound” is used, “a pharmaceutically acceptable salt or the compound and a solvate of the salt” is also referred to.
 本発明の化合物は、置換基の種類によって、酸付加塩を形成する場合や塩基との塩を形成する場合がある。かかる塩としては、製薬学的に許容しうる塩であれば特に限定されないが、例えば、金属塩、アンモニウム塩、有機塩基との塩、無機酸との塩、有機酸との塩、塩基性、又は酸性アミノ酸との塩などが挙げられる。金属塩の好適な例としては、例えば、リチウム塩、ナトリウム塩、カリウム塩、セシウム塩などのアルカリ金属塩、カルシウム塩、マグネシウム塩、バリウム塩などのアルカリ土類金属塩、アルミニウム塩などが挙げられる(例えば、モノ塩の他、二ナトリウム塩、二カリウム塩も含む)。有機塩基との塩の好適な例としては、例えば、メチルアミン、エチルアミン、t-ブチルアミン、t-オクチルアミン、ジエチルアミン、トリメチルアミン、トリエチルアミン、シクロヘキシルアミン、ジシクロヘキシルアミン、ジベンジルアミン、エタノールアミン、ジエタノールアミン、トリエタノールアミン、ピペリジン、モルホリン、ピリジン、ピコリン、リシン、アルギニン、オルニチン、エチレンジアミン、N-メチルグルカミン、グルコサミン、フェニルグリシンアルキルエステル、グアニジン、2,6-ルチジン、エタノールアミン、ジエタノールアミン、トリエタノールアミン、N,N'-ジベンジルエチレンジアミン等との塩が挙げられる。無機酸との塩の好適な例としては、例えば、塩酸、臭化水素酸、よう化水素酸、硝酸、硫酸、リン酸等との塩が挙げられる。有機酸との塩の好適な例としては、例えば、ギ酸、酢酸、トリフルオロ酢酸、プロピオン酸、酪酸、吉草酸、エナント酸、カプリン酸、ミリスチン酸、パルミチン酸、ステアリン酸、乳酸、ソルビン酸、マンデル酸等の脂肪族モノカルボン酸等との塩、シュウ酸、マロン酸、コハク酸、フマル酸、マレイン酸、リンゴ酸、酒石酸等の脂肪族ジカルボン酸との塩、クエン酸等の脂肪族トリカルボン酸との塩、安息香酸、サリチル酸等の芳香族モノカルボン酸との塩、フタル酸等の芳香族ジカルボン酸の塩、桂皮酸、グリコール酸、ピルビン酸、オキシル酸、サリチル酸、N-アセチルシステイン等の有機カルボン酸との塩、メタンスルホン酸、ベンゼンスルホン酸、p-トルエンスルホン酸等の有機スルホン酸との塩、アスパラギン酸、グルタミン酸等の酸性アミノ酸類との酸付加塩が挙げられる。塩基性アミノ酸との塩の好適な例としては、例えば、アルギニン、リジン、オルニチンなどとの塩が挙げられ、酸性アミノ酸との塩の好適な例としては、例えば、アスパラギン酸、グルタミン酸などとの塩が挙げられる。このうち、薬学的に許容し得る塩が好ましい。例えば、化合物内に酸性官能基を有する場合にはアルカリ金属塩(例、ナトリウム塩、カリウム塩など)、アルカリ土類金属塩(例、カルシウム塩、マグネシウム塩、バリウム塩など)などの無機塩、アンモニウム塩など、また、化合物内に塩基性官能基を有する場合には、例えば、塩酸、臭化水素酸、硝酸、硫酸、リン酸など無機酸との塩、又は酢酸、フタル酸、フマル酸、シュウ酸、酒石酸、マレイン酸、クエン酸、コハク酸、メタンスルホン酸、p-トルエンスルホン酸などの有機酸との塩が挙げられる。 The compound of the present invention may form an acid addition salt or a salt with a base depending on the type of substituent. Such a salt is not particularly limited as long as it is a pharmaceutically acceptable salt. For example, metal salts, ammonium salts, salts with organic bases, salts with inorganic acids, salts with organic acids, basics, Or the salt with an acidic amino acid etc. are mentioned. Preferable examples of the metal salt include alkali metal salts such as lithium salt, sodium salt, potassium salt and cesium salt, alkaline earth metal salts such as calcium salt, magnesium salt and barium salt, and aluminum salt. (For example, besides a mono salt, a disodium salt and a dipotassium salt are also included). Preferable examples of the salt with an organic base include, for example, methylamine, ethylamine, t-butylamine, t-octylamine, diethylamine, trimethylamine, triethylamine, cyclohexylamine, dicyclohexylamine, dibenzylamine, ethanolamine, diethanolamine, triamine. Ethanolamine, piperidine, morpholine, pyridine, picoline, lysine, arginine, ornithine, ethylenediamine, N-methylglucamine, glucosamine, phenylglycine alkyl ester, guanidine, 2,6-lutidine, ethanolamine, diethanolamine, triethanolamine, N , N′-dibenzylethylenediamine and the like. Preferable examples of the salt with inorganic acid include salts with hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, sulfuric acid, phosphoric acid and the like. Preferable examples of the salt with an organic acid include, for example, formic acid, acetic acid, trifluoroacetic acid, propionic acid, butyric acid, valeric acid, enanthic acid, capric acid, myristic acid, palmitic acid, stearic acid, lactic acid, sorbic acid, Salts with aliphatic monocarboxylic acids such as mandelic acid, salts with aliphatic dicarboxylic acids such as oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, malic acid, tartaric acid, and aliphatic tricarboxylic acids such as citric acid Salts with acids, salts with aromatic monocarboxylic acids such as benzoic acid and salicylic acid, salts of aromatic dicarboxylic acids such as phthalic acid, cinnamic acid, glycolic acid, pyruvic acid, oxylic acid, salicylic acid, N-acetylcysteine, etc. Salt with organic carboxylic acid, salt with organic sulfonic acid such as methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, aspartic acid, gluta Acid addition salts with acidic amino acids such as phosphate and the like. Preferable examples of salts with basic amino acids include, for example, salts with arginine, lysine, ornithine, and preferable examples of salts with acidic amino acids include, for example, salts with aspartic acid, glutamic acid, and the like. Is mentioned. Of these, pharmaceutically acceptable salts are preferred. For example, when the compound has an acidic functional group, an inorganic salt such as an alkali metal salt (eg, sodium salt, potassium salt), an alkaline earth metal salt (eg, calcium salt, magnesium salt, barium salt), In the case where the compound has a basic functional group, for example, a salt with an inorganic acid such as hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, or acetic acid, phthalic acid, fumaric acid, Examples thereof include salts with organic acids such as oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, methanesulfonic acid, and p-toluenesulfonic acid.
 上記塩は、常法に従い、例えば、本発明の化合物と適量の酸もしくは塩基を含む溶液を混合することにより目的の塩を形成させた後に分別濾取するか、もしくは該混合溶媒を留去することにより得ることができる。また、本発明の化合物またはその塩は、水、エタノール、グリセロール等の溶媒と溶媒和物を形成しうる。 According to a conventional method, for example, the salt is formed by mixing the compound of the present invention with a solution containing an appropriate amount of acid or base to form the desired salt, and then separated by filtration, or the mixed solvent is distilled off. Can be obtained. In addition, the compound of the present invention or a salt thereof can form a solvate with a solvent such as water, ethanol or glycerol.
 塩に関する総説として、Handbook of Pharmaceutical Salts:Properties,Selection,and Use、Stahl&Wermuth(Wiley-VCH、2002)が出版されており、本書に詳細な記載がなされている。 Handbook of Pharmaceutical Salts: Properties, Selection, and Use, Stahl & Wermuth (Wiley-VCH, 2002) are published as detailed reviews on salt, and are described in detail in this document.
 本発明の化合物は、非溶媒和形態もしくは溶媒和形態で存在することができる。本明細書において、「溶媒和物」は、本発明の化合物と1種または複数の薬学的に許容される溶媒分子(例えば、水、エタノール等)を含む分子複合体を意味する。上記溶媒分子が水であるとき、特に「水和物」と言う。 The compounds of the present invention can exist in unsolvated or solvated forms. As used herein, “solvate” means a molecular complex comprising a compound of the invention and one or more pharmaceutically acceptable solvent molecules (eg, water, ethanol, etc.). When the solvent molecule is water, it is particularly called “hydrate”.
 以下、本発明の化合物に関する記載においては、その塩、溶媒和物、ならびにその塩の溶媒和物に関する記載を包含する。 Hereinafter, the description relating to the compound of the present invention includes the description relating to the salt, solvate, and solvate of the salt.
 本発明の化合物の「プロドラッグ」も本発明の化合物に包含される。「プロドラッグ」とは、例えば、目的とする薬理活性を殆どもしくは全く示さない可能性のある、本発明の化合物のある種の誘導体を、身体内または身体上に投与した時、例えば、加水分解等により目的とする薬理活性を有する、本発明の化合物に変換される場合、投与前の化合物のことを「プロドラッグ」と呼ぶ。 “Prodrugs” of the compounds of the present invention are also encompassed by the compounds of the present invention. A “prodrug” is, for example, when a certain derivative of a compound of the invention, which may exhibit little or no desired pharmacological activity, is administered in or on the body, eg, hydrolysis. When converted to the compound of the present invention having the desired pharmacological activity by the like, the compound before administration is called “prodrug”.
 本発明の化合物の「プロドラッグ」は、例えば、本発明の化合物に存在する適切な官能基を、文献公知の方法、例えば、Design of Prodrugs、H.Bundgaard(Elsevier、1985)に記載されている方法に準じることで製造することができる。 The “prodrug” of the compound of the present invention can be prepared by, for example, converting an appropriate functional group present in the compound of the present invention into a method known in the literature, for example, Design of Products, H.P. It can be produced according to the method described in Bundgaard (Elsevier, 1985).
 本発明の化合物の「プロドラッグ」としては、具体的には、以下の(1)~(3)に示すものがあり得るが、これに限定されるものではない。
(1)本発明の化合物にカルボキシ基(-COOH)が置換している場合:そのエステル、即ち、該カルボキシ基(-COOH)の水素原子が「C1~6アルキル基」で置換されている化合物。
(2)本発明の化合物に水酸基(-OH)が置換している場合:そのアルカノイルオキシもしくはエーテル、即ち、該水酸基(-OH)の水素原子が「C2~7アルカノイル基」もしくは「C2~7アルカノイルオキシメチル基」で置換されている化合物。
(3)本発明の化合物にアミノ基(-NH2もしくは-NHR’(式中、R’≠H))が置換している場合:そのアミド、即ち、該アミノ基(-NH2もしくは-NHR’(式中、R’≠H))の一方または両方の水素原子が「C2~7アルカノイル基」で置換されている化合物。
Specific examples of the “prodrug” of the compound of the present invention include the following (1) to (3), but are not limited thereto.
(1) When the compound of the present invention is substituted with a carboxy group (—COOH): the ester, ie, the hydrogen atom of the carboxy group (—COOH) is substituted with a “C 1-6 alkyl group” Compound.
(2) When the hydroxyl group (-OH) to a compound of the present invention is substituted: Part alkanoyloxy or ethers, i.e., a hydrogen atom is "C 2 ~ 7 alkanoyl group" in the hydroxyl group (-OH) or "C 2 compounds substituted with 1-7 alkanoyloxymethyl group ".
(3) When the compound of the present invention is substituted with an amino group (—NH 2 or —NHR ′ (where R ′ ≠ H)): its amide, that is, the amino group (—NH 2 or —NHR) '(wherein, R' ≠ H)) one or both of the hydrogen atoms "C 2 ~ 7 alkanoyl group" compounds substituted with a.
 本発明の化合物が、幾何異性体(ジオメトリカルアイソマー)、配置異性体(コンフィギュレーショナルアイソマー)、互変異性体(トウトメリックアイソマー)、光学異性体(オプティカルアイソマー)、立体異性体(ジアステレオマー)、位置異性体(レジオアイソマー)、回転異性体(ロテイショナルアイソマー)などの異性体を有する場合には、いずれか一方の異性体も混合物も本発明の化合物に包含される。さらに、本発明の化合物に光学異性体が存在する場合には、ラセミ体から分割された光学異性体も本発明の化合物に包含される。 The compound of the present invention is a geometric isomer (geometric isomer), configurational isomer (configurational isomer), tautomer (tortomeric isomer), optical isomer (optical isomer), stereoisomer (diastereomer). , Positional isomers (Regio isomers), rotational isomers (rotational isomers) and the like, both isomers and mixtures are also included in the compound of the present invention. Furthermore, when an optical isomer exists in the compound of the present invention, the optical isomer resolved from the racemate is also encompassed in the compound of the present invention.
 本発明の化合物に、1つまたは複数の不斉炭素原子が有る場合には、2種以上の立体異性体が存在できる。また、本発明の化合物に、「C2~6アルケニル基」が含まれる場合、幾何異性体(シス/トランス、またはZ/E)が存在できる。また、構造異性体が低いエネルギー障壁により相互変換可能である場合には、互変異性(tautomeric isomerism)が生じうる。互変異性としては、例えば、イミノ、ケト、もしくはオキシム基を有する化合物においてプロトン互変異性の形態が挙げられる。 When the compound of the present invention has one or more asymmetric carbon atoms, two or more stereoisomers can exist. Further, the compounds of the present invention, if it contains "C 2 ~ 6 alkenyl group", geometric isomers (cis / trans or Z / E,) can be present. Also, tautomerism can occur when structural isomers can be interconverted by a low energy barrier. Examples of tautomerism include proton tautomerism in compounds having an imino, keto, or oxime group.
 本発明の化合物の式(I)における、ジアザビシクロ[2.2.2]オクタン骨格(式(OC)、但し式(OC)中、-C(O)-基はジアザビシクロ[2.2.2]オクタン骨格に含まない):
Figure JPOXMLDOC01-appb-C000022
には、光学異性体が存在し得る。本明細書中、特に断りが無い限り、式(OC)には(1S,4S)体(式(OC-1))、および(1R,4R)体(式(OC-2))の両異性体が含まれる事を意味する。
Figure JPOXMLDOC01-appb-C000023
A diazabicyclo [2.2.2] octane skeleton (formula (OC) in the formula (I) of the compound of the present invention, wherein —C (O) — group is diazabicyclo [2.2.2] Not included in the octane skeleton):
Figure JPOXMLDOC01-appb-C000022
May have optical isomers. In the present specification, unless otherwise specified, the formula (OC) includes both (1S, 4S) form (formula (OC-1)) and (1R, 4R) form (formula (OC-2)) isomers. It means that the body is included.
Figure JPOXMLDOC01-appb-C000023
 なお、本発明の実施例化合物において、式(OC)の1位および4位に*マークが有る化合物は、式(OC-1)、または式(OC-2)のいずれかの立体配置を有している化合物を意味する。
Figure JPOXMLDOC01-appb-C000024
In the example compounds of the present invention, the compounds having * mark at the 1-position and 4-position of the formula (OC) have either steric configuration of the formula (OC-1) or the formula (OC-2). Meaning a compound.
Figure JPOXMLDOC01-appb-C000024
 本発明の化合物に、幾何異性体、配置異性体、立体異性体、配座異性体等が存在する場合には、公知の手段によりそれぞれを単離することができる。 When there are geometrical isomers, configurational isomers, stereoisomers, conformational isomers and the like in the compound of the present invention, each can be isolated by a known means.
 また、本発明の化合物が光学活性体である場合には、ラセミ体を通常の光学分割手段により(+)体もしくは(-)体[D体もしくはL体]に分離することができる。 In addition, when the compound of the present invention is an optically active substance, the racemate can be separated into a (+) isomer or a (−) isomer [D isomer or L isomer] by an ordinary optical resolution means.
 本発明の化合物が、光学異性体、立体異性体、位置異性体、回転異性体、互変異性体を含有する場合には、自体公知の合成手法、分離手法により各々の異性体を単一の化合物として得ることができる。例えば、光学分割法としては、自体公知の方法、例えば、(1)分別再結晶法、(2)ジアステレオマー法、(3)キラルカラム法等が挙げられる。 When the compound of the present invention contains optical isomers, stereoisomers, positional isomers, rotational isomers, and tautomers, each isomer is converted to a single isomer by a known synthesis method or separation method. It can be obtained as a compound. For example, examples of the optical resolution method include methods known per se, such as (1) fractional recrystallization method, (2) diastereomer method, (3) chiral column method and the like.
 (1)分別再結晶法:ラセミ体に対して光学分割剤をイオン結合させることにより結晶性のジアステレオマーを得た後、これを分別再結晶法によって分離し、所望により、中和工程を経てフリーの光学的に純粋な化合物を得る方法である。光学分割剤としては、例えば、(+)-マンデル酸、(-)-マンデル酸、(+)-酒石酸、(-)-酒石酸、(+)-1-フェネチルアミン、(-)-1-フェネチルアミン、シンコニン、(-)-シンコニジン、ブルシン等が挙げられる。 (1) Fractionation recrystallization method: After obtaining a crystalline diastereomer by ion-bonding an optical resolving agent to a racemate, it is separated by a fractional recrystallization method and, if desired, a neutralization step is performed. This is a method for obtaining a free optically pure compound. Examples of the optical resolution agent include (+)-mandelic acid, (−)-mandelic acid, (+)-tartaric acid, (−)-tartaric acid, (+)-1-phenethylamine, (−)-1-phenethylamine, Examples include cinchonine, (−)-cinchonidine, brucine and the like.
 (2)ジアステレオマー法:ラセミ体の混合物に光学分割剤を共有結合(反応)させ、ジアステレオマーの混合物とした後、これを通常の分離手段(例、分別再結晶、シリカゲルカラムクロマトグラフィー、HPLC(高速液体クロマトグラフィー)等)等を経て光学的に純粋なジアステレオマーへ分離した後、加水分解反応等の化学的な処理により、光学分割剤を除去することにより、光学的に純粋な光学異性体を得る方法である。例えば、本発明の化合物に分子内水酸基または1級、2級アミノ基を有する場合、該化合物と光学活性な有機酸(例、MTPA〔α-メトキシ-α-(トリフルオロメチル)フェニル酢酸〕、(-)-メントキシ酢酸等)等とを縮合反応に付すことにより、それぞれエステル体またはアミド体のジアステレオマーが得られる。一方、本発明の化合物にカルボキシ基が有る場合、該化合物と光学活性アミンまたはアルコール試薬とを縮合反応に付すことにより、それぞれアミド体またはエステル体のジアステレオマーが得られる。上記の分離された各ジアステレオマーは、酸加水分解または塩基性加水分解反応に付すことにより、元の化合物の光学異性体に変換される。 (2) Diastereomer method: An optical resolution agent is covalently bonded (reacted) to a racemic mixture to obtain a mixture of diastereomers, which is then subjected to usual separation means (eg, fractional recrystallization, silica gel column chromatography). , HPLC (High Performance Liquid Chromatography, etc.) etc., and then optically pure by removing the optical resolving agent by chemical treatment such as hydrolysis reaction. This is a method for obtaining an optical isomer. For example, when the compound of the present invention has an intramolecular hydroxyl group or a primary or secondary amino group, the compound and an optically active organic acid (eg, MTPA [α-methoxy-α- (trifluoromethyl) phenylacetic acid], (-)-Menthoxyacetic acid and the like) are subjected to a condensation reaction to obtain ester or amide diastereomers, respectively. On the other hand, when the compound of the present invention has a carboxy group, an amide or ester diastereomer can be obtained by subjecting the compound and an optically active amine or alcohol reagent to a condensation reaction. Each of the separated diastereomers is converted to an optical isomer of the original compound by subjecting it to an acid hydrolysis or basic hydrolysis reaction.
 (3)キラルカラム法:ラセミ体またはその塩をキラルカラム(光学異性体分離用カラム)でのクロマトグラフィーに付すことで、直接光学分割する方法である。例えば、高速液体クロマトグラフィー(High performance liquid chromatography:HPLC)の場合、ダイセル社製CHIRALシリーズ等のキラルカラムに光学異性体の混合物を添加し、水、種々の緩衝液(例、リン酸緩衝液)、有機溶媒(例、エタノール、メタノール、イソプロパノール、アセトニトリル、トリフルオロ酢酸、ジエチルアミン)を単独で、または混合した溶液として用いて、展開させることにより、光学異性体を分離することができる。また、例えば、ガスクロマトグラフィーの場合、CP-Chirasil-DeX CB(ジーエルサイエンス社製)等のキラルカラムを使用して分離することができる。 (3) Chiral column method: a method in which a racemate or a salt thereof is subjected to direct optical resolution by subjecting to a chromatography on a chiral column (optical isomer separation column). For example, in the case of high performance liquid chromatography (High performance liquid chromatography: HPLC), a mixture of optical isomers is added to a chiral column such as Daicel's CHIRAL series, water, various buffers (eg, phosphate buffer), Optical isomers can be separated by developing using an organic solvent (eg, ethanol, methanol, isopropanol, acetonitrile, trifluoroacetic acid, diethylamine) alone or as a mixed solution. In addition, for example, in the case of gas chromatography, the separation can be performed using a chiral column such as CP-Chirasil-DeX CB (manufactured by GL Sciences).
 本発明の化合物が光学活性体である場合には、光学異性体の一方を選択的に合成する不斉合成により合成することができる。光学活性な化合物は、(1)ラセミ化合物をエナンチオ選択的に反応させ光学活性体に導く不斉合成反応、(2)天然に存在する光学活性化合物(糖、アミノ酸等)からジアステレオ選択的に合成する方法、により合成する事ができる。 When the compound of the present invention is an optically active substance, it can be synthesized by asymmetric synthesis in which one of optical isomers is selectively synthesized. Optically active compounds are: (1) asymmetric synthesis reaction in which a racemic compound is enantioselectively reacted to lead to an optically active form, (2) diastereoselective from a naturally occurring optically active compound (sugar, amino acid, etc.). It can be synthesized by the synthesis method.
 本発明の化合物は、結晶であってもよく、結晶形が単一であっても結晶形混合物であっても本発明の化合物に包含される。 The compound of the present invention may be a crystal, and the compound of the present invention includes a single crystal form or a crystal form mixture.
 本発明の化合物は、薬学的に許容され得る共結晶または共結晶塩であってもよい。ここで、共結晶または共結晶塩とは、各々が異なる物理的特性(例えば、構造、融点、融解熱、吸湿性、溶解性および安定性等)を持つ、室温で二種またはそれ以上の独特な固体から構成される結晶性物質を意味する。共結晶または共結晶塩は、自体公知の共結晶化法に従い製造することができる。 The compound of the present invention may be a pharmaceutically acceptable cocrystal or cocrystal salt. Here, co-crystals or co-crystal salts are two or more unique at room temperature, each having different physical properties (eg structure, melting point, heat of fusion, hygroscopicity, solubility and stability). It means a crystalline substance composed of a simple solid. The cocrystal or cocrystal salt can be produced according to a cocrystallization method known per se.
 本発明の化合物には、同位元素(例えば、水素の同位体、2Hおよび3Hなど、炭素の同位体、11C、13C、および14Cなど、塩素の同位体、36Clなど、フッ素の同位体、18Fなど、ヨウ素の同位体、123Iおよび125Iなど、窒素の同位体、13Nおよび15Nなど、酸素の同位体、15O、17O、および18Oなど、リンの同位体、32Pなど、ならびに硫黄の同位体、35Sなど)で標識、又は置換された化合物も包含される。 Compounds of the present invention include isotopes (eg, hydrogen isotopes, 2 H and 3 H, carbon isotopes, 11 C, 13 C, and 14 C, chlorine isotopes, 36 Cl, etc., fluorine Isotopes, 18 F, iodine isotopes, 123 I and 125 I, nitrogen isotopes, 13 N and 15 N, oxygen isotopes, 15 O, 17 O, and 18 O, phosphorus Also included are compounds labeled or substituted with isotopes, 32 P and the like, as well as sulfur isotopes, 35 S and the like.
 ある種の同位元素(例えば、11C、18F、15O、および13Nなどの陽電子放出同位元素)で標識または置換された本発明の化合物は、例えば、陽電子断層法(Positron Emission Tomography;PET)において使用するトレーサー(PETトレーサー)として用いることができ、医療診断などの分野において有用である。 Compounds of the invention labeled or substituted with certain isotopes (eg, positron emitting isotopes such as 11 C, 18 F, 15 O, and 13 N) can be synthesized, for example, by Positron Emission Tomography; PET ) Can be used as a tracer (PET tracer) for use in medical diagnosis and the like.
 ある種の同位体標識で標識または置換された本発明の化合物は、薬物および/または基質の組織分布研究において有用である。例えば、3Hおよび14Cは、それらの標識または置換が容易であり、かつ検出手段が容易である点から、該研究目的において有用である。 Compounds of the invention labeled or substituted with certain isotopic labels are useful in drug and / or substrate tissue distribution studies. For example, 3 H and 14 C are useful for this research purpose because they are easy to label or displace and easy to detect.
 同位体標識された本発明の化合物は、当業者に知られている通常の技法によって、または後述の実施例に記載する合成方法に類似する方法によって得る事ができる。また、非標識化合物の代わりに、得られた同位体標識化合物を薬理実験に用いる事ができる。 The isotope-labeled compound of the present invention can be obtained by a common technique known to those skilled in the art or by a method similar to the synthesis method described in the Examples below. In addition, the obtained isotope-labeled compound can be used for pharmacological experiments instead of the unlabeled compound.
 2H(あるいはDまたは重水素と表記する場合がある)で標識または置換された化合物(D化化合物、重水素化化合物)は、安定性が高くなることが期待され、活性化合物自体として有用である。例えば、代謝を受ける位置の水素原子を2Hに置換した化合物が挙げられ、化合物の性質にほとんど影響を与えずに、代謝反応速度を低下させることができる。また、代謝酵素と不可逆的に結合する位置を2Hに置換した化合物は、その代謝酵素の作用を阻害することを抑制し、併用時の薬物相互作用を軽減することができる。 A compound labeled or substituted with 2 H (or sometimes referred to as D or deuterium) (D compound, deuterated compound) is expected to have high stability and is useful as an active compound itself. is there. For example, a compound in which a hydrogen atom at a position to undergo metabolism is substituted with 2 H can be mentioned, and the metabolic reaction rate can be reduced with little influence on the properties of the compound. In addition, a compound in which the position of irreversibly binding to a metabolic enzyme is substituted with 2 H can suppress the inhibition of the action of the metabolic enzyme, and can reduce the drug interaction at the time of combined use.
 同位体標識された本発明の化合物は、当業者に知られている通常の技法によって、または後述の実施例に記載する合成方法に類似する方法によって得る事ができる。また、非標識化合物の代わりに、得られた同位体標識化合物を薬理実験に用いる事ができる。 The isotope-labeled compound of the present invention can be obtained by a common technique known to those skilled in the art or by a method similar to the synthesis method described in the Examples below. In addition, the obtained isotope-labeled compound can be used for pharmacological experiments instead of the unlabeled compound.
[本発明の化合物の製造方法]
 以下に、本発明の式(I)で表される化合物の製造方法について説明する。本発明の化合物である式(I)で表される化合物、その塩およびそれらの溶媒和物は、市販化合物または市販化合物から文献公知の製造方法により容易に得ることが出来る化合物を出発原料もしくは合成中間体として、既知の一般的化学的な製造方法を組み合わせることで容易に製造することが可能であり、以下に示す代表的な製造方法に従い製造することができる。また、本発明は以下に説明する製造方法に、何ら限定されるものではない。
[Method for producing compound of the present invention]
Below, the manufacturing method of the compound represented by Formula (I) of this invention is demonstrated. The compound of the present invention represented by the formula (I), a salt thereof, and a solvate thereof are commercially available compounds or compounds that can be easily obtained from commercially available compounds by known production methods in the literature as starting materials or synthesis. As an intermediate, it can be easily produced by combining known general chemical production methods, and can be produced according to the following typical production methods. Further, the present invention is not limited to the manufacturing method described below.
 以下の製造方法の各々の式中におけるQ、環A、環B、R1、R2、R3、m、nおよびp等の定義は、特に断らない限り、上記態様に記載された式(I)の各々の定義と同一である。製造方法中におけるMの定義は、特に断らない限り、リチウム、ナトリウム、カリウム等の金属である。製造方法中におけるXの定義は、特に断らない限り、ハロゲン原子、またはトリフルオロメタンスルホニルオキシ(OTf)である。製造方法中におけるARの定義は、特に断らない限り、C6~14アリール基もしくは5~7員ヘテロアリール基である。製造方法中におけるBの定義は、特に断らない限り、ボロン酸エステル、ボロン酸、トリフルオロボレート塩、ボロン酸、またはN-メチルイミノ二酢酸エステルである。製造方法中におけるP1、およびP2の定義は、特に断らない限り、水素原子もしくはイミノ基の保護基である。製造方法中におけるRαの定義は、特に断らない限り、C1~6アルキル基、C6~14アリール基またはベンジル基である。製造方法中におけるRβの定義は、特に断らない限り、C1~6アルキル基またはC3~8シクロアルキル基である。製造方法中におけるRγの定義は、特に断らない限り、C3~8シクロアルキル基、-O-C1~6アルキル基、-S-C1~6アルキル基、3~8員非芳香族複素環基、または-NRAB基である。 Unless otherwise specified, the definitions of Q, ring A, ring B, R 1 , R 2 , R 3 , m, n, p, and the like in each formula of the following production methods are the formulas ( It is the same as each definition of I). The definition of M in the production method is a metal such as lithium, sodium and potassium unless otherwise specified. The definition of X in the production method is a halogen atom or trifluoromethanesulfonyloxy (OTf) unless otherwise specified. Definition of AR in the production process, unless otherwise specified, a C 6 ~ 14 aryl group or 5- to 7-membered heteroaryl group. The definition of B in the production method is boronic acid ester, boronic acid, trifluoroborate salt, boronic acid, or N-methyliminodiacetic acid ester unless otherwise specified. Unless otherwise specified, the definitions of P 1 and P 2 in the production method are a protecting group for a hydrogen atom or an imino group. Defining R alpha in the production process, unless otherwise specified, is a C 1 ~ 6 alkyl group, C 6 ~ 14 aryl group or a benzyl group. Defining R beta in the production process, unless otherwise specified, is a C 1 ~ 6 alkyl group or a C 3 ~ 8 cycloalkyl group. The definition of R gamma in the manufacturing method, unless otherwise specified, C 3-8 cycloalkyl group, -O-C 1 ~ 6 alkyl group, -S-C 1 ~ 6 alkyl group, 3- to 8-membered non-aromatic A heterocyclic group, or a —NR A R B group;
 以下の各製造方法において、式(I)の製造に用いられる各原料化合物は、塩を形成していてもよく、このような塩としては、前述した式(I)の塩と同様のものが挙げられる。また、式(I)の製造に用いられる各原料化合物は、反応液のままか粗製物として次の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単離することもでき、それ自体が公知の手段、例えば、抽出、濃縮、中和、濾過、蒸留、再結晶、クロマトグラフィーなどの分離手段により容易に精製することができる。 In each of the following production methods, each raw material compound used for the production of formula (I) may form a salt, and as such a salt, the same salts as those of the aforementioned formula (I) can be mentioned. Can be mentioned. Each raw material compound used in the production of formula (I) can be used in the next reaction as a reaction solution or as a crude product, but can also be isolated from a reaction mixture according to a conventional method. It can be easily purified by known means, for example, separation means such as extraction, concentration, neutralization, filtration, distillation, recrystallization, chromatography and the like.
 上記再結晶に用いられる溶媒としては、例えば、水;メタノール、エタノール、2-プロパノール、ブタノール等のアルコール類;ジエチルエーテル、テトラヒドロフラン、1,4-ジオキサン等のエーテル類;n-ヘキサン、シクロヘキサン、ヘプタン等の炭化水素類、ベンゼン、トルエン、キシレン等の芳香族炭化水素類;N,N‐ジメチルホルムアミド、N,N‐ジメチルアセトアミド、1,3‐ジメチル‐2‐イミダゾリジノン等のアミド類;クロロホルム、塩化メチレン、1,2‐ジクロロエタン等のハロゲン化炭化水素類;アセトニトリル等のニトリル類;アセトン、ジフェニルケトン等のケトン類;酢酸メチル、酢酸エチル等のエステル類、ジメチルスルホキシド等のスルホキジド類;酢酸、トリフルオロ酢酸、メタンスルホン酸、p‐トルエンスルホン酸等の有機酸類;等が挙げられる。これらの溶媒は、単独で用いることもできるし、二種以上の溶媒を適当な割合、例えば、1:1~1:10の割合で混合して用いてもよい。また、式中の化合物が市販されている場合には市販品をそのまま用いることもでき、自体公知の方法、またはそれに準じた方法にて製造したものを用いることもできる。 Examples of the solvent used for the recrystallization include water; alcohols such as methanol, ethanol, 2-propanol and butanol; ethers such as diethyl ether, tetrahydrofuran and 1,4-dioxane; n-hexane, cyclohexane and heptane. Hydrocarbons such as benzene, toluene, xylene, etc .; amides such as N, N-dimethylformamide, N, N-dimethylacetamide, 1,3-dimethyl-2-imidazolidinone; chloroform , Halogenated hydrocarbons such as methylene chloride and 1,2-dichloroethane; nitriles such as acetonitrile; ketones such as acetone and diphenyl ketone; esters such as methyl acetate and ethyl acetate; sulfoxides such as dimethyl sulfoxide; , Trifluoroacetic acid, methanesulfur Phosphate, organic acids such as p- toluenesulfonic acid; and the like. These solvents can be used alone, or two or more kinds of solvents can be mixed at an appropriate ratio, for example, a ratio of 1: 1 to 1:10. Moreover, when the compound in a formula is marketed, a commercial item can also be used as it is, and what was manufactured by the method known per se or a method according to it can also be used.
 式(I)が有する置換基において、変換可能な官能基(例えば、カルボキシ基、アミノ基、水酸基、カルボニル基、メルカプト基、-C(O)-O-C1~6アルキル基、-C(O)-O-C6~14アリール基、-C(O)-O-ベンジル基、スルホ基、ハロゲン原子等)を含む場合、これらの官能基を自体公知の方法またはそれに準ずる方法によって変換することにより種々の化合物を製造することができる。 In substituent having the formula (I) is transformable functional group (e.g., carboxyl group, an amino group, a hydroxyl group, a carbonyl group, a mercapto group, -C (O) -O-C 1 ~ 6 alkyl group, -C ( O) -O-C 6 ~ 14 aryl group, -C (O) -O- benzyl group, a sulfo group, when containing a halogen atom, etc.), is converted by a method analogous to these functional groups per se known methods or in As a result, various compounds can be produced.
 「カルボキシ基」の場合、例えば、エステル化、還元、アミド化、保護されていてもよいアミノ基への変換反応等の反応により変換可能である。 In the case of a “carboxy group”, it can be converted by a reaction such as esterification, reduction, amidation, or a conversion reaction to an optionally protected amino group.
 「アミノ基」の場合、例えば、アミド化、スルホニル化、ニトロソ化、アルキル化、アリール化、イミド化等の反応により変換可能である。 In the case of an “amino group”, it can be converted by a reaction such as amidation, sulfonylation, nitrosation, alkylation, arylation, imidation and the like.
 「水酸基」の場合、例えば、エステル化、カルバモイル化、スルホニル化、アルキル化、アリール化、酸化、ハロゲン化等の反応により変換可能である。 In the case of “hydroxyl group”, it can be converted by a reaction such as esterification, carbamoylation, sulfonylation, alkylation, arylation, oxidation, halogenation, and the like.
 「カルボニル基」の場合、例えば、還元、酸化、イミノ化(オキシム化、ヒドラゾン化を含む)、(チオ)ケタール化、アルキリデン化、チオカルボニル化等の反応により変換可能である。 In the case of a “carbonyl group”, it can be converted by a reaction such as reduction, oxidation, iminization (including oximation and hydrazone formation), (thio) ketalization, alkylidene formation, thiocarbonylation and the like.
 「メルカプト基」の場合、例えば、アルキル化、酸化等の反応により変換可能である。 In the case of a “mercapto group”, it can be converted by a reaction such as alkylation or oxidation.
 「-C(O)-O-C1~6アルキル基」、「-C(O)-O-C6~14アリール基」、または「-C(O)-O-ベンジル基」の場合、例えば、還元、加水分解等の反応により変換可能である。 For - "C (O) -O-C 6 ~ 14 aryl group" or "-C (O) -O- benzyl group", "-C (O) -O-C 1 ~ 6 alkyl group", For example, it can be converted by a reaction such as reduction or hydrolysis.
 「スルホ基の場合、例えば、スルホンアミド化、還元等の反応により変換可能である。 "In the case of a sulfo group, it can be converted by a reaction such as sulfonamidation or reduction.
 「ハロゲン原子」の場合、例えば、各種求核置換反応、各種カップリング反応等により変換可能である。 In the case of a “halogen atom”, it can be converted by, for example, various nucleophilic substitution reactions, various coupling reactions, and the like.
 上記の各反応において、化合物が遊離の状態で得られる場合には、常法に従って塩に変換してもよく、また塩として得られる場合には、常法に従って遊離体またはその他の塩に変換することもできる。 In each of the above reactions, when the compound is obtained in a free state, it may be converted into a salt according to a conventional method. When it is obtained as a salt, it is converted into a free form or other salt according to a conventional method. You can also.
 これらの官能基の変換は、例えば、ラーロック(Richard C.Larock)らの、コンプリヘンシブ・オルガニック・トランスフォーメーション(Comprehensive Organic Transformations)、第2版、1999年10月刊、ウィリー ビーシーエッチ(Wiley-VCH)社、の成書に記載の方法等に準じて行う事ができる。 These functional group transformations are described in, for example, Comprehensive Organic Transformations, 2nd edition, published in October 1999, by Richard C. Larock, et al. VCH) can be carried out according to the method described in the book.
 また、本発明の式(I)で表される化合物の製造方法の各反応および原料化合物合成の各反応において、置換基として水酸基(アルコール性水酸基、フェノール性水酸基、複素環水酸基等)、アミノ基、カルボキシ基、チオール基等の反応性基がある場合には、各反応工程においてこれらの基を適宜保護し、適当な段階で当該保護基を除去することもできる。 Moreover, in each reaction of the manufacturing method of a compound represented by the formula (I) of this invention and each reaction of a raw material compound synthesis, a hydroxyl group (an alcoholic hydroxyl group, a phenolic hydroxyl group, a heterocyclic hydroxyl group, etc.), an amino group as a substituent When there are reactive groups such as carboxy group and thiol group, these groups can be appropriately protected in each reaction step, and the protecting group can be removed at an appropriate stage.
 当該水酸基(アルコール性水酸基、フェノール性水酸基、複素環水酸基等)の保護基としては、例えば、C1~6アルキル基(例えば、メチル基、エチル基、n-プロピル基、イソプロピル基、n-ブチル基、およびtert-ブチル基等);-C1~6アルキル-O-C1~6アルキル基(メトキシメチル基、メトキシエトキシメチル基等);テトラヒドロピラニル基;C7~20アラルキル基(例えば、ベンジル基、およびトリフェニルメチル基等);シリル基(例えば、トリメチルシリル基、トリエチルシリル基、t‐ブチルジメチルシリル基、およびt‐ブチルジフェニルシリル基等);-C(O)-C1~6アルキル基(例えば、アセチル基、エチルカルボニル基、およびピバロイル基等);-C(O)-C7~20アラルキル基(例えば、ベンジルカルボニル基等);-C(O)-C6~14アリール基(例えば、ベンゾイル基等);-C(O)-O-C1~6アルキル基(例えば、メトキシカルボニル基、エトキシカルボニル基、およびt‐ブトキシカルボニル基等);-C(O)-O-C7~20アラルキル基(例えば、ベンジルオキシカルボニル基等)等が用いられる。 Examples of the protective group for the hydroxyl group (alcoholic hydroxyl group, phenolic hydroxyl group, heterocyclic hydroxyl group, etc.) include, for example, C 1-6 alkyl groups (eg, methyl group, ethyl group, n-propyl group, isopropyl group, n-butyl group). group, and tert- butyl group); - C 1 ~ 6 alkyl -O-C 1 ~ 6 alkyl group (methoxymethyl group, methoxyethoxymethyl group, etc.); a tetrahydropyranyl group; C 7 ~ 20 aralkyl group (e.g. Silyl group (for example, trimethylsilyl group, triethylsilyl group, t-butyldimethylsilyl group, and t-butyldiphenylsilyl group); —C (O) —C 1- 6 alkyl group (e.g., acetyl group, ethylcarbonyl group, and pivaloyl group); - C (O) -C 7 ~ 20 aralkyl group (e.g., Ben Le carbonyl group); - C (O) -C 6 ~ 14 aryl group (e.g., benzoyl group); - C (O) -O -C 1 ~ 6 alkyl group (e.g., methoxycarbonyl group, ethoxycarbonyl group , and t- butoxycarbonyl group or the like); - C (O) -O -C 7 ~ 20 aralkyl group (e.g., a benzyl oxycarbonyl group) and the like.
 当該アミノ基(-NH2基)もしくはイミノ基(-NH-基)の保護基としては、例えば、-C(O)-C1~6アルキル基(例えば、アセチル基、エチルカルボニル基、およびピバロイル基等);-C(O)-O-C1~6アルキル基(例えば、メトキシカルボニル基、エトキシカルボニル基、およびt‐ブトキシカルボニル基等);-C(O)-O-C6~14アリール基(例えば、フェニルオキシカルボニル基);-C(O)-O-C7~20アラルキル基(例えば、フルオレニルメトキシカルボニル基、ベンジルオキシカルボニル基、パラメトキシベンジルオキシカルボニル基、およびパラニトロベンジルオキシカルボニル基等);-C(O)-O-C2~7アルケニル基(例えば、アリルオキシカルボニル基等);-C7~20アラルキル基(例えば、ベンジル基、およびトリフェニルメチル基等);-C(O)-C6~14アリール基(例えば、ベンゾイル基等);-C(O)-C7~20アラルキル基(例えば、ベンジルカルボニル基等);-SO2-C1~6アルキル基または-SO2-C6~14アリール基(例えば、メタンスルホニル基、p-トルエンスルホニル基、2,4-ジニトロベンゼンスルホニル基、およびベンゼンスルホニル基等);2-(トリメチルシリル)エトキシメチル基;フタロイル基;N,N-ジメチルアミノメチレン基等が用いられる。 The protecting group of the amino group (-NH 2 group) or an imino group (-NH- group), for example, -C (O) -C 1 ~ 6 alkyl group (e.g., acetyl group, ethylcarbonyl group, and pivaloyl group); - C (O) -O -C 1 ~ 6 alkyl group (e.g., methoxycarbonyl group, ethoxycarbonyl group, and t- butoxycarbonyl group or the like); - C (O) -O -C 6 ~ 14 an aryl group (e.g., phenyloxycarbonyl group); - C (O) -O -C 7 ~ 20 aralkyl group (e.g., fluorenyl methoxy carbonyl group, benzyloxycarbonyl group, para-methoxybenzyloxycarbonyl group, and para nitro benzyloxycarbonyl group, etc.); - C (O) -O -C 2 ~ 7 alkenyl group (e.g., an allyloxycarbonyl group, etc.); - C 7 ~ 20 aralkyl group (eg , Benzyl, and triphenylmethyl group and the like); - C (O) -C 6 ~ 14 aryl group (e.g., benzoyl group); - C (O) -C 7 ~ 20 aralkyl group (e.g., benzylcarbonyl group etc); - SO 2 -C 1 ~ 6 alkyl group or -SO 2 -C 6 ~ 14 aryl group (e.g., methanesulfonyl group, p- toluenesulfonyl group, 2,4-nitrobenzenesulfonyl group, and a benzenesulfonyl group Etc.); 2- (trimethylsilyl) ethoxymethyl group; phthaloyl group; N, N-dimethylaminomethylene group and the like are used.
 当該カルボキシ基の保護基としては、例えば、C1~6アルキル基(例えば、メチル基、エチル基、n-プロピル基、イソプロピル基、n-ブチル基、およびtert-ブチル基等);C2~7アルケニル基(ビニル基、およびアリル基等);C6~14アリール基(例えば、フェニル等);C7~20アラルキル基(例えば、ベンジル基、およびトリフェニルメチル基等);シリル基(例えば、トリメチルシリル基、トリエチルシリル基、t‐ブチルジメチルシリル基、およびt‐ブチルジフェニルシリル基等)等が用いられる。 The protecting group of the carboxyl group, for example, C 1 ~ 6 alkyl group (e.g., methyl group, ethyl group, n- propyl group, an isopropyl group, n- butyl group, and tert- butyl group); C 2 ~ 7 alkenyl group (vinyl group, and allyl group, etc.); C 6 ~ 14 aryl group (e.g., phenyl, etc.); C 7 ~ 20 aralkyl group (e.g., benzyl, and triphenylmethyl group); a silyl group (e.g. , Trimethylsilyl group, triethylsilyl group, t-butyldimethylsilyl group, t-butyldiphenylsilyl group, etc.).
 当該チオール基(-SH基)の保護基としては、例えば、C1~6アルキル基(例えば、メチル基、エチル基、n-プロピル基、イソプロピル基、n-ブチル基、およびtert-ブチル基等);C7~20アラルキル基(例えば、ベンジル基、およびトリフェニルメチル基等);-C(O)-C1~6アルキル基(例えば、アセチル基、エチルカルボニル基、およびピバロイル基等); -C(O)-C6~14アリール基(例えば、ベンゾイル等基)等が用いられる。 Examples of the protecting group for the thiol group (—SH group) include C 1-6 alkyl groups (eg, methyl group, ethyl group, n-propyl group, isopropyl group, n-butyl group, tert-butyl group, etc.) ); C 7 ~ 20 aralkyl group (e.g., benzyl, and triphenylmethyl group and the like); - C (O) -C 1 ~ 6 alkyl group (e.g., acetyl group, ethylcarbonyl group, and pivaloyl group); -C (O) -C 6 ~ 14 aryl group (e.g., benzoyl group) and the like.
 こうした保護基の導入・除去の方法は、保護される基あるいは保護基の種類により適宜行われるが、例えば、グリーン(Greene)らの『プロテクティブ・グループス・イン・オーガニック・シンセシス(Protective Groups in Organic Synthesis) 第4版、2007年、ジョン ウィリー アンド サンズ(John Wiley & Sons)』の成書に記載の方法により行うことができる。 Such protecting group introduction / removal method is appropriately carried out depending on the group to be protected or the type of protecting group. For example, Green, et al., Protective Groups in Organic Synthesis (Protective Groups in Organic). (Synthesis), 4th edition, 2007, John Wiley & Sons, can be performed by the method described in the book.
 保護基の脱保護法としては、例えば、-C(O)-C1~6アルキル基(例えば、アセチル基、エチルカルボニル基、およびピバロイル基等);-C(O)-O-C1~6アルキル基(例えば、メトキシカルボニル基、エトキシカルボニル基、およびt‐ブトキシカルボニル基等);-C(O)-C6~14アリール基(例えば、ベンゾイル基等)等のアシル型保護基では、例えば、水酸化リチウム、水酸化ナトリウムもしくは水酸化カリウムのような水酸化アルカリ金属等の適当な塩基を用いることにより加水分解し脱保護できる。 The deprotection methods of protecting groups, e.g., -C (O) -C 1 ~ 6 alkyl group (e.g., acetyl group, ethylcarbonyl group, and pivaloyl group); - C (O) -O -C 1 ~ 6 alkyl group (e.g., methoxycarbonyl group, ethoxycarbonyl group, and t- butoxycarbonyl group or the like); - acyl type protecting groups such as C (O) -C 6 ~ 14 aryl group (e.g., benzoyl group, etc.), For example, it can be hydrolyzed and deprotected by using a suitable base such as an alkali metal hydroxide such as lithium hydroxide, sodium hydroxide or potassium hydroxide.
 -C1~6アルキル-O-C1~6アルキル基(メトキシメチル基、メトキシエトキシメチル基等);テトラヒドロピラニル基; -C(O)-O-C1~6アルキル基(例えば、メトキシカルボニル基、エトキシカルボニル基、およびt‐ブトキシカルボニル基等);-C(O)-O-C7~20アラルキル基(例えば、ベンジルオキシカルボニル基、およびパラメトキシベンジルオキシカルボニル基等);シリル基(例えば、トリメチルシリル基、トリエチルシリル基、t‐ブチルジメチルシリル基、およびt‐ブチルジフェニルシリル基等)等の保護基は、例えば、酢酸、塩酸、臭化水素酸、硫酸、リン酸、トリフルオロ酢酸、トリフルオロメタンスルホン酸等の適当な酸、あるいはこれらを組み合わせた酸を用いることにより脱保護できる。 -C 1 ~ 6 alkyl -O-C 1 ~ 6 alkyl group (methoxymethyl group, methoxyethoxymethyl group, etc.); a tetrahydropyranyl group; -C (O) -O-C 1 ~ 6 alkyl group (e.g., methoxy group, an ethoxycarbonyl group, and t- butoxycarbonyl group or the like); - C (O) -O -C 7 ~ 20 aralkyl group (e.g., benzyloxycarbonyl group, and para-methoxybenzyloxycarbonyl group, etc.); silyl groups Protecting groups such as trimethylsilyl group, triethylsilyl group, t-butyldimethylsilyl group, t-butyldiphenylsilyl group and the like are, for example, acetic acid, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, trifluoro Deprotection can be achieved by using an appropriate acid such as acetic acid or trifluoromethanesulfonic acid, or a combination of these acids.
 また、上記シリル基は、適当なフッ素イオン(F-)発生試薬、例えばフッ化テトラブチルアンモニウム、フッ化水素等の試薬を用いることでも脱保護できる。 The silyl group can also be deprotected by using a suitable fluorine ion (F ) generating reagent, for example, a reagent such as tetrabutylammonium fluoride or hydrogen fluoride.
 -C(O)-O-C7~20アラルキル基(例えば、ベンジルオキシカルボニル基、パラメトキシベンジルオキシカルボニル基、およびパラニトロベンジルオキシカルボニル基等);C7~20アラルキル基(例えば、ベンジル基等)等の保護基は、例えば、パラジウム-炭素(Pd-C)触媒を用いる加水素分解により脱保護できる。 -C (O) -O-C 7 ~ 20 aralkyl group (e.g., benzyloxycarbonyl group, para-methoxybenzyloxycarbonyl group, and para-nitrobenzyloxycarbonyl group and the like); C 7 ~ 20 aralkyl group (e.g., benzyl group And the like can be deprotected by hydrogenolysis using, for example, a palladium-carbon (Pd—C) catalyst.
 また上記C7~20アラルキル基(例えば、ベンジル基等)の保護基は、例えば、液体アンモニア中、金属ナトリウムを用いるバーチ還元によっても脱保護できる。 The above C 7 ~ 20 aralkyl group (e.g., benzyl group) protecting groups are, for example, in liquid ammonia, can be deprotected by Birch reduction using metal sodium.
 トリフェニルメチル基は、適当な酸、例えば、酢酸、塩酸、臭化水素酸、硫酸、リン酸、トリフルオロ酢酸、トリフルオロメタンスルホン酸等の酸あるいはこれらを組み合わせた酸を用いることにより脱保護できる。また、液体アンモニア中、金属ナトリウムもしくは金属リチウムを用いるバーチ還元や、パラジウム炭素触媒を用いる加水素分解によっても脱保護できる。 The triphenylmethyl group can be deprotected by using an appropriate acid, for example, an acid such as acetic acid, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, trifluoroacetic acid, trifluoromethanesulfonic acid or a combination thereof. . It can also be deprotected by liquid Birch reduction using metallic sodium or metallic lithium or hydrogenolysis using a palladium carbon catalyst.
 -SO2-C1~6アルキル基または-SO2-C6~14アリール基(例えば、メタンスルホニル基、p-トルエンスルホニル基、2,4-ジニトロベンゼンスルホニル基、およびベンゼンスルホニル基等)は、例えば、低温下にてNa/アンスラセン、Na/ナフタレンを用いる一電子還元、もしくは、液体アンモニア中、金属ナトリウムもしくは金属リチウムを用いるバーチ還元等を用いることにより脱保護できる。また、-SO2-C6~14アリール基の中でも、2-ニトロベンゼンスルホニル基および2,4-ジニトロベンゼンスルホニル基は、例えば、炭酸カリウムもしくはトリエチルアミン等の塩基性試薬存在下、チオールを反応させる、穏和な条件にて脱保護できる。 -SO 2 -C 1 ~ 6 alkyl group or -SO 2 -C 6 ~ 14 aryl group (e.g., methanesulfonyl group, p- toluenesulfonyl group, 2,4-nitrobenzenesulfonyl group, and a benzenesulfonyl group, etc.) For example, deprotection can be achieved by using one-electron reduction using Na / anthracene or Na / naphthalene at low temperature or Birch reduction using metallic sodium or metallic lithium in liquid ammonia. Among the -SO 2 -C 6 ~ 14 aryl group, 2-nitrobenzenesulfonyl group and a 2,4-nitrobenzenesulfonyl group, for example, the presence of a basic reagent such as potassium carbonate or triethylamine, reacting the thiol, Can be deprotected under mild conditions.
 ここに示した、保護基の脱保護法は1例にしかなく、例えば、グリーン(Greene)らの『プロテクティブ・グループス・イン・オーガニック・シンセシス(Protective Groups in Organic Synthesis) 第4版、2007年、ジョン ウィリー アンド サンズ(John Wiley & Sons)』の成書に記載の方法もしくは公知に発表されている各種論文を適用することで、脱保護が可能である。 The protecting group deprotection method shown here is only one example. For example, Green et al., “Protective Groups in Organic Synthesis, 4th Edition, 2007. Deprotection is possible by applying the method described in the book of John Wiley & Sons or various published papers.
 下記に述べる製造方法中の反応条件については、特に断らない限り、以下の如きとする。反応温度は、-78℃から溶媒が還流する温度の範囲であり、特に温度が記載されていない場合は、室温(0~35℃)であり、反応時間は、反応が十分進行する時間である。 The reaction conditions in the production method described below are as follows unless otherwise specified. The reaction temperature is in the range from −78 ° C. to the temperature at which the solvent is refluxed. When the temperature is not described, it is room temperature (0 to 35 ° C.), and the reaction time is the time for which the reaction proceeds sufficiently. .
 また、製造方法中の各工程は、無溶媒、あるいは反応前に原料化合物を適当な反応に関与しない溶媒に溶解又は懸濁して行うことができる。反応に関与しない溶媒としては、具体的には、水;シクロヘキサン、ヘキサン等の飽和炭化水素系溶媒;ベンゼン、クロロベンゼン、トルエン、キシレン等の芳香族炭化水素溶媒;メタノール、エタノール、1-プロパノール、2-プロパノール、tert-ブチルアルコール、2-メトキシエタノール等のアルコール系溶媒;N,N-ジメチルホルムアミド、N,N-ジメチルアセトアミド、ヘキサメチルホスホリックトリアミド、1,3‐ジメチル‐2‐イミダゾリジノン等の極性アミド系溶媒:ジメチルスルホキシド等のスルホキシド系溶媒;アセトニトリル、プロピオニトリル等のニトリル系溶媒;ジエチルエーテル、ジイソプロピルエーテル、ジフェニルエーテル、テトラヒドロフラン、1,4-ジオキサン、1,2-ジメトキシエタン等のエーテル系溶媒;酢酸メチル、酢酸エチル、酢酸ブチル等のエステル系溶媒;アセトン、メチルエチルケトン等のケトン系溶媒;ジクロロメタン、クロロホルム、四塩化炭素、1,2-ジクロロエタン等のハロゲン化炭化水素系溶媒;トリエチルアミン、N,N-ジイソプロピルエチルアミン、ピリジン、ルチジン等の塩基性溶媒;無水酢酸等の酸無水物;ギ酸、酢酸、プロピオン酸、トリフルオロ酢酸、メタンスルホン酸等の有機酸;塩酸、硫酸等の無機酸;であるが、一種の溶媒を単独で用いてもよく、または反応条件により適宜選択し二種以上の溶媒を適宜の割合で混合して用いてもよい。 In addition, each step in the production method can be performed without solvent or by dissolving or suspending the raw material compound in a solvent not involved in an appropriate reaction before the reaction. Specific examples of solvents that do not participate in the reaction include water; saturated hydrocarbon solvents such as cyclohexane and hexane; aromatic hydrocarbon solvents such as benzene, chlorobenzene, toluene, and xylene; methanol, ethanol, 1-propanol, 2 Alcohol solvents such as propanol, tert-butyl alcohol, 2-methoxyethanol; N, N-dimethylformamide, N, N-dimethylacetamide, hexamethylphosphoric triamide, 1,3-dimethyl-2-imidazolidinone Polar amide solvents such as: sulfoxide solvents such as dimethyl sulfoxide; nitrile solvents such as acetonitrile and propionitrile; diethyl ether, diisopropyl ether, diphenyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxy Ether solvents such as tan; ester solvents such as methyl acetate, ethyl acetate and butyl acetate; ketone solvents such as acetone and methyl ethyl ketone; halogenated hydrocarbons such as dichloromethane, chloroform, carbon tetrachloride and 1,2-dichloroethane Solvent; basic solvent such as triethylamine, N, N-diisopropylethylamine, pyridine, lutidine; acid anhydride such as acetic anhydride; organic acid such as formic acid, acetic acid, propionic acid, trifluoroacetic acid, methanesulfonic acid; hydrochloric acid, sulfuric acid However, one kind of solvent may be used alone, or two or more kinds of solvents may be mixed and used at an appropriate ratio depending on the reaction conditions.
 本発明の化合物の製造方法中において用いられる塩基(又は脱酸剤)として、具体的には、水酸化リチウム、水酸化ナトリウム、水酸化カリウム、水酸化マグネシウム、炭酸リチウム、炭酸ナトリウム、炭酸カリウム、炭酸セシウム、炭酸カルシウム、炭酸水素ナトリウム等の無機塩基;トリエチルアミン、N,N-ジイソプロピルエチルアミン(DIPEA)、トリブチルアミン、シクロヘキシルジメチルアミン、ピリジン、ルチジン、4-ジメチルアミノピリジン(DMAP)、N,N-ジメチルアニリン、N-メチルピペリジン、N-メチルピロリジン、N-メチルモルホリン、1,5-ジアザビシクロ[4.3.0]-5-ノネン、1,4-ジアザビシクロ[2.2.2]オクタン、1,8-ジアザビシクロ[5.4.0]-7-ウンデセン、イミダゾール等の有機塩基;ナトリウムメトキシド、ナトリウムエトキシド、カリウムtert-ブトキシド、ナトリウムtert-ブトキシド等金属アルコキシルド類;水素化ナトリウム、水素化カリウム等のアルカリ金属水素化物;ナトリウムアミド、リチウムジイソプロピルアミド、リチウムヘキサメチルジシラジド等の金属アミド;メチルリチウム、n-ブチルリチウム、sec-ブチルリチウム、tert-ブチルリチウム等の有機リチウム試薬;が挙げられる。また、本発明の化合物の製造方法において用いられる酸、又は酸触媒として、具体的には、塩酸、硫酸、硝酸、臭化水素酸、リン酸等の無機酸;酢酸、トリフルオロ酢酸、シュウ酸、フタル酸、フマル酸、酒石酸、マレイン酸、クエン酸、コハク酸、メタンスルホン酸、p-トルエンスルホン酸、10-カンファースルホン酸等の有機酸;三フッ化ホウ素エーテル錯体、ヨウ化亜鉛、無水塩化アルミニウム、無水塩化亜鉛、無水塩化鉄等のルイス酸;が挙げられる。ただし、上記に記載したものに必ずしも限定されるわけではない。 Specific examples of the base (or deoxidizer) used in the method for producing the compound of the present invention include lithium hydroxide, sodium hydroxide, potassium hydroxide, magnesium hydroxide, lithium carbonate, sodium carbonate, potassium carbonate, Inorganic bases such as cesium carbonate, calcium carbonate, sodium bicarbonate; triethylamine, N, N-diisopropylethylamine (DIPEA), tributylamine, cyclohexyldimethylamine, pyridine, lutidine, 4-dimethylaminopyridine (DMAP), N, N- Dimethylaniline, N-methylpiperidine, N-methylpyrrolidine, N-methylmorpholine, 1,5-diazabicyclo [4.3.0] -5-nonene, 1,4-diazabicyclo [2.2.2] octane, 1 , 8-diazabicyclo [5.4.0] -7-undece Organic bases such as imidazole; sodium methoxide, sodium ethoxide, potassium tert-butoxide, sodium tert-butoxide and other metal alkoxides; sodium hydride, potassium hydride and other alkali metal hydrides; sodium amide, lithium diisopropylamide And metal amides such as lithium hexamethyldisilazide; organolithium reagents such as methyllithium, n-butyllithium, sec-butyllithium, and tert-butyllithium. In addition, as the acid or acid catalyst used in the method for producing the compound of the present invention, specifically, inorganic acids such as hydrochloric acid, sulfuric acid, nitric acid, hydrobromic acid, phosphoric acid; acetic acid, trifluoroacetic acid, oxalic acid , Organic acids such as phthalic acid, fumaric acid, tartaric acid, maleic acid, citric acid, succinic acid, methanesulfonic acid, p-toluenesulfonic acid, 10-camphorsulfonic acid; boron trifluoride ether complex, zinc iodide, anhydrous Lewis acids such as aluminum chloride, anhydrous zinc chloride, and anhydrous iron chloride. However, it is not necessarily limited to those described above.
 式(I)の塩は、それ自体公知の手段に従い、例えば、式(I)が塩基性化合物である場合には塩酸、臭化水素酸、硝酸、硫酸、リン酸等無機酸(鉱酸)またはギ酸、酢酸、トリフルオロ酢酸、フタル酸、フマル酸、シュウ酸、酒石酸、マレイン酸、クエン酸、コハク酸、リンゴ酸、メタンスルホン酸、ベンゼンスルホン酸、p-トルエンスルホン酸等有機酸を加えることによって、または式(I)が酸性化合物である場合にはアンモニア、トリメチルアミン、トリエチルアミン、ピリジン、ピコリン、2,6-ルチジン、エタノールアミン、ジエタノールアミン、トリエタノールアミン、シクロヘキシルアミン、ジシクロヘキシルアミン、N,N-ジイソプロピルエチルアミン、N,N'-ジベンジルエチレンジアミン、N,N‐ジアルキルアニリン等の有機塩基または炭酸リチウム、炭酸ナトリウム、炭酸カリウム、炭酸セシウム、水酸化リチウム、水酸化ナトリウム、水酸化カリウム、炭酸水素ナトリウム等無機塩基を加えることによって製造することができる。 The salt of the formula (I) is prepared according to a method known per se. For example, when the formula (I) is a basic compound, an inorganic acid (mineral acid) such as hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, etc. Or add organic acids such as formic acid, acetic acid, trifluoroacetic acid, phthalic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid Or when formula (I) is an acidic compound, ammonia, trimethylamine, triethylamine, pyridine, picoline, 2,6-lutidine, ethanolamine, diethanolamine, triethanolamine, cyclohexylamine, dicyclohexylamine, N, N -Diisopropylethylamine, N, N'-dibenzylethylenediamine, N, N-dialkyl It can be produced by adding an organic base such as ruaniline or an inorganic base such as lithium carbonate, sodium carbonate, potassium carbonate, cesium carbonate, lithium hydroxide, sodium hydroxide, potassium hydroxide, sodium bicarbonate.
 本発明の式(I)で表される化合物は、式(AM)で表されるアミン誘導体を出発原料として、製造方法A(P1=Hの場合:環A基の導入、続くP2基の脱保護、式(CA)で表されるカルボン酸誘導体の縮合反応)、もしくは製造方法B(P2=Hの場合:式(CA)で表されるカルボン酸誘導体の縮合反応、続くP1基の脱保護、環A基の導入)により得ることができる。
Figure JPOXMLDOC01-appb-C000025
The compound represented by the formula (I) of the present invention is prepared by using the amine derivative represented by the formula (AM) as a starting material and the production method A (when P 1 = H: introduction of the ring A group, followed by the P 2 group Deprotection of carboxylic acid derivative represented by formula (CA), or production method B (when P 2 = H: condensation reaction of carboxylic acid derivative represented by formula (CA), followed by P 1 Group deprotection, introduction of ring A group).
Figure JPOXMLDOC01-appb-C000025
以下に、式(I)、式(I-1)[式(I)で環B=フェニル(Ph)、または5~7員ヘテロアリール環;Q=C6~14アリール(Ar)の場合]、式(I-2)[式(I)で環A=3-置換-4-シアノ-2-ピリジンの場合]、および式(CA)[式(CA-3)、式(CA-4)、式(CA-5-1)、式(CA-5-2)、および式(CA-5-3)]で表される化合物の製造方法を示す。 The following formula (I), formula (I-1) [Formula (I) by ring B = phenyl (Ph), or 5-7 membered heteroaryl ring; in the case of Q = C 6 ~ 14 aryl (Ar)] And formula (I-2) [in the case of ring A = 3-substituted-4-cyano-2-pyridine in formula (I)], and formula (CA) [formula (CA-3), formula (CA-4) , Formula (CA-5-1), Formula (CA-5-2), and Formula (CA-5-3)].
[製造方法A]
Figure JPOXMLDOC01-appb-C000026
[Production Method A]
Figure JPOXMLDOC01-appb-C000026
<工程1>
[環A=5~7員ヘテロアリール基;X=ハロゲン原子の場合]
 式(AM-1)(式(AM)でP1=H)で表される化合物、および式(AR-1)で表される化合物(式(AM-1)および式(AR-1)で表される化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)を用いて、文献公知の方法、例えば、『テトラへドロン(Tetrahedron)、66(13)、2398-2403頁、2010年』等に記載された方法に準じて、無溶媒で反応温度120~150度にて反応を行い、式(AM-3)の化合物を製造することができる。
 また、式(AM-1)(式(AM)でP1=H)で表される化合物、および式(AR-1)で表される化合物を用いて、文献公知の方法、例えば、『ジャーナル オブ オルガニック ケミストリー (Journal of Organic Chemistry)、70(13)、5164-5173頁、2005年』等に記載された方法に準じて、ヨウ化銅(CuI)等のCu触媒、および(S)-プロリン、および炭酸カリウム、炭酸ナトリウム、炭酸セシウム、トリエチルアミン、N,N-ジイソプロピルエチルアミン、リン酸カリウム等塩基存在下、ジメチルスルホキシド、N,N-ジメチルホルムアミド、N,N-ジメチルアセトアミド、ジメトキシエタン、1,4-ジオキサン、テトラヒドロフラン、トルエン、キシレン、アセトニトリル等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(AM-3)で表される化合物を製造することができる。
<Step 1>
[Ring A = 5 to 7-membered heteroaryl group; X = halogen atom]
A compound represented by formula (AM-1) (P 1 = H in formula (AM)), and a compound represented by formula (AR-1) (in formula (AM-1) and formula (AR-1) The compound represented is a commercially available compound or a compound that can be produced from a commercially available compound by a production method known in the literature, and a method known in the literature, for example, “Tetrahedron, 66 (13), 2398”. The compound of formula (AM-3) can be produced by carrying out the reaction at a reaction temperature of 120 to 150 ° C. in the absence of a solvent in accordance with the method described in “-2403, 2010”.
Further, using a compound represented by the formula (AM-1) (P 1 = H in the formula (AM)) and a compound represented by the formula (AR-1), a method known in the literature, for example, “Journal According to the method described in Journal of Organic Chemistry, 70 (13), pages 5164-5173, 2005, etc., and (S)- Proline and potassium carbonate, sodium carbonate, cesium carbonate, triethylamine, N, N-diisopropylethylamine, potassium phosphate and the like in the presence of dimethyl sulfoxide, N, N-dimethylformamide, N, N-dimethylacetamide, dimethoxyethane, 1 , 4-dioxane, tetrahydrofuran, toluene, xylene, acetonito The compound represented by the formula (AM-3) can be produced by performing the reaction at a temperature at which the solvent is refluxed from 0 ° C. using a solvent such as ril, which does not participate in the reaction, or a mixed solvent thereof.
 更に、式(AM-1)(式(AM)でP1=H)で表される化合物、および式(AR-1)で表される化合物を用いて、文献公知の方法、例えば、『国際公開2012/076430号パンフレット』などに記載された方法に準じて、トリス(ジベンジリデンアセトン)ジパラジウム(Pd2(dba)3)等のPd触媒、および4,5’-ビス(ジフェニルホスフィノ)-9,9’-ジメチルキサンテン(XANTPHOS)等のホスフィン系試薬、およびトリエチルアミン、N,N-ジイソプロピルエチルアミン、リン酸カリウム、炭酸カリウム、炭酸セシウム等の有機または無機塩基存在下、トルエン、キシレン、N,N-ジメチルホルムアミド、N,N-ジメチルアセトアミド、ジメトキシエタン、アセトニトリル(アセトニトリル/水)、ジオキサン(ジオキサン/水)、テトラヒドロフラン(テトラヒドロフラン/水)等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(AM-3)で表される化合物を製造することができる。 Further, using a compound represented by the formula (AM-1) (P 1 = H in the formula (AM)) and a compound represented by the formula (AR-1), a method known in the literature, for example, “International In accordance with the method described in “Publication 2012/076430 pamphlet” and the like, a Pd catalyst such as tris (dibenzylideneacetone) dipalladium (Pd 2 (dba) 3 ), and 4,5′-bis (diphenylphosphino) In the presence of phosphine reagents such as -9,9'-dimethylxanthene (XANTPHOS) and organic or inorganic bases such as triethylamine, N, N-diisopropylethylamine, potassium phosphate, potassium carbonate, cesium carbonate, toluene, xylene, N , N-dimethylformamide, N, N-dimethylacetamide, dimethoxyethane, acetonitrile (acetonitrile / ), Dioxane (dioxane / water), tetrahydrofuran (tetrahydrofuran / water), or a solvent not involved in the reaction, or a mixed solvent thereof, and the reaction is carried out at a temperature at which the solvent is refluxed from 0 ° C. ) Can be produced.
[環A=5~7員ヘテロアリール基;X=OTfの場合]
 式(AM-1)(式(AM)でP1=H)で表される化合物、および式(AR-1)で表される化合物を用いて、文献公知の方法、例えば、『シンレット(Synlett)、(12)、1400-1402、1997年』等に記載された方法に準じて、ジクロロメタン、クロロホルム等のハロゲン系溶媒、ジエチルエーテル、テトラヒドロフラン、1,2-ジメトキシエタン、1,4-ジオキサン等のエーテル系溶媒、ベンゼン、トルエン等の芳香族炭化水素系溶媒、アセトニトリル、N,N-ジメチルホルムアミド、ジメチルスルホキシド等の極性溶媒など反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(AM-3)で表される化合物を製造することができる。
[Ring A = 5 to 7-membered heteroaryl group; when X = OTf]
Using a compound represented by the formula (AM-1) (P 1 = H in the formula (AM)) and a compound represented by the formula (AR-1), methods known in the literature, for example, “Synlett ), (12), 1400-1402, 1997 ”, etc., halogen solvents such as dichloromethane and chloroform, diethyl ether, tetrahydrofuran, 1,2-dimethoxyethane, 1,4-dioxane and the like 0 ° C. using a solvent that does not participate in the reaction, such as an ether solvent of the above, an aromatic hydrocarbon solvent such as benzene or toluene, a polar solvent such as acetonitrile, N, N-dimethylformamide, or dimethyl sulfoxide, or a mixed solvent thereof. The compound represented by the formula (AM-3) can be produced by reacting at a temperature at which the solvent is refluxed.
[環A=C6~14アリール基;X=ハロゲン原子の場合]
式(AM-1)(式(AM)でP1=H)で表される化合物、および式(AR-1)で表される化合物を用いて、文献公知の方法、例えば、『国際公開2009/059112号パンフレット』などに記載された方法に準じて、トリス(ジベンジリデンアセトン)ジパラジウム(Pd2(dba)3)、酢酸パラジウム(Pd(OAc)2)、クロロ(2-ジシクロヘキシルホスフィノ-2‘,6’-ジイソプロポキシ-1,1‘-ビフェニル)[2-(2’-アミノ-1,1‘-ビフェニル)]パラジウム(II)(RuPhos G2)等のPd触媒、および2-ジシクロヘキシルフォスフィノ-2’,4’,6’-トリイソプロピルビフェニル(X-phos)、4,5’-ビス(ジフェニルホスフィノ)-9,9’-ジメチルキサンテン(XANTPHOS)、(Ph2P)2-ビナフチル、2’,2-ビス(ジフェニルフォスフィノ)-1’,1-ビナフチル(BINAP)、 2-ジシクロヘキシルホスフィノ-2‘,6’-ジイソプロポキシ-1,1‘-ビフェニル(RuPhos)、[1’,1-ビフェニル]-2-イル ビス(1’,1-ジメチルエチル)-フォスフィン(JohnPhos)等のホスフィン系試薬、および炭酸セシウム、炭酸カリウム、リン酸カリウム、tert-BuONa、トリエチルアミン、N,N-ジイソプロピルエチルアミン、等の有機または無機塩基存在下、トルエン、N,N-ジメチルホルムアミド(DMF)、N-メチルピロリドン(NMP)、N,N-ジメチルアセトアミド、アセトニトリル、アセトニトリル/水、ジオキサン、ジオキサン/水、テトラヒドロフラン(THF)、テトラヒドロフラン/水、等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、室温から溶媒が還流する温度で反応を行い、式(AM-3)で表される化合物を製造することができる。
[; When X = halogen atom ring A = C 6 ~ 14 aryl group]
Using a compound represented by the formula (AM-1) (P 1 = H in the formula (AM)) and a compound represented by the formula (AR-1), methods known in the literature, for example, “International Publication 2009” / 059112 pamphlet "or the like, tris (dibenzylideneacetone) dipalladium (Pd 2 (dba) 3 ), palladium acetate (Pd (OAc) 2 ), chloro (2-dicyclohexylphosphino- A Pd catalyst such as 2 ′, 6′-diisopropoxy-1,1′-biphenyl) [2- (2′-amino-1,1′-biphenyl)] palladium (II) (RuPhos G2), and 2- Dicyclohexylphosphino-2 ′, 4 ′, 6′-triisopropylbiphenyl (X-phos), 4,5′-bis (diphenylphosphino) -9,9′-dimethylxanthene (XANT) HOS), (Ph 2 P) 2-binaphthyl, 2 ', 2-bis (diphenylphosphino) -1', 1 binaphthyl (BINAP), 2-dicyclohexyl phosphino-2 ', 6'-diisopropoxy - Phosphine reagents such as 1,1′-biphenyl (RuPhos), [1 ′, 1-biphenyl] -2-ylbis (1 ′, 1-dimethylethyl) -phosphine (JohnPhos), and cesium carbonate, potassium carbonate, In the presence of an organic or inorganic base such as potassium phosphate, tert-BuONa, triethylamine, N, N-diisopropylethylamine, etc., toluene, N, N-dimethylformamide (DMF), N-methylpyrrolidone (NMP), N, N— Dimethylacetamide, acetonitrile, acetonitrile / water, dioxane, dioxane / water, tet Using a solvent that does not participate in the reaction, such as lahydrofuran (THF), tetrahydrofuran / water, or a mixed solvent thereof, the reaction is performed from room temperature to the temperature at which the solvent is refluxed, and the compound represented by the formula (AM-3) is obtained. Can be manufactured.
<工程2>
[製造方法A]<工程1>で得られる式(AM-3)で表される化合物を用い、文献公知の方法、例えば、『プロテクティブ・グループス・イン・オーガニック・シンセシス(Protective Groups in Organic Synthesis 4thEdition) 第4版、2007年、ジョン ウィリー アンド サンズ(John Wiley & Sons)、グリーン(Greene)ら』の成書に記載された方法に準じて、保護基P2を保護基の種類に応じた方法で反応させることにより、P2基が脱保護された、式(AM-4)で表される化合物を製造することができる。
<Step 2>
[Production Method A] Using a compound represented by the formula (AM-3) obtained in <Step 1>, a method known in the literature, for example, “Protective Groups in Organic Synthesis” 4ThEdition) 4th edition, 2007, John Wiley & Sons (John Wiley & Sons), in accordance with the method described in textbooks of green (Greene) et al ", in accordance with the type of the protecting group the protecting group P 2 By reacting by the method, a compound represented by the formula (AM-4) in which the P 2 group is deprotected can be produced.
<工程3>
[製造方法A]<工程2>で得られる式(AM-4)のアミン、および式(CA)のカルボン酸を用いて、文献公知の方法、例えば、『実験化学講座 第4版 22 有機合成IV 酸・アミノ酸・ペプチド、191-309頁、1992年、丸善』等に記載された方法に準じて、1,3-ジシクロヘキシルカルボジイミド(DCC)、1-エチル-3-(3’-ジメチルアミノプロピル)カルボジイミド塩酸塩(EDCI)、1-ヒドロキシベンゾトリアゾール(HOBT)、ベンゾトリアゾール-1-イロキシトリス(ジメチルアミノ)ホスホニウムヘキサフルオロホスフェイト(BOP試薬)、ビス(2-オキソ-3-オキサゾリジニル)ホスフィニッククロリド(BOP-Cl)、2-クロロ-1,3-ジメチルイミダゾリニウムヘキサフルオロホスフェイト(CIP)、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(DMTMM)、ポリリン酸(PPA)、2-(1H-7-アザベンゾトリアゾール-1-イル)-1,1,3,3-テトラメチルウロニウム ヘキサフルオロホスフェート メタンアミニウム(HATU)、(1-シアノ-2-エトキシ-2-オキソエチリデンアミノオキシ)ジメチルアミノ-モルホリノ-カルベニウムヘキサフルオロリン酸塩(COMU)、プロピルホスホン酸無水物(T3P)等の縮合剤の存在下、ジクロロメタン、クロロホルム、ジエチルエーテル、テトラヒドロフラン、トルエン、ベンゼン、N,N-ジメチルホルムアミド、メタノール、エタノール、2-プロパノール等の反応に関与しない溶媒中、トリエチルアミン、N,N-ジイソプロピルエチルアミン、ピリジン等の塩基の存在下または非存在下、0℃から溶媒が還流する温度で反応させることにより、式(I)で表される化合物を製造することができる。
<Step 3>
[Production Method A] Using the amine of formula (AM-4) obtained in <Step 2> and the carboxylic acid of formula (CA), a method known in the literature, for example, “Experimental Chemistry Course 4th Edition 22 Organic Synthesis” In accordance with the method described in IV Acid / Amino Acid / Peptide, 191-309, 1992, Maruzen, etc., 1,3-dicyclohexylcarbodiimide (DCC), 1-ethyl-3- (3′-dimethylaminopropyl) ) Carbodiimide hydrochloride (EDCI), 1-hydroxybenzotriazole (HOBT), benzotriazole-1-iroxytris (dimethylamino) phosphonium hexafluorophosphate (BOP reagent), bis (2-oxo-3-oxazolidinyl) phosphinic Chloride (BOP-Cl), 2-chloro-1,3-dimethylimidazolinium hexaf Olophosphate (CIP), 4- (4,6-dimethoxy-1,3,5-triazin-2-yl) -4-methylmorpholinium chloride (DMTMM), polyphosphoric acid (PPA), 2- (1H -7-azabenzotriazol-1-yl) -1,1,3,3-tetramethyluronium hexafluorophosphate methanaminium (HATU), (1-cyano-2-ethoxy-2-oxoethylideneaminooxy) Dichloromethane, chloroform, diethyl ether, tetrahydrofuran, toluene, benzene, N, N-dimethyl in the presence of condensing agents such as dimethylamino-morpholino-carbenium hexafluorophosphate (COMU), propylphosphonic anhydride (T3P) Solvents that do not participate in the reaction, such as formamide, methanol, ethanol, 2-propanol , Triethylamine, N, N-diisopropylethylamine, pyridine and the like in the presence or absence of a reaction at a temperature at which the solvent is refluxed from 0 ° C. to produce the compound represented by formula (I). it can.
 また、式(CA)のカルボン酸を、文献公知の方法、例えば『ジャーナル・オブ・ザ・アメリカン・ケミカル・ソサエティ(Journal of the American Chemical Society)、109(24)、p7488-7494、1987年』等に記載された方法に準じて、トリエチルアミン、N,N-ジイソプロピルエチルアミン、N,N-ジメチルアミノピリジン等の塩基の存在もしくは非存在下、塩化チオニル、塩化オキサリル、塩化ホスホリル、塩化スルフリル、三塩化リン、五塩化リン、三臭化リン等のハロゲン化剤と、ジオキサン、テトラヒドロフラン、ベンゼン、トルエン、ジクロロメタン、1,2-ジクロロエタン、クロロホルム等の反応に不活性な溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い得られる酸ハライド、および[製造方法A]<工程2>で得られる式(AM-4)のアミンを用い、例えば『実験化学講座 第4版 22 有機合成IV酸・アミノ酸・ペプチド、144-146頁、1992年、丸善』等に記載された方法に準じて、トリエチルアミン、N,N-ジソプロピルエチルアミン、ピリジン、4-ジメチルアミノピリジン等の塩基の存在下、ジクロロメタン、クロロホルム、1,2-ジクロロエタン、ジエチルエーテル、テトラドロフラン、1,4-ジオキサン、トルエン、ベンゼン、N,N-ジメチルホルムアミド等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応させることにより、式(I)の化合物を同様に製造することができる。 Further, the carboxylic acid of the formula (CA) is converted into a method known in the literature, for example, “Journal of the American Chemical Society, 109 (24), p7488-7494, 1987”. In the presence or absence of a base such as triethylamine, N, N-diisopropylethylamine, N, N-dimethylaminopyridine, thionyl chloride, oxalyl chloride, phosphoryl chloride, sulfuryl chloride, trichloride. Use a halogenating agent such as phosphorus, phosphorus pentachloride or phosphorus tribromide and a solvent inert to the reaction such as dioxane, tetrahydrofuran, benzene, toluene, dichloromethane, 1,2-dichloroethane, chloroform, or a mixed solvent thereof. Using an acid halide obtained by reacting at a temperature at which the solvent refluxed from 0 ° C. and an amine of the formula (AM-4) obtained in [Production Method A] <Step 2>, for example, “Experimental Chemistry Course 4th Edition 22 Organic Synthetic IV Acids / Amino Acids / Peptides, 144-146, 1992, Maruzen ”etc., and bases such as triethylamine, N, N-disopropylethylamine, pyridine, 4-dimethylaminopyridine, etc. In the presence of dichloromethane, chloroform, 1,2-dichloroethane, diethyl ether, tetradrofuran, 1,4-dioxane, toluene, benzene, N, N-dimethylformamide and the like, or a mixed solvent thereof To react the compound of formula (I) in a similar manner by reacting from 0 ° C. to the temperature at which the solvent is refluxed. It is possible to elephants.
[製造方法B]
Figure JPOXMLDOC01-appb-C000027
[Production Method B]
Figure JPOXMLDOC01-appb-C000027
<工程1>
式(AM-2)(式(AM)でP2=H)の化合物、および式(CA)の化合物(式(AM-2)、および式(CA)の化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)を用いて、[製造方法A]<工程3>に準じる反応を行い、式(AM-5)の化合物を製造することができる。
<Step 1>
Compound of formula (AM-2) (P 2 = H in formula (AM)) and compound of formula (CA) (formula (AM-2) and compound of formula (CA) are commercially available compounds or commercially available compounds The compound of formula (AM-5) can be produced by carrying out a reaction according to [Production Method A] <Step 3>.
<工程2>
[製造方法B]<工程1>で得られる式(AM-5)の化合物を用い、文献公知の方法、例えば、『プロテクティブ・グループス・イン・オーガニック・シンセシス(Protective Groups in Organic Synthesis 4thEdition) 第4版、2007年、ジョン ウィリー アンド サンズ(John Wiley & Sons)、グリーン(Greene)ら』の成書に記載された方法に準じて、保護基P1を保護基の種類に応じた方法で反応させることにより、P1基が脱保護された、式(AM-6)で表される化合物を製造することができる。
<Step 2>
[Production Method B] Using a compound of formula (AM-5) obtained in <Step 1>, a method known in the literature, for example, “Protective Groups in Organic Synthesis 4th Edition” 4th edition, 2007, according to the method described in the book of John Wiley & Sons, Greene et al., The protecting group P 1 is reacted by a method according to the type of the protecting group. Thus, a compound represented by the formula (AM-6) in which the P 1 group is deprotected can be produced.
<工程3>
[製造方法B]<工程2>で得られる式(AM-6)の化合物、および式(AR-1)の化合物(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)を用いて、[製造方法A]<工程1>に準じる反応を行い、式(I)の化合物を製造することができる。
<Step 3>
[Production Method B] The compound of formula (AM-6) obtained in <Step 2> and the compound of formula (AR-1) (this compound is a commercially available compound or can be produced from a commercially available compound by a method known in the literature) The compound of formula (I) can be produced by carrying out a reaction according to [Production Method A] <Step 1>.
[製造方法C]式(I-1)[式(I)で環B=Ph、または5~7員ヘテロアリール環(但し、当該5~7員ヘテロアリール環は、ジアザビシクロ[2.2.2]オクタン環と環Bのアミド結合の隣接位(o-位)に窒素原子を有する様な、1-置換-1H-ピラゾール-5-イル基等の環を除く);Q=AR(C6~14アリール基もしくは5~7員ヘテロアリール基)の場合]の製造方法
Figure JPOXMLDOC01-appb-C000028
[製造方法C]
[Production Method C] Formula (I-1) [In formula (I), ring B = Ph, or a 5- to 7-membered heteroaryl ring (provided that the 5- to 7-membered heteroaryl ring is diazabicyclo [2.2.2] ] Except for a ring such as a 1-substituted-1H-pyrazol-5-yl group that has a nitrogen atom adjacent to the amide bond of the octane ring and ring B (o-position); Q = AR (C 6 To 14 aryl group or 5- to 7-membered heteroaryl group)]
Figure JPOXMLDOC01-appb-C000028
[Production Method C]
<工程1>
[製造方法A]<工程2>で得られる式(AM-4)の化合物、および式(CA-1)(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)の化合物を用いて、[製造方法A]<工程3>に準じる反応を行い、式(IM-1)の化合物を製造することができる。
<Step 1>
[Production Method A] Compound of formula (AM-4) obtained in <Step 2> and formula (CA-1) The compound of formula (IM-1) can be produced by carrying out a reaction according to [Production method A] <Step 3>.
<工程2>
[製造方法C]<工程1>で得られる式(IM-1)の化合物、および式(RG-1)のハロゲン化C6~14アリール化合物(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)を用い、文献公知の方法、例えば 『実験化学講座 第5版 18 有機化合物の合成 VI -金属を用いる有機合成-、327‐352頁、2004年、丸善』、および『ジャーナル・オブ・メディシナル・ケミストリー(Journal of Medicinal Chemistry)、48(20)、p6326‐6339、2005年』に記載された方法に準じて、酢酸パラジウム(II)(Pd(OAc)2)、テトラキストリフェニルホスフィンパラジウム(Pd(PPh34)、トリス(ジベンジリデンアセトン)ジパラジウム(Pd2(dba)3)、ビス(ジベンジリデンアセトン)パラジウム(Pd(dba)2)、[1,1’-ビス(ジフェニルホスフィノ)フェロセン]ジクロロパラジウム(II)(PdCl2(dppf))等のパラジウム触媒、トリフェニルホスフィン、トリス(tert-ブチル)ホスフィン、トリス(o-トリル)ホスフィン、2-ジシクロヘキシルホスフィノ-2’,6’-ジメトキシビフェニル、2-ジシクロヘキシルホスフィノ-2’,4’,6’-トリイソプロピルビフェニル等のホスフィン系試薬、およびトリエチルアミン、N,N-ジイソプロピルエチルアミン、リン酸カリウム、炭酸カリウム、炭酸セシウム等の有機または無機塩基存在下、トルエン、キシレン、N,N-ジメチルホルムアミド、N,N-ジメチルアセトアミド、ジメトキシエタン、アセトニトリル(アセトニトリル/水)、ジオキサン(ジオキサン/水)、テトラヒドロフラン(テトラヒドロフラン/水)等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(I-1)で表される化合物を製造することができる。またはホスフィン系試薬の替わりにテトラメチルアンモニウムクロリド、テトラブチルアンモニウムクロリド等を用いて、同様の方法にて製造することができる。
<Step 2>
[Method C] Compounds of <Step 1> In the resulting formula (IM-1), and halogenated C 6 ~ 14 aryl compound of formula (RG-1) (the compound document from commercially available compounds, or commercially available compound The compound is a compound that can be produced by a known production method), and a method known in the literature, for example, “Experimental Chemistry Course 5th edition 18 Synthesis of organic compounds VI —Organic synthesis using metals—327-352, 2004, Maruzen And palladium (II) acetate (Pd (OAc) 2 ) according to the methods described in Journal of Medicinal Chemistry, 48 (20), p 6326-6339, 2005. ), Tetrakistriphenylphosphine palladium (Pd (PPh 3 ) 4 ), tris (dibenzylidene) Acetone) dipalladium (Pd 2 (dba) 3 ), bis (dibenzylideneacetone) palladium (Pd (dba) 2 ), [1,1′-bis (diphenylphosphino) ferrocene] dichloropalladium (II) (PdCl 2 Palladium catalysts such as (dppf)), triphenylphosphine, tris (tert-butyl) phosphine, tris (o-tolyl) phosphine, 2-dicyclohexylphosphino-2 ′, 6′-dimethoxybiphenyl, 2-dicyclohexylphosphino- In the presence of a phosphine reagent such as 2 ′, 4 ′, 6′-triisopropylbiphenyl, and an organic or inorganic base such as triethylamine, N, N-diisopropylethylamine, potassium phosphate, potassium carbonate, cesium carbonate, toluene, xylene, N, N-dimethylformamide, N Using a solvent that does not participate in the reaction such as N-dimethylacetamide, dimethoxyethane, acetonitrile (acetonitrile / water), dioxane (dioxane / water), tetrahydrofuran (tetrahydrofuran / water), or a mixed solvent thereof, The reaction can be carried out at a refluxing temperature to produce the compound represented by the formula (I-1). Or it can manufacture by the same method using tetramethylammonium chloride, tetrabutylammonium chloride, etc. instead of a phosphine-type reagent.
<工程3>
[製造方法A]<工程2>で得られる式(AM-4)の化合物、および式(CA-2)(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)の化合物を用いて、[製造方法A]<工程3>に準じる反応を行い、式(IM-2)の化合物を製造することができる。
<Step 3>
[Production Method A] Compound of formula (AM-4) obtained in <Step 2> and formula (CA-2) The compound of formula (IM-2) can be produced by carrying out a reaction according to [Production Method A] <Step 3>.
<工程4>
[式(RG-2)のボロン酸試薬を用いる場合]
[製造方法C]<工程3>で得られる式(IM-2)の化合物、および式(RG-2)(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)の化合物を用いて、[製造方法C]<工程2>に準じる反応を行い、式(I-1)の化合物を製造することができる。
[式(RG-3)のC6~14アリールスズ試薬、ヘテロアリールスズ試薬を用いる場合]
[製造方法C]<工程3>で得られる式(IM-2)の化合物、および式(RG-3)(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)の化合物を用いて、文献公知の方法、例えば、『シンレット(Synlett)、25(5)、653-656頁、2014年』、『ヨーロピアン ジャーナル オブ オルガニック ケミストリー(Europian Journal of Organic Chemistry)、2013(28)、6404-6419頁、2013年』、『ケミカルコミュニケーション(Chemical Communication)、48(71)、8907‐8909、2012年』、『バイオオルガニック アンド メディシナル ケミストリー(Bioorganic & Medicinal Chemistry)、21(8)、2370-2378頁、2013年』等に記載された方法に準じて、テトラキストリフェニルホスフィンパラジウム(Pd(PPh34)、ビス(トリフェニルホスフィン)パラジウム(II)ジクロライド(Pd2(PPh32)等のパラジウム触媒の存在下(テトラキストリフェニルホスフィンパラジウム(Pd(PPh34)を用いる場合、ヨウ化銅(CuI)等の銅試薬の存在下または非存在下)、フッ化セシウム、塩化リチウム等の塩の存在下または非存在下、トルエン、テトラヒドロフラン、N,N-ジメチルホルムアミド、1,4-ジオキサン等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(I-1)の化合物を製造することができる。
<Step 4>
[When using a boronic acid reagent of the formula (RG-2)]
[Production Method C] Compound of formula (IM-2) obtained in <Step 3> and formula (RG-2) (This compound is a commercially available compound or a compound that can be produced from a commercially available compound by a known production method in the literature. The compound of formula (I-1) can be produced by carrying out a reaction according to [Production Method C] <Step 2> using the compound of (A).
[C 6 ~ 14 aryl tin reagent of formula (RG-3), when using a heteroaryl tin reagent]
[Production Method C] Compound of formula (IM-2) obtained in <Step 3> and formula (RG-3) (This compound is a commercially available compound or a compound that can be produced from a commercially available compound by a known production method in the literature. A compound known in the literature, for example, “Synlett, 25 (5), 653-656, 2014”, “European Journal of Organic Chemistry,” 2013 (28), 6404-6419, 2013 ”,“ Chemical Communication, 48 (71), 8907-8909, 2012 ”,“ Bioorgana and Medicinal Chemistry (Bioorga). ic & Medicinal Chemistry), 21 ( 8), pp. 2370-2378, in accordance with the method described in 2013 ", etc., tetrakis triphenylphosphine palladium (Pd (PPh 3) 4), bis (triphenylphosphine) palladium (II) In the presence of a palladium catalyst such as dichloride (Pd 2 (PPh 3 ) 2 ) (when tetrakistriphenylphosphine palladium (Pd (PPh 3 ) 4 ) is used, the presence of a copper reagent such as copper iodide (CuI) Or in the absence or presence of a salt such as cesium fluoride or lithium chloride, or a solvent not involved in the reaction, such as toluene, tetrahydrofuran, N, N-dimethylformamide, 1,4-dioxane, or the like The reaction is carried out at a temperature from 0 ° C. to the reflux of the solvent. , It is possible to produce a compound of formula (I-1).
<工程5>
[式(IM-1)でB=ボロン酸エステルの場合]
[製造方法C]<工程3>で得られる式(IM-2)の化合物を用い、文献公知の方法、例えば、『ザ・ジャーナル・オブ・オーガニック・ケミストリー(The Journal of Organic Chemistry)、60、7508‐2665、1995年』に記載された方法に準じて、ビス(ピナコラート)ジボロン、ビス(ネオペンチルグリコラート)ジボロン等のジボロンエステル存在下、酢酸パラジウム(II)、テトラキストリフェニルホスフィンパラジウム、トリス(ジベンジリデンアセトン)ジパラジウム、[1,1’-ビス(ジフェニルホスフィノ)フェロセン]ジクロロパラジウム(II)などのパラジウム触媒の存在下、トリフェニルホスフィン、トリス(tert-ブチル)ホスフィン、トリス(o-トリル)ホスフィン、2-ジシクロヘキシルホスフィノ-2’,6’-ジメトキシビフェニル等のホスフィン系試薬、およびトリエチルアミン、N,N-ジイソプロピルエチルアミン、酢酸カリウム等の有機または無機塩基存在下または非存在下、またはホスフィン系試薬の替わりにテトラメチルアンモニウムクロリド、テトラブチルアンモニウムクロリド等存在下または非存在下、トルエン、N,N-ジメチルホルムアミド、ジメチルスルホキシド、1,4-ジオキサン等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(IM-1)のボロン酸エステルを製造することができる。
<Step 5>
[In the formula (IM-1) where B = boronic acid ester]
[Production Method C] Using a compound of formula (IM-2) obtained in <Step 3>, a method known in the literature, for example, “The Journal of Organic Chemistry”, 60, 7508-2665, 1995 ”, in the presence of diboron esters such as bis (pinacolato) diboron and bis (neopentylglycolate) diboron, palladium (II) acetate, tetrakistriphenylphosphine palladium, In the presence of a palladium catalyst such as tris (dibenzylideneacetone) dipalladium, [1,1′-bis (diphenylphosphino) ferrocene] dichloropalladium (II), triphenylphosphine, tris (tert-butyl) phosphine, tris ( o-tolyl In the presence or absence of phosphine, phosphine reagents such as 2-dicyclohexylphosphino-2 ′, 6′-dimethoxybiphenyl, and organic or inorganic bases such as triethylamine, N, N-diisopropylethylamine, potassium acetate, or phosphine Solvents not involved in the reaction, such as toluene, N, N-dimethylformamide, dimethyl sulfoxide, 1,4-dioxane, or a mixture thereof, in the presence or absence of tetramethylammonium chloride, tetrabutylammonium chloride, etc. instead of the reagent The reaction can be carried out using a solvent at a temperature at which the solvent is refluxed from 0 ° C. to produce a boronic ester of the formula (IM-1).
[式(IM-1)でB=ボロン酸の場合]
[製造方法C]<工程3>で得られる式(IM-2)の化合物を用い、文献公知の方法、例えば、『ケミッシェ・ベリヒテ(Chemische Berichte)、42、3090、1909年』等に記載された方法に準じて、トルエン、テトラヒドロフラン、1,4-ジオキサン等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、n-ブチルリチウム、sec-ブチルリチウム等のアルキルリチウム、イソプロピルマグネシウムクロリド等のグリニャール(Grignard)試薬、または金属マグネシウムの存在下、トリメチルボレート、トリイソプロピルボレート等のトリアルキルボレートを加え、-78℃から室温で反応を行った後、塩酸、硫酸等の酸を加え、0℃から溶媒が還流する温度で反応を行い、式(IM-1)のボロン酸を製造することができる。
[In the formula (IM-1) where B = boronic acid]
[Production Method C] Using a compound of formula (IM-2) obtained in <Step 3>, it is described in literature known methods, for example, “Chemiche Berichte, 42, 3090, 1909” and the like. In accordance with the above method, using a solvent such as toluene, tetrahydrofuran, 1,4-dioxane or the like, or a mixed solvent thereof, alkyllithium such as n-butyllithium and sec-butyllithium, isopropylmagnesium chloride, etc. In the presence of Grignard reagent or magnesium metal, trialkylborate such as trimethylborate and triisopropylborate was added, and the reaction was carried out at −78 ° C. at room temperature, and then acid such as hydrochloric acid and sulfuric acid was added. The reaction is carried out at a temperature at which the solvent refluxes from -1) boronic acids can be produced.
[式(IM-1)でB=トリフルオロボレート塩の場合]
前述の方法にて得られるボロン酸エステル、またはボロン酸を用い、文献公知の方法、例えば、『ケミカル・レビューズ(Chemical Reviews)、108、288‐325、2008年』等に記載された方法に準じて、ジフッ化水素カリウム存在下、メタノール、エタノール、水等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(IM-1)のトリフルオロボレート塩を製造することができる。
[In the formula (IM-1) where B = trifluoroborate salt]
Using the boronic acid ester obtained by the above-mentioned method or boronic acid, a method known in the literature, for example, the method described in “Chemical Reviews, 108, 288-325, 2008”, etc. Similarly, the reaction is carried out at a temperature at which the solvent is refluxed from 0 ° C. using a solvent that does not participate in the reaction such as methanol, ethanol, water or the like in the presence of potassium hydrogen difluoride, or a mixed solvent thereof. ) Trifluoroborate salt can be produced.
[式(IM-1)でB=ボロン酸 N-メチルイミノ二酢酸エステルの場合]
[製造方法C]<工程3>で得られる式(IM-2)で表される化合物を用い、文献公知の方法、例えば、『ジャーナル・オブ・オルガノメタリック ケミストリー(Journal of Organometallic Chemistry)、307(1)、1‐6、1986年』等に記載された方法に準じて、N-メチルイミノ二酢酸(MIDA)の存在下、ベンゼン、トルエン、キシレンまたはジメチルスルホキシド等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(IM-1)のボロン酸 N-メチルイミノ二酢酸(MIDA)エステルを製造することができる。
[In the formula (IM-1), B = boronic acid N-methyliminodiacetic acid ester]
[Production Method C] Using a compound represented by the formula (IM-2) obtained in <Step 3>, a method known in the literature, for example, “Journal of Organometallic Chemistry”, 307 ( 1) 1-6, 1986 ”, etc., in the presence of N-methyliminodiacetic acid (MIDA), solvents such as benzene, toluene, xylene or dimethyl sulfoxide, or the like The reaction is carried out at a temperature at which the solvent is refluxed from 0 ° C. using the mixed solvent of (1) to produce a boronic acid N-methyliminodiacetic acid (MIDA) ester of the formula (IM-1).
<工程6>
[製造方法A]<工程2>で得られる式(AM-4)の化合物、および式(CA-3)(後述の[製造方法E]を参照)の化合物を用いて、[製造方法A]<工程3>に準じる反応を行い、式(I-1)の化合物を製造することができる。
<Step 6>
[Production Method A] [Production Method A] using the compound of formula (AM-4) obtained in <Step 2> and the compound of formula (CA-3) (see [Production Method E] described later) A reaction according to <Step 3> can be carried out to produce a compound of formula (I-1).
[製造方法D]式(I-2)[式(I)で環A=3-置換-4-シアノ-2-ピリジンの場合]の製造方法
Figure JPOXMLDOC01-appb-C000029
[製造方法D]
[Production Method D] Production method of formula (I-2) [in the case of formula A, ring A = 3-substituted-4-cyano-2-pyridine]
Figure JPOXMLDOC01-appb-C000029
[Production Method D]
<工程1>
[製造方法B]<工程2>で得られる式(AM-6)の化合物、および式(AR-2)(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)の化合物を用いて、[製造方法B]<工程3>に準じる反応を行い、式(IM-3)の化合物を製造することができる。
<Step 1>
[Production Method B] Compound of formula (AM-6) obtained in <Step 2> and formula (AR-2) (This compound is a commercially available compound or a compound that can be produced from a commercially available compound by a known production method in the literature. A compound of formula (IM-3) can be produced by carrying out a reaction according to [Production method B] <Step 3>.
<工程2>
[Rγ=C3~8シクロアルキル基の場合]
[製造方法D]<工程1>で得られる式(IM-3)の化合物、および式(RG-4)(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)の亜鉛化合物を用いて、文献公知の方法、例えば、『国際公開2014/066196号パンフレット』等に記載された方法に準じて、テトラキストリフェニルホスフィンパラジウム(Pd(PPh34)、ビス[トリス(1,1-ジメチルエチル)ホスフィン]パラジウム等のPd触媒存在下、テトラヒドロフラン、2,2-ジメトキシエタン等の反応に関与しない溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(I-2)の化合物を製造することができる。
<Step 2>
[For R γ = C 3 ~ 8 cycloalkyl radical
[Production Method D] Compound of formula (IM-3) obtained in <Step 1> and formula (RG-4) (This compound is a commercially available compound or a compound that can be produced from a commercially available compound by a known production method in the literature. In accordance with a method known in the literature, for example, the method described in “International Publication No. 2014/066196 Pamphlet”, etc., tetrakistriphenylphosphine palladium (Pd (PPh 3 ) 4 ), bis In the presence of a Pd catalyst such as [tris (1,1-dimethylethyl) phosphine] palladium, the reaction is carried out at a temperature at which the solvent refluxes from 0 ° C. using a solvent that does not participate in the reaction, such as tetrahydrofuran and 2,2-dimethoxyethane. And the compound of formula (I-2) can be produced.
[Rγ=非非芳香族複素環基(アジリジン、アゼチジン、ピロリジン、ピペリジン、モルホリン等)、または-NRab基の場合]
[製造方法D]<工程1>で得られる式(IM-3)の化合物、および式(RG-5)もしくは式(RG-6)(式(RG-5)、および式(RG-6)の化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)のアミンを用いて、文献公知の方法、例えば、『国際公開2005/111003号パンフレット』、『ジャーナル オブ メディシナル ケミストリー(Journal of Medicinal Chemistry)、48(23)、7374-7388頁、2005年』等に記載された方法に準じて、ジイソプロピルエチルアミン、トリエチルアミン等の塩基の存在下もしくは非存在下、水、N,N-ジメチルホルムアミド、N-メチルピロリドン、アセトニトリル、テトラヒドロフラン等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(I-2)の化合物を製造することができる。
[In the case of R γ = non-aromatic heterocyclic group (aziridine, azetidine, pyrrolidine, piperidine, morpholine, etc.) or —NR a R b group]
[Production Method D] Compound of formula (IM-3) obtained in <Step 1>, and formula (RG-5) or formula (RG-6) (formula (RG-5) and formula (RG-6)) Is a commercially available compound or a compound that can be produced from a commercially available compound by a known production method), using known amines such as “International Publication No. 2005/111003 pamphlet”, “Journal of Medicinal”. According to the method described in Chemistry (Journal of Medicinal Chemistry), 48 (23), pages 7374-7388, 2005, etc., in the presence or absence of a base such as diisopropylethylamine, triethylamine, N-dimethylformamide, N-methylpyrrolidone, acetonitrile, tetrahydroph The solvent is not involved in the reaction, such as down, or by using a mixed solvent thereof, the reaction was carried out in the temperature at which the solvent refluxes from 0 ° C., can be prepared a compound of formula (I-2).
[Rγ=C1~6アルキルチオ基の場合]
[製造方法D]<工程1>で得られる式(IM-3)の化合物、および式(RG-7)(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)の化合物を用いて、文献公知の方法、例えば、『国際公開2011/049222号パンフレット』等に記載された方法に準じて、N,N-ジメチルホルムアミド、N-メチルピロリドン等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(I-2)の化合物を製造することができる。
[In the case of R γ = C 1-6 alkylthio group]
[Production Method D] Compound of formula (IM-3) obtained in <Step 1> and formula (RG-7) (This compound is a commercially available compound or a compound that can be produced from a commercially available compound by a known production method in the literature. And a compound known in the literature, for example, according to the method described in “International Publication No. 2011/049222 pamphlet” or the like, is involved in the reaction of N, N-dimethylformamide, N-methylpyrrolidone, etc. The compound of formula (I-2) can be produced by carrying out the reaction at a temperature at which the solvent is refluxed from 0 ° C. using a solvent that is not used or a mixed solvent thereof.
[Rγ=C1~6アルコキシ基の場合]
[製造方法D]<工程1>で得られる式(IM-3)の化合物、および式(RG-8)(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)の化合物を用いて、『ジャーナル オブ メディシナル ケミストリー(Journal of Medicinal Chemistry)、55(22)、10118-10129頁、2012年』等に記載された方法に準じて、N,N-ジメチルホルムアミド、N-メチルピロリドン等の反応に関与しない溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(I-2)の化合物を製造することができる。
[In the case of R γ = C 1-6 alkoxy group]
[Production Method D] Compound of formula (IM-3) obtained in <Step 1> and formula (RG-8) (This compound is a commercially available compound or a compound that can be produced from a commercially available compound by a known production method in the literature. N, N-dimethylformamide, according to the method described in “Journal of Medicinal Chemistry, 55 (22), pages 10118-10129, 2012” and the like. The compound of formula (I-2) can be produced by performing the reaction at a temperature at which the solvent is refluxed from 0 ° C. using a solvent that does not participate in the reaction, such as N-methylpyrrolidone.
なお、式(IM-3)でX=Clの場合の式(IM-3-1)は、後述する[製造方法D]<工程3>から[製造方法D]<工程5>の方法でも製造する事ができる。 The formula (IM-3-1) when X = Cl in the formula (IM-3) can also be produced by the following [Production Method D] <Step 3> to [Production Method D] <Step 5>. I can do it.
<工程3>
式(AR-2-1)の化合物、および式(AM-1)(式(AR-2-1)、および式(AM-1)の化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)の化合物を用いて、[製造方法B]<工程3>に準じる反応を行い、式(D-1)の化合物を製造することができる。
<Step 3>
A compound of formula (AR-2-1) and a compound of formula (AM-1) (formula (AR-2-1) and compound of formula (AM-1) are commercially available compounds, or known production from literature The compound of formula (D-1) can be produced by carrying out a reaction according to [Production Method B] <Step 3> using a compound of (a compound that can be produced by the method).
<工程4>
[製造方法D]<工程3>で得られた式(D-1)の化合物を用いて、[製造方法A]<工程2>に準じる反応を行い、式(D-2)の化合物を製造することができる。
<Step 4>
[Production Method D] Using the compound of formula (D-1) obtained in <Step 3>, a reaction according to [Production Method A] <Step 2> is carried out to produce a compound of formula (D-2) can do.
<工程5>
[製造方法D]<工程4>で得られた式(D-2)の化合物、および式(CA)の化合物を用いて、[製造方法A]<工程3>に準じる反応を行い、式(IM-3-1)の化合物を製造することができる。
<Step 5>
[Production Method D] Using the compound of the formula (D-2) obtained in <Step 4> and the compound of the formula (CA), a reaction according to [Production Method A] <Step 3> is carried out to give a compound of the formula ( The compound of IM-3-1) can be produced.
また、式(IM-3)でX=Fの場合の式(IM-3-2)は、後述する[製造方法D]<工程6>から[製造方法D]<工程9>の方法でも製造する事ができる。 The formula (IM-3-2) when X = F in the formula (IM-3) can also be produced by the following [Production Method D] <Step 6> to [Production Method D] <Step 9>. I can do it.
<工程6>
式(AR-2-2)の化合物、および式(AM-1)(式(AR-2-2)、および式(AM-1)の化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)の化合物を用いて、[製造方法B]<工程3>に準じる反応を行い、式(D-3)の化合物を製造することができる。
<Step 6>
The compound of formula (AR-2-2), and the compound of formula (AM-1) (formula (AR-2-2) and compound of formula (AM-1) are commercially available compounds, or prepared from commercially available compounds in the literature The compound of formula (D-3) can be produced by carrying out a reaction according to [Production Method B] <Step 3>.
<工程7>
[製造方法D]<工程6>で得られた式(D-3)の化合物、およびトリフルオロ酢酸無水物(TFAA)を用いて、文献公知の方法、例えば、『国際公開2007/043677号パンフレット』等に記載された方法に準じて、トリエチルアミン等の塩基存在下、ジクロロメタン等の反応に関与しない溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(D-4)の化合物を製造することができる。
<Step 7>
[Production Method D] Using the compound of the formula (D-3) obtained in <Step 6> and trifluoroacetic anhydride (TFAA), a method known in the literature, for example, “Patent of International Publication No. WO 2007/043777. In the presence of a base such as triethylamine, the reaction is carried out at a temperature at which the solvent is refluxed from 0 ° C. in the presence of a base such as triethylamine. Compounds can be produced.
<工程8>
[製造方法D]<工程7>で得られた式(D-4)の化合物を用いて、[製造方法A]<工程2>に準じる反応を行い、式(D-5)の化合物を製造することができる。
<Step 8>
[Production Method D] Using the compound of formula (D-4) obtained in <Step 7>, a reaction according to [Production Method A] <Step 2> is carried out to produce a compound of formula (D-5) can do.
<工程9>
[製造方法D]<工程8>で得られた式(D-5)の化合物、および式(CA)の化合物を用いて、[製造方法A]<工程3>に準じる反応を行い、式(IM-3-2)の化合物を製造することができる。
<Step 9>
[Production Method D] Using the compound of formula (D-5) obtained in <Step 8> and the compound of formula (CA), a reaction according to [Production Method A] <Step 3> is carried out to give a compound of formula ( The compound of IM-3-2) can be produced.
[製造方法E]式(CA-3)[式(CA)で環B=Ph、または5~7員ヘテロアリール環(但し、当該5~7員ヘテロアリール環は、ジアザビシクロ[2.2.2]オクタン環と環Bのアミド結合の隣接位(o-位)に窒素原子を有する様な、1-置換-1H-ピラゾール-5-イル基等の環を除く);Q=AR(C6~14アリール基もしくは5~7員ヘテロアリール基)の場合]の製造方法
Figure JPOXMLDOC01-appb-C000030
[製造方法E]
[Production Method E] Formula (CA-3) [In formula (CA), ring B = Ph, or a 5- to 7-membered heteroaryl ring (provided that the 5- to 7-membered heteroaryl ring is diazabicyclo [2.2.2] ] Except for a ring such as a 1-substituted-1H-pyrazol-5-yl group that has a nitrogen atom adjacent to the amide bond of the octane ring and ring B (o-position); Q = AR (C 6 To 14 aryl group or 5- to 7-membered heteroaryl group)]
Figure JPOXMLDOC01-appb-C000030
[Production Method E]
<工程1>
式(E-1)の化合物(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)、および式(RG-2)もしくは式(RG-3)の化合物を用いて、[製造方法C]<工程4>に準じる反応を行い、式(E-2)の化合物を製造することができる。
<Step 1>
A compound of formula (E-1) (the compound is a commercially available compound or a compound that can be produced from a commercially available compound by a known production method in the literature), and a compound of formula (RG-2) or formula (RG-3) The compound of formula (E-2) can be produced by carrying out a reaction according to [Production method C] <Step 4>.
<工程2>
[Rα=C1~6アルキル基(例えば、メチル、エチル基など)の場合]
式(E-2)の化合物を用い、文献公知の方法、例えば、『実験化学講座 第4版 22 有機合成IV 酸・アミノ酸・ペプチド、1-43頁、1992年、丸善』などに記載された方法に準じて、水酸化リチウム、水酸化ナトリウム、水酸化カリウム、炭酸リチウム、炭酸ナトリウム、炭酸カリウム等の塩基存在下、水、メタノール、エタノール、2-プロパノール、N,N-ジメチルホルムアミド、1,4-ジオキサン、テトラヒドロフラン等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(CA-3)の化合物を製造することができる。
<Step 2>
[In the case of R α = C 1-6 alkyl group (eg, methyl, ethyl group, etc.)]
Using a compound of the formula (E-2), it is described in literature known methods such as “Experimental Chemistry Course 4th Edition 22 Organic Synthesis IV Acid / Amino Acid / Peptide, 1-43, 1992, Maruzen”. According to the method, in the presence of a base such as lithium hydroxide, sodium hydroxide, potassium hydroxide, lithium carbonate, sodium carbonate, potassium carbonate, water, methanol, ethanol, 2-propanol, N, N-dimethylformamide, 1, The compound of the formula (CA-3) can be produced by performing the reaction at a temperature at which the solvent is refluxed from 0 ° C. using a solvent that does not participate in the reaction such as 4-dioxane and tetrahydrofuran, or a mixed solvent thereof.
[Rα=tert-ブチル基の場合]
式(E-2)の化合物を用い、文献公知の方法、例えば、『プロテクティブ・グループス・イン・オーガニック・シンセシス(Protective Groups in Organic Synthesis 4thEdition) 第4版、2007年、ジョン ウィリー アンド サンズ(John Wiley & Sons)、グリーン(Greene)ら』の成書に記載された脱保護の方法に準じて、塩酸、硫酸、酢酸、トリフルオロ酢酸等の酸を用いて、0℃から溶媒が還流する温度で反応を行い、式(CA-3)の化合物を製造することができる。
[In the case of R α = tert-butyl group]
Using a compound of formula (E-2), a method known in the literature, for example, “Protective Groups in Organic Synthesis 4th Edition”, 4th edition, 2007, John Willy and Sons (John) The temperature at which the solvent is refluxed from 0 ° C. using an acid such as hydrochloric acid, sulfuric acid, acetic acid or trifluoroacetic acid according to the deprotection method described in the book of Wiley & Sons, Green et al. To produce a compound of formula (CA-3).
[Rα=ベンジル基等の場合]
式(E-2)の化合物を用い、文献公知の方法、例えば、『実験化学講座 第4版 26 有機合成VIII 不斉合成・還元・糖・標識化合物、159-266頁、1992年、丸善』等に記載された方法に準じて、パラジウム-炭素(Pd-C)、ラネーニッケル(Raney-Ni)、酸化白金(PtO2)、ジクロロトリトリフェニルホスフィンルテニウム等の触媒存在下、水素ガス雰囲気下にて、メタノール、エタノール、2-プロパノール、ジエチルエーテル、テトラヒドロフラン、1,2-ジメトキシエタン、1,4-ジオキサン、酢酸エチル、酢酸メチル等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(CA-3)の化合物を製造することができる。
[In the case of R α = benzyl group, etc.]
A method known in the literature using a compound of formula (E-2), for example, “Experimental Chemistry Course 4th Edition 26 Organic Synthesis VIII Asymmetric Synthesis / Reduction / Sugar / Labeled Compound, 159-266, 1992, Maruzen” In the presence of a catalyst such as palladium-carbon (Pd—C), Raney nickel (Raney-Ni), platinum oxide (PtO 2 ), dichlorotritriphenylphosphine ruthenium, etc. in a hydrogen gas atmosphere. , Methanol, ethanol, 2-propanol, diethyl ether, tetrahydrofuran, 1,2-dimethoxyethane, 1,4-dioxane, ethyl acetate, methyl acetate and the like, or a mixed solvent thereof, The reaction can be carried out at a temperature at which the solvent is refluxed from 0 ° C. to produce a compound of formula (CA-3).
<工程3>
式(E-3)の化合物(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)、および式(RG-1)の化合物を用いて、[製造方法C]<工程2>に準じる反応を行い、式(E-2)の化合物を製造することができる。
<Step 3>
Using a compound of formula (E-3) (this compound is a commercially available compound or a compound that can be produced from a commercially available compound by a production method known in the literature) and a compound of formula (RG-1), [Production Method C The compound of formula (E-2) can be produced by carrying out a reaction according to <Step 2>.
[製造方法F]式(CA-4)[式(CA)で環B=Ph、または5~7員ヘテロアリール(但し、当該5~7員ヘテロアリール環は、ジアザビシクロ[2.2.2]オクタン環と環Bのアミド結合の隣接位(o-位)に窒素原子を有する様な、1-置換-1H-ピラゾール-5-イル基等の環を除く);Q=イミダゾール、トリアゾール、ピロリジン、ピペラジン、モルホリン等の基の場合]の製造方法
Figure JPOXMLDOC01-appb-C000031
[製造方法F]
式(CA-2)の化合物、および式(F-1)の化合物(式(CA-2)、および式(F-1)の化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)を用いて、文献公知の方法、例えば、『アンゲヴァンテ ケミ、インターナショナル エディション(Angewandte Chemie,International Edition)、50、48、11511-11515頁、2011年』等に記載された方法に準じて、ヨウ化銅(CuI)等の銅触媒、および炭酸ナトリウム、炭酸カリウム、炭酸セシウム等の塩基存在下、アセトニトリル、テトラヒドロフラン、ジメチルホルムアミド等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(CA-4)の化合物を製造することができる。
[Production Method F] Formula (CA-4) [In formula (CA), ring B = Ph, or 5- to 7-membered heteroaryl (wherein the 5- to 7-membered heteroaryl ring is diazabicyclo [2.2.2] Q = imidazole, triazole, pyrrolidine, except for a ring such as a 1-substituted-1H-pyrazol-5-yl group having a nitrogen atom adjacent to the amide bond of the octane ring and ring B (o-position)) , A group such as piperazine, morpholine]
Figure JPOXMLDOC01-appb-C000031
[Production Method F]
The compound of the formula (CA-2) and the compound of the formula (F-1) (the compound of the formula (CA-2) and the formula (F-1) are commercially available compounds or commercially available compounds from the literature by known production methods. A compound known in the literature, for example, the method described in “Angewandte Chemie, International Edition, 50, 48, 11511-11515, 2011”, etc. Similarly, using a copper catalyst such as copper iodide (CuI) and a solvent that does not participate in the reaction such as acetonitrile, tetrahydrofuran, dimethylformamide, or a mixed solvent thereof in the presence of a base such as sodium carbonate, potassium carbonate, or cesium carbonate. And react at a temperature at which the solvent refluxes from 0 ° C. To produce a compound of formula (CA-4).
[製造方法G]式(CA-5-1)[式(CA)で環B=ピラゾールの場合(1)]の製造方法
Figure JPOXMLDOC01-appb-C000032
[製造方法G]
[Production Method G] Production Method of Formula (CA-5-1) [In the Formula (CA) where Ring B = Pyrazole (1)]
Figure JPOXMLDOC01-appb-C000032
[Production Method G]
<工程1>
式(G-1)の化合物(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)、および式(G-2)のヒドラジン化合物(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)を用いて、文献公知の方法、例えば、『国際公開2007/043677号パンフレット』等に記載された方法に準じて、酢酸存在下、テトラヒドロフラン、2-メトキシエタノール等の反応に関与しない溶媒、もしくはこれらの混合溶媒を用いて、0℃から溶媒が還流する温度で反応を行い、式(G-3)の化合物を製造することができる。
<Step 1>
A compound of the formula (G-1) (the compound is a commercially available compound or a compound that can be produced from a commercially available compound by a method known in the literature), and a hydrazine compound of the formula (G-2) (the compound is a commercially available compound) Or a compound that can be produced from a commercially available compound by a production method known in the literature), in the presence of acetic acid according to a method known in the literature, for example, the method described in “International Publication No. 2007/043777 pamphlet” or the like. Using a solvent that does not participate in the reaction, such as tetrahydrofuran, 2-methoxyethanol, or a mixed solvent thereof, at a temperature at which the solvent is refluxed from 0 ° C. to produce a compound of formula (G-3). it can.
<工程2>
[製造方法G]<工程1>で得られる式(G-3)の化合物を用いて、[製造方法E]<工程2>に準じる反応を行い、式(CA-5-1)の化合物を製造することができる。
<Step 2>
[Production Method G] Using the compound of formula (G-3) obtained in <Step 1>, a reaction according to [Production Method E] <Step 2> is carried out to give a compound of formula (CA-5-1). Can be manufactured.
[製造方法H]式(CA-5-2)[式(CA)で環B=ピラゾールの場合(2)]の製造方法
Figure JPOXMLDOC01-appb-C000033
[製造方法H]
[Production Method H] Production Method of Formula (CA-5-2) [In the Formula (CA) where Ring B = Pyrazole (2)]
Figure JPOXMLDOC01-appb-C000033
[Production Method H]
<工程1>
式(H-1)の化合物(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)、および式(G-2)のヒドラジン化合物を用いて、[製造方法G]<工程1>に準じる反応を行い、式(H-2)の化合物を製造することができる。
<Step 1>
Using the compound of the formula (H-1) (the compound is a commercially available compound or a compound that can be produced from a commercially available compound by a production method known in the literature) and a hydrazine compound of the formula (G-2), [Production Method G] A compound of formula (H-2) can be produced by carrying out a reaction according to <Step 1>.
<工程2>
[製造方法H]<工程1>で得られる式(H-2)の化合物を用いて、[製造方法E]<工程2>に準じる反応を行い、式(CA-5-2)の化合物を製造することができる。
<Step 2>
[Production Method H] Using the compound of formula (H-2) obtained in <Step 1>, a reaction according to [Production Method E] <Step 2> is carried out to give a compound of formula (CA-5-2). Can be manufactured.
[製造方法J]式(CA-5-3)[式(CA)で環B=ピラゾールの場合(3)]の製造方法
Figure JPOXMLDOC01-appb-C000034
[製造方法J]
 文献公知の方法、例えば、『バイオオルガニック アンド メディシナル ケミストリー レターズ(Bioorganic & Medicinal Chemistry Letters)、23、23、6341-6345頁、2013年』等に記載された方法に準じて、テトラヒドロフラン等の溶媒中、-78℃にて、ジイソプロピルアミン、およびn-ブチルリチウムを反応させ、リチウムジイソプロピルアミド(LDA)を調整し、同温にて式(J-1)の化合物(該化合物は、市販化合物、または市販化合物から文献公知の製造方法により製造できる化合物である)を加え、更に同温にて反応溶液に過剰のドライアイス(固体片)もしくは過剰の炭酸ガスを加え反応を行い、後処理段階で塩酸等の酸で酸性化する事で、式(CA-5-3)の化合物を製造することができる。
[Production Method J] Production Method of Formula (CA-5-3) [In the Formula (CA) where Ring B = Pyrazole (3)]
Figure JPOXMLDOC01-appb-C000034
[Production Method J]
In a solvent such as tetrahydrofuran in accordance with a method known in the literature, for example, the method described in “Bioorganic & Medicinal Chemistry Letters, 23, 23, 6341-6345, 2013”, etc. , Diisopropylamine and n-butyllithium were reacted at −78 ° C. to prepare lithium diisopropylamide (LDA), and the compound of formula (J-1) (the compound was a commercially available compound or It is a compound that can be produced from a commercially available compound by a known production method in the literature), and at the same temperature, the reaction solution is added with excess dry ice (solid piece) or excess carbon dioxide gas to carry out the reaction. By acidification with an acid such as (CA-5-3) It can be prepared of compounds.
[本発明の化合物を含有する併用剤]
 本発明の化合物や医薬組成物は、医療現場で行われている一般的な方法で、他の薬物もしくは薬剤と併用することも可能である。本発明の化合物と配合または併用しうる薬物としては、例えば、(A)睡眠障害関連薬、(B)睡眠障害を併発しやすい疾患の治療薬等が挙げられる。
[Combination agent containing the compound of the present invention]
The compound and pharmaceutical composition of the present invention can be used in combination with other drugs or drugs by a general method used in the medical field. Examples of the drug that can be combined or used in combination with the compound of the present invention include (A) a sleep disorder-related drug, (B) a therapeutic drug for a disease that easily causes sleep disorder, and the like.
 上記(A)の薬物としては、例えば、(1)睡眠導入剤((i)ベンゾジアゼピン系睡眠導入剤[具体的には、ニトラゼパム、エスタゾラム、塩酸フルラゼパム、ニメタゼパム、フルラゼパム、ハロキサゾラム、フルニトラゼパム、塩酸リルマザポン、ロルメタゼパム、トリアゾラム等]、(ii)チエノジアゼピン系眠導入剤[具体的には、ブロチゾラム等]、(iii)非ベンゾジアゼピン系睡眠導入剤[具体的には、ゾルピデム等]、(iv)メラトニン受容体作動薬[具体的には、ラメルテオン等])、(v)シクロピロロン系眠導入剤[具体的には、ゾピクロン等]、(vi)オレキシン受容体拮抗薬[具体的には、スボレキサント等]、(2)睡眠時無呼吸症候群で処方される薬剤[具体的にはアセタゾラミド等]、(3)レストレスレッグス症候群で処方される薬剤((i)ベンゾジアゼピン系睡眠導入剤[具体的には、クロナゼパム等]、(ii)ドーパミン作動薬[具体的には、レボドパ、塩酸アマンタジン、メシル酸ブロモクリプチン、メシル酸ペルゴリド、カベルゴリン、塩酸タリペキソール、塩酸プラミペキソール水和物、塩酸セレギリン、塩酸ロピニロール等]、(iii)オピオイド製剤[具体的には、コデイン等])等が挙げられる。 Examples of the drug (A) include: (1) sleep inducers ((i) benzodiazepine-based sleep inducers [specifically, nitrazepam, estazolam, flurazepam hydrochloride, nimetazepam, flurazepam, haloxazolam, flunitrazepam, rilmazapine hydrochloride, Lormetazepam, triazolam, etc.], (ii) thienodiazepine sleep inducer [specifically, brotizolam, etc.], (iii) non-benzodiazepine sleep inducer [specifically, zolpidem, etc.], (iv) melatonin receptor operation Drugs (specifically, ramelteon, etc.), (v) cyclopyrrolone sleep inducers [specifically, zopiclone, etc.], (vi) orexin receptor antagonists [specifically, suvorexant, etc.], ( 2) Drugs prescribed for sleep apnea syndrome [specifically, acetazolamide, etc.], (3) Drugs prescribed for Treslegs syndrome ((i) benzodiazepine sleep inducers [specifically, clonazepam, etc.], (ii) dopamine agonists [specifically, levodopa, amantadine hydrochloride, bromocriptine mesylate, mesylate] Pergolide, cabergoline, talipexol hydrochloride, pramipexole hydrochloride hydrate, selegiline hydrochloride, ropinirole hydrochloride, etc.], (iii) opioid preparations [specifically, codeine etc.]) and the like.
 上記(B)の薬物としては、例えば(4)非定型抗精神病薬[具体的には、オランザピン、クエチアピン、クロザピン、ジプラシドン、リスペリドン、パリペリドン、ペロスピロン、ブロナンセリン、ルラシドン、アリピプラゾール、セルチンドール、アミスルプリド、イロペリドン、ビフェプルノックス、アセナピン、メルペロン、ブレクスピプラゾール、ゾテピン等]、(5)定型抗精神病薬[具体的には、クロルプロマジン、プクロルペラジン、ペルフェナジン、レボメプロマジン、フルフェナジン、チオリダジン、プロペリシアジン、スピペロン、モペロン、ハロペリドール、チミペロン、ブロムペリドール、ピモジド、フロロピパミド、スルピリド、チアプリド、スルトプリド、ネモナプリド、オキシペルチン等]、(6)選択的セロトニン再取り込み阻害薬(SSRI)[具体的には、エスシタロプラム、シタロプラム、パロキセチン、セルトラリン、フルボキサミン、フルオキセチン等]、(7)選択的セロトニン・ノルアドレナリン再取り込み阻害薬(SNRI)[具体的には、ミルナシプラン、デュロキセチン、ベンラファキシン、ネファゾドン等]、(8)選択的ノルアドレナリン・ドーパミン再取り込み阻害薬(NDRI)[具体的には、ブブロピン等]、(9)ノルアドレナリン作動性・特異的セロトニン作動性抗うつ薬(NaSSA)[具体的には、ミルタザピン等]、(10)トリアゾロピリジン系抗うつ薬(SARI)[具体的には、トラゾドン等]、(11)四環系抗うつ薬[具体的には、セチプチリン、ミアンセリン、マプロチリン等]、(12)三環系抗うつ薬[具体的には、アミトリプチリン、トリミプラミン、イミプラミン、ノルトリプチリン、クロミプラミン、ロフェプラミン、アモキサピン、ドスレピン等]、(13)その他抗うつ薬[具体的には、NS-2359、Lu AA21004、DOV21947等]、(14)α7ニコチン受容体作動薬、(15)α7ニコチン受容体活性調節薬、(16)α7ニコチン受容体部分調節薬[具体的には、SSR-180711、PNU-120596等]、(17)PDE阻害薬[PDE1阻害薬、PDE2阻害薬、PDE4阻害薬、PDE5阻害薬、PDE7阻害薬、PDE9阻害薬、PDE10阻害薬等]、(18)NK2拮抗薬、(19)NK3拮抗薬、 Examples of the drug of (B) include (4) atypical antipsychotic drugs [specifically, olanzapine, quetiapine, clozapine, ziprasidone, risperidone, paliperidone, perospirone, blonanserin, lurasidone, aripiprazole, sertindole, amisulpride, Iloperidone, bifeprunox, asenapine, melperone, brexpiprazole, zotepine, etc.], (5) typical antipsychotics [specifically, chlorpromazine, pchlorperazine, perphenazine, levomepromazine, fluphenazine, thioridazine, propericazine, spiperone] , Moperon, haloperidol, timiperone, bromperidol, pimozide, fluropipamide, sulpiride, thioprid, sultopride, nemonapride, oxypertin, etc.], (6) selection Serotonin reuptake inhibitor (SSRI) [specifically, escitalopram, citalopram, paroxetine, sertraline, fluvoxamine, fluoxetine, etc.], (7) selective serotonin / noradrenaline reuptake inhibitor (SNRI) [specifically, MIL Nasiplan, duloxetine, venlafaxine, nefazodone, etc.], (8) selective noradrenaline / dopamine reuptake inhibitor (NDRI) [specifically, bubropine, etc.], (9) noradrenergic / specific serotonergic Antidepressant (NaSSA) [specifically, mirtazapine and the like], (10) Triazolopyridine antidepressant (SARI) [specifically, trazodone and the like], (11) Tetracyclic antidepressant [specifically In particular, cetiptiline, mianserin, maprotiline, etc.], ( 2) Tricyclic antidepressants [specifically, amitriptyline, trimipramine, imipramine, nortriptyline, clomipramine, lofepramine, amoxapine, dosrepin, etc.], (13) other antidepressants [specifically, NS-2359, Lu AA21004, DOV21947 etc.], (14) α7 nicotinic receptor agonist, (15) α7 nicotinic receptor activity modulator, (16) α7 nicotinic receptor partial modulator [specifically, SSR-180711, PNU-120596] Etc.], (17) PDE inhibitors [PDE1 inhibitors, PDE2 inhibitors, PDE4 inhibitors, PDE5 inhibitors, PDE7 inhibitors, PDE9 inhibitors, PDE10 inhibitors, etc.], (18) NK2 antagonists, (19) NK3 antagonist,
(20)ムスカリン型M1アセチルコリン受容体活性調節薬、(21)ムスカリン型M2アセチルコリン受容体活性調節薬、(22)アデノシン受容体調節薬、(23)ムスカリン型M4アセチルコリン受容体活性調節薬、(24)ムスカリン型M5アセチルコリン受容体活性調節薬、(25)アデノシン受容体調節薬、(26)グリシントランスポーター1(GlyT1)阻害薬[具体的には、ALX5407、SSR504734等]、(27)グルタミン酸増強薬[具体的には、アンパカイン]、(28)NMDA受容体阻害薬[具体的には、塩酸メマンチン等]、(29)代謝性グルタミン酸受容体調節薬(mGlu)[具体的には、CDPPB、MPEP等]、(30)抗不安薬((i)ベンゾジアゼピン系抗不安薬[具体的には、クロルジアゼポキシド、ジアゼパム、オキサゾラム、メダゼパム、クロキサゾラム、ロラゼパム、クロラゼプ酸二カリウム、プラゼパム、ブロマゼパム、フルジアゼパム、メキサゾラム、アルプラゾラム、フルトプラゼパム、フルタゾラム、ロフラゼプ酸エチル等]、(ii)チエノジアゼピン系抗不安薬[具体的には、エチゾラム、クロチアゼパム等]、(iii)セロトニン5-HT1A作動薬[具体的には、タンドスピロン等])、(31)βアミロイドワクチン、(32)βアミロイド分解酵素等、(33)脳機能賦活薬[具体的には、アニラセタム、ニセルゴリン等]、(34)カンナビノイド調節薬、(35)コリンエステラーゼ阻害薬[具体的には、塩酸ドネペジル、リバスチグミン、臭化水素酸ガランタミン等]、(36)MAO-B阻害剤[具体的には、ラサリジン等]、(37)パーキンソン病治療薬((i)ドーパミン受容体作動薬[具体的には、レボドパ、塩酸アマンタジン、メシル酸ブロモクリプチン、メシル酸ペルゴリド、カベルゴリン、塩酸タリペキソール、塩酸プラミペキソール水和物、塩酸セレギリン、塩酸ロピニロール等]、(ii)モノアミン酸化酵素阻害薬[具体的には、デプレニル、セルジリン(セレギリン)、レマセミド,リルゾール等]、(iii)抗コリン剤[具体的には、トリヘキシフェニジル、プロフェナミン、ビペリデン、塩酸ピロヘプチン、塩酸メチキセン、塩酸マザチコール等]、(iv)COMT阻害剤[具体的には、エンタカポン等]、(v)筋萎縮性側索硬化症治療薬[具体的には、リルゾール等、神経栄養因子等]、(vi)アポトーシス阻害薬[具体的には、CPI-1189、IDN-6556、CEP-1347等]、(vii)神経分化・再生促進剤[具体的には、レテプリニム(Leteprinim]、キサリプローデン(Xaliproden;SR-57746-A]、SB-216763等])、 (20) Muscarinic M1 acetylcholine receptor activity modulator, (21) Muscarinic M2 acetylcholine receptor activity modulator, (22) Adenosine receptor modulator, (23) Muscarinic M4 acetylcholine receptor activity regulator, (24 ) Muscarinic M5 acetylcholine receptor activity modulator, (25) adenosine receptor modulator, (26) glycine transporter 1 (GlyT1) inhibitor [specifically, ALX5407, SSR504734, etc.], (27) glutamate enhancer [Specifically, ampakine], (28) NMDA receptor inhibitor [specifically, memantine hydrochloride and the like], (29) metabolic glutamate receptor modulator (mGlu) [specifically, CDPPB, MPEP Etc.], (30) Anti-anxiety drugs ((i) benzodiazepine anxiolytic drugs [specifically Chlordiazepoxide, diazepam, oxazolam, medazepam, cloxazolam, lorazepam, dipotassium chlorazepate, prazepam, bromazepam, fludiazepam, mexazolam, alprazolam, fltoprazepam, flutazolam, ethyl loflazepate, etc. Ethiram, clothiazepam, etc.], (iii) serotonin 5-HT1A agonist [specifically, tandospirone, etc.)), (31) β-amyloid vaccine, (32) β-amyloid degrading enzyme, etc., (33) activation of brain function Drug [specifically, aniracetam, nicergoline, etc.], (34) cannabinoid modulator, (35) cholinesterase inhibitor [specifically, donepezil hydrochloride, rivastigmine, galantamine hydrobromide, etc.], (36) AO-B inhibitors [specifically, lasalidine, etc.], (37) Parkinson's disease therapeutic agent ((i) dopamine receptor agonists [specifically, levodopa, amantadine hydrochloride, bromocriptine mesylate, pergolide mesylate, Cabergoline, talipexol hydrochloride, pramipexole hydrochloride hydrate, selegiline hydrochloride, ropinirole hydrochloride, etc.], (ii) monoamine oxidase inhibitors [specifically, deprenyl, sergiline (selegiline), remasemide, riluzole, etc.], (iii) anti Choline agents [specifically, trihexyphenidyl, prophenamine, biperidene, pyroheptin hydrochloride, methixene hydrochloride, masaticol hydrochloride, etc.], (iv) COMT inhibitors [specifically, entacapone, etc.], (v) muscle atrophy Drugs for lateral sclerosis [specifically, riluzole and other neurotrophic factors Etc.], (vi) Apoptosis inhibitors [specifically CPI-1189, IDN-6556, CEP-1347, etc.], (vii) Neural differentiation / regeneration promoters [Specifically, leteprinim, xaliproden (Xaliproden; SR-57746-A], SB-216763 etc.),
(38)糖尿病治療薬((i)PPARγ作用薬(作動薬、阻害薬)[具体的には、ピオグリタゾン、ロシグリタゾン、トログリタゾン、シグリタゾン、ダルグリタゾン、エングリタゾン、ネトグリタゾン等]、(ii)インスリン分泌促進薬[(a)スルホニル尿素剤(具体的には、トルブタミド、アセトヘキサミド、クロルプロパミド、グリベンクラミド、グリクラジド、グリピジド、グリメピリド、グリペンチド、グリキドン、グリソラミド、トラザミド等)、(b)非スルホニル尿素剤等]、(iii)速効型インスリン分泌促進剤(具体的には、ナテグリニド、ミチグリニド、レパグリニド等)、(iv)αグルコシダーゼ阻害薬[具体的には、アカルボース、ボグリボース、ミグリトール、カミグリボース、アジポシン、エミグリテート、プラジミシン-Q、サルボスタチン等]、(v)インスリン抵抗性改善薬[具体的には、(a)PPAR-γ作用薬、(b)PTP-1B阻害薬、(c)DPP-4阻害薬[具体的には、シタグリプチン、ビルダグリプチン、アログリプチン、サクサグリプチン、NVP-DPP-728等]、(d)GLP-1及びGLP-1作動薬[具体的には、エキセナチド、リラグルチド等]、(e)11β-HSD阻害薬等、(f)GPR40作動薬、(g)GPR119作動薬、(h)GPR120作動薬]、(vi)肝糖新生抑制剤[具体的には、グルカゴン拮抗薬等]、(vii)ビグアナイド剤[具体的には、メトホルミン、ブホルミン、フェンホルミン等]、(viii)インスリンまたはインスリン誘導体[具体的には、インスリン亜鉛懸濁液、インスリンリスプロ、インスリンアスパルト、レギュラーインスリン、NPHインスリン、インスリングラルギン、インスリンデテミル、混合型インスリン等]、(ix)α2拮抗薬[具体的には、ミダグリゾール、イサグリドール、デリグリドール、イダゾキサン、エファロキサン等])、 (38) Antidiabetic agent ((i) PPARγ agonist (agonist, inhibitor) [specifically, pioglitazone, rosiglitazone, troglitazone, siglitazone, darglitazone, englitazone, netoglitazone, etc.], (ii) insulin Secretion enhancer [(a) sulfonylurea (specifically, tolbutamide, acetohexamide, chlorpropamide, glibenclamide, gliclazide, glipizide, glimepiride, glipentide, glyquidone, glisolamide, tolazamide, etc.), (b) non-sulfonylurea Agents, etc.], (iii) fast-acting insulin secretagogues (specifically, nateglinide, mitiglinide, repaglinide, etc.), (iv) α-glucosidase inhibitors [specifically, acarbose, voglibose, miglitol, camiglibose, adiposine, Emigli Tate, pradimicin-Q, sarvostatin, etc.], (v) insulin sensitizers [specifically, (a) PPAR-γ agonists, (b) PTP-1B inhibitors, (c) DPP-4 inhibition Drugs [specifically sitagliptin, vildagliptin, alogliptin, saxagliptin, NVP-DPP-728, etc.], (d) GLP-1 and GLP-1 agonists [specifically, exenatide, liraglutide, etc.], (e) 11β-HSD inhibitors, etc. (f) GPR40 agonists, (g) GPR119 agonists, (h) GPR120 agonists], (vi) hepatic gluconeogenesis inhibitors [specifically, glucagon antagonists, etc.], vii) biguanides [specifically, metformin, buformin, phenformin, etc.], (viii) insulin or insulin derivatives [specifically, insulin Phosphorus zinc suspension, insulin lispro, insulin aspart, regular insulin, NPH insulin, insulin glargine, insulin detemir, mixed insulin, etc.], (ix) α2 antagonist [specifically, midaglyzol, isagridol, deliglidol, idazoxan , Efaloxane, etc.]),
(39)抗肥満薬((i)アドレナリンβ3受容体作動薬[具体的には、KRP-204、TRK-380/TAC-301等]、(ii)CB-1受容体拮抗薬[具体的には、リモナバン、SR-147778、BAY-65-2520等]、(iii)ニューロペプチドY(NPY)受容体拮抗薬[具体的には、S-2367等]、(iv)摂食抑制薬[モノアミン再取り込み阻害剤[具体的には、シブトラミン、マジンドール等]]、(v)リパーゼ阻害薬[具体的には、オルリスタット、セチリスタット等]、(vi)ペプチドYY(PYY)受容体拮抗薬等)、(40)コレステロール低下薬等の高脂血症治療薬((i)ω3脂肪酸類[具体的には、イコサペント酸エチル(EPA-E製剤、例えば、製品名:エパデール(登録商標)等)、ドコサヘキサエン酸(DHA)、イコサペント酸エチルおよびドコサヘキサエン酸エチルの混合製剤(例えば、製品名:ロバザ(登録商標)、オマコール(登録商標)等)等]、(ii)HMG-CoA還元酵素阻害剤[具体的には、アトルバスタチン、シンバスタチン、ピタバスタチン、イタバスタチン、フルバスタチン、ロバスタチン、プラバスタチン、リバスタチン、ロスバスタチン等](iii)HMG-CoA合成酵素阻害剤、(iv)コレステロール吸収阻害剤[具体的には、エゼチミブ]、(v)アシル-CoA・コレステロールアシル転移酵素阻害剤、(vi)CETP阻害剤、(vii)スクアレン合成酵素阻害剤、(viii)抗酸化剤[具体的には、プロブコール等]、(ix)PPARα作動薬[具体的には、クロフィブラート、エトフィブラート、フェノフィブラート、ベザフィブラート、シプロフィブラート、ゲムフィブロジル、KRP-101等]、(x)PPARδ作動薬、(xi)LXR作動薬、(xii)FXR作動薬[具体的には、INT-747等]、(xiii)MTTP阻害剤、(xiv)スクアレンエポシダーゼ阻害剤等)、 (39) anti-obesity drugs ((i) adrenergic β3 receptor agonists [specifically, KRP-204, TRK-380 / TAC-301, etc.], (ii) CB-1 receptor antagonists [specifically Rimonabant, SR-147778, BAY-65-2520 etc.], (iii) Neuropeptide Y (NPY) receptor antagonist [specifically, S-2367 etc.], (iv) Antifeedant [monoamine] Reuptake inhibitors [specifically sibutramine, mazindol, etc.]], (v) lipase inhibitors [specifically, orlistat, cetiristat, etc.], (vi) peptide YY (PYY) receptor antagonists, etc.), (40) Antihyperlipidemic drugs such as cholesterol-lowering drugs ((i) ω3 fatty acids [specifically, ethyl icosapentate (EPA-E preparation, for example, product name: Epadale (registered trademark) ), Docosahexaenoic acid (DHA), ethyl icosapentate and ethyl docosahexaenoate (eg, product name: Lovaza (registered trademark), omacol (registered trademark), etc.), etc.], (ii) HMG-CoA reductase inhibitor [Specifically, atorvastatin, simvastatin, pitavastatin, itavastatin, fluvastatin, lovastatin, pravastatin, rivastatin, rosuvastatin, etc.] (iii) HMG-CoA synthase inhibitor, (iv) cholesterol absorption inhibitor [specifically , Ezetimibe], (v) acyl-CoA / cholesterol acyltransferase inhibitor, (vi) CETP inhibitor, (vii) squalene synthase inhibitor, (viii) antioxidant [specifically, probucol and the like], (Ix) PPARα agonist [specifically, black Ibrate, etofibrate, fenofibrate, bezafibrate, ciprofibrate, gemfibrozil, KRP-101, etc.], (x) PPARδ agonist, (xi) LXR agonist, (xii) FXR agonist [specifically, INT-747 Etc.], (xiii) MTTP inhibitor, (xiv) squalene eposidase inhibitor, etc.)
(41)降圧剤((i)利尿剤[具体的には、トリクロルメチアジド、ヒドロクロロチアジド、メフルシド、インダパミド、メチクラン、クロルタリドン、トリパミド、フロセミド、トラセミド、ブメタニド、エタクリン酸、スピロノラクトン、トリアムテレン、エプレレノン等]、(ii)カルシウム受容体拮抗薬[具体的には、アムロジピン、フェロジピン、ニカルジピン、ニフェジピン、ニモジピン、ニトレンジピン、ニルバジピン、アラニジピン、アゼルニジピン、マニジピン、バルニジピン、エホニジピン、シルニジピン、ベニジピン、ジルチアゼム等]、(iii)アンジオテンシン変換酵素阻害薬[具体的には、カプトプリル、リシノプリル、エナラプリル、デラプリル、ペリンドプリル、ベナゼプリル、トランドラプリル、キナプリル、アラセプリル、イミダプリル、テモカプリル、シラザプリル等]、(iv)アンジオテンシン受容体拮抗薬[具体的には、ロサルタン、オルメサルタン、テルミサルタン、バルサルタン、カンデサルタンシレキセチル、イルベサルタン等]、(v)直接的レニン阻害薬[具体的には、アリスキレン等]、(vi)α受容体遮断薬[具体的には、トラゾリン、フェントラミン、ドキサゾシン、プラゾシン、ブナゾシン、テラゾシン、ウラピジル等]、(vii)β受容体遮断薬[具体的には、ボピンドロール、ピンドロール、チモロール、ジクロロイソプレナリン、アルプレノロール、カルテオロール、インデノロール、ブニトロロール、ペンブトロール、プロプラノロール、ナドロール、ニプラジロール、チリソロール、アセブトロール、セリプロロール、メトプロロール、アテノロール、ビソプロロール、ベタキソロール、プラクトロール、ベバントロール、ブトキサミン、カルベジロール、アモスラロール、アロチノロール、ラベタロール等]、(viii)α1β遮断薬[具体的には、カルベジロール、ラベタロール、アロチノロール、ベバントロール等]、(ix)α2受容体刺激薬[具体的には、クロニジン、メチルドパ、グアンファシン等])、 (41) Antihypertensive agent ((i) diuretic [specifically, trichlormethiazide, hydrochlorothiazide, mefluside, indapamide, methiclan, chlorthalidone, tripamide, furosemide, torasemide, bumetanide, ethacrynic acid, spironolactone, triamterene, eplerenone, etc.] (Ii) Calcium receptor antagonists [specifically, amlodipine, felodipine, nicardipine, nifedipine, nimodipine, nitrendipine, nilvadipine, alanidipine, azelnidipine, manidipine, varnidipine, efonidipine, cilnidipine, benidipine, diltiazem etc.], (synten) Converting enzyme inhibitors [specifically, captopril, lisinopril, enalapril, delapril, perindopril, benazepril, trandolapril, Napril, alacepril, imidapril, temocapril, cilazapril, etc.], (iv) angiotensin receptor antagonist [specifically, losartan, olmesartan, telmisartan, valsartan, candesartan cilexetil, irbesartan, etc.], (v) direct renin inhibition Drugs [specifically, aliskiren, etc.], (vi) α receptor blockers [specifically, tolazoline, phentolamine, doxazosin, prazosin, bunazosin, terazosin, urapidil, etc.], (vii) β receptor blockers [ Specifically, bopindolol, pindolol, timolol, dichloroisoprenalin, alprenolol, carteolol, indenolol, bunitrolol, penbutolol, propranolol, nadolol, nipradilol, chilisolol, acebutol Lumpur, celiprolol, metoprolol, atenolol, bisoprolol, betaxolol, practolol, bevantolol, butoxamine, carvedilol, amosulalol, arotinolol, labetalol, etc.], the (viii) α 1 β-blockers [specifically, carvedilol, labetalol, Arotinolol, bevantolol, etc.], (ix) α 2 receptor stimulants [specifically, clonidine, methyldopa, guanfacine, etc.]),
(42)非ステロイド性抗炎症薬[具体的には、メロキシカム、テオキシカム、インドメタシン、イブプロフェン、セレコキシブ、ロフェコキシブ、アスピリン、インドメタシン等]、(43)疾患修飾性抗リウマチ薬、(44)抗サイトカイン薬[具体的には、TNF阻害薬、MAPキナーゼ阻害薬]、(45)ステロイド薬[具体的には、デキサメサゾン、ヘキセストロール、酢酸コルチゾン等]、(46)性ホルモンまたはその誘導体[具体的には、プロゲステロン、エストラジオール、安息香酸エストラジオール等]、(47)副甲状腺ホルモン、(48)オピオイド作動薬[具体的には、モルヒネ、ペンタゾシン、トラマドール]、(49)ピリン系解熱鎮痛薬[具体的には、スルピリン]、(50)非ピリン系解熱鎮痛薬[具体的には、アセトアミノフェン(パラセタモール)]、(51)非ステロイド性抗炎症薬(NSAIDs)[具体的には、アスピリン、メフェナム酸、ジクロフェナク、インドメタシン、イブプロフェン、ケトプロフェン、ナプロキセン、プラノプロフェン、ロキソプロフェン、ザルトプロフェン、ピロキシカム、ロルノキシカム、エピリゾール、チアラミド]、(52)COX-2選択的阻害薬[具体的には、セレコキシブ]、(53)末梢性神経障害性疼痛・線維筋痛症薬[具体的には、プレガバリン]が挙げられる。また、神経因性疼痛に転用し、処方されている以下の薬物、(54)抗てんかん薬[具体的には、ガバペンチン、フェニトイン、カルバマゼピン]等が挙げられる。 (42) Non-steroidal anti-inflammatory drugs [specifically, meloxicam, teoxicam, indomethacin, ibuprofen, celecoxib, rofecoxib, aspirin, indomethacin, etc.], (43) disease-modifying anti-rheumatic drugs, (44) anti-cytokine drugs [44] Specifically, TNF inhibitors, MAP kinase inhibitors], (45) steroid drugs [specifically, dexamethasone, hexestrol, cortisone acetate, etc.], (46) sex hormones or derivatives thereof [specifically, , Progesterone, estradiol, estradiol benzoate, etc.], (47) parathyroid hormone, (48) opioid agonist [specifically, morphine, pentazocine, tramadol], (49) pilin antipyretic analgesic [specifically, Sulpyrine], (50) non-pyrine antipyretic analgesics [specific Acetaminophen (paracetamol)], (51) non-steroidal anti-inflammatory drugs (NSAIDs) [specifically, aspirin, mefenamic acid, diclofenac, indomethacin, ibuprofen, ketoprofen, naproxen, pranoprofen, loxoprofen, Zaltoprofen, piroxicam, lornoxicam, epirisol, thiaramide], (52) COX-2 selective inhibitor [specifically, celecoxib], (53) peripheral neuropathic pain / fibromyalgia agent [specifically , Pregabalin]. In addition, the following drugs that have been diverted to neuropathic pain and are prescribed (54) antiepileptic drugs [specifically, gabapentin, phenytoin, carbamazepine] and the like can be mentioned.
 上記疾患に対して既存薬と併用することにより、既存薬の投薬量を下げることが可能であり、既存薬の副作用を軽減することが可能となる。もちろん、当該薬物を用いた併用方法は、上記疾患に限定されるものではなく、且つ併用される薬物は上記に例示した化合物に限定されない。
 本発明の化合物と併用される薬物とを組み合わせて使用する場合は、別々の製剤であっても、合剤であってもよい。また、別々の製剤においては、両者を同時に服用することも、時間をずらして投与することも可能である。
By using in combination with existing drugs for the above-mentioned diseases, it is possible to reduce the dosage of existing drugs and reduce the side effects of existing drugs. Of course, the combination method using the drug is not limited to the above diseases, and the drug used in combination is not limited to the compounds exemplified above.
When the compound of the present invention is used in combination with a drug used in combination, it may be a separate preparation or a combination. Moreover, in separate preparations, both can be taken simultaneously or can be administered at different times.
 本発明の化合物は、単回または多回投与のいずれかで、単独でまたは薬学的に許容できる担体と組み合わせて投与することができる。適切な医薬担体には、不活性固体希釈剤または充填剤、滅菌水溶液、および種々の有機溶媒が包含される。それにより形成される医薬組成物は次いで、錠剤、粉剤、ロゼンジ、液体調剤、シロップ剤、注射液などの様々な投与形態で容易に投与することができる。これらの医薬組成物は、香味剤、結合剤、賦形剤などの追加成分を場合により含有できる。したがって、本発明の化合物は、経口、口腔、鼻腔、非経口(例えば、静脈内、筋内、または皮下)、経皮(たとえば、パッチ)、もしくは直腸投与用に、または吸入もしくは注入(insufflation)による投与に適した形態で製剤化することができる。 The compounds of the present invention can be administered either alone or in combination with a pharmaceutically acceptable carrier, either single or multiple doses. Suitable pharmaceutical carriers include inert solid diluents or fillers, sterile aqueous solution, and various organic solvents. The pharmaceutical composition thereby formed can then be easily administered in various dosage forms such as tablets, powders, lozenges, liquid preparations, syrups, injection solutions and the like. These pharmaceutical compositions can optionally contain additional ingredients such as flavoring agents, binders, excipients and the like. Accordingly, the compounds of the present invention may be used for oral, buccal, nasal, parenteral (eg, intravenous, intramuscular, or subcutaneous), transdermal (eg, patch), or rectal administration, or by inhalation or insufflation. Can be formulated in a form suitable for administration by.
[併用・配合剤/組み合わせ剤の投与形態]
 本発明の化合物と併用薬物の投与形態は、特に限定されず、投与時に、本発明の化合物と併用薬物とが組み合わされていればよい。このような投与形態としては、例えば、
(1)本発明の化合物と併用薬物とを同時に製剤化して得られる単一の製剤の投与、
(2)本発明の化合物と併用薬物とを別々に製剤化して得られる2種の製剤の同一投与経路での同時投与、
(3)本発明の化合物と併用薬物とを別々に製剤化して得られる2種の製剤の同一投与経路での時間差をおいての投与、
(4)本発明の化合物と併用薬物とを別々に製剤化して得られる2種の製剤の異なる投与経路での同時投与、
(5)本発明の化合物と併用薬物とを別々に製剤化して得られる2種の製剤の異なる投与経路での時間差をおいての投与(例えば、本発明の化合物→併用薬物の順序での投与、または逆の順序での投与)などが用いられる。以下、これらの投与形態をまとめて、本発明の併用剤と略記する。
[Combination / Combination / Combination dosage form]
The administration mode of the compound of the present invention and the concomitant drug is not particularly limited as long as the compound of the present invention and the concomitant drug are combined at the time of administration. Such dosage forms include, for example,
(1) administration of a single preparation obtained by simultaneously formulating the compound of the present invention and a concomitant drug,
(2) Simultaneous administration by the same route of administration of two types of preparations obtained by separately formulating the compound of the present invention and a concomitant drug,
(3) Administration of two types of preparations obtained by separately formulating the compound of the present invention and a concomitant drug at the same administration route with a time difference,
(4) Simultaneous administration of two types of preparations obtained by separately formulating the compound of the present invention and the concomitant drug by different administration routes,
(5) Administration of two types of preparations obtained by separately formulating the compound of the present invention and a concomitant drug at different time intervals in different administration routes (for example, administration in the order of the compound of the present invention → the concomitant drug) Or administration in the reverse order). Hereinafter, these administration forms are collectively abbreviated as the combination agent of the present invention.
 本発明の併用剤を投与するに際しては、併用薬物と本発明の化合物とを同時期に投与してもよいが、併用薬物の投与の後、本発明の化合物を投与してもよいし、本発明の化合物の投与後、併用薬物を投与してもよい。時間差をおいて投与する場合、時間差は投与する有効成分、剤形、および投与方法により異なるが、例えば、併用薬物を先に投与する場合、併用薬物を投与した後1分~3日以内、好ましくは10分~1日以内、より好ましくは15分~1時間以内に本発明の化合物を投与する方法が挙げられる。本発明の化合物を先に投与する場合、本発明の化合物を投与した後、1分~1日以内、好ましくは10分~6時間以内、より好ましくは15分~1時間以内に併用薬物を投与する方法が挙げられる。 When administering the concomitant drug of the present invention, the concomitant drug and the compound of the present invention may be administered at the same time, but after administering the concomitant drug, the compound of the present invention may be administered. A concomitant drug may be administered after administration of the compound of the invention. When administered at a time difference, the time difference varies depending on the active ingredient to be administered, dosage form, and administration method. For example, when administering the concomitant drug first, preferably within 1 minute to 3 days after administering the concomitant drug. Includes a method of administering the compound of the present invention within 10 minutes to 1 day, more preferably within 15 minutes to 1 hour. When the compound of the present invention is administered first, the concomitant drug is administered within 1 minute to 1 day, preferably within 10 minutes to 6 hours, more preferably within 15 minutes to 1 hour after the administration of the compound of the present invention. The method of doing is mentioned.
 併用薬物は、副作用が問題とならなければ、どのような量を設定することも可能である。併用薬物としての一日投与量は、投与量は、投与対象、投与ルート、対象疾患、症状等によっても異なるが、例えば、統合失調症の患者(成人、体重約60kg)に経口投与する場合、通常1回量として約0.1~約20mg/kg体重、好ましくは約0.2~約10mg/kg体重、さらに好ましくは約0.5~約10mg/kg体重であり、この量を1日1回~数回(例、2回、3回、4回又は8回)投与するのが望ましい。
 本発明の化合物が併用薬物と組み合せて使用される場合には、お互いの剤の量は、それらの剤の反対効果を考えて安全な範囲内で低減できる。
 本発明の併用剤は、毒性が低く、例えば、本発明の化合物、または(および)上記併用薬物を公知の方法に従って、薬理学的に許容される担体と混合して医薬組成物、例えば、錠剤(糖衣錠、フィルムコーティング錠を含む)、散剤、顆粒剤、カプセル剤(ソフトカプセルを含む)、液剤、注射剤、坐剤、徐放剤などとすることができ、それらは、経口的、または非経口的(例、局所、直腸、静脈など)に安全に投与することができる。
Any amount of the concomitant drug can be set as long as side effects are not a problem. The daily dose as a concomitant drug varies depending on the administration subject, administration route, target disease, symptom, etc., for example, when orally administered to a patient with schizophrenia (adult, body weight about 60 kg), Usually, the dose is about 0.1 to about 20 mg / kg body weight, preferably about 0.2 to about 10 mg / kg body weight, more preferably about 0.5 to about 10 mg / kg body weight. It is desirable to administer once to several times (eg, 2, 3, 4 or 8 times).
When the compound of the present invention is used in combination with a concomitant drug, the amount of each agent can be reduced within a safe range in consideration of the opposite effect of those agents.
The concomitant drug of the present invention has low toxicity. For example, the compound of the present invention, or (and) the above concomitant drug is mixed with a pharmacologically acceptable carrier according to a known method, for example, a pharmaceutical composition such as a tablet. (Including sugar-coated tablets and film-coated tablets), powders, granules, capsules (including soft capsules), liquids, injections, suppositories, sustained-release agents, and the like, which can be oral or parenteral (Eg, topical, rectal, intravenous, etc.).
 本発明の併用剤の製造に用いられてもよい薬理学的に許容される担体としては、上記した本発明の医薬組成物に使用されるものと同様のものを使用することができる。
 本発明の併用剤における本発明の化合物と併用薬物との配合比は、投与対象、投与ルート、疾患などにより適宜選択することができる。
 上記併用薬物は、2種以上を適宜の割合で組み合せて用いてもよい。
 併用薬物の投与量は、臨床上用いられている用量を基準として適宜選択することができる。また、本発明の化合物と併用薬物の配合比は、投与対象、投与ルート、対象疾患、症状、組み合わせ等により適宜選択することができる。例えば、投与対象がヒトである場合、本発明の化合物1質量部に対し、併用薬物を0.01~100質量部用いればよい。
 例えば、本発明の併用剤における本発明の化合物の含有量は、製剤の形態によって相違するが、通常製剤全体に対して約0.01~99.9質量%の範囲であり、好ましくは約0.1~50質量%の範囲であり、さらに好ましくは約0.5~20質量%程度の範囲である。
 本発明の併用剤における併用薬物の含有量は、製剤の形態によって相違するが、通常製剤全体に対して約0.01~99.9質量%の範囲であり、好ましくは約0.1~約50質量%の範囲であり、さらに好ましくは約0.5~約20質量%の範囲である。
 本発明の併用剤における担体などの添加剤の含有量は、製剤の形態によって相違するが、通常製剤全体に対して約1~99.99質量%の範囲であり、好ましくは約10~約90質量%の範囲である。
 本発明の化合物、および併用薬物をそれぞれ別々に製剤化する場合も同様の含有量でよい。
 上記したように投与量は種々の条件で変動するので、上記投与量より少ない量で十分な場合もあり、また範囲を超えて投与する必要がある場合もある。
As the pharmacologically acceptable carrier that may be used in the production of the concomitant drug of the present invention, the same carriers as those used for the pharmaceutical composition of the present invention described above can be used.
The compounding ratio of the compound of the present invention and the concomitant drug in the concomitant drug of the present invention can be appropriately selected depending on the administration subject, administration route, disease and the like.
Two or more of the above concomitant drugs may be used in combination at an appropriate ratio.
The dose of the concomitant drug can be appropriately selected based on the clinically used dose. In addition, the compounding ratio of the compound of the present invention and the concomitant drug can be appropriately selected depending on the administration subject, administration route, target disease, symptom, combination and the like. For example, when the administration subject is a human, 0.01 to 100 parts by mass of the concomitant drug may be used per 1 part by mass of the compound of the present invention.
For example, the content of the compound of the present invention in the concomitant drug of the present invention varies depending on the form of the preparation, but is usually in the range of about 0.01 to 99.9% by mass, preferably about 0, based on the whole preparation. The range is from 1 to 50% by mass, and more preferably from about 0.5 to 20% by mass.
The content of the concomitant drug in the concomitant drug of the present invention varies depending on the form of the preparation, but is usually in the range of about 0.01 to 99.9% by mass, preferably about 0.1 to about It is in the range of 50% by weight, more preferably in the range of about 0.5 to about 20% by weight.
The content of an additive such as a carrier in the combination agent of the present invention varies depending on the form of the preparation, but is usually in the range of about 1 to 99.99% by mass, preferably about 10 to about 90% with respect to the whole preparation. It is the range of mass%.
The same content may be used when the compound of the present invention and the concomitant drug are formulated separately.
As described above, since the dosage varies depending on various conditions, an amount smaller than the above dosage may be sufficient, and it may be necessary to administer beyond the range.
[本発明の予防・治療剤の製剤化]
 本発明の医薬は、医薬組成物の形態で投与される。
 本発明の医薬組成物は、本発明の式(I)で表される化合物の少なくとも一つ以上を含んでいればよく、任意に医薬上許容される添加剤と組み合わせてつくられる。より詳細には、賦形剤(例;乳糖、白糖、マンニット、結晶セルロース、ケイ酸、トウモロコシデンプン、バレイショデンプン)、結合剤(例;セルロース類(ヒドロキシプロピルセルロース(HPC)、ヒドロキシプロピルメチルセルロース(HPMC))、結晶セルロース、糖類(乳糖、マンニット、白糖、ソルビトール、エリスリトール、キシリトール、)、デンプン類(トウモロコシデンプン、バレイショデンプン)、α化デンプン、デキストリン、ポリビニルピロリドン(PVP)、マクロゴール、ポリビニルアルコール(PVA))、滑沢剤(例;ステアリン酸マグネシウム、ステアリン酸カルシウム、タルク、カルボキシメチルセルロース)、崩壊剤(例;デンプン類(トウモロコシデンプン、バレイショデンプン)、カルボキシメチルスターチナトリウム、カルメロース、カルメロースカルシウム、クロスカルメロースナトリウム、クロスポピドン)、被膜剤(例;セルロース類(ヒドロキシプロピルセルロース(HPC)、ヒドロキシプロピルメチルセルロース(HPMC)、アミノアルキルメタクリレートコポリマーE、メタクリル酸コポリマーLD)、可塑剤(例;クエン酸トリエチル、マクロゴール)、隠蔽剤(例;酸化チタン)、着色剤、香味剤、防腐剤(例;塩化ベンザルコニウム、パラオキシ安息香酸エステル)、等張化剤(例;グリセリン、塩化ナトリウム、塩化カルシウム、マンニトール、ブドウ糖)、pH調節剤(例;水酸化ナトリウム、水酸化カリウム、炭酸ナトリウム、塩酸、硫酸、リン酸緩衝液などの緩衝液)、安定化剤(例;糖、糖アルコール、キサンタンガム)、分散剤、酸化防止剤(例;アスコルビン酸、ブチルヒドロキシアニソール(BHA)、没食子酸プロピル、dl-α-トコフェロール)、緩衝剤、保存剤(例;パラベン、ベンジルアルコール、塩化ベンザルコニウム)、芳香剤(例;バニリン、l-メントール、ローズ油)、溶解補助剤(例;ポリオキシエチレン硬化ヒマシ油、ポリソルベート80、ポリエチレングリコール、リン脂質コレステロール、トリエタノールアミン)、吸収促進剤(例;グリコール酸ナトリウム、エデト酸ナトリウム、カプリン酸ナトリウム、アシルカルニチン類、リモネン)、ゲル化剤、懸濁化剤、または乳化剤、一般的に用いられる適当な添加剤または溶媒の類を、本発明の化合物と適宜組み合わせて種々の剤形とすることが出来る。
[Formulation of the preventive / therapeutic agent of the present invention]
The medicament of the present invention is administered in the form of a pharmaceutical composition.
The pharmaceutical composition of the present invention only needs to contain at least one of the compounds represented by the formula (I) of the present invention, and is optionally combined with a pharmaceutically acceptable additive. More particularly, excipients (eg; lactose, sucrose, mannitol, crystalline cellulose, silicic acid, corn starch, potato starch), binders (eg; celluloses (hydroxypropylcellulose (HPC), hydroxypropylmethylcellulose ( HPMC)), crystalline cellulose, saccharides (lactose, mannitol, sucrose, sorbitol, erythritol, xylitol), starches (corn starch, potato starch), pregelatinized starch, dextrin, polyvinylpyrrolidone (PVP), macrogol, polyvinyl Alcohol (PVA)), lubricants (eg; magnesium stearate, calcium stearate, talc, carboxymethylcellulose), disintegrants (eg; starches (corn starch, potato starch), carboxymethyl starch sodium, potassium Lumellose, carmellose calcium, croscarmellose sodium, crospovidone), coating agent (eg, celluloses (hydroxypropylcellulose (HPC), hydroxypropylmethylcellulose (HPMC), aminoalkyl methacrylate copolymer E, methacrylic acid copolymer LD), plastic) Agents (e.g. triethyl citrate, macrogol), masking agents (e.g. titanium oxide), colorants, flavoring agents, preservatives (e.g. benzalkonium chloride, paraoxybenzoate), isotonic agents (e.g .; Glycerin, sodium chloride, calcium chloride, mannitol, glucose), pH adjuster (eg; buffer solution such as sodium hydroxide, potassium hydroxide, sodium carbonate, hydrochloric acid, sulfuric acid, phosphate buffer), stabilizer (eg; Sugar, sugar alcohol, xanthan gum), dispersant, antioxidant (eg Asco Binic acid, butylhydroxyanisole (BHA), propyl gallate, dl-α-tocopherol), buffer, preservative (eg, paraben, benzyl alcohol, benzalkonium chloride), fragrance (eg, vanillin, l-menthol) , Rose oil), solubilizers (eg, polyoxyethylene hydrogenated castor oil, polysorbate 80, polyethylene glycol, phospholipid cholesterol, triethanolamine), absorption enhancers (eg, sodium glycolate, sodium edetate, sodium caprate) , Acylcarnitines, limonene), gelling agents, suspending agents, or emulsifiers, commonly used suitable additives or solvents, and various combinations of the compounds of the present invention. I can do it.
 種々の剤形としては、例えば、錠剤、カプセル剤、顆粒剤、散剤、丸剤、エアゾール剤、吸入剤、軟膏剤、貼付剤、坐剤、注射剤、トローチ剤、液剤、酒精剤、懸濁剤、エキス剤、エリキシル剤等があげられる。また、本発明の医薬は、例えば、経口、皮下投与、筋肉内投与、鼻腔内投与、経皮投与、静脈内投与、動脈内投与、神経周囲投与、硬膜外投与、硬膜下腔内投与、脳室内投与、直腸内投与、吸入等により患者に投与し得る。 Various dosage forms include, for example, tablets, capsules, granules, powders, pills, aerosols, inhalants, ointments, patches, suppositories, injections, troches, liquids, spirits, suspensions Agents, extracts, elixirs and the like. The medicament of the present invention is, for example, oral, subcutaneous administration, intramuscular administration, intranasal administration, transdermal administration, intravenous administration, intraarterial administration, perineural administration, epidural administration, intradural administration. It can be administered to patients by intraventricular administration, rectal administration, inhalation and the like.
 本発明の化合物は、通常のカテーテル技法または注入(infusion)を用いることを含む、注射による非経口投与用に製剤化することができる。注射用製剤は、保存剤を添加して、たとえばアンプルまたは多回投与容器で、単位投与形態として提供することができる。これらの製剤は、油性または水性ビヒクル中の懸濁剤、液剤、またはエマルションなどの形態をとることができ、懸濁化剤、安定化剤、および/または分散剤などの製剤化剤を含有することができる。あるいは活性成分は、使用前に、適切なビヒクル、たとえば滅菌発熱物質除去水で再構成するための粉末形態であることもできる。 The compounds of the present invention can be formulated for parenteral administration by injection, including using conventional catheter techniques or infusion. Injectable formulations may be presented as unit dosage forms, for example, in ampoules or multi-dose containers, with the addition of preservatives. These formulations can take the form of suspensions, solutions, or emulsions in oily or aqueous vehicles, and contain formulation agents such as suspending, stabilizing, and / or dispersing agents. be able to. Alternatively, the active ingredient can be in powder form for reconstitution with a suitable vehicle, eg, sterile pyrogen-free water, before use.
 製品溶液が必要とされる場合、製品溶液は患者への経口または非経口投与に必要とされる強度の溶液を生じるのに充分な量で、水(または他の水性媒質)に単離包接複合体を溶解することによって製造できる。これらの化合物は、口腔内で活性成分が放出されるように設計されている、迅速分散投与形態(fddf)に製剤化することができる。これらの製剤は多くの場合、急速溶解性ゼラチンをベースとしたマトリクスを用いて製剤化されている。これらの投与形態はよく知られており、広範な薬物を送達するために用いることができる。ほとんどの迅速分散投与形態は、担体または構造形成剤としてゼラチンを利用する。典型的に、ゼラチンは、包装から取り出すときに破損を防ぐ充分な強度を投与形態に付与するために用いられるが、ひとたび口に入れると、ゼラチンはその投与形態が即時分解することを可能にする。あるいは、同じ効果を得るために、種々のデンプンが用いられる。 If a product solution is required, the product solution is isolated and included in water (or other aqueous medium) in an amount sufficient to produce a solution of the strength required for oral or parenteral administration to the patient. It can be produced by dissolving the complex. These compounds can be formulated into rapidly dispersed dosage forms (fddf) that are designed to release the active ingredient in the oral cavity. These formulations are often formulated using a matrix based on fast dissolving gelatin. These dosage forms are well known and can be used to deliver a wide range of drugs. Most rapid dispersion dosage forms utilize gelatin as a carrier or structure-forming agent. Gelatin is typically used to give a dosage form sufficient strength to prevent breakage when removed from the package, but once in the mouth, gelatin allows the dosage form to break down immediately. . Alternatively, various starches are used to achieve the same effect.
 本発明の化合物はまた、たとえば通常の坐剤基剤、たとえばカカオバターまたは他のグリセリドなどを含有する、坐剤または停留浣腸などの直腸組成物に製剤化することもできる。 The compounds of the invention can also be formulated in rectal compositions such as suppositories or retention enemas, eg containing conventional suppository bases such as cocoa butter or other glycerides.
 鼻腔内投与または吸入による投与の場合、本発明の化合物は、患者によって圧搾されるか、もしくはポンプで送り出されるポンプスプレー容器から溶液または懸濁液の形態で、または適切な噴射剤、たとえば、ジクロロジフルオロメタン、トリクロロフルオロメタン、ジクロロテトラフルオロエタン、二酸化炭素、もしくは他の適切なガスを用いて、加圧式容器もしくはネブライザからエアロゾルスプレー形(aerosol spray presentation)として、好都合に送達される。加圧式エアロゾルの場合、投与単位は、計量された量を送達する弁を提供することによって決定することができる。加圧式容器またはネブライザは、活性化合物の溶液または懸濁液を含有することができる。吸入器または注入器で用いるカプセル剤およびカートリッジ剤(たとえば、ゼラチンから製造される)は、本発明の化合物とラクトースまたはデンプンなどの適切な粉末基剤との混合粉末を含有させて製剤化することができる。 For intranasal administration or administration by inhalation, the compounds of the invention may be administered in the form of a solution or suspension from a pump spray container which is squeezed or pumped by the patient, or a suitable propellant such as dichloromethane. Conveniently delivered as an aerosol spray presentation from a pressurized container or nebulizer using difluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide, or other suitable gas. In the case of a pressurized aerosol, the dosage unit can be determined by providing a valve that delivers a metered amount. A pressurized container or nebulizer can contain a solution or suspension of the active compound. Capsules and cartridges (eg, made from gelatin) for use in an inhaler or insufflator should be formulated containing a mixed powder of a compound of the invention and a suitable powder base such as lactose or starch. Can do.
 平均的成人において上述の状態(たとえば、片頭痛)を治療するためのエアロゾル製剤は、好ましくはエアロゾルの各計量用量または「1吹き(puff)」が本発明の化合物約20mgから約1000mgを含有するように設定される。エアロゾルによる総日用量は、約100mgから約10mgの範囲内となる。投与は1日数回、たとえば2、3、4、または8回、たとえば各回1、2、または3用量の投与であることができる。 Aerosol formulations for treating the above-described conditions (eg, migraine) in the average adult preferably each metered dose or “puff” of the aerosol contains from about 20 mg to about 1000 mg of the compound of the invention. Is set as follows. The total daily dose with an aerosol will be within the range of about 100 mg to about 10 mg. Administration can be several times daily, for example 2, 3, 4 or 8 times, for example 1, 2 or 3 doses each time.
 上述の状態を治療するために、平均的成人に経口、非経口、直腸、または口腔投与される本発明の化合物の提案される日用量は、たとえば1日1から4回投与することのできる単位用量当たり、式(I)の活性成分約0.01mgから約2000mg、好ましくは約0.1mgから約200mgである。 Proposed daily doses of the compounds of the invention administered orally, parenterally, rectally, or buccally to the average adult to treat the above conditions are, for example, units that can be administered 1 to 4 times daily From about 0.01 mg to about 2000 mg, preferably from about 0.1 mg to about 200 mg, of the active ingredient of formula (I) per dose.
[薬理実験例]
 以下に実験例を挙げて、本発明を具体的に説明するが、本発明はこれらによって何ら限定されるものではない。
 以下の薬理実施例1ないし11は、本発明の化合物の有効性を試験する方法を提供する。
[Pharmacological experiment example]
Hereinafter, the present invention will be specifically described with reference to experimental examples, but the present invention is not limited to these examples.
The following Pharmacological Examples 1 to 11 provide methods for testing the effectiveness of the compounds of the present invention.
 薬理実験例1:オレキシン受容体拮抗活性評価
 ヒトオレキシン1受容体(hOX1R)、ヒトオレキシン2受容体(hOX2R)に対する拮抗活性の評価は、以下の方法で行った。hOX1R、hOX2Rを安定発現させたCHO-DXB11細胞を、96ウェル黒色ボトムプレート(Greiner)に40,000細胞/ウェルにて播種し、100units/mLペニシリン-0.1mg/mLストレプトマイシン溶液(Invitrogen)、0.4mg/mL G418(Invitrogen)、10% 非動化ウシ胎児血清(Cell Culture BioscienceもしくはSIGMA)を含むHam‘s F-12培地(SIGMA)中で37℃、5%CO2の条件下で一晩培養した。細胞外液を除去後、1.25mmol/Lプロべネシド(SIGMA)およびFLIPR Calcium 5 assay kit(Molecular Devices)を含むバッファー(20mmol/L HEPES(SIGMA)、Hank’s balanced salt solution(HBSS)(Invitrogen))を200μL添加し、60分間インキュベートした。被験化合物は、10mMとなるようにジメチルスルホキシド(和光純薬)溶液を調製し、終濃度10-10000nM(hOX1R)または終濃度3-3000nM(hOX2R)となるようにアッセイ用バッファー(0.1%ウシ血清アルブミン(BSA),20mmol/L HEPES(SIGMA),HBSS(Invitrogen))で希釈後、25μL添加し5分間インキュベートした。アゴニストであるオレキシン-A(Tocris bioscience)は、終濃度が同日に予め測定したEC80と同じ濃度になるようアッセイ用バッファーで調製し、各ウェルへ25μL添加することで反応を開始させた。反応開始から3分間、Functional Drug Screening System(FDSS:浜松ホトニクス)を用いて、細胞内Ca2+濃度の指標である蛍光値(Ex.480nm/Em.540nm)を経時的に測定した。
 被検化合物のOXR拮抗活性は、OX2RおよびOX1RのIC50値(IC50値が100nmol/L以下の化合物を++、IC50値が100nmol/Lより大きく1000nmol/L以下の本発明の化合物を+、IC50値が1000nmol/Lより大きい化合物を-と表記する)で表2に示す。
Pharmacological Experiment Example 1: Evaluation of orexin receptor antagonistic activity Evaluation of antagonistic activity against human orexin 1 receptor (hOX1R) and human orexin 2 receptor (hOX2R) was performed by the following method. CHO-DXB11 cells stably expressing hOX1R and hOX2R were seeded at 40,000 cells / well in a 96-well black bottom plate (Greiner), 100 units / mL penicillin-0.1 mg / mL streptomycin solution (Invitrogen), In Ham's F-12 medium (SIGMA) containing 0.4 mg / mL G418 (Invitrogen), 10% non-immobilized fetal calf serum (Cell Culture Bioscience or SIGMA) under conditions of 37 ° C. and 5% CO 2 Incubated overnight. After removing the extracellular fluid, a buffer containing 1.25 mmol / L probenecid (SIGMA) and FLIPR Calcium 5 assay kit (Molecular Devices) (20 mmol / L HEPES (SIGMA), Hank's balanced salt solution (HBSS) ( 200 μL of Invitrogen)) was added and incubated for 60 minutes. Prepare a dimethyl sulfoxide (Wako Pure Chemicals) solution so that the test compound is 10 mM, and assay buffer (0.1%) to a final concentration of 10-10000 nM (hOX1R) or a final concentration of 3-3000 nM (hOX2R). After dilution with bovine serum albumin (BSA), 20 mmol / L HEPES (SIGMA), HBSS (Invitrogen)), 25 μL was added and incubated for 5 minutes. The agonist orexin-A (Tocris bioscience) was prepared in assay buffer so that the final concentration was the same as EC80 measured in advance on the same day, and the reaction was started by adding 25 μL to each well. Using a Functional Drug Screening System (FDSS: Hamamatsu Photonics) for 3 minutes from the start of the reaction, the fluorescence value (Ex. 480 nm / Em. 540 nm), which is an index of intracellular Ca 2+ concentration, was measured over time.
OXR antagonistic activity of the test compounds, OX2R and IC 50 values of OXlR (an IC 50 value is ++ with 100 nmol / L the following compounds, an IC 50 value is the compound of greater than 1000 nmol / L or less of the present invention 100 nmol / L + The compounds with IC 50 values greater than 1000 nmol / L are expressed as −).
Figure JPOXMLDOC01-appb-T000035
Figure JPOXMLDOC01-appb-T000035
 薬理実験例2:[Ala11,D-Leu15]-オレキシンB誘発ロコモーター活性評価
 雄性Sprague-Dawleyラットを購入し、飼育施設に到着後、動物を少なくとも一週間の馴化期間をおいて実験に使用する。動物は、温度と湿度をコントロールした実験室で、12時間明暗サイクル下で飼育し、食餌と水を自由摂取させる。
 脳内オレキシンとオレキシン2受容体の相互作用に対する被験化合物の拮抗作用を確認するために、ラットを使用して[Ala11,D-Leu15]-オレキシンB(ADL-OXB)(Tocris bioscience)誘発自発運動量試験を行う。ADL-OXBはヒトオレキシンBの2アミノ酸を置換したペプチドであり、OX1Rと比較してOX2Rに400倍ほど高い親和性を持つ。ADL-OXBを脳室内投与するため、ラットにカニューレ埋め込み手術を行い、回復期間として1週間以上飼育する。回復期間を経て、正しい位置にカニューレが埋め込まれているかを確認するため、予備試験として100ng/5μLのアンジオテンシン2(ペプチド研究所)を脳室内投与し、投与後30分間の飲水量を測定する。飲水量が5gに達したラットのみ本試験に使用する。本試験まで、ラットを測定ケージ内で120分以上馴化させる。馴化後、溶媒又は被験化合物のいずれかを投与し、すぐに測定ケージに戻す。被験化合物の投与30~120分後に、再びラットを飼育ケージより取り出し、溶媒(生理食塩水)またはADL-OXB(3nmol/5μL/ラット)を脳室内投与し、すぐに測定ケージへ戻す。自発運動量測定は赤外線センサーの付いた自発運動量測定チャンバー(室町機械)に入れて行う。自発運動量は、10分ごとにカウントし、ADL-OXB投与後30、60、120分間の累積カウントを各処置群について計算する。全てのデータは平均値と平均値の標準誤差として表す。統計解析は、コントロール群とADL-OXB単独投与群の比較はStudent’s t-test(p<0.05で有意差あり)を使用し、ADL-OXB単独投与群と被験化合物投与群の比較はDunnett’s test(p<0.05で有意差あり)を使用する。
Pharmacological Experiment Example 2: Evaluation of [Ala 11 , D-Leu 15 ] -Orexin B-Induced Locomotor Activity After purchasing male Sprague-Dawley rats and arriving at the breeding facility, the animals were subjected to an experiment with an acclimatization period of at least one week. use. Animals are housed in a laboratory with controlled temperature and humidity under a 12 hour light / dark cycle, with free access to food and water.
[Ala 11 , D-Leu 15 ] -Orexin B (ADL-OXB) (Tocris bioscience) induction using rats to confirm the antagonism of test compounds against the interaction of orexin and orexin 2 receptor in the brain Perform spontaneous exercise test. ADL-OXB is a peptide in which two amino acids of human orexin B are substituted, and has a 400 times higher affinity for OX2R than OX1R. In order to administer ADL-OXB intracerebroventricularly, rats are cannulated and kept for at least 1 week as a recovery period. In order to confirm whether the cannula is implanted at the correct position after the recovery period, 100 ng / 5 μL of angiotensin 2 (Peptide Institute) is administered intraventricularly as a preliminary test, and the amount of water consumed for 30 minutes after the administration is measured. Only rats that have reached 5 g of drinking water are used in this study. Rats are acclimated for at least 120 minutes in the measurement cage until this test. After acclimatization, either vehicle or test compound is administered and immediately returned to the measurement cage. 30 to 120 minutes after administration of the test compound, the rat is again taken out from the housing cage, and the solvent (saline) or ADL-OXB (3 nmol / 5 μL / rat) is administered into the ventricle and immediately returned to the measurement cage. Spontaneous momentum is measured in a spontaneous momentum measurement chamber (Muromachi Kikai) equipped with an infrared sensor. Spontaneous exercise is counted every 10 minutes, and cumulative counts for 30, 60, 120 minutes after ADL-OXB administration are calculated for each treatment group. All data are expressed as mean and standard error of the mean. For statistical analysis, comparison between the control group and ADL-OXB single administration group was performed using Student's t-test (significant difference at p <0.05), and comparison between ADL-OXB single administration group and test compound administration group Uses Dunnett's test (significantly different at p <0.05).
 薬理実験例3:睡眠脳波測定試験 
 雄性Sprague-Dawleyラットを購入し、飼育施設に到着後、動物を少なくとも一週間の馴化期間をおいて実験に使用する。動物は、温度と湿度をコントロールした実験室で、12時間明暗サイクル下で飼育し、食餌と水を自由摂取させる。
 睡眠への効果を確認するためにラットにおける睡眠脳波試験を実施する。脳波(Electroencephalogram:EEG)および筋電図(Electromyogram:EMG)測定のためにラットにEEGおよびEMG電極埋め込み手術を行ない、回復期間として1週間以上飼育する。溶媒または被験化合物を投与した後、EEGおよびEMG信号を6時間ないし12時間記録する。
 解析は自動解析ソフトであるSleepSign(登録商標)(キッセイコムテック)を用いて、脳波周波数と筋電図の活動信号を分析し、覚醒、REM睡眠、NREM睡眠の3ステージの何れかを分類させる。各ステージの累積時間を計算し、覚醒時間の減少、総睡眠時間の上昇(REM睡眠+NREM睡眠)から被験化合物の睡眠作用を確認する。また総睡眠時間に対するREM睡眠のパーセンテージを算出し、コントロール群の結果と比較することにより生理的な睡眠パターンを示しているか検討する。
Pharmacological experiment example 3: Sleep EEG measurement test
After purchasing male Sprague-Dawley rats and arriving at the breeding facility, the animals are used for experiments with an acclimatization period of at least one week. Animals are housed in a laboratory with controlled temperature and humidity under a 12 hour light / dark cycle, with free access to food and water.
In order to confirm the effect on sleep, a sleep electroencephalogram test in rats is performed. Rats are subjected to EEG and EMG electrode implantation surgery for electroencephalogram (EEG) and electromyogram (EMG) measurement, and are kept for at least 1 week as a recovery period. After administration of vehicle or test compound, EEG and EMG signals are recorded for 6-12 hours.
The analysis is performed by using an automatic analysis software SleepSign (registered trademark) (Kissei Comtech) to analyze an electroencephalogram frequency and an electromyogram activity signal to classify one of three stages of arousal, REM sleep, and NREM sleep. The cumulative time of each stage is calculated, and the sleep action of the test compound is confirmed from the decrease in the awakening time and the increase in the total sleep time (REM sleep + NREM sleep). Moreover, the percentage of REM sleep with respect to the total sleep time is calculated, and it is examined whether a physiological sleep pattern is shown by comparing with the result of the control group.
 薬理実験例4:溶解性試験
(1)DMSO析出溶解性(Kinetic Solubility)
 本発明の化合物の10mMのDMSO溶液を最終濃度100μMとなるように50mMリン酸緩衝液(pH7.4)に添加する。その溶液を室温で1.5時間、600rpmにて撹拌しながらインキュベーションした後、フィルタープレート(MultiScreenHTS-PCFフィルタープレート(MerckMillipore))でろ過し、プレートリーダー(Powerscan HT(大日本製薬))を用いて、ろ液の吸光度を最大吸収波長で測定する。同時に、試験化合物の既知濃度(1、3、10、30、100μM)を添加したDMSO溶液を検量線標準溶液として各々の標準溶液吸光度を測定し、検量線を作成する。ろ液および標準溶液の吸光度値より化合物の溶解度(μM)を算出する。
(2)結晶溶解性(Thermodynamic Solubility)
 本発明の化合物を1mg/mLとなるように溶媒(例えば、水、緩衝液)に添加する。その溶液を25℃または37℃で24時間1000rpmにて撹拌しながらインキュベーションした後、フィルタープレートでろ過する。ろ液をHPLCにて分析し、最大吸収波長にてピークを検出し、ピーク面積を測定する。同様に試験化合物の既知濃度(例えば、0.01、0.03、0.1、0.3、1、3、10μg/mL)を添加した溶液(例えば、DMSO溶液、1,4-ジオキサン溶液、メタノール溶液)を検量線標準溶液として各々のピーク面積を測定し、検量線のピーク面積より化合物の溶解度(μg/mL)を算出する。
Pharmacological Experiment Example 4: Solubility Test (1) DMSO Precipitation Solubility (Kinetic Solubility)
A 10 mM DMSO solution of the compound of the present invention is added to 50 mM phosphate buffer (pH 7.4) to a final concentration of 100 μM. The solution was incubated at 600 rpm with stirring at room temperature for 1.5 hours, then filtered through a filter plate (MultiScreen HTS- PCF filter plate (Merck Millipore)), and using a plate reader (Powerscan HT (Dainippon Pharmaceutical)). Then, the absorbance of the filtrate is measured at the maximum absorption wavelength. At the same time, the absorbance of each standard solution is measured using a DMSO solution to which a known concentration (1, 3, 10, 30, 100 μM) of the test compound is added as a calibration curve standard solution, and a calibration curve is created. The solubility (μM) of the compound is calculated from the absorbance values of the filtrate and standard solution.
(2) Crystal solubility (Thermodynamic Solubility)
The compound of the present invention is added to a solvent (eg, water, buffer) so as to be 1 mg / mL. The solution is incubated with stirring at 1000 rpm for 24 hours at 25 ° C. or 37 ° C. and then filtered through a filter plate. The filtrate is analyzed by HPLC, the peak is detected at the maximum absorption wavelength, and the peak area is measured. Similarly, a solution (eg, DMSO solution, 1,4-dioxane solution) to which a known concentration of the test compound (eg, 0.01, 0.03, 0.1, 0.3, 1, 3, 10 μg / mL) is added. , Methanol solution) is used as a standard curve standard solution to measure each peak area, and the solubility (μg / mL) of the compound is calculated from the peak area of the standard curve.
 薬理実験例5:代謝安定性試験
(1)代謝安定性試験1
 本発明の化合物の10mMのDMSO溶液を最終濃度1μMとなるように肝ミクロソーム溶液(ヒト、ラット、マウス、イヌまたはサル;XenoTech)、NADPH生成溶液(β-NADP、Glucose-6-Phosphate、G-6-PDH(Y)、MgCl2を含む水)に添加する。その溶液を37℃で5、10、20または30分間インキュベートした後、アセトニトリルで反応停止する。反応液をフィルタープレート(MultiScreenHTS-HVプレート(MerckMillipore))で遠心ろ過し、高速液体クロマトグラム/マススペクトロメトリーを用いて、ろ液中の試験化合物を測定する。同様に反応時間0分のサンプルをコントロールとして測定し、各時点における残存率(%)を算出する。反応時間を横軸、残存率を縦軸としてプロットし、その傾きからクリアランス(μL/min/mg protein)を算出する。
(2)代謝安定性試験2
 本発明の化合物の10mMのDMSO溶液を最終濃度1μMとなるように肝細胞懸濁液(ヒト;GIBCO、ラット、イヌ、サル;XenoTech)に添加する。その溶液を37℃で5、15、30、45、60、120分間インキュベートした後、アセトニトリルで反応停止する。反応液をフィルタープレート(MultiScreenHTS-HVプレート(MerckMillipore))で遠心ろ過し、高速液体クロマトグラム/マススペクトロメトリーを用いて、ろ液中の試験化合物を測定する。同様に反応時間0分のサンプルをコントロールとして測定し、肝細胞反応サンプルとコントロールの比より、各時点における残存率(%)を算出する。反応時間を横軸、残存率を縦軸としてプロットし、その傾きからクリアランス(μL/min/mg protein)を算出する。
Pharmacological experiment example 5: Metabolic stability test (1) Metabolic stability test 1
Liver microsomal solution (human, rat, mouse, dog or monkey; XenoTech), NADPH generating solution (β-NADP, Glucose-6-phosphate, G- 6-PDH (Y), water containing MgCl 2 ). The solution is incubated at 37 ° C. for 5, 10, 20 or 30 minutes and then quenched with acetonitrile. The reaction solution is centrifuged through a filter plate (MultiScreen HTS- HV plate (Merck Millipore)), and the test compound in the filtrate is measured using high performance liquid chromatogram / mass spectrometry. Similarly, a sample with a reaction time of 0 minutes is measured as a control, and the residual rate (%) at each time point is calculated. The reaction time is plotted on the horizontal axis and the residual ratio is plotted on the vertical axis, and the clearance (μL / min / mg protein) is calculated from the slope.
(2) Metabolic stability test 2
A 10 mM DMSO solution of the compound of the present invention is added to a hepatocyte suspension (human; GIBCO, rat, dog, monkey; XenoTech) to a final concentration of 1 μM. The solution is incubated at 37 ° C. for 5, 15, 30, 45, 60, 120 minutes and then quenched with acetonitrile. The reaction solution is centrifuged through a filter plate (MultiScreenHTS-HV plate (Merck Millipore)), and the test compound in the filtrate is measured using high performance liquid chromatogram / mass spectrometry. Similarly, a sample with a reaction time of 0 minutes is measured as a control, and the residual rate (%) at each time point is calculated from the ratio of the hepatocyte reaction sample and the control. The reaction time is plotted on the horizontal axis and the residual ratio is plotted on the vertical axis, and the clearance (μL / min / mg protein) is calculated from the slope.
 薬理実験例6:パッチクランプ法によるhERG阻害試験
 hERG(human ether-a-go-go related gene)チャネルに対する作用を全自動パッチクランプシステム(QPatch HT(Sophion Bioscience))を用いて測定する。細胞(CHO hERG DUO(B’SYS))のhERG IKr電流を確認するため、膜電位を-80mVに保持し、-50mV、0.02秒間および20mV、4.8秒間の脱分極パルスに続く-50mV、5秒間の再分極パルスを15秒に1回の頻度で与える。試験化合物のhERGチャネルに対する作用は、再分極パルスによって誘導されるテール電流の変化によって確認する。測定は室温で行う。hERGチャネル阻害率は、試験化合物添加前のテール電流ピーク値に対する添加後4分のテール電流の減少率(抑制率)として算出する。
 この抑制率を算出することにより、薬物によるQT延長とそれに続く致死的な副作用(心室頻拍や突然死など)を誘発する可能性が示される。
Pharmacological Experiment Example 6 hERG Inhibition Test by Patch Clamp Method The effect on hERG (human ether-a-go related gene) channels is measured using a fully automatic patch clamp system (QPatch HT (Sophion Bioscience)). To confirm the hERG I Kr current of the cells (CHO hERG DUO (B'SYS)), the membrane potential is held at −80 mV, followed by a depolarization pulse of −50 mV, 0.02 seconds and 20 mV, 4.8 seconds. -50 mV, 5 seconds of repolarization pulse given once every 15 seconds. The effect of the test compound on the hERG channel is confirmed by the tail current change induced by the repolarization pulse. The measurement is performed at room temperature. The hERG channel inhibition rate is calculated as the reduction rate (suppression rate) of the tail current 4 minutes after the addition relative to the tail current peak value before the addition of the test compound.
By calculating this suppression rate, the possibility of inducing QT prolongation by drugs and subsequent fatal side effects (such as ventricular tachycardia and sudden death) is shown.
 薬理実験例7:ファーマコキネティクス(PK)試験
(1)ラットカセットPK試験
 本発明の化合物を7あるいは8週齢の雄性CD(SD)IGS Jclに1mg/kg(投与溶媒は、DMSO:Tween80:超純水=1:1:8、10mL/kg)で経口単回投与した後、0.5、1、2、4時間後に腹大静脈より採血する。血液を遠心分離(3000rpm、15分間、4℃)して得られた血漿を用いて、高速液体クロマトグラム/マススペクトロメトリーにて、血漿中の試験化合物を測定する。同様に試験化合物の既知濃度(0.01、0.02、0.05、0.1、0.2、0.5、1μg/mL)を添加した標準溶液を測定し、作成した検量線より血漿中濃度(μg/mL)を算出し、最高血漿中濃度をCmax(μg/mL)とする。
(2)PK試験
 本発明の化合物を動物(約7から8週齢の雄性Crl:CD(SD)ラット、もしくは、約4カ年齢の雄性ビーグル犬、もしくは、約6カ年齢の雄性カニクイザル)に1mg/kg(投与溶媒は、Dimethylacetamide:Tween80:超純水=4:4:2、1mL/kg)で静脈内投与あるいは10mg/kg(投与溶媒は、0.5% Methylcellulose、5mL/kg)で経口投与した後、0.083、0.25、0.5、1、2,4、8、24時間後に頚静脈より採血する。血液を遠心分離(3000rpm、15分間、4℃)して得られた血漿を用いて、高速液体クロマトグラム/マススペクトロメトリーにて、血漿中の試験化合物を測定する。同様に試験化合物の既知濃度(0.001、0.002、0.005、0.01、0.02、0.05、0.1、0.2、0.5、1、2、10μg/mL)を添加した標準溶液を測定し、作成した検量線より血漿中濃度(μg/mL)を算出し、最高血漿中濃度をCmax(μg/mL)とする。
Pharmacological Experiment Example 7: Pharmacokinetics (PK) Test (1) Rat Cassette PK Test The compound of the present invention was added to 7 or 8 weeks old male CD (SD) IGS Jcl at 1 mg / kg (administration solvent was DMSO: Tween 80: Ultrapure water = 1: 1: 8, 10 mL / kg), and blood is collected from the abdominal vena cava 0.5, 1, 2, and 4 hours later. Using plasma obtained by centrifuging blood (3000 rpm, 15 minutes, 4 ° C.), the test compound in plasma is measured by high performance liquid chromatogram / mass spectrometry. Similarly, a standard solution to which a known concentration of a test compound (0.01, 0.02, 0.05, 0.1, 0.2, 0.5, 1 μg / mL) was added was measured, and a calibration curve was prepared. The plasma concentration (μg / mL) is calculated, and the maximum plasma concentration is defined as Cmax (μg / mL).
(2) PK test The compound of the present invention was applied to animals (male Crl: CD (SD) rats of about 7 to 8 weeks of age, male beagle dogs of about 4 years of age, or male cynomolgus monkeys of about 6 years of age). 1 mg / kg (administration solvent is Dimethylacetamide: Tween 80: Ultrapure water = 4: 4: 2, 1 mL / kg) or 10 mg / kg (administration solvent is 0.5% Methylcellulose, 5 mL / kg) After oral administration, blood is collected from the jugular vein 0.083, 0.25, 0.5, 1, 2, 4, 8, and 24 hours later. Using plasma obtained by centrifuging blood (3000 rpm, 15 minutes, 4 ° C.), the test compound in plasma is measured by high performance liquid chromatogram / mass spectrometry. Similarly, known concentrations of test compounds (0.001, 0.002, 0.005, 0.01, 0.02, 0.05, 0.1, 0.2, 0.5, 1, 2, 10 μg / (mL) is measured, the plasma concentration (μg / mL) is calculated from the prepared calibration curve, and the maximum plasma concentration is defined as Cmax (μg / mL).
 薬理実験例8:タンパク結合試験
 本発明の化合物の10mMのDMSO溶液を最終濃度10μMとなるように正常血漿(ヒト、ラット、イヌ、サル)に添加する。簡易平衡透析装置(HTD96b Complete Unit(HTDialysis))にて37℃で4時間透析した後、透析膜の内側(血漿側)溶液および外側(PBS側)溶液中の試験化合物濃度を、高速液体クロマトグラム/マススペクトロメトリーを用いて測定する。PBS側と血漿側の比から非結合分率(%)を算出し、100-非結合分率(%)より蛋白結合率(%)を算出する。
 薬理実験例9:ファーマコキネティクス試験における各種パラメータの算出
  ラット、イヌ、サルの各動物種におけるPK試験(上記薬理実験例7)によって得られた血漿中濃度の時間推移についてモデル非依存的解析を行い、全身クリアランスCLtot(L/kg/hr)、定常状態における分布容積Vdss(L/kg)、血漿中濃度―時間曲線下面積AUC(μg・hr/mL)、半減期T1/2(hr)を算出する。また、静脈内投与時のAUCと経口投与時のAUCを比較してバイオアベイラビリティを算出する。
Pharmacological Experiment Example 8: Protein Binding Test A 10 mM DMSO solution of the compound of the present invention is added to normal plasma (human, rat, dog, monkey) to a final concentration of 10 μM. After dialyzing for 4 hours at 37 ° C. with a simple equilibrium dialysis machine (HTD96b Complete Unit (HTDialissis)), the test compound concentrations in the inner (plasma side) solution and outer (PBS side) solution of the dialysis membrane were measured using a high-performance liquid chromatogram. / Measure using mass spectrometry. The non-binding fraction (%) is calculated from the ratio between the PBS side and the plasma side, and the protein binding rate (%) is calculated from 100−non-binding fraction (%).
Pharmacological experiment example 9: Calculation of various parameters in pharmacokinetics test Model-independent analysis of the time course of plasma concentration obtained by PK test (the above pharmacological experiment example 7) in rat, dog and monkey animal species Systemic clearance CLtot (L / kg / hr), distribution volume Vdss in steady state (L / kg), plasma concentration-time curve area AUC (μg · hr / mL), half-life T1 / 2 (hr) Is calculated. In addition, the bioavailability is calculated by comparing the AUC at the time of intravenous administration with the AUC at the time of oral administration.
 薬理実験例10:ヒトにおける薬物動態パラメータの予測
  上記薬理実験例5、7または8等に記載の方法にて得られた、動物のファーマコキネティクス試験における各種パラメータ、インビトロ試験における代謝安定性、タンパク結合率などのパラメータを用い、アロメトリックスケーリングによる方法またはIVIVE(in vitro/in vivo extrapolation)法などの当業者に知られた方法により、ヒトにおける薬物動態パラメータを予測する。
Pharmacological experiment example 10: Prediction of pharmacokinetic parameters in humans Various parameters in animal pharmacokinetic tests, metabolic stability in in vitro tests, proteins obtained by the method described in pharmacological experiment examples 5, 7 or 8 above Using parameters such as binding rate, pharmacokinetic parameters in humans are predicted by methods known to those skilled in the art such as allometric scaling or IVIVE (in vitro / in vivo extrapolation).
 薬理実験例11:安全性試験
 本発明の化合物をマウスまたはラットに単回で経口投与し、死亡例は認められず、目立った行動異常も観察されないことにより、本発明の化合物の安全性が示される。
Pharmacological experiment example 11: Safety test The compound of the present invention is orally administered to a mouse or rat once, and no deaths are observed, and no remarkable behavioral abnormality is observed. It is.
 以上の結果より、本発明の化合物は、優れたオレキシン受容体拮抗作用を有することが示された。更に、ラットを用いた[Ala11,D-Leu15]-オレキシンB誘発自発運動量亢進に対する拮抗作用試験によって、オレキシン拮抗作用が示され、睡眠脳波測定試験によって、睡眠作用が示される。また、安全性試験において何ら異常が認められず、本発明の低い毒性が示される。更に、本発明の化合物は、上記の試験を行うことにより、溶解性、代謝安定性、薬物動態、hERGチャネル阻害作用の回避等の1つの点において良好であることが確認される。 From the above results, it was shown that the compound of the present invention has an excellent orexin receptor antagonistic action. Furthermore, orexin antagonism is shown by an antagonism test against [Ala 11 , D-Leu 15 ] -orexin B-induced locomotor activity enhancement using rats, and sleep action is shown by a sleep electroencephalogram measurement test. In addition, no abnormality is observed in the safety test, indicating the low toxicity of the present invention. Furthermore, it is confirmed that the compound of the present invention is good in one point such as solubility, metabolic stability, pharmacokinetics, and avoidance of hERG channel inhibitory effect by conducting the above test.
 従って、本発明の化合物は、オレキシン受容体拮抗剤として、睡眠障害(不眠症、概日リズム睡眠障害、睡眠時随伴症等)、精神疾患(うつ病、不安障害、双極性障害、注意欠陥多動性障害、自閉症、自閉症スペクトラム障害、薬物依存症等)、神経変性疾患(アルツハイマー病等)、記憶障害(認知症等)および摂食障害(過食症等)等の疾患の予防及び/または治療剤に用いることが期待される。
 本発明の化合物は、以下に示す各種疾患に対して有望な予防、あるいは治療効果を示すことが期待される。具体的には、不眠症、概日リズム睡眠障害、睡眠時随伴症、うつ病、不安障害、双極性障害、注意欠陥多動性障害、自閉症、自閉症スペクトラム障害、薬物依存症、アルツハイマー病、認知症、過食症等に対して有望な治療効果が期待できる。
Therefore, the compounds of the present invention are orexin receptor antagonists, such as sleep disorders (insomnia, circadian rhythm sleep disorders, sleep-related complications, etc.), mental disorders (depression, anxiety disorders, bipolar disorders, attention deficits Prevention of diseases such as mobility disorders, autism, autism spectrum disorders, drug addiction, etc.), neurodegenerative diseases (such as Alzheimer's disease), memory disorders (such as dementia), and eating disorders (such as bulimia) And / or expected to be used in therapeutic agents.
The compounds of the present invention are expected to show promising preventive or therapeutic effects for various diseases shown below. Specifically, insomnia, circadian rhythm sleep disorder, sleep-related comorbidities, depression, anxiety disorder, bipolar disorder, attention deficit hyperactivity disorder, autism, autism spectrum disorder, drug addiction, Promising therapeutic effects can be expected for Alzheimer's disease, dementia, bulimia and the like.
 次に、本発明をさらに詳細に説明するために実施例、製造例をあげるが、これらの例は単なる実施であって、本発明を限定するものではなく、また本発明の範囲を逸脱しない範囲で変化させてもよい。 Next, in order to describe the present invention in more detail, examples and production examples will be given. However, these examples are merely implementations, and do not limit the present invention, and do not depart from the scope of the present invention. It may be changed with.
 核磁気共鳴スペクトル(NMR)の測定には、JEOL JNM-ECX400 FT-NMR(日本電子)またはJEOL JNM-ECX300 FT-NMR(日本電子)を用いた。
 液体クロマトグラフィー-質量分析スペクトル(LC -Mass)は以下のいずれかの方法で測定した。[UPLC]Waters UPLC-ZQ MSシステム(Waters)とMGIII-Hカラム(2.1mm×5cm、3μm)(資生堂)を用い、移動相は、メタノール:0.05%トリフルオロ酢酸水溶液=5:95(0分)~100:0(1分)~100:0(2分)のグラジエント条件を用いた。[LCMS]Waters FractionLynx MSシステム(Waters)とSunFireカラム(4.6mm×5cm、5μm)(Waters)を用い、移動相は、メタノール:0.05%酢酸水溶液=10:90(0分)~100:0(5分)~100:0(7分)のグラジエント条件を用いた。分取系には化合物により適宜変更したグラジエント条件を用いた。
For the measurement of nuclear magnetic resonance spectrum (NMR), JEOL JNM-ECX400 FT-NMR (JEOL) or JEOL JNM-ECX300 FT-NMR (JEOL) was used.
Liquid chromatography-mass spectrometry spectrum (LC-Mass) was measured by one of the following methods. [UPLC] Waters UPLC-ZQ MS system (Waters) and MGIII-H column (2.1 mm × 5 cm, 3 μm) (Shiseido) were used, and the mobile phase was methanol: 0.05% trifluoroacetic acid aqueous solution = 5: 95. Gradient conditions from (0 minutes) to 100: 0 (1 minute) to 100: 0 (2 minutes) were used. [LCMS] Using a Waters FractionLynx MS system (Waters) and a SunFire column (4.6 mm × 5 cm, 5 μm) (Waters), the mobile phase was methanol: 0.05% acetic acid aqueous solution = 10: 90 (0 min) to 100 : Gradient conditions from 0 (5 minutes) to 100: 0 (7 minutes) were used. For the preparative system, gradient conditions appropriately changed depending on the compound were used.
 1H-NMRデータ中、NMRシグナルのパターンで、sはシングレット、dはダブレット、tはトリプレット、qはカルテット、mはマルチプレット、brはブロード、Jはカップリング定数、Hzはヘルツ、CDCl3は重クロロホルム、DMSO-d6は重ジメチルスルホキシドを意味する。1H-NMRデータ中、水酸基、アミノ基、カルボキシル基のプロトン等、ブロードバンドであるため確認ができないシグナルについては、データに記載していない。
 LC-Massデータは、各実施例において「UPLC」、「LCMS」として示した。LC-Massデータ中、[M+H]+、[M+Na]+は分子イオンピークを意味する。
1 H-NMR data, NMR signal pattern, s is singlet, d is doublet, t is triplet, q is quartet, m is multiplet, br is broad, J is coupling constant, Hz is Hertz, CDCl 3 Means deuterated chloroform, DMSO-d 6 means deuterated dimethyl sulfoxide. In the 1 H-NMR data, signals that cannot be confirmed due to broadband, such as protons of hydroxyl groups, amino groups, and carboxyl groups, are not described in the data.
The LC-Mass data is shown as “UPLC” and “LCMS” in each example. In the LC-Mass data, [M + H] + and [M + Na] + mean molecular ion peaks.
 実施例及び製造例中の「室温」は、実験室内の温度、通常は20±15℃の温度を示すものとする。 “Room temperature” in the examples and production examples represents the temperature in the laboratory, usually 20 ± 15 ° C.
(製造例1)2-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン塩酸塩(中間体1)の合成
<工程1>tert-ブチル 5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体1-1)の合成
 tert-ブチル 2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(0.50g)と炭酸セシウム(1.5g)のDMF(24mL)混合液に2-クロロ-4,6-ジメチルピリミジン(0.37g)を加え、90℃にて1日撹拌した。反応溶液に水を加えた後、酢酸エチルで抽出を行った。有機層を水で洗浄し、無水硫酸ナトリウムで乾燥した。減圧下溶媒を留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、ヘプタン:酢酸エチル=87:13~75:25)にて精製し、標記化合物(0.50g)を無色液体として得た。
UPLC:341[M+Na]+(保持時間0.93分)
1H-NMR (CDCl3, 400MHz) δ: 6.26 (1H, s), 5.08-5.01 (1H, m), 4.42-4.23 (1H, m), 3.84-3.75 (1H, m), 3.65-3.57 (2H, m), 3.51-3.43 (1H, m), 2.27 (6H, s), 2.10-1.95 (2H, m), 1.88-1.72 (2H, m), 1.47-1.44 (9H, m).
Production Example 1 Synthesis of 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane hydrochloride (intermediate 1) <Step 1> tert-butyl 5 Synthesis of — (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 1-1) tert-butyl 2,5-diazabicyclo [ 2.2.2] 2-Chloro-4,6-dimethylpyrimidine (0.37 g) was added to a DMF (24 mL) mixture of octane-2-carboxylate (0.50 g) and cesium carbonate (1.5 g). , And stirred at 90 ° C. for 1 day. Water was added to the reaction solution, followed by extraction with ethyl acetate. The organic layer was washed with water and dried over anhydrous sodium sulfate. The residue obtained by evaporating the solvent under reduced pressure was purified by column chromatography (silica gel, heptane: ethyl acetate = 87: 13 to 75:25) to obtain the title compound (0.50 g) as a colorless liquid. .
UPLC: 341 [M + Na] + (retention time 0.93 minutes)
1 H-NMR (CDCl 3 , 400 MHz) δ: 6.26 (1H, s), 5.08-5.01 (1H, m), 4.42-4.23 (1H, m), 3.84-3.75 (1H, m), 3.65-3.57 ( 2H, m), 3.51-3.43 (1H, m), 2.27 (6H, s), 2.10-1.95 (2H, m), 1.88-1.72 (2H, m), 1.47-1.44 (9H, m).
<工程2>2-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン塩酸塩(中間体1)の合成
 tert-ブチル 5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体1-1)(0.50g)に4規定塩酸-ジオキサン溶液(3.0mL)を加え、室温にて17時間撹拌した。減圧下溶媒を留去し、標記化合物(0.40g)を白色固体として得た。
UPLC:219[M+H]+(保持時間0.51分)
1H-NMR (CDCl3, 400MHz) δ: 10.42 (1H, br s), 10.09 (1H, br s), 6.56 (1H, s), 5.43-5.35 (1H, m), 4.66-4.57 (1H, m), 4.46-4.38 (1H, m), 4.24-4.13 (1H, m), 3.80-3.60 (2H, m), 2.78-2.54 (7H, m), 2.22-2.09 (2H, m), 2.02-1.89 (1H, m).
<Step 2> Synthesis of 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane hydrochloride (intermediate 1) tert-butyl 5- (4,6 -Dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 1-1) (0.50 g) in 4N hydrochloric acid-dioxane solution (3.0 mL) ) And stirred at room temperature for 17 hours. The solvent was distilled off under reduced pressure to obtain the title compound (0.40 g) as a white solid.
UPLC: 219 [M + H] + (retention time 0.51 minutes)
1 H-NMR (CDCl 3 , 400 MHz) δ: 10.42 (1H, br s), 10.09 (1H, br s), 6.56 (1H, s), 5.43-5.35 (1H, m), 4.66-4.57 (1H, m), 4.46-4.38 (1H, m), 4.24-4.13 (1H, m), 3.80-3.60 (2H, m), 2.78-2.54 (7H, m), 2.22-2.09 (2H, m), 2.02- 1.89 (1H, m).
(製造例2)2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリル(中間体2)の合成
<工程1>2-クロロ-3-メトキシイソニコチン酸(中間体2-1)の合成
 2-クロロ-3-メトキシピリジン(5.2g)のTHF(100mL)溶液を-78℃に冷却した後、1.6Mのn-BuLiヘキサン溶液(25mL)を徐々に加え、1時間撹拌した。その後、反応溶液にドライアイスを加え、徐々に室温まで昇温した。飽和塩化アンモニウム水溶液を加えてpH=6とした後に、酢酸エチルで逆抽出を行った。水層に1規定塩酸を加えてpH=2とし、析出した固体を濾取した後、減圧下乾燥を行い、標記化合物(6.3g)を無色固体として得た。
UPLC:188[M+H]+(保持時間0.71分)
1H-NMR (DMSO-d6, 300MHz) δ: 8.26 (1H, d, J = 5 Hz), 7.62 (1H, d, J = 5 Hz), 3.87 (3H, s).
Production Example 2 Synthesis of 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) -3-methoxyisonicotinonitrile (Intermediate 2) <Step 1> 2-Chloro-3 Synthesis of -methoxyisonicotinic acid (intermediate 2-1) A solution of 2-chloro-3-methoxypyridine (5.2 g) in THF (100 mL) was cooled to -78 ° C, and then 1.6 M n-BuLi hexane The solution (25 mL) was added slowly and stirred for 1 hour. Thereafter, dry ice was added to the reaction solution, and the temperature was gradually raised to room temperature. A saturated aqueous ammonium chloride solution was added to adjust to pH = 6, and then back extraction was performed with ethyl acetate. 1N Hydrochloric acid was added to the aqueous layer to adjust to pH = 2, and the precipitated solid was collected by filtration and dried under reduced pressure to give the title compound (6.3 g) as a colorless solid.
UPLC: 188 [M + H] + (retention time 0.71 minutes)
1 H-NMR (DMSO-d 6 , 300 MHz) δ: 8.26 (1H, d, J = 5 Hz), 7.62 (1H, d, J = 5 Hz), 3.87 (3H, s).
<工程2>2-クロロ-3-メトキシイソニコチンアミド(中間体2-2)の合成
 2-クロロ-3-メトキシイソニコチン酸(中間体2-1)(6.8g)、ピリジン(15mL)、炭酸水素アンモニウム(14g)の1,4-ジオキサン(100mL)混合溶液に二炭酸ジ-tert-ブチル(13mL)を加え、室温で1時間攪拌した。反応溶液に0.1規定塩酸水溶液を加えて中和した後、酢酸エチルで2回、抽出を行った。有機層を無水硫酸ナトリウムで乾燥し、減圧下溶媒を留去して、標記化合物の粗生成物(7.5g)を無色固体として得た。
UPLC:187[M+H]+(保持時間0.61分)
<Step 2> Synthesis of 2-chloro-3-methoxyisonicotinamide (Intermediate 2-2) 2-Chloro-3-methoxyisonicotinic acid (Intermediate 2-1) (6.8 g), pyridine (15 mL) To a mixed solution of ammonium hydrogen carbonate (14 g) in 1,4-dioxane (100 mL) was added di-tert-butyl dicarbonate (13 mL), and the mixture was stirred at room temperature for 1 hour. The reaction solution was neutralized with 0.1N aqueous hydrochloric acid solution, and extracted twice with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to obtain a crude product (7.5 g) of the title compound as a colorless solid.
UPLC: 187 [M + H] + (retention time 0.61 minutes)
<工程3>2-クロロ-3-メトキシイソニコチノニトリル(中間体2-3)の合成
 製造例2の工程2で得た2-クロロ-3-メトキシイソニコチンアミド(中間体2-2)の粗生成物(7.4g)、トリエチルアミン(15mL)のジクロロメタン(66mL)混合溶液を0℃に冷却した後、無水トリフルオロ酢酸(7.5mL)を徐々に加え、反応溶液を0℃で30分間撹拌した。反応溶液を室温に昇温した後に、飽和炭酸水素ナトリウム水溶液を加え、ジエチルエーテルで抽出した。有機層を無水硫酸ナトリウムで乾燥した後、減圧下溶媒を留去して得られた残渣を、カラムクロマトグラフィー(NHシリカゲル、ヘプタン:酢酸エチル=100:0~90:10)にて精製し、標記化合物(4.8g)を無色固体として得た。
UPLC:169[M+H]+(保持時間0.83分)
1H-NMR (CDCl3, 300MHz) δ: 8.24 (1H, d, J = 5 Hz), 7.42 (1H, d, J = 5 Hz), 4.17 (3H, s).
<Step 3> Synthesis of 2-chloro-3-methoxyisonicotinonitrile (Intermediate 2-3) 2-Chloro-3-methoxyisonicotinamide (Intermediate 2-2) obtained in Step 2 of Production Example 2 A crude product of 7.4 g, triethylamine (15 mL) in dichloromethane (66 mL) mixed solution was cooled to 0 ° C., trifluoroacetic anhydride (7.5 mL) was gradually added, and the reaction solution was added at 0 ° C. to 30 ° C. Stir for minutes. The reaction solution was warmed to room temperature, saturated aqueous sodium hydrogen carbonate solution was added, and the mixture was extracted with diethyl ether. After drying the organic layer over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure, and the resulting residue was purified by column chromatography (NH silica gel, heptane: ethyl acetate = 100: 0 to 90:10), The title compound (4.8 g) was obtained as a colorless solid.
UPLC: 169 [M + H] + (retention time 0.83 minutes)
1 H-NMR (CDCl 3 , 300 MHz) δ: 8.24 (1H, d, J = 5 Hz), 7.42 (1H, d, J = 5 Hz), 4.17 (3H, s).
<工程4>tert-ブチル 5-(4-シアノ-3-メトキシピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体2-4)の合成
 2-クロロ-3-メトキシイソニコチノニトリル(中間体2-3)(0.26g)、tert-ブチル 2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(0.30g)、RuPhos(66mg)、炭酸セシウム(0.69g)のテトラヒドロフラン(3.0mL)混合溶液に、RuPhos Pd G2(0.11g)を加え、90℃にて16時間撹拌した。反応溶液に水を加え、酢酸エチルで抽出した。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し、減圧下溶媒を留去して得られた残渣を、カラムクロマトグラフィー(シリカゲル、ヘプタン:酢酸エチル=95:5~80:20)にて精製し、標記化合物(0.28g)を黄色ガム状物質として得た。
UPLC:367[M+Na]+(保持時間1.13分)
<Step 4> of tert-butyl 5- (4-cyano-3-methoxypyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 2-4) Synthesis 2-chloro-3-methoxyisonicotinonitrile (intermediate 2-3) (0.26 g), tert-butyl 2,5-diazabicyclo [2.2.2] octane-2-carboxylate (0.30 g) ), RuPhos (66 mg), and a mixed solution of cesium carbonate (0.69 g) in tetrahydrofuran (3.0 mL) were added with RuPhos Pd G2 (0.11 g) and stirred at 90 ° C. for 16 hours. Water was added to the reaction solution, and the mixture was extracted with ethyl acetate. The organic layer was washed with saturated brine, dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure. The resulting residue was subjected to column chromatography (silica gel, heptane: ethyl acetate = 95: 5 to 80:20). The title compound (0.28 g) was obtained as a yellow gum.
UPLC: 367 [M + Na] + (retention time 1.13 minutes)
<工程5>2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリル(中間体2)の合成
 tert-ブチル 5-(4-シアノ-3-メトキシピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体2-4)(0.13g)にトリフルオロ酢酸(1.0mL)を加え、室温にて1時間攪拌した。溶媒を減圧下留去して得られた残渣に、1規定塩酸水溶液を加えた後、酢酸エチル-ヘプタン混合溶媒(1:1)で洗浄した。水層を1規定水酸化ナトリウム水溶液で塩基性にした後、食塩を加え、酢酸エチルで5回抽出を行った。有機層を無水硫酸ナトリウムで乾燥し、溶媒を減圧下留去して標記化合物(21mg)を淡黄色ガム状物質として得た。
UPLC:245[M+H]+(保持時間0.66分)
1H-NMR (CDCl3, 400MHz) δ: 7.95 (1H, d, J = 5 Hz), 6.72 (1H, d, J = 5 Hz), 4.50-4.45 (1H, m), 3.88 (3H, s), 3.86-3.80 (1H, m), 3.71-3.66 (1H, m), 3.44-3.39 (1H, 2m), 3.20-3.11 (2H, m), 2.17-1.76 (4H, m).
<Step 5> Synthesis of 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) -3-methoxyisonicotinonitrile (intermediate 2) tert-butyl 5- (4-cyano- 3-methoxypyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 2-4) (0.13 g) was added trifluoroacetic acid (1.0 mL). The mixture was further stirred at room temperature for 1 hour. The solvent was evaporated under reduced pressure, 1N aqueous hydrochloric acid solution was added to the resulting residue, and the mixture was washed with an ethyl acetate-heptane mixed solvent (1: 1). The aqueous layer was basified with 1N aqueous sodium hydroxide solution, sodium chloride was added, and the mixture was extracted 5 times with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to obtain the title compound (21 mg) as a pale yellow gum.
UPLC: 245 [M + H] + (retention time 0.66 minutes)
1 H-NMR (CDCl 3 , 400 MHz) δ: 7.95 (1H, d, J = 5 Hz), 6.72 (1H, d, J = 5 Hz), 4.50-4.45 (1H, m), 3.88 (3H, s ), 3.86-3.80 (1H, m), 3.71-3.66 (1H, m), 3.44-3.39 (1H, 2m), 3.20-3.11 (2H, m), 2.17-1.76 (4H, m).
(製造例3)2-(1H-ピラゾール-1-イル)ニコチン酸(中間体3)の合成
<工程1>2-(1H-ピラゾール-1-イル)ニコチン酸エチル(中間体3-1)の合成
 2-クロロニコチン酸エチル(25g)、1H-ピラゾール(18g)、炭酸カリウム(56g)、フッ化セシウム(20g)のDMSO(140mL)混合液を窒素雰囲気下、120℃(内温)で3時間加熱した。反応液を室温へ冷却後、水(300mL)を加え、ヘプタン-酢酸エチル(2:1)混合溶媒(600mL)で抽出した。抽出液を水(300mL×2)と飽和食塩水(200mL)で順次洗浄後、有機層を無水硫酸ナトリウムで乾燥させ、減圧下溶媒を留去して標記化合物(20g)を淡黄色オイルとして得た。
UPLC:218[M+H]+(保持時間0.86分)
Production Example 3 Synthesis of 2- (1H-pyrazol-1-yl) nicotinic acid (Intermediate 3) <Step 1> Ethyl 2- (1H-pyrazol-1-yl) nicotinate (Intermediate 3-1) Synthesis of DMSO (140 mL) mixture of ethyl 2-chloronicotinate (25 g), 1H-pyrazole (18 g), potassium carbonate (56 g), cesium fluoride (20 g) at 120 ° C. (internal temperature) under nitrogen atmosphere Heated for 3 hours. The reaction mixture was cooled to room temperature, water (300 mL) was added, and the mixture was extracted with a mixed solvent (600 mL) of heptane-ethyl acetate (2: 1). The extract was washed successively with water (300 mL × 2) and saturated brine (200 mL), then the organic layer was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to obtain the title compound (20 g) as a pale yellow oil. It was.
UPLC: 218 [M + H] + (retention time 0.86 minutes)
<工程2>2-(1H-ピラゾール-1-イル)ニコチン酸(中間体3)の合成
 2-(1H-ピラゾール-1-イル)ニコチン酸エチル(中間体3-1)(20g)と水酸化ナトリウム(7.3g)のTHF(150mL)-水(150mL)混合溶液を室温で3日間攪拌した。反応液を減圧下濃縮してTHFを留去した後、MTBE(100mL)で水層を洗浄した。水層を濃塩酸でpH=1にした後、酢酸エチル(450mL×2)で抽出した。集めた抽出液を飽和食塩水(90mL)で洗浄し、無水硫酸ナトリウムで乾燥させ、減圧下溶媒を留去して標記化合物(17g)を無色固体として得た。
UPLC:190[M+H]+(保持時間0.64分)
1H-NMR (DMSO-d6, 400MHz) δ: 13.18 (1H, brs), 8.57 (1H, dd, J = 5, 2 Hz), 8.48-8.46 (1H, m), 8.07 (1H, dd, J = 8, 2 Hz), 7.77-7.75 (1H, m), 7.48 (1H, dd, J = 8, 5 Hz), 6.56 (1H, dd, J = 3, 2 Hz).
<Step 2> Synthesis of 2- (1H-pyrazol-1-yl) nicotinic acid (intermediate 3) Ethyl 2- (1H-pyrazol-1-yl) nicotinate (intermediate 3-1) (20 g) and water A mixed solution of sodium oxide (7.3 g) in THF (150 mL) -water (150 mL) was stirred at room temperature for 3 days. The reaction solution was concentrated under reduced pressure to remove THF, and the aqueous layer was washed with MTBE (100 mL). The aqueous layer was adjusted to pH = 1 with concentrated hydrochloric acid, and extracted with ethyl acetate (450 mL × 2). The collected extract was washed with saturated brine (90 mL), dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to obtain the title compound (17 g) as a colorless solid.
UPLC: 190 [M + H] + (retention time 0.64 minutes)
1 H-NMR (DMSO-d 6 , 400 MHz) δ: 13.18 (1H, brs), 8.57 (1H, dd, J = 5, 2 Hz), 8.48-8.46 (1H, m), 8.07 (1H, dd, J = 8, 2 Hz), 7.77-7.75 (1H, m), 7.48 (1H, dd, J = 8, 5 Hz), 6.56 (1H, dd, J = 3, 2 Hz).
(製造例4)1-(ピリジン-2-イル)-1H-ピラゾール-5-カルボン酸(中間体4)の合成
<工程1>メチル 1-(ピリジン-2-イル)-1H-ピラゾール-5-カルボキシレート(中間体4-1)の合成
 2-ヒドラジニルピリジン(1.0g)、(E)-メチル 4-(ジメチルアミノ)-2-オキソブト-3-エノエート(2.9g)の酢酸(10mL)混合液を、110℃にて終夜攪拌した。減圧下溶媒を留去し、得られた残渣をカラムクロマトグラフィー(シリカゲル、ヘプタン:酢酸エチル=100:0~70:30)にて精製し、標記化合物(0.72g)を橙色液体として得た。
UPLC:204[M+H]+(保持時間0.75分)
Production Example 4 Synthesis of 1- (pyridin-2-yl) -1H-pyrazole-5-carboxylic acid (Intermediate 4) <Step 1> Methyl 1- (pyridin-2-yl) -1H-pyrazole-5 Synthesis of carboxylate (intermediate 4-1) Acetic acid of 2-hydrazinylpyridine (1.0 g), (E) -methyl 4- (dimethylamino) -2-oxobut-3-enoate (2.9 g) (10 mL) The mixture was stirred at 110 ° C. overnight. The solvent was distilled off under reduced pressure, and the resulting residue was purified by column chromatography (silica gel, heptane: ethyl acetate = 100: 0 to 70:30) to obtain the title compound (0.72 g) as an orange liquid. .
UPLC: 204 [M + H] + (retention time 0.75 minutes)
<工程2>1-(ピリジン-2-イル)-1H-ピラゾール-5-カルボン酸(中間体4)の合成
 メチル 1-(ピリジン-2-イル)-1H-ピラゾール-5-カルボキシレート(中間体3-1)(0.35g)のTHF(2.0mL)-水(0.50mL)混合溶液に水酸化リチウム1水和物(0.11g)を加え、窒素雰囲気下、60℃にて2時間攪拌した。1規定塩酸水溶液を加え反応を停止させた後に、酢酸エチルで抽出し飽和食塩水で洗浄した。有機層を無水硫酸ナトリウムで乾燥させ、減圧下溶媒を留去して標記化合物(0.33g)を白色固体として得た。
UPLC:212[M+Na]+(保持時間0.67分)
1H-NMR (CDCl3, 300MHz) δ: 8.41-8.37 (1H, m), 8.36-8.32 (1H, m), 8.10-8.03 (1H, m), 7.76 (1H, d, J = 2 Hz), 7.45-7.39 (2H, m).
<Step 2> Synthesis of 1- (pyridin-2-yl) -1H-pyrazole-5-carboxylic acid (intermediate 4) Methyl 1- (pyridin-2-yl) -1H-pyrazole-5-carboxylate (intermediate) Lithium hydroxide monohydrate (0.11 g) was added to a THF (2.0 mL) -water (0.50 mL) mixed solution of Compound 3-1) (0.35 g), and the mixture was heated at 60 ° C. under a nitrogen atmosphere. Stir for 2 hours. 1N Hydrochloric acid aqueous solution was added to stop the reaction, and the mixture was extracted with ethyl acetate and washed with saturated brine. The organic layer was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to obtain the title compound (0.33 g) as a white solid.
UPLC: 212 [M + Na] + (retention time 0.67 minutes)
1 H-NMR (CDCl 3 , 300 MHz) δ: 8.41-8.37 (1H, m), 8.36-8.32 (1H, m), 8.10-8.03 (1H, m), 7.76 (1H, d, J = 2 Hz) , 7.45-7.39 (2H, m).
(製造例5)2-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタンの光学異性体(中間体5-A)、(中間体5-B)の合成
<工程1>tert-ブチル 5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体1-1)の光学分割
 tert-ブチル 5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体1-1)(0.40g)を超臨界流体クロマトグラフィーを用いて光学分割を行うことで、表記化合物の各光学異性体を、第一分画((中間体5-1-A):0.14g、橙色ガム状物質、>99%ee、リテンションタイム(分析)1.44分)、第二分画((中間体5-1-B):0.13g、橙色ガム状物質、>99%ee、リテンションタイム2.05分(分析))として得た。
分取条件(カラム:CHIRALCEL OJ-H(20mm×250mm)、溶出液;3%イソプロパノール/CO2、流速:15mL/分)
分析条件(カラム:CHIRALCEL OJ-H(4.6mm×150mm)、溶出液;3%イソプロパノール/CO2、流速:3mL/分)
Production Example 5 2- (4,6-Dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane optical isomer (Intermediate 5-A), (Intermediate 5- Synthesis of B) <Step 1> tert-Butyl 5- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (Intermediate 1-1) ) Optical resolution of tert-butyl 5- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 1-1) (0. 40 g) was subjected to optical resolution using supercritical fluid chromatography, whereby each optical isomer of the title compound was converted into the first fraction (intermediate 5-1-A): 0.14 g, orange gum ,> 99% ee, retention time (analysis) .44 minutes), the second fraction ((Intermediate 5-1-B): 0.13 g, orange gum,> 99% ee, was obtained by retention time 2.05 min and (analysis)).
Preparative conditions (column: CHIRALCEL OJ-H (20 mm × 250 mm), eluent: 3% isopropanol / CO 2 , flow rate: 15 mL / min)
Analysis conditions (column: CHIRALCEL OJ-H (4.6 mm × 150 mm), eluent: 3% isopropanol / CO 2 , flow rate: 3 mL / min)
<工程2>2-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタンの光学異性体(中間体5-A)の合成
 製造例5の工程1で得られたtert-ブチル 5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレートの光学異性体(中間体5-1-A)(0.14g)に4規定塩酸-酢酸エチル溶液(2.2mL)を加え、室温にて1時間撹拌した。減圧下溶媒を留去し、2規定水酸化ナトリウム水溶液を加えて、pH=12~13とした後、酢酸エチル(20mL)で10回抽出を行った。有機層を無水硫酸ナトリウムで乾燥させ、減圧下溶媒を留去して標記化合物(97mg)を橙色液体として得た。
UPLC:219[M+H]+(保持時間0.51分)
<Step 2> Synthesis of 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane optical isomer (intermediate 5-A) Step of Production Example 5 Tert-butyl 5- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate obtained in 1 above (intermediate 5- To 1-A) (0.14 g) was added 4N hydrochloric acid-ethyl acetate solution (2.2 mL), and the mixture was stirred at room temperature for 1 hour. The solvent was distilled off under reduced pressure, 2N aqueous sodium hydroxide solution was added to adjust the pH to 12-13, and extraction was performed 10 times with ethyl acetate (20 mL). The organic layer was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to obtain the title compound (97 mg) as an orange liquid.
UPLC: 219 [M + H] + (retention time 0.51 minutes)
<工程3>2-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタンの光学異性体(中間体5-B)の合成
 製造例5の工程2と同様の方法もしくは、これに準ずる方法で、製造例5の工程1で得られたtert-ブチル 5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレートの光学異性体(中間体5-1-B)(0.13g)から標記化合物(85mg)を淡黄色固体として得た。
UPLC:219[M+H]+(保持時間0.51分)
<Step 3> Synthesis of 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane optical isomer (intermediate 5-B) Step of Production Example 5 2 or a method analogous thereto, tert-butyl 5- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2] obtained in Step 1 of Production Example 5 was used. .2] The title compound (85 mg) was obtained as a pale yellow solid from the optical isomer of Octane-2-carboxylate (Intermediate 5-1-B) (0.13 g).
UPLC: 219 [M + H] + (retention time 0.51 minutes)
(製造例6)2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの光学異性体(中間体6-A)、(中間体6-B)の合成
<工程1>tert-ブチル 5-(4-シアノ-3-メトキシピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体2-4)の光学分割
 tert-ブチル 5-(4-シアノ-3-メトキシピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体2-4)(0.15g)を超臨界流体クロマトグラフィーを用いて光学分割を行うことで、表記化合物の各光学異性体を、第一分画((中間体6-1-A):63mg、無色ガム状物質、>99%ee、リテンションタイム(分析)1.76分)、第二分画((中間体6-1-B):60mg、無色ガム状物質、>99%ee、リテンションタイム(分析)2.07分)として得た。
分取条件(カラム:CHIRALPAK OD-H(20mm×250mm)、溶出液;10%イソプロパノール/CO2、流速:15mL/分)
分析条件(カラム:CHIRALCEL OD-H(4.6mm×150mm)、溶出液;10%イソプロパノール/CO2、流速:3mL/分)
Production Example 6 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) -3-methoxyisonicotinonitrile optical isomer (Intermediate 6-A), (Intermediate 6) Synthesis of -B) <Step 1> tert-Butyl 5- (4-cyano-3-methoxypyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate) 2-4) optical resolution of tert-butyl 5- (4-cyano-3-methoxypyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 2- 4) By optically resolving (0.15 g) using supercritical fluid chromatography, each optical isomer of the title compound was converted into the first fraction ((intermediate 6-1-A): 63 mg, colorless Gum-like material,> 99% ee, retention Time (analysis) 1.76 minutes), second fraction ((intermediate 6-1-B): 60 mg, colorless gum,> 99% ee, retention time (analysis) 2.07 minutes) .
Preparative conditions (column: CHIRALPAK OD-H (20 mm × 250 mm), eluent: 10% isopropanol / CO 2 , flow rate: 15 mL / min)
Analysis conditions (column: CHIRALCEL OD-H (4.6 mm × 150 mm), eluent: 10% isopropanol / CO 2 , flow rate: 3 mL / min)
<工程2>2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの光学異性体(中間体6-A)の合成
 製造例5の工程2と同様の方法もしくは、これに準ずる方法で、製造例6の工程1で得られたtert-ブチル 5-(4-シアノ-3-メトキシピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレートの光学異性体(6-1-A)(60mg)から標記化合物(40mg)を橙色ガム状物質として得た。
UPLC:245[M+H]+(保持時間0.67分)
<Step 2> Synthesis of 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) -3-methoxyisonicotinonitrile optical isomer (intermediate 6-A) In the same manner as in Step 2 or a method analogous thereto, tert-butyl 5- (4-cyano-3-methoxypyridin-2-yl) -2,5-diazabicyclo [O] obtained in Step 1 of Production Example 6 2.2.2] The title compound (40 mg) was obtained as an orange gum from the optical isomer of the octane-2-carboxylate (6-1-A) (60 mg).
UPLC: 245 [M + H] + (retention time 0.67 minutes)
<工程3>2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの光学異性体(中間体6-B)の合成
 製造例5の工程2と同様の方法もしくは、これに準ずる方法で、製造例6の工程1で得られたtert-ブチル 5-(4-シアノ-3-メトキシピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレートの光学異性体(6-1-B)(58mg)から標記化合物(41mg)を橙色ガム状物質として得た。
UPLC:245[M+H]+(保持時間0.67分)
<Step 3> Synthesis of optical isomer of 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotinonitrile (intermediate 6-B) In the same manner as in Step 2 or a method analogous thereto, tert-butyl 5- (4-cyano-3-methoxypyridin-2-yl) -2,5-diazabicyclo [O] obtained in Step 1 of Production Example 6 2.2.2] The title compound (41 mg) was obtained as an orange gum from the optical isomer of the octane-2-carboxylate (6-1-B) (58 mg).
UPLC: 245 [M + H] + (retention time 0.67 minutes)
(製造例7)2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体(中間体7-A)、(中間体7-B)の合成
<工程1>tert-ブチル 5-(3-クロロ-4-シアノピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体7-1)の合成
 3-クロロ-2-フルオロイソニコチノニトリル(0.19g)、tert-ブチル 2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(0.20g)、炭酸カリウム(0.39g)のDMSO(3mL)混合液を100℃にて30分撹拌した。反応溶液に飽和塩化アンモニウム水溶液を加えた後、酢酸エチルで抽出を行った。集めた抽出液を無水硫酸ナトリウムで乾燥し、減圧下溶媒を留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、ヘプタン:酢酸エチル=100:0~75:25)にて精製し、標記化合物(0.26g)を黄色固体として得た。
UPLC:371,373[M+Na]+(保持時間1.16分)
(Production Example 7) 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropylisonicotinonitrile optical isomer (Intermediate 7-A), (Intermediate) Synthesis of 7-B) <Step 1> tert-butyl 5- (3-chloro-4-cyanopyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate) Synthesis of Compound 7-1) 3-Chloro-2-fluoroisonicotinonitrile (0.19 g), tert-butyl 2,5-diazabicyclo [2.2.2] octane-2-carboxylate (0.20 g) Then, a mixed solution of potassium carbonate (0.39 g) in DMSO (3 mL) was stirred at 100 ° C. for 30 minutes. A saturated aqueous ammonium chloride solution was added to the reaction solution, followed by extraction with ethyl acetate. The collected extract was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure, and the resulting residue was purified by column chromatography (silica gel, heptane: ethyl acetate = 100: 0 to 75:25). The compound (0.26 g) was obtained as a yellow solid.
UPLC: 371, 373 [M + Na] + (retention time 1.16 minutes)
<工程2>tert-ブチル 5-(4-シアノ-3-シクロプロピルピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体7-2)の合成
 tert-ブチル 5-(3-クロロ-4-シアノピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体7-1)(0.26g)、シクロプロピル亜鉛ブロミド(0.5規定 THF溶液)(9mL)のTHF(2.5mL)混合溶液にテトラキス(トリフェニルホスフィン)パラジウム(0.35g)を加え、窒素雰囲気下、100℃にて15時間撹拌した。反応溶液を室温に戻した後に飽和塩化アンモニウム水溶液を加え、酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥した後、減圧下溶媒を留去して得られた残渣をカラムクロマトグラフィー(NHシリカゲル、ヘプタン:酢酸エチル=100:0~80:20)にて精製し、標記化合物(0.13g)を無色オイル状物質として得た。
UPLC:377[M+Na]+(保持時間1.16分)
1H-NMR (CDCl3, 300MHz) δ: 8.10 (1H, d, J = 5 Hz), 6.88 (1H, d, J = 5 Hz), 4.59-4.48 (1H, m), 4.33-4.13 (1H, m), 3.94-3.68 (3H, m), 3.57-3.45 (1H, m), 2.30-1.68 (5H, m), 1.48 (9H, s), 1.19-1.10 (2H, m), 0.76-0.69 (2H, m).
<Step 2> tert-butyl 5- (4-cyano-3-cyclopropylpyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 7-2) Synthesis of tert-butyl 5- (3-chloro-4-cyanopyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 7-1) (0. 26 g), tetrakis (triphenylphosphine) palladium (0.35 g) was added to a THF (2.5 mL) mixed solution of cyclopropyl zinc bromide (0.5 N THF solution) (9 mL), and the mixture was heated to 100 ° C. under a nitrogen atmosphere. And stirred for 15 hours. The reaction solution was returned to room temperature, saturated aqueous ammonium chloride solution was added, and the mixture was extracted with ethyl acetate. After drying the organic layer over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure, and the resulting residue was purified by column chromatography (NH silica gel, heptane: ethyl acetate = 100: 0 to 80:20) The compound (0.13 g) was obtained as a colorless oily substance.
UPLC: 377 [M + Na] + (retention time 1.16 minutes)
1 H-NMR (CDCl 3 , 300 MHz) δ: 8.10 (1H, d, J = 5 Hz), 6.88 (1H, d, J = 5 Hz), 4.59-4.48 (1H, m), 4.33-4.13 (1H , m), 3.94-3.68 (3H, m), 3.57-3.45 (1H, m), 2.30-1.68 (5H, m), 1.48 (9H, s), 1.19-1.10 (2H, m), 0.76-0.69 (2H, m).
<工程3>tert-ブチル 5-(4-シアノ-3-シクロプロピルピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体7-2)の光学分割
 tert-ブチル 5-(4-シアノ-3-シクロプロピルピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体7-2)(0.13g)を超臨界流体クロマトグラフィーを用いて光学分割を行うことで、表記化合物の各光学異性体を、第一分画((中間体7-3-A):50mg、無色オイル状物質、>99%ee、リテンションタイム(分析)1.62分)、第二分画((中間体7-3-B):52mg、無色オイル状物質、>99%ee、リテンションタイム(分析)2.22分)として得た。
分取条件(カラム:CHIRALPAK OD-H(20mm×250mm)、溶出液;10%メタノール/CO2、流速:15mL/分)
分析条件(カラム:CHIRALCEL OD-H(4.6mm×150mm)、溶出液;10%メタノール/CO2、流速:3mL/分)
<Step 3> tert-butyl 5- (4-cyano-3-cyclopropylpyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 7-2) Optical resolution of tert-butyl 5- (4-cyano-3-cyclopropylpyridin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 7-2) 0.13 g) was optically resolved using supercritical fluid chromatography, whereby each optical isomer of the title compound was converted into the first fraction (intermediate 7-3-A): 50 mg, colorless oily substance ,> 99% ee, retention time (analysis) 1.62 minutes), second fraction ((intermediate 7-3-B): 52 mg, colorless oily substance,> 99% ee, retention time (analysis) 2 22 minutes) Obtained.
Preparative conditions (column: CHIRALPAK OD-H (20 mm × 250 mm), eluent: 10% methanol / CO 2 , flow rate: 15 mL / min)
Analysis conditions (column: CHIRALCEL OD-H (4.6 mm × 150 mm), eluent: 10% methanol / CO 2 , flow rate: 3 mL / min)
<工程4>2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体(中間体7-A)の合成
 製造例5の工程2と同様の方法もしくは、これに準ずる方法で、製造例7の工程3で得られたtert-ブチル 5-(4-シアノ-3-シクロプロピルピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレートの光学異性体(中間体7-3-A)(50mg)から標記化合物(44mg)を黄色オイル状物質として得た。
UPLC:255[M+H]+(保持時間0.73分)
<Step 4> Synthesis of 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropylisonicotinonitrile optical isomer (intermediate 7-A) Production Example 5 Tert-Butyl 5- (4-cyano-3-cyclopropylpyridin-2-yl) -2,5- The title compound (44 mg) was obtained as a yellow oil from the optical isomer of diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 7-3-A) (50 mg).
UPLC: 255 [M + H] + (retention time 0.73 minutes)
<工程5>2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体(中間体7-B)の合成
 製造例5の工程2と同様の方法もしくは、これに準ずる方法で、製造例7の工程3で得られたtert-ブチル 5-(4-シアノ-3-シクロプロピルピリジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレートの光学異性体(中間体7-3-B)(52mg)から標記化合物(42mg)を黄色オイル状物質として得た。
UPLC:255[M+H]+(保持時間0.72分)
<Step 5> Synthesis of optical isomer of 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropylisonicotinonitrile (intermediate 7-B) Production Example 5 Tert-Butyl 5- (4-cyano-3-cyclopropylpyridin-2-yl) -2,5- obtained in Step 3 of Production Example 7 in the same manner as in Step 2 above or a method analogous thereto. The title compound (42 mg) was obtained as a yellow oil from the optical isomer of diazabicyclo [2.2.2] octane-2-carboxylate (intermediate 7-3-B) (52 mg).
UPLC: 255 [M + H] + (retention time 0.72 minutes)
(実施例1)(5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)(5-メチル-2-(2H-1,2,3-トリアゾール-2-イル)フェニル)メタノンの合成
 tert-ブチル 5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-カルボキシレート(中間体1-1)(0.11g)のジクロロメタン溶液にトリフルオロ酢酸(0.88mL)を加え、反応液を室温にて2時間撹拌した。減圧下溶媒を留去して得られた残渣をトルエンで共沸した後、DMFで希釈し、5-メチル-2-(2H-1,2,3-トリアゾール-2-イル)安息香酸(70mg)、HATU(0.16g)、トリエチルアミン(0.19mL)を順次加え、室温にて攪拌した。1時間後、反応溶液に水を加え酢酸エチルで抽出した後、有機層を水で洗い、無水硫酸ナトリウムで乾燥した。減圧下溶媒を留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、ヘプタン:酢酸エチル=50:50~0:100)にて精製し、標記化合物(74mg)を白色固体として得た。
UPLC:404[M+H]+(保持時間0.91、0.95分、回転異性体)
1H-NMR (DMSO-d6, 300MHz) δ: 7.95-7.68 (3H, m), 7.43-7.33 (1H, m), 7.28-7.20 (1H, m), 6.39-6.33 (1H, m), 5.05-3.24 (6H, m), 2.70 (3H, s), 2.44-2.35 (2H, m), 2.26-2.18 (6H, m), 1.93-1.76 (2H, m).
Example 1 (5- (4,6-Dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-yl) (5-methyl-2- (2H-1 , 2,3-Triazol-2-yl) phenyl) methanone tert-butyl 5- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2- Trifluoroacetic acid (0.88 mL) was added to a dichloromethane solution of carboxylate (intermediate 1-1) (0.11 g), and the reaction solution was stirred at room temperature for 2 hours. The residue obtained by evaporating the solvent under reduced pressure was azeotroped with toluene and diluted with DMF to give 5-methyl-2- (2H-1,2,3-triazol-2-yl) benzoic acid (70 mg). ), HATU (0.16 g) and triethylamine (0.19 mL) were sequentially added, and the mixture was stirred at room temperature. After 1 hour, water was added to the reaction solution and the mixture was extracted with ethyl acetate. The organic layer was washed with water and dried over anhydrous sodium sulfate. The residue obtained by evaporating the solvent under reduced pressure was purified by column chromatography (silica gel, heptane: ethyl acetate = 50: 50 to 0: 100) to obtain the title compound (74 mg) as a white solid.
UPLC: 404 [M + H] + (retention time 0.91, 0.95 min, rotamer)
1 H-NMR (DMSO-d 6 , 300 MHz) δ: 7.95-7.68 (3H, m), 7.43-7.33 (1H, m), 7.28-7.20 (1H, m), 6.39-6.33 (1H, m), 5.05-3.24 (6H, m), 2.70 (3H, s), 2.44-2.35 (2H, m), 2.26-2.18 (6H, m), 1.93-1.76 (2H, m).
(実施例2)(2-(2H-1,2,3-トリアゾール-2-イル)フェニル)(5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)メタノンの合成
 2-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン塩酸塩(中間体1)(50mg)、2-(2H-1,2,3-トリアゾール-2-イル)安息香酸(32mg)、HATU(76mg)のDMF(1.0mL)混合溶液にトリエチルアミン(70μL)を加え、室温にて18時間撹拌した。反応液に水を加え、酢酸エチルで抽出を行い、有機層を水、飽和食塩水で順次洗浄し、無水硫酸ナトリウムで乾燥した。減圧下溶媒を留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、ヘプタン:酢酸エチル=60:40~25:75)にて精製し、標記化合物(37mg)を白色固体として得た。
UPLC:390[M+H]+(保持時間0.83、0.87分、回転異性体)
1H-NMR (DMSO-d6, 400MHz) δ: 8.10-7.83 (2H, m), 7.66-7.42 (4H, m), 6.40-6.35 (1H, m), 5.06-3.26 (6H, m), 2.90-2.67 (2H, m), 2.23 (6H, s), 1.94-1.75 (2H, m).
Example 2 (2- (2H-1,2,3-triazol-2-yl) phenyl) (5- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2 .2] Synthesis of octan-2-yl) methanone 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane hydrochloride (intermediate 1) (50 mg) Triethylamine (70 μL) was added to a mixed solution of 2- (2H-1,2,3-triazol-2-yl) benzoic acid (32 mg) and HATU (76 mg) in DMF (1.0 mL) at room temperature for 18 hours. Stir. Water was added to the reaction mixture, and the mixture was extracted with ethyl acetate. The organic layer was washed successively with water and saturated brine, and dried over anhydrous sodium sulfate. The residue obtained by evaporating the solvent under reduced pressure was purified by column chromatography (silica gel, heptane: ethyl acetate = 60: 40 to 25:75) to obtain the title compound (37 mg) as a white solid.
UPLC: 390 [M + H] + (retention time 0.83, 0.87 min, rotamer)
1 H-NMR (DMSO-d 6 , 400 MHz) δ: 8.10-7.83 (2H, m), 7.66-7.42 (4H, m), 6.40-6.35 (1H, m), 5.06-3.26 (6H, m), 2.90-2.67 (2H, m), 2.23 (6H, s), 1.94-1.75 (2H, m).
下記(実施例3)ないし(実施例6)は、上記(中間体1)または市販化合物を用いて、(実施例2)と同様の方法、もしくはこれに準ずる方法により合成した。
Figure JPOXMLDOC01-appb-T000036
The following (Example 3) to (Example 6) were synthesized by the same method as (Example 2) or a method analogous thereto using the above (Intermediate 1) or a commercially available compound.
Figure JPOXMLDOC01-appb-T000036
(実施例7)2-(5-(2-(2H-1,2,3-トリアゾール-2-イル)ベンゾイル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの合成
 2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリル(中間体2)(11mg)、2-(2H-1,2,3-トリアゾール-2-イル)安息香酸(9.5mg)、HATU(21mg)のDMF(1.0mL)混合溶液にトリエチルアミン(9.5μL)を加え、室温にて64時間撹拌した。反応液に水を加え、酢酸エチルで抽出を行い、有機層を水、飽和食塩水で順次洗浄し、無水硫酸ナトリウムで乾燥した。減圧下溶媒を留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、ヘプタン:酢酸エチル=70:30~25:75)にて精製し、標記化合物(13mg)を無色アモルファスとして得た。
UPLC:416[M+H]+(保持時間1.02、1.03分、回転異性体)
1H-NMR (CDCl3, 400MHz) δ: 8.05-7.62 (3H, m), 7.59-7.49 (1H, m), 7.48-7.36 (2H, m), 7.30-7.21 (1H, m), 6.82-6.73 (1H, m), 4.95-3.23 (10H, m), 2.39-1.67 (3H, m).
Example 7 2- (5- (2- (2H-1,2,3-triazol-2-yl) benzoyl) -2,5-diazabicyclo [2.2.2] octane-2-yl)- Synthesis of 3-methoxyisonicotinonitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotinonitrile (intermediate 2) (11 mg), 2- ( To a mixed solution of 2H-1,2,3-triazol-2-yl) benzoic acid (9.5 mg) and HATU (21 mg) in DMF (1.0 mL) was added triethylamine (9.5 μL), and the mixture was stirred at room temperature for 64 hours. Stir. Water was added to the reaction mixture, and the mixture was extracted with ethyl acetate. The organic layer was washed successively with water and saturated brine, and dried over anhydrous sodium sulfate. The residue obtained by evaporating the solvent under reduced pressure was purified by column chromatography (silica gel, heptane: ethyl acetate = 70: 30 to 25:75) to obtain the title compound (13 mg) as a colorless amorphous.
UPLC: 416 [M + H] + (retention time 1.02, 1.03 min, rotamer)
1 H-NMR (CDCl 3 , 400 MHz) δ: 8.05-7.62 (3H, m), 7.59-7.49 (1H, m), 7.48-7.36 (2H, m), 7.30-7.21 (1H, m), 6.82- 6.73 (1H, m), 4.95-3.23 (10H, m), 2.39-1.67 (3H, m).
(実施例8)2-(5-(2-(1H-ピラゾール-1-イル)ニコチノイル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの合成
 2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリル(中間体2)(25mg)、2-(1H-ピラゾール-1-イル)ニコチン酸(23mg)、HATU(58mg)のジクロロメタン(1.0mL)混合溶液に2,6-ルチジン(36μL)を加え、窒素雰囲気下、室温にて20時間撹拌した。反応液に飽和炭酸水素ナトリウム水溶液(10mL)を加え、酢酸エチル(20mL)で2回抽出を行った。有機層を飽和食塩水で洗浄し、無水硫酸ナトリウムで乾燥した。減圧下溶媒を留去して得られた残渣をカラムクロマトグラフィー(NHシリカゲル、ヘプタン:酢酸エチル=88:12~0:100)にて精製し、標記化合物(23mg)を無色アモルファスとして得た。
UPLC:416[M+H]+(保持時間0.97、1.00分、回転異性体)
1H-NMR (CDCl3, 400MHz) δ: 8.52-8.37 (2H, m), 8.01-7.80 (1H, m), 7.76-7.20 (3H, m), 6.83-6.70 (1H, m), 6.51-6.22 (1H, m), 5.00-4.79 (1H, m), 4.66-3.19 (9H, m), 2.59-1.68 (3H, m).
Example 8 2- (5- (2- (1H-pyrazol-1-yl) nicotinoyl) -2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotino Synthesis of nitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotinonitrile (intermediate 2) (25 mg), 2- (1H-pyrazole-1- Il) 2,6-lutidine (36 μL) was added to a mixed solution of nicotinic acid (23 mg) and HATU (58 mg) in dichloromethane (1.0 mL), and the mixture was stirred at room temperature for 20 hours under a nitrogen atmosphere. A saturated aqueous sodium hydrogen carbonate solution (10 mL) was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate (20 mL). The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The residue obtained by evaporating the solvent under reduced pressure was purified by column chromatography (NH silica gel, heptane: ethyl acetate = 88: 12 to 0: 100) to obtain the title compound (23 mg) as a colorless amorphous.
UPLC: 416 [M + H] + (retention time 0.97, 1.00 min, rotamer)
1 H-NMR (CDCl 3 , 400 MHz) δ: 8.52-8.37 (2H, m), 8.01-7.80 (1H, m), 7.76-7.20 (3H, m), 6.83-6.70 (1H, m), 6.51- 6.22 (1H, m), 5.00-4.79 (1H, m), 4.66-3.19 (9H, m), 2.59-1.68 (3H, m).
(実施例9)(2-(2H-1,2,3-トリアゾール-2-イル)フェニル)(5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)メタノン光学異性体の合成
 3-(2H-1,2,3-トリアゾール-2-イル)安息香酸(30mg)のジクロロメタン(0.80mL)溶液に塩化オキサリル(42μL)と触媒量のDMFを加え、窒素雰囲気下、室温にて30分撹拌後、減圧下溶媒を留去した。この残渣をジクロロメタン(0.60mL)に溶解し、2-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタンの光学異性体(中間体5-A)(35mg)とトリエチルアミン(0.11mL)のジクロロメタン(10mL)混合溶液へ0℃で滴下した。反応液を窒素雰囲気下、0℃にて1時間撹拌後、カラムクロマトグラフィー(シリカゲル、ヘプタン:酢酸エチル=90:10~0:100)にて精製し、標記化合物(57mg)を無色アモルファスとして得た。
UPLC:390[M+H]+(保持時間0.83、0.87分、回転異性体)
1H-NMRは、(実施例1)と一致した。
Example 9 (2- (2H-1,2,3-triazol-2-yl) phenyl) (5- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2 .2] Synthesis of Octan-2-yl) methanone optical isomer 3- (2H-1,2,3-triazol-2-yl) benzoic acid (30 mg) in dichloromethane (0.80 mL) in oxalyl chloride (42 μL) And a catalytic amount of DMF were added, and the mixture was stirred at room temperature for 30 minutes in a nitrogen atmosphere, and then the solvent was distilled off under reduced pressure. This residue was dissolved in dichloromethane (0.60 mL), and the optical isomer of 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane (intermediate 5- A) (35 mg) and triethylamine (0.11 mL) were added dropwise to a mixed solution of dichloromethane (10 mL) at 0 ° C. The reaction solution was stirred at 0 ° C. for 1 hour under a nitrogen atmosphere and then purified by column chromatography (silica gel, heptane: ethyl acetate = 90: 10 to 0: 100) to obtain the title compound (57 mg) as a colorless amorphous. It was.
UPLC: 390 [M + H] + (retention time 0.83, 0.87 min, rotamer)
1 H-NMR was consistent with (Example 1).
(実施例10)(2-(2H-1,2,3-トリアゾール-2-イル)フェニル)(5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)メタノン光学異性体の合成
 実施例9と同様の方法もしくは、これに準ずる方法で、2-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタンの光学異性体(中間体5-B)(35mg)から標記化合物(35mg)を無色アモルファスとして得た。
UPLC:390[M+H]+(保持時間0.83、0.87分、回転異性体)
1H-NMRは、(実施例1)と一致した。
Example 10 (2- (2H-1,2,3-triazol-2-yl) phenyl) (5- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2 .2] Synthesis of octan-2-yl) methanone optical isomer 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo The title compound (35 mg) was obtained as a colorless amorphous form from the optical isomer of [2.2.2] octane (Intermediate 5-B) (35 mg).
UPLC: 390 [M + H] + (retention time 0.83, 0.87 min, rotamer)
1 H-NMR was consistent with (Example 1).
(実施例11)(5-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)(1-(ピリジン-2-イル)-1H-ピラゾール-5-イル)メタノン光学異性体の合成
 2-(4,6-ジメチルピリミジン-2-イル)-2,5-ジアザビシクロ[2.2.2]オクタンの光学異性体(中間体5-B)(35mg)、1-(ピリジン-2-イル)-1H-ピラゾール-5-カルボン酸(中間体4)(31mg)、のDMF(1.0mL)混合溶液にHATU(63mg)、ジイソプロピルエチルアミン(72μL)を加え、室温にて30分間撹拌した。反応液に水を加え、酢酸エチル(5.0mL)で3回抽出を行った。有機層を飽和食塩水で洗浄し、無水硫酸ナトリウムで乾燥した。減圧下溶媒を留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、ヘプタン:酢酸エチル=80:20~40:60)にて精製し、標記化合物(14mg)を白色固体として得た。
UPLC:390[M+H]+(保持時間0.80、0.84分、回転異性体)
1H-NMR (CDCl3, 300MHz) δ: 8.28-8.22 (1H, m), 7.94-7.65 (3H, m), 7.20-7.09 (1H, m), 6.49-6.43 (1H, m), 6.31-6.25 (1H, m), 5.25-3.38 (7H, m), 2.32-2.20 (6H, m), 2.16-1.66 (3H, m).
Example 11 (5- (4,6-Dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane-2-yl) (1- (pyridin-2-yl)- Synthesis of 1H-pyrazol-5-yl) methanone optical isomer 2- (4,6-dimethylpyrimidin-2-yl) -2,5-diazabicyclo [2.2.2] octane optical isomer (intermediate 5 -B) (35 mg), 1- (pyridin-2-yl) -1H-pyrazole-5-carboxylic acid (Intermediate 4) (31 mg), in a mixed solution of DMF (1.0 mL), HATU (63 mg), diisopropyl Ethylamine (72 μL) was added and stirred at room temperature for 30 minutes. Water was added to the reaction mixture, and the mixture was extracted 3 times with ethyl acetate (5.0 mL). The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The residue obtained by evaporating the solvent under reduced pressure was purified by column chromatography (silica gel, heptane: ethyl acetate = 80: 20 to 40:60) to obtain the title compound (14 mg) as a white solid.
UPLC: 390 [M + H] + (retention time 0.80, 0.84 min, rotamer)
1 H-NMR (CDCl 3 , 300 MHz) δ: 8.28-8.22 (1H, m), 7.94-7.65 (3H, m), 7.20-7.09 (1H, m), 6.49-6.43 (1H, m), 6.31- 6.25 (1H, m), 5.25-3.38 (7H, m), 2.32-2.20 (6H, m), 2.16-1.66 (3H, m).
(実施例12)2-(5-(2-(2H-1,2,3-トリアゾール-2-イル)ベンゾイル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリル光学異性体の合成
 実施例7と同様の方法もしくは、これに準ずる方法で、2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの光学異性体(中間体6-A)(14mg)から標記化合物(13mg)を無色アモルファスとして得た。
UPLC:416[M+H]+(保持時間1.02分)
1H-NMRは、(実施例7)と一致した。
Example 12 2- (5- (2- (2H-1,2,3-triazol-2-yl) benzoyl) -2,5-diazabicyclo [2.2.2] octane-2-yl)- Synthesis of 3-methoxyisonicotinonitrile optical isomer 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) -3 in the same manner as in Example 7 or a method analogous thereto The title compound (13 mg) was obtained as a colorless amorphous form from the optical isomer of 2-methoxyisonicotinonitrile (Intermediate 6-A) (14 mg).
UPLC: 416 [M + H] + (retention time 1.02 minutes)
1 H-NMR was consistent with (Example 7).
(実施例13)2-(5-(2-(2H-1,2,3-トリアゾール-2-イル)ベンゾイル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリル光学異性体の合成
 実施例7と同様の方法もしくは、これに準ずる方法で、2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの光学異性体(中間体6-B)(18mg)から標記化合物(7.7mg)を無色アモルファスとして得た。
UPLC:416[M+H]+(保持時間1.02分)
1H-NMRは、(実施例7)と一致した。
Example 13 2- (5- (2- (2H-1,2,3-triazol-2-yl) benzoyl) -2,5-diazabicyclo [2.2.2] octane-2-yl)- Synthesis of 3-methoxyisonicotinonitrile optical isomer 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) -3 in the same manner as in Example 7 or a method analogous thereto The title compound (7.7 mg) was obtained as a colorless amorphous form from the optical isomer of 2-methoxyisonicotinonitrile (intermediate 6-B) (18 mg).
UPLC: 416 [M + H] + (retention time 1.02 minutes)
1 H-NMR was consistent with (Example 7).
(実施例14)2-(5-(2-(1H-ピラゾール-1-イル)ニコチノイル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの光学異性体の合成
 実施例7と同様の方法もしくは、これに準ずる方法で、2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの光学異性体(中間体6-A)(12mg)から標記化合物(6.2mg)を無色アモルファスとして得た。
UPLC:416[M+H]+(保持時間0.99、1.01分、回転異性体)
1H-NMRは、(実施例8)と一致した。
Example 14 2- (5- (2- (1H-pyrazol-1-yl) nicotinoyl) -2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotino Synthesis of optical isomer of nitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotino by the same method as in Example 7 or a method analogous thereto The title compound (6.2 mg) was obtained as a colorless amorphous form from the optical isomer of nitrile (Intermediate 6-A) (12 mg).
UPLC: 416 [M + H] + (retention time 0.99, 1.01 min, rotamer)
1 H-NMR was consistent with (Example 8).
(実施例15)2-(5-(2-(1H-ピラゾール-1-イル)ニコチノイル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの光学異性体の合成
 実施例7と同様の方法もしくは、これに準ずる方法で、2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-メトキシイソニコチノニトリルの光学異性体(中間体6-B)(12mg)から標記化合物(9.6mg)を無色アモルファスとして得た。
UPLC:416[M+H]+(保持時間0.99、1.01分、回転異性体)
1H-NMRは、(実施例8)と一致した。
Example 15 2- (5- (2- (1H-pyrazol-1-yl) nicotinoyl) -2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotino Synthesis of optical isomer of nitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-methoxyisonicotino by the same method as in Example 7 or a method analogous thereto The title compound (9.6 mg) was obtained as a colorless amorphous form from the optical isomer of nitrile (Intermediate 6-B) (12 mg).
UPLC: 416 [M + H] + (retention time 0.99, 1.01 min, rotamer)
1 H-NMR was consistent with (Example 8).
(実施例16)2-(5-(2-(1H-ピラゾール-1-イル)ニコチノイル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体の合成
 2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体(中間体7-A)(22mg)、2-(1H-ピラゾール-1-イル)ニコチン酸(16mg)、HATU(40mg)のジクロロメタン(2mL)混合溶液に2,6-ルチジン(20μL)を加え、窒素雰囲気下、室温にて15時間撹拌した。反応液に飽和炭酸水素ナトリウム水溶液を加え、酢酸エチルで抽出し、抽出液を無水硫酸ナトリウムで乾燥した。減圧下溶媒を留去して得られた残渣をカラムクロマトグラフィー(NHシリカゲル、ヘプタン:酢酸エチル=2:1~1:2)にて精製し、標記化合物(24mg)を無色アモルファスとして得た。
UPLC:426[M+H]+(保持時間1.03、1.07分、回転異性体)
1H-NMR (DMSO-D6, 400MHz) δ: 8.57-8.42 (2H, m), 8.19-7.18 (4H, m), 7.11-6.98 (1H, m), 6.60-6.35 (1H, m), 4.63-3.40 (6H, m), 2.30-0.98 (7H, m), 0.65-0.51 (2H, m).
Example 16 2- (5- (2- (1H-pyrazol-1-yl) nicotinoyl) -2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropylisonicoti Synthesis of optical isomers of nononitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropylisonicotinonitrile optical isomer (intermediate 7-A) ( 22 mg), 2- (1H-pyrazol-1-yl) nicotinic acid (16 mg), and HATU (40 mg) in dichloromethane (2 mL) were added with 2,6-lutidine (20 μL), and a nitrogen atmosphere at room temperature was added. Stir for 15 hours. A saturated aqueous sodium hydrogen carbonate solution was added to the reaction mixture, and the mixture was extracted with ethyl acetate. The extract was dried over anhydrous sodium sulfate. The solvent was distilled off under reduced pressure, and the resulting residue was purified by column chromatography (NH silica gel, heptane: ethyl acetate = 2: 1 to 1: 2) to obtain the title compound (24 mg) as a colorless amorphous.
UPLC: 426 [M + H] + (retention time 1.03, 1.07 min, rotamer)
1 H-NMR (DMSO-D 6 , 400 MHz) δ: 8.57-8.42 (2H, m), 8.19-7.18 (4H, m), 7.11-6.98 (1H, m), 6.60-6.35 (1H, m), 4.63-3.40 (6H, m), 2.30-0.98 (7H, m), 0.65-0.51 (2H, m).
(実施例17)2-(5-(2-(1H-ピラゾール-1-イル)ニコチノイル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体の合成
 実施例16と同様の方法もしくは、これに準ずる方法で、2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体(中間体7-B)(21mg)から標記化合物(26mg)を無色アモルファスとして得た。
UPLC:426[M+H]+(保持時間1.02、1.06分、回転異性体)
Example 17 2- (5- (2- (1H-pyrazol-1-yl) nicotinoyl) -2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropylisonicoti Synthesis of optical isomers of nononitrile 2- (2,5-diazabicyclo [2.2.2] octan-2-yl) -3-cyclopropyliso-isolated by a method similar to or equivalent to that in Example 16. The title compound (26 mg) was obtained as a colorless amorphous form from the optical isomer of nicotinonitrile (Intermediate 7-B) (21 mg).
UPLC: 426 [M + H] + (retention time 1.02, 1.06 min, rotamer)
(実施例18)2-(5-(2-(2H-1,2,3-トリアゾール-2-イル)ベンゾイル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体の合成
 実施例16と同様の方法もしくは、これに準ずる方法で、2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体(中間体7-A)(22mg)から標記化合物(26mg)を無色アモルファスとして得た。
UPLC:426[M+H]+(保持時間1.05、1.09分、回転異性体)
Example 18 2- (5- (2- (2H-1,2,3-triazol-2-yl) benzoyl) -2,5-diazabicyclo [2.2.2] octane-2-yl)- Synthesis of optical isomers of 3-cyclopropylisonicotinonitrile 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) in the same manner as in Example 16 or a method analogous thereto The title compound (26 mg) was obtained as a colorless amorphous form from the optical isomer of -3-cyclopropylisonicotinonitrile (Intermediate 7-A) (22 mg).
UPLC: 426 [M + H] + (retention time 1.05, 1.09 min, rotamer)
(実施例19)2-(5-(2-(2H-1,2,3-トリアゾール-2-イル)ベンゾイル)-2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体の合成
 実施例16と同様の方法もしくは、これに準ずる方法で、2-(2,5-ジアザビシクロ[2.2.2]オクタン-2-イル)-3-シクロプロピルイソニコチノニトリルの光学異性体(中間体7-B)(21mg)から標記化合物(23mg)を無色アモルファスとして得た。
UPLC:426[M+H]+(保持時間1.05、1.08分、回転異性体)
Example 19 2- (5- (2- (2H-1,2,3-triazol-2-yl) benzoyl) -2,5-diazabicyclo [2.2.2] octane-2-yl)- Synthesis of optical isomers of 3-cyclopropylisonicotinonitrile 2- (2,5-diazabicyclo [2.2.2] octane-2-yl) in the same manner as in Example 16 or a method analogous thereto The title compound (23 mg) was obtained as a colorless amorphous form from the optical isomer of -3-cyclopropylisonicotinonitrile (Intermediate 7-B) (21 mg).
UPLC: 426 [M + H] + (retention time 1.05, 1.08 min, rotamer)
上記の(実施例1)から(実施例19)で合成した最終化合物の構造を以下の[化32]および[化33]に示す。また、上記製造例中で得られた中間体化合物の構造を以下の[化34]および[化35]に示す。 The structures of the final compounds synthesized in the above (Example 1) to (Example 19) are shown in the following [Chemical Formula 32] and [Chemical Formula 33]. The structures of the intermediate compounds obtained in the above production examples are shown in the following [Chemical Formula 34] and [Chemical Formula 35].
Figure JPOXMLDOC01-appb-C000037
Figure JPOXMLDOC01-appb-C000037
Figure JPOXMLDOC01-appb-C000038
Figure JPOXMLDOC01-appb-C000038
Figure JPOXMLDOC01-appb-C000039
Figure JPOXMLDOC01-appb-C000039
Figure JPOXMLDOC01-appb-C000040
Figure JPOXMLDOC01-appb-C000040

Claims (7)

  1. 下記式(I):
    Figure JPOXMLDOC01-appb-C000001
    [式(I)中、mは、0~4の整数であり;nは、0~4の整数であり;環Aは、C6~14アリール基、または5~7員ヘテロアリール基を表し;環Bは、C6~14アリール基、または5~7員ヘテロアリール基を表し; Qは、C3~8シクロアルキル基、または-ORc、または部分構造式(SF-1)で表される基:
    Figure JPOXMLDOC01-appb-C000002
    [式(SF-1)中、環Cは、C6~14アリール基、5~7員ヘテロアリール基、または3~8非芳香族複素環基を表し;pは、0~5の整数であり;R3は、各々独立に、ハロゲン原子、シアノ基、-Rd、または-ORdを表し、Rdは、C1~6アルキル基、ハロゲン化C1~6アルキル基、またはC3~8シクロアルキル基を表す]を表し;R1は、各々独立に、ハロゲン原子、シアノ基、-Ra、-ORa、-C(O)Ra、-C(O)ORa、-SRa、-SO2a、-NRAB基、3~8員非芳香族複素環基、またはオキソ基を表し; R2は、各々独立に、ハロゲン原子、シアノ基、-Rb、-ORb、または5~7員ヘテロアリール基を表し、前記5~7員ヘテロアリール基は、1~5個のハロゲン原子または水酸基で置換されていても良く;前記R1中の-NRAB基におけるRAおよびRBは、各々独立に、-Ra、-C(O)Ra、-C(O)ORa、または-SO2aを表し;前記R1および-NRAB基中におけるRaは、水素原子、C1~6アルキル基、ハロゲン化C1~6アルキル基、ヒドロキシC1~6アルキル基、ヒドロキシハロゲン化C1~6アルキル基、またはC3~8シクロアルキル基を表し;前記R2中のRbは、C1~6アルキル基、またはハロゲン化C1~6アルキル基を表し;前記Q中のRcは、C1~6アルキル基、ハロゲン化C1~6アルキル基、またはC6~14アリール基を表す]で表される化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物。
    The following formula (I):
    Figure JPOXMLDOC01-appb-C000001
    [In formula (I), m is an integer from 0 to 4; n is an integer of 0-4; ring A represents a C 6 ~ 14 aryl group or a 5-7 membered heteroaryl group, ; ring B is C 6 ~ 14 aryl group, or represents a 5- to 7-membered heteroaryl group,; Q is Table with C 3 ~ 8 cycloalkyl group, or -OR c or partial structural formula,, (SF-1) Group:
    Figure JPOXMLDOC01-appb-C000002
    Wherein (SF-1), ring C is C 6 ~ 14 aryl group, 5- to 7-membered heteroaryl group or 3-8 represents a non-aromatic heterocyclic group,; p is an integer of 0 to 5 Yes; each R 3 independently represents a halogen atom, a cyano group, —R d , or —OR d , where R d is a C 1-6 alkyl group, a halogenated C 1-6 alkyl group, or C 3 represents ~ represents an 8 cycloalkyl group]; R 1 is each independently a halogen atom, a cyano group, -R a, -OR a, -C (O) R a, -C (O) OR a, - SR a represents —SO 2 R a , —NR A R B group, 3- to 8-membered non-aromatic heterocyclic group, or oxo group; R 2 each independently represents a halogen atom, a cyano group, —R b represents -OR b or 5-7 membered heteroaryl group, the 5-7 membered heteroaryl group, optionally substituted with 1-5 halogen atoms or a hydroxyl group Ku; R A and R B in -NR A R B group in the R 1 are each independently, -R a, -C (O) R a, -C (O) OR a or -SO 2 R, It represents a; R a in the R 1 and -NR a R B group, a hydrogen atom, C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl group, hydroxy C 1 ~ 6 alkyl group, hydroxy halides C 1 ~ 6 alkyl group or an C 3 ~ 8 cycloalkyl group; R b in the R 2 represents a C 1 ~ 6 alkyl group or halogenated C 1 ~ 6 alkyl group; in the Q R c is, C 1 ~ 6 alkyl group, a halogenated C 1 ~ 6 alkyl group or a compound represented by the representative] a C 6 ~ 14 aryl group, or a pharmaceutically acceptable salt thereof or their solvent, Japanese products.
  2.  請求項1に記載の化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の少なくとも1つを有効成分として含有することを特徴とする、医薬組成物。 A pharmaceutical composition comprising at least one of the compound according to claim 1 or a pharmaceutically acceptable salt thereof or a solvate thereof as an active ingredient.
  3.  請求項1に記載の化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の少なくとも1つを有効成分として含有することを特徴とする、オレキシン受容体が関与する疾患の予防及び/または治療剤。 The prevention of a disease involving orexin receptor comprising at least one of the compound according to claim 1 or a pharmaceutically acceptable salt thereof or a solvate thereof as an active ingredient / Or therapeutic agent.
  4.  請求項1に記載の化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の少なくとも1つを有効成分として含有することを特徴とする、睡眠障害の予防及び/または治療剤。 A prophylactic and / or therapeutic agent for sleep disorders, comprising at least one of the compound according to claim 1 or a pharmaceutically acceptable salt or solvate thereof as an active ingredient.
  5.  請求項1に記載の化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の1つ以上を含む、オレキシン受容体拮抗剤。 An orexin receptor antagonist comprising the compound according to claim 1 or a pharmaceutically acceptable salt or solvate thereof.
  6.  オレキシン2受容体選択的拮抗剤である、請求項5に記載のオレキシン受容体拮抗剤。 The orexin receptor antagonist according to claim 5, which is an orexin 2 receptor selective antagonist.
  7.  請求項1に記載の化合物、または製薬学的に許容されるその塩またはそれらの溶媒和物の1つ以上と、(1)睡眠導入剤、(2)睡眠時無呼吸症候群で処方される薬剤、(3)レストレスレッグス症候群で処方される薬剤、(4)非定型抗精神病薬、(5)定型抗精神病薬、(6)選択的セロトニン再取り込み阻害薬(SSRI)、(7)選択的セロトニン・ノルアドレナリン再取り込み阻害薬(SNRI)、(8)選択的ノルアドレナリン・ドーパミン再取り込み阻害薬(NDRI)、(9)ノルアドレナリン作動性・特異的セロトニン作動性抗うつ薬(NaSSA)、(10)トリアゾロピリジン系抗うつ薬(SARI)、(11)四環系抗うつ薬、(12)三環系抗うつ薬、(13)その他抗うつ薬、(14)α7ニコチン受容体作動薬、(15)α7ニコチン受容体活性調節薬、(16)α7ニコチン受容体部分調節薬、(17)PDE阻害薬、(18)NK2拮抗薬、(19)NK3拮抗薬、(20)ムスカリン型M1アセチルコリン受容体活性調節薬、(21)ムスカリン型M2アセチルコリン受容体活性調節薬、(22)アデノシン受容体調節薬、(23)ムスカリン型M4アセチルコリン受容体活性調節薬、(24)ムスカリン型M5アセチルコリン受容体活性調節薬、(25)アデノシン受容体調節薬、(26)グリシントランスポーター1(GlyT1)阻害薬、(27)グルタミン酸増強薬、(28)NMDA受容体阻害薬、(29)代謝性グルタミン酸受容体調節薬(mGlu)、(30)抗不安薬、(31)βアミロイドワクチン、(32)βアミロイド分解酵素等、(33)脳機能賦活薬、(34)カンナビノイド調節薬、(35)コリンエステラーゼ阻害薬、(36)MAO-B阻害剤、(37)パーキンソン病治療薬、(38)糖尿病治療薬、(39)抗肥満薬、(40)コレステロール低下薬等の高脂血症治療薬、(41)降圧剤、(42)非ステロイド性抗炎症薬、(43)疾患修飾性抗リウマチ薬、(44)抗サイトカイン薬、(45)ステロイド薬、(46)性ホルモンまたはその誘導体、(47)副甲状腺ホルモン、(48)オピオイド作動薬、(49)ピリン系解熱鎮痛薬、(50)非ピリン系解熱鎮痛薬、(51)非ステロイド性抗炎症薬(NSAIDs)、(52)COX-2選択的阻害薬、(53)末梢性神経障害性疼痛・線維筋痛症薬、および(54)抗てんかん薬からなる群より選択される薬剤の1種以上とを含有することを特徴とする医薬組成物。 One or more of the compounds according to claim 1, or a pharmaceutically acceptable salt thereof, or a solvate thereof, and (1) a sleep-inducing agent and (2) a drug prescribed for sleep apnea syndrome , (3) drugs prescribed for restless legs syndrome, (4) atypical antipsychotics, (5) typical antipsychotics, (6) selective serotonin reuptake inhibitors (SSRI), (7) selective Serotonin / noradrenaline reuptake inhibitor (SNRI), (8) Selective noradrenaline / dopamine reuptake inhibitor (NDRI), (9) Noradrenergic / specific serotonergic antidepressant (NaSSA), (10) Tria Zolopyridine antidepressants (SARI), (11) tetracyclic antidepressants, (12) tricyclic antidepressants, (13) other antidepressants, (14) α7 nicotine receptor production (15) α7 nicotinic receptor activity modulator, (16) α7 nicotinic receptor partial modulator, (17) PDE inhibitor, (18) NK2 antagonist, (19) NK3 antagonist, (20) muscarinic type M1 acetylcholine receptor activity modulator, (21) muscarinic M2 acetylcholine receptor activity regulator, (22) adenosine receptor modulator, (23) muscarinic M4 acetylcholine receptor activity regulator, (24) muscarinic M5 acetylcholine Receptor activity modulator, (25) adenosine receptor modulator, (26) glycine transporter 1 (GlyT1) inhibitor, (27) glutamate enhancer, (28) NMDA receptor inhibitor, (29) metabolic glutamate Receptor modulator (mGlu), (30) anxiolytic, (31) β-amyloid vaccine, (32) β-amyloid (33) brain function activator, (34) cannabinoid modulator, (35) cholinesterase inhibitor, (36) MAO-B inhibitor, (37) Parkinson's disease treatment, (38) diabetes treatment, (39) anti-obesity drugs, (40) anti-hyperlipidemic drugs such as cholesterol-lowering drugs, (41) antihypertensive drugs, (42) non-steroidal anti-inflammatory drugs, (43) disease-modifying anti-rheumatic drugs, (44 ) Anti-cytokine drugs, (45) steroid drugs, (46) sex hormones or derivatives thereof, (47) parathyroid hormone, (48) opioid agonists, (49) pilin antipyretic analgesics, (50) non-pyrine antipyretic analgesics , (51) non-steroidal anti-inflammatory drugs (NSAIDs), (52) COX-2 selective inhibitors, (53) peripheral neuropathic pain / fibromyalgia drugs, and (54) antiepileptic drugs Pharmaceutical composition characterized by containing a one or more agents selected from Ranaru group.
PCT/JP2015/083926 2014-12-03 2015-12-02 Novel diazabicyclo[2.2.2]octane derivative WO2016088813A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2014-245240 2014-12-03
JP2014245240A JP2018016544A (en) 2014-12-03 2014-12-03 Novel diazabicyclo [2.2.2]octane derivative

Publications (1)

Publication Number Publication Date
WO2016088813A1 true WO2016088813A1 (en) 2016-06-09

Family

ID=56091753

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2015/083926 WO2016088813A1 (en) 2014-12-03 2015-12-02 Novel diazabicyclo[2.2.2]octane derivative

Country Status (2)

Country Link
JP (1) JP2018016544A (en)
WO (1) WO2016088813A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018015196A1 (en) 2016-07-20 2018-01-25 Bayer Aktiengesellschaft Substituted diazahetero-bicyclic compounds and their use
WO2018228909A1 (en) 2017-06-14 2018-12-20 Bayer Pharma Aktiengesellschaft Substituted bridged diazepane derivatives and use thereof as task-1 and task-3 inhibitors
WO2018228907A1 (en) 2017-06-14 2018-12-20 Bayer Aktiengesellschaft Diazabicyclic substituted imidazopyrimidines and their use for the treatment of breathing disorders
US10414765B2 (en) 2015-12-10 2019-09-17 Bayer Pharma Aktiengesellschaft Substituted perhydropyrrolo[3,4-c]pyrrole derivatives and the use of same
US10759794B2 (en) 2015-12-10 2020-09-01 Bayer Pharma Aktiengesellschaft 2-phenyl-3-(piperazinomethyl)imidazo[1,2-A]pyridine derivatives as blockers of task-1 and task-2 channels, for the treatment of sleep-related breathing disorders
CN111892599A (en) * 2020-08-14 2020-11-06 黄芳 Synthesis method of 2, 5-diazabicyclo [2.2.2] octane-2-carboxylic acid tert-butyl ester
CN118598880A (en) * 2024-08-07 2024-09-06 艾斯拓康生物医药(天津)有限公司 Nitrogen-containing bridged ring compound, and preparation method and application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007537300A (en) * 2004-05-18 2007-12-20 シェーリング コーポレイション Substituted 2-quinolyl-oxazoles useful as PDE4 inhibitors
JP2009543785A (en) * 2006-07-14 2009-12-10 メルク エンド カムパニー インコーポレーテッド Cross-linked diazepan orexin receptor antagonist
WO2013050938A1 (en) * 2011-10-04 2013-04-11 Actelion Pharmaceuticals Ltd 3,7-diazabicyclo[3.3.1]nonane and 9-oxa-3,7-diazabicyclo[3.3.1]nonane derivatives
WO2013119639A1 (en) * 2012-02-07 2013-08-15 Eolas Therapeutics, Inc. Substituted prolines / piperidines as orexin receptor antagonists

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007537300A (en) * 2004-05-18 2007-12-20 シェーリング コーポレイション Substituted 2-quinolyl-oxazoles useful as PDE4 inhibitors
JP2009543785A (en) * 2006-07-14 2009-12-10 メルク エンド カムパニー インコーポレーテッド Cross-linked diazepan orexin receptor antagonist
WO2013050938A1 (en) * 2011-10-04 2013-04-11 Actelion Pharmaceuticals Ltd 3,7-diazabicyclo[3.3.1]nonane and 9-oxa-3,7-diazabicyclo[3.3.1]nonane derivatives
WO2013119639A1 (en) * 2012-02-07 2013-08-15 Eolas Therapeutics, Inc. Substituted prolines / piperidines as orexin receptor antagonists

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10414765B2 (en) 2015-12-10 2019-09-17 Bayer Pharma Aktiengesellschaft Substituted perhydropyrrolo[3,4-c]pyrrole derivatives and the use of same
US10759794B2 (en) 2015-12-10 2020-09-01 Bayer Pharma Aktiengesellschaft 2-phenyl-3-(piperazinomethyl)imidazo[1,2-A]pyridine derivatives as blockers of task-1 and task-2 channels, for the treatment of sleep-related breathing disorders
WO2018015196A1 (en) 2016-07-20 2018-01-25 Bayer Aktiengesellschaft Substituted diazahetero-bicyclic compounds and their use
WO2018228909A1 (en) 2017-06-14 2018-12-20 Bayer Pharma Aktiengesellschaft Substituted bridged diazepane derivatives and use thereof as task-1 and task-3 inhibitors
WO2018228907A1 (en) 2017-06-14 2018-12-20 Bayer Aktiengesellschaft Diazabicyclic substituted imidazopyrimidines and their use for the treatment of breathing disorders
US11098063B2 (en) 2017-06-14 2021-08-24 Bayer Aktiengesellschaft Diazabicyclic substituted imidazopyrimidines and their use for the treatment of breathing disorders
CN111892599A (en) * 2020-08-14 2020-11-06 黄芳 Synthesis method of 2, 5-diazabicyclo [2.2.2] octane-2-carboxylic acid tert-butyl ester
CN111892599B (en) * 2020-08-14 2023-01-13 黄芳 Synthesis method of 2,5-diazabicyclo [2.2.2] octane-2-tert-butyl carboxylate
CN118598880A (en) * 2024-08-07 2024-09-06 艾斯拓康生物医药(天津)有限公司 Nitrogen-containing bridged ring compound, and preparation method and application thereof

Also Published As

Publication number Publication date
JP2018016544A (en) 2018-02-01

Similar Documents

Publication Publication Date Title
EP2963037B1 (en) Novel pyrazole derivative
WO2016088813A1 (en) Novel diazabicyclo[2.2.2]octane derivative
WO2016084866A1 (en) Novel diazabicyclo derivative
CN110831926B (en) Tetrahydronaphtyl urea derivatives
JP2017095366A (en) Novel biaryl amide derivative
JP7514534B2 (en) Benzodiazepine derivatives, compositions and methods for treating cognitive disorders
JP6689856B2 (en) 6,7-Dihydropyrazolo [1,5-a] pyrazin-4 (5H) -one compounds and their use as negative allosteric modulators of the MGLUR2 receptor
CN103987710A (en) Novel oxazine derivatives and their use in the treatment of disease
JP2021526123A (en) Autotaxin inhibitors and their use
WO2018130869A1 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
JP2020526557A (en) Antagonist of muscarinic acetylcholine receptor M4
WO2016205739A1 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
CN109890388A (en) MAChR M4Positive allosteric modulators
WO2013005798A1 (en) Novel nitrogenated heterocyclic ring derivative
CN109071457A (en) The inhibitor of indole amine 2,3-dioxygenase
JP6745594B2 (en) Crystal of salt of pyrazole derivative
WO2017090716A1 (en) Crystals of pyrazole derivative
JP2022553284A (en) Inhibitors of TREK (TWIK-related K+ channels) channel function
JP2017075116A (en) Novel condensed heterocyclic compound
CN103998446A (en) Novel trifluoromethyl-oxadiazole derivatives and their use in the treatment of disease

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: 15866184

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15866184

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP