JPWO2020071550A1 - CDK8 Inhibitors and Their Applications - Google Patents

CDK8 Inhibitors and Their Applications Download PDF

Info

Publication number
JPWO2020071550A1
JPWO2020071550A1 JP2020551124A JP2020551124A JPWO2020071550A1 JP WO2020071550 A1 JPWO2020071550 A1 JP WO2020071550A1 JP 2020551124 A JP2020551124 A JP 2020551124A JP 2020551124 A JP2020551124 A JP 2020551124A JP WO2020071550 A1 JPWO2020071550 A1 JP WO2020071550A1
Authority
JP
Japan
Prior art keywords
group
compound
mmol
added
pharmaceutically acceptable
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
JP2020551124A
Other languages
Japanese (ja)
Inventor
弘明 白波瀬
弘明 白波瀬
達哉 北尾
達哉 北尾
高橋 健司
健司 高橋
幸倫 庄子
幸倫 庄子
滋充 武田
滋充 武田
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kyoto Pharmaceutical Industries Ltd
Original Assignee
Kyoto Pharmaceutical Industries Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Kyoto Pharmaceutical Industries Ltd filed Critical Kyoto Pharmaceutical Industries Ltd
Publication of JPWO2020071550A1 publication Critical patent/JPWO2020071550A1/en
Pending legal-status Critical Current

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
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/351Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom not condensed with another ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • A61K31/37Coumarins, e.g. psoralen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • 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
    • A61K31/397Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having four-membered rings, e.g. azetidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/47042-Quinolinones, e.g. carbostyril
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • 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
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/12Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/48Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D309/08Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D309/10Oxygen atoms
    • C07D309/12Oxygen atoms only hydrogen atoms and one oxygen atom directly attached to ring carbon atoms, e.g. tetrahydropyranyl ethers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/06Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2
    • C07D311/08Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring
    • C07D311/14Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring substituted in position 6 and unsubstituted in position 7
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/50Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D333/52Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes
    • C07D333/54Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to carbon atoms of the hetero ring
    • C07D333/56Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/10Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/10Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6571Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and oxygen atoms as the only ring hetero atoms
    • C07F9/6574Esters of oxyacids of phosphorus

Abstract

新規なCDK8阻害剤を提供する。本発明は、一般式(I):[式中の各記号は、明細書に記載のとおりである。]で表される化合物またはその医薬上許容される塩、および該化合物を有効成分として含有する医薬組成物に関する。本発明によれば、上記化合物がCDK8阻害活性を有しており、CDK8に関連する疾患、骨粗鬆症等の骨代謝に関連する疾患、癌等の細胞増殖性疾患の予防・治療剤として有用な医薬を提供することができる。To provide a novel CDK8 inhibitor. In the present invention, the general formula (I): [each symbol in the formula is as described in the specification. ], A pharmaceutically acceptable salt thereof, and a pharmaceutical composition containing the compound as an active ingredient. According to the present invention, the above-mentioned compound has CDK8 inhibitory activity, and is useful as a prophylactic / therapeutic agent for CDK8-related diseases, bone metabolism-related diseases such as osteoporosis, and cell proliferation diseases such as cancer. Can be provided.

Description

本発明は、サイクリン依存性キナーゼ(CDK)8阻害活性を有する化合物またはその医薬上許容される塩に関する。さらに本発明は、CDK8に関連する疾患、骨粗鬆症等の骨代謝に関連する疾患、癌等の細胞増殖性疾患の予防及び/または治療のために有用な化合物またはその医薬上許容される塩に関する。 The present invention relates to a compound having cyclin-dependent kinase (CDK) 8 inhibitory activity or a pharmaceutically acceptable salt thereof. Furthermore, the present invention relates to compounds useful for the prevention and / or treatment of CDK8-related diseases, bone metabolism-related diseases such as osteoporosis, and cell proliferation diseases such as cancer, or pharmaceutically acceptable salts thereof.

CDKは、サイクリンタンパク質と複合体を形成して活性化されるリン酸化酵素であり、細胞周期や転写調節に関与する因子として知られている。特に、CDK2、CDK4、CDK6は、主に細胞周期に、CDK7、CDK8、CDK9は、主に転写に関与することが知られている。その中で、CDK8はサイクリンC、MED12、MED13と複合体を形成し、RNAポリメラーゼIIのC末端ドメインなどのリン酸化を司ることで、主に転写を制御するキナーゼとして働いている(非特許文献1)。
CDK8は、一部の大腸癌細胞において癌遺伝子であること、また、大腸癌患者では、CDK8の発現が亢進し、このことがβカテニンシグナルを活性化させ、細胞増殖を正に制御することが報告されている(非特許文献2)。さらに、CDK8は癌細胞の未分化性の維持や上皮間葉転換にも関わっていることが知られている。
また、CDK8阻害剤Cortistatin Aは、種々のがん細胞の増殖を抑制し、特に白血病細胞に対する効果が著しいことが報告されている(非特許文献3)。
非特許文献4には、CDK8を阻害する化合物が癌の治療または予防に有用であること、特許文献1〜6には、抗癌作用を有するCDK阻害剤が開示されている。
CDK is a kinase that is activated by forming a complex with a cyclin protein, and is known as a factor involved in cell cycle and transcriptional regulation. In particular, it is known that CDK2, CDK4 and CDK6 are mainly involved in the cell cycle, and CDK7, CDK8 and CDK9 are mainly involved in transcription. Among them, CDK8 forms a complex with cyclin C, MED12, and MED13, and acts as a kinase that mainly controls transcription by controlling phosphorylation of the C-terminal domain of RNA polymerase II (non-patent literature). 1).
CDK8 is an oncogene in some colorectal cancer cells, and in colorectal cancer patients, the expression of CDK8 is enhanced, which activates β-catenin signal and positively regulates cell proliferation. It has been reported (Non-Patent Document 2). Furthermore, CDK8 is known to be involved in the maintenance of undifferentiated cancer cells and epithelial-mesenchymal transition.
In addition, it has been reported that the CDK8 inhibitor Cyclin A suppresses the growth of various cancer cells and has a particularly remarkable effect on leukemia cells (Non-Patent Document 3).
Non-Patent Document 4 discloses that a compound that inhibits CDK8 is useful for the treatment or prevention of cancer, and Patent Documents 1 to 6 disclose CDK inhibitors having an anticancer activity.

また、本発明化合物は、骨粗鬆症などの骨代謝に関連する疾患にも有用である。骨粗鬆症の治療薬としては破骨細胞の活性を抑える骨吸収抑制薬と骨芽細胞を活性化する骨形成促進薬がある。骨吸収抑制薬としては、カルシトニン、ビスホスホネート、エストロゲン受容体モジュレータなどが使用されている。しかしこれらの治療薬では、さらなる骨量の減少は防止されるが、失われた骨を再構築できない。一方、骨形成促進薬としてはヒトPTH(1-34)が用いられており、骨量および骨密度の上昇および骨構造の再構築に利用できる。しかしながら使用期間が1年半から2年に限定されており、また、長期にわたって皮下注射が必要で、患者がそれを遵守することは困難となる。 The compound of the present invention is also useful for diseases related to bone metabolism such as osteoporosis. As therapeutic agents for osteoporosis, there are bone resorption inhibitors that suppress the activity of osteoclasts and bone formation promoters that activate osteoblasts. As the bone resorption inhibitor, calcitonin, bisphosphonate, estrogen receptor modulator and the like are used. However, while these treatments prevent further loss of bone mass, they cannot reconstruct lost bone. On the other hand, human PTH (1-34) is used as a bone formation-promoting agent, and can be used for increasing bone mass and bone density and reconstructing bone structure. However, the period of use is limited to one and a half to two years, and long-term subcutaneous injection is required, making it difficult for patients to comply with it.

そこで、高い臨床効果を有する経口投与可能な骨形成促進剤の開発が望まれている。最近になって、アルカリホスファターゼ誘導活性を有するベンゾチエピン誘導体(特許文献7、8)、N−キノリルアントラニル酸誘導体(特許文献9)、トリアゾロピリダジン誘導体(特許文献10)、チエノピリジン誘導体(特許文献11)、[5,6]複素環化合物(特許文献12)および芳香族化合物(特許文献13)が、骨形成促進や骨代謝に関連する疾患の治療に有用であることが報告されている。しかし、その臨床上の有用性は不明である。 Therefore, it is desired to develop an orally administrable bone formation promoter having a high clinical effect. Recently, a benzothiepine derivative having alkaline phosphatase-inducing activity (Patent Documents 7 and 8), an N-quinolylanthranic acid derivative (Patent Document 9), a triazolopyridazine derivative (Patent Document 10), and a thienopyridine derivative (Patent Document 11). ), [5,6] Heterocyclic compounds (Patent Document 12) and aromatic compounds (Patent Document 13) have been reported to be useful for promoting bone formation and treating diseases related to bone metabolism. However, its clinical usefulness is unknown.

米国特許出願公開第2012/0071477号明細書U.S. Patent Application Publication No. 2012/0071477 WO2007/133772号WO2007 / 133772 WO2013/116786号WO2013 / 116786 WO2014/072435号WO2014 / 072435 WO2015/159937号WO2015 / 159937 WO2015/159938号WO2015 / 159938 US6346521US636521 US6632807US6632807 特開平9−188665号公報Japanese Unexamined Patent Publication No. 9-188665 US7173033US7173033 特開2007−131617号公報Japanese Unexamined Patent Publication No. 2007-131617 WO2011/136264号WO2011 / 136264 WO2015/030189号WO2015 / 030189

A. D. Clark et al., Crit Rev Biochem Mol Biol., 50(5), 393-426 (2015)A. D. Clark et al., Crit Rev Biochem Mol Biol., 50 (5), 393-426 (2015) R. Firestein et al., Nature 455, 547-551 (2008)R. Firestein et al., Nature 455, 547-551 (2008) H. E. Pelish et al., Nature 526, 273-276 (2015)H. E. Pelish et al., Nature 526, 273-276 (2015) T. Rzymski et al., Biochim. Biophys. Acta, Volume 1845, 1617-1629 (2015)T. Rzymski et al., Biochim. Biophys. Acta, Volume 1845, 1617-1629 (2015)

本発明の課題は、CDK8阻害作用を有し、且つ、CDK8に関連する疾患、骨粗鬆症等の骨代謝に関連する疾患、癌等の細胞増殖性疾患の予防及び/または治療剤を提供することである。さらには、安全性が高く経口投与が可能な化合物及び医薬上許容される塩を提供することである。 An object of the present invention is to provide a preventive and / or therapeutic agent for a CDK8-inhibiting action, a disease related to CDK8, a disease related to bone metabolism such as osteoporosis, and a cell proliferative disease such as cancer. be. Furthermore, it is to provide a compound which is highly safe and can be orally administered, and a pharmaceutically acceptable salt.

本発明者らは、かかる状況下、鋭意検討を重ねた結果、CDK8に対し強い阻害作用を示し、CDK8に関連する疾患、骨粗鬆症等の骨代謝に関連する疾患、癌等の細胞増殖性疾患の予防及び/または治療剤になり得る本発明の優れた化合物を見出し、本発明を完成するに至った。 As a result of diligent studies under such circumstances, the present inventors showed a strong inhibitory effect on CDK8, and found diseases related to CDK8, diseases related to bone metabolism such as osteoporosis, and cell proliferative diseases such as cancer. We have found an excellent compound of the present invention that can be a prophylactic and / or therapeutic agent, and have completed the present invention.

即ち、本発明は、
[1]一般式(I):
That is, the present invention
[1] General formula (I):

Figure 2020071550
Figure 2020071550

[式中、各置換基は以下のように定義される。
A環:
[In the formula, each substituent is defined as follows.
Ring A:

Figure 2020071550
Figure 2020071550

(R
1−6アルキルカルボニル基、C1−6アルキルチオカルボニル基、カルバモイル基、C1−6アルコキシカルボニル基またはカルボキシ基、

水素原子、C1−6アルキル基またはハロゲン原子、

1−6アルキル基、ハロゲン原子、アミノ基、ヒドロキシC1−6アルキル基、カルボキシC1−6アルキル基または水素原子(但し、Tが=CH−、Uが=N−の場合)、

水素原子またはC1−6アルキル基、

水素原子、C1−6アルキル基またはC1−6アルキルカルボニル基、

水素原子またはC1−6アルキル基)、

水素原子、ハロゲン原子、シアノ基、C1−6アルキル基、C1−6アルコキシ基、C1−6アルキルスルファニル基、C1−6アルキルカルボニル基、カルバモイル基または複素環基、
T及びU:
T及びUのいずれか一方が、=N−である場合、もう一方は=CR−、
T及びUの両方が、=CR−、

水素原子、ハロゲン原子、C1−6アルキル基またはC1−6アルコキシ基、

−B−D−E−G
(B:
6−14アリーレン基、C3−8シクロアルケニレン基、−複素環−または−飽和複素環−、
D:
単結合、−O(CH−、−(CHO−または−飽和複素環−、
n:
1〜4から選択されるいずれか1の整数、
E:
単結合、−O−、−N(R10)−、−CO−または−CONH−、
10
水素原子、置換基群αから選択されるいずれかの基で置換されていてもよいC1−6アルキル基、
G:
水素原子、置換基群αから選択されるいずれかの基で置換されていてもよいC1−6アルキル基、置換基群αから選択されるいずれかの基で置換されていてもよいC3−8シクロアルキル基、置換基群αから選択されるいずれかの基で置換されていてもよいC6−14アリール基、置換基群αから選択されるいずれかの基で置換されていてもよい複素環基または置換基群αから選択されるいずれかの基で置換されていてもよい飽和複素環基、
置換基群α:
複素環基、飽和複素環基、ハロゲン原子、シアノ基、C1−6アルキルカルボニル基、C1−6アルキルカルボニルオキシ基、C1−6アルキルカルボニルアミノ基、C1−6アルコキシカルボニル基、C1−6アルコキシC1−6アルキル基、ヒドロキシC1−6アルキル基、ヒドロキシC1−6アルコキシ基、C1−6アルキルスルホニル基、C1−6アルキルスルホニルアミノ基、アミノスルホニルアミノ基、カルボキシ基、カルボキシC1−6アルキルカルボニルオキシ基、カルバモイル基、ヒドロキシ基、C1−6アルコキシ基、C1−6アルキル基、C3−8シクロアルキル基、アミノ基、アミノC1−6アルキルカルボニルオキシ基、モノまたはジ−C1−6アルキルアミノ基、4−ヒドロキシテトラヒドロフラン−3−イルオキシ基、モノまたはジ−C1−6アルキルホスホノ基、
(R 4 :
C 1-6 alkylcarbonyl group, C 1-6 alkylthiocarbonyl group, carbamoyl group, C 1-6 alkoxycarbonyl group or carboxy group,
R 5 :
Hydrogen atom, C 1-6 alkyl group or halogen atom,
R 6 :
C 1-6 alkyl group, halogen atom, amino group, hydroxy C 1-6 alkyl group, carboxy C 1-6 alkyl group or hydrogen atom (provided that T is = CH- and U is = N-),
R 7 :
Hydrogen atom or C 1-6 alkyl group,
R 8 :
Hydrogen atom, C 1-6 alkyl group or C 1-6 alkyl carbonyl group,
R 9 :
Hydrogen atom or C 1-6 alkyl group),
R 1 :
Hydrogen atom, halogen atom, cyano group, C 1-6 alkyl group, C 1-6 alkoxy group, C 1-6 alkyl sulfanyl group, C 1-6 alkyl carbonyl group, carbamoyl group or heterocyclic group,
T and U:
If either T or U is = N-, the other is = CR 2- ,
Both T and U = CR 2- ,
R 2 :
Hydrogen atom, halogen atom, C 1-6 alkyl group or C 1-6 alkoxy group,
R 3 :
−BDEG
(B:
C 6-14 arylene group, C 3-8 cycloalkenylene group, -heterocycle-or-saturated heterocycle-,
D:
Single bond, -O (CH 2 ) n -,-(CH 2 ) n O- or -saturated heterocycle-,
n:
An integer of any one selected from 1 to 4,
E:
Single bond, -O-, -N (R 10 )-, -CO- or -CONH-,
R 10 :
A hydrogen atom, a C 1-6 alkyl group which may be substituted with any group selected from the substituent group α,
G:
Hydrogen atom, C 1-6 alkyl group may be substituted with any group selected from the substituent group α, C 3 which may be substituted with any group selected from the substituent group α -8 Cycloalkyl group, C 6-14 aryl group which may be substituted with any group selected from the substituent group α, or any group selected from the substituent group α Saturated heterocyclic groups, which may be substituted with a good heterocyclic group or any group selected from the substituent group α,
Substituent group α:
Heterocyclic group, saturated heterocyclic group, halogen atom, cyano group, C 1-6 alkylcarbonyl group, C 1-6 alkylcarbonyloxy group, C 1-6 alkylcarbonylamino group, C 1-6 alkoxycarbonyl group, C 1-6 alkoxy C 1-6 alkyl group, hydroxy C 1-6 alkyl group, hydroxy C 1-6 alkoxy group, C 1-6 alkylsulfonyl group, C 1-6 alkylsulfonylamino group, aminosulfonylamino group, carboxy Group, carboxy C 1-6 alkylcarbonyloxy group, carbamoyl group, hydroxy group, C 1-6 alkoxy group, C 1-6 alkyl group, C 3-8 cycloalkyl group, amino group, amino C 1-6 alkylcarbonyl Oxy group, mono or di-C 1-6 alkylamino group, 4-hydroxy tetrahydrofuran-3-yloxy group, mono or di-C 1-6 alkylphosphono group,

Figure 2020071550
Figure 2020071550

(nは、1〜4から選択されるいずれか1の整数))]で表わされる化合物(以下、「化合物(I)」と称することもある。)またはその医薬上許容される塩。
[2]A環が、
(N is an integer of any one selected from 1 to 4))] (hereinafter, may be referred to as “Compound (I)”) or a pharmaceutically acceptable salt thereof.
[2] Ring A

Figure 2020071550
Figure 2020071550

であり、Rが、アセチル基、カルバモイル基またはカルボキシ基であり、Rが、メチル基である、[1]に記載の化合物またはその医薬上許容される塩。
[3]A環が、
The compound according to [1] or a pharmaceutically acceptable salt thereof, wherein R 4 is an acetyl group, a carbamoyl group or a carboxy group, and R 5 is a methyl group.
[3] Ring A

Figure 2020071550
Figure 2020071550

であり、Rが、メチル基であり、Rが、水素原子である、[1]に記載の化合物またはその医薬上許容される塩。
[4]A環が、
The compound according to [1] or a pharmaceutically acceptable salt thereof, wherein R 7 is a methyl group and R 9 is a hydrogen atom.
[4] Ring A

Figure 2020071550
Figure 2020071550

であり、Rが、メチル基である、[1]に記載の化合物またはその医薬上許容される塩。
[5]A環が、
The compound according to [1] or a pharmaceutically acceptable salt thereof, wherein R 7 is a methyl group.
[5] Ring A

Figure 2020071550
Figure 2020071550

である、[1]に記載の化合物またはその医薬上許容される塩。
[6]A環が、
The compound according to [1] or a pharmaceutically acceptable salt thereof.
[6] Ring A

Figure 2020071550
Figure 2020071550

である、[1]に記載の化合物またはその医薬上許容される塩。
[7]A環が、
The compound according to [1] or a pharmaceutically acceptable salt thereof.
[7] Ring A

Figure 2020071550
Figure 2020071550

であり、Rが、塩素、メチル基またはカルボキシメチル基である、[1]に記載の化合物またはその医薬上許容される塩。
[8]Rが、シアノ基、メトキシ基、メチルスルファニル基、アセチル基、カルバモイル基またはイミダゾリル基である、[1]〜[7]のいずれか一項に記載の化合物またはその医薬上許容される塩。
[9]Tが、=CH−であり、Uが、=N−である、[1]〜[8]のいずれか一項に記載の化合物またはその医薬上許容される塩。
[10]Tが、=CH−であり、Uが、=CR−であり、Rが、水素原子、フッ素原子またはメトキシ基である、[1]〜[8]のいずれか一項に記載の化合物またはその医薬上許容される塩。
[11]Bが、
, And the salt R 6 is chlorine, a methyl group or a carboxymethyl group, that is acceptable compound or a medicament according to [1].
[8] The compound according to any one of [1] to [7], wherein R 1 is a cyano group, a methoxy group, a methylsulfanyl group, an acetyl group, a carbamoyl group or an imidazolyl group, or a pharmaceutically acceptable compound thereof. Salt.
[9] The compound according to any one of [1] to [8], wherein T is = CH- and U is = N-, or a pharmaceutically acceptable salt thereof.
[10] In any one of [1] to [8], where T is = CH-, U is = CR 2- , and R 2 is a hydrogen atom, a fluorine atom or a methoxy group. The compound described or a pharmaceutically acceptable salt thereof.
[11] B is

Figure 2020071550
Figure 2020071550

である、[1]〜[10]のいずれか一項に記載の化合物またはその医薬上許容される塩。
[12]Bが、
The compound according to any one of [1] to [10] or a pharmaceutically acceptable salt thereof.
[12] B is

Figure 2020071550
Figure 2020071550

である、[1]〜[10]のいずれか一項に記載の化合物またはその医薬上許容される塩。
[13]Dが、単結合、−O(CH−または−飽和複素環−であり、nが2である、[1]〜[12]のいずれか一項に記載の化合物またはその医薬上許容される塩。
[14]Eが、単結合または−O−である、[1]〜[13]のいずれか一項に記載の化合物またはその医薬上許容される塩。
[15]Gが、ヒドロキシ基、アミノ基、カルボキシ基、アミノスルホニルアミノ基、ジエチルホスホノ基または
The compound according to any one of [1] to [10] or a pharmaceutically acceptable salt thereof.
[13] The compound according to any one of [1] to [12], wherein D is a single bond, −O (CH 2 ) n − or − saturated heterocycle −, and n is 2. Pharmaceutically acceptable salt.
[14] The compound according to any one of [1] to [13] or a pharmaceutically acceptable salt thereof, wherein E is a single bond or —O—.
[15] G is a hydroxy group, an amino group, a carboxy group, an aminosulfonylamino group, a diethylphosphono group or

Figure 2020071550
Figure 2020071550

で置換されているC1−6アルキル基、フェニル基、テトラヒドロピラニル基、テトヒドロフラニル基、ピペラジニル基またはモルホリニル基である、[1]〜[14]のいずれか一項に記載の化合物またはその医薬上許容される塩。
[16]以下に記載の化合物群から選択されるいずれかの化合物またはその医薬上許容される塩。
(1)3-アセチル-5,6-ジメトキシ-2-メチル-1-[4-(モルホリン-4-イル)フェニル]インドール
The compound according to any one of [1] to [14], which is a C 1-6 alkyl group, a phenyl group, a tetrahydropyranyl group, a tethydrofuranyl group, a piperazinyl group or a morpholinyl group substituted with. Or its pharmaceutically acceptable salt.
[16] Any compound selected from the compound group described below or a pharmaceutically acceptable salt thereof.
(1) 3-Acetyl-5,6-dimethoxy-2-methyl-1- [4- (morpholine-4-yl) phenyl] indole

Figure 2020071550
Figure 2020071550

(2)3-アセチル-5,6-ジメトキシ-2-メチル-1-{4-[3-(2-オキソ-2λ5-[1,3,2]ジオキサホスフィナン-2-イル)プロポキシ]フェニル}インドール(2) 3-Acetyl-5,6-dimethoxy-2-methyl-1-{4- [3- (2-oxo-2λ 5- [1,3,2] dioxaphosphinan-2-yl) propoxy ] Phenyl} indole

Figure 2020071550
Figure 2020071550

(3)3-アセチル-1-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-6-メトキシ-2-メチルインドール (3) 3-Acetyl-1-{4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -6-methoxy-2-methylindole

Figure 2020071550
Figure 2020071550

(4)3-アセチル-1-{4-[4-(3-アミノプロピル)ピペリジン-1-イル]フェニル}-5,6-ジメトキシ-2-メチルインドール (4) 3-Acetyl-1-{4- [4- (3-aminopropyl) piperidine-1-yl] phenyl} -5,6-dimethoxy-2-methylindole

Figure 2020071550
Figure 2020071550

(5)3-アセチル-6-メトキシ-2-メチル-1-[4-(4-(モルホリン-4-イル)ブトキシ)フェニル]インドール 塩酸塩 (5) 3-Acetyl-6-methoxy-2-methyl-1- [4- (4- (morpholine-4-yl) butoxy) phenyl] indole hydrochloride

Figure 2020071550
Figure 2020071550

(6)3-アセチル-5-フルオロ-6-メトキシ-2-メチル-1-(1-フェニルピペリジン-4-イル)インドール (6) 3-Acetyl-5-Fluoro-6-Methoxy-2-methyl-1- (1-phenylpiperidine-4-yl) indole

Figure 2020071550
Figure 2020071550

(7)3-{1-[4-(3-アセチル-5,6-ジメトキシ-2-メチルインドール-1-イル)フェニル]ピペリジン-4-イル}プロピオン酸 (7) 3- {1- [4- (3-Acetyl-5,6-dimethoxy-2-methylindole-1-yl) phenyl] piperidin-4-yl} propionic acid

Figure 2020071550
Figure 2020071550

(8)1-[4-(4-アセチルピペラジン-1-イル)フェニル]-5,6-ジメトキシ-2-メチルインドール-3-カルボキサミド (8) 1- [4- (4-Acetylpiperazin-1-yl) phenyl] -5,6-dimethoxy-2-methylindole-3-carboxamide

Figure 2020071550
Figure 2020071550

(9)1-{4-[4-(2-ヒドロキシエトキシ)ピペリジン-1-イル]フェニル}-6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン-3-カルボキサミド (9) 1- {4- [4- (2-Hydroxyethoxy) piperidine-1-yl] phenyl} -6-methoxy-2-methyl-1H-pyrrolo [3,2-c] pyridine-3-carboxamide

Figure 2020071550
Figure 2020071550

[17]以下に記載の化合物群から選択されるいずれかの化合物またはその医薬上許容される塩。
(1)2-{1-[4-(6-メトキシ-2-メチルイミダゾ[4,5-c]ピリジン-1-イル)フェニル]アゼチジン-3-イルオキシ}エタノール
[17] Any compound selected from the compound group described below or a pharmaceutically acceptable salt thereof.
(1) 2- {1- [4- (6-Methoxy-2-methylimidazole [4,5-c] pyridin-1-yl) phenyl] azetidine-3-yloxy} ethanol

Figure 2020071550
Figure 2020071550

(2)ジエチル(1-(4-(5-フルオロ-6-メトキシ-2-メチル-1H-ベンゾ[d]イミダゾール-1-イル)フェニル)アゼチジン-3-イルオキシ)メチルホスホネート (2) Diethyl (1- (4- (5-fluoro-6-methoxy-2-methyl-1H-benzo [d] imidazol-1-yl) phenyl) azetidine-3-yloxy) methylphosphonate

Figure 2020071550
Figure 2020071550

[18]以下に記載の化合物またはその医薬上許容される塩。
(1)4-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-6-メトキシ-1-メチル-1H-[1,7]ナフチリジン-2-オン
[18] The compounds described below or pharmaceutically acceptable salts thereof.
(1) 4- {4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -6-methoxy-1-methyl-1H- [1,7] naphthididine-2-one

Figure 2020071550
Figure 2020071550

[19]以下に記載の化合物またはその医薬上許容される塩。
(1)7-メトキシ-1-[4-(3-(モルホリン-4-イル)アゼチジン-1-イル)フェニル]-1H-キノリン-4-オン 二塩酸塩
[19] The compound described below or a pharmaceutically acceptable salt thereof.
(1) 7-Methoxy-1- [4- (3- (morpholine-4-yl) azetidine-1-yl) phenyl] -1H-quinoline-4-one dihydrochloride

Figure 2020071550
Figure 2020071550

[20]以下に記載の化合物またはその医薬上許容される塩。
(1)2-オキソ-4-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}-2H-クロメン-6-カルボキサミド
[20] The compound described below or a pharmaceutically acceptable salt thereof.
(1) 2-oxo-4- {4- [2- (tetrahydropyran-4-yloxy) ethoxy] phenyl} -2H-chromen-6-carboxamide

Figure 2020071550
Figure 2020071550

[21]以下に記載の化合物群から選択されるいずれかの化合物またはその医薬上許容される塩。
(1)1-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-7-メトキシ-4-メチル-1,4-ジヒドロ-2H-ピリド[3,4-b]ピラジン-3-オン
[21] Any compound selected from the compound group described below or a pharmaceutically acceptable salt thereof.
(1) 1- {4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -7-methoxy-4-methyl-1,4-dihydro-2H-pyrido [3,4-b] Pyrazine-3-on

Figure 2020071550
Figure 2020071550

(2)1-{4-[3-(2-スルファモイルアミノエトキシ)アゼチジン-1-イル]フェニル}-7-メトキシ-4-メチル-1,4-ジヒドロ-2H-ピリド[3,4-b]ピラジン-3-オン (2) 1-{4- [3- (2-Sulfamoylaminoethoxy) azetidine-1-yl] phenyl} -7-methoxy-4-methyl-1,4-dihydro-2H-pyrido [3,4 -b] Pyrazine-3-on

Figure 2020071550
Figure 2020071550

[22]以下に記載の化合物群から選択されるいずれかの化合物またはその医薬上許容される塩。
(1)8-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ナフタレン-2-カルボキサミド
[22] Any compound selected from the compound group described below or a pharmaceutically acceptable salt thereof.
(1) 8- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] phenyl} naphthalene-2-carboxamide

Figure 2020071550
Figure 2020071550

(2)3-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ベンゾ[b]チオフェン-5-カルボキサミド (2) 3- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] phenyl} benzo [b] thiophene-5-carboxamide

Figure 2020071550
Figure 2020071550

[23][1]〜[22]のいずれか一項に記載の化合物またはその医薬上許容される塩を有効成分として含有する医薬組成物。
[24][1]〜[22]のいずれか一項に記載の化合物またはその医薬上許容される塩を有効成分として含有するCDK8阻害剤。
[25]CDK8に関連する疾患を予防又は治療するために用いられる、[1]〜[22]のいずれか一項に記載の化合物またはその医薬上許容される塩を有効成分として含有する医薬組成物。
[26]骨代謝に関連する疾患を予防又は治療するために用いられる、[1]〜[22]のいずれか一項に記載の化合物またはその医薬上許容される塩を有効成分として含有する医薬組成物。
[27]癌の予防又は治療するために用いられる、[1]〜[22]のいずれか一項に記載の化合物またはその医薬上許容される塩を有効成分として含有する医薬組成物。
[23] A pharmaceutical composition containing the compound according to any one of [1] to [22] or a pharmaceutically acceptable salt thereof as an active ingredient.
[24] A CDK8 inhibitor containing the compound according to any one of [1] to [22] or a pharmaceutically acceptable salt thereof as an active ingredient.
[25] A pharmaceutical composition containing the compound according to any one of [1] to [22] or a pharmaceutically acceptable salt thereof, which is used for preventing or treating a disease related to CDK8, as an active ingredient. thing.
[26] A drug containing the compound according to any one of [1] to [22] or a pharmaceutically acceptable salt thereof as an active ingredient, which is used for preventing or treating a disease related to bone metabolism. Composition.
[27] A pharmaceutical composition containing, as an active ingredient, the compound according to any one of [1] to [22] or a pharmaceutically acceptable salt thereof, which is used for preventing or treating cancer.

本発明の化合物(I)は、CDK8に対する優れた阻害活性を有する。したがって、本発明の化合物(I)を有効成分として含有する医薬は、CDK8阻害剤として使用することができ、また、CDK8に関連する疾患、骨粗鬆症等の骨代謝に関連する疾患、癌等の細胞増殖性疾患の予防剤及び/または治療剤として有用である。 Compound (I) of the present invention has excellent inhibitory activity against CDK8. Therefore, the drug containing the compound (I) of the present invention as an active ingredient can be used as a CDK8 inhibitor, and also has a CDK8-related disease, a bone metabolism-related disease such as osteoporosis, and cells such as cancer. It is useful as a prophylactic and / or therapeutic agent for proliferative diseases.

本明細書中に用いられる用語および各記号の定義について、以下に説明する。
本明細書において、「ハロゲン原子」とは、フッ素原子、塩素原子、臭素原子またはヨウ素原子をいう。
Definitions of terms and symbols used herein will be described below.
As used herein, the term "halogen atom" refers to a fluorine atom, a chlorine atom, a bromine atom or an iodine atom.

本明細書において、「C1−6アルキル基」は、炭素数が1〜6の直鎖状または分岐鎖状の1価の飽和炭化水素基を意味する。当該「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、4−メチルペンチル、へキシル等が挙げられる。As used herein, the term "C 1-6 alkyl group" means a linear or branched monovalent saturated hydrocarbon group having 1 to 6 carbon atoms. Examples of the "C 1-6 alkyl group" include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, 4-methylpentyl, hexyl and the like. Be done.

本明細書において、「C3−8シクロアルキル基」は、炭素数が3〜8の飽和炭化水素環から誘導される一価の基を意味する。また、当該「C3−8シクロアルキル基」は、架橋していてもよい。当該「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、シクロオクチル等が挙げられる。As used herein, "C 3-8 cycloalkyl group" means a monovalent group derived from a saturated hydrocarbon ring having 3 to 8 carbon atoms. Moreover, the said "C 3-8 cycloalkyl group" may be crosslinked. Examples of the "C 3-8 cycloalkyl group" include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl and the like.

本明細書において、「C1−6アルコキシ基」は、酸素原子に前記「C1−6アルキル基」が結合した基、すなわち、炭素数が1〜6の直鎖または分岐鎖アルコキシ基を意味する。当該「C1−6アルコキシ基」としては、例えば、メトキシ、エトキシ、プロポキシ、イソプロポキシ、ブトキシ、イソブトキシ、sec−ブトキシ、tert−ブトキシ、ペンチルオキシ、イソペンチルオキシ、ネオペンチルオキシ、へキシルオキシ等が挙げられる。In the present specification, the "C 1-6 alkoxy group" means a group in which the "C 1-6 alkyl group" is bonded to an oxygen atom, that is, a linear or branched alkoxy group having 1 to 6 carbon atoms. do. Examples of the "C 1-6 alkoxy group" include methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec-butoxy, tert-butoxy, pentyloxy, isopentyloxy, neopentyloxy, hexyloxy and the like. Can be mentioned.

本明細書において、「C1−6アルキルカルボニル基」は、カルボニルに前記「C1−6アルキル基」が結合した一価の基を意味する。当該「C1−6アルキルカルボニル基」としては、例えば、アセチル、プロピオニル、ブチリル、イソブチリル、ペンタノイル、イソペンタノイル、1−メチルブチリル、ピバロイル、ヘキサノイル、イソヘキサノイル、3,3−ジメチルブチリル、1−エチルブチリル、4−メチルヘキサノイル、ヘプタノイル等が挙げられる。As used herein, the "C 1-6 alkylcarbonyl group" means a monovalent group in which the "C 1-6 alkyl group" is bonded to a carbonyl. Examples of the "C 1-6 alkylcarbonyl group" include acetyl, propionyl, butyryl, isobutyryl, pentanoyl, isopentanoyl, 1-methylbutyryl, pivaloyl, hexanoyl, isohexanoyl, 3,3-dimethylbutyryl, 1 -Ethylbutyryl, 4-methylhexanoyl, heptanoyl and the like can be mentioned.

本明細書において、「C1−6アルコキシC1−6アルキル基」は、前記「C1−6アルキル基」に前記「C1−6アルコキシ基」が結合した一価の基を意味する。当該「C1−6アルコキシC1−6アルキル基」としては、例えば、メトキシメチル、2−メトキシエチル、エトキシメチル、2−エトキシエチル、3−エトキシプロピル、2−プロポキシエチル、2−イソプロポキシエチル、2−ブトキシエチル、2−イソブトキシエチル、sec−ブトキシメチル、tert−ブトキシメチル、2−ペンチルオキシエチル、イソペンチルオキシメチル、2−ネオペンチルオキシエチル、2−へキシルオキシエチル等が挙げられる。In the present specification, "C 1-6 alkoxy C 1-6 alkyl group" means the "C 1-6 alkyl group" in the monovalent radical "C 1-6 alkoxy group" is bonded. Examples of the "C 1-6 alkoxy C 1-6 alkyl group" include methoxymethyl, 2-methoxyethyl, ethoxymethyl, 2-ethoxyethyl, 3-ethoxypropyl, 2-propoxyethyl, and 2-isopropoxyethyl. , 2-Butoxyethyl, 2-Isobutoxyethyl, sec-Butoxymethyl, tert-Butoxymethyl, 2-Pentyloxyethyl, Isopentyloxymethyl, 2-Neopentyloxyethyl, 2-Hexyloxyethyl and the like. ..

本明細書において、「C1−6アルキルチオカルボニル基」は、チオカルボニルに前記「C1−6アルキル基」が結合した一価の基を意味する。当該「C1−6アルキルチオカルボニル基」としては、例えば、メチルチオカルボニル、エチルチオカルボニル、プロピルチオカルボニル、ブチルチオカルボニル、イソブチルチオカルボニル、ペンチルチオカルボニル、3,3−ジメチルブチルチオカルボニル、1−エチルブチルチオカルボニル、ヘキシルチオカルボニル等が挙げられる。As used herein, the "C 1-6 alkyl thiocarbonyl group" means a monovalent group in which the "C 1-6 alkyl group" is bonded to thiocarbonyl. Examples of the "C 1-6 alkylthiocarbonyl group" include methylthiocarbonyl, ethylthiocarbonyl, propylthiocarbonyl, butylthiocarbonyl, isobutylthiocarbonyl, pentylthiocarbonyl, 3,3-dimethylbutylthiocarbonyl and 1-ethyl. Examples thereof include butylthiocarbonyl and hexylthiocarbonyl.

本明細書において、「C1−6アルキルカルボニルオキシ基」は、酸素原子に前記「C1−6アルキルカルボニル基」が結合した一価の基を意味する。当該「C1−6アルキルカルボニルオキシ基」としては、例えば、アセチルオキシ、プロピオニルオキシ、ブチリルオキシ、イソブチリルオキシ、ペンタノイルオキシ、イソペンタノイルオキシ、1−メチルブチリルオキシ、ピバロイルオキシ、ヘキサノイルオキシ、イソヘキサノイルオキシ、3,3−ジメチルブチリルオキシ、1−エチルブチリルオキシ、4−メチルヘキサノイルオキシ、ヘプタノイルオキシ等が挙げられる。As used herein, the "C 1-6 alkylcarbonyloxy group" means a monovalent group in which the "C 1-6 alkylcarbonyl group" is bonded to an oxygen atom. Examples of the "C 1-6 alkylcarbonyloxy group" include acetyloxy, propionyloxy, butyryloxy, isobutyryloxy, pentanoyloxy, isopentanoyloxy, 1-methylbutyryloxy, pivaloyloxy and hexanoyloxy. , Isohexanoyloxy, 3,3-dimethylbutylyloxy, 1-ethylbutyryloxy, 4-methylhexanoyloxy, heptanoyloxy and the like.

本明細書において、「C1−6アルキルカルボニルアミノ基」は、窒素原子に前記「C1−6アルキルカルボニル基」が結合した一価の基を意味する。当該「C1−6アルキルカルボニルアミノ基」としては、例えば、メチルカルボニルアミノ、エチルカルボニルアミノ、プロピルカルボニルアミノ、イソプロピルカルボニルアミノ、ブチルカルボニルアミノ、イソブチルカルボニルアミノ、sec−ブチルカルボニルアミノ、tert−ブチルカルボニルアミノ、ペンチルカルボニルアミノ、イソペンチルカルボニルアミノ、ネオペンチルカルボニルアミノ、1−エチルプロピルカルボニルアミノ、へキシルカルボニルアミノ等が挙げられる。As used herein, the "C 1-6 alkylcarbonylamino group" means a monovalent group in which the "C 1-6 alkylcarbonyl group" is bonded to a nitrogen atom. Examples of the "C 1-6 alkylcarbonylamino group" include methylcarbonylamino, ethylcarbonylamino, propylcarbonylamino, isopropylcarbonylamino, butylcarbonylamino, isobutylcarbonylamino, sec-butylcarbonylamino, and tert-butylcarbonyl. Examples thereof include amino, pentylcarbonylamino, isopentylcarbonylamino, neopentylcarbonylamino, 1-ethylpropylcarbonylamino, hexylcarbonylamino and the like.

本明細書において、「C1−6アルキルスルホニル基」は、スルホニル基に前記「C1−6アルキル基」が結合した基、すなわち、炭素数が1−6の直鎖または分岐鎖アルキルスルホニル基を意味する。当該「C1−6アルキルスルホニル基」としては、例えば、メチルスルホニル、エチルスルホニル、プロピルスルホニル、イソプロピルスルホニル、ブチルスルホニル、イソブチルスルホニル、sec−ブチルスルホニル、tert−ブチルスルホニル、ペンチルスルホニル、イソペンチルスルホニル、ネオペンチルスルホニル、1−エチルプロピルスルホニル、へキシルスルホニル等が挙げられる。In the present specification, the "C 1-6 alkylsulfonyl group" is a group in which the "C 1-6 alkyl group" is bonded to a sulfonyl group, that is, a linear or branched alkylsulfonyl group having 1-6 carbon atoms. Means. Examples of the "C 1-6 alkylsulfonyl group" include methylsulfonyl, ethylsulfonyl, propylsulfonyl, isopropylsulfonyl, butylsulfonyl, isobutylsulfonyl, sec-butylsulfonyl, tert-butylsulfonyl, pentylsulfonyl, isopentylsulfonyl, and the like. Examples thereof include neopentylsulfonyl, 1-ethylpropylsulfonyl, and hexylsulfonyl.

本明細書において、「C1−6アルキルスルホニルアミノ基」は、窒素原子に前記「C1−6アルキルスルホニル基」が結合した一価の基を意味する。当該「C1−6アルキルスルホニルアミノ基」としては、例えば、メチルスルホニルアミノ、エチルスルホニルアミノ、プロピルスルホニルアミノ、イソプロピルスルホニルアミノ、ブチルスルホニルアミノ、イソブチルスルホニルアミノ、sec−ブチルスルホニルアミノ、tert−ブチルスルホニルアミノ、ペンチルスルホニルアミノ、イソペンチルスルホニルアミノ、ネオペンチルスルホニルアミノ、1−エチルプロピルスルホニルアミノ、へキシルスルホニルアミノ等が挙げられる。As used herein, the "C 1-6 alkylsulfonylamino group" means a monovalent group in which the "C 1-6 alkylsulfonyl group" is bonded to a nitrogen atom. Examples of the "C 1-6 alkylsulfonylamino group" include methylsulfonylamino, ethylsulfonylamino, propylsulfonylamino, isopropylsulfonylamino, butylsulfonylamino, isobutylsulfonylamino, sec-butylsulfonylamino, and tert-butylsulfonyl. Examples thereof include amino, pentylsulfonylamino, isopentylsulfonylamino, neopentylsulfonylamino, 1-ethylpropylsulfonylamino, hexylsulfonylamino and the like.

本明細書において、「C1−6アルキルスルファニル基」は、硫黄原子に前記「C1−6アルキル基」が結合した基、すなわち、炭素数が1−6の直鎖または分岐鎖アルキルスルファニル基を意味する。当該「C1−6アルキルスルファニル基」としては、例えば、メチルスルファニル、エチルスルファニル、プロピルスルファニル、イソプロピルスルファニル、ブチルスルファニル、イソブチルスルファニル、sec−ブチルスルファニル、tert−ブチルスルファニル、ペンチルスルファニル、イソペンチルスルファニル、ネオペンチルスルファニル、1−エチルプロピルスルファニル、へキシルスルファニル等が挙げられる。In the present specification, the "C 1-6 alkyl sulfanyl group" is a group in which the "C 1-6 alkyl group" is bonded to a sulfur atom, that is, a linear or branched alkyl sulfanyl group having 1-6 carbon atoms. Means. Examples of the "C 1-6 alkyl sulfanyl group" include methyl sulfanyl, ethyl sulfanyl, propyl sulfanyl, isopropyl sulfanyl, butyl sulfanyl, isobutyl sulfanyl, sec-butyl sulfanyl, tert-butyl sulfanyl, pentyl sulfanyl, and isopentyl sulfanyl. Examples thereof include neopentyl sulfanyl, 1-ethylpropyl sulfanyl, hexyl sulfanyl and the like.

本明細書において、「C3−8シクロアルキル基」は、炭素数が3−8の飽和炭化水素環から誘導される一価の基を意味する。また、当該「C3−8シクロアルキル基」は、架橋していてもよい。当該「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、シクロオクチル、ビシクロ[1,1,1]ペンチル等が挙げられる。As used herein, "C 3-8 cycloalkyl group" means a monovalent group derived from a saturated hydrocarbon ring having 3-8 carbon atoms. Moreover, the said "C 3-8 cycloalkyl group" may be crosslinked. Examples of the "C 3-8 cycloalkyl group" include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, bicyclo [1,1,1] pentyl and the like.

本明細書において、「アミノC1−6アルキルカルボニルオキシ基」は、前記「C1−6アルキルカルボニルオキシ基」にアミノ基が結合した基を意味する。当該「アミノC1−6アルキルカルボニルオキシ」としては、例えば、アミノアセチルオキシ、アミノプロピオニルオキシ、アミノブチリルオキシ、アミノイソブチリルオキシ、アミノペンタノイルオキシ、アミノイソペンタノイルオキシ、アミノ−1−メチルブチリルオキシ、アミノピバロイルオキシ、アミノヘキサノイルオキシ、アミノイソヘキサノイルオキシ、アミノ−3,3−ジメチルブチリルオキシ、アミノ−1−エチルブチリルオキシ、アミノ−4−メチルヘキサノイルオキシ、アミノヘプタノイルオキシ等が挙げられる。In the present specification, the "amino C 1-6 alkylcarbonyloxy group" means a group in which an amino group is bonded to the "C 1-6 alkylcarbonyloxy group". Examples of the "amino C 1-6 alkylcarbonyloxy" include aminoacetyloxy, aminopropionyloxy, aminobutyryloxy, aminoisobutyryloxy, aminopentanoyloxy, aminoisopentanoyloxy, and amino-1-. Methylbutyryloxy, Aminopivaloyloxy, Aminohexanoyloxy, Aminoisohexanoyloxy, Amino-3,3-Dimethylbutyryloxy, Amino-1-ethylbutyryloxy, Amino-4-methylhexanoyloxy , Aminoheptanoyloxy and the like.

本明細書において、「モノまたはジ−C1−6アルキルアミノ基」は、窒素原子に前記「C1−6アルキル基」が一つあるいは二つ結合した基、すなわち、炭素数が1−6の直鎖または分岐鎖アルキルアミノ基を意味する。当該「モノまたはジ−C1−6アルキルアミノ基」としては、例えば、メチルアミノ、エチルアミノ、プロピルアミノ、イソプロピルアミノ、ブチルアミノ、イソブチルアミノ、sec−ブチルアミノ、tert−ブチルアミノ、ペンチルアミノ、イソペンチルアミノ、ネオペンチルアミノ、1−エチルプロピルアミノ、へキシルアミノ、ジメチルアミノ、ジエチルアミノ、ジプロピルアミノ、ジイソプロピルアミノ、ジブチルアミノ、メチルエチルアミノ等が挙げられる。In the present specification, the "mono or di-C 1-6 alkylamino group" is a group in which one or two of the "C 1-6 alkyl groups" are bonded to a nitrogen atom, that is, the number of carbon atoms is 1-6. Means a linear or branched alkylamino group of. Examples of the "mono or di-C 1-6 alkylamino group" include methylamino, ethylamino, propylamino, isopropylamino, butylamino, isobutylamino, sec-butylamino, tert-butylamino and pentylamino. Examples thereof include isopentylamino, neopentylamino, 1-ethylpropylamino, hexylamino, dimethylamino, diethylamino, dipropylamino, diisopropylamino, dibutylamino and methylethylamino.

本明細書において、「C1−6アルコキシカルボニル基」は、カルボニルに前記「C1−6アルコキシ基」が結合した基、すなわち、炭素数が1〜6の直鎖または分岐鎖アルコキシカルボニル基を意味する。当該「C1−6アルコキシカルボニル基」としては、例えば、メトキシカルボニル、エトキシカルボニル、プロポキシカルボニル、イソプロポキシカルボニル、ブトキシカルボニル、イソブトキシカルボニル、sec−ブトキシカルボニル、tert−ブトキシカルボニル、ペンチルオキシカルボニル、イソペンチルオキシカルボニル、ネオペンチルオキシカルボニル、1−エチルプロピルオキシカルボニル、へキシルオキシカルボニル等が挙げられる。In the present specification, the "C 1-6 alkoxycarbonyl group" is a group in which the "C 1-6 alkoxy group" is bonded to carbonyl, that is, a linear or branched alkoxycarbonyl group having 1 to 6 carbon atoms. means. Examples of the "C 1-6 alkoxycarbonyl group" include methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, butoxycarbonyl, isobutoxycarbonyl, sec-butoxycarbonyl, tert-butoxycarbonyl, pentyloxycarbonyl and iso. Examples thereof include pentyloxycarbonyl, neopentyloxycarbonyl, 1-ethylpropyloxycarbonyl, hexyloxycarbonyl and the like.

本明細書において、「C3−8シクロアルケニレン基」は、1個以上の炭素−炭素二重結合を有し、炭素数が3−8の炭化水素環から誘導されるC3−8シクロアルケニル基由来の2価の基を意味する。当該C3−8シクロアルケニル基としては、例えば、3−シクロブテニル、3−シクロペンテニル、2−シクロヘキセニル、3−シクロヘキセニル、2−シクロヘプテニル、3−シクロヘプテニル、2−シクロオクテニル、3−シクロオクテニル等が挙げられる。As used herein, a "C 3-8 cycloalkenylene group" is a C 3-8 cycloalkenyl derived from a hydrocarbon ring having one or more carbon-carbon double bonds and 3-8 carbon atoms. It means a divalent group derived from a group. Examples of the C 3-8 cycloalkenyl group include 3-cyclobutenyl, 3-cyclopentenyl, 2-cyclohexenyl, 3-cyclohexenyl, 2-cycloheptenyl, 3-cycloheptenyl, 2-cyclooctenyl, 3-cyclooctenyl and the like. Be done.

本明細書において、「C6−14アリール基」は、炭素数が6−14の芳香族炭化水素基を意味する。当該「C6−14アリール基」としては、例えば、フェニル、ナフチル(例、1−ナフチル、2−ナフチル)、アセナフチレニル、アズレニル、アントリル、フェナントリル等が挙げられる。As used herein, the "C 6-14 aryl group" means an aromatic hydrocarbon group having 6-14 carbon atoms. Examples of the "C 6-14 aryl group" include phenyl, naphthyl (eg, 1-naphthyl, 2-naphthyl), acenaphthylenyl, azulenyl, anthryl, phenanthryl and the like.

本明細書において、「C6−14アリーレン基」は、炭素数が6−14の芳香族炭化水素基から誘導されるC6−14アリール基由来の2価の基を意味する。当該「C6−14アリール基」としては、例えば、フェニル、ナフチル(例、1−ナフチル、2−ナフチル)、アセナフチレニル、アズレニル、アントリル、フェナントリル等が挙げられる。As used herein, "C 6-14 arylene group" means a divalent group derived from a C 6-14 aryl group derived from an aromatic hydrocarbon group having 6-14 carbon atoms. Examples of the "C 6-14 aryl group" include phenyl, naphthyl (eg, 1-naphthyl, 2-naphthyl), acenaphthylenyl, azulenyl, anthryl, phenanthryl and the like.

本明細書において、「ヒドロキシC1−6アルキル基」は、前記「C1−6アルキル基」に1個のヒドロキシ基が結合した基であり、例えば、1−ヒドロキシメチル基、1−ヒドロキシエチル基、1−ヒドロキシプロピル基、2−ヒドロキシエチル基又は3−ヒドロキシプロピル基等が挙げられる。In the present specification, the "hydroxy C 1-6 alkyl group" is a group in which one hydroxy group is bonded to the "C 1-6 alkyl group", and is, for example, a 1-hydroxymethyl group or a 1-hydroxyethyl group. Groups, 1-hydroxypropyl groups, 2-hydroxyethyl groups, 3-hydroxypropyl groups and the like can be mentioned.

本明細書において、「ヒドロキシC1−6アルコキシ基」は、前記「C1−6アルコキシ基」に1個のヒドロキシ基が結合した基であり、例えば、1−ヒドロキシメトキシ基、1−ヒドロキシエトキシ基、1−ヒドロキシプロポキシ基、2−ヒドロキシエトキシ基又は3−ヒドロキシプロポキシ基等が挙げられる。In the present specification, the "hydroxy C 1-6 alkoxy group" is a group in which one hydroxy group is bonded to the "C 1-6 alkoxy group", and is, for example, a 1-hydroxymethoxy group or a 1-hydroxyethoxy group. Groups, 1-hydroxypropoxy groups, 2-hydroxyethoxy groups, 3-hydroxypropoxy groups and the like can be mentioned.

本明細書において、「カルボキシC1−6アルキル基」は、前記「C1−6アルキル基」にカルボキシ基が結合した基を意味する。当該「カルボキシC1−6アルキル基」としては、例えば、カルボキシメチル、カルボキシエチル、カルボキシプロピル、カルボキシイソプロピル、カルボキシブチル、カルボキシイソブチル、カルボキシ−sec−ブチル、カルボキシ−tert−ブチル、カルボキシペンチル、カルボキシイソペンチル、カルボキシネオペンチル、カルボキシ−1−エチルプロピル、カルボキシへキシル等が挙げられる。In the present specification, the "carboxy C 1-6 alkyl group" means a group in which a carboxy group is bonded to the "C 1-6 alkyl group". Examples of the "carboxyC 1-6 alkyl group" include carboxymethyl, carboxyethyl, carboxypropyl, carboxyisopropyl, carboxybutyl, carboxyisobutyl, carboxy-sec-butyl, carboxy-tert-butyl, carboxypentyl, and carboxyiso. Examples thereof include pentyl, carboxyneopentyl, carboxy-1-ethylpropyl, carboxyhexyl and the like.

本明細書において、「カルボキシC1−6アルキルカルボニルオキシ基」は、前記「C1−6アルキルカルボニルオキシ基」にカルボキシ基が結合した基を意味する。当該「カルボキシC1−6アルキルカルボニルオキシ」としては、例えば、カルボキシアセチルオキシ、カルボキシプロピオニルオキシ、カルボキシブチリルオキシ、カルボキシイソブチリルオキシ、カルボキシペンタノイルオキシ、カルボキシイソペンタノイルオキシ、カルボキシ−1−メチルブチリルオキシ、カルボキシピバロイルオキシ、カルボキシヘキサノイルオキシ、カルボキシイソヘキサノイルオキシ、カルボキシ−3,3−ジメチルブチリルオキシ、カルボキシ−1−エチルブチリルオキシ、カルボキシ−4−メチルヘキサノイルオキシ、カルボキシヘプタノイルオキシ等が挙げられる。In the present specification, the "carboxy C 1-6 alkylcarbonyloxy group" means a group in which a carboxy group is bonded to the "C 1-6 alkylcarbonyloxy group". Examples of the "carboxy C 1-6 alkylcarbonyloxy" include carboxyacetyloxy, carboxypropionyloxy, carboxybutylyloxy, carboxyisobutyryloxy, carboxypentanoyloxy, carboxyisopentanoyloxy, and carboxy-1- Methylbutyryloxy, carboxypivaloyloxy, carboxyhexanoyloxy, carboxyisohexanoyloxy, carboxy-3,3-dimethylbutylyloxy, carboxy-1-ethylbutyryloxy, carboxy-4-methylhexanoyloxy , Carboxyheptanoyloxy and the like.

本明細書において、「複素環基」は、酸素原子、窒素原子および硫黄原子からなる群より選択されるヘテロ原子を少なくとも1つ含有する1価の単環式芳香族複素環基および縮合芳香族複素環基を意味する。単環式芳香族複素環基としては、5または6員の単環式芳香族複素環基が挙げられ、例えば、フリル、チエニル、ピロリル、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、トリアゾリル、テトラゾリル、チアジアゾリル、オキサジアゾリル、ピリジル、ピリダジニル、ピリミジニル等が挙げられる。縮合芳香族複素環基としては、2環または3環系の縮合芳香族複素環基が挙げられ、例えば、インドリル、イソインドリル、インダゾリル、ベンゾフラニル、ベンゾチオフェニル、ベンズイミダゾリル、ベンゾオキサゾリル、ベンゾチアゾリル、キノリル、イソキノリル、ベンズオキサジニル、ベンゾチアジニル、フロ[2,3−b]ピリジル、チエノ[2,3−b]ピリジル、ナフチリジニル、イミダゾピリジル、オキサゾロピリジル、チアゾロピリジル、キノリル、カルバゾリル、ジベンゾチオフェニル等が挙げられる。 As used herein, the "heterocyclic group" is a monovalent monocyclic aromatic heterocyclic group containing at least one heteroatom selected from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, and a condensed aromatic. It means a heterocyclic group. Examples of the monocyclic aromatic heterocyclic group include a 5- or 6-membered monocyclic aromatic heterocyclic group, for example, frill, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, triazolyl, tetrazolyl, Examples thereof include thiadiazolyl, oxadiazolyl, pyridyl, pyridazinyl, pyrimidinyl and the like. Examples of the fused aromatic heterocyclic group include bicyclic or tricyclic condensed aromatic heterocyclic groups, for example, indolyl, isoindrill, indazolyl, benzofuranyl, benzothiophenyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, and the like. Quinoline, isoquinolyl, benzoxazinyl, benzothiazinyl, flo [2,3-b] pyridyl, thieno [2,3-b] pyridyl, naphthilidinyl, imidazolepyridyl, oxazolopyridyl, thiazolopyridyl, quinolyl, carbazolyl, dibenzo Examples include thiophenyl.

本明細書において、「飽和複素環基」は、硫黄原子、酸素原子又は/及び窒素原子を1−3個含む飽和5−7員複素環基であり、例えば、テトラヒドロピラニル、テトラヒドロフラニル、オキソテトラヒドロフラニル、モルホリニル、チオモルホリニル、1−オキソチオモルホリニル、1,1−ジオキソチオモルホリニル、ピロリジニル、ピロリニル、イミダゾリジニル、ピラゾリジニル、ピペリジニル、ピペラジニル、オキサゾリジニル、イソキサゾリジニル、チアゾリジニル、1,4−ジオキサニル、1,1−ジオキソヘキサヒドロチオピラニル等が挙げられる。 As used herein, a "saturated heterocyclic group" is a saturated 5-7-membered heterocyclic group containing 1-3 sulfur, oxygen or / / nitrogen atoms, eg, tetrahydropyranyl, tetrahydrofuranyl, oxo. Tetrahydrofuranyl, morpholinyl, thiomorpholinyl, 1-oxothiomorpholinyl, 1,1-dioxothiomorpholinyl, pyrrolidinyl, pyrrolinyl, imidazolidinyl, pyrazoridinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, 1, Examples thereof include 4-dioxanyl and 1,1-dioxohexahydrothiopyranyl.

本明細書において、「−複素環基−」は、複素環由来の2価の基を示し、当該複素環の単環式芳香族複素環としては、例えば、フラン、チオフェン、ピロール、イミダゾール、ピラゾール、オキサゾール、イソキサゾール、チアゾール、トリアゾール、テトラゾール、チアジアゾール、オキサジアゾール、ピリジン、ピリダジン、ピリミジン等が挙げられる。縮合芳香族複素環としては、例えば、インドリン、イソインドリン、インダゾリン、ベンゾフラン、ベンゾチオフェン、ベンズイミダゾール、ベンゾオキサゾール、ベンゾチアゾール、キノリン、イソキノリン、ベンズオキサジン、ベンゾチアジン、フロ[2,3−b]ピリジン、チエノ[2,3−b]ピリジン、ナフチリジン、イミダゾピリジン、オキサゾロピリジン、チアゾロピリジン、キノリン、カルバゾール、ジベンゾチオフェン等が挙げられる。 In the present specification, "-heterocyclic group-" indicates a divalent group derived from the heterocycle, and examples of the monocyclic aromatic heterocycle of the heterocycle include furan, thiophene, pyrrole, imidazole, and pyrazole. , Oxazole, isoxazole, thiazole, triazole, tetrazole, thiaziazole, oxaziazole, pyridine, pyridazine, pyrimidine and the like. Examples of the fused aromatic heterocycle include indolin, isoindolin, indazoline, benzofuran, benzothiophene, benzimidazole, benzoxazole, benzothiazole, quinoline, isoquinoline, benzoxazine, benzothiazine, flo [2,3-b] pyridine, and the like. Examples thereof include thieno [2,3-b] pyridine, naphthylidine, imidazopyridine, oxazolopyridine, thiazolopyridine, quinoline, carbazole, dibenzothiophene and the like.

本明細書において、「−飽和複素環基−」は、飽和複素環由来の2価の基を示し、当該飽和複素環としては、例えば、アゼチジン、テトラヒドロピラン、テトラヒドロフラン、モルホリン、チオモルホリン、1−オキソチオモルホリン、1,1−ジオキソチオモルホリン、ピロリジン、ピロリン、イミダゾリジン、ピラゾリジン、ピペリジン、ピペラジン、オキサゾリジン、イソキサゾリジン、チアゾリジン等が挙げられる。 In the present specification, "-saturated heterocyclic group-" indicates a divalent group derived from the saturated heterocycle, and examples of the saturated heterocycle include azetidine, tetrahydropyrrane, tetrahydrofuran, morpholine, thiomorpholine, 1-. Examples thereof include oxothiomorpholine, 1,1-dioxothiomorpholine, pyrrolidine, pyrrolidine, imidazolidine, pyrazolidine, piperazine, piperazine, oxazolidine, isoxazolidine, thiazolidine and the like.

「置換されていてもよい」とは、無置換又は1−3のいずれかの置換である。2又は3置換の場合、各置換基は同一であっても異なっていてもよい。 "May be substituted" is either non-replacement or substitution of 1-3. In the case of 2 or 3 substitutions, each substituent may be the same or different.

本明細書において、「その医薬上許容される塩」とは、医薬として使用することができる塩を示す。本発明の化合物では、酸性基または塩基性基を有する場合に、塩基または酸と反応させることにより、塩基性塩または酸性塩にすることができるので、その塩を示す。
本発明の化合物(I)の医薬上許容される「塩基性塩」としては、好適には、ナトリウム塩、カリウム塩、リチウム塩のようなアルカリ金属塩;マグネシウム塩、カルシウム塩のようなアルカリ土類金属塩;N−メチルモルホリン塩、トリエチルアミン塩、トリブチルアミン塩、ジイソプロピルエチルアミン塩、ジシクロヘキシルアミン塩、N−メチルピペリジン塩、ピリジン塩、4−ピロリジノピリジン塩、ピコリン塩のような有機塩基塩類またはグリシン塩、リジン塩、アルギニン塩、オルニチン塩、グルタミン酸塩、アスパラギン酸塩のようなアミノ酸塩であり、好適には、アルカリ金属塩である。
As used herein, the term "the pharmaceutically acceptable salt" refers to a salt that can be used as a medicine. In the compound of the present invention, when it has an acidic group or a basic group, it can be made into a basic salt or an acidic salt by reacting with a base or an acid, and thus the salt thereof is shown.
The pharmaceutically acceptable "basic salt" of the compound (I) of the present invention is preferably an alkali metal salt such as a sodium salt, a potassium salt or a lithium salt; an alkaline soil such as a magnesium salt or a calcium salt. Metal salts; organic base salts such as N-methylmorpholin salt, triethylamine salt, tributylamine salt, diisopropylethylamine salt, dicyclohexylamine salt, N-methylpiperidin salt, pyridine salt, 4-pyrrolidinopyridine salt, picolin salt or It is an amino acid salt such as glycine salt, lysine salt, arginine salt, ornithine salt, glutamate salt, asparaginate salt, and is preferably an alkali metal salt.

本発明の化合物(I)の医薬上許容される「酸性塩」としては、好適には、フッ化水素酸塩、塩酸塩、臭化水素酸塩、ヨウ化水素酸塩のようなハロゲン化水素酸塩、硝酸塩、過塩素酸塩、硫酸塩、リン酸塩等の無機酸塩;メタンスルホン酸塩、トリフルオロメタンスルホン酸塩、エタンスルホン酸塩のような低級アルカンスルホン酸塩、ベンゼンスルホン酸塩、p−トルエンスルホン酸塩のようなアリ−ルスルホン酸塩、酢酸塩、リンゴ酸塩、フマール酸塩、コハク酸塩、クエン酸塩、アスコルビン酸塩、酒石酸塩、蓚酸塩、マレイン酸塩等の有機酸塩;及び、グリシン塩、リジン塩、アルギニン塩、オルニチン塩、グルタミン酸塩、アスパラギン酸塩のようなアミノ酸塩であり、最も好適には、ハロゲン化水素酸塩(特に、塩酸塩)である。 The pharmaceutically acceptable "acidic salt" of the compound (I) of the present invention is preferably hydrogen halide such as hydrofluoride, hydrochloride, hydrobromide, hydroiodide. Inorganic acid salts such as acid salts, nitrates, perchlorates, sulfates, phosphates; lower alkane sulfonates such as methane sulfonate, trifluoromethane sulfonate, ethane sulfonate, benzene sulfonate , Allyl sulfonates such as p-toluene sulfonate, acetate, malate, fumarate, succinate, citrate, ascorbate, tartrate, oxalate, maleate, etc. Organic acid salts; and amino acid salts such as glycine salts, lysine salts, arginine salts, ornithine salts, glutamates, asparaginates, most preferably hydrohalogenates (particularly hydrochlorides). ..

本明細書において、「予防」とは、例えば、病気または症状に関連する何らかの因子により、発症の危険性が高いと予想される当該病気または症状を発症していない患者あるいは発症しているが自覚症状のない患者に対し、本発明の化合物(I)を含む医薬を投与すること、あるいは当該病気または症状の治療後、当該病気または症状の再発が懸念される患者に対し、本発明の化合物(I)を含む医薬を投与することである。
「治療」とは、病気または症状を治癒させることである。
As used herein, the term "prevention" means, for example, a patient who does not develop the disease or symptom that is expected to be at high risk of developing the disease or symptoms due to some factor related to the disease or symptom, or who is aware that the disease or symptom has developed. The compound of the present invention ( I) is to administer a drug containing.
"Treatment" is the cure of an illness or symptom.

以下に、式(I)中の各記号の定義について詳述する。
は、水素原子、ハロゲン原子、シアノ基、C1−6アルキル基、C1−6アルコキシ基、C1−6アルキルスルファニル基、C1−6アルキルカルボニル基、カルバモイル基または複素環基を示す。
は、好ましくは、ハロゲン原子(例、塩素原子)、シアノ基、C1−6アルコキシ基(例、メトキシ)、C1−6アルキルスルファニル基(例、メチルスルファニル)、C1−6アルキルカルボニル基(例、アセチル)、カルバモイル基または複素環基(例、イミダゾリル、フリル、ピリジル)であり、より好ましくは、シアノ基、メトキシ基、メチルスルファニル基、アセチル基、カルバモイル基またはイミダゾリル基である。
The definition of each symbol in the formula (I) will be described in detail below.
R 1 contains a hydrogen atom, a halogen atom, a cyano group, a C 1-6 alkyl group, a C 1-6 alkoxy group, a C 1-6 alkyl sulfanyl group, a C 1-6 alkyl carbonyl group, a carbamoyl group or a heterocyclic group. show.
R 1 is preferably a halogen atom (eg, chlorine atom), a cyano group, a C 1-6 alkoxy group (eg, methoxy), a C 1-6 alkyl sulfanyl group (eg, methyl sulfanyl), a C 1-6 alkyl. It is a carbonyl group (eg, acetyl), a carbamoyl group or a heterocyclic group (eg, imidazolyl, furyl, pyridyl), more preferably a cyano group, a methoxy group, a methylsulfanyl group, an acetyl group, a carbamoyl group or an imidazolyl group. ..

T及びUは、いずれか一方が=N−である場合、もう一方は=CR−を示し、あるいは、T及びUの両方が、=CR−を示す。
T及びUは、好ましくは、Tが=CR−であり、Uが=N−であるか、あるいは、T及びUの両方が、=CR−であり、より好ましくは、Tが=CH−であり、Uが=N−であるか、あるいは、Tが=CH−であり、Uが=CR−であり、Rが、水素原子、フッ素原子またはメトキシ基である。
When one of T and U is = N−, the other indicates = CR 2 −, or both T and U indicate = CR 2 −.
For T and U, T is preferably = CR 2- and U is = N-, or both T and U are = CR 2- , and more preferably T is = CH. -, U is = N-, or T is = CH-, U is = CR 2- , and R 2 is a hydrogen atom, a fluorine atom or a methoxy group.

は、水素原子、ハロゲン原子、C1−6アルキル基またはC1−6アルコキシ基を示す。
は、好ましくは、水素原子、ハロゲン原子(例、フッ素原子)、C1−6アルキル基(例、メチル)またはC1−6アルコキシ基(例、メトキシ)であり、より好ましくは、水素原子、フッ素原子またはメトキシ基である。
A環は、
R 2 represents a hydrogen atom, a halogen atom, a C 1-6 alkyl group or a C 1-6 alkoxy group.
R 2 is preferably a hydrogen atom, a halogen atom (eg, fluorine atom), a C 1-6 alkyl group (eg, methyl) or a C 1-6 alkoxy group (eg, methoxy), and more preferably hydrogen. It is an atom, a hydrogen atom or a methoxy group.
Ring A is

Figure 2020071550
Figure 2020071550

を示す。
は、−B−D−E−G[式中、Bは、C6−14アリーレン基、C3−8シクロアルケニレン基、−複素環−または−飽和複素環−を示し、Dは、単結合、−O(CH−、−(CHO−または−飽和複素環−を示し、nは、1〜4から選択されるいずれか1の整数を示し、Eは、単結合、−O−、−N(R10)−、−CO−または−CONH−を示し、R10は、水素原子または置換基群αから選択されるいずれかの基で置換されていてもよいC1−6アルキル基を示し、Gは、水素原子、置換基群αから選択されるいずれかの基で置換されていてもよいC1−6アルキル基、置換基群αから選択されるいずれかの基で置換されていてもよいC3−8シクロアルキル基、置換基群αから選択されるいずれかの基で置換されていてもよいC6−14アリール基、置換基群αから選択されるいずれかの基で置換されていてもよい複素環基または置換基群αから選択されるいずれかの基で置換されていてもよい飽和複素環基を示し、置換基群αは、複素環基、飽和複素環基、ハロゲン原子、シアノ基、C1−6アルキルカルボニル基、C1−6アルキルカルボニルオキシ基、C1−6アルキルカルボニルアミノ基、C1−6アルコキシカルボニル基、C1−6アルコキシC1−6アルキル基、ヒドロキシC1−6アルキル基、ヒドロキシC1−6アルコキシ基、C1−6アルキルスルホニル基、C1−6アルキルスルホニルアミノ基、アミノスルホニルアミノ基、カルボキシ基、カルボキシC1−6アルキルカルボニルオキシ基、カルバモイル基、ヒドロキシ基、C1−6アルコキシ基、C1−6アルキル基、C3−8シクロアルキル基、アミノ基、アミノC1−6アルキルカルボニルオキシ基、モノまたはジ−C1−6アルキルアミノ基、4−ヒドロキシテトラヒドロフラン−3−イルオキシ基、モノまたはジ−C1−6アルキルホスホノ基、
Is shown.
R 3 represents -BD-EG [in the formula, B represents a C 6-14 arylene group, a C 3-8 cycloalkenylene group, a-heterocycle-or a-saturated heterocycle-, and D is. Indicates a single bond, -O (CH 2 ) n -,-(CH 2 ) n O- or -saturated heterocycle-, where n represents an integer of any one selected from 1-4, where E is. It exhibits a single bond, -O-, -N (R 10 )-, -CO- or -CONH-, even if R 10 is substituted with a hydrogen atom or any group selected from the substituent group α. It represents a good C 1-6 alkyl group, where G is selected from the C 1-6 alkyl group, substituent group α, which may be substituted with any group selected from the hydrogen atom, substituent group α. C 3-8 cycloalkyl group optionally substituted with any group, C 6-14 aryl group optionally substituted with any group selected from substituent group α, from substituent group α A heterocyclic group which may be substituted with any of the selected groups or a saturated heterocyclic group which may be substituted with any of the groups selected from the substituent group α, wherein the substituent group α is Heterocyclic group, saturated heterocyclic group, halogen atom, cyano group, C 1-6 alkylcarbonyl group, C 1-6 alkylcarbonyloxy group, C 1-6 alkylcarbonylamino group, C 1-6 alkoxycarbonyl group, C 1-6 alkoxy C 1-6 alkyl group, hydroxy C 1-6 alkyl group, hydroxy C 1-6 alkoxy group, C 1-6 alkylsulfonyl group, C 1-6 alkylsulfonylamino group, aminosulfonylamino group, carboxy Group, carboxy C 1-6 alkylcarbonyloxy group, carbamoyl group, hydroxy group, C 1-6 alkoxy group, C 1-6 alkyl group, C 3-8 cycloalkyl group, amino group, amino C 1-6 alkylcarbonyl Oxy group, mono or di-C 1-6 alkylamino group, 4-hydroxy tetrahydrofuran-3-yloxy group, mono or di-C 1-6 alkylphosphono group,

Figure 2020071550
Figure 2020071550

(nは、1〜4から選択されるいずれか1の整数を示す。)を示す。]を示す。
Bは、好ましくは、C6−14アリーレン基(例、フェニレン)、C3−8シクロアルケニレン基(例、シクロヘキセニレン)、−複素環−(例、ピリジンジイル)または−飽和複素環−(例、ピぺリジンジイル)であり、より好ましくは、
(N represents an integer of any one selected from 1 to 4). ] Is shown.
B is preferably a C 6-14 arylene group (eg, phenylene), a C 3-8 cycloalkenylene group (eg, cyclohexenylene), a-heterocycle- (eg, pyridinediyl) or a-saturated heterocycle- (eg, pyridinediyl). For example, piperidine diyl), more preferably

Figure 2020071550
Figure 2020071550

であり、さらにより好ましくは、

Figure 2020071550
And even more preferably
Figure 2020071550

である。
Dは、好ましくは、単結合、−O(CH−(nは、1、2、3または4である。)、−(CHO−(nは、1である。)または−飽和複素環−(例、アゼチジンジイル、ピロリジンジイル、ピぺリジンジイル、ピペラジンジイル、テトラヒドロピリジンジイル)であり、より好ましくは、単結合、−O(CH−(nは、2である。)または−飽和複素環−(例、アゼチジンジイル、ピロリジンジイル、ピぺリジンジイル、ピペラジンジイル、テトラヒドロピリジンジイル)である。
Eは、好ましくは、単結合、−O−、−N(R10)−(R10は、水素原子またはヒドロキシ基で置換されていてもよいC1−6アルキル基(例、メチル、エチル)である。)、−CO−または−CONH−であり、より好ましくは、単結合または−O−である。
Is.
D is preferably a single bond, −O (CH 2 ) n − (n is 1, 2, 3 or 4), − (CH 2 ) n O − (n is 1). Or a -saturated heterocycle- (eg, azetidinediyl, pyrrolidinediyl, piperidinediyl, piperazindiyl, tetrahydropyridinediyl), more preferably a single bond, -O (CH 2 ) n- (n is 2). ) Or-Saturated heterocycle- (eg, azetidinediyl, pyrrolidinediyl, piperidinediyl, piperazindiyl, tetrahydropyridinediyl).
E is preferably a single bond, −O−, −N (R 10 ) − (where R 10 is a C 1-6 alkyl group optionally substituted with a hydrogen atom or a hydroxy group (eg, methyl, ethyl). ), -CO- or -CONH-, more preferably a single bond or -O-.

Gは、好ましくは、(1)複素環基(例、オキサゾリル、イミダゾリル、テトラゾリル)、飽和複素環基(例、テトラヒドロフリル、ピペラジニル、モルホリニル、テトラヒドロピラニル、1,4−ジオキサニル)、ハロゲン原子(例、フッ素原子)、C1−6アルキルカルボニルオキシ基(例、メチルカルボニルオキシ)、C1−6アルキルカルボニルアミノ基(例、メチルカルボニルアミノ)、C1−6アルコキシカルボニル基(例、エトキシカルボニル)、ヒドロキシC1−6アルコキシ基(例、ヒドロキシエトキシ)、アミノスルホニルアミノ基、カルボキシ基、カルボキシC1−6アルキルカルボニルオキシ基(例、カルボキシエチルカルボニルオキシ)、カルバモイル基、ヒドロキシ基、C1−6アルコキシ基(例、メトキシ、エトキシ、イソプロポキシ、tert−ブトキシ)、C3−8シクロアルキル基(例、シクロプロピル)、アミノ基、アミノC1−6アルキルカルボニルオキシ基(例、アミノメチルカルボニルオキシ)、モノまたはジ−C1−6アルキルアミノ基(例、ジメチルアミノ)、4−ヒドロキシテトラヒドロフラン−3−イルオキシ基、モノまたはジ−C1−6アルキルホスホノ基(例、ジエチルホスホノ)、およびG is preferably (1) a heterocyclic group (eg, oxazolyl, imidazolyl, tetrazolyl), a saturated heterocyclic group (eg, tetrahydrofuryl, piperazinyl, morpholinyl, tetrahydropyranyl, 1,4-dioxanyl), a halogen atom (eg, tetrahydrofuryl, piperazinyl, morpholinyl), halogen atom (eg, tetrahydrofuryl, piperazinyl, morpholinyl), halogen atom (eg, tetrahydrofuryl, piperazinyl, morpholinyl). For example, fluorine atom), C 1-6 alkylcarbonyloxy group (eg, methylcarbonyloxy), C 1-6 alkylcarbonylamino group (eg, methylcarbonylamino), C 1-6 alkoxycarbonyl group (eg, ethoxycarbonyl) ), Hydroxy C 1-6 alkoxy group (eg, hydroxyethoxy), aminosulfonylamino group, carboxy group, carboxy C 1-6 alkylcarbonyloxy group (eg, carboxyethylcarbonyloxy), carbamoyl group, hydroxy group, C 1 -6 alkoxy groups (eg, methoxy, ethoxy, isopropoxy, tert-butoxy), C 3-8 cycloalkyl groups (eg, cyclopropyl), amino groups, amino C 1-6 alkylcarbonyloxy groups (eg, aminomethyl) Carbonyloxy), mono or di-C 1-6 alkylamino groups (eg, dimethylamino), 4-hydroxy tetrahydrofuran-3-yloxy groups, mono or di-C 1-6 alkylphosphono groups (eg, diethylphosphono). ),and

Figure 2020071550
Figure 2020071550

(nは、3を示す。)から選択されるいずれかの基で置換されていてもよいC1−6アルキル基(例、メチル、エチル、プロピル、イソプロピル、ブチル、イソブチル、tert−ブチル、イソペンチル)、(2)ヒドロキシ基で置換されていてもよいC3−8シクロアルキル基(例、シクロプロピル、シクロペンチル)、(3)カルボキシ基で置換されていてもよいC6−14アリール(例、フェニル)、(4)C1−6アルコキシC1−6アルキル基(例、メトキシメチル)およびヒドロキシC1−6アルキル基(例、ヒドロキシメチル)から選択されるいずれかの基で置換されていてもよい複素環基(例、イミダゾリル、オキサゾリル、ピリジル)または(5)飽和複素環基(例、モルホリニル、チオモルホリニル、ジオキソチオモルホリニル)、ハロゲン原子(例、フッ素原子)、シアノ基、C1−6アルキルカルボニル基(例、メチルカルボニル)、C1−6アルキルカルボニルアミノ基(例、メチルカルボニルアミノ)、ヒドロキシC1−6アルキル基(例、ヒドロキシメチル)、C1−6アルキルスルホニル基(例、メチルスルホニル)、C1−6アルキルスルホニルアミノ基(例、メチルスルホニルアミノ)、カルボキシ基、カルバモイル基、ヒドロキシ基、C1−6アルコキシ基(例、メトキシ、エトキシ)、C1−6アルキル基(例、メチル)、アミノ基、およびモノまたはジ−C1−6アルキルアミノ基(例、ジメチルアミノ、ジエチルアミノ)から選択されるいずれかの基で置換されていてもよい飽和複素環基(例、アゼチジニル、テトラヒドロフリル、ピペリジニル、ピペラジニル、ジヒドロオキサゾリル、テトラヒドロピラニル、モルホリニル、チオモルホリニル、オキソピロリジニル、オキソピペリジニル、オキソチオモルホリニル、ジオキソチオモルホリニル、1,4−ジオキサニル、スルホラニル)であり、より好ましくは、(1)ヒドロキシ基、アミノ基、カルボキシ基、アミノスルホニルアミノ基、ジエチルホスホノ基または (N indicates 3) C 1-6 alkyl groups optionally substituted with any group selected from (eg, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, isopentyl). ), (2) C 3-8 cycloalkyl groups optionally substituted with hydroxy groups (eg, cyclopropyl, cyclopentyl), (3) C 6-14 aryls optionally substituted with carboxy groups (eg, eg. (Phenyl), (4) C 1-6 alkoxy C 1-6 alkyl group (eg, methoxymethyl) and hydroxy C 1-6 alkyl group (eg, hydroxymethyl) substituted with any group selected from May be heterocyclic groups (eg, imidazolyl, oxazolyl, pyridyl) or (5) saturated heterocyclic groups (eg, morpholinyl, thiomorpholinyl, dioxothiomorpholinyl), halogen atoms (eg, fluorine atoms), cyano groups, C. 1-6 alkylcarbonyl group (eg, methylcarbonyl), C 1-6 alkylcarbonylamino group (eg, methylcarbonylamino), hydroxy C 1-6 alkyl group (eg, hydroxymethyl), C 1-6 alkylsulfonyl group (Eg, methylsulfonyl), C 1-6 alkylsulfonylamino group (eg, methylsulfonylamino), carboxy group, carbamoyl group, hydroxy group, C 1-6 alkoxy group (eg, methoxy, ethoxy), C 1-6 A saturated heterocyclic group optionally substituted with an alkyl group (eg, methyl), an amino group, and any group selected from a mono or di-C 1-6 alkylamino group (eg, dimethylamino, diethylamino). (For example, azetidinyl, tetrahydrofuryl, piperidinyl, piperazinyl, dihydrooxazolyl, tetrahydropyranyl, morpholinyl, thiomorpholinyl, oxopyrrolidinyl, oxopiperidinyl, oxothiomorpholinyl, dioxothiomorpholinyl, 1, 4-Dioxanyl, sulfolanyl), more preferably (1) hydroxy group, amino group, carboxy group, aminosulfonylamino group, diethylphosphono group or

Figure 2020071550
Figure 2020071550

で置換されたC1−6アルキル基(例、メチル、エチル、プロピル、ブチル、イソブチル、イソペンチル)、(2)フェニル基、(3)テトラヒドロピラニル基、(4)テトヒドロフラニル基、(5)ピペラジニル基または(6)モルホリニル基である。 C 1-6 alkyl groups substituted with (eg, methyl, ethyl, propyl, butyl, isobutyl, isopentyl), (2) phenyl group, (3) tetrahydropyranyl group, (4) tethydrofuranyl group, ( 5) piperazinyl group or (6) morpholinyl group.

は、C1−6アルキルカルボニル基、C1−6アルキルチオカルボニル基、カルバモイル基、C1−6アルコキシカルボニル基またはカルボキシ基を示す。
は、好ましくは、C1−6アルキルカルボニル基(例、アセチル)、C1−6アルキルチオカルボニル基(例、メチルチオカルボニル)、カルバモイル基、C1−6アルコキシカルボニル基(例、メトキシカルボニル、エトキシカルボニル)またはカルボキシ基である。
R 4 represents a C 1-6 alkylcarbonyl group, a C 1-6 alkylthiocarbonyl group, a carbamoyl group, a C 1-6 alkoxycarbonyl group or a carboxy group.
R 4 is preferably, C 1-6 alkylcarbonyl group (e.g., acetyl), C 1-6 alkyl thio group (e.g., methylthiocarbonyl), a carbamoyl group, C 1-6 alkoxycarbonyl group (e.g., methoxycarbonyl, It is an ethoxycarbonyl) or a carboxy group.

は、水素原子、C1−6アルキル基またはハロゲン原子を示す。
は、好ましくは、水素原子、C1−6アルキル基(例、メチル)またはハロゲン原子(例、塩素原子)である。
R 5 represents a hydrogen atom, a C 1-6 alkyl group or a halogen atom.
R 5 is preferably a hydrogen atom, a C 1-6 alkyl group (eg, methyl) or a halogen atom (eg, chlorine atom).

は、C1−6アルキル基、ハロゲン原子、アミノ基、ヒドロキシC1−6アルキル基、カルボキシC1−6アルキル基または水素原子(但し、Tが=CH−、Uが=N−の場合)を示す。
は、好ましくは、C1−6アルキル基(例、メチル)、ハロゲン原子(例、塩素原子)、アミノ基、ヒドロキシC1−6アルキル基(例、ヒドロキシエチル)、カルボキシC1−6アルキル基(例、カルボキシメチル)または水素原子(但し、Tが=CH−、Uが=N−の場合)である。
R 6 is a C 1-6 alkyl group, a halogen atom, an amino group, a hydroxy C 1-6 alkyl group, a carboxy C 1-6 alkyl group or a hydrogen atom (where T is = CH− and U is = N−. If) is shown.
R 6 is preferably a C 1-6 alkyl group (eg, methyl), a halogen atom (eg, a chlorine atom), an amino group, a hydroxy C 1-6 alkyl group (eg, hydroxyethyl), a carboxy C 1-6. It is an alkyl group (eg, carboxymethyl) or a hydrogen atom (where T is = CH− and U is = N−).

は、水素原子またはC1−6アルキル基を示す。
は、好ましくは、水素原子またはC1−6アルキル基(例、メチル、エチル)である。
R 7 represents a hydrogen atom or a C 1-6 alkyl group.
R 7 is preferably a hydrogen atom or a C 1-6 alkyl group (eg, methyl, ethyl).

は、水素原子、C1−6アルキル基またはC1−6アルキルカルボニル基を示す。
は、好ましくは、水素原子またはC1−6アルキルカルボニル基(例、メチルカルボニル、エチルカルボニル)である。
R 8 represents a hydrogen atom, a C 1-6 alkyl group or a C 1-6 alkyl carbonyl group.
R 8 is preferably a hydrogen atom or a C 1-6 alkylcarbonyl group (eg, methylcarbonyl, ethylcarbonyl).

は、水素原子またはC1−6アルキル基を示す。
は、好ましくは、水素原子またはC1−6アルキル基(例、メチル)である。
R 9 represents a hydrogen atom or a C 1-6 alkyl group.
R 9 is preferably a hydrogen atom or a C 1-6 alkyl group (eg, methyl).

本発明の好適な実施態様において、
(1)A環は、
In a preferred embodiment of the invention
(1) Ring A is

Figure 2020071550
Figure 2020071550

であり、Rは、アセチル基、カルバモイル基またはカルボキシ基であり、Rは、メチル基である;
(2)A環は、
R 4 is an acetyl group, a carbamoyl group or a carboxy group, and R 5 is a methyl group;
(2) Ring A is

Figure 2020071550
Figure 2020071550

であり、Rは、メチル基であり、Rは、水素原子である;
(3)A環は、
R 7 is a methyl group and R 9 is a hydrogen atom;
(3) Ring A is

Figure 2020071550
Figure 2020071550

であり、Rは、メチル基である;
(4)A環は、
In and, R 7 is a methyl group;
(4) Ring A is

Figure 2020071550
Figure 2020071550

である;
(5)A環は、
Is;
(5) Ring A is

Figure 2020071550
Figure 2020071550

である;または
(6)A環は、
Or (6) Ring A is

Figure 2020071550
Figure 2020071550

であり、Rは、塩素、メチル基またはカルボキシメチル基である。And a, R 6 is chlorine, methyl or carboxymethyl groups.

化合物(I)の好適な例としては、以下の化合物が挙げられる。
[化合物I−1]
が、ハロゲン原子(例、塩素原子)、シアノ基、C1−6アルコキシ基(例、メトキシ、イソプロポキシ)、C1−6アルキルスルファニル基(例、メチルスルファニル)、C1−6アルキルカルボニル基(例、アセチル)、カルバモイル基または複素環基(例、イミダゾリル、フリル、ピリジル)であり;
T及びUが、Tが=CR−であり、Uが=N−であるか、あるいは、T及びUの両方が、=CR−であり;
が、水素原子、ハロゲン原子(例、フッ素原子)、C1−6アルキル基(例、メチル)またはC1−6アルコキシ基(例、メトキシ)であり;
A環が、
Preferable examples of compound (I) include the following compounds.
[Compound I-1]
R 1 is a halogen atom (eg, chlorine atom), cyano group, C 1-6 alkoxy group (eg, methoxy, isopropoxy), C 1-6 alkyl sulfanyl group (eg, methyl sulfanyl), C 1-6 alkyl. A carbonyl group (eg, acetyl), a carbamoyl group or a heterocyclic group (eg, imidazolyl, frill, pyridyl);
T and U have T = CR 2 − and U = N −, or both T and U have = CR 2 −;
R 2 is a hydrogen atom, a halogen atom (e.g., fluorine atom), be C 1-6 alkyl group (e.g., methyl) or C 1-6 alkoxy group (e.g., methoxy);
Ring A is

Figure 2020071550
Figure 2020071550

であり;
が、−B−D−E−Gであり;
Bが、C6−14アリーレン基(例、フェニレン)、C3−8シクロアルケニレン基(例、シクロヘキセニレン)、−複素環−(例、ピリジンジイル)または−飽和複素環−(例、ピぺリジンジイル)であり;
Dが、単結合、−O(CH−(nは、1、2、3または4である。)、−(CHO−(nは、1である。)または−飽和複素環−(例、アゼチジンジイル、ピロリジンジイル、ピぺリジンジイル、ピペラジンジイル、テトラヒドロピリジンジイル)であり;
Eが、単結合、−O−、−N(R10)−(R10は、水素原子またはヒドロキシ基で置換されていてもよいC1−6アルキル基(例、メチル、エチル)である。)、−CO−または−CONH−であり;
Gが、(1)複素環基(例、オキサゾリル、イミダゾリル、テトラゾリル)、飽和複素環基(例、テトラヒドロフリル、ピペラジニル、モルホリニル、テトラヒドロピラニル、1,4−ジオキサニル)、ハロゲン原子(例、フッ素原子)、C1−6アルキルカルボニルオキシ基(例、メチルカルボニルオキシ)、C1−6アルキルカルボニルアミノ基(例、メチルカルボニルアミノ)、C1−6アルコキシカルボニル基(例、エトキシカルボニル)、ヒドロキシC1−6アルコキシ基(例、ヒドロキシエトキシ)、アミノスルホニルアミノ基、カルボキシ基、カルボキシC1−6アルキルカルボニルオキシ基(例、カルボキシエチルカルボニルオキシ)、カルバモイル基、ヒドロキシ基、C1−6アルコキシ基(例、メトキシ、エトキシ、イソプロポキシ、tert−ブトキシ)、C3−8シクロアルキル基(例、シクロプロピル)、アミノ基、アミノC1−6アルキルカルボニルオキシ基(例、アミノメチルカルボニルオキシ)、モノまたはジ−C1−6アルキルアミノ基(例、ジメチルアミノ)、4−ヒドロキシテトラヒドロフラン−3−イルオキシ基、モノまたはジ−C1−6アルキルホスホノ基(例、ジエチルホスホノ)、および
Is;
R 3 is -BD-EG;
B is a C 6-14 arylene group (eg, phenylene), a C 3-8 cycloalkenylene group (eg, cyclohexenylene), a-heterocycle- (eg, pyridinediyl) or a-saturated heterocycle- (eg, pi). Perizinzyl);
D is single bond, -O (CH 2 ) n- (n is 1, 2, 3 or 4),-(CH 2 ) n O- (n is 1) or -saturated Heterocycle- (eg, azetidinediyl, pyrrolidinediyl, piperidinediyl, piperazindiyl, tetrahydropyridinediyl);
E is a single bond, -O-, -N (R 10 )-(R 10 is a C 1-6 alkyl group (eg, methyl, ethyl) that may be substituted with a hydrogen atom or a hydroxy group. ), -CO- or -CONH-;
G is (1) heterocyclic group (eg, oxazolyl, imidazolyl, tetrazolyl), saturated heterocyclic group (eg, tetrahydrofuryl, piperazinyl, morpholinyl, tetrahydropyranyl, 1,4-dioxanyl), halogen atom (eg, fluorine). Atomic), C 1-6 alkylcarbonyloxy group (eg, methylcarbonyloxy), C 1-6 alkylcarbonylamino group (eg, methylcarbonylamino), C 1-6 alkoxycarbonyl group (eg, ethoxycarbonyl), hydroxy C 1-6 alkoxy group (eg, hydroxyethoxy), aminosulfonylamino group, carboxy group, carboxy C 1-6 alkylcarbonyloxy group (eg, carboxyethylcarbonyloxy), carbamoyl group, hydroxy group, C 1-6 alkoxy Groups (eg, methoxy, ethoxy, isopropoxy, tert-butoxy), C 3-8 cycloalkyl groups (eg, cyclopropyl), amino groups, amino C 1-6 alkylcarbonyloxy groups (eg, aminomethylcarbonyloxy) , Mono or di-C 1-6 alkylamino groups (eg, dimethylamino), 4-hydroxy tetrahydrofuran-3-yloxy groups, mono or di-C 1-6 alkylphosphono groups (eg, diethylphosphono), and

Figure 2020071550
Figure 2020071550

(nは、3を示す。)から選択されるいずれかの基で置換されていてもよいC1−6アルキル基(例、メチル、エチル、プロピル、イソプロピル、ブチル、イソブチル、tert−ブチル、イソペンチル)、(2)ヒドロキシ基で置換されていてもよいC3−8シクロアルキル基(例、シクロプロピル、シクロペンチル)、(3)カルボキシ基で置換されていてもよいC6−14アリール(例、フェニル)、(4)C1−6アルコキシC1−6アルキル基(例、メトキシメチル)およびヒドロキシC1−6アルキル基(例、ヒドロキシメチル)から選択されるいずれかの基で置換されていてもよい複素環基(例、イミダゾリル、オキサゾリル、ピリジル)または(5)飽和複素環基(例、モルホリニル、チオモルホリニル、ジオキソチオモルホリニル)、ハロゲン原子(例、フッ素原子)、シアノ基、C1−6アルキルカルボニル基(例、メチルカルボニル)、C1−6アルキルカルボニルアミノ基(例、メチルカルボニルアミノ)、ヒドロキシC1−6アルキル基(例、ヒドロキシメチル)、C1−6アルキルスルホニル基(例、メチルスルホニル)、C1−6アルキルスルホニルアミノ基(例、メチルスルホニルアミノ)、カルボキシ基、カルバモイル基、ヒドロキシ基、C1−6アルコキシ基(例、メトキシ、エトキシ)、C1−6アルキル基(例、メチル)、アミノ基、およびモノまたはジ−C1−6アルキルアミノ基(例、ジメチルアミノ、ジエチルアミノ)から選択されるいずれかの基で置換されていてもよい飽和複素環基(例、アゼチジニル、テトラヒドロフリル、ピペリジニル、ピペラジニル、ジヒドロオキサゾリル、テトラヒドロピラニル、モルホリニル、チオモルホリニル、オキソピロリジニル、オキソピペリジニル、オキソチオモルホリニル、ジオキソチオモルホリニル、1,4−ジオキサニル、スルホラニル)であり;
が、C1−6アルキルカルボニル基(例、アセチル)、C1−6アルキルチオカルボニル基(例、メチルチオカルボニル)、カルバモイル基、C1−6アルコキシカルボニル基(例、メトキシカルボニル、エトキシカルボニル)またはカルボキシ基であり;
が、水素原子、C1−6アルキル基(例、メチル)またはハロゲン原子(例、塩素原子)であり;
が、C1−6アルキル基(例、メチル)、ハロゲン原子(例、塩素原子)、アミノ基、ヒドロキシC1−6アルキル基(例、ヒドロキシエチル)またはカルボキシC1−6アルキル基(例、カルボキシメチル)であり;
が、水素原子またはC1−6アルキル基(例、メチル、エチル)であり;
が、水素原子またはC1−6アルキルカルボニル基(例、メチルカルボニル、エチルカルボニル)であり;
が、水素原子またはC1−6アルキル基(例、メチル)である;
化合物(I)。
A C 1-6 alkyl group (eg, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, isopentyl) which may be substituted with any group selected from (n indicates 3). ), (2) C 3-8 cycloalkyl groups optionally substituted with hydroxy groups (eg, cyclopropyl, cyclopentyl), (3) C 6-14 aryls optionally substituted with carboxy groups (eg, eg. Substituted with any group selected from (phenyl), (4) C 1-6 alkoxy C 1-6 alkyl groups (eg, methoxymethyl) and hydroxy C 1-6 alkyl groups (eg, hydroxymethyl). May be heterocyclic groups (eg, imidazolyl, oxazolyl, pyridyl) or (5) saturated heterocyclic groups (eg, morpholinyl, thiomorpholinyl, dioxothiomorpholinyl), halogen atoms (eg, fluorine atoms), cyano groups, C. 1-6 alkylcarbonyl group (eg, methylcarbonyl), C 1-6 alkylcarbonylamino group (eg, methylcarbonylamino), hydroxy C 1-6 alkyl group (eg, hydroxymethyl), C 1-6 alkylsulfonyl group (Eg, methylsulfonyl), C 1-6 alkylsulfonylamino group (eg, methylsulfonylamino), carboxy group, carbamoyl group, hydroxy group, C 1-6 alkoxy group (eg, methoxy, ethoxy), C 1-6 Saturated heterocyclic groups optionally substituted with any group selected from alkyl groups (eg, methyl), amino groups, and mono or di-C 1-6 alkylamino groups (eg, dimethylamino, diethylamino). (For example, azetidinyl, tetrahydrofuryl, piperidinyl, piperazinyl, dihydrooxazolyl, tetrahydropyranyl, morpholinyl, thiomorpholinyl, oxopyrrolidinyl, oxopiperidinyl, oxothiomorpholinyl, dioxothiomorpholinyl, 1, 4-Dioxanyl, sulfolanyl);
R 4 is a C 1-6 alkylcarbonyl group (eg, acetyl), C 1-6 alkylthiocarbonyl group (eg, methylthiocarbonyl), carbamoyl group, C 1-6 alkoxycarbonyl group (eg, methoxycarbonyl, ethoxycarbonyl). Or a carboxy group;
R 5 is a hydrogen atom, C 1-6 alkyl group (eg, methyl) or halogen atom (eg, chlorine atom);
R 6 is a C 1-6 alkyl group (eg, methyl), a halogen atom (eg, chlorine atom), an amino group, a hydroxy C 1-6 alkyl group (eg, hydroxyethyl) or a carboxy C 1-6 alkyl group (eg, hydroxyethyl). For example, carboxymethyl);
R 7 is a hydrogen atom or C 1-6 alkyl group (eg, methyl, ethyl);
R 8 is a hydrogen atom or a C 1-6 alkylcarbonyl group (eg, methylcarbonyl, ethylcarbonyl);
R 9 is a hydrogen atom or a C 1-6 alkyl group (eg, methyl);
Compound (I).

[化合物I−2]
が、C1−6アルコキシ基(例、メトキシ)またはカルバモイル基であり;
T及びUが、Tが=CR−であり、Uが=N−であるか、あるいは、T及びUの両方が、=CR−であり;
が、水素原子、ハロゲン原子(例、フッ素原子)またはC1−6アルコキシ基(例、メトキシ)であり;
A環が、
[Compound I-2]
R 1 is a C 1-6 alkoxy group (eg, methoxy) or a carbamoyl group;
T and U have T = CR 2 − and U = N −, or both T and U have = CR 2 −;
R 2 is hydrogen atom, a halogen atom (e.g., fluorine atom) or a C 1-6 alkoxy group (e.g., methoxy);
Ring A is

Figure 2020071550
Figure 2020071550

であり;
が、−B−D−E−Gであり;
Bが、C6−14アリーレン基(例、フェニレン)または−飽和複素環−(例、ピぺリジンジイル)であり;
Dが、単結合、−O(CH−(nは、1、2、3または4である。)または−飽和複素環−(例、アゼチジンジイル、ピぺリジンジイル)であり;
Eが、単結合または−O−であり;
Gが、(1)飽和複素環基(例、モルホリニル)、アミノスルホニルアミノ基、カルボキシ基、ヒドロキシ基、アミノ基、モノまたはジ−C1−6アルキルホスホノ基(例、ジエチルホスホノ)、および
Is;
R 3 is -BD-EG;
B is a C 6-14 arylene group (eg, phenylene) or-saturated heterocycle- (eg, piperidinediyl);
D is a single bond, -O (CH 2 ) n- (n is 1, 2, 3 or 4) or -saturated heterocycle- (eg, azetidinediyl, piperidinediyl);
E is a single bond or -O-;
G is (1) a saturated heterocyclic group (eg, morpholinyl), an aminosulfonylamino group, a carboxy group, a hydroxy group, an amino group, a mono or di-C 1-6 alkylphosphono group (eg, diethylphosphono), and

Figure 2020071550
Figure 2020071550

(nは、3を示す。)から選択されるいずれかの基で置換されていてもよいC1−6アルキル基(例、メチル、エチル、プロピル、ブチル)、(2)C6−14アリール(例、フェニル)または(3)C1−6アルキルカルボニル基(例、メチルカルボニル)で置換されていてもよい飽和複素環基(例、ピペラジニル、テトラヒドロピラニル、モルホリニル)であり;
が、C1−6アルキルカルボニル基(例、アセチル)またはカルバモイル基であり;
が、水素原子またはC1−6アルキル基(例、メチル)であり;
が、C1−6アルキル基(例、メチル)であり;
が、C1−6アルキル基(例、メチル)であり;
が、水素原子である;
化合物(I)。
(N indicates 3) C 1-6 alkyl groups (eg, methyl, ethyl, propyl, butyl), which may be substituted with any group selected from (2) C 6-14 aryl. It is a saturated heterocyclic group (eg, piperazinyl, tetrahydropyranyl, morpholinyl) which may be substituted with (eg, phenyl) or (3) C 1-6 alkylcarbonyl group (eg, methylcarbonyl);
R 4 is located at C 1-6 alkylcarbonyl group (e.g., acetyl) or a carbamoyl group;
R 5 is a hydrogen atom or a C 1-6 alkyl group (eg, methyl);
R 6 is a C 1-6 alkyl group (eg, methyl);
R 7 is a C 1-6 alkyl group (eg, methyl);
R 9 is a hydrogen atom;
Compound (I).

[化合物I−3]
が、C1−6アルコキシ基(例、メトキシ)、カルバモイル基または複素環基(例、イミダゾリル)であり;
T及びUが、Tが=CR−であり、Uが=N−であるか、あるいは、T及びUの両方が、=CR−であり;
が、水素原子、ハロゲン原子(例、フッ素原子)またはC1−6アルコキシ基(例、メトキシ)であり;
A環が、
[Compound I-3]
R 1 is a C 1-6 alkoxy group (eg, methoxy), a carbamoyl group or a heterocyclic group (eg, imidazolyl);
T and U have T = CR 2 − and U = N −, or both T and U have = CR 2 −;
R 2 is hydrogen atom, a halogen atom (e.g., fluorine atom) or a C 1-6 alkoxy group (e.g., methoxy);
Ring A is

Figure 2020071550
Figure 2020071550

であり;
が、−B−D−E−Gであり;
Bが、C6−14アリーレン基(例、フェニレン)または−飽和複素環−(例、ピぺリジンジイル)であり;
Dが、単結合、−O(CH−(nは、1、2、3または4である。)、−(CHO−(nは、1である。)または−飽和複素環−(例、アゼチジンジイル、ピロリジンジイル、ピぺリジンジイル、ピペラジンジイル)であり;
Eが、単結合、−O−または−CO−であり;
Gが、(1)飽和複素環基(例、モルホリニル)、アミノスルホニルアミノ基、カルボキシ基、ヒドロキシ基、アミノ基、モノまたはジ−C1−6アルキルホスホノ基(例、ジエチルホスホノ)、および
Is;
R 3 is -BD-EG;
B is a C 6-14 arylene group (eg, phenylene) or-saturated heterocycle- (eg, piperidinediyl);
D is single bond, -O (CH 2 ) n- (n is 1, 2, 3 or 4),-(CH 2 ) n O- (n is 1) or -saturated Heterocycle- (eg, azetidinediyl, pyrrolidinediyl, piperidinediyl, piperazindiyl);
E is a single bond, -O- or -CO-;
G is (1) a saturated heterocyclic group (eg, morpholinyl), an aminosulfonylamino group, a carboxy group, a hydroxy group, an amino group, a mono or di-C 1-6 alkylphosphono group (eg, diethylphosphono), and

Figure 2020071550
Figure 2020071550

(nは、3を示す。)から選択されるいずれかの基で置換されていてもよいC1−6アルキル基(例、メチル、エチル、プロピル、ブチル)、(2)C6−14アリール(例、フェニル)または(3)C1−6アルキルカルボニル基(例、メチルカルボニル)、カルボキシ基、C1−6アルキル基(例、メチル)およびアミノ基から選択されるいずれかの基で置換されていてもよい飽和複素環基(例、ピペラジニル、ピペリジニル、テトラヒドロピラニル、モルホリニル)であり;
が、C1−6アルキルカルボニル基(例、アセチル)、C1−6アルキルチオカルボニル基(例、メチルチオカルボニル)またはカルバモイル基であり;
が、水素原子またはC1−6アルキル基(例、メチル)であり;
が、水素原子またはC1−6アルキル基(例、メチル)であり;
が、C1−6アルキル基(例、メチル)であり;
が、水素原子である;
化合物(I)。
(N indicates 3) C 1-6 alkyl groups (eg, methyl, ethyl, propyl, butyl), which may be substituted with any group selected from (2) C 6-14 aryl. Substituted with any group selected from (eg, phenyl) or (3) C 1-6 alkylcarbonyl group (eg, methylcarbonyl), carboxy group, C 1-6 alkyl group (eg, methyl) and amino group. Saturated heterocyclic groups that may be (eg, piperazinyl, piperidinyl, tetrahydropyranyl, morpholinyl);
R 4 is, C 1-6 alkylcarbonyl group (e.g., acetyl), C 1-6 alkyl thio group (e.g., methylthiocarbonyl) be or carbamoyl group;
R 5 is a hydrogen atom or a C 1-6 alkyl group (eg, methyl);
R 6 is a hydrogen atom or a C 1-6 alkyl group (eg, methyl);
R 7 is a C 1-6 alkyl group (eg, methyl);
R 9 is a hydrogen atom;
Compound (I).

化合物(I)の好ましい具体例としては、例えば、実施例1〜21及び下記表1−1〜1−71に記載の実施例22〜375の化合物(以下、化合物1〜375ともいう。)が挙げられ、中でも、
(1)3-アセチル-5,6-ジメトキシ-2-メチル-1-[4-(モルホリン-4-イル)フェニル]インドール
Preferred specific examples of the compound (I) include, for example, the compounds of Examples 1 to 21 and Examples 22 to 375 shown in Table 1-1 to 1-71 below (hereinafter, also referred to as compounds 1 to 375). Among them,
(1) 3-Acetyl-5,6-dimethoxy-2-methyl-1- [4- (morpholine-4-yl) phenyl] indole

Figure 2020071550
Figure 2020071550

(化合物1)、
(2)3-アセチル-5,6-ジメトキシ-2-メチル-1-{4-[3-(2-オキソ-2λ5-[1,3,2]ジオキサホスフィナン-2-イル)プロポキシ]フェニル}インドール
(Compound 1),
(2) 3-Acetyl-5,6-dimethoxy-2-methyl-1-{4- [3- (2-oxo-2λ 5- [1,3,2] dioxaphosphinan-2-yl) propoxy ] Phenyl} indole

Figure 2020071550
Figure 2020071550

(化合物38)、
(3)3-アセチル-1-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-6-メトキシ-2-メチルインドール
(Compound 38),
(3) 3-Acetyl-1-{4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -6-methoxy-2-methylindole

Figure 2020071550
Figure 2020071550

(化合物63)、
(4)2-{1-[4-(6-メトキシ-2-メチルイミダゾ[4,5-c]ピリジン-1-イル)フェニル]アゼチジン-3-イルオキシ}エタノール
(Compound 63),
(4) 2- {1- [4- (6-Methoxy-2-methylimidazole [4,5-c] pyridin-1-yl) phenyl] azetidine-3-yloxy} ethanol

Figure 2020071550
Figure 2020071550

(化合物19)、
(5)4-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-6-メトキシ-1-メチル-1H-[1,7]ナフチリジン-2-オン
(Compound 19),
(5) 4- {4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -6-methoxy-1-methyl-1H- [1,7] naphthididine-2-one

Figure 2020071550
Figure 2020071550

(化合物10)、
(6)7-メトキシ-1-[4-(3-(モルホリン-4-イル)アゼチジン-1-イル)フェニル]-1H-キノリン-4-オン 二塩酸塩
(Compound 10),
(6) 7-Methoxy-1- [4- (3- (morpholine-4-yl) azetidine-1-yl) phenyl] -1H-quinoline-4-one dihydrochloride

Figure 2020071550
Figure 2020071550

(化合物20)、
(7)2-オキソ-4-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}-2H-クロメン-6-カルボキサミド
(Compound 20),
(7) 2-oxo-4- {4- [2- (tetrahydropyran-4-yloxy) ethoxy] phenyl} -2H-chromen-6-carboxamide

Figure 2020071550
Figure 2020071550

(化合物163)、
(8)1-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-7-メトキシ-4-メチル-1,4-ジヒドロ-2H-ピリド[3,4-b]ピラジン-3-オン
(Compound 163),
(8) 1-{4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -7-methoxy-4-methyl-1,4-dihydro-2H-pyrazine [3,4-b] Pyrazine-3-on

Figure 2020071550
Figure 2020071550

(化合物120)、
(9)1-{4-[3-(2-スルファモイルアミノエトキシ)アゼチジン-1-イル]フェニル}-7-メトキシ-4-メチル-1,4-ジヒドロ-2H-ピリド[3,4-b]ピラジン-3-オン
(Compound 120),
(9) 1-{4- [3- (2-Sulfamoylaminoethoxy) azetidine-1-yl] phenyl} -7-methoxy-4-methyl-1,4-dihydro-2H-pyrido [3,4 -b] Pyrazine-3-on

Figure 2020071550
Figure 2020071550

(化合物149)、
(10)8-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ナフタレン-2-カルボキサミド
(Compound 149),
(10) 8- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] phenyl} naphthalene-2-carboxamide

Figure 2020071550
Figure 2020071550

(化合物21)、
(11)3-アセチル-1-{4-[4-(3-アミノプロピル)ピペリジン-1-イル]フェニル}-5,6-ジメトキシ-2-メチルインドール
(Compound 21),
(11) 3-Acetyl-1-{4- [4- (3-aminopropyl) piperidine-1-yl] phenyl} -5,6-dimethoxy-2-methylindole

Figure 2020071550
Figure 2020071550

(化合物31)、
(12)3-アセチル-6-メトキシ-2-メチル-1-[4-(4-(モルホリン-4-イル)ブトキシ)フェニル]インドール 塩酸塩
(Compound 31),
(12) 3-Acetyl-6-Methoxy-2-methyl-1- [4- (4- (morpholine-4-yl) butoxy) phenyl] indole hydrochloride

Figure 2020071550
Figure 2020071550

(化合物6)、
(13)3-アセチル-5-フルオロ-6-メトキシ-2-メチル-1-(1-フェニルピペリジン-4-イル)インドール
(Compound 6),
(13) 3-Acetyl-5-Fluoro-6-Methoxy-2-methyl-1- (1-phenylpiperidine-4-yl) indole

Figure 2020071550
Figure 2020071550

(化合物78)、
(14)3-{1-[4-(3-アセチル-5,6-ジメトキシ-2-メチルインドール-1-イル)フェニル]ピペリジン-4-イル}プロピオン酸
(Compound 78),
(14) 3- {1- [4- (3-Acetyl-5,6-dimethoxy-2-methylindole-1-yl) phenyl] piperidin-4-yl} propionic acid

Figure 2020071550
Figure 2020071550

(化合物28)、
(15)1-[4-(4-アセチルピペラジン-1-イル)フェニル]-5,6-ジメトキシ-2-メチルインドール-3-カルボキサミド
(Compound 28),
(15) 1- [4- (4-Acetylpiperazin-1-yl) phenyl] -5,6-dimethoxy-2-methylindole-3-carboxamide

Figure 2020071550
Figure 2020071550

(化合物48)、
(16)1-{4-[4-(2-ヒドロキシエトキシ)ピペリジン-1-イル]フェニル}-6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン-3-カルボキサミド
(Compound 48),
(16) 1- {4- [4- (2-Hydroxyethoxy) piperidine-1-yl] phenyl} -6-methoxy-2-methyl-1H-pyrrolo [3,2-c] pyridine-3-carboxamide

Figure 2020071550
Figure 2020071550

(化合物8)、
(17)ジエチル(1-(4-(5-フルオロ-6-メトキシ-2-メチル-1H-ベンゾ[d]イミダゾール-1-イル)フェニル)アゼチジン-3-イルオキシ)メチルホスホネート
(Compound 8),
(17) Diethyl (1- (4- (5-fluoro-6-methoxy-2-methyl-1H-benzo [d] imidazol-1-yl) phenyl) azetidine-3-yloxy) methylphosphonate

Figure 2020071550
Figure 2020071550

(化合物112)、および
(18)3-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ベンゾ[b]チオフェン-5-カルボキサミド
(Compound 112), and (18) 3- {4- [2- (tetrahydropyran-4-yloxy) ethoxy] phenyl} benzo [b] thiophene-5-carboxamide

Figure 2020071550
Figure 2020071550

(化合物14)が好ましい。 (Compound 14) is preferred.

(本発明の化合物(I)の製造方法)
本発明の化合物(I)は、その基本骨格あるいは置換基の種類に基づく特徴を利用し、各種の公知の製造方法を適用して製造することができる。公知の方法としては、例えば、「ORGANIC FUNCTIONAL GROUP PREPARATIONS」、第2版、ACADEMIC PRESS,INC.、1989年、「Comprehensive Organic Transformations」、VCHPublishers Inc.、1989年等に記載された方法がある。
その際、官能基の種類によっては、当該官能基を原料ないし中間体の段階で適当な保護基で保護、または当該官能基に容易に転化可能な基に置き換えておくことが製造技術上効果的な場合がある。
このような官能基としては、例えば、アミノ基、水酸基、カルボキシル基等があり、それらの保護基としては、例えば、T.W.Greene及びP.G. Wuts著、「Protective Groups in Organic Synthesis(第3版、1999年)」に記載の保護基があり、これらの反応条件に応じて適宜選択して用いればよい。このような方法によれば、当該置換基を導入して反応を行った後、必要に応じて保護基を除去、あるいは所望の基に転化することにより、所望の化合物を得ることができる。
また、本発明の化合物のプロドラッグは、上記保護基と同様に、原料ないし中間体の段階で特定の基を導入し、あるいは得られた本発明の化合物を用いて、反応を行うことで製造できる。反応は、通常のエステル化、アミド化、脱水、水素添加等、当業者により公知の方法を適用することにより行うことができる。
以下に本発明の化合物の製造方法について述べる。ただし、製造方法は、下記の方法に何ら限定されるものではない。
なお、各反応における原料化合物は、具体的製法を述べない場合、市販されているものを容易に入手して用いることができるか、または自体公知の方法、またはそれに準ずる方法に従って製造することもできる。
(Method for producing compound (I) of the present invention)
The compound (I) of the present invention can be produced by applying various known production methods by utilizing its characteristics based on the basic skeleton or the type of substituent. Known methods include, for example, the methods described in "ORGANIC FUNCTIONAL GROUP PREPARATIONS", 2nd Edition, ACADEMIC PRESS, INC., 1989, "Comprehensive Organic Transformations", VCH Publishers Inc., 1989 and the like.
At that time, depending on the type of the functional group, it is effective in terms of manufacturing technology to protect the functional group with an appropriate protecting group at the stage of the raw material or the intermediate, or to replace it with a group that can be easily converted to the functional group. In some cases.
Such functional groups include, for example, amino groups, hydroxyl groups, carboxyl groups and the like, and their protecting groups include, for example, TW Greene and PG Wuts, "Protective Groups in Organic Synthesis (3rd Edition, 1999)". ”, And may be appropriately selected and used according to these reaction conditions. According to such a method, a desired compound can be obtained by introducing the substituent, carrying out the reaction, and then removing the protecting group or converting to a desired group, if necessary.
Further, the prodrug of the compound of the present invention is produced by introducing a specific group at the stage of a raw material or an intermediate, or by carrying out a reaction using the obtained compound of the present invention, similarly to the above-mentioned protecting group. can. The reaction can be carried out by applying a method known to those skilled in the art, such as ordinary esterification, amidation, dehydration, hydrogenation and the like.
The method for producing the compound of the present invention will be described below. However, the manufacturing method is not limited to the following methods.
If the specific production method is not described, the raw material compound in each reaction can be easily obtained and used on the market, or can be produced according to a method known per se or a method similar thereto. ..

(製法A)
製法Aは、化合物(a-1)と化合物(a-2)とをカップリングさせることによって本発明化合物(I)のA環が
(Manufacturing method A)
In the production method A, the A ring of the compound (I) of the present invention is formed by coupling the compound (a-1) and the compound (a-2).

Figure 2020071550
Figure 2020071550

である化合物の中間体化合物(a-3)を製造する方法である。
本製法に用いられる化合物(a-1)は、自体公知の方法にて製造することができる。
This is a method for producing an intermediate compound (a-3) of the compound.
The compound (a-1) used in this production method can be produced by a method known per se.

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R3、R5、T及びUは、上記と同意義を示し、Xは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基などの脱離基を示す。In the above scheme, R 1 , R 3 , R 5 , T and U have the same meaning as above, and X indicates a leaving group such as a halogen atom or a trifluoromethanesulfonyloxy group.

StepAは、無機炭酸塩もしくはリン酸塩(例えば、炭酸カリウム、炭酸セシウム、リン酸三カリウム)などの塩基、銅化合物(例えば、CuBr、CuI)及びリガンド(例えば、N,N’-ジメチルエチレンジアミン、1,2-ジアミノシクロヘキサン、trans-N,N’-ジメチルシクロヘキサン-1,2-ジアミンなど)を用いた求核置換反応であり、不活性溶媒中、化合物(a-1)と化合物(a-2)から本発明化合物(I)の中間体化合物(a-3)を製造する工程である。 StepA is a base such as an inorganic carbonate or phosphate (eg, potassium carbonate, cesium carbonate, tripotassium phosphate), a copper compound (eg, CuBr, CuI) and a ligand (eg, N, N'-dimethylethylenediamine, This is a nucleophilic substitution reaction using 1,2-diaminocyclohexane, trans-N, N'-dimethylcyclohexane-1,2-diamine, etc.), in which compound (a-1) and compound (a-) are used in an inert solvent. This is a step of producing an intermediate compound (a-3) of the compound (I) of the present invention from 2).

StepAにおける化合物(a-2)の使用量は、化合物(a-1)に対して、通常1〜3当量、好ましくは1〜1.5当量である。無機炭酸塩もしくはリン酸塩の使用量は、化合物(a-1)に対して、通常1〜5当量、好ましくは1〜3当量である。銅化合物の使用量は、化合物(a-1)に対して、通常0.01〜0.3当量、好ましくは0.05〜0.1当量である。リガンドの使用量は、化合物(a-1)に対して、通常0.01〜0.3当量、好ましくは0.01〜0.15当量である。 The amount of compound (a-2) used in Step A is usually 1 to 3 equivalents, preferably 1 to 1.5 equivalents, relative to compound (a-1). The amount of the inorganic carbonate or phosphate used is usually 1 to 5 equivalents, preferably 1 to 3 equivalents, relative to compound (a-1). The amount of the copper compound used is generally 0.01 to 0.3 equivalents, preferably 0.05 to 0.1 equivalents, relative to compound (a-1). The amount of the ligand used is generally 0.01 to 0.3 equivalents, preferably 0.01 to 0.15 equivalents, relative to compound (a-1).

使用される不活性溶媒は、例えばメタノール、エタノール、プロパノール、2−プロパノールまたはブタノールのようなアルコール類;ベンゼン、トルエン、またはキシレンのような芳香族炭化水素類;ジエチルエーテル、ジイソプロピルエーテル、テトラヒドロフラン、ジオキサンまたは1,2−ジメトキシエタンのようなエーテル類;N,N−ジメチルホルムアミド、N,N−ジメチルアセトアミド、N−メチル−2−ピロリドン、N−メチルピロリジノンまたはヘキサメチルホスホロトリアミドのようなアミド類;或いはメチレンクロリド、クロロホルム、四塩化炭素、ジクロロエタン、クロロベンゼン、o−ジクロロベンゼン、m−ジクロロベンゼン、フルオロベンゼン、トリクロロメチルベンゼンまたはトリフルオロメチルベンゼンのようなハロゲン化炭化水素類、あるいはこれらの混合物である。 The inert solvent used is, for example, alcohols such as methanol, ethanol, propanol, 2-propanol or butanol; aromatic hydrocarbons such as benzene, toluene, or xylene; diethyl ether, diisopropyl ether, tetrahydrofuran, dioxane. Or ethers such as 1,2-dimethoxyethane; amides such as N, N-dimethylformamide, N, N-dimethylacetamide, N-methyl-2-pyrrolidone, N-methylpyrrolidinone or hexamethylphosphorotriamide. Or with halogenated hydrocarbons such as methylene chloride, chloroform, carbon tetrachloride, dichloroethane, chlorobenzene, o-dichlorobenzene, m-dichlorobenzene, fluorobenzene, trichloromethylbenzene or trifluoromethylbenzene, or mixtures thereof. be.

反応温度は原料化合物または使用される溶媒によって異なるが、通常、0℃から反応混合物の還流温度であり、好適には、室温から反応混合物の還流温度である。
反応時間は原料化合物、使用される溶媒または反応温度によって異なるが、通常30分間から72時間であり、好適には30分間から24時間である。
The reaction temperature varies depending on the starting compound or the solvent used, but is usually from 0 ° C. to the reflux temperature of the reaction mixture, preferably from room temperature to the reflux temperature of the reaction mixture.
The reaction time varies depending on the starting compound, the solvent used or the reaction temperature, but is usually 30 minutes to 72 hours, preferably 30 minutes to 24 hours.

(製法B)
製法Bは、本発明化合物(I)のA環が
(Manufacturing method B)
In the production method B, the A ring of the compound (I) of the present invention is used.

Figure 2020071550
Figure 2020071550

であり、Rが、C1−6低級アルキルカルボニル基である化合物(b-2)の製法であって、製法Aで得られた中間体化合物(a-3)に化合物(b-1)を反応させて本発明化合物(b-2)を製造する方法である。R 4 is a method for producing a compound (b-2) which is a C 1-6 lower alkylcarbonyl group, and compound (b-1) is added to the intermediate compound (a-3) obtained in the production method A. Is a method for producing the compound of the present invention (b-2).

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R3、R5、T及びUは、上記と同意義を示し、R11は、C1−6低級アルキルを示す。In the above scheme, R 1 , R 3 , R 5 , T and U have the same meaning as above, and R 11 indicates C 1-6 lower alkyl.

StepBは、不活性溶媒中、塩化チタン(IV)存在下、化合物(a-3)と化合物(b-1)から本発明化合物(b-2)を製造する工程である。
使用される不活性溶媒は、前述のものが挙げられる。
Step B is a step of producing the compound (b-2) of the present invention from the compound (a-3) and the compound (b-1) in the presence of titanium (IV) chloride in an inert solvent.
Examples of the inert solvent used include those mentioned above.

StepBにおける化合物(b-1)の使用量は、化合物(a-3)に対して、通常1〜3当量、好ましくは1〜1.5当量である。塩化チタン(IV)の使用量は、化合物(a-3)に対して、通常1〜2当量、好ましくは1〜1.5当量である。 The amount of compound (b-1) used in Step B is usually 1 to 3 equivalents, preferably 1 to 1.5 equivalents, relative to compound (a-3). The amount of titanium (IV) chloride used is usually 1 to 2 equivalents, preferably 1 to 1.5 equivalents, relative to compound (a-3).

反応温度は原料化合物または使用される溶媒によって異なるが、通常、0℃から50℃であり、好適には、0℃から室温である。
反応時間は原料化合物または使用される溶媒または反応温度によって異なるが、通常30分間から24時間であり、好適には30分間から2時間である。
The reaction temperature varies depending on the raw material compound or the solvent used, but is usually 0 ° C. to 50 ° C., preferably 0 ° C. to room temperature.
The reaction time varies depending on the starting compound, the solvent used or the reaction temperature, but is usually 30 minutes to 24 hours, preferably 30 minutes to 2 hours.

(製法C)
製法Cは、本発明化合物(I)のA環が
(Manufacturing method C)
In the production method C, the A ring of the compound (I) of the present invention is used.

Figure 2020071550
Figure 2020071550

であり、Rが、カルバモイル基である化合物(c-2)の製法であって、製法Aで得られた中間体化合物(a-3)にクロロスルホニルイソシアネートを反応させて得られる化合物(c-1)から本発明化合物(c-2)を得る方法である。R 4 is a method for producing a compound (c-2) which is a carbamoyl group, and is a compound (c) obtained by reacting the intermediate compound (a-3) obtained in the production method A with chlorosulfonyl isocyanate. This is a method for obtaining the compound of the present invention (c-2) from -1).

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R3、R5、T及びUは、上記と同意義を示す。In the above scheme, R 1 , R 3 , R 5 , T and U have the same meaning as above.

StepC1は、不活性溶媒中、化合物(a-3)にクロロスルホニルイソシアネートを反応させ、中間体化合物(c-1)を製造する工程である。
StepC2は、酢酸などにより、中間体化合物(c-1)のクロロスルホニル基を脱離し、本発明化合物(c-2)を製造する工程である。
使用される不活性溶媒は、前述のものが挙げられる。
Step C1 is a step of reacting compound (a-3) with chlorosulfonyl isocyanate in an inert solvent to produce intermediate compound (c-1).
Step C2 is a step of producing the compound (c-2) of the present invention by removing the chlorosulfonyl group of the intermediate compound (c-1) with acetic acid or the like.
Examples of the inert solvent used include those mentioned above.

StepC1におけるクロロスルホニルイソシアネートの使用量は、化合物(a-3)に対して、通常1〜3当量、好ましくは1〜1.5当量である。
反応温度は原料化合物または使用される溶媒によって異なるが、通常、0℃から50℃であり、好適には、0℃から室温である。
反応時間は原料化合物、使用される溶媒または反応温度によって異なるが、通常30分間から24時間であり、好適には30分間から2時間である。
The amount of chlorosulfonyl isocyanate used in Step C1 is usually 1 to 3 equivalents, preferably 1 to 1.5 equivalents, relative to compound (a-3).
The reaction temperature varies depending on the raw material compound or the solvent used, but is usually 0 ° C. to 50 ° C., preferably 0 ° C. to room temperature.
The reaction time varies depending on the starting compound, the solvent used or the reaction temperature, but is usually 30 minutes to 24 hours, preferably 30 minutes to 2 hours.

(製法D)
製法Dは、本発明化合物(I)のA環が
(Manufacturing method D)
In the production method D, the A ring of the compound (I) of the present invention is used.

Figure 2020071550
Figure 2020071550

であり、Rが、カルバモイル基である化合物(c-2)の製法であって、製法Aで得られた中間体化合物(a-3)のヨード化、中間体化合物(d-1)のシアノ化、中間体化合物(d-2)のシアノ基を加水分解することによって、本発明化合物(c-2)を得る方法である。R 4 is a method for producing the compound (c-2) which is a carbamoyl group, and the iodide of the intermediate compound (a-3) obtained in the production method A and the intermediate compound (d-1). This is a method for obtaining the compound (c-2) of the present invention by cyanation and hydrolysis of the cyano group of the intermediate compound (d-2).

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R3、R5、T及びUは、上記と同意義を示す。In the above scheme, R 1 , R 3 , R 5 , T and U have the same meaning as above.

StepD1は、不活性溶媒中、化合物(a-3)にN-ヨードスクシンイミドなどのヨード化剤を反応させ、中間体化合物(d-1)を製造する工程である。
StepD2は、不活性溶媒中、トリス(ジベンジリデンアセトン)ジパラジウム(0)及び1,1'-ビス(ジフェニルホスフィノ)フェロセンなどの触媒の存在下、化合物(d-1)にシアン化銅などのシアノ化剤を反応させ、中間体化合物(d-2)を製造する工程である。
StepD3は、不活性溶媒中、化合物(d-2)のシアノ基を加水分解反応に供し、カルバモイル基に変換し、本発明化合物(c-2)を製造する工程である。シアノ基の加水分解反応は、自体公知の方法に準じて行うことができる。
使用される不活性溶媒は、前述のものが挙げられる。
Step D1 is a step of reacting compound (a-3) with an iodizing agent such as N-iodosuccinimide in an inert solvent to produce intermediate compound (d-1).
In StepD2, in the presence of catalysts such as tris (dibenzylideneacetone) dipalladium (0) and 1,1'-bis (diphenylphosphino) ferrocene in an inert solvent, compound (d-1) was added to copper cyanate and the like. This is a step of producing the intermediate compound (d-2) by reacting with the cyanating agent of.
StepD3 is a step of subjecting the cyano group of the compound (d-2) to a hydrolysis reaction in an inert solvent to convert it into a carbamoyl group to produce the compound (c-2) of the present invention. The hydrolysis reaction of the cyano group can be carried out according to a method known per se.
Examples of the inert solvent used include those mentioned above.

StepD1におけるN-ヨードスクシンイミドなどのヨード化剤の使用量は、化合物(a-3)に対して、通常1〜2当量、好ましくは1〜1.1当量である。
反応温度は原料化合物または使用される溶媒によって異なるが、通常、0℃から50℃であり、好適には、0℃から室温である。
反応時間は原料化合物、使用される溶媒または反応温度によって異なるが、通常30分間から24時間であり、好適には30分間から2時間である。
The amount of an iodizing agent such as N-iodosuccinimide used in Step D1 is usually 1 to 2 equivalents, preferably 1 to 1.1 equivalents, relative to compound (a-3).
The reaction temperature varies depending on the raw material compound or the solvent used, but is usually 0 ° C. to 50 ° C., preferably 0 ° C. to room temperature.
The reaction time varies depending on the starting compound, the solvent used or the reaction temperature, but is usually 30 minutes to 24 hours, preferably 30 minutes to 2 hours.

StepD2におけるシアン化銅などのシアノ化剤の使用量は、化合物(d-1)に対して、通常1〜5当量、好ましくは1〜4当量である。トリス(ジベンジリデンアセトン)ジパラジウム(0)及び1,1'-ビス(ジフェニルホスフィノ)フェロセンなどの触媒の使用量は、化合物(d-1)に対して、通常0.01〜0.5当量、好ましくは0.01〜0.2当量である。
反応温度は原料化合物または使用される溶媒によって異なるが、通常、0℃から50℃であり、好適には、0℃から室温である。
反応時間は原料化合物、使用される溶媒または反応温度によって異なるが、通常30分間から24時間であり、好適には30分間から10時間である。
The amount of the cyanating agent such as copper cyanide used in Step D2 is usually 1 to 5 equivalents, preferably 1 to 4 equivalents, relative to compound (d-1). The amount of catalysts used, such as tris (dibenzylideneacetone) dipalladium (0) and 1,1'-bis (diphenylphosphino) ferrocene, is usually 0.01-0.5 relative to compound (d-1). Equivalent, preferably 0.01-0.2 equivalent.
The reaction temperature varies depending on the raw material compound or the solvent used, but is usually 0 ° C. to 50 ° C., preferably 0 ° C. to room temperature.
The reaction time varies depending on the starting compound, the solvent used or the reaction temperature, but is usually 30 minutes to 24 hours, preferably 30 minutes to 10 hours.

(製法E)
製法Eは、化合物(e-1)と化合物(e-2)とをカップリングさせることによって本発明化合物(I)のA環が
(Manufacturing method E)
In the production method E, the A ring of the compound (I) of the present invention is formed by coupling the compound (e-1) and the compound (e-2).

Figure 2020071550
Figure 2020071550

である本発明化合物(e-3)を製造する方法である。
本製法に用いられる化合物(e-1)は、自体公知の方法にて製造することができる。
This is a method for producing the compound of the present invention (e-3).
The compound (e-1) used in this production method can be produced by a method known per se.

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R3、R5、T、U及びXは、上記と同意義を示し、Zは、-N(R8)-、酸素原子または硫黄原子を示す。In the above scheme, R 1 , R 3 , R 5 , T, U and X have the same meaning as above, and Z indicates -N (R 8 )-, oxygen atom or sulfur atom.

StepEは、化合物(e-1)と化合物(e-2)を不活性溶媒中、塩基の存在下、有機金属触媒を用いたカップリング反応に付し、本発明化合物(e-3)を製造する工程である。また、本反応は、必要に応じて、ホスフィンリガンドの存在下に行ってもよい。
使用される不活性溶媒は、前述のものが挙げられる。
In Step E, the compound (e-1) and the compound (e-2) are subjected to a coupling reaction using an organometallic catalyst in an inert solvent in the presence of a base to produce the compound (e-3) of the present invention. It is a process to do. In addition, this reaction may be carried out in the presence of a phosphine ligand, if necessary.
Examples of the inert solvent used include those mentioned above.

塩基としては、無機炭酸塩もしくはリン酸塩(例えば、炭酸カリウム、炭酸セシウム、リン酸三カリウム)などが挙げられる。
有機金属触媒としては、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(0)、トリス(ジベンジリデンアセトン)ジパラジウム(0)、ジクロロビス(トリフェニルホスフィン)パラジウム(II)などが挙げられる。
ホスフィンリガンドとしては、2,2’−ビス(ジフェニルホスフィノ)−1,1’−ビナフチル(BINAP)、トリス(2−メチルフェニル)ホスフィン、1,1’−ビス(ジフェニルホスフィノ)フェロセン、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が挙げられる。
Examples of the base include inorganic carbonates or phosphates (for example, potassium carbonate, cesium carbonate, tripotassium phosphate) and the like.
Examples of the organometallic catalyst include palladium (II) acetate, tetrakis (triphenylphosphine) palladium (0), tris (dibenzylideneacetone) dipalladium (0), and dichlorobis (triphenylphosphine) palladium (II).
Phosphine ligands include 2,2'-bis (diphenylphosphine) -1,1'-binaphthyl (BINAP), tris (2-methylphenyl) phosphine, 1,1'-bis (diphenylphosphine) ferrocene, 2 -Dicyclohexylphosphine-2', 6'-dimethoxybiphenyl, 2-dicyclohexylphosphine-2', 4, 6'-triisopropylbiphenyl and the like can be mentioned.

反応温度や反応時間などの反応条件は、用いられる反応試薬、反応溶媒などによって異なるが、通常、反応温度は−30℃から150℃であり、反応時間は30分から20時間である。 The reaction conditions such as the reaction temperature and the reaction time vary depending on the reaction reagent used, the reaction solvent, and the like, but usually the reaction temperature is −30 ° C. to 150 ° C. and the reaction time is 30 minutes to 20 hours.

(製法F)
製法Fは、化合物(f-1)と化合物(e-2)とをカップリングさせることによって本発明化合物(I)のA環が
(Manufacturing method F)
In the production method F, the A ring of the compound (I) of the present invention is formed by coupling the compound (f-1) and the compound (e-2).

Figure 2020071550
Figure 2020071550

である本発明化合物(f-2)を製造する方法である。
本製法に用いられる化合物(f-1)は、自体公知の方法にて製造することができる。
This is a method for producing the compound of the present invention (f-2).
The compound (f-1) used in this production method can be produced by a method known per se.

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R3、R7、T、U及びXは、上記と同意義を示す。In the above scheme, R 1 , R 3 , R 7 , T, U and X have the same meaning as above.

StepFは、化合物(f-1)と化合物(e-2)を不活性溶媒中、塩基の存在下、有機金属触媒を用いたカップリング反応に付し、本発明化合物(f-2)を製造する工程であり、製法E記載のStepEの反応と同様の条件にて行うことができる。 In Step F, the compound (f-1) and the compound (e-2) are subjected to a coupling reaction using an organometallic catalyst in an inert solvent in the presence of a base to produce the compound (f-2) of the present invention. This step can be performed under the same conditions as the reaction of Step E described in Production Method E.

(製法G)
製法Gは、化合物(g-1)と化合物(e-2)とをカップリングさせることによって本発明化合物(I)のA環が
(Manufacturing method G)
In the production method G, the A ring of the compound (I) of the present invention is formed by coupling the compound (g-1) and the compound (e-2).

Figure 2020071550
Figure 2020071550

である本発明化合物(g-2)を製造する方法である。
本製法に用いられる化合物(g-1)は、自体公知の方法にて製造することができる。
This is a method for producing the compound of the present invention (g-2).
The compound (g-1) used in this production method can be produced by a method known per se.

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R3、T、U及びXは、上記と同意義を示す。In the above scheme, R 1 , R 3 , T, U and X have the same meaning as above.

StepGは、化合物(g-1)と化合物(e-2)を不活性溶媒中、塩基の存在下、有機金属触媒を用いたカップリング反応に付し、本発明化合物(g-2)を製造する工程であり、製法E記載のStepEの反応と同様の条件にて行うことができる。 StepG produces the compound (g-2) of the present invention by subjecting the compound (g-1) and the compound (e-2) to a coupling reaction using an organometallic catalyst in the presence of a base in an inert solvent. This step can be performed under the same conditions as the reaction of Step E described in Production Method E.

(製法H)
製法Hは、化合物(h-1)と化合物(a-2)とをカップリングさせることによって本発明化合物(I)のA環が
(Manufacturing method H)
In the production method H, the A ring of the compound (I) of the present invention is formed by coupling the compound (h-1) and the compound (a-2).

Figure 2020071550
Figure 2020071550

である本発明化合物(h-2)を製造する方法である。
本製法に用いられる化合物(h-1)は、自体公知の方法にて製造することができる。
This is a method for producing the compound of the present invention (h-2).
The compound (h-1) used in this production method can be produced by a method known per se.

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R、T、U及びXは、上記と同意義を示す。In the above scheme, R 1 , R 3 , T, U and X have the same meaning as above.

StepHは、無機炭酸塩もしくはリン酸塩(例えば、炭酸カリウム、炭酸セシウム、リン酸三カリウム)などの塩基、銅化合物(例えば、CuBr、CuI)及びリガンド(例えば、N,N’-ジメチルエチレンジアミン、1,2-ジアミノシクロヘキサン、trans-N,N’-ジメチルシクロヘキサン-1,2-ジアミンなど)を用いた求核置換反応であり、不活性溶媒中、化合物(h-1)と化合物(a-2)から本発明化合物(h-2)を製造する工程であり、製法A記載のStepAの反応と同様の条件にて行うことができる。 StepH refers to bases such as inorganic carbonates or phosphates (eg potassium carbonate, cesium carbonate, tripotassium phosphate), copper compounds (eg CuBr, CuI) and ligands (eg N, N'-dimethylethylenediamine, It is a nucleophilic substitution reaction using 1,2-diaminocyclohexane, trans-N, N'-dimethylcyclohexane-1,2-diamine, etc.), and is a compound (h-1) and compound (a-) in an inert solvent. This is a step of producing the compound (h-2) of the present invention from 2), and can be carried out under the same conditions as the reaction of Step A described in Production Method A.

(製法I)
製法Iは、化合物(i-1)と化合物(a-2)とを反応させ、ニトロ基を自体公知の方法にて還元し、得られる化合物(i-3)のアミノ基を塩化クロロアセチルにてアミド化後、閉環反応に付し、得られる化合物(i-5)と化合物(i-6)とを反応させることによって本発明化合物(I)のA環が
(Manufacturing method I)
In the production method I, the compound (i-1) is reacted with the compound (a-2), the nitro group is reduced by a method known per se, and the amino group of the obtained compound (i-3) is converted to chloroacetyl chloride. After amidation, the compound (i-5) is subjected to a ring-closing reaction, and the obtained compound (i-5) is reacted with the compound (i-6) to form the A ring of the compound (I) of the present invention.

Figure 2020071550
Figure 2020071550

である本発明化合物(i-7)を製造する方法である。 This is a method for producing the compound of the present invention (i-7).

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R3、R7、R9、T、U及びXは、上記と同意義を示す。In the above scheme, R 1 , R 3 , R 7 , R 9 , T, U and X have the same meaning as above.

StepI1は、化合物(i-1)と化合物(a-2)を不活性溶媒中、塩基の存在下、有機金属触媒を用いたカップリング反応に付し、化合物(i-2)を製造する工程である。また、本反応は、必要に応じて、ホスフィンリガンドの存在下に行ってもよく、製法E記載のStepEの反応と同様の条件にて行うことができる。
StepI2は、化合物(i-2)のニトロ基を、自体公知の方法にてアミノ基に還元し、化合物(i-3)を製造する工程である。
StepI3は、化合物(i-3)のアミノ基を不活性溶媒中、トリエチルアミンなどの塩基の存在下、塩化クロロアセチルを反応させ、化合物(i-4)を製造する工程である。
StepI4は、化合物(i-4)を不活性溶媒中、水素化ナトリウムなどの塩基の存在下、分子内閉環させ、化合物(i-5)を製造する工程である。
StepI5は、化合物(i-5)を不活性溶媒中、水素化ナトリウムなどの塩基の存在下、化合物(i-6)を反応させ、本発明化合物(i-7)を製造する工程である。
使用される不活性溶媒は、前述のものが挙げられる。
Step I1 is a step of producing compound (i-2) by subjecting compound (i-1) and compound (a-2) to a coupling reaction using an organometallic catalyst in the presence of a base in an inert solvent. Is. Further, this reaction may be carried out in the presence of a phosphine ligand, if necessary, and can be carried out under the same conditions as the reaction of Step E described in Production Method E.
StepI2 is a step of reducing the nitro group of compound (i-2) to an amino group by a method known per se to produce compound (i-3).
Step I3 is a step of producing compound (i-4) by reacting the amino group of compound (i-3) with chloroacetyl chloride in the presence of a base such as triethylamine in an inert solvent.
Step I4 is a step of producing compound (i-5) by intramolecularly closing the ring of compound (i-4) in an inert solvent in the presence of a base such as sodium hydride.
Step I5 is a step of producing the compound (i-7) of the present invention by reacting the compound (i-5) with the compound (i-6) in the presence of a base such as sodium hydride in an inert solvent.
Examples of the inert solvent used include those mentioned above.

StepI3における塩化クロロアセチルの使用量は、化合物(i-3)に対して、通常1〜2当量、好ましくは1〜1.2当量である。塩基の使用量は、化合物(i-3)に対して、通常1〜3当量、好ましくは1〜1.5当量である。反応温度や反応時間などの反応条件は、用いられる反応試薬、反応溶媒などによって異なるが、通常、反応温度は−30℃から50℃であり、反応時間は30分間から5時間である。
StepI4における塩基の使用量は、化合物(i-4)に対して、通常1〜3当量、好ましくは1〜1.5当量である。反応温度や反応時間などの反応条件は、用いられる反応試薬、反応溶媒などによって異なるが、通常、反応温度は−30℃から50℃であり、反応時間は30分間から5時間である。
StepI5における化合物(i-6)の使用量は、化合物(i-5)に対して、通常1〜3当量、好ましくは1〜1.5当量である。反応温度や反応時間などの反応条件は、用いられる反応試薬、反応溶媒などによって異なるが、通常、反応温度は−30℃から50℃であり、反応時間は30分間から5時間である。
The amount of chloroacetyl chloride used in Step I3 is usually 1 to 2 equivalents, preferably 1 to 1.2 equivalents, relative to compound (i-3). The amount of the base used is usually 1 to 3 equivalents, preferably 1 to 1.5 equivalents, relative to compound (i-3). The reaction conditions such as the reaction temperature and the reaction time vary depending on the reaction reagent used, the reaction solvent, and the like, but the reaction temperature is usually −30 ° C. to 50 ° C., and the reaction time is 30 minutes to 5 hours.
The amount of the base used in Step I4 is usually 1 to 3 equivalents, preferably 1 to 1.5 equivalents, relative to compound (i-4). The reaction conditions such as the reaction temperature and the reaction time vary depending on the reaction reagent used, the reaction solvent, and the like, but the reaction temperature is usually −30 ° C. to 50 ° C., and the reaction time is 30 minutes to 5 hours.
The amount of compound (i-6) used in Step I5 is usually 1 to 3 equivalents, preferably 1 to 1.5 equivalents, relative to compound (i-5). The reaction conditions such as the reaction temperature and the reaction time vary depending on the reaction reagent used, the reaction solvent, and the like, but the reaction temperature is usually −30 ° C. to 50 ° C., and the reaction time is 30 minutes to 5 hours.

(製法J)
製法Jは、化合物(j-1)と化合物(j-2)の反応から得られる化合物(j-3)と化合物(a-2)を反応させるか、または、製法Iで得られる化合物(i-4)と化合物(j-2)とを反応させて化合物(j-4)を得、化合物(j-4)を自体公知の方法にて閉環反応に付すことによって本発明化合物(I)のA環が
(Manufacturing method J)
In the production method J, the compound (j-3) obtained from the reaction of the compound (j-1) and the compound (j-2) is reacted with the compound (a-2), or the compound (i) obtained by the production method I is reacted. The compound (j-4) of the present invention is obtained by reacting -4) with the compound (j-2) to obtain the compound (j-4), and subjecting the compound (j-4) to a ring-closing reaction by a method known per se. A ring

Figure 2020071550
Figure 2020071550

である本発明化合物(j-5)を製造する方法である。 This is a method for producing the compound of the present invention (j-5).

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R3、R6、T、U及びXは、上記と同意義を示す。In the above scheme, R 1 , R 3 , R 6 , T, U and X have the same meaning as above.

StepJ1は、不活性溶媒中、化合物(j-1)と化合物(j-2)とを反応させ、化合物(j-1)のアミノ基をアミド化し、化合物(j-3)を製造する工程である。本反応は、必要に応じて、トリエチルアミン、ピリジン、4−ジメチルアミノピリジン、炭酸カリウム等の塩基の存在下に行ってもよい。
StepJ2は、化合物(j-3)と化合物(a-2)とを不活性溶媒中、塩基の存在下、有機金属触媒を用いたカップリング反応に付し、化合物(j-4)を製造する工程である。また、本反応は、必要に応じて、ホスフィンリガンドの存在下に行ってもよく、製法Eに記載のStepEの反応と同様の条件にて行うことができる。
StepJ3は、不活性溶媒中、製法Iで得られる化合物(i-4)と化合物(j-2)とを反応させ化合物(j-4)を製造する工程である。StepJ1の反応と同様の条件にて行うことができる。
StepJ4は、化合物(j-4)を不活性溶媒中、自体公知の方法で分子内閉環し、本発明化合物(j-5)を製造する工程である。
使用される不活性溶媒は、前述のものが挙げられる。
StepJ1 is a step of reacting compound (j-1) with compound (j-2) in an inert solvent to amidate the amino group of compound (j-1) to produce compound (j-3). be. This reaction may be carried out in the presence of bases such as triethylamine, pyridine, 4-dimethylaminopyridine and potassium carbonate, if necessary.
In StepJ2, compound (j-3) and compound (a-2) are subjected to a coupling reaction using an organometallic catalyst in an inert solvent in the presence of a base to produce compound (j-4). It is a process. Further, this reaction may be carried out in the presence of a phosphine ligand, if necessary, and can be carried out under the same conditions as the reaction of Step E described in Production Method E.
StepJ3 is a step of producing the compound (j-4) by reacting the compound (i-4) obtained by the production method I with the compound (j-2) in an inert solvent. It can be carried out under the same conditions as the reaction of Step J1.
StepJ4 is a step of producing the compound (j-5) of the present invention by intramolecularly closing the compound (j-4) in an inert solvent by a method known per se.
Examples of the inert solvent used include those mentioned above.

StepJ1において、化合物(j-2)は、遊離酸の形態だけでなく、塩(例えば、ナトリウム、カリウム、カルシウム、トリエチルアミン、ピリジン等との塩)や反応性誘導体(例えば、酸クロライド、酸ブロマイド等の酸ハライド;酸無水物;ジアルキルリン酸等の置換リン酸、モノエチル炭酸等のアルキル炭酸との混合酸無水物;イミダゾール等とのアミドである活性アミド;シアノメチルエステル、4−ニトロフェニルエステル等の活性エステル)等として当該反応に供される。 In StepJ1, the compound (j-2) is not only in the form of a free acid, but also a salt (for example, a salt with sodium, potassium, calcium, triethylamine, pyridine, etc.) or a reactive derivative (for example, acid chloride, acid bromide, etc.). Acid halide; Acid anhydride; Substituted phosphoric acid such as dialkyl phosphate, mixed acid anhydride with alkyl carbonate such as monoethyl carbonate; Active amide which is an amide with imidazole, etc .; Cyanomethyl ester, 4-nitrophenyl ester, etc. Is subjected to the reaction as an active ester) or the like.

また、StepJ1において化合物(j-2)を遊離酸または塩の状態で使用する場合には、縮合剤の存在下で反応を行うのが好ましく、縮合剤としては、例えばN,N’−ジシクロヘキシルカルボジイミド等のN,N’−ジ置換カルボジイミド類;1−エチル−3−(3’−ジメチルアミノプロピル)カルボジイミド、N−シクロヘキシル−N’−モルホリノエチルカルボジイミド、N−シクロヘキシル−N’−(4−ジエチルアミノシクロヘキシル)カルボジイミド等のカルボジイミド化合物;N,N’−カルボニルジイミダゾール、N,N’−チオニルジイミダゾール等のアゾライド化合物等が用いられる。上記縮合剤を用いた場合、反応は化合物(j-2)の反応性誘導体を経て反応が進行すると考えられる。 When the compound (j-2) is used in the state of a free acid or a salt in Step J1, the reaction is preferably carried out in the presence of a condensing agent, and the condensing agent is, for example, N, N'-dicyclohexylcarbodiimide. N, N'-di-substituted carbodiimides such as 1-ethyl-3- (3'-dimethylaminopropyl) carbodiimide, N-cyclohexyl-N'-morpholinoethyl carbodiimide, N-cyclohexyl-N'-(4-diethylamino). Carbodiimide compounds such as cyclohexyl) carbodiimide; azolide compounds such as N, N'-carbonyldiimidazole and N, N'-thionyldiimidazole are used. When the above condensing agent is used, it is considered that the reaction proceeds via the reactive derivative of compound (j-2).

StepJ1における化合物(j-2)の使用量は、化合物(j-1)に対して、通常1〜5当量、好ましくは1〜3当量である。塩基の使用量は、化合物(j-1)に対して、通常1〜5当量、好ましくは1〜3当量である。
反応温度や反応時間などの反応条件は、用いられる反応試薬、反応溶媒などによって異なるが、通常、反応温度は−30℃から100℃であり、反応時間は30分間から20時間である。
The amount of compound (j-2) used in Step J1 is usually 1 to 5 equivalents, preferably 1 to 3 equivalents, relative to compound (j-1). The amount of the base used is usually 1 to 5 equivalents, preferably 1 to 3 equivalents, relative to compound (j-1).
The reaction conditions such as the reaction temperature and the reaction time vary depending on the reaction reagent used, the reaction solvent, and the like, but usually the reaction temperature is −30 ° C. to 100 ° C., and the reaction time is 30 minutes to 20 hours.

(製法K)
製法Kは、化合物(k-1)と化合物(e-2)とをカップリングさせることによって本発明化合物(I)のA環が
(Manufacturing method K)
In the production method K, the A ring of the compound (I) of the present invention is formed by coupling the compound (k-1) and the compound (e-2).

Figure 2020071550
Figure 2020071550

である本発明化合物(k-2)を製造する方法である。
本製法に用いられる化合物(k-1)は、自体公知の方法にて製造することができる。
This is a method for producing the compound of the present invention (k-2).
The compound (k-1) used in this production method can be produced by a method known per se.

Figure 2020071550
Figure 2020071550

上記スキーム中、R1、R3、T、U及びXは、上記と同意義を示す。In the above scheme, R 1 , R 3 , T, U and X have the same meaning as above.

使用される不活性溶媒は、前述のものが挙げられる。 Examples of the inert solvent used include those mentioned above.

StepKは、化合物(k-1)と化合物(e-2)を不活性溶媒中、塩基の存在下、有機金属触媒を用いたカップリング反応に付し、本発明化合物(k-2)を製造する工程であり、製法E記載のStepEの反応と同様の条件にて行うことができる。 StepK produces the compound (k-2) of the present invention by subjecting the compound (k-1) and the compound (e-2) to a coupling reaction using an organometallic catalyst in the presence of a base in an inert solvent. This step can be performed under the same conditions as the reaction of Step E described in Production Method E.

上記の方法で製造された本発明の化合物(I)は、慣用の精製手段、例えば、濃縮、抽出、クロマトグラフィー、再沈殿、再結晶等の手段を施すことにより、任意の純度に精製することができる。また、必要に応じて適当な溶媒中、酸または塩基等と処理することでその医薬上許容される塩とすることができる。さらに、本発明の化合物(I)またはその医薬上許容される塩は、大気中に放置したり、水、含水溶媒またはその他の溶媒(例えば、アルコール等)と処理したり、または再結晶をすることにより、水分(または溶媒)を吸収し、吸着水(または吸着溶媒)が付いて、水和物(または溶媒和物)となったりする場合がある。そのような各種の水和物、溶媒和物及び結晶多形の化合物も包含する。 The compound (I) of the present invention produced by the above method is purified to an arbitrary purity by subject to conventional purification means such as concentration, extraction, chromatography, reprecipitation, recrystallization and the like. Can be done. Further, if necessary, the salt can be obtained as a pharmaceutically acceptable salt by treating with an acid, a base or the like in an appropriate solvent. Further, compound (I) of the present invention or a pharmaceutically acceptable salt thereof is left in the air, treated with water, a hydrated solvent or other solvent (for example, alcohol), or recrystallized. As a result, water (or solvent) may be absorbed and adsorbed water (or adsorbed solvent) may be attached to form a hydrate (or solvent). It also includes such various hydrates, solvates and polymorphic compounds.

本発明の化合物(I)またはその医薬上許容される塩は、置換基の種類や組み合わせによって、シス体、トランス体等の幾何異性体、互変異性体またはd体、l体等の光学異性体等の各種異性体が存在し得るが、本発明の化合物(I)は、特に限定していない場合はそれら全ての異性体、立体異性体及びいずれの比率のこれら異性体及び立体異性体混合物をも包含するものである。これら光学異性体や異性体の混合物は、公知の分割手段(例えば、J.Jacquesらの、「Enantiomers, Racemates and Resolution, John Wiley And Sons,Inc.」等参照)により単離することができる。また、不斉合成によっても上記異性体を製造することができる。また、本発明の化合物(I)は、ラベル体、すなわち、本発明の化合物(I)を構成する1または2以上の原子を同位元素(例えば、H、H、13C、14C、35S等)で置換した化合物も包含される。The compound (I) of the present invention or a pharmaceutically acceptable salt thereof is a geometric isomer such as a cis isomer or a trans isomer, or an optically isomer such as a t-form or a d-form or l-form, depending on the type and combination of substituents. Various isomers such as isomers may exist, but the compound (I) of the present invention is all isomers, steric isomers and mixtures of these isomers and steric isomers in any ratio unless otherwise specified. Also includes. These optical isomers and mixtures of isomers can be isolated by known partitioning means (see, eg, "Enantiomers, Racemates and Resolution, John Wiley And Sons, Inc." by J. Jacques et al.). The isomer can also be produced by asymmetric synthesis. The compounds (I) of the present invention, the label body, i.e., the compounds of the present invention (I) 1 or 2 or more atoms isotopes constituting the (e.g., 2 H, 3 H, 13 C, 14 C, Compounds substituted with 35 S etc.) are also included.

また、本発明には、本発明の化合物(I)の医薬上許容される、いわゆる、プロドラッグも包含される。医薬上許容されるプロドラッグとは、加水分解により、若しくは、生理学的条件下で、本発明の化合物(I)のアミノ基、水酸基、カルボキシ基等に変換し得る基を有する化合物であり、このようなプロドラッグを形成する基としては、Prog. Med.、第5巻、2157−2161ページ、1985年や、「医薬品の開発」(廣川書店、1990年)第7巻、分子設計163−198ページに記載の基である。当該プロドラッグとして、より具体的には、本発明の化合物(I)に、アミノ基が存在する場合には、そのアミノ基がアシル化、アルキル化、りん酸化された化合物(例えば、そのアミノ基がエイコサノイル化、アラニル化、ペンチルアミノカルボニル化、(5−メチル−2−オキソ−1,3−ジオキソレン−4−イル)メトキシカルボニル化、テトラヒドロフラニル化、ピロリジルメチル化、ピバロイルオキシメチル化、tert−ブチル化された化合物等である。)等を挙げることができ、本発明の化合物(I)に、水酸基が存在する場合には、その水酸基がアシル化、アルキル化、リン酸化、ホウ酸化された化合物(例えば、その水酸基がアセチル化、パルミトイル化、プロパノイル化、ピバロイル化、サクシニル化、フマリル化、アラニル化、ジメチルアミノメチルカルボニル化された化合物等である。)等を挙げることができる。また、本発明の化合物(I)に、カルボキシ基が存在する場合には、そのカルボキシ基がエステル化、アミド化された化合物(例えば、そのカルボキシ基がエチル エステル化、フェニル エステル化、カルボキシメチル エステル化、ジメチルアミノメチル エステル化、ピバロイルオキシメチル エステル化、1−エトキシカルボニルオキシエチル エステル化、1−シクロヘキシルオキシカルボニルオキシエチル エステル化、アミド化またはメチルアミド化された化合物等である。)等が挙げられる。 The present invention also includes so-called prodrugs, which are pharmaceutically acceptable, of the compound (I) of the present invention. A pharmaceutically acceptable prodrug is a compound having a group that can be converted into an amino group, a hydroxyl group, a carboxy group, etc. of the compound (I) of the present invention by hydrolysis or under physiological conditions. The groups that form such prodrugs include Prog. Med., Vol. 5, pp. 2157-2161, 1985, and "Pharmaceutical Development" (Hirokawa Shoten, 1990), Vol. 7, Molecular Design 163-198. It is the basis described on the page. More specifically, as the prodrug, when the compound (I) of the present invention has an amino group, the amino group is acylated, alkylated, or phosphorylated (for example, the amino group thereof). Eikosanoylation, alanylation, pentylaminocarbonylation, (5-methyl-2-oxo-1,3-dioxolen-4-yl) methoxycarbonylation, tetrahydrofuranylation, pyrrolidylmethylation, pivaloyloxymethylation , A tert-butylated compound, etc.), etc., and when the compound (I) of the present invention has a hydroxyl group, the hydroxyl group is acylated, alkylated, phosphorylated, or boroinated. Examples thereof include an oxidized compound (for example, a compound whose hydroxyl group is acetylated, palmitoylated, propanoylated, pivaloylated, succinylated, fumarylated, alanylated, dimethylaminomethylcarbonylated, etc.) and the like. .. When the compound (I) of the present invention has a carboxy group, the compound in which the carboxy group is esterified or amidated (for example, the carboxy group is ethyl esterified, phenyl esterified, or carboxymethyl ester). , Dimethylaminomethyl esterification, pivaloyloxymethyl esterification, 1-ethoxycarbonyloxyethyl esterification, 1-cyclohexyloxycarbonyloxyethyl esterification, amidated or methylamided compounds, etc.) Can be mentioned.

本発明の化合物(I)またはその医薬上許容される塩は、サイクリン依存性キナーゼ(CDK)8阻害活性を有し、哺乳動物(ヒト、ウマ、ウシ、イヌ、ネコ、ラット、マウス、ハムスター等)に対して、CDK8に関連する疾患、骨粗鬆症等の骨代謝に関連する疾患、癌等の細胞増殖性疾患の予防・治療剤として有用である。すなわち、本発明の化合物(I)またはその医薬上許容される塩は、例えば、骨粗鬆症、骨形成不全症、軟骨無形成症、線維性骨炎(副甲状腺機能亢進症)、骨軟化症、ページェット病、慢性関節リューマチ、変形性関節炎、癌、自己免疫疾患、炎症性疾患等の予防および治療剤として使用することができる。その場合、本発明の化合物(I)を、そのままあるいは医薬上許容される担体を配合し、医薬(すなわち、医薬組成物)として、哺乳動物(好ましくは、ヒト)に経口的または非経口的に投与することができる。 The compound (I) of the present invention or a pharmaceutically acceptable salt thereof has cyclin-dependent kinase (CDK) 8 inhibitory activity, and mammals (humans, horses, cows, dogs, cats, rats, mice, hamsters, etc.) ), It is useful as a prophylactic / therapeutic agent for CDK8-related diseases, bone metabolism-related diseases such as osteoporosis, and cell-proliferative diseases such as cancer. That is, the compound (I) of the present invention or a pharmaceutically acceptable salt thereof may be, for example, osteoporosis, osteodysplasia, chondropathy, fibroosteitis (hyperparathyroidism), osteomalacia, page. It can be used as a prophylactic and therapeutic agent for et's disease, rheumatoid arthritis, osteomalacia, cancer, autoimmune disease, inflammatory disease and the like. In that case, the compound (I) of the present invention may be used as it is or by blending a pharmaceutically acceptable carrier, orally or parenterally to a mammal (preferably human) as a medicine (that is, a pharmaceutical composition). Can be administered.

本発明の化合物(I)が適用される癌としては、例えば、大腸癌(例、結腸癌、直腸癌、肛門癌、家族性大腸癌、遺伝性非ポリポーシス大腸癌、消化管間質腫瘍)、肺癌(例、非小細胞肺癌、小細胞肺癌、悪性中皮腫)、中皮腫、膵臓癌(例、膵管癌、膵内分泌腫瘍)、咽頭癌、喉頭癌、食道癌、胃癌(例、乳頭腺癌、粘液性腺癌、腺扁平上皮癌)、十二指腸癌、小腸癌、乳癌(例、浸潤性乳管癌、非浸潤性乳管癌、炎症性乳癌)、卵巣癌(例、上皮性卵巣癌、性腺外胚細胞腫瘍、卵巣性胚細胞腫瘍、卵巣低悪性度腫瘍)、精巣腫瘍、前立腺癌(例、ホルモン依存性前立腺癌、ホルモン非依存性前立腺癌、去勢療法抵抗性前立腺癌)、肝臓癌(例、肝細胞癌、原発性肝癌、肝外胆管癌)、甲状腺癌(例、甲状腺髄様癌)、腎臓癌(例、腎細胞癌、腎盂と尿管の移行上皮癌)、子宮癌(例、子宮内膜癌、子宮頚部癌、子宮体部癌、子宮肉腫)、妊娠性絨毛癌、脳腫瘍(例、髄芽細胞腫、神経膠腫、松果体星細胞腫瘍、毛様細胞性星細胞腫、びまん性星細胞腫、退形成性星細胞腫、下垂体腺腫)、網膜芽細胞腫、皮膚癌(例、基底細胞腫、悪性黒色腫)、肉腫(例、横紋筋肉腫、平滑筋肉腫、軟部肉腫、紡錘細胞肉腫)、悪性骨腫瘍、膀胱癌、血液癌(例、多発性骨髄腫、白血病(例、急性骨髄性白血病)、悪性リンパ腫、ホジキン病、慢性骨髄増殖性疾患)、原発不明癌等が挙げられる。
中でも、本発明の化合物(I)は、乳癌、膵臓癌、膀胱癌、前立腺癌、食道癌、胃癌、子宮癌、卵巣癌、脳腫瘍、大腸癌(例、結腸癌、直腸癌)、血液癌(例、急性骨髄性白血病、多発性骨髄腫)、肝臓癌(例、肝細胞癌)、皮膚癌、肺癌及び甲状腺癌からなる群から選ばれる少なくとも1種の癌に対して有効である。
Cancers to which the compound (I) of the present invention is applied include, for example, colon cancer (eg, colon cancer, rectal cancer, anal cancer, familial colon cancer, hereditary non-polyposis colon cancer, gastrointestinal stromal tumor). Pulmonary cancer (eg, non-small cell lung cancer, small cell lung cancer, malignant mesoderma), mesopharyngoma, pancreatic cancer (eg, pancreatic duct cancer, pancreatic endocrine tumor), pharyngeal cancer, laryngeal cancer, esophageal cancer, gastric cancer (eg, papillary) Adenocarcinoma, mucinous adenocarcinoma, squamous epithelial cancer), duodenal cancer, small intestinal cancer, breast cancer (eg, invasive ductal carcinoma, non-invasive ductal carcinoma, inflammatory breast cancer), ovarian cancer (eg, epithelial ovarian cancer) , Extragonal embryonic cell tumor, ovarian embryonic cell tumor, low-grade ovarian tumor), testicular tumor, prostate cancer (eg, hormone-dependent prostate cancer, hormone-independent prostate cancer, castration-resistant prostate cancer), liver Cancer (eg, hepatocellular carcinoma, primary liver cancer, extrahepatic bile duct cancer), thyroid cancer (eg, thyroid medullary cancer), kidney cancer (eg, renal cell cancer, metastatic epithelial cancer of the renal pelvis and urinary tract), uterine cancer (Example, endometrial cancer, cervical cancer, uterine body cancer, uterine sarcoma), gestational chorionic villus cancer, brain tumor (eg, medullary cell tumor, glioma, pineapple stellate cell tumor, hairy cell type Star cell tumor, diffuse stell cell tumor, degenerative stell cell tumor, pituitary adenoma), retinoblastoma, skin cancer (eg, basal cell tumor, malignant melanoma), sarcoma (eg, rhizome myoma, Smooth muscle tumor, soft sarcoma, spindle cell sarcoma), malignant bone tumor, bladder cancer, hematological cancer (eg, multiple myeloma, leukemia (eg, acute myeloid leukemia), malignant lymphoma, Hodgkin's disease, chronic myeloproliferative disorder ), Cancer of unknown primary, etc.
Among them, the compound (I) of the present invention contains breast cancer, pancreatic cancer, bladder cancer, prostate cancer, esophageal cancer, gastric cancer, uterine cancer, ovarian cancer, brain tumor, colon cancer (eg, colon cancer, rectal cancer), and hematological cancer (eg, colon cancer, rectal cancer). It is effective against at least one cancer selected from the group consisting of eg, acute myeloid leukemia, multiple myeloma), liver cancer (eg, hepatocellular carcinoma), skin cancer, lung cancer and thyroid cancer.

本発明の医薬は、製剤技術分野で一般的に用いられている公知の製造方法(例、日本薬局方に記載の方法)により製造することができる。また、本発明の医薬には、必要に応じて、製剤分野において通常用いられる賦形剤、結合剤、崩壊剤、滑沢剤、甘味剤、界面活性剤、懸濁化剤、乳化剤、着色剤、保存剤、芳香剤、矯味剤、安定剤、粘稠剤等の添加剤を適宜、適量含有させることができる。
前記した医薬上に許容される担体としては、これらの添加剤が挙げられる。
The pharmaceutical product of the present invention can be produced by a known production method generally used in the field of pharmaceutical technology (eg, the method described in the Japanese Pharmacopoeia). In addition, the medicament of the present invention includes excipients, binders, disintegrants, lubricants, sweeteners, surfactants, suspending agents, emulsifiers, and colorants commonly used in the pharmaceutical field, if necessary. , Preservatives, fragrances, flavoring agents, stabilizers, thickeners and other additives can be appropriately contained.
Examples of the pharmaceutically acceptable carrier described above include these additives.

例えば、錠剤は、賦形剤、結合剤、崩壊剤、滑沢剤等を用いて製造することができ、丸剤及び顆粒剤は、賦形剤、結合剤、崩壊剤を用いて製造することができる。また、散剤及びカプセル剤は賦形剤等を、シロップ剤は甘味剤等を、乳剤または懸濁剤は懸濁化剤、界面活性剤、乳化剤等を用いて製造することができる。 For example, tablets can be produced using excipients, binders, disintegrants, lubricants and the like, and pills and granules can be produced using excipients, binders, disintegrants and the like. Can be done. Further, powders and capsules can be produced using excipients and the like, syrups can be produced using sweeteners and the like, and emulsions or suspending agents can be produced using suspending agents, surfactants, emulsifiers and the like.

賦形剤の例としては、乳糖、白糖、ブドウ糖、でんぷん、蔗糖、微結晶セルロース、カンゾウ末、マンニトール、炭酸水素ナトリウム、リン酸カルシウム、硫酸カルシウムが挙げられる。
結合剤の例としては、5ないし10重量%デンプンのり液、10ないし20重量%アラビアゴム液またはゼラチン液、1ないし5重量%トラガント液、カルボキシメチルセルロース液、アルギン酸ナトリウム液、グリセリンが挙げられる。
崩壊剤の例としては、でんぷん、炭酸カルシウムが挙げられる。
滑沢剤の例としては、ステアリン酸マグネシウム、ステアリン酸、ステアリン酸カルシウム、精製タルクが挙げられる。
甘味剤の例としては、ブドウ糖、果糖、転化糖、ソルビトール、キシリトール、グリセリン、単シロップが挙げられる。
界面活性剤の例としては、ラウリル硫酸ナトリウム、ポリソルベート80、ソルビタンモノ脂肪酸エステル、ステアリン酸ポリオキシル40が挙げられる。
懸濁化剤の例としては、アラビアゴム、アルギン酸ナトリウム、カルボキシメチルセルロースナトリウム、メチルセルロース、ベントナイトが挙げられる。
乳化剤の例としては、アラビアゴム、トラガント、ゼラチン、ポリソルベート80が挙げられる。
Examples of excipients include lactose, sucrose, glucose, starch, sucrose, microcrystalline cellulose, citrus powder, mannitol, sodium hydrogen carbonate, calcium phosphate, calcium sulfate.
Examples of the binder include 5 to 10% by weight starch paste solution, 10 to 20% by weight Arabic rubber solution or gelatin solution, 1 to 5% by weight tragant solution, carboxymethyl cellulose solution, sodium alginate solution, and glycerin.
Examples of disintegrants include starch and calcium carbonate.
Examples of lubricants include magnesium stearate, stearic acid, calcium stearate, and purified talc.
Examples of sweeteners include glucose, fructose, invert sugar, sorbitol, xylitol, glycerin, and simple syrup.
Examples of surfactants include sodium lauryl sulfate, polysorbate 80, sorbitan monofatty acid ester, and polyoxyl 40 stearate.
Examples of suspending agents include gum arabic, sodium alginate, sodium carboxymethyl cellulose, methyl cellulose and bentonite.
Examples of emulsifiers include gum arabic, tragant, gelatin and polysorbate 80.

例えば、本発明の医薬が錠剤である場合、該錠剤は、自体公知の方法に従い、本発明化合物に、例えば、賦形剤(例、乳糖、白糖、デンプン)、崩壊剤(例、デンプン、炭酸カルシウム)、結合剤(例、デンプン、アラビアゴム、カルボキシメチルセルロース、ポリビニルピロリドン、ヒドロキシプロピルセルロース)または滑沢剤(例、タルク、ステアリン酸マグネシウム、ポリエチレングリコール6000)を添加して圧縮成形し、次いで必要により、味のマスキング、腸溶性あるいは持続性の目的のため自体公知の方法でコーティングすることにより製造することができる。コーティングに用いられるコーティング剤としては、例えば、ヒドロキシプロピルメチルセルロース、エチルセルロース、ヒドロキシメチルセルロース、ヒドロキシプロピルセルロース、ポリオキシエチレングリコール、ツイーン80、プルロニックF68、セルロースアセテートフタレート、ヒドロキシプロピルメチルセルロースフタレート、ヒドロキシメチルセルロースアセテートサクシネート、オイドラギット(ローム社製、ドイツ、メタアクリル酸・アクリル酸共重合体)及び色素(例、ベンガラ、二酸化チタン)が用いられる。 For example, when the medicament of the present invention is a tablet, the tablet can be added to the compound of the present invention according to a method known per se, for example, an excipient (eg, lactose, sucrose, starch), a disintegrant (eg, starch, carbonate). Calcium), binders (eg starch, gum arabic, carboxymethyl cellulose, polyvinylpyrrolidone, hydroxypropyl cellulose) or lubricants (eg talc, magnesium stearate, polyethylene glycol 6000) are added and compression molded, then required It can be produced by coating by a method known per se for the purpose of taste masking, enteric or persistent. Examples of the coating agent used for coating include hydroxypropylmethyl cellulose, ethyl cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, polyoxyethylene glycol, Tween 80, Pluronic F68, cellulose acetate phthalate, hydroxypropyl methyl cellulose phthalate, and hydroxymethyl cellulose acetate succinate. Eudragit (Roam, Germany, methacrylic acid / acrylic acid copolymer) and dyes (eg, Bengala, titanium dioxide) are used.

前記注射剤としては、静脈注射剤のほか、皮下注射剤、皮内注射剤、筋肉注射剤、腹腔内注射剤、点滴注射剤等が含まれる。 In addition to intravenous injections, the injections include subcutaneous injections, intradermal injections, intramuscular injections, intraperitoneal injections, drip injections and the like.

かかる注射剤は、自体公知の方法、すなわち、本発明の化合物(I)を無菌の水性液もしくは油性液に溶解、懸濁または乳化することによって調製される。水性液としては、生理食塩水、ブドウ糖やその他の補助薬を含む等張液(例、D−ソルビトール、D−マンニトール、塩化ナトリウム)等が挙げられる。該水性液は適当な溶解補助剤、例えば、アルコール(例、エタノール)、ポリアルコール(例、プロピレングリコール、ポリエチレングリコール)、非イオン性界面活性剤(例、ポリソルベート80、HCO−50)を含んでいてもよい。油性液としては、ゴマ油、大豆油等が挙げられる。該油性液は適当な溶解補助剤を含んでいてもよい。該溶解補助剤としては、安息香酸ベンジル、ベンジルアルコール等が挙げられる。また、該注射剤には緩衝剤(例、リン酸緩衝液、酢酸ナトリウム緩衝液)、無痛化剤(例、塩化ベンザルコニウム、塩酸プロカイン)、安定剤(例、ヒト血清アルブミン、ポリエチレングリコール)、保存剤(例、ベンジルアルコール、フェノール)等を配合してもよい。調製された注射液は、通常、アンプルに充填される。 Such injections are prepared by a method known per se, that is, by dissolving, suspending or emulsifying compound (I) of the present invention in a sterile aqueous or oily solution. Examples of the aqueous solution include physiological saline, an isotonic solution containing glucose and other auxiliary agents (eg, D-sorbitol, D-mannitol, sodium chloride) and the like. The aqueous solution contains suitable solubilizers such as alcohols (eg ethanol), polyalcohols (eg propylene glycol, polyethylene glycol), nonionic surfactants (eg polysorbate 80, HCO-50). You may. Examples of the oily liquid include sesame oil and soybean oil. The oily liquid may contain a suitable dissolution aid. Examples of the solubilizing agent include benzyl benzoate and benzyl alcohol. In addition, the injection includes a buffer (eg, phosphate buffer, sodium acetate buffer), a soothing agent (eg, benzalkonium chloride, prokine hydrochloride), and a stabilizer (eg, human serum albumin, polyethylene glycol). , Preservatives (eg, benzyl alcohol, phenol) and the like may be blended. The prepared injection solution is usually filled in an ampoule.

上記以外に、常法を利用して適宜好ましい製剤とすることもできる。 In addition to the above, a preferable preparation can be appropriately prepared by using a conventional method.

本発明の医薬中の添加剤の含有量は、製剤の形態に応じて相違するが、通常、製剤全体に対して約1ないし約99.9重量%、好ましくは約10ないし約90重量%である。 The content of the additive in the pharmaceutical product of the present invention varies depending on the form of the preparation, but is usually about 1 to about 99.9% by weight, preferably about 10 to about 90% by weight, based on the entire preparation. be.

本発明の化合物またはその医薬上許容される塩の投与量は、投与対象、症状、その他の要因によって異なるが、例えば、成人(体重約60kg)に経口投与する場合には、1日当りの投与量は、本発明化合物として約1ないし約1000mg、好ましくは約3ないし約300mg、さらに好ましくは約10ないし約200mgであり、これらを1回又は数回に分けて投与するのが望ましい。 The dose of the compound of the present invention or a pharmaceutically acceptable salt thereof varies depending on the administration subject, symptoms, and other factors, but for example, when orally administered to an adult (body weight of about 60 kg), the daily dose is administered. As the compound of the present invention, it is about 1 to about 1000 mg, preferably about 3 to about 300 mg, more preferably about 10 to about 200 mg, and it is desirable to administer these in one or several divided doses.

また、本発明の化合物またはその医薬上許容される塩を抗癌剤として使用する場合、その薬効を損なわない限り、他の薬剤、例えば、既存の抗癌剤と併用することができる。この際、投与時期は限定されず、これらを投与対象に対し、同時に投与してもよいし、時間差をおいて投与してもよい。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、本発明の化合物またはその医薬上許容される塩等と併用薬の配合比は、投与対象、投与ルート、対象疾患、症状、組み合わせ等に応じて適宜選択することができる。 In addition, when the compound of the present invention or a pharmaceutically acceptable salt thereof is used as an anticancer agent, it can be used in combination with another agent, for example, an existing anticancer agent, as long as its efficacy is not impaired. At this time, the administration time is not limited, and these may be administered to the administration subject at the same time or may be administered at different times. The dose can be appropriately selected based on the clinically used dose. In addition, the compounding ratio of the compound of the present invention or a pharmaceutically acceptable salt thereof and the concomitant drug can be appropriately selected according to the administration target, administration route, target disease, symptom, combination and the like.

既存の抗癌剤としては、例えば、化学療法剤、ホルモン療法剤、免疫療法剤、分子標的薬、免疫チェックポイント阻害剤(抗PD−1抗体、抗PD−L1抗体)等が挙げられる。
「化学療法剤」としては、例えば、アルキル化剤、代謝拮抗剤、抗癌性抗生物質、植物由来抗癌剤等が用いられる。
「アルキル化剤」としては、例えば、ナイトロジェンマスタード、塩酸ナイトロジェンマスタード−N−オキシド、クロラムブシル、シクロフォスファミド、イホスファミド、チオテパ、カルボコン、トシル酸インプロスルファン、ブスルファン、塩酸ニムスチン、ミトブロニトール、メルファラン、ダカルバジン、ラニムスチン、ベンダムスチン、プロカルバジン、リン酸エストラムスチンナトリウム、トリエチレンメラミン、カルムスチン、ロムスチン、ストレプトゾシン、ピポブロマン、エトグルシド、カルボプラチン、シスプラチン、ミボプラチン、ネダプラチン、オキサリプラチン、アルトレタミン、アンバムスチン、塩酸ジブロスピジウム、フォテムスチン、プレドニムスチン、プミテパ、リボムスチン、テモゾロミド、トレオスルファン、トロフォスファミド、ジノスタチンスチマラマー、アドゼレシン、システムスチン、ビゼレシン、およびそれらのDDS製剤等が用いられる。
Examples of existing anticancer agents include chemotherapeutic agents, hormonal therapeutic agents, immunotherapeutic agents, molecular targeted agents, immune checkpoint inhibitors (anti-PD-1 antibody, anti-PD-L1 antibody) and the like.
As the "chemotherapeutic agent", for example, an alkylating agent, an antimetabolite, an anticancer antibiotic, a plant-derived anticancer agent and the like are used.
Examples of the "alkylating agent" include nitrogen mustard, nitrogen mustard-N-oxide hydrochloride, chlorambucil, cyclophosphamide, ifosfamide, thiotepa, carbocon, improsulfan tosylate, busulfan, nimustine hydrochloride, mitobronitol, and melphalan. Faran, dacarbazine, lanimustine, bendamustine, procarbazine, sodium estramustine phosphate, triethylenemelamine, carmustine, lomustine, streptozocin, pipobroman, etoglucid, carboplatin, cisplatin, miboplatin, nedaplatin, oxaliplatin, altretamine, ambamstine hydrochloride Pydium, fotemustine, prednimustine, pumitepa, ribomustine, temozolomid, treosfamide, trophosphamide, diostatinstimalamare, adzelesin, cisplatin, bizelesin, and their DDS preparations are used.

「代謝拮抗剤」としては、例えば、メルカプトプリン、6−メルカプトプリンリボシド、チオイノシン、メトトレキサート、ペメトレキセド、エノシタビン、シタラビン、シタラビンオクフォスファート、塩酸アンシタビン、5−FU系薬剤(例、フルオロウラシル、テガフール、UFT、ドキシフルリジン、カルモフール、ガロシタビン、エミテフール、カペシタビン)、アミノプテリン、ネルザラビン、ロイコボリンカルシウム、タブロイド、ブトシン、フォリネイトカルシウム、レボフォリネイトカルシウム、クラドリビン、クロファラビン、ネララビン、エミテフール、フルダラビン、ゲムシタビン、ヒドロキシカルバミド、ペントスタチン、ピリトレキシム、イドキシウリジン、ミトグアゾン、チアゾフリン、およびそれらのDDS製剤等が用いられる。 Examples of the "antimetabolite" include mercaptopurine, 6-mercaptopurine riboside, thioinosin, methotrexate, pemetrexed, enocitabine, cytarabine, cytarabine octofusphate, ancitabine hydrochloride, 5-FU drugs (eg, fluorouracil, tegafur, etc.). UFT, doxiflulysine, carmofur, galositabin, emitefur, capecitabine), aminopterin, nerzarabin, leucovorin calcium, tabloid, butocin, forinate calcium, levofolinate calcium, cladribine, clofarabine, nerarabine, emitefur, fludarabin Statins, pyritrexim, idoxyuridine, mitoguazone, cytarabine, and DDS preparations thereof and the like are used.

「抗癌性抗生物質」としては、例えば、アクチノマイシンD、アクチノマイシンC、マイトマイシンC、クロモマイシンA3、塩酸ブレオマイシン、硫酸ブレオマイシン、硫酸ペプロマイシン、塩酸ダウノルビシン、塩酸ドキソルビシン、塩酸アクラルビシン、塩酸ピラルビシン、塩酸エピルビシン、ネオカルチノスタチン、ミスラマイシン、ザルコマイシン、カルチノフィリン、ミトタン、塩酸ゾルビシン、塩酸ミトキサントロン、塩酸イダルビシン、およびそれらのDDS製剤等が用いられる。 Examples of the "anticancer antibiotic" include actinomycin D, actinomycin C, mitomycin C, chromomycin A3, bleomycin hydrochloride, bleomycin sulfate, pepromycin sulfate, daunorubicin hydrochloride, doxorubicin hydrochloride, acralubicin hydrochloride, pyrarubicin hydrochloride, and epirubicin hydrochloride. , Neocartinostatin, myslamycin, zarcomycin, cartinophylline, mittane, zorubicin hydrochloride, mitoxantrone hydrochloride, idarubicin hydrochloride, and DDS preparations thereof and the like are used.

「植物由来抗癌剤」としては、例えば、エトポシド、リン酸エトポシド、硫酸ビンブラスチン、硫酸ビンクリスチン、硫酸ビンデシン、テニポシド、パクリタキセル、ドセタクセル、ビノレルビン、およびそれらのDDS製剤等が用いられる。 As the "plant-derived anticancer agent", for example, etoposide, etoposide phosphate, vinblastine sulfate, vincristine sulfate, vindesine sulfate, teniposide, paclitaxel, docetaxel, vinorelbine, and DDS preparations thereof and the like are used.

「ホルモン療法剤」としては、例えば、ホスフェストロール、ジエチルスチルベストロール、クロロトリアニセン、酢酸メドロキシプロゲステロン、酢酸メゲストロール、酢酸クロルマジノン、酢酸シプロテロン、ダナゾール、アリルエストレノール、ゲストリノン、メパルトリシン、ラロキシフェン、オルメロキシフェン、レボルメロキシフェン、抗エストロゲン(例、クエン酸タモキシフェン、クエン酸トレミフェン)、ピル製剤、メピチオスタン、テストロラクトン、アミノグルテチイミド、LH−RHアゴニスト(例、酢酸ゴセレリン、ブセレリン、リュープロレリン)、ドロロキシフェン、エピチオスタノール、スルホン酸エチニルエストラジオール、アロマターゼ阻害薬(例、塩酸ファドロゾール、アナストロゾール、レトロゾール、エキセメスタン、ボロゾール、フォルメスタン)、抗アンドロゲン(例、フルタミド、ビカルタミド、ニルタミド)、5α−レダクターゼ阻害薬(例、フィナステリド、エプリステリド)、副腎皮質ホルモン系薬剤(例、デキサメタゾン、プレドニゾロン、ベタメタゾン、トリアムシノロン)、アンドロゲン合成阻害薬(例、アビラテロン)、レチノイドおよびレチノイドの代謝を遅らせる薬剤(例、リアロゾール)等が用いられる。 Examples of the "hormone therapeutic agent" include phosfestol, diethylstilbestrol, chlorotrianisen, medroxyprogesterone acetate, megestrol acetate, chlormaginone acetate, ciproterone acetate, dannazole, allylestradiol, guestlinone, mepartricin, and the like. Laroxyphene, olmeroxyphene, revolmeroxyphene, anti-estrogen (eg tamoxifen citrate, tremiphen citrate), pills, mepitiostane, testrolactone, aminoglutetiimide, LH-RH agonists (eg goseleline acetate, busererin) , Leuprorelin), droroxyfen, epithiostanol, ethinyl estradiol sulfonate, aromatase inhibitors (eg, fadrosole hydrochloride, anastrosol, retrosol, exemethan, borozol, formestan), antiandrogens (eg, flutamide, Bicalutamide, niltamide), 5α-reductase inhibitors (eg, finasteride, epristeride), corticohormonal agents (eg, dexamethasone, prednisolone, betamethasone, triamsinolone), androgen synthesis inhibitors (eg, avirateron), retinoid and retinoid metabolism Drugs that delay the disease (eg, riarozole) are used.

「免疫療法剤」としては、生物反応修飾物質(例えば、ピシバニール、クレスチン、シゾフィラン、レンチナン、ウベニメクス、インターフェロン、インターロイキン、マクロファージコロニー刺激因子、顆粒球コロニー刺激因子、エリスロポイエチン、リンホトキシン、BCGワクチン、コリネバクテリウムパルブム、レバミゾール、ポリサッカライドK、プロコダゾール、抗CTLA4抗体)等が用いられる。 "Immunotherapeutic agents" include biological reaction modifiers (eg, schizophyllan, krestin, schizophyllan, lentinan, ubenimex, interferon, interleukin, macrophage colony stimulating factor, granulocyte colony stimulating factor, erythropoietin, phosphotoxin, BCG vaccine, etc. Corinebacterium parbum, levamisol, polysaccharide K, procodazole, anti-CTLA4 antibody) and the like are used.

「分子標的薬」としては、例えば、トシツモマブ、イブリツモマブ、アレムツズマブ、アキシチニブ、ベバシズマブ、アファチニブ、オシメルチニブ、ボルテゾミブ、ボスチニブ、カルフィルゾミブ、セツキシマブ、ダサチニブ、デノスマブ、エドレコロマブ、エルロチニブ、エベロリムス、ビスモデギブ、ゲフィチニブ、ゲムツズマブオゾガマイシン、イマチニブ、イピリムマブ、ラパチニブ、レナリドミド、ニロチニブ、ニモツズマブ、オラパリブ、パニツムマブ、パゾパニブ、ペルツズマブ、リツキシマブ、オファツムマブ、モガムリズマブ、ブレンツキシマブベドチン、ラムシルマブ、エロツズマブ、シルツキシマブ、ソラフェニブ、スニチニブ、タミバロテン、テムシロリムス、サリドマイド、トラスツズマブ、トラスツズマブエムタンシン、トレチノイン、バンデタニブ、ボリノスタット、カボザンチニブ、トラメチニブ、ダブラフェニブ、クリゾチニブ、アレクチニブ、セリチニブ、ルキソリチニブ、イブルチニブ、パルボシクリブ、リボシクリブ、アベマシクリブ、レゴラフェニブ、ピララリシブ、レンバチニブ、ベムラフェニブ等が用いられる。 "Molecular-targeted drugs" include, for example, trastuzumab, ibritsumomab, alemtuzumab, axitinib, bebashizumab, afatinib, osimertinib, voltezomib, bostinib, calfilzomib, cetuximab, dasatinib, denosumab, edretimib Ozogamycin, imatinib, ipilimumab, lapatinib, renalidemid, nirotinib, nimotuzumab, olaparib, panitzumumab, pazopanib, pertuzumab, rituximab, ofatumumab, mogamurizumab, ofatumumab, mogamurizumab, brentsumumab Salidamide, trastuzumab, trastuzumab emtansine, tretinoin, bandetanib, bolinostat, cabozantinib, tramethinib, dabrafenib, crizotinib, alektinib, cetuximab, luxoritinib, seritinib, luxoritinib, seritinib, luxoritinib, ibrutinib, parvosirib

「免疫チェックポイント阻害剤」としては、例えば、イピリムマブ、トレメリムマブ、ニボルマブ、ペムブロリズマブ、アベルマブ、アテゾリズマブ、デュルバルマブ等が用いられる。 As the "immune checkpoint inhibitor", for example, ipilimumab, tremelimumab, nivolumab, pembrolizumab, avelumab, atezolizumab, durvalumab and the like are used.

以下に実施例を用いて本発明を詳述するが、本発明は以下の実施例に何ら限定されるものではない。また、使用する試薬及び材料は特に限定されない限り商業的に入手可能である。 The present invention will be described in detail with reference to the following examples, but the present invention is not limited to the following examples. In addition, the reagents and materials used are commercially available unless otherwise specified.

実施例1 Example 1

(1a) 1-(4,5-ジメトキシ-2-ニトロフェニル)プロパン-2-オン (1a) 1- (4,5-dimethoxy-2-nitrophenyl) propan-2-one

Figure 2020071550
Figure 2020071550

1-(4,5-ジメトキシフェニル)プロパン-2-オン1.00 g (5.2 mmol) をクロロホルム 10 mLに溶解し、-5℃で濃硝酸(P=69 %) 0.94 mL (10 mmol) を1 時間かけて滴下し、氷冷下1 時間撹拌した。水30 mLを加え、クロロホルムで抽出後、クロロホルム層を飽和食塩水で洗浄し、乾燥 (Na2SO4) した。減圧下クロロホルムを留去し、標題化合物の黄色粉末 1.22 g (収率:99%) を得た。
1H-NMR (CDCl3, 400 MHz) δ: 2.34 (3H, s), 3.95 (3H, s), 3.96 (3H, s), 4.08 (2H, s), 6.64 (1H, s), 7.74 (1H, s).
Dissolve 1.00 g (5.2 mmol) of 1- (4,5-dimethoxyphenyl) propan-2-one in 10 mL of chloroform and add 0.94 mL (10 mmol) of concentrated nitrate (P = 69%) at -5 ° C for 1 hour. The mixture was added dropwise, and the mixture was stirred under ice-cooling for 1 hour. After adding 30 mL of water and extracting with chloroform, the chloroform layer was washed with saturated brine and dried (Na 2 SO 4 ). Chloroform was distilled off under reduced pressure to obtain 1.22 g (yield: 99%) of a yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.34 (3H, s), 3.95 (3H, s), 3.96 (3H, s), 4.08 (2H, s), 6.64 (1H, s), 7.74 ( 1H, s).

(1b) 5,6-ジメトキシ-2-メチル-1H-インドール (1b) 5,6-dimethoxy-2-methyl-1H-indole

Figure 2020071550
Figure 2020071550

10 % Pd-C 100 mgをベンゼン 10 mLに懸濁し、実施例1 (1a) で製造した化合物 1.22 g (5.10 mmol) を加え、0.3 MPa、室温にて16.5時間接触水素添加した。Pd-Cをろ別後、減圧下ベンゼンを留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→3:1、V/V)にて精製し、標題化合物の赤褐色粉末 300 mg (収率:31%) を得た。
1H-NMR (CDCl3, 400 MHz) δ: 2.40 (3H, s), 3.89 (3H, s), 3.90 (3H, s), 6.09-6.10 (1H, m), 6.82 (1H, s), 6.98 (1H, s), 7.60-7.75 (1H, br).
100 mg of 10% Pd-C was suspended in 10 mL of benzene, 1.22 g (5.10 mmol) of the compound prepared in Example 1 (1a) was added, and catalytic hydrogenation was carried out at 0.3 MPa at room temperature for 16.5 hours. After filtering Pd-C, benzene was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 3: 1, V / V) to obtain 300 mg (yield: 31%) of a reddish brown powder of the title compound. ..
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.40 (3H, s), 3.89 (3H, s), 3.90 (3H, s), 6.09-6.10 (1H, m), 6.82 (1H, s), 6.98 (1H, s), 7.60-7.75 (1H, br).

(1c) 5,6-ジメトキシ-2-メチル-1-[4-(モルホリン-4-イル)フェニル]-1H-インドール (1c) 5,6-dimethoxy-2-methyl-1- [4- (morpholine-4-yl) phenyl] -1H-indole

Figure 2020071550
Figure 2020071550

実施例1 (1b) で製造した化合物 992 mg (5.19 mmol) をトルエン 7 mLに溶解し、4-(4-ヨードフェニル)モルホリン1.50 g (5.19 mmol)、trans-N,N’-ジメチルシクロヘキサン-1,2-ジアミン 0.12 mL (0.78 mmol)、ヨウ化銅 49 mg (0.26 mmol) および粉砕したリン酸三カリウム 2.20 g (10.4 mmol) を加え、窒素を 10分間バブリング後、窒素気流下110℃で13.5時間撹拌した。水50 mLを加え、水層を酢酸エチル 50 mLで抽出し、有機層を飽和食塩水で洗浄し、乾燥 (Na2SO4) 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(n-ヘキサン:酢酸エチル、10:1→5:1→3:1、V/V)にて精製し、標題化合物の微黄白色粉末 1.07 g (収率:58%) を得た。
1H-NMR (CDCl3, 400 MHz) δ: 2.23 (3H, s), 3.25-3.27 (4H, m), 3.78 (3H, s), 3.88-3.95 (7H, m), 6.24 (1H, s), 7.01-7.04 (3H, m), 7.22-7.24 (2H, m).
Compound 992 mg (5.19 mmol) prepared in Example 1 (1b) was dissolved in 7 mL of toluene, 4- (4-iodophenyl) morpholine 1.50 g (5.19 mmol), trans-N, N'-dimethylcyclohexane-. Add 0.12 mL (0.78 mmol) of 1,2-diamine, 49 mg (0.26 mmol) of copper iodide and 2.20 g (10.4 mmol) of ground tripotassium phosphate, bubbling nitrogen for 10 minutes, and then at 110 ° C. under a nitrogen stream. Stirred for 13.5 hours. 50 mL of water was added, the aqueous layer was extracted with 50 mL of ethyl acetate, the organic layer was washed with saturated brine and dried (Na 2 SO 4 ). The solvent was distilled off under reduced pressure, and the obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 5: 1 → 3: 1, V / V), and the title compound was slightly yellowed. A white powder of 1.07 g (yield: 58%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.23 (3H, s), 3.25-3.27 (4H, m), 3.78 (3H, s), 3.88-3.95 (7H, m), 6.24 (1H, s) ), 7.01-7.04 (3H, m), 7.22-7.24 (2H, m).

(1) 3-アセチル-5,6-ジメトキシ-2-メチル-1-[4-(モルホリン-4-イル)フェニル]-1H-インドール (1) 3-Acetyl-5,6-dimethoxy-2-methyl-1- [4- (morpholine-4-yl) phenyl] -1H-indole

Figure 2020071550
Figure 2020071550

実施例1 (1c) で製造した化合物 1.07 g (3.04 mmol) を塩化メチレン 10 mLに溶解させ、氷冷下塩化アセチル 0.32 mL (4. 6 mmol) および塩化チタン(IV) 0.50 mL (4.6 mmol) を順次滴下し、同温にて1時間撹拌した。水20 mL、酢酸エチル 20 mLを加え、30分間撹拌後、析出物をろ取し、水および酢酸エチルにて順次洗浄した。得られた粉末を塩化メチレン 25 mLに加熱溶解し、酢酸エチル 20 mLを加え、室温にて1時間撹拌した。析出物をろ取し、塩化メチレン-酢酸エチル (1:3) にて洗浄し、標題化合物の白色結晶性粉末 435 mg (収率:36%) を得た。
IR ν (ATR) cm-1:1618, 1514, 1120, 869.
1H-NMR (CDCl3, 400 MHz) δ:2.52 (3H, s), 2.65 (3H, s), 3.28-3.30 (4H, m), 3.77 (3H, s), 3.90-3.93 (7H, m), 3.98 (3H, s), 6.47 (1H, s), 7.05 (2H, m), 7.20 (2H, m), 7.69 (1H, s).
MS m/z: 395 [M+H]+, 417 [M+Na]+.
1.07 g (3.04 mmol) of the compound prepared in Example 1 (1c) was dissolved in 10 mL of methylene chloride, and under ice-cooling, 0.32 mL (4.6 mmol) of acetyl chloride and 0.50 mL (4.6 mmol) of titanium chloride (IV) were dissolved. Was sequentially added dropwise, and the mixture was stirred at the same temperature for 1 hour. 20 mL of water and 20 mL of ethyl acetate were added, and the mixture was stirred for 30 minutes, the precipitate was collected by filtration, and washed successively with water and ethyl acetate. The obtained powder was dissolved by heating in 25 mL of methylene chloride, 20 mL of ethyl acetate was added, and the mixture was stirred at room temperature for 1 hour. The precipitate was collected by filtration and washed with methylene chloride-ethyl acetate (1: 3) to give 435 mg (yield: 36%) of white crystalline powder of the title compound.
IR ν (ATR) cm -1 : 1618, 1514, 1120, 869.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.52 (3H, s), 2.65 (3H, s), 3.28-3.30 (4H, m), 3.77 (3H, s), 3.90-3.93 (7H, m) ), 3.98 (3H, s), 6.47 (1H, s), 7.05 (2H, m), 7.20 (2H, m), 7.69 (1H, s).
MS m / z: 395 [M + H] + , 417 [M + Na] + .

実施例2 Example 2

(2a) エチル 1-フェニルピペリジン-4-カルボキシレート (2a) Ethyl 1-phenylpiperidine-4-carboxylate

Figure 2020071550
Figure 2020071550

ヨードベンゼン 20.4 g (100 mmol)、エチル ピペリジン-4-カルボキシレート 23 mL (0.15 mol)、ヨウ化銅 1.90 g (9.98 mmol)、2-アセチルシクロヘキサノン 2.6 mL (20 mmol) および炭酸セシウム 48.9 g (150 mmol) をN,N-ジメチルホルムアミド 50 mLに懸濁させ、60 ℃にて37時間撹拌した。放冷後、氷水 500 mLに注加し、ジイソプロピルエーテル 200 mLを加え、不溶物をろ別した。二層を分離し、水層をジイソプロピルエーテル 200 mLにて抽出し、ジイソプロピルエーテル層を合わせ、水 200 mLにて洗浄し、減圧下ジイソプロピルエーテルを留去した。残渣にジイソプロピルエーテル 50 mLを加え、氷冷下1 M塩酸200 mLを加え、不溶物をろ別した。二層を分離し、ジイソプロピルエーテル層を1M塩酸 40 mLにて抽出し、水層を合わせ、重曹 20 gにて中和した。ジイソプロピルエーテル 200 mLおよび100 mLにて2回抽出し、ジイソプロピルエーテル層を合わせ、飽和食塩水にて洗浄、乾燥 (Na2SO4) した。減圧下ジイソプロピルエーテルを留去し、標題化合物の赤褐色オイル 17.1 g (収率:73%) を得た。
1H-NMR (CDCl3, 400 MHz) δ :1.27 (3H, t, J=7.1 Hz), 1.82-1.98 (2H, m), 1.99-2.09 (2H, m), 2.39-2.50 (1H, m), 2.79 (2H, td, J=12.4, 2.7 Hz), 3.65 (2H, dt, J=12.4, 3.4 Hz), 4.16 (2H, q, J=7.1 Hz), 6.82-6.87 (1H, m), 6.92-6.97 (2H, m), 7.21-7.28 (2H, m).
Iodobenzene 20.4 g (100 mmol), ethyl piperidine-4-carboxylate 23 mL (0.15 mol), copper iodide 1.90 g (9.98 mmol), 2-acetylcyclohexanone 2.6 mL (20 mmol) and cesium carbonate 48.9 g (150) mmol) was suspended in 50 mL of N, N-dimethylformamide and stirred at 60 ° C. for 37 hours. After allowing to cool, the mixture was poured into 500 mL of ice water, 200 mL of diisopropyl ether was added, and the insoluble material was filtered off. The two layers were separated, the aqueous layer was extracted with 200 mL of diisopropyl ether, the diisopropyl ether layers were combined, washed with 200 mL of water, and diisopropyl ether was distilled off under reduced pressure. 50 mL of diisopropyl ether was added to the residue, 200 mL of 1 M hydrochloric acid was added under ice-cooling, and the insoluble material was filtered off. The two layers were separated, the diisopropyl ether layer was extracted with 40 mL of 1M hydrochloric acid, the aqueous layers were combined, and the mixture was neutralized with 20 g of baking soda. Extraction was performed twice with 200 mL and 100 mL of diisopropyl ether, the diisopropyl ether layers were combined, washed with saturated brine, and dried (Na 2 SO 4 ). Diisopropyl ether was distilled off under reduced pressure to obtain 17.1 g (yield: 73%) of reddish brown oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.27 (3H, t, J = 7.1 Hz), 1.82-1.98 (2H, m), 1.99-2.09 (2H, m), 2.39-2.50 (1H, m) ), 2.79 (2H, td, J = 12.4, 2.7 Hz), 3.65 (2H, dt, J = 12.4, 3.4 Hz), 4.16 (2H, q, J = 7.1 Hz), 6.82-6.87 (1H, m) , 6.92-6.97 (2H, m), 7.21-7.28 (2H, m).

(2b) エチル 1-(4-ヨードフェニル)ピペリジン-4-カルボキシレート (2b) Ethyl 1- (4-iodophenyl) piperidine-4-carboxylate

Figure 2020071550
Figure 2020071550

実施例2 (2a) で製造した化合物 10.4 g (44.6 mmol) を塩化メチレン 100 mLに溶解し、水 40 mLおよび重曹 5.6 g (67 mmol) を加えた後、室温にてヨウ素 13.6 g (53.6 mmol) を1時間かけて分割添加し、さらに同温にて1時間撹拌した。塩化メチレン層を分離し、5 %チオ硫酸ナトリウム水、水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下塩化メチレンを留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、20:1→10:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の黄色粉末 13.6 g (収率:35%) を得た。
1H-NMR (CDCl3, 400 MHz)δ:1.27 (3H, t, J=7.2 Hz), 1.80-1.94 (2H, m), 1.94-2.09 (2H, m), 2.39-2.50 (1H, m), 2.73-2.88 (2H, m), 3.65 (2H, dt, J=12.9, 3.6 Hz), 4.15 (2H, q, J=7.2 Hz), 6.63-6.75 (2H, m), 7.46-7.54 (2H, m).
10.4 g (44.6 mmol) of the compound prepared in Example 2 (2a) was dissolved in 100 mL of methylene chloride, 40 mL of water and 5.6 g (67 mmol) of baking soda were added, and then 13.6 g (53.6 mmol) of iodine was added at room temperature. ) Was added in portions over 1 hour, and the mixture was further stirred at the same temperature for 1 hour. The methylene chloride layer was separated, washed sequentially with 5% sodium thiosulfate water, water, and saturated brine, dried (Na 2 SO 4 ), and then methylene chloride was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 20: 1 → 10: 1, V / V), and the solvent of the target fraction was distilled off under reduced pressure to obtain a yellow powder of the title compound. 13.6 g (yield: 35%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.27 (3H, t, J = 7.2 Hz), 1.80-1.94 (2H, m), 1.94-2.09 (2H, m), 2.39-2.50 (1H, m) ), 2.73-2.88 (2H, m), 3.65 (2H, dt, J = 12.9, 3.6 Hz), 4.15 (2H, q, J = 7.2 Hz), 6.63-6.75 (2H, m), 7.46-7.54 ( 2H, m).

(2c) エチル 1-[4-(5-フルオロ-6-メトキシ-2-メチルインドール-1-イル)フェニル]ピペリジン-4-カルボキシレート (2c) Ethyl 1- [4- (5-fluoro-6-methoxy-2-methylindole-1-yl) phenyl] piperidine-4-carboxylate

Figure 2020071550
Figure 2020071550

実施例2 (2b) で製造した化合物 837 mg (2.33 mmol)、5-フルオロ-6-メトキシ-2-メチルインドール347 mg (1.94 mmol)、ヨウ化銅 18 mg (0.095 mmol)、trans-N,N’-ジメチルシクロヘキサン-1,2-ジアミン 0.046 mL (0.29 mmol) およびリン酸三カリウム 906 mg (4.27 mmol) をトルエン 2 mLに懸濁させ、窒素気流下、80 ℃にて12時間撹拌した。放冷後、水20 mLおよび酢酸エチル 30 mLを加え、不溶物をろ別し、二層を分離した。酢酸エチル層を水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) し、減圧下酢酸エチルを留去した。残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→6:1、V/V)にて精製後、目的分画の溶媒を減圧下留去した。残渣にジイソプロピルエーテル 1 mLを加え撹拌後、n-ヘキサン 10 mLを加え、析出物をろ取し、標題化合物の微黄色粉末 273 mg (収率:34%) を得た。
1H-NMR (CDCl3, 400 MHz)δ:1.29 (3H, t, J=7.2 Hz), 1.86-2.00 (2H, m), 2.04-2.14 (2H, m), 2.22 (3H, s), 2.45-2.55 (1H, m), 2.87-2.97 (2H, m), 3.70-3.81 (2H, m), 3.79 (3H, s), 4.18 (2H, q, J=7.2 Hz), 6.24 (1H, s), 6.59 (1H, d, J=7.1 Hz), 7.02-7.08 (2H, m), 7.17-7.24 (3H, m).
Compound 837 mg (2.33 mmol) prepared in Example 2 (2b), 5-fluoro-6-methoxy-2-methylindole 347 mg (1.94 mmol), copper iodide 18 mg (0.095 mmol), trans-N, 0.046 mL (0.29 mmol) of N'-dimethylcyclohexane-1,2-diamine and 906 mg (4.27 mmol) of tripotassium phosphate were suspended in 2 mL of toluene, and the mixture was stirred at 80 ° C. for 12 hours under a nitrogen stream. After allowing to cool, 20 mL of water and 30 mL of ethyl acetate were added, the insoluble material was filtered off, and the two layers were separated. The ethyl acetate layer was washed successively with water and saturated brine, dried (Na 2 SO 4 ), and ethyl acetate was distilled off under reduced pressure. The residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 6: 1, V / V), and the solvent for the target fraction was distilled off under reduced pressure. After adding 1 mL of diisopropyl ether to the residue and stirring, 10 mL of n-hexane was added, and the precipitate was collected by filtration to obtain 273 mg (yield: 34%) of a slightly yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.29 (3H, t, J = 7.2 Hz), 1.86-2.00 (2H, m), 2.04-2.14 (2H, m), 2.22 (3H, s), 2.45-2.55 (1H, m), 2.87-2.97 (2H, m), 3.70-3.81 (2H, m), 3.79 (3H, s), 4.18 (2H, q, J = 7.2 Hz), 6.24 (1H, m) s), 6.59 (1H, d, J = 7.1 Hz), 7.02-7.08 (2H, m), 7.17-7.24 (3H, m).

(2d) エチル 1-[4-(2-アセチル-5-フルオロ-6-メトキシ-2-メチルインドール-1-イル)フェニル]ピペリジン-4-カルボキシレート (2d) Ethyl 1- [4- (2-acetyl-5-fluoro-6-methoxy-2-methylindole-1-yl) phenyl] piperidine-4-carboxylate

Figure 2020071550
Figure 2020071550

実施例2 (2c) で製造した化合物 265 mg (0.646 mmol) を塩化メチレン 3 mLに溶解させ、氷冷下塩化アセチル 0.070 mL (0.98 mmol) および塩化チタン(IV) 0.11 mL (1.0 mmol) を順次加え、同温にて30分間撹拌した。氷水 20 mLおよび塩化メチレン 20 mLを加え、30分間撹拌後、二層を分離した。塩化メチレン層を水、飽和食塩水にて順次洗浄後、乾燥(Na2SO4) し、減圧下塩化メチレンを留去した。残渣をカラムクロマトグラフィー(トルエン:酢酸エチル、10:1→5:1、V/V)にて精製後、目的分画の溶媒を減圧下留去した。得られた残渣にジエチルエーテル 5 mLを加え撹拌後、ろ取し、標題化合物の白色粉末 174 mg (収率:60%) を得た。
1H-NMR (CDCl3, 400 MHz)δ:1.29 (3H, t, J=7.1 Hz), 1.86-2.00 (2H, m), 2.04-2.14 (2H, m), 2.48-2.58 (1H, m), 2.52 (3H, s), 2.64 (3H, s), 2.90-3.03 (2H, m), 3.75-3.83 (2H, m), 3.79 (3H, s), 4.18 (2H, q, J=7.1 Hz), 6.53 (1H, d, J=7.3 Hz), 7.03-7.09 (2H, m), 7.12-7.18 (2H, m), 7.79 (1H, d, J=12.2 Hz).
Compound 265 mg (0.646 mmol) prepared in Example 2 (2c) was dissolved in 3 mL of methylene chloride, and 0.070 mL (0.98 mmol) of acetyl chloride and 0.11 mL (1.0 mmol) of titanium (IV) chloride were sequentially added under ice-cooling. In addition, the mixture was stirred at the same temperature for 30 minutes. 20 mL of ice water and 20 mL of methylene chloride were added, and the mixture was stirred for 30 minutes, and then the two layers were separated. The methylene chloride layer was washed successively with water and saturated brine, dried (Na 2 SO 4 ), and methylene chloride was distilled off under reduced pressure. The residue was purified by column chromatography (toluene: ethyl acetate, 10: 1 → 5: 1, V / V), and the solvent for the target fraction was evaporated under reduced pressure. 5 mL of diethyl ether was added to the obtained residue, and the mixture was stirred and collected by filtration to give 174 mg (yield: 60%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.29 (3H, t, J = 7.1 Hz), 1.86-2.00 (2H, m), 2.04-2.14 (2H, m), 2.48-2.58 (1H, m) ), 2.52 (3H, s), 2.64 (3H, s), 2.90-3.03 (2H, m), 3.75-3.83 (2H, m), 3.79 (3H, s), 4.18 (2H, q, J = 7.1) Hz), 6.53 (1H, d, J = 7.3 Hz), 7.03-7.09 (2H, m), 7.12-7.18 (2H, m), 7.79 (1H, d, J = 12.2 Hz).

(2) 1-[4-(2-アセチル-5-フルオロ-6-メトキシ-2-メチルインドール-1-イル)フェニル]ピペリジン-4-カルボン酸 (2) 1- [4- (2-Acetyl-5-fluoro-6-methoxy-2-methylindole-1-yl) phenyl] piperidine-4-carboxylic acid

Figure 2020071550
Figure 2020071550

実施例2 (2d) で製造した化合物 166 mg (0.367 mmol) を塩化メチレン-メタノール (1:1) の混液 4 mLに溶解し、室温にて5.0 M 水酸化ナトリウム水 0.37 mL (1.8 mmol) を加え、同温にて13時間撹拌した。減圧下溶媒を留去し、残渣に酢酸エチル 5 mLを加え、水 15 mLにて抽出した。水層に酢酸エチル 5 mLを加え、クエン酸にてpH 4以下に調整後、析出物をろ取し、標題化合物の白色粉末 110 mg (収率:70%) を得た。
IR ν(ATR) cm-1 ; 1724, 1707, 1579, 1510, 1489, 1192, 931, 733.
1H-NMR (DMSO-d6, 400 MHz)δ:1.60-1.73 (2H, m), 1.89-2.00 (2H, m), 2.42-2.50 (1H, m), 2.46 (3H, s), 2.55 (3H, s), 2.83-2.93 (2H, m), 3.71 (3H, s), 3.73-3.83 (2H, m), 6.57 (1H, d, J=7.3 Hz), 7.10-7.17 (2H, m), 7.22-7.28 (2H, m), 7.86 (1H, d, J=12.4 Hz), 12.05-12.40 (1H, br).
MS m/z: 425 [M+H]+, 423 [M-H]-.
Compound 166 mg (0.367 mmol) prepared in Example 2 (2d) was dissolved in 4 mL of a mixture of methylene chloride-methanol (1: 1), and 5.0 M sodium hydroxide water 0.37 mL (1.8 mmol) was added at room temperature. In addition, the mixture was stirred at the same temperature for 13 hours. The solvent was evaporated under reduced pressure, 5 mL of ethyl acetate was added to the residue, and the mixture was extracted with 15 mL of water. After adding 5 mL of ethyl acetate to the aqueous layer and adjusting the pH to 4 or less with citric acid, the precipitate was collected by filtration to obtain 110 mg (yield: 70%) of a white powder of the title compound.
IR ν (ATR) cm -1 ; 1724, 1707, 1579, 1510, 1489, 1192, 931, 733.
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 1.60-1.73 (2H, m), 1.89-2.00 (2H, m), 2.42-2.50 (1H, m), 2.46 (3H, s), 2.55 ( 3H, s), 2.83-2.93 (2H, m), 3.71 (3H, s), 3.73-3.83 (2H, m), 6.57 (1H, d, J = 7.3 Hz), 7.10-7.17 (2H, m) , 7.22-7.28 (2H, m), 7.86 (1H, d, J = 12.4 Hz), 12.05-12.40 (1H, br).
MS m / z: 425 [M + H] + , 423 [MH] - .

実施例3 Example 3

(3a) 8-フェニル-1,4-ジオキサ-8-アザスピロ[4,5]デカン (3a) 8-Phenyl-1,4-dioxa-8-azaspiro [4,5] decane

Figure 2020071550
Figure 2020071550

トリス(2-メチルフェニル)ホスフィン 3.80 g (12.5 mmol) および塩化パラジウム(II) 1.11 g (6.24 mmol) をトルエン 50 mLに懸濁し、窒素気流下30分間撹拌した。トルエン 650 mL、1,4-ジオキサ-8-アザスピロ[4.5]デカン 22.3 g (156 mmol)、ナトリウム tert-ブトキシド 22.5 g (234 mmol) およびブロモベンゼン 19.7 mL (187 mmol) を順次加え、窒素気流下100 ℃にて20時間撹拌した。放冷後、飽和食塩水650 mL、酢酸エチル 300 mLを加え、不溶物をろ別した。酢酸エチル層を分離し、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→5:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡橙色オイル 33.3 g (収率:98%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.80-1.88 (4H, m), 3.28-3.36 (4H, m), 3.99 (4H, s), 6.79-6.87 (1H, m), 6.91-6.98 (2H, m), 7.20-7.29 (2H, m).
3.80 g (12.5 mmol) of tris (2-methylphenyl) phosphine and 1.11 g (6.24 mmol) of palladium (II) chloride were suspended in 50 mL of toluene and stirred for 30 minutes under a nitrogen stream. Toluene 650 mL, 1,4-dioxa-8-azaspiro [4.5] decane 22.3 g (156 mmol), sodium tert-butoxide 22.5 g (234 mmol) and bromobenzene 19.7 mL (187 mmol) were added sequentially under a nitrogen stream. The mixture was stirred at 100 ° C. for 20 hours. After allowing to cool, 650 mL of saturated brine and 300 mL of ethyl acetate were added, and the insoluble material was filtered off. The ethyl acetate layer was separated, dried (Na 2 SO 4 ), and then the solvent was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 5: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 33.3 g (yield: 98%) of a light orange oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.80-1.88 (4H, m), 3.28-3.36 (4H, m), 3.99 (4H, s), 6.79-6.87 (1H, m), 6.91-6.98 (2H, m), 7.20-7.29 (2H, m).

(3b) 4-[2-(tert-ブチルジメチルシラニルオキシ)エトキシ]-1-フェニルピペリジン (3b) 4- [2- (tert-Butyldimethylsilanyloxy) ethoxy] -1-phenylpiperidine

Figure 2020071550
Figure 2020071550

塩化アルミニウム 17.6 g (132 mmol) をジエチルエーテル 200 mLに溶解し、-78 ℃にて30分間撹拌した。水素化アルミニウムリチウム 1.26 g (33.1 mmol) のジエチルエーテル 33 mL懸濁液を加え、同温にて30分間撹拌した。実施例3 (3a) で製造した化合物 14.5 g (66.1 mmol) のジエチルエーテル 66 mL溶液を滴下し、室温にて2時間撹拌した。氷冷下反応液に水200 mLを滴下し、水酸化ナトリウム 7.44 gを加え、ジエチルエーテルにて抽出した。ジエチルエーテル層を飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下ジエチルエーテルを留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、2:1→1:1→1:2、V/V)にて精製した。目的分画の溶媒を減圧下留去し、2-(1-フェニルピペリジン-4-イルオキシ)エタノールの褐色オイル15.3 gを得た。
2-(1-フェニルピペリジン-4-イルオキシ)エタノール 15.3 gをN,N-ジメチルホルムアミド 75 mLに溶解し、イミダゾール 8.99 g (132 mmol)、tert-ブチルジメチルクロロシラン 9.96 g (66.1 mmol) を加え、室温にて2.5時間撹拌した。反応液に水 300 mLを加え、ジエチルエーテル 300 mLにて抽出した。ジエチルエーテル層を水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下ジエチルエーテルを留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、20:1→10:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡橙色オイル 17.7 g (収率:80%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.08 (6H, s), 0.90 (9H, s), 1.66-1.78 (2H, m), 1.94-2.06 (2H, m), 2.87-2.97 (2H, m), 3.46-3.56 (3H, m), 3.56 (2H, t, J=5.6 Hz), 3.76 (2H, t, J=5.6 Hz), 6.78-6.87 (1H, m), 6.90-6.98 (2H, m), 7.20-7.28 (2H, m).
17.6 g (132 mmol) of aluminum chloride was dissolved in 200 mL of diethyl ether, and the mixture was stirred at −78 ° C. for 30 minutes. A 33 mL suspension of diethyl ether of 1.26 g (33.1 mmol) of lithium aluminum hydride was added, and the mixture was stirred at the same temperature for 30 minutes. A 66 mL solution of 14.5 g (66.1 mmol) of the compound prepared in Example 3 (3a) was added dropwise, and the mixture was stirred at room temperature for 2 hours. 200 mL of water was added dropwise to the reaction solution under ice-cooling, 7.44 g of sodium hydroxide was added, and the mixture was extracted with diethyl ether. The diethyl ether layer was washed with saturated brine and dried (Na 2 SO 4 ), and then diethyl ether was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 2: 1 → 1: 1 → 1: 2, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 15.3 g of a brown oil of 2- (1-phenylpiperidine-4-yloxy) ethanol.
Dissolve 15.3 g of 2- (1-phenylpiperidine-4-yloxy) ethanol in 75 mL of N, N-dimethylformamide, add 8.99 g (132 mmol) of imidazole and 9.96 g (66.1 mmol) of tert-butyldimethylchlorosilane. The mixture was stirred at room temperature for 2.5 hours. 300 mL of water was added to the reaction mixture, and the mixture was extracted with 300 mL of diethyl ether. The diethyl ether layer was washed successively with water and saturated brine, dried (Na 2 SO 4 ), and then diethyl ether was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 20: 1 → 10: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 17.7 g (yield: 80%) of a light orange oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.08 (6H, s), 0.90 (9H, s), 1.66-1.78 (2H, m), 1.94-2.06 (2H, m), 2.87-2.97 (2H) , m), 3.46-3.56 (3H, m), 3.56 (2H, t, J = 5.6 Hz), 3.76 (2H, t, J = 5.6 Hz), 6.78-6.87 (1H, m), 6.90-6.98 ( 2H, m), 7.20-7.28 (2H, m).

(3c) 4-[2-(tert-ブチルジメチルシラニルオキシ)エトキシ]-1-(4-ヨードフェニル)ピペリジン (3c) 4- [2- (tert-butyldimethylsilanyloxy) ethoxy] -1- (4-iodophenyl) piperidine

Figure 2020071550
Figure 2020071550

実施例3 (3b) で製造した化合物 17.7 g (52.7 mmol) を塩化メチレン-水 (4:3) 350 mLに溶解後、重曹 6.65 g (79.1 mmol) を加え、氷冷下ヨウ素 13.4 g (52.7 mmol) を分割添加し、室温にて2.5時間撹拌した。さらに氷冷下ヨウ素 2.68 g (10.5 mmol) を分割添加し、室温にて2時間撹拌した。反応液に水200 mLを加え、塩化メチレン 200 mLにて抽出した。塩化メチレン層を5 %チオ硫酸ナトリウム水、水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下塩化メチレンを留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、20:1→10:1、V/V)にて精製し、目的分画の溶媒を減圧下留去した。得られた残渣を再度カラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡桃色粉末 22.4 g (収率:92%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.07 (6H, s), 0.90 (9H, s), 1.64-1.76 (2H, m), 1.90-2.02 (2H, m), 2.87-2.97 (2H, m), 3.41-3.54 (3H, m), 3.55 (2H, t, J=5.4 Hz), 3.76 (2H, t, J=5.4 Hz), 6.65-6.72 (2H, m), 7.44-7.51 (2H, m).
After dissolving 17.7 g (52.7 mmol) of the compound prepared in Example 3 (3b) in 350 mL of methylene chloride-water (4: 3), 6.65 g (79.1 mmol) of baking soda was added, and 13.4 g (52.7) of iodine under ice cooling was added. mmol) was added in portions, and the mixture was stirred at room temperature for 2.5 hours. Further, 2.68 g (10.5 mmol) of iodine under ice-cooling was added in portions, and the mixture was stirred at room temperature for 2 hours. 200 mL of water was added to the reaction mixture, and the mixture was extracted with 200 mL of methylene chloride. The methylene chloride layer was washed successively with 5% sodium thiosulfate, water and saturated brine, dried (Na 2 SO 4 ), and then methylene chloride was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 20: 1 → 10: 1, V / V), and the solvent for the target fraction was evaporated under reduced pressure. The obtained residue was purified again by column chromatography (n-hexane: ethyl acetate, 10: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 22.4 g (yield: 92%) of a pale pink powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.07 (6H, s), 0.90 (9H, s), 1.64-1.76 (2H, m), 1.90-2.02 (2H, m), 2.87-2.97 (2H) , m), 3.41-3.54 (3H, m), 3.55 (2H, t, J = 5.4 Hz), 3.76 (2H, t, J = 5.4 Hz), 6.65-6.72 (2H, m), 7.44-7.51 ( 2H, m).

(3d) 1-(4-[4-(2-tert-ブチルジメチルシラニルオキシ)エトキシ]ピペリジン-1-イル}フェニル)-5,6-ジメトキシ-2-メチルインドール (3d) 1-(4- [4- (2-tert-butyldimethylsilanyloxy) ethoxy] piperidine-1-yl} phenyl) -5,6-dimethoxy-2-methylindole

Figure 2020071550
Figure 2020071550

実施例3 (3c) で製造した化合物 22.4 g (48.5 mmol)、実施例1 (1b) で製造した化合物 10.2 g (53.4 mmol)、ヨウ化銅 509 mg (2.67 mmol)、trans-N,N’-ジメチルシクロヘキサン-1,2-ジアミン 1.26 mL (8.01 mmol) およびリン酸三カリウム 23.8 g (112 mmol) をトルエン 70 mLに懸濁し、室温にて10分間窒素をバブリングさせ、窒素気流下110 ℃にて22時間撹拌した。放冷後、反応液に水 150 mLおよび酢酸エチル 150 mLを加え、不溶物をろ別し、二層を分離した。酢酸エチル層を飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、5:1→4:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 13.8 g (収率:54%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.09 (6H, s), 0.91 (9H, s), 1.71-1.83 (2H, m), 1.97-2.11 (2H, m), 2.22 (3H, s), 2.99-3.10 (2H, m), 3.53-3.66 (5H, m), 3.75-3.82 (2H, m), 3.78 (3H, s), 3.91 (3H, s), 6.23 (1H, s), 6.57 (1H, s), 7.00-7.07 (2H, m), 7.01 (1H, s), 7.15-7.22 (2H, m).
Compound 22.4 g (48.5 mmol) prepared in Example 3 (3c), compound 10.2 g (53.4 mmol) prepared in Example 1 (1b), copper iodide 509 mg (2.67 mmol), trans-N, N'. -Suspended 1.26 mL (8.01 mmol) of dimethylcyclohexane-1,2-diamine and 23.8 g (112 mmol) of tripotassium phosphate in 70 mL of toluene, bubbling nitrogen for 10 minutes at room temperature, and bringing to 110 ° C. under a nitrogen stream. Stirred for 22 hours. After allowing to cool, 150 mL of water and 150 mL of ethyl acetate were added to the reaction solution, the insoluble material was filtered off, and the two layers were separated. The ethyl acetate layer was washed successively with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 5: 1 → 4: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 13.8 g (yield: 54%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.09 (6H, s), 0.91 (9H, s), 1.71-1.83 (2H, m), 1.97-2.11 (2H, m), 2.22 (3H, s) ), 2.99-3.10 (2H, m), 3.53-3.66 (5H, m), 3.75-3.82 (2H, m), 3.78 (3H, s), 3.91 (3H, s), 6.23 (1H, s), 6.57 (1H, s), 7.00-7.07 (2H, m), 7.01 (1H, s), 7.15-7.22 (2H, m).

(3e) 3-アセチル-1-(4-[4-(2-tert-ブチルジメチルシラニルオキシ)エトキシ]ピペリジン-1-イル}フェニル)-5,6-ジメトキシ-2-メチルインドール (3e) 3-Acetyl-1-(4- [4- (2-tert-butyldimethylsilanyloxy) ethoxy] piperidine-1-yl} phenyl) -5,6-dimethoxy-2-methylindole

Figure 2020071550
Figure 2020071550

実施例3 (3d) で製造した化合物 13.8 g (26.3 mmol) を塩化メチレン 140 mLに溶解し、氷冷下塩化アセチル 2.81 mL (39.5 mmol) および塩化チタン(IV) 4.33 mL (39.5 mmol) を順次加え、同温にて1.5時間撹拌した。反応液に水300 mLを加え、塩化メチレン 200 mLにて抽出した。塩化メチレン層を飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下塩化メチレンを留去した。得られた残渣をカラムクロマトグラフィー(トルエン:酢酸エチル、2:1→1:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡黄色泡状粉末 3.00 g (収率:20%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.91 (9H, s), 1.71-1.84 (2H, m), 1.98-2.11 (2H, m), 2.51 (3H, s), 2.65 (3H, s), 3.03-3.15 (2H, m), 3.54-3.68 (5H, m), 3.77 (3H, s), 3.77-3.83 (2H, m), 3.98 (3H, s), 6.48 (1H, s), 7.02-7.09 (2H, m), 7.11-7.17 (2H, m), 7.69 (1H, s).
13.8 g (26.3 mmol) of the compound prepared in Example 3 (3d) was dissolved in 140 mL of methylene chloride, and 2.81 mL (39.5 mmol) of acetyl chloride and 4.33 mL (39.5 mmol) of titanium (IV) chloride were sequentially added under ice-cooling. In addition, the mixture was stirred at the same temperature for 1.5 hours. 300 mL of water was added to the reaction mixture, and the mixture was extracted with 200 mL of methylene chloride. The methylene chloride layer was washed with saturated brine and dried (Na 2 SO 4 ), and then methylene chloride was distilled off under reduced pressure. The obtained residue was purified by column chromatography (toluene: ethyl acetate, 2: 1 → 1: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 3.00 g (yield: 20%) of a pale yellow foamy powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.91 (9H, s), 1.71-1.84 (2H, m), 1.98-2.11 (2H, m), 2.51 (3H, s), 2.65 (3H, s) ), 3.03-3.15 (2H, m), 3.54-3.68 (5H, m), 3.77 (3H, s), 3.77-3.83 (2H, m), 3.98 (3H, s), 6.48 (1H, s), 7.02-7.09 (2H, m), 7.11-7.17 (2H, m), 7.69 (1H, s).

(3) 3-アセチル-1-{4-[4-(2-ヒドロキシエトキシ)ピペリジン-1-イル]フェニル}-5,6-ジメトキシ-2-メチルインドール (3) 3-Acetyl-1-{4- [4- (2-Hydroxyethoxy) piperidine-1-yl] phenyl} -5,6-dimethoxy-2-methylindole

Figure 2020071550
Figure 2020071550

実施例3 (3e) で製造した化合物 3.00 g (5.29 mmol) をテトラヒドロフラン 30 mLに溶解し、氷冷下1.0 Mフッ化テトラ-n-ブチルアンモニウム テトラヒドロフラン溶液 6.35 mL (6.4 mmol) を加え、同温にて1.5時間撹拌した。反応液に水 100 mLを加え、酢酸エチル 100 mLにて抽出した。酢酸エチル層を飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下酢酸エチルを留去した。得られた残渣をカラムクロマトグラフィー(酢酸エチル)にて精製した。目的分画の溶媒を減圧下留去し、ジイソプロピルエーテルを加え、ろ取し、標題化合物の淡黄色泡状粉末 1.64 g (収率:68%) を得た。
IR ν (ATR) cm-1 ; 3494, 1630, 1610.
1H-NMR (CDCl3, 400 MHz) δ:1.73-1.88 (2H, m), 1.99-2.15 (3H, m), 2.51 (3H, s), 2.65 (3H, s), 3.06-3.18 (2H, m), 3.55-3.70 (5H, m), 3.73-3.83 (2H, m), 3.78 (3H, s), 3.98 (3H, s), 6.48 (1H, s), 7.03-7.09 (2H, m), 7.12-7.18 (2H, m), 7.68 (1H, s).
MS m/z: 453 [M+H]+, 475 [M+Na]+.
3.00 g (5.29 mmol) of the compound prepared in Example 3 (3e) was dissolved in 30 mL of tetrahydrofuran, and 6.35 mL (6.4 mmol) of a 1.0 M tetra-n-butylammonium tetrahydrofuran tetrahydrofuran solution was added under ice-cooling to the same temperature. Was stirred for 1.5 hours. 100 mL of water was added to the reaction mixture, and the mixture was extracted with 100 mL of ethyl acetate. The ethyl acetate layer was washed with saturated brine and dried (Na 2 SO 4 ), and then ethyl acetate was distilled off under reduced pressure. The obtained residue was purified by column chromatography (ethyl acetate). The solvent of the target fraction was distilled off under reduced pressure, diisopropyl ether was added, and the mixture was collected by filtration to obtain 1.64 g (yield: 68%) of a pale yellow foamy powder of the title compound.
IR ν (ATR) cm -1 ; 3494, 1630, 1610.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.73-1.88 (2H, m), 1.99-2.15 (3H, m), 2.51 (3H, s), 2.65 (3H, s), 3.06-3.18 (2H) , m), 3.55-3.70 (5H, m), 3.73-3.83 (2H, m), 3.78 (3H, s), 3.98 (3H, s), 6.48 (1H, s), 7.03-7.09 (2H, m) ), 7.12-7.18 (2H, m), 7.68 (1H, s).
MS m / z: 453 [M + H] + , 475 [M + Na] + .

実施例4 Example 4

(4a) ベンジル 4-(4-ヨードフェニル)ピペラジン-1-カルボキシレート (4a) Benzyl 4- (4-iodophenyl) piperazin-1-carboxylate

Figure 2020071550
Figure 2020071550

1-(4-ヨードフェニル)ピペラジン 2.0 g (6.9 mmol) をN,N-ジメチルホルムアミド 10 mLに溶解し、氷冷下クロロぎ酸ベンジル 1.09 mL (7.63 mmol)、トリエチルアミン 1.16 mL (8.33 mmol) を加え、室温にて1.5時間撹拌した。反応液に水を加え、酢酸エチルにて2回抽出した。酢酸エチル層を5% クエン酸水、水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) した。減圧下酢酸エチルを留去し、得られた残渣にn-ヘキサンを加え、ろ取し、標題化合物の淡褐色粉末 2.36 g (収率:81%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:3.06-3.20 (4H, m), 3.61-3.70 (4H, m), 5.16 (2H, s), 6.66-6.68 (2H, m), 7.30-7.40 (5H, m), 7.51-7.53 (2H, m).
Dissolve 2.0 g (6.9 mmol) of 1- (4-iodophenyl) piperazin in 10 mL of N, N-dimethylformamide, and add 1.09 mL (7.63 mmol) of benzyl chloroformate and 1.16 mL (8.33 mmol) of triethylamine under ice-cooling. In addition, the mixture was stirred at room temperature for 1.5 hours. Water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The ethyl acetate layer was washed successively with 5% citric acid water, water and saturated brine, and dried (Na 2 SO 4 ). Ethyl acetate was distilled off under reduced pressure, n-hexane was added to the obtained residue, and the mixture was collected by filtration to give 2.36 g (yield: 81%) of a light brown powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 3.06-3.20 (4H, m), 3.61-3.70 (4H, m), 5.16 (2H, s), 6.66-6.68 (2H, m), 7.30-7.40 (5H, m), 7.51-7.53 (2H, m).

(4b) ベンジル [4-(5,6-ジメトキシ-2-メチルインドール-1-イル)フェニル]ピペラジン-1-カルボキシレート (4b) Benzyl [4- (5,6-dimethoxy-2-methylindole-1-yl) phenyl] piperazin-1-carboxylate

Figure 2020071550
Figure 2020071550

実施例4 (4a) で製造した化合物 2.36 g (5.59 mmol) をトルエン 10 mLに溶解し、実施例1 (1b) で製造した化合物 1.07 g (5.59 mmol)、ヨウ化銅 54 mg (0.28 mmol)、粉砕したリン酸三カリウム 2.49 g (11.7 mmol)、trans-N,N’-ジメチルシクロヘキサン-1,2-ジアミン 0.13 mL (0.84 mmol) を加え、窒素で10分間バブリング後、窒素雰囲気下110℃で20時間撹拌した。酢酸エチルおよび水を加え、不溶物をセライトにてろ別した。二層を分離し、有機層を飽和食塩水で洗浄後、乾燥 (Na2SO4) 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(酢酸エチル:クロロホルム、1:20、n-ヘキサン:酢酸エチル、1:2、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 1.78 g (収率:66%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.22 (3H, s), 3.20-3.32 (4H, m), 3.68-3.78 (4H, m), 3.77 (3H, s), 3.92 (3H, s), 5.18 (2H, s), 6.24 (1H, s), 6.56 (1H, s), 7.00-7.09 (3H, m), 7.20-7.30 (2H, m), 7.32-7.42 (5H, m).
Compound 2.36 g (5.59 mmol) prepared in Example 4 (4a) was dissolved in 10 mL of toluene, and compound 1.07 g (5.59 mmol) prepared in Example 1 (1b), copper iodide 54 mg (0.28 mmol). Add 2.49 g (11.7 mmol) of crushed tripotassium phosphate and 0.13 mL (0.84 mmol) of trans-N, N'-dimethylcyclohexane-1,2-diamine, bubbling with nitrogen for 10 minutes, and then 110 ° C. under a nitrogen atmosphere. Was stirred for 20 hours. Ethyl acetate and water were added, and the insoluble material was filtered off with Celite. The two layers were separated, and the organic layer was washed with saturated brine and dried (Na 2 SO 4 ). The solvent was evaporated under reduced pressure, and the obtained residue was purified by column chromatography (ethyl acetate: chloroform, 1:20, n-hexane: ethyl acetate, 1: 2, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 1.78 g (yield: 66%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.22 (3H, s), 3.20-3.32 (4H, m), 3.68-3.78 (4H, m), 3.77 (3H, s), 3.92 (3H, s) ), 5.18 (2H, s), 6.24 (1H, s), 6.56 (1H, s), 7.00-7.09 (3H, m), 7.20-7.30 (2H, m), 7.32-7.42 (5H, m).

(4c) ベンジル 4-[3-アセチル-4-(5,6-ジメトキシ-2-メチルインドール-1-イル)フェニル]ピペラジン-1-カルボキシレート (4c) Benzyl 4- [3-acetyl-4- (5,6-dimethoxy-2-methylindole-1-yl) phenyl] piperazin-1-carboxylate

Figure 2020071550
Figure 2020071550

実施例4 (4b) で製造した化合物 1.78 g (3.67 mmol) を塩化メチレン 20 mLに溶解し、氷冷下塩化アセチル 0.39 mL (5.5 mmol)および塩化チタン(IV) 0.61 mL (5.5 mmol) を順次加え、同温にて1時間撹拌した。反応液に水およびクロロホルムを加え、二層を分離し、クロロホルム層を水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下クロロホルムを留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、1:1→1:2、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 580 mg (収率:30%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.51 (3H, s), 2.65 (3H, s), 3.25-3.35 (4H, m), 3.68-3.74 (4H, m), 3.76 (3H, s), 3.98 (3H, s), 5.18 (2H, s), 6.46 (1H, s), 7.03-7.07 (2H, m), 7.17-7.21 (2H, m), 7.32-7.40 (5H, m), 7.68 (1H, s).
1.78 g (3.67 mmol) of the compound prepared in Example 4 (4b) was dissolved in 20 mL of methylene chloride, and 0.39 mL (5.5 mmol) of acetyl chloride and 0.61 mL (5.5 mmol) of titanium (IV) chloride were sequentially added under ice-cooling. In addition, the mixture was stirred at the same temperature for 1 hour. Water and chloroform were added to the reaction solution, the two layers were separated, the chloroform layer was washed successively with water and saturated brine, dried (Na 2 SO 4 ), and then chloroform was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 1: 1 → 1: 2, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 580 mg (yield: 30%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.51 (3H, s), 2.65 (3H, s), 3.25-3.35 (4H, m), 3.68-3.74 (4H, m), 3.76 (3H, s) ), 3.98 (3H, s), 5.18 (2H, s), 6.46 (1H, s), 7.03-7.07 (2H, m), 7.17-7.21 (2H, m), 7.32-7.40 (5H, m), 7.68 (1H, s).

(4d) 1-[3-アセチル-4-(5,6-ジメトキシ-2-メチルインドール-1-イル)フェニル]ピペラジン (4d) 1- [3-Acetyl-4- (5,6-dimethoxy-2-methylindole-1-yl) phenyl] piperazin

Figure 2020071550
Figure 2020071550

10%Pd-C 100 mg を塩化メチレン-メタノール (1:2) 30 mLに懸濁し、実施例4 (4c) で製造した化合物 580 mg (1.10 mmol) を加え、室温、0.40 MPa にて17時間接触水素添加した。Pd-C をろ別後、減圧下溶媒を留去し、得られた残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の淡黄色粉末 170 mg (収率:85%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.51 (3H, s), 2.65 (3H, s), 3.17-3.28 (4H, m), 3.37-3.47 (4H, m), 3.78 (3H, s), 3.98 (3H, s), 6.46 (1H, s), 7.06-7.08 (2H, m), 7.19-7.21 (2H, m), 7.68 (1H, s, indole C4-H).
Suspend 100 mg of 10% Pd-C in 30 mL of methylene chloride-methanol (1: 2), add 580 mg (1.10 mmol) of the compound prepared in Example 4 (4c), and add 580 mg (1.10 mmol) at room temperature at 0.40 MPa for 17 hours. Contact hydrogenation was added. After filtering Pd-C, the solvent was evaporated under reduced pressure, diisopropyl ether was added to the obtained residue, and the mixture was collected by filtration to give 170 mg (yield: 85%) of a pale yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.51 (3H, s), 2.65 (3H, s), 3.17-3.28 (4H, m), 3.37-3.47 (4H, m), 3.78 (3H, s) ), 3.98 (3H, s), 6.46 (1H, s), 7.06-7.08 (2H, m), 7.19-7.21 (2H, m), 7.68 (1H, s, indole C4-H).

(4) 3-アセチル-1-{4-[4-(3-ヒドロキシプロピル)ピペラジン-1-イル]フェニル}-5,6-ジメトキシ-2-メチルインドール (4) 3-Acetyl-1-{4- [4- (3-Hydroxypropyl) piperazine-1-yl] phenyl} -5,6-dimethoxy-2-methylindole

Figure 2020071550
Figure 2020071550

実施例4 (4d) で製造した化合物 555 mg (1.41 mmol) をN,N-ジメチルホルムアミド 5 mLに懸濁し、炭酸カリウム 390 mg (2.82 mmol)および3-ブロモ-1-プロパノール 196 mg (1.41 mmol) を加え、60 ℃にて24時間撹拌した。放冷後、水を加え、酢酸エチルにて抽出し、酢酸エチル層を水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) した。減圧下酢酸エチルを留去し、残渣をカラムクロマトグラフィー(塩化メチレン:メタノール、10:1、V/V)にて精製後、目的分画の溶媒を減圧下留去した。得られた残渣を酢酸エチルに加熱溶解後、ジイソプロピルエーテルを加え、析出物をろ取し、標題化合物の白色粉末 477 mg (収率:74%) を得た。
IR ν (ATR) cm-1 ; 1628, 1516, 1475, 1232, 1134, 1043, 818, 768
1H-NMR (CDCl3, 400 MHz) δ:1.78-1.89 (2H, m), 2.52 (3H, s), 2.66 (3H, s), 2.69-2.78 (6H, m), 3.30-3.42 (4H, m), 3.78 (3H, s), 3.86 (2H, t), 3.99 (3H, s), 6.41 (1H, s), 7.02-7.07 (2H, m), 7.16-7.21 (2H, m), 7.70 (1H, s).
MS m/z: 452 [M+H]+.
Compound 555 mg (1.41 mmol) prepared in Example 4 (4d) was suspended in 5 mL of N, N-dimethylformamide, potassium carbonate 390 mg (2.82 mmol) and 3-bromo-1-propanol 196 mg (1.41 mmol). ) Was added, and the mixture was stirred at 60 ° C. for 24 hours. After allowing to cool, water was added, the mixture was extracted with ethyl acetate, and the ethyl acetate layer was washed successively with water and saturated brine, and dried (Na 2 SO 4 ). Ethyl acetate was distilled off under reduced pressure, the residue was purified by column chromatography (methylene chloride: methanol, 10: 1, V / V), and the solvent for the target fraction was distilled off under reduced pressure. The obtained residue was dissolved by heating in ethyl acetate, diisopropyl ether was added, and the precipitate was collected by filtration to obtain 477 mg (yield: 74%) of a white powder of the title compound.
IR ν (ATR) cm -1 ; 1628, 1516, 1475, 1232, 1134, 1043, 818, 768
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.78-1.89 (2H, m), 2.52 (3H, s), 2.66 (3H, s), 2.69-2.78 (6H, m), 3.30-3.42 (4H) , m), 3.78 (3H, s), 3.86 (2H, t), 3.99 (3H, s), 6.41 (1H, s), 7.02-7.07 (2H, m), 7.16-7.21 (2H, m), 7.70 (1H, s).
MS m / z: 452 [M + H] + .

実施例5 Example 5

(5a) 1-[4-(tert-ブチルジメチルシラニルオキシ)フェニル]-6-メトキシ-2-メチルインドール (5a) 1- [4- (tert-butyldimethylsilanyloxy) phenyl] -6-methoxy-2-methylindole

Figure 2020071550
Figure 2020071550

6-メトキシ-2-メチルインドール 3.85 g (23.9 mmol)、tert-ブチル-(4-ヨードフェノキシ)ジメチルシラン 8.00 g (23.9 mmol)、ヨウ化銅 229 mg (1.20 mmol)、trans-N,N’-ジメチルシクロヘキサン-1,2-ジアミン 0.57 mL (3.6 mmol) およびリン酸三カリウム 10.7 g (50.2 mmol) をトルエン 27 mLに懸濁し、室温にて10分間窒素をバブリングし、窒素雰囲気下110℃ にて18時間撹拌した。放冷後、反応液に水 80 mLを加え、酢酸エチル 80 mLにて抽出した。酢酸エチル層を水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下酢酸エチルを留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、50:1→20:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡黄色粉末 4.38 g (収率:50%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.27 (6H, s), 1.03 (9H, s), 2.22 (3H, s), 3.74 (3H, s), 6.28 (1H, s), 6.54 (1H, d, J=2.2 Hz), 6.75 (1H, dd, J=8.3, 2.2 Hz), 6.93-7.00 (2H, m), 7.13-7.21 (2H, m), 7.41 (1H, d, J=8.3 Hz).
6-methoxy-2-methylindole 3.85 g (23.9 mmol), tert-butyl- (4-iodophenoxy) dimethylsilane 8.00 g (23.9 mmol), copper iodide 229 mg (1.20 mmol), trans-N, N' -Dimethylcyclohexane-1,2-diamine 0.57 mL (3.6 mmol) and tripotassium phosphate 10.7 g (50.2 mmol) were suspended in 27 mL of toluene, nitrogen was bubbled at room temperature for 10 minutes, and the temperature was 110 ° C. under a nitrogen atmosphere. Stirred for 18 hours. After allowing to cool, 80 mL of water was added to the reaction solution, and the mixture was extracted with 80 mL of ethyl acetate. The ethyl acetate layer was washed successively with water and saturated brine, dried (Na 2 SO 4 ), and then ethyl acetate was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 50: 1 → 20: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 4.38 g (yield: 50%) of a pale yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.27 (6H, s), 1.03 (9H, s), 2.22 (3H, s), 3.74 (3H, s), 6.28 (1H, s), 6.54 ( 1H, d, J = 2.2 Hz), 6.75 (1H, dd, J = 8.3, 2.2 Hz), 6.93-7.00 (2H, m), 7.13-7.21 (2H, m), 7.41 (1H, d, J = 8.3 Hz).

(5b) 3-アセチル-1-[4-(tert-ブチルジメチルシラニルオキシ)フェニル]-6-メトキシ-2-メチルインドール (5b) 3-Acetyl-1- [4- (tert-butyldimethylsilanyloxy) phenyl] -6-methoxy-2-methylindole

Figure 2020071550
Figure 2020071550

実施例5 (5a) で製造した化合物 4.36 g (11.9 mmol) を塩化メチレン 44 mLに溶解し、氷冷下塩化アセチル 1.27 mL (17.8 mmol) および塩化チタン(IV) 1.95 mL (17.8 mmol)を順次加え、同温にて5時間撹拌した。反応液を水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下塩化メチレンを留去し、得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、1:10→1:5、V/V)にて精製した。目的分画の溶媒を減圧下留去し、得られた残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の白色粉末 1.30 g (収率:27%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.29 (6H, s), 1.03 (9H, s), 2.53 (3H, s), 2.68 (3H, s), 3.75 (3H, s), 6.47 (1H, d, J=2.4 Hz), 6.91 (1H, dd, J=8.8, 2.4 Hz), 6.97-7.04 (2H, m), 7.12-7.19 (2H, m), 7.93 (1H, d, J=8.8 Hz).
4.36 g (11.9 mmol) of the compound prepared in Example 5 (5a) was dissolved in 44 mL of methylene chloride, and 1.27 mL (17.8 mmol) of acetyl chloride and 1.95 mL (17.8 mmol) of titanium (IV) chloride were sequentially added under ice-cooling. In addition, the mixture was stirred at the same temperature for 5 hours. The reaction mixture was washed successively with water and saturated brine, dried (Na 2 SO 4 ), methylene chloride was distilled off under reduced pressure, and the obtained residue was subjected to column chromatography (n-hexane: ethyl acetate, 1:10). → Purified by 1: 5, V / V). The solvent of the target fraction was distilled off under reduced pressure, diisopropyl ether was added to the obtained residue, and the mixture was collected by filtration to obtain 1.30 g (yield: 27%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.29 (6H, s), 1.03 (9H, s), 2.53 (3H, s), 2.68 (3H, s), 3.75 (3H, s), 6.47 ( 1H, d, J = 2.4 Hz), 6.91 (1H, dd, J = 8.8, 2.4 Hz), 6.97-7.04 (2H, m), 7.12-7.19 (2H, m), 7.93 (1H, d, J = 8.8 Hz).

(5c) 3-アセチル-1-(4-ヒドロキシフェニル)-6-メトキシ-2-メチルインドール (5c) 3-Acetyl-1- (4-Hydroxyphenyl) -6-methoxy-2-methylindole

Figure 2020071550
Figure 2020071550

実施例5 (5b) で製造した化合物 1.29 g (3.15 mmol)をテトラヒドロフラン 13 mLに溶解し、氷冷下1.0 Mフッ化テトラ-n-ブチルアンモニウム テトラヒドロフラン溶液 3.47 mL (3.5 mmol)を加え、同温にて1時間撹拌した。反応液に水 30 mLを加え、酢酸エチル 30 mLにて抽出した。酢酸エチル層を飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下酢酸エチルを留去した。得られた残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の淡桃色粉末 900 mg (収率:97%) を得た。
1H-NMR (DMSO-d6, 400 MHz) δ:2.45 (3H, s), 2.56 (3H, s), 3.66 (3H, s), 6.39 (1H, d, J=2.2 Hz), 6.85 (1H, dd, J=8.8, 2.2 Hz), 6.94-7.01 (2H, m), 7.17-7.23 (2H, m), 7.96 (1H, d, J=8.8 Hz).
1.29 g (3.15 mmol) of the compound prepared in Example 5 (5b) is dissolved in 13 mL of tetrahydrofuran, 1.0 M tetra-n-butylammonium fluoride tetrahydrofuran solution 3.47 mL (3.5 mmol) is added under ice-cooling, and the temperature is the same. Was stirred for 1 hour. 30 mL of water was added to the reaction mixture, and the mixture was extracted with 30 mL of ethyl acetate. The ethyl acetate layer was washed with saturated brine and dried (Na 2 SO 4 ), and then ethyl acetate was distilled off under reduced pressure. Diisopropyl ether was added to the obtained residue and collected by filtration to obtain 900 mg (yield: 97%) of a pale pink powder of the title compound.
1 H-NMR (DMSO-d6, 400 MHz) δ: 2.45 (3H, s), 2.56 (3H, s), 3.66 (3H, s), 6.39 (1H, d, J = 2.2 Hz), 6.85 (1H) , dd, J = 8.8, 2.2 Hz), 6.94-7.01 (2H, m), 7.17-7.23 (2H, m), 7.96 (1H, d, J = 8.8 Hz).

(5) 3-アセチル-6-メトキシ-2-メチル-1-{4-[3-(2-オキソ-2λ5-[1,3,2]ジオキサホスフィナン-2-イル)プロポキシ]フェニル}インドール(5) 3-Acetyl-6-Methoxy-2-methyl-1-{4- [3- (2-oxo-2λ 5- [1,3,2] dioxaphosphinan-2-yl) propoxy] phenyl } Indole

Figure 2020071550
Figure 2020071550

実施例5 (5c) で製造した化合物 400 mg (1.35 mmol) をN,N-ジメチルホルムアミド 4.0 mLに溶解し、炭酸カリウム 281 mg (2.03 mmol)、ヨウ化カリウム 45 mg (0.27 mmol) および2-(3-ブロモプロピル)-[1,3,2]ジオキサホスフィナン 2-オキシド 362 mg (1.49 mmol) を加え、室温にて4時間、60℃ にて3.5時間撹拌した。反応液に水 20 mLを加え、30分間撹拌後、析出物をろ取し、水 100 mLを加え、酢酸エチル-テトラヒドロフラン (1:1) 200 mLにて抽出した。有機層を飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(酢酸エチル:クロロホルム:メタノール、1:0:0→0:20:1、V/V)にて精製した。目的分画の溶媒を減圧下留去後、得られた残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の白色粉末 455 mg (収率:86%) を得た。
IR ν (ATR) cm-1 :1624
1H-NMR (CDCl3, 400 MHz) δ:1.94-2.30 (6H, m), 2.52 (3H, s), 2.68 (3H, s), 3.75 (3H, s), 4.14 (2H, t, J=5.6 Hz), 4.21-4.35 (2H, m), 4.52-4.66 (2H, m), 6.46 (1H, d, J=2.4 Hz), 6.91 (1H, dd, J=7.3, 2.4 Hz), 7.02-7.11 (2H, m), 7.17-7.25 (2H, m), 7.93 (1H, d, J=8.8 Hz).
MS m/z: 458 [M+H]+, 480 [M+Na]+.
400 mg (1.35 mmol) of the compound prepared in Example 5 (5c) was dissolved in 4.0 mL of N, N-dimethylformamide, potassium carbonate 281 mg (2.03 mmol), potassium iodide 45 mg (0.27 mmol) and 2-. (3-Bromopropyl)-[1,3,2] dioxaphosphinan 2-oxide 362 mg (1.49 mmol) was added, and the mixture was stirred at room temperature for 4 hours and at 60 ° C. for 3.5 hours. 20 mL of water was added to the reaction mixture, and the mixture was stirred for 30 minutes, the precipitate was collected by filtration, 100 mL of water was added, and the mixture was extracted with 200 mL of ethyl acetate-tetrahydrofuran (1: 1). The organic layer was washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (ethyl acetate: chloroform: methanol, 1: 0: 0 → 0: 20: 1, V / V). After distilling off the solvent of the target fraction under reduced pressure, diisopropyl ether was added to the obtained residue and collected by filtration to obtain 455 mg (yield: 86%) of a white powder of the title compound.
IR ν (ATR) cm -1 : 1624
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.94-2.30 (6H, m), 2.52 (3H, s), 2.68 (3H, s), 3.75 (3H, s), 4.14 (2H, t, J) = 5.6 Hz), 4.21-4.35 (2H, m), 4.52-4.66 (2H, m), 6.46 (1H, d, J = 2.4 Hz), 6.91 (1H, dd, J = 7.3, 2.4 Hz), 7.02 -7.11 (2H, m), 7.17-7.25 (2H, m), 7.93 (1H, d, J = 8.8 Hz).
MS m / z: 458 [M + H] + , 480 [M + Na] + .

実施例6 Example 6

(6a) 4-[4-(3-アセチル-6-メトキシ-2-メチルインドール-1-イル)フェノキシ]ブチル アセテート (6a) 4- [4- (3-Acetyl-6-methoxy-2-methylindole-1-yl) phenoxy] butyl acetate

Figure 2020071550
Figure 2020071550

実施例5 (5c) で製造した化合物 500 mg (1.69 mmol) をN,N-ジメチルホルムアミド 5.0 mLに溶解し、炭酸カリウム 351 mg (2.54 mmol)、ヨウ化カリウム 56 mg (0.34 mmol) および4-クロロブチル アセテート 0.36 mL (2.5 mmol) を加え、60℃ にて17時間撹拌した。反応液に水 30 mLを加え、30分間撹拌後、析出物をろ取し、水、ジイソプロピルエーテルにて順次洗浄した。得られた粉末をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、2:1→1:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡黄色粉末 586 mg (収率:85%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.83-1.99 (4H, m), 2.08 (3H, s), 2.52 (3H, s), 2.68 (3H, s), 3.74 (3H, s), 4.08 (2H, t, J=5.6 Hz), 4.18 (2H, t, J=6.4 Hz), 6.46 (1H, d, J=2.4 Hz), 6.91 (1H, dd, J=8.8, 2.4 Hz), 7.02-7.09 (2H, m), 7.17-7.24 (2H, m), 7.93 (1H, d, J=8.8 Hz).
500 mg (1.69 mmol) of the compound prepared in Example 5 (5c) was dissolved in 5.0 mL of N, N-dimethylformamide, potassium carbonate 351 mg (2.54 mmol), potassium iodide 56 mg (0.34 mmol) and 4-. 0.36 mL (2.5 mmol) of chlorobutyl acetate was added, and the mixture was stirred at 60 ° C. for 17 hours. 30 mL of water was added to the reaction solution, and the mixture was stirred for 30 minutes, the precipitate was collected by filtration, and washed successively with water and diisopropyl ether. The obtained powder was purified by column chromatography (n-hexane: ethyl acetate, 2: 1 → 1: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 586 mg (yield: 85%) of a pale yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.83-1.99 (4H, m), 2.08 (3H, s), 2.52 (3H, s), 2.68 (3H, s), 3.74 (3H, s), 4.08 (2H, t, J = 5.6 Hz), 4.18 (2H, t, J = 6.4 Hz), 6.46 (1H, d, J = 2.4 Hz), 6.91 (1H, dd, J = 8.8, 2.4 Hz), 7.02-7.09 (2H, m), 7.17-7.24 (2H, m), 7.93 (1H, d, J = 8.8 Hz).

(6b) 3-アセチル-1-[4-(4-ヒドロキシブトキシ)フェニル]-6-メトキシ-2-メチルインドール (6b) 3-Acetyl-1- [4- (4-Hydroxybutoxy) phenyl] -6-methoxy-2-methylindole

Figure 2020071550
Figure 2020071550

実施例6 (6a) で製造した化合物 578 mg (1.41 mmol) を塩化メチレン-メタノール (1:1) 6.0 mLに溶解し、5.0 M 水酸化ナトリウム水 1.41 mL (7.1 mmol) を加え、室温にて1時間撹拌した。減圧下溶媒を留去し、水 20 mLを加え、30分間撹拌した。析出物をろ取し、標題化合物の白色粉末 483 mg (収率:93%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.57 (1H, t, J=5.4 Hz), 1.77-1.87 (2H, m), 1.91-2.01 (2H, m), 2.52 (3H, s), 2.68 (3H, s), 3.74 (3H, s), 3.75-3.82 (2H, m), 4.10 (2H, t, J=6.1 Hz), 6.46 (1H, d, J=2.4 Hz), 6.91 (1H, dd, J=8.8, 2.4 Hz), 7.02-7.10 (2H, m), 7.17-7.24 (2H, m), 7.93 (1H, d, J=8.8 Hz).
578 mg (1.41 mmol) of the compound prepared in Example 6 (6a) was dissolved in 6.0 mL of methylene chloride-methanol (1: 1), 1.41 mL (7.1 mmol) of 5.0 M sodium hydroxide water was added, and the mixture was added at room temperature. Stirred for 1 hour. The solvent was distilled off under reduced pressure, 20 mL of water was added, and the mixture was stirred for 30 minutes. The precipitate was collected by filtration to give 483 mg (yield: 93%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.57 (1H, t, J = 5.4 Hz), 1.77-1.87 (2H, m), 1.91-2.01 (2H, m), 2.52 (3H, s), 2.68 (3H, s), 3.74 (3H, s), 3.75-3.82 (2H, m), 4.10 (2H, t, J = 6.1 Hz), 6.46 (1H, d, J = 2.4 Hz), 6.91 (1H) , dd, J = 8.8, 2.4 Hz), 7.02-7.10 (2H, m), 7.17-7.24 (2H, m), 7.93 (1H, d, J = 8.8 Hz).

(6c) 4-[4-(3-アセチル-6-メトキシ-2-メチルインドール-1-イル)フェノキシ]ブチル メタンスルホネート (6c) 4- [4- (3-Acetyl-6-methoxy-2-methylindole-1-yl) phenoxy] butyl methanesulfonate

Figure 2020071550
Figure 2020071550

実施例6 (6b) で製造した化合物 478 mg (1.30 mmol) を塩化メチレン 5.0 mLに溶解し、氷冷下トリエチルアミン 0.54 mL (3.9 mmol)、次いで塩化メタンスルホニル 0.15 mL (2.0 mmol) を加え、同温にて1時間撹拌した。反応液に水 30 mLを加え、塩化メチレンにて抽出した。塩化メチレン層を飽和食塩水にて洗浄、乾燥(Na2SO4) 後、減圧下塩化メチレンを留去し、標題化合物の淡黄色粉末 552 mg (収率:95%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.94-2.10 (4H, m), 2.52 (3H, s), 2.68 (3H, s), 3.04 (3H, s), 3.74 (3H, s), 4.10 (2H, t, J=5.4 Hz), 4.36 (2H, t, J=5.8 Hz), 6.46 (1H, d, J=2.4 Hz), 6.91 (1H, dd, J=8.8, 2.4 Hz), 7.02-7.10 (2H, m), 7.18-7.25 (2H, m), 7.93 (1H, d, J=8.8 Hz).
478 mg (1.30 mmol) of the compound prepared in Example 6 (6b) was dissolved in 5.0 mL of methylene chloride, 0.54 mL (3.9 mmol) of triethylamine under ice cooling, and then 0.15 mL (2.0 mmol) of methanesulfonyl chloride were added thereto. The mixture was stirred at warm temperature for 1 hour. 30 mL of water was added to the reaction mixture, and the mixture was extracted with methylene chloride. The methylene chloride layer was washed with saturated brine and dried (Na 2 SO 4 ), and then methylene chloride was distilled off under reduced pressure to obtain 552 mg (yield: 95%) of a pale yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.94-2.10 (4H, m), 2.52 (3H, s), 2.68 (3H, s), 3.04 (3H, s), 3.74 (3H, s), 4.10 (2H, t, J = 5.4 Hz), 4.36 (2H, t, J = 5.8 Hz), 6.46 (1H, d, J = 2.4 Hz), 6.91 (1H, dd, J = 8.8, 2.4 Hz), 7.02-7.10 (2H, m), 7.18-7.25 (2H, m), 7.93 (1H, d, J = 8.8 Hz).

(6d) 3-アセチル-6-メトキシ-2-メチル-1-[4-(4-(モルホリン-4-イル)ブトキシ)フェニル]インドール (6d) 3-Acetyl-6-Methoxy-2-methyl-1- [4- (4- (morpholine-4-yl) butoxy) phenyl] indole

Figure 2020071550
Figure 2020071550

実施例6 (6c) で製造した化合物 544 mg (1.22 mmol) をN,N-ジメチルホルムアミド 5.5 mLに溶解し、モルホリン 0.21 mL (2.4 mmol)、炭酸カリウム 337 mg (2.44 mmol) を加え、60℃ にて16時間撹拌した。反応液に水 30 mLを加え、酢酸エチル 30 mLにて抽出した。酢酸エチル層を飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下酢酸エチルを留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、1:1→0:1、クロロホルム:メタノール、20:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡黄色オイル 478 mg (収率:90%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.68-1.79 (2H, m), 1.84-1.94 (2H, m), 2.41-2.51 (6H, m), 2.52 (3H, s), 2.68 (3H, s), 3.70-3.76 (4H, m), 3.74 (3H, s), 4.07 (2H, t, J=6.4 Hz), 6.46 (1H, d, J=2.4 Hz), 6.91 (1H, dd, J=8.8, 2.4 Hz), 7.02-7.09 (2H, m), 7.16-7.24 (2H, m), 7.93 (1H, d, J=8.8 Hz).
544 mg (1.22 mmol) of the compound prepared in Example 6 (6c) was dissolved in 5.5 mL of N, N-dimethylformamide, 0.21 mL (2.4 mmol) of morpholine and 337 mg (2.44 mmol) of potassium carbonate were added, and 60 ° C. Was stirred for 16 hours. 30 mL of water was added to the reaction mixture, and the mixture was extracted with 30 mL of ethyl acetate. The ethyl acetate layer was washed with saturated brine and dried (Na 2 SO 4 ), and then ethyl acetate was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 1: 1 → 0: 1, chloroform: methanol, 20: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 478 mg (yield: 90%) of pale yellow oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.68-1.79 (2H, m), 1.84-1.94 (2H, m), 2.41-2.51 (6H, m), 2.52 (3H, s), 2.68 (3H) , s), 3.70-3.76 (4H, m), 3.74 (3H, s), 4.07 (2H, t, J = 6.4 Hz), 6.46 (1H, d, J = 2.4 Hz), 6.91 (1H, dd, J = 8.8, 2.4 Hz), 7.02-7.09 (2H, m), 7.16-7.24 (2H, m), 7.93 (1H, d, J = 8.8 Hz).

(6) 3-アセチル-6-メトキシ-2-メチル-1-[4-(4-(モルホリン-4-イル)ブトキシ)フェニル]インドール 塩酸塩 (6) 3-Acetyl-6-methoxy-2-methyl-1- [4- (4- (morpholine-4-yl) butoxy) phenyl] indole hydrochloride

Figure 2020071550
Figure 2020071550

実施例6 (6d) で製造した化合物 470 mg (1.08 mmol) をメタノール 10 mLに溶解し、氷冷下8.6 M 塩化水素 イソプロパノール溶液 0.25 mL (2.2 mmol) を加え、同温にて30分間撹拌した。減圧下溶媒を留去し、得られた残渣に酢酸エチルを加え、不溶物をろ取した。得られた粉末をエタノール 30 mLに加熱溶解し、ジエチルエーテル 150 mLを加え、室温にて30分間撹拌した。析出物をろ取し、標題化合物の淡桃色粉末 343 mg (収率:67%) を得た。
IR ν (ATR) cm-1 :1622
1H-NMR (DMSO-d6, 400 MHz) δ:1.77-2.00 (4H, m), 2.46 (3H, s), 2.57 (3H, s), 2.98-3.13 (2H, m), 3.13-3.23 (2H, m), 3.40-3.50 (2H, m), 3.66 (3H, s), 3.75-3.87 (2H, m), 3.90-4.05 (2H, m), 4.12 (2H, t, J=5.8 Hz), 6.38 (1H, d, J=2.4 Hz), 6.87 (1H, dd, J=8.8, 2.4 Hz), 7.14-7.21 (2H, m), 7.33-7.41 (2H, m), 7.97 (1H, d, J=8.8 Hz), 10.67-10.87 (1H, br).
MS m/z: 438 [M+H]+.
470 mg (1.08 mmol) of the compound prepared in Example 6 (6d) was dissolved in 10 mL of methanol, 0.25 mL (2.2 mmol) of an 8.6 M hydrogen chloride isopropanol solution was added under ice-cooling, and the mixture was stirred at the same temperature for 30 minutes. .. The solvent was evaporated under reduced pressure, ethyl acetate was added to the obtained residue, and the insoluble material was collected by filtration. The obtained powder was dissolved by heating in 30 mL of ethanol, 150 mL of diethyl ether was added, and the mixture was stirred at room temperature for 30 minutes. The precipitate was collected by filtration to give 343 mg (yield: 67%) of a pale pink powder of the title compound.
IR ν (ATR) cm -1 : 1622
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 1.77-2.00 (4H, m), 2.46 (3H, s), 2.57 (3H, s), 2.98-3.13 (2H, m), 3.13-3.23 ( 2H, m), 3.40-3.50 (2H, m), 3.66 (3H, s), 3.75-3.87 (2H, m), 3.90-4.05 (2H, m), 4.12 (2H, t, J = 5.8 Hz) , 6.38 (1H, d, J = 2.4 Hz), 6.87 (1H, dd, J = 8.8, 2.4 Hz), 7.14-7.21 (2H, m), 7.33-7.41 (2H, m), 7.97 (1H, d) , J = 8.8 Hz), 10.67-10.87 (1H, br).
MS m / z: 438 [M + H] + .

実施例7 Example 7

(7a) 5-ブロモ-2-メトキシピリジン (7a) 5-Bromo-2-methoxypyridine

Figure 2020071550
Figure 2020071550

2-メトキシピリジン 60.8 g (0.560 mol) を塩化メチレン 300 mLに溶解し、酢酸ナトリウム 50.5 g (0.620 mol) を加え、-10℃ にて臭素 28.7 mL (0.620 mol) を45分間かけて滴下し、同温にて1時間撹拌後、室温にて12時間撹拌した。不溶物をろ別後、ろ液を5.0 M 水酸化ナトリウム水にてpH 8以上に調整し、二層を分離した。有機層を水にて洗浄後、乾燥 (Na2SO4) 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(n-ヘキサン:酢酸エチル、1:20→1:10、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の無色油状物 84.5 g (収率:80%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:6.65 (1H, d, J=8.8 Hz), 7.63 (1H, dd, J=8.8, 2.7 Hz), 8.19 (1H, d, J=2.7 Hz).
Dissolve 60.8 g (0.560 mol) of 2-methoxypyridine in 300 mL of methylene chloride, add 50.5 g (0.620 mol) of sodium acetate, and add 28.7 mL (0.620 mol) of bromine at -10 ° C over 45 minutes. After stirring at the same temperature for 1 hour, the mixture was stirred at room temperature for 12 hours. After filtering the insoluble material, the filtrate was adjusted to pH 8 or higher with 5.0 M sodium hydroxide water, and the two layers were separated. The organic layer was washed with water and then dried (Na 2 SO 4 ). The solvent was evaporated under reduced pressure, and the obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 1:20 → 1:10, V / V). The solvent of the target fraction was evaporated under reduced pressure to give 84.5 g (yield: 80%) of a colorless oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 6.65 (1H, d, J = 8.8 Hz), 7.63 (1H, dd, J = 8.8, 2.7 Hz), 8.19 (1H, d, J = 2.7 Hz) ..

(7b) 5-ブロモ-2-メトキシピリジン-N-オキシド (7b) 5-Bromo-2-methoxypyridin-N-oxide

Figure 2020071550
Figure 2020071550

実施例7 (7a) で製造した化合物 84.5 g (0.450 mol) をクロロホルム 500 mLに溶解し、氷冷下、m-クロロ過安息香酸 (P=77%) 101 g (0.450 mol) を加え、室温にて19時間撹拌した。反応液を乾燥 (Na2SO4) 後、減圧下クロロホルムを留去し、残渣をカラムクロマトグラフィー(クロロホルム:酢酸エチル、1:0→1:1、クロロホルム:メタノール、10:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 43.9 g (収率:48%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:4.07 (3H, s), 6.78 (1H, d, J=9.0 Hz), 7.38 (1H, d, J=9.0 Hz), 8.38-8.42 (1H, m).
84.5 g (0.450 mol) of the compound prepared in Example 7 (7a) was dissolved in 500 mL of chloroform, and 101 g (0.450 mol) of m-chloroperbenzoic acid (P = 77%) was added under ice-cooling to room temperature. Was stirred for 19 hours. After the reaction solution was dried (Na 2 SO 4 ), chloroform was distilled off under reduced pressure, and the residue was subjected to column chromatography (chloroform: ethyl acetate, 1: 0 → 1: 1, chloroform: methanol, 10: 1, V / V. ). The solvent of the target fraction was distilled off under reduced pressure to obtain 43.9 g (yield: 48%) of a white powder of the title compound.
1 H-NMR (CDCl 3 , 400 MHz) δ: 4.07 (3H, s), 6.78 (1H, d, J = 9.0 Hz), 7.38 (1H, d, J = 9.0 Hz), 8.38-8.42 (1H, s) m).

(7c) 5-ブロモ-2-メトキシ-4-ニトロピリジン (7c) 5-Bromo-2-methoxy-4-nitropyridine

Figure 2020071550
Figure 2020071550

氷冷下、濃硫酸 80 mLに実施例7 (7b) で製造した化合物 43.9 g (0.220 mol) を分割添加し、発煙硝酸 101 mL (2.20 mol) を滴下した。反応液を徐々に昇温させ、80℃ にて3日間撹拌した。反応液を氷水中に注加し、炭酸カリウムにて中和し、クロロホルムにて抽出後、乾燥 (Na2SO4) した。減圧下溶媒を留去し、標題化合物の黄色粉末 30.0 g (収率:60%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:3.99 (3H, s), 7.11 (1H, s), 8.45 (1H, s).
Under ice-cooling, 43.9 g (0.220 mol) of the compound prepared in Example 7 (7b) was added in portions to 80 mL of concentrated sulfuric acid, and 101 mL (2.20 mol) of fuming nitric acid was added dropwise. The temperature of the reaction solution was gradually raised, and the mixture was stirred at 80 ° C. for 3 days. The reaction mixture was poured into ice water, neutralized with potassium carbonate, extracted with chloroform, and dried (Na 2 SO 4 ). The solvent was distilled off under reduced pressure to obtain 30.0 g (yield: 60%) of a yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 3.99 (3H, s), 7.11 (1H, s), 8.45 (1H, s).

(7d) ビス-tert-ブチル (5-ブロモ-2-メトキシピリジン-4-イル)カルバメート (7d) Bis-tert-butyl (5-bromo-2-methoxypyridin-4-yl) carbamate

Figure 2020071550
Figure 2020071550

実施例7 (7c) で製造した化合物 26.3 g (0.113 mol) を酢酸 100 mLに溶解し、鉄 30.7 g (0.550 mol) を加え、80℃ にて1時間撹拌した。不溶物をセライトにてろ別し、ろ液に水を加え、炭酸カリウムにて中和した。酢酸エチルにて2回抽出後、有機層を合わせ、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をテトラヒドロフラン 100 mLに溶解し、二炭酸ジ-tert-ブチル33.9 mL (0.148 mol)、N,N-ジメチルアミノピリジン 552 mg (4.90 mmol)を加え、1時間加熱還流した。テトラヒドロフランを減圧下留去し、酢酸エチルを加え、水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→3:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 18.2 g (収率:40%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.42 (18H, s), 3.94 (3H, s), 6.65 (1H, s), 8.28 (1H, s).
26.3 g (0.113 mol) of the compound prepared in Example 7 (7c) was dissolved in 100 mL of acetic acid, 30.7 g (0.550 mol) of iron was added, and the mixture was stirred at 80 ° C. for 1 hour. The insoluble material was filtered off with Celite, water was added to the filtrate, and the mixture was neutralized with potassium carbonate. After extraction with ethyl acetate twice, the organic layers were combined, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was dissolved in 100 mL of tetrahydrofuran, 33.9 mL (0.148 mol) of di-tert-butyl dicarbonate and 552 mg (4.90 mmol) of N, N-dimethylaminopyridine were added, and the mixture was heated under reflux for 1 hour. Tetrahydrofuran was evaporated under reduced pressure, ethyl acetate was added, the mixture was washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 3: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 18.2 g (yield: 40%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.42 (18H, s), 3.94 (3H, s), 6.65 (1H, s), 8.28 (1H, s).

(7e) ビス-tert-ブチル [2-メトキシ-5-(2-オキソプロピル)ピリジン-4-イル]カルバメート (7e) Bis-tert-butyl [2-methoxy-5- (2-oxopropyl) pyridin-4-yl] carbamate

Figure 2020071550
Figure 2020071550

窒素雰囲気下、実施例7 (7d) で製造した化合物 9.0 g (22 mmol) をトルエン 35 mLに溶解し、酢酸イソプロペニル 3.69 mL (33.5 mmol)、酢酸パラジウム 250 mg (1.12 mmol)、トリス(2-メチルフェニル)ホスフィン 678 mg (2.23 mmol)、トリブチルスズメトキシド 9.63 mL (33.5 mmol) を加え、80℃ にて20時間撹拌した。反応液を減圧下留去し、得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→5:1→3:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の無色油状物 1.40 g (収率:17%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.42 (18H, s), 2.15 (3H, s), 3.51 (2H, s), 3.95 (3H, s), 6.56 (1H, s), 8.06 (1H, s).
Under a nitrogen atmosphere, 9.0 g (22 mmol) of the compound prepared in Example 7 (7d) was dissolved in 35 mL of toluene, isopropenyl acetate 3.69 mL (33.5 mmol), palladium acetate 250 mg (1.12 mmol), Tris (2). -Methylphenyl) phosphine 678 mg (2.23 mmol) and tributyltin methoxyde 9.63 mL (33.5 mmol) were added, and the mixture was stirred at 80 ° C. for 20 hours. The reaction mixture was evaporated under reduced pressure, and the obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 5: 1 → 3: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure to give 1.40 g (yield: 17%) of a colorless oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.42 (18H, s), 2.15 (3H, s), 3.51 (2H, s), 3.95 (3H, s), 6.56 (1H, s), 8.06 ( 1H, s).

(7f) 6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン (7f) 6-Methoxy-2-methyl-1H-pyrrolo [3,2-c] pyridine

Figure 2020071550
Figure 2020071550

実施例7 (7e) で製造した化合物 1.40 g (3.68 mmol) を室温にてトリフルオロ酢酸 10 mLに溶解し、同温にて1時間撹拌した。減圧下トリフルオロ酢酸を留去し、残渣を飽和重曹水にて中和した。クロロホルムにて2回抽出後、有機層を合わせ、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、3:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 460 mg (収率:77%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.42 (3H, s), 3.89 (3H, s), 6.20 (1H, s), 6.65 (1H, s), 8.30 (1H, s), 9.27-9.50 (1H, br).
1.40 g (3.68 mmol) of the compound prepared in Example 7 (7e) was dissolved in 10 mL of trifluoroacetic acid at room temperature, and the mixture was stirred at the same temperature for 1 hour. Trifluoroacetic acid was distilled off under reduced pressure, and the residue was neutralized with saturated aqueous sodium hydrogen carbonate. After extraction with chloroform twice, the organic layers were combined, dried (Na 2 SO 4 ), and the solvent was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 3: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 460 mg (yield: 77%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.42 (3H, s), 3.89 (3H, s), 6.20 (1H, s), 6.65 (1H, s), 8.30 (1H, s), 9.27- 9.50 (1H, br).

(7g) 1-(4-{4-[2-(tert-ブチルジメチルシラニルオキシ)エトキシ]ピペリジン-1-イル}フェニル)-6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン (7g) 1- (4- {4- [2- (tert-butyldimethylsilanyloxy) ethoxy] piperidine-1-yl} phenyl) -6-methoxy-2-methyl-1H-pyrrolo [3,2- c] Pyridine

Figure 2020071550
Figure 2020071550

実施例7 (7f) で製造した化合物 450 mg (2.77 mmol) をトルエン 5 mLに溶解し、実施例3 (3c) で製造した化合物 1.54 g (3.32 mmol)、ヨウ化銅 27 mg (0.14 mmol)、粉砕したリン酸三カリウム 1.23 g (5.82 mmol)、trans-N,N’-ジメチルシクロヘキサン-1,2-ジアミン 0.066 mL (0.42 mmol) を加え、窒素を10分間バブリング後、窒素雰囲気下100℃ で28時間撹拌した。ヨウ化銅27 mg (0.14 mmol)、trans-N,N’-ジメチルシクロヘキサン-1,2-ジアミン 0.066 mL (0.42 mmol) を追加し、さらに13時間撹拌した。酢酸エチル、水を加え、不溶物をセライトにてろ別した。有機層を飽和食塩水で洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→6:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の褐色油状物 210 mg (収率:15%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.09 (6H, s), 0.91 (9H, s), 1.70-1.83 (2H, m), 2.00-2.10 (2H, m), 2.22 (3H, s), 2.97-3.10 (2H, m), 3.55-3.65 (5H, m), 3.78 (2H, t, J=5.4 Hz), 3.91(3H, s), 6.31 (1H, s), 6.35 (1H, s), 7.00-7.02 (2H, m), 7.13-7.15 (2H, m), 8.36 (1H, s).
Compound 450 mg (2.77 mmol) prepared in Example 7 (7f) was dissolved in 5 mL of toluene, and compound 1.54 g (3.32 mmol) prepared in Example 3 (3c), copper iodide 27 mg (0.14 mmol). Add 1.23 g (5.82 mmol) of crushed tripotassium phosphate and 0.066 mL (0.42 mmol) of trans-N, N'-dimethylcyclohexane-1,2-diamine, bubbling nitrogen for 10 minutes, and then 100 ° C. under a nitrogen atmosphere. Was stirred for 28 hours. 27 mg (0.14 mmol) of copper iodide and 0.066 mL (0.42 mmol) of trans-N, N'-dimethylcyclohexane-1,2-diamine were added, and the mixture was further stirred for 13 hours. Ethyl acetate and water were added, and the insoluble material was filtered off with Celite. The organic layer was washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 6: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 210 mg (yield: 15%) of a brown oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.09 (6H, s), 0.91 (9H, s), 1.70-1.83 (2H, m), 2.00-2.10 (2H, m), 2.22 (3H, s) ), 2.97-3.10 (2H, m), 3.55-3.65 (5H, m), 3.78 (2H, t, J = 5.4 Hz), 3.91 (3H, s), 6.31 (1H, s), 6.35 (1H, 1H, s), 7.00-7.02 (2H, m), 7.13-7.15 (2H, m), 8.36 (1H, s).

(7h) 2-{1-[4-(6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン-1-イル)フェニル]ピペリジン-4-イルオキシ}エチル アセテート (7h) 2- {1- [4- (6-Methoxy-2-methyl-1H-pyrrolo [3,2-c] pyridin-1-yl) phenyl] piperidine-4-yloxy} ethyl acetate

Figure 2020071550
Figure 2020071550

実施例7 (7g) で製造した化合物 210 mg (0.42 mmol) をテトラヒドロフラン 2 mL に溶解し、氷冷下1.0 M フッ化テトラ-n-ブチルアンモニウム テトラヒドロフラン溶液 1.3 mL (1.3 mmol) を加え、同温にて30分間撹拌後、1.0 M フッ化テトラ-n-ブチルアンモニウム テトラヒドロフラン溶液0.63 mL (0.63 mmol) を追加し、室温にて2時間撹拌した。反応液に水を加え、酢酸エチルにて2回抽出した。酢酸エチル層を合わせ、水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた無色油状物を塩化メチレン 2 mL に溶解し、氷冷下トリエチルアミン 0.18 mL (1.3 mmol)、塩化アセチル 60 μL (0.84 mmol) を加え、同温にて30分間撹拌した。反応液を水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、3:1→1:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の無色油状物 125 mg (収率:71%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.72-1.86 (2H, m), 2.00-2.08 (2H, m), 2.10 (3H, s), 2.22 (3H, s), 3.00-3.10 (2H, m), 3.50-3.66 (3H, m), 3.69-3.78 (2H, m), 3.91(3H, s), 4.24-4.30 (2H, m), 6.31 (1H, s), 6.35 (1H, s), 7.00-7.03 (2H, m), 7.14-7.16 (2H, m), 8.37 (1H, s).
210 mg (0.42 mmol) of the compound prepared in Example 7 (7 g) was dissolved in 2 mL of tetrahydrofuran, 1.3 mL (1.3 mmol) of a 1.0 M tetra-n-butylammonium tetrahydrofuran tetrahydrofuran solution was added under ice-cooling, and the temperature was the same. After stirring for 30 minutes, 0.63 mL (0.63 mmol) of a 1.0 M tetra-n-butylammonium tetrahydrofuran solution was added, and the mixture was stirred at room temperature for 2 hours. Water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The ethyl acetate layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained colorless oil was dissolved in 2 mL of methylene chloride, 0.18 mL (1.3 mmol) of triethylamine and 60 μL (0.84 mmol) of acetyl chloride were added under ice-cooling, and the mixture was stirred at the same temperature for 30 minutes. The reaction mixture was washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 3: 1 → 1: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure to give 125 mg (yield: 71%) of a colorless oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.72-1.86 (2H, m), 2.00-2.08 (2H, m), 2.10 (3H, s), 2.22 (3H, s), 3.00-3.10 (2H) , m), 3.50-3.66 (3H, m), 3.69-3.78 (2H, m), 3.91 (3H, s), 4.24-4.30 (2H, m), 6.31 (1H, s), 6.35 (1H, s) ), 7.00-7.03 (2H, m), 7.14-7.16 (2H, m), 8.37 (1H, s).

(7i) 2-{1-[4-(3-アセチル-6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン-1-イル)フェニル]ピペリジン-4-イルオキシ}エチル アセテート (7i) 2- {1- [4- (3-Acetyl-6-methoxy-2-methyl-1H-pyrrolo [3,2-c] pyridin-1-yl) phenyl] piperidine-4-yloxy} ethyl acetate

Figure 2020071550
Figure 2020071550

粉砕した塩化アルミニウム 60 mg (0.45 mmol) を塩化メチレン 1 mLに懸濁し、氷冷下塩化アセチル 39 μL (0.54 mmol) を加え、同温にて15分間撹拌した。実施例7 (7h) で製造した化合物 76 mg (0.18 mmol)の塩化メチレン 1 mLの溶液を反応液に滴下し、室温にて1時間撹拌した。反応液に塩化メチレンおよび水を加え、飽和重曹水にて中和した。二層を分離し、塩化メチレン層を乾燥(Na2SO4)した。減圧下溶媒を留去し、標題化合物の褐色油状物 42 mg (収率:50%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.73-1.85 (2H, m), 2.00-2.08 (2H, m), 2.04 (3H, s), 2.53 (3H, s), 2.69 (3H, s), 3.04-3.14 (2H, m), 3.54-3.66 (3H, m), 3.72 (2H, t, J=5.1 Hz), 3.94 (3H, s), 4.20-4.30 (2H, m), 6.33 (1H, s), 7.02-7.04 (2H, m), 7.10-7.12 (2H, m), 8.87 (1H, s).
60 mg (0.45 mmol) of pulverized aluminum chloride was suspended in 1 mL of methylene chloride, 39 μL (0.54 mmol) of acetyl chloride was added under ice-cooling, and the mixture was stirred at the same temperature for 15 minutes. A solution of 76 mg (0.18 mmol) of the compound prepared in Example 7 (7h) in 1 mL of methylene chloride was added dropwise to the reaction mixture, and the mixture was stirred at room temperature for 1 hour. Methylene chloride and water were added to the reaction mixture, and the mixture was neutralized with saturated aqueous sodium hydrogen carbonate. The two layers were separated and the methylene chloride layer was dried (Na 2 SO 4 ). The solvent was distilled off under reduced pressure to obtain 42 mg (yield: 50%) of a brown oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.73-1.85 (2H, m), 2.00-2.08 (2H, m), 2.04 (3H, s), 2.53 (3H, s), 2.69 (3H, s) ), 3.04-3.14 (2H, m), 3.54-3.66 (3H, m), 3.72 (2H, t, J = 5.1 Hz), 3.94 (3H, s), 4.20-4.30 (2H, m), 6.33 ( 1H, s), 7.02-7.04 (2H, m), 7.10-7.12 (2H, m), 8.87 (1H, s).

(7) 2-{1-[4-(3-アセチル-6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン-1-イル)フェニル]ピペリジン-4-イルオキシ}エタノール (7) 2- {1- [4- (3-Acetyl-6-methoxy-2-methyl-1H-pyrrolo [3,2-c] pyridin-1-yl) phenyl] piperidine-4-yloxy} ethanol

Figure 2020071550
Figure 2020071550

実施例7 (7i) で製造した化合物 40 mg (0.086 mmol) を塩化メチレン-メタノール (1:1)の混液 1 mLに溶解し、室温にて5.0 M 水酸化ナトリウム水 86 μL (0.43 mmol) を加え、同温にて30分間撹拌した。反応液に水を加え、塩化メチレンにて抽出後、乾燥(Na2SO4)し、減圧下溶媒を留去した。残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の白色粉末 20 mg (収率:56%) を得た。
IR ν (ATR) cm-1 :3350-3100, 1635
1H-NMR (CDCl3, 400 MHz) δ:1.75-1.83 (2H, m), 2.03-2.11 (2H, m), 2.54 (3H, s), 2.69 (3H, s), 3.05-3.15 (2H, m), 3.57-3.69 (5H, m), 3.73 (2H, m), 3.94 (3H, s), 6.33 (1H, s), 7.03-7.05 (2H, m), 7.11-7.13 (2H, m), 8.88 (1H, s).
MS m/z: 424 [M+H]+, 446 [M+Na]+.
40 mg (0.086 mmol) of the compound prepared in Example 7 (7i) was dissolved in 1 mL of a mixed solution of methylene chloride-methanol (1: 1), and at room temperature, 86 μL (0.43 mmol) of 5.0 M sodium hydroxide water was added. In addition, the mixture was stirred at the same temperature for 30 minutes. Water was added to the reaction mixture, the mixture was extracted with methylene chloride, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. Diisopropyl ether was added to the residue and collected by filtration to give 20 mg (yield: 56%) of a white powder of the title compound.
IR ν (ATR) cm -1 : 3350-3100, 1635
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.75-1.83 (2H, m), 2.03-2.11 (2H, m), 2.54 (3H, s), 2.69 (3H, s), 3.05-3.15 (2H) , m), 3.57-3.69 (5H, m), 3.73 (2H, m), 3.94 (3H, s), 6.33 (1H, s), 7.03-7.05 (2H, m), 7.11-7.13 (2H, m) ), 8.88 (1H, s).
MS m / z: 424 [M + H] + , 446 [M + Na] + .

実施例8 Example 8

(8a) 1-(4-{4-[2-(tert-ブチルジメチルシラニルオキシ)エトキシ]ピペリジン-1-イル}フェニル)-3-ヨード-6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン (8a) 1-(4- {4- [2- (tert-butyldimethylsilanyloxy) ethoxy] piperidine-1-yl} phenyl) -3-iodo-6-methoxy-2-methyl-1H-pyrrolo [ 3,2-c] pyridine

Figure 2020071550
Figure 2020071550

実施例7 (7g) で製造した化合物 3.03 g (6.11 mmol) をテトラヒドロフラン 30 mLに溶解させ、N-ヨードスクシンイミド 1.51 g (6.72 mmol) を加え、室温にて40分間撹拌した。反応液に5% チオ硫酸ナトリウム水 100 mLを加え、酢酸エチルにて2回抽出した。有機層を合わせ、飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、4:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の微黄色粉末 3.52 g (収率:93%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.09 (6H, s), 0.91 (9H, s), 1.70-1.84 (2H, m), 1.97-2.09 (2H, m), 2.26 (3H, s), 3.00-3.12 (2H, m), 3.53-3.64 (5H, m), 3.73-3.82 (2H, m), 3.92 (3H, s), 6.29 (1H, s), 6.98-7.04 (2H, m), 7.08-7.14 (2H, m), 8.20 (1H, s).
3.03 g (6.11 mmol) of the compound prepared in Example 7 (7 g) was dissolved in 30 mL of tetrahydrofuran, 1.51 g (6.72 mmol) of N-iodosuccinimide was added, and the mixture was stirred at room temperature for 40 minutes. 100 mL of 5% sodium thiosulfate solution was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 4: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 3.52 g (yield: 93%) of a slightly yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.09 (6H, s), 0.91 (9H, s), 1.70-1.84 (2H, m), 1.97-2.09 (2H, m), 2.26 (3H, s) ), 3.00-3.12 (2H, m), 3.53-3.64 (5H, m), 3.73-3.82 (2H, m), 3.92 (3H, s), 6.29 (1H, s), 6.98-7.04 (2H, m) ), 7.08-7.14 (2H, m), 8.20 (1H, s).

(8b) 1-(4-{4-[2-(tert-ブチルジメチルシラニルオキシ)エトキシ]ピペリジン-1-イル}フェニル)-6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン-3-カルボニトリル (8b) 1-(4- {4- [2- (tert-butyldimethylsilanyloxy) ethoxy] piperidine-1-yl} phenyl) -6-methoxy-2-methyl-1H-pyrrolo [3,2- c] Pyridine-3-carbonitrile

Figure 2020071550
Figure 2020071550

実施例8 (8a) で製造した化合物 3.49 g (5.61 mmol)、シアン化銅(I) 1.76 g (19.6 mmol)、トリス(ジベンジリデンアセトン)ジパラジウム(0) 257 mg (0.281 mmol)、1,1'-ビス(ジフェニルホスフィノ)フェロセン544 mg (0.982 mmol) を1,4-ジオキサン 60 mLに懸濁させ、15時間加熱還流後、室温にて7時間撹拌した。不溶物をセライトにてろ別後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、4:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の黄色粉末 2.27 g (収率:78%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.09 (6H, s), 0.91 (9H, s), 1.70-1.83 (2H, m), 1.97-2.09 (2H, m), 2.39 (3H, s), 3.03-3.15 (2H, m), 3.55-3.66 (5H, m), 3.73-3.82 (2H, m), 3.93 (3H, s), 6.36 (1H, s), 6.98-7.07 (2H, m), 7.08-7.15 (2H, m), 8.51 (1H, s).
Compound 3.49 g (5.61 mmol) prepared in Example 8 (8a), copper (I) cyanide (I) 1.76 g (19.6 mmol), tris (dibenzylideneacetone) dipalladium (0) 257 mg (0.281 mmol), 1, 544 mg (0.982 mmol) of 1'-bis (diphenylphosphino) ferrocene was suspended in 60 mL of 1,4-dioxane, heated under reflux for 15 hours, and stirred at room temperature for 7 hours. The insoluble material was filtered off with Celite, and the solvent was distilled off under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 4: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 2.27 g (yield: 78%) of a yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.09 (6H, s), 0.91 (9H, s), 1.70-1.83 (2H, m), 1.97-2.09 (2H, m), 2.39 (3H, s) ), 3.03-3.15 (2H, m), 3.55-3.66 (5H, m), 3.73-3.82 (2H, m), 3.93 (3H, s), 6.36 (1H, s), 6.98-7.07 (2H, m) ), 7.08-7.15 (2H, m), 8.51 (1H, s).

(8) 1-{4-[4-(2-ヒドロキシエトキシ)ピペリジン-1-イル]フェニル}-6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン-3-カルボキサミド (8) 1- {4- [4- (2-Hydroxyethoxy) piperidine-1-yl] phenyl} -6-methoxy-2-methyl-1H-pyrrolo [3,2-c] pyridine-3-carboxamide

Figure 2020071550
Figure 2020071550

氷冷下、実施例8 (8b) で製造した化合物 1.92 g (3.69 mmol) に濃硫酸15 mLを加え、室温にて3時間撹拌した。氷冷下、反応液に水 100 mLを加え、5.0 M 水酸化ナトリウム水にて中和後、クロロホルムにて13回抽出した。有機層を合わせ、飽和食塩水にて洗浄、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣を酢酸エチルにて洗浄後、10% 塩酸 8mLに溶解させた。トルエン 8 mLを加え、撹拌しながら5.0 M 水酸化ナトリウム水にて中和後、析出物をろ取し、標題化合物の微黄色粉末 754 mg (収率:48%) を得た。
IR ν(ATR) cm-1 :1641, 1513, 1352, 1144
1H-NMR (DMSO-d6, 400 MHz) δ:1.88-2.03 (2H, m), 2.38 (3H, s), 2.95-3.07 (2H, m), 3.44-3.56 (5H, m), 3.58-3.69 (2H, m), 3.82 (3H, s), 6.18 (1H, s), 7.07-7.15 (2H, m), 7.16-7.25 (2H, m), 8.65 (1H, s).
MS m/z: 425 [M+H]+, 447 [M+Na]+, 423 [M-H]-.
Under ice-cooling, 15 mL of concentrated sulfuric acid was added to 1.92 g (3.69 mmol) of the compound prepared in Example 8 (8b), and the mixture was stirred at room temperature for 3 hours. Under ice-cooling, 100 mL of water was added to the reaction solution, neutralized with 5.0 M sodium hydroxide water, and extracted 13 times with chloroform. The organic layers were combined, washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was washed with ethyl acetate and then dissolved in 8 mL of 10% hydrochloric acid. Toluene (8 mL) was added, and the mixture was neutralized with 5.0 M sodium hydroxide water with stirring, and the precipitate was collected by filtration to obtain 754 mg (yield: 48%) of a slightly yellow powder of the title compound.
IR ν (ATR) cm -1 : 1641, 1513, 1352, 1144
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 1.88-2.03 (2H, m), 2.38 (3H, s), 2.95-3.07 (2H, m), 3.44-3.56 (5H, m), 3.58- 3.69 (2H, m), 3.82 (3H, s), 6.18 (1H, s), 7.07-7.15 (2H, m), 7.16-7.25 (2H, m), 8.65 (1H, s).
MS m / z: 425 [M + H] + , 447 [M + Na] + , 423 [MH] - .

実施例9 Example 9

(9a) 4-ヒドロキシ-6,7-ジメトキシ-1-メチル-1H-キノリン-2-オン (9a) 4-Hydroxy-6,7-dimethoxy-1-methyl-1H-quinoline-2-one

Figure 2020071550
Figure 2020071550

3,4-ジメトキシアニリン 8.86 g (57.8 mmol) をテトラヒドロフラン 100 mLに溶解し、二炭酸ジ-tert-ブチル 14.0 mL (60.9 mmol) を加え、1.5時間加熱還流した。減圧下溶媒を留去し、n-ヘキサンを加え、再度減圧下溶媒を留去した。得られた残渣にジイソプロピルエーテルを加え、ろ取し、tert-ブチル (3,4-ジメトキシフェニル)カルバメートを含む褐色粉末10.3 gを得た。
得られた粉末 10.3 gをN,N-ジメチルホルムアミド 100 mLに溶解し、0℃ にて水素化ナトリウム (P=60%) 1.94 g (49 mmol) を分割添加し、同温にて15分間撹拌後、ヨウ化メチル 3.78 mL (60.7 mmol) を加え、同温にて40分間撹拌し、室温にて30分間撹拌した。反応液に水を加え、酢酸エチルにて2回抽出した。有機層を合わせ、水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去し、tert-ブチル (3,4-ジメトキシフェニル)メチルカルバメートを含む残渣を得た。
得られた残渣にぎ酸 30 mLを加え、氷冷下8.6 M 塩化水素 イソプロパノール溶液 15.3 mL (0.13 mol) を加え、同温にて20分間撹拌した。反応液を氷水1.2 Lに注加し、炭酸カリウムにて中和後、酢酸エチルにて2回抽出し、有機層を合わせ、飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去し、(3,4-ジメトキシフェニル)メチルアミンを含む残渣を得た。
得られた残渣にマロン酸 8.43 g (81.0 mmol) およびオキシ塩化リン 35.0 mL (378 mmol) を加え、90℃ にて2時間撹拌した。反応液を氷水に注加し、5.0 M 水酸化ナトリウム水 130 mLを加え、炭酸カリウムにて中和後、酢酸エチルにて2回、クロロホルムにて3回抽出した。有機層を合わせ、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣に酢酸エチルを加え、ろ取し、標題化合物の赤褐色粉末 1.96 g (収率:14%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:3.53 (3H, s), 3.80 (3H, s), 3.91 (3H, s), 5.75 (1H, s), 6.90 (1H, s), 7.27 (1H, s), 11.14 (1H, s).
8.86 g (57.8 mmol) of 3,4-dimethoxyaniline was dissolved in 100 mL of tetrahydrofuran, 14.0 mL (60.9 mmol) of di-tert-butyl dicarbonate was added, and the mixture was heated under reflux for 1.5 hours. The solvent was distilled off under reduced pressure, n-hexane was added, and the solvent was distilled off again under reduced pressure. Diisopropyl ether was added to the obtained residue and collected by filtration to obtain 10.3 g of a brown powder containing tert-butyl (3,4-dimethoxyphenyl) carbamate.
Dissolve 10.3 g of the obtained powder in 100 mL of N, N-dimethylformamide, add 1.94 g (49 mmol) of sodium hydride (P = 60%) in portions at 0 ° C, and stir at the same temperature for 15 minutes. After that, 3.78 mL (60.7 mmol) of methyl iodide was added, and the mixture was stirred at the same temperature for 40 minutes and at room temperature for 30 minutes. Water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers are combined, washed sequentially with water and saturated brine, dried (Na 2 SO 4 ), and then the solvent is distilled off under reduced pressure to obtain a residue containing tert-butyl (3,4-dimethoxyphenyl) methylcarbamate. rice field.
30 mL of the obtained residue was added, and 15.3 mL (0.13 mol) of an 8.6 M hydrogen chloride isopropanol solution was added under ice-cooling, and the mixture was stirred at the same temperature for 20 minutes. The reaction mixture is poured into 1.2 L of ice water, neutralized with potassium carbonate, extracted twice with ethyl acetate, combined with organic layers, washed with saturated brine, dried (Na 2 SO 4 ), and then reduced in pressure. The lower solvent was distilled off to obtain a residue containing (3,4-dimethoxyphenyl) methylamine.
To the obtained residue was added 8.43 g (81.0 mmol) of malonic acid and 35.0 mL (378 mmol) of phosphorus oxychloride, and the mixture was stirred at 90 ° C. for 2 hours. The reaction mixture was poured into ice water, 130 mL of 5.0 M sodium hydroxide water was added, neutralized with potassium carbonate, and extracted twice with ethyl acetate and three times with chloroform. The organic layers were combined, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. Ethyl acetate was added to the obtained residue and collected by filtration to obtain 1.96 g (yield: 14%) of a reddish brown powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 3.53 (3H, s), 3.80 (3H, s), 3.91 (3H, s), 5.75 (1H, s), 6.90 (1H, s), 7.27 ( 1H, s), 11.14 (1H, s).

(9b) 4-クロロ-6,7-ジメトキシ-1-メチル-1H-キノリン-2-オン (9b) 4-Chloro-6,7-dimethoxy-1-methyl-1H-quinoline-2-one

Figure 2020071550
Figure 2020071550

実施例9 (9a) で製造した化合物 948 mg (4.03 mmol) にオキシ塩化リン 3 5.63 mL (60.5 mmol) を加え、90℃ にて2時間撹拌した。反応液に水100 mLを加え、炭酸カリウムにてpH 11以上とし、酢酸エチルにて2回、クロロホルムにて2回抽出した。有機層を合わせ、飽和食塩水にて洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣に酢酸エチルを加え、ろ取し、標題化合物の赤褐色粉末 815 mg (収率:80%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:3.71 (3H, s), 3.98 (3H, s), 4.03 (3H, s), 6.77 (1H, s), 6.78 (1H, s), 7.36 (1H, s).
To 948 mg (4.03 mmol) of the compound prepared in Example 9 (9a) was added 3 5.63 mL (60.5 mmol) of phosphorus oxychloride, and the mixture was stirred at 90 ° C. for 2 hours. 100 mL of water was added to the reaction mixture, the pH was adjusted to 11 or higher with potassium carbonate, and the mixture was extracted twice with ethyl acetate and twice with chloroform. The organic layers were combined, washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. Ethyl acetate was added to the obtained residue and collected by filtration to obtain 815 mg (yield: 80%) of a reddish brown powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 3.71 (3H, s), 3.98 (3H, s), 4.03 (3H, s), 6.77 (1H, s), 6.78 (1H, s), 7.36 ( 1H, s).

(9c) 1-フェニル-3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン (9c) 1-Phenyl-3- [2- (Tetrahydropyran-2-yloxy) ethoxy] Azetidine

Figure 2020071550
Figure 2020071550

ベンズヒドリルアゼチジン-3-オール 20.0 g (83.6 mmol) をジメチルホルムアミド 150 mLに溶解し、氷冷下、水素化ナトリウム (P=60%) 5.02 g (125 mmol) を分割添加後、同温にて撹拌した。2-(2-ブロモエトキシ)テトラヒドロピラン 18.9 mL (125 mmol) を滴下し、60℃ にて30分間撹拌した。水素化ナトリウム (P=60%) 2.00 g (50 mmol) を追加し、さらに12時間撹拌後、室温に放冷した。反応液に水を加え、酢酸エチルにて2回抽出した。有機層を合わせ、水および飽和食塩水にて洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をシリカゲルカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、6:1→3:1、V/V) にて精製した。目的分画の溶媒を減圧下留去し、1-ベンズヒドリル-3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジンの淡褐色油状物 23.8 g、(収率:77%) を得た。
メタノール 200 mLにPd(OH)2-C 0.90 gを懸濁し、1-ベンズヒドリル-3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン17.7 g (48.2 mmol) を加え、室温、0.3 MPaにて46時間接触水素添加した。Pd(OH)2-Cをろ別後、減圧下溶媒を留去した。得られた残渣にヨウ化フェニル 7.0 mL (63 mmol)、ヨウ化銅 918 mg (4.82 mmol)、L-プロリン 1.11 g (9.64 mmol)、炭酸カリウム 13.3 g (96.2 mmol) およびジメチルスルホキシド 24 mLを加え、窒素を1分間バブリング後、窒素雰囲気下70℃ にて19時間撹拌した。放冷後、反応液を水 120 mLに注加し、酢酸エチル 100 mLにて2回抽出した。有機層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、5:1→2:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の黄色油状物 7.8 g (収率:55%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.40-1.90 (6H, m), 3.47-3.67 (4H, m), 3.69-3.78 (2H, m), 3.82-3.92 (2H, m), 4.08-4.16 (2H, m), 4.47-4.55 (1H, m), 4.62-4.68 (1H, m), 6.44-6.52 (2H, m), 6.70-6.78 (1H, m), 7.16-7.26 (2H, m).
Dissolve 20.0 g (83.6 mmol) of benzhydryl azetidine-3-ol in 150 mL of dimethylformamide, add 5.02 g (125 mmol) of sodium hydride (P = 60%) in portions under ice-cooling, and then add at the same temperature. Was stirred. 18.9 mL (125 mmol) of 2- (2-bromoethoxy) tetrahydropyran was added dropwise, and the mixture was stirred at 60 ° C. for 30 minutes. Sodium hydride (P = 60%) of 2.00 g (50 mmol) was added, and the mixture was further stirred for 12 hours and allowed to cool to room temperature. Water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by silica gel column chromatography (n-hexane: ethyl acetate, 6: 1 → 3: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 23.8 g of a light brown oil of 1-benzhydryl-3- [2- (tetrahydropyran-2-yloxy) ethoxy] azetidine (yield: 77%). ..
Suspend 0.90 g of Pd (OH) 2- C in 200 mL of methanol, add 17.7 g (48.2 mmol) of 1-benzhydryl-3- [2- (tetrahydropyran-2-yloxy) ethoxy] azetidine, room temperature, 0.3 MPa. Contact hydrogenation was added for 46 hours. After filtering Pd (OH) 2- C, the solvent was distilled off under reduced pressure. To the obtained residue was added 7.0 mL (63 mmol) of phenyl iodide, 918 mg (4.82 mmol) of copper iodide, 1.11 g (9.64 mmol) of L-proline, 13.3 g (96.2 mmol) of potassium carbonate and 24 mL of dimethyl sulfoxide. After bubbling nitrogen for 1 minute, the mixture was stirred at 70 ° C. for 19 hours under a nitrogen atmosphere. After allowing to cool, the reaction solution was poured into 120 mL of water and extracted twice with 100 mL of ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography (n-hexane: ethyl acetate, 5: 1 → 2: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure to give 7.8 g (yield: 55%) of a yellow oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.40-1.90 (6H, m), 3.47-3.67 (4H, m), 3.69-3.78 (2H, m), 3.82-3.92 (2H, m), 4.08 -4.16 (2H, m), 4.47-4.55 (1H, m), 4.62-4.68 (1H, m), 6.44-6.52 (2H, m), 6.70-6.78 (1H, m), 7.16-7.26 (2H, m) m).

(9d) 1-(4-ヨードフェニル)-3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン (9d) 1- (4-iodophenyl) -3- [2- (tetrahydropyran-2-yloxy) ethoxy] azetidine

Figure 2020071550
Figure 2020071550

実施例9 (9c) で製造した化合物 7.8 g (28 mmol) を塩化メチレン 40 mLに溶解し、重曹3.5 g (42 mmol) および水 40 mLを加え、室温にてヨード 7.46 g (29.4 mmol) を1時間かけて分割添加後、さらに1.5時間撹拌した。5% チオ硫酸ナトリウム水40 mLを加え、30分間撹拌後、減圧下塩化メチレンを留去した。残渣を酢酸エチル 200 mLにて抽出後、有機層を5% チオ硫酸ナトリウム水および飽和食塩水にて順次洗浄、乾燥 (Na2SO4) した。減圧下溶媒を留去し、残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、4:1→2:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の黄色油状物 10.34 g (収率:92%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.40-1.90 (6H, m), 3.47-3.65 (4H, m), 3.68-3.75 (2H, m), 3.83-3.91 (2H, m), 4.07 (2H, t, J=7.1 Hz), 4.44-4.52 (1H, m), 4.60-4.65 (1H, m), 6.20-6.25 (2H, m), 7.43-7.47 (2H, m).
7.8 g (28 mmol) of the compound prepared in Example 9 (9c) was dissolved in 40 mL of methylene chloride, 3.5 g (42 mmol) of baking soda and 40 mL of water were added, and 7.46 g (29.4 mmol) of iodine was added at room temperature. After the divided addition over 1 hour, the mixture was further stirred for 1.5 hours. 40 mL of 5% sodium thiosulfate water was added, and the mixture was stirred for 30 minutes, and then methylene chloride was distilled off under reduced pressure. The residue was extracted with 200 mL of ethyl acetate, and the organic layer was washed successively with 5% sodium thiosulfate solution and saturated brine, and dried (Na 2 SO 4 ). The solvent was evaporated under reduced pressure, and the residue was purified by column chromatography (n-hexane: ethyl acetate, 4: 1 → 2: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure to give 10.34 g (yield: 92%) of a yellow oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.40-1.90 (6H, m), 3.47-3.65 (4H, m), 3.68-3.75 (2H, m), 3.83-3.91 (2H, m), 4.07 (2H, t, J = 7.1 Hz), 4.44-4.52 (1H, m), 4.60-4.65 (1H, m), 6.20-6.25 (2H, m), 7.43-7.47 (2H, m).

(9e) 1-[4-(4,4,5,5-テトラメチル-1,3,2-ジオキサボロラン-2-イル)フェニル]-3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン (9e) 1- [4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) phenyl] -3- [2- (tetrahydropyran-2-yloxy) ethoxy] Azetidine

Figure 2020071550
Figure 2020071550

実施例9 (9d) で製造した化合物2.50 g (6.20 mmol)、[1,1'-ビス(ジフェニルホスフィノ)フェロセン]パラジウム(II)ジクロリド ジクロロメタン付加物 101 mg (0.124 mmol)、4,4,5,5-テトラメチル-1,3,2-ジオキサボロラン1.35 mL (9.30 mmol) を1,4-ジオキサン 20 mLに懸濁させ、トリエチルアミン 4.32 mL (31.0 mmol) を加え、窒素雰囲気下80℃ にて40分間撹拌した。反応液に水100 mLを加え、酢酸エチルにて2回抽出した。有機層を合わせ、飽和食塩水にて洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→5:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の微黄色油状物 1.83 g (収率:73%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.29-1.78 (18H, m), 3.46-3.54 (1H, m), 3.55-3.66 (3H, m), 3.73-3.81 (2H, m), 3.82-3.91 (2H, m), 4.08-4.17 (2H, m), 4.46-4.54 (1H, m), 4.59-4.65 (1H, m), 6.37-6.45 (2H, m).
Compound 2.50 g (6.20 mmol) prepared in Example 9 (9d), [1,1'-bis (diphenylphosphino) ferrocene] palladium (II) dichloride dichloromethane adduct 101 mg (0.124 mmol), 4,4, Suspend 1,35 mL (9.30 mmol) of 5,5-tetramethyl-1,3,2-dioxaborolane in 20 mL of 1,4-dioxane, add 4.32 mL (31.0 mmol) of triethylamine, and at 80 ° C. under a nitrogen atmosphere. Stirred for 40 minutes. 100 mL of water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 5: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure to give 1.83 g (yield: 73%) of a slightly yellow oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.29-1.78 (18H, m), 3.46-3.54 (1H, m), 3.55-3.66 (3H, m), 3.73-3.81 (2H, m), 3.82 -3.91 (2H, m), 4.08-4.17 (2H, m), 4.46-4.54 (1H, m), 4.59-4.65 (1H, m), 6.37-6.45 (2H, m).

(9f) 6,7-ジメトキシ-1-メチル-4-(4-{3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン-1-イル}フェニル)-1H-キノリン-2-オン (9f) 6,7-Dimethoxy-1-methyl-4- (4- {3- [2- (tetrahydropyran-2-yloxy) ethoxy] azetidine-1-yl} phenyl) -1H-quinoline-2-one

Figure 2020071550
Figure 2020071550

実施例9 (9b) で製造した化合物 200 mg (0.788 mmol)、実施例9 (9e) で製造した化合物 381 mg (0.946 mmol)、テトラキス(トリフェニルホスフィン)パラジウム(0) 92 mg (0.079 mmol)、炭酸セシウム 515 mg (1.58 mmol) を1,4-ジオキサン 8 mLに懸濁させ、窒素雰囲気下80℃ にて16時間撹拌した。反応液に水 80 mLを加え、酢酸エチルにて3回抽出した。有機層を合わせ、飽和食塩水にて洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(クロロホルム:酢酸エチル、1:1→1:2→1:3、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の褐色油状物 123 mg (収率:32%) を得た。Compound 200 mg (0.788 mmol) prepared in Example 9 (9b), compound 381 mg (0.946 mmol) prepared in Example 9 (9e), tetrakis (triphenylphosphine) palladium (0) 92 mg (0.079 mmol). , 515 mg (1.58 mmol) of cesium carbonate was suspended in 8 mL of 1,4-dioxane, and the mixture was stirred at 80 ° C. for 16 hours under a nitrogen atmosphere. 80 mL of water was added to the reaction mixture, and the mixture was extracted 3 times with ethyl acetate. The organic layers were combined, washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (chloroform: ethyl acetate, 1: 1 → 1: 2 → 1: 3, V / V). The solvent of the target fraction was evaporated under reduced pressure to give 123 mg (yield: 32%) of a brown oil of the title compound.

(9) 4-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-6,7-ジメトキシ-1-メチルキノリン-2(1H)-オン (9) 4- {4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -6,7-dimethoxy-1-methylquinoline-2 (1H) -one

Figure 2020071550
Figure 2020071550

実施例9 (9f) で製造した化合物 123 mg (0.294 mmol) をメタノール 2.5 mLに溶解させ、氷冷下8.6 M 塩化水素 イソプロパノール溶液 86 μL (0.75 mmol) を加え、同温にて1時間撹拌した。反応液を飽和重曹水にて中和し、水 50 mLを加え、酢酸エチルにて2回抽出した。有機層を合わせ、水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(クロロホルム:メタノール、100:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の黄色粉末 23 mg (収率:23%) を得た。
IR ν (ATR) cm-1 :1635, 1041
1H-NMR (DMSO-d6, 400 MHz) δ:3.41-3.47 (2H, m), 3.49-3.56 (2H, m), 3.63-3.73 (8H, m), 3.95 (3H, s), 4.09-4.18 (2H, m), 4.43-4.52 (1H, m), 4.60-4.75 (1H, br), 6.28 (1H, s), 6.54-6.61 (2H, m), 7.03 (1H, s), 7.04 (1H, s), 7.29-7.37 (2H, m).
MS m/z: 411 [M+H]+, 433 [M+Na]+, 409 [M-H]-.
123 mg (0.294 mmol) of the compound prepared in Example 9 (9f) was dissolved in 2.5 mL of methanol, 86 μL (0.75 mmol) of an 8.6 M hydrogen chloride isopropanol solution was added under ice-cooling, and the mixture was stirred at the same temperature for 1 hour. .. The reaction mixture was neutralized with saturated aqueous sodium hydrogen carbonate, 50 mL of water was added, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (chloroform: methanol, 100: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 23 mg (yield: 23%) of a yellow powder of the title compound.
IR ν (ATR) cm -1 : 1635, 1041
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 3.41-3.47 (2H, m), 3.49-3.56 (2H, m), 3.63-3.73 (8H, m), 3.95 (3H, s), 4.09- 4.18 (2H, m), 4.43-4.52 (1H, m), 4.60-4.75 (1H, br), 6.28 (1H, s), 6.54-6.61 (2H, m), 7.03 (1H, s), 7.04 ( 1H, s), 7.29-7.37 (2H, m).
MS m / z: 411 [M + H] + , 433 [M + Na] + , 409 [MH] - .

実施例10 Example 10

(10a) 5-tert-ブトキシカルボニルアミノ-2-メトキシイソニコチン酸 (10a) 5-tert-Butyloxycarbonylamino-2-methoxyisonicotinic acid

Figure 2020071550
Figure 2020071550

5-アミノ-2-メトキシピリジン 10.0 g (80.6 mmol) をテトラヒドロフラン 100 mLに溶解し、二炭酸ジ-tert-ブチル 18.5 mL (80.6 mmol) を加え、2時間加熱還流した。減圧下テトラヒドロフランを留去し、残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、2:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、tert-ブチル (6-メトキシピリジン-3-イル)カルバメートの淡褐色油状物(18.2 g、収率:定量的) を得た。
tert-ブチル (6-メトキシピリジン-3-イル)カルバメート 5.00 g (22.3 mmol) を脱水ジエチルエーテル 120 mLに溶解し、N,N,N,N-テトラメチルエチレンジアミン 10 mL (67 mmol) を加え、-78℃ にて2.6 M n-ブチルリチウム n-ヘキサン溶液 26 mL (67 mmol) を滴下した。-20℃ にて2時間撹拌後、-78℃ にて二酸化炭素ガスを吹き込みながら0℃ までゆっくり昇温させた。反応液に水を加え、二層を分離後、水層をジエチルエーテルにて洗浄した。水層に濃塩酸 13 mLを加え、析出物をろ取後、水にて洗浄し、標題化合物の微黄色粉末 2.94 g (収率:49%) を得た。
1H-NMR (DMSO-d6, 400 MHz) δ:1.45 (9H, s), 3.84 (3H, s), 7.12 (1H, s), 8.67 (1H, s), 9.40 (1H, s).
10.0 g (80.6 mmol) of 5-amino-2-methoxypyridine was dissolved in 100 mL of tetrahydrofuran, 18.5 mL (80.6 mmol) of di-tert-butyl dicarbonate was added, and the mixture was heated under reflux for 2 hours. Tetrahydrofuran was distilled off under reduced pressure, and the residue was purified by column chromatography (n-hexane: ethyl acetate, 2: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain a light brown oil (18.2 g, yield: quantitative) of tert-butyl (6-methoxypyridin-3-yl) carbamate.
5.00 g (22.3 mmol) of tert-butyl (6-methoxypyridin-3-yl) carbamate was dissolved in 120 mL of dehydrated diethyl ether, and 10 mL (67 mmol) of N, N, N, N-tetramethylethylenediamine was added. At -78 ° C, 26 mL (67 mmol) of a 2.6 M n-butyllithium n-hexane solution was added dropwise. After stirring at -20 ° C for 2 hours, the temperature was slowly raised to 0 ° C while blowing carbon dioxide gas at -78 ° C. Water was added to the reaction solution, the two layers were separated, and the aqueous layer was washed with diethyl ether. 13 mL of concentrated hydrochloric acid was added to the aqueous layer, and the precipitate was collected by filtration and washed with water to obtain 2.94 g (yield: 49%) of a slightly yellow powder of the title compound.
1 H-NMR (DMSO-d6, 400 MHz) δ: 1.45 (9H, s), 3.84 (3H, s), 7.12 (1H, s), 8.67 (1H, s), 9.40 (1H, s).

(10b) メチル 2-メトキシ-5-メチルアミノイソニコチネート (10b) Methyl 2-methoxy-5-methylaminoisonicotinate

Figure 2020071550
Figure 2020071550

実施例10 (10a) で製造した化合物 6.30 g (23.5 mmol) をN,N-ジメチルホルムアミド 50 mLに溶解し、氷冷下、水素化ナトリウム (P=60%) 2.82 g (71 mmol) を加え、同温にて10分間撹拌後、ヨウ化メチル 7.3 mL (120 mmol) を加え、室温にて30分間撹拌した。反応液に5% クエン酸水を加え、酢酸エチルにて2回抽出した。有機層を合わせ、水および飽和食塩水にて順次洗浄し、乾燥 (Na2SO4) した。減圧下溶媒を留去し、メチル 5-(tert-ブトキシカルボニルメチルアミノ)-2-メトキシイソニコチネートの褐色油状物 8.19 g (収率:定量的) を得た。
メチル 5-(tert-ブトキシカルボニルメチルアミノ)-2-メトキシイソニコチネート 8.19 g (23.5 mmol) をぎ酸 20 mLに溶解し、氷冷下、8.6 M 塩化水素 イソプロパノール溶液 8.2 mL (71 mmol) を加え、同温にて30分間撹拌した。反応液を冷水に注加し、炭酸カリウムにて中和後、酢酸エチルにて2回抽出し、有機層を合わせ、乾燥 (Na2SO4) した。減圧下溶媒を留去し、残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の黄色粉末2.50 g を得た。また、ろ液の溶媒を減圧下留去し、残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、5:1、V/V)にて精製し、さらに標題化合物の黄色粉末 2.02 g を得た。(合計収率:98%)
1H-NMR (CDCl3, 400 MHz) δ:2.93 (3H, m), 3.88 (3H, m), 3.89 (3H, m), 7.18 (1H, s), 7.74 (1H, m).
6.30 g (23.5 mmol) of the compound prepared in Example 10 (10a) was dissolved in 50 mL of N, N-dimethylformamide, and sodium hydride (P = 60%) 2.82 g (71 mmol) was added under ice-cooling. After stirring at the same temperature for 10 minutes, 7.3 mL (120 mmol) of methyl iodide was added, and the mixture was stirred at room temperature for 30 minutes. 5% water citrate was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, and dried (Na 2 SO 4 ). The solvent was distilled off under reduced pressure to obtain 8.19 g (yield: quantitative) of a brown oil of methyl 5- (tert-butoxycarbonylmethylamino) -2-methoxyisonicotinate.
Methyl 5- (tert-butoxycarbonylmethylamino) -2-methoxyisonicotinate 8.19 g (23.5 mmol) was dissolved in 20 mL of formic acid, and 8.6 M hydrogen chloride isopropanol solution 8.2 mL (71 mmol) was added under ice-cooling. In addition, the mixture was stirred at the same temperature for 30 minutes. The reaction mixture was poured into cold water, neutralized with potassium carbonate, extracted twice with ethyl acetate, the organic layers were combined, and dried (Na 2 SO 4 ). The solvent was evaporated under reduced pressure, diisopropyl ether was added to the residue, and the mixture was collected by filtration to give 2.50 g of a yellow powder of the title compound. The solvent of the filtrate was evaporated under reduced pressure, and the residue was purified by column chromatography (n-hexane: ethyl acetate, 5: 1, V / V) to obtain 2.02 g of yellow powder of the title compound. .. (Total yield: 98%)
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.93 (3H, m), 3.88 (3H, m), 3.89 (3H, m), 7.18 (1H, s), 7.74 (1H, m).

(10c) メチル 5-[(2-エトキシカルボニルアセチル)メチルアミノ]-2-メトキシイソニコチネート (10c) Methyl 5-[(2-ethoxycarbonylacetyl) Methylamino] -2-methoxyisonicotinate

Figure 2020071550
Figure 2020071550

実施例10 (10b) で製造した化合物 4.52 g (23.0 mmol) を塩化メチレン 40 mLに溶解し、氷冷下、トリエチルアミン 4.17 mL (29.9 mmol)、エチルマロニルクロリド 3.24 mL (25.3 mmol) を加え、同温にて30分間撹拌後、トリエチルアミン0.64 mL (4.6 mmol)、エチルマロニルクロリド 0.29 mL (2.3 mmol) を追加し、同温にてさらに30分間撹拌した。反応液を水および飽和食塩水にて順次洗浄し、乾燥 (Na2SO4) した。減圧下溶媒を留去し、残渣をカラムクロマトグラフィー(n-ヘキサン:メタノール、9:1→3:7、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の黄色油状物 5.37 g (収率:75
%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.23 (3H, t, J=7.3 Hz), 3.11 (2H, s), 3.20 (3H, m), 3.91 (3H, m), 3.99 (3H, m), 4.12 (2H, q, J=7.3 Hz), 7.24 (1H, s), 8.15 (1H, m).
4.52 g (23.0 mmol) of the compound prepared in Example 10 (10b) was dissolved in 40 mL of methylene chloride, and under ice-cooling, 4.17 mL (29.9 mmol) of triethylamine and 3.24 mL (25.3 mmol) of ethylmalonyl chloride were added. After stirring at warm temperature for 30 minutes, 0.64 mL (4.6 mmol) of triethylamine and 0.29 mL (2.3 mmol) of ethylmalonyl chloride were added, and the mixture was further stirred at the same temperature for 30 minutes. The reaction mixture was washed successively with water and saturated brine, and dried (Na 2 SO 4 ). The solvent was evaporated under reduced pressure, and the residue was purified by column chromatography (n-hexane: methanol, 9: 1 → 3: 7, V / V). The solvent of the target fraction was distilled off under reduced pressure, and 5.37 g of the yellow oil of the title compound (yield: 75).
%) Was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.23 (3H, t, J = 7.3 Hz), 3.11 (2H, s), 3.20 (3H, m), 3.91 (3H, m), 3.99 (3H, m) m), 4.12 (2H, q, J = 7.3 Hz), 7.24 (1H, s), 8.15 (1H, m).

(10d) 4-ヒドロキシ-6-メトキシ-1-メチル-1H-[1,7]ナフチリジン-2-オン (10d) 4-Hydroxy-6-methoxy-1-methyl-1H- [1,7] diazanaphthalene-2-one

Figure 2020071550
Figure 2020071550

実施例10 (10c) で製造した化合物 5.37 g (17.3 mmol) をエタノール 50 mL に懸濁し、室温にてナトリウムエトキシド 2.36 g (34.6 mmol) を分割添加後、エタノール 40 mLを追加し、同温にて1.5時間撹拌した。反応液を10% クエン酸水に注加し、酢酸エチルを加え、不溶物をろ取し、エチル 4-ヒドロキシ-6-メトキシ-1-メチル-2-オキソ-1,2-ジヒドロ-[1,7]ナフチリジン-3-カルボキシレートの黄色粉末 1.80 g を得た。また、ろ液を酢酸エチルにて2回抽出し、有機層を合わせ、水および飽和食塩水にて順次洗浄し、乾燥 (Na2SO4) した。減圧下溶媒を留去し、さらにエチル 4-ヒドロキシ-6-メトキシ-1-メチル-2-オキソ-1,2-ジヒドロ-[1,7]ナフチリジン-3-カルボキシレートの黄色粉末 2.94 g を得た。(合計収率99%)
エチル 4-ヒドロキシ-6-メトキシ-1-メチル-2-オキソ-1,2-ジヒドロ-[1,7]ナフチリジン-3-カルボキシレート 4.73 g (17.0 mmol) に濃塩酸 40 mLおよびトリフルオロ酢酸 40 mLを加え、80℃ にて13時間撹拌した。溶媒を減圧下留去し、得られた残渣を飽和重曹水にて中和後、析出した粉末をろ取し、標題化合物の黄色粉末 1.80 g を得た。また、ろ液を酢酸エチルにて2回抽出し、有機層を合わせ、水および飽和食塩水にて順次洗浄し、乾燥 (Na2SO4) した。減圧下溶媒を留去し、さらに標題化合物の黄色粉末 2.94 g を得た。(合計収率:99%)
1H-NMR (DMSO-d6, 400 MHz) δ:3.54 (3H, s), 3.89 (3H, s), 5.96 (1H, s), 7.08 (1H, s), 8.44 (1H, s), 11.61 (1H, s).
5.37 g (17.3 mmol) of the compound prepared in Example 10 (10c) was suspended in 50 mL of ethanol, 2.36 g (34.6 mmol) of sodium ethoxide was added in portions at room temperature, and then 40 mL of ethanol was added at the same temperature. Was stirred for 1.5 hours. The reaction mixture was poured into 10% aqueous citric acid, ethyl acetate was added, the insoluble material was collected by filtration, and ethyl 4-hydroxy-6-methoxy-1-methyl-2-oxo-1,2-dihydro- [1 , 7] 1.80 g of yellow powder of naphthylidine-3-carboxylate was obtained. The filtrate was extracted twice with ethyl acetate, the organic layers were combined, washed successively with water and saturated brine, and dried (Na 2 SO 4 ). The solvent was distilled off under reduced pressure to obtain 2.94 g of a yellow powder of ethyl 4-hydroxy-6-methoxy-1-methyl-2-oxo-1,2-dihydro- [1,7] naphthylidine-3-carboxylate. rice field. (Total yield 99%)
Ethyl 4-hydroxy-6-methoxy-1-methyl-2-oxo-1,2-dihydro- [1,7] diazanaphthalene-3-carboxylate 4.73 g (17.0 mmol) with 40 mL of concentrated hydrochloric acid and 40 of trifluoroacetic acid mL was added, and the mixture was stirred at 80 ° C. for 13 hours. The solvent was evaporated under reduced pressure, the obtained residue was neutralized with saturated aqueous sodium hydrogen carbonate, and the precipitated powder was collected by filtration to give 1.80 g of a yellow powder of the title compound. The filtrate was extracted twice with ethyl acetate, the organic layers were combined, washed successively with water and saturated brine, and dried (Na 2 SO 4 ). The solvent was distilled off under reduced pressure to obtain 2.94 g of a yellow powder of the title compound. (Total yield: 99%)
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 3.54 (3H, s), 3.89 (3H, s), 5.96 (1H, s), 7.08 (1H, s), 8.44 (1H, s), 11.61 (1H, s).

(10e) 4-クロロ-6-メトキシ-1-メチル-1H-[1,7]ナフチリジン-2-オン (10e) 4-Chloro-6-methoxy-1-methyl-1H- [1,7] diazanaphthalene-2-one

Figure 2020071550
Figure 2020071550

実施例10 (10d) で製造した化合物 964 mg (4.68 mmol) をオキシ塩化リン 6.54 mL (70.1 mmol) に懸濁し、90℃ にて1時間撹拌した。放冷後、反応液に水 100 mLを加え、酢酸エチル、次いでクロロホルムにて抽出した。有機層を合わせ、飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(クロロホルム:メタノール、50:1、V/V)にて精製した。目的分画の溶媒を減圧下留去後、残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の淡黄色粉末 754 mg (収率:72%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:3.71 (3H, s), 4.00 (3H, s), 7.00 (1H, s), 7.24 (1H, s), 8.38 (1H, s).
964 mg (4.68 mmol) of compound prepared in Example 10 (10d) was suspended in 6.54 mL (70.1 mmol) of phosphorus oxychloride and stirred at 90 ° C. for 1 hr. After allowing to cool, 100 mL of water was added to the reaction mixture, and the mixture was extracted with ethyl acetate and then chloroform. The organic layers were combined, washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (chloroform: methanol, 50: 1, V / V). After distilling off the solvent of the target fraction under reduced pressure, diisopropyl ether was added to the residue and collected by filtration to obtain 754 mg (yield: 72%) of a pale yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 3.71 (3H, s), 4.00 (3H, s), 7.00 (1H, s), 7.24 (1H, s), 8.38 (1H, s).

(10) 4-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-6-メトキシ-1-メチル-1H-[1,7]ナフチリジン-2-オン (10) 4- {4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -6-methoxy-1-methyl-1H- [1,7] naphthididine-2-one

Figure 2020071550
Figure 2020071550

実施例10 (10e) で製造した化合物 200 mg (0.890 mmol)、実施例9 (9e) で製造した化合物 432 mg (1.07 mmol) をN,N-ジメチルホルムアミド 4.0 mLに懸濁し、リン酸三カリウム 567 mg (2.67 mmol)、テトラキス(トリフェニルホスフィン)パラジウム(0) 103 mg (0.089 mmol) を加え、窒素雰囲気下90℃ にて80分間撹拌した。放冷後、反応液に水 20 mLを加え、酢酸エチルにて抽出した。有機層を水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、2:1→1:1→0:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、褐色油状物 480 mgを得た。
得られた油状物 480 mgを6.0 M塩酸 2.4 mLに溶解し、室温にて2時間撹拌した。反応液を飽和重曹水にて中和し、クロロホルムにて抽出した。有機層を飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(クロロホルム:メタノール、1:0→50:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の黄色粉末 242 mg (収率:71%) を得た。
IR ν (ATR) cm-1 :3556, 1639, 1610
1H-NMR (CDCl3, 400 MHz) δ:1.85-2.20 (1H, br), 3.55-3.62 (2H, m), 3.75 (3H, s), 3.76-3.82 (2H, m), 3.83 (2H, dd, J=8.3, 4.4 Hz), 3.94 (3H, s), 4.14-4.24 (2H, m), 4.47-4.57 (1H, m), 6.50-6.59 (2H, m), 6.74 (1H, s), 6.98 (1H, s), 7.23-7.32 (2H, m), 8.38 (1H, s).
MS m/z: 382 [M+H]+, 404 [M+Na]+, 380 [M-H]-.
200 mg (0.890 mmol) of the compound prepared in Example 10 (10e) and 432 mg (1.07 mmol) of the compound prepared in Example 9 (9e) were suspended in 4.0 mL of N, N-dimethylformamide and tripotassium phosphate. 567 mg (2.67 mmol) and 103 mg (0.089 mmol) of tetrakis (triphenylphosphine) palladium (0) were added, and the mixture was stirred at 90 ° C. for 80 minutes under a nitrogen atmosphere. After allowing to cool, 20 mL of water was added to the reaction solution, and the mixture was extracted with ethyl acetate. The organic layer was washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 2: 1 → 1: 1 → 0: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 480 mg of a brown oil.
480 mg of the obtained oil was dissolved in 2.4 mL of 6.0 M hydrochloric acid, and the mixture was stirred at room temperature for 2 hours. The reaction mixture was neutralized with saturated aqueous sodium hydrogen carbonate and extracted with chloroform. The organic layer was washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (chloroform: methanol, 1: 0 → 50: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure, diisopropyl ether was added to the residue, and the mixture was collected by filtration to give 242 mg (yield: 71%) of a yellow powder of the title compound.
IR ν (ATR) cm -1 : 3556, 1639, 1610
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.85-2.20 (1H, br), 3.55-3.62 (2H, m), 3.75 (3H, s), 3.76-3.82 (2H, m), 3.83 (2H) , dd, J = 8.3, 4.4 Hz), 3.94 (3H, s), 4.14-4.24 (2H, m), 4.47-4.57 (1H, m), 6.50-6.59 (2H, m), 6.74 (1H, s) ), 6.98 (1H, s), 7.23-7.32 (2H, m), 8.38 (1H, s).
MS m / z: 382 [M + H] + , 404 [M + Na] + , 380 [MH] - .

実施例11 Example 11

(11a) 4-ヒドロキシ-6-メトキシ-2H-クロメン-2-オン (11a) 4-Hydroxy-6-methoxy-2H-chromen-2-one

Figure 2020071550
Figure 2020071550

1-(2-ヒドロキシ-5-メトキシフェニル)エタノン5.00 g (30.1 mmol) をトルエン 50 mLに溶解し、炭酸ジエチル 5.5 mL (45 mmol)およびカリウム tert-ブトキシド 5.10 g (45.5 mmol) を加え、室温にて2時間撹拌後、炭酸ジエチル 5.5 mL (45 mmol) およびカリウム tert-ブトキシド 5.10 g (45.5 mmol) を追加し、60℃ にてさらに6時間撹拌した。氷冷下反応液に6.0 M塩酸17 mL (0.10 mol) および水 50 mLを加え、析出物をろ取した。得られた粉末を水、メタノールおよびジエチルエーテルにて順次洗浄し、標題化合物の淡褐色粉末 4.75 g (収率:82%) を得た。
1H-NMR (DMSO-d6, 400 MHz) δ:3.80 (3H, s), 5.60 (1H, s), 7.18-7.23 (2H, m), 7.29 (1H, d, J=8.5 Hz), 12.42-12.61 (1H, br).
Dissolve 5.00 g (30.1 mmol) of 1- (2-hydroxy-5-methoxyphenyl) etanone in 50 mL of toluene, add 5.5 mL (45 mmol) of diethyl carbonate and 5.10 g (45.5 mmol) of potassium tert-butoxide, and room temperature. After stirring at 60 ° C. for 2 hours, 5.5 mL (45 mmol) of diethyl carbonate and 5.10 g (45.5 mmol) of potassium tert-butoxide were added, and the mixture was further stirred at 60 ° C. for 6 hours. 17 mL (0.10 mol) of 6.0 M hydrochloric acid and 50 mL of water were added to the reaction solution under ice-cooling, and the precipitate was collected by filtration. The obtained powder was washed successively with water, methanol and diethyl ether to obtain 4.75 g (yield: 82%) of a light brown powder of the title compound.
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 3.80 (3H, s), 5.60 (1H, s), 7.18-7.23 (2H, m), 7.29 (1H, d, J = 8.5 Hz), 12.42 -12.61 (1H, br).

(11b) 4-クロロ-6-メトキシ-2H-クロメン-2-オン (11b) 4-Chloro-6-methoxy-2H-chromen-2-one

Figure 2020071550
Figure 2020071550

実施例11 (11a) で製造した化合物 4.64 g (24.1 mmol) をクロロホルム 80 mLに懸濁し、トリフェニルホスフィン 19.0 g (72.4 mmol) および四塩化炭素 7.2 mL (72 mmol) を加え、2時間加熱還流した。放冷後、反応液を減圧下濃縮乾固し、得られた残渣をカラムクロマトグラフィー(n-ヘキサン:クロロホルム、1:1→0:1、V/V)にて精製した。目的分画の溶媒を減圧下留去後、残渣をメタノール 100 mLおよびジイソプロピルエーテル 20 mLにて順次洗浄し、標題化合物の微褐色粉末 2.61 g (収率:51%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:3.89 (3H, s), 6.61 (1H, s), 7.18 (1H, dd, J=9.0, 2.9 Hz), 7.27 (1H, d, J=2.9 Hz), 7.29 (1H, d, J=9.0 Hz).
4.64 g (24.1 mmol) of the compound prepared in Example 11 (11a) was suspended in 80 mL of chloroform, 19.0 g (72.4 mmol) of triphenylphosphine and 7.2 mL (72 mmol) of carbon tetrachloride were added, and the mixture was heated under reflux for 2 hours. bottom. After allowing to cool, the reaction mixture was concentrated to dryness under reduced pressure, and the obtained residue was purified by column chromatography (n-hexane: chloroform, 1: 1 → 0: 1, V / V). After distilling off the solvent of the target fraction under reduced pressure, the residue was washed successively with 100 mL of methanol and 20 mL of diisopropyl ether to obtain 2.61 g (yield: 51%) of a light brown powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 3.89 (3H, s), 6.61 (1H, s), 7.18 (1H, dd, J = 9.0, 2.9 Hz), 7.27 (1H, d, J = 2.9) Hz), 7.29 (1H, d, J = 9.0 Hz).

(11c) 6-メトキシ-4-(4-{3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン-1-イル}フェニル)-2H-クロメン-2-オン (11c) 6-Methoxy-4- (4- {3- [2- (tetrahydropyran-2-yloxy) ethoxy] azetidine-1-yl} phenyl) -2H-chromen-2-one

Figure 2020071550
Figure 2020071550

実施例9 (9e) で製造した化合物 415 mg (1.03 mmol) をN,N-ジメチルホルムアミド 4 mLに溶解し、実施例11 (11b) で製造した化合物 217 mg (1.03 mmol)、リン酸三カリウム 656 mg (3.09 mmol) およびテトラキス(トリフェニルホスフィン)パラジウム(0) 119 mg (0.103 mmol) を加え、窒素雰囲気下90℃ にて1時間撹拌した。放冷後反応液を酢酸エチルにて希釈し、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) 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(n-ヘキサン:酢酸エチル、9:1→1:4、V/V)にて精製した。目的分画の溶媒を減圧下留去後、残渣をメタノール 100 mLおよびジイソプロピルエーテル 20 mLにて順次洗浄し、標題化合物の褐色油状物 250 mg (収率:54%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.50-1.92 (6H, m), 3.48-3.55 (1H, m), 3.59-3.71 (3H, m), 3.76 (3H, s), 3.83-3.93 (4H, m), 4.17-4.22 (2H, m), 4.52-4.58 (1H, m), 4.62-4.66 (1H, m), 6.31 (1H, s), 6.52-6.57 (2H, m), 7.07-7.12 (2H, m), 7.30-7.36 (3H, m).
Compound 415 mg (1.03 mmol) prepared in Example 9 (9e) was dissolved in 4 mL of N, N-dimethylformamide, and compound 217 mg (1.03 mmol) prepared in Example 11 (11b), tripotassium phosphate. 656 mg (3.09 mmol) and tetrakis (triphenylphosphine) palladium (0) 119 mg (0.103 mmol) were added, and the mixture was stirred at 90 ° C. for 1 hour under a nitrogen atmosphere. After allowing to cool, the reaction solution was diluted with ethyl acetate, washed successively with water and saturated brine, and dried (Na 2 SO 4 ). The solvent was evaporated under reduced pressure, and the obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 9: 1 → 1: 4, V / V). After distilling off the solvent of the target fraction under reduced pressure, the residue was washed successively with 100 mL of methanol and 20 mL of diisopropyl ether to obtain 250 mg (yield: 54%) of a brown oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.50-1.92 (6H, m), 3.48-3.55 (1H, m), 3.59-3.71 (3H, m), 3.76 (3H, s), 3.83-3.93 (4H, m), 4.17-4.22 (2H, m), 4.52-4.58 (1H, m), 4.62-4.66 (1H, m), 6.31 (1H, s), 6.52-6.57 (2H, m), 7.07 -7.12 (2H, m), 7.30-7.36 (3H, m).

(11) 4-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-6-メトキシ-2H-クロメン-2-オン (11) 4- {4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -6-methoxy-2H-chromen-2-one

Figure 2020071550
Figure 2020071550

実施例11 (11c) で製造した化合物 245 mg (0.543 mmol) をメタノール 4 mLに懸濁し、氷冷下テトラヒドロフラン 1 mLおよび8.6 M 塩化水素 イソプロパノール溶液 0.19 mL (1.6 mmol) を加え、同温にて0.5時間撹拌した。反応液に飽和重曹水を加え、pH を8-9とし、減圧下メタノールを留去後、残渣に水を加え、クロロホルムにて2回抽出した。有機層を合わせ、乾燥 (Na2SO4) 後、減圧下溶媒を留去し、残渣をカラムクロマトグラフィー(クロロホルム:メタノール、1:0→93:7、V/V)にて精製した。目的分画の溶媒を減圧下留去し、残渣を再度カラムクロマトグラフィー(n-ヘキサン:酢酸エチル、1:1→1:3→0:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、残渣にジイソプロピルエーテル 5 mLを加え、ろ取し、標題化合物の淡黄色粉末 99 mg (収率:50%) を得た。
IRν (ATR) cm-1 :3465, 1709, 1685
1H-NMR (DMSO-d6, 400 MHz) δ:3.43-3.48 (2H, m), 3.50-3.56 (2H, m), 3.70-3.75 (5H, m), 4.13-4.19 (2H, m), 4.45-4.52 (1H, m), 4.63-4.70 (1H, br), 6.30 (1H, s), 6.56-6.61 (2H, m), 7.04 (1H, d, J=2.9 Hz), 7.26 (1H, dd, J=9.0, 2.9 Hz), 7.40-7.46 (3H, m).
MS m/z: 368 [M+H]+, 366 [M-H]-.
245 mg (0.543 mmol) of the compound prepared in Example 11 (11c) was suspended in 4 mL of methanol, 1 mL of tetrahydrofuran and 8.6 M hydrogen chloride isopropanol solution 0.19 mL (1.6 mmol) were added under ice-cooling, and the temperature was the same. Stirred for 0.5 hours. Saturated aqueous sodium hydrogen carbonate was added to the reaction mixture to adjust the pH to 8-9, methanol was distilled off under reduced pressure, water was added to the residue, and the mixture was extracted twice with chloroform. The organic layers were combined, dried (Na 2 SO 4 ), the solvent was evaporated under reduced pressure, and the residue was purified by column chromatography (chloroform: methanol, 1: 0 → 93: 7, V / V). The solvent of the target fraction was evaporated under reduced pressure, and the residue was purified again by column chromatography (n-hexane: ethyl acetate, 1: 1 → 1: 3 → 0: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure, 5 mL of diisopropyl ether was added to the residue, and the mixture was collected by filtration to give 99 mg (yield: 50%) of a pale yellow powder of the title compound.
IRν (ATR) cm -1 : 3465, 1709, 1685
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 3.43-3.48 (2H, m), 3.50-3.56 (2H, m), 3.70-3.75 (5H, m), 4.13-4.19 (2H, m), 4.45-4.52 (1H, m), 4.63-4.70 (1H, br), 6.30 (1H, s), 6.56-6.61 (2H, m), 7.04 (1H, d, J = 2.9 Hz), 7.26 (1H, s) dd, J = 9.0, 2.9 Hz), 7.40-7.46 (3H, m).
MS m / z: 368 [M + H] + , 366 [MH] - .

実施例12 Example 12

(12a) 4-(1-ベンズヒドリルアゼチジン-3-イル)モルホリン (12a) 4- (1-Benz Hydrill Azetidine-3-yl) Morpholine

Figure 2020071550
Figure 2020071550

1-ベンズヒドリルアゼチジン-3-オール 5.00 g (20.9 mmol) を塩化メチレン 40 mLに溶解し、氷冷下トリエチルアミン 4.38 mL (31.4 mmol)、塩化メタンスルホニル 1.94 mL (25.1 mmol) を加え、同温にて30分間撹拌した。反応液に水 200 mLを加え、酢酸エチルにて2回抽出した。有機層を飽和食塩水にて洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。
得られた残渣をN,N-ジメチルホルムアミド 40 mLに溶解し、モルホリン 3.65 mL (41.8 mmol)、炭酸カリウム 5.78 g (41.8 mmol) を加え、70℃ にて20時間、室温にて42時間撹拌した。反応液に水 200 mLを加え、酢酸エチルにて2回抽出し、有機層を合わせ、水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、3:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の淡黄色油状物 5.51 g (収率:85%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.26-2.33 (4H, m), 2.87-3.02 (3H, m), 3.36-3.43 (2H, m), 3.68-3.73 (4H, m), 4.41 (1H, s), 7.16-7.43 (10H, m).
5.00 g (20.9 mmol) of 1-benzhydryl azetidine-3-ol was dissolved in 40 mL of methylene chloride, and 4.38 mL (31.4 mmol) of triethylamine and 1.94 mL (25.1 mmol) of methanesulfonyl chloride were added under ice-cooling. The mixture was stirred at warm temperature for 30 minutes. 200 mL of water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layer was washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure.
The obtained residue was dissolved in 40 mL of N, N-dimethylformamide, 3.65 mL (41.8 mmol) of morpholine and 5.78 g (41.8 mmol) of potassium carbonate were added, and the mixture was stirred at 70 ° C. for 20 hours and at room temperature for 42 hours. .. 200 mL of water was added to the reaction solution, the mixture was extracted twice with ethyl acetate, the organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. .. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 3: 1, V / V), and the solvent of the target fraction was distilled off under reduced pressure to obtain 5.51 g of a pale yellow oil of the title compound. (Yield: 85%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.26-2.33 (4H, m), 2.87-3.02 (3H, m), 3.36-3.43 (2H, m), 3.68-3.73 (4H, m), 4.41 (1H, s), 7.16-7.43 (10H, m).

(12b) 4-[1-(4-ニトロフェニル)アゼチジン-3-イル]モルホリン (12b) 4- [1- (4-Nitrophenyl) Azetidine-3-yl] Morpholine

Figure 2020071550
Figure 2020071550

メタノール 100 mLにアルゴン雰囲気下 20%Pd(OH)2-C (wet.) 800 mgおよび実施例12 (12a) で製造した化合物 5.50 g (17.8 mmol) を加え、0.4 MPa、室温にて3.5時間接触水素添加した。Pd(OH)2-Cをセライトにてろ別し、減圧下溶媒を留去した。
得られた残渣および1-フルオロ-4-ニトロベンゼン 3.27 g (23.1 mmol) をN,N-ジメチルホルムアミド 30 mLに溶解させ、炭酸カリウム 3.69 g (26.7 mmol) を加え、70℃ にて12時間撹拌した。反応液に水 200 mLを加え、酢酸エチルにて2回抽出した。有機層を合わせ、水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の黄色粉末 3.10 g (収率:66%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.41-2.52 (4H, m), 3.35-3.41 (1H, m), 3.72-3.81 (4H, m), 3.86-3.92 (2H, m), 4.07-4.11 (2H, m), 6.29-6.33 (2H, m), 8.08-8.12 (2H, m).
To 100 mL of methanol, 800 mg of 20% Pd (OH) 2- C (wet.) And 5.50 g (17.8 mmol) of the compound prepared in Example 12 (12a) were added under an argon atmosphere, and 0.4 MPa was added at room temperature for 3.5 hours. Contact hydrogenation was added. Pd (OH) 2- C was filtered off with Celite, and the solvent was distilled off under reduced pressure.
The obtained residue and 1.27 g (23.1 mmol) of 1-fluoro-4-nitrobenzene were dissolved in 30 mL of N, N-dimethylformamide, 3.69 g (26.7 mmol) of potassium carbonate was added, and the mixture was stirred at 70 ° C. for 12 hours. .. 200 mL of water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. Diisopropyl ether was added to the obtained residue and collected by filtration to obtain 3.10 g (yield: 66%) of a yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.41-2.52 (4H, m), 3.35-3.41 (1H, m), 3.72-3.81 (4H, m), 3.86-3.92 (2H, m), 4.07 -4.11 (2H, m), 6.29-6.33 (2H, m), 8.08-8.12 (2H, m).

(12c) (2-メトキシ-5-ニトロピリジン-4-イル)-[4-(3-(モルホリン-4-イル)アゼチジン-1-イル)フェニル]アミン (12c) (2-Methoxy-5-nitropyridin-4-yl)-[4- (3- (morpholine-4-yl) azetidine-1-yl) phenyl] amine

Figure 2020071550
Figure 2020071550

メタノール-テトラヒドロフラン (7:1) の混液80 mLにアルゴン雰囲気下 10% Pd(OH)2-C (wet.) 230 mgおよび実施例12 (12b) で製造した化合物 1.40 g (5.32 mmol) を加え、0.4 MPaにて35分間接触水素添加した。Pd(OH)2-Cをセライトにてろ別し、減圧下溶媒を留去した。
得られた残渣および4-クロロ-2-メトキシ-5-ニトロピリジン1.20 g (6.38 mmol) をN,N-ジメチルホルムアミド15 mLに溶解し、ジイソプロピルエチルアミン 1.85 mL (10.6 mmol) を加え、70℃ にて3.5時間撹拌した。反応液に水 150 mLを加え、酢酸エチルにて2回抽出した。有機層を合わせ、水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(クロロホルム:酢酸エチル、1:0→1:2、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の橙色粉末 1.81 g (収率:88%) を得た。
1H-NMR (CDCl3, 400 MHz)δ:2.41-2.52 (4H, m), 3.30-3.39 (1H, m), 3.72-3.81 (6H, m), 3. 90 (3H, s), 4.07-4.11 (2H, m), 6.06 (1H, s), 6.45-6.50 (2H, m), 7.04-7.09 (2H, m), 9.02 (1H, s), 9.27 (1H, s).
To 80 mL of a mixed solution of methanol-tetrahydrofuran (7: 1 ), add 230 mg of 10% Pd (OH) 2- C (wet.) Under an argon atmosphere and 1.40 g (5.32 mmol) of the compound prepared in Example 12 (12b). , 0.4 MPa was contact hydrogenated for 35 minutes. Pd (OH) 2- C was filtered off with Celite, and the solvent was distilled off under reduced pressure.
Dissolve the obtained residue and 1.20 g (6.38 mmol) of 4-chloro-2-methoxy-5-nitropyridine in 15 mL of N, N-dimethylformamide, add 1.85 mL (10.6 mmol) of diisopropylethylamine, and bring to 70 ° C. Stirred for 3.5 hours. 150 mL of water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (chloroform: ethyl acetate, 1: 0 → 1: 2, V / V), the solvent for the target fraction was distilled off under reduced pressure, and 1.81 g of the orange powder of the title compound was distilled off. (Yield: 88%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.41-2.52 (4H, m), 3.30-3.39 (1H, m), 3.72-3.81 (6H, m), 3.90 (3H, s), 4.07 -4.11 (2H, m), 6.06 (1H, s), 6.45-6.50 (2H, m), 7.04-7.09 (2H, m), 9.02 (1H, s), 9.27 (1H, s).

(12) 7-メトキシ-4-メチル-1-[4-(3-(モルホリン-4-イル)アゼチジン-1-イル)フェニル]-1,4-ジヒドロ-2H-ピリド[3,4-b]ピラジン-3-オン (12) 7-Methoxy-4-methyl-1- [4- (3- (morpholine-4-yl) azetidine-1-yl) phenyl] -1,4-dihydro-2H-pyrido [3,4-b ] Pyrazine-3-on

Figure 2020071550
Figure 2020071550

メタノール-テトラヒドロフラン (9:1) の混液100 mLにアルゴン雰囲気下 10% Pd-C 250 mgおよび実施例12 (12c) で製造した化合物 1.78 g (4.62 mmol) を加え、0.4 MPa、室温にて45分間接触水素添加した。Pd-Cをセライトにてろ別し、減圧下溶媒を留去した。
得られた残渣をテトラヒドロフラン (脱水) 10 mLに溶解し、氷冷下トリエチルアミン 966 μL (6.93 mmol)、塩化クロロアセチル 368μL (4.62 mmol) を順次加え、同温にて1時間撹拌後、トリエチルアミン 644μL (4.62 mmol)、塩化クロロアセチル 200μL (2.51 mmol) を順次追加し、同温にて30分間撹拌後、室温にてさらに30分間撹拌した。反応液に酢酸エチル 20 mL、水 100 mLを加え、二層を分離後、水層を酢酸エチルにて抽出した。有機層を合わせ、飽和食塩水にて洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。
得られた残渣をN,N-ジメチルホルムアミド 10 mLに溶解し、氷冷下水素化ナトリウム (P=60%) 408 mg (10 mmol) を加え、同温にて80分間撹拌した。酢酸エチル 40 mL、水 100 mLを加え、二層を分離後、水層を酢酸エチルにて2回抽出した。有機層を合わせ、水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。
得られた残渣をN,N-ジメチルホルムアミド (脱水) 10 mLに溶解させ、氷冷下、水素化ナトリウム (P=60%) 222 mg (5.5 mmol) を加え、同温にて15分間撹拌後、ヨウ化メチル 288μL (4.62 mmol) を加え、同温にて30分間撹拌した。反応液に酢酸エチル 20 mL、水 100 mLを加え、二層を分離後、水層を酢酸エチルにて2回抽出した。有機層を合わせ、飽和食塩水にて洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(クロロホルム:メタノール、1:0→98:2、V/V)にて精製後、目的分画の溶媒を減圧下留去した。得られた残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の白色粉末 715 mg (収率:38%) を得た。
IRν (ATR) cm-1 :1676
1H-NMR (DMSO-d6, 400 MHz) δ:2.30-2.40 (4H, m), 3.26-3.31 (1H, m), 3.32 (3H, s), 3.58-3.67 (6H, m), 3.69 (3H, s), 3.90-3.97 (2H, m), 4.21 (2H, s), 5.51 (1H, s), 6.48-6.53 (2H, m), 7.12-7.16 (2H, m), 7.72 (1H, s).
MS m/z: 410 [M+H]+.
To 100 mL of a mixed solution of methanol-tetrahydrofuran (9: 1), 250 mg of 10% Pd-C under an argon atmosphere and 1.78 g (4.62 mmol) of the compound prepared in Example 12 (12c) were added, and 0.4 MPa at room temperature 45. Contact hydrogenation was added for minutes. Pd-C was filtered off with Celite, and the solvent was distilled off under reduced pressure.
The obtained residue is dissolved in 10 mL of tetrahydrofuran (dehydrated), 966 μL (6.93 mmol) of triethylamine and 368 μL (4.62 mmol) of chloroacetyl chloride are sequentially added under ice-cooling, and the mixture is stirred at the same temperature for 1 hour, and then 644 μL of triethylamine (644 μL). 4.62 mmol) and 200 μL (2.51 mmol) of chloroacetyl chloride were sequentially added, and the mixture was stirred at the same temperature for 30 minutes and then at room temperature for another 30 minutes. 20 mL of ethyl acetate and 100 mL of water were added to the reaction mixture, the two layers were separated, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure.
The obtained residue was dissolved in 10 mL of N, N-dimethylformamide, 408 mg (10 mmol) of sodium hydride (P = 60%) was added under ice-cooling, and the mixture was stirred at the same temperature for 80 minutes. 40 mL of ethyl acetate and 100 mL of water were added, the two layers were separated, and the aqueous layer was extracted twice with ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure.
The obtained residue is dissolved in 10 mL of N, N-dimethylformamide (dehydrated), 222 mg (5.5 mmol) of sodium hydride (P = 60%) is added under ice-cooling, and the mixture is stirred at the same temperature for 15 minutes. , 288 μL (4.62 mmol) of methyl iodide was added, and the mixture was stirred at the same temperature for 30 minutes. 20 mL of ethyl acetate and 100 mL of water were added to the reaction mixture, the two layers were separated, and the aqueous layer was extracted twice with ethyl acetate. The organic layers were combined, washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (chloroform: methanol, 1: 0 → 98: 2, V / V), and the solvent for the target fraction was distilled off under reduced pressure. Diisopropyl ether was added to the obtained residue and collected by filtration to obtain 715 mg (yield: 38%) of a white powder of the title compound.
IRν (ATR) cm -1 : 1676
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 2.30-2.40 (4H, m), 3.26-3.31 (1H, m), 3.32 (3H, s), 3.58-3.67 (6H, m), 3.69 ( 3H, s), 3.90-3.97 (2H, m), 4.21 (2H, s), 5.51 (1H, s), 6.48-6.53 (2H, m), 7.12-7.16 (2H, m), 7.72 (1H, s) s).
MS m / z: 410 [M + H] + .

実施例13 Example 13

(13a) 6-ヨード-1-メチル-1H-ベンゾ[d][1,3]オキサジン-2,4-ジオン (13a) 6-iodo-1-methyl-1H-benzo [d] [1,3] oxazine-2,4-dione

Figure 2020071550
Figure 2020071550

2-メチルアミノ安息香酸 25.0 g (0.165 mol) を酢酸 150 mLおよび水 300 mLの混液に懸濁し、ヨウ素 42.0 g (0.165 mol) を分割添加後、室温にて3日間撹拌した。析出物をろ取し、水にて洗浄し、5-ヨード-2-メチルアミノ安息香酸を含む黒紫色粉末 37.7 gを得た。
得られた黒紫色粉末 37.7 gを水 400 mLに懸濁し、炭酸ナトリウム 14.5 g (0.137 mol) を加え、氷冷下トリホスゲン 13.5 g (45.5 mmol) のトルエン 65 mLの溶液を滴下し、同温にて2時間撹拌した。析出物をろ取し、得られた粉末を水 150 mL、エタノール-ジエチルエーテル (1:1) の混液150 mL、次いでジエチルエーテル 50 mLにて順次洗浄し、標題化合物の褐色粉末 17.3 g (収率:35%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:3.43 (3H, s), 7.26 (1H, d, J=8.8 Hz), 8.13 (1H, dd, J=8.8, 1.7 Hz), 8.20 (1H, d, J=1.7 Hz).
25.0 g (0.165 mol) of 2-methylaminobenzoic acid was suspended in a mixed solution of 150 mL of acetic acid and 300 mL of water, 42.0 g (0.165 mol) of iodine was added in portions, and the mixture was stirred at room temperature for 3 days. The precipitate was collected by filtration and washed with water to obtain 37.7 g of a black-purple powder containing 5-iodo-2-methylaminobenzoic acid.
Suspend 37.7 g of the obtained black-purple powder in 400 mL of water, add 14.5 g (0.137 mol) of sodium carbonate, add a solution of 13.5 g (45.5 mmol) of triphosgene in 65 mL of toluene under ice-cooling, and bring to the same temperature. Stirred for 2 hours. The precipitate was collected by filtration, and the obtained powder was washed successively with 150 mL of water, 150 mL of a mixed solution of ethanol-diethyl ether (1: 1), and then 50 mL of diethyl ether, and 17.3 g of brown powder of the title compound (yield). Rate: 35%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 3.43 (3H, s), 7.26 (1H, d, J = 8.8 Hz), 8.13 (1H, dd, J = 8.8, 1.7 Hz), 8.20 (1H, 1H, d, J = 1.7 Hz).

(13b) 4-クロロ-1-メチル-2-オキソ-1,2-ジヒドロキノリン-6-カルボニトリル (13b) 4-Chloro-1-methyl-2-oxo-1,2-dihydroquinoline-6-carbonitrile

Figure 2020071550
Figure 2020071550

マロン酸ジエチル 40 mL (0.26 mol) をN,N-ジメチルホルムアミド 150 mLに溶解し、水素化ナトリウム (P=60%) 2.74 g (69 mmol) を分割添加し、室温にて10分間撹拌後、実施例13 (13a) で製造した化合物 (3) 17.3 g (57.1 mmol) のN,N-ジメチルホルムアミド 50 mLの溶液を滴下し、120℃ にて2.5時間撹拌した。放冷後、反応液に酢酸エチル 200 mLを加え、不溶物をろ取し、得られた粉末を水 4.5 Lに溶解後、6.0 M塩酸にて溶液のpHを2-3に調整した。析出物をろ取し、水、エタノール-ジエチルエーテル (1:1) の混液、次いでジエチルエーテルにて順次洗浄し、淡灰色粉末 14.8 gを得た。
得られた淡灰色粉末 14.8 gをトリフルオロ酢酸 100 mLに懸濁し、濃塩酸 100 mLを加え、80℃ にて24時間撹拌した。減圧下反応液を濃縮乾固し、残渣に水を加え、不溶物をろ取後、水 100 mL、エタノール-ジエチルエーテル (1:1) の混液 50 mL、次いでジイソプロピルエーテル 50 mLにて順次洗浄し、淡灰色粉末 12.4 gを得た。
得られた淡灰色粉末 12.4 gをN,N-ジメチルホルムアミド 150 mLに溶解し、亜鉛ジシアニド 9.32 g (79.4 mmol) およびテトラキス(トリフェニルホスフィン)パラジウム(0) 4.59 g (3.97 mmol) を加え、窒素雰囲気下、90℃ にて2.5時間撹拌した。放冷後、反応液にテトラヒドロフラン 100 mLを加え、不溶物をセライトにてろ過し、ろ液に水 900 mLを加え、析出物をろ取した。得られた粉末にメタノール-テトラヒドロフラン(1:5) の混液2 Lを加え、不溶物をろ別後、ろ液を減圧下濃縮乾固し、残渣に酢酸エチル 30 mLを加え、不溶物をろ取した。得られた粉末を酢酸エチル 5 mLおよびクロロホルム 5 mLにて順次洗浄し、淡褐色粉末 7.25 gを得た。
得られた淡褐色粉末 7.25 gにオキシ塩化リン 70 mLを加え、90℃ にて6時間撹拌した。減圧下反応液を濃縮乾固し、残渣に水を加え、酢酸エチルにて2回抽出した。有機層を合わせ、水にて洗浄後、10% 炭酸カリウム水を加え、析出物をセライトにてろ別した。ろ液の二層を分離し、有機層を飽和食塩水にて洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(クロロホルム:n-ヘキサン、1:1→1:0、V/V)にて2回精製後、目的分画の溶媒を減圧下留去し、残渣に酢酸エチル-ジエチルエーテル (1:3) の混液10 mLを加え、不溶物をろ取した。得られた粉末をジエチルエーテルにて洗浄し、標題化合物の白色粉末 1.27 g (収率:10%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:3.75 (3H, s), 6.98 (1H, s), 7.47 (1H, d, J=8.8 Hz), 7.86 (1H, dd, J=8.8, 2.0 Hz), 8.33 (1H, d, J=2.0 Hz).
Dissolve 40 mL (0.26 mol) of diethyl malonate in 150 mL of N, N-dimethylformamide, add 2.74 g (69 mmol) of sodium hydride (P = 60%) in portions, and stir at room temperature for 10 minutes. A solution of 17.3 g (57.1 mmol) of compound (3) prepared in Example 13 (13a) in 50 mL of N, N-dimethylformamide was added dropwise, and the mixture was stirred at 120 ° C. for 2.5 hours. After allowing to cool, 200 mL of ethyl acetate was added to the reaction solution, the insoluble material was collected by filtration, the obtained powder was dissolved in 4.5 L of water, and the pH of the solution was adjusted to 2-3 with 6.0 M hydrochloric acid. The precipitate was collected by filtration and washed with water, a mixed solution of ethanol-diethyl ether (1: 1), and then with diethyl ether to obtain 14.8 g of a light gray powder.
14.8 g of the obtained light gray powder was suspended in 100 mL of trifluoroacetic acid, 100 mL of concentrated hydrochloric acid was added, and the mixture was stirred at 80 ° C. for 24 hours. The reaction solution is concentrated to dryness under reduced pressure, water is added to the residue, the insoluble material is collected by filtration, and then washed sequentially with 100 mL of water, 50 mL of a mixed solution of ethanol-diethyl ether (1: 1), and then 50 mL of diisopropyl ether. And 12.4 g of light gray powder was obtained.
12.4 g of the obtained light gray powder was dissolved in 150 mL of N, N-dimethylformamide, 9.32 g (79.4 mmol) of zinc disianide and 4.59 g (3.97 mmol) of tetrakis (triphenylphosphine) palladium (0) were added, and nitrogen was added. The mixture was stirred at 90 ° C. for 2.5 hours in an atmosphere. After allowing to cool, 100 mL of tetrahydrofuran was added to the reaction solution, the insoluble material was filtered through Celite, 900 mL of water was added to the filtrate, and the precipitate was collected by filtration. Add 2 L of a mixed solution of methanol-tetrahydrofuran (1: 5) to the obtained powder, filter out the insoluble material, concentrate and dry the filtrate under reduced pressure, add 30 mL of ethyl acetate to the residue, and filter out the insoluble material. I took it. The obtained powder was washed successively with 5 mL of ethyl acetate and 5 mL of chloroform to obtain 7.25 g of a light brown powder.
To 7.25 g of the obtained light brown powder, 70 mL of phosphorus oxychloride was added, and the mixture was stirred at 90 ° C. for 6 hours. The reaction mixture was concentrated to dryness under reduced pressure, water was added to the residue, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed with water, 10% potassium carbonate water was added, and the precipitate was filtered off with Celite. The two layers of the filtrate were separated, the organic layer was washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified twice by column chromatography (chloroform: n-hexane, 1: 1 → 1: 0, V / V), the solvent for the target fraction was distilled off under reduced pressure, and ethyl acetate was added to the residue. -10 mL of a mixed solution of diethyl ether (1: 3) was added, and the insoluble material was collected by filtration. The obtained powder was washed with diethyl ether to obtain 1.27 g (yield: 10%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 3.75 (3H, s), 6.98 (1H, s), 7.47 (1H, d, J = 8.8 Hz), 7.86 (1H, dd, J = 8.8, 2.0) Hz), 8.33 (1H, d, J = 2.0 Hz).

(13c) 4,4,5,5-テトラメチル-2-[4-(2-フェノキシエトキシ)フェニル][1,3,2]ジオキサボロラン (13c) 4,4,5,5-Tetramethyl-2- [4- (2-phenoxyethoxy) phenyl] [1,3,2] dioxaborolane

Figure 2020071550
Figure 2020071550

4-(4,4,5,5-テトラメチル-[1,3,2]ジオキサボロラン-2-イル)フェノール 1.00 g (4.54 mmol) をN,N-ジメチルホルムアミド 20 mLに溶解し、(2-ブロモエトキシ)ベンゼン 1.37 g (6.81 mmol) および炭酸セシウム 2.22 g (6.81 mmol) を加え、90℃ にて3時間撹拌した。放冷後、反応液を酢酸エチルにて希釈し、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、1:0→7:3、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 1.54 g (収率:定量的) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.33 (12H, s), 4.30-4.37 (4H, m), 6.92-6.70 (5H, m), 7.29 (2H, t, J=7.6 Hz), 7.73-7.78 (2H, m).
1.00 g (4.54 mmol) of 4- (4,4,5,5-tetramethyl- [1,3,2] dioxaborolan-2-yl) phenol was dissolved in 20 mL of N, N-dimethylformamide and (2- 1.37 g (6.81 mmol) of bromoethoxy) benzene and 2.22 g (6.81 mmol) of cesium carbonate were added, and the mixture was stirred at 90 ° C. for 3 hours. After allowing to cool, the reaction solution was diluted with ethyl acetate, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography (n-hexane: ethyl acetate, 1: 0 → 7: 3, V / V), the solvent of the target fraction was distilled off under reduced pressure, and 1.54 g of white powder of the title compound ( Yield: Quantitative) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.33 (12H, s), 4.30-4.37 (4H, m), 6.92-6.70 (5H, m), 7.29 (2H, t, J = 7.6 Hz), 7.73-7.78 (2H, m).

(13d) 1-メチル-2-オキソ-4-[4-(2-フェノキシエトキシ)フェニル]-1,2-ジヒドロキノリン-6-カルボニトリル (13d) 1-Methyl-2-oxo-4- [4- (2-phenoxyethoxy) phenyl] -1,2-dihydroquinoline-6-carbonitrile

Figure 2020071550
Figure 2020071550

実施例13 (13b) で製造した化合物 200 mg (0.915 mmol)および実施例13 (13c) で製造した化合物 342mg (1.01 mmol)をN,N-ジメチルホルムアミド 4 mLに溶解し、リン酸三カリウム 583 mg (2.75 mmol) およびテトラキス(トリフェニルホスフィン)パラジウム(0) 106 mg (0.917 mmol) を加え、窒素雰囲気下90℃ にて2時間撹拌した。放冷後反応液を酢酸エチルにて希釈し、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。残渣をカラムクロマトグラフィー(クロロホルム:酢酸エチル、1:0→3:2、V/V)にて精製後、目的分画の溶媒を減圧下留去し、残渣にメタノール 1 mLおよびジエチルエーテル 10 mLを加え、室温にて1時間撹拌した。不溶物をろ取し、標題化合物の白色粉末 333 mg (収率:92%) を得た。
1H-NMR (DMSO-d6, 400 MHz)δ:3.69 (3H, s), 4.34-4.38 (2H, m), 4.40-4.45 (2H, m), 6.62 (1H, s), 6.93-7.02 (3H, m), 7.16-7.20 (2H, m), 7.29-7.34 (2H, m), 7.43-7.47 (2H, m), 7.78 (1H, d, J=9.0 Hz), 7.82 (1H, d, J=2.0 Hz), 8.06 (1H, dd, J=9.0, 2.0 Hz).
200 mg (0.915 mmol) of compound prepared in Example 13 (13b) and 342 mg (1.01 mmol) of compound prepared in Example 13 (13c) were dissolved in 4 mL of N, N-dimethylformamide and tripotassium phosphate 583. Mg (2.75 mmol) and tetrakis (triphenylphosphine) palladium (0) 106 mg (0.917 mmol) were added, and the mixture was stirred at 90 ° C. for 2 hours under a nitrogen atmosphere. After allowing to cool, the reaction solution was diluted with ethyl acetate, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. After purifying the residue by column chromatography (chloroform: ethyl acetate, 1: 0 → 3: 2, V / V), the solvent of the target fraction was distilled off under reduced pressure, and 1 mL of methanol and 10 mL of diethyl ether were added to the residue. Was added, and the mixture was stirred at room temperature for 1 hour. The insoluble material was collected by filtration to give 333 mg (yield: 92%) of a white powder of the title compound.
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 3.69 (3H, s), 4.34-4.38 (2H, m), 4.40-4.45 (2H, m), 6.62 (1H, s), 6.93-7.02 ( 3H, m), 7.16-7.20 (2H, m), 7.29-7.34 (2H, m), 7.43-7.47 (2H, m), 7.78 (1H, d, J = 9.0 Hz), 7.82 (1H, d, J = 2.0 Hz), 8.06 (1H, dd, J = 9.0, 2.0 Hz).

(13) 1-メチル-2-オキソ-4-[4-(2-フェノキシエトキシ)フェニル]-1,2-ジヒドロキノリン-6-カルボキサミド (13) 1-Methyl-2-oxo-4- [4- (2-phenoxyethoxy) phenyl] -1,2-dihydroquinoline-6-carboxamide

Figure 2020071550
Figure 2020071550

実施例13 (13d) で製造した化合物 (5) 325 mg (0.820 mmol) に炭酸カリウム 68 mg (0.49 mmol) および30% 過酸化水素水 130 mg (1.2 mmol) のジメチルスルホキシド 4 mLの溶液を加え、室温にて3.5時間撹拌した。反応液に水 30 mLを加え、析出物をろ取後、エタノール 5 mLにて洗浄し、白色粉末 310 mgを得た。得られた白色粉末 310 mgにエタノール 15 mLを加え、15分間加熱還流後、室温まで放冷し、不溶物をろ取した。得られた粉末をエタノール 10 mLにて洗浄し、標題化合物の灰色粉末 260 mg (収率:77%) を得た。
IR ν (ATR) cm-1:3272, 3182, 1651
1H-NMR (DMSO-d6, 400 MHz) δ:3.70 (3H, s), 4.33-4.39 (2H, m), 4.39-4.46 (2H, m), 6.53 (1H, s), 6.93-7.04 (3H, m), 7.15-7.20 (2H, m), 7.29-7.34 (2H, m), 7.37-7.42 (1H, br), 7.41-7.46 (2H, m), 7.66 (1H, d, J=8.8 Hz), 8.02-8.09 (2H, m), 8.14 (1H, dd, J=8.8, 1.5 Hz).
MS m/z: 437 [M+Na]+, 413 [M-H]-.
Compound (5) prepared in Example 13 (13d) was added with a solution of 68 mg (0.49 mmol) of potassium carbonate and 4 mL of dimethyl sulfoxide of 130 mg (1.2 mmol) of 30% hydrogen peroxide solution to 325 mg (0.820 mmol). , Stirred at room temperature for 3.5 hours. 30 mL of water was added to the reaction solution, and the precipitate was collected by filtration and washed with 5 mL of ethanol to obtain 310 mg of white powder. To 310 mg of the obtained white powder, 15 mL of ethanol was added, and the mixture was heated under reflux for 15 minutes, allowed to cool to room temperature, and the insoluble material was collected by filtration. The obtained powder was washed with 10 mL of ethanol to obtain 260 mg (yield: 77%) of a gray powder of the title compound.
IR ν (ATR) cm -1 : 3272, 3182, 1651
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 3.70 (3H, s), 4.33-4.39 (2H, m), 4.39-4.46 (2H, m), 6.53 (1H, s), 6.93-7.04 ( 3H, m), 7.15-7.20 (2H, m), 7.29-7.34 (2H, m), 7.37-7.42 (1H, br), 7.41-7.46 (2H, m), 7.66 (1H, d, J = 8.8) Hz), 8.02-8.09 (2H, m), 8.14 (1H, dd, J = 8.8, 1.5 Hz).
MS m / z: 437 [M + Na] + , 413 [MH] - .

実施例14 Example 14

(14a) 2-(テトラヒドロピラン-4-イルオキシ)エタノール (14a) 2- (Tetrahydropyran-4-yloxy) Ethanol

Figure 2020071550
Figure 2020071550

水素化リチウムアルミニウム 1.37 g (36.0 mmol) をテトラヒドロフラン 20 mLに懸濁し、氷冷下、4-オキソテトラヒドロピラン 2.00 g (20.0 mmol) のテトラヒドロフラン 10 mLの溶液を滴下し、同温にて40分間撹拌した。反応液に水 1.5 mL、5.0 M 水酸化ナトリウム水 1.5 mL、水 1.5 mLを順次加え、不溶物をろ別した。ろ液を乾燥 (Na2SO4) 後、減圧下溶媒を留去し、テトラヒドロピラン-4-オールの無色油状物 1.81 g (収率:89%) を得た。
テトラヒドロピラン-4-オール 375 mg (3.67 mmol) を N,N-ジメチルホルムアミド 2.5 mL に溶解し、氷冷下水素化ナトリウム (P=60%) 190 mg (4.75 mmol) を加え、室温にて 15分間撹拌した。2-(2-ブロモエトキシ)テトラヒドロピラン 1.0 g (4.8 mmol) を加え、60℃ にて 3時間、室温にて 2 日間撹拌した。酢酸エチルを加え、水次いで飽和食塩水にて順次洗浄し、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→2:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、1-(テトラヒドロピラン-4-イルオキシ)-2-(テトラヒドロピラン-2-イルオキシ)エタンの無色油状物190 mg (収率:22%) を得た。
1-(テトラヒドロピラン-4-イルオキシ)-2-(テトラヒドロピラン-2-イルオキシ)エタン 180 mg (0.782 mmol) をメタノール 2.0 mL に溶解し、8.6 M 塩化水素 イソプロパノール溶液 0.27 mL (2.3 mmol) を加え、室温にて 15分間撹拌した。減圧下溶媒を留去し、標題化合物の無色油状物 110 mg (収率:96%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.54-1.66 (2H, m), 1.84 (1H, s), 1.86-1.97 (2H, m), 3.39-3.49 (2H, m), 3.49-3.57 (1H, m), 3.59 (2H, t, J=4.9 Hz), 3.74 (2H, t, J=4.9 Hz), 3.91-3.99 (2H, m).
1.37 g (36.0 mmol) of lithium aluminum hydride is suspended in 20 mL of tetrahydrofuran, a solution of 2.00 g (20.0 mmol) of 4-oxotetrahydropyran in 10 mL of tetrahydrofuran is added dropwise under ice-cooling, and the mixture is stirred at the same temperature for 40 minutes. bottom. 1.5 mL of water, 1.5 mL of 5.0 M sodium hydroxide water, and 1.5 mL of water were sequentially added to the reaction solution, and the insoluble material was filtered off. After drying the filtrate (Na 2 SO 4 ), the solvent was distilled off under reduced pressure to obtain 1.81 g (yield: 89%) of a colorless oil of tetrahydropyran-4-ol.
Dissolve 375 mg (3.67 mmol) of tetrahydropyran-4-ol in 2.5 mL of N, N-dimethylformamide, add 190 mg (4.75 mmol) of sodium hydride (P = 60%) under ice-cooling, and at room temperature 15 Stir for minutes. 1.0 g (4.8 mmol) of 2- (2-bromoethoxy) tetrahydropyran was added, and the mixture was stirred at 60 ° C. for 3 hours and at room temperature for 2 days. Ethyl acetate was added, and the mixture was washed successively with water and then saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 2: 1, V / V), and the solvent of the target fraction was distilled off under reduced pressure to obtain 1- (tetrahydropyran). A colorless oil of -4-yloxy) -2- (tetrahydropyran-2-yloxy) ethane was obtained in an amount of 190 mg (yield: 22%).
Dissolve 180 mg (0.782 mmol) of 1- (tetrahydropyran-4-yloxy) -2- (tetrahydropyran-2-yloxy) ethane in 2.0 mL of methanol, and add 0.27 mL (2.3 mmol) of 8.6 M hydrogen chloride isopropanol solution. , Stirred for 15 minutes at room temperature. The solvent was evaporated under reduced pressure to give 110 mg (yield: 96%) of a colorless oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.54-1.66 (2H, m), 1.84 (1H, s), 1.86-1.97 (2H, m), 3.39-3.49 (2H, m), 3.49-3.57 (1H, m), 3.59 (2H, t, J = 4.9 Hz), 3.74 (2H, t, J = 4.9 Hz), 3.91-3.99 (2H, m).

(14b) 4-{2-[4-(4,4,5,5-テトラメチル-[1,3,2]ジオキサボロラン-2-イル)フェノキシ]エトキシ}テトラヒドロピラン (14b) 4- {2- [4- (4,4,5,5-tetramethyl- [1,3,2] dioxaborolan-2-yl) phenoxy] ethoxy} tetrahydropyran

Figure 2020071550
Figure 2020071550

実施例14 (14a) で製造した化合物 1.37 g (9.37 mmol) を 塩化メチレン 25 mL に溶解し、氷冷下トリエチルアミン 1.96 mL (14.1 mmol)、塩化メタンスルホニル 0.94 mL (12 mmol) を加え、室温にて 2時間撹拌した。反応液に水を加え、酢酸エチルにて2回抽出した。有機層を合わせ、飽和食塩水にて洗浄し、乾燥 (Na2SO4) 後、減圧下溶媒を留去し、2-(テトラヒドロピラン-2-イルオキシ)エチル メタンスルホネートの淡黄色油状物 2.10 gを得た。
2-(テトラヒドロピラン-2-イルオキシ)エチル メタンスルホネート 2.10 g (9.37 mmol) をN,N-ジメチルホルムアミド 20 mL に溶解し、4-(4,4,5,5-テトラメチル-[1,3,2]ジオキサボロラン-2-イル)フェノール 2.27 g (10.3 mmol) および炭酸カリウム 2.59 g (18.7 mmol) を加え、80℃ にて 2時間撹拌した。反応液に水を加え、酢酸エチルにて2回抽出し、有機層を合わせ、水、飽和食塩水にて順次洗浄し、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→8:1、V/V)にて精製後した。目的分画の溶媒を減圧下留去し、標題化合物の黄色油状物 1.16 g (収率:35%) を得た。
1H-NMR (CDCl3, 400 MHz)δ:1.33 (12H, s), 1.57-1.70 (2H, m), 1.87-1.98 (2H, m), 3.40-3.49 (2H, m), 3.55-3.65 (1H, m), 3.83-3.87 (2H, m), 3.91-4.00 (2H, m), 4.13-4.19 (2H, m), 6.87-6.94 (2H, m), 7.70-7.78 (2H, m).
1.37 g (9.37 mmol) of the compound prepared in Example 14 (14a) was dissolved in 25 mL of methylene chloride, 1.96 mL (14.1 mmol) of triethylamine and 0.94 mL (12 mmol) of methanesulfonyl chloride were added under ice-cooling, and the mixture was brought to room temperature. Stir for 2 hours. Water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers are combined, washed with saturated brine, dried (Na 2 SO 4 ), the solvent is evaporated under reduced pressure, and a pale yellow oil of 2- (tetrahydropyran-2-yloxy) ethyl methanesulfonate is 2.10 g. Got
2.10 g (9.37 mmol) of 2- (tetrahydropyran-2-yloxy) ethyl methanesulfonate was dissolved in 20 mL of N, N-dimethylformamide and 4- (4,4,5,5-tetramethyl- [1,3] , 2] Dioxaborolan-2-yl) phenol 2.27 g (10.3 mmol) and potassium carbonate 2.59 g (18.7 mmol) were added, and the mixture was stirred at 80 ° C. for 2 hours. Water was added to the reaction mixture, the mixture was extracted twice with ethyl acetate, the organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 8: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 1.16 g (yield: 35%) of a yellow oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.33 (12H, s), 1.57-1.70 (2H, m), 1.87-1.98 (2H, m), 3.40-3.49 (2H, m), 3.55-3.65 (1H, m), 3.83-3.87 (2H, m), 3.91-4.00 (2H, m), 4.13-4.19 (2H, m), 6.87-6.94 (2H, m), 7.70-7.78 (2H, m) ..

(14c) 5-ブロモベンゾ[b]チオフェン (14c) 5-Bromobenzo [b] thiophene

Figure 2020071550
Figure 2020071550

メタノール 14 mLにナトリウムメトキシド メタノール溶液 (P=28%) 7.1 mL (29 mmol) を滴下後、氷冷下、4-ブロモチオフェノール 5.00 g (26.4 mmol) を加え、5分間撹拌した。ブロモアセトアルデヒドジエチルアセタール 4.5 mL (29 mmol) を加え、3時間加熱還流した。減圧下溶媒を留去後、水50 mLを加え、酢酸エチル 50 mLにて2回抽出した。有機層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、20:1→10:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、1-ブロモ-4-(2,2-ジエトキシエチルスルファニル)ベンゼンの黄色油状物 7.70 g (収率:96%) を得た。
ポリリン酸 3.3 gにクロロベンゼン 80 mLおよび1-ブロモ-4-(2,2-ジエトキシエチルスルファニル)ベンゼン 3.31 g (10.8 mmol) を加え、1.5時間加熱還流した。放冷後、デカントにて上澄みを分離し、残渣にn-ヘキサンを加え、さらに上澄みを分離した。分離した溶液に飽和重曹水を加え、二層を分離後、有機層を飽和重曹水および飽和食塩水にて順次洗浄、乾燥 (Na2SO4) し、減圧下溶媒を留去した。残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、20:1→10:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 191 mg (収率:8%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:7.27 (1H, d, J=5.4 Hz), 7.43 (1H, dd, J=8.5, 1.9 Hz), 7.47 (1H, d, J=5.4 Hz), 7.73 (1H, d, J=8.5 Hz), 7.96 (1H, d, J=1.9 Hz).
After adding 7.1 mL (29 mmol) of sodium methoxide methanol solution (P = 28%) to 14 mL of methanol, 5.00 g (26.4 mmol) of 4-bromothiophenol was added under ice-cooling, and the mixture was stirred for 5 minutes. Bromoacetaldehyde diethyl acetal 4.5 mL (29 mmol) was added, and the mixture was heated under reflux for 3 hours. After distilling off the solvent under reduced pressure, 50 mL of water was added, and the mixture was extracted twice with 50 mL of ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography (n-hexane: ethyl acetate, 20: 1 → 10: 1, V / V), and the solvent of the target fraction was distilled off under reduced pressure to obtain 1-bromo-4- (2). , 2-Diethoxyethylsulfanyl) A yellow oil of benzene was obtained in an amount of 7.70 g (yield: 96%).
To 3.3 g of polyphosphate, 80 mL of chlorobenzene and 3.31 g (10.8 mmol) of 1-bromo-4- (2,2-diethoxyethylsulfanyl) benzene were added, and the mixture was heated under reflux for 1.5 hours. After allowing to cool, the supernatant was separated by decanting, n-hexane was added to the residue, and the supernatant was further separated. Saturated aqueous sodium hydrogen carbonate was added to the separated solution, the two layers were separated, and then the organic layer was washed successively with saturated aqueous sodium hydrogen carbonate and saturated brine (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography (n-hexane: ethyl acetate, 20: 1 → 10: 1, V / V), the solvent of the target fraction was distilled off under reduced pressure, and the white powder of the title compound was 191 mg (1 mg). Yield: 8%) was obtained.
1 H-NMR (CDCl 3 , 400 MHz) δ: 7.27 (1H, d, J = 5.4 Hz), 7.43 (1H, dd, J = 8.5, 1.9 Hz), 7.47 (1H, d, J = 5.4 Hz) , 7.73 (1H, d, J = 8.5 Hz), 7.96 (1H, d, J = 1.9 Hz).

(14d) 3-ブロモ-5-シアノベンゾ[b]チオフェン (14d) 3-Bromo-5-cyanobenzo [b] thiophene

Figure 2020071550
Figure 2020071550

実施例14 (14c) で製造した化合物 191 mg (0.896 mmol) をN,N-ジメチルホルムアミド 4 mLに溶解させ、シアン化銅(I) 241 mg (2.69 mmol) を加え、150℃ にて12時間撹拌した。放冷後、水 20 mLおよび酢酸エチル 20 mLを加え、不溶物をセライトにてろ別した。ろ液の二層を分離後、水層を酢酸エチル 20 mLにて抽出した。有機層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、5-シアノベンゾ[b]チオフェンの白色粉末 100 mg (収率:70%) を得た。また、実施例14 (14c) で製造した化合物 100 mg (0.469 mmol) を用い、上記と同様に反応させ、5-シアノベンゾ[b]チオフェンの白色粉末 23 mg (収率:31%) を得た。
5-シアノベンゾ[b]チオフェン 121 mg (0.760 mmol) を酢酸 4 mLに溶解させ、氷冷下臭素 0.12 mL (2.3 mmol) を加え、室温にて1.5時間撹拌した。減圧下溶媒を留去し、飽和重曹水 20 mLを加え、酢酸エチル 20 mLにて抽出し、有機層を飽和重曹水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) した。減圧下溶媒を留去し、標題化合物の白色粉末 155 mg (収率:86%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:7.60 (1H, s), 7.63 (1H, dd, J=8.3, 1.5 Hz), 7.96 (1H, d, J=8.3 Hz), 8.16 (1H, d, J=1.5 Hz).
Compound 191 mg (0.896 mmol) prepared in Example 14 (14c) was dissolved in 4 mL of N, N-dimethylformamide, copper (I) cyanide (I) 241 mg (2.69 mmol) was added, and the mixture was added at 150 ° C. for 12 hours. Stirred. After allowing to cool, 20 mL of water and 20 mL of ethyl acetate were added, and the insoluble material was filtered off with Celite. After separating the two layers of the filtrate, the aqueous layer was extracted with 20 mL of ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1, V / V), the solvent for the target fraction was distilled off under reduced pressure, and 100 mg of a white powder of 5-cyanobenzo [b] thiophene was distilled off. (Yield: 70%) was obtained. Further, 100 mg (0.469 mmol) of the compound prepared in Example 14 (14c) was used and reacted in the same manner as above to obtain 23 mg (yield: 31%) of a white powder of 5-cyanobenzo [b] thiophene. ..
121 mg (0.760 mmol) of 5-cyanobenzo [b] thiophene was dissolved in 4 mL of acetic acid, 0.12 mL (2.3 mmol) of bromine was added under ice-cooling, and the mixture was stirred at room temperature for 1.5 hours. The solvent was distilled off under reduced pressure, 20 mL of saturated aqueous sodium hydrogen carbonate was added, the mixture was extracted with 20 mL of ethyl acetate, and the organic layer was washed successively with saturated aqueous sodium hydrogen carbonate and saturated brine, and dried (Na 2 SO 4 ). The solvent was distilled off under reduced pressure to obtain 155 mg (yield: 86%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 7.60 (1H, s), 7.63 (1H, dd, J = 8.3, 1.5 Hz), 7.96 (1H, d, J = 8.3 Hz), 8.16 (1H, 1H, d, J = 1.5 Hz).

(14e) 3-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ベンゾ[b]チオフェン-5-カルボニトリル (14e) 3- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] phenyl} benzo [b] thiophene-5-carbonitrile

Figure 2020071550
Figure 2020071550

実施例14 (14d) で製造した化合物 152 mg (0.638 mmol)、実施例14 (14b) で製造した化合物 222 mg (0.637 mmol)、[1,1'-ビス(ジフェニルホスフィノ)フェロセン]パラジウム(II)ジクロリド ジクロロメタン付加物 52 mg (0.064 mmol)、リン酸三カリウム407 mg (1.92 mmol) およびテトラブチルアンモニウムブロミド 21 mg (0.065 mmol) をアセトニトリル 4 mLに懸濁し、窒素雰囲気下、12時間加熱還流した。放冷後、減圧下溶媒を留去し、水 20 mLを加え、酢酸エチル 20 mLにて2回抽出した。有機層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、2:1→1:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の淡黄色粘性油状物 0.19 g (収率:78%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.62-1.72 (2H, m), 1.93-2.02 (2H, m), 3.43-3.51 (2H, m), 3.60-3.68 (1H, m), 3.89 (2H, t, J=4.9 Hz), 3.95-4.02 (2H, m), 4.21 (2H, t, J=4.9 Hz), 7.04-7.10 (2H, m), 7.43-7.49 (2H, m), 7.48 (1H, s), 7.58 (1H, dd, J=8.4, 1.3 Hz), 7.99 (1H, dd, J=8.4, 0.6 Hz), 8.18 (1H, dd, J=1.3, 0.6 Hz).
Compound 152 mg (0.638 mmol) prepared in Example 14 (14d), compound 222 mg (0.637 mmol) prepared in Example 14 (14b), [1,1'-bis (diphenylphosphino) ferrocene] palladium ( II) Dichloride dichloromethane adduct 52 mg (0.064 mmol), tripotassium phosphate 407 mg (1.92 mmol) and tetrabutylammonium bromide 21 mg (0.065 mmol) were suspended in 4 mL of acetonitrile and heated to reflux for 12 hours under a nitrogen atmosphere. bottom. After allowing to cool, the solvent was evaporated under reduced pressure, 20 mL of water was added, and the mixture was extracted twice with 20 mL of ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography (n-hexane: ethyl acetate, 2: 1 → 1: 1, V / V), and the solvent of the target fraction was distilled off under reduced pressure to obtain a pale yellow viscous oil of the title compound. 0.19 g (yield: 78%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.62-1.72 (2H, m), 1.93-2.02 (2H, m), 3.43-3.51 (2H, m), 3.60-3.68 (1H, m), 3.89 (2H, t, J = 4.9 Hz), 3.95-4.02 (2H, m), 4.21 (2H, t, J = 4.9 Hz), 7.04-7.10 (2H, m), 7.43-7.49 (2H, m), 7.48 (1H, s), 7.58 (1H, dd, J = 8.4, 1.3 Hz), 7.99 (1H, dd, J = 8.4, 0.6 Hz), 8.18 (1H, dd, J = 1.3, 0.6 Hz).

(14) 3-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ベンゾ[b]チオフェン-5-カルボキサミド (14) 3- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] phenyl} benzo [b] thiophene-5-carboxamide

Figure 2020071550
Figure 2020071550

実施例14 (14e) で製造した化合物 0.19 g (0.50 mmol) をジメチルスルホキシド 2.5 mLに溶解し、室温にて5.0 M 水酸化ナトリウム水 0.15 mL (0.75 mmol) および30% 過酸化水素水 0.068 mL (0.75 mmol) を加え、同温にて40分間撹拌した。水 20 mLおよびジエチルエーテル 10 mLを加え、30分間撹拌後、析出物をろ取し、標題化合物の白色粉末 135 mg (収率:68%) を得た。
IR ν (ATR) cm-1:1670
1H-NMR (DMSO-d6, 400 MHz) δ:1.38-1.48 (2H, m), 1.85-1.92 (2H, m), 3.29-3.41 (2H, m), 3.60-3.68 (1H, m), 3.78-3.85 (4H, m), 4.14-4.20 (2H, m), 7.10-7.15 (2H, m), 7.36-7.44 (1H, br), 7.53-7.58 (2H, m), 7.80 (1H, s), 7.89 (1H, dd, J=8.4, 1.3 Hz), 8.10-8.15 (2H, m), 8.35 (1H, d, J=1.3 Hz).
MS m/z: 420 [M+Na]+, 396 [M-H]-.
0.19 g (0.50 mmol) of the compound prepared in Example 14 (14e) was dissolved in 2.5 mL of dimethyl sulfoxide, and at room temperature 5.0 M sodium hydroxide water 0.15 mL (0.75 mmol) and 30% hydrogen peroxide solution 0.068 mL ( 0.75 mmol) was added, and the mixture was stirred at the same temperature for 40 minutes. 20 mL of water and 10 mL of diethyl ether were added, and the mixture was stirred for 30 minutes, and the precipitate was collected by filtration to give 135 mg (yield: 68%) of a white powder of the title compound.
IR ν (ATR) cm -1 : 1670
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 1.38-1.48 (2H, m), 1.85-1.92 (2H, m), 3.29-3.41 (2H, m), 3.60-3.68 (1H, m), 3.78-3.85 (4H, m), 4.14-4.20 (2H, m), 7.10-7.15 (2H, m), 7.36-7.44 (1H, br), 7.53-7.58 (2H, m), 7.80 (1H, s) ), 7.89 (1H, dd, J = 8.4, 1.3 Hz), 8.10-8.15 (2H, m), 8.35 (1H, d, J = 1.3 Hz).
MS m / z: 420 [M + Na] + , 396 [MH] - .

実施例15 Example 15

(15a) 3-エチニル-4-ヒドロキシベンゾニトリル (15a) 3-ethynyl-4-hydroxybenzonitrile

Figure 2020071550
Figure 2020071550

3-ブロモ-4-ヒドロキシベンゾニトリル 5.20 g (25.2 mmol) をピリジン 40 mL に溶解し、氷冷下、無水酢酸 7.15 mL (75.7 mmol) を加え、室温にて19時間撹拌した。反応液を減圧下濃縮し、ジイソプロピルエーテルを加え、析出物をろ取し、2-ブロモ-4-シアノフェニル アセテートの白色粉末 5.16 g (収率:85%) を得た。
2-ブロモ-4-シアノフェニル アセテート 300 mg (1.25 mmol) を1,4-ジオキサン 5 mLに溶解し、トリメチルシリルアセチレン 246 mg (2.50mmol)、ビス(トリフェニルホスフィン)パラジウム(II)ジクロリド 44 mg (0.063 mmol)、ヨウ化銅 12 mg (0.063 mmol)、トリエチルアミン 0.35 mL (2.5 mmol) を加え、2分間窒素をバブリング後、窒素雰囲気下、80℃ にて1時間撹拌した。放冷後、水および酢酸エチルを加え、不溶物をろ別した。ろ液の二層を分離し、水層を酢酸エチルにて抽出した。有機層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。2-ブロモ-4-シアノフェニル アセテート 4.86 g (20.2 mmol) を用いて同様の反応および処理を行い、先に得られた残渣と合わせ、カラムクロマトグラフィー(n-ヘキサン:酢酸エチル、9:1→7:3、V/V)にて精製した。目的分画の溶媒を減圧下留去し、4-シアノ-2-トリメチルシラニルエチニルフェニル アセテートの淡褐色粉末 4.75 g (収率:86%) を得た。
4-シアノ-2-トリメチルシラニルエチニルフェニル アセテート 370 mg (1.44 mmol) をメタノール 3 mL に溶解し、フッ化カリウム184 mg (3.16 mmol) を加え、室温にて2時間撹拌後、フッ化カリウム 84 mg (1.4 mmol) を追加し、同温にてさらに1時間撹拌した。反応液に5%クエン酸水を加え、酢酸エチルにて2回抽出した。有機層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。4-シアノ-2-トリメチルシラニルエチニルフェニル アセテート 4.34 g (16.9 mmol) を用いて同様の反応および処理を行い、先に得られた残渣と合わせ、カラムクロマトグラフィー(クロロホルム:酢酸エチル、9:1→7:3、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡褐色粉末 2.49 g (収率:95%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:3.57 (1H, s), 6.31 (1H, s), 7.04 (1H, d, J=8.5 Hz), 7.55 (1H, dd, J=8.5, 2.0 Hz), 7.69 (1H, d, J=2.0 Hz).
5.20 g (25.2 mmol) of 3-bromo-4-hydroxybenzonitrile was dissolved in 40 mL of pyridine, 7.15 mL (75.7 mmol) of acetic anhydride was added under ice-cooling, and the mixture was stirred at room temperature for 19 hours. The reaction mixture was concentrated under reduced pressure, diisopropyl ether was added, and the precipitate was collected by filtration to obtain 5.16 g (yield: 85%) of a white powder of 2-bromo-4-cyanophenyl acetate.
Dissolve 300 mg (1.25 mmol) of 2-bromo-4-cyanophenyl acetate in 5 mL of 1,4-dioxane, 246 mg (2.50 mmol) of trimethylsilylacetylene, 44 mg of bis (triphenylphosphine) palladium (II) dichloride (II). 0.063 mmol), copper iodide 12 mg (0.063 mmol), and triethylamine 0.35 mL (2.5 mmol) were added, nitrogen was bubbled for 2 minutes, and the mixture was stirred at 80 ° C. for 1 hour under a nitrogen atmosphere. After allowing to cool, water and ethyl acetate were added, and the insoluble material was filtered off. The two layers of the filtrate were separated, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. A similar reaction and treatment was carried out with 4.86 g (20.2 mmol) of 2-bromo-4-cyanophenyl acetate, which was combined with the previously obtained residue for column chromatography (n-hexane: ethyl acetate, 9: 1 → Purified by 7: 3, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 4.75 g (yield: 86%) of a light brown powder of 4-cyano-2-trimethylsilanylethynylphenyl phenyl acetate.
4-Cyano-2-trimethylsilanylethynylphenyl phenyl acetate 370 mg (1.44 mmol) is dissolved in 3 mL of methanol, potassium fluoride 184 mg (3.16 mmol) is added, and the mixture is stirred at room temperature for 2 hours, and then potassium fluoride 84. mg (1.4 mmol) was added, and the mixture was stirred at the same temperature for another 1 hour. 5% citric acid water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. Similar reactions and treatments were performed with 4.34 g (16.9 mmol) of 4-cyano-2-trimethylsilanylethynylphenyl acetate, combined with the previously obtained residue and column chromatography (chloroform: ethyl acetate, 9: 1). → Purified by 7: 3, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 2.49 g (yield: 95%) of a light brown powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 3.57 (1H, s), 6.31 (1H, s), 7.04 (1H, d, J = 8.5 Hz), 7.55 (1H, dd, J = 8.5, 2.0) Hz), 7.69 (1H, d, J = 2.0 Hz).

(15b) ベンゾフラン-5-カルボニトリル (15b) Benzofuran-5-Carbonitrile

Figure 2020071550
Figure 2020071550

実施例15 (15a) で製造した化合物 215 mg (1.50 mmol) をピリジン 2 mL に溶解し、シクロペンタジエニルビス-(トリフェニルホスフィン)ルテニウム(II) クロリド 55 mg (0.075 mmol) を加え、窒素雰囲気下、90℃ にて1.5時間撹拌後、シクロペンタジエニルビス-(トリフェニルホスフィン)ルテニウム(II) クロリド 28 mg (0.038 mmol) を追加し、同温にて更に1時間撹拌した。放冷後、水を加え、酢酸エチルにて2回抽出した。有機層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、9:1→7:3、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 60 mg (収率:28%) を得た。
実施例15 (15a) で製造した化合物 2.25 g (15.7 mmol) を用いて同様の反応および処理を行い、標題化合物の白色粉末 1.04 g (収率:46%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:6.85 (1H, d, J=2.2 Hz), 7.55-7.61 (2H, m), 7.74 (1H, d, J=2.2 Hz), 7.95 (1H, d, J=1.0 Hz).
Compound 215 mg (1.50 mmol) prepared in Example 15 (15a) was dissolved in 2 mL of pyridine, cyclopentadienylbis- (triphenylphosphine) ruthenium (II) chloride 55 mg (0.075 mmol) was added, and nitrogen was added. After stirring at 90 ° C. for 1.5 hours under an atmosphere, 28 mg (0.038 mmol) of cyclopentadienylbis- (triphenylphosphine) ruthenium (II) chloride was added, and the mixture was further stirred at the same temperature for 1 hour. After allowing to cool, water was added and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 9: 1 → 7: 3, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 60 mg (yield: 28%) of a white powder of the title compound.
The same reaction and treatment was carried out using 2.25 g (15.7 mmol) of the compound prepared in Example 15 (15a) to obtain 1.04 g (yield: 46%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 6.85 (1H, d, J = 2.2 Hz), 7.55-7.61 (2H, m), 7.74 (1H, d, J = 2.2 Hz), 7.95 (1H, 1H, d, J = 1.0 Hz).

(15c) 3-ブロモベンゾフラン-5-カルボニトリル (15c) 3-Bromobenzofuran-5-Carbonitrile

Figure 2020071550
Figure 2020071550

実施例15 (15b) で製造した化合物 500 mg (3.49 mmol) を塩化メチレン 10 mL に溶解し、室温にて臭素 0.36 mL (7.0 mmol) を加え、同温にて20分間撹拌した。反応液を1 M チオ硫酸ナトリウム水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をテトラヒドロフラン 10 mLに溶解し、室温にて2.0 M 水酸化カリウム メタノール溶液 1.75 mL (3.5 mmol) を加え、同温にて30分間撹拌した。反応液に水およびクロロホルムを加え、二層を分離した。水層をクロロホルムにて抽出し、有機層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の白色粉末 596 mg (収率:77%) を得た。1H-NMR (CDCl3, 400 MHz) δ:7.55-7.65 (2H, m), 7.78 (1H, s), 7.91 (1H, d, J=1.4 Hz).500 mg (3.49 mmol) of the compound prepared in Example 15 (15b) was dissolved in 10 mL of methylene chloride, 0.36 mL (7.0 mmol) of bromine was added at room temperature, and the mixture was stirred at the same temperature for 20 minutes. The reaction mixture was washed successively with 1 M aqueous sodium thiosulfate solution and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was dissolved in 10 mL of tetrahydrofuran, 1.75 mL (3.5 mmol) of 2.0 M potassium hydroxide methanol solution was added at room temperature, and the mixture was stirred at the same temperature for 30 minutes. Water and chloroform were added to the reaction solution, and the two layers were separated. The aqueous layer was extracted with chloroform, the organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. Diisopropyl ether was added to the obtained residue and collected by filtration to obtain 596 mg (yield: 77%) of a white powder of the title compound. 1 1 H-NMR (CDCl 3 , 400 MHz) δ: 7.55-7.65 (2H, m), 7.78 (1H, s), 7.91 (1H, d, J = 1.4 Hz).

(15d) 3-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ベンゾフラン-5-カルボニトリル (15d) 3- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] Phenyl} Benzofuran-5-Carbonitrile

Figure 2020071550
Figure 2020071550

実施例15 (15c) で製造した化合物 400 mg (1.80 mmol) をアセトニトリル 6 mL に溶解し、実施例14 (14b) で製造した化合物 570 mg (1.80 mmol)、[1,1'-ビス(ジフェニルホスフィノ)フェロセン]パラジウム(II)ジクロリド ジクロロメタン付加物 134 mg (0.18 mmol)、リン酸三カリウム 1.04 g (4.92 mmol)、テトラブチルアンモニウムブロミド 53 mg (0.18 mmol) を加え、2分間窒素をバブリングした。窒素雰囲気下、1時間加熱還流後、[1,1'-ビス(ジフェニルホスフィノ)フェロセン]パラジウム(II)ジクロリド ジクロロメタン付加物 134 mg (0.18 mmol) を追加し、さら2.5時間加熱還流した。放冷後、水および酢酸エチルを加え、不溶物をろ別した。ろ液の二層を分離後、水層を酢酸エチルにて抽出し、有機層を合わせ、飽和食塩水にて洗浄後、乾燥 (Na2SO4) 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(n-ヘキサン:酢酸エチル、9:1→1:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の無色油状物 460 mg (収率:77%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.62-1.72 (2H, m), 1.92-2.02 (2H, m), 3.43-3.52 (2H, m), 3.59-3.67 (1H, m), 3.85-3.91 (2H, m), 3.92-4.02 (2H, m), 4.15-4.24 (2H, m), 7.04-7.07 (2H, m), 7.49-7517 (2H, m), 7.60-7.64 (2H, m), 7.82 (1H, s), 8.12 (1H, s).
Compound 400 mg (1.80 mmol) prepared in Example 15 (15c) was dissolved in 6 mL of acetonitrile, and compound 570 mg (1.80 mmol) prepared in Example 14 (14b), [1,1'-bis (diphenyl). Phosphino) ferrocene] Palladium (II) dichloride dichloromethane adduct 134 mg (0.18 mmol), tripotassium phosphate 1.04 g (4.92 mmol), tetrabutylammonium bromide 53 mg (0.18 mmol) were added and nitrogen was bubbled for 2 minutes. .. After heating under reflux for 1 hour under a nitrogen atmosphere, 134 mg (0.18 mmol) of [1,1'-bis (diphenylphosphino) ferrocene] palladium (II) dichloride dichloromethane adduct was added, and the mixture was heated under reflux for another 2.5 hours. After allowing to cool, water and ethyl acetate were added, and the insoluble material was filtered off. After separating the two layers of the filtrate, the aqueous layer was extracted with ethyl acetate, the organic layers were combined, washed with saturated brine, and dried (Na 2 SO 4 ). The solvent was evaporated under reduced pressure, and the obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 9: 1 → 1: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure to give 460 mg (yield: 77%) of a colorless oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.62-1.72 (2H, m), 1.92-2.02 (2H, m), 3.43-3.52 (2H, m), 3.59-3.67 (1H, m), 3.85 -3.91 (2H, m), 3.92-4.02 (2H, m), 4.15-4.24 (2H, m), 7.04-7.07 (2H, m), 7.49-7517 (2H, m), 7.60-7.64 (2H, m) m), 7.82 (1H, s), 8.12 (1H, s).

(15e) 2-クロロ-3-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ベンゾフラン-5-カルボニトリル (15e) 2-Chloro-3- {4- [2- (tetrahydropyran-4-yloxy) ethoxy] phenyl} benzofuran-5-carbonitrile

Figure 2020071550
Figure 2020071550

実施例15 (15d) で製造した化合物 105 mg (0.289 mmol) をベンゼン 1 mLに溶解し、氷冷下、塩化スルフリル0.025 mL (0.30 mmol) を加え、同温にて30分間撹拌後、室温にて1.5時間撹拌した。反応液に飽和重曹水を加え、酢酸エチルにて2回抽出した。有機層を合わせ、飽和食塩水にて洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、9:1→2:3、V/V)にて精製し、目的分画の溶媒を減圧下留去し、標題化合物の無色油状物 80 mg (収率:70%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.62-1.72 (2H, m), 1.92-2.02 (2H, m), 3.43-3.52 (2H, m), 3.59-3.67 (1H, m), 3.85-3.91 (2H, m), 3.92-4.02 (2H, m), 4.18-4.24 (2H, m), 7.07-7.09 (2H, m), 7.51-7.53 (2H, m), 7.55-7.61 (2H, m), 7.95 (1H, s).
105 mg (0.289 mmol) of the compound prepared in Example 15 (15d) was dissolved in 1 mL of benzene, 0.025 mL (0.30 mmol) of sulfuryl chloride was added under ice-cooling, and the mixture was stirred at the same temperature for 30 minutes and then brought to room temperature. Stirred for 1.5 hours. Saturated aqueous sodium hydrogen carbonate was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 9: 1 → 2: 3, V / V), the solvent of the target fraction was evaporated under reduced pressure, and the title compound was a colorless oil. A product of 80 mg (yield: 70%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.62-1.72 (2H, m), 1.92-2.02 (2H, m), 3.43-3.52 (2H, m), 3.59-3.67 (1H, m), 3.85 -3.91 (2H, m), 3.92-4.02 (2H, m), 4.18-4.24 (2H, m), 7.07-7.09 (2H, m), 7.51-7.53 (2H, m), 7.55-7.61 (2H, m) m), 7.95 (1H, s).

(15) 2-クロロ-3-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ベンゾフラン-5-カルボキサミド (15) 2-Chloro-3- {4- [2- (tetrahydropyran-4-yloxy) ethoxy] phenyl} benzofuran-5-carboxamide

Figure 2020071550
Figure 2020071550

実施例15 (15e) で製造した化合物 80 mg (0.20 mmol) をジメチルスルホキシド 1 mL に溶解し、炭酸カリウム 83 mg (0.60 mmol)、30% 過酸化水素水 32 mg (1.0 mmol)を加え、室温にて1時間撹拌した。反応液に水を加え、酢酸エチルにて2回抽出した。有機層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の白色粉末 55 mg (収率:66%) を得た。
IR ν (ATR) cm-1 :3403, 3176, 1670
1H-NMR (CDCl3, 400 MHz) δ :1.60-1.72 (2H, m), 1.90-2.00 (2H, m), 3.43-3.52 (2H, m), 3.58-3.67 (1H, m), 3.86-3.91 (2H, m), 3.92-4.00 (2H, m), 4.17-4.24 (2H, m), 5.45-6.25 (2H, br), 7.06-7.08 (2H, m), 7.50-7.56 (3H, m), 7.78 (1H, dd, J=8.6, 1.7 Hz), 8.09 (1H, d, J=1.7 Hz).
MS (ESI) m/z:416 [M+H+].
80 mg (0.20 mmol) of the compound prepared in Example 15 (15e) was dissolved in 1 mL of dimethyl sulfoxide, 83 mg (0.60 mmol) of potassium carbonate and 32 mg (1.0 mmol) of 30% hydrogen peroxide solution were added, and room temperature was added. Was stirred for 1 hour. Water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. Diisopropyl ether was added to the residue and collected by filtration to give 55 mg (yield: 66%) of a white powder of the title compound.
IR ν (ATR) cm -1 : 3403, 3176, 1670
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.60-1.72 (2H, m), 1.90-2.00 (2H, m), 3.43-3.52 (2H, m), 3.58-3.67 (1H, m), 3.86 -3.91 (2H, m), 3.92-4.00 (2H, m), 4.17-4.24 (2H, m), 5.45-6.25 (2H, br), 7.06-7.08 (2H, m), 7.50-7.56 (3H, m) m), 7.78 (1H, dd, J = 8.6, 1.7 Hz), 8.09 (1H, d, J = 1.7 Hz).
MS (ESI) m / z: 416 [M + H + ].

実施例16 Example 16

(16a) 1,4-ジオキサスピロ[4.5]デカ-7-エン-8-イル トリフルオロメタンスルホネート (16a) 1,4-Dioxaspiro [4.5] Deca-7-en-8-yl trifluoromethanesulfonate

Figure 2020071550
Figure 2020071550

1,4-シクロヘキサンジオンモノエチレンケタール 2.40 g (15.4 mmol) およびN-フェニルビス(トリフルオロメタンスルホンイミド) 7.15 g (20.0 mmol) をテトラヒドロフラン 150 mLに溶解し、-78℃ にて0.5 M カリウムヘキサメチルジシラジド トルエン溶液 40 mL (20 mmol) を滴下し、同温にて2.5時間撹拌した。反応液を水に注加後、ジエチルエーテルにて抽出し、有機層を飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1、V/V)にて精製し、目的分画の溶媒を減圧下留去した。得られた残渣を再度カラムクロマトグラフィー(Chromatorex NH、n-ヘキサン:酢酸エチル、9:1→2:3、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の微黄色油状物 4.68 g (収率:定量的) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.91 (2H, t, J=6.6 Hz, 2.38-2.44 (2H, m), 2.51-2.58 (2H, m), 3.95-4.03 (4H, m), 5.64-5.69 (1H, m).
Dissolve 2.40 g (15.4 mmol) of 1,4-cyclohexanedione monoethylene ketal and 7.15 g (20.0 mmol) of N-phenylbis (trifluoromethanesulfonimide) in 150 mL of tetrahydrofuran and 0.5 M potassium hexamethyl at -78 ° C. 40 mL (20 mmol) of disilazide toluene solution was added dropwise, and the mixture was stirred at the same temperature for 2.5 hours. The reaction mixture was poured into water, extracted with diethyl ether, the organic layer was washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1, V / V), and the solvent for the target fraction was evaporated under reduced pressure. The obtained residue was purified again by column chromatography (Chromatorex NH, n-hexane: ethyl acetate, 9: 1 → 2: 3, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 4.68 g (yield: quantitative) of a slightly yellow oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.91 (2H, t, J = 6.6 Hz, 2.38-2.44 (2H, m), 2.51-2.58 (2H, m), 3.95-4.03 (4H, m) , 5.64-5.69 (1H, m).

(16b) 3-(1,4-ジオキサスピロ[4.5]デカ-7-エン-8-イル)ベンゾ[b]チオフェン-5-カルボニトリル (16b) 3- (1,4-dioxaspiro [4.5] deca-7-ene-8-yl) benzo [b] thiophene-5-carbonitrile

Figure 2020071550
Figure 2020071550

実施例14 (14d) で製造した化合物 500 mg (2.10 mmol) を1,4-オキサン 10 mLに溶解し、酢酸カリウム 720 mg (7.34 mmol)、ビス(ピナコラト)ジボロン 590 mg (2.32 mmol) およびビス(トリフェニルホスフィン)パラジウム(II)ジクロリド 150 mg (0.214 mmol) を加え、窒素雰囲気下100℃ にて4時間撹拌した。放冷後、反応液を酢酸エチルにて希釈し、水および飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、3-(4,4,5,5-テトラメチル[1,3,2]ジオキサボロラン-2-イル)ベンゾ[b]チオフェン-5-カルボニトリルを含む淡黄色粉末 486 mgを得た。
実施例16 (16a) で製造した化合物 330 mg (1.14 mmol) および3-(4,4,5,5-テトラメチル[1,3,2]ジオキサボロラン-2-イル)ベンゾ[b]チオフェン-5-カルボニトリルを含む粉末 486 mg を1,2-ジメトキシエタン 5 mLに溶解し、炭酸ナトリウム 350 mg (3.02 mmol) の水5 mLの溶液、塩化リチウム 50 mg (1.2 mmol) の水0.5 mLの溶液およびテトラキス(トリフェニルホスフィン)パラジウム(0) 66 mg (0.057 mmol) を順次加え、窒素雰囲気下70℃ にて14時間撹拌した。放冷後、反応液を酢酸エチルにて希釈し、水および飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→5:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の黄色油状物 308 mg (収率:90%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.98 (2H, t, J=6.6 Hz), 2.52-2.57 (2H, m), 2.64-2.71 (2H, m), 4.03-4.08 (4H, m), 5.91-5.96 (1H, m), 7.36 (1H, s), 7.54 (1H, dd, J=8.3, 1.5 Hz), 7.92 (1H, d, J=8.3 Hz), 8.21-8.25 (1H, m).
Compound 500 mg (2.10 mmol) prepared in Example 14 (14d) was dissolved in 10 mL of 1,4-oxane, potassium acetate 720 mg (7.34 mmol), bis (pinacolato) diboron 590 mg (2.32 mmol) and bis. (Triphenylphosphine) Palladium (II) dichloride 150 mg (0.214 mmol) was added, and the mixture was stirred at 100 ° C. for 4 hours under a nitrogen atmosphere. After allowing to cool, the reaction solution was diluted with ethyl acetate, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1, V / V), and the solvent for the target fraction was distilled off under reduced pressure to 3- (4,4,5, A pale yellow powder containing 5-tetramethyl [1,3,2] dioxaborolan-2-yl) benzo [b] thiophene-5-carbonitrile was obtained in an amount of 486 mg.
Compound 330 mg (1.14 mmol) and 3- (4,4,5,5-tetramethyl [1,3,2] dioxaborolan-2-yl) benzo [b] thiophene-5 prepared in Example 16 (16a). -Dissolve 486 mg of powder containing carbonitrile in 5 mL of 1,2-dimethoxyethane, a solution of 350 mg (3.02 mmol) of sodium carbonate in 5 mL of water, and a solution of lithium chloride 50 mg (1.2 mmol) in 0.5 mL of water. And tetrakis (triphenylphosphine) palladium (0) 66 mg (0.057 mmol) were sequentially added, and the mixture was stirred at 70 ° C. for 14 hours under a nitrogen atmosphere. After allowing to cool, the reaction solution was diluted with ethyl acetate, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 5: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 308 mg (yield: 90%) of a yellow oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.98 (2H, t, J = 6.6 Hz), 2.52-2.57 (2H, m), 2.64-2.71 (2H, m), 4.03-4.08 (4H, m) ), 5.91-5.96 (1H, m), 7.36 (1H, s), 7.54 (1H, dd, J = 8.3, 1.5 Hz), 7.92 (1H, d, J = 8.3 Hz), 8.21-8.25 (1H, s) m).

(16c) 3-(4-ヒドロキシシクロヘキサ-1-エンイル)ベンゾ[b]チオフェン-5-カルボニトリル (16c) 3- (4-Hydroxycyclohexa-1-enyl) benzo [b] thiophene-5-carbonitrile

Figure 2020071550
Figure 2020071550

実施例16 (16b) で製造した化合物 2.08 g (6.99 mmol) をテトラヒドロフラン 40 mLに溶解させ、1.0 M 塩酸21 mL (21 mmol) を加え、40℃ にて3時間撹拌した。反応液に酢酸エチルを加え、水および飽和食塩水にて順次洗浄、乾燥 (Na2SO4) した。減圧下溶媒を留去し、3-(4-オキソヘキサ-1-エンイル)ベンゾ[b]チオフェン-5-カルボニトリルの黄色粉末 1.72 g (収率:97%) を得た。
3-(4-オキソヘキサ-1-エンイル)ベンゾ[b]チオフェン-5-カルボニトリル 1.72 g (6.79 mmol) をエタノール 50 mLおよびテトラヒドロフラン 17 mLの混合溶媒に溶解させ、氷冷下、水素化ホウ素ナトリウム 514 mg (13.6 mmol) を加え、室温にて1時間撹拌後、反応液を水に注加し、酢酸エチルにて抽出した。有機層を水および飽和食塩水にて順次洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、3:1→2:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡黄色粉末 1.49 g (収率:86%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.50-1.60 (1H, br), 1.85-1.95 (1H, m), 2.03-2.11 (1H, m), 2.26-2.36 (1H, m), 2.47-2.72 (3H, m), 4.12-4.21 (1H, m), 5.92-5.98 (1H, m), 7.35 (1H, s), 7.54 (1H, dd, J=8.3, 1.5 Hz), 7.93 (1H, d, J=8.3 Hz), 8.20-8.24 (1H, m).
2.08 g (6.99 mmol) of the compound prepared in Example 16 (16b) was dissolved in 40 mL of tetrahydrofuran, 21 mL (21 mmol) of 1.0 M hydrochloric acid was added, and the mixture was stirred at 40 ° C. for 3 hours. Ethyl acetate was added to the reaction mixture, and the mixture was washed successively with water and saturated brine, and dried (Na 2 SO 4 ). The solvent was distilled off under reduced pressure to obtain 1.72 g (yield: 97%) of a yellow powder of 3- (4-oxohexa-1-eneyl) benzo [b] thiophene-5-carbonitrile.
Dissolve 1.72 g (6.79 mmol) of 3- (4-oxohexa-1-eneyl) benzo [b] thiophene-5-carbonitrile in a mixed solvent of 50 mL ethanol and 17 mL tetrahydrofuran, and sodium borohydride under ice-cooling. After adding 514 mg (13.6 mmol) and stirring at room temperature for 1 hr, the reaction solution was poured into water and extracted with ethyl acetate. The organic layer was washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 3: 1 → 2: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 1.49 g (yield: 86%) of a pale yellow powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.50-1.60 (1H, br), 1.85-1.95 (1H, m), 2.03-2.11 (1H, m), 2.26-2.36 (1H, m), 2.47 -2.72 (3H, m), 4.12-4.21 (1H, m), 5.92-5.98 (1H, m), 7.35 (1H, s), 7.54 (1H, dd, J = 8.3, 1.5 Hz), 7.93 (1H) , d, J = 8.3 Hz), 8.20-8.24 (1H, m).

(16d) 3-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]シクロヘキサ-1-エンイル}ベンゾ[b]チオフェン-5-カルボニトリル (16d) 3- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] cyclohex-1-enyl} benzo [b] thiophene-5-carbonitrile

Figure 2020071550
Figure 2020071550

実施例16 (16c) で製造した化合物 0.88 g (3.5 mmol) をN,N-ジメチルホルムアミド 10 mLに溶解させ、氷冷下、水素化ナトリウム (P=60%) 207 mg (5.2 mmol) を加え、室温にて15分間撹拌した。反応液に4-(2-ブロモエトキシ)テトラヒドロピラン 1.44 g (6.89 mmol) を加え、13.5時間撹拌後、反応液を水に注加し、酢酸エチルにて抽出した。有機層を飽和食塩水にて洗浄、乾燥 (Na2SO4) 後、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1、V/V)にて精製し、目的分画の溶媒を減圧下留去した。得られた残渣を再度カラムクロマトグラフィー(Chromatorex NH、n-ヘキサン:酢酸エチル、4:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の無色油状物 0.30 g (収率:25%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.57-1.67 (2H, m), 1.87-1.97 (3H, m), 1.96-2.04 (1H, m), 2.29-2.68 (4H, m), 3.39-3.80 (8H, m), 3.92-3.98 (2H, m), 5.91-5.97 (1H, m), 7.33 (1H, s), 7.54 (1H, dd, J=8.3, 1.5 Hz), 7.92 (1H, d, J=8.3 Hz), 8.21 (1H, d, J=1.5 Hz).
The compound 0.88 g (3.5 mmol) prepared in Example 16 (16c) was dissolved in 10 mL of N, N-dimethylformamide, and sodium hydride (P = 60%) 207 mg (5.2 mmol) was added under ice-cooling. , Stirred at room temperature for 15 minutes. 1.44 g (6.89 mmol) of 4- (2-bromoethoxy) tetrahydropyran was added to the reaction mixture, and the mixture was stirred for 13.5 hours, the reaction mixture was poured into water, and the mixture was extracted with ethyl acetate. The organic layer was washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1, V / V), and the solvent for the target fraction was evaporated under reduced pressure. The obtained residue was purified by column chromatography (Chromatorex NH, n-hexane: ethyl acetate, 4: 1, V / V) again, and the solvent of the target fraction was distilled off under reduced pressure to obtain a colorless oil of the title compound. A product of 0.30 g (yield: 25%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.57-1.67 (2H, m), 1.87-1.97 (3H, m), 1.96-2.04 (1H, m), 2.29-2.68 (4H, m), 3.39 -3.80 (8H, m), 3.92-3.98 (2H, m), 5.91-5.97 (1H, m), 7.33 (1H, s), 7.54 (1H, dd, J = 8.3, 1.5 Hz), 7.92 (1H) , d, J = 8.3 Hz), 8.21 (1H, d, J = 1.5 Hz).

(16) 3-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]シクロヘキサ-1-エンイル}ベンゾ[b]チオフェン-5-カルボキサミド (16) 3- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] cyclohex-1-enyl} benzo [b] thiophene-5-carboxamide

Figure 2020071550
Figure 2020071550

実施例16 (16d) で製造した化合物 0.30 g (0.78 mmol) をジメチルスルホキシド 1.5 mLに溶解し、室温にて5.0 M 水酸化ナトリウム水0.23 mL (1.2 mmol) および30% 過酸化水素水 0.12 mL (1.2 mmol) を加え、1時間撹拌後、5.0 M 水酸化ナトリウム水 0.12 mL (0.60 mmol) および30%過酸化水素水 0.06 mL (0.67 mmol) を加え、30分間撹拌した。水 10 mLを加え、酢酸エチル 20 mLにて2回抽出した。有機層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。残渣にジエチルエーテルを加え、ろ取し、標題化合物の白色粉末 161 mg (収率:51%) を得た。
IR ν (ATR) cm-1 :1655
1H-NMR (DMSO-d6, 400 MHz) δ:1.33-1.43 (2H, m), 1.68-1.78 (1H, m), 1.79-1.88 (2H, m), 1.98-2.08 (1H, m), 2.15-2.24 (1H, m), 2.47-2.64 (3H, m), 3.26-3.36 (2H, m), 3.46-3.54 (1H, m), 3.54-3.65 (4H, m), 3.67-3.74 (1H, m), 3.74-3.80 (2H, m), 5.98-6.03 (1H, m), 7.38-7.48 (1H, br), 7.64 (1H, s), 7.85 (1H, dd, J=8.3, 1.5 Hz), 8.03 (1H, d, J=8.3 Hz), 8.10-8.18 (1H, br), 8.38 (1H, d, J=1.3 Hz).
MS (ESI) m/z:402 [M+H]+.
0.30 g (0.78 mmol) of the compound prepared in Example 16 (16d) was dissolved in 1.5 mL of dimethyl sulfoxide, and at room temperature, 5.0 M sodium hydroxide water 0.23 mL (1.2 mmol) and 30% hydrogen hydrogen peroxide solution 0.12 mL ( 1.2 mmol) was added, and after stirring for 1 hour, 0.12 mL (0.60 mmol) of 5.0 M sodium hydroxide water and 0.06 mL (0.67 mmol) of 30% hydrogen hydrogen solution were added, and the mixture was stirred for 30 minutes. 10 mL of water was added, and the mixture was extracted twice with 20 mL of ethyl acetate. The organic layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. Diethyl ether was added to the residue and collected by filtration to give 161 mg (yield: 51%) of a white powder of the title compound.
IR ν (ATR) cm -1 : 1655
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 1.33-1.43 (2H, m), 1.68-1.78 (1H, m), 1.79-1.88 (2H, m), 1.98-2.08 (1H, m), 2.15-2.24 (1H, m), 2.47-2.64 (3H, m), 3.26-3.36 (2H, m), 3.46-3.54 (1H, m), 3.54-3.65 (4H, m), 3.67-3.74 (1H) , m), 3.74-3.80 (2H, m), 5.98-6.03 (1H, m), 7.38-7.48 (1H, br), 7.64 (1H, s), 7.85 (1H, dd, J = 8.3, 1.5 Hz) ), 8.03 (1H, d, J = 8.3 Hz), 8.10-8.18 (1H, br), 8.38 (1H, d, J = 1.3 Hz).
MS (ESI) m / z: 402 [M + H] + .

実施例17 Example 17

(17a) 1-フェニルアゼチジン-3-オール (17a) 1-Phenylazetidine-3-ol

Figure 2020071550
Figure 2020071550

20% Pd(OH)2-C (wet) 200 mgをメタノール 10 mLに懸濁させ、1-ベンズヒドリルアゼチジン-3-オール 2.00 g (8.36 mmol) を加え、0.3 MPa、室温にて18時間接触水素添加した。Pd(OH)2-Cをセライトにてろ過後、減圧下メタノールを留去し、残渣にn-ヘキサン 10 mLを加え、デカントにて上澄みを除いた。得られた残渣を減圧乾燥し、706 mgの粉末を得た。
得られた粉末 700 mg、ヨードベンゼン 2.77 g (13.6 mmol)、ヨウ化銅 159 mg (0.835 mmol)、2-アセチルシクロヘキサノン 0.22 mL (1.7 mmol) および炭酸セシウム 5.44 g (16.7 mmol) をN,N-ジメチルホルムアミド 4 mLに懸濁させ、50℃ にて15時間撹拌した。放冷後、水20 mLを加え、酢酸エチル 20 mLにて2回抽出した。酢酸エチル層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下酢酸エチルを留去した。残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→3:1→2:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の黄色油状物 875 mg (収率:70%) を得た。
1-ベンズヒドリルアゼチジン-3-オール 6.93 g (29.0 mmol) を用いて上記と同様に反応および処理し、標題化合物 3.34 g (収率:77%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.15-2.30 (1H, m), 3.65 (2H, dd, J=8.6, 4.5 Hz), 4.13-4.22 (2H, m), 4.69-4.80 (1H, m), 6.45-6.53 (2H, m), 6.73-6.82 (1H, m), 7.18-7.28 (2H, m).
Suspend 200 mg of 20% Pd (OH) 2- C (wet) in 10 mL of methanol, add 2.00 g (8.36 mmol) of 1-benzhydryl azetidine-3-ol, 0.3 MPa, at room temperature 18 Time contact hydrogenation was added. After filtering Pd (OH) 2- C with Celite, methanol was distilled off under reduced pressure, 10 mL of n-hexane was added to the residue, and the supernatant was removed by decanting. The obtained residue was dried under reduced pressure to obtain 706 mg of powder.
The resulting powder 700 mg, iodobenzene 2.77 g (13.6 mmol), copper iodide 159 mg (0.835 mmol), 2-acetylcyclohexanone 0.22 mL (1.7 mmol) and cesium carbonate 5.44 g (16.7 mmol) N, N- It was suspended in 4 mL of dimethylformamide and stirred at 50 ° C. for 15 hours. After allowing to cool, 20 mL of water was added, and the mixture was extracted twice with 20 mL of ethyl acetate. The ethyl acetate layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and ethyl acetate was distilled off under reduced pressure. The residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 3: 1 → 2: 1, V / V), the solvent of the target fraction was distilled off under reduced pressure, and the title compound was yellow. An oily substance of 875 mg (yield: 70%) was obtained.
Reaction and treatment with 1-benzhydryl azetidine-3-ol 6.93 g (29.0 mmol) in the same manner as above gave 3.34 g (yield: 77%) of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.15-2.30 (1H, m), 3.65 (2H, dd, J = 8.6, 4.5 Hz), 4.13-4.22 (2H, m), 4.69-4.80 (1H) , m), 6.45-6.53 (2H, m), 6.73-6.82 (1H, m), 7.18-7.28 (2H, m).

(17b) 3-[2-(tert-ブチルジメチルシラニルオキシ)エトキシ]-1-(4-ヨードフェニル)アゼチジン (17b) 3- [2- (tert-butyldimethylsilanyloxy) ethoxy] -1- (4-iodophenyl) azetidine

Figure 2020071550
Figure 2020071550

実施例17 (17a) で製造した化合物 1.81 g (12.1 mmol) および (2-ブロモエトキシ)-tert-ブチルジメチルシラン 3.19 g (13.3 mmol) をN,N-ジメチルホルムアミド 18 mLに溶解し、室温にて水素化ナトリウム (P=60%) 629 mg (16 mmol) を加え、窒素雰囲気下、同温にて4時間撹拌した。氷水 90 mLに注加し、ジエチルエーテル 100 mLおよび50 mLにて抽出した。ジエチルエーテル層を合わせ、水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下ジエチルエーテルを留去した。残渣をカラムクロマトグラフィー(ジエチルエーテル:n-ヘキサン、1:30→1:10→1:5、V/V)にて精製後、目的分画の溶媒を減圧下留去し、3-[2-(tert-ブチルジメチルシラニルオキシ)エトキシ]-1-フェニルアゼチジンの淡黄色油状物 827 mg (収率:22%) を得た。
3-[2-(tert-ブチルジメチルシラニルオキシ)エトキシ]-1-フェニルアゼチジン915 mg (2.96 mmol) を塩化メチレン 9 mLに溶解させ、重曹 373 mg (4.44 mmol) および水 9 mLを加え、室温にてヨウ素 827 mg (3.26 mmol) を15分間かけて分割添加後、同温にて1.5時間撹拌した。5% チオ硫酸ナトリウム水 10 mLを加え、減圧下塩化メチレンを留去後、酢酸エチル 20 mLにて抽出した。酢酸エチル層を5% チオ硫酸ナトリウム水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下酢酸エチルを留去した。残渣をカラムクロマトグラフィー(Chromatorex NH、ジエチルエーテル:n-ヘキサン、0:1→1:6、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の微黄色油状物 1.25 g (収率:97%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.07 (6H, s), 0.89 (9H, s), 3.51 (2H, t, J=5.1 Hz), 3.68 (2H, dd, J=8.2, 4.5 Hz), 3.76 (2H, t, J=5.1 Hz), 4.04-4.11 (2H, m), 4.43-4.53 (1H, m), 6.20-6.28 (2H, m), 7.42-7.49 (2H, m).
The compound 1.81 g (12.1 mmol) and (2-bromoethoxy) -tert-butyldimethylsilane 3.19 g (13.3 mmol) prepared in Example 17 (17a) were dissolved in 18 mL of N, N-dimethylformamide and brought to room temperature. Sodium hydride (P = 60%) 629 mg (16 mmol) was added, and the mixture was stirred under a nitrogen atmosphere at the same temperature for 4 hours. It was poured into 90 mL of ice water and extracted with 100 mL and 50 mL of diethyl ether. The diethyl ether layers were combined, washed successively with water and saturated brine, dried (Na 2 SO 4 ), and diethyl ether was distilled off under reduced pressure. The residue was purified by column chromatography (diethyl ether: n-hexane, 1:30 → 1:10 → 1: 5, V / V), the solvent for the target fraction was distilled off under reduced pressure, and 3- [2 A pale yellow oil of-(tert-butyldimethylsilanyloxy) ethoxy] -1-phenylazetidine was obtained in an amount of 827 mg (yield: 22%).
Dissolve 915 mg (2.96 mmol) of 3- [2- (tert-butyldimethylsilanyloxy) ethoxy] -1-phenylazetidine in 9 mL of methylene chloride, add 373 mg (4.44 mmol) of baking soda and 9 mL of water. After adding 827 mg (3.26 mmol) of iodine in portions over 15 minutes at room temperature, the mixture was stirred at the same temperature for 1.5 hours. 10 mL of 5% sodium thiosulfate was added, methylene chloride was distilled off under reduced pressure, and the mixture was extracted with 20 mL of ethyl acetate. The ethyl acetate layer was washed successively with 5% aqueous sodium thiosulfate solution and saturated brine, dried (Na 2 SO 4 ), and ethyl acetate was distilled off under reduced pressure. The residue was purified by column chromatography (Chromatorex NH, diethyl ether: n-hexane, 0: 1 → 1: 6, V / V), the solvent of the target fraction was distilled off under reduced pressure, and the title compound was slightly yellow. An oily substance of 1.25 g (yield: 97%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.07 (6H, s), 0.89 (9H, s), 3.51 (2H, t, J = 5.1 Hz), 3.68 (2H, dd, J = 8.2, 4.5) Hz), 3.76 (2H, t, J = 5.1 Hz), 4.04-4.11 (2H, m), 4.43-4.53 (1H, m), 6.20-6.28 (2H, m), 7.42-7.49 (2H, m) ..

(17c) 1-(4-{3-[2-(tert-ブチルジメチルシラニルオキシ)エトキシ]アゼチジン-1-イル}フェニル)-5,6-ジメトキシ-2-メチルインドール (17c) 1- (4- {3- [2- (tert-butyldimethylsilanyloxy) ethoxy] azetidine-1-yl} phenyl) -5,6-dimethoxy-2-methylindole

Figure 2020071550
Figure 2020071550

実施例1 (1b) で製造した化合物 658 mg (3.44 mmol)、実施例17 (17b) で製造した化合物 1.25 g (2.88 mmol)、ヨウ化銅 27 mg (0.14 mmol)、trans-N,N’-ジメチルシクロヘキサン-1,2-ジアミン 61 mg (0.43 mmol) およびリン酸三カリウム 1.28 g (6.03 mmol) をトルエン 3 mLに懸濁させ、窒素をバブリング後、窒素雰囲気下100℃ にて15時間撹拌した。放冷後、水 20 mLおよび酢酸エチル 40 mLを加え、不溶物をろ別後、ろ液の二層を分離した。酢酸エチル層を水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下酢酸エチルを留去した。残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、10:1→5:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の黄色粉末 755 mg (収率:53%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:0.09 (6H, s), 0.91 (9H, s), 2.21 (3H, s), 3.55 (2H, t, J=5.1 Hz), 3.78 (3H, s), 3.78-3.85 (4H, m), 3.91 (3H, s), 4.16-4.24 (2H, m), 4.50-4.60 (1H, m), 6.22 (1H, s), 6.53-6.60 (3H, m), 7.01 (1H, s), 7.12-7.19 (2H, m).
Compound 658 mg (3.44 mmol) prepared in Example 1 (1b), compound 1.25 g (2.88 mmol) prepared in Example 17 (17b), copper iodide 27 mg (0.14 mmol), trans-N, N'. -Dimethylcyclohexane-1,2-diamine 61 mg (0.43 mmol) and tripotassium phosphate 1.28 g (6.03 mmol) are suspended in 3 mL of toluene, nitrogen is bubbled, and then stirred at 100 ° C. for 15 hours under a nitrogen atmosphere. bottom. After allowing to cool, 20 mL of water and 40 mL of ethyl acetate were added, the insoluble material was filtered off, and the two layers of the filtrate were separated. The ethyl acetate layer was washed successively with water and saturated brine, dried (Na 2 SO 4 ), and ethyl acetate was distilled off under reduced pressure. The residue was purified by column chromatography (n-hexane: ethyl acetate, 10: 1 → 5: 1, V / V), the solvent of the target fraction was distilled off under reduced pressure, and the yellow powder of the title compound was 755 mg (5). Yield: 53%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 0.09 (6H, s), 0.91 (9H, s), 2.21 (3H, s), 3.55 (2H, t, J = 5.1 Hz), 3.78 (3H, s) s), 3.78-3.85 (4H, m), 3.91 (3H, s), 4.16-4.24 (2H, m), 4.50-4.60 (1H, m), 6.22 (1H, s), 6.53-6.60 (3H, s) m), 7.01 (1H, s), 7.12-7.19 (2H, m).

(17d) 2-{1-[4-(5,6-ジメトキシ-2-メチルインドール-1-イル)フェニル]アゼチジン-3-イルオキシ}エチル アセテート (17d) 2- {1- [4- (5,6-dimethoxy-2-methylindole-1-yl) phenyl] azetidine-3-yloxy} ethyl acetate

Figure 2020071550
Figure 2020071550

実施例17 (17c) で製造した化合物 743 mg (1.38 mmol) に室温にて1.0 M フッ化テトラ-n-ブチルアンモニウム テトラヒドロフラン溶液 4.1 mL (4.1 mmol) を加え、同温にて50分間撹拌した。減圧下テトラヒドロフランを留去後、残渣に水 20 mLを加え、酢酸エチル 50 mLにて抽出し、酢酸エチル層を水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下酢酸エチルを留去した。
得られた残渣を塩化メチレン 7 mLに溶解させ、氷冷下トリエチルアミン 0.38 mL (2.7 mmol) および塩化アセチル 0.15 mL (2.1 mmol) を加え、室温にて30分間撹拌した。水 20 mLおよび塩化メチレン 20 mLを加え、二層を分離後、塩化メチレン層を水および飽和食塩水にて順次洗浄し、乾燥(Na2SO4) した。減圧下塩化メチレンを留去し、残渣をカラムクロマトグラフィー(Chromatorex NH、n-ヘキサン:酢酸エチル、6:1→4:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 461 mg (収率:79%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.11 (3H, s), 2.21 (3H, s), 3.69 (2H, t, J=4.8 Hz), 3.78 (3H, s), 3.84 (2H, dd, J=8.3, 4.9 Hz), 3.91 (3H, s), 4.19-4.30 (4H, m), 4.49-4.59 (1H, m), 6.22 (1H, s), 6.54 (1H, s), 6.54-6.60 (3H, m), 7.01 (1H, s), 7.13-7.20 (2H, m).
To 743 mg (1.38 mmol) of the compound prepared in Example 17 (17c) was added 4.1 mL (4.1 mmol) of a 1.0 M tetra-n-butylammonium tetrahydrofuran tetrahydrofuran solution at room temperature, and the mixture was stirred at the same temperature for 50 minutes. After distilling off tetrahydrofuran under reduced pressure, add 20 mL of water to the residue, extract with 50 mL of ethyl acetate, wash the ethyl acetate layer sequentially with water and saturated brine, dry (Na 2 SO 4 ), and reduce the pressure. The lower ethyl acetate was distilled off.
The obtained residue was dissolved in 7 mL of methylene chloride, 0.38 mL (2.7 mmol) of triethylamine and 0.15 mL (2.1 mmol) of acetyl chloride were added under ice-cooling, and the mixture was stirred at room temperature for 30 minutes. After adding 20 mL of water and 20 mL of methylene chloride and separating the two layers, the methylene chloride layer was washed successively with water and saturated brine, and dried (Na 2 SO 4 ). Methylene chloride is distilled off under reduced pressure, the residue is purified by column chromatography (Chromatorex NH, n-hexane: ethyl acetate, 6: 1 → 4: 1, V / V), and then the solvent of the target fraction is reduced under reduced pressure. Distillation gave 461 mg (yield: 79%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.11 (3H, s), 2.21 (3H, s), 3.69 (2H, t, J = 4.8 Hz), 3.78 (3H, s), 3.84 (2H, 2H, dd, J = 8.3, 4.9 Hz), 3.91 (3H, s), 4.19-4.30 (4H, m), 4.49-4.59 (1H, m), 6.22 (1H, s), 6.54 (1H, s), 6.54 -6.60 (3H, m), 7.01 (1H, s), 7.13-7.20 (2H, m).

(17e) 2-{1-[4-(3-アセチル-5,6-ジメトキシ-2-メチルインドール-1-イル)フェニル]アゼチジン-3-イルオキシ}エチル アセテート (17e) 2- {1- [4- (3-Acetyl-5,6-dimethoxy-2-methylindole-1-yl) phenyl] azetidine-3-yloxy} ethyl acetate

Figure 2020071550
Figure 2020071550

実施例17 (17d) で製造した化合物 454 mg (1.07 mmol) を塩化メチレン 5.5 mLに溶解し、氷冷下塩化アセチル 0.11 mL (1.5 mmol) および塩化チタン(IV) 0.18 mL (1.6 mmol) を順に加え、同温にて30分間撹拌した。氷水 20 mLを加え、室温にて析出物が溶解するまで撹拌後、二層を分離した。塩化メチレン層を飽和重曹水および飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) し、減圧下塩化メチレンを留去した。残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、3:1→2:1→1:1→2:3、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の白色粉末 120 mg (収率:24%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.11 (3H, s), 2.50 (3H, s), 2.65 (3H, s), 3.70 (2H, t, J=4.8 Hz), 3.77 (3H, s), 3.86 (2H, dd, J=8.3, 4.6 Hz), 3.98 (3H, s), 4.20-4.30 (4H, m), 4.51-4.60 (1H, m), 6.45 (1H, s), 6.54-6.60 (2H, m), 7.01 (1H, s), 7.13-7.20 (2H, m).
Compound 454 mg (1.07 mmol) prepared in Example 17 (17d) was dissolved in 5.5 mL of methylene chloride, followed by ice-cooled acetyl chloride 0.11 mL (1.5 mmol) and titanium (IV) chloride 0.18 mL (1.6 mmol). In addition, the mixture was stirred at the same temperature for 30 minutes. 20 mL of ice water was added, and the mixture was stirred at room temperature until the precipitate dissolved, and then the two layers were separated. The methylene chloride layer was washed successively with saturated aqueous sodium hydrogen carbonate and saturated brine, dried (Na 2 SO 4 ), and methylene chloride was distilled off under reduced pressure. The residue was purified by column chromatography (n-hexane: ethyl acetate, 3: 1 → 2: 1 → 1: 1 → 2: 3, V / V), and then the solvent for the target fraction was distilled off under reduced pressure. A white powder of the title compound (120 mg) (yield: 24%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.11 (3H, s), 2.50 (3H, s), 2.65 (3H, s), 3.70 (2H, t, J = 4.8 Hz), 3.77 (3H, s) s), 3.86 (2H, dd, J = 8.3, 4.6 Hz), 3.98 (3H, s), 4.20-4.30 (4H, m), 4.51-4.60 (1H, m), 6.45 (1H, s), 6.54 -6.60 (2H, m), 7.01 (1H, s), 7.13-7.20 (2H, m).

(17) 2-{1-[4-(3-アセチル-5,6-ジメトキシ-2-メチルインドール-1-イル)フェニル]アゼチジン-3-イルオキシ}エタノール (17) 2- {1- [4- (3-Acetyl-5,6-dimethoxy-2-methylindole-1-yl) phenyl] azetidine-3-yloxy} ethanol

Figure 2020071550
Figure 2020071550

実施例17 (17e) で製造した化合物 115 mg (0.247 mmol) を塩化メチレン 1 mLに溶解し、室温にてメタノール 1 mLおよび5.0 M水酸化ナトリウム水 0.24 mL (1.2 mmol) を加え、同温にて2時間撹拌した。減圧下溶媒を留去後、水 10 mLを加え、析出物が溶解するまでメタノールを加えた。減圧下メタノールを留去後、析出物をろ取し、標題化合物の白色粉末 98 mg (収率:85%) を得た。
IR ν (ATR) cm-1 :1608
1H-NMR (CDCl3, 400 MHz) δ:1.85-2.00 (1H, br), 2.50 (3H, s), 2.64 (3H, s), 3.60 (2H, t, J=4.5 Hz), 3.76-3.84 (2H, m), 3.77 (3H, s), 3.87 (2H, dd, J=7.9, 4.3 Hz), 3.98 (3H, s), 4.20-4.28 (2H, m), 4.51-4.60 (1H, m), 6.45 (1H, s), 6.56-6.62 (2H, m), 7.09-7.16 (2H, m), 7.68 (1H, s).
MS m/z: 427 [M+H]+.
115 mg (0.247 mmol) of the compound prepared in Example 17 (17e) was dissolved in 1 mL of methylene chloride, and 1 mL of methanol and 0.24 mL (1.2 mmol) of 5.0 M sodium hydroxide water were added at room temperature to the same temperature. Stirred for 2 hours. After distilling off the solvent under reduced pressure, 10 mL of water was added, and methanol was added until the precipitate was dissolved. After distilling off methanol under reduced pressure, the precipitate was collected by filtration to give 98 mg (yield: 85%) of a white powder of the title compound.
IR ν (ATR) cm -1 : 1608
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.85-2.00 (1H, br), 2.50 (3H, s), 2.64 (3H, s), 3.60 (2H, t, J = 4.5 Hz), 3.76- 3.84 (2H, m), 3.77 (3H, s), 3.87 (2H, dd, J = 7.9, 4.3 Hz), 3.98 (3H, s), 4.20-4.28 (2H, m), 4.51-4.60 (1H, m) m), 6.45 (1H, s), 6.56-6.62 (2H, m), 7.09-7.16 (2H, m), 7.68 (1H, s).
MS m / z: 427 [M + H] + .

実施例18 Example 18

(18a) 3-カルバモイル-5,6-ジメトキシ-2-メチル-1-{4-[3-(2-アセトキシエトキシ)アゼチジン-1-イル]フェニル}インドール (18a) 3-Carbamoyl-5,6-dimethoxy-2-methyl-1-{4- [3- (2-acetoxyethoxy) azetidine-1-yl] phenyl} indole

Figure 2020071550
Figure 2020071550

実施例17 (17d) で製造した化合物 200 mg (0.470 mmol) を塩化メチレン 2 mLに溶解し、-78℃ にてクロロスルホニルイソシアネート 0.062 mL (0.71 mmol) を滴下し、同温にて5分間撹拌した。反応液にジイソプロピルエーテルを加え、析出した粉末をろ取し、白色粉末300 mgを得た。
得られた白色粉末 300 mg を酢酸 2 mLに溶解し、室温にて30分間撹拌した。反応液を飽和重曹水にて中和し、クロロホルムにて2回抽出した。クロロホルム層を合わせ、飽和食塩水にて洗浄後、乾燥 (Na2SO4) した。減圧下クロロホルムを留去し、標題化合物の白色粉末 200 mg (収率:91%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.11 (3H, s), 2.48 (3H, s), 3.65-3.72 (2H, m), 3.78 (3H, s), 3.83-3.87 (2H, m), 3.96 (3H, s), 4.17-4.30 (4H, m), 4.49-4.57 (1H, m), 5.55-5.67 (2H, br), 6.49 (1H, s), 6.56-6.58 (2H, m), 7.10-7.13 (2H, m), 7.37 (1H, s).
200 mg (0.470 mmol) of the compound prepared in Example 17 (17d) was dissolved in 2 mL of methylene chloride, 0.062 mL (0.71 mmol) of chlorosulfonyl isocyanate was added dropwise at -78 ° C, and the mixture was stirred at the same temperature for 5 minutes. bottom. Diisopropyl ether was added to the reaction solution, and the precipitated powder was collected by filtration to obtain 300 mg of white powder.
The obtained white powder (300 mg) was dissolved in 2 mL of acetic acid, and the mixture was stirred at room temperature for 30 minutes. The reaction mixture was neutralized with saturated aqueous sodium hydrogen carbonate and extracted twice with chloroform. The chloroform layers were combined, washed with saturated brine, and dried (Na 2 SO 4 ). Chloroform was distilled off under reduced pressure to obtain 200 mg (yield: 91%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.11 (3H, s), 2.48 (3H, s), 3.65-3.72 (2H, m), 3.78 (3H, s), 3.83-3.87 (2H, m) ), 3.96 (3H, s), 4.17-4.30 (4H, m), 4.49-4.57 (1H, m), 5.55-5.67 (2H, br), 6.49 (1H, s), 6.56-6.58 (2H, m) ), 7.10-7.13 (2H, m), 7.37 (1H, s).

(18) 3-カルバモイル-5,6-ジメトキシ-2-メチル-1-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}インドール (18) 3-Carbamoyl-5,6-dimethoxy-2-methyl-1-{4- [3- (2-hydroxyethoxy) azetidine-1-yl] phenyl} indole

Figure 2020071550
Figure 2020071550

実施例18 (18a) で製造した化合物 200 mg (0.430 mmol) を塩化メチレン-メタノール (1:1) の混液 4 mLに溶解し、室温にて5.0 M 水酸化ナトリウム水 0.43 mL (2.2 mmol) を加え、同温にて30分間撹拌した。反応液に水 4 mLを加え、減圧下溶媒を留去した。析出した粉末をろ取し、ジイソプロピルエーテルにて洗浄し、標題化合物の白色粉末 130 mg (収率:71%) を得た。
IR ν (ATR) cm-1 :3220-3050, 1648.
1H-NMR (CDCl3, 400 MHz) δ:1.96 (1H, t, J=6.0 Hz), 2.47 (3H, s), 3.57-3.62 (2H, m), 3.78 (3H, s), 3.78-3.89 (4H, m), 3.96 (3H, s), 4.20-4.23 (2H, m), 4.50-4.60 (1H, m), 5.58-5.70 (2H, br), 6.49 (1H, s), 6.57-6.59 (2H, m), 7.10-7.13 (2H, m), 7.37 (1H, s).
MS (EMS) m/z:426 [M+H+].
200 mg (0.430 mmol) of the compound prepared in Example 18 (18a) was dissolved in 4 mL of a mixture of methylene chloride-methanol (1: 1), and at room temperature 5.0 M sodium hydroxide water 0.43 mL (2.2 mmol) was added. In addition, the mixture was stirred at the same temperature for 30 minutes. 4 mL of water was added to the reaction mixture, and the solvent was distilled off under reduced pressure. The precipitated powder was collected by filtration and washed with diisopropyl ether to give 130 mg (yield: 71%) of a white powder of the title compound.
IR ν (ATR) cm -1 : 3220-3050, 1648.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.96 (1H, t, J = 6.0 Hz), 2.47 (3H, s), 3.57-3.62 (2H, m), 3.78 (3H, s), 3.78- 3.89 (4H, m), 3.96 (3H, s), 4.20-4.23 (2H, m), 4.50-4.60 (1H, m), 5.58-5.70 (2H, br), 6.49 (1H, s), 6.57- 6.59 (2H, m), 7.10-7.13 (2H, m), 7.37 (1H, s).
MS (EMS) m / z: 426 [M + H + ].

実施例19 Example 19

(19a) 1-(4-ニトロフェニル)-3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン (19a) 1- (4-Nitrophenyl) -3- [2- (Tetrahydropyran-2-yloxy) ethoxy] Azetidine

Figure 2020071550
Figure 2020071550

20% 水酸化パラジウム炭素 3.0 g をメタノール 250 mL に懸濁し、実施例9 (9c) で製造した 1-ベンズヒドリル-3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン 23.8 g (64.8 mmol) を加え、25℃、0.40 MPa にて3時間接触水素添加した。不溶物をろ別後、減圧下溶媒を留去し、3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジンの褐色油状物 23.3 g (収率:98%) を得た。
3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン 3.00 g (8.16 mmol) をN,N-ジメチルホルムアミド 30 mL に溶解し、4-ニトロフルオロベンゼン 1.04 mL (9.79 mmol)、ジイソプロピルエチルアミン 2.13 mL (12.2 mmol) を加え、窒素雰囲気下、60℃ にて15時間撹拌した。反応液を室温に放冷後、水を加え、酢酸エチルにて2回抽出した。有機層を合わせ、水および飽和食塩水にて洗浄後、乾燥 (Na2SO4) した。減圧下溶媒を留去し、得られた残渣をシリカゲルカラムクロマトグラフィー (n-ヘキサン:酢酸エチル、4:1→1:1、V/V) にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の黄色油状物 2.08 g (収率:79%) を得た。
1H-NMR (CDCl3, 400 MHz) δ : 1.50-1.88 (6H, m),3.48-3.64 (2H, m),3.65-3.69 (2H, m),3.84-3.98 (4H, m),4.20-4.26 (2H, m),4.53-4.57 (1H,m),4.61-4.63 (1H, m),6.30-6.32 (2H, m),8.08-8.11 (2H, m).
1-Benz hydryl-3- [2- (tetrahydropyran-2-yloxy) ethoxy] azetidine prepared in Example 9 (9c) by suspending 3.0 g of 20% palladium hydroxide in 250 mL of methanol 23.8 g (64.8 mmol). ) Was added, and catalytic hydrogenation was performed at 25 ° C. and 0.40 MPa for 3 hours. After the insoluble material was filtered off, the solvent was distilled off under reduced pressure to obtain 23.3 g (yield: 98%) of a brown oil of 3- [2- (tetrahydropyran-2-yloxy) ethoxy] azetidine.
3- [2- (Tetrahydropyran-2-yloxy) ethoxy] azetidine 3.00 g (8.16 mmol) was dissolved in 30 mL of N, N-dimethylformamide, 4-nitrofluorobenzene 1.04 mL (9.79 mmol), diisopropylethylamine 2.13. mL (12.2 mmol) was added, and the mixture was stirred at 60 ° C. for 15 hours under a nitrogen atmosphere. The reaction mixture was allowed to cool to room temperature, water was added, and the mixture was extracted twice with ethyl acetate. The organic layers were combined, washed with water and saturated brine, and dried (Na 2 SO 4 ). The solvent was evaporated under reduced pressure, and the obtained residue was purified by silica gel column chromatography (n-hexane: ethyl acetate, 4: 1 → 1: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 2.08 g (yield: 79%) of a yellow oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.50-1.88 (6H, m), 3.48-3.64 (2H, m), 3.65-3.69 (2H, m), 3.84-3.98 (4H, m), 4.20 -4.26 (2H, m), 4.53-4.57 (1H, m), 4.61-4.63 (1H, m), 6.30-6.32 (2H, m), 8.08-8.11 (2H, m).

(19b) 4-{3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン-1-イル}フェニルアミン (19b) 4- {3- [2- (Tetrahydropyran-2-yloxy) ethoxy] Azetidine-1-yl} Phenylamine

Figure 2020071550
Figure 2020071550

5%パラジウム炭素 500 mg をメタノール 30 mL に懸濁し、実施例19 (19a) で製造した化合物 2.07 g (6.42 mmol) を加え、25℃、0.40 MPa にて1時間接触水素添加した。不溶物をろ別後、減圧下溶媒を留去し、標題化合物の褐色油状物 1.9 g (収率:定量的) を得た。
1H-NMR (CDCl3, 400 MHz) δ : 1.46-1.88 (6H, m),3.25-3.40 (2H, br),3.45-3.55 (1H, m),3.56-3.65 (5H, m),3.83-3.90 (2H, m),4.02-4.06 (2H, m),4.44-4.47 (1H,m),4.61-4.63 (1H, m),6.35-6.37 (2H, m),6.61-6.63 (2H, m).
500 mg of 5% palladium carbon was suspended in 30 mL of methanol, 2.07 g (6.42 mmol) of the compound prepared in Example 19 (19a) was added, and catalytic hydrogenation was carried out at 25 ° C. and 0.40 MPa for 1 hour. After the insoluble material was filtered off, the solvent was distilled off under reduced pressure to obtain 1.9 g (yield: quantitative) of a brown oily substance of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.46-1.88 (6H, m), 3.25-3.40 (2H, br), 3.45-3.55 (1H, m), 3.56-3.65 (5H, m), 3.83 -3.90 (2H, m), 4.02-4.06 (2H, m), 4.44-4.47 (1H, m), 4.61-4.63 (1H, m), 6.35-6.37 (2H, m), 6.61-6.63 (2H, m) m).

(19c) N-(4-ヨード-6-メトキシピリジン-3-イル)-2,2-ジメチルプロピオンアミド (19c) N- (4-iodo-6-methoxypyridin-3-yl) -2,2-dimethylpropionamide

Figure 2020071550
Figure 2020071550

6-メトキシピリジン-3-イルアミン 13.2 g (10.6 mmol) を塩化メチレン 130 mLに溶解し、氷冷下トリエチルアミン 17.7 mL (12.7 mmol)、塩化ピバロイル 13.6 mL (11.1 mmol) を加え、同温にて30分間撹拌した。反応液を水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) した。減圧下塩化メチレンを留去し、N-(6-メトキシピリジン-3-イル)-2,2-ジメチルプロピオンアミドの微赤色粉末 22.0 g (収率:定量的) を得た。
N-(6-メトキシピリジン-3-イル)-2,2-ジメチルプロピオンアミド 25.2 g (0.120 mol) をテトラヒドロフラン 200 mLに溶解し、-78℃ にて1.7 M tert-ブチルリチウム ペンタン溶液 200 mL (0.34 mol) を1時間かけて滴下した。同温にて30分間撹拌後、ヨウ素 55.2 g (0.220 mol) のテトラヒドロフラン 200 mLの溶液を1時間かけて滴下後、室温にて2時間撹拌した。反応液に水を加え、酢酸エチルにて2回抽出した。酢酸エチル層を水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) した。減圧下酢酸エチルを留去し、得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、40:1→8:1→6:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡褐色粉末 15.0 g (収率:37%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.36 (9H, s), 3.90 (3H, s), 7.23 (1H, s), 7.33-7.40 (1H, br), 8.69 (1H, s).
13.2 g (10.6 mmol) of 6-methoxypyridin-3-ylamine is dissolved in 130 mL of methylene chloride, 17.7 mL (12.7 mmol) of triethylamine and 13.6 mL (11.1 mmol) of pivaloyl chloride are added under ice-cooling, and 30 at the same temperature. Stir for minutes. The reaction mixture was washed successively with water and saturated brine, and dried (Na 2 SO 4 ). Methylene chloride was distilled off under reduced pressure to obtain 22.0 g (yield: quantitative) of a slightly red powder of N- (6-methoxypyridin-3-yl) -2,2-dimethylpropionamide.
Dissolve 25.2 g (0.120 mol) of N- (6-methoxypyridin-3-yl) -2,2-dimethylpropionamide in 200 mL of tetrahydrofuran and at -78 ° C, add 200 mL of 1.7 M tert-butyllithium pentane solution (200 mL). 0.34 mol) was added dropwise over 1 hour. After stirring at the same temperature for 30 minutes, a solution of 55.2 g (0.220 mol) of iodine in 200 mL of tetrahydrofuran was added dropwise over 1 hour, and the mixture was stirred at room temperature for 2 hours. Water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate. The ethyl acetate layer was washed successively with water and saturated brine, and dried (Na 2 SO 4 ). Ethyl acetate was distilled off under reduced pressure, and the obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 40: 1 → 8: 1 → 6: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 15.0 g (yield: 37%) of a light brown powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.36 (9H, s), 3.90 (3H, s), 7.23 (1H, s), 7.33-7.40 (1H, br), 8.69 (1H, s).

(19d) 4-ヨード-6-メトキシピリジン-3-イルアセタミド (19d) 4-Iodine-6-methoxypyridin-3-ylacetamide

Figure 2020071550
Figure 2020071550

実施例19 (19c) で製造した化合物 15.0 g (44.9 mmol) に10% 硫酸水 165 mLを加え、8時間加熱還流した。反応液を重曹にて中和後、酢酸エチルにて2回抽出し、乾燥 (Na2SO4) した。減圧下酢酸エチルを留去し、得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、3:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、4-ヨード-6-メトキシピリジン-3-イルアミンの淡褐色粉末 6.0 g (収率:54%) を得た。
4-ヨード-6-メトキシピリジン-3-イルアミン5.10 g (20.4 mmol) を塩化メチレン 50 mLに溶解し、氷冷下トリエチルアミン 4.27 mL (30.6 mmol)、塩化アセチル 1.74 mL (24.5 mmol) を加え、室温にて1時間撹拌した。反応液を水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) した。減圧下塩化メチレンを留去し、残渣にジイソプロピルエーテルを加え、ろ取し、標題化合物の白色粉末 4.60 g (収率:77%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.24 (3H, s), 3.91 (3H, s), 6.96-7.10 (1H, br), 7.24 (1H, s), 8.59 (1H, s).
To 15.0 g (44.9 mmol) of the compound prepared in Example 19 (19c), 165 mL of 10% sulfuric acid water was added, and the mixture was heated under reflux for 8 hours. The reaction mixture was neutralized with sodium bicarbonate, extracted twice with ethyl acetate, and dried (Na 2 SO 4 ). Ethyl acetate was distilled off under reduced pressure, and the obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 3: 1, V / V). The solvent of the target fraction was distilled off under reduced pressure to obtain 6.0 g (yield: 54%) of a light brown powder of 4-iodo-6-methoxypyridin-3-ylamine.
Dissolve 5.10 g (20.4 mmol) of 4-iodo-6-methoxypyridin-3-ylamine in 50 mL of methylene chloride, add 4.27 mL (30.6 mmol) of triethylamine and 1.74 mL (24.5 mmol) of acetyl chloride under ice-cooling, and add to room temperature. Was stirred for 1 hour. The reaction mixture was washed successively with water and saturated brine, and dried (Na 2 SO 4 ). Dichloromethane chloride was distilled off under reduced pressure, diisopropyl ether was added to the residue, and the mixture was collected by filtration to obtain 4.60 g (yield: 77%) of a white powder of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.24 (3H, s), 3.91 (3H, s), 6.96-7.10 (1H, br), 7.24 (1H, s), 8.59 (1H, s).

(19e) N-[6-メトキシ-4-(4-{3-[2-(テトラヒドロピラン-2-イルオキシ)エトキシ]アゼチジン-1-イル}フェニルアミノ)ピリジン-3-イル]アセタミド (19e) N- [6-methoxy-4- (4- {3- [2- (tetrahydropyran-2-yloxy) ethoxy] azetidine-1-yl} phenylamino) pyridin-3-yl] acetamide

Figure 2020071550
Figure 2020071550

実施例19 (19b) で製造した化合物 3.46 g (11.8 mmol) をtert-ブタノール 35 mLに溶解し、実施例19 (19d) で製造した化合物 3.46 g (11.8 mmol)、トリス(ジベンジリデンアセトン)ジパラジウム(0) 540 mg (0.590 mmol)、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 562 mg (1.18 mmol)、リン酸三カリウム5.00 g (23.6 mmol) を加え、窒素雰囲気下、2時間加熱還流した。反応液に水を加え、酢酸エチルにて2回抽出し、有機層を合わせ、水、飽和食塩水にて順次洗浄後、乾燥 (Na2SO4) した。減圧下酢酸エチルを留去し、得られた残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、3:1、クロロホルム:メタノール、50:1→20:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡褐色油状物 4.50 g (収率:83%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.50-1.80 (6H, m), 1.96 (1H, s), 2.24 (2H, s), 3.46-3.92 (11H, m), 4.05-4.20 (2H, m), 4.45-4.55 (1H, m), 4.60-4.70 (1H, m), 6.07-6.30 (2H, m), 6.40-6.54 (2H, m), 6.95-7.10 (2H, m), 7.76-7.84 (1H, s).
Compound 3.46 g (11.8 mmol) prepared in Example 19 (19b) was dissolved in 35 mL of tert-butanol, and compound 3.46 g (11.8 mmol) prepared in Example 19 (19d), tris (dibenzylideneacetone) di Add 540 mg (0.590 mmol) of palladium (0), 562 mg (1.18 mmol) of 2-dicyclohexylphosphino-2', 4', 6'-triisopropylbiphenyl, 5.00 g (23.6 mmol) of tripotassium phosphate, and nitrogen. The mixture was heated and refluxed for 2 hours in an atmosphere. Water was added to the reaction solution, the mixture was extracted twice with ethyl acetate, the organic layers were combined, washed successively with water and saturated brine, and dried (Na 2 SO 4 ). Ethyl acetate was distilled off under reduced pressure, and the obtained residue was purified by column chromatography (n-hexane: ethyl acetate, 3: 1, chloroform: methanol, 50: 1 → 20: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure to give 4.50 g (yield: 83%) of a light brown oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.50-1.80 (6H, m), 1.96 (1H, s), 2.24 (2H, s), 3.46-3.92 (11H, m), 4.05-4.20 (2H) , m), 4.45-4.55 (1H, m), 4.60-4.70 (1H, m), 6.07-6.30 (2H, m), 6.40-6.54 (2H, m), 6.95-7.10 (2H, m), 7.76 -7.84 (1H, s).

(19f) 2-{1-[4-(6-メトキシ-2-メチル-イミダゾ[4,5-c]ピリジン-1-イル)フェニル]アゼチジン-3-イルオキシ}エチル アセテート (19f) 2- {1- [4- (6-Methoxy-2-methyl-imidazole [4,5-c] pyridin-1-yl) phenyl] azetidine-3-yloxy} ethyl acetate

Figure 2020071550
Figure 2020071550

実施例19 (19e) で製造した化合物 4.50 g (9.85 mmol) を酢酸 20 mL、無水酢酸 20 mLに溶解し、窒素雰囲気下、80℃ にて5時間撹拌後、酢酸 10mL、無水酢酸10 mLを追加し、同温にて更に3時間撹拌した。反応液を飽和重曹水にて中和し、酢酸エチルにて2回抽出後、水、飽和食塩水にて順次洗浄、乾燥 (Na2SO4) した。減圧下酢酸エチルを留去し、得られた残渣をカラムクロマトグラフィー(Chromatorex NH、n-ヘキサン:酢酸エチル、1:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、標題化合物の淡褐色油状物 2.07 g (収率:53%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:2.11 (3H, s), 2.43 (3H, s), 3.65-3.75 (2H, m), 3.80-3.88 (2H, m), 3.94 (3H, s), 4.18-4.30 (4H, m), 4.50-4.60 (1H, m), 6.39 (1H, s), 6.55-6.57 (2H, m), 7.11-7.14 (2H, m), 8.56 (1H, s).
4.50 g (9.85 mmol) of the compound prepared in Example 19 (19e) was dissolved in 20 mL of acetic acid and 20 mL of acetic anhydride, and after stirring at 80 ° C. for 5 hours in a nitrogen atmosphere, 10 mL of acetic acid and 10 mL of acetic anhydride were added. The mixture was added and stirred at the same temperature for another 3 hours. The reaction mixture was neutralized with saturated aqueous sodium hydrogen carbonate, extracted twice with ethyl acetate, washed successively with water and saturated brine, and dried (Na 2 SO 4 ). Ethyl acetate was distilled off under reduced pressure, and the obtained residue was purified by column chromatography (Chromatorex NH, n-hexane: ethyl acetate, 1: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure to give 2.07 g (yield: 53%) of a light brown oil of the title compound.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 2.11 (3H, s), 2.43 (3H, s), 3.65-3.75 (2H, m), 3.80-3.88 (2H, m), 3.94 (3H, s) ), 4.18-4.30 (4H, m), 4.50-4.60 (1H, m), 6.39 (1H, s), 6.55-6.57 (2H, m), 7.11-7.14 (2H, m), 8.56 (1H, s) ).

(19) 2-{1-[4-(6-メトキシ-2-メチルイミダゾ[4,5-c]ピリジン-1-イル)フェニル]アゼチジン-3-イルオキシ}エタノール (19) 2- {1- [4- (6-Methoxy-2-methylimidazole [4,5-c] pyridin-1-yl) phenyl] azetidine-3-yloxy} ethanol

Figure 2020071550
Figure 2020071550

実施例19 (19f) で製造した化合物 2.07 g (5.22 mmol) をメタノール 20 mLに溶解し、室温にて 5.0 M 水酸化ナトリウム水 3.1 mL (15 mmol) を加え、同温にて30分間撹拌した。減圧下メタノールを留去し、残渣に水を加え、クロロホルムにて抽出した。クロロホルム層を飽和食塩水にて洗浄後、乾燥(Na2SO4)し、減圧下溶媒を留去した。残渣にジイソプロピルエーテルを加え、不溶物をろ取し、得られた粉末をカラムクロマトグラフィー(Chromatorex NH、n-ヘキサン:酢酸エチル、1:1→0:1、V/V)にて精製した。目的分画の溶媒を減圧下留去し、残渣にジイソプロピルエーテルを加え、不溶物をろ取し、標題化合物の微紅色粉末 1.35 g (収率:73%) を得た。
IR ν (ATR) cm-1 :3100-3200, 1623
1H-NMR (DMSO-d6, 400 MHz) δ:2.35 (3H, s), 3.40-3.60 (4H, m), 3.68-3.78 (2H, m), 3.83 (3H, s), 4.10-4.20 (2H, m), 4.44-4.54 (1H, m), 4.65-4.73 (1H, m), 6.31 (1H, s), 6.58-6.60 (2H, m), 7.27-7.30 (2H, m), 8.45 (1H, s).
MS m/z: 355 [M+H]+, 353 [M-H]-.
2.07 g (5.22 mmol) of the compound prepared in Example 19 (19f) was dissolved in 20 mL of methanol, 3.1 mL (15 mmol) of 5.0 M sodium hydroxide water was added at room temperature, and the mixture was stirred at the same temperature for 30 minutes. .. Methanol was distilled off under reduced pressure, water was added to the residue, and the mixture was extracted with chloroform. The chloroform layer was washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. Diisopropyl ether was added to the residue, the insoluble material was collected by filtration, and the obtained powder was purified by column chromatography (Chromatorex NH, n-hexane: ethyl acetate, 1: 1 → 0: 1, V / V). The solvent of the target fraction was evaporated under reduced pressure, diisopropyl ether was added to the residue, and the insoluble material was collected by filtration to obtain 1.35 g (yield: 73%) of a slightly red powder of the title compound.
IR ν (ATR) cm -1 : 3100-3200, 1623
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 2.35 (3H, s), 3.40-3.60 (4H, m), 3.68-3.78 (2H, m), 3.83 (3H, s), 4.10-4.20 ( 2H, m), 4.44-4.54 (1H, m), 4.65-4.73 (1H, m), 6.31 (1H, s), 6.58-6.60 (2H, m), 7.27-7.30 (2H, m), 8.45 ( 1H, s).
MS m / z: 355 [M + H] + , 353 [MH] - .

実施例20 Example 20

(20a) 7-メトキシキノリン-4-オール (20a) 7-Methoxyquinoline-4-ol

Figure 2020071550
Figure 2020071550

3-メトキシアニリン 12.3 g (0.100 mol) にエトキシメチレンマロン酸ジエチル 21.6 g (0.100 mol) を加え、110℃ にて75分間撹拌後、ジフェニルエーテル 150 mL を加え、230℃ にて8.5時間撹拌した。反応液を室温まで冷却し、酢酸エチル 200 mLに注加した。析出物をろ取後、酢酸エチルにて洗浄し、エチル 4-ヒドロキシ-7-メトキシキノリン-3-カルボキシレートの淡褐色粉末 8.56 g (収率:35%) を得た。
エチル 4-ヒドロキシ-7-メトキシキノリン-3-カルボキシレート 8.56 g (34.6 mmol) に2.0 M 水酸化ナトリウム水 85 mL (170 mmol) を加え、1.0時間加熱還流した。反応液を室温まで冷却し、2.0 M塩酸 80 mLを加え、析出物をろ取し、水にて洗浄後、乾燥した。得られた粉末にジフェニルエーテル 40 mLを加え、40分間加熱還流した。反応液を室温まで冷却し、上清をデカントにて除去した。残渣に5.0 M水酸化ナトリウム水を加え、酢酸エチルにて2回洗浄し、水層に5% クエン酸水を加え、pH 2に調整した。析出物をろ取後、ジイソプロピルエーテルにて洗浄し、標題化合物の白色粉末 1.30 g (収率:21%) を得た。
1H-NMR (DMSO-d6, 400 MHz) δ:3.85 (3H, s), 5.93 (1H, d, J=6.8 Hz), 6.82-6.97 (2H, m), 7.72-7.84 (1H, m), 7.94-8.05 (1H, m), 11.45-11.60 (1H, br).
To 12.3 g (0.100 mol) of 3-methoxyaniline, 21.6 g (0.100 mol) of diethyl ethoxymethylene malonate was added, and the mixture was stirred at 110 ° C. for 75 minutes, 150 mL of diphenyl ether was added, and the mixture was stirred at 230 ° C. for 8.5 hours. The reaction was cooled to room temperature and poured into 200 mL of ethyl acetate. The precipitate was collected by filtration and washed with ethyl acetate to obtain 8.56 g (yield: 35%) of a light brown powder of ethyl 4-hydroxy-7-methoxyquinoline-3-carboxylate.
To 8.56 g (34.6 mmol) of ethyl 4-hydroxy-7-methoxyquinoline-3-carboxylate, 85 mL (170 mmol) of 2.0 M sodium hydroxide water was added, and the mixture was heated under reflux for 1.0 hour. The reaction mixture was cooled to room temperature, 80 mL of 2.0 M hydrochloric acid was added, the precipitate was collected by filtration, washed with water, and dried. 40 mL of diphenyl ether was added to the obtained powder, and the mixture was heated under reflux for 40 minutes. The reaction solution was cooled to room temperature, and the supernatant was removed by decanting. 5.0 M sodium hydroxide water was added to the residue, washed twice with ethyl acetate, and 5% citric acid water was added to the aqueous layer to adjust the pH to 2. The precipitate was collected by filtration and washed with diisopropyl ether to give 1.30 g (yield: 21%) of a white powder of the title compound.
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 3.85 (3H, s), 5.93 (1H, d, J = 6.8 Hz), 6.82-6.97 (2H, m), 7.72-7.84 (1H, m) , 7.94-8.05 (1H, m), 11.45-11.60 (1H, br).

(20) 7-メトキシ-1-[4-(3-(モルホリン-4-イル)アゼチジン-1-イル)フェニル]-1H-キノリン-4-オン 塩酸塩 (20) 7-Methoxy-1- [4- (3- (morpholine-4-yl) azetidine-1-yl) phenyl] -1H-quinoline-4-one hydrochloride

Figure 2020071550
Figure 2020071550

実施例20 (20a) で製造した化合物 594 mg (3.39 mmol)、4-[1-(4-ヨードフェニル)アゼチジン-3-イル]モルホリン 1.17 g (3.39 mmol) をジメチルスルホキシド 8 mLに懸濁し、2,2,6,6-テトラメチル-3,5-ヘプタンジオン141 μL (0.678 mmol)、ヨウ化銅 65 mg (0.34 mmol)、リン酸三カリウム 1.44 g (6.78 mmol) を加え、窒素を1分間バブリングし、窒素雰囲気下、120℃ にて9時間撹拌後、さらに室温にて5時間撹拌した。反応液に水 100 mL、クロロホルム 100 mLを加え、不溶物をろ別後、二層を分離し、水層をクロロホルムにて3回抽出した。有機層を合わせ、飽和食塩水にて洗浄後、乾燥 (Na2SO4) し、減圧下溶媒を留去した。得られた残渣をカラムクロマトグラフィー(クロロホルム:メタノール、98:2→95:5、V/V)にて精製後、目的分画の溶媒を減圧下留去し、7-メトキシ-1-[4-(3-(モルホリン-4-イル)アゼチジン-1-イル)フェニル]-1H-キノリン-4-オンの淡褐色粉末 975mg (収率:73%) を得た。
7-メトキシ-1-[4-(3-(モルホリン-4-イル)アゼチジン-1-イル)フェニル]-1H-キノリン-4-オン 800 mg (2.04 mmol) をメタノール 10 mLに懸濁させ、氷冷下8.6 M 塩化水素 イソプロパノール溶液 712 μL (6.1 mmol) を加え、室温にて5分間撹拌した。反応液にジイソプロピルエーテル 80 mLを加え、析出物をろ取し、標題化合物の橙色粉末 933 mg (収率:99%) を得た。
IR ν (ATR) cm-1 :1608
1H-NMR (DMSO-d6, 400 MHz) δ:3.00-3.14 (2H, m), 3.33-3.48 (2H, m), 3.72 (3H, s), 3.80-3.96 (2H, m), 3.97-4.08 (2H, m), 4.18-4.24 (2H, m), 4.25-4.38 (3H, m), 6.44 (1H, d, J=2.2 Hz), 6.66-6.74 (3H, m), 7.24 (1H, dd, J=8.6, 2.2 Hz), 7.41-7.46 (2H, m), 8.25 (1H, d, J=8.6 Hz), 8.31 (1H, d, J=7.2 Hz).
MS m/z: 392 [M+H]+.
Compound 594 mg (3.39 mmol), 4- [1- (4-iodophenyl) azetidine-3-yl] morpholine 1.17 g (3.39 mmol) prepared in Example 20 (20a) was suspended in 8 mL of dimethyl sulfoxide. Add 141 μL (0.678 mmol) of 2,2,6,6-tetramethyl-3,5-heptandione, 65 mg (0.34 mmol) of copper iodide, 1.44 g (6.78 mmol) of tripotassium phosphate, and add 1 nitrogen. Bubbling was carried out for 1 minute, and the mixture was stirred at 120 ° C. for 9 hours under a nitrogen atmosphere, and then further stirred at room temperature for 5 hours. 100 mL of water and 100 mL of chloroform were added to the reaction mixture, the insoluble material was filtered off, the two layers were separated, and the aqueous layer was extracted 3 times with chloroform. The organic layers were combined, washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure. The obtained residue was purified by column chromatography (chloroform: methanol, 98: 2 → 95: 5, V / V), the solvent of the target fraction was distilled off under reduced pressure, and 7-methoxy-1- [4]. -(3- (Morpholine-4-yl) azetidine-1-yl) phenyl] -1H-quinoline-4-one light brown powder 975 mg (yield: 73%) was obtained.
7-Methone-1- [4- (3- (morpholine-4-yl) azetidine-1-yl) phenyl] -1H-quinoline-4-one 800 mg (2.04 mmol) was suspended in 10 mL of methanol. 8.6 M hydrogen chloride isopropanol solution under ice cooling 712 μL (6.1 mmol) was added, and the mixture was stirred at room temperature for 5 minutes. 80 mL of diisopropyl ether was added to the reaction mixture, and the precipitate was collected by filtration to give 933 mg (yield: 99%) of orange powder of the title compound.
IR ν (ATR) cm -1 : 1608
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 3.00-3.14 (2H, m), 3.33-3.48 (2H, m), 3.72 (3H, s), 3.80-3.96 (2H, m), 3.97- 4.08 (2H, m), 4.18-4.24 (2H, m), 4.25-4.38 (3H, m), 6.44 (1H, d, J = 2.2 Hz), 6.66-6.74 (3H, m), 7.24 (1H, m) dd, J = 8.6, 2.2 Hz), 7.41-7.46 (2H, m), 8.25 (1H, d, J = 8.6 Hz), 8.31 (1H, d, J = 7.2 Hz).
MS m / z: 392 [M + H] + .

実施例21 Example 21

(21a) メチル 8-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ナフタレン-2-カルボキシレート (21a) Methyl 8- {4- [2- (tetrahydropyran-4-yloxy) ethoxy] phenyl} naphthalene-2-carboxylate

Figure 2020071550
Figure 2020071550

メチル 8-ブロモナフタレン-2-カルボキシレート 495 mg (1.87 mmol) および実施例14 (14b) で製造した化合物 650 mg (1.87 mmol) をN,N-ジメチルホルムアミド 10 mLに溶解し、リン酸三カリウム 793 mg (3.74 mmol) およびテトラキス(トリフェニルホスフィン)パラジウム(0) 216 mg (0.187 mmol) を加え、窒素雰囲気下90℃ にて7時間撹拌した。放冷後反応液に水および酢酸エチルを加え、不溶物をセライトろ過により除去後、ろ液の二層を分離し、有機層を飽和食塩水にて洗浄後、乾燥 (Na2SO4) した。減圧下溶媒を留去し、残渣をカラムクロマトグラフィー(n-ヘキサン:酢酸エチル、3:1、V/V)にて精製後、目的分画の溶媒を減圧下留去し、標題化合物の黄色油状物 565 mg (収率:74%) を得た。
1H-NMR (CDCl3, 400 MHz) δ:1.63-1.73 (2H, m), 1.94-2.02 (2H, m), 3.44-3.52 (2H, m), 3.61-3.68 (1H, m), 3.88-3.93 (5H, m), 3.99 (2H, dt, J=12.0, 4.2 Hz), 4.21-4.25 (2H, m), 7.05-7.09 (2H, m), 7.39-7.43 (2H, m), 7.46 (1H, dd, J=7.1, 1.0 Hz), 7.58-7.63 (1H, m), 7.30 (1H, d, J=8.0 Hz), 7.93 (1H, d, J=8.6 Hz), 8.06 (1H, dd, J=8.6, 1.5 Hz), 8.66-8.68 (1H, m).
Methyl 8-bromonaphthalene-2-carboxylate 495 mg (1.87 mmol) and compound 650 mg (1.87 mmol) prepared in Example 14 (14b) were dissolved in 10 mL of N, N-dimethylformamide and tripotassium phosphate. 793 mg (3.74 mmol) and tetrakis (triphenylphosphine) palladium (0) 216 mg (0.187 mmol) were added, and the mixture was stirred at 90 ° C. for 7 hours under a nitrogen atmosphere. After allowing to cool, water and ethyl acetate were added to the reaction solution, the insoluble material was removed by filtration through Celite, the two layers of the filtrate were separated, the organic layer was washed with saturated brine, and dried (Na 2 SO 4 ). .. The solvent was distilled off under reduced pressure, the residue was purified by column chromatography (n-hexane: ethyl acetate, 3: 1, V / V), and the solvent of the target fraction was distilled off under reduced pressure. An oily substance of 565 mg (yield: 74%) was obtained.
1 1 H-NMR (CDCl 3 , 400 MHz) δ: 1.63-1.73 (2H, m), 1.94-2.02 (2H, m), 3.44-3.52 (2H, m), 3.61-3.68 (1H, m), 3.88 -3.93 (5H, m), 3.99 (2H, dt, J = 12.0, 4.2 Hz), 4.21-4.25 (2H, m), 7.05-7.09 (2H, m), 7.39-7.43 (2H, m), 7.46 (1H, dd, J = 7.1, 1.0 Hz), 7.58-7.63 (1H, m), 7.30 (1H, d, J = 8.0 Hz), 7.93 (1H, d, J = 8.6 Hz), 8.06 (1H, 1H, dd, J = 8.6, 1.5 Hz), 8.66-8.68 (1H, m).

(21) 8-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ナフタレン-2-カルボキサミド (21) 8- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] phenyl} naphthalene-2-carboxamide

Figure 2020071550
Figure 2020071550

実施例21 (21a) で製造した化合物 350 mg (0.861 mmol) をテトラヒドロフラン 6 mL に溶解し、メタノール 3 mLおよび1.0 M 水酸化リチウム水 2.2 mL (2.2 mmol) を加え、室温にて9時間撹拌した。減圧下メタノールおよびテトラヒドロフランを留去し、水 10 mLおよび6.0 M塩酸0.40 mL (2.4 mmol) を加え、酢酸エチルにて抽出した。有機層を飽和食塩水にて洗浄し、乾燥 (Na2SO4) 後、減圧下溶媒を留去し、白色粉末 330 mgを得た。
得られた白色粉末330 mgをN,N-ジメチルホルムアミド 5 mLに溶解後、炭酸アンモニウム 500 mg、1-ヒドロキシベンゾトリアゾール 125 mg (0.925 mmol) および1-[3-(ジメチルアミノ)プロピル]-3-エチルカルボジイミド 塩酸塩 177 mg (0.923 mmol) を加え、室温にて16時間撹拌した。反応液に水を加え、酢酸エチルにて2回抽出後、有機層を合わせ、飽和食塩水にて洗浄し、乾燥 (Na2SO4) した。減圧下溶媒を留去し、残渣をカラムクロマトグラフィー(クロロホルム:メタノール、1:0→9:1、V/V)にて精製後、目的分画の溶媒を減圧下留去した。得られた粉末 260 mgを酢酸エチル 16 mLに加熱溶解し、室温に戻しながら3時間撹拌した。析出物をろ取し、標題化合物の白色粉末 221 mg (収率:67%) を得た。
IR ν (ATR) cm-1 :3406, 3195, 1668
1H-NMR (DMSO-d6, 400 MHz) δ:1.38-1.48 (2H, m), 1.85-1.92 (2H, m), 3.31-3.38 (2H, m), 3.55-3.63 (1H, m), 3.78-3.85 (4H, m), 4.16-4.20 (2H, m), 7.09-7.13 (2H, m), 7.38-7.46 (4H, m), 7.63 (1H, dd, J=8.3, 7.1 Hz), 7.93-7.98 (2H, m), 8.01-8.08 (2H, m), 8.38-8.40 (1H, m).
MS m/z: 414 [M+H]+, 390 [M-H]-.
350 mg (0.861 mmol) of the compound prepared in Example 21 (21a) was dissolved in 6 mL of tetrahydrofuran, 3 mL of methanol and 2.2 mL (2.2 mmol) of 1.0 M lithium hydroxide water were added, and the mixture was stirred at room temperature for 9 hours. .. Methanol and tetrahydrofuran were distilled off under reduced pressure, 10 mL of water and 0.40 mL (2.4 mmol) of 6.0 M hydrochloric acid were added, and the mixture was extracted with ethyl acetate. The organic layer was washed with saturated brine, dried (Na 2 SO 4 ), and the solvent was evaporated under reduced pressure to give 330 mg of white powder.
After dissolving 330 mg of the obtained white powder in 5 mL of N, N-dimethylformamide, 500 mg of ammonium carbonate, 125 mg (0.925 mmol) of 1-hydroxybenzotriazole and 1- [3- (dimethylamino) propyl] -3 -Ethylcarbodiimide hydrochloride 177 mg (0.923 mmol) was added, and the mixture was stirred at room temperature for 16 hours. Water was added to the reaction mixture, and the mixture was extracted twice with ethyl acetate, the organic layers were combined, washed with saturated brine, and dried (Na 2 SO 4 ). The solvent was evaporated under reduced pressure, the residue was purified by column chromatography (chloroform: methanol, 1: 0 → 9: 1, V / V), and the solvent of the target fraction was evaporated under reduced pressure. The obtained powder (260 mg) was dissolved in 16 mL of ethyl acetate by heating, and the mixture was stirred for 3 hours while returning to room temperature. The precipitate was collected by filtration to give 221 mg (yield: 67%) of a white powder of the title compound.
IR ν (ATR) cm -1 : 3406, 3195, 1668
1 1 H-NMR (DMSO-d6, 400 MHz) δ: 1.38-1.48 (2H, m), 1.85-1.92 (2H, m), 3.31-3.38 (2H, m), 3.55-3.63 (1H, m), 3.78-3.85 (4H, m), 4.16-4.20 (2H, m), 7.09-7.13 (2H, m), 7.38-7.46 (4H, m), 7.63 (1H, dd, J = 8.3, 7.1 Hz), 7.93-7.98 (2H, m), 8.01-8.08 (2H, m), 8.38-8.40 (1H, m).
MS m / z: 414 [M + H] + , 390 [MH] - .

以下の表1−1〜表1−71に示す化合物は、実施例1〜21に示す方法に準じて製造した。 The compounds shown in Tables 1-1 to 1-71 below were produced according to the methods shown in Examples 1 to 21.

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

Figure 2020071550
Figure 2020071550

(試験例1)
以下の方法により、実施例化合物のCDK8阻害活性を評価した。
(Test Example 1)
The CDK8 inhibitory activity of the Example compound was evaluated by the following method.

DMSOに溶解した被験化合物をアッセイバッファー(QSS Assist STK ELISA Kit(CDK8/CycC)、Carna Bioscience)で希釈し、DMSO濃度4%の一次希釈溶液を得た。上記一次希釈溶液10μLをストレプトアビジンコーティング96穴プレートに分注した後、1μM基質(Carna Bioscience)、20mM MgCl(Carna Bioscience)及び400μM ATP(Carna Bioscience)を含有したアッセイバッファー混液10μLを添加した。上記添加後、さらにキナーゼ溶液(CDK8阻害活性測定用にはアッセイバッファーで希釈した0.15ng/μL CDK8/CycC(Carna Bioscience)を用いた)を20μL添加した。上記添加後、20〜30℃で30分静置した。反応終了後、ウェル内の溶液を捨て、直ちにウェルあたり250μLのウォッシュバッファー(50mM Tris−HCl(pH7.5)、150mM NaCl、0.02%Tween−20)で4回洗浄した。各ウェルにブロッキングバッファー(0.1%BSA含有アッセイバッファー)を200μL添加し、室温にて30分間静置した。ウェル内の溶液を捨て、各ウェルに一次抗体溶液(Carna Bioscience)を100μL添加し、室温にて30分間静置した。ウェル内の溶液を捨て、直ちにウェルあたり250μLのウォッシュバッファーで4回洗浄した。各ウェルにHRP標識二次抗体溶液(Carna Bioscience)を100μL添加し、室温にて30分間静置した。ウェル内の溶液を捨て、直ちにウェルあたり250μLのウォッシュバッファーで4回洗浄した。各ウェルに発色試薬(ELISA POD基質TMBキット(HYPER)、ナカライテスク)を100μL添加し、室温にて5分反応させた。各ウェルに呈色反応停止薬(ナカライテスク)を100μL添加して反応を止めた後、プレートリーダーで吸光度(450nm)を測定した。The test compound dissolved in DMSO was diluted with assay buffer (QSS Assist STK ELISA Kit (CDK8 / CycC), Carna Bioscience) to obtain a primary dilution solution having a DMSO concentration of 4%. After dispensing 10 μL of the above primary dilution solution into a streptavidin-coated 96-well plate, 10 μL of an assay buffer mixture containing 1 μM substrate (Carna Biosciences), 20 mM MgCl 2 (Carna Biosciences) and 400 μM ATP (Carna Biosciences) was added. After the above addition, 20 μL of a kinase solution (0.15 ng / μL CDK8 / CycC (Carna Bioscience) diluted with an assay buffer was used for measuring the CDK8 inhibitory activity) was further added. After the above addition, the mixture was allowed to stand at 20 to 30 ° C. for 30 minutes. After completion of the reaction, the solution in the wells was discarded and immediately washed 4 times with 250 μL of wash buffer (50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 0.02% Tween-20) per well. 200 μL of blocking buffer (assay buffer containing 0.1% BSA) was added to each well, and the mixture was allowed to stand at room temperature for 30 minutes. The solution in the wells was discarded, 100 μL of the primary antibody solution (Carna Bioscience) was added to each well, and the mixture was allowed to stand at room temperature for 30 minutes. The solution in the wells was discarded and immediately washed 4 times with 250 μL wash buffer per well. 100 μL of HRP-labeled secondary antibody solution (Carna Biosciences) was added to each well, and the mixture was allowed to stand at room temperature for 30 minutes. The solution in the wells was discarded and immediately washed 4 times with 250 μL wash buffer per well. 100 μL of a coloring reagent (ELISA POD substrate TMB kit (HYPER), Nacalai Tesque) was added to each well, and the mixture was reacted at room temperature for 5 minutes. After stopping the reaction by adding 100 μL of a color reaction terminator (Nacalai Tesque) to each well, the absorbance (450 nm) was measured with a plate reader.

被験化合物非添加のウェルの吸光度をコントロール、酵素非添加のウェルの吸光度をブランクとして、被験化合物1μMにおけるCDK8阻害率を求めた。 The absorbance of the well without the test compound was controlled, and the absorbance of the well without the enzyme was used as a blank to determine the CDK8 inhibition rate at 1 μM of the test compound.

化合物1、6、8、10、14、19、20、21、28、31、38、48、57、63、78、112、117、120、149、163、172及び176は、1μMにおいて90%以上のCDK8阻害率を示した。 Compounds 1, 6, 8, 10, 14, 19, 20, 21, 28, 31, 38, 48, 57, 63, 78, 112, 117, 120, 149, 163, 172 and 176 are 90% at 1 μM. The above CDK8 inhibition rate was shown.

(試験例2)
以下の方法により、実施例化合物のヒト急性骨髄性白血病MV4;11 細胞増殖阻害活性を評価した。
(Test Example 2)
The human acute myeloid leukemia MV4; 11 cell proliferation inhibitory activity of the Example compound was evaluated by the following method.

ヒト急性骨髄性白血病 MV4;11(American Type Culture Collectionより購入)の細胞懸濁液100μL (10,000cells/ウェル)を96ウェルプレートに播き、5%炭酸ガスインキュベーター中、37℃で1日培養した。上記培養後、被験化合物濃度が1μMになるように添加し、さらに3日間培養した。上記培養後、40μLのMTS/PMS溶液(CellTiter 96(登録商標)AQueous One Solution Cell Proliferation Assay(Promega社))を96ウェルプレートに添加し、1時間反応させた後、プレートリーダーで吸光度(490nm)を測定した。 Human acute myeloid leukemia MV4; 11 (purchased from American Type Culture Collection) cell suspension 100 μL (10,000 cells / well) was seeded in 96-well plates and cultured in a 5% carbon dioxide incubator at 37 ° C. for 1 day. .. After the above culture, the test compound was added so as to have a concentration of 1 μM, and the cells were further cultured for 3 days. After the above culture, 40 μL of MTS / PMS solution (CellTiter 96 (registered trademark) AQueuus One Solution Cell Proliferation Assay (Promega)) was added to a 96-well plate, reacted for 1 hour, and then absorbed by a plate reader (490 nm). Was measured.

被験化合物非添加のウェルの吸光度をコントロール、細胞を播種していないウェルの吸光度をブランクとして、被験化合物 1μM における細胞増殖阻害率を求めた。 The absorbance of the wells to which the test compound was not added was controlled, and the absorbance of the wells in which cells were not seeded was used as a blank, and the cell proliferation inhibition rate at 1 μM of the test compound was determined.

化合物1、6、8、10、14、19、20、21、28、31、38、48、57、63、78、112、117、120、149、163、172及び176は、1μMにおいて50%以上の細胞増殖阻害率を示した。 Compounds 1, 6, 8, 10, 14, 19, 20, 21, 28, 31, 38, 48, 57, 63, 78, 112, 117, 120, 149, 163, 172 and 176 are 50% at 1 μM. The above cell proliferation inhibition rate was shown.

(試験例3)
以下の方法により、実施例化合物の骨芽細胞分化作用を評価した。
(Test Example 3)
The osteoblast differentiation effect of the Example compound was evaluated by the following method.

マウス骨髄由来間葉系細胞株ST2細胞(入手先:理化学研究所)を96 well plateにおいて、10%牛胎児血清を含むα-MEM培地(入手先:GIBCO BRL Cat.No.12000-022)中、4×103cells/0.1 mL/wellの細胞密度で播種し、37℃、5%CO2条件下で24h培養した。次いで、被験化合物を終濃度0.001〜10 μMになるように添加し、コントロールには最終濃度0.1%(v/v)のDMSOを添加した。さらに4日間培養後、アルカリホスファターゼ(ALP)活性を以下の方法で測定を行った。
培養した96 well plateの培地を除去した後、PBSバッファー(KCl 0.2 g/L,KH2PO4 0.2 g/L,Na2HPO4・12H2O 2.9 g/L,NaCl 8 g/L)100 μL/well で洗浄し、細胞溶解液 (10 mM MgCl2、2%(v/v)TritonX-100) 50 μL/wellを添加し、室温で3分撹拌した。基質溶液(50 mMジエタノールアミン(和光純薬 Cat.No.099-03112)、20 mM p-ニトロフェニルホスファイト(ナカライテスクCat.No.25019-81)) 50 μL/wellを添加し、室温で9分間静置後、1 N NaOH 50 μL/wellを加えて反応を停止し、マイクロプレートリーダー(大日本製薬社)を用いて405nmの吸光度を測定した。
Mouse bone marrow-derived mesenchymal cell line ST2 cells (obtained by: Institute of Physical and Chemical Research) in 96-well plate in α-MEM medium containing 10% fetal bovine serum (obtained from GIBCO BRL Cat.No.12000-022) , 4 × 10 3 cells / 0.1 mL / well, seeded and cultured for 24 hours under 37 ° C. and 5% CO 2 conditions. The test compound was then added to a final concentration of 0.001-10 μM, and DMSO with a final concentration of 0.1% (v / v) was added to the control. After culturing for another 4 days, the alkaline phosphatase (ALP) activity was measured by the following method.
After removal of the medium of 96 well plate cultured, PBS buffer (KCl 0.2 g / L, KH 2 PO 4 0.2 g / L, Na 2 HPO 4 · 12H 2 O 2.9 g / L, NaCl 8 g / L) 100 The cells were washed with μL / well, 50 μL / well of cell lysate (10 mM MgCl 2 , 2% (v / v) TritonX-100) was added, and the mixture was stirred at room temperature for 3 minutes. Substrate solution (50 mM diethanolamine (Wako Pure Chemicals Cat.No.099-03112), 20 mM p-nitrophenylphosphite (Nacalai Tesque Cat.No.25019-81)) 50 μL / well was added, and 9 at room temperature. After allowing to stand for 1 minute, 50 μL / well of 1 N NaOH was added to stop the reaction, and the absorbance at 405 nm was measured using a microplate reader (Dainippon Pharmaceutical Co., Ltd.).

コントロールの吸光度を100%とした際の被験化合物の吸光度の増加率(%)を算出し、骨芽細胞の分化度を評価した。 The rate of increase (%) in the absorbance of the test compound when the absorbance of the control was set to 100% was calculated, and the degree of differentiation of osteoblasts was evaluated.

本試験において、全ての実施例化合物が、0.1μg/mLで、200%以上のアルカリホスファターゼ活性を示した。 In this test, all Example compounds showed more than 200% alkaline phosphatase activity at 0.1 μg / mL.

(試験例4)
以下の方法により、実施例化合物の骨密度への影響を評価した。
(Test Example 4)
The effect of the Example compound on bone mineral density was evaluated by the following method.

12週齢雌性F344ラット(SLC)は麻酔下で卵巣摘出または偽手術を施した。手術翌々日より試験化合物を0.5%メチルセルロース溶液 (和光純薬Cat.No.133-14255) に懸濁し、一日一回、週6あるいは7日経口投与した。なお、対照群には0.5%メチルセルロース溶液を経口投与した。投与8週後、麻酔下で腹部大動脈より全採血して安楽死させ、左右大腿骨を摘出した。
摘出した大腿骨は、軟部組織を除去した後、DXA装置DCS-600R(アロカ株式会社)を用いて骨密度を測定した。骨密度は、大腿骨全体、並びに、全体を三等分して近位端、骨幹部及び遠位端部分に分けて評価した。
12-week-old female F344 rats (SLC) underwent ovariectomy or sham surgery under anesthesia. From the day after the operation, the test compound was suspended in a 0.5% methylcellulose solution (Wako Pure Chemical Industries, Ltd. Cat.No.133-14255) and orally administered once a day for 6 or 7 days a week. A 0.5% methylcellulose solution was orally administered to the control group. Eight weeks after administration, all blood was collected from the abdominal aorta under anesthesia and euthanized, and the left and right femurs were removed.
After removing the soft tissue from the removed femur, the bone density was measured using a DXA device DCS-600R (Aroca Co., Ltd.). Bone density was evaluated by dividing the entire femur and the entire femur into three equal parts, the proximal end, the diaphysis, and the distal end.

本試験において、化合物2、3、36、38、40、54、92、102、120及び167は、10 mg/kg以下で有意に骨密度を増加させた。 In this study, compounds 2, 3, 36, 38, 40, 54, 92, 102, 120 and 167 significantly increased bone mineral density below 10 mg / kg.

以上の結果より、本発明の化合物(I)は、サイクリン依存性キナーゼ(CDK)8に対する優れた阻害活性を有すること、ヒト急性骨髄性白血病 MV4;11細胞に対し、高い細胞増殖阻害活性を有すること、優れた骨芽細胞分化作用及び骨密度増加作用を有することが分かった。 From the above results, the compound (I) of the present invention has an excellent inhibitory activity on cyclin-dependent kinase (CDK) 8 and a high cell proliferation inhibitory activity on human acute myeloid leukemia MV4; 11 cells. It was found that it has an excellent osteoblast differentiation effect and a bone density increasing effect.

製剤例1(カプセルの製造)
1)実施例化合物(化合物(I)) 10 mg
2)微粉末セルロース 10 mg
3)乳糖 19 mg
4)ステアリン酸マグネシウム 1 mg
計 40 mg
1)、2)、3)及び4)を混合して、ゼラチンカプセルに充填する。
Pharmaceutical example 1 (manufacturing of capsule)
1) Example compound (Compound (I)) 10 mg
2) Fine powdered cellulose 10 mg
3) Lactose 19 mg
4) Magnesium stearate 1 mg
40 mg in total
1), 2), 3) and 4) are mixed and filled in gelatin capsules.

製剤例2(錠剤の製造)
1)実施例化合物(化合物(I)) 10 g
2)乳糖 50 g
3)トウモロコシデンプン 15 g
4)カルボキシメチルセルロースカルシウム 44 g
5)ステアリン酸マグネシウム 1 g
1000錠 計 120 g
1)、2)、3)の全量及び30gの4)を水で練合し、真空乾燥後、整粒を行う。この整粒末に14gの4)及び1gの5)を混合し、打錠機により打錠する。このようにして、1錠あたり実施例化合物10mgを含有する錠剤1000錠を得る。
Pharmaceutical example 2 (Manufacturing of tablets)
1) Example compound (Compound (I)) 10 g
2) Lactose 50 g
3) Corn starch 15 g
4) Carboxymethyl cellulose calcium 44 g
5) Magnesium stearate 1 g
1000 tablets total 120 g
The whole amount of 1), 2) and 3) and 30 g of 4) are kneaded with water, vacuum dried, and then granulated. 14 g of 4) and 1 g of 5) are mixed with this sized powder and locked with a lock press. In this way, 1000 tablets containing 10 mg of the Example compound per tablet are obtained.

本発明の化合物(I)は、サイクリン依存性キナーゼ(CDK)8に対する優れた阻害活性を有する。したがって、本発明によれば、優れたCDK8阻害剤を提供することができる。また、本発明によれば、CDK8に関連する疾患、骨粗鬆症等の骨代謝に関連する疾患、癌等の細胞増殖性疾患の予防及び/または治療剤として有用な医薬も提供することができる。
本出願は、日本で出願された特願2018−189561および特願2018−206050を基礎としており、それらの内容は本明細書にすべて包含されるものである。
Compound (I) of the present invention has excellent inhibitory activity against cyclin-dependent kinase (CDK) 8. Therefore, according to the present invention, it is possible to provide an excellent CDK8 inhibitor. Further, according to the present invention, it is possible to provide a medicine useful as a preventive and / or therapeutic agent for a disease related to CDK8, a disease related to bone metabolism such as osteoporosis, and a cell proliferation disease such as cancer.
This application is based on Japanese Patent Application No. 2018-189561 and Japanese Patent Application No. 2018-206050, the contents of which are incorporated herein by reference.

Claims (27)

一般式(I):
Figure 2020071550

[式中、各置換基は以下のように定義される。
A環:
Figure 2020071550

(R
1−6アルキルカルボニル基、C1−6アルキルチオカルボニル基、カルバモイル基、C1−6アルコキシカルボニル基またはカルボキシ基、

水素原子、C1−6アルキル基またはハロゲン原子、

1−6アルキル基、ハロゲン原子、アミノ基、ヒドロキシC1−6アルキル基、カルボキシC1−6アルキル基または水素原子(但し、Tが=CH−、Uが=N−の場合)、

水素原子またはC1−6アルキル基、

水素原子、C1−6アルキル基またはC1−6アルキルカルボニル基、

水素原子またはC1−6アルキル基)、

水素原子、ハロゲン原子、シアノ基、C1−6アルキル基、C1−6アルコキシ基、C1−6アルキルスルファニル基、C1−6アルキルカルボニル基、カルバモイル基または複素環基、
T及びU:
T及びUのいずれか一方が、=N−である場合、もう一方は=CR−、
T及びUの両方が、=CR−、

水素原子、ハロゲン原子、C1−6アルキル基またはC1−6アルコキシ基、

−B−D−E−G
(B:
6−14アリーレン基、C3−8シクロアルケニレン基、−複素環−または−飽和複素環−、
D:
単結合、−O(CH−、−(CHO−または−飽和複素環−、
n:
1〜4から選択されるいずれか1の整数、
E:
単結合、−O−、−N(R10)−、−CO−または−CONH−、
10
水素原子、置換基群αから選択されるいずれかの基で置換されていてもよいC1−6アルキル基、
G:
水素原子、置換基群αから選択されるいずれかの基で置換されていてもよいC1−6アルキル基、置換基群αから選択されるいずれかの基で置換されていてもよいC3−8シクロアルキル基、置換基群αから選択されるいずれかの基で置換されていてもよいC6−14アリール基、置換基群αから選択されるいずれかの基で置換されていてもよい複素環基または置換基群αから選択されるいずれかの基で置換されていてもよい飽和複素環基、
置換基群α:
複素環基、飽和複素環基、ハロゲン原子、シアノ基、C1−6アルキルカルボニル基、C1−6アルキルカルボニルオキシ基、C1−6アルキルカルボニルアミノ基、C1−6アルコキシカルボニル基、C1−6アルコキシC1−6アルキル基、ヒドロキシC1−6アルキル基、ヒドロキシC1−6アルコキシ基、C1−6アルキルスルホニル基、C1−6アルキルスルホニルアミノ基、アミノスルホニルアミノ基、カルボキシ基、カルボキシC1−6アルキルカルボニルオキシ基、カルバモイル基、ヒドロキシ基、C1−6アルコキシ基、C1−6アルキル基、C3−8シクロアルキル基、アミノ基、アミノC1−6アルキルカルボニルオキシ基、モノまたはジ−C1−6アルキルアミノ基、4−ヒドロキシテトラヒドロフラン−3−イルオキシ基、モノまたはジ−C1−6アルキルホスホノ基、
Figure 2020071550

(nは、1〜4から選択されるいずれか1の整数))]で表わされる化合物またはその医薬上許容される塩。
General formula (I):
Figure 2020071550

[In the formula, each substituent is defined as follows.
Ring A:
Figure 2020071550

(R 4 :
C 1-6 alkylcarbonyl group, C 1-6 alkylthiocarbonyl group, carbamoyl group, C 1-6 alkoxycarbonyl group or carboxy group,
R 5 :
Hydrogen atom, C 1-6 alkyl group or halogen atom,
R 6 :
C 1-6 alkyl group, halogen atom, amino group, hydroxy C 1-6 alkyl group, carboxy C 1-6 alkyl group or hydrogen atom (provided that T is = CH- and U is = N-),
R 7 :
Hydrogen atom or C 1-6 alkyl group,
R 8 :
Hydrogen atom, C 1-6 alkyl group or C 1-6 alkyl carbonyl group,
R 9 :
Hydrogen atom or C 1-6 alkyl group),
R 1 :
Hydrogen atom, halogen atom, cyano group, C 1-6 alkyl group, C 1-6 alkoxy group, C 1-6 alkyl sulfanyl group, C 1-6 alkyl carbonyl group, carbamoyl group or heterocyclic group,
T and U:
If either T or U is = N-, the other is = CR 2- ,
Both T and U = CR 2- ,
R 2 :
Hydrogen atom, halogen atom, C 1-6 alkyl group or C 1-6 alkoxy group,
R 3 :
−BDEG
(B:
C 6-14 arylene group, C 3-8 cycloalkenylene group, -heterocycle-or-saturated heterocycle-,
D:
Single bond, -O (CH 2 ) n -,-(CH 2 ) n O- or -saturated heterocycle-,
n:
An integer of any one selected from 1 to 4,
E:
Single bond, -O-, -N (R 10 )-, -CO- or -CONH-,
R 10 :
A hydrogen atom, a C 1-6 alkyl group which may be substituted with any group selected from the substituent group α,
G:
Hydrogen atom, C 1-6 alkyl group may be substituted with any group selected from the substituent group α, C 3 which may be substituted with any group selected from the substituent group α -8 Cycloalkyl group, C 6-14 aryl group which may be substituted with any group selected from the substituent group α, or any group selected from the substituent group α Saturated heterocyclic groups, which may be substituted with a good heterocyclic group or any group selected from the substituent group α,
Substituent group α:
Heterocyclic group, saturated heterocyclic group, halogen atom, cyano group, C 1-6 alkylcarbonyl group, C 1-6 alkylcarbonyloxy group, C 1-6 alkylcarbonylamino group, C 1-6 alkoxycarbonyl group, C 1-6 alkoxy C 1-6 alkyl group, hydroxy C 1-6 alkyl group, hydroxy C 1-6 alkoxy group, C 1-6 alkylsulfonyl group, C 1-6 alkylsulfonylamino group, aminosulfonylamino group, carboxy Group, carboxy C 1-6 alkylcarbonyloxy group, carbamoyl group, hydroxy group, C 1-6 alkoxy group, C 1-6 alkyl group, C 3-8 cycloalkyl group, amino group, amino C 1-6 alkylcarbonyl Oxy group, mono or di-C 1-6 alkylamino group, 4-hydroxy tetrahydrofuran-3-yloxy group, mono or di-C 1-6 alkylphosphono group,
Figure 2020071550

(N is an integer of any one selected from 1 to 4))] or a pharmaceutically acceptable salt thereof.
A環が、
Figure 2020071550

であり、Rが、アセチル基、カルバモイル基またはカルボキシ基であり、Rが、メチル基である、請求項1に記載の化合物またはその医薬上許容される塩。
Ring A is
Figure 2020071550

The compound according to claim 1, wherein R 4 is an acetyl group, a carbamoyl group or a carboxy group, and R 5 is a methyl group, or a pharmaceutically acceptable salt thereof.
A環が、
Figure 2020071550

であり、Rが、メチル基であり、Rが、水素原子である、請求項1に記載の化合物またはその医薬上許容される塩。
Ring A is
Figure 2020071550

The compound according to claim 1, wherein R 7 is a methyl group and R 9 is a hydrogen atom, or a pharmaceutically acceptable salt thereof.
A環が、
Figure 2020071550

であり、Rが、メチル基である、請求項1に記載の化合物またはその医薬上許容される塩。
Ring A is
Figure 2020071550

The compound according to claim 1 or a pharmaceutically acceptable salt thereof, wherein R 7 is a methyl group.
A環が、
Figure 2020071550

である、請求項1に記載の化合物またはその医薬上許容される塩。
Ring A is
Figure 2020071550

The compound according to claim 1 or a pharmaceutically acceptable salt thereof.
A環が、
Figure 2020071550

である、請求項1に記載の化合物またはその医薬上許容される塩。
Ring A is
Figure 2020071550

The compound according to claim 1 or a pharmaceutically acceptable salt thereof.
A環が、
Figure 2020071550

であり、Rが、塩素、メチル基またはカルボキシメチル基である、請求項1に記載の化合物またはその医薬上許容される塩。
Ring A is
Figure 2020071550

In and, R 6 is chlorine, a methyl group or a carboxymethyl group, a compound or a pharmaceutically acceptable salt thereof according to claim 1.
が、シアノ基、メトキシ基、メチルスルファニル基、アセチル基、カルバモイル基またはイミダゾリル基である、請求項1〜7のいずれか一項に記載の化合物またはその医薬上許容される塩。The compound according to any one of claims 1 to 7, wherein R 1 is a cyano group, a methoxy group, a methyl sulfanyl group, an acetyl group, a carbamoyl group or an imidazolyl group, or a pharmaceutically acceptable salt thereof. Tが、=CH−であり、Uが、=N−である、請求項1〜8のいずれか一項に記載の化合物またはその医薬上許容される塩。 The compound according to any one of claims 1 to 8, wherein T is = CH- and U is = N-, or a pharmaceutically acceptable salt thereof. Tが、=CH−であり、Uが、=CR−であり、Rが、水素原子、フッ素原子またはメトキシ基である、請求項1〜8のいずれか一項に記載の化合物またはその医薬上許容される塩。The compound according to any one of claims 1 to 8, wherein T is = CH-, U is = CR 2- , and R 2 is a hydrogen atom, a fluorine atom or a methoxy group. A pharmaceutically acceptable salt. Bが、
Figure 2020071550

である、請求項1〜10のいずれか一項に記載の化合物またはその医薬上許容される塩。
B is
Figure 2020071550

The compound according to any one of claims 1 to 10 or a pharmaceutically acceptable salt thereof.
Bが、
Figure 2020071550

である、請求項1〜10のいずれか一項に記載の化合物またはその医薬上許容される塩。
B is
Figure 2020071550

The compound according to any one of claims 1 to 10 or a pharmaceutically acceptable salt thereof.
Dが、単結合、−O(CH−または−飽和複素環−であり、nが2である、請求項1〜12のいずれか一項に記載の化合物またはその医薬上許容される塩。The compound according to any one of claims 1 to 12, wherein D is a single bond, −O (CH 2 ) n − or − saturated heterocycle −, and n is 2, or a pharmaceutically acceptable compound thereof. salt. Eが、単結合または−O−である、請求項1〜13のいずれか一項に記載の化合物またはその医薬上許容される塩。 The compound according to any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, wherein E is a single bond or -O-. Gが、ヒドロキシ基、アミノ基、カルボキシ基、アミノスルホニルアミノ基、ジエチルホスホノ基または
Figure 2020071550

で置換されているC1−6アルキル基、フェニル基、テトラヒドロピラニル基、テトヒドロフラニル基、ピペラジニル基またはモルホリニル基である、請求項1〜14のいずれか一項に記載の化合物またはその医薬上許容される塩。
G is a hydroxy group, an amino group, a carboxy group, an aminosulfonylamino group, a diethylphosphono group or
Figure 2020071550

The compound according to any one of claims 1 to 14, which is a C 1-6 alkyl group, a phenyl group, a tetrahydropyranyl group, a tethydrofuranyl group, a piperazinyl group or a morpholinyl group substituted with. A pharmaceutically acceptable salt.
以下に記載の化合物群から選択されるいずれかの化合物またはその医薬上許容される塩。
(1)3-アセチル-5,6-ジメトキシ-2-メチル-1-[4-(モルホリン-4-イル)フェニル]インドール
Figure 2020071550

(2)3-アセチル-5,6-ジメトキシ-2-メチル-1-{4-[3-(2-オキソ-2λ5-[1,3,2]ジオキサホスフィナン-2-イル)プロポキシ]フェニル}インドール
Figure 2020071550

(3)3-アセチル-1-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-6-メトキシ-2-メチルインドール
Figure 2020071550

(4)3-アセチル-1-{4-[4-(3-アミノプロピル)ピペリジン-1-イル]フェニル}-5,6-ジメトキシ-2-メチルインドール
Figure 2020071550

(5)3-アセチル-6-メトキシ-2-メチル-1-[4-(4-(モルホリン-4-イル)ブトキシ)フェニル]インドール 塩酸塩
Figure 2020071550

(6)3-アセチル-5-フルオロ-6-メトキシ-2-メチル-1-(1-フェニルピペリジン-4-イル)インドール
Figure 2020071550

(7)3-{1-[4-(3-アセチル-5,6-ジメトキシ-2-メチルインドール-1-イル)フェニル]ピペリジン-4-イル}プロピオン酸
Figure 2020071550

(8)1-[4-(4-アセチルピペラジン-1-イル)フェニル]-5,6-ジメトキシ-2-メチルインドール-3-カルボキサミド
Figure 2020071550

(9)1-{4-[4-(2-ヒドロキシエトキシ)ピペリジン-1-イル]フェニル}-6-メトキシ-2-メチル-1H-ピロロ[3,2-c]ピリジン-3-カルボキサミド
Figure 2020071550
Any compound selected from the group of compounds described below or a pharmaceutically acceptable salt thereof.
(1) 3-Acetyl-5,6-dimethoxy-2-methyl-1- [4- (morpholine-4-yl) phenyl] indole
Figure 2020071550

(2) 3-Acetyl-5,6-dimethoxy-2-methyl-1-{4- [3- (2-oxo-2λ 5- [1,3,2] dioxaphosphinan-2-yl) propoxy ] Phenyl} indole
Figure 2020071550

(3) 3-Acetyl-1-{4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -6-methoxy-2-methylindole
Figure 2020071550

(4) 3-Acetyl-1-{4- [4- (3-aminopropyl) piperidine-1-yl] phenyl} -5,6-dimethoxy-2-methylindole
Figure 2020071550

(5) 3-Acetyl-6-methoxy-2-methyl-1- [4- (4- (morpholine-4-yl) butoxy) phenyl] indole hydrochloride
Figure 2020071550

(6) 3-Acetyl-5-Fluoro-6-Methoxy-2-methyl-1- (1-phenylpiperidine-4-yl) indole
Figure 2020071550

(7) 3- {1- [4- (3-Acetyl-5,6-dimethoxy-2-methylindole-1-yl) phenyl] piperidin-4-yl} propionic acid
Figure 2020071550

(8) 1- [4- (4-Acetylpiperazin-1-yl) phenyl] -5,6-dimethoxy-2-methylindole-3-carboxamide
Figure 2020071550

(9) 1- {4- [4- (2-Hydroxyethoxy) piperidine-1-yl] phenyl} -6-methoxy-2-methyl-1H-pyrrolo [3,2-c] pyridine-3-carboxamide
Figure 2020071550
以下に記載の化合物群から選択されるいずれかの化合物またはその医薬上許容される塩。
(1)2-{1-[4-(6-メトキシ-2-メチルイミダゾ[4,5-c]ピリジン-1-イル)フェニル]アゼチジン-3-イルオキシ}エタノール
Figure 2020071550

(2)ジエチル(1-(4-(5-フルオロ-6-メトキシ-2-メチル-1H-ベンゾ[d]イミダゾール-1-イル)フェニル)アゼチジン-3-イルオキシ)メチルホスホネート
Figure 2020071550
Any compound selected from the group of compounds described below or a pharmaceutically acceptable salt thereof.
(1) 2- {1- [4- (6-Methoxy-2-methylimidazole [4,5-c] pyridin-1-yl) phenyl] azetidine-3-yloxy} ethanol
Figure 2020071550

(2) Diethyl (1- (4- (5-fluoro-6-methoxy-2-methyl-1H-benzo [d] imidazol-1-yl) phenyl) azetidine-3-yloxy) methylphosphonate
Figure 2020071550
以下に記載の化合物またはその医薬上許容される塩。
(1)4-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-6-メトキシ-1-メチル-1H-[1,7]ナフチリジン-2-オン
Figure 2020071550
The compounds listed below or pharmaceutically acceptable salts thereof.
(1) 4- {4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -6-methoxy-1-methyl-1H- [1,7] naphthididine-2-one
Figure 2020071550
以下に記載の化合物またはその医薬上許容される塩。
(1)7-メトキシ-1-[4-(3-(モルホリン-4-イル)アゼチジン-1-イル)フェニル]-1H-キノリン-4-オン 二塩酸塩
Figure 2020071550
The compounds listed below or pharmaceutically acceptable salts thereof.
(1) 7-Methoxy-1- [4- (3- (morpholine-4-yl) azetidine-1-yl) phenyl] -1H-quinoline-4-one dihydrochloride
Figure 2020071550
以下に記載の化合物またはその医薬上許容される塩。
(1)2-オキソ-4-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}-2H-クロメン-6-カルボキサミド
Figure 2020071550
The compounds listed below or pharmaceutically acceptable salts thereof.
(1) 2-oxo-4- {4- [2- (tetrahydropyran-4-yloxy) ethoxy] phenyl} -2H-chromen-6-carboxamide
Figure 2020071550
以下に記載の化合物群から選択されるいずれかの化合物またはその医薬上許容される塩。
(1)1-{4-[3-(2-ヒドロキシエトキシ)アゼチジン-1-イル]フェニル}-7-メトキシ-4-メチル-1,4-ジヒドロ-2H-ピリド[3,4-b]ピラジン-3-オン
Figure 2020071550

(2)1-{4-[3-(2-スルファモイルアミノエトキシ)アゼチジン-1-イル]フェニル}-7-メトキシ-4-メチル-1,4-ジヒドロ-2H-ピリド[3,4-b]ピラジン-3-オン
Figure 2020071550
Any compound selected from the group of compounds described below or a pharmaceutically acceptable salt thereof.
(1) 1- {4- [3- (2-Hydroxyethoxy) azetidine-1-yl] phenyl} -7-methoxy-4-methyl-1,4-dihydro-2H-pyrido [3,4-b] Pyrazine-3-on
Figure 2020071550

(2) 1-{4- [3- (2-Sulfamoylaminoethoxy) azetidine-1-yl] phenyl} -7-methoxy-4-methyl-1,4-dihydro-2H-pyrido [3,4 -b] Pyrazine-3-on
Figure 2020071550
以下に記載の化合物群から選択されるいずれかの化合物またはその医薬上許容される塩。
(1)8-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ナフタレン-2-カルボキサミド
Figure 2020071550

(2)3-{4-[2-(テトラヒドロピラン-4-イルオキシ)エトキシ]フェニル}ベンゾ[b]チオフェン-5-カルボキサミド
Figure 2020071550
Any compound selected from the group of compounds described below or a pharmaceutically acceptable salt thereof.
(1) 8- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] phenyl} naphthalene-2-carboxamide
Figure 2020071550

(2) 3- {4- [2- (Tetrahydropyran-4-yloxy) ethoxy] phenyl} benzo [b] thiophene-5-carboxamide
Figure 2020071550
請求項1〜22のいずれか一項に記載の化合物またはその医薬上許容される塩を有効成分として含有する医薬組成物。 A pharmaceutical composition containing the compound according to any one of claims 1 to 22 or a pharmaceutically acceptable salt thereof as an active ingredient. 請求項1〜22のいずれか一項に記載の化合物またはその医薬上許容される塩を有効成分として含有するCDK8阻害剤。 A CDK8 inhibitor containing the compound according to any one of claims 1 to 22 or a pharmaceutically acceptable salt thereof as an active ingredient. CDK8に関連する疾患を予防又は治療するために用いられる、請求項1〜22のいずれか一項に記載の化合物またはその医薬上許容される塩を有効成分として含有する医薬組成物。 A pharmaceutical composition containing, as an active ingredient, the compound according to any one of claims 1 to 22, or a pharmaceutically acceptable salt thereof, which is used for preventing or treating a disease related to CDK8. 骨代謝に関連する疾患を予防又は治療するために用いられる、請求項1〜22のいずれか一項に記載の化合物またはその医薬上許容される塩を有効成分として含有する医薬組成物。 A pharmaceutical composition containing, as an active ingredient, the compound according to any one of claims 1 to 22, or a pharmaceutically acceptable salt thereof, which is used for preventing or treating a disease related to bone metabolism. 癌の予防又は治療するために用いられる、請求項1〜22のいずれか一項に記載の化合物またはその医薬上許容される塩を有効成分として含有する医薬組成物。 A pharmaceutical composition containing, as an active ingredient, the compound according to any one of claims 1 to 22, or a pharmaceutically acceptable salt thereof, which is used for preventing or treating cancer.
JP2020551124A 2018-10-04 2019-10-04 CDK8 Inhibitors and Their Applications Pending JPWO2020071550A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
JP2018189561 2018-10-04
JP2018189561 2018-10-04
JP2018206050 2018-10-31
JP2018206050 2018-10-31
PCT/JP2019/039376 WO2020071550A1 (en) 2018-10-04 2019-10-04 Cdk8 inhibitor and use of same

Publications (1)

Publication Number Publication Date
JPWO2020071550A1 true JPWO2020071550A1 (en) 2021-09-24

Family

ID=70054688

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2020551124A Pending JPWO2020071550A1 (en) 2018-10-04 2019-10-04 CDK8 Inhibitors and Their Applications

Country Status (2)

Country Link
JP (1) JPWO2020071550A1 (en)
WO (1) WO2020071550A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4081524A1 (en) * 2019-12-23 2022-11-02 Bristol-Myers Squibb Company Substituted heteroaryl compounds useful as t cell activators
IL308122A (en) 2021-06-16 2023-12-01 Celgene Corp Azetidinyl compounds comprising a carboxylic acid group for the treatment of neurodegenerative diseases

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0500492D0 (en) * 2005-01-11 2005-02-16 Cyclacel Ltd Compound
WO2010118009A1 (en) * 2009-04-06 2010-10-14 Ptc Therapeutics, Inc. Hcv inhibitor and therapeutic agent combinations
WO2011136264A1 (en) * 2010-04-28 2011-11-03 第一三共株式会社 [5,6] heterocyclic compound
US20150018566A1 (en) * 2011-08-25 2015-01-15 The Provost, Fellows, Foundation Scholars, & the Other Members of Board, of The College of the Holy Tubulin binding agents
EP3082802B1 (en) * 2013-12-03 2020-02-26 Iomet Pharma Ltd. Tryptophan-2,3-dioxygenase (tdo) and/or indolamine-2,3-dioxygenase (ido) inhibitors and their use
JP2017522324A (en) * 2014-07-17 2017-08-10 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Novel naphthyridine and isoquinoline and their use as CDK8 / 19 inhibitors
CN106316942A (en) * 2015-06-25 2017-01-11 中山大学 A kind of Combretastatin A-4 analogs with a condensed ring structure as well as a preparation method, pharmaceutical compositions and applications thereof
WO2018013430A2 (en) * 2016-07-12 2018-01-18 Arisan Therapeutics Inc. Heterocyclic compounds for the treatment of arenavirus infection
AU2017395023B2 (en) * 2016-12-23 2022-04-07 Plexxikon Inc. Compounds and methods for CDK8 modulation and indications therefor
JP2021014409A (en) * 2017-11-21 2021-02-12 京都薬品工業株式会社 Protein-binding drug
CN108191834B (en) * 2017-12-28 2020-10-16 中山大学 Preparation method of benzo-fused N-heterocyclic compound

Also Published As

Publication number Publication date
WO2020071550A1 (en) 2020-04-09

Similar Documents

Publication Publication Date Title
ES2641478T3 (en) 3-Acetylamino-1- (phenyl-heteroaryl-aminocarbonyl or phenyl-heteroaryl-carbonylamino) benzene derivatives for the treatment of hyperproliferative disorders
EA027563B1 (en) Anticancer pyridopyrazines via the inhibition of fgfr kinases
US10662173B2 (en) Indole and pyrrole compounds, a process for their preparation and pharmaceutical compositions containing them
CA2935071A1 (en) Piperidine-dione derivatives
KR20160029808A (en) Substituted benzofuranyl and benzoxazolyl compounds and uses thereof
JP2004509893A (en) Substituted azepino [4,5b] indoline derivatives
CA2922933A1 (en) 2,8-diazaspiro[4.5]decane and 3,9-dispiro[5.5]undecane derivates and pharmaceutical compositions thereof useful as tryptophan hydroxylase inhibitors
EA010426B1 (en) Indazolopyridines and methods of making and using the same
CN106458983A (en) Novel compounds
JP2019206529A (en) Novel anticancer agent
EA022753B1 (en) Imidazothiadiazoles useful as inhibitors of kinases
EP3556761B1 (en) Pyrrolo-aromatic heterocyclic compound, preparation method therefor, and medical use thereof
EA010546B1 (en) Beta-carbolines useful for treating inflammatory diseases
CA3172478A1 (en) Bcl-2 inhibitor
JPWO2020045334A1 (en) Optically active azabicyclo ring derivative
JP2018522847A (en) 3-Amino-1,5,6,7-tetrahydro-4H-indol-4-ones
EP3194403A1 (en) Pyrrolopyrimidine derivatives as nr2b nmda receptor antagonists
WO2021226547A2 (en) Targeted nek7 inhibition for modulation of the nlrp3 inflammasome
JP7346425B2 (en) Dihydroceramide desaturase inhibitors for treating diseases
CA2933683A1 (en) Wnt pathway modulators
US20180118718A1 (en) Substituted Quinoxaline Derivatives
CA3189873A1 (en) 1h-benzo[d]imidazole derivatives as tlr9 inhibitors for the treatment of fibrosis
JPWO2020071550A1 (en) CDK8 Inhibitors and Their Applications
EP3882242A1 (en) Compound inhibiting yap-tead binding, and pharmaceutical composition for preventing or treating cancer, comprising compound as active ingredient
CN101663276A (en) 2-aminopyridine derivatives useful as kinase inhibitors

Legal Events

Date Code Title Description
A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20220929

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20231003

A02 Decision of refusal

Free format text: JAPANESE INTERMEDIATE CODE: A02

Effective date: 20240402