WO2016195083A1 - Novel heteroarylamino-3-pyrazole derivatives and pharmacologically acceptable salts thereof - Google Patents

Novel heteroarylamino-3-pyrazole derivatives and pharmacologically acceptable salts thereof Download PDF

Info

Publication number
WO2016195083A1
WO2016195083A1 PCT/JP2016/066617 JP2016066617W WO2016195083A1 WO 2016195083 A1 WO2016195083 A1 WO 2016195083A1 JP 2016066617 W JP2016066617 W JP 2016066617W WO 2016195083 A1 WO2016195083 A1 WO 2016195083A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
amino
pyrazol
pyrimidin
methanol
Prior art date
Application number
PCT/JP2016/066617
Other languages
French (fr)
Japanese (ja)
Inventor
誠治 堀
太志 長谷川
慎也 戸▲崎▼
雄介 今▲崎▼
智哉 柄澤
博之 上野
裕太 松村
チャン チア リー
Original Assignee
大日本住友製薬株式会社
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 大日本住友製薬株式会社 filed Critical 大日本住友製薬株式会社
Publication of WO2016195083A1 publication Critical patent/WO2016195083A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D403/12Heterocyclic 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 chain containing hetero atoms as chain links

Definitions

  • the present invention relates to a novel heteroarylamino-3-pyrazole derivative useful as a medicament and a pharmacologically acceptable salt thereof. More specifically, the present invention relates to a heteroarylamino-3-pyrazole derivative and a pharmacologically acceptable salt thereof, a pharmaceutical composition containing the derivative or a salt thereof, and a therapeutic agent for a pathological condition involving a kinase containing the composition. .
  • CDK cyclin-dependent kinase
  • CDC2 cyclin-dependent kinase
  • CDK2 CDK3, CDK4 and CDK6 have been shown to be involved in cell cycle progression.
  • CDK1 and CDK2 form a complex with cyclin E or cyclin A, respectively, and the transition from G1 phase to S phase or from S phase, respectively. It plays an essential role in the transition to G2.
  • Non-Patent Document 1 In mitosis, it has been shown that in addition to CDK1, a small number of specific kinases control a complex series of mitosis. Serine and threonine kinases have been shown to play an important role as a small number of specific kinases (Non-Patent Document 1). Among them, Aurora kinase is a chromosome with high fidelity. Is known to be controlled so as to be distributed to daughter cells (Non-patent Document 2).
  • AURKA Aurora kinase-A
  • AURKB Aurora kinase-B
  • CDK5 plays an important role in the development of nerves and the formation of brain functions.
  • CDK5 is related to neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease (Non-patent document 4), and a report that suggests an association with cancer (Non-patent document 5).
  • CDK1, cyclin A, cyclin E, AURKA and AURKB are overexpressed in many human solid cancers and blood cancers, and their expression and activity are reported to correlate with poor prognosis (non-patent) Therefore, it is considered that the utility value of a low molecular weight compound targeting CDK or Aurora as an anti-cancer agent is high because of Reference 6, Non-Patent Document 7, Non-Patent Document 8, Non-Patent Document 9, and Non-Patent Document 10).
  • Cancer stem cells are defined as a small number of cancer cells constituting cancer that have stem cell-like properties and high tumor-forming ability. Moreover, it may be called Tumor (Cancer) -Initiating Cell from a viewpoint of the main cause of malignant transformation of cancer. In recent years, the role of specific kinase signals in cancer stem cells has been elucidated (Non-patent Document 11).
  • the JAK2 / STAT3 signal is necessary for the survival of cancer stem cells in breast cancer and prostate cancer (Non-patent Document 12), and the TGF-beta signal is activated in cancer stem cells of breast cancer that are resistant to chemotherapy. (Non-Patent Document 13).
  • Myc known as a proto-oncogene, is considered to be one of the main causes of cancer malignancy, and it is known that the prognosis of breast cancer patients whose Myc signal is activated is extremely poor.
  • CDK1, CDK2, AURKA and AURKB inhibitors are used to suppress the growth of cancer cells that depend on the Myc signal and to selectively It is considered effective in inducing cell death (Non-Patent Document 14).
  • L is —O—
  • R 2 and R 2 ′ are —R, —T—W—R 6 and the like
  • R 3 is —R, etc.
  • R is a hydrogen atom
  • W is —C (R 6 ) 2 O— or the like
  • R 6 is a hydrogen atom or the like.
  • Patent Document 2 the following compound is disclosed in Patent Document 2.
  • K is NH, O, S or the like
  • A is aryl or the like
  • m is 0 or the like
  • X is O or S
  • R 1 is a hydrogen atom or the like
  • R 2 is amino or the like
  • R 3 is aryloxy or the like
  • R 4 is alkyl or the like.
  • Patent Document 3 wherein R 1 is a hydrogen atom, R 2 is a hydroxyl group, R 3 is an alkyl, R 4 is a hydrogen atom, etc., and R 5 is a hydrogen atom, etc. , R 6 is halogen or the like, R 7 is halogen or the like, n is 0 to 4, and p is 0 to 5. ]
  • Patent Document 4 the following compound is disclosed in Patent Document 4.
  • Patent Documents 1, 2, 3 and 4 do not disclose any compounds represented by the formula (1) of the present invention.
  • the problem to be solved by the present invention is to inhibit a plurality of kinases and signals selected from CDK1, CDK2, AURKA, AURKB, JAK2, CDK5 and the like necessary for cell cycle, cell proliferation or cancer stem cell survival as described above.
  • it is to provide a highly safe compound that exhibits an anticancer effect and has reduced side effects.
  • Another object of the present invention is to provide a highly safe compound that exerts cancer stem cell control action by inhibiting kinase and has reduced side effects.
  • the present inventors have found that a compound represented by the following formula (1) or a pharmacologically acceptable salt thereof (hereinafter sometimes referred to as “the compound of the present invention”) is CDK1, CDK2. , AURKA, AURKB, JAK2, CDK5, etc., have high inhibitory action on kinases selected from ARKKA, AURKB, JAK2, CDK5, etc. .
  • the compound has a PK profile based on suitable solubility and metabolic stability and / or weak cardiotoxicity and weak toxicity to normal cells. It came to complete.
  • the present invention is as follows.
  • X represents a nitrogen atom or CR 5 ;
  • Y is a hydrogen atom, C 1-6 alkyl (the group is substituted with the same or different 1 to 3 groups selected from a halogen atom, hydroxy, C 1-6 alkoxy and C 3-10 cycloalkyl) C 3-10 cycloalkyl (which is substituted with 1 to 4 groups identical or different selected from a halogen atom, hydroxy, C 1-6 alkyl and C 1-6 alkoxy).
  • R 1a , R 1b , R 1c , R 1d and R 1e are the same or different and each represents a hydrogen atom, a halogen atom, hydroxy, cyano, nitro, C 1-6 alkyl (the group is a halogen atom, hydroxy and C 1 Optionally substituted with 1 to 3 identical or different groups selected from -6 alkoxy), C 1-6 alkoxy (the same selected from halogen atom, hydroxy and C 1-6 alkoxy) Or optionally substituted with 1 to 3 different groups), amino, C 1-6 alkylamino (the group is the same or different 1 to 3 selected from halogen atom, hydroxy and C 1-6 alkoxy) ), C 2-12 dialkylamino (which is selected from halogen atom,
  • a C 1-6 alkylcarbonyl (the group may be the same or different 1-3 selected from halogen atom, hydroxy and C 1-6 alkoxy). Which may be substituted with R 4 represents a hydrogen atom, cyano, hydroxy, C 1-6 alkyl (the group may be substituted with the same or different 1 to 3 groups selected from a halogen atom, hydroxy and C 1-6 alkoxy) Or a C 3-10 cycloalkyl, which group may be substituted with 1 to 4 groups identical or different selected from a halogen atom, hydroxy and C 1-6 alkoxy; R 2 , R 3 and R 5 are the same or different and each represents a hydrogen atom, a halogen atom, hydroxy, cyano, nitro, C 1-6 alkyl (the group is a halogen atom, hydroxy, C 1-6 alkoxy and C 3 -10 may be substituted with the same or different 1 to 3 groups selected from cycloalkyl), C 1-6 alkoxy (the
  • X is CR 5 ; Item 4.
  • R 5 is a hydrogen atom, a halogen atom, or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or Its pharmacologically acceptable salt.
  • Item 4 The compound or a pharmacologically acceptable salt thereof according to any one of Items 1 to 3, wherein X is a nitrogen atom.
  • R 4 is a hydrogen atom, C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or C 3-10 cycloalkyl (the group is substituted with 1 to 4 fluorine atoms).
  • Item 7. The compound or a pharmacologically acceptable salt thereof according to any one of Items 1 to 6, which may be substituted with an atom.
  • R 4 is a hydrogen atom, C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or C 3-4 cycloalkyl (the group is substituted with 1 to 4 fluorine atoms).
  • Item 8 The compound or a pharmacologically acceptable salt thereof according to any one of Items 1 to 7, which may be substituted with an atom.
  • R 2 and R 3 are the same or different and are a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms), The compound according to any one of the above or a pharmacologically acceptable salt thereof.
  • R 1a , R 1b , R 1c , R 1d and R 1e are the same or different and are each a hydrogen atom, a halogen atom or C 1-6 alkyl (the same group selected from a fluorine atom and C 1-6 alkoxy) Or any one of items 1 to 9 which may be substituted with 1 to 3 different groups) or C 1-6 alkoxy (the group may be substituted with 1 to 3 fluorine atoms) Or a pharmacologically acceptable salt thereof.
  • R 1a , R 1b , R 1c , R 1d and R 1e are the same or different and each represents a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms)
  • Item 13 The compound or a pharmaceutically acceptable salt thereof according to any one of Items 1 to 12, wherein at least one of R 1a , R 1b , R 1c , R 1d and R 1e is a halogen atom.
  • Item 14 The compound or a pharmaceutically acceptable salt thereof according to any one of Items 1 to 13, wherein at least two of R 1a , R 1b , R 1c , R 1d and R 1e are halogen atoms.
  • Item 15 The compound or a pharmacologically acceptable salt thereof according to any one of Items 1 to 14, wherein at least two of R 1a , R 1b , R 1c , R 1d and R 1e are fluorine atoms.
  • Item 53 A pharmaceutical composition comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmaceutically acceptable salt thereof.
  • a kinase inhibitor comprising the compound according to any one of Items 1 to 17 or 52 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • Item 53 An anticancer agent comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof as an active ingredient.
  • the anticancer agent as described in.
  • Treating and / or treating cancer comprising administering to a patient in need of treatment a therapeutically effective amount of a compound according to any one of Items 1-17 or 52 or a pharmaceutically acceptable salt thereof. Or a way to prevent.
  • the cancer is breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain cancer, endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or blood cancer. Use as described in.
  • the cancer is breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor, endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or blood cancer 26.
  • a pharmacologically acceptable salt thereof is a pharmacologically acceptable salt thereof.
  • Item 20 The anticancer agent according to Item 20, wherein the cancer is blood cancer, myeloma, liver cancer, ovarian cancer, prostate cancer, lung cancer, osteosarcoma, colon cancer, breast cancer, skin cancer or epithelial cell cancer, 27.
  • Item 53 A cell cycle arrester comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof as an active ingredient.
  • Item 53 A cancer stem cell growth inhibitor comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof as an active ingredient.
  • Item 22 The anticancer agent according to Item 20 or Item 21, which has a stemness gene expression inhibitory action.
  • Item 53 A stemness gene expression inhibitor comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof as an active ingredient.
  • [Claim 35] An anticancer agent comprising a compound that inhibits three or more kinases.
  • Item 36 The anticancer agent according to Item 35, wherein the kinase is selected from the group consisting of CDK2, CDK5, JAK2, AURKA, AURKB, and CDK1.
  • An anticancer agent comprising a compound that inhibits two or more kinases selected from the group consisting of CDK2, CDK5 and JAK2.
  • [Claim 40] 40 The method according to Item 39, wherein the kinase is selected from the group consisting of CDK2, CDK5, JAK2, AURKA, AURKB, and CDK1.
  • [Section 41] A method for treating cancer, comprising inhibiting two or more kinases selected from the group consisting of CDK2, CDK5 and JAK2.
  • Item 51 A therapeutic agent for cancer having resistance to treatment with a chemotherapeutic agent, comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof as an active ingredient.
  • Item 46 The therapeutic agent according to Item 45, wherein the chemotherapeutic agent is a taxane anticancer agent.
  • Item 22 The anticancer agent according to Item 20 or Item 21, which has a cell cycle arresting action.
  • Item 48 The anticancer agent according to any one of Item 20, Item 21, or Item 47, which has an effect of inhibiting the growth of cancer stem cells.
  • Item 49 The anticancer agent according to any one of Items 20 to 21 or 47 to 48, which also has an effect of preventing cancer recurrence.
  • [Section 51] A method for suppressing the expression of a stemness gene, comprising inhibiting two or more kinases selected from the group consisting of CDK2, CDK5 and JAK2.
  • a hormonal therapeutic agent (2) a chemotherapeutic agent, (3) an immunizing agent comprising the compound according to any one of items 1 to 17 or 52 or a pharmacologically acceptable salt thereof as an active ingredient A therapeutic agent, and (4) an anticancer agent for use in combination with one or more drugs selected from the group consisting of cell growth factors or drugs that inhibit cell growth factor receptor action.
  • the compound represented by the formula (1), or a pharmacologically acceptable salt thereof may be combined with CDK1, CDK2, CDK5, JAK2, AURKA or AURKB, or other kinases.
  • CDK1, CDK2, CDK5, JAK2, AURKA or AURKB By having an excellent inhibitory effect on the interaction, it can be applied to the prevention and treatment of various symptoms involving kinases such as cancer diseases.
  • cardiotoxicity and toxicity to normal cells are weak, it can be applied to the prevention and treatment of various diseases involving kinases as an anticancer agent having particularly excellent therapeutic effects and few side effects.
  • FIG. 2 is a chart of a powder X-ray diffraction pattern of the compound of Example 89.
  • 2 is a DSC-TGA chart of the compound of Example 89.
  • 6 is a three-dimensional structure diagram obtained by X-ray crystal analysis of the crystal obtained in Example 90.
  • FIG. 2 is a chart of a powder X-ray diffraction pattern of the compound of Example 91.
  • 2 is a DSC-TGA chart of the compound of Example 91.
  • the vertical axis shows the change in tumor volume.
  • the horizontal axis shows the number of days after tumor transplantation.
  • the plot and error bar show the mean and standard error of the tumor volume, respectively.
  • # Indicates the result of a significant difference test (p ⁇ 0.05, Dunnett's test). About the compound obtained in Example 4, the measurement result of the body weight which performed the test shown in Test example 7 is represented. The vertical axis shows the change in body weight. The horizontal axis shows the number of days after tumor transplantation. The plot and error bar show the mean weight value and standard error, respectively. # Indicates the result of a significant difference test (p ⁇ 0.05, Students t-test). About the compound obtained in Example 4, the measurement result of the tumor diameter which performed the test shown in Test example 8 is represented. The vertical axis shows the change in tumor volume. The horizontal axis shows the number of days after tumor transplantation. The plot and error bar show the mean and standard error of the tumor volume, respectively.
  • # Indicates the result of a significant difference test (p ⁇ 0.025, Williams' test).
  • the compound obtained in Example 4 the measurement result of the body weight which performed the test shown in Test example 8 is represented.
  • the vertical axis shows the change in body weight.
  • the horizontal axis shows the number of days after tumor transplantation.
  • the plot and error bar show the mean weight value and standard error, respectively.
  • # Indicates the result of a significant difference test (p ⁇ 0.025, Williams' test).
  • the result of the Nanog expression level which performed the test shown in Test example 13 is shown.
  • the results of the control compounds Palbociclib and Docetaxel are also shown.
  • the compound obtained in Example 72 the result of the Nanog expression level which performed the test shown in Test example 13 is shown.
  • a group defined as “optionally substituted” is an unsubstituted or substituted group, and the number of substituents in the group is the number of substituents unless otherwise specified. There is no restriction
  • examples of the “halogen atom” include a fluorine atom, a chlorine atom, a bromine atom, and an iodine atom.
  • halogen atom include a fluorine atom, a chlorine atom, a bromine atom, and an iodine atom.
  • they are a fluorine atom or a chlorine atom, More preferably, it is a fluorine atom.
  • C 1-6 alkyl means a straight or branched saturated hydrocarbon group having 1 to 6 carbon atoms. Specific examples include, for example, methyl, ethyl, propyl, 1-methylethyl, butyl, 2-methylpropyl, 1-methylpropyl, 1,1-dimethylethyl, pentyl, 3-methylbutyl, 2-methylbutyl, 2,2 -Dimethylpropyl, 1-ethylpropyl, 1,1-dimethylpropyl, hexyl, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl and the like.
  • C 1-6 alkyl includes C 1-4 alkyl and the like, more preferably C 1-3 alkyl.
  • C 1-4 alkyl include methyl, ethyl, propyl, 1-methylethyl, butyl, 2-methylpropyl having 1 to 4 carbon atoms in the specific example of “C 1-6 alkyl”. , 1-methylpropyl, 1,1-dimethylethyl and the like.
  • C 1-3 alkyl include methyl, ethyl, propyl, 1-methylethyl and the like having 1 to 3 carbon atoms in the specific example of “C 1-6 alkyl”. In this document, for example, C 1-6 has 1 to 6 carbon atoms, C 1-4 has 1 to 4 carbon atoms, and C 1-3 has 1 to 3 carbon atoms. Yes, or C 6 represents 6 carbon atoms. The same applies to other numbers.
  • C 3-10 cycloalkyl means a cyclic alkyl having 3 to 10 carbon atoms, and includes a partially crosslinked structure. Specific examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, adamantyl and the like.
  • Preferred “C 3-10 cycloalkyl” includes C 3-8 cycloalkyl, more preferably C 3-6 cycloalkyl.
  • C 3-8 cycloalkyl means a cyclic alkyl of 3 to 8 carbon atoms.
  • C 3-10 cycloalkyl Specific examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl having 3 to 8 carbon atoms in the specific example of “C 3-10 cycloalkyl”.
  • C 3-6 cycloalkyl means a cyclic alkyl of 3 to 6 carbon atoms.
  • Specific examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like having 3 to 6 carbon atoms in the specific example of “C 3-10 cycloalkyl”.
  • C 1-6 alkoxy means “C 1-6 alkyloxy”, and the “C 1-6 alkyl” moiety is as defined above for “C 1-6 alkyl”. Specific examples include, for example, methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy, 2-methylpropoxy, 1-methylpropoxy, 1,1-dimethylethoxy, pentyloxy group, 3-methylbutoxy, 2-methylbutoxy, 2,2-dimethylpropoxy, 1-ethylpropoxy, 1,1-dimethylpropoxy, hexyloxy, 4-methylpentyloxy, 3-methylpentyloxy, 2-methylpentyloxy, 1-methylpentyloxy, 3,3-dimethyl Examples include butoxy, 2,2-dimethylbutoxy, 1,1-dimethylbutoxy, 1,2-dimethylbutoxy and the like.
  • C 1-6 alkoxy includes C 1-4 alkoxy.
  • Specific examples of “C 1-4 alkoxy” include methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy having 1 to 4 carbon atoms in the specific example of “C 1-6 alkoxy”.
  • C 1-6 alkyl moiety in “C 1-6 alkylcarbonyl” is synonymous with “C 1-6 alkyl” of the.
  • Specific examples of “C 1-6 alkylcarbonyl” include, for example, methylcarbonyl, ethylcarbonyl, propylcarbonyl, 1-methylethylcarbonyl, butylcarbonyl, 2-methylpropylcarbonyl, 1-methylpropylcarbonyl, 1,1- And dimethylethylcarbonyl.
  • Preferred “C 1-6 alkylcarbonyl” includes C 1-4 alkylcarbonyl.
  • C 1-4 alkylcarbonyl include, for example, methylcarbonyl, ethylcarbonyl, propylcarbonyl, 1-methylethylcarbonyl, butylcarbonyl, 2-methylpropylcarbonyl and the like.
  • C 1-6 alkyl moiety in “C 1-6 alkylamino” is the same as defined in the “C 1-6 alkyl”.
  • Specific examples of “C 1-6 alkylamino” include, for example, methylamino, ethylamino, propylamino, isopropylamino, butylamino, isobutylamino, tert-butylamino, pentylamino, hexylamino and the like.
  • Preferred “C 1-6 alkylamino” includes “C 1-4 alkylamino”.
  • C 1-4 alkylamino "C 1-6 alkylamino” methylamino carbon atoms is of 1 to 4 in the specific example of, ethylamino, propylamino, isopropylamino, butylamino, etc. Is mentioned.
  • C 2-12 dialkylamino means “amino in which two identical or different C 1-6 alkyls are bonded”, and the C 1-6 alkyl is as defined above.
  • Specific examples of “C 2-12 alkylamino” include, for example, dimethylamino, diethylamino, ethylmethylamino, methylpropylamino, ethylpropylamino, dipropylamino, isopropylmethylamino, isopropylethylamino, diisopropylamino, methylbutyl Examples include amino, ethylbutylamino, dibutylamino, diisobutylamino, ditert-butylamino, dipentylamino, dihexylamino and the like.
  • C 2-12 dialkylamino includes “C 2-8 dialkylamino”.
  • Specific examples of “C 2-8 dialkylamino” include, for example, dimethylamino, diethylamino, ethylmethylamino, methylpropylamino, ethylpropylamino, dipropylamino, isopropylmethylamino, isopropylethylamino, diisopropylamino, methylbutyl Amino, ethylbutylamino, dibutylamino, diisobutylamino, ditert-butylamino and the like can be mentioned.
  • “3- to 6-membered cyclic amino” includes 3- to 6-membered monocyclic cyclic amino containing at least one nitrogen atom.
  • the “3- to 6-membered cyclic amino” may further have one same or different heteroatom selected from a nitrogen atom, an oxygen atom or a sulfur atom.
  • the bond of the group is a nitrogen atom constituting the ring in “3- to 6-membered cyclic amino”.
  • the group also includes a cyclic amino in which a part of the ring contains an unsaturated bond.
  • 3- to 6-membered cyclic amino examples include aziridino, azetidino, pyrrolidino, imidazolidino, oxazolidino, thiazolidino, piperazino, piperidino, morpholino, thiomorpholino, tetrahydropyridino and the like.
  • Preferable “3- to 6-membered cyclic amino” includes “5- to 6-membered cyclic amino”.
  • 5-membered cyclic amino examples include pyrrolidino, imidazolidino, oxazolidino, thiazolidino, piperazino, piperidino, morpholino, thiomorpholino, tetrahydropyridino and the like.
  • “3- to 6-membered cyclic amino” and “5- or 6-membered cyclic amino” form a fused ring with C 3-6 cycloalkyl, 6-membered aryl or 5- or 6-membered heteroaryl. Also good.
  • Specific examples of the condensed ring include “group” represented by the following.
  • “3- to 8-membered saturated heterocyclic ring” means 3 to 8 atoms including 1 to 2 atoms independently selected from the group consisting of a nitrogen atom, an oxygen atom and a sulfur atom in addition to a carbon atom. It means a monocyclic or bicyclic saturated heterocyclic ring.
  • the group may be partially crosslinked or spirolated.
  • the group may be condensed with C 6-10 aryl or C 5-10 heteroaryl.
  • the nitrogen atom constituting the ring is not a bond of the “group”. That is, the group does not include concepts such as 1-pyrrolidino.
  • “3- to 8-membered saturated heterocycle” includes monocyclic 5- to 6-membered saturated heterocyclic groups (“5- to 6-membered monocyclic saturated heterocycle”).
  • “5- to 6-membered monocyclic saturated heterocycle” include pyrrolidine, piperidine, piperazine, morpholine, tetrahydrofuran, which are monocyclic 5- to 6-membered in the specific example of “3- to 8-membered saturated heterocycle” , Tetrahydropyran and the like.
  • C 6-10 monocyclic or polycyclic aryl means an aromatic hydrocarbon having 6 to 10 carbon atoms. Specific examples include phenyl, 1-naphthyl, 2-naphthyl group and the like. Preferably, phenyl is used.
  • the group may be C 4-6 cycloalkyl, or a 5- to 6-membered heterocyclic group having 1 to 3 of the same or different atoms selected from a nitrogen atom, an oxygen atom or a sulfur atom. .
  • the bond of the group is a carbon atom constituting the ring in “C 6-10 monocyclic or polycyclic aryl”. Specific examples include groups represented by the following.
  • 5- to 10-membered monocyclic or polycyclic heteroaryl refers to monocyclic 5 containing 1 to 4 atoms independently selected from the group consisting of a nitrogen atom, an oxygen atom and a sulfur atom. Means a 6-membered aromatic heterocyclic group or a bicyclic 8- to 10-membered aromatic heterocyclic group.
  • pyridyl pyridazinyl, isothiazolyl, pyrrolyl, furyl, thienyl, thiazolyl, imidazolyl, pyrimidinyl, thiadiazolyl, pyrazolyl, oxazolyl, isoxazolyl, pyrazinyl, triazinyl, triazolyl, imidazolidinyl, oxadiazolyl, triazolyl, indrylyl, , Quinolyl, isoquinolyl, benzofuranyl, benzothienyl, benzoxazolyl, benzothiazolyl, benzisoxazolyl, benzisothiazolyl, benzotriazolyl, benzimidazolyl, or 6,11-dihydrodibenzo [b, e] thiepinyl Is mentioned.
  • “5- to 10-membered monocyclic or polycyclic heteroaryl” includes 5- to 6-membered monocyclic heteroaryl.
  • Specific examples of the 5- to 6-membered monocyclic heteroaryl include monocyclic examples in the specific examples of “5- to 10-membered monocyclic or polycyclic heteroaryl”. More preferably, a 5- to 6-membered monocyclic aromatic heterocyclic ring having one or more nitrogen atoms in the ring is used.
  • Specific examples include pyridyl, pyrimidinyl and the like, and more preferably pyridyl.
  • the compounds of the present invention may have some or all atoms of isotopes (eg, D, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 35 S, 18 F, 125 I Etc.), and these compounds are also included in the compounds of the present invention.
  • isotopes eg, D, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 35 S, 18 F, 125 I Etc.
  • R 1a , R 1b , R 1c , R 1d , R 1e , R 2 , R 3 , R 4 , R 5 , X, Y and Ar in the compound represented by the formula (1) are shown below.
  • the technical scope of the invention is not limited thereto.
  • the pyrazole moiety of the compound represented by the formula (1) can take tautomers such as the following formulas (1a) and (1b), and all tautomers are included in the compound of the present invention. .
  • X is preferably a nitrogen atom or CR 5 . More preferably, a nitrogen atom is mentioned.
  • Y is preferably a hydrogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). More preferably, a hydrogen atom is mentioned.
  • R 1a ”, “R 1b ”, “R 1c ”, “R 1d ” or “R 1e ” are preferably the same or different and each represents a hydrogen atom, a halogen atom, C 1-6 alkyl (the group is fluorine Optionally substituted with the same or different 1 to 3 groups selected from an atom and C 1-6 alkoxy) or C 1-6 alkoxy (the group is substituted with 1 to 3 fluorine atoms) May be included). More preferably, they are the same or different and include a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). Even more preferably, a hydrogen atom or a halogen atom is the same or different.
  • R 2 ” and “R 3 ” are preferably the same or different and each represents a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). Can be mentioned.
  • R 4 is preferably a hydrogen atom, C 1-6 alkyl (optionally substituted with 1 to 3 fluorine atoms) or C 3-10 cycloalkyl (substituted with 1 to 4 fluorine atoms). May be included). More preferable examples include C 1-6 alkyl (which may be substituted with 1 to 3 fluorine atoms) and C 3-6 cycloalkyl (which may be substituted with 1 to 4 fluorine atoms). It is done. Even more preferred are C 1-3 alkyl ( optionally substituted with 1 to 3 fluorine atoms) and cyclopropyl.
  • R 5 is preferably a hydrogen atom, a halogen atom, or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms or C 1-6 alkoxy). More preferable examples include a hydrogen atom and C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). Even more preferred is a hydrogen atom.
  • Ar is preferably phenyl or pyridyl. More preferably, phenyl is mentioned.
  • “Pharmaceutically acceptable salts” include acid addition salts and base addition salts.
  • acid addition salts include inorganic acid salts such as hydrochloride, hydrobromide, sulfate, hydroiodide, nitrate, phosphate, or citrate, oxalate, acetate, formic acid Salt, propionate, benzoate, trifluoroacetate, methanesulfonate, benzenesulfonate, p-toluenesulfonate, camphorsulfonate, and other organic acid salts.
  • Inorganic base salts such as sodium salt, potassium salt, calcium salt, magnesium salt, ammonium salt, or organic base salts such as triethylammonium salt, triethanolammonium salt, pyridinium salt, diisopropylammonium salt, and the like, and arginine And amino acid salts such as basic or acidic amino acids such as aspartic acid and glutamic acid.
  • the compound of the present invention When it is desired to obtain the compound of the present invention as a salt, it can be purified as it is when the compound of the present invention is obtained in a salt form.
  • an appropriate organic solvent can be used. It may be dissolved or suspended in the solution, and an acid or base may be added to form a salt by a conventional method.
  • the compound of the present invention may exist in the form of an adduct (hydrate, solvate) with water or various solvents, and these adducts are also included in the present invention.
  • the present invention also includes all tautomers of the compounds of the present invention and crystal forms of all embodiments.
  • the compound of the present invention includes a prodrug of the compound represented by the formula (1) or a pharmacologically acceptable salt thereof. Moreover, solvates, such as these hydrates and ethanol solvates, are also included.
  • the term “prodrug of the compound represented by formula (1)” is a compound that is converted into a compound represented by formula (1) by a reaction with an enzyme, gastric acid or the like under physiological conditions in vivo.
  • it means a compound that is enzymatically oxidized, reduced, hydrolyzed, etc. and converted to a compound represented by formula (1).
  • the compound of the present invention includes compounds having one or more asymmetric carbon atoms in the molecule. Accordingly, the compound of the present invention includes optical isomers, racemates, diastereomers and the like. The compounds of the present invention also include cis-trans isomers, isomers resulting from axial asymmetry, and the like. As described above, the compound of the present invention includes all isomers and mixtures thereof.
  • Examples of the kinase in which the compound of the present invention exhibits inhibitory activity include various tyrosine kinases, various serine kinases, and various threonine kinases. CDK1, CDK2, CDK5, AURKA, AURKB, JAK2 and the like are preferable.
  • the compound of the present invention can be produced from a known compound by appropriately combining, for example, the following production method and a method analogous thereto, or a synthesis method well known to those skilled in the art.
  • the compound represented by the formula (1) can be synthesized, for example, by the following method.
  • LG and LG ′ are the same or different and each represents a leaving group such as a halogen atom, methanesulfonyloxy, p-toluenesulfonic acid oxy or trifluoromethanesulfonic acid oxy, phenoxy, trifluorophenoxy, Tetrafluorophenoxy, pentafluorophenoxy, nitrophenoxy and the like can be mentioned.
  • the other definitions are the same as those described in item 1.
  • Step 1-1 Synthesizing a compound of formula (A-3) by reacting a compound of formula (A-2) wherein X is a nitrogen atom with a compound of formula (A-1) in the presence of a base, if necessary. Can do.
  • the base is not particularly limited. For example, triethylamine, diisopropylethylamine, tributylamine, 1.5-diazabicyclo [4.3.0] non-5-ene (DBN), 1,8-diazabicyclo [5.4.
  • Organic bases such as undec-7-ene (DBU), pyridine, dimethylaminopyridine, picoline, N-methylmorpholine (NMM), or sodium bicarbonate, potassium bicarbonate, sodium carbonate, potassium carbonate, hydroxide
  • examples include inorganic bases such as sodium and potassium hydroxide.
  • the solvent should just be a solvent which does not react on the reaction conditions of this process. Specifically, for example, alcohol solvents such as methanol, ethanol, 2-propanol (isopropyl alcohol), t-butanol, such as diethyl ether, diisopropyl ether, tetrahydrofuran, methylcyclopentyl ether, anisole, 1,4-dioxane and the like.
  • Ether solvents aromatic hydrocarbon solvents such as benzene, toluene, chlorobenzene and xylene, ester solvents such as ethyl acetate and methyl acetate, N, N-dimethylformamide, N, N-dimethylacetamide, N-methyl-2
  • An aprotic solvent such as pyrrolidinone, 1,3-dimethyl-2-imidazolidinone, dimethyl sulfoxide, water, or a mixture thereof.
  • the reaction temperature is from ⁇ 80 ° C. under heating under reflux, and is usually from 25 ° C. to 90 ° C.
  • the reaction time is usually 30 minutes to 12 hours.
  • Step 1-2 A compound of the formula (A-2) in which X is a carbon atom is optionally combined with a compound of the formula (A-1) in Ulman type conditions (for example, in an aprotic solvent such as DMF, in the presence of a metal catalyst, Using a metal catalyst such as copper (II) acetate, or refluxing under Buchwald type conditions (for example, BINAP, Pd 2 (dba) 3 or Pd (OAc) 2 under a carbonate base such as cesium carbonate) The compound of the formula (A-3) is reacted by reacting using a palladium catalyst and a ligand such as dppf, Xantphos, etc.
  • Ulman type conditions for example, in an aprotic solvent such as DMF, in the presence of a metal catalyst, Using a metal catalyst such as copper (II) acetate, or refluxing under Buchwald type conditions (for example, BINAP, Pd 2 (dba) 3 or Pd (OAc) 2
  • reaction conditions such as heating and refluxing in an inert solvent such as toluene.
  • an inert solvent such as toluene.
  • a solvent what is necessary is just a solvent which does not react on the reaction conditions of this process.
  • Specific examples include ether solvents such as tetrahydrofuran and 1,4-dioxane.
  • the reaction temperature is from ⁇ 80 ° C. to heating under reflux, usually from 25 ° C. to 150 ° C.
  • the reaction time is usually 30 minutes to 12 hours.
  • Step 2 (Production method of the compound represented by the formula (A-5)): Synthesizing a compound of formula (A-5) by reacting a compound of formula (A-4) wherein X is a nitrogen atom with a compound of formula (A-1) in the presence of a base, if necessary. Can do.
  • the base is not particularly limited. For example, triethylamine, diisopropylethylamine, tributylamine, 1.5-diazabicyclo [4.3.0] non-5-ene (DBN), 1,8-diazabicyclo [5.4.
  • Organic bases such as undec-7-ene (DBU), pyridine, dimethylaminopyridine, picoline, N-methylmorpholine (NMM), or sodium bicarbonate, potassium bicarbonate, sodium carbonate, potassium carbonate, hydroxide
  • DBU undec-7-ene
  • NMM N-methylmorpholine
  • inorganic bases such as sodium and potassium hydroxide.
  • the solvent should just be a solvent which does not react on the reaction conditions of this process.
  • alcohol solvents such as methanol, ethanol, 2-propanol and t-butanol
  • ether solvents such as diethyl ether, diisopropyl ether, tetrahydrofuran, methylcyclopentyl ether, anisole and 1,4-dioxane
  • Aromatic hydrocarbon solvents such as benzene, toluene, chlorobenzene and xylene
  • ester solvents such as ethyl acetate and methyl acetate, N, N-dimethylformamide, N, N-dimethylacetamide, N-methyl-2-pyrrolidinone, 1 , 3-dimethyl-2-imidazolidinone
  • aprotic solvents such as dimethyl sulfoxide, water, or a mixture thereof.
  • the reaction temperature is from ⁇ 80 ° C. under heating under reflux, and is usually from 25 ° C. to 90 ° C.
  • the reaction time is usually 30 minutes to 12 hours.
  • Step 3 (Production method of compound represented by formula (A-3)):
  • a compound of the formula (A-3) can be synthesized by reacting a compound of the formula (A-5) in which X is a nitrogen atom with a cyanide ion in the presence of a base, if necessary.
  • the base is not particularly limited.
  • Organic bases such as octane (DABCO), 1,8-diazabicyclo [5.4.0] undec-7-ene (DBU), pyridine, dimethylaminopyridine, picoline, N-methylmorpholine (NMM) It is done.
  • DABCO octane
  • DBU 1,8-diazabicyclo [5.4.0] undec-7-ene
  • pyridine dimethylaminopyridine
  • picoline N-methylmorpholine (NMM) It is done.
  • NMM N-methylmorpholine
  • alcohol solvents such as methanol, ethanol, 2-propanol and t-butanol
  • ether solvents such as diethyl ether, diisopropyl ether, tetrahydrofuran, methylcyclopentyl ether, anisole and 1,4-dioxane
  • Aromatic hydrocarbon solvents such as benzene, toluene, chlorobenzene and xylene
  • ester solvents such as ethyl acetate and methyl acetate, N, N-dimethylformamide, N, N-dimethylacetamide, N-methyl-2-pyrrolidinone, 1 , 3-dimethyl-2-imidazolidinone
  • aprotic solvents such as dimethyl sulfoxide, water, or a mixture thereof.
  • the reaction temperature is usually from 25 ° C to 150 ° C.
  • the reaction time is usually 30 minutes to 12 hours.
  • Step 4 Synthesizing a compound of the formula (A-6) by reacting a compound of the formula (A-3) in which X is a carbon atom or a nitrogen atom with an organometallic species such as a substituted aryl Grignard reagent in an inert solvent.
  • the inert solvent should just be a solvent which does not react on the reaction conditions of this process. Specific examples include ether solvents such as tetrahydrofuran and diethyl ether.
  • the reaction temperature is from ⁇ 80 ° C. to heating under reflux, usually from ⁇ 10 ° C. to 25 ° C.
  • the reaction time is usually 30 minutes to 24 hours.
  • a compound represented by the formula (1) can be synthesized by reacting a compound of the formula (A-6) in which X is a carbon atom or a nitrogen atom with a reducing agent, an alkyl Grignard reagent, an alkylating reagent or the like.
  • a reducing agent include sodium borohydride, lithium aluminum hydride, diisobutylaluminum hydride and the like.
  • the alkyl Grignard reagent include methyl magnesium bromide and isopropyl magnesium bromide.
  • Examples of the alkylating reagent include trifluoromethyltrimethylsilane.
  • the solvent should just be a solvent which does not react on the reaction conditions of this process.
  • an alcohol solvent such as methanol or ethanol is used when a reducing agent is used
  • an ether solvent such as tetrahydrofuran or diethyl ether is used when an alkyl Grignard reagent is used
  • dimethyl is used when an alkylating reagent is used.
  • aprotic solvents such as acetamide and N-methyl-2-pyrrolidinone.
  • Step 5-2 A compound of the formula (A-6) in which X is a carbon atom or a nitrogen atom is reacted with hydrogen in the presence of a catalyst, in the presence or absence of a ligand, in the presence or absence of a base, to give a compound of the formula (1) Can be synthesized.
  • Catalysts include metals such as palladium and nickel, salts of transition metals such as ruthenium chloride and palladium acetate, complexes such as the BINAP complex of ruthenium acetate, (R) -RUCY TM -Xylbinap, (S) -RUCY TM -Xylbinap, etc. And illegal catalysts.
  • an asymmetric ligand such as BIAP can also be used.
  • the base include tert-butoxy potassium, tert-butoxy sodium, sodium ethoxide, sodium methoxide and the like.
  • the hydrogen source include ammonium formate in addition to hydrogen gas.
  • a solvent what is necessary is just a solvent which does not react on the reaction conditions of this process. Specific examples include alcohol solvents such as isopropyl alcohol, methanol and ethanol, and ether solvents such as tetrahydrofuran and diethyl ether.
  • the reaction temperature is usually from ⁇ 10 ° C. to 50 ° C. under heating to reflux from ⁇ 80 ° C.
  • the reaction time is usually 10 minutes to 48 hours.
  • any functional group other than the reactive site changes under the described reaction conditions, even when the use of a protecting group is not specifically stated, or the described method
  • the compound other than the reaction point is protected as necessary, and the target compound can be obtained by deprotection after completion of the reaction or after a series of reactions.
  • an ordinary protecting group described in literature for example, Protective Groups in Organic Synthesis, 3rd ed., TWGreene, John Wiley & Sons Inc. (1999) can be used.
  • examples of the amino protecting group include benzyloxycarbonyl, tert-butoxycarbonyl, acetyl, benzyl and the like
  • examples of the hydroxy protecting group include trialkylsilyl such as trimethylsilyl, tert-butyldimethylsilyl, Acetyl or benzyl can be mentioned respectively.
  • Introduction and elimination of protecting groups can be performed by methods commonly used in organic synthetic chemistry (see, for example, the above-mentioned Protective Groups in Organic Synthesis) or methods based thereon.
  • Suitable and pharmaceutically acceptable salts of the starting and target compounds are conventional non-toxic salts, such as organic acid salts (eg acetate, trifluoroacetate, maleate, fumarate) Citrate, tartrate, methanesulfonate, benzenesulfonate, formate, toluenesulfonate, etc.) and inorganic acid salts (eg hydrochloride, hydrobromide, hydroiodide, sulfate) Acid addition salts such as nitrates, phosphates, etc., salts with amino acids (eg arginine, aspartic acid, glutamic acid etc.), alkali metal salts (eg sodium salts, potassium salts etc.), alkaline earth metal salts (eg calcium Metal salts such as salts and magnesium salt
  • the intermediate or final product in the above production method may be converted as appropriate by its functional group (for example, protecting and deprotecting the functional group as necessary to establish an amino group, hydroxyl group, carbonyl group, halogen group, etc.) Can be converted into other compounds included in the present invention.
  • the functional group can be converted by a commonly used general method (for example, see Comprehensive Organic Transformations, R. C. Larock, John Wiley & Sons Inc. (1999)).
  • the intermediates and target compounds in each of the above production methods are isolated and purified by purification methods commonly used in synthetic organic chemistry such as neutralization, filtration, extraction, washing, drying, concentration, recrystallization, and various chromatography. can do.
  • the intermediate can be subjected to the next reaction without any particular purification.
  • optical isomers based on optically active centers there are optical isomers based on optically active centers, atropisomers based on axial or planar chirality generated by restraining intramolecular rotation, other stereoisomers, tautomerism Isomers, geometric isomers and the like may exist, but all possible isomers and mixtures thereof are included in the scope of the present invention.
  • optical isomers and atropisomers can be obtained as racemates, or can be obtained as optically active substances using optically active starting materials and intermediates.
  • the corresponding raw material, intermediate or final racemate is physically or chemically separated by a known separation method such as a method using an optically active column or a fractional crystallization method.
  • a known separation method such as a method using an optically active column or a fractional crystallization method.
  • a racemate can be reacted with an optically active resolving agent to synthesize two diastereomers, which can be separated by a method such as fractional crystallization utilizing the different physical properties (diastereomeric method).
  • the compound represented by the formula (1) when it is desired to obtain a pharmacologically acceptable salt of the compound of the present invention, if the compound represented by the formula (1) is obtained in the form of a pharmacologically acceptable salt, it may be purified as it is, When it is obtained in a free form, it may be dissolved or suspended in an appropriate organic solvent, and an acid or base may be added to form a salt by a conventional method.
  • the compound of the present invention is provided as, for example, an anticancer agent, and the cancer type to which it is applied is not limited, but specific examples include, for example, breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor And endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or hematological cancer.
  • preferable cancer types include breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor, or endometrial cancer, and more preferable cancer types are , Breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, or prostate cancer.
  • the cancer types to be applied are blood cancer, myeloma, liver cancer, osteosarcoma, skin cancer, epithelial cell carcinoma, breast cancer, lung cancer. Ovarian cancer, uterine cancer, colon cancer, prostate cancer or pharyngeal cancer.
  • preferable cancer types include blood cancer, myeloma, liver cancer, breast cancer, lung cancer, ovarian cancer, uterine cancer, colon cancer, prostate cancer or pharyngeal cancer
  • preferable cancer types include breast cancer, Mention may be made of lung cancer, ovarian cancer, uterine cancer, colon cancer, prostate cancer or pharyngeal cancer.
  • blood cancer is a concept including lymphoma and leukemia.
  • the “anticancer agent” has an effect of reducing or eliminating a carcinoma or not increasing the tumor when administered for the purpose of preventing and / or treating cancer.
  • prevention is an act of administering the active ingredient of the present invention to a healthy person who has not developed a disease, and is intended to prevent, for example, the onset of the disease.
  • Treatment is the act of administering the active ingredient of the present invention to a person (patient) diagnosed as having developed a disease by a doctor. For example, alleviating a disease or symptom, It is intended not to increase or to return to the state before the onset of the disease.
  • the therapeutic purpose also includes cases where the purpose of administration is prevention of worsening of diseases and symptoms or prevention of increase in carcinoma.
  • the compounds of the present invention are used orally or parenterally (for example, intravenous, subcutaneous or intramuscular injection, topical, rectal, transdermal or nasal) as pharmaceutical compositions when used for treatment.
  • examples of the composition for oral administration include tablets, capsules, pills, granules, powders, liquids, suspensions and the like. These preparations are prepared using conventionally known techniques, and can contain non-toxic and inert carriers or excipients usually used in the field of preparation.
  • the amount to be used varies depending on symptoms, age, administration method, etc.
  • the lower limit is 0.01 mg per day for adults (preferably 1 mg)
  • 5000 mg preferably 500 mg
  • intravenous injection for adults, 0.01 mg (preferably 0.1 mg) as the lower limit and 1000 mg (preferably 30 mg) as the upper limit per day, divided into one or several times, The effect is expected by administering according to the symptoms.
  • intermittent administration is also preferable in oral and intravenous injection depending on the patient's symptoms.
  • the compound of the present invention can be used in combination with other drugs for the purpose of enhancing its effect and / or reducing side effects.
  • the compound of the present invention can be used in combination with a drug such as a hormonal therapeutic agent, a chemotherapeutic agent, an immunotherapeutic agent, a cell growth factor or a drug that inhibits cell growth factor receptor action.
  • a drug that can be used in combination with the compound of the present invention is abbreviated as a concomitant drug.
  • hormone therapeutic agents include phosfestol, diethylstilbestrol, chlorotrianicene, medroxyprogesterone acetate, megestrol acetate, chlormadinone acetate, cyproterone acetate, danazol, allylestrenol, gestrinone, mepartricin, raloxifene, Olmeroxifene, levormeloxifene, antiestrogens (eg, tamoxifen citrate, toremifene citrate, etc.), pill formulations, mepithiostan, testrolactone, aminoglutethimide, LH-RH agonists (eg, goserelin acetate, buserelin, Leuprorelin, etc.), droloxifene, epithiostanol, ethinyl estradiol sulfonate, aromatase inhibitors (eg, fadrozole hydrochloride,
  • chemotherapeutic agent for example, alkylating agents, antimetabolites, anticancer antibiotics, plant-derived anticancer agents and the like are used. A typical example is described below.
  • alkylating agent examples include nitrogen mustard, nitrogen mustard hydrochloride-N-oxide, chlorambutyl, cyclophosphamide, ifosfamide, thiotepa, carbocon, improsulfan tosylate, busulfan, nimustine hydrochloride, mitoblonitol, melphalan, dacarbazine, ranimustine, estramustine phosphate sodium, triethylenemelamine, carmustine, lomustine, streptozidine, pipobrommann, etoglucid, carboplatin, cisplatin, dibrospium hydrochloride, fotemustine, prednimustine, pumitepa, ribomuthine, temozolomide, treosorphan, Trophosphamide, dinostatin stimamarer, adzeresin, systemustin, vizeresin and their D S formulation, and the like.
  • antimetabolite examples include mercaptopurine, 6-mercaptopurine riboside, thioinosine, methotrexate, pemetrexed, eocitabine, cytarabine, cytarabine okphosphat, ancitabine hydrochloride, 5-FU drugs (for example, fluorouracil, tegafur, UFT, Doxyfluridine, carmofur, galocitabine, emiteful, capecitabine, etc.), aminopterin, nerzarabine, leucoporin calcium, tabloid, butosine, folinate calcium, levofolinate calcium, cladribine, emitefur, fludarabine, gemcitabine, hydroxycarpamide treptostatine, pentostatin , Idoxyuridine, mitoguazone, thiazofurin, ambamustine, bendamustine and those DDS formulation, and the like.
  • 5-FU drugs for example, fluorouracil, tegafur
  • Anticancer antibiotics include, for example, actinomycin D, actinomycin C, mitomycin C, chromomycin A3, bleomycin hydrochloride, bleomycin sulfate, peplomycin sulfate, daunorubicin hydrochloride, doxorubicin hydrochloride, aclarubicin hydrochloride, pirarubicin hydrochloride, epirubicin hydrochloride, neo Examples include cartinostatin, myramicin, sarcomycin, carcinophylline, mitotane, zorubicin hydrochloride, mitoxantrone hydrochloride, idarubicin hydrochloride and their DDS preparations.
  • plant-derived anticancer agents include etoposide, etoposide phosphate, vinblastine sulfate, vincristine sulfate, vindesine sulfate, teniposide, paclitaxel, docetaxel, vinorelbine, and their DDS preparations.
  • immunotherapeutic agents include picibanil, krestin, schizophyllan, lentinan, ubenimex, interferon, interleukin, macrophage colony stimulating factor, granulocyte colony stimulating factor, erythropoietin, lymphotoxin, BCG vaccine, corynebacterium parvum, levamisole , Polysaccharide K, procodazole, anti-CTLA4 antibody, PD-1 antibody, Toll-like Receptors agonist (for example, TLR7 agonist, TLR8 agonist, TLR9 agonist, etc.).
  • the cell growth factor in the drug that inhibits the action of the cell growth factor and the receptor for the cell growth factor may be any substance as long as it is a substance that promotes cell growth, and usually has a molecular weight of 20,000 or less. Thus, a factor that exerts its action at a low concentration by binding to a receptor can be mentioned.
  • EGF epidermal growth factor
  • TGFalpha a substance having substantially the same activity
  • insulin or a substance having substantially the same activity
  • IGF insulin, IGF (insulin-insulin- like growth factor) -1, IGF-2, etc.
  • FGF fibroblast growth factor
  • CSF colony (stimukating factor), EPO (erythropoietin), IL-2 (interleukin-2), NGF (nerve growth factor), PDGF (platelet-derived growth factor), TGF) -Beta (transforming growth factor beta), HGF (hepatocyte growth factor) VEGF (vascular endothelial growth factor), heregulin, angiopoietin, etc.).
  • CSF colony (stimukating factor), EPO (erythropoietin), IL-2 (interleukin-2), NGF (nerve growth factor), PDGF (platelet-derived growth factor), TGF) -Beta (transforming growth factor beta), HGF (hepatocyte growth factor)
  • the administration period of the compound of the present invention and the concomitant drug is not limited, and these may be administered simultaneously to the administration subject or may be administered with a time difference. Moreover, it is good also as a mixture of this invention compound and a concomitant drug.
  • the dose of the concomitant drug can be appropriately selected based on the clinically used dose.
  • the compounding ratio of the compound of the present invention and the concomitant drug can be appropriately selected depending on the administration subject, administration route, target disease, symptom, combination and the like.
  • the concomitant drug may be used in an amount of 0.01 to 100 parts by weight per 1 part by weight of the compound of the present invention.
  • it can be used in combination with drugs such as antiemetics, sleep inducers, anticonvulsants and the like (these are also included in the concomitant drugs).
  • the compound of the present invention has a high inhibitory action on a kinase selected from CDK1, CDK2, AURKA, AURKB, JAK2, CDK5, etc., as shown in the test examples and the like in this specification, and various kinases involved. It can be applied to the prevention and treatment of symptoms such as cancer diseases.
  • preferred compounds exhibit a stemness gene expression suppressing action by inhibiting CDK5 (the stemness gene will be described later).
  • a substance that inhibits CDK5 other than the compound of the present invention for example, siRNA of CDK5 that can directly inhibit CDK5
  • the suppressive action of stemness gene expression by the CDK5 inhibitor can be confirmed.
  • a stemness gene is a gene that can induce self-replication and / or differentiation into different cells.
  • Examples of stemness genes include Nanog, Sox2, ⁇ -catenin, Oct4, and the like.
  • a preferred compound has an action of suppressing the expression of stemness gene (Test Example 11).
  • preferred compounds exhibit a sphere formation inhibitory action, and therefore can be expected to suppress the growth of cancer stem cells. Due to these effects, the preferred compounds of the present invention can be expected to be useful as anticancer agents having an effect of preventing cancer recurrence.
  • preferred compounds are also useful as therapeutic agents for cancer having resistance to treatment with chemotherapeutic agents.
  • chemotherapeutic agents here include, for example, taxane anticancer agents.
  • taxane anticancer agent in the present specification include an anticancer agent having a taxane skeleton, and examples thereof include paclitaxel and docetaxel.
  • G protein-coupled receptors novel targets for drug discovery in cancer.
  • Lappano R Maggiolini M. Nat Rev Drug Discov. 2011 Jan; 10 (1): 47 -60
  • Differential effect of adenosine receptors on growth of human colon cancer HCT 116 and HT-29 cell lines.Sakowicz-Burkiewicz M, Kitowska A, Grden M, Maciejewska I, Szutowicz A, Pawelczyk T. Arch Biochem Biophys. 2013 May; 533 (1 -2): 47-54 The adenosinergic system in cancer: Key therapeutic target. See Sorrentino R, Pinto A, Morello S. Oncoimmunology. 2013 Jan 1; 2 (1): e22448).
  • the compound of the present invention is a compound that is weak in agonistic and antagonistic properties to GPCR, and is considered to have little influence on GPCR and on tumor formation via GPCR.
  • THF Tetrahydrofuran
  • TFA Trifluoroacetic acid NaBH (OAc) 3 : Sodium triacetoxyborohydride
  • DMAP N, N-dimethyl-4-aminopyridine
  • Boc 2 O: Di-tert-butyldicarbonate
  • DMF N, N -Dimethylformamide
  • DIEA N-ethyldiisopropylamine
  • WSCI 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide
  • WSCI ⁇ HCl 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride
  • HOBt 1- Hydroxybenzotriazole HOBt ⁇ H 2 O: 1-hydroxybenzotriazole monohydrate Me: methyl Et: ethyl
  • DMA N, N-dimethylacetamide
  • NMP 1-methyl-2-pyrrolidinone Boc:
  • Reversed phase HPLC preparative purification was performed as follows. Purification was performed using a Gilson HPLC System. The column used was a YMC CombiPrep ODS-A column (5 ⁇ m, 50 ⁇ 20 mm ID), and the solvent was a mixed solvent system of CH 3 CN (containing 0.035% TFA) and water (containing 0.05% TFA). UV detection was performed at wavelengths of 210 nm, 220 nm, and 254 nm. The elution conditions are as follows.
  • the powder X-ray diffraction measurement was performed under the following conditions.
  • ⁇ Device X'pert-MPD (Spectris)
  • ⁇ X-ray Cu K ⁇ 1/45 kV / 40 mA -Incident slit: 15 mm (automatic) / Divergence prevention slit: 15 mm (automatic)
  • Sample plate Non-reflective Si plate
  • Step size 0.017 °
  • Scanning range 4-40 ° (2 ⁇ )
  • Accumulation time 100 seconds / step
  • the diffraction peak value at the diffraction angle 2 ⁇ (°) described in this specification may cause some measurement error depending on the measurement equipment or measurement conditions.
  • the measurement error may be within a range of ⁇ 0.2, preferably ⁇ 0.1.
  • DSC Differential scanning calorimetry
  • Thermogravimetry was performed under the following conditions.
  • ⁇ Device TGAQ500 (TA Instruments)
  • Measurement temperature range Room temperature to 250 °C
  • Temperature increase rate 10 °C / min
  • Vessel Platinum pan
  • Atmospheric gas flow rate Dry nitrogen, sample flow rate approx. 60 mL / min, balance flow rate approx. 40 mL / min
  • X-ray diffraction measurement was performed under the following conditions.
  • -X-ray diffractometer R-AXIS RAPID-R Rigaku (Control software: RAPID AUTO Ver.3.11 Rigaku, Analysis software: Crystal Structure Ver.4.01 Rigaku, Mercury Ver.3.1 Development (Build RC5) CCDC)
  • Measurement conditions X-ray: CuK ⁇ ray, output: 50kV-100mA, temperature: 93 ⁇ 1K
  • LC / MS analysis conditions for compound identification are as follows.
  • LC / MS measurement method Detector: ACQUITY (registered trademark) SQ deteceter (Waters)
  • HPLC ACQUITY UPLC (registered trademark) system
  • Column Waters ACQUITY UPLC (registered trademark) BEH C18 (1.7 ⁇ m, 2.1 mm ⁇ 30 mm)
  • Solvent A solution: 0.06% formic acid / H 2 O, B solution: 0.06% formic acid / MeCN Gradient condition: 0.0-1.3 min Linear gradient from B 2% to 96% Flow rate: 0.8 mL / min UV: 220 nm and 254 nm
  • Reference Example 2 6-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile To a solution of the compound obtained in Reference Example 1 (4.8 g) in DMSO (100 mL) was added sodium cyanide (2.24 g) and 1,4-diazabicyclo [2.2.2] octane (1.28 g). Stir for 3 hours at ° C. After cooling to room temperature, water was added to the reaction solution, and insoluble matters were removed by filtration. Ethyl acetate was added to the obtained filtrate, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and then dried over anhydrous sodium sulfate.
  • Reference Example 4 (3,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanone
  • THF 10 mL
  • 3,4-difluorophenylmagnesium bromide 0.5 M THF (3 mL) under ice cooling, and the mixture was stirred for 45 minutes under ice cooling.
  • 2N hydrochloric acid (1 mL) was added, the mixture was warmed to room temperature and stirred for 45 minutes.
  • Reference Example 5 (3,5-dimethoxyphenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanone
  • THF 120 mL
  • 3,5-dimethoxyphenylmagnesium bromide in 1M THF (80 mL)
  • 2N Hydrochloric acid 80 mL was added, and the mixture was stirred at room temperature for 13 hr.
  • Reference Example 8 tert-butyl 3-((6- (3,4-difluorobenzoyl) pyridin-2-yl) amino) -5-methyl-1H-pyrazole-1-carboxylate
  • a 1.5 M THF solution (12 mL) of 3,4-difluorophenylmagnesium bromide was added under ice cooling. The mixture was stirred at room temperature for 2 hours, and 10% aqueous potassium hydrogen sulfate solution (30 mL) was added.
  • Reference Example 11 (6-((5-Methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) (3,4,5-trifluorophenyl) methanone
  • THF 15 mL
  • a solution of 3,4,5-trifluorophenylmagnesium bromide in 0.3 M THF 8.4 mL
  • the mixture was stirred for 10 minutes and stirred at room temperature for 18.5 hours.
  • a saturated aqueous ammonium chloride solution was added to the reaction solution, and the mixture was stirred at room temperature for 3 hours.
  • Reference Example 12 (4-Chloro-3-fluorophenyl) (6-((5-methyl-1H-pyrazole) amino) pyridin-2-yl) methanone
  • a THF (8 mL) solution of the compound (250 mg) obtained in Reference Example 7 was added a 0.5 M THF solution (5 mL) of 3-fluoro-4-chlorophenylmagnesium bromide under ice cooling, and the mixture was stirred for 10 minutes under ice cooling. And stirred at room temperature for 18.5 hours. A saturated aqueous ammonium chloride solution was added to the reaction solution, and the mixture was stirred at room temperature for 3 hours.
  • Reference Example 13 tert-butyl 3-((6- (3,5-dimethoxybenzoyl) pyridin-2-yl) amino) -5-methyl-1H-pyrazole-1-carboxylate
  • a THF (6.7 mL) solution of the compound (500 mg) obtained in Reference Example 7 was added a 0.5 M THF solution (16 mL) of 3,5-dimethoxyphenylmagnesium bromide under ice cooling, and the mixture was stirred for 10 minutes under ice cooling. And stirred at room temperature for 18.5 hours.
  • a saturated aqueous ammonium chloride solution was added to the reaction solution, and the mixture was stirred at room temperature for 3 hours, and saturated aqueous sodium hydrogen carbonate was further added.
  • Reference Example 15 (4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanone
  • THF a 0.5 M THF solution
  • 2N hydrochloric acid 200 mL was added under ice cooling, and the mixture was warmed to room temperature and stirred for 14 hours.
  • Reference Example 17 4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile To a solution of the compound obtained in Reference Example 16 (1.24 g) in DMSO (100 mL) / isopropyl alcohol (3 mL), sodium cyanide (0.31 g) and 1,4-diazabicyclo [2.2.2] octane (0. 32 g) was added and stirred at 90 ° C. for 5 hours. After cooling to room temperature, water was added and insolubles were removed by filtration. Ethyl acetate was added to the obtained filtrate, and the target product was extracted into an organic layer.
  • Reference Example 18 (4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanone
  • a THF (4 mL) solution of the compound (0.2 g) obtained in Reference Example 17 a 1.0 M THF solution (5.6 mL) of 3,4,5-trifluorophenylmagnesium bromide was slowly added under ice cooling. .
  • 10% aqueous potassium hydrogen sulfate solution (30 mL) was added under ice cooling, and the mixture was warmed to room temperature and stirred for 16 hours.
  • Reference Example 19 (4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4-difluorophenyl) methanone
  • a solution of the compound (0.2 g) obtained in Reference Example 17 in THF 5 mL was slowly added a 0.5 M THF solution (7.5 mL) of 3,4-difluorophenylmagnesium bromide under ice cooling, and the mixture was stirred at room temperature. Stir for hours. 2N Hydrochloric acid (3.1 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 50 min.
  • Reference Example 21 (4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanone
  • a THF (30 mL) solution of the compound (4.2 g) obtained in Reference Example 2 2,4,5-trifluorophenylmagnesium bromide (120 mL of 1M THF solution) obtained in Reference Example 20 was slowly added under ice cooling. The mixture was stirred at room temperature for 2 hours and 50 minutes. 2N Hydrochloric acid (7 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 2 hr.
  • Reference Example 23 tert-butyl 3-((6-((3,4-difluorophenyl) (hydroxy) methyl) pyridin-2-yl) amino) -5-methyl-1H-pyrazole-1-carboxylate
  • Sodium borohydride 113 mg was added to a solution of the compound obtained in Reference Example 8 (1.00 g) in methanol (6 mL) and THF (6 mL), and the mixture was stirred at room temperature for 1 hour. Water and ethyl acetate were added to the reaction solution, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate.
  • Reference Example 25 6-((5-cyclopropyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile To a suspension of the compound obtained in Reference Example 24 (364 mg) in DMSO (1.4 mL) / isopropyl alcohol (0.70 mL), sodium cyanide (106 mg) and 1,4-diazabicyclo [2.2.2] octane ( 87 mg), and the mixture was stirred at 90 ° C. for 6 hours. After cooling to room temperature, water was added to the reaction mixture, and the mixture was extracted 3 times with ethyl acetate. The organic phase was washed with saturated brine, dried over sodium sulfate, and concentrated with an evaporator.
  • Reference Example 27 4-((5-cyclopropyl-1H-pyrazol-3-yl) amino) -6-methylpyrimidine-2-carbonitrile To a solution of the compound obtained in Reference Example 26 (0.32 g) in DMSO (1.4 mL) / isopropyl alcohol (0.70 mL), sodium cyanide (75 mg) and 1,4-diazabicyclo [2.2.2] octane ( 72 mg) and stirred at 90 ° C. for 6 hours. After cooling to room temperature, water was added to the reaction solution and filtered.
  • Reference Example 29 4-((5-ethyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile
  • DMSO 1.4 mL
  • isopropyl alcohol 0.70 mL
  • sodium cyanide 50 mg
  • 1,4-diazabicyclo [2.2.2] octane 47.7 mg
  • Reference Example 30 (4-((5-Cyclopropyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanone
  • a THF (10 mL) solution of the compound (87 mg) obtained in Reference Example 25 a 0.5 M THF solution (3.2 mL) of 3,4,5-trifluorophenylmagnesium bromide was slowly added under ice cooling. After stirring at room temperature for 2 hours, 2M hydrochloric acid (200 mL) was added under ice cooling, and the mixture was warmed to room temperature and stirred for 14 hours. Water (2.0 mL) and acetic acid (1.0 mL) were added and stirred for 1 hour.
  • Reference Example 31 (4-((5-Ethyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanone
  • THF 0.5 M THF solution
  • 2M hydrochloric acid 200 mL was added under ice cooling, and the mixture was warmed to room temperature and stirred for 14 hours.
  • Water (2.0 mL) and acetic acid (1.0 mL) were added and stirred for 1 hour.
  • Reference Example 33 6-Methyl-4- (methylthio) pyrimidin-2 (1H) -one
  • the compound (13.1 g) obtained in Reference Example 32 was added to 1M aqueous sodium hydroxide solution (287 mL), cooled to 0 ° C., iodomethane (6.0 mL) was added dropwise, and the mixture was stirred at 0 ° C. for 2 hours. did.
  • the mixture was further stirred at room temperature for 2.5 hours, cooled to 0 ° C., acetic acid (130 mL) was added, and the mixture was concentrated under reduced pressure. Water was added to the obtained solid and dissolved by heating to 100 ° C., then cooled to room temperature and recrystallized.
  • Reference Example 34 2-chloro-4-methyl-6- (methylthio) pyrimidine
  • the compound (442 mg) obtained in Reference Example 33 was added to phosphoryl chloride (4 mL) and stirred at 100 ° C. for 1.5 hours.
  • the mixture was allowed to cool to room temperature, and concentrated under reduced pressure, saturated aqueous sodium hydrogen carbonate and saturated brine were added, and the mixture was extracted with ethyl acetate.
  • the organic layer was washed with saturated brine, dried over sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (490 mg).
  • 1 H-NMR (CDCl 3 ) ⁇ 2.40 (s, 3H), 2.54 (s, 3H), 6.93 (s, 1H).
  • Reference Example 35 4-methyl-6- (methylthio) pyrimidine-2-carbonitrile To a solution of the compound obtained in Reference Example 34 (490 mg) in DMSO (3 mL) / isopropyl alcohol (1.7 mL) was added sodium cyanide (165 mg) and 1,4-diazabicyclo [2.2.2] octane (157 mg). And stirred at room temperature for 23 hours. Saturated aqueous sodium hydrogen carbonate, saturated brine and water were added to the reaction mixture, and the mixture was extracted with ethyl acetate. The organic layer was washed twice with saturated brine, dried over sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (436 mg). 1 H-NMR (CDCl 3 ) ⁇ 2.45 (s, 3H), 2.56 (s, 3H), 7.15 (s, 1H).
  • Reference Example 36 4-chloro-6-methylpyrimidine-2-carbonitrile
  • a solution of the compound obtained in Reference Example 35 (165 mg) in acetonitrile (10 mL) was cooled to 0 ° C., and sulfuryl chloride (0.4 mL) was added dropwise thereto, followed by stirring at 0 ° C. for 25 minutes.
  • a saturated aqueous sodium carbonate solution was added to the reaction mixture, and the mixture was extracted with chloroform. The organic layer was washed with saturated brine, dried over sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (159 mg).
  • 1 H-NMR (CDCl 3 ) ⁇ 2.59 (s, 3H), 7.40 (s, 1H).
  • Reference Example 37 4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile To a solution of the compound obtained in Reference Example 36 (154 mg) in DMSO (4 mL), 5-methyl-pyrazol-3-amine (146 mg) and N-ethyl-diisopropylamine (0.26 mL) were added and stirred at room temperature for 67 hours. did. Water was added, and the precipitated solid was filtered, washed with water, and then dried under reduced pressure to obtain the title compound (162 mg).
  • Reference Example 38 2,4-Dibromo-6-methyl-pyrimidine A solution of 2,4-dichloro-6-methylpyrimidine (40 g) in acetic acid (40 g) and methanesulfonic acid (160 g) was injected into a 30% hydrogen bromide / acetic acid solution (662 g) at 45 ° C. After stirring at 45 ° C. for 15 minutes, cooled toluene (200 g) was added, and the internal temperature was cooled to 0 ° C. Water (320 g), triethylamine (248 g) and 37.5% aqueous potassium carbonate solution (640 g) were successively added dropwise at an internal temperature of 5 ° C.
  • Reference Example 40 4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile 2-Bromo-6-methyl-N- (5-methyl-1H-pyrazol-3-yl) pyrimidin-4-amine (30 g) obtained in Reference Example 39 was added to DMF (240 g) / isopropyl alcohol (30 g). The mixture was suspended, sodium cyanide (7.4 g) was added, and the temperature was raised to 70 ° C. 1,4-diazabicyclo [2.2.2] octane (4 g) was added, the temperature was raised to 90 ° C., and the mixture was stirred for 11 hours.
  • Example 1 (3,4-Difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol
  • Synthesis Method A Sodium borohydride (3.6 mg) was added to a solution of the compound (30 mg) obtained in Reference Example 4 in methanol (2 mL), and the mixture was stirred at room temperature for 30 minutes. Water and ethyl acetate were added to the reaction solution, and the target product was extracted into the organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate.
  • Example 2 (-)-(3,4-Difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol and Example 3: (+)-(3,4-Difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol (Synthesis Method B) The racemic (3,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol (20 mg) obtained in Example 1 was added to hexane: isopropyl.
  • Example 6 ( ⁇ )-(6-((5-Methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) (3,4,5-trifluorophenyl) methanol and
  • Example 7 (+)-(6-((5-Methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) (3,4,5-trifluorophenyl) methanol
  • sodium borohydride 23 mg was added and stirred at room temperature for 3 hours and 20 minutes. Water and ethyl acetate were added to the reaction solution, and the target product was extracted into an organic layer.
  • Example 6 The racemic (6-((5-methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) (3,4,5-trifluorophenyl) methanol (28.7 g) was obtained in the same manner. )
  • Example 45 (3,4-Difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methane-d-ol
  • sodium deuteride 7.4 mg
  • 3N HCl and 5N NaOH were added to the reaction solution to adjust the pH of the reaction solution to 8, and then concentrated under reduced pressure. Water was added to the residue, and the precipitated solid was collected by filtration, washed with water and hexane, and then dried under reduced pressure to obtain the title compound (44 mg).
  • Example 46 1- (3-methoxyphenyl) -1- (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) ethane-1-ol
  • Methyl magnesium bromide (0.98M, 0.2 mL) was added to a THF (1 mL) solution of the compound (20 mg) obtained in Reference Example 3, and the mixture was stirred at room temperature for 20 hours. Thereafter, methylmagnesium bromide (0.98M, 1.5 mL) was added, and the mixture was stirred at room temperature for 20 hours. A saturated aqueous ammonium chloride solution and ethyl acetate were added to the reaction solution, and the target product was extracted into an organic layer.
  • Examples 47-52 The compounds of Examples 47 to 52 shown in the following table were obtained in the same manner as in Example 46 using the corresponding starting compounds.
  • Examples 53-70 The compounds of Examples 53 to 70 shown in the following table were obtained in the same manner as in Example 1 (Synthesis Method A) using the corresponding starting compounds.
  • Examples 71-86 The compounds of Examples 71 to 86 shown in the following table were obtained in the same manner as in Example 46 using the corresponding starting compounds.
  • Example 87 (+)- ⁇ 4-Methyl-6-[(5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl ⁇ (2,4,5-trifluorophenyl) methanol ethanol Japanese (4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanone (3.0 g) in isopropyl alcohol ( To a solution of 60 mL) / tetrahydrofuran (60 mL) were added potassium t-butoxy (58.2 mg) and (R) -RUCY TM -Xylbinap (40.9 mg) under a nitrogen atmosphere.
  • the reaction mixture was stirred at 40 ° C. for 2.5 hours under a hydrogen stream (5 atm). After replacing hydrogen in the reaction vessel with nitrogen, 3-mercaptopropyl silica gel (4.0 g) and activated carbon (2.0 g) were added to the reaction solution. The mixture was stirred for 1 hour, filtered through Celite, and the residue was washed with ethanol. The filtrate was concentrated with an evaporator to obtain a crude product (3.1 g). Ethanol (5 mL) was added thereto, and the mixture was stirred with an ultrasonic cleaner to confirm solid precipitation. The mixture was heated and stirred at 90 ° C. for 30 minutes, then slowly cooled to room temperature, and further cooled to 5 ° C. in an ice bath. The precipitated crystals were collected by filtration, further washed twice with cooled ethanol (2 mL), and dried under reduced pressure at 60 ° C. for 3 hours to obtain the title compound (2.39 g).
  • Example 88 (+)- ⁇ 4-Methyl-6-[(5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl ⁇ (2,4,5-trifluorophenyl) methanol (+)-(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) obtained in Example 87 Methanol Ethanolate (0.40 g) was suspended in water (8 mL). This suspension was heated and stirred at 90 ° C. for 1 hour. After cooling to room temperature, the precipitated crystals were collected by filtration. The crystals were further washed with water three times and dried under reduced pressure to obtain the title compound (0.34 g).
  • Example 89 (+)- ⁇ 4-Methyl-6-[(5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl ⁇ (2,4,5-trifluorophenyl) methanol (+)-(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-tri) obtained in the same manner as in Example 87.
  • Fluorophenyl) methanol Ethanolate (7.10 g) was suspended in water (71 mL).
  • the seed crystal obtained in Example 88 was added, and the mixture was heated with stirring at 90 ° C. for 6 hours. After cooling to room temperature, the precipitated crystals were collected by filtration. The crystals were further washed with water three times and dried under reduced pressure to obtain the title compound (6.08 g).
  • a chart of the powder X-ray diffraction pattern is shown in FIG. 1, and its main peak is shown in the table below.
  • a DSC-TGA chart is shown in FIG. From the DSC-TGA chart, since no weight change was observed at the endothermic peak, it was confirmed to be an anhydrous crystal.
  • Phosphoric acid 21 ⁇ L was added to a methanol (0.5 mL) suspension of methanol-ethanol product (100 mg), and the mixture was stirred at 50 ° C. for 1 hour. Further, the mixture was stirred at room temperature for 21 hours to confirm crystal precipitation. Ethyl acetate (2 mL) was added and stirred for 1.5 hours. The precipitated solid was collected by filtration, and the residue was washed three times with ethyl acetate (0.5 mL) and dried under reduced pressure at 50 ° C. to obtain the title compound (111 mg).
  • this compound (+)- ⁇ 4-methyl-6-[(5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl ⁇ (2,4,5-tri Since the absolute structure of (fluorophenyl) methanol phosphate) was found to be the (S) -form, the corresponding compounds ((+)-(4-methyl-6-((5- Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol) and the compound of Example 87 ((+)- ⁇ 4-methyl-6- [(5-Methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl ⁇ (2,4,5-trifluorophenyl) methanol ethanolate) was also determined to be the (S) -isomer, respectively. .
  • a chart of the powder X-ray diffraction pattern is shown in FIG.
  • a DSC-TGA chart is shown in FIG. From the DSC-TGA chart, since no weight change was observed at the endothermic peak, it was confirmed to be an anhydrous crystal.
  • Test Example 1 Cell Growth Inhibition Experiment NCI-H23 cells were obtained from ATCC (American Cultured Cell Line Storage Organization). NCI-H23 was cultured in RPMI 1640 medium containing 10% FCS and 1% penicillin / streptomycin, and HCT116 was cultured in DMEM medium containing 10% FCS and 1% penicillin / streptomycin in the presence of 37 ° C. and 5% CO 2 . Cells were seeded on a 96-well plate at 500-3000 cells / well, the test substance was added so that the final DMSO concentration was 0.1%, and the cells were cultured for 4-7 days. Thereafter, the number of viable cells was measured using Presto Blue (Life Technologies), and the concentration (Cytotoxicity IC 50 value; ⁇ M) of each test substance that inhibits cell growth was calculated. It was confirmed to have a growth inhibitory action.
  • Test Example 2 Cytotoxicity test using rat liver parenchymal cells
  • Test Example 3 Metabolic stability test of compound To 39.6 ml of 25 mM Kpi (pH 7.4), 0.4 ml of human liver microsome (Xenotech, approximately 20 mg protein / ml) was added to prepare a microsome solution. 10 ⁇ l of 1 mM test compound in DMSO was diluted 100-fold with acetonitrile. An intermediate diluted solution was prepared by adding 250 ⁇ l of Cofactor solution prepared by dissolving (6.5 mM) NADPH 300 mg in 55.2 ml of 125 mM Kpi (pH 7.4) to 5 ⁇ l of this solution.
  • Test Example 4 hERG inhibition test A test substance was added to the cultured hERG gene stably expressing CHO cell line so that the final DMSO concentration was 0.0135 to 0.5%. The hERG current was measured using QPatch HT (Sophion), the concentration at which each test substance suppresses 50% hERG current (IC 50 value; ⁇ M) was calculated, and the hERG inhibitory activity of the compound of the present invention was weak. confirmed.
  • Tests shown in Test Examples 1 to 4 were performed on the compounds obtained in the examples. The results are shown in the following table.
  • Test Example 5 Kinase inhibition experiment assay buffer (20 mM HEPES, 0.01% Triton X-100, 2 mM DTT, pH 7.5) 5 ⁇ L of 4-fold concentration test substance solution, 5 ⁇ L of 4-fold concentration substrate / ATP / The metal solution and 10 ⁇ L of the double concentration kinase solution were mixed in the wells of a polypropylene 384-well plate and reacted at room temperature for 1 hour. The reaction was stopped by adding 60 ⁇ L Termination Buffer (QuickScout Screening Assist MSA; Carna Biosciences). The substrate peptide and phosphorylated peptide in the reaction solution were separated and quantified by LabChip system (Perkin Elmer).
  • the kinase reaction was evaluated by the product ratio (P / (P + S)) calculated from the substrate peptide peak height (S) and the phosphorylated peptide peak height (P).
  • the average signal of the control well was 0% inhibition
  • the average signal of the background well was 100% inhibition
  • the inhibition rate was calculated from the average signal of each test substance well. From the results, it was confirmed that the compound of the present invention has kinase inhibitory activity.
  • Tests shown in Test Example 5 were performed on the compounds obtained in the examples. The test results are shown in the following table.
  • Test Example 6 Tumor Growth Suppression Experiment in HCT-116 Human Colon Adenocarcinoma Cell Carrying Mice HCT-116 cells (ATCC) were suspended in HBSS and 5-week-old female BALB / cAnNCrj-nu / nu mice (Charles Japan) 3 ⁇ 10 6 cells were transplanted subcutaneously on the ventral part of the river. From 7 days after transplantation, the test substance suspended in a 0.5% methylcellulose solution (Wako Pure Chemical Industries) was orally administered twice a day for 13 to 15 days to obtain a test substance administration group. A 0.5% methylcellulose solution was similarly administered to the vehicle administration group. The number of animals in each group was 7-8.
  • Tumor diameter and body weight were measured 1-3 times a week after the start of administration.
  • Tumor volume was calculated from the following equation.
  • Tumor volume (mm 3 ) major axis (mm) x minor axis (mm) x minor axis (mm) x 1/2
  • the tumor growth inhibition rate was calculated from the following formula using the average tumor volume of each administration group.
  • Tumor growth inhibition rate (%) 100-100 ⁇ (T-T0) / (C-C0) T: Average tumor volume after administration of test substance T0: Average tumor volume at the start of test substance administration
  • C Average tumor volume after vehicle administration
  • C0 Average tumor volume at the start of vehicle administration
  • the weight change rate was calculated from the following formula using the average body weight of each administration group.
  • Weight change rate (%) 100 x (T / T0) / (C / C0) T: Average body weight after administration of test substance T0: Average body weight at the start of test substance administration C: Average body weight after vehicle administration C0: Average body weight at the start of vehicle administration
  • Tests shown in Test Example 6 were performed on the compounds obtained in the examples. The results are shown in the following table.
  • Test Example 7 Tumor growth suppression experiment in HCC1806 human breast cancer cell-bearing mice HCC1806 cells (ATCC) were suspended in HBSS, and the ventral part of 5-week-old female BALB / cAnNCrj-nu / nu mice (Charles River Japan) 3 ⁇ 10 6 cells were transplanted subcutaneously.
  • test substance suspended in 0.5% methylcellulose solution (Wako Pure Chemical Industries) (200 mg / kg, twice a day, oral administration), docetaxel suspended in physiological saline (Otsuka Pharmaceutical) (5 mg / kg, once a week for 3 times, tail vein administration) or the compound obtained in Example 4 (150 mg / kg, twice a day, oral administration) and docetaxel (5 mg / kg, 1 time per week) 3 times, tail vein administration) was administered for 15 days.
  • 0.5% methylcellulose solution was administered twice a day for 15 days.
  • the number of animals in each group was 5-8.
  • the tumor diameter and body weight of all individuals were measured 1 to 3 times a week.
  • FIG. 6 shows the measurement results of the tumor diameter of the compound obtained in Example 4 in which the test shown in Test Example 7 was performed.
  • the tumor volume in the docetaxel administration group and the test substance and docetaxel combination administration group was significantly suppressed compared to that in the vehicle administration group (p ⁇ 0.05, Dunnett's test).
  • the tumor volume in the combination administration group of the test substance and docetaxel continued to be suppressed and disappeared completely.
  • no tumor recurrence was observed in all cases within the observation period.
  • FIG. 7 shows the measurement results of body weight of the compound obtained in Example 4 in which the test shown in Test Example 7 was performed.
  • the body weight (19th and 22nd days) of the docetaxel administration group was significantly suppressed compared with that of the vehicle administration group (p ⁇ 0.05, Student's ⁇ t-test).
  • the weight of the test substance and docetaxel combined administration group was not significantly changed from that of the vehicle administration group on any measurement day.
  • Test Example 8 Tumor growth inhibition experiment in tumor-bearing mice using FaDu human pharyngeal carcinoma cells FaDu cells (ATCC) were suspended in HBSS, and 5-week-old female BALB / cAnNCrj-nu / nu mice (Charles River Japan) 1 ⁇ 10 6 cells were transplanted subcutaneously in the ventral region of each other. From 7 days after transplantation, a test substance (50, 100, or 200 mg / kg, orally administered twice a day) suspended in a 0.5% methylcellulose solution (Wako Pure Chemical Industries) was administered for 15 days. In the vehicle administration group, 0.5% methylcellulose solution was administered twice a day for 15 days. The number of animals in each group was 10.
  • Tumor volume (mm 3 ) major axis (mm) x minor axis (mm) x minor axis (mm) x 1/2 8 and 9 show average values and standard errors.
  • FIG. 8 shows the measurement results of the tumor diameter of the compound obtained in Example 4 in which the test shown in Test Example 8 was performed. On the last day of measurement, the tumor volume of the test substance-administered group was significantly suppressed in a dose-dependent manner compared to that of the vehicle-administered group (p ⁇ 0.025, Williams' test).
  • FIG. 9 shows the measurement results of the body weight of the compound obtained in Example 4 in which the test shown in Test Example 8 was performed.
  • the body weight of the test substance-administered group significantly increased in a dose-dependent manner compared with that of the vehicle-administered group (p ⁇ 0.025, Williams' test). In this weight gain action, no abnormal cause such as edema was observed.
  • Test Example 9 Adenosine A3 antagonist activity test Coelenterazine h (125 nM) was loaded into CHO cells expressing adenosine A3 gene, aequorin and G ⁇ 16. Thereafter, a test substance was added, and 2 minutes later, a ligand (adenosine 1 ⁇ M) was added, aequorin luminescence was measured with FDSS 7000 (Hamamatsu Photonics), and the inhibition rate was calculated from the following formula.
  • Inhibition rate (luminescence intensity when no test substance is added-luminescence intensity when test substance is added) / Luminescence intensity with no test substance added * 100 From the results, it was confirmed that the compound of the present invention had no adenosine A3 antagonist activity at 1 ⁇ M.
  • Test Example 9 For the compound obtained in Example 4, the test shown in Test Example 9 was conducted. The test results are shown in the following table.
  • Test Example 10 Solubility Measurement A test substance (10 mM DMSO solution) was dispensed by 15 ⁇ L each into a Utube on a 96-well rack, set in a centrifugal evaporator, and evaporated to dryness. After 3 ⁇ L of DMSO was added and redissolved, 300 ⁇ L each of pH 7.4 and 1.2 buffers were added, and the mixture was shaken at 110 rpm for 90 minutes at 25 ° C. and then allowed to stand for 16 to 20 hours. Insoluble matters were separated by centrifugation at 2000 G for 15 minutes, and 100 ⁇ L of the supernatant was collected in a 96-well plate.
  • test substance 10 mM DMSO solution
  • a test substance 10 mM DMSO solution
  • a 100 ⁇ M standard solution was diluted 10-fold with 50% acetonitrile to obtain a 10 ⁇ M standard solution.
  • the solubility measurement sample and two standard solutions were analyzed by liquid chromatography, and the solubility was calculated from the area ratio with the standard solution. Tests shown in Test Example 10 were performed on the compounds obtained in the examples. The test results are shown in the following table.
  • Test Example 11 Bulk growth inhibition test HCC1806 cells were obtained from ATCC. HCC1806 cells were cultured in RPMI 1640 medium containing 10% fetal bovine serum and 1% penicillin / streptomycin in the presence of 37% at 5% CO 2 . Cells were seeded in a 96-well plate at 1,000 to 3,000 cells / well, a test substance was added so that the DMSO concentration was 0.1%, and the cells were cultured for 5 days. Thereafter, the number of viable cells was counted using Presto Blue (Life Technologies), and the concentration (IC 50 ; ⁇ M) at which 50% cell growth of each test substance was suppressed was calculated.
  • Presto Blue Presto Blue
  • Test Example 12 Cancer cell sphere formation inhibition experiment HCC1806 cells were obtained from ATCC. Cells were B27 Supplement (GIBCO), 20 ng / mL epidermal growth factor (EGF) (peprotech), 20 ng / mL basic fibroblast growth factor (bFGF) (peprotech), 5 ⁇ g / mL insulin (GIBCO) and 1 Cells were seeded at 1,000-3,000 cells / well in DMEM / F12 medium containing% methylcellulose (Nacalai Tesque) and seeded on Corning 96-well clear round bottom ultra-low attachment microplates (Corning, # 7007). A test substance was added so that the DMSO concentration was 0.1%, and the cells were cultured for 10 days. Thereafter, the number of spheres was counted, and the concentration (IC 50 : ⁇ M) at which 50% cell growth of each test substance was suppressed was calculated.
  • GEF epidermal growth factor
  • bFGF basic fibroblast growth factor
  • Test Example 13 Nanog Expression Inhibition Experiment with Test Substance HCC1806 cells were obtained from ATCC. The cells were subcultured in RPMI medium containing 10% fetal bovine serum and 1% penicillin / streptomycin in the presence of 5% CO 2 at 37 ° C. On the day before the addition of the test substance, the cells were seeded in 2 ⁇ 10 5 cells in a 6-well plate and cultured overnight. After adding a test substance and culturing for 24 hours, the supernatant was removed.
  • the cells were washed with ice-cold PBS, ice-cold RIPA buffer (Cell signaling technology, # 9806S) containing Proteinase / Phosphatase inhibitor cocktail (Cell signaling technology, # 5872S) was added, and the cells were collected using a cell scraper. After sonication, it was left on ice for 10 minutes. After centrifugation (15,000 rpm, 10 minutes), the supernatant was recovered and the protein concentration was quantified. A solution containing cell proteins adjusted to a constant concentration with RIPA buffer and SDS sample buffer (Nacalai Tesque, # 09499-14) was heated at 100 ° C. for 5 minutes, and then the proteins were separated by electrophoresis.
  • an anti-Nanog antibody (Cell signaling technology, # 4903S) or an anti- ⁇ -actin antibody (Cell signaling technology, # 5125S) is added using a 5% skim milk / TBST solution. Incubate overnight at 4 ° C. After washing the antibody-hybridized PVDF membrane with a TBST solution, a secondary antibody (HRP-labeled anti-rabbit IgG) was added and incubated at room temperature for 1 hour. After washing with TBST solution, a coloring reagent (ECL Prime western blotting detection reagent, GE healthcare, # RPN2232) was added, and luminescence was detected with a detector.
  • a coloring reagent ECL Prime western blotting detection reagent, GE healthcare, # RPN2232
  • Example 4 Using the compound obtained in Example 4 as a test substance, Palbociclib (selective CDK4 / 6 inhibitor; purchased from Selleck Chemicals) and Docetaxel (purchased from Sanofi Aventis) as control compounds were subjected to the test shown in Test Example 13. It was. A similar test was conducted using the compound obtained in Example 72 as a test substance. The results are shown in FIG. 10 (the compound of Example 4) and FIG. 11 (the compound of Example 72). The compounds in Example 4 and Example 72 decreased the expression level of Nanog in a concentration-dependent manner. On the other hand, Palbociclib and Docetaxel did not affect the expression level of Nanog. None of the test substances affected the expression level of ⁇ -actin.
  • Nanog expression inhibitory effect can be confirmed also about other Example compounds.
  • the effect of the compound of the present invention on the functions of other stemness genes can be confirmed.
  • the compound of Example 4 also showed an expression inhibitory effect on Sox2 and ⁇ -catenin.
  • the compound of the present invention is useful as a multikinase inhibitor for the prevention and / or treatment of diseases that may be affected by cell proliferation, for example, diseases such as cancer.

Abstract

Provided are highly safe compounds having reduced side effects including compounds represented by formula (1): [in formula (1), X indicates a nitrogen atom or CR5, Y indicates a hydrogen atom, C1-6 alkyl, or the like, R1a、R1b、R1c、R1d, and R1e are the same or different and indicate a hydrogen atom, halogen atom, hydroxy, or the like, R4 indicates a hydrogen atom, cyano, C1-6 alkyl, or the like, R2, R3, and R5 are the same or different and indicate a hydrogen atom, halogen atom, hydroxy, C1-6 alkyl, or the like, Ar indicates a C6-10 monocyclic or polycyclic aryl or 5-membered to 10-membered monocyclic or polycyclic heteroaryl] or a pharmacologically acceptable salt thereof that actualize an antitumor effect by inhibiting multiple kinases and signals such as CDK1、CDK2、CDK5、AURKA、AURKB、and JAK2 necessary for the cell cycle, cell proliferation, or survival of cancer stem cells.

Description

新規ヘテロアリールアミノ-3-ピラゾール誘導体およびその薬理学上許容される塩Novel heteroarylamino-3-pyrazole derivatives and pharmacologically acceptable salts thereof
 本発明は、医薬として有用な新規ヘテロアリールアミノ-3-ピラゾール誘導体およびその薬理学上許容される塩に関する。より詳しくは、ヘテロアリールアミノ-3-ピラゾール誘導体およびその薬理学上許容される塩、前記誘導体またはその塩を含有する医薬組成物、ならびに該組成物を含有するキナーゼが関与する病態の治療剤に関する。 The present invention relates to a novel heteroarylamino-3-pyrazole derivative useful as a medicament and a pharmacologically acceptable salt thereof. More specifically, the present invention relates to a heteroarylamino-3-pyrazole derivative and a pharmacologically acceptable salt thereof, a pharmaceutical composition containing the derivative or a salt thereof, and a therapeutic agent for a pathological condition involving a kinase containing the composition. .
 正常な体細胞が癌化する過程において、細胞分裂制御の異常は、根本的な特徴の一つとして知られている。細胞分裂は、4つの周期に分けられており、それらは、G1期、S期、G2期、および有糸分裂(M)期で、それぞれ異なったタンパク質が、その時期の制御に関わっている。 In the process of normal somatic cells becoming cancerous, abnormal cell division control is known as one of the fundamental features. Cell division is divided into four cycles, which are G1 phase, S phase, G2 phase, and mitosis (M) phase, and different proteins are involved in the control of that phase.
 現在使用されている細胞分裂の制御に関わる抗癌剤の多くは、これらの細胞周期の1つ、或いは複数期に関わっているタンパク質の活性を阻害するものであり、その臨床での有効性は様々な研究により証明されている。一方、現行の細胞分裂制御に関わる抗癌剤の多くが、阻害メカニズムに由来した副作用に加え、原因が明確でない様々な副作用を示すことも報告されている。そのため抗癌剤を十分投与出来ず、治療の限界が生じている場合がある。これらのことから、治療効果が優れかつ副作用の少ない抗癌剤の創生が望まれている。 Many of the currently used anticancer agents involved in the control of cell division inhibit the activity of proteins involved in one or more of these cell cycles, and their clinical effectiveness varies. Proven by research. On the other hand, it is reported that many of the current anticancer agents related to cell division control exhibit various side effects whose cause is not clear in addition to side effects derived from the inhibition mechanism. Therefore, anticancer drugs cannot be administered sufficiently, and there are cases where treatment limits have occurred. For these reasons, creation of an anticancer agent having an excellent therapeutic effect and few side effects is desired.
 細胞周期制御において、サイクリン依存性キナーゼ(cyclin-dependent kinase,CDK)は、その調節サブユニットであるサイクリンと同調し、G1期、S期、G2期およびM期の4段階を経て細胞周期の進行を制御する。CDKは、哺乳類細胞において少なくとも13種類が知られており、これらの内、CDK1(CDC2)、CDK2、CDK3、CDK4およびCDK6が、細胞周期進行に関わることが明らかとなっている。また、G1期からS期への移行にはすべてのCDKが関わるものの、特にCDK1とCDK2はサイクリンEまたはサイクリンAと複合体を形成し、それぞれ、G1期からS期への移行またはS期からG2期への移行に必須な役割を担っている。
 また、有糸分裂では、CDK1に加え少数の特定のキナーゼが複雑な一連の有糸分裂を制御することが明らかとなっている。少数の特定のキナーゼとしては、セリン・スレオニンキナーゼが重要な役割を担っていることが明らかとなっており(非特許文献1)、その中でも、オーロラキナーゼ(Aurora kinase)は、高い忠実度で染色体が娘細胞へ分配されるよう制御を行うことが知られている(非特許文献2)。オーロラキナーゼは、哺乳類細胞において3種類が知られており、これらの内、少なくともAURKA(オーロラキナーゼ-A)とAURKB(オーロラキナーゼ-B)は、染色体分離や細胞質分離に関与することが明らかとなっている。
 CDK5は、神経の発生や脳機能の形成に重要な役割を担っているとの報告がある(非特許文献3)。また、CDK5は、アルツハイマー病およびパーキンソン病のような神経変性疾患に関連するとの報告(非特許文献4)、および癌との関連性を示唆する報告がある(非特許文献5)。
In cell cycle control, cyclin-dependent kinase (CDK) synchronizes with its regulatory subunit, cyclin, and progresses through the four stages of G1, S, G2, and M phases. To control. At least 13 types of CDK are known in mammalian cells, and among these, CDK1 (CDC2), CDK2, CDK3, CDK4 and CDK6 have been shown to be involved in cell cycle progression. In addition, although all CDKs are involved in the transition from G1 phase to S phase, CDK1 and CDK2 form a complex with cyclin E or cyclin A, respectively, and the transition from G1 phase to S phase or from S phase, respectively. It plays an essential role in the transition to G2.
In mitosis, it has been shown that in addition to CDK1, a small number of specific kinases control a complex series of mitosis. Serine and threonine kinases have been shown to play an important role as a small number of specific kinases (Non-Patent Document 1). Among them, Aurora kinase is a chromosome with high fidelity. Is known to be controlled so as to be distributed to daughter cells (Non-patent Document 2). Three types of Aurora kinases are known in mammalian cells, and of these, at least AURKA (Aurora kinase-A) and AURKB (Aurora kinase-B) are known to be involved in chromosome separation and cytoplasm separation. ing.
There is a report that CDK5 plays an important role in the development of nerves and the formation of brain functions (Non-patent Document 3). In addition, there are reports that CDK5 is related to neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease (Non-patent document 4), and a report that suggests an association with cancer (Non-patent document 5).
 CDKおよびオーロラキナーゼの機能を阻害することにより、細胞周期進行の抑制、細胞増殖抑制、アポトーシスおよび分化や老化を促進し、抗腫瘍作用を示すことが報告されている。また、CDK1、サイクリンA、サイクリンE、AURKAおよびAURKBが、多くのヒト固形癌や血液癌において過剰発現しており、それらの発現や活性と予後不良が相関することも報告されている(非特許文献6,非特許文献7,非特許文献8,非特許文献9,非特許文献10)ため、CDKやAuroraを標的とした低分子化合物の抗癌剤としての利用価値は高いと考えられている。 It has been reported that inhibition of the functions of CDK and Aurora kinase promotes suppression of cell cycle progression, suppression of cell proliferation, apoptosis, differentiation and aging, and exhibits antitumor action. In addition, CDK1, cyclin A, cyclin E, AURKA and AURKB are overexpressed in many human solid cancers and blood cancers, and their expression and activity are reported to correlate with poor prognosis (non-patent) Therefore, it is considered that the utility value of a low molecular weight compound targeting CDK or Aurora as an anti-cancer agent is high because of Reference 6, Non-Patent Document 7, Non-Patent Document 8, Non-Patent Document 9, and Non-Patent Document 10).
 一方、血液癌、脳腫瘍、大腸癌、乳癌などいくつかの癌では、ガン幹細胞の存在が報告されている。癌幹細胞は癌を構成する癌細胞のうち、幹細胞様の性質を持ち、腫瘍形成能の高い少数の細胞と定義される。また、癌の悪性化の主原因という観点より、Tumor(Cancer)-Initiating Cellと呼称されることもある。近年、癌幹細胞における特定のキナーゼのシグナルの役割が明らかとなってきた(非特許文献11)。例えば、JAK2/STAT3シグナルは、乳癌や前立腺癌における癌幹細胞の生存に必要であること(非特許文献12)やTGF-betaシグナルが、化学療法に耐性を示す乳癌の癌幹細胞において活性化されていることが報告される(非特許文献13)。
 また、原癌遺伝子として知られるMycは、癌の悪性化の主原因の一要因と考えられており、Mycシグナルが活性化された乳癌患者の予後は極めて悪いことが知られている。さらにMycシグナルが恒常化された癌細胞株を用いた合成致死を誘導する標的分子の探索から、CDK1、CDK2、AURKAおよびAURKBの阻害剤が、Mycシグナルに依存する癌細胞の増殖抑制や選択的な細胞死の誘導に有効であると考えられている(非特許文献14)。
On the other hand, the presence of cancer stem cells has been reported in some cancers such as blood cancer, brain tumor, colon cancer, and breast cancer. Cancer stem cells are defined as a small number of cancer cells constituting cancer that have stem cell-like properties and high tumor-forming ability. Moreover, it may be called Tumor (Cancer) -Initiating Cell from a viewpoint of the main cause of malignant transformation of cancer. In recent years, the role of specific kinase signals in cancer stem cells has been elucidated (Non-patent Document 11). For example, the JAK2 / STAT3 signal is necessary for the survival of cancer stem cells in breast cancer and prostate cancer (Non-patent Document 12), and the TGF-beta signal is activated in cancer stem cells of breast cancer that are resistant to chemotherapy. (Non-Patent Document 13).
In addition, Myc, known as a proto-oncogene, is considered to be one of the main causes of cancer malignancy, and it is known that the prognosis of breast cancer patients whose Myc signal is activated is extremely poor. Furthermore, from the search for target molecules that induce synthetic lethality using cancer cell lines in which the Myc signal is constitutive, CDK1, CDK2, AURKA and AURKB inhibitors are used to suppress the growth of cancer cells that depend on the Myc signal and to selectively It is considered effective in inducing cell death (Non-Patent Document 14).
 これまでに、アリールアミノピラゾール誘導体としては、例えば下記化合物が、特許文献1に開示されている。
Figure JPOXMLDOC01-appb-C000002
[式中、ZまたはZは窒素原子を意味し、Qは-S-等であり、RおよびRは、-T-R、-L-Z-Rであり、Rは、-T-(D環)であり[D環は、5員から7員の単環等であり]、Tは、原子価結合等であり、Zは、C1-4アルキリデン鎖等であり、Lは、-O-等であり、R及びR2’は、-R、-T-W-R等であり、Rは、-R等であり、Rは、水素原子等であり、Wは、-C(RO-等であり、Rは、水素原子等である。]
So far, as the arylaminopyrazole derivatives, for example, the following compounds have been disclosed in Patent Document 1.
Figure JPOXMLDOC01-appb-C000002
[Wherein Z 1 or Z 2 represents a nitrogen atom, Q is —S— or the like, R x and R y are —T—R 3 , —L—Z—R 3 , and R 1 Is —T— (D ring) [D ring is a 5- to 7-membered monocycle, etc.], T is a valence bond, etc., Z is a C 1-4 alkylidene chain, etc. L is —O—, R 2 and R 2 ′ are —R, —T—W—R 6 and the like, R 3 is —R, etc., R is a hydrogen atom, etc. W is —C (R 6 ) 2 O— or the like, and R 6 is a hydrogen atom or the like. ]
 また、例えば下記化合物が、特許文献2に開示されている。
Figure JPOXMLDOC01-appb-C000003
[式中、Kは、NH、O、S等であり、Aは、アリール等であり、mは、0等であり、Xは、OまたはSであり、Rは、水素原子等であり、Rはアミノ等であり、Rは、アリールオキシ等であり、Rは、アルキル等である。]
Further, for example, the following compound is disclosed in Patent Document 2.
Figure JPOXMLDOC01-appb-C000003
[Wherein, K is NH, O, S or the like, A is aryl or the like, m is 0 or the like, X is O or S, and R 1 is a hydrogen atom or the like] , R 2 is amino or the like, R 3 is aryloxy or the like, and R 4 is alkyl or the like. ]
 また、例えば下記化合物が、特許文献3に開示されている。
Figure JPOXMLDOC01-appb-C000004
[式中、Rは、水素原子等であり、Rは水酸基等であり、Rは、アルキル等であり、Rは、水素原子等であり、Rは、水素原子等であり、Rは、ハロゲン等であり、Rは、ハロゲン等であり、nは、0~4であり、pは、0~5である。]
Further, for example, the following compounds are disclosed in Patent Document 3.
Figure JPOXMLDOC01-appb-C000004
[Wherein R 1 is a hydrogen atom, R 2 is a hydroxyl group, R 3 is an alkyl, R 4 is a hydrogen atom, etc., and R 5 is a hydrogen atom, etc. , R 6 is halogen or the like, R 7 is halogen or the like, n is 0 to 4, and p is 0 to 5. ]
 また、例えば下記化合物が、特許文献4に開示されている。
Figure JPOXMLDOC01-appb-C000005
 しかしながら、特許文献1、2、3及び4には、本発明の式(1)で表される化合物は、一切開示されない。
Further, for example, the following compound is disclosed in Patent Document 4.
Figure JPOXMLDOC01-appb-C000005
However, Patent Documents 1, 2, 3 and 4 do not disclose any compounds represented by the formula (1) of the present invention.
国際公開第2002/066461号International Publication No. 2002/066641 国際公開第2013/033862号International Publication No. 2013/033862 国際公開第2010/099379号International Publication No. 2010/099379 国際公開第2013/130600号International Publication No. 2013/130600
 本発明が解決しようとする課題は、上記のとおり細胞周期、細胞増殖または癌幹細胞の生存に必要な、CDK1、CDK2、AURKA、AURKB、JAK2、CDK5等から選ばれる複数のキナーゼおよびシグナルを阻害することにより抗癌作用を発揮し、副作用を低減した安全性の高い化合物を提供することである。また、キナーゼを阻害することにより癌幹細胞制御作用を発揮し、副作用を低減した安全性の高い化合物を提供することである。 The problem to be solved by the present invention is to inhibit a plurality of kinases and signals selected from CDK1, CDK2, AURKA, AURKB, JAK2, CDK5 and the like necessary for cell cycle, cell proliferation or cancer stem cell survival as described above. Thus, it is to provide a highly safe compound that exhibits an anticancer effect and has reduced side effects. Another object of the present invention is to provide a highly safe compound that exerts cancer stem cell control action by inhibiting kinase and has reduced side effects.
 本発明者らは、鋭意検討した結果、下記式(1)で表される化合物またはその薬理学上許容される塩(以下、「本発明化合物」と称することもある。)が、CDK1、CDK2、AURKA、AURKB、JAK2、CDK5等から選ばれるキナーゼに対して高い阻害作用を有すること、およびAdenosine A3受容体に対してアンタゴニスト活性を示さないことにより、優れた抗腫瘍作用を持つことを見出した。加えて、本発明化合物のうち好ましい化合物においては、該化合物が適した溶解度と代謝安定性に基づいたPKプロフィルを持つこと、および/または弱い心毒性と通常細胞に対する弱い毒性を確認し、本発明を完成させるに至った。 As a result of intensive studies, the present inventors have found that a compound represented by the following formula (1) or a pharmacologically acceptable salt thereof (hereinafter sometimes referred to as “the compound of the present invention”) is CDK1, CDK2. , AURKA, AURKB, JAK2, CDK5, etc., have high inhibitory action on kinases selected from ARKKA, AURKB, JAK2, CDK5, etc. . In addition, among the preferred compounds of the present invention, it is confirmed that the compound has a PK profile based on suitable solubility and metabolic stability and / or weak cardiotoxicity and weak toxicity to normal cells. It came to complete.
 すなわち、本発明は、以下の通りである。 That is, the present invention is as follows.
[項1]
式(1):
Figure JPOXMLDOC01-appb-C000006
[式(1)中、
 Xは、窒素原子またはCRを示し;
 Yは、水素原子、C1-6アルキル(該基は、ハロゲン原子、ヒドロキシ、C1-6アルコキシおよびC3-10シクロアルキルから選択される同一または異なる1~3個の基で置換されていてもよい)、C3-10シクロアルキル(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)または3員~8員の飽和複素環(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)を示し;
 R1a、R1b、R1c、R1dおよびR1eは、同一または異なって、水素原子、ハロゲン原子、ヒドロキシ、シアノ、ニトロ、C1-6アルキル(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)、C1-6アルコキシ(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)、アミノ、C1-6アルキルアミノ(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)、C2-12ジアルキルアミノ(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~6個の基で置換されていてもよい)、3~6員の環状アミノ(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)またはC1-6アルキルカルボニル(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)を示し;
 Rは、水素原子、シアノ、ヒドロキシ、C1-6アルキル(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)またはC3-10シクロアルキル(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)を示し;
 R、RおよびRは、同一または異なって、水素原子、ハロゲン原子、ヒドロキシ、シアノ、ニトロ、C1-6アルキル(該基は、ハロゲン原子、ヒドロキシ、C1-6アルコキシおよびC3-10シクロアルキルから選択される同一または異なる1~3個の基で置換されていてもよい)、C1-6アルコキシ(該基は、同一または異なる1~3個のハロゲン原子で置換されていてもよい)、アミノ、C1-6アルキルアミノ(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)、C2-12ジアルキルアミノ(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~6個の基で置換されていてもよい)、3~6員の環状アミノ(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)、C1-6アルキルカルボニル(該基は、同一または異なる1~3個のハロゲン原子で置換されていてもよい)、C3-10シクロアルキル(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)、3員~8員の飽和複素環(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)、C6-10アリール(該基は、ハロゲン原子、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)または5員~10員の単環式もしくは多環式のヘテロアリール(該基は、ハロゲン原子、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)を示し;
 Arは、C6-10の単環式もしくは多環式のアリールまたは5員~10員の単環式もしくは多環式のヘテロアリールを示す]
で表される化合物またはその薬理学上許容される塩。
[Claim 1]
Formula (1):
Figure JPOXMLDOC01-appb-C000006
[In Formula (1),
X represents a nitrogen atom or CR 5 ;
Y is a hydrogen atom, C 1-6 alkyl (the group is substituted with the same or different 1 to 3 groups selected from a halogen atom, hydroxy, C 1-6 alkoxy and C 3-10 cycloalkyl) C 3-10 cycloalkyl (which is substituted with 1 to 4 groups identical or different selected from a halogen atom, hydroxy, C 1-6 alkyl and C 1-6 alkoxy). Or a 3- to 8-membered saturated heterocyclic ring (the group is substituted with the same or different 1 to 4 groups selected from a halogen atom, hydroxy, C 1-6 alkyl and C 1-6 alkoxy) May be)
R 1a , R 1b , R 1c , R 1d and R 1e are the same or different and each represents a hydrogen atom, a halogen atom, hydroxy, cyano, nitro, C 1-6 alkyl (the group is a halogen atom, hydroxy and C 1 Optionally substituted with 1 to 3 identical or different groups selected from -6 alkoxy), C 1-6 alkoxy (the same selected from halogen atom, hydroxy and C 1-6 alkoxy) Or optionally substituted with 1 to 3 different groups), amino, C 1-6 alkylamino (the group is the same or different 1 to 3 selected from halogen atom, hydroxy and C 1-6 alkoxy) ), C 2-12 dialkylamino (which is selected from halogen atoms, hydroxy and C 1-6 alkoxy) 3 to 6-membered cyclic amino (which is selected from a halogen atom, hydroxy, C 1-6 alkyl and C 1-6 alkoxy), which may be substituted with the same or different 1-6 groups. Or a C 1-6 alkylcarbonyl (the group may be the same or different 1-3 selected from halogen atom, hydroxy and C 1-6 alkoxy). Which may be substituted with
R 4 represents a hydrogen atom, cyano, hydroxy, C 1-6 alkyl (the group may be substituted with the same or different 1 to 3 groups selected from a halogen atom, hydroxy and C 1-6 alkoxy) Or a C 3-10 cycloalkyl, which group may be substituted with 1 to 4 groups identical or different selected from a halogen atom, hydroxy and C 1-6 alkoxy;
R 2 , R 3 and R 5 are the same or different and each represents a hydrogen atom, a halogen atom, hydroxy, cyano, nitro, C 1-6 alkyl (the group is a halogen atom, hydroxy, C 1-6 alkoxy and C 3 -10 may be substituted with the same or different 1 to 3 groups selected from cycloalkyl), C 1-6 alkoxy (the group is substituted with 1 to 3 halogen atoms which are the same or different May be substituted with the same or different 1 to 3 groups selected from a halogen atom, hydroxy and C 1-6 alkoxy), amino, C 1-6 alkylamino, C 2-12 dialkylamino (in which the halogen atom, optionally substituted by the same or different 1-6 groups selected from hydroxy and C 1-6 alkoxy ), 3 cyclic amino (base to 6-membered, halogen atom, hydroxy, optionally substituted by the same or different 1 to 4 groups selected from C 1-6 alkyl and C 1-6 alkoxy ), C 1-6 alkylcarbonyl (the group may be substituted with the same or different 1 to 3 halogen atoms), C 3-10 cycloalkyl (the group is a halogen atom, hydroxy, C 1 3 to 8 membered saturated heterocyclic ring (which may be substituted with the same or different 1 to 4 groups selected from -6 alkyl and C 1-6 alkoxy), which is a halogen atom, hydroxy, Optionally substituted with 1 to 4 identical or different groups selected from C 1-6 alkyl and C 1-6 alkoxy), C 6-10 aryl (the group is a halogen atom, C 1-6 Archi And the same or different one to four monocyclic or heteroaryl (substrate polycyclic which may also be) or a 5- to 10-membered optionally substituted with a group selected from C 1-6 alkoxy, halogen And optionally substituted with 1-4 identical or different groups selected from atoms, C 1-6 alkyl and C 1-6 alkoxy;
Ar represents C 6-10 monocyclic or polycyclic aryl or 5- to 10-membered monocyclic or polycyclic heteroaryl]
Or a pharmacologically acceptable salt thereof.
[項2]
 Arが、フェニルまたはピリジルである項1に記載の化合物またはその薬理学上許容される塩。
[Section 2]
Item 2. The compound according to Item 1 or a pharmacologically acceptable salt thereof, wherein Ar is phenyl or pyridyl.
[項3]
 Arが、フェニルである項1または項2に記載の化合物またはその薬理学上許容される塩。
[Section 3]
Item 3. The compound or pharmacologically acceptable salt thereof according to Item 1 or 2, wherein Ar is phenyl.
[項4]
 Xが、CRであり;
 Rが、水素原子、ハロゲン原子またはC1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)である項1~3いずれか一項に記載の化合物またはその薬理学上許容される塩。
[Claim 4]
X is CR 5 ;
Item 4. The compound according to any one of Items 1 to 3, wherein R 5 is a hydrogen atom, a halogen atom, or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or Its pharmacologically acceptable salt.
[項5]
 Xが、窒素原子である項1~3いずれか一項に記載の化合物またはその薬理学上許容される塩。
[Section 5]
Item 4. The compound or a pharmacologically acceptable salt thereof according to any one of Items 1 to 3, wherein X is a nitrogen atom.
[項6]
 Yが、水素原子またはC1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)である項1~5いずれか一項に記載の化合物またはその薬理学上許容される塩。
[Claim 6]
Item 6. The compound according to any one of Items 1 to 5, wherein Y is a hydrogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or a pharmacologically thereof Acceptable salt.
[項7]
 Rが、水素原子、C1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)またはC3-10シクロアルキル(該基は、1~4個のフッ素原子で置換されていてもよい)である項1~6のいずれか一項に記載の化合物またはその薬理学上許容される塩。
[項8]
 Rが、水素原子、C1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)またはC3-4シクロアルキル(該基は、1~4個のフッ素原子で置換されていてもよい)である項1~7のいずれか一項に記載の化合物またはその薬理学上許容される塩。
[Claim 7]
R 4 is a hydrogen atom, C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or C 3-10 cycloalkyl (the group is substituted with 1 to 4 fluorine atoms). Item 7. The compound or a pharmacologically acceptable salt thereof according to any one of Items 1 to 6, which may be substituted with an atom.
[Section 8]
R 4 is a hydrogen atom, C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or C 3-4 cycloalkyl (the group is substituted with 1 to 4 fluorine atoms). Item 8. The compound or a pharmacologically acceptable salt thereof according to any one of Items 1 to 7, which may be substituted with an atom.
[項9]
 RおよびRが、同一または異なって、水素原子、ハロゲン原子またはC1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)である項1~8のいずれか一項に記載の化合物またはその薬理学上許容される塩。
[Claim 9]
R 2 and R 3 are the same or different and are a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms), The compound according to any one of the above or a pharmacologically acceptable salt thereof.
[項10]
 R1a、R1b、R1c、R1dおよびR1eが、同一または異なって、水素原子、ハロゲン原子、C1-6アルキル(該基は、フッ素原子およびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)またはC1-6アルコキシ(該基は、1~3個のフッ素原子で置換されていてもよい)である項1~9のいずれか一項に記載の化合物またはその薬理学上許容される塩。
[Section 10]
R 1a , R 1b , R 1c , R 1d and R 1e are the same or different and are each a hydrogen atom, a halogen atom or C 1-6 alkyl (the same group selected from a fluorine atom and C 1-6 alkoxy) Or any one of items 1 to 9 which may be substituted with 1 to 3 different groups) or C 1-6 alkoxy (the group may be substituted with 1 to 3 fluorine atoms) Or a pharmacologically acceptable salt thereof.
[項11]
 R1a、R1b、R1c、R1dおよびR1eが、同一または異なって、水素原子、ハロゲン原子またはC1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)である項1~10のいずれか一項に記載の化合物またはその薬理学上許容される塩。
[Section 11]
R 1a , R 1b , R 1c , R 1d and R 1e are the same or different and each represents a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) The compound according to any one of Items 1 to 10 or a pharmacologically acceptable salt thereof.
[項12]
 Rが、C1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)またはシクロプロピルである項1~11のいずれか一項に記載の化合物またはその薬理学上許容される塩。
[Claim 12]
Item 12. The compound or drug according to any one of Items 1 to 11, wherein R 4 is C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or cyclopropyl. A scientifically acceptable salt.
[項13]
 R1a、R1b、R1c、R1dおよびR1eのうち少なくとも1つが、ハロゲン原子である項1~12のいずれか一項に記載の化合物またはその薬理学上許容される塩。
[Claim 13]
Item 13. The compound or a pharmaceutically acceptable salt thereof according to any one of Items 1 to 12, wherein at least one of R 1a , R 1b , R 1c , R 1d and R 1e is a halogen atom.
[項14]
 R1a、R1b、R1c、R1dおよびR1eのうち少なくとも2つが、ハロゲン原子である項1~13のいずれか一項に記載の化合物またはその薬理学上許容される塩。
[Section 14]
Item 14. The compound or a pharmaceutically acceptable salt thereof according to any one of Items 1 to 13, wherein at least two of R 1a , R 1b , R 1c , R 1d and R 1e are halogen atoms.
[項15]
 R1a、R1b、R1c、R1dおよびR1eのうち少なくとも2つが、フッ素原子である項1~14のいずれか一項に記載の化合物またはその薬理学上許容される塩。
[Section 15]
Item 15. The compound or a pharmacologically acceptable salt thereof according to any one of Items 1 to 14, wherein at least two of R 1a , R 1b , R 1c , R 1d and R 1e are fluorine atoms.
[項16]
 以下の化合物群:
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール [実施例14等]、
(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール [実施例10等]、
(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,4-トリフルオロフェニル)メタノール [実施例21等]、
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,4-トリフルオロフェニル)メタノール [実施例31等]、
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール [実施例9]、
(4-(((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール [実施例8等]、
(3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例1等]、
(3,4-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例11]、
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール [実施例27]、
(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール [実施例26]、
(2,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例29]、
(2,4-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例30]、
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,6-トリフルオロフェニル)メタノール、
(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,6-トリフルオロフェニル)メタノール [実施例20]、
(2,3-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例35]、
(2,3-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(ペンタフルオロフェニル)メタノール[実施例22等]、
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(ペンタフルオロフェニル)メタノール、
(3,4-ジフルオロフェニル)(5-フルオロ-4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例24]、
(3,4-ジフルオロフェニル)(5-フルオロ-4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
(5-フルオロ-4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
(5-フルオロ-4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール [実施例33]、
(5-フルオロ-4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール、
(5-フルオロ-4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール [実施例25]、
(5-クロロ-4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール [実施例36]、
(5-クロロ-4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
1-(3,4-ジフルオロフェニル)-1-(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)エタン-1-オール [実施例49]、
1-(3,4-ジフルオロフェニル)-1-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)エタン-1-オール、
(4-((3-エチル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール [実施例54]、
(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール [実施例53]、
(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール [実施例57]、
1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)-1-(2,4,5-トリフルオロフェニル)エタン-1-オール [実施例72]、
(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(4-フルオロ-3-メチルフェニル)メタノール [実施例61]、
(4-クロロ-3-フルオロフェニル)(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)メタノール [実施例59]、
1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)-1-(4-フルオロ-3-メチルフェニル)エタン-1-オール [実施例79]、および
1-(4-クロロ-3-フルオロフェニル)-1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)エタン-1-オール [実施例73]
から選択される化合物である項1に記載の化合物またはその薬理学上許容される塩。
[Section 16]
The following compound groups:
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol [Example 14 etc.]
(4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol [Example 10 etc.],
(4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,4-trifluorophenyl) methanol [Example 21 etc.]
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,4-trifluorophenyl) methanol [Example 31, etc.]
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol [Example 9]
(4-(((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol [Example 8 etc.]
(3,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 1 etc.]
(3,4-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 11]
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol [Example 27]
(4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol [Example 26]
(2,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 29],
(2,4-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 30]
(4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,6-trifluorophenyl) methanol,
(4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,6-trifluorophenyl) methanol [Example 20],
(2,3-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 35],
(2,3-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
(4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (pentafluorophenyl) methanol [Example 22 etc.],
(4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (pentafluorophenyl) methanol,
(3,4-difluorophenyl) (5-fluoro-4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 24]
(3,4-difluorophenyl) (5-fluoro-4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
(5-fluoro-4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
(5-Fluoro-4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol [Example 33],
(5-fluoro-4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol,
(5-Fluoro-4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol [Example 25]
(5-Chloro-4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol [Example 36]
(5-chloro-4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
1- (3,4-difluorophenyl) -1- (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) ethane-1-ol [Example 49]
1- (3,4-difluorophenyl) -1- (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) ethane-1-ol,
(4-((3-Ethyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol [Example 54]
(4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol [Example 53]
(4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol [Example 57]
1- (4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) -1- (2,4,5-trifluorophenyl) ethane-1-ol [Examples] 72],
(4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (4-fluoro-3-methylphenyl) methanol [Example 61]
(4-Chloro-3-fluorophenyl) (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) methanol [Example 59]
1- (4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) -1- (4-fluoro-3-methylphenyl) ethan-1-ol Example 79 And 1- (4-chloro-3-fluorophenyl) -1- (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) ethane-1-ol [ Example 73]
Item 2. The compound according to Item 1, which is a compound selected from: or a pharmacologically acceptable salt thereof.
[項17]
 以下の化合物群:
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール [実施例14等]、
(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール [実施例10等]、
(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,4-トリフルオロフェニル)メタノール [実施例21等]、
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,4-トリフルオロフェニル)メタノール [実施例31等]、
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール [実施例9]、
(4-(((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール [実施例8等]、
(3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例1等]、
(3,4-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例11]、
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール [実施例27]、
(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール [実施例26]、
(2,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例29]、
(2,4-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例30]、
(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,6-トリフルオロフェニル)メタノール、
(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,6-トリフルオロフェニル)メタノール [実施例20]、
(2,3-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例35]、
(2,3-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
1-(3,4-ジフルオロフェニル)-1-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)エタン-1-オール、
(4-((3-エチル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール [実施例54]、
(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール [実施例53]、
(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール [実施例57]、
1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)-1-(2,4,5-トリフルオロフェニル)エタン-1-オール [実施例72]、
(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(4-フルオロ-3-メチルフェニル)メタノール [実施例61]、
(4-クロロ-3-フルオロフェニル)(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)メタノール [実施例59]、
1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)-1-(4-フルオロ-3-メチルフェニル)エタン-1-オール [実施例79]および
1-(4-クロロ-3-フルオロフェニル)-1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)エタン-1-オール [実施例73]
から選択される化合物である項1に記載の化合物またはその薬理学上許容される塩。
[Section 17]
The following compound groups:
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol [Example 14 etc.]
(4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol [Example 10 etc.],
(4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,4-trifluorophenyl) methanol [Example 21 etc.]
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,4-trifluorophenyl) methanol [Example 31, etc.]
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol [Example 9]
(4-(((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol [Example 8 etc.]
(3,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 1 etc.]
(3,4-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 11]
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol [Example 27]
(4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol [Example 26]
(2,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 29],
(2,4-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 30]
(4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,6-trifluorophenyl) methanol,
(4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,6-trifluorophenyl) methanol [Example 20],
(2,3-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 35],
(2,3-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
1- (3,4-difluorophenyl) -1- (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) ethane-1-ol,
(4-((3-Ethyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol [Example 54]
(4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol [Example 53]
(4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol [Example 57]
1- (4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) -1- (2,4,5-trifluorophenyl) ethane-1-ol [Examples] 72],
(4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (4-fluoro-3-methylphenyl) methanol [Example 61]
(4-Chloro-3-fluorophenyl) (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) methanol [Example 59]
1- (4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) -1- (4-fluoro-3-methylphenyl) ethan-1-ol Example 79 ] And 1- (4-chloro-3-fluorophenyl) -1- (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) ethane-1-ol Example 73]
Item 2. The compound according to Item 1, which is a compound selected from: or a pharmacologically acceptable salt thereof.
[項18]
 項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩を含有する医薬組成物。
[Section 18]
Item 53. A pharmaceutical composition comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmaceutically acceptable salt thereof.
[項19]
 項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩を有効成分として含有するキナーゼ阻害剤。
[Section 19]
53. A kinase inhibitor comprising the compound according to any one of Items 1 to 17 or 52 or a pharmaceutically acceptable salt thereof as an active ingredient.
[項20]
 項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩を有効成分として含有する抗癌剤。
[Section 20]
Item 53. An anticancer agent comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof as an active ingredient.
[項21]
 癌が、乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、前立腺癌、脳腫瘍、子宮内膜癌、膵臓癌、胃癌、骨肉腫、骨髄腫、または血液癌である項20に記載の抗癌剤。
[Claim 21]
Item 20 wherein the cancer is breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor, endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or blood cancer. The anticancer agent as described in.
[項22]
 治療が必要な患者に、治療上の有効量の項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩を投与することを含む、癌を治療および/または予防するための方法。
[Item 22]
Treating and / or treating cancer comprising administering to a patient in need of treatment a therapeutically effective amount of a compound according to any one of Items 1-17 or 52 or a pharmaceutically acceptable salt thereof. Or a way to prevent.
[項23]
 癌が、乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、前立腺癌、脳腫瘍、子宮内膜癌、膵臓癌、胃癌、骨肉腫、骨髄腫、または血液癌である項22に記載の治療および/または予防するための方法。
[Section 23]
Item 22 wherein the cancer is breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor, endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or blood cancer. A method for the treatment and / or prevention according to 1.
[項24]
 癌の治療剤および/または予防剤を製造するための、項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩の使用。
[Claim 24]
Item 53. Use of the compound according to any one of Items 1 to 17 or Item 52 or a pharmaceutically acceptable salt thereof for the manufacture of a therapeutic and / or prophylactic agent for cancer.
[項25]
 癌が、乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、前立腺癌、脳腫瘍、子宮内膜癌、膵臓癌、胃癌、骨肉腫、骨髄腫、または血液癌である項24に記載の使用。
[Claim 25]
Item 24. The cancer is breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain cancer, endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or blood cancer. Use as described in.
[項26]
 癌の治療および/または予防に使用するための項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩。
[Claim 26]
53. The compound or a pharmacologically acceptable salt thereof according to any one of Items 1 to 17 or Item 52 for use in the treatment and / or prevention of cancer.
[項27]
 癌が、乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、前立腺癌、脳腫瘍、子宮内膜癌、膵臓癌、胃癌、骨肉腫、骨髄腫、または血液癌である、項26に記載の化合物またはその薬理学上許容される塩。
[Section 27]
The cancer is breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor, endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or blood cancer 26. A pharmacologically acceptable salt thereof.
[項28]
 癌が、血液癌、骨髄腫、肝癌、卵巣癌、前立腺癌、肺癌、骨肉腫、大腸癌、乳癌、皮膚癌または上皮性細胞癌である項20に記載の抗癌剤、項22に記載の治療および/または予防するための方法、項24に記載の使用、または項26に記載の化合物またはその薬理学上許容される塩。
[Claim 28]
Item 20. The anticancer agent according to Item 20, wherein the cancer is blood cancer, myeloma, liver cancer, ovarian cancer, prostate cancer, lung cancer, osteosarcoma, colon cancer, breast cancer, skin cancer or epithelial cell cancer, 27. A method for preventing, a use according to Item 24, or a compound according to Item 26 or a pharmacologically acceptable salt thereof.
[項29]
 項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩を有効成分として含有する、細胞周期の停止剤。
[Item 29]
Item 53. A cell cycle arrester comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof as an active ingredient.
[項30]
 項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩を有効成分として含有する、がん幹細胞の増殖抑制剤。
[Section 30]
Item 53. A cancer stem cell growth inhibitor comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof as an active ingredient.
[項31]
 ステムネス遺伝子の発現抑制作用を有する、項20または項21に記載の抗癌剤。
[Claim 31]
Item 22. The anticancer agent according to Item 20 or Item 21, which has a stemness gene expression inhibitory action.
[項32]
 ステムネス遺伝子が、Nanog、Sox2、b-cateninおよびOct4からなる群から選ばれるいずれかである、項31に記載の抗癌剤。
[Section 32]
Item 32. The anticancer agent according to Item 31, wherein the stemness gene is any one selected from the group consisting of Nanog, Sox2, b-catenin and Oct4.
[項33]
 ステムネス遺伝子がNanogである、項31に記載の抗癌剤。
[Section 33]
Item 32. The anticancer agent according to Item 31, wherein the stemness gene is Nanog.
[項34]
 項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩を有効成分として含有する、ステムネス遺伝子の発現抑制剤。
[Section 34]
Item 53. A stemness gene expression inhibitor comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof as an active ingredient.
[項35]
 3種またはそれ以上のキナーゼを阻害する化合物を含有する、抗癌剤。
[Claim 35]
An anticancer agent comprising a compound that inhibits three or more kinases.
[項36]
 キナーゼがCDK2、CDK5、JAK2、AURKA、AURKBおよびCDK1からなる群から選ばれる、項35に記載の抗癌剤。
[Claim 36]
Item 36. The anticancer agent according to Item 35, wherein the kinase is selected from the group consisting of CDK2, CDK5, JAK2, AURKA, AURKB, and CDK1.
[項37]
 CDK2、CDK5およびJAK2からなる群から選ばれる2またはそれ以上のキナーゼを阻害する化合物を含有する、抗癌剤。
[Section 37]
An anticancer agent comprising a compound that inhibits two or more kinases selected from the group consisting of CDK2, CDK5 and JAK2.
[項38]
 CDK2、CDK5およびJAK2をいずれも阻害する化合物を含有する、抗癌剤。
[Section 38]
An anticancer agent comprising a compound that inhibits all of CDK2, CDK5, and JAK2.
[項39]
 3種またはそれ以上のキナーゼを阻害することを含む、癌の治療方法。
[Section 39]
A method of treating cancer comprising inhibiting three or more kinases.
[項40]
 キナーゼがCDK2、CDK5、JAK2、AURKA、AURKBおよびCDK1からなる群から選ばれる、項39に記載の方法。
[Claim 40]
40. The method according to Item 39, wherein the kinase is selected from the group consisting of CDK2, CDK5, JAK2, AURKA, AURKB, and CDK1.
[項41]
 CDK2、CDK5およびJAK2からなる群から選ばれる2またはそれ以上のキナーゼを阻害することを含む、癌の治療方法。
[Section 41]
A method for treating cancer, comprising inhibiting two or more kinases selected from the group consisting of CDK2, CDK5 and JAK2.
[項42]
 CDK2、CDK5およびJAK2をいずれも阻害することを含む、癌の治療方法。
[Section 42]
A method for treating cancer, comprising inhibiting all of CDK2, CDK5 and JAK2.
[項43]
 ステムネス遺伝子の発現を抑制することを含む、癌の治療方法。
[Section 43]
A method for treating cancer, comprising suppressing the expression of a stemness gene.
[項44]
 CDK5を阻害することを含む、ステムネス遺伝子の発現抑制方法。
[Item 44]
A method for suppressing the expression of a stemness gene, comprising inhibiting CDK5.
[項45]
 項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩を有効成分として含有する、化学療法剤による治療抵抗性を有する癌の治療剤。
[Section 45]
Item 51. A therapeutic agent for cancer having resistance to treatment with a chemotherapeutic agent, comprising the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof as an active ingredient.
[項46]
 化学療法剤がタキサン系抗癌剤である、項45記載の治療剤。
[Claim 46]
Item 46. The therapeutic agent according to Item 45, wherein the chemotherapeutic agent is a taxane anticancer agent.
[項47]
 細胞周期の停止作用を有する、項20または項21に記載の抗癌剤。
[Section 47]
Item 22. The anticancer agent according to Item 20 or Item 21, which has a cell cycle arresting action.
[項48]
 がん幹細胞の増殖抑制作用を有する、項20、項21または項47のいずれか一項に記載の抗癌剤。
[Section 48]
Item 48. The anticancer agent according to any one of Item 20, Item 21, or Item 47, which has an effect of inhibiting the growth of cancer stem cells.
[項49]
 癌の再発予防作用を併せ持つ、項20~21または項47~48のいずれか一項に記載の抗癌剤。
[Item 49]
Item 49. The anticancer agent according to any one of Items 20 to 21 or 47 to 48, which also has an effect of preventing cancer recurrence.
[項50]
 癌の転移抑制作用を併せ持つ、項20~21または項47~49のいずれか一項に記載の抗癌剤。
[Section 50]
Item 52. The anticancer agent according to any one of Items 20 to 21 or 47 to 49, which also has a cancer metastasis suppressing effect.
[項51]
 CDK2、CDK5およびJAK2からなる群から選ばれる2またはそれ以上のキナーゼを阻害することを含む、ステムネス遺伝子の発現抑制方法。
[Section 51]
A method for suppressing the expression of a stemness gene, comprising inhibiting two or more kinases selected from the group consisting of CDK2, CDK5 and JAK2.
[項52]
 以下の化合物群:
(-)-(3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例2]、
(+)-(3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール [実施例3]、
(+)-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール [実施例4]、
(-)-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール [実施例5]、
(-)-(6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール [実施例6]、
(+)-(6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール [実施例7]、
(+)-{4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,3,4-トリフルオロフェニル)メタノール [実施例32]、および
(-)-{4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,3,4-トリフルオロフェニル)メタノール [実施例31]
から選択される化合物である項1に記載の化合物またはその薬理学上許容される塩。
[Section 52]
The following compound groups:
(−)-(3,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 2]
(+)-(3,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol [Example 3]
(+)-(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol [Example 4] ,
(−)-(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol [Example 5] ,
(−)-(6-((5-Methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) (3,4,5-trifluorophenyl) methanol [Example 6]
(+)-(6-((5-methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) (3,4,5-trifluorophenyl) methanol [Example 7]
(+)-{4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,3,4-trifluorophenyl) methanol [Example 32] And (−)-{4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,3,4-trifluorophenyl) methanol [Examples] 31]
Item 2. The compound according to Item 1, which is a compound selected from: or a pharmacologically acceptable salt thereof.
[項53]
 項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩、並びに(1)ホルモン療法剤、(2)化学療法剤、(3)免疫療法剤、および(4)細胞増殖因子または細胞増殖因子の受容体作用を阻害する薬剤からなる群から選ばれる1以上の薬剤を含有する医薬組成物。
[Section 53]
Item 1-17 or Item 52 or a pharmacologically acceptable salt thereof, and (1) a hormonal therapeutic agent, (2) a chemotherapeutic agent, (3) an immunotherapeutic agent, and ( 4) A pharmaceutical composition comprising one or more drugs selected from the group consisting of cell growth factors or drugs that inhibit cell growth factor receptor action.
[項54]
 項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩を有効成分として含有する、(1)ホルモン療法剤、(2)化学療法剤、(3)免疫療法剤、および(4)細胞増殖因子または細胞増殖因子の受容体作用を阻害する薬剤からなる群から選ばれる1以上の薬剤と併用するための抗癌剤。
[Section 54]
(1) a hormonal therapeutic agent, (2) a chemotherapeutic agent, (3) an immunizing agent comprising the compound according to any one of items 1 to 17 or 52 or a pharmacologically acceptable salt thereof as an active ingredient A therapeutic agent, and (4) an anticancer agent for use in combination with one or more drugs selected from the group consisting of cell growth factors or drugs that inhibit cell growth factor receptor action.
[項55]
 治療が必要な患者に、治療上の有効量の項1~17または項52のいずれか一項に記載の化合物またはその薬理学上許容される塩、並びに(1)ホルモン療法剤、(2)化学療法剤、(3)免疫療法剤、および(4)細胞増殖因子または細胞増殖因子の受容体作用を阻害する薬剤からなる群から選ばれる1以上の薬剤の両方を投与することを含む、癌を治療および/または予防するための方法。
[Section 55]
A therapeutically effective amount of the compound according to any one of Items 1 to 17 or Item 52 or a pharmacologically acceptable salt thereof, and (1) a hormonal therapeutic agent, and (2) Cancer, comprising administering both a chemotherapeutic agent, (3) an immunotherapeutic agent, and (4) a cell growth factor or one or more agents selected from the group consisting of agents that inhibit the receptor action of cell growth factor For the treatment and / or prevention.
 式(1)で表される化合物、またはその薬理学上許容される塩(本発明化合物と称する場合もある。)は、CDK1、CDK2、CDK5、JAK2、AURKAまたはAURKB、或いは他のキナーゼとの相互作用に対して、優れた阻害作用を有することにより、キナーゼが関与する様々な症状、例えば癌疾患等の予防及び治療に適用することができる。また、心毒性や正常細胞に対する毒性が弱いことから、特に治療効果が優れかつ副作用の少ない抗癌剤として、キナーゼが関与する様々な疾患等の予防及び治療に適用することが出来る。 The compound represented by the formula (1), or a pharmacologically acceptable salt thereof (sometimes referred to as the compound of the present invention) may be combined with CDK1, CDK2, CDK5, JAK2, AURKA or AURKB, or other kinases. By having an excellent inhibitory effect on the interaction, it can be applied to the prevention and treatment of various symptoms involving kinases such as cancer diseases. In addition, since cardiotoxicity and toxicity to normal cells are weak, it can be applied to the prevention and treatment of various diseases involving kinases as an anticancer agent having particularly excellent therapeutic effects and few side effects.
実施例89の化合物の、粉末X線回折パターンのチャートである。2 is a chart of a powder X-ray diffraction pattern of the compound of Example 89. 実施例89の化合物の、DSC-TGAのチャートである。2 is a DSC-TGA chart of the compound of Example 89. 実施例90で得た結晶のX線結晶解析により得られた立体構造図である。6 is a three-dimensional structure diagram obtained by X-ray crystal analysis of the crystal obtained in Example 90. FIG. 実施例91の化合物の、粉末X線回折パターンのチャートである。2 is a chart of a powder X-ray diffraction pattern of the compound of Example 91. 実施例91の化合物の、DSC-TGAのチャートである。2 is a DSC-TGA chart of the compound of Example 91. 実施例4で得られた化合物について、試験例7に示す試験を行った腫瘍径の測定結果を表す。縦軸は、腫瘍体積の変化を示す。横軸は、腫瘍移植後の日数を示す。プロットとエラーバーは、それぞれ、腫瘍体積の平均値と標準誤差を示す。#は、有意差検定の結果(p<0.05,Dunnett's test)を示す。About the compound obtained in Example 4, the measurement result of the tumor diameter which performed the test shown in Test example 7 is represented. The vertical axis shows the change in tumor volume. The horizontal axis shows the number of days after tumor transplantation. The plot and error bar show the mean and standard error of the tumor volume, respectively. # Indicates the result of a significant difference test (p <0.05, Dunnett's test). 実施例4で得られた化合物について、試験例7に示す試験を行った体重の測定結果を表す。縦軸は、体重の変化を示す。横軸は、腫瘍移植後の日数を示す。プロットとエラーバーは、それぞれ、体重の平均値と標準誤差を示す。#は、有意差検定の結果(p<0.05,Students t-test)を示す。About the compound obtained in Example 4, the measurement result of the body weight which performed the test shown in Test example 7 is represented. The vertical axis shows the change in body weight. The horizontal axis shows the number of days after tumor transplantation. The plot and error bar show the mean weight value and standard error, respectively. # Indicates the result of a significant difference test (p <0.05, Students t-test). 実施例4で得られた化合物について、試験例8に示す試験を行った腫瘍径の測定結果を表す。縦軸は、腫瘍体積の変化を示す。横軸は、腫瘍移植後の日数を示す。プロットとエラーバーは、それぞれ、腫瘍体積の平均値と標準誤差を示す。#は、有意差検定の結果(p<0.025,Williams' test)を示す。About the compound obtained in Example 4, the measurement result of the tumor diameter which performed the test shown in Test example 8 is represented. The vertical axis shows the change in tumor volume. The horizontal axis shows the number of days after tumor transplantation. The plot and error bar show the mean and standard error of the tumor volume, respectively. # Indicates the result of a significant difference test (p <0.025, Williams' test). 実施例4で得られた化合物について、試験例8に示す試験を行った体重の測定結果を表す。縦軸は、体重の変化を示す。横軸は、腫瘍移植後の日数を示す。プロットとエラーバーは、それぞれ、体重の平均値と標準誤差を示す。#は、有意差検定の結果(p<0.025,Williams' test)を示す。About the compound obtained in Example 4, the measurement result of the body weight which performed the test shown in Test example 8 is represented. The vertical axis shows the change in body weight. The horizontal axis shows the number of days after tumor transplantation. The plot and error bar show the mean weight value and standard error, respectively. # Indicates the result of a significant difference test (p <0.025, Williams' test). 実施例4で得られた化合物について、試験例13に示す試験を行ったNanog発現レベルの結果を示す。併せて対照化合物のPalbociclibとDocetaxelの結果も示す。About the compound obtained in Example 4, the result of the Nanog expression level which performed the test shown in Test example 13 is shown. In addition, the results of the control compounds Palbociclib and Docetaxel are also shown. 実施例72で得られた化合物について、試験例13に示す試験を行ったNanog発現レベルの結果を示す。About the compound obtained in Example 72, the result of the Nanog expression level which performed the test shown in Test example 13 is shown.
 以下に、本発明をさらに詳細に説明する。
 本明細書において、「置換されていてもよい」で定義される基は、無置換および置換されている基であり、基における置換基の数は、明記した場合を除き、置換可能であれば特に制限はなく、1または複数である。また、特に指示した場合を除き、各々の基の説明はその基が他の基の一部分または置換基である場合にも該当する。
The present invention is described in further detail below.
In the present specification, a group defined as “optionally substituted” is an unsubstituted or substituted group, and the number of substituents in the group is the number of substituents unless otherwise specified. There is no restriction | limiting in particular and it is one or more. In addition, unless otherwise specified, the description of each group also applies when the group is a part of another group or a substituent.
 本明細書において、「ハロゲン原子」としては、例えば、フッ素原子、塩素原子、臭素原子、ヨウ素原子等が挙げられる。好ましくはフッ素原子、または塩素原子であり、より好ましくはフッ素原子である。 In the present specification, examples of the “halogen atom” include a fluorine atom, a chlorine atom, a bromine atom, and an iodine atom. Preferably they are a fluorine atom or a chlorine atom, More preferably, it is a fluorine atom.
 「C1-6アルキル」は炭素数1~6の直鎖状もしくは分枝状の飽和炭化水素基を意味する。具体例としては、例えば、メチル、エチル、プロピル、1-メチルエチル、ブチル、2-メチルプロピル、1-メチルプロピル、1,1-ジメチルエチル、ペンチル、3-メチルブチル、2-メチルブチル、2,2-ジメチルプロピル、1-エチルプロピル、1,1-ジメチルプロピル、ヘキシル、4-メチルペンチル、3-メチルペンチル、2-メチルペンチル、1-メチルペンチル等が挙げられる。好ましい「C1-6アルキル」としては、C1-4アルキル等が挙げられ、より好ましくはC1-3アルキルが挙げられる。
 「C1-4アルキル」の具体例としては、「C1-6アルキル」の具体例における炭素原子数が1~4であるメチル、エチル、プロピル、1-メチルエチル、ブチル、2-メチルプロピル、1-メチルプロピル、1,1-ジメチルエチル等が挙げられる。「C1-3アルキル」の具体例としては、「C1-6アルキル」の具体例における炭素原子数1~3であるメチル、エチル、プロピル、1-メチルエチル等が挙げられる。
 なお、本書において、例えば、C1-6とは炭素数が1~6であり、C1-4とは炭素数が1~4であり、C1-3とは炭素数が1~3であり、またはCとは炭素数が6であることを表す。他の数字の場合も同様である。
“C 1-6 alkyl” means a straight or branched saturated hydrocarbon group having 1 to 6 carbon atoms. Specific examples include, for example, methyl, ethyl, propyl, 1-methylethyl, butyl, 2-methylpropyl, 1-methylpropyl, 1,1-dimethylethyl, pentyl, 3-methylbutyl, 2-methylbutyl, 2,2 -Dimethylpropyl, 1-ethylpropyl, 1,1-dimethylpropyl, hexyl, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl and the like. Preferable “C 1-6 alkyl” includes C 1-4 alkyl and the like, more preferably C 1-3 alkyl.
Specific examples of “C 1-4 alkyl” include methyl, ethyl, propyl, 1-methylethyl, butyl, 2-methylpropyl having 1 to 4 carbon atoms in the specific example of “C 1-6 alkyl”. , 1-methylpropyl, 1,1-dimethylethyl and the like. Specific examples of “C 1-3 alkyl” include methyl, ethyl, propyl, 1-methylethyl and the like having 1 to 3 carbon atoms in the specific example of “C 1-6 alkyl”.
In this document, for example, C 1-6 has 1 to 6 carbon atoms, C 1-4 has 1 to 4 carbon atoms, and C 1-3 has 1 to 3 carbon atoms. Yes, or C 6 represents 6 carbon atoms. The same applies to other numbers.
 「C3-10シクロアルキル」とは、炭素原子数が3~10の環状アルキルを意味し、一部架橋された構造のものも含まれる。具体例としては、例えば、シクロプロピル、シクロブチル、シクロペンチル、シクロへキシル、シクロヘプチル、シクロオクチル、シクロノニル、シクロデシル、アダマンチル等が挙げられる。好ましい「C3-10シクロアルキル」としては、C3-8シクロアルキルが挙げられ、より好ましくは、C3-6シクロアルキルが挙げられる。
 「C3-8シクロアルキル」は炭素原子数が3~8の環状アルキルを意味する。具体例としては、「C3-10シクロアルキル」の具体例における炭素原子数が3~8であるシクロプロピル、シクロブチル、シクロペンチル、シクロへキシル、シクロヘプチル、シクロオクチル等が挙げられる。
 「C3-6シクロアルキル」は炭素原子数が3~6の環状アルキルを意味する。具体例としては、「C3-10シクロアルキル」の具体例における炭素原子数3~6であるシクロプロピル、シクロブチル、シクロペンチル、シクロへキシル等が挙げられる。
“C 3-10 cycloalkyl” means a cyclic alkyl having 3 to 10 carbon atoms, and includes a partially crosslinked structure. Specific examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, adamantyl and the like. Preferred “C 3-10 cycloalkyl” includes C 3-8 cycloalkyl, more preferably C 3-6 cycloalkyl.
“C 3-8 cycloalkyl” means a cyclic alkyl of 3 to 8 carbon atoms. Specific examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl having 3 to 8 carbon atoms in the specific example of “C 3-10 cycloalkyl”.
“C 3-6 cycloalkyl” means a cyclic alkyl of 3 to 6 carbon atoms. Specific examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like having 3 to 6 carbon atoms in the specific example of “C 3-10 cycloalkyl”.
 「C1-6アルコキシ」とは「C1-6アルキルオキシ」を意味し、「C1-6アルキル」部分は、前記「C1-6アルキル」と同義である。具体例としては、例えば、メトキシ、エトキシ、プロポキシ、1-メチルエトキシ、ブトキシ、2-メチルプロポキシ、1-メチルプロポキシ、1,1-ジメチルエトキシ、ペンチロキシ基、3-メチルブトキシ、2-メチルブトキシ、2,2-ジメチルプロポキシ、1-エチルプロポキシ、1,1-ジメチルプロポキシ、ヘキシロキシ、4-メチルペンチロキシ、3-メチルペンチロキシ、2-メチルペンチロキシ、1-メチルペンチロキシ、3,3-ジメチルブトキシ、2,2-ジメチルブトキシ、1,1-ジメチルブトキシ、1,2-ジメチルブトキシ等が挙げられる。好ましい「C1-6アルコキシ」としては、C1-4アルコキシが挙げられる。
 「C1-4アルコキシ」の具体例としては、「C1-6アルコキシ」の具体例における炭素原子数が1~4であるメトキシ、エトキシ、プロポキシ、1-メチルエトキシ、ブトキシ等が挙げられる。
“C 1-6 alkoxy” means “C 1-6 alkyloxy”, and the “C 1-6 alkyl” moiety is as defined above for “C 1-6 alkyl”. Specific examples include, for example, methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy, 2-methylpropoxy, 1-methylpropoxy, 1,1-dimethylethoxy, pentyloxy group, 3-methylbutoxy, 2-methylbutoxy, 2,2-dimethylpropoxy, 1-ethylpropoxy, 1,1-dimethylpropoxy, hexyloxy, 4-methylpentyloxy, 3-methylpentyloxy, 2-methylpentyloxy, 1-methylpentyloxy, 3,3-dimethyl Examples include butoxy, 2,2-dimethylbutoxy, 1,1-dimethylbutoxy, 1,2-dimethylbutoxy and the like. Preferred “C 1-6 alkoxy” includes C 1-4 alkoxy.
Specific examples of “C 1-4 alkoxy” include methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy having 1 to 4 carbon atoms in the specific example of “C 1-6 alkoxy”.
 「C1-6アルキルカルボニル」における「C1-6アルキル」部分は、前記の「C1-6アルキル」と同義である。「C1-6アルキルカルボニル」の具体例としては、例えば、メチルカルボニル、エチルカルボニル、プロピルカルボニル、1-メチルエチルカルボニル、ブチルカルボニル、2-メチルプロピルカルボニル、1-メチルプロピルカルボニル、1,1-ジメチルエチルカルボニル等が挙げられる。好ましい「C1-6アルキルカルボニル」としては、C1-4アルキルカルボニルが挙げられる。
 「C1-4アルキルカルボニル」の具体例としては、例えば、メチルカルボニル、エチルカルボニル、プロピルカルボニル、1-メチルエチルカルボニル、ブチルカルボニル、2-メチルプロピルカルボニル等が挙げられる。
"C 1-6 alkyl" moiety in "C 1-6 alkylcarbonyl" is synonymous with "C 1-6 alkyl" of the. Specific examples of “C 1-6 alkylcarbonyl” include, for example, methylcarbonyl, ethylcarbonyl, propylcarbonyl, 1-methylethylcarbonyl, butylcarbonyl, 2-methylpropylcarbonyl, 1-methylpropylcarbonyl, 1,1- And dimethylethylcarbonyl. Preferred “C 1-6 alkylcarbonyl” includes C 1-4 alkylcarbonyl.
Specific examples of “C 1-4 alkylcarbonyl” include, for example, methylcarbonyl, ethylcarbonyl, propylcarbonyl, 1-methylethylcarbonyl, butylcarbonyl, 2-methylpropylcarbonyl and the like.
 「C1-6アルキルアミノ」における「C1-6アルキル」部分は、前記「C1-6アルキル」と同義である。「C1-6アルキルアミノ」の具体例としては、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、tert-ブチルアミノ、ペンチルアミノ、ヘキシルアミノ等が挙げられる。好ましい「C1-6アルキルアミノ」としては「C1-4アルキルアミノ」が挙げられる。
 「C1-4アルキルアミノ」の具体例としては、「C1-6アルキルアミノ」の具体例における炭素原子数が1~4のであるメチルアミノ、エチルアミノ、プロピルアミノ、イソプロピルアミノ、ブチルアミノ等が挙げられる。
"C 1-6 alkyl" moiety in "C 1-6 alkylamino" is the same as defined in the "C 1-6 alkyl". Specific examples of “C 1-6 alkylamino” include, for example, methylamino, ethylamino, propylamino, isopropylamino, butylamino, isobutylamino, tert-butylamino, pentylamino, hexylamino and the like. Preferred “C 1-6 alkylamino” includes “C 1-4 alkylamino”.
Specific examples of the "C 1-4 alkylamino", "C 1-6 alkylamino" methylamino carbon atoms is of 1 to 4 in the specific example of, ethylamino, propylamino, isopropylamino, butylamino, etc. Is mentioned.
 「C2-12ジアルキルアミノ」は、「同一または異なったC1-6アルキルが2つ結合したアミノ」を意味し、該C1-6アルキルは前記と同義である。「C2-12アルキルアミノ」の具体例としては、例えば、ジメチルアミノ、ジエチルアミノ、エチルメチルアミノ、メチルプロピルアミノ、エチルプロピルアミノ、ジプロピルアミノ、イソプロピルメチルアミノ、イソプロピルエチルアミノ、ジイソプロピルアミノ、メチルブチルアミノ、エチルブチルアミノ、ジブチルアミノ、ジイソブチルアミノ、ジtert-ブチルアミノ、ジペンチルアミノ、ジヘキシルアミノ等が挙げられる。好ましい「C2-12ジアルキルアミノ」としては「C2-8ジアルキルアミノ」が挙げられる。
 「C2-8ジアルキルアミノ」の具体例としては、例えば、ジメチルアミノ、ジエチルアミノ、エチルメチルアミノ、メチルプロピルアミノ、エチルプロピルアミノ、ジプロピルアミノ、イソプロピルメチルアミノ、イソプロピルエチルアミノ、ジイソプロピルアミノ、メチルブチルアミノ、エチルブチルアミノ、ジブチルアミノ、ジイソブチルアミノ、ジtert-ブチルアミノ等が挙げられる。
“C 2-12 dialkylamino” means “amino in which two identical or different C 1-6 alkyls are bonded”, and the C 1-6 alkyl is as defined above. Specific examples of “C 2-12 alkylamino” include, for example, dimethylamino, diethylamino, ethylmethylamino, methylpropylamino, ethylpropylamino, dipropylamino, isopropylmethylamino, isopropylethylamino, diisopropylamino, methylbutyl Examples include amino, ethylbutylamino, dibutylamino, diisobutylamino, ditert-butylamino, dipentylamino, dihexylamino and the like. Preferred “C 2-12 dialkylamino” includes “C 2-8 dialkylamino”.
Specific examples of “C 2-8 dialkylamino” include, for example, dimethylamino, diethylamino, ethylmethylamino, methylpropylamino, ethylpropylamino, dipropylamino, isopropylmethylamino, isopropylethylamino, diisopropylamino, methylbutyl Amino, ethylbutylamino, dibutylamino, diisobutylamino, ditert-butylamino and the like can be mentioned.
 「3員~6員の環状アミノ」は、少なくとも一つの窒素原子を含む3員~6員の単環式環状アミノが挙げられる。当該「3員~6員の環状アミノ」は、さらに窒素原子、酸素原子または硫黄原子から選択される同種または異種のヘテロ原子を1個有していても良い。なお、該基の結合手となるのは「3員~6員の環状アミノ」における環を構成する窒素原子である。また、該基には、環の一部が不飽和結合を含む環状アミノも含まれる。「3員~6員の環状アミノ」の具体例としては、例えば、アジリジノ、アゼチジノ、ピロリジノ、イミダゾリジノ、オキサゾリジノ、チアゾリジノ、ピペラジノ、ピペリジノ、モルホリノ、チオモルホリノ、テトラヒドロピリジノ等が挙げられる。好ましい「3員~6員の環状アミノ」としては、「5員~6員の環状アミノ」が挙げられる。
 「5員~6員の環状アミノ」の具体例としては、例えば、ピロリジノ、イミダゾリジノ、オキサゾリジノ、チアゾリジノ、ピペラジノ、ピペリジノ、モルホリノ、チオモルホリノ、テトラヒドロピリジノ等が挙げられる。
“3- to 6-membered cyclic amino” includes 3- to 6-membered monocyclic cyclic amino containing at least one nitrogen atom. The “3- to 6-membered cyclic amino” may further have one same or different heteroatom selected from a nitrogen atom, an oxygen atom or a sulfur atom. The bond of the group is a nitrogen atom constituting the ring in “3- to 6-membered cyclic amino”. The group also includes a cyclic amino in which a part of the ring contains an unsaturated bond. Specific examples of the “3- to 6-membered cyclic amino” include aziridino, azetidino, pyrrolidino, imidazolidino, oxazolidino, thiazolidino, piperazino, piperidino, morpholino, thiomorpholino, tetrahydropyridino and the like. Preferable “3- to 6-membered cyclic amino” includes “5- to 6-membered cyclic amino”.
Specific examples of “5-membered cyclic amino” include pyrrolidino, imidazolidino, oxazolidino, thiazolidino, piperazino, piperidino, morpholino, thiomorpholino, tetrahydropyridino and the like.
 「3員~6員の環状アミノ」および「5員または6員の環状アミノ」は、C3-6シクロアルキル、6員のアリールまたは5員もしくは6員のヘテロアリールと縮合環を形成してもよい。該縮合環の具体例としては、下記で表される「基」等が挙げられる。
Figure JPOXMLDOC01-appb-C000007
“3- to 6-membered cyclic amino” and “5- or 6-membered cyclic amino” form a fused ring with C 3-6 cycloalkyl, 6-membered aryl or 5- or 6-membered heteroaryl. Also good. Specific examples of the condensed ring include “group” represented by the following.
Figure JPOXMLDOC01-appb-C000007
 「3~8員の飽和複素環」とは、炭素原子以外に窒素原子、酸素原子及び硫黄原子からなる群から独立して選択される1~2個の原子を含む3~8個の原子で構成される単環又は2環の飽和複素環を意味する。該基は一部が架橋またはスピロ化されてもよい。また、該基はC6-10アリールまたはC5-10ヘテロアリールが縮環してもよい。なお、該基は、環を構成する窒素原子が、「基」の結合手となることはない。すなわち、該基には、例えば、1-ピロリジノ等の概念は包含されない。具体例としては、例えば、アゼチジン、ピロリジン、ピペリジン、ピペラジン、モルホリン、ホモピペリジン、テトラヒドロフラン、テトラヒドロピラン基等が挙げられる。好ましい「3~8員の飽和複素環」としては、単環の5~6員の飽和複素環基(「5~6員の単環飽和複素環」)が挙げられる。
 「5~6員の単環飽和複素環」の具体例としては、「3~8員の飽和複素環」の具体例における単環の5~6員であるピロリジン、ピペリジン、ピペラジン、モルホリン、テトラヒドロフラン、テトラヒドロピラン等が挙げられる。
“3- to 8-membered saturated heterocyclic ring” means 3 to 8 atoms including 1 to 2 atoms independently selected from the group consisting of a nitrogen atom, an oxygen atom and a sulfur atom in addition to a carbon atom. It means a monocyclic or bicyclic saturated heterocyclic ring. The group may be partially crosslinked or spirolated. The group may be condensed with C 6-10 aryl or C 5-10 heteroaryl. In this group, the nitrogen atom constituting the ring is not a bond of the “group”. That is, the group does not include concepts such as 1-pyrrolidino. Specific examples include azetidine, pyrrolidine, piperidine, piperazine, morpholine, homopiperidine, tetrahydrofuran, tetrahydropyran group and the like. Preferable “3- to 8-membered saturated heterocycle” includes monocyclic 5- to 6-membered saturated heterocyclic groups (“5- to 6-membered monocyclic saturated heterocycle”).
Specific examples of “5- to 6-membered monocyclic saturated heterocycle” include pyrrolidine, piperidine, piperazine, morpholine, tetrahydrofuran, which are monocyclic 5- to 6-membered in the specific example of “3- to 8-membered saturated heterocycle” , Tetrahydropyran and the like.
 「C6-10単環式もしくは多環式のアリール」とは、炭素原子数が6~10の芳香族炭化水素を意味する。具体例としては、例えば、フェニル、1-ナフチル、2-ナフチル基等が挙げられる。好ましくは、フェニルが挙げられる。
 該基はC4-6シクロアルキル、あるいは窒素原子、酸素原子または硫黄原子から選択される同種または異種の原子を1~3個有する5員~6員の複素環基が縮環してもよい。なお、該基の結合手となるのは「C6-10単環式もしくは多環式のアリール」における環を構成する炭素原子である。具体例としては、例えば、下記で表される基等が挙げられる。
Figure JPOXMLDOC01-appb-C000008
“C 6-10 monocyclic or polycyclic aryl” means an aromatic hydrocarbon having 6 to 10 carbon atoms. Specific examples include phenyl, 1-naphthyl, 2-naphthyl group and the like. Preferably, phenyl is used.
The group may be C 4-6 cycloalkyl, or a 5- to 6-membered heterocyclic group having 1 to 3 of the same or different atoms selected from a nitrogen atom, an oxygen atom or a sulfur atom. . The bond of the group is a carbon atom constituting the ring in “C 6-10 monocyclic or polycyclic aryl”. Specific examples include groups represented by the following.
Figure JPOXMLDOC01-appb-C000008
 「5員~10員の単環式もしくは多環式のヘテロアリール」とは、窒素原子、酸素原子及び硫黄原子からなる群から独立して選ばれる1から4個の原子を含む単環の5~6員環の芳香族複素環基もしくは2環の8~10員の芳香族複素環基を意味する。具体例としては、例えば、ピリジル、ピリダジニル、イソチアゾリル、ピロリル、フリル、チエニル、チアゾリル、イミダゾリル、ピリミジニル、チアジアゾリル、ピラゾリル、オキサゾリル、イソオキサゾリル、ピラジニル、トリアジニル、トリアゾリル、イミダゾリジニル、オキサジアゾリル、トリアゾリル、テトラゾリル、インドリル、インダゾリル、キノリル、イソキノリル、ベンゾフラニル、ベンゾチエニル、ベンゾオキサゾリル、ベンゾチアゾリル、ベンズイソオキサゾリル、ベンズイソチアゾリル、ベンゾトリアゾリル、ベンズイミダゾリル、又は6,11-ジヒドロジベンゾ[b,e]チエピニル等が挙げられる。好ましい「5員~10員の単環式もしくは多環式のヘテロアリール」としては、5員~6員の単環式のヘテロアリールが挙げられる。
 5員~6員の単環式のヘテロアリールの具体例としては、例えば、「5員~10員の単環式もしくは多環式のヘテロアリール」の具体例における単環の例示が挙げられる。
 より好ましくは、環内に1つ以上の窒素原子を有する5~6員環の単環式の芳香族複素環が挙げられる。具体例としては、例えば、ピリジル、ピリミジニル等が挙げられ、さらにより好ましくは、ピリジルが挙げられる。
The term “5- to 10-membered monocyclic or polycyclic heteroaryl” refers to monocyclic 5 containing 1 to 4 atoms independently selected from the group consisting of a nitrogen atom, an oxygen atom and a sulfur atom. Means a 6-membered aromatic heterocyclic group or a bicyclic 8- to 10-membered aromatic heterocyclic group. Specific examples include, for example, pyridyl, pyridazinyl, isothiazolyl, pyrrolyl, furyl, thienyl, thiazolyl, imidazolyl, pyrimidinyl, thiadiazolyl, pyrazolyl, oxazolyl, isoxazolyl, pyrazinyl, triazinyl, triazolyl, imidazolidinyl, oxadiazolyl, triazolyl, indrylyl, , Quinolyl, isoquinolyl, benzofuranyl, benzothienyl, benzoxazolyl, benzothiazolyl, benzisoxazolyl, benzisothiazolyl, benzotriazolyl, benzimidazolyl, or 6,11-dihydrodibenzo [b, e] thiepinyl Is mentioned. Preferable “5- to 10-membered monocyclic or polycyclic heteroaryl” includes 5- to 6-membered monocyclic heteroaryl.
Specific examples of the 5- to 6-membered monocyclic heteroaryl include monocyclic examples in the specific examples of “5- to 10-membered monocyclic or polycyclic heteroaryl”.
More preferably, a 5- to 6-membered monocyclic aromatic heterocyclic ring having one or more nitrogen atoms in the ring is used. Specific examples include pyridyl, pyrimidinyl and the like, and more preferably pyridyl.
 本発明の化合物は、一部または全部の原子が同位体元素(例えば、D、H、11C、13C、14C、13N、15N、15O、35S、18F、125I等)で置換されていてもよく、これらの化合物も本発明の化合物に含まれる。 The compounds of the present invention may have some or all atoms of isotopes (eg, D, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 35 S, 18 F, 125 I Etc.), and these compounds are also included in the compounds of the present invention.
 本発明の好ましい態様について、更に説明する。 The preferred embodiment of the present invention will be further described.
 式(1)で表される化合物中の好ましいR1a、R1b、R1c、R1d、R1e、R、R、R、R、X、Y及びArを以下示すが、本発明の技術的範囲はそれらに限定されるものではない。 Preferred R 1a , R 1b , R 1c , R 1d , R 1e , R 2 , R 3 , R 4 , R 5 , X, Y and Ar in the compound represented by the formula (1) are shown below. The technical scope of the invention is not limited thereto.
 式(1)で表される化合物のピラゾール部分は、下記式(1a)及び(1b)のような互変異性体をとることができ、全ての互変異性体は本発明の化合物に含まれる。
Figure JPOXMLDOC01-appb-C000009
The pyrazole moiety of the compound represented by the formula (1) can take tautomers such as the following formulas (1a) and (1b), and all tautomers are included in the compound of the present invention. .
Figure JPOXMLDOC01-appb-C000009
 「X」として好ましくは、窒素原子またはCRが挙げられる。より好ましくは、窒素原子が挙げられる。 “X” is preferably a nitrogen atom or CR 5 . More preferably, a nitrogen atom is mentioned.
 「Y」として好ましくは、水素原子、C1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)が挙げられる。より好ましくは、水素原子が挙げられる。 “Y” is preferably a hydrogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). More preferably, a hydrogen atom is mentioned.
 「R1a」、「R1b」、「R1c」、「R1d」または「R1e」として好ましくは、同一または異なって、水素原子、ハロゲン原子、C1-6アルキル(該基は、フッ素原子およびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)またはC1-6アルコキシ(該基は、1~3個のフッ素原子で置換されていてもよい)が挙げられる。より好ましくは、同一または異なって、水素原子、ハロゲン原子またはC1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)が挙げられる。さらにより好ましくは、同一または異なって、水素原子またはハロゲン原子が挙げられる。 “R 1a ”, “R 1b ”, “R 1c ”, “R 1d ” or “R 1e ” are preferably the same or different and each represents a hydrogen atom, a halogen atom, C 1-6 alkyl (the group is fluorine Optionally substituted with the same or different 1 to 3 groups selected from an atom and C 1-6 alkoxy) or C 1-6 alkoxy (the group is substituted with 1 to 3 fluorine atoms) May be included). More preferably, they are the same or different and include a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). Even more preferably, a hydrogen atom or a halogen atom is the same or different.
 「R」および「R」として好ましくは、同一または異なって、水素原子、ハロゲン原子またはC1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)が挙げられる。 “R 2 ” and “R 3 ” are preferably the same or different and each represents a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). Can be mentioned.
 「R」として好ましくは、水素原子、C1-6アルキル(1~3個のフッ素原子で置換されていてもよい)またはC3-10シクロアルキル(1~4個のフッ素原子で置換されていてもよい)が挙げられる。より好ましくは、C1-6アルキル(1~3個のフッ素原子で置換されていてもよい)およびC3-6シクロアルキル(1~4個のフッ素原子で置換されていてもよい)が挙げられる。さらにより好ましくは、C1-3アルキル(1~3個のフッ素原子で置換されていてもよい)およびシクロプロピルが挙げられる。 “R 4 ” is preferably a hydrogen atom, C 1-6 alkyl (optionally substituted with 1 to 3 fluorine atoms) or C 3-10 cycloalkyl (substituted with 1 to 4 fluorine atoms). May be included). More preferable examples include C 1-6 alkyl (which may be substituted with 1 to 3 fluorine atoms) and C 3-6 cycloalkyl (which may be substituted with 1 to 4 fluorine atoms). It is done. Even more preferred are C 1-3 alkyl ( optionally substituted with 1 to 3 fluorine atoms) and cyclopropyl.
 「R」として好ましくは、水素原子、ハロゲン原子、C1-6アルキル(該基は、1~3個のフッ素原子、またはC1-6アルコキシで置換されていてもよい)が挙げられる。より好ましくは、水素原子、C1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)が挙げられる。さらにより好ましく水素原子が挙げられる。 “R 5 ” is preferably a hydrogen atom, a halogen atom, or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms or C 1-6 alkoxy). More preferable examples include a hydrogen atom and C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). Even more preferred is a hydrogen atom.
「Ar」として好ましくは、フェニルまたはピリジルが挙げられる。より好ましくは、フェニルが挙げられる。 “Ar” is preferably phenyl or pyridyl. More preferably, phenyl is mentioned.
 「薬理学上許容される塩」としては、酸付加塩及び塩基付加塩が挙げられる。例えば、酸付加塩としては、塩酸塩、臭化水素酸塩、硫酸塩、ヨウ化水素酸塩、硝酸塩、リン酸塩等の無機酸塩、またはクエン酸塩、シュウ酸塩、酢酸塩、ギ酸塩、プロピオン酸塩、安息香酸塩、トリフルオロ酢酸塩、メタンスルホン酸塩、ベンゼンスルホン酸塩、p-トルエンスルホン酸塩、カンファースルホン酸塩等の有機酸塩が挙げられ、塩基付加塩としては、ナトリウム塩、カリウム塩、カルシウム塩、マグネシウム塩、アンモニウム塩等の無機塩基塩、またはトリエチルアンモニウム塩、トリエタノールアンモニウム塩、ピリジニウム塩、ジイソプロピルアンモニウム塩等の有機塩基塩等が挙げられ、さらにはアルギニン、アスパラギン酸、グルタミン酸等の塩基性または酸性アミノ酸といったアミノ酸塩が挙げられる。 “Pharmaceutically acceptable salts” include acid addition salts and base addition salts. For example, acid addition salts include inorganic acid salts such as hydrochloride, hydrobromide, sulfate, hydroiodide, nitrate, phosphate, or citrate, oxalate, acetate, formic acid Salt, propionate, benzoate, trifluoroacetate, methanesulfonate, benzenesulfonate, p-toluenesulfonate, camphorsulfonate, and other organic acid salts. Inorganic base salts such as sodium salt, potassium salt, calcium salt, magnesium salt, ammonium salt, or organic base salts such as triethylammonium salt, triethanolammonium salt, pyridinium salt, diisopropylammonium salt, and the like, and arginine And amino acid salts such as basic or acidic amino acids such as aspartic acid and glutamic acid.
 本発明化合物を塩として取得したいとき、本発明化合物が塩の形で得られる場合には、そのまま精製すればよく、また、本発明化合物が遊離の形で得られる場合には、適当な有機溶媒に溶解もしくは懸濁させ、酸又は塩基を加えて通常の方法により塩を形成させればよい。
 本発明化合物は、水あるいは各種溶媒との付加物(水和物、溶媒和物)の形で存在することもあるが、これらの付加物も本発明に包含される。さらに、本発明は、本発明化合物のあらゆる互変異性体およびあらゆる態様の結晶形のものも包含する。
When it is desired to obtain the compound of the present invention as a salt, it can be purified as it is when the compound of the present invention is obtained in a salt form. When the compound of the present invention is obtained in a free form, an appropriate organic solvent can be used. It may be dissolved or suspended in the solution, and an acid or base may be added to form a salt by a conventional method.
The compound of the present invention may exist in the form of an adduct (hydrate, solvate) with water or various solvents, and these adducts are also included in the present invention. Furthermore, the present invention also includes all tautomers of the compounds of the present invention and crystal forms of all embodiments.
 本発明化合物は、式(1)で表される化合物のプロドラッグ、またはその薬理学上許容される塩も含まれる。また、これらの水和物、エタノール溶媒和物等の溶媒和物も含まれる。 The compound of the present invention includes a prodrug of the compound represented by the formula (1) or a pharmacologically acceptable salt thereof. Moreover, solvates, such as these hydrates and ethanol solvates, are also included.
 本明細書における「式(1)で表される化合物のプロドラッグ」なる用語は、生体内における生理条件下で酵素や胃酸等による反応により式(1)で表される化合物に変換される化合物、例えば、酵素的に酸化、還元、加水分解等されて式(1)で表される化合物に変換される化合物を意味する。 In the present specification, the term “prodrug of the compound represented by formula (1)” is a compound that is converted into a compound represented by formula (1) by a reaction with an enzyme, gastric acid or the like under physiological conditions in vivo. For example, it means a compound that is enzymatically oxidized, reduced, hydrolyzed, etc. and converted to a compound represented by formula (1).
 本発明化合物は、分子内に1または複数の不斉炭素原子を有する化合物も包含する。従って、本発明化合物は光学異性体、ラセミ体、ジアステレオマー等も包含する。また、本発明化合物は、シス-トランス異性体、軸不斉による異性体等も包含する。以上のように、本発明化合物は、存在するあらゆる異性体およびその混合物も包含する。 The compound of the present invention includes compounds having one or more asymmetric carbon atoms in the molecule. Accordingly, the compound of the present invention includes optical isomers, racemates, diastereomers and the like. The compounds of the present invention also include cis-trans isomers, isomers resulting from axial asymmetry, and the like. As described above, the compound of the present invention includes all isomers and mixtures thereof.
 本発明化合物が阻害活性を示すキナーゼとしては、種々のチロシンキナーゼ、種々のセリンキナーゼ、種々のスレオニンキナーゼが挙げられる。好ましくは、CDK1,CDK2、CDK5、AURKA、AURKB、JAK2等が挙げられる。 Examples of the kinase in which the compound of the present invention exhibits inhibitory activity include various tyrosine kinases, various serine kinases, and various threonine kinases. CDK1, CDK2, CDK5, AURKA, AURKB, JAK2 and the like are preferable.
 以下に、式(1)で表される化合物、またはその薬理学上許容される塩の製造方法について、例を挙げて説明するが、これに限定されるものではない。 Hereinafter, the method for producing the compound represented by the formula (1) or a pharmacologically acceptable salt thereof will be described by way of example, but the present invention is not limited thereto.
 本発明化合物は、公知化合物から、例えば以下の製造法およびそれに準じた方法、あるいは当業者に周知の合成方法を適宜組み合わせて製造することができる。 The compound of the present invention can be produced from a known compound by appropriately combining, for example, the following production method and a method analogous thereto, or a synthesis method well known to those skilled in the art.
製造法:
 式(1)で表される化合物は、例えば、次の方法により合成することができる。
Figure JPOXMLDOC01-appb-C000010
[式中LGおよびLG’は、それぞれ同一または異なって、脱離基を意味し、例えば、ハロゲン原子、メタンスルホニルオキシ、p-トルエンスルホン酸オキシまたはトリフルオロメタンスルホン酸オキシ、フェノキシ、トリフルオロフェノキシ、テトラフルオロフェノキシ、ペンタフルオロフェノキシ、ニトロフェノキシ等が挙げられる。他の定義は、前記項1に記載と同義である。]
Manufacturing method:
The compound represented by the formula (1) can be synthesized, for example, by the following method.
Figure JPOXMLDOC01-appb-C000010
[Wherein LG and LG ′ are the same or different and each represents a leaving group such as a halogen atom, methanesulfonyloxy, p-toluenesulfonic acid oxy or trifluoromethanesulfonic acid oxy, phenoxy, trifluorophenoxy, Tetrafluorophenoxy, pentafluorophenoxy, nitrophenoxy and the like can be mentioned. The other definitions are the same as those described in item 1. ]
式(A-3)で表される化合物の製法1
工程1:
工程1-1:
 Xが窒素原子である式(A-2)の化合物を必要に応じて塩基の存在下に、式(A-1)の化合物と反応させることにより式(A-3)の化合物を合成することができる。塩基としては特に限定はないが、例えば、トリエチルアミン、ジイソプロピルエチルアミン、トリブチルアミン、1.5-ジアザビシクロ[4.3.0]ノナ-5-エン(DBN)、1,8-ジアザビシクロ[5.4.0]ウンデカ-7-エン(DBU)、ピリジン、ジメチルアミノピリジン、ピコリン、N-メチルモルホリン(NMM)等の有機塩基類、または、炭酸水素ナトリウム、炭酸水素カリウム、炭酸ナトリウム、炭酸カリウム、水酸化ナトリウム、水酸化カリウム等の無機塩基類等が挙げられる。
 溶媒は、本工程の反応条件で反応しない溶媒であればよい。具体的には、例えばメタノール、エタノール、2-プロパノール(イソプロピルアルコール)、t-ブタノール等のアルコール系溶媒、例えば、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、メチルシクロペンチルエーテル、アニソール、1,4-ジオキサン等のエーテル系溶媒、ベンゼン、トルエン、クロロベンゼン、キシレン等の芳香族炭化水素系溶媒、酢酸エチル、酢酸メチル等のエステル系溶媒、N,N-ジメチルホルムアミド、N,N-ジメチルアセトアミド、N-メチル-2-ピロリジノン、1,3-ジメチル-2-イミダゾリジノン、ジメチルスルホキシド等の非プロトン系溶媒または水、あるいはそれらの混合物が挙げられる。
 反応温度は、-80℃から加熱還流下で行われ、通常25℃から90℃である。反応時間は、通常30分間から12時間である。
Production method 1 of compound represented by formula (A-3)
Step 1:
Step 1-1:
Synthesizing a compound of formula (A-3) by reacting a compound of formula (A-2) wherein X is a nitrogen atom with a compound of formula (A-1) in the presence of a base, if necessary. Can do. The base is not particularly limited. For example, triethylamine, diisopropylethylamine, tributylamine, 1.5-diazabicyclo [4.3.0] non-5-ene (DBN), 1,8-diazabicyclo [5.4. 0] Organic bases such as undec-7-ene (DBU), pyridine, dimethylaminopyridine, picoline, N-methylmorpholine (NMM), or sodium bicarbonate, potassium bicarbonate, sodium carbonate, potassium carbonate, hydroxide Examples include inorganic bases such as sodium and potassium hydroxide.
The solvent should just be a solvent which does not react on the reaction conditions of this process. Specifically, for example, alcohol solvents such as methanol, ethanol, 2-propanol (isopropyl alcohol), t-butanol, such as diethyl ether, diisopropyl ether, tetrahydrofuran, methylcyclopentyl ether, anisole, 1,4-dioxane and the like. Ether solvents, aromatic hydrocarbon solvents such as benzene, toluene, chlorobenzene and xylene, ester solvents such as ethyl acetate and methyl acetate, N, N-dimethylformamide, N, N-dimethylacetamide, N-methyl-2 An aprotic solvent such as pyrrolidinone, 1,3-dimethyl-2-imidazolidinone, dimethyl sulfoxide, water, or a mixture thereof.
The reaction temperature is from −80 ° C. under heating under reflux, and is usually from 25 ° C. to 90 ° C. The reaction time is usually 30 minutes to 12 hours.
工程1-2:
 Xが炭素原子である式(A-2)の化合物を、必要に応じて金属触媒の存在下、式(A-1)の化合物とUlmann型条件(例えば、DMF等の非プロトン性溶媒中、酢酸銅(II)等の金属触媒を用いて、加熱還流する)や、Buchwald型条件(例えば、炭酸セシウム等の炭酸塩基下、BINAP、Pd(dba)またはPd(OAc)のようなパラジウム触媒と、dppf、Xantphos等のようなリガンドを用いて、トルエン等の不活性溶媒中、加熱還流等する)等の反応条件を用いて、反応させることにより式(A-3)の化合物を合成することができる。
 溶媒としては、本工程の反応条件で反応しない溶媒であればよい。具体的には、例えば、テトラヒドロフランや1,4-ジオキサン等のエーテル系溶媒等があげられる。
 反応温度は、-80℃から加熱還流下であり、通常25℃から150℃で行われる。反応時間は、通常30分間から12時間である。
Step 1-2:
A compound of the formula (A-2) in which X is a carbon atom is optionally combined with a compound of the formula (A-1) in Ulman type conditions (for example, in an aprotic solvent such as DMF, in the presence of a metal catalyst, Using a metal catalyst such as copper (II) acetate, or refluxing under Buchwald type conditions (for example, BINAP, Pd 2 (dba) 3 or Pd (OAc) 2 under a carbonate base such as cesium carbonate) The compound of the formula (A-3) is reacted by reacting using a palladium catalyst and a ligand such as dppf, Xantphos, etc. under reaction conditions such as heating and refluxing in an inert solvent such as toluene. Can be synthesized.
As a solvent, what is necessary is just a solvent which does not react on the reaction conditions of this process. Specific examples include ether solvents such as tetrahydrofuran and 1,4-dioxane.
The reaction temperature is from −80 ° C. to heating under reflux, usually from 25 ° C. to 150 ° C. The reaction time is usually 30 minutes to 12 hours.
式(A-3)で表される化合物の製法2
工程2(式(A-5)で表される化合物の製法):
 Xが窒素原子である式(A-4)の化合物を必要に応じて塩基の存在下に、式(A-1)の化合物と反応させることにより式(A-5)の化合物を合成することができる。塩基としては特に限定はないが、例えば、トリエチルアミン、ジイソプロピルエチルアミン、トリブチルアミン、1.5-ジアザビシクロ[4.3.0]ノナ-5-エン(DBN)、1,8-ジアザビシクロ[5.4.0]ウンデカ-7-エン(DBU)、ピリジン、ジメチルアミノピリジン、ピコリン、N-メチルモルホリン(NMM)等の有機塩基類、または、炭酸水素ナトリウム、炭酸水素カリウム、炭酸ナトリウム、炭酸カリウム、水酸化ナトリウム、水酸化カリウム等の無機塩基類等が挙げられる。
 溶媒は、本工程の反応条件で反応しない溶媒であればよい。具体的には、例えばメタノール、エタノール、2-プロパノール、t-ブタノール等のアルコール系溶媒、例えば、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、メチルシクロペンチルエーテル、アニソール、1,4-ジオキサン等のエーテル系溶媒、ベンゼン、トルエン、クロロベンゼン、キシレン等の芳香族炭化水素系溶媒、酢酸エチル、酢酸メチル等のエステル系溶媒、N,N-ジメチルホルムアミド、N,N-ジメチルアセトアミド、N-メチル-2-ピロリジノン、1,3-ジメチル-2-イミダゾリジノン、ジメチルスルホキシド等の非プロトン系溶媒または水、あるいはそれらの混合物が挙げられる。
 反応温度は、-80℃から加熱還流下で行われ、通常25℃から90℃である。反応時間は、通常30分間から12時間である。
Production method 2 of compound represented by formula (A-3)
Step 2 (Production method of the compound represented by the formula (A-5)):
Synthesizing a compound of formula (A-5) by reacting a compound of formula (A-4) wherein X is a nitrogen atom with a compound of formula (A-1) in the presence of a base, if necessary. Can do. The base is not particularly limited. For example, triethylamine, diisopropylethylamine, tributylamine, 1.5-diazabicyclo [4.3.0] non-5-ene (DBN), 1,8-diazabicyclo [5.4. 0] Organic bases such as undec-7-ene (DBU), pyridine, dimethylaminopyridine, picoline, N-methylmorpholine (NMM), or sodium bicarbonate, potassium bicarbonate, sodium carbonate, potassium carbonate, hydroxide Examples include inorganic bases such as sodium and potassium hydroxide.
The solvent should just be a solvent which does not react on the reaction conditions of this process. Specifically, alcohol solvents such as methanol, ethanol, 2-propanol and t-butanol, for example, ether solvents such as diethyl ether, diisopropyl ether, tetrahydrofuran, methylcyclopentyl ether, anisole and 1,4-dioxane, Aromatic hydrocarbon solvents such as benzene, toluene, chlorobenzene and xylene, ester solvents such as ethyl acetate and methyl acetate, N, N-dimethylformamide, N, N-dimethylacetamide, N-methyl-2-pyrrolidinone, 1 , 3-dimethyl-2-imidazolidinone, aprotic solvents such as dimethyl sulfoxide, water, or a mixture thereof.
The reaction temperature is from −80 ° C. under heating under reflux, and is usually from 25 ° C. to 90 ° C. The reaction time is usually 30 minutes to 12 hours.
工程3(式(A-3)で表される化合物の製法):
 Xが窒素原子である式(A-5)の化合物を、必要に応じて塩基存在下、シアン化物イオンと反応させることにより式(A-3)の化合物を合成することができる。
 塩基としては特に限定はないが、例えば、トリエチルアミン、ジイソプロピルエチルアミン、トリブチルアミン、1.5-ジアザビシクロ[4.3.0]ノナ-5-エン(DBN)、1,4-ジアザビシクロ[2.2.2]オクタン(DABCO)、1,8-ジアザビシクロ[5.4.0]ウンデカ-7-エン(DBU)、ピリジン、ジメチルアミノピリジン、ピコリン、N-メチルモルホリン(NMM)等の有機塩基類が挙げられる。
 溶媒は、本工程の反応条件で反応しない溶媒であればよい。具体的には、例えばメタノール、エタノール、2-プロパノール、t-ブタノール等のアルコール系溶媒、例えば、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、メチルシクロペンチルエーテル、アニソール、1,4-ジオキサン等のエーテル系溶媒、ベンゼン、トルエン、クロロベンゼン、キシレン等の芳香族炭化水素系溶媒、酢酸エチル、酢酸メチル等のエステル系溶媒、N,N-ジメチルホルムアミド、N,N-ジメチルアセトアミド、N-メチル-2-ピロリジノン、1,3-ジメチル-2-イミダゾリジノン、ジメチルスルホキシド等の非プロトン系溶媒または水、あるいはそれらの混合物が挙げられる。
 反応温度は、通常25℃から150℃である。反応時間は、通常30分間から12時間である。
Step 3 (Production method of compound represented by formula (A-3)):
A compound of the formula (A-3) can be synthesized by reacting a compound of the formula (A-5) in which X is a nitrogen atom with a cyanide ion in the presence of a base, if necessary.
The base is not particularly limited. For example, triethylamine, diisopropylethylamine, tributylamine, 1.5-diazabicyclo [4.3.0] non-5-ene (DBN), 1,4-diazabicyclo [2.2. 2] Organic bases such as octane (DABCO), 1,8-diazabicyclo [5.4.0] undec-7-ene (DBU), pyridine, dimethylaminopyridine, picoline, N-methylmorpholine (NMM) It is done.
The solvent should just be a solvent which does not react on the reaction conditions of this process. Specifically, alcohol solvents such as methanol, ethanol, 2-propanol and t-butanol, for example, ether solvents such as diethyl ether, diisopropyl ether, tetrahydrofuran, methylcyclopentyl ether, anisole and 1,4-dioxane, Aromatic hydrocarbon solvents such as benzene, toluene, chlorobenzene and xylene, ester solvents such as ethyl acetate and methyl acetate, N, N-dimethylformamide, N, N-dimethylacetamide, N-methyl-2-pyrrolidinone, 1 , 3-dimethyl-2-imidazolidinone, aprotic solvents such as dimethyl sulfoxide, water, or a mixture thereof.
The reaction temperature is usually from 25 ° C to 150 ° C. The reaction time is usually 30 minutes to 12 hours.
式(A-6)で表される化合物の製法
工程4:
 Xが炭素原子または窒素原子である式(A-3)の化合物を不活性溶媒中、例えば置換アリールGrignard試薬等の有機金属種と反応させることにより式(A-6)の化合物を合成することができる。
 不活性溶媒は、本工程の反応条件で反応しない溶媒であればよい。具体的には、例えば、テトラヒドロフラン、ジエチルエーテル等のエーテル系溶媒等が挙げられる。
 反応温度は、-80℃から加熱還流下で行われ、通常-10℃から25℃である。反応時間は、通常30分間から24時間である。
Method for producing compound represented by formula (A-6)
Step 4:
Synthesizing a compound of the formula (A-6) by reacting a compound of the formula (A-3) in which X is a carbon atom or a nitrogen atom with an organometallic species such as a substituted aryl Grignard reagent in an inert solvent. Can do.
The inert solvent should just be a solvent which does not react on the reaction conditions of this process. Specific examples include ether solvents such as tetrahydrofuran and diethyl ether.
The reaction temperature is from −80 ° C. to heating under reflux, usually from −10 ° C. to 25 ° C. The reaction time is usually 30 minutes to 24 hours.
式(1)で表される化合物の製法
工程5:
工程5-1:
 Xが炭素原子または窒素原子である式(A-6)の化合物を還元剤、アルキルGrignard試薬、アルキル化試薬等と反応させることにより式(1)で表される化合物を合成することができる。
 還元剤としては、水素化ホウ素ナトリウム、水素化リチウムアルミニウム、水素化ジイソブチルアルミニウム等が挙げられる。アルキルGrignard試薬としては、メチルマグネシウムブロミド、イソプロピルマグネシウムブロミド等が挙げられる。アルキル化試薬としては、トリフルオロメチルトリメチルシラン等が挙げられる。
 溶媒は、本工程の反応条件で反応しない溶媒であればよい。具体的には、例えば、還元剤使用時はメタノールやエタノール等のアルコール系溶媒が挙げられ、アルキルGrignard試薬使用時はテトラヒドロフランやジエチルエーテル等のエーテル系溶媒が挙げられ、アルキル化試薬使用時はジメチルアセトアミドやN-メチル-2-ピロリジノン等の非プロトン性溶媒が挙げられる。
 反応温度は、-80℃から加熱還流下、通常-10℃から50℃である。反応時間は、通常10分間から48時間である。
Method for producing compound represented by formula (1)
Step 5:
Step 5-1
A compound represented by the formula (1) can be synthesized by reacting a compound of the formula (A-6) in which X is a carbon atom or a nitrogen atom with a reducing agent, an alkyl Grignard reagent, an alkylating reagent or the like.
Examples of the reducing agent include sodium borohydride, lithium aluminum hydride, diisobutylaluminum hydride and the like. Examples of the alkyl Grignard reagent include methyl magnesium bromide and isopropyl magnesium bromide. Examples of the alkylating reagent include trifluoromethyltrimethylsilane.
The solvent should just be a solvent which does not react on the reaction conditions of this process. Specifically, for example, an alcohol solvent such as methanol or ethanol is used when a reducing agent is used, an ether solvent such as tetrahydrofuran or diethyl ether is used when an alkyl Grignard reagent is used, and dimethyl is used when an alkylating reagent is used. Examples include aprotic solvents such as acetamide and N-methyl-2-pyrrolidinone.
The reaction temperature is usually from −10 ° C. to 50 ° C. under heating to reflux from −80 ° C. The reaction time is usually 10 minutes to 48 hours.
工程5-2:
 Xが炭素原子または窒素原子である式(A-6)の化合物を触媒存在下、配位子存在下または非存在下、塩基存在下または非存在下、水素と反応させることにより式(1)で表される化合物を合成することができる。
 触媒としては、パラジウムやニッケル等の金属、塩化ルテニウムや酢酸パラジウム等の遷移金属の塩、酢酸ルテニウムのBINAP錯体等の錯体、(R)-RUCYTM-Xylbinap、(S)-RUCYTM-Xylbinap等の不正触媒等が挙げられる。
 配位子としてはジフェニルホスフィノエタン等のリン系配位子の他、BIAP等の不斉配位子も使用できる。
 塩基としては、tert-ブトキシカリウム、tert-ブトキシナトリウム、ナトリウムエトキシド、ナトリウムメトキシド等が挙げられる。
 水素源としては、水素ガスの他にギ酸アンモニウム等が挙げられる。
 溶媒としては、本工程の反応条件で反応しない溶媒であればよい。具体的には、例えば、イソプロピルアルコール、メタノール、エタノール等のアルコール系溶媒、テトラヒドロフラン、ジエチルエーテル等のエーテル系溶媒が挙げられる。
 反応温度は、-80℃から加熱還流下、通常-10℃から50℃である。反応時間は、通常10分間から48時間である。
Step 5-2:
A compound of the formula (A-6) in which X is a carbon atom or a nitrogen atom is reacted with hydrogen in the presence of a catalyst, in the presence or absence of a ligand, in the presence or absence of a base, to give a compound of the formula (1) Can be synthesized.
Catalysts include metals such as palladium and nickel, salts of transition metals such as ruthenium chloride and palladium acetate, complexes such as the BINAP complex of ruthenium acetate, (R) -RUCY TM -Xylbinap, (S) -RUCY TM -Xylbinap, etc. And illegal catalysts.
As the ligand, in addition to a phosphorus-based ligand such as diphenylphosphinoethane, an asymmetric ligand such as BIAP can also be used.
Examples of the base include tert-butoxy potassium, tert-butoxy sodium, sodium ethoxide, sodium methoxide and the like.
Examples of the hydrogen source include ammonium formate in addition to hydrogen gas.
As a solvent, what is necessary is just a solvent which does not react on the reaction conditions of this process. Specific examples include alcohol solvents such as isopropyl alcohol, methanol and ethanol, and ether solvents such as tetrahydrofuran and diethyl ether.
The reaction temperature is usually from −10 ° C. to 50 ° C. under heating to reflux from −80 ° C. The reaction time is usually 10 minutes to 48 hours.
 上記において説明した製造法の各反応において、具体的に保護基の使用を明示した場合以外でも、反応点以外の何れかの官能基が説明した反応条件下で変化する、もしくは、説明した方法を実施するのに不適切な場合には、反応点以外を必要に応じて保護し、反応終了後または一連の反応を行った後に脱保護することにより目的化合物を得ることができる。
 保護基としては、文献(例えば、Protective Groups in Organic Synthesis, 3rd ed., T.W.Greene,John Wiley & Sons Inc.(1999)等)に記載されているような通常の保護基を用いることができ、更に具体的には、アミノの保護基としては、例えばベンジルオキシカルボニル、tert-ブトキシカルボニル、アセチル、ベンジル等を、またヒドロキシの保護基としては、例えばトリメチルシリル、tert-ブチルジメチルシリル等のトリアルキルシリル、アセチルまたはベンジル等を、それぞれ挙げることができる。
 保護基の導入および脱離は、有機合成化学で常用される方法(例えば前述のProtective Groups in Organic Synthesis参照)またはそれらに準じた方法により行うことができる。
In each reaction of the production method described above, any functional group other than the reactive site changes under the described reaction conditions, even when the use of a protecting group is not specifically stated, or the described method When it is inappropriate to carry out, the compound other than the reaction point is protected as necessary, and the target compound can be obtained by deprotection after completion of the reaction or after a series of reactions.
As the protecting group, an ordinary protecting group described in literature (for example, Protective Groups in Organic Synthesis, 3rd ed., TWGreene, John Wiley & Sons Inc. (1999)) can be used. Specifically, examples of the amino protecting group include benzyloxycarbonyl, tert-butoxycarbonyl, acetyl, benzyl and the like, and examples of the hydroxy protecting group include trialkylsilyl such as trimethylsilyl, tert-butyldimethylsilyl, Acetyl or benzyl can be mentioned respectively.
Introduction and elimination of protecting groups can be performed by methods commonly used in organic synthetic chemistry (see, for example, the above-mentioned Protective Groups in Organic Synthesis) or methods based thereon.
 本明細書を通じて、保護基、縮合剤等は、この技術分野において慣用されているIUPAC-IUB(生化学命名委員会)による略号で表わすことがある。
 出発化合物および目的化合物の好適な塩および医薬として許容しうる塩は、慣用の無毒性塩であり、それらとしては、有機酸塩(例えば酢酸塩、トリフルオロ酢酸塩、マレイン酸塩、フマル酸塩、クエン酸塩、酒石酸塩、メタンスルホン酸塩、ベンゼンスルホン酸塩、蟻酸塩、トルエンスルホン酸塩等)および無機酸塩(例えば塩酸塩、臭化水素酸塩、ヨウ化水素酸塩、硫酸塩、硝酸塩、燐酸塩等)のような酸付加塩、アミノ酸(例えばアルギニン、アスパラギン酸、グルタミン酸等)との塩、アルカリ金属塩(例えばナトリウム塩、カリウム塩等)、アルカリ土類金属塩(例えばカルシウム塩、マグネシウム塩等)等の金属塩、アンモニウム塩、有機塩基塩(例えばトリメチルアミン塩、トリエチルアミン塩、ピリジン塩、ピコリン塩、ジシクロヘキシルアミン塩、N,N’-ジベンジルエチレンジアミン塩等)等を挙げることができる。
Throughout this specification, protecting groups, condensing agents and the like may be represented by abbreviations by IUPAC-IUB (Biochemical Nomenclature Board) commonly used in this technical field.
Suitable and pharmaceutically acceptable salts of the starting and target compounds are conventional non-toxic salts, such as organic acid salts (eg acetate, trifluoroacetate, maleate, fumarate) Citrate, tartrate, methanesulfonate, benzenesulfonate, formate, toluenesulfonate, etc.) and inorganic acid salts (eg hydrochloride, hydrobromide, hydroiodide, sulfate) Acid addition salts such as nitrates, phosphates, etc., salts with amino acids (eg arginine, aspartic acid, glutamic acid etc.), alkali metal salts (eg sodium salts, potassium salts etc.), alkaline earth metal salts (eg calcium Metal salts such as salts and magnesium salts), ammonium salts, organic base salts (for example, trimethylamine salt, triethylamine salt, pyridine salt, picoline salt, dioxygen salt) Black hexylamine salt, N, N'- dibenzylethylenediamine salt, etc.) and the like.
 上記製造方法における中間体または最終生成物は、その官能基を適宜変換すること(例えば、必要に応じて官能基の保護、脱保護を行い、アミノ基、水酸基、カルボニル基、ハロゲン基等を足がかりに種々の変換をおこなうこと)によって、本発明に含まれる別の化合物へ導く事もできる。官能基の変換は、通常行われる一般的方法(例えば、Comprehensive Organic Transformations, R. C. Larock, John Wiley & Sons Inc.(1999)等を参照)によって行うことができる。 The intermediate or final product in the above production method may be converted as appropriate by its functional group (for example, protecting and deprotecting the functional group as necessary to establish an amino group, hydroxyl group, carbonyl group, halogen group, etc.) Can be converted into other compounds included in the present invention. The functional group can be converted by a commonly used general method (for example, see Comprehensive Organic Transformations, R. C. Larock, John Wiley & Sons Inc. (1999)).
 上記各製造法における中間体および目的化合物は、有機合成化学で常用される精製法、例えば中和、濾過、抽出、洗浄、乾燥、濃縮、再結晶、各種クロマトグラフィー等に付して単離精製することができる。また、中間体においては、特に精製することなく次の反応に供することも可能である。 The intermediates and target compounds in each of the above production methods are isolated and purified by purification methods commonly used in synthetic organic chemistry such as neutralization, filtration, extraction, washing, drying, concentration, recrystallization, and various chromatography. can do. In addition, the intermediate can be subjected to the next reaction without any particular purification.
 本発明化合物(1)の中には、光学活性中心に基づく光学異性体、分子内回転の束縛により生じた軸性または面性キラリティーに基づくアトロプ異性体、その他の立体異性体、互変異性体、および幾何異性体等が存在し得るものがあるが、これらを含め、全ての可能な異性体およびそれらの混合物は本発明の範囲に包含される。 Among the compounds (1) of the present invention, there are optical isomers based on optically active centers, atropisomers based on axial or planar chirality generated by restraining intramolecular rotation, other stereoisomers, tautomerism Isomers, geometric isomers and the like may exist, but all possible isomers and mixtures thereof are included in the scope of the present invention.
 特に光学異性体やアトロプ異性体は、ラセミ体として得ることも出来るし、あるいは光学活性の出発原料や中間体を用い、光学活性体として得ることもできる。また、前記製造法の適切な段階で、対応する原料、中間体または最終品のラセミ体を、光学活性カラムを用いた方法、分別結晶化法等の公知の分離方法によって、物理的にまたは化学的にそれらの光学対掌体に分割することができる。例えば、ラセミ体を光学活性分割剤と反応させ、2種のジアステレオマーを合成し、物理的性質が異なることを利用し分別結晶化等の方法によって分割することができる(ジアステレオマー法)。 In particular, optical isomers and atropisomers can be obtained as racemates, or can be obtained as optically active substances using optically active starting materials and intermediates. Further, at an appropriate stage of the production method, the corresponding raw material, intermediate or final racemate is physically or chemically separated by a known separation method such as a method using an optically active column or a fractional crystallization method. Can be divided into their optical enantiomers. For example, a racemate can be reacted with an optically active resolving agent to synthesize two diastereomers, which can be separated by a method such as fractional crystallization utilizing the different physical properties (diastereomeric method). .
 本発明化合物の薬理学上許容される塩を取得したい時は、式(1)で表される化合物が薬理学上許容される塩の形で得られる場合には、そのまま精製すればよく、また、遊離の形で得られる場合には、適当な有機溶媒に溶解もしくは懸濁させ、酸または塩基を加えて通常の方法により塩を形成させればよい。 When it is desired to obtain a pharmacologically acceptable salt of the compound of the present invention, if the compound represented by the formula (1) is obtained in the form of a pharmacologically acceptable salt, it may be purified as it is, When it is obtained in a free form, it may be dissolved or suspended in an appropriate organic solvent, and an acid or base may be added to form a salt by a conventional method.
 以上説明した各々の製造法における原料、中間体のうち、特にあらためてその製造法を記載しなかったものについては、市販化合物であるか、または市販化合物から当業者に公知の方法、もしくはそれに準じた方法によって合成することができる。 Among the raw materials and intermediates in each of the production methods described above, those that have not been described in particular are commercially available compounds, or methods known to those skilled in the art from commercially available compounds, or equivalents thereto. It can be synthesized by the method.
 本発明化合物は、例えば抗癌剤として提供され、その適応される癌種は問わないが、具体例としては例えば乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、前立腺癌、脳腫瘍、子宮内膜癌、膵臓癌、胃癌、骨肉腫、骨髄腫、または血液癌を挙げることができる。この中で好ましい癌種としては、乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、前立腺癌、脳腫瘍、または子宮内膜癌を挙げることができ、より好ましい癌種としては、乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、または前立腺癌を挙げることができる。
 本発明化合物を抗癌剤として適応する場合の別の好ましい態様としては、適応される癌種として、血液癌、骨髄腫、肝癌、骨肉腫、皮膚癌、上皮性細胞癌(midline carcinoma)、乳癌、肺癌、卵巣癌、子宮癌、大腸癌、前立腺癌または咽頭癌を挙げることができる。この中で好ましい癌種としては、血液癌、骨髄腫、肝癌、乳癌、肺癌、卵巣癌、子宮癌、大腸癌、前立腺癌または咽頭癌を挙げることができ、より好ましい癌種としては、乳癌、肺癌、卵巣癌、子宮癌、大腸癌、前立腺癌または咽頭癌を挙げることができる。
 本発明において「血液癌」とは、リンパ腫、白血病を含む概念である。
 本発明において「抗癌剤」とは、癌を予防および/または治療する目的で投与した際、癌腫を縮小若しくは消滅させるか又は癌腫を増大させない効果を有するものである。
 本発明において、「予防」とは、疾患を発症していない健常人に対して本発明の有効成分を投与する行為であり、例えば、疾患の発症を防止することを目的とするものである。「治療」とは、医師により疾患を発症していると診断をされた人(患者)に対して本発明の有効成分を投与する行為であり、例えば、疾患や症状を軽減すること、癌腫を増大させないこと又は疾患発症前の状態に戻すことを目的とするものである。また、投与の目的が疾患や症状の悪化防止又は癌腫の増大防止の場合も、治療行為に含まれる。
The compound of the present invention is provided as, for example, an anticancer agent, and the cancer type to which it is applied is not limited, but specific examples include, for example, breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor And endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or hematological cancer. Among these, preferable cancer types include breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor, or endometrial cancer, and more preferable cancer types are , Breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, or prostate cancer.
In another preferred embodiment when the compound of the present invention is applied as an anticancer agent, the cancer types to be applied are blood cancer, myeloma, liver cancer, osteosarcoma, skin cancer, epithelial cell carcinoma, breast cancer, lung cancer. Ovarian cancer, uterine cancer, colon cancer, prostate cancer or pharyngeal cancer. Among these, preferable cancer types include blood cancer, myeloma, liver cancer, breast cancer, lung cancer, ovarian cancer, uterine cancer, colon cancer, prostate cancer or pharyngeal cancer, and more preferable cancer types include breast cancer, Mention may be made of lung cancer, ovarian cancer, uterine cancer, colon cancer, prostate cancer or pharyngeal cancer.
In the present invention, “blood cancer” is a concept including lymphoma and leukemia.
In the present invention, the “anticancer agent” has an effect of reducing or eliminating a carcinoma or not increasing the tumor when administered for the purpose of preventing and / or treating cancer.
In the present invention, “prevention” is an act of administering the active ingredient of the present invention to a healthy person who has not developed a disease, and is intended to prevent, for example, the onset of the disease. “Treatment” is the act of administering the active ingredient of the present invention to a person (patient) diagnosed as having developed a disease by a doctor. For example, alleviating a disease or symptom, It is intended not to increase or to return to the state before the onset of the disease. In addition, the therapeutic purpose also includes cases where the purpose of administration is prevention of worsening of diseases and symptoms or prevention of increase in carcinoma.
 本発明化合物は、治療に使用する場合に、医薬組成物として、経口的または非経口的(例えば、静脈内、皮下または筋肉内注射、局所的、経直腸的、経皮的あるいは経鼻的)に投与することができる。経口投与のための組成物としては、例えば、錠剤、カプセル剤、丸剤、顆粒剤、散剤、液剤、懸濁剤等が挙げられる。これらの製剤は、従来公知の技術を用いて調整され、製剤分野において通常使用される無毒性かつ不活性な担体もしくは賦形剤を含有することができる。 The compounds of the present invention are used orally or parenterally (for example, intravenous, subcutaneous or intramuscular injection, topical, rectal, transdermal or nasal) as pharmaceutical compositions when used for treatment. Can be administered. Examples of the composition for oral administration include tablets, capsules, pills, granules, powders, liquids, suspensions and the like. These preparations are prepared using conventionally known techniques, and can contain non-toxic and inert carriers or excipients usually used in the field of preparation.
 本発明化合物を投与する場合、その使用量は、症状、年齢、投与方法等によって異なるが、例えば、経口投与の場合には、成人に対して、1日当たり、下限として、0.01mg(好ましくは1mg)、上限として、5000mg(好ましくは500mg)を、1回または数回に分けて、症状に応じて投与することが望ましい。静脈内注射の場合には、成人に対して、1日当たり、下限として、0.01mg(好ましくは0.1mg)、上限として、1000mg(好ましくは30mg)を、1回または数回に分けて、症状に応じて投与することにより効果が期待される。また、患者の症状により経口および静脈内注射において、間欠投与することも好ましい態様である。 When the compound of the present invention is administered, the amount to be used varies depending on symptoms, age, administration method, etc. For example, in the case of oral administration, the lower limit is 0.01 mg per day for adults (preferably 1 mg), and as an upper limit, 5000 mg (preferably 500 mg) is desirably administered once or several times according to symptoms. In the case of intravenous injection, for adults, 0.01 mg (preferably 0.1 mg) as the lower limit and 1000 mg (preferably 30 mg) as the upper limit per day, divided into one or several times, The effect is expected by administering according to the symptoms. In addition, intermittent administration is also preferable in oral and intravenous injection depending on the patient's symptoms.
 本発明化合物は、その効果の増強および/または副作用の軽減を目的として、他の薬剤と併用して用いることができる。具体的には、本発明化合物は、ホルモン療法剤、化学療法剤、免疫療法剤、細胞増殖因子または細胞増殖因子の受容体作用を阻害する薬剤等の薬剤と併用して用いることができる。以下、本発明化合物と併用し得る薬剤を併用薬剤と略記する。 The compound of the present invention can be used in combination with other drugs for the purpose of enhancing its effect and / or reducing side effects. Specifically, the compound of the present invention can be used in combination with a drug such as a hormonal therapeutic agent, a chemotherapeutic agent, an immunotherapeutic agent, a cell growth factor or a drug that inhibits cell growth factor receptor action. Hereinafter, a drug that can be used in combination with the compound of the present invention is abbreviated as a concomitant drug.
 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(例えば、クエン酸タモキシフェン、クエン酸トレミフェン等)、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(例えば、酢酸ゴセレリン、ブセレリン、リュープロレリン等)、ドロロキシフェン、エピチオスタノール、スルホン酸エチニルエストラジオール、アロマターゼ阻害薬(例えば、塩酸ファドロゾール、アナストロゾール、レトロゾール、エキセメスタン、ボロゾール、フォルメスタン等)、抗アンドロゲン(例えば、フルタミド、ビカルタミド、ニルタミド等)、副腎皮質ホルモン系薬剤(例えば、デキサメタゾン、プレドニゾロン、ベタメタゾン、トリアムシノロン等)、アンドロゲン合成阻害薬(例えば、アビラテロン等)、レチノイドおよびレチノイドの代謝を遅らせる薬剤(例えば、リアロゾール等)等が挙げられる。 Examples of hormone therapeutic agents include phosfestol, diethylstilbestrol, chlorotrianicene, medroxyprogesterone acetate, megestrol acetate, chlormadinone acetate, cyproterone acetate, danazol, allylestrenol, gestrinone, mepartricin, raloxifene, Olmeroxifene, levormeloxifene, antiestrogens (eg, tamoxifen citrate, toremifene citrate, etc.), pill formulations, mepithiostan, testrolactone, aminoglutethimide, LH-RH agonists (eg, goserelin acetate, buserelin, Leuprorelin, etc.), droloxifene, epithiostanol, ethinyl estradiol sulfonate, aromatase inhibitors (eg, fadrozole hydrochloride, anastrozo) , Letrozole, exemestane, borozole, formestane, etc.), antiandrogens (eg, flutamide, bicalutamide, nilutamide, etc.), corticosteroids (eg, dexamethasone, prednisolone, betamethasone, triamcinolone, etc.), androgen synthesis inhibitors (eg, For example, abiraterone etc.), retinoids and drugs that delay the metabolism of retinoids (eg riarosol etc.) and the like.
 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。代表的な例を次に記載する。 As the chemotherapeutic agent, for example, alkylating agents, antimetabolites, anticancer antibiotics, plant-derived anticancer agents and the like are used. A typical example is described below.
 アルキル化剤としては、例えば、ナイトロジェンマスタード、塩酸ナイトロジェンマスタード-N-オキシド、クロラムブチル、シクロフォスファミド、イホスファミド、チオテパ、カルボコン、トシル酸インプロスルファン、ブスルファン、塩酸ニムスチン、ミトブロニトール、メルファラン、ダカルバジン、ラニムスチン、リン酸エストラムスチンナトリウム、トリエチレンメラミン、カルムスチン、ロムスチン、ストレプトゾジン、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、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、ビゼレシン及びそれらのDDS製剤等が挙げられる。 Examples of the alkylating agent include nitrogen mustard, nitrogen mustard hydrochloride-N-oxide, chlorambutyl, cyclophosphamide, ifosfamide, thiotepa, carbocon, improsulfan tosylate, busulfan, nimustine hydrochloride, mitoblonitol, melphalan, Dacarbazine, ranimustine, estramustine phosphate sodium, triethylenemelamine, carmustine, lomustine, streptozidine, pipobrommann, etoglucid, carboplatin, cisplatin, dibrospium hydrochloride, fotemustine, prednimustine, pumitepa, ribomuthine, temozolomide, treosorphan, Trophosphamide, dinostatin stimamarer, adzeresin, systemustin, vizeresin and their D S formulation, and the like.
 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、エオシタビン、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(例えば、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、カペシタビン等)、アミノプテリン、ネルザラビン、ロイコポリンカルシウム、タブロイド、ブトシン、フォリネイトカルシウム、レボフォリネイトカルシウム、クラドリビン、エミテフール、フルダラビン、ゲムシタビン、ヒドロキシカルパミド、ペントスタチン、ピリトレキシム、イドキシウリジン、ミトグアゾン、チアゾフリン、アンバムスチン、ベンダムスチンおよびそれらのDDS製剤等が挙げられる。 Examples of the antimetabolite include mercaptopurine, 6-mercaptopurine riboside, thioinosine, methotrexate, pemetrexed, eocitabine, cytarabine, cytarabine okphosphat, ancitabine hydrochloride, 5-FU drugs (for example, fluorouracil, tegafur, UFT, Doxyfluridine, carmofur, galocitabine, emiteful, capecitabine, etc.), aminopterin, nerzarabine, leucoporin calcium, tabloid, butosine, folinate calcium, levofolinate calcium, cladribine, emitefur, fludarabine, gemcitabine, hydroxycarpamide treptostatine, pentostatin , Idoxyuridine, mitoguazone, thiazofurin, ambamustine, bendamustine and those DDS formulation, and the like.
 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製剤等が挙げられる。 Anticancer antibiotics include, for example, actinomycin D, actinomycin C, mitomycin C, chromomycin A3, bleomycin hydrochloride, bleomycin sulfate, peplomycin sulfate, daunorubicin hydrochloride, doxorubicin hydrochloride, aclarubicin hydrochloride, pirarubicin hydrochloride, epirubicin hydrochloride, neo Examples include cartinostatin, myramicin, sarcomycin, carcinophylline, mitotane, zorubicin hydrochloride, mitoxantrone hydrochloride, idarubicin hydrochloride and their DDS preparations.
 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、ドセタキセル、ビノレルビンおよびそれらのDDS製剤等が挙げられる。 Examples of plant-derived anticancer agents include etoposide, etoposide phosphate, vinblastine sulfate, vincristine sulfate, vindesine sulfate, teniposide, paclitaxel, docetaxel, vinorelbine, and their DDS preparations.
 免疫療法剤としては、例えば、ピシバニール、クレスチン、シゾフィラン、レンチナン、ウベニメクス、インターフェロン、インターロイキン、マクロファージコロニー刺激因子、顆粒球コロニー刺激因子、エリスロポイエチン、リンホトキシン、BCGワクチン、コリネバクテリウムパルブム、レバミゾール、ポリサッカライドK、プロコダゾール、抗CTLA4抗体、PD-1抗体、Toll-like Receptors作動薬(例えば、TLR7作動薬、TLR8作動薬、TLR9作動薬等)が挙げられる。 Examples of immunotherapeutic agents include picibanil, krestin, schizophyllan, lentinan, ubenimex, interferon, interleukin, macrophage colony stimulating factor, granulocyte colony stimulating factor, erythropoietin, lymphotoxin, BCG vaccine, corynebacterium parvum, levamisole , Polysaccharide K, procodazole, anti-CTLA4 antibody, PD-1 antibody, Toll-like Receptors agonist (for example, TLR7 agonist, TLR8 agonist, TLR9 agonist, etc.).
 細胞増殖因子および細胞増殖因子の受容体の作用を阻害する薬剤における細胞増殖因子としては、細胞増殖を促進する物質であれば、どのようなものでもよく、通常、分子量が20,000以下のペプチドで、受容体との結合により低濃度で作用が発揮される因子があげられる。具体的には、EGF(epidermal growth factor)またはそれと実質的に同一の活性を有する物質(例えば、TGFalpha等)、インスリンまたはそれと実質的に同一の活性を有する物質(例えば、インスリン、IGF(insulin-like growth factor)-1、IGF-2等)、FGF(fibroblast growth factor)またはそれと実質的に同一のアッセイを有する物質(例えば、酸性FGF、塩基性FGF、KGK(keratinocyte growth factor)、FGF-10等)および、その他の細胞増殖因子(例えば、CSF(colony stimukating factor)、EPO(erythropoietin)、IL-2(interleukin-2)、NGF(nerve growth factor)、PDGF(platelet-derived growth factor)、TGF-beta(transforming growth factor beta)、HGF(hepatocyte growth factor)、VEGF(vascular endothelial growth factor)、へレグリン、アンジオポエチン等)が挙げられる。 The cell growth factor in the drug that inhibits the action of the cell growth factor and the receptor for the cell growth factor may be any substance as long as it is a substance that promotes cell growth, and usually has a molecular weight of 20,000 or less. Thus, a factor that exerts its action at a low concentration by binding to a receptor can be mentioned. Specifically, EGF (epidermal grow factor) or a substance having substantially the same activity (for example, TGFalpha), insulin or a substance having substantially the same activity (for example, insulin, IGF (insulin-insulin- like growth factor) -1, IGF-2, etc.), FGF (fibroblast growth factor) or a substance having substantially the same assay as it (for example, acidic FGF, basic FGF, KGK (keratinocyte growth factor), FGF-10) And other cell growth factors (for example, CSF (colony (stimukating factor), EPO (erythropoietin), IL-2 (interleukin-2), NGF (nerve growth factor), PDGF (platelet-derived growth factor), TGF) -Beta (transforming growth factor beta), HGF (hepatocyte growth factor) VEGF (vascular endothelial growth factor), heregulin, angiopoietin, etc.).
 本発明化合物および併用薬剤の投与期間は限定されず、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。また、本発明化合物と併用薬剤の合剤としてもよい。併用薬剤の投与量は、臨床上用いられている用量を基準として適宜選択することができる。また、本発明化合物と併用薬剤の配合比は、投与対象、投与ルート、対象疾患、症状、組み合わせ等により適宜選択することができる。例えば投与対象がヒトである場合、本発明化合物1重量部に対し、併用薬剤を0.01~100重量部用いればよい。また、その副作用抑制の目的として、制吐剤、睡眠導入剤、抗痙攣薬等の薬剤(これらも併用薬剤に包含される)と組み合わせて用いることができる。 The administration period of the compound of the present invention and the concomitant drug is not limited, and these may be administered simultaneously to the administration subject or may be administered with a time difference. Moreover, it is good also as a mixture of this invention compound and a concomitant drug. The dose of the concomitant drug can be appropriately selected based on the clinically used dose. The compounding ratio of the compound of the present invention and the concomitant drug can be appropriately selected depending on the administration subject, administration route, target disease, symptom, combination and the like. For example, when the administration subject is a human, the concomitant drug may be used in an amount of 0.01 to 100 parts by weight per 1 part by weight of the compound of the present invention. In addition, for the purpose of suppressing the side effects, it can be used in combination with drugs such as antiemetics, sleep inducers, anticonvulsants and the like (these are also included in the concomitant drugs).
 本発明化合物は、本明細書中の試験例等で示されるとおり、CDK1、CDK2、AURKA、AURKB、JAK2、CDK5等から選ばれるキナーゼに対して高い阻害作用を有し、キナーゼが関与する様々な症状、例えば癌疾患等の予防及び治療に適用することができる。特に、本発明化合物のうち好ましいものは、CDK5を阻害することにより、ステムネス遺伝子の発現抑制作用を示す(ステムネス遺伝子については後述する)。本発明化合物以外のCDK5を阻害する物質(例えば、CDK5を直接に阻害することができる、CDK5のsiRNA等)を用いることにより、CDK5阻害剤によるステムネス遺伝子の発現抑制作用を確認することができる。また、CDK5の発現をなくした、もしくは発現を抑制した細胞(CDK5ノックダウン、CDK5ドミナントネガティブ等)におけるステムネス遺伝子の発現を確認することによっても、CDK5阻害とステムネス遺伝子の発現抑制との関連性を確認することができる。 The compound of the present invention has a high inhibitory action on a kinase selected from CDK1, CDK2, AURKA, AURKB, JAK2, CDK5, etc., as shown in the test examples and the like in this specification, and various kinases involved. It can be applied to the prevention and treatment of symptoms such as cancer diseases. In particular, among the compounds of the present invention, preferred compounds exhibit a stemness gene expression suppressing action by inhibiting CDK5 (the stemness gene will be described later). By using a substance that inhibits CDK5 other than the compound of the present invention (for example, siRNA of CDK5 that can directly inhibit CDK5), the suppressive action of stemness gene expression by the CDK5 inhibitor can be confirmed. Further, by confirming the expression of the stemness gene in cells (CDK5 knockdown, CDK5 dominant negative, etc.) in which the expression of CDK5 is eliminated or suppressed, the relationship between CDK5 inhibition and stemness gene expression suppression is also confirmed. Can be confirmed.
 ステムネス遺伝子とは、自己複製および/または異なった細胞への分化を誘導することができる遺伝子である。ステムネス遺伝子の例としては、例えばNanog、Sox2、β-cateninおよびOct4などが挙げられる。本発明化合物のうち好ましいものは、ステムネス遺伝子の発現抑制作用を有する(試験例11)。また、本発明化合物のうち好ましいものは、スフェア形成阻害作用を示し、それゆえ、がん幹細胞の増殖抑制作用が期待できる。
 これらの作用により、本発明化合物のうち好ましいものは、癌の再発予防作用を併せ持つ抗癌剤としても有用性が期待できる。
 また、本発明化合物のうち好ましいものは、化学療法剤による治療抵抗性を有する癌の治療剤としても有用である。ここでの化学療法剤として好ましくは、例えばタキサン系抗癌剤を挙げることができる。本明細書におけるタキサン系抗癌剤としては、タキサン骨格を有する抗癌剤を挙げることができ、例えばパクリタキセル、ドセタキセルなどが挙げられる。
A stemness gene is a gene that can induce self-replication and / or differentiation into different cells. Examples of stemness genes include Nanog, Sox2, β-catenin, Oct4, and the like. Among the compounds of the present invention, a preferred compound has an action of suppressing the expression of stemness gene (Test Example 11). Among the compounds of the present invention, preferred compounds exhibit a sphere formation inhibitory action, and therefore can be expected to suppress the growth of cancer stem cells.
Due to these effects, the preferred compounds of the present invention can be expected to be useful as anticancer agents having an effect of preventing cancer recurrence.
Among the compounds of the present invention, preferred compounds are also useful as therapeutic agents for cancer having resistance to treatment with chemotherapeutic agents. Preferable chemotherapeutic agents here include, for example, taxane anticancer agents. Examples of the taxane anticancer agent in the present specification include an anticancer agent having a taxane skeleton, and examples thereof include paclitaxel and docetaxel.
 近年、GPCRと腫瘍形成の関係が明らかとなってきている
(G protein-coupled receptors: novel targets for drug discovery in cancer. Lappano R, Maggiolini M. Nat Rev Drug Discov. 2011 Jan;10(1):47-60、
Differential effect of adenosine receptors on growth of human colon cancer HCT 116 and HT-29 cell lines. Sakowicz-Burkiewicz M, Kitowska A, Grden M, Maciejewska I, Szutowicz A, Pawelczyk T. Arch Biochem Biophys. 2013 May;533(1-2):47-54、
The adenosinergic system in cancer: Key therapeutic target. Sorrentino R, Pinto A, Morello S. Oncoimmunology. 2013 Jan 1;2(1):e22448 参照)。
 本発明化合物は、GPCRに対し、作動性、及び拮抗性が弱い化合物であり、GPCRへの影響およびGPCRを介した腫瘍形成への影響は小さいと考えられる。
In recent years, the relationship between GPCR and tumorigenesis has been clarified (G protein-coupled receptors: novel targets for drug discovery in cancer. Lappano R, Maggiolini M. Nat Rev Drug Discov. 2011 Jan; 10 (1): 47 -60,
Differential effect of adenosine receptors on growth of human colon cancer HCT 116 and HT-29 cell lines.Sakowicz-Burkiewicz M, Kitowska A, Grden M, Maciejewska I, Szutowicz A, Pawelczyk T. Arch Biochem Biophys. 2013 May; 533 (1 -2): 47-54,
The adenosinergic system in cancer: Key therapeutic target. See Sorrentino R, Pinto A, Morello S. Oncoimmunology. 2013 Jan 1; 2 (1): e22448).
The compound of the present invention is a compound that is weak in agonistic and antagonistic properties to GPCR, and is considered to have little influence on GPCR and on tumor formation via GPCR.
 以下に本発明を、参考例、実施例および試験例により、さらに具体的に説明するが、本発明はもとよりこれに限定されるものではない。尚、以下の参考例及び実施例において示された化合物名は、必ずしもIUPAC命名法に従うものではない。 Hereinafter, the present invention will be described more specifically with reference examples, examples and test examples. However, the present invention is not limited to these examples. In addition, the compound names shown in the following Reference Examples and Examples do not necessarily follow the IUPAC nomenclature.
 本明細書において、以下の略語を使用することがある。
THF:テトラヒドロフラン
TFA:トリフルオロ酢酸
NaBH(OAc):トリアセトキシ水素化ホウ素ナトリウム
DMAP:N,N-ジメチル-4-アミノピリジン
(Boc)O:ジ-tert-ブチルジカーボネート
DMF:N,N-ジメチルホルムアミド
DIEA:N-エチルジイソプロピルアミン
WSCI:1-(3-ジメチルアミノプロピル)-3-エチルカルボジイミド
WSCI・HCl: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
HOBt:1-ヒドロキシベンゾトリアゾール
HOBt・HO:1-ヒドロキシベンゾトリアゾール1水和物
Me:メチル
Et:エチル
DMA:N,N-ジメチルアセトアミド
NMP:1-メチル-2-ピロリジノン
Boc:tert-ブトキシカルボニル
CbzまたはZ:ベンジルオキシカルボニル
(R)-RUCYTM-XylBINAP:RuCl[(R)-daipena][(R)-xylbinap]、CAS番号:1384974-38-2
(S)-RUCYTM-XylBINAP:RuCl[(S)-daipena][(S)-xylbinap]、CAS番号:1312713-39-5
N:規定(例として2N HClは2規定塩酸を示す。)
M:モル濃度(mol/L)(例として2Mメチルアミンは2mol/Lメチルアミン溶液を示す。)
:保持時間
obs MS[M+1]:観測された分子の質量
In this specification, the following abbreviations may be used.
THF: Tetrahydrofuran TFA: Trifluoroacetic acid NaBH (OAc) 3 : Sodium triacetoxyborohydride DMAP: N, N-dimethyl-4-aminopyridine (Boc) 2 O: Di-tert-butyldicarbonate DMF: N, N -Dimethylformamide DIEA: N-ethyldiisopropylamine WSCI: 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide WSCI · HCl: 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride HOBt: 1- Hydroxybenzotriazole HOBt · H 2 O: 1-hydroxybenzotriazole monohydrate Me: methyl Et: ethyl DMA: N, N-dimethylacetamide NMP: 1-methyl-2-pyrrolidinone Boc: tert-butoxycarbo Nyl Cbz or Z: benzyloxycarbonyl (R) -RUCY -XylBINAP: RuCl [(R) -daipena] [(R) -xylbinap], CAS number: 1384974-38-2
(S) -RUCY -XylBINAP: RuCl [(S) -daipena] [(S) -xylbinap], CAS number: 1312713-39-5
N: Normal (for example, 2N HCl indicates 2N hydrochloric acid)
M: Molar concentration (mol / L) (2M methylamine as an example indicates a 2 mol / L methylamine solution)
t R : Retention time obs MS [M + 1]: Observed molecular mass
 逆相HPLC分取精製は以下のように実施した。
 精製はGilson HPLC Systemを用いて行った。カラムはYMC CombiPrep ODS-A column(5μm,50×20 mm I.D.)を使用し、溶媒はCHCN(0.035 % TFA含有)、水(0.05 % TFA含有)の混合溶媒系を用いた。UV検出は210 nm,220 nm,254 nmの各波長で行った。
 溶出の条件は以下の通り。
分取装置:Gilson HPLC System
カラム:YMC CombiPrep ODS-A 5μm,50×20 mm I.D.
溶媒:CH3CN(containing 0.035 % TFA)、water(containing 0.05 % TFA)
流速:35 mL/min
グラジェント:linear gradient from 1:99(v/v)CH3CN/water to 95:5(v/v)CH3CN/water within 13 min at 35 mL/min
Reversed phase HPLC preparative purification was performed as follows.
Purification was performed using a Gilson HPLC System. The column used was a YMC CombiPrep ODS-A column (5 μm, 50 × 20 mm ID), and the solvent was a mixed solvent system of CH 3 CN (containing 0.035% TFA) and water (containing 0.05% TFA). UV detection was performed at wavelengths of 210 nm, 220 nm, and 254 nm.
The elution conditions are as follows.
Preparative equipment: Gilson HPLC System
Column: YMC CombiPrep ODS-A 5μm, 50 × 20 mm ID
Solvent: CH 3 CN (containing 0.035% TFA), water (containing 0.05% TFA)
Flow rate: 35 mL / min
Gradient: linear gradient from 1:99 (v / v) CH 3 CN / water to 95: 5 (v / v) CH 3 CN / water within 13 min at 35 mL / min
 粉末X線回折測定は、次の条件で行った。
・装置:X'pert-MPD(スペクトリス社製)
・X線:Cu Kα1/45 kV/40 mA
・入射スリット:15 mm(オート)/発散防止スリット:15 mm(オート)
・試料板:無反射Si板
・ステップサイズ:0.017°
・走査範囲:4-40°(2θ)
・積算時間:100秒/ステップ
 なお、本明細書に記載した回折角2θ(°)における回折ピーク値は、測定機器により、もしくは測定条件等により多少の測定誤差を生じることがある。具体的には、測定誤差は±0.2、好ましくは±0.1の範囲内であってもよい。
The powder X-ray diffraction measurement was performed under the following conditions.
・ Device: X'pert-MPD (Spectris)
· X-ray: Cu Kα 1/45 kV / 40 mA
-Incident slit: 15 mm (automatic) / Divergence prevention slit: 15 mm (automatic)
・ Sample plate: Non-reflective Si plate ・ Step size: 0.017 °
・ Scanning range: 4-40 ° (2θ)
Accumulation time: 100 seconds / step Note that the diffraction peak value at the diffraction angle 2θ (°) described in this specification may cause some measurement error depending on the measurement equipment or measurement conditions. Specifically, the measurement error may be within a range of ± 0.2, preferably ± 0.1.
 示差走査熱量(DSC)測定は、次の条件で行った。
・装置:DSCQ1000(TAインスツルメント社製)
・測定温度範囲:10~250℃
・昇温速度: 10℃/分
・容器: アルミニウムハーメチックパン(Pin hole)
・雰囲気ガス流量: 乾燥窒素、約50 mL/分
Differential scanning calorimetry (DSC) measurement was performed under the following conditions.
・ Equipment: DSCQ1000 (TA Instruments)
・ Measurement temperature range: 10-250 ℃
・ Temperature increase rate: 10 ℃ / min ・ Vessel: Aluminum hermetic pan (Pin hole)
・ Atmospheric gas flow: Dry nitrogen, approx. 50 mL / min
 熱重量測定(TGA)は、次の条件で行った。
・装置:TGAQ500(TAインスツルメント社製)
・測定温度範囲: 室温~250℃
・昇温速度: 10℃/分
・容器: プラチナパン
・雰囲気ガス流量: 乾燥窒素、サンプル流量約60 mL/分、バランス流量約40 mL/分
Thermogravimetry (TGA) was performed under the following conditions.
・ Device: TGAQ500 (TA Instruments)
・ Measurement temperature range: Room temperature to 250 ℃
・ Temperature increase rate: 10 ℃ / min ・ Vessel: Platinum pan ・ Atmospheric gas flow rate: Dry nitrogen, sample flow rate approx. 60 mL / min, balance flow rate approx. 40 mL / min
X線回折測定は、次の条件で行った。
・X線回折装置:R-AXIS  RAPID-R リガク
(制御ソフトウェア:RAPID  AUTO Ver.3.11 リガク、
 解析ソフトウェア:Crystal Structure Ver.4.01  リガク、Mercury Ver.3.1 Development (Build RC5) CCDC)
・測定条件(X線:CuKα線、出力:50kV-100mA、温度:93±1K)
X-ray diffraction measurement was performed under the following conditions.
-X-ray diffractometer: R-AXIS RAPID-R Rigaku (Control software: RAPID AUTO Ver.3.11 Rigaku,
Analysis software: Crystal Structure Ver.4.01 Rigaku, Mercury Ver.3.1 Development (Build RC5) CCDC)
・ Measurement conditions (X-ray: CuKα ray, output: 50kV-100mA, temperature: 93 ± 1K)
 化合物同定のLC/MS分析条件は以下の通りである。
LC/MS 測定法:
検出機器:ACQUITY(登録商標)SQ deteceter(Waters社)
HPLC:ACQUITY UPLC(登録商標)system
Column:Waters ACQUITY UPLC(登録商標)BEH C18(1.7μm,2.1 mm×30 mm)
Solvent:A液:0.06 % ギ酸/H2O,B液:0.06 % ギ酸/MeCN
Gradient condition:0.0-1.3 min Linear gradient from B 2 % to 96 %
Flow rate:0.8 mL/min
UV:220 nm and 254 nm
The LC / MS analysis conditions for compound identification are as follows.
LC / MS measurement method:
Detector: ACQUITY (registered trademark) SQ deteceter (Waters)
HPLC: ACQUITY UPLC (registered trademark) system
Column: Waters ACQUITY UPLC (registered trademark) BEH C18 (1.7 μm, 2.1 mm × 30 mm)
Solvent: A solution: 0.06% formic acid / H 2 O, B solution: 0.06% formic acid / MeCN
Gradient condition: 0.0-1.3 min Linear gradient from B 2% to 96%
Flow rate: 0.8 mL / min
UV: 220 nm and 254 nm
参考例1:2-クロロ-N-(5-メチル-1H-ピラゾール-3-イル)ピリミジン-4-アミン
Figure JPOXMLDOC01-appb-C000011
 5-メチル-ピラゾール-3-アミン(18.0g)のエタノール(500mL)溶液に2,4-ジクロロピリミジン(25.0g)およびN-エチル-ジイソプロピルアミン(30.6mL)を加え、70℃にて9時間攪拌した。室温まで放冷した後、析出した固体をろ過し、エタノールとヘキサンで洗浄した後、減圧乾燥して表題化合物(14.9g)を得た。
1H-NMR(300 MHz, DMSO-d6)δ12.1 (s, 1H), 10.3 (s, 1H), 8.14 (d, 1H), 5.76 (br-s, 1H), 2.21 (s, 3H).
Reference Example 1: 2-Chloro-N- (5-methyl-1H-pyrazol-3-yl) pyrimidin-4-amine
Figure JPOXMLDOC01-appb-C000011
To a solution of 5-methyl-pyrazol-3-amine (18.0 g) in ethanol (500 mL) was added 2,4-dichloropyrimidine (25.0 g) and N-ethyl-diisopropylamine (30.6 mL), and the mixture was heated to 70 ° C. And stirred for 9 hours. After allowing to cool to room temperature, the precipitated solid was filtered, washed with ethanol and hexane, and dried under reduced pressure to give the title compound (14.9 g).
1 H-NMR (300 MHz, DMSO-d 6 ) δ12.1 (s, 1H), 10.3 (s, 1H), 8.14 (d, 1H), 5.76 (br-s, 1H), 2.21 (s, 3H ).
参考例2:6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-カルボニトリル
Figure JPOXMLDOC01-appb-C000012
 参考例1で得た化合物(4.8g)のDMSO(100mL)溶液にシアン化ナトリウム(2.24g)および1,4-ジアザビシクロ[2.2.2]オクタン(1.28g)を加え、120℃にて3時間攪拌した。室温に冷却後、反応液に水を加え、不溶物をろ過により除去した。得られたろ液に酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して表題化合物(3.67g)を得た。
1H-NMR(300 MHz, DMSO-d6)δ12.20 (s, 1H), 10.48 (s, 1H), 8.35 (s, 1H), 2.22 (s, 3H).
Reference Example 2: 6-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile
Figure JPOXMLDOC01-appb-C000012
To a solution of the compound obtained in Reference Example 1 (4.8 g) in DMSO (100 mL) was added sodium cyanide (2.24 g) and 1,4-diazabicyclo [2.2.2] octane (1.28 g). Stir for 3 hours at ° C. After cooling to room temperature, water was added to the reaction solution, and insoluble matters were removed by filtration. Ethyl acetate was added to the obtained filtrate, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and then dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure to obtain the title compound (3.67 g).
1 H-NMR (300 MHz, DMSO-d 6 ) δ 12.20 (s, 1H), 10.48 (s, 1H), 8.35 (s, 1H), 2.22 (s, 3H).
参考例3:(3-メトキシフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノン
Figure JPOXMLDOC01-appb-C000013
 参考例2で得た化合物(3.40g)のTHF(170mL)溶液に、3-メトキシフェニルマグネシウムブロミドの1.0M THF溶液(68mL)を加え、室温で1時間攪拌した。反応液に2規定塩酸(70mL)を加え室温にて18時間撹拌した後、6規定水酸化ナトリウム水溶液(12mL)を加え、THFを減圧留去した。析出した固体をろ取し、水および酢酸エチルで洗浄した後に減圧乾燥することによって、表題化合物(4.5g)を得た。
1H-NMR(300 MHz, DMSO-d6)δ12.07 (s, 1H), 10.19 (s, 1H), 8.40 (d, 1H), 7.48-7.39 (m, 3H), 7.29-7.25 (m, 1H), 3.80 (s, 3H), 2.16 (s, 3H).
Reference Example 3: (3-Methoxyphenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanone
Figure JPOXMLDOC01-appb-C000013
To a solution of the compound (3.40 g) obtained in Reference Example 2 in THF (170 mL) was added 1.0 M THF solution (68 mL) of 3-methoxyphenylmagnesium bromide, and the mixture was stirred at room temperature for 1 hour. 2N Hydrochloric acid (70 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 18 hr. 6N aqueous sodium hydroxide solution (12 mL) was added, and THF was evaporated under reduced pressure. The precipitated solid was collected by filtration, washed with water and ethyl acetate, and then dried under reduced pressure to obtain the title compound (4.5 g).
1 H-NMR (300 MHz, DMSO-d 6 ) δ12.07 (s, 1H), 10.19 (s, 1H), 8.40 (d, 1H), 7.48-7.39 (m, 3H), 7.29-7.25 (m , 1H), 3.80 (s, 3H), 2.16 (s, 3H).
参考例4:(3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノン
Figure JPOXMLDOC01-appb-C000014
 参考例2で得た化合物(100mg)のTHF(10mL)溶液に、氷冷下、3,4-ジフルオロフェニルマグネシウムブロミドの0.5M THF溶液(3mL)を加え、氷冷下45分攪拌した。2規定塩酸(1mL)を加えた後、室温に昇温し45分撹拌した。反応液に6規定水酸化ナトリウム水溶液(0.5mL)および水を加えた後、さらに酢酸エチルを加え、目的物を有機層に抽出した。有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して得られた残渣にヘキサン/酢酸エチル(1/1、3mL)を加え、超音波洗浄機にかけ、析出した固体をろ取し、ヘキサン/酢酸エチル(1/1)で洗浄(1mL×4)して表題化合物(91mg)を得た。
1H-NMR(400 MHz, DMSO-d6)δ12.10 (s, 1H), 10.24 (s, 1H), 8.43 (s, 1H), 8.07-8.01 (m, 1H), 7.93 (d, 1H), 7.84-7.82 (m, 1H), 2.18 (s, 3H).
Reference Example 4: (3,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanone
Figure JPOXMLDOC01-appb-C000014
To a solution of the compound obtained in Reference Example 2 (100 mg) in THF (10 mL) was added 3,4-difluorophenylmagnesium bromide in 0.5 M THF (3 mL) under ice cooling, and the mixture was stirred for 45 minutes under ice cooling. After 2N hydrochloric acid (1 mL) was added, the mixture was warmed to room temperature and stirred for 45 minutes. A 6N aqueous sodium hydroxide solution (0.5 mL) and water were added to the reaction mixture, and ethyl acetate was further added to extract the desired product into the organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. Hexane / ethyl acetate (1/1, 3 mL) was added to the residue obtained by concentrating the organic layer under reduced pressure, and the mixture was passed through an ultrasonic cleaner, and the precipitated solid was collected by filtration and filtered with hexane / ethyl acetate (1/1). Washing (1 mL × 4) gave the title compound (91 mg).
1 H-NMR (400 MHz, DMSO-d 6 ) δ12.10 (s, 1H), 10.24 (s, 1H), 8.43 (s, 1H), 8.07-8.01 (m, 1H), 7.93 (d, 1H ), 7.84-7.82 (m, 1H), 2.18 (s, 3H).
参考例5:(3,5-ジメトキシフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノン
Figure JPOXMLDOC01-appb-C000015
 参考例2で得た化合物(4.0g)のTHF(120mL)溶液に、3,5-ジメトキシフェニルマグネシウムブロミドの1M THF溶液(80mL)を加え、2時間20分攪拌した。2規定塩酸(80mL)を加えた後、室温で13時間撹拌した。反応液に6規定水酸化ナトリウム水溶液(15mL)および水を加えた後、さらに酢酸エチルを加え目的物を有機層に抽出した。有機層を飽和食塩水で洗浄後、無水硫酸マグネシウムで乾燥した。有機層を減圧濃縮して得られた残渣にヘキサン/酢酸エチル(1/1、40mL)を加え、超音波洗浄機にかけ、析出した固体をろ取し、ヘキサン/酢酸エチル(1/1)で洗浄(10mL×2)して表題化合物(4.73g)を得た。
1H-NMR(300 MHz, DMSO-d6)δ12.08 (s, 1H), 10.19 (s, 1H), 8.39 (d, 1H), 7.00 (s, 2H), 6.83 (s, 1H), 3.77 (s, 6H), 2.16 (s, 3H).
Reference Example 5: (3,5-dimethoxyphenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanone
Figure JPOXMLDOC01-appb-C000015
To a solution of the compound (4.0 g) obtained in Reference Example 2 in THF (120 mL) was added 3,5-dimethoxyphenylmagnesium bromide in 1M THF (80 mL), and the mixture was stirred for 2 hours and 20 minutes. 2N Hydrochloric acid (80 mL) was added, and the mixture was stirred at room temperature for 13 hr. A 6N aqueous sodium hydroxide solution (15 mL) and water were added to the reaction solution, and ethyl acetate was further added to extract the desired product into an organic layer. The organic layer was washed with saturated brine and dried over anhydrous magnesium sulfate. Hexane / ethyl acetate (1/1, 40 mL) was added to the residue obtained by concentrating the organic layer under reduced pressure, and the mixture was passed through an ultrasonic cleaner, and the precipitated solid was collected by filtration and filtered with hexane / ethyl acetate (1/1). Washing (10 mL × 2) gave the title compound (4.73 g).
1 H-NMR (300 MHz, DMSO-d 6 ) δ12.08 (s, 1H), 10.19 (s, 1H), 8.39 (d, 1H), 7.00 (s, 2H), 6.83 (s, 1H), 3.77 (s, 6H), 2.16 (s, 3H).
参考例6:tert-ブチル 5-アミノ-3-メチル-1H-ピラゾール-1-カルボキシレート
Figure JPOXMLDOC01-appb-C000016
 3-メチル-1H-ピラゾール-5-アミン(6.79g)のクロロホルム溶液(200mL)にBocO(15.26g)を室温で加えた。7時間攪拌後、溶媒を減圧留去し、残渣にヘキサン(50mL)を加えて撹拌した。析出した固体をろ取し、表題化合物(12.3g)を得た。
1H-NMR (300 MHz, DMSO-d6)δ 6.24 (br-s, 2H), 5.13 (s, 1H), 1.99 (s, 3H), 1.53 (s, 9H).
Reference Example 6: tert-butyl 5-amino-3-methyl-1H-pyrazole-1-carboxylate
Figure JPOXMLDOC01-appb-C000016
Boc 2 O (15.26 g) was added to a chloroform solution (200 mL) of 3-methyl-1H-pyrazol-5-amine (6.79 g) at room temperature. After stirring for 7 hours, the solvent was distilled off under reduced pressure, and hexane (50 mL) was added to the residue and stirred. The precipitated solid was collected by filtration to obtain the title compound (12.3 g).
1 H-NMR (300 MHz, DMSO-d 6 ) δ 6.24 (br-s, 2H), 5.13 (s, 1H), 1.99 (s, 3H), 1.53 (s, 9H).
参考例7:tert-ブチル 3-(6-シアノピリジン-2-イルアミノ)-5-メチル-1H-ピラゾール-1-カルボキシレート
Figure JPOXMLDOC01-appb-C000017
 参考例6で得た化合物(3.94g)、2-クロロ-6-シアノピリジン(2.76g)、Pd(OAc)(448mg)、Xantphos(1.15g)、炭酸セシウム(16.25g)およびジオキサン(100mL)を混合し、3時間加熱還流した。室温まで放冷し、セライトろ過後、ろ液を減圧濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、表題化合物(3.54g)を得た。
1H-NMR (300 MHz, DMSO-d6)δ 10.4 (s, 1H), 7.82(dd, 1H), 7.45 (d, 1H), 7.42 (d, 1H), 6.57 (s, 1H), 2.47 (s, 3H), 1.56 (s, 9H).
Reference Example 7: tert-butyl 3- (6-cyanopyridin-2-ylamino) -5-methyl-1H-pyrazole-1-carboxylate
Figure JPOXMLDOC01-appb-C000017
The compound (3.94 g) obtained in Reference Example 6, 2-chloro-6-cyanopyridine (2.76 g), Pd (OAc) 2 (448 mg), Xantphos (1.15 g), cesium carbonate (16.25 g) And dioxane (100 mL) were mixed and heated to reflux for 3 hours. The mixture was allowed to cool to room temperature, filtered through Celite, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane / ethyl acetate) to give the title compound (3.54 g).
1 H-NMR (300 MHz, DMSO-d 6 ) δ 10.4 (s, 1H), 7.82 (dd, 1H), 7.45 (d, 1H), 7.42 (d, 1H), 6.57 (s, 1H), 2.47 (s, 3H), 1.56 (s, 9H).
参考例8:tert-ブチル 3-((6-(3,4-ジフルオロベンゾイル)ピリジン-2-イル)アミノ)-5-メチル-1H-ピラゾール-1-カルボキシレート
Figure JPOXMLDOC01-appb-C000018
 参考例7で得た化合物(1.8g)のTHF(20mL)溶液に、氷冷下、3,4-ジフルオロフェニルマグネシウムブロミドの1.5M THF溶液(12mL)を加えた。室温下2時間攪拌し、10%硫酸水素カリウム水溶液(30mL)を加えた。室温下1時間撹拌した後飽和重曹水を加えて中性にし、反応混合物に酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して得られた残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、表題化合物(1.70g)を得た。
1H-NMR (300 MHz, DMSO-d6)δ 10.2 (s, 1H), 8.09 (m, 1H), 7.89-7.82 (m, 2H), 7.69-7.60 (m, 1H), 7.48-7.41 (m, 2H), 6.32 (s, 1H), 2.33 (s, 3H), 1.55 (s, 9H).
Reference Example 8: tert-butyl 3-((6- (3,4-difluorobenzoyl) pyridin-2-yl) amino) -5-methyl-1H-pyrazole-1-carboxylate
Figure JPOXMLDOC01-appb-C000018
To a THF (20 mL) solution of the compound obtained in Reference Example 7 (1.8 g), a 1.5 M THF solution (12 mL) of 3,4-difluorophenylmagnesium bromide was added under ice cooling. The mixture was stirred at room temperature for 2 hours, and 10% aqueous potassium hydrogen sulfate solution (30 mL) was added. After stirring at room temperature for 1 hour, the reaction mixture was neutralized with saturated aqueous sodium hydrogen carbonate, and ethyl acetate was added to the reaction mixture. The target product was extracted into the organic layer, and the organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. . The residue obtained by concentrating the organic layer under reduced pressure was purified by silica gel column chromatography (hexane / ethyl acetate) to obtain the title compound (1.70 g).
1 H-NMR (300 MHz, DMSO-d 6 ) δ 10.2 (s, 1H), 8.09 (m, 1H), 7.89-7.82 (m, 2H), 7.69-7.60 (m, 1H), 7.48-7.41 ( m, 2H), 6.32 (s, 1H), 2.33 (s, 3H), 1.55 (s, 9H).
参考例9:(3,4-ジフルオロフェニル)(6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリジン-2-イル)メタノン
Figure JPOXMLDOC01-appb-C000019
 参考例8で得た化合物(19mg)にジクロロメタン(0.5mL)およびトリフルオロ酢酸(0.2mL)を加え、室温で3時間撹拌した。反応液を減圧濃縮し、得られた残渣に飽和重曹水および酢酸エチルを加え、目的物を有機層に抽出した。有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮し、得られた粗生成物をジエチルエーテルで洗浄して表題化合物(8.0mg)を得た。
1H-NMR (300 MHz, DMSO-d6)δ 9.47 (s, 1H), 8.13 (m, 1H), 7.89 (m, 1H), 7.76 (m, 1H), 7.63 (m, 1H), 7.42-7.33 (m, 2H), 5.94 (s, 1H), 2.11 (s, 3H).
Reference Example 9: (3,4-Difluorophenyl) (6-((5-methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) methanone
Figure JPOXMLDOC01-appb-C000019
Dichloromethane (0.5 mL) and trifluoroacetic acid (0.2 mL) were added to the compound (19 mg) obtained in Reference Example 8, and the mixture was stirred at room temperature for 3 hours. The reaction mixture was concentrated under reduced pressure, saturated aqueous sodium hydrogen carbonate and ethyl acetate were added to the resulting residue, and the desired product was extracted into the organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure, and the resulting crude product was washed with diethyl ether to obtain the title compound (8.0 mg).
1 H-NMR (300 MHz, DMSO-d 6 ) δ 9.47 (s, 1H), 8.13 (m, 1H), 7.89 (m, 1H), 7.76 (m, 1H), 7.63 (m, 1H), 7.42 -7.33 (m, 2H), 5.94 (s, 1H), 2.11 (s, 3H).
参考例10:(4-エトキシフェニル)(6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリジン-2-イル)メタノン
Figure JPOXMLDOC01-appb-C000020
 参考例7で得た化合物(250mg)のTHF(8mL)溶液に、氷冷下、4-エトキシフェニルマグネシウムブロミドの1M THF溶液(2.5mL)を加え、氷冷下10分攪拌し、室温で18.5時間撹拌した。反応液に飽和塩化アンモニウム水溶液を加えた後、室温で3時間撹拌した。反応液に2規定塩酸(4mL)を加え室温で2時間撹拌し、その後飽和重曹水を加えた。反応混合物に酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して得られた残渣にジクロロメタン(4mL)およびトリフルオロ酢酸(2mL)を加え、室温で2時間撹拌後、減圧濃縮した。得られた残渣に飽和重曹水および酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮し、得られた残渣にヘキサン/酢酸エチル(1/1、6mL)を加え、超音波洗浄機にかけ、析出した固体をろ取し、さらにヘキサン/酢酸エチル(3/1)で洗浄(2mL×2)し、表題化合物(187mg)を得た。
LC/MS, tR 0.77 min, obs MS [M+1] 323.2.
Reference Example 10: (4-Ethoxyphenyl) (6-((5-methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) methanone
Figure JPOXMLDOC01-appb-C000020
To a THF (8 mL) solution of the compound (250 mg) obtained in Reference Example 7 was added a 1M THF solution (2.5 mL) of 4-ethoxyphenylmagnesium bromide under ice-cooling, and the mixture was stirred for 10 minutes under ice-cooling. Stir for 18.5 hours. A saturated aqueous ammonium chloride solution was added to the reaction solution, and the mixture was stirred at room temperature for 3 hours. 2N Hydrochloric acid (4 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 2 hr. After that, saturated aqueous sodium hydrogen carbonate was added. Ethyl acetate was added to the reaction mixture, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. Dichloromethane (4 mL) and trifluoroacetic acid (2 mL) were added to the residue obtained by concentrating the organic layer under reduced pressure, and the mixture was stirred at room temperature for 2 hours and concentrated under reduced pressure. Saturated aqueous sodium bicarbonate and ethyl acetate were added to the resulting residue, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine, and then dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure, and hexane / ethyl acetate (1/1, 6 mL) was added to the resulting residue. The mixture was subjected to an ultrasonic cleaner, and the precipitated solid was collected by filtration, and further hexane / ethyl acetate (3/1). (2 mL × 2) to give the title compound (187 mg).
LC / MS, t R 0.77 min, obs MS [M + 1] 323.2.
参考例11:(6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリジン-2-イル)(3,4,5-トリフルオロフェニル)メタノン
Figure JPOXMLDOC01-appb-C000021
 参考例7で得た化合物(250mg)のTHF(15mL)溶液に、氷冷下、3,4,5-トリフルオロフェニルマグネシウムブロミドの0.3M THF溶液(8.4mL)を加え、氷冷下10分攪拌し、室温で18.5時間撹拌した。反応液に飽和塩化アンモニウム水溶液を加えた後、室温で3時間撹拌した。反応液に2規定塩酸(4mL)を加え室温で2時間撹拌し、飽和重曹水を加えた。反応混合物に酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して得られた残渣にジクロロメタン(4mL)およびトリフルオロ酢酸(2mL)を加え、室温で3時間撹拌した。有機層を減圧濃縮し、得られた残渣に飽和重曹水および酢酸エチルを加え、目的物を有機層に抽出し、飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して得られた残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、表題化合物(250mg)を得た。
LC/MS, tR 0.87 min, obs MS [M+1] 333.1.
Reference Example 11: (6-((5-Methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) (3,4,5-trifluorophenyl) methanone
Figure JPOXMLDOC01-appb-C000021
To a solution of the compound obtained in Reference Example 7 (250 mg) in THF (15 mL) was added a solution of 3,4,5-trifluorophenylmagnesium bromide in 0.3 M THF (8.4 mL) under ice cooling, and the mixture was cooled under ice cooling. The mixture was stirred for 10 minutes and stirred at room temperature for 18.5 hours. A saturated aqueous ammonium chloride solution was added to the reaction solution, and the mixture was stirred at room temperature for 3 hours. 2N Hydrochloric acid (4 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 2 hr, and saturated aqueous sodium hydrogen carbonate was added. Ethyl acetate was added to the reaction mixture, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. Dichloromethane (4 mL) and trifluoroacetic acid (2 mL) were added to the residue obtained by concentrating the organic layer under reduced pressure, and the mixture was stirred at room temperature for 3 hours. The organic layer was concentrated under reduced pressure, saturated aqueous sodium hydrogen carbonate and ethyl acetate were added to the resulting residue, the target product was extracted into the organic layer, washed with saturated brine, and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (hexane / ethyl acetate) to obtain the title compound (250 mg).
LC / MS, t R 0.87 min, obs MS [M + 1] 333.1.
参考例12:(4-クロロ-3-フルオロフェニル)(6-((5-メチル-1H-ピラゾール)アミノ)ピリジン-2-イル)メタノン
Figure JPOXMLDOC01-appb-C000022
 参考例7で得た化合物(250mg)のTHF(8mL)溶液に、氷冷下、3-フルオロ-4-クロロフェニルマグネシウムブロミドの0.5M THF溶液(5mL)を加え、氷冷下10分攪拌し、室温で18.5時間撹拌した。反応液に飽和塩化アンモニウム水溶液を加えた後、室温で3時間撹拌した。反応液に2規定塩酸(4mL)を加え室温で2時間撹拌し、飽和重曹水を加えた。反応混合物に酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して得られた残渣にジクロロメタン(4mL)およびトリフルオロ酢酸(2mL)を加え、室温で3時間撹拌後、減圧濃縮した。得られた残渣に飽和重曹水および酢酸エチルを加え、目的物を有機層に抽出し、飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮し、得られた残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製して表題化合物(250mg)を得た。
LC/MS, tR 0.87 min, obs MS [M+1] 331.1.
Reference Example 12: (4-Chloro-3-fluorophenyl) (6-((5-methyl-1H-pyrazole) amino) pyridin-2-yl) methanone
Figure JPOXMLDOC01-appb-C000022
To a THF (8 mL) solution of the compound (250 mg) obtained in Reference Example 7 was added a 0.5 M THF solution (5 mL) of 3-fluoro-4-chlorophenylmagnesium bromide under ice cooling, and the mixture was stirred for 10 minutes under ice cooling. And stirred at room temperature for 18.5 hours. A saturated aqueous ammonium chloride solution was added to the reaction solution, and the mixture was stirred at room temperature for 3 hours. 2N Hydrochloric acid (4 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 2 hr, and saturated aqueous sodium hydrogen carbonate was added. Ethyl acetate was added to the reaction mixture, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. Dichloromethane (4 mL) and trifluoroacetic acid (2 mL) were added to the residue obtained by concentrating the organic layer under reduced pressure, and the mixture was stirred at room temperature for 3 hours and concentrated under reduced pressure. Saturated aqueous sodium hydrogen carbonate and ethyl acetate were added to the resulting residue, and the target product was extracted into the organic layer, washed with saturated brine, and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (hexane / ethyl acetate) to obtain the title compound (250 mg).
LC / MS, t R 0.87 min, obs MS [M + 1] 331.1.
参考例13:tert-ブチル 3-((6-(3,5-ジメトキシベンゾイル)ピリジン-2-イル)アミノ)-5-メチル-1H-ピラゾール-1-カルボキシレート
Figure JPOXMLDOC01-appb-C000023
 参考例7で得た化合物(500mg)のTHF(6.7mL)溶液に、氷冷下、3,5-ジメトキシフェニルマグネシウムブロミドの0.5M THF溶液(16mL)を加え、氷冷下10分攪拌し、室温で18.5時間撹拌した。反応液に飽和塩化アンモニウム水溶液を加えた後、室温で3時間撹拌し、さらに飽和重曹水を加えた。反応混合物に酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮し、得られた残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、表題化合物(450mg)を得た。
LC/MS, tR 1.07 min, obs MS [M+1] 439.6.
Reference Example 13: tert-butyl 3-((6- (3,5-dimethoxybenzoyl) pyridin-2-yl) amino) -5-methyl-1H-pyrazole-1-carboxylate
Figure JPOXMLDOC01-appb-C000023
To a THF (6.7 mL) solution of the compound (500 mg) obtained in Reference Example 7 was added a 0.5 M THF solution (16 mL) of 3,5-dimethoxyphenylmagnesium bromide under ice cooling, and the mixture was stirred for 10 minutes under ice cooling. And stirred at room temperature for 18.5 hours. A saturated aqueous ammonium chloride solution was added to the reaction solution, and the mixture was stirred at room temperature for 3 hours, and saturated aqueous sodium hydrogen carbonate was further added. Ethyl acetate was added to the reaction mixture, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (hexane / ethyl acetate) to obtain the title compound (450 mg).
LC / MS, t R 1.07 min, obs MS [M + 1] 439.6.
参考例14:tert-ブチル 3-((6-(3-メトキシベンゾイル)ピリジン-2-イル)アミノ)-5-メチル-1H-ピラゾール-1-カルボキシレート
Figure JPOXMLDOC01-appb-C000024
 参考例7で得た化合物(500mg)と3-メトキシフェニルマグネシウムブロマイドの0.5M THF溶液を用いて、参考例8と同様にして表題化合物(466mg)を得た。
LC/MS, tR 1.08 min, obs MS [M+1] 409.3.
Reference Example 14: tert-butyl 3-((6- (3-methoxybenzoyl) pyridin-2-yl) amino) -5-methyl-1H-pyrazole-1-carboxylate
Figure JPOXMLDOC01-appb-C000024
The title compound (466 mg) was obtained in the same manner as in Reference Example 8 using the compound (500 mg) obtained in Reference Example 7 and a 0.5 M THF solution of 3-methoxyphenylmagnesium bromide.
LC / MS, t R 1.08 min, obs MS [M + 1] 409.3.
参考例15:(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノン
Figure JPOXMLDOC01-appb-C000025
 参考例2で得た化合物(4.80g)のTHF(40mL)懸濁溶液に、氷冷下、3,4、5-トリフルオロフェニルマグネシウムブロミドの0.5M THF溶液(200mL)をゆっくり加えた。室温下2時間攪拌した後、氷冷下、2規定塩酸(200mL)を加え、室温に昇温して14時間撹拌した。6規定水酸化ナトリウム水溶液(55mL)を加え、反応混合物を減圧濃縮した。析出した固体をろ取し、水(40mL×2)、酢酸エチル(40mL×3)、およびTHF(10mL×3)で洗浄した後減圧乾燥して表題化合物(6.50g)を得た。
1H-NMR (300 MHz, DMSO-d6)δ 12.1 (s, 1H), 10.2 (s, 1H), 8.42 (d, 1H), 7.97-7.90 (m, 2H), 2.18 (s, 3H).
Reference Example 15: (4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanone
Figure JPOXMLDOC01-appb-C000025
To a suspension of the compound (4.80 g) obtained in Reference Example 2 in THF (40 mL) was slowly added a 0.5 M THF solution (200 mL) of 3,4,5-trifluorophenylmagnesium bromide under ice cooling. . After stirring at room temperature for 2 hours, 2N hydrochloric acid (200 mL) was added under ice cooling, and the mixture was warmed to room temperature and stirred for 14 hours. A 6N aqueous sodium hydroxide solution (55 mL) was added, and the reaction mixture was concentrated under reduced pressure. The precipitated solid was collected by filtration, washed with water (40 mL × 2), ethyl acetate (40 mL × 3), and THF (10 mL × 3) and then dried under reduced pressure to obtain the title compound (6.50 g).
1 H-NMR (300 MHz, DMSO-d 6 ) δ 12.1 (s, 1H), 10.2 (s, 1H), 8.42 (d, 1H), 7.97-7.90 (m, 2H), 2.18 (s, 3H) .
参考例16:2-クロロ-6-メチル-N-(5-メチル-1H-ピラゾール-3-イル)ピリミジン-4-アミン
Figure JPOXMLDOC01-appb-C000026
 5-メチル-ピラゾール-3-アミン(1.6g)のエタノール(19.2mL)溶液に2,4-ジクロロ‐6-メチルピリミジン(1.35g)および炭酸水素ナトリウム(2.47g)を加え、110℃にて8時間攪拌した。室温まで放冷した後、反応液に水および酢酸エチルを加え、目的物を有機層に抽出した。有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮し、得られた残渣をシリカゲルカラムクロマトグラフィー(酢酸エチル/ヘキサン)で精製して表題化合物(1.24g)を得た。
1H-NMR (400 MHz, DMSO-d6)δ 12.0 (s, 1H), 10.1 (s, 1H), 6.00 (br-s, 1H), 2.25 (s, 3H), 2.20(s, 3H).
Reference Example 16: 2-Chloro-6-methyl-N- (5-methyl-1H-pyrazol-3-yl) pyrimidin-4-amine
Figure JPOXMLDOC01-appb-C000026
To a solution of 5-methyl-pyrazol-3-amine (1.6 g) in ethanol (19.2 mL) was added 2,4-dichloro-6-methylpyrimidine (1.35 g) and sodium bicarbonate (2.47 g), Stir at 110 ° C. for 8 hours. After allowing to cool to room temperature, water and ethyl acetate were added to the reaction solution, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure, and the obtained residue was purified by silica gel column chromatography (ethyl acetate / hexane) to give the title compound (1.24 g).
1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.0 (s, 1H), 10.1 (s, 1H), 6.00 (br-s, 1H), 2.25 (s, 3H), 2.20 (s, 3H) .
参考例17:4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-カルボニトリル
Figure JPOXMLDOC01-appb-C000027
 参考例16で得た化合物(1.24g)のDMSO(100mL)/イソプロピルアルコール(3mL)溶液にシアン化ナトリウム(0.31g)および1,4-ジアザビシクロ[2.2.2]オクタン(0.32g)を加え、90℃にて5時間攪拌した。室温に冷却後、水を加え、不溶物をろ過により除去した。得られたろ液に酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して表題化合物(0.66g)を得た。
1H-NMR (400 MHz, DMSO-d6)δ 12.13 (s, 1H), 10.29 (s, 1H), 2.32 (s, 3H), 2.21 (s, 3H).
Reference Example 17: 4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile
Figure JPOXMLDOC01-appb-C000027
To a solution of the compound obtained in Reference Example 16 (1.24 g) in DMSO (100 mL) / isopropyl alcohol (3 mL), sodium cyanide (0.31 g) and 1,4-diazabicyclo [2.2.2] octane (0. 32 g) was added and stirred at 90 ° C. for 5 hours. After cooling to room temperature, water was added and insolubles were removed by filtration. Ethyl acetate was added to the obtained filtrate, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and then dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure to obtain the title compound (0.66 g).
1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.13 (s, 1H), 10.29 (s, 1H), 2.32 (s, 3H), 2.21 (s, 3H).
参考例18:(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノン
Figure JPOXMLDOC01-appb-C000028
 参考例17で得た化合物(0.2g)のTHF(4mL)溶液に、氷冷下、3,4,5-トリフルオロフェニルマグネシウムブロミドの1.0M THF溶液(5.6mL)をゆっくり加えた。室温下3時間攪拌した後、氷冷下、10%硫酸水素カリウム水溶液(30mL)を加えた後、室温に昇温し16時間撹拌した。反応液に飽和炭酸水素ナトリウム水溶液、酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮し、析出した固体をろ取した。さらにクロロホルムとヘキサンで洗浄した後減圧乾燥して表題化合物(0.22g)を得た。
1H-NMR (400 MHz, DMSO-d6)δ 12.0 (s, 1H), 10.0 (s, 1H), 7.93 (t, 2H), 2.36 (s, 3H) , 2.17 (s, 3H).
Reference Example 18: (4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanone
Figure JPOXMLDOC01-appb-C000028
To a THF (4 mL) solution of the compound (0.2 g) obtained in Reference Example 17, a 1.0 M THF solution (5.6 mL) of 3,4,5-trifluorophenylmagnesium bromide was slowly added under ice cooling. . After stirring at room temperature for 3 hours, 10% aqueous potassium hydrogen sulfate solution (30 mL) was added under ice cooling, and the mixture was warmed to room temperature and stirred for 16 hours. A saturated aqueous sodium hydrogen carbonate solution and ethyl acetate were added to the reaction solution, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine, and then dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure, and the precipitated solid was collected by filtration. The product was further washed with chloroform and hexane and dried under reduced pressure to obtain the title compound (0.22 g).
1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.0 (s, 1H), 10.0 (s, 1H), 7.93 (t, 2H), 2.36 (s, 3H), 2.17 (s, 3H).
参考例19:(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4-ジフルオロフェニル)メタノン
Figure JPOXMLDOC01-appb-C000029
 参考例17で得た化合物(0.2g)のTHF(5mL)溶液に、氷冷下、3,4-ジフルオロフェニルマグネシウムブロミドの0.5M THF溶液(7.5mL)をゆっくり加え、室温下15時間攪拌した。反応液に2規定塩酸(3.1mL)を加え、室温下50分撹拌した。反応液に飽和炭酸ナトリウム水溶液および酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮し、析出した固体をろ取し、ヘキサンと酢酸エチルで洗浄した後、減圧乾燥して表題化合物(0.28g)を得た。
LC/MS, tR 0.75 min, obs MS [M+1] 330.6.
Reference Example 19: (4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4-difluorophenyl) methanone
Figure JPOXMLDOC01-appb-C000029
To a solution of the compound (0.2 g) obtained in Reference Example 17 in THF (5 mL) was slowly added a 0.5 M THF solution (7.5 mL) of 3,4-difluorophenylmagnesium bromide under ice cooling, and the mixture was stirred at room temperature. Stir for hours. 2N Hydrochloric acid (3.1 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 50 min. A saturated aqueous sodium carbonate solution and ethyl acetate were added to the reaction solution, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine, and then dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure, and the precipitated solid was collected by filtration, washed with hexane and ethyl acetate, and then dried under reduced pressure to obtain the title compound (0.28 g).
LC / MS, t R 0.75 min, obs MS [M + 1] 330.6.
参考例20:(2,4,5-トリフルオロフェニル)マグネシウムブロミド
Figure JPOXMLDOC01-appb-C000030
 減圧下120℃で40分乾燥した削状マグネシウム(0.35g)にTHF(12mL)および1,2-ジブロモエタン(51μL)を加え、室温下20分撹拌した。反応混合物に氷冷下、2,4,5-トリフルオロフェニルブロミド(2.5g)のTHF(6mL)溶液をゆっくり加えた。反応混合物を室温に昇温し、1時間攪拌して表題化合物(1M THF溶液12mL)を得た。
Reference Example 20: (2,4,5-trifluorophenyl) magnesium bromide
Figure JPOXMLDOC01-appb-C000030
THF (12 mL) and 1,2-dibromoethane (51 μL) were added to ground magnesium (0.35 g) dried at 120 ° C. for 40 minutes under reduced pressure, and stirred at room temperature for 20 minutes. To the reaction mixture, a solution of 2,4,5-trifluorophenyl bromide (2.5 g) in THF (6 mL) was slowly added under ice cooling. The reaction mixture was warmed to room temperature and stirred for 1 hour to give the title compound (12 mL of 1M THF solution).
参考例21:(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノン
Figure JPOXMLDOC01-appb-C000031
 参考例2で得た化合物(4.2g)のTHF(30mL)溶液に、氷冷下、参考例20で得た2,4,5-トリフルオロフェニルマグネシウムブロミド(1M THF溶液120mL)をゆっくり加え、室温下2時間50分攪拌した。反応液に2規定塩酸(7mL)を加え、室温下2時間撹拌した。反応液に飽和炭酸ナトリウム水溶液および酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して得られた残渣をシリカゲルカラムクロマトグラフィー(クロロホルム/メタノール)で精製して表題化合物(2.49g)を得た。
LC/MS, tR 0.74 min, obs MS [M+1] 349.2.
Reference Example 21: (4-((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanone
Figure JPOXMLDOC01-appb-C000031
To a THF (30 mL) solution of the compound (4.2 g) obtained in Reference Example 2, 2,4,5-trifluorophenylmagnesium bromide (120 mL of 1M THF solution) obtained in Reference Example 20 was slowly added under ice cooling. The mixture was stirred at room temperature for 2 hours and 50 minutes. 2N Hydrochloric acid (7 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 2 hr. A saturated aqueous sodium carbonate solution and ethyl acetate were added to the reaction solution, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine, and then dried over anhydrous sodium sulfate. The residue obtained by concentrating the organic layer under reduced pressure was purified by silica gel column chromatography (chloroform / methanol) to obtain the title compound (2.49 g).
LC / MS, t R 0.74 min, obs MS [M + 1] 349.2.
参考例22:(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノン
Figure JPOXMLDOC01-appb-C000032
 参考例17で得た化合物(0.5g)のTHF(15mL)溶液に、氷冷下、参考例20で得た2,4,5-トリフルオロフェニルマグネシウムブロミド(1M THF溶液10mL)をゆっくり加え、室温下2時間50分攪拌した。反応液に2規定塩酸(7mL)を加え、室温下2時間撹拌した。反応混合物に飽和炭酸ナトリウム水溶液および酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮し、析出した固体をろ取し、ヘキサンと酢酸エチルで洗浄した後、減圧乾燥して表題化合物(0.62g)を得た。
LC/MS, tR 0.74 min, obs MS [M+1] 349.2.
Reference Example 22: (4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanone
Figure JPOXMLDOC01-appb-C000032
To a THF (15 mL) solution of the compound (0.5 g) obtained in Reference Example 17, 2,4,5-trifluorophenylmagnesium bromide (1 M THF solution 10 mL) obtained in Reference Example 20 was slowly added under ice cooling. The mixture was stirred at room temperature for 2 hours and 50 minutes. 2N Hydrochloric acid (7 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 2 hr. A saturated aqueous sodium carbonate solution and ethyl acetate were added to the reaction mixture, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine, and then dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure, and the precipitated solid was collected by filtration, washed with hexane and ethyl acetate, and then dried under reduced pressure to obtain the title compound (0.62 g).
LC / MS, t R 0.74 min, obs MS [M + 1] 349.2.
参考例23:tert-ブチル 3-((6-((3,4-ジフルオロフェニル)(ヒドロキシ)メチル)ピリジン-2-イル)アミノ)-5-メチル-1H-ピラゾール-1-カルボキシレート
Figure JPOXMLDOC01-appb-C000033
 参考例8で得た化合物(1.00g)のメタノール(6mL)およびTHF(6mL)溶液に水素化ホウ素ナトリウム(113mg)を加え、室温で1時間攪拌した。反応液に水および酢酸エチルを加えた後、目的物を有機層に抽出し、有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して得られた残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、表題化合物(0.950g)を得た。
LC/MS, tR 0.84 min, obs MS [M+1] 417.3.
Reference Example 23: tert-butyl 3-((6-((3,4-difluorophenyl) (hydroxy) methyl) pyridin-2-yl) amino) -5-methyl-1H-pyrazole-1-carboxylate
Figure JPOXMLDOC01-appb-C000033
Sodium borohydride (113 mg) was added to a solution of the compound obtained in Reference Example 8 (1.00 g) in methanol (6 mL) and THF (6 mL), and the mixture was stirred at room temperature for 1 hour. Water and ethyl acetate were added to the reaction solution, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The residue obtained by concentrating the organic layer under reduced pressure was purified by silica gel column chromatography (hexane / ethyl acetate) to obtain the title compound (0.950 g).
LC / MS, t R 0.84 min, obs MS [M + 1] 417.3.
参考例24:2-クロロ-N-(5-シクロプロピル-1H-ピラゾール-3-イル)ピリミジン-4-アミン
Figure JPOXMLDOC01-appb-C000034
 5-シクロプロピル-ピラゾール-3-アミン(1.10g)のエタノール(30mL)溶液に2,4-ジクロロピリミジン(1.21g)およびN-エチル-ジイソプロピルアミン(1.49mL)を加え、70℃にて11時間攪拌した。室温まで放冷した後、水を加え、酢酸エチルで3回抽出した。有機相を飽和食塩水で洗浄後、硫酸ナトリウムで、乾燥し、エバポレーターで濃縮した。残渣をシリカゲルカラムクロマトグラフィーで精製し、表題化合物(1.04g)を得た。
1H-NMR (400 MHz, CDCl3)δ 8.13 (d, J = 5.6 Hz, 1H), 7.96 (br-s, 1H), 7.05 (br-s, 1H), 5.87 (br-s, 1H), 1.80-1.87 (m, 1H), 0.94-1.01 (m, 2H), 0.72-0.74 (m, 2H).
LC/MS, tR 0.67 min, obs MS [M+1] 236.1
Reference Example 24: 2-chloro-N- (5-cyclopropyl-1H-pyrazol-3-yl) pyrimidin-4-amine
Figure JPOXMLDOC01-appb-C000034
2,4-Dichloropyrimidine (1.21 g) and N-ethyl-diisopropylamine (1.49 mL) were added to a solution of 5-cyclopropyl-pyrazol-3-amine (1.10 g) in ethanol (30 mL) at 70 ° C. For 11 hours. After allowing to cool to room temperature, water was added, and the mixture was extracted 3 times with ethyl acetate. The organic phase was washed with saturated brine, dried over sodium sulfate, and concentrated with an evaporator. The residue was purified by silica gel column chromatography to obtain the title compound (1.04 g).
1 H-NMR (400 MHz, CDCl 3 ) δ 8.13 (d, J = 5.6 Hz, 1H), 7.96 (br-s, 1H), 7.05 (br-s, 1H), 5.87 (br-s, 1H) , 1.80-1.87 (m, 1H), 0.94-1.01 (m, 2H), 0.72-0.74 (m, 2H).
LC / MS, t R 0.67 min, obs MS [M + 1] 236.1
参考例25:6-((5-シクロプロピル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-カルボニトリル
Figure JPOXMLDOC01-appb-C000035
 参考例24で得た化合物(364mg)のDMSO(1.4mL)/イソプロピルアルコール(0.70mL)懸濁液にシアン化ナトリウム(106mg)および1,4-ジアザビシクロ[2.2.2]オクタン(87mg)を加え、90℃にて6時間攪拌した。室温に冷却後、反応液に水を加え、酢酸エチルで3回抽出した。有機相を飽和食塩水で洗浄後、硫酸ナトリウムで乾燥し、エバポレーターで濃縮した。残渣を酢酸エチルで希釈し、その後ヘキサンで10倍に希釈した。生じた固形物をろ取、乾燥して表題化合物(0.309g)を得た。
1H-NMR (400 MHz, DMSO-d6)δ 12.12 (s, 1H), 10.38 (s, 1H), 8.30 (s, 1H), 1.82-1.88 (m, 1H), 0.86-0.91 (m, 2H), 0.62-0.66 (m, 2H).
LC/MS, tR 0.673 min, obs MS [M+1] 227.1
Reference Example 25: 6-((5-cyclopropyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile
Figure JPOXMLDOC01-appb-C000035
To a suspension of the compound obtained in Reference Example 24 (364 mg) in DMSO (1.4 mL) / isopropyl alcohol (0.70 mL), sodium cyanide (106 mg) and 1,4-diazabicyclo [2.2.2] octane ( 87 mg), and the mixture was stirred at 90 ° C. for 6 hours. After cooling to room temperature, water was added to the reaction mixture, and the mixture was extracted 3 times with ethyl acetate. The organic phase was washed with saturated brine, dried over sodium sulfate, and concentrated with an evaporator. The residue was diluted with ethyl acetate and then diluted 10-fold with hexane. The resulting solid was collected by filtration and dried to obtain the title compound (0.309 g).
1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.12 (s, 1H), 10.38 (s, 1H), 8.30 (s, 1H), 1.82-1.88 (m, 1H), 0.86-0.91 (m, 2H), 0.62-0.66 (m, 2H).
LC / MS, t R 0.673 min, obs MS [M + 1] 227.1
参考例26:2-クロロ-N-(5-シクロプロピル-1H-ピラゾール-3-イル)-6-メチルピリミジン-4-アミン
Figure JPOXMLDOC01-appb-C000036
 5-シクロプロピル-ピラゾール-3-アミン(1.11g)のエタノール(30mL)溶液に2,4-ジクロロ-6-メチルピリミジン(1.34g)およびN-エチル-ジイソプロピルアミン(1.50mL)を加え、70℃にて5時間攪拌した。室温まで放冷した後、水を加え、酢酸エチルで3回抽出した。有機相を飽和食塩水で洗浄後、硫酸ナトリウムで、乾燥し、エバポレーターで濃縮した。残渣をシリカゲルカラムクロマトグラフィーで精製し、表題化合物(0.96g)を得た。
1H-NMR (400 MHz, CDCl3)δ 7.53 (br-s, 1H), 6.95 (br-s, 1H), 5.87 (br-s, 1H), 2.37 (s, 3H), 1.81-1.88 (m, 1H), 0.97-1.02 (m, 2H), 0.72-0.76 (m, 2H).
LC/MS, tR 0.70 min, obs MS [M+1] 250.1.
Reference Example 26: 2-chloro-N- (5-cyclopropyl-1H-pyrazol-3-yl) -6-methylpyrimidin-4-amine
Figure JPOXMLDOC01-appb-C000036
To a solution of 5-cyclopropyl-pyrazol-3-amine (1.11 g) in ethanol (30 mL) was added 2,4-dichloro-6-methylpyrimidine (1.34 g) and N-ethyl-diisopropylamine (1.50 mL). In addition, the mixture was stirred at 70 ° C. for 5 hours. After allowing to cool to room temperature, water was added, and the mixture was extracted 3 times with ethyl acetate. The organic phase was washed with saturated brine, dried over sodium sulfate, and concentrated with an evaporator. The residue was purified by silica gel column chromatography to obtain the title compound (0.96 g).
1 H-NMR (400 MHz, CDCl 3 ) δ 7.53 (br-s, 1H), 6.95 (br-s, 1H), 5.87 (br-s, 1H), 2.37 (s, 3H), 1.81-1.88 ( m, 1H), 0.97-1.02 (m, 2H), 0.72-0.76 (m, 2H).
LC / MS, t R 0.70 min, obs MS [M + 1] 250.1.
参考例27:4-((5-シクロプロピル-1H-ピラゾール-3-イル)アミノ)-6-メチルピリミジン-2-カルボニトリル
Figure JPOXMLDOC01-appb-C000037
 参考例26で得た化合物(0.32g)のDMSO(1.4mL)/イソプロピルアルコール(0.70mL)溶液にシアン化ナトリウム(75mg)および1,4-ジアザビシクロ[2.2.2]オクタン(72mg)を加え、90℃にて6時間攪拌した。室温に冷却後、反応液に水を加え、ろ過にした。得られた残渣を水で3回、酢酸エチル/ヘキサン=1/10溶媒で2回洗浄後、減圧下乾燥して表題化合物(0.24g)を得た。
1H-NMR (400 MHz, DMSO-d6)δ 12.15 (br-s, 1H), 10.23 (br-s, 1H), 7.30 (br-s, 1H), 5.95 (br-s, 1H), 2.28 (s, 3H), 1.81-1.86 (m, 2H), 0.86-0.90 (m, 2H), 0.62-0.66 (m, 2H).
LC/MS, tR 0.72 min, obs MS[M+1] 241.1.
Reference Example 27: 4-((5-cyclopropyl-1H-pyrazol-3-yl) amino) -6-methylpyrimidine-2-carbonitrile
Figure JPOXMLDOC01-appb-C000037
To a solution of the compound obtained in Reference Example 26 (0.32 g) in DMSO (1.4 mL) / isopropyl alcohol (0.70 mL), sodium cyanide (75 mg) and 1,4-diazabicyclo [2.2.2] octane ( 72 mg) and stirred at 90 ° C. for 6 hours. After cooling to room temperature, water was added to the reaction solution and filtered. The obtained residue was washed 3 times with water and twice with ethyl acetate / hexane = 1/10 solvent, and dried under reduced pressure to give the title compound (0.24 g).
1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.15 (br-s, 1H), 10.23 (br-s, 1H), 7.30 (br-s, 1H), 5.95 (br-s, 1H), 2.28 (s, 3H), 1.81-1.86 (m, 2H), 0.86-0.90 (m, 2H), 0.62-0.66 (m, 2H).
LC / MS, t R 0.72 min, obs MS [M + 1] 241.1.
参考例28:2-クロロ-N-(5-エチル-1H-ピラゾール-3-イル)ピリミジン-4-アミン
Figure JPOXMLDOC01-appb-C000038
 5-エチル-ピラゾール-3-アミン(0.332g)のエタノール(10mL)溶液に2,4-ジクロロピリミジン(0.405g)およびN-エチル-ジイソプロピルアミン(0.497mL)を加え、70℃にて6時間攪拌した。室温まで放冷した後、濃縮し、残渣をシリカゲルカラムクロマトグラフィーで精製し、表題化合物(171mg)を得た。
1H-NMR (400 MHz, CDCl3)δ 8.18 (d, J = 5.2 Hz, 1H), 7.74 (br-s, 1H), 7.11 (br-s, 1H), 6.10 (br-s, 1H), 2.69 (q, J= 7.2Hz, 2H), 1.29 (t, J = 7.2 Hz, 3H).
LC/MS, tR 0.65 min, obs MS [M+1] 224.1.
Reference Example 28: 2-chloro-N- (5-ethyl-1H-pyrazol-3-yl) pyrimidin-4-amine
Figure JPOXMLDOC01-appb-C000038
2,4-Dichloropyrimidine (0.405 g) and N-ethyl-diisopropylamine (0.497 mL) were added to a solution of 5-ethyl-pyrazol-3-amine (0.332 g) in ethanol (10 mL), and the mixture was heated to 70 ° C. And stirred for 6 hours. The mixture was allowed to cool to room temperature, concentrated, and the residue was purified by silica gel column chromatography to give the title compound (171 mg).
1 H-NMR (400 MHz, CDCl 3 ) δ 8.18 (d, J = 5.2 Hz, 1H), 7.74 (br-s, 1H), 7.11 (br-s, 1H), 6.10 (br-s, 1H) , 2.69 (q, J = 7.2Hz, 2H), 1.29 (t, J = 7.2 Hz, 3H).
LC / MS, t R 0.65 min, obs MS [M + 1] 224.1.
参考例29:4-((5-エチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-カルボニトリル
Figure JPOXMLDOC01-appb-C000039
 参考例28で得た化合物(171mg)のDMSO(1.4mL)/イソプロピルアルコール(0.70mL)懸濁液にシアン化ナトリウム(50mg)および1,4-ジアザビシクロ[2.2.2]オクタン(47.7mg)を加え、90℃にて6時間攪拌した。室温に冷却後、反応液に水を加え、酢酸エチル/ヘキサン=1/1で希釈した。この混合物を酢酸エチルで2回抽出し、有機相を飽和食塩水で洗浄、硫酸ナトリウムで乾燥後、エバポレーターで濃縮した。得られた残渣を酢酸エチル/ヘキサン=1/10で、懸濁し、ろ過した。得られた残渣を酢酸エチル/ヘキサン=1/10溶媒で2回洗浄後、減圧下乾燥して表題化合物(102mg)を得た。
LC/MS, tR 0.66 min, obs MS [M+1] 215.2.
Reference Example 29: 4-((5-ethyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile
Figure JPOXMLDOC01-appb-C000039
To a suspension of the compound obtained in Reference Example 28 (171 mg) in DMSO (1.4 mL) / isopropyl alcohol (0.70 mL), sodium cyanide (50 mg) and 1,4-diazabicyclo [2.2.2] octane ( 47.7 mg) was added and the mixture was stirred at 90 ° C. for 6 hours. After cooling to room temperature, water was added to the reaction solution and diluted with ethyl acetate / hexane = 1/1. This mixture was extracted twice with ethyl acetate, and the organic phase was washed with saturated brine, dried over sodium sulfate, and concentrated by an evaporator. The obtained residue was suspended in ethyl acetate / hexane = 1/10 and filtered. The obtained residue was washed twice with ethyl acetate / hexane = 1/10 solvent and dried under reduced pressure to give the title compound (102 mg).
LC / MS, t R 0.66 min, obs MS [M + 1] 215.2.
参考例30:(4-((5-シクロプロピル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノン
Figure JPOXMLDOC01-appb-C000040
 参考例25で得た化合物(87mg)のTHF(10mL)溶液に、氷冷下、3,4、5-トリフルオロフェニルマグネシウムブロミドの0.5M THF溶液(3.2mL)をゆっくり加えた。室温下2時間攪拌した後、氷冷下、2M塩酸(200mL)を加え、室温に昇温して14時間撹拌した。水(2.0mL)と酢酸(1.0mL)を加え、1時間撹拌した。酢酸エチルで3回抽出し、有機相を飽和食塩水で洗浄後、硫酸ナトリウムで、乾燥し、エバポレーターで濃縮した。残渣をシリカゲルカラムクロマトグラフィーで精製し、表題化合物(45mg)を得た。
1H-NMR (400 MHz, CDCl3)δ 8.40-8.55 (m, 2H), 7.86-7.89 (m, 2H), 7.31 (br-s, 1H), 5.92 (br-s, 1H), 1.84-1.89 (m, 1H), 0.99-1.02 (m, 2H), 0.71-0.73 (m, 2H).
LC/MS, tR 0.90 min, obs MS [M+1] 360.2.
Reference Example 30: (4-((5-Cyclopropyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanone
Figure JPOXMLDOC01-appb-C000040
To a THF (10 mL) solution of the compound (87 mg) obtained in Reference Example 25, a 0.5 M THF solution (3.2 mL) of 3,4,5-trifluorophenylmagnesium bromide was slowly added under ice cooling. After stirring at room temperature for 2 hours, 2M hydrochloric acid (200 mL) was added under ice cooling, and the mixture was warmed to room temperature and stirred for 14 hours. Water (2.0 mL) and acetic acid (1.0 mL) were added and stirred for 1 hour. The mixture was extracted 3 times with ethyl acetate, and the organic phase was washed with saturated brine, dried over sodium sulfate, and concentrated with an evaporator. The residue was purified by silica gel column chromatography to obtain the title compound (45 mg).
1 H-NMR (400 MHz, CDCl 3 ) δ 8.40-8.55 (m, 2H), 7.86-7.89 (m, 2H), 7.31 (br-s, 1H), 5.92 (br-s, 1H), 1.84- 1.89 (m, 1H), 0.99-1.02 (m, 2H), 0.71-0.73 (m, 2H).
LC / MS, t R 0.90 min, obs MS [M + 1] 360.2.
参考例31:(4-((5-エチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノン
Figure JPOXMLDOC01-appb-C000041
 参考例29で得た化合物(62mg)のTHF(4mL)懸濁溶液に、氷冷下、3,4、5-トリフルオロフェニルマグネシウムブロミドの0.5M THF溶液(2.4mL)をゆっくり加えた。室温下2時間攪拌した後、氷冷下、2M塩酸(200mL)を加え、室温に昇温して14時間撹拌した。水(2.0mL)と酢酸(1.0mL)を加え、1時間撹拌した。酢酸エチルで3回抽出し、有機相を飽和食塩水で洗浄後、硫酸ナトリウムで、乾燥し、エバポレーターで濃縮した。残渣をシリカゲルカラムクロマトグラフィーで精製し、表題化合物(43mg)を得た。
1H-NMR (400 MHz, CDCl3)δ 9.23 (br-s, 1H), 8.41(d, J = 5.6Hz, 1H), 7.87-7.95 (m, 2H), 7.28 (br-s, 1H), 6.03 (br-s, 1H), 2.71 (q, J = 7.6 Hz, 2H), 1.26 (t, J = 7.6 Hz, 3H).
LC/MS, tR 0.90 min, obs MS[M+1] 348.2.
Reference Example 31: (4-((5-Ethyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanone
Figure JPOXMLDOC01-appb-C000041
To a suspension of the compound obtained in Reference Example 29 (62 mg) in THF (4 mL) was slowly added a 0.5 M THF solution (2.4 mL) of 3,4,5-trifluorophenylmagnesium bromide under ice cooling. . After stirring at room temperature for 2 hours, 2M hydrochloric acid (200 mL) was added under ice cooling, and the mixture was warmed to room temperature and stirred for 14 hours. Water (2.0 mL) and acetic acid (1.0 mL) were added and stirred for 1 hour. The mixture was extracted 3 times with ethyl acetate, and the organic phase was washed with saturated brine, dried over sodium sulfate, and concentrated with an evaporator. The residue was purified by silica gel column chromatography to obtain the title compound (43 mg).
1 H-NMR (400 MHz, CDCl 3 ) δ 9.23 (br-s, 1H), 8.41 (d, J = 5.6Hz, 1H), 7.87-7.95 (m, 2H), 7.28 (br-s, 1H) , 6.03 (br-s, 1H), 2.71 (q, J = 7.6 Hz, 2H), 1.26 (t, J = 7.6 Hz, 3H).
LC / MS, t R 0.90 min, obs MS [M + 1] 348.2.
参考例32:6-メチル-4-チオキソ-3,4-ジヒドロピリミジン-2(1H)-オン
Figure JPOXMLDOC01-appb-C000042
 6-メチルピリミジン-2,4(1H,3H)-ジオン(12.6g)のピリジン(100mL)溶液にローソン試薬(22.3g)を加え、140℃にて2時間攪拌した。室温まで放冷後、減圧濃縮して得られた固体に水(300mL)を加え、110℃にて10分攪拌した。室温まで放冷した後、析出した固体をろ過し、水にて洗浄した後、減圧乾燥して標題化合物(13.1g)を得た。
1H-NMR (DMSO-d6)δ 2.00 (s, 3H), 6.13 (s, 1H), 11.5 (s, 1H), 12.3 (s, 1H). 
Reference Example 32: 6-Methyl-4-thioxo-3,4-dihydropyrimidin-2 (1H) -one
Figure JPOXMLDOC01-appb-C000042
Lawesson's reagent (22.3 g) was added to a solution of 6-methylpyrimidine-2,4 (1H, 3H) -dione (12.6 g) in pyridine (100 mL), and the mixture was stirred at 140 ° C. for 2 hours. After cooling to room temperature, water (300 mL) was added to the solid obtained by concentration under reduced pressure, and the mixture was stirred at 110 ° C. for 10 minutes. After allowing to cool to room temperature, the precipitated solid was filtered, washed with water, and dried under reduced pressure to give the title compound (13.1 g).
1 H-NMR (DMSO-d 6 ) δ 2.00 (s, 3H), 6.13 (s, 1H), 11.5 (s, 1H), 12.3 (s, 1H).
参考例33:6-メチル-4-(メチルチオ)ピリミジン-2(1H)-オン
Figure JPOXMLDOC01-appb-C000043
 参考例32で得た化合物(13.1g)を1M水酸化ナトリウム水溶液(287mL)に加え、0℃に冷却した後、ヨードメタン(6.0mL)を滴下して加え、0℃にて2時間攪拌した。さらに室温で2.5時間攪拌後、0℃に冷却し、酢酸(130mL)を加え減圧濃縮した。得られた固体に水を加え、100℃に加熱して溶解させた後、室温まで冷却し再結晶化させた。析出固体をろ過し、水にて洗浄した後、減圧乾燥して標題化合物(10.5g)を得た。
1H-NMR (DMSO-d6)δ 2.10 (s, 3H), 2.40 (s, 3H), 6.19 (s, 1H), 11.4 (br-s, 1H).
Reference Example 33: 6-Methyl-4- (methylthio) pyrimidin-2 (1H) -one
Figure JPOXMLDOC01-appb-C000043
The compound (13.1 g) obtained in Reference Example 32 was added to 1M aqueous sodium hydroxide solution (287 mL), cooled to 0 ° C., iodomethane (6.0 mL) was added dropwise, and the mixture was stirred at 0 ° C. for 2 hours. did. The mixture was further stirred at room temperature for 2.5 hours, cooled to 0 ° C., acetic acid (130 mL) was added, and the mixture was concentrated under reduced pressure. Water was added to the obtained solid and dissolved by heating to 100 ° C., then cooled to room temperature and recrystallized. The precipitated solid was filtered, washed with water, and dried under reduced pressure to obtain the title compound (10.5 g).
1 H-NMR (DMSO-d 6 ) δ 2.10 (s, 3H), 2.40 (s, 3H), 6.19 (s, 1H), 11.4 (br-s, 1H).
参考例34:2-クロロ-4-メチル-6-(メチルチオ)ピリミジン
Figure JPOXMLDOC01-appb-C000044
 参考例33で得た化合物(442mg)を塩化ホスホリル(4mL)に加え、100℃にて1.5時間攪拌した。室温まで放冷後、減圧濃縮して得られた固体に飽和重曹水と飽和食塩水を加え、酢酸エチルで抽出した。有機層を飽和食塩水にて洗浄後、硫酸ナトリウムで乾燥し、ろ過後、ろ液を減圧濃縮して表題化合物(490mg)を得た。
1H-NMR (CDCl3)δ 2.40 (s, 3H), 2.54 (s, 3H), 6.93 (s, 1H).
Reference Example 34: 2-chloro-4-methyl-6- (methylthio) pyrimidine
Figure JPOXMLDOC01-appb-C000044
The compound (442 mg) obtained in Reference Example 33 was added to phosphoryl chloride (4 mL) and stirred at 100 ° C. for 1.5 hours. The mixture was allowed to cool to room temperature, and concentrated under reduced pressure, saturated aqueous sodium hydrogen carbonate and saturated brine were added, and the mixture was extracted with ethyl acetate. The organic layer was washed with saturated brine, dried over sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (490 mg).
1 H-NMR (CDCl 3 ) δ 2.40 (s, 3H), 2.54 (s, 3H), 6.93 (s, 1H).
参考例35:4-メチル-6-(メチルチオ)ピリミジン-2-カルボニトリル
Figure JPOXMLDOC01-appb-C000045
 参考例34で得た化合物(490mg)のDMSO(3mL)/イソプロピルアルコール(1.7mL)溶液にシアン化ナトリウム(165mg)、1,4-ジアザビシクロ[2.2.2]オクタン(157mg)を加え、室温にて23時間攪拌した。反応液に飽和重曹水、飽和食塩水、水を加え、酢酸エチルで抽出した。有機層を飽和食塩水にて2回洗浄後、硫酸ナトリウムで乾燥し、ろ過後、ろ液を減圧濃縮して表題化合物(436mg)を得た。
1H-NMR (CDCl3)δ 2.45 (s, 3H), 2.56 (s, 3H), 7.15 (s, 1H). 
Reference Example 35: 4-methyl-6- (methylthio) pyrimidine-2-carbonitrile
Figure JPOXMLDOC01-appb-C000045
To a solution of the compound obtained in Reference Example 34 (490 mg) in DMSO (3 mL) / isopropyl alcohol (1.7 mL) was added sodium cyanide (165 mg) and 1,4-diazabicyclo [2.2.2] octane (157 mg). And stirred at room temperature for 23 hours. Saturated aqueous sodium hydrogen carbonate, saturated brine and water were added to the reaction mixture, and the mixture was extracted with ethyl acetate. The organic layer was washed twice with saturated brine, dried over sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (436 mg).
1 H-NMR (CDCl 3 ) δ 2.45 (s, 3H), 2.56 (s, 3H), 7.15 (s, 1H).
参考例36:4-クロロ-6-メチルピリミジン-2-カルボニトリル
Figure JPOXMLDOC01-appb-C000046
 参考例35で得た化合物(165mg)のアセトニトリル(10mL)溶液を0℃に冷却した後、塩化スルフリル(0.4mL)を滴下して加え、0℃にて25分間攪拌した。反応液に飽和炭酸ナトリウム水溶液を加え、クロロホルムで抽出した。有機層を飽和食塩水にて洗浄後、硫酸ナトリウムで乾燥し、ろ過後、ろ液を減圧濃縮して表題化合物(159mg)を得た。
1H-NMR (CDCl3)δ 2.59 (s, 3H), 7.40 (s, 1H).
Reference Example 36: 4-chloro-6-methylpyrimidine-2-carbonitrile
Figure JPOXMLDOC01-appb-C000046
A solution of the compound obtained in Reference Example 35 (165 mg) in acetonitrile (10 mL) was cooled to 0 ° C., and sulfuryl chloride (0.4 mL) was added dropwise thereto, followed by stirring at 0 ° C. for 25 minutes. A saturated aqueous sodium carbonate solution was added to the reaction mixture, and the mixture was extracted with chloroform. The organic layer was washed with saturated brine, dried over sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (159 mg).
1 H-NMR (CDCl 3 ) δ 2.59 (s, 3H), 7.40 (s, 1H).
参考例37:4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-カルボニトリル
Figure JPOXMLDOC01-appb-C000047
 参考例36で得た化合物(154mg)のDMSO(4mL)溶液に5-メチル-ピラゾール-3-アミン(146mg)、N-エチル-ジイソプロピルアミン(0.26mL)を加え、室温にて67時間攪拌した。水を加え、析出した固体をろ過し、水にて洗浄した後、減圧乾燥して表題化合物(162mg)を得た。
1H-NMR (DMSO-d6)δ 2.22 (s, 3H), 2.33 (s, 3H), 5.98 (br-s, 1H), 7.55 (br-s, 1H), 10.3 (s, 1H), 12.1 (s, 1H). 
Reference Example 37: 4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile
Figure JPOXMLDOC01-appb-C000047
To a solution of the compound obtained in Reference Example 36 (154 mg) in DMSO (4 mL), 5-methyl-pyrazol-3-amine (146 mg) and N-ethyl-diisopropylamine (0.26 mL) were added and stirred at room temperature for 67 hours. did. Water was added, and the precipitated solid was filtered, washed with water, and then dried under reduced pressure to obtain the title compound (162 mg).
1 H-NMR (DMSO-d 6 ) δ 2.22 (s, 3H), 2.33 (s, 3H), 5.98 (br-s, 1H), 7.55 (br-s, 1H), 10.3 (s, 1H), 12.1 (s, 1H).
参考例38:2,4-ジブロモ-6-メチル-ピリミジン
Figure JPOXMLDOC01-appb-C000048
 45℃で30% 臭化水素/酢酸溶液(662g)に2,4-ジクロロ-6-メチルピリミジン(40g)の酢酸(40g)、メタンスルホン酸(160g)の溶液を注入した。45℃で15分攪拌後、冷却したトルエン(200g)を加え、内温0℃まで冷却した。内温5℃以下で水(320g)、トリエチルアミン(248g)および37.5%炭酸カリウム水溶液(640g)を順次滴下し、有機層を分取した。水層を酢酸イソプロピル(200g)で2回抽出し、最初の有機層と合わせ、内容量が148gになるまで濃縮した。イソプロピルアルコール(148g)を加え、再び内容量が148gになるまで濃縮することを2回繰り返した。イソプロピルアルコール(40g)を加え50℃に昇温した。水(200g)を50℃で滴下し、0℃まで5時間で冷却、終夜攪拌した。結晶をろ過し、冷却したイソプロピルアルコール/水(30g/120g)で洗浄、40℃で減圧乾燥し、表題化合物(45g)を得た。
LC/MS, tR 0.803 min, obs MS [M+1] 252.9.
Reference Example 38: 2,4-Dibromo-6-methyl-pyrimidine
Figure JPOXMLDOC01-appb-C000048
A solution of 2,4-dichloro-6-methylpyrimidine (40 g) in acetic acid (40 g) and methanesulfonic acid (160 g) was injected into a 30% hydrogen bromide / acetic acid solution (662 g) at 45 ° C. After stirring at 45 ° C. for 15 minutes, cooled toluene (200 g) was added, and the internal temperature was cooled to 0 ° C. Water (320 g), triethylamine (248 g) and 37.5% aqueous potassium carbonate solution (640 g) were successively added dropwise at an internal temperature of 5 ° C. or lower, and the organic layer was separated. The aqueous layer was extracted twice with isopropyl acetate (200 g), combined with the first organic layer, and concentrated to a content of 148 g. Isopropyl alcohol (148 g) was added, and concentration was repeated twice until the internal volume reached 148 g. Isopropyl alcohol (40 g) was added and the temperature was raised to 50 ° C. Water (200 g) was added dropwise at 50 ° C., cooled to 0 ° C. over 5 hours, and stirred overnight. The crystals were filtered, washed with cooled isopropyl alcohol / water (30 g / 120 g), and dried under reduced pressure at 40 ° C. to obtain the title compound (45 g).
LC / MS, t R 0.803 min, obs MS [M + 1] 252.9.
参考例39:2-ブロモ-6-メチル-N-(5-メチル-1H-ピラゾール-3-イル)ピリミジン-4-アミン
Figure JPOXMLDOC01-appb-C000049
 参考例38で得られた2,4-ジブロモ-6-メチル-ピリミジン(40g)、5-メチル-ピラゾール-3-アミン(22g)の2-ピロリドン(120g)溶液に水(280g)と炭酸水素ナトリウム(16g)を加え、60℃で16時間攪拌した。0℃まで6時間かけて冷却、0℃で終夜攪拌し、ろ過して得られた結晶を冷やしたアセトニトリル(80g)で洗浄、40~50℃で減圧乾燥し、表題化合物(36g)を得た。
LC/MS, tR 0.635 min, obs MS [M+1] 270.0.
Reference Example 39: 2-Bromo-6-methyl-N- (5-methyl-1H-pyrazol-3-yl) pyrimidin-4-amine
Figure JPOXMLDOC01-appb-C000049
To a solution of 2,4-dibromo-6-methyl-pyrimidine (40 g) and 5-methyl-pyrazol-3-amine (22 g) obtained in Reference Example 38 in 2-pyrrolidone (120 g), water (280 g) and hydrogen carbonate Sodium (16 g) was added and stirred at 60 ° C. for 16 hours. The mixture was cooled to 0 ° C. over 6 hours, stirred at 0 ° C. overnight, filtered, and the crystals obtained were washed with chilled acetonitrile (80 g) and dried under reduced pressure at 40-50 ° C. to give the title compound (36 g). .
LC / MS, t R 0.635 min, obs MS [M + 1] 270.0.
参考例40:4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-カルボニトリル
Figure JPOXMLDOC01-appb-C000050
 参考例39で得られた2-ブロモ-6-メチル-N-(5-メチル-1H-ピラゾール-3-イル)ピリミジン-4-アミン(30g)をDMF(240g)/イソプロピルアルコール(30g)に懸濁し、シアン化ナトリウム(7.4g)を加え、70℃に昇温した。1,4-ジアザビシクロ[2.2.2]オクタン(4g)を加え、90℃に昇温し、11時間攪拌した。50℃に冷却後、水(900g)を加え、0℃まで冷却した。結晶をろ過し、水(60g)、冷却したアセトニトリル(30g)で洗浄、50℃で減圧乾燥し、表題化合物(16g)を得た。
1H-NMR (400 MHz, DMSO-d6)δ 12.13 (s, 1H), 10.29 (s, 1H), 2.32 (s, 3H), 2.21 (s, 3H).
Reference Example 40: 4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidine-2-carbonitrile
Figure JPOXMLDOC01-appb-C000050
2-Bromo-6-methyl-N- (5-methyl-1H-pyrazol-3-yl) pyrimidin-4-amine (30 g) obtained in Reference Example 39 was added to DMF (240 g) / isopropyl alcohol (30 g). The mixture was suspended, sodium cyanide (7.4 g) was added, and the temperature was raised to 70 ° C. 1,4-diazabicyclo [2.2.2] octane (4 g) was added, the temperature was raised to 90 ° C., and the mixture was stirred for 11 hours. Water (900g) was added after cooling to 50 degreeC, and it cooled to 0 degreeC. The crystals were filtered, washed with water (60 g), cooled acetonitrile (30 g), and dried under reduced pressure at 50 ° C. to give the title compound (16 g).
1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.13 (s, 1H), 10.29 (s, 1H), 2.32 (s, 3H), 2.21 (s, 3H).
実施例1:(3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール
Figure JPOXMLDOC01-appb-C000051
(合成法A)
 参考例4で得た化合物(30mg)のメタノール(2mL)溶液に、水素化ホウ素ナトリウム(3.6mg)を加え、室温で30分攪拌した。反応液に水および酢酸エチルを加え目的物を有機層に抽出した。有機層を飽和食塩水で洗浄後、無水硫酸ナトリウムで乾燥した。有機層を減圧濃縮して得られた残渣をシリカゲルカラムクロマトグラフィー(酢酸エチル/メタノール)により精製して表題化合物(22mg)を得た。
1H-NMR (400 MHz, DMSO-d6)δ 11.98 (s, 1H), 9.88 (s, 1H), 8.22 (d, 1H), 7.51-7.46 (m, 1H), 7.40-7.33 (m, 1H), 7.28-7.25 (m, 1H), 5.77 (d, 1H), 5.56 (d, 1H), 2.19 (s, 3H).
Example 1: (3,4-Difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol
Figure JPOXMLDOC01-appb-C000051
(Synthesis Method A)
Sodium borohydride (3.6 mg) was added to a solution of the compound (30 mg) obtained in Reference Example 4 in methanol (2 mL), and the mixture was stirred at room temperature for 30 minutes. Water and ethyl acetate were added to the reaction solution, and the target product was extracted into the organic layer. The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (ethyl acetate / methanol) to obtain the title compound (22 mg).
1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.98 (s, 1H), 9.88 (s, 1H), 8.22 (d, 1H), 7.51-7.46 (m, 1H), 7.40-7.33 (m, 1H), 7.28-7.25 (m, 1H), 5.77 (d, 1H), 5.56 (d, 1H), 2.19 (s, 3H).
実施例2:(-)-(3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノールおよび
実施例3:(+)-(3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール
(合成法B)
 実施例1で得たラセミ体の(3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール(20mg)をヘキサン:イソプロピルアルコール:メタノール=7:3:5(1.5mL)に溶解し、2回に分けてShimadzu CR-6A CHROMATOPAC,HITACHI L-6000PUMP,Shimadzu SPD-6AよりなるHPLCシステムを用い、カラムDaicel Chiral PAK AD-H,20 mmφ×25 cm、溶媒はヘキサン:イソプロピルアルコール=70:30、流速10mL/minの条件で光学分割を行い、実施例2(10.9mg)および実施例3(10.7mg)の化合物をそれぞれ得た。
実施例2:保持時間19.9 min、[α] 20 =-22.8 (c=0.50, MeOH)
実施例3:保持時間36.3 min、[α] 20 =+21.9 (c=1.00, MeOH)
Example 2: (-)-(3,4-Difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol and
Example 3: (+)-(3,4-Difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol (Synthesis Method B)
The racemic (3,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol (20 mg) obtained in Example 1 was added to hexane: isopropyl. Dissolve in alcohol: methanol = 7: 3: 5 (1.5 mL) and divide into 2 steps using HPLC system consisting of Shimadzu CR-6A CHROMATOPAC, HITACHI L-6000PUMP, Shimadzu SPD-6A and column Daicel Chiral PAK AD -H, 20 mmφ × 25 cm, solvent was hexane: isopropyl alcohol = 70: 30, flow rate was 10 mL / min, and optical resolution was carried out. Example 2 (10.9 mg) and Example 3 (10.7 mg) Each compound was obtained.
Example 2: Retention time 19.9 min, [α] D 20 = −22.8 (c = 0.50, MeOH)
Example 3: Retention time 36.3 min, [α] D 20 = + 21.9 (c = 1.00, MeOH)
実施例4:(+)-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール
Figure JPOXMLDOC01-appb-C000052
(合成法C)
 参考例22と同様にして得た(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トルフルオロフェニル)メタノン(2.0g)のイソプロピルアルコール(10mL)溶液に、窒素雰囲気下で0.01Mのt-ブトキシカリウム イソプロピルアルコール溶液(30mL)および(R)-RUCYTM-Xylbinap(0.2g)を加えた。反応混合物を水素気流下(5気圧)、40℃で3時間撹拌した。反応容器内の水素を窒素で置換後、反応液を減圧濃縮し、残渣をシリカゲルカラムクロマトグラフィー(クロロホルム/メタノール)により精製し、表題化合物(1.45g)を得た。
1H-NMR (400 MHz, DMSO-d6)δ 11.89 (s, 1H), 9.70 (s, 1H), 7.50-7.64 (m, 1H), 7.43-7.48 (m, 1H), 6.09 (s, 1H), 5.87(s, 1H),5.70 (s, 1H), 2.27 (S, 1H), 2.14(s, 3H).
LC/MS,tR 0.58 min,obs MS[M+1] 351.3.
[α] 20 =-29.5 (c=1.0, MeOH)
LC分析(カラムDaicel Chiral PAK AD-H,4.6 mmφ×25 cm、ヘキサン:イソプロピルアルコール:メタノール:トリエチルアミン:酢酸=70:25:5:0.1:0.05、流速1 mL/min)保持時間11.37 min.
Example 4: (+)-(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol
Figure JPOXMLDOC01-appb-C000052
(Synthesis Method C)
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanone obtained in the same manner as in Reference Example 22. To a solution of (2.0 g) in isopropyl alcohol (10 mL) was added 0.01 M t-butoxypotassium isopropyl alcohol solution (30 mL) and (R) -RUCY -Xylbinap (0.2 g) under a nitrogen atmosphere. The reaction mixture was stirred at 40 ° C. for 3 hours under a hydrogen stream (5 atm). After replacing hydrogen in the reaction vessel with nitrogen, the reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (chloroform / methanol) to obtain the title compound (1.45 g).
1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.89 (s, 1H), 9.70 (s, 1H), 7.50-7.64 (m, 1H), 7.43-7.48 (m, 1H), 6.09 (s, 1H), 5.87 (s, 1H), 5.70 (s, 1H), 2.27 (S, 1H), 2.14 (s, 3H).
LC / MS, t R 0.58 min, obs MS [M + 1] 351.3.
[Α] D 20 = −29.5 (c = 1.0, MeOH)
LC analysis (column Daicel Chiral PAK AD-H, 4.6 mmφ × 25 cm, hexane: isopropyl alcohol: methanol: triethylamine: acetic acid = 70: 25: 5: 0.1: 0.05, flow rate 1 mL / min) Retention time 11.37 min.
実施例5:(-)-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール
(合成法D)
 触媒として(S)-RUCYTM-Xylbinapを用い、実施例4の方法と同様にして、(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トルフルオロフェニル)メタノン(100mg)より表題化合物(54mg)を得た。
LC/MS,tR 0.58min,obs MS[M+1]351.3.
LC分析(カラムDaicel Chiral PAK AD-H,4.6 mmφ×25 cm、ヘキサン:イソプロピルアルコール:メタノール:トリエチルアミン:酢酸=70:25:5:0.1:0.05、流速1 mL/min)保持時間8.66 min.
Example 5: (−)-(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol ( Synthesis method D)
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidine-2-) was used in the same manner as in Example 4 using (S) -RUCY -Xylbinap as the catalyst. Yl) (2,4,5-trifluorophenyl) methanone (100 mg) gave the title compound (54 mg).
LC / MS, t R 0.58min, obs MS [M + 1] 351.3.
LC analysis (column Daicel Chiral PAK AD-H, 4.6 mmφ × 25 cm, hexane: isopropyl alcohol: methanol: triethylamine: acetic acid = 70: 25: 5: 0.1: 0.05, flow rate 1 mL / min) Retention time 8.66 min.
実施例6:(-)-(6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール
および
実施例7:(+)-(6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール
Figure JPOXMLDOC01-appb-C000053
 参考例11で得た化合物(200mg)のメタノール(5mL)溶液に、水素化ホウ素ナトリウム(23mg)を加え、室温で3時間20分攪拌した。反応液に水および酢酸エチルを加え、目的物を有機層に抽出し、有機層を飽和食塩水にて洗浄後、硫酸ナトリウムで乾燥した。有機層を減圧濃縮して得られた残渣をアミノシリカゲルカラムクロマトグラフィー(酢酸エチル/メタノール)にて精製してラセミ体の(6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール(130mg)を得た。
LC/MS,tR 0.63 min,MS[M+1] 334.9.
 同様にして得られたラセミ体の(6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール(28.7g)をDAICEL社にて光学分割を行い、実施例6(10.3g)および実施例7(9.6g)の化合物をそれぞれ得た。
 実施例6および実施例7の化合物を、カラムDaicel Chiral PAK OJ-H,4.6 mmφ×25 cm、溶媒はヘキサン:エタノール:メタノール:エチレンジアミン=40:40:20:0.1、流速1.0 mL/minで分析した保持時間を以下に示す。
実施例6:3.69 min
実施例7:5.90 min
Example 6: (−)-(6-((5-Methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) (3,4,5-trifluorophenyl) methanol
and
Example 7: (+)-(6-((5-Methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) (3,4,5-trifluorophenyl) methanol
Figure JPOXMLDOC01-appb-C000053
To a solution of the compound (200 mg) obtained in Reference Example 11 in methanol (5 mL), sodium borohydride (23 mg) was added and stirred at room temperature for 3 hours and 20 minutes. Water and ethyl acetate were added to the reaction solution, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine and dried over sodium sulfate. The organic layer was concentrated under reduced pressure, and the resulting residue was purified by amino silica gel column chromatography (ethyl acetate / methanol) to obtain racemic (6-((5-methyl-1H-pyrazol-3-yl) amino) Pyridin-2-yl) (3,4,5-trifluorophenyl) methanol (130 mg) was obtained.
LC / MS, t R 0.63 min , MS [M + 1] 334.9.
The racemic (6-((5-methyl-1H-pyrazol-3-yl) amino) pyridin-2-yl) (3,4,5-trifluorophenyl) methanol (28.7 g) was obtained in the same manner. ) Were optically resolved by DAICEL to obtain the compounds of Example 6 (10.3 g) and Example 7 (9.6 g).
The compounds of Example 6 and Example 7 were analyzed by column Daicel Chiral PAK OJ-H, 4.6 mmφ × 25 cm, the solvent was hexane: ethanol: methanol: ethylenediamine = 40: 40: 20: 0.1, and the flow rate was 1.0 mL / min. The retention times obtained are shown below.
Example 6: 3.69 min
Example 7: 5.90 min
実施例8~44
 対応する原料化合物を用いて実施例1、2、3、4又は5と同様の方法(合成法A、C、D)により、下記表に示す実施例8~44の化合物を得た。キラルカラムによるLC分析を以下の条件で行い、その保持時間を示す。
カラムDaicel Chiral PAK AD-H,4.6 mmφ×25 cm、ヘキサン:イソプロピルアルコール:メタノール:トリエチルアミン:酢酸=70:25:5:0.1:0.05、流速1 mL/min
Examples 8-44
The compounds of Examples 8 to 44 shown in the following table were obtained in the same manner as in Example 1, 2, 3, 4 or 5 (Synthesis methods A, C and D) using the corresponding starting compounds. LC analysis using a chiral column is performed under the following conditions, and the retention time is shown.
Column Daicel Chiral PAK AD-H, 4.6 mmφ × 25 cm, hexane: isopropyl alcohol: methanol: triethylamine: acetic acid = 70: 25: 5: 0.1: 0.05, flow rate 1 mL / min
Figure JPOXMLDOC01-appb-C000054
Figure JPOXMLDOC01-appb-C000054
Figure JPOXMLDOC01-appb-T000055
Figure JPOXMLDOC01-appb-T000055
Figure JPOXMLDOC01-appb-T000056
Figure JPOXMLDOC01-appb-T000056
Figure JPOXMLDOC01-appb-T000057
Figure JPOXMLDOC01-appb-T000057
Figure JPOXMLDOC01-appb-T000058
Figure JPOXMLDOC01-appb-T000058
Figure JPOXMLDOC01-appb-T000059
Figure JPOXMLDOC01-appb-T000059
Figure JPOXMLDOC01-appb-T000060
Figure JPOXMLDOC01-appb-T000060
実施例45:(3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタン-d-オール
Figure JPOXMLDOC01-appb-C000061
 参考例4で得た化合物(47mg)のメタノール(0.8mL)溶液に、重水素化ホウ素ナトリウム(7.4mg)を加え、室温で2時間攪拌した。反応液に3規定HClおよび5規定NaOHを加え、反応液のpHを8に調整した後、減圧濃縮した。残渣に水を加え、析出した固体をろ取し、水、ヘキサンで洗浄した後、減圧乾燥して表題化合物(44mg)を得た。
1H-NMR(400 MHz, DMSO-d6)δ11.9 (s, 1H), 9.84 (s, 1H), 8.21 (s, 1H), 7.50-7.26 (m, 3H), 5.84 (s, 1H), 5.53 (d, 1H),2.18 (s, 3H).
Example 45: (3,4-Difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methane-d-ol
Figure JPOXMLDOC01-appb-C000061
To a solution of the compound obtained in Reference Example 4 (47 mg) in methanol (0.8 mL) was added sodium deuteride (7.4 mg), and the mixture was stirred at room temperature for 2 hours. 3N HCl and 5N NaOH were added to the reaction solution to adjust the pH of the reaction solution to 8, and then concentrated under reduced pressure. Water was added to the residue, and the precipitated solid was collected by filtration, washed with water and hexane, and then dried under reduced pressure to obtain the title compound (44 mg).
1 H-NMR (400 MHz, DMSO-d 6 ) δ11.9 (s, 1H), 9.84 (s, 1H), 8.21 (s, 1H), 7.50-7.26 (m, 3H), 5.84 (s, 1H ), 5.53 (d, 1H), 2.18 (s, 3H).
実施例46:1-(3-メトキシフェニル)-1-(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)エタン-1-オール
Figure JPOXMLDOC01-appb-C000062
 参考例3で得た化合物(20mg)のTHF(1mL)溶液にメチルマグネシウムブロミド(0.98M、0.2mL)を加え、室温下20時間攪拌した。その後、メチルマグネシウムブロミド(0.98M、1.5mL)を追加し、室温下20時間攪拌した。反応液に塩化アンモニウム飽和水溶液および酢酸エチルを加え、目的物を有機層に抽出した後、有機層を飽和食塩水で洗浄し、無水硫酸マグネシウムを加えて乾燥させた。有機層を減圧濃縮し、得られた残渣をシリカゲルカラムクロマトグラフィー(酢酸エチル)で精製し、表題化合物(7.8mg)を得た。
1H-NMR(300 MHz, DMSO-d6)δ12.10 (s, 1H), 10.00 (s, 1H), 8.29 (s, 1H), 7.12-7.30 (m, 3H), 6.82 (d, 1H), 5.72 (s, 1H), 3.77 (s, 3H), 2.28 (s, 3H), 1.87 (s, 3H).
Example 46: 1- (3-methoxyphenyl) -1- (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) ethane-1-ol
Figure JPOXMLDOC01-appb-C000062
Methyl magnesium bromide (0.98M, 0.2 mL) was added to a THF (1 mL) solution of the compound (20 mg) obtained in Reference Example 3, and the mixture was stirred at room temperature for 20 hours. Thereafter, methylmagnesium bromide (0.98M, 1.5 mL) was added, and the mixture was stirred at room temperature for 20 hours. A saturated aqueous ammonium chloride solution and ethyl acetate were added to the reaction solution, and the target product was extracted into an organic layer. The organic layer was washed with saturated brine, and dried over anhydrous magnesium sulfate. The organic layer was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (ethyl acetate) to obtain the title compound (7.8 mg).
1 H-NMR (300 MHz, DMSO-d 6 ) δ12.10 (s, 1H), 10.00 (s, 1H), 8.29 (s, 1H), 7.12-7.30 (m, 3H), 6.82 (d, 1H ), 5.72 (s, 1H), 3.77 (s, 3H), 2.28 (s, 3H), 1.87 (s, 3H).
実施例47~52
 対応する原料化合物を用いて実施例46と同様の方法により、下記表に示す実施例47~52の化合物を得た。
Figure JPOXMLDOC01-appb-C000063
Figure JPOXMLDOC01-appb-T000064
Examples 47-52
The compounds of Examples 47 to 52 shown in the following table were obtained in the same manner as in Example 46 using the corresponding starting compounds.
Figure JPOXMLDOC01-appb-C000063
Figure JPOXMLDOC01-appb-T000064
実施例53~70
 対応する原料化合物を用いて実施例1と同様の方法(合成法A)により、下記表に示す実施例53~70の化合物を得た。
Figure JPOXMLDOC01-appb-C000065
Figure JPOXMLDOC01-appb-T000066
Examples 53-70
The compounds of Examples 53 to 70 shown in the following table were obtained in the same manner as in Example 1 (Synthesis Method A) using the corresponding starting compounds.
Figure JPOXMLDOC01-appb-C000065
Figure JPOXMLDOC01-appb-T000066
Figure JPOXMLDOC01-appb-T000067
Figure JPOXMLDOC01-appb-T000067
実施例71~86
 対応する原料化合物を用いて実施例46と同様の方法により、下記表に示す実施例71~86の化合物を得た。
Figure JPOXMLDOC01-appb-C000068
Figure JPOXMLDOC01-appb-T000069
Examples 71-86
The compounds of Examples 71 to 86 shown in the following table were obtained in the same manner as in Example 46 using the corresponding starting compounds.
Figure JPOXMLDOC01-appb-C000068
Figure JPOXMLDOC01-appb-T000069
Figure JPOXMLDOC01-appb-T000070
Figure JPOXMLDOC01-appb-T000070
実施例87:(+)-{4-メチル-6-[(5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,4,5-トリフルオロフェニル)メタノール エタノール和物
Figure JPOXMLDOC01-appb-C000071
 (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トルフルオロフェニル)メタノン(3.0g)のイソプロピルアルコール(60mL)/テトラヒドロフラン(60mL)溶液に、窒素雰囲気下でt-ブトキシカリウム(58.2mg)および(R)-RUCYTM-Xylbinap(40.9mg)を加えた。反応混合物を水素気流下(5気圧)、40℃で2.5時間撹拌した。反応容器内の水素を窒素で置換後、反応液に3-メルカプトプロピルシリカゲル(4.0g)、活性炭(2.0g)を加えた。1時間撹拌後、セライトろ過し、残渣をエタノールで洗浄した。ろ液をエバポレーターで濃縮し、粗生成物(3.1g)を取得した。これにエタノール(5mL)を加え、超音波洗浄機で撹拌し、固体の析出を確認した。この混合物を90℃で30分加熱撹拌し、その後、ゆっくりと室温に冷却し、さらに氷浴で5℃に冷却した。析出した結晶をろ取し、さらに冷却したエタノール(2mL)で2度洗浄後、60℃で3時間減圧乾燥し、表題化合物(2.39g)を得た。
Example 87: (+)-{4-Methyl-6-[(5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,4,5-trifluorophenyl) methanol ethanol Japanese
Figure JPOXMLDOC01-appb-C000071
(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanone (3.0 g) in isopropyl alcohol ( To a solution of 60 mL) / tetrahydrofuran (60 mL) were added potassium t-butoxy (58.2 mg) and (R) -RUCY -Xylbinap (40.9 mg) under a nitrogen atmosphere. The reaction mixture was stirred at 40 ° C. for 2.5 hours under a hydrogen stream (5 atm). After replacing hydrogen in the reaction vessel with nitrogen, 3-mercaptopropyl silica gel (4.0 g) and activated carbon (2.0 g) were added to the reaction solution. The mixture was stirred for 1 hour, filtered through Celite, and the residue was washed with ethanol. The filtrate was concentrated with an evaporator to obtain a crude product (3.1 g). Ethanol (5 mL) was added thereto, and the mixture was stirred with an ultrasonic cleaner to confirm solid precipitation. The mixture was heated and stirred at 90 ° C. for 30 minutes, then slowly cooled to room temperature, and further cooled to 5 ° C. in an ice bath. The precipitated crystals were collected by filtration, further washed twice with cooled ethanol (2 mL), and dried under reduced pressure at 60 ° C. for 3 hours to obtain the title compound (2.39 g).
実施例88:(+)-{4-メチル-6-[(5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,4,5-トリフルオロフェニル)メタノール
Figure JPOXMLDOC01-appb-C000072
 実施例87で得た(+)-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール エタノール和物(0.40g)を水(8mL)に懸濁した。この懸濁液を、90℃で1時間加熱撹拌した。室温に冷却後、析出した結晶をろ取した。結晶をさらに水で3回洗浄し、減圧乾燥し、表題化合物(0.34g)を得た。
Example 88: (+)-{4-Methyl-6-[(5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,4,5-trifluorophenyl) methanol
Figure JPOXMLDOC01-appb-C000072
(+)-(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) obtained in Example 87 Methanol Ethanolate (0.40 g) was suspended in water (8 mL). This suspension was heated and stirred at 90 ° C. for 1 hour. After cooling to room temperature, the precipitated crystals were collected by filtration. The crystals were further washed with water three times and dried under reduced pressure to obtain the title compound (0.34 g).
実施例89:(+)-{4-メチル-6-[(5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,4,5-トリフルオロフェニル)メタノール
Figure JPOXMLDOC01-appb-C000073
 実施例87と同様にして得た(+)-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール エタノール和物(7.10g)を水(71mL)に懸濁した。実施例88で得た種晶を加え、90℃で6時間加熱撹拌した。室温に冷却後、析出した結晶をろ取した。結晶をさらに水で3回洗浄し、減圧乾燥し、表題化合物(6.08g)を得た。
Example 89: (+)-{4-Methyl-6-[(5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,4,5-trifluorophenyl) methanol
Figure JPOXMLDOC01-appb-C000073
(+)-(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-tri) obtained in the same manner as in Example 87. Fluorophenyl) methanol Ethanolate (7.10 g) was suspended in water (71 mL). The seed crystal obtained in Example 88 was added, and the mixture was heated with stirring at 90 ° C. for 6 hours. After cooling to room temperature, the precipitated crystals were collected by filtration. The crystals were further washed with water three times and dried under reduced pressure to obtain the title compound (6.08 g).
 粉末X線回折パターンのチャートを図1、その主要ピークを下表に示す。また、DSC-TGAのチャートを図2に示す。DSC-TGAのチャートより、吸熱ピークで重量変化が見られなかったため、無水物結晶であることが確認された。
Figure JPOXMLDOC01-appb-T000074
A chart of the powder X-ray diffraction pattern is shown in FIG. 1, and its main peak is shown in the table below. A DSC-TGA chart is shown in FIG. From the DSC-TGA chart, since no weight change was observed at the endothermic peak, it was confirmed to be an anhydrous crystal.
Figure JPOXMLDOC01-appb-T000074
実施例90:(+)-{4-メチル-6-[(5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,4,5-トリフルオロフェニル)メタノール リン酸塩Example 90: (+)-{4-Methyl-6-[(5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,4,5-trifluorophenyl) methanol phosphorus Acid salt
Figure JPOXMLDOC01-appb-C000075
Figure JPOXMLDOC01-appb-C000075
 実施例87で得た(+)-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール エタノール和物(100mg)のメタノール(0.5mL)懸濁液にリン酸(21μL)を加え、50℃で1時間撹拌した。さらに室温で21時間撹拌し、結晶の析出を確認した。酢酸エチル(2mL)を加え、1.5時間撹拌した。析出した固体をろ取し、残渣を酢酸エチル(0.5mL)で3回洗浄後、50℃で減圧乾燥して表題化合物(111mg)を得た。 (+)-(4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) obtained in Example 87 Phosphoric acid (21 μL) was added to a methanol (0.5 mL) suspension of methanol-ethanol product (100 mg), and the mixture was stirred at 50 ° C. for 1 hour. Further, the mixture was stirred at room temperature for 21 hours to confirm crystal precipitation. Ethyl acetate (2 mL) was added and stirred for 1.5 hours. The precipitated solid was collected by filtration, and the residue was washed three times with ethyl acetate (0.5 mL) and dried under reduced pressure at 50 ° C. to obtain the title compound (111 mg).
 得られた(+)-{4-メチル-6-[(5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,4,5-トリフルオロフェニル)メタノール リン酸塩(50mg)をメタノールに溶解し、メタノールの半量のエタノールと、エタノールと同量のイソプロピルアルコールを加えた。室温下ゆっくりと溶媒を揮発させ、太目の針状結晶が析出したところで、結晶をろ取し、X線結晶解析を行った。
解析結果を下記の5つの表に示す。また、解析結果より本化合物の絶対構造は(S)-体であると決定した。立体構造を図3に示す。
Figure JPOXMLDOC01-appb-T000076
Figure JPOXMLDOC01-appb-T000077
Figure JPOXMLDOC01-appb-T000078
Figure JPOXMLDOC01-appb-T000079
Figure JPOXMLDOC01-appb-T000080
 また、本化合物(実施例90:(+)-{4-メチル-6-[(5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,4,5-トリフルオロフェニル)メタノール リン酸塩)の絶対構造が(S)-体であるとわかったため、対応する実施例4および実施例89の化合物((+)-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール)、ならびに実施例87の化合物((+)-{4-メチル-6-[(5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,4,5-トリフルオロフェニル)メタノール エタノール和物)もそれぞれ(S)-体であると決定した。
The obtained (+)-{4-methyl-6-[(5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,4,5-trifluorophenyl) methanol phosphoric acid The salt (50 mg) was dissolved in methanol, and half of ethanol in methanol and isopropyl alcohol in the same amount as ethanol were added. The solvent was volatilized slowly at room temperature, and when thick needle-like crystals were precipitated, the crystals were collected by filtration and subjected to X-ray crystal analysis.
The analysis results are shown in the following five tables. From the analysis results, the absolute structure of this compound was determined to be (S) -isomer. The three-dimensional structure is shown in FIG.
Figure JPOXMLDOC01-appb-T000076
Figure JPOXMLDOC01-appb-T000077
Figure JPOXMLDOC01-appb-T000078
Figure JPOXMLDOC01-appb-T000079
Figure JPOXMLDOC01-appb-T000080
In addition, this compound (Example 90: (+)-{4-methyl-6-[(5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,4,5-tri Since the absolute structure of (fluorophenyl) methanol phosphate) was found to be the (S) -form, the corresponding compounds ((+)-(4-methyl-6-((5- Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol) and the compound of Example 87 ((+)-{4-methyl-6- [(5-Methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,4,5-trifluorophenyl) methanol ethanolate) was also determined to be the (S) -isomer, respectively. .
実施例91:(+)-{4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,3,4-トリフルオロフェニル)メタノールの結晶化および粉末X線結晶解析
Figure JPOXMLDOC01-appb-C000081
 実施例32で得た(+)-{4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ]ピリミジン-2-イル}(2,3,4-トリフルオロフェニル)メタノール(1.33g)に対し、ヘキサン/クロロホルム=1/1(4mL)を加え、1日撹拌した。結晶をろ取、ヘキサン/クロロホルム=1/1で洗浄後、乾燥して白色結晶(1.29g)を得た。
Example 91: (+)-{4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,3,4-trifluorophenyl) methanol Crystallization and powder X-ray crystallography
Figure JPOXMLDOC01-appb-C000081
(+)-{4-Methyl-6-((5-methyl-1H-pyrazol-3-yl) amino] pyrimidin-2-yl} (2,3,4-trifluorophenyl) obtained in Example 32 Hexane / chloroform = 1/1 (4 mL) was added to methanol (1.33 g), and the mixture was stirred for 1 day.The crystals were collected by filtration, washed with hexane / chloroform = 1/1, dried and dried to give white crystals (1 .29 g) was obtained.
 粉末X線回折パターンのチャートを図4、その主要ピークを下表に示す。また、DSC-TGAのチャートを図5に示す。DSC-TGAのチャートより、吸熱ピークで重量変化が見られなかったため、無水物結晶であることが確認された。
Figure JPOXMLDOC01-appb-T000082
A chart of the powder X-ray diffraction pattern is shown in FIG. A DSC-TGA chart is shown in FIG. From the DSC-TGA chart, since no weight change was observed at the endothermic peak, it was confirmed to be an anhydrous crystal.
Figure JPOXMLDOC01-appb-T000082
薬理試験
 以下に本発明の代表化合物についての薬理試験方法およびその結果について示すが、本発明はこれらの試験例に限定されるものではない。
Pharmacological test Although the pharmacological test method and the results of the representative compounds of the present invention are shown below, the present invention is not limited to these test examples.
試験例1:細胞増殖抑制実験
 NCI-H23細胞をATCC(アメリカ培養細胞系統保存機関)より入手した。NCI-H23は10% FCS、1%ペニシリン/ストレプトマイシン含有RPMI1640培地、HCT116は10% FCS、1%ペニシリン/ストレプトマイシン含有DMEM培地にて37℃、5% CO2存在下にて培養した。
 96 wellプレートに500-3000 cells/wellで細胞を播種し、DMSO終濃度が0.1 %となるように被験物質を添加し、4-7日培養した。その後、プレストブルー(ライフテクノロジーズ)を用いて生細胞数を計測し、各被験物質の50%細胞増殖を抑制する濃度(Cytotoxicity IC50値;μM)を算出し、本発明化合物ががん細胞に対する増殖抑制作用を有することを確認した。
Test Example 1: Cell Growth Inhibition Experiment NCI-H23 cells were obtained from ATCC (American Cultured Cell Line Storage Organization). NCI-H23 was cultured in RPMI 1640 medium containing 10% FCS and 1% penicillin / streptomycin, and HCT116 was cultured in DMEM medium containing 10% FCS and 1% penicillin / streptomycin in the presence of 37 ° C. and 5% CO 2 .
Cells were seeded on a 96-well plate at 500-3000 cells / well, the test substance was added so that the final DMSO concentration was 0.1%, and the cells were cultured for 4-7 days. Thereafter, the number of viable cells was measured using Presto Blue (Life Technologies), and the concentration (Cytotoxicity IC 50 value; μM) of each test substance that inhibits cell growth was calculated. It was confirmed to have a growth inhibitory action.
試験例2:ラット肝実質細胞を用いた細胞毒性試験
 6週齢のCrl:CD(SD)系雄ラットより、ラット肝実質細胞を単離した。懸濁液中の生細胞と死細胞数をトリパンブルー染色にてカウントし、viabilityを算出した後、1.0×104 cell/100μL肝細胞懸濁液を調製した。この懸濁液を60μL/wellでType 1コラーゲンコート済み384 well プレート(コーニング)へ添加し、37℃、5% CO2存在下で一晩培養した。384 wellの細胞培養プレートから培地を除き、被験物質含有培養液を50μL/well添加した(n=4)。添加終了後は順次37℃、5% CO2存在下で培養を継続した。2日後、WST-8試薬(Cell Counting Lit-8,同仁化学研究所)を5μL/well添加し攪拌後、37℃、5% CO2存在下で約3時間インキュベートした。インキュベート後、0.1N HClを15μL/well添加し発色を停止し、波長450 nmで吸光度を測定、生細胞数を計測し、各被験物質の50%細胞増殖を抑制する濃度(Cytotoxicity IC50値;μM)を算出した。本発明化合物の肝細胞に対する毒性が弱いことが確認できた。
Test Example 2: Cytotoxicity test using rat liver parenchymal cells Rat liver parenchymal cells were isolated from 6-week-old Crl: CD (SD) male rats. The number of viable cells and dead cells in the suspension was counted by trypan blue staining and viability was calculated, and then a 1.0 × 10 4 cell / 100 μL hepatocyte suspension was prepared. This suspension was added to a Type 1 collagen-coated 384 well plate (Corning) at 60 μL / well and cultured overnight at 37 ° C. in the presence of 5% CO 2 . The medium was removed from the 384-well cell culture plate, and 50 μL / well of a test substance-containing culture solution was added (n = 4). After completion of the addition, the culture was continued successively in the presence of 37 ° C. and 5% CO 2 . Two days later, 5 μL / well of WST-8 reagent (Cell Counting Lit-8, Dojindo Laboratories) was added and stirred, and incubated at 37 ° C. in the presence of 5% CO 2 for about 3 hours. After incubation, 0.1N HCl was added at 15 μL / well to stop color development, the absorbance was measured at a wavelength of 450 nm, the number of viable cells was counted, and the concentration at which each test substance inhibits 50% cell growth (Cytotoxicity IC 50 value; μM) was calculated. It was confirmed that the toxicity of the compound of the present invention to hepatocytes was weak.
試験例3:化合物の代謝安定性試験
 25 mM Kpi(pH 7.4)39.6 mlに、0.4 mlのヒト肝ミクロソーム(Xenotech社製、約20 mg protein/ml)を加え、ミクロソーム溶液を調製した。1 mM被検化合物のDMSO溶液10μlをアセトニトリルで100倍に希釈した。この溶液5μlに、NADPH 300 mgを125 mM Kpi(pH 7.4)55.2 mlに溶解(6.5 mM)して調製したCofactor液250μlを添加混合して中間希釈液を調製した。96ウェルプレート上でTecan社製スクリーニングロボットを用いて、中間希釈液50μlずつを2ウェルに取り、これにそれぞれミクロソーム溶液50μlを添加混合し、37℃で30分間振とうしながらインキュベートし、反応サンプルを調製した。別途、中間希釈液50μlずつを2ウェルに取り、ミクロソーム溶液を添加せずに同様にインキュベートし、未反応サンプルを調製した。インキュベート終了後、反応サンプル、未反応サンプルのウェルに対して400μlずつメタノールを添加調製した。さらに、未反応サンプルのウェルに対しては、メタノール添加の後ミクロソーム溶液50μlを添加混合調製した。-20℃で30分間静置後、遠心し、上清10μlをLC-MS/MSシステム(島津製作所製HPLC、AB Sciex社製API 4000)に注入し、反応サンプル2ウェル及び未反応サンプル2ウェルの計4ウェルを測定し、それぞれのクロマトピーク面積値の平均値を算出した。この値を用いて、自然対数(LN)を用いた以下の式により、化合物のクリアランスを求めた。
化合物のクリアランス=-LN{(反応サンプル平均値)÷(未反応サンプル平均値)}/30/0.1
この結果より、本発明化合物のミクロソーム中での安定性が向上していることを確認した。
Test Example 3: Metabolic stability test of compound To 39.6 ml of 25 mM Kpi (pH 7.4), 0.4 ml of human liver microsome (Xenotech, approximately 20 mg protein / ml) was added to prepare a microsome solution. 10 μl of 1 mM test compound in DMSO was diluted 100-fold with acetonitrile. An intermediate diluted solution was prepared by adding 250 μl of Cofactor solution prepared by dissolving (6.5 mM) NADPH 300 mg in 55.2 ml of 125 mM Kpi (pH 7.4) to 5 μl of this solution. Using a Tecan screening robot on a 96-well plate, take 50 μl of the intermediate dilution in 2 wells, add 50 μl of the microsomal solution to each well, and incubate with shaking at 37 ° C. for 30 minutes. Was prepared. Separately, 50 μl of the intermediate dilution was taken in 2 wells and incubated in the same manner without adding the microsome solution to prepare an unreacted sample. After incubation, 400 μl of methanol was added to each well of the reaction sample and unreacted sample. Furthermore, for the wells of the unreacted sample, 50 μl of microsome solution was added and mixed after methanol addition. Centrifuge for 30 minutes at -20 ° C, centrifuge, and inject 10 µl of the supernatant into an LC-MS / MS system (Shimadzu HPLC, AB Sciex API 4000). 2 reaction samples and 2 unreacted samples A total of 4 wells were measured, and the average value of each chromatographic peak area value was calculated. Using this value, the clearance of the compound was determined by the following equation using the natural logarithm (LN).
Compound clearance = −LN {(average value of reaction sample) ÷ (average value of unreacted sample)} / 30 / 0.1
From this result, it was confirmed that the stability of the compound of the present invention in microsomes was improved.
試験例4:hERG阻害試験
 培養したhERG遺伝子安定発現CHO細胞株細胞に、DMSO終濃度が0.0135~0.5 %となるように被験物質を添加した。そのhERG電流をQPatch HT(Sophion社)を用いて測定し、各被験物質が50 % hERG電流を抑制する濃度(IC50値;μM)を算出し、本発明化合物のhERG阻害活性が弱いことを確認した。
Test Example 4: hERG inhibition test A test substance was added to the cultured hERG gene stably expressing CHO cell line so that the final DMSO concentration was 0.0135 to 0.5%. The hERG current was measured using QPatch HT (Sophion), the concentration at which each test substance suppresses 50% hERG current (IC 50 value; μM) was calculated, and the hERG inhibitory activity of the compound of the present invention was weak. confirmed.
 実施例で得られた化合物について、試験例1~試験例4に示す試験を行った。結果について下記表に示す。
Figure JPOXMLDOC01-appb-T000083
Tests shown in Test Examples 1 to 4 were performed on the compounds obtained in the examples. The results are shown in the following table.
Figure JPOXMLDOC01-appb-T000083
Figure JPOXMLDOC01-appb-T000084
Figure JPOXMLDOC01-appb-T000084
Figure JPOXMLDOC01-appb-T000085
Figure JPOXMLDOC01-appb-T000085
試験例5:キナーゼ阻害実験
 アッセイバッファー(20 mM HEPES,0.01 % Triton X-100,2 mM DTT,pH 7.5)にて調製した5μLの4倍濃度被験物質溶液、5μLの4倍濃度基質/ATP/金属溶液および10μLの2倍濃度キナーゼ溶液をポリプロピレン製384ウェルプレートのウェル内で混合し、室温にて1時間反応させた。60μLのTermination Buffer(QuickScout Screening Assist MSA;Carna Biosciences)を添加して反応を停止させた。反応溶液中の基質ペプチドとリン酸化ペプチドをLabChip system(Perkin Elmer)にて分離、定量した。キナーゼ反応は基質ペプチドピーク高さ(S)とリン酸化ペプチドピーク高さ(P)から計算される生成物比(P/(P+S))にて評価した。また、データ解析において、コントロールウエルの平均シグナルを0 %阻害、バックグラウンドウエル(酵素非添加)の平均シグナルを100 %阻害とし、各被検体物質ウエルの平均シグナルから阻害率を計算した。結果により、本発明化合物がキナーゼ阻害活性を有することを確認した。
Test Example 5: Kinase inhibition experiment assay buffer (20 mM HEPES, 0.01% Triton X-100, 2 mM DTT, pH 7.5) 5 μL of 4-fold concentration test substance solution, 5 μL of 4-fold concentration substrate / ATP / The metal solution and 10 μL of the double concentration kinase solution were mixed in the wells of a polypropylene 384-well plate and reacted at room temperature for 1 hour. The reaction was stopped by adding 60 μL Termination Buffer (QuickScout Screening Assist MSA; Carna Biosciences). The substrate peptide and phosphorylated peptide in the reaction solution were separated and quantified by LabChip system (Perkin Elmer). The kinase reaction was evaluated by the product ratio (P / (P + S)) calculated from the substrate peptide peak height (S) and the phosphorylated peptide peak height (P). In the data analysis, the average signal of the control well was 0% inhibition, the average signal of the background well (no enzyme added) was 100% inhibition, and the inhibition rate was calculated from the average signal of each test substance well. From the results, it was confirmed that the compound of the present invention has kinase inhibitory activity.
 実施例で得られた化合物について、試験例5に示す試験を行った。試験結果について下記表に示す。
Figure JPOXMLDOC01-appb-T000086
Tests shown in Test Example 5 were performed on the compounds obtained in the examples. The test results are shown in the following table.
Figure JPOXMLDOC01-appb-T000086
Figure JPOXMLDOC01-appb-T000087
Figure JPOXMLDOC01-appb-T000087
Figure JPOXMLDOC01-appb-T000088
Figure JPOXMLDOC01-appb-T000088
試験例6:HCT-116ヒト結腸腺癌細胞担癌マウスにおける腫瘍増殖抑制実験
 HCT-116細胞(ATCC)をHBSSに懸濁し、5週齢の雌性BALB/cAnNCrj-nu/nuマウス(日本チャールス・リバー)の腹側部皮下に、3×106個移植した。移植7日後から、0.5 %メチルセルロース溶液(和光純薬)に懸濁した被験物質を、1日2回13~15日間経口投与し、被験物質投与群とした。ビークル投与群には、0.5 %メチルセルロース溶液を同様に投与した。各群の匹数は7~8匹とした。投与開始後、週1~3回、腫瘍径および体重を測定した。腫瘍体積は、以下の式から算出した。
腫瘍体積(mm3)=長径(mm)×短径(mm)×短径(mm)×1/2
各投与群の平均腫瘍体積を用いて、以下の式から腫瘍増殖抑制率を算出した。
腫瘍増殖抑制率(%)=100-100×(T-T0)/(C-C0)
T:被験物質投与終了後の平均腫瘍体積
T0:被験物質投与開始時の平均腫瘍体積
C:ビークル投与終了後の平均腫瘍体積
C0:ビークル投与開始時の平均腫瘍体積
各投与群の平均体重を用いて、以下の式から体重変化率を算出した。
体重変化率(%)=100×(T/T0)/(C/C0)
T:被験物質投与終了後の平均体重
T0:被験物質投与開始時の平均体重
C:ビークル投与終了後の平均体重
C0:ビークル投与開始時の平均体重
Test Example 6: Tumor Growth Suppression Experiment in HCT-116 Human Colon Adenocarcinoma Cell Carrying Mice HCT-116 cells (ATCC) were suspended in HBSS and 5-week-old female BALB / cAnNCrj-nu / nu mice (Charles Japan) 3 × 10 6 cells were transplanted subcutaneously on the ventral part of the river. From 7 days after transplantation, the test substance suspended in a 0.5% methylcellulose solution (Wako Pure Chemical Industries) was orally administered twice a day for 13 to 15 days to obtain a test substance administration group. A 0.5% methylcellulose solution was similarly administered to the vehicle administration group. The number of animals in each group was 7-8. Tumor diameter and body weight were measured 1-3 times a week after the start of administration. Tumor volume was calculated from the following equation.
Tumor volume (mm 3 ) = major axis (mm) x minor axis (mm) x minor axis (mm) x 1/2
The tumor growth inhibition rate was calculated from the following formula using the average tumor volume of each administration group.
Tumor growth inhibition rate (%) = 100-100 × (T-T0) / (C-C0)
T: Average tumor volume after administration of test substance
T0: Average tumor volume at the start of test substance administration
C: Average tumor volume after vehicle administration
C0: Average tumor volume at the start of vehicle administration The weight change rate was calculated from the following formula using the average body weight of each administration group.
Weight change rate (%) = 100 x (T / T0) / (C / C0)
T: Average body weight after administration of test substance
T0: Average body weight at the start of test substance administration
C: Average body weight after vehicle administration
C0: Average body weight at the start of vehicle administration
 実施例で得られた化合物について、試験例6に示す試験を行った。結果について下記表に示す。
Figure JPOXMLDOC01-appb-T000089
Tests shown in Test Example 6 were performed on the compounds obtained in the examples. The results are shown in the following table.
Figure JPOXMLDOC01-appb-T000089
試験例7:HCC1806ヒト乳癌細胞担癌マウスにおける腫瘍増殖抑制実験
 HCC1806細胞(ATCC)をHBSSに懸濁し、5週齢の雌性BALB/cAnNCrj-nu/nuマウス(日本チャールス・リバー)の腹側部皮下に、3×106個移植した。移植7日後から、0.5 %メチルセルロース溶液(和光純薬)に懸濁した被験物質(200 mg/kg、1日2回、経口投与)、生理食塩水(大塚製薬)に懸濁したドセタキセル(5 mg/kg、1週間1回を3回、尾静脈投与)、或いは実施例4で得られた化合物(150 mg/kg、1日2回、経口投与)とドセタキセル(5 mg/kg、1週間1回を3回、尾静脈投与)の併用を、15日間投与した。ビークル投与群には、0.5 %メチルセルロース溶液を、1日2回、15日間、投与した。各群の匹数は5~8匹とした。投与期間の15日間は、週1~3回、すべての個体の腫瘍径および体重を測定した。また、薬剤投与を伴わない投与終了後の35日間、ドセタキセル投与群と被験物質とドセタキセルの併用群の腫瘍径、及び体重を測定した。
腫瘍体積は、以下の式から算出した。
腫瘍体積(mm3)=長径(mm)×短径(mm)×短径(mm)×1/2
Test Example 7: Tumor growth suppression experiment in HCC1806 human breast cancer cell-bearing mice HCC1806 cells (ATCC) were suspended in HBSS, and the ventral part of 5-week-old female BALB / cAnNCrj-nu / nu mice (Charles River Japan) 3 × 10 6 cells were transplanted subcutaneously. 7 days after transplantation, test substance suspended in 0.5% methylcellulose solution (Wako Pure Chemical Industries) (200 mg / kg, twice a day, oral administration), docetaxel suspended in physiological saline (Otsuka Pharmaceutical) (5 mg / kg, once a week for 3 times, tail vein administration) or the compound obtained in Example 4 (150 mg / kg, twice a day, oral administration) and docetaxel (5 mg / kg, 1 time per week) 3 times, tail vein administration) was administered for 15 days. In the vehicle administration group, 0.5% methylcellulose solution was administered twice a day for 15 days. The number of animals in each group was 5-8. During the administration period of 15 days, the tumor diameter and body weight of all individuals were measured 1 to 3 times a week. In addition, the tumor diameter and body weight of the docetaxel administration group and the combination group of the test substance and docetaxel were measured for 35 days after the end of administration without drug administration.
Tumor volume was calculated from the following equation.
Tumor volume (mm 3 ) = major axis (mm) x minor axis (mm) x minor axis (mm) x 1/2
 実施例4で得られた化合物について、試験例7に示す試験を行った腫瘍径の測定結果を、図6に示す。薬物投与最終日にて、ドセタキセル投与群、及び被験物質とドセタキセルの併用投与群の腫瘍体積は、ビークル投与群のそれに対し、有意に抑制された(p<0.05,Dunnett's test)。さらに、薬物投与終了後、ドセタキセル投与群においては、腫瘍体積の増大が認められたが、被験物質とドセタキセルの併用投与群の腫瘍体積は抑制され続け、完全に消失した。さらに観察期間内において、腫瘍の再発を、全例において認めなかった。 FIG. 6 shows the measurement results of the tumor diameter of the compound obtained in Example 4 in which the test shown in Test Example 7 was performed. On the last day of drug administration, the tumor volume in the docetaxel administration group and the test substance and docetaxel combination administration group was significantly suppressed compared to that in the vehicle administration group (p <0.05, Dunnett's test). Furthermore, after the drug administration was completed, the tumor volume increased in the docetaxel administration group, but the tumor volume in the combination administration group of the test substance and docetaxel continued to be suppressed and disappeared completely. Furthermore, no tumor recurrence was observed in all cases within the observation period.
 実施例4で得られた化合物について、試験例7に示す試験を行った体重の測定結果を、図7に示す。ドセタキセル投与群の体重(19日と22日)は、ビークル投与群のそれに対し、有意に抑制された(p<0.05,Student's t-test)。一方、被験物質とドセタキセルの併用投与群の体重は、いずれの測定日においても、ビークル投与群のそれに対し、有意な変化は認められなかった。 FIG. 7 shows the measurement results of body weight of the compound obtained in Example 4 in which the test shown in Test Example 7 was performed. The body weight (19th and 22nd days) of the docetaxel administration group was significantly suppressed compared with that of the vehicle administration group (p <0.05, Student's < t-test). On the other hand, the weight of the test substance and docetaxel combined administration group was not significantly changed from that of the vehicle administration group on any measurement day.
試験例8:FaDuヒト咽頭癌細胞を用いた担癌マウスにおける腫瘍増殖抑制実験
 FaDu細胞(ATCC)をHBSSに懸濁し、5週齢の雌性BALB/cAnNCrj-nu/nuマウス(日本チャールス・リバー)の腹側部皮下に、1×106個移植した。移植7日後から、0.5 %メチルセルロース溶液(和光純薬)に懸濁した被験物質(50、100、或いは200 mg/kg、1日2回、経口投与)を、15日間投与した。ビークル投与群には、0.5 %メチルセルロース溶液を、1日2回、15日間、投与した。各群の匹数は10匹とした。投与期間の15日間は、週1~3回、すべての個体の腫瘍径および体重を測定した。
腫瘍体積は、以下の式から算出した。
腫瘍体積(mm3)=長径(mm)×短径(mm)×短径(mm)×1/2
図8および図9に、平均値と標準誤差を示した。
Test Example 8: Tumor growth inhibition experiment in tumor-bearing mice using FaDu human pharyngeal carcinoma cells FaDu cells (ATCC) were suspended in HBSS, and 5-week-old female BALB / cAnNCrj-nu / nu mice (Charles River Japan) 1 × 10 6 cells were transplanted subcutaneously in the ventral region of each other. From 7 days after transplantation, a test substance (50, 100, or 200 mg / kg, orally administered twice a day) suspended in a 0.5% methylcellulose solution (Wako Pure Chemical Industries) was administered for 15 days. In the vehicle administration group, 0.5% methylcellulose solution was administered twice a day for 15 days. The number of animals in each group was 10. During the administration period of 15 days, the tumor diameter and body weight of all individuals were measured 1 to 3 times a week.
Tumor volume was calculated from the following equation.
Tumor volume (mm 3 ) = major axis (mm) x minor axis (mm) x minor axis (mm) x 1/2
8 and 9 show average values and standard errors.
 実施例4で得られた化合物について、試験例8に示す試験を行った腫瘍径の測定結果を、図8に示す。測定最終日にて、被験物質投与群の腫瘍体積は、ビークル投与群のそれに対し、用量依存的に、有意に抑制された(p<0.025,Williams' test)。 FIG. 8 shows the measurement results of the tumor diameter of the compound obtained in Example 4 in which the test shown in Test Example 8 was performed. On the last day of measurement, the tumor volume of the test substance-administered group was significantly suppressed in a dose-dependent manner compared to that of the vehicle-administered group (p <0.025, Williams' test).
 実施例4で得られた化合物について、試験例8に示す試験を行った体重の測定結果を、図9に示す。測定最終日にて、被験物質投与群の体重は、ビークル投与群のそれに対し、用量依存的に、有意に増加した(p<0.025,Williams' test)。また、この体重増作用において、浮腫など異常な原因は観察されなかった。 FIG. 9 shows the measurement results of the body weight of the compound obtained in Example 4 in which the test shown in Test Example 8 was performed. On the last day of measurement, the body weight of the test substance-administered group significantly increased in a dose-dependent manner compared with that of the vehicle-administered group (p <0.025, Williams' test). In this weight gain action, no abnormal cause such as edema was observed.
試験例9:アデノシンA3アンタゴニスト活性試験
 アデノシンA3遺伝子、エクオリン、Gα16を発現するCHO細胞にセレンテラジンh(125 nM)をロードした。その後、被験物質を添加し、その2分後にリガンド(アデノシン1μM)を添加してエクオリンの発光をFDSS 7000(浜松ホトニクス)で測定し、以下の式より阻害率を計算した。
  阻害率=(被験物質無添加時の発光強度―被験物質添加時の発光強度)
                   /被験物質無添加の発光強度*100
 結果により、本発明化合物が1μMでアデノシンA3アンタゴニスト活性を有さないことを確認した。
Test Example 9: Adenosine A3 antagonist activity test Coelenterazine h (125 nM) was loaded into CHO cells expressing adenosine A3 gene, aequorin and Gα16. Thereafter, a test substance was added, and 2 minutes later, a ligand (adenosine 1 μM) was added, aequorin luminescence was measured with FDSS 7000 (Hamamatsu Photonics), and the inhibition rate was calculated from the following formula.
Inhibition rate = (luminescence intensity when no test substance is added-luminescence intensity when test substance is added)
/ Luminescence intensity with no test substance added * 100
From the results, it was confirmed that the compound of the present invention had no adenosine A3 antagonist activity at 1 μM.
 実施例4で得られた化合物について、試験例9に示す試験を行った。試験結果について下記表に示す。
Figure JPOXMLDOC01-appb-T000090
For the compound obtained in Example 4, the test shown in Test Example 9 was conducted. The test results are shown in the following table.
Figure JPOXMLDOC01-appb-T000090
試験例10:溶解度測定
 96 wellラック上のUtubeに被験物質(10 mM DMSO溶液)を15μLずつ分注後、遠心エバポレーターにセットし、蒸発乾固を行った。DMSO 3μLを添加して再溶解させたのち、pH 7.4及び1.2の緩衝液を各300μL添加、25℃で90分間110 rpmで振盪の後、16~20時間静置した。2000 G、15分間の遠心で不溶物を分離し、上清100μLを96 wellプレートに採集した。別途96wellプレートに被験物質(10 mM DMSO溶液)を2μL分注し、50 %アセトニトリル198μLで希釈し、100μM標準液を調製した。さらに50%アセトニトリルで100μM標準液を10倍希釈し10μM標準液とした。上記溶解度測定サンプル及び標準溶液2種を液体クロマトグラフィーで分析し、標準溶液との面積比より溶解度を算出した。
 実施例で得られた化合物について、試験例10に示す試験を行った。試験結果について下記表に示す。
Figure JPOXMLDOC01-appb-T000091
Test Example 10: Solubility Measurement A test substance (10 mM DMSO solution) was dispensed by 15 μL each into a Utube on a 96-well rack, set in a centrifugal evaporator, and evaporated to dryness. After 3 μL of DMSO was added and redissolved, 300 μL each of pH 7.4 and 1.2 buffers were added, and the mixture was shaken at 110 rpm for 90 minutes at 25 ° C. and then allowed to stand for 16 to 20 hours. Insoluble matters were separated by centrifugation at 2000 G for 15 minutes, and 100 μL of the supernatant was collected in a 96-well plate. Separately, 2 μL of a test substance (10 mM DMSO solution) was dispensed into a 96-well plate and diluted with 198 μL of 50% acetonitrile to prepare a 100 μM standard solution. Further, a 100 μM standard solution was diluted 10-fold with 50% acetonitrile to obtain a 10 μM standard solution. The solubility measurement sample and two standard solutions were analyzed by liquid chromatography, and the solubility was calculated from the area ratio with the standard solution.
Tests shown in Test Example 10 were performed on the compounds obtained in the examples. The test results are shown in the following table.
Figure JPOXMLDOC01-appb-T000091
試験例11:バルク増殖抑制試験
 HCC1806細胞をATCCより入手した。HCC1806細胞は、10 %ウシ胎児血清、1 %ペニシリン/ストレプトマイシン含RPMI1640培地にて37℃、5 % CO2存在下にて培養した。細胞を、96ウエルプレートに1,000~3,000細胞/ウエルで細胞を播種し、DMSO濃度が0.1%となるように被験物質を添加し、5日間培養した。その後、プレストブルー(ライフテクノロジーズ)を用いて生細胞数を計測し、各被験物質の50 %細胞増殖を抑制する濃度(IC50; μM)を算出した。
Test Example 11: Bulk growth inhibition test HCC1806 cells were obtained from ATCC. HCC1806 cells were cultured in RPMI 1640 medium containing 10% fetal bovine serum and 1% penicillin / streptomycin in the presence of 37% at 5% CO 2 . Cells were seeded in a 96-well plate at 1,000 to 3,000 cells / well, a test substance was added so that the DMSO concentration was 0.1%, and the cells were cultured for 5 days. Thereafter, the number of viable cells was counted using Presto Blue (Life Technologies), and the concentration (IC 50 ; μM) at which 50% cell growth of each test substance was suppressed was calculated.
試験例12:癌細胞スフェア形成阻害実験
 HCC1806細胞をATCCより入手した。細胞をB27 Supplement(GIBCO)、20 ng/mL上皮細胞成長因子(EGF)(peprotech)、20 ng/mL塩基性線維芽細胞増殖因子(bFGF)(peprotech)、5μg/mLインスリン(GIBCO)及び1%メチルセルロース(ナカライテスク)を含むDMEM/F12培地にて1,000~3,000細胞/ウエルで細胞を播種し、Corning 96ウェルクリアラウンドボトムウルトラロウアタッチメントマイクロプレート(Corning,#7007)に播種した。DMSO濃度が0.1 %となるように被験物質を添加し、10日間培養した。その後、スフェア数を計測し、各被験物質の50 %細胞増殖を抑制する濃度(IC50:μM)を算出した。
Test Example 12: Cancer cell sphere formation inhibition experiment HCC1806 cells were obtained from ATCC. Cells were B27 Supplement (GIBCO), 20 ng / mL epidermal growth factor (EGF) (peprotech), 20 ng / mL basic fibroblast growth factor (bFGF) (peprotech), 5 μg / mL insulin (GIBCO) and 1 Cells were seeded at 1,000-3,000 cells / well in DMEM / F12 medium containing% methylcellulose (Nacalai Tesque) and seeded on Corning 96-well clear round bottom ultra-low attachment microplates (Corning, # 7007). A test substance was added so that the DMSO concentration was 0.1%, and the cells were cultured for 10 days. Thereafter, the number of spheres was counted, and the concentration (IC 50 : μM) at which 50% cell growth of each test substance was suppressed was calculated.
 実施例で得られた化合物について、試験例11と試験例12に示す試験を行った。試験結果について下表に示す。
Figure JPOXMLDOC01-appb-T000092
The compounds shown in Examples were subjected to the tests shown in Test Example 11 and Test Example 12. The test results are shown in the table below.
Figure JPOXMLDOC01-appb-T000092
試験例13:被験物質によるNanog発現抑制実験
 HCC1806細胞をATCCより入手した。細胞を、10%ウシ胎児血清、1%ペニシリン/ストレプトマイシンを含むRPMI培地にて、37℃、5% CO2存在下で継代培養した。被験物質添加前日に、細胞を6ウエルプレートに2×105細胞を播種して一晩培養した。被験物質を添加し、24時間培養した後、上清を除去した。氷冷PBSで洗浄し、Proteinase/Phosphatase inhibitor cocktail(Cell signaling technology,#5872S)を含む、氷冷RIPA buffer(Cell signaling technology, #9806S)を添加し、セルスクレーパーを用いて細胞を回収した。超音波処置後、10分間氷上で静置した。遠心(15,000 rpm、10分間)を行った後、上清を回収し、タンパク質濃度を定量した。RIPA buffer、及びSDS sample buffer(ナカライテスク、#09499-14)により一定濃度に調整した細胞タンパク質を含む溶液を、100℃、5分間、加熱した後に、電気泳動によりタンパク質を分離した。タンパク質は、PVDF膜に転写した後、5 % Skim milk/TBST溶液を用いて抗Nanog抗体(Cell signaling technology,#4903S)、或いは抗β-actin抗体(Cell signaling technology,#5125S)を添加し、4℃で一晩インキュベートした。抗体がハイブリダイズされたPVDF膜を、TBST溶液で洗浄した後、2次抗体(HRP標識抗ウサギIgG)を添加し、室温にて1時間インキュベートした。TBST溶液で洗浄した後、発色試薬(ECL Prime western blotting detection reagent,GE healthcare,#RPN2232)を添加し、検出器にて発光を検出した。
Test Example 13 Nanog Expression Inhibition Experiment with Test Substance HCC1806 cells were obtained from ATCC. The cells were subcultured in RPMI medium containing 10% fetal bovine serum and 1% penicillin / streptomycin in the presence of 5% CO 2 at 37 ° C. On the day before the addition of the test substance, the cells were seeded in 2 × 10 5 cells in a 6-well plate and cultured overnight. After adding a test substance and culturing for 24 hours, the supernatant was removed. The cells were washed with ice-cold PBS, ice-cold RIPA buffer (Cell signaling technology, # 9806S) containing Proteinase / Phosphatase inhibitor cocktail (Cell signaling technology, # 5872S) was added, and the cells were collected using a cell scraper. After sonication, it was left on ice for 10 minutes. After centrifugation (15,000 rpm, 10 minutes), the supernatant was recovered and the protein concentration was quantified. A solution containing cell proteins adjusted to a constant concentration with RIPA buffer and SDS sample buffer (Nacalai Tesque, # 09499-14) was heated at 100 ° C. for 5 minutes, and then the proteins were separated by electrophoresis. After the protein is transferred to the PVDF membrane, an anti-Nanog antibody (Cell signaling technology, # 4903S) or an anti-β-actin antibody (Cell signaling technology, # 5125S) is added using a 5% skim milk / TBST solution. Incubate overnight at 4 ° C. After washing the antibody-hybridized PVDF membrane with a TBST solution, a secondary antibody (HRP-labeled anti-rabbit IgG) was added and incubated at room temperature for 1 hour. After washing with TBST solution, a coloring reagent (ECL Prime western blotting detection reagent, GE healthcare, # RPN2232) was added, and luminescence was detected with a detector.
 実施例4で得られた化合物を被験物質として用い、対照化合物としてPalbociclib(選択的CDK4/6阻害薬;Selleck Chemicalsより購入)、Docetaxel(Sanofi Aventisより購入)について、試験例13に示す試験を行った。また、実施例72で得られた化合物を被験物質として同様の試験を行った。結果を図10(実施例4の化合物)と図11(実施例72の化合物)に示す。実施例4、実施例72での化合物は、濃度依存的にNanogの発現レベルを低下させた。一方、Palbociclib、及びDocetaxelは、Nanogの発現レベルに影響を与えなかった。尚、いずれの被験物質もβ-actinの発現レベルへ影響を与えなかった。
 同様にして、他の実施例化合物についてもNanog発現抑制効果を確認することができる。また、同様にして、本発明化合物が他のステムネス遺伝子(Sox2、β-catenin、Oct4など)の機能に及ぼす効果を確認することができる。実施例4の化合物は、Sox2およびβ-cateninに対しても発現抑制効果を示した。
Using the compound obtained in Example 4 as a test substance, Palbociclib (selective CDK4 / 6 inhibitor; purchased from Selleck Chemicals) and Docetaxel (purchased from Sanofi Aventis) as control compounds were subjected to the test shown in Test Example 13. It was. A similar test was conducted using the compound obtained in Example 72 as a test substance. The results are shown in FIG. 10 (the compound of Example 4) and FIG. 11 (the compound of Example 72). The compounds in Example 4 and Example 72 decreased the expression level of Nanog in a concentration-dependent manner. On the other hand, Palbociclib and Docetaxel did not affect the expression level of Nanog. None of the test substances affected the expression level of β-actin.
Similarly, the Nanog expression inhibitory effect can be confirmed also about other Example compounds. Similarly, the effect of the compound of the present invention on the functions of other stemness genes (Sox2, β-catenin, Oct4, etc.) can be confirmed. The compound of Example 4 also showed an expression inhibitory effect on Sox2 and β-catenin.
 本発明化合物は、マルチキナーゼ阻害薬として、細胞増殖により影響される可能性のある疾患、例えば、癌などの疾患の予防および/または治療に有用である。 The compound of the present invention is useful as a multikinase inhibitor for the prevention and / or treatment of diseases that may be affected by cell proliferation, for example, diseases such as cancer.

Claims (34)

  1.  式(1):
    Figure JPOXMLDOC01-appb-C000001
    [式(1)中、
     Xは、窒素原子またはCRを示し;
     Yは、水素原子、C1-6アルキル(該基は、ハロゲン原子、ヒドロキシ、C1-6アルコキシおよびC3-10シクロアルキルから選択される同一または異なる1~3個の基で置換されていてもよい)、C3-10シクロアルキル(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)または3員~8員の飽和複素環(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)を示し;
     R1a、R1b、R1c、R1dおよびR1eは、同一または異なって、水素原子、ハロゲン原子、ヒドロキシ、シアノ、ニトロ、C1-6アルキル(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)、C1-6アルコキシ(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)、アミノ、C1-6アルキルアミノ(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)、C2-12ジアルキルアミノ(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~6個の基で置換されていてもよい)、3~6員の環状アミノ(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)またはC1-6アルキルカルボニル(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)を示し;
     Rは、水素原子、シアノ、ヒドロキシ、C1-6アルキル(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)またはC3-10シクロアルキル(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)を示し;
     R、RおよびRは、同一または異なって、水素原子、ハロゲン原子、ヒドロキシ、シアノ、ニトロ、C1-6アルキル(該基は、ハロゲン原子、ヒドロキシ、C1-6アルコキシおよびC3-10シクロアルキルから選択される同一または異なる1~3個の基で置換されていてもよい)、C1-6アルコキシ(該基は、同一または異なる1~3個のハロゲン原子で置換されていてもよい)、アミノ、C1-6アルキルアミノ(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)、C2-12ジアルキルアミノ(該基は、ハロゲン原子、ヒドロキシおよびC1-6アルコキシから選択される同一または異なる1~6個の基で置換されていてもよい)、3~6員の環状アミノ(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)、C1-6アルキルカルボニル(該基は、同一または異なる1~3個のハロゲン原子で置換されていてもよい)、C3-10シクロアルキル(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)、3員~8員の飽和複素環(該基は、ハロゲン原子、ヒドロキシ、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)、C6-10アリール(該基は、ハロゲン原子、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)または5員~10員の単環式もしくは多環式のヘテロアリール(該基は、ハロゲン原子、C1-6アルキルおよびC1-6アルコキシから選択される同一または異なる1~4個の基で置換されていてもよい)を示し;
     Arは、C6-10の単環式もしくは多環式のアリールまたは5員~10員の単環式もしくは多環式のヘテロアリールを示す]
    で表される化合物またはその薬理学上許容される塩。
    Formula (1):
    Figure JPOXMLDOC01-appb-C000001
    [In Formula (1),
    X represents a nitrogen atom or CR 5 ;
    Y is a hydrogen atom, C 1-6 alkyl (the group is substituted with the same or different 1 to 3 groups selected from a halogen atom, hydroxy, C 1-6 alkoxy and C 3-10 cycloalkyl) C 3-10 cycloalkyl (which is substituted with 1 to 4 groups identical or different selected from a halogen atom, hydroxy, C 1-6 alkyl and C 1-6 alkoxy). Or a 3- to 8-membered saturated heterocyclic ring (the group is substituted with the same or different 1 to 4 groups selected from a halogen atom, hydroxy, C 1-6 alkyl and C 1-6 alkoxy) May be)
    R 1a , R 1b , R 1c , R 1d and R 1e are the same or different and each represents a hydrogen atom, a halogen atom, hydroxy, cyano, nitro, C 1-6 alkyl (the group is a halogen atom, hydroxy and C 1 Optionally substituted with 1 to 3 identical or different groups selected from -6 alkoxy), C 1-6 alkoxy (the same selected from halogen atom, hydroxy and C 1-6 alkoxy) Or optionally substituted with 1 to 3 different groups), amino, C 1-6 alkylamino (the group is the same or different 1 to 3 selected from halogen atom, hydroxy and C 1-6 alkoxy) ), C 2-12 dialkylamino (which is selected from halogen atoms, hydroxy and C 1-6 alkoxy) 3 to 6-membered cyclic amino (which is selected from a halogen atom, hydroxy, C 1-6 alkyl and C 1-6 alkoxy), which may be substituted with the same or different 1-6 groups. Or a C 1-6 alkylcarbonyl (the group may be the same or different 1-3 selected from halogen atom, hydroxy and C 1-6 alkoxy). Which may be substituted with
    R 4 represents a hydrogen atom, cyano, hydroxy, C 1-6 alkyl (the group may be substituted with the same or different 1 to 3 groups selected from a halogen atom, hydroxy and C 1-6 alkoxy) Or a C 3-10 cycloalkyl, which group may be substituted with 1 to 4 groups identical or different selected from a halogen atom, hydroxy and C 1-6 alkoxy;
    R 2 , R 3 and R 5 are the same or different and each represents a hydrogen atom, a halogen atom, hydroxy, cyano, nitro, C 1-6 alkyl (the group is a halogen atom, hydroxy, C 1-6 alkoxy and C 3 -10 may be substituted with the same or different 1 to 3 groups selected from cycloalkyl), C 1-6 alkoxy (the group is substituted with 1 to 3 halogen atoms which are the same or different May be substituted with the same or different 1 to 3 groups selected from a halogen atom, hydroxy and C 1-6 alkoxy), amino, C 1-6 alkylamino, C 2-12 dialkylamino (in which the halogen atom, optionally substituted by the same or different 1-6 groups selected from hydroxy and C 1-6 alkoxy ), 3 cyclic amino (base to 6-membered, halogen atom, hydroxy, optionally substituted by the same or different 1 to 4 groups selected from C 1-6 alkyl and C 1-6 alkoxy ), C 1-6 alkylcarbonyl (the group may be substituted with the same or different 1 to 3 halogen atoms), C 3-10 cycloalkyl (the group is a halogen atom, hydroxy, C 1 3 to 8 membered saturated heterocyclic ring (which may be substituted with the same or different 1 to 4 groups selected from -6 alkyl and C 1-6 alkoxy), which is a halogen atom, hydroxy, Optionally substituted with 1 to 4 identical or different groups selected from C 1-6 alkyl and C 1-6 alkoxy), C 6-10 aryl (the group is a halogen atom, C 1-6 Archi And the same or different one to four monocyclic or heteroaryl (substrate polycyclic which may also be) or a 5- to 10-membered optionally substituted with a group selected from C 1-6 alkoxy, halogen And optionally substituted with 1-4 identical or different groups selected from atoms, C 1-6 alkyl and C 1-6 alkoxy;
    Ar represents C 6-10 monocyclic or polycyclic aryl or 5- to 10-membered monocyclic or polycyclic heteroaryl]
    Or a pharmacologically acceptable salt thereof.
  2.  Arが、フェニルまたはピリジルである請求項1に記載の化合物またはその薬理学上許容される塩。 Ar is phenyl or pyridyl, The compound or its pharmacologically acceptable salt of Claim 1 characterized by the above-mentioned.
  3.  Arが、フェニルである請求項1または請求項2に記載の化合物またはその薬理学上許容される塩。 Ar is phenyl, The compound of Claim 1 or Claim 2, or its pharmacologically acceptable salt.
  4.  Xが、CRであり;
     Rが、水素原子、ハロゲン原子またはC1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)である請求項1~3いずれか一項に記載の化合物またはその薬理学上許容される塩。
    X is CR 5 ;
    The compound according to any one of claims 1 to 3, wherein R 5 is a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). Or a pharmacologically acceptable salt thereof.
  5.  Xが、窒素原子である請求項1~3いずれか一項に記載の化合物またはその薬理学上許容される塩。 The compound according to any one of claims 1 to 3, or a pharmacologically acceptable salt thereof, wherein X is a nitrogen atom.
  6.  Yが、水素原子またはC1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)である請求項1~5いずれか一項に記載の化合物またはその薬理学上許容される塩。 The compound or pharmacology thereof according to any one of claims 1 to 5, wherein Y is a hydrogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). Top acceptable salt.
  7.  Rが、水素原子、C1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)またはC3-10シクロアルキル(該基は、1~4個のフッ素原子で置換されていてもよい)である請求項1~6のいずれか一項に記載の化合物またはその薬理学上許容される塩。 R 4 is a hydrogen atom, C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or C 3-10 cycloalkyl (the group is substituted with 1 to 4 fluorine atoms). The compound or a pharmacologically acceptable salt thereof according to any one of claims 1 to 6, which may be substituted with an atom.
  8.  Rが、水素原子、C1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)またはC3-4シクロアルキル(該基は、1~4個のフッ素原子で置換されていてもよい)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薬理学上許容される塩。 R 4 is a hydrogen atom, C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or C 3-4 cycloalkyl (the group is substituted with 1 to 4 fluorine atoms). The compound or a pharmacologically acceptable salt thereof according to any one of claims 1 to 7, which may be substituted with an atom.
  9.  RおよびRが、同一または異なって、水素原子、ハロゲン原子またはC1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)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薬理学上許容される塩。 R 2 and R 3 are the same or different and each is a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms). Or a pharmacologically acceptable salt thereof.
  10.  R1a、R1b、R1c、R1dおよびR1eが、同一または異なって、水素原子、ハロゲン原子、C1-6アルキル(該基は、フッ素原子およびC1-6アルコキシから選択される同一または異なる1~3個の基で置換されていてもよい)またはC1-6アルコキシ(該基は、1~3個のフッ素原子で置換されていてもよい)である請求項1~9のいずれか一項に記載の化合物またはその薬理学上許容される塩。 R 1a , R 1b , R 1c , R 1d and R 1e are the same or different and are each a hydrogen atom, a halogen atom or C 1-6 alkyl (the same group selected from a fluorine atom and C 1-6 alkoxy) Or a C 1-6 alkoxy (which may be substituted with 1 to 3 fluorine atoms) or a C 1-6 alkoxy (which may be substituted with 1 to 3 different groups). The compound according to any one of the above or a pharmacologically acceptable salt thereof.
  11.  R1a、R1b、R1c、R1dおよびR1eが、同一または異なって、水素原子、ハロゲン原子またはC1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)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薬理学上許容される塩。 R 1a , R 1b , R 1c , R 1d and R 1e are the same or different and each represents a hydrogen atom, a halogen atom or C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) The compound according to any one of claims 1 to 10 or a pharmacologically acceptable salt thereof.
  12.  Rが、C1-6アルキル(該基は、1~3個のフッ素原子で置換されていてもよい)またはシクロプロピルである請求項1~11のいずれか一項に記載の化合物またはその薬理学上許容される塩。 The compound according to any one of claims 1 to 11, wherein R 4 is C 1-6 alkyl (the group may be substituted with 1 to 3 fluorine atoms) or cyclopropyl. Pharmacologically acceptable salt.
  13.  以下の化合物群:
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
    (4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
    (4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,4-トリフルオロフェニル)メタノール、
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,4-トリフルオロフェニル)メタノール、
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール、
    (4-(((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール、
    (3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (3,4-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール、
    (4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール、
    (2,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (2,4-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,6-トリフルオロフェニル)メタノール、
    (4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,6-トリフルオロフェニル)メタノール、
    (2,3-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (2,3-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(ペンタフルオロフェニル)メタノール、
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(ペンタフルオロフェニル)メタノール、
    (3,4-ジフルオロフェニル)(5-フルオロ-4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (3,4-ジフルオロフェニル)(5-フルオロ-4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (5-フルオロ-4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
    (5-フルオロー4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
    (5-フルオロー4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール、
    (5-フルオロ-4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール、
    (5-クロロ-4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
    (5-クロロ-4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
    1-(3,4-ジフルオロフェニル)-1-(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)エタン-1-オール、
    1-(3,4-ジフルオロフェニル)-1-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)エタン-1-オール
    (4-((3-エチル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール、
    (4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール、
    (4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
    1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)-1-(2,4,5-トリフルオロフェニル)エタン-1-オール、
    (4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(4-フルオロ-3-メチルフェニル)メタノール、
    (4-クロロ-3-フルオロフェニル)(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)メタノール、
    1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)-1-(4-フルオロ-3-メチルフェニル)エタン-1-オール、および
    1-(4-クロロ-3-フルオロフェニル)-1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)エタン-1-オール
    から選択される化合物である請求項1に記載の化合物またはその薬理学上許容される塩。
    The following compound groups:
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
    (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
    (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,4-trifluorophenyl) methanol,
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,4-trifluorophenyl) methanol,
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol,
    (4-(((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol,
    (3,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (3,4-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol,
    (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol,
    (2,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (2,4-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,6-trifluorophenyl) methanol,
    (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,6-trifluorophenyl) methanol,
    (2,3-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (2,3-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (pentafluorophenyl) methanol,
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (pentafluorophenyl) methanol,
    (3,4-difluorophenyl) (5-fluoro-4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (3,4-difluorophenyl) (5-fluoro-4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (5-fluoro-4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
    (5-Fluoro-4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
    (5-Fluoro-4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol,
    (5-Fluoro-4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol,
    (5-chloro-4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
    (5-chloro-4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
    1- (3,4-difluorophenyl) -1- (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) ethane-1-ol,
    1- (3,4-Difluorophenyl) -1- (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) ethane-1-ol (4- ((3-Ethyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol,
    (4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol,
    (4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
    1- (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) -1- (2,4,5-trifluorophenyl) ethane-1-ol,
    (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (4-fluoro-3-methylphenyl) methanol,
    (4-chloro-3-fluorophenyl) (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) methanol,
    1- (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) -1- (4-fluoro-3-methylphenyl) ethan-1-ol, and 1- A compound selected from (4-chloro-3-fluorophenyl) -1- (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) ethane-1-ol The compound according to claim 1 or a pharmacologically acceptable salt thereof.
  14.  以下の化合物群:
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
    (4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
    (4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,4-トリフルオロフェニル)メタノール、
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,4-トリフルオロフェニル)メタノール、
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール、
    (4-(((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール、
    (3,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (3,4-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール、
    (4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,3,5-トリフルオロフェニル)メタノール、
    (2,4-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (2,4-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,6-トリフルオロフェニル)メタノール、
    (4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)(2,4,6-トリフルオロフェニル)メタノール、
    (2,3-ジフルオロフェニル)(4-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    (2,3-ジフルオロフェニル)(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)メタノール、
    1-(3,4-ジフルオロフェニル)-1-(4-メチル-6-((5-メチル-1H-ピラゾール-3-イル)アミノ)ピリミジン-2-イル)エタン-1-オール、
    (4-((3-エチル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール、
    (4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(3,4,5-トリフルオロフェニル)メタノール、
    (4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(2,4,5-トリフルオロフェニル)メタノール、
    1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)-1-(2,4,5-トリフルオロフェニル)エタン-1-オール、
    (4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)(4-フルオロ-3-メチルフェニル)メタノール、
    (4-クロロ-3-フルオロフェニル)(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)メタノール、
    1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)-1-(4-フルオロ-3-メチルフェニル)エタン-1-オールおよび
    1-(4-クロロ-3-フルオロフェニル)-1-(4-((3-シクロプロピル-1H-ピラゾール-5-イル)アミノ)ピリミジン-2-イル)エタン-1-オール
    から選択される化合物である請求項1に記載の化合物またはその薬理学上許容される塩。
    The following compound groups:
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
    (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
    (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,4-trifluorophenyl) methanol,
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,4-trifluorophenyl) methanol,
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol,
    (4-(((5-Methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol,
    (3,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (3,4-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol,
    (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,3,5-trifluorophenyl) methanol,
    (2,4-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (2,4-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,6-trifluorophenyl) methanol,
    (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) (2,4,6-trifluorophenyl) methanol,
    (2,3-difluorophenyl) (4-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    (2,3-difluorophenyl) (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) methanol,
    1- (3,4-difluorophenyl) -1- (4-methyl-6-((5-methyl-1H-pyrazol-3-yl) amino) pyrimidin-2-yl) ethane-1-ol,
    (4-((3-Ethyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol,
    (4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (3,4,5-trifluorophenyl) methanol,
    (4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (2,4,5-trifluorophenyl) methanol,
    1- (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) -1- (2,4,5-trifluorophenyl) ethane-1-ol,
    (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) (4-fluoro-3-methylphenyl) methanol,
    (4-chloro-3-fluorophenyl) (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) methanol,
    1- (4-((3-Cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) -1- (4-fluoro-3-methylphenyl) ethan-1-ol and 1- ( 4-chloro-3-fluorophenyl) -1- (4-((3-cyclopropyl-1H-pyrazol-5-yl) amino) pyrimidin-2-yl) ethan-1-ol The compound according to claim 1 or a pharmacologically acceptable salt thereof.
  15.  請求項1~14のいずれか一項に記載の化合物またはその薬理学上許容される塩を含有する医薬組成物。 A pharmaceutical composition comprising the compound according to any one of claims 1 to 14 or a pharmacologically acceptable salt thereof.
  16.  請求項1~14のいずれか一項に記載の化合物またはその薬理学上許容される塩を有効成分として含有するキナーゼ阻害剤。 A kinase inhibitor comprising the compound according to any one of claims 1 to 14 or a pharmacologically acceptable salt thereof as an active ingredient.
  17.  請求項1~14のいずれか一項に記載の化合物またはその薬理学上許容される塩を有効成分として含有する抗癌剤。 An anticancer agent comprising the compound according to any one of claims 1 to 14 or a pharmacologically acceptable salt thereof as an active ingredient.
  18.  癌が、乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、前立腺癌、脳腫瘍、子宮内膜癌、膵臓癌、胃癌、骨肉腫、骨髄腫、または血液癌である請求項17に記載の抗癌剤。 The cancer is breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor, endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or blood cancer 18. The anticancer agent according to 17.
  19.  治療が必要な患者に、治療上の有効量の請求項1~14のいずれか一項に記載の化合物またはその薬理学上許容される塩を投与することを含む、癌を治療および/または予防するための方法。 Treatment and / or prevention of cancer comprising administering to a patient in need of treatment a therapeutically effective amount of a compound according to any one of claims 1-14 or a pharmacologically acceptable salt thereof. How to do.
  20.  癌が、乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、前立腺癌、脳腫瘍、子宮内膜癌、膵臓癌、胃癌、骨肉腫、骨髄腫、または血液癌である請求項19に記載の治療および/または予防するための方法。 The cancer is breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor, endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or blood cancer 20. The method for treatment and / or prevention according to 19.
  21.  癌の治療剤および/または予防剤を製造するための、請求項1~14のいずれか一項に記載の化合物またはその薬理学上許容される塩の使用。 Use of the compound according to any one of claims 1 to 14 or a pharmacologically acceptable salt thereof for producing a therapeutic and / or preventive agent for cancer.
  22.  癌が、乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、前立腺癌、脳腫瘍、子宮内膜癌、膵臓癌、胃癌、骨肉腫、骨髄腫、または血液癌である請求項21に記載の使用。 The cancer is breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor, endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or blood cancer 21. Use according to 21.
  23.  癌の治療および/または予防に使用するための請求項1~14のいずれか一項に記載の化合物またはその薬理学上許容される塩。 The compound or a pharmacologically acceptable salt thereof according to any one of claims 1 to 14, for use in the treatment and / or prevention of cancer.
  24.  癌が、乳癌、卵巣癌、頭頸部癌、肺癌、大腸癌、皮膚癌、肝癌、前立腺癌、脳腫瘍、子宮内膜癌、膵臓癌、胃癌、骨肉腫、骨髄腫、または血液癌である、請求項23に記載の化合物またはその薬理学上許容される塩。 The cancer is breast cancer, ovarian cancer, head and neck cancer, lung cancer, colon cancer, skin cancer, liver cancer, prostate cancer, brain tumor, endometrial cancer, pancreatic cancer, gastric cancer, osteosarcoma, myeloma, or hematological cancer Item 24 or a pharmacologically acceptable salt thereof.
  25.  ステムネス遺伝子の発現抑制作用を有する、請求項17または請求項18に記載の抗癌剤。 The anticancer agent according to claim 17 or 18, which has an action of suppressing expression of a stemness gene.
  26.  ステムネス遺伝子が、Nanog、Sox2、β-cateninおよびOct4からなる群から選ばれるいずれかである、請求項25に記載の抗癌剤。 26. The anticancer agent according to claim 25, wherein the stemness gene is any one selected from the group consisting of Nanog, Sox2, β-catenin and Oct4.
  27.  ステムネス遺伝子がNanogである、請求項25に記載の抗癌剤。 The anticancer agent according to claim 25, wherein the stemness gene is Nanog.
  28.  請求項1~14のいずれか一項に記載の化合物またはその薬理学上許容される塩を有効成分として含有する、ステムネス遺伝子の発現抑制剤。 A stemness gene expression inhibitor comprising the compound according to any one of claims 1 to 14 or a pharmacologically acceptable salt thereof as an active ingredient.
  29.  3種またはそれ以上のキナーゼを阻害する化合物を含有する、抗癌剤。 An anticancer agent containing a compound that inhibits three or more kinases.
  30.  キナーゼがCDK2、CDK5、JAK2、AURKA、AURKBおよびCDK1からなる群から選ばれる、請求項29に記載の抗癌剤。 30. The anticancer agent according to claim 29, wherein the kinase is selected from the group consisting of CDK2, CDK5, JAK2, AURKA, AURKB, and CDK1.
  31.  CDK2、CDK5およびJAK2からなる群から選ばれる2またはそれ以上のキナーゼを阻害する化合物を含有する、抗癌剤。 An anticancer agent comprising a compound that inhibits two or more kinases selected from the group consisting of CDK2, CDK5 and JAK2.
  32.  3種またはそれ以上のキナーゼを阻害することを含む、癌の治療方法。 A method of treating cancer comprising inhibiting three or more kinases.
  33.  ステムネス遺伝子の発現を抑制することを含む、癌の治療方法。 A method for treating cancer comprising suppressing the expression of a stemness gene.
  34.  CDK5を阻害することを含む、ステムネス遺伝子の発現抑制方法。 A method for suppressing the expression of a stemness gene, comprising inhibiting CDK5.
PCT/JP2016/066617 2015-06-05 2016-06-03 Novel heteroarylamino-3-pyrazole derivatives and pharmacologically acceptable salts thereof WO2016195083A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2015-114860 2015-06-05
JP2015114860A JP2018135268A (en) 2015-06-05 2015-06-05 Novel heteroaryl amino-3-pyrazole derivative and pharmacologically acceptable salt thereof

Publications (1)

Publication Number Publication Date
WO2016195083A1 true WO2016195083A1 (en) 2016-12-08

Family

ID=57441283

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2016/066617 WO2016195083A1 (en) 2015-06-05 2016-06-03 Novel heteroarylamino-3-pyrazole derivatives and pharmacologically acceptable salts thereof

Country Status (3)

Country Link
JP (1) JP2018135268A (en)
TW (1) TW201704227A (en)
WO (1) WO2016195083A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111362923A (en) * 2020-03-25 2020-07-03 魏威 Method for preparing RET inhibitor pracetib, intermediate of pracetib and preparation method of pracetib
CN111440151A (en) * 2020-03-25 2020-07-24 魏威 Method for preparing antitumor drug prasutinib

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017156493A1 (en) 2016-03-11 2017-09-14 Denali Therapeutics Inc. Compounds, compositions, and methods
WO2017218843A1 (en) 2016-06-16 2017-12-21 Denali Therapeutics Inc. Pyrimidin-2-ylamino-1h-pyrazols as lrrk2 inhibitors for use in the treatment of neurodegenerative disorders
US20200157081A1 (en) * 2017-05-24 2020-05-21 Denali Therapeutics Inc. Compounds, compositions and methods

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2004518703A (en) * 2000-12-21 2004-06-24 バーテックス ファーマシューティカルズ インコーポレイテッド Pyrazole compounds useful as protein kinase inhibitors
JP2009539374A (en) * 2006-06-06 2009-11-19 ユニバーシティ・オブ・テネシー・リサーチ・ファウンデーション Composition enriched with neoplastic stem cells and method comprising the same
JP2011509994A (en) * 2008-01-18 2011-03-31 サーントゥル ナシオナル ドゥ ラ ルシェルシュ シャーンティフィク Tetrahydrocyclopenta [c] acridine derivatives as kinase inhibitors and their biological applications
WO2012030914A1 (en) * 2010-09-01 2012-03-08 Ambit Boisciences Corporation 4-azolylaminoquinazoline derivatives and methods of use thereof
WO2012030924A1 (en) * 2010-09-01 2012-03-08 Ambit Biosciences Corporation Azolopyridine and azolopyrimidine compounds and methods of use thereof
WO2012030944A2 (en) * 2010-09-01 2012-03-08 Ambit Biosciences Corporation Quinoline and isoquinoline compounds and methods of use thereof
JP2012519179A (en) * 2009-02-27 2012-08-23 アムビト ビオスシエンセス コルポラチオン JAK kinase-regulated quinazoline derivatives and methods of use thereof
JP2013538811A (en) * 2010-09-01 2013-10-17 アムビト ビオスシエンセス コルポラチオン Quinazoline compounds and methods of using the same
JP2014129341A (en) * 2012-11-27 2014-07-10 Kokuritsu Iyakuhin Shokuhin Eisei Kenkyusho Multi-kinase inhibitor, anticancer agent, anti-metastasis agent, drug resistance inhibitor, pain inhibitor, and antipruritic agent

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2004518703A (en) * 2000-12-21 2004-06-24 バーテックス ファーマシューティカルズ インコーポレイテッド Pyrazole compounds useful as protein kinase inhibitors
JP2009539374A (en) * 2006-06-06 2009-11-19 ユニバーシティ・オブ・テネシー・リサーチ・ファウンデーション Composition enriched with neoplastic stem cells and method comprising the same
JP2011509994A (en) * 2008-01-18 2011-03-31 サーントゥル ナシオナル ドゥ ラ ルシェルシュ シャーンティフィク Tetrahydrocyclopenta [c] acridine derivatives as kinase inhibitors and their biological applications
JP2012519179A (en) * 2009-02-27 2012-08-23 アムビト ビオスシエンセス コルポラチオン JAK kinase-regulated quinazoline derivatives and methods of use thereof
WO2012030914A1 (en) * 2010-09-01 2012-03-08 Ambit Boisciences Corporation 4-azolylaminoquinazoline derivatives and methods of use thereof
WO2012030924A1 (en) * 2010-09-01 2012-03-08 Ambit Biosciences Corporation Azolopyridine and azolopyrimidine compounds and methods of use thereof
WO2012030944A2 (en) * 2010-09-01 2012-03-08 Ambit Biosciences Corporation Quinoline and isoquinoline compounds and methods of use thereof
JP2013538811A (en) * 2010-09-01 2013-10-17 アムビト ビオスシエンセス コルポラチオン Quinazoline compounds and methods of using the same
JP2014129341A (en) * 2012-11-27 2014-07-10 Kokuritsu Iyakuhin Shokuhin Eisei Kenkyusho Multi-kinase inhibitor, anticancer agent, anti-metastasis agent, drug resistance inhibitor, pain inhibitor, and antipruritic agent

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BEBBINGTON, DAVID ET AL.: "The discovery of the potent aurora inhibitor MK -0457 (VX-680", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 19, no. 13, 2009, pages 3586 - 3592 *
ZHANG, BAIDONG ET AL.: "3D-QSAR and molecular docking studies on serivatives of MK -0457, GSK1070916 and SNS-314 as inhibitors against Aurora B kinase", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 11, no. 11, 2010, pages 4326 - 4347, XP055334396 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111362923A (en) * 2020-03-25 2020-07-03 魏威 Method for preparing RET inhibitor pracetib, intermediate of pracetib and preparation method of pracetib
CN111440151A (en) * 2020-03-25 2020-07-24 魏威 Method for preparing antitumor drug prasutinib

Also Published As

Publication number Publication date
TW201704227A (en) 2017-02-01
JP2018135268A (en) 2018-08-30

Similar Documents

Publication Publication Date Title
JP6921101B2 (en) Protein kinase inhibitor and its preparation method and pharmaceutical use
ES2702450T3 (en) Quinoxaline derivatives useful as modulators of the FGFR kinase
WO2016195083A1 (en) Novel heteroarylamino-3-pyrazole derivatives and pharmacologically acceptable salts thereof
KR101828187B1 (en) Novel fused pyrimidine compound or salt thereof
JP5719770B2 (en) Icotinib hydrochloride, compound, crystallographic form, concomitant drug and its use
CA3177261A1 (en) Benzothiazolyl biaryl compound, and preparation method and use
JP7008064B2 (en) Heterocyclic compound as an FGFR4 inhibitor
CN104910137B (en) CDK kinase inhibitor
KR20120081164A (en) Pi3k(delta) selective inhibitors
WO2017071516A1 (en) Kinase inhibitor, and preparing method and pharmaceutical use thereof
TW201018696A (en) Fused bicyclic and tricyclic pyrimidine compounds as tyrosine kinase inhibitors
CN112771049B (en) FGFR4 inhibitor and application thereof
WO2021043208A1 (en) 3, 5-disubstituted pyrazole compounds as kinase inhibitors and uses thereof
EP2139893A2 (en) 2,6,9-substituted purine derivatives having antiproliferative properties
CN112094269B (en) Saturated six-membered ring heterocyclic compound, preparation method and application
WO2019070167A1 (en) Epidermal growth factor receptor inhibitors
WO2023083201A1 (en) Aminopyrazole derivative, and preparation method therefor and use thereof
RU2669696C2 (en) Conformationally restricted pi3k and mtor inhibitors
CN108456214B (en) Quinazoline compound containing oxazole or imidazole structure and application thereof
CN111836819A (en) Arylamine-substituted pyrrolopyrimidine compound, and preparation method and application thereof
CN115433207A (en) Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof
JP2018090546A (en) Medicine composed of heteroaryl amino-3-pyrazole derivative and pharmacologically acceptable salt thereof
CN107721937B (en) Preparation method and application of compound containing pyrimidone skeleton
CN115703760A (en) 2,4-disubstituted pyrimidines cyclin dependent kinase inhibitor and preparation method and application thereof
CN116583287A (en) Pyridazinone heterocyclic compound, preparation method and application thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16803514

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16803514

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP