[go: up one dir, main page]

WO2024111626A1 - Novel thiazole derivative - Google Patents

Novel thiazole derivative Download PDF

Info

Publication number
WO2024111626A1
WO2024111626A1 PCT/JP2023/041996 JP2023041996W WO2024111626A1 WO 2024111626 A1 WO2024111626 A1 WO 2024111626A1 JP 2023041996 W JP2023041996 W JP 2023041996W WO 2024111626 A1 WO2024111626 A1 WO 2024111626A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
reaction
amino
pyrazol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/JP2023/041996
Other languages
French (fr)
Japanese (ja)
Inventor
匡明 澤
真以 新井
充治 花田
大本 弘志
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Carna Biosciences Inc
Original Assignee
Carna Biosciences Inc
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 Carna Biosciences Inc filed Critical Carna Biosciences Inc
Publication of WO2024111626A1 publication Critical patent/WO2024111626A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • the present invention relates to a medicine, in particular to a novel thiazole derivative or a pharma- ceutical acceptable salt thereof that has TGF ⁇ type I receptor (ALK5) inhibitory activity.
  • TGF- ⁇ Transforming growth factor ⁇
  • TGF- ⁇ a multifunctional cytokine
  • TGF- ⁇ 1, ⁇ 2, ⁇ 3 three isoforms in mammals and is involved in physiological functions such as cell proliferation, differentiation control, migration, and adhesion
  • TGF- ⁇ binds to a complex formed by a single-pass transmembrane type I receptor (ALK5) with a serine/threonine kinase domain and a type II receptor, and transmits a signal into the cell.
  • ALK5 is activated by the type II receptor upon binding of TGF- ⁇ , ALK5 phosphorylates the transcription factors Smad2 and Smad3 in the cell.
  • Non-Patent Documents 1 and 2 After forming a complex with Smad4, phosphorylated Smad2 and Smad3 migrate to the nucleus and induce the transcription of target genes directly or together with other transcription factors (Non-Patent Documents 1 and 2).
  • TGF- ⁇ signaling plays an important role in pathological conditions such as wound healing, inflammation/immunity, and cancer invasion/metastasis (Non-Patent Document 3).
  • EMT epithelial-mesenchymal transition
  • angiogenesis angiogenesis
  • TGF- ⁇ is involved in the proliferation of osteosarcoma
  • ALK5 inhibitors suppress the proliferation of osteosarcoma cells stimulated with TGF- ⁇
  • anti-TGF- ⁇ antibodies suppress lung metastasis of osteosarcoma
  • ALK5 inhibitors are useful for the treatment of solid cancers and metastasis in which TGF- ⁇ is involved.
  • TGF- ⁇ controls the differentiation induction of regulatory T cells (Treg), which are responsible for immune control, and it is thought that in the cancer microenvironment, Treg cells suppress antitumor immunity and promote tumor growth (Non-Patent Document 8). Therefore, suppressing TGF- ⁇ signaling is expected to have an antitumor effect by cancer immunotherapy and a combined effect with immune checkpoint inhibitors.
  • TGF- ⁇ is also involved in fibrosis, a pathological excess state of the normal tissue repair process, and it has been reported that abnormal excess signaling of TGF- ⁇ is the cause of fibrosis (Non-Patent Document 9). Therefore, a compound having an ALK5 inhibitory activity is useful for treating diseases in which TGF- ⁇ signaling is involved, such as cancer, and for expanding and enhancing the therapeutic effect in cancer immunotherapy by using it in combination with an immune checkpoint inhibitor, etc. It is also useful for treating and preventing fibrotic diseases, etc.
  • the objective of the present invention is to provide a pharmaceutical, in particular a novel thiazole derivative or a pharma- ceutical acceptable salt thereof that has TGF ⁇ type I receptor (ALK5) inhibitory activity.
  • the present invention can be achieved by the following thiazole derivatives or pharma- ceutically acceptable salts thereof: (1) The following formula (I): (wherein R 1 represents an optionally substituted alkyl group, an optionally substituted saturated heterocyclic group or an optionally substituted amino group; R 2 represents an optionally substituted aryl group, an optionally substituted heteroaryl group or an optionally substituted alkynyl group; and Z represents an optionally substituted alkyl group, an optionally substituted alkenyl group, an optionally substituted alkynyl group, an optionally substituted saturated heterocyclic group or an optionally substituted cycloalkyl group), or a pharmacy acceptable salt thereof.
  • R 1 represents an optionally substituted alkyl group, an optionally substituted saturated heterocyclic group or an optionally substituted amino group
  • R 2 represents an optionally substituted aryl group, an optionally substituted heteroaryl group or an optionally substituted alkynyl group
  • Z represents an optionally substituted alky
  • the present inventors conducted various studies to solve the above problems, and found that the novel thiazole derivative represented by the above formula (I) and its pharma- ceutical acceptable salt exhibit ALK5 inhibitory activity, and thus completed the present invention.
  • the compounds provided by the present invention are useful for treating diseases known to be associated with abnormal cell responses via the TGF ⁇ signal pathway, particularly cancer, and are also useful for preventing tumor metastasis and recurrence by targeting cancer stem cells. Furthermore, by using them in combination with immune checkpoint inhibitors, they are useful for expanding and enhancing the therapeutic effect in cancer immunotherapy. They are also useful for treating and preventing fibrotic diseases and the like. They are also useful as experimental and research reagents as ALK5 inhibitors and TGF ⁇ signal inhibitors.
  • the novel thiazole derivative of the present invention has the following formula (I):
  • R1 represents an alkyl group which may have a substituent, a saturated heterocyclic group which may have a substituent, or an amino group which may have a substituent
  • R2 represents an aryl group which may have a substituent, a heteroaryl group which may have a substituent, or an alkynyl group which may have a substituent
  • Z represents an alkyl group which may have a substituent, an alkenyl group which may have a substituent, an alkynyl group which may have a substituent, a saturated heterocyclic group which may have a substituent, or a cycloalkyl group which may have a substituent.
  • It is a compound represented by the formula:
  • halogen means a fluorine atom, a chlorine atom, a bromine atom, or an iodine atom.
  • C 1-6 alkyl means a linear or branched saturated hydrocarbon group having 1-6 carbon atoms
  • C 6 alkyl means a saturated hydrocarbon group having 6 carbon atoms. The same applies to other numbers.
  • C 1-3 alkyl specifically means a methyl group, an ethyl group, a propyl group, or an isopropyl group.
  • examples of “C 1-6 alkyl” include a butyl group, a 1-methylpropyl group, a 2-methylpropyl group, a tert-butyl group, a pentyl group, a 1,1-dimethylpropyl group, a 1,2-dimethylpropyl group, a 1-methylbutyl group, a 2-methylbutyl group, a 4-methylpentyl group, a 3-methylpentyl group, a 2-methylpentyl group, a 1-methylpentyl group, and an n-hexyl group.
  • C 3-8 cycloalkyl refers to a cyclic saturated hydrocarbon group having 3 to 8 carbon atoms.
  • Specific examples include a cyclopropyl group, a cyclobutyl group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, and a cyclooctyl group.
  • C 1-6 alkoxy means “C 1-6 alkyloxy”
  • the “C 1-6 alkyl” portion has the same meaning as the above-mentioned "C 1-6 alkyl”.
  • the substituents may be one or more of any type of substituent at any chemically possible position, and when there are two or more substituents, the respective substituents may be the same or different.
  • R1 R 1 represents an alkyl group which may have a substituent, a saturated heterocyclic group which may have a substituent, or an amino group which may have a substituent.
  • the alkyl group moiety of the alkyl group which may have a substituent may be any of C1-6 alkyl, and particularly preferably C1-3 alkyl.
  • a specific example of a preferred substituent in the C 1-6 alkyl or C 1-3 alkyl is a substituted amino group --NR 11 R 12.
  • R 11 and R 12 each independently represent , a 4- to 6-membered oxygen-containing saturated heterocycle, or a C 1-6 alkyl, or a 4- to 6-membered ring which, taken together with the nitrogen atom to which it is bonded, may further contain a nitrogen atom or an oxygen atom.
  • the oxygen-containing saturated heterocycle include an oxetanyl group, a tetrahydropyranyl group, and a tetrahydrofuryl group
  • examples of the nitrogen-containing saturated heterocycle include pyrrolidine, piperidine, piperazine, and morpholine. These may be substituted with halogen, a methyl group, a methoxy group, or the like.
  • a preferred embodiment of the alkyl group which may have a substituent is -CH 2 NR 11 R 12 in which the above-mentioned substituted amino group is substituted with a methyl group.
  • the saturated heterocyclic group portion of the optionally substituted saturated heterocyclic group is a 4- to 6-membered heterocyclic group containing 1 to 3 heteroatoms arbitrarily selected from a sulfur atom, a nitrogen atom, and an oxygen atom. and can be substituted at any position where chemical substitution is possible.
  • the substituent can be an azetidinyl group, a pyrrolidinyl group, a piperidinyl group, a morpholinyl group, a piperazinyl group, an oxetanyl group, or , tetrahydrofuranyl group, tetrahydrothienyl group, tetrahydropyranyl group, thiomorpholinyl group, etc.
  • the substituent of the saturated heterocyclic group may be a halogen atom, a hydroxy group, a methyl group, a methoxy group, a methanesulfonyl group, etc.
  • Examples include a 2-hydroxyethyl group, an oxetanyl group, a tetrahydropyranyl group, a methyl-piperidinyl group, a methyl-piperazinyl group, a morpholinyl group, and a thiomorpholinyl group.
  • the amino group which may have a substituent in R 1 is represented by the atomic group -NR 13 R 14 , R 13 and R 14 each independently represent a C 1-6 alkyl, The 6- alkyl may be further substituted with a dimethylamino group, a methoxy group, etc.
  • a preferred embodiment of the amino group which may have a substituent is a dimethylamino group.
  • R2 R2 represents an aryl group which may have a substituent, a heteroaryl group which may have a substituent, or an alkynyl group which may have a substituent.
  • Examples of the aryl group moiety of the aryl group which may have a substituent include a phenyl group and a naphthyl group.
  • the heteroaryl group portion of the heteroaryl group which may have a substituent is a 4- to 6-membered heteroaryl ring containing 1 to 4 heteroatoms arbitrarily selected from a sulfur atom, a nitrogen atom, and an oxygen atom.
  • the formula includes a pyrrolyl group, a furanyl group, a thienyl group, an imidazolyl group, a pyrazolyl group, an oxazolyl group, a thiazolyl group, an imidazolyl group, a triazolyl group, a tetrazolyl group, a pyridyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, a triazinyl group, etc. are examples.
  • the alkynyl group portion of the alkynyl group which may have a substituent is a straight-chain or branched-chain unsaturated hydrocarbon having at least one carbon-carbon triple bond and 2 to 6 carbon atoms. Specifically, it means an ethynyl group, a 2-propynyl group, a 2-butynyl group, a 2-pentynyl group, a 3-pentynyl group, a 2-hexynyl group, a 3-hexynyl group. etc. are examples.
  • the substituents in the above-mentioned aryl group which may have a substituent, the heteroaryl group which may have a substituent, and the alkynyl group which may have a substituent are preferably C1
  • the substituents of the C 1-6 alkyl and C 1-6 alkoxy include halogen, cyano, hydroxyl, and benzyl.
  • the substituents include 1 to 3 halogens, a hydroxyl group, a cyclopropyl group, a phenyl group, etc.
  • Examples of the substituents that form a ring include a methylenedioxy group or an ethylenedioxy group. Examples can be given.
  • Z Z represents an alkyl group which may have a substituent, an alkenyl group which may have a substituent, an alkynyl group which may have a substituent, a saturated heterocyclic group which may have a substituent, or a cycloalkyl group which may have a substituent.
  • the alkyl group portion of the alkyl group which may have a substituent is a C 1-6 alkyl.
  • Examples of the substituent of this C 1-6 alkyl include a C 3-6 cycloalkyl group, a halogeno C 3-6 cycloalkyl group, a C 1-3 alkoxy group, a halogen atom, a hydroxy group, a benzyloxy group, a methylamino group, a dimethylamino group, a pyranyl group, a piperidinyl group, and a C 1-3 alkylsulfonyl group.
  • the alkenyl group moiety of the alkenyl group which may have a substituent may be any of linear or branched alkenyl groups having 2 to 6 carbon atoms, such as an allyl group, a 2-butenyl group, an isobutenyl group, a 2-pentenyl group, a 3-pentenyl group, and a 2-hexenyl group.
  • the alkynyl group which may have a substituent in Z is the same as the alkynyl group which may have a substituent in R2 .
  • the cycloalkyl group portion of the cycloalkyl group which may have a substituent may be any cyclic saturated hydrocarbon group having 3 to 8 carbon atoms (C cycloalkyl ), with a cyclopropyl group being particularly preferred.
  • Examples of the substituent of this C cycloalkyl include halogen atoms.
  • the saturated heterocyclic group portion of the optionally substituted saturated heterocyclic group may be any 4- to 6-membered saturated heterocyclic group containing 1 to 3 heteroatoms arbitrarily selected from sulfur atoms, nitrogen atoms, and oxygen atoms, and may be substituted at any chemically substitutable position.
  • pyrrolidinyl group examples include a pyrrolidinyl group, a piperidinyl group, a morpholinyl group, a piperazinyl group, an oxetanyl group, a tetrahydrofuranyl group, a tetrahydrothienyl group, a tetrahydropyranyl group, a thiomorpholinyl group, and the like.
  • An embodiment of the compound (I) of the present invention is the above thiazole derivative (I) or a pharma- ceutically acceptable salt thereof, wherein Z is a cycloalkyl group which may be substituted.
  • An embodiment of compound (I) of the present invention is the above thiazole derivative (I) or a pharma- ceutically acceptable salt thereof, wherein Z is an alkyl group which may have a substituent.
  • Another embodiment of the compound (I) of the present invention is the above thiazole derivative (I) or a pharma- ceutically acceptable salt thereof, wherein R2 is a phenyl group which may be substituted.
  • Another embodiment of the compound (I) of the present invention is the above thiazole derivative (I) or a pharma- ceutically acceptable salt thereof, wherein R2 is an alkynyl group which may be substituted.
  • Another embodiment of the compound (I) of the present invention is the thiazole derivative (I) or a pharma- ceutically acceptable salt thereof, which is selected from the group consisting of Examples 1 to 226.
  • Compound (I) of the present invention may have isomers depending on, for example, the type of substituent.
  • the chemical structure of only one form of the isomer may be described, but the present invention also includes all isomers (geometric isomers, optical isomers, tautomers, etc.) that may occur due to the structure, and also includes a single isomer or a mixture thereof.
  • Pharmaceutically acceptable salts of compound (I) of the present invention include inorganic acid salts with hydrochloric acid, sulfuric acid, carbonic acid, phosphoric acid, etc., and organic acid salts with formic acid, fumaric acid, maleic acid, methanesulfonic acid, p-toluenesulfonic acid, etc.
  • the present invention also includes alkali metal salts with sodium, potassium, etc., alkaline earth metal salts with magnesium, calcium, etc., organic amine salts with lower alkylamines, lower alcoholamines, etc., basic amino acid salts with lysine, arginine, ornithine, etc., as well as ammonium salts.
  • Compound (I) of the present invention and its pharma- ceutical acceptable salts can be prepared, for example, by the following method. In the preparation methods shown below, if the defined groups change under the conditions of the method or are not suitable for carrying out the method, the compound can be easily prepared by applying a method commonly used in organic synthesis chemistry, such as protection and deprotection of functional groups [T. W. Greene, Protective Groups in Organic Synthesis 4th Edition, John Wiley & Sons, Inc., 2007]. In addition, the order of reaction steps such as introduction of substituents can be changed as necessary.
  • the compound (I) of the present invention can be produced by Ullmann condensation reaction or nucleophilic substitution reaction between the compound (II) and a compound R 2 -X. That is, when R 2 is an aryl group which may have a substituent or a heteroaryl group which may have a substituent, a mixture of compound (II) and compound R 2 -X is subjected to Ullmann condensation reaction in a solvent in the presence of a metal catalyst such as copper, using a base and an additive as necessary, to produce compound (I).
  • the solvent used in the reaction may be any solvent inert to the reaction, and is not particularly limited, but preferably includes dioxane, toluene, dimethoxyethane, DMF, and the like.
  • Compound R 2 -X is preferably used in a molar equivalent or excess amount relative to compound (II), more preferably 1 to 10 molar equivalents.
  • a base may be added to accelerate the reaction as necessary, and sodium carbonate, cesium carbonate, potassium carbonate, and the like are usually used as the base.
  • the amount of the base used is 1 to 10 molar equivalents relative to compound (II), preferably 1 to 5 molar equivalents.
  • the metal catalyst a commercially available copper catalyst (e.g., CuI, etc.) used in coupling can be used, and it is preferable to add a catalytic amount to compound (II).
  • the reaction can be carried out by heating at 30 to 150° C. for 1 to 48 hours, preferably at 80 to 130° C.
  • Compound (I) can also be produced under Buchwald-Hartwig reaction conditions using a palladium catalyst instead of a copper catalyst.
  • Compound (I) can also be produced by a Chan-Lam-Evans coupling reaction using a compound in which the halogen X of compound R 2 -X is converted to a boronic acid or a boronic acid ester thereof.
  • compound (I) of the present invention can be obtained by subjecting compound (II) to a nucleophilic substitution reaction in the presence of 1 to 5 molar equivalents, preferably 1 to 2.5 molar equivalents of compound R 2 -X and 1 to 5 molar equivalents, preferably 1 to 3.5 molar equivalents of a base such as cesium carbonate, in a solvent.
  • the solvent may be any solvent inactive to the reaction, and is not particularly limited, but preferably DMSO or DMF can be used.
  • the reaction can be carried out by heating at 30 to 150° C. for 1 to 24 hours, and preferably at 50 to 80° C. for 1 to 7 hours.
  • Compound (I) of the present invention can also be produced by the method shown in Scheme 2, for example, using compound (III). (In the formula, R 1 , R 2 and Z are as defined above.)
  • Compound (I) of the present invention can be produced by Chan-Lam-Evans coupling reaction using compound (III) and boronic acid (IV). That is, compound (I) can be produced by reacting a mixture of compound (III) and boronic acid (IV) in a solvent in the presence of a metal catalyst such as copper or nickel, using a base and an additive as necessary.
  • the solvent used in the reaction may be any solvent inert to the reaction, and is not particularly limited, but preferably dichloromethane or dichloroethane can be used.
  • the reaction can be carried out by heating at 30 to 150° C. for 1 to 24 hours, preferably at 50 to 80° C. for 1 to 7 hours.
  • the boronyl group of compound (IV) can be replaced with a boronic acid ester group for use in the reaction.
  • Compound (I) can also be produced by subjecting compound (III) to a nucleophilic substitution reaction with a Z-introducing agent in which the boronic acid moiety of compound (IV) is a suitable leaving group, in the presence of a suitable base.
  • the compound (I) of the present invention can also be produced, for example, according to Scheme 3.
  • the compound (I) of the present invention can be produced by converting the ester group of the compound (V) to a carboxamide group by aminolysis reaction. That is, the compound (I) can be produced by reacting the compound (V) with ammonia in a solvent under heating.
  • the solvent may be any solvent inert to the reaction, and is not particularly limited, but preferably ethanol or methanol can be used.
  • ammonia an aqueous solution, an alcohol solution, etc. can be used, or ammonia gas can be directly blown into the reaction solution.
  • the reaction can be carried out by heating at 50 to 150° C. for 1 to 48 hours, preferably at 80 to 100° C. for 1 to 15 hours.
  • the conversion to the carboxamide group can also be carried out by a method commonly used in organic synthesis, namely, hydrolysis of an ester followed by an amidation reaction using a coupling agent.
  • the compound (I) of the present invention can also be produced, for example, according to Scheme 4.
  • Scheme 4 In the formula, R 1 , R 2 and Z are defined as above, and X represents a halogen.
  • Compound (I) of the present invention can be produced by a cross-coupling reaction such as Suzuki coupling reaction using compound (VI) and compound (VII) (for example, see known literature (N. Miyaura, et al., J. Am. Chem. Soc., 107, 972 (1985)., N. Miyaura, A. Suzuki, Chem. Rev. 95, 2457 (1995)) for the conditions of Suzuki coupling reaction).
  • compound (I) can be produced by reacting a mixture of compound (VI) and compound (VII) in a solvent in the presence of a metal catalyst such as palladium or nickel, and using a base and an additive as necessary.
  • a metal catalyst such as palladium or nickel
  • the solvent used in the reaction include THF, dioxane, toluene, dimethoxyethane, methanol, ethanol, and acetonitrile. It is also suitable to use two or more of these solvents in combination, or to further mix them with water.
  • a mixed solvent of dioxane and water, or a mixed solvent of toluene, methanol, and water is preferred.
  • Compound (VII) is preferably used in a molar equivalent or excess amount relative to compound (VI), more preferably 1 to 10 molar equivalents.
  • a base may be added to accelerate the reaction, and sodium carbonate, cesium carbonate, potassium carbonate, cesium fluoride, and the like are usually used as the base.
  • the amount of the base used is 1 to 10 molar equivalents relative to compound (VI), preferably 1 to 5 molar equivalents.
  • a commercially available palladium catalyst e.g., PdCl2 (Amphos), PdCl2 (dppf), Pd2 (dba) 3 , Pd( PPh3 ) 4 , etc.
  • a catalytic amount i.e., 0.1 to 0.5 molar equivalents
  • the boronyl group of compound (VII) can be replaced with a boronic acid ester group for use in the reaction.
  • Compound (VII) which is one of the raw materials in Scheme 4, is a commercially available product or can be obtained by a known method or a method analogous thereto.
  • the compound (Ia) of the present invention in which R 1 in formula (I) is an amino group which may have a substituent, can also be produced according to scheme 5, for example.
  • R 2 , R 13 , R 14 and Z are the same as defined above.
  • Compound (I-a) can be produced by condensation reaction of the carboxyl group of compound (VIII) with amine (IX). That is, compound (VIII) can be obtained by reacting 0.9 to 5 molar equivalents, preferably 1 to 2 molar equivalents, of amine (IX) in a solvent in the presence of an amide condensing agent commonly used in organic chemistry.
  • the solvent may be any solvent inert to the reaction, and is not particularly limited, but for example, DMF, THF, etc. can be used.
  • the reaction can be carried out by stirring at 0 to 150°C for 1 to 48 hours, but preferably by reacting at room temperature to 80°C for 1 hour to overnight.
  • the amine (IX) which is one of the raw materials in Scheme 5, is a commercially available product or can be obtained by a known method or a method similar thereto.
  • compound (I-b) of the present invention having an "amino group which may have a substituent" as a substituent can also be produced using compound (I-c) of the present invention in which R 1 is represented by an alkyl group which may have a substituent and has a halogen as a substituent, for example, according to Scheme 6.
  • R 2 , Z, R 11 and R 12 are the same as defined above, and X represents a halogen.
  • Compound (I-b) can be produced by reacting compound (I-c) with amine (IX).
  • compound (I-c) can be reacted with 0.9 to 5 molar equivalents, preferably 1 to 2 molar equivalents, of amine (IX) in a solvent in the presence of a base.
  • the solvent may be any solvent inert to the reaction, and is not particularly limited.
  • DMF, THF, etc. can be used.
  • the reaction can be carried out at 0 to 150° C. with stirring for 1 to 48 hours, preferably at room temperature to 80° C. for 1 hour to overnight.
  • Compound (II) used as the starting material in Scheme 1 can be produced, for example, by the method shown in Scheme 7.
  • Scheme 7 In the formula, R1 and Z are defined as above, X represents a halogen, and PG represents a protecting group.
  • Compound (II) can be produced by deprotecting the product obtained by a cross-coupling reaction such as Suzuki coupling reaction using compound (X) and compound (VII). That is, a protected form of compound (II) can be obtained by reacting compound (X) and compound (VII) under the same conditions as in Scheme 4.
  • the deprotection reaction of the obtained protected form of compound (II) can be carried out by subjecting it to deprotection conditions generally used in organic chemistry that are suitable for the protecting group used.
  • the boronyl group of compound (VII) can also be replaced with a boronic acid ester group and used in the reaction.
  • Compound (X) used as the starting material in Scheme 7 can be produced, for example, by the method shown in Scheme 8.
  • Z is defined as above, X represents a halogen, R3 represents a lower alkyl group, and PG represents a protecting group.
  • Compound (X) can be produced by converting the ester group of compound (XI) to a carboxamide group by aminolysis reaction. The reaction can be carried out under the same conditions as in Scheme 3. The conversion to the carboxamide group can also be carried out by a method used in ordinary organic synthesis, that is, by hydrolysis of the ester followed by an amidation reaction using a coupling agent.
  • Compound (XI) used as the starting material in Scheme 8 can be produced, for example, by the method shown in Scheme 9.
  • Z is as defined above, X represents a halogen, R3 represents a lower alkyl group, and PG represents a protecting group.
  • Compound (XI) can be produced by treating compound (XII) with a halogenating agent typically used in organic chemistry, such as N-bromosuccinimide (NBS) or N-chlorosuccinimide (NCS), etc. That is, compound (XI) can be obtained by reacting compound (XII) with 0.5 to 2 molar equivalents, preferably 0.8 to 1.1 molar equivalents, of NBS or NCS in a solvent.
  • NBS N-bromosuccinimide
  • NCS N-chlorosuccinimide
  • the solvent may be any solvent inert to the reaction, and is not particularly limited, but preferably dichloromethane or DMF can be used.
  • the reaction can be carried out by stirring at -20 to 30°C for 0.25 to 10 hours, preferably at -10 to 10°C for 0.5 to 5 hours.
  • Compound (XII) used as the starting material in Scheme 9 can be produced, for example, by the method shown in Scheme 10.
  • Z is defined as above, R3 represents a lower alkyl group, and PG represents a protecting group.
  • Compound (XII) can be produced by Chan-Lam-Evans coupling reaction using compound (XIII) and boronic acid (IV). The reaction conditions can be the same as those in Scheme 2. Furthermore, the boronyl group of compound (IV) can be replaced with a boronic acid ester group for use in the reaction.
  • Compound (XII) can also be produced by subjecting compound (XIII) to a nucleophilic substitution reaction with a Z-introducing agent in which the boronic acid moiety of compound (IV) is a suitable leaving group, in the presence of a suitable base.
  • Compound (XIII) used as the starting material in Scheme 10 can be produced, for example, by the method shown in Scheme 11. (In the formula, R3 represents a lower alkyl group, and PG represents a protecting group.) Compound (XIII) can be produced by reacting aminopyrazole (XIV) with thiophosgene, treating with aqueous ammonia to obtain thioamide (XV), and reacting the thioamide (XV) with ethyl bromopyruvate to form a thiazole ring.
  • aminopyrazole (XIV) is reacted with 0.5 to 2 molar equivalents, preferably 0.9 to 1.5 molar equivalents of thiophosgene, if necessary, in the presence of a base, and then an aqueous ammonia solution is added to obtain thioamide (XV).
  • Compound (XIII) can be obtained by reacting this thioamide (XV) with 0.5 to 2 molar equivalents, preferably 0.8 to 1.5 molar equivalents of ethyl bromopyruvate in a solvent such as ethanol in the presence of a base such as potassium carbonate.
  • the aminopyrazole (XIV) used as a starting material in Scheme 11 is either commercially available or can be easily prepared using a method commonly used in organic synthesis using 4-nitropyrazole or the like as a starting material.
  • Compound (III) used as a starting material in Scheme 2 can be produced, for example, by the method shown in Scheme 12.
  • R 1 and R 2 are defined as above, and X represents a halogen.
  • Compound (III) can be produced by a cross-coupling reaction such as Suzuki coupling reaction using compound (XVI) and compound (VII).
  • the cross-coupling reaction can be carried out under the same conditions as in Scheme 4.
  • the boronyl group of compound (VII) can also be replaced with a boronic acid ester group for use in the reaction.
  • Compound (XVI) used as the starting material in Scheme 12 can be produced, for example, by the method shown in Scheme 13. (wherein R2 is as defined above, R3 represents a lower alkyl group, and X represents a halogen.)
  • Compound (XVI) can be produced by converting the ester group of compound (XVII) to a carboxamide group by aminolysis reaction. The reaction can be carried out under the same conditions as in Scheme 3. The conversion to the carboxamide group can also be carried out by a method used in ordinary organic synthesis, that is, hydrolysis of the ester followed by an amidation reaction using a coupling agent.
  • Compound (XVII) used as the starting material in Scheme 13 can be produced, for example, by the method shown in Scheme 14. (wherein R2 is as defined above, R3 represents a lower alkyl group, and X represents a halogen.)
  • Compound (XVII) can be produced by treating compound (XVIII) with a halogenating agent commonly used in organic chemistry. The reaction conditions can be the same as those in Scheme 9.
  • Compound (XVIII) used as a starting material in Scheme 14 can be produced, for example, in Scheme 10, by using an aminopyrazole derivative having an R 2 group instead of the protecting group of compound (XIII) as a starting material.
  • Compound (V) used as a starting material in Scheme 3 can be produced, for example, by the method shown in Scheme 15.
  • R 1 , R 2 and Z are defined as above, R 3 represents a lower alkyl group, PG represents a protecting group, and X represents a halogen.
  • R 2 is an optionally substituted aryl group or an optionally substituted heteroaryl group
  • compound (V) can be produced by deprotecting the protecting group of compound (XIX) and then subjecting the resulting compound to a coupling reaction such as Ullmann condensation reaction or Buchwald-Hartwig reaction with compound R 2 -X.
  • the reaction conditions can be the same as those in Scheme 1.
  • Compound (V) can also be produced by a Chan-Lam-Evans coupling reaction using a compound in which the halogen X of compound R 2 -X is converted to a boronic acid or a boronic acid ester thereof.
  • R 2 is an alkynyl group which may have a substituent
  • compound (V) can be produced by deprotecting the protecting group of compound (XIX) and then subjecting it to a nucleophilic substitution reaction with compound R 2 -X.
  • the reaction can be carried out under the same reaction conditions as in Scheme 1.
  • Compound (XIX) used as the starting material in Scheme 15 can be produced, for example, by the method shown in Scheme 16.
  • R1 and Z are defined as above, R3 represents a lower alkyl group, PG represents a protecting group, and X represents a halogen.
  • Compound (XIX) can be produced by a cross-coupling reaction such as Suzuki coupling reaction using compound (XI) and compound (VII).
  • the cross-coupling reaction can be carried out under the same conditions as in Scheme 4.
  • the boronyl group of compound (VII) can also be replaced with a boronic acid ester group for use in the reaction.
  • Compound (VI) used as the starting material in Scheme 4 can be produced, for example, by the method shown in Scheme 17.
  • R2 and Z are defined as above, PG represents a protecting group, and X represents a halogen.
  • compound (VI) can be produced by deprotecting the protecting group of compound (X) and then subjecting the compound to a coupling reaction such as Ullmann condensation reaction or Buchwald-Hartwig reaction with compound R 2 -X.
  • the reaction conditions can be the same as those in Scheme 1.
  • Compound (VI) can also be produced by a Chan-Lam-Evans coupling reaction using a compound in which the halogen X of compound R 2 -X is converted into a boronic acid or a boronic acid ester thereof.
  • R 2 is an alkynyl group which may have a substituent
  • compound (VI) can be produced by deprotecting the protecting group of compound (X) and then subjecting the compound to a nucleophilic substitution reaction with compound R 2 -X.
  • the reaction can be carried out under the same reaction conditions as in Scheme 1.
  • Compound (VIII) used as the starting material in Scheme 5 can be produced, for example, by the method shown in Scheme 18.
  • R2 and Z are as defined above, and R6 represents lower alkyl.
  • Compound (VIII) can be produced by hydrolyzing the ester group of compound (XX).
  • the hydrolysis reaction can be carried out under reaction conditions generally used in organic chemistry, and alkaline conditions (sodium hydroxide, lithium hydroxide, etc.) or acidic conditions (hydrochloric acid, sulfuric acid, etc.) can be used.
  • Compound (XX) used as the starting material in Scheme 18 can be produced, for example, by the method shown in Scheme 19.
  • R2 and Z are as defined above, R6 represents lower alkyl, and X represents halogen.
  • compound (XX) can be produced by deprotecting the protecting group of compound (XXI) and then subjecting the compound to a coupling reaction such as Ullmann condensation reaction or Buchwald-Hartwig reaction with compound R 2 -X. The reaction can be carried out under the same reaction conditions as in Scheme 1.
  • Compound (XX) can also be produced by a Chan-Lam-Evans coupling reaction using a compound in which the halogen X of compound R 2 -X is converted to a boronic acid or a boronic acid ester thereof.
  • R2 is an alkynyl group which may have a substituent
  • compound (XX) can be produced by deprotecting the protecting group of compound (XXI) and then subjecting the compound to a nucleophilic substitution reaction with compound R2 -X. The reaction can be carried out under the same reaction conditions as in Scheme 1.
  • Compound (XXI) used as the starting material in Scheme 19 can be produced, for example, by the method shown in Scheme 20.
  • Z is as defined above, R6 represents a lower alkyl group, PG represents a protecting group, and X represents a halogen.
  • Compound (XXI) can be produced by a cross-coupling reaction such as Suzuki coupling reaction using compound (X) and compound (XXII). The cross-coupling reaction can be carried out under the same conditions as in Scheme 4.
  • the boronyl group of compound (XXII) can be replaced with a boronic acid ester group and used in the reaction.
  • the compound (Ic) of the present invention used as the starting material in Scheme 6 can be produced from the compound (Id) of the present invention, which is represented by formula (I) in which R 1 is a methyl group, by, for example, the method shown in Scheme 21. (In the formula, R2 and Z are defined as above, and X represents a halogen.)
  • Compound (I-c) can be produced by treating compound (I-d) with a halogenating agent used in ordinary organic chemistry, such as benzyltrimethylammonium dichloroiodide, NBS, NCS, etc.
  • compound (I-c) can be obtained by reacting compound (I-d) with 0.5 to 5.0 molar equivalents, preferably 0.8 to 2.0 molar equivalents of a halogenating agent in a solvent.
  • the solvent may be any one inert to the reaction, and is not particularly limited, and dichloromethane, THF, DMF, etc. can be used.
  • the reaction can be carried out by stirring at -20 to 100°C for 0.25 to 10 hours, but preferably at room temperature to 80°C for 0.5 to 5 hours.
  • the compound (I) of the present invention or a pharma- ceutically acceptable salt thereof can be prepared in the form of a conventional pharmaceutical preparation (pharmaceutical composition) suitable for oral, parenteral or topical administration.
  • a conventional pharmaceutical preparation pharmaceutical composition
  • Preparations for oral administration include solid preparations such as tablets, granules, powders, capsules, and liquid preparations such as syrups. These preparations can be prepared by conventional methods.
  • Solid preparations can be prepared by using conventional pharmaceutical carriers such as lactose, starch such as corn starch, crystalline cellulose such as microcrystalline cellulose, hydroxypropyl cellulose, calcium carboxymethyl cellulose, talc, magnesium stearate, etc.
  • Capsules can be prepared by encapsulating the granules or powders thus prepared.
  • Syrups can be prepared by dissolving or suspending the compound (I) of the present invention or a pharma- ceutically acceptable salt thereof in an aqueous solution containing sucrose, carboxymethyl cellulose, etc.
  • Preparations for parenteral administration include injections such as drip infusions.
  • Injection preparations can also be prepared by conventional methods, and can be appropriately incorporated into isotonic agents (e.g., mannitol, sodium chloride, glucose, sorbitol, glycerol, xylitol, fructose, maltose, mannose), stabilizers (e.g., sodium sulfite, albumin), and preservatives (e.g., benzyl alcohol, methyl p-oxybenzoate).
  • isotonic agents e.g., mannitol, sodium chloride, glucose, sorbitol, glycerol, xylitol, fructose, maltose, mannose
  • stabilizers e.g., sodium sulfite, albumin
  • preservatives e.g., benzyl alcohol, methyl p-oxybenzoate
  • the dose of the compound (I) of the present invention or a pharma- ceutically acceptable salt thereof can be varied according to the severity of the disease, the age and body weight of the patient, the dosage form, etc., but is usually in the range of 1 mg to 1,000 mg per day for an adult, which can be administered orally or parenterally in a single dose, or in two or three divided doses.
  • the compound (I) of the present invention or a pharma- ceutical acceptable salt thereof can be used as an ALK5 inhibitor and as a reagent for experiments and research.
  • the compound (I) of the present invention that is radiolabeled can also be used as a molecular probe for PET.
  • reaction mixture was diluted with water and extracted with ethyl acetate.
  • organic layer was dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the residue was purified using a HPLC preparative chromatography system to obtain the title compound (21 mg).
  • reaction mixture was diluted with water and extracted with ethyl acetate.
  • organic layer was dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the residue was purified using a HPLC preparative chromatography system to obtain the title compound (21.3 mg).
  • Examples 9 to 183 The following example compounds [Table 1] were produced using the corresponding raw materials (commercially available products, or compounds derived from commercially available compounds by known methods or methods similar thereto) according to the methods described in the above examples, and, if necessary, by appropriately combining methods commonly used in organic synthetic chemistry. The physicochemical data of each compound is shown in [Table 2].
  • Step 8 Preparation of 2-[(1-(but-2-yn-1-yl)-1H-pyrazol-4-yl)(cyclopropylmethyl)amino)-5-[4-(dimethylcarbamoyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide Potassium carbonate (103 mg, 0.75 mmol) was added to a solution of 2-[(cyclopropylmethyl)(1H-pyrazol-4-yl)amino]-5-[4-(dimethylcarbamoyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide (150 mg, 0.38 mmol) and 1-bromo-2-butyne (103 mg, 0.75 mmol) in DMF (3 mL), and the mixture was reacted at 120° C.
  • Examples 185 to 226 The following example compounds [Table 3] were produced using the corresponding raw materials (commercially available products, or compounds derived from commercially available compounds by known methods or methods similar thereto) according to the methods described in the above examples, and, if necessary, by appropriately combining methods commonly used in organic synthetic chemistry. The physicochemical data of each compound is shown in [Table 4].
  • TR-FRET time-resolved fluorescence resonance energy transfer
  • a 10 mM DMSO solution of the test compound was further diluted with DMSO to 10 concentrations (0.00003 mM, 0.0001 mM, 0.0003 mM, 0.001 mM, 0.003 mM, 0.01 mM, 0.03 mM, 0.1 mM, 0.3 mM, 1 mM), and each was diluted 25-fold with assay buffer to prepare a drug solution (4% DMSO solution).
  • Drug or control solution (5 ⁇ L), substrate mixture (5 ⁇ L), and enzyme solution (10 ⁇ L) were mixed in wells of a 384-well black plate and reacted at room temperature for 1 hour.
  • the TR-FRET signal was measured using a multimode plate reader (EnVision, PerkinElmer) according to the protocol attached to the kit. (Method of evaluating inhibitory activity) As a blank, an assay buffer was added instead of the enzyme solution to measure. The inhibition rate (%) of the test compound was calculated according to the following formula.
  • Inhibition rate (%) (1 - (C - A) / (B - A)) x 100
  • A, B, and C respectively indicate the TR-FRET signal of a blank well, the TR-FRET signal of a control solution well, and the TR-FRET signal of a compound-added well.
  • the IC 50 value was calculated by regression analysis of the inhibition rate and the test compound concentration (logarithm).
  • the inhibitory activity against ALK5 of representative compounds of the present invention is shown in Table 5.
  • IC50 values of less than 0.05 ⁇ M are indicated by ***, 0.05 ⁇ M or more and less than 0.5 ⁇ M are indicated by **, and 0.5 ⁇ M or more and less than 5 ⁇ M are indicated by *.
  • Test Example 2 Inhibition test of Smad3 phosphorylation by intracellular ALK5 (culture of cells used) A549 cells (ATCC No. CCL-185) were cultured in a T75 flask at 37°C in a 5% CO2 incubator using D-MEM medium (Nacalai, #08459-64) (hereinafter referred to as growth medium) supplemented with 10% fetal bovine serum (hereinafter referred to as FBS) (AusGene) and 1% penicillin-streptomycin (Nacalai).
  • D-MEM medium Nacalai, #08459-644
  • FBS fetal bovine serum
  • penicillin-streptomycin Nacalai
  • the cultured A549 cells were diluted with growth medium to a cell density of 3.3 x 105 cells/mL, and the resulting cell suspension was seeded in a 24-well culture plate (Falcon, 353226) at 3.3 x 105 cells/mL/well, and cultured overnight at 37°C in a 5% CO2 incubator. The next day, the growth medium was removed with an aspirator and immediately washed with FBS-free D-MEM medium (hereinafter referred to as medium) kept at 37°C. The cell plate was prepared by adding 0.989 mL of medium to each well.
  • TGF ⁇ 1 (Peprotec, PEP-100-21-10) adjusted to 200 ng/mL in culture medium was added to each well of the cell plate (10 ⁇ L of culture medium alone was added to the control well), and the plate was further incubated for 30 minutes at 37° C. in a 5% CO 2 incubator.
  • the mixture was mixed with SDS-sample buffer and reacted at 95°C for 5 minutes to denature the proteins, and a sample solution was prepared. 5 ⁇ g/12 ⁇ L of the sample solution was applied to each well of a 5-20% gradient acrylamide gel (Nacalai, No. 13064-04) and electrophoresis was performed. The proteins in the gel were then transferred to a PVDF membrane using a Transblot Turbo transfer system (Bio-Rad). (Detection of phosphorylated Smad3) The transferred PVDF membrane was subjected to blocking treatment with 2% ECL prime blocking agent (GE Healthcare), and then reacted overnight at 4° C.
  • ECL prime blocking agent GE Healthcare
  • the combinations and dilution concentrations of the primary and secondary antibodies used in this test are as follows. (Method of evaluating inhibitory activity) The detected bands were quantified by densitometry (ImageQuant TL analysis software), and the inhibition rate was calculated from the band intensity in each group, taking the luminescence of the phosphorylated Smad3 band in the compound-free and TGF ⁇ -stimulated group as 100% and the luminescence of the phosphorylated Smad3 band in the compound-free and TGF ⁇ -unstimulated group as 0%. Each phosphorylated Smad3 band was corrected with ⁇ -actin. The inhibition rate (%) of Smad3 phosphorylation by the test compound was calculated according to the following formula.
  • Inhibition rate (%) (1 - (C - A) / (B - A)) x 100
  • A, B, and C respectively show the luminescence of the phosphorylated Smad3 band in the group without addition of compound and without stimulation with TGF ⁇ , the group without addition of compound and stimulation with TGF ⁇ , and the group with addition of compound and stimulation with TGF ⁇ .
  • the IC 50 value was calculated by regression analysis of the inhibition rate and the test compound concentration (logarithm).
  • the inhibitory activity of intracellular Smad3 phosphorylation of representative compounds of the present invention is shown in Table 7.
  • IC50 values of less than 0.1 ⁇ M are indicated by ***, those of 0.1 ⁇ M or more and less than 0.3 ⁇ M are indicated by **, and those of 0.3 ⁇ M or more are indicated by *.
  • Test Example 3 Inhibition test of regulatory T cell (Treg) differentiation induction by TGF- ⁇ stimulation
  • Spleens were collected from mice (BALB/cCrSlc, female, 7 weeks old), and mouse lymphocytes were separated from a suspension of splenocytes (FBS-free IMDM medium) prepared using a cell strainer using HISTOPAQUE-1083 (Sigma).
  • CD4 positive T cells were isolated from these mouse lymphocytes using EasySep Mouse Naive CD4+ T Cell Isolation Kit (manufactured by STEMCELL technologies) according to the protocol attached to the kit.
  • a compound solution prepared by diluting a DMSO solution of a test compound 100-fold with IMDM medium supplemented with IL-2-containing FBS was added to each well.
  • CD4-positive T cells were suspended in IMDM medium supplemented with IL-2-containing FBS (2 ⁇ 10 5 cells/mL), 0.1 mL of the solution was added to each well, and the mixture was incubated for 1 hour in a CO 2 incubator.
  • the cells were collected from each well, stained with FITC anti-mouse CD4 Antibody (BioLegend), APC anti-mouse CD25 Antibody (BioLegend), and FOXP3 Monoclonal Antibody (FJK-16s) PE (eBioscience), and the proportion of the Treg fraction (CD4+/CD25+/Foxp3+) was measured using a flow cytometer.
  • IMDM medium supplemented with IL-2-containing FBS was used instead of TGF- ⁇ 1.
  • the IC 50 value was calculated by regression analysis of the change in the proportion of the Treg fraction and the test compound concentration (logarithm).
  • the inhibitory activity of representative compounds of the present invention in inducing Treg differentiation by TGF- ⁇ stimulation is shown in Table 8.
  • IC50 values of less than 0.1 ⁇ M are indicated by ***, 0.1 ⁇ M or more and less than 0.5 ⁇ M by **, and 0.5 ⁇ M or more by *.
  • Table 8 In this test, as shown in Table 8, compound (I) of the present invention inhibited TGF- ⁇ signaling in naive T cells and strongly suppressed the induction of differentiation into Treg by TGF- ⁇ stimulation.
  • the results of Test Example 3 show that compound (I) of the present invention inhibits intracellular TGF- ⁇ signaling and has a strong suppressive effect on the induction of Treg differentiation.
  • Test Example 4 Effect of Combination with Anti-PD-1 Antibody in an Allograft Mouse Model of Mouse Colon Cancer Cell Line CT26.WT
  • the effect of combination with an immune checkpoint inhibitor in cancer immunotherapy was examined using a syngeneic mouse tumor model (subcutaneous transplant) of the mouse colon cancer cell line CT26.WT.
  • CT26.WT cells were adjusted to a cell density of 1 x 107 cells/mL in HBSS(-) medium (manufactured by Nacalai) to prepare a cell preparation for transplantation.
  • 0.1 mL of this cell preparation for transplantation was subcutaneously transplanted into the back of a BALB/cCrslc mouse (female, 7 weeks old, Japan SLC).
  • mice Four days after the cancer cells were transplanted, the mice were divided into groups so that the average tumor volume (see the calculation formula below) of the cancer-bearing mice was close to each other.
  • Anti-PD-1 antibody Bio X Cell, clone RMP1-14; catalog No.
  • BE0146 was prepared with physiological saline to a concentration of 1 mg/mL immediately before administration.
  • Each mouse (6 mice per group) transplanted with cancer cells was forced to orally administer 0.1 mL of the test substance (100 mg/kg) or the solvent per 10 g of body weight on the day once a day from the 4th to the 20th day after transplantation. Drugs were suspended on the 9th, 10th, 16th, and 17th days after transplantation.
  • the antibody administration group and the drug combination group were intraperitoneally administered 0.1 mL of the antibody solution (10 mg/kg) per 10 g of body weight on the day twice a week (5 times in total), and saline was administered intraperitoneally to the other groups.
  • FIG. 1 shows the change in tumor volume over time in each group.
  • the compounds of the present invention, Example 6 and Example 7, showed a significant tumor growth suppression/tumor regression effect when used in combination with an anti-PD-1 antibody. This confirmed that the compounds of the present invention showed an excellent antitumor effect when used in combination with an immune checkpoint inhibitor, and are useful in the treatment of cancer.
  • Test Example 5 Combination effect of anti-PD-1 antibody in allograft mouse model of mouse colon cancer cell line CT26.WT 2 (Preparation of tumor-bearing model) CT26.WT cells were adjusted to a cell density of 1 x 107 cells/mL with HBSS(-) medium to prepare a cell preparation for transplantation. 0.1 mL of this cell preparation for transplantation was subcutaneously transplanted into the back of a BALB/cCrslc mouse (female, 7 weeks old, Japan SLC). On the third day after the cancer cells were transplanted, the mice were divided into groups so that the average tumor volume (see the calculation formula in Test Example 4) of the cancer-bearing mice was close to each other.
  • test substance preparation of sample solution for administration of test substance
  • Each mouse transplanted with cancer cells (10 mice in the solvent group, 9 mice in the compound group of Example 184, 9 mice in the antibody PD-1 antibody group, and 8 mice in the compound of Example 184 and antibody PD-1 antibody combination group) was forcibly administered 0.1 mL of the test substance (30 mg/kg) or the solvent per 10 g of body weight on the day once a day from the 3rd to the 21st day after transplantation. Drugs were suspended on the 8th, 9th, 15th, and 16th days after transplantation.
  • the antibody administration group and the drug combination group were intraperitoneally administered 0.1 mL of antibody solution (10 mg/kg) per 10 g of body weight on the day twice a week (6 times in total), and saline was intraperitoneally administered to the other groups.
  • the mice were observed until the 21st day, and the tumor diameter was measured several times a week to calculate the tumor volume of each mouse using the formula in Test Example 4 to evaluate the antitumor effect.
  • Figure 2 shows the change in tumor volume over time in each group.
  • the compound of the present invention, Example 184 showed a significant tumor growth inhibitory and tumor regression effect when used alone or in combination with an anti-PD-1 antibody. This confirmed that the compound of the present invention showed an excellent antitumor effect when used in combination with an immune checkpoint inhibitor, and is useful in the treatment of cancer.
  • the compounds provided by the present invention are useful for treating diseases known to be associated with abnormal cell responses via the TGF ⁇ signal pathway, particularly cancer, and are also useful for preventing tumor metastasis and recurrence by targeting cancer stem cells. Furthermore, when used in combination with immune checkpoint inhibitors, they are useful for expanding and enhancing the therapeutic effect in cancer immunotherapy. They are also useful for treating and preventing fibrotic diseases, etc. Furthermore, they are useful as experimental and research reagents as ALK5 inhibitors and TGF ⁇ signal inhibitors.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention provides a thiazole derivative that has an ALK5 inhibitory action and that is represented by formula (1) (refer to the description with respect to R1, R2, and Z in the formula), or a pharmaceutically acceptable salt thereof.

Description

新規チアゾール誘導体New thiazole derivatives

 本発明は、医薬、特にTGFβI型受容体(ALK5)阻害作用を有する新規チアゾール誘導体またはその薬学的に許容される塩に関するものである。 The present invention relates to a medicine, in particular to a novel thiazole derivative or a pharma- ceutical acceptable salt thereof that has TGFβ type I receptor (ALK5) inhibitory activity.

 多機能性サイトカインであるトランスフォーミング増殖因子β(TGF-β)は、哺乳動物で3種類のアイソフォーム(TGF-β1、β2、β3)が存在し、細胞増殖、分化の制御、遊走や接着などの生理機能に関与している(非特許文献1)。TGF-βは、セリン/スレオニンキナーゼ領域を持つ一回膜貫通型のI型受容体(ALK5)とII型受容体で形成される複合体に結合し、細胞内にシグナルを伝達する。TGF-βの結合により、ALK5がII型受容体により活性化されると、ALK5は、細胞内で転写因子であるSmad2やSmad3をリン酸化する。リン酸化されたSmad2やSmad3は、Smad4と複合体を形成したのち、核へ移行し、直接または他の転写因子とともに標的遺伝子の転写を誘導する(非特許文献1、2)。
近年の研究から、TGF-βシグナルは、創傷治癒、炎症・免疫、癌の浸潤転移など病態においても、重要な役割を果たしていることが報告されている(非特許文献3)。特にがん細胞においては、上皮間葉転換(EMT)の促進、血管新生、がん幹細胞の維持に直接関与していることが報告されている(非特許文献4、5)。
例えば、骨肉腫の増殖にTGF-βが関わっていることが知られており、TGF-βで刺激した骨肉腫細胞の増殖をALK5阻害剤が抑制することが報告されている(非特許文献6)。また、抗TGF-β抗体が骨肉腫の肺転移を抑制することも報告されていることから(非特許文献7)、ALK5阻害剤は、TGF-βが関わっている固形がんや転移などの治療に有用であると考えられる。
Transforming growth factor β (TGF-β), a multifunctional cytokine, exists in three isoforms (TGF-β1, β2, β3) in mammals and is involved in physiological functions such as cell proliferation, differentiation control, migration, and adhesion (Non-Patent Document 1). TGF-β binds to a complex formed by a single-pass transmembrane type I receptor (ALK5) with a serine/threonine kinase domain and a type II receptor, and transmits a signal into the cell. When ALK5 is activated by the type II receptor upon binding of TGF-β, ALK5 phosphorylates the transcription factors Smad2 and Smad3 in the cell. After forming a complex with Smad4, phosphorylated Smad2 and Smad3 migrate to the nucleus and induce the transcription of target genes directly or together with other transcription factors (Non-Patent Documents 1 and 2).
Recent studies have reported that TGF-β signaling plays an important role in pathological conditions such as wound healing, inflammation/immunity, and cancer invasion/metastasis (Non-Patent Document 3). In particular, it has been reported that TGF-β signaling is directly involved in the promotion of epithelial-mesenchymal transition (EMT), angiogenesis, and the maintenance of cancer stem cells in cancer cells (Non-Patent Documents 4 and 5).
For example, it is known that TGF-β is involved in the proliferation of osteosarcoma, and it has been reported that ALK5 inhibitors suppress the proliferation of osteosarcoma cells stimulated with TGF-β (Non-Patent Document 6). In addition, it has been reported that anti-TGF-β antibodies suppress lung metastasis of osteosarcoma (Non-Patent Document 7), so it is considered that ALK5 inhibitors are useful for the treatment of solid cancers and metastasis in which TGF-β is involved.

 近年、がんの治療において、抗CTLA-4(Cyototoxic T lymphocyte antigen 4)抗体、抗PD-1(Programmed death receptor 1)抗体や抗PD-L1(Programmed death ligand 1)抗体などの免疫チェックポイント阻害剤を用いて、ホストの抗腫瘍免疫応答を増強させて抗腫瘍効果を得るがん免疫療法が注目されている。TGF-βは、免疫制御をつかさどる制御性T細胞(Treg)の分化誘導を制御しており、がんの微小環境において、Treg細胞は抗腫瘍免疫を抑制し、腫瘍増殖を促進すると考えられている(非特許文献8)。したがって、TGF-βシグナルを抑制することは、癌免疫療法による抗腫瘍効果や免疫チェックポイント阻害剤との併用効果が期待される。
またTGF-βは、正常な組織修復過程の病的過剰状態である線維化にも関与しており、TGF-βの異常な過剰シグナルは線維症の病因になっていることが報告されている(非特許文献9)。
 従って、ALK5阻害活性を有する化合物は、TGF-βシグナルが関与している疾患、例えば、がんの治療や、免疫チェックポイント阻害剤などと併用することで、がん免疫療法において治療効果の拡大および増強に有用である。また、線維化疾患等の治療や予防に有用である。
In recent years, in the treatment of cancer, attention has been focused on cancer immunotherapy, which uses immune checkpoint inhibitors such as anti-CTLA-4 (cytotoxic T lymphocyte antigen 4) antibody, anti-PD-1 (programmed death receptor 1) antibody, and anti-PD-L1 (programmed death ligand 1) antibody to enhance the host's antitumor immune response and obtain an antitumor effect. TGF-β controls the differentiation induction of regulatory T cells (Treg), which are responsible for immune control, and it is thought that in the cancer microenvironment, Treg cells suppress antitumor immunity and promote tumor growth (Non-Patent Document 8). Therefore, suppressing TGF-β signaling is expected to have an antitumor effect by cancer immunotherapy and a combined effect with immune checkpoint inhibitors.
TGF-β is also involved in fibrosis, a pathological excess state of the normal tissue repair process, and it has been reported that abnormal excess signaling of TGF-β is the cause of fibrosis (Non-Patent Document 9).
Therefore, a compound having an ALK5 inhibitory activity is useful for treating diseases in which TGF-β signaling is involved, such as cancer, and for expanding and enhancing the therapeutic effect in cancer immunotherapy by using it in combination with an immune checkpoint inhibitor, etc. It is also useful for treating and preventing fibrotic diseases, etc.

Massague,J.,Annu.Rev.Biochem.,1998,67,753-791.Massague, J., Annu. Rev. Biochem., 1998, 67, 753-791. Derynck,R.,Zhang,Y.E.,Nature,2003,425,577.Derynck, R. , Zhang, Y. E. , Nature, 2003, 425, 577. Akhurst,R.J.,Hata,A.,Nat.Rev.Drug Discov.,2012,11,790-811.Akhurst, R. J., Hata, A., Nat. Rev. Drug Discov., 2012, 11, 790-811. Xu,J.,Lamouille, S., Derynck, R.,Cell Res.,2009,19,156-172.Xu, J. , Lamouille, S. , Derynck, R. , Cell Res. , 2009, 19, 156-172. Padua, D.,Massague,J., Cell Res.,2009,19,89-102.Padua, D., Massague, J., Cell Res., 2009, 19, 89-102. Matsuyama,S.,et al.,Cancer Res.,2003,63(22),7791-7798.Matsuyama, S., et al., Cancer Res., 2003, 63(22), 7791-7798. Kawano,M.,et al.,Clin.Orthop.Relat.Res.,2012,470(8):2288-2294.Kawano, M. , et al. , Clin. Orthop. Relat. Res. , 2012, 470(8):2288-2294. Tauriello,D.V.F.,Palomo-Ponce,S.,Stork,D.,Berenguer-Llergo,A.,Nature,2018,554,538-543.Tauriello, D.V.F., Palomo-Ponce, S., Stork, D., Berenguer-Llergo, A. , Nature, 2018, 554, 538-543. Verrecchia,F,Mauviel,A., World J. Gastroenterol.,2007,13(22),3056-3062.Verrecchia, F, Mauviel, A., World J. Gastroenterol., 2007, 13(22), 3056-3062.

 本発明は、医薬、特にTGFβI型受容体(ALK5)阻害作用を有する新規チアゾール誘導体またはその薬学的に許容される塩を提供することを課題とする。 The objective of the present invention is to provide a pharmaceutical, in particular a novel thiazole derivative or a pharma- ceutical acceptable salt thereof that has TGFβ type I receptor (ALK5) inhibitory activity.

 本発明は、以下のチアゾール誘導体またはその薬学的に許容される塩によって達成される。
(1)下式(I):

Figure JPOXMLDOC01-appb-C000002
(式中、Rは置換基を有してもよいアルキル基、置換基を有してもよい飽和ヘテロ環式基または置換基を有してもよいアミノ基を表わし、Rは置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基または置換基を有してもよいアルキニル基を表し、Zは置換基を有してもよいアルキル基、置換基を有してもよいアルケニル基、置換基を有してもよいアルキニル基、置換基を有してもよい飽和ヘテロ環式基または置換基を有してもよいシクロアルキル基を表す。)で示されるチアゾール誘導体またはその薬学的に許容される塩。
(2)Rが置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基である、上記(1)に記載のチアゾール誘導体またはその薬学的に許容される塩。
(3)Rが置換基を有してもよいアルキニル基である、上記(1)に記載のチアゾール誘導体またはその薬学的に許容される塩。
(4)後記実施例1から226よりなる群から選択される、チアゾール誘導体またはその薬学的に許容される塩。 The present invention can be achieved by the following thiazole derivatives or pharma- ceutically acceptable salts thereof:
(1) The following formula (I):
Figure JPOXMLDOC01-appb-C000002
(wherein R 1 represents an optionally substituted alkyl group, an optionally substituted saturated heterocyclic group or an optionally substituted amino group; R 2 represents an optionally substituted aryl group, an optionally substituted heteroaryl group or an optionally substituted alkynyl group; and Z represents an optionally substituted alkyl group, an optionally substituted alkenyl group, an optionally substituted alkynyl group, an optionally substituted saturated heterocyclic group or an optionally substituted cycloalkyl group), or a pharmacy acceptable salt thereof.
(2) The thiazole derivative or a pharma- ceutically acceptable salt thereof according to the above (1), wherein R2 is an aryl group which may have a substituent, or a heteroaryl group which may have a substituent.
(3) The thiazole derivative or a pharma- ceutically acceptable salt thereof according to the above (1), wherein R2 is an alkynyl group which may have a substituent.
(4) A thiazole derivative or a pharma- ceutically acceptable salt thereof selected from the group consisting of Examples 1 to 226 described below.

 本発明者らは、上記の課題を解決するために種々検討を重ねた結果、前記式(I)で示される新規なチアゾール誘導体およびその薬学的に許容される塩が、ALK5阻害作用を示すことを見出し、本発明を完成させた。本発明により提供される化合物は、TGFβシグナル経路を介した異常な細胞応答に関連していることが知られている疾患、特にがんの治療に有用であり、また、がん幹細胞を標的とした腫瘍の転移および再発予防にも有用である。さらに免疫チェックポイント阻害剤などと併用することで、がん免疫療法において治療効果の拡大および増強に有用である。また、線維化疾患等の治療や予防に有用である。また、ALK5阻害剤、TGFβシグナル阻害剤として、実験用、研究用の試薬としても有用である。 The present inventors conducted various studies to solve the above problems, and found that the novel thiazole derivative represented by the above formula (I) and its pharma- ceutical acceptable salt exhibit ALK5 inhibitory activity, and thus completed the present invention. The compounds provided by the present invention are useful for treating diseases known to be associated with abnormal cell responses via the TGFβ signal pathway, particularly cancer, and are also useful for preventing tumor metastasis and recurrence by targeting cancer stem cells. Furthermore, by using them in combination with immune checkpoint inhibitors, they are useful for expanding and enhancing the therapeutic effect in cancer immunotherapy. They are also useful for treating and preventing fibrotic diseases and the like. They are also useful as experimental and research reagents as ALK5 inhibitors and TGFβ signal inhibitors.

実施例代表化合物と抗PD-1抗体との併用における、腫瘍増殖抑制・腫瘍退縮効果を示す(試験例4)Showing the tumor growth suppression and tumor regression effect in the combined use of a representative compound of the embodiment and an anti-PD-1 antibody (Test Example 4) 実施例代表化合物の腫瘍増殖抑制・腫瘍退縮効果(単剤および抗PD-1抗体との併用)を示す(試験例5)Tumor growth inhibition and tumor regression effects of representative compounds (single agent and in combination with anti-PD-1 antibody) are shown (Test Example 5)

 以下、本発明を詳細に説明する。
 本発明の新規なチアゾール誘導体は、下式(I):

Figure JPOXMLDOC01-appb-C000003
(式中、Rは置換基を有してもよいアルキル基、置換基を有してもよい飽和ヘテロ環式基または置換基を有してもよいアミノ基を表わし、Rは置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基または置換基を有してもよいアルキニル基を表し、Zは置換基を有してもよいアルキル基、置換基を有してもよいアルケニル基、置換基を有してもよいアルキニル基、置換基を有してもよい飽和ヘテロ環式基または置換基を有してもよいシクロアルキル基を表す。)
で示される化合物である。 The present invention will be described in detail below.
The novel thiazole derivative of the present invention has the following formula (I):
Figure JPOXMLDOC01-appb-C000003
(In the formula, R1 represents an alkyl group which may have a substituent, a saturated heterocyclic group which may have a substituent, or an amino group which may have a substituent, R2 represents an aryl group which may have a substituent, a heteroaryl group which may have a substituent, or an alkynyl group which may have a substituent, and Z represents an alkyl group which may have a substituent, an alkenyl group which may have a substituent, an alkynyl group which may have a substituent, a saturated heterocyclic group which may have a substituent, or a cycloalkyl group which may have a substituent.)
It is a compound represented by the formula:

 本願明細書において使用する術語を、以下に、より詳細に説明する。
 以下の説明において、「ハロゲン」とは、フッ素原子、塩素原子、臭素原子、またはヨウ素原子を意味する。
 「C1-6アルキル」とは、炭素原子数が1-6の直鎖状または分枝状の飽和炭化水素基を意味し、「Cアルキル」とは、炭素原子数が6の飽和炭化水素基を意味する。他の数字の場合も同様である。「C1-3アルキル」とは、具体的には、メチル基、エチル基、プロピル基またはイソプロピル基である。「C1-6アルキル」としては、例えば、上記「C1-3アルキル」に加えて、ブチル基、1-メチルプロピル基、2-メチルプロピル基、tert-ブチル基、ペンチル基、1,1-ジメチルプロピル基、1,2-ジメチルプロピル基、1-メチルブチル基、2-メチルブチル基、4-メチルペンチル基、3-メチルペンチル基、2-メチルペンチル基、1-メチルペンチル基、n-ヘキシル基等を挙げることができる。
 同様に「C3-8シクロアルキル」とは、炭素原子数が3-8の環状飽和炭化水素基を意味する。他の数字の場合も同様である。その具体例としては、シクロプロピル基、シクロブチル基、シクロペンチル基、シクロヘキシル基、シクロヘプチル基、シクロオクチル基等が挙げられる。
 また、「C1-6アルコキシ」とは「C1-6アルキルオキシ」を意味し、当該「C1-6アルキル」部分は、前記「C1-6アルキル」と同義である。他の数字の場合も同様である。
 置換基としては、特に記載のない限り、1または2個以上の任意の種類の置換基を、化学的に可能な任意の位置に有することができ、置換基が2個以上の場合、それぞれの置換基は同一であっても異なっていてもよい。
The terminology used in this specification is explained in more detail below.
In the following description, "halogen" means a fluorine atom, a chlorine atom, a bromine atom, or an iodine atom.
"C 1-6 alkyl" means a linear or branched saturated hydrocarbon group having 1-6 carbon atoms, and "C 6 alkyl" means a saturated hydrocarbon group having 6 carbon atoms. The same applies to other numbers. "C 1-3 alkyl" specifically means a methyl group, an ethyl group, a propyl group, or an isopropyl group. In addition to the above "C 1-3 alkyl", examples of "C 1-6 alkyl" include a butyl group, a 1-methylpropyl group, a 2-methylpropyl group, a tert-butyl group, a pentyl group, a 1,1-dimethylpropyl group, a 1,2-dimethylpropyl group, a 1-methylbutyl group, a 2-methylbutyl group, a 4-methylpentyl group, a 3-methylpentyl group, a 2-methylpentyl group, a 1-methylpentyl group, and an n-hexyl group.
Similarly, "C 3-8 cycloalkyl" refers to a cyclic saturated hydrocarbon group having 3 to 8 carbon atoms. The same applies to other numbers. Specific examples include a cyclopropyl group, a cyclobutyl group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, and a cyclooctyl group.
In addition, "C 1-6 alkoxy" means "C 1-6 alkyloxy", and the "C 1-6 alkyl" portion has the same meaning as the above-mentioned "C 1-6 alkyl". The same applies to other numbers.
Unless otherwise specified, the substituents may be one or more of any type of substituent at any chemically possible position, and when there are two or more substituents, the respective substituents may be the same or different.

 1.R
 Rは置換基を有してもよいアルキル基、置換基を有してもよい飽和ヘテロ環式基または置換基を有してもよいアミノ基を表わす。
 置換基を有してもよいアルキル基のアルキル基部分としては、C1-6アルキルのいずれでもよい。特に好ましいのは、C1-3アルキルである。
 当該C1-6アルキルまたはC1-3アルキルにおける好ましい置換基の具体例として、置換アミノ基-NR1112を挙げることができる。ここで、R11、R12としては、それぞれ独立して、4~6員の含酸素飽和ヘテロ環またはC1-6アルキルを表すか、またはそれぞれ結合する窒素原子と一緒になり、さらに窒素原子または酸素原子を追加的に含むことのある4~6員の含窒素飽和ヘテロ環を表す。含酸素飽和ヘテロ環としては、オキセタニル基、テトラヒドロピラニル基またはテトラヒドロフリル基等、該含窒素飽和ヘテロ環としては、ピロリジン、ピペリジン、ピペラジン、モルホリン等がそれぞれ例示され、これらはハロゲン、メチル基またはメトキシ基等で置換されていてもよい。
 置換基を有してもよいアルキル基の好ましい態様としては、上記置換アミノ基がメチル基に置換した-CHNR1112を例示することができる。
 置換基を有してもよい飽和ヘテロ環式基の飽和ヘテロ環式基部分としては、硫黄原子、窒素原子および酸素原子から任意に選択される1~3個のヘテロ原子を含む4~6員の飽和ヘテロ環式基のいずれでもよく、化学的に置換可能な任意の位置で置換することができて、具体的には、アゼチジニル基、ピロリジニル基、ピペリジニル基、モルホリニル基、ピペラジニル基、オキセタニル基、テトラヒドロフラニル基、テトラヒドロチエニル基、テトラヒドロピラニル基、チオモルホリニル基等が挙げられる。そして、該飽和ヘテロ環式基の置換基としては、ハロゲン原子、ヒドロキシ基、メチル基、メトキシ基、メタンスルホニル基、2-ヒドロキシエチル基、オキセタニル基、テトラヒドロピラニル基、メチル-ピペリジニル基、メチル-ピペラジニル基、モルホリニル基、チオモルホリニル基等が例示される。
 Rにおける置換基を有してもよいアミノ基は、原子団-NR1314で表され、R13、R14はそれぞれ独立して、C1-6アルキルであって、該C1-6アルキルはさらにジメチルアミノ基、メトキシ基等で置換されていてもよい。置換基を有してもよいアミノ基の好ましい態様としては、ジメチルアミノ基である。
1. R1
R 1 represents an alkyl group which may have a substituent, a saturated heterocyclic group which may have a substituent, or an amino group which may have a substituent.
The alkyl group moiety of the alkyl group which may have a substituent may be any of C1-6 alkyl, and particularly preferably C1-3 alkyl.
A specific example of a preferred substituent in the C 1-6 alkyl or C 1-3 alkyl is a substituted amino group --NR 11 R 12. Here, R 11 and R 12 each independently represent , a 4- to 6-membered oxygen-containing saturated heterocycle, or a C 1-6 alkyl, or a 4- to 6-membered ring which, taken together with the nitrogen atom to which it is bonded, may further contain a nitrogen atom or an oxygen atom. Examples of the oxygen-containing saturated heterocycle include an oxetanyl group, a tetrahydropyranyl group, and a tetrahydrofuryl group, and examples of the nitrogen-containing saturated heterocycle include pyrrolidine, piperidine, piperazine, and morpholine. These may be substituted with halogen, a methyl group, a methoxy group, or the like.
A preferred embodiment of the alkyl group which may have a substituent is -CH 2 NR 11 R 12 in which the above-mentioned substituted amino group is substituted with a methyl group.
The saturated heterocyclic group portion of the optionally substituted saturated heterocyclic group is a 4- to 6-membered heterocyclic group containing 1 to 3 heteroatoms arbitrarily selected from a sulfur atom, a nitrogen atom, and an oxygen atom. and can be substituted at any position where chemical substitution is possible. Specifically, the substituent can be an azetidinyl group, a pyrrolidinyl group, a piperidinyl group, a morpholinyl group, a piperazinyl group, an oxetanyl group, or , tetrahydrofuranyl group, tetrahydrothienyl group, tetrahydropyranyl group, thiomorpholinyl group, etc. The substituent of the saturated heterocyclic group may be a halogen atom, a hydroxy group, a methyl group, a methoxy group, a methanesulfonyl group, etc. Examples include a 2-hydroxyethyl group, an oxetanyl group, a tetrahydropyranyl group, a methyl-piperidinyl group, a methyl-piperazinyl group, a morpholinyl group, and a thiomorpholinyl group.
The amino group which may have a substituent in R 1 is represented by the atomic group -NR 13 R 14 , R 13 and R 14 each independently represent a C 1-6 alkyl, The 6- alkyl may be further substituted with a dimethylamino group, a methoxy group, etc. A preferred embodiment of the amino group which may have a substituent is a dimethylamino group.

 2.R
 Rは置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基または置換基を有してもよいアルキニル基を表す。
 置換基を有してもよいアリール基のアリール基部分としては、フェニル基、ナフチル基等が例示される。
 置換基を有してもよいヘテロアリール基のヘテロアリール基部分としては、硫黄原子、窒素原子および酸素原子から任意に選択される1~4個のヘテロ原子を含む4~6員のヘテロアリール環式基が該当し、ピロリル基、フラニル基、チエニル基、イミダゾリル基、ピラゾリル基、オキサゾリル基、チアゾリル基、イミダゾリル基、トリアゾリル基、テトラゾリル基、ピリジル基、ピラジニル基、ピリミジニル基、ピリダジニル基、トリアジニル基等が例示される。
 置換基を有してもよいアルキニル基のアルキニル基部分としては、少なくとも一つの炭素-炭素間三重結合を含み、炭素原子数が2~6の直鎖状又は分枝鎖状の不飽和炭化水素基(C2-6アルキニル基)を意味し、具体的には、エチニル基、2-プロピニル基、2-ブチニル基、2-ペンチニル基、3-ペンチニル基、2-ヘキシニル基、3-ヘキシニル基等が例示される。
 上記置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基および置換基を有してもよいアルキニル基における置換基としては、置換基を有してもよいC1-6アルキル、置換基を有してもよいC1-6アルコキシ、ハロゲン、シアノ基、水酸基、ベンジル基等を例示することができ、これらC1-6アルキルおよびC1-6アルコキシの置換基としては、1~3個のハロゲン、水酸基、シクロプロピル基、フェニル基等を例示することができる。また、隣接する置換基が環形成する例としては、メチレンジオキシ基またはエチレンジオキシ基を例示することができる。
2. R2
R2 represents an aryl group which may have a substituent, a heteroaryl group which may have a substituent, or an alkynyl group which may have a substituent.
Examples of the aryl group moiety of the aryl group which may have a substituent include a phenyl group and a naphthyl group.
The heteroaryl group portion of the heteroaryl group which may have a substituent is a 4- to 6-membered heteroaryl ring containing 1 to 4 heteroatoms arbitrarily selected from a sulfur atom, a nitrogen atom, and an oxygen atom. The formula includes a pyrrolyl group, a furanyl group, a thienyl group, an imidazolyl group, a pyrazolyl group, an oxazolyl group, a thiazolyl group, an imidazolyl group, a triazolyl group, a tetrazolyl group, a pyridyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, a triazinyl group, etc. are examples.
The alkynyl group portion of the alkynyl group which may have a substituent is a straight-chain or branched-chain unsaturated hydrocarbon having at least one carbon-carbon triple bond and 2 to 6 carbon atoms. Specifically, it means an ethynyl group, a 2-propynyl group, a 2-butynyl group, a 2-pentynyl group, a 3-pentynyl group, a 2-hexynyl group, a 3-hexynyl group. etc. are examples.
The substituents in the above-mentioned aryl group which may have a substituent, the heteroaryl group which may have a substituent, and the alkynyl group which may have a substituent are preferably C1 Examples of the substituents of the C 1-6 alkyl and C 1-6 alkoxy include halogen, cyano, hydroxyl, and benzyl. Examples of the substituents include 1 to 3 halogens, a hydroxyl group, a cyclopropyl group, a phenyl group, etc. Examples of the substituents that form a ring include a methylenedioxy group or an ethylenedioxy group. Examples can be given.

 3.Z
 Zは置換基を有してもよいアルキル基、置換基を有してもよいアルケニル基、置換基を有してもよいアルキニル基、置換基を有してもよい飽和ヘテロ環式基または置換基を有してもよいシクロアルキル基を表す。
 置換基を有してもよいアルキル基のアルキル基部分は、C1-6アルキルである。このC1-6アルキルの置換基としては、C3-6シクロアルキル基、ハロゲノC3-6シクロアルキル基、C1-3アルコキシ基、ハロゲン原子、ヒドロキシ基、ベンジルオキシ基、メチルアミノ基、ジメチルアミノ基、ピラニル基、ピペリジニル基、C1-3アルキルスルホニル基等を例示できる。
 置換基を有してもよいアルケニル基のアルケニル基部分としては、炭素数2から6の直鎖状または分枝状のアルケニル基のいずれでもよく、例えば、アリル基、2-ブテニル基、イソブテニル基、2-ペンテニル基、3-ペンテニル基、2-ヘキセニル基、等を挙げることができる。
 Zにおける置換基を有してもよいアルキニル基は、上記Rの置換基を有してもよいアルキニル基と同様である。
 置換基を有してもよいシクロアルキル基のシクロアルキル基部分としては、炭素数3から8の環状の飽和炭化水素基(C3-8シクロアルキル)のいずれでもよいが、特にシクロプロピル基が好ましい。このC3-8シクロアルキルの置換基としては、ハロゲン原子等を例示できる。
 置換基を有してもよい飽和ヘテロ環式基の飽和ヘテロ環式基部分としては、硫黄原子、窒素原子および酸素原子から任意に選択される1~3個のヘテロ原子を含む4~6員の飽和ヘテロ環式基のいずれでもよく、化学的に置換可能な任意の位置で置換することができて、具体的には、ピロリジニル基、ピペリジニル基、モルホリニル基、ピペラジニル基、オキセタニル基、テトラヒドロフラニル基、テトラヒドロチエニル基、テトラヒドロピラニル基、チオモルホリニル基等が挙げられる。
3. Z
Z represents an alkyl group which may have a substituent, an alkenyl group which may have a substituent, an alkynyl group which may have a substituent, a saturated heterocyclic group which may have a substituent, or a cycloalkyl group which may have a substituent.
The alkyl group portion of the alkyl group which may have a substituent is a C 1-6 alkyl. Examples of the substituent of this C 1-6 alkyl include a C 3-6 cycloalkyl group, a halogeno C 3-6 cycloalkyl group, a C 1-3 alkoxy group, a halogen atom, a hydroxy group, a benzyloxy group, a methylamino group, a dimethylamino group, a pyranyl group, a piperidinyl group, and a C 1-3 alkylsulfonyl group.
The alkenyl group moiety of the alkenyl group which may have a substituent may be any of linear or branched alkenyl groups having 2 to 6 carbon atoms, such as an allyl group, a 2-butenyl group, an isobutenyl group, a 2-pentenyl group, a 3-pentenyl group, and a 2-hexenyl group.
The alkynyl group which may have a substituent in Z is the same as the alkynyl group which may have a substituent in R2 .
The cycloalkyl group portion of the cycloalkyl group which may have a substituent may be any cyclic saturated hydrocarbon group having 3 to 8 carbon atoms (C cycloalkyl ), with a cyclopropyl group being particularly preferred. Examples of the substituent of this C cycloalkyl include halogen atoms.
The saturated heterocyclic group portion of the optionally substituted saturated heterocyclic group may be any 4- to 6-membered saturated heterocyclic group containing 1 to 3 heteroatoms arbitrarily selected from sulfur atoms, nitrogen atoms, and oxygen atoms, and may be substituted at any chemically substitutable position. Specific examples thereof include a pyrrolidinyl group, a piperidinyl group, a morpholinyl group, a piperazinyl group, an oxetanyl group, a tetrahydrofuranyl group, a tetrahydrothienyl group, a tetrahydropyranyl group, a thiomorpholinyl group, and the like.

 本発明の化合物(I)のある態様は、Zが置換基を有してもよいシクロアルキル基である、前記チアゾール誘導体(I)またはその薬学的に許容される塩である。
 本発明の化合物(I)のある態様は、Zが置換基を有してもよいアルキル基である、前記チアゾール誘導体(I)またはその薬学的に許容される塩である。
 本発明の化合物(I)のまたひとつの態様は、Rが置換基を有してもよいフェニル基である、前記チアゾール誘導体(I)またはその薬学的に許容される塩である。
 本発明の化合物(I)のまたひとつの態様は、Rが置換基を有してもよいアルキニル基である、前記チアゾール誘導体(I)またはその薬学的に許容される塩である。
 本発明の化合物(I)のまたひとつの態様は、実施例1から226よりなる群から選択される、前記チアゾール誘導体(I)またはその薬学的に許容される塩である。
An embodiment of the compound (I) of the present invention is the above thiazole derivative (I) or a pharma- ceutically acceptable salt thereof, wherein Z is a cycloalkyl group which may be substituted.
An embodiment of compound (I) of the present invention is the above thiazole derivative (I) or a pharma- ceutically acceptable salt thereof, wherein Z is an alkyl group which may have a substituent.
Another embodiment of the compound (I) of the present invention is the above thiazole derivative (I) or a pharma- ceutically acceptable salt thereof, wherein R2 is a phenyl group which may be substituted.
Another embodiment of the compound (I) of the present invention is the above thiazole derivative (I) or a pharma- ceutically acceptable salt thereof, wherein R2 is an alkynyl group which may be substituted.
Another embodiment of the compound (I) of the present invention is the thiazole derivative (I) or a pharma- ceutically acceptable salt thereof, which is selected from the group consisting of Examples 1 to 226.

 本発明の化合物(I)は、例えば、置換基の種類によって、異性体が存在する場合がある。本明細書において、それらの異性体の一形態のみの化学構造で記載することがあるが、本発明には、構造上生じ得るすべての異性体(幾何異性体、光学異性体、互変異性体など)も含有し、異性体単体、またはそれらの混合物も含有する。
 また、本発明の化合物(I)の薬学的に許容される塩としては、塩酸、硫酸、炭酸、リン酸等との無機酸塩、ギ酸、フマル酸、マレイン酸、メタンスルホン酸、p-トルエンスルホン酸等との有機酸塩等が挙げられる。また、ナトリウム、カリウム等とのアルカリ金属塩、マグネシウム、カルシウム等とのアルカリ土類金属塩、低級アルキルアミン、低級アルコールアミン等との有機アミン塩、リジン、アルギニン、オルニチン等との塩基性アミノ酸塩の他、アンモニウム塩等も本発明に包含される。
 本発明の化合物(I)およびその薬学的に許容される塩は、例えば以下の方法によって製造することができる。なお、以下に示した製造法において、定義した基が実施方法の条件下で変化するか、または当該方法を実施するのに不向きな場合、有機合成化学で通常用いられる方法、例えば、官能基の保護、脱保護[T.W.Greene,Protective Groups in Organic Synthesis 4th Edition, John Wiley&Sons,Inc.,2007]等の手段を付すことにより容易に製造することができる。また、必要に応じて置換基導入等の反応工程の順序を変えることもできる。
Compound (I) of the present invention may have isomers depending on, for example, the type of substituent. In this specification, the chemical structure of only one form of the isomer may be described, but the present invention also includes all isomers (geometric isomers, optical isomers, tautomers, etc.) that may occur due to the structure, and also includes a single isomer or a mixture thereof.
Pharmaceutically acceptable salts of compound (I) of the present invention include inorganic acid salts with hydrochloric acid, sulfuric acid, carbonic acid, phosphoric acid, etc., and organic acid salts with formic acid, fumaric acid, maleic acid, methanesulfonic acid, p-toluenesulfonic acid, etc. In addition, the present invention also includes alkali metal salts with sodium, potassium, etc., alkaline earth metal salts with magnesium, calcium, etc., organic amine salts with lower alkylamines, lower alcoholamines, etc., basic amino acid salts with lysine, arginine, ornithine, etc., as well as ammonium salts.
Compound (I) of the present invention and its pharma- ceutical acceptable salts can be prepared, for example, by the following method. In the preparation methods shown below, if the defined groups change under the conditions of the method or are not suitable for carrying out the method, the compound can be easily prepared by applying a method commonly used in organic synthesis chemistry, such as protection and deprotection of functional groups [T. W. Greene, Protective Groups in Organic Synthesis 4th Edition, John Wiley & Sons, Inc., 2007]. In addition, the order of reaction steps such as introduction of substituents can be changed as necessary.

以下の説明で使用される略語、記号の意味は次の通りである。
DMF:ジメチルホルムアミド
DMSO:ジメチルスルホキシド
HATU:O-(7-アザベンゾトリアゾール-1-イル)-N,N,N’,N’-テトラメチルウロニウム ヘキサフルオロホスファート
DIPEA:N,N-ジイソプロピルエチルアミン
THF:テトラヒドロフラン
The abbreviations and symbols used in the following description have the following meanings:
DMF: Dimethylformamide DMSO: Dimethylsulfoxide HATU: O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate DIPEA: N,N-diisopropylethylamine THF: Tetrahydrofuran

[本発明の化合物(I)の製法]
 以下に本発明化合物(I)の製造方法を説明する。
 式(I)で表される本発明の化合物は、例えばスキーム1によって製造することができる。

Figure JPOXMLDOC01-appb-C000004
(式中、R、R及びZは前記と同義であり、Xはハロゲンを表す。) [Method for producing compound (I) of the present invention]
The process for producing the compound (I) of the present invention will be explained below.
The compound of the present invention represented by formula (I) can be produced, for example, according to Scheme 1.
Figure JPOXMLDOC01-appb-C000004
(In the formula, R 1 , R 2 and Z are defined as above, and X represents a halogen.)

 本発明の化合物(I)は、化合物(II)と化合物R-XとのUllmann縮合反応あるいは求核置換反応により製造することができる。
すなわち、Rが置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基の場合、化合物(II)と化合物R-Xの混合物を溶媒中、銅などの金属触媒の存在下、必要に応じて塩基および添加剤を使用してUllmann縮合反応させることによって、化合物(I)を製造することができる。反応に用いる溶媒としては反応に不活性なものであればいずれでも良く、特に限定されるものではないが、好ましくは、ジオキサン、トルエン、ジメトキシエタン、DMFなどが挙げられる。化合物R-Xは、化合物(II)に対しモル当量または過剰量用いることが好ましく、より好ましくは1モル当量から10モル当量である。必要に応じて反応を加速させるために塩基を添加してもよく、塩基としては炭酸ナトリウム、炭酸セシウム、炭酸カリウムなどが通常用いられる。使用する塩基の用量は化合物(II)に対し1モル当量から10モル当量が挙げられ、好ましくは1モル当量から5モル当量である。金属触媒としては、カップリングに用いられている市販で入手容易な銅触媒(例えば、CuIなど)を用いることができ、化合物(II)に対して触媒量を添加することが好ましい。反応は30~150℃において、1~48時間加熱することで実施することができるが、好ましくは80~130℃において12~24時間反応させることにより実施することができる。化合物(I)は、銅触媒の代わりにパラジウム触媒を用いたBuchwald-Hartwig反応条件でも製造することができる。
 また、化合物R-XのハロゲンXをボロン酸もしくはそのボロン酸エステルに変換した化合物を用いたChan-Lam-Evansカップリング反応によっても化合物(I)を製造することができる。
 Rが置換基を有してもよいアルキニル基である場合、本発明の化合物(I)は、化合物(II)と1~5モル当量、好ましくは1~2.5モル当量の化合物R-X、および1~5モル当量、好ましくは1~3.5モル当量の炭酸セシウムのような塩基存在下、溶媒中、求核置換反応させることによって得ることができる。
 溶媒は反応に不活性なものであればいずれでもよく、特に限定されるものではないが、好ましくはDMSO、もしくはDMFを用いることができる。反応は30~150℃において、1~24時間加熱することで実施することができるが、好ましくは50~80℃において1~7時間反応させることにより実施することができる。
The compound (I) of the present invention can be produced by Ullmann condensation reaction or nucleophilic substitution reaction between the compound (II) and a compound R 2 -X.
That is, when R 2 is an aryl group which may have a substituent or a heteroaryl group which may have a substituent, a mixture of compound (II) and compound R 2 -X is subjected to Ullmann condensation reaction in a solvent in the presence of a metal catalyst such as copper, using a base and an additive as necessary, to produce compound (I). The solvent used in the reaction may be any solvent inert to the reaction, and is not particularly limited, but preferably includes dioxane, toluene, dimethoxyethane, DMF, and the like. Compound R 2 -X is preferably used in a molar equivalent or excess amount relative to compound (II), more preferably 1 to 10 molar equivalents. A base may be added to accelerate the reaction as necessary, and sodium carbonate, cesium carbonate, potassium carbonate, and the like are usually used as the base. The amount of the base used is 1 to 10 molar equivalents relative to compound (II), preferably 1 to 5 molar equivalents. As the metal catalyst, a commercially available copper catalyst (e.g., CuI, etc.) used in coupling can be used, and it is preferable to add a catalytic amount to compound (II). The reaction can be carried out by heating at 30 to 150° C. for 1 to 48 hours, preferably at 80 to 130° C. for 12 to 24 hours. Compound (I) can also be produced under Buchwald-Hartwig reaction conditions using a palladium catalyst instead of a copper catalyst.
Compound (I) can also be produced by a Chan-Lam-Evans coupling reaction using a compound in which the halogen X of compound R 2 -X is converted to a boronic acid or a boronic acid ester thereof.
When R 2 is an alkynyl group which may have a substituent, compound (I) of the present invention can be obtained by subjecting compound (II) to a nucleophilic substitution reaction in the presence of 1 to 5 molar equivalents, preferably 1 to 2.5 molar equivalents of compound R 2 -X and 1 to 5 molar equivalents, preferably 1 to 3.5 molar equivalents of a base such as cesium carbonate, in a solvent.
The solvent may be any solvent inactive to the reaction, and is not particularly limited, but preferably DMSO or DMF can be used. The reaction can be carried out by heating at 30 to 150° C. for 1 to 24 hours, and preferably at 50 to 80° C. for 1 to 7 hours.

 また本発明の化合物(I)は、化合物(III)を用いて、例えばスキーム2に示す方法によっても製造することができる。

Figure JPOXMLDOC01-appb-C000005
(式中、R、R及びZは前記と同義である。) Compound (I) of the present invention can also be produced by the method shown in Scheme 2, for example, using compound (III).
Figure JPOXMLDOC01-appb-C000005
(In the formula, R 1 , R 2 and Z are as defined above.)

 本発明の化合物(I)は、化合物(III)およびボロン酸(IV)を用い、Chan-Lam-Evansカップリング反応により製造することができる。すなわち、化合物(III)およびボロン酸(IV)の混合物を溶媒中、銅やニッケルなどの金属触媒の存在下、必要に応じて塩基および添加剤を使用して反応させることによって、化合物(I)を製造することができる。反応に用いる溶媒としては反応に不活性なものであればいずれでも良く、特に限定されるものではないが、好ましくはジクロロメタン、もしくはジクロロエタンを用いることができる。
反応は30~150℃において、1~24時間加熱することで実施することができるが、好ましくは50~80℃において1~7時間反応させることにより実施することができる。
 また、化合物(IV)のボロニル基は、ボロン酸エステル基に置き換えて反応に使用することもできる。
 また、化合物(III)を、化合物(IV)のボロン酸部分が適当な脱離基であるZ導入剤等と、適当な塩基存在下、求核置換反応を行うことによっても化合物(I)を製造することができる。
Compound (I) of the present invention can be produced by Chan-Lam-Evans coupling reaction using compound (III) and boronic acid (IV). That is, compound (I) can be produced by reacting a mixture of compound (III) and boronic acid (IV) in a solvent in the presence of a metal catalyst such as copper or nickel, using a base and an additive as necessary. The solvent used in the reaction may be any solvent inert to the reaction, and is not particularly limited, but preferably dichloromethane or dichloroethane can be used.
The reaction can be carried out by heating at 30 to 150° C. for 1 to 24 hours, preferably at 50 to 80° C. for 1 to 7 hours.
Furthermore, the boronyl group of compound (IV) can be replaced with a boronic acid ester group for use in the reaction.
Compound (I) can also be produced by subjecting compound (III) to a nucleophilic substitution reaction with a Z-introducing agent in which the boronic acid moiety of compound (IV) is a suitable leaving group, in the presence of a suitable base.

 また本発明の化合物(I)は、例えばスキーム3によっても製造することができる。

Figure JPOXMLDOC01-appb-C000006
(式中、R、R及びZは前記と同義であり、Rは低級アルキル基を表わす。)
 本発明の化合物(I)は、化合物(V)のエステル基をアミノリシス反応によりカルボキサミド基に変換することで製造することができる。すなわち、化合物(V)を溶媒中、アンモニアと加熱反応させることによって、化合物(I)を製造することができる。溶媒は反応に不活性なものであればいずれでも良く、特に限定されるものではないが、好ましくはエタノール、もしくはメタノールを用いることができ、アンモニアとしては、水溶液、アルコール溶液などを用いるか、直接、反応液にアンモニアガスを吹き込むことができる。
 反応は50~150℃において、1~48時間加熱することで実施することができるが、好ましくは80~100℃において1~15時間反応させることにより実施することができる。
 当該カルボキサミド基への変換は、通常の有機合成で用いられる手法、すなわち、エステルの加水分解後、カップリング剤を用いたアミド化反応でも実施可能である。 Moreover, the compound (I) of the present invention can also be produced, for example, according to Scheme 3.
Figure JPOXMLDOC01-appb-C000006
(In the formula, R 1 , R 2 and Z are defined as above, and R 3 represents a lower alkyl group.)
The compound (I) of the present invention can be produced by converting the ester group of the compound (V) to a carboxamide group by aminolysis reaction. That is, the compound (I) can be produced by reacting the compound (V) with ammonia in a solvent under heating. The solvent may be any solvent inert to the reaction, and is not particularly limited, but preferably ethanol or methanol can be used. As the ammonia, an aqueous solution, an alcohol solution, etc. can be used, or ammonia gas can be directly blown into the reaction solution.
The reaction can be carried out by heating at 50 to 150° C. for 1 to 48 hours, preferably at 80 to 100° C. for 1 to 15 hours.
The conversion to the carboxamide group can also be carried out by a method commonly used in organic synthesis, namely, hydrolysis of an ester followed by an amidation reaction using a coupling agent.

 また本発明の化合物(I)は、例えばスキーム4によっても製造することができる。

Figure JPOXMLDOC01-appb-C000007
(式中、R、R及びZは前記と同義であり、Xはハロゲンを表す。)
 本発明の化合物(I)は、化合物(VI)と化合物(VII)を用いて鈴木カップリング反応などのクロスカップリング反応により製造することができる(例えば、鈴木カップリング反応の条件については公知文献(N.Miyaura,et al,J.Am.Chem.Soc.,107,972(1985).,N.Miyaura,A.Suzuki,Chem.Rev.95,2457(1995))などを参照)。すなわち、化合物(VI)と化合物(VII)の混合物を溶媒中、パラジウムやニッケルなどの金属触媒の存在下、必要に応じて塩基および添加剤を使用して反応させることによって、化合物(I)を製造することができる。
 反応に用いる溶媒としてはTHF、ジオキサン、トルエン、ジメトキシエタン、メタノール、エタノール、アセトニトリルなどが挙げられる。また、これらの溶媒を2種以上混合して、或いはそれらを更に水と混合して用いても好適である。好ましくは、ジオキサンと水の混合溶媒、トルエンとメタノールおよび水との混合溶媒である。化合物(VII)は化合物(VI)に対しモル当量または過剰量用いることが好ましく、より好ましくは1モル当量から10モル当量である。必要に応じて反応を加速させるために塩基を添加してもよく、塩基としては炭酸ナトリウム、炭酸セシウム、炭酸カリウム、フッ化セシウムなどが通常用いられる。使用する塩基の用量は化合物(VI)に対し1モル当量から10モル当量が挙げられ、好ましくは1モル当量から5モル当量である。金属触媒としては、クロスカップリングに用いられている市販で入手容易なパラジウム触媒(例えば、PdCl(Amphos)、PdCl(dppf)、Pd(dba)、Pd(PPhなど)を用いることができ、化合物(VI)に対して触媒量、すなわち0.1モル当量から0.5モル当量を添加することが好ましい。また、化合物(VII)のボロニル基は、ボロン酸エステル基に置き換えて反応に使用することもできる。
 スキーム4の原料の一つである化合物(VII)は市販品として、または公知の方法もしくはそれに準じた方法により得ることができる。 Moreover, the compound (I) of the present invention can also be produced, for example, according to Scheme 4.
Figure JPOXMLDOC01-appb-C000007
(In the formula, R 1 , R 2 and Z are defined as above, and X represents a halogen.)
Compound (I) of the present invention can be produced by a cross-coupling reaction such as Suzuki coupling reaction using compound (VI) and compound (VII) (for example, see known literature (N. Miyaura, et al., J. Am. Chem. Soc., 107, 972 (1985)., N. Miyaura, A. Suzuki, Chem. Rev. 95, 2457 (1995)) for the conditions of Suzuki coupling reaction). That is, compound (I) can be produced by reacting a mixture of compound (VI) and compound (VII) in a solvent in the presence of a metal catalyst such as palladium or nickel, and using a base and an additive as necessary.
Examples of the solvent used in the reaction include THF, dioxane, toluene, dimethoxyethane, methanol, ethanol, and acetonitrile. It is also suitable to use two or more of these solvents in combination, or to further mix them with water. A mixed solvent of dioxane and water, or a mixed solvent of toluene, methanol, and water is preferred. Compound (VII) is preferably used in a molar equivalent or excess amount relative to compound (VI), more preferably 1 to 10 molar equivalents. If necessary, a base may be added to accelerate the reaction, and sodium carbonate, cesium carbonate, potassium carbonate, cesium fluoride, and the like are usually used as the base. The amount of the base used is 1 to 10 molar equivalents relative to compound (VI), preferably 1 to 5 molar equivalents. As the metal catalyst, a commercially available palladium catalyst (e.g., PdCl2 (Amphos), PdCl2 (dppf), Pd2 (dba) 3 , Pd( PPh3 ) 4 , etc.) used in cross-coupling can be used, and it is preferable to add a catalytic amount, i.e., 0.1 to 0.5 molar equivalents, to compound (VI). In addition, the boronyl group of compound (VII) can be replaced with a boronic acid ester group for use in the reaction.
Compound (VII), which is one of the raw materials in Scheme 4, is a commercially available product or can be obtained by a known method or a method analogous thereto.

 また式(I)において、Rが置換基を有してもよいアミノ基で表される本発明の化合物(I-a)は、例えばスキーム5によっても製造することができる。

Figure JPOXMLDOC01-appb-C000008
(式中、R、R13、R14及びZは前記と同義である。)
 化合物(I-a)は、化合物(VIII)のカルボキシル基とアミン(IX)を縮合反応させることで製造できる。すなわち、化合物(VIII)と0.9~5モル当量、好ましくは1~2モル当量のアミン(IX)を溶媒中、有機化学で一般的に用いられるアミド縮合剤存在下、反応させることによって得ることができる。溶媒は反応に不活性なものであればいずれでも良く、特に限定されるものではないが、例えば、DMF、THFなどを用いることができる。反応は0~150℃において、1~48時間撹拌することで実施することができるが、好ましくは室温~80℃において1時間から終夜反応させることにより実施することができる。
 スキーム5の原料の一つであるアミン(IX)は市販品として、または公知の方法もしくはそれに準じた方法により得ることができる。 Moreover, the compound (Ia) of the present invention, in which R 1 in formula (I) is an amino group which may have a substituent, can also be produced according to scheme 5, for example.
Figure JPOXMLDOC01-appb-C000008
(In the formula, R 2 , R 13 , R 14 and Z are the same as defined above.)
Compound (I-a) can be produced by condensation reaction of the carboxyl group of compound (VIII) with amine (IX). That is, compound (VIII) can be obtained by reacting 0.9 to 5 molar equivalents, preferably 1 to 2 molar equivalents, of amine (IX) in a solvent in the presence of an amide condensing agent commonly used in organic chemistry. The solvent may be any solvent inert to the reaction, and is not particularly limited, but for example, DMF, THF, etc. can be used. The reaction can be carried out by stirring at 0 to 150°C for 1 to 48 hours, but preferably by reacting at room temperature to 80°C for 1 hour to overnight.
The amine (IX), which is one of the raw materials in Scheme 5, is a commercially available product or can be obtained by a known method or a method similar thereto.

 また、式(I)において、Rが置換基を有してもよいアルキル基で表される化合物のうち、置換基が「置換基を有してもよいアミノ基」をもつ本発明の化合物(I-b)は、Rが置換基を有してもよいアルキル基で表される化合物のうち、ハロゲンを置換基にもつ本発明の化合物(I-c)を用いて、例えばスキーム6によっても製造することができる。

Figure JPOXMLDOC01-appb-C000009
(式中、R、Z、R11およびR12は前記と同義であり、Xはハロゲンを表わす。)
 化合物(I-b)は、化合物(I-c)とアミン(IX)を反応させることで製造することができる。すなわち、化合物(I-c)と0.9~5モル当量、好ましくは1~2モル当量のアミン(IX)を溶媒中、塩基存在下、反応させることによって得ることができる。溶媒は反応に不活性なものであればいずれでも良く、特に限定されるものではないが、例えば、DMF、THFなどを用いることができる。
 反応は0~150℃において、1~48時間撹拌することで実施することができるが、好ましくは室温~80℃において1時間から終夜反応させることにより実施することができる。 Furthermore, among the compounds in formula (I) in which R 1 is represented by an alkyl group which may have a substituent, compound (I-b) of the present invention having an "amino group which may have a substituent" as a substituent can also be produced using compound (I-c) of the present invention in which R 1 is represented by an alkyl group which may have a substituent and has a halogen as a substituent, for example, according to Scheme 6.
Figure JPOXMLDOC01-appb-C000009
(In the formula, R 2 , Z, R 11 and R 12 are the same as defined above, and X represents a halogen.)
Compound (I-b) can be produced by reacting compound (I-c) with amine (IX). That is, compound (I-c) can be reacted with 0.9 to 5 molar equivalents, preferably 1 to 2 molar equivalents, of amine (IX) in a solvent in the presence of a base. The solvent may be any solvent inert to the reaction, and is not particularly limited. For example, DMF, THF, etc. can be used.
The reaction can be carried out at 0 to 150° C. with stirring for 1 to 48 hours, preferably at room temperature to 80° C. for 1 hour to overnight.

 スキーム1の原料として用いられる化合物(II)は、例えばスキーム7に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000010
(式中、R及びZは前記と同義であり、Xはハロゲンを表し、PGは保護基を表わす。)
 化合物(II)は、化合物(X)と化合物(VII)を用いて鈴木カップリング反応などのクロスカップリング反応により得た生成物を脱保護することに製造することができる。すなわち、化合物(X)と化合物(VII)をスキーム4と同様の条件で反応させることにより、化合物(II)の保護体を得ることができる。得られた化合物(II)の保護体の脱保護反応は、用いた保護基に適した有機化学で一般的に用いられる脱保護条件に供することで実施できる。化合物(VII)のボロニル基は、ボロン酸エステル基に置き換えて反応に使用することもできる。 Compound (II) used as the starting material in Scheme 1 can be produced, for example, by the method shown in Scheme 7.
Figure JPOXMLDOC01-appb-C000010
(In the formula, R1 and Z are defined as above, X represents a halogen, and PG represents a protecting group.)
Compound (II) can be produced by deprotecting the product obtained by a cross-coupling reaction such as Suzuki coupling reaction using compound (X) and compound (VII). That is, a protected form of compound (II) can be obtained by reacting compound (X) and compound (VII) under the same conditions as in Scheme 4. The deprotection reaction of the obtained protected form of compound (II) can be carried out by subjecting it to deprotection conditions generally used in organic chemistry that are suitable for the protecting group used. The boronyl group of compound (VII) can also be replaced with a boronic acid ester group and used in the reaction.

スキーム7の原料として用いられる化合物(X)は、例えばスキーム8に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000011
(式中、Zは前記と同義であり、Xはハロゲンを表し、Rは低級アルキル基を表わし、PGは保護基を表わす。)
 化合物(X)は、化合物(XI)のエステル基をアミノリシス反応によりカルボキサミド基に変換することで製造することができる。反応は、スキーム3と同様の条件を用いることで実施できる。また当該カルボキサミド基への変換は、通常の有機合成で用いられる手法、すなわち、エステルの加水分解後、カップリング剤を用いたアミド化反応でも実施可能である。 Compound (X) used as the starting material in Scheme 7 can be produced, for example, by the method shown in Scheme 8.
Figure JPOXMLDOC01-appb-C000011
(In the formula, Z is defined as above, X represents a halogen, R3 represents a lower alkyl group, and PG represents a protecting group.)
Compound (X) can be produced by converting the ester group of compound (XI) to a carboxamide group by aminolysis reaction. The reaction can be carried out under the same conditions as in Scheme 3. The conversion to the carboxamide group can also be carried out by a method used in ordinary organic synthesis, that is, by hydrolysis of the ester followed by an amidation reaction using a coupling agent.

スキーム8の原料として用いられる化合物(XI)は、例えばスキーム9に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000012
(式中、Zは前記と同義であり、Xはハロゲンを表し、Rは低級アルキル基を表わし、PGは保護基を表わす。)
 化合物(XI)は、化合物(XII)を通常の有機化学で用いられるハロゲン化剤、例えばN-ブロモコハク酸イミド(NBS)やN-クロロコハク酸イミド(NCS)などで処理することで製造することができる。すなわち、化合物(XI)は、溶媒中、化合物(XII)と、0.5~2モル当量、好ましくは0.8~1.1モル当量のNBSあるいはNCSを反応させることによって得ることができる。
 溶媒は反応に不活性なものであればいずれでもよく、特に限定されるものではないが、好ましくはジクロロメタン、もしくはDMFを用いることができる。反応は-20~30℃において、0.25~10時間撹拌することで実施することができるが、好ましくは-10~10℃において0.5~5時間撹拌することで実施することができる。 Compound (XI) used as the starting material in Scheme 8 can be produced, for example, by the method shown in Scheme 9.
Figure JPOXMLDOC01-appb-C000012
(In the formula, Z is as defined above, X represents a halogen, R3 represents a lower alkyl group, and PG represents a protecting group.)
Compound (XI) can be produced by treating compound (XII) with a halogenating agent typically used in organic chemistry, such as N-bromosuccinimide (NBS) or N-chlorosuccinimide (NCS), etc. That is, compound (XI) can be obtained by reacting compound (XII) with 0.5 to 2 molar equivalents, preferably 0.8 to 1.1 molar equivalents, of NBS or NCS in a solvent.
The solvent may be any solvent inert to the reaction, and is not particularly limited, but preferably dichloromethane or DMF can be used. The reaction can be carried out by stirring at -20 to 30°C for 0.25 to 10 hours, preferably at -10 to 10°C for 0.5 to 5 hours.

 スキーム9の原料として用いられる化合物(XII)は、例えばスキーム10に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000013
(式中、Zは前記と同義であり、Rは低級アルキル基を表わし、PGは保護基を表わす。)
 化合物(XII)は、化合物(XIII)とボロン酸(IV)を用いて、Chan-Lam-Evansカップリング反応により製造することができる。反応条件は、スキーム2と同様の条件で実施可能である。
また、化合物(IV)のボロニル基は、ボロン酸エステル基に置き換えて反応に使用することもできる。
 また、化合物(XIII)を、化合物(IV)のボロン酸部分が適当な脱離基であるZ導入剤等と、適当な塩基存在下、求核置換反応を行うことによっても化合物(XII)を製造することができる。 Compound (XII) used as the starting material in Scheme 9 can be produced, for example, by the method shown in Scheme 10.
Figure JPOXMLDOC01-appb-C000013
(In the formula, Z is defined as above, R3 represents a lower alkyl group, and PG represents a protecting group.)
Compound (XII) can be produced by Chan-Lam-Evans coupling reaction using compound (XIII) and boronic acid (IV). The reaction conditions can be the same as those in Scheme 2.
Furthermore, the boronyl group of compound (IV) can be replaced with a boronic acid ester group for use in the reaction.
Compound (XII) can also be produced by subjecting compound (XIII) to a nucleophilic substitution reaction with a Z-introducing agent in which the boronic acid moiety of compound (IV) is a suitable leaving group, in the presence of a suitable base.

 スキーム10の原料として用いられる化合物(XIII)は、例えばスキーム11に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000014
(式中、Rは低級アルキル基を表わし、PGは保護基を表わす。)
 化合物(XIII)は、アミノピラゾール(XIV)をチオホスゲンと反応させたのち、アンモニア水溶液で処理することで得られるチオアミド(XV)を、ブロモピルビン酸エチルと反応させてチアゾール環を形成させることで製造することができる。すなわち、ジクロロメタンなどの溶媒中、アミノピラゾール(XIV)を、必要に応じて塩基存在下、0.5~2モル当量、好ましくは0.9~1.5モル当量のチオホスゲンと反応させたのち、アンモニア水溶液を加えることでチオアミド(XV)を得ることができる。このチオアミド(XV)を、エタノールなどの溶媒中、炭酸カリウムなどの塩基存在下、0.5~2モル当量、好ましくは0.8~1.5モル当量のブロモピルビン酸エチルと反応させることで、化合物(XIII)を得ることができる。
 スキーム11の原料として用いられるアミノピラゾール(XIV)は、市販品として入手するか、または4-ニトロピラゾールなどを出発原料として有機合成化学で通常用いられる方法を用いて容易に製造することができる。 Compound (XIII) used as the starting material in Scheme 10 can be produced, for example, by the method shown in Scheme 11.
Figure JPOXMLDOC01-appb-C000014
(In the formula, R3 represents a lower alkyl group, and PG represents a protecting group.)
Compound (XIII) can be produced by reacting aminopyrazole (XIV) with thiophosgene, treating with aqueous ammonia to obtain thioamide (XV), and reacting the thioamide (XV) with ethyl bromopyruvate to form a thiazole ring. That is, in a solvent such as dichloromethane, aminopyrazole (XIV) is reacted with 0.5 to 2 molar equivalents, preferably 0.9 to 1.5 molar equivalents of thiophosgene, if necessary, in the presence of a base, and then an aqueous ammonia solution is added to obtain thioamide (XV). Compound (XIII) can be obtained by reacting this thioamide (XV) with 0.5 to 2 molar equivalents, preferably 0.8 to 1.5 molar equivalents of ethyl bromopyruvate in a solvent such as ethanol in the presence of a base such as potassium carbonate.
The aminopyrazole (XIV) used as a starting material in Scheme 11 is either commercially available or can be easily prepared using a method commonly used in organic synthesis using 4-nitropyrazole or the like as a starting material.

 スキーム2の原料として用いられる化合物(III)は、例えばスキーム12に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000015
(式中、R、Rは前記と同義であり、Xはハロゲンを表わす。)
 化合物(III)は、化合物(XVI)と化合物(VII)を用いて鈴木カップリング反応などのクロスカップリング反応により製造することができる。当該クロスカップリング反応は、スキーム4と同様の条件で実施することができる。また化合物(VII)のボロニル基は、ボロン酸エステル基に置き換えて反応に使用することもできる。 Compound (III) used as a starting material in Scheme 2 can be produced, for example, by the method shown in Scheme 12.
Figure JPOXMLDOC01-appb-C000015
(In the formula, R 1 and R 2 are defined as above, and X represents a halogen.)
Compound (III) can be produced by a cross-coupling reaction such as Suzuki coupling reaction using compound (XVI) and compound (VII). The cross-coupling reaction can be carried out under the same conditions as in Scheme 4. The boronyl group of compound (VII) can also be replaced with a boronic acid ester group for use in the reaction.

 スキーム12の原料として用いられる化合物(XVI)は、例えばスキーム13に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000016
(式中、R2は前記と同義であり、Rは低級アルキル基を表わし、Xはハロゲンを表わす。)
 化合物(XVI)は、化合物(XVII)のエステル基をアミノリシス反応によりカルボキサミド基に変換することで製造することができる。反応は、スキーム3と同様の条件を用いることで実施できる。また当該カルボキサミド基への変換は、通常の有機合成で用いられる手法、すなわち、エステルの加水分解後、カップリング剤を用いたアミド化反応でも実施可能である。 Compound (XVI) used as the starting material in Scheme 12 can be produced, for example, by the method shown in Scheme 13.
Figure JPOXMLDOC01-appb-C000016
(wherein R2 is as defined above, R3 represents a lower alkyl group, and X represents a halogen.)
Compound (XVI) can be produced by converting the ester group of compound (XVII) to a carboxamide group by aminolysis reaction. The reaction can be carried out under the same conditions as in Scheme 3. The conversion to the carboxamide group can also be carried out by a method used in ordinary organic synthesis, that is, hydrolysis of the ester followed by an amidation reaction using a coupling agent.

 スキーム13の原料として用いられる化合物(XVII)は、例えばスキーム14に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000017
(式中、R2は前記と同義であり、Rは低級アルキル基を表わし、Xはハロゲンを表わす。)
 化合物(XVII)は、化合物(XVIII)を通常の有機化学で用いられるハロゲン化剤で処理することで製造することができる。反応条件は、スキーム9と同様の条件で実施可能である。
 スキーム14の原料として用いられる化合物(XVIII)は、例えばスキーム10において、化合物(XIII)の保護基の代わりにR基を有するアミノピラゾール誘導体を出発原料に用いることで製造することができる。 Compound (XVII) used as the starting material in Scheme 13 can be produced, for example, by the method shown in Scheme 14.
Figure JPOXMLDOC01-appb-C000017
(wherein R2 is as defined above, R3 represents a lower alkyl group, and X represents a halogen.)
Compound (XVII) can be produced by treating compound (XVIII) with a halogenating agent commonly used in organic chemistry. The reaction conditions can be the same as those in Scheme 9.
Compound (XVIII) used as a starting material in Scheme 14 can be produced, for example, in Scheme 10, by using an aminopyrazole derivative having an R 2 group instead of the protecting group of compound (XIII) as a starting material.

 スキーム3の原料として用いられる化合物(V)は、例えばスキーム15に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000018
(式中、R、R及びZは前記と同義であり、Rは低級アルキル基を表わし、PGは保護基を表わし、Xはハロゲンを表わす。)
 Rが置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基の場合、化合物(V)は、化合物(XIX)の保護基を脱保護した後、化合物R-XとのUllmann縮合反応あるいはBuchwald-Hartwig反応などのカップリング反応により製造することができる。反応条件は、スキーム1と同様の条件で実施可能である。
 また、化合物R-XのハロゲンXをボロン酸もしくはそのボロン酸エステルに変換した化合物を用いたChan-Lam-Evansカップリング反応によっても化合物(V)を製造することができる。
 Rが置換基を有してもよいアルキニル基である場合、化合物(V)は、化合物(XIX)の保護基を脱保護した後、化合物R-Xとの求核置換反応により製造することができる。反応条件は、スキーム1と同様の条件で実施可能である。 Compound (V) used as a starting material in Scheme 3 can be produced, for example, by the method shown in Scheme 15.
Figure JPOXMLDOC01-appb-C000018
(In the formula, R 1 , R 2 and Z are defined as above, R 3 represents a lower alkyl group, PG represents a protecting group, and X represents a halogen.)
When R 2 is an optionally substituted aryl group or an optionally substituted heteroaryl group, compound (V) can be produced by deprotecting the protecting group of compound (XIX) and then subjecting the resulting compound to a coupling reaction such as Ullmann condensation reaction or Buchwald-Hartwig reaction with compound R 2 -X. The reaction conditions can be the same as those in Scheme 1.
Compound (V) can also be produced by a Chan-Lam-Evans coupling reaction using a compound in which the halogen X of compound R 2 -X is converted to a boronic acid or a boronic acid ester thereof.
When R 2 is an alkynyl group which may have a substituent, compound (V) can be produced by deprotecting the protecting group of compound (XIX) and then subjecting it to a nucleophilic substitution reaction with compound R 2 -X. The reaction can be carried out under the same reaction conditions as in Scheme 1.

 スキーム15の原料として用いられる化合物(XIX)は、例えばスキーム16に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000019
(式中、R及びZは前記と同義であり、Rは低級アルキル基を表わし、PGは保護基を表わし、Xはハロゲンを表わす。)
 化合物(XIX)は、化合物(XI)と化合物(VII)を用いて、鈴木カップリング反応などのクロスカップリング反応により製造することができる。当該クロスカップリング反応は、スキーム4と同様の条件で実施することができる。また化合物(VII)のボロニル基は、ボロン酸エステル基に置き換えて反応に使用することもできる。 Compound (XIX) used as the starting material in Scheme 15 can be produced, for example, by the method shown in Scheme 16.
Figure JPOXMLDOC01-appb-C000019
(In the formula, R1 and Z are defined as above, R3 represents a lower alkyl group, PG represents a protecting group, and X represents a halogen.)
Compound (XIX) can be produced by a cross-coupling reaction such as Suzuki coupling reaction using compound (XI) and compound (VII). The cross-coupling reaction can be carried out under the same conditions as in Scheme 4. The boronyl group of compound (VII) can also be replaced with a boronic acid ester group for use in the reaction.

 スキーム4の原料として用いられる化合物(VI)は、例えばスキーム17に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000020
(式中、R及びZは前記と同義であり、PGは保護基を表わし、Xはハロゲンを表わす。)
 Rが置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基の場合、化合物(VI)は、化合物(X)の保護基を脱保護したのち、化合物R-XとのUllmann縮合反応あるいはBuchwald-Hartwig反応などのカップリング反応により製造することができる。反応条件は、スキーム1と同様の条件で実施可能である。
 また、化合物R-XのハロゲンXをボロン酸もしくはそのボロン酸エステルに変換した化合物を用いたChan-Lam-Evansカップリング反応によっても化合物(VI)を製造することができる。
が置換基を有してもよいアルキニル基である場合、化合物(VI)は、化合物(X)の保護基を脱保護したのち、化合物R-Xとの求核置換反応により製造することができる。反応条件は、スキーム1と同様の条件で実施可能である。 Compound (VI) used as the starting material in Scheme 4 can be produced, for example, by the method shown in Scheme 17.
Figure JPOXMLDOC01-appb-C000020
(In the formula, R2 and Z are defined as above, PG represents a protecting group, and X represents a halogen.)
When R 2 is an optionally substituted aryl group or an optionally substituted heteroaryl group, compound (VI) can be produced by deprotecting the protecting group of compound (X) and then subjecting the compound to a coupling reaction such as Ullmann condensation reaction or Buchwald-Hartwig reaction with compound R 2 -X. The reaction conditions can be the same as those in Scheme 1.
Compound (VI) can also be produced by a Chan-Lam-Evans coupling reaction using a compound in which the halogen X of compound R 2 -X is converted into a boronic acid or a boronic acid ester thereof.
When R 2 is an alkynyl group which may have a substituent, compound (VI) can be produced by deprotecting the protecting group of compound (X) and then subjecting the compound to a nucleophilic substitution reaction with compound R 2 -X. The reaction can be carried out under the same reaction conditions as in Scheme 1.

 スキーム5の原料として用いられる化合物(VIII)は、例えばスキーム18に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000021
(式中、R及びZは前記と同義であり、Rは低級アルキルを表わす。)
 化合物(VIII)は、化合物(XX)のエステル基を加水分解することにより製造できる。加水分解反応は、有機化学で一般的に用いられる反応条件で実施することができ、アルカリ性条件(水酸化ナトリウム、水酸化リチウムなど)もしくは酸性条件(塩酸、硫酸など)を用いることができる。 Compound (VIII) used as the starting material in Scheme 5 can be produced, for example, by the method shown in Scheme 18.
Figure JPOXMLDOC01-appb-C000021
(In the formula, R2 and Z are as defined above, and R6 represents lower alkyl.)
Compound (VIII) can be produced by hydrolyzing the ester group of compound (XX). The hydrolysis reaction can be carried out under reaction conditions generally used in organic chemistry, and alkaline conditions (sodium hydroxide, lithium hydroxide, etc.) or acidic conditions (hydrochloric acid, sulfuric acid, etc.) can be used.

 スキーム18の原料として用いられる化合物(XX)は、例えばスキーム19に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000022
(式中、R及びZは前記と同義であり、Rは低級アルキルを表わし、Xはハロゲンを表わす。)
 Rが置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基の場合、化合物(XX)は、化合物(XXI)の保護基を脱保護した後、化合物R-XとのUllmann縮合反応あるいはBuchwald-Hartwig反応などのカップリング反応により製造することができる。反応条件は、スキーム1と同様の条件で実施可能である。
 また、化合物R-XのハロゲンXをボロン酸もしくはそのボロン酸エステルに変換した化合物を用いたChan-Lam-Evansカップリング反応によっても化合物(XX)を製造することができる。
が置換基を有してもよいアルキニル基である場合、化合物(XX)は、化合物(XXI)の保護基を脱保護した後、化合物R-Xとの求核置換反応により製造することができる。反応条件は、スキーム1と同様の条件で実施可能である。 Compound (XX) used as the starting material in Scheme 18 can be produced, for example, by the method shown in Scheme 19.
Figure JPOXMLDOC01-appb-C000022
(In the formula, R2 and Z are as defined above, R6 represents lower alkyl, and X represents halogen.)
When R 2 is an optionally substituted aryl group or an optionally substituted heteroaryl group, compound (XX) can be produced by deprotecting the protecting group of compound (XXI) and then subjecting the compound to a coupling reaction such as Ullmann condensation reaction or Buchwald-Hartwig reaction with compound R 2 -X. The reaction can be carried out under the same reaction conditions as in Scheme 1.
Compound (XX) can also be produced by a Chan-Lam-Evans coupling reaction using a compound in which the halogen X of compound R 2 -X is converted to a boronic acid or a boronic acid ester thereof.
When R2 is an alkynyl group which may have a substituent, compound (XX) can be produced by deprotecting the protecting group of compound (XXI) and then subjecting the compound to a nucleophilic substitution reaction with compound R2 -X. The reaction can be carried out under the same reaction conditions as in Scheme 1.

 スキーム19の原料として用いられる化合物(XXI)は、例えばスキーム20に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000023
(式中、Zは前記と同義であり、Rは低級アルキルを表わし、PGは保護基を表わし、Xはハロゲンを表わす。)
 化合物(XXI)は、化合物(X)と化合物(XXII)を用いて、鈴木カップリング反応などのクロスカップリング反応により製造することができる。当該クロスカップリング反応は、スキーム4と同様の条件で実施することができる。また化合物(XXII)のボロニル基は、ボロン酸エステル基に置き換えて反応に使用することもできる。 Compound (XXI) used as the starting material in Scheme 19 can be produced, for example, by the method shown in Scheme 20.
Figure JPOXMLDOC01-appb-C000023
(In the formula, Z is as defined above, R6 represents a lower alkyl group, PG represents a protecting group, and X represents a halogen.)
Compound (XXI) can be produced by a cross-coupling reaction such as Suzuki coupling reaction using compound (X) and compound (XXII). The cross-coupling reaction can be carried out under the same conditions as in Scheme 4. In addition, the boronyl group of compound (XXII) can be replaced with a boronic acid ester group and used in the reaction.

 スキーム6の原料として用いられる本発明の化合物(I-c)は、式(I)においてRがメチル基である本発明の化合物(I-d)から、例えばスキーム21に表す方法によって製造することができる。

Figure JPOXMLDOC01-appb-C000024
(式中、R及びZは前記と同義であり、Xはハロゲンを表わす。)
 化合物(I-c)は、化合物(I-d)を通常の有機化学で用いられるハロゲン化剤、例えばベンジルトリメチルアンモニウムジクロロヨージド、NBS、NCSなどで処理することで製造することができる。すなわち、化合物(I-c)は、溶媒中、化合物(I-d)と、0.5~5.0モル当量、好ましくは0.8~2.0モル当量のハロゲン化剤を反応させることによって得ることができる。溶媒は反応に不活性なものであればいずれでも良く、特に限定されるものではなく、ジクロロメタン、THF、DMFなどを用いることができる。反応は-20~100℃において、0.25~10時間撹拌することで実施することができるが、好ましくは室温~80℃において0.5~5時間撹拌することで実施することができる。
 なお、上記の方法を適宜組み合わせ、有機合成化学で通常用いられる方法(例えば、アミノ基のアルキル化反応、アルキルチオ基をスルホキシド基もしくはスルホン基へ酸化する反応、アルコキシ基をヒドロキシル基、もしくはその逆へ変換する反応)を実施することにより、所望の位置に所望の官能基を有する本発明の化合物(I)を得ることができる。 The compound (Ic) of the present invention used as the starting material in Scheme 6 can be produced from the compound (Id) of the present invention, which is represented by formula (I) in which R 1 is a methyl group, by, for example, the method shown in Scheme 21.
Figure JPOXMLDOC01-appb-C000024
(In the formula, R2 and Z are defined as above, and X represents a halogen.)
Compound (I-c) can be produced by treating compound (I-d) with a halogenating agent used in ordinary organic chemistry, such as benzyltrimethylammonium dichloroiodide, NBS, NCS, etc. That is, compound (I-c) can be obtained by reacting compound (I-d) with 0.5 to 5.0 molar equivalents, preferably 0.8 to 2.0 molar equivalents of a halogenating agent in a solvent. The solvent may be any one inert to the reaction, and is not particularly limited, and dichloromethane, THF, DMF, etc. can be used. The reaction can be carried out by stirring at -20 to 100°C for 0.25 to 10 hours, but preferably at room temperature to 80°C for 0.5 to 5 hours.
In addition, by appropriately combining the above methods and carrying out a method commonly used in organic synthetic chemistry (e.g., alkylation reaction of an amino group, oxidation reaction of an alkylthio group to a sulfoxide group or a sulfone group, conversion of an alkoxy group to a hydroxyl group, or vice versa), it is possible to obtain compound (I) of the present invention having a desired functional group at a desired position.

[本発明の化合物(I)の用途]
 本発明の化合物(I)またはその薬学的に許容される塩は、経口投与、非経口投与または局所的投与に適した従来の薬学製剤(医薬組成物)の形態に調製することができる。
経口投与のための製剤は、錠剤、顆粒、粉末、カプセルなどの固形剤、およびシロップなどの液体製剤を含む。これらの製剤は従来の方法によって調製することができる。固形剤は、ラクトース、コーンスターチなどのデンプン、微結晶性セルロースなどの結晶セルロース、ヒドロキシプロピルセルロース、カルシウムカルボキシメチルセルロース、タルク、ステアリン酸マグネシウムなどのような従来の薬学的担体を用いることによって調製することができる。カプセルは、このように調製した顆粒または粉末をカプセルに包むことによって調製することができる。シロップは、ショ糖、カルボキシメチルセルロースなどを含む水溶液中で、本発明の化合物(I)またはその薬学的に許容される塩を溶解または懸濁することによって調製することができる。
 非経口投与のための製剤は、点滴注入などの注入物を含む。注入製剤もまた従来の方法によって調製することができ、等張化剤(例えば、マンニトール、塩化ナトリウム、グルコース、ソルビトール、グリセロール、キシリトール、フルクトース、マルトース、マンノース)、安定化剤(例えば、亜硫酸ナトリウム、アルブミン)、防腐剤(例えば、ベンジルアルコール、p-オキシ安息香酸メチル)中に適宜組み入れることができる。
 本発明の化合物(I)またはその薬学的に許容される塩の用量は、疾患の重症度、患者の年齢および体重、投薬形態などに従って変化させることができるが、通常は成人において1日あたり1mg~1,000mgの範囲であり、それは経口経路または非経口経路によって、1回、または2回もしくは3回に分割して投与することができる。
 また、本発明の化合物(I)またはその薬学的に許容される塩は、ALK5阻害剤として、実験用、研究用の試薬として用いることもできる。さらに、本発明の化合物(I)を放射性標識した化合物は、PET用分子プローブとしても用いることもできる。
[Use of compound (I) of the present invention]
The compound (I) of the present invention or a pharma- ceutically acceptable salt thereof can be prepared in the form of a conventional pharmaceutical preparation (pharmaceutical composition) suitable for oral, parenteral or topical administration.
Preparations for oral administration include solid preparations such as tablets, granules, powders, capsules, and liquid preparations such as syrups. These preparations can be prepared by conventional methods. Solid preparations can be prepared by using conventional pharmaceutical carriers such as lactose, starch such as corn starch, crystalline cellulose such as microcrystalline cellulose, hydroxypropyl cellulose, calcium carboxymethyl cellulose, talc, magnesium stearate, etc. Capsules can be prepared by encapsulating the granules or powders thus prepared. Syrups can be prepared by dissolving or suspending the compound (I) of the present invention or a pharma- ceutically acceptable salt thereof in an aqueous solution containing sucrose, carboxymethyl cellulose, etc.
Preparations for parenteral administration include injections such as drip infusions. Injection preparations can also be prepared by conventional methods, and can be appropriately incorporated into isotonic agents (e.g., mannitol, sodium chloride, glucose, sorbitol, glycerol, xylitol, fructose, maltose, mannose), stabilizers (e.g., sodium sulfite, albumin), and preservatives (e.g., benzyl alcohol, methyl p-oxybenzoate).
The dose of the compound (I) of the present invention or a pharma- ceutically acceptable salt thereof can be varied according to the severity of the disease, the age and body weight of the patient, the dosage form, etc., but is usually in the range of 1 mg to 1,000 mg per day for an adult, which can be administered orally or parenterally in a single dose, or in two or three divided doses.
In addition, the compound (I) of the present invention or a pharma- ceutical acceptable salt thereof can be used as an ALK5 inhibitor and as a reagent for experiments and research. Furthermore, the compound (I) of the present invention that is radiolabeled can also be used as a molecular probe for PET.

以下に実施例および試験例などを挙げて本発明をさらに具体的に説明するが、これらの実施例により本発明が限定されるものではない。
 化合物の同定は水素核磁気共鳴スペクトル(1H-NMR)およびマススペクトル(MS)により行った。1H-NMRは、特に指示のないかぎりは400MHzで測定されたものであり、また化合物および測定条件によっては交換性水素が明瞭に観測されない場合がある。なお、br.は幅広いシグナル(ブロード)を意味する。HPLC分取クロマトグラフィーは、市販のODSカラムを用い、特に記載のない限りは水/メタノール(ギ酸を含む)を溶出液としてグラジェントモードにて分取した。
The present invention will be explained in more detail below by way of examples and test examples, but the present invention is not limited to these examples.
Compounds were identified by hydrogen nuclear magnetic resonance spectroscopy ( 1 H-NMR) and mass spectroscopy (MS). 1 H-NMR was measured at 400 MHz unless otherwise specified, and exchangeable hydrogen may not be clearly observed depending on the compound and measurement conditions. Note that br. means broad signal (broad). For HPLC preparative chromatography, a commercially available ODS column was used, and unless otherwise specified, separation was performed in gradient mode using water/methanol (containing formic acid) as the eluent.

参考例1
5-ブロモ-2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000025
(第1工程)
1-[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]チオ尿素
Figure JPOXMLDOC01-appb-C000026
 1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-アミン(80g,479mmol)のジクロロメタン溶液(750mL)に、0℃でチオホスゲン(36.7mL,479mmol)とDIPEA(41.8mL,239.5mmol)を加え、混合物を室温で2時間撹拌した。反応混合物を減圧下で濃縮し、4-イソチオシアナート-1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール(80g)を得た。得られた化合物(80g,382mmol)のジクロロメタン溶液(750mL)に、0℃で濃アンモニア水溶液(250mL)を滴下し、混合物を室温で3時間撹拌した。反応混合物に水を加え、ジクロロメタンで抽出した。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、溶媒を減圧留去した。残渣をカラムクロマトグラフィー(シリカゲル、酢酸エチル)で精製し、標記化合物(33g)を得た。
LCMS (m/z): 227.2 [M+H]+. Reference Example 1
Preparation of 5-bromo-2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000025
(First step)
1-[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]thiourea
Figure JPOXMLDOC01-appb-C000026
Thiophosgene (36.7 mL, 479 mmol) and DIPEA (41.8 mL, 239.5 mmol) were added to a dichloromethane solution (750 mL) of 1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-amine (80 g, 479 mmol) at 0° C., and the mixture was stirred at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure to obtain 4-isothiocyanato-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (80 g). A concentrated aqueous ammonia solution (250 mL) was added dropwise to a dichloromethane solution (750 mL) of the obtained compound (80 g, 382 mmol) at 0° C., and the mixture was stirred at room temperature for 3 hours. Water was added to the reaction mixture, and the mixture was extracted with dichloromethane. The organic layer was washed with saturated saline, dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure. The residue was purified by column chromatography (silica gel, ethyl acetate) to obtain the title compound (33 g).
LCMS (m/z): 227.2 [M+H] + .

(第2工程)
2-{[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル

Figure JPOXMLDOC01-appb-C000027
 第1工程を繰り返し得られた1-[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]チオ尿素(65g,287.6mmol)のエタノール溶液(200mL)に、0℃で炭酸カリウム(119.1g,862.8mmol)および3-ブロモ-2-オキソプロパン酸(84.1g,431.4mmol)を加え、混合物を室温で16時間撹拌した。反応混合物を水で希釈し、酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥し、減圧下濃縮した。残渣をカラムクロマトグラフィー(シリカゲル、酢酸エチル)で精製し、標記化合物(60g)を得た。
LC-MS (m/z): 323.3 [M+H]+. (Second step)
2-{[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylate ethyl
Figure JPOXMLDOC01-appb-C000027
To an ethanol solution (200 mL) of 1-[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]thiourea (65 g, 287.6 mmol) obtained by repeating the first step, potassium carbonate (119.1 g, 862.8 mmol) and 3-bromo-2-oxopropanoic acid (84.1 g, 431.4 mmol) were added at 0° C., and the mixture was stirred at room temperature for 16 hours. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, ethyl acetate) to obtain the title compound (60 g).
LC-MS (m/z): 323.3 [M+H] + .

(第3工程)
2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル

Figure JPOXMLDOC01-appb-C000028
 2-{[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル(30g,93.2mmol)の1,2-ジクロロエタン溶液(500mL)にシクロプロピルボロン酸 (39.6g,465.8mmol)、酢酸銅(II)(33.7g,186.3mmol)、2,2’-ビピリジン(17.4g,111.8mmol)および炭酸ナトリウム(19.8g,186.3mmol)を室温で加え、混合物を酸素雰囲気下、50℃で2時間撹拌した。反応混合物を水で希釈し、酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥し、減圧下濃縮した。残渣をカラムクロマトグラフィー(シリカゲル、石油エーテル/酢酸エチル)で精製し、標記化合物(17g)を得た。
LC-MS (m/z): 363.2 [M+H]+. (Third process)
2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylate ethyl
Figure JPOXMLDOC01-appb-C000028
Cyclopropylboronic acid (39.6 g, 465.8 mmol), copper(II) acetate (33.7 g, 186.3 mmol), 2,2'-bipyridine (17.4 g, 111.8 mmol) and sodium carbonate (19.8 g, 186.3 mmol) were added to a 1,2-dichloroethane solution (500 mL) of ethyl 2-{[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylate (30 g, 93.2 mmol), and the mixture was stirred at 50°C for 2 hours under an oxygen atmosphere. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate) to obtain the title compound (17 g).
LC-MS (m/z): 363.2 [M+H] + .

(第4工程)
5-ブロモ-2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル

Figure JPOXMLDOC01-appb-C000029
 第3工程を繰り返し得られた2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル(35g,96.7mmol)のジクロロメタン溶液(500mL)に、0℃でNBS(13.8g,77.4mmol)を加え、混合物を0℃で2時間撹拌した。反応混合物に氷水を加え、酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥後、溶媒を減圧留去した。残渣をカラムクロマトグラフィー(シリカゲル、石油エーテル/酢酸エチル)で精製し、標記化合物(22g)を得た。
1H-NMR (500 MHz, CDCl3) δ 8.22 (d, J = 0.8 Hz, 1H), 7.75 (d, J = 0.8 Hz, 1H), 5.37 (dd, J = 9.6, 2.7 Hz, 1H), 4.37 (q, J = 7.1 Hz, 2H), 4.11 - 4.00 (m, 1H), 3.76 - 3.62 (m, 1H), 3.04 - 2.91 (m, 1H), 2.24 - 1.96 (m, 3H), 1.84 - 1.52 (m, 1H), 1.41 (t, J = 7.1 Hz, 3H), 1.35 - 1.17 (m, 2H), 1.15 - 1.04 (m, 2H), 0.96 - 0.90 (m, 2H); LC-MS (m/z): 443.1 ([M+2+H]+, isotopic mass). (Fourth step)
5-Bromo-2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylate ethyl
Figure JPOXMLDOC01-appb-C000029
NBS (13.8 g, 77.4 mmol) was added to a dichloromethane solution (500 mL) of ethyl 2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylate (35 g, 96.7 mmol) obtained by repeating the third step, and the mixture was stirred at 0° C. for 2 hours. Ice water was added to the reaction mixture, and the mixture was extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure. The residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate) to obtain the title compound (22 g).
1H -NMR (500 MHz, CDCl3 ) δ 8.22 (d, J = 0.8 Hz, 1H), 7.75 (d, J = 0.8 Hz, 1H), 5.37 (dd, J = 9.6, 2.7 Hz, 1H), 4.37 (q, J = 7.1 Hz, 2H), 4.11 - 4.00 (m, 1H), 3.76 - 3.62 (m, 1H), 3.04 - 2.91 (m, 1H), 2.24 - 1.96 (m, 3H), 1.84 - 1.52 (m, 1H), 1.41 (t, J = 7.1 Hz, 3H), 1.35 - 1.17 (m, 2H), 1.15 - 1.04 (m, 2H), 0.96 - 0.90 (m, 2H); LC-MS (m/z): 443.1 ([M+2+H] + , isotopic mass).

(第5工程)
5-ブロモ-2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボキサミドの製造
 5-ブロモ-2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル(1g,2.7mmol)のエタノール溶液(10mL)に濃アンモニア水溶液(10mL)を加え、混合物を100℃で一晩攪拌した。反応混合物を室温まで冷却し、水と酢酸エチルを加えた。有機層を無水硫酸ナトリウムで乾燥後、溶媒を減圧留去し、標記化合物(0.82g)を得た。
1H-NMR (DMSO-d6) δ 8.27 (d, J = 0.8 Hz, 1H), 7.80 (d, J = 0.8 Hz, 1H), 7.61 - 7.47 (m, 2H), 5.39 (dd, J = 10.1, 2.5 Hz, 1H), 3.99 - 3.80 (m, 1H), 3.70 - 3.49 (m, 1H), 3.14 - 2.96 (m, 1H), 2.26 - 2.05 (m, 1H), 2.01 - 1.81 (m, 2H), 1.76 - 1.58 (m, 1H), 1.59 - 1.44 (m, 2H), 1.14 - 1.01 (m, 2H), 0.84 - 0.69 (m, 2H); LC-MS (m/z): 412.1 ([M+2+H]+, isotopic mass).
(Fifth step)
Preparation of 5-bromo-2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxamide Concentrated aqueous ammonia (10 mL) was added to an ethanol solution (10 mL) of ethyl 5-bromo-2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylate (1 g, 2.7 mmol), and the mixture was stirred at 100° C. overnight. The reaction mixture was cooled to room temperature, and water and ethyl acetate were added. The organic layer was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to obtain the title compound (0.82 g).
1H -NMR (DMSO-d6) δ 8.27 (d, J = 0.8 Hz, 1H), 7.80 (d, J = 0.8 Hz, 1H), 7.61 - 7.47 (m, 2H), 5.39 (dd, J = 10.1, 2.5 Hz, 1H), 3.99 - 3.80 (m, 1H), 3.70 - 3.49 (m, 1H), 3.14 - 2.96 (m, 1H), 2.26 - 2.05 (m, 1H), 2.01 - 1.81 (m, 2H), 1.76 - 1.58 (m, 1H), 1.59 - 1.44 (m, 2H), 1.14 - 1.01 (m, 2H), 0.84 - 0.69 (m, 2H); LC-MS (m/z): 412.1 ([M+2+H] + , isotopic mass).

参考例2
1-[5-(4,4,5,5-テトラメチル-1,3,2-ジオキサボロラン-2-イル)-1H-ピロール-3-イル]エタノンの製造

Figure JPOXMLDOC01-appb-C000030
 1-(1H-ピロール-3-イル)エタン-1-オン(200mg,1.8mmol)と4,4,4’,4’,5,5,5’,5’-オクタメチル-2,2’-ビ(1,3,2-ジオキサボロラン)(233mg,0.92mmol)および4,4’-ジ-tert-ブチル-2,2’-ビピリジン(7.4mg,0.027mmol)のシクロヘキサン溶液(3mL)に、(1,5-シクロオクタジエン)(メトキシ)イリジウム(I)(ダイマー)(9.4mg,0.014mmol)を室温で加えて、混合物を90℃で6時間攪拌した。反応混合物を室温まで冷却し、析出した固体をろ取し、n-ヘキサンと水で洗浄して、標記化合物(270mg)を得た。
1H-NMR (DMSO-d6) δ 11.81 (s, 1H), 7.62 (dd, J = 3.2, 1.5 Hz, 1H), 6.95 - 6.91 (m, 1H), 2.32 (s, 3H), 1.28 (s, 12H); LC-MS (m/z): 236.2 [M+H]+. Reference Example 2
Preparation of 1-[5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrrol-3-yl]ethanone
Figure JPOXMLDOC01-appb-C000030
1-(1H-pyrrol-3-yl)ethan-1-one (200 mg, 1.8 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (233 mg, 0.92 mmol), and 4,4'-di-tert-butyl-2,2'-bipyridine (7.4 mg, 0.027 mmol) were added to a cyclohexane solution (3 mL) at room temperature, and the mixture was stirred at 90° C. for 6 hours. The reaction mixture was cooled to room temperature, and the precipitated solid was collected by filtration and washed with n-hexane and water to obtain the title compound (270 mg).
1H -NMR (DMSO-d6) δ 11.81 (s, 1H), 7.62 (dd, J = 3.2, 1.5 Hz, 1H), 6.95 - 6.91 (m, 1H), 2.32 (s, 3H), 1.28 (s, 12H); LC-MS (m/z): 236.2 [M+H] + .

参考例3
5-(4,4,5,5-テトラメチル-1,3,2-ジオキサボロラン-2-イル)-1H-ピロール-3-カルボン酸メチルの製造

Figure JPOXMLDOC01-appb-C000031
 1H-ピロール-3-カルボン酸メチル(250mg,2mmol)と4,4,4’,4’,5,5,5’,5’-オクタメチル-2,2’-ビ(1,3,2-ジオキサボロラン)(254mg,1mmol)および4,4’-ジ-tert-ブチル-2,2’-ビピリジン(8mg,0.03mmol)のシクロヘキサン溶液(3mL)に(1,5-シクロオクタジエン)(メトキシ)イリジウム(I)(ダイマー)(9.9mg,0.015mmol)を室温で加えて、混合物を90℃で2時間攪拌した。反応混合物を室温まで冷却し、析出した固体をろ取、n-ヘキサンと水で洗浄し、標記化合物(314mg)を得た。
1H-NMR (DMSO-d6) δ 11.84 (s, 1H), 7.48 (dd, J = 3.2, 1.4 Hz, 1H), 6.88 (dd, J = 2.5, 1.4 Hz, 1H), 3.69 (s, 3H), 1.27 (s, 12H); LC-MS (m/z): 252.2 [M+H]+. Reference Example 3
Preparation of methyl 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrrole-3-carboxylate
Figure JPOXMLDOC01-appb-C000031
To a cyclohexane solution (3 mL) of 1H-pyrrole-3-methyl carboxylate (250 mg, 2 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (254 mg, 1 mmol), and 4,4'-di-tert-butyl-2,2'-bipyridine (8 mg, 0.03 mmol), (1,5-cyclooctadiene)(methoxy)iridium(I)(dimer) (9.9 mg, 0.015 mmol) was added at room temperature, and the mixture was stirred at 90° C. for 2 hours. The reaction mixture was cooled to room temperature, and the precipitated solid was collected by filtration and washed with n-hexane and water to obtain the title compound (314 mg).
1H -NMR (DMSO-d6) δ 11.84 (s, 1H), 7.48 (dd, J = 3.2, 1.4 Hz, 1H), 6.88 (dd, J = 2.5, 1.4 Hz, 1H), 3.69 (s, 3H), 1.27 (s, 12H); LC-MS (m/z): 252.2 [M+H] + .

参考例4
5-(4-カルバモイル-2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-5-イル)-1H-ピロール-3-カルボン酸メチルの製造

Figure JPOXMLDOC01-appb-C000032
 5-ブロモ-2-(シクロプロピル(1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル)アミノ)チアゾール-4-カルボキサミド(参考例1)(5.0g,12.2mmol)と5-(4,4,5,5-テトラメチル-1,3,2-ジオキサボロラン-2-イル)-1H-ピロール-3-カルボン酸メチル(参考例3)(4.5g,18.2mmol)の1,4-ジオキサン/水の混合液(100mL/10mL)に、ジクロロビス[ジ‐tert‐ブチル(4‐ジメチルアミノフェニル)ホスフィン]パラジウム(II)(856mg,1.22mmol)とフッ化セシウム(3.65g,24.3mmol)を加え、混合物を窒素気流下、100℃で2時間攪拌した。反応混合物を室温まで冷却後、水を加えて析出した固体をろ取し、標記化合物(5g)を得た。
1H-NMR (DMSO-d6) δ 13.25 (br.s, 1H), 8.28 (s, 1H), 8.03 (s, 1H), 7.83 - 7.80 (m, 2H), 7.59 (t, J = 0.8 Hz, 1H), 6.67 (m. 1H), 5.40 (m, 1H), 3.86 (d, J = 4.3 Hz, 1H), 3.71 (s, 3H), 3.68 - 3.61 (m, 2H), 2.21 - 2.03 (m, 2H), 1.94 - 1.88 (m, 2H), 1.55 - 1.35 (m, 2H), 1.16 - 1.10 (m, 2H), 0.82 - 0.77 (m, 2H); LC-MS (m/z): 457.1 [M+H]+. Reference Example 4
Preparation of methyl 5-(4-carbamoyl-2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazol-5-yl)-1H-pyrrole-3-carboxylate
Figure JPOXMLDOC01-appb-C000032
To a mixture (100 mL/10 mL) of 5-bromo-2-(cyclopropyl(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)amino)thiazole-4-carboxamide (Reference Example 1) (5.0 g, 12.2 mmol) and 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrrole-3-methyl carboxylate (Reference Example 3) (4.5 g, 18.2 mmol) in 1,4-dioxane/water, dichlorobis[di-tert-butyl(4-dimethylaminophenyl)phosphine]palladium(II) (856 mg, 1.22 mmol) and cesium fluoride (3.65 g, 24.3 mmol) were added, and the mixture was stirred at 100° C. for 2 hours under a nitrogen stream. The reaction mixture was cooled to room temperature, water was added, and the precipitated solid was collected by filtration to obtain the title compound (5 g).
1H -NMR (DMSO- d6 ) δ 13.25 (br.s, 1H), 8.28 (s, 1H), 8.03 (s, 1H), 7.83 - 7.80 (m, 2H), 7.59 (t, J = 0.8 Hz, 1H), 6.67 (m. 1H), 5.40 (m, 1H), 3.86 (d, J = 4.3 Hz, 1H), 3.71 (s, 3H), 3.68 - 3.61 (m, 2H), 2.21 - 2.03 (m, 2H), 1.94 - 1.88 (m, 2H), 1.55 - 1.35 (m, 2H), 1.16 - 1.10 (m, 2H), 0.82 - 0.77 (m, 2H); LC-MS (m/z): 457.1 [M+H] + .

参考例5
5-{4-カルバモイル-2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ]チアゾール-5-イル}-1H-ピロール-3-カルボン酸メチルの製造

Figure JPOXMLDOC01-appb-C000033
 5-(4-カルバモイル-2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-5-イル)-1H-ピロール-3-カルボン酸メチル(参考例4)(5g,10.9mmol)の1,4-ジオキサン(50mL)溶液に、塩化水素・1,4-ジオキサン溶液(約4mol/L)(50mL)を加え、室温で16時間攪拌した。溶媒を減圧留去し、標記化合物(4g)を得た。
1H-NMR (DMSO-d6) δ 13.24 (br.s, 1H), 12.86 (br.s, 1H), 8.01 - 7.81 (m, 3H), 7.58 (br.s, 1H), 7.25 - 7.16 (m, 1H), 6.65 (s, 1H), 3.71 (s, 3H), 3.16 - 3.14 (m, 1H), 1.08 - 1.05 (m, 2H), 0.86 - 0.76 (m, 2H); LC-MS (m/z): 373.2 [M+H]+. Reference Example 5
Preparation of methyl 5-{4-carbamoyl-2-[cyclopropyl(1H-pyrazol-4-yl)amino]thiazol-5-yl}-1H-pyrrole-3-carboxylate
Figure JPOXMLDOC01-appb-C000033
To a solution of 5-(4-carbamoyl-2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazol-5-yl)-1H-pyrrole-3-methyl carboxylate (Reference Example 4) (5 g, 10.9 mmol) in 1,4-dioxane (50 mL), hydrogen chloride-1,4-dioxane solution (about 4 mol/L) (50 mL) was added and stirred at room temperature for 16 hours. The solvent was evaporated under reduced pressure to obtain the title compound (4 g).
1H -NMR (DMSO- d6 ) δ 13.24 (br.s, 1H), 12.86 (br.s, 1H), 8.01 - 7.81 (m, 3H), 7.58 (br.s, 1H), 7.25 - 7.16 (m, 1H), 6.65 (s, 1H), 3.71 (s, 3H), 3.16 - 3.14 (m, 1H), 1.08 - 1.05 (m, 2H), 0.86 - 0.76 (m, 2H); LC-MS (m/z): 373.2 [M+H] + .

参考例6
5-{4-カルバモイル-2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ]チアゾール-5-イル}-1H-ピロール-3-カルボン酸の製造

Figure JPOXMLDOC01-appb-C000034
 5-{4-カルバモイル-2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ]チアゾール-5-イル}-1H-ピロール-3-カルボン酸メチル(参考例5)(0.78g,2.1mmol)のメタノール溶液(7mL)に4M水酸化ナトリウム水溶液(7mL)を加え、混合物を70℃で3時間攪拌した。反応混合物に濃塩酸を加えて酸性にし、析出した固体をろ取し、標記化合物(0.54g)を得た。
1H-NMR (DMSO-d6) δ 13.18 (br.s, 1H), 8.00 - 7.94 (m, 3H), 7.77 (br.s, 1H), 7.52 - 7.48 (m, 1H), 6.60 (t, J = 0.4 Hz, 1H), 3.19 - 3.12 (m, 1H), 1.23 - 1.12 (m, 2H), 0.85 - 0.76 (m, 2H); LC-MS (m/z): 359.1 [M+H]+. Reference Example 6
Preparation of 5-{4-carbamoyl-2-[cyclopropyl(1H-pyrazol-4-yl)amino]thiazol-5-yl}-1H-pyrrole-3-carboxylic acid
Figure JPOXMLDOC01-appb-C000034
A 4M aqueous sodium hydroxide solution (7 mL) was added to a methanol solution (7 mL) of methyl 5-{4-carbamoyl-2-[cyclopropyl(1H-pyrazol-4-yl)amino]thiazol-5-yl}-1H-pyrrole-3-carboxylate (Reference Example 5) (0.78 g, 2.1 mmol), and the mixture was stirred for 3 hours at 70° C. The reaction mixture was acidified with concentrated hydrochloric acid, and the precipitated solid was collected by filtration to obtain the title compound (0.54 g).
1H -NMR (DMSO- d6 ) δ 13.18 (br.s, 1H), 8.00 - 7.94 (m, 3H), 7.77 (br.s, 1H), 7.52 - 7.48 (m, 1H), 6.60 (t, J = 0.4 Hz, 1H), 3.19 - 3.12 (m, 1H), 1.23 - 1.12 (m, 2H), 0.85 - 0.76 (m, 2H); LC-MS (m/z): 359.1 [M+H] + .

参考例7
2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ]-5-[4-(ジメチルカルバモイル)-1H-ピロール-2-イル]チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000035
 5-{4-カルバモイル-2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ]チアゾール-5-イル}-1H-ピロール-3-カルボン酸(参考例6)(2g,5.59mol)のDMF溶液(10mL)に、ジメチルアミン塩酸塩(0.91g,11.17mol)及びEDC・HCl(2.14g,11.17mol)を加え、室温で3時間攪拌した。反応混合物を水で希釈し、析出した固体をろ取し、標記化合物(0.9g)を得た。
1H-NMR (500 MHz, DMSO-d6) δ 13.08 (br.s, 1H), 12.79 (br.s, 1H), 8.16 (s, 1H), 7.96 (br.s, 1H), 7.74 (d, J = 9.8 Hz, 2H), 7.33 (dd, J = 1.5, 2.7 Hz, 1H), 6.55 (dd, J = 1.7, 2.3 Hz, 1H), 3.18 - 3.14 (m, 1H), 3.13 - 2.95 (m, 6H), 1.11 - 1.01 (m, 2H), 0.82 - 0.75 (m, 2H); LC-MS (m/z): 386.1 [M+H]+. Reference Example 7
Preparation of 2-[cyclopropyl(1H-pyrazol-4-yl)amino]-5-[4-(dimethylcarbamoyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000035
Dimethylamine hydrochloride (0.91 g, 11.17 mol) and EDC.HCl (2.14 g, 11.17 mol) were added to a DMF solution (10 mL) of 5-{4-carbamoyl-2-[cyclopropyl(1H-pyrazol-4-yl)amino]thiazol-5-yl}-1H-pyrrole-3-carboxylic acid (Reference Example 6) (2 g, 5.59 mol), and the mixture was stirred at room temperature for 3 hours. The reaction mixture was diluted with water, and the precipitated solid was collected by filtration to obtain the title compound (0.9 g).
1H -NMR (500 MHz, DMSO- d6 ) δ 13.08 (br.s, 1H), 12.79 (br.s, 1H), 8.16 (s, 1H), 7.96 (br.s, 1H), 7.74 (d, J = 9.8 Hz, 2H), 7.33 (dd, J = 1.5, 2.7 Hz, 1H), 6.55 (dd, J = 1.7, 2.3 Hz, 1H), 3.18 - 3.14 (m, 1H), 3.13 - 2.95 (m, 6H), 1.11 - 1.01 (m, 2H), 0.82 - 0.75 (m, 2H); LC-MS (m/z): 386.1 [M+H] + .

参考例8
2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ]-5-[4-(モルホリン-4-カルボニル)-1H-ピロール-2-イル]チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000036
 5-{4-カルバモイル-2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ]チアゾール-5-イル}-1H-ピロール-3-カルボン酸(参考例6)(2g,5.59mol)のDMF溶液(10mL)にモルホリン(0.97g,11.17mol)、HATU(4.24g,11.17mol)及びDIPEA(1.94ml,11.17mol)を加え、室温で一晩攪拌した。混合物を水で希釈し、析出した固体をろ取した。固体をカラムクロマトグラフィー(シリカゲル、クロロホルム/メタノール)で精製し、標記化合物(0.8g)を得た。
1H-NMR (DMSO-d6) δ 13.0 (br.s, 1H), 8.23 - 8.16 (m, 2H), 7.80 (s, 2H), 7.38 (d, J = 9.7 Hz, 1H), 7.13 (t, J = 9.7 Hz, 1H), 6.56 (dd, J = 1.7, 2.3 Hz, 1H), 4.11 - 4.07 (m, 1H), 3.60 - 3.41 (m, 8H), 1.22 - 1.05 (m, 2H), 0.90 - 0.89 (m, 2H); LC-MS (m/z): 428.2 [M+H]+. Reference Example 8
Preparation of 2-[cyclopropyl(1H-pyrazol-4-yl)amino]-5-[4-(morpholine-4-carbonyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000036
Morpholine (0.97 g, 11.17 mol), HATU (4.24 g, 11.17 mol) and DIPEA (1.94 ml, 11.17 mol) were added to a DMF solution (10 mL) of 5-{4-carbamoyl-2-[cyclopropyl(1H-pyrazol-4-yl)amino]thiazol-5-yl}-1H-pyrrole-3-carboxylic acid (Reference Example 6) (2 g, 5.59 mol), and the mixture was stirred at room temperature overnight. The mixture was diluted with water, and the precipitated solid was collected by filtration. The solid was purified by column chromatography (silica gel, chloroform/methanol) to obtain the title compound (0.8 g).
1H -NMR (DMSO-d6) δ 13.0 (br.s, 1H), 8.23 - 8.16 (m, 2H), 7.80 (s, 2H), 7.38 (d, J = 9.7 Hz, 1H), 7.13 (t, J = 9.7 Hz, 1H), 6.56 (dd, J = 1.7, 2.3 Hz, 1H), 4.11 - 4.07 (m, 1H), 3.60 - 3.41 (m, 8H), 1.22 - 1.05 (m, 2H), 0.90 - 0.89 (m, 2H); LC-MS (m/z): 428.2 [M+H] + .

参考例9
5-(4-アセチル-1H-ピロール-2-イル)-2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000037
 5-ブロモ-2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボキサミド(参考例1)(5g,12.2mmol)と1-[5-(4,4,5,5-テトラメチル-1,3,2-ジオキサボロラン-2-イル)-1H-ピロール-3-イル]エタノン(参考例2)(4.28g,18.25mmol)の1,4-ジオキサン/水の混合液(100mL/10mL)に、ジクロロビス[ジ‐tert‐ブチル(4‐ジメチルアミノフェニル)ホスフィン]パラジウム(II)(861mg,1.22mmol)とフッ化セシウム(3.65g,24.3mmol)を加え、混合物を窒素気流下、100℃で2時間攪拌した。反応混合物を室温まで冷却後、溶媒を減圧留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、石油エーテル/酢酸エチル)で精製し、標記化合物(4g)を得た。
1H-NMR (CDCl3) δ 13.16 (br.s, 1H), 7.77 (dd, J = 0.7, 14.2 Hz, 2H), 7.44 (dd, J = 1.6, 3.1 Hz, 1H), 7.36 (br.s, 1H), 6.83 (dd, J = 1.5, 2.2 Hz, 1H), 5.64 (d, J = 3.2 Hz, 1H), 5.37 (dd, J = 3.7, 8.6 Hz, 1H), 4.12 - 4.05 (m, 1H), 3.78 - 3.68 (m, 1H), 3.06 - 3.01 (m, 1H), 2.43 (s, 3H), 2.17 - 2.01 (m, 3H), 1.76 - 1.62 (m, 3H), 1.11 - 1.01 (m, 2H), 0.94 - 0.88 (m, 2H); LC-MS(m/z): 441.08 [M+H]+. Reference Example 9
Preparation of 5-(4-acetyl-1H-pyrrol-2-yl)-2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000037
5-Bromo-2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxamide (Reference Example 1) (5 g, 12.2 mmol) and 1-[5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrrol-3-yl]ethanone (Reference Example 2) (4.28 g, 18.25 mmol) in 1,4-dioxane/water mixture (100 mL/10 mL) were added dichlorobis[di-tert-butyl(4-dimethylaminophenyl)phosphine]palladium(II) (861 mg, 1.22 mmol) and cesium fluoride (3.65 g, 24.3 mmol), and the mixture was stirred at 100° C. for 2 hours under a nitrogen stream. The reaction mixture was cooled to room temperature, the solvent was distilled off under reduced pressure, and the resulting residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate) to obtain the title compound (4 g).
1H -NMR ( CDCl3 ) δ 13.16 (br.s, 1H), 7.77 (dd, J = 0.7, 14.2 Hz, 2H), 7.44 (dd, J = 1.6, 3.1 Hz, 1H), 7.36 (br.s, 1H), 6.83 (dd, J = 1.5, 2.2 Hz, 1H), 5.64 (d, J = 3.2 Hz, 1H), 5.37 (dd, J = 3.7, 8.6 Hz, 1H), 4.12 - 4.05 (m, 1H), 3.78 - 3.68 (m, 1H), 3.06 - 3.01 (m, 1H), 2.43 (s, 3H), 2.17 - 2.01 (m, 3H), 1.76 - 1.62 (m, 3H), 1.11 - 1.01 (m, 2H), 0.94 - 0.88 (m, 2H); LC-MS(m/z): 441.08 [M+H] + .

参考例10
5-(4-アセチル-1H-ピロール-2-イル)-2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ)チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000038
 5-(4-アセチル-1H-ピロール-2-イル)-2-{シクロプロピル[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボキサミド(参考例9)(1.5g,3.41mmol)の1,4-ジオキサン(15mL)溶液に、塩化水素・1,4-ジオキサン溶液(約4mol/L)(10mL)を加え、室温で4時間攪拌した。溶媒を減圧留去し、標記化合物(1g)を得た。
1H-NMR (DMSO-d6) δ 13.27 (br.s, 1H), 12.80 (br.s, 1H), 8.16 (br.s, 1H), 8.00 (s, 1H), 7.76 (br.s, 2H), 7.68 (dd, J = 1.6, 2.8 Hz, 1H), 6.68 (s, 1H), 3.22 - 3.13 (m, 1H), 2.33 (s, 3H), 1.13 - 1.01 (m, 2H), 0.83 - 0.76 (m, 2H); LC-MS(m/z): 357.09 [M+H]+. Reference Example 10
Preparation of 5-(4-acetyl-1H-pyrrol-2-yl)-2-[cyclopropyl(1H-pyrazol-4-yl)amino)thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000038
To a solution of 5-(4-acetyl-1H-pyrrol-2-yl)-2-{cyclopropyl[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxamide (Reference Example 9) (1.5 g, 3.41 mmol) in 1,4-dioxane (15 mL), hydrogen chloride-1,4-dioxane solution (about 4 mol/L) (10 mL) was added and stirred at room temperature for 4 hours. The solvent was distilled off under reduced pressure to obtain the title compound (1 g).
1H -NMR (DMSO- d6 ) δ 13.27 (br.s, 1H), 12.80 (br.s, 1H), 8.16 (br.s, 1H), 8.00 (s, 1H), 7.76 (br.s, 2H), 7.68 (dd, J = 1.6, 2.8 Hz, 1H), 6.68 (s, 1H), 3.22 - 3.13 (m, 1H), 2.33 (s, 3H), 1.13 - 1.01 (m, 2H), 0.83 - 0.76 (m, 2H); LC-MS(m/z): 357.09 [M+H] + .

実施例1
5-(4-アセチル-1H-ピロール-2-イル)-2-{シクロプロピル[1-(3-フルオロピリジン-4-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000039
 5-(4-アセチル-1H-ピロール-2-イル)-2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ]チアゾール-4-カルボキサミド(参考例10)(1.85g,5.191mmol)と3-フルオロ-4-ヨードピリジン(1.73g,7.79mmol)の1,4-ジオキサン(40mL)溶液に、炭酸カリウム(2.15g,15.58mmol)とヨウ化銅(I)(0.197g,1.02mmol)と(1R,2R)-N1,N2-ジメチルシクロヘキサン-1,2-ジアミン(0.295g,2.08mmol)を加え、混合物を窒素気流下、130℃で一晩攪拌した。反応混合物を室温まで冷却後、セライトを用いて固形物をろ過し、ろ液を減圧濃縮した。残渣をカラムクロマトグラフィー(シリカゲル、クロロホルム/メタノール)で精製し、標記化合物(0.7g)を得た。
1H-NMR (DMSO-d6) δ13.27 (s, 1H), 8.79 (d, J = 3.7 Hz, 1H), 8.77 (d, J = 2.4 Hz, 1H), 8.54 (d, J = 5.3 Hz, 1H), 8.36 (s, 1H), 8.07 (s, 1H), 7.96 (dd, J = 7.0, 5.3 Hz, 1H), 7.87 (s, 1H), 7.72 (dd, J = 3.1, 1.6 Hz, 1H), 6.77 (dd, J = 2.3, 1.6 Hz, 1H), 2.57 (p, J = 1.9 Hz, 1H), 2.35 (s, 3H), 1.26 - 1.17 (m, 2H), 0.97 - 0.90 (m, 2H); LC-MS (m/z): 452.2 [M+H]+. Example 1
Preparation of 5-(4-acetyl-1H-pyrrol-2-yl)-2-{cyclopropyl[1-(3-fluoropyridin-4-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000039
To a solution of 5-(4-acetyl-1H-pyrrol-2-yl)-2-[cyclopropyl(1H-pyrazol-4-yl)amino]thiazole-4-carboxamide (Reference Example 10) (1.85 g, 5.191 mmol) and 3-fluoro-4-iodopyridine (1.73 g, 7.79 mmol) in 1,4-dioxane (40 mL), potassium carbonate (2.15 g, 15.58 mmol), copper (I) iodide (0.197 g, 1.02 mmol), and (1R, 2R)-N1,N2-dimethylcyclohexane-1,2-diamine (0.295 g, 2.08 mmol) were added, and the mixture was stirred overnight under a nitrogen stream at 130° C. After cooling the reaction mixture to room temperature, the solid matter was filtered using Celite, and the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, chloroform/methanol) to obtain the title compound (0.7 g).
1H -NMR (DMSO-d6) δ13.27 (s, 1H), 8.79 (d, J = 3.7 Hz, 1H), 8.77 (d, J = 2.4 Hz, 1H), 8.54 (d, J = 5.3 Hz, 1H), 8.36 (s, 1H), 8.07 (s, 1H), 7.96 (dd, J = 7.0, 5.3 Hz, 1H), 7.87 (s, 1H), 7.72 (dd, J = 3.1, 1.6 Hz, 1H), 6.77 (dd, J = 2.3, 1.6 Hz, 1H), 2.57 (p, J = 1.9 Hz, 1H), 2.35 (s, 3H), 1.26 - 1.17 (m, 2H), 0.97 - 0.90 (m, 2H); LC-MS (m/z): 452.2 [M+H] + .

実施例2
2-{シクロプロピル[1-(4-フルオロフェニル)-1H-ピラゾール-4-イル]アミノ}-5-[4-(ジメチルカルバモイル)-1H-ピロール-2-イル]チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000040
 2-(シクロプロピル[1H-ピラゾール-4-イル]アミノ)-5-(4-[ジメチルカルバモイル]-1H-ピロール-2-イル)チアゾール-4-カルボキサミド(参考例7)(0.08g,0.21mol)のジクロロメタン溶液(4mL)に(4-フルオロフェニルボロン酸(58mg,0.42mol)、ピリジン(164mg,2.08mol)及び酢酸銅(II)(75mg,0.42mol)を加え、混合液を酸素雰囲気下、室温で一晩攪拌した。溶媒を減圧留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、クロロホルム/メタノール)で精製し、標記化合物(23mg)を得た。
1H-NMR (DMSO-d6) δ 13.08 (s, 1H), 8.80 (s, 1H), 8.12 (s, 1H), 7.97 (s, 1H), 7.95 - 7.88 (m, 3H), 7.42 - 7.32 (m, 3H), 6.62 (s, 1H), 3.20 (s, 1H), 3.06 (s, 6H), 1.21 - 1.14 (m, 2H), 0.92 - 0.89 (m, 2H); LC-MS (m/z): 480.3 [M+H]+. Example 2
Preparation of 2-{cyclopropyl[1-(4-fluorophenyl)-1H-pyrazol-4-yl]amino}-5-[4-(dimethylcarbamoyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000040
4-Fluorophenylboronic acid (58 mg, 0.42 mol), pyridine (164 mg, 2.08 mol) and copper (II) acetate (75 mg, 0.42 mol) were added to a dichloromethane solution (4 mL) of 2-(cyclopropyl[1H-pyrazol-4-yl]amino)-5-(4-[dimethylcarbamoyl]-1H-pyrrol-2-yl)thiazole-4-carboxamide (Reference Example 7) (0.08 g, 0.21 mol), and the mixture was stirred overnight at room temperature under an oxygen atmosphere. The solvent was distilled off under reduced pressure, and the resulting residue was purified by column chromatography (silica gel, chloroform/methanol) to obtain the title compound (23 mg).
1H -NMR (DMSO-d6) δ 13.08 (s, 1H), 8.80 (s, 1H), 8.12 (s, 1H), 7.97 (s, 1H), 7.95 - 7.88 (m, 3H), 7.42 - 7.32 (m, 3H), 6.62 (s, 1H), 3.20 (s, 1H), 3.06 (s, 6H), 1.21 - 1.14 (m, 2H), 0.92 - 0.89 (m, 2H); LC-MS (m/z): 480.3 [M+H] + .

実施例3
2-{シクロプロピル[1-(4-フルオロ-2-メトキシフェニル)-1H-ピラゾール-4-イル]アミノ}-5-{4-[4-(オキセタン-3-イル)ピペラジン-1-カルボニル]-1H-ピロール-2-イル}チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000041
(第1工程)
5-(4-カルバモイル-2-{シクロプロピル[1-(4-フルオロ-2-メトキシフェニル)-1H-ピラゾール-4-イル]アミノ}チアゾール-5-イル)-1H-ピロール-3-カルボン酸メチル
Figure JPOXMLDOC01-appb-C000042
 5-{4-カルバモイル-2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ]チアゾール-5-イル}-1H-ピロール-3-カルボン酸メチル(参考例5)(1g,2.69mol)のジクロロメタン溶液(10mL)に4-フルオロ-2-メトキシフェニルボロン酸(0.9g,5.38mol)、ピリジン(6.4g,80.64mol)及び酢酸銅(II)(0.243g,1.34mol)を加え、混合液を酸素雰囲気下、室温で一晩攪拌した。反応混合物に水を加え、酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥後、溶媒を減圧留去し、標記化合物(0.8g)を得た。
1H-NMR (DMSO-d6) δ 13.25 (br.s, 1H), 8.54 (s, 1H), 8.08 - 8.03 (m, 2H), 7.77 (br.s, 1H), 7.64 - 7.61 (m, 2H), 7.20 (dd, J = 2.5, 11.0 Hz, 1H), 6.96 - 6.89 (m, 1H), 6.70 (t, J = 1.9 Hz, 1H), 3.87 (s, 3H), 3.72 (s, 3H), 3.22 - 3.17 (m, 1H), 1.20 - 1.12 (m, 2H), 0.91 - 0.87 (m, 2H); LC-MS (M/Z): 497.4 [M+H]+. Example 3
Preparation of 2-{cyclopropyl[1-(4-fluoro-2-methoxyphenyl)-1H-pyrazol-4-yl]amino}-5-{4-[4-(oxetan-3-yl)piperazine-1-carbonyl]-1H-pyrrol-2-yl}thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000041
(First step)
5-(4-carbamoyl-2-{cyclopropyl[1-(4-fluoro-2-methoxyphenyl)-1H-pyrazol-4-yl]amino}thiazol-5-yl)-1H-pyrrole-3-carboxylate methyl
Figure JPOXMLDOC01-appb-C000042
4-Fluoro-2-methoxyphenylboronic acid (0.9 g, 5.38 mol), pyridine (6.4 g, 80.64 mol) and copper (II) acetate (0.243 g, 1.34 mol) were added to a dichloromethane solution (10 mL) of 5-{4-carbamoyl-2-[cyclopropyl(1H-pyrazol-4-yl)amino]thiazol-5-yl}-1H-pyrrole-3-methyl carboxylate (Reference Example 5) (1 g, 2.69 mol), and the mixture was stirred overnight at room temperature under an oxygen atmosphere. Water was added to the reaction mixture, and the mixture was extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to obtain the title compound (0.8 g).
1H -NMR (DMSO- d6 ) δ 13.25 (br.s, 1H), 8.54 (s, 1H), 8.08 - 8.03 (m, 2H), 7.77 (br.s, 1H), 7.64 - 7.61 (m, 2H), 7.20 (dd, J = 2.5, 11.0 Hz, 1H), 6.96 - 6.89 (m, 1H), 6.70 (t, J = 1.9 Hz, 1H), 3.87 (s, 3H), 3.72 (s, 3H), 3.22 - 3.17 (m, 1H), 1.20 - 1.12 (m, 2H), 0.91 - 0.87 (m, 2H); LC-MS (M/Z): 497.4 [M+H] + .

(第2工程)
5-(4-カルバモイル-2-{シクロプロピル[1-(4-フルオロ-2-メトキシフェニル)-1H-ピラゾール-4-イル]アミノ}チアゾール-5-イル)-1H-ピロール-3-カルボン酸

Figure JPOXMLDOC01-appb-C000043
 5-(4-カルバモイル-2-(シクロプロピル(1-(4-フルオロ-2-メトキシフェニル)-1H-ピラゾール-4-イル)アミノ)チアゾール-5-イル)-1H-ピロール-3-カルボン酸メチル(0.8g,1.61mol)の1,4-ジオキサン/メタノールの混合液(6mL/6mL)に、3M水酸化ナトリウム水溶液(3mL)を加え70℃で5時間攪拌した。溶媒を減圧留去して得られた残渣に10%クエン酸水溶液を加え、析出した固体をろ取し、標記化合物(0.6g)を得た。
1H-NMR (DMSO-d6) δ 13.17 (br.s, 1H), 11.98 (br.s, 1H), 8.54 (s, 1H), 8.08 - 8.03 (m, 2H), 7.77 (br.s, 1H), 7.62 (t, J = 7.2 Hz, 1H), 7.53 (br.s, 1H), 7.22 - 7.19 (m, 1H), 6.96 - 6.89 (m, 1H), 6.65 (s, 1H), 3.87 (s, 3H), 3.22 - 3.17 (m, 1H), 1.19 - 1.11 (m, 2H), 0.92 - 0.84 (m, 2H); LC-MS (M/Z): 483.2 [M+H]+. (Second step)
5-(4-carbamoyl-2-{cyclopropyl[1-(4-fluoro-2-methoxyphenyl)-1H-pyrazol-4-yl]amino}thiazol-5-yl)-1H-pyrrole-3-carboxylic acid
Figure JPOXMLDOC01-appb-C000043
A 3M aqueous solution of sodium hydroxide (3 mL) was added to a mixture (6 mL/6 mL) of 5-(4-carbamoyl-2-(cyclopropyl(1-(4-fluoro-2-methoxyphenyl)-1H-pyrazol-4-yl)amino)thiazol-5-yl)-1H-pyrrole-3-methyl carboxylate (0.8 g, 1.61 mol) in 1,4-dioxane/methanol, and the mixture was stirred at 70° C. for 5 hours. The solvent was distilled off under reduced pressure, and a 10% aqueous solution of citric acid was added to the resulting residue. The precipitated solid was collected by filtration to obtain the title compound (0.6 g).
1H -NMR (DMSO- d6 ) δ 13.17 (br.s, 1H), 11.98 (br.s, 1H), 8.54 (s, 1H), 8.08 - 8.03 (m, 2H), 7.77 (br.s, 1H), 7.62 (t, J = 7.2 Hz, 1H), 7.53 (br.s, 1H), 7.22 - 7.19 (m, 1H), 6.96 - 6.89 (m, 1H), 6.65 (s, 1H), 3.87 (s, 3H), 3.22 - 3.17 (m, 1H), 1.19 - 1.11 (m, 2H), 0.92 - 0.84 (m, 2H); LC-MS (M/Z): 483.2 [M+H] + .

(第3工程)
2-{シクロプロピル[1-(4-フルオロ-2-メトキシフェニル)-1H-ピラゾール-4-イル]アミノ}-5-{4-[4-(オキセタン-3-イル)ピペラジン-1-カルボニル]-1H-ピロール-2-イル}チアゾール-4-カルボキサミドの製造
 5-(4-カルバモイル-2-(シクロプロピル(1-(4-フルオロ-2-メトキシフェニル)-1H-ピラゾール-4-イル)アミノ)チアゾール-5-イル)-1H-ピロール-3-カルボン酸(0.06g,0.12mol)のDMF溶液(3mL)に1-(オキセタン-3-イル)ピペラジン(0.035g,0.25mol)、HATU(0.094g,0.25mol)及びDIPEA(0.04mL,0.25mol)を加え、室温で16時間攪拌した。反応混合物を水で希釈し、酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥後、溶媒を減圧留去し、残渣をHPLC分取クロマトグラフィーシステムを用いて精製し標記化合物(21mg)を得た。
1H-NMR (500 MHz, DMSO-d6) δ 13.08 (br.s, 1H), 8.53 (s, 1H), 8.02 (br.s, 2H), 7.72 (br.s, 1H), 7.62 (dd, J = 6.4, 8.9 Hz, 1H), 7.29 (s, 1H), 7.19 (dd, J = 3.0, 11.0 Hz, 1H),  6.93 (dt, J = 2.6, 8.5 Hz, 1H), 6.53 (s, 1H), 4.54 (t, J = 6.6 Hz, 2H), 4.45 (t, J = 6.1 Hz, 2H), 3.87 (s, 3H), 3.65 - 3.62 (m, 4H), 3.45 - 3.40 (m, 1H), 3.21 - 3.17 (m, 1H), 2.30 - 2.24 (m, 4H), 1.17 - 1.11 (m, 2H), 0.91 - 0.85 (m, 2H); LC-MS (m/z): 607.4 [M+H]+.
(Third process)
Preparation of 2-{cyclopropyl[1-(4-fluoro-2-methoxyphenyl)-1H-pyrazol-4-yl]amino}-5-{4-[4-(oxetan-3-yl)piperazine-1-carbonyl]-1H-pyrrol-2-yl}thiazole-4-carboxamide 1-(oxetan-3-yl)piperazine (0.035 g, 0.25 mol), HATU (0.094 g, 0.25 mol) and DIPEA (0.04 mL, 0.25 mol) were added to a DMF solution (3 mL) of 5-(4-carbamoyl-2-(cyclopropyl(1-(4-fluoro-2-methoxyphenyl)-1H-pyrazol-4-yl)amino)thiazol-5-yl)-1H-pyrrole-3-carboxylic acid (0.06 g, 0.12 mol), and the mixture was stirred at room temperature for 16 hours. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the residue was purified using a HPLC preparative chromatography system to obtain the title compound (21 mg).
1H -NMR (500 MHz, DMSO-d6) δ 13.08 (br.s, 1H), 8.53 (s, 1H), 8.02 (br.s, 2H), 7.72 (br.s, 1H), 7.62 (dd, J = 6.4, 8.9 Hz, 1H), 7.29 (s, 1H), 7.19 (dd, J = 3.0, 11.0 Hz, 1H), 6.93 (dt, J = 2.6, 8.5 Hz, 1H), 6.53 (s, 1H), 4.54 (t, J = 6.6 Hz, 2H), 4.45 (t, J = 6.1 Hz, 2H), 3.87 (s, 3H), 3.65 - 3.62 (m, 4H), 3.45 - 3.40 (m, 1H), 3.21 - 3.17 (m, 1H), 2.30 - 2.24 (m, 4H), 1.17 - 1.11 (m, 2H), 0.91 - 0.85 (m, 2H); LC-MS (m/z): 607.4 [M+H] + .

実施例4
2-{シクロプロピル[1-(5-フルオロ-2-メトキシフェニル)-1H-ピラゾール-4-イル]アミノ}-5-[4-(モルホリン-4-カルボニル)-1H-ピロール-2-イル]チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000044
 2-(シクロプロピル(1H-ピラゾール-4-イル)アミノ)-5-(4-(モルホリン-4-カルボニル)-1H-ピロール-2-イル)チアゾール-4-カルボキサミド(参考例8)(0.04g,0.094mol)のジクロロメタン溶液(2mL)に5-フルオロ-2-メトキシフェニルボロン酸(32mg,0.187mol)、ピリジン(74mg,0.936mol)及び酢酸銅(II)(34mg,0.187mol)を加え、混合物を酸素雰囲気下、室温で一晩攪拌した。溶媒を減圧留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、クロロホルム/メタノール)で精製し、標記化合物(19.2mg)を得た。
1H-NMR (DMSO-d6) δ 13.25 - 13.00 (m, 1H), 8.68 (d, J = 0.8 Hz, 1H), 8.10 (s, 1H), 8.07 (d, J = 2.7 Hz, 1H), 7.72 (d, J = 2.8 Hz, 1H), 7.54 (dd, J = 9.5, 3.1 Hz, 1H), 7.35 - 7.31 (m, 1H), 7.31 - 7.20 (m, 2H), 6.58 (dd, J = 2.3, 1.6 Hz, 1H), 3.88 (s, 3H), 3.65 - 3.57 (m, 8H), 3.22 (tt, J = 6.8, 3.7 Hz, 1H), 1.19 - 1.14 (m, 2H), 0.93 - 0.87 (m, 2H); LC-MS (m/z): 552.3 [M+H]+. Example 4
Preparation of 2-{cyclopropyl[1-(5-fluoro-2-methoxyphenyl)-1H-pyrazol-4-yl]amino}-5-[4-(morpholine-4-carbonyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000044
5-Fluoro-2-methoxyphenylboronic acid (32 mg, 0.187 mol), pyridine (74 mg, 0.936 mol) and copper (II) acetate (34 mg, 0.187 mol) were added to a dichloromethane solution (2 mL) of 2-(cyclopropyl(1H-pyrazol-4-yl)amino)-5-(4-(morpholine-4-carbonyl)-1H-pyrrol-2-yl)thiazole-4-carboxamide (Reference Example 8) (0.04 g, 0.094 mol), and the mixture was stirred overnight at room temperature under an oxygen atmosphere. The residue obtained by evaporating the solvent under reduced pressure was purified by column chromatography (silica gel, chloroform/methanol) to obtain the title compound (19.2 mg).
1H -NMR (DMSO-d6) δ 13.25 - 13.00 (m, 1H), 8.68 (d, J = 0.8 Hz, 1H), 8.10 (s, 1H), 8.07 (d, J = 2.7 Hz, 1H), 7.72 (d, J = 2.8 Hz, 1H), 7.54 (dd, J = 9.5, 3.1 Hz, 1H), 7.35 - 7.31 (m, 1H), 7.31 - 7.20 (m, 2H), 6.58 (dd, J = 2.3, 1.6 Hz, 1H), 3.88 (s, 3H), 3.65 - 3.57 (m, 8H), 3.22 (tt, J = 6.8, 3.7 Hz, 1H), 1.19 - 1.14 (m, 2H), 0.93 - 0.87 (m, 2H); LC-MS (m/z): 552.3 [M+H] + .

実施例5
2-{シクロプロピル[1-(3,4-ジフルオロフェニル)-1H-ピラゾール-4-イル]アミノ}-5-(4-(ジメチルカルバモイル)-1H-ピロール-2-イル)チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000045
 2-(シクロプロピル(1H-ピラゾール-4-イル)アミノ)-5-(4-(ジメチルカルバモイル)-1H-ピロール-2-イル)チアゾール-4-カルボキサミド(参考例7)(0.05g,0.13mol)のジクロロメタン溶液(3mL)に、3,4-ジフルオロフェニルボロン酸(44mg,0.26mol)、ピリジン(103mg,1.3mol)及び酢酸銅(II)(47mg,0.26mol)を加え、混合液を酸素雰囲気下、室温で一晩攪拌した。溶媒を減圧留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、クロロホルム/メタノール)で精製し、標記化合物(9mg)を得た。
1H-NMR (DMSO-d6) δ 13.10 (s, 1H), 8.86 (d, J = 0.7 Hz, 1H), 8.17 (s, 1H), 8.04 - 8.00 (m, 2H), 7.98 (s, 1H), 7.66 - 7.58 (m, 2H), 7.36 (dd, J = 2.9, 1.6 Hz, 1H), 6.63 (dd, J = 2.4, 1.6 Hz, 1H), 3.24 - 3.16 (m, 1H), 3.06 (m, 6H), 1.22 - 1.12 (m, 2H), 0.95 - 0.84 (m, 2H); LC-MS (m/z): 498.2 [M+H]+. Example 5
Preparation of 2-{cyclopropyl[1-(3,4-difluorophenyl)-1H-pyrazol-4-yl]amino}-5-(4-(dimethylcarbamoyl)-1H-pyrrol-2-yl)thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000045
3,4-Difluorophenylboronic acid (44 mg, 0.26 mol), pyridine (103 mg, 1.3 mol) and copper(II) acetate (47 mg, 0.26 mol) were added to a dichloromethane solution (3 mL) of 2-(cyclopropyl(1H-pyrazol-4-yl)amino)-5-(4-(dimethylcarbamoyl)-1H-pyrrol-2-yl)thiazole-4-carboxamide (Reference Example 7) (0.05 g, 0.13 mol), and the mixture was stirred overnight at room temperature under an oxygen atmosphere. The solvent was distilled off under reduced pressure and the resulting residue was purified by column chromatography (silica gel, chloroform/methanol) to obtain the title compound (9 mg).
1H -NMR (DMSO-d6) δ 13.10 (s, 1H), 8.86 (d, J = 0.7 Hz, 1H), 8.17 (s, 1H), 8.04 - 8.00 (m, 2H), 7.98 (s, 1H), 7.66 - 7.58 (m, 2H), 7.36 (dd, J = 2.9, 1.6 Hz, 1H), 6.63 (dd, J = 2.4, 1.6 Hz, 1H), 3.24 - 3.16 (m, 1H), 3.06 (m, 6H), 1.22 - 1.12 (m, 2H), 0.95 - 0.84 (m, 2H); LC-MS (m/z): 498.2 [M+H] + .

実施例6
2-{シクロプロピル[1-(4-フルオロ-3-メチルフェニル)-1H-ピラゾール-4-イル]アミノ}-5-[4-(ジメチルカルバモイル)-1H-ピロール-2-イル]チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000046
 2-(シクロプロピル(1H-ピラゾール-4-イル)アミノ)-5-(4-(ジメチルカルバモイル)-1H-ピロール-2-イル)チアゾール-4-カルボキサミド(参考例7)(0.05g,0.13mol)のジクロロメタン溶液(3mL)に、(4-フルオロ-3-メトキシフェニル)ボロン酸(44mg,0.26mol)、ピリジン(103mg,1.3mol)及び酢酸銅(II)(47mg,0.26mol)を加え、混合物を酸素雰囲気下、室温で一晩攪拌した。溶媒を減圧留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、クロロホルム/メタノール)で精製し、標記化合物(10.5mg)を得た。
1H-NMR (DMSO-d6) δ 13.08 (s, 1H), 8.77 (d, J = 0.7 Hz, 1H), 7.99 (s, 1H), 7.92 (s, 1H), 7.84 - 7.79 (m, 1H), 7.71 (m, 2H), 7.35 (dd, J = 2.9, 1.6 Hz, 1H), 7.29 (t, J = 9.1 Hz, 1H), 6.62 (dd, J = 2.3, 1.6 Hz, 1H), 3.23 - 3.16 (m, 1H), 2.97 (m, 6H), 2.32 (s, 3H), 1.20 - 1.13 (m, 2H), 0.90 - 0.87 (m, 2H); LC-MS (m/z): 494.1 [M+H]+. Example 6
Preparation of 2-{cyclopropyl[1-(4-fluoro-3-methylphenyl)-1H-pyrazol-4-yl]amino}-5-[4-(dimethylcarbamoyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000046
To a dichloromethane solution (3 mL) of 2-(cyclopropyl(1H-pyrazol-4-yl)amino)-5-(4-(dimethylcarbamoyl)-1H-pyrrol-2-yl)thiazole-4-carboxamide (Reference Example 7) (0.05 g, 0.13 mol), (4-fluoro-3-methoxyphenyl)boronic acid (44 mg, 0.26 mol), pyridine (103 mg, 1.3 mol) and copper(II) acetate (47 mg, 0.26 mol) were added, and the mixture was stirred overnight at room temperature under an oxygen atmosphere. The residue obtained by distilling off the solvent under reduced pressure was purified by column chromatography (silica gel, chloroform/methanol) to obtain the title compound (10.5 mg).
1H -NMR (DMSO-d6) δ 13.08 (s, 1H), 8.77 (d, J = 0.7 Hz, 1H), 7.99 (s, 1H), 7.92 (s, 1H), 7.84 - 7.79 (m, 1H), 7.71 (m, 2H), 7.35 (dd, J = 2.9, 1.6 Hz, 1H), 7.29 (t, J = 9.1 Hz, 1H), 6.62 (dd, J = 2.3, 1.6 Hz, 1H), 3.23 - 3.16 (m, 1H), 2.97 (m, 6H), 2.32 (s, 3H), 1.20 - 1.13 (m, 2H), 0.90 - 0.87 (m, 2H); LC-MS (m/z): 494.1 [M+H] + .

実施例7
2-{シクロプロピル[1-(5-フルオロ-2-メトキシフェニル)-1H-ピラゾール-4-イル]アミノ}-5-[4-(ジメチルカルバモイル)-1H-ピロール-2-イル]チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000047
 2-(シクロプロピル[1H-ピラゾール-4-イル]アミノ)-5-(4-[ジメチルカルバモイル]-1H-ピロール-2-イル)チアゾール-4-カルボキサミド(参考例7)(0.05g,0.13mol)のジクロロメタン溶液(3mL)に5-フルオロ-2-メトキシフェニルボロン酸(44mg,0.26mol)、ピリジン(103mg,1.3mol)及び酢酸銅(II)(47mg,0.26mol)を加え、混合液を酸素雰囲気下、室温で一晩攪拌した。溶媒を減圧留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、クロロホルム/メタノール)で精製し、標記化合物(7.6mg)を得た。
1H-NMR (DMSO-d6) δ 13.07 (s, 1H), 8.68 (d, J = 0.7 Hz, 1H), 8.11 (d, J = 0.7 Hz, 1H), 8.08 (s, 1H), 7.74 (s, 1H), 7.54 (dd, J = 9.4, 3.1 Hz, 1H), 7.36 (dd, J = 2.9, 1.6 Hz, 1H), 7.29 (d, J = 5.0 Hz, 1H), 7.25 - 7.21 (m, 1H), 6.62 (dd, J = 2.4, 1.6 Hz, 1H), 3.88 (s, 3H), 3.24 - 3.16 (m, 1H), 2.99 (d, J = 12.0 Hz, 6H), 1.19 - 1.15 (m, 2H), 0.93 - 0.88 (m, 2H); LC-MS (m/z): 510.1 [M+H]+. Example 7
Preparation of 2-{cyclopropyl[1-(5-fluoro-2-methoxyphenyl)-1H-pyrazol-4-yl]amino}-5-[4-(dimethylcarbamoyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000047
5-Fluoro-2-methoxyphenylboronic acid (44 mg, 0.26 mol), pyridine (103 mg, 1.3 mol) and copper (II) acetate (47 mg, 0.26 mol) were added to a dichloromethane solution (3 mL) of 2-(cyclopropyl[1H-pyrazol-4-yl]amino)-5-(4-[dimethylcarbamoyl]-1H-pyrrol-2-yl)thiazole-4-carboxamide (Reference Example 7) (0.05 g, 0.13 mol), and the mixture was stirred overnight at room temperature under an oxygen atmosphere. The residue obtained by distilling off the solvent under reduced pressure was purified by column chromatography (silica gel, chloroform/methanol) to obtain the title compound (7.6 mg).
1H -NMR (DMSO-d6) δ 13.07 (s, 1H), 8.68 (d, J = 0.7 Hz, 1H), 8.11 (d, J = 0.7 Hz, 1H), 8.08 (s, 1H), 7.74 (s, 1H), 7.54 (dd, J = 9.4, 3.1 Hz, 1H), 7.36 (dd, J = 2.9, 1.6 Hz, 1H), 7.29 (d, J = 5.0 Hz, 1H), 7.25 - 7.21 (m, 1H), 6.62 (dd, J = 2.4, 1.6 Hz, 1H), 3.88 (s, 3H), 3.24 - 3.16 (m, 1H), 2.99 (d, J = 12.0 Hz, 6H), 1.19 - 1.15 (m, 2H), 0.93 - 0.88 (m, 2H); LC-MS (m/z): 510.1 [M+H] + .

実施例8
2-{シクロプロピル[1-(2,4-ジメトキシフェニル)-1H-ピラゾール-4-イル]アミノ}-5-{4-[4-(オキセタン-3-イル)ピペラジン-1-カルボニル]-1H-ピロール-2-イル}チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000048
(第1工程)
5-(4-カルバモイル-2-{シクロプロピル[1-(2,4-ジメトキシフェニル)-1H-ピラゾール-4-イル]アミノ}チアゾール-5-イル)-1H-ピロール-3-カルボン酸メチル
Figure JPOXMLDOC01-appb-C000049
 5-{4-カルバモイル-2-[シクロプロピル(1H-ピラゾール-4-イル)アミノ]チアゾール-5-イル}-1H-ピロール-3-カルボン酸メチル(参考例5)(2g,5.38mol)のジクロロメタン溶液(60mL)に、2,4-ジメトキシフェニルボロン酸(1.95g,10.75mol)、ピリジン(13mL)及び酢酸銅(II)(0.195g,1.07mol)を加え、混合物を酸素雰囲気下、室温で一晩攪拌した。溶媒を減圧留去して得られた残渣をカラムクロマトグラフィー(シリカゲル、クロロホルム/メタノール)で精製し、標記化合物(0.8g)を得た。
1H NMR (400 MHz, DMSO-d6) δ 13.17 (br. s, 1H), 8.43 (s, 1H), 8.05 (s, 1H), 7.97 (m, 1H), 7.73 (s, 1H), 7.56 - 7.46 (m, 2H), 6.78 (d, J = 2.5 Hz, 1H), 6.68 - 6.62 (m, 2H), 3.84 (s, 6H), 3.72 (s, 3H), 3.23 - 3.14 (m, 1H), 1.16 - 1.13 (m, 2H), 0.92 - 0.85 (m, 2H).LC-MS (M/Z): 509.3 [M+H]+. Example 8
Preparation of 2-{cyclopropyl[1-(2,4-dimethoxyphenyl)-1H-pyrazol-4-yl]amino}-5-{4-[4-(oxetan-3-yl)piperazine-1-carbonyl]-1H-pyrrol-2-yl}thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000048
(First step)
5-(4-carbamoyl-2-{cyclopropyl[1-(2,4-dimethoxyphenyl)-1H-pyrazol-4-yl]amino}thiazol-5-yl)-1H-pyrrole-3-carboxylate methyl
Figure JPOXMLDOC01-appb-C000049
2,4-Dimethoxyphenylboronic acid (1.95 g, 10.75 mol), pyridine (13 mL) and copper(II) acetate (0.195 g, 1.07 mol) were added to a dichloromethane solution (60 mL) of 5-{4-carbamoyl-2-[cyclopropyl(1H-pyrazol-4-yl)amino]thiazol-5-yl}-1H-pyrrole-3-methyl carboxylate (Reference Example 5) (2 g, 5.38 mol), and the mixture was stirred overnight at room temperature under an oxygen atmosphere. The residue obtained by distilling off the solvent under reduced pressure was purified by column chromatography (silica gel, chloroform/methanol) to obtain the title compound (0.8 g).
1H NMR (400 MHz, DMSO-d6) δ 13.17 (br. s, 1H), 8.43 (s, 1H), 8.05 (s, 1H), 7.97 (m, 1H), 7.73 (s, 1H), 7.56 - 7.46 (m, 2H), 6.78 (d, J = 2.5 Hz, 1H), 6.68 - 6.62 (m, 2H), 3.84 (s, 6H), 3.72 (s, 3H), 3.23 - 3.14 (m, 1H), 1.16 - 1.13 (m, 2H), 0.92 - 0.85 (m, 2H).LC-MS (M/Z): 509.3 [M+H] + .

(第2工程)
5-(4-カルバモイル-2-{シクロプロピル[1-(2,4-ジメトキシフェニル)-1H-ピラゾール-4-イル]アミノ}チアゾール-5-イル)-1H-ピロール-3-カルボン酸

Figure JPOXMLDOC01-appb-C000050
 5-(4-カルバモイル-2-(シクロプロピル(1-(2,4-ジメトキシフェニル)-1H-ピラゾール-4-イル)アミノ)チアゾール-5-イル)-1H-ピロール-3-カルボン酸メチル(0.8g,1.57mol)の1,4-ジオキサン/メタノールの混合液(40mL/40mL)に、3M水酸化ナトリウム水溶液(1.6mL,4.72mmol)を加え80℃で16時間攪拌した。溶媒を減圧留去して得られた残渣に10%クエン酸水溶液を加え、析出した固体をろ取し、標記化合物(0.7g)を得た。
1H-NMR (400 MHz, DMSO-d6) δ 13.17 (br.s, 1H), 11.99 (br.s, 1H), 8.43 (s, 1H), 8.05 (s, 1H), 7.97 (m, 1H), 7.73 (s, 1H), 7.56 - 7.46 (m, 2H), 6.78 (d, J = 2.5 Hz, 1H), 6.68 - 6.62 (m, 2H), 3.84 (s, 6H), 3.23 - 3.14 (m, 1H), 1.16 - 1.13 (m, 2H), 0.92 - 0.85 (m, 2H); LC-MS (M/Z): 495.2 [M+H]+. (Second step)
5-(4-carbamoyl-2-{cyclopropyl[1-(2,4-dimethoxyphenyl)-1H-pyrazol-4-yl]amino}thiazol-5-yl)-1H-pyrrole-3-carboxylic acid
Figure JPOXMLDOC01-appb-C000050
A 3M aqueous solution of sodium hydroxide (1.6 mL, 4.72 mmol) was added to a mixture (40 mL/40 mL) of 5-(4-carbamoyl-2-(cyclopropyl(1-(2,4-dimethoxyphenyl)-1H-pyrazol-4-yl)amino)thiazol-5-yl)-1H-pyrrole-3-methyl carboxylate (0.8 g, 1.57 mol) in 1,4-dioxane/methanol, and the mixture was stirred at 80° C. for 16 hours. The solvent was distilled off under reduced pressure, and a 10% aqueous solution of citric acid was added to the resulting residue. The precipitated solid was collected by filtration to obtain the title compound (0.7 g).
1H -NMR (400 MHz, DMSO-d6) δ 13.17 (br.s, 1H), 11.99 (br.s, 1H), 8.43 (s, 1H), 8.05 (s, 1H), 7.97 (m, 1H), 7.73 (s, 1H), 7.56 - 7.46 (m, 2H), 6.78 (d, J = 2.5 Hz, 1H), 6.68 - 6.62 (m, 2H), 3.84 (s, 6H), 3.23 - 3.14 (m, 1H), 1.16 - 1.13 (m, 2H), 0.92 - 0.85 (m, 2H); LC-MS (M/Z): 495.2 [M+H] + .

(第3工程)
2-(シクロプロピル(1-(2,4-ジメトキシフェニル)-1H-ピラゾール-4-イル)アミノ)-5-(4-(4-(オキセタン-3-イル)ピペラジン-1-カルボニル)-1H-ピロール-2-イル)チアゾール-4-カルボキサミド
 5-(4-カルバモイル-2-(シクロプロピル(1-(2,4-ジメトキシフェニル)-1H-ピラゾール-4-イル)アミノ)チアゾール-5-イル)-1H-ピロール-3-カルボン酸(0.07g,0.14mol)のTHF溶液(7mL)に1-(オキセタン-3-イル)ピペラジン(0.04g,0.28mol)、HATU(0.108g,0.28mol)及びDIPEA(0.049mL,0.28mol)を加え、室温で16時間攪拌した。反応混合物を水で希釈し、酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥後、溶媒を減圧留去し、残渣をHPLC分取クロマトグラフィーシステムを用いて精製し標記化合物(21.3mg)を得た。
1H-NMR (DMSO-d6) δ 13.08 (br.s, 1H), 8.43 (s, 1H), 8.02 (s, 1H), 7.97 (s, 1H), 7.71 (br.s, 1H), 7.49 (d, J = 8.4 Hz, 1H), 7.29 (br.s, 1H), 6.78 (br.s, 1H), 6.65 (d, J = 7.6 Hz, 1H), 6.52 (br.s, 1H), 4.54 (t, J = 6.0 Hz, 2H), 4.45 (t, J = 5.6 Hz, 2H), 3.83 (d, J = 5.1 Hz, 6H), 3.71 - 3.60 (m, 4H), 3.44 - 3.39 (m, 1H), 3.21 - 3.13 (m, 1H), 2.33 - 2.27 (m, 4H), 1.16 - 1.09 (m, 2H), 0.88 - 0.84 (m, 2H); LC-MS (m/z): 619.4 [M+H]+.
(Third process)
2-(cyclopropyl(1-(2,4-dimethoxyphenyl)-1H-pyrazol-4-yl)amino)-5-(4-(4-(oxetan-3-yl)piperazine-1-carbonyl)-1H-pyrrol-2-yl)thiazole-4-carboxamide 5-(4-carbamoyl-2-(cyclopropyl(1-(2,4-dimethoxyphenyl)-1H-pyrazol-4-yl)amino)thiazole-5-yl)-1H-pyrrole-3-carboxylic acid (0.07 g, 0.14 mol) in THF solution (7 mL) was added with 1-(oxetan-3-yl)piperazine (0.04 g, 0.28 mol), HATU (0.108 g, 0.28 mol) and DIPEA (0.049 mL, 0.28 mol), and the mixture was stirred at room temperature for 16 hours. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the residue was purified using a HPLC preparative chromatography system to obtain the title compound (21.3 mg).
1H -NMR (DMSO-d6) δ 13.08 (br.s, 1H), 8.43 (s, 1H), 8.02 (s, 1H), 7.97 (s, 1H), 7.71 (br.s, 1H), 7.49 (d, J = 8.4 Hz, 1H), 7.29 (br.s, 1H), 6.78 (br.s, 1H), 6.65 (d, J = 7.6 Hz, 1H), 6.52 (br.s, 1H), 4.54 (t, J = 6.0 Hz, 2H), 4.45 (t, J = 5.6 Hz, 2H), 3.83 (d, J = 5.1 Hz, 6H), 3.71 - 3.60 (m, 4H), 3.44 - 3.39 (m, 1H), 3.21 - 3.13 (m, 1H), 2.33 - 2.27 (m, 4H), 1.16 - 1.09 (m, 2H), 0.88 - 0.84 (m, 2H); LC-MS (m/z): 619.4 [M+H] + .

実施例9~183
以下の実施例化合物[表1]は、それぞれ対応する原料(市販品、または市販化合物から公知の方法もしくはそれに準じた方法により誘導体化した化合物)を用い、上述の実施例記載の方法に従い、必要に応じて、有機合成化学で通常用いられる方法を適宜組み合わせて製造した。また、各々の化合物の物理化学データを[表2]に示した。
Examples 9 to 183
The following example compounds [Table 1] were produced using the corresponding raw materials (commercially available products, or compounds derived from commercially available compounds by known methods or methods similar thereto) according to the methods described in the above examples, and, if necessary, by appropriately combining methods commonly used in organic synthetic chemistry. The physicochemical data of each compound is shown in [Table 2].

[表1]

Figure JPOXMLDOC01-appb-T000051
Figure JPOXMLDOC01-appb-T000052
Figure JPOXMLDOC01-appb-T000053
Figure JPOXMLDOC01-appb-T000054
Figure JPOXMLDOC01-appb-T000055
Figure JPOXMLDOC01-appb-T000056
Figure JPOXMLDOC01-appb-T000057
Figure JPOXMLDOC01-appb-T000058
Figure JPOXMLDOC01-appb-T000059
Figure JPOXMLDOC01-appb-T000060
Figure JPOXMLDOC01-appb-T000061
Figure JPOXMLDOC01-appb-T000062
Figure JPOXMLDOC01-appb-T000063
Figure JPOXMLDOC01-appb-T000064
Figure JPOXMLDOC01-appb-T000065
Figure JPOXMLDOC01-appb-T000066
Figure JPOXMLDOC01-appb-T000067
Figure JPOXMLDOC01-appb-T000068
Figure JPOXMLDOC01-appb-T000069
Figure JPOXMLDOC01-appb-T000070
Figure JPOXMLDOC01-appb-T000071
Figure JPOXMLDOC01-appb-T000072
Figure JPOXMLDOC01-appb-T000073
Figure JPOXMLDOC01-appb-T000074
Figure JPOXMLDOC01-appb-T000075
Figure JPOXMLDOC01-appb-T000076
Figure JPOXMLDOC01-appb-T000077
Figure JPOXMLDOC01-appb-T000078
Figure JPOXMLDOC01-appb-T000079
Figure JPOXMLDOC01-appb-T000080
[Table 1]
Figure JPOXMLDOC01-appb-T000051
Figure JPOXMLDOC01-appb-T000052
Figure JPOXMLDOC01-appb-T000053
Figure JPOXMLDOC01-appb-T000054
Figure JPOXMLDOC01-appb-T000055
Figure JPOXMLDOC01-appb-T000056
Figure JPOXMLDOC01-appb-T000057
Figure JPOXMLDOC01-appb-T000058
Figure JPOXMLDOC01-appb-T000059
Figure JPOXMLDOC01-appb-T000060
Figure JPOXMLDOC01-appb-T000061
Figure JPOXMLDOC01-appb-T000062
Figure JPOXMLDOC01-appb-T000063
Figure JPOXMLDOC01-appb-T000064
Figure JPOXMLDOC01-appb-T000065
Figure JPOXMLDOC01-appb-T000066
Figure JPOXMLDOC01-appb-T000067
Figure JPOXMLDOC01-appb-T000068
Figure JPOXMLDOC01-appb-T000069
Figure JPOXMLDOC01-appb-T000070
Figure JPOXMLDOC01-appb-T000071
Figure JPOXMLDOC01-appb-T000072
Figure JPOXMLDOC01-appb-T000073
Figure JPOXMLDOC01-appb-T000074
Figure JPOXMLDOC01-appb-T000075
Figure JPOXMLDOC01-appb-T000076
Figure JPOXMLDOC01-appb-T000077
Figure JPOXMLDOC01-appb-T000078
Figure JPOXMLDOC01-appb-T000079
Figure JPOXMLDOC01-appb-T000080

[表2]

Figure JPOXMLDOC01-appb-T000081
Figure JPOXMLDOC01-appb-T000082
Figure JPOXMLDOC01-appb-T000083
Figure JPOXMLDOC01-appb-T000084
Figure JPOXMLDOC01-appb-T000085
Figure JPOXMLDOC01-appb-T000086
Figure JPOXMLDOC01-appb-T000087
Figure JPOXMLDOC01-appb-T000088
Figure JPOXMLDOC01-appb-T000089
Figure JPOXMLDOC01-appb-T000090
Figure JPOXMLDOC01-appb-T000091
Figure JPOXMLDOC01-appb-T000092
Figure JPOXMLDOC01-appb-T000093
Figure JPOXMLDOC01-appb-T000094
Figure JPOXMLDOC01-appb-T000095
Figure JPOXMLDOC01-appb-T000096
Figure JPOXMLDOC01-appb-T000097
Figure JPOXMLDOC01-appb-T000098
Figure JPOXMLDOC01-appb-T000099
Figure JPOXMLDOC01-appb-T000100
Figure JPOXMLDOC01-appb-T000101
Figure JPOXMLDOC01-appb-T000102
Figure JPOXMLDOC01-appb-T000103
Figure JPOXMLDOC01-appb-T000104
Figure JPOXMLDOC01-appb-T000105
Figure JPOXMLDOC01-appb-T000106
Figure JPOXMLDOC01-appb-T000107
Figure JPOXMLDOC01-appb-T000108
Figure JPOXMLDOC01-appb-T000109
Figure JPOXMLDOC01-appb-T000110
Figure JPOXMLDOC01-appb-T000111
Figure JPOXMLDOC01-appb-T000112
Figure JPOXMLDOC01-appb-T000113
Figure JPOXMLDOC01-appb-T000114
Figure JPOXMLDOC01-appb-T000115
[Table 2]
Figure JPOXMLDOC01-appb-T000081
Figure JPOXMLDOC01-appb-T000082
Figure JPOXMLDOC01-appb-T000083
Figure JPOXMLDOC01-appb-T000084
Figure JPOXMLDOC01-appb-T000085
Figure JPOXMLDOC01-appb-T000086
Figure JPOXMLDOC01-appb-T000087
Figure JPOXMLDOC01-appb-T000088
Figure JPOXMLDOC01-appb-T000089
Figure JPOXMLDOC01-appb-T000090
Figure JPOXMLDOC01-appb-T000091
Figure JPOXMLDOC01-appb-T000092
Figure JPOXMLDOC01-appb-T000093
Figure JPOXMLDOC01-appb-T000094
Figure JPOXMLDOC01-appb-T000095
Figure JPOXMLDOC01-appb-T000096
Figure JPOXMLDOC01-appb-T000097
Figure JPOXMLDOC01-appb-T000098
Figure JPOXMLDOC01-appb-T000099
Figure JPOXMLDOC01-appb-T000100
Figure JPOXMLDOC01-appb-T000101
Figure JPOXMLDOC01-appb-T000102
Figure JPOXMLDOC01-appb-T000103
Figure JPOXMLDOC01-appb-T000104
Figure JPOXMLDOC01-appb-T000105
Figure JPOXMLDOC01-appb-T000106
Figure JPOXMLDOC01-appb-T000107
Figure JPOXMLDOC01-appb-T000108
Figure JPOXMLDOC01-appb-T000109
Figure JPOXMLDOC01-appb-T000110
Figure JPOXMLDOC01-appb-T000111
Figure JPOXMLDOC01-appb-T000112
Figure JPOXMLDOC01-appb-T000113
Figure JPOXMLDOC01-appb-T000114
Figure JPOXMLDOC01-appb-T000115

実施例184
2-{[1-(ブタ-2-イン-1-イル)-1H-ピラゾール-4-イル](シクロプロピルメチル)アミノ}-5-[4-(ジメチルカルバモイル)-1H-ピロール-2-イル]チアゾール-4-カルボキサミドの製造

Figure JPOXMLDOC01-appb-C000116
(第1工程)
2-{(シクロプロピルメチル)[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル
Figure JPOXMLDOC01-appb-C000117
 2-{[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル(参考例1第2工程の化合物)(1.5g,4.7mmol)の1,4-ジオキサン溶液(25mL)に(ブロモメチル)シクロプロパン(1.9g,14mmol)および炭酸カリウム(1.9g,14mmol)を室温で加え、混合物を100℃で16時間撹拌した。反応混合物を水で希釈し、酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥し、減圧下濃縮した。残渣をカラムクロマトグラフィー(シリカゲル、石油エーテル/酢酸エチル)で精製し、標記化合物(1.5g)を得た。
1H-NMR (500 MHz, Chloroform-d) δ 7.81 (d, J = 0.8 Hz, 1H), 7.59 (d, J = 0.8 Hz, 1H), 7.33 (s, 1H), 5.42 - 5.33 (m, 1H), 4.34 (q, J = 7.1 Hz, 2H), 4.10 - 4.02 (m, 2H), 3.82 - 3.72 (m, 3H), 2.11 (m, 2H), 2.07 - 2.00 (m, 1H), 1.71 (m, 2H), 1.37 (t, J = 7.1 Hz, 3H), 1.12 (m, 1H), 0.54 - 0.46 (m, 2H), 0.29 - 0.22 (m, 2H); LC-MS (m/z): 377.33 [M+H]+. Example 184
Preparation of 2-{[1-(but-2-yn-1-yl)-1H-pyrazol-4-yl](cyclopropylmethyl)amino}-5-[4-(dimethylcarbamoyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000116
(First step)
2-{(cyclopropylmethyl)[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylate ethyl
Figure JPOXMLDOC01-appb-C000117
To a solution (25 mL) of ethyl 2-{[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylate (compound from Step 2 of Reference Example 1) (1.5 g, 4.7 mmol) in 1,4-dioxane, (bromomethyl)cyclopropane (1.9 g, 14 mmol) and potassium carbonate (1.9 g, 14 mmol) were added at room temperature, and the mixture was stirred at 100° C. for 16 hours. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate) to give the title compound (1.5 g).
1H -NMR (500 MHz, Chloroform-d) δ 7.81 (d, J = 0.8 Hz, 1H), 7.59 (d, J = 0.8 Hz, 1H), 7.33 (s, 1H), 5.42 - 5.33 (m, 1H), 4.34 (q, J = 7.1 Hz, 2H), 4.10 - 4.02 (m, 2H), 3.82 - 3.72 (m, 3H), 2.11 (m, 2H), 2.07 - 2.00 (m, 1H), 1.71 (m, 2H), 1.37 (t, J = 7.1 Hz, 3H), 1.12 (m, 1H), 0.54 - 0.46 (m, 2H), 0.29 - 0.22 (m, 2H); LC-MS (m/z): 377.33 [M+H] + .

(第2工程)
5-ブロモ-2-{(シクロプロピルメチル)[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル

Figure JPOXMLDOC01-appb-C000118
 2-{(シクロプロピルメチル)[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル(1.5g,3.9mmol)のジクロロメタン溶液(20mL)に、0℃でNBS(637mg,3.6mmol)を加え、混合物を室温で3時間撹拌した。反応混合物に氷水を加え、酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥後、溶媒を減圧留去し、標記化合物(1.4g)を得た。
1H-NMR (Chloroform-d) δ 7.78 (s, 1H), 7.57 (s, 1H), 5.38 (dd, J = 8.5, 3.6 Hz, 1H), 4.37 (q, J = 7.1 Hz, 2H), 4.07 (d, J = 12.3 Hz, 1H), 3.81 - 3.61 (m, 3H), 2.17 - 1.98 (m, 3H), 1.68 (m, 3H), 1.40 (t, J = 7.1 Hz, 3H), 1.09 (m, 1H), 0.55 - 0.42 (m, 2H), 0.26 (t, J = 5.1 Hz, 2H).; LC-MS (m/z): 455.12 ([M+H]+. (Second step)
5-Bromo-2-{(cyclopropylmethyl)[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylate ethyl
Figure JPOXMLDOC01-appb-C000118
NBS (637 mg, 3.6 mmol) was added to a dichloromethane solution (20 mL) of ethyl 2-{(cyclopropylmethyl)[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylate (1.5 g, 3.9 mmol) at 0° C., and the mixture was stirred at room temperature for 3 hours. Ice water was added to the reaction mixture, and the mixture was extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure to obtain the title compound (1.4 g).
1H -NMR (Chloroform-d) δ 7.78 (s, 1H), 7.57 (s, 1H), 5.38 (dd, J = 8.5, 3.6 Hz, 1H), 4.37 (q, J = 7.1 Hz, 2H), 4.07 (d, J = 12.3 Hz, 1H), 3.81 - 3.61 (m, 3H), 2.17 - 1.98 (m, 3H), 1.68 (m, 3H), 1.40 (t, J = 7.1 Hz, 3H), 1.09 (m, 1H), 0.55 - 0.42 (m, 2H), 0.26 (t, J = 5.1 Hz, 2H).; LC-MS (m/z): 455.12 ([M+H] + .

(第3工程)
5-ブロモ-2-{(シクロプロピルメチル)[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸

Figure JPOXMLDOC01-appb-C000119
 5-ブロモ-2-{(シクロプロピルメチル)[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸エチル(1.4g,3.1mmol)の1,4-ジオキサン/メタノールの混合液(1:1,10mL)に、4M水酸化ナトリウム水溶液(5mL)を加え、室温で16時間攪拌した。反応混合物に塩酸を加えて中和した後、クロロホルムで抽出した。有機層を無水硫酸ナトリウムで乾燥後、溶媒を減圧留去し、標記化合物(1.2g)を得た。
LC-MS (m/z): 427.06 [M+H]+. (Third process)
5-Bromo-2-{(cyclopropylmethyl)[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylic acid
Figure JPOXMLDOC01-appb-C000119
A 4M aqueous sodium hydroxide solution (5 mL) was added to a mixture of 5-bromo-2-{(cyclopropylmethyl)[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-ethyl carboxylate (1.4 g, 3.1 mmol) in 1,4-dioxane/methanol (1:1, 10 mL), and the mixture was stirred at room temperature for 16 hours. The reaction mixture was neutralized with hydrochloric acid, and then extracted with chloroform. The organic layer was dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to obtain the title compound (1.2 g).
LC-MS (m/z): 427.06 [M+H] + .

(第4工程)
5-ブロモ-2-{(シクロプロピルメチル)[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボキサミド

Figure JPOXMLDOC01-appb-C000120
 5-ブロモ-2-{(シクロプロピルメチル)[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボン酸(1.2g,2.8mmol)のTHF溶液(10mL)に1,1’-カルボニルジイミダゾール(0.9g,5.6mmol)を加え、室温で2時間撹拌した後、アンモニア水(5mL)を加えて、さらに室温で1時間撹拌した。析出物をろ取し、乾燥させて標記化合物(1.0g)を得た。
LC-MS (m/z): 426.14 ([M+H]+. (Fourth step)
5-Bromo-2-{(cyclopropylmethyl)[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000120
1,1'-carbonyldiimidazole (0.9 g, 5.6 mmol) was added to a THF solution (10 mL) of 5-bromo-2-{(cyclopropylmethyl)[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxylic acid (1.2 g, 2.8 mmol), and the mixture was stirred at room temperature for 2 hours, and then aqueous ammonia (5 mL) was added and the mixture was further stirred at room temperature for 1 hour. The precipitate was collected by filtration and dried to obtain the title compound (1.0 g).
LC-MS (m/z): 426.14 ([M+H] + .

(第5工程)
5-[4-カルバモイル-2-{(シクロプロピルメチル)(1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル)アミノ}チアゾール-5-イル]-1H-ピロール-3-カルボン酸メチル

Figure JPOXMLDOC01-appb-C000121
 5-ブロモ-2-{(シクロプロピルメチル)[1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル]アミノ}チアゾール-4-カルボキサミド(1.0g,2.4mmol)と5-(4,4,5,5-テトラメチル-1,3,2-ジオキサボロラン-2-イル)-1H-ピロール-3-カルボン酸メチル(参考例3)(885mg,3.5mmol)の1,4-ジオキサン/水の混合液(15mL/1.5mL)に、テトラキス(トリフェニルホスフィン)パラジウム(0)(27.2mg,0.2mmol)と炭酸カリウム(975mg,7.1mmol)を加え、混合物を100℃で2時間攪拌した。反応混合物に氷水を加え、酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥後、溶媒を減圧留去した。残渣をカラムクロマトグラフィー(シリカゲル、酢酸エチル)で精製し、標記化合物(800mg)を得た。
1H-NMR (DMSO-d6) δ 13.27 (s, 1H), 8.24 (d, J = 0.8 Hz, 1H), 7.95 (d, J = 8.6 Hz, 2H), 7.73 (d, J = 0.8 Hz, 1H), 7.61 - 7.59 (m, 1H), 6.47 (dd, J = 2.3, 1.6 Hz, 1H), 5.42 (dd, J = 10.1, 2.1 Hz, 1H), 3.96 (d, J = 11.9 Hz, 1H), 3.92 (s, 1H), 3.76 (d, J = 7.1 Hz, 2H), 3.68 (m, 5H), 2.12 (dt, J = 11.9, 9.4 Hz, 1H), 1.98 - 1.91 (m, 2H), 1.59 - 1.48 (m, 2H), 0.50 - 0.40 (m, 2H), 0.30 - 0.21 (m, 2H); LC-MS (m/z): 471.26 [M+H]+. (Fifth step)
5-[4-carbamoyl-2-{(cyclopropylmethyl)(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)amino}thiazol-5-yl]-1H-pyrrole-3-carboxylate methyl
Figure JPOXMLDOC01-appb-C000121
Tetrakis(triphenylphosphine)palladium(0) (27.2 mg, 0.2 mmol) and potassium carbonate (975 mg, 7.1 mmol) were added to a mixture (15 mL/1.5 mL) of 5-bromo-2-{(cyclopropylmethyl)[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]amino}thiazole-4-carboxamide (1.0 g, 2.4 mmol) and 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrrole-3-methyl carboxylate (Reference Example 3) (885 mg, 3.5 mmol) in 1,4-dioxane/water, and the mixture was stirred at 100° C. for 2 hours. Ice water was added to the reaction mixture, and the mixture was extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure. The residue was purified by column chromatography (silica gel, ethyl acetate) to obtain the title compound (800 mg).
1H -NMR (DMSO-d6) δ 13.27 (s, 1H), 8.24 (d, J = 0.8 Hz, 1H), 7.95 (d, J = 8.6 Hz, 2H), 7.73 (d, J = 0.8 Hz, 1H), 7.61 - 7.59 (m, 1H), 6.47 (dd, J = 2.3, 1.6 Hz, 1H), 5.42 (dd, J = 10.1, 2.1 Hz, 1H), 3.96 (d, J = 11.9 Hz, 1H), 3.92 (s, 1H), 3.76 (d, J = 7.1 Hz, 2H), 3.68 (m, 5H), 2.12 (dt, J = 11.9, 9.4 Hz, 1H), 1.98 - 1.91 (m, 2H), 1.59 - 1.48 (m, 2H), 0.50 - 0.40 (m, 2H), 0.30 - 0.21 (m, 2H); LC-MS (m/z): 471.26 [M+H] + .

(第6工程)
5-{4-カルバモイル-2-[(シクロプロピルメチル)(1H-ピラゾール-4-イル)アミノ]チアゾール-5-イル}-1H-ピロール-3-カルボン酸

Figure JPOXMLDOC01-appb-C000122
 5-[4-カルバモイル-2-{(シクロプロピルメチル)(1-(テトラヒドロ-2H-ピラン-2-イル)-1H-ピラゾール-4-イル)アミノ}チアゾール-5-イル]-1H-ピロール-3-カルボン酸メチル(800mg,1.7mmol)の1,4-ジオキサン/メタノールの混合液(1:1,14mL)に4M水酸化ナトリウム水溶液(5mL)を加え、混合物を70℃で1時間攪拌した。反応混合物に濃塩酸を加えて酸性にし、析出した固体をろ取し、標記化合物(220mg)を得た。
1H-NMR (DMSO-d6) δ 13.17 (m, 2H), 7.90 (q, J = 2.8 Hz, 1H), 7.85 (s, 1H), 7.43 (dd, J = 3.0, 1.6 Hz, 1H), 7.29 (dt, J = 3.3, 1.8 Hz, 1H), 6.73 (q, J = 2.4 Hz, 1H), 6.36 (t, J = 1.9 Hz, 1H), 3.71 (d, J = 7.0 Hz, 2H), 1.05 (tt, J = 7.6, 5.2 Hz, 1H), 0.44 - 0.38 (m, 2H), 0.25 - 0.17 (m, 2H); LC-MS (m/z): 373.16 [M+H]+. (Sixth step)
5-{4-carbamoyl-2-[(cyclopropylmethyl)(1H-pyrazol-4-yl)amino]thiazol-5-yl}-1H-pyrrole-3-carboxylic acid
Figure JPOXMLDOC01-appb-C000122
A 4M aqueous sodium hydroxide solution (5 mL) was added to a mixture of 5-[4-carbamoyl-2-{(cyclopropylmethyl)(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)amino}thiazol-5-yl]-1H-pyrrole-3-methyl carboxylate (800 mg, 1.7 mmol) in 1,4-dioxane/methanol (1:1, 14 mL), and the mixture was stirred at 70° C. for 1 hour. The reaction mixture was acidified with concentrated hydrochloric acid, and the precipitated solid was collected by filtration to obtain the title compound (220 mg).
1H -NMR (DMSO-d6) δ 13.17 (m, 2H), 7.90 (q, J = 2.8 Hz, 1H), 7.85 (s, 1H), 7.43 (dd, J = 3.0, 1.6 Hz, 1H), 7.29 (dt, J = 3.3, 1.8 Hz, 1H), 6.73 (q, J = 2.4 Hz, 1H), 6.36 (t, J = 1.9 Hz, 1H), 3.71 (d, J = 7.0 Hz, 2H), 1.05 (tt, J = 7.6, 5.2 Hz, 1H), 0.44 - 0.38 (m, 2H), 0.25 - 0.17 (m, 2H); LC-MS (m/z): 373.16 [M+H] + .

(第7工程)
2-[(シクロプロピルメチル)(1H-ピラゾール-4-イル)アミノ]-5-[4-(ジメチルカルバモイル)-1H-ピロール-2-イル]チアゾール-4-カルボキサミド

Figure JPOXMLDOC01-appb-C000123
 5-{4-カルバモイル-2-[(シクロプロピルメチル)(1H-ピラゾール-4-イル)アミノ]チアゾール-5-イル}-1H-ピロール-3-カルボン酸(200mg,0.5mol)のDMF溶液(3mL)に、ジメチルアミン塩酸塩(203mg,1.1mol)及びEDC・HCl(204mg,1.1mol)を加え、室温で3時間攪拌した。反応混合物を水で希釈し、析出した固体をろ取し、標記化合物(170mg)を得た。
LC-MS (m/z): 400.22 [M+H]+. (Seventh step)
2-[(cyclopropylmethyl)(1H-pyrazol-4-yl)amino]-5-[4-(dimethylcarbamoyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide
Figure JPOXMLDOC01-appb-C000123
Dimethylamine hydrochloride (203 mg, 1.1 mol) and EDC.HCl (204 mg, 1.1 mol) were added to a DMF solution (3 mL) of 5-{4-carbamoyl-2-[(cyclopropylmethyl)(1H-pyrazol-4-yl)amino]thiazol-5-yl}-1H-pyrrole-3-carboxylic acid (200 mg, 0.5 mol), and the mixture was stirred at room temperature for 3 hours. The reaction mixture was diluted with water, and the precipitated solid was collected by filtration to obtain the title compound (170 mg).
LC-MS (m/z): 400.22 [M+H] + .

(第8工程)
2-[(1-(ブタ-2-イン-1-イル)-1H-ピラゾール-4-イル)(シクロプロピルメチル)アミノ)-5-[4-(ジメチルカルバモイル)-1H-ピロール-2-イル]チアゾール-4-カルボキサミドの製造
 2-[(シクロプロピルメチル)(1H-ピラゾール-4-イル)アミノ]-5-[4-(ジメチルカルバモイル)-1H-ピロール-2-イル]チアゾール-4-カルボキサミド(150mg,0.38mmol)と1-ブロモ-2-ブチン(103mg,0.75mmol)のDMF(3mL)溶液に、炭酸カリウム(103mg,0.75mmol)を加え、マイクロウェーブ合成装置を用いて120℃で1時間反応させた。反応混合物を室温まで冷却後、セライトを用いて固形物をろ過し、ろ液を減圧濃縮した。残渣をカラムクロマトグラフィー(シリカゲル、クロロホルム/メタノール)で精製し、標記化合物(30mg)を得た。
1H-NMR (DMSO-d6) δ 13.10 (s, 1H), 8.15 (d, J = 0.8 Hz, 1H), 7.99 - 7.84 (m, 2H), 7.69 (d, J = 0.8 Hz, 1H), 7.29 (dd, J = 2.8, 1.6 Hz, 1H), 6.42 - 6.39 (m, 1H), 5.00 (q, J = 2.5 Hz, 2H), 3.74 (d, J = 7.1 Hz, 2H), 2.97 (d, J = 34.5 Hz, 6H), 1.85 (t, J = 2.5 Hz, 3H), 1.15 - 1.04 (m, 1H), 0.48 - 0.44 (m, 2H), 0.27 (dd, J = 4.8, 1.8 Hz, 2H); LC-MS (m/z): 452.2 [M+H]+.
(Step 8)
Preparation of 2-[(1-(but-2-yn-1-yl)-1H-pyrazol-4-yl)(cyclopropylmethyl)amino)-5-[4-(dimethylcarbamoyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide Potassium carbonate (103 mg, 0.75 mmol) was added to a solution of 2-[(cyclopropylmethyl)(1H-pyrazol-4-yl)amino]-5-[4-(dimethylcarbamoyl)-1H-pyrrol-2-yl]thiazole-4-carboxamide (150 mg, 0.38 mmol) and 1-bromo-2-butyne (103 mg, 0.75 mmol) in DMF (3 mL), and the mixture was reacted at 120° C. for 1 hour using a microwave synthesis apparatus. After cooling the reaction mixture to room temperature, solids were filtered using Celite, and the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, chloroform/methanol) to obtain the title compound (30 mg).
1H -NMR (DMSO-d6) δ 13.10 (s, 1H), 8.15 (d, J = 0.8 Hz, 1H), 7.99 - 7.84 (m, 2H), 7.69 (d, J = 0.8 Hz, 1H), 7.29 (dd, J = 2.8, 1.6 Hz, 1H), 6.42 - 6.39 (m, 1H), 5.00 (q, J = 2.5 Hz, 2H), 3.74 (d, J = 7.1 Hz, 2H), 2.97 (d, J = 34.5 Hz, 6H), 1.85 (t, J = 2.5 Hz, 3H), 1.15 - 1.04 (m, 1H), 0.48 - 0.44 (m, 2H), 0.27 (dd, J = 4.8, 1.8 Hz, 2H); LC-MS (m/z): 452.2 [M+H] + .

実施例185~226
 以下の実施例化合物[表3]は、それぞれ対応する原料(市販品、または市販化合物から公知の方法もしくはそれに準じた方法により誘導体化した化合物)を用い、上述の実施例記載の方法に従い、必要に応じて、有機合成化学で通常用いられる方法を適宜組み合わせて製造した。また、各々の化合物の物理化学データを[表4]に示した。
Examples 185 to 226
The following example compounds [Table 3] were produced using the corresponding raw materials (commercially available products, or compounds derived from commercially available compounds by known methods or methods similar thereto) according to the methods described in the above examples, and, if necessary, by appropriately combining methods commonly used in organic synthetic chemistry. The physicochemical data of each compound is shown in [Table 4].

[表3]

Figure JPOXMLDOC01-appb-T000124
Figure JPOXMLDOC01-appb-T000125
Figure JPOXMLDOC01-appb-T000126
Figure JPOXMLDOC01-appb-T000127
Figure JPOXMLDOC01-appb-T000128
Figure JPOXMLDOC01-appb-T000129
Figure JPOXMLDOC01-appb-T000130
Figure JPOXMLDOC01-appb-T000131
[Table 3]
Figure JPOXMLDOC01-appb-T000124
Figure JPOXMLDOC01-appb-T000125
Figure JPOXMLDOC01-appb-T000126
Figure JPOXMLDOC01-appb-T000127
Figure JPOXMLDOC01-appb-T000128
Figure JPOXMLDOC01-appb-T000129
Figure JPOXMLDOC01-appb-T000130
Figure JPOXMLDOC01-appb-T000131

[表4]

Figure JPOXMLDOC01-appb-T000132
Figure JPOXMLDOC01-appb-T000133
Figure JPOXMLDOC01-appb-T000134
Figure JPOXMLDOC01-appb-T000135
Figure JPOXMLDOC01-appb-T000136
Figure JPOXMLDOC01-appb-T000137
Figure JPOXMLDOC01-appb-T000138
Figure JPOXMLDOC01-appb-T000139
[Table 4]
Figure JPOXMLDOC01-appb-T000132
Figure JPOXMLDOC01-appb-T000133
Figure JPOXMLDOC01-appb-T000134
Figure JPOXMLDOC01-appb-T000135
Figure JPOXMLDOC01-appb-T000136
Figure JPOXMLDOC01-appb-T000137
Figure JPOXMLDOC01-appb-T000138
Figure JPOXMLDOC01-appb-T000139

試験例1
ALK5に対する活性阻害試験(TR-FRET法)
 QuickScout Screening Assist(商標)TR-FRETアッセイキット(GST-TGFβR1;カルナバイオサイエンス社製、カタログNo.09-141TR)を用い、時間分解蛍光-共鳴エネルギー転移(TR-FRET)法にてキナーゼ活性を測定した。
 被験化合物の10mM DMSO溶液から、10濃度(0.00003mM、0.0001mM、0.0003mM、0.001mM、0.003mM、0.01mM、0.03mM、0.1mM、0.3mM、1mM)にDMSOでさらに希釈し、それぞれをアッセイバッファーで25倍希釈して、薬物溶液とした(4%DMSO溶液)。
 薬物溶液もしくはコントロール溶液(5μL)、基質混合液(5μL)、および酵素溶液(10μL)を384穴黒プレートのウェル中で混合し、1時間室温で反応させた。検出溶液[15mM Tris-HCl(pH7.5),0.01% Tween20,20mM EDTA,0.53nM Eu標識抗リン酸化TopoIIa抗体(PerkinElmer社製)]を60μL添加して反応を停止させた後、キットに添付のプロトコルに準じ、TR-FRETシグナルをマルチモードプレートリーダー(EnVision、PerkinElmer社製)を用いて測定した。
(阻害活性の評価方法)
 ブランクとして酵素溶液の代わりにアッセイバッファーを添加したものを測定した。
被験化合物の阻害率(%)は、次の式に従って算出した。
  阻害率(%)=(1-(C-A)/(B-A))×100
ただし、A、B、Cは、それぞれブランクウェルのTR-FRETシグナル、コントロール溶液ウェルのTR-FRETシグナル、化合物添加ウェルのTR-FRETシグナルを示す。
 また、IC50値は、阻害率と被験化合物濃度(対数)の回帰分析により算出した。
Test Example 1
ALK5 activity inhibition test (TR-FRET method)
Kinase activity was measured by time-resolved fluorescence resonance energy transfer (TR-FRET) using a QuickScout Screening Assist™ TR-FRET Assay Kit (GST-TGFβR1; Karna Biosciences, Catalog No. 09-141TR).
A 10 mM DMSO solution of the test compound was further diluted with DMSO to 10 concentrations (0.00003 mM, 0.0001 mM, 0.0003 mM, 0.001 mM, 0.003 mM, 0.01 mM, 0.03 mM, 0.1 mM, 0.3 mM, 1 mM), and each was diluted 25-fold with assay buffer to prepare a drug solution (4% DMSO solution).
Drug or control solution (5 μL), substrate mixture (5 μL), and enzyme solution (10 μL) were mixed in wells of a 384-well black plate and reacted at room temperature for 1 hour. After stopping the reaction by adding 60 μL of detection solution [15 mM Tris-HCl (pH 7.5), 0.01% Tween 20, 20 mM EDTA, 0.53 nM Eu-labeled anti-phosphorylated TopoIIa antibody (PerkinElmer)], the TR-FRET signal was measured using a multimode plate reader (EnVision, PerkinElmer) according to the protocol attached to the kit.
(Method of evaluating inhibitory activity)
As a blank, an assay buffer was added instead of the enzyme solution to measure.
The inhibition rate (%) of the test compound was calculated according to the following formula.
Inhibition rate (%) = (1 - (C - A) / (B - A)) x 100
Here, A, B, and C respectively indicate the TR-FRET signal of a blank well, the TR-FRET signal of a control solution well, and the TR-FRET signal of a compound-added well.
The IC 50 value was calculated by regression analysis of the inhibition rate and the test compound concentration (logarithm).

(評価結果)
 本発明の代表化合物のALK5に対する阻害活性を表5に示す。キナーゼ活性阻害作用としてIC50値が、0.05μM未満を***印、0.05μM以上0.5μM未満を**印、0.5μM以上5μM未満を*印で示した。
(Evaluation results)
The inhibitory activity against ALK5 of representative compounds of the present invention is shown in Table 5. As kinase activity inhibitory action, IC50 values of less than 0.05 μM are indicated by ***, 0.05 μM or more and less than 0.5 μM are indicated by **, and 0.5 μM or more and less than 5 μM are indicated by *.

[表5]

Figure JPOXMLDOC01-appb-T000140
Figure JPOXMLDOC01-appb-T000141
Figure JPOXMLDOC01-appb-T000142
Figure JPOXMLDOC01-appb-T000143
Figure JPOXMLDOC01-appb-T000144
Figure JPOXMLDOC01-appb-T000145
Figure JPOXMLDOC01-appb-T000146
Figure JPOXMLDOC01-appb-T000147

 この結果は、本発明の化合物(I)が、強いALK5阻害活性を有することを示している。 [Table 5]
Figure JPOXMLDOC01-appb-T000140
Figure JPOXMLDOC01-appb-T000141
Figure JPOXMLDOC01-appb-T000142
Figure JPOXMLDOC01-appb-T000143
Figure JPOXMLDOC01-appb-T000144
Figure JPOXMLDOC01-appb-T000145
Figure JPOXMLDOC01-appb-T000146
Figure JPOXMLDOC01-appb-T000147

This result indicates that compound (I) of the present invention has strong ALK5 inhibitory activity.

試験例2
細胞内ALK5によるSmad3リン酸化阻害試験
(使用する細胞の培養)
 A549細胞(ATCC社No.CCL-185)は、T75フラスコ中、10%ウシ胎仔血清(以下、FBS)(AusGene社)および1%ペニシリン・ストレプトマイシン(ナカライ社)を添加したD-MEM培地(ナカライ社、#08459-64)(以下、増殖培地)を用いて37℃、5%CO2インキュベーター内で培養した。培養したA549細胞を細胞密度3.3×105cells/mLになるように増殖培地で希釈して得られた細胞懸濁液を24ウェル培養プレート(Falcon社、353226)に3.3×105cells/mL/wellになるように播種して、37℃、5%COインキュベーター内で一晩培養した。翌日、増殖培地をアスピレーターで除去し、すぐに37℃で保温したFBSを含まないD-MEM培地(以下、培地)で洗浄した。各ウェルに培地を0.989mLずつ添加して細胞プレートを調製した。
(被験化合物の添加およびTGFβによる刺激)
 細胞プレートの各ウェルに被験化合物のDMSO溶液(1000~3μM)を1μL添加し(コントロールウェルはDMSOのみを1μL添加)、5%COインキュベーター内にて37℃で1時間インキュベートした。培地で200ng/mLに調製したTGFβ1(Peprotec社,PEP-100-21-10)を、細胞プレートに各ウェル10μLずつ添加し(コントロールウェルは培地のみ10μL添加)、さらに5%COインキュベーター内にて37℃で30分間インキュベートした。
Test Example 2
Inhibition test of Smad3 phosphorylation by intracellular ALK5 (culture of cells used)
A549 cells (ATCC No. CCL-185) were cultured in a T75 flask at 37°C in a 5% CO2 incubator using D-MEM medium (Nacalai, #08459-64) (hereinafter referred to as growth medium) supplemented with 10% fetal bovine serum (hereinafter referred to as FBS) (AusGene) and 1% penicillin-streptomycin (Nacalai). The cultured A549 cells were diluted with growth medium to a cell density of 3.3 x 105 cells/mL, and the resulting cell suspension was seeded in a 24-well culture plate (Falcon, 353226) at 3.3 x 105 cells/mL/well, and cultured overnight at 37°C in a 5% CO2 incubator. The next day, the growth medium was removed with an aspirator and immediately washed with FBS-free D-MEM medium (hereinafter referred to as medium) kept at 37°C. The cell plate was prepared by adding 0.989 mL of medium to each well.
(Addition of Test Compound and Stimulation with TGFβ)
1 μL of a DMSO solution of the test compound (1000-3 μM) was added to each well of the cell plate (1 μL of DMSO alone was added to the control well), and the plate was incubated for 1 hour at 37° C. in a 5% CO 2 incubator. 10 μL of TGFβ1 (Peprotec, PEP-100-21-10) adjusted to 200 ng/mL in culture medium was added to each well of the cell plate (10 μL of culture medium alone was added to the control well), and the plate was further incubated for 30 minutes at 37° C. in a 5% CO 2 incubator.

(タンパク質の抽出)
 細胞プレートから培地をアスピレーターで除去し、PBS(-)で2回洗浄後、Lysisバッファー[RIPA Buffer(×1)(Cell Signaling Technology社)に、1% Phosphatase inhibitor Cacktail 3(Sigma社、No.P0044)、1% Phosphatase inhibitor Cacktail (ナカライ社、No.07575)および1mM フッ化フェニルメチルスルホニル(PMSF)を添加したもの]を100μL添加し、軽く攪拌したのち10分間静置した。遠心操作(15,000rpm、15分間)により上清を回収し、タンパク質量を定量した。SDS-サンプルバッファーと混合し、95℃5分間反応させてタンパク質を変性させて、サンプル溶液とした。5-20%のグラジエントアクリルアミドゲル(ナカライ社、No.13064-04)の各ウェルにサンプル溶液を5μg/12μLずつアプライし、電気泳動を行った。その後、トランスブロット Turbo転写システム(バイオラッド社)を用いてPVDF膜にゲル中のタンパク質を転写した。
(リン酸化Smad3の検出)
 転写したPVDF膜を2%ECL prime blocking agent(GEヘルスケア社)でブロッキング処理した後、一次抗体としてウサギ抗リン酸化Smad3抗体(Abcam社、ab51451)あるいはマウス抗βアクチン抗体(Abcam社、ab6276)を用い、4℃で一晩反応させた。未反応の一次抗体をTBSTバッファー(10mM Tris-HCl(pH7.5),150mM NaCl,0.1%Tween20)で洗浄後、二次抗体としてHRP標識ヤギ抗ウサギIgG抗体(Cell Signaling Technology社、No.7074)あるいはHRP標識ヤギ抗マウスIgG抗体(Cell Signaling Technology社、No.7076)を用い、2%ECL prime blocking agentを添加したTBSTバッファー中で、室温で1時間反応させた。未反応の二次抗体をTBSTバッファーで洗浄後、ケミルミワンSuper(ナカライ社)を用いて添付のプロトコルどおりに反応させた後、CCDカメラ(GEヘルスケア社、ImageQuant LAS500)を用いて、それぞれのバンドを化学発光で検出した。
(Protein Extraction)
The medium was removed from the cell plate with an aspirator, washed twice with PBS (-), and then 100 μL of lysis buffer [RIPA Buffer (×1) (Cell Signaling Technology) to which 1% Phosphatase inhibitor Cacktail 3 (Sigma, No. P0044), 1% Phosphatase inhibitor Cacktail (Nacalai, No. 07575) and 1 mM phenylmethylsulfonyl fluoride (PMSF) were added] was added, gently stirred, and then allowed to stand for 10 minutes. The supernatant was collected by centrifugation (15,000 rpm, 15 minutes), and the protein amount was quantified. The mixture was mixed with SDS-sample buffer and reacted at 95°C for 5 minutes to denature the proteins, and a sample solution was prepared. 5 μg/12 μL of the sample solution was applied to each well of a 5-20% gradient acrylamide gel (Nacalai, No. 13064-04) and electrophoresis was performed. The proteins in the gel were then transferred to a PVDF membrane using a Transblot Turbo transfer system (Bio-Rad).
(Detection of phosphorylated Smad3)
The transferred PVDF membrane was subjected to blocking treatment with 2% ECL prime blocking agent (GE Healthcare), and then reacted overnight at 4° C. with rabbit anti-phosphorylated Smad3 antibody (Abeam, ab51451) or mouse anti-β-actin antibody (Abeam, ab6276) as the primary antibody. After washing off the unreacted primary antibody with TBST buffer (10 mM Tris-HCl (pH 7.5), 150 mM NaCl, 0.1% Tween 20), the sections were reacted with HRP-labeled goat anti-rabbit IgG antibody (Cell Signaling Technology, No. 7074) or HRP-labeled goat anti-mouse IgG antibody (Cell Signaling Technology, No. 7076) as the secondary antibody at room temperature for 1 hour in TBST buffer supplemented with 2% ECL prime blocking agent. After washing off unreacted secondary antibody with TBST buffer, the antibody was reacted with Chemiluminescent One Super (Nacalai) according to the attached protocol, and each band was detected by chemiluminescence using a CCD camera (GE Healthcare, ImageQuant LAS500).

 本試験で用いた一次抗体と二次抗体の組み合わせおよび希釈濃度は以下の通りである。

Figure JPOXMLDOC01-appb-T000148
(阻害活性の評価方法)
 検出されたバンドをデンシトメトリー(ImageQuant TL解析ソフトウェア)により数値化し、化合物非添加かつTGFβ刺激群のリン酸化Smad3のバンドの発光を100%、化合物非添加かつTGFβ無刺激群のリン酸化Smad3のバンドの発光を0%として、各群におけるバンドの強度から阻害率を算出した。なお、それぞれのリン酸化Smad3のバンドは、βアクチンにより補正を行なった。
 被験化合物のSmad3リン酸化の阻害率(%)は、次の式に従って算出した。
阻害率(%)=(1-(C-A)/(B-A))×100
ただし、A、B、Cは、それぞれ化合物非添加かつTGFβ無刺激群のリン酸化Smad3のバンドの発光、化合物非添加かつTGFβによる刺激群のリン酸化Smad3のバンドの発光、化合物添加かつTGFβによる刺激群のリン酸化Smad3のバンドの発光を示す。
また、IC50値は、阻害率と被験化合物濃度(対数)の回帰分析により算出した。
 本発明の代表化合物の細胞内Smad3リン酸化阻害活性を、表7に示す。細胞内Smad3リン酸化阻害活性としてIC50値が、0.1μM未満を***印、0.1μM以上0.3μM未満を**印、0.3μM以上を*印で示した。 The combinations and dilution concentrations of the primary and secondary antibodies used in this test are as follows.
Figure JPOXMLDOC01-appb-T000148
(Method of evaluating inhibitory activity)
The detected bands were quantified by densitometry (ImageQuant TL analysis software), and the inhibition rate was calculated from the band intensity in each group, taking the luminescence of the phosphorylated Smad3 band in the compound-free and TGFβ-stimulated group as 100% and the luminescence of the phosphorylated Smad3 band in the compound-free and TGFβ-unstimulated group as 0%. Each phosphorylated Smad3 band was corrected with β-actin.
The inhibition rate (%) of Smad3 phosphorylation by the test compound was calculated according to the following formula.
Inhibition rate (%) = (1 - (C - A) / (B - A)) x 100
Here, A, B, and C respectively show the luminescence of the phosphorylated Smad3 band in the group without addition of compound and without stimulation with TGFβ, the group without addition of compound and stimulation with TGFβ, and the group with addition of compound and stimulation with TGFβ.
The IC 50 value was calculated by regression analysis of the inhibition rate and the test compound concentration (logarithm).
The inhibitory activity of intracellular Smad3 phosphorylation of representative compounds of the present invention is shown in Table 7. As the inhibitory activity of intracellular Smad3 phosphorylation, IC50 values of less than 0.1 μM are indicated by ***, those of 0.1 μM or more and less than 0.3 μM are indicated by **, and those of 0.3 μM or more are indicated by *.

[表7]

Figure JPOXMLDOC01-appb-T000149
Figure JPOXMLDOC01-appb-T000150
Figure JPOXMLDOC01-appb-T000151

 本試験において、表7に示すとおり、本発明化合物(I)は細胞内においてALK5のSmad3リン酸化を強く阻害した。
 試験例2の結果は、本発明の化合物(I)が、“細胞内ALK5のSmad3リン酸化作用”についても、強い阻害作用を有することを示している。 [Table 7]
Figure JPOXMLDOC01-appb-T000149
Figure JPOXMLDOC01-appb-T000150
Figure JPOXMLDOC01-appb-T000151

In this test, as shown in Table 7, compound (I) of the present invention strongly inhibited Smad3 phosphorylation of ALK5 in cells.
The results of Test Example 2 show that compound (I) of the present invention also has a strong inhibitory effect on the "Smad3 phosphorylation effect of intracellular ALK5."

試験例3
TGF-β刺激による制御性T細胞(Treg)分化誘導の阻害試験
 マウス(BALB/cCrSlc、雌、7週齢)から脾臓を採取し、セルストレイナーを使って調整した脾細胞の懸濁液(FBS-free IMDM培地)からHISTOPAQUE-1083(SIGMA社製)を用いてマウスリンパ球細胞を分離した。
 このマウスリンパ球細胞からEasySep Mouse Naive CD4+ T Cell Isolation Kit(STEMCELL technologies社製)を用い、キット付属のプロトコルに従い、CD4陽性T細胞を分離した。
96ウェルプレートをanti-CD3ε抗体(BioXCell社製)でコートしたのち、被験化合物のDMSO溶液をIL-2含有FBS添加IMDM培地で100倍に希釈した化合物溶液30μLを、各ウェルに添加した。CD4陽性T細胞をIL-2含有FBS添加IMDM培地に懸濁し(2x10cells/mL)、各ウェルに0.1mLずつ添加し、COインキュベーターで1時間インキュベートした。IL-2含有FBS添加IMDM培地で希釈したanti-CD28抗体(BioXCell社製)およびTGF-β1(Gibco社製)をそれぞれ30μLずつ各ウェルに添加し、さらにIL-2含有FBS添加IMDM培地110μLを添加してCOインキュベーターで5日間培養した。
 各ウェルの細胞を回収し、FITC anti-mouse CD4 Antibody(BioLegend社製)、APC anti-mouse CD25 Antibody(BioLegend社製)、FOXP3 Monoclonal Antibody(FJK-16s) PE(eBioscience社製)で細胞を染色し、フローサイトメーターを用いてTreg画分(CD4+/CD25+/Foxp3+)の割合を測定した。TGF-β1の代わりにIL-2含有FBS添加IMDM培地を添加したものをコントロールとして用いた。IC50値は、Treg画分の割合変化と被験化合物濃度(対数)の回帰分析により算出した。
Test Example 3
Inhibition test of regulatory T cell (Treg) differentiation induction by TGF-β stimulation Spleens were collected from mice (BALB/cCrSlc, female, 7 weeks old), and mouse lymphocytes were separated from a suspension of splenocytes (FBS-free IMDM medium) prepared using a cell strainer using HISTOPAQUE-1083 (Sigma).
CD4 positive T cells were isolated from these mouse lymphocytes using EasySep Mouse Naive CD4+ T Cell Isolation Kit (manufactured by STEMCELL technologies) according to the protocol attached to the kit.
After coating a 96-well plate with anti-CD3ε antibody (manufactured by BioXCell), 30 μL of a compound solution prepared by diluting a DMSO solution of a test compound 100-fold with IMDM medium supplemented with IL-2-containing FBS was added to each well. CD4-positive T cells were suspended in IMDM medium supplemented with IL-2-containing FBS (2×10 5 cells/mL), 0.1 mL of the solution was added to each well, and the mixture was incubated for 1 hour in a CO 2 incubator. 30 μL of anti-CD28 antibody (manufactured by BioXCell) and TGF-β1 (manufactured by Gibco) diluted with IMDM medium supplemented with IL-2-containing FBS were added to each well, and 110 μL of IMDM medium supplemented with IL-2-containing FBS was added, followed by incubation for 5 days in a CO 2 incubator.
The cells were collected from each well, stained with FITC anti-mouse CD4 Antibody (BioLegend), APC anti-mouse CD25 Antibody (BioLegend), and FOXP3 Monoclonal Antibody (FJK-16s) PE (eBioscience), and the proportion of the Treg fraction (CD4+/CD25+/Foxp3+) was measured using a flow cytometer. As a control, IMDM medium supplemented with IL-2-containing FBS was used instead of TGF-β1. The IC 50 value was calculated by regression analysis of the change in the proportion of the Treg fraction and the test compound concentration (logarithm).

 本発明の代表化合物のTGF-β刺激によるTreg分化誘導阻害活性を、表8に示す。Treg分化誘導阻害活性としてIC50値が、0.1μM未満を***印、0.1μM以上0.5μM未満を**印、0.5μM以上を*印で示した。
[表8]

Figure JPOXMLDOC01-appb-T000152


 本試験において、表8に示すとおり、本発明化合物(I)はナイーブT細胞においてTGF-βシグナルを阻害し、TGF-β刺激によるTregへの分化誘導を強く抑制した。
 試験例3の結果は、本発明の化合物(I)が、細胞内TGF-βシグナルを阻害し、Tregの分化誘導に対して強い抑制作用を有することを示している。 The inhibitory activity of representative compounds of the present invention in inducing Treg differentiation by TGF-β stimulation is shown in Table 8. As the inhibitory activity inducing Treg differentiation, IC50 values of less than 0.1 μM are indicated by ***, 0.1 μM or more and less than 0.5 μM by **, and 0.5 μM or more by *.
[Table 8]
Figure JPOXMLDOC01-appb-T000152


In this test, as shown in Table 8, compound (I) of the present invention inhibited TGF-β signaling in naive T cells and strongly suppressed the induction of differentiation into Treg by TGF-β stimulation.
The results of Test Example 3 show that compound (I) of the present invention inhibits intracellular TGF-β signaling and has a strong suppressive effect on the induction of Treg differentiation.

試験例4
マウス大腸がん細胞株CT26.WTの同種移植マウスモデルにおける抗PD-1抗体との併用効果
 本発明化合物の癌免疫療法による免疫チェックポイント阻害剤との併用効果を、マウス大腸がん細胞株CT26.WTの同系マウス腫瘍モデル(皮下移植)を用いて検討した。
(担癌モデルの作製)
 CT26.WT細胞を細胞密度1×10個/mLになるように、HBSS(-)培地(ナカライ社製)で調整し、移植用細胞調製液を作製した。この移植用細胞調製液0.1mLを、BALB/cCrslcマウス(雌、7週齢、日本エスエルシー社)の背部皮下に移植した。がん細胞を移植してから4日目に担癌マウスの腫瘍体積(下記計算式参照)の平均値が近似するように群分けを行った。
(被験物質の投与用試料溶液の調製)
 被験物質を10mg/mLになるように投与溶媒(Solutol HS15(BASF社製)/0.5w/v%メチルセルロース400溶液(富士フイルム和光純薬社製)=1:4)に懸濁させ、30分間ソニケーションして投与用試料溶液を調製した。
(抗体溶液の調製)
 抗PD-1抗体(Bio X cell社製、clone RMP1-14;カタログNo.BE0146)を投与直前に生理食塩水で1mg/mLになるように調製した。
(被験物質の抗腫瘍効果試験)
 がん細胞を移植した各マウス(各群6匹)に、それぞれの当日の体重10g当たり0.1mLの被験物質(100mg/kg)もしくは溶媒を1日1回、移植後4日目から20日目までの強制経口投与を行なった。移植後9、10、16、17日目は休薬とした。また、抗体投与群、薬剤併用群には、週に2回(計5回)各マウスに当日の体重10g当たり0.1mLの抗体溶液(10mg/kg)、それ以外の群には生理食塩水を腹腔内投与した。移植後、21日目まで経過観察し、週に数回、腫瘍径を測定して各マウスの腫瘍体積を以下の式を用いて算出し抗腫瘍効果を評価した。
数式
腫瘍体積=長径×短径×短径×0.5
(評価結果)
 図1に、各群における腫瘍体積の経時変化を示す。図1に示すように、本発明化合物である実施例6および実施例7の化合物は抗PD-1抗体と併用することで、顕著な腫瘍増殖抑制・腫瘍退縮効果を示した。このことから、本発明化合物が免疫チェックポイント阻害剤との併用で優れた抗腫瘍効果を示し、癌の治療において有用であることが確認された。
Test Example 4
Effect of Combination with Anti-PD-1 Antibody in an Allograft Mouse Model of Mouse Colon Cancer Cell Line CT26.WT The effect of combination with an immune checkpoint inhibitor in cancer immunotherapy was examined using a syngeneic mouse tumor model (subcutaneous transplant) of the mouse colon cancer cell line CT26.WT.
(Preparation of tumor-bearing model)
CT26.WT cells were adjusted to a cell density of 1 x 107 cells/mL in HBSS(-) medium (manufactured by Nacalai) to prepare a cell preparation for transplantation. 0.1 mL of this cell preparation for transplantation was subcutaneously transplanted into the back of a BALB/cCrslc mouse (female, 7 weeks old, Japan SLC). Four days after the cancer cells were transplanted, the mice were divided into groups so that the average tumor volume (see the calculation formula below) of the cancer-bearing mice was close to each other.
(Preparation of sample solution for administration of test substance)
The test substance was suspended in an administration solvent (Solutol HS15 (BASF)/0.5 w/v % methylcellulose 400 solution (Fujifilm Wako Pure Chemical Industries, Ltd.) = 1:4) to a concentration of 10 mg/mL, and the suspension was sonicated for 30 minutes to prepare a sample solution for administration.
(Preparation of antibody solution)
Anti-PD-1 antibody (Bio X Cell, clone RMP1-14; catalog No. BE0146) was prepared with physiological saline to a concentration of 1 mg/mL immediately before administration.
(Antitumor effect test of test substance)
Each mouse (6 mice per group) transplanted with cancer cells was forced to orally administer 0.1 mL of the test substance (100 mg/kg) or the solvent per 10 g of body weight on the day once a day from the 4th to the 20th day after transplantation. Drugs were suspended on the 9th, 10th, 16th, and 17th days after transplantation. In addition, the antibody administration group and the drug combination group were intraperitoneally administered 0.1 mL of the antibody solution (10 mg/kg) per 10 g of body weight on the day twice a week (5 times in total), and saline was administered intraperitoneally to the other groups. After transplantation, the mice were observed until the 21st day, and the tumor diameter was measured several times a week to calculate the tumor volume of each mouse using the following formula to evaluate the antitumor effect.
Formula Tumor volume = major axis x minor axis x minor axis x 0.5
(Evaluation results)
Figure 1 shows the change in tumor volume over time in each group. As shown in Figure 1, the compounds of the present invention, Example 6 and Example 7, showed a significant tumor growth suppression/tumor regression effect when used in combination with an anti-PD-1 antibody. This confirmed that the compounds of the present invention showed an excellent antitumor effect when used in combination with an immune checkpoint inhibitor, and are useful in the treatment of cancer.

試験例5
マウス大腸がん細胞株CT26.WTの同種移植マウスモデルにおける抗PD-1抗体との併用効果2
(担癌モデルの作製)
 CT26.WT細胞を細胞密度1×10個/mLになるように、HBSS(-)培地で調整し、移植用細胞調製液を作製した。この移植用細胞調製液0.1mLを、BALB/cCrslcマウス(雌、7週齢、日本エスエルシー社)の背部皮下に移植した。がん細胞を移植してから3日目に担癌マウスの腫瘍体積(試験例4の計算式参照)の平均値が近似するように群分けを行った。
(被験物質の投与用試料溶液の調製)
 被験物質を3mg/mLになるように投与溶媒(DMSO/ポリエチレングリコール400溶液/30%ヒドロキシプロピル-β-シクロデキストリン=1:4:15)に溶解させ、投与用試料溶液を調製した。
(被験物質の抗腫瘍効果試験)
 がん細胞を移植した各マウス(溶媒群10匹、実施例184の化合物群9匹、抗体PD-1抗体群9匹、実施例184の化合物と抗体PD-1抗体併用群8匹)に、それぞれの当日の体重10g当たり0.1mLの被験物質(30mg/kg)もしくは溶媒を1日1回、移植後3日目から21日目までの強制経口投与を行なった。移植後8、9、15、16日目は休薬とした。また、抗体投与群、薬剤併用群には、週に2回(計6回)各マウスに当日の体重10g当たり0.1mLの抗体溶液(10mg/kg)、それ以外の群には生理食塩水を腹腔内投与した。移植後、21日目まで経過観察し、週に数回、腫瘍径を測定して各マウスの腫瘍体積を試験例4の計算式を用いて算出し抗腫瘍効果を評価した。
(評価結果)
図2に、各群における腫瘍体積の経時変化を示す。図2に示すように、本発明化合物である実施例184の化合物は単剤もしくは抗PD-1抗体と併用することで、顕著な腫瘍増殖抑制・腫瘍退縮効果を示した。このことから、本発明化合物が免疫チェックポイント阻害剤との併用で優れた抗腫瘍効果を示し、癌の治療において有用であることが確認された。
Test Example 5
Combination effect of anti-PD-1 antibody in allograft mouse model of mouse colon cancer cell line CT26.WT 2
(Preparation of tumor-bearing model)
CT26.WT cells were adjusted to a cell density of 1 x 107 cells/mL with HBSS(-) medium to prepare a cell preparation for transplantation. 0.1 mL of this cell preparation for transplantation was subcutaneously transplanted into the back of a BALB/cCrslc mouse (female, 7 weeks old, Japan SLC). On the third day after the cancer cells were transplanted, the mice were divided into groups so that the average tumor volume (see the calculation formula in Test Example 4) of the cancer-bearing mice was close to each other.
(Preparation of sample solution for administration of test substance)
The test substance was dissolved in an administration solvent (DMSO/polyethylene glycol 400 solution/30% hydroxypropyl-β-cyclodextrin=1:4:15) to a concentration of 3 mg/mL to prepare a sample solution for administration.
(Antitumor effect test of test substance)
Each mouse transplanted with cancer cells (10 mice in the solvent group, 9 mice in the compound group of Example 184, 9 mice in the antibody PD-1 antibody group, and 8 mice in the compound of Example 184 and antibody PD-1 antibody combination group) was forcibly administered 0.1 mL of the test substance (30 mg/kg) or the solvent per 10 g of body weight on the day once a day from the 3rd to the 21st day after transplantation. Drugs were suspended on the 8th, 9th, 15th, and 16th days after transplantation. In addition, the antibody administration group and the drug combination group were intraperitoneally administered 0.1 mL of antibody solution (10 mg/kg) per 10 g of body weight on the day twice a week (6 times in total), and saline was intraperitoneally administered to the other groups. After transplantation, the mice were observed until the 21st day, and the tumor diameter was measured several times a week to calculate the tumor volume of each mouse using the formula in Test Example 4 to evaluate the antitumor effect.
(Evaluation results)
Figure 2 shows the change in tumor volume over time in each group. As shown in Figure 2, the compound of the present invention, Example 184, showed a significant tumor growth inhibitory and tumor regression effect when used alone or in combination with an anti-PD-1 antibody. This confirmed that the compound of the present invention showed an excellent antitumor effect when used in combination with an immune checkpoint inhibitor, and is useful in the treatment of cancer.

 本発明により提供される化合物は、TGFβシグナル経路を介した異常な細胞応答に関連していることが知られている疾患、特にがんの治療に有用であり、また、がん幹細胞を標的とした腫瘍の転移および再発予防にも有用である。さらに免疫チェックポイント阻害剤などと併用することで、がん免疫療法において治療効果の拡大および増強に有用である。また、線維化疾患等の治療や予防に有用である。さらに、ALK5阻害剤、TGFβシグナル阻害剤として、実験用、研究用の試薬としても有用である。 The compounds provided by the present invention are useful for treating diseases known to be associated with abnormal cell responses via the TGFβ signal pathway, particularly cancer, and are also useful for preventing tumor metastasis and recurrence by targeting cancer stem cells. Furthermore, when used in combination with immune checkpoint inhibitors, they are useful for expanding and enhancing the therapeutic effect in cancer immunotherapy. They are also useful for treating and preventing fibrotic diseases, etc. Furthermore, they are useful as experimental and research reagents as ALK5 inhibitors and TGFβ signal inhibitors.

Claims (4)

 下式(I):
Figure JPOXMLDOC01-appb-C000001
(式中、Rは置換基を有してもよいアルキル基、置換基を有してもよい飽和ヘテロ環式基または置換基を有してもよいアミノ基を表わし、Rは置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基または置換基を有してもよいアルキニル基を表し、Zは置換基を有してもよいアルキル基、置換基を有してもよいアルケニル基、置換基を有してもよいアルキニル基、置換基を有してもよい飽和ヘテロ環式基または置換基を有してもよいシクロアルキル基を表す。)で示されるチアゾール誘導体またはその薬学的に許容される塩。
The following formula (I):
Figure JPOXMLDOC01-appb-C000001
(wherein R 1 represents an optionally substituted alkyl group, an optionally substituted saturated heterocyclic group or an optionally substituted amino group; R 2 represents an optionally substituted aryl group, an optionally substituted heteroaryl group or an optionally substituted alkynyl group; and Z represents an optionally substituted alkyl group, an optionally substituted alkenyl group, an optionally substituted alkynyl group, an optionally substituted saturated heterocyclic group or an optionally substituted cycloalkyl group), or a pharmacy acceptable salt thereof.
 Rが置換基を有してもよいアリール基、置換基を有してもよいヘテロアリール基である、請求項1に記載のチアゾール誘導体またはその薬学的に許容される塩。 The thiazole derivative or a pharma- ceutically acceptable salt thereof according to claim 1, wherein R2 is an optionally substituted aryl group or an optionally substituted heteroaryl group.  Rが置換基を有してもよいアルキニル基である、請求項1に記載のチアゾール誘導体またはその薬学的に許容される塩。 The thiazole derivative or a pharma- ceutically acceptable salt thereof according to claim 1, wherein R2 is an alkynyl group which may have a substituent.  実施例1から226よりなる群から選択される、チアゾール誘導体またはその薬学的に許容される塩。 A thiazole derivative or a pharma- ceutically acceptable salt thereof selected from the group consisting of Examples 1 to 226.
PCT/JP2023/041996 2022-11-25 2023-11-22 Novel thiazole derivative Ceased WO2024111626A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2022188246 2022-11-25
JP2022-188246 2022-11-25
JP2023-104088 2023-06-26
JP2023104088 2023-06-26

Publications (1)

Publication Number Publication Date
WO2024111626A1 true WO2024111626A1 (en) 2024-05-30

Family

ID=91196177

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2023/041996 Ceased WO2024111626A1 (en) 2022-11-25 2023-11-22 Novel thiazole derivative

Country Status (1)

Country Link
WO (1) WO2024111626A1 (en)

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2005527590A (en) * 2002-04-04 2005-09-15 バイオジェン, インコーポレイテッド Trisubstituted heteroaryls and methods of making and using them
JP2006516603A (en) * 2003-01-27 2006-07-06 ファイザー・プロダクツ・インク Isothiazole derivatives
JP2006517592A (en) * 2003-02-12 2006-07-27 バイオジェン・アイデック・エムエイ・インコーポレイテッド Pyrazole and methods of making and using them
JP2008511631A (en) * 2004-08-31 2008-04-17 バイオジェン・アイデック・エムエイ・インコーポレイテッド Pyrimidinylimidazoles as TGF-β inhibitors
JP2013507932A (en) * 2009-10-16 2013-03-07 ザ スクリプス リサーチ インスティチュート How to induce pluripotent cells
JP2015517453A (en) * 2012-05-24 2015-06-22 カルナバイオサイエンス株式会社 New bicyclic thiazole compounds
JP2017537886A (en) * 2014-10-22 2017-12-21 ダナ−ファーバー キャンサー インスティテュート, インコーポレイテッド Thiazolyl-containing compounds for treating proliferative diseases
WO2020012357A1 (en) * 2018-07-10 2020-01-16 Integral Biosciences Pvt. Ltd. Heterocyclic compounds and methods of use
WO2020103817A1 (en) * 2018-11-20 2020-05-28 南京圣和药业股份有限公司 TGF-βR1 INHIBITOR AND USE THEREOF
WO2020146819A1 (en) * 2019-01-10 2020-07-16 Tolero Pharmaceuticals, Inc. Alk5 inhibitors for treating myelodysplastic syndrome

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2005527590A (en) * 2002-04-04 2005-09-15 バイオジェン, インコーポレイテッド Trisubstituted heteroaryls and methods of making and using them
JP2006516603A (en) * 2003-01-27 2006-07-06 ファイザー・プロダクツ・インク Isothiazole derivatives
JP2006517592A (en) * 2003-02-12 2006-07-27 バイオジェン・アイデック・エムエイ・インコーポレイテッド Pyrazole and methods of making and using them
JP2008511631A (en) * 2004-08-31 2008-04-17 バイオジェン・アイデック・エムエイ・インコーポレイテッド Pyrimidinylimidazoles as TGF-β inhibitors
JP2013507932A (en) * 2009-10-16 2013-03-07 ザ スクリプス リサーチ インスティチュート How to induce pluripotent cells
JP2015517453A (en) * 2012-05-24 2015-06-22 カルナバイオサイエンス株式会社 New bicyclic thiazole compounds
JP2017537886A (en) * 2014-10-22 2017-12-21 ダナ−ファーバー キャンサー インスティテュート, インコーポレイテッド Thiazolyl-containing compounds for treating proliferative diseases
WO2020012357A1 (en) * 2018-07-10 2020-01-16 Integral Biosciences Pvt. Ltd. Heterocyclic compounds and methods of use
WO2020103817A1 (en) * 2018-11-20 2020-05-28 南京圣和药业股份有限公司 TGF-βR1 INHIBITOR AND USE THEREOF
WO2020146819A1 (en) * 2019-01-10 2020-07-16 Tolero Pharmaceuticals, Inc. Alk5 inhibitors for treating myelodysplastic syndrome

Similar Documents

Publication Publication Date Title
KR101816942B1 (en) Nitrogen-containing heterocyclic compound having kynurenine production inhibitory activity
JP2021176859A (en) Heterocyclic compounds as immunomodulators
CN113490495A (en) Small molecule degradation agent of HELIOS and use method thereof
TWI865480B (en) Compounds for inhibiting pge2/ep4 signaling, preparation method thereof and use in medicine
TW201910306A (en) Selective inhibitor of NLRP3 inflammatory body
JP7741142B2 (en) Certain pladienolide compounds and methods of use
JP2022542669A (en) IRAK4 kinase inhibitor and method of preparation thereof
CN110857293A (en) Novel quinoline derivative inhibitor
JPWO2016031987A1 (en) Pyrimidinone derivatives with autotaxin inhibitory activity
AU2018337138B2 (en) 2-substituted pyrazole amino-4-substituted amino-5-pyrimidine formamide compound, composition, and application thereof
JP2021525810A (en) Tryptophan catabolism modulator
WO2024111626A1 (en) Novel thiazole derivative
US10550125B2 (en) Prodrugs of imidazotriazine compounds as CK2 inhibitors
KR101718299B1 (en) Azaindole derivative
KR20240014050A (en) Compounds as PD1/PD-L1 inhibitors and methods thereof
WO2025231105A1 (en) Processes of making csf-1r inhibitors and methods of use thereof
HK40051501B (en) Compound for inhibiting pge2/ep4 signaling transduction inhibiting, preparation method therefor, and medical uses thereof
HK40051501A (en) Compound for inhibiting pge2/ep4 signaling transduction inhibiting, preparation method therefor, and medical uses 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: 23894630

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: 23894630

Country of ref document: EP

Kind code of ref document: A1