CN108069955B - 3-pyridyl-4-benzothiazolylpyrazole derivatives, preparation method and medical application thereof - Google Patents

3-pyridyl-4-benzothiazolylpyrazole derivatives, preparation method and medical application thereof Download PDF

Info

Publication number
CN108069955B
CN108069955B CN201711111958.8A CN201711111958A CN108069955B CN 108069955 B CN108069955 B CN 108069955B CN 201711111958 A CN201711111958 A CN 201711111958A CN 108069955 B CN108069955 B CN 108069955B
Authority
CN
China
Prior art keywords
compound
cancer
alkyl
general formula
added
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.)
Active
Application number
CN201711111958.8A
Other languages
Chinese (zh)
Other versions
CN108069955A (en
Inventor
杨方龙
沈光远
韩吉慧
贺峰
陶维康
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.)
Jiangsu Hengrui Medicine Co Ltd
Shanghai Hengrui Pharmaceutical Co Ltd
Original Assignee
Jiangsu Hengrui Medicine Co Ltd
Shanghai Hengrui Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jiangsu Hengrui Medicine Co Ltd, Shanghai Hengrui Pharmaceutical Co Ltd filed Critical Jiangsu Hengrui Medicine Co Ltd
Publication of CN108069955A publication Critical patent/CN108069955A/en
Application granted granted Critical
Publication of CN108069955B publication Critical patent/CN108069955B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings

Abstract

The invention relates to 3-pyridyl-4-benzothiazolyl pyrazole derivatives, a preparation method and application thereof in medicines. Specifically, the invention relates to a novel 3-pyridyl-4-benzothiazolylpyrazole derivative shown in a general formula (I), a preparation method thereof, a pharmaceutical composition containing the derivative, and application thereof as a therapeutic agent, especially application thereof as a TGF-beta inhibitor and application thereof in preparing a medicament for treating, preventing or reducing cancer mediated by TGF-beta overexpression, wherein each substituent in the general formula (I) is as defined in the specification.

Description

3-pyridyl-4-benzothiazolylpyrazole derivatives, preparation method and medical application thereof
Technical Field
The invention belongs to the field of medicines, and relates to a novel 3-pyridyl-4-benzothiazolylpyrazole derivative, a preparation method thereof, a pharmaceutical composition containing the derivative, application of the derivative as a therapeutic agent, especially as a TGF-beta inhibitor, and application of the derivative in preparation of medicines for treating, preventing or reducing cancers mediated by TGF-beta overexpression.
Background
Transforming Growth Factor TGF- β (Transforming Growth Factor β) is a member of the dimeric polypeptide Growth Factor superfamily, which includes, for example, activins, inhibins, Bone Morphogenetic Proteins (BMPs), Growth and Differentiation Factors (GDFs) and Müllerian-inhibiting substance,MIS.
TGF-beta has three subtypes, TGF-beta 1, TGF-beta 2 and TGF-beta 3, which are involved in regulation of cell proliferation and differentiation, wound healing, extracellular matrix production and immunosuppression. See, e.g., Massague, J.Ann.Rev, cell.biol.6:594-641 (1990); roberts, A.B.peptide Growth Factor and theory receptors,95:419-472Berlin, Springer-Verlag (1990); roberts, A.B. and Sporn M.B.growth Factor 8:1-9 (1993); and Alexandrow, M.G., Moses, H.L.cancer Res.55: 1452-. Three subtypes of TGF-. beta.are present in most cells along with their receptors. Each TGF-. beta.subtype is synthesized as a precursor protein that is cleaved intracellularly into the C-terminal region (LAP) and the N-terminal portion, referred to as mature or active TGF-. beta.s. LAP is typically linked to mature TGF-. beta.in a non-covalent manner prior to secretion from the cell. The LAP-TGF-beta complex is unable to bind to the TGF-beta receptor and is biologically inactive. TGF-. beta.s are typically released (and active) from the complex by a variety of mechanisms including, for example, interaction with thrombospondin-1 or plasmin. TGF-. beta.1 transduces signals through two highly conserved single transmembrane serine/threonine kinases, the type I (ALK5) and the type II TGF-. beta.receptor. Once ligand-induced oligomerization, type II receptors hyperphosphorylate serine/threonine residues in the ALK5GS region, leading to activation of ALK5 by creating a binding site for Smad proteins. The activated ALK5 in turn phosphorylates Smad2 and Smad3 proteins at the C-terminal SSXS-motif, causing them to dissociate from the receptor and form heterocomplexes with Smad4 (heterocomplex). Smad complexes are readily localized to the nucleus, assemble with specific DNA-binding cofactors and coregulators, and ultimately activate transcription of extracellular matrix components and matrix-degrading protease inhibitors.
Hyperactive TGF- β signaling pathway is responsible for many human diseases such as excessive deposition of extracellular matrix, abnormally high levels of inflammatory response, fibrotic disorders, and progressive cancer. In the advanced stages of various cancers, tumor cells and stromal cells within the tumor typically overexpress TGF-. beta.s. This causes stimulation of angiogenesis and cellular motility, suppression of the immune system, and increased interaction of tumor cells with the extracellular matrix (e.g., Hojo, M. et al, Nature 397: 530-. Therefore, tumor cells become more invasive, metastasizing to distal organs such as, for example, Maehara, Y, et al, J.Clin.Oncol.17:607-614 (1999); picon, A. et al, Cancer epidemic. Biomarkers Prev.7:497-504 (1998)).
A number of experimental animal studies have demonstrated associations between TGF-. beta.glomerular expression and fibrosis, including the Thy-1 rat model of proliferative glomerulonephritis, the rabbit anti-GBM glomerulonephritis and the 5/6 nephrectomized rat model of focal segmental glomerulosclerosis, recently reviewed (e.g., Bitzer, M. et al, Kidney Blood Press. Res.21:1-12 (1998)). Neutralizing antibodies to TGF-. beta.improve glomerular histology in a model of Thy-1 nephritis (e.g., Border, W.A. et al, Nature 346:371-374 (1990)).
TGF-. beta.1 and its receptors are overexpressed in damaged blood vessels and fibroproliferative vascular injury, causing overproduction of extracellular matrix (e.g., Saltis, J. et al, Clin. Exp. Pharmacol. physiol.23:193- "200 (1996); McCaffrey, T.A. et al, J.Clin. invest.96: 2667-" 2675 (1995)).
TGF-. beta.2 levels are increased in most aqueous humor tumor eyes with juvenile glaucoma and in almost half of eyes with Primary Open Angle Glaucoma (POAG) (e.g., Picht, G., et al, Graefes Arch. Clin. exp. Ophthalmol.239:199- "207 (2001)).
It would therefore be desirable to develop inhibitors of TGF- β family members to prevent and/or treat diseases that include such signaling pathways. Published patent applications for modulators (e.g., antagonists) of TGF- β family member receptors include WO2004111046, WO2012000595, WO2012002680, WO2013009140, WO 2016106266.
In order to achieve better therapeutic effects and better meet the market demand, the inventors hope to develop a new generation of TGF-beta receptor kinase inhibitor with high efficiency and low toxicity. The invention provides a TGF-beta receptor kinase inhibitor with a novel structure, and finds that the compound with the structure has good activity and shows excellent TGF-beta receptor inhibition effect.
Disclosure of Invention
The invention aims to provide a compound shown in a general formula (I) or a tautomer, a meso form, a racemate, an enantiomer, a diastereomer or a mixture form thereof or a pharmaceutically acceptable salt form thereof,
Figure BDA0001465460610000021
wherein:
R1selected from-S (O)mR4Cycloalkyl and heterocyclyl, wherein said cycloalkyl and heterocyclyl are each independently optionally substituted by a group selected from hydrogen, alkyl, alkoxy, halogen, oxo, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, -OR5、-C(O)R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7、-NR6R7and-S (O)mNR6R7Is substituted with one or more substituents of (1);
R2the same or different, and each is independently selected from the group consisting of a hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, and heterocyclyl;
R3the same or different, and each is independently selected from the group consisting of a hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, and nitro;
R4selected from the group consisting of hydrogen atoms, alkyl groups, haloalkyl groups, hydroxyalkyl groups, amino groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
R5selected from the group consisting of hydrogen atoms, alkyl groups, haloalkyl groups, hydroxyalkyl groups, amino groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
R6and R7Each independently selected from the group consisting of hydrogen atoms, alkyl groups, haloalkyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
n is 0, 1,2, 3 or 4;
p is 0, 1,2 or 3; and is
m is 0, 1 or 2.
In a preferred embodiment of the present invention, the compound represented by the general formula (I) wherein R is3Is a hydrogen atom.
In a preferred embodiment of the present invention, the compound represented by the general formula (I) is a compound represented by the general formula (II):
Figure BDA0001465460610000031
or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof,
wherein:
g is selected from CH, N, S (O)mAnd O;
when G is S (O)mOr O is, R8Is absent;
when G is CH or N, R8Selected from the group consisting of hydrogen, alkyl, alkoxy, halogen, oxo, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, -OR5、-C(O)R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7、-NR6R7and-S (O)mNR6R7
s is 1 or 2;
r is 0, 1 or 2;
R2、R5~R7m and n are as defined in formula (I).
In a preferred embodiment of the present invention, the compound represented by the general formula (II) is a compound represented by the general formula (III):
Figure BDA0001465460610000041
or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof,
wherein:
R9selected from the group consisting of alkyl, alkoxy, halogen, oxo, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl, -OR5、-C(O)R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7、-NR6R7and-S (O)mNR6R7
R2、R5~R7N, m, s and r are as defined in formula (II).
In a preferred embodiment of the present invention, the compound represented by the general formula (I) is a compound represented by the general formula (III-A):
Figure BDA0001465460610000051
or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof,
wherein:
R2n, s and r are as defined in formula (II) or (III).
In a preferred embodiment of the present invention, the compound represented by the general formula (I) wherein R is2Are the same or different and are each independently selected from the group consisting of a hydrogen atom, a halogen, and an alkyl group.
The compounds of the present invention include all conformational isomers thereof, such as cis and trans isomers; and all optical isomers and stereoisomers thereof and mixtures thereof. The compounds of the invention have asymmetric centers and thus exist as different enantiomers and diastereomers. The present invention relates to the use of the compounds of the present invention, and all pharmaceutical compositions and methods of treatment which may be employed and which contain them. The present invention relates to the use of all such isomers and mixtures thereof.
Typical compounds of the invention include, but are not limited to:
Figure BDA0001465460610000052
Figure BDA0001465460610000061
Figure BDA0001465460610000071
Figure BDA0001465460610000081
Figure BDA0001465460610000091
Figure BDA0001465460610000101
Figure BDA0001465460610000111
Figure BDA0001465460610000121
Figure BDA0001465460610000131
Figure BDA0001465460610000141
Figure BDA0001465460610000151
Figure BDA0001465460610000161
Figure BDA0001465460610000171
Figure BDA0001465460610000181
Figure BDA0001465460610000191
Figure BDA0001465460610000201
or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof.
The invention also relates to a compound shown in the general formula (I-A), which is an intermediate for synthesizing the compound shown in the general formula (I):
Figure BDA0001465460610000202
or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof,
wherein:
x is halogen;
R1、R2and n is as defined in formula (I).
Compounds of formula (I-A) include, but are not limited to:
Figure BDA0001465460610000203
Figure BDA0001465460610000211
Figure BDA0001465460610000221
Figure BDA0001465460610000231
another aspect of the present invention relates to a method of preparing a compound of formula (I), the method comprising:
Figure BDA0001465460610000232
the compound of the general formula (I-A) and the compound of the general formula (I-B) are subjected to a Suzuki reaction under alkaline conditions in the presence of a catalyst to obtain the compound of the general formula (I),
wherein:
G1is composed of
Figure BDA0001465460610000241
X is halogen, preferably bromine;
R1~R3n and p are as defined in formula (I).
Another aspect of the present invention relates to a method of preparing a compound of formula (III), comprising:
Figure BDA0001465460610000242
a compound of the formula (III-A) or a hydrochloride thereof and R9-G2To give a compound of the general formula (III),
wherein:
G2selected from halogen, amino, -OC (O) R10Hejia teaA silicon-based group; wherein R is10Is a hydrogen atom or an alkyl group;
R2、R9n, s and r are as defined in formula (III).
Another aspect of the present invention relates to a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients. The invention also relates to a method for preparing the composition, which comprises mixing the compound shown in the general formula (I) or the tautomer, the mesomer, the racemate, the enantiomer, the diastereomer or the mixture form thereof, or the pharmaceutically acceptable salt thereof with one or more pharmaceutically acceptable carriers, diluents or excipients.
The invention further relates to the use of a compound of general formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for the manufacture of a medicament for inhibiting the TGF-beta and/or activin (in particular human TGF-beta and/or activin) signalling pathway.
The invention further relates to a compound shown in the general formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture form thereof, or a pharmaceutically acceptable salt or a pharmaceutical composition containing the compound, and an application of the compound in preparing medicines for treating, preventing or reducing the metastasis of tumor cells (particularly human tumor cells).
The invention further relates to the use of a compound of general formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for the manufacture of a medicament for the treatment, prevention or reduction of cancer mediated by TGF- β overexpression, in particular for the treatment, prevention or reduction of cancer mediated by TGF- β overexpression by inhibiting the human TGF- β signalling pathway.
The invention further relates to the use of a compound of general formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising same, for the preparation of a medicament for the treatment, prevention or alleviation, particularly of a human, of a disease selected from the group consisting of: vascular injury, glomerulonephritis, diabetic nephropathy, lupus nephritis, hypertension-induced nephropathy, renal interstitial fibrosis, renal fibrosis caused by drug exposure complications, HIV-related nephropathy, graft nephropathy, liver fibrosis of various etiologies, liver dysfunction attributable to infection, alcohol-induced hepatitis, cystic fibrosis, interstitial lung disease, acute lung injury, adult respiratory distress syndrome, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, lung disease caused by infectious or toxic factors, post-infarction cardiac fibrosis, congestive heart failure, dilated cardiomyopathy, myocarditis, intimal thickening, vascular stenosis, vascular remodeling caused by hypertension, pulmonary hypertension, coronary restenosis, peripheral arterial restenosis, carotid restenosis, stent-induced restenosis, atherosclerosis, coronary heart disease, peripheral arterial restenosis, coronary artery restenosis, stent-induced restenosis, coronary heart disease, peripheral arterial restenosis, hypertension, coronary heart disease, ocular scarring, corneal scarring, proliferative vitreoretinopathy, glaucoma, ocular hypertension, excessive or hypertrophic dermal scarring or keloid formation that occurs during wound healing from trauma or surgical wounds, peritoneal and subcutaneous adhesions, scleroderma, fibrosis sclerosis, progressive systemic sclerosis, dermatomyositis, polymyositis, arthritis, osteoporosis, ulcers, reduced nervous system function, male erectile dysfunction, pelonetz, duplet's contracture, alzheimer's disease, raynaud's syndrome, radiation-induced fibrosis, thrombosis, tumor metastatic growth, multiple myeloma, melanoma, glioma, glioblastoma, leukemia, sarcoma, leiomyoma, mesothelioma, breast cancer, cervical cancer, lung cancer, gastric cancer, rectal cancer, colon cancer, pancreatic cancer, liver cancer, pancreatic cancer, bone loss of function, bone loss of muscle, bone marrow, muscle loss of muscle mass, muscle loss, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, and liver cancer.
The present invention further relates to a method for treating, preventing or reducing metastasis of human tumor cells, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same.
The present invention further relates to a method of treating, preventing or reducing cancer mediated by TGF- β overexpression, in particular by inhibiting the TGF- β signalling pathway, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
The invention further relates to a method for the treatment, prevention or alleviation (in particular of humans) of a disease selected from the group consisting of: vascular injury, glomerulonephritis, diabetic nephropathy, lupus nephritis, hypertension-induced nephropathy, renal interstitial fibrosis, renal fibrosis caused by drug exposure complications, HIV-related nephropathy, graft nephropathy, liver fibrosis of various etiologies, liver dysfunction attributable to infection, alcohol-induced hepatitis, cystic fibrosis, interstitial lung disease, acute lung injury, adult respiratory distress syndrome, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, lung disease caused by infectious or toxic factors, post-infarction cardiac fibrosis, congestive heart failure, dilated cardiomyopathy, myocarditis, intimal thickening, vascular stenosis, vascular remodeling caused by hypertension, pulmonary hypertension, coronary restenosis, peripheral arterial restenosis, carotid restenosis, stent-induced restenosis, atherosclerosis, coronary heart disease, peripheral arterial restenosis, coronary artery restenosis, stent-induced restenosis, coronary heart disease, peripheral arterial restenosis, hypertension, coronary heart disease, ocular scarring, corneal scarring, proliferative vitreoretinopathy, glaucoma, ocular hypertension, excessive or hypertrophic dermal scarring or keloid formation that occurs during wound healing from trauma or surgical wounds, peritoneal and subcutaneous adhesions, scleroderma, fibrosis sclerosis, progressive systemic sclerosis, dermatomyositis, polymyositis, arthritis, osteoporosis, ulcers, reduced nervous system function, male erectile dysfunction, pelonetz, duplet's contracture, alzheimer's disease, raynaud's syndrome, radiation-induced fibrosis, thrombosis, tumor metastatic growth, multiple myeloma, melanoma, glioma, glioblastoma, leukemia, sarcoma, leiomyoma, mesothelioma, breast cancer, cervical cancer, lung cancer, gastric cancer, rectal cancer, colon cancer, pancreatic cancer, liver cancer, pancreatic cancer, bone loss of function, bone loss of muscle, bone marrow, muscle loss of muscle mass, muscle loss, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, and liver cancer, which comprises administering to a patient in need thereof a therapeutically effective amount of a compound represented by the general formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
The invention further relates to a compound shown in the general formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof or a pharmaceutical composition containing the same, which acts as a medicament.
The invention further relates to a compound represented by the general formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition containing the same, which acts as a TGF-beta receptor kinase inhibitor.
The invention further relates to a compound shown in the general formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof or a pharmaceutical composition containing the compound, which is used for treating, preventing or reducing the metastasis of tumor cells (particularly human tumor cells).
The invention further relates to a compound shown in the general formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof or a pharmaceutical composition containing the same, which is used for treating, preventing or reducing cancers mediated by TGF-beta overexpression, particularly treating, preventing or reducing the cancers mediated by the TGF-beta overexpression by inhibiting a TGF-beta signaling pathway.
The invention further relates to a compound of general formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising same, for use in the treatment, prevention, or alleviation (particularly of humans) of a disease selected from the group consisting of: vascular injury, glomerulonephritis, diabetic nephropathy, lupus nephritis, hypertension-induced nephropathy, renal interstitial fibrosis, renal fibrosis caused by drug exposure complications, HIV-related nephropathy, graft nephropathy, liver fibrosis of various etiologies, liver dysfunction attributable to infection, alcohol-induced hepatitis, cystic fibrosis, interstitial lung disease, acute lung injury, adult respiratory distress syndrome, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, lung disease caused by infectious or toxic factors, post-infarction cardiac fibrosis, congestive heart failure, dilated cardiomyopathy, myocarditis, intimal thickening, vascular stenosis, vascular remodeling caused by hypertension, pulmonary hypertension, coronary restenosis, peripheral arterial restenosis, carotid restenosis, stent-induced restenosis, atherosclerosis, coronary heart disease, peripheral arterial restenosis, coronary artery restenosis, stent-induced restenosis, coronary heart disease, peripheral arterial restenosis, hypertension, coronary heart disease, ocular scarring, corneal scarring, proliferative vitreoretinopathy, glaucoma, ocular hypertension, excessive or hypertrophic dermal scarring or keloid formation that occurs during wound healing from trauma or surgical wounds, peritoneal and subcutaneous adhesions, scleroderma, fibrosis sclerosis, progressive systemic sclerosis, dermatomyositis, polymyositis, arthritis, osteoporosis, ulcers, reduced nervous system function, male erectile dysfunction, pelonetz, duplet's contracture, alzheimer's disease, raynaud's syndrome, radiation-induced fibrosis, thrombosis, tumor metastatic growth, multiple myeloma, melanoma, glioma, glioblastoma, leukemia, sarcoma, leiomyoma, mesothelioma, breast cancer, cervical cancer, lung cancer, gastric cancer, rectal cancer, colon cancer, pancreatic cancer, liver cancer, pancreatic cancer, bone loss of function, bone loss of muscle, bone marrow, muscle loss of muscle mass, muscle loss, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, and liver cancer.
The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Oral compositions may be prepared according to any method known in the art for preparing pharmaceutical compositions, and such compositions may contain one or more ingredients selected from the group consisting of: sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide a pleasant to the eye and palatable pharmaceutical preparation. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents.
Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil. The oil suspension may contain a thickener. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable preparation. These compositions can be preserved by the addition of antioxidants.
Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent or one or more preservatives. Suitable dispersing or wetting agents and suspending agents are illustrative of the examples given above. Other excipients, for example sweetening, flavoring and coloring agents, may also be present. These compositions were preserved by the addition of an antioxidant.
The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oil phase may be a vegetable oil. Suitable emulsifiers may be naturally occurring phospholipids. The emulsions may also contain sweetening agents, flavouring agents, preservatives and antioxidants. Such formulations may also contain a demulcent, a preservative, a colorant and an antioxidant.
The pharmaceutical compositions of the present invention may be in the form of a sterile injectable aqueous solution. The sterile injectable preparation may be a sterile injectable oil-in-water microemulsion in which the active ingredient is dissolved in the oil phase.
The pharmaceutical compositions of the present invention may be in the form of sterile injectable aqueous or oleaginous suspensions for intramuscular and subcutaneous administration. The suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a parenterally-acceptable, non-toxic diluent or solvent. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
The compounds of the present invention may be administered in the form of suppositories for rectal administration. These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid in the rectum and therefore will melt in the rectum to release the drug.
As is well known to those skilled in the art, the dosage of a drug administered depends on a variety of factors, including, but not limited to: the activity of the particular compound employed, the age of the patient, the weight of the patient, the health condition of the patient, the behavior of the patient, the diet of the patient, the time of administration, the mode of administration, the rate of excretion, the combination of drugs, and the like; in addition, the optimal treatment regimen, such as the mode of treatment, the daily amount of compound (I) of the formula or the type of pharmaceutically acceptable salt, can be verified according to conventional treatment protocols.
Detailed description of the invention
Unless stated to the contrary, terms used in the specification and claims have the following meanings.
The term "alkyl" refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing 1 to 20 carbon atoms, preferably an alkyl group containing 1 to 12 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylpentyl, 2, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, 2-heptyl-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylpentyl, 2-dimethylpentyl, 3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2, 3-dimethylhexyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 2-dimethylhexyl, 3-dimethylhexyl, 4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 2, 4-dimethylhexyl, 2,4-, 2, 2-diethylpentyl, n-decyl, 3-diethylhexyl, 2-diethylhexyl, and various branched isomers thereof. More preferred are lower alkyl groups having 1 to 6 carbon atoms, non-limiting examples of which include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl and the like. Alkyl groups may be substituted OR unsubstituted, and when substituted, substituents may be substituted at any available point of attachment, preferably independently optionally selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, -OR5、-C(O)R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7、-NR6R7and-S (O)mNR6R7Is substituted with one or more substituents.
The term "cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably from 3 to 10 carbon atoms, most preferably from 3 to 6 carbon atoms, for example 3,4, 5 or 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl and the like, with cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl being preferred; polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups.
The term "spirocycloalkyl" refers to a 5 to 20 membered polycyclic group sharing one carbon atom (referred to as a spiro atom) between monocyclic rings, which may contain one or more double bonds, but none of the rings have a completely conjugated pi-electron system. Preferably 6 to 14, more preferably 7 to 10. Spirocycloalkyl groups are classified into a single spirocycloalkyl group, a double spirocycloalkyl group or a multi spirocycloalkyl group, preferably a single spirocycloalkyl group and a double spirocycloalkyl group, according to the number of spiro atoms shared between rings. More preferably 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered. Non-limiting examples of spirocycloalkyl groups include:
Figure BDA0001465460610000291
the term "fused cyclic alkyl" refers to a 5 to 20 membered all carbon polycyclic group in which each ring in the system shares an adjacent pair of carbon atoms with other rings in the system, wherein one or more of the rings may contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system. Preferably 6 to 14, more preferably 7 to 10. They may be classified into bicyclic, tricyclic, tetracyclic or polycyclic fused ring alkyls according to the number of constituent rings, preferably bicyclic or tricyclic, more preferably 5-or 6-membered bicycloalkyl. Non-limiting examples of fused ring alkyl groups include:
Figure BDA0001465460610000301
the term "bridged cycloalkyl" refers to a 5 to 20 membered all carbon polycyclic group in which any two rings share two carbon atoms not directly attached, which may contain one or more double bonds, but none of the rings have a completely conjugated pi-electron system. Preferably 6 to 14, more preferably 7 to 10. They may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl groups, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic, depending on the number of constituent rings. Non-limiting examples of bridged cycloalkyl groups include:
Figure BDA0001465460610000302
the cycloalkyl ring may be fused to an aryl, heteroaryl or heterocycloalkyl ring, where the ring to which the parent structure is attached is cycloalkyl, non-limiting examples of which include indanyl, tetrahydronaphthyl, benzocycloheptanyl, and the like. Cycloalkyl groups may be optionally substituted OR unsubstituted, and when substituted, the substituents are preferably independently optionally selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, -OR5、-C(O)R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7、-NR6R7and-S (O)mNR6R7Is substituted with one or more substituents.
The term "heterocyclyl" refers to a saturated and/or partially unsaturated mono-or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms wherein one or more of the ring atoms is selected from nitrogen, oxygen, or S (O)m(wherein m is an integer from 0 to 2) but excludes the ring moiety of-O-O-, -O-S-, or-S-S-, the remaining ring atoms being carbon. Preferably 3 to 12 ring atoms, of which 1 to 4 are heteroatoms, for example 1,2, 3 or 4 are heteroatoms; more preferably heterocyclyl contains 3 to 10 ring atoms, most preferably heterocyclyl contains 3 to 6 ring atoms, for example 3,4, 5 or 6 ring atoms. Non-limiting examples of monocyclic heterocyclyl groups include oxetanyl, azetidinyl, tetrahydrofuryl, tetrahydropyranyl, pyrrolyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and
Figure BDA0001465460610000303
etc., preferably azetidinyl, oxetanyl, pyrrolyl and piperidinyl; polycyclic heterocyclic groups include spiro, fused and bridged heterocyclic groups.
The term "spiroheterocyclyl" refers to a 5-to 20-membered polycyclic heterocyclic group in which one atom (referred to as the spiro atom) is shared between monocyclic rings, and in which one or more ring atoms is selected from nitrogen, oxygen, or S (O)m(wherein m is an integer of 0 to 2) and the remaining ring atoms are carbon. It may contain one or more double bonds, but no ring has a completely conjugated pi-electron system. Preferably 6 to 14, more preferably 7 to 10. The spiro heterocyclic group is classified into a mono-spiro heterocyclic group, a di-spiro heterocyclic group or a multi-spiro heterocyclic group according to the number of spiro atoms shared between rings, and preferably into a mono-spiro cycloalkyl group and a di-spiro cycloalkyl group. More preferred are 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered mono spiroheterocyclic groups. Non-limiting examples of spiro heterocyclic groups include:
Figure BDA0001465460610000311
the term "fused heterocyclyl" refers to a 5 to 20 membered polycyclic heterocyclic group in which each ring in the system shares an adjacent pair of atoms with other rings in the system, one or more rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system in which one or more ring atoms is selected from nitrogen, oxygen or S (O)m(wherein m is an integer of 0 to 2) and the remaining ring atoms are carbon. Preferably 6 to 14, more preferably 7 to 10. They may be classified into bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic groups according to the number of constituent rings, preferably bicyclic or tricyclic, more preferably 5-or 6-membered bicyclic fused heterocyclic groups. Non-limiting examples of fused heterocyclic groups include:
Figure BDA0001465460610000312
the term "bridgeHeterocyclyl "means a 5-to 14-membered polycyclic heterocyclic group in which any two rings share two atoms not directly attached, which may contain one or more double bonds, but none of the rings have a completely conjugated pi-electron system in which one or more of the ring atoms is selected from nitrogen, oxygen or S (O)m(wherein m is an integer of 0 to 2) and the remaining ring atoms are carbon. Preferably 6 to 14, more preferably 7 to 10. 7 to 10 yuan. They may be classified into bicyclic, tricyclic, tetracyclic or polycyclic bridged heterocyclic groups according to the number of constituent rings, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of bridged heterocyclic groups include:
Figure BDA0001465460610000313
the heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring to which the parent structure is attached is heterocyclyl, non-limiting examples of which include:
Figure BDA0001465460610000321
the heterocyclyl group may be optionally substituted OR unsubstituted, and when substituted, the substituents are preferably one OR more groups independently optionally selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, -OR5、-C(O)R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7、-NR6R7and-S (O)mNR6R7Is substituted with one or more substituents.
The term "aryl" refers to a 6 to 14 membered all carbon monocyclic or fused polycyclic (i.e., rings which share adjacent pairs of carbon atoms) group which is a polycyclic (i.e., rings which carry adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 10 membered, such as phenyl and naphthyl. The aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring attached to the parent structure is an aryl ring, non-limiting examples of which include:
Figure BDA0001465460610000322
aryl groups may be substituted OR unsubstituted, and when substituted, the substituents are preferably one OR more groups independently optionally selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, -OR5、-C(O)R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7、-NR6R7and-S (O)mNR6R7Is substituted with one or more substituents.
The term "heteroaryl" refers to a heteroaromatic system comprising 1 to 4 heteroatoms, 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen. Heteroaryl is preferably 5 to 10 membered, more preferably 5 or 6 membered, for example furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, pyrazolyl, tetrazolyl and the like. The heteroaryl ring may be fused to an aryl, heterocyclyl or cycloalkyl ring, wherein the ring joined together with the parent structure is a heteroaryl ring, non-limiting examples of which include:
Figure BDA0001465460610000323
Figure BDA0001465460610000331
heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, are substitutedThe substituents are preferably one OR more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, -OR5、-C(O)R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7、-NR6R7and-S (O)mNR6R7Is substituted with one or more substituents.
The term "alkoxy" refers to-O- (alkyl) and-O- (unsubstituted cycloalkyl), wherein alkyl is as defined above. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy. Alkoxy may be optionally substituted OR unsubstituted, and when substituted, the substituents are preferably one OR more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, thiol, hydroxy, amino, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, -OR5、-C(O)R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7、-NR6R7and-S (O)mNR6R7Is substituted with one or more substituents.
The term "hydroxyalkyl" refers to an alkyl group substituted with a hydroxy group, wherein alkyl is as defined above.
The term "haloalkyl" refers to an alkyl group substituted with one or more halogens, wherein alkyl is as defined above.
The term "hydroxy" refers to an-OH group.
The term "halogen" refers to fluorine, chlorine, bromine or iodine.
The term "amino" refers to the group-NH2
The term "cyano" refers to — CN.
The term "nitro" means-NO2
The term "oxo" refers to ═ O.
"optional" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not. For example, "a heterocyclic group optionally substituted with an alkyl" means that an alkyl may, but need not, be present, and the description includes the case where the heterocyclic group is substituted with an alkyl and the heterocyclic group is not substituted with an alkyl.
"substituted" means that one or more, preferably up to 5, more preferably 1 to 3, hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (experimentally or theoretically) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable in combination with carbon atoms having unsaturated (e.g., olefinic) bonds.
"pharmaceutical composition" means a mixture containing one or more compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof in admixture with other chemical components, as well as other components such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of the active ingredient and exert biological activity.
"pharmaceutically acceptable salts" refers to salts of the compounds of the present invention which are safe and effective for use in the body of a mammal and which possess the requisite biological activity.
m and R5~R7As defined in formula (I).
Synthesis of the Compounds of the invention
In order to achieve the purpose of the invention, the invention adopts the following technical scheme:
scheme one
The invention relates to a method for preparing a compound shown as a general formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereoisomer or a mixture form thereof, or a pharmaceutically acceptable salt form thereof, which comprises the following steps:
Figure BDA0001465460610000341
a first step of reacting a compound of the general formula (I-1) with a compound of the general formula (I-2) under basic conditions to obtain a compound of the general formula (I-3);
secondly, reacting the compound of the general formula (I-3) with a halogenating reagent to obtain a compound of the general formula (I-A);
thirdly, carrying out Suzuki reaction on the compound of the general formula (I-A) and the compound of the general formula (I-B) under alkaline conditions and in the presence of a catalyst to obtain the compound of the general formula (I);
the reagents that provide basic conditions include organic bases including, but not limited to, triethylamine, N-diisopropylethylamine, N-butyllithium, lithium diisopropylamide, lithium bistrimethylsilylamide, potassium acetate, sodium t-butoxide, or potassium t-butoxide, and inorganic bases including, but not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium acetate, cesium carbonate, sodium hydroxide, and lithium hydroxide;
catalysts include, but are not limited to, palladium on carbon, raney nickel, tetrakis-triphenylphosphine palladium, palladium dichloride, palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride, 1' -bis (dibenzylphosphine) dichlorodiamantapalladium, or tris (dibenzylideneacetone) dipalladium, preferably [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride or tetrakis-triphenylphosphine palladium;
halogenated agents include, but are not limited to, liquid bromine, hydrogen bromide, N-bromosuccinimide (NBS), PBr3、POBr3Pyridine Perbromide Hydrobromide (PHP), Tetrabromocycloketone (TBCO), diethyl bromomalonate, tetrabutylammonium bromide, N-chlorosuccinimide, PCl3And POCl3
The above reaction is preferably carried out in a solvent including, but not limited to: acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, N-hexane, dimethyl sulfoxide, 1, 4-dioxane, water, N-dimethylformamide and a mixture thereof;
wherein:
G1is selected from
Figure BDA0001465460610000351
X is halogen, preferably bromine;
R1~R3n and p are as defined in formula (I).
Scheme two
The invention relates to a method for preparing a compound shown as a general formula (III) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, which comprises the following steps:
Figure BDA0001465460610000352
a compound of the formula (III-A) or a hydrochloride thereof and R9-G2Under alkaline conditions to give compounds of the general formula (III),
the reagents that provide basic conditions include organic bases including but not limited to triethylamine, pyridine, N-diisopropylethylamine, N-butyllithium, lithium diisopropylamide, lithium bistrimethylsilylamide, potassium acetate, sodium t-butoxide or potassium t-butoxide, preferably triethylamine or pyridine, and inorganic bases including but not limited to sodium hydride, potassium phosphate, sodium carbonate, potassium acetate, cesium carbonate, sodium hydroxide and lithium hydroxide;
the above reaction is preferably carried out in a solvent including, but not limited to: acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, N-hexane, dimethyl sulfoxide, 1, 4-dioxane, water, N-dimethylformamide and a mixture thereof;
wherein:
G2selected from halogen, amino, -OC (O) R10And a trimethylsilyl group; wherein R is10Is a hydrogen atom or an alkyl group;
R2、R9n, s and r are as defined in formula (III).
Detailed Description
Examples
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or/and Mass Spectrometry (MS). NMR shift (. delta.) of 10-6The units in (ppm) are given. NMR was measured using a Bruker AVANCE-400 NMR spectrometer using deuterated dimethyl sulfoxide (DMSO-d)6) Deuterated chloroform (CDCl)3) Deuterated methanol (CD)3OD), internal standard Tetramethylsilane (TMS).
MS was determined using a FINNIGAN LCQAD (ESI) mass spectrometer (manufacturer: Thermo, model: Finnigan LCQ advantage MAX).
HPLC was carried out using an Agilent 1200DAD high pressure liquid chromatograph (Sunfire C18150X 4.6mm column) and a Waters 2695-2996 high pressure liquid chromatograph (Gimini C18150X 4.6mm column).
Chiral HPLC analytical determination using LC-10A vp (Shimadzu) or SFC-analytical (Berger Instruments Inc.);
the thin layer chromatography silica gel plate adopts HSGF254 of tobacco yellow sea or GF254 of Qingdao, the specification of the silica gel plate used by Thin Layer Chromatography (TLC) is 0.15 mm-0.2 mm, and the specification of the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm.
The column chromatography generally uses 200-300 mesh silica gel of the Tibet Huanghai silica gel as a carrier.
Chiral preparative column chromatography used Prep Star SD-1(Varian Instruments Inc.) or SFC-multigram (Berger Instruments Inc.).
The CombiFlash rapid preparation instrument uses CombiFlash Rf200(TELEDYNE ISCO).
Average inhibition rate of kinase and IC50The values were determined with a NovoStar microplate reader (BMG, Germany).
Known starting materials of the present invention may be synthesized by or according to methods known in the art, or may be purchased from companies such as ABCR GmbH & Co.KG, Acros Organics, Aldrich Chemical Company, Shao Yuan Chemical technology (Accela ChemBio Inc), Darri Chemicals, and the like.
In the examples, the reaction can be carried out in an argon atmosphere or a nitrogen atmosphere, unless otherwise specified.
An argon atmosphere or nitrogen atmosphere means that the reaction flask is connected to a balloon of argon or nitrogen with a volume of about 1L.
The hydrogen atmosphere refers to a reaction flask connected with a hydrogen balloon with a volume of about 1L.
The pressure hydrogenation reaction used a hydrogenation apparatus of Parr 3916EKX type and a hydrogen generator of Qinglan QL-500 type or a hydrogenation apparatus of HC2-SS type.
The hydrogenation reaction was usually evacuated and charged with hydrogen and repeated 3 times.
The microwave reaction was carried out using a CEM Discover-S908860 type microwave reactor.
In the examples, the solution means an aqueous solution unless otherwise specified.
In the examples, the reaction temperature is, unless otherwise specified, from 20 ℃ to 30 ℃ at room temperature.
The monitoring of the progress of the reaction in the examples employed Thin Layer Chromatography (TLC), a developing solvent used for the reaction, a system of eluents for column chromatography used for purifying compounds and a developing solvent system for thin layer chromatography including: a: dichloromethane/methanol system, B: n-hexane/ethyl acetate system, C: petroleum ether/ethyl acetate system, D: acetone, E: dichloromethane/acetone system, F: ethyl acetate/dichloromethane system, G: ethyl acetate/dichloromethane/n-hexane, H: ethyl acetate/dichloromethane/acetone, the volume ratio of the solvent is adjusted according to the polarity of the compound, and a small amount of basic or acidic reagents such as triethylamine, acetic acid and the like can be added for adjustment.
Example 1
4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidine-1-carboxylic acid tert-butyl ester 1
Figure BDA0001465460610000371
First step of
4- (3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidine-1-carboxylic acid tert-butyl ester 1c
2-methyl-6- (1H-pyrazol-3-yl) pyridine 1a (800mg, 5mmol, prepared by the known method "Bioorganic and Medicinal Chemistry,2015,23(6),1260, 1275") was charged into a 100mL single-neck flask, 30mL of N, N-dimethylformamide was added, and tert-butyl (4-toluenesulfonyloxy) piperidine-1-carboxylate 1b (1.78g, 5mmol, prepared by the known method "Journal of Medicinal Chemistry,2012,55(22),9958, 9972") and cesium carbonate (3.2g, 10mmol) were added, and the reaction was stirred at 60 ℃ for 16 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, then added with 50mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 1c (700mg, white solid) in yield: 40.9 percent.
MS m/z(ESI):343.5[M+1]
Second step of
4- (4-bromo-3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidine-1-carboxylic acid tert-butyl ester 1d
1c (700mg, 2mmol) was dissolved in 10mL of dichloromethane, N-bromosuccinimide (356mg,2mmol) was added, and the reaction was stirred at room temperature for 16 hours. Concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent B to give the title compound 1d (800mg, white solid), yield: 95.0 percent.
MS m/s(ESI):423.0[M+1]
The third step
4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidine-1-carboxylic acid tert-butyl ester 1
1d (220mg, 0.5mmol), 6- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) benzo [ d ] thiazole 1e (204mg, 0.78mmol, prepared by the method disclosed in patent application "WO 2016090296"), [1,1 '-bis (diphenylphosphino) ferrocene ] dichloropalladium (36mg, 0.05mmol), potassium carbonate (138mg, 1mmol) and 1,1' -bis (diphenylphosphino) ferrocene (28mg, 0.05mmol) were dissolved in 10mL of a mixed solvent of 1, 4-dioxane and water (V: V4: 1), and the reaction was stirred at 80 ℃ for 12 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, added with 50mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 1(150mg, white solid), yield: and (3) 63.3%.
MS m/z(ESI):476.5[M+1]
1H NMR(400MHz,CDCl3)δ9.01(s,1H),8.08(d,1H),8.02(s,1H),7.66(s,1H),7.62-7.58(m,1H),7.50(d,1H),7.29(d,1H),7.19-7.11(m,1H),4.53-4.45(m,1H),4.40-4.29(m,2H),3.03-2.89(m,2H),2.69-2.56(m,3H),2.37-2.28(m,2H),2.09-1.97(m,2H),1.52(s,9H)。
Example 2
4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidine-1-sulfonamide 2
Figure BDA0001465460610000381
Figure BDA0001465460610000391
First step of
6- (3- (6-methylpyridin-2-yl) -1- (piperidin-4-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 2a
1(150mg, 0.31mmol) was added to 5mL of ethyl acetate, 0.5mL of a 4M solution of hydrogen chloride in 1, 4-dioxane was added dropwise, and the reaction was stirred for 2 hours. Saturated sodium bicarbonate solution was added to the reaction solution to pH 7-8, extracted with ethyl acetate (30mL × 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to give the title compound 2a (130mg, yellow oil), yield: 100 percent.
MS m/z(ESI):376.4[M+1]
Second step of
4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidine-1-sulfonamide 2
Crude 2a (100mg, 0.24mmol) and sulfonamide (138mg, 1.44mmol) were dissolved in 2mL pyridine and reacted with microwave at 130 ℃ for 30 min. Concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to give the title compound 2(28mg, orange solid), yield: 25.0 percent.
MS m/z(ESI):455.4[M+1]
1H NMR(400MHz,CD3OD)δ9.20(s,1H),8.11(s,1H),8.03(d,1H),7.98-7.96(d,1H),7.36-7.70(m,1H),7.47-7.44(dd,1H),7.36-7.34(d,1H),7.29-7.27(d,1H),4.39-4.46(m,1H),3.86-3.83(d,2H),2.94-2.87(m,2H),2.49(s,3H),2.34-2.23(m,4H)。
Example 3
6-3- (6-methylpyridin-2-yl) -1- (1- (methylsulfonyl) piperidin-4-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 3
Figure BDA0001465460610000392
Figure BDA0001465460610000401
2a (40mg, 0.11mmol) was dissolved in 5mL of dichloromethane, triethylamine (22mg, 0.22mmol) and methanesulfonyl chloride (15mg, 0.13mmol) were added, and the reaction was stirred for 3 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 3(20mg, white solid), yield: 33.8 percent.
MS m/z(ESI):454.5[M+1]
1H NMR(400MHz,CDCl3)δ9.03(s,1H),8.10(d,1H),8.01(s,1H),7.69(s,1H),7.62-7.58(m,1H),7.49(d,1H),7.29(d,1H),7.27-7.22(m,1H),4.52-4.28(m,1H),4.05-4.00(m,2H),3.10-3.00(m,2H),2.92(s,3H),2.81-2.66(m,3H),2.50-2.42(m,2H),2.33-2.22(m,2H)。
Examples 4 and 5
(1R,4R) -4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) cyclohexanecarboxamide 4
(1S,4S) -4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) cyclohexanecarboxamide 5
Figure BDA0001465460610000402
First step of
(1R,4R) -4- (p-toluenesulfonyloxy) cyclohexyl carboxylic acid methyl ester 4b
Methyl (1R,4R) -4-hydroxycyclohexanecarboxylate 4a (3.16g, 20mmol, prepared as disclosed in patent application "US 2009118287A 1") was dissolved in 20mL of pyridine, p-toluenesulfonyl chloride (4.2g, 22mmol) was added and the reaction stirred for 12 hours. The reaction solution was concentrated under reduced pressure, 50mL of water was added, extraction was performed with ethyl acetate (50mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to obtain the title compound 4B (3.5g, white solid), yield: 56.1 percent.
MS m/z(ESI):330.5[M+1]
Second step of
(1S,4S) -4- (3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) cyclohexanecarboxylic acid methyl ester 4c
1a (960mg, 6mmol) was charged into a 100mL single-neck flask, 30mL of N, N-dimethylformamide was added, 4b (2.8g, 9mmol) and cesium carbonate (3.9g, 12mmol) were added, and the reaction was stirred at 50 ℃ for 16 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, then added with 50mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 4c (1.0g, colorless oil), yield: 55.6 percent.
The third step
(1S,4S) -4- (4-bromo-3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) cyclohexanecarboxylic acid methyl ester 4d
4c (1g, 3.34mmol) was dissolved in 30mL of dichloromethane, N-bromosuccinimide (0.71mg, 4.01mmol) was added, and the reaction was stirred at room temperature for 16 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system B gave the title compound 4d (600mg, light yellow solid), yield: 47.5 percent
MS m/z(ESI):[M+1]
The fourth step
(1R,4R) -4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) cyclohexanecarboxylic acid methyl ester 4e
4d (300mg, 0.79mmol), 1e (414mg, 1.59mmol), [1,1 '-bis (diphenylphosphino) ferrocene ] dichloropalladium (58mg, 0.08mmol), potassium carbonate (219mg, 1.59mmol) and 1,1' -bis (diphenylphosphino) ferrocene (44mg, 0.08mmol) were dissolved in 11mL of a mixed solvent of 1, 4-dioxane and water (V: V ═ 10:1), and the reaction was stirred at 80 ℃ for 18 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, added with 50mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 4e (200mg, pale yellow viscous solid), yield: 58.3 percent.
MS m/z(ESI):449.5[M+1]
The fifth step
4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) cyclohexanecarboxylic acid 4f
4e (200mg, 0.46mmol) was dissolved in 10mL of a mixed solvent of tetrahydrofuran and methanol (V: V ═ 3:2), and 1.15mL of a 2M sodium hydroxide solution was added. The reaction was stirred for 2 hours. Most of the solvent was evaporated under reduced pressure, 15mL of water was added, pH was adjusted to 5-6 with 1M hydrochloric acid, a solid precipitated, filtered, and the filter cake was dried to give the title compound 4f (180mg, pale yellow solid), yield: 93.0 percent
MS m/z(ESI):419.5[M+1]
The sixth step
(1R,4R) -4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) cyclohexanecarboxamide 4
(1S,4S) -4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) cyclohexanecarboxamide 5
4f (180mg, 0.43mmol) was dissolved in 10mL of N, N-dimethylformamide, ammonium chloride (34.51mg, 0.65mmol), N, N-diisopropylethylamine (0.22mL, 1.29mmol), 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (99mg, 0.52mmol) and 1-hydroxybenzotriazole (65mg, 0.43mmol) were added, the reaction was stirred for 18 hours, 30mL of water was added, extraction was performed with ethyl acetate (30 mL. times.2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by high performance liquid chromatography to give the title compounds 4(40mg, white solid) and 5(20mg, white solid).
Example 4
MS m/z(ESI):418.5[M+1]
1H NMR(400MHz,CD3OD)δ9.19(s,1H),8.05(s,1H),8.00(d,1H),7.95(d,1H),7.71(t,1H),7.43(dd,1H),7.33(d,1H),7.27(d,1H),4.28-4.35(m,1H),2.48(s,3H),2.41-2.35(m,1H),2.33-2.27(m,2H),2.09-1.97(m,4H),1.80-1.68(m,2H)。
Example 5
MS m/z(ESI):418.5[M+1]
1H NMR(400MHz,CD3OD)δ9.18(s,1H),8.08(s,1H),8.03(d,1H),7.94(d,1H),7.71(t,1H),7.45(dd,1H),7.35(d,1H),7.26(d,1H),4.48-4.43(m,1H),2.62-2.55(m,1H),2.47(s,3H),2.43-2.35(m,2H),2.15-2.05(m,4H),1.85-1.76(m,2H)。
Example 6
(R) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 6
Figure BDA0001465460610000421
Figure BDA0001465460610000431
First step of
(R) -3- (3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 6b
1a (960mg, 6mmol) was charged in a 100mL single-neck flask, 30mL of N, N-dimethylformamide was added, and then (S) -3- (p-toluenesulfonyloxy) pyrrolidine-1-carboxylic acid tert-butyl ester 6a (3.07g, 9mmol, prepared by a known method "Bioorganic and Medicinal Chemistry Letters,2013,23(14), 4044-. The reaction was cooled to room temperature, 100mL of water was added, extraction was performed with ethyl acetate (100mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 6B (1.3g, colorless oil), yield: 66.0 percent.
MS m/z(ESI):329.1[M+1]
Second step of
(R) -3- (4-bromo-3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 6c
6b (1.3g,3.96mmol) was dissolved in 30mL of dichloromethane, N-bromosuccinimide (705mg, 3.96mmol) was added, and the reaction was stirred at room temperature for 16 hours. The reaction was concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 6c (1.5g, colorless oil), yield: 93.1 percent.
MS m/z(ESI):407.1[M+1]
The third step
(R) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 6
6c (640mg, 1.5mmol), 1e (640mg, 2.25mmol), tris (dibenzylideneacetone) dipalladium (274mg, 0.3mmol), potassium carbonate (414mg, 3mmol) and tricyclohexylphosphine (210mg, 0.75mmol) were dissolved in 6.6mL of a mixed solvent of 1, 4-dioxane and water (V: V ═ 10:1), and the reaction was stirred at 80 ℃ for 12 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, added with 20mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 6(380mg, white solid), yield: 55.0 percent.
MS m/z(ESI):462.5[M+1]
1H NMR(400MHz,CDCl3)δ8.97(s,1H),8.04(d,1H),8.00(s,1H),7.62(s,1H),7.51(t,1H),7.47(dd,1H),7.26(d,1H),7.10(d,1H),5.08(m,1H),3.92-3.50(m,4H),2.55(s,3H),2.50-2.43(m,2H),1.49(s,9H)。
Example 7
(R) -6- (3- (6-methylpyridin-2-yl) -1- (pyrrolidin-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 7
Figure BDA0001465460610000441
6(362mg, 0.78mmol) was added to 10mL ethyl acetate, 4mL of a 4M solution of hydrogen chloride in 1, 4-dioxane was added dropwise, the reaction was stirred for 1 hour and monitored by LC-MS to completion. Saturated sodium bicarbonate solution was added to the reaction solution to pH 7-8, extracted with ethyl acetate (30mL × 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to give the title compound 7(250mg, yellow solid), yield: 88.3 percent.
MS m/z(ESI):362.4[M+1]
1H NMR(400MHz,CD3OD)δ9.67(s,1H),8.32-8.28(m,3H),8.17(d,1H),7.86(d,1H),7.67(s,1H),7.58(d,1H),5.53-5.51(m,1H),4.203(d,1H),3.91-3.80(m,2H),3.65-3.59(m,1H),3.01(s,3H),2.77-2.71(m,1H),2.63-2.55(m,1H)。
Example 8
(R) -6- (1- (1- (cyclopropylsulfonyl) pyrrolidin-3-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 8
Figure BDA0001465460610000442
Figure BDA0001465460610000451
7(40mg, 0.11mmol) was dissolved in 2mL of dichloromethane, triethylamine (33mg, 0.22mmol) and cyclopropylsulfonyl chloride (19mg, 0.13mmol) were added, and the reaction was stirred for 1 hour. 10mL of water was added, dichloromethane (20 mL. times.2) was extracted, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title compound 8(30mg, white solid), yield: 58.0 percent.
MS m/z(ESI):466.6[M+1]
1H NMR(400MHz,CDCl3)δ9.01(s,1H),8.09(d,1H),8.02(d,1H),7.76(s,1H),7.60-7.55(m,1H),7.49(dd,1H),7.27(d,1H),7.16(d,1H),5.20-5.10(m,1H),4.01-3.95(m,2H),3.85-3.79(m,1H),3.71-3.63(m,1H),2.71-2.52(m,6H),1.27-1.23(m,2H),1.04-1.01(m,2H)。
Example 9
(R) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-sulfonamide
Figure BDA0001465460610000452
Figure BDA0001465460610000461
First step of
3-fluoro-2-methyl-6- (1H-pyrazol-3-yl) pyridine 9c
1H-pyrazole-3-boronic acid 9a (3.09g, 27.6mmol), 6-bromo-3-fluoro-2-methylpyridine 9b (6.30g, 33.1mmol), tetrakistriphenylphosphine palladium (1.6g, 1.38mmol), and 27.6mL of a 2M sodium carbonate solution were added to 120mL1, 4-dioxane, and heated under reflux for 16 hours. Cooled to room temperature, concentrated under reduced pressure, added 20mL of water, extracted with ethyl acetate (20mL × 3), the organic phases combined, dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, and the resulting residue purified by silica gel column chromatography with eluent system a to give the title compound 9c (2.6g, yellow oil), yield: 53.2 percent.
MS m/z(ESI):178.4[M+1]
Second step of
(R) -3- (3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 9d
9c (800mg, 4.5mmol) was charged into a 100mL single-neck flask, 30mL of N, N-dimethylformamide was added, 6a (2.32g, 6.8mmol) and cesium carbonate (2.94g, 9mmol) were further added, and the reaction was stirred at 60 ℃ for 16 hours. The reaction was cooled to room temperature, 100mL of water was added, extraction was performed with ethyl acetate (100 mL. times.3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the crude title compound 9d (1.52g, yellow oil) which was directly subjected to the next reaction without purification.
MS m/z(ESI):347.2[M+1]
The third step
(R) -3- (4-bromo-3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 9e
Crude 9d (1.52g,4.39mmol) was dissolved in 10mL of dichloromethane, N-bromosuccinimide (791mg, 4.39mmol) was added, and the reaction was stirred at room temperature for 16 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system B gave the title compound 9e (1.3g, colorless oil) in 69.9% yield.
MS m/z(ESI):425.1[M+1]
The fourth step
(R) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 9f
9e (1.3g, 3.1mmol), 1e (1.2g, 4.58mmol), tris (dibenzylideneacetone) dipalladium (0.56g, 0.6mmol), potassium carbonate (0.84g, 6.11mmol) and tricyclohexylphosphine (430mg, 1.53mmol) were dissolved in 33mL of a mixed solvent of 1, 4-dioxane and water (V: V ═ 10:1), and the reaction was stirred at 80 ℃ for 12 hours. The reaction was cooled to room temperature, concentrated under reduced pressure, added with 50mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 9f (1.4g, white solid), yield: 65.9 percent.
MS m/z(ESI):480.2[M+1]
The fifth step
(R) -6- (3- (5-fluoro-6-methylpyridin-2-yl) -1- (pyrrolidin-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole
9f (1.4g, 2.92mmol) was added to 3mL of dichloromethane, 3mL of a 4M solution of hydrogen chloride in 1, 4-dioxane was added dropwise, and the reaction was stirred for 2 hours. Saturated sodium bicarbonate solution was added to the reaction solution to pH 7-8, extracted with ethyl acetate (30mL × 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 9g (1.29g, yellow solid), yield: 73.3 percent.
MS m/z(ESI):380.1[M+1]
The sixth step
(R) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-sulfonamide 9
9g (77mg, 0.2mmol) and sulfonamide (117mg, 1.22mmol) were dissolved in 2mL pyridine and reacted with microwave at 120 ℃ for 30 minutes. Concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography with eluent system a to give the title compound 9(22mg, orange solid), yield: 22.7 percent.
MS m/z(ESI):459.4[M+1]
1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),8.25(d,1H),8.22(s,1H),7.99(d,1H),7.70-7.50(m,3H),6.94(s,2H),5.11(t,1H),3.73-3.60(m,1H),3.55-3.50(m,1H),3.48-3.42(m,1H),3.36-3.31(m,1H),2.46-2.41(m,2H),2.30(m,3H)。
Example 10
6-1- (1- (cyclopropylsulfonyl) piperidin-4-yl) 3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 10
Figure BDA0001465460610000481
2a (45mg, 0.11mmol) was dissolved in 5mL of dichloromethane, triethylamine (33mg, 0.33mmol) and cyclopropylsulfonyl chloride (19mg, 0.13mmol) were added, and the reaction was stirred for 1 hour. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 10(26mg, white solid), yield: 50.0 percent.
MS m/z(ESI):480.4[M+1]
1H NMR(400MHz,CD3OD)δ9.19(s,1H),8.10(s,1H),8.02(d,1H),7.96-7.94(m,1H),7.70(t,1H),7.45-7.43(mm,1H),7.34(d,1H),7.26(d,1H),4.51-4.44(m,1H),3.96-3.92(m,2H),3.19-3.12(m,2H),2.59-2.53(m,1H),2.47(s,3H),2.33-2.19(m,4H),1.10-1.06(m,4H)。
Example 11
1- (4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidin-1-yl) ethanone 11
Figure BDA0001465460610000482
Figure BDA0001465460610000491
2a (40mg, 0.11mmol) was dissolved in 5mL of dichloromethane, triethylamine (22mg, 0.22mmol) and acetic anhydride (16mg, 0.16mmol) were added, and the reaction was stirred for 1 hour. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 11(20mg, white solid), yield: 43.5 percent.
MS m/z(ESI):418.5[M+1]
1H NMR(400MHz,CDCl3)δ9.01(s,1H),8.08(d,1H),8.02(s,1H),7.66(s,1H),7.62-7.58(m,1H),7.49(d,1H),7.29(d,1H),7.27-7.22(m,1H),4.88-4.83(m,1H),4.60-4.52(m,1H),4.10-4.02(m,1H),3.31(t,1H),2.83(t,1H),2.69-2.58(m,3H),2.43-2.40(m,1H),2.36-2.30(m,1H),2.18(s,3H),2.13-2.03(m,2H)。
Example 12
6-1- (1- (ethylsulfonyl) piperidin-4-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 12
Figure BDA0001465460610000492
Dissolve 2a (45mg, 0.11mmol) in 5mL of dichloromethane, add 0.3mL of triethylamine and 2 drops of ethanesulfonyl chloride, and stir for 1 hour. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 12(28mg, white solid), yield: 54.9 percent.
MS m/z(ESI):468.4[M+1]
1H NMR(400MHz,CD3OD)δ9.19(s,1H),8.09(s,1H),8.02(d,1H),7.96-7.94(m,1H),7.70(t,1H),7.45-7.43(m,1H),7.33(d,1H),7.26(d,1H),4.51-4.44(m,1H),3.96-3.93(m,2H),3.15-3.08(m,4H),2.47(s,3H),2.32-2.16(m,4H),1.36(t,3H)。
Example 13
4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidine-1-carboxamide 13
Figure BDA0001465460610000501
Dissolve 2a (45mg, 0.11mmol) in 5mL of dichloromethane, add triethylamine (33mg, 0.33mmol) and 2 drops of trimethylsilyl isocyanate, and stir for 12 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 13(8mg, white solid), yield: 17.4 percent.
MS m/z(ESI):419.5[M+1]
1H NMR(400MHz,CD3OD)δ9.19(s,1H),8.08(s,1H),8.01(d,1H),7.94(d,1H),7.70(t,1H),7.45-7.43(m,1H),7.33(d,1H),7.26(d,1H),4.54-4.48(m,1H),4.23-4.19(m,2H),3.09-3.02(m,2H),2.47(s,3H),2.23-2.07(m,4H)。
Example 14
4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) -N, N-dimethylpiperidin-1-sulfonic acid
Amides 14
Figure BDA0001465460610000502
Figure BDA0001465460610000511
2(20mg, 0.04mmol) was dissolved in 1mL of N, N-dimethylformamide, cesium carbonate (28mg, 0.09mmol) was added, methyl iodide (0.01mL, 0.09mmol) was added dropwise, and the reaction was stirred for 18 hours. Concentrated under reduced pressure, added 10mL of water, extracted with ethyl acetate (20mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to give the title compound 14(6mg, off-white solid) in yield: 26.8 percent.
MS m/z(ESI):483.5[M+1]
1H NMR(400MHz,CD3OD)δ9.18(s,1H),8.07(s,1H),8.01(d,1H),7.94(d,1H),7.68(t,1H),7.45-7.42(m,1H),7.33(d,1H),7.25(d,1H),4.47-4.42(m,1H),3.87-3.84(m,2H),3.12-3.06(m,2H),2.86(s,6H),2.47(s,3H),2.25-2.20(m,4H)。
Example 15
4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) -N-methylpiperidine-1-sulfonamide 15
Figure BDA0001465460610000512
2(20mg, 0.04mmol) was dissolved in 1mL of N, N-dimethylformamide, cesium carbonate (28mg, 0.09mmol) was added, methyl iodide (14mg, 0.04mmol) was added dropwise at 0 ℃, the temperature was raised to 25 ℃, and the reaction was stirred for 2 hours. Concentrated under reduced pressure, added 10mL of water, extracted with ethyl acetate (20mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to give the title compound 15(8mg, off-white solid) in yield: 36.0 percent.
MS m/z(ESI):469.5[M+1]
1H NMR(400MHz,CD3OD)δ9.18(s,1H),8.08(s,1H),8.01(d,1H),7.94(d,1H),7.70(t,1H),7.45-7.43(m,1H),7.33(d,1H),7.26(d,1H),4.45-4.41(m,1H),3.85-3.82(m,2H),3.05-2.99(m,2H),2.68(s,3H),2.56(s,3H),2.27-2.19(m,4H)。
Example 16
(S) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 16
Figure BDA0001465460610000521
Using the synthetic route of example 6, the first step starting material 6a was replaced with tert-butyl (R) -3- (p-toluenesulphonic acid) pyrrolidine-1-carboxylate (prepared using the well-known method "Bioorganic and Medicinal Chemistry Letters,2013,23(14), 4044-.
MS m/z(ESI):462.4[M+1]
1H NMR(400MHz,CDCl3)δ8.98(s,1H),8.06(d,1H),7.99(s,1H),7.63(s,1H),7.55(t,1H),7.45(d,1H),7.26(d,1H),7.13(d,1H),5.07(m,1H),3.91-3.58(m,4H),2.60(s,3H),2.45(m,2H),1.47(s,9H)。
Example 17
(S) -6- (3- (6-methylpyridin-2-yl) -1- (pyrrolidin-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 17
Figure BDA0001465460610000522
Figure BDA0001465460610000531
16(250mg, 0.54mmol) was added to 2mL of dichloromethane, 2mL of a 4M solution of hydrogen chloride in 1, 4-dioxane was added dropwise, and the reaction was stirred for 12 hours. Saturated sodium bicarbonate solution was added to the reaction solution to pH 7-8, extracted with ethyl acetate (30mL × 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to give the title compound 17(220mg, yellow solid), yield: 100 percent.
MS m/z(ESI):362.4[M+1]
1H NMR(400MHz,CDCl3)δ8.97(s,1H),8.04(d,1H),7.99(d,1H),7.68(s,1H),7.51(t,1H),7.46(dd,1H),7.26(d,1H),7.09(d,1H),4.96(m,1H),3.44(dd,1H),3.37(m,1H),3.29(m,1H),3.07(m,1H),2.52(s,3H),2.40(m,1H),2.32(m,1H)。
Example 18
(S) -6- (3- (6-methylpyridin-2-yl) -1- (1- (methylsulfonyl) pyrrolidin-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 18
Figure BDA0001465460610000532
17(50mg, 0.1mmol) was dissolved in 2mL of dichloromethane, triethylamine (30mg, 0.3mmol) and methanesulfonyl chloride (20mg, 0.15mmol) were added, and the reaction was stirred for 16 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 18(20mg, white solid), yield: 45.4 percent.
MS m/z(ESI):440.4[M+1]
1H NMR(400MHz,CDCl3)δ8.98(s,1H),8.05(d,1H),8.00(s,1H),7.70(s,1H),7.55(t,1H),7.46(d,1H),7.25(d,1H),7.13(d,1H),5.08(m,1H),3.95(m,2H),3.72(m,1H),3.64(m,1H),2.97(s,3H),2.56(br,5H)。
Example 19
(S) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid methyl ester 19
Figure BDA0001465460610000541
17(100mg, 0.25mmol) was dissolved in 5mL of dichloromethane, triethylamine (127mg, 1.25mmol) and methyl chloroformate (47mg, 0.50mmol) were added, and the reaction was stirred for 16 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 19(70mg, white solid), yield: 65.1 percent.
MS m/z(ESI):420.5[M+1]
1H NMR(400MHz,CDCl3)δ8.97(s,1H),8.04(d,1H),7.99(s,1H),7.61(s,1H),7.53(t,1H),7.46(d,1H),7.26(d,1H),7.14(d,1H),5.08(m,1H),3.95(m,2H),3.74(s,3H),3.65(m,2H),2.56(m,5H)。
Example 20
(S) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid ethyl ester 20
Figure BDA0001465460610000542
Figure BDA0001465460610000551
17(100mg, 0.25mmol) was dissolved in 5mL of dichloromethane, triethylamine (127mg, 1.25mmol) and ethyl chloroformate (54mg, 0.50mmol) were added, and the reaction was stirred for 16 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 20(60mg, white solid), yield: 54.0 percent.
MS m/z(ESI):434.5[M+1]
1H NMR(400MHz,CDCl3)δ8.97(s,1H),8.04(d,1H),8.00(s,1H),7.62(s,1H),7.51(t,1H),7.47(d,1H),7.26(d,1H),7.09(d,1H),5.08(m,1H),4.17(m,2H),3.98-3.90(m,2H),3.74-3.62(m,2H),2.54-2.45(m,5H),1.28(t,3H)。
Example 21
(S) -1- (3- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidin-1-yl) ethanone 21
Figure BDA0001465460610000552
17(50mg, 0.1mmol) was dissolved in 2mL of dichloromethane, triethylamine (30mg, 0.3mmol) and acetic anhydride (20mg, 0.2mmol) were added, and the reaction was stirred for 2 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 21(35mg, white solid), yield: 87.5 percent.
MS m/z(ESI):404.4[M+1]
1H NMR(400MHz,CDCl3)δ8.98(s,1H),8.05(d,1H),7.99(s,1H),7.63(d,1H),7.54(m,1H),7.46(d,1H),7.27(t,1H),7.13(d,1H),5.14-5.06(m,1H),4.05(m,2H),3.83(m,1H),3.69(m,1H),2.59-2.47(m,5H),2.12(d,3H)。
Example 22
(S) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid isopropyl ester 22
Figure BDA0001465460610000561
17(80mg, 0.2mmol) was dissolved in 5mL of dichloromethane, triethylamine (81mg, 0.8mmol) and isopropyl chloride (49mg, 0.4mmol) were added, and the reaction was stirred for 16 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 22(50mg, white solid), yield: 54.4 percent.
MS m/z(ESI):448.4[M+1]
1H NMR(400MHz,CDCl3)δ8.98(s,1H),8.05(d,1H),8.00(s,1H),7.63(s,1H),7.54(t,1H),7.47(d,1H),7.26(d,1H),7.12(d,1H),5.08(m,1H),4.97(m,1H),3.69-3.55(m,4H),2.58-2.45(m,5H),1.28(d,6H)。
Example 23
(R) -6- (1- (1- (ethylsulfonyl) pyrrolidin-3-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 23
Figure BDA0001465460610000562
7(40mg, 0.11mmol) was dissolved in 2mL of dichloromethane, triethylamine (33mg, 0.22mmol) and ethylsulfonyl chloride (17mg, 0.13mmol) were added, and the reaction was stirred for 1 hour. 10mL of water was added, dichloromethane (20 mL. times.2) was extracted, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title compound 23(17mg, yellow solid), yield: 34.0 percent.
MS m/z(ESI):454.6[M+1]
1H NMR(400MHz,CD3OD)δ9.21(s,1H),8.11(s,1H),8.04(d,1H),7.97(d,1H),7.71(t,1H),7.46(dd,1H),7.36(d,1H),7.27(d,1H),5.19-5.14(m,1H),3.97-3.90(m,2H),3.77-3.73(m,1H),3.66-3.62(m,1H),3.21-3.15(q,2H),2.62-2.57(m,2H),2.48(s,3H),1.34(t,3H)。
Example 24
(R) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-sulfonamide 24
Figure BDA0001465460610000571
7(72mg, 0.2mmol) and sulfonamide (77mg, 0.8mmol) were dissolved in 3mL of 1, 4-dioxane, and 2mL of triethylamine was added to react at 100 ℃ for 5 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 24(15mg, yellow white solid), yield: 17.0 percent.
MS m/z(ESI):441.5[M+1]
1H NMR(400MHz,CD3OD)δ9.21(s,1H),8.11(s,1H),8.03(d,1H),7.97(d,1H),7.69(t,1H),7.46(dd,1H),7.32(d,1H),7.27(d,1H),5.18-5.14(m,1H),3.85-3.77(m,2H),3.65-3.61(m,1H),3.52-3.48(m,1H),2.60-2.55(m,2H),2.50(s,3H)。
Example 25
(R) -6- (3- (6-methylpyridin-2-yl) -1- (1- (methylsulfonyl) pyrrolidin-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 25
Figure BDA0001465460610000581
7(40mg, 0.11mmol) was dissolved in 2mL of dichloromethane, triethylamine (33mg, 0.22mmol) and methanesulfonyl chloride (15mg, 0.13mmol) were added, and the reaction was stirred for 1 hour. 10mL of water was added, dichloromethane (20 mL. times.2) was extracted, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title compound 25(20mg, yellow solid), yield: 41.0 percent.
MS m/z(ESI):440.5[M+1]
1H NMR(400MHz,CDCl3)δ9.00(s,1H),8.07(d,1H),8.03(s,1H),7.73(s,1H),7.55(t,1H),7.49(dd,1H),7.27(d,1H),7.13(d,1H),5.12-5.09(m,1H),3.99-3.92(m,2H),3.76-3.72(m,1H),3.69-3.65(m,1H),2.99(s,3H),2.61-2.56(m,2H),2.54(s,3H)。
Example 26
3- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) azetidine-1-carboxylic acid tert-butyl ester 26
Figure BDA0001465460610000582
Using the synthetic route of example 6, the first step starting material 6a was replaced with tert-butyl 3- (p-toluenesulfonyloxy) azetidine-1-carboxylate (prepared by the well-known procedure "Organic Letters,2014,16(7), 1992-.
MS m/z(ESI):448.4[M+1]
1H NMR(400MHz,CD3OD)δ9.19(s,1H),8.09(s,1H),8.02(d,1H),7.96-7.94(m,1H),7.70(t,1H),7.45-7.43(m,1H),7.36(d,1H),7.26(d,1H),5.34-5.28(m,1H),4.48-4.41(m,4H),2.47(s,3H),1.48(s,9H)。
Example 27
6- (1- (azetidin-3-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 27
Figure BDA0001465460610000591
26(110mg, 0.25mmol) was added to 12mL of ethyl acetate, and 1.2mL of a 4M solution of hydrogen chloride in 1, 4-dioxane was added dropwise thereto, followed by stirring and reacting for 1 hour. Saturated sodium bicarbonate solution was added to the reaction solution to pH 7-8, extracted with ethyl acetate (30mL × 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by high performance liquid chromatography to give the title compound 27(83mg, yellow solid), yield: 100 percent.
MS m/z(ESI):348.4[M+1]
1H NMR(400MHz,CD3OD)δ9.20(s,1H),8.06(s,1H),8.03(d,1H),7.99-7.97(m,1H),7.69(t,1H),7.46-7.44(m,1H),7.36(d,1H),7.27(d,1H),5.51-5.44(m,1H),4.57-4.44(m,4H),2.51(s,3H)。
Example 28
6- (3- (6-methylpyridin-2-yl) -1- (1- (methylsulfonyl) azetidin-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 28
Figure BDA0001465460610000592
27(70mg, 0.05mmol) was dissolved in 5mL of dichloromethane, 0.5mL of triethylamine and 0.05mL of methanesulfonyl chloride were added, and the reaction was stirred for 1 hour. Concentration under reduced pressure and purification of the resulting residue by high performance liquid chromatography gave the title compound 28(16mg, white solid), yield: 76.0 percent.
MS m/z(ESI):426.4[M+1]
1H NMR(400MHz,CD3OD)δ9.20(s,1H),8.09(s,1H),8.03(d,1H),7.97-7.95(m,1H),7.71(t,1H),7.46-7.44(mm,1H),7.40(d,1H),7.27(d,1H),5.40-5.33(m,1H),4.58-4.54(m,2H),4.45-4.41(m,2H),3.13(s,3H),2.46(s,3H)。
Example 29
1- (3- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) azetidin-1-yl) ethanone 29
Figure BDA0001465460610000601
27(18mg, 0.05mmol) was dissolved in 5mL of dichloromethane, 0.5mL of triethylamine and 0.05mL of acetic anhydride were added, and the reaction was stirred for 1 hour. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 29(5mg, yellow solid), yield: 75.0 percent.
MS m/z(ESI):390.5[M+1]
1H NMR(400MHz,CD3OD)δ9.20(s,1H),8.12(s,1H),8.03(d,1H),7.97-7.95(m,1H),7.70(t,1H),7.46-7.43(m,1H),7.34(d,1H),7.28(d,1H),5.41-5.34(m,1H),4.74(d,2H),4.54-4.45(m,2H),2.48(s,3H),1.97(s,3H)。
Example 30
6-3- (5-fluoro-6-methylpyridin-2-yl) -1- (1- (methylsulfonyl) piperidin-4-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 30
Figure BDA0001465460610000602
Figure BDA0001465460610000611
First step of
4- (3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidine-1-carboxylic acid tert-butyl ester 30a
9c (885mg, 5mmol) was charged into a 100mL single-neck flask, 10mL of N, N-dimethylformamide was added, 1b (1.78g, 5mmol) and cesium carbonate (3.2g, 10mmol) were added, and the reaction was stirred at 45 ℃ for 16 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, then added with 50mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 30a (720mg, white solid) in yield: 40.0 percent.
Second step of
4- (4-bromo-3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidine-1-carboxylic acid tert-butyl ester 30b
30a (720mg, 2mmol) was dissolved in 10mL of dichloromethane, N-bromosuccinimide (356mg,2mmol) was added, and the reaction was stirred at room temperature for 16 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system B gave the title compound 30B (700mg, white solid), yield: 80.0 percent.
The third step
4- (4- (benzo [ d ] thiazol-6-yl) -3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) piperidine-1-carboxylic acid tert-butyl ester 30c
30b (200mg, 0.46mmol), 1e (179mg, 0.68mmol), tris (dibenzylideneacetone) dipalladium (0.84g, 0.09mmol), potassium carbonate (138mg, 1mmol) and tricyclohexylphosphine (64mg, 0.23mmol) were dissolved in 5mL of a mixed solvent of 1, 4-dioxane and water (V: V ═ 10:1), and the reaction was stirred at 80 ℃ for 12 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, added with 50mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 30c (227mg, white solid) in yield: 100 percent.
The fourth step
6- (3- (5-fluoro-6-methylpyridin-2-yl) -1- (piperidin-4-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 30d
30c (227mg, 0.46mmol) was added to 20mL of dichloromethane, 5mL of trifluoroacetic acid was added dropwise, and the reaction was stirred for 12 hours. Saturated sodium bicarbonate solution was added to the reaction solution to pH 7-8, extracted with ethyl acetate (30mL × 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 30d (180mg, yellow solid), yield: 100.0 percent.
The fifth step
6-3- (5-fluoro-6-methylpyridin-2-yl) -1- (1- (methylsulfonyl) piperidin-4-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 30
30d (180mg, 0.46mmol) was dissolved in 5mL of dichloromethane, triethylamine (139mg, 1.38mmol) and methanesulfonyl chloride (105mg, 0.9mmol) were added, and the reaction was stirred for 2 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 30(30mg, light yellow solid), yield: 11.1 percent.
MS m/z(ESI):472.4[M+1]
1H NMR(400MHz,CDCl3)δ9.15(s,1H),8.16-8.14(m,1H),7.94(s,1H),7.67(m,1H),7.60-7.58(m,1H),7.38-7.34(m,1H),7.26(m,1H),4.47(m,1H),4.0-3.97(m,2H),2.99-2.94(m,2H),2.87(s,3H),2.76-2.75(m,3H),2.39-2.36(m,2H),2.17-2.13(m,2H)。
Example 31
(R) -6- (3- (5-fluoro-6-methylpyridin-2-yl) -1- (1-methanesulfonyl) pyrrolidin-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 31
Figure BDA0001465460610000621
9g (60mg, 0.16mmol) was dissolved in 3mL of dichloromethane, triethylamine (64mg, 0.63mmol) and methanesulfonyl chloride (27mg, 0.24mmol) were added, and the reaction was stirred for 2 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 31(35mg, light yellow solid), yield: 48.4 percent.
MS m/z(ESI):458.4[M+1]
1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),8.27-8.25(m,2H),7.99(d,1H),7.67-7.50(m,3H),5.13(t,1H),3.90-3.80(m,1H),3.74-3.68(m,1H),3.60-3.45(m,2H),2.95(s,3H),2.46-2.41(m,2H),2.30(m,3H)。
Example 32
(R) -6- (1- (1- (cyclopropylsulfonyl) pyrrolidin-3-yl) -3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 32
Figure BDA0001465460610000631
9g (70mg, 0.18mmol) was dissolved in 3mL of dichloromethane, triethylamine (74mg, 0.74mmol) and cyclopropylsulfonyl chloride (39mg, 0.28mmol) were added, and the reaction was stirred for 2 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 32(40mg, light yellow solid), yield: 42.0 percent.
MS m/z(ESI):484.4[M+1]
1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),8.27-8.25(m,2H),7.99(d,1H),7.67-7.51(m,3H),5.15(t,1H),3.90-3.85(m,1H),3.79-3.74(m,1H),3.69-3.51(m,2H),2.78-2.73(m,1H),2.54-2.50(m,2H),2.30(d,3H),0.96-0.92(m,4H)。
Example 33
(S) -6- (3- (5-fluoro-6-methylpyridin-2-yl) -1- (1-methanesulfonyl) pyrrolidin-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 33
Figure BDA0001465460610000632
Figure BDA0001465460610000641
First step of
(S) -3- (3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 33b
9c (600mg, 3.4mmol) was put into a 100mL single-neck flask, 25mL of N, N-dimethylformamide was added, and tert-butyl (R) -3- (p-toluenesulfonyloxy) pyrrolidine-1-carboxylate 33a (1.74g, 5.1mmol, prepared by a known method "Bioorganic and Medicinal Chemistry Letters,2013,23(14),4044 and 4047") and cesium carbonate (2.2g, 6.8mmol) were further added, and the reaction was stirred at 60 ℃ for 2 hours. The reaction was cooled to room temperature, 100mL of water was added, extraction was performed with ethyl acetate (100mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 33B (700mg, yellow oil), yield: 60.0 percent.
MS m/z(ESI):347.1[M+1]
Second step of
(S) -3- (4-bromo-3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 33c
33b (700mg, 2.02mmol) was dissolved in 5mL of dichloromethane, N-bromosuccinimide (360mg, 2.02mmol) was added, and the reaction was stirred at room temperature for 16 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system B gave the title compound 33c (870mg, pale yellow oil), yield: 100.0 percent.
MS m/z(ESI):425.0[M+1]
The third step
(S) -3- (4- (benzo [ d ] thiazol-6-yl) -3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-1-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 33d
33c (385mg, 0.91mmol), 1e (355mg, 1.35mmol), tris (dibenzylideneacetone) dipalladium (167mg, 0.18mmol), potassium carbonate (252mg, 1.82mmol) and tricyclohexylphosphine (128mg, 0.45mmol) were dissolved in 5mL of a mixed solvent of 1, 4-dioxane and water (V: V ═ 10:1), and the reaction was stirred at 80 ℃ for 12 hours. The reaction was cooled to room temperature, concentrated under reduced pressure, added with 50mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to give the title compound 33d (440mg, brown oil) in yield: 100.0 percent.
MS m/z(ESI):480.1[M+1]
The fourth step
(S) -6- (3- (5-fluoro-6-methylpyridin-2-yl) -1- (pyrrolidin-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 33e
33d (440mg, 0.91mmol) was added to 3mL of dichloromethane, 3mL of a 4M solution of hydrogen chloride in 1, 4-dioxane was added dropwise, and the reaction was stirred for 2 hours. Saturated sodium bicarbonate solution was added to the reaction solution to pH 7-8, extracted with ethyl acetate (30mL × 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 33e (350mg, yellow solid), yield: 100.0 percent.
MS m/z(ESI):380.1[M+1]
The fifth step
(S) -6- (3- (5-fluoro-6-methylpyridin-2-yl) -1- (1-methanesulfonyl) pyrrolidin-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 33
33e (70mg, 0.18mmol) was dissolved in 3mL of dichloromethane, triethylamine (56mg, 0.55mmol) and methanesulfonyl chloride (31mg, 0.28mmol) were added, and the reaction was stirred for 2 hours. Concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to give the title compound 33(40mg, pale yellow solid), yield: 47.6 percent.
MS m/z(ESI):458.4[M+1]
1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),8.25-8.27(m,2H),7.99(d,1H),7.50-7.67(m,3H),5.13(t,1H),3.80-3.90(m,1H),3.68-3.74(m,1H),3.45-3.60(m,2H),2.95(s,3H),2.41-2.46(m,2H),2.30(m,3H)。
Example 34
(S) -6- (1- (1- (cyclopropylsulfonyl) pyrrolidin-3-yl) -3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 34
Figure BDA0001465460610000651
Figure BDA0001465460610000661
33e (70mg, 0.18mmol) was dissolved in 3mL of dichloromethane, triethylamine (56mg, 0.56mmol) and cyclopropylsulfonyl chloride (39mg, 0.28mmol) were added, and the reaction was stirred for 2 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system a gave the title compound 34(30mg, light yellow solid), yield: 33.7 percent.
MS m/z(ESI):484.4[M+1]
1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),8.25-8.27(m,2H),7.99(d,1H),7.51-7.67(m,3H),5.15(t,1H),3.85-3.90(m,1H),3.74-3.79(m,1H),3.51-3.69(m,2H),2.73-2.78(m,1H),2.50-2.54(m,2H),2.30(d,3H),0.92-0.96(m,4H)。
Example 35
6- (1-cyclopropyl-3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 35
Figure BDA0001465460610000662
First step of
6- (1-cyclopropyl-1H-pyrazol-3-yl) -3-fluoro-2-methylpyridine 35b
9c (1.58g, 8.92mmol), cyclopropylboronic acid 35a (1.53g, 17.83mmol), copper acetate monohydrate (3.56g, 17.83mmol), sodium carbonate (1.89g, 17.83mmol) and 2, 2-bipyridine (2.79g, 17.83mmol) were dissolved in 50mL of 1, 2-dichloroethane and the reaction was stirred at 50 ℃ for 16 hours. After the reaction was completed, filtration was performed, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to obtain the title compound 35B (455mg, yellow viscous substance), yield: 23.5 percent.
Second step of
6- (4-bromo-1-cyclopropyl-1H-pyrazol-3-yl) -3-fluoro-2-methylpyridine 35c
35b (344mg, 1.58mmol) was dissolved in 30mL of dichloromethane, N-bromosuccinimide (282mg, 1.58mmol) was added, and the mixture was stirred at room temperature for 1 hour. After the reaction was completed, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to obtain the title compound 35c (380mg, colorless oil), yield: 81.0 percent.
The third step
6- (1-cyclopropyl-3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 35
35c (75mg, 0.25mmol), 1e (99mg, 0.38mmol), potassium carbonate (70mg, 0.51mmol) and [1,1 '-bis (diphenylphosphino) ferrocene ] dichloropalladium (19mg, 0.025mmol), 1,1' -bis (diphenylphosphino) ferrocene (14mg, 0.025mmol) were dissolved in 5.5mL of a mixed solvent of 1, 4-dioxane and water (V/V ═ 10:1), and the reaction was stirred at 80 ℃ for 16 hours. The reaction solution was concentrated under reduced pressure, and the obtained residue was added with water, extracted with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by high performance liquid chromatography to obtain the title compound 35(46mg, white solid), yield: 51.7 percent.
MS m/z(ESI):351.4[M+1]
1H NMR(400MHz,CDCl3)δ8.97(s,1H),8.05(d,1H),7.97(s,1H),7.64(s,1H),7.46(dd,1H),7.33-7.31(m,1H),7.26(d,1H),3.76-3.71(m,1H),2.50(d,3H),1.29-1.25(m,2H),1.12-1.09(m,2H)。
Example 36
6- (1-cyclobutyl-3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 36
Figure BDA0001465460610000671
First step of
2- (1-cyclobutyl-1H-pyrazol-3-yl) -6-methylpyridine 36b
1a (2.01g, 12.6mmol) was dissolved in 100mL of N, N-dimethylformamide, followed by addition of toluene-4-sulfonic acid cyclobutyl ester 36a (4.29g, 18.9mmol, prepared by the method disclosed in patent application "WO 200993029") and cesium carbonate (8.23g, 25.3mmol), and heating to 60 ℃ for stirring reaction for 16 hours. Cooling to room temperature, addition of water, extraction with ethyl acetate, combination of the organic phases, washing with saturated sodium chloride solution, drying over anhydrous sodium sulfate, filtration, concentration of the filtrate under reduced pressure, and purification of the resulting residue by silica gel column chromatography with eluent system B to give the title compound 36B (1.81g, colorless oil), yield: 67.3 percent.
Second step of
2- (4-bromo-1-cyclobutyl-1H-pyrazol-3-yl) -6-methylpyridine 36c
36b (1.81g, 8.49mmol) was dissolved in 100mL of dichloromethane, N-bromosuccinimide (1.51g, 8.49mmol) was added, and the mixture was stirred at room temperature for 72 hours. After completion of the reaction, the reaction solution was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to obtain the title compound 36c (2.13g, yellow oil), yield: 85.9 percent.
The third step
6- (1-cyclobutyl-3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 36
36c (77mg, 0.26mmol), 1e (103mg, 0.40mmol), 1,1 '-bis (diphenylphosphino) ferrocene (15mg, 0.026mmol), potassium phosphate (168mg, 0.79mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (10mg, 0.013mmol) were dissolved in 10mL of 1, 4-dioxane, and the reaction was stirred at 80 ℃ for 3 hours. The reaction solution was concentrated under reduced pressure, and the obtained residue was added with water, extracted with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by high performance liquid chromatography to obtain the title compound 36(6mg, white solid), yield: 6.6 percent.
MS m/z(ESI):347.4[M+1]
1H NMR(400MHz,CDCl3)δ8.95(s,1H),8.03(d,1H),7.99(s,1H),7.69(s,1H),7.51-7.47(m,2H),7.25(d,1H),7.08(d,1H),4.96-4.88(m,1H),2.64-2.57(m,4H),2.55(s,3H),1.94-1.87(m,2H)。
Example 37
6- (1-cyclopropyl-3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 37
Figure BDA0001465460610000681
First step of
2- (1-cyclopropyl-1H-pyrazol-3-yl) -6-methylpyridine 37a
1a (700mg, 4.40mmol), 35a (756mg, 8.80mmol), copper acetate monohydrate (1.76g, 8.80mmol), sodium carbonate (933mg, 8.80mmol) and 2, 2-bipyridine (1.37g, 8.80mmol) were dissolved in 25mL1, 2-dichloroethane, and the reaction was stirred at 50 ℃ for 16 hours. After completion of the reaction, filtration and concentration of the filtrate under reduced pressure, the resulting residue was purified by silica gel column chromatography with eluent system B to obtain the title compound 37a (440mg, yellow dope), yield: 50.2 percent.
MS m/z(ESI):200.4[M+1]
Second step of
2- (4-bromo-1-cyclopropyl-1H-pyrazol-3-yl) -6-methylpyridine 37b
37a (330mg, 1.66mmol) was dissolved in 8mL of dichloromethane, N-bromosuccinimide (295mg, 1.66mmol) was added, and the mixture was stirred at room temperature for 1 hour. After completion of the reaction, 10mL of water was added to the reaction solution, extracted with dichloromethane (10mL × 3), the organic phases were combined, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to obtain the title compound 37b (335mg, yellow dope), yield: 72.6 percent.
MS m/z(ESI):280.1[M+1]
The third step
6- (1-cyclopropyl-3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 37
37b (85mg, 0.3mmol), 1e (94mg, 0.36mmol), tetrakistriphenylphosphine palladium (35mg, 0.03mmol) and 0.6mL of 2M sodium carbonate solution were dissolved in 3mL of ethylene glycol dimethyl ether and microwave reacted at 110 ℃ for 30 minutes. The reaction solution was concentrated under reduced pressure, and the obtained residue was added with water, extracted with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by high performance liquid chromatography to obtain the title compound 37(10mg, yellow solid), yield: 10.0 percent.
MS m/z(ESI):333.4[M+1]
1H NMR(400MHz,CD3OD)δ9.20(s,1H),8.06(s,1H),8.01(s,1H),7.95(d,1H),7.71(t,1H),7.43(dd,1H),7.33(d,1H),7.27(d,1H),3.83-3.78(m,1H),2.48(s,3H),1.28-1.24(m,2H),1.16-1.11(m,2H)。
Example 38
4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) tetrahydro-2H-thiopyran 1, 1-dioxide 38
Figure BDA0001465460610000691
First step of
4- (3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) tetrahydro-2H-thiopyran 1, 1-dioxide 38b
1a (146mg, 0.92mmol) was charged into a 100mL single neck flask, 30mL of N, N-dimethylformamide was added, 1-dioxide tetrahydro-2H-thiopyran-4-yl 4-tosylate 38a (280mg, 0.92mmol, prepared by the method disclosed in patent application "WO 2014036897A 1") and cesium carbonate (599mg, 1.84mmol) were added, and the reaction was stirred at 60 ℃ for 16 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, then added with 50mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 38B (250mg, white solid) in yield: 88.6 percent.
MS m/z(ESI):292.3[M+1]
Second step of
4- (4-bromo-3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) tetrahydro-2H-thiopyran 1, 1-dioxide 38c
38b (250mg, 0.86mmol) was dissolved in 20mL of dichloromethane, N-bromosuccinimide (229mg, 1.29mmol) was added, and the reaction was stirred at room temperature for 36 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel chromatography with eluent system B gave the title compound 38c (380mg, white solid) in yield: 100.0 percent.
MS m/z(ESI):372.3[M+1]
The third step
4- (4- (benzo [ d ] thiazol-6-yl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-1-yl) tetrahydro-2H-thiopyran 1, 1-dioxide 38
38c (50mg, 0.14mmol), 1e (70mg, 0.27mmol), [1,1 '-bis (diphenylphosphino) ferrocene ] dichloropalladium (10mg, 0.01mmol), potassium carbonate (37mg, 0.27mmol) and 1,1' -bis (diphenylphosphino) ferrocene (8mg, 0.01mmol) were added to 10mL of a mixed solvent of 1, 4-dioxane and water (V: V ═ 10:1), and the reaction was stirred at 80 ℃ for 12 hours under argon atmosphere. The reaction solution was cooled to room temperature, concentrated under reduced pressure, added with 50mL of water, extracted with ethyl acetate (30mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to obtain the title compound 38(5mg, white solid), yield: 7.6 percent.
MS m/z(ESI):425.4[M+1]
1H NMR(400MHz,CD3OD)δ9.20(s,1H),8.09(s,1H),8.02(d,1H),7.95(d,1H),7.70(t,1H),7.45-7.43(mm,1H),7.33(d,1H),7.26(d,1H),4.93-4.89(m,1H),3.82-3.75(m,1H),3.62-3.59(m,1H),3.19-3.12(m,2H),2.47(s,3H),2.34-2.21(m,4H)。
Example 39
6- (3- (6-methylpyridin-2-yl) -1- (tetrahydro-2H-pyran-4-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 39
Figure BDA0001465460610000701
Using the synthetic route of example 38, the first step starting material 38a was replaced with toluene-4-sulfonic acid-tetrahydro-2H-pyran-4-yl ester (prepared using the well known procedure "MedChemComm, 2012,3(9), 1077-.
MS m/z(ESI):377.4[M+1]
1H NMR(400MHz,CDCl3)δ8.97(s,1H),8.05(d,1H),8.01(s,1H),7.66(s,1H),7.54(t,1H),7.49(d,1H),7.26(d,1H),7.12(d,1H),4.65-4.50(m,1H),4.16(d,2H),3.59(t,2H),2.61(s,3H),2.29-2.23(m,2H),2.17-2.09(m,2H)。
Example 40
6- (3- (6-methylpyridin-2-yl) -1- (oxetan-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 40
Figure BDA0001465460610000711
Using the synthetic route of example 38, the starting material 38a from the first step was replaced with toluene-4-sulfonic acid oxetan-3-yl ester (prepared as disclosed in patent application "US 2015336982") to give the title product 40(31mg, yellow solid).
MS m/z(ESI):349.4[M+1]
1H NMR(400MHz,CDCl3)δ8.99(s,1H),8.07(d,1H),8.01(s,1H),7.89(s,1H),7.56-7.48(m,2H),7.28(d,1H),7.13(d,1H),5.66-5.61(m,1H),5.18-5.12(m,4H),2.59(s,3H)。
EXAMPLE 41
(R) -6- (3- (6-methylpyridin-2-yl) -1- (tetrahydrofuran-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 41
Figure BDA0001465460610000712
Using the synthetic route of example 38, the first step starting material 38a was replaced with (S) -toluene-4-sulfonic acid tetrahydrofuran-3-yl ester (prepared by a well-known method "European Journal of Organic Chemistry,2011,32, 6527-.
MS m/z(ESI):363.4[M+1]
1H NMR(400MHz,CDCl3)δ8.97(s,1H),8.05(d,1H),8.00(s,1H),7.71(s,1H),7.52(t,1H),7.46(d,1H),7.25(d,1H),7.13(d,1H),5.22-5.17(m,1H),4.25-4.20(m,2H),4.11-4.07(m,1H),3.98-3.92(m,1H),2.62(s,3H),2.58-2.09(m,2H)。
Example 42
(S) -6- (3- (6-methylpyridin-2-yl) -1- (tetrahydrofuran-3-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 42
Figure BDA0001465460610000721
Using the synthetic route for example 38, the first step starting material 38a was replaced with (R) -toluene-4-sulfonic acid tetrahydrofuran-3-yl ester (prepared using the well-known method "Journal of Medicinal Chemistry,2011,54(12), 4092-.
MS m/z(ESI):363.4[M+1]
1H NMR(400MHz,CD3OD)δ9.20(s,1H),8.07(s,1H),8.03(d,1H),7.96(d,1H),7.71(t,1H),7.46(dd,1H),7.36(d,1H),7.27(d,1H),5.20-5.14(m,1H),4.22-4.19(m,2H),4.15-4.11(m,1H),4.01-3.95(m,1H),2.61-2.51(m,2H),2.48(s,3H)。
Example 43
6- (1- (cyclopropylsulfonyl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 43
Figure BDA0001465460610000722
First step of
2- (4-bromo-1H-pyrazol-3-yl) -6-methylpyridine 43a
1a (5.59g, 35.08mmol) was dissolved in 200mL of dichloromethane, N-bromosuccinimide (6.24g, 35.08mmol) was added in portions, and the reaction was stirred at room temperature for 16 hours. Concentration under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system B gave the title compound 43a (8.01g, white solid), yield: 95.41 percent.
MS m/z(ESI):[M+1]
Second step of
2- (4-bromo-1- (tetrahydro-2H-pyran-2-yl) -1H-pyrazol-3-yl) -6-methylpyridine 43b
43a (8g, 33.6mmol), 3, 4-dihydro-2H-pyran (4.2g, 50.4mmol), 1mL of trifluoroacetic acid was added to 200mL of toluene, and the reaction was stirred at 80 ℃ for 12 hours. After the reaction was completed, the reaction mixture was washed with saturated sodium bicarbonate solution, the organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to obtain the title compound 43B (8.1g, white solid), yield: 74.8 percent.
MS m/z(ESI):[M+1]
The third step
6- (3- (6-methylpyridin-2-yl) -1- (tetrahydro-2H-pyran-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 43c
43b (3.41g, 10.58mmol), 1e (4.15g, 15.88mmol), [1,1 '-bis (diphenylphosphino) ferrocene ] dichloropalladium (0.77g, 1.06mmol), potassium carbonate (2.93g, 21.17mmol) and 1,1' -bis (diphenylphosphino) ferrocene (0.59g, 1.06mmol) were dissolved in 220mL of a mixed solvent of 1, 4-dioxane and water (V: V ═ 10:1), and the reaction was stirred at 80 ℃ for 48 hours. The reaction was cooled to room temperature, concentrated under reduced pressure, added 300mL of water, extracted with ethyl acetate (200mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title compound 43c (3.09g, white solid), yield: 77.6 percent.
MS m/z(ESI):[M+1]
The fourth step
6- (3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 43d
43c (3.09g, 8.21mmol) was added to 50mL of methanol, 10mL of a 4M solution of hydrogen chloride in 1, 4-dioxane was added dropwise, and the reaction was stirred for 12 hours. Saturated sodium bicarbonate solution was added to the reaction solution to pH 7-8, extracted with ethyl acetate (100mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to give the title compound 43d (2.11g, white solid), yield: 87.9 percent.
MS m/z(ESI):[M+1]
The fifth step
6- (1- (cyclopropylsulfonyl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 43
43d (30mg, 0.10mmol) was dissolved in 3mL of dichloromethane, triethylamine (30mg, 0.3mmol) and cyclopropylsulfonyl chloride (20mg, 0.15mmol) were added, and the reaction was stirred for 2 hours. Concentration under reduced pressure and purification of the resulting residue by high performance liquid chromatography gave the title compound 43(2mg, pale yellow solid), yield: 5.0 percent.
MS m/z(ESI):397.4[M+1]
1H NMR(400MHz,DMSO-d6)δ9.39(s,1H),8.69(s,1H),8.33(d,1H),8.01(dd,1H),7.92(t,1H),7.58-7.53(m,2H),7.31(d,1H),3.3-3.25(m,1H),2.34(s,3H),1.40-1.37(m,2H),1.29-1.25(m,2H)。
Example 44
6- (3- (6-methylpyridin-2-yl) -1- (methylsulfonyl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 44
Figure BDA0001465460610000741
43d (80mg, 0.27mmol) was dissolved in 2mL of pyridine, methanesulfonyl chloride (0.06mL, 0.82mmol) was added dropwise, and the reaction was stirred for 12 hours. Concentrated under reduced pressure, 20mL of a saturated copper sulfate solution was added, extraction was performed with ethyl acetate (10mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to obtain the title compound 44(88mg, off-white solid), yield: 81.1 percent.
MS m/z(ESI):371.4[M+1]
1H NMR(400MHz,CDCl3)δ9.02(s,1H),8.25(s,1H),8.09(s,1H),8.07(s,1H),7.65(t,1H),7.52-7.48(m,2H),7.20(d,1H),3.51(s,3H),2.50(s,3H)。
Example 45
6- (1- (ethylsulfonyl) -3- (6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 45
Figure BDA0001465460610000742
43d (81mg, 0.28mmol) was dissolved in 2mL of pyridine, ethanesulfonyl chloride (0.08mL, 0.83mmol) was added dropwise, and the reaction was stirred for 12 hours. Concentrated under reduced pressure, 20mL of a saturated copper sulfate solution was added, extraction was performed with ethyl acetate (10mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system a to obtain the title compound 45(90mg, off-white solid), yield: 82.3 percent.
MS m/z(ESI):385.4[M+1]
1H NMR(400MHz,CDCl3)δ9.01(s,1H),8.24(s,1H),8.10(s,1H),8.07(s,1H),7.64(t,1H),7.57(d,1H),7.51(dd,1H),7.18(d,1H),3.66(dd,2H),2.46(s,3H),1.41(t,3H)。
Example 46
6- (1- (cyclopropylsulfonyl) -3- (5-fluoro-6-methylpyridin-2-yl) -1H-pyrazol-4-yl) benzo [ d ] thiazole 46
Figure BDA0001465460610000751
Using the synthetic route to example 43, substituting the first step starting material 1a for 9c, the title product 46 was obtained (31mg, white solid).
MS m/z(ESI):415.3[M+1]
1H NMR(400MHz,CDCl3)δ9.03(s,1H),8.18(s,1H),8.10(s,1H),8.08(s,1H),7.68(dd,1H),7.51(d,1H),7.38(t,1H),2.98-2.93(m,1H),2.40(d,3H),1.58-1.56(m,2H),1.29-1.25(m,2H)。
Test example:
biological evaluation
Test example 1 determination of inhibitory Effect of the Compound of the present invention on TGF-. beta.RI kinase Activity
Inhibition of TGF β RI kinase activity in vitro was tested by the following method.
The inhibitory effect of the compounds of the present invention on the activity of TGF β RI kinase ALK5 was determined by the following experimental method:
enzymatic Activity detection Using TGF-. beta.RI kinase detection kit (V4093, Promega), 2. mu.l of a reaction buffer (40mM Tris pH7.5, 20mM MgCl. sub.L) was sequentially added to a 384-well plate (4514, Corning)20.1mg/ml BSA), 1. mu.l of a 3-fold gradient diluted compound dissolved in 5% DMSO, 2. mu.l of a mixed solution of ATP and TGF. beta. RI substrate polypeptide (ATP final concentration of 50. mu.M, substrate final concentration of 0.2. mu.g/. mu.L), and after reacting at 27 ℃ for 2.5 hours, 5. mu.l of ADP-Glo solution in the kit was added to each well, and allowed to stand at 27 ℃ for 40 minutes, 10. mu.l of a kinase assay reagent was added to each well, and allowed to stand at 27 ℃ for 30 minutes. Chemiluminescence signal values were detected using a Victor 3(PerkinElmer) multifunctional microplate reader. Calculating IC of the inhibitory effect of the compound on enzyme by Graphpad prism software according to each concentration of the compound and corresponding signal value50The value is obtained.
The biological activity of the compounds of the invention is determined by the above assay, the IC determined50The values are given in table 1 below.
TABLE 1 IC inhibition of TGF-. beta.RI kinase ALK5 Activity by Compounds of the invention50
Figure BDA0001465460610000752
Figure BDA0001465460610000761
And (4) conclusion: the compounds of the embodiment of the invention have obvious inhibition effect on the activity of TGF beta RI kinase ALK 5.
Test example 2 determination of inhibitory Effect of the Compounds of the present invention on VEGFR2 kinase Activity
The inhibition of VEGFR2 kinase activity in vitro was tested by the following method.
The following assay was used to determine the inhibitory effect of the compounds of the invention on VEGFR2 kinase activity:
the enzyme activity was measured using Z-
Figure BDA0001465460610000771
Mu.l of reaction buffer (50mM HEPES pH7.5, 10mM MgCl. sub.1) was sequentially added to a 384-well plate (4513, Corning) using the Kit of Kinase Assay Kit-Tyrosine 1Peptide (PV3190, Invitrogen)21mM EGTA, 0.05% BRIJ-35) and VEGFR2 substrate polypeptide (final enzyme concentration of 0.14 ng/. mu.L and final substrate concentration of 2. mu.M in the reaction system), 2.5. mu.L of 2-fold gradient diluted compound dissolved in 5% DMSO, 2.5. mu.L of ATP solution (final ATP concentration of 50. mu.M), 5. mu.L of detection reagent was added to each well after 2 hours of reaction at 25 ℃, and after 1 hour of reaction at 25 ℃, fluorescence signals with emission wavelengths of 445nm and 520nm were measured using a NOVOstar (BMG) multifunctional plate reader. Calculating IC of the inhibitory effect of the compound on enzyme by Graphpad prism software according to each concentration of the compound and corresponding signal value50The value is obtained.
The biological activity of the compounds of the invention is determined by the above assay, the IC determined50The values are given in Table 2 below.
TABLE 2 IC of inhibition of VEGFR2 kinase Activity by Compounds of the invention50
Example numbering IC50(nM)
3 2331
4 >10000
5 819
8 729
9 1320
16 2284
18 407
19 1687
23 386
39 3279
41 744
And (4) conclusion: the compounds of the embodiments of the invention do not show obvious inhibition effect on VEGFR2 kinase activity, which shows that the compounds of the embodiments of the invention have selective inhibition effect on TGF beta RI kinase.
Test example 3 determination of the inhibitory Effect of the Compound of the present invention on the p 38. alpha. kinase Activity
Inhibition of p38 α kinase activity in vitro was tested by the following method.
The following assay was used to determine the inhibition of p38 α kinase activity by the compounds of the present invention:
enzyme Activity detection Using p 38. alpha. kinase detection kit (V9591, Promega), 2. mu.L of reaction buffer (40mM Tris pH7.5, 20mM MgCl. sub.L) was sequentially added to a 384-well plate (4514, Corning)20.1mg/mL BSA), 1. mu.L of a 3-fold gradient diluted compound dissolved in 5% DMSO, 2. mu.L of a mixed solution of ATP and p38 substrate polypeptide (ATP final concentration of 50. mu.M, substrate final concentration of 0.2. mu.g/. mu.L), and after 2.5 hours of reaction at 27 ℃, 5. mu.L of ADP-Glo solution in the kit was added to each well, and the mixture was left at 27 ℃ for 40 minutes, 10. mu.L of a kinase assay reagent was added to each well, and left at 27 ℃ for 30 minutes. Chemiluminescence signal values were detected using a Victor 3(PerkinElmer) multifunctional microplate reader. Calculating IC of the inhibitory effect of the compound on enzyme by Graphpad prism software according to each concentration of the compound and corresponding signal value50The value is obtained.
The biological activity of the compounds of the invention is determined by the above assay, the IC determined50The values are given in Table 3 below.
TABLE 3 IC of inhibition of p38 alpha kinase activity by the compounds of the invention50
Example numbering IC50(nM)
2 591
3 750
4 890
5 1078
8 1295
16 2841
18 859
19 4358
22 517
23 655
31 732
33 892
34 2772
39 1754
41 595
43 >10000
And (4) conclusion: the compounds of the embodiment of the invention do not show obvious inhibition effect on the activity of p38 alpha kinase, which shows that the compounds of the embodiment of the invention have selective inhibition effect on TGF beta RI kinase.
Test example 4 inhibition assay of NIH3T3 cell proliferation by Compounds of the invention
The following in vitro assays were used to determine the inhibitory activity of the compounds of the invention on the proliferation of NIH3T3 cells.
The following assay was used to determine the inhibitory effect of the compounds of the invention on the proliferation of NIH3T3 cells:
100. mu.L of NIH3T3 cells (GNM6, cell bank of the national academy of sciences type culture Collection) were seeded in 96-well transparent bottom plates (3903, Corning) in a medium of DMEM (SH30243.01, GE) containing 10% FBS at a density of 2000 cells/well at 37 ℃ in 5% CO2Incubated under conditions overnight. After overnight incubation, each well was replaced with 90. mu.L of DMEM medium containing 0.5% FBS, then 10. mu.L of compound diluted in 3-fold gradient in DMEM medium containing 0.5% FBS was added and left at 37 ℃ with 5% CO2The cells were cultured in a cell incubator for 72 hours. Finally 50. mu.L of CellTiter-Glo (G7573, Promega) was added to each well and the chemiluminescent signal was read using a Victor3 microplate reader (Perkinelmer) after 10 minutes incubation at room temperature. IC of the compound was calculated from each concentration of the compound and the corresponding signal value using Graphpad Prism software50The value is obtained.
The biological activity of the compounds of the invention is determined by the above analysis and the calculated IC50The values are given in Table 4 below:
TABLE 4 Compounds of the inventionIC for inhibition of NIH3T3 cell proliferation50
Figure BDA0001465460610000791
Figure BDA0001465460610000801
And (4) conclusion: the compound of the invention has obvious inhibitory activity on NIH3T3 cell proliferation.
Test example 5 measurement of inhibitory Activity of the Compound of the present invention on Smad Signaling pathway of TGF-. beta.RI
The following in vitro assays were used to determine the inhibitory activity of the compounds of the invention on the Smad signaling pathway of TGF β RI.
The following experimental methods were used to determine the inhibitory activity of the compounds of the invention on the Smad signaling pathway of TGF β RI:
mu.L of HepG2 (TCtu 72, cell bank of the culture Collection for type culture Collection of China academy of sciences) cells were seeded in 96-well plates at a density of 2.5X10 per well with 10% FBS-containing EMEM medium (42360-4Cells/well, cells at 37 ℃ 5% CO2Incubated under conditions overnight. The fresh EMEM medium containing 10% FBS was replaced and transfected with 0.1. mu.g of 3TP-lux plasmid (11767, proeg Biotech (Beijing) Ltd.) per well and the cells were continued at 37 ℃ with 5% CO2Incubated under conditions for 24 hours. 90 μ L of EMEM medium containing 0.5% FBS was replaced per well and starved for 6 hours. Compounds were prepared as 20mM stock, diluted to 400X concentration with a 100% DMSO gradient, and diluted 40-fold with EMEM containing 0.5% FBS. The cell culture plate was removed, 10. mu.L of diluted compound or control (0.25% DMSO) was added to each well, mixed by gentle shaking, and left at 37 ℃ with 5% CO2Culturing in incubator for 18 hr, and adding 100 μ L detection reagent ONE-Glo into each wellTMLuciferase Assay (E6110, Promega) was left at room temperature for 10 minutes in the absence of light and the chemiluminescent signal was read using Victor3.0 (Perkinelmer). The concentration of each compound and the corresponding signal were determined by Graphpad Prism softwareValue calculation IC of Compounds50The value is obtained.
The biological activity of the compounds of the invention is determined by the above analysis and the calculated IC50The values are given in Table 5 below:
TABLE 5 Smad signaling pathway inhibition of TGF-beta RI by compounds of the invention IC50
Figure BDA0001465460610000802
Figure BDA0001465460610000811
Figure BDA0001465460610000821
And (4) conclusion: the compounds of the invention all have obvious inhibitory activity on Smad signaling pathway of TGF beta RI.
Pharmacokinetic evaluation
Test example 6 pharmacokinetic testing of the Compound of the invention
1. Abstract
Using rats as test animals, the drug concentrations in plasma at various times after gavage administration of the compound of example 2, the compound of example 24 and the compound of example 37 to rats were determined by LC/MS/MS method. The pharmacokinetic behavior of the compounds of the invention in rats was studied and their pharmacokinetic profile was evaluated.
2. Test protocol
2.1 test drugs
The compound of example 2, the compound of example 24 and the compound of example 37.
2.2 test animals
Healthy adult SD rats 12 with half male and female, divided into 3 groups on average, 4 per group, purchased from shanghai jestie laboratory animals ltd, animal production license number: SCXK (Shanghai) 2013 and 0006.
2.3 pharmaceutical formulation
A certain amount of the drug is weighed, and then 5% volume of DMSO, 5% volume of Tween 80 and 90% volume of physiological saline are added to prepare colorless clear transparent liquid of 0.2 mg/mL.
2.4 administration
SD rats are subjected to gastric lavage after being fasted overnight, the administration dose is 2.0mg/kg, and the administration volume is 10.0 mL/kg.
3. Operation of
Rats were gavaged with the compound of example 2, the compound of example 24 and the compound of example 37, and 0.2mL of the periorbital blood was collected before and after administration at 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 11.0, 24.0 hours, placed in heparinized tubes, centrifuged at 4 ℃ and 3500 rpm for 10 minutes to separate plasma, stored at-20 ℃ and fed 2 hours after administration.
Determining the content of the compound to be tested in rat plasma after the drug with different concentrations is administered by gastric lavage: rat plasma at each time post-dose was taken at 25. mu.L, added with camptothecin as an internal standard solution at 80. mu.L (100ng/mL), acetonitrile at 200. mu.L, vortexed for 5 minutes, centrifuged for 10 minutes (4000 rpm), and plasma samples were taken at 1.0. mu.L of supernatant for LC/MS/MS analysis.
4. Pharmacokinetic parameter results
The pharmacokinetic parameters of the compounds of the invention are as follows:
Figure BDA0001465460610000831
and (4) conclusion: the compound of the invention has better drug absorption and pharmacokinetic advantage.

Claims (16)

1. A compound of the general formula (I):
Figure FDA0002888084080000011
or a racemate, enantiomers or mixture thereof or pharmaceutically acceptable salts thereof,
wherein:
R1selected from-S (O)mR4、C3-6Cycloalkyl and 3 to 6 membered heterocyclyl, wherein said C is3-6Cycloalkyl and 3-to 6-membered heterocyclyl are each independently optionally selected from hydrogen atom, C1-6Alkyl, halogen, halogeno C1-6Alkyl, hydroxy C1-6Alkyl, -C (O) R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7and-S (O)mNR6R7Is substituted with one or more substituents of (1);
R2are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C1-6Alkyl, halo C1-6Alkyl and hydroxy C1-6An alkyl group;
R3are the same or different and are each independently selected from the group consisting of hydrogen, halogen and C1-6An alkyl group;
R4selected from hydrogen atoms, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino, C3-6Cycloalkyl and 3 to 6 membered heterocyclyl;
R5selected from hydrogen atoms, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino, C3-6Cycloalkyl and 3 to 6 membered heterocyclyl;
R6and R7Each independently is a hydrogen atom or C1-6An alkyl group;
n is 0, 1,2, 3 or 4;
p is 0, 1,2 or 3; and is
m is 2.
2. A compound of formula (I) according to claim 1, wherein R3Is a hydrogen atom.
3. The compound represented by the general formula (I) according to claim 1 or 2, which is a compound represented by the general formula (II):
Figure FDA0002888084080000021
or a racemate, enantiomers or mixture thereof or pharmaceutically acceptable salts thereof,
wherein:
g is selected from CH, N, S (O)mAnd O;
when G is S (O)mOr O is, R8Is absent;
when G is CH or N, R8Selected from hydrogen atoms, C1-6Alkyl, halogen, halogeno C1-6Alkyl, hydroxy C1-6Alkyl, -C (O) R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7and-S (O)mNR6R7
s is 1 or 2;
r is 0, 1 or 2;
R2、R5~R7m and n are as defined in claim 1.
4. The compound of formula (I) according to claim 3, which is a compound of formula (III):
Figure FDA0002888084080000022
or a racemate, enantiomers or mixture thereof or pharmaceutically acceptable salts thereof,
wherein:
R9is selected from C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl, -C (O) R5、-C(O)OR5、-S(O)mR5、-C(O)NR6R7and-S (O)mNR6R7
R2、R5~R7N, m, s and r are as defined in claim 3.
5. The compound represented by the general formula (I) according to claim 3, which is a compound represented by the general formula (III-a):
Figure FDA0002888084080000031
or a racemate, enantiomers or mixture thereof or pharmaceutically acceptable salts thereof,
wherein:
R2n, s and r are as defined in claim 3.
6. A compound of formula (I) according to claim 1, wherein R2Are the same or different and are each independently selected from the group consisting of hydrogen, halogen and C1-6An alkyl group.
7. A compound of formula (I) according to claim 1, selected from:
Figure FDA0002888084080000032
Figure FDA0002888084080000041
Figure FDA0002888084080000051
Figure FDA0002888084080000061
Figure FDA0002888084080000071
Figure FDA0002888084080000081
8. a compound represented by the general formula (I-A):
Figure FDA0002888084080000082
or a racemate, enantiomers or mixture thereof or pharmaceutically acceptable salts thereof,
wherein:
x is halogen;
R1、R2and n is as defined in claim 1.
9. The compound of formula (I-a) according to claim 8, selected from:
Figure FDA0002888084080000083
Figure FDA0002888084080000091
10. a process for the preparation of a compound of formula (I) according to claim 1, which process comprises:
Figure FDA0002888084080000092
the compound of the general formula (I-A) and the compound of the general formula (I-B) are subjected to a Suzuki reaction under alkaline conditions in the presence of a catalyst to obtain the compound of the general formula (I),
wherein:
G1is composed of
Figure FDA0002888084080000093
X is halogen;
R1~R3n and p are as defined in claim 1.
11. The method of claim 10, wherein X is bromine.
12. A process for the preparation of a compound of formula (III) according to claim 4, which process comprises:
Figure FDA0002888084080000101
a compound of the formula (III-A) or a hydrochloride thereof and R9-G2To give a compound of the general formula (III),
wherein:
G2selected from halogen, amino, -OC (O) R10And a trimethylsilyl group; wherein R is10Is a hydrogen atom or C1-6An alkyl group;
R2、R9n, s and r are as defined in claim 4.
13. A pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I) according to any one of claims 1 to 7, or a racemate, an enantiomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
14. Use of a compound of general formula (I) according to any one of claims 1 to 7, or a racemate, an enantiomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 13, for the preparation of a medicament for the treatment, prevention or reduction of tumor cell metastasis.
15. Use of a compound of general formula (I) according to any one of claims 1 to 7, or a racemate, an enantiomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 13, for the preparation of a medicament for the treatment, prevention or reduction of cancer mediated by TGF- β overexpression.
16. Use of a compound of general formula (I) according to any one of claims 1 to 7, or a racemate, an enantiomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 13, for the preparation of a medicament for the treatment, prevention or alleviation of a disease selected from the group consisting of: vascular injury, glomerulonephritis, diabetic nephropathy, lupus nephritis, hypertension-induced nephropathy, renal interstitial fibrosis, renal fibrosis caused by drug exposure complications, HIV-related nephropathy, graft nephropathy, liver fibrosis of various etiologies, liver dysfunction attributable to infection, alcohol-induced hepatitis, cystic fibrosis, interstitial lung disease, acute lung injury, adult respiratory distress syndrome, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, lung disease caused by infectious or toxic factors, post-infarction cardiac fibrosis, congestive heart failure, dilated cardiomyopathy, myocarditis, intimal thickening, vascular stenosis, vascular remodeling caused by hypertension, pulmonary hypertension, coronary restenosis, peripheral arterial restenosis, carotid restenosis, stent-induced restenosis, atherosclerosis, coronary heart disease, peripheral arterial restenosis, coronary artery restenosis, stent-induced restenosis, coronary heart disease, peripheral arterial restenosis, hypertension, coronary heart disease, ocular scarring, corneal scarring, proliferative vitreoretinopathy, glaucoma, ocular hypertension, excessive or hypertrophic dermal scarring or keloid formation that occurs during wound healing from trauma or surgical wounds, peritoneal and subcutaneous adhesions, scleroderma, fibrosis sclerosis, progressive systemic sclerosis, dermatomyositis, polymyositis, arthritis, osteoporosis, ulcers, reduced nervous system function, male erectile dysfunction, peyronie's disease, dupuytren's contracture, alzheimer's disease, raynaud's syndrome, radiation-induced fibrosis, thrombosis, tumor metastatic growth, multiple myeloma, melanoma, glioma, glioblastoma, leukemia, sarcoma, leiomyoma, mesothelioma, breast cancer, cervical cancer, lung cancer, gastric cancer, rectal cancer, colon cancer, pancreatic cancer, and the like, Brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, and liver cancer.
CN201711111958.8A 2016-11-14 2017-11-13 3-pyridyl-4-benzothiazolylpyrazole derivatives, preparation method and medical application thereof Active CN108069955B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610998808 2016-11-14
CN2016109988082 2016-11-14

Publications (2)

Publication Number Publication Date
CN108069955A CN108069955A (en) 2018-05-25
CN108069955B true CN108069955B (en) 2021-04-06

Family

ID=62159813

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201711111958.8A Active CN108069955B (en) 2016-11-14 2017-11-13 3-pyridyl-4-benzothiazolylpyrazole derivatives, preparation method and medical application thereof

Country Status (1)

Country Link
CN (1) CN108069955B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108779100A (en) * 2016-11-14 2018-11-09 江苏恒瑞医药股份有限公司 3,4- bipyridyls pyrazole derivatives, preparation method and its application in medicine

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021120890A1 (en) * 2019-12-20 2021-06-24 Novartis Ag Pyrazolyl derivatives useful as anti-cancer agents
US20240116900A1 (en) * 2020-10-30 2024-04-11 Novartis Ag New crystalline forms of a kras g12c inhibitor compound
CN112625030A (en) * 2020-12-25 2021-04-09 杭州澳赛诺生物科技有限公司 Synthetic method for synthesizing N-protected 3-bromopyrazole by one-pot method

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004050659A1 (en) * 2002-11-27 2004-06-17 Eli Lilly And Company Novel compounds as pharmaceutical agents
CN1770980A (en) * 2003-02-12 2006-05-10 比奥根艾迪克Ma公司 Pyrazoles and methods of making and using the same
WO2010046780A2 (en) * 2008-10-22 2010-04-29 Institut Pasteur Korea Anti viral compounds

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004050659A1 (en) * 2002-11-27 2004-06-17 Eli Lilly And Company Novel compounds as pharmaceutical agents
CN1770980A (en) * 2003-02-12 2006-05-10 比奥根艾迪克Ma公司 Pyrazoles and methods of making and using the same
WO2010046780A2 (en) * 2008-10-22 2010-04-29 Institut Pasteur Korea Anti viral compounds

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108779100A (en) * 2016-11-14 2018-11-09 江苏恒瑞医药股份有限公司 3,4- bipyridyls pyrazole derivatives, preparation method and its application in medicine

Also Published As

Publication number Publication date
CN108069955A (en) 2018-05-25

Similar Documents

Publication Publication Date Title
CN113166103B (en) EGFR inhibitor and application thereof
CN108069955B (en) 3-pyridyl-4-benzothiazolylpyrazole derivatives, preparation method and medical application thereof
JP6109969B2 (en) Novel pyrimidine and pyridine compounds and their use
CN101553482B (en) Kinase inhibitor compounds
CN112243439A (en) Pyrrolo [2,3-B ] pyridines or pyrrolo [2,3-B ] pyrazines as HPK1 inhibitors and uses thereof
TW202039498A (en) Pyrimidine five-membered nitrogen heterocyclic derivatives, a preparation method thereof and pharmaceutical use thereof
CN107531679B (en) Aromatic amide derivative, preparation method and medical application thereof
WO2020238791A1 (en) Hydropyridopyrimidine derivative, preparation method therefor and medical use thereof
CN112955432A (en) Fused aromatic ring derivative, preparation method and medical application thereof
WO2021249475A1 (en) Fused quinazoline derivative, preparation method therefor and application thereof in medicine
CN108779115B (en) Five-membered heteroaromatic ring bridged ring derivative, preparation method thereof and application thereof in medicine
WO2023025320A1 (en) Nitrogen-containing heterocyclic derivative inhibitor, and preparation method therefor and use thereof
CN113874354B (en) Pyridone derivative, preparation method thereof and application thereof in medicine
TW202110848A (en) A substituted fused bicyclic derivative, a preparation method thereof, and medical applications thereof
KR20230142504A (en) CDK inhibitors
CN110494431A (en) Nitrogen heterocycles derivative, preparation method and its medical usage
CN113166141B (en) Hexa-membered and hexa-membered heterocyclic compounds and application thereof as protein receptor kinase inhibitors
CN108779100B (en) 3, 4-dipyridyl pyrazole derivatives, preparation method and medical application thereof
CN109983015B (en) 6-pyrazole- [1,2,4] triazolo [4,3-a ] pyridine-3-amide derivatives, preparation method and medical application thereof
WO2021027647A1 (en) Bridged heterocyclyl-substituted pyrimidine compound, preparation method therefor, and pharmaceutical use thereof
WO2022100738A1 (en) Crystal form of free base of inhibitor containing bicyclic ring derivative and preparation method and application of crystal form
WO2023064586A1 (en) Novel modulators of ehmt1 and ehmt2 and therapeutic use thereof
CN112812105A (en) Aminopyridyloxypyrazole derivative and preparation method and application thereof
JP2023536603A (en) FGFR inhibitor compounds and uses thereof
CN113493453A (en) Fused aromatic ring derivative, preparation method and medical application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant