CN113773335A - Compound as protein kinase inhibitor and preparation method and application thereof - Google Patents

Compound as protein kinase inhibitor and preparation method and application thereof Download PDF

Info

Publication number
CN113773335A
CN113773335A CN202111080921.XA CN202111080921A CN113773335A CN 113773335 A CN113773335 A CN 113773335A CN 202111080921 A CN202111080921 A CN 202111080921A CN 113773335 A CN113773335 A CN 113773335A
Authority
CN
China
Prior art keywords
substituted
unsubstituted
compound
heterocycloalkyl
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202111080921.XA
Other languages
Chinese (zh)
Inventor
刘冠锋
杨茂
刘龙飞
李英富
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.)
Chengdu Haibowei Pharmaceutical Co ltd
Original Assignee
Chengdu Haibowei 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 Chengdu Haibowei Pharmaceutical Co ltd filed Critical Chengdu Haibowei Pharmaceutical Co ltd
Publication of CN113773335A publication Critical patent/CN113773335A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/22Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed systems contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention discloses a compound, which has obvious inhibition effect on the activity of a protein kinase inhibitor, has obvious advantages in the aspect of pharmacokinetics compared with clinical second-stage TPX-0005, can be used as the protein kinase inhibitor, and has wide application prospect in resisting malignant tumor diseases or inflammatory diseases.

Description

Compound as protein kinase inhibitor and preparation method and application thereof
The invention is a divisional application of Chinese patent application CN202010525613.2, and the application date of the parent application is 2020.06.10.
Technical Field
The invention relates to the technical field of medicines, in particular to a compound serving as a protein kinase inhibitor, and a preparation method and application thereof.
Background
Lung cancer is one of the most rapidly growing malignancies with the greatest threat to human health and life. Of these, non-small cell lung cancer accounts for approximately 80% of all lung cancers, so research on non-small cell lung cancer has been a focus of both past and present research. The current main method for treating lung cancer is still molecular targeted therapy, and mainly comprises ALK, EGFR, C-Met, ROS1, TRK and other targets. ALK (anaplastic lymphoma kinase) is a receptor tyrosine kinase, involved in the development of many human cancers and humans, is a member of the insulin receptor family, has high homology with leukocyte tyrosine kinase, and is associated with three major types of tumors, including blood, stroma, and solid. The EML4 gene exon and ALK gene exon are fused in the tumor chromosome of about 3-7% of patients with non-small cell lung cancer (NSCLC) to form EML4-ALK fusion tyrosine kinase, the EML4-ALK fusion variant has high carcinogenicity, and ALK is highly expressed in various tumor cells. ROS1 is called c-ROS protooncogene, and is a receptor tyrosine kinase gene for transmembrane. Patients with the ROS1 mutation were more young, non-smoking lung cancer patients, with a majority of lung adenocarcinoma. The mutant accounted for approximately 3% of the total NSCLC population. TRK (Tropomyosin-related kinase) is a tyrosine kinase of neurotrophic receptors present in a variety of tissues and activates a variety of downstream processes during cell proliferation and survival. There are three members of the TRK proto-oncogene family: TRKA, B and C, encoded by NTRK1, NTRK2, NTRK3, respectively. Since 2018, the first worldwide drug targeting species, larotinib, was marketed, yet another "diamond" target gene, NTRK, has quickly gone through the circle of cancer. However, in the common lung cancer, breast cancer and colorectal cancer in China, only 1-5% of patients have the mutation. Although the incidence of NTRK fusion in NSCLC is very low, once the fusion mutation is found, the targeting drug has high effective rate, can actively treat TKI, and has huge potential and broad market prospect.
The breakthrough progress of using ALK as a new target for treating lung cancer has been achieved, and the fourth generation of macrocyclic inhibitors has been developed. The first generation of ALK inhibitor Crizotinib (Crizotinib) developed by pyroxene was generated based on the discovery of the EML4-ALK target. Crizotinib is an ATP competitive multi-target protein kinase inhibitor, can effectively inhibit the cellular biological activity of MET/ALK/ROS, and shows higher clinical efficacy in tumor patients with abnormal ALK, ROS1 or MET kinase activity respectively. Crizotinib is one of the fastest drugs in the development history of tumor drugs, and causes bombing in the united states in 2011. The united states drug administration (FDA) approved Crizotinib (Crizotinib) for the treatment of advanced (metastatic) non-small cell lung cancer (NSCLC) patients carrying mutations in the ROS-1 gene in 2016.
However, with the first ALK drug, crizotinib, on the market, patients develop resistance within two years of continued dosing. The drug resistance mainly comprises three types, namely, the change of an ALK gene such as a 'gatekeeper gene' mutation L1196M and a common mutation C1156Y, an insertion mutation 1151Tins and the like; continuous amplification of ALK fusion gene; third, activation of the ALK signaling pathway bypass signal. To address the resistance issues arising from crizotinib, the FDA announced exenatide (CH5424802) developed by roche in 2014 and norwalk's ceritinib (LDK378) as a second generation ALK inhibitor drug for treating lung cancer. In addition, another second-generation ALK inhibitor, namely Bugatinib (Brigatinib), is also listed on the FDA in 2017 in succession, and is the only EGFR and ALK double-target drug at present. Although acquired drug resistant patients have gained many benefits from these marketed drugs, the efficacy of these therapies is often limited due to side effects or the problem of tumors acquiring drug resistance again, and there is an urgent need to develop new, highly effective, broad-spectrum kinase inhibitors.
On day 2/11 in 2018, pharmaceutical giant-tailed company announces that the third-generation ALK kinase inhibitor, loratidib (loratidib), developed by the company is approved by FDA to be marketed for treating ALK-positive metastatic non-small cell lung cancer (NSCLC) patients. These patients developed resistance to treatment with crizotinib (crizotinib) or at least one other ALK inhibitor, or with treatment with elotinib (aletinib) or ceritinib (ceritinib) as the first ALK inhibitor. Although loratinib had a better blocking effect against most secondary drug-resistant mutations, including the refractory ALK G1202R mutation. But the clinical application of the medicine is severely limited due to the side effects of hypercholesterolemia, hypertriglyceridemia, edema, peripheral neuropathy and the like.
At present, a fourth generation ALK inhibitor TPX-0005 (lopertinib) developed by TP Therapeutics company enters a clinical second-stage research, is a broad-spectrum ALK, ROS1 and TRK inhibitor, and effectively aims at various acquired mutations, including ALK G1202R, ROS 1G 2032R, TRKA G595R and the like. The compound has high inhibitor activity on drug-resistant mutation generated by crizotinib, has good inhibitor effect on drug-resistant mutation generated by using a second-generation ALK inhibitor, and has small side effect. On 28.6.2017, TP Therapeutics announced that FDA has awarded orphan drug qualification to its on-study clinical new drug compound TPX-0005 for the treatment of NSCLC patients carrying ALK, ROS1 or NTRK oncogene rearrangements.
TPX-0005 (lopertinib) has potential to comprehensively solve the problem of drug tolerance caused by drug resistance mutation after ALK, ROS1 and NTRK are targeted, but the pharmacodynamic and pharmacokinetic properties of the TPX-0005 (lopertinib) still have great improvement space.
Disclosure of Invention
The invention mainly solves the technical problem of providing a compound capable of inhibiting protein kinase. In order to solve the technical problems, the invention adopts a technical scheme that:
providing a compound having the structure of formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, pharmaceutically acceptable hydrate, solvate, or salt thereof:
Figure BDA0003263976850000031
wherein:
M1selected from the group consisting of CR15Or N, M2Selected from the group consisting of CR16Or N;
ring E is selected from substituted or unsubstituted C3~C15Wherein the heterocycloalkyl group contains one or more O, NR24S, S (O), S (O)2, the substituents each being independently selected from the group consisting of halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxy, cyano, amino, ester, amide, aryl, heteroaryl, sulfonyl;
A1、A2、A3、A4、A5、A6each independently selected from C, CR17NH, N, and A1、A2、A3、A4、A5、A6At least one of which is N or NH;
y is selected from CR26 R27、O、NR18、S、S(O)、S(O)2、C=O、C=S;
n is selected from 0-5;
R1、R2、R3、R9、R15、R16、R17、R18、R26、R27each independently selected from hydrogen, halogen, substituted or non-substitutedSubstituted alkyl or cycloalkyl, substituted or unsubstituted heteroalkyl or heterocycloalkyl, hydroxy, cyano, amino, ester, amido, aryl, heteroaryl, sulfonyl, wherein the substituents are each independently selected from the group consisting of halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxy, cyano, amino, ester, amido, aryl, heteroaryl, sulfonyl;
R4selected from the group consisting of hydrogen, halogen, substituted or unsubstituted alkyl or cycloalkyl, substituted or unsubstituted heteroalkyl or heterocycloalkyl, hydroxyl, cyano, amino, ester, amide, aryl, heteroaryl, sulfonyl, boronic acid, phosphoryl, wherein the substituents are each independently selected from the group consisting of halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, amino, ester, amide, aryl, heteroaryl, sulfonyl;
R5、R6、R7、R8、R22、R23are respectively and independently selected from hydrogen, halogen, substituted or unsubstituted alkyl or heteroalkyl of C1-C6, substituted or unsubstituted cycloalkyl or heterocycloalkyl of C3-C6, or R5、R6The carbon atoms connected with the substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl, or R7、R8The carbon atoms connected with the substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl, or R22、R23The carbon atoms connected with the substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl, or R5、R8The carbon atoms connected with the carbon atoms respectively and the saturated chain segment of the macrocyclic molecule between the two carbon atoms jointly form substituted or non-substituted monocyclic, bicyclic or tricyclic C3-10 cycloalkyl or heterocycloalkyl, or R5、R9The carbon atom and the nitrogen atom which are respectively connected with the above-mentioned two groups form a substituted or unsubstituted C3-10 heterocycloalkyl group, or R23、R9The carbon atoms and the nitrogen atoms which are respectively connected with the substituted or unsubstituted C3-10 heterocycloalkyl group are formed by the saturated chain segments of the macrocyclic molecules between the carbon atoms and the nitrogen atoms;
wherein said heterocycloalkyl group contains one or more O, NR25、S、S (O), S (O)2, wherein the substituents are respectively and independently selected from halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, amino, ester group, amido, aryl, heteroaryl and sulfonyl;
R24、R25selected from hydrogen, substituted or unsubstituted alkyl or cycloalkyl, substituted or unsubstituted heteroalkyl or heterocycloalkyl, hydroxyl, amido, aryl, heteroaryl, sulfonyl, wherein the substituents are each independently selected from halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, amino, ester, amido, aryl, heteroaryl, sulfonyl.
Further, ring E is selected from substituted or unsubstituted C3~C15Monocyclic cycloalkyl or monocyclic heterocycloalkyl of (1).
Further alternatively, ring E is selected from substituted or unsubstituted C3~C6Monocyclic cycloalkyl or monocyclic heterocycloalkyl of (1).
Further, the heterocycloalkyl group is an N-heterocycloalkyl group.
Further, the N-heterocycloalkyl is
Figure BDA0003263976850000041
Wherein R is24Selected from hydrogen, substituted or unsubstituted alkyl.
Further, said R24Selected from hydrogen, unsubstituted alkyl; further, said R24Selected from hydrogen and methyl.
Further, it has the structure shown in formula (II) or its isomer, tautomer, meso form, racemate, enantiomer, diastereoisomer or mixture form, pharmaceutically acceptable hydrate, solvate or salt thereof:
Figure BDA0003263976850000042
wherein R is10、R11、R12、R13Each independently selected from hydrogen, halogen, hydroxyl, cyano, amino, substituted or non-substitutedThe aryl-substituted alkyl-substituted cycloalkyl-substituted heterocycloalkyl group comprises C1-6 substituted alkyl or heteroalkyl, and substituted or unsubstituted C3-6 substituted cycloalkyl or heterocycloalkyl, wherein the substituents are respectively and independently selected from halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, amino, ester, amido, aryl, heteroaryl and sulfonyl.
Further, A4 is selected from N, A2 and A5 are independently selected from CR17,A1、A3、A6Each independently selected from C, N, and A3、A6Are not identical.
Further, M1Selected from the group consisting of CR15,M2Selected from the group consisting of CR16Or N.
Further, R16Selected from H, halogen, C1-3 alkyl or heteroalkyl.
Further, R16Selected from H, F, chloro, trifluoromethyl, difluoromethyl, methyl, methoxy.
Further, it has the structure shown in formula (III) or a tautomer, mesomer, racemate, enantiomer, diastereomer or mixture form, pharmaceutically acceptable hydrate, solvate or salt thereof:
Figure BDA0003263976850000051
wherein Y is selected from C R26R27、O、NR18、S、S(O)、S(O)2、C=O;
n is selected from 0, 1 or 2;
R2、R3、R4、R9、R10、R11、R12、R13、R14、R18、R26、R27independently selected from hydrogen, halogen, hydroxyl, cyano, amino, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the substituents are independently selected from halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, amino, ester, amide, aryl, carboxyl, amino, or amino,Heteroaryl, sulfonyl;
R5、R6、R7、R8、R22、R23independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, or R5、R6The carbon atoms connected with the substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, or R7、R8The carbon atoms connected with the substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, or R22、R23The carbon atoms connected with the substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, or R5、R8The carbon atoms connected with the carbon atoms respectively and the saturated chain segment of the macrocyclic molecule between the two carbon atoms jointly form substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, or R5、R9The carbon atom and the nitrogen atom which are respectively connected with the N-substituted heterocyclic ring form substituted or unsubstituted C3-6 heterocycloalkyl, or R23、R9The carbon atoms and the nitrogen atoms which are respectively connected with the saturated chain segments of the macrocyclic molecules form a substituted or unsubstituted C3-6 heterocycloalkyl together, wherein the heterocycloalkyl contains one or more O or NR25The substituents are each independently selected from the group consisting of halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxy, cyano, amino, ester, amide, aryl, heteroaryl, sulfonyl;
R25selected from hydrogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl; the substituents are each independently selected from halogen, hydroxyl, cyano, amino, ester group, amide group.
10. A compound according to any one of claims 1 to 9, characterized in that: y is selected from CR26R27、O、NR18(ii) a Further, Y is selected from CH2、O、NH。
Further, n is selected from 0 and 1.
Further, R5、R6、R7、R8、R22、R23Are respectively and independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl, or R5、R6The carbon atoms connected with the substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, or R7、R8The carbon atoms connected with the substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, or R22、R23The carbon atoms connected with the aryl form substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl.
Further, R5、R6、R7、R8、R22、R23Are respectively and independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl, or R5、R6The carbon atoms connected with the aryl form substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl.
In a particular embodiment of the invention, R of said compound5、R6、R7、R8、R9、R22、R23In the middle, at least one ring is formed between two rings.
Here, at least one ring formation between two rings means that at R5、R6、R7、R8、R9、R22、R23In the presence of at least one looping event according to claim 1, namely: r5、R6The carbon atoms connected with the substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl; ② R7、R8The carbon atoms connected with the substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl; (iii) R22、R23The carbon atoms connected with the substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl; r5、R8The carbon atoms connected with the carbon atoms respectively and the saturated chain segment of the macrocyclic molecule between the two carbon atoms jointly form a substituted or non-substituted monocyclic, bicyclic or tricyclic cycloalkyl or heterocycloalkyl of C3-10; fifthly, R5、R9The carbon atoms and the nitrogen atoms which are respectively connected with the substituted or unsubstituted C3-10 heterocycloalkyl group are formed together; sixthly, theR23、R9The carbon atoms and the nitrogen atoms which are respectively connected with the substituted or unsubstituted C3-10 heterocycloalkyl group are formed by the saturated chain segments of the macrocyclic molecules between the carbon atoms and the nitrogen atoms; in six ring formation cases, at least one is present.
Further, R5、R6、R7、R8、R9、R22、R23In the middle, there is one and only one looping condition between two.
Further, R5、R6、R7、R9、R8、R22、R23Wherein the acyclic is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl.
Further, R5、R6、R7、R9、R8、R22、R23Wherein the acyclic alkyl group is selected from hydrogen, halogen, substituted or unsubstituted C1-3 alkyl.
Further, R5、R6、R7、R9、R8、R22、R23Wherein, the acyclic ones are all selected from H.
Further, it has the structure shown in formula (IV) or a tautomer, a meso form, a racemate, an enantiomer, a diastereomer or a mixture form, a pharmaceutically acceptable hydrate, a solvate or a salt thereof:
Figure BDA0003263976850000071
wherein:
n is selected from 0, 1 or 2;
R2selected from halogens;
R4、R9、R10、R11、R14are respectively and independently selected from hydrogen, halogen, hydroxyl, cyano, amino, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the substituents are respectively and independentlySelected from halogen, hydroxyl, cyano, amino, ester, amido and sulfonyl;
R5、R6、R22、R23are respectively and independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl;
or R5、R6The carbon atoms connected with the substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl;
the heterocycloalkyl group containing one or more O or NR25The substituents are respectively and independently selected from halogen, hydroxyl, cyano, amino, ester group, amido and sulfonyl;
or R5、R9The carbon atom and the nitrogen atom which are respectively connected with the N-substituted heterocyclic alkyl form a substituted or unsubstituted C3-6N heterocyclic alkyl group, or R23、R9The N-heterocyclic alkyl is substituted or unsubstituted C3-6 and is formed by carbon atoms, nitrogen atoms and saturated chain segments of macrocyclic molecules between the carbon atoms and the nitrogen atoms, wherein the substituents are respectively and independently selected from halogen, C1-6 alkyl or heteroalkyl, hydroxyl, cyano, amino and ester groups;
R25is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C3-6 cycloalkyl, wherein the substituent is selected from halogen, hydroxyl, cyano and amino.
Further, R4The compound is selected from H, halogen, hydroxyl, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the substituents are independently selected from halogen, hydroxyl, cyano, amino, ester, amido and sulfonyl;
R5、R6、R22、R23、R9are respectively and independently selected from H, halogen and C1-3 alkyl;
or R5、R6The carbon atoms connected with the heterocyclic ring form a substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the heterocycloalkyl contains one or more O or NR25The substituents are respectively and independently selected fromF. Cl, Br, hydroxyl, cyano, amino;
or R5、R9The carbon atom and the nitrogen atom which are respectively connected with the N-substituted heterocyclic ring form the N-heterocyclic alkyl of C3-6, or R23、R9The C3-6N heterocycloalkyl is formed by carbon atoms, nitrogen atoms and saturated chain segments of macrocyclic molecules between the carbon atoms and the nitrogen atoms which are respectively connected with the N and the nitrogen atoms;
R25selected from hydrogen, substituted or unsubstituted C1-3 alkyl, wherein the substituents are independently selected from hydroxyl or amino.
Further: r10、R11、R14Selected from H, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl; further, R10、R11、R14Is selected from H.
In a particular embodiment of the invention, R4Selected from H, F, Cl, Br, methyl, trifluoromethyl, difluoromethyl, methoxy, ethyl, propyl, isopropyl, cyclopropyl, hydroxy,
Figure BDA0003263976850000081
In a particular embodiment of the invention, n is selected from 0 or 1.
Further, R5、R6、R9、R22、R23In the middle, at least one ring is formed between two rings.
Further, R5、R6、R9、R22、R23In the middle, there is one and only one looping condition between two.
Further, R5、R6、R9、R22、R23Wherein, the acyclic ones are all selected from H.
Further, R5、R6The carbon atoms connected with the aryl form a substituted or unsubstituted C3-6 cycloalkyl, or R5、R9The substituted or unsubstituted 4-6-membered N-containing heterocycloalkyl group is formed by the carbon atom and the nitrogen atom which are respectively connected with the substituted or unsubstituted N-containing heterocycloalkyl group, and N is selected from 0; further, R5、R6To which the carbon atom is attachedSubstituted or unsubstituted cyclopropyl, or R5、R9Together with the carbon atom and the nitrogen atom to which they are respectively attached, form a substituted or unsubstituted 5-membered N-containing heterocycloalkyl group.
Further, the air conditioner is provided with a fan,
Figure BDA0003263976850000082
is composed of
Figure BDA0003263976850000083
Further, the
Figure BDA0003263976850000084
Is composed of
Figure BDA0003263976850000085
Further, it has a structure shown in formula (V) or a tautomer, mesomer, racemate, enantiomer, diastereomer or mixture form, pharmaceutically acceptable hydrate, solvate or salt thereof:
Figure BDA0003263976850000086
wherein:
n is selected from 0, 1 or 2;
R2selected from halogens;
R4、R9、R10、R11、R14the substituent groups are respectively and independently selected from hydrogen, halogen, hydroxyl, cyano, amino, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the substituent groups are respectively and independently selected from halogen, hydroxyl, cyano, amino, ester group, amido and sulfonyl;
R23selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl;
R5、R8to which are respectively attached carbon atoms and between twoSaturated segments of macrocyclic molecules between carbon atoms together form a substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl group, wherein the heterocycloalkyl group contains one or more O or NR25The substituents are respectively and independently selected from halogen, hydroxyl, cyano, amino, ester group, amido and sulfonyl;
R25is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C3-6 cycloalkyl, wherein the substituent is selected from halogen, hydroxyl, cyano and amino.
Further, R4The compound is selected from H, halogen, hydroxyl, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the substituents are independently selected from halogen, hydroxyl, cyano, amino, ester, amido and sulfonyl;
R9an alkyl or heteroalkyl group selected from H, C1-3;
R23selected from H, halogen and C1-3 alkyl;
R5、R8the carbon atoms connected with the heterocyclic compound and the saturated chain segment of the macrocyclic molecule between two carbon atoms jointly form a substituted or non-substituted C3-6 cycloalkyl or heterocycloalkyl, wherein the heterocycloalkyl contains one or more O or NR25The substituents are respectively and independently selected from F, Cl, Br, hydroxyl, cyano and amino;
R25selected from hydrogen, substituted or unsubstituted C1-3 alkyl, wherein the substituents are independently selected from hydroxyl or amino.
Further, R10、R11、R14Selected from H, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl.
Further, R10、R11、R14Is selected from H.
In a particular embodiment of the invention, R4Selected from H, F, Cl, Br, methyl, trifluoromethyl, difluoromethyl, methoxy, ethyl, propyl, isopropyl, cyclopropyl, hydroxy,
Figure BDA0003263976850000091
Further, R5、R8Carbon atoms connected with the substituted or unsubstituted C3-6 cycloalkyl group are respectively and independently selected from F, Cl, Br, hydroxyl, cyano and amino; y is O.
Further, R5、R8The carbon atoms to which they are respectively attached and the saturated segment of the macrocyclic molecule between the two carbon atoms together constitute a substituted or unsubstituted cyclobutyl, cyclopentyl, cyclohexyl, preferably cyclopentyl, cyclohexyl.
Further, the air conditioner is provided with a fan,
Figure BDA0003263976850000092
is composed of
Figure BDA0003263976850000093
Further comprises
Figure BDA0003263976850000094
Further, n is selected from 0 or 1.
Further, R4Selected from H and halogen.
In a particular embodiment of the invention, the compound structure is selected from one of the following:
Figure BDA0003263976850000095
Figure BDA0003263976850000101
Figure BDA0003263976850000111
Figure BDA0003263976850000121
the invention also provides a medicinal composition, wherein the active ingredient of the medicinal composition is selected from one or the combination of more than two of the compounds or the stereoisomer, the solvate, the hydrate, the pharmaceutically acceptable salt or the eutectic crystal thereof.
The pharmaceutical composition may include, in addition to the aforementioned compounds, at least one additional therapeutic agent. The additional therapeutic agent may be selected from immunomodulators, cancer chemotherapeutics, anti-infectives, and the like. Such cancer chemotherapeutic agents include, but are not limited to, alkylating agents (e.g., nitrogen mustards), nucleoside analogs (e.g., gemcitabine), nitrosoureas (e.g., carmustine), platinum-based drugs (e.g., cisplatin), DNA-cleaving agents (e.g., bleomycin), antimetabolites (folic acid antagonists such as methotrexate, pyrimidine antagonists such as fluorouracil, purine antagonists such as pentostatin, etc.), tubulin-interacting agents (e.g., vincristine, docetaxel, etc.), hormones, and the like.
The invention also provides the application of the compound or the stereoisomer, the solvate, the hydrate, the pharmaceutically acceptable salt or the eutectic crystal thereof in preparing a protein kinase inhibitor; further, the kinase inhibitor is selected from one or more of an ALK inhibitor, a ROS1 inhibitor, a TRK inhibitor, a MET inhibitor, a JAK inhibitor and a CSF-1R inhibitor.
The invention also provides the use of the above compound or a stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or co-crystal thereof in the preparation of a medicament for the treatment of pain, cancer, inflammation, autoimmune disease, neurodegenerative disorder or trypanosoma cruzi infection in a mammal.
Further, the cancer is selected from: non-small cell lung cancer, gastric cancer, neuroblastoma, lung cancer, ovarian cancer, appendiceal cancer, colorectal cancer, melanoma, head and neck cancer, breast cancer, glioblastoma, medulloblastoma, secretory breast cancer, papillary thyroid cancer, adult myeloid leukemia, pancreatic cancer, prostate cancer, cholangiocarcinoma, salivary gland carcinoma, gastrointestinal stromal tumor, infantile fibrosarcoma.
The invention also provides application of the compound or a stereoisomer, a solvate, a hydrate, a pharmaceutically acceptable salt or a co-crystal thereof in preparing a medicament for inhibiting cell proliferation.
The invention also provides application of the compound or a stereoisomer, a solvate, a hydrate, a pharmaceutically acceptable salt or a eutectic crystal thereof in preparing a medicament for treating diseases causing ALK, ROS1, TRK, MET, JAK and CSF-1R kinase overexpression.
The invention also provides application of the compound or stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or eutectic thereof in preparing a medicament for treating diseases caused by over-expression of ALK, ROS1, TRK, MET, JAK and CSF-1R kinase.
The invention also provides a preparation method of the compound, which comprises the following steps:
Figure BDA0003263976850000131
(1) reacting the compound a with the compound b under the action of alkali to generate a compound c, and then reacting a substituent R of the compound c28Removing to obtain a compound d;
wherein R is28Selected from benzyl, silyl, p-methoxybenzyl, 2, 4-dimethoxybenzyl, allyl, acyl, C1-3 alkyl;
(2) dehydrating two molecules of hydroxyl of the compound d and the compound e into ether to generate a compound f;
(3) removing the Boc protecting group from the compound f under acidic conditions to generate a compound g;
(4) carrying out hydrolysis reaction on the compound g under the action of alkali to generate a compound h;
(5) and (3) generating the compound i by the compound h under the action of alkali and a condensation reagent.
In a specific embodiment of the present invention, compound f is produced by a Mitsunobu reaction in step (2). The invention also provides another preparation method of the compound, which comprises the following steps:
Figure BDA0003263976850000141
(1) the substituent R of the compound j28Removing to obtain a compound k, and reacting the compound k with a compound b under the action of alkali to generate a compound m;
wherein R is29Selected from benzyl, silyl, p-methoxybenzyl, 2, 4-dimethoxybenzyl, allyl, acyl, C1-3 alkyl;
(2) the compound m and the compound n are condensed under the action of alkali to generate a compound o,
(3) removing the Boc protecting group from the compound o under acidic conditions to generate a compound p;
(4) carrying out hydrolysis reaction on the compound p under the action of alkali to generate a compound h;
(5) and (3) generating a compound r by the compound h under the action of alkali and a condensation reagent.
The invention also provides another preparation method of the compound, which comprises the following steps:
Figure BDA0003263976850000142
(1) reacting the compound s with the compound t to generate a compound u;
(2) compound u is hydrogenated to generate compound v;
(3) removing the Boc protective group from the compound v under acidic conditions to generate a compound w;
(4) reacting the compound w with the compound b under the action of alkali to generate a compound x;
(5) carrying out hydrolysis reaction on the compound x under the action of alkali to generate a compound y;
(5) generating a compound z by the compound y under the action of alkali and a condensation reagent;
wherein R is30Selected from Cl, Br, I, OTf.
The invention also provides another preparation method of the compound, which comprises the following steps:
Figure BDA0003263976850000151
(1) reacting the compound aa with the compound t to generate a compound ab;
(2) compound ab is subjected to hydrogenation reaction to generate compound ac;
(3) removing the Boc protecting group from the compound ac under an acidic condition to generate a compound ad;
(4) reacting the compound ad with the compound b under the action of alkali to generate a compound ae;
(5) compound ae is hydrolyzed under the action of alkali to generate compound af;
(5) generating a compound ag by the compound af under the action of alkali and a condensation reagent;
wherein R is31Selected from Br, I, OTf.
The invention also provides a compound VI, the structural formula of which is as follows:
Figure BDA0003263976850000161
wherein R is19Selected from halogen, NR20、OR20,R20Selected from Boc, benzyl, p-methoxybenzyl, 2, 4-dimethoxybenzyl, methyl, allyl, trimethylsilyl.
The invention also provides a preparation method of the compound VI, which comprises the following steps:
mixing 5-fluoro-2-methoxy nicotinonitrile, tetraisopropyl titanate and a solvent, and adding ethyl magnesium bromide into the mixed system at the temperature of-80 to-50 ℃ in the atmosphere of nitrogen or inert gas for reaction, wherein the preferable temperature is-70 ℃;
based on the molar amount of the 5-fluoro-2-methoxy nicotinonitrile, the amount of the tetraisopropyl titanate is 1-2 equivalents, preferably 1-1.5 equivalents; the amount of Grignard reagent is 1.5 to 3 equivalents, preferably 2 to 2.5 equivalents.
Further, the solvent is one or more selected from tetrahydrofuran, diethyl ether and toluene, and tetrahydrofuran is preferred.
Further, the ethyl magnesium bromide is added in portions, preferably dropwise.
Further, after the Grignard reagent is added, reacting for 5-30 min, preferably 10-15 min, at the original low temperature; then the reaction system is warmed up to room temperature for reaction.
Further, the post-processing method comprises the following steps: mixing the reaction system with 1-4 equivalents of boron trifluoride ethyl ether, and adding dilute hydrochloric acid to quench the reaction; furthermore, the amount of boron trifluoride diethyl etherate is 2-3 equivalents.
The present invention also provides compound VII, having the structural formula:
Figure BDA0003263976850000162
wherein R is21Selected from H, fluorine, chlorine, bromine, methyl, trifluoromethyl, difluoromethyl, methoxy, ethyl, isopropyl, cyclopropyl.
The invention also provides a preparation method of the compound VII, which comprises the following steps:
(1) 2-fluoro malonic acid, 5-amino-1H-pyrazole-4-carboxylic acid ethyl ester and POCl3Mixing DMF and N, N-diethylaniline, and reacting at 80-100 ℃ to obtain 5, 7-dichloro-6-fluoropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester; the using amount of the 5-amino-1H-pyrazole-4-ethyl formate is 0.8-1.5 equivalents, preferably 1 equivalent, based on the molar using amount of the 2-fluoro malonic acid; the dosage of the N, N-diethylaniline is 0.8 to 1.5 equivalents, preferably 1 equivalent;
(2) reacting 5, 7-dichloro-6-fluoropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester, zinc powder and NH4Mixing Cl and a solvent; the solvent is a mixed solvent of EtOH, THF and water which are 2-4: 1: 1.5-2.5; the solvent is a mixed solvent of EtOH, THF, water and 3:1: 2; with 5, 7-dichloro-6-fluoropyrazolo [1,5-a ]]The molar amount of the pyrimidine-3-ethyl formate is used as a reference, and the amount of the zinc powder is 1-2 equivalents, preferably 1-1.5 equivalents; NH (NH)4The amount of Cl is 2 to 3 equivalents, preferably 2.5 to 3 equivalents.
Further, POCl3The dosage of the composition is as follows: 0.5 to 1.5ml of OCCl is used per millimole of 2-fluoro-malonic acid3Preferably 1 mL; the dosage of the N, N-diethylaniline is as follows: 0.1 to 0.5mL of N-diethylaniline, preferably 0.2mL, is used per mmol of 2-fluoropropanedioic acid.
Further, in the step (2), the amount of the solvent is as follows: 10-20 mL of solvent, preferably 15-18 mL, is used per millimole of ethyl 5, 7-dichloro-6-fluoropyrazolo [1,5-a ] pyrimidine-3-carboxylate.
The invention also provides application of the compound VI and the compound VII in preparing the compound.
The "equivalent" in the present invention refers to the molar weight ratio between the compounds, for example: the phrase "the amount of tetraisopropyl titanate is 1 to 2 equivalents based on the molar amount of 5-fluoro-2-methoxynicotinonitrile" means that the amount of tetraisopropyl titanate (molar amount) is 1 to 2 times the amount of 5-fluoro-2-methoxynicotinonitrile (molar amount), and so on.
The pharmaceutical composition containing the compound of the invention or the stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or cocrystal thereof can contain pharmaceutically acceptable auxiliary materials.
As used herein, "pharmaceutically acceptable" is meant to include any material that does not interfere with the effectiveness of the biological activity of the active ingredient and is not toxic to the host to which it is administered.
The pharmaceutically acceptable auxiliary materials are general names of all the additional materials except the main medicine in the medicine, and the auxiliary materials have the following properties: (1) no toxic effect on human body and few side effects; (2) the chemical property is stable and is not easily influenced by temperature, pH, storage time and the like; (3) has no incompatibility with the main drug, and does not influence the curative effect and quality inspection of the main drug; (4) does not interact with the packaging material. The auxiliary materials in the invention include, but are not limited to, a filler (diluent), a lubricant (glidant or anti-adhesion agent), a dispersing agent, a wetting agent, an adhesive, a regulator, a solubilizer, an antioxidant, a bacteriostatic agent, an emulsifier, a disintegrating agent and the like. The binder comprises syrup, acacia, gelatin, sorbitol, tragacanth, cellulose and its derivatives (such as microcrystalline cellulose, sodium carboxymethylcellulose, ethyl cellulose or hydroxypropyl methylcellulose), gelatin slurry, syrup, starch slurry or polyvinylpyrrolidone; the filler comprises lactose, sugar powder, dextrin, starch and its derivatives, cellulose and its derivatives, inorganic calcium salt (such as calcium sulfate, calcium phosphate, calcium hydrogen phosphate, precipitated calcium carbonate, etc.), sorbitol or glycine, etc.; the lubricant comprises superfine silica gel powder, magnesium stearate, talcum powder, aluminum hydroxide, boric acid, hydrogenated vegetable oil, polyethylene glycol and the like; the disintegrating agent comprises starch and its derivatives (such as sodium carboxymethyl starch, sodium starch glycolate, pregelatinized starch, modified starch, hydroxypropyl starch, corn starch, etc.), polyvinylpyrrolidone or microcrystalline cellulose, etc.; the wetting agent comprises sodium lauryl sulfate, water or alcohol, etc.; the antioxidant comprises sodium sulfite, sodium bisulfite, sodium pyrosulfite, dibutylbenzoic acid, etc.; the bacteriostatic agent comprises 0.5% of phenol, 0.3% of cresol, 0.5% of chlorobutanol and the like; the regulator comprises hydrochloric acid, citric acid, potassium (sodium) hydroxide, sodium citrate, and buffer (including sodium dihydrogen phosphate and disodium hydrogen phosphate); the emulsifier comprises polysorbate-80, sorbitan fatty acid, pluronic F-68, lecithin, soybean lecithin, etc.; the solubilizer comprises Tween-80, bile, glycerol, etc. The term "pharmaceutically acceptable salt" refers to a salt of a compound of the present invention with an acid or base that is suitable for use as a pharmaceutical. The acid base is a generalized Lewis acid base. Suitable acids for forming the salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, etc., organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, phenylmethanesulfonic acid, benzenesulfonic acid, etc.; and acidic amino acids such as aspartic acid and glutamic acid.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, parenteral (intravenous, intramuscular, or subcutaneous), and topical administration.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers, e.g., ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, and oils, especially cottonseed, groundnut, corn germ, olive, castor and sesame oils, or mixtures of such materials, and the like.
In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms for topical administration of the compounds of the present invention include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
The compounds of the invention can likewise be used in injectable preparations. Wherein the injection is selected from liquid injection (water injection), sterile powder for injection (powder injection) or tablet for injection (refers to impression tablet or machine pressing tablet prepared by aseptic operation method of medicine, and is dissolved with water for injection for subcutaneous or intramuscular injection when in use).
Wherein the powder for injection contains at least an excipient in addition to the above compound. The excipients, which are components intentionally added to a drug in the present invention, should not have pharmacological properties in the amounts used, however, the excipients may aid in the processing, dissolution or dissolution of the drug, delivery by a targeted route of administration, or stability.
"alkyl" refers to an aliphatic hydrocarbon group and to a saturated hydrocarbon group. The alkyl moiety may be a straight chain or branched chain alkyl.
The C1-n used in the invention comprises C1-2 and C1-3 … … C1-n. n is an integer greater than one. Typical alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, hexyl, and the like.
"amide" is a chemical structure having the formula-C (O) NHR or-NHC (O) R, wherein R is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl.
"ester" means having the formula-COOR, wherein R is selected from the group consisting of alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl.
"Ring" refers to any covalently closed structure, including, for example, carbocycles (e.g., aryl or cycloalkyl), heterocycles (e.g., heteroaryl or heterocycloalkyl), aryls (e.g., aryl or heteroaryl), nonaromatic (e.g., cycloalkyl or heterocycloalkyl). The rings may be optionally substituted, and may be monocyclic or polycyclic. Typical polycyclic rings generally include bicyclic and tricyclic rings.
"element" means the number of skeleton atoms constituting a ring. Typical 5-membered rings may include cyclopentyl, pyrrole, imidazole, thiazole, furan, thiophene, and the like; typical 6-membered rings include cyclohexyl, pyridine, pyran, pyrazine, thiopyran, pyridazine, pyrimidine, benzene, and the like. Wherein, the skeleton atom contains the ring of heteroatom, namely is the heterocycle; aryl consisting of a heterocycle is heteroaryl; the nonaromatic group consisting of a heterocycle is a heterocycloalkyl group.
"heteroalkyl" refers to an alkyl group containing a heteroatom, including, but not limited to O, S, N, P, and the like; alkoxy, thioalkyl, aminoalkyl and the like are all intended to be heteroalkyl.
"C1-n" heteroalkyl or heterocycloalkyl group, wherein n is an integer greater than one and is the number of carbon atoms in the heteroalkyl group or the number of carbon atoms in the backbone of the ring formed by the heterocycloalkyl group is n.
Typical heterocycloalkyl groups include, but are not limited to:
Figure BDA0003263976850000201
typical heteroaryl or heteroaryl groups include, but are not limited to:
Figure BDA0003263976850000202
"aryl" means a planar ring having a delocalized pi-electron system and containing 4n +2 pi electrons, where n is an integer. An aryl ring may be composed of five, six, seven, eight, nine or more than nine atoms. Aromatic groups include, but are not limited to, phenyl, naphthyl, phenanthryl, anthracyl, fluorenyl, indenyl, and the like.
"cycloalkyl" refers to a monocyclic or polycyclic group containing approximately carbon and hydrogen, which may be saturated or unsaturated. Typical cycloalkyl structures include, but are not limited to:
Figure BDA0003263976850000203
"halogen" "or" halo "refers to fluorine, chlorine, bromine or iodine. "haloalkyl" means an alkyl group wherein at least one hydrogen is replaced by a halogen atom.
The present invention also provides the following expression: r5、R8The carbon atoms connected with the carbon atoms respectively and the saturated chain segment of the macrocyclic molecule between the two carbon atoms jointly form a substituted or non-substituted monocyclic, bicyclic or tricyclic cycloalkyl or heterocycloalkyl of C3-10; or R23、R9And carbon atoms and nitrogen atoms which are respectively connected with the substituted or unsubstituted C3-10 heterocycloalkyl group are formed by the saturated chain segments of the macrocyclic molecules between the carbon atoms and the nitrogen atoms. Wherein the "saturated segment of the macrocyclic molecule" is explained as follows: since the compounds of the present invention are macrocyclic compounds, the macrocyclic chain as previously described as "between two carbon atoms" or "between carbon and nitrogen atoms" has two, one long, and the other relatively short, and as can be seen from the structural formula, the relatively short macrocyclic chain is defined herein as the saturated segment of the macrocyclic molecule.
For example:
in the compound of formula (I), R5、R8The carbon atoms connected with the carbon atoms and the saturated chain segment of the macrocyclic molecule between the two carbon atoms jointly form a substituted or non-substituted monocyclic, bicyclic or tricyclic cycloalkyl or heterocycloalkyl of C3-10,
namely, it is
Figure BDA0003263976850000211
In this paragraph, R5、R8To the carbon atom to which it is attached, with- (CR)22R23)-nWherein the carbon chain on the macrocyclic molecule is combined to form a substituted or unsubstituted monocyclic, bicyclic or tricyclic C3-10 cycloalkyl or heterocycloalkyl group, such as
Figure BDA0003263976850000212
Etc.; and so on.
The amino group, ester group, carbonyl group, amide group and the like described herein may be an unsubstituted amino group, ester group, carbonyl group, amide group, or a substituted amino group, ester group, carbonyl group, amide group.
Hereinbefore, except where already indicated, "substituted" in said "substituted or unsubstituted" means that the mentioned groups may be substituted by one or more additional groups each and independently selected from alkyl, cycloalkyl, aryl, carboxy, heteroaryl, heterocycloalkyl, hydroxy, alkoxy, alkylthio, aryloxy, nitro, acyl, halogen, haloalkyl, amino and the like.
"inhibitor" refers to a substance that decreases the activity of an enzyme.
Obviously, many modifications, substitutions, and variations are possible in light of the above teachings of the invention, without departing from the basic technical spirit of the invention, as defined by the following claims.
The foregoing aspects of the present invention are explained in further detail below with reference to specific embodiments. It should not be understood that the scope of the above-described subject matter of the present invention is limited to the following examples. All the technologies realized based on the above contents of the present invention belong to the scope of the present invention.
The invention has the beneficial effects that:
(1) the invention provides a series of compounds capable of inhibiting ALK, ROS1 and TRK kinase activity, tests show that the compounds have obvious inhibition effect on protein kinase inhibitor activity, and a new scheme is provided for treating diseases taking protein kinase as a treatment target, such as mammal pain, cancer, inflammation, autoimmune diseases, neurodegenerative diseases, trypanosoma cruzi infection and other diseases.
(2) Compared with clinical second-stage TPX-0005, the compound has obvious advantages in the aspect of pharmacokinetics, can be used as a protein kinase inhibitor, and has wide application prospect in resisting malignant tumor diseases or inflammatory diseases.
Detailed Description
The technical solutions of the present invention are described clearly and completely below, and it is obvious that the described embodiments are some, not all embodiments of the present invention. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
In the examples, the following abbreviations have the meanings indicated below:
NMP: n-methyl pyrrolidone;
EA: ethyl acetate;
m means molar concentration units mol/L, e.g., 1M means 1 mol/L;
THF: tetrahydrofuran;
n means equivalent concentration, e.g., 1N HCl means hydrochloric acid at a concentration of 1 mol/L;
MTBE: methyl tert-butyl ether;
POCl3: phosphorus oxychloride;
DMF: n, N-dimethylformamide;
TLC: thin layer chromatography;
PE refers to petroleum ether (with a boiling point of 60-90 ℃);
DCM: dichloromethane;
H2o: distilled water;
DIEA: n, N-diisopropylethylamine;
TMSCl: trimethylchlorosilane;
the DIAD: diisopropyl azodicarboxylate;
a Dioxane: 1, 4-dioxane;
HATU: o- (7-azabenzotriazol-1-yl) -N, N' -tetramethyluronium hexafluorophosphate;
TsCl: p-toluenesulfonyl chloride;
1, 4-dioxane: 1, 4-dioxane;
Pd2(dba)3: 3,3,6, 6-tetramethyl-9- (1,2,3, 4-tetrahydroxybutyl) -4,5,7, 9-tetrahydro-2H-heteroanthracene-1, 8-dione);
x-phos: 2-dicyclohexylphosphonium-2, 4, 6-triisopropylbiphenyl;
xant-phos 4, 5-bis diphenylphosphine-9, 9-dimethylxanthene.
Preparation of intermediates
Intermediate 1: preparation of 1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropane-1-amine
Figure BDA0003263976850000221
Step 1: preparation of 5-fluoro-2-methoxynicotinonitrile
3-bromo-5-fluoro-2-methoxypyridine (11.0g,53.7mmol) was dissolved in NMP (110mL) and cuprous cyanide (24.0g,268.5mmol) in N was added to it2The temperature was raised to 160 ℃ with protection and stirred for 6 hours. TLC showed the starting material was reacted, the solid was filtered and the filter cake was washed with EA, water (1.1L) was added to the filtrate, and after stirring, the solid was removed by filtration. The filtrate was extracted twice with EA, the filtrates were combined and taken with anhydrous Na2SO4Dry and concentrate to dryness to give the crude product as a brown solid. The solid was dissolved in EA (20mL) and petroleum ether (200mL) was added slowly to precipitate a solid. The precipitated solid was filtered and dried to obtain the objective compound (7.3g, yield 90%) as an off-white solid.
Step 2: preparation of 1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropane-1-amine
5-fluoro-2-methoxynicotinonitrile (7.3g,48.0mmol) and tetraisopropyl titanate (16.4g,57.6mmol) were dissolved in anhydrous tetrahydrofuran (80mL), and the reaction system was purged with N2Displaced and cooled to-70 ℃. Ethyl magnesium bromide (106mL, 106mmol,1M in THF), the internal temperature was controlled to not higher than-65 ℃. After the addition, the reaction was continued for 10 minutes while maintaining the temperature, the low temperature bath was removed and slowly returned to room temperature, and stirring was carried out for 2 hours. Boron trifluoride ethyl ether (13.6g, 96.0mmol) is added into the system, the reaction is quenched by 1N diluted hydrochloric acid, and the pH is adjusted to 9-10 by NaOH. Filtered and the filtrate concentrated to dryness. The residue was purified by column chromatography (PE/EA ═ 10/1) using crude product to give the target compound (3.1g, yield 35%) as a yellow solid.
EM (calculated): 182.1; MS (ESI) M/z (M + H) +: 183.2
1H NMR(400MHz,CDCl3)δ1.05-1.08(2H,m),1.45-1.48(2H,m),4.00(3H,s),7.28(1H,dd,J=8.2Hz,2.8Hz),8.02(1H,d,J=2.8Hz).
Intermediate 2: preparation of ethyl 5-chloro-6-fluoropyrazolo [1,5-a ] pyrimidine-3-carboxylate
Figure BDA0003263976850000231
Step 1: preparation of 2-fluoro malonic acid
NaOH (4.5g,113mmol) was dissolved in water (25mL), to which was added EtOH (100mL) and warmed to 60 ℃. Diethyl 2-fluoromalonate (10.0g,56.2mmol) was slowly added dropwise to the reaction system, and a large amount of white solid was precipitated. After stirring for 2 hours with incubation, TLC indicated the starting material was reacted. The solid was filtered and spun dry, dissolved in 4N hydrochloric acid and stirred at room temperature for 1 hour. The reaction solution was concentrated to dryness, cooled, added with MTBE, stirred and filtered, and the filtrate was concentrated to dryness to obtain the objective compound (6.7g, yield 98%) as a white solid.
EM (calculated): 122.0; MS (ESI) M/z (M-H) -: 121.0
Step 2: preparation of ethyl 5, 7-dichloro-6-fluoropyrazolo [1,5-a ] pyrimidine-3-carboxylate
2-Fluoromalonic acid (3.0g,24.6mmol) and ethyl 5-amino-1H-pyrazole-4-carboxylate (3.8g,24.6mmol) were dissolved in POCl3To the reaction mixture (25mL) were added DMF (5mL) and N, N-diethylaniline (3.7g,24.6mmol), and the mixture was heated to 90 ℃ to react for 6 hours. TLC showed starting materialAnd (5) finishing the reaction. The reaction solution was concentrated to dryness, and saturated Na was slowly added to the residue2CO3Aqueous solution, stirred and extracted 3 times with DCM, the organic phases were combined and taken over anhydrous Na2SO4Drying and concentrating to dryness. The crude product was purified by column chromatography (PE/EA ═ 10/1) to give the desired compound (870mg, yield 13%) as a yellow solid.
EM (calculated): 277.0; MS (ESI) M/z (M + H) +: 278.1
And step 3: preparation of ethyl 5-chloro-6-fluoropyrazolo [1,5-a ] pyrimidine-3-carboxylate
Reacting 5, 7-dichloro-6-fluoropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (850mg,3.1mmol), zinc powder (212mg,3.3mmol) and NH4Cl (425mg,8.0mmol) was dispersed in a mixed solvent of EtOH/THF/water (48mL,3/1/2) and stirred at room temperature for 20 min. TLC showed the starting material was reacted. The reaction solution was filtered, and the filtrate was concentrated to dryness to give the objective compound (750mg, yield 100%) as a yellow solid.
EM (calculated): 243.0, respectively; MS (ESI) M/z (M + H) +: 244.1
1H NMR(400MHz,d6-DMSO)δ1.44(3H,t,J=6.8Hz),4.44(2H,q,J=6.8Hz),8.59(1H,s),8.74(1H,d,J=3.2Hz).
Intermediate 3: preparation of ethyl 5- ((1- (5-fluoro-2-hydroxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
Figure BDA0003263976850000241
Step 1: preparation of ethyl 5- ((1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropane-1-amine (500mg,2.7mmol), 5-chloropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (618mg,2.7mmol) was dissolved in acetonitrile (10mL), DIEA (697mg,5.4mmol) was added, and the reaction was stirred at 80 ℃ for 5 h. TLC showed the consumption of starting material was complete. Concentrating the reaction system to dryness, adding water to the residue, extracting with DCM for 3 times, combining the organic phases, anhydrous Na2SO4Drying and spin-drying. The crude product was purified by column chromatography (DCM/MeOH 100/1-20/1) to give the title compound (620mg, 62% yield) as a yellow solid.
EM (calculated): 371.1, respectively; MS (ESI) M/z (M + H) +: 372.2
Step 2: preparation of ethyl 5- ((1- (5-fluoro-2-hydroxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 5- ((1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (600mg,1.6mmol) was dissolved in DCM (20mL), TMSCl (1.7g,16.2mmol) and potassium iodide (2.7g,16.2mmol) were added in that order, and the mixture was stirred at room temperature for 3 hours. TLC showed the consumption of starting material was complete. Slowly adding 20% Na into the reaction solution2S2O3Aqueous solution, organic phase after washing, and anhydrous Na2SO4Drying and concentrating to dryness. The crude product was purified by column chromatography (DCM/MeOH 100/1-20/1) to give the title compound (520mg, 91% yield) as a yellow solid.
EM (calculated): 357.1; MS (ESI) M/z (M + H) +: 358.2
1H NMR(400MHz,d6-DMSO)δ1.04-1.10(4H,m),1.35(3H,t,J=8.0Hz),4.30(2H,q,J=8.0Hz),6.39(1H,d,J=8.0Hz),7.48(1H,s),8.16(1H,s),8.20(1H,dd,J=8.0Hz,3.2Hz),8.50(1H,d,J=8.0Hz),8.68(1H,s),11.47(1H,brs).
Intermediate 4: preparation of ethyl 6-fluoro-5- ((1- (5-fluoro-2-hydroxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
Figure BDA0003263976850000251
Step 1: preparation of ethyl 6-fluoro-5- ((1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropane-1-amine (328mg,1.8mmol), 5-chloro-6-fluoropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (437mg,1.8mmol) dissolved in CH3CN (12mL), DIEA (697mg,5.4mmol) was added to the reaction systemStirred at 80 ℃ overnight. TLC showed the consumption of starting material was complete. Concentrating the reaction system to dryness, adding water to the residue, extracting with DCM for 3 times, combining the organic phases, anhydrous Na2SO4Drying and spin-drying. The crude product was purified by column chromatography (PE/EA/DCM/═ 10/1/1) to give the title compound (550mg, yield 79%) as a yellow solid.
EM (calculated): 389.1, respectively; MS (ESI) M/z (M + H) +: 390.2
Step 2: preparation of ethyl 6-fluoro-5- ((1- (5-fluoro-2-hydroxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
Mixing compound 6-fluoro-5- ((1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (540mg,1.4mmol) was dissolved in DCM (20mL), and TMSCl (1.53g,14.0mmol) and potassium iodide (2.32g,14.0mmol) were added to the solution in this order, and the mixture was stirred at room temperature for 3 hours. TLC showed the consumption of starting material was complete. The reaction solution was mixed with 10% Na2S2O3Washing, separating liquid, collecting organic phase, and adding anhydrous Na2SO4Drying and concentration to dryness gave the title compound (480mg, yield 91%) as a yellow solid.
EM (calculated): 375.1 of the total weight of the product; MS (ESI) M/z (M + H) +: 376.2
1H NMR(400MHz,d6-DMSO)δ1.18-1.19(4H,m),1.34(3H,t,J=7.2Hz),4.30(2H,q,J=7.2Hz),7.48(1H,s),8.17(1H,dd,J=8.4Hz,3.2Hz),8.21(1H,s),8.71(1H,s),9.06(1H,d,J=6.4Hz),11.44(1H,brs).
Intermediate 5: preparation of ethyl 5- ((1- (5-fluoro-2-hydroxyphenyl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
Figure BDA0003263976850000261
Step 1: preparation of 1- (5-fluoro-2-methoxyphenyl) cyclopropane-1-amine
The compound 5-fluoro-2-methoxybenzonitrile (5.0g,33.1mmol) and tetraisopropyl titanate (11.3g,39.7mmol) were dissolved in anhydrous THF (50mL), and the reaction system was purged with N2Displaced and cooled to-70 ℃.Ethyl magnesium bromide (73mL, 73mmol, 1M in THF) was slowly added dropwise to the system, with the internal temperature controlled to be not higher than-65 ℃. After the addition, the reaction was continued for 10 minutes while maintaining the temperature, the low temperature bath was removed and slowly returned to room temperature, and stirring was carried out for 2 hours. Boron trifluoride ethyl ether (7.1g, 49.7mmol) is added into the system, the reaction is quenched by 1N diluted hydrochloric acid, and the pH is adjusted to 9-10 by sodium hydroxide. Filtered and the filtrate concentrated to dryness. The residue was purified by column chromatography (PE/EA 10/1) to obtain the objective compound (2.4g, yield 40%) as a yellow liquid.
EM (calculated): 181.1, respectively; MS (ESI) M/z (M + H) +: 182.2
Step 2: preparation of 2- (1-aminocyclopropyl) -4-fluorophenol
Compound 1- (5-fluoro-2-methoxyphenyl) cyclopropane-1-amine (2.3g,12.7mmol) is dissolved in anhydrous DCM (30mL) and the reaction system is treated with N2The displacement was performed 3 times and cooled to-70 ℃. Boron tribromide (93.6g, 63.5mmol, 17% solution in dichloromethane) was slowly added dropwise to the system with the internal temperature controlled to be not higher than-60 ℃. After the addition, the reaction was continued for 20 minutes while maintaining the temperature, the low temperature bath was removed and slowly returned to room temperature, and stirring was carried out for 2 hours. After completion of the reaction, methanol was slowly added dropwise to the reaction system to quench, the reaction solution was concentrated to dryness, and the residue was purified by column chromatography (DCM/MeOH 20/1) to obtain the objective compound (1.1g, yield 52%) as a yellow oil.
EM (calculated): 167.1; MS (ESI) M/z (M + H) +: 168.2
And step 3: preparation of ethyl 5- ((1- (5-fluoro-2-hydroxyphenyl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 2- (1-aminocyclopropyl) -4-fluorophenol (500mg,3.0mmol), 5-chloro-6-fluoropyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (675mg,3.0mmol) dissolved in CH3CN (12mL), DIEA (774mg,6.0mmol) was added and the reaction stirred at 80 ℃ overnight. TLC showed the consumption of starting material was complete. Concentrating the reaction system to dryness, adding water to the residue, extracting with DCM for 3 times, combining the organic phases, anhydrous Na2SO4Drying and spin-drying. The crude product was purified by column chromatography (PE/EA/DCM/═ 10/1/1) to give the title compound (415mg, 39% yield)) It is a yellow solid.
EM (calculated): 356.1; MS (ESI) M/z (M + H) +: 357.2
1H NMR(400MHz,d6-DMSO)δ1.07-1.12(4H,m),1.37(3H,t,J=8.0Hz),4.35(2H,q,J=8.0Hz),6.36(1H,d,J=8.0Hz),7.42(1H,s),8.03(1H,s),8.08(1H,d,J=7.2Hz),8.15(1H,dd,J=7.2Hz,2.4Hz),8.37(1H,d,J=8.0Hz),8.56(1H,s),10.12(1H,brs).
Intermediate 6: preparation of ethyl 6-bromo-5- ((1- (2- ((1- (((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
Figure BDA0003263976850000271
Step 1: preparation of ethyl 6-bromo-5- ((1- (5-fluoro-2-hydroxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 2- (1-aminocyclopropyl) -4-fluorophenol (1.0g,6.0mmol), 6-bromo-5-chloropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (1.8g,6.0mmol) dissolved in CH3CN (30mL), DIEA (930mg,7.2mmol) was added, and the reaction was stirred at 80 ℃ for 6 h. TLC showed the consumption of starting material was complete. Concentrating the reaction system to dryness, adding water to the residue, extracting with DCM for 3 times, combining the organic phases, anhydrous Na2SO4Drying and spin-drying. The crude product was purified by column chromatography (PE/EA/DCM/═ 10/1/1) to give the title compound (940mg, 36% yield) as a yellow solid.
EM (calculated): 435.0, respectively; MS (ESI) M/z (M + H) +: 436.1
Step 2: preparation of ethyl 6-bromo-5- ((1- (2- ((1- (((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 6-bromo-5- ((1- (5-fluoro-2-hydroxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (920mg, 2.1mmol), (1- (hydroxymethyl) cyclopropyl) carbamic acid tert-butyl ester (598mg, 3.2mmol) and PPh3(1.1g, 4.2mmol) was dissolved in anhydrous THF (30mL)Is made of N2The substitution was carried out three times. DIAD (850mg, 4.2mmol) was slowly added to the reaction system, which was yellow and clear. After 1 hour at room temperature, the reaction was completed. Concentrating the reaction solution, adding the residue into water, extracting twice with ethyl acetate, combining the organic phases and using anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by column chromatography (DCM/MeOH ═ 30/1) to give the title compound (523mg, yield 41%) as a yellow solid.
EM (calculated): 604.1; MS (ESI) M/z (M + H) +: 605.2
EXAMPLES Compounds
Example 15 oxa-2, 8-diaza-45-fluoro-1 (5,3) -pyrazolo [1,5-a]Preparation of pyrimidino-4 (3,2) -pyridino-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one
Figure BDA0003263976850000281
Step 1: preparation of ethyl 5- ((1- (2- ((1- ((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
Compound 5- ((1- (5-fluoro-2-hydroxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (500mg, 1.40mmol), (1- (hydroxymethyl) cyclopropyl) carbamic acid tert-butyl ester (314mg, 1.68mmol) and PPh3(734mg, 2.80mmol) was dissolved in anhydrous THF (20mL) using N2The substitution was carried out three times. DIAD (566mg, 2.80mmol) was slowly added to the reaction system, which was yellow and clear. After 1 hour at room temperature, the reaction was completed. Concentrating the reaction solution, adding the residue into water, extracting twice with ethyl acetate, combining the organic phases and using anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by column chromatography (DCM/MeOH ═ 25/1) to give the title compound (278mg, yield 38%) as a yellow oily solid.
EM (calculated): 526.2, respectively; MS (ESI) M/z (M + H) +: 527.3
Step 2: preparation of ethyl 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 5- ((1- (2- ((1- ((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (250mg, 0.48mmol) was dissolved in HCl/EA (15mL) and stirred at room temperature for 1 hour. After the reaction, the reaction solution was concentrated to dryness. The residue was dissolved in water (15mL) and extracted with EA three times. Collecting the water phase, adjusting the pH value to 8-9 by using saturated sodium carbonate water solution, and extracting for three times by using DCM. The organic phases were combined and washed with anhydrous Na2SO4After drying and concentration, the title compound (183mg, yield 89%) was obtained as an off-white solid.
EM (calculated): 426.2 of the total weight of the mixture; MS (ESI) M/z (M + H) +: 427.2
And step 3: preparation of 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
Mixing compound 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (175mg, 0.41mmol) dissolved in EtOH/H2To O (10mL/1mL), NaOH (49mg, 1.23mmol) was added to the reaction mixture, and the mixture was heated to 60 ℃ to react overnight. After completion of the reaction, the reaction solution was adjusted to pH 7 with dilute hydrochloric acid. Concentrate to dryness to give the crude product as a white solid. The solid was washed with DCM/EtOH (30mL,5/1), filtered and the filtrate was concentrated to give the title compound (120mg, 74% yield) as a pale yellow oil.
EM (calculated): 398.2; MS (ESI) M/z (M + H) +: 399.2
And 4, step 4: 5-oxa-2, 8-diaza-45-fluoro-1 (5,3) -pyrazolo [1,5-a]Preparation of pyrimidino-4 (3,2) -pyridino-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one
Mixing compound 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (115mg, 0.29mmol), HATU (165mg, 0.44mmol) and DIEA (75mg, 0.58mmol) were dispersed in THF (20mL) and stirred at room temperature for 4 h. After the reaction is finished, concentrating the reaction solution, and adding the remainder into the reaction solutionExtracting with DCM twice in water, combining the organic phases and adding anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by column chromatography (DCM/MeOH ═ 30/1) to give the title compound (65mg, yield 59%) as a white solid.
EM (calculated): 380.1, respectively; MS (ESI) M/z (M + H) +: 381.2
1H NMR(400MHz,d6-DMSO)δ0.79-0.85(2H,m),0.89-0.99(2H,m),1.21-1.31(2H,m),2.00-2.04(1H,m),2.08-2.12(1H,m),3.63(1H,d,J=10.4Hz),4.72(1H,d,J=9.2Hz),6.28(1H,d,J=7.6Hz),7.85(1H,dd,J=8.6Hz,2.8Hz),8.02(1H,s),8.03(1H,d,J=2.8Hz),8.57(1H,d,J=7.6Hz),9.04(1H,s),9.51(1H,s).
The following example compounds were synthesized according to the procedure set forth in example 1:
TABLE 1
Figure BDA0003263976850000291
Figure BDA0003263976850000301
Figure BDA0003263976850000311
Example 65 oxa-2, 8-diaza-16,45-difluoro-1 (5,3) -pyrazolo [1,5-a]Preparation of pyrimidino-4 (3,2) -pyridino-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one
Figure BDA0003263976850000321
Step 1: preparation of ethyl 5- ((1- (2- ((1- ((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6-fluoropyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 6-fluoro-5- ((1- (5-fluoro-2-hydroxypyridine-3-)Yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (460mg, 1.23mmol), (1- (hydroxymethyl) cyclopropyl) carbamic acid tert-butyl ester (346mg, 1.85mmol) and PPh3(645mg, 2.46mmol) was dissolved in anhydrous THF (20mL) using N2The substitution was carried out three times. DIAD (497mg, 2.46mmol) was slowly added to the reaction system, which was yellow and clear. After 1 hour at room temperature, the reaction was completed. Concentrating the reaction solution, adding the residue into water, extracting twice with ethyl acetate, combining the organic phases and using anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by column chromatography (DCM/MeOH ═ 25/1) to give the title compound (294mg, yield 44%) as a yellow oily solid.
EM (calculated): 544.2, respectively; MS (ESI) M/z (M + H) +: 545.3
Step 2: preparation of ethyl 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6-fluoropyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 5- ((1- (2- ((1- ((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6-fluoropyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (280mg, 0.51mmol) was dissolved in HCl/EA (15mL) and stirred at room temperature for 1 hour. After the reaction, the reaction solution was concentrated to dryness. The residue was dissolved in water (15mL) and extracted with EA three times. Collecting the water phase, adjusting the pH value to 8-9 by using saturated sodium carbonate water solution, and extracting for three times by using DCM. The organic phases were combined and washed with anhydrous Na2SO4After drying and concentration, the title compound (193mg, yield 85%) was obtained as an off-white solid.
EM (calculated): 444.2, respectively; MS (ESI) M/z (M + H) +: 445.3
And step 3: preparation of 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6-fluoropyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
Mixing compound 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6-fluoropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (175mg, 0.39mmol) dissolved in EtOH/H2To O (10mL/1mL), NaOH (47mg, 1.17mmol) was added to the reaction mixture, and the mixture was heated to 60 ℃ to react overnight. After the reaction is completedThe reaction solution was adjusted to pH 7 with dilute hydrochloric acid. Concentrate to dryness to give the crude product as a white solid. The solid was washed with DCM/EtOH (30mL,5/1), filtered and the filtrate was concentrated to give the title compound (123mg, 76% yield) as a pale yellow oil.
EM (calculated): 416.1; MS (ESI) M/z (M + H) +: 417.2
Step 45-oxa-2, 8-diaza-16,45-difluoro-1 (5,3) -pyrazolo [1,5-a]Preparation of pyrimidino-4 (3,2) -pyridino-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one
Mixing compound 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6-fluoropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid (115mg, 0.28mmol), HATU (165mg, 0.44mmol) and DIEA (75mg, 0.58mmol) were dispersed in THF (20mL) and stirred at room temperature for 4 h. After the reaction, the reaction solution was concentrated, the residue was added to water, extracted twice with DCM, and the organic phases were combined and extracted with anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by column chromatography (DCM/MeOH ═ 30/1) to give the title compound (29mg, yield 26%) as a white solid.
EM (calculated): 398.1; MS (ESI) M/z (M + H) +: 399.2
1H NMR(400MHz,d6-DMSO)δ0.81-0.84(2H,m),0.96-0.98(2H,m),1.17-1.20(1H,m),1.39-1.42(1H,m),2.01-2.07(1H,m),2.09-2.13(1H,m),3.64(1H,d,J=11.2Hz),4.70(1H,d,J=11.2Hz),7.99(1H,dd,J=8.8Hz,2.8Hz),8.04(1H,d,J=3.2Hz),8.06(1H,s),9.15(1H,d,J=6.4Hz),9.27(1H,s),9.36(1H,s).
The following example compounds were synthesized according to the procedure set forth in example 6:
TABLE 2
Figure BDA0003263976850000331
Figure BDA0003263976850000341
Examples75-oxa-2, 8-diaza-44-fluoro-1 (5,3) -pyrazolo [1,5-a]Preparation of pyrimidino-4 (2,1) -benz-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one
Figure BDA0003263976850000342
Step 1: preparation of (1- (tert-butoxycarbonyl) amino) cyclopropyl) methyl 4-methylbenzenesulfonate
The compound tert-butyl (1- (hydroxymethyl) cyclopropyl) carbamate (5.0g, 26.7mmol) was dissolved in DCM (50mL), to which TsCl (5.6g, 29.4mmol) and DIEA (5.2g, 40.1mmol) were added in order, and the reaction was stirred at room temperature overnight. After the reaction, the reaction solution was added to water, and the solution was separated. The aqueous phase was extracted twice with DCM, the organic phases were combined and washed with anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by column chromatography (PE/EA ═ 5/1) to give the objective compound (7.3g, yield 80%) as a white solid.
EM (calculated): 341.1, respectively; MS (ESI) M/z (M + H) +: 342.2
Step 2: preparation of ethyl 5- ((1- (2- ((1- ((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluorophenyl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
Compound 5- ((1- (5-fluoro-2-hydroxyphenyl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (400mg,1.1mmol) was dissolved in DMF (10mL), and (1- (tert-butoxycarbonyl) amino) cyclopropyl) methyl 4-methylbenzenesulfonate (375mg,1.1mmol) and K were added to the reaction in this order2CO3(304mg,2.2mmol) and the reaction was stirred at 100 ℃ overnight. Adding water to the reaction solution, extracting with DCM for 3 times, combining organic phases, washing with saturated saline solution, separating liquid, collecting organic phase, and collecting anhydrous Na2SO4Drying and spin-drying. The crude product was purified by column chromatography (DCM/MeOH ═ 30/1) to give the title compound (286mg, 49% yield) as a yellow solid.
EM (calculated): 525.2, respectively; MS (ESI) M/z (M + H) +: 526.2
And step 3: preparation of ethyl 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluorophenyl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 5- ((1- (2- ((1- ((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluorophenyl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (270mg, 0.51mmol) was dissolved in HCl/EA (15mL) and stirred at room temperature for 1 hour. After the reaction, the reaction solution was concentrated to dryness. The residue was dissolved in water (15mL) and extracted with EA three times. Collecting the aqueous phase, adding saturated Na2CO3Adjusting the pH value of the aqueous solution to 8-9, and extracting with DCM for three times. The organic phases were combined and washed with anhydrous Na2SO4After drying and concentration, the title compound (156mg, yield 72%) was obtained as an off-white solid.
EM (calculated): 425.2; MS (ESI) M/z (M + H) +: 426.3
And 4, step 4: preparation of 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluorophenyl) cyclopropyl) amino) pyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
Mixing compound 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluorophenyl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (150mg, 0.35mmol) dissolved in EtOH/H2To O (10mL/1mL), NaOH (56mg, 1.40mmol) was added to the reaction mixture, and the mixture was heated to 70 ℃ to react overnight. After completion of the reaction, the reaction solution was adjusted to pH 7 with dilute hydrochloric acid. Concentrate to dryness to give the crude product as a white solid. The solid was washed with DCM/EtOH (30mL,5/1), filtered and the filtrate was concentrated to give the title compound (103mg, 74% yield) as a pale yellow oil.
EM (calculated): 397.2; MS (ESI) M/z (M + H) +: 398.2
And 5: 5-oxa-2, 8-diaza-44-fluoro-1 (5,3) -pyrazolo [1,5-a]Preparation of pyrimidino-4 (2,1) -benz-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one
Mixing compound 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluorophenyl) cyclopropyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (98mg, 0.25mmol), HBTU (144mg, 0.38mmol) and DIEA (65mg, 0.50mmol) were dispersed in THF (10mL) and stirred at room temperature for 4 h. After the reaction, the reaction solution was concentrated, and the residue was added to water and extracted twice with DCMCombining the organic phases and using anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by TLC (DCM/MeOH ═ 20/1) to give the title compound (19mg, yield 20%) as a white solid.
EM (calculated): 379.1, respectively; MS (ESI) M/z (M + H) +: 380.2
1H NMR(400MHz,d6-DMSO)δ0.78-0.87(4H,m),0.94-0.97(1H,m),1.28-1.30(1H,m),2.05-2.08(1H,m),2.12-2.15(1H,m),3.70(1H,d,J=9.2Hz),4.16(1H,d,J=9.2Hz),6.27(1H,d,J=7.6Hz),6.82(1H,dd,J=9.2Hz,4.8Hz),6.98-7.03(1H,m),7.35(1H,dd,J=10.0Hz,3.2Hz),8.02(1H,s),8.56(1H,d,J=7.6Hz),9.08(1H,s),9.71(1H,s).
Example 85-oxa-2, 8-diaza-45-fluoro-1 (5,3) -pyrazolo [1-a]Preparation of pyridina-4 (3,2) -pyridina-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane-heterocyclic nonapan-9-ones
Figure BDA0003263976850000361
Step 1: preparation of ethyl 5- ((tert-butoxycarbonyl) amino) pyrazolo [1,5-a ] pyridine-3-carboxylate
The compound tert-butyl pyridin-4-ylcarbamate (7.0g, 36.06mmol) was dissolved in acetonitrile (70mL), to which O- (2, 4-dinitrophenyl) hydroxylamine (7.2g, 36.06mmol) was added and stirred at room temperature overnight. The reaction was concentrated to dryness to give a yellow solid. The solid was dissolved in DMF (100mL) and K was added to it sequentially2CO3(6.0g, 43.27mmol) and ethyl propiolate (3.5g, 36.06mmol), and the mixture was stirred at room temperature for 3 hours. After completion of the reaction, the reaction mixture was added to water (1000mL) with stirring, and extracted three times with EA. The combined organic phases were washed successively with water and saturated aqueous NaCl solution, dried over anhydrous sodium sulfate and concentrated to dryness. The residue was purified by column chromatography (PE/EA 4/1) to give the objective compound (3.3g, yield 30%) as a yellow solid.
EM (calculated): 305.1; MS (ESI) M/z (M + H) +: 306.2
Step 2: preparation of ethyl 5-iodopyrazolo [1,5-a ] pyridine-3-carboxylate
Compound ethyl 5- ((tert-butoxycarbonyl) amino) pyrazolo [1,5-a ] pyridine-3-carboxylate (3.3g, 10.82mmol) was dissolved in DCM (50mL), cooled with stirring in an ice-water bath and TFA (15mL) was added dropwise thereto. After the dropwise addition, the ice-water bath was removed, and the mixture was returned to room temperature and stirred for 3 hours. After TLC showed the reaction was complete, the reaction was concentrated to dryness to give a brown oil.
The oil was dissolved in concentrated hydrochloric acid (30mL), to which concentrated sulfuric acid (3mL) was added dropwise under ice-cooling. Adding NaNO2(821mg, 11.90mmol) was dissolved in water (15mL), slowly added dropwise to the reaction system, and stirred for 15min while maintaining an ice-water bath. A small amount of urea (50mg) was added to the reaction system and stirring was continued for 15 min. KI (2.89g, 17.31mmol) solid was added to the reaction system at one time, and the reaction solution was returned to room temperature and stirred for 30 min. The reaction solution was adjusted to pH 3 with solid NaOH and extracted 2 times with DCM. The organic phases were combined and dried over anhydrous sodium sulfate and concentrated to dryness. The residue was purified by column chromatography (PE/EA 10/1-7/1) to give the title compound (2.0g, yield 58%) as a yellow solid.
EM (calculated): 316.0; MS (ESI) M/z (M + H) +: 317.1
And step 3: preparation of ethyl 5- ((1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyridine-3-carboxylate
The compound 5-iodopyrazolo [1, 5-a)]Pyridine-3-carboxylic acid ethyl ester (2.0g, 6.33mmol), 1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropane-1-amine (1.4g, 7.60mmol), Pd2(dba)3(577mg, 0.63mmol), X-phos (600mg, 1.26mmol) and cesium carbonate (6.2g, 18.99mmol) were dispersed in toluene (30 mL). The reaction flask was placed in an oil bath pan preheated to 95 ℃ and stirred for 2h under nitrogen protection. After the reaction was complete, the reaction was cooled and filtered, the filter cake was washed with DCM and the filtrate was concentrated to dryness. The residue was purified by column chromatography (PE/EA 10/1-2/1) to give the title compound (600mg, 26% yield) as a yellow solid.
EM (calculated): 370.1 of the total weight of the mixture; MS (ESI) M/z (M + H) +: 371.2
And 4, step 4: preparation of ethyl 5- ((1- (5-fluoro-2-hydroxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyridine-3-carboxylate
The compound 5- ((1- (5-fluoro-2-methoxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyridine-3-carboxylic acid ethyl ester (600mg, 1.62mmol) was dissolved in acetonitrile (15mL) and stirred at room temperature. KI (2.69g, 16.2mmol) and TMSCl (2.69g, 16.2mmol) were added successively thereto, and stirring was continued for 2 h. After the reaction was complete, most of the acetonitrile was concentrated. The residue was dissolved in DCM and Na was added2SO3And (4) washing with an aqueous solution. The organic phase was collected, dried over anhydrous sodium sulfate and concentrated to dryness. The residue was washed with PE/EA (30mL,10/1), and dried to give the title compound (520mg, yield 90%) as a yellow solid.
EM (calculated): 356.1; MS (ESI) M/z (M + H) +: 357.2
And 5: preparation of ethyl 5- ((1- (1- (2- ((1- ((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyridine-3-carboxylate
Compound 5- ((1- (5-fluoro-2-hydroxypyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyridine-3-carboxylic acid ethyl ester (300mg, 0.84mmol), (1- (hydroxymethyl) cyclopropyl) carbamic acid tert-butyl ester (236mg, 1.26mmol) and PPh3(440mg, 1.68mmol) was dissolved in anhydrous THF (10mL) using N2The substitution was carried out three times. DIAD (340mg, 1.68mmol) was slowly added to the reaction system, which was yellow and clear. After 1 hour at room temperature, the reaction was completed. Concentrating the reaction solution, adding the residue into water, extracting twice with ethyl acetate, combining the organic phases and using anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by column chromatography (DCM/MeOH ═ 50/1) to give the title compound (207mg, yield 47%) as a yellow oily solid.
EM (calculated): 525.2, respectively; MS (ESI) M/z (M + H) +: 526.3
Step 6: preparation of 5- ((1- (2- (((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a ] pyridine-3-carboxylic acid
The compound 5- ((1- (1- (2- ((1- ((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyridine-3-carboxylic acid ethyl ester (200mg, 0.3)8mmol) was dissolved in HCl/EA (10mL) and stirred at room temperature for 1 hour. After the reaction, the reaction solution was concentrated to dryness. The residue was dissolved in EtOH/H2To O (11mL,10/1), NaOH (152mg, 3.80mmol) was added to the reaction mixture, and the temperature was raised to 50 ℃ to react for 4 hours. After completion of the reaction, the reaction solution was adjusted to pH 7 with dilute hydrochloric acid. Concentrate to dryness to give the crude product as a white solid. The solid was washed with DCM/EtOH (20mL,5/1), filtered and the filtrate was concentrated to give the title compound (75mg, 50% yield) as a pale yellow solid.
EM (calculated): 397.2; MS (ESI) M/z (M + H) +: 398.3
And 7: 5-oxa-2, 8-diaza-45-fluoro-1 (5,3) -pyrazolo [1-a]Preparation of pyridina-4 (3,2) -pyridina-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane-heterocyclic nonapan-9-ones
The compound 5- ((1- (2- (((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo [1,5-a]Pyridine-3-carboxylic acid (75mg, 0.19mmol), HATU (144mg, 0.38mmol) and DIEA (74mg, 0.57mmol) were dispersed in THF (15mL) and stirred at room temperature for 4 h. After the reaction, the reaction solution was concentrated, the residue was added to water, extracted twice with DCM, and the organic phases were combined and extracted with anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by column chromatography (DCM/MeOH ═ 30/1) to give the title compound (27mg, yield 38%) as an off-white solid.
EM (calculated): 379.1, respectively; MS (ESI) M/z (M + H) +: 380.2
1H NMR(400MHz,d6-DMSO)δ0.79-0.83(1H,m),0.90-1.05(4H,m),1.13-1.21(2H,m),1.73-1.78(1H,m),3.62(1H,d,J=11.6Hz),4.74(1H,d,J=11.6Hz),6.33(1H,dd,J=7.4Hz,2.4Hz),7.60(1H,s),7.88(1H,s),7.92(1H,dd,J=8.8Hz,2.8Hz),8.04(1H,d,J=3.2Hz),8.09(1H,s),8.28(1H,d,J=2.0Hz),8.31(1H,d,J=7.6Hz).
Example 155-oxa-2, 8-diaza-16-hydroxy-45-fluoro-1 (5,3) -pyrazolo [1,5-a]Preparation of pyrimidino-4 (3,2) -pyridino-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one
Figure BDA0003263976850000381
Step 1: preparation of 5- ((1- (2- (((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6-hydroxypyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
Mixing compound 5- ((1- (2- ((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6-fluoropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (200mg, 0.45mmol) dissolved in EtOH/H2To O (10mL/5mL), NaOH (180mg, 4.5mmol) was added to the reaction mixture, and the mixture was heated to 80 ℃ to react overnight. After completion of the reaction, the reaction solution was adjusted to pH 7 with dilute hydrochloric acid. Concentrate to dryness to give the crude product as a white solid. The solid was washed with DCM/EtOH (10mL,5/1), filtered and the filtrate was concentrated to give the title compound (97mg, 52% yield) as an off-white solid.
EM (calculated): 414.1, respectively; MS (ESI) M/z (M-H) -: 413.0
Step 2: 5-oxa-2, 8-diaza-16-hydroxy-45-fluoro-1 (5,3) -pyrazolo [1,5-a]Preparation of pyrimidino-4 (3,2) -pyridino-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one
The compound 5- ((1- (2- (((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6-hydroxypyrazolo [1, 5-a)]Pyrimidine-3-carboxylic acid (90mg, 0.22mmol), HATU (165mg, 0.44mmol) and DIEA (85mg, 0.66mmol) were dispersed in THF (30mL) and stirred at room temperature for 4 h. After the reaction, the reaction solution was concentrated, the residue was added to water, extracted twice with DCM, and the organic phases were combined and extracted with anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by column chromatography (DCM/MeOH ═ 30/1) to give the title compound (12mg, yield 14%) as a white solid.
EM (calculated): 396.1, respectively; MS (ESI) M/z (M + H) +: 397.2
1H NMR(400MHz,d6-DMSO)δ0.82-0.84(2H,m),0.96-0.99(2H,m),1.18-1.23(1H,m),1.40-1.43(1H,m),2.07-2.13(2H,m),3.65(1H,d,J=11.0Hz),4.68(1H,d,J=11.2Hz),7.96-8.04(3H,m),9.14(1H,s),9.25(1H,s),9.38(1H,s),9.56(1H,brs).
Example 165-oxa-2, 8-diaza-16-methoxy-45-fluoro-1 (5,3) -pyrazolo [1,5-a]Preparation of pyrimidino-4 (3,2) -pyridino-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one
Figure BDA0003263976850000391
The compound 5-oxa-2, 8-diaza-16-hydroxy-45-fluoro-1 (5,3) -pyrazolo [1,5-a]Pyrimidizidine-4 (3,2) -pyrididine-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one (10mg, 0.03mmol), CH3I (4mg, 0.03mmol) and K2CO3(8mg, 0.06mmol) was dispersed in acetonitrile (2mL) and stirred at room temperature for 2 hours. After completion of the reaction, the reaction was concentrated, and the residue was purified by TLC (DCM/MeOH ═ 40/1) to give the title compound (3mg, yield 24%) as a white solid.
EM (calculated): 410.2; MS (ESI) M/z (M + H) +: 411.2
1H NMR(400MHz,d6-DMSO)δ0.80-0.84(2H,m),0.93-0.99(2H,m),1.25-1.34(2H,m),2.09-2.20(2H,m),3.67(1H,d,J=11.0Hz),3.95(3H,s),4.73(1H,d,J=11.2Hz),7.93-8.00(3H,m),9.12(1H,s),9.20(1H,s),9.36(1H,s).
Example 175 oxa-2, 8-diaza-16-dimethylphosphoryl-45-fluoro-1 (5,3) -pyrazolo [1,5-a]Preparation of pyrimidino-4 (3,2) -pyridino-3 (1,1) -cyclopropane-7 (1,1) -cyclopropane heterocycle nonapan-9-one
Figure BDA0003263976850000401
Step 1: preparation of ethyl 5- ((1- (1- (2- ((1- ((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) aminoethyl-6- (dimethylphosphoryl) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
The compound 6-bromo-5- ((1- (2- ((1- (((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) pyrazolo[1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (100mg, 0.17mmol), dimethylphosphine oxide (53mg, 0.68mmol), Pd2(dba)3(18mg, 0.02mmol), xant-phos (23mg, 0.04mmol) and Et3N (52mg, 0.51mmol) was dispersed in 1,4-dioxane (5mL) and reacted overnight at 100 ℃ under nitrogen. After completion of the reaction, the reaction solution was concentrated, and the residue was purified by column chromatography (DCM/MeOH 40/1) to obtain the objective compound (77mg, yield 75%) as an off-white solid.
EM (calculated): 602.2; MS (ESI) M/z (M + H) +: 603.2
Step 2: preparation of 5- ((1- (2- (((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6- (dimethylphosphoryl) pyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
The compound 5- ((1- (1- (2- ((1- ((tert-butoxycarbonyl) amino) cyclopropyl) methoxy) -5-fluoropyridinyl-3-yl) cyclopropyl) aminoethyl-6- (dimethylphosphoryl) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (75mg, 0.12mmol) was dissolved in HCl/EA (10mL) and stirred at room temperature for 1 hour. After the reaction, the reaction solution was concentrated to dryness. The residue was dissolved in EtOH/H2To O (11mL,10/1), NaOH (20mg, 0.48mmol) was added to the reaction mixture, and the temperature was raised to 60 ℃ to react for 4 hours. After the reaction was complete, the reaction was concentrated to dryness to give a crude product as a white solid. The solid was washed with DCM/EtOH (20mL,5/1), filtered and the filtrate was concentrated to give the title compound (39mg, 68% yield) as a pale yellow solid.
EM (calculated): 474.2; MS (ESI) M/z (M + H) +: 475.2
And step 3: preparation of 5-oxa-2, 8-diaza-16-dimethylphosphoryl-45-fluoro-1 (5,3) -pyrazolo [1,5-a ] pyrimidinehetero-4 (3,2) -pyridiz-3 (1,1) -cyclopropanehetero-7 (1,1) -cyclopropaneheterocyclic nonapan-9-one
Reacting 5- ((1- (2- (((1-aminocyclopropyl) methoxy) -5-fluoropyridin-3-yl) cyclopropyl) amino) -6- (dimethylphosphoryl) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (39mg, 0.08mmol), HATU (61mg, 0.16mmol) and DIEA (21mg, 0.16mmol) were dispersed in THF (10mL) and stirred at room temperature for 4 h. After the reaction, the reaction solution was concentrated, the residue was added to water, extracted twice with DCM, the organic phases were combined andwith anhydrous Na2SO4And (5) drying. The crude product obtained by concentration was purified by TLC (DCM/MeOH ═ 20/1) to give the title compound (8mg, yield 22%) as an off-white solid.
EM (calculated): 456.1, respectively; MS (ESI) M/z (M + H) +: 457.1
1H NMR(400MHz,d6-DMSO)δ0.83-0.87(4H,m),0.93-0.98(1H,m),1.28-1.30(1H,m),1.83(3H,s),1.85(3H,s),2.03-2.07(1H,m),2.10-2.14(1H,m),3.58(1H,d,J=9.0Hz),4.12(1H,d,J=9.0Hz),7.99(1H,dd,J=6.8Hz,2.8Hz),8.01(1H,d,J=11.6Hz),8.05(1H,d,J=6.8Hz),9.15(1H,d,J=6.4Hz),9.27(1H,s),9.36(1H,s).
The following example compounds were synthesized according to the procedure set forth in example 17:
TABLE 3
Figure BDA0003263976850000411
Figure BDA0003263976850000421
Test example 1 inhibitory Effect of Compounds on kinase Activity
1: test materials:
ALK(Carna,No.11CBS-0934L),ALK L1196M(Carna,No.15CBS-0496E),TRKA(Carna,No.13CBS-0565G),ROS1(Carna,No.11CBS-0883C),Kinase substrate22(GL,No.P190116-SL112393),DMSO(Sigma,No.SHBG3288V),384well white plate(PerkinElmer,No.810712),Staurosporine(selleckchem,No.S142105)
2: the experimental method comprises the following steps:
2.1 preparation of Compounds
Compounds were received by the administrator and dissolved in 100% DMSO, formulated into 10mM stock, -20 cryopreserved.
2.2 kinase reaction Processes
(1) A1 XKinase buffer was prepared.
(2) Preparation of compound concentration gradient: test compounds were tested at 1000nM, diluted to 100-fold final concentration in 100% DMSO solutions in 384source plates, and compounds were diluted 3-fold with Precision, 10 concentrations. Using a dispenser Echo 550 to the target plate OptiPlate-384F transfer 250nL 100 times the final concentration of compounds.
(3) A2.5 fold final concentration of Kinase solution was prepared using a 1 XKinase buffer.
(4) Add 10. mu.L of 2.5 fold final concentration kinase solution to the compound well and positive control well, respectively; mu.L of 1 XKinase buffer was added to the negative control wells.
(5) Centrifuge at 1000rpm for 30 seconds, shake the plate and incubate at room temperature for 10 minutes.
(6) A mixture of ATP and Kinase substrate22 was made up at 5/3 fold final concentration using 1 XKinase buffer.
(7) The reaction was initiated by adding 15. mu.L of a mixed solution of ATP and substrate at 5/3-fold final concentration.
(8) The 384 well plates were centrifuged at 1000rpm for 30 seconds, shaken and mixed and incubated at room temperature for the appropriate time.
(9) Add 30. mu.L of termination detection solution to stop the kinase reaction, centrifuge at 1000rpm for 30 seconds, shake and mix.
(10) The conversion was read using a Caliper EZ Reader.
2.3 data analysis
Formula for calculation
Figure BDA0003263976850000431
Wherein: conversion% _ sample is the Conversion reading for the sample; conversion% _ min: negative control well mean, representing conversion readings without enzyme live wells; conversion% _ max: positive control wells are averaged for conversion readings in wells without compound inhibition.
Fitting a dose-response curve:
the log values of the concentrations were taken as the X-axis and the percent inhibition as the Y-axis, and the log (inhibitor) vs. response-Variable slope of the analytical software GraphPad Prism5 was used to fit the dose-effect curves to obtain IC50 values for the enzyme activities of the respective compounds. The calculation formula is:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))
the test results are shown in table 4:
TABLE 4 inhibitory Activity of Compounds on wild-type and mutant ALK, TRK, ROS1 kinase (IC)50)
Figure BDA0003263976850000432
Figure BDA0003263976850000441
Test example 2 inhibitory Effect of Compounds on cell Activity
1: cell lines
Cell lines Cell type Cell number/well Culture medium
Ba/F3EML4-ALK-WT Suspended in water 3000 RPMI-1640+10%FBS
Ba/F3EML4-ALK-L1196M Suspended in water 3000 RPMI-1640+10%FBS
Ba/F3LMNA-NTRK1-G595R Suspended in water 3000 RPMI-1640+10%FBS
Placing at 37 ℃ and 5% CO2And culturing under 95% humidity.
2: reagent and consumable
Fetal bovine serum FBS (GBICO, Cat #10099-141)
CellTiter-
Figure BDA0003263976850000442
Luminescent Cell Viability Assay(Promega,Cat#G7573)
96-hole transparent flat-bottom black wall plate (
Figure BDA0003263976850000443
Cat#165305)
RPMI-1640(Hyclone,Cat#SH30809.01)
3: cell culture and inoculation:
(1) cells in the logarithmic growth phase were harvested and counted using a platelet counter. Detecting the cell viability by using a trypan blue exclusion method to ensure that the cell viability is over 90 percent;
(2) adjusting the cell concentration; add 90 μ Ι _ of cell suspension to 96-well plates, respectively;
(3) cells in 96-well plates were incubated at 37 ℃ with 5% CO2And cultured overnight under 95% humidity conditions.
4: drug dilution and dosing:
(1) preparing 10 times of drug solution, wherein the highest concentration is 10 mu M, the concentration is 9, the dilution is 3.16 times (refer to appendix 1), 10 mu L of drug solution is added into each hole of a 96-hole plate inoculated with cells, three compound holes are arranged for each drug concentration, the final acting concentration of the compound is 1 mu M, the concentration is 9, the dilution is 3.16 times, and the final DMSO concentration is 0.1%;
(2) the cells in the dosed 96-well plate were placed at 37 ℃ in 5% CO2And culturing for 72 hours under the condition of 95% humidity,CTG analysis was then performed.
5: reading the plate at the end:
(1) melt CTG reagents and equilibrate cell plates to room temperature for 30 minutes;
(2) adding equal volume of CTG solution into each well;
(3) the cells were lysed by shaking on an orbital shaker for 5 minutes;
(4) the cell plate was left at room temperature for 20 minutes to stabilize the luminescence signal;
(5) and reading the cold light value.
6: data processing
Data were analyzed using GraphPad Prism 7.0 software, fitted to the data using non-linear S-curve regression to derive a dose-effect curve, and IC50 values were calculated therefrom.
Cell survival rate (%) ═ (Lum)Drug to be tested-LumCulture fluid control)/(LumCell controls-LumCulture fluid control)×100%。
The test results are shown in table 5:
TABLE 5 inhibitory Activity of Compounds IC on Ba/F3ALK, TRK, ROS1 cells50(nm)
Figure BDA0003263976850000451
Test example 3 detection of Compound liver microsome stability test
1: materials and methods
Buffer solution:
(1)100mM potassium phosphate buffer, pH 7.4; (2)10mM MgCl2
Preparation of compound solution:
(1) preparation of 100. mu.M working solution: mu.L of the stock solution (10mM) of the test or control group was diluted with 495. mu.L of methanol to give a compound concentration of 100. mu.M (99% MeOH).
(2) Preparation of 10 μ M working solution: mu.L of 100. mu.M working solution was diluted with 450. mu.L of 100mM potassium phosphate buffer to give a compound concentration of 10. mu.M (9.9% MeOH).
Composition of NADPH (prototype coenzyme II) regeneration System (final concentration of isocitrate dehydrogenase in culture broth 1.0 unit/mL):
β -nicotinamide adenine dinucleotide phosphate, supplier: chem-impex international shipment number: preparation of a solution of N00616 liver microsomes (final concentration of 0.5mg protein/mL), the liver microsomes were of the type shown in Table 6:
TABLE 6
Figure BDA0003263976850000452
Stopping liquid:
acetonitrile ice-cold solution containing 100ng/mL tolbutamide and 100ng/mL labetalol as internal standards.
The method comprises the following operation steps:
(1) in addition to the blank matrix plate wells, 10 μ L of working solution of test or control drug was added to each of the plate wells (T0, T5, T10, T20, T30, T60, and NCF 60).
(2) 80 μ L/well of the microsome solution was dispensed onto each plate using Apricot and the mixture of microsome solution and compound was incubated at 37 ℃ for about 10 minutes.
(3) To NCF60 was added 10. mu.L of 100mM potassium phosphate buffer/well, incubated at 37 ℃ and timer 1 was started for the time shown in Table 7.
TABLE 7
Figure BDA0003263976850000461
(4) After preheating, 10. mu.L/well of NADPH regeneration system was dispensed to each plate using Apricot to start the reaction.
TABLE 8 Final concentrations of each component in the incubation Medium
Figure BDA0003263976850000462
(5) Incubate at 37 ℃ and start timer 2, see table 9 for data.
TABLE 9
Figure BDA0003263976850000463
(6) The reaction was stopped by adding pre-cooled stop solutions (containing 100ng/mL tolbutamide and 100ng/mL albuterol) at 4 ℃ to each well.
(7) The sample plate was then shaken on a shaking trigger for about 10 minutes.
(8) The samples were centrifuged at 4000rpm for 20min at 4 ℃.
(9) And adding water of 300 mu LHPLC grade into each hole of a 96-hole plate, adding supernatant obtained by 100 mu L centrifugation into the corresponding hole position, and uniformly mixing the supernatant and the hole position for LC/MS/MS detection.
And (3) data analysis:
calculating t 1/2 and clint (mic) values according to first order elimination kinetics
The first order elimination kinetics equation is:
Figure BDA0003263976850000471
Figure BDA0003263976850000472
Figure BDA0003263976850000473
Figure BDA0003263976850000474
Figure BDA0003263976850000475
the results of the partial compound liver microsome stability test are shown in table 10:
watch 10
Figure BDA0003263976850000476
Test example 4 detection of Compounds rat in vivo PK assay
SD rats, male (purchased from shanghai siepal-bika laboratory animals ltd). The test compounds were administered to SD rats in a single dose in both oral (10mg/kg, 3 per group) and intravenous (1mg/kg, 3 per group) modes of administration for pharmacokinetic studies. The test compound is prepared on the day of administration, and the test compound is dissolved by using 5% DMSO + 10% solutol + 85% saline, and is prepared into an administration solution after vortex for 2min and ultrasonic treatment for 5 min. Animals were fasted for 10-14 hours prior to oral administration and returned to food 4 hours after administration. After the SD rat is orally taken through the gavage and intravenously administered, pharmacokinetic samples are collected through jugular veins, and the collection time points are as follows: before administration, 5min, 15min, 30min, 1h, 2h, 4h, 6h, 8h and 24h after administration, 3 whole blood samples were collected at each time point, in an amount of about 0.2mL, and anticoagulated with heparin sodium. Immediately after the blood sample was collected, the blood was placed on ice and the plasma was centrifuged within 1 hour (centrifugation conditions: 6800 rpm, 6 minutes, 2-8 ℃). The collected plasma was stored in a-80 ℃ freezer before analysis.
The results of the pharmacokinetic testing of some of the compounds of the invention are shown in table 11 below:
TABLE 11 pharmacokinetic test results for some of the compounds of the invention
Examples T1/2(iv)h Tmax(po)h Cmax(po)ng/ml AUC(po)ng/ml*h Cl(iv)ml/hr/kg F(po)%
Example 1 4.32 4.00 16004 139106 66.07 93.27
Example 6 2.58 4.00 5256 47980 300.20 147.23
Example 10 2.62 4.00 5087 42423 322.41 130.17
Example 11 5.35 4.00 18978 138207 60.74 92.66
Example 12 3.79 4.00 15303 139776 70.11 93.71
TPX-0005 8.17 3.33 1574 8319 1136.20 102.57
From the research data of the patent drug property, the compound has obvious inhibition effect on the activity of the protein kinase inhibitor, has obvious advantages in the aspect of pharmacokinetics compared with clinical second-stage TPX-0005, can be used as the protein kinase inhibitor, and has wide application prospect in resisting malignant tumor diseases or inflammatory diseases.

Claims (28)

1. A compound having the structure of formula (II) or an isomer, tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, pharmaceutically acceptable hydrate, solvate, or salt thereof:
Figure FDA0003263976840000011
wherein M is1Selected from the group consisting of CR15Or N, M2Selected from the group consisting of CR16Or N;
A4is selected from N, A2、A5Are each independently selected from CR17,A1、A3、A6Each independently selected from C, N, and A3、A6Different;
y is selected from CR26R27、O、NR18、S、S(O)、S(O)2、C=O、C=S;
n is selected from 0-5;
R1、R2、R3、R9、R15、R16、R17、R18、R26、R27each independently selected from hydrogen, halogen, substituted or unsubstituted alkyl or cycloalkyl, substituted or unsubstituted heteroalkyl or heterocycloalkyl, hydroxy, cyano, amino, ester, amide, aryl, heteroaryl, sulfonyl, wherein the substituents are each independently selected from halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxy, cyano, amino, ester, amide, aryl, heteroaryl, sulfonyl;
R4selected from the group consisting of hydrogen, halogen, substituted or unsubstituted alkyl or cycloalkyl, substituted or unsubstituted heteroalkyl or heterocycloalkyl, hydroxyl, cyano, amino, ester, amide, aryl, heteroaryl, sulfonyl, boronic acid, phosphoryl, wherein the substituents are each independently selected from the group consisting of halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, amino, ester, amide, aryl, heteroaryl, sulfonyl;
R22、R23are respectively and independently selected from hydrogen, halogen, substituted or unsubstituted alkyl or heteroalkyl with C1-C6, substituted or unsubstituted cycloalkyl or heterocycloalkyl with C3-C6; r5、R6The carbon atoms connected with the substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl, or R7、R8The carbon atoms connected with the substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl, or R22、R23The carbon atoms connected with the substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl, or R5、R8Carbon attached thereto respectivelyThe atoms and the saturated chain segment of the macrocyclic molecule between two carbon atoms jointly form a substituted or non-substituted monocyclic, bicyclic or tricyclic C3-10 cycloalkyl or heterocycloalkyl, or R5、R9The carbon atom and the nitrogen atom which are respectively connected with the above-mentioned two groups form a substituted or unsubstituted C3-10 heterocycloalkyl group, or R23、R9The carbon atoms and the nitrogen atoms which are respectively connected with the substituted or unsubstituted C3-10 heterocycloalkyl group are formed by the saturated chain segments of the macrocyclic molecules between the carbon atoms and the nitrogen atoms; wherein said heterocycloalkyl group contains one or more O, NR25S, S (O), S (O)2, the substituents each being independently selected from the group consisting of halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxy, cyano, amino, ester, amide, aryl, heteroaryl, sulfonyl;
R10、R11、R12、R13the substituent groups are respectively and independently selected from hydrogen, halogen, hydroxyl, cyano, amino, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the substituent groups are respectively and independently selected from halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, amino, ester group, amido, aryl, heteroaryl and sulfonyl;
R25selected from hydrogen, substituted or unsubstituted alkyl or cycloalkyl, substituted or unsubstituted heteroalkyl or heterocycloalkyl, hydroxyl, amido, aryl, heteroaryl, sulfonyl, wherein the substituents are each independently selected from halogen, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, amino, ester, amido, aryl, heteroaryl, sulfonyl.
2. The compound of claim 1, wherein M is1Selected from the group consisting of CR15,M2Selected from the group consisting of CR16Or N; further, R16Selected from H, halogen, C1-3 alkyl or heteroalkyl; further, R16Selected from H, F, chloro, trifluoromethyl, difluoromethyl, methyl, methoxy.
3. The compound of claim 1, wherein a is1、A6Is selected from C.
4. The compound of claim 1, having the structure of formula (IV) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable hydrate, solvate, or salt thereof:
Figure FDA0003263976840000021
wherein:
n is selected from 0, 1 or 2;
R2selected from halogens;
R4、R9、R10、R11、R14the substituent groups are respectively and independently selected from hydrogen, halogen, hydroxyl, cyano, amino, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the substituent groups are respectively and independently selected from halogen, hydroxyl, cyano, amino, ester group, amido and sulfonyl;
R22、R23are respectively and independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl;
R5、R6the carbon atoms connected with the substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl;
the heterocycloalkyl group containing one or more O or NR25The substituents are respectively and independently selected from halogen, hydroxyl, cyano, amino, ester group, amido and sulfonyl;
or R5、R9The carbon atom and the nitrogen atom which are respectively connected with the N-substituted heterocyclic alkyl form a substituted or unsubstituted C3-6N heterocyclic alkyl group, or R23、R9Carbon atoms, nitrogen atoms and saturated chain segments of macrocyclic molecules between carbon atoms and nitrogen atomsThe same composition of substituted or unsubstituted C3-6N heterocycloalkyl, wherein the substituents are independently selected from halogen, C1-6 alkyl or heteroalkyl, hydroxyl, cyano, amino and ester;
R25is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C3-6 cycloalkyl, wherein the substituent is selected from halogen, hydroxyl, cyano and amino.
5. The compound of claim 4, wherein:
R4the compound is selected from H, halogen, hydroxyl, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the substituents are independently selected from halogen, hydroxyl, cyano, amino, ester, amido and sulfonyl;
R22、R23、R9are respectively and independently selected from H, halogen and C1-3 alkyl;
R5、R6the carbon atoms connected with the heterocyclic ring form a substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the heterocycloalkyl contains one or more O or NR25The substituents are respectively and independently selected from F, Cl, Br, hydroxyl, cyano and amino;
or R5、R9The carbon atom and the nitrogen atom which are respectively connected with the N-substituted heterocyclic ring form the N-heterocyclic alkyl of C3-6, or R23、R9The C3-6N heterocycloalkyl is formed by carbon atoms, nitrogen atoms and saturated chain segments of macrocyclic molecules between the carbon atoms and the nitrogen atoms which are respectively connected with the N and the nitrogen atoms;
R25selected from hydrogen, substituted or unsubstituted C1-3 alkyl, wherein the substituents are independently selected from hydroxyl or amino.
6. The compound of claim 4 or 5, wherein: r10、R11、R14Selected from H, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl; further, R10、R11、R14Is selected from H.
7. The compound of claim 5, wherein: r4Selected from H, F, Cl, Br, methyl, trifluoromethyl, difluoromethyl, methoxy, ethyl, propyl, isopropyl, cyclopropyl, hydroxy,
Figure FDA0003263976840000031
8. The compound of claim 4, wherein: n is selected from 0 or 1.
9. A compound according to claim 4 or 5, wherein R is5、R6、R9、R22、R23Wherein, the acyclic ones are all selected from H.
10. A compound of claim 4, wherein R is5、R6The carbon atoms connected with the aryl form a substituted or unsubstituted C3-6 cycloalkyl, or R5、R9The substituted or unsubstituted 4-6-membered N-containing heterocycloalkyl group is formed by the carbon atom and the nitrogen atom which are respectively connected with the substituted or unsubstituted N-containing heterocycloalkyl group, and N is selected from 0; further, R5、R6The carbon atom to which it is attached constitutes a substituted or unsubstituted cyclopropyl, or R5、R9Together with the carbon atom and the nitrogen atom to which they are respectively attached, form a substituted or unsubstituted 5-membered N-containing heterocycloalkyl group.
11. The compound of claim 4,
Figure FDA0003263976840000032
is composed of
Figure FDA0003263976840000033
Further, the
Figure FDA0003263976840000034
Is composed of
Figure FDA0003263976840000035
12. The compound of claim 1, having the structure of formula (V) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable hydrate, solvate, or salt thereof:
Figure FDA0003263976840000041
wherein:
n is selected from 0, 1 or 2;
R2selected from halogens;
R4、R9、R10、R11、R14the substituent groups are respectively and independently selected from hydrogen, halogen, hydroxyl, cyano, amino, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the substituent groups are respectively and independently selected from halogen, hydroxyl, cyano, amino, ester group, amido and sulfonyl;
R23selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl;
R5、R8the carbon atoms connected with the heterocyclic compound and the saturated chain segment of the macrocyclic molecule between two carbon atoms jointly form a substituted or non-substituted C3-6 cycloalkyl or heterocycloalkyl, wherein the heterocycloalkyl contains one or more O or NR25The substituents are respectively and independently selected from halogen, hydroxyl, cyano, amino, ester group, amido and sulfonyl;
R25is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C3-6 cycloalkyl, wherein the substituent is selected from halogen, hydroxyl, cyano and amino.
13. The compound of claim 12, wherein:
R4the compound is selected from H, halogen, hydroxyl, substituted or unsubstituted C1-6 alkyl or heteroalkyl, substituted or unsubstituted C3-6 cycloalkyl or heterocycloalkyl, wherein the substituents are independently selected from halogen, hydroxyl, cyano, amino, ester, amido and sulfonyl;
R9an alkyl or heteroalkyl group selected from H, C1-3;
R23selected from H, halogen and C1-3 alkyl;
R5、R8the carbon atoms connected with the heterocyclic compound and the saturated chain segment of the macrocyclic molecule between two carbon atoms jointly form a substituted or non-substituted C3-6 cycloalkyl or heterocycloalkyl, wherein the heterocycloalkyl contains one or more O or NR25The substituents are respectively and independently selected from F, Cl, Br, hydroxyl, cyano and amino;
R25selected from hydrogen, substituted or unsubstituted C1-3 alkyl, wherein the substituents are independently selected from hydroxyl or amino.
14. The compound of claim 13, wherein: r10、R11、R14Selected from H, halogen, substituted or unsubstituted C1-6 alkyl or heteroalkyl; further, R10、R11、R14Is selected from H.
15. The compound of any one of claims 12-14, wherein: r4Selected from H, F, Cl, Br, methyl, trifluoromethyl, difluoromethyl, methoxy, ethyl, propyl, isopropyl, cyclopropyl, hydroxy,
Figure FDA0003263976840000051
16. The compound according to any one of claims 12 to 14, characterized in thatCharacterized in that: r5、R8Carbon atoms connected with the substituted or unsubstituted C3-6 cycloalkyl group are respectively and independently selected from F, Cl, Br, hydroxyl, cyano and amino; y is O.
17. The compound of claim 16, wherein: r5、R8The carbon atoms to which they are respectively attached and the saturated segment of the macrocyclic molecule between the two carbon atoms together constitute a substituted or unsubstituted cyclobutyl, cyclopentyl, cyclohexyl, preferably cyclopentyl, cyclohexyl.
18. The compound of claim 12, wherein:
Figure FDA0003263976840000052
is composed of
Figure FDA0003263976840000053
Further comprises
Figure FDA0003263976840000054
19. A compound according to any one of claims 12 to 17, wherein: n is selected from 0 or 1.
20. A compound according to any one of claims 1 to 19, wherein: r4Selected from H and halogen.
21. The compound of claim 1, wherein the compound structure is selected from one of the following:
Figure FDA0003263976840000055
Figure FDA0003263976840000061
Figure FDA0003263976840000071
Figure FDA0003263976840000081
22. a pharmaceutical composition, wherein the active ingredient of the pharmaceutical composition is selected from the compounds of any one of claims 1 to 21, or one or more of stereoisomers, solvates, hydrates, pharmaceutically acceptable salts and co-crystals thereof.
23. Use of a compound of any one of claims 1 to 21, or a stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or co-crystal thereof, in the preparation of a protein kinase inhibitor; further, the kinase inhibitor is selected from one or more of an ALK inhibitor, a ROS1 inhibitor, a TRK inhibitor, a MET inhibitor, a JAK inhibitor and a CSF-1R inhibitor.
24. Use of a compound of any one of claims 1 to 21, or a stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or co-crystal thereof, for the manufacture of a medicament for the treatment of pain, cancer, inflammation, autoimmune disease, neurodegenerative disorder or trypanosoma cruzi infection in a mammal.
25. The use according to claim 24, wherein the cancer is selected from: non-small cell lung cancer, gastric cancer, neuroblastoma, lung cancer, ovarian cancer, appendiceal cancer, colorectal cancer, melanoma, head and neck cancer, breast cancer, glioblastoma, medulloblastoma, secretory breast cancer, papillary thyroid cancer, adult myeloid leukemia, pancreatic cancer, prostate cancer, cholangiocarcinoma, salivary gland carcinoma, gastrointestinal stromal tumor, infantile fibrosarcoma.
26. Use of a compound of any one of claims 1 to 21, or a stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or co-crystal thereof, in the manufacture of a medicament for inhibiting cell proliferation.
27. Use of a compound of any one of claims 1 to 21, or a stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or co-crystal thereof, for the manufacture of a medicament for the treatment of a disease that causes overexpression of ALK, ROS1, TRK, MET, JAK, CSF-1R kinase.
28. Use of a compound of any one of claims 1 to 21, or a stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or co-crystal thereof, for the manufacture of a medicament for the treatment of a disease caused by overexpression of ALK, ROS1, TRK, MET, JAK, CSF-1R kinase.
CN202111080921.XA 2019-06-21 2020-06-10 Compound as protein kinase inhibitor and preparation method and application thereof Pending CN113773335A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN201910543645 2019-06-21
CN2019105436452 2019-06-21
CN202010525613.2A CN112110938B (en) 2019-06-21 2020-06-10 Compound as protein kinase inhibitor and preparation method and application thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN202010525613.2A Division CN112110938B (en) 2019-06-21 2020-06-10 Compound as protein kinase inhibitor and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN113773335A true CN113773335A (en) 2021-12-10

Family

ID=73799467

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202010525613.2A Active CN112110938B (en) 2019-06-21 2020-06-10 Compound as protein kinase inhibitor and preparation method and application thereof
CN202111080921.XA Pending CN113773335A (en) 2019-06-21 2020-06-10 Compound as protein kinase inhibitor and preparation method and application thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202010525613.2A Active CN112110938B (en) 2019-06-21 2020-06-10 Compound as protein kinase inhibitor and preparation method and application thereof

Country Status (1)

Country Link
CN (2) CN112110938B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117412970A (en) * 2021-04-12 2024-01-16 成都倍特药业股份有限公司 Solid form of fluorine-containing macrocyclic structure compound, preparation method and application
CN113582994B (en) * 2021-09-28 2022-02-11 北京鑫开元医药科技有限公司 Compound with TRK kinase inhibitory activity, preparation method, composition and application thereof
CN117427079A (en) * 2022-07-20 2024-01-23 广州嘉越医药科技有限公司 Use of cyano-substituted macrocyclic compounds

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109516999A (en) * 2017-11-01 2019-03-26 郑州泰基鸿诺医药股份有限公司 Compound and its application as protein kinase regulator
CN111971287A (en) * 2018-03-28 2020-11-20 重庆复创医药研究有限公司 Macrocyclic compounds as TRK kinase inhibitors

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL3205654T3 (en) * 2010-05-20 2019-08-30 Array Biopharma, Inc. Macrocyclic compounds as trk kinase inhibitors
EA201892241A1 (en) * 2014-01-24 2019-02-28 ТиПи ТЕРАПЬЮТИКС, ИНК. DIARYLIC MACRO CYCLES AS PROTEINKINASE MODULATORS
US9273067B2 (en) * 2014-02-19 2016-03-01 Bristol-Myers Squibb Company Pyrazolopyrimidine macrocycles as inhibitors of human immunodeficiency virus replication
JP2019527230A (en) * 2016-07-28 2019-09-26 ターニング・ポイント・セラピューティクス・インコーポレイテッドTurning Point Therapeutics,Inc. Macrocyclic kinase inhibitor
BR112020003783A2 (en) * 2017-08-23 2020-09-01 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. macrocycle containing aminopyrazole and pyrimidine and pharmaceutical composition and use thereof
CN110156813B (en) * 2018-02-13 2023-07-25 北京诺诚健华医药科技有限公司 Heterocyclic compounds as TRK inhibitors
US11358973B2 (en) * 2018-04-16 2022-06-14 Shenzhen Targetrx, Inc. Di(hetero)aryl macrocyclic compound for inhibiting protein kinase activity

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109516999A (en) * 2017-11-01 2019-03-26 郑州泰基鸿诺医药股份有限公司 Compound and its application as protein kinase regulator
CN111971287A (en) * 2018-03-28 2020-11-20 重庆复创医药研究有限公司 Macrocyclic compounds as TRK kinase inhibitors

Also Published As

Publication number Publication date
CN112110938B (en) 2021-11-09
CN112110938A (en) 2020-12-22

Similar Documents

Publication Publication Date Title
AU2016225896B2 (en) Heterocyclic compounds useful as PDK1 inhibitors
US9567342B2 (en) Certain protein kinase inhibitors
EP3746424B1 (en) Erbb/btk inhibitors
CN112110938B (en) Compound as protein kinase inhibitor and preparation method and application thereof
CN113307811B (en) Tetrahydropyranyl amino-pyrrolopyrimidinones and methods of use thereof
JP5490790B2 (en) Substituted pyrimidin-4-one derivatives
KR20090102756A (en) Substituted heteroaryl pyridopyrimidone derivatives
JP6986032B2 (en) Crystals of pyrrolopyrimidine compounds as JAK inhibitors
CN112608318A (en) Compound serving as protein kinase inhibitor and application thereof
JP5508256B2 (en) Heteroarylamide substituted pyrimidone derivatives for the treatment of neurodegenerative diseases
CN111763215B (en) Compound with nitrogen-containing heterocyclic structure and preparation method and application thereof
TW202229297A (en) Modulators of cystic fibrosis transmembrane conductance regulator
KR20160137566A (en) SUBSTITUTED 4,5,6,7-TETRAHYDRO-PYRAZOLO[1,5-a]PYRIMIDINE DERIVATIVES AND 2,3-DIHYDRO-1H-IMIDAZO[1,2-b]PYRAZOLE DERIVATIVES AS ROS1 INHIBITORS
CN109790156B (en) Chiral heterocyclic compound with hedgehog pathway antagonist activity and preparation method and application thereof
EP3967691A1 (en) Pyrimido five-membered heterocyclic compound and use thereof as mutant idh2 inhibitor
CN111170986A (en) Inhibitors of bruton's tyrosine kinase
EP3105232B1 (en) Substituted triazolobenzodiazepines
EP3988551A1 (en) Cdk kinase inhibitor
CN112574255A (en) Organic arsine-based CDK inhibitor and preparation method and 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