CN116789670A - Selective CSF1R inhibitor and application thereof - Google Patents

Selective CSF1R inhibitor and application thereof Download PDF

Info

Publication number
CN116789670A
CN116789670A CN202210262929.6A CN202210262929A CN116789670A CN 116789670 A CN116789670 A CN 116789670A CN 202210262929 A CN202210262929 A CN 202210262929A CN 116789670 A CN116789670 A CN 116789670A
Authority
CN
China
Prior art keywords
phenyl
pyrrolo
reaction
pyrimidin
trifluoromethyl
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
CN202210262929.6A
Other languages
Chinese (zh)
Inventor
刘青松
刘静
梁小飞
王纯
王蓓蕾
王傲莉
亓爽
齐紫平
刘青旺
刘娟
操江艳
王文超
王黎
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.)
Hefei Institutes of Physical Science of CAS
Original Assignee
Hefei Institutes of Physical Science of CAS
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 Hefei Institutes of Physical Science of CAS filed Critical Hefei Institutes of Physical Science of CAS
Priority to CN202210262929.6A priority Critical patent/CN116789670A/en
Priority to PCT/CN2022/083651 priority patent/WO2023173480A1/en
Publication of CN116789670A publication Critical patent/CN116789670A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Landscapes

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

Abstract

The present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof and its use in inhibiting CSF1R kinase and related signaling pathway activity, or treating M2-type macrophage-based cancers or tumors in a tumor microenvironment, wherein L, ar and R 1 As defined in the specification.

Description

Selective CSF1R inhibitor and application thereof
Technical Field
The invention belongs to the field of biological medicine, and in particular relates to a selective CSF1R inhibitor and application thereof in anticancer drugs.
Background
Members of the class III receptor tyrosine kinase (RTK class III) family include colony stimulating factor 1 (CSF 1R), platelet-derived growth factor (PDGFR alpha/beta), KIT, FLT3, etc., which are very similar in gene sequence and have a protein structure comprising an extracellular region of 5 Ig-like domains, a transmembrane region, a membrane-proximal region, and two tyrosine kinase regions separated by a kinase insert region (Abu-Duhier FM, et al Mutational analysis of class III receptor tyrosine kinases (C-KIT, C-FMS, FLT 3) in idiopathic myeleofibrosis.Br.Haemal.2003, 120 (3): 464-470). Wherein the KIT protein is used as a receptor of stem cell factors and participates in the proliferation and differentiation process of hematopoietic stem cells through a series of signal paths. FLT3 protein is also known as FMS-like tyrosine kinase 3, which regulates the proliferation and differentiation of hematopoietic cells.
CSF1 is a dimeric glycoprotein linked by disulfide bonds, which is mainly present in the bone marrow cavity and is responsible for cellular processes such as growth, proliferation and differentiation of macrophages. CSF1R plays an important role as a cell surface receptor for CSF1 and IL34 in regulating survival, proliferation and differentiation of hematopoietic precursors. The signal axis formed by CSF1 and CSF1R is abnormally expressed in various malignant tumors such as breast cancer, ovarian cancer, nasopharyngeal carcinoma and the like, and is closely related to the occurrence and development of tumors. In the existing kinase inhibitors, the simultaneous inhibition of FLT3 and KIT can generate myelosuppression toxicity, so that the selective inhibition of CSF1R without inhibiting FLT3 and KIT can reduce the toxic and side effects of the medicament. While FLT3, KIT have high homology to the ATP binding pocket of CSF1R kinase, there is a constant need in the art to obtain selective CSF1R inhibitors because of the difficulty in achieving selectivity.
Disclosure of Invention
It is an object of the present invention to provide a class of compounds which selectively inhibit CSF1R kinase and related signaling pathway activity and thus are capable of activating autoimmunity by inhibiting phosphorylation of CSF1R for use in the treatment of cancer.
Accordingly, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof:
Wherein, the liquid crystal display device comprises a liquid crystal display device,
wherein L is selected from
Ar is selected from optionally 1 to 3 independently R 2 Aryl and heteroaryl groups substituted with groups;
R 1 selected from the group consisting of C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 heterocycloalkyl, hydroxyC 1-C6 alkyl, nitriloC 1-C6 alkyl, carboxyC 1-C6 alkyl, C3-C6 cycloalkylC 1-C6 alkyl, C1-C6 heterocycloalkylC 1-C6 alkyl, C1-C6 alkoxyC 1-C6 alkyl, C1-C6 alkylaminoC 1-C6 alkyl, C1-C6 alkylsulphonyl, C1-C6 alkanoyl, C1-C6 heterocycloalkylaminoacyl, hydroxyC 1-C6 alkylamineacyl, C3-C6 cycloakylAlkylamineacyl, C1-C6 alkoxycarbonyl, aryl, aryloxycarbonyl, and heteroaryl, wherein aryl, heteroaryl, and heterocycloalkyl are optionally substituted with 1R 2 Group substitution;
each R is 2 Independently selected from halogen, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, halogenated C1-C6 alkoxy, and nitrogen protecting groups.
In a preferred embodiment, L is
In a further preferred embodiment Ar is selected from the group consisting of optionally 1 to 2 independently R 2 Phenyl, pyridyl, furyl, benzofuryl, thienyl, and pyrrolyl groups substituted with groups; further preferably, each R 2 Independently selected from halogen, C1-C3 alkyl, C1-C3 alkoxy, C1-C3 haloalkyl, halogenated C1-C3 alkoxy, pivaloyl, t-butoxycarbonyl, benzyloxycarbonyl, 9-fluorenylmethoxycarbonyl, benzyl, p-methoxybenzyl, allyloxycarbonyl, and trifluoroacetyl.
In a particularly preferred embodiment Ar is selected from the group consisting of 1R 2 Phenyl or pyridyl substituted with a group; more preferably Ar is selected from para-quilt R 2 Phenyl or pyridin-2-yl substituted with a group; further preferably, R 2 Selected from C1-C6 haloalkyl, more preferably trifluoromethane.
In other preferred embodiments, R 1 Selected from the group consisting of C1-C3 alkyl, C1-C3 haloalkyl, C3-C5 heterocycloalkyl, hydroxyC 1-C3 alkyl, nitriloC 1-C3 alkyl, carboxyC 1-C3 alkyl, C3-C5 cycloalkylC 1-C3 alkyl, C1-C3 alkoxyC 1-C3 alkyl, C1-C4 alkylaminoC 1-C3 alkyl, C1-C3 alkylsulfonyl, C2-C4 alkanoyl, C3-C5 heterocycloalkylaminoacyl, hydroxyC 1-C3 alkylamineacyl, C1-C4 alkylaminoacyl, C3-C6 cycloalkylaminoacyl, C1-C3 alkoxycarbonyl, phenyl, phenoxycarbonyl, thienyl and pyridyl, wherein phenyl and heterocycloalkyl are optionally substituted with methyl, halogen or t-butoxycarbonyl; more preferably, R 1 Selected from methyl, acetyl, propionyl, butyryl, and methylaminoacyl.
In another aspect, the application also provides pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite, or prodrug thereof, and a pharmaceutically acceptable diluent or carrier, and optionally other active pharmaceutical compounds.
In other aspects, the application also relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof, in the manufacture of a medicament for inhibiting CSF1R kinase and related signaling pathway activity, or for treating cancer or tumour associated with CSF1R kinase activity.
In a preferred aspect, the application also relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof for the manufacture of a medicament for the treatment of M2-type macrophage-based cancer or tumor in a tumor microenvironment (tumor micro environment, TME).
The cancer or tumor is selected from one or more of ovarian cancer, cervical cancer, membranous adenocarcinoma, prostate cancer, bladder cancer, lung cancer, thyroid cancer, breast cancer, pancreatic cancer, renal cancer, gastric cancer, liver cancer, cervical cancer, endometrial cancer, colorectal cancer, nasopharyngeal cancer, esophageal cancer, cholangiocarcinoma, bone metastatic cancer, papillary thyroid cancer, non-small cell lung cancer, colon cancer, solid tumor, brain tumor, lymphoma, glioma, glioblastoma, melanoma, mesothelioma, glioblastoma, osteosarcoma, gastrointestinal stromal tumor, multiple myeloma, biliary tract carcinoma sarcoma, and leukemia. Wherein the lymphoma comprises non-hodgkin's lymphoma, B-cell or T-cell lymphoma; the leukemia includes chronic myelogenous leukemia or acute myelogenous leukemia.
The invention also relates to a method of inhibiting CSF1R kinase and associated signaling pathway activity, or treating M2-type macrophage-based cancer or tumor in a tumor microenvironment, comprising administering to a patient a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite, or prodrug thereof, or a pharmaceutical composition comprising the same.
Drawings
FIG. 1 shows the promotion of macrophage transformation from M2 to M1 by compound 54 and BLZ945, PLC 3397.
FIG. 2 shows the effect of compound 54 and PLX3397 on mouse body weight after administration in an M-NFS-60 cell tumor engrafting mouse model.
FIG. 3 shows the effect of compound 54 and PLX3397 on tumor volume following administration in a M-NFS-60 cell tumor engrafting mouse model.
FIG. 4 shows the effect of compound 54 and PLX3397 on tumor weight following administration in an M-NFS-60 cell tumor engrafting mouse model.
Figure 5 shows the effect of compound 54 and PLX3397 on mouse body weight following administration in a MC38 cell tumor engrafting mouse model.
Figure 6 shows the effect of compound 54 and PLX3397 on tumor volume following administration in a MC38 cell tumor engrafting mouse model.
Figure 7 shows the effect of compound 54 and PLX3397 on tumor weight following administration in a MC38 cell tumor engrafting mouse model.
Detailed Description
Terminology
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which claimed subject matter belongs.
The present invention employs, unless otherwise indicated, conventional methods of mass spectrometry, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology within the skill of the art. Unless specifically defined otherwise, nomenclature and laboratory procedures and techniques related to analytical chemistry, synthetic organic chemistry, and chemistry such as medical and pharmaceutical chemistry described herein are known to those skilled in the art. In general, the foregoing techniques and steps may be implemented by conventional methods well known in the art and described in various general and more specific documents, which are cited and discussed in this specification.
The term "alkyl" refers to an aliphatic hydrocarbon group, which may be branched or straight chain. Depending on the structure, the alkyl group may be a monovalent group or a divalent group (i.e., alkylene). In the present invention, the alkyl group is preferably an alkyl group having 1 to 8 carbon atoms, more preferably a "lower alkyl group" having 1 to 6 carbon atoms, even more preferably an alkyl group having 1 to 4 carbon atoms. Typical alkyl groups include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, and the like. It is to be understood that references herein to "alkyl" include such alkyl groups in all configurations and conformations that may be present, e.g., references herein to "propyl" include n-propyl and isopropyl, "butyl" includes n-butyl, isobutyl and tert-butyl, and references to "pentyl" include n-pentyl, isopentyl, neopentyl, tert-pentyl, and pent-3-yl, and the like.
The term "alkoxy" refers to an-O-alkyl group, wherein alkyl is as defined herein. Typical alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, pentoxy, hexoxy, and the like.
The term "cycloalkyl" refers to a monocyclic or polycyclic group containing only carbon and hydrogen. Cycloalkyl includes groups having 3 to 12 ring atoms. Cycloalkyl groups may be monovalent or divalent (e.g., cycloalkylene) depending on the structure. In the present invention, the cycloalkyl group is preferably a cycloalkyl group having 3 to 8 carbon atoms, more preferably a "lower cycloalkyl group" having 3 to 6 carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, and adamantyl.
The term "aryl" refers to a planar ring having a delocalized pi-electron system and containing 4n+2 pi electrons, where n is an integer. The aryl ring may be composed of five, six, seven, eight, nine or more than nine atoms. The aryl group may be optionally substituted. The term "aryl" includes carbocyclic aryl (e.g., phenyl) and heterocyclic aryl (or "heteroaryl") groups (e.g., pyridine). The term includes monocyclic or fused ring polycyclic (i.e., rings that share adjacent pairs of carbon atoms) groups.
The term "aryl" as used herein means that each of the atoms making up the ring in the aromatic ring is a carbon atom. The aryl ring may be composed of five, six, seven, eight, nine or more than nine atoms. Aryl groups may be optionally substituted. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, phenanthryl, anthracyl, fluorenyl, and indenyl. Depending on the structure, the aryl group may be a monovalent group or a divalent group (i.e., arylene).
The term "aryloxy" refers to an-O-aryl group, wherein aryl is as defined herein.
The term "heteroaryl" refers to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen, and sulfur. An "heteroaryl" moiety containing N means that at least one backbone atom in the ring of the aromatic group is a nitrogen atom. Depending on the structure, the heteroaryl group may be a monovalent group or a divalent group (i.e., heteroarylene). Examples of heteroaryl groups include, but are not limited to, pyridyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothienyl, benzothiazolyl, benzoxazolyl, quinazolinyl, naphthyridinyl, furopyridinyl, and the like.
The term "heteroalkyl" as used herein means that one or more of the backbone chain atoms in the alkyl groups defined herein are heteroatoms such as oxygen, nitrogen, sulfur, silicon, phosphorus, or combinations thereof. The heteroatom(s) may be located at any position within the heteroalkyl group or where the heteroalkyl group is attached to the remainder of the molecule.
The term "heterocycloalkyl" or "heterocyclyl" as used herein means that one or more of the atoms making up the ring in the non-aromatic ring is a heteroatom selected from nitrogen, oxygen and sulfur. The heterocycloalkyl ring may be made up of three, four, five, six, seven, eight, nine or more than nine atoms. The heterocycloalkyl ring may be optionally substituted. Examples of heterocycloalkyl groups include, but are not limited to, lactams, lactones, cyclic imines, cyclic thioimines, cyclic carbamates, tetrahydrothiopyrans, 4H-pyrans, tetrahydropyrans, piperidines, 1, 3-dioxanes, 1, 4-dioxanes, piperazines, 1, 3-oxathiolanes, 1, 4-oxathiolanes, tetrahydro-1, 4-thiazines, 2H-1, 2-oxazines, maleimides, succinimides, barbituric acid, thiobarbituric acid, dioxopiperazines, hydantoins, dihydropyrimidines, morpholines, trioxane, hexahydro-1, 3, 5-triazines, tetrahydrothiophenes, tetrahydrofurans, pyrrolines, pyrrolidines, imidazolidines, pyrazolines, pyrazolidines, imidazolines, imidazoles, 1, 3-dioxoles, 1, 3-dioxanes, 1, 3-dithianes, 1, 3-dithiazolines, 1, 3-oxazanes, 3-oxazalidines, isoxazolines, oxazalidines, and thiazoles. Depending on the structure, the heterocycloalkyl group may be a monovalent group or a divalent group (i.e., heterocycloalkylene).
The term "halogen" or "halo" refers to fluorine, chlorine, bromine and iodine.
The terms "haloalkyl", "haloalkoxy" and "haloalkylalkyl" include structures of alkyl, alkoxy or heteroalkyl groups in which at least one hydrogen is replaced with a halogen atom. In certain embodiments, if two or more hydrogen atoms are replaced with halogen atoms, the halogen atoms are the same or different from each other.
The term "amino" refers to-NH 2 A group.
The term "hydroxy" refers to an-OH group.
The term "cyano" refers to a-CN group.
The term "ester group" refers to a chemical moiety having the formula-COOR, wherein R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (attached through a ring carbon) and heterocyclyl (attached through a ring carbon).
The term "amide" or "amido" refers to-NR-CO-R ', wherein R and R' are each independently hydrogen or alkyl.
The term "aminoacyl" or "aminoacyl" refers to-CO-NH 2 A group.
The term "alkylaminoacyl" or "alkylaminoacyl" refers to a-CO-NH-R group, wherein R is an alkyl group as defined herein.
The term "optional" means that one or more of the subsequently described events may or may not occur, and that both events occurring and events that do not occur are included. The term "optionally substituted" or "substituted" means that the mentioned groups may be substituted with one or more additional groups each and independently selected from alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl, hydroxy, alkoxy, cyano, halogen, amide, nitro, haloalkyl, amino, methanesulfonyl, alkylcarbonyl, alkoxycarbonyl, heteroarylalkyl, heterocycloalkyl, aminoacyl, amino protecting groups, and the like. Among them, the amino protecting group is preferably selected from pivaloyl, t-butoxycarbonyl, benzyloxycarbonyl, 9-fluorenylmethoxycarbonyl, benzyl, p-methoxybenzyl, allyloxycarbonyl, trifluoroacetyl and the like.
The term "tyrosine protein kinase (tyrosine protein kinase, TPK)" as used herein is a class of kinases that catalyze the transfer of gamma-phosphate on ATP to protein tyrosine residues, and which catalyze the phosphorylation of various substrate protein tyrosine residues, playing an important role in cell growth, proliferation, and differentiation.
The terms "inhibit", "inhibited" or "inhibitor" of a kinase as used herein refer to an inhibition of phosphotransferase activity.
A "metabolite" of a compound disclosed herein is a derivative of a compound that is formed when the compound is metabolized. The term "active metabolite" refers to a biologically active derivative of a compound that is formed when the compound is metabolized. The term "metabolized" as used herein refers to the sum of the processes by which a particular substance is altered by an organism (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes, such as oxidation reactions). Thus, enzymes can produce specific structural transformations into compounds. For example, cytochrome P450 catalyzes a variety of oxidation and reduction reactions, while the enzyme phosphoglucomutase catalyzes the conversion of activated glucuronic acid molecules to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulfhydryl groups. Further information on metabolism can be obtained from The Pharmacological Basis of Therapeutics, ninth edition, mcGraw-Hill (1996). Metabolites of the compounds disclosed herein can be identified by administering the compounds to a host and analyzing tissue samples from the host, or by incubating the compounds with hepatocytes in vitro and analyzing the resulting compounds. Both of these methods are known in the art. In some embodiments, the metabolites of the compounds are formed by an oxidation process and correspond to the corresponding hydroxyl containing compounds. In some embodiments, the compound is metabolized to a pharmaceutically active metabolite. The term "modulate" as used herein refers to directly or indirectly interacting with a target to alter the activity of the target, including, by way of example only, enhancing the activity of the target, inhibiting the activity of the target, limiting the activity of the target, or prolonging the activity of the target.
IC50 as used herein refers to the amount, concentration or dose of a particular test compound that achieves 50% inhibition of the maximum effect in an assay that measures such effect.
EC50 as used herein refers to the dose, concentration or amount of an assay compound that causes a dose-dependent response that induces, stimulates or enhances 50% of the maximum expression of a particular response by a particular assay compound.
GI50, as used herein, refers to the concentration of drug required to inhibit 50% of cell growth, i.e., the concentration of drug at which 50% of cell (e.g., cancer cell) growth is inhibited or controlled.
Novel kinase inhibitors of the invention
The present invention provides a compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof:
wherein, the liquid crystal display device comprises a liquid crystal display device,
wherein L is selected from
Ar is selected from optionally 1 to 3 independently R 2 Aryl and heteroaryl groups substituted with groups;
R 1 Selected from the group consisting of C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 heterocycloalkyl, hydroxyC 1-C6 alkyl, nitriloC 1-C6 alkyl, carboxyC 1-C6 alkyl, C3-C6 cycloalkylC 1-C6 alkyl, C1-C6 heterocycloalkylC 1-C6 alkyl, C1-C6 alkoxyC 1-C6 alkyl, C1-C6 alkylaminoC 1-C6 alkyl, C1-C6 alkylsulfonyl, C1-C6 alkanoyl, C1-C6 heterocycloalkylaminoacyl, C1-C6 alkylaminoacyl, C3-C6 cycloalkylaminoacyl, C1-C6 alkoxycarbonyl, aryl, aryloxycarbonyl, and heteroaryl, wherein aryl, heteroaryl and heterocycloalkylare optionally substituted with 1R 2 Group substitution;
each R is 2 Independently selected from halogen, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, halogenated C1-C6 alkoxy, and nitrogen protecting groups.
In a preferred embodiment, L is
In a further preferred embodiment Ar is selected from the group consisting of optionally 1 to 2 independently R 2 Phenyl, pyridyl, furyl, benzofuryl, thienyl, and pyrrolyl groups substituted with groups; further preferably, each R 2 Independently selected from halogen, C1-C3 alkyl, C1-C3 alkoxy, C1-C3 haloalkyl, halogenated C1-C3 alkoxy, pivaloyl, t-butoxycarbonyl, benzyloxycarbonyl, 9-fluorenylmethoxycarbonyl, benzyl, p-methoxybenzyl, allyloxycarbonyl, and trifluoroacetyl.
In a particularly preferred embodiment Ar is selected from the group consisting of 1R 2 Phenyl or pyridyl substituted with a group; more preferably Ar is selected from para-quilt R 2 Phenyl or pyridin-2-yl substituted with a group; further preferably, R 2 Selected from C1-C6 haloalkyl, more preferably trifluoromethane.
In other preferred embodiments, R 1 Selected from the group consisting of C1-C3 alkyl, C1-C3 haloalkyl, C3-C5 heterocycloalkyl, hydroxyC 1-C3 alkyl, nitriloC 1-C3 alkyl, carboxyC 1-C3 alkyl, C3-C5 cycloalkylC 1-C3 alkyl, C3-C5 heterocycloalkyl C1-C3 alkyl, C1-C3 alkoxyC 1-C3 alkyl, C1-C4 alkylamino C1-C3 alkyl, C1-C3 alkanoylamino C1-C3 alkyl, C1-C3 alkylsulfonyl, C2-C4 alkanoyl, C3-C5 heterocycloalkylaminoacyl, C3-C5 heterocycloalkyl C1-C3 alkylaminoacyl, hydroxyC 1-C3 alkylamineacyl, C1-C4 alkylamineacyl, C3-C6 cycloalkylaminoacyl, C1-C3 alkoxycarbonyl, phenyl, phenoxycarbonyl, thienyl and pyridyl, wherein phenyl and heterocycloalkyl are optionally substituted with methyl, halogen or t-butoxycarbonyl; more preferably, R 1 Selected from methyl, acetyl, propionyl, butyryl, and methylaminoacyl.
In a preferred embodiment, the present invention relates to a compound of table 1 below or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof.
TABLE 1
/>
/>
Any combination of the above groups for each variable is also contemplated herein. It will be appreciated that: substituents and substitution patterns on the compounds provided herein can be selected by one of skill in the art to provide compounds that are chemically stable and that can be synthesized using techniques known in the art and set forth herein.
Pharmaceutically acceptable salts, solvates, esters, acids, metabolites, or prodrugs of such compounds are also described herein.
In additional or further embodiments, the compounds described herein are metabolized in vivo to produce metabolites in organisms in need thereof, which are then used to produce desired effects, including desired therapeutic effects.
The compounds described herein may be formulated and/or used as pharmaceutically acceptable salts. Types of pharmaceutically acceptable salts include, but are not limited to: (1) Acid addition salts formed by reacting the free base form of the compound with a pharmaceutically acceptable inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, metaphosphoric acid, and the like; or with an organic acid such as acetic acid, propionic acid, caproic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, malic acid, citric acid, succinic acid, maleic acid, tartaric acid, fumaric acid, trifluoroacetic acid, benzoic acid, 3- (4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1, 2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, 4-methylbicyclo- [2.2.2] oct-2-ene-1-carboxylic acid, 2-naphthalenesulfonic acid, t-butylacetic acid, glucoheptonic acid, 4' -methylenebis- (3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, dodecylsulfuric acid, gluconic acid, glutamic acid, salicylic acid, hydroxynaphthoic acid, stearic acid, muconic acid, and the like; (2) A base addition salt, which is formed when an acidic proton in the parent compound is replaced with a metal ion, such as an alkali metal ion (e.g., lithium, sodium, potassium), alkaline earth metal ion (e.g., magnesium or calcium), or aluminum ion; or with organic or inorganic bases, acceptable organic bases including ethanolamine, diethanolamine, triethanolamine, trimethylamine, N-methylglucamine, and the like; acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.
The corresponding counterions of the pharmaceutically acceptable salts can be analyzed and identified using a variety of methods including, but not limited to, ion exchange chromatography, ion chromatography, capillary electrophoresis, inductively coupled plasma, atomic absorption spectroscopy, mass spectrometry, or any combination thereof.
Recovering the salt using at least one of the following techniques: filtration, precipitation with a non-solvent followed by filtration, solvent evaporation, or lyophilization in the case of aqueous solutions.
Screening and characterization of pharmaceutically acceptable salts, polymorphs, and/or solvates may be accomplished using a variety of techniques including, but not limited to, thermal analysis, X-ray diffraction, spectroscopy, microscopy, elemental analysis. Various spectroscopic techniques are used including, but not limited to Raman, FTIR, UVIS and NMR (liquid and solid states). Various microscopy techniques include, but are not limited to, IR microscopy and Raman (Raman) microscopy.
The pharmaceutical use of the invention
The compound of the formula (I) or the pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof can convert macrophages in TME from M2 type to M1 type which promote tumor growth, thereby changing TME and achieving the aim of treating cancers. In addition, the compounds of formula (I) or pharmaceutically acceptable salts, solvates, esters, acids, metabolites or prodrugs thereof of the present invention inhibit CSF1R kinase and related signaling pathway activity, thereby achieving the goal of treating cancer.
Thus, the use of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof of the application in the manufacture of a medicament for inhibiting CSF1R kinase and associated signal pathway activity, or for treating cancer or tumour associated with CSF1R kinase activity.
In addition, the compounds of formula (I) of the present application, or pharmaceutically acceptable salts, solvates, esters, acids, metabolites or prodrugs thereof, can be used for the treatment of cancers or tumors that are predominantly M2 type macrophages in the tumor microenvironment.
The cancer or tumor is selected from one or more of ovarian cancer, cervical cancer, membranous adenocarcinoma, prostate cancer, bladder cancer, lung cancer, thyroid cancer, breast cancer, pancreatic cancer, renal cancer, gastric cancer, liver cancer, cervical cancer, endometrial cancer, colorectal cancer, nasopharyngeal cancer, esophageal cancer, cholangiocarcinoma, bone metastatic cancer, papillary thyroid cancer, non-small cell lung cancer, colon cancer, solid tumor, brain tumor, lymphoma, glioma, glioblastoma, melanoma, mesothelioma, glioblastoma, osteosarcoma, gastrointestinal stromal tumor, multiple myeloma, biliary tract carcinoma sarcoma, and leukemia. Wherein the lymphoma comprises non-hodgkin's lymphoma, B-cell or T-cell lymphoma; the leukemia includes chronic myelogenous leukemia or acute myelogenous leukemia.
In embodiments of the present invention, a medicament comprising a compound of the present invention may be administered to a patient by at least one of injection, oral, inhalation, rectal and transdermal administration. The amount of a given drug in treating a patient according to the present invention will depend on a number of factors, such as the particular dosage regimen, the type of disease or disorder and its severity, the uniqueness of the subject or host in need of treatment (e.g., body weight), but depending on the particular circumstances, including, for example, the particular drug employed, the route of administration, the disorder being treated, and the subject or host being treated, the dosage administered can be routinely determined by methods known in the art. Generally, for dosages used in adult treatment, the dosage administered is typically in the range of 0.02-5000 mg/day, for example about 1-1500 mg/day. The desired dosage may conveniently be presented as a single dose, or as divided doses administered simultaneously (or in short time periods) or at appropriate intervals, for example two, three, four or more divided doses per day. It will be appreciated by those skilled in the art that, although the above dosage ranges are given, the specific effective amount may be suitably adjusted depending on the patient's condition in combination with a physician's diagnosis.
Preparation of the Compounds
The compounds of formula (I) may be synthesized using standard synthetic techniques known to those skilled in the art or using methods known in the art in combination with the methods described herein. In addition, the solvents, temperatures, and other reaction conditions set forth herein may vary according to the skill in the art. As a further guidance, the following synthetic methods may also be used.
The reactions may be used sequentially to provide the compounds described herein; or they may be used to synthesize fragments that are subsequently added by methods described herein and/or known in the art.
In certain embodiments, provided herein are methods of making the tyrosine kinase inhibitor compounds described herein and methods of use thereof. In certain embodiments, the compounds described herein can be synthesized using the following synthetic schemes. The compounds can be synthesized using a method similar to that described below, by using appropriate alternative starting materials.
The starting materials for the synthesis of the compounds described herein may be synthesized or may be obtained from commercial sources. The compounds described herein and other related compounds having different substituents can be synthesized using techniques and starting materials known to those skilled in the art. The general methods of preparing the compounds disclosed herein may be from reactions known in the art, and the reactions may be modified by reagents and conditions deemed appropriate by one of skill in the art to incorporate various moieties in the molecules provided herein.
If desired, the reaction product may be isolated and purified using conventional techniques including, but not limited to, filtration, distillation, crystallization, chromatography, and the like. These products can be characterized using conventional methods, including physical constants and profile data.
Example 1N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (3- (trifluoromethoxy) phenyl) acetamide
Into a 25mL round bottom flask was charged 20mL of N, N-dimethylformamide, and tert-butyl (4-hydroxyphenyl) carbamate (1.4 g), 4-chloro was added with stirring at room temperature-7-methylpyrrolo [2,3-D]Pyrimidine (1 g) and cesium carbonate (3.9 g). The reaction system was heated to 50℃and then reacted for 14 hours. After the completion of the reaction, the reaction was neutralized with 50mL of water. The mixed solution was extracted three times with 50mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 30mL of water, 30mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase was distilled under reduced pressure to give a crude product, which was chromatographed on a silica gel column to give a yellow product (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) carbamic acid tert-butyl ester. MS m/z (ESI): 341.16[ M+H ]] +
5mL of methylene chloride was added to a 25mL round bottom flask and the mixture was stirred at room temperature (4- ((7-methyl-7H-pyrrolo [2, 3-D) ]Pyrimidin-4-yl) oxy) phenyl) carbamic acid tert-butyl ester (200 mg) and trifluoroacetic acid (5 mL). The reaction system reacts for 4 hours at room temperature, and after the reaction is finished, the organic solvent is removed by reduced pressure distillation to obtain the product 4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) aniline. MS m/z (ESI): 241.11[ M+H ]] +
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 3-trifluoromethoxyphenylacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 1, N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (3- (trifluoromethoxy) phenyl) acetamide. MS m/z (ESI) 443.13[ M+H ]] +
Example 2N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (2- (trifluoromethoxy) phenyl) acetamide
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 2-trifluoromethoxyphenylacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 2, N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (2- (trifluoromethoxy) phenyl) acetamide. MS m/z (ESI) 443.13[ M+H ]] +
Example 3.2- (4- (difluoromethoxy) phenyl) -N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) acetamide
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 4-difluoromethoxy phenylacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 3,2- (4- (difluoromethoxy) phenyl) -N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) acetamide. MS m/z (ESI): 425.14[ M+H ]] +
EXAMPLE 4N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 4-trifluoromethylphenylacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 4, N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 427.14[ M+H ]] +
Example 5N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethoxy) phenyl) acetamide
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 4-trifluoromethoxyphenylacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added, respectively, with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 5,N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethoxy) phenyl) acetamide. MS m/z (ESI) 443.13[ M+H ]] +
EXAMPLE 6N- (4- (difluoromethoxy) benzyl) -4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) aniline
1mL of N, N-dimethylformamide was added to a 25mL round-bottomed flask, and 1-bromomethyl-4-difluoromethoxybenzene (30 mg) and Cs were added with stirring at room temperature 2 CO 3 (50 mg) and 4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 6, N- (4- (difluoromethoxy) benzyl) -4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) aniline. MS m/z (ESI): 397.15[ M+H ]] +
EXAMPLE 7N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (6- (trifluoromethyl) pyridin-3-yl) acetamide
1mL of tetrahydrofuran was added to a 10mL round bottom flask, and 6-trifluoromethyl-3-pyridineacetic acid (5 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 7, N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (6- (trifluoromethyl) pyridin-3-yl) acetamide. MS m/z (ESI): 428.13[ M+H ]] +
Example 8.4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) -N- ((6- (trifluoromethyl) pyridin-3-yl) methyl) aniline
5mL of methanol was added to a 25mL round bottom flask and 4- ((7-methyl-7H-pyrrolo [2, 3-D) was added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (20 mg) and acetic acid (0.1 mL). 6- (trifluoromethyl) nicotinaldehyde (30 mg) was added under ice-bath conditions. The reaction mixture was stirred at 0 ℃ for 0.5 hours, then the ice bath was removed and stirring was continued at room temperature for 3 hours. After completion of the reaction, sodium cyanoborohydride (10 mg) was added under ice-bath conditions. The reaction mixture was stirred at 0 ℃ for 2 hours. After the reaction is completed, saturated NaHCO is used 3 The reaction was quenched. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 8,4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) -N- ((6- (trifluoromethyl) pyridin-3-yl) methyl) aniline. MS m/z (ESI) 400.14[ M+H ]] +
EXAMPLE 9N- (4- ((7- (methylsulfonyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
Into a 25mL round bottom flask was added 5mL of tetrahydrofuran, and tert-butyl (4-hydroxyphenyl) carbamate (105 mg) and NaH (21 mg in 60% mineral oil) were added with stirring at 0deg.C. After 30 minutes of reaction at 0deg.C, 4-bromo-7H-pyrrolo [2,3-D ] is added ]Pyrimidine (100 mg) was reacted for 60 minutes, and then the reaction was continued at room temperature for 12 hours. After the reaction was completed, 1mL of saturated NH was used 4 And (3) performing a Cl neutralization reaction. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined.The organic phase was washed with 20mL of water, 20mL of saturated NaCl, followed by anhydrous Na 2 SO 4 And (5) drying. The organic phase was distilled under reduced pressure to give a crude product, which was chromatographed on a silica gel column to give a yellow product (4- ((7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) carbamic acid tert-butyl ester. MS m/z (ESI): 327.15[ M+H ]] +
1mL of methylene chloride was added to a 25mL round bottom flask, and the mixture was stirred at room temperature (4- ((7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) carbamic acid tert-butyl ester (50 mg) and trifluoroacetic acid (1 mL). The reaction system reacts for 4 hours at room temperature, and after the reaction is finished, the organic solvent is removed by reduced pressure distillation to obtain the product (4- ((7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) aniline. MS m/z (ESI): 227.09[ M+H ]] +
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 4-trifluoromethylphenylacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and (4- ((7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. Distilling the organic phase under reduced pressure to obtain crude product, and subjecting the crude product to silica gel column chromatography to obtain yellow product N- (4- ((7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 413.12[ M+H ]] + .
1mL of tetrahydrofuran was added to a 25mL round bottom flask, and methanesulfonyl chloride (10 mg), TEA (0.03 mL) and N- (4- ((7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (30 mg). The reaction system was allowed to react at room temperature for 3 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 9,N- (4- ((7- (methylsulfonyl) -7H-pyrrolo [ 2),3-D]pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 491.10[ M+H ]] +
Example 10N- (4- ((7-acetyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of tetrahydrofuran was added to a 25mL round bottom flask, and acetic anhydride (15 mg) and N- (4- ((7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature ]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (30 mg). The reaction system was allowed to react at room temperature for 3 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 10, N- (4- ((7-acetyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 455.13[ M+H ]]+。
EXAMPLE 11N- (4- ((7-isopropyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 25mL round bottom flask, and 2-iodopropane (25 mg) and N- (4- ((7H-pyrrolo [2, 3-D) were added with stirring at room temperature, respectively]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (30 mg) and cesium carbonate (50 mg). The reaction system was allowed to react at room temperature for 3 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. After distillation of the organic phase under reduced pressureObtaining a crude product, and obtaining a yellow solid compound 11, N- (4- ((7-isopropyl-7H-pyrrolo [2, 3-D) after the crude product is subjected to silica gel column chromatography]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 455.17[ M+H ]]+。
EXAMPLE 12N- (4- ((7- (2, 2-trifluoroethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 25mL round bottom flask, and 1, 1-trifluoro-2-iodoethane (30 mg) and N- (4- ((7H-pyrrolo [2, 3-D) were added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (30 mg) and cesium carbonate (50 mg). The reaction system was allowed to react at room temperature for 3 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 12, N- (4- ((7- (2, 2-trifluoroethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 495.13[ M+H ] ] +
EXAMPLE 13N- (4- ((7- (2, 2-difluoroethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 25mL round bottom flask, and 1, 1-difluoro-2-iodoethane (25 mg) and N- (4- ((7H-pyrrolo [2, 3-D) were added with stirring at room temperature, respectively]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (30 mg) and cesium carbonate (50 mg). The reaction system was allowed to react at room temperature for 3 hours. After the reaction, the reaction mass was diluted with 10mL of waterIs tied up. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 13, N- (4- ((7- (2, 2-difluoroethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 477.13[ M+H ]] +
EXAMPLE 14N- (4- ((7- (methoxyethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature ]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 2-bromoethyl methyl ether (50 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 14, N- (4- ((7- (methoxyethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 471.16[ M+H ]] +
EXAMPLE 15N- (4- ((7-ethyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) Phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), ethyl iodide (45 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 15, N- (4- ((7-ethyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 441.15[ M+H ]] +
EXAMPLE 16N- (4- ((7-propionyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
/>
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg) and propionic anhydride (62 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 16, N- (4- ((7-propionyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 469.15[ M+H ]] +
EXAMPLE 17N- (4- ((7-isobutyryl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
At 101mL of N, N-dimethylformamide was added to a mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg) and isobutyric anhydride (76 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 17, N- (4- ((7-isobutyryl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 483.16[ M+H ]] +
EXAMPLE 18N- (4- ((7-butyryl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg) and butyric anhydride (76 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 18, N- (4- ((7-butyryl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 483.16[ M+H ]] +
EXAMPLE 19N- (4- ((7- (2- (diethylamino) ethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), (2-bromoethyl) - (diethyl) amine (76 mg) and cesium carbonate (50 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 19, N- (4- ((7- (2- (diethylamino) ethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 512.23[ M+H ] ] +
EXAMPLE 20N- (4- ((7- (2-fluoroethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 1-bromo-2-fluoroethane (37 mg) and cesium carbonate (50 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 20, N- (4- ((7- (2-fluoroethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 459.14[ M+H ]] +
EXAMPLE 21N- (4- ((7- (cyclopentylmethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature ]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), bromomethylcyclopentane (47 mg) and cesium carbonate (50 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 21, N- (4- ((7- (cyclopentylmethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 495.20[ M+H ]] +
EXAMPLE 22N- (4- ((7- (2-ethoxyethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 2-bromoethyl ether (50 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 22, N- (4- ((7- (2-ethyl) B)Oxyethyl) -7H-pyrrolo [2,3-D]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 485.18[ M+H ]] +
Example 23N- (4- ((7- ((3-Methyloxetan-3-yl) methyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 3-bromomethyl-3-methyl-1-oxetane (50 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 23, N- (4- ((7- ((3-methyl oxetan-3-yl) methyl) -7H-pyrrolo [2, 3-D) ]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 497.18[ M+H ]] +
EXAMPLE 24N- (4- ((7- (2- (dimethylamino) ethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), (2-bromoethyl) dimethylamine (50 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. Mixed solutionThe organic phases were combined by extraction three times with 20mL ethyl acetate. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 24, N- (4- ((7- (2- (dimethylamino) ethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 484.20[ M+H ]] +
EXAMPLE 25N- (4- ((7- (cyanomethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), iodoacetonitrile (50 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 25, N- (4- ((7- (cyanomethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 452.13[ M+H ]] +
EXAMPLE 26N- (4- ((7- (fluoromethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl)Phenyl) acetamide (100 mg), fluoroiodomethane (77 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 26, N- (4- ((7- (fluoromethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 445.13[ M+H ]] +
EXAMPLE 27N- (4- ((7- (2-hydroxyethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 2-bromoethanol (50 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 27, N- (4- ((7- (2-hydroxyethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 457.15[ M+H ] ] +
EXAMPLE 28N- (4- ((7-acetyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethoxy) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask, and 4-trifluoromethoxy-phenylacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7H-pyrrolo [2, 3-D) were added, respectively, with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. Distilling the organic phase under reduced pressure to obtain crude product, and subjecting the crude product to silica gel column chromatography to obtain N- (4- ((7H-pyrrolo [2, 3-d)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethoxy) phenyl) acetamide. MS m/z (ESI) 429.12[ M+H ]] +
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-d) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethoxy) phenyl) acetamide (100 mg) and acetic anhydride. The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 28, N- (4- ((7-acetyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethoxy) phenyl) acetamide. MS m/z (ESI): 471.14[ M+H ]] +
EXAMPLE 29N- (4- ((7- (oxetan-3-yl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 3-iodooxetane (53 mg) and cesium carbonate (184 mg). Reverse-rotationThe reaction system was then reacted at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase was distilled under reduced pressure to give a crude product, which was subjected to silica gel column chromatography to give the compound 29, N- (4- ((7- (oxetan-3-yl) -7H-pyrrolo [2, 3-D), as a yellow solid]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 469.15[ M+H ] ] +
EXAMPLE 30N- (4- ((7- (3-hydroxypropyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 3-bromopropanol (50 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 30, N- (4- ((7- (3-hydroxypropyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 471.16[ M+H ]] +
EXAMPLE 31 (4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidin-7-yl) acetic acid
1mL of N, N-dimethylformamide was added to a 10mL round bottom flaskAdding N- (4- ((7H-pyrrolo [2, 3-D) to the mixture at room temperature under stirring ]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 2-iodoacetic acid (55 mg) and cesium carbonate (235 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase was distilled under reduced pressure to give a crude product, which was subjected to silica gel column chromatography to give Compound 31, (4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) as a yellow solid]Pyrimidin-7-yl) acetic acid. MS m/z (ESI): 471.13[ M+H ]] +
EXAMPLE 32 phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxylate
Into a 25mL round bottom flask was added 10mL tetrahydrofuran, and phenyl chloroformate (91 mg), TEA (0.14 mL) and N- (4- ((7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (200 mg). The reaction was then allowed to react at room temperature for 3 hours. And after the reaction is finished, distilling under reduced pressure to obtain a crude product. To the crude product was added 20mL of water, the mixed solution was extracted three times with 30mL of ethyl acetate, and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 32, phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxylic acid esters. MS m/z (ESI): 533.14[ M+H ]] +
EXAMPLE 33N-methyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), methylamine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction was then allowed to react at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 33, N-methyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI) 470.14[ M+H ] ] +
EXAMPLE 34N-ethyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), ethylamine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction was then allowed to react at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 34, N-ethyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI) 484.16[ M+H ]] +
Example 35N- (4- ((7- (pentan-3-yl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature ]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 3-bromopentane (45 mg) and cesium carbonate (235 mg). The reaction system was heated to 50℃and then reacted for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 35, N- (4- ((7- (pentane-3-yl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 483.20[ M+H ]] +
EXAMPLE 36N- (4- ((7-isopropyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethoxy) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-d) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethoxy) phenyl) acetamide (100 mg), 2-iodo-propane (45 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 36, N- (4- ((7-isopropyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) benzenePhenyl) -2- (4- (trifluoromethoxy) phenyl) acetamide. MS m/z (ESI): 471.45[ M+H ]] +
EXAMPLE 37N- (4- ((7- (piperidin-4-ylmethyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 4- (bromomethyl) piperidine (50 mg) and cesium carbonate (245 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 37, N- (4- ((7- (piperidin-4-ylmethyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 510.21[ M+H ] ] +
EXAMPLE 38N- (4- ((7- (azetidin-3-yl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 3-iodo-azetidine (82 mg) and cesium carbonate (245 mg). The reaction system was heated to 50℃and then reacted for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, respectively, and thenBy anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 38, N- (4- ((7- (azetidin-3-yl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 468.16[ M+H ]] +
EXAMPLE 39N- (4- ((7- (3-methoxypropyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature ]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 1-iodo-3-methoxypropane (45 mg) and cesium carbonate (184 mg). The reaction was then allowed to react at room temperature for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 39, N- (4- ((7- (3-methoxypropyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 484.24[ M+H ]] + .
EXAMPLE 40N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (pyridin-2-yl) acetamide
1mL of tetrahydrofuran was added to a 25mL round bottom flask, and 2- (pyridin-2-yl) acetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (20 mg). The reaction system was reacted at room temperature for 14 hours. Reaction junctionAfter the ligation, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 40, N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (pyridin-2-yl) acetamide. MS m/z (ESI) 360.15[ M+H ]] +
EXAMPLE 41N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (pyridin-3-yl) acetamide
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 2- (pyridin-3-yl) acetic acid (20 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 41, N- (4- ((7-methyl-7H-pyrrole [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (pyridin-3-yl) acetamide. MS m/z (ESI) 360.15[ M+H ]] +
EXAMPLE 42N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (pyridin-4-yl) acetamide
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 2- (pyridin-4-yl) acetic acid (20 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) anilines30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 42, N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (pyridin-4-yl) acetamide. MS m/z (ESI) 360.15[ M+H ]] +
EXAMPLE 43.2- (furan-2-yl) -N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) acetamide
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 2-furoacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 43,2- (furan-2-yl) -N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) acetamide. MS m/z (ESI): 349.13[ M+H ]] +
EXAMPLE 44.2- (benzofuran-2-yl) -N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) acetamide
Into a 25mL round bottom flask was charged 1mL of tetrahydrofuran, and 2- (benzofuran-2-yl) acetic acid (25 mg), HATU (30 mg), DIPEA (0) were added, respectively, with stirring at room temperature05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 44,2- (benzofuran-2-yl) -N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) acetamide. MS m/z (ESI): 399.15[ M+H ]] +
EXAMPLE 45N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (thiophen-3-yl) acetamide
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 2- (thiophen-3-yl) acetic acid (30 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added, respectively, with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 45, N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (thiophen-3-yl) acetamide. MS m/z (ESI): 365.11[ M+H ]] +
EXAMPLE 46N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (thiophen-2-yl) acetamide
1mL of tetrahydrofuran was added to a 10mL round bottom flask and stirred at room temperature2- (thiophen-2-yl) acetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 46, N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (thiophen-2-yl) acetamide. MS m/z (ESI): 365.11[ M+H ]] +
Example 47.2- (2-fluoro-4- (trifluoromethyl) phenyl) -N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) amino) phenyl) acetamide
1mL of tetrahydrofuran was added to a 10mL round bottom flask, and 2-fluoro-4- (trifluoromethyl) phenylacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 47,2- (2-fluoro-4- (trifluoromethyl) phenyl) -N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) amino) phenyl) acetamide. MS m/z (ESI): 445.13[ M+H ]] +
EXAMPLE 48N-isopropyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), isopropylamine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction was then allowed to react at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 48, N-isopropyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI): 498.18[ M+H ]] +
EXAMPLE 49N-propyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), propylamine (50 mg) and N, N-diisopropylethylamine (0.1 mL). The reaction was then allowed to react at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 49, N-propyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI): 498.18[ M+H ]] +
EXAMPLE 50N-butyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), butylamine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction was then allowed to react at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 50, N-butyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI) 512.19[ M+H ] ] +
EXAMPLE 51N-cyclopropyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), cyclopropylamine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction was then allowed to react at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 51, N-cyclopropyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyridinePyrrolo [2,3-D]Pyrimidine-7-carboxamide. MS m/z (ESI): 496.16[ M+H ]] +
EXAMPLE 52N-cyclobutyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature ]Pyrimidine-7-carboxylate (50 mg), cyclobutylamine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction was then allowed to react at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 52, N-cyclobutyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI) 510.18[ M+H ]] +
EXAMPLE 53N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (1H-pyrrol-2-yl) acetamide
Into a 10mL round bottom flask was added 1mL tetrahydrofuran, and pyrrole-2-acetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. Distilling the organic phase under reduced pressure to obtain crude productThe crude product is subjected to silica gel column chromatography to obtain yellow solid compound 53, N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (1H-pyrrol-2-yl) acetamide. MS m/z (ESI) 348.15[ M+H ]] +
EXAMPLE 54.2- (3-fluoro-4- (trifluoromethyl) phenyl) -N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) acetamide
1mL of tetrahydrofuran was added to a 10mL round bottom flask, and 3-fluoro-4- (trifluoromethyl) phenylacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 54,2- (3-fluoro-4- (trifluoromethyl) phenyl) -N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) acetamide. MS m/z (ESI): 445.13[ M+H ]] +
EXAMPLE 55N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -3- (6- (trifluoromethyl) pyridin-3-yl) propanamide
1mL of tetrahydrofuran was added to a 10mL round bottom flask, and 3- (6- (trifluoromethyl) pyridin-3-yl) propionic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 55, N- (4- ((7-methyl-7H-pyrrole [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -3- (6- (trifluoromethyl) pyridin-3-yl) propanamide. MS m/z (ESI) 442.15[ M+H ]] +
EXAMPLE 56N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (5- (trifluoromethyl) pyridin-2-yl) acetamide
1mL of tetrahydrofuran was added to a 10mL round bottom flask, and 2- (5- (trifluoromethyl) pyridin-2-yl) acetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 56, N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (5- (trifluoromethyl) pyridin-2-yl) acetamide. MS m/z (ESI): 428.13[ M+H ]] +
Example 57.2- (furan-3-yl) -N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) acetamide
1mL of tetrahydrofuran was added to a 10mL round bottom flask, and 2- (furan-3-yl) acetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system is reversed at room temperatureShould be 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 57,2- (furan-3-yl) -N- (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) acetamide. MS m/z (ESI): 349.13[ M+H ]] +
EXAMPLE 58N-cyclopentyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), cyclopentylamine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 58, N-cyclopentyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI) 524.19[ M+H ]] +
EXAMPLE 59N-cyclohexyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask, and phenyl was added with stirring at room temperature4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxylate (50 mg), cyclohexylamine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 59, N-cyclohexyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI): 538.21[ M+H ]] +
EXAMPLE 60N-isobutyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), 2-methylpropylamine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 60, N-isobutyl-4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI) 512.19[ M+H ] ] +
EXAMPLE 61N- (tetrahydro-2H-pyran-4-yl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), 4-aminotetrahydropyran (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 61, N- (tetrahydro-2H-pyran-4-yl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI): 540.19[ M+H ]] +
EXAMPLE 62N- (oxetan-3-yl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature ]Pyrimidine-7-carboxylate (50 mg), 3-oxetane (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 62, N- (oxetan-3-yl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. S m/z (ESI) 512.15[ M+H ]] +
EXAMPLE 63N- ((tetrahydro-2H-pyran-4-yl) methyl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
/>
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), (tetrahydro-2H-pyran-4-yl) methylamine (50 mg) and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase was distilled under reduced pressure to give a crude product, which was chromatographed on a silica gel column to give compound 63, N- ((tetrahydro-2H-pyran-4-yl) methyl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) as a yellow solid]Pyrimidine-7-carboxamide. MS m/z (ESI): 554.20[ M+H ]] +
EXAMPLE 64N- (4- ((7- (pyridin-3-yl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
3mL of 1, 4-dioxane was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 3-iodopyridine (59 mg), cuprous iodide (5 mg) and potassium phosphate (102 mg). The reaction system was then reacted for 14 hours while heating to 130 ℃. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 64, N- (4- ((7- (pyridine-3-yl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 490.15[ M+H ] ] +
EXAMPLE 65N- (4- ((7- (o-tolyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
3mL of 1, 4-dioxane was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 1-iodo-2-toluene (63 mg), cuprous iodide (5 mg) and potassium phosphate (102 mg). The reaction system was then reacted for 14 hours while heating to 130 ℃. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 65, N- (4- ((7- (o-tolyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 503.17[ M+H ]] +
EXAMPLE 66N- (4- ((7- (pyridin-2-yl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
3mL of 1, 4-dioxane was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature ]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 2-iodopyridine (53 mg), cuprous iodide (5 mg) and potassium phosphate (102 mg). The reaction system is heated to 130 DEG CThe reaction was continued for 14 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 66, N- (4- ((7- (pyridine-2-yl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 490.15[ M+H ]] +
EXAMPLE 67N- (4- ((7- (thiophen-2-yl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
3mL of 1, 4-dioxane was added to a 10mL round bottom flask and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 2-iodothiophene (61 mg), cuprous iodide (5 mg) and potassium phosphate (102 mg). The reaction system was then reacted for 14 hours while heating to 130 ℃. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 67, N- (4- ((7- (thiophene-2-yl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 495.11[ M+H ]] +
EXAMPLE 68N- (4- ((7- (2-fluorophenyl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
Into a 10mL round bottom flask was added 3mL of 1, 4-dioxane in the chamberAdding N- (4- ((7H-pyrrolo [2, 3-D) to the mixture under stirring at a temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 1-fluoro-2-iodobenzene (64 mg), cuprous iodide (5 mg) and potassium phosphate (102 mg). The reaction system was then reacted for 14 hours while heating to 130 ℃. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 68, N- (4- ((7- (2-fluorophenyl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 507.14[ M+H ] ] +
EXAMPLE 69N- (morpholin-4-yl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask, and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at zero degrees]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 4-aminomorpholine (20 mg), triphosgene (50 mg) and triethylamine TEA (184 mg). The reaction was then allowed to react at zero degrees for 1 hour. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 69, N- (morpholin-4-yl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI) 541.50[ M+H ]] +
EXAMPLE 70N- (4-methylpiperazin-1-yl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature ]Pyrimidine-7-carboxylate (50 mg), 4-methylpiperazin-1-amine (50 mg) and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 70, N- (4-methylpiperazin-1-yl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI): 554.21[ M+H ]] +
EXAMPLE 71N- (1-methylpiperidin-4-yl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), 4-amino-1-methylpiperidine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow product N- (1-methylpiperidin-4-yl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. M is MS m/z(ESI):553.22[M+H] +
EXAMPLE 72N- (2- (morpholin-4-yl) ethyl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), N- (2-aminoethyl) morpholine (50 mg) and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 72, N- (2- (morpholin-4-yl) ethyl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) ]Pyrimidine-7-carboxamide. MS m/z (ESI): 569.21[ M+H ]] +
EXAMPLE 73N- (3- (4-methylpiperazin-1-yl) propyl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), 3- (4-methylpiperazin-1-yl) propan-1-amine (50 mg) and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was separated with 20mL of water,20mL of saturated NaCl followed by anhydrous Na 2 SO 4 And (5) drying. The organic phase was distilled under reduced pressure to give a crude product, which was subjected to silica gel column chromatography to give compound 73, N- (3- (4-methylpiperazin-1-yl) propyl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) as a yellow solid]Pyrimidine-7-carboxamide. MS m/z (ESI): 596.26[ M+H ]]+。
Example 74N- (2-hydroxyethyl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), ethanolamine (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 74, N- (2-hydroxyethyl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI) 500.15[ M+H ]] +
EXAMPLE 75N- (3-hydroxypropyl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxamide
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylic acid ester (50 mg), 3-aminopropyl ester Alkyl-1-ol (50 mg) and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 75, N- (3-hydroxypropyl) -4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxamide. MS m/z (ESI) 514.17[ M+H ]] +
EXAMPLE 76.4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carboxylic acid methyl ester
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), methanol (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound, namely 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carboxylic acid methyl ester. MS m/z (ESI): 471.13[ M+H ]] +
EXAMPLE 77N- (4- ((7- (pyridin-4-yl) -7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide
/>
Firing at 10mL of round bottom3mL of 1, 4-dioxane was added to the flask, and N- (4- ((7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide (100 mg), 4-iodopyridine (53 mg), cuprous iodide (5 mg) and potassium phosphate (102 mg). The reaction system was then reacted for 14 hours while heating to 130 ℃. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 77, N- (4- ((7- (pyridin-4-yl) -7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 490.15[ M+H ] ] +
Example 78.4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) -N- ((6- (trifluoromethyl) pyridin-3-yl) methyl) aniline
5mL of methanol was added to a 25mL round bottom flask and 4- ((7-methyl-7H-pyrrolo [2, 3-D) was added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (20 mg) and acetic acid (0.1 mL). 6-trifluoromethylpyridine-3-aldehyde (30 mg) was added under ice-bath conditions. The reaction mixture was stirred at 0 ℃ for 0.5 hours, then the ice bath was removed and stirring was continued at room temperature for 3 hours. After completion of the reaction, sodium cyanoborohydride (10 mg) was added under ice-bath conditions. The reaction mixture was stirred at 0 ℃ for 2 hours. After the reaction is completed, saturated NaHCO is used 3 The reaction was quenched. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 78,4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) -N- ((6- (trifluoromethyl) pyridin-3-yl) methyl) aniline. MS m/z (ESI) 400.14[ M+H ]] +
Example 79N- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) benzyl) -2- (4- (trifluoromethyl) phenyl) acetamide
Into a 25mL round bottom flask was added 10mL of N, N-dimethylformamide, tert-butyl (4-hydroxybenzyl) carbamate (500 mg), 4-chloro-7-methylpyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine (310 mg) and cesium carbonate (1.4 g). The reaction system was added to 50℃followed by a reaction for 14 hours. After the completion of the reaction, the reaction was neutralized with 50mL of water. The mixed solution was extracted three times with 50mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 30mL of water, 30mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to give a crude product, which is chromatographed on a silica gel column to give a yellow product (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) benzyl) carbamic acid tert-butyl ester. MS m/z (ESI): 355.18[ M+H ]] +
5mL of methylene chloride was added to a 25mL round bottom flask and the mixture was stirred at room temperature (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy benzyl) carbamic acid tert-butyl ester (500 mg) and trifluoroacetic acid (5 mL). The reaction system reacts for 4 hours at room temperature, and after the reaction is finished, the organic solvent is removed by reduced pressure distillation to obtain the product (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl) methylamine. MS m/z (ESI) 255.12[ M+H ]] +
Into a 25mL round bottom flask was added 1mL tetrahydrofuran, and 4-trifluoromethylphenylacetic acid (42 mg), HATU (80 mg), DIPEA (0.1 mL) and (4- ((7-methyl-7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature ]Pyrimidin-4-yl) oxy) phenyl methylamine (50 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 79, N- (4- ((7-methyl-7H-pyrrolo)[2,3-D]Pyrimidin-4-yl) oxy) benzyl) -2- (4- (trifluoromethyl) phenyl) acetamide. MS m/z (ESI): 441.15[ M+H ]] +
EXAMPLE 80 (4- ((7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-4-yl) oxy) benzyl) - ((6- (trifluoromethyl) pyridin-3-yl) methyl) amine
5mL of methanol was added to a 25mL round bottom flask and the mixture was stirred at room temperature and added separately (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl methylamine (50 mg) and acetic acid (0.1 mL). 6-trifluoromethylpyridine-3-aldehyde (37 mg) was added under ice-bath conditions. The reaction mixture was stirred at 0 ℃ for 0.5 hours, then the ice bath was removed and stirring was continued at room temperature for 3 hours. After completion of the reaction, sodium cyanoborohydride (17 mg) was added under ice-bath conditions. The reaction mixture was stirred at 0 ℃ for 2 hours. After the reaction is completed, saturated NaHCO is used 3 The reaction was quenched. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase was distilled under reduced pressure to give a crude product, which was chromatographed on a silica gel column to give compound 80 as a yellow solid, (4- ((7-methyl-7H-pyrrolo [2, 3-d)]Pyrimidin-4-yl) oxy) benzyl) - ((6- (trifluoromethyl) pyridin-3-yl) methyl) amine. MS m/z (ESI) 414.15[ M+H ]] +
Example 81.4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) -N- (4- (trifluoromethyl) benzyl) aniline
5mL of methanol was added to a 25mL round bottom flask and 4- ((7-methyl-7H-pyrrolo [2, 3-D) was added separately with stirring at room temperature]Pyrimidin-4-yl) oxy) aniline (50 mg) and acetic acid (0.1 mL). 4-trifluoromethylbenzaldehyde (31 mg) was added thereto under ice-bath conditions. The reaction mixture was stirred at 0 ℃ for 0.5 hours, then the ice bath was removed and continued at room temperatureStirring for 3 hours. After completion of the reaction, sodium cyanoborohydride (14 mg) was added under ice-bath conditions. The reaction mixture was stirred at 0 ℃ for 2 hours. After the reaction is completed, saturated NaHCO is used 3 The reaction was quenched. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 81,4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) -N- (4- (trifluoromethyl) benzyl) aniline. MS m/z (ESI): 399.14[ M+H ]] +
EXAMPLE 82.4- (4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carbonyl) piperidine-1-carboxylic acid tert-butyl ester
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), 4-aminopiperidine-1-carboxylic acid tert-butyl ester (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 82,4- (4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carbonyl) piperidine-1-carboxylic acid tert-butyl ester. MS m/z (ESI): 639.25[ M+H ] ] +
Example 83.3- (4- (4- (2- (4- (trifluoromethyl) phenyl) acetamide) phenoxy) -7H-pyrrolo [2,3-D ] pyrimidine-7-carbonyl) piperidine-1-carboxylic acid tert-butyl ester
Into a 10mL round bottom flask was added 1mL of N, N-dimethylformamide and phenyl 4- (4- (2- (4- (trifluoromethyl) phenyl) acetamido) phenoxy) -7H-pyrrolo [2, 3-D) was added with stirring at room temperature]Pyrimidine-7-carboxylate (50 mg), 3-aminopiperidine-1-carboxylic acid tert-butyl ester (50 mg), and N, N-diisopropylethylamine (0.1 mL). The reaction system was reacted at room temperature for 10 hours. After the completion of the reaction, the reaction was neutralized with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain a yellow solid compound 83,3- (4- (4- (2- (4- (trifluoromethyl) phenyl) acetamide) phenoxy) -7H-pyrrolo [2, 3-D)]Pyrimidine-7-carbonyl) piperidine-1-carboxylic acid tert-butyl ester. MS m/z (ESI): 639.25[ M+H ]] +
Comparative example 1N- (4- ((7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -2- (3-trifluoromethyl) phenyl) acetamide
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask, and 3-trifluoromethylphenylacetic acid (25 mg), HATU (30 mg), DIPEA (0.05 mL) and 4- ((7H-pyrrolo [2, 3-D) were added separately with stirring at room temperature ]Pyrimidin-4-yl) oxy) aniline (30 mg). The reaction system was reacted at room temperature for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain the compound 1, N- (4- ((7H-pyrrolo [2, 3-D) of comparative example]Pyrimidin-4-yl) oxy) phenyl) -2- (3-trifluoromethyl) phenyl) acetamide. MS m/z (ESI) 413.12[ M+H ]] +
Comparative example 2.1- (4- ((7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -3- (4- (trifluoromethyl) phenyl) urea
Into a 25mL round bottom flask was added 10mL tetrahydrofuran, and 4- ((7H-pyrrolo [2, 3-D) was added separately with stirring at room temperature]Pyrimidin-4-yl) oxy-aniline (500 mg), phenyl chloroformate (200 mg) and N, N-diisopropylethylamine (0.5 mL). The reaction system was allowed to react at room temperature for 2 hours. After the completion of the reaction, the reaction system was diluted with 50mL of water. The mixed solution was extracted three times with 30mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 30mL of water, 30mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain phenyl (4- ((7H-pyrrolo [2, 3-D) ]Pyrimidin-4-yl) oxy) phenyl) carbamate. MS m/z (ESI): 347.35[ M+H ]] +
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask, and phenyl groups (4- ((7H-pyrrolo [2, 3-D) were added with stirring at room temperature]Pyrimidin-4-yl) oxy) phenyl) carbamate (50 mg), 4-trifluoromethylaniline (20 mg) and DIPEA (0.05 mL). The reaction system was heated to 50℃and reacted for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain the compound 2,1- (4- ((7H-pyrrolo [2, 3-D) of comparative example]Pyrimidin-4-yl) oxy) phenyl) -3- (4- (trifluoromethyl) phenyl) urea. MS m/z (ESI) 414.37[ M+H ]] +
Comparative example 3.1- (4- ((7-methyl-7H-pyrrolo [2,3-D ] pyrimidin-4-yl) oxy) phenyl) -3- (3-trifluoromethyl-4-chlorophenyl) urea
Into a 25mL round bottom flask was added 10mL tetrahydrofuran, and the mixture was stirred at room temperatureAdding 4- ((7-methyl-7H-pyrrolo [2, 3-D) respectively]Pyrimidin-4-yl) oxy-aniline (500 mg), phenyl chloroformate (200 mg) and N, N-diisopropylethylamine (0.5 mL). The reaction system was allowed to react at room temperature for 2 hours. After the completion of the reaction, the reaction system was diluted with 50mL of water. The mixed solution was extracted three times with 30mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 30mL of water, 30mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase was distilled under reduced pressure to give a crude product, which was chromatographed on a silica gel column to give a yellow product (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl carbamate. MS m/z (ESI): 361.38[ M+H ]] +
1mL of N, N-dimethylformamide was added to a 10mL round bottom flask, and the mixture was stirred at room temperature and then added to each of the two (4- ((7-methyl-7H-pyrrolo [2, 3-D)]Pyrimidin-4-yl) oxy) phenyl carbamate (50 mg), 3-trifluoromethyl-4-chloroaniline (20 mg), and DIPEA (0.05 mL). The reaction system was heated to 50℃and reacted for 14 hours. After the completion of the reaction, the reaction system was diluted with 10mL of water. The mixed solution was extracted three times with 20mL of ethyl acetate and the organic phases were combined. The organic phase was washed with 20mL of water, 20mL of saturated NaCl, and then with anhydrous Na 2 SO 4 And (5) drying. The organic phase is distilled under reduced pressure to obtain a crude product, and the crude product is subjected to silica gel column chromatography to obtain the compound 3,1- (4- ((7-methyl-7H-pyrrolo [2, 3-D) of the comparative example]Pyrimidin-4-yl) oxy) phenyl) -3- (3-trifluoromethyl-4-chlorophenyl) urea. MS m/z (ESI) 462.09[ M+H ]] +
Example 84 effects on proliferation of cancer cells
The inhibition of cancer cell proliferation by the compounds herein was further evaluated by testing the effect of the compounds of the invention on cancer cell growth. In the examples, different concentrations (0.000508. Mu.M, 0.00152. Mu.M, 0.00457. Mu.M, 0.0137. Mu.M, 0.0411. Mu.M, 0.123. Mu.M, 0.370. Mu.M, 1.11. Mu.M, 3.33. Mu.M, 10. Mu.M in DMSO) of a compound of the invention were added to BaF3 (purchased from ATCC), TEL-cKIT-BaF3 (built in this laboratory), TEL-FLT3-BaF3 (built in this laboratory), TEL-CSF1R-BaF3 (built in this laboratory) and M-NSF-60 (purchased from Nanjac, china, a M-CSF (CSF-1) mediated mouse myeloblast cell line, which was used to evaluate the inhibition of cancer cell proliferation by the compound At the time of preparation, M-CSF at a concentration of 50 ng/mL) was added to the cell culture medium and incubated for 72 hours. The incubated cells were examined by CCK-8 (purchased from MedChem Express, china) cell viability assay kit (CCK-8 was reduced by dehydrogenase in living cells to yellow formazan product with high water solubility, the amount of formazan produced was proportional to the number of living cells), the number of living cells was quantified by a microplate reader, and GI of each compound was calculated 50 (the results are shown in Table 2).
The construction methods of TEL-CSF1R-BaF3 (cell line stably expressing CSF1R kinase), TEL-cKIT-BaF3 (cell line stably expressing cKIT kinase) and TEL-FLT3-BaF3 (cell line stably expressing FLT3 kinase) cell lines are as follows: PCR amplified human CSF1R, cKIT, FLT kinase domain sequences, respectively, and inserted into MSCV-Puro vector (Clontech) with N-terminal TEL fragment, stably transferred into mouse BaF3 cells (purchased from ATCC in the United states) by retrovirus method, and IL-3 growth factor was removed, finally obtaining cell line dependent on CSF1R, cKIT, FLT kinase transfer protein.
As shown in Table 2, the compounds of the invention have strong inhibition effect on proliferation of both TEL-CSF1R-BaF3 and M-CSF mediated mouse myeloblasts line M-NSF-60 expressing CSF1R kinase, which indicates that the series of compounds have strong inhibition effect on CSF1R target, and can be used for treating related diseases caused by activation of CSF1R kinase mediated signaling pathway. In addition, the compounds of the invention are capable of selectively inhibiting proliferation of cell TEL-CSF1R-BaF3 expressing CSF1R kinase relative to cKIT and FLT3 kinase. In addition, since CSF1R kinase is a key protein controlling macrophage polarization in tumor microenvironment, the conversion of macrophages into M2 type macrophages that promote cancer progression can be reduced by inhibiting the activity of CSF1R kinase, increasing the conversion of M1 type macrophages that inhibit cancer progression.
TABLE 2
/>
/>
Example 85: cell transformation experiments
RAW264.7 cells (purchased from ATCC, usa) were blown off using a pipette gun, the cells were added to a centrifuge tube, placed in a centrifuge, and centrifuged at 800rpm for 4 minutes. Cell supernatants were discarded according to 0.5X10 5 The density per ml was plated in 6 well transparent plates and incubated with 1640 medium containing 10% FBS (1640 medium from Corning, USA, FBS from Excel, australia) at 37℃with 5% CO 2 Culturing in an incubator for 12 hours; then, taking out the 6-hole transparent plate, adding 20ng/ml interleukin 4 factor (purchased from Beijing Yiqiao Shenzhou of China) into 1640 culture medium, and then adding the interleukin 4 factor and the interleukin 4 factor into the cell holes of the 6-hole transparent plate together, and culturing the cells in the culture medium for 24 hours; observing the cell morphology, if the cells gradually change from round to spindle, detecting the secretion of interleukin 10 in the culture medium supernatant by using a mouse IL-10ELISA kit (purchased from Wuhansai Peltier in China) according to the kit instruction, and if the secretion concentration of interleukin 10 increases, the RAW264.7 cells are induced to be M2 type macrophages. After successful induction of cells, the invention compound 54, control compound BLZ945 and PLX3397 (purchased from med chem Express in china) were treated with a random lottery of gradient dilutions for 72 hours, respectively, and then the cells and supernatants were harvested for qPCR and ELISA experiments, respectively.
qPCR-cell RNA extraction
The experiment used an RNA extraction kit (purchased from China Tiangen Biochemical, cat# DP 451) and used a NanoDrop nucleic acid concentration meter to measure the concentration of RNA in the sample according to the instructions and dilute the RNA solution to the same concentration.
Real-time fluorescent quantitative PCR experiment
The experiment used One Step TB of TaKaRaPrimeScript TM PLUS RT-PCR Kit (available from Takara Bio Inc., cat# RR 096A) containing the following components:
the operation steps are as follows:
(1) the PCR primers were removed and placed on ice. The primer sequences used were as follows:
(2) RT-PCR reaction solutions were prepared on ice as follows.
(3) After the RT-PCR reaction reagent is prepared, the mixture is mixed and split into 8 connecting pipes, 2 microlitres of RNA sample is added into the pipes, the pipe bottom is flicked to the solution, the solution is mixed uniformly, and the mixture is put into a centrifuge for slight centrifugation.
(4) Opening the fluorescent quantitative PCR instrument until the self-detection of the instrument is finished, placing a sample into the instrument, and performing PCR reaction according to the following procedures: the first step is reverse transcription: 42 ℃ for 5 minutes, 95 ℃ for 10 seconds, 1Cycle; the second step is PCR amplification: 95℃for 5 seconds, 60℃for 20 seconds, 40Cycles.
(5) After the experiment was completed, the sample was taken out and the instrument was turned off. Using Roche analysis software 96SW 1.1 analysis data, and the results of the analysis were plotted by introducing the analyzed data into Graphpad prism8.0, see FIG. 1.
Experiments show that after cells are induced into M2 type macrophages, the level of secreted IL-10 in the M2 type macrophages is far higher than that of the M1 type macrophages, which indicates that the IL-10 is mainly secreted by the M2 type cells and the M1 type macrophages are less secreted. As shown in FIG. 1, M0 represents the amount of IL-10 secreted by RAW264.7 cells without any treatment, M2 represents the amount of IL-10 secreted by RAW264.7 cells polarized to M2 type macrophages after treatment with interleukin 4 factor as described in this example, and the other columns represent the amount of IL-10 secreted by M2 type macrophages after polarization to M2 type macrophages before addition of compound 54 or control compound BLZ945, PLC3397 of the invention. The level of IL-10 secretion in M2-type macrophages showed a dose-dependent decrease with increasing drug concentration, indicating that M2-type cells in macrophages gradually decrease and gradually convert to M1-type cells. The compound disclosed by the invention can reduce the conversion of macrophages to M2 type and promote the conversion of macrophages to M1 type, so that the growth of tumors is inhibited and the immune curative effect of cancer is enhanced.
EXAMPLE 86 pharmacokinetic parameter detection
The present example used male SD rats, 180-220g (purchased from university of Anhui medical science laboratory animal center, china). Animals were kept in separate air-feeding cages, 6 per cage. The feeding conditions were as follows: the temperature is 20-26 ℃, the humidity is 35-75%, the illumination is 12 hours, the darkness is achieved, the corncob padding is replaced once a week, the corncob padding is free to eat, free to drink water and the tail is marked by a digital mark. During the course of the experiment, animal feeding and use strictly followed the specifications of the international laboratory animal assessment regulatory committee.
The preparation method of the compound is as follows. Precisely weighing 10mg of a compound to be measured, placing the compound into a sterile small bottle, dissolving the compound with a small amount of DMSO, and then adding 5% glucose solution to a volume of 5mL to obtain a gastric lavage test solution with the concentration of 2 mg/mL; precisely measuring 0.5mL of 2mg/mL of the gastric lavage test solution, adding 4.5mL of 5% glucose solution to 5mL, and obtaining intravenous injection test solution with the concentration of 0.2 mg/mL. Is prepared at present in experiments.
The 6 SD rats were randomly divided into two groups and the synthesized compounds were administered by intragastric and tail vein injection, respectively. The stomach-filling group is used for 0h before administration and 5min, 15min, 30min, 1h, 1.5h, 2h, 4h, 6h, 9h, 12h and 24h after administration; tail vein groups were subjected to blood sample collection from the retroorbital venous plexus at 0h before and 2min, 5min, 15min, 30min, 1h, 2h, 4h, 6h, 9h, 12h after administration by about 0.3mL, placed in a centrifuge tube of 1.5mL with heparin (Sigma, usa), centrifuged at 6000rpm for 3min to separate plasma, and then the upper 100 μl of plasma was taken in a new centrifuge tube of 1.5mL, stored at-80 ℃ and tested.
Precisely weighing 10mg of a compound standard substance to be measured in a 10mL volumetric flask, adding methanol to dissolve and fix the volume, and uniformly mixing to obtain 1mg/mL stock solution. Then the mixture is diluted step by methanol to obtain serial working solutions with the concentration of 0.01, 0.02, 0.05, 0.1, 0.2, 0.5, 1, 2, 5, 10 and 20 mug/mL respectively, and the serial working solutions are put into a refrigerator with the temperature of 4 ℃ for standby.
Taking 11 centrifuge tubes, respectively adding 10 mu L of the working solution, then adding 90 mu L of blank rat plasma, uniformly mixing to ensure that the concentration of the compound in the rat plasma is 1, 2, 5, 10, 20, 50, 100, 200, 500, 1000 and 2000ng/mL, then adding 20 mu L of internal standard caffeine solution (200 ng/mL) (Chinese food and drug assay institute), swirling for 10s, then adding 400 mu L of methanol, swirling for 10min, centrifuging at 16000rpm for 5min, taking 70 mu L of supernatant into an inner tube of a sample injection bottle, and injecting 5 mu L for LC-MS/MS analysis and determination. And (3) carrying out linear regression by taking the peak area ratio As/Ais of the sample and the internal standard As an ordinate and the concentration C (mug/mL) As an abscissa and taking 1/C2 As a weight coefficient to obtain a standard curve of the compound in the rat plasma.
Taking 100 mu L of rat plasma to be tested stored at the temperature of minus 80 ℃, adding 20 mu L of an internal standard caffeine solution (200 ng/mL), swirling for 10s, then adding 400 mu L of methanol, swirling for 10min, centrifuging at 16000rpm for 5min, taking 70 mu L of supernatant into an inner tube of a sample injection bottle, and injecting 5 mu L of sample for LC-MS/MS analysis and determination.
The LC-MS/MS assay method is as follows. Experimental instrument: API 4000 triple quaternary rod detector (AB SCIEX Co., U.S.A.), operating software was Analyst 1.5.1 (applied biosystems Co., U.S.A.); the device comprises an Shimadzu LC-30AD liquid pump, an Shimadzu DGU-20A degassing unit, an Shimadzu CTO-30A column incubator and a SIL-30AC automatic sampler (Shimadzu corporation). Chromatographic conditions: the column was Hedera ODS-2 (Jiangsu Hanbang technology Co., ltd., china), dim. (mm): 150X 2.1, pro.No.: h18100205.15; ser.no: c981210513; column temperature 40 ℃; mobile phase A is water phase (containing 0.1% formic acid), mobile phase B is methanol, gradient elution is 0-0.5min, B10%; 0.5-1.0min, B90%; 1.0 to 5.0min, B90%; 5.0-5.5min, B10%; 5.5-7min, B10%. The flow rate was 0.3mL/min. Sample injection amount: 5. Mu.L. Mass spectrometry conditions: the ion source is a Turbo Spray source, and CAD is 10; curtain gas (CUR) 25; the heating temperature is 500 ℃; the ion source gas GS1 is 45; the ion source gas GS2 is 45; spray voltage 5500V; the temperature in the source was 500 ℃.
The results are shown in Table 3 below. The results show that compound 54, whether administered intravenously or intragastrically, has higher Cmax, AUC and oral availability at the highest concentration in the blood than the compounds of comparative examples 1 and 2. Therefore, by introducing substituents on the pyrimidopyrrole ring, the absorption of the compounds of the invention in rats is greatly improved, and the oral availability is also improved to some extent.
TABLE 3 Table 3
Parameters (parameters) Unit (B) Compound 54 Comparative example 1 Comparative example 2
Cmax ng/ml 1193±270 38±12 311±101
AUC0-t hr*ng/ml 13703±2514 214±83 783±121
F 111%±1.35% 4.2%±2.66% 11.1%±2.8%
EXAMPLE 87 in vivo efficacy detection
In this example, experimental results of compound 54 and control compound PLX3397 (purchased from MedChem Express, china) in a mouse model of M-NFS-60 and MC38 cells, respectively, were tested.
The experimental procedure was as follows:
(1) Balb/C nu male mice used for feeding the 6-week-old M-NFS-60 cell transplantation tumor model and C57BL/6J male mice used for the MC38 cell transplantation tumor model were purchased from Jiangsu Ji-kang biotechnology Co., ltd, all of which were fed into SPF-class laboratories, and were autoclaved with drinking water and padding, and all of the operations on the mice were performed under aseptic conditions.
(2) About 1X 10 injections were made subcutaneously on day 0 in the left dorsum of Balb/c nu mice, respectively 6 M-NFS-60 cells (purchased from ATCC) were subcutaneously injected at the left back of C57BL/6J mice with about 1X 10 injections, respectively 6 Individual MC38 cells (purchased from ATCC).
(3) For the mouse model of M-NFS-60 cells, the corresponding mice were orally administered methyl cellulose (HKI) (purchased from China) vehicle once daily (5 mice) daily, starting on day 6; compound 54 at doses of 25, 50, 100mg/kg murine weight once daily (5 mice); PLX-3397 at a dose of 50mg/kg murine weight was administered once daily (5 mice). For the mouse model of MC38 cells, from day 6, the corresponding mice were orally dosed with 10% hs-15 (purchased from basf, germany) vehicle (5 mice) daily; compound 54 at a dose of 25mg/kg murine weight once daily (5 mice); PLX-3397 at a dose of 50mg/kg murine weight was administered once daily (5 mice).
(4) From day 6, the body weights of the mice were recorded daily, and the effect of compound 54 and control compound PLX-3397 on the body weights of the mice in the different model mice were determined, respectively, and the results are shown in fig. 2 and 5.
(5) From day 6, measuring the length/width of subcutaneous tumor with vernier caliper every day, and counting the growth trend of the subcutaneous tumor of mice with different models respectively, wherein the tumor volume calculation method comprises the following steps: length x width/2 mm 3 The results are shown in fig. 3 and 6.
(6) Mice were euthanized, tumor tissue from the mice was dissected with tissue forceps, placed on an analytical balance and the tumor mass was weighed and the results are shown in fig. 4 and 7.
As shown in fig. 2-4 and fig. 5-7, compound 54 and control compound PLX3397 have no obvious toxicity in the mouse tumor models of tumor immune models M-NFS-60 and MC38, and show good tumor inhibition effect on mice, and are equivalent to the tumor inhibition effect of control compound PLX 3397. Experimental results indicate that compound 54 of the invention is capable of dose-dependently inhibiting proliferation of tumors in M-NFS-60 mouse transplant tumor model as well as MC38 mouse transplant tumor model, demonstrating that inhibition of CSF1R kinase can be used to treat tumors activated by CSF1R signaling pathway. Furthermore, the literature shows that MC38 is an immune model, and inhibition of CSF1R kinase can transform M1 type macrophages, which are beneficial for tumor proliferation, to M2 type macrophages, which kill tumors in vivo, and this example also demonstrates that inhibition of CSF1R can inhibit tumor proliferation in an MC38 immune model.
The present invention provides a class of selective CSF1R inhibitors that selectively inhibit CSF1R kinase and related signaling pathway activity, and thus are capable of activating autoimmunity by inhibiting phosphorylation of CSF1R for use in the treatment of cancer. Thus, the present invention is suitable for industrial applications.
Although the present invention has been described in detail herein, the present invention is not limited thereto, and modifications may be made by those skilled in the art in light of the present principles, and it is therefore intended that all such modifications as fall within the scope of the present invention.

Claims (11)

1. A compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof:
wherein, the liquid crystal display device comprises a liquid crystal display device,
wherein L is selected from
Ar is selected from optionally 1 to 3 independently R 2 Aryl and heteroaryl groups substituted with groups;
R 1 selected from the group consisting of C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 heterocycloalkyl, hydroxyC 1-C6 alkyl, nitriloC 1-C6 alkyl, carboxyC 1-C6 alkyl, C3-C6 cycloalkylC 1-C6 alkyl, C1-C6 heterocycloalkylC 1-C6 alkyl, C1-C6 alkoxyC 1-C6 alkyl, C1-C6 alkylaminoC 1-C6 alkyl, C1-C6 alkylsulfonyl, C1-C6 alkanoyl, C1-C6 heterocycloalkylaminoacyl, C1-C6 alkylaminoacyl, C3-C6 cycloalkylaminoacyl, C1-C6 alkoxycarbonyl, aryl, aryloxycarbonyl, and heteroaryl, wherein aryl, heteroaryl and heterocycloalkylare optionally substituted with 1R 2 Group substitution;
each R is 2 Independently selected from halogen, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, halogenated C1-C6 alkoxy, and nitrogen protecting groups.
2. According to the weightsA compound of formula (I) according to claim 1, wherein L is
3. A compound of formula (I) according to claim 1 or 2, or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof, wherein Ar is selected from the group consisting of optionally 1-2 independently R 2 Phenyl, pyridyl, furyl, benzofuryl, thienyl, and pyrrolyl groups substituted with groups; optionally, each R 2 Independently selected from halogen, C1-C3 alkyl, C1-C3 alkoxy, C1-C3 haloalkyl, halogenated C1-C3 alkoxy, pivaloyl, t-butoxycarbonyl, benzyloxycarbonyl, 9-fluorenylmethoxycarbonyl, benzyl, p-methoxybenzyl, allyloxycarbonyl, and trifluoroacetyl.
4. A compound of formula (I) according to claim 3, or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof, wherein Ar is selected from the group consisting of 1R 2 Phenyl or pyridyl substituted with a group; optionally Ar is selected from para-position quilt R 2 Phenyl or pyridin-2-yl substituted with a group; optionally R 2 Selected from C1-C6 haloalkyl.
5. A compound of formula (I) according to claim 1 or 2, or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof, wherein R 1 Selected from the group consisting of C1-C3 alkyl, C1-C3 haloalkyl, C3-C5 heterocycloalkyl, hydroxyC 1-C3 alkyl, nitriloC 1-C3 alkyl, carboxyC 1-C3 alkyl, C3-C5 cycloalkylC 1-C3 alkyl, C1-C3 alkoxyC 1-C3 alkyl, C1-C4 alkylaminoC 1-C3 alkyl, C1-C3 alkylsulfonyl, C2-C4 alkanoyl, C3-C5 heterocycloalkylaminoacyl, hydroxyC 1-C3 alkylamineacyl, C1-C4 alkylamineaminoacyl, C3-C6 cycloalkylaminoacyl, C1-C3 alkoxycarbonyl, phenyl, phenoxyCarbonyl, thienyl and pyridyl, wherein phenyl and heterocycloalkyl are optionally substituted with methyl, halogen or t-butoxycarbonyl.
6. A compound of formula (I) according to claim 1 or 2, or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof, comprising the following compounds, or pharmaceutically acceptable salts, solvates, esters, acids, metabolites or prodrugs thereof:
7. a pharmaceutical composition comprising a compound of formula (I) as defined in any one of claims 1 to 6 or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof, and a pharmaceutically acceptable carrier or excipient, and optionally other therapeutic agents.
8. Use of a compound of formula (I), or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof, as claimed in any one of claims 1 to 6 in the manufacture of a medicament for inhibiting CSF1R kinase and associated signal pathway activity, or for treating cancer or tumour associated with CSF1R kinase activity.
9. Use of a compound of formula (I) as defined in any one of claims 1 to 6 or a pharmaceutically acceptable salt, solvate, ester, acid, metabolite or prodrug thereof in the manufacture of a medicament for the treatment of M2-type macrophage based cancer or tumour in a tumour microenvironment.
10. The use of claim 8 or 9, wherein the cancer or tumor is selected from one or more of ovarian cancer, cervical cancer, membranous adenocarcinoma, prostate cancer, bladder cancer, lung cancer, thyroid cancer, breast cancer, pancreatic cancer, renal cancer, gastric cancer, liver cancer, cervical cancer, endometrial cancer, colorectal cancer, nasopharyngeal cancer, esophageal cancer, cholangiocarcinoma, bone metastatic cancer, papillary thyroid cancer, non-small cell lung cancer, colon cancer, solid tumor, brain tumor, lymphoma, glioma, glioblastoma, melanoma, mesothelioma, glioblastoma, osteosarcoma, gastrointestinal stromal tumor, multiple myeloma, biliary tract carcinoma sarcoma, and leukemia.
11. The use of claim 10, wherein the lymphoma comprises non-hodgkin's lymphoma, B-cell or T-cell lymphoma; the leukemia includes chronic myelogenous leukemia or acute myelogenous leukemia.
CN202210262929.6A 2022-03-17 2022-03-17 Selective CSF1R inhibitor and application thereof Pending CN116789670A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202210262929.6A CN116789670A (en) 2022-03-17 2022-03-17 Selective CSF1R inhibitor and application thereof
PCT/CN2022/083651 WO2023173480A1 (en) 2022-03-17 2022-03-29 Selective csf1r inhibitor and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210262929.6A CN116789670A (en) 2022-03-17 2022-03-17 Selective CSF1R inhibitor and application thereof

Publications (1)

Publication Number Publication Date
CN116789670A true CN116789670A (en) 2023-09-22

Family

ID=88022047

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210262929.6A Pending CN116789670A (en) 2022-03-17 2022-03-17 Selective CSF1R inhibitor and application thereof

Country Status (2)

Country Link
CN (1) CN116789670A (en)
WO (1) WO2023173480A1 (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW200740820A (en) * 2005-07-05 2007-11-01 Takeda Pharmaceuticals Co Fused heterocyclic derivatives and use thereof
PL1940844T3 (en) * 2005-10-28 2010-03-31 Irm Llc Compounds and compositions as protein kinase inhibitors
CN102827186A (en) * 2011-06-16 2012-12-19 中国科学院上海药物研究所 Pyridino five-membered heterocyclic derivative as well as preparation method and applications thereof

Also Published As

Publication number Publication date
WO2023173480A1 (en) 2023-09-21

Similar Documents

Publication Publication Date Title
JP6791979B2 (en) Nitrogen-containing heterocyclic compounds, production methods, intermediates, compositions and applications
CN106928231B (en) Novel EGFR wild type and mutant kinase inhibitors
US8501755B2 (en) Tetrahydropyridothienopyrimidine compounds and methods of use thereof
RU2633694C2 (en) Dyetherned phenylaminopyrimidine and pharmaceutical composition containing such connection
CN103588792A (en) Pyrazolopyrimidine or pyrimidinopyrimidine compounds, preparation method therefor, pharmaceutical compositions thereof and applications thereof
US20220017520A1 (en) Macrocyclic compound as cdk inhibitor, preparation method therefor, and use thereof in medicine
CN109970717B (en) 4- (alicyclic pyrimidine/pyridine substituted) amino-1H-3-pyrazolecarboxamide FLT3 inhibitor and application thereof
CN116283953B (en) Indoline compound containing thiazole structure, and preparation method and application thereof
CN109810100A (en) The bis- target spot inhibitor of PARP-1 and PI3K containing benzofuran
US11572359B2 (en) PARP/PI3K double-target inhibit containing pyridopyrimidine structure
CN110467637B (en) Bisaminyl chloropyrimidine compound containing phosphine oxide substituted aniline, preparation method and application thereof
CN113105434A (en) Novel CDK4/6 inhibitor and preparation method and application thereof
CN111825719A (en) Arylamine-substituted pyrrolopyrimidine compound, and preparation method and application thereof
CN116789670A (en) Selective CSF1R inhibitor and application thereof
JP2022521964A (en) New pan-RAF kinase inhibitor and its use
CN113135898B (en) Anticancer quinoxaline pyrimidine amine heterocyclic compound and preparation method and application thereof
CN111662271B (en) Compound with IDH mutant inhibitory activity and preparation method and application thereof
CN110577546B (en) VEGFR inhibitor and preparation method and application thereof
CN114751899A (en) Diaryl urea mTOR kinase inhibitor and pharmaceutical composition and application thereof
CN110283160B (en) PDGFR kinase inhibitor
JP2023536634A (en) Small molecules for the treatment of autoimmune diseases and cancer
TW201738227A (en) Fused ring compound and preparation method, application and intermediate compound thereof
CN109942544B (en) Novel indazole derivative kinase inhibitor
CN111303024B (en) Quinoline-structured pan-KIT kinase inhibitor and application thereof
US11021479B2 (en) Pyridoquinazoline derivatives useful as protein kinase inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication