CN107793413B - Pyrimidine heterocyclic compound and preparation method and application thereof - Google Patents

Pyrimidine heterocyclic compound and preparation method and application thereof Download PDF

Info

Publication number
CN107793413B
CN107793413B CN201610803436.3A CN201610803436A CN107793413B CN 107793413 B CN107793413 B CN 107793413B CN 201610803436 A CN201610803436 A CN 201610803436A CN 107793413 B CN107793413 B CN 107793413B
Authority
CN
China
Prior art keywords
methyl
compound
reaction
added
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201610803436.3A
Other languages
Chinese (zh)
Other versions
CN107793413A (en
Inventor
田红旗
黄功超
程瑛
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.)
KECHOW PHARMA, Inc.
Original Assignee
KECHOW PHARMA Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by KECHOW PHARMA Inc filed Critical KECHOW PHARMA Inc
Priority to CN201610803436.3A priority Critical patent/CN107793413B/en
Publication of CN107793413A publication Critical patent/CN107793413A/en
Application granted granted Critical
Publication of CN107793413B publication Critical patent/CN107793413B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings

Abstract

The present invention relates to compounds of formula (I) and pharmaceutically acceptable salts, prodrugs and solvates thereof, which are useful for treating cancer and inflammation in a mammal. The invention also discloses a preparation method of the compound shown in the formula (I) and a pharmaceutical composition containing the compound.

Description

Pyrimidine heterocyclic compound and preparation method and application thereof
Technical Field
The present invention relates to certain fluoroheterocyclic compounds and pharmaceutically acceptable salts thereof, which are useful for the treatment and prevention of diseases or conditions mediated by certain variant forms of epidermal growth factor receptor (e.g., L858R activation mutant, Exon19 deletion activation mutant, and T790M resistance mutant). Such compounds and their salts are useful in the treatment or prevention of many different cancers. The invention is also directed to pharmaceutical compositions comprising the compounds and salts thereof, to intermediates in the preparation of the compounds, and to methods of using the compounds and salts thereof to treat various forms of EGFR-mediated diseases.
Background
EGFR is a member of the erbB receptor family of transmembrane protein tyrosine kinases. When bound to a growth factor ligand, such as Epidermal Growth Factor (EGF), the receptor may homodimerize with additional EGFR molecules or heterodimerize with another family member, such as erbB2(HER2), erbB3(HER3), or erbB4(HER 4).
Homodimerization and/or heterodimerization of erbB receptors results in phosphorylation of key tyrosine residues in the intracellular domain and in stimulation of many intracellular signaling pathways involved in cell proliferation and survival. Dysregulation of erbB family signaling promotes proliferation, invasion, metastasis, angiogenesis, and tumor cell survival, and has been described in many human cancers, including lung, head and neck, and breast cancers.
Thus, the erbB family represents a reasonable target for the development of anticancer drugs, and many agents targeting EGFR or erbB2 are now clinically available, including gefitinib (IRESSA)TM) Erlotinib (TARCEVA)TM) Lapatinib (TYKERB)TM,TYVERBTM). A detailed discussion of erbB receptor signaling and its involvement in tumorigenesis is provided in New England Journal of medicine (2008) stage 358, 1160-74 and Bi deg.C chemical and biological Research communication (2004) Vol.319, 1-11.
Activating mutations of EGFR in non-small cell lung cancer (NSCLC) reported in 2004 (Science [2004] stage 304, 1497-500 and New England Journal of medicine [2004] stage 350, 2129-39) were associated with response to gefitinib treatment. The most common EGFR activating mutations (L858R and del 746_ a750) result in increased affinity for small molecule tyrosine kinase inhibitors (such as gefitinib and erlotinib) and decreased affinity for adenosine monophosphate (ATP) relative to wild-type (WT) EGFR. Finally, acquired resistance to gefitinib or erlotinib treatment, such as the mutation at gatekeeper residue T790M, which is reported to be detected in 50% of clinically resistant patients, is not considered to sterically hinder gefitinib or erlotinib binding to EGFR, but only alters the affinity for ATP to levels comparable to WT EGFR.
Given the importance of such mutations in the resistance of existing therapies targeting EGFR. Drugs that can inhibit EGFR including a mutation in the gatekeeping gene are considered to be particularly useful in the treatment of cancer.
There remains a need for compounds that can exhibit advantageous potency properties for WT EGFR, and/or selectivity with respect to other enzyme receptors, which make these compounds particularly promising for development as therapeutic agents, relative to activating mutant forms of EGFR (e.g., L858R EGFR mutant, or del e746_ a750 mutant or Exon19 deletion EGFR mutant) and/or resistant mutant forms of EGFR (e.g., T790M EGFR mutant). In this regard, there is a need for compounds that exhibit high inhibition of certain activating or resistant mutant forms of EGFR while exhibiting relatively low inhibition of WT EGFR. Due to the reduced toxicology associated with inhibition of wild-type EGFR, it is expected that such compounds may be more suitable for use as therapeutic agents, particularly for cancer treatment. Such toxicology is known to manifest as rashes and/or diarrhea in humans.
The applicant has found that one or more fluoro-heterocyclic compounds have high potency relative to several EGFR mutant forms, while showing relatively low inhibition of WTEGFR. The compounds of the invention may also exhibit advantageous physical properties (e.g., higher aqueous solubility, permeability, and/or lower plasma protein binding) and/or advantageous toxicity profiles (e.g., reduced tendency to hERG block) and/or advantageous metabolic profiles compared to other known EGFR/EGFR mutant inhibitors. Thus, such compounds are particularly useful in the treatment of disease conditions that involve EGFR and/or activating mutations of EGFR and/or resistance mutations of EGFR, for example, in the treatment of cancer.
Disclosure of Invention
One aspect of the present invention provides compounds of formula (I) and pharmaceutically acceptable salts, prodrugs and solvates thereof,
Figure BDA0001109816280000021
wherein the content of the first and second substances,
R1selected from: hydrogen, deuterium, halogen, unsubstituted or substituted C1-6 alkyl, -NR6R7Nitro, cyano, -OR6、-C(O)OR6、-C(O)R6、-C(O)NR6R7、-S(O)t-R6、-S(O)2-NR6R7
R2Selected from: hydrogen, halogen, C1-6Alkyl, halogen substituted C1-6Alkyl, -S (O)t-R6、-OR13
R3Selected from: hydrogen, halogen, unsubstituted or substituted C1-6Alkyl, unsubstituted OR substituted aromatic OR non-aromatic heterocyclyl, unsubstituted OR substituted heterocycloalkyl, -OR6、-NR6R7、-C(O)R6、-S(O)t-R6
R4And R5Independently selected from: hydrogen, unsubstituted or substituted C1-6Alkyl, -C (O) R6、-S(O)t-R6(ii) a Or, R4And R3Together with the nitrogen atom to which they are attached and the phenyl ring form a substituted or unsubstituted aromatic or non-aromatic heterocyclic group;
t is 0, 1, 2;
R6and R7Independently selected from: hydrogen, substituted or unsubstituted C1-6Alkyl, substituted or unsubstituted C3-6Cycloalkyl, substituted or unsubstituted C1-6An alkenyl group, a substituted or unsubstituted aryl group, a substituted or unsubstituted aromatic or non-aromatic heterocyclic group, a substituted or unsubstituted alkoxy group, a substituted or unsubstituted aryloxy group, a substituted or unsubstituted amino group, a halogen;
R13selected from halogen substituted C 1-6An alkyl group;
g is selected from the following groups:
Figure BDA0001109816280000022
x is selected from carbon, nitrogen (CH, N);
R8selected from: hydrogen, deuterium, substituted or unsubstituted C1-6Alkyl, substituted or unsubstituted C3-6Cycloalkyl, -C (O) R6、-S(O)t-R6;
R9Selected from: hydrogen, halogen, substituted OR unsubstituted alkyl, -OR6、-S(O)t-R6
Preferably, R8Selected from: hydrogen, deuterium, substituted or unsubstituted C1-3Alkyl, substituted or unsubstituted C3-5Cycloalkyl, -S (O)2-R6,R6Selected from: substituted or unsubstituted C1-6Alkyl, substituted or unsubstituted C3-6Cycloalkyl groups of (a); more preferably, R8Selected from: hydrogen, halogen substituted or unsubstituted C1-3Alkyl (e.g., methyl, ethyl, isopropyl, n-propyl), -S (O)2-R6,R6Selected from: substituted or unsubstituted C1-3Alkyl, substitutedOr unsubstituted cyclopropyl; further preferably, R8Selected from: -CH3、-CH2CF3Cyclopropane, isopropyl, methylsulfonyl;
preferably, R9Selected from: hydrogen, halogen, substituted or unsubstituted C1-6Alkyl, -OR of6、-S(O)2-R6,R6Is substituted or unsubstituted C1-6Alkyl groups of (a); more preferably, R9Selected from: hydrogen, halogen substituted or unsubstituted C1-3Alkyl (e.g., methyl, ethyl, isopropyl, n-propyl); further preferably, R9Selected from: hydrogen, -CH3、-CF3、-CH2CF3
In one embodiment of the invention, R9Is hydrogen;
More preferably, G is selected from:
Figure BDA0001109816280000031
the compounds of formula (I) herein have the general formula:
Figure BDA0001109816280000032
in a preferred embodiment of the invention, G is
Figure BDA0001109816280000033
R8Selected from: -CH3、-CH2CF3Cyclopropane, isopropyl, methylsulfonyl; preferably selected from: -CH3、-CH2CF3
In a preferred embodiment of the invention, in said compound, R3Selected from: -C (O) R6、-S(O)-R6、-S(O)2-R6(ii) a Or, R4And R3Together with the nitrogen atom to which they are attached and the phenyl ring form a substituted or unsubstituted aromatic or non-aromatic heterocyclic group;
preferably, R6Selected from: c1-3Alkyl, halogen substituted C1-3Alkyl group of (i), (ii), (iii), (iv) and (iv)2)PNR10R11、-NR12(CH2)PNR10R11、-NR10R11Wherein p is selected from an integer of 1 to 6, R10、R11、R12Independently selected from H, C1-3Alkyl groups of (a); more preferably, R6Selected from: -CH3、-CF3、-CH2F、-CHF2、-N(CH3)CH2CH2N(CH3)2、-CH2CH2N(CH3)2、-N(CH3)2
Preferably, R7Selected from: H. c1-3Alkyl, halogen substituted C1-3Alkyl groups of (a);
in a preferred embodiment of the present invention, in the compound, R3Selected from: -C (O) NR6R7、-SOR6、-SO2R6,R2Selected from: hydrogen, deuterium, halogen, -OR13Halogen-substituted C1-6Alkyl, -S (O)t-R6
In another preferred embodiment of the present invention, in the compound, R3Selected from: c1-3Alkyl, -OR of13、-SR6,R2Selected from: hydrogen, deuterium, halogen substituted C1-6Alkyl, -S (O)t-R6
Preferably, R1Selected from: hydrogen, deuterium, halogen, fluorine substituted or unsubstituted C1-3Alkyl, nitro, cyano; further preferably, R 1Selected from: hydrogen, deuterium, halogen, cyano, methyl, trifluoromethyl; even more preferably, R1Selected from: hydrogen, chloro, cyano, trifluoromethyl; most preferably, R1Selected from: hydrogen, chlorine;
in another preferred embodiment of the invention, R4And R5Independently selected from: hydrogen, unsubstituted or substituted C1-3Alkyl, -C (O) R6、-S(O)2-R6,R6Selected from: hydrogen and oxygen generationSubstituted or unsubstituted C1-3Alkyl, substituted or unsubstituted C3-6Cycloalkyl, substituted or unsubstituted C1-3An alkenyl group; more preferably, R4And R5Independently selected from: hydrogen, methyl, -C (O) R6、-S(O)2-R6,R6Selected from: vinyl, methylvinyl, 2- (dimethylamino) ethyl, 2- (diethylamino) ethyl, cyclopropyl;
in one embodiment of the invention, R4Is hydrogen, R5Selected from: acryloyl, 3- (dimethylamino) propionyl, cyclopropaneformyl;
in another preferred embodiment of the invention, R4And R3Together with the nitrogen atom to which they are attached and the phenyl ring form a substituted or unsubstituted aromatic or non-aromatic heterocyclic group, said compound having the general structure:
Figure BDA0001109816280000034
n is 1 or 2;
R6selected from: hydrogen, substituted or unsubstituted C1-6Alkyl groups of (a); preferably selected from: hydrogen, substituted or unsubstituted C1-3Alkyl groups of (a);
In a preferred embodiment of the invention, R6Is 2- (dimethylamino) ethyl;
preferably, the compound has the structure of formula II-1;
in some preferred embodiments of the invention, the compound of formula i is selected from:
Figure BDA0001109816280000041
Figure BDA0001109816280000051
Figure BDA0001109816280000061
Figure BDA0001109816280000071
Figure BDA0001109816280000081
Figure BDA0001109816280000091
Figure BDA0001109816280000101
in some preferred embodiments of the invention, the compound of formula i is selected from:
Figure BDA0001109816280000111
the above groups, in addition to oxygen and halogen, may be linked to each other to form a cycloalkyl or heterocycloalkyl group, which may be optionally substituted by substituents selected from: -ORa, -NRaRb, C optionally substituted by-ORa1-5An alkyl group;
ra and Rb may be the same or different and are each independently selected from hydrogen and C1-5An alkyl group which may be optionally substituted with one to three substituents selected from: hydroxy, C1-5Alkoxy, halogen, amino.
Another aspect of the invention provides a process for the preparation of a compound of formula (I), preferably, such as:
Figure BDA0001109816280000112
wherein R is1,R2,R3,R4As defined above. The base is organic base including triethylamine, pyridine, diisopropylethylamine, etc. or inorganic base including potassium carbonate, sodium carbonate, cesium carbonate, sodium hydride, etc.
Yet another aspect of the present invention provides pharmaceutical compositions comprising a compound of formula (I) or pharmaceutically acceptable salts, prodrugs and solvates thereof.
In a further aspect of the present invention, there is provided a use of a compound of formula (I) or a pharmaceutically acceptable salt, prodrug and solvate thereof, and a pharmaceutical composition thereof for the manufacture of a medicament for preventing and/or treating tumors, acute and chronic inflammatory diseases, inflammatory bowel diseases, skin diseases, diabetes, ocular diseases, diseases associated with angiogenesis or revascularization in mammals, and diseases associated with chronic pain.
A further aspect of the present invention provides the use of a compound of formula (I) or a pharmaceutically acceptable salt, prodrug and solvate thereof, and pharmaceutical compositions thereof, in the manufacture of a medicament for the treatment and/or prevention of a disorder or disease mediated by activated or resistant mutant forms of EGRF, preferably ovarian cancer, cervical cancer, colorectal cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, melanoma, prostate cancer, leukemia, lymphoma, non-hodgkin lymphoma, gastric cancer, lung cancer, hepatocellular carcinoma, gastrointestinal stromal tumor, thyroid cancer, cholangiocarcinoma, endometrial cancer, renal cancer, anaplastic large cell lymphoma, acute myelogenous leukemia, multiple myeloma or mesothelioma.
The compound of the invention has better physical and chemical properties and safe toxicity parameters, and can be used for treating cancer and inflammation of mammals.
Detailed Description
The present invention is defined throughout the disclosure with the following terms, if not otherwise indicated:
the term "prodrug" refers to any derivative that can be converted in vivo to the corresponding active drug compound. In one embodiment, when a compound of the present invention contains a hydroxyl group, a prodrug thereof may be an ester thereof with a suitable acid, including, for example, lactic acid, citric acid, ascorbic acid, and the like.
The term "pharmaceutically acceptable salt", unless otherwise specified, includes salts of acidic groups (such as, but not limited to, potassium, sodium, magnesium, calcium, and the like) or salts of basic groups (such as, but not limited to, sulfate, hydrochloride, phosphate, nitrate, carbonate, and the like) that may be present in the compounds of the invention.
The term "solvate" refers to a complex molecular compound formed by solute molecules or ions attracting adjacent solvent molecules in solution through intermolecular forces such as coulomb forces, van der waals forces, charge transfer forces, hydrogen bonding, and the like. In one embodiment, the solvent is water, i.e. the compound of the invention forms a hydrate.
The alkyl, alkenyl, alkynyl, cycloalkyl moieties may each independently be optionally substituted with one or more groups selected from: hydroxyl, oxo, halogen, cyano, nitro, trifluoromethyl, azido, amino, carboxyl, mercapto.
Saturated or unsaturated hydrocarbon radicals, such as alkyl, alkanediyl or alkenyl, including combinations with heteroatoms, such as alkoxy, can each be linear or branched, respectively.
Depending on the substituents, the compounds of the formula (I) may, depending on the substituents, be present as optical isomers or as mixtures of isomers of different composition, which mixtures, if appropriate, may be separated off in a customary manner. The present invention provides pure isomers and isomer mixtures, processes for their preparation and their use, as well as compositions comprising them. For the sake of simplicity, this is referred to hereinafter as the compound of the formula (I), which refers both to the pure optical isomers and, where appropriate, to mixtures of isomers in different proportions.
Synthesis of
Suitable solvents commonly used in organic reactions can be used in the following steps of the preparation process of the present invention, such as, but not limited to: aliphatic and aromatic, optionally hydrocarbon or halogenated hydrocarbon (e.g., pentane, hexane, heptane, cyclohexane, petroleum ether, gasoline, volatile oil, benzene, toluene, xylene, methylene chloride, dichloroethane, chloroform, carbon tetrachloride, chlorobenzene and o-dichlorobenzene), aliphatic and aromatic, optionally alcohol (e.g., methanol, ethanol, propanol, isopropanol, t-butanol, ethylene glycol, etc.), ether (e.g., diethyl ether and dibutyl ether, ethylene glycol dimethyl ether and diglyme, tetrahydrofuran and dioxane, etc.), ester (e.g., methyl acetate or ethyl acetate, etc.), nitrile (e.g., acetonitrile or propionitrile, etc.), ketone (e.g., acetone, butanone, etc.), amide (e.g., dimethylformamide, dimethylacetamide, N-methylpyrrolidone, etc.), and dimethyl sulfoxide, tetramethylene sulfone and hexamethylphosphoric triamide and N, N-dimethylpropylene urea (DMPU) and the like.
Synthesis examples:
example 1: synthesis of 2-acrylamido-N- (2- (dimethylamino) ethyl) -5-methoxy-N-methyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) aniline
Figure BDA0001109816280000121
Step 1: synthesis of methyl 5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzoate:
the compound 3- (2-chloropyrimidin-4-yl) -1-methylindole (2.4g,9.85mmol) and methyl 4-amino-5-methoxy-2-nitrobenzoate (2.67g,11.82mmol) were charged into a reaction flask, followed by addition of 1, 4-dioxane (20mL), addition of p-toluenesulfonic acid (2.03g,11.82mmol) with stirring, heating to 85 ℃ for reaction for 48 hours, after completion of the reaction, cooling to room temperature to precipitate a solid, filtration, washing of the cake with acetonitrile, and drying to give a yellow solid (4.0g, yield: 93.7%).1H NMR(400MHz,CDCl3):δ9.66(s,1H),8.44(d,J=4Hz,1H),8.21-8.19(m,1H), 8.17(s,1H),7.94(s,1H),7.44-7.41(m,2H),7.28(s,1H),7.17(s,1H),4.06(s,3H),3.95(s,3H),3.93(s,3H).
Step 2: synthesis of 5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzoic acid:
the compound 5-methoxy-4- ((4-(1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzoic acid methyl ester (1.0g,2.3mmol) was added to a reaction flask, then methanol (10mL) was added to dissolve it, sodium hydroxide (1M,4.62mL,4.62mmol) was added with stirring, heated to 75 ℃ for 8 hours, after the reaction was complete, cooled to room temperature, adjusted to pH 4 with 10% hydrochloric acid, filtered, the filter cake was washed with methanol, dried to give a yellow solid (0.86g, yield: 88.9%). 1H NMR(400MHz,DMSO-d6):δ9.10(s,1H),8.46-8.43(m,4H),7.57-7.55(d,J=8Hz,1H),7.41-7.39(m,2H),7.32-7.28(m,1H),7.22-7.19(m,1H),4.07(s,3H),3.90(s,3H).
And step 3: synthesis of N- (2- (dimethylamino) ethyl) -5-methoxy-N-methyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzamide:
the compound 5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzoic acid (850mg,2.03mmol) was added to a reaction flask, after DCM (10mL) was added and dissolved, HOBT (411mg,3.04mmol) and EDCI (583mg,3.04mmol) were added, and after 30 minutes of reaction N was added1,N1-dimethylethane-1, 2-diamine (310mg,3.04mmol), stirred at room temperature for reaction for 1 hour, after completion of the reaction, the reaction solution was diluted with saturated brine, extracted with dichloromethane, the organic phase was washed with saturated brine three times, dried over anhydrous sodium sulfate, and concentrated to give a crude product which was used directly in the next step (900mg, yield: 88.27%).1H NMR(400MHz,MeOD-d4):δ9.74(s,1H),8.38(m,1H),8.24(m,2H),7.48(s,1H),7.29-7.25(m,3H),7.15(s,1H),4.11(s,3H),3.92(s,3H),3.80(t,2H),2.96(s,3H),2.87(t,2H),2.51(s,6H).
And 4, step 4: synthesis of 2-amino-N- (2- (dimethylamino) ethyl) -5-methoxy-N-methyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzamide:
the compound N- (2- (dimethylamino) ethyl) -5-methoxy-N-methyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzamide (500mg,0.99mmol), iron powder (305mg,5.46mmol) and ammonium chloride (425mg,7.94mmol) were added to a reaction flask, followed by EtOH/H 2O-4/1 (10mL) under reflux for 2 hours, cooled, filtered, extracted with dichloromethane, dried over anhydrous sodium sulfate,concentration gave the objective compound (400mg, yield: 85.07%).1H NMR(400MHz,CDCl3)δ8.47(d,J=8.8Hz,1H),8.34(d,J=4.8Hz,1H),8.24(m,2H),7.78(s,1H),7.37(m,2H),7.05(d,J=4.8Hz,1H),6.73(s,1H),4.11(s,3H),4.08(s,3H),3.87(m,2H),3.10(s,3H),2.70(s,2H),2.60(s,2H),2.38(s,6H)。
And 5: synthesis of 2-acrylamido-N- (2- (dimethylamino) ethyl) -5-methoxy-N-methyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) aniline
A solution of acryloyl chloride (73mg,0.80mmol) in dichloromethane was added dropwise to a stirred mixture of 2-amino-N- (2- (dimethylamino) ethyl) -5-methoxy-N-methyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzamide (380mg,0.80mmol) and DIPEA (114mg,0.88mmol) in dichloromethane (2mL) cooled in an ice-water bath. After the reaction for 1.5 hours, it was diluted with dichloromethane, washed with saturated sodium bicarbonate, the aqueous phase was extracted twice with dichloromethane, and the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give the objective compound (250mg, yield: 59.05%).1H NMR(400MHz,CDCl3)δ9.60(s,1H),9.27(s,1H),8.77(s,1H),8.40(d,J=5.3Hz,1H),8.10(d,J=7.4Hz,1H),7.88(s,1H),7.38(s,1H),7.28(d,J=3.0Hz,2H),7.23(d,J=5.3Hz,1H),6.95(s,1H),6.40(s,2H),5.74(d,J=9.4Hz,1H),3.95(d,J=8.9Hz,6H),3.86–3.74(m,2H),3.14(s,3H),2.91(m,2H),2.59(s,6H).MS(ESI)m/z 528.62[M+H].
Example 2: synthesis of N- (2- (dimethylamino) ethyl) -2- (3- (dimethylamino) propionylamino) -5-methoxy-N-methyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzamide
Figure BDA0001109816280000131
The compound 2-acrylamido-N- (2- (dimethylamino) ethyl) -5-methoxy-N-methyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) aniline prepared in example 1 (200mg,0.38mmol) was added to a reaction flask, followed by dissolution in ethanol (5mL), and 30% dimethylaminoethanol was added with stirring The reaction mixture (2mL) was stirred at room temperature for 30 minutes, and after completion of the TLC detection, the reaction mixture was concentrated and purified by TLC to obtain the objective compound as an off-white solid (100mg, yield: 46.06%).1H NMR(400MHz,CDCl3)δ10.27(s,1H),9.16(s,1H),8.39(d,J=5.3Hz,2H),8.21(s,1H),7.80(s,1H),7.42–7.37(m,1H),7.31(dd,J=8.9,5.2Hz,2H),7.17(d,J=5.3Hz,1H),6.77(s,1H),3.94(s,3H),3.87(s,3H),3.10(s,3H),2.81(m,6H),2.55(s,6H),2.04(m,8H).MS(ESI)m/z573.32[M+H].
Example 3: synthesis of N- (5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (2,2, 2-trifluoroacetyl) -4- (trifluoromethoxy) phenyl) acrylamide:
Figure BDA0001109816280000141
step 1: synthesis of 2,2, 2-trifluoro-1- (4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) oxy) -2-nitro-5- (trifluoromethoxy) phenyl) ethanone:
the compounds 3- (2-chloropyrimidin-4-yl) -1-methylindole (1.5g,6.16mmol) and 1- (4-amino-2-nitro-5- (trifluoromethoxy) phenyl) -2,2, 2-trifluoroacetone (1.96g,6.16mmol) were added to a reaction flask, followed by 2-pentanol (100mL), p-toluenesulphonic acid monohydrate (1.27g,7.39mmol) was added with stirring, heated to 105 ℃ for 2.5 hours, after completion of the reaction, cooled to room temperature to precipitate a solid, filtered, the filter cake was washed with acetonitrile, and dried to give a yellow solid (2.3g, yield: 71%).1H NMR(400MHz,CDCl3):δ9.46(s,1H),8.56(d,1H),8.17(m,1H),7.59(m,1H),7.56(s,1H),7.50(s,1H),7.42-7.41(m,2H),7.19(s,1H),3.69(s,3H).
Step 2: synthesis of 1- (2-amino-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -5- (trifluoromethoxy) phenyl) -2,2, 2-trifluoroacetone:
the compound 2,2, 2-trifluoro-1- (4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) oxy) -2-nitro-5- (trifluoromethoxy) phenyl) ethanone (500mg,0.95mmol), iron powder (293mg,5.23mmol) and ammonium chloride (407mg,7.61mmol) were added to a reaction flask, followed by EtOH/H 2O-4/1 (5mL) solutionThe reaction mixture was refluxed for 2 hours, cooled, filtered, extracted with dichloromethane, dried over anhydrous sodium sulfate, and concentrated to obtain the objective compound (400mg, yield: 85%).1H NMR(400MHz,CDCl3):δ8.53(s,1H),8.32(d,1H),8.17(m,1H),7.59(m,1H),7.42-7.41(m,2H),7.24(d,1H),7.19(s,1H),6.99(s,1H),5.77(s,1H),4.57(br s,2H),3.69(s,3H).
And step 3: synthesis of N- (5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (2,2, 2-trifluoroacetyl) -4- (trifluoromethoxy) phenyl) acrylamide:
a solution of acryloyl chloride (19mg,0.20mmol) in dichloromethane was added dropwise to a stirred mixture of 1- (2-amino-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -5- (trifluoromethoxy) phenyl) -2,2, 2-trifluoroacetone (100mg,0.20mmol) and DIPEA (65mg,0.50mmol) in dichloromethane (2mL) cooled in an ice-water bath. After 1.5 hours of the reaction, it was diluted with dichloromethane, washed with saturated sodium bicarbonate, the aqueous phase was extracted twice with dichloromethane, and the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give the objective compound (55mg, yield: 50%).1H NMR(400MHz,CDCl3):δ9.07(s,1H),8.81(s,1H),8.58(d,1H),8.17(m,1H),7.59(m,1H),7.42-7.41(m,2H),7.24(d,1H),7.22(s,1H),7.19(s,1H),6.95(s,1H,),6.48(m,1H),6.01(m,1H),5.51(m,1H),3.69(s,3H).MS(ESI)m/z 550.42[M+H].
Example 4 Synthesis of 2-acrylamido-N- (2- (dimethylamino) ethyl) -4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -5- (trifluoromethoxy) benzamide:
Figure BDA0001109816280000142
step 1: synthesis of methyl 4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitro-5- (trifluoromethoxy) benzoate:
The compound 3- (2-chloropyrimidin-4-yl) -1-methylindole (500mg,2.05mmol) and methyl 4-amino-2-nitro-5- (trifluoromethoxy) benzoate (575mg,2.05mmol) were added to a reaction flask, followed by 2-pentanol (10mL), and p-methyl was added with stirringPhenylbenzenesulfonic acid monohydrate (424mg,2.46mmol), was heated to 105 ℃ to react for 2.5 hours, after the reaction was completed, cooled to room temperature to precipitate a solid, which was filtered, and the filter cake was washed with acetonitrile and dried to give a yellow solid (650mg, yield: 65%).1H NMR(400MHz,CDCl3):δ9.46(s,1H),8.43(d,1H),8.17(m,1H),7.61(s,1H),7.59(m,1H),7.56(s,1H),7.42-7.41(m,2H),7.24(d,1H),7.19(s,1H),3.98(s,3H),3.69(s,3H).
Step 2: synthesis of 4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitro-5- (trifluoromethoxy) benzoic acid:
methyl 4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitro-5- (trifluoromethoxy) benzoate (600mg,1.23mmol) was added to a reaction flask, THF/MeOH ═ 4/1(10mL) was added, then an aqueous solution of 1M lithium hydroxide (2.46mL,2.46mmol) was added, and the mixture was stirred at room temperature for 4 hours, after completion of the reaction, pH was adjusted to 3 to 4 with 1M hydrochloric acid, extraction was performed with ethyl acetate, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to obtain the objective compound (580mg, yield: 99.5%). MS (ESI) M/z 474.10[ M + H ]
And step 3: synthesis of N- (2- (dimethylamino) ethyl) -4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitro-5- (trifluoromethoxy) benzamide:
The compound 4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitro-5- (trifluoromethoxy) benzoic acid (200mg, 0.42mmol) was added to a reaction flask, after dissolution in DMF (5mL), HOBT (86mg,0.63mmol) and EDCI (121mg,0.63mmol) were added, and after 30 minutes of reaction N was added1,N1-dimethylethane-1, 2-diamine (56mg,0.63mmol), stirred at room temperature for reaction for 1 hour, after completion of the reaction, the reaction mixture was poured into water, extracted with ethyl acetate, the organic phase was washed with saturated brine three times, dried over anhydrous sodium sulfate, concentrated, and subjected to column chromatography to give the objective compound (180mg, yield: 78.4%).1H NMR(400MHz,CDCl3):δ9.46(s,1H),8.43(d,1H),8.17(m,1H),8.03(s,1H),7.63(s,1H),7.59(m,2H),7.42-7.41(m,2H),7.24(d,1H),7.19(s,1H),3.69(s,3H),3.59(m,2H),2.57(m,2H),2.26(s,6H).
And 4, step 4: synthesis of 2-amino-N- (2- (dimethylamino) ethyl) -4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -5- (trifluoromethoxy) benzamide:
the compound N- (2- (dimethylamino) ethyl) -4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitro-5- (trifluoromethoxy) benzamide (160mg,0.29mmol) was added to a reaction flask and EtOH/H was added2O-4/1 (5mL), followed by Fe (91mg,1.62mmol) and NH4Cl (126mg,2.36 mmol). After the reaction was completed, it was cooled to room temperature, filtered, washed with ethanol, dried over anhydrous sodium sulfate, and concentrated to obtain the objective compound (130mg, yield: 86.0%). MS (ESI) M/z 514.51[ M + H ] ]
And 5: synthesis of 2-acrylamido-N- (2- (dimethylamino) ethyl) -4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -5- (trifluoromethoxy) benzamide:
a solution of acryloyl chloride (18mg, 0.19mmol) in dichloromethane was added dropwise to a stirred mixture of 2-amino-N- (2- (dimethylamino) ethyl) -4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -5- (trifluoromethoxy) benzamide (100mg, 0.19mmol) and DIPEA (31mg, 0.23mmol) in dichloromethane (2mL) cooled in an ice-water bath. After the reaction for 1.5 hours, it was diluted with dichloromethane, washed with saturated sodium bicarbonate, the aqueous phase was extracted twice with dichloromethane, and the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give the objective compound (55mg, yield: 49.7%).1H NMR(400MHz,CDCl3):δ10.12(s,1H),9.46(s,1H),8.43(d,1H),8.17(m,1H),8.03(s,1H),7.59(m,1H),7.42-7.41(m,2H),7.31(s,1H),7.24(d,1H),7.19(s,1H),7.02(s,1H),6.48(m,1H),6.01(m,1H),5.51(m,1H),3.69(s,3H),3.59(m,2H),2.57(m,2H),2.26(s,6H).MS(ESI)m/z:568.56[M+H].
Example 5 Synthesis of 2-acrylamido-N- (2- (dimethylamino) ethyl) -5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzamide:
Figure BDA0001109816280000151
step 1: synthesis of methyl 5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzoate:
the compound 3- (2-chloropyrimidin-4-yl) -1-methylindole (1.0g, 4.1mmol) and methyl 4-amino-5-methoxy-2-nitrobenzoate (928mg,4.1mmol) were charged into a reaction flask, followed by addition of 2-pentanol (10mL), addition of p-toluenesulfonic acid monohydrate (848mg,4.92mmol) with stirring, reaction to 105 ℃ for 2.5 hours, after completion of the reaction, cooling to room temperature, precipitation of a solid, filtration, cake washing with acetonitrile, and drying to give a yellow solid (1.5g, yield: 84.3%). 1H NMR(CDCl3):δ9.46(s,1H),8.43(d,1H),8.17(m,1H),7.61(s,1H),7.59(m,1H),7.56(s,1H),7.42-7.41(m,2H),7.24(d,1H),7.19(s,1H),3.98(s,3H),3.83(s,3H),3.69(s,3H).
Step 2: synthesis of 5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzoic acid:
the compound methyl 5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzoate (1.0g,2.31mmol) was added to a reaction flask, THF/MeOH (4/1) (20mL) was added, then a 1M aqueous solution of lithium hydroxide (4.62mL,4.62mmol) was added, the mixture was stirred at room temperature for 4 hours, after completion of the reaction, the pH was adjusted to 3 to 4 with 1M hydrochloric acid, extraction was performed with ethyl acetate, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to obtain the objective compound (900mg, yield: 93.0%).1H NMR(400MHz,DMSO-d6):δ9.10(s,1H),8.46-8.43(m,4H),7.57-7.55(d,J=8Hz,1H),7.41-7.39(m,2H),7.32-7.28(m,1H),7.22-7.19(m,1H),4.07(s,3H),3.90(s,3H).
And step 3: synthesis of N- (2- (dimethylamino) ethyl) -5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzamide:
the compound 5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzoic acid (200mg,0.48mmol) was added to a reaction flask, after dissolution in DMF (5mL), HOBT (97mg,0.72mmol) and EDCI (138mg,0.72mmol) were added, and after 30 minutes of reaction N was added1,N1Dimethylethane-1, 2-diamine (63mg,0.72mmol),after completion of the reaction, the reaction mixture was poured into water, extracted with ethyl acetate, and the organic phase was washed with saturated brine three times, dried over anhydrous sodium sulfate, concentrated, and subjected to column chromatography to give the objective compound (160mg, yield: 68.5%). 1H NMR(400MHz,CDCl3):δ9.46(s,1H),8.43(d,1H),8.17(m,1H),8.03(s,1H),7.63(s,1H),7.59(m,2H),7.42-7.41(m,2H),7.24(d,1H),7.19(s,1H),3.83(s,3H),3.69(s,3H),3.59(m, 2H),2.57(m,2H),2.26(s,6H).
And 4, step 4: synthesis of 2-amino-N- (2- (dimethylamino) ethyl) -5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzamide:
the compound N- (2- (dimethylamino) ethyl) -5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzamide (150mg,0.31mmol) was added to a reaction flask and EtOH/H was added2O-4/1 (5mL), followed by Fe (94mg,1.69mmol) and NH4Cl (131mg,2.45 mmol). After the reaction was completed, it was cooled to room temperature, filtered, washed with ethanol, dried over anhydrous sodium sulfate, and concentrated to obtain the objective compound (120mg, yield: 85.2%). MS (ESI) M/z 460.54[ M + H ]].
And 5: synthesis of 2-acrylamido-N- (2- (dimethylamino) ethyl) -5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzamide:
a solution of acryloyl chloride (22mg, 0.24mmol) in dichloromethane was added dropwise to a stirred mixture of 2-amino-N- (2- (dimethylamino) ethyl) -5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzamide (110mg, 0.24mmol) and DIPEA (38mg, 0.29mmol) in dichloromethane (2mL) cooled in an ice-water bath. After the reaction for 1.5 hours, it was diluted with dichloromethane, washed with saturated sodium bicarbonate, the aqueous phase was extracted twice with dichloromethane, and the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give the objective compound (40mg, yield: 35.4%). 1H NMR(400MHz,CDCl3):δ10.12(s,1H),9.46(s,1H),8.43(d,1H),8.17(m,1H),8.03(s,1H),7.59(m,1H),7.42-7.41(m,2H),7.31(s,1H),7.24(d,1H),7.19(s,1H),7.02(s,1H),6.48(m,1H),6.01(m,1H),5.51(m,1H),3.83(s,3H),3.69(s,3H),3.59(m,2H),2.57(m,2H),2.26(s,6H).MS(ESI):m/z 514.59[M+H].
Example 6: synthesis of N- (2- (2,2, 2-trifluoroacetyl) -5- ((4- (1- (2,2, 2-trifluoroethyl) -1H-indol-3-yl) pyrimidin-2-yl) amino) -4- (trifluoromethoxy) phenyl) acrylamide:
Figure BDA0001109816280000161
step 1: synthesis of 1- (2,2, 2-trifluoroethyl) -1H-indole
Compound 1H-indole (5.0g, 42.68mmol) was charged to a reaction flask, acetonitrile: DMF ═ 3:2(100mL) was added, and Cs was then added2CO3(16.69g,51.22mmol) and 2,2, 2-trifluoroethyl trifluoromethanesulfonate (11.89g,51.22mmol) were reacted at room temperature under nitrogen atmosphere overnight, after completion of the reaction, the solid was removed by filtration, the reaction solution was added to water, extracted with ethyl acetate, the organic phase was washed three times with saturated brine, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give the objective product (6.5g, yield: 76.5%).1H NMR(400MHz,CDCl3):δ7.59-7.55(m,2H),7.42(t,1H),7.33(d,1H),7.04(t,1H),6.41(d,1H),4.37(m,2H).
Step 2: synthesis of 3- (2-chloropyrimidin-4-yl) -1- (2,2, 2-trifluoroethyl) -1H-indole:
the compound 2, 4-dichloropyrimidine (3.5g,23.49mmol) was added to a reaction flask, ethylene glycol dimethyl ether (30mL) was added to dissolve the compound, after dissolution, ferric trichloride (3.81g,23.49mmol) and 1-methylindole (5.15g,25.84mmol) were added, and the mixture was heated to 60 ℃ and stirred for reaction overnight. After the completion of the reaction, the reaction mixture was cooled to room temperature, water (100mL) was added to precipitate a large amount of solid, which was filtered, and the filter cake was washed with methanol and dried to obtain a purple solid (5.5g, yield: 75.1%). 1H NMR(400MHz,CDCl3):δ9.15(d,1H),8.17(m,1H),7.72(d,1H),7.59(m,1H),7.42-7.39(m,2H),7.19(s,1H),4.37(m,2H).
And step 3: synthesis of 2,2, 2-trifluoro-1- (2-nitro-4- ((4- (1- (2,2, 2-trifluoroethyl) -1H-indol-3-yl) pyrimidin-2-yl) amino) -5- (trifluoromethoxy) phenyl) ethanone:
the compounds 3- (2-chloropyrimidin-4-yl) -1- (2,2, 2-trifluoroethyl) -1H-indole (500mg, 1.60mmol) and 1- (4-amino-2-nitro-5- (trifluoromethoxy) phenyl) -2,2, 2-trifluoroacetone (510mg,1.60mmol) were added to a reaction flask, then 2-pentanol (10mL) was added, p-toluenesulphonic acid monohydrate (332mg,1.92mmol) was added under stirring, heated to 105 ℃ for 2.5 hours, after completion of the reaction, cooled to room temperature, a solid precipitated, filtered, the filter cake was washed with acetonitrile, and dried to give a yellow solid (750mg, yield: 78.8%).1H NMR(400MHz,CDCl3):δ9.47(s,1H),8.43(d,1H),8.17(m,1H),7.59(m,1H),7.56(s,1H),7.50(s,1H),7.42-7.39(m,2H),7.24(d,1H),7.19(s,1H),4.37(m,2H).
And 4, step 4: synthesis of 1- (2-amino-4- ((4- (1- (2,2, 2-trifluoroethyl) -1H-indol-3-yl) pyrimidin-2-yl) amino) -5- (trifluoromethoxy) phenyl) -2,2, 2-trifluoroacetone:
the compound 2,2, 2-trifluoro-1- (2-nitro-4- ((4- (1- (2,2, 2-trifluoroethyl) -1H-indol-3-yl) pyrimidin-2-yl) amino) -5- (trifluoromethoxy) phenyl) ethanone (300mg,0.51mmol) was added to a reaction flask, EtOH: H was added2O4: 1(5mL), followed by Fe (156mg,2.78mmol) and NH4Cl (217mg,4.04 mmol). After the reaction was completed, it was cooled to room temperature, filtered, washed with ethanol, dried over anhydrous sodium sulfate, and concentrated to obtain the objective compound (260mg, yield: 91.3%). MS (ESI) M/z 564.38[ M + H ] ]
And 5: synthesis of N- (2- (2,2, 2-trifluoroacetyl) -5- ((4- (1- (2,2, 2-trifluoroethyl) -1H-indol-3-yl) pyrimidin-2-yl) amino) -4- (trifluoromethoxy) phenyl) acrylamide:
a solution of acryloyl chloride (24mg, 0.27mmol) in dichloromethane was added dropwise to a stirred mixture of 1- (2-amino-4- ((4- (1- (2,2, 2-trifluoroethyl) -1H-indol-3-yl) pyrimidin-2-yl) amino) -5- (trifluoromethoxy) phenyl) -2,2, 2-trifluoroacetone (150mg,0.27mmol) and DIPEA (42mg,0.32mmol) in dichloromethane (2mL) cooled in an ice-water bath. After 1.5 hours of reaction, diluted with dichloromethane, washed with saturated sodium bicarbonate, the aqueous phase extracted twice with dichloromethane, the organic phases combined, dried over anhydrous sodium sulfate and concentratedColumn chromatography gave the title compound (100mg, yield: 60.8%).1H NMR(400MHz,CDCl3):10.12(s,1H),9.47(s,1H),8.43(d,1H),8.17(m,1H),7.59(m,1H),7.56(s,1H),7.50(s,1H),7.42-7.39(m,2H),7.24(d,1H),7.22(s,1H),6.95(s,1H),6.48(m,1H),6.01(m,1H),5.52(m,1H),4.37(m,2H).MS(ESI):m/z:618.42[M+H]
Example 7: synthesis of 2-acrylamido-N- (2- (dimethylamino) ethyl) -5-methoxy-4- ((4- (pyrazolo [1,5-a ] pyridin-3-yl) pyrimidin-2-yl) amino) benzamide
Figure BDA0001109816280000171
Step 1: synthesis of 3-iodopyrazolo [1,5-a ] pyridine:
the compound pyrazolo [1,5-a]Pyridine (5.0g,42.32mmol) was added to a reaction flask, acetonitrile (20mL) was added to dissolve the pyridine, and then NIS (10.47g,46.56mmol) was added in portions, and the mixture was stirred under reflux for 1 hour. The reaction mixture was poured into water and extracted twice with MTBE and the organic phase was washed with 2N NaOH and 15% Na in that order 2S2O3The solution was washed with water, saturated brine and dried over anhydrous sodium sulfate, and concentrated to give the objective compound (8.7g, yield: 84.23%).1H-NMR(400MHz,CDCl3):δ8.46(d,1H),7.96(s,1H),7.48(d,1H),7.22-7.17(m,1H),6.80(td,1H);
Step 2: synthesis of 3- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazolo [1,5-a ] pyridine:
the compound 3-iodopyrazolo [1, 5-a)]Pyridine (8.5g,34.83mmol) and 2-isopropoxy-4, 4,5, 5-tetramethyl-1, 3, 2-dioxaborolan (9.72g,52.25mmol) were added to a reaction flask, then anhydrous tetrahydrofuran (60mL) was added, cooled with an ice-water bath, a tetrahydrofuran solution of isopropyl magnesium chloride (1.5M,23.22mL,34.83mmol) was added dropwise under nitrogen protection, reacted for 2 hours under ice-water bath cooling, after completion of the reaction, the reaction was quenched, extracted with ethyl acetate, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give a white solid (7.5g, yield: 88.2%).1H-NMR(400MHz,CDCl3):δ8.51(dt,1H),8.23(s,1H),7.96(dt,1H,),7.21(m,1H),6.81(td,1H),1.36(s,12H);
And step 3: synthesis of 3- (2-chloropyrimidin-4-yl) pyrazolo [1,5-a ] pyridine:
the compound 3- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazolo [1,5-a]Pyridine (7g,28.68mmol) and 2, 4-dichloropyrimidine (5.13g,34.41mmol) were added to a reaction flask, ethylene glycol dimethyl ether (120mL) was added, followed by bis (di-tert-butyl (4-dimethylaminophenyl) phosphine) palladium (II) dichloride (Pd (Amhos) Cl 2) (1.31g,1.85mmol) and sodium carbonate (2M,31.55mL,63.10mmol), replacing nitrogen, heating to 80 ℃ under nitrogen atmosphere for four hours, cooling to room temperature after completion of the reaction, diluting with water, extracting twice with ethyl acetate, drying the organic phase with anhydrous sodium sulfate, concentrating, and separating by column chromatography to obtain the objective compound (4g, yield: 60.47%).1H-NMR(400MHz,CDCl3)δ8.92(s,1H),8.91-8.85(m,1H),8.61(d,1H),8.49(m,1H),7.95(d,1H),7.65(m,1H),7.19(m,1H).
And 4, step 4: synthesis of methyl 5-methoxy-2-nitro-4- ((4- (pyrazolo [1,5-a ] pyridin-3-yl) pyrimidin-2-yl) amino) benzoate:
the compound 3- (2-chloropyrimidin-4-yl) pyrazolo [1,5-a]Pyridine (500mg, 2.17mmol) and methyl 4-amino-5-methoxy-2-nitrobenzoate (490mg,2.17mmol) were added to a reaction flask, followed by 2-pentanol (5mL), p-toluenesulphonic acid monohydrate (498mg, 2.60mmol) with stirring, heated to 105 ℃ for 2.5 hours, after completion of the reaction, cooled to room temperature, solid precipitated, filtered, the filter cake was washed with acetonitrile and dried to give a yellow solid (650mg, yield: 71.3%).1H NMR(400MHz,CDCl3):δ9.46(s,1H),8.60(d,1H),8.43(m,1H),7.81(s,1H),7.61(s,1H),7.56(s,1H),7.45(m,1H),7.24(d,1H),7.01-6.65(m,2H),3.98(s,3H),3.83(s,3H).
And 5: synthesis of 5-methoxy-2-nitro-4- ((4- (pyrazolo [1,5-a ] pyridin-3-yl) pyrimidin-2-yl) amino) benzoic acid:
the compound methyl 5-methoxy-2-nitro-4- ((4- (pyrazolo [1,5-a ] pyridin-3-yl) pyrimidin-2-yl) amino) benzoate (500mg,1.19mmol) was charged in a reaction flask, THF/MeOH (4/1) (10mL) was added, then 1M aqueous solution of lithium hydroxide (2.38mL, 2.38mmol) was added, and the mixture was stirred at room temperature for 4 hours, after completion of the reaction, pH was adjusted to 3 to 4 with 1M hydrochloric acid, extraction was performed with ethyl acetate, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to obtain the objective compound (470mg, yield: 97.2%).
Step 6: synthesis of N- (2- (dimethylamino) ethyl) -5-methoxy-2-nitro-4- ((4- (pyrazolo [1,5-a ] pyridin-3-yl) pyrimidin-2-yl) amino) benzamide:
mixing the compound 5-methoxy-2-nitro-4- ((4- (pyrazolo [1, 5-a))]Pyridin-3-yl) pyrimidin-2-yl) amino) benzoic acid (400mg, 0.98mmol) was added to a reaction flask, DMF (5mL) was added and dissolved, HOBT (200mg,1.48mmol) and EDCI (284mg,1.48mmol) were added, and N was added after 30 minutes of reaction1,N1-dimethylethane-1, 2-diamine (131mg, 1.48mmol), stirred at room temperature for reaction for 1 hour, after completion of the reaction, the reaction mixture was poured into water, extracted with ethyl acetate, the organic phase was washed with saturated brine three times, dried over anhydrous sodium sulfate, concentrated, and subjected to column chromatography to give the objective compound (350mg, yield: 74.6%).1H NMR(400MHz,CDCl3):δ9.46(s,1H),8.43(d,1H),8.17(m,1H),8.03(s,1H),7.63(s,1H),7.59(m,2H),7.42-7.41(m,2H),7.24(d,1H),7.19(s,1H),3.83(s,3H),3.69(s,3H),3.59(m,2H),2.57(m,2H),2.26(s,6H).
And 7: synthesis of 2-amino-N- (2- (dimethylamino) ethyl) -5-methoxy-4- ((4- (pyrazolo [1,5-a ] pyridin-3-yl) pyrimidin-2-yl) amino) benzamide:
mixing the compound N- (2- (dimethylamino) ethyl) -5-methoxy-2-nitro-4- ((4- (pyrazolo [1, 5-a))]Pyridin-3-yl) pyrimidin-2-yl) amino) benzamide (300mg, 0.63mmol) was added to the reaction flask and EtOH/H was added2O-4/1 (5mL), followed by Fe (193mg, 3.46mmol) and NH 4Cl (270mg, 5.04 mmol). After the reaction was completed, it was cooled to room temperature, filtered, washed with ethanol, dried over anhydrous sodium sulfate, and concentrated to obtain the objective compound (200mg, yield: 71.4%). MS (ESI) M/z 447.5[ M + H ]].
And 8: synthesis of 2-acrylamido-N- (2- (dimethylamino) ethyl) -5-methoxy-4- ((4- (pyrazolo [1,5-a ] pyridin-3-yl) pyrimidin-2-yl) amino) benzamide:
a solution of acryloyl chloride (37mg, 0.43mmol) in dichloromethane was added dropwise to 2-amino-N- (2- (dimethylamino) ethyl) -5-methoxy-4- ((4- (pyrazolo [1,5-a ] ethyl) cooled in an ice-water bath]Pyridin-3-yl) pyrimidin-2-yl) amino) benzamide (150mg, 0.34mmol) and DIPEA (65mg, 0.51mmol) in a stirred mixture of dichloromethane (3 mL). After the reaction for 1.5 hours, it was diluted with dichloromethane, washed with saturated sodium bicarbonate, the aqueous phase was extracted twice with dichloromethane, and the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give the objective compound (80mg, yield: 47.5%).1H NMR(400Hz,CDCl3):δ11.94(s,1H),10.12(s,1H),9.47(s,1H),8.43(d,1H),8.24(d,1H),7.91(s,1H),7.56(d,1H),7.50(s,1H),7.27-7.21(m,2H),7.15(t,1H),7.05(s,1H),6.44(dd,1H),6.28(dd,1H),5.77(dd,1H),3.93(m,3H),3.86(s,3H),2.89(t,2H),2.72(s,3H),2.29(t,2H),2.21(s,6H).MS(ES+):m/z:501.55[M+1].
Example 8: synthesis of N- (4-methoxy-5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) -2- (trifluoromethyl) phenyl) acrylamide:
Figure BDA0001109816280000181
Step 1: synthesis of 4-methoxy-1-nitro-2- (trifluoromethyl) benzene:
4-chloro-1-nitro-2- (trifluoromethyl) benzene (250mg,1.11mmol) was dissolved in anhydrous methanol (5mL) and a sodium block (38mg,1.67mmol) was added at room temperature. After the sodium cake was reacted, the reaction solution was refluxed for 6 hours. After the reaction was completed, it was cooled to room temperature. Water (20mL) was added and the mixture was extracted with methylene chloride (20 mL. times.3). The organic phases were combined, washed with saturated brine (40mL), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (eluent petroleum ether: ethyl acetate ═ 20:1) to give a yellow oil (180mg, yield: 81.4%).1H NMR(400MHz,CDCl3)δ8.01(d,J=9.2Hz,1H),7.10(dd,J=3.2Hz,J2=9.2H,1H),7.26(d,J=12.0Hz,1H),3.93(s,3H).
Step 2: synthesis of 4-methoxy-2- (trifluoromethyl) aniline
4-methoxy-1-nitro-2- (trifluoromethyl) benzene (9.0g,40.7mmol) was dissolved in ethyl acetate (50mL) and methanol (100mL), 10% palladium on carbon (1.2g, 50% water) was added, hydrogen balloon was added, and the mixture was stirred at room temperature overnight. After the reaction was completed, the reaction solution was filtered, and the filtrate was concentrated under reduced pressure to obtain a yellow oil (7.7g, 99.0%) which was used in the next step.
And step 3: synthesis of N- (4-methoxy-2- (trifluoromethyl) phenyl) acetamide
4-methoxy-2- (trifluoromethyl) aniline (4.8g,25.1mmol) was dissolved in dichloromethane (10mL) and triethylamine (5.0g,50.0mmol) and acetic anhydride (5.0g,50.0mmol) were added. The reaction solution was stirred at room temperature overnight. After completion of the reaction, water (100mL) was added and the mixture was extracted with methylene chloride (100 mL. times.3). The organic phases were combined, washed with saturated brine (100mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a gray solid. The gray solid was washed with a mixture of petroleum ether and ethyl acetate (20: 1) to give a white solid (5.4g, yield: 92.3%). 1H NMR(400MHz,CDCl3)δ7.84(d,J=9.2Hz,1H),7.21(br,1H),7.10(d,J=2.4Hz,1H),7.05(d,J=2.8Hz,1H),7.03(d,J=2.4Hz,1H),3.81(s,3H),2.17(s,3H).
And 4, step 4: synthesis of N- (4-methoxy-5-nitro-2- (trifluoromethyl) phenyl) acetamide
N- (4-methoxy-2- (trifluoromethyl) phenyl) acetamide (5.4g,23.2mmol) was dissolved in concentrated sulfuric acid (50mL) and placed in a chilled salt bath. Potassium nitrate (2.8g,27.8mmol) was dissolved in concentrated sulfuric acid (25mL) and slowly added dropwise to the reaction flask, maintaining the temperature below 0 ℃. After the addition was complete, the temperature was slowly raised to room temperature and stirred for 1 hour. After completion of the reaction, the reaction mixture was poured into crushed ice (300g) and extracted with ethyl acetate (200 mL. times.3). The organic phases were combined, washed with saturated brine (200mL), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (eluent petroleum ether: ethyl acetate 1:1) to give a white solid (5.1g, yield: 79.1%).1H NMR(400MHz,DMSO-d6)δ9.64(s,1H),7.94(s,1H),7.53(s,1H),3.98(s,3H),2.02(s,3H).
And 5: synthesis of N- (5-amino-4-methoxy-2- (trifluoromethyl) phenyl) acetamide
N- (4-methoxy-5-nitro-2- (trifluoromethyl) phenyl) acetamide (5.1g,18.35mmol) was dissolved in ethanol (100mL) and saturated ammonium chloride (50 mL). Zinc powder was added and the reaction solution was refluxed for 1.5 hours. After the reaction was completed, about half of the ethanol was removed by concentration under reduced pressure. The solid was filtered, washed with water, and then with a mixture of petroleum ether and ethyl acetate (1:1) to give a white solid (4.3g, yield: 84.2%). 1H NMR(400MHz,CDCl3)δ7.35(s,1H),7.20(br,1H),6.89(s,1H),4.12(b,2H),3.84(s,3H),2.15(s,3H).
Step 6: synthesis of N- (4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (trifluoromethyl) phenyl) acetamide
N- (5-amino-4-methoxy-2- (trifluoromethyl) phenyl) acetamide (496mg,2.0mmol), 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (704mg,4.0mmol) were dissolved in 2-pentanol (10mL), followed by addition of p-toluenesulfonic acid monohydrate (688mg,4.0 mmol). The reaction mixture was stirred at 130 ℃ for 4 hours, after completion of the reaction, the reaction mixture was cooled to room temperature, and saturated sodium bicarbonate (20mL) was added to the reaction mixture, followed by extraction with dichloromethane (100 mL. times.3). The organic phases were combined, washed with saturated brine (100mL), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by column chromatography on silica gel (eluent petroleum ether: ethyl acetate ═ 2:1) to give a yellow solid (113mg, yield: 12.4%).1H NMR(400MHz,MeOD/CDCl3)δ8.88(s,1H),8.25(d,J=5.6Hz,1H),8.18(m,2H),7.51(s,1H),7.40(m 1H),7.29(m,2H),7.20(m,1H),7.11(s,1H),3.96(s,3H),3.88(s,3H),2.14(s,3H).
And 7: 6-methoxy-N1Synthesis of- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -4- (trifluoromethyl) benzene-1, 3-diamine
N- (4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (trifluoromethyl) phenyl) acetamide (91mg,0.2mmol) was dissolved in concentrated hydrochloric acid. The reaction solution was heated to 110 ℃ in a sealed tube and stirred for 5 hours. After the reaction was completed, the reaction mixture was cooled to room temperature, and a 1N aqueous sodium hydroxide solution was added thereto until the pH was 8. Extracting with dichloromethane Take (10 mL. times.3). The organic phases were combined, washed with saturated brine (10mL), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (eluent petroleum ether: ethyl acetate 1:1) to give a yellow oil (48mg, yield: 58.1%).1H NMR(400MHz,CDCl3)δ8.47(m,1H),8.32(m,1H),7.93(br,1H),7.80(s,1H),7.38(m,1H),7.30(m,2H),7.07(d,J=5.2Hz,1H),6.90(s,1H),3.95(br,2H),3.88(s,6H).
And 8: synthesis of N- (4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amine) -2- (trifluoromethyl) benzene) acetyl
6-methoxy-N1- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -4- (trifluoromethyl) benzene-1, 3-diamine (28mg, 0.068mmol) was dissolved in anhydrous tetrahydrofuran (2mL) and diisopropylethylamine (13mg,0.1mmol) was added. Acryloyl chloride (7mg, 0.078mmol) was added at-40 ℃. The reaction solution was slowly warmed to room temperature and stirred for 1 hour. After completion of the reaction, water (5mL) was added, and the mixture was extracted with ethyl acetate (10 mL. times.3). The organic phases were combined, washed with saturated brine (10mL), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by column chromatography on silica gel (eluent methanol: dichloromethane ═ 1:1) to give a yellow solid (18mg, yield: 56.6%).1H NMR(400MHz,DMSO-d6)δ9.70(s,1H),8.60(s,1H),8.34(m,3H),8.12(s,1H),7.50(m,1H),7.31(d,J=5.2Hz,2H),7.23(m,3H),6.48(m,1H),6.20(dd,J1=1.0Hz,J2=8.4Hz,1H),5.72(m,1H),3.97(s,3H),3.84(s,3H).13C NMR(100MHz,DMSO-d6)δ33.51,56.92,108.18,109.09,110.97,112.60,119.94,121.80,121.86,122.66,122.87,125.58,125.64,127.27,128.69,131.72,133.22,134.01,138.11,146.34,157.83,159.45,162.53,164.84.MS(ESI):m/z 468.17[M+H].
Example 9: synthesis of N- (2-fluoro-4-methoxy-5- (4- (1-methyl-1H-indol-3-yl) pyridin-2-ylamino) phenyl) acrylamide:
Figure BDA0001109816280000191
Step 1: synthesis of 2, 4-difluoro-5-nitroaniline
2, 4-difluoroaniline (24.0g,186mmol) was dissolved in concentrated sulfuric acid (200mL) and nitric acid (11.7g,186mmol) was added slowly dropwise at 0 ℃ with the temperature maintained below 4 ℃ during the addition. Stirring in an ice water bath for half an hour. After the reaction was completed, the reaction solution was poured into ice water (1L), and sodium hydroxide was slowly added until the pH was 8. Extraction with ethyl acetate (1L × 3), combination of organic phases, washing of the organic phases with saturated brine (1L), drying over anhydrous sodium sulfate, concentration under reduced pressure, and separation and purification of the crude product by silica gel column chromatography (eluent petroleum ether: ethyl acetate ═ 10:1) gave a red solid (22.8g, yield: 70.4%).1H NMR(400MHz,CDCl3)δ7.49(m,1H),6.95(t,J=10.0Hz,1H),3.89(s,1H)。
Step 2: synthesis of 4-fluoro-2-methoxy-5-nitroaniline
2, 4-difluoro-5-nitroaniline (3.48g,20mmol) was dissolved in anhydrous methanol (50mL), and sodium methoxide (1.30g,24mmol) was added at ordinary temperature and stirred for 48 hours. After completion of the reaction, water (100mL) was added and the mixture was extracted with methylene chloride (100 mL. times.3). The organic phases were combined, washed with saturated brine (100mL), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (eluent petroleum ether: ethyl acetate ═ 6:1) to give a red solid (3.26g, yield: 87.6%). 1H NMR(400MHz,CDCl3)δ7.38(d,J=7.6Hz,1H),6.62(d,J=12.4Hz,1H),3.92(s,3H),3.88(br,2H).MS(ESI)m/z:187.1[M+H].
And step 3: synthesis of N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (1-methyl-1H-indol-3-yl) pyrimidin-2-amine
4-fluoro-2-methoxy-5-nitroaniline (1.40g,7.52mmol), 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (1.83g,7.52mmol) was dissolved in 2-pentanol (50mL), followed by addition of p-toluenesulfonic acid monohydrate (1.55g,9.0 mmol). The reaction mixture was stirred at 105 ℃ for 3 hours, and after completion of the reaction, it was cooled to room temperature and filtered through a suction filter funnel. The solid was washed with 2-pentanol (50mL) and then with a mixed solvent of dichloromethane (50mL) and petroleum ether (50 mL). Then, it was dried in vacuo to give a yellow solid (2.80g, yield: 94.6%).1H NMR(400MHz,MeOD/CDCl3)δ9.82(br,1H),8.65(s,1H),8.48(s,1H),8.12(s,1H),7.57(d,J=8.0Hz,1H),7.49(m,2H),7.28(m,1H),7.10(m,1H),5.72(s,1H),3.96(s,3H),3.88(s,3H).MS(ESI)m/z:394.1[M+H].
And 4, step 4: 4-fluoro-6-methoxy-N1Synthesis of- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) benzene-1, 3-diamine
N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (1-methyl-1H-indol-3-yl) pyrimidin-2-amine (796mg,2.0mmol) was then added 10% palladium on carbon catalyst (80mg) under a nitrogen atmosphere, then the nitrogen was replaced with hydrogen three times, and the reaction mixture was stirred at room temperature overnight. After completion of the reaction, the palladium-carbon catalyst was filtered off, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (eluent dichloromethane: methanol 20:1) to obtain a brown solid (582mg, yield: 80.2%). 1H NMR(400MHz,DMSO-d6)δ8.33(d,J=6.8Hz,1H),8.21(d,J=4.8Hz,1H),8.05(d,J=10.0Hz,1H),7.91(s,1H),7.42(s,1H),7.32(d,J=8.4Hz,1H),7.18(m,2H),6.99(d,J=5.2Hz,1H),6.60(d,J=12.0Hz,1H),3.94(br,2H),3.81(s,3H),3.75(s,3H).MS(ES+):m/z 364.2[M+H].
And 5: synthesis of N- (2-fluoro-4-methoxy-5- (4- (1-methyl-1H-indol-3-yl) pyridin-2-ylamino) phenyl) acrylamide
4-fluoro-6-methoxy-N1- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) benzene-1, 3-diamine (137mg, 0.377mmol) was dissolved in anhydrous tetrahydrofuran (5mL), and diisopropylethylamine (97mg,0.75mmol) was added. Acryloyl chloride (40mg, 0.45mmol) was added at-5 ℃. The reaction solution was stirred at 5 ℃ for 2 hours. After completion of the reaction, water (0.5mL) was added, the reaction mixture was concentrated under reduced pressure, and the crude solid was washed with methylene chloride to give a yellow solid (87mg, yield: 55.3%).1H NMR(400MHz,DMSO-d6)δ9.78(s,1H),8.81(d,J=8.8Hz,1H),8.44(s,1H),8.25(m,2H),7.81(s,1H),7.44(d,J=8.0Hz,1H),7.17(m,3H),7.00(d,J=12.0Hz,1H),6.57(m,1H),6.25(d,J=16.8Hz,1H),5.70(d,J=12.0Hz,1H),3.86(s,6H).13C NMR(100MHz,DMSO-d6)δ33.29,56.69,99.67,99.92,107.85,110.71,112.89,117.05,117.72,117.88,121.33,121.79,122.36,125.16,125.79,126.81,132.10,133.86,138.07,157.89,160.12,162.18,163.71.MS(ESI)m/z:418.2[M+H].
Example 10: synthesis of N- (2- ((2- (methylamino) ethyl) (methyl) amino) -4-methoxy-5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) phenyl) cyclopropanecarboxamide:
Figure BDA0001109816280000201
step 1: n is a radical of1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4Synthesis of- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -2-nitrophenyl-1, 4-diamine:
n- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (1-methyl-1H-indol-3-yl) pyrimidin-2-amine (393mg, 1.0mmol) was dissolved in dimethylacetamide (20mL), and N, N, N' -trimethylethylenediamine (122mg, 1.2mmol) and N, N-diisopropylethylamine (168mg, 1.3mmol) were added. The reaction solution was stirred at 85 ℃ for 5 hours. After the reaction was completed, it was cooled to room temperature, and 20mL of water was added. A solid precipitate appeared, which was then filtered, washed with water, and then dried in vacuo to give a yellow solid (421mg, yield: 88.5%). 1H NMR(400MHz,MeOD/CDCl3)δ9.34(s,1H),8.24(d,J=5.2Hz,1H),8.13(m,2H),7.60(s,1H),7.37(d,J=5.2Hz,1H),7.22(m,2H),7.15(d,J=5.2Hz,1H),6.72(s,1H),3.98(s,3H),3.86(s,3H),3.29(m,2H),2.85(s,3H),2.70(m,2H),2.35(s,6H).
Step 2: n is a radical of1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4Synthesis of- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) benzene-1, 2, 4-triamine:
will N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -2-nitrobenzene-1, 4-diamine (666mg, 1.4mmol) was dissolved in ethanol (40mL), and iron powder (470mg,8.4mmol) and ammonium chloride (241mg,4.5mmol) were added. The reaction was stirred at reflux for 2 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the crude product was purified by column chromatography on silica gel (eluent dichloromethane: methanol 20:1) to give a brown oil (552mg, yield: 88.6%).1H NMR(400MHz,CDCl3)δ8.48(d,J=8.8Hz,1H),8.29(d,J=5.2Hz,1H),8.14(s,1H),7.74(s,1H),7.57(s,1H),7.29(m,3H),6.97(d,J=5.2Hz,1H),6.70(s,1H),3.94(br,2H),3.83(s,3H),3.82(s,3H),2.96(m,2H),2.67(s,3H),2.41(m,2H),2.26(s,6H)。
And step 3: synthesis of N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) cyclopropanecarboxamide:
will N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) benzene-1, 2, 4-triamine (73mg,0.164mmol) was dissolved in anhydrous tetrahydrofuran (5mL), and diisopropylethylamine (32mg,0.250mmol) was added. Acryloyl chloride (21mg,0.200mmol) was added at-10 ℃. The reaction solution was stirred at 10 ℃ for 1 hour. After completion of the reaction, water (0.5mL) was added, the reaction mixture was concentrated under reduced pressure, and the crude solid was washed with dichloromethane to give a yellow solid (62mg, yield: 73.6%). 1H NMR(400MHz,DMSO-d6)δ9.99(s,1H),9.72(s,1H),9.03(s,1H),8.34(d,J=5.6Hz,1H),8.04(d,J=8.8Hz,1H),7.71(s,1H),7.37(d,J=6.8Hz,1H),7.24(m,3H),7.17(d,J=5.6Hz,1H),6.76(s,1H),3.95(s,3H),3.86(s,3H),3.47(s,1H),2.93(m,2H),2.69(s,3H),2.34(m,8H),1.22(m,2H),1.09(m,2H).13C NMR(100MHz,DMSO-d6)δ7.40,7.61,15.50,32.91,43.75,45.20,55.96,57.08,104.46,107.82,109.24,110.12,113.59,120.25,120.86,121.72,125.94,127.56,129.91,133.87,135.05,138.20,143.67,157.75,159.63,162.13,171.17.MS(ES+):m/z 513.8[M+H].
Example 11: synthesis of N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) methacrylamide:
Figure BDA0001109816280000211
will N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) benzene-1, 2, 4-triamine (50mg,0.337mmol) was dissolved in anhydrous tetrahydrofuran (5mL), and diisopropylethylamine (56mg,0.438mmol) was added. Acryloyl chloride (42mg, 0.400mmol) was added at-10 ℃. The reaction solution isStirred at 10 ℃ for 1 hour. After completion of the reaction, water (0.5mL) was added, the reaction mixture was concentrated under reduced pressure, and the crude solid was washed with methylene chloride to give a yellow solid (124mg, yield: 71.6%).1H NMR(400MHz,DMSO-d6)δ9.78(s,1H),9.74(s,1H),9.06(s,1H),8.36(d,J=5.2Hz,1H),8.05(d,J=8.4Hz,1H),7.73(s,1H),7.36(d,J=8.8Hz,1H),7.23(m,3H),6.78(s,1H),5.87(s,3H),5.44(s,3H),3.95(s,3H),3.86(s,3H),2.99(m,2H),2.65(s,3H),2.31(m,2H),2.21(s,6H),2.11(s,3H).13C NMR(100MHz,DMSO-d6)δ18.94,32.97,44.58,45.13,54.97,56.12,57.14,76.79,77.11,77.43,104.35,107.89,109.50,110.02,113.58,119.19,120.23,120.89,121.75,125.93,133.96,135.04,138.22,141.44,144.19,157.84,159.54,162.08,165.67.MS(ESI)m/z:513.8[M+H].
Example 12: synthesis of 3- (dimethylamino) -N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) propionamide:
Figure BDA0001109816280000212
step 1: synthesis of N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide:
a solution of acryloyl chloride (41mg, 0.45mmol) in methylene chloride was added dropwise to N cooled in an ice-water bath 1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4A stirred mixture of (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) benzene-1, 2, 4-triamine (200mg, 0.45mmol) and DIPEA (70mg, 0.54mmol) in dichloromethane (5 mL). After the reaction for 1.5 hours, it was diluted with dichloromethane, washed with saturated sodium bicarbonate, the aqueous phase was extracted twice with dichloromethane, and the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give the objective compound (130mg, yield: 57.97%).1H NMR(400MHz,CDCl3):δ10.22(s,1H),9.14(s,1H),8.68(s,1H),8.33(d,1H),8.24(d,1H),7.91(s,1H),7.53(d,1H),7.27-7.20(m,2H),7.15(t,1H),7.04(s,1H),6.43(dd,1H),6.27(dd,1H),5.77(dd,1H),3.92(s,3H),3.86(s,3H),2.89(t,2H),2.72(s,3H),2.29(t,2H),2.21(s,6H).MS(ESI):m/z 500.42[M+H].
Step 2: synthesis of 3- (dimethylamino) -N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) propionamide:
the compound N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide (100mg,0.20mmol) was added to a reaction flask, followed by addition of ethanol (5mL) for dissolution, addition of 30% dimethylamine ethanol solution (1mL) with stirring, reaction with stirring at room temperature for 30 minutes, concentration after completion of the TLC detection reaction, and TLC purification to give the objective compound as an off-white solid (80mg, yield: 73.38%).1H NMR(400MHz,DMSO-d6)δ10.65(s,1H),10.07(s,1H),9.91(s,1H),8.58(s,2H),8.28(s,1H),7.56(d,J=7.8Hz,1H),7.25(dt,J=28.2,7.2Hz,3H),6.99(s,1H),3.92(s,3H),3.86(s,3H),3.23(m,4H),2.77(s,12H),2.65(m,4H).MS(ESI)m/z:545.33[M+H].
Example 13: synthesis of 2-acrylamido-5-methoxy-N, N-dimethyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) aniline:
Figure BDA0001109816280000221
Step 1: synthesis of 2-amino-5-methoxy-N, N-dimethyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzamide:
2-amino-5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzoic acid (117mg,0.300mmol) and dimethylamine hydrochloride (41mg, 0.500mmol) were dissolved in DMF (3mL) and diisopropylethylamine (129mg,1.0mmol) and HATU (171mg,0.450mmol) were added. The reaction mixture was stirred at room temperature overnight, and after completion of the reaction, the mixture was diluted with methylene chloride (20mL), water (20mL) was added, and the mixture was extracted with methylene chloride (20 mL. times.3). The organic phases were combined, washed with saturated brine (50mL), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (washing)The remover is dichloromethane: methanol 40: 1) to give a yellow solid (110mg, 88.1%).1H NMR(400MHz,CDCl3)δ8.45(d,J=6.8Hz,1H),8.29(d,J=5.6Hz,1H),8.20(s,1H),7.98(s,1H),7.85(s,1H),7.80(s,1H),7.36(m,1H),7.28(m,2H),7.05(d,J=5.2Hz,1H),6.67(s,1H),3.85(s,3H),3.82(s,3H),3.07(s,6H).MS(ESI)m/z 417.10[M+H].
Step 2: synthesis of 2-acrylamido-5-methoxy-N, N-dimethyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) aniline:
2-amino-5-methoxy-N, N-dimethyl-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzamide (29) (100mg, 0.240mmol) was dissolved in anhydrous tetrahydrofuran (5mL) and diisopropylethylamine (46mg, 0.360mmol) was added. Acryloyl chloride (24mg, 0.270mmol) was added at-10 ℃. Stirred at-10 ℃ for 1 hour. After completion of the reaction, water (5mL) was added and the mixture was extracted with methylene chloride (10 mL. times.3). The organic phases were combined, washed with saturated brine (20mL), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by column chromatography on silica gel (eluent dichloromethane: methanol ═ 40: 1) to give a yellow solid (38m, yield: 33.7%). 1H NMR(400MHz,DMSO-d6)δ9.57(m,2H),8.78(s,1H),8.33(d,J=5.2Hz,1H),8.03(d,J=8.4Hz,1H),7.85(s,1H),7.33(d,J=6.0Hz,1H),7.22(m,3H),6.70(s,1H),6.30(m,2H),5.71(d,J=10.0Hz,1H),3.89(s,3H),3.81(s,3H),3.05(s,6H).MS(ESI)m/z:471.10[M+H].
Example 14: synthesis of methyl 2-acrylamido-5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzoate:
Figure BDA0001109816280000222
step 1: synthesis of methyl 2-amino-5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzoate:
the compound methyl 5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2-nitrobenzoate (500mg,1.15mmol), iron powder (354mg,6.34mmol) and ammonium chloride (494mg,9.23mmol) were added to a reaction vesselThe flask was charged with EtOH/H2O-4/1 (10mL) solution was refluxed for 2 hours, cooled, filtered through celite, washed with dichloromethane, the organic phase was extracted with dichloromethane, dried over anhydrous sodium sulfate, and concentrated to give the objective compound (400mg, yield: 85.95%). 1H NMR (400MHz, CDCl3) δ 8.47(d, J ═ 9.2Hz,1H),8.33(d, J ═ 5.2Hz,1H),8.23(s,1H),7.97(M,2H),7.31(M,5H),7.04(d, J ═ 5.2Hz,1H),3.84(M,9H) ms (esi) M/z 471.10[ M + H471.10 [ M + esi ]].
Step 2: synthesis of methyl 2-acrylamido-5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzoate:
methyl 2-amino-5-methoxy-4- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) benzoate (30) (28mg,0.070mmol) was dissolved in THF (5mL) and diisopropylethylamine (9mg, 0.10mmol) was added. Acryloyl chloride (10mg, 0.080mmol) was added at-10 ℃. The reaction solution was stirred at-10 ℃ for 1 hour. After completion of the reaction, water (5mL) was added and the mixture was extracted with methylene chloride (10 mL. times.3). The organic phases were combined, washed with saturated brine (20mL), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by column chromatography on silica gel (eluent dichloromethane: methanol ═ 100: 1) to give a yellow solid (21mg, yield: 65.6%). 1H NMR (400MHz, DMSO-d6) δ 11.48(s,1H),10.09(s,1H),9.04(s,1H),8.40(d, J ═ 5.2Hz,1H),8.05(M,2H),7.44(s,1H),7.36(M,1H),7.25(M,3H),6.40(M,2H),5.77(M,1H),3.94(s,3H),3.91(s,3H),3.89(s,3H). ms (esi) M/z 458.11[ M + H ].
Example 15: synthesis of 1- (6-methoxy-7- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -3, 4-dihydroquinolin-1 (2H) -yl) prop-2-en-1-one:
Figure BDA0001109816280000231
step 1: synthesis of 6-methoxy-3, 4-dihydroquinolin-2 (1H) -one:
the compound 6-hydroxy-3, 4-dihydroquinolin-2 (1H) -one (2.0g, 12.26mmol), K2CO3(2.5g, 18.38mmol) in a three-neck flask, adding 15mL of DMF, heating to 42 ℃ for reaction for 1h, and then dropwise adding under the condition of keeping out of the lightMethyl iodide (2.1g, 14.72mmol) was added and the progress of the reaction was checked by TLC spot plate, after 3h the reaction was complete, and after moving to room temperature 75mL ice water was added and standing for 0.5h and filtration gave a white solid (2.0g, 92.1% yield).1H NMR(400MHz,CDCl3)δ8.40(s,1H),6.72(t,J=2.0Hz,3H),3.78(s,3H),2.96–2.91(m,2H),2.64–2.59(m,2H).
Step 2: synthesis of 6-methoxy-1, 2,3, 4-tetrahydroquinoline:
the compound 6-methoxy-3, 4-dihydroquinolin-2 (1H) -one (1.1g, 6.21mmol) was placed in a three-necked flask with 50mL of anhydrous THF and LiAlH was added in portions under ice bath conditions4(0.7g, 18.63mmol), heating and refluxing after the addition is finished, detecting the progress of the reaction by a TLC point plate, completing the reaction for about 2 hours, carrying out post-treatment, moving to an ice bath condition, dropwise adding 0.7g of water, stirring for 15 minutes, dropwise adding 15% sodium hydroxide solution (0.7g), stirring for 15 minutes, dropwise adding 2.1g of water, stirring for 0.5 hours, then adding anhydrous magnesium sulfate (0.7g), stirring for 0.5 hours, filtering, washing a filter cake with THF, drying a filtrate with anhydrous magnesium sulfate, and concentrating to obtain an oil (0.92g, yield 91.1%). 1H NMR(400MHz,CDCl3)δ6.64–6.58(m,1H),6.48(d,J=8.5Hz,0H),3.75(s,2H),3.30–3.25(m,1H),2.78(t,J=6.5Hz,1H),2.00–1.91(m,1H).
And step 3: synthesis of 1- (6-methoxy-3, 4-dihydroquinolin-1 (2H) -yl) prop-2-en-1-one:
the compound 6-methoxy-1, 2,3, 4-tetrahydroquinoline (0.9g, 5.54mmol) is put into a three-neck flask, DCM (10mL) is added, DIPEA (0.9g, 7.20mmol) is added under ice bath conditions, allylacyl chloride (0.6g, 6.65mmol) is added dropwise and the reaction is continued under ice bath conditions, the progress of the reaction is checked by TLC spot plate, about 3h reaction is completed, post-treatment is carried out, DCM (20mL) is added to dilute the reaction solution, the reaction solution is washed with saturated sodium bicarbonate solution (20mL), the organic phase is dried with anhydrous sodium sulfate and concentrated to obtain (1.12g, yield 93.3%).1H NMR(400MHz,CDCl3)δ6.77–6.66(m,1H),6.59–6.46(m,1H),6.43–6.36(m,1H),6.23–6.05(m,0H),5.63(dd,J=10.0,2.2Hz,1H),3.84(t,J=6.7Hz,2H),3.80(s,3H),2.68(t,J=6.6Hz,2H),1.96(p,J=6.6Hz,2H).
And 4, step 4: synthesis of 1- (6-methoxy-7-nitro-3, 4-dihydroquinolin-1 (2H) -yl) prop-2-en-1-one:
after the compound 1- (6-methoxy-3, 4-dihydroquinolin-1 (2H) -yl) prop-2-en-1-one (1.12g, 5.16mmol) was added with TFA (18mL) under ice bath conditions, sodium nitrite (0.37g, 5.41mmol) was added in portions, the mixture was allowed to stand at room temperature to continue the reaction, after completion of the reaction for about 1.5 hours, ice water (200mL) was added, extraction was performed with DCM (30 mL. times.3), the organic layers were combined, dried over anhydrous magnesium sulfate, concentrated, and purified by column chromatography to obtain a solid (0.3g, yield: 22.2%).1H NMR(400MHz,CDCl3)δ6.89(s,1H),6.56–6.42(m,2H),5.77(dd,J=7.7,4.4Hz,1H),5.34–5.20(m,1H),3.96(s,3H),3.86(t,J=6.6Hz,2H),2.80(t,J=6.5Hz,2H),2.05–1.96(m,2H).
And 5: synthesis of 1- (7-amino-6-methoxy-3, 4-dihydroquinolin-1 (2H) -yl) prop-2-en-1-one:
The compound 1- (6-methoxy-7-nitro-3, 4-dihydroquinolin-1 (2H) -yl) prop-2-en-1-one (0.3g, 1.14mmol), reduced iron powder (0.38g, 6.84mmol) and ammonium chloride (0.61g, 11.4mmol) were placed in a two-necked flask, ethanol (21mL) and water (7mL) were added, heated under reflux, the reaction progress was checked by TLC spot plate, after about 1.5H, post-treatment, filtration while hot, the cake was washed twice with ethanol, the filtrate was concentrated to remove ethanol, after which the aqueous layer was extracted with DCM (10 mL. times.3), the organic layers were combined, dried over anhydrous magnesium sulfate, and desolventized to give the product (0.23g, yield 88.5%).1H NMR(400MHz,CDCl3)δ6.86(s,1H),6.54–6.43(m,2H),5.75(dd,J=7.7,4.4Hz,1H),5.32–5.24(m,1H),4.18(s,2H),3.94(s,3H),3.87(t,J=6.6Hz,2H),2.80(t,J=6.5Hz,2H),2.05–1.96(m,2H).
Step 6: synthesis of 1- (6-methoxy-7- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -3, 4-dihydroquinolin-1 (2H) -yl) prop-2-en-1-one:
the compound 1- (7-amino-6-methoxy-3, 4-dihydroquinolin-1 (2H) -yl) prop-2-en-1-one (0.22g, 0.95mmol), 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (0.19g, 0.79mmol) and p-chlorobenzoic acid (0.16g, 0.95mmol) were placed in a two-necked flask, 1, 4-dioxane (6mL) was added and heated to 90 ℃ to effect a reaction, and the progress of the reaction was checked by TLC dot plateAfter about 5 hours of completion of the reaction, the reaction was post-treated, cooled to room temperature, quenched by addition of 25% aqueous ammonia (0.2mL) and water (0.97mL), desolventized, and purified by column chromatography to give the product (83mg, yield 24.0%). 1H NMR(400MHz,CDCl3)δ8.68(s,1H),8.36(d,J=5.3Hz,1H),8.24(d,J=7.3Hz,1H),7.91(s,1H),7.74(s,1H),7.42(d,J=7.6Hz,1H),7.33(dd,J=16.3,7.4Hz,2H),7.14(d,J=5.3Hz,1H),6.84–6.70(m,2H),6.40(dd,J=16.8,2.0Hz,1H),5.58(d,J=10.4Hz,1H),3.99–3.87(m,8H),2.74(t,J=6.6Hz,2H),2.03(p,J=6.6Hz,2H).MS(ESI)m/z 440.21[M+H].
Example 16: synthesis of 1- (5-methoxy-6- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) indol-1-yl) prop-2-en-1-one:
Figure BDA0001109816280000241
step 1: synthesis of 5-methoxyindole:
weighing 5-methoxy-1H-indole (0.4g, 2.72mmol), adding acetic acid (25mL) into a three-neck flask, adding sodium cyanoborohydride (0.51g, 8.16mmol) in batches, continuing the reaction at room temperature after the addition is finished, detecting the progress of the reaction by a TLC (thin layer chromatography) spot plate, finishing the reaction after about 2.5H, adding water (3mL) for quenching, adjusting the pH to 5-6 by using saturated sodium bicarbonate solution, extracting DCM (10mL multiplied by 4), combining organic layers, drying over anhydrous magnesium sulfate, and concentrating to obtain the target compound (0.35g, yield 86.3%).1H NMR(400MHz,CDCl3)δ6.78–6.75(m,1H),6.67(d,J=8.4Hz,1H),6.61(dd,J=8.4,2.5Hz,1H),3.75(s,3H),3.56(t,J=8.2Hz,2H),3.36(s,2H),3.01(t,J=8.2Hz,2H).
Step 2: synthesis of 1- (5-methoxyindol-1-yl) prop-2-en-1-one:
weighing 5-methoxyindole (0.35g, 2.35mmol), adding DCM (7mL) to dissolve, adding DIPEA (0.36g, 2.82mmol) under an ice bath condition, dropwise adding allyl chloride (0.23g, 2.59mmol), continuing to react under the ice bath condition after the addition is finished, detecting the reaction progress through a TLC (thin layer chromatography) point plate, after about 5h, carrying out post-treatment, adding DCM (20mL) to dilute the reaction solution, and diluting the reaction solution with DCM (20mL)The saturated sodium bicarbonate solution (20mL) was washed, and the sodium bicarbonate solution layer was extracted with DCM (10mL × 3), dried over anhydrous magnesium sulfate, concentrated, and purified by column chromatography to give the objective compound (0.28g, yield 58.7%). 1H NMR(400MHz,CDCl3)δ8.23(d,J=8.9Hz,1H),6.75(d,J=8.2Hz,2H),6.62–6.44(m,2H),5.77(dd,J=9.6,2.4Hz,1H),4.20–4.15(m,2H),3.79(s,3H),3.19(t,J=8.3Hz,2H).
And step 3: synthesis of 1- (5-methoxy-6-nitroindol-1-yl) prop-2-en-1-one:
weighing 1- (5-methoxyindol-1-yl) prop-2-en-1-one (0.27g, 1.33mmol) in a two-neck flask, adding TFA (5mL) under ice bath conditions, adding sodium nitrite (0.097g, 1.40mmol) in batches, moving to room temperature for continuing the reaction, adding ice water (50mL) into the reaction solution after about 2h of reaction completion, precipitating a solid, standing for 1h, filtering, and drying a filter cake to obtain a product (0.25g, yield 75.8%).1H NMR(400MHz,CDCl3)δ8.77(s,1H),6.92(d,J=10.0Hz,1H),6.54(d,J=5.5Hz,2H),5.84(t,J=6.0Hz,1H),4.24(t,J=8.5Hz,2H),3.92(s,3H),3.28(t,J=8.3Hz,2H).
And 4, step 4: synthesis of 1- (-amino-5-methoxyindol-1-yl) -2-en-1-one:
1- (5-methoxy-6-nitroindol-1-yl) prop-2-en-1-one (0.25g, 1.01mmol), reduced iron powder (0.34g, 6.06mmol) and ammonium chloride (0.54g, 10.1mmol) were weighed into a two-necked flask, ethanol (21mL) and water (7mL) were added and heated under reflux, the progress of the reaction was checked by TLC spotting, after-treatment was carried out for about 1h, filtration was carried out while hot, the cake was washed twice with ethanol, ethanol was removed by concentration, the aqueous layer was extracted with DCM (10 mL. times.3), dried over anhydrous magnesium sulfate and concentrated to give a solid (0.16g, yield 72.7%).1H NMR(400MHz,DMSO)δ7.64(s,1H),6.73(d,J=8.0Hz,2H),6.24(d,J=16.1Hz,1H),5.76(t,J=6.0Hz,1H),4.66(s,2H),4.14(t,J=7.5Hz,2H),3.73(s,3H),3.02(t,J=7.3Hz,2H).
And 5: synthesis of 1- (5-methoxy-6- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) indol-1-yl) prop-2-en-1-one:
Weighing 1- (-amino-5-methoxyindol-1-yl) -2-en-1-one (0.16g, 0.73mmol), 3- (2-chloropyrimidine)Pyridine-4-yl) -1-methyl-1H-indole (0.15g, 0.61mmol) and p-chlorobenzoic acid (0.13g, 0.73mmol) are added into a double-neck flask, 1, 4-dioxane (6mL) is added and heated to 90 ℃ for reaction, the reaction progress is detected by a TLC point plate, the reaction is finished after about 24H, the post-treatment is carried out, the temperature is reduced to the room temperature, 25% ammonia water (0.15mL) and water (0.75mL) are added for quenching, desolventization and purification are carried out by column chromatography, and the product is obtained (117mg, yield 45.2%).1H NMR(400MHz,CDCl3)δ9.82(s,1H),9.07(s,1H),8.38(d,J=5.3Hz,1H),8.06(d,J=6.7Hz,1H),7.76(s,1H),7.39(d,J=7.0Hz,1H),7.32–7.23(m,2H),7.20(d,J=5.2Hz,1H),6.76(s,1H),6.66(dd,J=16.7,10.1Hz,1H),6.53(d,J=15.2Hz,1H),5.78(d,J=9.0Hz,1H),4.21(t,J=8.3Hz,2H),4.00(s,3H),3.89(s,3H),3.19(t,J=8.2Hz,2H).MS(ESI)m/z:426.28[M+H].
Example 17: synthesis of N- (2- ((2- (dimethylamino) ethyl) sulfinyl) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide:
Figure BDA0001109816280000242
step 1: synthesis of N- (4- ((2- (dimethylamino) ethyl) thio) -2-methoxy-5-nitrophenyl) -4- (1-methyl-1H-indol-3-yl) pyrimidin-2-amine
NaH (60%, 2.03g,50.84mmol) was charged into a reaction flask, DMF (25mL) was added, a solution of 2- (dimethylamino) ethanethiol hydrochloride (3.60g,25.42mmol) in DMF (25mL) was added under nitrogen, the reaction mixture was stirred at room temperature for 45 minutes, then a solution of N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (1-methyl-1H-indol-3-yl) pyrimidin-2-amine (2.0g,5.08mmol) in DMF (25mL) was slowly added dropwise, and after completion of the addition, the mixture was stirred at room temperature overnight. After completion of the reaction, the reaction mixture was poured into water, filtered, and the filter cake was washed with water and dried in vacuo to give a yellow solid (2.2g, yield: 90.42%).
Step 2: 4- ((2- (dimethylamino) ethyl) thio) -6-methoxy-N1Synthesis of- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) benzene-1, 3-diamine:
taking Fe powder (0.77g,12mmoL) and NH4Cl (1.3g,24mmoL) in water (10mL) and ethanol (10mL) was added the compound N- (4- ((2- (dimethylamino) ethyl) thio) -2-methoxy-5-nitrophenyl) -4- (1-methyl-1H-indol-3-yl) pyrimidin-2-amine (0.96g,2mmoL) in ethanol (20mL), refluxed in an oil bath at 80 ℃ for 2 hours, the reaction was completed (DCM: MeOH ═ 20:1), solids were removed by hot filtration, the filtrate was concentrated, water was added in 50mL, extracted with dichloromethane, dried over anhydrous sodium sulfate of the organic phase, and concentrated to give the product (0.5g, yield: 56%).1H NMR(400MHz,DMSO-d6)δ8.45(d,J=7.8Hz,1H),8.36(d,J=4.6Hz,2H),7.92(s,1H),7.87(s,1H),7.55(d,J=8.0Hz,1H),7.32–7.19(m,3H),7.02(s,1H),3.90(s,3H),3.84(s,3H),3.17(dd,J=8.8,4.9Hz,3H),3.09(dd,J=8.9,4.8Hz,2H),2.74(s,6H).
And step 3: synthesis of N- (2- ((2- (dimethylamino) ethyl) thio) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide:
taking a compound 4- ((2- (dimethylamino) ethyl) sulfenyl) -6-methoxyl-N1- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) benzene-1, 3-diamine (1.1g,2.45mmol) was dissolved in 20mL of dichloromethane, stirred in an ice-water bath for 10 minutes, diisopropylethylamine (613mg,4.9mmol) was added, a solution of acryloyl chloride (246mg,2.7mmol) in dichloromethane (2 mL) was added, stirred in an ice-water bath for 2 hours, the reaction mixture was quenched (DCM: MeOH ═ 20:1), dichloromethane (100 mL) was added, a solution of sodium bicarbonate (100 mL) was added, the mixture was separated, dried over anhydrous sodium sulfate, the crude product was concentrated, and the crude product was purified by column chromatography (DCM: MeOH ═ 100:1) to obtain a product (600mg, yield 49%). 1H NMR(400MHz,CDCl3)δ10.14(s,1H),9.77(s,1H),8.98(s,1H),8.42(d,J=5.3Hz,1H),8.09(dd,J=6.3,2.2Hz,1H),7.88(s,1H),7.42(dd,J=6.5,2.3Hz,1H),7.35–7.23(m,3H),7.11(s,1H),6.51(dd,J=16.9,1.8Hz,1H),6.39(dd,J=16.9,10.0Hz,1H),5.78(dd,J=10.0,1.7Hz,1H),4.01(s,3H),3.94(s,3H),2.89(t,J=6.2Hz,2H),2.38(t,J=6.2Hz,2H),2.29(s,6H).
And 4, step 4: synthesis of N- (2- ((2- (dimethylamino) ethyl) sulfinyl) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide
Taking a compound N- (2- ((2- (dimethyl)Amino) ethyl) thio) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide (580mg,1.15mmol) was dissolved in dichloromethane (30mL), stirred in an ice-water bath for 10 minutes, 1mL trifluoroacetic acid was added, a solution of 70% m-chloroperoxybenzoic acid (290mg,1.2mmol) in dichloromethane (2mL) was added, stirred in an ice-water bath for 0.5 hour, the reaction was spotted (DCM: MeOH ═ 20:1), dichloromethane (100mL) was added to the reaction mixture, 100mL sodium bicarbonate solution was added, the mixture was separated, the organic phase was dried over anhydrous sodium sulfate, the concentrated crude product was purified by column chromatography (DCM: MeOH ═ 100:1 to 20:1) gave the product (300mg, yield: 47%).1H NMR(400MHz,CDCl3)δ10.43(s,1H),9.74(s,1H),8.84(s,1H),8.43(d,J=5.3Hz,1H),8.13–8.06(m,1H),7.98(s,1H),7.41(dd,J=6.6,2.3Hz,1H),7.35–7.24(m,3H),7.11(s,1H),6.47(dd,J=17.0,1.2Hz,1H),6.28(dd,J=17.0,10.3Hz,1H),5.80(dd,J=10.3,1.0Hz,1H),3.97(d,J=4.8Hz,6H),3.58–3.46(m,1H),3.03(dd,J=7.8,5.3Hz,1H),2.60(dd,J=11.8,6.5Hz,1H),2.49(dd,J=8.9,5.7Hz,1H),2.32(s,6H).MS(HESI)m/z:519.22[M+H].
Example 18: synthesis of 3- (dimethylamino) -N- (2- ((2- (dimethylamino) ethyl) sulfinyl) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) propionamide:
Figure BDA0001109816280000251
the compound N- (2- ((2- (dimethylamino) ethyl) sulfinyl) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide (50mg,0.096mmol) was taken in tetrahydrofuran (5mL), stirred in an ice-water bath for 10 minutes, dimethylamine-tetrahydrofuran solution (1M,2mL,2mmol) was added, stirred in an ice-water bath for 0.5 hour, the reaction was stopped (DCM: MeOH 10:1), the reaction was concentrated to give a crude product, and the crude product was purified by column chromatography (DCM: MeOH 100:1 to 20:1) to give a product (12mg, 21% yield). 1H NMR(400MHz,CDCl3)δ10.74(s,1H),9.37(s,1H),8.53(s,1H),8.43(s,1H),8.18(s,1H),7.94(s,1H),7.42(s,1H),7.28(s,2H),7.26–7.19(m,2H),3.99(s,3H),3.96(s,3H),3.31(dt,J=13.5,6.9Hz,1H),3.11–3.01(m,1H),2.86(t,J=6.4Hz,2H),2.76–2.69(m,1H),2.65(t,J=6.6Hz,2H),2.52(dt,J=13.5,6.9Hz,1H),2.40(d,J=14.8Hz,6H),2.34(s,6H).MS(ESI)m/z:564.27[M+H].
Example 19: synthesis of N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-fluoro-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide:
Figure BDA0001109816280000261
step 1: synthesis of N- (2, 4-difluoro-5-nitrophenyl) -4- (1-methyl-1H-indol-3-yl) pyrimidin-2-amine:
the compound 3- (2-chloropyrimidin-4-yl) -1-methylindole (300mg,1.23mmol) and methyl 4-amino-2-nitro-5- (trifluoromethoxy) benzoate (257mg,1.48mmol) were charged into a reaction flask, followed by addition of 1, 4-dioxane (10mL), addition of p-toluenesulfonic acid (254mg,1.48mmol) with stirring, heating to 85 ℃ for 10 hours, reaction completion, cooling to room temperature, precipitation of a solid, filtration, washing of the filter cake with acetonitrile, and drying to give a yellow solid (460mg, yield: 98.08%).1H NMR(400MHz,CDCl3)δ9.59(s,1H),8.32(d,J=5.7Hz,1H),8.19(s,1H),8.10(d,J=7.9Hz,1H),7.82(d,J=8.0Hz,1H),7.42(d,J=8.0Hz,1H),7.35-7.28(m,2H),7.18(d,J=8.0Hz,1H),7.12(t,J=10.1Hz,1H),3.94(s,3H).
Step 2: n is a radical of1- (2- (dimethylamino) ethyl) -5-fluoro-N1-methyl-N4Synthesis of- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -2-nitrophenyl-1, 4-diamine:
the compound N- (2, 4-difluoro-5-nitrophenyl) -4- (1-methyl-1H-indol-3-yl) pyrimidin-2-amine (200mg,0.52mmol) was added to a reaction flask, followed by DMF (5mL) and N added with stirring1,N1,N2-trimethylethane-1, 2-diamine (53mg,0.52mmol), stirred at room temperature for reaction for 10 hours, after completion of the reaction, the reaction mixture was poured into water, extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated. Separating by column chromatography to obtain target compound (90mg, yield: 38.18%).1H NMR(400MHz,CDCl3)δ9.42(d,J=8.7Hz,1H),8.39(d,J=5.3Hz,1H),8.26–8.18(m,1H),8.09(s,1H),7.40(s,1H),7.32(m,2H),7.22(d,J=5.4Hz,2H),7.01(d,J=12.9Hz,1H),3.93(s,3H),3.39(m,2H),2.86(m,5H),2.50(s,6H).
And step 3: n is a radical of1- (2- (dimethylamino) ethyl) -5-fluoro-N1-methyl-N4Synthesis of- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) benzene-1, 2, 4-triamine:
compound N1- (2- (dimethylamino) ethyl) -5-fluoro-N1-methyl-N4- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -2-nitrobenzene-1, 4-diamine (70mg,0.15mmol), iron powder (46mg,0.83mmol) and ammonium chloride (65mg,1.21mmol) were added to a reaction flask, followed by EtOH/H2O-4/1 (8mL) solution was refluxed for 2 hours, cooled, filtered through a silica gel sheet, washed with dichloromethane, extracted with dichloromethane, dried over anhydrous sodium sulfate, and concentrated to obtain the objective compound (60mg, yield: 91.64%).1H NMR(400MHz,MeOD)δ8.41(d,J=8.1Hz,1H),8.22(d,J=5.5Hz,1H),8.07(d,J=2.9Hz,1H),7.56(d,J=8.2Hz,1H),7.45(d,J=8.1Hz,1H),7.32–7.23(m,2H),7.20–7.15(m,1H),7.02–6.97(m,1H),3.90(s,3H),3.12(t,J=6.5Hz,2H),2.70(s,5H),2.49(s,6H).
And 4, step 4: synthesis of N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-fluoro-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide:
a solution of acryloyl chloride (13mg,0.14mmol) in methylene chloride was added dropwise to N cooled in an ice-water bath1- (2- (dimethylamino) ethyl) -5-fluoro-N1-methyl-N4- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) benzene-1, 2, 4-triamine (60mg,0.14mmol) and DIPEA (20mg,15mmol) in a stirred mixture of dichloromethane (2 mL). After 30 minutes of the reaction, it was diluted with dichloromethane, washed with saturated sodium bicarbonate, the aqueous phase was extracted twice with dichloromethane, the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give the objective compound (25mg, yield: 37.05%). 1H NMR(400MHz,MeOD)δ8.54(d,J=8.4Hz,1H),8.33(d,J=8.0Hz,1H),8.29(d,J=5.5Hz,1H),8.20(s,1H),7.47(d,J=8.1Hz,1H),7.30–7.21(m,2H),7.18(t,J=7.2Hz,1H),6.64–6.53(m,1H),6.49(dd,J=16.9,1.9Hz,1H),6.28–6.09(m,1H),5.88(dd,J=9.8,1.9Hz,1H),3.92(s,3H),3.40(t,J=5.9Hz,2H),3.19(t,J=5.9Hz,2H),2.81(s,6H),2.75(s,3H).MS(ESI)m/z:488.25[M+H].
Synthesis of methyl 4-amino-5-methoxy-2-nitrobenzoate:
concentrated sulfuric acid (20mL) was charged into a reaction flask, and the compound methyl 4-amino-3-methoxybenzoate (5.0g,27.6mmol) was added to the reaction mixture under ice bath conditions, after complete dissolution, potassium nitrate (3.35g,33.11mmol) was added in portions, and after reaction for 30 minutes in ice water bath, the reaction mixture was slowly poured into ice water, extracted with ethyl acetate, and the organic phase was washed successively with a saturated sodium bicarbonate solution, a saturated saline solution, dried over anhydrous sodium sulfate, and concentrated to give a yellow solid (5.8g, yield: 93%).1H NMR(400MHz,CDCl3):7.12(1H,s),7.10(1H,s),4.37(2H,s),3.97(3H,s),3.88(3H,s)。
Example 20: cell viability assay
1. Cell: NCI-H1975, PC-9 and A431, all from the Shanghai cell Bank of Chinese academy of sciences.
2. Reagent: hyclone DMEM high-sugar medium, RPMI 1640 medium, MEM medium, Tryple, MTT (5mg/mL), DMSO, DPBS.
3. The instrument comprises the following steps: 37 ℃ and 5% CO2Incubator, UTRAO enzyme-linked immunosorbent assay (ELISA) reader, biological safety cabinet, cell counting plate, and ott optical microscope.
4. Experiment consumables: stock number of 96-well plate: 3599, 96-hole round bottom dispensing plate.
Activity testing of NCI-H1975 cells Experimental procedure:
1. plate paving: cells in logarithmic growth phase were digested with Tryple, terminated with fresh medium, counted, adjusted to 11111 cells/mL with fresh medium, 90 μ L per well, and the other edges filled with sterile DPBS.
2. At 37 ℃ with 5% CO2Incubate in incubator for 24 hours to allow cells to fill about 50% of the bottom of the well.
3. The experimental group is prepared as follows: dissolving the drug in DMSO to prepare 20mmol/L stock solution, diluting with DMSO to prepare 2mmol/L, sequentially diluting 3 times and 8 concentration gradients to obtain 200 × compound gradient solution, adding 10 μ L gradient compound solution into 190 μ L RPMI1640 culture medium to obtain 10 × gradient compound solution, adding 10 μ L10 × compound solution into 90 μ L96-well cell culture plate, repeating three gradients, wherein the concentration gradients of the compound in the 96-well cell culture plate are 0.05080526nM, 1.524158nM, 4.572474nM, 13.717420nM, 41.152260nM, 123.456800nM, 370.370400nM, 1111.111000nM, 3333.333000nM and 10000.000000nM, each well is 100 μ L, and the final concentration of DMSO is 0.5%.
The control group, containing the same volume of solvent as the experimental group, was diluted with complete medium at 100. mu.L per well.
4. At 37 ℃ in 5% CO2Incubate in incubator for 72 h.
After 5.72h, 20. mu.L of MTT solution (5mg/mL) was added to each well and incubation was continued for 4 h.
6. The culture was terminated and the culture medium in the wells was carefully aspirated.
7. And (3) zeroing the wells, adding 150 mu L of dimethyl sulfoxide (DMSO) into each well of the experimental group and the control group, oscillating at a medium speed for 10s in a microplate reader, fully dissolving crystals, and measuring the light absorption value at the position of 492 nm.
IC of all Compounds 1 to 1450The values are shown in Table 1.
Activity testing of PC-9 cells Experimental procedure:
1. plate paving: cells in logarithmic growth phase were digested with Tryple, terminated with fresh medium, counted, adjusted to 11111 cells/mL with fresh medium, 90 μ L per well, and the other edges filled with sterile DPBS.
2. At 37 ℃ with 5% CO2Incubate in incubator for 24 hours to allow cells to fill about 50% of the bottom of the well.
3. The experimental group is prepared as follows: dissolving the drug in DMSO to prepare 20mmol/L stock solution, diluting with DMSO to prepare 2mmol/L, sequentially diluting 3 times and 8 concentration gradients to obtain 200 × compound gradient solution, adding 10 μ L gradient compound solution into 190 μ L RPMI1640 culture medium to obtain 10 × gradient compound solution, adding 10 μ L10 × compound solution into 90 μ L96-well cell culture plate, repeating three gradients, wherein the concentration gradients of the compound in the 96-well cell culture plate are 0.05080526nM, 1.524158nM, 4.572474nM, 13.717420nM, 41.152260nM, 123.456800nM, 370.370400nM, 1111.111000nM, 3333.333000nM and 10000.000000nM, each well is 100 μ L, and the final concentration of DMSO is 0.5%.
The control group, containing the same volume of solvent as the experimental group, was diluted with complete medium at 100. mu.L per well.
4. At 37 ℃ in 5% CO2Incubate in incubator for 72 h.
After 5.72h, 20 μ of LMTT solution (5mg/mL) was added to each well and incubation continued for 4 h.
6. The culture was terminated and the culture medium in the wells was carefully aspirated.
7. And (3) zeroing the wells, adding 150 mu L of dimethyl sulfoxide (DMSO) into each well of the experimental group and the control group, oscillating at a medium speed for 10s in a microplate reader, fully dissolving crystals, and measuring the light absorption value at the position of 492 nm.
IC of all Compounds 1 to 1450The values are shown in Table 1.
Activity test of a431 cells experimental procedure:
1. plate paving: cells in logarithmic growth phase were digested with Tryple, terminated with fresh medium, counted, adjusted to 11111 cells/mL with fresh medium, 90 μ L per well, and the other edges filled with sterile DPBS.
2. At 37 ℃ with 5% CO2Incubate in incubator for 24 hours to allow cells to fill about 50% of the bottom of the well.
3. The experimental group was dosed. Dissolving the drug in DMSO to prepare 20mmol/L stock solution, diluting with DMSO to prepare 2mmol/L, sequentially diluting 3 times and 8 concentration gradients to obtain 200 × compound gradient solution, adding 10 μ L gradient compound solution into 190 μ L RPMI1640 culture medium to obtain 10 × gradient compound solution, adding 10 μ L10 × compound solution into 90 μ L96 well cell culture plate, repeating three gradients, wherein the concentration gradients of the compound in 96 well cell culture plate are 0.05080526nM, 1.524158nM, 4.572474nM, 13.717420nM, 41.152260nM, 123.456800nM, 370.370400nM, 1111.111000nM, 3333.333000nM and 10000.000000nM, each well is 100 μ L, and the final concentration of DMSO is 0.5%.
The control group, containing the same volume of solvent as the experimental group, was diluted with complete medium at 100. mu.L per well.
4. At 37 ℃ in 5% CO2Incubate in incubator for 72 h.
After 5.72 h, 20. mu.L of MTT solution (5mg/mL) was added to each well and incubation was continued for 4 h.
6. The culture was terminated and the culture medium in the wells was carefully aspirated.
7. And (3) zeroing a hole, adding 150 mu L of dimethyl sulfoxide (DMSO) into each hole of the experimental group and the control group, oscillating at a medium speed of a microplate reader for 10s, fully dissolving a crystal, and measuring the light absorption value at the wavelength of 492 nm.
IC of all Compounds 1 to 1450The values are shown in Table 1:
TABLE 1 results of cell Activity assays for Compounds 1-14
Figure BDA0001109816280000281
Figure BDA0001109816280000291
- -means no detection.

Claims (10)

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof,
Figure FDA0003104327230000011
wherein the content of the first and second substances,
R1selected from: hydrogen, deuterium, and halogen;
R2selected from: halogen, C1-6Alkyl, halogen substituted C1-6Alkyl and-OR13Wherein R is13Selected from halogen substituted C1-6An alkyl group;
R3selected from: -C (O) R6、-SO-R6and-SO2-R6Wherein R is6Is- (CH)2)PNR10R11and-NR12(CH2)PNR10R11Wherein p is selected from an integer of 1 to 6, R10、R11And R12Independently selected from H and C1-3An alkyl group;
R4is H; and R5is-C (O) R6Wherein R is6Is C2-6An alkenyl group;
g is
Figure FDA0003104327230000012
Wherein X is CH; and R8Selected from: hydrogen, halogen substituted or unsubstituted C1-3Alkyl and C3-6A cycloalkyl group; and R9Is hydrogen.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R2Is halogen.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R3Selected from: -C (O) R6、-SO-R6and-SO2-R6Wherein R is6Is selected from-N (CH)3)CH2CH2N(CH3)2and-CH2CH2N(CH3)2
4. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R4is-C (O) R6Wherein R is6Is a vinyl group.
5. A compound of formula (II) or a pharmaceutically acceptable salt thereof,
Figure FDA0003104327230000013
wherein the content of the first and second substances,
R1selected from: hydrogen, deuterium, and halogen;
R2selected from: halogen;
R3is-NR6R7Wherein R is6Is selected from- (CH)2)PNR10R11Wherein p is selected from an integer of 1 to 6, R10And R11Independently selected from H and C1-3An alkyl group; and R7Independently selected from: hydrogen and C1-3An alkyl group;
R4is H; and R5is-C (O) R6Wherein R is6Is C2-6An alkenyl group;
g is
Figure FDA0003104327230000014
Wherein X is CH; and R8Selected from: hydrogen or C1-3An alkyl group; and R9Is hydrogen.
6. The compound of claim 5, or a pharmaceutically acceptable salt thereof, wherein R3is-N (CH)3)CH2CH2N(CH3)2
7. A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure FDA0003104327230000015
8. a pharmaceutical composition comprising a compound according to any one of claims 1 to 7 and pharmaceutically acceptable salts thereof.
9. Use of a compound according to any one of claims 1 to 7, and pharmaceutically acceptable salts thereof, and a pharmaceutical composition according to claim 8, for the manufacture of a medicament for the treatment and/or prevention of a disorder or disease mediated by the activated or resistant mutant form of EGFR.
10. The use of claim 9, wherein the EGFR-mediated disorder or disease in the form of an activating or resistant mutant is ovarian cancer, cervical cancer, colorectal cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, melanoma, prostate cancer, leukemia, lymphoma, non-hodgkin lymphoma, gastric cancer, lung cancer, hepatocellular carcinoma, gastrointestinal stromal tumor, thyroid cancer, cholangiocarcinoma, endometrial cancer, renal cancer, anaplastic large cell lymphoma, acute myelogenous leukemia, multiple myeloma, or mesothelioma.
CN201610803436.3A 2016-09-05 2016-09-05 Pyrimidine heterocyclic compound and preparation method and application thereof Active CN107793413B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201610803436.3A CN107793413B (en) 2016-09-05 2016-09-05 Pyrimidine heterocyclic compound and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201610803436.3A CN107793413B (en) 2016-09-05 2016-09-05 Pyrimidine heterocyclic compound and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN107793413A CN107793413A (en) 2018-03-13
CN107793413B true CN107793413B (en) 2021-09-28

Family

ID=61529855

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201610803436.3A Active CN107793413B (en) 2016-09-05 2016-09-05 Pyrimidine heterocyclic compound and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN107793413B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11851426B2 (en) 2019-10-11 2023-12-26 Incyte Corporation Bicyclic amines as CDK2 inhibitors

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107935995A (en) * 2017-11-28 2018-04-20 中山大学 A kind of new 2 anilino-pyrimidine derivative and its application in antitumor drug is prepared
US11066404B2 (en) 2018-10-11 2021-07-20 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
CN111574503A (en) * 2019-02-15 2020-08-25 药雅科技(上海)有限公司 Pyrimidine double aromatic ring derivative epidermal growth factor inhibitor and preparation method and application thereof
WO2020168197A1 (en) 2019-02-15 2020-08-20 Incyte Corporation Pyrrolo[2,3-d]pyrimidinone compounds as cdk2 inhibitors
CN111620854A (en) * 2019-02-28 2020-09-04 上海医药工业研究院 4-aminopyrimidine substituted phenylamide compound and preparation method thereof
CN111617083B (en) * 2019-02-28 2023-10-27 上海医药工业研究院 Application of methoxy substituted phenylamide aminopyrimidine derivative
WO2020180959A1 (en) 2019-03-05 2020-09-10 Incyte Corporation Pyrazolyl pyrimidinylamine compounds as cdk2 inhibitors
EP3941905A1 (en) * 2019-03-19 2022-01-26 Suvalent Therapeutics, Inc. Sumo inhibitor compounds and uses thereof
US11919904B2 (en) 2019-03-29 2024-03-05 Incyte Corporation Sulfonylamide compounds as CDK2 inhibitors
CN111777599A (en) * 2019-04-03 2020-10-16 药雅科技(上海)有限公司 Quinazoline, pyridopyrimidine or bipyrimidine bi-aromatic ring derivative EGFR inhibitor and preparation method and application thereof
US11447494B2 (en) 2019-05-01 2022-09-20 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
US11440914B2 (en) 2019-05-01 2022-09-13 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
CN111909101B (en) * 2019-05-10 2022-07-19 浙江大学 EGFR kinase inhibitor and application thereof in preparation of anti-cancer drugs
CA3138648A1 (en) * 2019-05-22 2020-11-26 Shanghai Hansoh Biomedical Co., Ltd. Indole derivative-containing inhibitor, preparation method therefor and application thereof
CN116348458A (en) 2019-08-14 2023-06-27 因赛特公司 Imidazolylpyrimidinylamine compounds as CDK2 inhibitors
CN110357863B (en) * 2019-08-27 2023-04-14 药雅科技(上海)有限公司 Triazine double aromatic ring derivative epidermal growth factor inhibitor and preparation method and application thereof
CN111057073A (en) * 2019-12-26 2020-04-24 浙江工业大学 4-indole-2-arylamino pyrimidine compound and application thereof in inflammation treatment
CN113121528B (en) * 2020-01-15 2022-12-13 中国科学院上海药物研究所 Multi-target inhibition compound, composition, functional molecule and application thereof
CN113354622B (en) * 2020-03-06 2022-11-01 沈阳药科大学 P-phenylenediamine LSD1 inhibitor and preparation method thereof
WO2021243596A1 (en) * 2020-06-03 2021-12-09 InventisBio Co., Ltd. Aminopyrimidine compounds, preparation methods and uses thereof
CN113563310B (en) * 2021-06-25 2022-10-11 浙江工业大学 4- (1-methylindol-3-yl) pyrimidine derivative and application thereof
CN113387935B (en) * 2021-07-23 2022-06-10 苏州雅深智慧科技有限公司 Compound for inhibiting triple-mutation epidermal growth factor receptor tyrosine kinase and application thereof
CN113620809B (en) * 2021-08-13 2023-11-14 南京信息工程大学 Recycling treatment method of industrial wastewater generated in production of 5-chloro-2-nitrobenzotrifluoride
CA3229553A1 (en) * 2021-09-02 2023-03-09 Synthon B.V. A process for making osimertinib
CN114805304A (en) * 2022-04-19 2022-07-29 辽宁大学 4-methoxyphenyl-1, 3-diamine derivatives containing 1-methyl-1H-indole structure and application thereof
CN115974845A (en) * 2023-01-19 2023-04-18 奥锐特药业股份有限公司 Preparation method of oxitinib intermediate

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105646454A (en) * 2016-01-19 2016-06-08 浙江大学 2-arylamine pyrimidine derivatives containing hydroxamic acid fragments and preparation and application

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104860941B (en) * 2014-02-25 2017-03-22 上海海雁医药科技有限公司 2,4-disubstituted phenyl-1,5-diamine derivatives and use thereof, and pharmaceutical composition and medicinal composition prepared from 2,4-disubstituted phenyl-1,5-diamine derivative
CA2949793C (en) * 2014-06-19 2024-02-27 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
BR112017006713B1 (en) * 2014-10-11 2023-11-21 Shanghai Hansoh Biomedical Co., Ltd. EGFR INHIBITORS, THEIR USES AND THEIR PREPARATION PROCESS, AND PHARMACEUTICAL COMPOSITION

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105646454A (en) * 2016-01-19 2016-06-08 浙江大学 2-arylamine pyrimidine derivatives containing hydroxamic acid fragments and preparation and application

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11851426B2 (en) 2019-10-11 2023-12-26 Incyte Corporation Bicyclic amines as CDK2 inhibitors

Also Published As

Publication number Publication date
CN107793413A (en) 2018-03-13

Similar Documents

Publication Publication Date Title
CN107793413B (en) Pyrimidine heterocyclic compound and preparation method and application thereof
CN106661000B (en) EGFR inhibitor and its preparation and application
TWI597268B (en) Fgfr4 inhibitors
TWI618704B (en) Substituted 2-anilinopyrimidine derivatives as egfr modulators
KR101904632B1 (en) 2,4-Disubstituted phenylene-1,5-Diamine Derivatives and Applications Thereof, and Pharmaceutical Compositions and Pharmaceutically Acceptable Compositions Prepared Therefrom
JP5414794B2 (en) Amidophenoxyindazoles useful as inhibitors of C-MET
CN109310671B (en) Bruton's tyrosine kinase inhibitors
KR102051609B1 (en) 2-arylaminopyridine, pyrimidine or triazine derivatives and methods for their preparation and use
ES2768694T3 (en) Quinolinone pyrimidine compositions as mutant isocitrate dehydrogenase inhibitors
CN108349896B (en) Heterocyclic compounds as FGFR inhibitors
JP2009527562A (en) Cinnoline derivatives as phosphodiesterase 10 inhibitors
KR20170063734A (en) Mk2 inhibitors and uses thereof
CN105315285A (en) 2,4-disubstituted 7H-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medical uses thereof
CN113748114A (en) Quinazoline compound and application thereof in medicine
WO2022166974A1 (en) Pyridopyrimidinone derivative, preparation method therefor, and use thereof
CN114728926A (en) ROCK inhibitor and preparation method and application thereof
WO2020233669A1 (en) Indole derivative-containing inhibitor, preparation method therefor and application thereof
CN112313207B (en) Cyano-substituted pyridine and cyano-substituted pyrimidine compounds, and preparation methods and applications thereof
WO2022017408A1 (en) Arylamine derivative and preparation method therefor and medical use thereof
WO2019196720A1 (en) Arginine methyltransferase inhibitor, pharmaceutical composition thereof and use thereof
WO2022100738A1 (en) Crystal form of free base of inhibitor containing bicyclic ring derivative and preparation method and application of crystal form
WO2022033455A1 (en) Triazine derivative having egfr inhibitory activity, preparation method therefor and use thereof
KR20150105302A (en) Cyclic sulfonamide containing derivatives as inhibitors of hedgehog signaling pathway
WO2022033410A1 (en) Efgr inhibitor, preparation method therefor, and application thereof
WO2024022286A1 (en) Macrocyclic egfr inhibitor, and preparation method therefor and pharmaceutical use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right

Effective date of registration: 20210222

Address after: 201203, block m, 3 / F, 780 Cailun Road, Zhangjiang High Tech Park, Pudong New Area, Shanghai, 200120

Applicant after: KECHOW PHARMA, Inc.

Address before: N1701, 17th floor, experimental building, Tianjin International Joint Research Institute of biomedicine, 220 Dongting Road, Binhai New Area Development Zone, Tianjin, 300457

Applicant before: TIANJIN BINJIANG PHARMACEUTICAL Inc.

TA01 Transfer of patent application right
GR01 Patent grant
GR01 Patent grant