CN109608444B - Isoindolinone-containing ERK inhibitor and preparation method and application thereof - Google Patents

Isoindolinone-containing ERK inhibitor and preparation method and application thereof Download PDF

Info

Publication number
CN109608444B
CN109608444B CN201811424530.3A CN201811424530A CN109608444B CN 109608444 B CN109608444 B CN 109608444B CN 201811424530 A CN201811424530 A CN 201811424530A CN 109608444 B CN109608444 B CN 109608444B
Authority
CN
China
Prior art keywords
arh
compound
acid
dichloromethane
pharmaceutically acceptable
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
CN201811424530.3A
Other languages
Chinese (zh)
Other versions
CN109608444A (en
Inventor
徐云根
纪德重
张灵芝
朱启华
柏英
吴尧尧
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.)
China Pharmaceutical University
Original Assignee
China Pharmaceutical University
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 China Pharmaceutical University filed Critical China Pharmaceutical University
Priority to CN201811424530.3A priority Critical patent/CN109608444B/en
Publication of CN109608444A publication Critical patent/CN109608444A/en
Priority to PCT/CN2019/104110 priority patent/WO2020107987A1/en
Application granted granted Critical
Publication of CN109608444B publication Critical patent/CN109608444B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

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

Abstract

The invention discloses isoindolinone derivatives, in particular compounds containing a 6- (2-aminopyrimidine-4-yl) isoindoline-1-one structure. The invention also discloses a preparation method of the isoindolinone derivative. The invention also discloses the isoindolinone derivatives, stereoisomers, hydrates, solvates or crystals thereof, application of a composition containing the isoindolinone derivatives or pharmaceutically acceptable salts thereof and pharmaceutically acceptable carriers in preparation of ERK kinase inhibitors, and application of a pharmaceutical composition containing the compounds in preparation of antitumor drugs.

Description

Isoindolinone-containing ERK inhibitor and preparation method and application thereof
Technical Field
The invention belongs to the pharmaceutical chemistry technology, and particularly relates to ERK kinase inhibitors containing 6- (2-aminopyrimidine-4-yl) isoindoline-1-ketone structures, preparation methods thereof, pharmaceutical compositions containing the compounds and application thereof in preparing antitumor drugs.
Technical Field
The Mitogen-activated protein kinases (MAPKs) signal transduction pathway is an important signal pathway for transducing cell surface signals to cell nucleus, and the pathway influences the transcription and regulation of genes in animal cells, thereby causing biological reactions such as cell proliferation, differentiation, transformation, apoptosis and the like. Currently, 5 MAPKs signaling pathways are commonly found in mammalian cells, including Extracellular signal-regulated kinases (ERK), c-Jun N-terminal kinases (JNK/SAPK, c-Jun N-terminal kinase/stress-activated protein kinase), p38 kinase isozymes (p38A, p38B, p38C, and p38D), ERK3/ERK4, and ERK 5.
The RAS-RAF-MEK-ERK signal transduction pathway (ERK pathway) is an evolutionarily conserved signal cascade pathway, which can transfer to cell surface receptor signals, thereby promoting cell proliferation and survival. The ERK signaling pathway plays an important role in maintaining cell stability and regulating cell growth under normal physiological conditions, and is tightly controlled by a multi-stage feedback regulatory pathway.
To date, BRAF inhibitors and MEK inhibitors have met with great success in the anti-tumor field, but with the development of clinical applications, it has been found that most patients will again worsen within one year (acquired resistance) whether BRAF inhibitors are used alone or in combination with MEK inhibitors. In addition, about 10% to 15% of the carrier carries B-RafV600EMutant tumor patients are not sensitive to BRAF inhibitors as well as MEK inhibitors (intrinsic resistance). Therefore, the problem of resistance to BRAF and MEK inhibitors is a key scientific problem that needs to be addressed at present.
Compared with the BRAF and MEK inhibitors, the ERK inhibitor firstly blocks the reactivation of the MAPKs expressed by the tumor cells resistant to the BRAF and MEK inhibitors again, and overcomes the existing drug resistance problem. Secondly, tumors inevitably develop drug-resistant mutations, such as the numerous BRAF and MEK mutations mentioned above, over time through drug treatment with targeted kinases. However, to date, little mutation in ERK1/2 was observed in tumor cells. Meanwhile, preclinical experimental results of ERK inhibitors show better effects compared to BRAF and MEK inhibitors. Therefore, compared with inhibiting the upstream kinase of ERK, the ERK inhibitor can block the signal transduction of the ERK pathway more effectively, and overcome the drug resistance of tumor cells to BRAF inhibitors and MEK inhibitors.
Disclosure of Invention
The purpose of the invention is as follows: the invention provides an isoindolinone derivative containing a 6- (2-aminopyrimidine-4-yl) isoindoline-1-one structure, a specific preparation method of the derivative and pharmaceutical application of the derivative in preparing an ERK kinase inhibitor.
The technical scheme is as follows: the invention discloses an isoindolinone derivative shown as a general formula I or pharmaceutically acceptable salt thereof:
Figure BDA0001881258540000011
wherein:
R1is selected from
Figure BDA0001881258540000021
Figure BDA0001881258540000022
Wherein X is selected from CH2O, NH or CH-OH; r4Selected from H or C1-C6 alkyl, R5Selected from H, CH3、-CH2OH、-CH2CH2OH、-CH2CH2CH2OH or-CH2CH2CH2CH2OH; y is selected from CH2O, NH or S; r6Selected from F, Cl, Br, CH3、NH2Or NHCOCH3
R2Is H or-CH2OH;
R3Is selected from
Figure BDA0001881258540000023
Wherein R is7Is H, F, Cl, Br or OCH3,R7Is mono-, di-or tri-substituted; r8Is H or CH3
Preferably, R1Is selected from
Figure BDA0001881258540000024
Figure BDA0001881258540000025
Preferably, R3Is selected from
Figure BDA0001881258540000026
When R is in the specification2is-CH2OH, the compound I or a pharmaceutically acceptable salt thereof includes chiral isomers:
Figure BDA0001881258540000027
preferably, the isoindolinone derivatives described herein are selected from the group consisting of I-1 to I-28:
Figure BDA0001881258540000028
Figure BDA0001881258540000031
Figure BDA0001881258540000041
Figure BDA0001881258540000051
the pharmaceutically acceptable salt is an acid addition salt of the compound shown in the general formula I, wherein the acid for forming the salt is: hydrogen chloride, hydrogen bromide, sulfuric acid, carbonic acid, oxalic acid, citric acid, succinic acid, tartaric acid, phosphoric acid, lactic acid, pyruvic acid, acetic acid, maleic acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid or ferulic acid.
The compounds of the general formula (I) according to the invention can be prepared by the following process:
the preparation method of the compound of the general formula (I) comprises the following steps:
Figure BDA0001881258540000061
wherein R is1Represents:
Figure BDA0001881258540000062
wherein X represents CH2、O、CH-OH;R4Represents an alkyl group of H, C1 to C6, R5Representative H, CH3、-CH2OH、-CH2CH2OH、-CH2CH2CH2OH or-CH2CH2CH2CH2OH;R2Represents: h or-CH2OH;R3Represents:
Figure BDA0001881258540000063
Figure BDA0001881258540000064
when R is2=CH2OH, compound I configuration includes: s configuration, R configuration and a mixed racemate of the two configurations.
Further, the compound IX is 1-methyl-1H-pyrazol-4-amine, 1-methyl-1H-pyrazol-3-amine, 4-aminotetrahydropyran, 4-aminocyclohexanol, 4-aminopiperidine, 3-aminopyrrolidine, ethanolamine, cyclohexylamine, 3-amino-1-propanol, isopropylamine, 4-aminopyridine; the solvent is selected from sec-butyl alcohol, tert-butyl alcohol, ethanol, tetrahydrofuran, ethyl acetate, methanol or a mixed solvent of any two or three of the solvents, preferably sec-butyl alcohol; the reaction temperature is selected from 90-150 ℃, preferably 110-130 ℃; the reaction time is 4-48 h, and the reaction time is preferably 8-24 h. The compound VIII: the molar ratio of the compound IX is 1: 1-1: 20, preferably 1: 1.5-1: 3.
A second process for the preparation of a compound of formula (I) comprising the steps of:
Figure BDA0001881258540000065
reacting a compound VIII with tert-butyldimethylsilyl chloride TBSCl to prepare a compound X; then preparing a compound XI from the compound X and IX through substitution reaction; finally, removing a hydroxyl protecting group from a compound XI and then preparing a salt to prepare a compound I.A.
Wherein R is1Represents:
Figure BDA0001881258540000066
R3represents:
Figure BDA0001881258540000067
Figure BDA0001881258540000068
the configuration of the compound I.A comprises: s configuration, R configuration and a mixed racemate of the two configurations.
It is specifically mentioned that the second preparation process described above is also suitable for R2=H。
Process for the preparation of compound X by reaction of compound VIII with tert-butyldimethylsilyl chloride (TBSCl): the base is selected from triethylamine, 4-dimethylaminopyridine, imidazole, sodium carbonate or potassium carbonate, preferably imidazole; the reaction solvent is preferably ethyl acetate, tetrahydrofuran, chloroform, acetonitrile, dichloromethane, toluene or a mixed solvent of any two or three of the solvents, preferably dichloromethane; the temperature is selected from 0 ℃ to 50 ℃, preferably 30 ℃ to 40 ℃; the reaction time is selected from 1h to 12h, preferably 7h to 9 h; compound VIII: the molar ratio of tert-butyldimethylsilyl chloride (TBSCl) is 1: 1-1: 10, preferably 1: 1.5-1: 2.
Preparing a compound XI from a compound X and an IX through a substitution reaction, wherein the compound IX is 2-aminothiazole, 2-aminooxazole, 1-methyl-5-aminotetrazole, 1-methyl-1H-3-aminopyrazole or 1-methyl-1H-5-aminopyrazole; the base used is selected from lithium bis (trimethylsilyl) amide, sodium bis (trimethylsilyl) amide or sodium hydride, preferably lithium bis (trimethylsilyl) amide; the solvent is tetrahydrofuran, N-dimethylformamide, dioxane, N-methylpyrrolidone or a mixed solvent of any two or three of the solvents, preferably tetrahydrofuran and N, N-dimethylformamide; the reaction temperature is selected from-80 ℃ to 0 ℃, preferably from-80 ℃ to-25 ℃; the reaction time is selected from 0.1h to 10h, preferably 1h to 3 h; compound X: compound IX: the ratio of the alkali (molar ratio) is 1:1: 1-1: 6:6, preferably 1:1.5: 1.5-1: 2: 2.
The process for preparing the compound I.A by removing a hydroxyl protecting group from a compound XI and then forming a salt, wherein the used deprotection reagent (A) is selected from hydrogen chloride, hydrogen bromide, sulfuric acid or trifluoroacetic acid; the solvent is methanol, ethanol, dichloromethane, acetone, ethyl acetate, tetrahydrofuran or a mixed solvent of any two of the above solvents; the reaction temperature is selected from 0 ℃ to 50 ℃, preferably 0 ℃ to 25 ℃; the reaction time is selected from 0.1h to 6h, preferably 1h to 3 h.
The first preparation method of the key intermediate VIII in the preparation method of the general formula (I) comprises the following steps:
Figure BDA0001881258540000071
wherein R is2Represents: h or-CH2OH;R3Represents:
Figure BDA0001881258540000072
wherein R is7Represents H, F, Cl, Br or OCH3,R7May be mono-, di-or tri-substituted; r8Represents H or CH3. When R is2=CH2OH, compound VIII configuration includes: s configuration, R configuration and a mixed racemate of the two configurations.
Procedure for preparation of compound IV from compound II and compound III by Suzuki coupling reaction: the catalyst used is selected from palladium tetratriphenylphosphine (Pd (PPh)3)4) [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride (Pd (dppf) Cl2) Bis (triphenylphosphine) palladium dichloride (Pd (PPh3)2Cl2) Palladium acetate (Pd (OAc)2) Or (1,1' -bis (diphenylphosphino) ferrocene) nickel dichloride (NiCl)2(dppf)), preferably palladium tetratriphenylphosphine (Pd (PPh)3)4). The base is selected from sodium ethoxide, sodium acetate, potassium phosphate, sodium carbonate or potassium carbonate, preferably sodium carbonate; the reaction solvent is selected from N, N-dimethylformamide, N-dimethylacetamide, ethylene glycol dimethyl ether, 1, 4-dioxane, tetrahydrofuran, toluene, ethanol, water or a mixed solution of any two or three of the solventsAn agent, preferably a toluene/methanol/water mixed solvent; the reaction temperature is selected from 60 ℃ to 120 ℃, and preferably 70 ℃ to 80 ℃; the reaction time is selected from 4 to 24 hours, preferably 7 to 10 hours; compound II: compound III: catalyst: the ratio of the alkali (molar ratio) is 1:1:0.02: 1-6: 1:0.2:6, preferably 1:1:0.03: 1-1: 1.05:0.1: 2.
Process for preparing compound V by oxidation of compound IV: the oxidant is selected from m-chloroperoxybenzoic acid (m-CPBA), hydrogen peroxide, sodium hypochlorite, periodic acid, sodium periodate, potassium permanganate or tert-butyl peroxy alcohol, preferably m-chloroperoxybenzoic acid; the reaction solvent is selected from tetrahydrofuran, acetone, dichloromethane, 1, 4-dioxane or acetonitrile, preferably dichloromethane; the reaction temperature is selected from 10 ℃ to 80 ℃, preferably 20 ℃ to 30 ℃; the reaction time is selected from 0.5 h-5 h, preferably 1 h-2 h; compound IV: the molar ratio of the oxidant is 1: 1-1: 10, preferably 1: 1.5-1: 3.
Process for preparing compound VI from compound V by bromination: the brominating agent used is selected from N-bromosuccinimide (NBS) or liquid bromine, preferably N-bromosuccinimide; the solvent is selected from benzene, carbon tetrachloride, chloroform, acetonitrile, dichloromethane, methanol or toluene, preferably carbon tetrachloride; the reaction temperature is selected from 50 ℃ to 100 ℃, and preferably 70 ℃ to 90 ℃; the reaction time is selected from 1h to 18h, preferably 4h to 7 h; compound V: the ratio of the brominating agent (molar ratio) is 1: 1-1: 20, preferably 1: 1-1: 2.
Process for preparing compound VIII by cyclization of compound VI with compound VII: the base used is selected from triethylamine, benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (PyBop), N-Diisopropylethylamine (DIEA), sodium carbonate or potassium carbonate, preferably triethylamine; the reaction solvent is selected from tetrahydrofuran, acetone, acetonitrile, methanol, dichloromethane or a mixed solvent of any two, preferably acetonitrile; the reaction temperature is selected from 50 ℃ to 100 ℃, preferably 60 ℃ to 90 ℃; the reaction time is selected from 1h to 24h, preferably 5h to 9 h; the molar ratio of the compound VI to the compound VII to the base is 1:1: 1-1: 4:8, preferably 1:1: 1-1: 1.5: 1.5.
The second preparation method of the key intermediate VIII in the preparation method of the general formula (I) comprises the following steps:
Figure BDA0001881258540000081
wherein R is2Represents: h; r3Represents:
Figure BDA0001881258540000082
wherein R is7Represents H, F, Cl, Br or OCH3,R7May be mono-, di-or tri-substituted; r8Represents H or CH3
Procedure for preparation of compound XIII from compound II and compound XII by Suzuki coupling reaction: the reaction is carried out under the condition of adding a catalyst, a base and a reaction solvent, wherein the catalyst is selected from bis (triphenylphosphine) palladium dichloride, tetrakis (triphenylphosphine) palladium or [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride, preferably tetrakis (triphenylphosphine) palladium; the base is selected from sodium ethoxide, sodium acetate, potassium phosphate, sodium carbonate or potassium carbonate, preferably sodium carbonate; the reaction solvent is selected from N, N-dimethylformamide, N-dimethylacetamide, ethylene glycol dimethyl ether, 1, 4-dioxane, tetrahydrofuran, toluene, methanol, ethanol, water or a mixed solvent of any two or three of the solvents, preferably a mixed solvent of toluene/methanol/water; the reaction temperature is selected from 50 ℃ to 120 ℃, preferably 60 ℃ to 80 ℃; the reaction time is selected from 3 to 30 hours, preferably 7 to 12 hours; compound II: compound XII: catalyst: the ratio of the alkali (molar ratio) is 1:1:0.02: 1-6: 1:0.3:10, preferably 1:1:0.05: 1-1: 1.05:0.1: 2.
Process for the preparation of compound XIV from compound XIII by deprotection with a deprotecting agent selected from hydrogen chloride, trifluoroacetic acid or boron trifluoride, preferably trifluoroacetic acid; the reaction solvent is selected from tetrahydrofuran, acetone, dichloromethane, 1, 4-dioxane or acetonitrile, preferably dichloromethane; the reaction temperature is selected from 0 ℃ to 50 ℃, preferably 20 ℃ to 30 ℃; the reaction time is selected from 0.5h to 6h, preferably 1h to 2 h.
Process for the preparation of Compound XVI by substitution reaction of Compound XIV and Compound XV: the base is selected from potassium carbonate, sodium carbonate, potassium phosphate, potassium acetate, potassium hydroxide, sodium hydride or sodium acetate, preferably sodium hydride; the reaction solvent is selected from toluene, methanol, water, 1, 4-dioxane, glycol dimethyl ether or a mixed solvent of any three of the above, preferably 1, 4-dioxane; the reaction temperature is selected from 0 ℃ to 120 ℃, preferably 50 ℃ to 80 ℃; the reaction time is selected from 3 to 24 hours, preferably 9 to 12 hours; the molar ratio of the compound XIV to the compound XV to the base is selected from 1:1:1 to 1:6:6, preferably 1:1:1 to 1:1.2: 1.5.
Procedure for preparation of compound VIII by oxidation of compound XVI: the oxidant is selected from m-chloroperoxybenzoic acid (m-CPBA), hydrogen peroxide, sodium hypochlorite, periodic acid, sodium periodate, potassium permanganate or tert-butyl peroxy alcohol, preferably m-chloroperoxybenzoic acid; the reaction solvent is selected from tetrahydrofuran, acetone, dichloromethane, 1, 4-dioxane or acetonitrile, preferably dichloromethane; the reaction temperature is selected from 10 ℃ to 80 ℃, preferably 20 ℃ to 30 ℃; the reaction time is selected from 0.1h to 10h, preferably 1h to 2 h; compound IV: the proportion of the oxidant (molar ratio) is 1: 1-1: 6, preferably 1: 1.5-1: 3.
The invention also discloses a pharmaceutical composition which contains the compound of the general formula I or pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier. The compound can be added with pharmaceutically acceptable carriers to prepare common pharmaceutical preparations, such as tablets, capsules, syrup and suspending agents, and common pharmaceutical excipients such as spices, sweeteners, liquid or solid fillers or diluents and the like can be added.
The application of the isoindolinone derivative shown in the general formula I and the stereoisomer, hydrate, solvate or crystal thereof in preparing ERK kinase inhibitor drugs is also within the protection scope of the invention.
Furthermore, the ERK kinase inhibitor is used for preparing the medicine for treating malignant tumors.
Has the advantages that: the invention discloses a novel isoindolinone derivative shown in a general formula (I), and pharmacological experiments show that the compound I can generate good inhibition effect on ERK kinase and can be used for preparing a medicament for treating malignant tumors with over-activated ERK pathways; the invention also discloses a preparation method of the isoindolinone derivative.
Drawings
FIG. 1 shows the results of HCT116 nude mouse transplantable tumor experiments with Compound I-8.
Detailed Description
The present application will be described in detail with reference to specific examples.
Example 1
Synthesis of 2- (3-chlorobenzyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-1)
Synthesis of tert-butyl 6- (2- (methylthio) pyrimidin-4-yl) -1-oxoisoindoline-2-carboxylate (XIII)
4-chloro-2- (methylthio) pyrimidine (XII) (6.14g,38.24mmol), tert-butyl 6- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) isoindoline-2-carboxylate (II) (13.74g,38.24mmol), sodium carbonate (8.11g,76.48mmol) and tetratriphenylphosphine palladium (2.21g,1.91mmol) were dissolved in a mixed solvent composed of toluene (150mL), ethanol (50mL) and water (50mL), reacted at 85 ℃ for about 12h under nitrogen protection, after TLC (dichloromethane: methanol ═ 35:1) detection of reaction completion, cooled to room temperature, evaporated to dryness under reduced pressure, the residue was slurried with ethyl acetate (300mL), filtered, the filter cake was washed with ethyl acetate and separated by column chromatography (eluent: dichloromethane: methanol: 100: 1), 12.3g of solid yellow soil is obtained, the yield is 90.0%, and m.p.197.7-199.2 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.60(d,J=5.2Hz,1H,ArH),8.53(s,1H,ArH),8.47(dd,J=8.0,1.6Hz,1H,ArH),7.61(d,J=8.0Hz,1H,ArH),7.44(d,J=5.3Hz,1H,ArH),4.84(s,2H,CH2),2.65(s,3H,SCH3),1.61(s,9H,3×CH3).
Synthesis of 6- (2- (methylthio) pyrimidin-4-yl) isoindolin-1-one (XIV)
Dissolving compound XIII (8.0g,22.4mmol) in dichloromethane (80mL), adding trifluoroacetic acid (20mL), stirring at room temperature for about 1h, stopping the reaction until the starting material reaction is complete as detected by TLC (dichloromethane: methanol ═ 25:1), evaporating the solvent under reduced pressure, adding water (100mL) to the residue, adjusting pH to 6-7 with saturated sodium hydroxide solution, precipitating a white solid, filtering, and drying to obtain 5.5g of a white solid, yield: 95.5%, m.p. >250.0 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):8.69(s,2H,ArH),8.44(s,1H,ArH),7.88(s,1H,ArH),7.74(d,J=7.8Hz,1H,ArH),4.47(s,2H,CH2),2.61(s,3H,CH3).
Synthesis of 2- (3-chlorobenzyl) -6- (2- (methylthio) pyrimidin-4-yl) isoindolin-1-one (XVI-1)
Dissolving compound XIV (2g,7.8mmol) in 1, 4-dioxane (50mL), adding 60% sodium hydride (376mg,9.4mmol), stirring at room temperature for 1h, adding 3-chlorobenzyl bromide (2.4g,11.7mmol), stirring at 70 ℃ for reaction for about 12h, detecting by TLC (petroleum ether: ethyl acetate ═ 2:1) that the reaction of the raw material XIV is complete, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: petroleum ether: ethyl acetate ═ 6: 1) to obtain 1.93g of yellow solid, yield: 64.8 percent and m.p.152.2-153.4 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.61(d,J=5.3Hz,1H,ArH),8.55(d,J=1.0Hz,1H,ArH),8.40(dd,J=8.0,1.6Hz,1H,ArH),7.55(d,J=8.0Hz,1H,ArH),7.47(d,J=5.3Hz,1H,ArH),7.34-7.27(m,3H,ArH),7.25-7.17(m,1H,ArH),4.83(s,2H,CH2),4.37(s,2H,CH2),2.66(s,3H,SCH3).
Synthesis of 2- (3-chlorobenzyl) -6- (2- (methylsulfonyl) pyrimidin-4-yl) isoindolin-1-one (VIII-1)
Dissolving compound XVI-1(1.0g,2.6mmol) in dichloromethane (20mL), adding m-chloroperoxybenzoic acid (1.31g,6.5mmol) under ice bath, stirring at room temperature for about 2h, detecting complete reaction of the raw material XVI-1 by TLC (dichloromethane: methanol ═ 35:1), adding saturated sodium thiosulfate solution (30mL) to the reaction solution, stirring vigorously for 10min, standing for layering, extracting the aqueous layer with dichloromethane (30mLx3), combining the organic layers, washing with saturated sodium carbonate solution (30mLx3), washing with saturated sodium chloride solution (30mL), drying with anhydrous sodium sulfate, filtering, and evaporating the solvent under reduced pressure to obtain white solid 890mg, yield: 82.7 percent and m.p.145.3-146.1 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.99(d,J=5.3Hz,1H,ArH),8.59(d,J=0.6Hz,1H,ArH),8.53(dd,J=8.0,1.5Hz,1H,ArH),8.03(d,J=5.3Hz,1H,ArH),7.62(d,J=8.0Hz,1H,ArH),7.36-7.28(m,3H,ArH),7.26-7.19(m,1H,ArH),4.83(s,2H,CH2),4.41(s,2H,CH2),3.46(s,3H,SO2CH3).
Synthesis of 2- (3-chlorobenzyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-1)
Dissolving compound VIII-1(200mg,0.48mmol) and 4-aminotetrahydropyran (IX-1) (74mg,0.73mmol) in sec-butanol (3mL), heating to 125 ℃ with a sealed tube, reacting for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that the raw materials are completely reacted, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1) to obtain 116mg of yellow solid, yield: 55.6 percent and m.p.181.9-183.0 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.49(s,1H,ArH),8.37(d,J=4.0Hz,1H,ArH),8.27(d,J=7.6Hz,1H,ArH),7.51(d,J=7.9Hz,1H,ArH),7.35-7.25(m,3H,ArH),7.21(d,J=3.4Hz,1H,ArH),7.06(d,J=4.7Hz,1H,ArH),5.37-5.22(m,1H,NH),4.81(s,2H,CH2),4.35(s,2H,CH2),4.26-4.10(m,1H,NHCH),4.04-4.00(m,2H,OC 2H),3.63-3.56(m,2H,OC 2H),2.12-2.08(m,2H,NHCHC 2H),1.66-1.56(m,2H,NHCHC 2H).
13C-NMR(75MHz,CDCl3)δ(ppm):168.10,163.96,161.79,158.75,143.23,138.92,133.06,130.89,130.32,130.18,128.18,128.03,126.27,123.25,122.58,114.84,106.71,66.80,49.51,47.22,46.02,33.29.
HRMS(ESI):m/z[M+H]+.Calcd for C24H24ClN4O2:435.1582;Found:435.1590.
IR(cm-1):3434.79,3253.87,1703.98,1600.62,1572.80,1531.13,1415.70,1261.52,1197.50,1137.32,803.41,765.66,709.03,611.12.
Example 2
Synthesis of 2- (3-fluoro-4-chlorobenzyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-2) Synthesis of 2- (4-chloro-3-fluorobenzyl) -6- (2- (methylthio) pyrimidin-4-yl) isoindolin-1-one (XVI-2)
Dissolving compound XIV (1g,3.9mmol) in 1, 4-dioxane (50mL), adding 60% sodium hydride (188mg,4.7mmol), stirring at room temperature for 1h, adding 4-chloro-3-fluorobenzyl bromide (1.3g,5.85mmol), stirring at 70 ℃ for about 12h, detecting the completion of the reaction of the raw material XIV by TLC (petroleum ether: ethyl acetate ═ 2:1), evaporating the solvent under reduced pressure, and separating the residue by column chromatography (eluent: petroleum ether: ethyl acetate ═ 6: 1) to obtain 855mg of white solid, yield: 54.8 percent and m.p.143.2-144.5 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.63(d,J=5.2Hz,1H,ArH),8.57(s,1H,ArH),8.43(dd,J=8.0,1.6Hz,1H,ArH),7.58(d,J=8.0Hz,1H,ArH),7.49(d,J=5.3Hz,1H,ArH),7.41(t,J=7.8Hz,1H,ArH),7.12(ddd,J=9.5,8.9,1.6Hz,2H,ArH),4.83(s,2H,CH2),4.40(s,2H,CH2),2.70(s,3H,SCH3).
Synthesis of 2- (4-chloro-3-fluorobenzyl) -6- (2- (methylsulfonyl) pyrimidin-4-yl) isoindolin-1-one (VIII-2)
Dissolving compound XVI-2(500mg,1.3mmol) in dichloromethane (20mL), adding m-chloroperoxybenzoic acid (657mg,3.25mmol) under ice bath, stirring at room temperature for about 2h, after TLC (dichloromethane: methanol ═ 35:1) detects that the raw material-2 is completely reacted, adding saturated sodium thiosulfate solution (20mL) into the reaction solution, stirring vigorously for 10min, standing for layering, extracting the aqueous layer with dichloromethane (20mLx3), combining the organic layers, washing with saturated sodium carbonate (20mLx3), washing with saturated sodium chloride solution (20mL), drying with anhydrous sodium sulfate, filtering, evaporating the solvent under reduced pressure to dryness to obtain white solid 447mg, yield: 79.6 percent and m.p.153.8-155.2 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.99(d,J=5.3Hz,1H,ArH),8.58(s,1H,ArH),8.53(d,J=7.9Hz,1H,ArH),8.03(d,J=5.3Hz,1H,ArH),7.62(d,J=8.0Hz,1H,ArH),7.38(t,J=7.9Hz,1H,ArH),7.13(d,J=9.1Hz,1H,ArH),7.07(d,J=8.2Hz,1H,ArH),4.81(s,2H,CH2),4.41(s,2H,CH2),3.45(s,3H,SO2CH3).
Synthesis of 2- (3-fluoro-4-chlorobenzyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-2)
Dissolving compound VIII-2(207mg,0.48mmol) and 4-aminotetrahydropyran (IX-1) (74mg,0.73mmol) in sec-butanol (3mL), heating to 125 ℃ with a sealed tube to react for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that the raw materials are completely reacted, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1) to obtain 115mg of white solid, yield: 52.9 percent and m.p.180.4-181.9 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.49(s,1H,ArH),8.37(d,J=3.8Hz,1H,ArH),8.28(dd,J=8.0,1.5Hz,1H,ArH),7.52(d,J=7.9Hz,1H,ArH),7.37(t,J=7.8Hz,1H,ArH),7.19-6.99(m,3H,ArH),5.30(brs,1H,NH),4.80(s,2H,CH2),4.36(s,2H,C 2H),4.26-4.11(m,1H,NHCH),4.04-4.00(m,2H,OC 2H),3.63-3.56(m,2H,OC 2H),2.12-2.08(m,2H,NHCHC 2H),1.68-1.55(m,2H,NHCHC 2H).
13C-NMR(75MHz,CDCl3)δ(ppm):168.11,164.03,160.77(d,J=124.1Hz),158.52,156.63,143.19,137.86(d,J=10.9Hz),133.16,132.92,131.04,130.45,124.39(d,J=3.7Hz),123.29,122.64,116.26(d,J=21.3Hz),115.00,106.65,66.78,49.50,47.25,45.66,33.26.
HRMS(ESI):m/z[M+H]+.Calcd for C24H23ClFN4O2:453.1488;Found:453.1494.
IR(cm-1):3427.73,3248.25,2922.60,2843.90,1701.76,1602.04,1570.09,1528.32,1414.52,1365.96,1162.42,1196.23,1055.28,867.41,802.31,768.02,612.67,535.91.
Example 3
Synthesis of 2- (3-chloro-4-fluorobenzyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-3)
Synthesis of 2- (3-chloro-4-fluorobenzyl) -6- (2- (methylthio) pyrimidin-4-yl) isoindolin-1-one (XVI-3)
Dissolving compound XIV (1g,3.9mmol) in 1, 4-dioxane (50mL), adding 60% sodium hydride (188mg,4.7mmol), stirring at room temperature for 1h, adding 4-fluoro-3-chlorobenzyl bromide (1.3g,5.85mmol), stirring at 70 ℃ for about 12h, detecting the completion of the reaction of the raw material XIV by TLC (petroleum ether: ethyl acetate ═ 2:1), evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: petroleum ether: ethyl acetate ═ 6: 1) to obtain 933mg of orange solid, yield: 59.8 percent and m.p.174.9-176.3 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.60(d,J=5.0Hz,1H,ArH),8.54(s,1H,ArH),8.40(d,J=8.0Hz,1H,ArH),7.55(d,J=7.9Hz,1H,ArH),7.46(d,J=5.2Hz,1H,ArH),7.38(d,J=6.8Hz,1H,ArH),7.21(brs,1H,ArH),7.12(t,J=8.5Hz,1H,ArH),4.78(s,2H,CH2),4.37(s,2H,CH2),2.67(s,3H,SCH3).
Synthesis of 2- (3-chloro-4-fluorobenzyl) -6- (2- (methylsulfonyl) pyrimidin-4-yl) isoindolin-1-one (VIII-3)
Dissolving compound XVI-3(500mg,1.25mmol) in dichloromethane (20mL), adding m-chloroperoxybenzoic acid (657mg,3.25mmol) under ice bath, stirring at room temperature for about 2h, after TLC (dichloromethane: methanol ═ 35:1) detects that the raw material has reacted completely, adding saturated sodium thiosulfate solution (20mL) to the reaction solution, stirring vigorously for 10min, standing for layering, extracting the aqueous layer with dichloromethane (20mLx3), combining the organic layers, washing with saturated sodium carbonate (20mLx3), washing with saturated sodium chloride solution (20mL), drying with anhydrous sodium sulfate, filtering, evaporating the solvent under reduced pressure to dryness to obtain white solid 446mg, yield: 82.6 percent and m.p.173.2-174.4 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.99(d,J=5.3Hz,1H,ArH),8.57(s,1H,ArH),8.53(dd,J=8.0,1.7Hz,1H,ArH),8.02(d,J=5.3Hz,1H,ArH),7.62(d,J=8.0Hz,1H,ArH),7.39(dd,J=6.8,2.1Hz,1H,ArH),7.26-7.18(m,1H,ArH),7.13(t,J=8.6Hz,1H,ArH),4.80(s,2H,CH2),4.40(s,2H,CH2),3.46(s,3H,SO2CH3).
Synthesis of 2- (3-chloro-4-fluorobenzyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-3)
Dissolving compound VIII-3(207mg,0.48mmol) and 4-aminotetrahydropyran (IX-1) (74mg,0.73mmol) in sec-butanol (3mL), heating to 125 ℃ with a sealed tube, reacting for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that the reaction of the raw material VIII-3 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1) to obtain 104mg of white solid, yield: 47.8 percent and m.p.210.5-211.7 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.49(s,1H,ArH),8.37(d,J=5.2Hz,1H,ArH),8.27(dd,J=8.0,1.4Hz,1H,ArH),7.52(d,J=7.8Hz,1H,ArH),7.38(dd,J=6.8,1.9Hz,1H,ArH),7.23-7.19(m,1H,ArH),7.16-7.01(m,2H,ArH),5.30(s,1H,NH),4.78(s,2H,CH2),4.35(s,2H,CH2),4.26-4.09(m,1H,NHCH),4.04-4.00(m,2H,OC 2H),3.63-3.56(m,2H,OC 2H),2.12-2.08(m,2H,NHCHC 2H),1.68-1.55(m,2H,NHCHC 2H).
13C-NMR(75MHz,CDCl3)δ(ppm):168.07,164.00,163.26,160.49(d,J=171.3Hz),158.58,143.19,137.92,134.01(d,J=3.6Hz),132.99,130.40,130.29,127.95,127.86,123.28,122.62,116.99(d,J=21.3Hz),106.65,66.78,49.45,47.24,45.49,33.27.
HRMS(ESI):m/z[M+H]+.Calcd for C24H23ClFN4O2:453.1488;Found:453.1492.
IR(cm-1):3469.03,3249.92,2924.39,2847.62,1701.05,1604.79,1571.85,1531.09,1493.95,1261.83,1238.03,868.32,803.01,761.81,628.16.
Example 4
Synthesis of 2- (3-chlorobenzyl) -6- (2- ((1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-4)
Compound VIII-1(200mg,0.48mmol) and 1-methyl-1H-pyrazol-5-amine (IX-2) (71mg,0.73mmol) were dissolved in tetrahydrofuran (5mL), cooled to-25 ℃, 1N LiHMDS (0.73mL,0.73mmol) was added, reacted at-25 ℃ for about 3H, after TLC (dichloromethane: methanol ═ 25:1) detection that the raw material VIII-1 reacted completely, the solvent was evaporated under reduced pressure, and the residue was separated by column chromatography (dichloromethane: methanol ═ 150: 1) to give 52mg of a white solid, yield: 25.1 percent and m.p.167.9-169.2 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.58-8.46(m,2H,ArH),8.30(d,J=7.3Hz,1H,ArH),7.61-7.46(m,2H,ArH),7.41-7.16(m,6H,ArH,NH),6.39(s,1H,ArH),4.81(s,2H,CH2),4.36(s,2H,CH2),3.84(s,3H,CH3).
13C-NMR(75MHz,CDCl3)δ(ppm):168.43,164.92,160.79,159.57,144.19,139.27,138.84,137.77,137.51,135.25,133.62,130.96,130.63-130.53(m),128.66,128.55,126.75,124.01,123.04,109.59,99.89,50.02,46.50,36.07.
HRMS(ESI):m/z[M+H]+.Calcd for C23H20ClN6O:431.1382;Found:431.1389.
IR(cm-1):3446.17,3227.14,1680.22,1581.58,1554.06,1446.36,1408.42,1263.38,1200.56,812.52,769.37,709.28,682.87,609.28.
Example 5
Synthesis of 2- (3-chlorobenzyl) -6- (2- ((1-methyl-1H-pyrazol-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-5)
Dissolving compound VIII-1(200mg,0.48mmol) and 1-methyl-1H-pyrazol-4-amine (IX-3) (71mg,0.73mmol) in sec-butanol (3mL), heating to 125 ℃ in a sealed tube to react for about 12H, detecting completion of the reaction of VIII-1 by TLC (dichloromethane: methanol ═ 25:1), cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (dichloromethane: methanol ═ 150: 1) to obtain 107mg of a light yellow solid, yield: 51.7 percent and m.p.160.1-161.3 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.56(s,1H,NH),8.5-8.47(m,2H,ArH),8.37(d,J=8.1Hz,1H,ArH),7.90(s,1H,ArH),7.72(d,J=7.6Hz,1H,ArH),7.57(s,1H,ArH),7.45-7.34(m,4H,ArH),7.27-7.25(m,1H,ArH),4.77(s,2H,CH2),4.48(s,2H,CH2),3.82(s,3H,CH3).
13C-NMR(75MHz,DMSO-d6)δ(ppm):167.58,163.42,160.26,159.87,144.74,140.50,133.78,133.19,131.12,130.50,129.40,128.04,127.88,126.88,124.67,123.68,121.69,120.95,117.71,107.29,50.00,45.44,39.26.
HRMS(ESI):m/z[M+H]+.Calcd for C23H20ClN6O:431.1382;Found:431.1386.
IR(cm-1):3446.28,1682.78,1625.78,1573.42,1433.39,1199.84,798.84,769.01,710.30,611.06.
Example 6
Synthesis of 2- (3-chloro-4-fluorobenzyl) -6- (2- ((1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-6)
Dissolving compound VIII-3(207mg,0.48mmol) and 1-methyl-1H-pyrazol-5-amine (IX-2) (71mg,0.73mmol) in tetrahydrofuran (5mL), cooling to-25 ℃, adding 1N LiHMDS (0.73mL,0.73mmol), reacting at-25 ℃ for about 3 hours, detecting by TLC (dichloromethane: methanol ═ 25:1) that the reaction of the raw material VIII-3 is complete, evaporating the solvent under reduced pressure, and performing column chromatography (dichloromethane: methanol ═ 150: 1) to obtain a light yellow solid 62mg, yield: 28.8 percent and m.p.108.1-109.2 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.55-8.45(m,2H,ArH),8.29(d,J=8.0Hz,1H,ArH),7.54-7.46(m,2H,ArH),7.38(d,J=6.2Hz,1H,ArH),7.30(d,J=4.7Hz,1H,ArH),7.20(brs,1H,NH),7.15-7.09(m,1H,ArH),7.02(s,1H,ArH),6.37(s,1H,ArH),4.78(s,2H,CH2),4.36(s,2H,CH2),3.83(s,3H,CH3).
13C-NMR(75MHz,CDCl3)δ(ppm):167.94,164.44,160.22,159.86(d,J=92.7Hz),159.03,143.64,138.32,137.27,137.06,133.87(d,J=3.8Hz),133.05,130.55,130.30,127.96,127.87,123.55,122.60,117.01(d,J=21.3Hz),109.13,99.50,49.50,45.49,35.56.
HRMS(ESI):m/z[M+H]+.Calcd for C23H19ClFN6O:449.1287;Found:449.1290.
IR(cm-1):3431.63,1685.96,1583.06,1554.62,1499.00,1445.94,1405.63,1315.27,1247.74,1200.54,1133.99,816.71,764.94,609.57,541.08.
Example 7
Synthesis of 2- (1- (3, 4-dichlorophenyl) -2-hydroxyethyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-7)
Synthesis of methyl 2-methyl-5- (2- (methylthio) pyrimidin-4-yl) benzoate (IV)
Dissolving 4-chloro-2- (methylthio) pyrimidine (XII) (19.8g,123.4mmol), methyl 2-methyl-5- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) benzoate (III) (34.1g,123.4mmol), tetratriphenylphosphine palladium (7.1g,6.17mmol) and sodium carbonate (26.2g,246.8mmol) in a mixed solvent composed of toluene (90mL), methanol (30mL) and water (30mL), reacting at 70 ℃ for about 8h under the protection of nitrogen, allowing TLC (petroleum ether: ethyl acetate: 10:1) to detect that the raw materials are completely reacted, cooling to room temperature, adding ethyl acetate (100mL), standing for layering, evaporating the solvent under reduced pressure to obtain a residue, separating by column chromatography (petroleum ether: ethyl acetate: 25:1) to obtain 29.8g of a white solid, yield: 88.0 percent and m.p.66.2-67.3 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.58(d,J=1.9Hz,1H,ArH),8.54(d,J=5.3Hz,1H,ArH),8.16(dd,J=8.0,2.0Hz,1H,ArH),7.40-7.35(m,2H,ArH),3.92(s,3H,OCH3),2.65(s,3H,SCH3),2.64(s,3H,CH3).
Synthesis of methyl 2-methyl-5- (2- (methylsulfonyl) pyrimidin-4-yl) benzoate (V)
Dissolving compound IV (24.2g,88.3mmol) in dichloromethane (150mL), adding m-chloroperoxybenzoic acid (38.1g,220.8mmol) in portions under ice bath, stirring at room temperature for about 2h, detecting that the raw material IV completely reacts by TLC (dichloromethane: methanol ═ 20:1), adding dichloromethane (100mL) for dilution, adding saturated sodium thiosulfate solution (100mL), stirring for 20min, standing for layering, extracting an aqueous layer with dichloromethane (100mL × 3), combining organic layers, washing with saturated sodium carbonate solution (100mL), washing with saturated sodium chloride solution (100mL), drying with anhydrous sodium sulfate, filtering, evaporating the solvent under reduced pressure to obtain 25.7g of light yellow solid, yield: 95.0 percent and m.p.121.8-123.3 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.91(d,J=5.3Hz,1H,ArH),8.62(d,J=1.9Hz,1H,ArH),8.23(dd,J=8.1,2.0Hz,1H,ArH),7.93(d,J=5.3Hz,1H,ArH),7.41(d,J=8.1Hz,1H,ArH),3.93(s,3H,OCH3),3.42(s,3H,SCH3),2.66(s,3H,CH3).
Synthesis of methyl 2- (bromomethyl) -5- (2- (methylsulfonyl) pyrimidin-4-yl) benzoate (VI)
The starting material V (23.6g,77.0mmol) and NBS (15.1g,84.7mmol) were dissolved in benzene (150mL), azobisisobutyronitrile (2.5g,15.4mmol) was added and reacted at 80 ℃ for about 3h, TLC (petroleum ether: ethyl acetate 1:1) checked for completion of the starting material reaction, cooled to room temperature, concentrated to a red oil and put into the next step without further treatment.
Synthesis of 2- (1- (3, 4-dichlorophenyl) -2-hydroxyethyl) -6- (2- (methylsulfonyl) pyrimidin-4-yl) isoindolin-1-one (VIII-4)
Compound VI (1g,2.6mmol), 2-amino-2- (3, 4-dichlorophenyl) ethyl-1-ol (VII-1) (533mg,2.6mmol) and triethylamine (540 μ L,3.9mmol) were dissolved in methanol (10mL), refluxed for about 6 hours, TLC (dichloromethane: methanol ═ 20:1) checked for completion of the starting material reaction, cooled to below 10 ℃ in an ice bath, stirred for 30min, solid precipitated, and suction filtered to give 694mg of white solid, yield: 55.8 percent and m.p.203.9-204.9 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.12(d,J=5.1Hz,1H,ArH),8.59(s,1H,ArH),8.52(d,J=8.4Hz,1H,ArH),8.39(d,J=4.8Hz,1H,ArH),7.87(d,J=8.5Hz,1H,ArH),7.69-7.56(m,2H,ArH),7.39(d,J=9.6Hz,1H,ArH),5.50-5.37(m,1H,HOCH2CH),4.82(d,J=19.0Hz,1H,CONC 2H),4.59(d,J=18.4Hz,1H,CONC 2H),4.24-4.02(m,2H,HOC 2H),3.55(s,3H,SO2C 3H).
Synthesis of 2- (1- (3, 4-dichlorophenyl) -2-hydroxyethyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-7)
Dissolving compound VIII-4(229mg,0.48mmol) and 4-aminotetrahydropyran (IX-1) (74mg,0.73mmol) in sec-butanol (3mL), heating to 125 ℃ in a sealed tube to react for about 12h, detecting complete reaction by TLC (dichloromethane: methanol ═ 25:1), cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (dichloromethane: methanol ═ 250: 1-50: 1) to obtain 124mg of white solid, yield: 51.7 percent and m.p.217.9-219.3 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):8.42(s,1H,ArH),8.39(d,J=5.2Hz,1H,ArH),8.34(d,J=8.3Hz,1H,ArH),7.72(d,J=8.2Hz,1H,ArH),7.64(d,J=1.4Hz,1H,ArH),7.61(d,J=8.4Hz,1H,ArH),7.35(dd,J=8.3,1.4Hz,1H,ArH),7.31(s,1H,NH),7.26(d,J=5.3Hz,1H,ArH),5.36(t,J=6.6Hz,1H,HOCH2CH),4.72(d,J=17.9Hz,1H,CONC 2H),4.48(d,J=18.2Hz,1H,CONC 2H),4.12-3.96(m,3H,HOC 2H,NHCH),3.91-3.88(m,2H,O(C 2 2H)),3.59-3.40(m,2H,O(C 2 2H)),1.61-1.57(m,2H,NHC 2H),1.51-1.51(m,2H,NHC 2H).
13C-NMR(75MHz,DMSO-d6)δ(ppm):167.41,163.63,161.62,158.95,144.38,139.78,137.03,132.67,131.16,130.66,130.07,129.86,129.50,128.02,123.92,120.96,105.66,66.09,60.84,55.84,47.28,46.77,32.46.
HRMS(ESI):m/z[M+H]+.Calcd for C25H25Cl2N4O3:499.1298;Found:499.1295.
IR(cm-1):3427.73,3298.57,2939.08,2839.35,1567.99,1524.11,1455.27,1411.81,1161.99,1137.06,1084.73,871.24,802.94,772.14,606.59,524.48.
Example 8
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-8)
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- (methylsulfonyl) pyrimidin-4-yl) isoindolin-1-one (VIII-5)
Compound VI (9.63g,25.0mmol), 2-amino-2- (3-chlorophenyl) ethyl-1-ol (VII-2) (4.29g,25.0mmol) and triethylamine (10.4mL,74.9mmol) were dissolved in methanol (100mL), refluxed for about 6 hours, and after completion of the reaction of the starting materials by TLC (dichloromethane: methanol ═ 20:1), cooled in ice bath to 10 ℃ or less, stirred for 30min, solid precipitated, and suction filtered to obtain 7.2g of a white solid, yield: 64.9 percent and m.p.166.2-167.4 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):9.01(d,J=5.3Hz,1H,ArH),8.49(s,1H,ArH),8.44(dd,J=8.0,1.5Hz,1H,ArH),8.01(d,J=5.3Hz,1H,ArH),7.58(d,J=8.0Hz,1H,ArH),7.50-7.31(m,3H,ArH),7.28-7.26(m,1H,ArH),5.40(dd,J=7.9,4.4Hz,1H,CHCH2OH),4.63(d,J=17.8Hz,1H,C 2H),4.43-4.32(m,2H,HOC 2HCH),4.28(dd,J=11.9,4.4Hz,1H,C 2H),3.49(s,3H,SO2C 3H).
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-8)
Dissolving compound VIII-5(213mg,0.48mmol) and 4-aminotetrahydropyran (IX-1) (74mg,0.73mmol) in sec-butanol (3mL), heating to 125 ℃ with a sealed tube, reacting for about 12h, detecting by TLC (dichloromethane: methanol: 25:1) that the reaction of the raw material VIII-5 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and performing column chromatography (dichloromethane: methanol: 250: 1-50: 1) to obtain 117mg of a light yellow solid, yield: 52.4 percent and m.p.141.0-142.3 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.47(s,1H,ArH),8.34-8.26(m,2H,ArH),7.54-7.51(m,1H,ArH),7.34-7.30(m.3H,ArH),7.24-7.21(m,1H,ArH),7.11-7.05(m,1H,ArH),5.30-5.24(m,1H,HOCH2CH),4.51(d,J=17.0Hz,1H,CONC 2H),4.37-4.19(m,4H,CONC 2H,HOC 2H,NHCH),4.04-4.01(m,2H,O(C 2 2H)),3.63-3.57(m,2H,O(C 2 2H)),2.11-2.08(m,2H,NHC 2H),1.68-1.64(m,2H,NHC 2H).
13C-NMR(75MHz,CDCl3)δ(ppm):169.71,165.54,162.32,159.43,143.86,139.89,138.43,135.35,133.52,130.93,130.75,128.89,128.09,126.24,123.60,122.91,107.12,67.31,63.65,60.08,49.66,47.68,33.76.
HRMS(ESI):m/z[M+H]+.Calcd for C25H26ClN4O3:465.1688;Found:465.1688.
IR(cm-1):3421.83,3266.10,2946.95,2851.70,1677.23,1571.70,1454.84,1454.84,1203.44,1084.97,802.24,771.67,521.54.
Example 9
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-9)
Synthesis of 2- (2- ((tert-butyldimethylsilyl) oxy) -1- (3-chlorophenyl) ethyl) -6- (2- (methylsulfonyl) pyrimidin-4-yl) isoindolin-1-one (X-1)
Dissolving compound VIII-5(1.0g,2.26mmol) and imidazole (0.31g,4.52mmol) in dichloromethane (15mL), adding TBSCl (0.68g,4.52mmol), stirring at 35 ℃ for about 8h, detecting the complete reaction of the raw material VIII-5, washing with saturated sodium bicarbonate (6mL), washing with saturated sodium chloride solution (6mL), drying with anhydrous sodium sulfate, filtering, evaporating the solvent under reduced pressure, adding petroleum ether (6mL), pulping, and filtering to obtain a white solid 1.20g, yield 95.1%, m.p.131.7-133.2 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.99(d,J=5.3Hz,1H,ArH),8.58-8.49(m,2H,ArH),8.02(d,J=5.4Hz,1H,ArH),7.63(d,J=7.9Hz,1H,ArH),7.44(s,1H,ArH),7.29(d,J=6.5Hz,3H,ArH),5.58(t,J=5.4Hz,1H,CHCH2OSi),4.70(d,J=17.9Hz,1H,C 2H),4.36(d,J=18.0Hz,1H,C 2H),4.28-4.21(m,2H,SiOC 2HCH),3.46(s,3H,SO2C 3H),0.84(s,9H,SiC(C 3 3H)),0.07(s,3H,SiC 3H),0.05(s,3H,SiC 3H).
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-9)
Dissolving a compound X-1(200mg,0.36mmol) and 1-methyl-1H-pyrazol-5-amine (IX-2) (52mg,0.54mmol) in anhydrous tetrahydrofuran (5mL), cooling to-25 ℃, adding 1N LiHMDS (0.54mL,0.54mmol), reacting at 25 ℃ for about 3H, detecting that the raw material X-1 is completely reacted by TLC (dichloromethane: methanol ═ 25:1), evaporating the solvent under reduced pressure, adding ethyl acetate (5mL), dissolving, adjusting the pH to 2-3 by using an ethyl acetate solution of HCl, continuously stirring for about 3H, detecting that the intermediate is completely reacted by TLC (dichloromethane: methanol ═ 25:1), evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1), so as to obtain 35mg of a white solid, wherein the yield: 21.1 percent and m.p.118.2-119.4 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.45(d,J=5.0Hz,1H,ArH),8.32(s,1H,ArH),8.07(d,J=7.5Hz,1H,ArH),7.84(s,1H,ArH),7.41-7.35(m,3H,ArH),7.28(m,2H,ArH),7.17(d,J=5.0Hz,1H,ArH),6.15(s,1H,ArH),5.41(s,1H,HOCH2CH),4.52(d,J=17.4Hz,1H,CONC 2H),4.47-4.12(m,3H,CONC 2H,HOC 2H),3.81(s,3H,CH3).
13C-NMR(75MHz,CDCl3)δ(ppm):169.37,165.04,161.12,159.75,144.28,139.92,138.64,137.93,137.60,135.33,133.56,130.91,130.71,128.84,128.18,126.33,123.75,122.87,109.67,100.15,63.15,59.25,49.13,36.09.
HRMS(ESI):m/z[M+H]+.Calcd for C24H22ClN6O2:461.1487;Found:461.1489.
IR(cm-1):3411.10,3233.04,2938.05,1673.72,1588.57,1554.41,1446.65,1406.90,1267.97,1202.18,815.17,770.16,610.50.
Example 10
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((1-methyl-1H-pyrazol-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-10)
Dissolving compound VIII-5(160mg,0.36mmol) and 1-methyl-1H-pyrazol-4-amine (IX-3) (52mg,0.54mmol) in sec-butyl alcohol (3mL), heating to 125 ℃ in a sealed tube, reacting for about 12H, detecting complete reaction by TLC (dichloromethane: methanol 25:1), cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol 200: 1-50: 1) to obtain an earthy yellow solid 93mg, yield: 56.0 percent and m.p.218.2-219.4 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.59(s,1H,NH),8.82-8.20(m,3H,ArH),7.91(s,1H,ArH),7.75(s,1H,ArH),7.56(s,1H,ArH),7.50-6.91(m,5H,ArH),5.38(s,1H,OH),5.21(s,1H,HOCH2CH),4.73(d,J=16.7Hz,1H,CONC 2H),4.47(d,J=17.8Hz,1H,CONC 2H),4.01(s,2H,HOC 2H),3.82(s,3H,CH3).
13C-NMR(75MHz,DMSO-d6)δ(ppm):167.88,163.44,160.25,159.86,144.96,141.58,133.72,133.36,130.96,130.52,130.45,127.96,127.73,126.74,124.61,123.68,121.57,120.93,111.49,107.28,61.41,56.74,47.67,39.27.
HRMS(ESI):m/z[M+H]+.Calcd for C24H22ClN6O2:461.1487;Found:461.1494.
IR(cm-1):3427.46,3261.74,3062.46,2920.09,1680.07,1626.00,1573.30,1451.93,1433.52,1333.65,1200.93,991.89,802.54,769.16,617.20.
Example 11
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- (isopropylamino) pyrimidin-4-yl) isoindolin-1-one (I-11)
Dissolving compound VIII-5(200mg,0.45mmol) and isopropylamine (IX-3) (40mg,0.68mmol) in sec-butyl alcohol (3mL), sealing the tube, heating to 125 ℃ for reaction for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that the reaction of the raw material VIII-5 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain 96mg of white solid, yield: 50.4 percent and m.p.140.1-142.4 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.42(s,1H,ArH),8.36-8.32(m,1H,ArH),8.24(d,J=7.6Hz,1H,ArH),7.46-7.44(m,1H,ArH),7.33-7.29(m.3H,ArH),7.26-7.23(m,1H,ArH),6.98(d,J=5.2Hz,1H,ArH),5.33-5.26(m,2H,HOCH2CH,NHCH),4.52(d,J=17.5Hz,1H,CONC 2H),4.31-4.25(m,4H,CONC 2H,HOC 2H,NHCH),1.30(d,J=6.5Hz,6H,NHCH 3 2(CH)).
13C-NMR(75MHz,CDCl3)δ(ppm):158.27,142.81,138.92,137.47,134.37,132.40,130.01,129.79,127.91,127.10,125.28,122.86,122.56,122.05,121.86,110.05,105.54,62.46,58.94,48.58,42.33,22.40.
HRMS(ESI):m/z[M+H]+.Calcd for C23H24ClN4O2:423.1588;Found:423.1588.
IR(cm-1):3478.65,3245.37,1660.99,1597.27,1573.23,1537.51,1459.55,1408.28,1203.39,1178.35,1177.90,773.23.
Example 12
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- (2-hydroxyethylamino) pyrimidin-4-yl) isoindolin-1-one (I-12)
Dissolving compound VIII-5(300mg,0.68mmol) and ethanolamine (IX-5) (124mg,2.03mmol) in sec-butyl alcohol (3mL), heating to 125 ℃ in a sealed tube, reacting for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that raw material VIII-5 completely reacts, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain white solid 160mg, yield: 55.7 percent and m.p.157.5-160.1 ℃.
1H-NMR(300MHz,CD3OD)δ(ppm):8.51(s,1H,ArH),8.35-8.32(m,2H,ArH),7.66-7.64(m,1H,ArH),7.44-7.34(m.4H,ArH),7.17-7.16(m,1H,ArH),5.53-5.49(m,1H,HOCH2CH),4.80-4.72(m,1H,CONC 2H),4.44(d,J=18.1Hz,1H,CONC 2H),4.27-4.17(m,2H,HO 2CHCH),3.77-3.76(m,2H,HO 2CHCH2NH),3.67-3.62(m,2H,HOCH2 2CHNH).
13C-NMR(75MHz,CD3OD)δ(ppm):169.02,163.52,161.88,157.75,143.99,139.34,136.96,133.85,131.92,129.97,129.67,127.34,126.86,125.21,122.97,121.03,105.35,60.49,59.98,56.40,42.78,25.31.
HRMS(ESI):m/z[M+H]+.Calcd for C22H22ClN4O3:425.1380;Found:425.1376.
IR(cm-1):3253.11,1655.42,1624.36,1575.99,1545.27,1458.44,1403.24,1360.94,1272.46,1203.64,1201.80,1065.99.
Example 13
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- (3-hydroxypropylamino) pyrimidin-4-yl) isoindolin-1-one (I-13)
Dissolving compound VIII-5(300mg,0.68mmol) and propanolamine (IX-6) (152mg,2.03mmol) in sec-butyl alcohol (3mL), sealing the tube, heating to 125 ℃ for reaction for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that raw material VIII-5 completely reacts, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain 130mg of white solid, yield: 43.6 percent and m.p.64.2-66.3 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.37(s,1H,ArH),8.28(s,1H,ArH),8.17(d,J=7.7Hz,1H,ArH),7.46(d,J=7.7Hz,1H,ArH),7.35-7.26(m,4H,ArH),6.98-6.96(m,1H,ArH),5.99(s,1H,HNCH2),5.44-5.40(m,1H,HOCH2 CH),4.59(d,J=17.5Hz,1H,CONC 2H),4.38-4.21(m,3H,CONC 2H,HO 2CHCH),4.02(brs,2H,HOCH2CH,HOCH2CH2),3.75-3.68(m,4H,HO 2CHCH2 2CHNH),1.88-1.85(m,2H,HOCH2 2CHCH2NH).
13C-NMR(75MHz,CDCl3)δ(ppm):168.75,163.48,162.27,158.06,143.11,138.85,136.96,134.34,132.47,129.88,129.78,127.89,127.11,125.31,122.72,121.96,105.98,62.08,58.26,48.17,37.08,32.37,26.46.
HRMS(ESI):m/z[M+H]+.Calcd for C23H24ClN4O3:439.1537;Found:439.1532.
IR(cm-1):3439.85,1668.07,1572.74,1493.88,1454.98,1407.24,1342.27,1266.70,1201.84,1067.90,806.12,770.92,695.77,613.93.
Example 14
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- (cyclohexylamino) pyrimidin-4-yl) isoindolin-1-one (I-14)
Dissolving compound VIII-5(400mg,0.90mmol) and cyclohexylamine (IX-7) (268mg,2.70mmol) in sec-butyl alcohol (3mL), heating to 125 ℃ in a sealed tube, reacting for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that raw material VIII-5 completely reacts, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain 180mg of white solid, yield: 43.2 percent and m.p.150.4-151.8 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.37(s,1H,ArH),8.25(d,J=7.8Hz,1H,ArH),8.16(s,1H,ArH),7.56(d,J=7.9Hz,1H,ArH),7.40(s,1H,ArH),7.32-7.29(m,3H,ArH),7.06(d,J=5.7Hz,1H,ArH),5.50-5.46(m,1H,HOCH2 CH),4.73(d,J=17.7Hz,1H,CONC 2H),4.45-4.23(m,4H,CONC 2H,HO 2CHCH,CH2 CHNH),4.02(s,1H,HOCH2CH),2.11-2.04(m,2H,cyclohexyl-H),1.91-1.83(m,2H,cyclohexyl-H),1.68-1.36(m,6H,cyclohexyl-H).
13C-NMR(75MHz,CDCl3)δ(ppm):168.08,164.51,157.71,143.69,138.87,135.44,134.33,132.83,130.25,129.78,128.51,127.89,127.16,125.29,123.00,122.35,104.63,61.86,57.74,49.82,47.92,32,02,25.02,24.07
HRMS(ESI):m/z[M+H]+.Calcd for C26H28ClN4O2:463.1901;Found:463.1896.
IR(cm-1):3297.74,2928.85,1673.28,1570.45,1524.32,1455.75,1419.22,1283.98,1205.37,798.16,770.61,709.39,682.87.
Example 15
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((4-hydroxycyclohexyl) amino) pyrimidin-4-yl) isoindolin-1-one (I-15)
Dissolving compound VIII-5(400mg,0.90mmol) and 4-aminocyclohexanol (IX-8) (311mg,2.70mmol) in sec-butanol (3mL), sealing the tube and heating to 125 ℃ for reaction for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that the reaction of the raw material VIII-5 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain 210mg of white solid, yield: 48.7 percent and m.p.170.3-171.6 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.42(s,1H,ArH),8.32-8.31(m,1H,ArH),8.24(d,J=8.0Hz,1H,ArH),7.49(d,J=8.0Hz,1H,ArH),7.35(s,1H,ArH),7.32-7.26(m,3H,ArH),7.01(d,J=5.4Hz,1H,ArH),5.73(brs,1H,HNCH),5.40-5.36(m,1H,HOCH2 CH),4.57(d,J=17.5Hz,1H,CONCH 2),4.38-4.22(m,3H,CONCH 2,HO 2CHCH),3.99-3.96(m,1H,(CH2)2 CHOH),3.78-3.68(m,1H,(CH2)2 CHNH),2.23-2.05(m,4H,hydroxycyclohexyl-H),1.59-1.33(m,4H,hydroxycyclohexyl-H).
13C-NMR(75MHz,CDCl3)δ(ppm):168.57,153.71,143.49,138.88,136.74,134.34,132.54,130.03,129.78,128.51,127.89,127.13,127.03,125.29,122.75,122.01,105.55,69.32,62.17,58.21,48.94,48.13,33.39,30.13.
HRMS(ESI):m/z[M+H]+.Calcd for C26H28ClN4O3:479.1850;Found:479.1846.
IR(cm-1):3429.06,2929.86,2848.98,1654.59,1594.29,1575.12,1524.49,1495.10,1456.75,1414.30,1347.78,1203.31,1068.26,808.64,771.78,689.80,612.24.
Example 16
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((tetrahydrofuran-3-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-16)
Dissolving compound VIII-5(400mg,0.90mmol) and 3-aminotetrahydrofuran (IX-8) (48mg,1.35mmol) in sec-butanol (3mL), heating to 125 ℃ with a sealed tube, reacting for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that the reaction of the raw material VIII-5 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain 220mg of white solid, yield: 54.2 percent and m.p.192.3-193.9 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.38-8.34(m,2H,ArH),8.20(d,J=7.9Hz,1H,ArH),7.43(d,J=7.7Hz,1H,ArH),7.32-7.28(m,3H,ArH),7.24-7.23(m,1H,ArH),7.03-7.02(m,1H,ArH),5.72(s,1H,NHCH),5.35-5.30(m,1H,HOCH2CH),4.70-4.68(m,1H,NHCH),4.54(d,J=17.6Hz,1H,CONCH 2),4.34-4.20(m,3H,CONCH 2,HO 2CH),4.05-4.01(m,2H,O 2CH),3.91-3.89(m,1H,OCH 2),3.80-3.78(m,1H,OCH 2),2.40-2.34(m,1H,CHCH 2CH2O),1.98-1.94(m,1H,CHCH 2CH2O).
13C-NMR(75MHz,CDCl3)δ(ppm):169.04,163.03,161.58,158.33,142.98,138.89,137.27,134.35,132.41,129.94,129.78,127.89,127.10,125.29,122.59,121.83,106.20,73.27,66.61,62.23,58.43,51.51,48.27,32.81.
HRMS(ESI):m/z[M+H]+.Calcd for C24H24ClN4O3:451.1537;Found:451.1533.
IR(cm-1):3239.95,1669.85,1566.82,1532.89,1458.47,1426.89,1407.90,1334.69,1200.00,800.56,771.43,685.71,612.24.
Example 17
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- (2-aminothiazole) pyrimidin-4-yl) isoindolin-1-one (I-17)
Dissolving 2-aminothiazole (IX-10) (36mg,0.36mmol) in anhydrous tetrahydrofuran (5mL), cooling to-78 ℃, adding 1N LiHMDS (0.36mL,0.36mmol), stirring for 30min, dissolving compound X-1(100mg,0.18mmol) in anhydrous tetrahydrofuran (3mL), slowly dropwise adding the reaction solution at-78 ℃ to react at-78 ℃ for about 1h, detecting the completion of the reaction of raw material X-1 by TLC (dichloromethane: methanol: 20:1), evaporating the solvent under reduced pressure, adding ethyl acetate (5mL) to dissolve, adjusting the pH to 2-3 with an ethyl acetate solution of HCl, continuing stirring for about 3h, detecting the intermediate reaction by TLC (dichloromethane: methanol: 15: 1), evaporating the solvent under reduced pressure, separating by column chromatography (eluent: dichloromethane: methanol: 1-30: 1), yield 45mg of the target compound hydrochloride as a golden yellow solid: 50.0%, m.p. >220.1 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):8.73-8.72(m,1H,ArH),8.61(s,1H,ArH),8.55-8.52(m,1H,ArH),7.82-7.76(m,2H,ArH),7.55-7.50(m,1H,ArH),7.43-7.36(m,4H,ArH),7.23(s,1H,ArH),5.39-5.32(m,1H,HOCH2 CH),4.76(d,J=17.9Hz,1H,CONCH 2),4.49(d,J=18.2Hz,1H,CONCH 2),4.07-4.01(m,2H,HO 2CHCH).
13C-NMR(75MHz,DMSO-d6)δ(ppm):158.69,155.65,152.61,148.10,136.88,132.54,127.24,126.17,124.71,124.55,121.96,120.09,118.97,118.74,117.74,115.83,113.39,106.05,103.98,101.06,52.36,47.83,38.78.
HRMS(ESI):m/z[M+H]+.Calcd for C23H19ClN5O2S:464.0948;Found:464.0947.
IR(cm-1):3404.23,1666.47,1549.64,1453.87,1395.88,1203.23,1166.75,1072.97,771.35,691.31,614.43.
Example 18
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((1-methyl-1H-pyrazol-3-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-18)
Dissolving N-methyl-3-aminopyrazole (IX-11) (35mg,0.36mmol) in anhydrous tetrahydrofuran (5mL), cooling to-78 ℃, adding 1N LiHMDS (0.36mL,0.36mmol), stirring for 30min, dissolving compound X-1(100mg,0.18mmol) in anhydrous tetrahydrofuran (3mL), slowly dropwise adding the reaction solution at-78 ℃ to react at-78 ℃ for about 1h, detecting by TLC (dichloromethane: methanol ═ 20:1) that raw material X-1 has completely reacted, evaporating the solvent under reduced pressure, adding ethyl acetate (5mL) to dissolve, adjusting pH to 2-3 with HCl ethyl acetate solution, stirring for about 3h, detecting by TLC (dichloromethane: methanol ═ 15: 1) that the intermediate has completely reacted, evaporating the solvent under reduced pressure, separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-30: 1), yield 50mg of the hydrochloride of the target compound as a white solid: 55.8 percent and m.p.218.2-219.8 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):8.50-8.49(m,1H,ArH),8.44(s,1H,ArH),8.38-8.36(m,1H,ArH),7.75-7.72(m,1H,ArH),7.58(s,1H,ArH),7.46-7.35(m,5H,ArH),6.64(s,1H,ArH),5.38-5.33(m,1H,HOCH2 CH),5.24(s,1H,HNC),4.72(d,J=18.7Hz,1H,CONCH 2),4.44(d,J=18.6Hz,1H,CONCH 2),4.04-3.97(m,2H,HO 2CHCH),3.74(s,3H, 3CHN).
13C-NMR(75MHz,DMSO-d6)δ(ppm):159.21,154.35,151.09,150.89,139.21,135.97,132.21,128.26,124.74,124.10,122.47,122.05,121.59,119.06,118.61,117.53,115.67,112.53,99.15,88.07,83.25,52.13,47.79,38.66
HRMS(ESI):m/z[M+H]+.Calcd for C24H22ClN6O2:461.1493;Found:461.1483.
IR(cm-1):3416.33,1660.65,1569.89,1404.85,1204.08,1073.47,795.92,771.43,608.16.
Example 19
Synthesis of 2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- (1-methyl-1H-tetrazol-5-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-19)
Dissolving a compound X-1(500mg,0.90mmol) and 1-methyl-5-aminotetrazole (IX-12) (228mg,2.30mmol) in anhydrous N, N-dimethylformamide (4mL), cooling to-70 ℃, adding 1N LiHMDS (1.35mL,1.35mmol), reacting at 70 ℃ for about 2 hours, detecting the reaction by TLC (dichloromethane: methanol ═ 20:1), evaporating the solvent under reduced pressure, adding ethyl acetate (5mL), dissolving, adjusting the pH to 2-3 by using an ethyl acetate solution of HCl, continuously stirring for about 3 hours, detecting the reaction completion of an intermediate by TLC (dichloromethane: methanol ═ 15: 1), evaporating the solvent under reduced pressure, and carrying out column chromatography (eluent: dichloromethane: methanol ═ 200: 1-30: 1) to obtain a light yellow solid, wherein the target product hydrochloride is 180mg, and the yield: 40.1 percent and m.p.86.3-88.0 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):8.62-8.60(m,1H,ArH),8.43(s,1H,ArH),8.35-8.32(m,1H,ArH),7.75-7.73(m,2H,ArH),7.42-7.32(m,4H,ArH),5.39-5.35(m,1H,HOCH2 CH),5.19(s,1H,HNCH),4.73(d,J=18.2Hz,1H,CONCH 2),4.45(d,J=18.2Hz,1H,CONCH 2),4.08-4.02(m,2H,HO 2CHCH),3.91(s,3H,N 3CH)
13C-NMR(75MHz,DMSO-d6)δ(ppm):159.11,154.89,151.34,150.70,142.71,136.40,132.10,127.27,124.75,124.16,122.04,121.62,119.08,118.58,117.51,115.78,112.65,101.76,52.09,47.78,38.66,25.12
HRMS(ESI):m/z[M+H]+.Calcd for C22H20ClN8O2:463.1398;Found:463.1397.
IR(cm-1):3417.98,1664.86,1626.84,1594.43,1556.37,1495.65,1439.49,1410.86,1372.67,1227.31,1204.91,1061.42,822.97,770.34,686.55.
Example 20
Synthesis of (R) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-20)
Synthesis of (R) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- (methylsulfonyl) pyrimidin-4-yl) isoindolin-1-one (VIII-6)
Dissolving compound VI (1.57g,4.08mmol), (R) -2-amino-2- (3-chlorophenyl) ethyl-1-ol (VII-3) (700mg,4.08mmol) and triethylamine (1.70mL,12.24mmol) in acetonitrile (30mL), heating under reflux for about 6h, evaporating the solvent under reduced pressure after the reaction is detected by TLC (dichloromethane: methanol ═ 20:1), adding methanol (10mL), cooling in ice bath to below 10 ℃, stirring for 30min, precipitating a solid, and performing suction filtration to obtain 1.03g of a white solid, yield: 56.9 percent and m.p.78.2-79.8 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.15(d,J=5.3Hz,1H,ArH),8.60-8.55(m,3H,ArH),7.84(d,J=8.0,1H,ArH),7.44(s,1H,ArH),7.38-7.32(m,3H,ArH),5.41-5.37(m,1H,CHCH2OH),5.22-5.19(m,1H,CHCH2 OH),4.78(d,J=18.6Hz,1H,CH 2N),4.51(d,J=18.6Hz,1H,CH 2N)4.08 4.01(m,2H,HO 2CHCH),3.51(s,3H, 3CH).
Synthesis of (R) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-20)
Dissolving compound VIII-6(400mg,0.90mmol) and 4-aminotetrahydropyran (IX-1) (182mg,1.80mmol) in sec-butanol (5mL), heating to 125 ℃ with a sealed tube, reacting for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that the reaction of the raw material VIII-6 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain 208mg of white solid, yield: 49.7 percent and m.p.83.8-85.7 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):8.47-8.37(m,3H,ArH),7.77(d,J=7.8Hz,1H,ArH),7.48(s,1H,ArH),7.42-7.37(m,3H,ArH),7.30-7.29(m,1H,ArH),5.45-5.41(m,1H,HOCH2 CH),5.27-5.24(m,1H,HOCH2CH),4.78(d,J=18.3Hz,1H,CONC 2H),4.50(d,J=18.3Hz,1H,CONC 2H),4.09-4.07(m,3H,NHCH,HO 2CH),3.96-3.93(m,2H,O(C 2 2H)),3.56-3.47(m,2H,O(C 2 2H)),1.96-1.92(m,2H,NHCH(C 2 2H)),1.66-1.59(m,2H,NHCH(C 2 2H)).
13C-NMR(75MHz,DMSO-d6)δ(ppm):167.91,162.73,161.52,159.11,144.21,140.61,136.99,133.28,132.43,130.54,130.03,127.58,127.10,125.98,124.01,120.93,105.74,66.07,60.63,56.34,47.19,46.61,32.29.
HRMS(ESI):m/z[M+H]+.Calcd for C25H26ClN4O3:465.1693;Found:465.1687.
IR(cm-1):3415.44,2949.76,2359.85,1673.13,1573.18,1524.05,1454.41,1408.20,1201.03,1136.01,1077.26,806.04,767.72,605.57.
Example 21
Synthesis of (R) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((1-methyl-1H-pyrazol-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-21)
Dissolving compound VIII-6(400mg,0.90mmol) and 1-methyl-1H-pyrazol-4-amine (IX-3) (175mg,1.80mmol) in sec-butyl alcohol (5mL), heating to 125 ℃ in a sealed tube to react for about 12H, detecting by TLC (dichloromethane: methanol: 20:1) that the reaction of the raw material VIII-6 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol: 200: 1-50: 1) to obtain a golden yellow solid 100mg, wherein the yield is: 24.1 percent and m.p.229.7-231.6 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.61(s,1H,NH),8.58-8.56(m,1H,ArH),8.52(s,1H,ArH),8.45-8.43(m,1H,ArH),7.97(s,1H,ArH),7.83-7.80(m,1H,ArH),7.64(s,1H,ArH),7.49-7.39(m,5H,ArH),5.47-5.42(m,1H,HOCH2 CH),5.27-5.24(m,1H,HOCH2CH),4.79(d,J=18.0Hz,1H,CONCH 2),4.52(d,J=18.2Hz,1H,CONCH 2),4.14-4.07(m,2H,HO 2CH),3.88(s,3H,N 3CH).
13C-NMR(75MHz,DMSO-d6)δ(ppm):167.80,163.00,159.62,159.37,144.39,140.69,136.89,133.27,132.59,130.54,130.12,129.99,127.57,127.12,126.04,124.18,122.93,121.03,120.75,106.93,60.67,56.33,47.20,38.58.
HRMS(ESI):m/z[M+H]+.Calcd for C24H22ClN6O2:461.1493;Found:461.1492.
IR(cm-1):3421.26,3262.27,1674.63,1626.32,1572.86,1454.67,1433.10,1405.87,1333.36,1299.81,1201.52,1039.41,992.27,803.19,768.62,615.10.
Example 22
Synthesis of (S) -2- (2-hydroxy-1-phenylethyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-22)
Synthesis of (S) -2- (2-hydroxy-1-phenylethyl) -6- (2- (methylsulfonyl) pyrimidin-4-yl) isoindolin-1-one (VIII-7)
Dissolving compound VI (2.81g,7.29mmol), L-phenylglycinol (VII-4) (1g,7.29mmol) and triethylamine (2.03mL,14.58mmol) in acetonitrile (30mL), heating and refluxing for about 6h, detecting by TLC (dichloromethane: methanol 20:1) that the raw materials have reacted, evaporating the solvent under reduced pressure, adding methanol (10mL), cooling in ice bath to below 10 ℃, stirring for 30min, precipitating solids, and performing suction filtration to obtain 1.65g of white solid, wherein the yield is: 55.3 percent and m.p.154.2-155.8 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.13-9.12(m,1H,ArH),8.56-8.53(m,3H,ArH),7.81(d,J=7.8Hz,1H,ArH),7.34-7.26(m,5H,ArH),5.41-5.37(m,1H,CHCH2OH),5.16-5.14(m,1H,CHCH2 OH),4.75(d,J=18.7Hz,1H,CH 2N),4.43(d,J=18.5Hz,1H,CH 2N),4.06-3.95(m,2H,HO 2CHCH),3.48(s,3H,SO2 3CH).
Synthesis of (S) -2- (2-hydroxy-1-phenylethyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-22)
Dissolving compound VIII-7(400mg,0.98mmol) and 4-aminotetrahydropyran (IX-1) (198mg,1.96mmol) in sec-butanol (5mL), sealing the tube and heating to 125 ℃ for reaction for about 12h, detecting by TLC (dichloromethane: methanol ═ 20:1) that the reaction of the raw material VIII-7 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain 260mg of white solid, yield: 61.6 percent and m.p.143.9-145.6 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):8.42-8.31(m,3H,ArH),7.71(d,J=8.0Hz,1H,ArH),7.35-7.24(m,6H,ArH),5.42-5.34(m,1H,HOCH2 CH),5.17-5.13(m,1H,HOCH2CH),4.71(d,J=18.2Hz,1H,CONC 2H),4.40(d,J=18.1Hz,1H,CONC 2H),4.09-3.98(m,3H,HO 2CHCH,HNCH),3.91-3.88(m,2H,CH2 CH 2OCH 2CH2),3.44-3.42(m,2H,CH2 CH 2OCH 2CH2),1.91-1.85(m,2H,CH2 CH 2CHCH 2CH2),1.57-1.53(m,2H,CH2 CH 2CHCH 2CH2).
13C-NMR(75MHz,DMSO-d6)δ(ppm):168.23,162.07,159.67,144.66,138.63,137.45,133.24,130.36,129.12,128.08,127.74,126.10,124.46,121.37,106.18,66.58,61.30,57.14,47.38,47.11,32.84.
HRMS(ESI):m/z[M+H]+.Calcd for C25H27N4O3:431.2083;Found:431.2073.
IR(cm-1):3376.06,3252.41,2932.72,2845.54,1676.71,1567.26,1526.08,1453.81,1410.60,1364.66,1305.55,1274.66,1201.98,1136.46,1088.16,769.49,703.07.
Example 23
Synthesis of (S) -2- (2-hydroxy-1-phenylethyl) -6- (2- ((1-methyl-1H-pyrazol-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-23)
Dissolving compound VIII-7(400mg,0.98mmol) and 1-methyl-1H-pyrazol-4-amine (IX-3) (190mg,1.96mmol) in sec-butyl alcohol (5mL), heating to 125 ℃ in a sealed tube to react for about 12H, detecting by TLC (dichloromethane: methanol ═ 20:1) that the reaction of the raw material VIII-7 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain 206mg of white solid, yield: 49.3 percent and m.p.227.9-229.7 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.58(s,1H,HN),8.52-8.51(m,1H,ArH),8.46(s,1H,ArH),8.38(d,J=8.4Hz,1H,ArH),7.91(s,1H,ArH),7.76(d,J=8.1Hz,1H,ArH),7.57(s,1H,ArH),7.41(d,J=5.1Hz,1H,ArH),7.36-7.31(m,5H,ArH),5.43-5.38(m,1H,HOCH2 CH),5.18-5.14(m,1H,HOCH2),4.73(d,J=18.2Hz,1H,CONCH 2),4.42(d,J=18.4Hz,1H,CONCH 2),4.06-3.97(m,2H,HO 2CHCH),3.83(s,3H,N 3CH).
13C-NMR(75MHz,DMSO-d6)δ(ppm):168.28,163.53,160.10,159.90,144.85,138.55,135.38,133.30,130.54,130.45,129.18,128.13,127.73,124.67,123.43,121.48,121.25,107.44,61.30,57.17,47.42,39.17.
HRMS(ESI):m/z[M+H]+.Calcd for C24H23N6O2:427.1882;Found:427.1871.
IR(cm-1):3409.61,3267.17,2920.35,1677.93,1627.82,1574.07 1493.34,1450.57,1435.03,1365.18,1335.15,1299.97,1201.55,805.63,763.49,707.73,605.10.
Example 24
Synthesis of (S) -2- (2-hydroxy-1-phenylethyl) -6- (2- ((1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-24)
Synthesis of (S) -2- (2- ((tert-butyldimethylsilyl) oxy) -1-phenylethyl) -6- (2- (methylsulfonyl) pyrimidin-4-yl) isoindolin-1-one (X-2)
Dissolving a compound VIII-7(400mg,0.98mmol) and imidazole (133mg,1.96mmol) in dichloromethane (4mL), adding TBSCl (295mg,1.96mmol), stirring at 35 ℃ for reaction for about 8h, after TLC (dichloromethane: methanol ═ 30: 1) detection of the reaction of the raw material VIII-7, washing with saturated sodium bicarbonate (4mL), washing with saturated sodium chloride solution (4mL), drying with anhydrous sodium sulfate, filtering, evaporating the solvent under reduced pressure, adding petroleum ether (4mL), pulping, filtering to obtain a white solid 480mg, the yield is 93.6%, and m.p.70.2-72.1 ℃.
1H-NMR(300MHz,DMSO-d6))δ(ppm):9.13(d,J=5.4Hz,1H,ArH),8.58-8.52(m,3H,ArH),7.83(d,J=7.92Hz,1H,ArH),7.38-7.30(m.5H,ArH),5.45-5.41(m,1H,HOCH2 CH),4.69(d,J=18.5Hz,1H,CONCH 2),4.46(d,J=18.4Hz,1H,CONCH 2),4.26-4.14(m,2H,O 2CH),3.49(s,3H,S 3CH),0.73(S,9H,C 3 3(CH)),0.01(d,J=4.7Hz,6H,Si 3 2(CH)).
Synthesis of (S) -2- (2-hydroxy-1-phenylethyl) -6- (2- ((1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-24)
Dissolving 1-methyl-1H-pyrazole-5-amine (IX-2) (74mg,0.76mmol) in anhydrous tetrahydrofuran (3mL), cooling to-78 ℃, adding 1N LiHMDS (0.76mL,0.76mmol), stirring for 30min, dissolving compound X-2(200mg,0.38mmol) in anhydrous tetrahydrofuran (3mL), slowly dropwise adding the reaction solution at-78 ℃, reacting at-78 ℃ for about 1H, detecting by TLC (dichloromethane: methanol ═ 20:1) that raw material X-2 is completely reacted, evaporating the solvent under reduced pressure, adding ethyl acetate (5mL) to dissolve, adjusting pH to 2-3 with HCl ethyl acetate solution, continuing stirring for about 3H, detecting by TLC (dichloromethane: methanol ═ 15: 1) that the intermediate is completely reacted, evaporating the solvent under reduced pressure, separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-30: 1), yield 55mg of the target compound hydrochloride as a golden yellow solid: 31.3 percent and m.p.202.3-204.1 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.59(brs,1H,HN),8.61-8.60(m,1H,ArH),8.50(s,1H,ArH),8.40(d,J=7.8Hz,1H,ArH),7.80(d,J=8.3Hz,1H,ArH),7.63-7.61(m,1H,ArH),7.42-7.34(m,6H,ArH),6.37-6.33(m,1H,ArH),5.48-5.43(m,1H,HOCH2 CH),5.22-5.18(m,1H,HOCH2),4.78(d,J=18.3Hz,1H,CH 2NCO),4.47(d,J=18.4Hz,1H,CH 2NCO),4.12-4.03(m,2H,HO 2CHCH),3.77(s,3H,N 3CH).
13C-NMR(75MHz,DMSO-d6)δ(ppm):166.86,162.33,159.45,158.79,143.74,137.19,136.63,135.53,131.99,129.17,127.85,126.81,126.40,123.34,120.21,115.02,107.79,99.19,59.93,55.85,46.09,34.44.
HRMS(ESI):m/z[M+H]+.Calcd for C24H23N6O2:427.1882;Found:427.1867.
IR(cm-1):3396.69,3233.24,2923.20,1666.59,1558.09,1449.72,1407.88,1307.11,1269.59,1203.11,1062.83,1042.64,820.99,768.85,700.35,613.30.
Example 25
Synthesis of (S) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-25)
Synthesis of (S) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- (methylsulfonyl) pyrimidin-4-yl) isoindolin-1-one (VIII-8)
Dissolving compound VI (2,25g,5.83mmol), (S) -2-amino-2- (3-chlorophenyl) ethyl-1-ol (VII-5) (1g,5,83mmol) and triethylamine (1.6mL,11.65mmol) in acetonitrile (30mL), heating and refluxing for about 6h, until the raw materials react completely by TLC (dichloromethane: methanol ═ 20:1), evaporating the solvent under reduced pressure, adding methanol (10mL), cooling in ice bath to below 10 ℃, stirring for 30min, precipitating a solid, and performing suction filtration to obtain 1.72g of a white solid, wherein the yield is: 66.5 percent and m.p.71.8-73.5 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.16-9.14(m,1H,ArH),8.60-8.55(m,3H,ArH),7.84(d,J=8.8Hz,1H,ArH),7.44-7.34(m,4H,ArH),5.41-5.38(m,1H,CHCH2OH),5.23-5.21(m,1H,CHCH2 OH),4.78(d,J=19.1Hz,1H,CH 2N),4.51(d,J=18.9Hz,1H,CH 2N),4.08-4.00(m,2H,HO 2CHCH),3.51(s,3H,S 3CH).
Synthesis of (S) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-25)
Dissolving compound VIII-8(400mg,0.90mmol) and 4-aminotetrahydropyran (IX-1) (182mg,1.80mmol) in sec-butanol (3mL), heating to 125 ℃ with a sealed tube, reacting for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that the reaction of the raw material VIII-8 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain 220mg of a light yellow solid, yield: 52.6 percent and m.p.86.2-87.9 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):8.44(s,1H,ArH),8.40-8.39(m,1H,ArH),8.34(d,J=7.9Hz,1H,ArH),7.73(d,J=7.9Hz,1H,ArH),7.45(s,1H,ArH),7.38-7.30(m,3H,ArH),7.26-7.25(m,1H,ArH),5.42-5.38(m,1H,HOCH2 CH),5.24-5.23(m,1H,HOCH2CH),4.74(d,J=18.3Hz,1H,CONCH 2),4.46(d,J=18.0Hz,1H,CONCH 2),4.05-4.03(m,3H,HO 2CH,NHCH),3.92-3.89(m,2H,O(C 2H) 2),3.46-3.43(m,2H,O(C 2H) 2),1.92-1.88(m,2H,NHCH( 2CH) 2),1.62-1.51(m,2H,NHCH( 2CH) 2).
13C-NMR(75MHz,DMSO-d6)δ(ppm):168.30,161.98,159.72,150.22,144.68,141.21,137.42,133.74,132.98,130.99,130.51,128.04,127.61,126.51,124.52,121.45,106.05,66.57,61.07,56.84,47.66,47.10,32.82.
HRMS(ESI):m/z[M+H]+.Calcd for C25H26ClN4O3:465.1693;Found:465.1681.
IR(cm-1):3324.61,2952.51,2843.82,1674.86,1573.21,1524.39,1492.66,1454.00,1410.36,1367.09,1248.13,1200.68,1138.06,1085.54,1011.43,979.63,868.32,807.30,770.97,613.89.
Example 26
Synthesis of (S) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) -6- (2- ((1-methyl-1H-pyrazol-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-26)
Dissolving compound VIII-8(400mg,0.90mmol) and 1-methyl-1H-pyrazol-4-amine (IX-3) (175mg,1.80mmol) as raw materials in sec-butyl alcohol (3mL), heating to 125 ℃ in a sealed tube to react for about 12 hours, detecting that the raw material VIII-8 completely reacts by TLC (dichloromethane: methanol: 25:1), cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol: 200: 1-50: 1) to obtain a golden yellow solid 126mg, wherein the yield: 30.4 percent and m.p.207.8-209.6 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.58(s,1H,NH),8.53-8.51(m,1H,ArH),8.47(s,1H,ArH),8.39(d,J=8.4Hz,1H,ArH),7.92(s,1H,ArH),7.77(d,J=8.0Hz,1H,ArH),7.58(brs,1H,ArH),7.44-7.32(m,5H,ArH),5.41-5.37(m,1H,HOCH2 CH),5.24-5.20(m,1H,HOCH2CH),4.74(d,J=17.9Hz,1H,CONCH 2),4.48(d,J=18.0Hz,1H,CONCH 2),4.07-4.00(m,2H,HO 2CH),3.83(s,3H,N 3CH).
13C-NMR(75MHz,DMSO-d6)δ(ppm):168.27,163.47,160.10,159.89,144.91,141.20,137.36,133.76,133.09,131.06,130.61,130.45,128.08,127.62,126.55,124.70,123.44,121.52,121.21,107.41,61.16,56.80,47.68,39.09.
HRMS(ESI):m/z[M+H]+.Calcd for C24H22ClN6O2:461.1493;Found:461.1482.
IR(cm-1):3263.50,3064.40,1673.56,1626.77,1594.58,1573.68,1455.10,1434.10,1333.59,1300.77,1262.55,1201.99,1076.02,1040.00,803.48,768.99,691.89,616.86.
Example 27
Synthesis of 2- (3-chlorobenzyl) -6- (2- ((4-fluorophenyl) amino) pyrimidin-4-yl) isoindolin-1-one (I-27)
Dissolving a compound VIII-1(200mg,0.48mmol) and para-fluoroaniline IX-13(81mg,0.73mmol) in sec-butyl alcohol (5mL), sealing a tube, heating to 125 ℃ for reaction for about 12h, detecting by TLC (dichloromethane: methanol ═ 25:1) that the reaction of the raw material VIII-1 is complete, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, and separating by column chromatography (eluent: dichloromethane: methanol ═ 200: 1-50: 1) to obtain a gray solid 98mg, wherein the yield is as follows: 45.9 percent and 196.2 to 197.3 ℃.
1H-NMR(300MHz,DMSO-d6)δ(ppm):9.76(s,1H,NH),8.58(d,J=5.2Hz,1H,ArH),8.52(s,1H,ArH),8.40(dd,J=8.1,1.5Hz,1H,ArH),7.83(dd,J=8.8,4.8Hz,2H,ArH),7.74(d,J=8.0Hz,1H,ArH),7.54(d,J=5.2Hz,1H,ArH),7.19-7.13(m,3H,ArH),7.28(d,J=6.4Hz,1H,ArH),7.16(t,J=8.9Hz,2H,ArH),4.78(s,2H,CH2),4.50(s,2H,CH2).
13C-NMR(75MHz,DMSO-d6)δ(ppm):167.05,162.88,161.41,159.71(d,J=66.2Hz),159.22,144.36,139.97,136.79(d,J=8.5Hz),136.71,132.72,130.60,130.02,128.80,127.54,127.37,126.38,124.18,121.32,120.62(d,J=7.5Hz),114.96(d,J=22.1Hz),108.11,49.53,44.96.
HRMS(ESI):m/z[M+H]+.Calcd for C25H19ClFN4O:445.1226;Found:445.1235.
IR(cm-1):3465.54,3265.05,1703.97,1570.27,1537.36,1507.27,1463.32,1453.48,1430.72,1303.10,1199.33,829.97,801.87,764.94,711.27,612.21,548.43.
Example 28
Synthesis of 2- ((6-methylpyridin-2-yl) methyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-28)
Synthesis of methyl 2- (bromomethyl) -5- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) benzoate (XVII)
Compound III (1g,3.6mmol) and NBS (765mg,4.3mmol) were dissolved in carbon tetrachloride (15mL), benzoyl peroxide (12mg,0.036mmol) was added, heated to 80 ℃ for about 4h, and starting material III was detected by TLC (petroleum ether: ethyl acetate ═ 20:1) for completion of the reaction. Cooling to room temperature, filtering off insoluble substances, concentrating the filtrate to obtain red oily substance, and directly feeding into the next step without treatment.
Synthesis of 2- ((6-methylpyridin-2-yl) methyl) -6- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) isoindolin-1-one (XVIII)
6-methyl-2-aminomethylpyridine (VII-5) (194mg,1.8mmol) and triethylamine (182mg,1.8mmol) were dissolved in acetonitrile (10mL), and compound XVII (714mg,2mmol) was added dropwise, followed by heating to 80 ℃ to react for about 10 hours. TLC (dichloromethane: methanol 20:1) detects complete reaction of the XVII starting material, the solvent is evaporated to dryness under reduced pressure, ethyl acetate (15mL) is dissolved, washed with water, dried, filtered, the filtrate is evaporated under reduced pressure to remove the solvent, and the residue is separated by column chromatography (eluent: dichloromethane: methanol 75: 1) to give 157mg of a dark green oil, yield: 23.9 percent.
1H-NMR(300MHz,CDCl3)δ(ppm):8.40(s,1H,ArH),7.99(d,J=7.6Hz,1H ArH),7.57(t,J=7.7Hz,1H ArH),7.45(d,J=7.6Hz,1H ArH),7.14(d,J=7.8Hz,1H ArH),7.10(d,J=7.6Hz,1H ArH),4.97(s,2H,CH2),4.49(s,2H,CH2),2.60(s,3H,CH3),1.39(s,12H,CH3).
Synthesis of 6- (2-chloropyrimidin-4-yl) -2- ((6-methylpyridin-2-yl) methyl) isoindolin-1-one (XIX)
XVIII (500mg,1.4mmol), 2, 4-dichloropyrimidine (XX) (207mg,1.4mmol), palladium tetratriphenylphosphine (162mg,0.14mmol) and potassium phosphate (890mg,4.2mmol) were dissolved in a dioxane/water mixed solution (20/5mL), and the mixture was heated to 80 ℃ under nitrogen atmosphere to react for about 12 hours. TLC (dichloromethane: methanol 25:1) detects complete reaction of the starting material XVIII, cools to room temperature, filters, concentrates the filtrate under reduced pressure, and separates the residue by column chromatography (eluent: dichloromethane: methanol 50:1) to give 272mg of a yellow solid, yield: 55.4 percent and m.p.208.3-209.2 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.74-8.72(m,1H,ArH),8.54(s,1H,ArH),8.47(d,J=7.9Hz,1H,ArH),7.82-7.74(m,1H,ArH),7.65-7.57(m,2H,ArH),7.17(d,J=7.1Hz,1H,ArH),7.12(d,J=6.8Hz,1H,ArH),4.97(s,2H,CH2),4.60(s,2H,CH2),2.60(s,3H,CH3).
Synthesis of 2- ((6-methylpyridin-2-yl) methyl) -6- (2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) isoindolin-1-one (I-28)
Mixing XIX (50mg,0.14mmol), 4-aminotetrahydropyran (IX-1) (14mg,0.14mmol), Pd (OAc)2(3mg,0.014mmol), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (35mg,0.06mmol) and Cs2CO3Dissolving (68mg,0.21mmol) in dioxane (3mL), heating to 100 ℃ under the protection of nitrogen, reacting for about 12 hours, detecting complete reaction of the raw material XIX by TLC (dichloromethane: methanol 20:1), cooling the reaction liquid to room temperature, adding ethyl acetate for dilution, washing with water (2mL) and saturated salt water (2mL), drying an organic layer with anhydrous sodium sulfate, filtering, evaporating the filtrate under reduced pressure to remove the solvent, and separating residue by column chromatography (eluent: dichloromethane: methanol 200: 1-50: 1) to obtain a light yellow solid 24mg, wherein the yield is as follows: 41.3 percent and m.p.186.1-187.2 ℃.
1H-NMR(300MHz,CDCl3)δ(ppm):8.48(s,1H,ArH),8.36(d,J=5.3Hz,1H,ArH),8.27(d,J=7.7Hz,1H,ArH),7.59-7.52(m,2H,ArH),7.15(d,J=6.9Hz,1H,ArH),7.12-7.02(m,2H,ArH),5.32(s,1H,NH),4.95(s,2H,CH2),4.55(s,2H,CH2),4.29-4.10(m,1H,NHCH),4.04-4.00(m,2H,OCH2),3.63-3.56(m,2H,OCH2),2.58(s,3H,CH3),2.13-2.08(m,2H,NHCHCH2),1.67-1.57(m,2H,NHCHCH2).
13C-NMR(75MHz,CDCl3)δ(ppm):168.09,164.20,161.57,158.39,158.02,155.94,143.75,137.67,137.61,133.24,130.21,123.23,122.51,122.31,119.51,106.65,66.76,50.34,48.21,47.25,33.27,24.21.
HRMS(ESI):m/z[M+H]+.Calcd for C24H26N5O2:416.2087;Found:416.2081.
IR(cm-1):3437.25,3260.22,2959.37,2920.76,2839.89,1570.28,1593.93,1529.95,1462.49,1412.85,1369.02,1136.31,1109.76,872.80,822.00,773.29,612.73,531.56.
Example 29
The pharmacological experiments and results of the partial compounds prepared in the above are as follows:
first, ERK2 enzyme inhibition activity experiment
(I) an experimental method
All compounds were dissolved in DMSO to prepare 10mM stock, and then the compounds were added to the screening system at concentrations ranging from 0.05nM to 1. mu.M for detection, and diluted in 3-fold gradients, each concentration being in duplicate wells. The experimental results are converted into the activity percentage, dose-effect curves are drawn, and inhibition IC is calculated by GRAPHPAD PEISM 5 nonlinear regression50The value is obtained.
The enzyme reaction system consists of: 20mM Hepes (pH 7.5),10mM MgCl2,1mM EGTA,0.02%Brij35,0.02mg/mL BSA,0.1mM Na3VO42mM DTT,10 μ M ATP, kinase substrate; and simultaneously adding compounds to be screened with different concentrations to form a 50 mu l reaction system, reacting at room temperature for 2h, detecting the ADP content in the system by using a luciferase method, reacting for 5min, and detecting a chemiluminescence signal on an MD-SpectraMax M5 multifunctional enzyme-labeled instrument, wherein the value intensity of the chemiluminescence signal is in direct proportion to the inhibition of enzyme activity. Substituting the detected chemiluminescence signal value into the formula:
percentage of enzyme activity (%) { (Lu drug-Lu background)/(Lu enzyme-Lu background) } x 100%
IC of the compound was then calculated using Graphpad Prism5 software50The value is obtained.
Test results:
partial compounds of the invention were screened for ERK2 kinase inhibitory activity in vitro and the results are shown in table 1.
TABLE 1 IC of part of the tested compounds for ERK2 kinase50Value of
Figure BDA0001881258540000251
Figure BDA0001881258540000261
The results in Table 1 show that the compounds of the invention have better inhibitory activity on ERK2 kinase, wherein the compounds I-8 and I-10 have better activity and IC of ERK2 kinase500.7nM and 1.2nM, respectively. Meanwhile, the pharmacological activities of the S configuration and the R configuration of the compounds I-8 and I-10 are respectively compared, and the activity of the compound with the S configuration is far better than that of the compound with the R configuration.
Second, experiment of inhibition of human tumor cell proliferation
(I) an experimental method
Test grouping and drug concentration selection
All compound concentrations were selected: 1nM,10nM,100nM,500nM,1000 nM.
Cell Culture conditions (Cell Culture)
The cells were subcultured for 10-15 passages under conditions of medium containing penicillin (final concentration of 100U/mL), streptomycin (final concentration of 100. mu.g/mL), 10% FBS (CT26, Colo-205, WM-266-4: RPMI-1640; HCT116, HT 29: McCoy's 5 a; SW 626: L-15), when the cells fused to 90%, the old medium was discarded, the cells were washed with 2mL of PBS for 2 times, after discarding PBS, 2mL of 0.25% (w/v) Trypsin-0.53mM EDTA mixed digest was added, and when the cells were gently rounded, 2mL of complete medium was rapidly added to terminate the digestion, and the cells were collected by pipetting. Centrifuging at 800rpm and 4 deg.C for 5min, discarding supernatant, suspending cells with complete culture medium, culturing in bottles, and changing the culture medium every other day.
MTT assay
Cells in logarithmic growth phase were seeded at 1X 105 cells/well in 96-well plates, placed at 37 ℃ and 5% CO2After culturing under conditions until the cells are 90% confluent, the cells are synchronized by incubating for 2h with serum-free L-15 medium. Subsequently, the supernatant was discarded, and each of the compounds (final dilution concentration 1nM,10nM,100nM,500nM,1000nM after compound addition) in L-15 medium was added for 72h, and 20. mu.l of MTT solution (5mg/mL) was added to each well 4h before the end of incubation. After incubation, the supernatant from each well was discarded, 150. mu.L of DMSO was added to each well, and the cells were shaken on a cell shakerAnd (5) after 10min, fully dissolving the crystal, and measuring OD570 by using an enzyme labeling instrument.
The percentage (%) of proliferation inhibitory activity was (OD value administration-OD value background)/(OD value control-OD value background) x 100%
The concentration of the drug was plotted on the abscissa and the percentage of the proliferation inhibitory activity corresponding to each concentration was plotted on the ordinate, and the IC of each compound was calculated by nonlinear regression using Graphpd Prism550The value is obtained.
Test results:
selecting a part of compounds with better ERK2 kinase inhibition activity, and determining the antiproliferative capability of the compounds on tumor cell lines containing RAS-RAF-MEK-ERK mutation, namely human colon cancer cell Colo-205(BRAFV600E), human melanoma cell WM-266-4(BRAFV600D), human ovarian cancer cell SW-626(KRASG12V) and human colon cancer cell HCT-116(KRASG 13D). The experimental results show (Table 2) that the tested compound has certain proliferation inhibition capacity on four cell lines, and the inhibition activity is equivalent to that of the positive drug BVD-523.
TABLE 2 antiproliferative effect of partial compounds on four ERK pathway-activating cell lines
Figure BDA0001881258540000262
On this basis, the proliferation inhibitory ability of the above compounds on human chronic myelogenous leukemia cell K562, a tumor cell line without RAS-RAF-MEK-ERK mutation, was determined (Table 3). The results of the experiments show that the test compound and BVD-523 have inhibitory activity (IC) on K562 cell proliferation50All are more than 1 mu M) is obviously weaker than the 4 tumor cell lines containing RAS-RAF-MEK-ERK mutation, the antiproliferative activity is greatly weakened, and the tested compound has certain selectivity on kinase and can selectively inhibit ERK kinase.
TABLE 3 antiproliferative effect of partial compounds on ERK pathway inactive cell lines
Figure BDA0001881258540000271
Third, NCM-460 cytotoxicity assay
(I) an experimental method
Test grouping and drug concentration selection
All compound concentrations were selected: 10nM,100nM,500nM,1000nM,10000 nM.
Cell Culture conditions (Cell Culture)
Subculturing the cells under the condition of RPMI-1640 medium containing penicillin (final concentration of 100U/mL), streptomycin (final concentration of 100. mu.g/mL) and 10% FBS, discarding the old medium when the cells are fused to 90%, washing the cells with 2mL of PBS for 2 times, discarding the PBS, adding 2mL of 0.25% (w/v) Trypsin-0.53mM EDTA mixed digestive juice, observing under a microscope for about 30s, rapidly adding 2mL of complete medium to stop digestion after the cells are rounded, gently blowing and beating, and collecting the cells. Centrifuging at 800rpm and 4 deg.C for 5min, discarding supernatant, suspending cells with complete culture medium, culturing in bottles, and changing the culture medium every other day.
MTT assay
Cells in logarithmic growth phase were grown at 1X 105cells/well were seeded in 96-well plates at 37 ℃ with 5% CO2After culturing under conditions until the cells are 90% confluent, the cells are synchronized by incubating for 2h with serum-free L-15 medium. Subsequently, the supernatant was discarded, and each of them was incubated for 72 hours in RPMI-1640 medium containing each compound (10nM,100nM,1000nM,5000nM,10000nM) for 4 hours before the end of the incubation, and 20. mu.L of MTT solution (5mg/mL) was added to each well. After incubation is finished, discarding supernatant of each well, adding 150 mu L DMSO into each well, oscillating on a cell oscillator for 10min, and measuring OD (optical density) by using an enzyme-labeling instrument after crystals are fully dissolved570
Test results:
anti-proliferative Effect of some Compounds of Table 4 on NCM-460 cells
Figure BDA0001881258540000272
We selected compounds I-8 and I-10 and tested their antiproliferative potential on normal cells using the NCM-460 cell line (Table 4). The results show that two compounds grow on NCM-460 cellsAll showed very weak proliferation inhibition, IC50The values were all > 10000 nM.
From the above data we found that compounds I-8 and I-10 were not significantly toxic to normal cell growth of NCM-460.
Fourth, HCT-116 nude mouse transplanted tumor inhibition experiment
(I) an experimental method
Grouping conditions of experimental animals:
negative control group (model group): 10 patients were administered intraperitoneally with physiological saline containing 5% DMSO;
group I-8: 10, the administration dose is 50 mg/kg;
BVD-523 group: 10, the administration dose is 50 mg/kg;
the administration mode comprises the following steps: the preparation is administered by intragastric administration once a day.
Establishment of a transplantation tumor model and drug treatment:
HCT116 cells are subcultured for 10-15 generations under the condition of 1640 medium containing penicillin (final concentration of 100U/mL), streptomycin (final concentration of 100. mu.g/mL) and 10% FBS, when the cells are fused to 90%, the old medium is discarded, the cells are washed with 2mL of PBS for 2 times, 2mL of 0.25% trypsin-0.02% EDTA mixed digestive solution is added after the PBS is discarded, the mixture is observed under a microscope for about 30s, when the cells are rounded, 2mL of complete medium is rapidly added to stop digestion, the cells are lightly blown and collected. Centrifuging at 800rpm and 4 deg.C for 5min, discarding supernatant, suspending cells with complete culture medium, culturing in bottles, and changing the culture medium every other day.
Taking HCT116 cells in logarithmic growth phase, preparing into 5 × 10 under aseptic condition with serum-free culture medium6Selecting 30 healthy nude mice from the above experiment, feeding for one week to reach a weight of 20g, injecting 0.2mL of cell suspension into left armpit of each nude mouse, with cell concentration of 5 × 107and/mL. Measuring the diameter of the transplanted tumor of the nude mouse by using a vernier caliper until the tumor grows to 100mm3Animals were randomized after left and right as day 0 and then started dosing according to the dosing method in the group. The antitumor effect of the test compound was dynamically observed using a method of measuring the tumor diameter. The number of measurements of the tumor diameter isEvery 3 days. The administration volume was different for each group. After 30 days, the mice were sacrificed and the tumor mass was surgically removed and weighed. The formula for Tumor Volume (TV) is:
TV=1/2×a×b2
wherein a and b represent length and width, respectively.
Statistical analysis:
statistical differences between the data groups were determined to be significant using one-way ANOVA and Tukey's tests, with P values less than 0.05.
Test results:
we selected compound I-8, evaluated the in vivo anti-tumor activity of compound I-8 using a mouse transplantation tumor model of HCT-116 tumor cells, and compared it to the positive drug BVD-523.
As shown in figure 1, after the compound I-8 and the BVD-523 are continuously administered by gavage for 30 days at 50mg/kg/day, the increase of the volume and the weight of the transplanted tumor of the HCT-116 nude mice can be remarkably inhibited (p is less than 0.05), the tumor inhibition rate of the I-8 is 71 percent, the tumor inhibition rate of the BVD-523 is 54 percent, and the both do not influence the body weight of the mice. Experimental results show that the anti-tumor effect of the I-8 is better than that of a positive drug BVD-523 under the condition of intragastric administration of 50 mg/kg.

Claims (10)

1. An isoindolinone derivative represented by the general formula I or a pharmaceutically acceptable salt thereof:
Figure FDA0003351074190000011
wherein:
R1is selected from
Figure FDA0003351074190000012
Figure FDA0003351074190000013
Wherein X is selected from CH2O or CH-OH; r4Selected from H or C1-C6 alkyl, R5Selected from H, CH3、-CH2OH、-CH2CH2OH、-CH2CH2CH2OH or-CH2CH2CH2CH2OH; y is selected from O, NH or S; r6Selected from F, Cl, Br, CH3、NH2Or NHCOCH3
R2Is H or-CH2OH;
R3Is selected from
Figure FDA0003351074190000014
Wherein R is7Is H, F, Cl, Br or OCH3,R7Is mono-, di-or tri-substituted.
2. Isoindolinone derivative I or a pharmaceutically acceptable salt thereof according to claim 1, wherein:
R1is selected from
Figure FDA0003351074190000015
Figure FDA0003351074190000016
3. Isoindolinone derivative I or a pharmaceutically acceptable salt thereof according to claim 1, wherein:
R3is selected from
Figure FDA0003351074190000017
4. Isoindolinone derivative I or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 3, wherein: r2is-CH2OH, the compound I or a pharmaceutically acceptable salt thereof includes chiral isomers:
Figure FDA0003351074190000018
5. isoindolinone derivative I according to claim 1, or a pharmaceutically acceptable salt thereof, which is selected from I-1 to I-26:
Figure FDA0003351074190000021
Figure FDA0003351074190000031
Figure FDA0003351074190000041
6. isoindolinone derivative I or a pharmaceutically acceptable salt thereof according to claim 1, wherein the pharmaceutically acceptable salt is an acid addition salt of the compound of the general formula I, wherein the acid used for salt formation is: hydrogen chloride, hydrogen bromide, sulfuric acid, carbonic acid, oxalic acid, citric acid, succinic acid, tartaric acid, phosphoric acid, lactic acid, pyruvic acid, acetic acid, maleic acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid or ferulic acid.
7. A process for the preparation of isoindolinone derivatives according to claim 1, wherein compound I is prepared by substitution of compound VIII with compound IX:
Figure FDA0003351074190000051
8. a process for the preparation of isoindolinone derivatives according to claim 1, comprising the steps of:
Figure FDA0003351074190000052
reacting a compound VIII with tert-butyldimethylsilyl chloride TBSCl to prepare a compound X; then preparing a compound XI from the compound X and IX through substitution reaction; finally, compound XI is subjected to hydroxyl protecting group removal and salification to prepare compound I.A。
9. A pharmaceutical composition comprising a compound of formula I of claim 1 or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
10. Use of the isoindolinone derivative according to claim 1 or a pharmaceutically acceptable salt thereof for the preparation of an ERK kinase inhibitor medicament.
CN201811424530.3A 2018-11-27 2018-11-27 Isoindolinone-containing ERK inhibitor and preparation method and application thereof Active CN109608444B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201811424530.3A CN109608444B (en) 2018-11-27 2018-11-27 Isoindolinone-containing ERK inhibitor and preparation method and application thereof
PCT/CN2019/104110 WO2020107987A1 (en) 2018-11-27 2019-09-03 Erk inhibitor containing isoindoline, preparation method therefor and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811424530.3A CN109608444B (en) 2018-11-27 2018-11-27 Isoindolinone-containing ERK inhibitor and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN109608444A CN109608444A (en) 2019-04-12
CN109608444B true CN109608444B (en) 2022-02-11

Family

ID=66005187

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811424530.3A Active CN109608444B (en) 2018-11-27 2018-11-27 Isoindolinone-containing ERK inhibitor and preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN109608444B (en)
WO (1) WO2020107987A1 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109608444B (en) * 2018-11-27 2022-02-11 中国药科大学 Isoindolinone-containing ERK inhibitor and preparation method and application thereof
CN114829365B (en) * 2019-12-06 2023-04-25 南京明德新药研发有限公司 Thiazolo lactams as ERK inhibitors and uses thereof
CN113248482B (en) * 2020-02-10 2022-08-26 中国药科大学 Compound containing benzo five-membered heterocyclic structure and preparation method and application thereof
JP7296017B2 (en) * 2020-04-30 2023-06-21 メッドシャイン ディスカバリー インコーポレイテッド Compounds containing benzosultam
WO2022253186A1 (en) * 2021-06-02 2022-12-08 南京明德新药研发有限公司 Crystal form of dimethyl substituted thiazolopyrrolone compound and preparation method therefor
CN115594681B (en) * 2022-10-21 2024-06-18 广州六顺生物科技有限公司 Isoindoline-1, 3-dione derivative and application thereof
CN117164597B (en) * 2023-11-02 2024-02-09 深圳创元生物医药科技有限公司 SMTP-0 synthesis method

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1494541A (en) * 2001-02-09 2004-05-05 ��̩��˹ҩ��ɷ����޹�˾ Heterocyclic inhibitors of ERK 2 and uses thereof
CN1953974A (en) * 2004-05-14 2007-04-25 辉瑞产品有限公司 Pyrimidine derivatives for the treatment of abnormal cell growth
CN107922387A (en) * 2015-06-15 2018-04-17 阿沙纳生物科学公司 The heterocycle inhibitor of ERK1 and ERK2 and its application in treatment of cancer
CN108617166A (en) * 2015-10-21 2018-10-02 大冢制药株式会社 Kinases inhibitor benzolactam compounds
WO2018193410A1 (en) * 2017-04-20 2018-10-25 Otsuka Pharmaceutical Co., Ltd. 6-pyrimidin-isoindole derivative as erk1/2 inhibitor

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8338439B2 (en) * 2008-06-27 2012-12-25 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
US9938269B2 (en) * 2011-06-30 2018-04-10 Abbvie Inc. Inhibitor compounds of phosphodiesterase type 10A
TN2016000270A1 (en) * 2014-01-14 2017-10-06 Millennium Pharm Inc Heteroaryls and uses thereof.
CN109608444B (en) * 2018-11-27 2022-02-11 中国药科大学 Isoindolinone-containing ERK inhibitor and preparation method and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1494541A (en) * 2001-02-09 2004-05-05 ��̩��˹ҩ��ɷ����޹�˾ Heterocyclic inhibitors of ERK 2 and uses thereof
CN1953974A (en) * 2004-05-14 2007-04-25 辉瑞产品有限公司 Pyrimidine derivatives for the treatment of abnormal cell growth
CN107922387A (en) * 2015-06-15 2018-04-17 阿沙纳生物科学公司 The heterocycle inhibitor of ERK1 and ERK2 and its application in treatment of cancer
CN108617166A (en) * 2015-10-21 2018-10-02 大冢制药株式会社 Kinases inhibitor benzolactam compounds
WO2018193410A1 (en) * 2017-04-20 2018-10-25 Otsuka Pharmaceutical Co., Ltd. 6-pyrimidin-isoindole derivative as erk1/2 inhibitor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Fragment-Based Discovery of a Potent, Orally Bioavailable Inhibitor That Modulates the Phosphorylation and Catalytic Activity of ERK1/2;Tom D,等;《Journal of Medicinal Chemistry》;20180518;第61卷(第11期);4978-4992 *

Also Published As

Publication number Publication date
CN109608444A (en) 2019-04-12
WO2020107987A1 (en) 2020-06-04

Similar Documents

Publication Publication Date Title
CN109608444B (en) Isoindolinone-containing ERK inhibitor and preparation method and application thereof
EP2044051B1 (en) Pyridine and pyrazine derivatives as mnk kinase inhibitors
CN112608318B (en) Compound as protein kinase inhibitor and application thereof
JP2010523639A (en) Pharmaceutical compounds
KR20100016431A (en) Pharmaceutical compounds
CN102112449A (en) Phenyl or pyridinyl substituted indazoles derivatives
KR20100016432A (en) 2-morpholin-4-yl-pyrimidines as pi3k inhibitors
EP3536692B1 (en) Pyrazolopyrimidinyl inhibitors of ubiquitin-activating enzyme
CA2728408A1 (en) Substituted pyridazine carboxamide compounds as kinase inhibitor compounds
EP2933248A1 (en) Novel renin inhibitor
CN115536656A (en) Heterocyclic compounds as HPK1 inhibitors
CN117957219A (en) Pyridine derivative and application thereof
CN112457326A (en) Aromatic heterocyclic lactam compound, preparation method and application
KR102649802B1 (en) Pyrimidine derivatives
WO2020057669A1 (en) Aromatic heterocyclic compound with kinase inhibitory activity
CN111303128B (en) Polycyclic compound, preparation method and application thereof
CN113321645A (en) 5-aminoisoxazole derivative and application thereof in preparation of multi-kinase inhibitor
EP3189060A1 (en) Derivatives of macrocyclic n-aryl-2-amino-4-aryl-pyrimidine polyethers as inhibitors of ftl3 and jak
EP3988551A1 (en) Cdk kinase inhibitor
CN114075199A (en) JAK inhibitor compounds and uses thereof
EP3697786A1 (en) Substituted pyrrolopyridines as inhibitors of activin receptor-like kinase
KR102344185B1 (en) Novel Pim Kinase Inhibitor and Uses Thereof
CN113004252B (en) Aryl imidazole derivative and application thereof
WO2024109788A1 (en) Parp7 inhibitor, preparation method therefor, and use thereof
EP4293019A1 (en) 4-substituted (1h-benzo[d]imidazol-2-yl)-1h-pyrazoles as bub1 inhibitors useful for treating cancers

Legal Events

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