CN115197130B - Aryl urea derivative and preparation method and application thereof - Google Patents

Aryl urea derivative and preparation method and application thereof Download PDF

Info

Publication number
CN115197130B
CN115197130B CN202210891463.6A CN202210891463A CN115197130B CN 115197130 B CN115197130 B CN 115197130B CN 202210891463 A CN202210891463 A CN 202210891463A CN 115197130 B CN115197130 B CN 115197130B
Authority
CN
China
Prior art keywords
compound
synthesis
phenyl
aryl urea
reaction
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
CN202210891463.6A
Other languages
Chinese (zh)
Other versions
CN115197130A (en
Inventor
刘新华
刘明明
石静波
闫尧瑶
张兴星
王宇蒙
肖云
江瀚
佘梓濠
吴磊
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.)
Anhui Medical University
Original Assignee
Anhui Medical 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 Anhui Medical University filed Critical Anhui Medical University
Priority to CN202210891463.6A priority Critical patent/CN115197130B/en
Publication of CN115197130A publication Critical patent/CN115197130A/en
Application granted granted Critical
Publication of CN115197130B publication Critical patent/CN115197130B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/73Unsubstituted amino or imino radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention discloses an aryl urea derivative, a preparation method and application thereof, and relates to the technical field of pharmaceutical chemistry, wherein the structure of the aryl urea derivative is shown as formulas I and II:
Figure DDA0003767738190000011
the result of in vitro CDK8 kinase activity screening of the aryl urea compound shows that the aryl urea compound has stronger inhibitory activity on CDK 8; and the result of in vitro anti-tumor activity screening shows that the anti-tumor extract has stronger inhibition activity on tumor cells; the aryl urea compound disclosed by the invention is novel in structure, simple in synthesis process, high in product purity and good in application prospect.

Description

Aryl urea derivative and preparation method and application thereof
Technical field:
the invention relates to the technical field of pharmaceutical chemistry, in particular to an aryl urea derivative, a preparation method and application thereof.
The background technology is as follows:
current treatments for cancer are primarily chemotherapy, CAR-T cell immunotherapy and the use of targeted inhibitors. Chemotherapy causes the patient to have reduced immunity, is easy to infect and has great side effect. CAR-T treatment has many adverse effects such as cytokine release syndrome CRS, off-target effects, neurotoxicity, anaphylaxis, graft versus host disease, oncolytic syndrome, etc. Among these, CRS is the most serious, which is a fatal uncontrolled systemic inflammatory response. The increased risk of TRM often makes these patients unacceptable for optimal chemotherapy or stem cell transplantation. Thus, new targeted therapies offer the hope of potent anti-tumor activity, reducing the toxicity of off-target effects.
The cyclin-dependent protease 8 (cdk 8) was originally referred to as protein K35 and was found to be a putative kinase partner for cyclin C. The CDK8 gene is located on human chromosome 13q12.13, transcribed into a 53kDa protein containing 464 amino acids, whose kinase activity is regulated by association with Cyc-C, and the 13q12.13 chromosomal region is amplified in most colon cancers. Among the many cellular functions of CDK8, the most notable is the involvement in transcription. CDK8 and MED12, MED13, cys-C constitute an intermediate complex, a large multi-subunit protein complex, which is the center for regulating transcription in eukaryotes. In 2008, hahn and his co-workers in the dana-farbo cancer institute in the united states proposed CDK8 for the first time as an oncogene for colorectal cancer by regulating β -catenin, and later studies showed that CDK8 was overexpressed in melanoma, breast cancer, acute myeloid leukemia, pancreatic cancer, prostate cancer, and other cancers. Studies have shown that CDK8 kinase activity weakens natural killer cell defenses against malignant cells and inhibits tumor surveillance of pre-cells. CDK8 was validated by gene knockout as having an important role in these cancer survival. These evidence suggests that CDK8 carcinogenesis in these cancers and inhibition of CDK8 protein activity can inhibit tumorigenesis. Thus, potent and selective small molecule CDK8 inhibitors were found to be useful in the treatment of cancer as a new strategy for the treatment of cancer.
The invention comprises the following steps:
the technical problem to be solved by the invention is to provide aryl urea derivatives and a preparation method thereof, wherein the compounds belong to a potential class of 'CDK 8 Type II inhibitors', wherein the compound 29 has significance superior to that of a positive medicine sorafenib at a protein level and a cell level, and can be applied to research for preparing anti-tumor medicines.
One of the purposes of the present invention is to provide an aryl urea derivative, as shown in formulas I and II:
Figure BDA0003767738170000021
wherein R is 1 ,R 2 ,R 3 ,R 4 Selected from CH 3 、OCH 3 、CF 3 Any of the groups CN, H, F, cl, br;
R 5 any one group selected from 2-aminopyridine, 7-azaindole and derivatives thereof;
x is selected from CH 2 Any one of NH, O and S.
The structural formula of the aryl urea compound is shown as the following (compounds 1-54):
Figure BDA0003767738170000022
Figure BDA0003767738170000031
Figure BDA0003767738170000041
Figure BDA0003767738170000051
the second purpose of the invention is to provide a preparation method of the aryl urea derivative, which comprises the following steps:
(1) Nucleophilic substitution reaction is carried out on 2-nitro-5-bromopyridine and (3-hydroxyphenyl) carbamic acid tert-butyl ester to obtain an intermediate M1;
(2) Removing protecting groups from the intermediate M1 to obtain an intermediate M2;
(3) Intermediate M2 reacts with an aromatic amine derivative to obtain intermediate M3;
(4) Intermediate M3 undergoes a reduction reaction to give compounds 1-19.
The reaction equation is as follows:
Figure BDA0003767738170000052
(5) The synthesis route conditions of the compounds 20-42 are the same as those of the compounds 1-19, except that the raw material (3-hydroxyphenyl) carbamic acid tert-butyl ester is converted into (4-hydroxyphenyl) carbamic acid tert-butyl ester; wherein compound 29 is brominated to afford compound 53.
The reaction equation is as follows:
Figure BDA0003767738170000061
(6) Nucleophilic substitution reaction is carried out on the 5-hydroxy-7-azaindole and 3-fluoronitrobenzene to obtain an intermediate M7;
(7) Intermediate M7 undergoes a reduction reaction to obtain intermediate M8;
(8) Intermediate M8 and the aromatic amine derivative are reacted to give compounds 43-46.
The reaction equation is as follows:
Figure BDA0003767738170000062
(10) The synthesis route of the compound 47-50 is the same as that of the compound 43-46, and the raw material 3-fluoronitrobenzene is changed into 4-fluoronitrobenzene.
The reaction equation is as follows:
Figure BDA0003767738170000063
(11) Intermediate M4 is reduced to give intermediate M11;
(12) Intermediate M11 is acetylated to afford intermediate M12;
(13) Removing Boc from the intermediate M12 under trifluoroacetic acid to obtain an intermediate M13;
(14) Intermediate M13 and the aromatic amine derivative are reacted to give compounds 51-52.
The reaction equation is as follows:
Figure BDA0003767738170000071
(15) Compound 54 is synthesized in the same manner as in Compound 1, except that the starting material, tert-butyl (3-hydroxyphenyl) carbamate, is converted to tert-butyl (4-mercaptophenyl) carbamate.
The reaction equation is as follows:
Figure BDA0003767738170000072
the invention also provides a pharmaceutical composition containing the aryl urea derivative or the pharmaceutically acceptable salt thereof.
The invention also provides a pharmaceutical preparation, which comprises an active ingredient and pharmaceutically acceptable auxiliary materials and/or carriers, wherein the active ingredient contains the aryl urea derivative or pharmaceutically acceptable salts thereof.
The fifth object of the invention is to provide the use of the aryl urea derivative or the pharmaceutically acceptable salt thereof in preparing CDK8 inhibitor.
The invention aims at providing the application of the aryl urea derivative or the pharmaceutically acceptable salt thereof in preparing antitumor drugs. The tumor is selected from melanoma, breast cancer, acute myelogenous leukemia, pancreatic cancer, prostate cancer, colorectal cancer and the like.
The beneficial effects of the invention are as follows:
(1) The aryl urea compound disclosed by the invention can be used for biological or pharmacological phenomena, signal path conduction research involving CDK8 and evaluation of novel CDK8 inhibitors;
(2) The result of in vitro CDK8 kinase activity screening of the aryl urea compound shows that the aryl urea compound has stronger inhibitory activity on CDK8 and lower toxicity;
(3) The result of in vitro anti-tumor activity screening of the aryl urea compound shows that the aryl urea compound has stronger inhibition activity on tumor cells and lower toxicity;
(4) The aryl urea compound disclosed by the invention is novel in structure, simple in synthesis process, high in product purity and good in application prospect.
Description of the drawings:
FIG. 1 shows the results of an in vivo antitumor activity test of Compound 29 of the present invention.
The specific embodiment is as follows:
the invention is further described in connection with the following embodiments in order to make the technical means, the creation features, the achievement of the purpose and the effect of the invention easy to understand.
Example 1
Synthesis of tert-butyl (3- ((6-nitropyridin-3-yl) oxy) phenyl) carbamate (intermediate M1):
the compound 2-nitro-5-bromopyridine (10 g,49.27 mmol), tert-butyl (3-hydroxyphenyl) carbamate (10.3 g,49.27 mmol), cesium carbonate (20.9 g,64.05 mmol), N, N-dimethylformamide (100 mL) was reacted at 40℃for 20h. Adding water and ethyl acetate to stir after the reaction is finished, filtering with diatomite, and washing a filter cake with ethyl acetate; standing for separating, re-extracting the water phase with ethyl acetate, sequentially combining the organic phases, washing with saturated saline, drying with anhydrous sodium sulfate, and performing column chromatography to obtain 8.35g of yellow solid with a yield of 51.2%.
Synthesis of (3- ((6-nitropyridin-3-yl) oxy) aniline (intermediate M2)
Tert-butyl (8.3 g,25.07 mmol) carbamate (3- ((6-nitropyridin-3-yl) oxy) phenyl) compound M1 was added to dichloromethane (50 mL), stirred in an ice bath, trifluoroacetic acid (8.6 g,75.21 mmol) was added dropwise, and the mixture was reacted at 25℃for 2h; directly concentrating to obtain yellow solid, adding ethyl acetate and sodium bicarbonate water solution, stirring, separating liquid, drying and concentrating to obtain brownish red solid 5g, and the yield is 85.6%.
Synthesis of 1- (3- ((6-nitropyridin-3-yl) oxy) phenyl) -3-phenylurea (intermediate M3)
Triphosgene (100 mg,1.07 mmol) was first dissolved in tetrahydrofuran (10 mL) and the solution (compound aniline (100 mg,1.07 mmol) and N, N-diisopropylethylamine (276 mg,2.14 mmol) were dissolved in tetrahydrofuran (2 mL)) was added dropwise under ice-bath. Stirring for 40min in ice bath, adding compound M2 (3- ((6-nitropyridin-3-yl) oxy) aniline (230 mg,1.00 mmol) and reacting at 30deg.C for 5h, adding water and ethyl acetate after the reaction, stirring, standing for separating liquid, re-extracting the water phase with ethyl acetate, combining organic phases, washing with saturated common salt, drying with anhydrous sodium sulfate, and separating the product by column chromatography to obtain 263mg of yellow solid with 75% yield.
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3-phenylurea (Compound 1):
intermediate M3 (263 mg,0.75 mmol), methylene chloride (10 mL), methanol (10 mL), and saturated aqueous ammonium chloride (10 mL) were reacted at 30℃for 1 hour with 150mg of zinc powder added in an ice bath. Adding water and dichloromethane after the reaction, stirring, suction filtering, standing for separating liquid, sequentially extracting the aqueous phase with dichloromethane, combining the organic phases, washing with saturated saline, drying with anhydrous sodium sulfate, and performing column chromatography to obtain a product with 135mg of white solid and a yield of 56.3%. 1 H NMR(400MHz,DMSO-d 6 )δ8.75(s,1H),8.59(s,1H),7.77(d,J=2.8Hz,1H),7.42(d,J=7.6Hz,2H),7.27(t,J=7.9Hz,2H),7.23(dd,J=5.1,3.8Hz,1H),7.20(d,J=8.3Hz,1H),7.11(t,J=2.1Hz,1H),7.05(dd,J=8.1,1.0Hz,1H),6.97(t,J=7.3Hz,1H),6.55–6.47(m,2H),5.90(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 16 N 4 O 2 :321.1346;found:321.1349.
Example 2
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (o-tolyl) urea (Compound 2):
the synthesis procedure was as in example 1, substituting 2-methylaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ9.07(s,1H),7.85(s,1H),7.81(d,J=7.9Hz,1H),7.77(d,J=2.8Hz,1H),7.33–7.19(m,2H),7.15(dd,J=14.4,7.6Hz,2H),7.09(d,J=8.1Hz,1H),7.06(t,J=2.1Hz,1H),6.94(t,J=7.0Hz,1H),6.54(d,J=3.5Hz,1H),6.52–6.46(m,1H),5.89(s,2H),2.22(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 18 N 4 O 2 :335.1503;found:335.1502.
Example 3
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (2-fluorophenyl) urea (compound 3):
the synthesis procedure was as in example 1, substituting 2-fluoroaniline for aniline. 1 H NMR(400MHz,DMSO)δ9.14(s,1H),8.48(d,J=2.4Hz,1H),8.12(td,J=8.3,1.6Hz,1H),7.77(d,J=2.6Hz,1H),7.28–7.16(m,3H),7.16–7.08(m,2H),7.08–6.97(m,2H),6.53(ddd,J=9.3,5.6,0.6Hz,2H),5.92(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 F:339.1252;found:339.1297.
Example 4
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (2-chlorophenyl) urea (compound 4):
the synthesis procedure was the same as in example 1, substituting 2-chloroaniline for aniline. 1 H NMR(400MHz,DMSO)δ9.47(s,1H),8.26(s,1H),8.14(dd,J=8.3,1.5Hz,1H),7.78(d,J=2.8Hz,1H),7.45(dd,J=8.0,1.4Hz,1H),7.33–7.26(m,1H),7.26–7.19(m,2H),7.11–6.99(m,3H),6.60–6.46(m,2H),5.92(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 Cl:355.0956;found:355.0998.
Example 5
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-fluorophenyl) urea (compound 5):
the synthesis procedure was as in example 1, substituting 3-fluoroaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.82(d,J=2.1Hz,2H),7.77(d,J=2.9Hz,1H),7.45(dt,J=12.0,2.2Hz,1H),7.29(dd,J=15.2,8.1Hz,1H),7.25–7.18(m,2H),7.15–7.02(m,3H),6.78(td,J=8.4,2.4Hz,1H),6.63–6.39(m,2H),5.89(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 F:339.1252;found:339.1255.
Example 6
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-chlorophenyl) urea (compound 6):
the synthesis procedure was the same as in example 1, substituting 3-chloroaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ9.03(d,J=6.6Hz,2H),7.76(d,J=2.7Hz,1H),7.66(t,J=1.9Hz,1H),7.32–7.24(m,2H),7.24–7.19(m,2H),7.12(t,J=2.2Hz,1H),7.06(dd,J=8.1,1.1Hz,1H),7.03–6.98(m,1H),6.53(s,1H),6.51(s,1H),5.90(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 Cl:355.0956;found:355.0954.
Example 7\
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-bromophenyl) urea (compound 7):
the synthesis procedure was as in example 1, substituting 3-bromoaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.83(s,1H),8.80(s,1H),7.81(s,1H),7.76(d,J=2.7Hz,1H),7.29(d,J=8.2Hz,1H),7.22(dt,J=7.7,5.1Hz,3H),7.15(d,J=7.8Hz,1H),7.08(dd,J=8.7,5.0Hz,2H),6.61–6.45(m,2H),5.90(s,2H).HRMS(ESI):m/z[M+H]+calcd for C 18 H 15 N 4 O 2 Br:399.0451;found:399.0453.
Example 8
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-methylphenyl) urea (compound 8):
the synthesis procedure was as in example 1, substituting 3-methylaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.72(s,1H),8.50(s,1H),7.77(d,J=2.8Hz,1H),7.26(s,1H),7.22(dd,J=8.9,3.0Hz,2H),7.19(d,J=1.6Hz,1H),7.15(t,J=7.7Hz,1H),7.10(t,J=2.2Hz,1H),7.05(d,J=8.0Hz,1H),6.79(d,J=7.2Hz,1H),6.58–6.34(m,2H),5.90(s,2H),2.27(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 18 N 4 O 2 :335.1503;found:335.1503.
Example 9
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-methoxyphenyl) urea (Compound 9):
the synthesis procedure was as in example 1, substituting 3-methoxyaniline for aniline. 1 H NMR(400MHz,DMSO)δ8.87(s,1H),8.74(s,1H),7.76(d,J=2.7Hz,1H),7.22(dd,J=8.9,3.0Hz,2H),7.19–7.16(m,1H),7.15–7.13(m,1H),7.10(t,J=2.2Hz,1H),7.06(dd,J=8.1,1.0Hz,1H),6.92(dd,J=8.1,1.2Hz,1H),6.58–6.53(m,1H),6.50(ddd,J=5.5,3.7,3.1Hz,2H),5.89(s,2H),3.73(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 18 N 4 O 3 :351.1452;found:351.1453.
Example 10
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-cyanophenyl) urea (Compound 10):
the synthesis procedure was as in example 1, substituting 3-cyanoaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ9.51(s,1H),9.35(s,1H),7.94(s,1H),7.76(d,J=2.8Hz,1H),7.65(d,J=8.2Hz,1H),7.48(t,J=7.9Hz,1H),7.40(d,J=7.6Hz,1H),7.28–7.18(m,2H),7.16(s,1H),7.05(d,J=8.0Hz,1H),6.52(d,J=8.8Hz,2H),5.89(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 15 N 5 O 2 :346.1299;found:346.1295.
Example 11
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-trifluoromethylphenyl) urea (Compound 11):
the synthesis procedure was as in example 1, substituting 3-trifluoromethylaniline for aniline. 1 H NMR(500MHz,DMSO-d 6 )δ8.97(s,1H),8.88(s,1H),7.95(s,1H),7.77(d,J=1.1Hz,1H),7.58(d,J=8.2Hz,1H),7.52(t,J=7.9Hz,1H),7.33(d,J=7.6Hz,1H),7.30–7.21(m,2H),7.17–7.04(m,2H),6.61–6.49(m,2H),5.88(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 15 N 4 O 2 F 3 :389.1220;found:389.1221.
Example 12
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-fluorophenyl) urea (Compound 12):
the synthesis procedure was as in example 1, substituting 4-fluoroaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.74(s,1H),8.62(s,1H),7.76(d,J=2.8Hz,1H),7.48–7.39(m,2H),7.24–7.17(m,2H),7.16–7.08(m,3H),7.04(dd,J=8.1,1.1Hz,1H),6.51(d,J=8.7Hz,2H),5.89(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 F:339.1252;found:339.1254.
Example 13
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-fluorophenyl) urea (Compound 13):
the synthesis procedure was the same as in example 1, substituting 4-chloroaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.83(s,1H),8.78(s,1H),7.76(d,J=2.8Hz,1H),7.50–7.40(m,2H),7.35–7.28(m,2H),7.25–7.17(m,2H),7.11(t,J=2.1Hz,1H),7.04(dd,J=8.1,1.1Hz,1H),6.56–6.48(m,2H),5.89(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 Cl355.0956;found:355.0955.
Example 14
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-methylphenyl) urea (Compound 14):
the synthesis procedure was as in example 1, substituting 4-methylaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.69(s,1H),8.47(s,1H),7.76(d,J=2.9Hz,1H),7.31(s,1H),7.29(d,J=2.4Hz,1H),7.25–7.20(m,1H),7.19(d,J=8.2Hz,1H),7.10–7.07(m,2H),7.06(s,1H),7.03(dd,J=8.1,1.1Hz,1H),6.50(ddd,J=4.8,4.0,2.8Hz,2H),5.88(s,2H),2.24(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 18 N 4 O 2 :335.1503;found:335.1507.
Example 15
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-fluoro-3-trifluoromethylphenyl) urea (Compound 15):
the synthesis procedure was as in example 1, substituting 4-fluoro-3-trifluoromethylaniline for aniline. 1 H NMR(500MHz,DMSO-d 6 )δ9.53(s,1H),9.38(s,1H),8.02–7.94(m,1H),7.90–7.83(m,2H),7.70–7.62(m,1H),7.44(t,J=9.7Hz,1H),7.32(dd,J=15.2,7.0Hz,2H),7.16(dd,J=8.2,0.8Hz,1H),7.10(d,J=9.4Hz,1H),6.67(dd,J=8.1,2.2Hz,1H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 15 N 4 O 2 F 3 :407.1126;found:407.1122.
Example 16
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-chloro-3-trifluoromethylphenyl) urea (Compound 16):
the synthesis procedure was as in example 1, substituting 4-chloro-3-trifluoromethylaniline for aniline. 1 H NMR(500MHz,DMSO-d 6 )δ9.96(s,1H),9.65(s,1H),8.05(d,J=2.3Hz,1H),7.93(d,J=2.7Hz,1H),7.86(dd,J=9.6,2.7Hz,1H),7.66–7.62(m,1H),7.61(d,J=8.8Hz,1H),7.33(t,J=2.1Hz,1H),7.29(t,J=8.2Hz,1H),7.13(dd,J=8.1,1.1Hz,1H),7.08(d,J=9.6Hz,1H),6.65(dd,J=8.1,2.0Hz,1H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 14 N 4 O 2 F 3 Cl:423.0830;found:423.0831.
Example 17
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-chloro-4-methylphenyl) urea (Compound 17):
the synthesis procedure was as in example 1, substituting 3-chloro-4-methylaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.78(s,1H),8.68(s,1H),7.76(d,J=2.8Hz,1H),7.64(d,J=2.1Hz,1H),7.27–7.13(m,4H),7.07(dt,J=9.2,1.6Hz,2H),6.57–6.45(m,2H),5.89(s,2H),2.26(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 17 N 4 O 2 Cl:369.1113;found:369.1111.
Example 18
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-chloro-4-fluorophenyl) urea (Compound 18):
the synthesis procedure was as in example 1 substituting 3-chloro-4-fluoroaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.86(s,1H),8.82(s,1H),7.75(dd,J=6.7,2.3Hz,2H),7.35–7.27(m,2H),7.24–7.18(m,2H),7.08(dt,J=9.1,1.6Hz,2H),6.56–6.47(m,2H),5.89(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 14 N 4 O 2 FCl:373.0862;found:373.0861.
Example 19
Synthesis of 1- (3- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3, 4-dichlorophenyl) urea (Compound 19):
the synthesis procedure was the same as in example 1, substituting 3, 4-dichlorobenzylamine for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.91(d,J=10.8Hz,2H),7.83(d,J=2.5Hz,1H),7.76(d,J=2.7Hz,1H),7.51(d,J=8.8Hz,1H),7.32(dd,J=8.8,2.5Hz,1H),7.26–7.15(m,2H),7.08(dt,J=9.1,1.6Hz,2H),6.60–6.38(m,2H),5.89(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 14 N 4 O 2 Cl 2 :389.0567;found:389.0565.
Example 20
Synthesis of tert-butyl (4- ((6-nitropyridin-3-yl) oxy) phenyl) carbamate (intermediate M4):
the compound 2-nitro-5-bromopyridine (10 g,49.27 mmol), tert-butyl (4-hydroxyphenyl) carbamate (10.3 g,49.27 mmol), cesium carbonate (20.9 g,64.05 mmol), N, N-dimethylformamide (100 mL) was reacted at 40℃for 20h. Adding water and ethyl acetate to stir after the reaction is finished, filtering with diatomite, and washing a filter cake with ethyl acetate; standing for separating, re-extracting the water phase with ethyl acetate, sequentially combining the organic phases, washing with saturated saline, drying with anhydrous sodium sulfate, and performing column chromatography to obtain a yellow solid with a yield of 10.19g and 62.5%.
Synthesis of (4- ((6-nitropyridin-3-yl) oxy) aniline (intermediate M5)
Tert-butyl (10.19 g,30.78 mmol) carbamate (4- ((6-nitropyridin-3-yl) oxy) phenyl) compound M4 (100 mL) was added to dichloromethane, stirred in an ice bath, trifluoroacetic acid (11.43 g,100 mmol) was added dropwise, and the mixture was reacted at 25℃for 2h; directly concentrating to obtain yellow solid, adding ethyl acetate and sodium bicarbonate water solution, stirring, separating liquid, drying and concentrating to obtain brownish red solid 7.5g, and the yield is 81.5%.
Synthesis of 1- (4- ((6-nitropyridin-3-yl) oxy) phenyl) -3-phenylurea (intermediate M6)
Triphosgene (100 mg,1.07 mmol) was first dissolved in tetrahydrofuran (10 mL) and the solution (compound aniline (100 mg,1.07 mmol) and N, N-diisopropylethylamine (276 mg,2.14 mmol) were dissolved in tetrahydrofuran (2 mL)) was added dropwise under ice-bath. Stirring for 40min in ice bath, adding compound M5 (4- ((6-nitropyridin-3-yl) oxy) aniline (230 mg,1.00 mmol) and reacting at 30deg.C for 5h, adding water and ethyl acetate after the reaction, stirring, standing for separating liquid, re-extracting the water phase with ethyl acetate, combining organic phases, washing with saturated common salt, drying with anhydrous sodium sulfate, and separating the product by column chromatography to obtain 245mg of yellow solid with a yield of 70%.
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3-phenylurea (Compound 20):
to a saturated aqueous ammonium chloride solution (10 mL) of intermediate M6 (245 mg,0.70 mmol), methylene chloride (10 mL), methanol (10 mL) was added 150mg of zinc powder in an ice bath, and the mixture was reacted at 30℃for 1 hour. Adding water and dichloromethane after the reaction, stirring, suction filtering, standing for separating liquid, sequentially extracting the aqueous phase with dichloromethane, combining the organic phases, washing with saturated saline, drying with anhydrous sodium sulfate, and performing column chromatography to obtain a product, wherein 100mg of white solid is obtained. 1 H NMR(400MHz,DMSO-d 6 )δ8.59(d,J=5.0Hz,2H),7.73(d,J=2.4Hz,1H),7.42(dd,J=17.9,8.4Hz,4H),7.27(t,J=7.7Hz,2H),7.20(dd,J=8.8,2.7Hz,1H),6.97(d,J=7.2Hz,1H),6.87(d,J=8.8Hz,2H),6.51(d,J=8.9Hz,1H),5.89(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 16 N 4 O 2 :321.1346;found:321.1345.
Example 21
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (2-fluorophenyl) urea (Compound 21):
the procedure of the synthesis was as in example 20,the aniline was replaced with 2-fluoroaniline. 1 H NMR(400MHz,DMSO)δ9.00(s,1H),8.49(d,J=2.4Hz,1H),8.15(td,J=8.3,1.5Hz,1H),7.73(d,J=2.8Hz,1H),7.45–7.35(m,2H),7.23(ddd,J=11.7,8.2,1.3Hz,1H),7.20–7.16(m,1H),7.13(t,J=7.7Hz,1H),7.03–6.96(m,1H),6.92–6.82(m,2H),6.49(d,J=8.9Hz,1H),5.85(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 F:339.1252;found:339.1190.
Example 22
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (2-chlorophenyl) urea (Compound 22):
the synthesis procedure was as in example 20 substituting 2-chloroaniline for aniline. 1 H NMR(400MHz,DMSO)δ9.36(s,1H),8.25(s,1H),8.17(dd,J=8.3,1.5Hz,1H),7.74(d,J=2.6Hz,1H),7.43(ddd,J=14.7,7.4,1.8Hz,3H),7.29(s,1H),7.19(dd,J=8.9,3.0Hz,1H),7.03(dd,J=7.7,1.3Hz,1H),6.91–6.84(m,2H),6.49(dd,J=8.9,0.4Hz,1H),5.86(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 Cl:355.0956;found:355.0999.
Example 23
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-fluorophenyl) urea (Compound 23):
the synthesis procedure was as in example 20 substituting 3-fluoroaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ9.91(s,1H),9.59(s,1H),7.72(d,J=2.7Hz,1H),7.50(d,J=12.0Hz,1H),7.40(d,J=8.9Hz,2H),7.28(dd,J=15.3,8.0Hz,1H),7.17(dd,J=8.8,2.8Hz,1H),7.10(d,J=8.1Hz,1H),6.86(d,J=8.9Hz,2H),6.74(td,J=8.4,2.0Hz,1H),6.50(d,J=8.9Hz,1H),5.86(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 F:339.1252;found:339.1257.
Example 24
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-chlorophenyl) urea (Compound 24):
the synthesis procedure was as in example 20 substituting 3-chloroaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ10.06(s,1H),9.71(s,1H),7.79–7.63(m,2H),7.40(d,J=8.9Hz,2H),7.28(d,J=6.1Hz,2H),7.17(dd,J=8.9,2.8Hz,1H),7.03–6.92(m,1H),6.85(d,J=8.9Hz,2H),6.50(d,J=8.9Hz,1H),5.85(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 Cl:355.0956;found:355.0959.
Example 25
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-bromophenyl) urea (Compound 25):
the synthesis procedure was as in example 20 substituting 3-bromoaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.82(s,1H),8.67(s,1H),7.85(t,J=1.9Hz,1H),7.73(d,J=2.9Hz,1H),7.40(d,J=2.1Hz,1H),7.39(d,J=2.2Hz,1H),7.34–7.27(m,1H),7.23(t,J=8.0Hz,1H),7.18(dd,J=8.9,3.0Hz,1H),7.15–7.11(m,1H),6.87(d,J=2.1Hz,1H),6.86(d,J=2.1Hz,1H),6.49(d,J=8.9Hz,1H),5.84(s,2H).HRMS(ESI):m/z[M+H]+calcd for C 18 H 15 N 4 O 2 Br:399.0451;found:399.0451.
Example 26
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-methylphenyl) urea (Compound 26):
the synthesis procedure was as in example 20 substituting 3-methylaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ9.71(s,1H),9.63(s,1H),7.72(d,J=2.8Hz,1H),7.42(d,J=2.1Hz,1H),7.40(d,J=2.1Hz,1H),7.27(d,J=7.3Hz,2H),7.17(dd,J=8.9,3.0Hz,1H),7.13(d,J=7.5Hz,1H),6.85(d,J=2.0Hz,1H),6.83(d,J=2.1Hz,1H),6.74(d,J=7.2Hz,1H),6.52–6.47(m,1H),5.85(s,2H),2.26(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 18 N 4 O 2 :335.1503;found:335.1506.
Example 27
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-methoxyphenyl) urea (Compound 27):
the synthesis procedure was as in example 20 substituting 3-methoxyaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ9.09(d,J=15.3Hz,2H),7.73(d,J=2.0Hz,1H),7.41(d,J=8.6Hz,2H),7.21(s,1H),7.16(dd,J=15.5,5.6Hz,2H),6.95(s,1H),6.85(d,J=8.6Hz,2H),6.51(dd,J=13.4,8.6Hz,2H),5.84(s,2H),3.72(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 18 N 4 O 3 :351.1452;found:351.1457.
Example 28
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-cyanophenyl) urea (Compound 28):
the synthesis procedure was as in example 20 substituting 3-cyanoaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.96(s,1H),8.76(s,1H),8.01–7.90(m,1H),7.74(d,J=2.9Hz,1H),7.70–7.63(m,1H),7.48(t,J=7.9Hz,1H),7.41(d,J=9.0Hz,3H),7.18(dd,J=8.9,2.9Hz,1H),6.93–6.78(m,2H),6.49(d,J=8.9Hz,1H),5.84(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 15 N 5 O 2 :346.1299;found:346.1298.
Example 29
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-trifluoromethylphenyl) urea (Compound 29):
the synthesis procedure was as in example 20 substituting 3-trifluoromethylaniline for aniline. 1 H NMR(500MHz,DMSO-d 6 )δ9.56(s,1H),9.30(s,1H),8.00(s,1H),7.83(d,J=9.6Hz,1H),7.78(d,J=2.6Hz,1H),7.61(d,J=8.2Hz,1H),7.56–7.47(m,3H),7.31(d,J=7.7Hz,1H),7.12–7.01(m,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 15 N 4 O 2 F 3 :389.1220;found:389.1220.
Example 30
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-fluorophenyl) urea (Compound 30):
the synthesis procedure was as in example 20 substituting 4-fluoroaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.68(s,1H),8.62(s,1H),7.72(d,J=2.9Hz,1H),7.48–7.42(m,2H),7.39(d,J=2.2Hz,1H),7.39–7.36(m,1H),7.17(dd,J=8.9,3.0Hz,1H),7.14–7.07(m,2H),6.86(d,J=2.1Hz,1H),6.85(d,J=2.1Hz,1H),6.49(d,J=8.9Hz,1H),5.82(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 F:339.1252;found:339.1252.
Example 31
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-chlorophenyl) urea (Compound 31):
the synthesis procedure was as in example 20 substituting 4-chloroaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.81(s,1H),8.68(s,1H),7.80(dd,J=6.8,2.0Hz,1H),7.73(d,J=2.8Hz,1H),7.43–7.39(m,1H),7.39–7.36(m,1H),7.30(td,J=6.7,4.2Hz,2H),7.18(dd,J=8.9,3.0Hz,1H),6.92–6.73(m,2H),6.49(d,J=8.9Hz,1H),5.84(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 15 N 4 O 2 Cl355.0956;found:355.0958.
Example 32
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-methylphenyl) urea (Compound 32):
the synthesis procedure was as in example 20 substituting 4-methylaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.54(s,1H),8.49(s,1H),7.73(d,J=2.8Hz,1H),7.40(d,J=2.2Hz,1H),7.38(d,J=2.2Hz,1H),7.33(s,1H),7.31(s,1H),7.20(dd,J=8.9,3.0Hz,1H),7.08(s,1H),7.06(s,1H),6.87(d,J=2.1Hz,1H),6.85(d,J=2.2Hz,1H),6.51(d,J=8.9Hz,1H),5.90(s,2H),2.24(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 18 N 4 O 2 :335.1503;found:335.1504.
Example 33
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-methoxyphenyl) urea (Compound 33):
the synthesis procedure was as in example 20 substituting 4-methoxyaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.50(s,1H),8.40(s,1H),7.72(d,J=2.8Hz,1H),7.36(dd,J=16.5,9.0Hz,4H),7.20(dd,J=8.9,2.9Hz,1H),6.86(dd,J=8.9,1.1Hz,4H),6.52(d,J=8.9Hz,1H),5.91(s,2H),3.71(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 18 N 4 O 3 :351.1452;found:351.1451.
Example 34
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-trifluoromethylphenyl) urea (Compound 34):
the synthesis procedure was as in example 20 substituting 4-trifluoromethylaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ9.08(s,1H),8.75(s,1H),7.73(d,J=2.9Hz,1H),7.64(q,J=9.0Hz,4H),7.48–7.41(m,1H),7.41–7.38(m,1H),7.18(dd,J=8.9,3.0Hz,1H),6.99–6.87(m,1H),6.87–6.83(m,1H),6.49(d,J=8.9Hz,1H),5.84(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 15 N 4 O 2 F 3 :389.1220;found:389.1223.
Example 35
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-fluoro-3- (trifluoromethyl) phenyl) urea (Compound 35):
the synthesis procedure was as in example 20 substituting 4-fluoro-3-trifluoromethylaniline for aniline. 1 H NMR(500MHz,DMSO-d 6 )δ9.56(s,1H),9.28(s,1H),8.06–7.96(m,1H),7.83(dd,J=9.6,1.8Hz,1H),7.81–7.74(m,1H),7.70–7.62(m,1H),7.50(d,J=8.9Hz,2H),7.43(t,J=9.8Hz,1H),7.08(d,J=9.6Hz,1H),7.06–7.02(m,2H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 15 N 4 O 2 F 3 :407.1126;found:407.1126.
Example 36
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (4-chloro-3- (trifluoromethyl) phenyl) urea (Compound 36):
the synthesis procedure was as in example 20 substituting 4-chloro-3-trifluoromethylaniline for aniline. 1 H NMR(500MHz,DMSO-d 6 )δ9.94(s,1H),9.50(s,1H),8.10(d,J=2.4Hz,1H),7.86–7.80(m,2H),7.64(dd,J=8.8,2.4Hz,1H),7.60(d,J=8.8Hz,1H),7.49(d,J=9.0Hz,2H),7.10–7.01(m,3H),3.44(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 14 N 4 O 2 F 3 Cl:423.0830;found:423.0831.
Example 37
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-chloro-4-fluorophenyl) urea (Compound 37):
the synthesis procedure was as in example 20 substituting 3-chloro-4-fluoroaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.82(s,1H),8.68(s,1H),7.73(d,J=2.7Hz,1H),7.49(d,J=2.1Hz,1H),7.47(d,J=2.1Hz,1H),7.40(d,J=2.2Hz,1H),7.38(d,J=2.2Hz,1H),7.32(d,J=2.1Hz,1H),7.30(d,J=2.0Hz,1H),7.18(dd,J=8.9,3.0Hz,1H),6.87(d,J=2.2Hz,1H),6.85(d,J=2.2Hz,1H),6.54–6.29(m,1H),5.83(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 14 N 4 O 2 FCl:373.0862;found:373.0861.
Example 38
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3, 4-dichlorophenyl) urea (Compound 38):
the synthesis procedure was the same as in example 20 substituting 3, 4-dichlorobenzylamine for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.94(s,1H),8.73(s,1H),7.88(d,J=2.5Hz,1H),7.73(d,J=2.8Hz,1H),7.50(d,J=8.8Hz,1H),7.40(d,J=2.1Hz,1H),7.38(d,J=2.1Hz,1H),7.32(dd,J=8.8,2.5Hz,1H),7.18(dd,J=8.9,3.0Hz,1H),6.91–6.83(m,2H),6.49(d,J=8.9Hz,1H),5.85(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 14 N 4 O 2 Cl 2 :389.0567;;found:389.0566.
Example 39
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3-chloro-4 methylphenyl) urea (Compound 39):
the synthesis procedure was as in example 20 substituting 3-chloro-4 methylaniline for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.99(s,1H),8.88(s,1H),7.73(d,J=2.8Hz,1H),7.68(d,J=2.0Hz,1H),7.42–7.39(m,1H),7.39–7.36(m,1H),7.22(d,J=8.4Hz,1H),7.20–7.14(m,2H),6.88–6.85(m,1H),6.85–6.82(m,1H),6.49(d,J=8.9Hz,1H),5.84(s,2H),2.25(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 17 N 4 O 2 Cl:369.1113;found:369.1115.
Example 40
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (2, 3-dichlorophenyl) urea (Compound 40):
the synthesis procedure was the same as in example 20 substituting 2, 3-dichlorobenzylamine for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ9.42(s,1H),8.41(s,1H),8.17(dd,J=8.1,1.7Hz,1H),7.74(d,J=2.9Hz,1H),7.42(d,J=2.1Hz,1H),7.40(d,J=2.1Hz,1H),7.32(t,J=8.1Hz,1H),7.27(dd,J=8.0,1.7Hz,1H),7.18(dd,J=8.9,2.9Hz,1H),6.95–6.78(m,2H),6.49(d,J=8.9Hz,1H),5.84(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 14 N 4 O 2 Cl 2 :389.0567;found:389.0568.
Example 41
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (3, 5-dichlorophenyl) urea (Compound 41):
the synthesis procedure was the same as in example 20 substituting 3, 5-dichlorobenzylamine for aniline. 1 H NMR(400MHz,DMSO-d 6 )δ8.99(s,1H),8.79(s,1H),7.73(d,J=2.8Hz,1H),7.53(d,J=1.8Hz,2H),7.41(d,J=2.1Hz,1H),7.40–7.37(m,1H),7.18(dd,J=8.9,3.0Hz,1H),7.14(t,J=1.8Hz,1H),6.94–6.87(m,1H),6.87–6.84(m,1H),6.49(d,J=8.9Hz,1H),5.84(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 14 N 4 O 2 Cl 2 :389.0567;found:389.0568.
Example 42
Synthesis of 1- (4- ((6-aminopyridin-3-yl) oxy) phenyl) -3- (6- (trifluoromethyl) pyridin-2-yl) urea (Compound 42):
the synthesis procedure was as in example 20 substituting 6- (trifluoromethyl) -2 aminopyridine for aniline. 1 H NMR(500MHz,DMSO-d 6 )δ10.08(d,J=9.4Hz,1H),9.84(d,J=3.1Hz,1H),8.09–8.00(m,2H),7.91–7.72(m,2H),7.51(t,J=8.2Hz,3H),7.17–7.03(m,3H).HRMS(ESI):m/z[M+H] + calcd for C 18 H 14 N 5 O 2 F 3 :390.1172;found:390.1177
Example 43
Synthesis of 5- (3-nitrophenoxy) -1H-pyrrolo [2,3-b ] pyridine (intermediate M7):
5-hydroxy-7-azaindole (2.5 g,17.73 mmol) and 3-fluoronitrobenzene (2.62 g,19.50 mmol) were dissolved in N, N-dimethylformamide (50 mL), and then potassium carbonate (4.9 g,35.46 mmol) was added and the reaction stirred at 80℃for 14h. After the completion of the reaction, ethyl acetate (300 mL) was added, followed by suction filtration, washing of the filtrate with saturated brine, drying over anhydrous sodium sulfate, and column chromatography to give intermediate M7.
Synthesis of 5- (3-aminophenoxy) -1H-pyrrolo [2,3-b ] pyridine (intermediate M8)
Intermediate M7 (2 g,7.84 mmol) and CH 3 OH (40 mL) was added to the flask, and then Raney nickel (300 mg) was added and the reaction stirred at 25℃under hydrogen balloon pressure for 6h. And after the reaction is finished, carrying out suction filtration, and carrying out column chromatography on the filtrate to obtain an intermediate M8.
Synthesis of 1- (3- ((1H-pyrrolo [2,3-b ] pyridin-5-yl) oxy) phenyl) -3- (3- (trifluoromethyl) phenyl) urea (Compound 43)
Triphosgene (147 mg,0.50 mmol) was dissolved in tetrahydrofuran (15 mL) and then a solution of 3-trifluoromethylaniline (217 mg,1.35 mmol) and DIPEA (387 mg,3 mmol) in tetrahydrofuran (5 mL) was added under ice-bath. The reaction was stirred at 0deg.C for one hour, and intermediate M8 (281mg, 1.25 mmol) was added to the reaction, followed by stirring at 25deg.C for 5 hours. The reaction mixture was quenched with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate, and subjected to column chromatography to give compound 43. 1 H NMR(500MHz,DMSO-d 6 )δ11.75(s,1H),8.91(d,J=37.6Hz,2H),8.09(s,1H),7.94(s,1H),7.72(s,1H),7.60–7.52(m,2H),7.49(t,J=7.7Hz,1H),7.30(d,J=7.2Hz,1H),7.26(t,J=8.0Hz,1H),7.15(d,J=13.8Hz,2H),6.61(d,J=7.5Hz,1H),6.45(s,1H).HRMS(ESI):m/z[M+H] + calcd for C 21 H 15 N 4 O 2 F 3 :413.1220found:413.1223.
Example 44
Synthesis of 1- (3- ((1H-pyrrolo [2,3-b ] pyridin-5-yl) oxy) phenyl) -3- (3 chlorophenyl) urea (Compound 44):
the procedure is as in example 43, substituting 3-chloroaniline for 3-trifluoromethylaniline. 1 H NMR(400MHz,DMSO-d 6 )δ11.76(s,1H),8.84(s,1H),8.80(s,1H),8.09(d,J=2.6Hz,1H),7.72(d,J=2.5Hz,1H),7.65(t,J=1.9Hz,1H),7.57–7.51(m,1H),7.26(ddd,J=12.8,9.9,7.2Hz,3H),7.16–7.09(m,2H),7.04–6.97(m,1H),6.63–6.56(m,1H),6.45(dd,J=3.4,1.9Hz,1H).HRMS(ESI):m/z[M+H] + calcd for C 20 H 15 N 4 O 2 Cl:379.0956;found:379.0961.
Example 45
Synthesis of 1- (3- ((1H-pyrrolo [2,3-b ] pyridin-5-yl) oxy) phenyl) -3- (4-methyl-3-trifluoromethylphenyl) urea (Compound 45):
the synthesis procedure was as in example 43 substituting 4-methyl-3-trifluoromethylaniline for 3-trifluoromethylaniline. 1 H NMR(500MHz,DMSO-d 6 )δ11.74(s,1H),8.81(d,J=2.4Hz,2H),8.09(d,J=2.4Hz,1H),7.85(d,J=1.3Hz,1H),7.71(d,J=2.3Hz,1H),7.54(t,J=2.8Hz,1H),7.48(d,J=8.2Hz,1H),7.31(d,J=8.3Hz,1H),7.25(t,J=8.0Hz,1H),7.14(d,J=8.5Hz,2H),6.64–6.55(m,1H),6.45(dt,J=7.0,3.5Hz,1H),2.36(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 22 H 17 N 4 O 2 F 3 :427.1376;found:427.1377.
Example 46
Synthesis of 1- (3- ((1H-pyrrolo [2,3-b ] pyridin-5-yl) oxy) phenyl) -3- (4-chloro-3-trifluoromethylphenyl) urea (Compound 46):
the procedure was as in example 43, substituting 4-chloro-3-trifluoromethylaniline for 3-trifluoromethylaniline. 1 H NMR(500MHz,DMSO-d 6 )δ11.73(s,1H),9.05(s,1H),8.91(s,1H),8.07(d,J=2.6Hz,1H),8.01(d,J=2.2Hz,1H),7.70(d,J=2.5Hz,1H),7.60(dt,J=16.1,5.5Hz,2H),7.53(t,J=2.9Hz,1H),7.29–7.21(m,1H),7.12(dd,J=8.5,5.2Hz,2H),6.60(dd,J=8.1,1.8Hz,1H),6.44(dd,J=3.3,1.9Hz,1H).HRMS(ESI):m/z[M+H] + calcd for C 21 H 14 N 4 O 2 F 3 Cl:447.0830;found:447.0833.
Example 47
Synthesis of 5- (4-nitrophenoxy) -1H-pyrrolo [2,3-b ] pyridine (intermediate M9):
5-hydroxy-7-azaindole (2.5 g,17.73 mmol) and 4-fluoronitrobenzene (2.62 g,19.50 mmol) were dissolved in N, N-dimethylformamide (50 mL), and then potassium carbonate (4.9 g,35.46 mmol) was added and the reaction stirred at 80℃for 14h. After the completion of the reaction, ethyl acetate (300 mL) was added, followed by suction filtration, washing of the filtrate with saturated brine, drying over anhydrous sodium sulfate, and column chromatography to give intermediate M9.
Synthesis of 5- (4-aminophenoxy) -1H-pyrrolo [2,3-b ] pyridine (intermediate M10)
Intermediate M9 (2 g,7.84 mmol) and CH 3 OH (40 mL) was added to the flask, and then Raney nickel (300 mg) was added and the reaction stirred at 25℃under hydrogen balloon pressure for 10h. And after the reaction is finished, carrying out suction filtration, and carrying out column chromatography on the filtrate to obtain an intermediate M10.
Synthesis of 1- (4- ((1H-pyrrolo [2,3-b ] pyridin-5-yl) oxy) phenyl) -3- (3- (trifluoromethyl) phenyl) urea (Compound 47):
triphosgene (147 mg,0.50 mmol) was dissolved in tetrahydrofuran (15 mL) and then a solution of 3-trifluoromethylaniline (217 mg,1.35 mmol) and DIPEA (387 mg,3 mmol) in tetrahydrofuran (5 mL) was added under ice-bath. The reaction was stirred at 0deg.C for one hour, and intermediate M10 (281mg, 1.25 mmol) was added to the reaction, followed by stirring at 25deg.C for 5 hours. The reaction mixture was quenched with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate, and subjected to column chromatography to give compound 47. 1 H NMR(500MHz,DMSO-d 6 )δ11.70(s,1H),9.00(s,1H),8.75(s,1H),8.07(d,J=2.6Hz,1H),8.02(s,1H),7.63(d,J=2.5Hz,1H),7.59(d,J=8.4Hz,1H),7.53–7.49(m,2H),7.47–7.43(m,2H),7.30(d,J=7.7Hz,1H),6.99–6.91(m,2H),6.42(dd,J=3.3,1.9Hz,1H).HRMS(ESI):m/z[M+H] + calcd for C 21 H 15 N 4 O 2 F 3 :413.1220found:413.1220.
Example 48
Synthesis of 1- (4- ((1H-pyrrolo [2,3-b ] pyridin-5-yl) oxy) phenyl) -3- (3-chlorophenyl) urea (Compound 48):
the procedure is as in example 47, substituting 3-chloroaniline for 3-trifluoromethylaniline. 1 H NMR(400MHz,DMSO-d 6 )δ11.70(s,1H),8.89(s,1H),8.76(s,1H),8.06(d,J=2.6Hz,1H),7.67(dd,J=37.9,2.1Hz,2H),7.57–7.47(m,1H),7.45–7.37(m,2H),7.28(d,J=2.7Hz,2H),7.00(dt,J=6.6,2.2Hz,1H),6.96–6.88(m,2H),6.42(dd,J=3.4,1.9Hz,1H).HRMS(ESI):m/z[M+H] + calcd for C 20 H 15 N 4 O 2 Cl:379.0956;found:379.0958.
Example 49
Synthesis of 1- (4- ((1H-pyrrolo [2,3-b ] pyridin-5-yl) oxy) phenyl) -3- (4-chloro-3-trifluoromethylphenyl) urea (Compound 49):
the procedure was as in example 47, substituting 4-chloro-3-trifluoromethylaniline for 3-trifluoromethylaniline. 1 H NMR(400MHz,DMSO)δ11.72(s,1H),9.30(s,1H),8.94(s,1H),8.11(d,J=2.3Hz,1H),8.06(d,J=2.6Hz,1H),7.68–7.58(m,3H),7.56–7.48(m,1H),7.48–7.40(m,2H),6.99–6.88(m,2H),6.42(dd,J=3.3,1.9Hz,1H).HRMS(ESI):m/z[M+H] + calcd for C 21 H 14 N 4 O 2 F 3 Cl:447.0830;found:447.0829.
Example 50
Synthesis of 1- (4- ((1H-pyrrolo [2,3-b ] pyridin-5-yl) oxy) phenyl) -3- (4-methyl-3-trifluoromethylphenyl) urea (Compound 50):
the procedure was as in example 47, substituting 4-methyl-3-trifluoromethylaniline for 3-trifluoromethylaniline. 1 H NMR(500MHz,DMSO-d 6 )δ11.69(s,1H),8.85(s,1H),8.67(s,1H),8.06(d,J=2.5Hz,1H),7.93(d,J=1.6Hz,1H),7.62(d,J=2.4Hz,1H),7.51(dd,J=7.6,5.1Hz,2H),7.44(d,J=8.9Hz,2H),7.32(d,J=8.3Hz,1H),6.94(d,J=8.9Hz,2H),6.42(dd,J=3.1,1.9Hz,1H),2.37(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 22 H 17 N 4 O 2 F 3 :427.1376;found:427.1380.
Example 51
Synthesis of tert-butyl (4- ((6-aminopyridin-3-yl) oxy) phenyl) carbamate (intermediate M11):
intermediate M4 (2 g,7.84 mmol) and methanol (40 mL) were added to the flask, and Raney nickel (350 mg) was added and the reaction stirred at 25℃under hydrogen balloon pressure for 12h. Filtering after the reaction is finished, and obtaining an intermediate M11 through filtrate column chromatography.
Synthesis of tert-butyl (4- ((6-acetamidopyridin-3-yl) oxy) phenyl) carbamate (intermediate M12):
intermediate M11 (301 mg,1 mmol), 4-dimethylaminopyridine (5 mg) and triethylamine (120 mg,1.2 mmol) were dissolved in dichloromethane (20 mL), then acetyl chloride (87 mg,1.1 mmol) was added dropwise, the reaction mixture was stirred at 30℃for 2h, after the completion of the reaction, dichloromethane was added, and then column chromatography was performed by water washing to obtain intermediate M12.
Synthesis of N- (5- (4-aminophenoxy) pyridin-2-yl) acetamide (intermediate M13):
intermediate M12 (200 mg,0.58 mmol) was dissolved in dichloromethane (15 mL) and trifluoroacetic acid (200 mg,1.74 mmol) was added under ice-bath. The reaction solution was stirred at 30℃for 1 hour, after the completion of the reaction, concentrated directly, ethyl acetate and saturated sodium bicarbonate solution were added, and after sufficient stirring, the solution was separated, and the filtrate was dried over anhydrous sodium sulfate, followed by column chromatography to give intermediate M13.
Synthesis of N- (5- (4- (3- (3- (trifluoromethyl) phenyl) ureido) phenoxy) pyridin-2-yl) acetamide (Compound 51):
triphosgene (53 mg,0.18 mmol) was dissolved in tetrahydrofuran (15 mL) and then a solution of 3-trifluoromethylaniline (74 mg,0.45 mmol) and DIPEA (135 mg,1.04 mmol) in tetrahydrofuran (5 mL) was added at 0deg.C, and the reaction stirred at 0deg.C for 1h and intermediate M13 (106 mg,0.45 mmol) w was added. The mixture was stirred at 35℃for 5h. After the completion of the reaction, water and ethyl acetate were added thereto, followed by washing with saturated brine, drying and column chromatography to obtain Compound 51. 1 H NMR(400MHz,DMSO)δ10.53(s,1H),9.04(s,1H),8.82(s,1H),8.07(dd,J=17.9,14.7Hz,3H),7.58(d,J=8.5Hz,1H),7.54–7.44(m,4H),7.30(d,J=7.6Hz,1H),7.05–6.93(m,2H),2.09(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 21 H 17 N 4 O 3 F 3 :431.1326;found:431.1299.
Example 52
Synthesis of N- (5- (4- (3- (4-chloro-3- (trifluoromethyl) phenyl) ureido) phenoxy) pyridin-2-yl) (Compound 52):
the procedure was as in example 51, substituting 4-chloro-3-trifluoromethylaniline for 3-trifluoromethylaniline. 1 H NMR(500MHz,DMSO-d 6 )δ10.48(s,1H),9.25(s,1H),8.94(s,1H),8.09(dd,J=14.3,2.6Hz,3H),7.64(dd,J=8.8,2.1Hz,1H),7.60(d,J=8.8Hz,1H),7.50–7.43(m,3H),7.00(d,J=8.9Hz,2H),2.08(s,3H).HRMS(ESI):m/z[M+H] + calcd for C 21 H 16 N 4 O 3 F 3 Cl:465.0936;found:465.0940.
Example 53
Synthesis of 1- (4- ((6-amino-5-bromopyridin-3-yl) oxy) phenyl) -3- (3- (trifluoromethyl) phenyl) urea (Compound 53):
compound 29 (200 mg,0.52 mmol) was dissolved in N, N-dimethylformamide (15 mL), followed by the addition of NBS (116 mg,0.65 mmol). The reaction solution was stirred at 25℃for 1 hour, after the completion of the reaction, ethyl acetate and an aqueous solution of sodium thiosulfate were added, and after sufficient stirring, the mixture was separated, washed with water and dried, and column chromatography was performed to obtain Compound 53.
1 H NMR(400MHz,DMSO-d 6 )δ9.01(s,1H),8.77(s,1H),8.01(s,1H),7.83(d,J=2.5Hz,1H),7.63–7.54(m,2H),7.51(d,J=7.8Hz,1H),7.43(d,J=8.9Hz,2H),7.30(d,J=7.5Hz,1H),6.92(d,J=8.9Hz,2H),6.14(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 14 N 4 O 2 F 3 Br:467.0325;found:467.0324.
Example 54
Synthesis of tert-butyl (4- ((6-nitropyridin-3-yl) thio) phenyl) carbamate (intermediate M14):
the compound 2-nitro-5-bromopyridine (10 g,49.27 mmol) (4-mercaptophenyl) carbamic acid tert-butyl ester (10.3 g,49.27 mmol), cesium carbonate (20.9 g,64.05 mmol), N, N-dimethylformamide (100 mL) was reacted at 40℃for 20h. Adding water and ethyl acetate to stir after the reaction is finished, filtering with diatomite, and washing a filter cake with ethyl acetate; standing for separating, re-extracting the water phase with ethyl acetate, sequentially combining the organic phases, washing with saturated saline, drying with anhydrous sodium sulfate, and performing column chromatography to obtain a yellow solid with a yield of 10.19g and 62.5%.
Synthesis of (4- ((6-nitropyridin-3-yl) thio) aniline (intermediate M15)
Tert-butyl (10.19 g,30.78 mmol) carbamate (4- ((6-nitropyridin-3-yl) thio) of compound M4 (2 mmol) was added to dichloromethane (100 mL), stirred in an ice bath, trifluoroacetic acid (11.43 g,100 mmol) was added dropwise and the mixture was reacted at 25℃for 2h; directly concentrating to obtain yellow solid, adding ethyl acetate and sodium bicarbonate water solution, stirring, separating liquid, drying and concentrating to obtain brownish red solid 7.5g, and the yield is 81.5%.
Synthesis of 1- (4- ((6-nitropyridin-3-yl) thio) phenyl) -3-phenylurea (intermediate M16)
Triphosgene (100 mg,1.07 mmol) was first dissolved in tetrahydrofuran (10 mL) and the solution (compound aniline (100 mg,1.07 mmol) and N, N-diisopropylethylamine (276 mg,2.14 mmol) were dissolved in tetrahydrofuran (2 mL)) was added dropwise under ice-bath. Stirring for 40min in ice bath, adding compound M15 (4- ((6-nitropyridin-3-yl) thio) aniline (230 mg,1.00 mmol) for reaction at 30 ℃ for 5h, adding water and ethyl acetate after the reaction, stirring, standing for separating liquid, re-extracting the water phase with ethyl acetate, combining organic phases sequentially, washing with saturated common salt, drying with anhydrous sodium sulfate, and separating the product by column chromatography to obtain 245mg of yellow solid with the yield of 70%.
Synthesis of 1- (4- ((6-aminopyridin-3-yl) thio) phenyl) -3- (3- (trifluoromethyl) phenyl) urea (Compound 54):
to a saturated aqueous ammonium chloride solution (10 mL) of intermediate M16 (245 mg,0.70 mmol), methylene chloride (10 mL), methanol (10 mL) was added 150mg of zinc powder in an ice bath, and the mixture was reacted at 30℃for 1 hour. Adding water and dichloromethane after the reaction, stirring, suction filtering, standing for separating liquid, sequentially extracting the aqueous phase with dichloromethane, combining the organic phases, washing with saturated saline, drying with anhydrous sodium sulfate, and performing column chromatography to obtain a product, wherein 100mg of white solid is obtained. 1 H NMR(400MHz,DMSO)δ9.03(s,1H),8.84(s,1H),8.07–7.97(m,2H),7.50(ddd,J=14.4,11.1,5.5Hz,3H),7.44–7.36(m,2H),7.30(d,J=7.5Hz,1H),7.13–7.03(m,2H),6.49(d,J=8.6Hz,1H),6.36(s,2H).HRMS(ESI):m/z[M+H] + calcd for C 19 H 15 N 4 OSF 3 :405.1169;found:405.1110.
Example 55
In vitro CDK8 kinase inhibitory activity and in vitro antitumor activity evaluation:
compounds 1-54 were tested for CDK8 Kinase inhibitory activity by ADP-Glo Kinase Assay (Promega) using black 384 well plates. Will be aliveThe CDK8 kinase was diluted in a mixture (5 ng CDK8 kinase, 0.5. Mu.g substrate, 50. Mu.M DTT, 1. Mu.L buffer, dd H was added to each well) 2 O to 3. Mu.L) and then 1. Mu.L of 1. Mu.M compound solution (corresponding to 5-fold dilution in the system, 200nM final concentration) was added to each well, followed by 1. Mu.L of ATP (adenosine triphosphate) to give a final concentration of 50. Mu.M. After incubation for 1h at room temperature, ADP-Glo solution and kinase detection reagent were added and the data collected by a microplate reader.
Compounds 1-54 of the present invention all show a certain CDK8 kinase inhibitory activity at a concentration of 200nM, preferably compounds with a higher inhibition rate are tested in MTT assay to test the activity of these compounds on normal cells and colorectal cancer cells. Sorafenib was selected as a positive control and the results are shown in table 1.
MTT test: HCT-116, HT-29, SW-480, CT-26, GES-1 cells were seeded at 6000 cells/well in 96-well plates and at 37℃with 5% CO 2 Culturing for 24 hours in an incubator; the medium was discarded, and then 100. Mu.L of each concentration of compound solution ( concentrations 100, 20, 4, 0.8, 0.016. Mu.M) was added and cultured for 48 hours; MTT (5 mg/mL, 20. Mu.L) was added and incubated at 37℃for 4h; removing the culture medium, and adding 150 mu L of DMSO for dissolution; the absorbance at 492nm was measured by a microplate reader (PerkinElmer Envision), and the GI was calculated 50 Values.
TABLE 1
Figure BDA0003767738170000251
/>
Figure BDA0003767738170000261
As can be seen from table 1, the kinase activity and cellular activity of compound 29 was significantly better than sorafenib.
Example 56
Evaluation of antitumor Activity in vivo:
to evaluate the in vivo antitumor activity of compound 29, different doses (10 mg/kg and 40 mg/kg) of compound 29 were orally administered once daily to Balb/c mice of subcutaneous CT-26 tumor cell xenografts. Tumor volume and body weight were recorded every two days. As shown in fig. 1A-B, the tumor volume increase change was most pronounced in the control group, and significantly slowed in the treated group compared to the control group. Tumor Growth Inhibition (TGI) was used to evaluate the inhibition of tumor growth in vivo by compound 29, with the TGI increasing with increasing dose of compound 29 (fig. 1C). The results show that compound 29 effectively reduced the tumor volume in CT-26 tumor-bearing mice. H & E staining of tumor tissue showed significant changes after treatment, indicating the in vivo efficiency and specificity of compound 29 (FIG. 1D). To investigate the effect of compound 29 on CDK8 in vivo, the expression level of β -catenin in tumor samples was examined. IHC analysis showed that β -catenin was down-regulated following treatment with compound 29 (FIGS. 1E-F). To investigate the effect of compound 29 on molecular targets in vivo, the expression levels of β -catenin, E2F1, c-Myc associated with CDK8 in tumor samples were examined. Western blot analysis shows that after the compound 29 is treated, beta-catenin and c-Myc are down-regulated, E2F1 is up-regulated, CDK8 has no obvious change, and the WNT/beta-catenin signal channel related to CDK8 is involved in the in-vivo anti-tumor effect of the compound 29.
The foregoing has shown and described the basic principles and main features of the present invention and the advantages of the present invention. It will be understood by those skilled in the art that the present invention is not limited to the embodiments described above, and that the above embodiments and descriptions are merely illustrative of the principles of the present invention, and various changes and modifications may be made without departing from the spirit and scope of the invention, which is defined in the appended claims. The scope of the invention is defined by the appended claims and equivalents thereof.

Claims (6)

1. The aryl urea derivative is characterized in that the structural formula of the aryl urea compound is shown as follows:
Figure FDA0004235902070000011
Figure FDA0004235902070000021
Figure FDA0004235902070000031
Figure FDA0004235902070000041
2. the process for producing an arylurea derivative according to claim 1, wherein: the method comprises the following steps:
(1) Nucleophilic substitution reaction is carried out on 2-nitro-5-bromopyridine and (3-hydroxyphenyl) carbamic acid tert-butyl ester to obtain an intermediate M1;
(2) Removing protecting groups from the intermediate M1 to obtain an intermediate M2;
(3) Intermediate M2 reacts with an aromatic amine derivative to obtain intermediate M3;
(4) Intermediate M3 is subjected to reduction reaction to obtain compounds 1-19;
the reaction equation is as follows:
Figure FDA0004235902070000042
(5) The synthesis route conditions of the compounds 20-27,29-42 are the same as those of the compounds 1-19, except that the raw material (3-hydroxyphenyl) carbamic acid tert-butyl ester is replaced by (4-hydroxyphenyl) carbamic acid tert-butyl ester; wherein compound 29 is brominated to afford compound 53;
the reaction equation is as follows:
Figure FDA0004235902070000051
(6) Nucleophilic substitution reaction is carried out on the 5-hydroxy-7-azaindole and 3-fluoronitrobenzene to obtain an intermediate M7;
(7) Intermediate M7 undergoes a reduction reaction to obtain intermediate M8;
(8) Intermediate M8 reacts with an aromatic amine derivative to obtain a compound 43,45,46;
the reaction equation is as follows:
Figure FDA0004235902070000052
(10) The synthesis route of the compound 47-50 is the same as that of the compound 43,45,46, and the raw material 3-fluoronitrobenzene is changed into 4-fluoronitrobenzene; the reaction equation is as follows:
Figure FDA0004235902070000053
(11) Intermediate M4 is reduced to give intermediate M11;
(12) Intermediate M11 is acetylated to afford intermediate M12;
(13) Removing Boc from the intermediate M12 under trifluoroacetic acid to obtain an intermediate M13;
(14) Intermediate M13 reacts with aromatic amine derivative to obtain compounds 51-52;
the reaction equation is as follows:
Figure FDA0004235902070000061
(15) Compound 54 is synthesized by the same route as compound 1, except that the raw material (3-hydroxyphenyl) carbamic acid tert-butyl ester is converted into (4-mercaptophenyl) carbamic acid tert-butyl ester;
the reaction equation is as follows:
Figure FDA0004235902070000062
3. a pharmaceutical composition comprising the aryl urea derivative of claim 1 or a pharmaceutically acceptable salt thereof.
4. A pharmaceutical preparation comprising an active ingredient and a pharmaceutically acceptable adjuvant, wherein the active ingredient comprises the aryl urea derivative or a pharmaceutically acceptable salt thereof according to claim 1.
5. Use of an aryl urea derivative according to claim 1 or a pharmaceutically acceptable salt thereof in the preparation of a CDK8 inhibitor.
6. The use of an aryl urea derivative or a pharmaceutically acceptable salt thereof as claimed in claim 1 in the preparation of an antitumor drug; the tumor is colorectal cancer.
CN202210891463.6A 2022-07-27 2022-07-27 Aryl urea derivative and preparation method and application thereof Active CN115197130B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210891463.6A CN115197130B (en) 2022-07-27 2022-07-27 Aryl urea derivative and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210891463.6A CN115197130B (en) 2022-07-27 2022-07-27 Aryl urea derivative and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN115197130A CN115197130A (en) 2022-10-18
CN115197130B true CN115197130B (en) 2023-06-27

Family

ID=83584998

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210891463.6A Active CN115197130B (en) 2022-07-27 2022-07-27 Aryl urea derivative and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN115197130B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115141197A (en) * 2022-07-27 2022-10-04 安徽医科大学 3-aromatic heterocyclic substituted phenyl derivative and preparation method and application thereof
CN115991705A (en) * 2022-12-26 2023-04-21 安徽医科大学 3- (1H pyrrolo [2,3-b ] pyridin-5-yl) benzoyl derivatives, preparation and application thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5072595B2 (en) * 2005-08-05 2012-11-14 中外製薬株式会社 Multikinase inhibitor
HUE058998T2 (en) * 2018-03-15 2022-09-28 Fujian Haixi Pharmaceuticals Co Ltd Heteroaryl compounds as kinase inhibitor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115141197A (en) * 2022-07-27 2022-10-04 安徽医科大学 3-aromatic heterocyclic substituted phenyl derivative and preparation method and application thereof
CN115991705A (en) * 2022-12-26 2023-04-21 安徽医科大学 3- (1H pyrrolo [2,3-b ] pyridin-5-yl) benzoyl derivatives, preparation and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
"Design and Development of a Series of Potent and Selective Type II Inhibitors of CDK8";Philippe Bergeron et al.;《ACS Med. Chem. Lett.》;第7卷;第595-600页 *

Also Published As

Publication number Publication date
CN115197130A (en) 2022-10-18

Similar Documents

Publication Publication Date Title
EP2949647A1 (en) Deuterated phenyl amino pyrimidine compound and pharmaceutical composition containing same
US10689361B2 (en) Quinoline derivative and use thereof
EA018539B1 (en) Phenylamino isonicotinamide compounds
RO120541B1 (en) Cyanoguanidine derivatives as cell proliferation inhibitors
WO2011153197A1 (en) Hydroxypyridone derivatives, pharmaceutical compositions thereof, and their therapeutic use for treating proliferative diseases
RU2195452C2 (en) Cyanoguanidines, methods of their synthesis and pharmaceutical preparation based on thereof
CN104230952A (en) Compound containing pyrimidine skeleton, and preparation method and use of compound
EP3403651B1 (en) Isocorydine derivatives, preparation method and use thereof
EP3617198B1 (en) Guanidine derivative
CN115197130B (en) Aryl urea derivative and preparation method and application thereof
CN105622507B (en) A kind of naphthalimide derivative and its preparation method and application
CN111943906B (en) Amidine derivatives, preparation method, pharmaceutical composition and application thereof
CN112979659B (en) Preparation and application of HIF-2 alpha small molecule inhibitor
CN107513089B (en) Novel cytidine derivative dimer and application thereof
CN113444074B (en) Compound with EGFR (epidermal growth factor receptor) and Wnt dual inhibition effects as well as preparation method and application thereof
CN114685382A (en) Quinazoline-4-amine derivative with HDACs (HDACs) inhibitory activity and preparation method and application thereof
EP3133069B1 (en) Bcr-abl diploid inhibitor, preparation method therefor, and uses thereof
CN110183503B (en) Sulfonyl azaspirodecadienone compounds and application thereof
CN106892859A (en) (H) ketone polyamines conjugate of benzo [c, d] indoles 2 and its preparation method and application
CN113754587A (en) Phenylpyrazole compound and application thereof
CN111533732A (en) N-heterocyclic condensed tryptamine-beta-lactam derivative and preparation method and application thereof
CN102485735B (en) 6-fructosamine-4-arylamidoquinazoline derivative and purpose thereof
CN107235994B (en) Simultaneously [3,2-a] pyrimidine -3- acetamide derivative and the application of 5,7- diphenyl -5H- thiazole
CN115073361B (en) Rosin diterpenoid compound and preparation method and application thereof
CN109293588A (en) A kind of small molecule compound and the preparation method and application thereof with the bis- target spots of IDO1/TDO

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