CN116178248A - Naphthyl urea-piperidine compounds, and preparation method and application thereof - Google Patents

Naphthyl urea-piperidine compounds, and preparation method and application thereof Download PDF

Info

Publication number
CN116178248A
CN116178248A CN202310176580.9A CN202310176580A CN116178248A CN 116178248 A CN116178248 A CN 116178248A CN 202310176580 A CN202310176580 A CN 202310176580A CN 116178248 A CN116178248 A CN 116178248A
Authority
CN
China
Prior art keywords
acid
compound
cancer
cancer cells
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202310176580.9A
Other languages
Chinese (zh)
Inventor
徐学军
杨玉坡
郭伟凯
杨争艳
刘亚青
段超群
徐红运
李岑
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.)
Henan Radiomedical Science And Technology Co ltd
Original Assignee
Henan Radiomedical Science And Technology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Henan Radiomedical Science And Technology Co ltd filed Critical Henan Radiomedical Science And Technology Co ltd
Priority to CN202310176580.9A priority Critical patent/CN116178248A/en
Publication of CN116178248A publication Critical patent/CN116178248A/en
Pending legal-status Critical Current

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
    • C07D213/24Heterocyclic 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 substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/36Radicals substituted by singly-bound nitrogen atoms
    • C07D213/40Acylated substituent nitrogen atom
    • 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
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Landscapes

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

Abstract

The invention discloses a naphthyl urea-piperidine compound, a preparation method and application thereof, and the structural formula is shown as follows:
Figure ZY_1
or (b)
Figure ZY_2
Wherein R is 1 Selected from the group consisting of
Figure ZY_3
Figure ZY_4
Figure ZY_5
Figure ZY_6

Description

Naphthyl urea-piperidine compounds, and preparation method and application thereof
Technical Field
The invention belongs to the field of tumor targeted therapy, and particularly relates to a naphtyl urea-piperidine compound, a preparation method and application thereof.
Background
There have been extensive studies demonstrating that abnormal activation of cell transduction and transcriptional activator 3 (Signal transducer and activators of transcriptions 3, stat 3) plays an important role in the development and progression of tumors and immune disorders. Overexpression and constitutive activation of STAT3 is common in a variety of solid tumors and hematological tumors. STAT3 is a substrate protein of the non-receptor tyrosine protein kinase JAK2 (Janus kinase 2). Under normal conditions, STAT3 exists in the cytoplasm as an inactive monomer, and has a strict negative feedback regulatory mechanism, involved in many important biological processes such as proliferation, differentiation, apoptosis and immune regulation of cells. When the negative feedback regulation mechanism of JAK2 or STAT3 is abnormal or the gene mutation, the phosphorylation level of STAT3 is continuously increased and endogenous is enhanced, homodimer or heterodimer is formed with the SH2 domain of another STAT3 protein to enter the nucleus, and the protein expression and transcription of downstream target genes are started through the binding of a DNA binding domain to a specific gene promoter sequence, wherein the protein expression comprises BCL-2 family (such as BCL-2 and BCL-XL) related to mitochondrial apoptosis, cyclin D1 (Cyclin D1) related to cell cycle regulation and the like.
Cyclin D1 is a regulator of Cyclin-dependent kinase CDKs, and has the main functions of promoting cell cycle progression, promoting cell proliferation, combining histone deacetylase P/CAF or transcription factor TF II D and the like, and promoting target gene transcription and overexpression to cause uncontrolled cell proliferation and malignant transformation. In animal tumor models or in vitro cultured tumor cells, inhibiting STAT3 protein can effectively inhibit growth of tumor cells or induce apoptosis of tumor cells, and reduce metastasis of tumor cells. STAT3 has become a popular target for tumor therapy.
The invention aims to disclose the antitumor effect and the potential pharmacological mechanism of a novel naphthyridine compound and derivatives thereof, and the potential application of the compound in clinical treatment of breast cancer, liver cancer, lung cancer, oxatinib-resistant lung cancer, colon cancer, leukemia, gastric cancer, pancreatic cancer, prostate cancer and esophageal cancer.
Disclosure of Invention
The invention aims to provide a naphthalene urea-piperidine compound, and a preparation method and application thereof.
Based on the above purpose, the invention adopts the following technical scheme:
the structural formula of the naphtyl urea-piperidine compounds is shown as a general formula I:
Figure BDA0004101053920000021
wherein R is 1 Selected from the group consisting of
Figure BDA0004101053920000022
Specifically, the compound has the following structure:
Figure BDA0004101053920000023
the preparation process of the compound IY220816B-1 is as follows:
Figure BDA0004101053920000031
the preparation method comprises the following specific steps:
(1) Compound 1A, boc 2 O and Pd/C are dissolved in methanol, and the reaction is completed under stirring of hydrogen; the reaction solution was filtered through celite, rinsed with ethyl acetate, and subjected to organic phase spin-dry column chromatography to give intermediate 2A, compound 1A, boc 2 The mol ratio of O to Pd/C is 1:1:0.1;
(2) Compound 2A, hbtu and DIEA were dissolved in DMF and stirred at room temperature to complete the reaction; diluting the reaction solution with ethyl acetate, washing with saturated saline, and performing organic phase drying spin-drying column chromatography to obtain a compound 3A; the molar ratio of compound 2A, HBTU and DIEA is 1:1:1.2:1.2;
(3) Dissolving the compound 3A in methanol, adding an ethyl acetate solution of hydrogen chloride, and stirring at room temperature to react completely; directly spin-drying the reaction solution; the molar ratio of the compound 3A to the hydrogen chloride is 1.29:40;
(4) Dissolving a compound 4A and a compound 5 in tetrahydrofuran, and stirring at 110-130 ℃ to react completely; and (3) directly spin-drying the reaction solution for column chromatography to obtain the IY220816B-1, wherein the molar ratio of the compound 4A to the compound 5 is 1:1.
The synthesis of compound ID220825A-1 was as follows:
Figure BDA0004101053920000032
the preparation method comprises the following specific steps:
(1) Dissolving the compound 1a and potassium tert-butoxide in tetrahydrofuran, stirring for 20-40 min, adding the compound 1B, and continuing stirring at room temperature to react completely; directly spin-drying the reaction solution for column chromatography to obtain a compound 2B, wherein the molar ratio of the compound 1B to the compound 1a to the potassium tert-butoxide is 1:1.2:1.2;
(2) Dissolving the compound 2B in ethanol, heating to 45-55 ℃, adding iron powder at 45-55 ℃ and continuously stirring for complete reaction; filtering the reaction solution by using diatomite, and directly spin-drying the filtrate by column chromatography to obtain a compound 3B, wherein the molar ratio of the compound 2B to the iron powder is 1:5;
(3) Dissolving a compound 3B and a compound 5 in tetrahydrofuran, and stirring at 110-130 ℃ to react completely; and (3) directly spin-drying the reaction solution for column chromatography to obtain ID220825A-1, wherein the molar ratio of the compound 3B to the compound 5 is 1:1.
The preparation process of the compound IY220825A-1 is as follows:
Figure BDA0004101053920000041
(1) Dissolving a compound 1C and a compound 5 in tetrahydrofuran, and stirring at 110-130 ℃ to react completely; directly spin-drying the reaction solution for column chromatography to obtain a compound 2C, wherein the molar ratio of the compound 1C to the compound 5 is 1:1;
(2) Compound 2C, compound 2b, K 2 CO 3 And Pd (dppf) Cl 2 Dissolving in a mixture of 1, 4-dioxane and water, and stirring at 100-110 ℃ to react completely; directly spin-drying the reaction solution to obtain IY220825A-1, compound 2C, compound 2b and K 2 CO 3 And Pd (dppf) Cl 2 The molar ratio of 1:1.2:1.2:0.1, the volume ratio of 1, 4-dioxane to water was 10:1. The preparation process of the compound IY221116B-1 is as follows:
Figure BDA0004101053920000051
(1) Dissolving compound 1D, compound 1b, HBTU and DIEA in DMF, stirring at room temperature for reacting for a period of time (0.5-1.5 h), and then reacting at 110-130 ℃ completely; directly spin-drying the reaction solution for column chromatography to obtain a compound 2D, wherein the molar ratio of the compound 1D to the compound 1b to the HBTU to the DIEA is 1:1:1.2:1.2;
(2) Compound 2D, compound 2c, BINAP, pd (OAc) 2 And Cs 2 CO 3 Dissolving in toluene under the protection of nitrogen at 110-120 DEG CStirring and reacting completely; the reaction solution was directly subjected to spin-drying column chromatography to give intermediate 3D, compound 2c, BINAP, pd (OAc) 2 And Cs 2 CO 3 The molar ratio of (2) is 1:1:0.1:0.05:1.2;
(3) Dissolving the compound 3D in dichloromethane, dropwise adding trifluoroacetic acid at the temperature of 0 ℃, and stirring to react completely; directly spin-drying the reaction solution, diluting with ethyl acetate, adjusting the pH to 8 with saturated sodium bicarbonate aqueous solution, and performing organic phase drying spin-drying column chromatography to obtain a compound 4D;
the molar volume ratio of compound 3D to trifluoroacetic acid was 0.88mmol:5mL;
(4) Dissolving a compound 4D and a compound 5 in tetrahydrofuran, and stirring at 110-130 ℃ to react completely; and (3) directly spin-drying the reaction solution for column chromatography to obtain the IY221116B-1, wherein the molar ratio of the compound 4D to the compound 5 is 1:1.
The preparation process of the compound ID221028A-1 is as follows:
Figure BDA0004101053920000061
(1) Dissolving a compound 1C and a compound 5 in tetrahydrofuran, and stirring at 110-130 ℃ to react completely; directly spin-drying the reaction solution for column chromatography to obtain a compound 2C, wherein the molar ratio of the compound 1C to the compound 5 is 1:1;
(2) Compound 2C, compound 2d, pd (PPh 3 )Cl 2 Dissolving CuI and triethylamine in DMF, heating to 45-55 ℃ and stirring to react completely; the reaction solution was directly spin-dried to give ID221028A-1, compound 2C, compound 2d, pd (PPh) 3 )Cl 2 The molar ratio of CuI to triethylamine was 1:1:0.1:0.1:4.
The preparation process of the compound IY220831B-1 is as follows:
Figure BDA0004101053920000062
/>
(1) Dissolving a compound 2A, a compound 1c and CDI in tetrahydrofuran, and stirring at 70-75 ℃ to react completely; directly spin-drying and mixing the reaction solution, and passing through a column to obtain a compound 2F, wherein the molar ratio of the compound 2A to the compound 1c to the CDI is 1:1:1;
(2) Dissolving compound 2F in methanol, and adding ethyl acetate solution of hydrogen chloride; stirring at room temperature to react completely; directly spinning the reaction solution to obtain a compound 3F, wherein the molar ratio of the compound 2F to hydrogen chloride is 1:40;
(3) Dissolving a compound 3F and a compound 5 in tetrahydrofuran, and stirring at 110-130 ℃ to react completely; and (3) directly spin-drying the reaction solution for column chromatography to obtain the IY220831B-1, wherein the molar ratio of the compound 3F to the compound 5 is 1:1.
A biologically acceptable salt of the above described naphthylurea-piperidines with at least one of acetic acid, dihydrofolic acid, benzoic acid, citric acid, sorbic acid, propionic acid, oxalic acid, fumaric acid, maleic acid, hydrochloric acid, malic acid, phosphoric acid, sulfurous acid, sulfuric acid, vanillic acid, tartaric acid, ascorbic acid, boric acid, lactic acid and ethylenediamine tetraacetic acid.
The application of the naphthyridine urea-piperidine compounds and the biologically acceptable salts thereof in preparing anti-tumor drugs, wherein the anti-tumor drugs are drugs for treating tumors related to STAT3 signaling.
Preferably, the antitumor drug is a drug for treating breast cancer, liver cancer, lung cancer, drug-resistant lung cancer, colon cancer, leukemia, gastric cancer, pancreatic cancer, prostate cancer and esophageal cancer.
It is another object of the present invention to provide a class of small molecule compounds with targeted antitumor activity.
The tumor may be specifically a STAT3 high-expression or constitutively activated tumor including, but not limited to, breast cancer, liver cancer, lung cancer, drug-resistant lung cancer, colon cancer, leukemia, stomach cancer, pancreatic cancer, prostate cancer, esophageal cancer, and the like.
Specifically, the invention synthesizes a class of naphthylurea-piperidine compounds IY220816B-1, ID 2208235A-1, IY221116B-1, ID221028A-1 and IY220831B-1 with brand new structures. The proliferation inhibition effect of the compounds on tumor cells is detected by an MTT method, the influence of the compounds on the cell cycle and apoptosis of the tumor cells is detected by a flow cytometry method, and the inhibition effect of the compounds on JAK2/STAT3 signals is clarified by immunoblotting and other methods.
The results show that the compounds IY220816B-1, ID 2208235A-1, IY221116B-1, ID221028A-1 and IY220831B-1 can effectively inhibit proliferation of cells such as breast cancer, liver cancer, lung cancer, drug-resistant lung cancer, colon cancer, leukemia, gastric cancer, pancreatic cancer, prostate cancer, esophageal cancer and the like, and inhibit activation of STAT3 signal channels.
The tumor cells are breast cancer cells MDA-MB-468, liver cancer cells HepG2, HCC97H, lung cancer cells PC9, octreotide-resistant lung cancer cells PC9-AR, colon cancer cells HT-29, leukemia cells Jurkat, gastric cancer cells HGC27, pancreatic cancer cells BxPC-3, prostate cancer cells Lncap and esophageal cancer cells KYSE450.
In summary, the present invention provides a novel naphthylurea-piperidine compound and its derivatives for use in tumor therapy and potential molecular mechanisms.
Drawings
FIG. 1 shows the results of detection of half-number inhibition (IC 50 value) of breast cancer cells MDA-MB-468, liver cancer cells HepG2, lung cancer cells PC9, octenib-resistant lung cancer cells PC9-AR, colon cancer cells HT29, leukemia cells Jurkat, gastric cancer cells HGC27, pancreatic cancer cells BxPC-3, prostate cancer cells Lncap and esophageal cancer cells KYSE450, etc. by IY220816B-1, ID 2208235A-1, IY 2205A-1, IY221116B-1, ID221028A-1, IY220831B-1, etc.;
FIG. 2 shows the inhibition of STAT3, p-STAT3 (Tyr 705) and Cyclin D1 protein expression in MDA-MB-468 cells by different concentrations of IY 220816B-1.
Detailed Description
In order to make the technical purpose, technical scheme and beneficial effect of the present invention more clear, the technical scheme of the present invention is further described below with reference to the accompanying drawings and specific embodiments.
In the process according to the invention for the synthesis of the compounds of the formula I, the various starting materials used for the reaction are preparable by the person skilled in the art according to the prior art, or can be prepared by methods known from the literature, or can be obtained commercially. The intermediates, raw materials, reagents, reaction conditions and the like used in the above reaction schemes may be appropriately changed according to the knowledge already known to those skilled in the art.
In the present invention, unless otherwise specified, wherein: (i) The temperature is expressed in degrees centigrade (DEG C), and the operation is performed in a room temperature environment; more specifically, the room temperature is 20-30 ℃; (ii) Drying the organic solvent by a common drying method, evaporating the solvent by a rotary evaporator under reduced pressure, wherein the bath temperature is not higher than 50 ℃; the volume ratio of the developing agent to the eluent is equal; (iii) the reaction process is followed by Thin Layer Chromatography (TLC); (iv) The final product has satisfactory proton nuclear magnetic resonance 1 H-NMR)。
EXAMPLE 1 Synthesis of IY220816B-1 Compound
Figure BDA0004101053920000081
The compound IY220816B-1 is named 4- (2- (piperidin-1-yl) methoxy) -N- (4- (3- (pyridin-4-ylmethyl) ureido) naphthalen-1-yl) benzamide,
the synthetic route is as follows:
Figure BDA0004101053920000082
tert-butyl (4-aminophthalen-1-yl) carbamate (intermediate 2A)
Raw material 1A (1 g,5.31mmol,1.0 eq), boc 2 O (1.16 g,5.31mmol,1.0 eq) and Pd/C (100 mg,0.1 eq) were dissolved in 150ml methanol and reacted under hydrogen for 12 hours with stirring; TLC (PE/ea=2/1, r f Product=0.25) shows that the starting material has reacted, a new point has been created; the reaction solution was filtered through celite, rinsed with ethyl acetate, and the organic phase was spin-dry stirred through the column and rinsed with (PE/ea=10/1 to 1/1) to give intermediate 2A (1.05 g, 76.6%) as a yellow solid.
Step 2.Tert-butyl (4- (4- (2- (pin-1-yl) methoxy) benzamido) naphthalen-1-yl) carbamate (intermediate 3A)
Raw material 2A (400 mg,1.55mmol,1.0 eq) as suchMaterial 2a (383 mg,1.55mmol,1.0 eq), HBTU (704 mg,1.86mmol,1.2 eq) and DIEA (240 mg,1.86mmol,1.2 eq) were dissolved in 10ml DMF and stirred at room temperature for 12 hours; TLC (DCM/MeOH=20/1, R) f Product=0.35) shows that the starting material has reacted, a new point has been created; the reaction was diluted with 100ml ethyl acetate and washed 3 times with saturated brine (100 ml x 3), the organic phase was dried, spin-dried and column-passed, and rinsed with (DCM/meoh=100/1-20/1) to give intermediate 3A (580 mg, 76.5%) as a yellow solid.
Step 3N- (4-phosphoraphtalen-1-yl) -4- (2- (piprolin-1-yl) ethoxy) benzamide (intermediate 4A)
Raw material 3A (630 mg,1.29mmol,1.0 eq) was dissolved in 20ml of methanol, and 10ml of 4mol/L ethyl acetate solution of hydrogen chloride was added thereto, followed by stirring at room temperature for reaction for 2 hours; TLC (DCM/MeOH=20/1, R) f Product=0) shows that the raw material reaction is completed and a new point is generated; the reaction solution was directly dried by spin to give 4A (600 mg, 97%) as a white solid.
Step 4.4- (2- (pireidin-1-yl) method) -N- (4- (3- (pyridin-4-ylmethyl) ureido) naphthalen-1-yl) benzamide (IY 220816B-1)
Raw material 4A (100 mg,0.22mmol,1.0 eq) and raw material 5 (44 mg,0.22mmol,1.0 eq) were dissolved in 20ml tetrahydrofuran and reacted for 10 hours with stirring at 120 ℃. TLC (DCM/MeOH=10/1, R) f Product=0.2) shows that the raw material reaction is completed and a new point is generated; the reaction was directly spin-dried through the column and rinsed with (DCM/meoh=50/1 to 10/1) to give IY220816B-1 (55 mg, 48.7%) as a yellow solid.
1 H NMR(CDCl 3 ,300MHz)δ:10.16(s,1H),8.82(s,1H),8.54(d,J=6.0Hz,2H),8.07(d,J=9.0Hz,1H),7.95(d,J=9.0Hz,2H),7.60(d,J=9.0Hz,2H),7.55-7.47(m,2H),7.37(d,J=6.0Hz,2H),7.10(d,J=6.0Hz,2H),4.43(d,J=6.0Hz,2H),4.18(t,J=6.0Hz,2H),2.70(t,J=6.0Hz,2H),2.51(t,J=6.0Hz,2H),1.52(t,J=6.0Hz,2H),1.25(m,2H).
EXAMPLE 2 Synthesis of ID2208235A-1 Compound
Figure BDA0004101053920000101
The compound ID220825A-1, named 1- (4- (2- (1-yl) ethanoxy) phenyl) ethanoxy) naphthalen-1-yl) -3- (pyridin-4-ylmethyl) urea, was synthesized as follows:
Figure BDA0004101053920000102
step 1.1- (2- (4- (1- ((4-nitrothiophen-1-yl) oxy) ethyl) phenyl) ethyl) piperidine (intermediate 2B)
Raw material 1a (1.57 g,6.28mmol,1.2 eq) and potassium tert-butoxide (704 mg,6.28mmol,1.2 eq) were dissolved in 20ml tetrahydrofuran, after stirring for 30 minutes raw material 1B (1.0 g,5.23mmol,1.0 eq) was added and the reaction was continued at room temperature for 30 minutes; TLC (DCM/MeOH=20/1, R) f Product=0.45) shows that the starting material has reacted, a new point has been created; the reaction was directly spun-dried over the column and rinsed with DCM/meoh=50/1 to 10/1 to give intermediate 2B (1.7 g, 77.3%) as a yellow oily liquid.
Step 2.4- (1- (4- (2- (1-yl) ethoxy) phenyl) ethoxy) naphthalen-1-amine (intermediate 3B)
Raw material 2B (1 g,2.38mmol,1.0 eq) was dissolved in ethanol, warmed to 50℃and iron powder (664 mg,11.89mmol,5.0 eq) was added and the reaction stirred for 30 minutes at 50℃further; TLC (DCM/MeOH=20/1, R) f Product=0.25) shows that the starting material has reacted, a new point has been created; the reaction was filtered through celite and the filtrate was directly spin-dried through the column and rinsed with DCM/meoh=50/1-10/1 to give intermediate 3B (750 mg, 80.8%) as a reddish brown oil.
Step 3.1- (4- (1- (4- (2- (pi-idin-1-yl) ethoxy) phenyl)
ethoxy)naphthalen-1-yl)-3-(pyridin-4-ylmethyl)urea(ID220825A-1)
Raw material 3B (200 mg,0.51mmol,1.0 eq) and raw material 5 (103 mg,0.51mmol,1.0 eq) were dissolved in 20ml tetrahydrofuran and reacted at 120℃for 10 hours with stirring; TLC (DCM/MeOH=10/1, R) f Product=0.2) shows that the raw material reaction is completed and a new point is generated; the reaction mixture was directly spin-dried through the column and rinsed with (DCM/meoh=50/1 to 10/1) to give ID220825a-1 (120 mg, 44.7%) as whiteA solid.
1 H NMR(CDCl 3 ,300MHz)δ:8.0(d,J=9.0Hz,2H),6.93(d,J=9.0Hz,2H),4.17(t,J=6.0Hz,2H),3.90(q,1H),2.82(t,J=6.0Hz,2H),2.58-2.55(m,4H),1.66-1.61(m,4H),1.50(t,J=3.0Hz,2H),1.25(d,J=6.0Hz,3H).
EXAMPLE 3 Synthesis of IY 2208235A-1 Compound
Figure BDA0004101053920000111
The compound IY220825A-1, named 1- (4- (2- (piperidin-1-yl) method) phenyl) naphthalen-1-yl) -3- (pyridin-4-ylmethyl) urea, was synthesized as follows:
Figure BDA0004101053920000112
step 1.1- (4-bromoshape-1-yl) -3- (pyridin-4-ylmethyl) urea (intermediate 2C)
Raw material 1C (1 g,4.5mmol,1.0 eq) and raw material 5 (910 mg,4.5mmol,1.0 eq) were dissolved in 100ml tetrahydrofuran and reacted at 120℃for 10 hours with stirring; TLC (DCM/MeOH=10/1, R) f Product=0.6) shows that the raw material reaction is completed and a new point is generated; the reaction was directly spin-dried through the column and rinsed with (DCM/meoh=50/1 to 30/1) to give intermediate 2C (810 mg, 50.6%) as a white solid.
Step 2.1- (4- (4- (2- (1-yl) ethanoxy) phenyl) naphthalen-1-yl) -3- (pyridin-4-ylmethyl) urea (IY 220825A-1)
Starting material 2C (200 mg,0.56mmol,1.0 eq), starting material 2b (186 mg,0.56mmol,1.2 eq), K 2 CO 3 (93 mg,0.67mmol,1.2 eq) and Pd (dppf) Cl 2 (12 mg,0.056mmol,0.1 eq) in 1, 4-dioxane (20 ml) and water (2 ml) at 100℃for 3 hours with stirring; TLC (DCM/MeOH=10/1, R) f Product=0.2) shows that the raw material reaction is completed and a new point is generated; the reaction was directly spin-dried through the column and rinsed with (DCM/meoh=50/1 to 10/1) to give IY220825a-1 (220 mg, 81.5%) as a white solid.
1 H NMR(CDCl 3 ,300MHz)δ:8.0(d,J=9.0Hz,2H),6.93(d,J=9.0Hz,2H),4.17(t,J=6.0Hz,2H),2.82(t,J=6.0Hz,2H),2.58-2.55(m,4H),1.66-1.61(m,4H),1.50(t,J=3.0Hz,2H).
EXAMPLE 4 Synthesis of IY221116B-1 Compound
Figure BDA0004101053920000121
The compound IY221116B-1 is named 1- (4- (3- (4- (2- (pixel-1-yl) ethoxy) phenyl) -1,2, 4-oxazo-5-yl) naphthalen-1-yl) -3- (pyridin-4-ylmethyl) urea,
the synthetic route is as follows:
Figure BDA0004101053920000131
step 1.5- (4-bromoshape-1-yl) -3- (4- (2- (piperidine-1-yl) ethoxy) phenyl) -1,2, 4-oxoadiazole (intermediate 2D)
Raw material 1D (1 g,3.98mmol,1.0 eq), raw material 1b (1.05 g,3.98mmol,1.0 eq), HBTU (1.8 g,4.8mmol,1.2 eq) and DIEA (618 mg,4.8mmol,1.2 eq) were dissolved in 100ml DMF and reacted for 1 hour at room temperature with stirring, then for 10 hours at 120 ℃. TLC (DCM/meoh=10/1, rf/product=0.35) showed that the starting material was reacted completely, a new spot was generated; the reaction was directly spin-dried through the column and rinsed with (DCM/meoh=50/1 to 10/1) to give intermediate 2D (1.7 g, 89.1%) as an off-white solid.
Step 2N- (2, 4-Dimethoxybenzyl) -4- (3- (4- (2- (pi-idin-1-yl) ethoxy)
phenyl) -1,2, 4-oxazol-5-yl-naphthalen-1-amine (intermediate 3D)
Raw material 2D (1 g,2.09mmol,1.0 eq), raw material 2c (349 mg,2.09mmol,1.0 eq), BINAP (130 mg,0.209mmol,0.1 eq), pd (OAc) 2 (24 mg,0.104mmol,0.05 eq) and Cs 2 CO 3 (817 mg,2.51mmol,1.2 eq) in 100ml toluene under nitrogen, stirring at 110℃for 12 hours; TLC (DCM/MeOH=10/1, R) f Product=0.25) shows that the starting material has reacted, new pointGenerating; the reaction was directly spin-dried through the column and rinsed with (DCM/meoh=50/1 to 10/1) to give intermediate 3D (650 mg, 55.1%) as a white solid.
Step 3.4- (3- (4- (2-yl) ethoxy) phenyl) -1,2, 4-oxazo-5-yl) naphthalen-1-amine (intermediate 4D)
Raw material 3D (500 mg,0.88mmol,1.0 eq) was dissolved in 20ml dichloromethane, 5ml trifluoroacetic acid (67 mmol) was added dropwise at 0℃and the reaction was stirred at room temperature for 1 hour; TLC (DCM/MeOH=10/1, R) f Product=0.25) shows that the raw material reaction is completed and a new point is generated; the reaction was dried directly by spin-drying, diluted with ethyl acetate (150 ml), ph=8 was adjusted with saturated aqueous sodium bicarbonate, the organic phase was dried by spin-drying and applied to the column, and rinsed with (DCM/meoh=50/1-10/1) to give intermediate 4D (350 mg, 95.4%) as a yellow solid.
Step 4.1- (4- (3- (4- (2- (pi-idin-1-yl) ethoxy) phenyl) -1,2, 4-oxoadiazol-5-yl)
naphthalen-1-yl)-3-(pyridin-4-ylmethyl)urea(IY221116B-1)
Raw material 4D (200 mg,0.48mmol,1.0 eq) and raw material 5 (97 mg,0.48mmol,1.0 eq) were dissolved in 20ml tetrahydrofuran and reacted at 120℃for 10 hours with stirring; TLC (DCM/MeOH=10/1, R) f Product=0.1) shows that the raw material reaction is completed and a new point is generated; the reaction was directly spin-dried through the column and rinsed with (DCM/meoh=50/1 to 10/1) to give IY221116B-1 (102 mg, 39.2%) as a yellow solid.
1 H NMR(CDCl 3 ,300MHz)δ:8.02(d,J=9.0Hz,2H),6.90(d,J=9.0Hz,2H),4.12(t,J=6.0Hz,2H),2.82(t,J=6.0Hz,2H),2.58-2.53(m,4H),1.66-1.60(m,4H),1.51(t,J=3.0Hz,2H).
EXAMPLE 5 Synthesis of ID221028A-1 Compound
Figure BDA0004101053920000141
The compound ID221028A-1 is named 1- (4- ((4- (2- (1-yl) method) phenyl) ethyl) naphthalen-1-yl) -3- (pyridin-4-ylmethyl) urea, and the synthetic route is as follows:
Figure BDA0004101053920000151
step 1.1- (4-bromoshape-1-yl) -3- (pyridin-4-ylmethyl) urea (intermediate 2C)
See example 3, step 1 for a specific preparation procedure.
Step 2.1- (4- ((4- (2- (1-yl) ethanoxy) phenyl) ethyl) naphthalen-1-yl) -3- (pyridin-4-ylmethyl) urea (ID 221028A-1)
Starting material 2C (200 mg,0.56mmol,1.0 eq), starting material 2d (129 mg,0.56mmol,1.0 eq), pd (PPh 3 )Cl 2 (24 mg,0.056mmol,0.1 eq), cuI (10.69 mg,0.056mmol,0.1 eq) and triethylamine (227 mg,2.25mmol,4 eq) were dissolved in 10ml DMF and heated to 50deg.C and stirred for 2 hours; TLC (DCM/meoh=10/1, rf/product=0.2) showed that the starting material was reacted completely, a new spot was generated; the reaction was directly spin-dried through the column and rinsed with (DCM/meoh=50/1 to 10/1) to give ID221028A-1 (80 mg, 28.5%) as a gray solid.
1 H NMR(CDCl 3 ,300MHz)δ:8.10(d,J=9.0Hz,2H),6.99(d,J=9.0Hz,2H),4.25(t,J=6.0Hz,2H),2.85(t,J=6.0Hz,2H),2.56-2.53(m,4H),1.63-1.60(m,4H),1.52(t,J=3.0Hz,2H).
EXAMPLE 6 Synthesis of IY220831B-1 Compound
Figure BDA0004101053920000152
The compound IY220831B-1, named 1- (4- (2- (piceridin-1-yl) methoxy) phenyl) -3- (4- (3- (pyridin-4-ylmethyl) ureido) naphthalen-1-yl) urea, is synthesized as follows:
Figure BDA0004101053920000161
step 1.Tert-butyl (4- (3- (4- (2- (pi-1-yl) method) phenyl) ureido) napthlien-1-yl) carbamate (intermediate 2F)
Raw material 2A (1 g,3.87mmol, 1).0 eq), raw material 1c (0.85 g,3.87mmol,1.0 eq) and CDI (0.63 g,3.87mmol,1.0 eq) were dissolved in 50ml tetrahydrofuran and reacted for 10 hours with stirring at 70 ℃. TLC (DCM/MeOH=10/1, R) f Product=0.3) shows that the raw material reaction is completed and a new point is generated; the reaction was directly spin-dried through the column and rinsed with (DCM/meoh=50/1 to 10/1) to give intermediate 2F (1.02 g, 52.3%) as a yellow solid.
Step 2.Tert-butyl (4- (3- (4- (2- (pi-1-yl) method) phenyl) ureido) napthlien-1-yl) carbamate (intermediate 3F)
Raw material 2F (1 g,1.98mmol,1.0 eq) was dissolved in 20ml of methanol, and a 20ml, 4mol/L solution of hydrogen chloride in ethyl acetate was added; stirring at room temperature for reaction for 2 hours; TLC (DCM/MeOH=10/1, R) f Product=0) shows that the raw material reaction is completed and a new point is generated; the reaction solution was directly poured to give intermediate 3F (800 mg, 99%) as a white solid.
Step 3.1- (4- (2- (piperidine-1-yl) method) phenyl) -3- (4- (3- (pyridin-4-ylmethyl) ureido) naphthalen-1-yl) urea (IY 220831B-1)
Raw material 3F (200 mg,0.49mmol,1.0 eq) and raw material 5 (99 mg,0.49mmol,1.0 eq) were dissolved in 20ml tetrahydrofuran and reacted at 120℃for 10 hours with stirring; TLC (DCM/MeOH=10/1, R) f Product=0.1) shows that the raw material reaction is completed and a new point is generated; the reaction was directly spin-dried through the column and rinsed with (DCM/meoh=50/1 to 10/1) to give IY220831B-1 (101 mg, 39.1%) as a yellow solid.
1 H NMR(CDCl 3 ,300MHz)δ:8.05(d,J=9.0Hz,2H),6.93(d,J=9.0Hz,2H),4.15(t,J=6.0Hz,2H),2.85(t,J=6.0Hz,2H),2.55-2.53(m,4H),1.63-1.60(m,4H),1.51(t,J=3.0Hz,2H).
Example 7: proliferation inhibiting effect of IY220816B-1, ID 2208235A-1, IY221116B-1, ID221028A-1 and IY220831B-1 on tumor cells such as breast cancer, liver cancer and lung cancer
Respectively collecting tumor cells in logarithmic growth phase, and adjusting cell suspension concentration to 5×10 4 Each mL was added to a 96-well cell culture plate at 100ul per well volume. The DMSO is used as solvent for comparison, and the accepted STAT3 signal inhibitor WP1066 (Chinese name, (2E) -3- (6-bromo-2-pyridyl) -2-cyano is used-N- [ (1S) -1-phenylethyl]2-acrylamide, CAS 857064-38-1, of the structure
Figure BDA0004101053920000171
As positive control, the novel naphthyridine-piperidine compounds IY220816B-1, ID 2208235A-1, IY221116B-1, ID221028A-1, IY220831B-1 and the like are diluted by DMSO and added into culture wells, so that the final concentrations of the compounds in the system are respectively 0.1, 0.3, 1, 3, 10, 30, 100 and 300 (mu mol/L). After further culturing for 48h, 10. Mu.L of MTT solvent (5 mg/ml) was added to each well, incubated for 4h at 37℃and the culture supernatant was aspirated off, 150. Mu.L of DMSO was added to each well, shaking and decolorizing was performed for 10min on a shaker, reading was performed on a microplate reader, the OD at an absorbance wavelength of 490nm was measured, the results were recorded, and the cell growth curve was drawn with the dose of the compound as the abscissa and the absorbance as the ordinate. The statistical results of half inhibition rates (IC 50 values) of IY220816B-1, ID 2208235A-1, IY221116B-1, ID221028A-1, IY220831B-1 and the like on tumor cells are shown in FIG. 1.
The results shown in FIG. 1 indicate that compared with the positive control medicines WP1066, IY220816B-1, ID 2208235A-1, IY221116B-1, ID221028A-1, IY220831B-1 and the like have better proliferation inhibition effects on breast cancer, liver cancer, lung cancer, drug-resistant lung cancer, colon cancer, leukemia, stomach cancer, pancreatic cancer, prostate cancer and esophageal cancer cells, and especially the IY220816B-1 has the strongest tumor inhibition activity on breast cancer. The application takes IY220816B-1 as an important research object, and further researches on the anti-tumor effect and action mechanism of the compounds.
Example 8: inhibition of STAT3 signaling in breast cancer cells by IY220816B-1
1. Cell culture and drug addition A. MDA-MB-468 cells in logarithmic phase are digested with pancreatin, and prepared into 4×10 density with L-15 medium containing 10% foetal calf serum 5 individual/mL of single cell suspension was seeded into 6-well plates at 2mL of cell suspension per well. Incubator incubation at 37℃ (MDA-MB-468 cell culture without CO 2 ). After cell attachment, IY220816B-1 was added to give final concentrations of 0,0.015,0.03 and 0.06. Mu.M, respectively. c. After further culturing for 48h, cells were lysed with RIPA lysate and protein was collected。
2. Cell collection and lysis: a. the upper medium was discarded and the cells were washed twice with pre-chilled PBS. 100. Mu.L of precooled RIPA cell lysate (protease inhibitor and PMSF were added to the lysate at a ratio of 1:100) was added per well.
b. Lysing on ice for 3min, scraping off the cells with a cell scraper and collecting into a 1.5mL EP tube; the mixture was then placed on ice for 30min and vortexed once every 6 min. c.4℃and 12000g for 10min. d. Cell supernatants were transferred to new EP tubes. Cell supernatants are divided into two parts: 5 mu L of the sample is added into an EP tube of 1.5mL for BCA protein content measurement, and 45 mu L of 1 XPBS is added for even mixing; the remaining cell supernatants were quantified by taking 80. Mu.L, adding 5X SDS Loading Buffer. Mu.L, mixing well, boiling in boiling water for 10min, centrifuging, and loading or storing in a refrigerator at-20deg.C. e. Protein concentration determination:
(1) BCA working solution preparation: the total required amount of A and B mixed working fluid is calculated according to the number of the standard substance and the sample to be tested. The volume ratio of the solid agent A to the solid agent B is 50:1, preparing working solution, and uniformly mixing by vortex oscillation for standby.
(2) 1 XPBS diluted protein standard:
7 pipe number 1×PBS(μl) BSA standard dosage BSA standard (μg/ml)
A 0 100 2000
B 200 200 1000
C 200 200 (from tube B) 500
D 200 200 (taken from the C-tube) 250
E 200 200 (taken from the D-tube) 125
F 400 100 (taken from E-tube) 25
G 200 0 0 (blank)
(3) Protein standards and sample supernatants diluted with PBS (10-fold dilution) were each taken at 25 μl and added to a new 96-well plate. Then 200. Mu.L of BCA working solution prepared in advance was added respectively and mixed well. The bubbles are not generated by blowing, the cover of the 96-well plate is tightly covered, and the reaction is carried out for 30min in a constant temperature box at 37 ℃. (4) Taking out the 96-well plate, recovering to room temperature for 3-5 min, measuring the absorbance value of A562 on an enzyme label instrument, copying out the obtained value and storing the obtained value in an Excel table. A standard curve was made and the protein content of 1 μl per sample was calculated for protein loading.
3. SDS-PAGE: (1) The gel plate was fixed and 10% SDS-PAGE separating gel was prepared.
The release gel was prepared according to the following table: 10mL
Figure BDA0004101053920000181
Figure BDA0004101053920000191
(2) And (3) adding the mixed separating glue into 2 glue plates respectively, adding the glue plates to a position 1.0cm away from the top, filling the glue plates with absolute ethyl alcohol, and standing for 30-45 min. (3) After the gel is separated, the residual absolute ethyl alcohol is poured out, and the residual absolute ethyl alcohol is sucked clean by filter paper. (4) 5mL of 5% concentrated gel was prepared according to the following Table
Deionized water 2.77mL
30%(m/v)Acrylamide 830μL
0.5M Tris-HCl (pH 6.8) buffer 1.26mL
10%(m/v)SDS 50μL
10%(m/v)APS 50μL
TEMED 5μL
Total 5mL
(5) Slowly adding the prepared concentrated glue into a glue plate to avoid generating bubbles, inserting a comb, and standing for 30-45 min.
(6) The protein sample was removed and heated in a water bath at 100deg.C for 5min at 10000rpm and centrifuged for 5min. (7) The gel plate was fixed in an electrophoresis tank, SDS-PAGE electrophoresis buffer was added, the comb was pulled out, and the treated protein samples were sequentially added to the sample tank, 50. Mu.g per well of protein. (8) 80V electrophoresis for 40min. (9) Changing the voltage to 120V for electrophoresis for about 1.5 hours until bromophenol blue goes out of the colloid;
4. western-blot: (1) And (3) placing the SDS-PAGE gel subjected to electrophoresis into TBST buffer solution for rinsing once, and placing the protein gel into transfer buffer solution for soaking. (2) Soaking a layer of cotton pad in a membrane transfer buffer solution, clamping onto a membrane transferring instrument by using forceps, placing a blackboard, the cotton pad, filter paper, albumin glue, PVDF membrane, filter paper, the cotton pad and a whiteboard in order, clamping, and placing into the membrane transferring instrument. If bubbles exist between each two layers, the bubbles are removed by using a glass solid tube to lightly roll. And (3) opening a film transfer instrument, and performing constant-current transfer printing for 80 minutes at 300 mA. (4) The membranes were placed in TBST buffer and rinsed 3 times for 8min each. (5) blocking with 20mL of 5% BSA-TBST blocking solution at room temperature for 2h. (6) Primary antibody was added and incubated at 4℃at 60rpm overnight. (7) The membranes were washed three times with TBST at room temperature, 60rpm, 10min each. (8) adding a secondary antibody, and incubating for 1h at room temperature. (9) The membranes were washed three times with TBST at room temperature, 60rpm, 10min each. (10) 1ml of each of chemiluminescent substrate solid solution A and solution B is taken and developed for 2min at room temperature. (11) the liquid on the membrane is sucked by filter paper, and the membrane is subjected to machine aeration.
5. And (3) preparation of a reagent:
(1) 10% SDS: 1g of high purity (electrophoretic grade) SDS was weighed into a 10mL centrifuge tube, added with about 8mL deionized water, dissolved by heating, and stored at room temperature to a volume of 10 mL.
(2) 10% ammonium persulfate (Ammonium persulfate, AP): 1g of ammonium persulfate was weighed, dissolved by stirring after adding about 10mL of deionized water, and stored at 4 ℃.
(3) 5 Xrunning buffer: 15.1g of Tris, 94g of Glycine and 5.0g of SDS are weighed in a beaker, added with 1L of double distilled water for dissolution, stored at room temperature and diluted 5 times with time.
(4) Transfer buffer: 5.8g of Tris, 11.6g of glycine and 0.75g of SDS are weighed into a beaker, 700mL of double distilled water is added, the volume is fixed to 800mL after dissolution, and finally 200mL of methanol is added.
(5) 1.5mol/L Tris-HCl,100ml: then 18.15gtris is dissolved in 80ml water and adjusted to 8.8 by 4N HCl, and the volume is fixed to 100 ml.
(6) 0.5mol/L Tris-HCl,1000ml: 60.5 g of tris base was weighed, water was added to 850ml, concentrated hydrochloric acid was added and stirred until all dissolved, the pH was adjusted to 6.8, and water was added to 1L.
(7) TBS buffer: 8.8g of NaCl was weighed out in 800mL of distilled water, dissolved, added with 10mL of 1mol/L Tris HCl (pH 7.5), and kept at room temperature to a volume of 1L.
(8) TBST buffer: to 1L TBS buffer, 500. Mu.L of 20% Tween20 was added to give a final concentration of 0.1% Tween20, and the mixture was prepared immediately.
(9) Blocking solution, antibody dilution: 5% skim milk powder or BSA was added to TBST buffer and ready-to-use.
As shown in FIG. 2, the Western blot results show that 0.06 mu M of IY220816B-1 can significantly inhibit the expression levels of STAT3 total protein (T-STAT 3), a key marker p-STAT3 for STAT3 activation (Tyr 705) and a downstream target protein Cyclin D1 of STAT3 in MDA-MB-468 cells.
In conclusion, the result shows that the piperazine compound represented by IY220816B-1 can effectively inhibit proliferation of various tumor cells, has obvious inhibition effect on STAT3 and related proteins, and shows good targeted anti-tumor effect. According to the general way of drug development (conventional anti-tumor in-vitro screening is carried out firstly and then targeted research is carried out), the compound can be applied to tumor therapeutic drugs related to abnormal cell proliferation, and the anti-tumor drugs can be prepared by salifying with human bodies or mixing with a medicinal carrier.
Finally, what should be said is: the above embodiments are only for illustrating the technical solution of the present invention, but any equivalent replacement of the present invention and modification or partial replacement without departing from the spirit and scope of the present invention should be covered in the scope of the claims of the present invention.

Claims (7)

1. The naphtyl urea-piperidine compounds are characterized by having the following structural formula:
Figure FDA0004101053910000011
wherein R is 1 Selected from the group consisting of
Figure FDA0004101053910000012
2. The naphthylurea-piperidine type compound according to claim 1, characterized in that it is a compound of the following structure:
Figure FDA0004101053910000013
3. a biologically acceptable salt of the naphthylurea-piperidine compound of claim 1 or 2 with at least one of acetic acid, dihydrofolate, benzoic acid, citric acid, sorbic acid, propionic acid, oxalic acid, fumaric acid, maleic acid, hydrochloric acid, malic acid, phosphoric acid, sulfurous acid, sulfuric acid, vanillic acid, tartaric acid, ascorbic acid, boric acid, lactic acid, and ethylenediamine tetraacetic acid.
4. Use of a naphthylurea-piperidine compound according to any one of claims 1 to 3, and a biologically acceptable salt thereof, in the manufacture of an antitumor medicament.
5. Use of a naphthylurea-piperidine compound according to any one of claims 1 to 3, and a biologically acceptable salt thereof, for the manufacture of a medicament for the treatment of a disease associated with abnormal STAT3 cell signalling.
6. The use according to claim 4, wherein the antitumor drug is a drug for treating breast cancer, liver cancer, lung cancer, colon cancer, leukemia, stomach cancer, pancreatic cancer, prostate cancer and esophageal cancer.
7. The use according to claim 6, wherein the naphtyl urea-piperidine compounds are used for preparing medicines for inhibiting proliferation, inducing apoptosis or promoting apoptosis of breast cancer cells MDA-MB-468, liver cancer cells HepG2, HCC97H, lung cancer cells PC9, octenib-resistant lung cancer cells PC9-AR, colon cancer cells HT-29, leukemia cells Jurkat, gastric cancer cells HGC27, pancreatic cancer cells BxPC-3, prostate cancer cells Lncap and esophageal cancer cells KYSE450.
CN202310176580.9A 2023-02-28 2023-02-28 Naphthyl urea-piperidine compounds, and preparation method and application thereof Pending CN116178248A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310176580.9A CN116178248A (en) 2023-02-28 2023-02-28 Naphthyl urea-piperidine compounds, and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310176580.9A CN116178248A (en) 2023-02-28 2023-02-28 Naphthyl urea-piperidine compounds, and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN116178248A true CN116178248A (en) 2023-05-30

Family

ID=86434309

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310176580.9A Pending CN116178248A (en) 2023-02-28 2023-02-28 Naphthyl urea-piperidine compounds, and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN116178248A (en)

Similar Documents

Publication Publication Date Title
RU2528408C2 (en) Method for preparing dihydroindenamide compounds, pharmaceutical compositions containing these compounds and using them as protein kinase inhibitor
US20220098162A1 (en) Aphthylamine compound and biologically acceptable salt thereof, preparation method therefor, and application thereof
CN113444035B (en) Naphthyl urea compounds with anticancer effect and preparation method and application thereof
CN114702439B (en) Naphthyl urea-piperazine compound and preparation method and application thereof
CN110563706B (en) MDM2 protein degradation targeting chimera and preparation method and application thereof
CN101163684A (en) Quinazoline derivative having tyrosine kinase inhibitory activity
CN112047950B (en) Imidazo pyrazine derivative and synthetic method and application thereof
US20210101881A1 (en) Pyrimidine compound, preparation method thereof and medical use thereof
JP6917448B2 (en) Crystal form, salt form and production method of tyrosine kinase inhibitor
WO2007019884A1 (en) Novel 4-amino-thieno[3,2-c]pyridine-7-carboxylic acid amides
CN109820866A (en) A kind of c-Myc gene expression inhibitor and its application
Jing et al. SAR study on N 2, N 4-disubstituted pyrimidine-2, 4-diamines as effective CDK2/CDK9 inhibitors and antiproliferative agents
CN110885332B (en) PDE delta protein degradation targeting chimera and preparation method and application thereof
CN116178248A (en) Naphthyl urea-piperidine compounds, and preparation method and application thereof
CN109369620B (en) Pyridine compound, preparation method thereof and application thereof in resisting gastric cancer
CN114292270B (en) BTK inhibitor and preparation method and application thereof
CN110467637B (en) Bisaminyl chloropyrimidine compound containing phosphine oxide substituted aniline, preparation method and application thereof
CN108530337B (en) Indoleamide compound capable of selectively inhibiting gastric cancer cells
CN113956233B (en) Amide compound or pharmaceutically acceptable salt thereof, and preparation method and application thereof
WO2022068860A1 (en) Crystal form of pyrrolo heterocyclic derivative and preparation method therefor
CN117263851A (en) Pyridine-naphthyl urea-piperazine derivative, and preparation method and application thereof
CN107163028A (en) A kind of benzamides Hedgehog inhibitor and its preparation method and application
CN112759564B (en) Diaryl urea compound and its prepn and medicinal use
JP2005537258A (en) Novel anthracene derivatives and their use as pharmaceuticals
CN117567361A (en) Aryl alkyne compound as well as preparation method and application thereof

Legal Events

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