CN111153817B - phenoxy-N-phenylaniline derivatives and application thereof - Google Patents

phenoxy-N-phenylaniline derivatives and application thereof Download PDF

Info

Publication number
CN111153817B
CN111153817B CN202010066925.1A CN202010066925A CN111153817B CN 111153817 B CN111153817 B CN 111153817B CN 202010066925 A CN202010066925 A CN 202010066925A CN 111153817 B CN111153817 B CN 111153817B
Authority
CN
China
Prior art keywords
compound
hydrogen
phenoxy
nmr
cdcl
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
CN202010066925.1A
Other languages
Chinese (zh)
Other versions
CN111153817A (en
Inventor
李锐
杨杰
魏于全
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.)
Sichuan University
Original Assignee
Sichuan 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 Sichuan University filed Critical Sichuan University
Priority to CN202010066925.1A priority Critical patent/CN111153817B/en
Publication of CN111153817A publication Critical patent/CN111153817A/en
Application granted granted Critical
Publication of CN111153817B publication Critical patent/CN111153817B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C217/00Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton
    • C07C217/78Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having amino groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton
    • C07C217/80Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having amino groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton having amino groups and etherified hydroxy groups bound to carbon atoms of non-condensed six-membered aromatic rings
    • C07C217/82Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having amino groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton having amino groups and etherified hydroxy groups bound to carbon atoms of non-condensed six-membered aromatic rings of the same non-condensed six-membered aromatic ring
    • C07C217/92Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having amino groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton having amino groups and etherified hydroxy groups bound to carbon atoms of non-condensed six-membered aromatic rings of the same non-condensed six-membered aromatic ring the nitrogen atom of at least one of the amino groups being further bound to a carbon atom of a six-membered aromatic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/34Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups
    • C07C233/35Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/36Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom having the carbon atom of the carboxamide group bound to a hydrogen atom or to a carbon atom of an acyclic saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/08Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms
    • C07D295/096Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/44Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D317/46Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D317/48Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring
    • C07D317/50Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to atoms of the carbocyclic ring
    • C07D317/58Radicals substituted by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/02Systems containing only non-condensed rings with a three-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/56Ring systems containing bridged rings
    • C07C2603/58Ring systems containing bridged rings containing three rings
    • C07C2603/70Ring systems containing bridged rings containing three rings containing only six-membered rings
    • C07C2603/74Adamantanes

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

The invention relates to a phenoxy-N-phenylaniline derivative and application thereof, belonging to the technical field of novel colorectal cancer drugs. The present invention solves the problem of providing novel compounds having c-Myc inhibitory activity. The structural formula of the compound is shown as formula I. The invention designs and synthesizes a series of novel phenoxy-N-phenylaniline derivatives, and the compounds can be used as c-Myc inhibitors, have better inhibition effect on the proliferation of colorectal cancer cells, and provide a new choice for colorectal cancer drugs.
Figure DDA0002376249280000011

Description

phenoxy-N-phenylaniline derivatives and application thereof
Technical Field
The invention relates to a phenoxy-N-phenylaniline derivative and application thereof, belonging to the technical field of novel colorectal cancer drugs.
Background
Globally, colorectal cancer (CRC) is the second leading cause of cancer death, ranked second in common cancers in women, and third in common cancers in men. Statistically, in 2018, over 630 thousands of patients with colorectal cancer worldwide are newly diagnosed, and 180 thousands of patients are newly diagnosed, so that 880792 people die. The world economic forum estimates that the costs of examination and treatment associated with colorectal cancer in 2010 are around $ 316 billion, and CRC poses a huge economic burden to patients and society. Although there are some CRC treatments such as chemotherapy, surgery, radiotherapy and molecular targeted therapy, the clinical prognosis of these treatments is still not satisfactory, and especially for patients with CRC metastasis, the treatment effect is not significant. Furthermore, tumor heterogeneity will also lead to resistance responses in humans to chemotherapy and targeted therapies. At present, first-line medicaments for colorectal cancer patients, such as irinotecan, 5-fluorouracil and derivative prodrugs thereof, platinum medicaments and the like, have the defects of poor clinical curative effect, poor patient prognosis, serious drug resistance reaction and the like. Therefore, there is a need to find new effective drugs for CRC treatment.
The c-Myc is a member of Myc proto-oncogene family, the common genetic abnormality in CRC is the expression increase of the c-Myc, the importance of the expression of the c-Myc in CRC is proved in experimental researches of transgenic mice, and intestinal polyps can be inhibited by down-regulating the c-Myc, which is the most important lesion in the process of colon cancer. Therefore, expression of c-Myc can help to inhibit tumor development and progression. Currently, the types of action of existing c-Myc inhibitors mainly include: inhibition of Myc-Max protein-protein interactions, e.g., 10058-F4, 10074-G5, SF-4-017, NUCC-0176242; inhibition of Myc-Max protein-DNA binding interactions, e.g., Mycro 3, KJ Pyr 9; in combination with a c-Myc G-quadruplex (G4) stabilizer, such as indene triisoquinoline 5, IZCZ-3, and the like. The structural formula of the existing c-Myc inhibitor is shown in figure 1, and no clinically available c-Myc inhibitor medicine exists at present. Furthermore, many of the known c-Myc inhibitors are not suitable for clinical studies due to low in vivo efficacy or poor antitumor activity, and there is an urgent need to develop more structurally novel c-Myc inhibitors. Therefore, research and exploration of c-Myc small molecule inhibitors with novel frameworks are particularly important.
Disclosure of Invention
In view of the above drawbacks, the present invention provides a novel compound having c-Myc inhibitory activity.
The structural formula of the phenoxy-N-phenylaniline derivative is shown as formula I:
Figure BDA0002376249260000021
wherein R is1Is hydrogen or C1-C3 alkoxy; r2Is hydrogen or C1-C3 alkoxy; r3Is hydrogen or C1-C3 alkoxy; r4Is hydrogen or C1-C3 alkoxy;
R5is composed of
Figure BDA0002376249260000022
Figure BDA0002376249260000023
R6Is hydrogen, methyl, ethyl, hydroxyl, methoxy or halogen, n is any integer of 1-3, R7Is hydrogen or C1-C3 alkoxy.
Preferably, R1Is hydrogen or methoxy; r2Is hydrogen or methoxy; r3Is hydrogen or methoxy; r4Is hydrogen or methoxy; r5Is composed of
Figure BDA0002376249260000024
Figure BDA0002376249260000025
R6Is hydrogen, methyl, ethyl, hydroxyl, methoxy or halogen, n is any integer of 1-3, R7Is hydrogen or C1-C3 alkoxy.
Preferably, the phenoxy-N-phenylaniline derivative has a structural formula shown in formula ii or formula iii:
Figure BDA0002376249260000026
preferably, R1Is hydrogen or C1-C3 alkoxy; r2Is hydrogen or C1-C3 alkoxy; r3Is hydrogen or C1-C3 alkoxy; r4Is hydrogen or C1-C3 alkoxy; r5Is composed of
Figure BDA0002376249260000027
R6Is hydrogen, methyl, hydroxyl, methoxy, fluorine, chlorine or bromine. Preferably, R1Is hydrogen or methoxy; r2Is hydrogen or methoxy; r3Is hydrogen or methoxy; r4Is hydrogen or methoxy; r5Is composed of
Figure BDA0002376249260000028
R6Is hydrogen, methyl, hydroxyl, methoxy, fluorine, chlorine or bromine.
More preferably, R1Is hydrogen or C1-C3 alkoxy; r2Is hydrogen or C1-C3 alkoxy; r3Is hydrogen or C1-C3 alkoxy; r4Is hydrogen or C1-C3 alkoxy; r5Is composed of
Figure BDA0002376249260000031
Preferably, R is1Is hydrogen or methoxy; r2Is hydrogen or methoxy; r3Is hydrogen or methoxy; r4Is hydrogen or methoxy; r5Is composed of
Figure BDA0002376249260000032
Preferably, R1Is hydrogen or C1-C3 alkoxy; r2Is hydrogen or C1-C3 alkoxy; r3Is hydrogen or C1-C3 alkoxy; r4Is hydrogen or C1-C3 alkoxy; r5Is composed of
Figure BDA0002376249260000033
R7Is hydrogen or methoxy. Preferably, R1Is hydrogen or methoxy; r2Is hydrogen or methoxy; r3Is hydrogen or methoxy; r4Is hydrogen or methoxy; r5Is composed of
Figure BDA0002376249260000034
R7Is hydrogen or methoxy.
The invention also provides application of the phenoxy-N-phenylaniline derivative in preparing a medicine for treating colorectal cancer.
The compound has a certain curative effect on colorectal cancer, and can be used for preparing a colorectal cancer medicament.
The invention also provides application of the phenoxy-N-phenylaniline derivative in preparation of a c-Myc inhibitor.
The phenoxy-N-phenylaniline derivative can be used as a c-Myc inhibitor.
The invention also provides isomers, pharmaceutically acceptable salts and hydrates of the phenoxy-N-phenylaniline derivatives.
The invention also provides application of the isomer, the pharmaceutically acceptable salt and the hydrate of the phenoxy-N-phenylaniline derivative in preparing medicaments for treating colorectal cancer.
The invention also provides an application of the isomer, the pharmaceutically acceptable salt and the hydrate of the phenoxy-N-phenylaniline derivative in the preparation of a c-Myc inhibitor.
The invention also provides a pharmaceutical composition, which consists of an effective component and pharmaceutically acceptable auxiliary materials, wherein the effective component contains a therapeutically effective amount of the phenoxy-N-phenylaniline derivative or the isomer thereof or pharmaceutically acceptable salt or hydrate thereof.
Compared with the prior art, the invention has the following beneficial effects:
the invention designs and synthesizes a series of novel phenoxy-N-phenylaniline derivatives, and the compounds can be used as c-Myc inhibitors, have better inhibition effect on the proliferation of colorectal cancer cells, and provide a new choice for colorectal cancer drugs.
Drawings
FIG. 1 is a structural formula of some existing c-Myc inhibitors.
FIG. 2 shows the effect of compound 42 of the present invention on the viability of CRC and normal hepatocyte LO 2. Wherein A is the IC50 value of the compound 42 to HT29, HCT15, HCT116 and DLD-1, and B is the toxicity of the compound 42, 5-fluorouracil and cisplatin to LO 2. Relative to DMSO, # P <0.01, # P < 0.001.
FIG. 3 is a graph showing the binding pattern of compound 42 of the present invention to c-Myc/Max, wherein A is the binding pattern of compound 42 to key amino acid residues of c-Myc/Max; b is the binding mode of compound 42 in the surface of the c-Myc/Max dimer; c is a computer binding mode of a complex with compound 42 of Myc-Max-DNA.
FIG. 4 is a colony cluster of HT29 cells and HCT15 cells detected after 15 days of treatment with different concentrations of compound 42.
FIG. 5 shows the results of flow cytometry analysis of Compound 42, wherein A is the cell cycle distribution analysis of HT29 and HCT15 cells treated with different concentrations of Compound 42 for 24 h; b is the level of apoptosis and apoptotic cell analysis of cells treated with compound 42 at a concentration for 24 h. P <0.05,. P <0.01 relative to DMSO; p < 0.001.
FIG. 6 is the effect of compound 42 on c-Myc expression as determined by Western blotting.
FIG. 7 shows the results of an in vivo experiment of Compound 42 of the present invention in a HT29 tumor xenograft model. Wherein A is the growth inhibition of HT29 xenograft tumor in female BALB/c nude mice treated by compound 42; b is the average body weight of mice in each group during the 30 day treatment period; c is the tumor weight of each group of mice at the end of treatment. P <0.01 relative to DMSO; p < 0.001.
Detailed Description
The structural formula of the phenoxy-N-phenylaniline derivative is shown as formula I:
Figure BDA0002376249260000041
wherein R is1Is hydrogen or C1-C3 alkoxy; r2Is hydrogen or C1-C3 alkoxy; r3Is hydrogen or C1-C3 alkoxy; r4Is hydrogen or C1-C3 alkoxy;
R5is composed of
Figure BDA0002376249260000042
Figure BDA0002376249260000043
R6Is hydrogen, methyl, ethylHydroxyl, methoxyl or halogen, n is any integer of 1-3, R7Is hydrogen or C1-C3 alkoxy.
The C1-C3 alkoxy group is an alkoxy group having 1-3 carbon atoms, such as methoxy, ethoxy, propoxy and the like.
The c-Myc protein is a member of the Intrinsically Disordered (ID) protein, characterized by a lack of stable structure and extensive backbone flexibility, a relevant but challenging goal of drug discovery, and c-Myc-Max interaction inhibitors are of greatest value for clinical research in order to seek new c-Myc inhibitors. Studies of the core structure of existing small molecule inhibitors have found that most inhibitors comprise long molecular structures because the c-Myc-Max dimer interface is a left-handed, parallel, and four-helix bundle, with each monomer consisting of two R helices separated by a loop. Accordingly, the invention designs a long molecule consisting of three benzene rings, namely, the phenoxy-N-phenylaniline derivative shown in the formula I, by combining a core structure.
The compound is designed as follows:
Figure BDA0002376249260000051
the study shows that R5The choice of groups, the linkage of region II to region I, and the linkage of region I to region III may all contribute significantly to biological activity.
Preferably, R1Is hydrogen or methoxy; r2Is hydrogen or methoxy; r3Is hydrogen or methoxy; r4Is hydrogen or methoxy; r5Is composed of
Figure BDA0002376249260000052
Figure BDA0002376249260000053
R6Is hydrogen, methyl, ethyl, hydroxyl, methoxy or halogen, n is any integer of 1-3, R7Is hydrogen or C1-C3 alkoxy.
In one embodiment, the point of attachment of zone II to zone I is at the C-3 position, i.e., a compound of formula II:
Figure BDA0002376249260000054
as another embodiment, the point of attachment of zone II to zone I is at the C-4 position, i.e., a compound of formula III:
Figure BDA0002376249260000055
as one of the preferred embodiments, R1Is hydrogen or C1-C3 alkoxy; r2Is hydrogen or C1-C3 alkoxy; r3Is hydrogen or C1-C3 alkoxy; r4Is hydrogen or C1-C3 alkoxy; r5Is composed of
Figure BDA0002376249260000056
R6Is hydrogen, methyl, hydroxyl, methoxy, fluorine, chlorine or bromine.
Preferably, R1Is hydrogen or methoxy; r2Is hydrogen or methoxy; r3Is hydrogen or methoxy; r4Is hydrogen or methoxy; r5Is composed of
Figure BDA0002376249260000061
R6Is hydrogen, methyl, hydroxyl, methoxy, fluorine, chlorine or bromine.
More preferably, R1Is hydrogen or C1-C3 alkoxy; r2Is hydrogen or C1-C3 alkoxy; r3Is hydrogen or C1-C3 alkoxy; r4Is hydrogen or C1-C3 alkoxy; r5Is composed of
Figure BDA0002376249260000062
Preferably, R is1Is hydrogen or methoxy; r2Is hydrogen or methoxy; r3Is hydrogen or methoxy; r4Is hydrogen or methoxy; r5Is composed of
Figure BDA0002376249260000063
As another preferred embodiment, R1Is hydrogen or C1-C3 alkoxy; r2Is hydrogen or C1-C3 alkoxy; r3Is hydrogen or C1-C3 alkoxy; r4Is hydrogen or C1-C3 alkoxy; r5Is composed of
Figure BDA0002376249260000064
R7Is hydrogen or methoxy. Preferably, R1Is hydrogen or methoxy; r2Is hydrogen or methoxy; r3Is hydrogen or methoxy; r4Is hydrogen or methoxy; r5Is composed of
Figure BDA0002376249260000065
R7Is hydrogen or methoxy.
The following are structural formulae of some specific preferred compounds of the present invention.
Figure BDA0002376249260000066
Figure BDA0002376249260000071
The preparation method of the phenoxy-N-phenylaniline derivative can adopt a conventional method, and the specific preparation method is described in example 1.
The phenoxy-N-phenylaniline derivative can be used for preparing a colorectal cancer medicament and has a certain curative effect on colorectal cancer.
The phenoxy-N-phenylaniline derivative can be used as a c-Myc inhibitor.
The invention also provides an isomer, a pharmaceutically acceptable salt and a hydrate of the phenoxy-N-phenylaniline derivative shown as the general formula I. Among the pharmaceutically acceptable salts, the compounds of formula I include, but are not limited to, salts with inorganic acids such as hydrochloric acid, sulfuric acid, phosphoric acid, phosphorous acid, hydrobromic acid and nitric acid, and salts with various organic acids such as malic acid, maleic acid, citric acid, fumaric acid, tartaric acid, succinic acid, acetic acid, lactic acid, p-toluenesulfonic acid, methanesulfonic acid, palmitic acid, and the like. Some of the compounds of the present invention may be crystallized or recrystallized using water or various organic solvents, in which case various solvates may be formed. The present invention includes those stoichiometric solvates, including hydrates, as well as compounds containing variable amounts of water that are formed when prepared by a low pressure sublimation drying process.
The invention also provides application of the isomer, the pharmaceutically acceptable salt and the hydrate of the phenoxy-N-phenylaniline derivative in preparing medicaments for treating colorectal cancer.
The invention also provides an application of the isomer, the pharmaceutically acceptable salt and the hydrate of the phenoxy-N-phenylaniline derivative in the preparation of a c-Myc inhibitor.
The phenoxy-N-phenylaniline derivative or its isomer or a pharmaceutically acceptable salt or hydrate thereof of the present invention can be used alone or in the form of a pharmaceutical composition together with a pharmaceutically acceptable carrier or excipient, and when used in the form of a pharmaceutical composition, a therapeutically effective amount of the compound of the present invention or a pharmaceutically acceptable salt or hydrate thereof and one or more pharmaceutically acceptable carriers or diluents are usually combined to make an appropriate administration form or dosage form. Accordingly, the present invention also provides a pharmaceutical composition comprising a therapeutically effective amount of a phenoxy-N-phenylaniline derivative according to the invention, all possible isomers thereof or a pharmaceutically acceptable salt or hydrate thereof and at least one pharmaceutically acceptable carrier.
Pharmaceutical compositions of the compounds of the present invention may be administered in any of the following ways: oral, aerosol inhalation, rectal, nasal, vaginal, topical, parenteral such as subcutaneous, intravenous, intramuscular, intraperitoneal, intrapin, intraventricular, intrasternal or intracranial injection or infusion, or by means of an explanted reservoir, with oral, intramuscular, intraperitoneal or intravenous administration being preferred.
The compounds of the present invention or pharmaceutical compositions containing them may be administered in unit dosage form. The administration dosage form can be liquid dosage form or solid dosage form. The liquid dosage form can be true solution, colloid, microparticle, emulsion, or warm suspension. Other dosage forms such as tablet, capsule, dripping pill, aerosol, pill, powder, solution, warm suspension, emulsion, granule, suppository, lyophilized powder for injection, clathrate, implant, patch, liniment, etc.
The pharmaceutical compositions of the present invention may also contain conventional carriers, including but not limited to: ion exchangers, aluminum oxide, aluminum stearate, lecithin, serum proteins such as human serum albumin, buffer substances such as phosphates, glycerol, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, sodium hydrogen phosphate, potassium hydrogen phosphate, sodium oxide, zinc salts, colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, cellulosic substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, beeswax, lanolin and the like. The carrier may be present in the pharmaceutical composition in an amount of 1 to 98% by weight, typically about 80% by weight. For convenience, the local anesthetic, preservative, buffer, etc. may be dissolved directly in the vehicle.
The following examples are provided to further illustrate the embodiments of the present invention and are not intended to limit the scope of the present invention.
Preparation of the Compound of example 1
The phenoxy-N-phenylaniline derivative is synthesized and designed according to the following process:
Figure BDA0002376249260000081
wherein, the synthetic process of the compounds 7-38 is as follows:
Figure BDA0002376249260000091
reagents and conditions: 1-fluoro-3, 5-dimethoxybenzene, POCl3DMF, 0 ℃ to 60 ℃, 4h, yield 62.30 percent; (ii) 3-aminophenol, bromobenzene, t-BuONa, chlorine (2-dicyclohexylphosphino-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2-aminoethylphenyl)]Palladium (II) (Brettphos Palladicycle), 1, 4-dioxane, 90 ℃, 2h, 86.01% yield; (iii) 3- (phenylamino) phenol, K2CO3DMSO, 100 ℃, 3h, and the yield is 73-85%; (iv) NaBH (AcO)3, AcOH, DMF, 80 ℃ overnight. The yield is 45-60%.
Synthesis of Compound 3 (4-fluoro-2, 6-dimethoxybenzaldehyde): 1-fluoro-3, 5-dimethoxybenzene (Compound 1) (10g, 64.06mmol) was added to anhydrous DMF (20mL) under argon, stirred at 0 ℃ for 20min, POCl was added over 15min3(11.77g, 76.87mmol) and the reaction mixture was stirred at room temperature for 30min and then heated at 60 ℃ for 4 h. After completion of the reaction, it was cooled to room temperature and poured into ice water (400 mL). The aqueous phase was adjusted to pH 9-10 with 4N NaOH, extracted twice with ethyl acetate (2 × 400mL), the organic phases combined and washed with brine, anhydrous MgSO4And (5) drying. The organic layer was removed by vacuum rotary evaporation and purified by column chromatography to give 7.35g of the product as a white solid. The yield was 62.30%.1H NMR(400MHz,CDCl3)δ10.44(s,1H),6.77(s,2H),3.91(s,6H).ESI-MS:mass calcd for[M+H]+(C9H9FO3)185.05;found m/z,185.06.
Synthesis of Compound 5(3- (phenylamino) phenol): 3-aminophenol (5g, 45.82mmol) and bromobenzene (8.63g, 54.98mmol), sodium tert-butoxide (1.32g, 137.46mmol), Brettphos Palladacycle (182.78mg, 0.23mmol, 0.5 mmol%) were added to 25mL of anhydrous 1, 4-dioxane and stirred at 90 ℃ for 2h under argon. Completion of the reaction was monitored by TLC. The reaction mixture was concentrated in vacuo, 500mL of water was added, extracted twice with ethyl acetate (2X 400mL), and the organic phases were combined, washed with brine, and anhydrous MgSO4And (5) drying. The organic layer was concentrated in vacuo and purified by silica gel chromatography to give 4.31g of a brown solid. The yield was 86.01%.1H NMR(400MHz,CDCl3)δ7.28(dd,J=7.0,1.5Hz,1H),7.25(s,1H),7.14–7.06(m,3H),6.95(dd,J=10.5,4.2Hz,1H),6.64–6.58(m,1H),6.56(t,J=2.2Hz,1H),6.37(ddd,J=8.0,2.3,0.7Hz,1H),5.68(s,1H),4.71(s,1H).ESI-MS:mass calcd for[M+H]+(C12H11NO)186.08;found m/z,186.09.
Synthesis of Compounds 6a-6d general procedure A: to 3- (phenylamino) phenol (3g, 16.20mmol) and K in anhydrous DMSO (15mL) under an argon atmosphere2CO3(6.72g, 48.59mmol) was added to a solution of 4-fluorobenzaldehyde (16.20mmol) with various substituents with stirring. The reaction was left at 100 ℃ for 3h and checked for completion by thin layer silica gel plate (TLC). After completion of the reaction, the reaction was quenched with cold water (100mL), extracted with DCM, and extracted with anhydrous MgSO4Drying and concentration in vacuo afforded the crude product, which was purified by silica gel chromatography to afford the desired compound.
Wherein, compound 6 a: prepared according to the above procedure A from compound 5 and 4-fluoro-2, 6-dimethoxybenzaldehyde. Yield 85.21%, brown solid.1H NMR(400MHz,CDCl3)δ10.30(s,1H),7.25–7.20(m,2H),7.17(s,1H),7.07–7.02(m,2H),6.92(t,J=7.4Hz,1H),6.80(ddd,J=8.1,2.2,0.8Hz,1H),6.73(t,J=2.2Hz,1H),6.53(ddd,J=8.1,2.3,0.8Hz,1H),6.10(s,2H),3.74(s,6H).ESI-MS:mass calcd for[M+H]+(C21H19NO4)350.13;found m/z,350.23.
Compound 6 b: prepared according to the above procedure A from compound 5 and 4-fluoro-2-methoxybenzaldehyde. Yield 82.35%, white solid.1H NMR(400MHz,CDCl3)δ10.31(s,1H),7.77(d,J=8.6Hz,1H),7.30–7.23(m,3H),7.09(d,J=7.6Hz,2H),6.96(t,J=7.4Hz,1H),6.87(dd,J=8.1,1.6Hz,1H),6.78(t,J=2.1Hz,1H),6.62–6.52(m,3H),5.90(s,1H),3.85(s,3H).ESI-MS:mass calcd for[M+H]+(C20H17NO3)320.12;found m/z,320.13.
Compound 6 c: prepared according to the above procedure A from compound 5 and 4-fluoro-3-methoxybenzaldehyde. Yield 78.82%, colorless oil.1H NMR(400MHz,CDCl3)1H NMR(400MHz,CDCl3)δ9.89(s,1H),7.51(d,J=1.7Hz,1H),7.39(dd,J=8.2,1.8Hz,1H),7.29–7.22(m,3H),7.08(d,J=7.8Hz,2H),6.97(dd,J=15.6,7.8Hz,2H),6.83(dd,J=8.1,2.0Hz,1H),6.77(t,J=2.1Hz,1H),6.56(dd,J=8.1,2.2Hz,1H),3.96(s,3H).ESI-MS:mass calcd for[M+H]+(C20H17NO3)320.12;found m/z,320.12.
Compound 6 d: prepared according to the above method a from compound 5 and 4-fluorobenzaldehyde. Yield 73.55%, colorless oil.1H NMR(400MHz,CDCl3)δ9.92(s,1H),7.88–7.82(m,2H),7.31–7.25(m,3H),7.09(ddd,J=7.9,4.8,1.8Hz,4H),6.97(dd,J=10.5,4.2Hz,1H),6.90–6.85(m,1H),6.78(t,J=2.2Hz,1H),6.59(ddd,J=8.1,2.2,0.7Hz,1H),5.79(s,1H).ESI-MS:mass calcd for[M+H]+(C19H15NO2)290.11;found m/z,290.12.
Synthesis of compounds 7-38 general procedure B: in anhydrous DMF (2mL), under argon atmosphere, compounds 6a-6d (0.30mmol), sodium triacetoxyborohydride (127mg, 0.60mmol), various amines (0.36mmol) and acetic acid (0.03mmol) were added, stirred overnight at 80 ℃ and checked for completion with thin layer silica gel plate (TLC). After completion of the reaction, the reaction was quenched with cold water (100mL), extracted with ethyl acetate, and then extracted with anhydrous MgSO4Drying, concentration in vacuo afforded the crude product, which was purified by flash column chromatography to afford the desired compound.
Synthesis of compound 7: prepared according to method B above from compound 6a and methylamine hydrochloride. Yield 52.50% brown solid.1H NMR(400MHz,CDCl3)δ7.28(d,J=7.8Hz,2H),7.22(t,J=8.1Hz,1H),7.10(d,J=7.9Hz,2H),6.96(t,J=7.3Hz,1H),6.84(d,J=8.1Hz,1H),6.73(s,1H),6.56(d,J=7.9Hz,1H),6.23(s,2H),4.21(s,2H),3.81(s,6H),2.54(s,3H).13C NMR(101MHz,CDCl3)δ160.42,160.03,157.26,145.23,142.27,130.47,129.41,121.76,118.68,112.52,111.12,107.78,101.09,94.70,56.01,40.47,31.19.ESI-MS:mass calcd for[M+H]+(C22H24N2O3)365.18;found m/z,365.19.
Synthesis of compound 8: prepared according to method B above from compound 6a and cyclopropylamine. Yield 61.05% brown solid.1H NMR(400MHz,CDCl3)δ7.25(d,J=7.2Hz,2H),7.19(t,J=8.1Hz,1H),7.09(d,J=7.6Hz,2H),6.95(t,J=7.4Hz,1H),6.80(dd,J=8.0,2.0Hz,1H),6.71(t,J=2.2Hz,1H),6.54(dd,J=8.1,2.2Hz,1H),6.26(s,2H),5.81(s,1H),4.02(s,2H),3.77(s,6H),2.20(tt,J=7.2,3.7Hz,1H),0.71(m,2H),0.52(q,J=6.6Hz,2H).13C NMR(101MHz,CDCl3)δ159.82,145.11,142.36,130.39,129.38,121.64,118.56,112.16,110.77,107.44,95.18,55.92,40.47,29.37,4.40.ESI-MS:mass calcd for[M+H]+(C24H26N2O3)391.19;found m/z,391.20.
Synthesis of compound 9: prepared according to method B above from compound 6a and 2-propylamine. Yield 52.36%, brown solid.1H NMR(400MHz,CDCl3)δ7.29(s,2H),7.25(s,1H),7.20(t,J=8.1Hz,1H),7.10(d,J=7.7Hz,2H),6.95(t,J=7.3Hz,1H),6.82(d,J=8.1Hz,1H),6.70(d,J=2.0Hz,1H),6.55(dd,J=8.1,1.7Hz,1H),6.25(s,2H),5.92(s,1H),4.02(s,2H),3.77(s,6H),2.95(dt,J=12.9,6.4Hz,1H),1.23(d,J=6.4Hz,6H).13C NMR(101MHz,CDCl3)δ159.57,157.93,145.09,142.36,130.38,129.38,121.64,118.55,112.05,110.67,107.33,95.42,55.88,46.94,40.29,37.81,23.17.ESI-MS:mass calcd for[M+H]+(C24H28N2O3)393.21;found m/z,393.21.
Synthesis of compound 10: prepared according to the above method B from compound 6a and dimethylamine. Yield 45.77%, brown solid.1H NMR(400MHz,CDCl3)δ7.31–7.27(m,2H),7.23(d,J=8.1Hz,1H),7.11(d,J=7.6Hz,2H),6.97(t,J=7.3Hz,1H),6.86(dd,J=8.1,1.9Hz,1H),6.76(t,J=2.2Hz,1H),6.58(dd,J=8.0,2.1Hz,1H),6.25(s,2H),5.89(s,1H),4.22(s,2H),3.81(s,6H),2.72(s,6H).13C NMR(101MHz,CDCl3)δ161.35,160.49,156.76,145.32,142.18,130.58,129.43,121.90,118.78,112.95,111.44,108.02,99.54,94.50,56.04,49.03,42.40,29.70.ESI-MS:mass calcd for[M+H]+(C23H26N2O3)379.19;found m/z,379.20.
Synthesis of compound 11: prepared according to method B above from compound 6a and 2,2' -azadiylbis (ethan-1-ol). Yield 56.15%, yellow oil.1H NMR(400MHz,CDCl3)δ7.29(d,J=7.8Hz,1H),7.23(d,J=8.1Hz,1H),7.11(d,J=7.9Hz,1H),6.97(t,J=7.3Hz,1H),6.86(dd,J=8.2,1.5Hz,1H),6.76(s,1H),6.58(dd,J=7.9,1.6Hz,1H),6.25(s,1H),5.88(s,1H),4.35(s,1H),3.99(s,2H),3.81(s,3H),3.28(s,2H).13C NMR(101MHz,CDCl3)δ160.02,130.58,129.41,121.84,118.76,112.88,111.44,108.05,94.65,56.15,56.10,55.35,47.58.ESI-MS:mass calcd for[M+H]+(C23H26N2O3)379.19;found m/z,379.20.ESI-MS:mass calcd for[M+H]+(C25H30N2O5)439.22;found m/z,439.23.
Synthesis of compound 12: prepared according to method B above from compound 6a and N- (2-aminoethyl) acetamide. Yield 61.42%, brown oil.1H NMR(400MHz,CDCl3)δ7.27(d,J=7.0Hz,2H),7.20(t,J=8.1Hz,1H),7.13–7.08(m,2H),7.00(s,1H),6.94(t,J=7.3Hz,1H),6.83(dd,J=8.1,1.5Hz,1H),6.64(t,J=2.2Hz,1H),6.55(dd,J=8.1,1.6Hz,1H),6.26(s,2H),6.08(s,1H),3.92(s,2H),3.77(s,6H),3.39(d,J=5.3Hz,2H),2.84–2.73(m,2H),1.97(s,3H).13C NMR(101MHz,CDCl3)δ170.65,159.51,158.60,158.02,145.15,142.43,130.37,129.36,121.57,118.51,111.85,110.59,107.22,95.44,55.88,46.94,40.29,37.81,23.17.ESI-MS:mass calcd for[M+H]+(C25H29N3O4)436.22;found m/z,436.23.
Synthesis of compound 13: prepared according to method B above from compound 6a and 2- (piperazin-1-yl) ethan-1-ol. Yield 53.18%, brown oil.1H NMR(400MHz,CDCl3)δ7.28(dd,J=7.0,1.4Hz,2H),7.23(t,J=8.1Hz,1H),7.14–7.08(m,2H),6.96(t,J=7.4Hz,1H),6.84(dd,J=8.1,1.5Hz,1H),6.75(t,J=2.2Hz,1H),6.57(dd,J=8.1,1.6Hz,1H),6.25(s,2H),5.88(s,1H),4.08(s,2H),3.76(s,6H),3.70–3.65(m,2H),2.93(s,7H),2.72–2.66(m,2H).13C NMR(101MHz,CDCl3)δ160.54,157.23,145.23,142.25,130.49,129.41,121.81,118.69,112.62,111.18,107.79,99.99,94.89,59.16,57.64,55.93,50.55,48.17.ESI-MS:mass calcd for[M+H]+(C27H33N3O4)464.25;found m/z,464.26.
Synthesis of compound 14: prepared according to method B above from compound 6a and 1-ethylpiperazine. Yield 62.33%, brown solid.1H NMR(400MHz,CDCl3)δ7.29(s,1H),7.25(s,1H),7.20(t,J=8.1Hz,1H),7.10(d,J=8.0Hz,2H),6.95(t,J=7.3Hz,1H),6.81(d,J=8.1Hz,1H),6.71(s,1H),6.55(dd,J=8.1,1.9Hz,1H),6.24(s,2H),5.88(s,1H),3.84(s,2H),3.73(s,6H),2.72(d,J=46.0Hz,8H),2.50–2.44(m,2H),1.10(t,J=7.1Hz,3H).13C NMR(101MHz,CDCl3)δ160.30,158.01,145.04,142.40,130.34,129.38,121.61,118.55,112.07,110.73,107.38,95.41,55.82,52.10,51.92,51.50,48.48,11.67.ESI-MS:mass calcd for[M+H]+(C27H33N3O3)448.25;found m/z,448.25.
Synthesis of compound 15: prepared according to method B above from compound 6a and amantadine. Yield 41.27%, yellow solid.1H NMR(400MHz,CDCl3)δ7.28(s,1H),7.24(s,1H),7.21(d,J=8.1Hz,1H),7.12(d,J=7.7Hz,2H),6.94(t,J=7.3Hz,1H),6.85(d,J=8.1Hz,1H),6.69(s,1H),6.55(dd,J=8.0,1.6Hz,1H),6.23(d,J=8.1Hz,2H),4.16(s,2H),3.82(s,6H),2.13(s,3H),2.00(s,6H),1.64(s,6H).13C NMR(101MHz,CDCl3)δ159.91,159.42,157.66,145.22,142.36,130.38,129.34,121.57,118.62,111.91,110.82,107.57,102.98,95.45,58.30,56.16,38.68,35.58,33.64,29.11.ESI-MS:mass calcd for[M+H]+(C31H36N2O3)485.27;found m/z,485.28.
Synthesis of compound 16: following method B above, starting from Compound 6a and NaBH4(2mg, 0.057mmol) in methanol at room temperature for 2 min. After the reaction, concentration in vacuo afforded the crude product, which was purified by silica gel chromatography to afford compound 16. Yield 65.25%, colorless oil.1H NMR(400MHz,CDCl3)δ7.28–7.24(m,2H),7.19(t,J=8.1Hz,1H),7.08(d,J=7.6Hz,2H),6.96(d,J=7.3Hz,1H),6.79(dd,J=8.0,1.9Hz,1H),6.72(t,J=2.2Hz,1H),6.54(dd,J=8.1,1.8Hz,1H),6.26(s,2H),5.78(s,1H),4.73(s,2H),3.77(s,6H).ESI-MS:mass calcd for[M+H]+(C21H21NO4)352.15;found m/z,352.16.
Synthesis of compound 17: prepared according to method B above from compound 6a and benzylamine. Yield 58.02%, brown solid.1H NMR(400MHz,CDCl3)δ7.31(dt,J=9.7,4.2Hz,5H),7.23(d,J=6.4Hz,2H),7.19(t,J=8.1Hz,1H),7.08(d,J=7.8Hz,2H),6.94(t,J=7.3Hz,1H),6.78(dd,J=8.0,1.6Hz,1H),6.72(t,J=2.1Hz,1H),6.54(dd,J=8.1,2.0Hz,1H),6.26(s,2H),5.75(s,1H),3.84(s,2H),3.76(s,2H),3.73(s,6H).13C NMR(101MHz,CDCl3)δ159.45,158.45,157.27,144.94,142.43,140.86,130.30,129.39,128.20,126.67,121.62,118.50,111.91,111.74,110.49,107.19,95.62,55.73,53.12,41.05.ESI-MS:mass calcd for[M+H]+(C28H28N2O3)441.21;found m/z,441.21.
Synthesis of compound 18: prepared according to method B above from compound 6a and (4-fluorophenyl) methylamine. Yield 53.72% yellow solid.1H NMR(400MHz,CDCl3)δ7.32–7.23(m,4H),7.18(t,J=8.1Hz,1H),7.07(d,J=7.7Hz,2H),7.02–6.91(m,3H),6.77(dd,J=8.1,1.4Hz,1H),6.71(t,J=2.0Hz,1H),6.53(dd,J=8.1,1.7Hz,1H),6.25(s,2H),5.80(s,1H),3.82(s,2H),3.72(s,8H).13C NMR(101MHz,CDCl3)δ163.03,160.61,159.44,158.39,157.37,144.99,142.46,136.61,136.58,130.32,129.75,129.67,129.59,129.39,121.60,121.25,118.50,115.01,114.80,111.97,111.59,110.50,107.23,95.59,55.73,52.33,40.92.ESI-MS:mass calcd for[M+H]+(C28H27FN2O3)459.20;found m/z,459.21.
Synthesis of compound 19: prepared according to the above procedure B from compound 6a and (4-chlorophenyl) methylamine. Yield 58.87%, brown solid.1H NMR(400MHz,CDCl3)δ7.27–7.22(m,6H),7.19(d,J=8.1Hz,1H),7.10–7.05(m,2H),6.93(t,J=7.4Hz,1H),6.80–6.75(m,1H),6.71(t,J=2.2Hz,1H),6.56–6.51(m,1H),6.25(s,2H),5.80(s,1H),3.81(s,2H),3.72(s,8H).13C NMR(101MHz,CDCl3)δ159.44,158.34,157.46,144.99,142.45,139.26,132.34,130.33,129.58,129.39,128.28,121.61,118.51,112.01,111.35,110.53,107.26,95.54,55.73,52.25,40.86.ESI-MS:mass calcd for[M+H]+(C28H27ClN2O3)475.17;found m/z,475.17.
Synthesis of compound 20: prepared according to method B above from compound 6a and (4-bromophenyl) methylamine. Yield 61.25%, brown solid.1H NMR(400MHz,CDCl3)δ7.42(d,J=8.3Hz,2H),7.26–7.17(m,5H),7.08(d,J=7.6Hz,2H),6.94(t,J=7.4Hz,1H),6.80–6.76(m,1H),6.72(t,J=2.2Hz,1H),6.56–6.52(m,1H),6.25(s,2H),5.77(s,1H),3.81(s,2H),3.72(d,J=6.5Hz,8H).13C NMR(101MHz,CDCl3)δ159.43,158.33,157.46,144.97,142.44,139.79,131.23,130.33,129.96,129.39,121.63,120.42,118.51,112.01,111.35,110.55,107.26,95.54,55.73,52.28,40.85.ESI-MS:mass calcd for[M+H]+(C28H27BrN2O3)519.12,521.12;found m/z,519.13,521.13.
Synthesis of compound 21: prepared according to method B above from compound 6a and 4- (aminomethyl) phenol. Yield 48.73% brown oil.1H NMR(400MHz,CDCl3)δ7.26–7.22(m,2H),7.18(t,J=8.1Hz,1H),7.07(dd,J=8.5,1.0Hz,2H),7.02(d,J=8.4Hz,2H),6.93(t,J=7.3Hz,1H),6.81–6.77(m,1H),6.69(t,J=2.2Hz,1H),6.58–6.51(m,3H),6.23(s,2H),5.84(s,1H),3.89(s,2H),3.70(s,6H),3.62(s,2H).13C NMR(101MHz,CDCl3)δ159.46,158.21,157.75,156.24,145.01,142.45,130.33,129.97,129.71,129.37,121.60,118.51,115.76,111.97,110.61,110.16,107.34,95.46,55.74,52.09,40.74.ESI-MS:mass calcd for[M+H]+(C28H28N2O4)457.20;found m/z,457.21.
Synthesis of compound 22: prepared according to method B above from compound 6a and (4-methoxyphenyl) methylamine. Yield 55.61%, brown solid.1H NMR(400MHz,CDCl3)δ7.35(d,J=8.5Hz,2H),7.26(s,2H),7.20(t,J=8.1Hz,1H),7.10(d,J=7.7Hz,2H),6.95(t,J=7.3Hz,1H),6.88–6.83(m,3H),6.66(t,J=2.1Hz,1H),6.53(dd,J=8.0,1.9Hz,1H),6.20(s,2H),5.98(s,1H),4.07(s,2H),3.91(s,2H),3.77(s,3H),3.73(s,6H).13C NMR(101MHz,CDCl3)δ159.85,159.65,157.58,145.19,142.35,131.06,130.41,129.39,124.88,121.68,118.60,114.11,112.13,110.86,107.53,95.06,55.90,55.29,49.65,39.00.ESI-MS:mass calcd for[M+H]+(C29H30N2O4)471.22;found m/z,471.22.
Synthesis of compound 23: prepared according to the above method B from compound 6a and p-toluidine. Yield 52.72%, brown solid.1H NMR(400MHz,CDCl3)δ7.21–7.18(m,2H),7.15(dd,J=10.1,3.5Hz,3H),7.11(t,J=8.1Hz,1H),7.04(d,J=7.9Hz,2H),7.01(dd,J=8.5,1.0Hz,2H),6.86(dd,J=10.5,4.2Hz,1H),6.72(dd,J=8.1,1.5Hz,1H),6.64(t,J=2.2Hz,1H),6.48–6.44(m,1H),6.17(s,2H),5.74(s,1H),3.80(s,2H),3.68(s,2H),3.65(s,6H),2.25(s,3H).13C NMR(101MHz,CDCl3)δ159.49,158.27,157.70,145.00,142.43,136.62,136.36,130.31,129.38,129.00,128.39,121.61,118.52,111.97,110.55,110.29,107.28,95.50,55.75,52.35,40.73,21.11.ESI-MS:mass calcd for[M+H]+(C29H30N2O3)455.23;found m/z,455.24.
Synthesis of compound 24: according to the above process B, starting from compound 6a and benzo [ d ]][1,3]Dioxazol-5-ylmethylamine. Yield 58.36%, brown solid.1H NMR(400MHz,CDCl3)δ7.28(d,J=8.5Hz,2H),7.20(t,J=8.1Hz,1H),7.10(d,J=7.6Hz,2H),6.99(s,1H),6.95(t,J=7.4Hz,1H),6.90(d,J=7.9Hz,1H),6.84(dd,J=8.1,1.6Hz,1H),6.74(d,J=7.9Hz,1H),6.67(t,J=2.2Hz,1H),6.52(dd,J=8.0,1.7Hz,1H),6.19(s,2H),5.92(s,2H),4.09(s,2H),3.90(s,2H),3.75(s,6H).13C NMR(101MHz,CDCl3)δ160.08,159.67,157.35,148.17,147.94,145.21,142.31,130.44,129.41,125.31,123.86,121.74,118.64,112.32,111.01,110.18,108.43,107.69,102.21,101.31,94.87,55.95,49.76,38.85.ESI-MS:mass calcd for[M+H]+(C29H28N2O5)485.20;found m/z,485.20.
Synthesis of compound 25: prepared according to method B above from compound 6a and (R) -1-phenylethane-1-amine. Yield 51.83%, brown solid.1H NMR(400MHz,CDCl3)δ7.51–7.45(m,2H),7.32(ddd,J=9.4,6.6,1.9Hz,4H),7.24(t,J=1.9Hz,1H),7.17(t,J=8.1Hz,1H),7.09(dd,J=8.5,1.0Hz,2H),6.95(t,J=7.3Hz,1H),6.81(dd,J=8.1,1.5Hz,1H),6.67(t,J=2.2Hz,1H),6.50(dd,J=8.1,1.6Hz,1H),6.16(s,2H),5.92(s,1H),3.98(d,J=6.7Hz,1H),3.97–3.92(m,2H),3.71(s,6H),1.64(d,J=6.8Hz,3H).13C NMR(101MHz,CDCl3)δ159.64,159.27,157.75,145.10,142.40,130.33,129.39,128.70,128.30,127.57,121.62,118.54,112.10,110.75,107.49,95.04,57.98,55.80,38.52,21.10.ESI-MS:mass calcd for[M+H]+(C29H30N2O3)455.23;found m/z,455.23.
Synthesis of compound 26: prepared according to method B above from compound 6a and (R) -1- (4-methoxyphenyl) ethan-1-amine. Yield 50.07% brown solid.1H NMR(400MHz,CDCl3)δ7.30–7.25(m,4H),7.18(t,J=8.1Hz,1H),7.09(d,J=7.7Hz,2H),6.94(t,J=7.3Hz,1H),6.86(d,J=8.6Hz,2H),6.79(dd,J=8.1,1.4Hz,1H),6.69(t,J=2.1Hz,1H),6.53(dd,J=8.1,1.6Hz,1H),6.23(s,2H),5.79(s,1H),3.80(s,3H),3.71(s,6H),3.69(s,2H),1.34(d,J=6.6Hz,3H).13C NMR(101MHz,CDCl3)δ159.40,158.47,158.46,157.24,144.94,142.46,130.28,129.38,127.86,121.59,118.49,113.57,111.81,110.47,107.16,95.65,56.83,55.68,55.28,39.50,24.26.ESI-MS:mass calcd for[M+H]+(C30H32N2O4)485.24;found m/z,485.25.
Synthesis of compound 27: prepared according to method B above from compound 6a and 2-phenylethylamine. Yield 61.09% brown solid.1H NMR(400MHz,CDCl3)δ7.28(d,J=4.6Hz,3H),7.22(dd,J=7.6,2.9Hz,2H),7.17–7.12(m,2H),7.09(d,J=7.6Hz,2H),6.96(t,J=7.4Hz,1H),6.83(dd,J=8.1,1.5Hz,1H),6.71(t,J=2.2Hz,1H),6.53(dd,J=8.1,1.7Hz,1H),6.18(s,2H),5.85(s,1H),4.19(s,2H),3.65(s,6H),3.11(d,J=6.2Hz,2H),3.06(d,J=6.2Hz,2H).13C NMR(101MHz,CDCl3)δ159.92,159.62,157.36,145.21,142.24,136.87,130.48,129.42,128.92,128.90,127.05,121.83,118.69,112.52,110.96,107.59,94.94,55.88,47.27,40.17,32.62.ESI-MS:mass calcd for[M+H]+(C29H30N2O3)455.23;found m/z,455.24.
Synthesis of compound 28: prepared according to method B above from compound 6a and 2- (4-chlorophenyl) ethane-1-amine. Yield 52.36%, brown solid.1H NMR(400MHz,CDCl3)δ7.29(s,2H),7.25(d,J=2.5Hz,2H),7.21(d,J=8.1Hz,1H),7.09(dd,J=7.5,5.3Hz,4H),6.96(t,J=7.3Hz,1H),6.83(dd,J=8.1,2.1Hz,1H),6.71(t,J=2.1Hz,1H),6.53(dd,J=8.1,2.2Hz,1H),6.19(s,2H),5.86(s,1H),4.17(s,2H),3.67(s,6H),3.07(d,J=6.4Hz,2H),3.02(d,J=6.4Hz,2H).13C NMR(101MHz,CDCl3)δ160.43,159.67,157.11,145.27,142.19,134.94,133.15,130.55,130.30,129.44,129.10,121.89,112.72,111.03,107.69,94.81,55.95,46.78,40.05,31.53.ESI-MS:mass calcd for[M+H]+(C29H29ClN2O3)489.19;found m/z,489.20.
Synthesis of compound 29: according to the above formulaMethod B, prepared from compound 6a and 2- (4-fluorophenyl) ethan-1-amine. Yield 48.28%, yellow solid.1H NMR(400MHz,CDCl3)δ7.25(t,J=7.9Hz,2H),7.20–7.06(m,5H),6.99–6.90(m,3H),6.77(dd,J=8.0,1.5Hz,1H),6.70(t,J=2.0Hz,1H),6.52(dd,J=8.1,1.7Hz,1H),6.23(s,2H),5.81(s,1H),3.81(s,2H),3.67(s,6H),2.80(dd,J=11.0,5.3Hz,4H).13C NMR(101MHz,CDCl3)δ162.60,160.18,159.34,158.41,157.28,144.98,142.46,136.02,135.99,130.30,130.14,130.07,129.38,121.59,118.49,115.12,114.92,111.94,111.39,110.42,107.17,95.56,55.66,50.15,41.15,35.31.ESI-MS:mass calcd for[M+H]+(C29H29FN2O3)473.22;found m/z,473.23.
Synthesis of compound 30: prepared according to method B above from compound 6a and 2- (p-tolyl) ethan-1-amine. Yield 55.37%, yellow oil.1H NMR(400MHz,CDCl3)1H NMR(400MHz,CDCl3)δ7.26(s,1H),7.23(d,J=5.4Hz,1H),7.17(t,J=8.1Hz,1H),7.10–7.03(m,6H),6.93(t,J=7.3Hz,1H),6.77(dd,J=8.0,1.6Hz,1H),6.70(t,J=2.2Hz,1H),6.52(dd,J=8.1,1.8Hz,1H),6.22(s,2H),5.89(s,1H),3.86(s,2H),3.65(s,6H),2.83(dd,J=10.6,5.3Hz,4H),2.29(s,3H).13C NMR(101MHz,CDCl3)δ159.43,158.29,157.67,145.06,142.48,136.73,135.53,130.31,129.38,129.09,128.68,121.58,118.52,111.98,110.47,110.08,107.24,95.47,55.68,49.79,40.94,35.14,21.02.ESI-MS:mass calcd for[M+H]+(C30H32N2O3)469.24;found m/z,469.25.
Synthesis of compound 31: prepared according to method B above from compound 6a and 4- (2-aminoethyl) phenol. Yield 51.03%, brown oil.1H NMR(400MHz,MeOD)δ7.21(td,J=8.2,4.6Hz,3H),7.11–7.04(m,4H),6.89–6.84(m,2H),6.77(s,1H),6.74(dd,J=4.4,2.2Hz,2H),6.51–6.47(m,1H),6.36(s,2H),4.21(s,2H),3.79(s,6H),3.17–3.12(m,2H),2.94–2.87(m,2H).13C NMR(101MHz,CDCl3)δ165.12,163.81,160.91,160.36,150.01,146.90,134.04,133.31,132.73,130.79,124.57,121.86,119.31,116.20,114.04,110.78,105.23,98.17,59.12,43.41,34.69.ESI-MS:mass calcd for[M+H]+(C29H30N2O4)471.22;found m/z,471.23.
Synthesis of compound 32: prepared according to method B above from compound 6a and 2- (4-methoxyphenyl) ethan-1-amine. Yield 58.95%, brown solid.1H NMR(400MHz,CDCl3)δ7.25(t,J=7.9Hz,2H),7.18(t,J=8.1Hz,1H),7.08(dd,J=8.5,2.1Hz,4H),6.94(t,J=7.3Hz,1H),6.85–6.76(m,3H),6.70(d,J=1.9Hz,1H),6.52(dd,J=8.1,2.1Hz,1H),6.22(s,2H),5.85(s,1H),3.84(s,2H),3.77(s,3H),3.67(s,6H),2.83(d,J=5.8Hz,2H),2.79(d,J=6.0Hz,2H).13C NMR(101MHz,CDCl3)δ160.25,159.73,158.67,157.23,145.26,142.26,130.48,129.92,129.41,128.46,121.78,118.68,114.29,112.52,111.00,107.67,101.40,94.81,55.91,55.29,47.02,39.72,31.23.ESI-MS:mass calcd for[M+H]+(C30H32N2O4)485.24;found m/z,484.25.
Synthesis of compound 33: prepared according to method B above from compound 6a and 2- (2-methoxyphenyl) ethan-1-amine. Yield 47.64%, brown solid.1H NMR(400MHz,CDCl3)δ7.25(t,J=7.9Hz,2H),7.20–7.11(m,3H),7.08(d,J=7.6Hz,2H),6.94(t,J=7.3Hz,1H),6.88–6.76(m,3H),6.70(t,J=2.1Hz,1H),6.51(dd,J=8.1,1.7Hz,1H),6.22(s,2H),5.83(s,1H),3.87(s,2H),3.75(s,3H),3.67(s,6H),2.87(s,4H).13C NMR(101MHz,CDCl3)δ159.40,158.37,157.68,157.45,145.01,142.45,130.42,130.29,129.38,128.29,127.38,121.59,120.40,118.52,111.92,110.40,110.38,107.17,95.52,55.70,55.26,48.33,41.06,30.36.ESI-MS:mass calcd for[M+H]+(C30H32N2O4)485.24;found m/z,484.25.
Synthesis of compound 34: prepared according to method B above from compound 6a and 2- (3-methoxyphenyl) ethan-1-amine. Yield 52.11%, brown solid.1H NMR(400MHz,CDCl3)δ7.24(t,J=7.9Hz,2H),7.17(t,J=8.0Hz,2H),7.07(d,J=7.7Hz,2H),6.93(t,J=7.3Hz,1H),6.81–6.67(m,5H),6.52(dd,J=8.1,1.8Hz,1H),6.22(s,2H),5.87(s,1H),3.83(s,2H),3.76(s,3H),3.66(s,6H),2.83(dd,J=15.7,5.8Hz,4H).13C NMR(101MHz,CDCl3)δ159.68,159.38,158.42,157.33,145.01,142.49,141.90,130.29,129.38,129.31,121.55,121.22,118.48,114.43,111.87,111.42,111.17,110.41,107.16,95.56,55.66,55.14,49.87,41.13,36.07.ESI-MS:mass calcd for[M+H]+(C30H32N2O4)485.24;found m/z,484.25.
Synthesis of compound 35: prepared according to method B above from compound 6a and 3- (4-methoxyphenyl) propan-1-amine. Yield 55.61%, brown oil.1H NMR(400MHz,CDCl3)δ7.28–7.22(m,2H),7.18(t,J=8.1Hz,1H),7.11–7.05(m,4H),6.93(t,J=7.3Hz,1H),6.83–6.77(m,3H),6.70(t,J=2.2Hz,1H),6.56–6.51(m,1H),6.24(s,2H),5.87(s,1H),3.83(s,2H),3.76(s,3H),3.73(s,6H),2.60(dt,J=24.7,7.5Hz,4H),1.88–1.77(m,2H).13C NMR(101MHz,CDCl3)δ159.47,158.32,157.72,157.59,145.03,142.49,134.26,130.31,129.38,129.29,121.56,118.50,113.75,111.94,110.60,110.49,107.26,95.52,55.77,55.27,48.19,40.99,32.66,31.33.ESI-MS:mass calcd for[M+H]+(C31H34N2O4)499.25;found m/z,499.26.
Synthesis of compound 36: prepared according to method B above from compound 6B and 2- (4-methoxyphenyl) ethan-1-amine. Yield 50.09%, brown solid.1H NMR(400MHz,CDCl3)δ7.24(dd,J=11.2,4.6Hz,2H),7.16(t,J=8.1Hz,1H),7.12(d,J=8.1Hz,1H),7.10–7.04(m,4H),6.92(t,J=7.3Hz,1H),6.84–6.79(m,2H),6.79–6.75(m,1H),6.69(t,J=2.2Hz,1H),6.56(d,J=2.2Hz,1H),6.51(ddd,J=7.6,5.6,2.0Hz,2H),5.83(s,1H),3.76(s,5H),3.67(s,3H),2.84(dd,J=10.9,4.2Hz,2H),2.77(dd,J=10.7,3.9Hz,2H).13C NMR(101MHz,CDCl3)δ158.71,158.44,158.06,157.29,144.99,142.49,131.96,130.64,130.31,129.69,129.38,122.74,121.54,118.50,113.90,111.96,110.61,110.32,107.37,102.46,55.34,55.29,50.33,48.72,35.08.ESI-MS:mass calcd for[M+H]+(C29H30N2O3)455.23;found m/z,455.23.
Synthesis of compound 37: prepared according to method B above from compound 6c and 2- (4-methoxyphenyl) ethan-1-amine. Yield 55.62%, brown oil.1H NMR(400MHz,CDCl3)δ7.24–7.20(m,2H),7.12(t,J=8.2Hz,3H),7.06–7.02(m,2H),6.96–6.88(m,3H),6.85–6.78(m,3H),6.71(dd,J=8.0,1.5Hz,1H),6.64(t,J=2.2Hz,1H),6.45(dd,J=7.9,2.0Hz,1H),5.76(s,1H),3.80(s,3H),3.76(s,5H),2.88(t,J=6.8Hz,2H),2.78(t,J=6.9Hz,2H).13C NMR(101MHz,CDCl3)δ159.21,158.13,151.46,144.70,143.78,142.69,136.86,131.86,130.09,129.68,129.32,121.28,121.08,120.63,118.30,113.98,112.67,111.45,109.28,106.14,56.02,55.29,53.54,50.62,35.21.ESI-MS:mass calcd for[M+H]+(C29H30N2O3)455.23;found m/z,455.24.
Synthesis of compound 38: prepared according to method B above from compound 6d and 2- (4-methoxyphenyl) ethan-1-amine. Yield 52.36%, brown oil.1H NMR(400MHz,CDCl3)δ7.25–7.19(m,4H),7.14(t,J=8.1Hz,1H),7.09(d,J=8.6Hz,2H),7.03(dd,J=8.5,1.0Hz,2H),6.98–6.93(m,2H),6.90(dd,J=10.5,4.2Hz,1H),6.84–6.79(m,2H),6.76–6.72(m,1H),6.67(t,J=2.2Hz,1H),6.51–6.47(m,1H),5.79(s,1H),3.74(s,3H),3.73(s,2H),2.85(dd,J=10.8,3.8Hz,2H),2.74(t,J=7.0Hz,2H).13C NMR(101MHz,CDCl3)δ158.64,158.12,156.01,145.01,142.56,135.30,132.06,130.35,129.71,129.54,129.42,121.53,119.11,118.52,114.01,111.96,110.70,107.49,55.31,53.32,50.76,35.41.ESI-MS:mass calcd for[M+H]+(C28H28N2O2)425.22;found m/z,425.23.
The synthetic scheme for compounds 42-45 is as follows:
Figure BDA0002376249260000191
reagents and conditions: 3-aminophenol, bromobenzene, t-BuONa, chlorine (2-dicyclohexylphosphino-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2-aminoethylphenyl)]Palladium (II) (Brettphos Palladicycle), 1, 4-dioxane, 90 ℃,2 h. Yield 82.37%; (ii) 4- (phenylamino) phenol, K2CO3, DMSO, 100 ℃, 3 h. Yield 76.55%; (iii) NaBH (AcO)3AcOH, DMF, 80 ℃ overnight. The yield is 47% -56%.
Synthesis of compound 40: prepared from 4-aminophenol and bromobenzene in the same procedure as compound 5. Yield 82.37%, brown solid.1H NMR(400MHz,CDCl3)δ7.24–7.18(m,2H),7.04–6.99(m,2H),6.93–6.86(m,2H),6.83(t,J=7.3Hz,1H),6.80–6.74(m,2H),5.46(s,1H),4.64(s,1H).ESI-MS:mass calcd for[M+H]+(C12H11NO)186.08;found m/z,186.09.
Synthesis of compound 41: prepared from the compound 39 and 4-fluoro-2, 6-dimethoxybenzaldehyde by the method A. Yield 76.55%, colorless oil.1H NMR(400MHz,CDCl3)δ10.32(s,1H),7.26–7.21(m,2H),7.07–7.02(m,4H),6.96(d,J=8.8Hz,2H),6.93(t,J=7.4Hz,1H),6.12(s,2H),3.72(s,6H).ESI-MS:mass calcd for[M+H]+(C21H19NO4)350.13;found m/z,350.14.
Synthesis of compound 42: prepared from compound 41 and 2- (4-methoxyphenyl) ethan-1-amine by the method B. Yield 56.35%, brown oil.1H NMR(400MHz,CDCl3)δ7.25(t,J=7.8Hz,2H),7.08(t,J=8.3Hz,4H),7.01(d,J=8.1Hz,2H),6.95(d,J=8.8Hz,2H),6.90(t,J=7.3Hz,1H),6.81(d,J=8.5Hz,2H),6.17(s,2H),5.68(s,1H),3.84(s,2H),3.77(s,3H),3.66(s,6H),2.81(dd,J=13.1,5.6Hz,4H).13C NMR(101MHz,CDCl3)δ159.38,158.75,157.99,150.95,143.82,138.93,132.05,129.69,129.40,120.63,120.28,120.09,116.99,113.82,109.89,94.39,55.66,55.28,49.97,41.03,34.86.ESI-MS:mass calcd for[M+H]+(C30H32N2O4)485.24;found m/z,485.25.
Synthesis of compound 43: prepared from compound 41 and 2- (3-methoxyphenyl) ethan-1-amine by the method B. Yield 51.21%, brown oil.1H NMR(400MHz,CDCl3)δ7.28–7.25(m,1H),7.24(dd,J=5.7,3.8Hz,1H),7.21–7.16(m,1H),7.09–7.05(m,2H),7.01(dd,J=8.6,1.0Hz,2H),6.98–6.93(m,2H),6.90(t,J=7.3Hz,1H),6.79–6.71(m,3H),6.17(s,2H),5.66(s,1H),3.85(s,2H),3.77(s,3H),3.65(d,J=7.9Hz,6H),2.85(dd,J=12.5,5.6Hz,4H).13C NMR(101MHz,CDCl3)δ161.02,159.93,159.71,149.71,143.49,139.68,138.95,129.77,129.42,121.13,120.95,120.90,119.69,117.32,114.23,112.53,93.70,55.82,55.22,47.29,40.00,32.87.ESI-MS:mass calcd for[M+H]+(C30H32N2O4)485.24;found m/z,485.25.
Synthesis of compound 44: prepared from compound 41 and 2- (2-methoxyphenyl) ethan-1-amine by the method B. Yield 47.38%, brown oil.1H NMR(400MHz,CDCl3)δ7.19(d,J=7.2Hz,1H),7.16(s,1H),7.12–7.05(m,2H),7.02–6.98(m,2H),6.94(dd,J=8.5,0.9Hz,2H),6.90–6.85(m,2H),6.84(d,J=7.3Hz,1H),6.80–6.77(m,1H),6.75(d,J=8.2Hz,1H),6.10(s,2H),5.60(s,1H),3.79(s,2H),3.70(s,3H),3.59(s,6H),2.79(s,4H).13C NMR(101MHz,CDCl3)δ160.92,159.60,157.50,149.69,143.45,139.75,131.18,129.42,128.49,125.30,120.93,120.86,120.83,119.65,117.36,110.48,93.75,55.82,55.20,45.68,40.21,28.44.ESI-MS:mass calcd for[M+H]+(C30H32N2O4)485.24;found m/z,485.25.
Synthesis of compound 45: prepared from compound 41 and 3- (4-methoxyphenyl) propan-1-amine using method B described above. Yield 53.13%, brown oil.1H NMR(400MHz,CDCl3)δ7.28(s,1H),7.24(s,1H),7.10–7.06(m,3H),7.05–7.01(m,3H),6.98–6.94(m,2H),6.90(dd,J=11.6,4.1Hz,1H),6.81–6.77(m,2H),6.18(s,2H),5.69(s,1H),3.94(s,2H),3.76(s,3H),3.73(s,6H),2.71–2.66(m,2H),2.59–2.54(m,2H),1.97–1.90(m,2H).13C NMR(101MHz,CDCl3)δ160.89,159.85,157.92,149.86,143.53,139.59,132.78,129.42,129.29,120.90,120.87,119.76,117.28,113.83,93.71,55.85,55.24,45.80,39.55,32.16,28.18.ESI-MS:mass calcd for[M+H]+(C31H34N2O4)499.25;found m/z,499.26.
EXAMPLE 2 inhibitory Activity of Compounds on tumor cells
The tumor cell lines used were human colorectal cancer cell lines including HT29, HCT15, HCT116, DLD-1 and SW620, which were purchased from ATCC, USA.
The MTT method was used to evaluate the tumor cell inhibitory activity. The cells are treated at 2-4X 103Cells were seeded in 96-well plates at a cell density of 0.1 mL/well. After 24h incubation compound was added and after 72 h treatment 20. mu.L MTT solution (5mg/mL) was added to each well and incubated at 37 ℃ for a further 2-4 h. Discarding the supernatant, and returning toAnd adding 150 mu LDMSO into each hole, and fully shaking to dissolve for 10-15 min. The relative inhibition of tumor cell proliferation was calculated for each target compound by measuring the absorbance (OD) of 96-well plates at 570nm using a Spectra MAX M5 microplate spectrophotometer (Molecular Devices, CA, USA) and calculating the IC50 value from the software. The results are shown in Table 1. IC50 values are the average of at least three independent experiments and were calculated by GraphPad Prism5 software.
TABLE 1
Figure BDA0002376249260000211
Figure BDA0002376249260000221
As can be seen from Table 1, compounds 7 to 38 and compounds 42 to 45 of the present invention can inhibit the proliferation of colorectal cancer cells, wherein the inhibitory activity of compound 42 is the best.
FIG. 2 shows the effect of compound 42 of the present invention on the viability of tumor cells (CRC cells) and normal hepatocytes LO 2. Wherein A is the IC50 value of the compound 42 to HT29, HCT15, HCT116 and DLD-1, and B is the toxicity of the compound 42, 5-fluorouracil and cisplatin to LO 2. Data were obtained from triplicate experiments. Relative to DMSO, P <0.01, P < 0.001.
The point of attachment of region ii to region i was changed to C-4 and the resulting compound 42 had an antitumor activity against HT29 with an IC50 value of 0.32 μ M (as shown in a in fig. 2), 3-fold higher than compound 30, and 31-fold higher than the positive drug (cisplatin: IC50 ═ 10.75 μ M, 5-fluorouracil: IC50 ═ 13.37 μ M). The toxicity of compound 42 to a human normal liver cell line (LO2) was analyzed by MTT assay, and the results are shown in B in FIG. 2, where the cytotoxicity of compound 42 is significantly lower than that of the positive drugs (cisplatin and 5-fluorouracil), demonstrating that compound 42 has good safety in vitro.
EXAMPLE 3 interaction of Compounds with c-Myc/MAX proteins
The electrophoretic mobility assay (EMSA) was used to analyze the interaction of compounds with c-Myc/MAX proteins as follows:
1. purification of c-Myc and Max
The human c-Myc gene (residue 353-439) containing the b-HLH-Zip domain and Max (151 amino acid residues) were expressed in recombinant E.coli BL21DE3 by a backbone vector pET151D of N-terminal hexahistidine (His 6) tagged protein. Cultivation of bacteria and purification of proteins the bacteria were cultivated overnight at 37 ℃ and 225rpm for 20h using the methods known per se, and the proteins were expressed in 0.8mM isopropyl-L-thio-B-D-galactopyranoside (IPTG, Sigma). Then, the culture was harvested and buffered (8M urea, 100mM NaH)2PO4And 10mM Tris), purified by NTA-Ni-agarose chromatography column (Qiagen, inc. chatsworth) and dialyzed in storage buffer (TrisHCl 50mM, pH 6.5, 150mM NaCl and 30% glycerol). The protein was quantified by using Nanodrop and used for electrophoretic mobility assay (EMSA).
2. Electrophoretic mobility determination
Double-stranded DNA biotinylated oligonucleotides (5-CACCCGGTCACGTGGCCTACAC-3, 50nM) that formed a binding site for the c-Myc/Max dimer and purified protein complexes (Myc/Max complexes, 50nM) were prepared as reported in the literature. The binding reaction buffer contained 10mM EDTA, 1 XPBS (pH 7.0), 500mM KCl, 30mM MgCl 25% glycerol, 0.1% NP40, 1.5mM DTT. 10% DMSO. Compounds were tested dissolved in DMSO as a control. Protein interaction complexes and 20. mu.M compound were prepared at room temperature for 1.5h, then DNA oligo was added for 0.5h, samples were loaded into 8% SDS gels in 10% glycerol, 0.5TBE solution, Image J was detected by Bio-Rad and data was analyzed. The results are shown in Table 2. All data in table 2 are the average of three trials.
TABLE 2
Compound numbering Relative inhibition ratio (%)a Compound numbering Relative inhibition ratio (%)a
7 2.5 26 36.1
8 1.7 27 45.1
9 N/A 28 43.7
10 3.2 29 40.5
11 6.1 30 46.2
12 10.2 31 51.7
13 N/A 32 75.1
14 8.9 33 67.4
15 12.6 34 60.8
16 N/A 35 63.4
17 26.4 36 42.1
18 21.2 37 42.3
19 25.7 38 35.6
20 29.3 42 83.2
21 31.8 43 65.2
22 48.7 44 57.3
23 32.4 45 70.6
24 42.1 DMSO 0
25 30.5
aRelative inhibition calculated by the ratio of experimental to control (DMSO).
N/A was not determined.
As can be seen from Table 2, the compounds of the present invention have different binding inhibition rates with c-Myc/MAX protein, and among these molecules, compound 42 is most effective, indicating that compound 42 has excellent strong destructive activity of the c-Myc-Max/DNA complex. The results correspond to an in vitro antiproliferative activity.
The following focuses on the possible binding patterns of compound 42 to c-Myc/MAX. The protein crystal structure of Myc/Max (PDB ID: 1NKP) was used. As shown in FIG. 3, the predicted binding pattern and detailed interaction of compound 42 with Myc/Max are illustrated. Compound 42 can bind tightly to the basic/helix-loop-helix/leucine zipper (bHLHZ) domain of Myc-Max. It is clearly seen that NH and Ala237 of the N-phenylaniline (domain I) form hydrogen bonds and that N-phenylaniline interacts with ILe242, arg226, His223 respectively by hydrophobic character. In addition, the two methoxy groups of region II interact with Arg913 through hydrogen bonds. More importantly, NH and Arg913 of region II form an additional hydrogen bond. Furthermore, the phenyl groups of the iii-region in Lys939, Lys918, Arg914 form hydrophobic interactions, respectively. Furthermore, the entire molecule is located at the Myc-Max dimer interface and the protein-DNA interface, indicating that the stability of the binding of the molecule to the Myc-Max protein may disrupt the formation of the c-Myc/Max/DNA complex. All of the characteristics of the molecular binding pattern are predictive of the superior antitumor activity of compound 42.
EXAMPLE 4 cell clonogenic assay for Compound 42
To test the viability of compound 42-treated cells, HT29 cells and HCT15 cells (500 cells/well) were plated in 6-well plates and incubated overnight at 37 ℃, followed by addition of the indicated dose of compound 42(0-5.0 μ M), with DMSO as control) and cultured with fresh medium for 15 days. Cell culture was terminated when colonies of cell formation were observed, then the supernatant was removed and washed twice with PBS buffer solution, then fixed with 4% paraformaldehyde for 15 minutes before the solution was discarded. Colonies were fixed with 4% paraformaldehyde and stained with 0.5% crystal violet solution for 15 minutes, then the crystal violet solution was removed and the staining solution was washed with PBS buffer solution. The results are shown in fig. 4, with the data in fig. 4 being from three separate experiments. The number of colonies in the treated culture was expressed as a percentage of the control culture. As can be seen, the colony formation rate of HCT15 cells and HT29 cells was significantly reduced in the experimental group. As shown in FIG. 4, when the concentration of 42 was increased to 1.25. mu.M, almost no colony formation was observed in HT29 cells. Similarly, when the concentration of 42 in HCT15 cells reached 2.5. mu.M, cell colony formation was almost completely inhibited. The results of plate colony formation correspond to the results of the MTT assay.
EXAMPLE 5 cell cycle distribution and apoptosis Rate induced by Compound 42
The effect of compound 42 on cell cycle distribution and apoptosis rate was studied using Flow Cytometry (FCM). HT29 cells and HCT15 cells at 3X 10 per well5The amount of cells was seeded in 6-well plates and incubated for 24 hours, followed by treatment with compound 42 for 24 hours, respectively. Cells were collected and fixed with ice-cold 70% ethanol at 4 ℃ for 12 hours, then ethanol was removed and cells were washed with cold PBS. Thereafter, cells were detached from the plate using trypsinization, and collected by centrifugation at 1000rpm for 5 min. The cells were then resuspended in binding buffer and stained by cell cycle detection kit (keygenetec, KGA512) and annexin V-FITC/PI dual label (keygenetec, KGA 107). The resulting samples were analyzed using a flow cytometer (ACEA NovoCyte Advanteon, Ason biosciences, USA). The results are shown in FIG. 5.
FIG. 5 shows the results of flow cytometry analysis of Compound 42, wherein A is the cell cycle distribution analysis of HT29 and HCT15 cells treated with different concentrations of Compound 42 for 24 h; b is treating cells with compound 42 at a concentration for 24h and assessing levels of apoptosis by FITC-Annexin V/PI and analyzing apoptotic cells by flow cytometry. All data were from three parallel experiments, P <0.05 versus DMSO, P <0.01 versus DMSO; p <0.001 compared to DMSO.
As shown in a in figure 5, in both HT29 and HCT15 cells, the cell cycle progression affected by compound 42 was significantly arrested in the G0/G1 phase, and compound 42 clearly differentiated the cell cycle distribution in a dose-dependent manner. Inhibition of the cell cycle halted herein inhibited proliferation of these cells, reflecting their ability to disrupt DNA binding to the c-Myc-Max heterodimer.
As shown in B in FIG. 5, it was found that 42 compounds at different concentrations significantly affected apoptosis in HT29 and HCT15 in a dose-dependent manner. After HT29 and HCT15 cells are treated for 24 hours, the compound 42 with the concentration of 10 mu M can obviously cause apoptosis, and indicates that the inhibition capability of c-Myc on proliferating cells is strong.
EXAMPLE 6 Western blot analysis of Compound 42
Western blot was used to analyze the ability of compound 42 to inhibit c-Myc expression. The specific Western blot method comprises the following steps: cells were seeded overnight in 60 x 15mm petri dishes before treatment, then incubated with the indicated dose of compound 42 at 37 ℃ for 24 hours, then cell pellets were collected and resuspended in NP40 lysis buffer (Beyotime), and additional-proteasome inhibitor PMSF and phosphatase inhibitor cocktail (sigma) were added. Whole cell protein lysates were incubated on ice for 30min and centrifuged at 12000rpm and 4 ℃ for 20 min. The supernatant was assayed using the BCA ProteinAssay Kit (Solambio PC 0020). The samples were separated by SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) and transferred to polyvinylidene difluoride membranes (Millipore). The membrane was incubated with the primary antibody in 5% BSA/TBST buffer overnight at 4 ℃ with gentle shaking. After washing, the membranes were incubated with an appropriate peroxidase-conjugated secondary antibody (anti-IgG-HRP 1: 5000, CST) for 1 hour at room temperature, then washed again, and then the protein bands were visualized using an enhanced ECL immunoblot detection reagent (Millipore) and photographs taken using an imager (Abbkine). Antibody information: c-Myc antibody (1/1000 dilution, CST #9402), β -actin (1/1000 dilution, CST # 4970).
All experiments were independently repeated 3 times, and the results showed that compound 42 decreased the expression levels of c-Myc protein in HT29 cells and HCT15 cells. As shown in FIG. 6, it is clear that the down-regulation of c-Myc protein by compound 42 is essentially dose-dependent. The results indicate that compound 42 is likely to target c-Myc/Max in HT29 cells and HCT15 cells, thus presumably down-regulating its expression.
EXAMPLE 7 in vivo assay of Compound 42
Animal studies were conducted with approval from the institutional animal care committee of university of sichuan. Female BALB/c nude mice (Beijing HFK biosciences, Inc., Beijing, China) were purchased. HT29 cells (8X 10)6) Subcutaneously injected into 6-7 week old female BALB/c mice. Once the mean tumor volume increased to about 100mm3Randomly divide the mice into fiveAnd (5) each group. Compound 42(40mg/kg, 80mg/kg, 120mg/kg in 10% DMSO, 1% Tween-80 and saline), 5-fluorouracil (30mg/kg in 10% DMSO, 1% Tween-80 and saline) was administered by oral gavage (10% DMSO, 1% Tween-80 and saline). Tumor size and body weight were measured every two days. Tumor volume was measured with a vernier caliper and calculated as [0.5 × shortest diameter 2 × longest diameter]. The inhibition rate of tumor growth was calculated using the following formula: 100 x {1- [ final tumor volume in treatment group-initial tumor volume]/[ final tumor volume-initial tumor volume in vehicle-treated group]]}. The results are shown in FIG. 7.
FIG. 7 shows the results of an in vivo experiment of Compound 42 of the present invention in a HT29 tumor xenograft model. Wherein A is the growth inhibition of HT29 xenograft tumor in female BALB/c nude mice treated by compound 42; b is the average body weight of mice in each group during the 30 day treatment period; c is the tumor weight of each group of mice at the end of treatment. P <0.01 relative to DMSO; p <0.001 compared to DMSO.
As shown in fig. 7, compound 42 significantly inhibited tumor growth in a dose-dependent manner. In the HT29 xenograft model, Tumor Growth Inhibition (TGI) was observed at doses of 120mg/kg, 80mg/kg, 40mg/kg of 71.58%, 61.21%, 55.17%, respectively. 5-Fluorouracil was used as a positive control at a dose of 30mg/kg, and the body weight of mice was significantly reduced after 10 days of treatment. In contrast, treatment with 42 over 30 days showed excellent antitumor efficacy without causing significant weight loss and toxicity.
In conclusion, all the compounds of the present invention, especially compound 42, showed excellent antitumor activity against colon cancer tumor cells, and is expected to be used in the development of novel drugs for the treatment of CRC in the future.

Claims (12)

1. A phenoxy-N-phenylaniline derivative characterized in that: the structural formula is shown as formula II or formula III:
Figure FDA0002815714170000011
wherein R is1Is hydrogen or C1-C3 alkoxy;
R2is hydrogen or C1-C3 alkoxy;
R3is hydrogen or C1-C3 alkoxy;
R4is hydrogen or C1-C3 alkoxy;
R5is composed of
Figure FDA0002815714170000012
Figure FDA0002815714170000013
R6Is hydrogen, methyl, ethyl, hydroxyl, methoxy or halogen, n is any integer of 1-3, R7Is hydrogen or C1-C3 alkoxy.
2. The phenoxy-N-phenylaniline derivative according to claim 1, characterized in that: r1Is hydrogen or methoxy; r2Is hydrogen or methoxy; r3Is hydrogen or methoxy; r4Is hydrogen or methoxy.
3. The phenoxy-N-phenylaniline derivative according to claim 1 or 2, characterized in that: the R is5Is composed of
Figure FDA0002815714170000014
R6Is hydrogen, methyl, hydroxyl, methoxy, fluorine, chlorine or bromine.
4. A phenoxy-N-phenylaniline derivative according to claim 3 characterized in that: the R is5Is composed of
Figure FDA0002815714170000015
5. The phenoxy-N-phenylaniline derivative according to claim 1 or 2, characterized in that: the R is5Is composed of
Figure FDA0002815714170000016
R7Is hydrogen or methoxy.
6. The phenoxy-N-phenylaniline derivative of claim 1 having the formula any one of the following compound formulae:
Figure FDA0002815714170000021
7. use of a phenoxy-N-phenylaniline derivative according to any one of claims 1 to 6 in the manufacture of a medicament for the treatment of colorectal cancer.
8. Use of a phenoxy-N-phenylaniline derivative according to any one of claims 1 to 6 in the preparation of a c-Myc inhibitor.
9. A pharmaceutically acceptable salt of the phenoxy-N-phenylaniline derivative according to any one of claims 1 to 6.
10. Use of a pharmaceutically acceptable salt of a phenoxy-N-phenylaniline derivative according to claim 9 in the manufacture of a medicament for the treatment of colorectal cancer.
11. Use of a pharmaceutically acceptable salt of a phenoxy-N-phenylaniline derivative according to claim 9 in the preparation of a c-Myc inhibitor.
12. A pharmaceutical composition characterized by: the compound comprises an effective component and pharmaceutically acceptable auxiliary materials, wherein the effective component comprises a therapeutically effective amount of the phenoxy-N-phenylaniline derivative or pharmaceutically acceptable salt thereof according to any one of claims 1-6.
CN202010066925.1A 2020-01-20 2020-01-20 phenoxy-N-phenylaniline derivatives and application thereof Active CN111153817B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010066925.1A CN111153817B (en) 2020-01-20 2020-01-20 phenoxy-N-phenylaniline derivatives and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010066925.1A CN111153817B (en) 2020-01-20 2020-01-20 phenoxy-N-phenylaniline derivatives and application thereof

Publications (2)

Publication Number Publication Date
CN111153817A CN111153817A (en) 2020-05-15
CN111153817B true CN111153817B (en) 2021-02-02

Family

ID=70564667

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010066925.1A Active CN111153817B (en) 2020-01-20 2020-01-20 phenoxy-N-phenylaniline derivatives and application thereof

Country Status (1)

Country Link
CN (1) CN111153817B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107434779A (en) * 2011-12-21 2017-12-05 洛桑联邦理工学院(Epfl) Notch signal pathway inhibitors and its purposes in treatment of cancer
WO2017212414A1 (en) * 2016-06-08 2017-12-14 Università Degli Studi Del Piemonte Orientale "Amedeo Avogadro" Modulators of soce, compositions, and uses thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3485232D1 (en) * 1983-03-23 1991-12-12 Hitachi Ltd ANTHRACHINONE DYES AND LIQUID CRYSTALLINE CONTAINING THE SAME.
CN1506355A (en) * 2002-12-05 2004-06-23 中国医学科学院药物研究所 Diphenyl benzimidazole compound containing sulfonyl group and its prepn process, medicinal composition and use
EP1969930A1 (en) * 2007-03-12 2008-09-17 Bayer CropScience AG Phenoxy phenylamidines and their use as fungicides
CA2826034A1 (en) * 2011-01-28 2012-08-02 University Of Kentucky Research Foundation Stilbene analogs and methods of treating cancer
CN103772293B (en) * 2012-10-25 2015-09-09 中国中化股份有限公司 Fluorine-containing pyrimidines and purposes

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107434779A (en) * 2011-12-21 2017-12-05 洛桑联邦理工学院(Epfl) Notch signal pathway inhibitors and its purposes in treatment of cancer
WO2017212414A1 (en) * 2016-06-08 2017-12-14 Università Degli Studi Del Piemonte Orientale "Amedeo Avogadro" Modulators of soce, compositions, and uses thereof

Also Published As

Publication number Publication date
CN111153817A (en) 2020-05-15

Similar Documents

Publication Publication Date Title
CN108524482B (en) Use of 2- (substituted phenylamino) benzoic acid FTO inhibitors for treating leukemia
JP5047627B2 (en) Pyridinium and quinolinium derivatives
CN105968115B (en) Quinolines, preparation method, intermediate, pharmaceutical composition and application
TWI597062B (en) Nitrile derivatives and their pharmaceutical use and compositions
EP2784061B1 (en) Acyl-hydrazone and oxadiazole compounds, pharmaceutical compositions containing the same and uses thereof
KR101069735B1 (en) Benzophenone thiazole derivatives useful for inhibiting formation of microtubule and process for preparing the same
EP3388419A1 (en) Gli1 inhibitors and uses thereof
CN111153817B (en) phenoxy-N-phenylaniline derivatives and application thereof
Yang et al. Discovery of 4-(3, 5-dimethoxy-4-(((4-methoxyphenethyl) amino) methyl) phenoxy)-N-phenylaniline as a novel c-myc inhibitor against colorectal cancer in vitro and in vivo
CN104230912A (en) Quinoline derivative as well as preparation method and application thereof
WO2013000395A1 (en) Novel histone deacetylase inhibitor of benzamides and use thereof
JP2021527071A (en) Thieno [2,3-B] pyridine derivative as an EPAC inhibitor and its pharmaceutical use
Huang et al. Synthesis and biological evaluation of a novel c-Myc inhibitor against colorectal cancer via blocking c-Myc/Max heterodimerization and disturbing its DNA binding
US8883798B2 (en) Pyrimidine compounds inhibiting the formation of nitric oxide and prostaglandin E2, method of production thereof and use thereof
AU2018278283B2 (en) Pyridoquinazoline derivatives useful as protein kinase inhibitors
KR102709756B1 (en) Phenylene dibenzamide based compounds and composition for preventing and treating cancer diseases comprising the same as effective ingredient
CA3086099C (en) Mono- and di-amidine endo-exonuclease inhibitors and methods for inhibiting endo-exonuclease activity
WO2014000686A1 (en) Pyrimidobenzoazepine compound and use thereof as antitumour drug
CN111039940B (en) Aurora A kinase inhibitor, preparation method, pharmaceutical composition and application thereof
TWI415611B (en) Quinoline derivatives and uses of the same
CA3191223A1 (en) Methods of treating cancer
WO2022098808A1 (en) Therapeutic agents for treating hepatocellular carcinoma
CN111320557A (en) Semicarbazone compound and use thereof
CN103058927B (en) Quinoline and application thereof
CN117886750A (en) Azaphenanthrene compound with BRD4 protein inhibition effect, and 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
GR01 Patent grant
GR01 Patent grant