CN111793011A - Styrene sulfone NLRP3 inflammatory corpuscle inhibitor and preparation method and application thereof - Google Patents

Styrene sulfone NLRP3 inflammatory corpuscle inhibitor and preparation method and application thereof Download PDF

Info

Publication number
CN111793011A
CN111793011A CN202010794558.7A CN202010794558A CN111793011A CN 111793011 A CN111793011 A CN 111793011A CN 202010794558 A CN202010794558 A CN 202010794558A CN 111793011 A CN111793011 A CN 111793011A
Authority
CN
China
Prior art keywords
compound
polymorph
stereoisomer
prodrug
solvate
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.)
Granted
Application number
CN202010794558.7A
Other languages
Chinese (zh)
Other versions
CN111793011B (en
Inventor
吴敬德
张祥娜
徐阿娜
冉莹瑛
魏超
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.)
Shandong University
Original Assignee
Shandong 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 Shandong University filed Critical Shandong University
Priority to CN202010794558.7A priority Critical patent/CN111793011B/en
Publication of CN111793011A publication Critical patent/CN111793011A/en
Application granted granted Critical
Publication of CN111793011B publication Critical patent/CN111793011B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/44Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/14Radicals substituted by nitrogen atoms, not forming part of a nitro radical
    • 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

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General 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)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The disclosure relates to the field of styrene sulfone compounds and NLRP3 inhibitors, and particularly provides a styrene sulfone NLRP3 inflammatory corpuscle inhibitor and a preparation method and application thereof. The inhibitor is shown in a formula (1),
Figure DDA0002625071370000011
wherein n is selected from 0, 1; x is selected from N, O; r1Selected from different electron withdrawing or electron donating substituents; r2Selected from different aliphatic or aromatic substituents. Through verification, the compounds in the general formula have NLRP3 inhibitory activity.

Description

Styrene sulfone NLRP3 inflammatory corpuscle inhibitor and preparation method and application thereof
Technical Field
The disclosure relates to the field of styrene sulfone compounds and NLRP3 inflammatory corpuscle inhibitors, and particularly provides a styrene sulfone NLRP3 inflammatory corpuscle inhibitor and a preparation method and application thereof.
Background
The statements herein merely provide background information related to the present disclosure and may not necessarily constitute prior art.
Innate immunity (also called non-specific immunity) is the first line of defense against pathogens such as foreign viruses and bacteria or against internal damage in the body. The innate immune system can recognize dangerous molecules through pattern recognition receptors and mediate inflammatory responses to repair injury and preserve the balance of body tissues. The inflammasome, as a component of the innate immune system, plays a crucial role in immune regulation. Intracellular NOD-like receptors and AIM 2-like receptors are known to assemble into structurally common inflammatory bodies, of which NLRP3 is currently the clearest form of inflammatory body studied. It is a macromolecular complex of polyproteins consisting of the NLRP3 protein, connexin ASC and effector caspase-1, usually distributed among macrophages, dendritic cells, microglia and endothelial cells. The content of NLRP3 protein in normal cells is low, when cells are stimulated by Pathogen-associated molecular patterns (PAMPs) or Danger-associated molecular patterns (DAMPs), NLRP3 and gene expression of proinflammatory factors are up-regulated through NF-kB signaling pathway, NLRP3 protein is assembled into NLRP3 inflammatory corpuscle through recruitment of pro-caspase-1 by ASC after oligomerization, caspase-1 is converted from precursor form to active form, and release of the inflammatory factors is promoted and inflammatory scorching of cells is accompanied.
NLRP3 inflammasome activation is involved in the pathology of a variety of diseases with a severe inflammatory response. Cold porphyrin associated periodic syndrome is a hereditary immune inflammation characterized by fever, mainly due to the sustained activation of NLRP3 inflammasome; activation of NLRP3 inflammasome on microglia can mediate tau protein phosphorylation and other processes to aggravate alzheimer disease; there are also a number of associations between neuroinflammation-related diseases such as parkinson, multiple sclerosis and depression with NLRP3 inflammatory bodies; the NLRP3 small inflammatory body is reported to have good treatment effect on congenital immunity diseases such as gout, rheumatoid arthritis, inflammatory bowel disease and the like; the onset of metabolic inflammation such as obesity, type II diabetes, atherosclerosis, etc. is also closely related to NLRP3 inflammasome. NLRP3 inflammasome is a potential therapeutic target for the above diseases. Therefore, the reasonable design and synthesis of the compound with NLRP3 inflammatory body inhibitory activity has important value for treating inflammatory diseases.
Disclosure of Invention
The present disclosure provides a novel class of NLRP3 inflammasome inhibitors.
In one or some embodiments of the present disclosure, there is provided a compound represented by formula (1), a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof,
Figure BDA0002625071350000021
wherein n is selected from 0, 1; x is selected from N, O; r1Selected from different electron withdrawing or electron donating substituents; r2Selected from different aliphatic or aromatic substituents.
In one or more embodiments of the present disclosure, methods of making the above-described compounds, pharmaceutically acceptable salts, stereoisomers, isotopic labels, solvates, polymorphs, or prodrugs thereof, are provided, which synthetic routes are described in the detailed description.
In one or more embodiments of the present disclosure, there is provided a use of the compound, a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof, or a product obtained by the preparation method for preparing an NLRP3 inflammasome inhibitor.
In one or more embodiments of the present disclosure, there is provided a use of the compound, a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof, or a product prepared by the preparation method for preparing an anti-inflammatory drug.
In one or more embodiments of the present disclosure, there is provided an application of the compound, a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof, or a product prepared by the preparation method in preparing an anticancer drug.
In one or more embodiments of the present disclosure, a pharmaceutical composition is provided, which comprises the compound, a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof, or a product prepared by the preparation method.
One of the above technical solutions has the following advantages or beneficial effects:
1) the invention synthesizes 28 phenyl vinyl sulfone NLRP3 inflammatory corpuscle inhibitors, and all the compounds have the structure1HNMR,13The confirmation of CNMR and ESI-MS, the result of cytotoxicity evaluation shows that the cytotoxicity of the compound is greatly related to the structure of alpha, beta-unsaturated amide or alpha, beta-unsaturated ester, the cytotoxicity of the compound containing amide or ester is reduced compared with that of the compound containing cyano, but the toxicity of the amide compound is higher than that of the ester compound; r2The substituent has a large influence on cytotoxicity, when R2The cytotoxicity is obviously reduced when the compound is benzene sulfonamide. The primary activity evaluation result shows that the compound mostly containing an alpha, beta-unsaturated amide or alpha, beta-unsaturated ester structure shows better NLRP3 inflammatory corpuscle inhibitory activity; r1Substituents are useful for enhancing inhibitory activity; r2The substituent has great inhibitory activity on inflammasomeEffects, two of the compounds 7a and 5b showed comparable inhibitory activity to the lead compound BAY 11-7082, with a significant reduction in cytotoxicity; 7a still has better inhibitory activity in animals, therefore, the compounds disclosed by the disclosure can be used for preparing anti-inflammatory drugs.
2) In the evaluation of biological activity, the compound cytotoxicity and IL-1 beta inhibitory activity are firstly screened by MTT and ELISA experiments, the toxicity of most compounds is lower than that of BAY 11-7082, the cytotoxicity of compounds 7a and 5b is obviously reduced compared with that of a lead compound, the IL-1 beta inhibitory activity of the compounds 7a and 5b is respectively 1.83 mu M and 0.91 mu M, the inhibitory activity of 5b relative to BAY 11-7082 is improved, and the rationality of the design idea of the compound is preliminarily verified; the activity of 7a in mice is proved by an in vivo LPS-induced NLRP3 inflammasome model, and a basis is provided for the application of the compound in diseases.
Drawings
The accompanying drawings, which are incorporated in and constitute a part of this disclosure, illustrate embodiments of the disclosure and, together with the description, serve to explain the disclosure and not to limit the disclosure.
FIG. 1 shows the results of preliminary screening of partial compounds in example 3 for the evaluation of the activity of NLRP3 inflammasome.
FIG. 2 shows the results of the evaluation of the inhibitory activity of some compounds of example 4 on the in vivo NLRP3 inflammasome.
Detailed Description
The technical solutions in the embodiments of the present invention will be clearly and completely described below, and it is obvious that the described embodiments are only a part of the embodiments of the present invention, and not all embodiments. All other embodiments, which can be derived by a person skilled in the art from the embodiments of the present invention without making any creative effort, shall fall within the protection scope of the present invention.
In one or some embodiments of the present disclosure, there is provided a compound represented by formula (1), a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof,
Figure BDA0002625071350000031
wherein n is selected from 0, 1; x is selected from N, O; r1Selected from different electron withdrawing or electron donating substituents; r2Selected from different aliphatic or aromatic substituents.
Preferably, wherein X is an N or O atom; r1Is 4-methyl; 2-chloro group; 4-chloro group; 4-tert-butyl; one of 4-methoxy;
R2is that
Figure BDA0002625071350000041
Figure BDA0002625071350000042
One kind of (1).
In one or some embodiments of the present disclosure, there is provided a compound represented by formula (2) or formula (3), a pharmaceutically acceptable salt, stereoisomer, isotopic label, solvate, polymorph or prodrug thereof,
Figure BDA0002625071350000043
wherein R is1Selected from different electron withdrawing or electron donating substituents; r2Selected from different aliphatic or aromatic substituents, further preferably, R1Is 4-methyl; 2-chloro group; 4-chloro group; 4-tert-butyl; one of 4-methoxy; r2The same as claim 2.
Preferably, the compound, its pharmaceutically acceptable salt, stereoisomer, isotopic label, solvate, polymorph or prodrug thereof, wherein the compound is selected from the group consisting of:
Figure BDA0002625071350000051
further, the compound is one of 7a, 10a, 21a or 5b,
further, the compounds are 7a, 5 b.
In one or more embodiments of the present disclosure, there is provided a method for preparing the above compound, a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof, wherein the synthetic route is as follows:
Figure BDA0002625071350000061
preferably, wherein, a is the addition of Na2SO3、H2O, reacting for 3 to 6 hours at the temperature of between 50 and 100 ℃,
further preferably, the condition a comprises the addition of Na2SO3、H2O, reacting for 5 hours at the temperature of 80 ℃,
preferably, the b condition comprises the addition of NaOH, MeOH, H2O, reacting at the temperature of 20-50 ℃,
further preferably, the conditions b comprise adding NaOH, MeOH, H2O, reacting at 40 ℃,
preferably, the c condition comprises the addition of Na2SO3、Na2CO3、H2O, reacting for 1 to 5 hours,
further preferably, the c condition comprises addition of Na2SO3、Na2CO3、H2O, reacting for 4 hours,
preferably, the d condition comprises adding dibromopropionic acid and DMF, reacting for 10-15h at 60-90 ℃,
further preferably, the d condition comprises adding dibromopropionic acid and DMF, reacting for 12h at 80 ℃,
preferably, the e conditions comprise addition of amine, TBTU, TEA, anhydrous tetrahydrofuran,
preferably, the f conditions include the addition of alcohol, PPh3, DEAD, anhydrous tetrahydrofuran,
preferably, the g conditions include addition of MeOH, SOCl2
Preferably, the h condition comprises adding TEA and MeOH, reacting for 15-20h at 50-70 ℃,
further preferably, the h conditions comprise addition of TEA, MeOH, and reaction at 60 ℃ for 18 h.
In one or more embodiments of the present disclosure, there is provided a use of the compound, a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof, or a product obtained by the preparation method for preparing an NLRP3 inflammasome inhibitor.
In one or more embodiments of the present disclosure, there is provided a use of the compound, a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof, or a product prepared by the preparation method for preparing an anti-inflammatory drug.
In one or more embodiments of the present disclosure, there is provided an application of the compound, a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof, or a product prepared by the preparation method in preparing an anticancer drug.
In one or more embodiments of the present disclosure, a pharmaceutical composition is provided, which comprises the compound, a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof, or a product prepared by the preparation method.
Example 1
This example provides methods for the synthesis of compounds 2a-28a, 5B-8B, wherein the synthetic pathways for series a and B are as described above, and in particular, the specific pathways for the synthesis of compounds 6a, 9a, 6B, 8B are as follows:
Figure BDA0002625071350000071
in particular, the specific synthetic route of compound 13a is shown below:
Figure BDA0002625071350000081
in particular, the specific synthetic route for compounds 21a-23a is shown below:
Figure BDA0002625071350000082
the synthesis of compounds 2a-28a, 5b-8b specifically comprises the following steps:
2-Chlorobenzenesulfinic acid (2a)
2-Chlorobenzenesulfonyl chloride (5g, 23.7mmol), sodium sulfite (8.96g, 71.1mmol) were dissolved in 50mL of water at room temperature, heated at 80 ℃ for 5h, and the reaction was detected by TLC to be complete. The reaction solution is cooled, washed twice with dichloromethane (50mL × 2mL), the pH is adjusted to about 2-3 with 6M hydrochloric acid, a white flocculent precipitate is separated out, stirring is continued, a large amount of precipitate is separated out, filtering is carried out, and the process is repeated for 2-3 times, so that a white needle-shaped product 2a (3.99g, the yield is 95.68%) is obtained.
2-Chlorobenzenesulfonic acid sodium salt (3a)
Dissolving 2a (6g, 34.1mmol) in 20ml of methanol, adding NaOH (1.37g, 34.1mmol), heating in a water bath at 40 ℃ for 2h, and evaporating the reaction solution to dryness to obtain 6.6g of white powder with the yield of 97.8%.
4-Methylbenzenesulfinic acid (2b)
4-tosyl chloride (10g, 52.5mmol), sodium sulfite (19.8g, 157.4mmol) were dissolved in 70mL of water at room temperature, heated at 80 ℃ for 5h, and the reaction was checked by TLC to completion. The reaction solution was cooled, washed twice with dichloromethane (50 mL. times.2), precipitated as white flocculent precipitates with 6M hydrochloric acid at pH of about 2 to 3, stirred continuously, precipitated in large amounts, and filtered to obtain white needle-like product 2b (8.0g, 97.79% yield).
4-Methylbenzenesulfonic acid sodium salt (3b)
Dissolving 2b (4.5g, 26.2mmol) in 20ml methanol, adding NaOH (1.05g, 26.2mmol), heating in 40 deg.C water bath for 2h, evaporating reaction solution to dryness to obtain white powder 4.9g, yield 96.6%.
Sodium benzene sulfinate synthesis steps (3c-3 f):
benzenesulfonyl chloride (11mmol), sodium sulfite (22mmol) and sodium bicarbonate (22mmol) with different substituents were dissolved in 50ml of water at room temperature for 4h, and detected by TLC. And after the reaction is completed, evaporating the reaction solution to dryness, adding 30ml of methanol, heating and reacting at 78 ℃ for 1h, filtering insoluble substances while the solution is hot, evaporating the reaction solution to dryness, and repeating the steps to obtain sodium sulfinate 3c-3f with different substituents.
4-Chlorobenzenesulfonic acid sodium salt (3c)
White solid, yield 96.0%.
4-Methoxybenzenesulfonic acid sodium salt (3d)
White solid, yield 51%.
4-Tert-Butylphenylsulfinic acid sodium salt (3e)
White solid, yield 80.9%.
Sodium benzene sulfinate (3f)
Pale yellow solid, yield 100%.
4a-4f Synthesis:
adding sodium benzene sulfinate (1mmol) and 2, 3-dibromopropionic acid (0.67mmol) with different substituents into anhydrous DMF20 mL respectively, heating at 80 ℃ for 10-12 h, adding 50mL of purified water after TLC detection reaction is completed, extracting for 3-5 times by using ethyl acetate, washing an organic phase by using water, drying sodium chloride anhydrous magnesium sulfate overnight, filtering, evaporating a solvent under reduced pressure, mixing a sample by using 100-200-mesh silica gel, and purifying by using a silica gel column chromatography (PE: EA is 10:1) to obtain a powdery solid.
(E) -3- ((2-chlorophenyl) sulfonyl) acrylic acid (4a)
Pale yellow solid, yield 36.1%. Mp: 152-.1H NMR(600MHz,DMSO-d6)13.73(s,1H),8.12(dd,J=7.9,1.6Hz,1H),7.82(td,J=7.7,1.7Hz,1H),7.77(dd,J=8.0,1.3Hz,1H),7.71–7.66(m,2H),6.84(d,J=15.2Hz,1H).ESI-MS m/z:245.2[M-H]-
(E) -3-p-methylbenzenesulfonic acid (4b)
White solid, yield 46.7%. Mp:101-102 ℃.1H NMR(600MHz,DMSO-d6)13.52(s,1H),7.85–7.80(m,2H),7.65(d,J=15.2Hz,1H),7.50(d,J=8.0Hz,2H),6.66(d,J=15.2Hz,1H),2.43(s,3H).ESI-MS m/z:225.3[M-H]-
(E) -3- ((4-chlorophenyl) sulfonyl) acrylic acid (4c)
White solid, yield 62.9%. Mp:153-]-
(E) -3- ((4-methoxyphenyl) sulfonyl) acrylic acid (4d)
Pale yellow solid, yield 17%. Mp:97-98℃.1H NMR(600MHz,DMSO-d6)13.52(s,1H),7.87(d,J=9.0Hz,1H),7.64(d,J=15.2Hz,1H),7.20(d,J=9.0Hz,1H),6.63(d,J=15.1Hz,1H),3.88(s,3H).ESI-MS m/z:241.2[M-H]-
(E) -3- ((4- (tert-butyl) phenyl) sulfonyl) acrylic acid (4e)
White solid, yield 39.4%. Mp:140-142 ℃.1H NMR(600MHz,DMSO-d6)13.52(s,1H),7.89–7.85(m,2H),7.71(d,J=8.6Hz,2H),7.67(d,J=15.2Hz,1H),6.69(d,J=15.2Hz,1H),1.31(s,9H).ESI-MS m/z:267.3[M-H]-
(E) -3- (phenylsulfonyl) acrylic acid (4f)
Pale yellow solid, yield 34.36%. Mp 74-76 ℃.1H NMR(600MHz,DMSO-d6)7.97–7.94(m,2H),7.82–7.78(m,1H),7.72–7.68(m,2H),7.64(d,J=15.2Hz,1H),6.72(d,J=15.2Hz,1H);ESI-MS m/z:211.3[M-H]-。
(E) -3- ((2-chlorophenyl) sulfonyl) -N-phenethylacrylamide (5a)
Dissolving 4a (0.4g,1.63mmol) in 8mL of anhydrous tetrahydrofuran, adding O-benzotriazole-N, N, N' -tetramethylurea tetrafluoroborate (TBTU, 0.785g,2.445mmol) and triethylamine (0.494g, 4.89mmol) under ice bath, activating for 1h under ice bath, adding phenethylamine (0.24g, 1.95mmol), reacting for 12h at room temperature, and detecting the reaction completion by TLC. The organic phase was evaporated to dryness under reduced pressure, ethyl acetate 15mL was added, washed twice with water (3 × 25mL), washed three times with 2M HCl (3 × 25mL), washed three times with saturated sodium bicarbonate (3 × 25mL), washed three times with saturated sodium chloride (3 × 25mL), the organic phase was dried over anhydrous magnesium sulfate overnight, anhydrous magnesium sulfate was filtered off, the solvent was evaporated to dryness under reduced pressure, 100-200 mesh silica gel was stirred and purified by column chromatography to give a white solid 150mg, 24% yield. Mp 138-142 ℃.1H NMR(600MHz,DMSO-d6)8.83(t,J=5.7Hz,1H),8.11(dd,J=7.9,1.6Hz,1H),7.83–7.72(m,2H),7.70–7.65(m,1H),7.44(d,J=14.9Hz,1H),7.29(t,J=7.5Hz,2H),7.21(d,J=7.6Hz,3H),7.15(d,J=14.9Hz,1H),3.41(q,J=6.8Hz,2H),2.76(t,J=7.3Hz,2H);13C NMR(151MHz,DMSO)161.59,139.49,138.04,137.17,136.61,136.54,132.63,132.08,131.42,129.10,128.93,128.80,126.68,41.11,40.41,40.27,40.14,40.00,39.86,39.72,39.58,35.06.ESI-MS m/z:350.3[M+H]+
Methyl (E) -4- (3- ((2-chlorophenyl) sulfonyl) acrylamido) butanoate methyl ester (6a)
4a (0.1g, 0.406mmol) was dissolved in 5mL dry tetrahydrofuran, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI, 0.157g, 0.49mmol), 1-hydroxybenzotriazole (HOBT, 0.083g, 0.609mmol) triethylamine (0.124g, 1.218mmol) were added under ice bath, activation was carried out for 1h under ice bath, ethyl methyl-4-aminobutyrate (0.058g, 0.49mmol) was added, reaction was carried out for 6h at room temperature, and TLC was performed to detect completion of the reaction. The reaction solution was evaporated to dryness under reduced pressure, 10mL of water was added, extraction was carried out three times with ethyl acetate (3 × 20mL), washing was carried out two times with water (3 × 20mL), washing was carried out two times with 2M HCl (3 × 20mL), washing was carried out three times with saturated sodium bicarbonate (3 × 20mL), washing was carried out three times with saturated sodium chloride (3 × 20mL), the organic phase was dried over anhydrous magnesium sulfate overnight, filtration was carried out, the reaction solution was evaporated to dryness under reduced pressure, a sample was stirred with 100-200 mesh silica gel, and purification was carried out by column chromatography to obtain 80mg of a white solid with a yield. Mp:134-136 ℃.1H NMR(600MHz,DMSO-d6)8.31(d,J=6.2Hz,1H),8.29(d,J=8.5Hz,1H),8.21(d,J=13.6Hz,1H),7.97(d,J=8.4Hz,1H),7.83–7.79(m,1H),7.55(dd,J=8.3,7.1Hz,1H),6.60(d,J=13.6Hz,1H),3.60(s,3H),3.22(q,J=6.6Hz,2H),2.37(t,J=7.4Hz,2H),1.73(p,J=7.2Hz,2H).;13C NMR(151MHz,DMSO)173.40,161.67,138.02,137.13,136.60,136.56,132.62,132.09,131.42,128.92,51.75,38.84,31.06,24.52.ESI-MS m/z:346.2[M+H]+
(E) -3- ((2-chlorophenyl) sulfonyl) -N- (4-sulfamoylphenethyl) acrylamide (7a)
4a (0.6g, 2.44mmol) was dissolved in 8mL of dry tetrahydrofuran, TBTU (1.38g, 4.3mmol), triethylamine (0.58g, 5.7mmol) were added under ice bath, 4- (2-aminoethyl) benzenesulfonamide (0.685g, 3.4mmol), triethylamine (0.58g, 5.7mmol) were added, the reaction was allowed to react overnight at room temperature, and the reaction was complete by TLC. Filtering to remove insoluble precipitate, evaporating reaction solution under reduced pressure, adding 20mL water, extracting with ethyl acetate (3X 10mL) for three times, combining organic phases, washing organic phase with water for three times (3X 20mL), washing with saturated sodium carbonate for three times (3X 20mL), washing with 2N HCl for three times (3X 20mL), washing with saturated sodium chloride solution for three times(3X 20mL), drying over night with anhydrous magnesium sulfate, and carrying out sample mixing on 100-200 meshes of silica gel and passing through a column to obtain a white solid. The solid obtained is heated and dissolved by using a proper amount of MeOH, cooled in a refrigerator, white precipitate is separated out, filtered and washed twice by using the MeOH, and 108mg of white solid is obtained, and the yield is 9.15%. Mp 222-225 ℃.1H NMR(600MHz,DMSO-d6)8.85(t,J=5.7Hz,1H),8.11(dd,J=7.9,1.6Hz,1H),7.80(td,J=7.6,1.6Hz,1H),7.77–7.73(m,3H),7.67(td,J=7.6,1.3Hz,1H),7.45(d,J=15.0Hz,1H),7.41(d,J=8.2Hz,2H),7.30(s,2H),7.14(d,J=14.9Hz,1H),3.44(q,J=6.8Hz,2H),2.84(t,J=7.1Hz,2H);13C NMR(151MHz,DMSO)161.67,143.71,142.65,137.92,137.26,136.63,136.50,132.63,132.08,131.43,129.60,128.93,126.17,40.71,34.75.ESI-MS m/z:429.2[M+H]+
(E) -N-phenethyl-3-tosylacrylamide (8a)
White solid, yield 26.7%. Mp:191-193 ℃.1H NMR(600MHz,DMSO-d6)8.75(t,J=5.7Hz,1H),7.81–7.78(m,2H),7.49(d,J=8.0Hz,2H),7.41(d,J=14.9Hz,1H),7.31–7.27(m,2H),7.21(d,J=7.4Hz,3H),6.96(d,J=14.9Hz,1H),3.38(td,J=7.3,5.8Hz,2H),2.74(t,J=7.3Hz,2H),2.42(s,3H);13C NMR(151MHz,DMSO)161.87,145.53,139.50,139.34,136.42,134.89,130.71,129.06,128.81,128.23,126.67,41.03,35.11,21.60.ESI-MS m/z:330.4[M+H]+
Methyl- (E) -4- (3-tosylacrylamide) butanoate (9a)
White solid, yield 56.2%. Mp:214-216 ℃.1H NMR(600MHz,DMSO-d6)8.69(t,J=5.7Hz,1H),7.83–7.79(m,2H),7.51–7.47(m,2H),7.42(d,J=14.9Hz,1H),6.94(d,J=15.0Hz,1H),3.57(s,3H),3.15(td,J=6.9,5.6Hz,2H),2.42(s,3H),2.33(t,J=7.4Hz,2H),1.68(p,J=7.1Hz,2H);13C NMR(151MHz,DMSO)173.41,161.94,145.53,139.26,136.42,134.91,130.70,128.25,51.74,38.76,31.06,28.97,24.55,21.59.ESI-MS m/z:326.4[M+H]+
(E) -N- (4-Aminosulfonylphenethyl) -3-tosylacrylamide (10a)
Pale yellow solid, yield 13.5%. Mp 115-116 ℃.1H NMR(600MHz,DMSO-d6)8.78(t,J=5.7Hz,1H),7.82–7.78(m,2H),7.75–7.71(m,2H),7.49(d,J=8.1Hz,2H),7.45–7.38(m,3H),7.30(s,2H),6.95(d,J=14.9Hz,1H),3.42(q,J=6.9Hz,2H),2.82(t,J=7.1Hz,2H),2.42(s,3H);13C NMR(151MHz,DMSO)161.95,145.56,143.72,142.63,139.42,136.38,134.77,130.72,129.57,128.24,126.17,40.61,34.80,28.94,21.60.ESI-MS m/z:409.2[M+H]+
(E) -3- ((2-chlorophenyl) sulfonyl) -N- (4-methoxyphenethyl) acrylamide (11a)
White solid, yield 54.7%. Mp: 152-.1H NMR(600MHz,DMSO-d6)8.80(t,J=5.7Hz,1H),8.11(dd,J=7.9,1.6Hz,1H),7.83–7.74(m,2H),7.67(td,J=7.6,1.4Hz,1H),7.44(d,J=14.9Hz,1H),7.19–7.09(m,3H),6.89–6.81(m,2H),3.36(q,J=7.0Hz,2H),2.69(t,J=7.2Hz,2H);13C NMR(151MHz,DMSO)161.56,158.21,138.09,137.13,136.61,136.54,132.62,132.08,131.42,131.32,130.07,128.92,114.22,55.43,41.37,34.19.ESI-MS m/z:380.3[M+H]+
(E) -N- (4-methoxyphenylethyl) -3-tosylacrylamide (12a)
White solid, yield 26.4%. Mp 146-148 ℃.1H NMR(600MHz,DMSO-d6)8.72(t,J=5.7Hz,1H),7.80(d,J=8.2Hz,2H),7.49(d,J=8.0Hz,2H),7.41(d,J=14.9Hz,1H),7.15–7.10(m,2H),6.97(d,J=14.9Hz,1H),6.88–6.82(m,2H),3.72(s,3H),3.38–3.33(m,2H),2.68(t,J=7.3Hz,2H),2.42(s,3H);13C NMR(151MHz,DMSO)161.84,158.20,145.53,139.30,136.43,134.93,131.33,130.71,130.03,128.22,114.23,55.43,41.29,34.23,21.60.ESI-MS m/z:360.3[M+H]+
(E) -3- ((2-chlorophenyl) sulfonyl) -N- (4- (N- (cyclohexylcarbamoyl) aminosulfonyl) phenethyl) acrylamide (13a)
4a (0.4g,1.63mmol) was dissolved in 8mL dry tetrahydrofuran, activated for 2h with TBTU (0.81g, 2.5mmol), triethylamine (0.33g, 3.26mmol) added under ice-bath, 33(0.64g,1.95mmol), triethylamine (0.33g, 3,26mmol) added, reacted at room temperature, monitored by TLC. After the reaction is completed, the reaction solution is evaporated to dryness, dissolved by 20mL of ethyl acetate and washed twice by 10mL of water,washing with saturated sodium carbonate for three times (3 × 20mL), washing with 2N diluted hydrochloric acid for two times (3 × 20mL), washing with saturated sodium chloride for 2 times (3 × 20mL), drying with anhydrous magnesium sulfate overnight, filtering, evaporating the solvent under reduced pressure, mixing with 100-200 mesh silica gel, purifying by column chromatography to obtain white powdery solid, recrystallizing with MeOH to obtain white solid, washing with MeOH for two times to obtain 98mg of white product, and obtaining the yield of 11.13%. Mp: 199-.1H NMR(600MHz,DMSO-d6)10.32(s,1H),8.85(t,J=5.7Hz,1H),8.10(dd,J=7.9,1.7Hz,1H),7.81(dd,J=8.9,2.0Hz,2H),7.75(dd,J=8.0,1.3Hz,1H),7.67(td,J=7.6,1.3Hz,1H),7.48–7.42(m,3H),7.13(d,J=15.0Hz,1H),6.32(d,J=7.9Hz,1H),3.45(q,J=6.8Hz,2H),3.35–3.22(m,1H),2.86(t,J=7.1Hz,2H),1.68–1.43(m,6H),1.25–1.19(m,2H),1.12–1.08(m,2H).13C NMR(151MHz,DMSO)161.72,150.95,145.26,138.81,137.91,137.27,136.60,132.63,132.10,131.42,129.67,128.92,127.76,56.50,48.53,40.54,34.83,32.73,25.45,24.62,19.02.ESI-MS m/z:554.3[M+H]+
(E) -3- ((2-chlorophenyl) sulfonyl) -N- (4-methylphenylethyl) acrylamide (14a)
White solid, 45.7%. Mp 162-164 ℃.1H NMR(600MHz,DMSO-d6)8.79(s,1H),8.11(dd,J=7.9,1.5Hz,1H),7.82–7.78(m,1H),7.77–7.72(m,1H),7.68(t,J=7.4Hz,1H),7.43(d,J=14.9Hz,1H),7.14(d,J=15.0Hz,1H),7.09(s,4H),3.38(q,J=6.8Hz,2H),2.72(t,J=7.2Hz,2H),2.26(s,3H);13C NMR(151MHz,DMSO)161.58,138.05,137.16,136.59,136.36,135.60,132.63,132.09,131.40,129.37,128.96,128.92,41.20,34.64,21.09.ESI-MS m/z:364.2[M+H]+。
(E) -3- ((2-chlorophenyl) sulfonyl) -N- (2- (5-methoxy-1H-indol-3-yl) ethyl) acrylamide (15a)
Yellow solid, yield 29.5%. 147 ℃.1H NMR (600MHz, DMSO-d6)10.68(s,1H),8.88(t, J ═ 5.8Hz,1H),8.11(dd, J ═ 7.9,1.6Hz,1H),7.80(td, J ═ 7.7,1.7Hz,1H),7.75(dd, J ═ 8.0,1.3Hz,1H),7.67(td, J ═ 7.6,1.3Hz,1H),7.46(d, J ═ 14.9Hz,1H),7.22(d, J ═ 8.7Hz,1H),7.17(d, J ═ 14.9Hz,1H),7.11(d, J ═ 2.4Hz,1H),7.00(d, 2.7H, 1H), 7.8.7.5 (d, J ═ 2.4H, 7.84H), 7.6H, 7.5 (dd, 3H), 7.5H, 3H); 13CNMR (151MHz, DMSO))161.62,153.49,138.17,137.12,136.62,136.57,132.63,132.12,131.85,131.39,128.90,127.95,123.93,112.49,111.70,111.58,100.53,55.80,40.56,25.20.ESI-MS m/z:419.2[M+H]+
(E) -3- ((2-chlorophenyl) sulfonyl) -N- (4- (trifluoromethyl) phenethyl) acrylamide (16a)
White solid, yield 41%. Mp: 182-.1H NMR(600MHz,DMSO-d6)8.85(t,J=5.8Hz,1H),8.11(dd,J=7.9,1.7Hz,1H),7.80(td,J=7.6,1.7Hz,1H),7.76(dd,J=8.0,1.3Hz,1H),7.70–7.62(m,3H),7.48–7.41(m,3H),7.14(d,J=14.9Hz,1H),3.46(q,J=6.7Hz,2H),2.87(t,J=7.1Hz,2H);13C NMR(151MHz,DMSO)161.70,144.51,137.92,137.24,136.58,136.56,132.60,132.08,131.41,130.00,128.90,127.61,125.58,125.56,123.97,40.61,34.79.ESI-MS m/z:418.4[M+H]+
(E) -N- (3, 4-dihydroxyphenethyl) -3-tosylacrylamide (17a)
White solid, yield 12.9%. Mp:164 ℃.1H NMR (400MHz, DMSO-d6) 8.84-8.71 (m,3H),7.86(d, J ═ 7.9Hz,2H),7.55(d, J ═ 7.9Hz,2H),7.46(d, J ═ 14.9Hz,1H),7.02(d, J ═ 14.9Hz,1H),6.68(d, J ═ 7.9Hz,1H), 6.65-6.60 (m,1H), 6.52-6.45 (m,1H),3.34(d, J ═ 6.9Hz,2H),2.61(d, J ═ 6.7Hz,2H),2.48(s,3H).13C NMR(151MHz,DMSO)161.80,145.55,145.52,144.09,139.26,136.46,135.01,130.72,130.23,128.21,119.64,116.40,115.97,41.43,34.58,21.60.ESI-MS m/z:362.3[M+H]+。
(E) -N- (3, 4-Dimethoxyphenylethyl) -3-toluenesulfonylacrylamide (18a)
White solid, yield 57.43%. Mp:148-150 ℃.1H NMR(600MHz,DMSO-d6)8.71(t,J=5.7Hz,1H),7.82–7.78(m,2H),7.52–7.48(m,2H),7.41(d,J=14.9Hz,1H),6.98(d,J=14.9Hz,1H),6.85(d,J=8.1Hz,1H),6.79(d,J=2.0Hz,1H),6.71(dd,J=8.1,2.0Hz,1H),3.71(d,J=1.2Hz,6H),3.37(td,J=7.3,5.8Hz,2H),2.68(t,J=7.2Hz,2H),2.43(s,3H).13C NMR(151MHz,DMSO-d6)161.84,149.04,147.74,145.54,139.32,136.43,134.96,131.91,130.72,128.22,120.89,112.95,112.31,55.94,55.81,41.21,34.64,21.60.ESI-MS m/z:390.3[M+H]+。
(E) -3- ((2-chlorophenyl) sulfonyl) -N- (2- (pyridin-2-yl) ethyl) acrylamide (19a)
White powdery solid, yield 21.94%. Mp:143-144 ℃.1H NMR(600MHz,DMSO-d6)8.82(t,J=5.7Hz,1H),8.52–8.48(m,1H),8.11(dd,J=8.0,1.6Hz,1H),7.80(td,J=7.6,1.6Hz,1H),7.75(dd,J=8.1,1.3Hz,1H),7.69(dtd,J=16.5,7.6,1.6Hz,2H),7.44(d,J=14.9Hz,1H),7.26(d,J=7.8Hz,1H),7.23(ddd,J=7.6,4.8,1.1Hz,1H),7.14(d,J=14.9Hz,1H),3.56(td,J=7.2,5.7Hz,2H),2.93(t,J=7.2Hz,2H);13C NMR(151MHz,DMSO)161.65,159.08,149.55,138.03,137.20,136.93,136.72,136.61,136.59,132.74,132.63,132.24,132.10,131.41,128.92,128.85,123.65,122.06,39.34,37.26.ESI-MS m/z:351.2[M+H]+
(E) -3- ((2-chlorophenyl) sulfonyl) -N- (3-phenylpropyl) acrylamide (20a)
White powdery solid, yield 48.5%. Mp:174-176 ℃.1H NMR(600MHz,DMSO-d6)8.77(t,J=5.7Hz,1H),8.12(dd,J=7.9,1.7Hz,1H),7.80(td,J=7.6,1.6Hz,1H),7.76(dd,J=8.0,1.3Hz,1H),7.68(td,J=7.6,1.4Hz,1H),7.45(d,J=14.9Hz,1H),7.28(t,J=7.6Hz,2H),7.22–7.19(m,2H),7.19–7.16(m,2H),3.18(td,J=7.0,5.6Hz,2H),2.60(t,J=7.7Hz,2H),1.76(dq,J=9.0,7.1Hz,2H);13C NMR(151MHz,DMSO)161.60,141.90,138.14,137.09,136.65,136.59,132.64,132.11,131.41,128.93,128.76,126.26,39.18,32.88,30.83.ESI-MS m/z:364.2[M+H]+
(E) -N- (4- (N-methylaminosulfonyl) phenethyl) -3-toluenesulfonylacrylamide (21a)
4a (0.5g, 2.03mmol) was dissolved in 10mL of dry THF solution, TBTU (0.782g, 2.44mmol) and TEA (0.41g, 4.06mmol) were added, activated for 2h in ice bath, 37a (0.522g, 2.44mmol) and TEA (0.41g, 4.06mmol) were added to the reaction mixture, reacted at room temperature and checked by TLC. After completion of the reaction, the reaction mixture was evaporated to dryness, dissolved in ethyl acetate (20mL), washed twice with 2M dilute hydrochloric acid (2X 20mL), washed three times with saturated sodium carbonate (2X 20mL), washed three times with saturated sodium chloride (3X 20mL), dried over anhydrous magnesium sulfate overnight, evaporated to dryness under reduced pressure, and recrystallized from methanol to give 0.392g of a pale pink powdery solid with a yield of 36.64%。Mp:186-188℃.1H NMR(600MHz,DMSO-d6)8.85(t,J(3×20mL)=5.7Hz,1H),7.90–7.85(m,2H),7.80–7.75(m,2H),7.59–7.56(m,2H),7.53–7.51(m,2H),7.47(s,1H),7.45(t,J=5.0Hz,1H),7.03(d,J=14.9Hz,1H),3.51(q,J=6.9Hz,2H),2.92(t,J=7.1Hz,2H),2.50(s,3H),2.46(d,J=5.0Hz,3H);13C NMR(151MHz,DMSO)161.96,145.56,144.42,139.40,137.67,136.38,134.77,130.72,129.89,128.23,127.22,40.50,34.86,29.10,21.60.ESI-MS m/z:423.3[M+H]+
(E) -N- (4- (N- (4-methoxybenzyl) aminosulfonyl) phenethyl) -3-p-toluenesulfonylacrylamide (22a) was white solid in 19% yield. Mp:170-172 ℃.1H NMR(600MHz,DMSO-d6)8.77(t,J=5.7Hz,1H),7.98(t,J=6.3Hz,1H),7.82–7.77(m,2H),7.72–7.67(m,2H),7.51–7.45(m,2H),7.44–7.38(m,3H),7.14–7.09(m,2H),6.96(d,J=14.9Hz,1H),6.85–6.79(m,2H),3.87(d,J=6.3Hz,2H),3.71(s,3H),3.42(q,J=6.8Hz,2H),2.83(t,J=7.2Hz,2H),2.42(s,3H);13C NMR(151MHz,DMSO)161.97,158.89,145.55,144.27,139.40,139.17,136.38,134.79,130.72,129.92,129.80,129.39,128.22,127.06,114.07,55.52,46.12,40.57,34.88,21.60.ESI-MS m/z:529.3[M+H]+
(E) -N- (4- (N-Propylaminosulfonyl) phenethyl) -3-p-toluenesulfonylacrylamide (23a)
White solid, yield 36.1%. Mp 199-.1H NMR(600MHz,DMSO-d6)8.76(t,J=5.7Hz,1H),7.82–7.77(m,2H),7.73–7.67(m,2H),7.54–7.45(m,3H),7.45–7.38(m,3H),6.95(d,J=14.9Hz,1H),3.45–3.40(m,2H),2.83(t,J=7.1Hz,2H),2.70–2.62(m,2H),2.42(s,3H),1.35(h,J=7.3Hz,2H),0.77(t,J=7.4Hz,3H);13C NMR(151MHz,DMSO)161.94,145.55,144.24,139.39,139.05,136.39,134.78,130.71,129.83,128.22,127.00,44.80,40.50,34.85,22.84,21.60,11.60.ESI-MS m/z:451.3[M+H]+
(E) -3- ((4-methoxyphenyl) sulfonyl) -N-phenethylacrylamide (24a)
White solid, yield 42%. Mp:184-187 ℃.1H NMR(600MHz,DMSO-d6)8.77(t,J=5.7Hz,1H),7.92–7.85(m,2H),7.43(d,J=14.9Hz,1H),7.34(dd,J=8.3,6.9Hz,2H),7.29–7.23(m,5H),6.98(d,J=14.9Hz,1H),3.92(s,3H),3.44(td,J=7.4,5.9Hz,2H),2.80(t,J=7.3Hz,2H);13C NMR(151MHz,DMSO)164.18,161.98,139.69,139.51,134.22,130.66,130.59,129.05,128.81,126.66,115.53,56.38,41.02,35.12.ESI-MS m/z:346.2[M+H]+
(E) -3- ((4-Chloroethyl) sulfonyl) -N- (4-aminosulfonylphenethyl) acrylamide (25a)
White solid, yield 29.92%. Mp:207-208 ℃.1H NMR(600MHz,DMSO-d6)8.79(t,J=5.8Hz,1H),7.93(d,J=8.5Hz,2H),7.75(dd,J=14.8,8.2Hz,4H),7.52(d,J=14.9Hz,1H),7.40(d,J=8.0Hz,2H),7.29(s,2H),7.01(d,J=14.9Hz,1H),3.43(q,J=6.7Hz,2H),2.83(t,J=7.2Hz,2H);13C NMR(151MHz,DMSO)161.82,143.70,142.66,139.93,138.82,138.16,135.78,130.41,130.21,129.56,126.18,40.62,34.80.ESI-MS m/z:429.3[M+H]+
(E) -3- ((4-chlorophenyl) sulfonyl) -N-phenethylacrylamide (26a)
White solid, yield 36.7%. Mp:174-175 ℃.1H NMR(600MHz,DMSO-d6)8.77(t,J=5.7Hz,1H),7.96–7.93(m,2H),7.79–7.75(m,2H),7.50(d,J=14.9Hz,1H),7.29(td,J=7.2,1.5Hz,2H),7.23–7.20(m,3H),7.02(d,J=14.9Hz,1H),3.40(td,J=7.3,5.8Hz,2H),2.76(t,J=7.3Hz,2H);13C NMR(151MHz,DMSO)161.75,139.92,139.49,138.74,138.19,135.89,130.41,130.20,129.05,128.81,126.67,41.05,35.10.ESI-MS m/z:350.3[M+H]+
(E) -3- ((4- (tert-butylethylethyl) sulfonyl) -N- (4-phenethylsulfonamide) acrylamide (27a)
Pale yellow solid, yield 28.4%. Mp:210-212 ℃.1H NMR(600MHz,DMSO-d6)8.80(d,J=5.9Hz,1H),7.86–7.82(m,2H),7.77–7.74(m,2H),7.72–7.69(m,2H),7.46–7.39(m,3H),7.28(s,2H),7.00(d,J=14.9Hz,1H),3.43(q,J=6.8Hz,2H),2.84(t,J=7.1Hz,2H),1.32(s,9H);13C NMR(151MHz,DMSO)161.98,158.07,143.72,142.66,139.36,136.49,134.97,129.56,128.10,127.18,126.18,40.62,35.54,34.81,31.17.ESI-MS m/z:451.2[M+H]+,468.3[M+NH4]+
(E) -3- (phenylsulfonyl) -N- (4-aminosulfonylphenethyl) acrylamide (28a)
White solid, yield 39.8%. Mp 213-214 ℃.1H NMR(600MHz,DMSO-d6)8.78(t,J=5.7Hz,1H),7.93(dt,J=7.3,1.3Hz,2H),7.81–7.77(m,1H),7.76–7.74(m,2H),7.72–7.68(m,2H),7.47(d,J=14.9Hz,1H),7.41(d,J=8.2Hz,2H),7.28(s,2H),7.01(d,J=15.0Hz,1H),3.46–3.41(m,1H),2.84(t,J=7.1Hz,2H);13C NMR(151MHz,DMSO)161.92,143.73,142.67,139.35,139.17,135.35,134.83,130.29,129.57,128.17,126.19,40.63,34.81.ESI-MS m/z:395.2[M+H]+
Phenethyl (E) -3- ((2-chlorophenyl) sulfonyl) acrylate (5b)
4a (0.3g, 1.22mmol) was dissolved in dry THF, phenethyl alcohol (0.23g, 2.44mmol) was added, triphenylphosphine (0.64g, 2.44mmol) was added under ice-bath, DEAD (0.43g, 2.44mmol) was slowly added dropwise, the reaction at room temperature, monitored by TCL. After the reaction is completed, evaporating tetrahydrofuran to dryness, dissolving with a proper amount of diethyl ether, standing overnight in a refrigerator at 4 ℃, separating out a large amount of white precipitate, filtering out the precipitate, evaporating the filtrate to dryness under reduced pressure, mixing the filtrate with 100-200 meshes of silica gel, and purifying by column chromatography to obtain 0.189g of a yellow oily product with the yield of 44.37%. Mp is 38-40 deg.C,1H NMR(600MHz,DMSO-d6)8.12(ddd,J=8.0,3.5,1.6Hz,1H),7.85–7.79(m,1H),7.79–7.76(m,1H),7.74(d,J=15.2Hz,1H),7.69(td,J=7.6,1.3Hz,1H),7.29–7.25(m,3H),7.23–7.20(m,1H),6.87(d,J=15.2Hz,1H),4.38(t,J=6.8Hz,2H),2.95(t,J=6.8Hz,2H).13C NMR(151MHz,DMSO)163.28,141.78,138.09,136.88,136.00,133.62,132.64,132.20,131.72,129.36,128.95,128.82,126.93,66.51,34.52.ESI-MS m/z:351.3[M+H]+
methyl (E) -4- ((3- ((2-chlorophenyl) sulfonyl) acryloyl) oxy) butanoate (6b)
Pale yellow oil, yield 47.6%,1H NMR(400MHz,DMSO-d6)8.13(dd,J=7.9,1.6Hz,1H),7.85–7.74(m,3H),7.69(td,J=7.6,1.4Hz,1H),6.90(d,J=15.3Hz,1H),4.19(t,J=6.3Hz,2H),3.57(s,3H),2.44(t,J=7.3Hz,2H),1.90(q,J=6.8Hz,2H);13C NMR(101MHz,DMSO)173.34,163.41,141.73,136.87,135.98,133.70,132.65,132.25,131.76,128.94,65.29,51.76,30.24,23.84.ESI-MS m/z:347.3[M+H]+,364.3[M+NH4]+
phenethyl (E) -3-p-methylbenzenesulfonylacrylamide (7b)
White powdery solid, yield 47.9%. Mp is 76-78 deg.C.1H NMR(600MHz,DMSO-d6)7.85–7.79(m,2H),7.72(d,J=15.2Hz,1H),7.50(d,J=8.0Hz,2H),7.32–7.17(m,5H),6.71(d,J=15.2Hz,1H),4.35(t,J=6.9Hz,2H),2.93(t,J=6.9Hz,2H),2.43(s,3H).13C NMR(151MHz,DMSO)163.52,145.95,143.76,138.05,135.74,130.85,130.76,129.32,128.83,128.55,126.93,66.37,34.52,21.62.ESI-MS m/z:331.3[M+H]+,348.3[M+NH4]+
Methyl (E) -4- ((3-tosylacryloyl) oxy) butanoate (8b)
White powder, yield 47.14%. Mp 40-42 ℃.1H NMR(600MHz,DMSO-d6)7.87–7.81(m,2H),7.78(d,J=15.2Hz,1H),7.53–7.48(m,2H),6.72(d,J=15.2Hz,1H),4.16(t,J=6.3Hz,2H),3.56(s,3H),2.51(p,J=1.8Hz,2H),2.45–2.40(m,5H),1.90–1.84(m,2H).13CNMR(151MHz,DMSO)173.32,163.59,145.93,143.64,135.77,130.93,130.75,128.56,65.12,51.74,40.46,40.32,40.18,40.03,39.89,39.75,39.61,30.26,23.86,21.61.ESI-MS m/z:327.3[M+H]+,344.3[M+NH4]+
Methyl-4-aminobutyric acid methyl ester (29a)
4-aminobutyric acid (1g, 9.7mmol) is dissolved in 5mL of methanol, thionyl chloride is slowly dropped under ice bath, the reaction is carried out for 2h at room temperature, the methanol is evaporated after the TLC detection reaction is completed, a crude product is obtained, and the crude product is directly put into the next step without further purification. ESI-MS M/z 118.2[ M + H ]]+
Methyl 4-hydroxybutyric acid (29b)
Dissolving gamma-butyrolactone (5g, 23.3mmol) in 19mL of methanol, adding triethylamine (35g, 139.4mmol), heating at 60 ℃, reacting for 18h, evaporating the reaction solution after the reaction is completed, stirring with 100-mesh silica gel, purifying by column chromatography to obtain 2.51g of light yellow oily substance, and drying in an oven overnight. The yield was 36.64%。ESI-MS m/z:119.2[M+H]+. Tert-butyl- (4-aminosulfonylphenethyl) carbamate (31)
4- (2-aminoethyl) benzenesulfonamide (30) (1g, 5.0mmol) was dissolved in 12mL of dry N, N-dimethylformamide, di-tert-butyl dicarbonate (1.1g, 5.0mmol) was added slowly and the reaction was allowed to proceed at room temperature with TLC monitoring. After the reaction was completed, 30mL of water was added to precipitate a white precipitate, which was further stirred at room temperature for 1h, and the precipitate was filtered off, washed with water, and dried to obtain 1.29g of a white powder. The yield was 86%. ESI-MS M/z 301.3[ M + H ]]+,318.3[M+NH4]+
Tert-butyl- (4- (N- (cyclohexylcarbamoyl) sulfamoyl) phenethyl) carbamate (32)
Compound 31(1.5g, 5mmol), potassium carbonate (2.073g, 15mmol) were dissolved in acetone, heated to 55 deg.C, and cyclohexyl isocyanate (3.034g, 20mmol) was added dropwise slowly to reflux and monitored by TLC. After the reaction is completed, cooling to room temperature, adding water (water: acetone ≈ 1:1), stirring for 30min, filtering, washing filter residue with acetone and 2.5% potassium carbonate solution, filtering, mixing filtrates, and evaporating to obtain white powder product 0.86g with yield of 40.41%. 1H NMR (600MHz, DMSO-d6) 7.63-7.59 (M,2H),7.13(d, J ═ 8.1Hz,2H),6.88(t, J ═ 5.7Hz,1H),5.58(d, J ═ 11.8Hz,1H), 3.44-3.25 (M,1H), 3.15-3.08 (M,2H), 2.72-2.66 (M,2H), 1.67-1.46 (M,6H),1.37(s,9H), 1.27-1.13 (M,2H), 1.10-0.96 (M,2H), ESI-MS M/z 426.4[ M + H ] +.
4- (2-Aminoethyl) -N- (cyclohexylcarbamoyl) benzenesulfonamide (33)
Compound 32(0.1g, 0.24mmol) was dissolved in 4mL of ethyl acetate, and a saturated solution of ethyl acetate with hydrochloric acid was slowly added thereto to react at room temperature for 12 hours, whereupon the solution became clear from turbidity and then a precipitate was precipitated, and filtration was carried out to obtain 51.2mg of a white precipitate with a yield of 76.5%.1H NMR(400MHz,DMSO-d6)7.63(d,J=7.8Hz,2H),7.17(d,J=7.8Hz,2H),5.76(s,1H),3.21–3.10(m,1H),2.75(t,J=7.3Hz,2H),2.64(t,J=7.2Hz,2H),1.72–1.43(m,6H),1.40–1.08(m,2H),1.08–0.96(m,2H).ESI-MS m/z:326.3[M+H]+
N-phenethylacetamide (34)
Beta-phenylethylamine (0.5g 4.13 mm) was added at room temperatureol) was dissolved in dichloromethane (4mL), acetic anhydride (0.484g, 4.75mmol) was slowly added under ice bath, TLC monitored for reaction completion, ice water was slowly added, extraction was performed, the organic phase was separated, the aqueous phase was extracted once with dichloro, the organic phases were combined, dried over anhydrous magnesium sulfate and evaporated to dryness to give a pale yellow oil with 100% yield. ESI-MS M/z 164.3[ M + H ]]+
4- (2-Acetylamidoethyl) benzenesulfonyl chloride (35)
Adding compound 34(3g, 18.38mmol) into a reaction bottle under ice bath, slowly dropwise adding chlorosulfonic acid, reacting at room temperature, monitoring the reaction by TLC (thin layer chromatography), adding a large amount of ice water into the reaction solution to separate out a white precipitate, stirring for about 15-30min, filtering, draining, and carrying out sample mixing and column passing on 100-200-mesh silica gel to obtain 2.67g of white precipitate with the yield of 55.66%.1H NMR(600MHz,DMSO-d6)7.91(d,J=5.8Hz,1H),7.30–7.27(m,2H),7.20(d,J=6.6Hz,2H),3.28–3.21(m,2H),2.69(t,J=7.5Hz,2H),1.78(s,3H).ESI-MS m/z:262.0[M+H]+
N- (4- (N-Methylsulfonamido) phenethyl) acetic acid amine (36a)
Compound 35(0.7g, 2.68mmol) is dissolved in dichloromethane (10mL) at room temperature and methylamine hydrochloride (0.216g, 3.2mmol), K is added2CO3(0.812g, 8.04mmol), reaction at room temperature, and completion of reaction monitored by TLC. The reaction solution was evaporated to dryness, dissolved in ethyl acetate, washed twice with 1M hydrochloric acid (3 × 20mL), twice with anhydrous sodium carbonate (3 × 20mL), twice with saturated sodium chloride (3 × 20mL), dried over anhydrous magnesium sulfate overnight, stirred with 100-200 mesh silica gel, and subjected to column chromatography to obtain a pale yellow precipitate of 0.523g with a yield of 76.13%.1H NMR(600MHz,DMSO-d6)7.94(t,J=5.7Hz,1H),7.71–7.67(m,2H),7.45–7.41(m,2H),7.39(q,J=5.0Hz,1H),3.29(td,J=7.3,5.7Hz,2H),2.79(t,J=7.2Hz,2H),2.39(d,J=5.0Hz,3H),1.78(s,3H).ESI-MS m/z:257.2[M+H]+
N- (4- (N- (4-methoxybenzyl) sulfonamide) phenethyl) acrylamide (36b)
Compound 35(1.2g, 4.67mmol) was dissolved in dry THF at room temperature, p-methoxyphenethylamine (3.2g, 23.35mmol) and TEA (2.36g, 23.35mmol) were added, and the reaction was allowed to proceed at room temperature overnight. Filtering to remove precipitate after reaction, and evaporating to drynessDissolving the solid with ethyl acetate, washing twice with 2M dilute hydrochloric acid (3X 20mL), washing twice with saturated sodium chloride (3X 20mL), drying over night with anhydrous magnesium sulfate, mixing with 100-200 mesh silica gel, and purifying by column chromatography to obtain 0.5g of white powdery solid with a yield of 29.4%.1H NMR(600MHz,DMSO-d6)7.97(t,J=6.3Hz,1H),7.92(t,J=5.6Hz,1H),7.73–7.70(m,2H),7.42–7.38(m,2H),7.15–7.12(m,2H),6.85–6.82(m,2H),3.90(d,J=6.3Hz,2H),3.72(s,3H),3.31–3.27(m,2H),2.79(t,J=7.3Hz,2H),1.79(s,3H).ESI-MS m/z:363.4[M+H]+
N- (4- (N-Propanesulfonyl) phenethyl) acrylamide (36c)
White powdery solid, yield 35%. ESI-MS M/z 285.3[ M + H ]]+
4- (2-aminoethyl) -N-methylbenzenesulfonamide (37a)
36(0.4g, 1.56mmol) was dissolved in 2N sodium hydroxide solution at room temperature and refluxed for 12 h; after the reaction was completed, the pH was adjusted to be weakly alkaline, a large amount of precipitate was precipitated, filtered, extracted with dichloromethane (15mL), the residue was washed three times with dichloromethane (3X 10mL), the organic phases were combined, washed twice with saturated sodium chloride (3X 20mL), dried over anhydrous magnesium sulfate overnight, and the organic solvent was distilled off under reduced pressure to obtain 0.31g of a crude product as a white powder with a yield of 92.8%.1H NMR(600MHz,DMSO-d6)7.81–7.70(m,2H),7.49(d,J=8.1Hz,2H),2.86(t,J=6.8Hz,2H),2.78(t,J=7.1Hz,2H),2.46(s,3H).ESI-MS m/z:215.3[M+H]+
4- (2-aminoethyl) -N- (4-methoxybenzyl) benzenesulfonamide (37b)
White powdery solid, yield 93.2%.1H NMR(600MHz,DMSO-d6)7.74–7.64(m,2H),7.42–7.36(m,2H),7.16–7.09(m,2H),6.86–6.77(m,2H),3.89(s,2H),3.71(s,3H),2.82–2.78(m,2H),2.72(t,J=7.1Hz,2H).ESI-MS m/z:321.3[M+H]+
4- (2-aminoethyl) -N-propylbenzenesulfonamide (37c)
Off-white solid, yield 63%. ESI-MS M/z 243.3[ M + H ]]+
Example 2
This example performed cytotoxicity evaluation of compounds 2a-26a, 5b-8 b:
experimental materials and instruments: DMEM medium; fetal bovine serum FBS; 100 Xcyan streptomycin, trypsin, MTT powder, PBS buffer solution powder, sterile DMSO and sterile deionized water; BAY 11-7082; multifunctional enzyme mark instrument
The experimental steps are as follows: the mouse mononuclear macrophage J774A.1 cell culture is started to be used for experiments when the 3 rd generation cell culture is carried out, when the cell density is about 80%, the cell is scraped by a cell scraper, the cell is centrifuged at 1000rm for 5min, the cell is resuspended, the cell is blown uniformly, the cell density is adjusted to 1X105, 100 mu L of cell culture medium is sucked into a 96-well plate, PBS is added to the outermost circle of the 96-well plate, 37 ℃ is carried out, and the cell culture medium stays overnight (12h) in a 5% CO2 culture box. The test compound dissolved in DMSO and prepared in a concentration of 25mM and a positive control stock solution Bay 11-7082 were diluted with DMEM to a test solution (100. mu.M-6.25. mu.M) by a gradient dilution method, and 100. mu.M (3.125. mu.M-50. mu.M) of the test solution was added to each well. Each compound was set at 5 concentrations, and each concentration was set at 5 replicate wells. Blank control wells containing no cells and medium and 100% control containing no drug were also set. After the addition of the drug, the mixture is cultured for 24 hours in an incubator at 37 ℃ and 5% CO 2. In a clean bench, 20. mu.L of MTT solution was added to each well, the mixture was incubated in the same culture environment for 4 hours, the medium in a 96-well plate was discarded, DMSO was added thereto at a volume of 150. mu.L/well to dissolve formazan formed, the resulting solution was shaken on a shaker at room temperature for 10 minutes, and the OD value of absorbance was measured at a wavelength of 490 nm.
Figure BDA0002625071350000221
Calculating different inhibition rates of the test compound according to the formula, fitting an S curve through GrphpPad Prism 5.0, and calculating the IC of the compound50
The experimental results are as follows: the results of cytotoxicity evaluation of the compounds 2a-26a, 5b-8 and the positive drug BAY 11-7082 are shown in Table 1.
Results of cytotoxicity evaluation of the Compounds of Table 1
Figure BDA0002625071350000231
Figure BDA0002625071350000241
[a] The experimental results are expressed as mean ± standard deviation from three independent experiments.
As can be seen from the experimental results, almost all compounds were less cytotoxic or comparable to BAY 11-7082, with compounds 7a, 10a, 21a, 5b and 9b being less cytotoxic than the positive compounds.
Example 3
This example was evaluated on part of the compounds for NLRP3 inflammatory body inhibitory activity:
experimental materials and instruments: LPS (Escherichia coli 0111: B4) L2361, ATP A6419 mouse IL-1 beta ELISA kit, centrifuge, 37 degree shaking table, common shaking table, multifunctional microplate reader
The experimental steps are as follows: preparing LPS mother liquor: accurately weigh 1mg of LPS into 1mL sterile deionized water, vortex and sonicate until the LPS powder is dissolved, dispense 20. mu.L/tube and store at-20 ℃.
Sodium hydroxide solution: 24mg of sodium hydroxide is accurately weighed, 600 mul of sterile deionized water is added into a sterile super clean bench to obtain 1M NaOH solution which is prepared on site.
Preparing an ATP solution: ATP 27mg was accurately weighed into 400. mu.L of the medium in a sterile clean bench, adjusted to pH 7 with 1M sodium hydroxide solution, and supplemented to 500. mu.L of the medium, ready for use.
NLRP3 inflammasome activation
Mouse mononuclear macrophage J774A.1 cultured to the third generation is used for experiment, and the mouse mononuclear macrophage J774A.1 is plated when the cell density is 70-80%. Carefully scraping cells by using a cell scraper, centrifuging, counting, adjusting the cell density to 106 cells/mL, adding 100 mu L of cells into each hole, supplementing 80 mu L of fresh culture medium, staying overnight (12h) in an incubator, adding LPS diluted by the culture medium into the culture medium on the next day, keeping the final concentration to be 1 mu g/mL, continuously culturing for 5.5h in the incubator, adding a solution of a compound to be detected diluted by the culture medium, culturing for 30min in the incubator, adding an ATP solution with the final concentration of 5mM into each hole except a blank group, culturing for 30min in the incubator, collecting supernatant, centrifuging at 1500rpm in a centrifuge at 4 ℃, centrifuging for 15min, transferring to a new EP tube, marking, and immediately detecting or storing at-20 ℃.
IL-1 beta content detection
(1) Pre-encapsulated capture antibody
Taking an enzyme label plate with N +8 holes according to the number of the samples to be measured, and according to the capture antibody: preparing a capture antibody solution at a ratio of ddH 2O-1: 99, covering a sealing plate membrane on each well by 100 mu L, and incubating for 12-16h at 4 ℃;
(2) cleaning pre-coated ELISA plate
Taking out the ELISA plate from a 4-degree refrigerator before the next day to be tested, placing the ELISA plate to room temperature, cleaning the ELISA plate for 4 times by using 1X PBST, wherein each hole is at least 300 mu L, throwing out liquid after each cleaning, and drying the liquid in a sanitary way until no residual liquid exists in the hole, adding 200 mu L of 1X Assay Diluent A into each hole after cleaning, and incubating the holes on a shaking table for 1h at room temperature;
(3) adding a sample to be tested
Preparing an IL-1 beta standard: dissolving 26ng IL-1 beta standard in 200 μ L sample diluent, standing at room temperature for 15min, packaging after completely dissolving, and storing at-70 deg.C.
Preparing a standard solution: dissolving 15.6 μ L of the mother solution of the standard substance in 984.6 μ L of the sample diluent to obtain 2000pg/mL of the solution of the standard substance, and performing gradient dilution to obtain 1000pg/mL, 500pg/mL, 250pg/mL, 125pg/mL, 62.5pg/mL, 31.25pg/mL and 0pg/mL of the standard substance.
After the Assay Diluent A is sealed, PBST is used for cleaning for 4 times, the minimum washing liquid of 300 mu L is taken from each hole, 100 mu L of standard substance and sample are added into an enzyme label plate, a sealing plate membrane is covered, and the incubation is carried out for 2h at room temperature;
(4) adding a detection antibody
According to Detection antibody: preparing a detection antibody according to the ratio of 1:99 of the diluent, after the incubation of the ELISA plate is finished, washing PBST for 4 times, adding 100 mu L of detection antibody into each hole, and incubating for 1h at room temperature;
(5) adding biotinylated antibody
Under dark conditions, according to biotinylated antibody: 1-diluted solution: 999, preparing a biotinylated antibody solution, after the incubation at room temperature is finished, washing PBST for 4 times, washing Detection antibody, adding 100 mu L of biotinylated antibody into each hole, and incubating at room temperature for 30 min;
(6) adding substrate for color development
After the biotinylated antibody incubation is finished, washing the PBST for 5 times by 1X, keeping the PBST for 1min each time, then patting the PBST dry after washing, adding 100 mu L of substrate into each hole, and incubating for 30min in a dark place at 37 ℃;
(7) the reaction was terminated and detected
After the substrate incubation, 100 mu L of stop solution is added into each hole, and the OD value under the wavelength of 450nm is detected on a multifunctional enzyme-labeling instrument within 15min, and 630nm is taken as the wavelength reference.
(8) Data processing
And (3) taking the absorbance as an abscissa and the content of the IL-1 beta as an ordinate, drawing a standard Curve through the Curve Expert 14, and bringing the absorbance of all samples into the standard Curve to obtain the IL-1 beta concentration of the sample to be detected. The inhibition rate of the compound was calculated according to the following formula.
Figure BDA0002625071350000261
S-curve fitting by Graphpad Prism5 and calculation of IC50
The experimental results are as follows:
this example screens 5a, 7a, 10a, 21a, 26a, 28a and 5b for IC by preliminary evaluation of cytotoxicity and activity50The results of the assay (Table 4.2) show that the inhibitory activity of compounds 5a and 5b is 0.38 + -0.07. mu.M and 0.91 + -0.06. mu.M, respectively, which are improved over the positive control BAY 11-7082, and the inhibitory activity of the compound of formula 1 on NLRP3 inflammasome is shown in FIG. 1 and Table 2.
From the above figure, it can be seen that all the final products have activities equal to or superior to those of the positive drugs, and the compounds were screened for further IC based on the results of cytotoxicity and activity evaluation50The results of the measurements are shown in Table 2.
Table 2 evaluation results of activity of some compounds on NLRP3 inflammasome
Figure BDA0002625071350000262
Figure BDA0002625071350000271
The experimental results are expressed as mean values. + -. standard deviation, and the experimental results are more than or equal to two times.
As can be seen, 7a and 5b are two head compounds.
Example 4
This example was evaluated on partial compounds for in vivo NLRP3 inflammatory body inhibitory activity:
c57BL/6 mice were fasted for 12h before the experiment without water deprivation, and the mice were randomly divided into four groups, i.e., blank group, LPS model group, positive control BAY 11-7082 group (20mg/kg), Compound 7a group (20mg/kg), and 5b group (20 mg/kg).
Injecting the prepared drug solution into the abdominal cavity of the mice of the experimental group, wherein the administration volume is 100 mu L/20g, and the blank group and the LPS model group are administered with the same dosage of physiological saline; LPS solution (20mg/kg) was intraperitoneally injected 1h after the administration in a volume of 200. mu.L/20 g, and 2.5h after the administration, blood was taken from the mouse eyes (the mouse was cut with whiskers to prevent hemolysis). The mice are bled and placed in an EP tube, the room temperature is kept for 1h, the rotation speed is 3500rpm under the condition of 4 ℃, the centrifugation is carried out for 10min, and the serum is subpackaged and stored at the temperature of minus 80 ℃.
The results of the experiment are shown in FIG. 2.
The above disclosure is only for the purpose of illustrating the preferred embodiments of the present invention and is not to be construed as limiting the scope of the present invention, therefore, the present invention is not limited by the appended claims.

Claims (10)

1. A compound represented by the formula (1), a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof,
Figure FDA0002625071340000011
wherein n is selected from 0, 1; x is selected from N, O; r1Is selected from differentAn electron-withdrawing or electron-donating substituent; r2Selected from different aliphatic or aromatic substituents.
2. The compound of claim 1, a pharmaceutically acceptable salt, stereoisomer, isotopic label, solvate, polymorph or prodrug thereof, wherein
X is an N or O atom; r1Is 4-methyl; 2-chloro group; 4-chloro group; 4-tert-butyl; one of 4-methoxy;
R2is that
Figure FDA0002625071340000012
Figure FDA0002625071340000013
One kind of (1).
3. A compound represented by formula (2) or formula (3), a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof,
Figure FDA0002625071340000014
wherein R is1Selected from different electron withdrawing or electron donating substituents; r2 is selected from different aliphatic or aromatic substituents, more preferably, R1Is 4-methyl; 2-chloro group; 4-chloro group; 4-tert-butyl; one of 4-methoxy; r2The same as claim 2.
4. The compound of claim 3, a pharmaceutically acceptable salt, stereoisomer, isotopic label, solvate, polymorph or prodrug thereof, wherein the compound is selected from the group consisting of:
Figure FDA0002625071340000021
further, the compound is one of 7a, 10a, 21aa or 5b,
further, the compounds are 7a, 5 b.
5. A process for the preparation of a compound according to any one of claims 1 to 4, or a pharmaceutically acceptable salt, stereoisomer, isotopic label, solvate, polymorph or prodrug thereof, according to the synthetic route shown below:
Figure FDA0002625071340000031
6. the method for preparing a compound according to claim 5, a pharmaceutically acceptable salt, stereoisomer, isotopic label, solvate, polymorph or prodrug thereof, wherein a is provided by the addition of Na2SO3、H2O, reacting for 3 to 6 hours at the temperature of between 50 and 100 ℃,
further preferably, the condition a comprises the addition of Na2SO3、H2O, reacting for 5 hours at the temperature of 80 ℃,
preferably, the b condition comprises the addition of NaOH, MeOH, H2O, reacting at the temperature of 20-50 ℃,
further preferably, the condition b comprises the addition of NaOH, MeOH, H2O, reacting at the temperature of 40 ℃,
preferably, the c condition comprises the addition of Na2SO3、Na2CO3、H2O, reacting for 1 to 5 hours,
further preferably, the c condition comprises addition of Na2SO3、Na2CO3、H2O, reacting for 4 hours,
preferably, the d condition comprises adding dibromopropionic acid and DMF, reacting for 10-15h at 60-90 ℃,
further preferably, the d condition comprises adding dibromopropionic acid and DMF, reacting for 12h at 80 ℃,
preferably, the e conditions comprise addition of amine, TBTU, TEA, anhydrous tetrahydrofuran,
preferably, the condition f comprises the addition of alcohol, PPh3DEAD, anhydrous tetrahydrofuran,
preferably, the g conditions include addition of MeOH, SOCl2
Preferably, the h condition comprises adding TEA and MeOH, reacting for 15-20h at 50-70 ℃,
further preferably, the h conditions comprise addition of TEA, MeOH, and reaction at 60 ℃ for 18 h.
7. Use of a compound according to claims 1-4, a pharmaceutically acceptable salt, a stereoisomer, an isotopic label, a solvate, a polymorph or a prodrug thereof, or a product obtained by the preparation method according to claim 5 or 6, for the preparation of an NLRP3 inflammasome inhibitor.
8. Use of a compound according to claims 1-4, a pharmaceutically acceptable salt, stereoisomer, isotopic label, solvate, polymorph or prodrug thereof, or a product thereof prepared by a process according to claims 5 or 6, for the manufacture of an anti-inflammatory medicament.
9. Use of a compound of claims 1-4, a pharmaceutically acceptable salt, stereoisomer, isotopic label, solvate, polymorph or prodrug thereof, or a product prepared by the method of claim 5 or 6, for the manufacture of a medicament for the treatment of cancer.
10. A pharmaceutical composition comprising a compound of claims 1-4, a pharmaceutically acceptable salt, stereoisomer, isotopic label, solvate, polymorph or prodrug thereof, or a product of a process of claim 5 or 6.
CN202010794558.7A 2020-08-10 2020-08-10 Styrene sulfone NLRP3 inflammatory corpuscle inhibitor and preparation method and application thereof Active CN111793011B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010794558.7A CN111793011B (en) 2020-08-10 2020-08-10 Styrene sulfone NLRP3 inflammatory corpuscle inhibitor and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010794558.7A CN111793011B (en) 2020-08-10 2020-08-10 Styrene sulfone NLRP3 inflammatory corpuscle inhibitor and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN111793011A true CN111793011A (en) 2020-10-20
CN111793011B CN111793011B (en) 2021-08-20

Family

ID=72833725

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010794558.7A Active CN111793011B (en) 2020-08-10 2020-08-10 Styrene sulfone NLRP3 inflammatory corpuscle inhibitor and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN111793011B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4105436A (en) * 1976-10-18 1978-08-08 Kao Soap Co., Ltd. Herbicide
CN103476752A (en) * 2011-02-24 2013-12-25 赛福伦公司 Substituted aromatic sulfur compounds and methods of their use

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4105436A (en) * 1976-10-18 1978-08-08 Kao Soap Co., Ltd. Herbicide
CN103476752A (en) * 2011-02-24 2013-12-25 赛福伦公司 Substituted aromatic sulfur compounds and methods of their use

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BLAYA, SALVADOR 等: "Stereoselective synthesis of β-alkoxy- and β-alkylthioacrylic esters and amides from β-tosylacrylic derivatives", 《TETRAHEDRON》 *
FALCAO, ANA S. 等: "Dipeptidyl Vinyl Sulfone as a Novel Chemical Tool to Inhibit HMGB1/NLRP3-Inflammasome and Inflamma-miRs in Aβ-Mediated Microglial Inflammation", 《ACS CHEMICAL NEUROSCIENCE 》 *
JIANG, JUN 等: "A Mild and Rapid Synthesis of (Z)-β-Sulfonyl Enoates from Sodium Sulfinates and Propargyl Esters", 《CHINESE JOURNAL OF CHEMISTRY》 *
JULIANA, CHRISTINE 等: "Anti-inflammatory Compounds Parthenolide and Bay11-7082 Are Direct Inhibitors of the Inflammasome", 《JOURNAL OF BIOLOGICAL CHEMISTRY》 *
TASSAPORN SAWANGPHON 等: "An Improved Synthesis of Vinyl- and β- Iodovinyl Sulfones by a Molecular Iodine-Mediated One-Pot Iodosulfonation-Dehydroiodination Reaction", 《SYNTHETIC COMMUNICATIONS》 *
WENYI LI 等: "Regioselective and stereoselective sulfonylation of alkynylcarbonyl compounds in water", 《GREEN CHEMISTRY》 *

Also Published As

Publication number Publication date
CN111793011B (en) 2021-08-20

Similar Documents

Publication Publication Date Title
AU2020260400B2 (en) Human plasma kallikrein inhibitors
TW204348B (en)
CN110719902A (en) SSAO inhibitors
UA75891C2 (en) Pharmaceutically active sulfonamides derivatives having either lipophilic fragments and those capable to ionize as inhibitors of protein- jun-kinases, a pharmaceutical composition, a method for obtaining these compounds (variants) and intermediary compounds
KR20010101675A (en) 2h-phthalazin-1-one derivatives and drugs comprising these derivatives as the active ingredient
JP2014525443A (en) Kynurenin-3-monooxygenase inhibitors, pharmaceutical compositions, and methods of use thereof
CN105919987A (en) N-acyloxysulfonamide And N-hydroxy-n-acylsulfonamide Derivatives And Use Thereof
PT1320526E (en) Pharmaceutically active benzsulfonamide derivatives as inhibitors of protein junkinases
CN115304583B (en) 5-pyridine-1H-indazole compound for targeted inhibition of CLK2 and application thereof
CN105658641B (en) Benzazole amide derivatives, its preparation method and its application in medicine
CN110078663B (en) Sulfonamide compound with tetrahydroquinoline as parent nucleus and preparation method and application thereof
CN103524559B (en) Ester derivant of polysubstituted 4-methylamino benzenecarboximidamide and its production and use
EP2040690A1 (en) Inhibitors of cxcr2
CN104910894B (en) Benzimidazole hERG potassium ion channel small-molecular fluorescent probe and preparation method and applications thereof
JP4975641B2 (en) Benzothiophene-2-carbonylguanidine derivative, process for producing the same and pharmaceutical composition containing the derivative
CN111793011B (en) Styrene sulfone NLRP3 inflammatory corpuscle inhibitor and preparation method and application thereof
CN103012381B (en) Benzofuran compound, preparation method thereof and application of benzofuran compound in preparation of antiarrhythmic drugs
US6727266B2 (en) Substituted tryptophan derivatives
CN103396351B (en) Pyrrolidines Bcl-2 protein micromolecular inhibitor compound and preparation, pharmaceutical composition and pharmaceutical applications
CN104016944B (en) N-(2-(amine methyl) phenyl) thiazole-4-carboxamide derivative and preparation method thereof and application
CN104817519B (en) The derivant of one class CA-4, its preparation method and medical usage thereof
CN100519544C (en) Furancarbonylguanidine derivatives, their preparation and pharmaceutical compositions containing them
Bui et al. Synthesis of sulfonylurea derivatives and their α-glucosidase inhibitory activity
KR100820173B1 (en) 4-methylimidazol-5-ylcarbonylguanidine derivatives, pharmaceutically acceptable salts thereof, preparation method, and pharmaceutical compositions for the prevention and treatment of the ischemic heart diseases containing the same as an active ingredient
KR101857714B1 (en) Novel amide compounds and use thereof

Legal Events

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