CN112321620B - Keap1-Nrf2 PPI inhibitor prodrug, preparation method and application - Google Patents

Keap1-Nrf2 PPI inhibitor prodrug, preparation method and application Download PDF

Info

Publication number
CN112321620B
CN112321620B CN202011244101.5A CN202011244101A CN112321620B CN 112321620 B CN112321620 B CN 112321620B CN 202011244101 A CN202011244101 A CN 202011244101A CN 112321620 B CN112321620 B CN 112321620B
Authority
CN
China
Prior art keywords
prodrug
compound
nrf2
keap1
stirring
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
CN202011244101.5A
Other languages
Chinese (zh)
Other versions
CN112321620A (en
Inventor
尤启冬
姜正羽
王小鹿
吕逸菲
张贤
陆朦辰
郭小可
徐晓莉
王磊
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
China Pharmaceutical University
Original Assignee
China Pharmaceutical University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by China Pharmaceutical University filed Critical China Pharmaceutical University
Priority to CN202011244101.5A priority Critical patent/CN112321620B/en
Publication of CN112321620A publication Critical patent/CN112321620A/en
Application granted granted Critical
Publication of CN112321620B publication Critical patent/CN112321620B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic Table
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • 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]
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09KMATERIALS FOR MISCELLANEOUS APPLICATIONS, NOT PROVIDED FOR ELSEWHERE
    • C09K11/00Luminescent, e.g. electroluminescent, chemiluminescent materials
    • C09K11/06Luminescent, e.g. electroluminescent, chemiluminescent materials containing organic luminescent materials
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09KMATERIALS FOR MISCELLANEOUS APPLICATIONS, NOT PROVIDED FOR ELSEWHERE
    • C09K2211/00Chemical nature of organic luminescent or tenebrescent compounds
    • C09K2211/10Non-macromolecular compounds
    • C09K2211/1003Carbocyclic compounds
    • C09K2211/1014Carbocyclic compounds bridged by heteroatoms, e.g. N, P, Si or B
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09KMATERIALS FOR MISCELLANEOUS APPLICATIONS, NOT PROVIDED FOR ELSEWHERE
    • C09K2211/00Chemical nature of organic luminescent or tenebrescent compounds
    • C09K2211/10Non-macromolecular compounds
    • C09K2211/1018Heterocyclic compounds
    • C09K2211/1025Heterocyclic compounds characterised by ligands
    • C09K2211/1088Heterocyclic compounds characterised by ligands containing oxygen as the only heteroatom
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09KMATERIALS FOR MISCELLANEOUS APPLICATIONS, NOT PROVIDED FOR ELSEWHERE
    • C09K2211/00Chemical nature of organic luminescent or tenebrescent compounds
    • C09K2211/10Non-macromolecular compounds
    • C09K2211/1018Heterocyclic compounds
    • C09K2211/1025Heterocyclic compounds characterised by ligands
    • C09K2211/1096Heterocyclic compounds characterised by ligands containing other heteroatoms

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Materials Engineering (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a Keap1-Nrf2 PPI inhibitor prodrug, a preparation method and application thereof. The prodrug P-168 provided by the invention seals polar carboxylic acid groups of an active compound 168, improves fat solubility, and effectively improves membrane permeability and pharmacy; the prodrug P-168 is reduced under the condition of high ROS, a pharmacophore 168 and a fluorescent group coumarin are released, the expression of Nrf2 and downstream genes thereof is activated, the anti-inflammatory activity is exerted, and the fluorescence is released to realize visual monitoring.

Description

Keap1-Nrf2 PPI inhibitor prodrug, preparation method and application
Technical Field
The invention belongs to the field of medicinal chemistry, and particularly relates to a Keap1-Nrf2 PPI inhibitor prodrug, a preparation method and application thereof.
Background
One of the most important protection mechanisms of cells is the Keap1-Nrf2-ARE signaling pathway, which regulates oxidative stress in vivo and maintains redox balance. The signaling pathway regulates the expression of over 100 genes and functions associated with oxidative stress and cell survival, including direct antioxidant proteins, phase I and II electrophilic detoxification enzymes, free radical metabolism, inhibition of inflammation, glutathione homeostasis, proteasome function, recognition of DNA damage, and expression of various related growth factors and transcription factors. The discovery of Keap1-Nrf2 PPI inhibitors has become a promising strategy for developing novel molecules against a variety of stresses.
The subject group has found the first compound 168 with nanomolar inhibitory activity against Keap1-Nrf2 PPI, having an EC of50It was 28.6 nM. However, the compound contains polar carboxylic acid group, so that the fat solubility is poor, and the membrane permeability and the drug property are influenced.
Disclosure of Invention
The invention aims to overcome the defects of the prior art and provides a Keap1-Nrf2 PPI inhibitor prodrug, a preparation method and application thereof. The above purpose of the invention is realized by the following technical scheme:
a Keap1-Nrf2 PPI inhibitor prodrug having the chemical structure:
Figure BDA0002769374250000011
the preparation method of the Keap1-Nrf2 PPI inhibitor prodrug comprises the following steps:
Figure BDA0002769374250000021
the reagents and reaction conditions for each step were as follows:
(a) refluxing and stirring the compound 5 and a proper amount of sodium propionate, propionic anhydride and piperidine at 120 ℃ for 6 hours;
(b) CCl to Compound 64Adding a proper amount of NBS and AIBN into the solution, carrying out thermal reflux reaction for 8 hours, and removing the solvent under reduced pressure; adding a proper amount of NaOAc and acetic acid into the obtained residue, and carrying out a thermal reflux reaction for 12 hours; subsequently, an appropriate amount of hydrochloric acid was added to the hot reaction mixture, the reaction was continued for 30 minutes, and stirred at room temperature overnight;
(c) dissolving the compound 7 and an appropriate amount of pyridine in dichloromethane, stirring in an ice bath for 10 minutes, slowly adding an appropriate amount of trifluoromethanesulfonic anhydride, and stirring the reaction mixture for 2 hours;
(d) adding an appropriate amount of compound 8 to stirred bis (pinacolato) diboron, Pd (dppf) Cl2·CH2Cl2And KOA in dioxane, the reaction mixture was refluxed under nitrogen for 8 hours; after completion of the reaction, the solvent was removed under reduced pressure, and the reaction mixture was diluted with EtOAc, washed with brine, anhydrous Na2SO4Drying, filtering, and removing the solvent under reduced pressure; the crude product was dissolved in MeOH and the appropriate amount of NaBH added4Stirring and reacting for 1 hour at normal temperature;
(e) dissolving the compound 168 in dichloromethane, adding a proper amount of DMAP and DCC, stirring for half an hour at normal temperature, adding a proper amount of a compound 9, and stirring overnight;
wherein the chemical structural formula of the compound 168 in the step (e) is:
Figure BDA0002769374250000031
the application of the Keap1-Nrf2 PPI inhibitor prodrug in preparing anti-inflammatory drugs.
The application of the Keap1-Nrf2 PPI inhibitor prodrug in preparing the medicines for resisting inflammation and monitoring the inflammation relieving degree.
Has the advantages that:
1. the prodrug P-168 provided by the invention seals polar carboxylic acid groups of an active compound 168, improves fat solubility, and effectively improves membrane permeability and pharmacy;
2. the prodrug P-168 provided by the invention contains three functional fragments: (1) active compound 168, a Keap1-Nrf2 PPI inhibitor with superior activity; (2) a coumarin fluorophore; (3) an ROS activatable phenylboronate group; the compound is reduced under the condition of high ROS, a pharmacophore 168 and a fluorescent group coumarin are released, the expression of Nrf2 and downstream genes thereof is activated, the anti-inflammatory activity is exerted, meanwhile, fluorescence is released to realize visual monitoring, and the reduction of fluorescence intensity indicates that inflammation is relieved;
3. the prodrug P-168 provided by the invention is coupled with H2O2Is more selective than other biologically relevant ROS (e.g., hydroxyl radical, t-butoxy radical, superoxide, hypochlorite anion, and t-butyl hydroperoxide), and H2O2Is the most typical ROS in inflammation, thus ensuring that the prodrug P-168 is not easily interfered with for the selectivity of the visible monitoring of inflammation.
Drawings
FIG. 1A is H2O2Schematic HPLC peak profile of the activated prodrug P-168 to release prodrug and coumarin group; 1B is a time gradient technical release curve detected by HPLC; 1C is the fluorescence spectrum of the prodrug P-168 under the time gradient (0-60min), and the fluorescence intensity at the wavelength of 465nm is taken to draw a time gradient release curve; 1D is H2O2The fluorescence spectrum of prodrug P-168 under the concentration trend (0-10mM) is taken to plot the fluorescence intensity at 465nm as H2O2Concentration gradient release profile.
FIG. 2A is a fluorescence spectrum of prodrug P-168 detected in the presence of various ROS; 2B is the calculated percentage of prodrug P-168 released in the presence of each ROS.
FIG. 3A shows the effect of prodrug P-168 and prodrug 168 on H2O2LC-MS analysis after pretreatment of RAW264.7 cells; 3B is the prodrug P-168 acting on H2O2The fluorescence intensity (fold) after pretreatment of RAW264.7 cells was varied with time gradient; 3C acting on H as prodrug P-1682O2The fluorescence intensity (fold) after pretreatment of RAW264.7 cells was varied with concentration gradient.
FIG. 4 is a comparison of the ARE inducing activity and selectivity of LPS activation, with 4A being the ARE inducing activity of prodrugs P-168 and 168 in HepG2-ARE-C8 cells; 4B is the ARE induction activity of P-168 in RAW264.7 cells under different concentrations of LPS; 4C is ARE induction activity of adding P-168 with different concentrations after LPS treatment in RAW264.7 cells; d is the time trend of ARE induction activity by the addition of P-168 after LPS treatment in RAW264.7 cells.
FIGS. 5A-D are real-time quantitative PCR analyses of Nrf2(A), HO-1(B), NQO1(C) and GCLM (D) in RAW264.7 cells; 5E is the determination of GSH/GSSG levels in RAW264.7 cells; 5F is the Western blot analysis of Nrf2 downstream protein and inflammatory factors IL-1 beta and IL-6 in RAW264.7 cells.
FIGS. 6A-B are real-time quantitative PCR analyses of IL-1. beta. (A) and IL-6(B) in RAW264.7 cells; C-F the concentrations of IL-1 β (D), IL-6(E), TNF- α (F) and NO (G) in the culture supernatants of RAW264.7 cells were investigated for ELISA assays.
FIG. 7 is the chemical structural formula of prodrug P-168.
Detailed Description
The following detailed description of the present invention is provided with reference to the accompanying drawings, which are not intended to limit the scope of the present invention.
Firstly, synthesis and structure confirmation of target compound
1.1 Experimental instruments and general rules
All chemicals purchased from commercial suppliers were used as received, unless otherwise indicated. All solvents were reagent grade, purified and dried by standard methods if necessary. All reactions were monitored by Thin Layer Chromatography (TLC) on a visual silica gel plate (GF-254). Melting points were determined on a Mel-TEMP II melting point apparatus without calibration. Determined on a Bruker Avance-300 Instrument1HNMR and13CNMR spectroscopy. Using CDCl3Or DMSO-d6 as solvent, and the chemical shifts (δ) reported are parts per million (ppm) of Tetramethylsilane (TMS). HR-MS spectra were recorded on a Water Q-Tof micro mass spectrometer. Flash column chromatography was performed on 100-200 mesh silica gel to obtain chromatographically and spectroscopically pure compounds. The purity of the compounds was verified by HPLC studies performed on an Agilent C18(4.6mm X150 mm, 3.5 μm) column using a mixture of solvents methanol/water at a flow rate of 0.5mL/min and monitored by UV absorption at 254 nm.
1.2 Synthesis and characterization of Compounds
The synthetic route is as follows:
Figure BDA0002769374250000051
Reagents and conditions:(a)NH2OH·HCl,ethanol,MeOH,60℃,2h;(b)Pd/C,H2,rt,4h;(c)4-Methoxybenzenesulfonyl chloride,toluene,pyridine,100℃,2h;(d)DMF,K2CO3,ethyl bromoacetate,rt,3h;(e)NaOH,MeOH,H2O,2h.
Figure BDA0002769374250000052
Reagents and conditions:(a)propionic anhydride,piperidine,reflux,6h;(b)(1)NBS,CCl4,reflux,8h;(2)NaOAc,AcOH,reflux,12h;(3)2N HCl,reflux,rt,overnight;(c)Tf2O,Py,DCM,2h;(d)(1)Bis(pinacolatol)diboron,PdCl2dppf,KOAc,dioxane,reflux,8h;(2)NaBH4,MeOH,rt,1h;(e)DCC,DMAP,DCM,rt,overnight.
synthesis of 4-nitronaphthalen-1-amine (2):
alpha-nitronaphthalene (1) (3g, 11.5mmol) and hydroxylamine hydrochloride (5g, 72mmol) were dissolved in 150ml 95% ethanol, heated to 60 ℃ and stirred for 1 hour. A solution of 10g of potassium hydroxide in 60ml of methanol was gradually added with stirring. Stirring was continued for one hour and the warm solution was poured slowly into ice water. The solid was filtered and washed with water. Recrystallization from acetonitrile gave compound 2 as a yellow solid in 55% yield. m.p. 191-192 ℃.1H-NMR(300MHz,DMSO-d6):δ8.89(d,1H,J=8.9),8.37(d,1H,J=9.0),8.30(d,1H,J=8.9),7.74(dd,1H,J=6.9,1.0),7.52(dd,1H,J=6.9,1.0),7.11(s,1H),6.67(d,1H,J=9.0).HRMS(ESI):189.0659[M+H]+.
Synthesis of N, N' - (naphthalene-1, 4-diyl) bis (4-methoxybenzenesulfonamide) (3):
to a solution of 4-nitro-1-naphthylamine (2) (1.81g, 10mmol) in THF was added Pd/C. The reaction mixture was stirred under hydrogen for 4 hours. The solution was filtered through celite to remove the catalyst. The filtrate was concentrated under reduced pressure to give a pale yellow oil. The crude naphthalene-1, 4-diamine was not further purified. 4-Methoxybenzenesulfonyl chloride (3.75g, 22mmol) and pyridine (2.37g, 30mmol) were added to a solution of naphthalene-1, 4-diamine in toluene. The reaction mixture was stirred at 100 ℃ for 2 hours under nitrogen. After cooling to room temperature, the reaction mixture was diluted by pouring 20mL of petroleum ether. After filtration, the solid was collected and recrystallized from acetonitrile to give compound 3 as a grey solid in 54% yield. m.p.: 217 ℃ to 219 ℃.1HNMR(300MHz,DMSO-d6):δ10.04(s,2H),7.95(dd,2H,J=6.5±3.2),7.55(d,4H,J=8.9),7.39(dd,2H,J=6.5±3.2),7.00(m,6H),3.77(s,6H).HRMS(ESI):521.1026[M+Na]+.
Synthesis of diethyl 2, 2' - (naphthalene-1, 4-diylbis (((4-methoxyphenyl) sulfonyl) aza) diacetate (4):
to a solution of compound 3(997mg, 2mmol) in DMF (5ml) was added K2CO3(830mg, 6mmol) followed by ethyl bromoacetate (836mg, 5 mmol). After stirring at room temperature for 3 hours, the reaction mixture was then poured into 30mL of water and adjusted to pH 5 with 2M hydrochloric acid. Filtration gave the crude product which was recrystallized from ethyl acetate/n-hexane to give a light pink solid 4 in 64% yield. m.p.: 71-73 ℃.1H-NMR(300MHz,DMSO-d6):δ8.30(m,1H),8.18(m,1H),7.58(m,6H),7.11(m,4H),7.04(s,1H),6.86(s,1H),4.48(m,4H),4.01(m,4H),3.88(s,3H),3.84(s,3H),1.06(m,6H).HRMS(ESI):693.1558[M+Na]+.
Synthesis of 2' - (naphthalene-1, 4-diylbis (((4-methoxyphenyl) sulfonyl) azenediyl)) diacetic acid (168):
compound 4(670mg, 1mmol) was added to NaOH (4g) in MeOH/H2O (20/20ml) solution. The reaction mixture was heated in an oil bath at 65 ℃ for 2 hours, then adjusted to pH 2 with 2M hydrochloric acid and diluted with 75mL of water. The precipitate was filtered, washed with 5X 10mL of water and dried to give 168 as a white solid in 64% yield. m.p.: 236 ℃ and 238 ℃.1H-NMR(300MHz,DMSO-d6):δ12.8(s,2H),8.30(m,1H),8.18(m,1H),7.58(m,6H),7.16(d,4H,J=9.5),7.05(s,1H),6.89(s,1H),4.38(m,4H),3.88(s,3H),3.84(s,3H);HRMS(ESI):637.0934[M+Na]+.
Synthesis of 3-methyl-2-oxo-2H-chromen-7-yl propionate (6):
a mixture of 2, 4-dihydroxybenzaldehyde (3.0g, 21.72mmol), sodium propionate (4.59g, 47.78mmol), propionic anhydride (7.63g, 58.64mmol) and piperidine (0.4ml) was stirred at 120 ℃ under reflux for 6 hours. The reaction solution was poured into ice water, the precipitate was collected by filtration and dissolved in EtOAc, and the organic layer was washed with water and 1N HCl, followed by anhydrous Na2SO4And (5) drying. Subjecting the residue to column chromatography to obtain compoundProduct 6 was a white crystal with a yield of 50%. m.p.: 110 ℃ and 112 ℃.1H NMR(300MHz,CDCl3)δ7.50(s,1H),7.40(d,1H,J=8Hz),7.08(d,1H,J=2Hz),7.00(dd,1H,J=8.2Hz),2.64-2.59(2H),2.20(s,3H),1.29-1.25(3H);HRMS(ESI):233.0808[M+H]+.
Synthesis of 7-hydroxy-2-oxo-2H-chromene-3-carbaldehyde (7):
to compound 6(2.7g, 11.9mmol) in 75mL CCl4NBS (5.31g, 29mmol) and a catalytic amount of AIBN were added to the solution in (1), and the mixture was refluxed. After 8 hours of reaction, the solvent was removed under reduced pressure. To the resulting residue were added NaOAc (8.78g, 107mmol) and acetic acid (80mL), and the mixture was heated under reflux for 12 hours. Subsequently, 2N HCl (60mL) was added to the hot reaction mixture and the reaction was allowed to continue for 30 minutes and stirred at room temperature overnight. The reaction mixture was then evaporated to dryness and the residue was directly purified by column chromatography to give compound 7 as a yellow powder in 50% yield. m.p.: 118 ℃ and 120 ℃.1HNMR(300MHz,DMSO-d6)δ10.86(s,1H),8.20(s,1H),7.89(d,1H,J=8.2Hz),7.23(dd,1H,J=8.2,2Hz),7.21(d,1H,J=2Hz)。HRMS(ESI):191.0339[M+H]+.
Synthesis of 3-formyl-2-oxo-2H-chromen-7-yl trifluoromethanesulfonate (8):
compound 7(100mg, 0.52mmol) and pyridine (0.06mL, 0.73mmol) were dissolved in 20mL of dichloromethane and stirred in an ice bath for 10 min. Trifluoromethanesulfonic anhydride (106. mu.L, 0.63mmol) was added slowly. The reaction mixture was stirred for 2 hours. After completion of the reaction, the reaction mixture was diluted with dichloromethane, washed twice with brine, over anhydrous Na2SO4Dried and then filtered, and the solvent removed under reduced pressure. The crude product was purified by silica gel column chromatography. Compound 8 was obtained as a white solid in 71% yield. m.p.: 130 ℃ and 131 ℃.1HNMR(300MHz,CDCl3):δ7.24-7.31(m,1H),7.34(d,J=2.0Hz,1H),7.80(d,J=8.6Hz,1H),8.40(s,1H),10.23(s,1H)。HRMS(ESI):322.9837[M+H]+.
Synthesis of 3- (hydroxymethyl) -7- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2H-pyran-2-one (9):
compound 8(100mg, 0.31mmol) is added to stirred bis (pinacolato) diboron (95mg, 0.37mmol), Pd (dppf) Cl2·CH2Cl2(12mg, 0.01mmol) and KOAc (91mg, 0.93mmol) in dioxane (5 mL). The reaction mixture was refluxed for 8 hours under nitrogen blanket. After completion of the reaction, the solvent was removed under reduced pressure, and the reaction mixture was diluted with EtOAc, washed twice with brine, over anhydrous Na2SO4Dry, filter, and remove the solvent under reduced pressure. The crude product was not further purified. The crude product (180mg) was dissolved in 10mL MeOH, then NaBH was added4(38mg, 1.02 mmol). The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was quenched with saturated ammonium chloride solution, then diluted with EtOAc, washed twice with brine, over anhydrous Na2SO4Drying, filtering and removing the solvent to obtain the compound 9. The crude product was not purified.
Synthesis of target compound P-168 (chemical structural formula shown in fig. 7):
compound 168(0.5g, 0.813mmol) was dissolved in dichloromethane, DMAP (0.15g, 1.22mmol) and DCC (0.25g, 1.22mmol) were added, and after stirring at room temperature for half an hour, compound 9(0.27g, 0.89mmol) was added and stirring was continued overnight. After the reaction was completed, the by-product solid was removed by filtration, the filtrate was washed with brine, and the organic phase was collected and dried. The compound P-168 is obtained by column chromatography as a yellow solid with a yield of 46%. m.p.: 210 ℃ and 211 ℃.1HNMR(300MHz,DMSO-d6)δ7.91–7.84(m,2H),7.70–7.65(m,2H),7.58–7.52(m,2H),7.52–7.44(m,3H),7.01–6.95(m,2H),5.14(d,J=0.9Hz,2H),4.67(s,1H),3.78(s,2H),1.24(s,9H)。HRMS(ESI):1183.3541[M+H]+.
Release study of prodrug P-168
2.1 Experimental methods
2.1.1 HPLC methods study prodrug Compound Release in buffer
Prodrug compound was dissolved in DMSO to make a final concentration of 10mM stock solution, stored at-20 ℃. The prodrugsThe compound stock solution (10mM) was added to Phosphate Buffered Saline (PBS) (10mM, pH 7.4) to a final concentration of 50. mu.M, followed by addition of H2O2After vortex mixing, the solution was incubated at 37 ℃ and repeated three times. Samples were taken at each time point and analyzed by HPLC injection, and peak areas were recorded to calculate the percent of compound. Using Agilent 1260HPLC and DAD detector, chromatographic conditions: agilent C18 column (4.6X 150mm, 3.5 μm); mobile phase: methanol: water 7: 3; flow rate: 0.5 mL/min. A standard curve of percent compound release versus concentration is plotted and the release rate is calculated from the standard curve.
2.1.2 fluorescent Spectroscopy study of prodrug Compound Release
In PBS buffer: prodrug compound stock solution (10mM) was added to PBS (10mM, pH 7.4) to a final concentration of 0.5. mu.M, added to a 96-well plate, and then added to concentration gradient H2O2Three multiple wells were set, incubated at 37 ℃ for 30min and then subjected to fluorescence detection using a Molecular Devices SpectraMax i3 multifunctional microplate reader. In the cell: RAW264.7 cells were seeded into 96-well plates and H was added2O2The treatment was carried out for 12 hours. Control group was not added H2O2The other culture conditions were the same. After 12h, the medium was discarded and washed three times with PBS, and prodrug compound solution (0.5. mu.M) was added and incubation continued at 37 ℃ for 30 min. Fluorescence detection was performed using a Molecular Devices SpectraMax i3 multifunctional microplate reader.
2.1.3 study of prodrug Compound reactivity with other related ROS
Superoxide (O)2 -) The concentration used was 5mM and the other reactive ROS was 100. mu.M. Solid KO for superoxide2(ii) a Hydrogen peroxide (H)2O2) Tert-butyl hydroperoxide (TBHP) and hypochlorite ion (OCl)-) Commercial 30%, 70% and 5% aqueous solutions were used; by Fe2+(1mM) with TBHP (100. mu.M) or H2O2(100. mu.M) reaction to prepare tert-butoxy (. O) respectivelytBu) and hydroxyl (. OH). The prodrug compound was incubated with each of the above solutions in PBS for 30min, and then samples were taken for HPLC analysis and fluorescence spectroscopy analysis.
2.1.4LC-MS method for determining the intracellular Release of prodrug Compounds
RAW264.7 cells were plated in large dishes overnight and cultured with H2O2Cells were stimulated (10mM) for 12 hours, then washed with fresh medium and treatment with 10. mu.M prodrug compound was continued for 12 hours. After the administration was completed, the cells were washed with fresh medium, 1mL of methanol was added to the cell pellet, the mixture was mixed well by vortexing, the polymer component was removed by high-speed centrifugation for 10 minutes, and the supernatant was subjected to LC-MS analysis and stored at-80 ℃. Analysis was performed using an LC-MS/MS spectrometer (6410Triple Quad LC/MS/MS, Agilent Technologies, Willington) equipped with a chromatography column (Agilent C18 column 4.6X 150mm, 3.5 μm).
2.2 results of the experiment
2.2.1 prodrug Compound P-168 in buffer by H2O2Activation
First, to confirm H2O2The phenylboronate group can be activated and release the original drug, and the release behavior of the prodrug is researched and a release curve is drawn by a High Performance Liquid Chromatography (HPLC) method. The prodrug was incubated with Phosphate Buffered Saline (PBS) and H was added2O2Activation, the solution was analyzed by HPLC injection to observe the appearance of the prodrug and prodrug peaks. As shown in fig. 1A and 1B, consumption of prodrug P-168 and production of the active compound can be observed, and the release profile is time dependent, with time the prodrug is activated and after 60 minutes the prodrug is completely released.
In addition, the application also researches the release of the prodrug by adopting fluorescence spectroscopy to verify H2O2The boronic ester moiety in the prodrug can be activated and thus release the coumarin fluorophore. First, the prodrug pair H was examined2O2Time responsiveness of (C), as shown in FIG. 1C, when not activated (H)2O2Concentration 0mM), the prodrug showed weak fluorescence centered at 460nm, in PBS buffer via H2O2The fluorescence after the treatment is rapidly and smoothly recovered, the maximum wavelength is slightly red-shifted, and the fluorescence intensity at the position of lambda max being 465nm is obviously enhanced. The fluorescence intensity at λ max of 465nm is also determined by the time gradientGradually enhanced and released fluorescence and original drug with time dependence, indicating that the prodrug has H2O2Has good responsiveness. Subsequently, H was investigated2O2Effect of concentration on prodrug release behavior, as shown in FIG. 1D, with H2O2The concentration increased and the fluorescence intensity at λ max 465nm increased progressively, indicating that the boronic ester moiety of the prodrug is susceptible to being substituted by H2O2Activating to release coumarin fluorescence and original medicine in a concentration-dependent mode.
2.2.2 prodrug pairs H2O2Is more selective than other biologically relevant ROS species
To assess the potential for additional interference, prodrug P-168 was reacted with other classes of ROS, including hydroxyl radicals, t-butoxy radicals, superoxide, hypochlorite anion, and t-butyl hydroperoxide. In and H2O2Other ROS were tested under the same reaction conditions and fluorescence intensity was monitored. When the prodrug is reacted with H2O2Upon reaction, an increase in fluorescence intensity was observed, whereas no significant change in fluorescence intensity was observed upon addition of the other ROS (see fig. 2A), indicating that the prodrug can react with H2O2And (4) carrying out selective reaction.
2.2.3 prodrug P-168 is substituted with H in cells2O2Activated release of prodrug and coumarin fluorescence
LC-MS analysis was used to further confirm that prodrug P-168 was able to be activated in cells to produce the active compound. Selecting mouse mononuclear macrophage leukemia cell RAW264.7 cell and using H2O2Pretreatment for 12 hours, then replacing the medium with fresh medium. Prodrug P-168 is added to the filtrate H2O2Treated cells and untreated control cells were incubated and cell lysates were analyzed by LC-MS. As shown in fig. 3A, there is a mass spectrum peak of the prodrug compound in the activated cells, which is the same as the standard prodrug peak in the unactivated cells, indicating that the prodrug compound is easily taken up by the cells and activated by ROS to produce the prodrug. In addition, H was performed on RAW264.7 cells2O2Pretreatment followed by prodrug administration and detection of intracellular coumarin fluorescence generation, prodrugs P-1, as shown in FIGS. 3B and 3C68 can release coumarin fluorescence in a concentration-dependent and time-dependent manner, and the trend of the original drug compound is not existed.
Third, evaluation of biological Effect
3.1 Experimental methods
3.1.1 ARE luciferase reporter Gene assays
HepG2-ARE-C8 cell culture at 4X 104Cell/well density was seeded into 96-well plates and incubated overnight. Concentration gradients of test prodrug compound were formulated with fresh medium and added to cells and incubated for 12 hours. DMSO was negative control, luciferase cell lysis reagent was blank. After dosing was complete, the medium was discarded and 100 μ L of pre-cooled PBS was added to each well to wash the cells, which was repeated three times. PBS was removed, 30. mu.L luciferase cell lysis reagent was added, cells were harvested after 15 minutes of ice lysis and centrifuged, and 20. mu.L of supernatant was collected by Thermo Scientific Luminoskan according to protocol supplied by PromegaTMAnd (4) performing luciferase activity determination on the Ascent, repeating the determination three times in parallel, and calculating the determination data and the ratio of the control group to obtain the induction multiple.
3.1.2 real-time fluorescent quantitative PCR
RNA extraction: after dosing RAW264.7 cells, the old medium was discarded and washed three times with PBS. After trypsinization, the cells were collected into 1.5mL EP tubes and centrifuged at 3000rpm for 5 minutes. The supernatant was discarded, 1ml of PBS was added to resuspend the cells, centrifuged at 3000rpm for 5 minutes, and the supernatant was discarded. 1ml of LTrizol was added to each tube, and the cells were blown off and lysed on ice for 15 minutes. Add 200. mu.L of chloroform to each tube, after standing for 10 minutes, shake vigorously for 15 seconds, stand at room temperature for 2-3 minutes, and centrifuge at 12000rpm for 15 minutes at 4 ℃. Absorbing 500 mu L of upper-layer water phase, adding isopropanol with the same volume, uniformly mixing, standing at room temperature for 10 minutes, centrifuging at 4 ℃ and 12000rpm for 15 minutes, and removing supernatant. Adding 75% ethanol (prepared with DEPC water), and centrifuging at 7500rpm for 3min at 4 deg.C. Alcohol was evaporated on ice, 20. mu.L of DEPC water was added to each tube to dissolve RNA, and the mixture was mixed well to quantify nucleic acid. Reverse transcription: according to
Figure BDA0002769374250000101
III RT Supermix for qPCR (+ gDNAwiper) instructions. Amplification: in the qPCR reactionmu.L DEPC water, 0.5. mu.L forward primer, 0.5. mu.L reverse primer, 2. mu.L cDNA, 10. mu.L 2 xchamQ SYBR Master Mix (High ROX premix) were added to the plate in this order. In Thermo StepOne&The amplification and quantification process is completed by StepOneNus Real-Time PCR Systems. The primers used for qRT-PCR were as follows: nrf2 (forward primer: AACCACCCTGAAAGCACGC; reverse primer: TGAAATGCCGGAGTCAGAATC); HO-1 (forward primer: ATGGCCTCCCTGTACCACATC, reverse primer: TGGTGCGCTCAATCTCCTCCT); NQO1 (forward primer: CGCAGACCTTGTGATATTCCAG, reverse primer: CGTTTCTTCCATCCTTCCAGG); GCLM (forward primer: TTGGAGTTGCACAGCTGGATTC, reverse primer: TGGTTTTACCTGTGCCCACTG).
3.1.3 Western immunoblotting
Protein sample preparation was performed after administration of RAW264.7 cells, the old medium was discarded and washed three times with PBS, 1mL of trypsinized cells were added to each dish, collected into 1.5mL EP tubes, and centrifuged at 3000rpm for 5 min. The supernatant was discarded, 1ml of LPBS was added to resuspend the cells, and the cells were centrifuged at 3000rpm for 5 min. The supernatant was discarded, 100. mu.L of a lysate (containing a proteasome inhibitor) was added to each tube, lysed on ice for 45 minutes, centrifuged at 12000rpm for 15 minutes, collected, and 1. mu.L of the supernatant was diluted ten-fold with up water for BCA quantification. Adding one fourth of 5Xprotein loading buffer into the residual supernatant, mixing, and boiling for 10 min. SDS-PAGE: after being aligned, the dry and clean glass plate is placed into a clamp for clamping, and after being vertically clamped on a frame, glue is poured. The glue is poured until the water is close to the center line of the green band without generating bubbles in the glue pouring process. Sealing with isopropanol, removing isopropanol after gelation, washing with deionized water, filling with concentrated gel, and inserting into comb. After the concentrated gel is solidified, the comb is carefully pulled out. The loading was done at a total protein of 100. mu.g per well, constant voltage of 60V until Marker separation, and increasing voltage to 120V until completion. Film transfer: the PVDF membrane is used for membrane conversion. The process of assembling the transfer film is carried out in a wet transfer buffer. The whole process must be ensured to be bubble-free, and the film is transferred in an ice bath. The film transfer condition is 300mA,2 h. And (3) sealing: after the membrane transfer is finished, taking out the PVDF membrane, dyeing the PVDF membrane for 30s, and washing away excessive Lichun by deionized water. The region corresponding to the molecular weight is reduced to bands, deionized and washed clean ponceau. The strips were soaked in 5% skim milk (TBST configuration) and shaken slowly for 1 hour. Primary antibody hybridization: after blocking, the PVDF membrane was placed in a hybridization bag, an appropriate amount of primary antibody solution was added, sealed, and slowly shaken overnight on a shaker at 4 ℃. And (3) hybridization of a second antibody: after the primary antibody incubation was complete, the PVDF membrane was removed and washed three times with TBST, 10min each time. Then, the PVDF membrane was soaked in the prepared corresponding fluorescent secondary antibody (secondary antibody: blocking solution: 1:20000), and incubated on a shaker for 1 hour slowly in the absence of light. And (3) detection: after the secondary antibody incubation was completed, the PVDF membrane was removed and washed three times with TBST for 10min each time. Detection of scanned membrane Imaging was performed by the Bio-Rad ChemiDoc XRS + Imaging Workstation and Tanon 5200 Multi Imaging Workstation. anti-Nrf 2(ab62352), anti-IL-1 β (ab45692) antibodies were purchased from Abcam Technology. anti-HO-1 (SC-136960) and anti-NQO 1(SC-271116) antibodies were purchased from Santa Cruz Biotechnology. Anti- β -Actin (60008-1-lg), anti-GCLM (14241-1-AP) antibody was purchased from Proteitech Group.
3.1.4 detection of IL-1 β, IL-18, IL-6, TNF- α and NO
IL-1 β (Mouse IL-1 β ELISA kit, EK0394, Boster), IL-18(Mouse IL-18 ELISA kit, EMC011, Neobioscience), IL-6(Mouse IL-6ELISA kit EK0411, Boster), TNF- α (Mouse TNF- α ELISA kit, EK0527, Boster) and NO (nitrate/nitrite assay kit, S0023, Beyotime) were all detected using commercially available kits according to the manufacturer' S instructions.
3.2 results of the experiment
3.2.1 LPS-induced prodrug activation of the Keap1-Nrf2-ARE signaling pathway
Lipopolysaccharide (LPS) is a widely used inflammatory inducer that induces the production of intracellular ROS. To mimic the high level ROS environment of inflamed tissues, cells are exposed to LPS to induce the production of high levels of ROS, which can be used to endogenously activate prodrugs. To verify that the prodrug is effective at activating Nrf2 at the cellular level, it was tested using the ARE luciferase reporter assay. HepG2-ARE-C8 cells were used, and when not induced by LPS, the ARE-inducing activity of the compound was examined after the prodrug/prodrug compound was added to act. In addition, cells were pretreated with LPS to generate endogenous ROS, and the effect was continued by adding a gradient prodrug compound to test ARE activity. As shown in fig. 4A, the prodrug had no significant ARE-inducing activity even at the highest concentrations when not activated, and Nrf2 was not activated when the prodrug was not released. After LPS induction, the prodrug releases original drug in cells, ARE induction activity is obviously enhanced, and Nrf2 can be effectively activated at a cellular level (as shown in figure 4B). Furthermore, the induction activity was shown to be enhanced in the concentration dependence of the original drug compound and LPS, and also time-dependent (see FIGS. 4C and 4D). In conclusion, the prodrug can be selectively activated by LPS (LPS) to generate ROS (reactive oxygen species), and active medicaments ARE released so as to activate a Keap1-Nrf2-ARE signal pathway.
3.2.2 LPS-induced prodrug-activated Nrf2 antioxidant System
mRNA levels of Nrf2 and regulatory genes, heme-oxygenase-1 (HO-1), glutamylcysteine ligase (GCLM) and NAD (P) H quinone oxidoreductase 1(NAD (P) H dehydrogenase, quinone 1, NQO-1), were determined by qRT-PCR. RAW264.7 cells were pretreated with LPS (10ng/mL), and the effect was continued by adding prodrug (500nM), and then the gene was detected using the qRT-PCR assay. As shown in fig. 5A-D, prodrug P-168 was able to significantly activate transcription of Nrf2 and its downstream genes under activation of endogenous ROS. Furthermore, protein levels of the above genes were detected by western blotting (fig. 5F), and the experimental results were consistent with mRNA levels of these genes, and LPS-induced prodrug activation significantly increased Nrf2 and downstream antioxidant enzyme protein levels, more potent than the parent drug at the same concentration. Subsequently, to continue to investigate the effect of the prodrug on antioxidant capacity under inflammatory conditions, the GSH/GSSG ratio was examined, an important marker indicative of oxidative stress. As shown in fig. 5E, exposure to LPS resulted in a large decrease in the GSH/GSSG ratio, indicating that LPS can cause oxidative stress, and the addition of prodrug P-168 can restore these indices to almost normal and exhibit concentration dependence.
3.2.3 prodrugs reduce LPS-induced inflammatory factor production in RAW264.7 cells
mRNA levels of IL-1 β and IL-6, which were inhibited by Nrf2, were measured using the qRT-PCR assay. As shown in FIGS. 6A and 6B, the expression of IL-1. beta. and IL-6mRNA in RAW264.7 cells was significantly increased after stimulation with 20ng/mL LPS for 8 h. Treatment with prodrug P-168 significantly inhibited the transcription of IL-1. beta. and IL-6 in RAW264.7 cells. Several inflammatory mediators closely related to ROS were subsequently evaluated by ELISA, including IL-1 β, IL-6, TNF- α and NO. All inflammatory factor levels were significantly increased after LPS treatment compared to the control group. The prodrug P-168 shows obvious advantages in inhibiting IL-1 beta, IL-6 and TNF-alpha production, and remarkably reduces the increase of extracellular NO concentration caused by LPS stimulation. In conclusion, the prodrug P-168 can be activated by ROS induced by LPS in RAW264.7 cells, and plays a remarkable role in cytoprotection and anti-inflammation.
In conclusion, it can be seen that:
1. the prodrug P-168 provided by the invention seals polar carboxylic acid groups of an active compound 168, improves fat solubility, and effectively improves membrane permeability and pharmacy;
2. the prodrug P-168 provided by the invention contains three functional fragments: (1) active compound 168, a Keap1-Nrf2 PPI inhibitor with superior activity; (2) a coumarin fluorophore; (3) an ROS activatable phenylboronate group; the compound is reduced under the condition of high ROS, a pharmacophore 168 and a fluorescent group coumarin are released, the expression of Nrf2 and downstream genes thereof is activated, the anti-inflammatory activity is exerted, meanwhile, fluorescence is released to realize visual monitoring, and the reduction of fluorescence intensity indicates that inflammation is relieved;
3. the prodrug P-168 provided by the invention is coupled with H2O2Is more selective than other biologically relevant ROS (e.g., hydroxyl radical, t-butoxy radical, superoxide, hypochlorite anion, and t-butyl hydroperoxide), and H2O2Is the most typical ROS in inflammation, thus ensuring that the prodrug P-168 is not easily interfered with for the selectivity of the visible monitoring of inflammation.
The above-described embodiments are intended to be illustrative of the nature of the invention, but those skilled in the art will recognize that the scope of the invention is not limited to the specific embodiments.

Claims (4)

1. A Keap1-Nrf2 PPI inhibitor prodrug is characterized in that the chemical structure is as follows
Figure FDA0002769374240000011
2. A process for the preparation of a Keap1-Nrf2 PPI inhibitor prodrug as claimed in claim 1, comprising the steps of:
Figure FDA0002769374240000012
the reagents and reaction conditions for each step were as follows:
(a) refluxing and stirring the compound 5 and a proper amount of sodium propionate, propionic anhydride and piperidine at 120 ℃ for 6 hours;
(b) CCl to Compound 64Adding a proper amount of NBS and AIBN into the solution, carrying out thermal reflux reaction for 8 hours, and removing the solvent under reduced pressure; adding a proper amount of NaOAc and acetic acid into the obtained residue, and carrying out a thermal reflux reaction for 12 hours; subsequently, an appropriate amount of hydrochloric acid was added to the hot reaction mixture, the reaction was continued for 30 minutes, and stirred at room temperature overnight;
(c) dissolving the compound 7 and an appropriate amount of pyridine in dichloromethane, stirring in an ice bath for 10 minutes, slowly adding an appropriate amount of trifluoromethanesulfonic anhydride, and stirring the reaction mixture for 2 hours;
(d) adding an appropriate amount of compound 8 to stirred bis (pinacolato) diboron, Pd (dppf) Cl2·CH2Cl2And KOA in dioxane, the reaction mixture was refluxed under nitrogen for 8 hours; after completion of the reaction, the solvent was removed under reduced pressure, and the reaction mixture was diluted with EtOAc, washed with brine, anhydrous Na2SO4Drying, filtering, and removing the solvent under reduced pressure; the crude product was dissolved in MeOH and the appropriate amount of NaBH added4Stirring and reacting for 1 hour at normal temperature;
(e) dissolving the compound 168 in dichloromethane, adding a proper amount of DMAP and DCC, stirring for half an hour at normal temperature, adding a proper amount of a compound 9, and stirring overnight;
wherein the chemical structural formula of the compound 168 in the step (e) is:
Figure FDA0002769374240000021
3. use of a Keap1-Nrf2 PPI inhibitor prodrug as claimed in claim 1 for the preparation of an anti-inflammatory drug.
4. Use of a Keap1-Nrf2 PPI inhibitor pro-drug as claimed in claim 1 for the manufacture of a medicament for anti-inflammatory and monitoring the degree of inflammatory remission.
CN202011244101.5A 2020-11-10 2020-11-10 Keap1-Nrf2 PPI inhibitor prodrug, preparation method and application Active CN112321620B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011244101.5A CN112321620B (en) 2020-11-10 2020-11-10 Keap1-Nrf2 PPI inhibitor prodrug, preparation method and application

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011244101.5A CN112321620B (en) 2020-11-10 2020-11-10 Keap1-Nrf2 PPI inhibitor prodrug, preparation method and application

Publications (2)

Publication Number Publication Date
CN112321620A CN112321620A (en) 2021-02-05
CN112321620B true CN112321620B (en) 2022-03-29

Family

ID=74315545

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011244101.5A Active CN112321620B (en) 2020-11-10 2020-11-10 Keap1-Nrf2 PPI inhibitor prodrug, preparation method and application

Country Status (1)

Country Link
CN (1) CN112321620B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3108534A1 (en) 2018-08-20 2020-02-27 Janssen Pharmaceutica Nv Inhibitors of keap1-nrf2 protein-protein interaction

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107266483A (en) * 2017-06-08 2017-10-20 浙江工业大学 A kind of hydrogen peroxide that responds kills photaesthesia targeting anti-tumor prodrug of tumour cell and preparation method and application

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107266483A (en) * 2017-06-08 2017-10-20 浙江工业大学 A kind of hydrogen peroxide that responds kills photaesthesia targeting anti-tumor prodrug of tumour cell and preparation method and application

Also Published As

Publication number Publication date
CN112321620A (en) 2021-02-05

Similar Documents

Publication Publication Date Title
AU2018244180B2 (en) Synthesis of Mcl-1 inhibitor
US8791258B2 (en) Pro-fluorescent probes
Leonard et al. Reaction of diethyl pyrocarbonate with nucleic acid components. Adenosine
Chen et al. Synthesis and biological evaluation of 1, 9-disubstituted β-carbolines as potent DNA intercalating and cytotoxic agents
Chen et al. Synthesis, cytotoxic activities and DNA binding properties of β-carboline derivatives
CN112110969B (en) Triazole glucoside derivative of 3-nitro-1-ethyl formate-7-azaindole, and preparation method and application thereof
EP3689875B1 (en) Use of triazolopyrimidine, triazolopyridine compounds and composition thereof for treating prc2-mediated diseases
CN109336835B (en) Fluorescent probe for detecting activity of myeloperoxidase and preparation method and application thereof
EP3823971B1 (en) Substituted triazolo quinoxaline derivatives
CN112321620B (en) Keap1-Nrf2 PPI inhibitor prodrug, preparation method and application
CN112912380A (en) Crystal form, amorphous form and application of MEK inhibitor
WO1999051586A1 (en) Reagent for singlet oxygen determination
CN113121599B (en) Fluorescent probe for targeting mitochondrial G-quadruplex DNA and preparation method and application thereof
WO2019114741A1 (en) Salt serving as akt inhibitor and crystal thereof
CN114957231A (en) GPX4 protein targeted degradation chimera and preparation method and application thereof
EP2683714B1 (en) Ii (pi)-conjugated fluoroionophores and method for determining an alkali ion
Formica et al. New branched macrocyclic ligand and its side-arm, two urea-based receptors for anions: synthesis, binding studies and crystal structure
CN112500293B (en) 1,1' -biphenyl-2, 6-diphenol compound and application thereof
Zhou et al. Synthesis, biological evaluation and cellular localization study of fluorescent derivatives of Jiyuan Oridonin A
CN107417598B (en) Fluorescent probe for detecting G-quadruplex DNAs (deoxyribonucleic acids) and preparation method thereof
CN114292226B (en) Compound with hydroxamic acid structure and preparation method and application thereof
Banerjee et al. A small-molecule fluorogenic probe for the detection of hypochlorite and its application in the bio-imaging of human breast cancer cells
Feng et al. Several Golgi targeting fluorescence markers based on 1, 8-naphthalimide derivatives with amide and long carbon chain
CN111606888B (en) Pyrrole derivative and preparation method and application thereof
CN112898260A (en) 3-carbonyl-2H-benzopyran compound or pharmaceutically acceptable salt thereof, preparation method and application

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