CN110790750B - Phthalimide selective androgen receptor degradation agent and preparation method and application thereof - Google Patents

Phthalimide selective androgen receptor degradation agent and preparation method and application thereof Download PDF

Info

Publication number
CN110790750B
CN110790750B CN201911080765.XA CN201911080765A CN110790750B CN 110790750 B CN110790750 B CN 110790750B CN 201911080765 A CN201911080765 A CN 201911080765A CN 110790750 B CN110790750 B CN 110790750B
Authority
CN
China
Prior art keywords
nmr
preparation
phthalimide
cdcl
101mhz
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
CN201911080765.XA
Other languages
Chinese (zh)
Other versions
CN110790750A (en
Inventor
徐霞
刘宏民
可钰
梁坚家
谢航
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.)
Zhengzhou University
Original Assignee
Zhengzhou 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 Zhengzhou University filed Critical Zhengzhou University
Priority to CN201911080765.XA priority Critical patent/CN110790750B/en
Publication of CN110790750A publication Critical patent/CN110790750A/en
Application granted granted Critical
Publication of CN110790750B publication Critical patent/CN110790750B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

The invention discloses a phthalimide chimeric molecule containing 1,2, 3-triazolyl, a preparation method and a selection thereofAn application of sex Androgen Receptor (AR) inhibitor in the field of AR-resistant high-expression cancer belongs to the field of pharmaceutical chemistry. It has the following structural general formula:
Figure DEST_PATH_IMAGE002
the compounds have good combination effect on AR and AR degradation activity, and can be used as further developed candidates or lead compounds for preparing anti-AR high-expression cancer drugs.

Description

Phthalimide selective androgen receptor degradation agent and preparation method and application thereof
Technical Field
The invention relates to the field of medicinal chemistry, in particular to a phthalimide chimeric molecule containing 1,2, 3-triazolyl, a preparation method thereof and application thereof as a selective Androgen Receptor (AR) degradation agent in the field of resisting prostate cancer.
Background
Prostate cancer (PCa) is common in middle-aged and elderly men, and is a common genitourinary malignancy, the worldwide incidence of which is second only to lung cancer, the second highest incidence of cancer, and the mortality of the sixth highest incidence of all male malignancies. The PCa therapeutic drug market is growing at an annual rate of 15-20% worldwide.
Androgen-deprivation therapy of surgical castration or drug castration is the standard of treatment for advanced stage prostate cancer, but after a median time of treatment of 14-30 months, almost all patient lesions develop from androgen-dependent prostate cancer (HDPC) to androgen-independent prostate cancer (HIPC), also known as castration-resistant prostate cancer (CRPC), with a median survival of less than 20 months in patients, resulting in endocrine treatment failure, such that antiandrogen therapy has essentially no effect on such patients, who eventually die from prostate cancer; approximately 50,000 patients will develop metastatic disease each year, which is known as metastatic crpc (mcrpc).
The most major obstacle to the progression of CRPC is the inability of AR signaling antagonists acting through LBD (e.g., bicalutamide, enzalutamide, abiraterone) to inhibit growth driven by N-terminal domain (NTD) dependent constitutively active AR-SV. There is currently no effective treatment for CRPC patients, and although the molecular biological mechanisms involved in their development are not completely understood, a number of studies have shown high AR expression in 80% of advanced CRPC. Degrading agent molecules degrade AR by direct targeting, thereby truly blocking this pathway at the AR level, and may be a potential alternative strategy for treating CRPC.
In 2004, the Ohwen Rous, arone Cheranoo and Alfamil Helichi families achieved the Nobel prize due to the discovery of ubiquitin-mediated protein degradation processes. In 2008, the Crews group reported that nutlin 3a has high binding force (Ki) with E3 ligase Mdm2, and the Ki value is nmol.L-1Horizontally, the androgen receptor antagonist bicalutamide analogue is covalently linked with nutlin 3a by polyethylene glycol Linker to generate PROTAC molecules (the structure is shown in the specification), the chimera can effectively degrade AR in cells with high androgen expression, and can prevent the degradation of AR after a proteasome inhibitor is added, and the process is proved to occur through a ubiquitin-proteasome pathway.
Figure BDA0002263882020000021
Mdm 2-based E3 ligase mediates AR targeted degradation of PROTAC.
Disclosure of Invention
The invention aims to provide a novel phthalimide androgen receptor degradation agent and a preparation method thereof; another purpose is to provide the application of the compound as a lead compound for preparing the anti-prostate cancer.
The specific general formula of the phthalimide androgen receptor degrading agent is as follows:
Figure BDA0002263882020000022
wherein R is C0-10Straight chain alkyl, phenyl;
m=1-6;
R1is- (CH)2)a-,-(CH2CH2O)b-,-(CH2CH2CH2O)c-, a ═ 1 to 10, b ═ 1 to 10, c ═ 1 to 10; w is an O or S atom;
preferred in formula I: r is phenyl or C1-5A linear alkyl group; m is 1 or 2; w is an O or S atom;
R1is- (CH)2)a-,-(CH2CH2O)b-,-(CH2CH2CH2O)c-,a=1-5,b=1-5,c=1-2;
The following compounds are preferred:
Figure BDA0002263882020000023
Figure BDA0002263882020000031
the invention relates to a triazolyl-containing phthalimide compound which is mainly prepared by the following method:
1. a process for the preparation of a compound of formula II:
the compound of the following general formula II is an intermediate for synthesizing the compound of the general formula I, and R is para-substituted phenyl or straight-chain alkyl; m is 1-6 and has the structure of
Figure BDA0002263882020000033
The alkyl chain of (A) may be-CH2-、-CH2CH2-、-CH2CH2CH2-and the like; w is an O or S atom;
Figure BDA0002263882020000032
in a solvent, carrying out electrophilic substitution reaction on commercially available 4-fluoro-2-trifluoromethyl benzonitrile and 5, 5-dimethyl hydantoin under an alkaline condition, wherein the used alkali is one of sodium hydroxide, potassium carbonate, sodium carbonate and the like; the method comprises the following steps of (1) using one or any two of N, N-dimethylformamide, acetonitrile, acetone, dioxane, methanol and ethanol as a solvent, purifying by column chromatography or recrystallization to obtain an intermediate 1, using one or two of ethyl acetate, diethyl ether and N-hexane as a solvent for recrystallization, carrying out nucleophilic substitution reaction on the intermediate 1 and dibromoalkane or phenyl azide with different lengths under an alkaline condition, and using one of sodium hydride, potassium hydride, sodium hydroxide, potassium hydroxide and the like as a base; the solvent is one of tetrahydrofuran, N-dimethylformamide, acetonitrile, dioxane, methanol and ethanol, and finally nucleophilic substitution reaction is carried out on the solvent and sodium azide under the heating condition, the solvent is one of N, N-dimethylformamide, ethyl acetate/water and acetone/water, and the obtained products L-1 and L-2 are purified by column chromatography to obtain pure products.
In a solvent, commercially available methyl 2-aminoisobutyrate hydrochloride undergoes nucleophilic substitution reaction with bromoalkanol and benzyl bromoazide respectively under alkaline conditions to obtain an intermediate 3 or 4 respectively. The alkali is one of potassium carbonate, sodium hydroxide, etc.; the solvent is one of acetonitrile, N-dimethylformamide, acetone, methanol and ethanol; the intermediate 3 or 4 and cyanobenzene isocyanuric acid ester are subjected to cyclization reaction with acetyl under alkaline conditions, the used alkali is one of triethylamine and diisopropylethylamine, and the used solvent is one of tetrahydrofuran, dichloromethane, chloroform, acetone, N-dimethylformamide and the like; the obtained products L-3 and L-4 are purified by column chromatography or recrystallization and the like to obtain pure products. The solvent used for recrystallization is one or a mixture of two of ethyl acetate, diethyl ether, isopropyl ether, ethanol and acetone.
The invention provides a preparation method of the compound, which is synthesized by the following reaction route:
Figure BDA0002263882020000041
Figure BDA0002263882020000051
2. a process for the preparation of a compound of formula III:
the compound of the following general formula III is an intermediate for synthesizing the compound of the general formula I, R1Is C1-10Straight chain alkyl or C1-5A polyalkyldiol chain.
Figure BDA0002263882020000052
In a solvent, commercially available 3-hydroxy phthalic anhydride and 3-amino-2, 6-piperidedione hydrochloride react under the condition of heating acetic acid and acetic anhydride, the obtained product is recrystallized in water to obtain an intermediate 10, and then the intermediate is subjected to alkylation reaction with a p-methyl benzene sulfonate alkynyl intermediate with alkyl or polyalkyl glycol chains of different chain lengths under an alkaline condition, wherein the base is one of sodium carbonate and sodium bicarbonate, and the solvent is one of N, N-dimethylformamide, acetonitrile, dioxane, tetrahydrofuran and the like; purifying the obtained product by column chromatography or recrystallization to obtain a pure product, wherein the solvent used for recrystallization is one or a mixture of two of ethyl acetate, diethyl ether, isopropyl ether and the like.
The invention provides a preparation method of the compound, which is synthesized by the following reaction route:
Figure BDA0002263882020000061
3. process for the preparation of compounds of the general formula I
In organic solvent, compound of formula II and compound of formula III in CuSO4Sodium ascorbate, CuI/organic base or Cu/CuSO41, 3-cycloaddition reaction is carried out under the condition, and the used alkali is diisopropylethylamine and triethylamine; the organic solvent is acetonitrile, tetrahydrofuran/water, tert-butyl alcohol/water, N, N-dimethylformamide/water, methanol/water, etc.; the reaction temperature is between 0 and 120 ℃, usuallyAt room temperature. The obtained product is purified by recrystallization, column chromatography and the like to obtain a pure product. The solvent used for recrystallization is one or a mixture of two of acetonitrile, methanol, ethanol, acetone, ethyl acetate, dichloromethane and chloroform.
The invention provides a preparation method of the compound, which is synthesized by the following reaction route:
Figure BDA0002263882020000062
the compounds of the invention are targeted proteolytic chimeras (PROTACs) that selectively induce degradation of AR proteins.
The invention has the advantages that: the AR non-steroid antagonist and an E3 ligase complex cereblon protein ligand are connected by a 1,2, 3-triazole Linker to obtain the bifunctional micromolecule. Compared with the prior art, the 1,2, 3-triazole active unit is combined with the E3 ligase ligand phthalimide and the AR ligand to form the targeting chimeric molecule by click chemistry for the first time, the triazole-containing phthalimide compound is synthesized simply, efficiently and environmentally, and in-vitro AR activity combination experiments show that the triazole-containing phthalimide compound has better combination effect and certain degradation effect on AR protein, can be used as an active ingredient for preparing an anti-prostatic cancer antagonistic or degradation drug, and is expected to be well applied to treatment of prostatic cancer, advanced prostatic cancer, castration-resistant prostatic cancer or other hyperandrogenism skin diseases.
Drawings
FIG. 1 is a western blot of AR protein expression in LnCAP cells with the compounds of the present invention;
FIG. 2 is a bar graph showing the effect of the compounds of the present invention on the expression of AR proteins in LnCAP cells.
Detailed Description
The starting materials may be obtained from commercial sources or prepared by methods known in the art or according to the methods described herein.
The compound structure passes through the nucleusMagnetic resonance (1H-NMR,13C-NMR) and mass spectrometry (HRMS).1H-NMR and13C-NMR was measured using a nuclear magnetic resonance apparatus model DPX-400 (Bruker, Germany); the high resolution mass spectrum is measured by a Q-exact mass spectrometer of Saimer Feishale; 200-mesh 300-mesh silica gel (Qingdao ocean chemical production) is used for column chromatography.
EXAMPLE 1 preparation of intermediate L-1
(1) Preparation of intermediate 1
Weighing 508mg of 5, 5-dimethylhydantoin, placing in a bottle shaped like a eggplant, adding 5mL of DMF while stirring to completely dissolve the 5, 5mg of K2CO3(1eq), stirring for half an hour at 45 ℃ to fully mix the components, finally slowly adding DMF solution of 4-fluoro-2-trifluoromethylbenzonitrile (300mg), stirring for 5 hours under heating, cooling to room temperature after full reaction, adding 30mL of ethyl acetate for dilution, extracting for 3 times by 20mL of saturated ammonium chloride aqueous solution, extracting for 1 time by saturated salt water, drying an organic layer for 6 hours by anhydrous magnesium sulfate, carrying out suction filtration, concentrating, and finally carrying out column chromatography with 200-300 meshes to obtain 330mg of white solid with the yield of 79%.
Intermediate 1, white solid:1H NMR(400MHz,DMSO-d6)δ8.83(s,1H),8.30(d,J=8.4Hz,1H),8.19(d,J=1.6Hz,1H),8.04(dd,J=8.4,1.8Hz,1H),1.43(s,6H).13C NMR(101MHz,DMSO-d6)δ175.72,152.94,136.98,135.98,131.19,130.87,129.93,124.04,123.99,123.60,120.88,115.20,106.55,57.94,24.56.
(2) preparation of intermediate 2
50mg of NaH (3eq) was weighed into a two-neck flask and charged with N2Replacing air in a two-neck flask for 3-4 times, slowly dropwise adding an intermediate 1(200mg) dissolved in dry DMF (dimethyl formamide) by using a constant-pressure dropping funnel under ice bath, stirring for 30min, slowly dropwise adding dibromopropane (2eq) diluted by 2 times of DMF, stirring for 2h at room temperature, fully reacting, then adding 30mL ethyl acetate for dilution, extracting for 3 times by using 20mL saturated ammonium chloride aqueous solution, extracting for 2 times by using saturated saline solution, drying an organic layer for 5h by using anhydrous magnesium sulfate, performing suction filtration, concentrating, and finally performing column chromatography by using 200-300 meshes to obtain 230mg of colorless liquid, wherein the yield is 85%.
The intermediate 2 is a mixture of two or more,colorless liquid:1H NMR(400MHz,CDCl3)δ8.15(s,1H),8.00(dd,J=8.4,1.8Hz,1H),7.92(d,J=8.4Hz,1H),3.65(t,J=6.0Hz,2H),3.57–3.45(m,2H),2.36–2.16(m,2H),1.56(d,J=2.5Hz,6H).13C NMR(101MHz,DMSO-d6)δ172.67,155.80,139.03,138.78,132.19,130.87,129.93,125.14,123.59,123.60,121.88,114.32,106.55,57.63,44.06,30.02,30.07,23.56.
(3) preparation of intermediate L-1
200mg of intermediate 2 is weighed, completely dissolved in 3mL of acetone solution, and 1mL of water-soluble NaN is slowly dropped into the solution under stirring at room temperature3(87mg), refluxing for 6h at 80 ℃, evaporating acetone after the reaction is finished, extracting for 3 times by using ethyl acetate, back extracting once by using saturated saline, combining organic layers, drying by using anhydrous magnesium sulfate, performing suction filtration, concentrating under reduced pressure, and purifying by using a column chromatography method to obtain 210mg of a colorless liquid of an intermediate L-1, wherein the yield is 90%.
Intermediate L-1, colorless liquid:1H NMR(400MHz,CDCl3)δ8.00–7.93(m,1H),7.92(s,1H),7.79(d,J=8.2Hz,1H),3.78(dd,J=17.0,8.7Hz,2H),3.48(t,J=6.2Hz,2H),2.14(d,J=5.8Hz,2H),1.68(s,J=64.4Hz,6H).13C NMR(101MHz,CDCl3)δ178.64,175.09,137.12,135.19,133.82,132.83,132.16,130.11,127.08,126.94,126.02,123.29,120.57,114.89,109.89,49.11,41.71,27.47,23.04.
EXAMPLE 2 preparation of intermediate L-2
Weighing p-methylaniline, adding into a flask, sequentially adding 1mL of concentrated hydrochloric acid and 1mL of water, stirring at room temperature for half an hour, then carrying out ice bath to below zero ℃, and quickly adding NaNO dissolved by 1mL of ice water below the liquid level of the system2(1.1eq) solution, stirring for 30min, cooling to-4 deg.C in a salt bath, and adding dropwise NaN dissolved in 1.5mL of water3(3.0eq) solution is stirred for 4-8 h until TLC monitoring reaction is complete, a neutralization system of sodium carbonate is slowly added under the condition of ice bath stirring, then ethyl acetate is used for extraction, saturated salt solution is used for washing for 2 times, low-temperature spin drying is carried out, a phenylazide product is obtained, then NBS and AIBN are taken, and the mixture is subjected to CCl reaction4Refluxing to obtain benzyl bromide azide intermediate, and finally weighing intermediate 1, and intermediate L-1180mg of L-2 was obtained as a tan solid in 65% yield.
Intermediate L-2, tan solid:1H NMR(400MHz,DMSO-d6)δ8.33(d,J=8.4Hz,1H),8.26(s,1H),8.10(d,J=8.3Hz,1H),7.51(d,J=8.3Hz,2H),7.10(d,J=8.3Hz,2H),4.62(s,2H),1.41(s,6H).13C NMR(101MHz,DMSO-d6)δ174.55,153.06,138.34,136.83,135.97,134.86,131.51,131.19,130.87,130.55,129.99,129.38,126.31,124.07,124.02,123.59,120.87,118.98,115.16,106.73,106.71,61.92,41.91,22.62.
EXAMPLE 3 preparation of intermediates L-3 and L-4
Methyl 2-aminoisobutyrate hydrochloride (200mg) was weighed to react with bromopropanol (1.2eq) to give intermediate 3, which was then reacted with less toxic phenylisothiocyanato to give 3-a in high yield, followed by reaction of 3-a with TsCl (1.5eq) and DABCO (2eq) to give intermediate 3-b, and finally with NaN3(3eq) refluxing for 6h at 80 ℃, evaporating acetone after the reaction is finished, extracting for 3 times by using ethyl acetate, performing back extraction once by using saturated saline solution, combining organic layers, drying by using anhydrous magnesium sulfate, performing suction filtration, performing reduced pressure concentration, and purifying by using a column chromatography method to obtain 210mg of ligand L-3 yellow solid with the yield of 90%.
Intermediate 3-a, colorless oily liquid:1H NMR(400MHz,CDCl3)δ7.96(d,J=8.3Hz,1H),7.90(d,J=1.4Hz,1H),7.78(dd,J=8.3,1.8Hz,1H),3.96–3.87(m,2H),3.75(s,2H),2.30(s,1H),2.06(dt,J=12.2,6.0Hz,2H),1.61(s,6H).13C NMR(101MHz,DMSO-d6)δ182.68,175.73,138.23,135.26,132.96,130.64,123.67,117.98,116.93,105.07,66.74,59.56,44.25,33.14,24.53.
intermediate 3-b, a colorless oil,1H NMR(400MHz,CDCl3)δ7.94(t,J=8.0Hz,1H),7.88(d,J=12.3Hz,1H),7.77(t,J=11.1Hz,3H),7.34(t,J=9.8Hz,2H),4.30–4.11(m,6H),3.81(dd,J=21.2,13.3Hz,2H),2.44(d,J=8.8Hz,3H),2.25(dt,J=15.2,6.0Hz,2H),2.04(p,J=6.0Hz,2H),1.58(d,J=12.5Hz,6H).13C NMR(101MHz,CDCl3)δ171.71,79.63,74.58,70.62,70.52,70.46,69.25,69.09,62.96,58.39,48.82,42.33,36.73,32.76,28.61.
the intermediate L-3, a yellow solid,1H NMR(400MHz,CDCl3)δ8.00–7.93(m,1H),7.92(s,1H),7.79(d,J=8.2Hz,1H),3.78(dd,J=17.0,8.7Hz,2H),3.48(t,J=6.2Hz,2H),2.14(d,J=5.8Hz,2H),1.68(s,J=64.4Hz,6H).13C NMR(101MHz,CDCl3)δ178.64,175.09,137.12,135.19,133.82,133.49,133.16,132.83,132.16,130.11,127.08,127.04,126.99,126.94,126.02,123.29,120.57,117.84,114.89,109.89,65.22,49.11,41.71,27.47,23.04.
methyl 2-aminoisobutyrate hydrochloride (200mg) was weighed to react with p-benzyl bromoazide (1.2eq) in acetonitrile as solvent and potassium carbonate as base to give intermediate 4, which was then reacted with less toxic phenylisothiocyanic acid ester to give intermediate L-4 at 140mg with 75% yield.
Intermediate L-4, tan solid:1H NMR(400MHz,DMSO-d6)δ8.33(d,J=8.4Hz,1H),8.26(s,1H),8.10(d,J=8.3Hz,1H),7.51(d,J=8.3Hz,2H),7.10(d,J=8.3Hz,2H),4.62(s,2H),1.41(s,6H).13C NMR(101MHz,DMSO-d6)δ174.55,153.06,138.34,136.83,135.97,134.86,131.51,131.19,130.87,130.55,129.99,129.38,126.31,124.07,124.02,123.59,120.87,118.98,115.16,106.73,106.71,61.92,41.91,22.62.
example 4 Synthesis of intermediate 10
Weighing 3-hydroxyphthalic anhydride, completely dissolving the 3-hydroxyphthalic anhydride by using acetic acid as a solvent, then adding sodium acetate (2eq) and 3-amino-2, 6-piperidinedione hydrochloride (1eq), stirring for 10 hours at 110 ℃, fully and completely reacting, evaporating the acetic acid solvent under reduced pressure, adding water, stirring for half an hour, precipitating a gray solid, filtering by suction, and drying to obtain an intermediate 10 with the yield of 85%.
Intermediate 10, off white solid:1H NMR(400MHz,DMSO-d6)δ11.19(s,1H),11.09(s,1H),7.66(dd,J=8.3,7.3Hz,1H),7.32(d,J=7.1Hz,1H),7.25(d,J=8.2Hz,1H),5.08(dd,J=12.9,5.4Hz,1H),2.89(ddd,J=17.3,14.0,5.4Hz,1H),2.56(dd,J=20.8,10.8Hz,2H),2.09–1.96(m,1H).13C NMR(101MHz,DMSO-d6)δ173.71,171.58,169.97,168.61,156.58,136.46,134.21,122.40,115.74,112.53,52.77,29.68,24.11.
EXAMPLE 5 preparation of intermediate 6
Potassium tert-butoxide 64mg (1.2eq) was weighed out and dissolved in dry THF solution, N2Adding polyethylene glycol chains of different lengths 200mg (1eq) under the environment, stirring for 30min under an ice salt bath, then dropwise adding a bromopropyne solution diluted by one time of THF by using a constant-pressure dropping funnel, stirring overnight at room temperature, fully reacting, finally removing insoluble substances by using a suction filtering funnel, removing THF at a low temperature by spinning, concentrating under reduced pressure, and purifying by using column chromatography to obtain a yellow liquid 6 with the yield of 38%.
EXAMPLE 6 Synthesis of intermediate III-1
200mg of intermediate 10 are weighed out and completely dissolved in DMF solution, and 56mg of K are added2CO3(1.2eq) fully stirring, finally slowly dropwise adding a DMF solution of alkynyl p-methylbenzenesulfonate with an alkyl chain or a polyethylene glycol chain, stirring for 6 hours at 85 ℃, detecting by TLC until the reaction is complete, cooling to room temperature, adding 30mL of dichloromethane diluted solution, extracting for 3 times by using 20mL of saturated ammonium chloride aqueous solution, extracting twice by using saturated saline solution, drying an organic layer for 5 hours by using anhydrous sodium sulfate, carrying out suction filtration, adding a small amount of ethyl acetate for recrystallization, and separating out white solid to obtain the target compound III-1.
Intermediate III-1, a white solid,1H NMR(400MHz,DMSO-d6)δ11.10(s,1H),7.85–7.77(m,1H),7.54(d,J=8.5Hz,1H),7.47(d,J=7.2Hz,1H),5.08(dd,J=12.8,5.4Hz,1H),4.32(t,J=6.6Hz,2H),2.97–2.85(m,2H),2.70(td,J=6.5,2.6Hz,2H),2.66–2.51(m,2H),2.14–1.94(m,1H).13C NMR(101MHz,DMSO-d6)δ173.71,169.97,169.74,168.61,155.71,136.92,133.20,118.92,118.87,116.95,84.40,70.14,68.24,52.77,29.68,28.29,24.11,18.81.
example 7: preparation of 4- (3- (4- (4- (2- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) ethyl) -1H-1,2, 3-triazol-1-yl) benzyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-1) as shown in the general formula (I).
120mg of alkynyl III-1 ligand is weighed, 5mL of tetrahydrofuran is added, 2eq of azide L-4 ligand 297mg are added after ultrasonic dissolution, and 41mg of CuSO is added4And 45mg VcNa, and finally 1.5mL of H2And O, stirring for 7-11 h at room temperature until the reaction is complete, performing suction filtration by using diatomite to remove metal insoluble substances, performing rotary evaporation at 39 ℃ to remove most tetrahydrofuran, adding 40mL of ethyl acetate to dilute the system, extracting for three times by using 24mL of saturated saline solution, adding anhydrous magnesium sulfate to an organic layer, drying, performing suction filtration and concentration, and performing column chromatography to obtain a white solid 170mg with the yield of 80%. Yellow solid, m.p. 158.2-159.5 ℃,1H NMR(400MHz,CDCl3)δ8.94(s,1H),8.74(s,1H),8.13(s,1H),8.01(d,J=8.6Hz,1H),7.94(d,J=8.4Hz,1H),7.80(d,J=8.4Hz,2H),7.71(t,J=7.9Hz,1H),7.56(d,J=8.4Hz,2H),7.50(d,J=7.2Hz,1H),7.24(d,J=8.6Hz,1H),4.97(dd,J=12.0,5.2Hz,1H),4.75(d,J=15.7Hz,1H),4.55(d,J=15.8Hz,1H),4.52–4.43(m,1H),4.26(dd,J=13.8,6.6Hz,1H),3.40(d,J=4.8Hz,2H),2.84(dd,J=26.3,13.9Hz,2H),2.74(dd,J=28.1,12.3Hz,2H),2.21–2.09(m,1H),1.67(s,2H),1.49(d,J=27.4Hz,6H).13C NMR(101MHz,CDCl3)δ173.71,172.68,169.97,169.74,168.61,155.71,155.21,152.26,138.65,138.61,136.92,134.79,133.20,132.75,132.32,132.13,131.76,123.67,122.79,118.92,118.87,116.95,116.93,116.67,115.34,113.05,68.74,64.49,52.77,39.74,29.68,27.72,24.53,24.11.ESI-HRMS:m/z cacld.For C37H29F3N8O7[M+H]+:755.2111,found 755.2132.
example 8: preparation of 4- (3- (4- (4- (3- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) propyl) -1H-1,2, 3-triazol-1-yl) benzyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-2).
The synthesis procedure is as in example 7, yellow brown solid, m.p. 156.5-157.9 ℃,1H NMR(400MHz,CDCl3)δ8.37(s,1H),8.18(s,1H),8.07–8.01(m,1H),7.99–7.91(m,2H),7.75(d,J=8.4Hz,2H),7.70–7.64(m,2H),7.52(d,J=8.4Hz,2H),7.45(d,J=7.3Hz,1H),7.24(d,J=8.5Hz,1H),4.95(dd,J=11.9,5.3Hz,1H),4.68(d,J=7.1Hz,2H),4.32–4.22(m,3H),3.09(t,J=6.9Hz,3H),2.91–2.78(m,2H),2.79–2.66(m,2H),2.36(dd,J=12.7,6.4Hz,2H),2.16–2.07(m,1H),1.70(s,2H),1.47(d,J=4.8Hz,6H).13C NMR(101MHz,CDCl3)δ173.71,172.68,169.97,169.74,168.61,155.71,155.21,149.66,138.65,138.61,136.92,134.79,133.20,132.75,132.32,132.13,131.76,123.67,122.90,122.79,118.92,118.87,116.95,116.93,116.67,113.05,69.99,64.49,52.77,39.74,29.68,25.79,24.53,24.11,23.95.ESI-HRMS:m/z cacld.For C38H31F3N8O7[M+H]+:769.2268,found 769.2261.
example 9: preparation of 4- (3- (4- (4- ((3- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) propoxy) methyl) -1H-1,2, 3-triazol-1-yl) benzyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-3).
The synthesis procedure was as in example 7, yellow brown solid, m.p.:215.2-216.1 ℃,1H NMR(400MHz,DMSO-d6)δ11.10(s,1H),8.77(s,1H),8.34(d,J=8.4Hz,1H),8.26(d,J=1.5Hz,1H),8.11(d,J=8.4Hz,1H),7.82(d,J=8.4Hz,1H),7.79(dd,J=8.4,7.4Hz,1H),7.64(d,J=8.5Hz,2H),7.50(d,J=8.5Hz,1H),7.41(d,J=7.2Hz,1H),5.05(dd,J=12.7,5.4Hz,1H),4.69(s,2H),4.62(s,2H),4.27(t,J=6.1Hz,2H),3.71(t,J=6.2Hz,2H),2.95–2.81(m,1H),2.58(d,J=17.8Hz,1H),2.28–2.15(m,1H),2.03(dd,J=11.5,5.5Hz,2H),1.44(s,6H).13C NMR(101MHz,DMSO-d6)δ174.62,172.80,169.90,166.80,155.80,153.16,145.15,138.37,137.05,136.79,136.02,133.10,130.15,128.94,124.22,121.99,119.87,116.14,115.23,65.76,63.21,62.04,48.69,28.75,22.60,21.94.ESI-HRMS:m/z cacld.For C39H33F3N8O8[M+H]+:799.2373,found 799.2329.
example 10: preparation of 4- (3- (4- (4- ((2- (2- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) ethoxy) methyl) -1H-1,2, 3-triazol-1-yl) benzyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-4).
The synthesis procedure was as in example 7, yellow brown solid, m.p. 226.5-227.9 ℃,1H NMR(400MHz,DMSO-d6)δ11.10(s,1H),8.76(s,1H),8.34(d,J=8.4Hz,1H),8.26(s,1H),8.11(d,J=8.3Hz,1H),7.86(d,J=8.4Hz,2H),7.82–7.74(m,1H),7.65(d,J=8.5Hz,2H),7.52(d,J=8.5Hz,1H),7.44(t,J=7.0Hz,1H),5.07(dd,J=12.6,5.4Hz,1H),4.70(s,2H),4.63(s,2H),4.38–4.30(m,2H),3.85–3.79(m,2H),3.70(dd,J=5.9,3.2Hz,2H),3.65(dd,J=5.7,3.2Hz,2H),2.93–2.80(m,1H),2.63–2.51(m,2H),2.31–2.22(m,2H),2.08–1.99(m,1H),1.42(d,J=15.1Hz,6H).13C NMR(101MHz,DMSO-d6)δ174.62,172.80,169.90,166.80,155.80,153.16,145.15,138.37,137.05,136.79,136.02,133.10,130.15,128.94,124.22,121.99,119.87,116.14,115.23,65.76,63.21,62.04,48.69,28.75,22.60,21.94.ESI-HRMS:m/z cacld.For C40H35F3N8O9[M+H]+:829.2479,found 829.2470.
example 11: preparation of 4- (3- (4- ((2- (2- (2- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) ethoxy) methyl) -1H-1,2, 3-triazol-1-yl) benzyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-5).
The synthesis procedure was as in example 7, yellow brown solid, m.p.:214.1-215.6 ℃,1H NMR(400MHz,CDCl3)δ8.47(s,1H),8.19(d,J=1.5Hz,1H),8.10–8.02(m,2H),7.94(d,J=8.4Hz,1H),7.74(d,J=8.3Hz,2H),7.69–7.62(m,1H),7.53(d,J=8.3Hz,2H),7.44(d,J=7.2Hz,1H),7.25(d,J=8.5Hz,1H),4.93(dd,J=11.7,5.4Hz,1H),4.77(s,2H),4.67(s,2H),4.37–4.30(m,2H),3.97–3.90(m,2H),3.79(dd,J=9.1,4.2Hz,2H),3.76(d,J=14.2Hz,3H),3.69(dd,J=10.0,5.1Hz,5H),2.85(dd,J=12.1,8.4Hz,1H),2.81–2.68(m,2H),2.27–2.18(m,1H),2.10(dd,J=12.8,7.9Hz,1H),1.46(s,6H).13C NMR(101MHz,CDCl3)δ174.32,171.13,168.53,167.01,165.82,156.54,153.38,136.64,136.20,135.39,133.74,133.44,128.07,123.08,123.03,119.11,117.08,115.92,114.98,108.44,77.25,66.45,66.08,64.37,62.12,49.13,47.81,37.62,31.40,29.87,29.20,23.37,23.34,22.64.ESI-HRMS:m/z cacld.For C42H39F3N8O10[M+H]+:873.2741,found 873.2751.
example 12: preparation of 4- (3- (4- (4- (13- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) -2,5,8, 11-ethoxy) -1H-1,2, 3-triazol-1-yl) benzyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-6).
The synthesis procedure was as in example 7, yellow brown solid, m.p. 236.3-237.5 ℃,1H NMR(400MHz,CDCl3)δ8.43(s,1H),8.19(d,J=1.5Hz,1H),8.11–8.02(m,1H),7.95(d,J=8.5Hz,1H),7.74(d,J=8.5Hz,1H),7.66(dd,J=8.3,7.4Hz,1H),7.54(d,J=8.5Hz,1H),7.44(d,J=7.2Hz,1H),7.25(s,1H),4.93(dd,J=12.0,5.3Hz,1H),4.79(d,J=12.9Hz,1H),4.68(s,1H),4.35–4.28(m,1H),3.96–3.88(m,1H),3.79–3.72(m,2H),3.71–3.64(m,3H),2.90–2.79(m,1H),2.73(dd,J=33.7,18.4,11.7Hz,1H),2.34(t,J=6.7Hz,1H),2.15–2.05(m,1H),1.87(dt,J=12.7,6.2Hz,2H),1.46(s,6H).13C NMR(101MHz,DMSO-d6)δ174.62,172.80,169.90,166.80,155.80,153.16,145.15,138.37,137.05,136.79,136.02,133.10,130.15,128.94,124.22,121.99,119.87,116.14,115.23,65.76,63.21,62.04,48.69,28.75,22.60,21.94.ESI-HRMS:m/z cacld.For C44H43F3N8O11[M+H]+:917.3003,found917.3014.
example 14: preparation of 4- (3- (3- (4- (2- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) ethyl) -1H-1,2, 3-triazol-1-yl) propyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-7).
The synthesis procedure was the same as in example 7, white solid, m.p. 185.1-186.8 ℃,1H NMR(400MHz,DMSO-d6)δ11.11(s,1H),8.32(d,J=8.4Hz,1H),8.19(d,J=1.8Hz,1H),8.04(dd,J=8.4,1.8Hz,1H),7.96(s,1H),7.80(dd,J=8.4,7.3Hz,1H),7.51(d,J=8.5Hz,1H),7.45(d,J=7.2Hz,1H),5.09(dd,J=12.9,5.4Hz,1H),4.43(t,J=7.2Hz,2H),4.33–4.20(m,2H),3.43–3.35(m,2H),2.96–2.88(m,1H),2.84(d,J=7.6Hz,2H),2.66–2.52(m,2H),2.17(dt,J=12.8,6.4Hz,2H),2.13–2.06(m,2H),2.08–2.00(m,1H),1.45(s,6H).13C NMR(101MHz,DMSO-d6)δ173.71,172.68,169.97,169.74,168.61,155.81,155.71,150.80,138.65,136.92,133.20,132.75,132.32,131.76,123.67,120.49,118.92,118.87,116.95,116.93,116.67,113.05,69.99,64.60,52.77,48.05,36.61,29.68,25.79,24.53,24.11,23.95,23.91.ESI-HRMS:m/z cacld.For C33H29F3N8O7[M+H]+:707.2111,found 707.2121.
example 15: preparation of 4- (3- (4- (4- (3- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) propyl) -1H-1,2, 3-triazol-1-yl) butyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-8).
The synthesis procedure was the same as in example 7, white solid, m.p. 174.3-175.6 ℃,1H NMR(400MHz,CDCl3)δ8.68(s,1H),8.13(s,1H),8.00(d,J=8.3Hz,1H),7.92(d,J=8.4Hz,1H),7.67(t,J=7.9Hz,1H),7.52(s,1H),7.45(d,J=7.2Hz,1H),7.22(d,J=8.5Hz,1H),4.96(dd,J=11.9,5.2Hz,1H),4.38(t,J=6.8Hz,2H),4.32–4.16(m,2H),3.37(t,J=7.3Hz,2H),3.00(dd,J=13.8,7.0Hz,2H),2.85(dd,J=22.0,9.3Hz,2H),2.76(t,J=7.4Hz,2H),2.29(dd,J=12.8,6.4Hz,2H),2.15(dd,J=15.7,7.9Hz,1H),2.01–1.92(m,2H),1.70(dt,J=14.8,7.4Hz,2H),1.49(s,6H).13C NMR(101MHz,CDCl3)δ173.71,172.68,169.97,169.74,168.61,155.81,155.71,150.80,138.65,136.92,133.20,132.75,132.32,131.76,123.67,120.49,118.92,118.87,116.95,116.93,116.67,113.05,69.99,64.60,52.77,51.79,40.72,29.68,27.72,25.79,24.88,24.53,24.11,23.95.ESI-HRMS:m/z cacld.For C35H33F3N8O7[M+H]+:735.2424,found 735.2421.
example 16: preparation of 4- (3- (3- (4- ((3- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) propoxy) methyl) -1H-1,2, 3-triazol-1-yl) propyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-9).
The synthesis procedure was the same as in example 7, white solid, m.p. 196.4-197.5 ℃,1H NMR(400MHz,DMSO-d6)δ11.11(s,1H),8.31(d,J=8.4Hz,1H),8.19(d,J=1.7Hz,1H),8.13(s,1H),8.04(dd,J=8.4,1.8Hz,1H),7.80(dd,J=8.4,7.4Hz,1H),7.50(d,J=8.5Hz,1H),7.44(d,J=7.2Hz,1H),5.09(dd,J=12.8,5.4Hz,1H),4.53(s,2H),4.45(t,J=7.2Hz,2H),4.26(t,J=6.2Hz,2H),3.65(t,J=6.2Hz,2H),3.39(t,J=7.3Hz,2H),2.97–2.80(m,1H),2.65–2.52(m,2H),2.19(dd,J=13.7,6.6Hz,2H),2.08–2.03(m,1H),2.02–2.00(m,1H),1.46(d,J=5.3Hz,6H).13C NMR(101MHz,CDCl3)δ174.32,171.13,168.53,167.01,165.82,156.54,153.38,136.64,136.20,135.39,133.74,133.44,128.07,123.08,123.03,119.11,117.08,115.92,114.98,108.44,77.25,66.45,66.08,64.37,62.12,49.13,47.81,37.62,31.40,29.87,29.20,23.37,23.34,22.64.ESI-HRMS:m/z cacld.For C35H33F3N8O8[M+H]+:751.2373,found751.2365.
example 17, 4- (3- (4- (4- ((2- (2- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) ethoxy) methyl) -1H-1,2, 3-triazol-1-yl) butyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-10) is prepared.
The synthesis procedure was the same as in example 7, white solid, m.p.:197.2-198.6 ℃,1H NMR(400MHz,CDCl3)δ8.48(s,1H),8.13(d,J=1.8Hz,1H),7.99(dd,J=8.5,1.9Hz,1H),7.92(d,J=8.4Hz,1H),7.67(dd,J=8.4,7.4Hz,1H),7.59(s,1H),7.46(d,J=7.2Hz,1H),7.29–7.23(m,2H),4.95(dd,J=12.1,5.3Hz,1H),4.67(s,2H),4.40(t,J=6.9Hz,2H),4.35(t,J=4.6Hz,2H),3.95–3.90(m,2H),3.80(dd,J=5.7,2.8Hz,2H),3.74–3.69(m,2H),3.48(s,1H),3.40–3.33(m,2H),2.85(dd,J=18.3,16.0Hz,1H),2.80–2.69(m,2H),2.16–2.08(m,1H),2.02–1.94(m,2H),1.71(t,J=7.3Hz,2H),1.50(s,6H).13C NMR(101MHz,DMSO-d6)δ174.62,172.80,169.90,166.80,155.80,153.16,145.15,138.37,137.05,136.79,136.02,133.10,130.15,128.94,124.22,121.99,119.87,116.14,115.23,65.76,63.21,62.04,48.69,28.75,22.60,21.94.ESI-HRMS:m/z cacld.For C37H37F3N8O9[M+H]+:795.2636,found 795.2616.
example 18 preparation of 4- (3- (4- (4- ((2- (2- (2- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) ethoxy) methyl) -1H-1,2, 3-triazol-1-yl) butyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-11).
The synthesis procedure was the same as in example 7, white solid, m.p. 203.2-204.5 deg.C,1H NMR(400MHz,CDCl3)δ8.85(d,J=18.9Hz,1H),8.13(d,J=1.3Hz,1H),8.03–7.89(m,1H),7.67(dt,J=17.0,6.1Hz,2H),7.55(s,1H),7.45(d,J=7.2Hz,1H),7.38(dd,J=8.7,2.2Hz,1H),7.26(s,1H),4.96(dd,J=11.1,4.4Hz,1H),4.65(d,J=13.1Hz,2H),4.34(dd,J=9.2,5.5Hz,3H),3.97–3.90(m,2H),3.78(dd,J=5.5,3.6Hz,4H),3.66(d,J=9.4Hz,6H),3.37(d,J=6.4Hz,2H),2.84(dd,J=16.0,12.1Hz,2H),2.76(dt,J=13.5,9.9Hz,2H),2.15–2.08(m,2H),1.99(dd,J=13.6,6.6Hz,2H),1.91(s,1H),1.69(dt,J=16.1,6.7Hz,2H),1.52(d,J=17.4Hz,6H).13C NMR(101MHz,CDCl3)δ174.32,171.13,168.53,167.01,165.82,156.54,153.38,136.64,136.20,135.39,133.74,133.44,128.07,123.08,123.03,119.11,117.08,115.92,114.98,108.44,77.25,66.45,66.08,64.37,62.12,49.13,47.81,37.62,31.40,29.87,29.20,23.37,23.34,22.64.ESI-HRMS:m/z cacld.For C39H41F3N8O10[M+H]+:839.2898,found 839.2887.
example 19 preparation of 4- (3- (4- (4- (13- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) -2,5,8, 11-ethoxy) -1H-1,2, 3-triazol-1-yl) butyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-12).
The synthesis procedure was the same as in example 7, white solid, m.p. 216.3-217.7 ℃,1H NMR(400MHz,CDCl3)δ8.98(s,1H),8.14(s,1H),8.01(d,J=8.3Hz,1H),7.93(d,J=8.4Hz,1H),7.67(t,J=7.8Hz,1H),7.59(s,1H),7.45(d,J=7.2Hz,1H),7.27(d,J=8.5Hz,1H),4.97(dd,J=7.2,3.4Hz,1H),4.67(s,2H),4.36(t,J=6.1Hz,5H),3.94(d,J=4.4Hz,2H),3.84–3.56(m,12H),2.91–2.81(m,1H),2.81(s,2H),2.15–2.06(m,1H),2.01–1.91(m,2H),1.73(d,J=6.8Hz,2H),1.51(s,6H),1.43–1.36(m,2H).13C NMR(101MHz,CDCl3)δ174.59,171.40,168.45,167.01,165.61,156.38,152.88,145.25,136.52,136.45,135.34,133.73,133.30,128.04,123.35,123.04,122.99,122.61,120.62,119.46,117.25,116.09,115.06,108.15,77.32,71.12,70.52,70.48,69.76,69.31,69.25,64.55,61.91,49.91,49.11,40.00,31.39,29.72,28.79,23.83,23.43,22.62.ESI-HRMS:m/z cacld.For C41H45F3N8O11[M+H]+:883.3160,found 883.3158.
example 20 preparation of 4- (3- (5- (4- ((2- (2- (2- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) ethoxy) methyl) -1H-1,2, 3-triazol-1-yl) pentyl) -4, 4-dimethyl-2, 5-dioxoimidazolidin-1-yl) -2- (trifluoromethyl) benzonitrile (I-13).
The synthesis procedure was the same as in example 7, white solid, m.p. 206.4-207.7 ℃,1H NMR(400MHz,CDCl3)δ8.64(s,1H),8.14(s,2H),8.00(d,J=8.4Hz,2H),7.92(d,J=8.4Hz,2H),7.70–7.64(m,2H),7.58(s,2H),7.46(d,J=7.2Hz,2H),7.26(s,2H),4.95(dd,J=11.7,5.5Hz,1H),4.68(d,J=4.4Hz,3H),4.35(dd,J=12.2,5.8Hz,6H),3.96–3.91(m,3H),3.78(dd,J=10.6,5.6Hz,3H),3.67(dd,J=6.4,4.1Hz,10H),3.36–3.28(m,2H),2.92–2.81(m,1H),2.80–2.71(m,2H),2.16–2.08(m,1H),2.01–1.94(m,2H),1.73(d,J=6.8Hz,2H),1.56–1.48(s,6H).13C NMR(101MHz,DMSO-d6)δ174.62,172.80,169.90,166.80,155.80,153.16,145.15,138.37,137.05,136.79,136.02,133.10,130.15,128.94,124.22,121.99,119.87,116.14,115.23,65.76,63.21,62.04,48.69,28.75,22.60,21.94.ESI-HRMS:m/z cacld.For C40H43F3N8O10[M+H]+:853.3054,found 853.3051.
example 21, 4- (3- (3- (4- ((2- (2- (2- ((2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione-isoindolin-4-yl) oxy) ethoxy) methyl) -1H-1,2, 3-triazol-1-yl) propyl) -4, 4-dimethyl-5-oxo-2-thioimidazolidin-1-yl) -2 (trifluoromethyl) benzonitrile (I-14) was prepared.
The synthesis procedure is as in example 7, red brown solid, m.p. 221.3-222.6 ℃,1H NMR(400MHz,CDCl3)δ8.67(s,1H),7.95(t,J=6.9Hz,1H),7.88(d,J=1.5Hz,1H),7.80–7.73(m,1H),7.67(dd,J=8.7,7.1Hz,2H),7.46(t,J=6.0Hz,1H),7.25(s,1H),4.96(dd,J=11.3,4.9Hz,1H),4.70(s,2H),4.47(t,J=6.6Hz,2H),4.37–4.31(m,3H),3.94(dd,J=10.8,6.0Hz,3H),3.81–3.75(m,5H),3.74–3.64(m,8H),2.90–2.81(m,2H),2.80–2.70(m,2H),2.55–2.44(m,2H),2.12(dd,J=7.4,4.9Hz,2H),1.53(s,6H).13C NMR(101MHz,CDCl3)δ174.32,171.13,168.53,167.01,165.82,156.54,153.38,136.64,136.20,135.39,133.74,133.44,128.07,123.08,123.03,119.11,117.08,115.92,114.98,108.44,77.25,66.45,66.08,64.37,62.12,49.13,47.81,37.62,31.40,29.87,29.20,23.37,23.34,22.64.ESI-HRMS:m/z cacld.For C38H39F3N8O9S[M+H]+:841.2513,found 841.2516.
example 22 AR protein binding level Activity assay of Compounds of the invention
The compounds of the invention were tested for in vitro AR affinity by Fluorescence Polarization (FP) to verify the binding interaction between the chimeric molecule and the AR protein. The primary sieving concentration of the compound of the present invention was 10 μm.
Preparing complete Buffer solution by using 1ml Buffer solution and 2 mu L1M DTT, then diluting the compound to be tested to 4X final concentration, controlling DMSO not to exceed 1%, firstly preparing 4X fluoromone by using fresh complete Buffer solutionTMTracer solution, then 5. mu.L of 4X FluormoneTMTracer solution was added to 384 well assay plates followed by 4X AR solution with complete AR buffer, 5 μ L of 4X AR-LBD solution was added to assay plates, then diluted test compound was added to 384 well plates 10 μ L per well, mixed well and the plates were covered to protect the test agent from light. Setting 1X FluormoneTM Tracer,1X AR-LBD\FluormoneTMTracer and 1X AR-LBD \ FluormoneTMTracer \ R1881 and solvent control group, each group 3 multiple wells, at least 3h incubation under dark at room temperature, finally placing the test board in a multifunctional microplate reader, measuring the fluorescence polarization value of each well at 485nm excitation wavelength and 535nm emission wavelength, taking the average value of three independent experiments, and analyzing the data by GrapHpad-prism software.
The fluorescence polarization inhibitory activity of the test compound at a concentration of 10. mu.M was calculated according to the following formula:
Figure BDA0002263882020000171
mP100%100% inhibition, mP0%Inhibition 0%, mpcound — the polarization value of the compound.
mP and corresponding concentrations of chimeric molecules at different concentrations IC was calculated using the formula GraphPad Prism fitted curves50
TABLE 1 binding Activity of preferred Compounds for AR
Figure BDA0002263882020000172
Example 23 degradation of Androgen Receptor (AR) by Compounds of the invention
The degradation test of the compound synthesized by the invention on Androgen Receptor (AR) is verified by extracting protein from prostate cancer cells LNCaP with high expression of androgen receptor through Western Blot experiment. The experimental result shows that the chimeric molecule has better degradation activity on androgen receptor.
(I) Experimental method
(1) Preparing a BCA standard curve: and adding PBS into 5mg/ml of standard protein to dilute the standard protein into standard protein with different concentrations. And (3) mixing solution A of the BCA working solution: mixing the solution B according to the volume ratio of 50: 1. In a 96-well plate, 19. mu.l of PBS, 1. mu.l of standard protein, and 200. mu.l of working solution were sequentially added, and incubated at 37 ℃ for 20 min. After the incubation, the OD value was measured at a wavelength of 562nm using a microplate reader. The absorbance values were substituted into the standard curve to calculate the protein concentration.
Cell plating: LNCaP which grows well in logarithmic phase is taken to prepare single cell suspension, and the single cell suspension is evenly inoculated in a six-hole plate in equal cell number.
Adding medicine: stock solutions of the preferred compounds of the present invention were diluted to 10. mu.M each, and 2ml of each of the drug-containing medium and the control medium was added to the corresponding six-well plate.
Collecting cells: after 24h of drug action, the primary medium and the cells collected by digestion were collected by centrifugation and washed 3 times with PBS to obtain a white cell pellet in a 1.5ml EP tube.
Cracking: and (3) adding the prepared RIPA lysate according to the cell number, then placing the cell on ice for lysis, and lightly blowing and beating an EP tube to ensure that the cell can be fully lysed. After full cracking, centrifuging at 12000rpm at 4 deg.C for 10-15min with a low temperature high speed centrifuge, transferring the supernatant to 1.5ml EP tube with a 1ml pipette, and discarding the bottom solid.
Protein quantification: mu.l of the sample was taken for protein quantification and the concentration of protein in each sample was calculated from the standard curve.
Protein denaturation: the collected protein lysate was added with 1/5 volume Loading Buffer and completely denatured by boiling in 100 ℃ boiling water in a water bath for 10 min. Storing at-20 ℃ in dark for later use.
Preparing glue, loading, electrophoresis, membrane transferring and dyeing: and (3) taking out the NC membrane after the electrophoresis membrane conversion is finished, washing the NC membrane once by using PBS, placing the NC membrane into ponceau red dye solution for protein dyeing, and judging whether the protein is smoothly converted into the membrane and the amount of the protein on the membrane according to the dyeing condition. The desired proteins of different molecular weights were cut according to the pre-stained protein Maker and then washed 3 times with TBST.
Sealing milk, sealing primary antibody, sealing secondary antibody, exposing, taking out NC membrane, washing with TBST for 3 times, each time for about 10 min. Primary antibody was diluted according to the instructions, AR and internal control GAPDH used in this experiment were diluted 1000-fold with TBST and incubated overnight at 4 ℃. The goat anti-rabbit and goat anti-mouse secondary antibodies used were both 5000-fold and 10000-fold diluted with TBST. And (3) adding the diluted secondary antibody to a corresponding NC membrane after uniformly mixing in a vortex mode, and incubating for 2h at room temperature. Uniformly mixing the solution A and the solution B of the ECL luminous liquid according to the size of the strip in a ratio of 1:1, adding the mixture on a film for reaction for 2min, and placing the film in a dark box for exposure.
Data processing: after scanning the films, the films were subjected to gray scale analysis using Image J software, and their data were statistically analyzed using GrapHpad-prism software.
(II) Experimental data and results
The AR degradation activity profile of preferred compounds of the invention is shown in FIGS. 1 and 2.
From the above biological activity evaluation, at a concentration of 10 μ M, all of the triazolyl-containing phthalimide compounds showed the ability to competitively bind to the AR-LBD ligand binding pocket with the fluorescence-labeled androgen fluorone, indicating that the compounds could inhibit the transcription activity of AR by competitively binding to AR with DHT, and some of the compounds also showed significant AR degradation activity. The compound of the invention is used as an active ingredient for preparing a new anticancer drug, and has potential application value.

Claims (1)

1. The phthalimide compound containing 1,2, 3-triazolyl is characterized in that: the compound has the following structural formula:
Figure 336610DEST_PATH_IMAGE001
I-2。
CN201911080765.XA 2019-11-07 2019-11-07 Phthalimide selective androgen receptor degradation agent and preparation method and application thereof Active CN110790750B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201911080765.XA CN110790750B (en) 2019-11-07 2019-11-07 Phthalimide selective androgen receptor degradation agent and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201911080765.XA CN110790750B (en) 2019-11-07 2019-11-07 Phthalimide selective androgen receptor degradation agent and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN110790750A CN110790750A (en) 2020-02-14
CN110790750B true CN110790750B (en) 2021-09-21

Family

ID=69443128

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201911080765.XA Active CN110790750B (en) 2019-11-07 2019-11-07 Phthalimide selective androgen receptor degradation agent and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN110790750B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4240733A1 (en) * 2020-11-06 2023-09-13 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of androgen receptor and associated methods of use

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20210132233A (en) * 2014-04-14 2021-11-03 아비나스 오퍼레이션스, 인코포레이티드 Imide-based modulators of proteolysis and associated methods of use
WO2016197032A1 (en) * 2015-06-04 2016-12-08 Arvinas, Inc. Imide-based modulators of proteolysis and associated methods of use
US20180147202A1 (en) * 2015-06-05 2018-05-31 Arvinas, Inc. TANK-BINDING KINASE-1 PROTACs AND ASSOCIATED METHODS OF USE
KR102173463B1 (en) * 2016-10-11 2020-11-04 아비나스 오퍼레이션스, 인코포레이티드 Compounds and methods for the targeted degradation of androgen receptor
US11220515B2 (en) * 2018-01-26 2022-01-11 Yale University Imide-based modulators of proteolysis and associated methods of use
CN110317192A (en) * 2018-03-28 2019-10-11 上海美志医药科技有限公司 One kind has the compound of degradation estrogen receptor activity

Also Published As

Publication number Publication date
CN110790750A (en) 2020-02-14

Similar Documents

Publication Publication Date Title
KR101287713B1 (en) Imidazole carboxamides
TWI406856B (en) Inhibitors of human immunodeficiency virus replication
FI112475B (en) Novel, optionally substituted phenylimidazolidines, process for their preparation, their use as pharmaceuticals and pharmaceutical compositions comprising them
Patil et al. Synthesis and antiulcer activity of 2-[5-substituted-1-H-benzo (d) imidazol-2-yl sulfinyl] methyl-3-substituted quinazoline-4-(3 H) ones
UA86196C2 (en) P38 inhibitors and methods of use thereof
JPH01279866A (en) Quinoline-based mevalonolactones
AU2005204428A1 (en) Conjugated small molecules
JPH07291939A (en) New (substituted) phenylimidazolidines, their production, use of them as medicines and pharmaceutical compositions containing them
JP2002514162A (en) Amidinoindoles, amidinoazoles, and analogs thereof as factor Xa inhibitors and thrombin inhibitors
WO2014183555A1 (en) Cycloalkyl acid derivative, preparation method thereof, and pharmaceutical application thereof
CN101189229A (en) Synthesis of avrainvillamide, stephacidin B, and analogues thereof
JP7406691B2 (en) Crystal forms and salt types of triazolopyrimidine compounds and their preparation methods
CN110790750B (en) Phthalimide selective androgen receptor degradation agent and preparation method and application thereof
US20190365768A1 (en) N-{[2-(piperidin-1-yl)phenyl](phenyl)methyl}-2-(3-oxo-3,4-dihydro-2h-1,4-benzoxa zin-7-yl)acetamide derivatives and related compounds as ror-gamma modulators for treating autoimmune diseases
CN1970548A (en) Medicinal methylene blue synthesis method
EP2160381A2 (en) Spirocyclic quinazoline derivatives and their use as pde7 inhibitors
BR112021000950A2 (en) substituted triazole quinoxaline derivatives
RU2198167C2 (en) Crystalline modifications of 1-(2,6-difluorobenzyl)-1h-1,2,3-trazol-4-carboxamide and pharmaceutical composition
CN110759897A (en) Triazole-containing hydrophobic adamantane type selective androgen receptor degradation agent and preparation method thereof
JP2004506043A (en) Method for producing cilostazol
CN102219803B (en) Preparation method of ezetimibe intermediate
CN110746400B (en) Androgen receptor targeted fluorescent probe and preparation method thereof
Ayyad et al. Anti-Inflammatory, Proton Pump Inhibitor and Synthesis of Some New Benzimidazole Derivatives
CN109020976A (en) Pyrimido triazole-Benzazole compounds and its preparation method and application
ES2264641B1 (en) PROCEDURE FOR OBTAINING DERIVATIVES OF BENCIMIDAZOL AND ITS INTERMEDIATES.

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