CN117603225A - Protein degradation targeting chimeric and preparation method thereof - Google Patents

Protein degradation targeting chimeric and preparation method thereof Download PDF

Info

Publication number
CN117603225A
CN117603225A CN202311620492.XA CN202311620492A CN117603225A CN 117603225 A CN117603225 A CN 117603225A CN 202311620492 A CN202311620492 A CN 202311620492A CN 117603225 A CN117603225 A CN 117603225A
Authority
CN
China
Prior art keywords
preparation
compound
protein degradation
ligand
solvent
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202311620492.XA
Other languages
Chinese (zh)
Inventor
董城
石磊
陈东星
孙吉跃
李艳冉
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.)
Tianjin Medical University
Original Assignee
Tianjin Medical 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 Tianjin Medical University filed Critical Tianjin Medical University
Priority to CN202311620492.XA priority Critical patent/CN117603225A/en
Publication of CN117603225A publication Critical patent/CN117603225A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/12Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D495/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

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

Abstract

The invention provides a protein degradation targeting chimeric body and a preparation method thereof, wherein the targeting chimeric body comprises a target protein ligand, an E3 ligase ligand and a connector, one end of the connector is connected with the target protein ligand, the other end of the connector is connected with the E3 ligase ligand, and the ligase ligand has the structural formula:the preparation method of the protein degradation targeting chimeric adopts a novel E3 ligase ligand, can obtain the protein degradation targeting chimeric compound with high yield and high purity, can effectively degrade various proteins in cells, and expands the species of the protein degradation targeting chimericThe class has wide application prospect.

Description

Protein degradation targeting chimeric and preparation method thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a protein degradation targeting chimeric and a preparation method thereof.
Background
Targeting Protein Degradation (TPD) is an emerging therapeutic approach that has been of interest because of its therapeutic potential to modulate proteins that are difficult to target by conventional small molecules. Protein degradation targeting chimeras (PROTACs) utilize the intracellular natural protein degradation system ubiquitin-proteasome system (UPS) to achieve targeted degradation of a protein of interest (POI). The PROTACs are heterobifunctional molecules and consist of three parts: the target protein ligand, the E3 ligase ligand, and the linking moiety connect the two. In cells, one end of the PROTACs molecule is combined with target protein, and the other end is combined with E3 ligase to form a target protein-PROTACs-E3 ligase ternary complex so as to bring the target protein and the E3 ligase closer, and the E3 ligase is promoted to catalyze ubiquitin protein to transfer to the target protein. The ubiquitinated target protein is then recognized and degraded by the proteasome, and the physiological functions carried by the target protein will disappear as the target protein dies. The physiological function of the target protein is regulated by degrading the target protein, and an unprecedented new way for researching and preparing biological medicine is opened up.
At present, only a few of these types of ligases, mainly VHL, CRBN, MDM, IAPs, are used for all PROTACs molecules. The main reason for this is the difficulty in developing new ligands that bind E3 ligase efficiently; and there is also a lack of compounds capable of undergoing efficient degradation.
Disclosure of Invention
In view of the above, the present invention aims to provide a protein degradation targeting chimeric body and a preparation method thereof, so as to provide a novel E3 ligase ligand as a part of the PROTACs to perform complete chemical synthesis of the PROTACs, and to effectively degrade a target protein.
In order to achieve the above purpose, the technical scheme of the invention is realized as follows:
the utility model provides a protein degradation target gomphosis body, includes target protein ligand, E3 ligase ligand and connector, the one end of connector links to each other with target protein ligand, and the other end links to each other with E3 ligase ligand, the structural formula of ligase ligand is:
further, the structural formula of the connector is selected from one of the following structural formulas:
wherein n is any integer from 1 to 6, m is any integer from 2 to 5, and x is 1 or 2.
Further, the structural formula of the target protein ligand is selected from one of the following structural formulas:
further, the structural formula of the protein degradation targeting chimera is selected from one of the following structural formulas:
the method for preparing the protein degradation targeting chimera, which comprises the following steps:
to the chloroacetyl chloride solution, (1S, 4S) -4-aminocyclohexane-1-carboxylic acid methyl ester hydrochloride and Et were slowly added dropwise 3 N solution was stirred overnight at room temperature, followed by successive treatment with 0.1M HCl solution, naHCO 3 The organic phase was washed with brine, dried over anhydrous Na 2 SO 4 Drying, and removing solvent under vacuumAn agent, yielding an oily residue; the residue was dissolved in MeCN and then (1H-indol-2-yl) methylamine, naHCO were added 3 And KI, rectifying for 12 hours, filtering, removing the solvent in vacuum, and purifying the residue by column chromatography to obtain a first intermediate;
dissolving the first intermediate in MeOH and water, adding LiOH-H 2 O, stirring overnight at room temperature, adjusting the pH value to 7, and removing the solvent under vacuum to obtain a second intermediate;
dissolving the second intermediate in 1, 4-dioxane and water, adding Na 2 CO 3 And Fmoc-Osu, stirring overnight at room temperature, adjusting pH to 6-7, removing solvent in vacuo, diluting the residue with water, adjusting pH to 4, extracting with ethyl acetate, combining organic layers, and extracting with anhydrous Na 2 SO 4 Drying, filtration, removal of solvent in vacuo, and recrystallization from EtOAc and petroleum ether gave the compound of formula:
compared with the prior art, the protein degradation targeting chimeric and the preparation method thereof have the following advantages:
the preparation method of the protein degradation targeting chimeric adopts a novel E3 ligase ligand, can obtain the protein degradation targeting chimeric compound with high yield and high purity, can effectively degrade various proteins in cells, expands the variety of the protein degradation targeting chimeric, and has wide application prospect.
Drawings
The accompanying drawings, which are included to provide a further understanding of the invention and are incorporated in and constitute a part of this specification, illustrate embodiments of the invention and together with the description serve to explain the invention. In the drawings:
FIG. 1 is a schematic diagram showing the degradation test results of application example 1;
FIG. 2 is a schematic diagram showing the degradation test results of application example 2;
FIG. 3 is a schematic diagram showing the degradation test results of application example 3;
FIG. 4 is a schematic diagram showing the degradation test results of application example 4;
FIG. 5 is a schematic diagram showing the degradation test results of application example 5.
Detailed Description
It should be noted that, without conflict, the embodiments of the present invention and features of the embodiments may be combined with each other.
The invention will be described in detail below with reference to the drawings in connection with embodiments.
Preparation example 1
To a solution of chloroacetyl chloride (431 ul,5.4mmol,1.5 eq.) in DCM at 0deg.C was slowly added dropwise methyl (1S, 4S) -4-aminocyclohexane-1-carboxylate hydrochloride (0.7 g,3.6mmol,1 eq.) and Et 3 A solution of N (1.5 mL,10.8mmol,3 eq.) in DCM. The mixture was then stirred at room temperature overnight. The solution was sequentially treated with 0.1M HCl solution, naHCO 3 The solution and brine were washed. The organic phase was treated with anhydrous Na 2 SO 4 Drying. The solvent was removed under vacuum to give an oily residue. The residue was dissolved in MeCN and then (1H-indol-2-yl) methylamine (789 mg,5.4mmol,1.5 eq.) NaHCO was added 3 (328 mg,10.8mmol,3 eq.) and KI (60 mg,0.36mmol,0.1 eq.). The mixture was distilled back for 12 hours. After filtration, the solvent was removed in vacuo and the residue was purified by column chromatography to give a first intermediate (1.2 g).
The first intermediate (1.2 g,3.49mmol,1 eq.) was dissolved in MeOH and water at 0deg.C, followed by addition of LiOH-H 2 O (1.5 g,34.9mmol,10 eq.). The mixture was then stirred at room temperature overnight. The pH of the mixture was adjusted to 7 at 0deg.C, and then the solvent was removed under vacuum to give a second intermediate. Dissolving the second intermediate in 1, 4-dioxane and water at 0deg.C, then adding Na 2 CO 3 And Fmoc-OSu (1.75 g,5.2mmol,1.5 eq.). The mixture was stirred at room temperature overnight. The pH of the mixture was adjusted to 6-7 at 0deg.C, and then the solvent was removed in vacuo. The residue was diluted with water and the pH of the mixture was adjusted to 4. The mixture was extracted with ethyl acetate, combined withThe machine layer is made of anhydrous Na 2 SO 4 And (5) drying. After filtration, the solvent was removed in vacuo. The residue was recrystallized from EtOAc and petroleum ether to give the compound of the formula (1.3 g, 65% yield):
1 H NMR(400MHz,DMSO-d 6 )δ12.04(s,1H),11.17(s,1H),11.07(s,1H),7.94–7.80(m,3H),7.63(d,J=7.4Hz,1H),7.50–7.39(m,3H),7.39–7.27(m,3H),7.14–7.01(m,2H),7.01–6.91(m,1H),6.29(s,1H),6.04(s,1H),4.59(d,J=20.1Hz,2H),4.37–4.20(m,3H),3.91(s,2H),3.82–3.67(m,1H),2.41–2.34(m,1H),1.90–1.75(m,2H),1.61–1.50(m,4H),1.50–1.38(m,2H).
HRMS(ESI)for C 33 H 34 N 3 O 5 [M+H] + calculated 552.2493,found 552.2491。
preparation example 2
To tert-butyl (3-hydroxypropyl) carbamate (1.00 g,5.70 mmol), et at 0deg.C 3 To a solution of N (1.59 mL,11.40 mmol) and DMAP (0.35 g,2.85 mmol) in DCM was slowly added 4-methylbenzenesulfonyl chloride (2.17 g,11.40 mmol). The mixture was then stirred at room temperature overnight. The solution was sequentially treated with 0.1M HCl solution, naHCO 3 The solution and brine were washed. The organic layer was treated with anhydrous Na 2 SO 4 And (5) drying. After filtration, the solvent was removed in vacuo and the residue was purified by column chromatography (PE/EA) to give a pale yellow oil (1.03 g, yield 55%). Pale yellow oil (1.03 g,3.13 mmol), 4-amino-3-nitrophenol (0.96 g,6.26 mmol) and K 2 CO 3 (0.87 g,6.26 mmol) in MeCN for 12 hours. After filtration, the solvent was removed in vacuo. The residue was washed with 0.1M NaOH solution and extracted with dichloromethane. The organic layer was treated with anhydrous Na 2 SO 4 And (5) drying. After filtration, the solvent was removed in vacuo and the residue was purified by column chromatography (PE/EA) to give the compound of formula (0.39 g, 40% yield):
1 H NMR(400MHz,Chloroform-d)δ7.54(d,J=3.0Hz,1H),7.06(dd,J=9.1,2.9Hz,1H),6.76(d,J=9.1Hz,1H),5.89(s,2H),4.71(brs,1H),3.99(t,J=5.9Hz,2H),3.32(q,J=6.5Hz,2H),1.97(p,J=6.4Hz,2H),1.44(s,9H).
preparation example 3
The difference from preparation 2 is that tert-butyl (3-hydroxypropyl) carbamate is replaced by tert-butyl (4-hydroxybutyl) carbamate, giving the compound of formula (yield 20%):
1 H NMR(400MHz,Chloroform-d)δ7.55–7.49(m,1H),7.10–7.01(m,1H),6.79–6.71(m,1H),5.88(s,2H),4.60(brs,1H),3.98–3.88(m,2H),3.24–3.10(m,2H),1.86–1.75(m,2H),1.71–1.61(m,2H),1.44(s,9H).
preparation example 4
The difference from preparation 2 is that tert-butyl (3-hydroxypropyl) carbamate is replaced by tert-butyl (5-hydroxypentyl) carbamate, giving the compound of the formula (yield 31%):
1 H NMR(400MHz,Chloroform-d)δ7.53(d,J=2.9Hz,1H),7.06(dd,J=9.1,2.9Hz,1H),6.75(d,J=9.2Hz,1H),5.88(s,2H),4.54(brs,1H),3.92(t,J=6.4Hz,2H),3.15(q,J=6.9,6.4Hz,2H),1.79(p,J=6.4Hz,2H),1.59–1.47(m,4H),1.45(s,9H).
preparation example 5
The difference from preparation 2 is that tert-butyl (3-hydroxypropyl) carbamate is replaced by tert-butyl (6-hydroxyhexyl) carbamate, giving the compound of formula (29% yield):
1 H NMR(400MHz,Chloroform-d)δ7.53(d,J=3.0Hz,1H),7.06(dd,J=9.0,2.9Hz,1H),6.75(d,J=9.2Hz,1H),5.86(s,2H),4.51(brs,1H),3.92(t,J=6.5Hz,2H),3.19–3.06(m,2H),1.82–1.72(m,2H),1.54–1.36(m,15H).
preparation example 6
The difference from preparation 2 is that tert-butyl (3-hydroxypropyl) carbamate is replaced by tert-butyl (7-hydroxyheptyl) carbamate, giving the compound of the formula (yield 22%):
1 H NMR(400MHz,Chloroform-d)δ7.53(d,J=2.9Hz,1H),7.06(dd,J=9.1,2.9Hz,1H),6.75(d,J=9.2Hz,1H),5.88(s,2H),4.50(brs,1H),3.92(t,J=6.5Hz,2H),3.12(q,J=6.4Hz,2H),1.82–1.70(m,2H),1.53–1.45(m,4H),1.44(s,9H),1.40–1.31(m,4H).
preparation example 7
The difference from preparation 2 is that tert-butyl (3-hydroxypropyl) carbamate is replaced by amino tert-butyl-di-polyethylene glycol, giving the compound of formula (yield 32%):
1 H NMR(400MHz,Chloroform-d)δ7.59(d,J=3.0Hz,1H),7.12(dd,J=9.1,2.9Hz,1H),6.76(d,J=9.1Hz,1H),5.88(s,2H),4.95(brs,1H),4.13–4.07(m,2H),3.84–3.77(m,2H),3.60(t,J=5.2Hz,2H),3.42–3.28(m,2H),1.44(s,9H).
preparation example 8
The difference from preparation 2 is that tert-butyl (3-hydroxypropyl) carbamate is replaced by amino tert-butyl-tri-polyethylene glycol, giving the compound of formula (yield 25%):
1 H NMR(400MHz,Chloroform-d)δ7.58(d,J=2.9Hz,1H),7.12(dd,J=9.1,2.9Hz,1H),6.76(d,J=9.0Hz,1H),5.89(s,2H),4.99(brs,1H),4.11–4.09(m,2H),3.89–3.81(m,2H),3.74–3.67(m,2H),3.67–3.62(m,2H),3.55(t,J=5.2Hz,2H),3.37–3.25(m,2H),1.43(s,9H).
preparation example 9
The difference from preparation 2 is that tert-butyl (3-hydroxypropyl) carbamate is replaced by amino tert-butyl-tetrapolyethylene glycol, giving the compound of formula (yield 32%):
1 H NMR(400MHz,Chloroform-d)δ7.57(d,J=2.9Hz,1H),7.11(dd,J=9.1,2.9Hz,1H),6.76(d,J=9.0Hz,1H),5.93(s,2H),5.05(brs,1H),4.12–4.09(m,2H),3.88–3.82(m,2H),3.75–3.60(m,8H),3.54(t,J=5.2Hz,2H),3.31(q,J=5.5Hz,2H),1.44(s,9H).
preparation example 10
The difference from preparation 2 is that tert-butyl (3-hydroxypropyl) carbamate is replaced by amino tert-butyl-pentapolyethylene glycol, giving the compound of formula (33% yield):
1 H NMR(400MHz,Chloroform-d)δ7.57(d,J=3.0Hz,1H),7.11(dd,J=9.1,2.9Hz,1H),6.76(d,J=9.0Hz,1H),5.92(s,2H),5.03(brs,1H),4.12–4.08(m,2H),3.88–3.81(m,2H),3.74–3.59(m,12H),3.53(t,J=5.2Hz,2H),3.31(q,J=5.8Hz,2H),1.44(s,9H).
PREPARATION EXAMPLE 11
A mixture of the compound from preparation 2 (0.39 g,1.25 mmol) and 10% Pd/C (wet) (78 mg) in MeOH was degassed in vacuo and then flushed with H 2 . The mixture was stirred at room temperature overnight. After filtration, the solvent was removed in vacuo. The crude product was used in the next step without purification. To a solution of the compound obtained in preparation 1 (120 mg,0.22 mmol), crude product (80 mg,0.28 mmol) and HBTU (107 mg,0.28 mmol) in MeCN was slowly added DIPEA (96 ml,0.55 mmol) at 0deg.C. The mixture was stirred at room temperature overnight. The solvent was removed in vacuo. The residue was dissolved with dichloromethane and washed with brine. The organic layer was treated with anhydrous Na 2 SO 4 And (5) drying. After filtration, the solvent was removed in vacuo. The residue was dissolved with HOAc and the mixture was stirred at 70 ℃ for 1 hour. After evaporation of the solvent in vacuo, the residue was purified by flash chromatography to give the compound of formula (119 mg, 68% yield):
1 H NMR(400MHz,Chloroform-d)δ9.38(s,1H),7.65(d,J=7.7Hz,2H),7.54–7.41(m,2H),7.39–7.26(m,4H),7.23–7.20(m,1H),7.15–6.99(m,4H),6.98–6.91(m,1H),6.82–6.73(m,1H),6.31(s,1H),6.18(s,1H),4.95–4.84(m,1H),4.49–4.33(m,3H),4.16–3.74(m,7H),3.33–3.15(m,2H),2.90–2.77(m,1H),1.96–1.78(m,4H),1.78–1.64(m,2H),1.61–1.47(m,4H),1.42(s,9H).
preparation example 12
The difference from preparation 11 is that the compound obtained in preparation 2 is replaced with the compound obtained in preparation 3 to give a compound of the following structural formula (yield 57%):
1 H NMR(400MHz,Chloroform-d)δ9.43(s,1H),7.71–7.62(m,2H),7.55–7.41(m,2H),7.40–7.27(m,4H),7.24–7.21(m,1H),7.20–6.87(m,5H),6.85–6.78(m,1H),6.19(s,1H),4.81–4.67(m,1H),4.64–4.27(m,4H),4.19–4.05(m,1H),4.00–3.73(m,5H),3.21–3.04(m,2H),2.90–2.74(m,1H),1.90–1.44(m,12H),1.43(s,9H)
preparation example 13
The difference from preparation 11 is that the compound obtained in preparation 2 was replaced with the compound obtained in preparation 4 to give a compound of the following structural formula (yield 58%):
1 H NMR(400MHz,Chloroform-d)δ9.41(s,1H),7.65(d,J=7.7Hz,2H),7.53–7.42(m,2H),7.41–7.26(m,4H),7.24–7.15(m,2H),7.14–6.95(m,4H),6.82(d,J=9.0Hz,1H),6.20(s,1H),4.76–4.54(m,2H),4.53–4.23(m,3H),4.19–4.04(m,1H),4.03–3.72(m,5H),3.20–2.99(m,2H),2.92–2.78(m,1H),1.96–1.81(m,2H),1.81–1.65(m,4H),1.64–1.45(m,8H),1.43(s,9H)
PREPARATION EXAMPLE 14
The difference from preparation 11 is that the compound obtained in preparation 2 is replaced with the compound obtained in preparation 5 to give the compound of the following structural formula (yield 56%):
1 H NMR(400MHz,Chloroform-d)δ9.37(s,1H),7.66(d,J=7.7Hz,2H),7.52–7.43(m,2H),7.42–7.37(m,1H),7.36–7.25(m,4H),7.14–6.96(m,5H),6.84(d,J=8.8Hz,1H),6.18(s,1H),4.63–4.59(m,1H),4.39(d,J=6.1Hz,2H),4.17–4.06(m,1H),4.04–3.76(m,6H),3.13–3.01(m,2H),2.88–2.77(m,1H),1.97–1.82(m,2H),1.76–1.64(m,4H),1.63–1.46(m,6H),1.44(s,9H),1.37–1.27(m,4H)
preparation example 15
The difference from preparation 11 is that the compound obtained in preparation 2 is replaced with the compound obtained in preparation 6 to give the compound of the following structural formula (yield 60%):
1 H NMR(400MHz,Chloroform-d)δ7.65(d,J=7.5Hz,2H),7.53–7.42(m,2H),7.41–7.30(m,2H),7.30–7.25(m,3H),7.23–7.16(m,1H),7.15–6.95(m,4H),6.88–6.81(m,1H),6.34(s,1H),6.19(s,1H),4.64–4.57(m,2H),4.38(d,J=6.3Hz,2H),4.17–3.75(m,6H),3.14–3.00(m,2H),2.87–2.79(m,1H),1.94–1.44(m,12H),1.43(s,9H),1.36–1.23(m,6H)
PREPARATION EXAMPLE 16
The difference from preparation 11 is that the compound obtained in preparation 2 is replaced with the compound obtained in preparation 7 to give the compound of the following structural formula (yield 62%):
1 H NMR(400MHz,Chloroform-d)δ7.70(d,J=7.4Hz,2H),7.53–7.40(m,3H),7.40–7.30(m,4H),7.22–6.98(m,5H),6.96–6.85(m,1H),6.40(s,1H),6.20(s,1H),5.00(brs,1H),4.67–4.59(m,1H),4.59–4.41(m,2H),4.38–4.28(m,1H),4.22–4.04(m,4H),4.01–3.76(m,5H),3.66–3.52(m,3H),3.41–3.28(m,2H),2.96–2.85(m,1H),1.86–1.68(m,4H),1.68–1.51(m,4H),1.44(s,9H).
preparation example 17
The difference from preparation 11 is that the compound obtained in preparation 2 is replaced with the compound obtained in preparation 8 to give a compound of the following structural formula (yield 58%):
1 H NMR(400MHz,Chloroform-d)δ7.70(d,J=7.3Hz,2H),7.59–7.42(m,3H),7.42–7.27(m,4H),7.24–6.99(m,5H),6.96–6.86(m,1H),6.20(s,1H),5.11–4.94(m,1H),4.68–4.42(m,3H),4.38–4.28(m,1H),4.26–4.06(m,3H),3.96–3.79(m,4H),3.77–3.67(m,2H),3.67–3.59(m,2H),3.55(t,J=5.1Hz,2H),3.36–3.27(m,2H),2.98–2.86(m,1H),1.90–1.70(m,4H),1.69–1.52(m,4H),1.43(s,9H).
HRMS(ESI)for C 50 H 59 N 6 O 8 [M+H] + calculated 871.4389,found 871.4389.
PREPARATION EXAMPLE 18
The difference from preparation 11 is that the compound obtained in preparation 2 is replaced with the compound obtained in preparation 9 to give the compound of the following structural formula (yield 55%):
1 H NMR(400MHz,Chloroform-d)δ7.66(d,J=7.8Hz,2H),7.48(d,J=9.0Hz,2H),7.41–7.27(m,4H),7.24–7.15(m,2H),7.15–6.95(m,5H),6.91–6.80(m,1H),6.19(s,1H),5.27(s,1H),5.11(t,J=5.8Hz,1H),4.61(s,1H),4.39(d,J=6.3Hz,2H),4.14–4.02(m,3H),4.02–3.96(m,1H),3.92(s,2H),3.89–3.77(m,3H),3.73–3.52(m,10H),3.48(t,J=5.2Hz,2H),3.30–3.19(m,2H),2.94–2.79(m,1H),2.00–1.84(m,2H),1.82–1.67(m,2H),1.65–1.45(m,4H),1.42(s,9H)
preparation example 19
The difference from preparation 11 is that the compound obtained in preparation 2 was replaced with the compound obtained in preparation 10 to give a compound of the following structural formula (yield 51%):
1 H NMR(400MHz,Chloroform-d)δ7.73–7.61(m,2H),7.51–7.44(m,2H),7.43–7.37(m,1H),7.35–7.27(m,3H),7.24–7.16(m,2H),7.15–7.05(m,2H),7.05–7.01(m,2H),6.82(d,J=8.8Hz,1H),6.19(s,1H),5.16–5.06(m,1H),4.66–4.53(m,1H),4.44–4.35(m,3H),4.11–4.04(m,2H),3.94(s,2H),3.84–3.77(m,2H),3.71–3.66(m,2H),3.66–3.58(m,6H),3.58–3.49(m,5H),3.47–3.37(m,2H),3.26–3.15(m,2H),2.95–2.82(m,1H),1.97–1.83(m,2H),1.83–1.70(m,2H),1.63–1.49(m,4H),1.42(s,9H)
preparation example 20
To a solution of the compound produced in preparation 11 (60 mg,0.07 mmol) in DCM at 0deg.C was slowly added dropwise trifluoroacetic acid. The mixture was stirred at 0℃for 1 hour. The solvent was then removed under vacuum, the residue was dissolved in DMF and JQ1-COOH (32 mg,0.08 mmol), HATU (42 mg,0.11 mmol) and DIPEA (61. Mu.L, 0.35 mmol) were added. The reaction was stirred at room temperature overnight. The mixture was diluted with EtOAc and washed with brine. The organic phase was purified by Na 2 SO 4 Drying. After removal of the solvent in vacuo, the residue was purified by column chromatography to afford the intermediate. Piperidine was slowly added dropwise to the intermediate in DCM and MeCN solution to remove Fmoc groups. After stirring at room temperature for 2 hours, the solvent was removed in vacuo and the residue was purified by HPLC to give a compound of formula (30% yield):
1 H NMR(400MHz,Chloroform-d)δ10.35(s,1H),7.57–7.49(m,2H),7.31(d,J=8.7Hz,3H),7.25–7.21(m,3H),7.07–6.99(m,2H),6.76(dd,J=8.8,2.4Hz,1H),6.37–6.32(m,1H),4.71(dd,J=9.3,4.6Hz,1H),4.22(s,1H),4.00(s,1H),3.91(s,2H),3.82–3.72(m,2H),3.44–3.29(m,3H),3.27(s,2H),2.87–2.74(m,1H),2.36(s,3H),2.28(s,3H),2.03–1.85(m,4H),1.76–1.66(m,4H),1.59(s,3H),1.57–1.49(m,2H).
HRMS(ESI)for C 46 H 50 ClN 10 O 3 S[M+H] + calculated 857.3477,found 857.3486
preparation example 21
The difference from preparation 20 is that the compound prepared in preparation 11 is replaced with the compound prepared in preparation 12 to give the compound of the following formula (yield 35%):
1 H NMR(400MHz,Chloroform-d)δ10.30(s,1H),7.51–7.45(m,2H),7.37(d,J=8.5Hz,3H),7.29–7.27(m,1H),7.25–7.24(m,1H),7.22–7.16(m,2H),7.03–6.95(m,2H),6.86(brs,1H),6.76–6.69(m,1H),6.28(s,1H),4.66(t,J=6.9Hz,1H),4.00–3.91(m,1H),3.88–3.75(m,4H),3.63–3.53(m,1H),3.43–3.34(m,1H),3.33–3.22(m,2H),3.17(s,2H),2.92–2.81(m,1H),2.59(s,3H),2.34(s,3H),2.02–1.89(m,2H),1.86–1.62(m,8H),1.61(s,3H),1.57–1.46(m,2H).
HRMS(ESI)for C 47 H 52 ClN 10 O 3 S[M+H] + calculated 871.3628,found 871.3623.
PREPARATION EXAMPLE 22
The difference from preparation 20 is that the compound prepared in preparation 11 is replaced with the compound prepared in preparation 13 to give the compound of the following formula (yield 34%):
1 H NMR(400MHz,Chloroform-d)δ11.35(brs,1H),10.32(s,1H),7.57–7.46(m,2H),7.37(d,J=8.7Hz,2H),7.28(d,J=8.8Hz,3H),7.24–7.18(m,1H),7.06–6.96(m,2H),6.76(dd,J=8.8,2.4Hz,1H),6.30(d,J=2.1Hz,1H),4.67(t,J=7.0Hz,1H),4.02–3.97(m,1H),3.88–3.81(m,4H),3.63–3.53(m,1H),3.49(s,1H),3.45–3.36(m,1H),3.31–3.23(m,2H),3.20(s,2H),2.94–2.84(m,1H),2.60(s,3H),2.34(s,3H),2.04–1.91(m,2H),1.86–1.79(m,2H),1.77–1.65(m,4H),1.61(s,3H),1.59–1.49(m,4H),1.49–1.38(m,2H).
HRMS(ESI)for C 48 H 54 ClN 10 O 3 S[M+H] + calculated 885.3790,found 885.3800
preparation example 23
The difference from preparation 20 is that the compound prepared in preparation 11 was replaced with the compound prepared in preparation 14 to give the compound of the following formula (yield 29%):
1 H NMR(400MHz,Chloroform-d)δ10.19(s,1H),7.54–7.46(m,2H),7.43–7.34(m,3H),7.30(d,J=8.4Hz,3H),7.24–7.19(m,1H),7.05–6.98(m,2H),6.93–6.85(m,2H),6.78(d,J=8.3Hz,1H),6.32(s,1H),4.65(t,J=6.9Hz,1H),4.03–3.99(m,1H),3.92–3.84(m,4H),3.55(dd,J=14.4,7.0Hz,1H),3.39(dd,J=14.6,6.3Hz,1H),3.30–3.21(m,4H),2.96–2.87(m,1H),2.61(s,3H),2.35(s,3H),2.04–1.93(m,2H),1.89–1.81(m,2H),1.78–1.68(m,4H),1.54(dd,J=14.1,9.0Hz,4H),1.45–1.32(m,4H).
HRMS(ESI)for C 49 H 56 ClN 10 O 3 S[M+H] + calculated 899.3941,found 899.3940
PREPARATION EXAMPLE 24
The difference from preparation 20 is that the compound prepared in preparation 11 is replaced with the compound prepared in preparation 15 to give the compound of the following formula (yield: 31%):
1 H NMR(400MHz,Chloroform-d)δ10.30(s,1H),7.57–7.46(m,2H),7.43–7.34(m,3H),7.30(d,J=8.8Hz,2H),7.25–7.18(m,1H),7.07–6.97(m,2H),6.95–6.89(m,1H),6.79(dd,J=8.8,2.4Hz,1H),6.33–6.28(m,1H),4.64(t,J=7.0Hz,1H),4.03–3.98(m,1H),3.91–3.82(m,4H),3.55(dd,J=14.6,7.3Hz,1H),3.49(s,1H),3.37(dd,J=14.6,6.5Hz,1H),3.32–3.18(m,4H),2.96–2.85(m,1H),2.62(s,3H),2.35(s,3H),2.05–1.91(m,2H),1.89–1.80(m,2H),1.78–1.66(m,4H),1.61(s,3H),1.58–1.44(m,4H),1.43–1.34(m,2H),1.33–1.26(m,4H).
HRMS(ESI)for C 50 H 58 ClN 10 O 3 S[M+H] + calculated 913.4097,found 913.4094
preparation example 25
The difference from preparation 20 is that the compound prepared in preparation 11 is replaced with the compound prepared in preparation 16 to give the compound of the following formula (yield: 30%):
1 H NMR(400MHz,Chloroform-d)δ10.22(s,1H),7.58(d,J=8.3Hz,1H),7.54–7.47(m,1H),7.41(d,J=8.5Hz,2H),7.34–7.27(m,3H),7.25–7.20(m,2H),7.08–6.95(m,2H),6.95–6.79(m,2H),6.32(d,J=1.8Hz,1H),4.68(t,J=7.1Hz,1H),4.17(t,J=4.5Hz,2H),4.05(s,1H),3.89(s,2H),3.84–3.72(m,2H),3.68–3.53(m,3H),3.53–3.39(m,5H),3.25(s,2H),3.00–2.86(m,2H),2.61(s,3H),2.39(s,3H),2.09–1.93(m,2H),1.90–1.80(m,2H),1.79–1.70(m,2H),1.66(s,3H),1.63–1.53(m,2H).
13 C NMR(101MHz,CDCl 3 )δ170.71,170.30,163.99,158.53,155.87,155.10,150.01,136.97,136.60,136.33,131.76,131.27,130.97,130.71,129.90,128.72,128.35,121.40,120.08,119.39,111.25,100.99,69.82,69.68,68.90,54.18,51.90,47.09,44.17,39.34,38.85,36.62,29.23,26.60,14.38,13.09,11.68.
HRMS(ESI)for C 47 H 52 ClN 10 O 4 S[M+H] + calculated 887.3577,found 887.3580
PREPARATION EXAMPLE 26
The difference from preparation 20 is that the compound prepared in preparation 11 is replaced with the compound prepared in preparation 17 to give the compound of the following formula (yield 28%):
1 H NMR(400MHz,Chloroform-d)δ11.74(brs,1H),10.25(s,1H),7.54(d,J=7.8Hz,1H),7.52–7.47(m,1H),7.43–7.36(m,2H),7.31(d,J=8.8Hz,2H),7.21(dd,J=7.0,3.7Hz,2H),7.08–6.96(m,3H),6.81(dd,J=8.8,2.5Hz,1H),6.31(d,J=2.1Hz,1H),4.71(t,J=6.9Hz,1H),4.22–4.09(m,2H),4.09–3.99(m,1H),3.87(s,2H),3.83(t,J=5.0Hz,2H),3.75–3.63(m,4H),3.63–3.54(m,2H),3.54–3.42(m,4H),3.25(s,2H),3.02–2.93(m,1H),2.64(s,3H),2.39(s,3H),2.12–1.97(m,2H),1.94–1.83(m,2H),1.81–1.70(m,2H),1.65(s,3H),1.63–1.53(m,2H).
13 C NMR(101MHz,CDCl 3 )δ170.79,170.45,164.03,155.85,155.04,149.90,136.97,136.90,136.62,136.49,131.83,131.19,131.01,130.69,129.90,128.72,128.33,121.36,120.05,119.35,111.25,100.97,70.65,70.18,69.82,69.43,68.30,54.17,51.91,47.08,44.32,39.44,38.74,36.43,29.22,26.70,26.61,14.37,13.08,11.74.
HRMS(ESI)for C 49 H 56 ClN 10 O 5 S[M+H] + calculated 931.3839,found 931.3841
preparation example 27
The difference from preparation 20 is that the compound prepared in preparation 11 was replaced with the compound prepared in preparation 18 to give the compound of the following formula (yield: 32%):
1 H NMR(400MHz,Chloroform-d)δ11.71(brs,1H),10.34(s,1H),7.58–7.45(m,3H),7.40(d,J=8.5Hz,2H),7.30(d,J=8.7Hz,2H),7.24–7.18(m,1H),7.07–6.97(m,3H),6.80(d,J=8.3Hz,1H),6.30(d,J=2.0Hz,1H),4.69(t,J=7.0Hz,1H),4.09(t,J=5.0Hz,2H),4.04–3.98(m,1H),3.85(s,2H),3.83–3.78(m,2H),3.75–3.56(m,9H),3.56–3.49(m,3H),3.48(s,2H),3.43–3.33(m,2H),3.22(s,2H),2.97–2.86(m,1H),2.63(s,3H),2.37(s,3H),2.04–1.95(m,2H),1.91–1.80(m,2H),1.76–1.67(m,2H),1.63(s,3H),1.61–1.52(m,2H).
13 C NMR(101MHz,CDCl 3 )δ170.87,170.68,164.04,155.80,155.09,149.90,137.04,136.86,136.63,136.51,131.95,131.15,130.90,130.51,129.89,128.72,128.33,121.34,120.05,119.34,118.77,111.21,100.94,96.65,70.77,70.63,70.30,69.79,69.73,68.28,54.29,51.87,50.19,47.05,44.32,39.54,38.76,36.41,29.21,26.72,26.59,14.36,13.07,11.75.
HRMS(ESI)for C 51 H 60 ClN 10 O 6 S[M+H] + calculated 975.4107,found 975.4111
PREPARATION EXAMPLE 28
The difference from preparation 20 is that the compound prepared in preparation 11 is replaced with the compound prepared in preparation 19 to give the compound of the following formula (yield 34%):
1 H NMR(400MHz,Chloroform-d)δ11.47(brs,1H),10.32(s,1H),7.55(d,J=7.8Hz,1H),7.49(dt,J=7.4,2.8Hz,1H),7.42–7.36(m,2H),7.31(d,J=8.8Hz,2H),7.25–7.16(m,2H),7.06–6.92(m,3H),6.80(dd,J=8.7,2.4Hz,1H),6.29(d,J=2.1Hz,1H),4.67(t,J=7.0Hz,1H),4.13–3.96(m,4H),3.84(s,2H),3.80(t,J=4.8Hz,2H),3.73–3.67(m,2H),3.67–3.61(m,6H),3.61–3.57(m,3H),3.57–3.50(m,4H),3.50–3.42(m,4H),3.22(s,2H),2.97–2.87(m,1H),2.62(s,3H),2.36(s,3H),2.05–1.93(m,2H),1.89–1.80(m,2H),1.75–1.66(m,2H),1.62(s,3H),1.60–1.51(m,2H).
HRMS(ESI)for C 53 H 63 ClN 10 O 7 SNa[M+Na] + calculated 1041.4183,found 1041.4176
preparation example 29
The difference from preparation 2 is that tert-butyl (3-hydroxypropyl) carbamate is replaced by tert-butyl-4- (2-hydroxyethyl) piperazine-1-carboxylate, giving the compound of the formula (yield 45%):
1 H NMR(400MHz,Chloroform-d)δ7.57(d,J=3.0Hz,1H),7.09(dd,J=9.1,2.9Hz,1H),6.76(d,J=9.2Hz,1H),5.91(s,2H),4.07(t,J=5.6Hz,2H),3.46(t,J=5.1Hz,4H),2.80(t,J=5.6Hz,2H),2.51(t,J=5.1Hz,4H),1.46(s,9H)
preparation example 30
The difference from preparation 29 is that tert-butyl-4- (2-hydroxyethyl) piperazine-1-carboxylate is replaced by tert-butyl-4- (3-hydroxypropyl) piperazine-1-carboxylate, giving the compound of the formula (yield 36%):
1 H NMR(400MHz,Chloroform-d)δ7.55(d,J=3.0Hz,1H),7.06(dd,J=9.0,2.9Hz,1H),6.75(d,J=9.2Hz,1H),5.88(s,2H),3.99(t,J=6.3Hz,2H),3.44(t,J=5.2Hz,4H),2.52(t,J=7.3Hz,2H),2.41(t,J=5.1Hz,4H),1.96(p,J=6.5Hz,2H),1.46(s,9H).
preparation example 31
The difference from preparation 11 is that the compound from preparation 2 is replaced by the compound from preparation 29, giving the compound of formula (yield 51%):
1 H NMR(400MHz,Chloroform-d)δ9.14(s,1H),7.66(d,J=7.7Hz,2H),7.53–7.42(m,2H),7.42–7.33(m,1H),7.33–7.26(m,3H),7.24–7.22(m,1H),7.20–6.96(m,5H),6.88–6.75(m,1H),6.18(s,1H),4.63–4.54(m,1H),4.45–4.31(m,3H),4.14–4.00(m,3H),3.99–3.94(m,1H),3.86(s,2H),3.49–3.37(m,4H),2.90–2.82(m,1H),2.82–2.69(m,2H),2.55–2.42(m,4H),1.93–1.65(m,4H),1.64–1.48(m,4H),1.46(s,9H)
PREPARATION EXAMPLE 32
The difference from preparation 31 is that the compound from preparation 29 is replaced by the compound from preparation 30, giving the compound of formula (54% yield):
1 H NMR(400MHz,Chloroform-d)δ7.66(d,J=7.8Hz,2H),7.52–7.41(m,2H),7.37–7.26(m,4H),7.24–7.22(m,1H),7.17–6.97(m,5H),6.88–6.78(m,1H),6.17(s,1H),4.61–4.56(m,1H),4.44–4.27(m,3H),4.02–3.79(m,6H),3.45–3.37(m,4H),2.91–2.75(m,1H),2.53–2.43(m,2H),2.41–2.34(m,4H),1.99–1.73(m,5H),1.65–1.50(m,5H),1.46(s,9H)
PREPARATION EXAMPLE 33
The difference from preparation 20 is that the compound prepared in preparation 11 is replaced with the compound prepared in preparation 31 to give the compound of the following formula (yield: 30%):
1 H NMR(400MHz,Chloroform-d)δ10.38(s,1H),7.64–7.57(m,1H),7.53–7.46(m,1H),7.43–7.37(m,3H),7.35–7.28(m,3H),7.24–7.19(m,1H),7.05–6.97(m,3H),6.85–6.78(m,1H),6.31(s,1H),5.29(s,1H),4.81(t,J=6.7Hz,1H),4.06–3.96(m,3H),3.85(s,2H),3.78–3.60(m,4H),3.60–3.48(m,3H),3.24(s,2H),2.98–2.88(m,1H),2.78–2.70(m,2H),2.63(s,3H),2.59–2.41(m,4H),2.37(s,3H),2.08–1.96(m,2H),1.90–1.83(m,2H),1.79–1.72(m,2H),1.64(s,3H),1.60–1.51(m,2H).
HRMS(ESI)for C 49 H 55 ClN 11 O 3 S[M+H] + calculated 912.3893,found 912.3897
PREPARATION EXAMPLE 34
The difference from preparation 33 is that the compound obtained in preparation 31 was replaced with the compound obtained in preparation 32 to give a compound of the following structural formula (yield: 28%):
1 H NMR(400MHz,Methanol-d 4 )δ7.49–7.36(m,6H),7.22(d,J=8.0Hz,1H),7.09–7.04(m,1H),7.03–6.95(m,1H),6.95–6.85(m,2H),6.30(s,1H),4.73–4.66(m,1H),4.64–4.59(m,1H),4.11(t,J=6.2Hz,2H),3.98–3.92(m,1H),3.91(s,2H),3.84–3.71(m,2H),3.68–3.61(m,2H),3.61–3.51(m,2H),3.29(s,2H),3.05–2.94(m,1H),2.70(s,3H),2.67–2.60(m,4H),2.58–2.48(m,2H),2.45(s,3H),2.12–1.98(m,4H),1.96–1.86(m,2H),1.70(s,3H),1.69–1.64(m,4H).
HRMS(ESI)for C 50 H 56 ClN 11 O 3 SNa[M+Na] + calculated 948.3869,found 948.3867
preparation example 35
To a solution of the compound produced in preparation 11 (60 mg,0.07 mmol) in DCM at 0deg.C was slowly added dropwise trifluoroacetic acid. The mixture was stirred at 0℃for 1 hour. The solvent was then removed in vacuo, the residue was dissolved in DMF and 2- (4- (1, 2-diphenylbut-1-en-1-yl) phenoxy) acetic acid (32 mg,0.08 mmol), HATU (42 mg,0.11 mmol) and DIPEA (61. Mu.L, 0.35 mmol) were added. The reaction was stirred at room temperature overnight. The mixture was diluted with EtOAc and washed with brine. The organic phase was purified by Na 2 SO 4 And (5) drying. After removal of the solvent in vacuo, the residue was purified by column chromatography to afford the intermediate. Piperidine was slowly added dropwise to a solution of intermediate in DCM and MeCN to remove Fmoc groups. After stirring at room temperature for 2 hours, the solvent was removed in vacuo and the residue was purified by HPLC to give a compound of formula (31% yield):
1 H NMR(400MHz,Chloroform-d)δ10.00(d,J=28.9Hz,1H),9.69(d,J=21.5Hz,1H),7.56–7.46(m,3H),7.23–6.93(m,15H),6.91–6.83(m,4H),6.39–6.34(m,1H),4.54–4.46(m,2H),4.07–3.96(m,2H),3.96–3.85(m,3H),3.64–3.48(m,2H),3.26(s,2H),2.97–2.84(m,1H),2.46(q,J=6.4,5.9Hz,2H),2.09–1.95(m,4H),1.94–1.79(m,5H),1.69–1.57(m,2H),0.92(t,J=7.4Hz,3H).
HRMS(ESI)for C 51 H 55 N 6 O 4 [M+H] + calculated 815.4279,found 815.4276
preparation example 36
The difference from preparation 35 is that the compound produced in preparation 11 was replaced with the compound produced in preparation 16 to give the compound of the following structural formula (yield: 31%):
1 H NMR(400MHz,Chloroform-d)δ9.94(s,1H),7.44–7.35(m,2H),7.29–7.16(m,3H),7.09(d,J=7.0Hz,2H),7.05–6.93(m,6H),6.93–6.83(m,4H),6.81–6.61(m,5H),6.37(d,J=8.5Hz,1H),6.21(s,1H),4.35(s,1H),4.18(s,1H),3.94–3.84(m,3H),3.76(s,2H),3.65–3.54(m,2H),3.52–3.47(m,1H),3.47–3.40(m,2H),3.40–3.33(m,2H),3.14(s,2H),2.77–2.72(m,1H),2.38–2.26(m,2H),1.95–1.79(m,2H),1.78–1.57(m,4H),1.52–1.38(m,2H),1.13(s,2H),0.78(t,J=7.4Hz,3H).
HRMS(ESI)for C 52 H 57 N 6 O 5 [M+H] + calculated 845.4385,found 845.4389
preparation example 37
To a solution of the compound produced in preparation 11 (50 mg,0.06 mmol) in DCM at 0deg.C was slowly added dropwise trifluoroacetic acid. The mixture was stirred at 0℃for 1 hour. The solvent was then removed in vacuo and the residue was dissolved in DMF and 2- (4- (3-amino-6- (2-hydroxyphenyl) pyridazin-4-yl) piperazin-1-yl) acetic acid (23 mg,0.06 mmol), HATU (38 mg,0.10 mmol) and DIPEA (55. Mu.L, 0.32 mmol) were added. The reaction was stirred at room temperature overnight. The mixture was diluted with EtOAc and washed with brine. The organic phase was purified by Na 2 SO 4 And (5) drying. After removal of the solvent in vacuo, the residue was purified by column chromatography to afford the intermediate. Piperidine was slowly added dropwise to a solution of intermediate in DCM and MeCN to remove Fmoc groups. After stirring at room temperature for 2 hours, the solvent was removed in vacuo and the residue was purified by HPLC to give a compound of the formula (yield 34%)):
1 H NMR(400MHz,Methanol-d 4 )δ7.56–7.49(m,1H),7.33–7.24(m,2H),7.20–7.14(m,2H),7.14–7.07(m,1H),6.99(s,1H),6.92–6.77(m,6H),6.18(s,1H),4.02(t,J=5.8Hz,2H),3.85–3.81(m,1H),3.80(s,2H),3.43(t,J=6.5Hz,2H),3.19(s,2H),3.01(s,2H),2.98–2.90(m,4H),2.81–2.70(m,1H),2.63–2.56(m,4H),1.97(p,J=6.1Hz,2H),1.91–1.78(m,2H),1.78–1.66(m,2H),1.57–1.48(m,4H).
HRMS(ESI)for C 52 H 57 N 6 O 5 [M+H] + calculated 786.4198,found 786.4200
Preparation example 38
The difference from preparation 37 is that the compound prepared in preparation 11 is replaced by the compound prepared in preparation 16, giving the compound of the formula (yield: 30%):
1 H NMR(400MHz,Methanol-d 4 )δ7.58(d,J=8.3Hz,1H),7.38(d,J=7.7Hz,1H),7.32–7.17(m,4H),7.05(s,1H),6.98(t,J=7.4Hz,1H),6.94–6.82(m,4H),6.28(s,1H),4.20–4.14(m,2H),3.96–3.84(m,5H),3.70(t,J=5.2Hz,2H),3.52(t,J=5.2Hz,2H),3.30(s,2H),3.10(s,2H),3.07–2.98(m,4H),2.92–2.82(m,1H),2.72–2.65(m,4H),2.02–1.90(m,2H),1.88–1.79(m,2H),1.72–1.58(m,4H).
HRMS(ESI)for C 44 H 54 N 11 O 5 [M+H] + calculated 816.4304,found 816.4309
preparation example 39
The difference from preparation 26 is that JQ1-COOH is replaced by 2- (4- (3-amino-6- (2-hydroxyphenyl) pyridazin-4-yl) piperazin-1-yl) acetic acid to give a compound of the formula (yield 29%):
1 H NMR(400MHz,Methanol-d 4 )δ7.75–7.69(m,1H),7.44–7.35(m,2H),7.35–7.18(m,3H),7.02–6.87(m,5H),6.82(dd,J=8.7,2.4Hz,1H),6.29(s,1H),4.14(t,J=4.5Hz,2H),3.96–3.84(m,5H),3.79–3.73(m,2H),3.73–3.66(m,2H),3.62(t,J=5.2Hz,2H),3.47(t,J=5.2Hz,2H),3.30(s,2H),3.20–3.12(m,4H),3.10(s,2H),3.01–2.91(m,1H),2.75–2.68(m,4H),2.08–1.96(m,2H),1.94–1.85(m,2H),1.72–1.62(m,4H).
HRMS(ESI)for C 46 H 58 N 11 O 6 [M+H] + calculated 860.4566,found 860.4565
preparation example 40
The difference from preparation 27 is that JQ1-COOH is replaced with 2- (4- (3-amino-6- (2-hydroxyphenyl) pyridazin-4-yl) piperazin-1-yl) acetic acid to give a compound of the formula (yield 27%):
1 H NMR(400MHz,Methanol-d 4 )δ7.77(d,J=8.0Hz,1H),7.48(s,1H),7.39(d,J=7.9Hz,2H),7.29–7.18(m,2H),7.02–6.88(m,5H),6.83(dd,J=8.7,2.4Hz,1H),6.30(s,1H),4.14–4.08(m,2H),3.92(s,2H),3.82(t,J=4.4Hz,2H),3.73–3.61(m,9H),3.58(t,J=5.3Hz,2H),3.44(t,J=5.1Hz,2H),3.30(s,2H),3.23–3.14(m,4H),3.10(s,2H),3.01–2.95(m,1H),2.77–2.68(m,4H),2.10–1.97(m,2H),1.95–1.86(m,2H),1.73–1.63(m,4H).
HRMS(ESI)for C 48 H 62 N 11 O 7 [M+H] + calculated 904.4828,found 904.4823.HRMS(ESI)for C48H61N11O7Na[M+Na] + calculated 926.4648,found 926.4641
PREPARATION EXAMPLE 41
The difference from preparation 20 is that JQ1-COOH is replaced by 2- (4- (3-amino-6- (2-hydroxyphenyl) pyridazin-4-yl) piperazin-1-yl) acetic acid to give a compound of the formula (yield 33%):
1 H NMR(400MHz,Methanol-d 4 )δ7.84–7.77(m,1H),7.53(s,1H),7.46–7.36(m,2H),7.30–7.18(m,2H),7.05(s,1H),7.02–6.84(m,5H),6.30(s,1H),4.11(t,J=6.1Hz,2H),3.91(d,J=10.2Hz,3H),3.74–3.69(m,2H),3.68–3.63(m,2H),3.35(s,2H),3.30(s,2H),3.26–3.18(m,4H),3.04–2.95(m,1H),2.80–2.72(m,4H),2.65(t,J=7.3Hz,2H),2.62–2.57(m,2H),2.56–2.50(m,2H),2.11–1.99(m,4H),1.96–1.87(m,2H),1.72–1.64(m,4H).
HRMS(ESI)for C 47 H 59 N 12 O 4 [M+H] + calculated 855.4777,found 855.4780
application example 1
The compounds prepared in preparation 25 and preparation 34 were tested for their degradation effect on target proteins by immunoblotting experiments.
The detection method comprises the following steps:
u2OS cells were treated with DMSO solvent or different concentrations of compounds for 8 hours. BRD2, BRD4 (long), BRD4 (short) proteins and control β -actin were detected by immunoblotting, and the detection results are shown in fig. 1.
Application example 2
The degradation effect of the compound prepared in preparation example 35 on the target protein is detected by immunoblotting experiment.
The detection method comprises the following steps:
MCF-7 cells were treated with DMSO solvents or compounds at various concentrations for 18 hours. The ERalpha protein and the control beta-actin were detected by immunoblotting, and the detection results are shown in FIG. 2.
Application example 3
The degradation effect of the compound prepared in preparation 39 on the target protein is detected by immunoblotting experiment.
The detection method comprises the following steps:
u2OS cells were treated with DMSO solvent or different concentrations of compounds for 8 hours. The SMARCA2 protein and the control beta-actin were detected by immunoblotting, and the detection results are shown in FIG. 3.
Application example 4
The compounds prepared in preparation examples 20 to 28 were tested for the degradation effect on the target protein by immunoblotting experiments.
The detection method comprises the following steps:
u2OS cells were treated with DMSO solvent or compound at a concentration of 500nM for 8 hours. BRD4 (long), BRD4 (short) protein and control beta-actin were detected by immunoblotting, and the detection results are shown in FIG. 4.
Application example 5
The compounds prepared in preparation 33 and preparation 34 were tested for their degradation effect on target proteins by immunoblotting experiments.
The detection method comprises the following steps:
u2OS cells were treated with DMSO solvent or compound at a concentration of 500nM for 8 hours. BRD4 (long), BRD4 (short) protein and control beta-actin were detected by immunoblotting, and the detection results are shown in FIG. 5.
From the detection results shown in fig. 1-5, the protein degradation targeting chimeric body prepared by the invention can effectively degrade BRD2, BRD4 (long), BRD4 (short), ER alpha and SMARCA2 proteins, and can also carry out further research on degradation of other proteins, including but not limited to EGFR, WDR5, SMURF1 and PI3K, KRAS, TGF-TGF-beta, and the protein degradation targeting chimeric body prepared by the invention has wide application prospects in the field of protein degradation.

Claims (5)

1. A protein degradation targeting chimera, characterized in that: the kit comprises a target protein ligand, an E3 ligase ligand and a connector, wherein one end of the connector is connected with the target protein ligand, the other end of the connector is connected with the E3 ligase ligand, and the structural formula of the ligase ligand is as follows:
2. the protein degradation targeting chimera of claim 1, wherein: the structural formula of the connector is selected from one of the following structural formulas:
wherein n is any integer from 1 to 6, m is any integer from 2 to 5, and x is 1 or 2.
3. The protein degradation targeting chimera of claim 1, wherein: the structural formula of the target protein ligand is selected from one of the following structural formulas:
4. the protein degradation targeting chimera of claim 1, wherein: the structural formula of the protein degradation targeting chimeric body is selected from one of the following structural formulas:
5. the method for preparing a protein degradation targeting chimera according to any one of claims 1-4, characterized in that it comprises the following steps:
to the chloroacetyl chloride solution, (1S, 4S) -4-aminocyclohexane-1-carboxylic acid methyl ester hydrochloride and Et were slowly added dropwise 3 N solution was stirred overnight at room temperature, followed by successive treatment with 0.1M HCl solution, naHCO 3 The organic phase was washed with brine, dried over anhydrous Na 2 SO 4 Drying, and removing the solvent under vacuum to obtain oily residue; the residue was dissolved in MeCN and then (1H-indol-2-yl) methylamine, naHCO were added 3 And KI, rectifying for 12 hours, filtering, removing the solvent in vacuum, and purifying the residue by column chromatography to obtain a first intermediate;
dissolving the first intermediate in MeOH and water, adding LiOH-H 2 O, stirring overnight at room temperature, adjusting the pH value to 7, and removing the solvent under vacuum to obtain a second intermediate;
dissolving the second intermediate in 1, 4-dioxane and water, adding Na 2 CO 3 And Fmoc-Osu, stirring overnight at room temperature, adjusting pH to 6-7, removing solvent in vacuo, diluting the residue with water, adjusting pH to 4, extracting with ethyl acetate, combining organic layers, and extracting with anhydrous Na 2 SO 4 Drying, filtration, removal of solvent in vacuo, and recrystallization from EtOAc and petroleum ether gave the compound of formula:
CN202311620492.XA 2023-11-30 2023-11-30 Protein degradation targeting chimeric and preparation method thereof Pending CN117603225A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311620492.XA CN117603225A (en) 2023-11-30 2023-11-30 Protein degradation targeting chimeric and preparation method thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311620492.XA CN117603225A (en) 2023-11-30 2023-11-30 Protein degradation targeting chimeric and preparation method thereof

Publications (1)

Publication Number Publication Date
CN117603225A true CN117603225A (en) 2024-02-27

Family

ID=89953102

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311620492.XA Pending CN117603225A (en) 2023-11-30 2023-11-30 Protein degradation targeting chimeric and preparation method thereof

Country Status (1)

Country Link
CN (1) CN117603225A (en)

Similar Documents

Publication Publication Date Title
US10981921B2 (en) Fragment synthesis of substituted cyclic peptides
KR20200013058A (en) SSAO inhibitor
JP4012145B2 (en) Solid phase synthesis of pyrrole-imidazole polyamide
CN108727468B (en) Cyclic peptide, pharmaceutical or cosmetic composition comprising the same, and preparation method thereof
CN107474107B (en) Process for the preparation of GLYX-13 and compounds useful for the preparation of GLYX-13
EP3735987A1 (en) Amanitin antibody conjugate
CN110092740B (en) Fused ring compound and application thereof
KR20170020808A (en) Method for producing synthetic pentapeptide
CN117603225A (en) Protein degradation targeting chimeric and preparation method thereof
CN113845485B (en) Amino acid derivative and preparation method and application thereof
CN108191849B (en) Preparation method of anti-epidermal growth factor receptor drug resistance mutation inhibitor, related intermediate and application
CN113402438A (en) Preparation method of tryptophan derivative medical intermediate
CN114315679A (en) Preparation method of Upactinib chiral intermediate
CN107739316B (en) Bromotyrosine alkaloid compound and preparation method and application thereof
CN111574463A (en) Riagliptin intermediate compound IV
CN111825659A (en) Impurity hydrochloride of dalatavir hydrochloride, preparation method and application thereof
CN111410650B (en) Process for the preparation of sulfonamides
CN111574520A (en) Riagliptin intermediate compound V
Ge et al. Iron-catalyzed C–O bond functionalization of butyrolactam derivatives with various N-/C-nucleophiles
CN114853729B (en) Quinazoline-2-ketone compound and preparation method and application thereof
CN111978213B (en) Multifunctional compound for polypeptide cyclization and preparation method and application thereof
CN118164889A (en) Synthesis method of chiral pyrrole glycine derivative
CN116606233A (en) Preparation method of S-trityl-L-cysteamine
Bergel et al. 734. Cyto-active amino-acids and peptides. Part IX. Further studies of N-acyl-dipeptides from melphalan
CN115322120A (en) Small molecule compound and its preparation method application of DHODH mediated disease medicine

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