CN113754680A - Alpha fluoroacyl piperazine derivative and preparation and application thereof - Google Patents

Alpha fluoroacyl piperazine derivative and preparation and application thereof Download PDF

Info

Publication number
CN113754680A
CN113754680A CN202111140554.8A CN202111140554A CN113754680A CN 113754680 A CN113754680 A CN 113754680A CN 202111140554 A CN202111140554 A CN 202111140554A CN 113754680 A CN113754680 A CN 113754680A
Authority
CN
China
Prior art keywords
cancer
derivative
reaction
compound
alpha
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202111140554.8A
Other languages
Chinese (zh)
Other versions
CN113754680B (en
Inventor
沈征武
邓斌
江亮
查雨峰
张梦麒
边泓竹
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.)
Yunbaiyao Zhengwu Technology Shanghai Co ltd
Original Assignee
Yunbaiyao Zhengwu Technology Shanghai Co ltd
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 Yunbaiyao Zhengwu Technology Shanghai Co ltd filed Critical Yunbaiyao Zhengwu Technology Shanghai Co ltd
Priority to CN202111140554.8A priority Critical patent/CN113754680B/en
Publication of CN113754680A publication Critical patent/CN113754680A/en
Priority to PCT/CN2022/105698 priority patent/WO2023050975A1/en
Application granted granted Critical
Publication of CN113754680B publication Critical patent/CN113754680B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • C07D495/02Heterocyclic 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 two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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

Abstract

The invention discloses an alpha fluoroacyl piperazine derivative, which has an alpha fluoroacyl piperazine derivative with a general formula (I) or an isomer thereof, or a pharmaceutically acceptable salt thereof, or a prodrug molecule thereof, wherein the chemical structure of the general formula (I) is as follows:

Description

Alpha fluoroacyl piperazine derivative and preparation and application thereof
Technical Field
The invention belongs to the field of medicinal chemistry, and particularly relates to an alpha fluoroacyl piperazine derivative, and preparation and application thereof.
Background
PI3K has a close role in intracellular signal transduction and plays a role in regulation of many important cellular processes, such as cell growth, differentiation, proliferation, apoptosis, intracellular transport, etc. The PI3K-Akt-mTOR signal path plays an important role in the aspects of cell growth, differentiation, apoptosis and the like, wherein a plurality of member molecules of signal transduction are key drug targets in the processes of cancer, immunity, thrombosis control and the like. When this signaling pathway is abnormally activated in the human body, cancer often occurs.
The PI3K kinase is divided into I, II and III, wherein the I type PI3K is the most studied, and the I type PI3K is a heterodimer and consists of a regulatory subunit and a catalytic subunit. There are 4 catalytic subunits, p110 α, β, δ, γ. Today, the target-pathological role of cancer and PI3K has been identified.
Each of the four catalytic subunits of class I PI3K kinases preferentially regulates specific signal transduction, depending on the type of malignancy and the genetic or epigenetic changes that occur. For example, p110 α is critical for the growth of PIK3CA mutations or tumor cells driven by the oncogene RAS and receptor tyrosine kinases; p110 β mediates PTEN-deficient tumorigenesis; and p110 delta is highly expressed in leukocytes, so that the p110 delta becomes an ideal target for treating hematological malignancies. p110 γ may play an important role in the pathogenesis of IPF (idiopathic pulmonary interstitial fibrosis) and may be a specific pharmacological target of IPF.
Due to the great potential of PI3K inhibitors in the treatment of tumors and a variety of diseases, numerous drug enterprises and research institutions devote significant resources to the development of such drugs. PI3K inhibitors currently approved by the FDA for marketing include: idelalisib, a PI3K δ selective inhibitor, for the treatment of lymphoma; copanlisib, PI 3K-alpha/delta inhibitors, for the treatment of recurrent follicular lymphoma; duvelisib, PI3K δ/γ inhibitor, for treating lymphoma; an inhibitor of Alpelisib, PI 3K-alpha, for the treatment of advanced metastatic breast cancer; inhibitors of ubbralisib, PI3K δ and CK1 ∈ for the treatment of lymphomas.
The PI3K inhibitor (Apitolisib, GDC-0980, RG7422, CAS:1032754-93-0) in the second clinical stage developed by gene Take has the following structural formula, and can be used for treating breast cancer, prostate cancer, endometrial cancer and renal tumor;
Figure BDA0003283738550000021
disclosure of Invention
The invention aims to provide an alpha fluoroacyl piperazine derivative, and preparation and application thereof, and aims to solve the technical problem that a medicament in the prior art is poor in effect on treating cancer.
The invention provides an alpha fluoroacylpiperazine derivative, which has an alpha fluoroacylpiperazine derivative with a general formula (I) or an isomer thereof, or a pharmaceutically acceptable salt thereof, or a prodrug molecule thereof, wherein the chemical structure of the general formula (I) is as follows:
Figure BDA0003283738550000031
in the general formula (I), X ═ O or S; wherein the acyl alpha position of the acyl piperazine contains at least one fluorine atom;
in the general formula (I), R1Is H, halogen, cyano, alkyl, alkenyl, alkynyl of 1-15 carbons or derivatives thereof; a monocyclic or fused ring aryl group having 5 to 22 carbon atoms or a derivative thereof; a 5-to 8-membered heterocyclic or heterocyclic ring containing 1 to 4 heteroatoms or a derivative thereof; a carboxyl group or a derivative thereof; a hydroxyl group or a derivative thereof; amino or a derivative thereof; a mercapto group or a derivative thereof; a sulfone or sulfoxide derivative; a sulfonate or sulfonate; a phosphate or a phosphate;
in the general formula (I), R2Is H, halogen, cyano, alkyl, alkenyl, alkynyl of 1-15 carbons or derivatives thereof; a monocyclic or fused ring aryl group having 5 to 22 carbon atoms or a derivative thereof; a 5-to 8-membered heterocyclic or heterocyclic ring containing 1 to 4 heteroatoms or a derivative thereof;a carboxyl group or a derivative thereof; a hydroxyl group or a derivative thereof; amino or a derivative thereof; a mercapto group or a derivative thereof; a sulfone or sulfoxide derivative; a sulfonate or sulfonate; a phosphate or a phosphate.
Further, in the general formula (I), the alpha position of acyl piperazine is a single optical configuration or is a racemate; r1And R2May be the same or different; in the general formula (I), R1And R2May be joined to form a ring structure.
Further, the preferred structure of the alpha fluoroacyl piperazine derivative is as follows:
Figure BDA0003283738550000041
the invention also provides a pharmaceutical composition, which contains the alpha fluoroacylpiperazine derivative or the isomer thereof, or the pharmaceutically acceptable salt thereof, or the prodrug molecule thereof with a therapeutically effective amount and one or more pharmaceutically acceptable carriers, diluents or excipients.
The invention also provides a preparation method of the alpha fluoroacyl piperazine derivative, and the reaction equation is as follows:
Figure BDA0003283738550000051
or as follows:
Figure BDA0003283738550000052
in the first reaction formula, P1Is H or an amino protecting group, P2Is H or an amino protecting group; p1、P2Not H at the same time;
the amino-protecting group includes formyl, acetyl, trifluoroacetyl, benzoyl, t-butoxycarbonyl, benzyloxycarbonyl, 9-fluorenylmethoxycarbonyl, phthaloyl, cyclosuccinyl, 2-biphenyl-2-propoxycarbonyl, p-toluenesulfonyl, trityl and the like;
the reductive amination reaction solvent can be anhydrous or water-containing, and comprises dichloromethane, tetrahydrofuran, methanol, ethanol, isopropanol, 1, 2-dichloroethane, ethylene glycol dimethyl ether, di (ethylene glycol) dimethyl ether and the like;
the reducing agent used in the reductive amination comprises NaBH4,KBH4,LiBH4,Zn(BH4)2,NaBH3CN,NaBH(OAc)3Borane complex, Bu3SnH,PhSiH4Etc.;
catalysts used for reductive amination include protic acids and Lewis acids, e.g. hydrochloric acid, sulfuric acid, phosphoric acid, formic acid, acetic acid, trifluoroacetic acid, BF3,SnCl2,SnCl4,Ti(OiPr)4,SiO2,BuSnCl2Etc.;
the acid used for deprotection includes protonic acids and Lewis acids, such as hydrochloric acid, sulfuric acid, phosphoric acid, formic acid, acetic acid, p-toluenesulfonic acid, trifluoromethanesulfonic acid, trifluoroacetic acid, BF3,SnCl2,SnCl4,Ti(OiPr)4,SiO2,BuSnCl2,AlCl3Etc.;
the base used for deprotection includes inorganic bases and organic bases, and the inorganic bases include: sodium hydroxide, potassium hydroxide, sodium hydride, calcium fluoride, cesium fluoride, sodium carbonate, potassium carbonate, cesium carbonate, potassium phosphate, and the like; the organic base includes: hydrazine hydrate, lithium diisopropylamide, butyllithium, lithium bis (trimethylsilyl) amide, triethylamine, diisopropylethylamine, pyridine, pyrrole, piperidine, morpholine, N-methylmorpholine, 1, 8-diazabicycloundec-7-ene, and the like;
hydrogen sources used for catalytic reduction deprotection reaction comprise hydrogen, formic acid, ammonium formate and the like; the catalyst comprises metal catalysts such as Pd, Pt, Ni, Cu and the like;
the solvent used for the deprotection reaction may be a protic solvent, an aprotic solvent or a mixed solvent. Preferably dichloromethane, dichloroethane, tetrahydrofuran, acetonitrile, methanol, ethanol, water, ethylene glycol dimethyl ether, N-dimethylformamide, dimethylsulfoxide, etc.;
the thionating agent is P2S52, 4-bis (methylthio) -1,3,2, 4-dithiadiphosphobutylene-2, 4-disulfide, 2, 4-bis (phenylthio) -1, 3-disulfide-2, 4-diphosphetane-2, 4-disulfide, or 2, 4-bis (4-phenoxyphenyl) -1,3,2, 4-dithiodiphosphetane-2, 4-disulfide; the solvent used in the reaction is any one or the mixture of two of a protic solvent and an aprotic solvent;
in the second reaction scheme, the fluorinating agent used includes HF and its salt, SF4Diethylaminosulfur trifluoride, bis (2-methoxyethyl) aminosulfur trifluoride, 4-tert-butyl-2, 6-dimethylphenylthio trifluoride, pyridine-2-sulfonylfluoride, bis (2-methoxyethyl) aminosulfur trifluoride, diethylamino) difluorosulfonium tetrafluoroborate, difluoro (4-morpholinyl) sulfonium tetrafluoroborate, 1, 3-bis (2, 6-diisopropylphenyl) -2, 2-difluoroimidazoline, 4-chloro-N- [ (4-methylphenyl) sulfonyl ] sulfonyl]-benzenesulfonamide fluoride; the solvent used for the reaction is an aprotic solvent or a mixed solvent. Preferred are methylene chloride, dichloroethane, tetrahydrofuran, acetonitrile, ethylene glycol dimethyl ether, 1, 4-dioxane, etc.
The reaction temperature is-78-180 ℃;
the crude product of the final product in the reaction can be further purified by solvent extraction, precipitation and crystallization, or column chromatography, the filler is silica gel, macroporous resin or alumina, and the eluent can be petroleum ether-acetone, petroleum ether-ethyl acetate, petroleum ether-dichloromethane and the like which are mixed in different proportions.
The invention also provides application of the alpha fluoroacyl piperazine derivative or isomer thereof, or pharmaceutically acceptable salt thereof, or prodrug molecule thereof in preparing a medicament for treating cancer.
Further, the cancer is brain cancer, brain glioma, endometrial cancer, ovarian cancer, cervical cancer, breast cancer, colon cancer, lung cancer, prostate cancer, liver cancer, leukemia, lymphoma, skin cancer, basal cell tumor, hemangioma, uterine cancer, laryngeal cancer, stomach cancer, lip cancer, esophageal cancer, nasopharyngeal cancer, gallbladder cancer, pancreatic cancer, kidney cancer, tongue cancer, bladder cancer, melanoma, lipoma, thyroid cancer, thymus cancer, or bone cancer.
The invention also provides application of the alpha fluoroacylpiperazine derivative or isomer thereof, or pharmaceutically acceptable salt thereof, or prodrug molecule thereof and at least one other anticancer agent in preparation of a medicament for treating cancer.
Further, the additional anticancer agent is any one or a combination of two or more of adriamycin, bleomycin, vinblastine, taxane, etoposide, 5-fluorouracil, cyclophosphamide, methotrexate, cisplatin, tretinoin, temozolomide, actinomycin, imatinib, gefitinib, sorafenib, erlotinib, sunitinib, afatinib, cabozantinib, ostoxib, rituximab, cetuximab, trastuzumab, nivolumab, palivizumab, attentizumab, daclizumab, or avizumab.
The alpha-fluoro-amide piperazine derivative introduces fluorine atoms with strong electron withdrawing ability at the ortho position of the amide group, changes the electron cloud distribution of the amide piperazine, not only enhances the PI3K kinase inhibition activity of the molecules, but also optimizes the lipid-water distribution coefficient of the compound, so that the medicine can permeate cell membranes more easily, and has extremely strong anti-tumor activity and very wide application prospect.
Detailed Description
The technical solutions in the embodiments of the present invention are clearly and completely described below, and it is obvious that the described embodiments are only a part of the embodiments of the present invention, and not all embodiments. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
The first embodiment is as follows: preparation of compound 1:
Figure BDA0003283738550000081
(1) preparation of Compound 1aPreparing: under the protection of nitrogen, 2-chloro-7-methyl-4-morpholin-4-yl thieno [3,2-d]Pyrimidine-6-carbaldehyde (CAS:955979-02-9,190 g, 0.640mol), 2- (N, N-di-tert. -butoxycarbonyl) aminopyrimidine-5-boronic acid pinacol ester (350 g, 0.831mol), Pd (dppf) Cl2(4.7 g, 0.0064mol), K2CO3(180 g, 1.28mol) were added to 1, 4-dioxane (2l) and H2O (0.2L) was added to the mixed solution, and the mixture was heated to 80 ℃ and stirred for 2 hours. After completion of the reaction, the reaction mixture was cooled to room temperature by TLC, and extracted with saturated ammonium chloride (2L) and ethyl acetate (2L). The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the crude product was subjected to separation and purification by silica gel column chromatography (eluent: petroleum ether, ethyl acetate/petroleum ether (1/1), ethyl acetate) to obtain 160g of a yellow solid in a yield of 45%.
MS:[M+1]+=557.2
1H NMR(400MHz,DMSO-d6)δ:10.4(s,1H),9.67(s,2H),3.98-4.03(m,4H),3.77-3.81(m,4H),2.73(s,3H),1.43(s,18H).
(2) Preparation of compound 1 b: compound 1a (500 mg, 0.9mmol) was dissolved in dichloromethane (5ml) followed by the addition of 1- (α -fluoropropionyl) piperazine (1.44 g, 9mmol) and acetic acid (54 mg). The reaction was stirred at room temperature for half an hour and then sodium cyanoborohydride (226 mg, 3.6mmol) was added. The reaction solution was further stirred at room temperature for 12 hours. 5ml of saturated aqueous sodium bicarbonate solution were added and the aqueous phase was extracted twice with 5ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 2/1) gave 300mg of the product as a white solid in 47% yield.
MS:[M+1]+=701.3
1H NMR(400MHz,DMSO-d6)δ:9.73(s,2H),5.21and 5.33(dq,J=48.2,6.6Hz,1H),4.09-4.01(m,4H),3.94-3.87(m,4H),3.84(s,2H),3.60-3.80(m,4H),2.50-2.65(m,4H),2.44(s,3H),1.55and 1.62(dd,J=27.0,6.6Hz,3H),1.47(s,18H).
(3) Preparation of compound 1: compound 1b (300mg,0.43mmol) was dissolved in methanol (3mL) and then hydrochloric acid (37%, 1mL) was added at 0 ℃. The reaction was stirred at room temperature overnight. When the thin layer silica gel chromatography indicated the reaction was complete, cold saturated aqueous sodium bicarbonate (20ml) was added dropwise to the reaction and extracted 3 times with 10ml ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography and lyophilized to give 60 mg of a white solid product in 30% yield.
MS:[M+1]+=501.2
1H NMR(400MHz,CDCl3)δ:9.33(s,2H),5.51(s,2H),5.30and5.20(dq,J=6.6,48.4Hz,1H),4.00-4.04(m,4H),3.80-3.95(m,6H),3.60-3.80(m,4H),2.55-2.65(m,4H),2.42(s,3H),1.60and 1.54(dd,J=6.6,24.6Hz,3H).
Example two: preparation of compound 2:
Figure BDA0003283738550000101
starting material 13(50mg,0.1mmol) was dissolved in dichloromethane (1mL) under argon followed by 4 drops of DAST dropwise. After the reaction solution was stirred at room temperature for 5 hours, the reaction solution was poured into 10ml of a saturated aqueous sodium bicarbonate solution and extracted twice with 20ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by preparative liquid chromatography gave 31 mg of a white solid.
MS:[M+1]+=501.2
1H NMR(400MHz,DMSO-d6)δ:9.16(s,2H),7.10(s,2H),5.57 and5.55(dq,J=6.6,48.4Hz,1H),3.90-4.00(m,4H),3.86(s,2H),3.70-3.80(m,4H),3.30-3.60(m,8H),2,35(s,3H),1.42and 1.36(dd,J=6.6,24.8Hz,3H).
Example three: preparation of Compound 3
Figure BDA0003283738550000111
Under argon protection, compound 1(200mg,0.4mmol) and lawson's reagent (170mg,0.42mmol) were dissolved in anhydrous tetrahydrofuran (12mL) and refluxed overnight. The reaction was concentrated under reduced pressure and the residue was purified by preparative liquid chromatography and lyophilized to give 80 mg of a white product in 39% yield.
MS:[M+1]+=517.2
1H NMR(400MHz,CDCl3)δ:9.34(s,2H),5.80and 5.67(dq,J=6.8,49.9Hz,1H),5.49(s,2H),4.25-4.50(m,2H),3.95-4.15(m,5H),3.75-3.95(m,7H),2.60-2.80(m,4H),2.42(s,3H),1.72and 1.66(dd,J=6.8,24.7Hz,3H).
Example four: preparation of Compound 4
Figure BDA0003283738550000112
(1) Preparation of compound 4 a: compound 1a (150mg,0.27mmol) was dissolved in dichloromethane (3ml), followed by addition of 1- (2-fluoro-2-methylpropionyl) piperazine (469.48mg,2.69mmol) and acetic acid (16 mg). The reaction was stirred at room temperature for half an hour and then sodium cyanoborohydride (68mg,1.08mmol) was added. The reaction solution was further stirred at room temperature for 12 hours. 10ml of saturated aqueous sodium bicarbonate solution were added and the aqueous phase was extracted twice with 10ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 2/1) gave 100mg of the product as a white solid in 52% yield.
MS:[M+1]+=715.3
1H NMR(400MHz,CDCl3)δ:9.72(s,2H),3.90–4.00(m,4H),3.75–3.90(m,8H),3.70(s,2H),2.55-2.70(m,4H),2.43(s,3H),1.59and 1.64(d,J=21.8Hz,6H),1.46(s,18H).
(2) Preparation of compound 4: compound 4a (100mg,0.14mmol) was dissolved in methanol (3mL) and then hydrochloric acid (37%, 1mL) was added at 0 ℃. The reaction was stirred at room temperature overnight. When the thin layer silica gel chromatography indicated the reaction was complete, cold saturated aqueous sodium bicarbonate (20ml) was added dropwise to the reaction and extracted 3 times with 10ml ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography and lyophilized to give 53 mg of a white solid product in 74% yield.
MS:[M+1]+=515.2
1H NMR(400MHz,CDCl3)δ:9.33(s,2H),5.49(brs,2H),4.03(m,4H),3.88(m,4H),3.82(s,2H),3.60-3.75(m,4H),2.55-2.65(m,4H),2.42(s,3H),1.64and 1.59(d,J=21.8Hz,6H).
Example five: preparation of Compound 5
Figure BDA0003283738550000131
(1) Preparation of compound 5 a: compound 1a (150mg,0.27mmol) was dissolved in dichloromethane (3ml), followed by addition of 1- (2, 2-difluoropropionyl) piperazine (479mg,2.69mmol) and acetic acid (16 mg). The reaction was stirred at room temperature for half an hour and then sodium cyanoborohydride (68mg,1.08mmol) was added. The reaction solution was further stirred at room temperature for 12 hours. 10ml of saturated aqueous sodium bicarbonate solution were added and the aqueous phase was extracted twice with 10ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 2/1) gave 105mg of the product as a white solid in 54% yield.
MS:[M+1]+=719.3
1H NMR(400MHz,CDCl3)δ:9.73(s,2H),4.00-4.10(m,4H),3.75-3.95(m,8H),3.72(s,2H),2.55-2.70(m,4H),2.44(s,3H),1.84(t,J=19.9Hz,3H),1.47(s,18H).
(2) Preparation of compound 5: compound 5a (105mg,0.15mmol) was dissolved in methanol (3mL) and then hydrochloric acid (37%, 1mL) was added at 0 ℃. The reaction was stirred at room temperature overnight. When the thin layer silica gel chromatography indicated the reaction was complete, cold saturated aqueous sodium bicarbonate (20ml) was added dropwise to the reaction and extracted 3 times with 10ml ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography and lyophilized to give 57 mg of a white solid product in 75% yield.
MS:[M+1]+=519.2
1H NMR(400MHz,CDCl3)δ:9.33(s,2H),5.29(brs,2H),3.90-4.10(m,4H),3.70-3.90(m,4H),3.82(s,2H),3.60-3.80(m,4H),2.58-2.64(m,4H),2.42(s,3H),1.84(t,J=19.9Hz,3H).
Example six: preparation of Compound 6
Figure BDA0003283738550000141
(1) Preparation of compound 6 a: compound 1a (150mg,0.27mmol) was dissolved in dichloromethane (3ml) followed by the addition of 1- (2,2, 2-trifluoroacetyl) piperazine (489mg,2.69mmol) and acetic acid (16 mg). The reaction was stirred at room temperature for half an hour and then sodium cyanoborohydride (68mg,1.08mmol) was added. The reaction solution was further stirred at room temperature for 12 hours. 10ml of saturated aqueous sodium bicarbonate solution were added and the aqueous phase was extracted twice with 10ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 3/1) gave 100mg of the product as a white solid in 51% yield.
MS:[M+1]+=723.3
1H NMR(400MHz,CDCl3)δ:9.73(s,2H),4.00-4.10(m,4H),3.80-3.95(m,6H),3.76(s,2H),3.60-3.70(m,2H),2.60-2.70(m,4H),2.44(s,3H),1.48(s,18H).
(2) Preparation of compound 6: compound 6a (100mg,0.14mmol) was dissolved in methanol (3mL) and then hydrochloric acid (37%, 1mL) was added at 0 ℃. The reaction was stirred at room temperature overnight. When the thin layer silica gel chromatography indicated the reaction was complete, cold saturated aqueous sodium bicarbonate (20ml) was added dropwise to the reaction and extracted 3 times with 10ml ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography and lyophilized to give 65 mg of a white solid product in 86% yield.
MS:[M+1]+=523.2
1H NMR(400MHz,CDCl3)δ:9.32(s,2H),5.41(brs,2H),3.90-4.10(m,4H),3.75-3.90(m,4H),3.84(s,2H),3.60-3.80(m,4H),2.60-2.70(m,4H),2.42(s,3H).
Example seven: preparation of Compound 7
Figure BDA0003283738550000151
(1) Preparation of compound 7 a: 1-Boc-piperazine (9.01g,48.35mmol) and triethylamine (9.79g,96.71mmol) were dissolved in dichloromethane (30mL) and cooled to 0 deg.C, then a solution of 2, 2-difluorobutyryl chloride (3.4 g, 23.8mmol) in dichloromethane (20mL) was added dropwise. The reaction was warmed to room temperature and stirring was continued for 1.5 hours. 30mL of water was added dropwise to the reaction mixture, the organic phase was separated, and the aqueous phase was extracted twice with 20mL of dichloromethane. The organic phases were combined and washed with anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: ethyl acetate/petroleum ether: 1/10) gave 4.9 g of the product as a white solid in 69% yield.
1H NMR(400MHz,CDCl3)δ:3.67-3.72(m,2H),3.58-3.63(m,2H),3.43-3.50(m,4H),2.08-2.24(m,2H),1.47(s,9H),1.07(t,J=7.44Hz,3H).
(2) Preparation of compound 7 b: compound 7a (3g,10.3mmol) was dissolved in dichloromethane (10 mL). Trifluoroacetic acid (10mL) was added dropwise at 0 ℃ and the reaction was stirred at room temperature for 1.5 hours after completion of the addition. When the thin layer silica gel chromatography showed the reaction was complete, 10mL of saturated cold NaHCO was added to the reaction3The aqueous phase was extracted 3 times with 10mL of dichloromethane. The combined organic phases were washed once with 20mL of saturated brine and then over anhydrous Na2SO4After drying, the mixture was concentrated under reduced pressure and dried in vacuo, and the resulting yellow oil was used in the next reaction without further purification. .
MS:[M+1]+=193.1
(3) Preparation of compound 7 c: compound 1a (150mg,0.27mmol) was dissolved in dichloromethane (3mL)Then 7b (516mg,2.69mmol) and acetic acid (16mg) were added. The reaction was stirred at room temperature for half an hour and then sodium cyanoborohydride (68mg,1.08mmol) was added. The reaction solution was further stirred at room temperature for 12 hours. 10ml of saturated aqueous sodium bicarbonate solution were added and the aqueous phase was extracted twice with 10ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 3/1) gave 100mg of the product as a white solid in 50% yield.
MS:[M+1]+=733.3
1H NMR(400MHz,CDCl3)δ:9.72(s,2H),4.07-4.03(m,4H),3.90-3.86(m,4H),3.83(s,2H),3.77(m,2H),3.70(m,2H),2.60(m,4H),2.43(s,3H),2.05-2.30(m,2H),1.46(s,18H),1.07(t,J=7.4Hz,3H).
(4) Preparation of compound 7: compound 7c (100mg,0.14mmol) was dissolved in methanol (3mL) and then hydrochloric acid (37%, 1mL) was added at 0 ℃. The reaction was stirred at room temperature overnight. When the thin layer silica gel chromatography indicated the reaction was complete, cold saturated aqueous sodium bicarbonate (20ml) was added dropwise to the reaction and extracted 3 times with 10ml ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography and lyophilized to give 54 mg of a white solid product in 74% yield.
MS:[M+1]+=533.2
1H NMR(400MHz,CDCl3)δ:9.32(s,2H),5.27(brs,2H),3.95-4.05(m,4H),3.83-3.90(m,4H),3.82(s,2H),3.65-3.81(m,4H),2.58-2.61(m,4H),2.41(s,3H),2.05-2.35(m,2H),1.07(t,J=7.4Hz,3H).
Example eight: preparation of Compound 8
Figure BDA0003283738550000181
(1) Preparation of compound 8 a: 1-Boc-piperazine (10.74g,57.65mmol) and triethylamine (11.67g,115.30mmol) were dissolved in dichloromethane (30mL) and cooled to 0 deg.C, then 1-fluorocyclopropanoyl chloride (C.) (30mL) was added dropwise3.5 g, 28.8mmol) in dichloromethane (20 mL). The reaction was warmed to room temperature and stirring was continued for 1.5 hours. 30mL of water was added dropwise to the reaction mixture, the organic phase was separated, and the aqueous phase was extracted twice with 20mL of dichloromethane. The organic phases were combined and washed with anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: ethyl acetate/petroleum ether: 1/6) gave 4.2 g of the product as a white solid in 54% yield.
1H NMR(400MHz,CDCl3)δ:3.50-3.75(m,4H),3.30-3.50(m,4H),1.40(s,9H),1.10–1.30(m,4H).
(2) Preparation of compound 8 b: compound 8a (3g,11.2mmol) was dissolved in dichloromethane (10 mL). Trifluoroacetic acid (10mL) was added dropwise at 0 ℃ and the reaction was stirred at room temperature for 1.5 hours after completion of the addition. When the thin layer silica gel chromatography showed the reaction was complete, 20mL of saturated cold NaHCO was added to the reaction3The aqueous phase was extracted 3 times with 10mL of dichloromethane. The combined organic phases were washed once with 20mL of saturated brine and then over anhydrous Na2SO4After drying, the mixture was concentrated under reduced pressure and dried in vacuo, and the resulting yellow oil was used in the next reaction without further purification.
MS:[M+1]+=173.1
(3) Preparation of compound 8 c: compound 1a (150mg,0.27mmol) was dissolved in dichloromethane (3mL) followed by the addition of 8b (462mg,2.69mmol) and acetic acid (16 mg). The reaction was stirred at room temperature for half an hour and then sodium cyanoborohydride (68mg,1.08mmol) was added. The reaction solution was further stirred at room temperature for 12 hours. 10ml of saturated aqueous sodium bicarbonate solution were added and the aqueous phase was extracted twice with 10ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 2/1) gave 98mg of the product as a white solid in 51% yield.
MS:[M+1]+=713.3
1H NMR(400MHz,CDCl3)δ:9.73(s,2H),4.00-4.10(m,4H),3.60-3.90(m,10H),2.58-2.66(m,4H),2.45(s,3H),1.47(s,18H),1.15-1.34(m,4H).
(4) Preparation of compound 8: compound 8c (98mg,0.14mmol) was dissolved in methanol (3mL) and then hydrochloric acid (37%, 1mL) was added at 0 ℃. The reaction was stirred at room temperature overnight. When the thin layer silica gel chromatography indicated the reaction was complete, cold saturated aqueous sodium bicarbonate (20ml) was added dropwise to the reaction and extracted 3 times with 10ml ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography and lyophilized to give 53 mg of a white solid product in 75% yield.
MS:[M+1]+=513.2
1H NMR(400MHz,CDCl3)δ:9.33(s,2H),5.53(brs,2H),4.00-4.10(m,4H),3.85-3.90(m,4H),3.84(s,2H),3.65-3.80(m,4H),2.60-2.65(m,4H),2.42(s,3H),1.15-1.40(m,4H).
Example nine: preparation of Compound 9
Figure BDA0003283738550000201
(1) Preparation of compound 9 a: 1-Fluorocyclohexylcarboxylic acid (1g,6.84mmol), 1-Boc-piperazine (2.55g,13.68mmol), EDCI hydrochloride (1.57g,8.21mmol), DMAP (1.67g,13.68mmol) were dissolved in dichloromethane (50mL), and the reaction mixture was stirred at room temperature for 10 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether: 1/10) to give 1.3g of a white solid product in a yield of 60%.
1H NMR(400MHz,CDCl3)δ:3.7-3.80(m,2H),3.50-3.70(m,2H),3.4-0-3.49(m,4H),1.70-2.00(m,4H),1.55-1.70(m,5H),1.47(s,9H),1.20-1.40(m,1H).
(2) Preparation of compound 9 b: compound 9a (1.3g,4.3mmol) was dissolved in dichloromethane (6 mL). Trifluoroacetic acid (6mL) was added dropwise at 0 ℃ and the reaction was stirred at room temperature for 1.5 hours after completion of the addition. When the thin layer silica gel chromatography showed the reaction was complete, 10mL of saturated cold NaHCO was added to the reaction3The aqueous phase was extracted 3 times with 10mL of dichloromethane. The combined organic phases were washed once with 20mL of saturated brine and then over anhydrous Na2SO4After drying, the mixture was concentrated under reduced pressure and dried in vacuo, and the resulting yellow oil was used in the next reaction without further purification.
MS:[M+1]+=215.2
(3) Preparation of compound 9 c: compound 1a (150mg,0.27mmol) was dissolved in dichloromethane (3mL) followed by the addition of 9b (577mg,2.69mmol) and acetic acid (16 mg). The reaction was stirred at room temperature for half an hour and then sodium cyanoborohydride (68mg,1.08mmol) was added. The reaction solution was further stirred at room temperature for 12 hours. 10ml of saturated aqueous sodium bicarbonate solution were added and the aqueous phase was extracted twice with 10ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 2/1) gave 110mg of the product as a white solid in 54% yield.
MS:[M+1]+=755.4
1H NMR(400MHz,CDCl3)δ:9.73(s,2H),4.00-4.10(m,4H),3.87-3.91(m,4H),3.83(s,2H),3.55-3.80(m,4H),2.50-2.63(m,4H),2.44(s,3H),1.80-2.01(m,4H),1.55-1.80(m,5H),1.47(s,18H),1.20-1.40(m,1H).
(4) Preparation of compound 9: compound 9c (110mg,0.15mmol) was dissolved in methanol (3mL) and then hydrochloric acid (37%, 1mL) was added at 0 ℃. The reaction was stirred at room temperature overnight. When the thin layer silica gel chromatography indicated the reaction was complete, cold saturated aqueous sodium bicarbonate (20ml) was added dropwise to the reaction and extracted 3 times with 10ml ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography and lyophilized to give 43 mg of a white solid product in 39% yield.
MS:[M+1]+=555.2
1H NMR(400MHz,CDCl3)δ:9.33(s,2H),5.38(brs,2H),4.00-4.08(m,4H),3.82-3.90(m,6H),3.82(s,2H),3.60-3.75(m,2H),2.55-2.65(m,4H),2.42(s,3H),1.80-2.00(m,4H),1.50-1.75(m,5H),1.20-1.35(m,1H).
Example ten: preparation of Compound 10
Figure BDA0003283738550000221
(1) Preparation of compound 10 a: 4-Fluoropiperidine-4-carboxylic acid hydrochloride (2.00g,10.89mmol) was dissolved in tetrahydrofuran (27mL) and water (27mL) followed by addition of CbzCl (2.23g,13.07mmol) and K2CO3(3.76g,27.23), and the reaction mixture was stirred at room temperature for 3 hours. When the reaction is complete by thin-layer silica gel chromatography, the pH is adjusted to 5 with 1M dilute hydrochloric acid and then extracted three times with 20ml of ethyl acetate. The organic phases were combined, dried over anhydrous sodium sulfate, concentrated under reduced pressure, dried in vacuo and the colorless oil was used in the next reaction without purification.
MS:[M-1]-=280.1
(2) Preparation of compound 10 b: 10a (3g,10.89mmol), 1-Boc-piperazine (4.07g,21.83mmol), EDCI hydrochloride (2.51g,13.10mmol), DMAP (2.67g,21.83mmol) were dissolved in dichloromethane (50mL), and the reaction mixture was stirred at room temperature for 10 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether: 1/10) to give 2.7g of a white solid product in 55% yield.
1H NMR(400MHz,CDCl3)δ:7.28-7.45(m,5H),5.17(s,2H),3.95-4.20(m,2H),3.55-3.60(m,4H),3.40-3.50(m,4H),3.15-3.30(m,2H),1.80-2.30(m,4H),1.47(s,9H).
(3) Preparation of compound 10 c: compound 10b (2.7g,6.01mmol) was dissolved in dichloromethane (10 mL). Trifluoroacetic acid (10mL) was added dropwise at 0 ℃ and the reaction was stirred at room temperature for 1.5 hours after completion of the addition. When the reaction was complete by TLC, 20mL of saturated NaHCO was added to the reaction3The aqueous phase was extracted 3 times with 20mL of dichloromethane. The combined organic phases were washed once with 30mL of saturated brine and then over anhydrous Na2SO4After drying, the mixture was concentrated under reduced pressure and dried in vacuo, and the resulting yellow oil was used in the next reaction without further purification.
MS:[M+1]+=350.2
(4) Preparation of compound 10 d: compound 1a (270.00mg,0.49 m)mol) was dissolved in dichloromethane (5ml) followed by the addition of 10c (1.6g,4.85mmol) and acetic acid (29 mg). The reaction mixture was stirred at room temperature for half an hour, and then sodium cyanoborohydride (122mg,1.96mmol) was added. The reaction solution was further stirred at room temperature for 12 hours. 20ml of saturated aqueous sodium bicarbonate solution were added and the aqueous phase was extracted twice with 20ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 2/1) gave 200mg of the product as a white solid in 46% yield.
MS:[M+1]+=890.4
1H NMR(400MHz,CDCl3)δ:9.73(s,2H),7.28-7.45(m,5H),5.14(s,2H),4.00-4.20(m,6H),3.60-3.90(m,10H),3.10-3.25(m,2H),2.55-2.65(m,4H),2.44(s,3H),2.05-2.40(m,4H),1.47(s,18H).
(5) Preparation of compound 10 e: compound 10d (200mg,0.22mmol) was dissolved in methanol (3mL) and then hydrochloric acid (37%, 1mL) was added at 0 ℃. The reaction was stirred at room temperature overnight. When the thin layer silica gel chromatography indicated the reaction was complete, cold saturated aqueous sodium bicarbonate (20ml) was added dropwise to the reaction and extracted 3 times with 10ml ethyl acetate. The organic phases were combined, washed once with 20ml of saturated brine, dried over anhydrous sodium sulfate, concentrated under reduced pressure and dried in vacuo to give 170mg of a white crude product which was used in the next reaction without purification.
MS:[M+1]+=690.3
(6) Preparation of compound 10: compound 10e (170mg,0.22mmol) was dissolved in methanol (5mL), and 15% Pd/C (34mg) was added, followed by hydrogenation at room temperature for 12 hours. The reaction was filtered through celite, the filtrate was concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography and lyophilized to give 20 mg of a white solid product in 16% yield.
MS:[M+1]+=556.2
1H NMR(400MHz,CDCl3)δ:9.32(s,2H),5.33(brs,2H),3.95-4.15(m,4H),3.75-3.90(m,8H),3.60-3.75(m,2H),2.90-3.10(m,3H),2.50-2.65(m,4H),2.41(s,3H),1.80-2.00(m,4H),1.70-2.30(m,6H).
Example eleven: preparation of Compound 11
Figure BDA0003283738550000251
(1) Preparation of compound 11 a: 2-fluoro-2-phenylacetic acid (1g,6.49mmol) was dissolved in dichloromethane (10mL) and cooled to 0 deg.C, then oxalyl chloride (894.58mg,7.79mmol) was added dropwise. After completion of the oxalyl chloride addition, 1 drop of DMF was added. After the reaction solution was warmed up to room temperature and stirred for 1.5 hours, a 2-fluoro-2-phenylacetyl chloride solution was obtained and used for the next reaction without any treatment.
1-Boc-piperazine (2.42g,12.98mmol) and triethylamine (2.63g,25.95mmol) were dissolved in dichloromethane (10mL) and cooled to 0 deg.C, and then a solution of the above 2-fluoro-2-phenylacetyl chloride in dichloromethane (10mL) was added dropwise to the reaction solution. After the completion of the dropwise addition, the reaction solution was further stirred at room temperature for 1.5 hours. 30ml of water are added and the aqueous phase is extracted three more times with 10ml of dichloromethane. The organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether: 1/7) to give 1.0g of a white solid product in 48% yield.
MS:[M+1]+=323.2
1H NMR(400MHz,CDCl3)δ:7.37-7.47(m,5H),6.09(d,J=49.4Hz,1H),3.00-3.80(m,8H),1.43(s,9H).
(2) Preparation of compound 11 b: compound 11a (1g,3.1mmol) was dissolved in dichloromethane (6 mL). Trifluoroacetic acid (6mL) was added dropwise at 0 ℃ and the reaction was stirred at room temperature for 1.5 hours after completion of the addition. When the thin layer silica gel chromatography showed the reaction was complete, 10mL of saturated cold NaHCO was added to the reaction3The aqueous phase was extracted 3 times with 10mL of dichloromethane. The combined organic phases were washed once with 20mL of saturated brine and then over anhydrous Na2SO4After drying, the mixture was concentrated under reduced pressure and dried in vacuo, and the resulting yellow oil was used in the next reaction without further purification.
MS:[M+1]+=223.1
(3) Preparation of compound 11 c: compound 1a (150mg,0.27mmol) was dissolved in dichloromethane (3mL) followed by the addition of 11b (599mg,2.69mmol) and acetic acid (16 mg). The reaction was stirred at room temperature for half an hour and then sodium cyanoborohydride (68mg,1.08mmol) was added. The reaction solution was further stirred at room temperature for 12 hours. 10ml of saturated aqueous sodium bicarbonate solution were added and the aqueous phase was extracted twice with 10ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 2/1) gave 118mg of the product as a white solid in 57% yield.
MS:[M+1]+=763.3
1H NMR(400MHz,CDCl3)δ:9.72(s,2H),7.34-7.49(m,5H),6.08(d,J=49.4Hz,1H),4.00-4.09(m,4H),3.84-3.93(m,4H),3.76(s,2H),3.70-3.76(m,2H),3.40-3.50(m,2H),2.50-2.60(m,2H),2.39(s,3H),2.25-2.35(m,2H),1.47(s,18H).
(4) Preparation of compound 11: compound 11c (118mg,0.15mmol) was dissolved in methanol (3mL) and then hydrochloric acid (37%, 1mL) was added at 0 ℃. The reaction was stirred at room temperature overnight. When the thin layer silica gel chromatography indicated the reaction was complete, cold saturated aqueous sodium bicarbonate (20ml) was added dropwise to the reaction and extracted 3 times with 10ml ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography and lyophilized to give 50mg of a white solid product in 57% yield.
MS:[M+1]+=563.2
1H NMR(400MHz,CDCl3)δ:9.31(s,2H),7.30-7.50(m,5H),6.07(d,J=49.4Hz,1H),5.31(brs,2H),3.95-4.05(m,4H),3.80-3.90(m,4H),3.60-3.80(m,4H),3.35-3.50(m,2H),2.50-2.69(m,2H),2.37(s,3H),2.20-2.37(m,2H).
Example twelve: preparation of Compound 12
Figure BDA0003283738550000271
(1) Preparation of compound 12 a: 2, 2-difluoro-2- (2-pyridyl) acetic acid (1g,5.78mmol), 1-Boc-piperazine (2.15g,11.55mmol), EDCI hydrochloride (1.33g,6.93mmol), DMAP (1.41g,11.55mmol) were dissolved in dichloromethane (30mL), and the reaction mixture was stirred at room temperature for 10 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether: 1/10) to give 1.0g of a white solid product in 51% yield.
1H NMR(400MHz,CDCl3)δ:8.60(d,J=4.4Hz,1H),7.88(m,1H),7.74(d,J=7.9Hz,1H),7.44(m,1H),3.65-3.72(m,2H),3.55-3.62(m,2H),3.45-3.51(m,2H),3.32-3.38(m,2H),1.46(s,9H).
(2) Preparation of compound 12 b: compound 12a (1.0g,2.93mmol) was dissolved in dichloromethane (5 mL). Trifluoroacetic acid (5mL) was added dropwise at 0 ℃ and the reaction was stirred at room temperature for 1.5 hours after completion of the addition. When the thin layer silica gel chromatography showed the reaction was complete, 20mL of saturated cold NaHCO was added to the reaction3The aqueous solution was extracted 3 times with 20mL of dichloromethane. The combined organic phases were washed once with 30mL of saturated brine and then over anhydrous Na2SO4After drying, the mixture was concentrated under reduced pressure and dried in vacuo, and the resulting yellow oil was used in the next reaction without further purification.
MS:[M+1]+=242.1
(3) Preparation of compound 12 c: compound 1a (150mg,0.27mmol) was dissolved in dichloromethane (3mL) followed by the addition of 12b (325mg,1.35mmol) and acetic acid (16 mg). The reaction was stirred at room temperature for half an hour and then sodium cyanoborohydride (68mg,1.08mmol) was added. The reaction solution was further stirred at room temperature for 12 hours. 10ml of saturated aqueous sodium bicarbonate solution were added and the aqueous phase was extracted twice with 10ml of ethyl acetate. The organic phases were combined and passed over anhydrous Na2SO4After drying, concentration under reduced pressure and purification of the residue by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate 2/1) gave 50mg of the product as a white solid in 26% yield.
MS:[M+1]+=782.3
1H NMR(400MHz,CDCl3)δ:9.72(s,2H),8.66(d,J=4.7Hz,1H),7.87(m,1H),7.72(d,J=7.9Hz,1H),7.43(m,1H),4.02-4.08(m,4H),3.86-3.91(m,4H),3.81(s,2H),3.70-3.80(m,2H),3.60-3.66(m,2H),2.61(m,2H),2.48(m,2H),2.42(s,3H),1.47(s,18H).
(4) Preparation of compound 12: compound 12c (50mg,0.06mmol) was dissolved in methanol (3mL) and then hydrochloric acid (37%, 1mL) was added at 0 ℃. The reaction was stirred at room temperature overnight. When the thin layer silica gel chromatography indicated the reaction was complete, cold saturated aqueous sodium bicarbonate (20ml) was added dropwise to the reaction and extracted 3 times with 10ml ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography and lyophilized to give 20 mg of a white solid product in 54% yield.
MS:[M+1]+=582.2
1H NMR(400MHz,CDCl3)δ:9.32(s,2H),8.66(d,J=4.4Hz,1H),7.80-7.90(m,1H),7.71(d,J=7.9Hz,1H),7.43(m,1H),5.36(brs,2H),3.95-4.10(m,4H),3.80-3.90(m,4H),3.70-3.80(m,4H),3.55-3.65(m,2H),2.55-2.60(m,2H),2.40-2.50(m,2H),2.40(s,3H).
Example thirteen: calculation of Compound clogP
clogP value refers to the logarithm of the partition coefficient ratio of a substance in n-octanol (oil) and water. Reflecting the distribution of the substances in the oil-water two phases. The larger the clogP value, the more lipophilic the substance, whereas the smaller the clogP value, the more hydrophilic, i.e. the better the water solubility. The dissolution, absorption, distribution and transport of a drug in vivo are related to the water solubility and lipid solubility of the drug, i.e. the oil-water partition coefficient clogP. Using the online tool provided by VCCLAB (http:// www.vcclab.org /), the results as shown in Table 1 were calculated:
TABLE 1 clogP of Compounds 1-12
Figure BDA0003283738550000301
The data obtained by calculation results show that clogP corresponding to the compounds 1-12 is between 1 and 5, and the ester water distribution coefficient is good.
Example fourteen: compound PI3K kinase Activity assay
Purpose of the experiment: testing compound pair PI by ADP-Glo luminescence method3Inhibitory Activity of Kalpha/beta/delta/gamma kinase
Experimental procedure the highest concentration of compound was set at 10uM, diluted 4-fold down with DMSO for a total of 10 concentrations: 10uM,2.5uM,0.625uM,0.156uM,0.039uM,0.0098uM,0.0024uM,0.0006uM,0.00015uM,0.000038 uM. Adding a corresponding PI3K alpha/beta/delta/gamma kinase solution into each hole of the test plate containing the compound DMSO solution, and uniformly mixing; PIP2 solution and ATP solution were added to each well to start the kinase reaction and incubated for 1h at room temperature. ADP Glo reagent equilibrated to room temperature was added to the wells to stop the kinase reaction, mixed by centrifugation and then gently shaken on a shaker to equilibrate for 120 minutes. Kinase test reagents were added to each well, mixed well and equilibrated for 30 minutes and readings recorded. Data curves were plotted using XLFit and IC was calculated50The value is obtained. The results are shown in Table 2.
The results of the experiment are shown in the following table:
TABLE 2 PI3K four subtype ICs for Compounds 1-12 and other Compounds50Data of
Figure BDA0003283738550000311
The results show that for the tested PI3K alpha/beta/delta/gamma kinase inhibitory activity, compounds 1-12 showed good activity on all four subtypes of PI3K and were stronger than apigenin. Particularly, compound 12 is the strongest PI3K α inhibitor found at present, and compared with apigenin, the compound 12 has 31 times of inhibitory activity on PI3K α and 29 times of inhibitory activity on PI3K β; the inhibitory activity to PI3K delta was 8 times that of Apixaglisa,
example fifteen: proliferation inhibition of tumor cells by compounds:
purpose of the experiment: MTT method was used to verify the proliferation inhibitory toxicity of the compound on MCF-7 cells (human breast cancer cells), NCI-H460 cells (human large cell lung cancer cells), HCT116 cells (human colon cancer cells), PC-3 cells (human prostate cancer cells), and HeLa cells (human cervical cancer cells).
The experimental method comprises the following steps: the sample was initially diluted 5 μ M, down 4-fold in order, to give 10 concentrations: 5000nM, 1250nM,312nM,78nM,19.5nM,4.9nM,1.2nM, 0.30nM, 0.076nM, 0.019 nM. Taking cancer cells in logarithmic growth phase, counting under microscope, and adjusting cell concentration to 7 × 10 with corresponding complete culture solution4seeds/mL are planted in 96-well culture plates, 100 mu L/well, placed at 37 ℃ and 5% CO2The incubator is used for 24 h. The culture solution is aspirated by a syringe (after suspension cells are centrifuged, the culture solution is aspirated again, adherent cells are directly aspirated), then 200 mul/well of the culture solution containing samples to be tested with different concentrations is respectively added, 200 mul of complete culture solution is directly added to a blank control well, and 3 duplicate wells are arranged for each concentration. After dosing, the cells were incubated at 37 ℃ with 5% CO2The incubator was incubated for a further 72 h. The culture solution was aspirated by syringe (suspension cells were centrifuged and then aspirated, adherent cells were directly aspirated), 200. mu.L of freshly prepared complete culture medium containing 10% MTT was added, and culture was continued for 4 h. The supernatant was aspirated, 150. mu.L DMSO was added to each well, shaken in the dark for 10min, and the OD of each well was measured at 490 nm. IC calculation Using SPSS17.0 software50The value is obtained.
The results are shown in Table 3:
TABLE 3 IC inhibition of proliferation of various tumor cells by Compounds 1-12 and other Compounds50Data of
Figure BDA0003283738550000331
The results show that for 5 tumor cells tested, the IC of the compounds 1-12 in vitro tumor cell proliferation inhibitory activity50All are lower than 0.2 mu M, and the activity of inhibiting the proliferation of tumor cells is stronger than that of the Apixagli. And wherein the IC of Compound 12 on five tumor cells tested50Are all less than 50nM and are 9-20 times the activity of Apigium melegueta. Has extremely high application prospect.
The above-mentioned embodiments only express the embodiments of the present invention, and the description thereof is more specific and detailed, but not construed as limiting the scope of the present invention. It should be noted that, for a person skilled in the art, several variations and modifications can be made without departing from the inventive concept, which falls within the scope of the present invention. Therefore, the protection scope of the present patent shall be subject to the appended claims.

Claims (10)

1. An α -fluoroacylpiperazine derivative characterized by: an α -fluoroacylpiperazine derivative having the general formula (I) or an isomer thereof, or a pharmaceutically acceptable salt thereof, or a prodrug molecule thereof, wherein the chemical structure of the general formula (I) is:
Figure FDA0003283738540000011
in the general formula (I), X ═ O or S; wherein the acyl alpha position of the acyl piperazine contains at least one fluorine atom;
in the general formula (I), R1Is H, halogen, cyano, alkyl, alkenyl, alkynyl of 1-15 carbons or derivatives thereof; or a monocyclic or fused ring aryl group having 5 to 22 carbon atoms or a derivative thereof; or a 5-to 8-membered heterocyclic or heterocyclic ring or derivative thereof containing 1 to 4 heteroatoms; or a carboxyl group or a derivative thereof; or hydroxy or a derivative thereof; or an amino group or a derivative thereof; or a mercapto group or a derivative thereof; or a sulfone or sulfoxide derivative; or a sulfonate or sulfonate salt; or a phosphate or phosphonate;
in the general formula (I), R2Is H, halogen, cyano, alkyl, alkenyl, alkynyl of 1-15 carbons or derivatives thereof; or a monocyclic or fused ring aryl group having 5 to 22 carbon atoms or a derivative thereof; or a 5-to 8-membered heterocyclic or heterocyclic ring or derivative thereof containing 1 to 4 heteroatoms; or a carboxyl group or a derivative thereof; or hydroxy or a derivative thereof; or an amino group or a derivative thereof; or a mercapto group or a derivative thereof; or a sulfone or sulfoxide derivative; or a sulfonate or sulfonate salt; or a phosphate or a phosphonate.
2. The alpha fluoroacylpiperazine derivative of claim 1An article, characterized in that: in the general formula (I), the alpha position of acyl piperazine is a single optical configuration or a racemate; r1And R2May be the same or different; in the general formula (I), R1And R2May be joined to form a ring structure.
3. The α -fluoroacylpiperazine derivative according to claim 1, wherein: the preferred structure is as follows:
Figure FDA0003283738540000021
4. a pharmaceutical composition characterized by: comprising a therapeutically effective amount of the α -fluoroacylpiperazine derivative of claim 1, or an isomer thereof, or a pharmaceutically acceptable salt thereof, or a prodrug molecule thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
5. The method for producing an α -fluoroacylpiperazine derivative according to claim 1, wherein: the reaction equation is as follows:
Figure FDA0003283738540000031
P1is H or an amino protecting group, P2Is H or an amino protecting group; p1、P2Not H at the same time;
the amino protecting group is formyl, acetyl, trifluoroacetyl, benzoyl, tert-butyloxycarbonyl, benzyloxycarbonyl, 9-fluorenylmethoxycarbonyl, phthaloyl, cyclobutanecarbonyl, 2-biphenyl-2-propoxycarbonyl, p-toluenesulfonyl or trityl;
the reducing agent used in the reductive amination is NaBH4、KBH4、LiBH4、Zn(BH4)2、NaBH3CN、NaBH(OAc)3Borane complex, Bu3SnH or PhSiH4
The catalyst used in reductive amination is protonic acid or Lewis acid;
the reductive amination reaction solvent is dichloromethane, tetrahydrofuran, methanol, ethanol, isopropanol, 1, 2-dichloroethane, ethylene glycol dimethyl ether or di (ethylene glycol) dimethyl ether;
the acid used for deprotection is protonic acid or Lewis acid;
the base used for deprotection is inorganic base or organic base;
the hydrogen source used in the catalytic reduction deprotection reaction is hydrogen, formic acid or ammonium formate; the catalyst is a metal catalyst;
the solvent used in the deprotection reaction is any one or the mixture of two of a protic solvent or an aprotic solvent;
the thionating agent is P2S5Lawson's reagent, 2, 4-bis (methylthio) -1,3,2, 4-dithiadiphosphobutylene-2, 4-disulfide, 2, 4-bis (phenylthio) -1, 3-disulfide-2, 4-diphosphetane-2, 4-disulfide, or 2, 4-bis (4-phenoxyphenyl) -1,3,2, 4-dithiodiphosphetane-2, 4-disulfide; the solvent used in the reaction is any one or the mixture of two of a protic solvent and an aprotic solvent;
the reaction temperature is-78-180 ℃.
6. The method for producing an α -fluoroacylpiperazine derivative according to claim 1, wherein: the reaction equation is as follows:
Figure FDA0003283738540000041
the fluorinating reagent is HF or salt thereof, SF4Diethylaminosulfur trifluoride, bis (2-methoxyethyl) aminosulfur trifluoride, 4-tert-butyl-2, 6-dimethylphenylsulfur trifluoride, pyridine-2-sulfonyl fluoride, sulfur trifluoride, sulfur dioxide, sulfur trifluoride, sulfur dioxide, sulfur trifluoride, sulfur trifluoride, and sulfur dioxide,Bis (2-methoxyethyl) aminosulfur trifluoride, diethylamino) difluorosulfonium tetrafluoroborate, difluoro (4-morpholino) sulfonium tetrafluoroborate, 1, 3-bis (2, 6-diisopropylphenyl) -2, 2-difluoroimidazoline, 4-chloro-N- [ (4-methylphenyl) sulfonyl chloride]-benzenesulfonamide fluoride; the solvent used in the reaction is an aprotic solvent or a mixed solvent, and the reaction temperature is-78-180 ℃.
The solvent used in the reaction is dichloromethane, dichloroethane, tetrahydrofuran, acetonitrile, ethylene glycol dimethyl ether or 1, 4-dioxane.
7. Use of an α -fluoroacylpiperazine derivative according to claim 1, or an isomer thereof, or a pharmaceutically acceptable salt thereof, or a prodrug molecule thereof, for the manufacture of a medicament for the treatment of cancer.
8. Use according to claim 7, characterized in that: the cancer is brain cancer, brain glioma, endometrial cancer, ovarian cancer, cervical cancer, breast cancer, colon cancer, lung cancer, prostate cancer, liver cancer, leukemia, lymph cancer, skin cancer, basal cell tumor, hemangioma, uterine cancer, laryngeal cancer, stomach cancer, lip cancer, esophageal cancer, nasopharyngeal cancer, gallbladder cancer, pancreatic cancer, kidney cancer, tongue cancer, bladder cancer, melanoma, lipoma, thyroid cancer, thymus cancer or bone cancer.
9. Use of an α -fluoroacylpiperazine derivative or an isomer thereof, or a pharmaceutically acceptable salt thereof, or a prodrug molecule thereof according to claim 1, in combination with at least one additional anticancer agent, for the manufacture of a medicament for the treatment of cancer.
10. Use according to claim 9, characterized in that: the additional anticancer agent is any one or a combination of more than two of adriamycin, bleomycin, vinblastine, taxanes, etoposide, 5-fluorouracil, cyclophosphamide, methotrexate, cisplatin, retinoic acid, temozolomide, actinomycin, imatinib, gefitinib, sorafenib, erlotinib, sunitinib, afatinib, cabozantinib, ostwalzumab, cetuximab, trastuzumab, nivolumab, panlizumab, attelizumab, daclizumab and avizumab.
CN202111140554.8A 2021-09-28 2021-09-28 Alpha fluoroacyl piperazine derivative and preparation and application thereof Active CN113754680B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202111140554.8A CN113754680B (en) 2021-09-28 2021-09-28 Alpha fluoroacyl piperazine derivative and preparation and application thereof
PCT/CN2022/105698 WO2023050975A1 (en) 2021-09-28 2022-07-14 α-FLUOROACYL PIPERAZINE DERIVATIVE, AND PREPARATION AND APPLICATION THEREOF

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111140554.8A CN113754680B (en) 2021-09-28 2021-09-28 Alpha fluoroacyl piperazine derivative and preparation and application thereof

Publications (2)

Publication Number Publication Date
CN113754680A true CN113754680A (en) 2021-12-07
CN113754680B CN113754680B (en) 2022-07-22

Family

ID=78797849

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111140554.8A Active CN113754680B (en) 2021-09-28 2021-09-28 Alpha fluoroacyl piperazine derivative and preparation and application thereof

Country Status (2)

Country Link
CN (1) CN113754680B (en)
WO (1) WO2023050975A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023050975A1 (en) * 2021-09-28 2023-04-06 云白药征武科技(上海)有限公司 α-FLUOROACYL PIPERAZINE DERIVATIVE, AND PREPARATION AND APPLICATION THEREOF
WO2023226176A1 (en) * 2022-05-23 2023-11-30 云白药征武科技(上海)有限公司 Thioamide derivatives, preparation method therefor, and use thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101600720A (en) * 2006-12-07 2009-12-09 霍夫曼-拉罗奇有限公司 Phosphoinositide 3-kinase inhibitor compounds and using method
WO2010080996A1 (en) * 2009-01-08 2010-07-15 Curis, Inc. Phosphoinositide 3-kinase inhibitors with a zinc binding moiety
CN102625708A (en) * 2009-09-09 2012-08-01 阿维拉制药公司 Pi3 kinase inhibitors and uses thereof
CN113045582A (en) * 2021-02-05 2021-06-29 中国药科大学 PARP-1/PI3K double-target inhibitor or pharmaceutically acceptable salt thereof, and preparation method and application thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113754680B (en) * 2021-09-28 2022-07-22 云白药征武科技(上海)有限公司 Alpha fluoroacyl piperazine derivative and preparation and application thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101600720A (en) * 2006-12-07 2009-12-09 霍夫曼-拉罗奇有限公司 Phosphoinositide 3-kinase inhibitor compounds and using method
WO2010080996A1 (en) * 2009-01-08 2010-07-15 Curis, Inc. Phosphoinositide 3-kinase inhibitors with a zinc binding moiety
CN102625708A (en) * 2009-09-09 2012-08-01 阿维拉制药公司 Pi3 kinase inhibitors and uses thereof
CN113045582A (en) * 2021-02-05 2021-06-29 中国药科大学 PARP-1/PI3K double-target inhibitor or pharmaceutically acceptable salt thereof, and preparation method and application thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023050975A1 (en) * 2021-09-28 2023-04-06 云白药征武科技(上海)有限公司 α-FLUOROACYL PIPERAZINE DERIVATIVE, AND PREPARATION AND APPLICATION THEREOF
WO2023226176A1 (en) * 2022-05-23 2023-11-30 云白药征武科技(上海)有限公司 Thioamide derivatives, preparation method therefor, and use thereof

Also Published As

Publication number Publication date
WO2023050975A1 (en) 2023-04-06
CN113754680B (en) 2022-07-22

Similar Documents

Publication Publication Date Title
KR102399848B1 (en) Novel JAK1 selective inhibitors and uses thereof
CN113754680B (en) Alpha fluoroacyl piperazine derivative and preparation and application thereof
US10016427B2 (en) Hydrochloride salt of((1S,2S,4R)-4-{4-[(1S)-2,3-dihydro-1H-inden-1-ylamino]-7H-pyrrolo[2,3-D]pyrimidin-7-YL}-2-hydroxycyclopentyl) methyl sulfamate
BR122023026304A2 (en) PYRIMIDOHETEROCYCLIC COMPOUNDS, PHARMACEUTICAL COMPOSITION AND USE OF SAID COMPOUNDS
RU2738654C1 (en) Novel cyclin-dependent kinase cdk9 inhibitor
KR20180051523A (en) Salt of LSD1 inhibitor
WO2015141616A1 (en) 1,3-benzodioxole derivative
TW201938556A (en) Compounds and their use in treating cancer
CN108473479B (en) Solid state forms of pladienolide pyridine compounds and methods of use
BR112015017963A2 (en) deuterated phenyl amino pyrimidine compound, method for preparing the pharmaceutical composition, pharmaceutical composition and use of the compound
WO2022087624A1 (en) Compounds as ras inhibitors and uses thereof
CN113825757B (en) Substituted fused bicyclic derivatives, preparation method thereof and application thereof in medicine
EP4143166A1 (en) Heterocyclic compounds as bet inhibitors
CN115942937A (en) Pyrimido-cyclic compounds
EP3988098A1 (en) Heterocyclo alkyl compounds used as ccr2/ccr5 antagonist
JP2023501110A (en) Aminopyrimidine compounds as CDK2/4/6 triple inhibitors
EP3964518A1 (en) Cd73 inhibitor, preparation method therefor and application thereof
CA3177298C (en) Compounds containing benzosultam as erk inhibitors
EP4349828A1 (en) Nitrogen-containing heterocyclic compound, preparation method therefor and application thereof
CA3199295A1 (en) New crystalline forms of a kras g12c inhibitor compound
WO2021053158A1 (en) Novel histone methyltransferase inhibitors
CN116783183A (en) 1- (2- (4-cyclopropyl-1H-1, 2, 3-triazol-1-yl) acetyl) -4-hydroxy-N- (benzyl) pyrrolidine-2-carboxamide derivatives as VHL inhibitors for the treatment of anemia and cancer
JP2022536574A (en) Solid Form BRD4 Inhibitor Compounds and Methods for Their Preparation and Uses
TWI839738B (en) Nitrogen heterocyclic compound, and preparation method and application thereof
CN116940581A (en) Novel protein degradation agent and application thereof

Legal Events

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