CN111423407A - Caffeoylquinic acid derivative and preparation method and application thereof - Google Patents

Caffeoylquinic acid derivative and preparation method and application thereof Download PDF

Info

Publication number
CN111423407A
CN111423407A CN201910022953.0A CN201910022953A CN111423407A CN 111423407 A CN111423407 A CN 111423407A CN 201910022953 A CN201910022953 A CN 201910022953A CN 111423407 A CN111423407 A CN 111423407A
Authority
CN
China
Prior art keywords
caffeoylquinic acid
acid derivative
caffeoylquinic
preparation
reaction
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
CN201910022953.0A
Other languages
Chinese (zh)
Other versions
CN111423407B (en
Inventor
田瑜
许旭东
郭鹏
吴崇明
尚海
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.)
Institute of Medicinal Plant Development of CAMS and PUMC
Original Assignee
Institute of Medicinal Plant Development of CAMS and PUMC
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 Institute of Medicinal Plant Development of CAMS and PUMC filed Critical Institute of Medicinal Plant Development of CAMS and PUMC
Priority to CN201910022953.0A priority Critical patent/CN111423407B/en
Publication of CN111423407A publication Critical patent/CN111423407A/en
Application granted granted Critical
Publication of CN111423407B publication Critical patent/CN111423407B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/44Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D317/46Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C231/00Preparation of carboxylic acid amides
    • C07C231/02Preparation of carboxylic acid amides from carboxylic acids or from esters, anhydrides, or halides thereof by reaction with ammonia or amines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/40Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of rings other than six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated
    • 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

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Urology & Nephrology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a caffeoylquinic acid derivative, a preparation method and application thereof, and the caffeoylquinic acid derivative has a structure shown in a general formula (I). The invention takes 3-O-caffeoylquinic acid as a raw material, reserves the structural skeleton of the 3-O-caffeoylquinic acid and is positioned at 1 positionDifferent amino substituent groups are introduced into carboxyl, 4, 5-position hydroxyl groups are substituted or exposed by ketal, and the novel caffeoylquinic acid derivative with lipid-lowering activity is synthesized, the synthesis method is simple and convenient, and the product purity is high. Compared with 3-O-caffeoylquinic acid, the caffeoylquinic acid derivative provided by the invention has better lipid-lowering activity.
Figure DDA0001941435280000011

Description

Caffeoylquinic acid derivative and preparation method and application thereof
Technical Field
The invention relates to the field of pharmaceutical chemistry and therapeutics, in particular to a caffeoylquinic acid derivative, a preparation method and application thereof.
Background
The hyperlipidemia (hyperlipidemia, H L P) can induce clinical common cardiovascular and cerebrovascular diseases, and is one of the potential factors threatening human health, at present, the dyslipidemia patients in China reach 2.0 hundred million, the morbidity of normal people reaches 35-40%, and the middle-aged and elderly people reach more than 60%.
Currently, the common lipid-lowering drugs in clinic mainly include statin lipid-regulating drugs (atorvastatin and lovastatin), fibrate drugs (gemfibrozil and fenofibrate), nicotinic acid drugs (acipimox) and the like. However, these drugs acting on a single target point have various unsatisfactory side effects in clinical application, such as severe myopathy and rhabdomyolysis, and hepatotoxicity in the current first ten drugs of the world sold atorvastatin (also known as lipitor) as an HMG-CoA reductase inhibitor; even the new drug lomitapide, which has just been approved by the FDA to be marketed in 12 months 2012 for the treatment of familial hypercholesterolemia, is indicated to be at risk for gastrointestinal reactions and hepatotoxicity. Therefore, the search for lipid-lowering drugs with significant efficacy, safety and reliability is a subject worthy of long-term research by medical workers.
However, from the actual situation of our country, we completely imitate the western countries to find the lead compound from the artificially synthesized product, and create a new lipid-lowering drug with high efficiency and low toxicity, which is difficult to get a major breakthrough in a short period, but the natural product rich in the traditional clinically effective drug has the specific structural diversity and multi-target action mechanism, and is more and more favored by researchers at home and abroad, such as berberine which is the main lipid-lowering active ingredient in the Chinese medicine coptis root.
The action mechanism of traditional Chinese medicine lipid regulation is complex compared with western medicines, adverse reactions are less, and lipid reduction is achieved mainly by reducing absorption of exogenous lipid in intestinal tracts, inhibiting synthesis of endogenous lipid and regulating lipid metabolism. For example, a single Chinese medicine (kudzu root, ginseng, salvia miltiorrhiza, ginkgo leaf and the like) with the function of regulating blood fat; the lipid-lowering natural product can be polysaccharides (fructus Lycii polysaccharide, fructus Cucurbitae Moschatae polysaccharide), flavonoids (dihydrotanshinone, folium Ginkgo flavone) and polyphenols (catechin). In recent years, many folk traditional medicines are found to have good lipid regulating effect and are gradually developed into medicines, for example, a Xuezhikang capsule (main component red yeast) developed by limited northern Weixin biotechnology has completed phase II clinical research in the United states at present, and Jianjiandong subject group of institute of biotechnology of Chinese academy of medicine sciences finds that berberine also has good lipid lowering activity, is completely different from the action mechanism of statin lipid lowering medicines, and theoretically provides a new molecular target for searching for novel lipid lowering medicines. Different from the single-target western medicines which are clinically and frequently adopted at the present stage and easily cause adverse reactions, the multi-target characteristics of the natural medicines are probably the reasons for less toxic and side effects, so the method has epoch significance for the deep exploration and research of the natural medicines.
Disclosure of Invention
In view of the above, the present invention aims to provide a caffeoylquinic acid derivative, and a preparation method and a use thereof, wherein the caffeoylquinic acid derivative has a good lipid-lowering effect.
In view of the above object, the present invention provides a caffeoylquinic acid derivative having a structure represented by general formula (i):
Figure BDA0001941435260000021
in the formula: r6Is an H atom;
R4and R5Is an H atom;
R2and R3Is H atom or propylidene;
R1selected from alkylamines containing 1-18 carbons, arylamines containing 6-10 carbons or saturated or unsaturated cyclic amines containing 3-7 carbons.
In some embodiments of the invention, R1Selected from alkylamines containing 3-8 carbons, arylamines containing 7-8 carbons or saturated cyclic amines containing 5-6 carbons.
In some embodiments of the invention, R1Selected from n-butylamino, isobutylamino, n-octylamino, propargylamino, benzylamino, piperidinyl or cyclohexylamino.
The invention also provides a preparation method of the caffeoylquinic acid derivative, which comprises the following steps:
(1) reacting 3-O-caffeoylquinic acid with 2, 2-dimethoxypropane to generate an intermediate;
(2) and carrying out condensation reaction on the intermediate and amine to generate a target compound caffeoylquinic acid derivative.
In some embodiments of the present invention, after step (2), further comprising:
(3): carrying out deprotection reaction on the target compound caffeoylquinic acid derivative.
In some embodiments of the present invention, the method for preparing caffeoylquinic acid derivatives comprises the following steps:
(1) reacting 3-O-caffeoylquinic acid and 2, 2-dimethoxypropane in anhydrous acetone by taking p-toluenesulfonic acid as a catalyst to generate an intermediate shown in a formula (II);
Figure BDA0001941435260000031
(2) the intermediate shown in the formula (II) is subjected to condensation reaction with amine in a mixed solvent of anhydrous acetonitrile and tetrahydrofuran under the condensation condition of BOP and DIEA to generate the target compound caffeoylquinic acid derivative.
In some embodiments of the present invention, after step (2), further comprising:
(3): removing acetonylidene protection from a target compound caffeoylquinic acid derivative in a mixed solution of trifluoroacetic acid, dichloromethane and water.
The invention also provides a pharmaceutical composition, which comprises the caffeoylquinic acid derivative and a medicinal carrier.
The invention also provides application of the caffeoylquinic acid derivatives in preparing a medicament for preventing and/or treating hyperlipidemia.
The invention also provides application of the caffeoylquinic acid derivatives in preparing medicaments for preventing and/or treating atherosclerosis.
From the above, the invention can be seen in that 3-O-caffeoylquinic acid is used as a raw material, the structural skeleton of the 3-O-caffeoylquinic acid is kept, different amino substituent groups are introduced to the carboxyl group at the 1 position, and the hydroxyl groups at the 4 and 5 positions are substituted or exposed by ketal, so that the novel caffeoylquinic acid derivative with lipid-lowering activity is synthesized, the synthesis method is simple and convenient, and the product purity is high. Compared with 3-O-caffeoylquinic acid, the caffeoylquinic acid derivative provided by the invention has better lipid-lowering activity. The invention fully utilizes medicinal plant resources found from natural products, deeply researches and develops to find compounds with unique chemical structures, and finds the lipid-lowering medicament for clinical application.
Drawings
FIG. 1 is a graph of the modulating effect of chlorogenic acid and its derivatives on oleic acid-induced lipid accumulation in HepG2 hepatocytes; wherein, A picture shows the regulation effect of caffeoylquinic acid derivatives 3a, 3b, 3c, 3d, 3e and 3f on lipid accumulation caused by oleic acid in HepG2 liver cells; the B picture shows that the caffeoylquinic acid derivatives 4a, 4B, 4c, 4d, 4e and 4f have the regulating effect on lipid accumulation initiated by oleic acid in HepG2 liver cells, CA is chlorogenic acid, S is simvastatin, M is oleic acid, K is a control, and # p <0.001 is very obvious compared with the cells (K) treated by the control group; p <0.05 was significantly different relative to cells treated with oleic acid (M); p <0.01 was significant relative to oleic acid (M) -treated cells; the cells differed significantly with p <0.001 versus oleic acid (M) treated cells.
Detailed Description
In order to make the objects, technical solutions and advantages of the present invention more apparent, the present invention is further described in detail with reference to specific embodiments below.
The invention provides a caffeoylquinic acid derivative, which has a structure shown in a general formula (I):
Figure BDA0001941435260000041
in the formula: r6Is an H atom;
R4and R5Is an H atom;
R2and R3Is H atom or propylidene;
R1selected from alkylamines containing 1-18 carbons, arylamines containing 6-10 carbons or saturated or unsaturated cyclic amines containing 3-7 carbons.
As an embodiment of the present invention, R1Selected from alkylamines containing 3-8 carbons, arylamines containing 7-8 carbons or saturated cyclic amines containing 5-6 carbons.
As an embodiment of the present invention, R1Selected from n-butylamino, isobutylamino, n-octylamino, propargylamino, benzylamino, piperidinyl or cyclohexylamino.
As an embodiment of the present invention, when R6Is an H atom; r4And R5Is an H atom; r2And R3Is propylidene; the structure shown in the formula (I) is as follows:
Figure BDA0001941435260000051
when R is1Is n-butylamino, and is a tertiary amino,
Figure BDA0001941435260000052
the caffeoylquinic acid derivative is now designated 3 a.
When R is1Is an isobutylamino group, and the like,
Figure BDA0001941435260000053
the caffeoylquinic acid derivative is named 3 b.
When R is1Is a n-octylamino group, and the amino acid is,
Figure BDA0001941435260000054
the caffeoylquinic acid derivative is named 3 c.
When R is1Is a propargylamine group, and is a propargylamine group,
Figure BDA0001941435260000055
the caffeoylquinic acid derivative is named 3 d.
When R is1Is a benzylamino group, and is a benzyl amine group,
Figure BDA0001941435260000056
the caffeoylquinic acid derivative is named 3 e.
When R is1Is a piperidine group, and is a cyclic or cyclic piperidine group,
Figure BDA0001941435260000057
the caffeoylquinic acid derivative is named 3 f.
When R is1Is a cyclohexylamino group, and is a cyclohexylamino group,
Figure BDA0001941435260000058
the caffeoylquinic acid derivative was named 3g at this time.
As an embodiment of the present invention, when R6Is an H atom; r4And R5Is an H atom; r2And R3Is an H atom; the structure shown in the formula (I) is as follows:
Figure BDA0001941435260000061
when R is1Is n-butylamino, and is a tertiary amino,
Figure BDA0001941435260000062
the caffeoylquinic acid derivative is now designated 4 a.
When R is1Is an isobutylamino group, and the like,
Figure BDA0001941435260000063
the caffeoylquinic acid derivative is named 4 b.
When R is1Is a n-octylamino group, and the amino acid is,
Figure BDA0001941435260000064
the caffeoylquinic acid derivative is named 4 c.
When R is1Is a propargylamine group, and is a propargylamine group,
Figure BDA0001941435260000065
the caffeoylquinic acid derivative is named 4 d.
When R is1Is a benzylamino group, and is a benzyl amine group,
Figure BDA0001941435260000066
the caffeoylquinic acid derivative is named 4 e.
When R is1Is a piperidine group, and is a cyclic or cyclic piperidine group,
Figure BDA0001941435260000067
the caffeoylquinic acid derivative is named 4 f.
When R is1Is a cyclohexylamino group, and is a cyclohexylamino group,
Figure BDA0001941435260000068
the caffeoylquinic acid derivative was named 4g at this time.
The invention also provides a preparation method of the caffeoylquinic acid derivative, which comprises the following steps:
(1) reacting 3-O-caffeoylquinic acid with 2, 2-dimethoxypropane to generate an intermediate;
(2) and carrying out condensation reaction on the intermediate and amine to generate a target compound caffeoylquinic acid derivative.
Specifically, the synthetic route of the caffeoylquinic acid derivative is as follows:
(1) synthesis reaction of intermediate 2
Figure BDA0001941435260000071
Taking a compound 1 (3-O-caffeoylquinic acid) as an initial raw material, and reacting 4, 5-hydroxy of the 3-O-caffeoylquinic acid with 2, 2-Dimethyloxypropane (DMP) in anhydrous acetone under the catalysis of p-toluenesulfonic acid (TsOH) to obtain an intermediate 2;
wherein, the structural formula of the 2, 2-dimethoxypropane is as follows:
Figure BDA0001941435260000072
(2) synthesis reaction of target Compound
Figure BDA0001941435260000073
Intermediate 2 was condensed with BOP (benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate) and DIEA (N, N-diisopropylethylamine) in anhydrous acetonitrile (CH)3CN) and Tetrahydrofuran (THF) to generate a target compound caffeoylquinic acid derivative through a condensation reaction with amine, wherein the target compound caffeoylquinic acid derivative comprises 3a, 3b, 3c, 3d, 3e, 3f and 3 g.
In one embodiment of the present invention, the target compound caffeoylquinic acid derivative of step (2) is subjected to deprotection reaction to obtain the target compounds caffeoylquinic acid derivatives 4a, 4b, 4c, 4d, 4e, 4f and 4 g.
Specifically, the synthetic route is as follows:
Figure BDA0001941435260000081
the caffeoylquinic acid derivatives of the target compounds comprise 3a, 3b, 3c, 3d, 3e, 3f and 3g, and the acetonylidene protection is removed from a mixed solution of trifluoroacetic acid (TFA), Dichloromethane (DCM) and water, so that caffeoylquinic acid derivatives of the target compounds, 4a, 4b, 4c, 4d, 4e, 4f and 4g, are respectively obtained.
The general synthetic route of the target compound caffeoylquinic acid derivative is as follows:
Figure BDA0001941435260000082
wherein, (A) DMP, TsOH, acetone; (B) BOP, DIEA, amines; (C) TFA, DCM, H2O(9:1:1).
Therefore, according to the structural characteristics of the 3-O-caffeoylquinic acid, the hydroxyl at the 4, 5-position of the 3-O-caffeoylquinic acid is reacted with 2, 2-Dimethoxypropane (DMP), so that the hydroxyl at the 4, 5-position of the 3-O-caffeoylquinic acid is substituted by ketal to obtain an intermediate 2; then reacting the intermediate 2 with various amines, and introducing different amino substituent groups on the 1-position carboxyl group to obtain the caffeoylquinic acid derivatives; or the acetonylidene protection is further removed from the obtained caffeoylquinic acid derivative, so that the hydroxyl groups at the 4 and 5 positions are exposed. The preparation method of the caffeoylquinic acid derivative provided by the invention has the advantages of simple and convenient synthesis method and high product purity.
Example 1 preparation of caffeoylquinic acid derivative 3a
(1) Preparation of intermediate 2
Accurately weighing 7.0g (19.8mmol) of 3-O-caffeoylquinic acid (compound 1) in a round-bottom flask with the thickness of 250m L, adding 60m L anhydrous acetone and 40m L DMP (2, 2-dimethoxypropane), stirring to react to form suspension, adding a catalytic amount of 50mg TsOH (p-toluenesulfonic acid) into the reaction liquid, stirring to react until the reaction liquid is clear, reacting at room temperature, monitoring by T L C, and after the reaction is finished, using anhydrous Na2CO3And neutralizing until the pH value is 5-6, performing suction filtration, performing reduced pressure evaporation to remove the solvent, and purifying the residue by silica gel column chromatography to obtain an intermediate 2 with the yield of 87%.
(2) Preparation of caffeoylquinic acid derivative 3a
In a 250m L round-bottom flask, exactly 21.2 g (3mmol) of intermediate and 1.5g (3mmol) of BOP are weighed, and 70m L of anhydrous mixed solvent THF/CH is added under the protection of argon3CN (v/v ═ 3:2) was stirred until dissolved, 0.8g (6mmol) of DIEA was added, 3mmol of n-butylamine was added dropwise to the reaction solution until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 3a as pale yellow powder with a yield of 81%.
1H-NMR(600MHz,DMSO):9.58(s,1H,Ph-OH),9.14(s,1H,Ph-OH),7.71 (t,J=6.0Hz,1H,NH),7.48(d,J=15.8Hz,1H,H-7′),7.05(d,J=1.7Hz,1H,H- 2′),7.00(dd,J=8.3Hz,1.7Hz,1H,H-6’),6.77(d,J=8.2Hz,1H,H-5’),6.24(d, J=15.9Hz,1H,H-8′),5.41(s,1H,H-1),5.37-5.32(m,1H,H-3),4.43-4.41(m, 1H,H-4),4.13-4.11(m,1H,H-5),3.11-3.01(m,2H,NH-CH2),2.26-2.22(m,1H, H-2),1.99-1.94(m,1H,H-2),1.80-1.73(m,2H,H-6),1.42(s,3H,CH3-C-O), 1.39-1.36(m,2H,NH-CH2-CH2),1.26-1.22(m,5H,CH3-C-O,CH2-CH3),0.84(t, J=7.3Hz,3H,CH2-CH3);13C-NMR(150MHz,DMSO):175.0,165.9,148.4,145.6,145.3,125.6,121.4,115.8,114.9,114.1,108.0,76.9,74.1,73.5,70.9,38.2,37.0,34.5,31.2,28.0,25.9,19.4,13.7;HRMS(ESI):Calcd for[M+Na]+C23H31NNaO8:472.1947,found 472.1948。
Example 2 preparation of caffeoylquinic acid derivative 4a
(3) Preparation of caffeoylquinic acid derivative 4a
In a 50m L round-bottomed flask, accurately weighing 2mmol caffeoylquinic acid derivative 3a, respectively, and adding TFA/DCM/H2O (v/v ═ 9:1:1)20m L, the reaction was stirred at room temperature for 0.5 h until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 4a as a pale yellow powder in 78% yield.
1H-NMR(600MHz,MeOD):7.58(d,J=15.9Hz,1H,H-7′),7.05(d,J=1.9 Hz,1H,H-2′),6.95(dd,J=8.2Hz,1.9Hz,1H,H-6’),6.78(d,J=8.1Hz,1H,H- 5’),6.29(d,J=15.9Hz,1H,H-8′),5.42-5.38(m,1H,H-3),4.24-4.23(m,1H,H- 4),3.72-3.70(m,1H,H-5),3.21-3.19(m,2H,NH-CH2),2.12-2.07(m,2H,H-2), 2.02-2.00(m,1H,H-6),1.95-1.91(m,1H,H-6),1.52-1.47(m,2H,NH-CH2-CH2), 1.36-1.32(m,2H,CH2-CH3),0.93(t,J=7.4Hz,3H,CH2-CH3);13C-NMR(150 MHz,MeOD):176.6,169.0,149.6,147.0,146.8,127.8,122.9,116.5,115.4, 115.2,77.8,74.4,72.7,71.9,40.0,39.9,38.7,32.6,21.0,14.1;HRMS(ESI):Calcd for[M+Na]+C20H27NNaO8:432.1634,found 432.1636。
Example 3 preparation of caffeoylquinic acid derivative 3b
(1) Preparation of intermediate 2
Accurately weighing 7.0g (19.8mmol) of 3-O-caffeoylquinic acid (compound 1) in a round-bottom flask with the thickness of 250m L, adding 60m L anhydrous acetone and 40m L DMP (2, 2-dimethoxypropane), stirring to react to form suspension, adding a catalytic amount of 50mg TsOH (p-toluenesulfonic acid) into the reaction liquid, stirring to react until the reaction liquid is clear, reacting at room temperature, monitoring by T L C, and after the reaction is finished, using anhydrous Na2CO3And neutralizing until the pH value is 5-6, performing suction filtration, performing reduced pressure evaporation to remove the solvent, and purifying the residue by silica gel column chromatography to obtain an intermediate 2 with the yield of 87%.
(2) Preparation of caffeoylquinic acid derivative 3b
In a 250m L round-bottom flask, exactly 21.2 g (3mmol) of intermediate and 1.5g (3mmol) of BOP are weighed, and 70m L of anhydrous mixed solvent THF/CH is added under the protection of argon3CN (v/v ═ 3:2) was stirred until dissolved, 0.8g (6mmol) of DIEA was added, 3mmol of isobutylamine was added dropwise to the reaction solution until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 3b as pale yellow powder in 77% yield.
1H-NMR(600MHz,DMSO):7.70(t,J=6.1Hz,1H,NH),7.47(d,J=15.8 Hz,1H,H-7′),7.04(d,J=2.0Hz,1H,H-2′),7.00(dd,J=8.2Hz,2.0Hz,1H,H- 6’),6.76(d,J=8.2Hz,1H,H-5’),6.24(d,J=15.9Hz,1H,H-8′),5.47(br,1H,H- 1),5.37-5.33(m,1H,H-3),4.43-4.41(m,1H,H-4),4.14-4.11(m,1H,H-5), 2.94-2.86(m,2H,NH-CH2),2.26-2.22(m,1H,H-2),1.99-1.96(m,1H,H-2), 1.80-1.75(m,2H,H-6),1.74-1.69(m,1H,NH-CH2-CH),1.42(s,3H,CH3-C-O), 1.26(s,3H,CH3-C-O),0.82-0.80(m,6H,2×CH3);13C-NMR(150MHz,DMSO) :175.1,165.9,148.5,145.6,145.4,125.6,121.4,115.8,114.9,114.1,108.0,76.9, 74.2,73.6,70.9,45.9,37.1,34.4,28.1,26.0,20.0;HRMS(ESI):Calcd for[M+ Na]+C23H31NNaO8:472.1947,found 472.1946。
Example 4 preparation of caffeoylquinic acid derivative 4b
(3) Preparation of caffeoylquinic acid derivative 4b
In a 50m L round-bottomed flask, 2mmol of caffeoylquinic acid derivative 3b was accurately weighed out, respectively, and TFA/DCM/H was added2O (v/v ═ 9:1:1)20m L, the reaction was stirred at room temperature for 0.5 h until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 4b as a pale yellow powder with a yield of 85%.
1H-NMR(600MHz,MeOD):7.57(d,J=15.7Hz,1H,H-7′),7.05(s,1H,H- 2′),6.99-6.89(m,1H,H-6’),6.78(d,J=7.7Hz,1H,H-5’),6.29(d,J=15.7Hz, 1H,H-8′),5.46-5.37(m,1H,H-3),4.30-4.18(m,1H,H-4),3.78-3.66(m,1H,H- 5),3.09-2.95(m,2H,NH-CH2),2.11-1.94(m,1H,H-2,6),1.85-1.73(m,1H, NH-CH2-CH),0.94-0.84(m,6H,2×CH3);13C-NMR(150MHz,MeOD):176.7, 169.0,149.5,147.0,146.7,127.7,122.9,116.5,115.3,115.2,77.8,74.3,72.6,71.9, 47.6,40.0,38.7,29.6,20.3;HRMS(ESI):Calcd for[M+Na]+C20H27NNaO8: 432.1634,found 432.1638.
Example 5 preparation of caffeoylquinic acid derivative 3c
(1) Preparation of intermediate 2
In a 250m L round bottom flask, 7.0g (19.8mmol) of 3-O-caffeoylquinic acid (Compound 1) was accurately weighed and 60 was addedm L anhydrous acetone and 40m L DMP (2, 2-dimethoxypropane), stirring to obtain suspension, adding a catalytic amount of 50mg TsOH (p-toluenesulfonic acid) into the reaction solution, stirring to react until the reaction solution is clear, reacting at room temperature, and monitoring with T L C, and reacting with anhydrous Na2CO3And neutralizing until the pH value is 5-6, performing suction filtration, performing reduced pressure evaporation to remove the solvent, and purifying the residue by silica gel column chromatography to obtain an intermediate 2 with the yield of 87%.
(2) Preparation of caffeoylquinic acid derivative 3c
In a 250m L round-bottom flask, exactly 21.2 g (3mmol) of intermediate and 1.5g (3mmol) of BOP are weighed, and 70m L of anhydrous mixed solvent THF/CH is added under the protection of argon3CN (v/v ═ 3:2) was stirred until dissolved, 0.8g (6mmol) of DIEA was added, 3mmol of n-octylamine was added dropwise to the reaction solution until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 3C as pale yellow powder in 82% yield.
1H-NMR(600MHz,DMSO):7.74(t,J=5.9Hz,1H,NH),7.47(d,J=15.8 Hz,1H,H-7′),7.04(d,J=2.0Hz,1H,H-2′),7.00(dd,J=8.2Hz,2.0Hz,1H, H-6’),6.76(d,J=8.2Hz,1H,H-5’),6.24(d,J=15.9Hz,1H,H-8′),5.44(br,1H, H-1),5.36-5.31(m,1H,H-3),4.43-4.40(m,1H,H-4),4.13-4.11(m,1H,H-5), 3.10-2.98(m,2H,NH-CH2),2.25-2.22(m,1H,H-2),1.97-1.94(m,1H,H-2), 1.75-1.74(m,2H,H-6),1.41(s,3H,CH3-C-O),1.40-1.37(m,2H,NH-CH2-CH2), 1.25(s,3H,CH3-C-O),1.25-1.19(m,10H,(CH2)5-CH3),0.83(t,J=7.1Hz,3H, (CH2)5-CH3);13C-NMR(150MHz,DMSO):174.9,165.8,148.4,145.5,145.2,125.5,121.3,115.7,114.8,114.0,107.9,76.8,73.9,73.4,70.8,38.4,37.0,34.5, 31.1,28.9,28.6,28.5,27.9,26.1,25.8,22.0,13.8;HRMS(ESI):Calcd for[M+ Na]+C27H39NNaO8:528.2573,found 528.2578。
EXAMPLE 6 preparation of caffeoylquinic acid derivative 4c
(3) Preparation of caffeoylquinic acid derivative 4c
At 50m L round bottomIn a flask, 2mmol of caffeoylquinic acid derivative 3c is accurately weighed, and TFA/DCM/H is added2O (v/v ═ 9:1:1)20m L, the reaction was stirred at room temperature for 0.5 h until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 4C as a pale yellow powder with a yield of 76%.
1H-NMR(400MHz,MeOD):7.58(d,J=15.9Hz,1H,H-7′),7.05(d,J=1.9 Hz,1H,H-2′),6.95(dd,J=8.2Hz,1.9Hz,1H,H-6’),6.78(d,J=8.2Hz,1H,H- 5’),6.29(d,J=15.9Hz,1H,H-8′),5.43-5.37(m,1H,H-3),4.24-4.23(m,1H,H- 4),3.72-3.69(m,1H,H-5),3.20-3.17(m,2H,NH-CH2),2.13-1.90(m,1H,H-2, 6),1.52-1.49(m,2H,NH-CH2-CH2),1.34-1.26(m,10H,(CH2)5-CH3),0.87(t,J =7.1Hz,3H,-CH3);13C-NMR(100MHz,MeOD):176.6,169.0,149.6,147.0, 146.8,127.8,123.0,116.5,115.3,115.1,77.7,74.4,72.7,72.0,40.2,40.0,38.7, 33.0,30.5,30.4,27.9,23.7,14.4;HRMS(ESI):Calcd for[M+Na]+C24H35NNaO8:488.2260,found 488.2252。
Example 7 preparation of caffeoylquinic acid derivative 3d
(1) Preparation of intermediate 2
Accurately weighing 7.0g (19.8mmol) of 3-O-caffeoylquinic acid (compound 1) in a round-bottom flask with the thickness of 250m L, adding 60m L anhydrous acetone and 40m L DMP (2, 2-dimethoxypropane), stirring to react to form suspension, adding a catalytic amount of 50mg TsOH (p-toluenesulfonic acid) into the reaction liquid, stirring to react until the reaction liquid is clear, reacting at room temperature, monitoring by T L C, and after the reaction is finished, using anhydrous Na2CO3And neutralizing until the pH value is 5-6, performing suction filtration, performing reduced pressure evaporation to remove the solvent, and purifying the residue by silica gel column chromatography to obtain an intermediate 2 with the yield of 87%.
(2) Preparation of caffeoylquinic acid derivative 3d
In a 250m L round-bottom flask, exactly 21.2 g (3mmol) of intermediate and 1.5g (3mmol) of BOP are weighed, and 70m L of anhydrous mixed solvent THF/CH is added under the protection of argon3CN (v/v ═ 3:2) was stirred until dissolved. 0.8g (6 m) ofmol) of DIEA, 3mmol of propargylamine was added dropwise to the reaction solution until the reaction was monitored by T L C, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 3d as a pale yellow powder in a yield of 75%.
1H-NMR(600MHz,DMSO):8.16(t,J=5.8Hz,1H,NH),7.47(d,J=15.8 Hz,1H,H-7′),7.04(d,J=2.0Hz,1H,H-2′),7.00(dd,J=8.2Hz,2.0Hz,1H,H- 6’),6.76(d,J=8.2Hz,1H,H-5’),6.24(d,J=15.8Hz,1H,H-8′),5.50(s,1H,H-1), 5.35-5.31(m,1H,H-3),4.43-4.41(m,1H,H-4),4.14-4.12(m,1H,H-5),3.84(dd, J=5.8Hz,2.5Hz,2H,NH-CH2),3.03(t,J=2.5Hz,1H,CCH),2.25-2.22(m,1H, H-2),2.00-1.95(m,1H,H-2),1.79-1.70(m,2H,H-6),1.42(s,3H,CH3-C-O), 1.26(s,3H,CH3-C-O);13C-NMR(150MHz,DMSO):175.3,166.0,148.5,145.7, 145.5,125.7,121.5,115.9,114.9,114.2,108.2,81.2,76.9,74.2,73.6,72.6,70.9, 37.0,34.3,28.4,28.1,26.0;HRMS(ESI):Calcd for[M+Na]+C22H25NNaO8:454.1478,found 454.1480。
EXAMPLE 8 preparation of caffeoylquinic acid derivative 4d
(3) Preparation of caffeoylquinic acid derivative 4d
In a 50m L round-bottomed flask, 2mmol of caffeoylquinic acid derivative 3d was accurately weighed out, respectively, and TFA/DCM/H was added2O (v/v ═ 9:1:1)20m L, the reaction was stirred at room temperature for 0.5 h until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 4d as pale yellow powder in 69% yield.
1H-NMR(400MHz,MeOD):7.57(d,J=15.9Hz,1H,H-7′),7.11-7.00(m, 1H,H-2′),6.99-6.89(m,1H,H-6’),6.78(d,J=8.0Hz,1H,H-5’),6.29(d,J=15.9 Hz,1H,H-8′),5.43-5.37(m,1H,H-3),4.29-4.19(m,1H,H-4),4.03-4.92(m,2H, NH-CH2),3.74-3.71(m,1H,H-5),2.61-2.49(m,1H,CCH),2.15-1.95(m,4H,H- 2,6);13C-NMR(100MHz,MeOD):176.5,169.0,149.5,147.0,146.7,127.7, 123.0,116.5,115.3,115.1,80.5,77.8,74.3,72.5,72.1,71.8,39.8,38.5,29.4; HRMS(ESI):Calcd for[M+Na]+C19H21NNaO8:414.1165,found 414.1667。
EXAMPLE 9 preparation of caffeoylquinic acid derivative 3e
(1) Preparation of intermediate 2
Accurately weighing 7.0g (19.8mmol) of 3-O-caffeoylquinic acid (compound 1) in a round-bottom flask with the thickness of 250m L, adding 60m L anhydrous acetone and 40m L DMP (2, 2-dimethoxypropane), stirring to react to form suspension, adding a catalytic amount of 50mg TsOH (p-toluenesulfonic acid) into the reaction liquid, stirring to react until the reaction liquid is clear, reacting at room temperature, monitoring by T L C, and after the reaction is finished, using anhydrous Na2CO3And neutralizing until the pH value is 5-6, performing suction filtration, performing reduced pressure evaporation to remove the solvent, and purifying the residue by silica gel column chromatography to obtain an intermediate 2 with the yield of 87%.
(2) Preparation of caffeoylquinic acid derivative 3e
In a 250m L round-bottom flask, exactly 21.2 g (3mmol) of intermediate and 1.5g (3mmol) of BOP are weighed, and 70m L of anhydrous mixed solvent THF/CH is added under the protection of argon3CN (v/v ═ 3:2) was stirred to dissolve, 0.8g (6mmol) of DIEA was added, 3mmol of benzylamine was added dropwise to the reaction solution until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 3e as pale yellow powder with a yield of 79%.
1H-NMR(600MHz,DMSO):8.32(t,J=6.2Hz,1H,NH),7.48(d,J=15.8 Hz,1H,H-7′),7.31-7.28(m,2H,Ph-H),7.24-7.20(m,3H,Ph-H),7.05(d,J=2.0 Hz,1H,H-2′),7.01(dd,J=8.2Hz,2.0Hz,1H,H-6’),6.77(d,J=8.2Hz,1H,H- 5’),6.25(d,J=15.9Hz,1H,H-8′),5.55(br,1H,H-1),5.39-5.35(m,1H,H-3), 4.45-4.42(m,1H,H-4),4.32-4.25(m,2H,NH-CH2),4.15-4.13(m,1H,H-5), 2.29-2.25(m,1H,H-2),2.04-2.01(m,1H,H-2),1.84-1.76(m,2H,H-6),1.43(s, 3H,CH3-C-O),1.26(s,3H,CH3-C-O);13C-NMR(150MHz,DMSO):175.2, 165.8,148.4,145.5,145.3,139.5,128.1,126.9,126.7,125.5,121.3,115.7,114.8, 114.0,108.0,76.8,74.1,73.4,70.8,42.0,37.0,34.5,27.9,25.8;HRMS(ESI):Calcd for[M+Na]+C26H29NNaO8:506.1791,found 506.1790。
EXAMPLE 10 preparation of caffeoylquinic acid derivative 4e
(3) Preparation of caffeoylquinic acid derivative 4e
In a 50m L round-bottomed flask, 2mmol of caffeoylquinic acid derivative 3e was accurately weighed out, respectively, and TFA/DCM/H was added2O (v/v ═ 9:1:1)20m L, the reaction was stirred at room temperature for 0.5 h until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 4e as a pale yellow powder with a yield of 73%.
1H-NMR(600MHz,MeOD):7.58(d,J=15.9Hz,1H,H-7′),7.32-7.22(m, 5H,Ph-H),7.05(s,1H,H-2′),6.96-6.94(m,1H,H-6’),6.78(d,J=8.1Hz,1H,H- 5’),6.29(d,J=15.8Hz,1H,H-8′),5.45-5.39(m,1H,H-3),4.39(m,2H,NH-CH2), 4.26-4.25(m,1H,H-4),3.73-3.69(m,1H,H-5),2.17-1.97(m,4H,H-2,6);13C- NMR(150MHz,MeOD):176.8,169.0,149.5,147.0,146.8,139.9,129.5,128.2, 128.2,128.0,122.9,116.5,115.4,115.2,77.9,74.4,72.7,71.9,43.8,40.0,38.8; HRMS(ESI):Calcd for[M+Na]+C23H25NNaO8:466.1448,found 466.1484。
EXAMPLE 11 preparation of caffeoylquinic acid derivative 3f
(1) Preparation of intermediate 2
Accurately weighing 7.0g (19.8mmol) of 3-O-caffeoylquinic acid (compound 1) in a round-bottom flask with the thickness of 250m L, adding 60m L anhydrous acetone and 40m L DMP (2, 2-dimethoxypropane), stirring to react to form suspension, adding a catalytic amount of 50mg TsOH (p-toluenesulfonic acid) into the reaction liquid, stirring to react until the reaction liquid is clear, reacting at room temperature, monitoring by T L C, and after the reaction is finished, using anhydrous Na2CO3And neutralizing until the pH value is 5-6, performing suction filtration, performing reduced pressure evaporation to remove the solvent, and purifying the residue by silica gel column chromatography to obtain an intermediate 2 with the yield of 87%.
(2) Preparation of caffeoylquinic acid derivative 3f
In a 250m L round-bottom flask, exactly 21.2 g (3mmol) of intermediate and 1.5g (3mmol) of BOP are weighed, and 70m L of anhydrous mixed solvent THF/CH is added under the protection of argon3CN (v/v=3:2) stirring to dissolve, after adding 0.8g (6mmol) of DIEA, 3mmol of piperidine was added dropwise to the reaction solution until T L C monitoring the end of the reaction, the solvent was evaporated under reduced pressure and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 3f as a pale yellow powder in 64% yield.
1H-NMR(600MHz,DMSO):9.59(s,1H,Ph-OH),9.15(s,1H,Ph-OH),7.48 (d,J=15.8Hz,1H,H-7′),7.04(d,J=1.5Hz,1H,H-2′),7.00(dd,J=8.1Hz,1.5 Hz,1H,H-6’),6.77(d,J=8.0Hz,1H,H-5’),6.22(d,J=15.9Hz,1H,H-8′),5.62 (s,1H,H-1),5.32-5.29(m,1H,H-3),4.45-4.42(m,1H,H-4),4.08-4.06(m,1H, H-5),3.97-3.59(m,2H,NH-CH2),3.55-3.43(m,1H,NH-CH),3.35-3.17(m,1H, NH-CH),2.33-2.30(m,1H,H-2),2.01-1.98(m,1H,H-2),1.90-1.86(m,2H,H-6), 1.58-1.50(m,2H,CH2),1.49-1.37(m,2H,CH3-C-O,(CH2)2),1.25(m,3H,CH3- C-O);13C-NMR(150MHz,DMSO):171.5,165.8,148.5,145.6,145.4,125.5, 121.3,115.8,114.8,114.0,108.0,76.1,74.8,73.2,70.6,37.0,36.8,34.5,29.0,27.9, 25.8,25.5,24.2;HRMS(ESI):Calcd for[M+Na]+C24H31NNaO8:484.1947,found 484.1949。
EXAMPLE 12 preparation of caffeoylquinic acid derivative 4f
(3) Preparation of caffeoylquinic acid derivative 4f
In a 50m L round-bottomed flask, 2mmol of caffeoylquinic acid derivative 3f was accurately weighed out, and TFA/DCM/H was added2O (v/v ═ 9:1:1)20m L, the reaction was stirred at room temperature for 0.5 h until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give caffeoylquinic acid derivative 4f as a pale yellow powder with a yield of 61%.
1H-NMR(600MHz,MeOD):7.57(d,J=15.9Hz,1H,H-7′),7.05(d,J=1.9 Hz,1H,H-2′),6.95(dd,J=8.2Hz,2.0Hz,1H,H-6’),6.78(d,J=8.2Hz,1H,H- 5’),6.26(d,J=15.9Hz,1H,H-8′),5.36-5.33(m,1H,H-3),4.28-4.27(m,1H,H- 4),4.06-3.79(m,2H,NH-CH2),3.73-3.71(m,1H,H-5),3.63-3.38(m,2H,NH- CH2),2.33-2.01(m,4H,H-2,6),1.68-1.61(m,2H,CH2),1.60-1.48(m,4H, (CH2)2),1.90-1.86(m,2H,H-6),1.58-1.50(m,2H,CH2),1.49-1.37(m,2H, CH3-C-O,(CH2)2);13C-NMR(150MHz,MeOD):173.2,168.8,149.6,147.2, 146.8,127.8,123.0,116.5,115.3,115.2,78.6,73.3,72.4,71.6,40.0,39.2,38.7, 30.7,25.6;HRMS(ESI):Calcd for[M+Na]+C21H27NNaO8:444.1634,found 444.1637。
EXAMPLE 13 preparation of caffeoylquinic acid derivatives 3g
(1) Preparation of intermediate 2
Accurately weighing 7.0g (19.8mmol) of 3-O-caffeoylquinic acid (compound 1) in a round-bottom flask with the thickness of 250m L, adding 60m L anhydrous acetone and 40m L DMP (2, 2-dimethoxypropane), stirring to react to form suspension, adding a catalytic amount of 50mg TsOH (p-toluenesulfonic acid) into the reaction liquid, stirring to react until the reaction liquid is clear, reacting at room temperature, monitoring by T L C, and after the reaction is finished, using anhydrous Na2CO3And neutralizing until the pH value is 5-6, performing suction filtration, performing reduced pressure evaporation to remove the solvent, and purifying the residue by silica gel column chromatography to obtain an intermediate 2 with the yield of 87%.
(2) Preparation of caffeoylquinic acid derivative 3g
In a 250m L round-bottom flask, exactly 21.2 g (3mmol) of intermediate and 1.5g (3mmol) of BOP are weighed, and 70m L of anhydrous mixed solvent THF/CH is added under the protection of argon3CN (v/v ═ 3:2) was stirred until dissolved, 0.8g (6mmol) of DIEA was added, 3mmol of cyclohexylamine was added dropwise to the reaction solution until T L C monitored the end of the reaction, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give 3g of caffeoylquinic acid derivative as pale yellow powder with a yield of 71%.
1H-NMR(400MHz,DMSO):7.47(d,J=15.8Hz,1H,H-7′),7.38(d,J=8.2 Hz,1H,NH),7.04-6.99(m,2H,H-2′,6’),6.76(d,J=8.1Hz,1H,H-5’),6.24(d,J =15.8Hz,1H,H-8′),5.44(br,1H,H-1),5.37-5.30(m,1H,H-3),4.42-4.41(m,1H, H-4),4.14-4.11(m,1H,H-5),3.51-3.49(m,1H,NH-CH),2.25-2.20(m,1H,H-2), 1.97-1.91(m,1H,H-2),1.77-1.75(m,2H,H-6),1.67-1.65(m,4H,CH(CH2)2), 1.41(s,3H,CH3-C-O),1.26-1.10(m,9H,CH3-C-O,(CH2)3);13C-NMR(100 MHz,DMSO):174.1,165.8,148.4,145.5,145.3,125.5,121.3,115.7,114.8, 114.0,108.0,76.8,74.0,73.5,70.9,47.4,37.0,34.4,32.1,28.0,25.9,25.1,24.4; HRMS(ESI):Calcd for[M+Na]+C25H33NNaO8:498.2104,found 498.2106。
EXAMPLE 14 preparation of caffeoylquinic acid derivative 4g
(3) Preparation of caffeoylquinic acid derivative 4g
In a 50m L round-bottomed flask, 3g of 2mmol caffeoylquinic acid derivative was accurately weighed out, and TFA/DCM/H was added2O (v/v ═ 9:1:1)20m L, and the reaction was stirred at room temperature for 0.5 h until T L C monitored that the reaction was complete, the solvent was distilled off under reduced pressure, and the residue was purified by silica gel column chromatography to give 4g of caffeoylquinic acid derivative as a pale yellow powder with a yield of 64%.
1H-NMR(600MHz,MeOD):7.57(d,J=15.9Hz,1H,H-7′),7.05(d,J=2.0 Hz,1H,H-2′),6.95(dd,J=8.2Hz,2.0Hz,1H,H-6’),6.78(d,J=8.2Hz,1H,H- 5’),6.29(d,J=16.0Hz,1H,H-8′),5.42-5.36(m,1H,H-3),4.24-4.22(m,1H,H- 4),3.72-3.69(m,1H,H-5),3.65-3.58(m,1H,NH-CH),2.12-1.89(m,4H,H-2,6), 1.84-1.82(m,2H,CH2),1.77-1.73(m,2H,CH2),1.65-1.58(m,2H,CH2), 1.40-1.34(m,2H,CH2),1.26-1.23(m,2H,CH2);13C-NMR(150MHz,MeOD): 175.7,169.0,149.6,147.0,146.8,127.8,122.9,116.5,115.4,115.2,77.6,74.4, 72.7,72.0,49.6,40.0,38.7,33.6,30.7,26.5,26.1;HRMS(ESI):Calcd for[M+ Na]+C22H29NNaO8:458.1791,found 458.1795。
Test example 1 biological Activity test of Caffeoylquinic acid derivatives
1.1 cytology methods
The human liver HepG2 cell line was cultured in DMEM medium containing 10% fetal bovine serum and penicillin/streptomycin (100. mu.g/m L) at 37 ℃ in 5% CO2When the cells grow to 70-80% of the culture dish, stimulating the cells for 12h by using 100 mu mol/L oleic acid (M) and palmitic acid mixed liquor to prepare a lipid accumulation model, and then giving different caffeoylquinic acid derivativesBiological (10 mu mol/L) and chlorogenic acid (namely 3-O-Caffeoylquinic Acid) (CA) and simvastatin (S) positive control with the same concentration, a blank group (K) control is added, the positive control and the simvastatin (S) positive control are incubated for 24h, oil red O staining is used after the experiment is finished, the absorbance (A) value is measured at 358nm through a spectrophotometer, and effective compounds for inhibiting lipid accumulation in the caffeoylquinic acid compounds are observed, each experiment is repeated for 3 times, and according to three independent experiments, data are expressed as
Figure BDA0001941435260000171
The value is obtained.
1.2 results of lipid-lowering Activity
The lipid-lowering effect of chlorogenic acid and its derivatives on oleic acid-induced lipid accumulation in HepG2 hepatocytes was examined using an oil red O staining test, the results of oil red O staining indicate that some derivatives have a moderate to good modulating effect on oleic acid-induced lipid accumulation in HepG2 hepatocytes as shown in fig. 1, preliminary testing at 10 μmol/L showed that caffeoylquinic acid derivatives 3d, 3f, 3g, 4a and 4c more moderate lipid-lowering effect on HepG2 cells, treatment of HepG2 hepatocytes with caffeoylquinic acid derivatives 3d, 3g, 4a and 4c had better modulating effect than Chlorogenic Acid (CA), after treatment with 10 μmol/L caffeoylquinic acid derivatives 3d, 3f and 3g in fig. 1A, respectively, the absorbance was reduced from 0.261 (treated with oleic acid alone) to 0.249,0.255 and 0.249, and 0.249 in fig. 1B, after treatment with 10 μmol/L caffeoylquinic derivatives 3d, 3f and 3g, respectively, to 0.267 c, to 0.271, where the potential modulating effect of the lipid-accumulation of the derivatives was found to 0.271, 3d, 3c, 3d, 3c, and 267 c, 3c, respectively, to show a, as strong modulating effects after treatment with oleic acid derivatives, to 0.271, which were studied.
1.3 Structure-activity relationship analysis
Preliminary structure-activity relationship (SARs) analysis showed that amide derivatives of chlorogenic acid with ketal-substituted 4,5 hydroxyl groups were slightly less active than the amide derivatives with naked 4,5 hydroxyl groups compared to simvastatin (S) and Chlorogenic Acid (CA) positive controls. In addition, the 3d, 4d, 3f, 4f, 3g and 4g derivatives containing propargylamine, piperidyl and cyclohexylamine groups can better regulate lipid accumulation after oleic acid treatment, and show that the introduction of propargylamine, piperidyl and cyclohexylamine groups can improve lipid accumulation. Among them, derivatives 3d and 4d showed stronger lipid-lowering effect compared to other compounds, which means that propargylamine group is a favorable substituent for regulating lipid accumulation in HepG2 hepatocytes. However, the derivatives 3b and 4b (including the isobutylamino group) were less active, indicating that the introduction of the isobutylamino group was not favorable for the derivatives to exert lipid-lowering activity. In addition, the above derivatives show lipid-lowering effect in fig. 1, which shows that the 1-position carboxyl group and the 4, 5-position hydroxyl groups of chlorogenic acid are not essential groups for regulating lipid effect, and the influence of different amino substituents on the 1-position carboxyl group on lipid-lowering activity is more obvious.
Those of ordinary skill in the art will understand that: the discussion of any embodiment above is meant to be exemplary only, and is not intended to intimate that the scope of the disclosure, including the claims, is limited to these examples; within the inventive idea, also technical features in the above embodiments or in different embodiments may be combined and there are many other variations of the different aspects of the invention as described above, which are not provided in detail for the sake of brevity. Therefore, any omissions, modifications, substitutions, improvements and the like that may be made without departing from the spirit and principles of the invention are intended to be included within the scope of the invention.

Claims (10)

1. A caffeoylquinic acid derivative, which is characterized by having a structure shown in a general formula (I):
Figure FDA0001941435250000011
in the formula: r6Is an H atom;
R4and R5Is an H atom;
R2and R3Is H atom or propylidene;
R1selected from 1-18 carbonsAlkyl amines, aryl amines having 6 to 10 carbons or saturated or unsaturated cyclic amines having 3 to 7 carbon atoms.
2. The caffeoylquinic acid derivative of claim 1, wherein R is1Selected from alkylamines containing 3-8 carbons, arylamines containing 7-8 carbons or saturated cyclic amines containing 5-6 carbons.
3. The caffeoylquinic acid derivative of claim 2, wherein R is1Selected from n-butylamino, isobutylamino, n-octylamino, propargylamino, benzylamino, piperidinyl or cyclohexylamino.
4. A process for the preparation of caffeoylquinic acid derivatives, as claimed in any one of claims 1-3, comprising the steps of:
(1) reacting 3-O-caffeoylquinic acid with 2, 2-dimethoxypropane to generate an intermediate;
(2) and carrying out condensation reaction on the intermediate and amine to generate a target compound caffeoylquinic acid derivative.
5. The process for producing a caffeoylquinic acid derivative according to claim 4, which further comprises, after the step (2):
(3): carrying out deprotection reaction on the target compound caffeoylquinic acid derivative.
6. The process for producing a caffeoylquinic acid derivative according to claim 4, which comprises the steps of:
(1) reacting 3-O-caffeoylquinic acid and 2, 2-dimethoxypropane in anhydrous acetone by taking p-toluenesulfonic acid as a catalyst to generate an intermediate shown in a formula (II);
Figure FDA0001941435250000021
(2) the intermediate shown in the formula (II) is subjected to condensation reaction with amine in a mixed solvent of anhydrous acetonitrile and tetrahydrofuran under the condensation condition of BOP and DIEA to generate the target compound caffeoylquinic acid derivative.
7. The process for producing a caffeoylquinic acid derivative according to claim 6, which further comprises, after the step (2):
(3): removing acetonylidene protection from a target compound caffeoylquinic acid derivative in a mixed solution of trifluoroacetic acid, dichloromethane and water.
8. A pharmaceutical composition comprising the caffeoylquinic acid derivative according to any one of claims 1-3 and a pharmaceutically acceptable carrier.
9. Use of the caffeoylquinic acid derivatives of any one of claims 1-3 in the preparation of a medicament for the prevention and/or treatment of hyperlipidemia.
10. Use of a caffeoylquinic acid derivative as defined in any of claims 1-3 in the preparation of a medicament for the prevention and/or treatment of atherosclerosis.
CN201910022953.0A 2019-01-10 2019-01-10 Caffeoylquinic acid derivative and preparation method and application thereof Active CN111423407B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910022953.0A CN111423407B (en) 2019-01-10 2019-01-10 Caffeoylquinic acid derivative and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910022953.0A CN111423407B (en) 2019-01-10 2019-01-10 Caffeoylquinic acid derivative and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN111423407A true CN111423407A (en) 2020-07-17
CN111423407B CN111423407B (en) 2021-06-08

Family

ID=71546056

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910022953.0A Active CN111423407B (en) 2019-01-10 2019-01-10 Caffeoylquinic acid derivative and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN111423407B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112341355A (en) * 2020-11-09 2021-02-09 中国医学科学院药用植物研究所 Chlorogenic acid derivative and preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101284799A (en) * 2007-03-23 2008-10-15 浙江医药股份有限公司新昌制药厂 Nitrogen-contained derivates of caffeoylquinic acid, method for preparing same, pharmaceutical compositions thereof and uses
CN105130838A (en) * 2015-07-15 2015-12-09 遵义医学院 Chlorogenic acid amide derivative, synthesis and application thereof
WO2017186779A1 (en) * 2016-04-26 2017-11-02 Plant Advanced Technologies Pat Amide derivatives of polycaffeoylquinic acids, process for producing same and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101284799A (en) * 2007-03-23 2008-10-15 浙江医药股份有限公司新昌制药厂 Nitrogen-contained derivates of caffeoylquinic acid, method for preparing same, pharmaceutical compositions thereof and uses
CN105130838A (en) * 2015-07-15 2015-12-09 遵义医学院 Chlorogenic acid amide derivative, synthesis and application thereof
WO2017186779A1 (en) * 2016-04-26 2017-11-02 Plant Advanced Technologies Pat Amide derivatives of polycaffeoylquinic acids, process for producing same and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
吴崇明 等: "露兜簕来源咖啡酰奎宁酸抑制HepG2 肝细胞脂质积聚并调节脂代谢相关基因的表达", 《药学学报》 *
田瑜 等,: "露兜簕单体化合物结构修饰及生物活性研究", 《中草药》 *
田瑜 等: "绿原酸酰胺二聚体的合成及体外抗脂质代谢紊乱活性研究", 《现代药物与临床》 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112341355A (en) * 2020-11-09 2021-02-09 中国医学科学院药用植物研究所 Chlorogenic acid derivative and preparation method and application thereof

Also Published As

Publication number Publication date
CN111423407B (en) 2021-06-08

Similar Documents

Publication Publication Date Title
ITOKAWA et al. New antitumor principles, casearins AF, for Casearia sylvestris Sw.(Flacourtiaceae)
US6017892A (en) Method for accelerating marrow cell proliferation
EP0609437A1 (en) Novel sphingoglycolipid and use thereof
CN109464675B (en) Preparation method and application of triptolide-carboxylated chitosan coupling drug
AU2012276114A1 (en) Substituted cinnamamide derivative, preparation method and use thereof
CN111423407B (en) Caffeoylquinic acid derivative and preparation method and application thereof
EP0315113A2 (en) Inner esters of gangliosides with analgesic-antiinflammatory activity
CN113336765B (en) Curcumenol esterified product, preparation method and application of curcumenol esterified product in medicine for treating colorectal cancer
CN111777758B (en) Water-soluble azelaic acid polyethylene glycol monoester and synthesis method and application thereof
CN103494806B (en) Application of benzene a pair of horses going side by side alpha-pyrone compound and preparation method thereof
TW201335177A (en) Sterol derivatives and preparation method and use thereof
CN108840899A (en) A kind of pentacyclic triterpenoid and its derivative and application
JP5903166B2 (en) Derivatives of butylphthalide and methods for producing and using the same
DE2512609A1 (en) PHARMACEUTICAL PREPARATION
AU2022282314B2 (en) Guaiane sesquiterpene derivatives and pharmaceutical use thereof
CN114292241B (en) Cannabidiol-2-dioxopiperazinoate and application thereof
EP0041235A2 (en) Alpha,beta-dihydropolyprenyl derivatives for treating liver diseases
CN112939910B (en) Santamarine derivative, pharmaceutical composition thereof, preparation method and application thereof
CN109096133B (en) 3-amino-4- (2,4, 5-trifluorophenyl) menthyl butyrate hydrochloride and preparation method and application thereof
CN116715706B (en) Chemical modification method for prolonging in-vivo half-life of theanine
CN111825608A (en) Tetrahydroquinoline and tetrahydroisoquinoline compounds and application thereof
CN115403571B (en) Arctigenin derivative, preparation method and application
CN104974116B (en) Vibralactone amide derivatives and preparation method and application
CN114456190B (en) Kashan alkane diterpenoid phaginin A derivative, pharmaceutical composition and application thereof
CN110128498B (en) Diosgenin derivative, pharmaceutical composition thereof, preparation method 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