CN114105807B - Hydroxycyclohexanamide compound and preparation method and application thereof - Google Patents

Hydroxycyclohexanamide compound and preparation method and application thereof Download PDF

Info

Publication number
CN114105807B
CN114105807B CN202111384289.8A CN202111384289A CN114105807B CN 114105807 B CN114105807 B CN 114105807B CN 202111384289 A CN202111384289 A CN 202111384289A CN 114105807 B CN114105807 B CN 114105807B
Authority
CN
China
Prior art keywords
benzyl ether
reacting
synthesis
dissolving
cooh
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202111384289.8A
Other languages
Chinese (zh)
Other versions
CN114105807A (en
Inventor
陈河如
李药兰
唐维
饶舒文
刘志军
朱皓月
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.)
Jinan University
Original Assignee
Jinan University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jinan University filed Critical Jinan University
Priority to CN202111384289.8A priority Critical patent/CN114105807B/en
Publication of CN114105807A publication Critical patent/CN114105807A/en
Application granted granted Critical
Publication of CN114105807B publication Critical patent/CN114105807B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/16Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms
    • C07C233/23Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a ring other than a six-membered aromatic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/166Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the carbon of a carboxamide group directly attached to the aromatic ring, e.g. procainamide, procarbazine, metoclopramide, labetalol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C213/00Preparation of compounds containing amino and hydroxy, amino and etherified hydroxy or amino and esterified hydroxy groups bound to the same carbon skeleton
    • C07C213/02Preparation of compounds containing amino and hydroxy, amino and etherified hydroxy or amino and esterified hydroxy groups bound to the same carbon skeleton by reactions involving the formation of amino groups from compounds containing hydroxy groups or etherified or esterified hydroxy groups
    • 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
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/32Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • C07C235/36Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton containing six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a ring other than a six-membered aromatic ring
    • 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
    • C07C235/42Carboxylic 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 six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/44Carboxylic 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 six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C235/54Carboxylic 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 six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a ring other than a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C247/00Compounds containing azido groups
    • C07C247/14Compounds containing azido groups with azido groups bound to carbon atoms of rings other than six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C29/00Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom not belonging to a six-membered aromatic ring
    • C07C29/132Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom not belonging to a six-membered aromatic ring by reduction of an oxygen containing functional group
    • C07C29/136Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom not belonging to a six-membered aromatic ring by reduction of an oxygen containing functional group of >C=O containing groups, e.g. —COOH
    • C07C29/143Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom not belonging to a six-membered aromatic ring by reduction of an oxygen containing functional group of >C=O containing groups, e.g. —COOH of ketones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C303/00Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides
    • C07C303/26Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides of esters of sulfonic acids
    • C07C303/28Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides of esters of sulfonic acids by reaction of hydroxy compounds with sulfonic acids or derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C303/00Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides
    • C07C303/26Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides of esters of sulfonic acids
    • C07C303/30Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides of esters of sulfonic acids by reactions not involving the formation of esterified sulfo groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C41/00Preparation of ethers; Preparation of compounds having groups, groups or groups
    • C07C41/01Preparation of ethers
    • C07C41/16Preparation of ethers by reaction of esters of mineral or organic acids with hydroxy or O-metal groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C41/00Preparation of ethers; Preparation of compounds having groups, groups or groups
    • C07C41/01Preparation of ethers
    • C07C41/18Preparation of ethers by reactions not forming ether-oxygen bonds
    • C07C41/26Preparation of ethers by reactions not forming ether-oxygen bonds by introduction of hydroxy or O-metal groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • 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
    • 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
    • C07C2601/16Systems containing only non-condensed rings with a six-membered ring the ring being unsaturated
    • 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 a hydroxycyclohexanediylThe amine compound has a structure shown in a formula I, and the structure of an R group is shown in A1-A5. According to the principle of medicine core similarity and the principle of bioisostere, 1 alpha, 2 beta-O, O-dicaffeoyl cyclopenta-3 beta-alcohol which is a natural source is used as a lead, and the hydroxycyclohexanamide compound with antiviral effect is designed and synthesized. The synthesized hydroxycyclohexanamide compounds designed by the invention have strong anti-respiratory syncytial virus activity and low cytotoxicity, the selectivity index of the anti-RSV is higher than that of positive control drug ribavirin and natural lead 3,4-O, O-dicaffeoylquinic acid methyl ester, and partial compounds are better than 1 alpha, 2 beta-O, O-dicaffeoylcyclopenta-3 beta-alcohol. And simultaneously, the half life period is prolonged and the oxidation stability is improved.

Description

Hydroxycyclohexanamide compound and preparation method and application thereof
Technical Field
The invention belongs to the fields of chemical medicines and medicines for resisting viral infection, and particularly relates to a hydroxycyclohexanamide compound as well as a preparation method and application thereof.
Background
Human respiratory syncytial virus (respiratory syncytial virus, RSV) is a enveloped, non-segmented, single-stranded negative-strand RNA virus of the genus pneumovirus of the family Paramyxoviridae, which is the major causative agent of bronchiolitis and pneumonia in infants worldwide, and infants less than 6 months are at the highest risk of infection with RSV, and cannot obtain life-long immunity after infection with RSV, therefore, RSV can also cause severe respiratory diseases in the elderly and immunodeficiency people, the major symptoms of which are fever, cough, rhinitis and dyspnea.
At present, there is no clinically safe RSV vaccine and small molecule medicine for specific treatment of RSV infection. Humanized monoclonal antibodies and ribavirin (ribavirin) are recognized therapeutic agents with a certain therapeutic effect on RSV, the latter being mainly achieved by inhibiting the activity of host cell inosine phosphate dehydrogenase (IMP dehydrogenase) to inhibit viral replication, so that the humanized monoclonal antibodies and ribavirin have higher toxicity to host cells; both have certain problems in the aspects of curative effect, cost, safety, usability and the like of aerosol administration, and limit the clinical application of the aerosol. Therefore, the development of the high-efficiency and low-toxicity specific anti-RSV small-molecule drug has important clinical significance.
The herba Hedyotidis Diffusae (Elephantopus scaber) has antiviral, antibacterial, antitumor, and antipyretic effects. Chinese patent No. CN102219687B discloses 1 a, 2 β -O, O-dicaffeoylcyclopent-3 β -ol extracted from elephantopus scaber, which has anti-RSV viral activity beliba Wei Linjiang and toxicity to host cells beliba Wei Linxiao. But 1 alpha, 2 beta-O, O-dicaffeoyl cyclopent-3 beta-alcohol is unstable in structure, is easy to hydrolyze in blood, has short half-life and directly influences the clinical application of the same.
The Chinese patent No. CN110156628A partially solves the problem of instability of 1 alpha, 2 beta-O, O-dicaffeoyl cyclopenta-3 beta-ol, but still has the problem that candidate compounds are easy to oxidize.
Disclosure of Invention
The invention aims to provide a hydroxycyclohexanamide compound, a preparation method and application thereof, wherein the compound reduces cytotoxicity, improves the inhibition activity on Respiratory Syncytial Virus (RSV), prolongs the half-life period and improves the oxidation stability.
The aim of the invention is achieved by the following technical scheme:
a hydroxycyclohexanamide compound having a structure as shown in formula I:
in formula I, the structure of the R group is as follows:
* Representing either the R configuration or the S configuration.
The hydroxycyclohexanediamide compounds are preferably compounds CL-A1-1 to CL-A1-8, CL-A2-1 to CL-A2-8, CL-A3-1 to CL-A3-8, CL-A4-1 to CL-A4-8 and CL-A5-1 to CL-A5-8, and the structures are as follows:
the hydroxycyclohexanamide compounds are more preferably compounds CL-A3-1 to CL-A3-8 and CL-A5-1 to CL-A5-8.
The preparation method of the hydroxycyclohexanediamide compound comprises the following steps:
(1) Synthesis of carboxylic acids comprising the A1-A5 Structure
The chemical structure of the carboxylic acid comprising the A1-A5 structure is shown below:
wherein A1-COOH and A2-COOH are purchased from commercial sources; A3-COOH and A4-COOH can be purchased commercially, and can be synthesized as in A5-COOH by the following steps.
(1.1) Synthesis of A3-COOH and A4-COOH
2-bromo-4-fluorobenzaldehyde or 4-nitrobenzaldehyde is taken as a starting material, and is dissolved in pyridine with maleic acid, and organic alkali is added for reaction to obtain A3-COOH or A4-COOH;
the molar ratio of 2-bromo-4-fluorobenzaldehyde or 4-nitrobenzaldehyde to maleic acid in step (1.1) is 1.0:1.0 to 3.0, preferably 1.0:2.5.
In step (1.1) a catalyst may be added, said catalyst being a tetrahydropyrrole or a piperidine, preferably a piperidine.
The dosage of the catalyst in the step (1.1) is mixed with the molar ratio of the catalyst to the 2-bromo-4-fluorobenzaldehyde or the 4-nitrobenzaldehyde is 0.1-0.3:1.0; preferably 0.15:1.0.
The reaction temperature in the step (1.1) is 100-150 ℃; preferably 110 ℃.
The reaction time in the step (1.1) is 10-20 h; preferably 15h.
(1.2) Synthesis of A5-COOH
Synthesizing 4-hydroxy phenyl acrylic acid according to the step (1.1); the method comprises the following steps: dissolving 4-hydroxybenzaldehyde serving as a starting material and maleic acid in pyridine, adding organic base, and reacting to obtain 4-hydroxyphenyl acrylic acid;
dissolving 4-hydroxy phenyl acrylic acid, formaldehyde and an acid medium in paraxylene, and heating for reaction to obtain A5-COOH.
The molar ratio of 4-hydroxy phenyl acrylic acid to formaldehyde in the step (1.2) is 1.0:2.0-10.0, preferably 1.0:9.0.
The acid medium in the step (1.2) is boric acid, 5-35% hydrochloric acid solution or 5-35% sulfuric acid solution; boric acid is preferred.
The dosage of the acid medium in the step (1.2) is mixed with the molar ratio of the acid medium to the 4-hydroxy phenyl acrylic acid to be 1.0-1.0:5.0; preferably 1.0:3.0 ratio.
The reaction temperature in the step (1.2) is 100-160 ℃; preferably 150 ℃.
The reaction time in the step (1.2) is 8.0-30 h; preferably 12h.
(2) Synthesis of 2, 3-diaminocyclohexanol
Comprises the following steps of
(2.1) Synthesis of 2-cyclohexen-1-ol
2-cyclohexene-1-one is taken as a raw material, dissolved in methanol, added with cerium trichloride, reacted, added with sodium borohydride in batches, and reacted to obtain 2-cyclohexene-1-ol;
the dosage of the cerium trichloride, the sodium borohydride and the 2-cyclohexene-1-ketone in the step (2.1) is calculated according to the molar ratio of 1-3:1; preferably in a molar ratio of 1.3:1.3:1.
In the step (2.1), the reaction after adding the cerium trichloride is carried out for 0.3 to 3 hours at the temperature of-30 to 0 ℃; preferably at-15 deg.c for 0.5 hr; the reaction after adding the sodium borohydride in batches is carried out for 1 to 5 hours at the temperature of-30 to 0 ℃; preferably at-15 deg.c for 3 hr.
The sodium borohydride is added in portions as described in step (2.1), preferably in three portions within 15 minutes.
(2.2) Synthesis of 2-cyclohexene-1-benzyl ether
Dissolving the 2-cyclohexene-1-alcohol obtained in the step (2.1) in redistilled tetrahydrofuran, adding tetrabutylammonium bromide, balancing, adding sodium hydride, reacting, dropwise adding benzyl bromide, reacting, moving to room temperature, and stirring for reacting to obtain 2-cyclohexene-1-benzyl ether;
the molar ratio of tetrabutylammonium bromide to sodium hydride, benzyl bromide and 2-cyclohexene-1-ol in the step (2.2) is 1:20-60:6-20:6-20; preferably in a molar ratio of 1:40:12:8.
In the step (2.2), the reaction after adding sodium hydride is carried out for 5-30 min at the temperature of-20-0 ℃, preferably for 10min at the temperature of-5 ℃; the reaction after the benzyl bromide is dripped is carried out for 20 to 50min at the temperature of-20 to 0 ℃, preferably for 30min at the temperature of-5 ℃; the reaction time of stirring after the reaction is moved to room temperature is 3 to 6 hours, preferably 5 hours.
In step (2.2), the sodium hydride is added in multiple portions, preferably three portions for 10 minutes.
(2.3) Synthesis of 2, 3-dihydroxycyclohexyl-1-benzyl ether
Dissolving the 2-cyclohexene-1-benzyl ether obtained in the step (2.2) in acetonitrile solution, adding an oxidant after balancing, reacting under ice bath condition, moving to room temperature, and stirring for reacting to obtain 2, 3-dihydroxycyclohexyl-1-benzyl ether;
the acetonitrile solution in the step (2.3) is obtained by mixing acetonitrile and water according to the volume ratio of 1.0-5.0:1.0, preferably 3.0:1.0.
The oxidant in the step (2.3) is at least one of potassium osmium oxide/N-methylmorpholine oxide/potassium carbonate, peracetic acid, hydrogen peroxide and m-chloroperbenzoic acid, preferably potassium osmium oxide/N-methylmorpholine oxide/potassium carbonate.
When the oxidant is potassium osmium oxide/N-methylmorpholine oxide/potassium carbonate, the dosage of the N-methylmorpholine oxide, the potassium carbonate and the 2-cyclohexene-1-benzyl ether is calculated according to the molar ratio of 0.5-2.0:0.5-2.0:1; preferably calculated according to the molar ratio of 1.3:1.3:1; the potassium osmium is used in a catalytic amount, preferably 10% of the molar amount of 2-cyclohexene-1-benzyl ether.
The reaction time under the ice bath condition in the step (2.3) is 10-60 min; preferably 40min.
The reaction time of stirring after moving to room temperature in the step (2.3) is 8-30 h; preferably 12h.
(2.4) Synthesis of 3-Benzyloxycyclohexyl-1, 2-dimesylate
Dissolving the 2, 3-dihydroxycyclohexyl-1-benzyl ether obtained in the step (2.3) in redistilled methylene dichloride, adding 4- (N, N-dimethyl) aminopyridine, adding triethylamine under ice bath condition, dripping a sulfonylating reagent after balancing, reacting, stirring at room temperature, and reacting to obtain 3-benzyloxycyclohexyl-1, 2-dimethyl sulfonate;
the sulfonylating agent in the step (2.4) is methanesulfonyl chloride or p-toluenesulfonyl chloride, preferably methanesulfonyl chloride.
The dosage of the sulfonylation reagent in the step (2.4) is calculated according to the molar ratio of the sulfonylation reagent to the 2, 3-dihydroxycyclohexane-1-benzyl ether of 1.0-5.0:1.0; preferably in a molar ratio of 3.0:1.0 to the 2, 3-dihydroxycyclohexyl-1-benzyl ether.
In the step (2.4), the reaction time after the sulfonyl reagent is dripped is 20-60 min; preferably 40min; stirring and reacting for 5-24 h at room temperature; preferably 12h.
(2.5) Synthesis of 2, 3-diazidocyclohexane-1-benzyl ether
Dissolving 3-benzyloxy-1, 2-dimethyl sulfonate obtained in the step (2.4) in a mixed solvent of N, N-Dimethylformamide (DMF) and hexamethylphosphoric triamide (HMPA), adding sodium azide, and reacting to obtain 1-benzyloxy-3-azidocyclohexyl-2-methane sulfonate;
Dissolving the obtained 1-benzyloxy-3-azidocyclohexyl-2-methanesulfonate in a mixed solvent of N, N-Dimethylformamide (DMF) and hexamethylphosphoric triamide (HMPA), adding sodium azide, and reacting to obtain the 2, 3-diazacyclhexyl-1-benzyl ether.
The mixed solvent of the N, N-dimethylformamide and the hexamethylphosphoric triamide in the step (2.5) is obtained by proportioning the N, N-dimethylformamide and the hexamethylphosphoric triamide according to the volume ratio of 3:1.
In the step (2.5), the dosage of the sodium azide added for the first time is calculated according to the molar ratio of the sodium azide to the 3-benzyloxy cyclohexyl-1, 2-dimethyl sulfonate of 1-5:1; preferably calculated according to the molar ratio of 3:1 of 3-benzyloxy cyclohexyl-1, 2-dimethyl sulfonate; the dosage of the sodium azide added for the second time is calculated according to the molar ratio of the sodium azide to the 1-benzyloxy-3-azidocyclohexyl-2-methane sulfonate of 2-8:1; preferably calculated according to the molar ratio of the 1-benzyloxy-3-azidocyclohexyl-2-methanesulfonate to the 1:1 ratio.
In the step (2.5), the first reaction is carried out for 5 to 20 hours under the oil bath condition of 50 to 100 ℃; preferably at 75 ℃ for 12 hours in an oil bath; the second reaction is carried out for 10-30 h under the oil bath condition of 100-150 ℃; preferably at 125℃in an oil bath.
(2.6) Synthesis of 2, 3-diaminocyclohexane-1-benzyl ether
Dissolving the 2, 3-diaza-cyclohexane-1-benzyl ether obtained in the step (2.5) in tetrahydrofuran, adding a reducing agent, and reacting to obtain a residue containing the 2, 3-diaminocyclohexane-1-benzyl ether;
the methanol solution in the step (2.6) is prepared by mixing methanol and water according to a volume ratio of 5:1.
The reducing agent in the step (2.6) is triphenylphosphine or LiAlH 4 Preferably LiAlH 4 . The dosage is calculated according to the mol ratio of the 2, 3-diazidocyclohexane-1-benzyl ether to the mixture ratio of 0.5-3:1; preferably in a molar ratio of 1.2:1 with the 2, 3-diazabicyclo-1-benzyl ether.
The reaction in the step (2.6) is carried out under stirring at-15 ℃ for 5 minutes, and 2.5M LiAlH is slowly added dropwise 4 The solution was stirred for 30min after dropping, and the reaction was continued with stirring for 10h after warming to room temperature.
(3) Synthesis of hydroxycyclohexanamide compounds
Comprises the following steps of
(3.1) dissolving A1-COOH, A2-COOH, A3-COOH, A4-COOH and A5-COOH in redistilled N, N-dimethylformamide, adding a condensing agent and 1-hydroxybenzotriazole (HOBt), reacting, adding N, N-Diisopropylethylamine (DIPEA), dissolving 2, 3-diaminocyclohexane-1-benzyl ether obtained in the step 2.6 in N, N-dimethylformamide, and then dropwise adding into a reaction system to react to obtain an optical isomer mixture of 3-benzyloxycyclohexane-1, 2-diamide;
The condensing agent in the step (3.1) is one of N, N' -Diisopropylcarbodiimide (DIC), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (EDCI) and ethyl chloroformate; preferably 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (EDCI).
The dosage of the condensing agent in the step (3.1) is calculated according to the molar ratio of the condensing agent to the An-COOH (n=1-5) of 1.0-3.0:1.0; preferably calculated as a molar ratio to An-COOH (n=1 to 5) of 1.2:1.
The amount of HOBt in the step (3.1) is calculated according to the molar ratio of the HOBt to the An-COOH (n=1-5) of 1.0-3.0:1.0; preferably calculated as a molar ratio to An-COOH (n=1 to 5) of 1.2:1.
The amount of the DIPEA in the step (3.1) is calculated according to the molar ratio of the DIPEA to the An-COOH (n=1-5) of 1.0-3.0:1.0; preferably calculated as a molar ratio to An-COOH (n=1 to 5) of 2.2:1.
The reaction time for adding the condensing agent and HOBt in the step (3.1) is 0.5 to 1.0 hours, preferably 0.5 hours.
The dosage of the 2, 3-diaminocyclohexane-1-benzyl ether in the step (3.1) is calculated according to the molar ratio of the 2, 3-diaminocyclohexane-1-benzyl ether to the An-COOH (n=1 to 5) of 0.2 to 1.0:1.0; preferably calculated as a molar ratio of 0.4:1 to the An-COOH (n=1 to 5).
The reaction time after the 2, 3-diaminocyclohexane-1-benzyl ether is added in the step (3.1) is 5 to 20 hours; preferably 12h.
(3.2) dissolving the optical isomer mixture of 3-benzyloxycyclohexyl-1, 2-diamide obtained in the step (3.1) in redistilled methylene dichloride, dropwise adding a deprotection agent after balancing, reacting, moving to room temperature, stirring and reacting to obtain optical isomer mixtures CL-A1-CLC-A5 of 3-hydroxycyclohexyl-1, 2-diamide.
The step (3.2) also comprises the step of separating and purifying the eight optical isomers of the CL-A1 to CLC-A5 by one or at least two of column chromatography, a high performance liquid phase method and a recrystallization method.
The deprotection agent in the step (3.2) is one of BCl3, BBr3 and H2/Pd; preferably BCl3.
The amount of the deprotection agent in the step (3.2) is calculated according to the molar ratio of the deprotection agent to the optical isomer mixture of the 3-benzyloxy cyclohexyl-1, 2-diamide of 1-5:1; preferably calculated as a molar ratio to the mixture of optical isomers of said 3-benzyloxycyclohexyl-1, 2-diamide of 2.5:1.
In the step (3.2), the first reaction is carried out for 15-45 min at the temperature of minus 20-0 ℃; preferably at-10deg.C for 30min; the reaction time of stirring after the second time to room temperature is 1-5 h, preferably 2h.
An antiviral agent comprising the above hydroxycyclohexanamide compound;
the virus is Respiratory Syncytial Virus (RSV);
the antiviral drug can also contain one or at least two pharmaceutically acceptable carriers or auxiliary materials.
The auxiliary materials are preferably sustained release agents, excipients, fillers, adhesives, wetting agents, disintegrating agents, absorption promoters, adsorption carriers, surfactants or lubricants and the like.
The carrier is at least one of microcapsule, microsphere, nanoparticle and liposome.
The antiviral medicament can be further prepared into various dosage forms, and the medicaments in various dosage forms can be prepared according to a conventional method in the pharmaceutical field.
Compared with the prior art, the invention has the following advantages and effects:
(1) According to the principle of medicine core similarity and the principle of bioisostere, 1 alpha, 2 beta-O, O-dicaffeoyl cyclopenta-3 beta-alcohol which is a natural source is used as a lead, and the hydroxycyclohexanamide compound with antiviral effect is designed and synthesized. In vitro antiviral activity of the series of hydroxycyclohexanamide compounds is detected by using a plaque reduction experiment, and cytotoxicity of the series of hydroxycyclohexanamide compounds is respectively estimated by using an MMT method. The results show that the hydroxycyclohexanamide compounds designed and synthesized by the invention have strong anti-Respiratory Syncytial Virus (RSV) activity and low cytotoxicity, and the selectivity index (SI value, SI=CC) of the anti-RSV 50 /IC 50 ) Compared with the positive control drug ribavirin and natural lead 3,4-O, O-dicaffeoylquinic acid methyl ester, partial compounds are better than 1 alpha, 2 beta-O, O-dicaffeoylcyclopenta-3 beta-alcohol.
(2) The hydroxycyclohexanamide compound has higher stability in fetal bovine serum, and the stability in fetal bovine serum is higher than that of a natural lead, namely 3,4-O, O-dicaffeoylquinic acid methyl ester and 1 alpha, 2 beta-O, O-dicaffeoylcyclopent-3 beta-alcohol.
(3) The anti-oxidation stability of the hydroxycyclohexanamide compounds of the invention is higher than that of the natural precursors 3,4-O, O-dicaffeoylquinic acid methyl ester and 1 alpha, 2 beta-O, O-dicaffeoylcyclopenta-3 beta-ol, and (1S, 2S, 3S) -2-hydroxycyclohexa-1, 3-dicaffeamide (CLC-H15).
(4) The hydroxycyclohexanamide compounds of the invention bind RSV fusion proteins with a nucleophilicity higher than the natural precursors 3,4-O, O-dicaffeoylquinic acid methyl ester and 1 a, 2 β -O, O-dicaffeoylcyclopenta-3 β -ol, but comparable to (1 s,2s,3 s) -2-hydroxycyclohexa-1, 3-dicaffeamide.
(5) The preparation method of the hydroxycyclohexanediamide compound has mild reaction conditions, is simple and safe to operate, and is easy to realize industrial production.
Detailed Description
The present invention will be described in further detail with reference to examples, but embodiments of the present invention are not limited thereto.
Example 1.
Preparation, separation and purification of 3-hydroxycyclohexyl-1, 2-di (beta-naphthyl) acetamide (CL-A1-CL-A1-8)
(1) Preparation of beta-naphthylacetic acid: purchased through conventional market channels.
(2) Preparation of 2, 3-diaminocyclohexane-1-benzyl ether
2.1.2 preparation of cyclohexen-1-ol
2-cyclohexen-1-one (0.49 mL,5 mmol) was weighed into a dry 50mL round bottom flask, dissolved with an appropriate amount of dry methanol, placed in a reaction bath stirred at low temperature and constant temperature, cerium trichloride (1.60 g,6.5 mmol) was then weighed into the flask, stirred at-15℃for 0.5h, sodium borohydride (245.92 mg,6.5 mmol) was slowly added 3 times over 15 min, and the reaction was maintained at-15℃for 3h. After the reaction, 3mL of saturated ammonium chloride solution was slowly added dropwise at-10℃to quench the reaction, the reaction mixture was then transferred to a separatory funnel, extracted with DCM (3X 50 mL), the organic phases were combined and washed with saturated brine (2X 100 mL), and the organic phase was taken up in anhydrous Na 2 SO 4 Drying, filtration, spin evaporation of solvent in vacuo, TLC developer conditions were EtOAc: PE=1:5 (V: V), R f The value was 0.34. Chromatography on silica gel (EtOAc: pe=1:10, v:v) afforded 439.2mg as a colorless oily liquid in 89.5% yield. 1 H NMR(300MHz,CDCl 3 )δ:5.96(m,1H),5.83(m,1H),4.84(m,1H),2.57-1.73(m,4H),1.71-1.53(m,2H); 13 C NMR(75MHz,CDCl 3 )δ:135.4,133.7,77.8,33.7,31.6,20.3。
Preparation of 2.2.2-cyclohexene-1-benzyl ether
2-cyclohexen-1-ol (392.6 mg,4.0 mmol) obtained in step 2.1 was weighed into a dry 100mL round bottom flask, dissolved with an appropriate amount of redistilled tetrahydrofuran, and tetrabutylammonium bromide (161 mg,0.5 mmol) was added, and the flask was then equilibrated for 5min in a low temperature reaction bath at-5 ℃. After equilibration, sodium hydride (480 mg,20 mmol) was slowly added to the reaction solution over 3 portions of 10 minutes, and the reaction was continued at-5℃for 10 minutes. Benzyl bromide (0.71 mL,6.0 mmol) was then slowly added dropwise to the reaction system, and after reaction at-5℃for 30min, the reaction mixture was allowed to warm to room temperature and stirring was continued for 5h. After the reaction is finished, under the ice bath conditionThe reaction was quenched by slowly dropping 5mL of methanol, then transferring the reaction solution to a separating funnel, diluting the reaction solution with an appropriate amount of saturated saline, extracting with DCM (3X 50 mL), combining the organic phases, washing with saturated saline (2X 100 mL), and washing the organic phase with anhydrous Na 2 SO 4 Drying, filtration, spin evaporation of solvent in vacuo, TLC developer conditions were EtOAc: PE=1:50 (V: V), R f The value was 0.42. The colorless oily liquid 653.0mg was obtained by silica gel column chromatography (pure petroleum ether) in 86.7% yield. 1 H NMR(300MHz,CDCl 3 )δ:7.57-7.46(m,5H,H-Ph),6.17(m,1H),6.08(m,1H),4.84(m,1H),4.68(s,2H,-CH 2 -Ph),2.73-1.95(m,4H),1.81-1.75(m,2H); 13 C NMR(75MHz,CDCl 3 )δ:138.8,135.5,130.6,128.4(2C),127.5(2C),127.3,84.3,70.1,30.8,29.6,20.4。
Preparation of 2.3.2,3-dihydroxycyclohexyl-1-benzyl ether
The 2-cyclohexene-1-benzyl ether from step 2.2 (564.8 mg,3.0 mmol) was weighed into a dry 100mL round bottom flask and mixed with an appropriate amount of acetonitrile and water (CH 3 CN:H 2 O=3:1, V:V) and the flask was equilibrated for 5min under ice bath conditions, then N-methylmorpholine oxide (458 mg,3.9 mmol) was slowly added followed by potassium osmium carbonate (99.7 mg,0.3 mmol) and potassium carbonate (539.5 mg,3.9 mmol) and the reaction was continued for 40min under ice bath conditions before moving to room temperature and stirring continued for 12h. After the reaction, the reaction mixture was transferred to a separatory funnel, diluted with an appropriate amount of saturated brine, extracted with DCM (3X 50 mL), the organic phases were combined, washed with saturated brine (2X 100 mL), and the organic phase was washed with anhydrous Na 2 SO 4 Drying, filtering, removing solvent by rotary evaporation under vacuum, and developing under the condition of CHCl as TLC developing agent 3 :MeOH=15:1(V:V),R f The value was 0.38. Separating by silica gel column chromatography (CHCl) 3 Meoh=30:1, v:v) to afford 583.5mg as a colorless oily liquid in 87.6% yield. 1 H NMR(300MHz,CDCl 3 )δ:7.42-7.28(m,5H,H-Ph),5.82(br,1H),4.57(s,2H,-CH 2 -Ph),4.33(br,1H),4.11(m,1H),3.95(m,2H),2.22-1.51(m,4H),1.51-1.43(m,2H); 13 C NMR(75MHz,CDCl 3 )δ:138.6,128.7(2C),127.9(2C),127.5,84.2,78.3,72.2,71.7,29.1,26.9,20.5。
Preparation of 2, 4, 3-Benzyloxycyclohexyl-1, 2-dimesylate
2, 3-Dihydroxycyclohexyl-1-benzyl ether (533.5 mg,2.4 mmol) obtained in step 2.3 was weighed into a dry 100mL round bottom flask, dissolved with an appropriate amount of redistilled dichloromethane, and 4- (N, N-dimethyl) aminopyridine (37 mg,0.3 mmol) was added, then the flask was placed under ice bath conditions, the acid-binding agent triethylamine (1 mL,7.2 mmol) was added, equilibrated for 5min, and then methanesulfonyl chloride (0.56 mL,7.2 mmol) was slowly added dropwise to the reaction solution, and after maintaining the ice bath conditions for 40min, the reaction was allowed to stand still at room temperature and stirring was continued for 12h. After the reaction, 5mL of pure water was slowly added dropwise under ice-bath conditions to quench the reaction, the reaction mixture was then transferred to a separatory funnel, diluted with an appropriate amount of saturated brine, extracted with DCM (3X 50 mL), the organic phases were combined and washed with saturated brine (2X 100 mL), and the organic phase was taken up in anhydrous Na 2 SO 4 Drying, filtration, spin evaporation of solvent in vacuo, TLC developer conditions were EtOAc: PE=1:2 (V: V), R f The value was 0.33. Chromatography on silica gel (EtOAc: pe=1:5, v: v) afforded 713.0mg as a pale yellow oily liquid in 78.5% yield. 1 H NMR(300MHz,CDCl 3 )δ:7.38-7.29(m,5H,H-Ph),5.08(q,J=6.0Hz,1H),4.86(t,J=6.0Hz,1H),4.55(s,2H,-CH 2 -Ph),4.18(m,1H),3.06(s,3H,-SO 2 CH 3 ),3.05(s,3H,-SO 2 CH 3 ),2.33-1.62(m,4H),1.51-1.42(m,2H); 13 C NMR(75MHz,CDCl 3 )δ:137.6,128.8(2C),128.1,127.8(2C),83.5,80.4,79.6,72.2,38.4(2C),27.2,25.8,20.5;ESI-MS(m/z):401.5[M+Na] +
Preparation of 2.5.2,3-diazidocyclohexyl-1-benzyl ether
3-Benzyloxycyclohexyl-1, 2-dimesylate (567.7 mg,1.5 mmol) obtained in step 2.4 was weighed into a dry 50mL round bottom flask, dissolved with a suitable amount of a mixed solvent of DMF and HMPA (DMF: HMPA=3:1, V: V), and sodium azide (293 mg,4.5 mmol) was added to react for 12h under an oil bath at 75 ℃. After the reaction, 5mL of pure water is slowly added dropwise under ice bath condition to quench the reaction, then the reaction solution is transferred to a separating funnel, a proper amount of saturated saline is added to dilute the reaction solution, DCM (3X 50 mL) is used for extraction,the organic phases were combined and washed with saturated brine (2X 100 mL), and the organic phase was washed with anhydrous Na 2 SO 4 Drying, filtration, spin evaporation in vacuo to remove low boiling solvent, TLC developing solvent conditions were EtOAc: PE=1:5 (V: V), R f The value was 0.35. The residue was purified by RP-HPLC with the eluent conditions of MeOH: H 2 O=70:30 (V: V), rt=21.3 min, yielding 412.4mg of yellow oil in 84.5% yield. 1 H NMR(300MHz,CDCl 3 )δ:7.37-7.25(m,5H,H-Ph),4.82(t,J=6.0Hz,1H),4.58(s,2H,-CH 2 -Ph),4.03(m,1H),3.87(m,1H),3.05(s,3H,-SO 2 CH 3 ),2.11-1.77(m,4H),1.53-1.44(m,2H); 13 C NMR(75MHz,CDCl 3 ) Delta 137.5,128.7 (2C), 128.3,128.2 (2C), 89.4,81.6,71.8,64.4,38.9,27.7,27.3,20.6. This is 1-benzyloxy-3-azidocyclohexyl-2-methanesulfonate.
1-benzyloxy-3-azidocyclohexane-2-mesylate (390.4 mg,1.2 mmol) was weighed into a dry 50mL round bottom flask, dissolved with a suitable amount of mixed solvent of DMF and HMPA (DMF: HMPA=3:1, V: V), and sodium azide (390 mg,6.0 mmol) was added and reacted under oil bath conditions at 125℃for 24h. After the reaction, 5mL of pure water was slowly added dropwise under ice-bath conditions to quench the reaction, the reaction mixture was then transferred to a separatory funnel, diluted with an appropriate amount of saturated brine, extracted with DCM (3X 50 mL), the organic phases were combined and washed with saturated brine (2X 100 mL), and the organic phase was taken up in anhydrous Na 2 SO 4 Drying, filtration, spin evaporation in vacuo to remove low boiling solvent, TLC developing solvent conditions were EtOAc: PE=1:8 (V: V), R f The value was 0.53. The residue was purified by RP-HPLC with the eluent conditions of MeOH: H 2 O=75:25 (V: V), rt=25.2 min, giving 248.0mg of a deep yellow oil, 75.9% yield. 1 H NMR(300MHz,CDCl 3 )δ:7.43-7.28(m,5H,H-Ph),4.57(s,2H,-CH 2 -Ph),3.98(m,1H),3.86(t,J=6.0Hz,1H),3.67(m,1H),2.03-1.86(m,4H),1.55-1.46(m,2H); 13 C NMR(75MHz,CDCl 3 )δ:137.7,128.5(2C),128.3,128.2(2C),89.4,81.6,71.8,64.2,38.7,27.9,27.3,20.5;ESI-MS(m/z):295.2[M+Na] + ,311.6[M+K] + This is 2, 3-diazabicyclo-1-benzyl ether.
2.6.2,3 preparation of diaminocyclohexane-1-benzyl ether.
2, 3-Diazidocyclohexyl-1-benzyl ether (272.3 mg,1.0 mmol) obtained in the step 2.5 is weighed, dissolved in tetrahydrofuran, protected by nitrogen, placed under the condition of-15 ℃ and stirred for 5min, a 2.5M lithium aluminum hydride tetrahydrofuran solution (0.48 mL,1.2 mmol) is slowly added dropwise, stirred for 30min, transferred to room temperature for reaction for 10h, transferred to an ice bath after the reaction is finished, stirred for 5min, slowly added dropwise with methanol until no bubbles are generated, the reaction solution is rotationally evaporated to remove the solvent, filtered, extracted by a dichloromethane and saturated saline system, pH is adjusted to 3 by adding 5mL hydrochloric acid in water, extracted by dichloromethane for 5 times, pH is adjusted to 8 by adding 10mg sodium hydroxide after extraction for 3 times, the organic phases are combined, and the solvent is removed by vacuum rotary evaporation. TLC detection as pure compound (TLC developing solvent condition V) DCM :V CH3OH =25:1,R f =0.46). The product was 178.5mg of red liquid in 81% yield. 1 H NMR(400MHz,DMSO-d 6 )δ7.35-7.23(m,5H),4.49(dd,J=27.5,12.0Hz,2H),3.31(d,J=10.2Hz,1H),3.14(s,1H),2.54-2.45(m,1H),2.29(s,4H),1.61-1.02(m,6H). 13 C NMR(75MHz,CD 3 OD)δ140.03,129.42,128.82,128.67,79.94,71.61,54.29,52.92,29.81,26.44,19.71.HR-ESI-MS(m/z):calcd for C 13 H 21 N 2 O 1 [M+H] + 221.1654,found:221.1664。
(3) Preparation, separation and purification of 3-hydroxycyclohexyl-1, 2-di (beta-naphthyl) acetamide (CL-A1-CL-A1-8)
3.1. Beta-naphthylacetic acid (372.4 mg,2.0 mmol) was weighed into a dry 50mL round bottom flask, dissolved with an appropriate amount of redistilled DMF, and EDCI (460 mg,2.4 mmol) and HOBt (324 mg,2.4 mmol) were slowly added, respectively, and reacted at room temperature for 30min, followed by DIPEA (0.77 mL,4.4 mmol), and 2, 3-diaminocyclohexyl-1-benzyl ether (180.7 mg,0.82 mmol) obtained by the reaction of step 2.6 was dissolved with dry DMF, then slowly added dropwise to the reaction system, and the reaction was continued at room temperature for 12h. After the reaction, 5mL of pure water was slowly added dropwise under ice bath conditions to quench the reaction, the reaction mixture was then transferred to a separatory funnel, diluted with an appropriate amount of saturated brine, extracted with DCM (3X 50 mL), and the organic phases were combined and then usedSaturated saline (2X 100 mL) and the organic phase was washed with anhydrous Na 2 SO 4 Drying, filtering, removing solvent by vacuum rotary evaporation, and developing under TLC developing agent condition V DCM :V CH3OH =25:1, silica gel column chromatography separation and purification, collection R f Fraction=0.61 to yield 356.1mg of red solid in 78% yield. 1 H NMR(400MHz,CDCl 3 )δ8.15(s,2H),7.95(d,J=8.4Hz,2H),7.89(d,J=8.4Hz,2H),7.81(d,J=8.4Hz,2H),7.67(s,2H),7.62-7.50(m,4H),7.38(d,J=3.2Hz,2H),7.29-7.26(m,5H),5.65(d,J=8.9Hz,1H),4.24(dd,J=8.6,3.8Hz,1H),3.98-3.88(m,1H),3.63-3.42(m,5H),3.25(d,J=15.8Hz,1H),2.90(d,J=19.7Hz,1H),1.70(d,J=12.4Hz,2H),1.50-1.40(m,1H),1.20(dd,J=18.4,7.3Hz,3H); 13 CNMR(101MHz,CDCl 3 )δ170.72,169.95,137.14,133.66,133.45,132.44(2),128.80,128.36,128.12-127.73(7),127.70-127.38(7),127.11,126.74,126.38(2),126.01,75.37,69.83,49.34,48.19,44.08,43.80,29.79,26.84,14.30.HR-ESI-MS(m/z):calcd for C 37 H 37 N 2 O 3 ([M+H] + ) 557.2804, found:557.2812. All data confirm that this material is a mixture of optical isomers of 3-benzyloxycyclohexyl-1, 2-bis (β -naphthyl) acetamide.
3.2. 3-Benzyloxycyclohexyl-1, 2-bis (. Beta. -naphthyl) acetamide (278.4 mg,0.5 mmol) was weighed into a dry 100mL round bottom flask, dissolved with an appropriate amount of distilled dichloromethane, and the flask was left to equilibrate for 5min at-10 ℃. After equilibration, a 1M DCM solution of boron trichloride (1.8 mL,1.8 mmol) was slowly added dropwise to the reaction solution, and after reaction at-10deg.C for 30min, the solution was allowed to warm to room temperature and stirring was continued for 2h. After the reaction, 3mL of methanol was slowly added dropwise under ice bath to quench the reaction, the reaction mixture was then transferred to a separatory funnel, diluted with an appropriate amount of saturated brine, extracted with DCM (3X 50 mL), the organic phases were combined and washed with saturated brine (2X 100 mL), and the organic phase was taken up in anhydrous Na 2 SO 4 Drying, filtering, removing solvent by vacuum rotary evaporation, and developing by TLC under pure V DCM :V CH3OH =25:1,R f =0.81, product was 175.0mg as red solid with 75% yield. 1 HNMR(400MHz,CDCl 3 )δ7.96-7.81(m,3H),7.71(s,1H),7.68(s,1H),7.56-7.50(m,3H),7.42(dd,J=6.2,3.3Hz,2H),7.32-7.27(m,1H),7.16(s,1H),6.89(d,J=8.4Hz,1H),6.58(d,J=9.2Hz,1H),5.68(d,J=8.4Hz,1H),4.19(dd,J=8.7,4.0Hz,1H),3.94-3.87(m,1H),3.79(d,J=2.8Hz,1H),3.64(s,2H),3.50(d,J=16.1Hz,1H),3.31(d,J=16.1Hz,1H),1.62(d,J=7.2Hz,6H). 13 C NMR(101MHz,CDCl 3 )δ170.89,170.55,133.64,133.42,132.61,132.28,132.25,132.21,128.88,128.40,128.16,127.82,127.63,127.48,127.42,127.17,126.72,126.41,126.29,125.91,125.86,69.45,53.47,50.18,48.20,43.96,43.88,29.73,29.40,14.17.HR-ESI-MS(m/z):calcd for C 30 H 31 N 2 O 3 ([M+H] + ) 467.2335, found:467.2334. All data confirm that the material is a mixture of optical isomers of 3-hydroxycyclohexyl-1, 2-bis (β -naphthyl) acetamide.
The resulting optical isomer mixture of 3-hydroxycyclohexyl-1, 2-bis (. Beta. -naphthyl) acetamide was isolated and purified by RP-HPLC: by C 18 A chromatographic column; the mobile phase A is water containing isopropanol and oxalic acid, the volume percentage concentration of the isopropanol is 0.6%, and the concentration of the oxalic acid is 0.5g/mL; the mobile phase B is methanol containing isopropanol and oxalic acid, the volume percentage concentration of the isopropanol is 0.6%, and the concentration of the oxalic acid is 0.5g/mL; elution procedure: 0-5min,5% phase b; 5-10min,5% -25% of phase B; 10-12min,25% -38% of phase B; 12-35min,38% phase B; the flow rate was 0.8mL/min.
(1S, 2S, 3R) -3-hydroxycyclohexa-1, 2-bis (. Beta. -naphthyl) acetamide (CL-A1-1): the fraction with retention time rt=27.8 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 65.5mg of a red solid.
(1S, 2R, 3R) -3-hydroxycyclohexa-1, 2-bis (. Beta. -naphthyl) acetamide (CL-A1-2): the fraction with retention time rt=26.3 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 36.6mg of a red solid.
(1R, 2S, 3R) -3-hydroxycyclohexa-1, 2-bis (. Beta. -naphthyl) acetamide (CL-A1-3): the fraction with retention time rt=25.6 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 16.7mg of a red solid.
(1R, 2R, 3R) -3-hydroxycyclohexa-1, 2-di (. Beta. -naphthyl) acetamide (CL-A1-4): the fraction with retention time rt=24.4 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 12.8mg of a red solid.
(1S, 2S, 3S) -3-hydroxycyclohexa-1, 2-bis (. Beta. -naphthyl) acetamide (CL-A1-5): the fraction with retention time rt=23.3 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 14.2mg of a red solid.
(1S, 2R, 3S) -3-hydroxycyclohexa-1, 2-bis (. Beta. -naphthyl) acetamide (CL-A1-6): the fraction with retention time rt=22.6 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 14.9mg of a red solid.
(1R, 2S, 3S) -3-hydroxycyclohexa-1, 2-bis (. Beta. -naphthyl) acetamide (CL-A1-7): the fraction with retention time rt=21.5 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 8.3mg of a red solid.
(1R, 2R, 3S) -3-hydroxycyclohexa-1, 2-di (. Beta. -naphthyl) acetamide (CL-A1-8): the fraction with retention time rt=20.9 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 6.0mg of a red solid.
Example 2
Preparation, separation and purification of 3-hydroxycyclohexyl-1, 2-bis (3, 4-dimethoxyphenyl) formamide (CL-A2-1-CL-A2-8)
(1) Preparation of 3, 4-dimethoxy benzoic acid. Purchased from commercial sources.
(2) Preparation of 2, 3-diaminocyclohexane-1-benzyl ether. Prepared according to example 1, steps 2.1-2.6.
(3) Preparation, separation and purification of 3-hydroxycyclohexyl-1, 2-bis (3, 4-dimethoxyphenyl) formamide (CL-A2-1-CL-A2-8).
3.1. The procedure is as in example 1, step 3.1, with the replacement of the starting material β -naphthylacetic acid by 3, 4-dimethoxybenzoic acid (364.4 mg,2.0 mmol) and the use of 2, 3-diaminocyclohexane-1-benzyl ether (180.7 mg,0.82 mmol). TLC developing agent condition V DCM :V CH3OH =25:1, silica gel column chromatography separation and purification, collection R f Fraction=0.61 to give 242.9mg of white solid in 54% yield. 1 H NMR(400MHz,CDCl 3 )δ8.17(d,J=9.1Hz,1H),7.84(d,J=8.6Hz,1H),7.14(dd,J=26.9,8.8Hz,2H),6.49-6.45(m,2H),6.34(d,J=7.3Hz,3H),5.81(dd,J=8.8,1.9Hz,2H),5.75(d,J=2.2Hz,1H),5.67(d,J=2.2Hz,1H),3.94(d,J=12.0Hz,1H),3.81-3.75(m,1H),3.65(d,J=12.0Hz,1H),3.55(d,J=3.8Hz,1H),3.14(s,1H),3.03(d,J=4.5Hz,6H),2.70(s,6H),1.39(d,J=13.1Hz,1H),1.09-0.96(m,3H),0.83-0.70(m,2H); 13 C NMR(101MHz,CDCl 3 )δ167.18,166.54,160.77,160.46,139.73(2),134.24(2),129.35(2),128.63(2),115.19(2),114.39(2),106.98(2),106.75(2),99.37(2),77.96,72.60,56.40(2),56.09,51.88,30.75,28.46,16.28.HR-ESI-MS(m/z):calcd for C 31 H 37 N 2 O 7 ([M+H] + ) 549.2604, found:549.2601. All data confirm that the material is a mixture of optical isomers of 3-benzyloxycyclohexyl-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide.
3.2. By proceeding as in example 1, step 3.2, replacing the starting material with a mixture of optical isomers of 3-benzyloxycyclohexyl-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide (274.3 mg,0.5 mmol) in a dry 100mL round bottom flask, the TLC developing solvent conditions were prepared as V DCM :V CH3OH =25:1, silica gel column chromatography separation and purification, collection R f Fraction=0.72, giving 174.2mg of white solid in 76% yield. 1 H NMR(400MHz,CDCl 3 )δ8.17(d,J=9.1Hz,1H),7.84(d,J=8.6Hz,1H),7.14(dd,J=26.9,8.8Hz,2H),5.76(d,J=2.2Hz,1H),5.65(d,J=2.2Hz,1H),3.93(d,J=12.0Hz,1H),3.82-3.74(m,1H),3.63(d,J=12.0Hz,1H),3.57(d,J=3.8Hz,1H),3.15(s,1H),3.03(d,J=4.5Hz,6H),2.73(s,6H),1.41(d,J=13.1Hz,1H),1.07-0.98(m,3H),0.85-0.72(m,2H). 13 C NMR(101MHz,CDCl 3 )δ167.21,166.96,153.42,153.35,149.8(2),128.63(2),120.92(2),115.19,114.39,111.89(2),63.12,58.6,56.09(4),51.88,32.95,32.83,16.85.HR-ESI-MS(m/z):calcd for C 24 H 31 N 2 O 7 ([M+H] + ) 459.2131, found:459.2136. All data confirm that the material is an optical isomer of 3-hydroxycyclohexyl-1, 2-bis (3, 4-dimethoxyphenyl) carboxamideA bulk mixture.
The resulting optical isomer mixture of 3-hydroxycyclohexyl-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide was isolated and purified by RP-HPLC: by C 18 A chromatographic column; the mobile phase A is water containing isopropanol and oxalic acid, the volume percentage concentration of the isopropanol is 0.6%, and the concentration of the oxalic acid is 0.5g/mL; the mobile phase B is methanol containing isopropanol and oxalic acid, the volume percentage concentration of the isopropanol is 0.6%, and the concentration of the oxalic acid is 0.5g/mL; elution procedure: 0-5min,5% phase b; 5-10min,5% -25% of phase B; 10-12min,25% -38% of phase B; 12-35min,38% phase B; the flow rate was 0.8mL/min.
(1S, 2S, 3R) -3-hydroxycyclohexa-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide (CL-A2-1): the fraction with retention time rt=26.7 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 63.9mg of a white solid.
(1S, 2R, 3R) -3-hydroxycyclohexa-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide (CL-A2-2): the fraction with retention time rt=25.2 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 35.1mg of a white solid.
(1R, 2S, 3R) -3-hydroxycyclohexa-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide (CL-A2-3): the fraction with retention time rt=24.5 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 15.8mg of a white solid.
(1 r,2r,3 r) -3-hydroxycyclohexa-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide (CL-A2-4): the fraction with retention time rt=23.2 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 13.7mg of a white solid.
(1S, 2S, 3S) -3-hydroxycyclohexa-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide (CL-A2-5): the fraction with retention time rt=22.1 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 15.1mg of a white solid.
(1S, 2R, 3S) -3-hydroxycyclohexa-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide (CL-A2-6): the fraction with retention time rt=21.5 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 14.7mg of a white solid.
(1R, 2S, 3S) -3-hydroxycyclohexa-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide (CL-A2-7): the fraction with retention time rt=20.4 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 8.7mg of a white solid.
(1 r,2r,3 s) -3-hydroxycyclohexa-1, 2-bis (3, 4-dimethoxyphenyl) carboxamide (CL-A2-8): the fraction with retention time rt=19.8 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 7.2mg of a white solid.
Example 3
Preparation, separation and purification of 3-hydroxycyclohexyl-1, 2-di ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-1-CL-A3-8)
(1) Preparation of (E) -3- (2-bromo-4-fluorophenyl) acrylic acid
1.1. 2-bromo-4-fluorobenzaldehyde (203.0 mg,1.0 mmol) and maleic acid (260.2 mg,2.5 mmol) were dissolved in 30mL of pyridine, then a catalytic amount of piperidine (12.8 mg,0.15 mmol) was added, the mixture was slowly warmed to 110℃and the reaction stirred for 15 hours at this temperature and TLC was followed until the reaction was completed. The reaction mixture was cooled to room temperature, 30mL of 1m NaOH solution was added until the solution became clear, washed with ethyl acetate (20 ml×2), the aqueous layer was adjusted to pH 2.0 with 50% sulfuric acid solution, precipitated out, cooled in an ice bath, the solid compound was collected by suction filtration, washed with cold water, cold diethyl ether, sucked dry, and dried under vacuum. 225.4mg of white solid was obtained in 92% yield. 1 H NMR(300MHz,DMSO-d 6 )δ(ppm):12.66(s,1H,COOH),8.07-7.98(m,1H,Ar);7.81-7.76(d,J=15.9Hz,1H),7.71-7.67(m,1H,Ar),7.36-7.33(m,1H,Ar),6.57-6.52(d,J=15.0Hz,1H); 13 C NMR(75MHz,DMSO-d 6 )δ(ppm):167.54,164.65-161.30(d,J=251Hz,Ar-C attached to F,-1C),140.69(trans C=C,-1C),130.79(Ar,-1C),130.50-130.38(d,Ar,-1C),125.47-125.34(d,Ar,-1C),122.75-122.73(d,trans C=C,-1C),120.84-120.51(d,Ar,-1C),116.34-116.06(d,Ar,-1C);ESI-MS(-ve)m/z:244.7[M-H] +
(2) Preparation of 2, 3-diaminocyclohexane-1-benzyl ether. Prepared according to example 1, steps 2.1-2.6.
(3) Preparation, separation and purification of 3-hydroxycyclohexyl-1, 2-bis ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-1 to CL-A3-8).
3.1. The procedure was followed as in example 1, step 3.1, except that the starting material was changed to (E) -3- (2-bromo-4-fluorophenyl) acrylic acid (490.1 mg,2.0 mmol) and 2, 3-diaminocyclohexane-1-benzyl ether was used in an amount of (180.7 mg,0.82 mmol). TLC developing agent condition V DCM :V CH3OH =25:1, silica gel column chromatography separation and purification, collection R f Fraction=0.38, yielding 365.0mg of brown solid with a yield of 66%. 1 H NMR(400MHz,CDCl 3 )δ7.75(d,J=15.5Hz,1H),7.67-7.58(m,3H),7.43-7.39(m,1H),7.24-7.19(m,5H),6.90(d,J=8.0Hz,2H),6.26(dd,J=18.2,11.6Hz,2H),5.91(d,J=15.5Hz,1H),4.58(d,J=11.0Hz,1H),4.31(d,J=11.0Hz,2H),4.22-4.16(m,1H),3.95-3.89(m,1H),1.78(d,J=3.3Hz,1H),1.67-1.38(m,5H). 13 C NMR(101MHz,CDCl 3 )δ165.33(s),163.96(s),138.93(s),137.95(d,J=12.9Hz),128.78(s),128.06(d,J=18.9Hz),126.61(s),125.99(s),123.79(s),123.14(s),120.64(s),120.40(s),115.06(dd,J=21.5,13.9Hz),110.99(s),77.22(d,J=8.1Hz),71.31(s),50.73(s),29.72(s),27.17(s),15.34(s).HR-ESI-MS(m/z):calcd for C 31 H 29 N 2 O 3 F 2 Br 2 ([M+H] + ) 675.0483, found:675.0508. All data confirm that this material is a mixture of optical isomers of 3-benzyloxycyclohexyl-1, 2-bis ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide.
3.2. The procedure was followed as in example 1, step 3.2, substituting 3-benzyloxycyclohexyl-1, 2-bis ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (337.2 mg,0.5 mmol). TLC developing agent condition V DCM :V CH3OH =25:1, silica gel column chromatography separation and purification, collection R f Fraction=0.61 to yield 128.5mg of red solid in 44%; 1 H NMR(400MHz,CD Cl 3 )δ7.85(dt,J=15.6,3.5Hz,2H),7.71(s,1H),7.54-7.43(m,2H),7.31-7.26(m,2H),7.01-6.87(m,2H),6.74(d,J=58.1Hz,1H),6.48-6.29(m,2H),4.44(d,J=2.2Hz,1H),4.25(s,1H),4.16(d,J=2.3Hz,1H),1.91-1.68(m,6H); 13 C NMR(101MHz,CDCl 3 )δ166.00(s),165.63(s),164.10(s),161.58(d,J=1.7Hz),138.92(s),138.35(s),131.27(d,J=3.8Hz),128.79(d,J=8.6Hz),125.50(d,J=9.5Hz),124.17(s),123.52(s),120.76(s),120.52(s),115.36(s),115.15(s),69.80(s),52.30(s),49.37(s),31.55(d,J=3.8Hz),29.30(s),17.58(s).HR-ESI-MS(m/z):calcd for C 24 H 23 N 2 O 3 F 2 Br 2 ([M+H] + ) 583.0043, found:583.0050. All data confirm that this material is a mixture of optical isomers of 3-hydroxycyclohexyl-1, 2-bis ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-1-CL-A3-8).
The resulting optical isomer mixture of 3-hydroxycyclohexyl-1, 2-bis ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide was isolated and purified by RP-HPLC: by C 18 A chromatographic column; the mobile phase A is water containing isopropanol and oxalic acid, the volume percentage concentration of the isopropanol is 0.6%, and the concentration of the oxalic acid is 0.5g/mL; the mobile phase B is methanol containing isopropanol and oxalic acid, the volume percentage concentration of the isopropanol is 0.6%, and the concentration of the oxalic acid is 0.5g/mL; elution procedure: 0-5min,5% phase b; 5-10min,5% -25% of phase B; 10-12min,25% -38% of phase B; 12-35min,38% phase B; the flow rate is 0.8mL/min;
(1S, 2S, 3R) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-1): the fraction with retention time rt=28.3 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 48.1mg of a red solid.
(1 s,2r,3 r) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-2): the fraction with retention time rt=26.8 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 26.9mg of a red solid.
(1 r,2s,3 r) -3-hydroxycyclohexa-1, 2-di ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-3): the fraction with retention time rt=26.2 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 12.3mg of a red solid.
(1 r,2r,3 r) -3-hydroxycyclohexa-1, 2-di ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-4): the fraction with retention time rt=25.3 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 9.4mg of a red solid.
(1S, 2S, 3S) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-5): the fraction with retention time rt=24.1 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 10.4mg of a red solid.
(1 s,2r,3 s) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-6): the fraction with retention time rt=23.5 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 10.9mg of a red solid.
(1R, 2S, 3S) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-7): the fraction with retention time rt=22.4 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 6.1mg of a red solid.
(1 r,2r,3 s) -3-hydroxycyclohexa-1, 2-di ((E) -3- (2-bromo-4-fluorophenyl)) acrylamide (CL-A3-8): the fraction with retention time rt=21.2 min was collected, the solvent was removed by rotary evaporation in vacuo and the residue was freeze-dried to give 4.4mg of a red solid.
Example 4
Preparation, separation and purification of 3-hydroxycyclohexyl-1, 2-di ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-1-CL-A4-8)
(1) Preparation of (E) -3- (4-nitrophenyl) acrylic acid
1.1. The procedure was followed as in example 4, step 1.1, substituting 4-nitrobenzaldehyde (151.1 mg,1.0 mmol) for the starting material. 177.7mg of yellow solid was obtained in 92% yield. Melting point 287 ℃; IR (KBr) v max in cm -1 :2513,1683,1425,1225(COOH),1629,1550(C=C alkene),1305,868(NO 2 ),1225(Ar–C=C),1108(Ar–C–H); 1 H NMR(DMSO-d 6 ,500MHz)δ(ppm):12.68(s,1H),8.24(d,J=8.6Hz,2H),7.98–7.97(d,J=8.6Hz,2H),7.70–7.68(d,J=15.6Hz,1H),6.75–6.73(d,J=15.6Hz,1H)。
(2) Preparation of 2, 3-diaminocyclohexane-1-benzyl ether. Prepared according to example 1, steps 2.1-2.6.
(3) Preparation, separation and purification of 3-hydroxycyclohexyl-1, 2-bis ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-1-CL-A4-8).
3.1. The procedure was followed as in example 1, step 3.1, substituting starting material with (E) -3- (4-nitrophenyl) acrylic acid (386.3 mg,2.0 mmol) and 2, 3-diaminocyclohexane-1-benzyl ether in an amount of (180.7 mg,0.82 mmol). TLC developing agent condition is pure V DCM :V CH3OH =25:1, silica gel column chromatography separation and purification, collection R f Fraction=0.24, yielding 332.2mg of yellow solid in 71% yield. 1 H NMR(400MHz,CD 3 OD)δ(ppm):8.17(d,J=8.7Hz,4H),8.01(s,2H),7.69(dd,J=21.9,8.7Hz,4H),7.55(d,J=2.4Hz,2H),7.44(d,J=7.2Hz,2H),7.33(d,J=7.6Hz,3H),7.04(d,J=15.8Hz,1H),6.71(d,J=15.8Hz,1H),4.79(s,1H),4.64(d,J=13.4Hz,2H),4.28(s,1H),3.86-3.80(m,1H),2.02-1.72(m,5H),1.50(dd,J=8.5,3.9Hz,1H).HR-ESI-MS(m/z):calcd for C 31 H 31 N 4 O 7 ([M+H] + ) 571.2193, found:571.2198. All data confirm that this material is a mixture of optical isomers of 3-benzyloxycyclohexyl-1, 2-bis ((E) -3- (4-nitrophenyl)) acrylamide.
3.2. By proceeding as in example 1, step 3.2, the starting material was replaced by 3-benzyloxycyclopentane-1, 2-bis ((E) -3- (4-nitrophenyl)) acrylamide (285.3 mg,0.5 mmol); TLC developing agent condition V DCM :V CH3OH =25:1, silica gel column chromatography separation and purification, collection R f Fraction=0.31, yielding 124.8mg of yellow solid in 52% yield. 1 H NMR(400MHz,CD 3 OD)δ(ppm):8.25(d,J=1.0Hz,2H),8.23(d,J=1.0Hz,2H),7.78(dd,J=8.8,5.1Hz,4H),7.65-7.54(m,2H),7.01(d,J=15.8Hz,1H),6.81(d,J=15.8Hz,1H),4.51-4.40(m,1H),4.29-4.21(m,1H),3.99(dd,J=7.2,3.6Hz,1H),1.93-1.65(m,6H).HR-ESI-MS(m/z):calcd for C 24 H 25 N 4 O 7 [M+H] + 481.1723, found:481.1722. All data confirm that this material is a mixture of optical isomers of 3-hydroxycyclohexyl-1, 2-bis ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-1-CL-A4-8).
The resulting optical isomer mixture of 3-hydroxycyclohexyl-1, 2-bis ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-1 to CL-A4-8) was isolated and purified by RP-HPLC: by C 18 A chromatographic column; the mobile phase A is water containing isopropanol and oxalic acid, the volume percentage concentration of the isopropanol is 0.6%, and the concentration of the oxalic acid is 0.5g/mL; the mobile phase B is methanol containing isopropanol and oxalic acid, the volume percentage concentration of the isopropanol is 0.6%, and the concentration of the oxalic acid is 0.5g/mL; elution procedure: 0-5min,5% phase b; 5-10min,5% -25% of phase B; 10-12min,25% -38% of phase B; 12-35min,38% phase B; the flow rate is 0.8mL/min;
(1S, 2S, 3R) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-1): the fraction with retention time rt=27.2 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 46.0mg of a yellow solid.
(1S, 2R, 3R) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-2): the fraction with retention time rt=25.7 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 25.3mg of yellow solid.
(1R, 2S, 3R) -3-hydroxycyclohexa-1, 2-di ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-3): the fraction with retention time rt=25.0 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 11.4mg of yellow solid.
(1R, 2R, 3R) -3-hydroxycyclohexa-1, 2-di ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-4): the fraction with retention time rt=23.7 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 9.9mg of a yellow solid.
(1S, 2S, 3S) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-5): the fraction with retention time rt=22.6 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 15.1mg of a yellow solid.
(1S, 2R, 3S) -3-hydroxycyclohexa-1, 2-di ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-6): the fraction with retention time rt=22.1 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 10.8mg of yellow solid.
(1R, 2S, 3S) -3-hydroxycyclohexa-1, 2-di ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-7): the fraction with retention time rt=20.8 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 6.3mg of a yellow solid.
(1R, 2R, 3S) -3-hydroxycyclohexa-1, 2-di ((E) -3- (4-nitrophenyl)) acrylamide (CL-A4-8): the fraction with retention time rt=20.3 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 5.1mg of a yellow solid.
Example 5
Preparation, separation and purification of 3-hydroxycyclohexyl-1, 2-di ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-1 to CL-A5-8)
(1) Preparation of (E) -3- (3-hydroxymethyl-4-hydroxyphenyl) acrylic acid
1.1. The procedure was followed as in example 4, step 1.1, substituting 4-hydroxybenzaldehyde (122.1 mg,1.0 mmol) for the starting material. 149.4mg of yellow needle-like solid was obtained in 91% yield. 1 H NMR(300MHz,CD 3 OD)δ:7.63(d,J=15.9Hz,1H),7.55(d,J=8.1Hz,2H),6.87(d,J=8.1Hz,2H),6.32(d,J=15.9Hz,1H); 13 C NMR(300MHz,CD 3 OD) δ 168.1,160.3,145.7,131.1,127.2,116.5,115.8. All data confirm that the material is (E) -3- (4-hydroxyphenyl) acrylic acid.
1.2. (E) -3- (4-hydroxyphenyl) acrylic acid (328.3 mg,2.0 mmol), formaldehyde (540.5 mg,18.0 mmol) and boric acid (371.0 mg,6.0 mmol) were dissolved in 20mL of p-xylene, heated to 150℃in an oil bath, and reacted under stirring at this temperature for 12 hours. Cooling to room temperature, precipitating a large amount of yellow precipitate, filtering, washing the obtained precipitate with a proper amount of water, and then drying to obtain 318.5mg of (E) -3- (3-hydroxymethyl-4-hydroxyphenyl) acrylic acid, yield 82%, melting point (mp): 185.1 to 185.7 ℃. 1 H NMR(DMSO-d 6 ,300MHz)δ(ppm):12.68(s,1H),9.71(s,1H),7.52(d,J=15.8Hz,1H),7.43(d,J=8.3Hz,1H),7.29(d,J=8.3Hz,1H),7.13(s,1H),6.37(d,J=15.8Hz,1H)。
(2) Preparation of 2, 3-diaminocyclohexane-1-benzyl ether. Prepared according to example 1, steps 2.1-2.6.
(3) Preparation, separation and purification of 3-hydroxycyclohexyl-1, 2-di ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-1 to CL-A5-8)
3.1. The procedure was followed as in example 1, step 3.1, except that the starting material was changed to (E) -3- (3-hydroxymethyl-4-hydroxyphenyl) acrylic acid (388.4 mg,2.0 mmol) and 2, 3-diaminocyclohexane-1-benzyl ether was used in an amount of (180.7 mg,0.82 mmol). TLC developing agent condition is pure V DCM :V CH3OH =25:1, silica gel column chromatography separation and purification, collection R f Fraction=0.19, yielding 3295.8mg of yellow solid in 63% yield. 1 H NMR(300MHz,DMSO-d 6 )δ(ppm):9.71(s,2H),8.35(br,2H),7.43(d,J=8.1Hz,2H),7.39(d,J=15.3Hz,2H),7.33-7.31(m,5H,H-Ph),7.29(d,J=8.1Hz,2H),7.12(m,2H),7.02(m,2H),6.53(d,J=15.3Hz,2H),4.63-4.48(m,4H,-CH 2 -Ph),4.38(m,1H),4.17(m,1H),3.87(m,1H),1.72(m,2H),1.55-1.46(m,4H); 13 C NMR(75MHz,DMSO-d 6 )δ(ppm):167.1,166.7,155.6(2C),141.9(2C),137.8,130.2(2C),128.9(2C),128.7(2C),128.3(2C),127.9,127.5(2C),126.2(2C),120.4(2C),116.1(2C),74.6,72.9,60.7(2C),55.8,51.1,32.3,29.4,17.2;HR-ESI-MS(m/z):calcd for C 33 H 36 N 2 O 7 ([M+H] + ) 573.2601, found:573.2605. All data confirm that this material is a mixture of optical isomers of 3-benzyloxycyclohexyl-1, 2-bis ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide.
3.2. By proceeding as in example 1, step 3.2, the starting material was replaced with 3-benzyloxycyclohexyl-1, 2-bis ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (286.3 mg,0.5 mmol); TLC developing agent condition is pure V DCM :V CH3OH =25:1, silica gel column chromatography separation and purification, collection R f The value was 0.31 to give a fraction. 185.8mg of yellow solid was obtained in 77% yield. 1 H NMR(300MHz,DMSO-d 6 )δ(ppm):9.69(s,2H),8.34(br,2H),7.42(d,J=8.1Hz,2H),7.38(d,J=15.3Hz,2H),7.27(d,J=8.1Hz,2H),7.13(m,2H),7.01(m,2H),6.54(d,J=15.3Hz,2H),5.36(br,1H),4.64-4.49(m,4H,-CH 2 -Ph),4.21-4.16(m,3H),1.73(m,2H),1.56-1.47(m,4H); 13 C NMR(75MHz,DMSO-d 6 )δ(ppm):167.3,166.5,155.5(2C),141.8(2C),130.1(2C),128.8(2C),128.1(2C),126.3(2C),120.1(2C),116.2(2C),62.3,60.5(2C),58.0,50.8,32.9,32.4,16.9;HR-ESI-MS(m/z):calcd for C 26 H 30 N 2 O 7 ([M+H] + ) 483.2131, found:483.2136. All data confirm that this material is a mixture of optical isomers of 3-hydroxycyclohexa-1, 2-bis ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-1-CL-A5-8).
The resulting optical isomer mixture of 3-hydroxycyclohexyl-1, 2-bis ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-1 to CL-A5-8) was isolated and purified by RP-HPLC: by C 18 A chromatographic column; the mobile phase A is water containing isopropanol and oxalic acid, the volume percentage concentration of the isopropanol is 0.6%, and the concentration of the oxalic acid is 0.5g/mL; the mobile phase B is methanol containing isopropanol and oxalic acid, the volume percentage concentration of the isopropanol is 0.6%, and the concentration of the oxalic acid is 0.5g/mL; elution procedure: 0-5min,5% phase b; 5-10min,5% -25% of phase B; 10-12min,25% -38% of phase B; 12-35min,38% phase B; the flow rate is 0.8mL/min;
(1S, 2S, 3R) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-1): the fraction with retention time rt=26.2 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 66.7mg of yellow solid.
(1S, 2R, 3R) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-2): the fraction with retention time rt=24.9 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 36.5mg of a yellow solid.
(1R, 2S, 3R) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-3): the fraction with retention time rt=24.2 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 16.5mg of yellow solid.
(1 r,2r,3 r) -3-hydroxycyclohexa-1, 2-di ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-4): the fraction with retention time rt=22.8 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 14.4mg of yellow solid.
(1S, 2S, 3S) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-5): the fraction with retention time rt=21.9 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 21.9mg of a yellow solid.
(1S, 2R, 3S) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-6): the fraction with retention time rt=21.2 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 15.7mg of a yellow solid.
(1R, 2S, 3S) -3-hydroxycyclohexa-1, 2-bis ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-7): the fraction with retention time rt=19.7 min was collected, the solvent was removed by rotary evaporation in vacuo, and the residue was freeze-dried to give 9.1mg of a yellow solid.
(1 r,2r,3 s) -3-hydroxycyclohexa-1, 2-di ((E) -3- (3-hydroxymethyl-4-hydroxyphenyl)) acrylamide (CL-A5-8): the fraction with retention time rt=18.6 min was collected, the solvent was removed by rotary evaporation in vacuo and the residue was freeze-dried to give 7.4mg of a yellow solid.
EXAMPLE 6 anti-respiratory viral Activity assay of hydroxycyclohexanamide compounds
Virus, host cell material: respiratory Syncytial Virus (RSV) is selected, and the host cell is laryngeal cancer cell (HEp-2). RSV was purchased from Shanghai, biotechnology Inc., and HEp-2 cells were purchased from the China academy of sciences cell bank.
The cell growth liquid is MEM medium containing 10% by mass of calf serum (FBS), and the cell maintenance liquid is MEM medium containing 1% by mass of FBS.
The positive control drugs were ribavirin, methyl 3, 4-O-dicaffeoylquinic acid, which is a natural product from elephantopus scaber, and 1α,2β -O, O-dicaffeoylcyclopent-3β -ol.
Tetrazolium salt (MMT) solution formulation: a5 mg/mL solution was prepared with buffer PBS (0.1M, pH 7.4).
Sample solution preparation: the hydroxycyclohexanamide compounds prepared in examples 1 to 5, methyl 3, 4-O-dicaffeoylquinic acid, 1. Alpha., 2. Beta. -O, O-dicaffeoylcyclopent-3. Beta. -ol and ribavirin were prepared into 200. Mu.g/mL and 100. Mu.g/mL of sample solutions, respectively, using cell maintenance solution.
(1) Detection of compounds by MMTCytotoxicity: HEp-2 cells were seeded in 96-well plates at a density of 1X 10 4 After each well, the monolayer cells were grown, the sample solution prepared by diluting the cell-maintaining solution was added to a serial concentration (3.1 to 200. Mu.g/mL), and at 37℃5% CO 2 Culturing in an incubator for 3 days, adding 10 mu L of the prepared MTT solution, and culturing for 4 hours. Sucking out the sample solution, adding 100 mu L of dimethyl sulfoxide (DMSO), placing the 96-well plate in a micro-hollow plate oscillator at room temperature, vibrating for 10min, detecting an OD value at 570nm by using an enzyme-labeling instrument, and calculating the survival rate of HEp-2 cells; each group was set up with 4 balance holes, repeated 3 times. The calculation result was plotted to determine the median toxicity concentration (CC 50 ) The detection results are shown in Table 1.
(2) Antiviral activity was determined by observing the extent of inhibition of cytopathic effects by the samples: HEp-2 cells were cultured in 96-well plates, after a monolayer of cells was grown, 100-fold and half-number of infections (100 TCID) diluted with cell-retaining solution were added 50 ) The RSV virus solution of (2) is diluted with a cell-retaining solution to a serial concentration (0.4-100.0. Mu.g/mL) and is diluted with 5% CO at 37 DEG C 2 Culturing in an incubator for 3-4 days. The extent of cytopathic effect (CPE) was observed daily under an inverted microscope and recorded. -indicating no CPE; + means that 0 to 25% of the cells have CPE;2+ means that 25 to 50% of the cells have CPE;3+ means that 50 to 70% of the cells have CPE;4+ means that 75 to 100% of the cells have CPE. Finally, half maximal Inhibitory Concentration (IC) 50 ). Selectivity Index (SI) =cc 50 /IC 50 . The experimental results are shown in Table 1.
TABLE 1 anti-respiratory syncytial virus Activity of hydroxycyclohexanamide Compounds and positive controls
/>
* And (3) injection: the host cell of Respiratory Syncytial Virus (RSV) is HEp-2, CC 50 IC at half toxic concentration 50 At half inhibition concentration, SI is the selectivity index, si=cc 50 /IC 50 The method comprises the steps of carrying out a first treatment on the surface of the CL-An (n=1 to 5) represents a mixture of all optical isomers.
The data in Table 1 shows that all hydroxycyclohexanamide compounds have better anti-RSV activity and selectivity than the positive control ribavirin; most hydroxycyclohexanamide compounds have better anti-RSV activity and selectivity than methyl 3,4-O, O-dicaffeoylquinic acid; only the series CL-A3 and CL-A5 had better anti-RSV viral activity and selectivity than 1 a, 2 β -O, O-dicaffeoylcyclopent-3 β -ol; among them, the combination evaluation of CL-A3-7 and CL-A5-7 is particularly preferable.
EXAMPLE 7 stability of partial hydroxycyclohexanamide Compounds in fetal bovine serum
Experimental materials: both calf serum (FBS) and medium DMEM were purchased from GIBCO corporation.
The experimental process comprises the following steps: 1) Establishing a standard working curve of a detection sample by using a high performance liquid phase method; 2) Preparing a sample solution with the final concentration of 20mM, adding 200 mu L of the sample solution and 1.8mL of mouse whole blood into a 6-hole plate, vortex shaking for 5min, standing at 37 ℃ for incubation, taking 10 mu L of mixed samples at 0.25h, 0.5h, 1h, 2h, 4h, 8h, 12h and 24h respectively, adding 90 mu L of 20% (v/v) perchloric acid methanol solution, standing for 30min after shaking and mixing, and centrifuging for 5min at 10000 rpm. The supernatant was analyzed by HPLC. The half-life of the sample to be tested in fetal bovine serum was obtained by plotting the time-concentration and the results are shown in table 2.
TABLE 2 stability of hydroxycyclohexanamide compounds in fetal bovine serum
Sample of t 1/2 (h)
3, 4-O-dicaffeoylquinic acid methyl ester 3.28±0.11
1 alpha, 2 beta-O, O-dicaffeoylcyclopent-3 beta-ol 3.86±0.08
CLC-H15 6.56±0.13
CL-A3 9.78±0.21
CL-A5 8.47±0.17
Note that: CLC-H15 is a compound of invention patent CN110156628A (1S, 2S, 3S) -2-hydroxycyclohexa-1, 3-dicaffeamide.
As can be seen from table 2, the half-lives of the natural products 3,4-O, O-dicaffeoylquinic acid methyl ester and 1α,2β -O, O-dicaffeoylcyclopent-3β -ol from elephantopus scaber in mouse whole blood are 3.28 and 3.86h, respectively; the half-life of the compound CLC-H15 in the whole blood of the mice is 6.56 hours, so that the half-life of the compound CLC-H15 in the whole blood of the mice is improved; whereas the half-lives of CL-A3 and CL-A5 in the whole blood of mice were 9.78 and 8.47 hours, respectively, further improving the stability.
EXAMPLE 8 antioxidant value of partial hydroxycyclohexanamide Compounds
The experimental process comprises the following steps: accurately sucking 5.0mL of sample liquid to be detected (0.1 g/mL), adding 0.20mL of 1mol/L H 2 SO 4 And 3.00mL of 0.25mol/L H 2 O 2 Reacting at room temperature for 10min, adding 10.00mL of 10% KI solution, standing in dark for 5min, and adding 0.05mol/L Na 2 S 2 O 3 And (3) titrating the solution, indicating the end point by using 1% -2% of starch solution, and simultaneously making blank control. The antioxidant value is calculated according to the following formula:
antioxidant value = (V Blank space -V Sample of )×M×34/2V
In the formula V Blank space Na consumed for blank solution 2 S 2 O 3 Volume of solution (mL); v (V) Sample of Na consumed for the sample to be measured 2 S 2 O 3 Volume of solution (mL); v is the volume of the sample (mL), which is 5.0mL in this experiment; m is Na 2 S 2 O 3 The concentration of the solution; 34 is H 2 O 2 Molar mass of (c) is determined.
The experimental results are shown in Table 3.
TABLE 3 antioxidant value of hydroxycyclohexanamide compounds
Sample of AOV(mg/mL)
3, 4-O-dicaffeoylquinic acid methyl ester 0.087±0.002
1 alpha, 2 beta-O, O-dicaffeoylcyclopent-3 beta-ol 0.076±0.001
CLC-H15 0.069±0.003
CL-A3 0.015±0.001
CL-A5 0.021±0.003
Note that: CLC-H15 is a compound of invention patent CN110156628A (1S, 2S, 3S) -2-hydroxycyclohexa-1, 3-dicaffeamide.
As can be seen from Table 2, the antioxidant values of the natural products 3,4-O, O-dicaffeoylquinic acid methyl ester and 1 alpha, 2 beta-O, O-dicaffeoylcyclopent-3 beta-ol and the compound CLC-H15 are relatively high, which indicates that the natural products are relatively easy to oxidize and have poor oxidation stability; in contrast, the compounds CL-A3 and CL-A5 have relatively small oxidation resistance and relatively high oxidation stability. The antioxidant chemical stability of the primary structure was evaluated here with mixtures of optical isomers, with optically pure monomers and mixtures without involving biological enzyme oxidation.
EXAMPLE 9 investigation of partial Cyclic triol derivatives interactions with RSV pre-fusion F protein Using Biacore method
The pre-RSV (pre-fusion) F protein was coupled to the CM-5 sensor chip using an amino direct coupling method (ref: K.Hiroaki, E.Tomohiro, O.Noriko, N.Hiromasa, I.Mariko, U.Haruhisa, J.Pharm.And biomed. Analysis,2010,54 (1), 258-263), hydroxycyclohexanamide compounds (50. Mu.M, 25. Mu.M, 12.5. Mu.M, 6.25. Mu.M and 3.125. Mu.M) were set at different concentration gradients, positive control groups 3,4-O, O-dicaffeoylquinic acid methyl ester and 1α,2β -O, O-dicaffeoylcyclopenta-3β -ol, at concentration gradients of 100nM, 50nM, 25nM, 12.5nM, 6.25nM, 3.125nM and 1.0625nM, while solvent correction was set at the beginning of the experiment and between groups to eliminate curve shift due to DMSO content variation. And (3) obtaining affinity and kinetic constants between the point mutant protein and DH by using Biacore analysis software on the obtained experimental data. The experimental results are shown in Table 4.
TABLE 4 interaction of hydroxycyclohexanamide compounds with RSV pre-fusion F protein
Sample of K d (nM)
3, 4-O-dicaffeoylquinic acid methyl ester 2.5±0.1
1 alpha, 2 beta-O, O-dicaffeoylcyclopent-3 beta-ol 2.3±0.2
CLC-H15 1.4±0.2
CL-A3-7 1.7±0.3
CL-A5-7 1.3±0.1
As seen in Table 4, hydroxycyclohexanamide CL-A3-7 and CL-A5-7 are both capable of binding to protein F and bind more strongly than the natural products methyl 3, 4-O-dicaffeoylquinic acid and 1 alpha, 2 beta-O, O-dicaffeoylcyclopent-3 beta-ol from elephantopus scaber, but comparable to CLC-H15.
The above examples are preferred embodiments of the present invention, but the embodiments of the present invention are not limited to the above examples, and any other changes, modifications, substitutions, combinations, and simplifications that do not depart from the spirit and principle of the present invention should be made in the equivalent manner, and the embodiments are included in the protection scope of the present invention.

Claims (8)

1. A hydroxycyclohexanamide compound, which is characterized in that: the hydroxycyclohexanediamide compounds are compounds CL-A1-1 to CL-A1-8, CL-A2-1 to CL-A2-8, CL-A3-1 to CL-A3-8 and CL-A5-1 to CL-A5-8, and the structures are as follows:
wherein the structures of the groups A1, A2, A3 and A5 are as follows:
2. the method for producing a hydroxycyclohexanediamide compound according to claim 1, which comprises the steps of:
(1) Synthesis of carboxylic acids comprising the structures A3 and A5;
(2) Synthesis of 2, 3-diaminocyclohexanol
(2.1) Synthesis of 2-cyclohexen-1-ol;
(2.2) Synthesis of 2-cyclohexene-1-benzyl ether
Dissolving 2-cyclohexene-1-alcohol in redistilled tetrahydrofuran, adding tetrabutylammonium bromide, adding sodium hydride after balancing, reacting, dropwise adding benzyl bromide, reacting, moving to room temperature, and stirring to react to obtain 2-cyclohexene-1-benzyl ether;
(2.3) Synthesis of 2, 3-dihydroxycyclohexyl-1-benzyl ether
Dissolving the 2-cyclohexene-1-benzyl ether obtained in the step (2.2) in acetonitrile solution, adding an oxidant after balancing, reacting under ice bath condition, moving to room temperature, and stirring for reacting to obtain 2, 3-dihydroxycyclohexyl-1-benzyl ether;
(2.4) Synthesis of 3-Benzyloxycyclohexyl-1, 2-dimesylate
Dissolving the 2, 3-dihydroxycyclohexyl-1-benzyl ether obtained in the step (2.3) in redistilled methylene dichloride, adding 4- (N, N-dimethyl) aminopyridine, adding triethylamine under ice bath condition, dripping a sulfonylating reagent after balancing, reacting, stirring at room temperature, and reacting to obtain 3-benzyloxycyclohexyl-1, 2-dimethyl sulfonate;
(2.5) Synthesis of 2, 3-diazidocyclohexane-1-benzyl ether
Dissolving 3-benzyloxy-1, 2-dimethyl sulfonate obtained in the step (2.4) in a mixed solvent of N, N-Dimethylformamide (DMF) and hexamethylphosphoric triamide (HMPA), adding sodium azide, and reacting to obtain 1-benzyloxy-3-azidocyclohexyl-2-methane sulfonate;
dissolving the obtained 1-benzyloxy-3-azidocyclohexyl-2-methanesulfonate in a mixed solvent of N, N-Dimethylformamide (DMF) and hexamethylphosphoric triamide (HMPA), adding sodium azide, and reacting to obtain 2, 3-diazidocyclohexyl-1-benzyl ether;
(2.6) Synthesis of 2, 3-diaminocyclohexane-1-benzyl ether
Dissolving the 2, 3-diaza-cyclohexane-1-benzyl ether obtained in the step (2.5) in tetrahydrofuran, adding a reducing agent, and reacting to obtain a residue containing the 2, 3-diaminocyclohexane-1-benzyl ether;
(3) The synthesis of the hydroxycyclohexanamide compound comprises the following steps:
(3.1) respectively dissolving A1-COOH, A2-COOH, A3-COOH and A5-COOH in redistilled N, N-dimethylformamide, adding a condensing agent and 1-hydroxybenzotriazole (HOBt), reacting, adding N, N-Diisopropylethylamine (DIPEA), dissolving the 2, 3-diaminocyclohexane-1-benzyl ether obtained in the step (2.6) in N, N-dimethylformamide, and then dropwise adding into a reaction system to react to obtain an optical isomer mixture of 3-benzyloxycyclohexane-1, 2-diamide;
(3.2) dissolving the optical isomer mixture of 3-benzyloxycyclohexyl-1, 2-diamide obtained in the step (3.1) in redistilled methylene dichloride, dropwise adding a deprotection agent after balancing, reacting, moving to room temperature, stirring and reacting to obtain optical isomer mixtures CL-A1, CL-A2, CL-A3 and CL-A5 of 3-hydroxycyclohexyl-1, 2-diamide.
3. The preparation method according to claim 2, characterized in that: the molar ratio of tetrabutylammonium bromide to sodium hydride, benzyl bromide and 2-cyclohexene-1-ol in the step (2.2) is 1:20-60:6-20:6-20.
4. The preparation method according to claim 2, characterized in that: the amount of the 2, 3-diaminocyclohexane-1-benzyl ether in the step (3.1) is calculated according to the molar ratio of the 2, 3-diaminocyclohexane-1-benzyl ether to the An-COOH (n=1, 2,3, 5) of 0.2-1.0:1.0.
5. The preparation method according to claim 2, characterized in that: step (2.1) synthesis of 2-cyclohexen-1-ol, comprising the steps of:
2-cyclohexene-1-one is taken as a raw material, dissolved in methanol, added with cerium trichloride, reacted, added with sodium borohydride in batches, and reacted to obtain 2-cyclohexene-1-ol.
6. The method of manufacturing according to claim 5, wherein: the dosages of the cerium trichloride, the sodium borohydride and the 2-cyclohexene-1-one are calculated according to the molar ratio of 1-3:1.
7. The preparation method according to claim 2, characterized in that: the synthesis of A5-COOH comprises the following steps:
taking 4-hydroxy benzaldehyde as a starting material, dissolving the 4-hydroxy benzaldehyde and malonic acid in pyridine, adding organic base, and reacting to obtain 4-hydroxy phenyl acrylic acid;
dissolving 4-hydroxy phenyl acrylic acid, formaldehyde and an acid medium in paraxylene, and heating for reaction to obtain A5-COOH.
8. An anti-respiratory syncytial virus drug comprising a hydroxycyclohexanamide compound according to claim 1.
CN202111384289.8A 2021-11-18 2021-11-18 Hydroxycyclohexanamide compound and preparation method and application thereof Active CN114105807B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111384289.8A CN114105807B (en) 2021-11-18 2021-11-18 Hydroxycyclohexanamide compound and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111384289.8A CN114105807B (en) 2021-11-18 2021-11-18 Hydroxycyclohexanamide compound and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN114105807A CN114105807A (en) 2022-03-01
CN114105807B true CN114105807B (en) 2023-08-18

Family

ID=80439081

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111384289.8A Active CN114105807B (en) 2021-11-18 2021-11-18 Hydroxycyclohexanamide compound and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN114105807B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102219687A (en) * 2011-04-22 2011-10-19 暨南大学 Elephantopus scaber extract as well as preparation method and applications thereof in preparing antiviral drugs
CN102219686A (en) * 2011-04-22 2011-10-19 暨南大学 Caffeoyl derivative and use of coffeeoyl derivative in preparing drugs against respiratory syncytial viruses
DE102014016495A1 (en) * 2014-11-07 2016-05-12 Klaus Düring Process for the recovery of high purity 4,5-dicaffeoylquinic acid (4,5-DCQA) and use thereof for the treatment of respiratory syncytial virus (RSV) infections
CN110156628A (en) * 2019-06-19 2019-08-23 暨南大学 A kind of ring triol derivates and the preparation method and application thereof
CN110218160A (en) * 2019-06-19 2019-09-10 暨南大学 A kind of quinine acid derivative and the preparation method and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102219687A (en) * 2011-04-22 2011-10-19 暨南大学 Elephantopus scaber extract as well as preparation method and applications thereof in preparing antiviral drugs
CN102219686A (en) * 2011-04-22 2011-10-19 暨南大学 Caffeoyl derivative and use of coffeeoyl derivative in preparing drugs against respiratory syncytial viruses
DE102014016495A1 (en) * 2014-11-07 2016-05-12 Klaus Düring Process for the recovery of high purity 4,5-dicaffeoylquinic acid (4,5-DCQA) and use thereof for the treatment of respiratory syncytial virus (RSV) infections
CN110156628A (en) * 2019-06-19 2019-08-23 暨南大学 A kind of ring triol derivates and the preparation method and application thereof
CN110218160A (en) * 2019-06-19 2019-09-10 暨南大学 A kind of quinine acid derivative and the preparation method and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
应敏等.沙美特罗的合成.《广东化工》.2009,第36卷(第12期),第58、77页. *

Also Published As

Publication number Publication date
CN114105807A (en) 2022-03-01

Similar Documents

Publication Publication Date Title
CN114507221B (en) Triazine compound and application thereof in preparation of antiviral drugs
CA2693552A1 (en) Pyridone compound
CN111233929B (en) Deuterated nucleoside analogue and preparation method and application thereof
JP2002508347A (en) Cyclohexene carboxylate as a neuraminidase inhibitor
CN108473477A (en) The pyrimidine of aryl substitution for being used in influenza infection
CN103304547A (en) Preparation method of antidepressant drug-vilazodone
JP2022534067A (en) Compounds and their use as RET kinase inhibitors
WO2013181913A1 (en) Method for preparing artemisinin by artemisinic acid
CN106749446B (en) A kind of intermediate of epirubicin hydrochloride compound V
WO2010037210A1 (en) Viral polymerase inhibitors
CN110156628B (en) Cyclotriol derivative and preparation method and application thereof
CN114105807B (en) Hydroxycyclohexanamide compound and preparation method and application thereof
CN106749445A (en) A kind of intermediate of epirubicin hydrochloride compound III
CN112771048B (en) Inhibitors of influenza virus replication and intermediates and uses thereof
CN113321694A (en) N4-hydroxycytidine derivative and preparation method and application thereof
WO2010090305A1 (en) Substituted acylguanidine derivative
CN110218160B (en) Quinic acid derivative, and preparation method and application thereof
CN111635373A (en) Polycyclic sulfonamide ROR gamma modulators
CN106632275B (en) The preparation method of Lei Dipawei a kind of and the intermediate for preparing Lei Dipawei
CN115518058A (en) Application of N-cycloalkyl substituted aromatic methylamine compound in preparation of antiviral drug, structure and preparation method
CN112079785B (en) Novel anti-influenza virus oseltamivir derivative, and preparation method and application thereof
CN111100042B (en) Preparation method of 2-methoxy-5-sulfonamide benzoic acid
CN111777638A (en) Quinoline compound, preparation method, pharmaceutical composition and application thereof
CN106977543A (en) The preparation technology of improved Suo Feibuwei intermediates
CN112961081B (en) Bibenzamide urea compound and 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