CN115181096B - 3TC-PA compound bonded through ester bonds and application thereof - Google Patents

3TC-PA compound bonded through ester bonds and application thereof Download PDF

Info

Publication number
CN115181096B
CN115181096B CN202210777474.1A CN202210777474A CN115181096B CN 115181096 B CN115181096 B CN 115181096B CN 202210777474 A CN202210777474 A CN 202210777474A CN 115181096 B CN115181096 B CN 115181096B
Authority
CN
China
Prior art keywords
tert
butyldimethylsilyl
oxy
compound
oxathiolan
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
CN202210777474.1A
Other languages
Chinese (zh)
Other versions
CN115181096A (en
Inventor
韩凤梅
任家强
王俊俊
续威
陈祺
唐兰如
贺文丽
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.)
Hubei University
Original Assignee
Hubei 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 Hubei University filed Critical Hubei University
Priority to CN202210777474.1A priority Critical patent/CN115181096B/en
Publication of CN115181096A publication Critical patent/CN115181096A/en
Application granted granted Critical
Publication of CN115181096B publication Critical patent/CN115181096B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D411/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms
    • C07D411/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D411/04Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • A61P31/20Antivirals for DNA viruses
    • 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 relates to the field of medicine synthesis, in particular to a 3TC-PA compound bonded by ester bonds and application thereof. The structural formula of the 3TC-PA compound provided by the invention is shown as a formula (I), the compound maintains the advantages of two medicaments of 3TC and PA and the different anti-HBV mechanisms of the two medicaments, overcomes the problems of lack of inhibition of 3TC on HBV antigen secretion and low single-use oral bioavailability of PA, and is expected to be widely applied to preparing medicaments for treating CHB.

Description

3TC-PA compound bonded through ester bonds and application thereof
Technical Field
The invention relates to the field of medicine synthesis, in particular to a 3TC-PA compound bonded by ester bonds and application thereof.
Background
Lamivudine, the trans enantiomer of 2',3' -dideoxy-3 ' -thiocytosine (3 TC), was approved by the FDA in 1998 for sale and was the first antiviral nucleotide analog available for the treatment of Chronic Hepatitis B (CHB). After 3TC enters the cell, its 5' -OH can be converted to the active form lamivudine triphosphate (3 TC-TP) by a three-time phosphorylation process, and the structure is shown below.
Subsequently, 3TC-TP may be used as a chain terminator to add to the extending HBV DNA strand, or as a competitive inhibitor of deoxycytidine triphosphate. Thus, 3TC-TP can inhibit viral DNA synthesis, but not mitochondrial DNA synthesis. Namely lamivudine acts as an inhibitor of viral DNA-dependent DNA polymerase and RNA-dependent DNA polymerase (RNA-dependent DNA polymerase, RT).
The 3TC has good absorption after oral administration, the average bioavailability is 86-88%, the combination with plasma protein is moderate (36%), the distribution is wide, and the safety is high. However, 3TC only has good inhibition effect on HBV DNA, and has no obvious inhibition effect on HBV antigen secretion. Meanwhile, 3TC has obvious drug resistance after long-term use, and virus rebound is also common after drug withdrawal. Therefore, at present, most of the clinical application of 3TC for HBV is combined with other medicines.
Protocatechuic acid (Protocatechuic acid, PA) is known as 3, 4-dihydroxybenzoic acid and has the chemical structure shown below. The compound is a polyphenol substance, has better water solubility, is widely used in common Chinese herbal medicines such as acanthopanax sessiliflorus, orienter, phyllanthus urinaria, and the like, has pharmacological effects of resisting bacteria, easing pain, reducing myocardial oxygen consumption, improving myocardial oxygen resistance and the like, and has obvious anti-HBV effect.
By researching the inhibition mechanism of PA on HBV DNA and antigen (HBeAg and HBsAg) of HepG2.2.15 cells, the PA can lower the expression of hepatocyte nuclear factors HNF4α and HNF1α by activating ERK channel, reduce the interaction between HNF4α/1α and HBV promoter, inhibit the transcription and replication of HBV, and play the role of anti-HBV. Meanwhile, in vitro anti-HBV research shows that the combined use of 3TC and PA has additive phase inhibition effect on HepG2.2.15 cell HBeAg and HBV DNA, and synergistic inhibition effect on HBsAg, compared with the independent use of 3TC or PA, the combined use improves the drug effect, and PA can obviously enhance the capability of 3TC for inhibiting HBV X protein from entering the nucleus and reduce the overall activity level of HBV, which is one of important ways for inhibiting HBV replication by the combination of 3TC and PA. In duck primary hepatocytes infected with DHBV, the combination of PA and 3TC can remarkably enhance the inhibition of 3TC on the adsorption and entry of DHBV, the infection activity and the replication capacity. In vivo pharmacodynamicsExperiments show that: the 3TC and the PA are combined to effectively inhibit the DHBV infection of the duckling, the inhibition effect of the combination on the DHBV is stronger than that of single use, and the obvious difference between the medicine stopping and the medicine pre-taking shows that the combination can improve the medicine stopping rebound of the 3TC and the drug resistance problem of long-term use. The combination has stronger inhibition effect on the contents of glutamic-oxaloacetic transaminase, glutamic-pyruvic transaminase, alkaline phosphatase, albumin, total bilirubin and other enzymes in serum than 3TC and PA which are used independently, so that the combination of the two medicines can relieve liver injury, and the observation of pathological sections of liver can show that the combination of the two medicines can also improve liver tissue lesions. In vivo pharmacokinetic experiments show that after the two are combined, the apparent distribution volume of the 3TC is obviously reduced, which shows that the combined use can improve the bioavailability of the 3 TC; t is t l/2ka Extremely significant increases indicate a slow rate of 3TC absorption and AUC 0-t And T max Significantly increased but C max And t l/2β No significant difference, indicating an increase in total 3TC absorption after combination; t of PA after combination l/2β Significantly increase, t l/2ka ,T max The increase is very remarkable, which indicates that the absorption and elimination are slow and the in vivo action time is prolonged. Thus the combination of 3TC and PA has a significant pharmacokinetic interaction.
The study shows that the combination of 3TC and PA has obvious synergistic anti-HBV effect, and improves the rebound phenomenon of the drug withdrawal virus existing after the 3TC is used for a long time. However, PA has a relatively high molecular polarity and poor lipid solubility, which results in poor oral absorption and low bioavailability. Therefore, the synthesis of the chemical bond of 3TC and PA is hopeful to keep the advantages of the combination of the two medicines and the different anti-HBV mechanisms of the two medicines, make up for the lack of inhibition of 3TC on HBV antigen secretion, and simultaneously overcome the problem of low bioavailability of single oral administration of PA.
Disclosure of Invention
Based on this, the invention aims to provide a 3TC-PA compound bonded by ester bonds, and the structural formula of the compound is shown as a formula (I):
the 3TC-PA compound provided by the invention maintains the advantages of two drugs of 3TC and PA and the different anti-HBV mechanisms of the two drugs, and solves the problems of lack of inhibition of 3TC on HBV antigen secretion and low bioavailability of single oral administration of PA.
The invention also provides a preparation method of the 3TC-PA compound, which comprises the following steps:
step 1, synthesis of ((2 r,5 s) -5- (4- ((tert-butyloxycarbonyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-di ((tert-butyldimethylsilyl) oxy) benzoate (v);
step 2, synthesizing ((2R, 5S) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-dihydroxybenzoate (VI);
step 3, methyl ((2R, 5S) -5- (4-amino-2-oxopyrimidin-1 (2H) -yl) -1, 3-oxathiolan-2-yl) -3, 4-dihydroxybenzoate (I) is synthesized.
Wherein ((2R, 5S) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-di ((tert-butyldimethylsilyl) oxy) benzoate (V) in step 1 is prepared from tert-butyl (1- (((2R, 5S) -2- (hydroxymethyl) -1, 3-oxathiopyran-5-yl) -2-oxo-1, 2-dihydropyrimidin-4-yl) carbamate (II), (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((tert-butyldimethylsilyl) oxy) phenyl) methanone (IV) and 4-Dimethylaminopyridine (DMAP).
In the step, the molar ratio of the tert-butyl (1- (((2R, 5S) -2- (hydroxymethyl) -1, 3-oxathiopyran-5-yl) -2-oxo-1, 2-dihydropyrimidin-4-yl) carbamate (II) to the (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((tert-butyldimethylsilyl) oxy) phenyl) methanone (IV) to DMAP is 1 (1.1-1.5): (1.8-2.5) after the reaction is completed under the protection of nitrogen at 20-30 ℃, the reaction solution is decompressed and concentrated, and the residue is separated by column chromatography.
Further, the synthesis of ((2 r,5 s) -5- (4- ((tert-butyldimethylsilyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-di ((tert-butyldimethylsilyl) oxy) benzoate (v) in step 1 comprises the steps of:
step 1-1, using lamivudine, di-tert-butyl dicarbonate and anhydrous DMF as raw materials to prepare tert-butyl (1- (((2R, 5S) -2- (hydroxymethyl) -1, 3-oxathiopyran-5-yl) -2-oxo-1, 2-dihydropyrimidine-4-yl) carbamate (II), wherein in the step, the mol ratio of the lamivudine to the di-tert-butyl dicarbonate is 1 (1.5-2.0), the dosage of the anhydrous DMF can be 42mL/1g lamivudine after the reaction is finished, the reaction solution is diluted by distilled water and then extracted by ethyl acetate, the organic phase is dried by anhydrous sodium sulfate, filtered, decompressed, concentrated and separated by column chromatography, petroleum ether and ethyl acetate with the volume ratio of 1 (1-6) are adopted to obtain the compound;
step 1-2, (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((tert-butyldimethylsilyl) oxy) phenyl) methanone (IV) was prepared starting from 3, 4-bis ((tert-butyldimethylsilyl) oxy) benzoate (III), benzotriazole, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride. In this step, the molar ratio of 3, 4-bis ((t-butyldimethylsilyl) oxy) benzoate (III), benzotriazole (BTA), 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) was 1: (1-1.2): (1.4-1.8), after the reaction is finished, the reaction solution is concentrated under reduced pressure, and the residue is separated by column chromatography.
Further, 3, 4-bis ((t-butyldimethylsilyl) oxy) benzoate (III) may be prepared from a DMF solution of protocatechuic acid and imidazole, a DMF solution of t-butyldimethylsilyl chloride; in the preparation, the mol ratio of protocatechuic acid, imidazole and tertiary butyl dimethyl chlorosilane is (1-2) 1: (4-6), and when preparing DMF solution of protocatechuic acid and imidazole, the solution is firstly cooled to 0-1 ℃, the aim is to control the reaction temperature, avoid exothermic in the process of dripping tertiary butyl dimethyl chlorosilane, then react under the protection of nitrogen, the temperature is controlled to be optimal at 20-30 ℃, the reaction time is generally 20-30 h, after the reaction is finished, the reaction solution is firstly poured into 150-300mL of saturated sodium bicarbonate solution, then the ethyl ether is used for extraction, the organic phase is washed by saturated saline solution, then dried by anhydrous sodium sulfate, filtered and decompressed and concentrated, and white viscous liquid is obtained; then adding the viscous liquid into the mixed solution of tetrahydrofuran and methanol for dissolution, stirring and reacting with the aqueous solution of potassium carbonate (0.60-0.90 mol/L) at 20-30 ℃, concentrating the reaction liquid after the reaction is finished, then adding saturated saline for dilution, adjusting the pH value to 4-5 by using potassium bisulfate, extracting by using diethyl ether, washing the organic phase by using the saturated saline, drying by using anhydrous sodium sulfate, filtering, concentrating under reduced pressure, and drying in vacuum to obtain the product.
The method for synthesizing ((2R, 5S) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-dihydroxybenzoate (VI) in step 2 of the present invention comprises the steps of: the preparation method comprises the steps of taking (2R, 5S) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxypyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-di ((tert-butyldimethylsilyl) oxy) benzoate (V) and tetrabutylammonium fluoride tetrahydrofuran solution prepared in the step 1 as raw materials, reacting at 0-4 ℃ and separating and purifying.
In this step, (2 r,5 s) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-di ((tert-butyldimethylsilyl) oxy) benzoate and tetrabutylammonium fluoride tetrahydrofuran were in a molar ratio of 1: (3-4), after the reaction is completed at 0-4 ℃, more white granular solids appear in the bottle, methanol and/or ethanol are added for dissolution, then decompression concentration and residue column chromatography separation are carried out, thus obtaining the catalyst.
The method for synthesizing ((2R, 5S) -5- (4-amino-2-oxypyrimidin-1 (2H) -yl) -1, 3-oxathiolan-2-yl) -3, 4-dihydroxybenzoic acid methyl ester (I) in the step 3 comprises the following steps: the ((2R, 5S) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxypyrimidine-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-dihydroxybenzoate (VI), trifluoroacetic acid and chloroform synthesized in the step 2 are used as raw materials, reacted at 0-4 ℃ and separated and purified to obtain the compound.
In this step, the molar ratio of ((2 r,5 s) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-dihydroxybenzoate to trifluoroacetic acid is 1: (55-70).
The synthetic route is as follows:
the invention also aims to protect the application of the 3TC-PA compound prepared by the method in preparing medicines for treating CHB.
Drawings
FIG. 1 is the percent remaining of 3TC-PA in a solution of varying pH, wherein A: pure water, B: ph=4.0 buffer, C: ph=1.4 solution, D: ph=6.86 buffer, E: ph=7.80 buffer;
FIG. 2 is the percent remaining 3TC-PA in the artificial simulated gastrointestinal fluids, wherein A: artificial gastric juice, B: artificial intestinal juice, C: gastric content, D: intestinal content;
FIG. 3 is a graph showing the residual percentage of 3TC-PA in rat liver homogenates;
FIG. 4 is the percent remaining 3TC-PA in rat whole blood;
FIG. 5 is a graph showing the toxicity of CCK-8 assay on 3TC-PA HepG2.2.15 cells (n=3, 6);
fig. 6 is the inhibition of HBeAg antigen by 3TC-PA (n=3);
fig. 7 is the inhibition of HBsAg antigen by 3TC-PA (n=3);
FIG. 8 shows the inhibition of HBV-DNA by 3TC-PA (n=3).
Detailed Description
The present invention will be described in further detail with reference to specific examples so as to more clearly understand the present invention by those skilled in the art.
The following examples are given by way of illustration of the invention and are not intended to limit the scope of the invention. All other embodiments obtained by those skilled in the art without creative efforts are within the protection scope of the present invention based on the specific embodiments of the present invention.
In the examples of the present invention, all raw material components are commercially available products well known to those skilled in the art unless specified otherwise; in the embodiments of the present invention, unless specifically indicated, all technical means used are conventional means well known to those skilled in the art.
Example 1
This example provides a method for preparing a 3TC-PA compound bonded by an ester linkage, comprising the steps of:
step S1 Synthesis of tert-butyl (1- (((2R, 5S) -2- (hydroxymethyl) -1, 3-oxathiopyran-5-yl) -2-oxo-1, 2-dihydropyrimidin-4-yl) carbamate (II)
Lamivudine (119.8 mg,0.52 mmol) and di-tert-butyl dicarbonate (195.6 mg,0.90 mmol) were taken and anhydrous DMF (5 mL) was stirred in a 100mL single-neck flask at room temperature of 30℃for 24h in an oil bath. The reaction mixture was diluted with water, extracted with ethyl acetate, and the organic phase was dried over anhydrous sodium sulfate. The dried organic phase was filtered and separated by column chromatography (petroleum ether: ethyl acetate=1:4) under reduced pressure to give 92.70mg of intermediate (ii) as a white solid in 53.9% yield.
Analysis of the synthesized product structure by nuclear magnetic resonance spectroscopy proves that the prepared substance is truly of the structure shown in the formula (II), and the specific result is as follows:
1 H NMR(500MHz,CDCl 3 )δ8.31-8.32(d,1H),7.24-7.25(d,1H),6.32-6.34(dd,1H),5.35-5.36(dd,1H),4.12-4.15(dd,1H),3.94-3.97(dd,1H),3.60-3.63(dd,1H),3.20-3.23(dd,1H),1.52(s,9H)。
step S2 Synthesis of (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((t-butyldimethylsilyl) oxy) phenyl) methanone (IV)
The synthesis of (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((tert-butyldimethylsilyl) oxy) phenyl) methanone (IV) comprises two steps S2-1 and S2-2, specifically as follows:
step S2-1,3, 4-bis ((t-butyldimethylsilyl) oxy) benzoate (III) Synthesis
Protocatechuic acid (3.08 g,19.98 mmol) and imidazole (1.12 g,16.45 mmol) are taken and dissolved in anhydrous DMF (30 mL) and cooled to 0-1 ℃; t-butyldimethylchlorosilane (12.06 g,80.02 mmol) was taken and dissolved in anhydrous DMF (60 mL) and slowly added dropwise to the above solution under nitrogen protection; after the dripping is finished, heating to 25 ℃, and stirring for 24 hours; after the completion of the reaction, the reaction solution was poured into a saturated sodium hydrogencarbonate solution (200 mL), extracted with diethyl ether, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a white viscous liquid. The viscous liquid was used directly for the next hydrolysis without purification.
Adding a mixed solution of tetrahydrofuran (16 mL) and methanol (48 mL) to the viscous liquid for dissolution; then adding aqueous potassium carbonate solution (16 mL,0.72 mol/L) into the system, and stirring at room temperature for 24h; after the reaction, the reaction mixture was concentrated to one fourth of the original volume, and diluted with saturated brine (40 mL); adjusting pH to 4-5 with potassium bisulfate aqueous solution (1 mol/L), extracting with diethyl ether, washing the organic phase with saturated saline solution, and drying with anhydrous sodium sulfate; concentrated by filtration under reduced pressure, and dried in vacuo to give 7.37g of intermediate (III) as a white powder in 96.4% yield.
The synthesized product structure is analyzed through nuclear magnetic hydrogen spectrum characterization and carbon spectrum, and the prepared substance is proved to be the structure shown in the formula (III), and the specific result is as follows:
1 H NMR(400MHz,CDCl 3 )δ10.99(s,1H),7.60-7.65(m,2H),6.87-6.89(d,1H),0.99-1.02(d,18H),0.24-0.25(d,12H)。
13 C NMR(100MHz,CDCl 3 )δ172.33,152.66,146.94,124.60,122.90,122.64,120.69,77.48,77.16,76.84,26.05,25.99,18.66,18.60,0.14,-3.63,-3.71,-3.91,-3.99,-4.20,-4.28。
ESI-MS(m/z)[M-H] - C 19 H 33 O 4 Si 2 calculated values: 381.64, detection value: 381.
step S2-2 Synthesis of (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((t-butyldimethylsilyl) oxy) phenyl) methanone (IV)
Intermediate (III) (766 mg,2.00 mmol), benzotriazole (BTA, 257mg,2.16 mmol), 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI, 625mg,3.26 mmol), anhydrous dichloromethane (100 mL) were taken in a50 mL three port round bottom flask and reacted at 25℃under nitrogen. After 24h of reaction, the reaction mixture was concentrated under reduced pressure, and the residue was separated by column chromatography (petroleum ether: ethyl acetate=200:1) to give 251mg of intermediate (iv) as a white solid in 25.9% yield.
The synthesized product structure is analyzed through nuclear magnetic resonance spectrum characterization, and the prepared substance is proved to be truly the structure shown in the formula (IV), and the specific result is as follows:
1 H NMR(400MHz,CDCl 3 )δ8.36-8.38(dt,1H),8.15-8.17(dt,1H),7.85-7.88(dt,2H),7.66-7.70(m,1H),7.51-7.55(m,1H),6.98-7.00(m,1H),1.01-1.05(s,18H),0.27-0.29(s,12H)。
step S3 Synthesis of((2R, 5S) -5- (4- ((tert-Butoxycarbonyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-di ((tert-butyldimethylsilyl) oxy) benzoate (V)
Intermediate IV (48.4 mg,0.10 mmol), intermediate II (41.5 mg,0.13 mmol), DMAP (25.1 mg,0.21 mmol) and anhydrous dichloromethane (20 mL) were taken and reacted in a50 mL three-necked round bottom flask under nitrogen protection, after stirring at 25℃for 24h, the reaction solution was concentrated under reduced pressure and the residue was separated by column chromatography (petroleum ether: ethyl acetate=3:2) to give 26.2mg of intermediate (V) as a white solid in 37.8% yield.
The synthesized product structure is analyzed through nuclear magnetic hydrogen spectrum characterization and carbon spectrum, and the prepared substance is proved to be the structure shown in the formula (V), and the specific result is as follows:
1 H NMR(400MHz,CDCl 3 )δ8.09-8.11(d,1H),7.53-7.56(d,2H),7.13-7.15(d,1H),6.87-6.90(m,1H),6.32-6.34(dd,1H),5.47-5.49(dd,1H),4.66-4.76(m,2H),3.61-3.66(dd,1H),3.18-3.22(dd,1H),1.48(s,9H),0.96-0.97(t,18H),0.19-0.22(m,12H)。
13 C NMR(100MHz,CDCl 3 )δ165.63,162.85,154.71,152.36,151.01,147.04,143.86,123.63,122.50,122.23,120.62,94.68,87.84,85.03,82.71,77.42,77.30,77.10,76.78,63.71,39.08,29.69,28.00,25.88,25.84,18.50,18.42,0.01,-3.75,-3.83,-4.04,-4.12,-4.32,-4.41,-5.38。
step S4 Synthesis of((2R, 5S) -5- (4- ((tert-Butoxycarbonyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-dihydroxybenzoate (VI)
A1M solution of tetrabutylammonium fluoride in tetrahydrofuran (1.59 mL,1.59 mmol) was taken, intermediate V (277.6 mg,0.4 mmol) was placed in a single-necked round bottom flask, and after 24 hours of reaction at 0-4 ℃, a large amount of white granular solid appeared in the flask, after dissolution with methanol, the reaction solution was concentrated under reduced pressure, and the residue was separated by column chromatography (dichloromethane: methanol=20:1) to give 168.1mg of white solid, intermediate (VI), in 90.3% yield.
Step S5 Synthesis of methyl (((2R, 5S) -5- (4-amino-2-oxopyrimidin-1 (2H) -yl) -1, 3-oxathiolan-2-yl) -3, 4-dihydroxybenzoate (I)
Intermediate vi (381.8 mg,0.82 mmol), trifluoroacetic acid (4 mL,52.24 mmol) and anhydrous dichloromethane (12 mL) were taken and reacted in a single neck flask at 0-4 ℃ for 24h, the reaction solution was concentrated under reduced pressure, and the residue was separated by column chromatography (dichloromethane: methanol=8:1). 153mg of intermediate (I) as a white solid was obtained in a yield of 51.1%.
The synthesized product structure is analyzed through nuclear magnetic hydrogen spectrum characterization and carbon spectrum, and the prepared substance is proved to be the structure shown in the formula (I), and the specific result is as follows:
1 H NMR(400MHz,MeOD-d 4 )δ8.01-8.03(d,1H),7.41-7.43(d,2H),6.81-6.83(d,1H),6.28(s,1H),5.78-5.80(d,1H),5.49(s,1H),4.65-4.73(d,2H),3.54-3.59(dd,1H),3.25(s,1H)。
13 C NMR(100MHz,MeOD-d 4 )δ167.39,164.50,152.16,146.37,143.81,123.88,121.86,117.54,116.09,95.53,88.50,86.40,64.36,49.64,49.43,49.21,49.00,48.79,48.57,48.36,39.04。
example 2
This example provides a method for preparing a 3TC-PA compound bonded by an ester linkage, comprising the steps of:
step S1 Synthesis of tert-butyl (1- (((2R, 5S) -2- (hydroxymethyl) -1, 3-oxathiopyran-5-yl) -2-oxo-1, 2-dihydropyrimidin-4-yl) carbamate (II)
Lamivudine (2.40 g,10.47 mmol) and di-tert-butyl dicarbonate (3.92 g,17.96 mmol) were put in a 200mL single-neck flask, anhydrous DMF (100 mL) was added, and the mixture was fixed in an oil bath at room temperature of 30℃and stirred for 24 hours. The reaction mixture was diluted with distilled water, extracted with ethyl acetate, and the organic phase was dried over anhydrous sodium sulfate. The dried organic phase was filtered and separated by column chromatography (petroleum ether: ethyl acetate=1:4) under reduced pressure to give 2.13g of intermediate (ii) as a white solid in 61.8% yield.
The nuclear magnetic resonance spectrum characterization result is the same as in example 1.
Step S2 Synthesis of (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((t-butyldimethylsilyl) oxy) phenyl) methanone (IV)
The synthesis of (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((tert-butyldimethylsilyl) oxy) phenyl) methanone (IV) comprises two steps S2-1 and S2-2, specifically as follows:
step S2-1,3, 4-bis ((t-butyldimethylsilyl) oxy) benzoate (III) Synthesis
Protocatechuic acid (3.08 g,19.98 mmol) and imidazole (1.12 g,16.45 mmol) are taken and dissolved in anhydrous DMF (30 mL) and cooled to 0-1 ℃; t-butyldimethylchlorosilane (12.06 g,80.02 mmol) was taken and dissolved in anhydrous DMF (60 mL) and slowly added dropwise to the above solution under nitrogen protection; after the dripping is finished, heating to 25 ℃, and stirring for 24 hours; after the completion of the reaction, the reaction solution was poured into a saturated sodium hydrogencarbonate solution (200 mL), extracted with diethyl ether, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a white viscous liquid. The viscous liquid was used directly for the next hydrolysis without purification.
Adding a mixed solution of tetrahydrofuran (16 mL) and methanol (48 mL) to the viscous liquid for dissolution; then adding aqueous potassium carbonate solution (16 mL,0.72 mol/L) into the system, and stirring at room temperature for 24h; after the reaction, the reaction mixture was concentrated to one fourth of the original volume, and diluted with saturated brine (40 mL); adjusting pH to 4-5 with potassium bisulfate aqueous solution (1 mol/L), extracting with diethyl ether, washing the organic phase with saturated saline solution, and drying with anhydrous sodium sulfate; concentrated by filtration under reduced pressure, and dried in vacuo to give 7.37g of intermediate (III) as a white powder in 96.4% yield.
The nuclear magnetic hydrogen spectrum characterization and the carbon spectrum characterization result are as in example 1.
Step S2-2 Synthesis of (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((t-butyldimethylsilyl) oxy) phenyl) methanone (IV)
Intermediate (III) (3.83 g,10.01 mmol), benzotriazole (BTA, 1.29g,10.83 mmol), 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI, 3.13g,16.33 mmol), dichloromethane (200 mL) were taken in a 250mL three port round bottom flask and reacted at room temperature under nitrogen. After 24h of reaction, the reaction mixture was concentrated under reduced pressure, and the residue was separated by column chromatography (petroleum ether: ethyl acetate=200:1) to give 1.48g of intermediate (iv) as a white solid with a yield of 30.6%.
The nuclear magnetic resonance spectrum is characterized as in example 1.
Step S3 Synthesis of((2R, 5S) -5- (4- ((tert-Butoxycarbonyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-di ((tert-butyldimethylsilyl) oxy) benzoate (V)
Intermediate IV (1.45 g,3.00 mmol), intermediate II (1.25 g,3.80 mmol), DMAP (753 mg,6.16 mmol) and anhydrous dichloromethane (200 mL) were taken and reacted in a 250mL three-necked round bottom flask under nitrogen protection, after stirring at 25 ℃ for 24h, the reaction solution was concentrated under reduced pressure and the residue was separated by column chromatography (petroleum ether: ethyl acetate=3:2) to give 0.89g of intermediate (V) as a white solid with a yield of 42.7%.
The nuclear magnetic hydrogen spectrum characterization and the carbon spectrum characterization result are as in example 1.
Step S4 Synthesis of((2R, 5S) -5- (4- ((tert-Butoxycarbonyl) amino) -2-oxopyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-dihydroxybenzoate (VI)
A1M solution of tetrabutylammonium fluoride in tetrahydrofuran (3.18 mL,3.18 mmol) was taken, intermediate V (555 mg,0.8 mmol) was placed in a single-necked round-bottomed flask, and after 24 hours of reaction at 0 to 4℃more white granular solid appeared in the flask, after dissolution with methanol, the reaction mixture was concentrated under reduced pressure and the residue was separated by column chromatography (dichloromethane: methanol=20:1) to give 340mg of intermediate (VI) as a white solid in 91.3% yield.
Step S5 Synthesis of methyl (((2R, 5S) -5- (4-amino-2-oxopyrimidin-1 (2H) -yl) -1, 3-oxathiolan-2-yl) -3, 4-dihydroxybenzoate (I)
Intermediate vi (381.8 mg,0.82 mmol), trifluoroacetic acid (4 mL,52.24 mmol) and anhydrous dichloromethane (12 mL) were taken and reacted in a single neck flask at 0-4 ℃ for 24h, the reaction solution was concentrated under reduced pressure, and the residue was separated by column chromatography (dichloromethane: methanol=8:1). 153mg of intermediate (I) as a white solid was obtained in a yield of 51.1%.
The nuclear magnetic hydrogen spectrum characterization and the carbon spectrum characterization result are as in example 1.
Example 3 in vitro stability test
(1) Stability of 3TC-PA in solutions of different pH
0.00541g of the 3TC-PA compound prepared in example 1 was dissolved in methanol (1 mg/mL), 100. Mu.L of the compound was taken and each of the pH mediums was fixed to a volume of 10mL, the solution was placed at 37℃and sampled and examined at 0, 30, 60, 120, 240, 360, 480, 720 and 1440 minutes after incubation, and the percentage of 3TC-PA in each pH medium was as shown in FIG. 1, wherein A: pure water; b: ph=4.0 buffer; c: ph=1.4 solution; d: ph=6.86 buffer; e: ph=7.80 buffer
Wherein the ph=4.0 buffer has the following composition (or preparation method): accurate weighing CH 3 COONa1.8000g, dissolved to a volume of 100mL, and adjusted to pH 4.0 with glacial acetic acid.
The composition (or preparation method) of the ph=1.4 solution is: accurately measuring 0.90mL of concentrated hydrochloric acid, and adding water to fix the volume to 100mL.
The composition (or preparation method) of the ph=6.86 buffer is: 0.6800g of potassium dihydrogen phosphate is precisely weighed, dissolved in water, and then fixed to 100mL of water, and the pH value is adjusted to 6.8 by 0.1mol/L of sodium hydroxide solution.
The composition (or preparation method) of the ph=7.80 buffer is: 0.5590g of dipotassium hydrogen phosphate and 0.0410g of monopotassium hydrogen phosphate are precisely weighed, and water is added to dissolve the dipotassium hydrogen phosphate and the monopotassium hydrogen phosphate to reach 100mL.
The experimental result shows that the pH=1.4 of the 3TC-PA has no decomposition within 8 hours, the content of the 3TC-PA is reduced by 30% compared with the content before incubation for 12-24 hours, the 3TC-PA is stable in pure water and other pH buffer media, and no obvious decomposition phenomenon (degradation rate is less than 10%) is observed.
(2) Stability of 3TC-PA in artificially simulated gastrointestinal fluids
The in vitro stability of 3TC-PA in the gastrointestinal tract was examined and the percentage of 3TC-PA content was quantitatively analyzed at various time points in each medium. The specific operation is as follows:
precisely measuring 1mg/mL of 3TC-PA solution, taking 100 mu L of artificial gastric juice, artificial intestinal juice, gastric content and intestinal content to constant volume to 10mL, incubating at 37 ℃, sampling and detecting the gastric content, the small intestinal content and the artificial gastric juice for 0, 0.5, 1,2, 4, 6 and 8 hours after incubating, and sampling and detecting the artificial intestinal juice for 0, 1,2, 4, 8, 12 and 24 hours after incubating.
The percent 3TC-PA content versus time for the various media is shown in FIG. 2, where A: artificial gastric juice; b: artificial intestinal juice; c: gastric contents; d: intestinal contents.
The experimental result shows that 3TC-PA has no obvious decomposition (degradation rate is less than 10%) within 4 hours after incubation in gastric contents, has little decomposition (10% -16%) within 6-8 hours, has no obvious decomposition (degradation rate is less than 10%) in artificial gastric juice, artificial intestinal juice and intestinal contents, and is relatively stable.
(3) In vitro stability of 3TC-PA in rat liver homogenates
The in vitro stability of 3TC-PA in liver homogenates was examined, and the percentage of 3TC-PA content at different time points of incubation of liver homogenates was quantitatively analyzed. The specific operation is as follows:
precisely transferring 250 μl of 4mg/mL 3TC-PA methanol solution, adding blank liver homogenate to 5mL, shaking to mix thoroughly, incubating at 37deg.C, sampling and detecting 0, 1,2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24 hr after incubation.
The percentage of 3TC-PA in the liver homogenate versus time is shown in FIG. 3. The experimental result shows that 3TC-PA has no obvious decomposition within 6 hours (degradation rate < 10%) and has a small decomposition within 6-24 hours (degradation rate < 20%) in rat liver homogenate.
(4) In vitro stability of 3TC-PA in rat Whole blood
The in vitro stability of 3TC-PA in rat whole blood was examined, and the percent 3TC-PA content versus time plot at various time points of the whole blood incubation is shown in FIG. 4. The specific operation is as follows:
precisely transferring 4mg/mL of 3TC-PA methanol solution, adding blank whole blood to 5mL, mixing by shaking, and incubating at 37deg.C for 0, 1,2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, and 24 hr.
The 3TC-PA content was almost unchanged in the whole rat blood for 24 hours, indicating that the 3TC-PA was more stable in the whole rat blood.
The in vitro stability experiment of the 3TC-PA shows that the ester bond-bonded 3TC-PA is a compound which exists relatively stably in vivo.
Example 4 evaluation of in vitro anti-HBV Activity Effect
4.1 experimental materials:
hepg2.2.15 cells: the cells were given away by the national emphasis laboratory of university of martial arts virology and stored by the present laboratory.
pHBV1.3 plasmid: the plasmid is presented by the national emphasis laboratory of university of martial arts virology and amplified and stored by the laboratory.
DMEM medium: purchased from Gibco company, usa.
Serum: purchased from PAN company, germany.
Penicillin, streptomycin, pancreatin: purchased from Hangzhou Jino biomedical technologies Inc.
CCK-8 reagent: purchased from japan homozygote.
HBV-DNA primer: purchased from wuhan hundred biotechnology limited.
Adefovir dipivoxil (Adv): purchased from Shanghai Dingrui chemical Co.
Viral DNA/RNA extraction kit: purchased from Beijing kang as century biotechnology Co.
Human hepatitis b surface antigen (HBsAg) enzyme-linked immunoassay (ELISA) kit, human hepatitis b E antigen (HBeAg) enzyme-linked immunoassay (ELISA) kit: purchased from Xiamen Huija biological Co., ltd.
4.2 laboratory apparatus:
HEARCell CO2 incubator: purchased from Heraeus company, germany.
Level II biosafety cabinet: purchased from the su-net group su-state antai air technologies inc.
-80 ℃ low temperature refrigerator, 4 ℃ medical refrigerator: purchased from Haier company, shandong.
Real-time fluorescence quantitative analyzer: purchased from BIO-RAD corporation, usa.
ENVISION multifunctional enzyme labeling instrument: purchased from us PERKINELMER company.
5415R desk-top low temperature high speed centrifuge: purchased from Eppendorf, germany.
4.3 detection of cell viability (CCk-8 method)
4.3.1 preparation of cell samples
(1) Taking a culture flask with good HepG2.2.15 cell growth state, taking out proper amount of cells from the culture flask after proper digestion and dilution, inoculating the cells into a 96-well cell culture plate, and placing the cells in CO 2 Culturing in an incubator. When the cell growth density reached 50%, 96-well cell culture plates were removed, the medium in each well was aspirated, and 100. Mu.L of 3TC-PA prodrug solutions (0, 5, 10, 20, 40, 80, 160. Mu.M) of different concentrations were added to the 96-well plates (0. Mu.M concentration of the solution was used as a negative control, and the DMSO content in the culture solution was 0.1%). 5 duplicate wells were set for each concentration and blank.
(2) The culture plate is incubated in an incubator for 3 and 6 days, the liquid medicine is replaced on the 3 rd day, and when the liquid medicine is replaced, the original liquid medicine is firstly sucked and then 100 mu L of fresh liquid medicine with corresponding concentration is replaced. This experiment was repeated 3 times. 4.3.2CCK-8 method for detecting cytotoxicity
(1) After the incubation of the liquid in the incubator for 6 days, 10. Mu. LCCK-8 reagent was directly added to each well, and care was taken not to generate bubbles in the wells during the procedure, which would otherwise affect the OD readings.
(2) Placing the culture plate in CO 2 Incubating for 30min in an incubator.
(3) Absorbance values at 450nm were measured with a microplate reader.
(4) The absorbance value was positively correlated with cell viability compared to the negative control, with higher absorbance values indicating higher cell viability.
(5) Cell viability was calculated as follows:
CellviabilityLiving (%) = (OD sample-OD blank)/(OD control-OD blank) ×100%.
4.4 preparation of cell samples
(1) A culture flask in which hepg2.2.15 cells grew well was taken, and after proper digestion and dilution, appropriate amount of cells were taken out of the flask and inoculated into a 12-well plate. The 12-well plate was then placed in CO 2 Culturing in an incubator, and starting administration when the cell density grows to 50%.
(2) According to the results of the previous cytotoxicity test, the concentration of the drug is selected to be low, medium and high by 3 in the non-toxic concentration of the drug to cells. 3TC-PA prodrug and 3TC liquid medicine 1mL (serum content in culture solution is 3%, DMSO content is 0.1%) with different concentrations are added into 12-well plate cells, cell wells with only culture medium (containing 0.1% DMSO) are set as virus controls, and Adv (10 μm) is added as positive drug controls. And 3 multiple holes are provided each. CO at 37 DEG C 2 The culture was performed in the incubator for 6 days, and the liquid medicine was replaced every 3 days.
(3) Collecting the medicine cell culture solution replaced in the experimental process, centrifuging for 20min at 3000r/min, collecting the supernatant, marking, and transferring to a refrigerator at-80 ℃ for storage for later use.
4.5ELISA method for detecting expression of HBeAg and HBsAg in cell supernatant
After removal of the ELISA kit from the 4℃chromatography cabinet, equilibration was performed by standing at room temperature for 20 minutes. At the same time, the cell culture solution is rapidly taken out from a refrigerator at-80 ℃, thawed under a low-temperature environment, and then vibrated uniformly for standby (the operation steps of HBeAg and HBsAg antigens are the same).
(1) Dilution and sample addition of standard: setting a standard substance hole 10 holes on an enzyme-labeled coating plate, respectively adding 100 mu L of standard substance into the first hole and the second hole, then adding 50 mu L of standard substance diluent into the first hole and the second hole, and uniformly mixing; then 100 mu L of each of the first hole and the second hole is added into a third hole and a fourth hole respectively, 50 mu L of standard substance diluent is added into the third hole and the fourth hole respectively, and the mixture is uniformly mixed; then 50 mu L of each of the third hole and the fourth hole is discarded, 50 mu L of each of the third hole and the fourth hole is added into the fifth hole and the sixth hole respectively, 50 mu L of standard substance diluent is added into the fifth hole and the sixth hole respectively, and the mixture is uniformly mixed; mixing, adding 50 μL of each of the fifth and sixth holes into the seventh and eighth holes, adding 50 μL of each of the standard substance diluents into the seventh and eighth holes, mixing, adding 50 μL of each of the seventh and eighth holes into the ninth and tenth holes, adding 50 μL of each of the standard substance diluents into the ninth and tenth holes, mixing, and discarding 50 μL of each of the ninth and tenth holes. (50. Mu.L of each well after dilution, and 54IU/L,36IU/L,18IU/L,9IU/L, and 4.5IU/L of each well were added).
(2) Sample adding: blank holes (blank control holes are not added with samples and enzyme-labeled reagents, and the rest steps are the same) and sample holes to be tested are respectively arranged, and each sample is provided with 2 repeats. The sample dilution liquid is added into 40 mu L of the sample to be detected in the sample hole on the enzyme-labeled coated plate, and then 10 mu L of the sample to be detected is added (the final dilution of the sample is 5 times). And (3) adding a sample to the bottom of the ELISA plate hole, so as not to touch the hole wall as much as possible, and slightly shaking and uniformly mixing.
(3) Incubation: the plates were then covered with a plate membrane and incubated at 37℃for 30 minutes.
(4) Preparing liquid: the 30 (20 times of 48T) concentrated washing solution was diluted with distilled water 30 (20 times of 48T) for use.
(5) Washing: carefully removing the sealing plate film, discarding the liquid, spin-drying, filling each hole with the washing liquid, standing for 30 seconds, discarding, repeating the process for 5 times, and beating.
(6) Adding enzyme: 50. Mu.L of enzyme-labeled reagent was added to each well, except for blank wells.
(7) Incubation: the operation is the same as 3.
(8) Washing: the operation is the same as 5.
(9) Color development: 50 mu L of a color developing agent A and 50 mu L of a color developing agent B are added into each hole, the mixture is gently vibrated and mixed uniformly, and the color is developed for 15 minutes at 37 ℃ in a dark place.
(10) And (3) terminating: the reaction was stopped by adding 50. Mu.L of stop solution to each well (blue turned yellow immediately).
(11) And (3) measuring: the absorbance (OD value) of each well was measured sequentially at the wavelength of Kong Diaoling blank, 450 nm.
(12) Drawing a standard curve by adopting the gradient concentration and the OD value of the standard substance, bringing the OD value of the sample into the standard curve, and solving the actual concentration of HBeAg and HBsAg antigens in the sample.
4.6qRT-PCR detection of HBV-DNA expression
After removal of the ELISA kit from the 4℃chromatography cabinet, equilibration was performed by standing at room temperature for 20 minutes. And at the same time, the cell culture solution is quickly taken out from a refrigerator at the temperature of-80 ℃, thawed in a low-temperature environment, and then vibrated uniformly for standby.
4.6.1 concentration determination
Detection was performed using an ultra-trace biological detector: DNA solution (2. Mu.L) was sampled and then its purity and concentration (including concentration, OD) 260 /OD 230 OD (optical density) 260 /OD 280 )。
4.6.2SYBRGreen real-time fluorescent quantitative PCR reactions
(1) Primer: HBVForwardprimer: AGAAACAACACATAGCGCCTCAT;
HBVReverseprimer:TGCCCCATGCTGTAGATCTTG。
(2) The reaction system comprises: three times of water (8. Mu.L), SYBRGreenSupermix (10. Mu.L), the prepared upstream and downstream primers (1. Mu.L) and DNA template (1. Mu.L) were sterilized and the total volume was 20. Mu.L.
(3) The mixture ratio is carried out according to the reaction system, and then all materials are added to the bottom of a 96-well plate special for PCR and are uniformly mixed. 3 auxiliary holes are respectively arranged, the sealing plate films are attached, and then the sealing plate films are put into Q-PCR to collect signals. The amplification conditions were: pre-denaturation at 95℃for 5min, 95℃for 15s, 58℃for 15s, 72℃for 30s for 40 cycles. Signal collection was set as SYBR fluorescence channel.
(4) According to the absolute quantification method, the negative logarithm of the concentration of the pHBV1.3 plasmid after equal gradient dilution is used for drawing a standard curve equation of the CT value of the plasmid.
(5) Substituting the CT value of the sample to be measured into an equation, then calculating to obtain the initial copy number of the sample, and finally processing the result.
4.6 experimental results and analysis
4.6.1CCK-8 method for detecting cytotoxicity of 3TC-PA
The CCK-8 method is a relatively convenient and accurate method for detecting cell growth and survival. The non-toxic concentration of 3TC-PA prodrug was screened by 3 and 6 days of 3TC-PA on HepG2.2.15 cells, as shown in FIG. 5, the concentration of 3TC-PA was between 0 and 160. Mu.M at 3 and 6 days of 3TC-PA treatment, and no obvious toxicity was observed on proliferation of HepG2.2.15 cells, so 10, 20 and 40. Mu.M were selected as the concentrations for the subsequent drug activity test at 3 and 6 days of 3TC-PA treatment.
4.6.2 Inhibition of HBsAg and HBeAg antigens by 3TC-PA
HBsAg is a surface protein of hepatitis B virus, which is one of the markers for detecting hepatitis B virus infection. And HBeAg is a soluble protein in the core particle of hepatitis B virus, and is a second serological antigen marker after HBV infection because HBeAg appears later than HBsAg but disappears earlier than HBsAg. In the experiment, hepG2.2.15 cells are treated for 3 days and 6 days by adopting 3TC-PA with different concentrations, and after culture solution supernatant is collected, the concentrations of HBeAg and HBsAg in the cell supernatant are measured by adopting an ELISA kit. The results are shown in FIGS. 6 and 7, and the 3TC-PA has a certain inhibition effect on HBeAg and HBsAg antigen in experiments of 3 and 6 days, but the inhibition effect is not significantly different from that of the 3TC with the same concentration.
4.6.3 Inhibition of HBV-DNA by 3TC-PA
HBV-DNA is the most direct index for detecting hepatitis B virus infection, and the index has the characteristics of strong specificity and high sensitivity. In the experiment, the effect of 3TC-PA on the absolute copy number of HBV-DNA was examined by treating HepG2.2.15 cells with different concentrations of 3TC-PA for 3,6 days. As shown in fig. 8, 3TC-PA had a higher inhibition rate for HBV-DNA, but the inhibition rate was not significantly different from that of 3TC at the same concentration. The results indicate that 3TC-PA still retains the anti-HBV-DNA replication capacity of 3 TC.
It should be noted that the above examples are only for further illustrating and describing the technical solution of the present invention, and are not intended to limit the technical solution of the present invention, and the method of the present invention is only a preferred embodiment and is not intended to limit the scope of the present invention. Any modification, equivalent replacement, improvement, etc. made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (6)

1. A 3TC-PA compound bonded by an ester bond, wherein the structural formula of the compound is represented by formula (i):
2. the method for producing a 3TC-PA compound according to claim 1, characterized by comprising the steps of:
step 1, synthesizing ((2 r,5 s) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxapyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-di ((tert-butyldimethylsilyl) oxy) benzoate) starting from tert-butyl (1- (((2 r,5 s) -2- (hydroxymethyl) -1, 3-oxathiopyran-5-yl) -2-oxo-1, 2-dihydropyrimidin-4-yl) carbamate, (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((tert-butyldimethylsilyl) oxy) phenyl) methanone and 4-dimethylaminopyridine;
step 2, taking (2R, 5S) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxypyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-di ((tert-butyldimethylsilyl) oxy) benzoate and tetrabutylammonium fluoride tetrahydrofuran solution prepared in the step 1 as raw materials, reacting at 0-4 ℃ and separating and purifying to obtain ((2R, 5S) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxypyrimidin-1-1 (2H) -yl) -1, 3-oxathiolan-2-yl) methyl-3, 4-dihydroxybenzoate;
step 3, using ((2R, 5S) -5- (4- ((tert-butoxycarbonyl) amino) -2-oxo pyrimidine-1-1 (2H) -group) -1, 3-oxathiolan-2-group) methyl-3, 4-dihydroxybenzoate, trifluoroacetic acid and dichloromethane synthesized in step 2 as raw materials, reacting at 0-4 ℃ and separating and purifying to obtain ((2R, 5S) -5- (4-amino-2-oxo pyrimidine-1 (2H) -group) -1, 3-oxathiolan-2-group) -3, 4-dihydroxybenzoic acid methyl ester.
3. The process for preparing a 3TC-PA compound according to claim 2 wherein in step 1, the process for preparing tert-butyl (1- (((2 r,5 s) -2- (hydroxymethyl) -1, 3-oxathiopyran-5-yl) -2-oxo-1, 2-dihydropyrimidin-4-yl) carbamate is as follows:
tert-butyl (1- (((2R, 5S) -2- (hydroxymethyl) -1, 3-oxathiopyran-5-yl) -2-oxo-1, 2-dihydropyrimidin-4-yl) carbamate is prepared by using lamivudine and di-tert-butyl dicarbonate and anhydrous DMF as raw materials.
4. The method for producing a 3TC-PA compound according to claim 2 wherein said method for producing (1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((t-butyldimethylsilyl) oxy) phenyl) methanone in step 1 is as follows:
(1H-benzo [ d ] [1,2,3] triazol-1-yl) (3, 4-di ((tert-butyldimethylsilyl) oxy) phenyl) methanone was prepared from 3, 4-bis ((tert-butyldimethylsilyl) oxy) benzoate, benzotriazole, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride as a raw material.
5. The method for producing a 3TC-PA compound according to claim 4, wherein the method for producing 3, 4-bis ((t-butyldimethylsilyl) oxy) benzoate is as follows:
the 3, 4-bis ((tert-butyldimethylsilyl) oxy) benzoate is prepared by taking a DMF solution of protocatechuic acid and imidazole and a DMF solution of tert-butyldimethylsilyl chlorosilane as raw materials.
6. Use of a 3TC-PA compound as claimed in claim 1 or a 3TC-PA compound as prepared according to any one of claims 2 to 5 in the manufacture of a medicament for the treatment of CHB.
CN202210777474.1A 2022-06-30 2022-06-30 3TC-PA compound bonded through ester bonds and application thereof Active CN115181096B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210777474.1A CN115181096B (en) 2022-06-30 2022-06-30 3TC-PA compound bonded through ester bonds and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210777474.1A CN115181096B (en) 2022-06-30 2022-06-30 3TC-PA compound bonded through ester bonds and application thereof

Publications (2)

Publication Number Publication Date
CN115181096A CN115181096A (en) 2022-10-14
CN115181096B true CN115181096B (en) 2024-03-19

Family

ID=83516383

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210777474.1A Active CN115181096B (en) 2022-06-30 2022-06-30 3TC-PA compound bonded through ester bonds and application thereof

Country Status (1)

Country Link
CN (1) CN115181096B (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101224209A (en) * 2008-01-25 2008-07-23 湖北大学 Novel anti- hepatitis B virus medicine compounds-lamivudine and protocatechuic acid

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2697238B1 (en) * 2011-04-08 2019-03-20 Laurus Labs Limited Solid forms of antiretroviral compounds, process for the preparation and their pharmaceutical composition thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101224209A (en) * 2008-01-25 2008-07-23 湖北大学 Novel anti- hepatitis B virus medicine compounds-lamivudine and protocatechuic acid

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
原儿茶酸对Huh7细胞中HNF1α表达的抑制影响;代小青;刘纯;陈勇;韩凤梅;;湖北大学学报(自然科学版)(第04期);第37-38页 *
拉米夫定与原儿茶酸药物组合体内抗鸭乙肝病毒;潘琪;王玲;王俊俊;陈勇;韩凤梅;;中华中医药杂志(第11期);第2500-2504页 *
拉米夫定联合原儿茶酸对鸭乙肝病毒的体外抑制;王玲;潘琪;孟沫然;陈勇;韩凤梅;;中华中医药杂志(第09期);第2308-2313页 *

Also Published As

Publication number Publication date
CN115181096A (en) 2022-10-14

Similar Documents

Publication Publication Date Title
US8835615B2 (en) 2′-fluorine-4′-substituted-nucleoside analogues, preparation methods and uses thereof
US8633309B2 (en) Nucleoside phosphoramidates
US9284342B2 (en) Nucleoside phosphoramidates
JP2024503755A (en) Nucleoside compounds for the treatment of viral infections and their uses
CN108530449B (en) Compound for treating or preventing hepatitis B virus infection and preparation method and application thereof
CN111196801B (en) Aporphine alkaloid derivative and preparation method and application thereof
JP2022549923A (en) Crystal forms of N-hetero pentacyclic ring-containing capsid protein assembly inhibitors and uses thereof
CN115181096B (en) 3TC-PA compound bonded through ester bonds and application thereof
CN114315855A (en) Curcumenol derivatives, preparation method and application thereof in preparation of anti-inflammatory drugs
CN108727403B (en) Nodosin derivative and preparation method and application thereof
TWI742419B (en) Crystal form of tricyclic compounds and application thereof
CN110204539A (en) A kind of dihydropyridine prodrug and its preparation method and application
CN103508920B (en) A kind of preparation method and its usage of optical isomer of phenylalaninol compound
CN107382944B (en) Coumarin gossypol derivatives with anti-tumor activity and synthesis method thereof
CN108623630A (en) Deuterated nucleotide analog and application thereof
US11807647B2 (en) Crystal form of hepatitis B surface antigen inhibitor and application thereof
CN112110880B (en) Androstane derivative and preparation method and application thereof
CN105130896B (en) The naphthalimide derivative of a kind of substituent containing thiocarbamide, its preparation method and application
CN115304605B (en) Oxetane derivatives with antitumor activity, and preparation method and application thereof
CN108864130A (en) Enmein derivative and its preparation method and application
CN115141206A (en) Alpha-lipoic acid lycorine conjugate and preparation method and application thereof
CN112679489B (en) N-sulfonyl heterocyclic derivative and pharmaceutical use thereof
CN110437264B (en) Homocamptothecin 5, 6-dibromo norcantharidinate derivative and regioselective synthesis method thereof
CN109824655B (en) Andrographolide compound and preparation method and application thereof
CN111018885B (en) 1, 2-dioxycyclohexene [3,4-f ] nitrogen oxo cyclononane derivative and synthetic 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