CN112604002A - Disulfide-bond bridged docetaxel-fatty acid prodrug and self-assembled nanoparticles thereof - Google Patents

Disulfide-bond bridged docetaxel-fatty acid prodrug and self-assembled nanoparticles thereof Download PDF

Info

Publication number
CN112604002A
CN112604002A CN202011492954.0A CN202011492954A CN112604002A CN 112604002 A CN112604002 A CN 112604002A CN 202011492954 A CN202011492954 A CN 202011492954A CN 112604002 A CN112604002 A CN 112604002A
Authority
CN
China
Prior art keywords
acid
docetaxel
prodrug
disulfide
bridged
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202011492954.0A
Other languages
Chinese (zh)
Inventor
孙进
王悦全
何仲贵
王永军
邱诗
罗聪
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.)
Suzhou Yutai Pharmaceutical Technology Co ltd
Original Assignee
Suzhou Yutai Pharmaceutical Technology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Suzhou Yutai Pharmaceutical Technology Co ltd filed Critical Suzhou Yutai Pharmaceutical Technology Co ltd
Publication of CN112604002A publication Critical patent/CN112604002A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/542Carboxylic acids, e.g. a fatty acid or an amino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Abstract

The invention designs and synthesizes a series of reduction-sensitive disulfide bond bridged docetaxel-fatty acid prodrugs, and the prodrugs can be self-assembled into a nano drug delivery system by a one-step nano precipitation method. The nano drug delivery system has the following advantages: docetaxel is prepared into a prodrug connected by disulfide bonds, so that the systematic toxicity of docetaxel is reduced, and the parent drug can be specifically released in a high-reduction environment in tumor cells, so that the effects of synergy and attenuation are realized; the drug loading is high, the use of a solubilizer with high toxicity is avoided, and the tolerance and compliance of patients are expected to be improved; the preparation method is simple in preparation process and easy for large-scale production by a one-step nano precipitation method; the nanoparticles have small and uniform particle size and are easy to enrich in tumor parts through an EPR effect; is easy to surface modify, and can be modified by PEG and the like to slow down the clearance of the reticuloendothelial system.

Description

Disulfide-bond bridged docetaxel-fatty acid prodrug and self-assembled nanoparticles thereof
Technical Field
The invention belongs to the field of new auxiliary materials and new dosage forms of medicinal preparations, and relates to a disulfide bond bridged docetaxel-fatty acid prodrug, construction of self-assembled nanoparticles of the disulfide bond bridged docetaxel-fatty acid prodrug, and application of the disulfide bond bridged docetaxel-fatty acid prodrug in medicament delivery.
Background
Cancer remains one of the diseases with high mortality rate today, seriously threatening human health. Common cancer treatment regimens include primarily surgical resection, chemotherapy, radiation therapy, and biological therapy. Among them, surgical resection has significant efficacy for early stage tumors, but chemotherapy has significant advantages for late stage or metastatic tumors. Taxane chemotherapeutic drugs are a class of antineoplastic drugs acting on microtubules, can promote the rapid aggregation of tubulin into microtubules, and bind to the microtubules to inhibit the depolymerization of the microtubules, thereby terminating the mitosis of the cells and inducing the apoptosis of tumor cells, and represent drugs of Paclitaxel (PTX) and Docetaxel (DTX). The research proves that DTX shows outstanding curative effect in the chemotherapy treatment of tumors regardless of single medicine or combined medicine. The DTX injection Taxotere (Taxotere) applied to clinic at present has good anti-tumor activity, is mainly applied to the treatment of advanced or metastatic breast cancer and non-small cell lung cancer, but is easy to generate systemic toxicity such as anaphylactic reaction, hemolysis and the like, and seriously influences the clinic application of the DTX injection Taxotere (Taxotere).
In recent years, prodrug strategies and nanotechnology have been widely used to reduce the systemic toxicity of taxanes and to improve drug potency. Wherein, the nanometer preparation of paclitaxel comprises paclitaxel liposome, paclitaxel albumin nanometer particle, etc. which are already applied to the clinical treatment of cancer; paclitaxel-docosahexaenoic acid prodrugs have also been subjected to phase III clinical studies, but docetaxel-based nano-dosage forms have not been clinically applied. At present, a stimulus-responsive drug delivery system based on a tumor microenvironment becomes a hot spot for research in the anti-tumor field. Glutathione (GSH) is present in tumor cells at higher concentrations than in normal cells, and this special tumor cell reducing microenvironment has been widely used to achieve specific drug release at the tumor site. Research finds that disulfide bonds have reduction sensitivity and can be rapidly broken in the presence of GSH, so that the insertion of disulfide bonds into the design of a prodrug is expected to realize tumor-specific drug release.
Disclosure of Invention
Based on this, we design and synthesize a series of disulfide bond bridged docetaxel-fatty acid prodrugs by combining with prodrug strategies, connect docetaxel with multiple fatty acids through reduction-sensitive disulfide bonds respectively, can stably exist in normal physiological environment, and rapidly break in abnormal high-reduction environment in tumor cells, thereby realizing specific release of the parent drug in the tumor cells. The synthesized prodrug is further prepared into prodrug self-assembly nanoparticles by a one-step nano precipitation method. The prepared prodrug self-assembly nanoparticles have the advantages of high drug loading rate (> 50%), small and uniform particle size, good stability, capability of quickly releasing parent drugs in a reducing environment and the like, and have excellent clinical transformation prospect.
The invention aims to design and synthesize a series of disulfide bond bridged docetaxel-fatty acid prodrugs, and respectively prepare the prodrugs into prodrug self-assembled nanoparticles to investigate the influence of different fatty acid side chains on the properties of the prodrugs. Provides a new strategy for developing a tumor microenvironment stimulus response type drug delivery system, and meets the requirement of synergy and attenuation of docetaxel preparation in clinic.
The invention realizes the aim through the following technical scheme:
the invention provides a series of disulfide bond bridged docetaxel-fatty acid prodrugs, wherein the fatty acid is stearic acid, oleic acid, elaidic acid, linoleic acid, alpha-linolenic acid, docosahexaenoic acid, acetic acid, propionic acid, butyric acid, valeric acid, caproic acid, heptanoic acid, caprylic acid, pelargonic acid, capric acid, lauric acid, myristic acid, palmitic acid, pearlescent aliphatic acid, arachidic acid, behenic acid, (E) -11-eicosenoic acid, erucic acid or nervonic acid.
The series of disulfide-bridged docetaxel-fatty acid prodrugs of the present invention comprise: (a) disulfide bridged docetaxel-stearic acid prodrug (n ═ 17), (b) disulfide bridged docetaxel-oleic acid prodrug (n ═ 7), (c) disulfide bridged docetaxel-elaidic acid prodrug, (d) disulfide bridged docetaxel-linoleic acid prodrug, (e) disulfide bridged docetaxel- α -linolenic acid prodrug, (f) disulfide bridged docetaxel-docosahexaenoic acid prodrug, having the following structures:
Figure BDA0002845330290000021
the invention provides a method for synthesizing series disulfide bond bridged docetaxel-fatty acid small molecule prodrugs, which comprises the following steps:
fatty acid reacts with glycol under the catalysis of p-toluenesulfonic acid to obtain fatty acid-glycol ester, the fatty acid-glycol ester reacts with 2,2' -dithiodiacetic anhydride under the catalysis of HOBt, EDCI and DMAP to obtain an intermediate product, the intermediate product reacts with docetaxel under the catalysis of HOBt and EDCI to obtain the compound, and the compound is separated and purified, wherein the reaction process is carried out in the whole process of N2Under protection. Ethylene glycol may also be replaced with ethylene diamine.
The preparation method of the series of disulfide bond bridged docetaxel-fatty acid small molecule prodrug self-assembly nanoparticles provided by the invention comprises the following steps:
dissolving a certain amount of small molecule prodrug into a proper amount of ethanol solution, slowly dripping the ethanol solution into deionized water under the stirring of a magnetic stirrer, and spontaneously assembling the prodrug into nanoparticles with uniform particle size.
The small molecule prodrug nanoparticles can be non-PEG small molecule prodrug nanoparticles and PEG modified small molecule prodrug nanoparticles.
(1) The preparation method of the non-PEG small molecule prodrug self-assembly nanoparticle comprises the following steps: dissolving a certain amount of small molecule prodrug into a proper amount of ethanol solution, slowly dropwise adding the ethanol solution into deionized water under the stirring of a magnetic stirrer, spontaneously assembling the prodrug into nanoparticles with uniform particle size, and removing ethanol in the preparation by adopting a reduced-pressure rotary evaporation method to obtain ethanol-free nanoparticles.
(2) The preparation method of the PEG micromolecule prodrug self-assembly nanoparticle comprises the following steps: mixing a certain amount of small molecule prodrug and DSPE-PEG2000Dissolving the mixture into a proper amount of ethanol solution, slowly dropwise adding the ethanol solution into deionized water under the stirring of a magnetic stirrer, spontaneously assembling the prodrug into nanoparticles with uniform particle size, and removing ethanol in the preparation by adopting a reduced-pressure rotary evaporation method to obtain the PEG nanoparticles without ethanol.
The series disulfide bond bridged docetaxel-fatty acid prodrug self-assembly nano drug delivery system has the following advantages: (1) docetaxel is prepared into a prodrug connected by disulfide bonds, so that the systematic toxicity of docetaxel is reduced, the parent drug can be specifically released in a high-reduction environment in tumor cells, and the effects of synergy and attenuation are realized; (2) the drug loading is high, the use of a solubilizer with high toxicity is avoided, and the tolerance and compliance of patients are expected to be improved; (3) the preparation is prepared by a one-step nano precipitation method, the preparation process is simple, and the large-scale production of the preparation is easy; (4) the nanoparticles have small and uniform particle size and are easy to enrich in tumor parts through an EPR effect; (5) is easy to surface modify, and can be modified by PEG and the like to slow down the clearance of the reticuloendothelial system.
Drawings
FIG. 1 is a high resolution mass spectrum of the disulfide bridged docetaxel-stearic acid prodrug (DTX-S-S-SA) of example 1
FIG. 2 is a high resolution mass spectrum of disulfide bridged docetaxel-oleic acid prodrug (DTX-S-S-OA) of example 2
FIG. 3 is a high resolution mass spectrum of the disulfide bridged docetaxel-elaidic acid prodrug (DTX-S-S-EA) of example 3
FIG. 4 is a high resolution mass spectrum of the disulfide bridged docetaxel-linoleic acid prodrug (DTX-S-S-LA) of example 4
FIG. 5 is a high resolution mass spectrum of the disulfide bridged docetaxel-linolenic acid prodrug (DTX-S-S-LNA) of example 5
FIG. 6 is a high resolution mass spectrum of the disulfide-bridged docetaxel-docosahexaenoic acid prodrug (DTX-S-S-DHA) of example 6
FIG. 7 is a transmission electron microscope micrograph of PEG-modified small molecule prodrug self-assembled nanoparticles of example 8
FIG. 8 is a graph of particle size versus storage time for PEG-modified small molecule prodrug self-assembled nanoparticles of example 9
Fig. 9 is a graph of an in vitro release assay for PEG-modified small molecule prodrug self-assembled nanoparticles of example 10.
Fig. 10 is a cytotoxicity diagram of PEG-modified docetaxel prodrug self-assembly nanoparticles of example 11 of the present invention.
Fig. 11 is a blood concentration-time curve diagram of PEG-modified docetaxel prodrug self-assembled nanoparticles of example 12 of the present invention.
Fig. 12 is an in vivo anti-tumor experimental graph of PEG-modified docetaxel prodrug self-assembly nanoparticles of example 13 of the present invention
Detailed Description
The invention is further illustrated by the following examples, which are not intended to limit the invention thereto.
Example 1: synthesis of disulfide bridged docetaxel-stearic acid prodrugs (DTX-S-S-SA)
Excess ethylene glycol was measured and placed in a 50mL three-necked flaskIn a bottle, adding a proper amount of p-toluenesulfonic acid into a three-necked bottle, refluxing by a spherical condenser pipe, and performing reaction on N2And (4) protecting, heating to 110 ℃, slowly dripping a stearic acid solution dissolved in toluene into a reaction bottle through a constant pressure dropping funnel, and reacting for 2 hours. After the reaction is finished, standing for layering, extracting for three times by using methylbenzene, combining methylbenzene layers, and then using saturated NaHCO for3Washing the solution to be neutral, drying the solution by anhydrous sodium sulfate, filtering the solution, evaporating the solution to dryness, separating and purifying the solution to obtain the stearic acid-2-hydroxyethyl ester. Using CH for 2,2' -dithiodiacetic anhydride2Cl2Dissolving and transferring to 100mL eggplant-shaped bottle, adding HOBt, EDCI and 2-hydroxyethyl stearate, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolving DMAP, stirring for 30min, transferring to room temp, reacting for 12 hr, TLC monitoring reaction, evaporating, acidifying with diluted hydrochloric acid, and reacting with CH2Cl2Extracting until TLC of water layer has no product point, washing CH with saturated NaCl solution2Cl2The layers were dried over anhydrous sodium sulfate, filtered and evaporated to dryness to give intermediate (2-oxo-2- (2-stearoyloxy-ethoxy) ethanedithiol) acetic acid. Dissolving (2-oxo-2- (2-stearoyloxy) ethoxy) ethyldithio) acetic acid, EDCI, HOBt and docetaxel in CH2Cl2Then put into a 100mL eggplant-shaped bottle, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolved DMAP, stirred for 1 hour, then transferred to 25 ℃ for reaction for 48 hours, monitored by TLC for completion of the reaction, and washed with saturated NaCl solution2Cl2The layer was dried over anhydrous sodium sulfate, filtered, evaporated to dryness and purified to give a white solid.
The prodrug structure of example 1 was determined by high resolution mass spectrometry using acetonitrile as the solvent, the result is shown in FIG. 1, and the molecular formula is C67H95NO19S2,m/z:1281.6。
Example 2: synthesis of disulfide bridged docetaxel-oleic acid prodrugs (DTX-S-S-OA)
Measuring excessive glycol, placing in a 50mL three-necked flask, adding appropriate amount of p-toluenesulfonic acid into the three-necked flask, refluxing with a spherical condenser tube, and performing N2Protecting, heating to 110 deg.C, dissolving toluene-dissolved oleic acidThe solution was slowly dripped into the reaction flask through a constant pressure dropping funnel, reacted for 2h, and the completion of the reaction was monitored by TLC. After the reaction is finished, standing for layering, extracting with toluene until no product point exists in a TLC (thin layer chromatography) of an ethylene glycol layer, combining toluene layers, and then using saturated NaHCO for carrying out secondary reaction3Washing the solution to be neutral, drying the solution by anhydrous sodium sulfate, filtering the solution, evaporating the solution to dryness, separating and purifying the solution to obtain the oleic acid-2-hydroxyethyl ester. Using CH for 2,2' -dithiodiacetic anhydride2Cl2Dissolving and transferring to 100mL eggplant-shaped bottle, adding HOBt, EDCI and oleic acid-2-hydroxyethyl ester, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolving DMAP, stirring for 30min, transferring to room temp, reacting for 12 hr, TLC monitoring reaction, evaporating, acidifying with diluted hydrochloric acid, and reacting with CH2Cl2Extracting until TLC of water layer has no product point, washing CH with saturated NaCl solution2Cl2The layers were dried over anhydrous sodium sulfate, filtered and evaporated to dryness to give intermediate (2-oxo-2- (2-oleoyloxy-ethoxy) ethanedithio) acetic acid. Dissolving (2-oxo-2- (2-oleoyloxy) ethoxy) ethanedithiol) acetic acid, EDCI, HOBt and docetaxel in CH2Cl2Then put into a 100mL eggplant-shaped bottle, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolved DMAP, stirred for 1 hour, then transferred to 25 ℃ for reaction for 48 hours, monitored by TLC for completion of the reaction, and washed with saturated NaCl solution2Cl2The layer was dried over anhydrous sodium sulfate, filtered, evaporated to dryness and purified to give a white solid.
The prodrug structure of example 2 was determined by high resolution mass spectrometry using acetonitrile as the solvent, the result is shown in FIG. 2, and the molecular formula is C67H93NO19S2,m/z:1279.6。
Example 3: synthesis of disulfide bridged docetaxel-elaidic acid prodrugs (DTX-S-S-EA)
Measuring excessive glycol, placing in a 50mL three-necked flask, adding appropriate amount of p-toluenesulfonic acid into the three-necked flask, refluxing with a spherical condenser tube, and performing N2And (4) protecting, heating to 110 ℃, slowly dripping the toluene-dissolved elaidic acid solution into a reaction bottle through a constant-pressure dropping funnel, reacting for 2 hours, and monitoring the reaction completion by TLC. After the reaction is finishedStanding for layering, extracting with toluene until no product is produced in TLC of ethylene glycol layer, mixing toluene layers, and adding saturated NaHCO3Washing the solution to be neutral, drying the solution by anhydrous sodium sulfate, filtering the solution, evaporating the solution to dryness, separating and purifying the solution to obtain the elaidic acid-2-hydroxyethyl ester. Using CH for 2,2' -dithiodiacetic anhydride2Cl2Dissolving and transferring to 100mL eggplant-shaped bottle, adding HOBt, EDCI and trans-oleic acid-2-hydroxyethyl ester, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolving DMAP, stirring for 30min, transferring to room temp, reacting for 12 hr, TLC monitoring reaction, evaporating, acidifying with diluted hydrochloric acid, and reacting with CH2Cl2Extracting until TLC of water layer has no product point, washing CH with saturated NaCl solution2Cl2The layers were dried over anhydrous sodium sulfate, filtered and evaporated to dryness to give intermediate (2-oxo-2- (2-elaidoyloxy-ethoxy) ethanedithio) acetic acid. Dissolving (2-oxo-2- (2-elaidoyloxy) ethoxy) ethanedithiol) acetic acid, EDCI, HOBt and docetaxel in CH2Cl2Then put into a 100mL eggplant-shaped bottle, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolved DMAP, stirred for 1 hour, then transferred to 25 ℃ for reaction for 48 hours, monitored by TLC for completion of the reaction, and washed with saturated NaCl solution2Cl2The layer was dried over anhydrous sodium sulfate, filtered, evaporated to dryness and purified to give a white solid.
The prodrug structure of example 3 was determined by high resolution mass spectrometry using acetonitrile as the solvent, the result is shown in FIG. 3, and the molecular formula is C67H93NO19S2,m/z:1279.6。
Example 4: synthesis of disulfide bridged docetaxel-linoleic acid prodrugs (DTX-S-S-LA)
Measuring excessive glycol, placing in a 50mL three-necked flask, adding appropriate amount of p-toluenesulfonic acid into the three-necked flask, refluxing with a spherical condenser tube, and performing N2And (4) protecting, heating to 110 ℃, slowly dripping the linoleic acid solution dissolved in toluene into a reaction bottle through a constant pressure dropping funnel, reacting for 2 hours, and monitoring the reaction completion by TLC. After the reaction is finished, standing for layering, extracting with toluene until no product point exists in TLC of an ethylene glycol layer, combining toluene layers, and then saturatingAnd NaHCO3Washing the solution to be neutral, drying the solution by anhydrous sodium sulfate, filtering the solution, evaporating the solution to dryness, and separating and purifying the solution to obtain the linoleic acid-2-hydroxyethyl ester. Using CH for 2,2' -dithiodiacetic anhydride2Cl2Dissolving and transferring to 100mL eggplant-shaped bottle, adding HOBt, EDCI and linoleic acid-2-hydroxyethyl ester, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolving DMAP, stirring for 30min, transferring to room temp, reacting for 12 hr, TLC monitoring reaction, evaporating, acidifying with diluted hydrochloric acid, and reacting with CH2Cl2Extracting until TLC of water layer has no product point, washing CH with saturated NaCl solution2Cl2The layers were dried over anhydrous sodium sulfate, filtered and evaporated to dryness to give intermediate (2-oxo-2- (2-linoleoyloxy-ethoxy) ethanedithio) acetic acid. Dissolving (2-oxo-2- (2-linoleoyloxy) ethoxy) ethanedithiol) acetic acid, EDCI, HOBt and docetaxel in CH2Cl2Then put into a 100mL eggplant-shaped bottle, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolved DMAP, stirred for 1 hour, then transferred to 25 ℃ for reaction for 48 hours, monitored by TLC for completion of the reaction, and washed with saturated NaCl solution2Cl2The layer was dried over anhydrous sodium sulfate, filtered, evaporated to dryness and purified to give a white solid.
The prodrug structure of example 4 was determined by high resolution mass spectrometry using acetonitrile as the solvent, the result is shown in FIG. 4, and the molecular formula is C67H91NO19S2,m/z:1277.6。
Example 5: synthesis of disulfide bridged docetaxel-alpha-linolenic acid prodrugs (DTX-S-S-LNA)
Measuring excessive glycol, placing in a 50mL three-necked flask, adding appropriate amount of p-toluenesulfonic acid into the three-necked flask, refluxing with a spherical condenser tube, and performing N2And (3) protecting, heating to 110 ℃, slowly dripping the alpha-linolenic acid solution dissolved in the toluene into a reaction bottle through a constant pressure dropping funnel, reacting for 2 hours, and monitoring the reaction completion by TLC. After the reaction is finished, standing for layering, extracting with toluene until no product point exists in a TLC (thin layer chromatography) of an ethylene glycol layer, combining toluene layers, and then using saturated NaHCO for carrying out secondary reaction3Washing the solution to neutral, drying with anhydrous sodium sulfate, filtering, evaporating to dryness, and separatingPurifying to obtain the alpha-linolenic acid-2-hydroxyethyl ester. Using CH for 2,2' -dithiodiacetic anhydride2Cl2Dissolving and transferring to 100mL eggplant-shaped bottle, adding HOBt, EDCI and alpha-linolenic acid-2-hydroxyethyl ester, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolving DMAP, stirring for 30min, transferring to room temp, reacting for 12 hr, TLC monitoring reaction, evaporating, acidifying with diluted hydrochloric acid, and reacting with CH2Cl2Extracting until TLC of water layer has no product point, washing CH with saturated NaCl solution2Cl2The layers were dried over anhydrous sodium sulfate, filtered and evaporated to dryness to give intermediate (2-oxo-2- (2- α -linolenoyloxy-ethoxy) ethanedithio) acetic acid. Dissolving (2-oxo-2- (2-alpha-linolenoyloxy) ethoxy) ethanedithiol) acetic acid, EDCI, HOBt and docetaxel in CH2Cl2Then put into a 100mL eggplant-shaped bottle, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolved DMAP, stirred for 1 hour, then transferred to 25 ℃ for reaction for 48 hours, monitored by TLC for completion of the reaction, and washed with saturated NaCl solution2Cl2The layer was dried over anhydrous sodium sulfate, filtered, evaporated to dryness and purified to give a white solid.
The prodrug structure of example 5 was determined by high resolution mass spectrometry using acetonitrile as the solvent, the result is shown in FIG. 5, and the molecular formula is C67H89NO19S2,m/z:1277.6。
Example 6: synthesis of disulfide-bridged docetaxel-docosahexaenoic acid prodrugs (DTX-S-S-DHA)
Measuring excessive glycol, placing in a 50mL three-necked flask, adding appropriate amount of p-toluenesulfonic acid into the three-necked flask, refluxing with a spherical condenser tube, and performing N2And (3) protecting, heating to 110 ℃, slowly dripping the toluene-dissolved docosahexaenoic acid solution into a reaction bottle through a constant pressure dropping funnel, reacting for 2 hours, and monitoring the reaction completion by TLC. After the reaction is finished, standing for layering, extracting with toluene until no product point exists in a TLC (thin layer chromatography) of an ethylene glycol layer, combining toluene layers, and then using saturated NaHCO for carrying out secondary reaction3Washing the solution to neutrality, drying with anhydrous sodium sulfate, filtering, evaporating to dryness, separating and purifying to obtain docosahexaenoic acid-2-hydroxyethyl ester. 2,2CH for' -dithiodiacetic anhydride2Cl2Dissolving and transferring into 100mL eggplant-shaped bottle, adding HOBt, EDCI and docosahexaenoic acid-2-hydroxyethyl ester, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolving DMAP, stirring for 30min, transferring to room temp, reacting for 12 hr, TLC monitoring reaction, evaporating, acidifying with diluted hydrochloric acid, and reacting with CH2Cl2Extracting until TLC of water layer has no product point, washing CH with saturated NaCl solution2Cl2The layers were dried over anhydrous sodium sulfate, filtered and evaporated to dryness to give intermediate (2-oxo-2- (2-docosahexenoyloxy-ethoxy) ethanedidisulfide) acetic acid. Dissolving (2-oxo-2- (2-docosahexenoyloxy) ethoxy) ethanedithiol) acetic acid, EDCI, HOBt and docetaxel in CH2Cl2Then put into a 100mL eggplant-shaped bottle, N2Cooling to 0 ℃ under the protection condition, and slowly dripping CH2Cl2Dissolved DMAP, stirred for 1 hour, then transferred to 25 ℃ for reaction for 48 hours, monitored by TLC for completion of the reaction, and washed with saturated NaCl solution2Cl2The layer was dried over anhydrous sodium sulfate, filtered, evaporated to dryness and purified to give a white solid.
The prodrug structure of example 6 was determined by high resolution mass spectrometry using acetonitrile as the solvent, the result is shown in FIG. 6, and the molecular formula is C71H91NO19S2,m/z:1325.6。
Example 7: preparation of non-PEG small molecule prodrug self-assembly nanoparticles
Precisely weighing 8mg of prodrug, dissolving the prodrug in 1mL of ethanol, slowly dripping the ethanol solution into 4mL of deionized water under stirring to spontaneously form uniform nanoparticles (DTX-S-S-SA nanoparticles, DTX-S-S-OA nanoparticles, DTX-S-S-EA nanoparticles, DTX-S-S-LA nanoparticles, DTX-S-S-LNA nanoparticles, DTX-S-S-DHA nanoparticles)
Example 8: preparation of PEG small molecule prodrug self-assembly nanoparticles
Precisely weighing prodrug 8mg and DSPE-PEG20001.6mg of the compound, dissolved in 1mL of ethanol, and added slowly dropwise to 4mL of deionized water with stirring to spontaneously form uniform nanoparticles (DTX-S-S-SA)/DSPE-PEG2000Nanoparticles, DTX-S-S-OA/DSPE-PEG2000Nanoparticles, DTX-S-S-EA/DSPE-PEG2000Nanoparticles, DTX-S-S-LA/DSPE-PEG2000Nanoparticles, DTX-S-S-LNA/DSPE-PEG2000Nanoparticles, DTX-S-S-DHA/DSPE-PEG2000Nano granule)
As shown in table 1, the prodrug nanoparticles have a small particle size, a particle size distribution of less than 0.2, and a high drug loading (> 50%). The particle size and morphology of the small molecule prodrug self-assembled nanoparticles prepared in example 8 were determined by transmission electron microscopy, and the results are shown in fig. 7, where the transmission electron microscopy shows that the drug-loaded nanoparticles are uniform spheres with a particle size of about 100 nm.
TABLE 1 particle size, particle size distribution and drug loading of PEG-modified prodrug self-assembled nanoparticles
Figure BDA0002845330290000091
Example 9: storage stability test of PEG modified small molecule prodrug self-assembly nanoparticles
The PEG modified small molecule prodrug prepared in the example 8 is self-assembled nanoparticle (DTX-S-S-SA/DSPE-PEG)2000Nanoparticles, DTX-S-S-OA/DSPE-PEG2000Nanoparticles, DTX-S-S-EA/DSPE-PEG2000Nanoparticles, DTX-S-S-LA/DSPE-PEG2000Nanoparticles, DTX-S-S-LNA/DSPE-PEG2000Nanoparticles, DTX-S-S-DHA/DSPE-PEG2000Nanoparticles) were stored at 4 ℃ for 2 months. During this time, the particle size change was measured by dynamic light scattering. The results are shown in FIG. 8, where only DTX-S-S-SA/DSPE-PEG2000Nanoparticles and DTX-S-S-OA/DSPE-PEG2000The particle size of the nanoparticles showed no significant change, and the other 4 nanoparticles showed an increase in particle size. The above results show that DTX-S-S-SA/DSPE-PEG2000Nanoparticles and DTX-S-S-OA/DSPE-PEG2000The nanoparticles have good storage stability, and the prepared DTX-S-S-EA/DSPE-PEG2000Nanoparticles, DTX-S-S-LA/DSPE-PEG2000Nanoparticles, DTX-S-S-LNA/DSPE-PEG2000Nanoparticles, DTX-S-S-DHA/DSPE-PEG2000Storage of nanoparticlesThe stability is poor.
Example 10: in vitro release test of PEG modified small molecule prodrug self-assembled nanoparticles
The in vitro release condition of the PEG modified small molecule prodrug self-assembly nanoparticles is examined by taking pH 7.4 phosphate buffer solution containing ethanol as a release medium. The PEG-modified small molecule prodrug self-assembled nanoparticle prepared in example 8 was added to 30mL of release medium and Dithiothreitol (DTT) at a certain concentration was added to the release medium, sampling was performed at 37 ℃ at a set time point, and the concentration of released docetaxel was determined by high performance liquid chromatography to examine the release of nanoparticles under reducing conditions.
The results are shown in fig. 9, and experimental results show that the series of docetaxel-fatty acid small molecule prodrugs designed by the invention can quickly release the parent drug in a reducing environment, have the characteristic of reduction-sensitive drug release, can respond to the oxidation-reduction environment specific to a tumor part, realize the drug release specific to the tumor part, are expected to improve the anti-tumor effect of docetaxel and reduce the toxic and side effects on an organism, and the release speeds of different fatty acid coupled docetaxel prodrugs are not obviously different, which suggests that different fatty acid side chains have no obvious influence on the release of the parent drug.
Example 11: cytotoxicity experiment of PEG (polyethylene glycol) -modified docetaxel prodrug self-assembled nanoparticles
The toxicity of the PEG-modified docetaxel-fatty acid prodrug self-assembly nanoparticles on tumor cells (4T1) is examined by adopting an MTT method. Firstly, digesting cells with good state, diluting the cells with a culture solution to 1 ten thousand cells/mL, uniformly blowing the cells, adding 100 mu L of cell suspension into each hole of a 96-hole plate, and placing the cells in an incubator for incubation for 24 hours to adhere to the walls. Docetaxel solution diluted by culture solution or the pegylated docetaxel prodrug nanoparticle prepared in example 8 is added after the cells adhere to the wall. 200 μ L of medium containing different concentrations of drug was added to each well, 3 parallel wells per concentration. In the control group, 200 mul of culture solution is singly supplemented without adding the liquid medicine to be detected, and the control group is placed in an incubator to be incubated with cells together. And (3) taking out the 96-well plate 48h and 72h after adding the drug, adding 25 mu L of 5mg/mL MTT solution into each well, putting the plate in an incubator for incubation for 4h, throwing the plate, turning the 96-well plate over on filter paper, fully sucking the residual liquid, adding 200 mu L of DMSO into each well, and shaking the plate on a shaker for 10min to dissolve the bluish purple crystals. The A1 well (containing only 200. mu.L DMSO) was set as the zeroing well. The absorbance value after zeroing of each well was measured at 490nm using a microplate reader.
The cytotoxicity results are shown in fig. 10. Compared with the docetaxel solution group, the docetaxel prodrug nanoparticles have slightly weakened cytotoxicity. However, due to the special high-reduction environment of tumor cells, docetaxel can be rapidly released from the parent drug, and docetaxel prodrug nanoparticles still have strong cytotoxicity.
Example 12: pharmacokinetic study of PEGylated docetaxel-fatty acid prodrug self-assembled nanoparticles
SD rats with the body weight between 180-220g are taken, 5 rats in each group are randomly grouped, and fasting is performed for 12h before administration, and water is freely drunk. Docetaxel solution and pegylated docetaxel-fatty acid prodrug self-assembled nanoparticles prepared in example 8 were injected intravenously, respectively. The equivalent dose was 5 mg/kg. Blood was collected from the orbit at a predetermined time point and separated to obtain plasma. The concentrations of the prodrug and the released docetaxel in the plasma were measured by ultra performance liquid chromatography-mass spectrometry (UPLC-MS).
The results are shown in figure 11, the docetaxel solution was rapidly cleared by blood. In contrast, the circulation time of the pegylated docetaxel-fatty acid prodrug nanoparticles was significantly prolonged. Meanwhile, different fatty acid side chains also have obvious influence on the circulation time of the docetaxel prodrug. DTX-S-S-SA/DSPE-PEG2000Nanoparticles and DTX-S-S-OA/DSPE-PEG2000The nanoparticles showed higher AUC. While DTX-S-S-EA/DSPE-PEG2000Nanoparticles, DTX-S-S-LA/DSPE-PEG2000Nanoparticles, DTX-S-S-LNA/DSPE-PEG2000Nanoparticles, DTX-S-S-DHA/DSPE-PEG2000The AUC of nanoparticles is relatively small due to their poor colloidal stability.
Example 13: in-vivo anti-tumor experiment of PEG (polyethylene glycol) -modified docetaxel prodrug self-assembled nanoparticles
4T1 cell suspension (5 x 10)6cells/100. mu.L) was inoculated inFemale BALB/c mice were ventral subcutaneous. When the tumor volume grows to 150mm3In the meantime, tumor-bearing mice were randomly grouped into five groups, and physiological saline, a docetaxel solution and the PEG-modified docetaxel prodrug self-assembly nanoparticles prepared in example 8 were administered to each group. The administration was performed 1 time every 1 day and 5 times continuously, and the survival state of the mice was observed every day, and the tumor volume was measured by weighing.
The in vivo antitumor effect is shown in fig. 12A-B, and the docetaxel solution has a significant antitumor effect compared with the normal saline group. However, the docetaxel solution had severe side effects and the weight loss of mice was significant after administration (see fig. 12C). Compared with a docetaxel solution, the PEG docetaxel prodrug self-assembly nanoparticle has good safety, and the body weight of a mouse is not remarkably reduced. In addition, different antitumor effects are shown among different prodrug nanoparticles, SA-S-S-DTX nanoparticles and OA-S-S-DTX nanoparticles show better antitumor effects due to stronger colloidal stability and higher AUC, and other nanoparticles show poorer in vivo antitumor activity due to shorter circulation time.
In addition, a high-dose group is designed to investigate the safety of the docetaxel prodrug nanoparticles, and the results are shown in fig. 12D, so that even if the administration dose reaches 20mg/kg, mice in the prodrug nanoparticle group hardly die, and even after a docetaxel solution of 10mg/kg is administered, the mice die completely in 20 days.

Claims (9)

1. Disulfide bridged docetaxel-fatty acid prodrugs comprising (a) a disulfide bridged docetaxel-stearic acid prodrug (n ═ 17), (b) a disulfide bridged docetaxel-oleic acid prodrug (n ═ 7), (c) a disulfide bridged docetaxel-elaidic acid prodrug, (d) a disulfide bridged docetaxel-linoleic acid prodrug, (e) a disulfide bridged docetaxel- α -linolenic acid prodrug, (f) a disulfide bridged docetaxel-docosahexaenoic acid prodrug having the structure:
Figure FDA0002845330280000011
2. the disulfide bridged docetaxel-fatty acid prodrug of claim 1, wherein the docetaxel is a taxane compound.
3. The disulfide bridged docetaxel-fatty acid prodrug in accordance with claim 1, wherein the fatty acid is one or more of stearic acid, oleic acid, elaidic acid, linoleic acid, α -linolenic acid, docosahexaenoic acid, acetic acid, propionic acid, butyric acid, valeric acid, caproic acid, enanthic acid, caprylic acid, pelargonic acid, capric acid, lauric acid, myristic acid, palmitic acid, perlitic acid, arachidic acid, behenic acid, (E) -11-eicosenoic acid, erucic acid, nervonic acid.
4. The disulfide bridged docetaxel-fatty acid prodrug of claim 1, wherein the disulfide bond is further one of a hydrazone bond, a carbonate bond, a monothioether bond, a spacer disulfide bond or a diselenide bond.
5. The disulfide bridged docetaxel-fatty acid prodrug of claim 1, synthesized by: reacting fatty acid with ethylene glycol under the catalysis of p-toluenesulfonic acid to obtain fatty acid-ethylene glycol ester, carrying out ring-opening esterification reaction on the fatty acid-ethylene glycol ester and 2,2' -dithiodiacetic anhydride under the catalysis of HOBt, EDCI and DMAP to obtain an intermediate product, carrying out esterification reaction on the intermediate product and docetaxel under the catalysis of HOBt and EDCI, and separating and purifying to obtain the product, wherein the reaction is carried out in the whole process of N2Under protection.
6. Disulfide-bridged docetaxel-fatty acid-series prodrug self-assembled nanoparticles comprising the disulfide-bridged docetaxel-fatty acid prodrug of any one of claims 1 to 5, prepared by the following process:
dissolving a certain amount of disulfide-bond bridged docetaxel-fatty acid prodrug and a PEG stabilizer into a proper amount of ethanol, slowly dropwise adding the ethanol solution into deionized water under stirring, and spontaneously assembling the disulfide-bond bridged docetaxel-fatty acid prodrug into nanoparticles with uniform particle size.
7. The disulfide-bridged docetaxel-fatty acid prodrug self-assembled nanoparticle of claim 6, wherein the PEG is one of TPGS, DSPE-PEG, PLGA-PEG, PE-PEG and DSPE-PEG-AA.
8. Use of the disulfide-bridged docetaxel-fatty acid prodrug of any one of claims 1 to 5 or the disulfide-bridged docetaxel-fatty acid prodrug self-assembled nanoparticle of claims 6 to 7 for the preparation of an antitumor drug.
9. Use of the disulfide bridged docetaxel-fatty acid prodrug of any one of claims 1 to 5 or the disulfide bridged docetaxel-fatty acid prodrug self-assembled nanoparticle of claims 6 to 7 in a medicament for oral, injectable or topical administration.
CN202011492954.0A 2020-07-12 2020-12-18 Disulfide-bond bridged docetaxel-fatty acid prodrug and self-assembled nanoparticles thereof Pending CN112604002A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2020106659037 2020-07-12
CN202010665903 2020-07-12

Publications (1)

Publication Number Publication Date
CN112604002A true CN112604002A (en) 2021-04-06

Family

ID=75240848

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011492954.0A Pending CN112604002A (en) 2020-07-12 2020-12-18 Disulfide-bond bridged docetaxel-fatty acid prodrug and self-assembled nanoparticles thereof

Country Status (1)

Country Link
CN (1) CN112604002A (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115089720A (en) * 2022-06-20 2022-09-23 沈阳药科大学 Tumor treatment medicine composition
CN115252804A (en) * 2021-04-30 2022-11-01 苏州裕泰医药科技有限公司 Lalotataxel-fatty alcohol small molecule prodrug and construction of self-assembled nanoparticles thereof
WO2022227556A1 (en) * 2021-04-30 2022-11-03 苏州裕泰医药科技有限公司 Docetaxel-fatty alcohol small molecule prodrug and construction of self-assembled nanoparticles thereof
WO2022227555A1 (en) * 2021-04-30 2022-11-03 苏州裕泰医药科技有限公司 Cabazitaxel-fatty alcohol small molecule prodrug and construction of self-assembled nanoparticle thereof
CN115624631A (en) * 2021-12-14 2023-01-20 盐城工学院 Bufalin prodrug self-assembly nanoparticles and preparation method and application thereof
WO2023071900A1 (en) * 2021-10-29 2023-05-04 南京科润工业介质股份有限公司 Quenching oil membrane breaking additive and quenching oil containing membrane breaking additive
WO2023109562A1 (en) * 2021-12-15 2023-06-22 沈阳药科大学 Docetaxel prodrug anti-tumor preparation

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003063960A (en) * 1996-03-12 2003-03-05 Pg-Txl Co Lp Water-soluble paclitaxel prodrug
CN105833284A (en) * 2016-03-31 2016-08-10 沈阳药科大学 Construction of paclitaxel-oleic acid small-molecular prodrug self-assembled nanoparticles
CN110101872A (en) * 2019-06-13 2019-08-09 成都大学 A kind of reduction-sensitive nano-micelle and preparation method and application

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003063960A (en) * 1996-03-12 2003-03-05 Pg-Txl Co Lp Water-soluble paclitaxel prodrug
CN105833284A (en) * 2016-03-31 2016-08-10 沈阳药科大学 Construction of paclitaxel-oleic acid small-molecular prodrug self-assembled nanoparticles
CN110101872A (en) * 2019-06-13 2019-08-09 成都大学 A kind of reduction-sensitive nano-micelle and preparation method and application

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115252804A (en) * 2021-04-30 2022-11-01 苏州裕泰医药科技有限公司 Lalotataxel-fatty alcohol small molecule prodrug and construction of self-assembled nanoparticles thereof
WO2022227556A1 (en) * 2021-04-30 2022-11-03 苏州裕泰医药科技有限公司 Docetaxel-fatty alcohol small molecule prodrug and construction of self-assembled nanoparticles thereof
WO2022227554A1 (en) * 2021-04-30 2022-11-03 苏州裕泰医药科技有限公司 Larotaxel-fatty alcohol small molecule prodrug and construction of self-assembling nanoparticle thereof
WO2022227555A1 (en) * 2021-04-30 2022-11-03 苏州裕泰医药科技有限公司 Cabazitaxel-fatty alcohol small molecule prodrug and construction of self-assembled nanoparticle thereof
WO2023071900A1 (en) * 2021-10-29 2023-05-04 南京科润工业介质股份有限公司 Quenching oil membrane breaking additive and quenching oil containing membrane breaking additive
CN115624631A (en) * 2021-12-14 2023-01-20 盐城工学院 Bufalin prodrug self-assembly nanoparticles and preparation method and application thereof
WO2023109562A1 (en) * 2021-12-15 2023-06-22 沈阳药科大学 Docetaxel prodrug anti-tumor preparation
CN115089720A (en) * 2022-06-20 2022-09-23 沈阳药科大学 Tumor treatment medicine composition
CN115089720B (en) * 2022-06-20 2023-12-26 沈阳药科大学 Tumor treatment medicine composition

Similar Documents

Publication Publication Date Title
CN112604002A (en) Disulfide-bond bridged docetaxel-fatty acid prodrug and self-assembled nanoparticles thereof
CN109350748B (en) Redox double-sensitive bond bridged micromolecule prodrug and self-assembled nanoparticle thereof
CN113398277B (en) Fatty acid/fatty alcohol-antitumor substance prodrug and preparation method of self-assembled nanoparticles thereof
CN113264906B (en) Docetaxel dimer micromolecular prodrug and construction of self-assembled nanoparticle thereof
JP2017114913A (en) Nanoparticle formulations and uses thereof
CN111494640B (en) Redox double-sensitive trithio bond bridged dimer prodrug and self-assembled nanoparticles thereof
CN105833284A (en) Construction of paclitaxel-oleic acid small-molecular prodrug self-assembled nanoparticles
JP2011517683A (en) Composition of hydrophobic taxane derivative and use thereof
CN112494458B (en) Construction of triglyceride-like prodrug intravenous injection self-assembly nanoparticles
CN111116521B (en) Solanesol modified taxol prodrug and preparation method and application thereof
CN106946975B (en) Triptolide derivative and preparation method and preparation thereof
CN105997943B (en) A kind of nano particle and its preparation method and application of human serum albumins load camptothecine
CN112089845B (en) Taxane drug-adriamycin prodrug self-assembly nanoparticles and application thereof
CN110251685B (en) Synthesis method and application of taxol-berberine nano-drug
CN109846857B (en) Preparation method and application of active natural supramolecular photosensitizer
CN114796513B (en) Diselenide bridge Lian Duo cetime dimer prodrug and self-assembled nanoparticles thereof
CN1927203A (en) Nano micelle preparation of Catharanthus roseus alkaloids antineoplastic drugs with coating of phospholipid derived from polyethylene glycol
CN108836937B (en) Cisplatin nano-medicine preparation, preparation method and application
CN115300637B (en) Chalcogen hybrid bond bridged dimer prodrug, self-assembled nanoparticle thereof, preparation method and application
WO2022227555A1 (en) Cabazitaxel-fatty alcohol small molecule prodrug and construction of self-assembled nanoparticle thereof
CN113278092B (en) Polymer carrier material, preparation and application thereof
CN113135875B (en) Photosensitizer-driven dimer prodrug co-assembled nanoparticles and preparation method and application thereof
CN116120333B (en) Podophyllotoxin nano prodrug and preparation method and application thereof
WO2022227554A1 (en) Larotaxel-fatty alcohol small molecule prodrug and construction of self-assembling nanoparticle thereof
CN111643679B (en) Preparation method and application of chitosan oligosaccharide modified betulinic acid drug delivery system

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