CN113694027B - Polymer vesicle and preparation method thereof, and drug-loaded vesicle - Google Patents

Polymer vesicle and preparation method thereof, and drug-loaded vesicle Download PDF

Info

Publication number
CN113694027B
CN113694027B CN202111000584.9A CN202111000584A CN113694027B CN 113694027 B CN113694027 B CN 113694027B CN 202111000584 A CN202111000584 A CN 202111000584A CN 113694027 B CN113694027 B CN 113694027B
Authority
CN
China
Prior art keywords
drug
vesicle
polymer
polymersome
loaded
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
CN202111000584.9A
Other languages
Chinese (zh)
Other versions
CN113694027A (en
Inventor
张国庆
胡衎
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.)
University of Science and Technology of China USTC
Original Assignee
University of Science and Technology of China USTC
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 University of Science and Technology of China USTC filed Critical University of Science and Technology of China USTC
Priority to CN202111000584.9A priority Critical patent/CN113694027B/en
Publication of CN113694027A publication Critical patent/CN113694027A/en
Application granted granted Critical
Publication of CN113694027B publication Critical patent/CN113694027B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1273Polymersomes; Liposomes with polymerisable or polymerised bilayer-forming substances
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Dispersion Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Inorganic Chemistry (AREA)
  • Medicinal Preparation (AREA)

Abstract

The disclosure provides a polymer vesicle and a preparation method thereof, and a drug-loaded vesicle, wherein,the polymersome includes: a self-assembly of a polylactic acid zwitterionic polymer, the polylactic acid zwitterionic polymer comprising at least one of: formula (I):
Figure DDA0003233758190000011
formula (II):
Figure DDA0003233758190000012
wherein R is selected from any one of the following groups: hydroxyl-modified C10-C20 amino acid group, substituted or unsubstituted glycerophosphorylcholine; r' is selected from any one of the following groups: hydroxyl modified C5-C10 amino acid group, substituted or unsubstituted glycerophosphorylcholine and hydroxyl modified phosphorylcholine; x includes an integer greater than or equal to 1, and y and z each include an integer greater than or equal to 0.

Description

Polymer vesicle and preparation method thereof, and drug-loaded vesicle
Technical Field
The disclosure relates to the field of nano materials, in particular to a polymer vesicle and a preparation method thereof, and a drug-loaded vesicle.
Background
Vesicles are amphiphilic molecules, such as many natural synthetic surfactants and phospholipids that cannot simply associate into micelles, that spontaneously form ordered assemblies of molecules with a closed bilayer structure when dispersed in water, also known as liposomes. Are commonly used to store, transport and digest cellular products and waste products in some cells.
The vesicle has a bilayer membrane structure to make it become a drug carrier in vivo, which can carry hydrophilic drugs such as amino acids, polypeptides, proteins and nucleic acid vaccines, encapsulate the drugs in the water phase in the vesicle, and carry oil-soluble drugs such as natural anticancer drugs such as paclitaxel and superparamagnetic iron oxide nanoparticles, and solubilize the drugs in the hydrophobic region in the bilayer. The vesicle is used as a drug carrier, has the characteristics of drug transportation, drug distribution change in vivo, drug degradation inactivation prevention, drug action time prolonging, toxic and side effects reduction and the like, and can enhance the long circulation and targeting of the vesicle in blood after surface modification.
However, the synthesis process of the polymersome in the related art is complicated, resulting in high cost and difficulty in mass production. Meanwhile, the polymersome has potential toxicity and non-degradability, so that the application of the polymersome in clinic is limited.
Disclosure of Invention
In view of the above, the present disclosure provides a polymersome, a preparation method thereof, and a drug-loaded vesicle, so as to at least partially solve one of the above technical problems.
Embodiments of the present disclosure provide a polymersome, comprising: a self-assembly of a polylactic acid zwitterionic polymer, said polylactic acid zwitterionic polymer comprising at least one of:
formula (I):
Figure BDA0003233758170000021
formula (II):
Figure BDA0003233758170000022
wherein the content of the first and second substances,
r is selected from any one of the following groups: hydroxyl-modified C10-C20 amino acid group, substituted or unsubstituted glycerophosphorylcholine;
r' is selected from any one of the following groups: hydroxyl modified C5-C10 amino acid group, substituted or unsubstituted glycerophosphorylcholine and hydroxyl modified phosphorylcholine;
x includes an integer greater than or equal to 1, and y and z each include an integer greater than or equal to 0.
According to an embodiment of the present disclosure, the hydroxyl-modified C10 to C20 amino acid group includes a molecular structure represented by formula (III):
Figure BDA0003233758170000023
wherein R is 1 、R 2 Any one selected from the following groups: substituted or unsubstituted C2 to C7 hydroxyl.
According to an embodiment of the present disclosure, the hydroxyl-modified C5 to C10 amino acid group includes a molecular structure represented by formula (IV):
Figure BDA0003233758170000024
wherein the content of the first and second substances,
R 3 any one selected from the following groups: hydrogen, alkyl;
R 4 any one selected from the following groups: substituted or unsubstituted C1 to C7 hydroxyl groups.
Further embodiments of the present disclosure provide a method of preparing the polymersome described above, comprising: dissolving the polylactic acid zwitterionic polymer in an organic solvent to obtain an organic phase; and adding the organic phase into the water phase under continuous stirring, and stirring until the organic solvent is volatilized to obtain the polymer vesicle.
According to an embodiment of the present disclosure, the organic solvent includes at least one of: acetone, tetrahydrofuran, N-dimethylformamide; the aqueous phase comprises at least one of: water, phosphate buffer.
According to an embodiment of the present disclosure, the volume ratio of the organic phase to the aqueous phase includes 1:5-1.
Still other embodiments of the present disclosure provide a drug-loaded vesicle comprising the polymersome described above and a drug encapsulated in the polymersome described above.
According to an embodiment of the present disclosure, the above-mentioned medicament includes at least one of: hydrophobic drugs, hydrophilic drugs.
According to the embodiment of the disclosure, the mass ratio of the polymersome to the drug comprises 8:1-10.
According to an embodiment of the present disclosure, the particle size of the polymersome includes 100 to 300nm.
By the embodiment of the disclosure, the polylactic acid zwitterionic polymer forming the polymersome is derived from amino acid containing hydroxyl, glycerophosphorylcholine and other biosafety small molecules. The polylactic acid zwitterionic polymer has degradability, and degradation products of lactic acid, glycerophosphorylcholine, amino acid and the like are all naturally-occurring small molecules in organisms, so that the polymer vesicle is degradable and high in safety.
Drawings
Fig. 1 schematically shows an electron microscope image of polymersomes prepared in example 1;
fig. 2 schematically shows an electron microscope image of the polymersome prepared in example 2;
fig. 3 schematically shows an electron microscope picture of the polymersome prepared in example 3;
fig. 4 schematically shows an electron microscope image of the polymersome prepared in example 4;
fig. 5 schematically shows an electron microscope picture of polymersomes prepared in example 5;
fig. 6 schematically shows an electron microscope image of polymersomes prepared in example 6;
fig. 7 schematically shows the particle size distribution of the polymersome prepared in examples 1 to 6;
FIG. 8 schematically shows the change in the average particle diameter of polymersome prepared in examples 1 to 6 with time;
fig. 9 schematically shows the results of cytotoxicity tests of the polymersomes prepared in examples 1 to 6;
fig. 10 schematically shows in vitro drug release profiles of the drug-loaded vesicles of examples 7-10;
fig. 11 schematically shows the in vitro drug release profile of the drug-loaded vesicles of examples 11-14.
Detailed Description
For the purpose of promoting a better understanding of the objects, aspects and advantages of the present disclosure, reference is made to the following detailed description taken in conjunction with the accompanying drawings.
The commonly used polymersome materials are amphiphilic block copolymers, including three types of diblock, triblock and graft, as shown in table 1:
table 1 common block copolymers for the preparation of polymersomes
Figure BDA0003233758170000041
The vesicle is easy to be recognized and phagocytized by the immune system when circulating in the body and is quickly eliminated. Therefore, some hydrophilic polymers such as polyethylene glycol, poloxamer, polysorbate and polysaccharides such as dextran can be used in designing the block copolymer to change the surface properties of the nanoparticles. These polymers provide a dynamic hydrophilic and neutral chain cloud on the surface of vesicles to repel plasma protein adhesion. Polyethylene glycol-containing copolymers are currently the most common surface modification method to increase surface hydrophilicity and improve circulation half-life by reducing the interaction of blood proteins and MPS cells, but it has been reported that the rapid clearance of pegylated nanoparticles from blood after repeated injections is still a serious problem, easily causing cumulative toxicity.
In view of the above, the disclosed embodiments provide a polymersome, including: a self-assembly of a polylactic acid zwitterionic polymer, the polylactic acid zwitterionic polymer comprising at least one of:
formula (I):
Figure BDA0003233758170000051
formula (II):
Figure BDA0003233758170000052
wherein R is selected from any one of the following groups: hydroxyl-modified C10-C20 amino acid group, substituted or unsubstituted glycerophosphorylcholine; r' is selected from any one of the following groups: hydroxyl modified C5-C10 amino acid group, substituted or unsubstituted glycerophosphorylcholine and hydroxyl modified phosphorylcholine; x includes an integer greater than or equal to 1, and y and z each include an integer greater than or equal to 0.
According to embodiments of the present disclosure, polylactic acid zwitterionic polymers include, but are not limited to, compounds represented by formulas (one) - (five):
Figure BDA0003233758170000053
Figure BDA0003233758170000061
wherein x comprises an integer greater than or equal to 1, and y and z each comprise an integer greater than or equal to 0.
By the embodiment of the disclosure, the polylactic acid zwitterionic polymer forming the polymersome is derived from biosafety small molecules such as amino acid containing hydroxyl, glycerophosphorylcholine and the like. The polylactic acid zwitterionic polymer has degradability, and degradation products of lactic acid, glycerophosphorylcholine, amino acid and the like are all naturally-occurring small molecules in organisms, so that the polymer vesicle is degradable and has high safety.
According to an embodiment of the present disclosure, a hydroxyl-modified C10-C20 amino acid group includes a molecular structure represented by formula (III):
formula (III):
Figure BDA0003233758170000062
wherein R is 1 、R 2 Any one selected from the following groups: substituted or unsubstituted C2 to C7 hydroxyl.
According to embodiments of the present disclosure, hydroxy-modified C10-C20 amino acid groups include, but are not limited to, compounds represented by formulas (six) to (eleven):
Figure BDA0003233758170000071
according to the embodiment of the disclosure, hydroxyl groups which are not dehydrogenated are used as active sites to be connected to form a polymer, and the polymer can be hydrolyzed to generate amino acid and lactic acid, so that the polymer vesicle has degradability, can be degraded in a living body to form water-soluble small molecules such as amino acid and lactic acid, and is originally existed in the living body, and therefore, the toxicity of the polymer vesicle is low.
According to an embodiment of the present disclosure, a hydroxyl-modified C5-C10 amino acid group includes a molecular structure represented by formula (IV):
formula (IV)
Figure BDA0003233758170000081
Wherein the content of the first and second substances,
R 3 any one selected from the following groups: hydrogen, alkyl;
R 4 any one selected from the following groups: substituted or unsubstituted C1 to C7 hydroxyl groups.
According to embodiments of the present disclosure, a hydroxy-modified C5-C10 amino acid group includes, but is not limited to, compounds represented by the formulas (twelve) to (fourteen):
Figure BDA0003233758170000082
according to the embodiment disclosed by the invention, hydroxyl groups losing hydrogen are taken as active sites to be connected to form a zwitterionic polymer, and the characteristics of the zwitterionic polymer are utilized, so that the vesicle can be helped to avoid protein adhesion, and the in vivo circulation stability is improved.
Further embodiments of the present disclosure provide a method of preparing the polymersome described above, comprising:
dissolving polylactic acid amphoteric ion polymer in an organic solvent to obtain an organic phase; and adding the organic phase into the water phase under continuous stirring, and stirring until the organic solvent is volatilized to obtain the polymer vesicle.
According to the embodiment of the disclosure, a polylactic acid zwitterionic polymer is dissolved in an organic solvent to obtain an organic phase, wherein the concentration range of the polymer zwitterionic polymer comprises 0.5-50 mg/mL. For example: may be 0.5mg/mL, 10mg/mL, 30mg/mL, 50mg/mL, and the like.
According to the embodiment of the disclosure, the solvent volatilization method is adopted to enable the polylactic acid zwitterionic polymer to quickly form vesicles in the water phase, and the method is simple to operate and suitable for large-scale production.
According to an embodiment of the present disclosure, the organic solvent includes at least one of: acetone, tetrahydrofuran, N-dimethylformamide; the aqueous phase comprises at least one of: water, phosphate buffer.
According to the embodiment disclosed by the disclosure, the organic phase is formed by mixing a solvent with strong volatility and a protic solvent, so that the solubility of the amphoteric polymer solution in the solvent is improved, the using amount of the solvent is reduced, and the volatilization speed of the solvent is increased. The water phase adopts water or phosphate buffer solution, the subsequent treatment is simple and convenient, and the mass production is convenient.
According to the disclosed embodiment, the volume ratio of the organic phase to the aqueous phase comprises 1:5-1, for example, can be 1:5, 1.
By the embodiment of the disclosure, the volume ratio of the organic phase to the aqueous phase is controlled, so that the zwitterionic polymer can quickly form vesicles in the system.
Still further embodiments of the present disclosure provide a drug-loaded vesicle comprising the polymersome described above and a drug entrapped in the polymersome.
According to an embodiment of the present disclosure, the drug includes a hydrophobic drug and a hydrophilic drug. Hydrophobic drugs include, but are not limited to, paclitaxel. Hydrophilic drugs include, but are not limited to, doxorubicin hydrochloride.
According to embodiments of the present disclosure, the mass ratio of polymersome to drug includes 8:1-10, for example: 8:1, 9:1, 10.
According to the embodiment of the disclosure, the medicine is encapsulated in the hydrophobic shell layer or the hydrophilic inner core by the amphiphilic ionic polymer vesicle, so that the blood stability and the biocompatibility are good, and the effect of layer-by-layer release is achieved.
According to embodiments of the present disclosure, the particle size of the polymersome comprises 100-300nm, for example: 100nm, 200nm and 300nm.
Through the disclosed embodiment, the drug-loaded polymer vesicle has micron-sized particle size and is stable in blood circulation.
The present disclosure is described in further detail below with reference to specific examples.
Example 1
Weighing 80mg of polymer shown in formula (fifteen), and dissolving in 8ml of acetone to obtain a concentration of 10mg/ml;
Figure BDA0003233758170000101
100ml of PBS (phosphate buffered saline) was prepared in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring at 500rpm for 48 hours, volatilizing the organic solvent to form a polymer vesicle solution, and freeze-drying to obtain the polymer vesicles. Fig. 1 schematically shows an electron microscope image of the polymersome prepared in example 1.
Example 2
Weighing 80mg of the polymer shown in the formula (sixteen), and dissolving the polymer in 8ml of acetone to obtain a concentration of 10mg/ml;
Figure BDA0003233758170000102
preparing 100ml of water in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring at 500rpm for 48h, volatilizing the organic solvent to form a polymer vesicle solution, and freeze-drying to obtain the polymer vesicle. Fig. 2 schematically shows an electron microscope image of the polymersome prepared in example 2.
Example 3
Weighing 50mg of the polymer represented by the formula (seventeen), and dissolving the polymer in 10ml of acetone to obtain a solution with a concentration of 5mg/ml;
Figure BDA0003233758170000103
preparing 100ml of water in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring at 500rpm for 48h, volatilizing the organic solvent to form a polymer vesicle solution, and freeze-drying to obtain the polymer vesicle. FIG. 3 schematically shows an electron micrograph of polymersome prepared in example 3
Example 4
Weighing 50mg of the polymer shown in the formula (eighteen), and dissolving the polymer in 10ml of acetone to obtain a solution with the concentration of 5mg/ml;
Figure BDA0003233758170000111
preparing 100ml of water in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring at 500rpm for 48h, volatilizing the organic solvent to form a polymer vesicle solution, and freeze-drying to obtain the polymer vesicle. Fig. 4 schematically shows an electron microscope image of the polymersome prepared in example 4.
Example 5
Weighing 60mg of the polymer shown in the formula (nineteen), and dissolving the polymer in 10ml of acetone to obtain a concentration of 6mg/ml;
Figure BDA0003233758170000112
preparing 100ml of water in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring at 500rpm for 48h, volatilizing the organic solvent to form a polymer vesicle solution, and freeze-drying to obtain the polymer vesicle. FIG. 5 schematically shows an electron micrograph of polymersome prepared in example 5
Example 6
Weighing 60mg of the polymer shown in the formula (twenty), and dissolving the polymer in 10ml of acetone to obtain a concentration of 6mg/ml;
Figure BDA0003233758170000113
preparing 100ml of water in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring at 500rpm for 48 hours, volatilizing the organic solvent to form a polymer vesicle solution, and freeze-drying to obtain the polymer vesicles. FIG. 6 schematically shows an electron micrograph of polymersome prepared in example 6
Fig. 7 schematically shows the particle size distribution of the polymersome prepared in examples 1 to 6.
As shown in fig. 7, the maximum strength of the polymersome of example 1 may be 12, the maximum strength of the polymersome of examples 2 to 4 may be 14 to 15, the maximum strength of the polymersome of example 5 may be 17, and the maximum strength of the polymersome of example 6 may be 20. And the particle sizes of the maximum intensity of examples 1 to 6 were all distributed between 200 and 300nm.
Fig. 8 schematically shows the change in the average particle diameter of the polymersome prepared in examples 1 to 6 with time.
As shown in fig. 8, the average particle size of the polymersome in example 1 was varied from 200 to 250nm, the average particle size of the polymersome in example 2 was varied from 250 to 275nm, the polymersome in example 3 was varied from 200 to 220nm, the polymersome in example 4 was varied about 250nm, the polymersome in example 5 was varied from 240 to 275nm, and the polymersome in example 6 was varied from 210 to 250 nm. From this, it can be seen that the polymersomes of examples 1 to 6 were all relatively stable.
Fig. 9 schematically shows the results of the cytotoxicity test of the polymersome prepared in examples 1 to 6.
Inoculating hela cells in a 96-well plate, and culturing overnight by adopting a DMEM nutrient solution to ensure that the cells are completely attached to the wall; then changing the nutrient solution into DMEM nutrient solution containing 200ug/ml prepared polymersome, and continuing culturing for 24h; then washing with fresh DMEM nutrient solution for 3 times, adding DMEM nutrient solution containing 10% of CCK-8, and continuing culturing for 4h; and finally, testing the absorbance of the cells in the 96-well plate at 450nm by using an enzyme labeling instrument, and comparing the absorbance with a blank control group to obtain the survival rate data of the cells treated by the polymer vesicles. The blank control was cells that were not treated with polymersomes. Of these, CCK-8 reagent is redox by NAD + to a water-soluble yellow formazan product in the presence of an electron coupling reagent (i.e., when the cell is in a viable state). The more surviving cells produced formazan, the darker the color and the higher the absorbance, and thus the absorbance was considered to be proportional to the number of surviving cells
As shown in fig. 9, the cell survival rates of the polymersomes prepared in examples 1-6 and the control group were close to 100%, indicating that the polymersomes prepared in examples 1-6 had low toxicity.
According to some embodiments of the present disclosure, a drug-loaded vesicle is provided, wherein the polymer vesicle is used as a drug delivery carrier for encapsulating various drug analyses such as nucleic acid vaccines, amino acids, proteins and the like. Hereinafter, a polymer vesicle encapsulating paclitaxel is prepared by taking the hydrophobic drug paclitaxel as an example.
Example 7
Weighing 80mg of polymer shown in formula (fifteen) and 10mg of paclitaxel, and dissolving in 8ml of acetone;
prepare 100ml PBS in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring at 500rpm for 72h until the organic solvent is volatilized, thus forming the drug-loaded vesicle, and freeze-drying and storing.
Example 8
Weighing 80mg of polymer represented by formula (sixteen) and 10mg of paclitaxel, and dissolving in 8ml of acetone;
prepare 100ml PBS in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring at 500rpm for 72h until the organic solvent is volatilized, thus forming the drug-loaded vesicle, and freeze-drying and storing.
Example 9
Weighing 80mg of polymer shown in formula (eighteen) and 10mg of paclitaxel, and dissolving in 8ml of acetone;
prepare 100ml PBS in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring at 500rpm for 72h until the organic solvent is volatilized, so as to form the drug-loaded vesicle, and freeze-drying and storing.
Example 10
Weighing 80mg of polymer shown in formula (nineteen), and 10mg of paclitaxel, and dissolving in 8ml of acetone;
prepare 100ml PBS in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring at 500rpm for 72h until the organic solvent is volatilized, thus forming the drug-loaded vesicle, and freeze-drying and storing.
The drug-loaded vesicles prepared in examples 7-10 were subjected to HPLC to determine the paclitaxel loading and encapsulation efficiency, as follows:
accurately weighing 5mg of drug-loaded nanoparticles, dissolving the drug-loaded nanoparticles in 5ml of acetonitrile, adding ultrapure water to fix the volume to 10ml, measuring the peak area of paclitaxel in the solution by using HPLC, and calculating the drug loading rate and the encapsulation rate according to the following formula by combining a paclitaxel standard curve:
Figure BDA0003233758170000131
Figure BDA0003233758170000141
the test results are shown in table 2:
table 2 paclitaxel drug loading and encapsulation efficiency of drug-loaded vesicles prepared in examples 7 to 10
Drug-loaded vesicle Drug loading (%) Encapsulation efficiency (%) Average particle diameter (nm)
Example 7 4.62 41.03 210.4
Example 8 5.34 51.34 228.9
Example 9 7.81 58.12 301.6
Example 10 5.18 49.25 234.6
As can be seen from the test results in table 2, the hydrophobic drug paclitaxel has been successfully encapsulated in the drug-loaded vesicles of examples 7 to 10.
Fig. 10 schematically shows the in vitro drug release profile of the drug-loaded vesicles of examples 7-10.
10mg of the drug-loaded vesicles obtained in examples 7 to 10 were weighed, dispersed in 1mL of pH =7.4PBS, and added to a dialysis bag (M) W = 4000), the dialysis bag is placed in 20mL of pH =7.4PBS containing 0.5% tween-80, stirred constantly at 37 ℃, and at the indicated time points, 5mL of release medium is removed from the release system and then an equal volume of fresh medium is replenished. Drug released and no drug released were quantified by HPLC.
The test result is shown in fig. 10, the drug release amount of the drug-loaded vesicle prepared in example 7 gradually increases to 44% within 12h, slowly increases to 45% between 12 and 24h, and gradually levels to 45% between 24 and 48h, and is basically stabilized at 45%. The drug release amount of the drug-loaded vesicle prepared in example 8 gradually increases to 42% within 12, slowly increases to 46% between 12 and 24, and gradually becomes flat and stable at 46% between 24 and 48 hours. The drug release amount of the drug-loaded vesicle prepared in example 9 gradually increases to 47% within 12, slowly increases to 52% between 12 and 24, and gradually levels to 52% between 24 and 48h, and is basically stabilized at 52%. The drug release amount of the drug-loaded vesicle prepared in the embodiment 10 gradually increases to 38% within 12, slowly increases to 40% between 12 and 24, and gradually becomes gentle and substantially stable at 40% between 24 and 48 hours.
Hereinafter, taking hydrophilic drug doxorubicin hydrochloride as an example, a polymersome loaded with doxorubicin hydrochloride was prepared.
Example 11
Weighing 80mg of polymer shown in formula (fifteen) and 10mg of doxorubicin hydrochloride, and dissolving in 8ml of tetrahydrofuran/methanol (volume ratio of 3:1) mixed solution;
prepare 100ml PBS in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring for 72h at 500rmp, and volatilizing the organic solvent to form the drug-loaded vesicle.
The prepared drug-loaded vesicle solution is loaded into PBS for dialysis for 12h to remove unencapsulated doxorubicin hydrochloride.
Example 12
Weighing 80mg of polymer shown in formula (sixteen) and 10mg of doxorubicin hydrochloride, and dissolving in 8ml of tetrahydrofuran/methanol (3:1 in volume ratio) mixed solution;
prepare 100ml PBS in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring for 72h at 500rmp, and volatilizing the organic solvent to form the drug-loaded vesicle.
The prepared drug-loaded vesicle solution is loaded into PBS for dialysis for 12h to remove unencapsulated doxorubicin hydrochloride.
Example 13
Weighing 80mg of polymer shown in formula (eighteen) and 10mg of doxorubicin hydrochloride, and dissolving in 8ml of tetrahydrofuran/methanol (volume ratio of 3:1) mixed solution;
prepare 100ml PBS in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring for 72h at 500rmp, and volatilizing the organic solvent to form the drug-loaded vesicle.
The prepared drug-loaded vesicle solution is loaded into PBS for dialysis for 12h to remove unencapsulated doxorubicin hydrochloride.
Example 14
Weighing 80mg of polymer shown in formula (nineteen), 10mg of doxorubicin hydrochloride, and dissolving in 8ml of tetrahydrofuran/methanol (3:1 in volume ratio);
prepare 100ml PBS in a beaker;
and (3) dropwise adding the organic phase into the water phase under continuous stirring, stirring for 72h at 500rmp, and volatilizing the organic solvent to form the drug-loaded vesicle.
The prepared drug-loaded vesicle solution is loaded into PBS for dialysis for 12h to remove unencapsulated doxorubicin hydrochloride.
The drug-loaded vesicles prepared in examples 11 to 14 were measured for doxorubicin hydrochloride drug-loaded amount by an absorbance method. The specific method comprises the following steps:
dissolving in tetrahydrofuran/methanol (V/V = 3:1) mixed solvent, diluting in a multiple ratio to obtain a series of gradient solutions, measuring the absorption values of the solutions at 485nm by an ultraviolet spectrophotometer (UV-vis, shimadzu) to obtain the corresponding relation between the concentration of the doxorubicin hydrochloride solution and the ultraviolet absorption value, and drawing a standard curve of the doxorubicin hydrochloride in the tetrahydrofuran/methanol (V/V = 3:1) mixed solvent.
The drug loading rate and the encapsulation efficiency are calculated by using the absorption of the nanoparticle solution at 485 nm. Freeze-drying the prepared PBS solution of the medicine-carrying vesicle, dissolving the medicine-carrying vesicle in 8mL tetrahydrofuran/methanol (V/V = 3:1) mixed solvent, and finally measuring the 485nm absorption value by a spectrophotometer, and measuring the medicine-carrying quantity according to the doxorubicin hydrochloride standard curve.
Figure BDA0003233758170000161
Figure BDA0003233758170000162
The test results are shown in table 3:
table 3 paclitaxel drug loading and encapsulation efficiency of drug-loaded vesicles prepared in examples 11-14
Drug-loaded vesicle Drug loading (%) Encapsulation efficiency (%) Average particle diameter (nm)
Example 11 3.78 21.74 228.4
Example 12 2.15 16.56 245.6
Example 13 2.56 17.68 278.7
Example 14 4.28 39.25 258.9
As can be seen from table 3, the hydrophilic drug doxorubicin hydrochloride was successfully encapsulated into the polymersome.
Fig. 11 schematically shows the in vitro drug release profile of the drug-loaded vesicles of examples 11-14.
The drug-loaded vesicles prepared in examples 11 to 14 were each weighed and dispersed in 1mL of ph =7.4PBS, and added to a dialysis bag (M) W = 4000), the dialysis bag was placed in 20mL ph =7.4PBS, stirred continuously at 37 ℃, and at the indicated time point, 5mL of release medium was taken out of the release system, and then an equal volume of fresh medium was replenished. The released drug was estimated by the absorbance of the solution at 485 nm.
The test result is shown in fig. 11, the drug release amount of the drug-loaded vesicle prepared in example 11 gradually increases to 15% within 12h, slowly increases to 20% between 12h and 24h, and slowly increases to 30% between 24h and 48 h. The drug-loaded vesicle prepared in example 12 gradually increases the drug release amount to 24% within 12, gradually increases the drug release amount to 35% between 12 and 24 hours, and slowly increases the drug release amount to 45% between 24 and 48 hours. The drug-loaded vesicle prepared in example 13 gradually increases the drug release amount to 35% within 12, gradually increases the drug release amount to 40% between 12 and 24, and slowly increases the drug release amount to 45% between 24 and 48 hours. The drug-loaded vesicle prepared in example 14 gradually increases the drug release amount to 35% within 12, gradually increases the drug release amount to 45% between 12 and 24 hours, and slowly increases the drug release amount to 50% between 24 and 48 hours.
In summary, the polymersome provided in the embodiments of the present disclosure serves as a drug carrier, and can stably release a drug.
The above-mentioned embodiments are intended to illustrate the objects, aspects and advantages of the present disclosure in further detail, and it should be understood that the above-mentioned embodiments are only illustrative of the present disclosure and are not intended to limit the present disclosure, and any modifications, equivalents, improvements and the like made within the spirit and principle of the present disclosure should be included in the scope of the present disclosure.

Claims (7)

1. A polymersome, comprising: a self-assembly of polylactic acid zwitterionic polymer,
the structural formula of the polylactic acid zwitterionic polymer comprises any one of the following formulas (fifteen) to (twenty):
Figure FDA0003833666110000011
Figure FDA0003833666110000021
the preparation method of the polymersome comprises the following steps:
dissolving the polylactic acid zwitterionic polymer in an organic solvent to obtain an organic phase;
and adding the organic phase into the water phase under continuous stirring, and stirring until the organic solvent is volatilized to obtain the polymer vesicle.
2. A method of making the polymersome of claim 1, comprising:
dissolving the polylactic acid zwitterionic polymer in an organic solvent to obtain an organic phase;
and adding the organic phase into the water phase under continuous stirring, and stirring until the organic solvent is volatilized to obtain the polymer vesicle.
3. The method of claim 2, wherein,
the organic solvent includes at least one of: acetone, tetrahydrofuran, N-dimethylformamide;
the aqueous phase comprises at least one of: water, phosphate buffer.
4. The method of claim 2, wherein the volume ratio of the organic phase to the aqueous phase is 1:5-1.
5. A drug-loaded vesicle comprising: the polymersome of claim 1 and a drug entrapped in the polymersome;
wherein the medicament comprises any one of the following: paclitaxel, and doxorubicin hydrochloride.
6. The drug-loaded vesicle of claim 5, wherein the mass ratio of the polymervesicle to the drug is 8:1-10.
7. The drug-loaded vesicle of claim 5, wherein the polymersome has a particle size of 100-300nm.
CN202111000584.9A 2021-08-27 2021-08-27 Polymer vesicle and preparation method thereof, and drug-loaded vesicle Active CN113694027B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111000584.9A CN113694027B (en) 2021-08-27 2021-08-27 Polymer vesicle and preparation method thereof, and drug-loaded vesicle

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111000584.9A CN113694027B (en) 2021-08-27 2021-08-27 Polymer vesicle and preparation method thereof, and drug-loaded vesicle

Publications (2)

Publication Number Publication Date
CN113694027A CN113694027A (en) 2021-11-26
CN113694027B true CN113694027B (en) 2022-12-30

Family

ID=78656455

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111000584.9A Active CN113694027B (en) 2021-08-27 2021-08-27 Polymer vesicle and preparation method thereof, and drug-loaded vesicle

Country Status (1)

Country Link
CN (1) CN113694027B (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101390826A (en) * 2008-10-24 2009-03-25 中山大学 Magnetic tumour target polymer nano vesicle and preparation method thereof
CN101953778A (en) * 2010-09-30 2011-01-26 中国医学科学院生物医学工程研究所 Folate-targeted hydrophobic medicine-loaded polymeric vesicle and preparation method and use thereof
CN107513145A (en) * 2017-08-23 2017-12-26 同济大学 The synthesis of biodegradable amphiphilic block class antibacterial peptide and the preparation method and application of class antibacterial peptide vesica

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104667286B (en) * 2015-01-30 2019-04-23 四川大学 A kind of size monodisperse polymer nano vesicle and its preparation method and application

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101390826A (en) * 2008-10-24 2009-03-25 中山大学 Magnetic tumour target polymer nano vesicle and preparation method thereof
CN101953778A (en) * 2010-09-30 2011-01-26 中国医学科学院生物医学工程研究所 Folate-targeted hydrophobic medicine-loaded polymeric vesicle and preparation method and use thereof
CN107513145A (en) * 2017-08-23 2017-12-26 同济大学 The synthesis of biodegradable amphiphilic block class antibacterial peptide and the preparation method and application of class antibacterial peptide vesica

Also Published As

Publication number Publication date
CN113694027A (en) 2021-11-26

Similar Documents

Publication Publication Date Title
Chan et al. PLGA–lecithin–PEG core–shell nanoparticles for controlled drug delivery
Dou et al. Preparation and evaluation in vitro and in vivo of docetaxel loaded mixed micelles for oral administration
Riley et al. Colloidal stability and drug incorporation aspects of micellar-like PLA–PEG nanoparticles
Avgoustakis Pegylated poly (lactide) and poly (lactide-co-glycolide) nanoparticles: preparation, properties and possible applications in drug delivery
Suksiriworapong et al. Development and characterization of lyophilized diazepam-loaded polymeric micelles
Li et al. PEG–PLA diblock copolymer micelle-like nanoparticles as all-trans-retinoic acid carrier: in vitro and in vivo characterizations
Zhang et al. Pullulan acetate nanoparticles prepared by solvent diffusion method for epirubicin chemotherapy
Numata et al. A slow-release system of bacterial cellulose gel and nanoparticles for hydrophobic active ingredients
Shi et al. Stealth MePEG-PCL micelles: effects of polymer composition on micelle physicochemical characteristics, in vitro drug release, in vivo pharmacokinetics in rats and biodistribution in S 180 tumor bearing mice
Varan et al. Development of polycationic amphiphilic cyclodextrin nanoparticles for anticancer drug delivery
Tao et al. Reduction-responsive gold-nanoparticle-conjugated Pluronic micelles: an effective anti-cancer drug delivery system
CN105833287B (en) Slow-release nano-drug carrier and preparation method and application thereof
Yadav et al. Long circulating nanoparticles of etoposide using PLGA‐MPEG and PLGA‐pluronic block copolymers: characterization, drug‐release, blood‐clearance, and biodistribution studies
Zhang et al. PEGylated nanostructured lipid carriers loaded with 10‐hydroxycamptothecin: an efficient carrier with enhanced anti‐tumour effects against lung cancer
KR100837860B1 (en) Hydrophilic polymer nanocapsules and method for preparing the same
Jain et al. Lyotropic liquid crystalline nanoparticles of amphotericin B: Implication of phytantriol and glyceryl monooleate on bioavailability enhancement
Das et al. Preparation of a size selective nanocomposite through temperature assisted co-assembly of gelatin and pluronic F127 for passive targeting of doxorubicin
Nguyen et al. Cell interaction studies of PLA-MePEG nanoparticles
Lee et al. Ionically fixed polymeric nanoparticles as a novel drug carrier
Pippa et al. Preparation, development and in vitro release evaluation of amphotericin B–loaded amphiphilic block copolymer vectors
Liang et al. 5-Fluorouracil-loaded self-assembled pH-sensitive nanoparticles as novel drug carrier for treatment of malignant tumors
Liaskoni et al. Paclitaxel controlled delivery using a pH-responsive functional-AuNP/block-copolymer vesicular nanocarrier composite system
CN113694027B (en) Polymer vesicle and preparation method thereof, and drug-loaded vesicle
Li et al. A novel monomethoxy polyethylene glycol–polylactic acid polymeric micelles with higher loading capacity for docetaxel and well-reconstitution characteristics and its anti-metastasis study
CA2746555A1 (en) Nanoparticle compositions

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