CN113786492B - Polymer carrier for photodynamic therapy and preparation method and application thereof - Google Patents

Polymer carrier for photodynamic therapy and preparation method and application thereof Download PDF

Info

Publication number
CN113786492B
CN113786492B CN202110932558.3A CN202110932558A CN113786492B CN 113786492 B CN113786492 B CN 113786492B CN 202110932558 A CN202110932558 A CN 202110932558A CN 113786492 B CN113786492 B CN 113786492B
Authority
CN
China
Prior art keywords
cell membrane
parts
tumor
polymer
glycosyl
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
CN202110932558.3A
Other languages
Chinese (zh)
Other versions
CN113786492A (en
Inventor
罗奎
罗强
段振宇
龚启勇
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.)
West China Hospital of Sichuan University
Original Assignee
West China Hospital of Sichuan University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by West China Hospital of Sichuan University filed Critical West China Hospital of Sichuan University
Priority to CN202110932558.3A priority Critical patent/CN113786492B/en
Publication of CN113786492A publication Critical patent/CN113786492A/en
Application granted granted Critical
Publication of CN113786492B publication Critical patent/CN113786492B/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
    • 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
    • A61K47/58Medicinal 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 by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly[meth]acrylate, polyacrylamide, polystyrene, polyvinylpyrrolidone, polyvinylalcohol or polystyrene sulfonic acid resin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0057Photodynamic therapy with a photosensitizer, i.e. agent able to produce reactive oxygen species upon exposure to light or radiation, e.g. UV or visible light; photocleavage of nucleic acids with an agent
    • A61K41/0076PDT with expanded (metallo)porphyrins, i.e. having more than 20 ring atoms, e.g. texaphyrins, sapphyrins, hexaphyrins, pentaphyrins, porphocyanines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5176Compounds of unknown constitution, e.g. material from plants or animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F8/00Chemical modification by after-treatment
    • C08F8/30Introducing nitrogen atoms or nitrogen-containing groups
    • C08F8/32Introducing nitrogen atoms or nitrogen-containing groups by reaction with amines
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • C12N2509/10Mechanical dissociation

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Botany (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Optics & Photonics (AREA)
  • Molecular Biology (AREA)
  • Oncology (AREA)
  • Nanotechnology (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Polymers & Plastics (AREA)
  • General Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides a novel processA polymer carrier for photodynamic therapy and a preparation method and application thereof belong to the technical field of drug delivery systems. Specifically provides a cell membrane-glycosyl polymer, which is formed by wrapping a layer of cell membrane outside the glycosyl polymer with the structure shown in formula I. The polymer has good photodynamic effect, good targeting effect when being used as a photosensitizer, can be gathered at a tumor part, and has good photodynamic treatment effect and strong killing effect on tumor cells. In addition, the polymer has excellent entrapment effect on the drug as a polymer carrier, and the drug loading is high; the tumor drug encapsulated by the coating has excellent tumor treatment effect and excellent application prospect.
Figure DDA0003211650020000011

Description

Polymer carrier for photodynamic therapy and preparation method and application thereof
Technical Field
The invention belongs to the technical field of drug delivery systems, and particularly relates to a polymer carrier for photodynamic therapy, and a preparation method and application thereof.
Background
Cancer (cancer) refers to a malignant tumor that originates in epithelial tissue. The data show that in 2020, there are 1929 new cancer cases worldwide, of which 1006 ten thousand cases in men and 923 ten thousand cases in women; 996 ten thousand cases of cancer death worldwide in 2020, of which 553 thousand in men and 443 ten thousand in women. The incidence rate and the mortality rate of cancer are high, and the cancer becomes one of global diseases seriously threatening human health, and is one of the hot spots and difficulties of the current basic and clinical research. According to the latest data in 2020, the first ten cancers with global incidence are respectively breast cancer, lung cancer, colorectal cancer, prostate cancer, stomach cancer, liver cancer, cervical cancer, esophageal cancer, thyroid cancer and bladder cancer. The current treatment modes of cancer mainly comprise surgery, radiotherapy, chemotherapy, immunotherapy and the like. Although the early treatment effect of the treatment modes is better, the problems of tumor metastasis, easy relapse, body injury, immune damage and the like still exist.
In recent years, photodynamic therapy (PDT) has been used as an emerging treatment modality. Photodynamic therapy induces apoptosis of tumor cells and causes DNA damage through Reactive Oxygen Species (ROS) generated by photosensitizers after specific laser irradiation. PDT has the advantages of high selectivity, low systemic toxic and side effects, repeated treatment, and difficulty in generating tolerance, and is receiving more and more attention from researchers. However, most photosensitizers used in PDT have problems of poor water solubility, lack of targeting property, low ROS yield, etc., insufficient accumulation at tumor sites, weak tumor killing effect, and limited application in cancer therapy.
How to increase the concentration of photosensitizer at the tumor site, enhance the killing effect of PDT on tumor, and enhance the effect of PDT on cancer treatment is an urgent need in the current cancer treatment.
Disclosure of Invention
In order to solve the above problems, the present invention provides a polymer carrier useful for photodynamic therapy, a method for preparing the same, and use thereof.
The invention provides a cell membrane-glycosyl polymer, which is formed by wrapping a layer of cell membrane outside a glycosyl polymer with a structure shown in a formula I:
Figure BDA0003211650000000021
formula I
Wherein the content of the first and second substances,
(x + y + z) o: p: n: m in a molar ratio of 2.9;
r is selected from
Figure BDA0003211650000000022
Or->
Figure BDA0003211650000000023
Further, the cell membrane is a cell membrane of a tumor cell;
and/or the glycosyl polymer is prepared from the following raw materials in parts by weight: B-gal-SH 100-500 parts, maleimide-Ppa-100 parts;
the structural formula of the Maleimide-Ppa is as follows:
Figure BDA0003211650000000024
/>
the B-gala-SH is prepared from the following raw materials in parts by weight: 1000-1500 parts of B-gal-PySS and 1000-1500 parts of dithiothreitol;
the B-gala-PySS is prepared from the following raw materials in parts by weight: 1000-1500 parts of MA-D-Galactosamine, 100-200 parts of MA-PySS, 10-50 parts of MA-GFLGKGLFG-MA and 50-100 parts of MA-GFLGK-CTA;
the MA-D-Galactosamine has the structure
Figure BDA0003211650000000031
The structure of the MA-PySS is
Figure BDA0003211650000000032
The MA-GFLGKGLFG-MA has the structure
Figure BDA0003211650000000033
The structure of the MA-GFLGK-CTA is
Figure BDA0003211650000000034
Further, the glycosyl polymer is prepared from the following raw materials in parts by weight: B-gala-SH 500 parts, maleimide-Ppa parts;
and/or the B-gala-SH is prepared from the following raw materials in parts by weight: 1300 parts of B-gal-PySS and 1000 parts of dithiothreitol;
and/or the B-gala-PySS is prepared from the following raw materials in parts by weight: 1446 parts of MA-D-Galactosamine, 100 parts of MA-PySS, 23.5 parts of MA-GFLGKGLFG-MA and 53 parts of MA-GFLGK-CTA.
Further, the preparation method of the cell membrane comprises the following steps:
extruding the cells by a miniature extruder until the cells are destroyed, centrifuging, and taking supernate containing cell membranes to obtain the cell-free cell culture medium;
preferably, the preparation method of the cell membrane comprises the following steps:
preparing PBS cell suspension containing protease inhibitor, adopting Avanit micro extruder without polycarbonate porous membrane to press cells until the cells are destroyed, centrifuging, and taking supernatant containing cell membrane to obtain the product.
Further, the preparation method of the B-gal-SH comprises the following steps:
reacting B-gala-PySS with dithiothreitol to obtain the product.
Further, the preparation method of the B-gala-PySS comprises the following steps:
dissolving MA-D-Galactosamine, MA-PySS, MA-GFLGKGLFG-MA and MA-GFLGK-CTA in solvent, reacting under the action of initiator, and lyophilizing to obtain the final product;
preferably, the solvent is a mixed solution of water and methanol;
and/or, oxygen is removed prior to the reaction;
and/or the reaction is a reaction protected from light;
more preferably, the water to methanol volume ratio is 1:4;
and/or the initiator is VA044;
and/or the reaction is carried out in an oil bath at the temperature of between 40 and 50 ℃ for 10 to 12 hours.
Further, the method for preparing the glycosyl polymer comprises the following steps:
(1) Dissolving B-gal-SH in water, adding DMSO, and preparing into B-gal-SH/DMSO mixed solution;
(2) Dissolving Maleimide-Ppa in DMSO, and adding the dissolved solution into a B-gala-SH/DMSO mixed solution for reaction;
(3) Purifying the reaction solution obtained in the step (2) to obtain the compound;
preferably, the first and second electrodes are formed of a metal,
in the step (1), the volume ratio of the water to the DMSO is 1: (1-5);
and/or, in the step (2), the reaction is carried out at room temperature in a dark place.
The present invention also provides a method for preparing the aforementioned cell membrane-glycosyl polymer, which comprises the steps of:
mixing the cell membrane with a glycosyl polymer aqueous solution, and centrifuging to obtain the cell membrane-glycosyl polymer aqueous solution;
preferably, the mixture is stirred overnight at a temperature of 0 to 4 ℃;
and/or, the centrifugation is 20000g for 30min.
The invention also provides the application of the cell membrane-glycosyl polymer in preparing a photosensitizer medicament;
preferably, the photosensitizer is a drug for photodynamic therapy of tumors.
The invention also provides the application of the cell membrane-glycosyl polymer in preparing a drug carrier;
preferably, the drug used for entrapment in the drug carrier is an anti-tumor drug;
more preferably, the anti-tumor drug is olaparib.
The invention also provides a medicament which is a preparation prepared by taking the cell membrane-glycosyl polymer as an active ingredient, or taking the cell membrane-glycosyl polymer as a carrier to entrap the medicament as the active ingredient and adding pharmaceutically acceptable auxiliary materials or auxiliary ingredients;
preferably, the entrapped drug is an anti-tumor drug;
more preferably, the entrapped drug is olaparib.
The invention provides a cell membrane-glycosyl polymer, which overcomes the defects of insufficient accumulation at a tumor part and weak killing effect on tumors of the existing PDT photosensitizer, can be accumulated at the tumor part, and has good targeting effect; it has photodynamic effect, high ROS generating efficiency when used as photosensitizer for photodynamic therapy, and strong killing effect on tumor cells. Meanwhile, the cell membrane-glycosyl polymer can also be used as a drug delivery system to entrap drugs, particularly tumor drugs such as olaparib, and is used for treating tumors with excellent effect. The cell membrane-glycosyl polymer has excellent application prospect in preparing medicaments for treating tumors.
Obviously, many modifications, substitutions, and variations are possible in light of the above teachings of the invention, without departing from the basic technical spirit of the invention, as defined by the following claims.
The present invention will be described in further detail with reference to the following examples. This should not be understood as limiting the scope of the above-described subject matter of the present invention to the following examples. All the technologies realized based on the above contents of the present invention belong to the scope of the present invention.
Drawings
FIG. 1 shows the structural formula of cathepsin B sensitive functionalized chain transfer agent MA-GFLGK-CTA and the preparation route thereof.
FIG. 2 shows the structural formula of cathepsin B sensitive functional cross-linking agent MA-GFLGKGLFG-MA and the preparation route thereof.
FIG. 3 is a schematic diagram of the synthetic route and structure of Branched-GFLG-poly-D-galactosamine-S-Ppa (BSP).
FIG. 4 shows the hydrogen spectrum of the polymer precursor Branche-GFLG-poly D-galactochitosan-PySS (solvent D6-DMSO).
FIG. 5 shows the hydrogen spectrum of the polymer precursor Branched-GFLG-poly D-galactamine-SH (solvent is D6-DMSO).
FIG. 6 shows the hydrogen spectrum of polymer Branche-GFLG-poly D-galactochitosan-S-Ppa (solvent is D6-DMSO).
FIG. 7 is a TEM image of the cell membrane and the CM-BSPO formed after the BSPO is modified by the cell membrane: a is a TEM picture of the cell membrane; b is TEM picture of CM-BSPO formed after BSPO is modified by cell membrane.
FIG. 8 shows the structural formula of Maleimide-hexyl-Ppa and its synthetic route.
FIG. 9 shows the hydrogen spectrum of the polymer Branched-GFLG-poly D-galactochitosan-S-hexyl-Ppa (solvent is D6-DMSO).
FIG. 10 is a synthetic route for a linear glycopolymer photodynamic therapy system.
FIG. 11 shows the hydrogen spectrum of the polymer precursor Linear-poly D-galactochitosan-PySS (solvent D) 2 O)。
FIG. 12 shows the hydrogen spectrum of the polymer precursor Linear-poly D-galactochitosan-SH (solvent D6-DMSO).
FIG. 13 shows the hydrogen spectrum of polymer Linear-poly D-galactochitosan-S-Ppa (solvent is D6-DMSO).
Fig. 14 is a synthetic route for a HPMA-based degradable branched/crosslinked polymer photodynamic therapy system.
FIG. 15 shows the hydrogen spectrum of Branche-GFLG-poly HPMA-PySS polymer precursor (solvent D) 2 O)。
FIG. 16 shows a hydrogen spectrum of Branche-GFLG-poly HPMA-SH polymer precursor (solvent D) 2 O)。
FIG. 17 shows the hydrogen spectrum of polymer Branched-GFLG-poly HPMA-S-Ppa (solvent D) 2 O)。
FIG. 18 shows the chemical structures, self-assembly diagrams and TEM results of BSP, BShP and LSP (scale: 200 nm).
Fig. 19 shows DLS detection results of BSP, BShP, and LSP: a is a particle size detection result; b is zeta potential detection result.
FIG. 20 shows the change of fluorescence intensity with increasing laser irradiation time when a singlet oxygen fluorescent probe (SOSG) is added to BSP, BShP, and LSP polymer solutions.
FIG. 21 shows the results of in vitro cytotoxicity assays for BSP, BShP, and LSP.
FIG. 22 shows the statistical results of fluorescence signals of the images and tumor sites of the three groups of BSP, LSP and BShP polymers in the tumor mouse model: a is a distribution image; b is the fluorescent signal statistical result of the tumor part.
FIG. 23 shows the content of Olaparib in polymer LSP, BSP, BShP and BHSP by HPLC measurement, which is calculated by a standard curve; HRMS results show that the encapsulated olaparib is a raw drug and does not influence the activity of the olaparib.
FIG. 24 is a graph showing the results of the fluorescence intensity of the tumor site after tail vein injection of different polymers at different time points.
FIG. 25 is a graph showing the results of the fluorescence intensity of the tumor site after tail vein injection of different polymers.
FIG. 26 is a tumor treatment flowchart of the mouse tumor model.
FIG. 27 shows the tumor volume changes in groups of mice for different treatment modalities.
Figure 28 summarizes the change in tumor volume in mice after different treatments (. Star.p < 0.01).
FIG. 29 shows tumor inhibition rates of mice of different treatment groups.
Fig. 30 shows the tumor volume magnetic resonance image (red circles indicate tumors), lung CT image (white arrows indicate lung metastases) and lung tissue specimen (black circles indicate lung metastases) for each group of illumination groups.
Fig. 31 shows the tumor volume magnetic resonance image (red circles indicate tumors), lung CT image (white arrows indicate lung metastases) and lung tissue specimen (black circles indicate lung metastases) for each group of non-illuminated groups.
FIG. 32 is HE staining of lungs of mice in different treatment groups, and red arrows indicate metastases (scale: 200 μm).
FIG. 33 shows the tumor CD31 immunohistochemical staining (scale: 200 μm) after treatment of the different groups.
FIG. 34 shows Ki-67 immunohistochemical staining of tumors after treatment in different groups (scale: 200 μm).
FIG. 35 statistical analysis of Ki-67 immunohistochemical staining for each group of tumors (. Star. P.)<0.01, ns No significant difference).
Detailed Description
The raw materials and equipment used in the embodiment of the present invention are known products and obtained by purchasing commercially available products.
Murine 4T1 breast cancer cell lines were purchased from cell banks of Chinese academy of sciences (Shanghai) and cultured in RPMI 1640 medium containing 1% diabody and 10% fetal bovine serum under 5% CO 2 95% air, 37 ℃ and constant humidity environment. All animal experiments were performed strictly according to the animal study guidelines approved by the ethical committee of the hospital. Experimental female Balb/c mice were purchased from Duoduosho laboratory animals Ltd. Establishment of subcutaneous tumor tumors with 4T1 cellsModels, and for in vivo distribution and imaging studies.
Example 1 preparation of sugar-based polymers according to the invention
The compounds Maleimide-Ppa, MA-PySS and MA-D-galactosamine were synthesized according to the methods reported in documents 1 and 2.
The synthesis of Maleimide-Ppa and MA-PySS is reported in document 1 (Pan D, zheng X, zhang Q, et al. Degraded-Polymer Disturbating Cells' Stress Protection by Targeting strategies [ J ]. Advanced Materials,2020, 1907490.), along the following synthetic routes:
Maleimide-Ppa:
Figure BDA0003211650000000061
MA-PySS:
Figure BDA0003211650000000062
the Synthesis of MA-D-galactosamine is reported in literature 2 (Wartchow C A, wang P, bednarski M D, et al. Carbohydrate Protease Conjugates: stabilized proteins for Peptide Synthesis [ J ]. The Journal of Organic Chemistry,1995, 60.
The structure of Maleimide-Ppa is:
Figure BDA0003211650000000063
wherein Ppa is the photosensitizer Pyropheophorbide-a (Pyropheophorbide a) moiety. The structure of MA-Pyss is: />
Figure BDA0003211650000000064
The MA-D-galactamine has the structure as follows: />
Figure BDA0003211650000000065
1. Preparation of enzyme-sensitive functionalized chain transfer agent MA-GFLGK-CTA
The synthetic route of the enzyme-sensitive functionalized chain transfer agent MA-GFLGK-CTA is shown in FIG. 1. MA-GFLGK-CTA was synthesized by a similar method reported in reference 3 (reference 3. The specific synthesis method comprises the following steps:
MA-GFLG-OH (4.6g, 10mmol), HOBt (1.49g, 11mmol) and HBTU (4.26g, 11mmol) as a condensing agent were placed in a round-bottom flask, and dissolved by adding ultra-dry DMF (50 mL) under nitrogen protection in ice bath. DIPEA (6.7mL, 40mmol) was added dropwise to the system and allowed to react for 0.5 hour. Weighing H-Lys (O) t Bu) -Fmoc & HCl (4.62g, 10 mmol) was added to the system, and the mixture was reacted for 0.5 hour in an ice bath and then returned to room temperature for 10 hours. Dissolving in 450mL Ethyl Acetate (EA), washing with saturated sodium bicarbonate solution (100 mL × 3), 1M dilute hydrochloric acid (100 mL × 3) and saturated sodium chloride solution (100 mL × 3), collecting organic phase, drying with anhydrous magnesium sulfate, concentrating, standing at 4 deg.C to obtain crystalline white solid MA-GFLGK (O-GFLGK) t Bu) -NHFmoc (6.51g, 7.51mmol, 75.1% yield). 1 H NMR(400MHz,δin ppm), 13 C NMR(100MHz,d 6 -DMSO,δin ppm),LC-MS(ES+):m/z=867.2[M+H] + 。MALDI-HRMS:m/z=889.4474[M+Na] +
In ice bath, MA-GFLGK (O) t Bu) -NHFmoc (4.33g, 5 mmol) was dissolved with 40mL of a mixed solution of DCM/TFA (v: v = 1:9) and reacted back to room temperature. TLC monitoring, after the reaction was complete the solvent was removed by rotary evaporation, and after addition of ether a white solid powder precipitated which was washed twice with ether and dried to give the product MA-GFLGK (OH) -NHFmoc (3.61g, 4.45mmol, 89.0% yield). 1 H NMR(400MHz,δin ppm), 13 C NMR(100MHz,d 6 -DMSO,δin ppm),LC-MS(ES+):m/z=811.3[M+H] + ,834.3[M+Na] + ,MALDI-HRMS:m/z=833.3842[M+Na] +
Using MA-GFLGK (OH) -NHFmoc as raw material, adopting solid phase polypeptide synthesis method, synthesizing functional monomer MA-GFLGK-CT by the following stepsA. MA-GFLGK (OH) -NHFmoc (1.62g, 2mmol) and DIPEA (0.67mL, 4mmol) were dissolved in 10mL of DMF and reacted with chlorotrityl chloride resin (5.0 g, 1.15mmol/g) for 2 hours. The resin was transferred to a polypropylene tube, washed 3 times with a mixed solution of DCM: meOH: DIPEA (v: v: v = 17. The Fmoc protecting group was treated three times with 50mL of 20% piperidine in DMF and cyanovaleric acid dithiobenzoic acid (CTA-COOH, 2.79g, 10mmol), DIC (1.26g, 10mmol) and HOBt (1.35g, 10mmol) were added and reacted for 12 hours. The product was treated with TFE/DCM (v: v = 3:7) at room temperature for 2 hours and the resin was removed by filtration. The mother liquor was concentrated under reduced pressure, dissolved in a little methanol and precipitated and purified by adding ether, and further purified by high performance liquid chromatography to obtain MA-GFLGK-CTA (892mg, 1.05mmol, 52.5% yield) as a pink solid product. 1 H NMR(400MHz,δin ppm), 13 C NMR(100MHz,d 6 -DMSO,δin ppm),LC-MS(ES+):m/z=851.3[M+H] + 。MALDI-HRMS:m/z=848.3408[M-H] - ,870.3181[M+Na-2H] -
2. Preparation of enzyme-sensitive functional cross-linking agent MA-GFLGKGLFG-MA
The synthesis route of the cathepsin B sensitive functional cross-linking agent MA-GFLGKGLFG-MA is shown in figure 2. MA-GFLGKGLFG-MA was synthesized by a similar method as reported in reference 3 (reference 3. The specific synthesis method comprises the following steps:
BocNH-GFLG-OH (2.46g, 5 mmol), HOBt (743mg, 5.5 mmol) and HBTU (2.13g, 5.5 mmol) were placed in a round bottom flask and 20mL of ultra dry DMF was added under nitrogen. DIPEA (3.35mL, 20mmol) was added under ice-cooling to react for 0.5 hour. Adding H-Lys (OCH) into the system 3 ) OH.2 HCl (583mg, 2.5 mmol) and then returned to room temperature for 20 h. The reaction mixture was added to 250mL of ethyl acetate, washed with a saturated sodium bicarbonate solution (20 mL. Times.3), 1M dilute hydrochloric acid (20 mL. Times.3) and a saturated sodium chloride solution (20 mL. Times.3) in this order, the organic phase was collected, dried over anhydrous magnesium sulfate and concentratedCrystallization at 4 ℃ gave the product BocNH-GFLGKGLFG-NHBoc (1.62g, 1.46mmol, 58.4% yield) as a white solid. 1 H NMR(400MHz,δin ppm), 13 C NMR(100MHz,δin ppm),LC-MS(ES+):m/z=1109.4[M+H] + ,MALDI-HRMS:m/z=1131.6045[M+Na] +
BocNH-GFLGKGLFG-NHBoc (1.33g, 1.2 mmol) was placed in a nitrogen protected round bottom flask, 25mL of DCM/TFA (v: v = 1:1) mixed solution was added under ice bath, after 12 hours reaction at room temperature the solvent was spun off, and after adding twice anhydrous ether, it was dried by suction to give a white solid intermediate which was dissolved in 100mL H 2 In a mixed solution of O/ACN (v: v = 4:1), 1M NaOH solution is added dropwise under ice bath to adjust pH to 7-8, then methacryloyl chloride (0.3 mL,3 mmol) is dissolved in Acetonitrile (ACN) and added dropwise to the system, meanwhile, 1M NaOH solution is slowly added dropwise to maintain the system at pH =10, the system is reacted for 1 hour under ice bath and then placed at room temperature for 4 hours, the reaction solution is added into 200mL ethyl acetate, 1M diluted hydrochloric acid is used to adjust pH to 2-3, aqueous phase is extracted three times by EA, organic phase is washed by sodium chloride solution (20 mL × 3), finally dried by anhydrous magnesium sulfate and concentrated, the obtained concentrated solution is placed at 4 ℃ for crystallization to obtain white solid product MA-GFLGKGLFG-MA (700mg, 0.6842 mmol, yield 56.7%). 1 H NMR(400MHz,δin ppm), 13 C NMR(100MHz,δin ppm),LC-MS(ES+):m/z=516.3[M+2H] 2+ ,1031.4[M+H] + 。MALDI-HRMS:m/z=1031.5506[M+H] + ,1053.5377[M+Na] + ,1029.5364[M-H] -
3. Construction and preparation of degradable branched/crosslinked glycosyl polymer photodynamic therapy system
The synthetic route for the degradable branched/cross-linked glycopolymer photodynamic therapy system is shown in figure 3. The specific synthesis method comprises the following steps:
monomers MA-D-Galactosamine (1446mg, 4.63mmol), MA-PySS (100mg, 0.39mmol), crosslinker MA-GFLGKGLFG-MA (23.5mg, 22.8. Mu. Mol), chain transfer agent MA-GFLGK-CTA (53.0mg, 62.4. Mu. Mol) and initiator VA044 (6.7mg, 20.8. Mu. Mol) were dissolved in H 2 O/CH 3 In a mixed solution of OH (7.2mL, 1, 4,v/v), the polymerization flask was placed on ice away from lightAnd introducing argon into the bath, bubbling the argon for deoxygenation for 45 minutes, sealing the bath, keeping the bath in the dark, placing the bath in an oil bath at 45 ℃ for reaction for 12 hours, quenching the reaction by using liquid nitrogen, dialyzing the reaction liquid at low temperature, and freeze-drying the reaction liquid to obtain a light red solid Branche-GFLG-poly D-galactosamine-PySS (B-gal-PySS, 1480mg, and the yield is 91.2%). The hydrogen spectrum of the polymer precursor Branche-GFLG-poly D-galactosamine-PySS (B-gal-PySS) is shown in FIG. 4.
1300mg of B-gal-PySS was dissolved in 10mL of an aqueous solution, treated with 1000mg of Dithiothreitol (DTT) overnight and dialyzed (MWCO 3.5KDa at 4 ℃ C.) for 2 days, and the sample was treated with an aqueous phase filter and lyophilized to obtain Branche-GFLG-poly D-galactosamine-SH (B-gal-SH, 1150mg, 88.5% yield) as a white solid powder. The hydrogen spectrum of the polymer precursor Branched-GFLG-poly D-galactosamine-SH (B-gal-SH) is shown in FIG. 5.
Branche-GFLG-poly D-galactosamine-S-Ppa (BSP) was prepared as follows: 500mg of B-gal-SH in 5mL of RO H 2 Adding 10mL of DMSO after O to prepare a B-gala-SH/DMSO mixed solution, dissolving 75mg of Maleimide-Ppa in 2mL of DMSO, dropwise adding the dissolved solution into the B-gala-SH/DMSO mixed solution under stirring, reacting at room temperature in a dark place overnight, dialyzing (MWCO 3.5 KDa) for 2 days after the dark place to remove the DMSO, centrifuging (7000 rpm is multiplied by 5 min), taking supernatant, filtering the supernatant by using an aqueous phase filter head (0.45 mu m), and freeze-drying the filtrate to obtain a crude dark green solid (504 mg, yield 87.7%). Dissolving the crude product with 2mL of water, then dropwise adding the solution into 200mL of acetonitrile to generate a large amount of precipitates, centrifuging (10000 rpm multiplied by 5 min), collecting solid residues, repeatedly treating and purifying for three times, dissolving the obtained solid in 20mL of deionized water again, and freeze-drying to obtain a product (BSP, 420 mg). The hydrogen spectrum of the polymer Branched-GFLG-poly D-galactosamine-S-Ppa (BSP) is shown in FIG. 6.
The results of the amino acid analysis of BSP are shown in Table 1.
TABLE 1 amino acid analysis results of sugar-based branched/crosslinked Polymer BSP
Figure BDA0003211650000000081
In general, the renal threshold for effective metabolism of polymeric carriers in vivo is about 50kDa, and thus requiresIt is ensured that the molecular weight of the polymer backbone after degradation is below this level. It was found that the sugar-based polymer material of the invention was rapidly degraded by the action of the enzyme, degradation products after 4 hours (Mn =32.4 × 10) 3 ,Mw=46.7×10 3 PDI = 1.44) has a better homogeneity with a molecular weight below the renal threshold.
Example 2 preparation of BSP-entrapped drug Olaparib
Prepared by dialysis, 300mg BSP was weighed out and dissolved in 5mL deionized water and 10mL DMSO was added. 30mg of Olaparib (Olaparib) was dissolved in 2mL of DMSO and added dropwise to BSP-containing H 2 O/DMSO mixed solution. The reaction was stirred overnight in the dark, dialyzed (3.5 kDa MWCO) for 2 days to remove the DMSO solvent, then treated with an aqueous phase filter and lyophilized to give an Olaparib-coated polymer designated BSPO (O for olaparib).
Example 3 preparation of the cell Membrane-glycosyl Polymer of the invention
Normally cultured 4T1 cells (adherent density 80-90%) were scraped off and collected in Phosphate Buffered Saline (PBS), and cell pellets were collected by centrifugation (700 g × 7 min). It was then resuspended in PBS containing a mixture of protease inhibitors. Cells were completely destroyed by 20 squeezes with an Avanit micro-extruder without polycarbonate porous membrane. The resulting solution was centrifuged at low temperature (1000 g.times.15min, 3000g.times.15min, 10000g.times.30min, 4 ℃ C.) in this order, and the cell membrane-containing supernatant was collected (TEM result of cell membrane is shown in FIG. 7 a). Mixing the supernatant with an aqueous solution containing BSPO, stirring overnight (4 ℃), centrifuging and collecting (20000 g multiplied by 30 min), wherein the obtained precipitate is a target product named as CM-BSPO, and observing the morphology of the product by using a TEM after resuspension, as shown in FIG. 7b, the cell membrane structure at the periphery of the glycosyl polymer can be obviously observed, which indicates the successful wrapping of the cell membrane.
Comparative example 1 preparation of Branched-GFLG-poly D-galactosamine-S-hexyl-Ppa (BShP)
1. Preparation of functional photosensitizer Maleimide-hexyl-Ppa
The synthetic route for the functionalized photosensitizer is shown in FIG. 8.
Maleimide-COOH (1.17g, 6.9mm) was weighed outol) and a condensing agent HATU (3.74g, 9.85mmol) are dissolved in 10mL of DMF, DIEA (3.2mL, 18.3mmol) is added under ice bath to react for 5 minutes, and then N-Boc-1,6-hexanediamine hydrochloride (1.80g, 8.3mmol) is added to the system to react for 2 hours. 50mL of saturated sodium bicarbonate solution and 30mL of DCM were added to the reaction solution, the organic phases were collected and the aqueous phase was extracted three times with DCM, the organic phases were combined and washed twice with saturated sodium bicarbonate solution, dilute hydrochloric acid and saturated sodium chloride solution, and the solvent was removed after drying over anhydrous magnesium sulfate to give the crude product as a yellow solid. Purifying by column Chromatography (CH) 3 DCM =1, 20,v/v) followed by further isolation and purification by high performance liquid chromatography gave the product Maleimide-hexyl-NHBoc as a colorless liquid (1.66 g, 65.4% yield). 1 H NMR(400MHz,δin ppm), 13 C NMR(100MHz,δin ppm),LC-MS(ES+):m/z=368.3m/z[M+H] + ,390.2m/z[M+Na] + 。MALDI-HRMS:m/z=390.2000[M+Na] +
Maleimide-hexyl-NHBoc (220mg, 0.60mmol) was dissolved in 10mL DCM at 0 deg.C, 10mL TFA was added, the reaction was allowed to proceed overnight, the solvent was removed, and the residue was treated with dry ether to give a solid powder. The solid powder obtained in the above step, pyropheophorbide a (270mg, 0.50mmol) and condensing agent HATU (285mg, 0.75mmol) were dissolved in DCM, DIEA (0.23mL, 1.33mmol) was added, the reaction solution was reacted at room temperature for 1.5 hours, the solvent was removed, and column purification was performed (DCM: CH) 3 OH =30, 1-20, v/v) to yield Maleimide-hexyl-Ppa as a black solid powder (237 mg, 50.5% yield). 1 H NMR(400MHz,δin ppm),LC-MS(ES+):m/z=784.4m/z[M+H] + 。MALDI-HRMS:m/z=806.3995[M+H] + ,m/z=822.3678[M+K] +
2. Preparation of Branched-GFLG-poly D-galactosamine-S-hexyl-Ppa (BShP)
Branch-GFLG-poly D-galactosamine-S-hexyl-Ppa (BShP) was prepared as follows: 500mg of B-gal-SH in 5mL of RO H 2 Adding 10mL of DMSO after O to prepare a B-gala-SH/DMSO mixed solution, dissolving 80mg of Maleimide-hexyl-Ppa in 2mL of DMSO, dropwise adding the dissolved solution into the B-gala-SH/DMSO mixed solution under stirring to obtain a dark green solution, reacting overnight in a dark environment at room temperature, dialyzing (MWCO 3.5KDa in a dark environment) for 2 days to remove DMSO, and centrifuging (7000 rpm)X 5 min), the supernatant was filtered through a water phase filter (0.45 μm), and the filtrate was lyophilized to give crude dark green solid (520 mg, 89.7% yield). The crude product was dissolved in 2mL of water and then added dropwise to 200mL of acetonitrile to precipitate a large amount, after centrifugation (9000 rpm. Times.5 min), the solid residue was collected and purified repeatedly three times, and the obtained solid was dissolved in 20mL of deionized water again and lyophilized to obtain the product (BShP, 450 mg). The hydrogen spectrum of the polymer Branched-GFLG-poly D-galactosamine-S-hexyl-Ppa (BShP) is shown in FIG. 9.
Comparative example 2 preparation of Linear glycosyl Polymer photodynamic therapy System
The synthetic route for the linear glycosyl polymer photodynamic therapy system is shown in figure 10:
as a control, a linear glycopolymer photodynamic therapy system was designed and synthesized. The monomers MA-D-Galactosamine (722mg, 2.31mmol), MA-PySS (50mg, 0.20mmol), chain transfer agent 4-cyano-4- (thiobenzoyl) pentanoic acid (CTA, 5.3mg, 19.0. Mu. Mol), and initiator VA044 (2.1mg, 6.5. Mu. Mol) were dissolved in H 2 O/CH 3 In the mixed solution of OH (3.5 mL,1, 4, v/v), a polymerization bottle is placed in an ice bath in a dark place, argon is introduced for bubbling to remove oxygen for 45 minutes, then the polymerization bottle is sealed, placed in an oil bath at 45 ℃ in a dark place for reaction for 12 hours, the reaction liquid is quenched by liquid nitrogen, and then the reaction liquid is dialyzed at a low temperature for freeze-drying to obtain a light red solid, namely, linear-poly D-galactosamine-PySS (L-gal-PySS: 725mg, the yield is 93.3%). The hydrogen spectrum of the polymer precursor, linear-poly D-galactosamine-PySS, is shown in FIG. 11.
620mg of L-gal-PySS was dissolved in 10mL of an aqueous solution, treated overnight with 600mg of DTT, dialyzed (MWCO 3.5KDa at 4 ℃ C.) for 2 days, and the sample was treated with an aqueous phase frit and lyophilized to give Linear-poly D-galactosamine-SH (L-gal-SH, 560mg, 93.3% yield) as a white solid powder. The hydrogen spectrum of the polymer precursor Linear-poly D-galactochitosan-SH is shown in FIG. 12.
The preparation of Linear-poly D-galactosamine-S-Ppa (LSP) is similar to the preparation method of degradable branched/crosslinked glycosyl polymer photodynamic therapy system: 500mgL-gala-SH in 5mL RO H 2 Adding 10mL of DMSO after O to prepare L-gala-SH/DMSO mixed solution, dissolving 80mg of Maleimide-Ppa in 2mL of DMSO, and dropwise adding the mixture into the L-gala-SH/DMSO mixed solution under stirring to obtain inkThe green solution was reacted overnight under ambient light-shielded conditions, dialyzed (MWCO 3.5KDa under light-shielded conditions) for 2 days to remove DMSO, centrifuged (7000 rpm × 5 min), and the supernatant was filtered through a water phase filter (0.45 μm), and the filtrate was lyophilized to obtain crude dark green solid (512 mg, yield 88.3%). Dissolving the crude product with 2mL of water, then dropwise adding the solution into 200mL of acetonitrile to generate a large amount of precipitates, centrifuging (10000 rpm multiplied by 5 min), collecting solid residues, repeatedly treating and purifying for three times, dissolving the obtained solid in 20mL of deionized water again, and freeze-drying to obtain a product (LSP, 496 mg). The hydrogen spectrum of the polymer Linear-poly D-galactosamine-S-Ppa (LSP) is shown in FIG. 13.
COMPARATIVE EXAMPLE 3 preparation of HPMA-based degradable branched/crosslinked Polymer photodynamic therapy System
As a control for sugar-based materials, HPMA-based degradable branched/cross-linked polymer photodynamic therapy systems were designed and synthesized. The synthetic route for the HPMA-based degradable branched/crosslinked polymer photodynamic therapy system is shown in fig. 14.
The specific synthesis method comprises the following steps:
monomer HPMA (722mg, 5.04mmol), MA-PySS (50mg, 0.20mmol), crosslinking agent MA-GFLGKGLFG-MA (11.8mg, 11.4. Mu. Mol), chain transfer agent MA-GFLGK-CTA (26.5mg, 31.2. Mu. Mol) and initiator VA044 (3.4mg, 10.4. Mu. Mol) were dissolved in H 2 O/CH 3 In a mixed solution of OH (3.6 mL, 1). The hydrogen spectrum of the polymer precursor Branched-GFLG-poly HPMA-PySS is shown in FIG. 15.
640mg of B-HPMA-PySS was dissolved in 10mL of an aqueous solution, treated with 1000mg of DTT overnight and dialyzed (4 ℃ C., MWCO 3.5 KDa) for 2 days, and the sample was treated with an aqueous phase cartridge and lyophilized to obtain Branched-GFLG-poly HPMA-SH (B-HPMA-SH, 600mg, 93.8% yield) as a white solid powder. The hydrogen spectrum of the polymer precursor Branched-GFLG-poly HPMA-SH is shown in FIG. 16.
Branche-GFLG-poly-HPMA-S-Ppa (BHSP) was prepared as follows: 500mg of B-HPMA-SH dissolved in 5mL of RO H 2 Adding 10mL of DMSO after O to prepare a B-HPMA-SH/DMSO mixed solution, dissolving 80mg of Maleimide-hexyl-Ppa in 2mL of DMSO, dropwise adding a dark green solution obtained in the B-HPMA-SH/DMSO mixed solution under stirring, reacting overnight in a dark environment at room temperature, dialyzing (MWCO 3.5 KDa) for 2 days to remove DMSO, centrifuging (7000 rpm is multiplied by 5 min), taking a supernatant, filtering the supernatant by using an aqueous phase filter head (0.45 mu m), and freeze-drying the filtrate to obtain a dark green solid crude product (500 mg, yield 86.2%). The crude product was dissolved in 2mL of water and then added dropwise to 200mL of acetone to cause a large amount of precipitation, after centrifugation (9000 rpm. Times.5 min), the solid residue was collected and purified by repeating the treatment three times, and the obtained solid was dissolved in 20mL of deionized water again and then lyophilized to obtain a product (BHSP, 460 mg). The hydrogen spectrum of the polymer Branched-GFLG-poly HPMA-S-Ppa (BHSP) is shown in FIG. 17.
The following specific test examples prove the beneficial effects of the invention:
test example 1 characterization of sugar-based Polymer
1. Particle size and potential measurement of sugar-based Polymer
BSP, BShP and LSP are weighed, dissolved and diluted by pure water to a final concentration of 1.0mg/mL, a particle size analyzer is used for representing the particle size and the Zata potential of a sample (the measurement is repeated three times each time), and finally, graphPad Prism software is used for analyzing final data. Meanwhile, a sample with the concentration of 200 mug/mL is dripped on a copper net for natural air drying, and the size and the shape of the particle size are observed by using a transmission electron microscope.
The morphology of each group of materials observed by transmission electron microscopy is shown in fig. 18. The assembly of BShP is a very uniform round nanoparticle; compared with the BSP, the formed nanoparticles are not regular, but the similar round shape can be observed; while LSP without branched/cross-linked structure is in loose filamentous distribution without aggregation to form special morphology.
As shown in fig. 19: the particle sizes of BSP, BShP and LSP were measured by DLS to be about 238nm, 164nm and 374nm, respectively, i.e. BShP formed the most dense particles, while BSP was relatively loose but retained the nanoparticle morphology. The Zeta potentials of the three groups of materials are respectively-16.81 mV, -19.23mV and-21.97 mV, which are all electronegativity.
2. In vitro ROS production assay for carbohydrate-based polymers
BSP, BShP and LSP were dissolved and diluted to a photosensitizer Pyropheophorbide a (Ppa) concentration of 5.0. Mu.g/mL, while SOSG detection reagent was added to a final concentration of 2.5. Mu.M. Each set of the solutions was pipetted at 100. Mu.L into a 96-well plate and provided with 3 multiple wells, followed by irradiation with 660nm laser (power density: 10 mW/cm) 2 The duration is as follows: 15 min). Fluorescence intensity (excitation wavelength 490nm, emission wavelength 525 nm) was measured with a multifunctional microplate reader at time points 0.5, 1, 1.5, 2, 3, 5, 10, 15min and the fluorescence intensity of each sample at the wavelength 525nm was recorded. PBS was used as blank control.
Ppa is capable of absorbing photons of a particular wavelength to transition from a ground state to an excited state, and subsequently transferring the absorbed energy to nearby oxygen molecules to produce singlet oxygen. 5363 ROS produced by excitation of Ppa by light can be detected by the kit. The Singlet oxygen fluorescent probe (SOSG) is a detection reagent with high selectivity on Singlet oxygen, the indicator initially has weak blue fluorescence, and can emit green fluorescence similar to fluorescein after the action of Singlet oxygen (the maximum excitation/emission wavelength is about 504/525 nm), and the intensity of the green fluorescence is in positive correlation with the amount of generated ROS. As shown in FIG. 20, BSP produced the strongest fluorescence after absorption of 660nm excitation energy, the second LSP, the weakest BShP, and the BSP fluorescence intensity was significantly higher than that of LPS and BShP. Indicating that BSP produces higher amounts and efficiency of ROS production in vitro, while BShP and LSP produce relatively lower efficiencies of ROS production. Further shows that BSP has better killing effect on tumors.
3. In vitro IC50 value determination of carbohydrate polymers
4T1 cells were plated at 5X 10 4 The cell concentration per well was seeded in 96-well plates, the medium was aspirated off after the cells were attached (about 12 hours of incubation), the incubation was continued for 6 hours after addition of a gradient (Ppa concentration: 50, 20, 10, 5, 2, 1, 0.5, 0.1, 0.01. Mu.g/mL) solution containing BSP, LSP and BShP formulated with the medium, and 2J/cm/g was given after completion of the incubation 2 The dose is illuminated, after incubation for 12 hours, CCK8 reagent is used according to the instruction of the kit, and the absorbance of about 450nm is detected by a microplate reader. The results of the assay were plotted using GraphPad Prism (vison 8.0) software.
IC was performed by in vitro cell experiments 50 Comparison of values (fig. 21). Under the same Ppa concentration gradient, illumination dose and culture condition, the IC50 values of the three groups of polymers are respectively BSP 0.25 mu g/mL, LSP 0.91 mu g/mL and BShP 4.8 mu g/mL, which indicates that the BSP tumor cells have the strongest killing effect.
4. Glycosyl polymer in vivo tumor site signal detection
1×10 6 4T1 cells were inoculated into the flank of Balb/c mice (20 g, 6-8 weeks old) and the experiment was started when the tumor grew to about 5mm in diameter. Mice were randomly divided into 3 groups (5 per group) and injected via tail vein with Ppa, respectively, of a 5mg/kg concentration of different groups of material (BSP, BShP and LSP). The tumor site fluorescence signals were observed by a live fluorescence imaging system at different time points (5 min,30 min, 1h, 3h, 6h, 12h and 24 h) at the completion of the injection, and the data were processed and analyzed by a data analysis system.
The results show that the fluorescence intensity of the BSP material at the tumor site gradually increased after injection, and reached the highest in about 6 hours, and the fluorescence intensity at the tumor site at each time point was much higher than that of the LSP and BShP groups (FIG. 22).
At the same concentration of Ppa, BSP, both in vitro ROS production, toxic effects on tumor cells and in tumor tissue accumulation, are higher than LSP and BShP, which ensures the efficacy of photodynamic therapy. BSP can be used as photosensitizer, and has excellent effect for photodynamic therapy of tumor.
Test example 2 sugar-based Polymer-Encapsulated Olaparib and characterization thereof
1. Polymer-encapsulated olaparib
The polymer-coated olaparib is prepared by a dialysis method, 300mg of LSP, BSP, BShP and BHSP are weighed and respectively dissolved in 5mL of deionized water, 10mL of DMSO is added, and H containing the material is obtained 2 O/DMSO mixed solution. 30mg of Olaparib (Olaparib) was dissolved in 2mL of DMSO and added dropwise to H containing material 2 O/DMSO mixed solution. Stirring overnight in dark, dialyzing the reaction solution (3.5 kDa MWCO) for 2 days to remove DMSO solvent, treating with water phase filter membrane, and lyophilizing to obtain Olaparib-coated polymer, which is sequentially named as LSPO, BSPO, BShPO and BHSPO (O stands for olaparib).
2. Determination of Olaparib entrapment
The loading of LSP, BSP, BShP and BHSP for olaparib was calculated by HPLC results. Drug loading (wt%) = (mass of drug entrapped/total mass of polymer and entrapped drug) × 100%. Dissolving each group of materials by using a mobile phase, performing ultrasonic treatment, and performing HPLC detection on the filtrate after the organic phase filter head is treated. Column model number (Column name: shim-pack GLST, C18,5 μm. Size: 250X 4.6mm I.D.), test Column temperature 30 ℃, flow rate 1.0mL/min, sample size 20 μ L, sample concentration about 100 μ g/mL, detector selected as UV light (276 nm), mobile phase CH 3 OH/H 2 Mixed solution of O (v: v = 1:1).
The results are shown in Table 2. The highest packing capacity of the BSP polymer reaches 4.03%; the BShP loading was 2.17% worse than BSP. In contrast, the control materials LSP and BHSP were not able to effectively encapsulate small molecule drugs, and the encapsulation amount was only 0.33% and 0.13%, respectively. Indicating that the BSP is in a proper assembled stacked form to be capable of loading more small molecule drugs. The entrapped small molecule drug was collected by HPLC for detection and HRMS results indicated that olaparib was still the active drug (fig. 23).
TABLE 2 Ppa content of each group of polymers and the corresponding amount of Olaparib entrapped
Figure BDA0003211650000000111
Test example 3 Effect of cell Membrane-glycosyl Polymer
Statistical Analysis of multiple comparisons was performed in this study using Analysis of variance (ANOVA for short), indicating a very significant difference in p <0.01, a significant difference in p <0.05, and no significant difference in ns.
1. In vivo fluorescence detection of tumor site accumulation
1. Test method
Will be 1 × 10 6 4T1 cells were inoculated on the backs of Balb/c mice (20 g, 6-8 weeks old) until tumors grewVolume of about 50mm 3 Experiments were started. Tumor model mice were randomly divided into 4 groups of 5 mice each, and Ppa was injected via tail vein into different groups of 5mg/kg material (free Ppa, BSP prepared in example 1, BSPO prepared in example 2, and CM-BSPO prepared in example 3). After completion of the injection, tumor site fluorescence signals were observed by an in vivo fluorescence imaging system (IVIS, perkinElmer, usa) at various time points (1 hour, 4 hours, 8 hours, 1 day, 2 days, 4 days, and 7 days), and the data were processed and analyzed by a data analysis system.
2. Test results
As shown in fig. 24, the highest fluorescence signal intensity was observed about 4 hours after injection of free Ppa into mice, and then the fluorescence signal intensity at the tumor site gradually decreased within a time point, and the fluorescence signal at the tumor site was not substantially observed about 2 days later. The free Ppa molecular weight of the small molecular photosensitizer is about 548, and the small molecular photosensitizer can be rapidly discharged out of the body through renal metabolism in vivo; at the same time, due to the absence of nanostructures and size, efficient aggregation at the tumor site by passive targeting is not possible, so a rapid decrease and disappearance of the fluorescence signal is observed. After the BSP and BSPO groups are injected into mice, the fluorescence signals of tumor tissues gradually increase within the first 8 hours and then slowly decrease, but higher fluorescence signals can still be detected on the 7 th day, which indicates that the BSP and BSPO can be accumulated and effectively retained at the tumor sites of the mice. Compared with BSP and BSPO, the fluorescence signal of CM-BSPO with the surface wrapped by tumor cell membranes at the tumor site of the mouse is obviously higher in the first hours, and the duration of the high fluorescence intensity signal is longer. The semiquantitative fluorescence signal and the signal change curve chart (as shown in figure 25) show that the signal change curve of the free Ppa is steeper, and rapidly decreases after 4 hours until disappears, while the signals of the BSP and BSPO groups do not start to slowly decrease until about 1 day, and the fluorescence signal can still be detected after 7 days. The CM-BSPO group showed higher signal intensity at each time point than BSP and BSPO, indicating that it was more significant at the tumor site. Compared with free Ppa, the in vivo metabolism time of the polymer drug delivery system is obviously prolonged, so that the aggregation intensity of the photosensitizer and the olaparib at a tumor part is increased, the window period of photodynamic therapy is prolonged, and the action time of the combined application of the tumor photodynamic therapy and the olaparib is effectively prolonged.
2. In vivo anti-tumor therapy in mouse tumor model
1. Test method
A tumor treatment flowchart for the mouse tumor model is shown in fig. 26. Will be 1 × 10 6 4T1 cells were inoculated onto the backs of Balb/c mice (20 g, 6-8 weeks old) until tumors grew to a volume of about 50mm 3 The experiment was started on the left and right (about 7 days). Tumor model mice were randomly divided into 10 groups of 5 mice each:
the illumination group (+ L) was divided into 5 groups: CM-BSPO + L, BSPO + L, BSP + L, ppa + L and Saline + L;
the non-illuminated group (-L) has 5 groups, and is divided into: CM-BSPO-L, BSPO-L, BSP-L, ppa-L and Saline-L.
BSP, BSPO, CM-BSPO and physiological saline with the concentration of 5mg/kg are injected into Ppa through tail vein respectively. 12 hours after the injection, the illumination group was irradiated with 660nm laser at an irradiation dose of 2J/cm 2 Photodynamic therapy is carried out every 2 days for 3 times, and the tumor volume is measured and monitored in the treatment process.
(1) Detecting tumor treatment effect by MRI, CT and other imaging means
The size of the tumor is measured by Magnetic Resonance Imaging (MRI) while photodynamic therapy is performed. MR images of the tumor site were acquired by a 7T MRI scanner. The parameters of the MRI sequence are as follows: TR =100ms, te =2.5ms, fov:40mm × 40mm, matrix: 256 × 256, scan time =128s. After 20 days of concurrent treatment, breast scans of the mice were performed by Micro-CT apparatus to observe the metastasis of the tumor in both lungs of the mice.
(2) HE staining and immunohistochemical staining of tumor tissue sections
After the treatment (day 19 of the experiment), lung and tumor tissues were collected after euthanasia of mice, fixed in paraformaldehyde (4% in PBS pH 7.4) for 24 hours, embedded in paraffin and sectioned. The lungs were imaged after HE staining to obtain images. Tumor tissues were treated and subjected to immunohistochemical staining for CD31 and Ki67 and images were taken by photography.
2. Test results
(1) Tumor volume
The tumor volume changes in each mouse are shown in fig. 27 for the different treatment groups, and the illumination group (+ L) is divided into: CM-BSPO + L, BSPO + L, BSP + L, ppa + L and Saline + L; also, the non-illuminated group (-L) was divided into: CM-BSPO-L, BSPO-L, BSP-L, ppa-L and Saline-L. The tumor volumes of mice in different treatment groups were counted, as shown in fig. 28, the tumor volumes of the saline group without light treatment and only light treatment and the Ppa group of mice were not significantly different (ns), and were all much larger than the CM-BSPO, BSPO and BSP groups with light treatment, and the tumor sizes increased with time during observation. The groups without light exposure, in which CM-BSPO, BSPO and BSP had a large accumulation of Ppa at the tumor site, did not produce ROS killing of tumor cells without light exposure, and thus had no significant antitumor effect. In each group of the light treatment, the normal saline group does not have Ppa as a photosensitizer, and the normal saline group also cannot generate ROS to kill tumor cells, and has no obvious anti-tumor effect. The residence time of the free Ppa at the tumor site is only a few hours, and illumination is performed 24 hours after administration, the amount of free Ppa remaining at the tumor site is insufficient to generate a large amount of ROS to kill tumor cells. Compared with free Ppa, the polymer Ppa has relatively longer aggregation time in a tumor body, provides a longer treatment window period, and obviously improves the treatment effect of the tumor.
The experimental results show that BSP + L, BSPO + L and CM-BSPO + L group Ppa are highly concentrated at the tumor site, and a large amount of ROS can be generated by illumination, so that the tumor cells are killed. Volume monitoring revealed significantly smaller tumor volumes in mice, and no significant increase in tumor volume during treatment. In the BSPO + L group loaded with olaparib, olaparib can enhance DNA damage of tumor cells, thereby increasing the effect of killing tumor cells, so that the tumor volume is significantly reduced compared to the BSP group, with statistical difference (p <0.01, n = 5). The CM-BSPO + L with the surface wrapped by the cell membrane has the advantages that the accumulation degree of the tumor part is increased, the cell uptake is increased, the treatment effect is obviously improved, the tumor volume is not obviously increased in the whole treatment period, and the tumor volume after treatment is obviously statistically different compared with the BSPO + L group.
(2) Tumor inhibition rate
The tumor inhibition rate of tumor treatment can be calculated by comparing the other groups with the group without adding light by normal saline, as shown in fig. 29, the tumor inhibition rate is sequentially as follows: CM-BSPO + L87.66%, BSPO + L81.34%, BSP + L69.55%, ppa + L36.99%, saline + L26.18%, and the tumor inhibition rate of the rest non-illuminated groups is only about 12%. The therapeutic effect of CM-BSPO plus light exposure is thus seen to be most pronounced.
(3) Magnetic resonance imaging
Tumor scans were performed every other week during treatment by magnetic resonance imaging, i.e. on days 1, 7, 14 and 19 of treatment, and the results are shown in fig. 30 and 31. The magnetic resonance image reflects the same therapeutic effect. The tumor volumes of the BSP + L, BSPO + L and CM-BSPO + L groups were significantly reduced. On day 19 of treatment, mice in each treatment group were subjected to pulmonary CT scan, and mice were observed for pulmonary metastasis while lung tissue was taken out for tissue imaging and HE staining.
The CT scan results in FIGS. 30 and 31 show that no distinct metastases were observed in the lungs of the CM-BSPO + L and BSPO + L group of mice, and no abnormal tissue was observed by visual observation of the specimen tissue. In other groups, one or more high-density isolated nodules can be observed in the CT lung image, and nodular protrusions can be observed in general specimen observation; and HE staining (fig. 32) observed deeply enlarged nuclei, with heterogeneity of cell morphology with surrounding normal lung tissue.
(4) Immunohistochemical staining
The tumor tissue sections were subjected to CD31 immunohistochemical staining to observe the vascularity of the tumor sites, as shown in FIG. 33, the expression levels of CD31 in CM-BSPO + L, BSPO + L and BSP + L tumors with obvious tumor treatment effects were lower than those in other groups, indicating that the tumor treatment effects were better and the number of new vessels in the tumors was less.
Ki-67 immunohistochemical staining was also performed to observe cell proliferation in the tumors, as shown in FIG. 34. The semi-quantitative statistical analysis result is shown in FIG. 35, the expression level of Ki-67 in tumors between the CM-BSPO + L, BSPO + L group and the BSP + L group is not significantly different, and is lower than that of Ppa + L group and that of Saline + L group, and the statistical difference is achieved. The results show that the intracellular proliferation of the tumors of CM-BSPO + L, BSPO + L and BSP + L is inhibited, and the treatment effect is better than that of other control groups.
The test results show that: the glycosyl polymer prepared by the invention, especially the cell membrane-glycosyl polymer has good photodynamic effect, can be used as a photosensitizer for photodynamic therapy, and has excellent killing effect on tumors. Meanwhile, the glycosyl polymer has good effect as a drug carrier and high drug loading, and the treatment effect is further enhanced after drug loading.
In conclusion, the invention provides a cell membrane-glycosyl polymer, which overcomes the defects of insufficient accumulation at the tumor part and weak killing effect on the tumor of the existing PDT photosensitizer, can be accumulated at the tumor part, and has good targeting effect; it has photodynamic effect, high ROS generating efficiency when used as photosensitizer for photodynamic therapy, and strong killing effect on tumor cells. Meanwhile, the cell membrane-glycosyl polymer can also be used as a drug delivery system to entrap drugs, particularly tumor drugs such as olaparib, and is used for treating tumors with excellent effect. The cell membrane-glycosyl polymer has excellent application prospect in preparing medicaments for treating tumors.

Claims (19)

1. A cell membrane-sugar based polymer characterized by: the glycosyl polymer with the structure shown in formula I is coated with a layer of cell membrane:
Figure QLYQS_1
wherein the content of the first and second substances,
(x + y + z) o: p: n: m in a molar ratio of 2.9;
r is selected from
Figure QLYQS_2
2. The cell membrane-glycosyl polymer according to claim 1, wherein: the cell membrane is the cell membrane of a tumor cell;
and/or the glycosyl polymer is prepared from the following raw materials in parts by weight: 100 to 500 portions of B-gala-SH and 1 to 100 portions of Maleimide-Ppa;
the structural formula of the Maleimide-Ppa is as follows:
Figure QLYQS_3
the B-gala-SH is prepared from the following raw materials in parts by weight: 1000-1500 parts of B-gal-PySS and 1000-1500 parts of dithiothreitol;
the B-gala-PySS is prepared from the following raw materials in parts by weight: 1000-1500 parts of MA-D-Galactosamine, 100-200 parts of MA-PySS, 10-50 parts of MA-GFLGKGLFG-MA and 50-100 parts of MA-GFLGK-CTA;
the MA-D-Galactosamine has the structure
Figure QLYQS_4
The structure of the MA-PySS is
Figure QLYQS_5
The MA-GFLGKGLFG-MA has the structure
Figure QLYQS_6
The structure of the MA-GFLGK-CTA is
Figure QLYQS_7
3. The cell membrane-glycosyl polymer according to claim 2, wherein: the glycosyl polymer is prepared from the following raw materials in parts by weight: B-gala-SH 500 parts, maleimide-Ppa parts;
and/or the B-gala-SH is prepared from the following raw materials in parts by weight: 1300 parts of B-gal-PySS and 1000 parts of dithiothreitol;
and/or the B-gala-PySS is prepared from the following raw materials in parts by weight: 1446 parts of MA-D-Galactosamine, 100 parts of MA-PySS, 23.5 parts of MA-GFLGKGLFG-MA and 53 parts of MA-GFLGK-CTA.
4. The cell membrane-sugar based polymer according to claim 2, wherein: the preparation method of the cell membrane comprises the following steps:
squeezing the cells with a miniature extruder until the cells are destroyed, centrifuging, and collecting the supernatant containing cell membranes;
and/or, the preparation method of the B-gal-SH comprises the following steps:
reacting B-gala-PySS with dithiothreitol to obtain the product.
5. The cell membrane-glycosyl polymer according to claim 2 or 3, wherein: the preparation method of the B-gala-PySS comprises the following steps:
dissolving MA-D-Galactosamine, MA-PySS, MA-GFLGKGLFG-MA and MA-GFLGK-CTA in solvent, reacting under the action of initiator, and lyophilizing.
6. The cell membrane-glycosyl polymer according to claim 5, wherein:
the solvent is a mixed solution of water and methanol;
and/or, deoxygenating prior to said reaction;
and/or the reaction is a reaction protected from light.
7. The cell membrane-glycosyl polymer according to claim 6, wherein:
the volume ratio of the water to the methanol is 1:4;
and/or the initiator is VA044;
and/or the reaction is carried out in an oil bath at the temperature of 40-50 ℃ for 10-12 hours.
8. The cell membrane-glycosyl polymer according to claim 2 or 3, wherein: the preparation method of the glycosyl polymer comprises the following steps:
(1) Dissolving B-gala-SH in water, adding DMSO, and preparing a B-gala-SH/DMSO mixed solution;
(2) Dissolving Maleimide-Ppa in DMSO, and adding the dissolved solution into a B-gala-SH/DMSO mixed solution for reaction;
(3) And (3) purifying the reaction liquid obtained in the step (2).
9. The cell membrane-glycosyl polymer according to claim 8, wherein:
in the step (1), the volume ratio of the water to the DMSO is 1: (1-5);
and/or, in the step (2), the reaction is carried out at room temperature in a dark place.
10. A method for preparing the cell membrane-sugar based polymer according to any one of claims 1 to 9, characterized in that: it comprises the following steps:
mixing the cell membrane with glycosyl polymer water solution, and centrifuging to obtain the final product.
11. The method of claim 10, wherein:
after mixing, stirring overnight at the temperature of 0-4 ℃;
and/or, the centrifugation is 20000g for 30min.
12. Use of the cell membrane-sugar based polymer according to any one of claims 1 to 9 in the manufacture of a medicament for photosensitizing agents.
13. Use according to claim 12, characterized in that: the photosensitizer is a medicine for photodynamic tumor treatment.
14. Use of the cell membrane-glycosyl polymer of any one of claims 1-9 in the preparation of a pharmaceutical carrier.
15. Use according to claim 14, characterized in that: the drug used for being encapsulated by the drug carrier is an anti-tumor drug.
16. Use according to claim 15, characterized in that: the anti-tumor drug is olaparib.
17. A medicament, characterized by: the preparation is prepared by taking the cell membrane-glycosyl polymer as an active ingredient according to any one of claims 1 to 9, or taking the cell membrane-glycosyl polymer as a carrier to entrap a medicine as the active ingredient according to any one of claims 1 to 9 and adding pharmaceutically acceptable auxiliary materials.
18. The medicament of claim 17, wherein: the entrapped medicine is an anti-tumor medicine.
19. The medicament of claim 18, wherein: the entrapped drug is olaparib.
CN202110932558.3A 2021-08-13 2021-08-13 Polymer carrier for photodynamic therapy and preparation method and application thereof Active CN113786492B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110932558.3A CN113786492B (en) 2021-08-13 2021-08-13 Polymer carrier for photodynamic therapy and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110932558.3A CN113786492B (en) 2021-08-13 2021-08-13 Polymer carrier for photodynamic therapy and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN113786492A CN113786492A (en) 2021-12-14
CN113786492B true CN113786492B (en) 2023-03-28

Family

ID=79181846

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110932558.3A Active CN113786492B (en) 2021-08-13 2021-08-13 Polymer carrier for photodynamic therapy and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN113786492B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115160514B (en) * 2022-04-13 2023-12-08 四川大学华西医院 Photodynamic-micromolecular targeting-gene therapy combined drug system and preparation method thereof

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104758949A (en) * 2014-01-08 2015-07-08 张雅洁 Tumor-targeted adenovirus polymer drug delivery system and preparation method thereof
CN104888219A (en) * 2015-06-10 2015-09-09 东南大学 Tumor phototherapy reagent based on cell membrane coating, as well as preparation method and application of tumor phototherapy reagent
CN105288622A (en) * 2015-11-03 2016-02-03 浙江大学 Preparation method of cell membrane microcapsule simultaneously loaded with chemotherapeutic drug and photodynamic therapeutic drug
CN105412924A (en) * 2014-08-22 2016-03-23 苏州大学 Sugar-containing photodynamic therapy nano particles with blood stability and target ability and preparing method of sugar-containing photodynamic therapy nano particles
WO2016079330A1 (en) * 2014-11-21 2016-05-26 Technical University Of Denmark Gel formulations for local drug release
CN106581690A (en) * 2016-12-23 2017-04-26 四川大学 Tumor microenvironment stimulation degradable amphiphilic block HPMA (hydroxypropyl methacrylate) polymer delivery system and preparation method thereof
CN106880848A (en) * 2017-02-14 2017-06-23 四川大学 Biodegradable poly HPMA Gd magnetic resonance imaging probes and preparation method thereof
CN110279856A (en) * 2019-06-18 2019-09-27 四川大学华西医院 A kind of PEG-Peptide light power-chemotherapy combination drug delivery system and its preparation method and application
CN110591075A (en) * 2019-06-28 2019-12-20 四川大学华西医院 PEG-Peptide linear-tree-shaped drug delivery system and preparation method and application thereof
CN112755182A (en) * 2019-11-01 2021-05-07 深圳先进技术研究院 Preparation and application of nano material for specifically activating immune system
CN112972420A (en) * 2021-02-24 2021-06-18 中国药科大学 Bionic cell membrane nanoparticle and preparation method and application thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL160126A0 (en) * 2001-07-31 2004-06-20 Immunomedics Inc A kit for targeting a target site containing a polymer conjugate
US8703167B2 (en) * 2006-06-05 2014-04-22 Advanced Cardiovascular Systems, Inc. Coatings for implantable medical devices for controlled release of a hydrophilic drug and a hydrophobic drug

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104758949A (en) * 2014-01-08 2015-07-08 张雅洁 Tumor-targeted adenovirus polymer drug delivery system and preparation method thereof
CN105412924A (en) * 2014-08-22 2016-03-23 苏州大学 Sugar-containing photodynamic therapy nano particles with blood stability and target ability and preparing method of sugar-containing photodynamic therapy nano particles
WO2016079330A1 (en) * 2014-11-21 2016-05-26 Technical University Of Denmark Gel formulations for local drug release
CN104888219A (en) * 2015-06-10 2015-09-09 东南大学 Tumor phototherapy reagent based on cell membrane coating, as well as preparation method and application of tumor phototherapy reagent
CN105288622A (en) * 2015-11-03 2016-02-03 浙江大学 Preparation method of cell membrane microcapsule simultaneously loaded with chemotherapeutic drug and photodynamic therapeutic drug
CN106581690A (en) * 2016-12-23 2017-04-26 四川大学 Tumor microenvironment stimulation degradable amphiphilic block HPMA (hydroxypropyl methacrylate) polymer delivery system and preparation method thereof
CN106880848A (en) * 2017-02-14 2017-06-23 四川大学 Biodegradable poly HPMA Gd magnetic resonance imaging probes and preparation method thereof
CN110279856A (en) * 2019-06-18 2019-09-27 四川大学华西医院 A kind of PEG-Peptide light power-chemotherapy combination drug delivery system and its preparation method and application
CN110591075A (en) * 2019-06-28 2019-12-20 四川大学华西医院 PEG-Peptide linear-tree-shaped drug delivery system and preparation method and application thereof
CN112755182A (en) * 2019-11-01 2021-05-07 深圳先进技术研究院 Preparation and application of nano material for specifically activating immune system
CN112972420A (en) * 2021-02-24 2021-06-18 中国药科大学 Bionic cell membrane nanoparticle and preparation method and application thereof

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Wang Wenjia等.Dendron‐Functionalized Polyglutamate‐Pyropheophorbide‐a Conjugates as Nanomedicines for Breast Cancer Photodynamic Therapy.《Macromolecular Rapid Communications》.2021,42(9),第e2100013页. *
Zhenyu Duan等.Synergistic Therapy of a Naturally Inspired Glycopolymer-Based Biomimetic Nanomedicine Harnessing Tumor Genomic Instability.《Advanced Materials》.2021,33第1-13页. *
张沛,等.光动力疗法用糖基光敏剂的研究进展.《有机化学》.2010,30(6),第775-782页. *
焉绍辉,等.红细胞/肿瘤细胞膜仿生载体介导高靶向性光动力抗肿瘤的体外研究.《中国细胞生物学学报》.2022,(8),第1635-1643页. *

Also Published As

Publication number Publication date
CN113786492A (en) 2021-12-14

Similar Documents

Publication Publication Date Title
EP2609935B1 (en) Switching fluorescent nanoparticle probe and fluorescent particle imaging method using same
EP2958946B1 (en) Near-infrared dye-conjugated hyaluronic acid derivative and contrast agent for optical imaging including them
EP1731550A1 (en) Novel water-soluble fullerene, process for producing the same and active oxygen generator containing the fullerene
CN107802840B (en) Peptide-based dendrimer-modified fluorescent carbon dot-based tumor microenvironment response nanoparticle and preparation method thereof
CN112451680B (en) ROS sensitive nano reagent with synergistic induction of photodynamic therapy and iron death and preparation method thereof
CN114377149B (en) Mn-based degradable MOF nano-reactor and preparation method and application thereof
JP6867084B2 (en) New cationic polyphosphazene compounds, polyphosphazene-drug conjugate compounds and methods for producing them
CN111544588B (en) Immunity active peptide-biliverdin conjugate, preparation method and application thereof
CN105001426B (en) A kind of polyaminoacid graft copolymer with tumor-targeting and preparation method thereof
CN113786492B (en) Polymer carrier for photodynamic therapy and preparation method and application thereof
CN113797350B (en) Glycosyl polymer and preparation method and application thereof
CN113663060B (en) Whole-cell tumor nano vaccine, preparation method and application thereof
CN110917349B (en) Bowl-shaped ISP (internet service provider) composite functional nano particle as well as preparation method and application thereof
RU2382787C2 (en) Water-soluble mono-pegylated tetrapyrrole derivatives for photodynamic therapy and method of obtaining them
CN114904012B (en) Active oxygen self-supplementing amphiphilic block copolymer-drug conjugate, preparation method and application thereof
CN110279856B (en) PEG-Peptide photodynamic-chemotherapy combined drug delivery system and preparation method and application thereof
CN111995745B (en) Double-locking polymer and preparation method and application thereof
CN110152013B (en) Pectin-adriamycin conjugate and preparation method and application thereof
CN113135984A (en) In-situ self-assembly polypeptide derivative responding to pathological microenvironment and application thereof
CN116059357B (en) PH responsive charge-reversal nano composite preparation and preparation method and application thereof
CN116041712B (en) Protoporphyrin modified cellulose grafted polylactic acid, tumor targeting stereocomplex drug-loaded nano micelle thereof, and preparation method and application thereof
CN117298294A (en) Antibody modified drug-loaded nano micelle and preparation method and application thereof
CN111298140B (en) Reduction of the T of the response1/T2Switching type MRI contrast agent, preparation method and application thereof
CN1673252A (en) Terminal amino oxyethylene block polymer modified Gd coordination compound and its synthesis process
CN113698555A (en) PH/cathepsin B stepwise response polymer-drug conjugate, micelle, preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant