CN111840305A - Nano particles and preparation method and application thereof - Google Patents

Nano particles and preparation method and application thereof Download PDF

Info

Publication number
CN111840305A
CN111840305A CN202010708342.4A CN202010708342A CN111840305A CN 111840305 A CN111840305 A CN 111840305A CN 202010708342 A CN202010708342 A CN 202010708342A CN 111840305 A CN111840305 A CN 111840305A
Authority
CN
China
Prior art keywords
solution
homotypic
digoxin
tumor cell
nano
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010708342.4A
Other languages
Chinese (zh)
Inventor
孔德新
钟玉绪
赵宝全
张哲�
王营营
马谦
侯岚娇
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.)
Academy of Military Medical Sciences AMMS of PLA
Tianjin Medical University
Original Assignee
Tianjin Medical 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 Tianjin Medical University filed Critical Tianjin Medical University
Priority to CN202010708342.4A priority Critical patent/CN111840305A/en
Publication of CN111840305A publication Critical patent/CN111840305A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • 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
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y40/00Manufacture or treatment of nanostructures
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Nanotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Optics & Photonics (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Botany (AREA)
  • Medical Informatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Condensed Matter Physics & Semiconductors (AREA)
  • General Physics & Mathematics (AREA)
  • Manufacturing & Machinery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

The invention provides a nano particle and a preparation method and application thereof, relating to the technical field of pharmaceutical preparations, wherein the nano particle comprises a nano self-assembly particle and a homotypic tumor cell membrane; the nano self-assembly particles are mainly prepared by self-assembly of digoxin and adriamycin, and the prepared nano self-assembly particles do not need to introduce a carrier material, so that the effect of high drug loading capacity can be realized; simultaneously the surface cladding of this application nanometer self-assembly granule has the homotypic tumor cell membrane, because tumor cell membrane surface has multiple antigen for this application nanoparticle has the characteristics of immune escape and homotypic combination with tumor cell, and then improves its tumor targeting nature, avoids host immune system. Therefore, the nano particles prepared by the application have the advantages of high drug loading and strong tumor targeting property, have stronger targeting property while improving the existing anti-tumor treatment effect, and can effectively reduce the toxicity of the drug to normal tissues.

Description

Nano particles and preparation method and application thereof
Technical Field
The invention relates to the technical field of pharmaceutical preparations, in particular to a nanoparticle and a preparation method and application thereof.
Background
Cancer has become one of the most serious public health problems worldwide. With the continuous development of molecular diagnosis and treatment, the diagnosis and treatment of cancer have been greatly improved in recent 10 years, and the current treatment methods mainly include chemotherapy, radiotherapy, surgery, gene targeting treatment and the like.
Many genotoxic anticancer drugs, such as doxorubicin, exert their anticancer effects by attacking DNA, causing a variety of DNA damage on tumor cells. However, tumor cells can clear DNA damage caused by genotoxic anticancer drugs through the DNA Damage Repair (DDR) pathway. Inhibition of DNA damage repair may therefore enhance the anticancer effect of genotoxic anticancer drugs. Inhibition of DNA damage repair has been reported to sensitize tumor cells to doxorubicin. Digoxin, a major drug in the treatment of heart failure for more than two centuries, has been shown to inhibit DNA double strand break repair. Since cancer chemotherapy is often hampered by rapid DNA Damage Repair (DDR) during and after treatment, DDR targeting has become a powerful complement to conventional chemotherapy. The research proves that digoxin can play the role of resisting tumors by inhibiting the repair of double-stranded and single-stranded breaks of DNA.
However, digoxin is used in a narrow concentration range (0.5-2.0 ng/mL) in clinical application and treatment, and toxicity is caused when the concentration range is beyond the treatment range. On the other hand, as a genotoxic anticancer drug, adriamycin lacks tumor targeting, and meanwhile, adriamycin has cardiotoxicity, so that the clinical application of adriamycin is limited to a certain extent. Although earlier studies showed that the combined use of digoxin and doxorubicin resulted in a synergistic effect in vivo and in vitro in the treatment of cancer, the actual therapeutic effect was not ideal.
Therefore, research and development of a nanoparticle which has the advantages of high drug loading and strong tumor targeting property, so that the problems of narrow digoxin treatment window and lack of tumor targeting property of adriamycin in the prior art are effectively improved, the treatment effect of cancer is improved, and the nanoparticle becomes necessary and urgent, and the nanoparticle is not reported at present.
In view of the above, the present invention is particularly proposed.
Disclosure of Invention
The invention aims to provide a nanoparticle which has the advantages of high drug loading and strong tumor targeting property, has stronger targeting property while improving the existing anti-tumor treatment effect, and can effectively reduce the toxicity of the drug to normal tissues; furthermore, the nano-particle can be widely applied to the preparation process of the medicine for treating cancer.
In order to achieve the above purpose of the present invention, the following technical solutions are adopted:
in a first aspect, the present invention provides a nanoparticle comprising a nano self-assembled particle and a homotypic tumor cell membrane;
the nano self-assembled particles are mainly prepared from digoxin and adriamycin;
the homotypic tumor cell membrane is coated on the surface of the nano self-assembly particles.
In a second aspect, the present invention provides a method for preparing the above nanoparticles, the method comprising the following steps:
(A) preparing nano self-assembled particles by using a precipitation method;
(B) providing homotypic tumor cell membranes, uniformly mixing the homotypic tumor cell membranes with the nano self-assembly particles prepared in the step (A), and then extruding in a grading manner to obtain the nano particles.
In a third aspect, the invention provides an application of the nanoparticles in preparing a medicament for treating cancer.
Preferably, the cancer is non-small cell lung cancer.
Compared with the prior art, the invention has the beneficial effects that:
the nano particle provided by the invention comprises a nano self-assembly particle and a homotypic tumor cell membrane; the nano self-assembled particles are mainly prepared by self-assembling digoxin and adriamycin, and the prepared nano self-assembled particles can realize the effect of high drug-loading rate without introducing a carrier material (equivalent to 100 percent of drug-loading rate); simultaneously the surface cladding of this application nanometer self-assembly granule has homotypic tumor cell membrane, because homotypic tumor cell membrane surface has multiple antigen for this application nanometer particle has the characteristics of immune escape and homotypic combination with tumor cell, and then improves its tumor targeting nature, avoids host immune system. Therefore, the nano particles prepared by the application have the advantages of high drug loading and strong tumor targeting property, have stronger targeting property while improving the existing anti-tumor treatment effect, and can effectively reduce the toxicity of the drug to normal tissues.
The preparation method of the nano-particles provided by the invention comprises the following steps: firstly, preparing nano self-assembly particles by using a precipitation method; and then, uniformly mixing the homotypic tumor cell membrane with the nano self-assembly particles, and then extruding in a grading manner to obtain the nano particles. The preparation method has the advantages of simple preparation process and easy operation.
The nano particles provided by the invention can be widely applied to the preparation process of medicaments for treating cancers, in particular to the preparation process of medicaments for treating non-small cell lung cancer.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below, and it is obvious that the drawings in the following description are some embodiments of the present invention, and other drawings can be obtained by those skilled in the art without creative efforts.
FIG. 1 is a Tyndall effect diagram of the nanoparticles and nano self-assembled particles of the present invention provided in example 4 of the present invention;
FIG. 2 is a transmission electron microscope observation image of the nano-particles and nano self-assembly particles provided in example 4 of the present invention;
FIG. 3 is a graph showing the proliferation inhibitory effect of the nanoparticles and nano self-assembled particles of the present invention on A549 cells, provided in example 5 of the present invention;
FIG. 4 is a graph showing the effect of the nanoparticles and nano self-assembled particles of the present invention on the apoptosis induction rate of A549 cells, which is provided in example 5 of the present invention;
FIG. 5 is a fluorescence microscope observation image of the experiment of zebrafish with nano particles and nano self-assembled particles provided in example 6 of the present invention;
FIG. 6 is a tumor growth observation chart of nude mouse experiment conducted by the nanoparticles and nano self-assembled particles of the present invention provided in example 6 of the present invention;
FIG. 7 is a graph showing the effect of tumor weight of nude mouse experiments with the nanoparticles and nano self-assembled particles of the present invention provided in example 6 of the present invention;
FIG. 8 is a graph showing the effect of tumor volume in nude mouse experiments with the nanoparticles and nano self-assembled particles of the present invention provided in example 6 of the present invention.
Detailed Description
The technical solutions of the present invention will be described clearly and completely with reference to the following embodiments, and it should be understood that the described embodiments are some, but not all, embodiments of the present invention. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
According to one aspect of the present invention, a nanoparticle comprising a nano self-assembled particle and a homotypic tumor cell membrane;
the nano self-assembled particles are mainly prepared from digoxin and adriamycin;
the homotypic tumor cell membrane is coated on the surface of the nano self-assembly particles.
The nano particle provided by the invention comprises a nano self-assembly particle and a homotypic tumor cell membrane; the nano self-assembled particles are mainly prepared by self-assembling digoxin and adriamycin, and the prepared nano self-assembled particles can realize the effect of high drug-loading rate without introducing a carrier material (equivalent to 100 percent of drug-loading rate); simultaneously the surface cladding of this application nanometer self-assembly granule has homotypic tumor cell membrane, because homotypic tumor cell membrane surface has multiple antigen for this application nanometer particle has the characteristics of immune escape and homotypic combination with tumor cell, and then improves its tumor targeting nature, avoids host immune system. Therefore, the nano particles prepared by the application have the advantages of high drug loading and strong tumor targeting property, have stronger targeting property while improving the existing anti-tumor treatment effect, and can effectively reduce the toxicity of the drug to normal tissues.
In a preferred embodiment of the present invention, the hydrated particle size of the nanoparticle is 150 to 200 nm; the hydrated particle size of the nano self-assembly particles is 100-150 nm.
As a preferred embodiment, nanotechnology, as an emerging field of research with rapid development, provides opportunities for breakthrough of specific anti-tumor immunotherapies. The nano drug-loaded system has the characteristics of targeting, sustained and controlled release, skin penetration, physical response, self long circulation and the like, and can overcome the defects of poor stability, short pharmacological action time, low bioavailability, great toxic and side effects and the like of the traditional drugs. Due to the physical size of the nanoparticles, the nanoparticles have the advantage of producing enhanced penetration and retention effects (EPR effect for short) in tumor vessels. The nano-particles prepared from the drug have the effects of reducing the nano-particles from reaching normal tissues and increasing the accumulation of the nano-particles at pathological positions of tumor cells, thereby effectively improving the targeting function of the nano-particles. Meanwhile, the nano particles are accumulated in a tumor part in vivo through active or passive targeting action, so that the drug dosage can be reduced, and the toxic and side effects of the drug on normal tissue cells can be reduced. Therefore, the nano particles with the hydrated particle size of 150-200 nm and the nano self-assembly particles with the hydrated particle size of 100-150 nm have stronger tumor targeting property and can effectively reduce the toxicity of the medicine to normal tissues.
Note: the EPR effect, i.e. the high permeability and retention effect (enhanced permeability and retention effect) of solid tumors, refers to the property that molecules or particles of certain sizes tend to aggregate more in tumor tissue than in normal tissue. The capillary endothelium clearance in normal tissue is compact, the structure is complete, and macromolecule and lipid particle are difficult for permeating the vascular wall, and the blood vessel is abundant in the solid tumor tissue, vascular wall clearance broad, structural integrity is poor, and lymph backflow is lacked, causes macromolecule class material and lipid particle to have selectivity high permeability and detention nature, and this kind of phenomenon is called the high permeability and the effect of detention of solid tumor tissue, for short EPR effect.
In a preferred embodiment of the invention, said homotypic tumor cell membrane comprises a non-small cell lung cancer cell membrane, preferably an a549 tumor cell membrane.
Preferably, the doxorubicin is doxorubicin hydrochloride.
According to an aspect of the present invention, a method for preparing the above nanoparticle comprises the following steps:
(A) preparing nano self-assembled particles by using a precipitation method;
(B) providing homotypic tumor cell membranes, uniformly mixing the homotypic tumor cell membranes with the nano self-assembly particles prepared in the step (A), and then extruding in a grading manner to obtain the nano particles.
The preparation method of the nano-particles provided by the invention comprises the following steps: firstly, preparing nano self-assembly particles by using a precipitation method; and (3) uniformly mixing the homotypic tumor cell membrane with the nano self-assembly particles prepared in the step (A), and then extruding in a grading manner to obtain the nano particles. The preparation method has the advantages of simple preparation process and easy operation.
In a preferred embodiment of the present invention, the step (a) comprises the steps of:
(a) dissolving digoxin in an organic solution to obtain a digoxin solution;
(b) dissolving adriamycin in deionized water to obtain adriamycin solution;
(c) under the condition of stirring, firstly injecting the digoxin solution obtained in the step (a) into deionized water to obtain a solution A; and (c) adding the adriamycin solution obtained in the step (b) into the solution A, and uniformly mixing to obtain the nano self-assembled particles.
In a preferred embodiment of the present invention, the organic solution in the step (a) comprises any one of absolute ethyl alcohol, methanol, isopropanol or n-butanol, preferably absolute ethyl alcohol;
preferably, when the organic solution in the step (a) is absolute ethyl alcohol, the molar concentration of the digoxin solution is 0.1-2 mmol/L, and 1mmol/L is preferred;
Preferably, the molar concentration of the adriamycin solution in the step (b) is 0.1-10 mmol/L, and 1mmol/L is preferable;
in a preferred embodiment of the present invention, the molar concentration of digoxin in the solution A in the step (c) is 0.1 mmol/L;
preferably, the volume ratio of the solution A to the adriamycin solution in the step (c) is 18-20: 1, and is preferably 19: 1;
preferably, the stirring reaction condition of step (c) at least satisfies at least one of the following conditions:
stirring at the rotating speed of 1000-1200 r/min, at the stirring temperature of 48-52 ℃ for 0.8-1.5 h;
more preferably, the stirring reaction condition of step (c) at least satisfies at least one of the following conditions:
stirring at the rotation speed of 1000r/min and the temperature of 50 ℃ for 1 h;
in a preferred embodiment of the present invention, the step (B) comprises the steps of:
(d) providing a homotypic tumor cell membrane containing solution, and then uniformly mixing the homotypic tumor cell membrane with the nano self-assembly particles prepared in the step (c) to obtain a solution B;
(e) extruding the solution B through a filter membrane with the aperture of 200-400 nm for 3-6 cycles in a grading manner by using a liposome extruder to obtain nanoparticles;
preferably, the homotypic tumor cell membrane content in the homotypic tumor cell membrane-containing solution in the step (d) is 0.1 mg/mL;
Preferably, the volume ratio of the homotypic tumor cell membrane-containing solution in the step (d) to the nano self-assembly particles prepared in the step (c) is 1-10: 100-500.
In a preferred embodiment of the present invention, the preparation method comprises the steps of:
(a) dissolving digoxin in absolute ethyl alcohol to obtain a digoxin solution with the molar concentration of 0.1-2 mmol/L;
(b) dissolving adriamycin in deionized water to obtain an adriamycin solution with the molar concentration of 0.1-10 mmol/L;
(c) injecting the digoxin solution obtained in the step (a) into deionized water to obtain a solution A with a digoxin molar concentration of 0.1mmol/L under the stirring conditions that the stirring speed is 1000-1200 r/min and the temperature is 48-52 ℃; adding the adriamycin solution obtained in the step (b) into the solution A, and stirring for 0.8-1.5 hours to obtain nano self-assembled particles; the volume ratio of the solution A to the adriamycin solution is 18-20: 1;
(d) providing a homotypic tumor cell membrane-containing solution with the concentration of 0.1mg/mL, and then uniformly mixing homotypic tumor cell membranes and the nano self-assembly particles prepared in the step (c) in a volume ratio of 1-10: 100-500 to obtain a solution B;
(e) extruding the solution B through a filter membrane with the aperture of 400nm for 3-6 cycles by using a liposome extruder, and then extruding through a filter membrane with the aperture of 200nm for 3-6 cycles to obtain nano particles;
Preferably, the preparation method comprises the following steps:
(a) dissolving digoxin in absolute ethyl alcohol to obtain a digoxin solution with the molar concentration of 1 mmol/L;
(b) dissolving the adriamycin in deionized water to obtain an adriamycin solution with the molar concentration of 1 mmol/L;
(c) injecting the digoxin solution obtained in the step (a) into deionized water to obtain a solution A with the digoxin molar concentration of 0.1mmol/L under the stirring conditions that the stirring speed is 1000r/min and the temperature is 50 ℃; adding the adriamycin solution obtained in the step (b) into the solution A, and stirring for 1h to obtain nano self-assembled particles; the volume ratio of the solution A to the adriamycin solution is 19: 1;
(d) providing a homotypic tumor cell membrane-containing solution with the concentration of 0.1mg/mL, and then uniformly mixing homotypic tumor cell membranes and the nano self-assembly particles prepared in the step (c) in a volume ratio of 1-10: 100-500 to obtain a solution B;
(e) and extruding the solution B through a filter membrane with the aperture of 400nm for 3 cycles by using a liposome extruder, and then extruding through a filter membrane with the aperture of 200nm for 3 cycles to obtain the nano particles.
According to one aspect of the invention, the application of the nanoparticle in preparing a medicament for treating cancer is provided.
Preferably, the cancer is non-small cell lung cancer.
The nano particles provided by the invention can be widely applied to the preparation process of medicaments for treating cancers, in particular to the preparation process of medicaments for treating non-small cell lung cancer.
The technical solution of the present invention will be further described with reference to the following examples.
Example 1 extraction of A549 cell membranes
A method for extracting A549 cell membranes comprises the following steps:
(1) and preparation of reagents: dissolving the membrane protein extraction reagent at room temperature, uniformly mixing, placing a proper amount of the membrane protein extraction reagent on ice, and adding PMSF (phenylmethylsulfonyl fluoride) into the membrane protein extraction reagent before use to ensure that the final concentration of PMSF in the membrane protein extraction reagent is 1 mmol/L.
(2) And preparing cells: selecting A549 cells in logarithmic growth phase and good state, washing once with PBS, scraping the cells, centrifugally collecting the cells, discarding supernatant, and leaving cell precipitate for later use. Cells were washed twice with ice PBS and counted. Adding 2X 10 membrane protein extraction reagent into 1mL of PMSF-added membrane protein extraction reagent7And (4) gently mixing the mixture in each cell, and carrying out ice bath for 10-15 min.
(3) And cell disruption: and (3) breaking the cells by adopting a freeze-thaw method, repeatedly freezing and thawing the sample in the step (2) in a water bath at 37 ℃ and liquid nitrogen for 3 times, taking a small amount of sample, observing the state of the cells under a microscope, and increasing the freeze-thaw times if the breaking degree is not enough until the cell breaking degree is more than 70%.
(4) Removal of nuclei and unbroken cells: centrifugation at 700g for 10min at 4 ℃ was carried out, and the supernatant was carefully collected and placed in a new centrifuge tube without touching the pellet.
(5) Precipitation of cell membrane debris: centrifuging at 14000g for 30min at 4 deg.C to precipitate cell membrane debris, adding 20 μ L DW, mixing, and extracting to obtain A549 cell membrane.
The membrane protein extraction reagent is as follows: bi yun tian, P0033.
Example 2 preparation of nano self-assembled particles
A preparation method of nano self-assembled particles comprises the following steps:
(a) dissolving digoxin in absolute ethyl alcohol to prepare a solution with the digoxin final concentration of 1mmol/L, and then filtering the solution through a 0.22 mu M microporous membrane to obtain a digoxin absolute ethyl alcohol solution with the concentration of 1 mmol/L;
(b) adding 17mL of ultrapure water and a stirrer into a eggplant-shaped bottle, and placing the bottle on a water bath at 50 ℃ for preheating; under the magnetic stirring of 1000r/min, slowly and uniformly dripping 2mL of fully dissolved 1mmol/L digoxin absolute ethyl alcohol solution by using a syringe pump, uncovering the cover, and continuously stirring for 5min to obtain a solution A;
(c) dissolving adriamycin hydrochloride into deionized water to prepare adriamycin hydrochloride (ADR-HCl) aqueous solution with the concentration of 1mmol/L, then adding 1mL of 1mmol/L ADR-HCl aqueous solution into the solution A, closing a cover, and continuing stirring for 1h to obtain the nano self-assembled particles.
EXAMPLE 3 preparation of nanoparticles
A method for preparing nanoparticles, comprising the steps of:
(d) 4mL of the nano self-assembled particles prepared in example 2 were collected in A5 mL centrifuge tube, and then 12. mu.L of the A549 cell membrane solution prepared in example 1 was added thereto and uniformly blown by a pipette to obtain a solution B.
(e) And after the liposome extrusion instrument is cleaned by ultrapure water, the liposome extrusion instrument is utilized to extrude the solution B for at least 3 cycles through a filter membrane with the aperture of 400nm, and the solution B is continuously extruded for at least 3 cycles through the filter membrane with the aperture of 200nm to obtain the nano particles.
Example 4 characterization of nanoparticles
In this example, the nano self-assembled particle prepared in example 2 and the nano particle prepared in example 3 are substantially characterized by the following specific methods:
(1) the tyndall effect: placing the prepared nano self-assembled particles and nano particles in an absorption cell, and observing the Tyndall effect by laser pen irradiation;
the Tyndall effect is an important part in colloid chemistry, some important concepts in nano materials are established on the classical theory and the mature idea of colloid chemistry, colloid is used as a dispersion system and comprises a dispersing agent and a dispersoid, and when the particle size of the dispersoid is in the range of 1-100 nm, the formed dispersion system is called colloid. When the nano particles are dispersed in a liquid phase system, a liquid sol is formed, and when the particle size is smaller than the visible wavelength (400-700 nm), the particles are highly dispersed in the liquid sol, and the obvious scattering effect, namely the Tyndall effect, is achieved.
As shown in fig. 1, the nano self-assembled particles (DANPs) prepared in example 2 and the nano particles (DAMNPs) prepared in example 3 both showed the tyndall phenomenon, and the digoxin doxorubicin hydrochloride protocol (D + a) showed no light scattering.
(2) And Transmission Electron Microscope (TEM) observation: the nano self-assembled particles prepared in example 2 and the nano particles prepared in example 3 are observed by a Transmission Electron Microscope (TEM), which is a common and intuitive method for observing the size and shape of the nano particles.
As shown in FIG. 2, it can be seen from the transmission electron micrograph that the dispersibility of the DANPs and the DAMNPs is relatively good, and the nano self-assembled particles (DANPs) prepared in example 2 are spherical and have uniform particle size distribution and particle size ranging from about 100nm to about 150 nm. And the nanoparticles (DAMNP) prepared in example 3S) The particle size of the particles is larger, and the A549 tumor cell membrane with stronger light transmittance which is obviously observed in the particles wraps the D with weaker light transmittanceANPs。
Example 5 in vitro antitumor assay of nanoparticles
(one), evaluation of nanoparticles (DAMNP) prepared in example 3S) For the proliferation inhibition of A549 cells, the cell survival rate is detected by adopting an MTT method. The MTT method mainly utilizes succinate dehydrogenase in mitochondria of living cells to reduce MTT to Formazan (Formazan), which is blue-violet crystalline and insoluble in water and deposited in cells, and such a reaction does not occur in dead cells. Formazan can be dissolved in DMSO, and the proliferation and survival conditions of cells can be indirectly detected by measuring the optical density value by using an enzyme-labeling instrument. The detection method has the advantages of low cost, convenient operation and high sensitivity.
Therefore, the MTT method is adopted to detect the influence of digoxin adriamycin hydrochloride (D + A), DANPs and DANMNPs on the proliferation of A549 cells. Three groups of D + A, DANPs and DANMNPs are dosed according to the drug concentration gradient, and the concentration ratio of Digoxin (DIG) to Adriamycin (ADR) is 2: 1.
The results are shown in fig. 3, and the cytotoxic effect of the DAMNPs on a549 cells was dose-dependent 72h after administration. When the concentration of the medicament is lower, the proliferation inhibition effect of the three groups on A549 cells is not obviously different, but the proliferation inhibition effect of the DAMNPs is obviously enhanced when the concentration of DIG is 0.5 mu M, ADR and the concentration of DIG is 0.25 mu M compared with the other two groups along with the increase of the administration concentration. The results indicate that the inhibition of a549 cells by the DAMNPs is dose-dependent.
And (II) apoptosis refers to the autonomous and orderly cell death of cells in a gene regulation mode in order to ensure that the self life activities can be normally carried out. The occurrence of apoptosis is an active death process chosen for better adaptation to the living environment. Many studies have shown that induction of apoptosis of tumor cells can effectively treat cancer, and various drugs exert tumor cell proliferation inhibitory effects by inducing apoptosis of tumor cells.
In order to investigate whether the inhibition effect of D + A, DANPs and DAMNPs on the proliferation of A549 cells is caused by apoptosis, the present example employs an APC single staining method to detect the effect of D + A, DANPs and DAMNPs on the apoptosis of A549 cells.
The results are shown in fig. 4, and the apoptosis induction rates of a549 cells in the D + a group, DANPs group and damnpps group were 9.4%, 14.7% and 17.2%, respectively, which were significantly higher than 5.5% of the control group, with the highest apoptosis rate induced by damnpps. The experimental results are tested by using GraphPad Prism 7 software, and compared with the apoptosis numbers of a D + A group, a DANPS group and a DAMNPs group (control), the numbers have significant differences.
Example 6 in vivo antitumor assay of nanoparticles
(I), zebra fish experiment:
(1) and feeding zebra fish: the experimental species used was wild AB zebra fish, the circulating water temperature was (28.5 + -0.5) deg.C, the pH was about 7.2, and the total hardness of the water was 62mg/L (as CaCO)3Meter), the conductivity was 485 mus. The illumination time is 14h, and the treatment is carried out for 10h in the dark. The zebra fish is fed with fairy shrimp 1 time respectively in the morning and evening.
(2) Establishment of zebra fish tumor model
1. Obtaining zebra fish embryos: the male and female adult AB-line zebra fish are separately placed in a breeding tank 1: 1 in the evening before the experiment, and the male and female fish are separated by a drawing plate in the middle. The next day 8: and (5) pulling the plate at 00am, and collecting the roes in time after spawning for 15 min. The eggs are washed by using E3 culture solution, and dead eggs are removed so as not to affect healthy eggs. Subsequently, the cells were placed in a 10cm petri dish, a culture solution containing 0.2mmol/L of PTU was added thereto, and the mixture was incubated in an incubator at 32 ℃ for 48 hours.
2. Preparation of a549 cell suspension: selecting A549 cells with good cell state and in logarithmic growth phase, washing with PBS, digesting with pancreatin, and making into cell suspension with PBS. Adding DIO dye, incubating at 37 deg.C for 20min, washing two sides with PBS, and making into 5 × 106Cell suspension/mL.
3. Injecting zebra fish embryos in a microinjection mode: zebrafish embryos were anesthetized with 1.2mmol/L tricaine at 48h, and subsequently placed on a 2% agarose gel. Approximately 20nL volumes of cell suspension containing around 100 cells per yolk sac of zebrafish embryos were injected with a microinjector. After cell injection, embryos were screened using a fluorescence microscope. Selecting embryos with basically consistent fluorescence intensity and cell area size for subsequent experiments. The embryos are transferred into 24-well plates, 2mL of E3 culture solution containing different drugs is added into each well, and the mixture is placed into an incubator at 32 ℃ for culturing for 48 hours.
4. Grouping and administration modes: the embryos were divided into four groups, namely a control group, a 1.4. mu.M DIG + 0.7. mu.M ADR group, a DANPS group (1.4. mu.M DIG + 0.7. mu.M ADR), and a DANMNPs group (1.4. mu.M Digoxin + 0.7. mu.M ADR), the administration method was a dipping method, the administration was continued for 48 hours, and the fluorescence intensity of A549 cells in zebrafish bodies was observed by a fluorescence microscope after 48 hours.
The results are shown in fig. 5, and fluorescence microscope observation after the control group and each administration group act for 48 hours shows that the fluorescence intensity of the tumors in the zebra fish bodies is obviously reduced compared with the D + A group, the DANPS group and the DANMNPs and the control group, which indicates that three groups can effectively inhibit the proliferation of the tumor cells in the zebra fish bodies. Compared with the D + A group and the DANPs group, the fluorescence intensity of the tumor is also obviously reduced. In fig. 5, the abscissa indicates zebra fish (Bright field) photographed in the Bright field, DIO group photographed under fluorescence (cell membrane staining), and the first two groups were overlaid, i.e., the same fish were photographed under fluorescence under different light sources at the same position (Merge). As can be seen from FIG. 5, when the same concentration of DAMNPs acts on zebrafish, the effect on tumor cells is enhanced and the inhibition effect is more remarkable than that of the D + A group and the DANPS group.
The first step of nude mouse experiment:
(1) establishment of mouse subcutaneous transplantation tumor model
1. Preparation of a549 cell suspension: selecting A549 cells with good cell state and in logarithmic phase, washing with PBS, digesting with pancreatin, adding culture medium to prepare cell suspension, and counting. Centrifuging at 1000rpm for 5min, washing with PBS twice to obtain final product with density of 1 × 107Cell suspension/mL.
2. Culturing the transplantation tumor: the right flank of the nude mice was injected subcutaneously with 200. mu.L of cell suspension, approximately 2X 10 per nude mouse6And (4) cells.
3. And (4) transplanting tumor masses, and autoclaving the surgical scissors, the surgical forceps and the inoculating needle in advance. When the tumor grows to meet the number and size of tumor blocks required by the transplantation experiment, the tumor tissue in the nude mouse is taken out and placed in a culture dish, and then normal saline is added to cut the tissue into pieces20~30mm3The tumor mass is then transplanted into healthy BALB/c nude mice by an inoculation needle for about 4-5 weeks.
4. Grouping and administration modes: when the tumor area of the nude mouse grows to 80-100 mm3The samples were randomly divided into 4 groups, namely, control group, 1mg/kg DIG +0.4mg/kg ADR group, DANPS (1mg/kg DIG +0.4mg/kg ADR) group, and DAMNPs (1mg/kg DIG +0.4mg/kg ADR) group. The control group was injected with a corresponding volume of physiological saline every three days, and the body weight was weighed and the tumor volume (tumor volume: tumor length × tumor width) was measured2/2). Nude mice were treated after 15 days, tumor tissues were taken out, photographed and weighed, and the change of tumor volume and the average tumor weight of each group at the time of sacrifice were counted. After the treatment, half of the tissues of the tumor, heart, liver, spleen, lung and kidney are frozen in a refrigerator at the temperature of minus 80 ℃, and half of the tissues are fixed in 4% paraformaldehyde.
As a result, as shown in fig. 6, 7 and 8, the tumor volume increased rapidly with time in the control group, and the tumor volume increased with time in the D + a group and DANPs group, respectively, in the administration process, while the growth of the dannps group was much slower than that of the other three groups. Each group was sacrificed 18 days after dosing and the tumor volume and tumor weight of each group were statistically analyzed. The D + A group and the DANPs group were decreased in tumor volume and tumor weight at 18 days after administration, and were statistically different from those of the control group. The DAMNPs group not only has obviously reduced tumor volume compared with the control group, but also has obviously reduced tumor weight. Compared with the D + A group and the DANPs group, the tumor volume and the tumor weight are also obviously reduced and have statistical significance.
In summary, the nanoparticles prepared by the method have the advantages of high drug loading and strong tumor targeting property, improve the existing anti-tumor treatment effect, have stronger targeting property, and can effectively reduce the toxicity of the drugs to normal tissues.
Finally, it should be noted that: the above embodiments are only used to illustrate the technical solution of the present invention, and not to limit the same; while the invention has been described in detail and with reference to the foregoing embodiments, it will be understood by those skilled in the art that: the technical solutions described in the foregoing embodiments may still be modified, or some or all of the technical features may be equivalently replaced; and the modifications or the substitutions do not make the essence of the corresponding technical solutions depart from the scope of the technical solutions of the embodiments of the present invention.

Claims (10)

1. A nanoparticle comprising a nano self-assembled particle and a homotypic tumor cell membrane;
the nano self-assembled particles are mainly prepared from digoxin and adriamycin;
the homotypic tumor cell membrane is coated on the surface of the nano self-assembly particles.
2. Nanoparticles according to claim 1, wherein the hydrated particle size of the nanoparticles is 150-200 nm;
preferably, the hydrated particle size of the nano self-assembly particles is 100-150 nm.
3. Nanoparticle according to claim 1, wherein said homotypic tumor cell membrane comprises a non-small cell lung cancer cell membrane, preferably an a549 tumor cell membrane.
4. A method for preparing nanoparticles according to any one of claims 1 to 3, comprising the following steps:
(A) preparing nano self-assembled particles by using a precipitation method;
(B) providing homotypic tumor cell membranes, uniformly mixing the homotypic tumor cell membranes with the nano self-assembly particles prepared in the step (A), and then extruding in a grading manner to obtain the nano particles.
5. The method for preparing nanoparticles according to claim 4, wherein the step (A) comprises the steps of:
(a) Dissolving digoxin in an organic solution to obtain a digoxin solution;
(b) dissolving adriamycin in deionized water to obtain adriamycin solution;
(c) under the condition of stirring, firstly injecting the digoxin solution obtained in the step (a) into deionized water at a constant speed to obtain a solution A; and (c) adding the adriamycin solution obtained in the step (b) into the solution A, and uniformly mixing to obtain the nano self-assembled particles.
6. The method for preparing nanoparticles according to claim 5, wherein the organic solution in the step (a) comprises any one of absolute ethyl alcohol, methanol, isopropanol or n-butanol, preferably absolute ethyl alcohol;
preferably, when the organic solution in the step (a) is absolute ethyl alcohol, the molar concentration of the digoxin solution is 0.1-2 mmol/L, and 1mmol/L is preferred;
preferably, when the injection speed of the digoxin solution in the step (c) is 20-30ml/h, preferably 25 ml/h;
preferably, the molar concentration of the adriamycin solution in the step (b) is 0.1-10 mmol/L, and 1mmol/L is preferred.
7. The method for preparing nanoparticles according to claim 5, wherein the molar concentration of digoxin in the solution A in the step (c) is 0.1 mmol/L;
preferably, the volume ratio of the solution A to the adriamycin solution in the step (c) is 18-20: 1, and is preferably 19: 1;
Preferably, the stirring reaction condition of step (c) at least satisfies at least one of the following conditions:
stirring at the rotating speed of 1000-1200 r/min, at the stirring temperature of 48-52 ℃, at the dropping speed of 25ml/h of digoxin solution and for 0.8-1.5 h;
more preferably, the stirring reaction condition of step (c) at least satisfies at least one of the following conditions:
the stirring speed is 1000r/min, the stirring temperature is 50 ℃, the dropping speed is 25ml/h, and the stirring time is 1 h.
8. The method for preparing nanoparticles according to claim 4, wherein the step (B) comprises the steps of:
(d) providing a homotypic tumor cell membrane containing solution, and then uniformly mixing the homotypic tumor cell membrane with the nano self-assembly particles prepared in the step (c) to obtain a solution B;
(e) extruding the solution B through a filter membrane with the aperture of 200-400 nm for 3-6 cycles in a grading manner by using a liposome extruder to obtain nanoparticles;
preferably, the homotypic tumor cell membrane content in the homotypic tumor cell membrane-containing solution in the step (d) is 0.1 mg/mL;
preferably, the volume ratio of the homotypic tumor cell membrane-containing solution in the step (d) to the nano self-assembly particles prepared in the step (c) is 1-10: 100-500.
9. A method for the preparation of nanoparticles according to claim 4, characterized in that it comprises the following steps:
(a) dissolving digoxin in absolute ethyl alcohol to obtain a digoxin solution with the molar concentration of 0.1-2 mmol/L;
(b) dissolving adriamycin in deionized water to obtain an adriamycin solution with the molar concentration of 0.1-10 mmol/L;
(c) injecting the digoxin solution obtained in the step (a) into deionized water to obtain a solution A with a digoxin molar concentration of 0.1mmol/L under the stirring conditions that the stirring speed is 1000-1200 r/min and the temperature is 48-52 ℃; adding the adriamycin solution obtained in the step (b) into the solution A, and stirring for 0.8-1.5 hours to obtain nano self-assembled particles; the volume ratio of the solution A to the adriamycin solution is 18-20: 1;
(d) providing a homotypic tumor cell membrane-containing solution with the concentration of 0.1mg/mL, and then uniformly mixing homotypic tumor cell membranes and the nano self-assembly particles prepared in the step (c) in a volume ratio of 1-10: 100-500 to obtain a solution B;
(e) extruding the solution B through a filter membrane with the aperture of 400nm for 3-6 cycles by using a liposome extruder, and then extruding through a filter membrane with the aperture of 200nm for 3-6 cycles to obtain nano particles;
Preferably, the preparation method comprises the following steps:
(a) dissolving digoxin in absolute ethyl alcohol to obtain a digoxin solution with the molar concentration of 1 mmol/L;
(b) dissolving the adriamycin in deionized water to obtain an adriamycin solution with the molar concentration of 1 mmol/L;
(c) injecting the digoxin solution obtained in the step (a) into deionized water to obtain a solution A with the digoxin molar concentration of 0.1mmol/L under the stirring conditions that the stirring speed is 1000r/min and the temperature is 50 ℃; adding the adriamycin solution obtained in the step (b) into the solution A, and stirring for 1h to obtain nano self-assembled particles; the volume ratio of the solution A to the adriamycin solution is 19: 1;
(d) providing a homotypic tumor cell membrane-containing solution with the concentration of 0.1mg/mL, and then uniformly mixing homotypic tumor cell membranes and the nano self-assembly particles prepared in the step (c) in a volume ratio of 1-10: 100-500 to obtain a solution B;
(e) and extruding the solution B through a filter membrane with the aperture of 400nm for 3 cycles by using a liposome extruder, and then extruding through a filter membrane with the aperture of 200nm for 3 cycles to obtain the nano particles.
10. Use of the nanoparticle according to any one of claims 1 to 3 for the preparation of a medicament for the treatment of a tumour.
CN202010708342.4A 2020-07-15 2020-07-15 Nano particles and preparation method and application thereof Pending CN111840305A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010708342.4A CN111840305A (en) 2020-07-15 2020-07-15 Nano particles and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010708342.4A CN111840305A (en) 2020-07-15 2020-07-15 Nano particles and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN111840305A true CN111840305A (en) 2020-10-30

Family

ID=73001428

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010708342.4A Pending CN111840305A (en) 2020-07-15 2020-07-15 Nano particles and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN111840305A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113368079A (en) * 2021-05-11 2021-09-10 暨南大学 Cancer cell membrane-coated drug-loaded lignin nanoparticle and preparation method and application thereof
CN113416752A (en) * 2021-06-23 2021-09-21 周娟 Mog1 gene knockout zebra fish model and application

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113368079A (en) * 2021-05-11 2021-09-10 暨南大学 Cancer cell membrane-coated drug-loaded lignin nanoparticle and preparation method and application thereof
CN113416752A (en) * 2021-06-23 2021-09-21 周娟 Mog1 gene knockout zebra fish model and application

Similar Documents

Publication Publication Date Title
CN110215438B (en) Preparation method and application of mesoporous silicon nanoparticles carrying anthracyclines and photosensitizers
CN106821985B (en) Aptamer-modified oxygen-carrying and drug-carrying multifunctional liposome compound
CN110368374A (en) A kind of Antitumor platinum drugs mineralization protein nanoparticle and its preparation method and application
CN111840305A (en) Nano particles and preparation method and application thereof
KR102063571B1 (en) Method for producing surface-modified exosomes from various cells
CN112494495B (en) Preparation method of cancer cell membrane chimeric liposome nano drug delivery system
CN108653733A (en) Polymer vesicle and the preparation of double load anthracene nucleus medicaments and photosensitizer with bubble formation function
Xuan et al. Bismuth particles imbedded degradable nanohydrogel prepared by one-step method for tumor dual-mode imaging and chemo-photothermal combined therapy
CN114831931A (en) Medicinal solution with anti-tumor synergistic attenuation effects and medicinal composition containing medicinal solution
CN108178803A (en) A kind of preparation of cinnamic acid-dextran polymer self-assembled nanometer grain for carrying medicine and its antitumor application thereof
Cao et al. Heparin modified photosensitizer-loaded liposomes for tumor treatment and alleviating metastasis in phototherapy
CN106344924A (en) Nano-formulation combined with metabolic block and drug resistant reverse application thereof
Ostovar et al. Nanocomposite of chitosan/gelatin/carbon quantum dots as a biocompatible and efficient nanocarrier for improving the Curcumin delivery restrictions to treat brain cancer
CN109200021B (en) Lysosome targeted pH sensitive nanoparticle and preparation method and application thereof
CN111956806B (en) Drug carrier, micelle, medicament, and preparation method and application thereof
CN113648415A (en) Organic metal nano drug-loaded particles for targeted therapy of tumors and preparation method thereof
CN111249235B (en) Brain targeting nanoliposome loaded with positive polymer/miR-195 compound, and preparation method and application thereof
CN110859820A (en) Biocompatible alkaline earth metal peroxide nano preparation, preparation method and application thereof
CN109364253A (en) One kind is for improving infiltrative nanoparticle of tumor tissues and its preparation method and application
CN115068444A (en) Liposome nanoparticle wrapped by macrophage membrane and preparation method thereof
CN102716137A (en) Preparation method and application of fluorescence nanometer diamond-doxycycline compound
CN108451905B (en) Gambogic acid nanoemulsion preparation and preparation method and application thereof
CN110151728A (en) A kind of double erythrocyte membrane composite nano-granules for carrying the pure and mild IR780 of Taxotere
CN115252580B (en) Drug-loaded erythrocyte membrane nanoparticle and preparation method and application thereof
CN108354896B (en) Raltitrexed hydrogel and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
TA01 Transfer of patent application right

Effective date of registration: 20220830

Address after: 300070 No. 22 Meteorological Observatory Road, Heping District, Tianjin

Applicant after: Tianjin Medical University

Applicant after: ACADEMY OF MILITARY MEDICAL SCIENCES

Address before: 300070 No. 22 Meteorological Observatory Road, Heping District, Tianjin

Applicant before: Tianjin Medical University

TA01 Transfer of patent application right
DD01 Delivery of document by public notice

Addressee: Wang Yingying

Document name: Notice of Conformity

DD01 Delivery of document by public notice
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination