CN114099706A - MOF drug carrier for treating breast cancer and preparation method thereof - Google Patents

MOF drug carrier for treating breast cancer and preparation method thereof Download PDF

Info

Publication number
CN114099706A
CN114099706A CN202111442947.4A CN202111442947A CN114099706A CN 114099706 A CN114099706 A CN 114099706A CN 202111442947 A CN202111442947 A CN 202111442947A CN 114099706 A CN114099706 A CN 114099706A
Authority
CN
China
Prior art keywords
mof
dox
solution
antisense
drug
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
CN202111442947.4A
Other languages
Chinese (zh)
Inventor
杨兆琪
龚森怡
尚孟迪
金坚
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.)
Jiangnan University
Original Assignee
Jiangnan 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 Jiangnan University filed Critical Jiangnan University
Priority to CN202111442947.4A priority Critical patent/CN114099706A/en
Publication of CN114099706A publication Critical patent/CN114099706A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6949Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit inclusion complexes, e.g. clathrates, cavitates or fullerenes
    • 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
    • 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
    • A61K41/0071PDT with porphyrins having exactly 20 ring atoms, i.e. based on the non-expanded tetrapyrrolic ring system, e.g. bacteriochlorin, chlorin-e6, or phthalocyanines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

The invention relates to the technical field of medicines, in particular to an MOF (metal-organic framework) medicine carrier, a preparation method thereof and a medicine carrying system containing the MOF medicine carrier. The novel reduction response type metal organic framework is synthesized by taking zinc nitrate and 2,2' -dipyridyl disulfide as main raw materials, has a proper particle size (less than 200nm) and a large specific surface area, can be used as a drug carrier, loads adriamycin and antisense nucleic acid and wraps hyaluronic acid. In a cytotoxicity test, compared with the IC50 of single Dox on MCF-7/ADM cells, the IC50 of the Dox on the MCF-7/ADM cells after the Dox is loaded in a nano system (Dox/Antisense/HA @ MOF) is greatly reduced, and the nano system is proved to have the effect of resisting drugs and reduce the dose of chemotherapeutic drugs; the drug resistance effect of the novel nano system is determined by measuring the content change of glutathione in cells, the expression condition of related protein and the active oxygen generating capacity.

Description

MOF drug carrier for treating breast cancer and preparation method thereof
Technical Field
The invention relates to the technical field of medicines, in particular to an MOF (Metal-organic framework) medicine carrier for treating breast cancer and a preparation method thereof.
Background
The current cancer incidence and mortality in china is high. Like most other countries, breast cancer is also the most common cancer among women in china. Chemotherapy is one of the most important means of treating malignancies today. However, almost all current drugs, over the course of time of use by a patient, develop resistance to the drug by cancer cells, such that the effect of the drug on the cancer cells is reduced. Drug resistance in cancer cells is one of the major causes of failure of cancer therapy, which may lead to rapid recurrence of the cancer, ultimately leading to death of the patient.
With the continuous development of medicine and molecular biology, mechanisms of tumor drug resistance have been revealed, such as overexpression of atp binding cassette transporters (p-glycoproteins), overexpression of anti-apoptotic proteins, repair of DNA damage and detoxification systems (presence of intracellular glutathione in tumors), epigenetic changes, inhibition of cell death, alteration of drug targets, Epithelial Mesenchymal Transition (EMT), and the like. Many therapeutic strategies have been developed for these tumor resistance mechanisms, such as p-glycoprotein reversal strategies, gene therapy, and the like. With the development of nanotechnology, designing reasonable nanomaterials to resist the drug resistance of tumor cells is becoming a new strategy to improve the effect of chemotherapy.
At present, the research at home and abroad mainly focuses on improving the tumor drug resistance problem by researching a single drug resistance mechanism. However, no research for designing a novel nano material to improve the drug resistance problem of breast cancer by combining the following drug resistance mechanisms, namely p-glycoprotein overexpression, anti-apoptosis protein overexpression and intracellular high-level glutathione) is available.
Disclosure of Invention
Aiming at the problems, the invention designs and successfully optimizes and synthesizes a novel reduction response type Metal Organic Framework (MOF) system, uses the MOF system as a nano-drug carrier, and constructs a drug carrying system containing the nano-drug carrier by combining a drug resistance mechanism of p-glycoprotein overexpression, anti-apoptosis protein overexpression and intracellular high-level glutathione, thereby improving the drug resistance of breast cancer to a certain extent and being beneficial to improving the treatment effect of drugs in the drug carrying system on the breast cancer.
The invention provides a method for synthesizing a novel reduction-responsive metal organic framework, which is characterized in that doxorubicin (Dox) and an Antisense nucleic acid (Antisense) of HIF-1 alpha are loaded, and the 5' end of the Antisense nucleic acid is modified with a Ce6 photosensitizer (purchased from Takara company, see Aptamer loaded MoS)2nanoplates as nanoprobes for detection of intracellular ATP and controllable photonic thermal. Li, J., Lin, D., Jiang, w.T., Lei, Bao, Yao, p.H., Huang, x.J., and Jun, S.Y.2015.Nanoscale,38.doi:10.1039/C5NR02224J), and further coated with Hyaluronic Acid (HA) to form a novel nanosystem.
The principle of the invention is shown in fig. 19: the nano material contains disulfide bonds and can consume Glutathione (GSH), so that iron death is induced to resist drug resistance; the nano system contains antisense nucleic acid and Ce6 photosensitizer, and the antisense nucleic acid can inhibit the expression of p-glycoprotein to reduce drug efflux and relieve drug resistance; the Ce6 photosensitizer can inhibit the overexpression of anti-apoptotic proteins by generating Reactive Oxygen Species (ROS) through photodynamic therapy. Based on the three drug resistance mechanisms (p-glycoprotein overexpression, anti-apoptosis protein overexpression and intracellular high-level glutathione), the invention specifies the strategy and the technical scheme for solving the drug resistance, improves the effect of the adriamycin drug on breast cancer drug-resistant cells through the nano-system combined photodynamic therapy, and provides a new solution for solving the technical problem of the drug resistance of breast cancer.
The technical scheme of the invention is as follows:
the first aspect of the invention provides a preparation method of an MOF drug carrier, which comprises the following steps:
preparation of solution A: dissolving zinc nitrate in N, N-dimethylformamide, and removing insoluble impurities to obtain solution A;
and (3) preparation of a solution B: dissolving 2,2' -dipyridyl disulfide and 4-dimethylaminopyridine in dimethyl sulfoxide, removing insoluble impurities, adding polyvinylpyrrolidone, and mixing to obtain solution B;
and (3) quickly adding the solution B into the solution A, heating and stirring for reaction, centrifuging the obtained reaction solution, taking precipitate, and washing with ultrapure water to obtain MOF nano particles, namely the MOF drug carrier.
In a preferred embodiment of the present invention, the mass ratio of zinc nitrate to 2,2' -dithiodipyridine in the mixed system of solution A and solution B is (1.1-1.3): 1. Furthermore, the concentration of zinc nitrate in the solution A is 0.85 mug/mL; the concentration of 2,2' -dithiodipyridine in the solution B is 0.7 mug/mL; the mixing volume ratio of the solution A to the solution B is 1: 1.
In a preferred embodiment of the present invention, the temperature of the heating and stirring reaction is 50 to 80 ℃.
A second aspect of the invention provides MOF nanoparticles made by the foregoing method.
A third aspect of the invention provides the use of the MOF nanoparticles described above as drug carriers.
The fourth aspect of the invention provides a medicine carrying system based on MOF nanoparticles, which is prepared by fully mixing the components including the MOF nanoparticles, HIF-1 alpha antisense nucleic acid modified with photosensitizer and adriamycin in a solvent.
The reaction equation is as follows:
Figure BDA0003384176380000021
as a preferred embodiment of the present invention, the drug delivery system further comprises hyaluronic acid.
As a preferred embodiment of the present invention, the final concentration of hyaluronic acid is 200. mu.g/mL.
As a preferred embodiment of the invention, the final concentration of the MOF nanoparticles is 200. mu.g/mL.
In a preferred embodiment of the present invention, the photosensitizer-modified HIF-1 α antisense nucleic acid is present at a final concentration of 200 nM.
In a preferred embodiment of the present invention, the final doxorubicin concentration is 0.2 to 5. mu.g/mL.
The third aspect of the present invention is to provide an application of the aforementioned drug-carrying system in preparing anti-breast cancer drugs. In particular, the drug-loaded system has good application prospect in the aspect of delaying the drug resistance of the anti-tumor drug. The drug-loading system has obvious effects in the aspects of reducing the content of glutathione in drug-resistant tumor cells, inhibiting the expression of p-glycoprotein in the drug-resistant tumor cells, inhibiting the expression of anti-apoptosis protein in the drug-resistant tumor cells and the like.
The beneficial technical effects of the invention are as follows:
1. the novel reduction response type metal organic framework is synthesized by taking zinc nitrate and 2,2' -dipyridyl disulfide as main raw materials, has a proper particle size (less than 200nm) and a large specific surface area, can be used as a drug carrier, loads adriamycin and antisense nucleic acid and wraps hyaluronic acid. IC on MCF-7/ADM cells with Dox alone in cytotoxicity assays50In contrast, IC of Dox on MCF-7/ADM cells after Loading of Dox on nanosystems (Dox/Antisense/HA @ MOF)50The dosage of the chemotherapeutic drug is reduced; the drug resistance effect of the novel nano system is determined by measuring the content change of glutathione in cells, the expression condition of related protein and the active oxygen generating capacity.
2. The novel nano system can improve the drug-resistant cell effect of the adriamycin drug on the breast cancer, has certain guiding significance for exploring the aspects of improving the multi-drug resistance of the cancer and the like by utilizing nano materials, and makes the nano system possible to be used as a new anti-tumor drug in the field of drug research and development.
Drawings
FIG. 1 is a schematic diagram of the morphology characterization of the nanomaterial prepared in example 1 of the present invention. FIG. 1(A) is a transmission electron microscope image of MOF nanoparticles prepared from solution A and solution B at a reaction volume ratio of 1:2, and FIG. 1(B) is a transmission electron microscope image of MOF nanoparticles prepared from solution A and solution B at a reaction volume ratio of 1: 2.
FIG. 2 is a graph of the average particle size of MOF nanoparticles prepared in example 1 using a reaction volume ratio of solution A to solution B of 1:2 over 15 days using Dynamic Light Scattering (DLS) measurements.
FIG. 3 shows the dispersity of MOF nanoparticles prepared in example 1, in which the reaction volume ratio of solution A to solution B is 1:2 within 15 days.
FIG. 4 shows the disintegration of MOF nanoparticles prepared from example 1, wherein the reaction volume ratio of solution A to solution B is 1:2, in Glutathione (GSH) with different concentrations
FIG. 5 is an X-ray diffraction (XRD) pattern of MOF nanoparticles prepared in example 1 with a 1:2 reaction volume ratio of solution A to solution B.
FIG. 6 is the BET surface analysis spectrum and the numerical values of the specific surface area and pore volume obtained by the BET analysis of MOF nanoparticles prepared in example 1 in which the reaction volume ratio of liquid A to liquid B is 1: 2.
FIG. 7 is a schematic diagram of the concentration optimization of MOF nanoparticles in MCF-7/ADM cells in the second embodiment of the invention 1.
FIG. 8 is a schematic diagram of the MOF nanoparticle-loaded HIF-1 α Antisense nucleic acid modified with Ce6 photosensitizer (denoted as Antisense) for maximum concentration optimization in example 1, scheme II of the present invention.
FIG. 9 is a schematic diagram of the Dox release capacity of Dox/Antisense @ MOF nanoparticles under different conditions according to an embodiment of the present invention.
FIG. 10 is a schematic diagram of toxicity of different nanosystems on MCF-7/ADM cells obtained using the MTT method.
FIG. 11 shows Dox loading after nanosystems (Dox/Antisense/HA @ MOF) [ FIG. 11(A)]And Dox alone [ FIG. 11(B)]IC on MCF-7/ADM cells50
FIG. 12 is a graph of viable and dead MCF-7/ADM cells after treatment with different groups.
FIG. 13 is a graph of apoptosis of MCF-7/ADM cells after treatment with different groups.
Fig. 14 is a schematic diagram of the results of active oxygen detection of nanoparticles added with different concentrations of Antisense under the stimulation of light with specific wavelength in example 9 of the present invention.
FIG. 15 is a graph showing the level of reactive oxygen species obtained by analyzing the fluorescence pattern of FIG. 14.
FIG. 16 is a measurement of glutathione content in nanomaterial-depleted cells of example 10 of the present invention.
FIG. 17 is a study of the uptake of Dox/Antisense @ MOF nanoparticles from example 11 by MCF-7/ADM cells.
FIG. 18 is a study of the inhibitory effect of Dox/Antisense @ MOF nanoparticles on related proteins in accordance with an embodiment of the present invention: (A) is a western blot of BCL-2 protein in MCF-7/ADM after the processing of a nano system loaded with different concentrations of Antisense, and (B) is a protein level graph obtained by gray scale analysis of (A); (C) is a western blot of P-gp protein in MCF-7/ADM after being processed by a nano system loaded with antibiotics with different concentrations, and (D) is a protein level graph obtained by gray level analysis of (C); (E) is a western blot of Gpx4 protein in MCF-7/ADM after being processed by a nano system loaded with different concentrations of Antisense, and (F) is a protein level graph obtained by gray scale analysis of (E).
Fig. 19 is a schematic diagram of the present invention.
Detailed Description
The present invention will be described in detail with reference to the accompanying drawings and examples.
Materials, reagents and the like used in the following examples are commercially available unless otherwise specified.
HIF-1 alpha Antisense nucleic acid (Antisense) and 5' end of the Antisense nucleic acid modified with Ce6 photosensitizer (hereinafter abbreviated as Antisense, available from Takara corporation, for preparation see Aptamer loaded MoS2 nanoplates as nanoprobes for detection of intracellular ATP and controllable photodynamic therapy.Li,J.,Lin,D.,Jiang,w.T.,Lei,Bao.,Yao,p.H.,,Huang,x.J.,and Jun,S.Y.2015.Nanoscale,38.doi:10.1039/C5NR02224J)。
Example 1: synthesis and drug-loading performance evaluation of novel metal organic framework MOF
Solution A: to 7mL of N, N-Dimethylformamide (DMF), 0.6g of zinc nitrate (0.85. mu.g/mL) was added, which was precisely weighed, sufficiently dissolved, and then the dissolved zinc nitrate solution was filtered through a 0.8 μm filter to remove insoluble impurities in the starting material.
And B, liquid B: to 7mL of Dimethylsulfoxide (DMSO), exactly weighed 0.5g of 2,2' -dithiodipyridine (concentration: 0.7. mu.g/mL) was added, and 0.5g of 4-dimethylaminopyridine was added and sufficiently dissolved, and then the solution was filtered with a 0.8 μm filter to remove insoluble impurities from the starting material. Then adding 80mg of polyvinylpyrrolidone accurately weighed into the dissolved solution after the membrane is passed through, and fully and uniformly stirring;
optimization of different raw material proportions:
the first scheme is as follows: and (3) quickly adding 7mL of the solution B into 3.5mL of the solution A (the mixing volume ratio of the solution A to the solution B is 1:2), stirring and reacting for 6 hours at 60 ℃, finally centrifuging and washing the reaction solution for multiple times by using ultrapure water (12000rpm, 10min, and repeatedly washing for at least three times) to fully remove the organic solvent, and finally obtaining the MOF nanoparticles, wherein a TEM transmission electron microscope image of the MOF nanoparticles is shown in FIG. 1(A), and the particles are poor in dispersity and uneven in shape at the moment.
Scheme II: referring to the first scheme, the difference is that the mixing volume ratio of the solution A to the solution B is adjusted to 1:1, that is, 7mL of the solution B is rapidly added into 7mL of the solution A, and other steps are completely the same, so that the MOF nanoparticles are finally obtained, a TEM transmission electron microscope image of the MOF nanoparticles is shown as a picture in FIG. 1(B), and the MOF nanoparticles have good dispersibility, uniform shape and size smaller than 200nm and meet the drug loading requirement. Therefore, the mixing volume ratio of the solution A to the solution B is optimized to be 1: 1.
Carrying out drug loading performance evaluation on the MOF nano particles prepared by the scheme II:
average particle size of MOF nanoparticles prepared in protocol two over 15 days using Dynamic Light Scattering (DLS) [ FIG. 1(C)]Particle dispersity [ FIG. 1(D) ]]The measurement is performed. The test result proves that the particle size of the MOF nano particles prepared by the scheme II is less than 200nm, the particle size is uniform, the dispersity is excellent, and the drug loading requirement is met; measurement protocol two disintegration of MOF nanoparticles in Glutathione (GSH) at different concentrations [ FIG. 1(E)]The result proves that the MOF nano particles prepared by the scheme II can be disintegrated in glutathione, and the design idea is met; x-ray diffraction (XRD) proves that the MOF nano-particles have a certain framework structure (figure (1F)](ii) a The porous structure of the MOF nano-particles is analyzed by using BET surface area [ figure 1(G),1(H)), which proves that the MOF nano-particles are porous structures and the specific surface area reaches 557cm2Per g, pore volume up to 0.18cm3/g。
Example 2:
the preparation method of the Dox @ MOF nanoparticles comprises the following steps:
and mixing 400 mu L of MOF nanoparticle dispersion liquid (4mg/mL) prepared in the second scheme of the example 1 with a proper amount of Dox solution (2mg/mL), and stirring for 24h at 25 ℃ in a dark place to obtain a mixed system with the final concentration of MOF of 200 mu g/mL and the final concentration of Dox of 1, 3 and 5 mu g/mL, namely Dox @ MOF nanoparticles.
Example 3:
the preparation method of the Dox/Antisense @ MOF nano-particles comprises the following steps:
160 mu.L of Antisense solution (10mM) is added into the Dox @ MOF nanoparticle dispersion liquid prepared in example 2, the mixture is mixed by vortex and is kept stand for 2h at room temperature so as to facilitate the adsorption of the Antisense, and a mixed system with the final concentration of MOF of 200 mu g/mL, the final concentration of the Antisense of 200nM and the final concentration of Dox of 1, 3 and 5 mu g/mL is obtained, and the mixture is fully reacted to obtain Dox/Antisense @ MOF nanoparticles.
Example 4:
the preparation method of the HA/Dox/Antisense/@ MOF nano-particle comprises the following steps:
200 mu L of hyaluronic acid solution (4mg/mL) is added into the Dox/Antisense @ MOF nanoparticle dispersion liquid prepared in example 3 to obtain a mixed system with the final MOF concentration of 200 mu g/mL, the final Antisense concentration of 200nM, the final Dox concentration of 1, 3, 5 mu g/mL and the final HA concentration of 200 mu g/mL, and the mixed system is fully reacted to obtain HA/Dox/Antisense @ MOF nanoparticles.
Example 5: exploration of usage amount of MOF nanoparticles
The in vitro cytotoxicity of the MOF nanoparticles of scheme two of example 1 on MCF-7/ADM cells was investigated by the MTT (thiazole blue) method.
Firstly, MCF-7/ADM cells are added at 1 × 104Cell/well Density was plated in 96-well plates and incubated for 36h (37 ℃, 5% CO)2). Then, the medium in the 96-well plate was removed with a pipette gun, and MOF nanoparticle dispersions (140. mu.g/mL, 160. mu.g/mL, 180. mu.g/mL, 200. mu.g/mL, 220. mu.g/mL) diluted with medium were added to the well plates, three replicates per concentration were set, and incubation was continued for 24 h. After incubation was complete, the MOF nanoparticle dispersion was removed and 100. mu.L of fresh medium and 10. mu.L of MTT solution (5mg/mL) were added and incubation continued for 4 h. Subsequently, 100. mu.L of MTT Buffer (triple lysis) was added and the plate was read overnight. The Optical Density (OD) at 570nm of each well was recorded on a microplate reader). Each experiment was repeated three times. The cell viability calculation formula is as follows: cell viability (OD)Experiment of/ODControl
As shown in FIG. 7, the MTT method was used to measure the inhibition rate of the MOF nanoparticles on the cells at different concentrations, taking MCF-7/ADM cells as an example. It can be seen that the survival rate of MCF-7/ADM cells can still reach 80% when the concentration of the MOF nano particles is 200 mug/mL. Therefore, when the concentration of the MOF nanoparticles is lower than 200 μ g/mL, the range is reasonable, and the concentration of the MOF nanoparticles is selected to be 200 μ g/mL for the following experiment by taking the carrying capacity of the subsequent experiment drug into consideration.
Example 6: MOF-Loading Activity assay
After determining the most suitable concentration of MOF nanoparticles entering the cells, in order to conveniently study the total amount of the antibodies capable of being loaded by the MOF nanoparticles at the concentration, the 5' -end of the antibodies is modified with a fluorescent group Cy5, different volumes of the solutions of the antibodies with the concentration of 10 μ M are mixed with a proper amount of MOF nanoparticles, and the volume of the system is supplemented to 200mL by TE (Tris-EDTAbuffer) buffer solution, so that the final concentration of the MOF nanoparticles in the final mixed system is 200 μ g/mL, and the final concentrations of the antibodies are respectively 160, 180, 200, 220 and 240 nM. After vortex mixing, the mixture reacts for 30min, and the fluorescence at 625nm is detected by a microplate reader.
As shown in FIG. 8, the fluorescence value increased after the concentration of Antisense exceeded 200nM, demonstrating that the Antisense was already supersaturated at this time, and thus the loading of Antisense was determined to be 200 nM.
Example 7: determination of the Release Capacity of Dox in Dox/Antisense @ MOF nanoparticles under different conditions
Release of Dox in PBS buffer containing GSH or at different pH was investigated by dialysis.
Dox/Antisense @ MOF nanoparticles prepared in example 3 (wherein final MOF concentration was 200 μ g/mL, final Antisense concentration was 200nM, and final Dox concentration was 3 μ g/mL) were dispersed in 1mL of ultrapure water, transferred to a dialysis bag (molecular cut-off of 3.5kDa), subjected to dialysis experiments by immersing them in PBS buffer (30mL) having pH 7.4, pH 5.6, and pH 5.6, respectively, and containing 10mM GSH, and then placed on a shaker 100r/min, slowly shaken at 37 ℃ to release Dox. Samples were taken at the set time points (0, 0.5, 1, 2, 4, 6, 8, 10, 12, 24, 36, 48, 72h), and 3mL of dialysate was withdrawn each time and supplemented with 3mL of fresh PBS buffer. After sampling, the fluorescence intensity at 570nm of the sample was measured, and the concentration was calculated from the Dox standard curve. Each experiment was repeated three times. The cumulative Dox release Er is calculated by the following formula:
Figure BDA0003384176380000071
wherein, V0Total volume of PBS, FnThe fluorescence intensity at the time of the nth sampling, VeVolume per sample, FtotalThe total fluorescence value of the added nucleic acid was used.
As shown in fig. 9, Dox/Antisense @ MOF nanoparticles of the present invention released slowly in PBS buffer at pH 7.4, and the cumulative release rate was only 23%; in PBS buffer solution with pH value of 5.6, Dox release is accelerated, and the cumulative release rate reaches 60%; under the condition that the pH value is 5.6 and 10mM glutathione exists, Dox is rapidly released in the first 10h, and the Dox/Antisense @ MOF nano-particles can rapidly decompose and release the drug under the condition, and after 72h, the accumulated release rate of Dox reaches 78%. Thus, it can be proved that the MOF nanoparticles prepared by the invention can effectively deliver drugs and realize the release of the drugs (such as Dox) in the microenvironment with weak acidity and high glutathione in tumor cells.
Example 8: cytotoxicity determination of Dox/Antisense @ MOF nanoparticles
Group MOF: the final concentration of MOF is 200 mug/mL;
group DOX: the final concentration of Dox is 1, 3, 5 mu g/mL;
dox @ MOF (MOF + Dox) group: the final MOF concentration is 200 mug/mL, and the final Dox concentration is 1, 3, 5 mug/mL;
Dox/Antisense @ MOF (MOF + Dox + Antisense) group: the final MOF concentration is 200 mug/mL, the final Antisense concentration is 200nM, and the final Dox concentration is 1, 3, 5 mug/mL;
Dox/Antisense @ MOF + Laser (MOF + Dox + Antisense + Laser) group: the final MOF concentration is 200 mug/mL, the final Antisense concentration is 200nM, and the final Dox concentration is 1, 3, 5 mug/mL;
HA/Dox/Antisense @ MOF + Laser (MOF + Dox + Antisense + HA + Laser) group: the final MOF concentration is 200. mu.g/mL, the final HA concentration is 200. mu.g/mL, the final Antisense concentration is 200nM, and the final Dox concentration is 1, 3, 5. mu.g/mL.
(1) MTT test:
the in vitro cytotoxicity of Dox/Antisense @ MOF nanoparticles on MCF-7/ADM cells was studied by MTT assay. Firstly, MCF-7/ADM cells are added at 1 × 104The density of individual cells/well was plated in 96-well plates and cultured for 36h (37 ℃ C., 5% CO)2) Then, each experimental group was added.
After 24h dosing, the drug was removed and washed with PBS, then 100 μ L of fresh medium was added and the illuminants were irradiated with a 660nm laser, with the conditions per well: 660nm, 5min, 0.75w/cm2And irradiating for 5min at intervals of 12h under the same conditions, culturing for 12h after irradiation is finished, adding the MTT solution, adding the MTT Buffer after 4h, and reading the plate overnight. The Optical density value (OD) at 570nm of each well was recorded on a microplate reader. Each experiment was repeated three times.
The cell viability calculation formula is as follows: cell viability (OD)Experiment of/ODControl
In order to prove the drug resistance effect of the Dox/Antisense @ MOF nanoparticles, the IC of MCF-7/ADM cells before and after Dox loading in a nano system is determined50
As shown in FIG. 10, MCF-7/ADM cytotoxicity in different treatment groups all showed dose-dependence on Dox. The Dox/Antisense @ MOF (MOF + Dox + Antisense) group showed stronger cytotoxicity than the Dox @ MOF (MOF + DOX) group even in the absence of irradiation, demonstrating that Antisense can be loaded into cells by MOF nanoparticles and improve drug resistance. The cytotoxicity of MCF-7 was further enhanced by the group Dox/Antisense @ MOF (MOF + DOX + Antisense) following combined PDT photodynamic therapy (laser irradiation), demonstrating that Ce6 is able to efficiently generate ROS and increase cytotoxicity towards MCF-7. After the coating is carried out by Hyaluronic Acid (HA) (HA/Dox/Antisense @ MOF + Laser (MOF + Dox + Antisense + HA + Laser)), the nano system HAs certain targeting property, can be better taken up by cells, and improves cytotoxicity.
IC with Dox alone on MCF-7/ADM cells50[ FIG. 11(B)]In contrast, IC of Dox on MCF-7/ADM cells after Loading of Dox on nanosystems (Dox/Antisense/HA @ MOF)50[ FIG. 11(A)]The dosage of the chemotherapeutic drug is reduced by greatly reducing the dosage of the Dox/Antisense/HA @ MOF nano system.
(2) Determination of live and dead cells:
firstly, MCF-7/ADM cells are added at 2X 104The density of individual cells/well was seeded in 6-well plates and then cultured continuously for 36h (37 ℃, 5% CO)2). Thereafter, Dox (concentration 3. mu.g/mL), Dox/Antisense @ MOF nanoparticles (final concentration of MOF 200. mu.g/mL, final concentration of Antisense 200nM, final concentration of Dox 3. mu.g/mL) were added to the well plate. After 24h dosing, the drug was removed and washed with PBS, then 100 μ L fresh medium was added and the 6-well plate was irradiated with a 660nm laser under the following conditions per well: 660nm, 5min, 0.75w/cm2. After the irradiation, the cells were incubated for another 12 hours and washed gently with PBS 2 to 3 times to ensure removal of the active esterase contained in the medium. Sufficient staining solution (2. mu.M calcein AM and 8. mu.M PI) was added to ensure that a monolayer of cells was submerged. Incubate at 37 ℃ for 15-30 minutes, observe live cells simultaneously under fluorescence microscope using 490nm excitation wavelength (yellow green fluorescence), and separately observe dead cells using 545nm excitation wavelength (red fluorescence).
As shown in FIG. 12, the Dox/Antisense @ MOF nanoparticles (labeled as Dox @ MOF/Antisense in the figure) had the least viable cells and the most dead cells, as compared to the non-dosed control group and the Dox group, and were consistent with the MTT assay.
(3) Apoptosis assay:
firstly, MCF-7/ADM cells are added at 2X 104The density of individual cells/well was seeded in 6-well plates and then cultured continuously for 36h (37 ℃, 5% CO)2). Thereafter, the illuminated group of Dox, Dox @ MOF, Dox/Antisense @ MOF (final MOF concentration of 200. mu.g @)mL, final Antisense concentration 200nM, final Dox concentration 3 μ g/mL) was added to the well plate. After 24h dosing, the drug was removed and washed with PBS, then 100 μ L fresh medium was added and the 6-well plate was irradiated with a 660nm laser under the following conditions per well: 660nm, 5min, 0.75w/cm2. After the irradiation, the cells were cultured for 12 hours again, washed gently with PBS for 2 times, digested with trypsin without EDTA, added with the cell culture medium, gently blown down, transferred into a centrifuge tube, and centrifuged to collect the cells. After the cells were collected, a precooled PBS solution was added, blown and washed, and the cells were collected by centrifugation again for two washes. Add 1 × Binding buffer working solution to the cell pellet, resuspend the cells to a cell concentration of 1 × 106 cells/mL. Then, 100. mu.L of the cell suspension (total number of cells: 1X 10) was aspirated5cell) into a new tube, add 5 μ L Annexin V-FITC and 10 μ L PI, mix gently, incubate for 15min at room temperature in the dark. After staining incubation, 400. mu.L of 1 XBinding Buffer working solution was added to each tube, mixed well and detected using a flow cytometer. And drawing a double-dispersion dot diagram, wherein FITC is an abscissa, and PI is an ordinate. When Annexin V-FITC and PI are used in combination, live cells only have very low-intensity background fluorescence, early apoptotic cells only have strong green fluorescence, and late apoptotic cells have dual fluorescence of green and red.
As shown in FIG. 13, the late apoptotic cells of the Dox/Antisense @ MOF group were more abundant than those of the Dox @ MOF group, demonstrating the therapeutic effect of the Antisense. The group of Dox/Antisense @ MOF exhibited the greatest number of late apoptotic cells following combination PDT treatment (+ Laser), demonstrating that Ce6 is able to efficiently produce ROS and increase cytotoxicity toward MCF-7/ADM.
Example 9: detection of ROS generated by Ce6 under specific wavelength illumination stimulation
First, 1mL of 1X 10 dense medium was added to a 24-well plate4And (4) putting the MCF-7/ADM cell sap of each/mL into an incubator to incubate for 48 hours. Serum-free DMEM dispersed Antisense @ MOF (0, 80, 150, 200nM) was then added and incubation continued for 12 h. The ability to produce ROS was then tested using an ROS detection kit, as detailed below: washing residual medicine and culture medium in the well plate with PBS, placing on a 200r/min shaking table, washing for 3 times, 10min each time, and keeping away from light. Subsequent application of avascular bloodDiluting DCFH-DA with clear DMEM to reach concentration of 10 μ M, incubating with cells in incubator for 30min, washing with serum-free culture medium in dark place, washing on 200r/min shaking table for 3 times (10 min each time), irradiating with light of specific wavelength to obtain 24-well plate with irradiation parameters of 660nm, 5min and 0.75w/cm2The ROS production was then observed using the FITC channel of a fluorescence microscope.
As shown in fig. 14-15, the MOF nanoparticles prepared in the second embodiment of example 1 successfully load and release Antisense into cells, and generate a large amount of ROS under the irradiation of a specific wavelength, and DCF generated in the ROS kit is combined to show strong green fluorescence and has concentration dependence. Dox/Antisense @ MOF nanoparticles were demonstrated to have the ability to generate ROS, consistent with the expectations and objectives of the design of the present invention.
Example 10: MOF nanoparticle-depleted intracellular glutathione assay of the invention
(1) Example 1 study of the inhibitory effect of MOF nanoparticles of scheme two on BCL-2 protein:
the ability of MOF nanoparticles of scheme two of example 1 to consume intracellular glutathione was tested by glutathione assay kit (Nanjing institute).
First, 1mL of a 2X 10 dense solution was added to a 6-well plate4MCF-7/ADM cell fluid of each/mL is put into an incubator until the cell fluid is fully attached to the wall, the MOF nano particle dispersion liquid of the second scheme of the embodiment 1 (the final concentration of MOF is 200 mug/mL) dispersed by serum-free DMEM is added, and after 2, 4 and 6 hours of incubation, the drug is removed and washed by PBS. Then, 100. mu.L of RIPA (containing 1 XPSF and EDTA) was added thereto and the mixture was lysed on ice for 5 min. After the lysis is completed, the bottom of the pore plate is carefully scraped by a cell scraper, and the lysate is collected and taken out of the cell room. Cells were disrupted with an ultrasonic cell disruptor. After disruption, centrifugation (4 ℃, 12000rpm, 20 min). After centrifugation, the supernatant was removed and the protein concentration of the sample was determined using the BCA kit. Taking 0.1mL of the crushed cell suspension, adding 0.1mL of the reagent I, mixing uniformly, 3500r/min, centrifuging for 10 minutes, and taking the supernatant to be tested. mu.L of the supernatant, 100. mu.L of reagent two, and 25. mu.L of reagent three were added to the assay well. Add 100. mu.L reagent one, 100. mu.L reagent two, and 25. mu.L reagent three to the blank wells. Standard hole100 μ L of 20 μmol/L GSH standard, 100 μ L of reagent two, and 25 μ L of reagent three are added. Standing for 5 minutes, and measuring the absorbance value of each hole at 405nm by using a microplate reader.
Figure BDA0003384176380000101
As shown in FIG. 16, the content of glutathione GSH decreased with the increase of time after the addition of the MOF nanoparticles of the second embodiment of example 1, which proves that the MOF nanoparticles of the present invention have the ability to consume glutathione in cells, and thus, it can be concluded that both the Dox/Antisense @ MOF nanoparticles and the Dox/Antisense/HA MOF @ nanoparticles of the present invention have the ability to consume glutathione in cells.
Example 11: study on uptake condition of Dox/Antisense @ MOF nanoparticles by MCF-7/ADM cells
The distribution of intracellular drugs and nucleic acids at different emission wavelengths is photographed by a laser confocal microscope. 1mL of culture dish with density of 1 multiplied by 10 is added into a special culture dish for laser confocal5MCF-7/ADM cells per mL, and incubating in the incubator until the cells are fully adherent. Media discard Dox (3. mu.g/mL)/Antisense (200nM) @ MOF (200. mu.g/mL) was co-incubated with MCF-7/ADM cells in serum-free DMEM media for 1, 3, 6, 12h, respectively. After completion, the drug was removed, and the plate was washed five times with 10min each time in a shaker at 200r/min with 1mL of PBS having a pH of 7.4, to sufficiently remove the drug remaining in the well plate, and then 1mL of 4% paraformaldehyde solution was added to fix the cells. After 15min, 1mL PBS was added and washed five times in a shaker for 10min each to remove residual paraformaldehyde. Subsequently 500 μ L DAPI staining solution was added and after 10min the staining of the nuclei was completed and washed five times with 1mL PBS on a shaker for 10min each to remove the remaining DAPI staining solution. After washing, 1mL of PBS was added to each dish and stored in a refrigerator at 4 ℃ in the dark. The phagocytosis effect of MCF-7 cells on a drug-loading system and the distribution of intracellular drugs and nucleic acid under different emission wavelengths are observed by using a laser scanning confocal microscope (CLSM).
As shown in FIG. 17, after the Dox/Antisense @ MOF nanoparticles are incubated with MCF-7/ADM cells for 1, 3, 6 and 12 hours, fluorescence of Dox and Cy5-Antisense in the cells is successfully captured by a laser confocal microscope, which proves that the Dox/Antisense @ MOF nanoparticles can be rapidly taken up by tumor cells and successfully enter the cells, and therefore, the feasibility of the Dox/Antisense @ MOF nanoparticles in treating breast cancer is proved.
Example 12: research on inhibition effect of Dox/Antisense @ MOF nanoparticles on related proteins
(1) Study of the inhibitory effect of Dox/Antisense @ MOF nanoparticles on BCL-2 protein:
the inhibition capability of the Dox/Antisense @ MOF nanoparticles on the BCL-2 protein is researched through a Western Blot experiment. First, 1mL of a 2X 10 dense solution was added to a 6-well plate4MCF-7/ADM cell fluid of each/mL is put into an incubator until the cell fluid is fully attached, serum-free DMEM dispersed Dox/Antisense @ MOF (MOF final concentration is 200 mug/mL, and Dox final concentration is 3 mug/mL) dispersion liquid loaded with different concentrations of Antisense (50, 100, 200nM) is added, the drug is removed after continuous incubation for 6 hours, PBS is used for washing, then fresh culture medium is added, and a laser with 660nM is used for irradiating 6-well plates, wherein the irradiation condition of each well is as follows: 660nm, 5min, 0.75w/cm2. After the irradiation was completed, the culture was further incubated for 4 hours, the medium was discarded and washed with PBS, and 100. mu.L of RIPA (containing 1 XPSF and EDTA) was added thereto and lysed on ice for 5 min. After the lysis is completed, the bottom of the pore plate is carefully scraped by a cell scraper, and the lysate is collected and taken out of the cell room. Cells were disrupted with an ultrasonic cell disruptor. After disruption, centrifugation (4 ℃, 12000rpm, 20 min). After centrifugation, the supernatant was removed, and the protein concentration of the sample was determined using the BCA kit, and the sample was adjusted to the same protein concentration with PBS. To the adjusted sample was added 1/4 volumes of 5 × Loading Buffer followed by a 5min boiling water bath. And (3) cooling the sample, then loading the sample, wherein the loading volume is 10 mu L, adding 5 mu L Marker into other holes, carrying out electrophoresis by using 5% concentrated gel on the upper layer and 110V and 1h according to a mode of 80V and 30min and 10% separation gel on the lower layer. And after the film is finished, the PVDF film with proper size is cut, the black glue film and the white film are sequentially filled into a mold, and the film rotating parameters are set to be 110V and 60 min. After the membrane transfer is finished, the membrane is sealed for 2h by using 5% skimmed milk, and then the membrane is washed 3 times by using 1 XTSST10min each time. After the completion of the washing, the primary antibody of BCL-2 protein was incubated overnight at 4 ℃. The next day, the primary antibody is recovered, and the membrane is cleaned for 3 times; after completion, the secondary antibody was incubated for 2h, and the membrane was washed 3 times. And opening the gel imager in advance for cooling, uniformly covering with an ECL reagent during development, and carrying out exposure and photographing.
As shown in FIGS. 18(A) and 18(B), BCL-2 is an anti-apoptotic protein and plays an important role in a drug resistance mechanism, the apoptosis effect of the Dox/Antisense @ MOF nanoparticle combined PDT treatment of the invention can inhibit the expression of BCL-2, and the expression amount of the protein is reduced along with the increase of the Antisense, so that a dose-dependent rule is presented, and the result is matched with the result of a cytotoxicity experiment, thereby conforming to the assumption of a gene therapy system of the invention.
(2) Research on the inhibition effect of Dox/Antisense @ MOF nanoparticles on P-gp protein:
the inhibition capability of the Dox/Antisense @ MOF nano ions on P-gp protein is researched through a Western Blot experiment. First, 1mL of a 2X 10 dense solution was added to a 6-well plate4MCF-7/ADM cell fluid of each/mL is put into an incubator until the cell fluid is fully attached, serum-free DMEM dispersed Dox/Antisense @ MOF (final MOF concentration is 200 mug/mL, final Dox concentration is 3 mug/mL) dispersion liquid loaded with different concentrations of Antisense (50, 100, 200nM) is added, the incubation is continued for 10 hours, the culture medium is discarded and washed by PBS, 100 mug RIPA (containing 1 XPSF and EDTA) is added into the dispersion liquid, and the mixture is placed on ice for lysis for 5 minutes. After the lysis is completed, the bottom of the pore plate is carefully scraped by a cell scraper, and the lysate is collected and taken out of the cell room. Cells were disrupted with an ultrasonic cell disruptor. After disruption, centrifugation (4 ℃, 12000rpm, 20 min). After centrifugation, the supernatant was removed, and the protein concentration of the sample was determined using the BCA kit, and the sample was adjusted to the same protein concentration with PBS. To the adjusted sample was added 1/4 volumes of 5 × Loading Buffer followed by a 5min boiling water bath. And (3) cooling the sample, then loading the sample, wherein the loading volume is 10 mu L, adding 5 mu L Marker into other holes, carrying out electrophoresis by using 5% concentrated gel on the upper layer and 110V and 1h according to a mode of 80V and 30min and 10% separation gel on the lower layer. And after the film is finished, the PVDF film with proper size is cut, the black glue film and the white film are sequentially filled into a mold, and the film rotating parameters are set to be 110V and 60 min. After the film transfer is finished, degreasing with 5 percentThe milk was blocked for 2h, then the membranes were washed 3 times with 1 × TBST for 10min each. After the washing, the primary antibody of the P-gp protein is incubated, and the temperature is 4 ℃ overnight. The next day, the primary antibody is recovered, and the membrane is cleaned for 3 times; after completion, the secondary antibody was incubated for 2h, and the membrane was washed 3 times. And opening the gel imager in advance for cooling, uniformly covering with an ECL reagent during development, and carrying out exposure and photographing.
As shown in FIGS. 18(C) and 18(D), since P-gp plays an important role in the resistance mechanism, HIF-1 α affects the expression of P-gp, i.e., when HIF-1 α is inhibited, P-gp is also inhibited. Therefore, the expression of P-gp can be further inhibited after HIF-1 alpha is inhibited by Dox/Antisense @ MOF nano particles, the expression amount of protein is reduced along with the increase of Antisense, a dose-dependent rule is presented, and the method is matched with the result of a cytotoxicity experiment and accords with the assumption of a gene therapy system.
(3) Research on the inhibitory effect of Dox/Antisense @ MOF nanoparticles on Gpx4 protein:
the inhibition capacity of Dox/Antisense @ MOF nanoparticles on Gpx4 protein was studied by Western Blot experiment. First, 1mL of a 2X 10 dense solution was added to a 6-well plate4MCF-7/ADM cell fluid of each/mL is put into an incubator until the cell fluid is fully attached, serum-free DMEM dispersed Dox/Antisense @ MOF (final MOF concentration is 200 mug/mL, final Dox concentration is 3 mug/mL) dispersion liquid loaded with different concentrations of Antisense (50, 100, 200nM) is added, the incubation is continued for 10 hours, the culture medium is discarded and washed by PBS, 100 mug RIPA (containing 1 XPSF and EDTA) is added into the dispersion liquid, and the mixture is placed on ice for lysis for 5 minutes. After the lysis is completed, the bottom of the pore plate is carefully scraped by a cell scraper, and the lysate is collected and taken out of the cell room. Cells were disrupted with an ultrasonic cell disruptor. After disruption, centrifugation (4 ℃, 12000rpm, 20 min). After centrifugation, the supernatant was removed, and the protein concentration of the sample was determined using the BCA kit, and the sample was adjusted to the same protein concentration with PBS. To the adjusted sample was added 1/4 volumes of 5 × Loading Buffer followed by a 5min boiling water bath. And (3) cooling the sample, then loading the sample, wherein the loading volume is 10 mu L, adding 5 mu L Marker into other holes, carrying out electrophoresis by using 5% concentrated gel on the upper layer and 110V and 1h according to a mode of 80V and 30min and 10% separation gel on the lower layer. After the end of the film transfer, cutting the PVDF film with proper size according to the black glueSequentially filling the white films into a mold, and setting film transfer parameters to be 110V and 60 min. After the membrane transfer was completed, the membrane was blocked with 5% skim milk for 2h, and then washed 3 times with 1 × TBST for 10min each. After washing, the primary antibody of Gpx4 protein was incubated, and the temperature was 4 ℃ overnight. The next day, the primary antibody is recovered, and the membrane is cleaned for 3 times; after completion, the secondary antibody was incubated for 2h, and the membrane was washed 3 times. And opening the gel imager in advance for cooling, uniformly covering with an ECL reagent during development, and carrying out exposure and photographing.
As shown in fig. 18(E) and 18(F), Gpx4 is a key regulatory factor for iron death, and once Gpx4 is inhibited, iron death is caused, drug sensitivity is increased, and drug resistance is improved. After MOF is added, the expression level of Gpx4 is obviously reduced, and after the Antisense with different concentrations is added, the expression level of protein is further reduced, a dose-dependent rule is presented, and the expression level is matched with the result of a cytotoxicity experiment, thereby conforming to the assumption of the gene therapy system of the invention.

Claims (10)

1. A preparation method of an MOF drug carrier is characterized by comprising the following steps:
preparation of solution A: dissolving zinc nitrate in N, N-dimethylformamide, and removing insoluble impurities to obtain solution A;
and (3) preparation of a solution B: dissolving 2,2' -dipyridyl disulfide and 4-dimethylaminopyridine in dimethyl sulfoxide, removing insoluble impurities, adding polyvinylpyrrolidone, and mixing to obtain solution B;
and (3) quickly adding the solution B into the solution A, heating and stirring for reaction, centrifuging the obtained reaction solution, taking precipitate, and washing with ultrapure water to obtain MOF nano particles, namely the MOF drug carrier.
2. The method according to claim 1, wherein the mass ratio of zinc nitrate to 2,2' -dithiodipyridine in the mixed system of solution A and solution B is (1.1-1.3): 1.
3. The method according to claim 1, wherein the temperature for the heating and stirring reaction is 50 to 80 ℃.
4. MOF nanoparticles produced by the method of any one of claims 1 to 3.
5. Use of the MOF nanoparticles of claim 4 as a drug carrier.
6. A drug carrier system based on MOF nanoparticles is characterized in that the drug carrier system is prepared by fully mixing the components including the MOF nanoparticles, HIF-1 alpha antisense nucleic acid modified with photosensitizer and adriamycin in a solvent.
7. The drug delivery system of claim 6, further comprising hyaluronic acid.
8. The drug carrier of claim 6 or 7, wherein the final concentration of the MOF nanoparticles is 200 μ g/mL.
9. The drug delivery system of claim 8, wherein the photosensitizer-modified HIF-1 α antisense nucleic acid has a final concentration of 200nM and doxorubicin has a final concentration of 0.2-5 μ g/mL.
10. The use of a drug delivery system according to any one of claims 6 to 9 in the preparation of an anti-breast cancer drug.
CN202111442947.4A 2021-11-30 2021-11-30 MOF drug carrier for treating breast cancer and preparation method thereof Pending CN114099706A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111442947.4A CN114099706A (en) 2021-11-30 2021-11-30 MOF drug carrier for treating breast cancer and preparation method thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111442947.4A CN114099706A (en) 2021-11-30 2021-11-30 MOF drug carrier for treating breast cancer and preparation method thereof

Publications (1)

Publication Number Publication Date
CN114099706A true CN114099706A (en) 2022-03-01

Family

ID=80368804

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111442947.4A Pending CN114099706A (en) 2021-11-30 2021-11-30 MOF drug carrier for treating breast cancer and preparation method thereof

Country Status (1)

Country Link
CN (1) CN114099706A (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105873569A (en) * 2013-11-06 2016-08-17 芝加哥大学 Nanoscale carriers for the delivery or co-delivery of chemotherapeutics, nucleic acids and photosensitizers
CN108619526A (en) * 2018-05-21 2018-10-09 中国医学科学院生物医学工程研究所 The sensitive preparation method for carrying chemotherapeutics and the nano-delivery system of P-gp reversal agent of drug resistance altogether of targeting reduction
CN111643673A (en) * 2020-07-08 2020-09-11 福建医科大学孟超肝胆医院(福州市传染病医院) Tumor-targeted nano-drug simultaneously encapsulating photosensitizer and protein and application thereof
US20210000971A1 (en) * 2017-11-28 2021-01-07 Unist(Ulsan National Institute Of Science And Technology) Fusion protein comprising glutathione-s-transferase and protein having binding affinity target cell or target protein, and use thereof
CN112773899A (en) * 2019-11-04 2021-05-11 天津大学 Drug delivery carrier based on biological metal organic framework material and preparation method and application thereof
CN113583244A (en) * 2021-06-17 2021-11-02 浙江大学 Metal organic framework material and preparation method and application thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105873569A (en) * 2013-11-06 2016-08-17 芝加哥大学 Nanoscale carriers for the delivery or co-delivery of chemotherapeutics, nucleic acids and photosensitizers
US20210000971A1 (en) * 2017-11-28 2021-01-07 Unist(Ulsan National Institute Of Science And Technology) Fusion protein comprising glutathione-s-transferase and protein having binding affinity target cell or target protein, and use thereof
CN108619526A (en) * 2018-05-21 2018-10-09 中国医学科学院生物医学工程研究所 The sensitive preparation method for carrying chemotherapeutics and the nano-delivery system of P-gp reversal agent of drug resistance altogether of targeting reduction
CN112773899A (en) * 2019-11-04 2021-05-11 天津大学 Drug delivery carrier based on biological metal organic framework material and preparation method and application thereof
CN111643673A (en) * 2020-07-08 2020-09-11 福建医科大学孟超肝胆医院(福州市传染病医院) Tumor-targeted nano-drug simultaneously encapsulating photosensitizer and protein and application thereof
CN113583244A (en) * 2021-06-17 2021-11-02 浙江大学 Metal organic framework material and preparation method and application thereof

Similar Documents

Publication Publication Date Title
Liang et al. A protein@ metal–organic framework nanocomposite for pH-triggered anticancer drug delivery
He et al. Enhanced up/down-conversion luminescence and heat: simultaneously achieving in one single core-shell structure for multimodal imaging guided therapy
Gonzales et al. Light-triggered carbon monoxide delivery with Al-MCM-41-based nanoparticles bearing a designed manganese carbonyl complex
Liu et al. A multifunctional nanoplatform based on mesoporous silica nanoparticles for imaging-guided chemo/photodynamic synergetic therapy
Dong et al. Ultrasmall biomolecule-anchored hybrid GdVO 4 nanophosphors as a metabolizable multimodal bioimaging contrast agent
CN108114290A (en) Preparation method that is a kind of while loading chemicals and the excretion body of nano material
Liu et al. Graphitic carbon nitride nanosheets as a multifunctional nanoplatform for photochemical internalization-enhanced photodynamic therapy
Kang et al. Boron-based nanosheets for combined cancer photothermal and photodynamic therapy
Wu et al. Methotrexate–Mn 2+ based nanoscale coordination polymers as a theranostic nanoplatform for MRI guided chemotherapy
CN102784397B (en) Method for loading doxorubicine (DOX) anti-cancer medicine by laponite (LAP) clay nanoparticles
Singh et al. Novel hybrid nanorod carriers of fluorescent hydroxyapatite shelled with mesoporous silica effective for drug delivery and cell imaging
Guan et al. Multifunctional Fe3O4@ SiO2-CDs magnetic fluorescent nanoparticles as effective carrier of gambogic acid for inhibiting VX2 tumor cells
Ren et al. Red emissive carbon dots prepared from polymers as an efficient nanocarrier for coptisine delivery in vivo and in vitro
Fan et al. AIE luminogen-functionalised mesoporous silica nanoparticles as nanotheranostic agents for imaging guided synergetic chemo-/photothermal therapy
CN103908676A (en) Folic-acid-modified laponite nanometer particle, preparation thereof and applications thereof
Atabaev et al. Highly mesoporous silica nanoparticles for potential drug delivery applications
Zhou et al. DNA-templated porous nanoplatform towards programmed “double-hit” cancer therapy via hyperthermia and immunogenicity activation
Kshirsagar et al. Synthesis of fluorescent metal nanoparticles in aqueous solution by photochemical reduction
Li et al. 5-FU@ DHA-UIO-66-NH2 potentiates chemotherapy sensitivity of breast cancer cells through a microRNA let-7a-dependent mechanism
CN114099706A (en) MOF drug carrier for treating breast cancer and preparation method thereof
CN104147608A (en) Lithium amide soapstone nano particles modified by polyethylene glycol-folic acid as well as preparation and application of lithium amide soapstone nano particles
Dong et al. Load and release of gambogic acid via dual-target ellipsoidal-Fe3O4@ SiO2@ mSiO2-C18@ dopamine hydrochloride-graphene quantum dots-folic acid and its inhibition to VX2 tumor cells
CN108159432B (en) Targeted nanoparticle for inhibiting breast cancer and preparation and application thereof
Zhang et al. Nanomicelles for GLUT1-targeting hepatocellular carcinoma therapy based on NADPH depletion
Zhong et al. In vivo study of a novel, safe, rapid, and targeted red carbon dot probe for recognition of tumors with high expression of folate enzyme

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