CN112791185A - Nano medicine for treating tumor by combining photothermal treatment with iron agent and preparation method thereof - Google Patents

Nano medicine for treating tumor by combining photothermal treatment with iron agent and preparation method thereof Download PDF

Info

Publication number
CN112791185A
CN112791185A CN202110073109.8A CN202110073109A CN112791185A CN 112791185 A CN112791185 A CN 112791185A CN 202110073109 A CN202110073109 A CN 202110073109A CN 112791185 A CN112791185 A CN 112791185A
Authority
CN
China
Prior art keywords
protein
metal
nano
drug
nanoparticle
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.)
Granted
Application number
CN202110073109.8A
Other languages
Chinese (zh)
Other versions
CN112791185B (en
Inventor
杨斌
余馨颖
韩世松
谢国喜
李雨微
蔡燕君
尚同祎
郑国栋
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.)
Guangzhou Medical University
Original Assignee
Guangzhou 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 Guangzhou Medical University filed Critical Guangzhou Medical University
Priority to CN202110073109.8A priority Critical patent/CN112791185B/en
Publication of CN112791185A publication Critical patent/CN112791185A/en
Application granted granted Critical
Publication of CN112791185B publication Critical patent/CN112791185B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0052Thermotherapy; Hyperthermia; Magnetic induction; Induction heating therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • 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
    • B82Y20/00Nanooptics, e.g. quantum optics or photonic crystals
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • 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

  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Health & Medical Sciences (AREA)
  • Nanotechnology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • General Physics & Mathematics (AREA)
  • Condensed Matter Physics & Semiconductors (AREA)
  • Molecular Biology (AREA)
  • Composite Materials (AREA)
  • Materials Engineering (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medical Informatics (AREA)
  • Manufacturing & Machinery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Optics & Photonics (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a nano-drug for treating tumor by combining photo-thermal and iron-based treatment and a preparation method thereof, wherein Fe is compounded on carrier protein of metal-protein nano-particles3+Ions while passing Fe3+Ion chelating pharmaceutically acceptableThe polyphenol compound of (1). The inventor finds that, through experiments, different from the traditional fact that the Fenton reagent generated in the tumor photothermal treatment process can cause the oxidative degradation failure of drug molecules or the uncontrollable drug effect, after the specific iron death drug is compounded with the metal-protein nanoparticle containing the polyphenol compound and used for tumor photothermal treatment, the two drugs can unexpectedly act in a synergistic manner, and have a better killing effect on tumor cells.

Description

Nano medicine for treating tumor by combining photothermal treatment with iron agent and preparation method thereof
Technical Field
The invention relates to a metal-protein nano-drug carrier, in particular to a metal-protein nano-drug for combined tumor photothermal therapy and iron death, and a preparation method and application thereof.
Background
Cancer is a serious disease which threatens human health worldwide nowadays, and the main means for treating cancer at present are chemotherapy, radiotherapy, surgical resection and the like, however, the methods are often accompanied by higher toxicity and recurrence rate and low targeting. In recent years, various novel biomedical nanomaterials have been applied to cancer therapy, including photothermal and photodynamic therapy materials, nano-drug carrier materials, and nanomaterials that induce iron death of tumor cells by using Fenton reaction (Fenton reaction) induced by the microenvironment of tumor cells. The photothermal therapy and the iron death combined tumor therapy are a novel effective method, and the defects of single photothermal therapy and iron therapy can be overcome. The key of the implementation of the technology is to construct a nano material with photo-thermal treatment and iron agent treatment, so that high iron loading capacity and low toxicity are realized while photo-thermal conversion efficiency is ensured, and the material can effectively carry therapeutic drugs, further induce the death of tumor cells, improve the treatment efficiency while the dosage is reduced, and realize the multi-means synergy.
The polyphenol compounds are also called flavonoid compounds, are compounds with a plurality of phenol groups, and have the functions of resisting oxidation, strengthening vessel walls and the like. The phenolic group of the polyphenol compound may be chelated with a metal ion to form a complex. However, the complex has the characteristics of uncontrollable appearance and size, poor biocompatibility and the like, and the application of the complex is limited. The metal-protein nano-particles are an effective strategy for constructing a stable multifunctional nano-biomaterial by utilizing the electrostatic interaction between proteins and the metal nano-particles and the good biocompatibility of the proteins.
Disclosure of Invention
The invention aims to overcome at least one defect of the prior art and provides a metal-protein nano-drug for combined tumor photothermal therapy and iron death and a preparation method and application thereof.
The technical scheme adopted by the invention is as follows:
in a first aspect of the present invention, there is provided:
a metal-protein nanoparticle for use in a medical device, comprising a carrier protein having Fe complexed thereon3+Ions while passing Fe3+Ion-chelating pharmaceutically acceptable polyphenolic compounds.
In some examples, the carrier protein is selected from bovine serum albumin, human serum albumin, ovalbumin.
In some examples, the polyphenolic compound is selected from Gallic Acid (GA), Tannic Acid (TA), Ellagic Acid (EA), epigallocatechin gallate (EGCG), baicalin (Baicalein), Myricetin (Myricetin), Delphinidin (Delphinidin), and the like.
In some examples, the Fe3+The molar ratio of the ions to the polyphenol compound is (17-67): (30-60).
In some examples, a therapeutic drug that enhances iron death is also loaded.
In some examples, the therapeutic drug is selected from at least one of sorafenib, elastine, FIN56, RSL3, lanpiriropine, sulfasalazine.
In some examples, the therapeutic drug is sorafenib, Fe3+: the molar ratio of the sorafenib is 1 (0.8-5).
In a second aspect of the present invention, there is provided:
use of a metal-protein nanoparticle for the manufacture of a medicament for photothermal therapy of a tumour in combination with iron death, said metal-protein nanoparticle being as described in the first aspect of the invention.
In some examples, the tumor is selected from the group consisting of models of breast cancer, colorectal cancer, liver cancer, pancreatic cancer, ovarian cancer, squamous cell carcinoma of the tongue, melanoma, fibrosarcoma, and the like.
In a third aspect of the present invention, there is provided:
a method for preparing metal-protein nanoparticles, comprising the steps of:
s1) fully stirring the carrier protein and the ferric salt solution to ensure that Fe3+Compounding the Fe-protein complex on carrier protein to obtain Fe-protein complex;
s2) adding a polyphenol compound into the obtained Fe-protein compound, stirring and standing to obtain a crude product of the metal-protein nano-particles;
s3) purifying the crude metal-protein nano-particles to obtain the finished metal-protein nano-particles.
In some examples, the ferric salt is selected from FeCl3、Fe2(SO4)3、Fe(NO3)3FeBr3 and Fe3O4And the like.
In some examples, a therapeutic agent that enhances iron death is added to the finished metal-protein nanoparticle product and reacted sufficiently to yield metal-protein nanoparticle particles for tumor photothermal therapy in combination with iron death.
The invention has the beneficial effects that:
the inventor finds that, through experiments, different from the traditional fact that the Fenton reagent generated in the tumor photothermal treatment process can cause the oxidative degradation failure of drug molecules or the uncontrollable drug effect, after the specific iron death drug is compounded with the metal-protein nanoparticle containing the polyphenol compound and used for tumor photothermal treatment, the two drugs can unexpectedly act in a synergistic manner, and have a better killing effect on tumor cells.
According to the metal-protein nanoparticles provided by some examples of the invention, the protein can play a role in improving the stability and biocompatibility of the nano material, and meanwhile, the metal-protein nanoparticles have a function of being widely combined with small molecules, so that a drug can be carried on the related nanoparticles to realize efficient drug delivery. The polyphenol compounds and iron ions can promote the Fenton reaction under the tumor microenvironment, and the photo-thermal property of the material and the drug-induced iron death effect are combined, so that the tumor is treated in a combined manner.
Metal-protein nanoparticles, Fe, of some embodiments of the invention3+The feed ratio of the protein and the polyphenol compound can be flexibly adjusted according to the use requirement.
The particle size of the metal-protein nanoparticles of some embodiments of the present invention can be controlled by selecting different polyphenolic compounds and proteins, or by varying the reaction time.
Some embodiments of the invention provide metal-protein nanoparticles as drugs and Fe3+When the feed ratio is changed, the treatment effect is changed, and the Fe is properly improved3+When the feed amount is reduced, the good treatment effect can be achieved when the medicine concentration is low, so that the toxic and side effects are reduced.
The metal-protein nanoparticles of the present invention can induce the death of tumor cells by using drugs, and compared with other antitumor drugs, the mode of synergistic drug administration and combined photo-thermal treatment has better tumor treatment effect.
Drawings
FIG. 1 is an atomic force microscope photograph of Fe-GA @ BSA nanoparticles prepared in example 1;
FIG. 2 shows the in vitro photothermal temperature profile of Fe-GA @ BSA nanoparticles prepared in example 1;
FIG. 3 shows the results of the catalytic efficiency of the in vitro Fenton reaction of Fe-GA @ BSA nanoparticles prepared in example 1;
FIG. 4 is an atomic force microscope photograph of Fe-GA @ BSA-SRF nanoparticles prepared in example 1;
FIG. 5 shows the changes of mitochondrial membrane potential of Fe-GA @ BSA and Fe-GA @ BSA-SRF prepared in example 1 in mouse breast cancer cell 4T1, which are observed by JC-1 detection kit through a fluorescence confocal microscope with a scale of 50 μm;
FIG. 6 is a graph of the effect of Fe-GA @ BSA and Fe-GA @ BSA-SRF prepared in example 1 on the induction of Reactive Oxygen Species (ROS) production by 4T1 in mouse breast cancer cells under different power light, as quantified by flow cytometry.
Detailed Description
In order that the invention may be more fully understood, reference will now be made to the following description. This invention may, however, be embodied in many different forms and should not be construed as limited to the embodiments set forth herein. Rather, these embodiments are provided so that this disclosure will be thorough and complete.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. The terminology used in the description of the invention herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used herein, the term "and/or" includes any and all combinations of one or more of the associated listed items.
Example 1
A preparation method of metal-protein nano-drug particles Fe-GA @ BSA-SRF comprises the following steps:
(1) weighing 66mg of BSA, dissolving in ultrapure water, and stirring at room temperature for 30 minutes;
(2) 54 μ L of FeCl3Dropwise adding the solution (100mg/mL) into the BSA solution under the condition of stirring at room temperature, and stirring at room temperature for 30 min;
(3) dropwise adding 1.02mL of gallic acid solution (10mg/mL) into the Fe-BSA mixed solution under the condition of stirring, and then standing for 3 hours at room temperature in a dark place;
(4) filtering the obtained solution containing the nano particles by using an ultrafiltration tube, and washing for 2 times by using ultrapure water to obtain purified Fe-GA @ BSA nano particles; the atomic force microscope result is shown in figure 1, and the figure shows that the nano particles have uniform size and the size is about 4 nm; the results of the in vitro photothermal temperature-rising curves of the obtained nanoparticles are shown in FIG. 2, and it can be seen from the graph that under 808nm near-infrared illumination, the temperature of nanoparticle solutions with different concentrations is increased compared with that of pure water, and the temperature rises along with illuminationThe longer the time, the higher the concentration, the larger the temperature rise amplitude of the nanoparticles; the results of the catalytic efficiency of the in vitro Fenton reaction are shown in FIG. 3, and it can be seen from the graph that Fe ions catalyze H under the 808nm near-infrared illumination and acidic condition2O2The capability of degrading methylene blue is obviously improved, namely the efficiency of Fenton reaction is enhanced.
(5) 1mL of 5mM Fe-GA @ BSA prepared in step (4) is added into 1mL of LPBS, 50 μ L of sorafenib solution (40mg/mL) is added under the condition of vigorous stirring at room temperature, ultrasonic treatment is carried out for 30 minutes by using a water bath after stirring for one hour, and then centrifugation is carried out for 5 minutes at 1000rpm to obtain Fe-GA @ BSA-SRF, wherein the atomic force microscope result is shown in FIG. 4, and the graph shows that the size of the nano-drug is uniform and is about 40 nm.
(6) The effects of the prepared Fe-GA @ BSA and Fe-GA @ BSA-SRF on photothermal combined iron treatment of mouse breast cancer cells 4T1 were studied, including killing effects on 4T1 cells, changes in mitochondrial membrane potential (see FIG. 5) and induction of Reactive Oxygen Species (ROS) production in cells (see FIG. 6). As can be seen from FIG. 5, the groups Fe-GA @ BSA and Fe-GA @ BSA-SRF both cause significant changes in mitochondrial membrane potential, so that the green fluorescence of the JC-1 probe is significantly enhanced, the red fluorescence is significantly reduced, and the changes in mitochondrial membrane potential are the important features of cell pig death. As shown in FIG. 6, Fe-GA @ BSA-SRF can induce the mouse breast cancer cell 4T1 to generate higher level of ROS under different power illumination compared with Fe-GA @ BSA, and the increased level of ROS is another important mark of cell iron death, which indicates that SRF drugs and iron death act synergistically and can achieve enhanced anti-tumor effect in combination with photothermal therapy when iron death occurs.
Example 2
The invention discloses a metal-protein nano-drug particle, which is a Fe-GA @ BSA-SRF protein-metal nano-drug particle prepared by the method, and comprises the following steps:
step (1) As in example 1
(2) 27 μ L of FeCl3Dropwise adding the solution (100mg/mL) into the BSA solution under the condition of stirring at room temperature, and stirring at room temperature for 30 min;
the steps (2) to (6) are the same as the example 1, and Fe-GA @ BSA-SRF nano-drug particles with lower unit Fe content are obtained.
Example 3
The invention discloses a metal-protein nano-drug particle, which is a Fe-TA @ OVA-SRF protein-metal nano-drug particle prepared by the method, and comprises the following steps:
(1) weighing 45mg of OVA, dissolving in ultrapure water, and stirring at room temperature for 30 minutes;
(2) 54 μ L of FeCl3Dropwise adding the solution (100mg/mL) into the OVA solution under the condition of stirring at room temperature, and stirring at room temperature for 30 min;
(3) dropwise adding 2.04mL of Tannic Acid (TA) solution (25mg/mL) into the Fe-OVA mixed solution under the condition of stirring, and then standing for 24 hours at room temperature in a dark place;
(4) filtering the obtained solution containing the nano particles by using an ultrafiltration tube, and washing for 2 times by using ultrapure water to obtain purified Fe-TA @ OVA nano particles;
(5) adding 1mL of 5mM Fe-TA @ OVA prepared in the step (4) into 1mL of PBS, adding 50 μ L of sorafenib solution (40mg/mL) under the condition of vigorous stirring at room temperature, stirring for one hour, then performing ultrasonic treatment for 30 minutes by using a water bath, and then centrifuging at 1000rpm for 5 minutes to obtain Fe-TA @ OVA-SRF;
step (6) as in example 1, the nanoparticles were obtained in which OVA was a protein and TA was a polyphenol.
Example 4
The invention discloses a metal-protein nano-drug particle, which is a Fe-GA @ BSA-RSL3 protein-metal nano-drug particle prepared by the method, and comprises the following steps:
the steps (1), (2), (3) and (4) in example 1 are reserved, and sorafenib in (5) is changed into RSL3 to obtain Fe-GA @ BSA-RSL3 nano drug particles.
Comparative example 1
The comparative example is a preparation method of metal-protein nano-drug particles, compared with the examples, the selected therapeutic drug can not induce the death of tumor cells, and the Fe-GA @ BSA-DOX protein-metal nano-drug particles prepared by the method comprise the following steps:
the steps (1), (2), (3) and (4) in the example 1 are reserved, and the sorafenib in the step (5) is changed into adriamycin to obtain Fe-GA @ BSA-DOX nano drug particles.
Comparative example 2
The comparative example is a preparation method of metal-protein nano-drug particles, compared with the examples, the selected therapeutic drug can not induce the death of tumor cells, and the Fe-GA @ BSA-CPT protein-metal nano-drug particles prepared by the method comprise the following steps:
the steps (1), (2), (3) and (4) in the example 1 are reserved, and the sorafenib in the step (5) is changed into camptothecin to obtain Fe-GA @ BSA-CPT nano-drug particles.
Comparative example 3
The comparative example is a preparation method of metal-protein nano-drug particles, compared with the examples, the selected therapeutic drugs can not induce the death of tumor cells, and the Fe-GA @ BSA-Irinotecan protein-metal nano-drug particles prepared by the method comprise the following steps:
the steps (1), (2), (3) and (4) in the example 1 are reserved, and the sorafenib in (5) is changed into Irinotecan to obtain Fe-GA @ BSA-Irinotecan nano-drug particles.
In order to confirm that a therapeutic drug capable of inducing iron death can act synergistically with the metal-protein nanoparticles prepared according to the present invention, and achieve an enhanced therapeutic effect in combination with photothermal therapy, experiments were conducted in examples 1, 4, comparative examples 1, 2 and 3, in which the drugs used in examples 1 and 4 were therapeutic drugs capable of inducing iron death, and comparative examples 1, 2 and 3 were therapeutic drugs incapable of inducing iron death. IC of each set of metal-protein nano-drug particles was calculated in 4T1 cells by MTT assay50Values, in free drug concentration as control, experimental data are shown in the following table:
Figure BDA0002906620120000061
as can be seen from the above table, the therapeutic drug inducing iron death acts synergistically with the metal-protein nanoparticles to significantly reduce the IC of the drug50Value, therapeutic drugs that kill tumor cells at lower concentrations without inducing iron death, after interaction with metal-protein nanoparticles, have little effect on their IC50The value is obtained. On the other hand, under near infrared illumination, each group had IC's compared to the unirradiated group50The values are all obviously reduced, which indicates that the photo-thermal combined iron death can further enhance the treatment effect.
The technical features of the embodiments described above may be arbitrarily combined, and for the sake of brevity, all possible combinations of the technical features in the embodiments described above are not described, but should be considered as being within the scope of the present specification as long as there is no contradiction between the combinations of the technical features. The following table lists the ratio of the amounts of the components in a series of synthesis steps, and the amounts of the components of the metal-protein nano-drug particles described in the present invention can be referred to, but not limited to, the following series of combinations:
combination of Protein (mu mol) Fe3+(μmol) Polyphenol (mu mol) Medicine (mu mol)
1 1 67 60 (Gallic acid) 67 (Sorafenib)
2 1 33 60 (Gallic acid) 33 (Sorafenib)
3 1 67 60 (Gallic acid) 134 (Sorafenib)
4 1 67 30 (tannic acid) 67 (Sorafenib)
5 1 67 50 (ellagic acid) 67 (Sorafenib)
The above-mentioned embodiments only express several embodiments of the present invention, and the description thereof is more specific and detailed, but not construed as limiting the scope of the invention. It should be noted that, for a person skilled in the art, several variations and modifications can be made without departing from the inventive concept, which falls within the scope of the present invention. Therefore, the protection scope of the present patent shall be subject to the appended claims.

Claims (10)

1. A metal-protein nanoparticle comprising a carrier protein, wherein: the carrier protein is compounded with Fe3+Ions while passing Fe3+Ion-chelating pharmaceutically acceptable polyphenolic compounds.
2. The metal-protein nanoparticle of claim 1, wherein: the carrier protein is selected from bovine serum albumin, human serum albumin and ovalbumin.
3. The metal-protein nanoparticle of claim 1, wherein: the polyphenol compound is selected from gallic acid, tannic acid, ellagic acid, epigallocatechin gallate, baicalin, myricetin, and delphinidin.
4. The metal-protein nanoparticle of claim 1, wherein: said Fe3+The molar ratio of the ions to the polyphenol compound is (17-67): (30-60).
5. The metal-protein nanoparticle of any one of claims 1 to 4, wherein: also loaded with therapeutic drugs that enhance iron death.
6. The metal-protein nanoparticle of claim 5, wherein: the therapeutic drug is at least one selected from sorafenib, elastine, FIN56, RSL3, lanpiriropine and sulfasalazine.
7. The metal-protein nanoparticle of claim 6, wherein: the therapeutic drug is sorafenib, Fe3+: the molar ratio of the sorafenib is 1 (0.8-5).
8. Use of metal-protein nanoparticles as claimed in any one of claims 1 to 7 in the preparation of a medicament for photothermal therapy of a tumor in combination with iron death.
9. A method for preparing a metal-protein nanoparticle according to any one of claims 1 to 7, comprising the steps of:
s1) fully stirring the carrier protein and the ferric salt solution to ensure that Fe3+Compounding the Fe-protein complex on carrier protein to obtain Fe-protein complex;
s2) adding a polyphenol compound into the obtained Fe-protein compound, stirring and standing to obtain a crude product of the metal-protein nano-particles;
s3) purifying the crude metal-protein nano-particles to obtain the finished metal-protein nano-particles.
10. The method of claim 9, wherein: adding therapeutic drugs for enhancing iron death into the metal-protein nanoparticle finished product and fully reacting to obtain the metal-protein nanoparticle for combined tumor photothermal therapy and iron death.
CN202110073109.8A 2021-01-20 2021-01-20 Nano medicine for treating tumor by combining photo-thermal treatment with iron agent and preparation method thereof Active CN112791185B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110073109.8A CN112791185B (en) 2021-01-20 2021-01-20 Nano medicine for treating tumor by combining photo-thermal treatment with iron agent and preparation method thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110073109.8A CN112791185B (en) 2021-01-20 2021-01-20 Nano medicine for treating tumor by combining photo-thermal treatment with iron agent and preparation method thereof

Publications (2)

Publication Number Publication Date
CN112791185A true CN112791185A (en) 2021-05-14
CN112791185B CN112791185B (en) 2022-07-08

Family

ID=75810662

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110073109.8A Active CN112791185B (en) 2021-01-20 2021-01-20 Nano medicine for treating tumor by combining photo-thermal treatment with iron agent and preparation method thereof

Country Status (1)

Country Link
CN (1) CN112791185B (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113647607A (en) * 2021-08-06 2021-11-16 广州大学 Mineral-loaded ovalbumin-polyphenol nanoparticles and preparation method and application thereof
CN114451511A (en) * 2022-01-05 2022-05-10 华南理工大学 Antibacterial nano-particles and preparation method and application thereof
CN114558150A (en) * 2022-03-03 2022-05-31 四川大学华西医院 Preparation method of magnetic resonance imaging nano probe for pH visualization
CN114632078A (en) * 2022-02-16 2022-06-17 中山大学附属第七医院(深圳) Heat shock protein inhibitor and preparation method and application thereof
CN114917191A (en) * 2022-04-08 2022-08-19 南通大学 Preparation method and application of vitamin B2-based acousto (photo) sensitive nanoparticle VFNS
CN115054688A (en) * 2022-05-10 2022-09-16 中国海洋大学 Cuttlefish ink chelated iron nanoparticles and preparation method and application thereof
CN115192544A (en) * 2022-07-12 2022-10-18 北京大学 Iron chelate nanoparticle for inducing iron death and preparation and application thereof
CN115429881A (en) * 2022-09-02 2022-12-06 中国医学科学院基础医学研究所 Iron chelators for treatment of tumors with beta-catenin activating mutations
CN116473944A (en) * 2023-04-27 2023-07-25 东北林业大学 Preparation method of FMMSNs capable of degrading tumor microenvironment responsiveness, prepared FMMSNs and application of FMMSNs

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014087413A1 (en) * 2012-12-03 2014-06-12 Vishwa Vidya Peetham University Amrita Nanoparticles comprising sorafenib
WO2015180325A1 (en) * 2014-05-28 2015-12-03 中国科学院生物物理研究所 Drug carrier for tumour-specific targeted drug delivery and use thereof
CN108619534A (en) * 2018-05-11 2018-10-09 上海师范大学 Multi-functional self-assemblies of a kind of GA-Fe@BSA-PTX and its preparation method and application
CN109806252A (en) * 2019-01-29 2019-05-28 中国药科大学 Tri compound nanometer system and its preparation method and application
CN110507821A (en) * 2019-08-29 2019-11-29 上海师范大学 A kind of pharmaceutical agent and preparation method thereof for treatment of colon cancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014087413A1 (en) * 2012-12-03 2014-06-12 Vishwa Vidya Peetham University Amrita Nanoparticles comprising sorafenib
WO2015180325A1 (en) * 2014-05-28 2015-12-03 中国科学院生物物理研究所 Drug carrier for tumour-specific targeted drug delivery and use thereof
CN108619534A (en) * 2018-05-11 2018-10-09 上海师范大学 Multi-functional self-assemblies of a kind of GA-Fe@BSA-PTX and its preparation method and application
CN109806252A (en) * 2019-01-29 2019-05-28 中国药科大学 Tri compound nanometer system and its preparation method and application
CN110507821A (en) * 2019-08-29 2019-11-29 上海师范大学 A kind of pharmaceutical agent and preparation method thereof for treatment of colon cancer

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
TAO LIU等: ""Ferrous-Supply-Regeneration Nanoengineering for Cancer-Cell-Specific Ferroptosis in Combination with ImagingGuided Photodynamic Therapy"", 《ACS NANO》 *
XUELING MU等: ""Bsa-assisted synthesis of ultrasmall gallic acid–Fe(III) coordination polymer nanoparticles for cancer theranostics"", 《INTERNATIONAL JOURNAL OF NANOMEDICINE》 *
王雅琼: ""功能蛋白引导制备铁-没食子酸纳米网用于肿瘤光热治疗"", 《中国优秀硕士学位论文全文数据库医药卫生科技辑》 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113647607A (en) * 2021-08-06 2021-11-16 广州大学 Mineral-loaded ovalbumin-polyphenol nanoparticles and preparation method and application thereof
CN113647607B (en) * 2021-08-06 2023-12-22 广州大学 Mineral-loaded ovalbumin-polyphenol nano-particles and preparation method and application thereof
CN114451511B (en) * 2022-01-05 2023-09-26 华南理工大学 Antibacterial nano-particles and preparation method and application thereof
CN114451511A (en) * 2022-01-05 2022-05-10 华南理工大学 Antibacterial nano-particles and preparation method and application thereof
CN114632078A (en) * 2022-02-16 2022-06-17 中山大学附属第七医院(深圳) Heat shock protein inhibitor and preparation method and application thereof
CN114632078B (en) * 2022-02-16 2023-12-19 中山大学附属第七医院(深圳) Heat shock protein inhibitor and preparation method and application thereof
CN114558150A (en) * 2022-03-03 2022-05-31 四川大学华西医院 Preparation method of magnetic resonance imaging nano probe for pH visualization
CN114558150B (en) * 2022-03-03 2023-06-23 四川大学华西医院 Preparation method of magnetic resonance imaging nano probe for pH visualization
CN114917191A (en) * 2022-04-08 2022-08-19 南通大学 Preparation method and application of vitamin B2-based acousto (photo) sensitive nanoparticle VFNS
CN115054688A (en) * 2022-05-10 2022-09-16 中国海洋大学 Cuttlefish ink chelated iron nanoparticles and preparation method and application thereof
CN115192544A (en) * 2022-07-12 2022-10-18 北京大学 Iron chelate nanoparticle for inducing iron death and preparation and application thereof
CN115192544B (en) * 2022-07-12 2024-01-02 北京大学 Iron chelate nanoparticle for inducing iron death and preparation and application thereof
CN115429881A (en) * 2022-09-02 2022-12-06 中国医学科学院基础医学研究所 Iron chelators for treatment of tumors with beta-catenin activating mutations
CN116473944A (en) * 2023-04-27 2023-07-25 东北林业大学 Preparation method of FMMSNs capable of degrading tumor microenvironment responsiveness, prepared FMMSNs and application of FMMSNs

Also Published As

Publication number Publication date
CN112791185B (en) 2022-07-08

Similar Documents

Publication Publication Date Title
CN112791185B (en) Nano medicine for treating tumor by combining photo-thermal treatment with iron agent and preparation method thereof
Yang et al. Red-light-triggered self-destructive mesoporous silica nanoparticles for cascade-amplifying chemo-photodynamic therapy favoring antitumor immune responses
Zhou et al. Photothermally triggered copper payload release for cuproptosis‐promoted cancer synergistic therapy
He et al. Magnetic graphene oxide: synthesis approaches, physicochemical characteristics, and biomedical applications
Shao et al. Cascade catalytic nanoplatform based on “butterfly effect” for enhanced immunotherapy
CN105682677B (en) Enhanced sensitivity composition using electromagnetic wave for the heat therapy of cancer, and the cancer therapy using it
CN113521098B (en) Platinum (IV) and cRGD modified GA/Fe nano-particle carried doxorubicin and method for targeted therapy of tumors by using same
CN113577101A (en) Tea polyphenol-metal nanoparticles, drug-loaded nanoparticles, preparation method and application thereof
Rehman et al. Mammalian cells: A unique scaffold for in situ biosynthesis of metallic nanomaterials and biomedical applications
Deng et al. Novel T7-modified pH-responsive targeted nanosystem for co-delivery of docetaxel and curcumin in the treatment of esophageal cancer
CN110403916B (en) Nano therapeutic agent and preparation method and application thereof
Liang et al. BSA-assisted synthesis of nanoreactors with dual pH and glutathione responses for ferroptosis and photodynamic synergistic therapy of colorectal cancer
Yang et al. Fabrication of magnetic nanochains linked with CTX and curcumin for dual modal imaging detection and limitation of early tumour
Hu et al. Mitochondria-targeted and multistage synergistic ROS-elevated drug delivery system based on surface decorated MnO2 with CeO2 for enhanced chemodynamic/chemotherapy
Huang et al. Bortezomib prodrug catalytic nanoreactor for chemo/chemodynamic therapy and macrophage re-education
Singh et al. Folic-acid adorned alginate-polydopamine modified paclitaxel/Zn-CuO nanocomplex for pH triggered drug release and synergistic antitumor efficacy
Wang et al. Applications of metal–phenolic networks in nanomedicine: a review
Zhang et al. An oxygen-economical nano-photosensitizer with a high photodynamic therapeutic outcome via simultaneous reduction of the cellular respiration and oxygen depletion of PDT
CN114177305A (en) Prodrug nanoparticle for inducing multi-mechanism death of tumor cells and preparation method and application thereof
CN109846857A (en) A kind of preparation method and applications of the natural supermolecule photosensitizer of activity
CN116327979B (en) Transition metal-based mesoporous nano catalytic medicine, preparation method and application
KR101513841B1 (en) Agglomerating magnetic alkoxysilane-coated nanoparticles
Erdoğan et al. Treatment of glioblastoma by photodynamic therapy with the aid of synthesized silver nanoparticles by green chemistry from Citrus aurantium
Ji et al. Evaluation of antioxidant, cytotoxicity, and anti-ovarian cancer properties of the Fe3O4@ CS-Starch/Cu bio-nanocomposite
Pan et al. Construction of novel multifunctional luminescent nanoparticles based on DNA bridging and their inhibitory effect on tumor growth

Legal Events

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