CN111529510A - Application of nanoparticles as tumor microenvironment responsive drug or imaging agent - Google Patents

Application of nanoparticles as tumor microenvironment responsive drug or imaging agent Download PDF

Info

Publication number
CN111529510A
CN111529510A CN202010387805.1A CN202010387805A CN111529510A CN 111529510 A CN111529510 A CN 111529510A CN 202010387805 A CN202010387805 A CN 202010387805A CN 111529510 A CN111529510 A CN 111529510A
Authority
CN
China
Prior art keywords
solution
dftnps
nanoparticles
tumor
mixture
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
CN202010387805.1A
Other languages
Chinese (zh)
Inventor
任建丽
周志益
罗远利
乔斌
***
冉海涛
李攀
郝兰
曹阳
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.)
Chongqing Medical University
Original Assignee
Chongqing 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 Chongqing Medical University filed Critical Chongqing Medical University
Priority to CN202010387805.1A priority Critical patent/CN111529510A/en
Publication of CN111529510A publication Critical patent/CN111529510A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5123Organic compounds, e.g. fats, sugars
    • 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
    • A61K41/0052Thermotherapy; Hyperthermia; Magnetic induction; Induction heating therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0002General or multifunctional contrast agents, e.g. chelated agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/08Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier
    • A61K49/10Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1821Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles
    • A61K49/1824Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles
    • A61K49/1827Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle
    • A61K49/1833Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with a small organic molecule
    • A61K49/1845Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with a small organic molecule the small organic molecule being a carbohydrate (monosaccharides, discacharides)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/22Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations
    • A61K49/222Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations characterised by a special physical form, e.g. emulsions, liposomes
    • A61K49/225Microparticles, microcapsules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5115Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5192Processes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

The invention belongs to the field of biomedicine, relates to application of nanoparticles, and particularly relates to application of nanoparticles as a tumor microenvironment responsive drug or an imaging agent. The nanoparticles comprise a chelate shell formed by tannic acid and ferric ions, and chemotherapeutic drugs are loaded in the chelate shell. The nano particles can slowly release chemotherapeutic drugs carried in the nano particles in a tumor microenvironment, and iron ions in the nano particles are slowly released to enhance the nuclear magnetic resonance imaging effect. The technical scheme can be applied to the medical practice of tumor imaging and tumor treatment.

Description

Application of nanoparticles as tumor microenvironment responsive drug or imaging agent
Technical Field
The invention belongs to the field of biomedicine, relates to application of nanoparticles, and particularly relates to application of nanoparticles as a tumor microenvironment responsive drug or an imaging agent.
Background
Chemotherapy is a classic and effective tumor treatment technique and has been widely used in clinical practice. However, the chemotherapy medicament lacks targeting, and is easy to be phagocytized and absorbed at non-pathological parts; moreover, some chemotherapeutic drugs have short half-life in vivo and are easy to inactivate in the in vivo transportation process; in addition, the burst release of chemotherapeutic agents also increases their systemic toxicity. The above disadvantages prevent the clinical application of chemotherapy. Therefore, more and more researchers and clinicians are dedicated to the study of loading chemotherapeutic drugs into nanoparticle drug delivery systems, and the rationally designed nanoparticle drug delivery systems are of great significance in improving the efficacy and biological safety of chemotherapy.
In the prior art, researchers encapsulated chemotherapeutic drugs in liposomes or polylactic acid-glycolic acid copolymer shells to form nanoparticles encapsulating chemotherapeutic drugs, thereby reducing the toxicity of chemotherapeutic drugs to normal tissues and preventing the chemotherapeutic drugs from being prematurely inactivated during in vivo operation. However, the prior art solutions have the following drawbacks: after the nanoparticles enter biological tissues, the tissue distribution and aggregation conditions of the chemotherapeutic drugs cannot be monitored in real time, the fixed-point release of the chemotherapeutic drugs at tumor tissues cannot be realized, and the control on the release speed of the chemotherapeutic drugs cannot be mentioned.
Disclosure of Invention
The invention aims to provide application of a nanoparticle as a tumor microenvironment responsive drug or an imaging agent, wherein the nanoparticle can slowly release chemotherapeutic drugs carried in the nanoparticle and iron ions in the nanoparticle so as to enhance nuclear magnetic resonance imaging effect in a tumor microenvironment.
In order to solve the technical problems, the technical scheme of the invention provides application of nanoparticles as a tumor microenvironment responsive drug or an imaging agent, wherein the nanoparticles comprise a chelate shell formed by tannic acid and ferric ions, and chemotherapy drugs are loaded in the chelate shell.
By adopting the technical scheme, the technical principle is as follows: the chelate shell formed by two substances of tannic acid and ferric ion is unstable in an acidic or glutathione environment and gradually dissociates, so that the chemotherapeutic drug wrapped in the chelate shell is gradually released, and ferric ion is released. The tumor microenvironment is an acidic and glutathione-rich environment, while the normal tissue microenvironment is neutral or weakly alkaline and does not contain glutathione. After the nano particles are injected into organisms, the nano particles are stable in nature and are not dissociated in normal tissues, but after the nano particles enter tumor tissues, the stability of the nano particles is reduced under the influence of pH value and glutathione (the nano particles have the characteristics of responding to the pH value and the glutathione), chemotherapeutic drugs entrapped in the nano particles are gradually released, the proliferation of tumor cells is inhibited, ferric ions in chelate shells are also gradually released, and the T1 weighted magnetic resonance imaging effect is enhanced.
Has the advantages that:
in the stimulus-responsive nanoparticle drug delivery system in the prior art, metal nanoparticles or liquid fluorocarbon microbubble substances are generally added to the nanoparticle drug delivery system, and the release of chemotherapeutic drugs in the nanoparticle drug delivery system is usually realized through external light, heat, ultrasonic or other stimuli. However, the additional external stimulation condition may cause some side effects to the living body, such as skin burn, increased radiation exposure, etc., and the process of promoting the release of the chemotherapy drug by using the external stimulation requires additional medical equipment, which increases the complexity of the operation and is not favorable for the clinical popularization of the technology. The inventor researches the response phenomenon of external light and thermal stimulation of the chelate nanoparticle formed by tannic acid and ferric ions, and unexpectedly finds that the chelate nanoparticle can be broken under the external light and thermal stimulation, the chelate nanoparticle can be dissociated in a tumor microenvironment, the dissociation process is carried out step by step, and the chemotherapeutic drug carried in the chelate nanoparticle can be released step by step, so that the slow release of the drug is realized, and the system toxicity of the drug is reduced. The inventor further researches the principle that the chelate nanoparticle formed by tannic acid and ferric ion can be dissociated in the tumor microenvironment. The oxygen content in the tumor cells is low, and the excessive secretion of glutathione into intercellular substance of the tumor cells is stimulated; in addition, in tumor cells, the excessive secretion of lactate or other tumor cell metabolites leads to an acidic microenvironment outside the tumor cell. The low pH value and the high glutathione environment stimulate the dissociation of the chelate nanoparticles, thereby promoting the release of the chemotherapeutic drugs in the chelate nanoparticles. In this scheme, the tumor microenvironment mainly refers to an acidic environment and an environment containing glutathione.
In conclusion, the nanoparticles in the technical scheme are applied to tumor microenvironment responsive drugs or imaging agents, and have the following beneficial effects:
(1) the nano particles in the technical scheme are stable in normal tissues, and release chemotherapeutic drugs in tumor tissues, so that the fixed-point release of the chemotherapeutic drugs at tumor parts is realized, and the fixed-point release does not need external stimulation such as light, heat or ultrasound and the like, is convenient to operate, and is suitable for clinical popularization.
(2) The shell of the nano particles in the technical scheme is gradually dissociated at the tumor tissue, so that the control of the drug release rate is realized, and the system toxicity caused by the burst release of the chemotherapeutic drug is avoided.
(3) The shell of the nano-particle in the technical scheme is gradually dissociated at the tumor tissue to gradually release ferric ions, so that T1 weighted magnetic resonance imaging can be enhanced, and the nano-particle can be used as an imaging agent to be applied to diagnosis and detection of tumors.
(4) The nano-particles in the technical scheme also have the properties of photothermal imaging (PTI) and photoacoustic imaging (PAI). After being chelated, the ferric ions and the tannic acid have the photoacoustic imaging property and can be monitored by a photoacoustic imager; the photo-thermal imaging material has photo-thermal imaging property under illumination and can be monitored by a thermal infrared imager.
(5) The nanoparticles in the technical scheme also have the effect of photothermal therapy (PTT). After chelation of ferric ions and tannic acid, the compound has high photo-thermal conversion efficiency, and can generate thermal effect under the irradiation of 600-900nm laser, thereby realizing the photo-thermal treatment of tumors.
(6) The chelate formed by the tannic acid and the ferric ions coats and modifies the chemotherapeutic drug, reduces the toxicity of the chemotherapeutic drug and increases the circulation time of the chemotherapeutic drug in blood, thereby preventing the chemotherapeutic drug from being prematurely inactivated during in vivo operation.
Further, the ferric ion is provided by ferric chloride.
By adopting the technical scheme, the ferric trichloride is a common chemical reagent, is easy to obtain and has low cost.
Further, the chemotherapeutic agent is doxorubriosin.
By adopting the technical scheme, the Tourethricin is a medicament widely applied to chemotherapy, and has clear pharmacological action and good treatment effect.
Further, the nanoparticles are prepared by the following method:
step (1) preparation of hydrophobic topiroxomicin: dissolving hydrochloric acid carbendazim in dimethyl sulfoxide solution, adding trimethylamine, and uniformly mixing to obtain a mixture;
step (2) preparation of the shell: dripping high-purity water into the mixture obtained in the step (1) and stirring the mixture simultaneously to obtain dispersion liquid; sequentially adding a tannic acid solution and a ferric trichloride hexahydrate solution into the dispersion liquid to obtain a mixed dispersion liquid, and carrying out ultrasonic treatment on the mixed dispersion liquid to obtain a crude product solution, wherein the crude product solution contains nano particles;
and (3) post-treatment: adjusting the pH value of the crude product solution to be neutral, and then washing and collecting the nano particles in the crude product solution.
By adopting the technical scheme, the nanoparticles with uniform particle size, stable property, good dispersion degree and high chemotherapeutic drug encapsulation rate can be prepared. In the step (1), hydrophobic polyoxins nanoparticles (mixture B is a hydrophobic polyoxins nanoparticle solution) are prepared, namely hydrophobic polyoxins self-assemble into a nano inner core template, and then on the basis of the inner core template, a chelate film formed by tannic acid and ferric ions is covered on the inner core template. Tannic acid is a naturally occurring polyphenol that can act as a chelating site for ferric ions at room temperature. The nano particles (DOX-Fe-TA-nanoparticles, DFTNPs) of the technical scheme are formed by the synergistic effects of coordination, hydrophobicity, P-P accumulation and the like on the surface of a nano core template formed by self-assembly of hydrophobic polyoxins. The hydrophobic polyoxins nanoparticles are used as the core, and a chelate film is formed on the core, so that compared with the traditional scheme of preparing an outer film and then wrapping a medicament, the preparation scheme is simpler, and the medicament encapsulation rate is higher.
High purity water (High purity water) is referred to in this case as: water having a conductivity of less than 0.1. mu.s/cm at 25 ℃ and a residual salt content of less than 0.3 mg/L.
Furthermore, in the step (1), the dosage ratio of the hydrochloric acid carbendazim to the dimethyl sulfoxide solution to the trimethylamine is (3-50) mg, (3-50) ml and (0.3-5) ml.
By adopting the technical scheme, the hydrophobic Toxoplasma variegated nanoparticles with stable properties and proper particle size can be obtained (namely, the hydrophobic Toxoplasma variegated nanoparticles are self-assembled into a nano inner core template).
Further, in the step (2), the volume ratio of the mixture, the high-purity water, the tannic acid solution and the ferric chloride hexahydrate solution is 100:980 (10-100) to (10-100); the concentration of tannic acid in the tannic acid solution is 40mg/ml, and the concentration of ferric chloride hexahydrate in the ferric chloride hexahydrate solution is 2-20 mg/ml.
By adopting the technical scheme, the tannic acid and ferric ions can quickly form cross-linking, and a chelate film is formed outside the hydrophobic Toxoplasma nanoparticles.
Further, in the step (2), high pure water is dropped to the mixture at a dropping speed of 100 to 1000. mu.l/min while stirring the mixture at a rotation speed of 300 to 1000 rpm.
By adopting the technical scheme, the mixture is stirred at a high speed, and high pure water is simultaneously dripped, so that hydrophobic multi-ratio soft star can be fully dispersed, nano-scale multi-ratio soft star particles are obtained, and subsequent chelation and attachment of tannic acid and ferric ions are facilitated.
Further, in step (3), the pH of the crude solution was adjusted to neutral using 1mM NaOH.
By adopting the technical scheme, 1mM NaOH is a common pH value adjusting reagent, has stable property and is easy to obtain.
Furthermore, the particle diameter of the nano-particles is 84.0-209.8 nm, and the surface potential is-26.7-16.1 mV.
By adopting the technical scheme, the nanoparticles with the particle sizes have better bioavailability and can pass through the vascular wall to reach the target tissue with higher efficiency.
Further, the polymer dispersion coefficient of the nano particles is 0.100-0.200.
By adopting the technical scheme, the dispersion coefficient of the polymer is not more than 0.2, the nano particles are uniform and dispersed, and the agglomeration tendency is small. The lower the polymer dispersion coefficient pdi (polymer dispersion index), the better the nanoparticle dispersion and the smaller the tendency to agglomerate.
Drawings
FIG. 1 is a transmission electron micrograph of DFTNPs of Experimental example 1;
FIG. 2 is a graph showing particle sizes of DFTNPs in Experimental example 1;
FIG. 3 is a potential diagram of DFTNPs of Experimental example 1;
FIG. 4 is a graph showing UV absorption of DFTNPs of Experimental example 1;
FIG. 5 is a graph of the in vitro drug release profiles (different GSH content) of DFTNPs from Experimental example 2;
FIG. 6 is a graph showing in vitro drug release profiles (different pH values) of DFTNPs of Experimental example 2;
FIG. 7 is a graph showing the results of photothermal effect experiments (different concentrations of DFTNPs) of DFTNPs of Experimental example 3;
FIG. 8 is a graph showing the results of photothermal effect experiments (different irradiation intensities) of DFTNPs of Experimental example 3;
FIG. 9 is a thermal imaging graph of different concentrations of DFTNPs in Experimental example 3 under 808nm laser irradiation;
FIG. 10 is a graph showing temperature change curves of different components in DFTNPs of Experimental example 3 under 808nm laser irradiation;
FIG. 11 shows the results of the CCK-8 method for evaluating cytotoxicity of DFTNPs in Experimental example 4;
FIG. 12 is a result of the CCK-8 method for evaluating phototoxicity of DFTNPs in Experimental example 4;
FIG. 13 is a graph showing the evaluation of apoptosis of DFTNPs by flow cytometry under different treatment conditions in Experimental example 4;
FIG. 14 shows the phagocytosis of DFTNPs by MDA-MB-231 cells of Experimental example 5 at different observation time points;
FIG. 15 shows the results of photoacoustic imaging in vitro using DFTNPs from Experimental example 6;
FIG. 16 shows the results of in vivo photoacoustic imaging experiments using DFTNPs from Experimental example 6;
FIG. 17 is an MRI image of DFTNPs from Experimental example 7 at different GSHs;
FIG. 18 is an MRI image of DFTNPs of Experimental example 7 at different pH;
FIG. 19 is an in vivo MRI image of DFTNPs of Experimental example 7;
FIG. 20 is the photothermographic signals of the tumor sites of tumor-bearing nude mice with DFTNPs of Experimental example 8 at different time points;
FIG. 21 is the temperature changes of the tumor sites of nude mice bearing tumor at different time points for DFTNPs of Experimental example 8;
FIG. 22 shows the tumor volume changes of the DFTNPs of Experimental example 8 in tumor-bearing nude mice at different observation time points;
FIG. 23 shows the body weight changes of tumor-bearing nude mice with DFTNPs of Experimental example 8 at different observation time points.
Detailed Description
The following is further detailed by way of specific embodiments:
example 1: preparation of nanoparticles
10mg DOX & HCl (doxorubicin hydrochloride) was dissolved in 10ml DMSO (dimethyl sulfoxide) solution. Then, the mixture is stirred moderately at room temperature, 1ml of trimethylamine is added, hydrochloric acid is removed to convert hydrophilic DOX & HCl into hydrophobic DOX nano particles, and the mixture is obtained after the mixture is stirred uniformly. Then, 100. mu.l of the prepared mixture (DOX/DMSO solution neutralized with trimethylamine) was dropped into 980. mu.l of high purity water with vigorous stirring (stirring at 700 rpm), the dropping speed of the high purity water being 800. mu.l/min, to obtain a dispersion. Then 10. mu.l of Ta (tannic acid) solution (40mg/ml) and 10. mu.l of FeCl3·6H2O solution (20mg/ml) was added to the above dispersion in order, followed by sonication for 50s and neutralization with 1. mu.M NaOH solution. Centrifugally washing the obtained product with deionized water to remove excessive TA and FeCl3. And re-dispersing the nano particles in high-purity water to obtain the DFTNPs.
In the preparation process, the dosage ratio of DOX & HCl, DMSO solution and trimethylamine can be (3-50) mg, (3-50) ml and (0.3-5) ml. Mixture, high purity water, Ta solution and FeCl3·6H2The volume ratio of the O solution can be 100:980, (10-100) to (10-100). FeCl3·6H2FeCl in O solution3·6H2The concentration of O can be 2-20 mg/ml. Dropping highly pure water into the mixture at a dropping speed of 100 to 1000. mu.l/min while stirring the mixture at a rotation speed of 300 to 1000 rpm.
Example 2
10mg DOX & HCl (doxorubicin hydrochloride) was dissolved in 10ml DMSO (dimethyl sulfoxide) solution. Then, with moderate stirring at room temperature, 1ml of trimethylamine was added and the hydrochloric acid was removed to convert the hydrophilic DOX · HCl into hydrophobic DOX nanoparticles. Then, 100. mu.l of prepared DOX/DMSO was dissolvedThe liquid was dropped into 980. mu.l of high purity water with vigorous stirring (stirring at 1000 rpm) at a rate of 1000. mu.l/min to obtain a dispersion. Then 100. mu.l of Ta (tannic acid) solution (40mg/ml) and 100. mu.l of FeCl3·6H2O solution (20mg/ml) was added to the above dispersion in order, followed by sonication for 50s and neutralization with 1. mu.M NaOH solution. Centrifugally washing the obtained product with deionized water to remove excessive TA and FeCl3. And re-dispersing the nano particles in high-purity water to obtain the DFTNPs.
Example 3
10mg DOX & HCl (doxorubicin hydrochloride) was dissolved in 10ml DMSO (dimethyl sulfoxide) solution. Then, with moderate stirring at room temperature, 1ml of trimethylamine was added and the hydrochloric acid was removed to convert the hydrophilic DOX · HCl into hydrophobic DOX nanoparticles. Then, 980. mu.l of high purity water was dropped into 100. mu.l of the prepared DOX/DMSO solution under vigorous stirring (stirring at 300 rpm), and the dropping rate of the high purity water was 100. mu.l/min, to obtain a dispersion. Then 50. mu.l of Ta (tannic acid) solution (40mg/ml) and 50. mu.l of FeCl3·6H2O solution (20mg/ml) was added to the above dispersion in order, followed by sonication for 50s and neutralization with 1. mu.M NaOH solution. Centrifugally washing the obtained product with deionized water to remove excessive TA and FeCl3. And re-dispersing the nano particles in high-purity water to obtain the DFTNPs.
Experimental example 1: characteristics and physical Properties of the nanoparticles
For the DFTNPs prepared in example 1, the morphology of the nanoparticles was observed using a transmission electron microscope, and the particle size and potential of DOXNPs and DFTNPs were measured using a potentiostat (see fig. 1, 2 and 3). The particle size of DFTNPs is: 146.9 +/-62.9 nm, potential: -21.4 ± 5.3 mV. The preparation method of the DOXNPs comprises the following steps: 10mg DOX & HCl (doxorubicin hydrochloride) was dissolved in 10ml DMSO (dimethyl sulfoxide) solution. Then, with moderate stirring at room temperature, 1ml of trimethylamine was added to the mixture and the hydrochloric acid was removed to convert hydrophilic DOX · HCl into hydrophobic DOX nanoparticles. Detection of DOXNPs, DFTNPs and Fe using UV absorption spectroscopy3+UV absorption spectrum of/TA (see FIG. 4). Wherein, Fe3+The term/TA means direct conversion of Fe3+Chelating with tannic acid and then sonicatingDispersed nanoparticles formed.
Experimental example 2: encapsulation efficiency and in vitro drug release
And (3) determining the encapsulation efficiency: UV-Vis absorption peaks of DFTNPs were measured and standard curves for DOX were plotted by measuring the UV absorption spectra at 480nm of solutions of various concentrations of DOX (300,150,75,37.5,0.3125 μ g/mL). By comparing the ultraviolet absorption of DOX in the supernatant, the encapsulation efficiency of DFTNPs to DOX is calculated to be 76.3% by combining a standard curve.
In vitro drug release of DOX: drug release experiments were performed at different pH and GSH concentrations (glutathione). 5mL of DFTNPs solution was placed in a dialysis bag (MWCO: 3500Da) and immersed in 15mL of buffer and the buffer was stirred at 160rpm for 120 h. The release of DOX from DFTNPs was measured at pH 5.5,6.5,7.4 and GSH concentrations 0mM,2mM,10mM (calculated according to the standard curve). The results are shown in fig. 5 and 6, which demonstrate that the release rate of DOX varies with the pH and GSH concentration. When the nano-particle moves in vivo, the pH value of a cancer part is low, the GSH content is high, and the release of DOX can be promoted, so that the therapeutic effect of the nano-particle is realized. However, in normal tissues, if the pH is higher and the GSH content is lower, the release amount of DOX is reduced, and the side effect of the DOX on the normal tissues is reduced. The nanoparticle DFTNPs have a selective release function, so that side effects are reduced, and the treatment effect is improved.
Experimental example 3: in vitro photothermal effect study
DFTNPs were added to a 96-well plate, and the relationship between the concentration of DFTNPs and the photothermal effect was investigated by irradiating DFTNPs (50,100,200,300,400. mu.g/mL) at different concentrations for 10min with 808nm laser. Irradiation with 808nm laser light of different intensities (0.5,1.0,1.5,2.0W/cm2) And (5) irradiating the DFTNPs for 10min, and researching the relation between the irradiation intensity and the photothermal effect. As shown in fig. 7 and 8, the higher the concentration of DFTNPs and the stronger the irradiation intensity, the stronger the photothermal effect of the nanoparticle. Meanwhile, the photothermal effects of DFTNPs with different irradiation times are also researched, and the experimental result is shown in FIG. 9, wherein the longer the irradiation time is, the higher the concentration of DFTNPs is, and the stronger the photothermal effect of the nanoparticle is.
To investigate the photothermal sources of DFTNPs, DFTNPs (400. mu.g/mL), DOXNPs (400. mu.g/mL), tannic acid (400. mu.g/mL), FeCl3(400. mu.g/mL) and Fe 3+200. mu.L of each of the solutions/TA was added to a 96-well plate as a control group, and a 808nm laser (2.0W/cm)2) The irradiation was carried out for 10min and the temperature change was recorded using a near infrared imager (Fotric 226). The results of the experiment are shown in FIG. 10, FeCl3Tannic acid and DOXNPs all have no photothermal effect, Fe3+the/TA has a certain photothermal effect, and the DFTNPs formed by adding DOX into the nanoparticles have a stronger heat supply effect.
Experimental example 4: cytotoxicity and proliferation assay
The cytotoxicity and photothermal therapeutic effects of DFTNPs were evaluated using the CCK-8 assay. MDA-MB-231 cells were plated on a 96-well plate overnight, 100. mu.L of DFTNPs (8.13-65. mu.g/ml) at various concentrations were added to the plate, and incubated for 36h with the cells, 10. mu.L of CCK-8 solution was added to the plate, and the cell activity was measured using a plate reader. The experimental results are shown in FIG. 11, and the DFTNPs have lower cytotoxicity and are safer than DOXNPs.
To further evaluate the photothermal chemotherapeutic synergistic therapeutic effect of DFTNPs, different concentrations of DFTNPs (25-200. mu.g/ml) were incubated with cells for 36 h. Then the cells were irradiated with laser at 808nm (2.0W/cm)2) Irradiating for 10min, incubating for 12h, and detecting cell activity by CCK-8 method. As shown in fig. 12, after laser irradiation, DFTNPs showed cytotoxicity and were applicable to photothermal chemotherapy of cancer cells.
To further evaluate the apoptosis of MDA-MB-231 cells after DFTNPs treatment, MDA-MB-231 cells were placed in a 6-well plate for 12h to allow complete cell adhesion, and then DOXNPs (50. mu.g/ml) and DFTNPs (50. mu.g/ml) were added to incubate for 24h, followed by laser irradiation of the cells at 808nm (2.0W/cm)2) Irradiating for 10min, and detecting cell activity by flow cytometry. The experimental result is shown in fig. 13, the DFTNPs can promote more apoptosis after laser irradiation, and have the effect of promoting cancer cell apoptosis.
Example 5: endocytosis assay
To investigate that DFTNPs could be efficiently phagocytosed by MDA-MB-231 cells, MDA-MB-231 cells (5 × 10)4One) was planted on a glass petri dish and incubated for 12h, after which the existing culture was discardedAnd (3) adding DFTNPs into nutrient, incubating for different times (1h, 2h, 4h and 8h), fully washing cells, and staining cell nuclei for 15min by using DAPI. Cell phagocytosis was observed using confocal laser microscopy. The experimental results are shown in fig. 14, and the experimental results prove that the DFTNPs can be effectively phagocytized by cells.
Example 6: in vivo and in vitro photoacoustic imaging
To investigate the potential of DFTNPs as contrast agents for photoacoustic imaging, photoacoustic imaging evaluations were performed using a photoacoustic imager (Visual sonic). Different concentrations of DFTNPs (40-200 mug/ml) are placed in a gel model, and photoacoustic imaging signals of the DFTNPs are detected by a photoacoustic imager. As shown in fig. 15, the photoacoustic imaging signal becomes stronger as the ion concentration increases.
Tumor-bearing mice were used to evaluate the photoacoustic imaging effect of DFTNPs in vivo. Injecting DFTNPs (5mg/mL) into tail vein, and observing the photoacoustic imaging signal intensity of the DFTNPs at the tumor site at different time points (0h, 2h, 4h and 24 h). As shown in fig. 16, DFTNPs have photoacoustic imaging signals at tumor sites in tumor-bearing nude mice.
Example 7: in vivo and in vitro MRI imaging signals
To investigate the potential of DFTNPs as contrast agents for magnetic resonance imaging, magnetic resonance imaging evaluations were performed using a magnetic resonance imager (simensmrc 406903.0 t MRI imager). Different concentrations of DFTNPs (40-200. mu.g/ml) were placed in a test tube model. The DFTNPs were adjusted to different pH values (4.5,7.4) and different GSH contents (2mM, 10mM), and the magnetic resonance imaging signals of the DFTNPs were detected by a magnetic resonance imager. The experimental results are shown in fig. 17 and 18, and the GSH concentration and the pH value have a certain influence on the nuclear magnetic imaging intensity of the DFTNPs.
Tumor-bearing mice were used to evaluate the MRI imaging effect of DFTNPs in vivo. DFTNPs (5mg/mL) were injected into the tail vein, and the MRI imaging signal intensity of the DFTNPs at the tumor site was observed at different time points (0h, 2h, 4h, and 24 h). The experimental result is shown in fig. 19, and the DFTNPs are proved to have the function of tumor MRI imaging.
Example 8: in vivo antitumor therapeutic study
By subcutaneous injection of MDA-MB-231 cells (100. mu.L, 10) into the right hip of nude mice7one/mL) to establish a tumor-bearing mouse model.When the tumor of the mouse grows to 100mm3Tumor-bearing nude mice were randomly divided into 6 groups (n ═ 5): the control group, the simple laser group, the DOXNPs group, the DFTNPs group, the DOXNPs plus laser group and the DFTNPs plus laser group are respectively. Tumor-bearing mice were injected with 5% glucose solution, DOXNPs solution and DFTNPs solution via tail vein, respectively. After 24h, the tumors were irradiated with 808nm laser (2W/cm)210 min). The temperature change at the tumor site was recorded using a thermal infrared imager. The body weight of the nude mice and the major axis (L) and minor axis (W) of the tumor were recorded every two days. Tumor volume (V) according to V ═ tumor major diameter (L)2× minor diameter (W)/2, relative tumor volume was calculated using V/V0, where V represents tumor volume and V0 represents initial tumor volume the results of the experiments are shown in FIGS. 20-23, and the in vivo experiments show that DFTNPs have better tumor therapeutic effect than DOXNPs.
The foregoing is merely an example of the present invention and common general knowledge in the art of designing and/or characterizing particular aspects and/or features is not described in any greater detail herein. It should be noted that, for those skilled in the art, without departing from the technical solution of the present invention, several variations and modifications can be made, which should also be regarded as the protection scope of the present invention, and these will not affect the effect of the implementation of the present invention and the practicability of the patent. The scope of the claims of the present application shall be determined by the contents of the claims, and the description of the embodiments and the like in the specification shall be used to explain the contents of the claims.

Claims (10)

1. The application of the nanoparticle as a tumor microenvironment responsive drug or an imaging agent is characterized in that the nanoparticle comprises a chelate shell formed by tannic acid and ferric ion, and chemotherapeutic drugs are loaded in the chelate shell.
2. Use according to claim 1, wherein the ferric ions are provided by ferric chloride.
3. The use of claim 2, wherein the chemotherapeutic agent is doxorubriosin.
4. Use according to claims 1-3, wherein the nanoparticles are prepared by a process comprising:
step (1) preparation of hydrophobic topiroxomicin: dissolving hydrochloric acid carbendazim in dimethyl sulfoxide solution, adding trimethylamine, and uniformly mixing to obtain a mixture;
step (2) preparation of the shell: dripping high-purity water into the mixture obtained in the step (1) and stirring the mixture simultaneously to obtain dispersion liquid; sequentially adding a tannic acid solution and a ferric trichloride hexahydrate solution into the dispersion liquid to obtain a mixed dispersion liquid, and carrying out ultrasonic treatment on the mixed dispersion liquid to obtain a crude product solution, wherein the crude product solution contains nano particles;
and (3) post-treatment: adjusting the pH value of the crude product solution to be neutral, and then washing and collecting the nano particles in the crude product solution.
5. The use of claim 4, wherein in the step (1), the dosage ratio of the hydrochloric acid carbendazim to the dimethyl sulfoxide solution to the trimethylamine is (3-50) mg, (3-50) ml, (0.3-5) ml.
6. The use as claimed in claim 5, wherein in the step (2), the volume ratio of the mixture, the high-purity water, the tannic acid solution and the ferric chloride hexahydrate solution is 100:980 (10-100): 10-100); the concentration of tannic acid in the tannic acid solution is 40mg/ml, and the concentration of ferric chloride hexahydrate in the ferric chloride hexahydrate solution is 2-20 mg/ml.
7. The use according to claim 6, wherein in the step (2), the dropping speed of high pure water into the mixture is 100 to 1000. mu.l/min, and the mixture is stirred at a rotation speed of 300 to 1000rpm at the same time.
8. The use as claimed in claim 7, wherein in step (3), 1mM NaOH is used to adjust the pH of the crude solution to neutral.
9. The use according to claim 8, wherein the nanoparticles have a particle size of 84.0 to 209.8nm and a surface potential of-26.7 to-16.1 mV.
10. The use according to claim 9, wherein the nanoparticles have a polymer dispersion factor of 0.100 to 0.200.
CN202010387805.1A 2020-05-09 2020-05-09 Application of nanoparticles as tumor microenvironment responsive drug or imaging agent Pending CN111529510A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010387805.1A CN111529510A (en) 2020-05-09 2020-05-09 Application of nanoparticles as tumor microenvironment responsive drug or imaging agent

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010387805.1A CN111529510A (en) 2020-05-09 2020-05-09 Application of nanoparticles as tumor microenvironment responsive drug or imaging agent

Publications (1)

Publication Number Publication Date
CN111529510A true CN111529510A (en) 2020-08-14

Family

ID=71973483

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010387805.1A Pending CN111529510A (en) 2020-05-09 2020-05-09 Application of nanoparticles as tumor microenvironment responsive drug or imaging agent

Country Status (1)

Country Link
CN (1) CN111529510A (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111821470A (en) * 2020-09-01 2020-10-27 中南大学 Methotrexate-entrapped iron-tannic acid complex and preparation method and application thereof
CN113181211A (en) * 2021-05-12 2021-07-30 南京大学深圳研究院 Fe2O3@ TA-Pt nano composite material and preparation method and application thereof
CN113262203A (en) * 2021-06-02 2021-08-17 郑州大学 Preparation method and application of self-assembled nano-drug liposome for resisting Alzheimer's disease
CN113413468A (en) * 2021-06-29 2021-09-21 首都医科大学附属北京儿童医院 Photo-thermal-hardening combined therapy targeted nano-drug delivery system
CN113521298A (en) * 2021-07-01 2021-10-22 东华大学 Responsive dendrimer drug-loaded material coated by tannic acid/iron complex
CN114404429A (en) * 2021-11-30 2022-04-29 重庆医科大学附属第二医院 Nano-silver modified tannin-iron network drug-loaded nano-composite, preparation method thereof and application thereof in reversing tumor drug resistance
CN114470246A (en) * 2022-01-21 2022-05-13 南方医科大学 Tumor microenvironment dual-response type composite nano material for anti-contrast magnetic resonance imaging and preparation method thereof

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009133413A1 (en) * 2008-04-28 2009-11-05 Novatech D.O.O. Formulation based on micronized clinoptilolite as a therapeutic agent for removal of toxins, bacteria and viruses from organism
CN103948934A (en) * 2014-04-09 2014-07-30 中国科学院过程工程研究所 Drug nanoparticle preparation based on complexing coating and preparation method and application
CN104194460A (en) * 2014-08-12 2014-12-10 西南交通大学 Preparation method of chelate coating with copper ions capable of controllably catalyzing release of nitrogen monoxide and polyphenol complex
CN107115322A (en) * 2017-05-23 2017-09-01 江苏省原子医学研究所 A kind of nanoparticle vector
CN107381580A (en) * 2017-07-13 2017-11-24 福州大学 A kind of preparation method of the interior doping metal net shaped Biodegradable silica dioxide granule of polyphenol
CN107441513A (en) * 2017-09-30 2017-12-08 中国科学院长春应用化学研究所 A kind of coordination polymer nano particle based on polyphenol and preparation method thereof
CN108653741A (en) * 2018-05-24 2018-10-16 华中科技大学同济医学院附属协和医院 A kind of natural sericin microballoon and its preparation method and application of metal organic coordination polymer package
CN109106952A (en) * 2018-07-28 2019-01-01 华南理工大学 A kind of preparation method of the drug-carrying nanometer particle of targeted therapy malignant lymphoma
CN109200021A (en) * 2018-09-17 2019-01-15 重庆医科大学 A kind of pH sensitive nanoparticles and the preparation method and application thereof of lysosome targeting
CN110237264A (en) * 2019-01-31 2019-09-17 华东理工大学 A kind of PLGA nano particle and preparation method thereof for TA-Fe (III) modification containing doxorubicin hydrochloride
CN110665005A (en) * 2019-10-29 2020-01-10 重庆医科大学 Iron-doped polymer nanoparticle and preparation method and application thereof

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009133413A1 (en) * 2008-04-28 2009-11-05 Novatech D.O.O. Formulation based on micronized clinoptilolite as a therapeutic agent for removal of toxins, bacteria and viruses from organism
CN103948934A (en) * 2014-04-09 2014-07-30 中国科学院过程工程研究所 Drug nanoparticle preparation based on complexing coating and preparation method and application
CN104194460A (en) * 2014-08-12 2014-12-10 西南交通大学 Preparation method of chelate coating with copper ions capable of controllably catalyzing release of nitrogen monoxide and polyphenol complex
CN107115322A (en) * 2017-05-23 2017-09-01 江苏省原子医学研究所 A kind of nanoparticle vector
CN107381580A (en) * 2017-07-13 2017-11-24 福州大学 A kind of preparation method of the interior doping metal net shaped Biodegradable silica dioxide granule of polyphenol
CN107441513A (en) * 2017-09-30 2017-12-08 中国科学院长春应用化学研究所 A kind of coordination polymer nano particle based on polyphenol and preparation method thereof
CN108653741A (en) * 2018-05-24 2018-10-16 华中科技大学同济医学院附属协和医院 A kind of natural sericin microballoon and its preparation method and application of metal organic coordination polymer package
CN109106952A (en) * 2018-07-28 2019-01-01 华南理工大学 A kind of preparation method of the drug-carrying nanometer particle of targeted therapy malignant lymphoma
CN109200021A (en) * 2018-09-17 2019-01-15 重庆医科大学 A kind of pH sensitive nanoparticles and the preparation method and application thereof of lysosome targeting
CN110237264A (en) * 2019-01-31 2019-09-17 华东理工大学 A kind of PLGA nano particle and preparation method thereof for TA-Fe (III) modification containing doxorubicin hydrochloride
CN110665005A (en) * 2019-10-29 2020-01-10 重庆医科大学 Iron-doped polymer nanoparticle and preparation method and application thereof

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
GUIZHI SHEN,等: "Interfacial Cohesion and Assembly of Bioadhesive Molecules for Design of Long-Term Stable Hydrophobic Nanodrugs toward Effective Anticancer Therapy", 《ACS NANO》 *
SANGHOON KIM,等: "pH- and glutathione-responsive release of curcumin from mesoporous silica nanoparticles coated using tannic acid–Fe(III) complex", 《RSC ADV.》 *
TAO LIU,等: "Ferrous-Supply-Regeneration Nanoengineering for Cancer-Cell-Specific Ferroptosis in Combination with Imaging-Guided Photodynamic Therapy", 《ACS NANO》 *
TAO LIU,等: "Metal Ion/Tannic Acid Assembly as a Versatile Photothermal Platform in Engineering Multimodal Nanotheranostics for Advanced Applications", 《ACS NANO》 *
XIANGYU MENG,等: "A metal–phenolic network-based multifunctional nanocomposite with pH-responsive ROS generation and drug release for synergistic chemodynamic/photothermal/chemo-therapy", 《JOURNAL OF MATERIALS CHEMISTRY B》 *
YANG CHAO,等: "Dual ultrasound-activatable nanodroplets for highly-penetrative and efficient ovarian cancer theranostics", 《JOURNAL OF MATERIALS CHEMISTRY B》 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111821470A (en) * 2020-09-01 2020-10-27 中南大学 Methotrexate-entrapped iron-tannic acid complex and preparation method and application thereof
CN111821470B (en) * 2020-09-01 2022-08-12 中南大学 Methotrexate-entrapped iron-tannic acid complex and preparation method and application thereof
CN113181211A (en) * 2021-05-12 2021-07-30 南京大学深圳研究院 Fe2O3@ TA-Pt nano composite material and preparation method and application thereof
CN113262203A (en) * 2021-06-02 2021-08-17 郑州大学 Preparation method and application of self-assembled nano-drug liposome for resisting Alzheimer's disease
CN113262203B (en) * 2021-06-02 2022-11-11 郑州大学 Preparation method and application of self-assembled nano-drug liposome for resisting Alzheimer disease
CN113413468A (en) * 2021-06-29 2021-09-21 首都医科大学附属北京儿童医院 Photo-thermal-hardening combined therapy targeted nano-drug delivery system
CN113413468B (en) * 2021-06-29 2023-05-12 首都医科大学附属北京儿童医院 Photothermal-hardening combined treatment targeting nano-drug delivery system
CN113521298A (en) * 2021-07-01 2021-10-22 东华大学 Responsive dendrimer drug-loaded material coated by tannic acid/iron complex
CN113521298B (en) * 2021-07-01 2022-11-01 东华大学 Responsive dendrimer drug-loaded material coated by tannic acid/iron complex
CN114404429A (en) * 2021-11-30 2022-04-29 重庆医科大学附属第二医院 Nano-silver modified tannin-iron network drug-loaded nano-composite, preparation method thereof and application thereof in reversing tumor drug resistance
CN114470246A (en) * 2022-01-21 2022-05-13 南方医科大学 Tumor microenvironment dual-response type composite nano material for anti-contrast magnetic resonance imaging and preparation method thereof
CN114470246B (en) * 2022-01-21 2023-01-03 南方医科大学 Tumor microenvironment dual-response type composite nano material for anti-contrast magnetic resonance imaging and preparation method thereof

Similar Documents

Publication Publication Date Title
CN111529510A (en) Application of nanoparticles as tumor microenvironment responsive drug or imaging agent
Li et al. Nanoscale metal‐organic frameworks: synthesis, biocompatibility, imaging applications, and thermal and dynamic therapy of tumors
Zhang et al. Janus γ-Fe2O3/SiO2-based nanotheranostics for dual-modal imaging and enhanced synergistic cancer starvation/chemodynamic therapy
Chen et al. NIR-II light activated photodynamic therapy with protein-capped gold nanoclusters
Cheng et al. Gold nanosphere gated mesoporous silica nanoparticle responsive to near-infrared light and redox potential as a theranostic platform for cancer therapy
Zhou et al. Doxorubicin‐loaded single wall nanotube thermo‐sensitive hydrogel for gastric cancer chemo‐photothermal therapy
Fan et al. A smart upconversion-based mesoporous silica nanotheranostic system for synergetic chemo-/radio-/photodynamic therapy and simultaneous MR/UCL imaging
Ma et al. Au capped magnetic core/mesoporous silica shell nanoparticles for combined photothermo-/chemo-therapy and multimodal imaging
Jing et al. Mn-porphyrin conjugated Au nanoshells encapsulating doxorubicin for potential magnetic resonance imaging and light triggered synergistic therapy of cancer
Gao et al. AuNRs@ MIL-101-based stimuli-responsive nanoplatform with supramolecular gates for image-guided chemo-photothermal therapy
Wang et al. Preparation and synergistic chemo-photothermal therapy of redox-responsive carboxymethyl cellulose/chitosan complex nanoparticles
Xiong et al. Polydopamine-mediated bio-inspired synthesis of copper sulfide nanoparticles for T1-weighted magnetic resonance imaging guided photothermal cancer therapy
Chen et al. Graphene quantum dots mediated magnetic chitosan drug delivery nanosystems for targeting synergistic photothermal-chemotherapy of hepatocellular carcinoma
Sun et al. MnO 2 nanoflowers as a multifunctional nano-platform for enhanced photothermal/photodynamic therapy and MR imaging
Vu-Quang et al. Carboxylic mannan-coated iron oxide nanoparticles targeted to immune cells for lymph node-specific MRI in vivo
Pitchaimani et al. Gd3+ tethered gold nanorods for combined magnetic resonance imaging and photo-thermal therapy
Wang et al. Construction of ICG encapsulated W18O49@ MSN as a fluorescence carrier for real-time tracked photothermal therapy
Liu et al. pH-responsive hollow Fe–gallic acid coordination polymer for multimodal synergistic-therapy and MRI of cancer
Wang et al. Tumor-targeting multi-shelled hollow nanospheres as drug loading platforms for imaging-guided combinational cancer therapy
Harvey et al. Recent advances in nanoscale metal–organic frameworks towards cancer cell cytotoxicity: an overview
Tian et al. Gadolinium-doped hollow silica nanospheres loaded with curcumin for magnetic resonance imaging-guided synergistic cancer sonodynamic-chemotherapy
Hou et al. Remodeling the tumor microenvironment with core–shell nanosensitizer featuring dual-modal imaging and multimodal therapy for breast cancer
Pal et al. NIR-responsive 5-Fluorouracil delivery using polydopamine coated polygonal CuS nanoplates for synergistic chemo-photothermal therapy on breast cancer
Li et al. H2O2/pH dual-responsive biomimetic nanoenzyme drugs delivery system for enhanced tumor photodynamic therapy
Huang et al. Tumor microenvironment–responsive versatile “Trojan horse” theranostic nanoplatform for magnetic resonance imaging–guided multimodal synergistic antitumor treatment

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
RJ01 Rejection of invention patent application after publication
RJ01 Rejection of invention patent application after publication

Application publication date: 20200814