CN112546062B - Perfluorocarbon silicon plastid and preparation method and application thereof - Google Patents

Perfluorocarbon silicon plastid and preparation method and application thereof Download PDF

Info

Publication number
CN112546062B
CN112546062B CN202011166195.9A CN202011166195A CN112546062B CN 112546062 B CN112546062 B CN 112546062B CN 202011166195 A CN202011166195 A CN 202011166195A CN 112546062 B CN112546062 B CN 112546062B
Authority
CN
China
Prior art keywords
preparation
tumor
silica
drug
hifu
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202011166195.9A
Other languages
Chinese (zh)
Other versions
CN112546062A (en
Inventor
王凡
梁晓龙
马晓途
史继云
姚美男
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.)
Institute of Biophysics of CAS
Peking University Third Hospital Peking University Third Clinical Medical College
Original Assignee
Institute of Biophysics of CAS
Peking University Third Hospital Peking University Third Clinical Medical College
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 Institute of Biophysics of CAS, Peking University Third Hospital Peking University Third Clinical Medical College filed Critical Institute of Biophysics of CAS
Priority to CN202011166195.9A priority Critical patent/CN112546062B/en
Publication of CN112546062A publication Critical patent/CN112546062A/en
Application granted granted Critical
Publication of CN112546062B publication Critical patent/CN112546062B/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
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • 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
    • 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
    • A61K47/02Inorganic compounds
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Inorganic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Organic Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Acoustics & Sound (AREA)
  • Radiology & Medical Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

The invention discloses a silicon plastid-encapsulated perfluorocarbon preparation, and a preparation method and application thereof. The silicate network structure on the surface of the siliceous body ensures that the preparation has good structural stability and drug-loading stability, can reduce the early drug leakage of the preparation in a blood circulation system and reduce the toxic and side effects of the drug. The preparation can also enhance the effect of ultrasonic imaging, and realize drug delivery and drug release under image monitoring.

Description

Perfluorocarbon silicon plastid and preparation method and application thereof
Technical Field
The invention belongs to the field of biomedical materials, and particularly relates to a perfluorocarbon preparation wrapped by a silicon body, and a preparation method and application thereof.
Background
Tumor hypoxia is one of the most common characteristics of solid tumors. Hypoxia is a malignant feature of the tumor microenvironment, which is closely related to the resistance of tumors to various therapies, including chemotherapy, radiation therapy and photodynamic therapy. In particular with respect to resistance to chemotherapy, hypoxia may lead to multidrug resistance (multidrug resistance) in tumors, which may ultimately lead to failure of chemotherapy. During the process of multidrug resistance, tumor hypoxia firstly increases the relative expression level of hypoxia inducible factor-1 alpha (HIF-1 alpha) of tumor cells, thereby increasing the transcription level of multidrug resistance gene 1(MDR1), and finally increasing the protein expression level of P-glycoprotein (P-gp), and P-gp is used as an 'efflux pump' of various chemotherapeutic drugs and can pump the drugs entering the tumor cells back to the outside of cells. Therefore, improving hypoxia is an effective method for enhancing the efficacy of chemotherapy. Tumor metastasis is the leading cause of death in most cancer patients. Hypoxia has been reported to promote epithelial-mesenchymal transition (epithelial-mesenchymal transition) of tumor cells, accelerating tumor metastasis. Therefore, improving hypoxia can increase the curative effect of chemotherapy and inhibit the metastasis of tumor.
Researchers have currently explored a number of approaches to improve tumor hypoxia. Hyperbaric oxygen therapy can directly increase the concentration of oxygen in the systemic blood, although it has been used clinically, it lacks tumor specificity, increases the damage of radiotherapy, chemotherapy to normal tissues, and risks hyperoxicosis. There is an urgent need to develop a tumor-specific oxygen delivery system. Among the different types of oxygen delivery materials, perfluorocarbons (perfluorcarbon) have a great advantage due to their higher oxygen solubility and better biocompatibility. Perfluorocarbons have been widely used in clinical settings as contrast agents for ultrasound imaging, to prevent ischemia and reperfusion injury, and are expected to be used as artificial blood. Perfluorocarbon-based nanodroplets have been reported to alleviate tumor hypoxia and enhance the efficacy of radiotherapy and photodynamic therapy.
Although perfluorocarbons have a high oxygen solubility, their rate of oxygen release is inefficient. Unlike hemoglobin, which releases oxygen molecules rapidly under hypoxic conditions by virtue of a synergistic effect between its four subunits, perfluorocarbons release oxygen solely by virtue of the free diffusion of oxygen along a concentration gradient. Therefore, to enhance the efficiency of oxygen release from perfluorocarbons, it is necessary to rely on external stimuli to trigger the release of oxygen. Previous reports have utilized near infrared light or ultrasound to trigger the release of oxygen, but clinical applications have been limited because of limited tissue penetration by near infrared light. Common ultrasound has high tissue penetrability, but ultrasonic waves cannot be focused, the release of oxygen cannot be controlled at fixed points, the controlled release is not accurate enough, and normal tissues near tumors can also be subjected to ultrasonic treatment.
Perfluorocarbons can be used not only as carriers for oxygen delivery, but also as drug delivery systems (drug delivery systems). Small-molecule chemotherapeutic drugs are often limited in therapeutic effect and have serious side effects, and researchers have been concerned about the development of intelligent drug delivery systems for a long time to realize the synergy and the attenuation of the chemotherapeutic drugs. Perfluorocarbons enable drug delivery under ultrasound imaging monitoring, as well as ultrasound-triggered drug release, and hence perfluorocarbon-based drug delivery systems have unique advantages.
Good structural and drug loading stability is critical to the drug delivery system because it avoids premature release of the drug from the delivery system in the blood circulation system, thereby reducing side effects of the drugThe application is as follows. The wrapping agent of the perfluorocarbon nano material reported in the literature mainly comprises phospholipid or other surfactants, albumin, erythrocyte membranes and mesoporous nano materials, such as Bi 2 Se and SiO 2 . These encapsulates have limited drug loading stability and are complex to synthesize. There is a need to develop a perfluorocarbon drug delivery system with a simple synthetic process and high stability.
Disclosure of Invention
One of the purposes of the invention is to use the silica as a wrapping agent of the carbon fluoride, improve the structural stability and the drug-loading stability of a carbon fluoride drug delivery system, reduce the early leakage of the drug in blood circulation and reduce the side effect of the drug.
The second purpose of the invention is to trigger the carbon fluoride preparation to release oxygen and the medicine by using High Intensity Focused Ultrasound (HIFU), so as to realize the high intensity focused ultrasound stimulation response type oxygen and medicine controlled release, improve the enrichment amount of the medicine in the tumor and increase the medicine effect of the medicine.
The third purpose of the invention is to utilize the carbon fluoride preparation carried by the silica plastid to deliver oxygen to the tumor in a targeted way, improve the hypoxia of the tumor, relieve the multi-drug resistance and epithelial-mesenchymal transition of the tumor, improve the drug effect of chemotherapy and reduce the metastasis of the tumor.
The purpose of the invention is realized by the following technical scheme:
a silica body encapsulating a fluorocarbon-based compound, the silica body being a particle having a core-shell structure, the shell being a single lipid molecule layer consisting of a silica-forming lipid and a phospholipid component not containing Si, the silica-forming lipid forming a silicate network structure of the form-Si-O-Si-on an outer surface of the shell; the core contains a fluorocarbon-based compound.
Further, according to the present invention, the mole percentage of the liposome-forming lipid is 50 to 100% and the mole percentage of the phospholipid component not containing Si is 0 to 50% based on the total number of moles of the monolayer of lipid molecules. In some embodiments of the invention, the mole percentage of the liposome forming lipids is, for example, 94-95%, 60%, 50%, and the balance is a phospholipid component that is free of Si.
Further, according to the present invention, the carbon fluoride compound refers to a compound formed by replacing all or part of hydrogen bonded to carbon in an organic compound with fluorine. The organic compounds include, but are not limited to, hydrocarbons, alcohols, esters, ethers. The hydrocarbon is, for example, a saturated or unsaturated chain or cyclic hydrocarbon of C3-10. The alcohol is, for example, a C1-10 monohydric or polyhydric alcohol. The esters are, for example, C2-10 esters. The ethers are, for example, C2-30 chain or cyclic mono-or poly-ethers. In some embodiments of the invention, the fluorocarbon-based compound is selected from the group consisting of perfluoropropane, perfluoropentane, perfluorohexane, perfluorooctane bromide, perfluorocrown ether. According to the present invention, preferred are fluorocarbon-based compounds having a boiling point of 40-80 ℃ at atmospheric pressure, including but not limited to perfluorohexane, perfluoroheptane, dodecafluorohexane, monobromododecafluorohexane. The inventor of the invention finds that the carbon fluoride compound in the boiling point range can meet the requirements of targeted tumor administration, proper drug encapsulation rate and drug loading rate, oxygen carrying and drug release under the guidance of HIFU.
Further, according to the present invention, the liposome-forming lipid comprises an inorganic precursor, a linker and a hydrophobic tail, which has the following structural formula: a. the m CH (3-m) —CH 2 —Y—(CH 2 ) p —Z—R q
Wherein A is: (R) 1 O) 3 Si—(CH 2 ) 3 —N—C(O)—X—(CH 2 ) n —;R 1 Is alkyl, preferably C1-10 alkyl, more preferably methyl, ethyl; x is-O-or a bond; n is an integer from 0 to 10, such as 0, 1,2, 3, 4, 5, 6, 7, 8, 9, or 10, preferably 0, 1, 2;
m is an integer of 1 to 3; when m is 2 or 3, the 2 or 3A can be the same or different;
y is-CH 2 -, -O-C (O) -or-C (O) -O-;
p is an integer from 0 to 10, such as 0, 1,2, 3, 4, 5, 6, 7, 8, 9 or 10, preferably 0, 1, 2;
z is-C (O) -NH ( 2-q ) -or-CH ( 3-q )—;
R is alkyl with more than 5 carbon atoms, preferably linear saturated alkyl with C7-20 carbon atoms;
when Z is-C (O) -NH ( 2-q ) -q is an integer from 1 to 2; when Z is-CH ( 3 -q) is an integer from 1 to 3; when q is 2 or 3, the 2 or 3R may be the same or different.
In some embodiments of the invention, the liposome-forming lipids are selected from the group consisting of complex lipids a-D as shown below. The synthesis procedure of the four complex lipids is described in the literature (chem. eur. j.2013,19, 16113-16121).
Figure BDA0002745855000000041
Further, according to the present invention, the silicate network structure on the surface of the silica body can be formed by in situ sol-gel reaction (in situ sol-gel reaction) from the formation of silica body lipid, for example, by the following reaction mechanism:
-Si-OCH 2 CH 3 +H 2 O→-Si-OH+CH 3 CH 2 OH
2-Si-OH→-Si-O-Si-+H 2 O
further, according to the present invention, the Si-free phospholipid component may be selected from Si-free phospholipid components known in the art, including but not limited to: polyethylene glycol derivative DSPE-mPEG of distearoylphosphatidylethanolamine capable of prolonging blood circulation time 1000~20000 Such as DSPE-mPEG2000 (distearoylphosphatidylethanolamine-polyethylene glycol 2000) and DSPE-mPEG5000 (distearoylphosphatidylethanolamine-polyethylene glycol 5000); maleimide-modified distearoylphosphatidylethanolamine or polyethylene glycol derivative of Maleimide-modified distearoylphosphatidylethanolamine DSPE-mPEG that can be used to link targeting molecules 1000~20000 For example, DSPE-Maleimide (distearoylphosphatidylethanolamine-modified Maleimide), DSPE-mPEG 2000 -Maleimide (II)Stearoyl phosphatidylethanolamine-polyethylene glycol 2000-maleimide); dipalmitoylphosphatidylcholine (DPPC) and 1-myristoyl-2-stearoyl lecithin (MSPC) which can regulate the transition temperature of the siliceous body; hydrogenated soy lecithin (HSPC); distearoylphosphatidylcholine (DSPC). The non-Si containing phospholipid component may be a mixture of one or more non-Si containing phospholipids.
Further, according to the present invention, the surface of the silica plastid may be modified with a tumor targeting molecule selected from any one of an antibody, a polypeptide, an aptamer, folic acid or a folic acid derivative, which can target a tumor. In one embodiment of the invention, the tumor targeting molecule is var7 polypeptide (ref: mol. pharmaceuticals 2014,11,2896-2905) with an amino acid sequence of ACEEQNPWARYLEWLFPTETLLLEL. In some embodiments of the invention, the tumor targeting molecule is a folate derivative, such as a sulfhydryl-polyethylene glycol 2000~6000 Folic acid, in particular mercapto-polyethylene glycol 5000 -folic acid.
Further, according to the invention, the particle size of the silica body is 10nm-1 μm.
Further, according to the invention, the core of the silica body can encapsulate a small molecule drug. The small molecule drug is a water insoluble or hydrophobic drug, including but not limited to one or more of a chemotherapeutic drug, a protein kinase inhibitor, a photodynamic drug. In some embodiments of the invention, the small molecule drug is selected from the group consisting of doxorubicin, cisplatin, paclitaxel, irinotecan, sorafenib, gefitinib, porfimer sodium, verteporfin, indocyanine green.
Further, according to the invention, the core of the silica body may carry oxygen. The capacity of the silica body to carry oxygen is realized by dissolving oxygen by carbon fluoride compounds. When the adsorption of the silica body to oxygen reaches equilibrium, 0-2 ml of oxygen can be dissolved in each ml of carbon fluoride compound in the environment with the temperature of 15-40 ℃ and the oxygen partial pressure of 0-101325 Pa. When the partial pressure of oxygen in the environment in which the silica bodies are located increases (or decreases), the amount of oxygen adsorbed by the silica bodies increases (or decreases) accordingly. When the silica body enters a human body through intravenous injection and is transported to the area B from the area A, if the oxygen concentration of the area B is higher than that of the area A, the silica body preparation adsorbs oxygen in the area B; if the oxygen concentration in zone B is lower than that in zone a, the silica will release oxygen in zone B. The blood oxygen content of the vein of the normal human body is 110-180mL/L, the blood oxygen content of the artery is 150-230mL/L, the oxygen concentration of the vein blood is lower than that of the artery blood, and the oxygen concentration of the artery blood is higher than that of the tumor tissue outside the tumor blood vessel (including the tumor cells and the tumor extracellular stroma). When the silica body enters arterial blood from venous blood after intravenous injection into a human body, the silica body adsorbs oxygen; when arterial blood enters the tumor tissue outside the tumor vessel, the silica will release oxygen. Thus, the silica can carry oxygen in arterial blood to the tumor tissue.
Further, according to the invention, the silica body has good stability: after being treated by 0-100 mu M surfactant Triton-100, the drug release rate of the silica is less than 20%. Compared with the liposome composed of common phospholipids and carrying the carbon fluoride compounds, the liposome has higher stability: after being treated by 10-100 mu M surfactant Triton-100, the drug release rate of the silica liposome is 30-90% lower than that of the liposome.
Further, according to the present invention, the silica body can respond to the stimulation of the high intensity focused ultrasound to achieve the drug release and oxygen release controlled by the high intensity focused ultrasound: the silica body is processed by high-intensity focused ultrasound with the ultrasonic intensity (pulse width modulation) of 1% -10% for 0.5-5 minutes, the oxygen release rate of the silica body can reach 5% -100%, and the drug release rate of the silica body can reach 5% -100%.
Figure BDA0002745855000000051
Further, according to the present invention, the silica also has the ability to enhance ultrasound imaging.
The invention further provides a preparation method of the silica body.
A method for preparing the aforementioned silica bodies, comprising the steps of: adding fluorocarbon compound, liposome-forming lipid and phospholipid component without Si, or further containing drug into buffer solution or water, homogenizing, and high-pressure extruding to obtain uniform particle size distribution.
Further, according to the present invention, the fluorocarbon-based compound, the liposome-forming lipid and the phospholipid component not containing Si, or the drug further contained therein, may be dissolved in an organic solvent prior to the addition of the buffer solution or water. The organic solvent may be alcohols (e.g., ethanol, propanol, etc.), dimethyl sulfoxide, etc.
Further, according to the present invention, the buffer solution may be a PBS buffer, a Tris-HCl buffer, or the like. The pH of the buffer solution may be 5.0-8.0.
The silica body of the invention can also be prepared by another method. The preparation method comprises the following steps: 1) hydrolyzing the formed siliceous plastid lipids in an acidic alcohol solution; 2) dissolving the phospholipid component without Si or the medicine further contained in the phospholipid component in an organic solvent, and adding the dissolved phospholipid component or the medicine further contained in the phospholipid component into the hydrolysate in the step 1) to form a mixture; 3) volatilizing the mixture of step 2) to obtain a phospholipid membrane, hydrating the membrane, and homogenizing; the fluorocarbon-based compound is added during the hydration operation or during the homogenization operation.
Further, according to the present invention, the acidic alcohol solution is prepared by dissolving an alcohol in an aqueous dilute acid solution. The alcohol may be ethanol, propanol, isopropanol, etc.; the acid may be hydrochloric acid, sulfuric acid, nitric acid, etc.
Further, according to the present invention, the organic solvent may be alcohols (e.g., ethanol, etc.), dichloromethane, chloroform, dimethylformamide, etc.
Further, according to the present invention, the hydration may use water, PBS buffer, Tris-HCl buffer, or the like.
According to the invention, the silica preparation is obtained by centrifugation after homogenization. After homogenization, the mixture may be further passed through a high-pressure extruder to obtain a silica preparation having a more uniform particle size.
According to the invention, after obtaining a preparation of silica, the preparation is modified with a tumor targeting molecule.
According to the invention, after the preparation of the silica, the material which is not encapsulated in the silica is removed, for example by washing, filtration and/or chromatography on a column.
According to the invention, if the medicine is contained, the mass ratio of the medicine to the phospholipid component (the sum of the liposome forming the silica body and the phospholipid without Si) is preferably 1: 10-1: 2, and the ratio can influence the medicine loading amount and the encapsulation rate of the silica body, and the medicine loading amount and the encapsulation rate of the silica body are optimal within the preferable mass ratio range of the invention.
The invention further provides a pharmaceutical application of the silica body.
The application of the silica body in preparing oxygen delivery carriers or medicines is provided. Based on the application, the silica can be prepared into a medicament for improving tumor hypoxia.
The invention relates to application of a silica body in preparation of a carrier for delivering a drug. Based on the application, the silica body can be prepared into a tumor-targeted medicament, wherein the tumor-targeted medicament is encapsulated, so that the tumor-targeted medicament can be simultaneously administered with oxygen and the medicament, the tumor hypoxia is improved, the medicament treatment effect is improved, and the tumor metastasis incidence rate is reduced. The application of the silica body in preparing tumor treatment medicines.
The application of the silica body in preparing the medicine for treating the tumor by combining with the HIFU is disclosed. The medicine can improve tumor hypoxia and reduce the incidence rate of tumor metastasis. The medicine can be used as new adjuvant chemotherapy medicine for thermotherapy. The HIFU can increase the temperature of target tissues (such as tumors) from normal body temperature (37 ℃) to target temperature (40-80 ℃) within 20 minutes and maintain the target temperature for more than 30 minutes; adjusting the power of the HIFU may change the target temperature.
In one embodiment of the invention, the tumor is a breast cancer, e.g., a triple negative breast cancer.
In one embodiment of the invention, the tumor therapy drug is doxorubicin.
The application of the silica body in preparing the ultrasonic imaging agent is provided.
The invention has the beneficial effects that:
one of the advantages of the invention is that the silica body is used as a wrapping agent of the carbon fluoride, which can improve the stability of a drug delivery system, reduce the early leakage of the drug in blood circulation and reduce the side effect of the drug. The silica body is an inorganic-organic hybrid material, the structure of the silica body is similar to that of a liposome consisting of common phospholipid, and the amphiphilic composite lipid is self-assembled to form micro or nano particles. However, compared with the traditional phospholipid liposome, the surface of the liposome is of a single-atom-layer network structure formed by organosilicate, and the liposome has higher structural stability and drug-loading stability. Compared with silicon nanoparticles, the phospholipid layer structure of the silica reduces the overall hardness and density of the nanoparticles, reduces the injection dosage of silicon element, and improves the biological safety. The carbon fluoride compound has strong hydrophobicity and very low loading content by adopting the silica body. The invention improves the microstructure of the silica body through research, skillfully solves the problem of high content entrapment of the highly hydrophobic carbon fluoride compounds by adopting a monomolecular layer silica body structure, and keeps the stability of the silica body as a delivery system.
The preparation of the carbon fluoride compound encapsulated by the silica body can respond to the stimulation of high-intensity focused ultrasound, and the high-intensity focused ultrasound can trigger the rapid release of the medicine and the oxygen in a target area. High-intensity focused ultrasound (HIFU) is a non-invasive technique used clinically for thermal ablation therapy. HIFU can precisely control and elevate the temperature of target tissue by converting ultrasonic energy into thermal and mechanical energy. HIFU can slightly elevate the temperature of the target tissue (<43 ℃), and has been used for the heat-sensitive release of drugs without damaging the tissue. A slight increase in temperature increases tumor blood flow, so HIFU itself may also alleviate tumor hypoxia to some extent. The siliceous body can respond to the stimulation of the HIFU, so as to release the oxygen or the medicine carried in the siliceous body, realize the accurate fixed-point control of the release of the oxygen or the medicine, and better relieve the hypoxic of the tumor by combining the heating effect of the HIFU.
The third advantage of the invention is that the preparation of the carbon fluoride compound carried by the silica plastid can deliver the drug and the oxygen to the tumor site in a targeted way. The targeted delivery of the medicine can increase the drug effect of the medicine, and the targeted delivery of the oxygen can improve tumor hypoxia, relieve the multidrug resistance and epithelial-mesenchymal transition of tumors, improve the drug effect and reduce tumor metastasis.
The preparation of the carbon fluoride compound encapsulated by the silica body has the effect of enhancing ultrasonic imaging, can realize drug delivery and drug release under image monitoring, and can estimate the treatment effect.
Drawings
FIG. 1: the invention discloses a structural schematic diagram of a silicon plastid-encapsulated carbon fluoride preparation.
FIG. 2: the cryoetching scanning electron microscope image of the silicon plastid-entrapped fluorocarbon preparation prepared in example 3 shows that the nano preparation is spherical and has an average particle size of about 100 nm.
FIG. 3: the drug loading stability of the silica-encapsulated fluorocarbon formulations prepared in example 3 and example 5, respectively, was compared to that of the common phospholipid-encapsulated fluorocarbon formulations.
FIG. 4 is a schematic view of: the controlled release effect of the drug after the high intensity focused ultrasound treatment of the silicon plastid-entrapped fluorocarbon preparation prepared in example 3 is evaluated.
FIG. 5: evaluation of oxygen controlled release effect of the silica-encapsulated fluorocarbon formulation prepared in example 3.
FIG. 6: ultrasonic imaging effect evaluation of the silica-encapsulated fluorocarbon formulations prepared in example 3.
FIG. 7: the drug-loading stability of the preparation consisting of different kinds of carbon fluoride.
FIG. 8: sonogram plots of different formulations before (a-d) and after (I-V) 1 minute of focused ultrasound treatment with M-HIFU (PWM ═ 2%): (a) and (I) D-PFP-vPCs, (b) and (II) D-PFH-vPCs, (c) and (III) D-PFOB-vPCs, (D) and (IV) D-PFCE-vPCs, (V) PBS.
FIG. 9: therapeutic effect of the preparation at cellular level.
FIG. 10: the preparation can improve the distribution of the medicine in vivo.
FIG. 11: the in vivo chemotherapy effect of the preparation.
FIG. 12: the preparation has effect in reducing tumor metastasis.
FIG. 13: the preparation has therapeutic effect as new adjuvant chemotherapy means.
Detailed Description
The present invention will be described in further detail with reference to examples. However, those skilled in the art will appreciate that the scope of the present invention is not limited to the following examples. In light of the present disclosure, those skilled in the art will recognize that many variations and modifications may be made to the embodiments described above without departing from the spirit and scope of the present invention.
Unless otherwise indicated, the starting materials and reagents used in the following examples are all commercially available products or can be prepared by known methods; the manipulations performed are all known in the art or performed according to the user's manual of commercially available products.
Example 1 preparation of silica-Encapsulated fluorocarbon formulations
(1) Mu.mol of complex lipid A, 0.5. mu. mol of DSPE-mPEG2000, 4. mu. mol of the small molecule drug doxorubicin and 20. mu.L of perfluorohexane were dissolved in 2mL of ethanol, injected into 20mL of PBS, and during and within 15 minutes after injection, the 20mL of PBS was sonicated using a water bath sonicator (Prima, UK, model PM3-900 TL). Thereafter, sonication was continued for 10 minutes at 50% output power using a probe-type sonicator (Qsonica, usa, model Q700) under ice bath conditions. The liquid was repeatedly extruded through a polycarbonate filter (pore size: 100nm, Whatman, USA) 3 times by means of a high-pressure extruder (Northern Lipid, USA) to obtain a preparation having an average particle size of about 100 nm.
(2) To remove doxorubicin that was not encapsulated in the siliceous preparation, the preparation obtained in step (1) was first filtered through a 0.45 μm filter and free doxorubicin was removed using exclusion chromatography column Sephadex G-50.
(3) The formulation was tested for carbon fluoride and drug loading: taking 0.1mL of 100mg/mL of the preparation (the concentration of the preparation is calculated by taking the weight of the preparation after freeze-drying as the weight of a solute, the solute is equivalent to 10mg of the freeze-dried silica, and the freeze-dried silica does not contain perfluorohexane due to volatilization of the perfluorohexane), dissolving the silica with 500 muL of methanol, detecting the content of the perfluorohexane by a gas chromatograph, and determining that 6.24mg of the perfluorohexane is contained in each 10mg of the freeze-dried silica. Similarly, 0.1mL of 100mg/mL of the preparation was dissolved in an acidified isopropanol solution (10% of 1mol/L hydrochloric acid, 90% isopropanol), and the content of doxorubicin was measured by fluorescence spectrophotometry, whereby 1.75mg of doxorubicin was contained per 10mg of lyophilized silica, and from this, the drug loading of doxorubicin was calculated to be 17.5%, and the encapsulation efficiency was calculated to be 84.9%.
(4) Detecting whether the outer layer of the silicon plastid of the preparation is a single-layer lipid molecular layer or not, wherein the method I comprises the following steps: the thickness of the outer shell of the preparation is observed by a cryo-electron microscope, and the result shows that the thickness of the outer shell of the preparation is about 2nm, and the thickness of the phospholipid bilayer is about 4.5nm, so that the outer shell of the preparation is a lipid monolayer. The second method comprises the following steps: dissolving 9.5 mu mol of the compound lipid A, 0.5 mu mol of DSPE-mPEG2000, 2 mu mol of micromolecular drug adriamycin and 20 mu L of perfluorohexane, 0.01 mu mol of water-soluble dye sodium calcein and 0.01 mu mol of fat-soluble dye nile red in 2mL of ethanol, and detecting the ultraviolet-visible light absorption curve of the prepared preparation by the same subsequent treatment method as the steps (1) and (2). The detection result shows that the preparation contains a characteristic absorption peak of nile red and does not have a characteristic absorption peak of calcein sodium, which indicates that the preparation does not have a water cavity, so that the outer layer is a monolayer lipid molecular layer.
Example 2 preparation of silica-Encapsulated fluorocarbon formulations
(1) Mu. mol of complex lipid B was hydrolyzed in acidic ethanol at 40 ℃ for 30 minutes. The acidic ethanol solution is prepared by adding dilute hydrochloric acid into ethanol solution, and adjusting pH value to 3.0 by using a pH meter.
(2) Dissolving 1 mu mol of paclitaxel, a small molecule drug, 4 mu mol of phospholipid components without Si (including 3 mu mol of hydrogenated soybean lecithin (HSPC) and 1 mu mol of DSPE-mPEG2000) in 2mL of chloroform, adding the hydrolysate obtained in step (1), volatilizing the liquid to form a phospholipid film, vacuum-drying overnight, adding 5mL of ultrapure water and 20 mu L of perfluorocrown ether, shaking and hydrating for 60 minutes, and then continuously performing ultrasonic treatment for 10 minutes by using a probe type ultrasonic instrument (Qsonica, model Q700, USA) at an output power of 50% under an ice bath condition to obtain a preparation with an average particle size of about 200 nm.
(3) To remove paclitaxel that is not encapsulated in the siliceous preparation, the preparation obtained in step (2) is first filtered through a 0.45 μm filter and then freed paclitaxel is removed using exclusion chromatography column Sephadex G-50.
(4) Using the same assay as in example 1, it was determined that 5.1mg of perfluorocrown ether and 0.9mg of paclitaxel were contained per 10mg of lyophilized siliceous mass.
Example 3 preparation of silica-Encapsulated fluorocarbon formulations with tumor targeting molecules
(1) Mu.mol of complex lipid A, 0.2. mu. mol of DSPE-mPEG2000, 4. mu. mol of small molecule drug doxorubicin, 20. mu.L of perfluorohexane, 0.3. mu. mol of DSPE-mPEG2000-Maleimide were dissolved in 2mL of ethanol, injected into 20mL of PBS, and the 20mL of PBS was sonicated using a water bath sonicator (Prima, UK, model PM3-900TL) during and within 15 minutes after injection. Thereafter, sonication was continued for 10 minutes at 50% output under ice bath conditions using a probe-type sonicator (Qsonica, usa, model Q700). The liquid was repeatedly extruded through a polycarbonate filter (pore size: 100nm, Whatman, USA) 3 times by means of a high-pressure extruder (Northern Lipid, USA) to obtain a preparation having an average particle size of about 100 nm.
(2) 0.6 mu Mol of tumor targeting molecule var7 polypeptide (var7 can be targeted to the tumor site by virtue of the acidic microenvironment of the tumor, the amino acid sequence is ACEEQNPWARYLEWLFPTETLLLEL, the detailed information is described in the literature: Mol Imaging Biol (2016)18:686-696), and the thiol group of var7 polypeptide is coupled with Maleimide by stirring at 4 ℃ for 24 hours.
(3) To remove doxorubicin that was not encapsulated in the siliceous preparation and var7 that was not attached to the surface of the siliceous body, the preparation obtained in step (2) was first filtered through a 0.45 μm filter and free doxorubicin and var7 were removed using exclusion chromatography column Sephadex G-50.
(4) Using the same assay method as in example 1, the assay result was 5.9mg of perfluorohexane and 1.6mg of doxorubicin per 10mg of lyophilized silica.
Example 4 preparation of a silica-entrapped fluorocarbon formulation with tumor targeting molecules
(1) Dissolving 9.5 μmol of complex lipid B, 0.2 μmol of DSPE-mPEG5000, 2 μmol of small molecule drug porfimer sodium, 20 μ L of perfluorooctyl bromide and 0.3 μmol of DSPE-Maleimide (distearoyl phosphatidyl ethanolamine modified Maleimide) in 2mL of ethanol, injecting into 20mL of PBS, and subjecting the 20mL of PBS to ultrasonic treatment by using a water bath ultrasonic instrument (Prima corporation, UK, model PM3-900TL) during and within 15 minutes after injection. The liquid was repeatedly extruded through a polycarbonate filter (10 μm in pore size, Whatman, USA) 3 times by means of a high-pressure extruder (northern lipid, USA) to obtain a preparation having a particle size of about 10 μm.
(2) 0.6 mu mol of tumor targeting molecule SH-PEG5000-Folate (sulfhydryl-polyethylene glycol 5000-folic acid) is added into the preparation, and the mixture is stirred for 24 hours at 4 ℃ so that the sulfhydryl of the SH-PEG5000-Folate is coupled with Maleimide.
(3) In order to remove porfimer sodium not encapsulated in the siliceous preparation and tumor targeting molecules not attached to the surface of the siliceous body, the preparation obtained in step (2) was repeatedly washed three times with 50% ethanol and stored in PBS for future use.
(4) The same test method as in example 1 was used, and the test results showed that 7.3mg of perfluorooctyl bromide and 0.7mg of porfimer sodium were contained per 10mg of the lyophilized silica.
Example 5 preparation of Liposome Encapsulated fluorocarbon formulations consisting of common Phospholipids
(1) Mu.mol Distearylphosphatidylcholine (DSPC), 0.2. mu. mol DSPE-mPEG2000, 3.0. mu. mol cholesterol, 2.0. mu. mol small molecule drug doxorubicin, 20. mu.L perfluorohexane, 0.3. mu. mol DSPE-mPEG2000-Maleimide were dissolved in 2mL ethanol and injected into 20mL PBS, and during and within 15 minutes after injection, 20mL PBS was sonicated using a water bath sonicator (Prima, UK, model PM3-900 TL). Thereafter, sonication was continued for 10 minutes at 50% output under ice bath conditions using a probe-type sonicator (Qsonica, usa, model Q700). The liquid was repeatedly extruded through a polycarbonate filter (pore diameter: 100nm, Whatman, USA) 3 times by means of a high-pressure extruder (Northern Lipid, USA) to obtain a preparation having an average particle diameter of about 100 nm.
(2) 0.6 mu mol of tumor targeting molecule var7 polypeptide is added into the preparation, and the mixture is stirred for 24 hours at 4 ℃ so that the sulfhydryl of var7 polypeptide is coupled with Maleimide.
(3) To remove doxorubicin that was not encapsulated in the formulation and var7 that was not attached to the surface of the liposomes, the formulation obtained in step (2) was first filtered through a 0.45 μm filter and free doxorubicin and var7 were removed using exclusion chromatography column Sephadex G-50.
(4) Using the same examination method as in example 1, it was determined that 5.3mg of perfluorohexane and 1.4mg of doxorubicin were contained per 10mg of the lyophilized liposomes.
Example 6 evaluation of drug-loaded stability of silica-encapsulated fluorocarbon formulations
(1) The surfactant Triton-100 solubilization method was used to compare the drug loading stability of different fluorocarbon formulations. The fluorocarbon formulations prepared in example 3 and example 5 were dispersed in Triton-100 solutions of different concentrations, respectively.
(2) After shaking for 10 minutes at room temperature, sampling and detecting the fluorescence intensity of the adriamycin, detecting the drug amount of the adriamycin released from the preparation by a fluorescence quenching method, and calculating the percentage of the released adriamycin in the total adriamycin in the preparation, wherein the formula is as follows:
Figure BDA0002745855000000121
wherein F (t) is the fluorescence intensity of doxorubicin of the Triton-100-treated sample, F 0 Is the fluorescence intensity of doxorubicin of the control sample not treated with Triton-100, whereas Fc is the doxorubicin of five samples after complete destruction by 1% Triton-100 and 1M hydrochloric acidMean fluorescence intensity of the mycin. The fluorescence intensity of the adriamycin is detected by a fluorescence spectrophotometer, the detected excitation wavelength is 480nm, and the detected emission wavelength is 580 nm.
As shown in FIG. 3, with the increase of Triton-100 concentration, doxorubicin in the liposome-entrapped fluorocarbon formulation composed of common phospholipids was released in a large amount, but only a small amount of doxorubicin in the liposome-entrapped fluorocarbon formulation was released. The result shows that the carbon fluoride coated by the silica liposome has higher stability than the carbon fluoride coated by the liposome and can resist the solubilization of the surfactant.
Example 7 evaluation of the drug Release ability of silica-encapsulated fluorocarbon formulations under high intensity focused ultrasound control
(1) The silica-encapsulated fluorocarbon preparation prepared in example 3 was dispersed in PBS at 37 ℃ and treated with high intensity focused ultrasound for 4 minutes with a Pulse Width Modulation (PWM) of 2%. At various times during sonication, the solution was sampled and immediately cooled on ice to stop drug release.
(2) The release rate of the drug is detected by a fluorescence quenching method, and the percentage of released adriamycin in the total adriamycin in the preparation is calculated by the following formula:
Figure BDA0002745855000000122
where F (t) is the fluorescence intensity of doxorubicin for the sample after high intensity focused ultrasound treatment, cooled on ice and equilibrated to room temperature, F0 is the fluorescence intensity of doxorubicin for the control sample without high intensity focused ultrasound treatment, and Fc is the average fluorescence intensity of doxorubicin for five samples after extensive destruction by 1% Triton-100 and 1M hydrochloric acid. The fluorescence intensity of the adriamycin is detected by a fluorescence spectrophotometer, the detected excitation wavelength is 480nm, and the detected emission wavelength is 580 nm.
The detection result is shown in figure 4, and the carbon fluoride preparation encapsulated by the silica can rapidly release the adriamycin under the treatment of high-intensity focused ultrasound. The high-intensity focused ultrasound can trigger the preparation to release the drug, so that the controlled release of the drug is realized.
Example 8 evaluation of oxygen delivery and controlled release capabilities of silica-encapsulated fluorocarbon formulations
(1) In a sterile oxygen chamber (O) 2 Flow rate 5L/min), 1mL of the preparation prepared in example 3 was oxygenated for 1 minute and then rapidly injected into 4mL of PBS.
(2) The dissolved oxygen concentration of 4ml pbs was monitored using a portable dissolved oxygen meter (Rex, model JPBJ-608, china) before and after injection. 1mL of oxygenated or non-oxygenated PBS containing no silica preparation was injected into 4mL PBS to serve as a control.
(3) The time of injection was defined as 0 minutes, and the injected solution was irradiated with high intensity focused ultrasound (PWM ═ 2%) for 2 minutes at 20, 30, 40, and 50 minutes after injection, respectively, to trigger the release of oxygen from the formulation. The oxygen concentration was recorded using an oxygen dissolver for a total of about 60 minutes.
As shown in FIG. 5, after 1mL of oxygenated preparation was injected into 4mL of PBS, the oxygen content of PBS increased rapidly and the preparation released oxygen rapidly each time it was treated with high intensity focused ultrasound, forming a prominent "fluctuating peak" in the oxygen concentration profile, indicating that the preparation was able to carry oxygen efficiently and that high intensity focused ultrasound triggered the release of oxygen.
Example 9 evaluation of ultrasound imaging Effect and carbon fluoride controlled Release Effect of silica-Encapsulated carbon fluoride preparations
(1) Establishing a mouse subcutaneous tumor model: 4T1 cells were administered at 5X 10 per mouse 6 The amount of each cell is inoculated to the subcutaneous side of the thigh of a BALB/c mouse, and the mouse is bred in an SPF level environment;
(2) tumor size up to about 100mm 3 200 μ L of the formulation prepared in example 3 was injected via tail vein at a dose of 5mg/kg of doxorubicin;
(3) the tumor site was treated with high intensity focused ultrasound (PWM ═ 2%) irradiation, and ultrasound imaging was performed before the high intensity focused ultrasound treatment and after different durations of continuous irradiation, respectively. Ultrasound images of the tumor site were acquired using a Resona 7 ultrasound system (Mindray, China) equipped with an L12-3E ultrasound probe (3-12MHz) with a Mechanical Index (MI) set at 0.044.
The detection result is shown in figure 6, and after the preparation is injected, the ultrasonic signal of the tumor area is not obviously changed. However, after the injection of the formulation and the treatment of the tumor with High Intensity Focused Ultrasound (HIFU), the ultrasound signal of the tumor becomes stronger with the increase of the HIFU treatment time, and finally, a significantly enhanced ultrasound signal appears in the whole tumor area, which indicates that the treatment of HIFU can convert the carbon fluoride formulation into bubbles to generate the ultrasound signal. This also suggests that HIFU treatment may facilitate the release of carbon fluoride from the formulation.
Example 10 evaluation of drug-loaded stability of formulations consisting of different kinds of fluorocarbons
Formulations consisting of perfluoropentane, perfluorohexane, perfluorooctane bromide and perfluorocrown ether, respectively designated D-PFP-vPCs, D-PFH-vPCs, D-PFOB-vPCs, D-PFCE-vPCs, were prepared according to the method described in example 3; a preparation of perfluorohexane, designated D-PFH-vPLs, encapsulated with conventional phospholipids was prepared according to the procedure described in example 5.1 mL of this formulation was dispersed in 10mL of fetal bovine serum at 37 ℃ with constant stirring (500 rpm), simulating the environment in which different formulations would be in the blood circulation system in vivo, during which time the solution was sampled at different times and the amount of doxorubicin-releasing drug from the formulation was determined using the fluorescence dequenching method described in example 6.
The results are shown in FIG. 7, where D-PFP-vPCs leaked about 50% of doxorubicin while the other three D-vPCs leaked only < 10% of doxorubicin within 48 hours, indicating poor drug loading stability of the formulation consisting of perfluoropentane. The doxorubicin leakage rate of D-PFH-vPLs is far higher than that of D-PFH-vPCs, and the doxorubicin leakage rate is respectively 78.5% and 8.4%, which shows that the preparation has better drug-loading stability compared with a common phospholipid-loaded perfluorohexane preparation.
Example 11 evaluation of the in vitro ultrasound contrast Effect of formulations consisting of different types of carbon fluoride
Formulations consisting of different types of fluorocarbons prepared in the previous example 10 were loaded in latex tubes, immersed in water at 37 c, and examined for ultrasonic signals before and after HIFU treatment using a 2% PWM (Pulse-Width Modulation) power using a Resona 7 ultrasonic imaging system (Mindray, china) using an L12-3E ultrasonic examination probe (3-12MHz) with a Mechanical Index (MI) of 0.044.
As shown in fig. 8, the formulations of four fluorinated carbons did not have significant ultrasound signals without HIFU pretreatment. After 3 minutes of HIFU (PWM ═ 2%) pretreatment, obvious ultrasonic signals appear in D-PFP-vPCs and D-PFH-vPCs. However, the ultrasonic signals of D-PFOB-PCs and D-PFCE-vPCs are not obvious. Therefore, after HIFU pretreatment, only the formulation consisting of perfluoropentane and perfluorohexane had ultrasound contrast ability, and the formulation consisting of perfluorooctyl bromide and perfluorocrown ether had no ultrasound contrast ability.
Example 12 evaluation of the therapeutic Effect of the preparations on the cellular level
4T1 cells were seeded into 24-well plates (10 per well) 4 One cell) filled with 1% O 2 The cells are induced to enter a hypoxic state by culturing in a hypoxic incubator for 24 hours. The D-PFH-vPCs preparation prepared according to the method of example 3 was dispersed in 2 mlpmi-1640 medium (final concentration of doxorubicin 5 μ g/mL), saturated with nitrogen or oxygen, respectively (filtration of nitrogen or oxygen through a 0.22 μm gas filter), added to the cells, immediately treated with HIFU (PWM ═ 2%) for 3 minutes, incubated for 4 hours, the cells were washed three times with the medium, cultured for further 24 hours, and the survival rate of the cells was examined using CCK-8 kit (Dojindo, japan).
The results are shown in fig. 9, and the experimental group "the preparation saturated with nitrogen + HIFU treatment" has lower survival rate of tumor cells than the experimental group "the preparation saturated with nitrogen", which indicates that HIFU can promote the preparation to release adriamycin. Compared with the experimental group of 'the preparation saturated with oxygen and the HIFU treatment', the survival rate of the tumor cells is lower, which shows that the oxygen carried by the preparation can improve the hypoxia state of the tumor cells and reduce the survival rate of the tumor cells. And the anti-tumor effect of the experimental group 'oxygen-saturated preparation + HIFU treatment' is much higher than that of the experimental group 'free adriamycin'.
The present inventors further investigated the effect of the relevant treatment methods on the expression levels of the relevant genes and proteins (HIF-1. alpha., MDR1 and P-gp) of tumor cell multidrug resistance using PFH-vPCs without doxorubicin in nitrogen or oxygen saturation conditions, using PBS as a control (relative expression level of MDR1 gene was quantified by RT-qPCR, relative expression levels of HIF-1. alpha. and P-gp proteins were quantified by western blotting). As a result, it was found that: cell passage through vPCs (O) 2 ) After + HIFU treatment, the expression levels of all three markers were significantly reduced, while vPCs (N) 2 ) + HIFU or PBS (O) 2 ) + HIFU had no significant effect on the expression levels of the three markers. The results show that PFH-vPCs are capable of carrying oxygen and that PFH-vPCs (O) 2 ) The + HIFU can effectively reduce the expression level of the genes and proteins related to the multiple drug resistance of the tumor cells after relieving the tumor hypoxia.
Example 13 evaluation of the formulations for improvement in vivo drug distribution
4T1 tumors were implanted subcutaneously in the hind legs of BALB/c mice on both sides until the tumor volumes increased to 100mm 3 In this case, the preparation (D-vPCs) prepared in example 3, the perfluorohexane preparation (D-vPLs) coated with common phospholipid prepared in example 5, Free doxorubicin (Free DOX), and doxorubicin liposome were injected intravenously, respectively
Figure BDA0002745855000000151
The dose was 5mg of doxorubicin per kg of mouse body weight. Immediately after injection, tumors on one side were sonicated with HIFU (PWM ═ 2%) for 15 minutes. After 24 hours, the mice were sacrificed, the tumors and major organs were collected, weighed, 0.5mL of PBS was added per 50mg of tissue, and after clipping, thoroughly homogenized using an electric tissue homogenizer. To 100. mu.L of the homogenate was added 900. mu.L of precipitation buffer (containing 800. mu.L of isopropanol, 100. mu.L of 10 vol% Triton-100, and 100. mu.L of water). After standing at 4 ℃ for 24 hours, 12000g of the mixture was centrifuged, and the fluorescence intensity of doxorubicin in the supernatant was measured by a multifunctional microplate reader. Dispersing doxorubicin in a known concentration in a homogenate of mouse tissue not subjected to the administration treatment, and detecting doxorubicin by the same treatment as described aboveFluorescence intensity, a standard calibration curve for each tissue was prepared. The tissue content of doxorubicin was calculated from the corresponding standard calibration curve and the final result was expressed as the percentage of doxorubicin content per gram of tissue (ID%/g) of the total dose of doxorubicin injected.
As a result, as shown in FIG. 10, the tumors on one side treated with HIFU had higher adriamycin uptake (34.2vs 1.2% ID/g) than the tumors on the other side not treated with HIFU after the injection of D-vPCs, which indicates that D-vPCs have good HIFU responsiveness. The experimental group "D-vPCs + HIFU" had the highest tumor doxorubicin uptake, 16.3 times (34.2vs 2.1% ID/g) that of the experimental group "Free DOX", and was the experimental group
Figure BDA0002745855000000161
8.8 times higher (34.2vs 3.9% ID/g).
Myocardial damage is one of the most serious side effects of doxorubicin. Although in contrast to Free DOX,
Figure BDA0002745855000000167
the myocardial uptake of doxorubicin was somewhat reduced, however, the experimental group "D-vPCs + HIFU" had a lower myocardial uptake, which is compared with
Figure BDA0002745855000000162
Compared with the prior art, the reduction is 75.0% (0.3vs 1.2% ID/g); compared with D-vPLs, a reduction of 81.3% (0.3vs 1.6% ID/g) was achieved. With the Free DOX, the process is simple,
Figure BDA0002745855000000163
compared with D-vPLs, the doxorubicin intake in the kidney and lung of the mice treated with the D-vPCs + HIFU is also lowest.
EXAMPLE 14 evaluation of the in vivo chemotherapeutic Effect of the formulations
Preparing BALB/c mouse with 4T1 tumor until the tumor volume grows to about 100mm 3 On the left and right, mice were randomly divided into nine groups of 5 mice each: (1) PBS; (2) vPCs (O) 2 )+HIFU;(3)Free DOX;(4)
Figure BDA0002745855000000164
(5)D-vPCs;(6)D-vPLs+HIFU;(7)D-vPCs+HIFU;(8)D-PCs+HIFU;(9)D-vPCs(O 2 ) + HIFU. D-vPCs and vPCs are doxorubicin-loaded and non-doxorubicin-loaded formulations modified with tumor targeting molecule var7, respectively, prepared according to the method of example 3, D-PCs are doxorubicin-loaded, non-modified tumor targeting molecule var7, prepared according to the method of example 3, and D-vPLs are common phospholipid-loaded perfluorocarbon formulations (containing doxorubicin), prepared according to example 5. For groups (2) and (9), "(O) 2 ) "means that the mice are given hyperoxic respiratory treatment: starting 5 minutes before injection, mice were fitted with an oxygen breathing mask and continuously breathed with high concentration oxygen for 20 minutes. All groups of the administered agents were dispersed in PBS and injected into mice via tail vein in a volume of 200. mu.L per mouse. For the group treated with HIFU sonication, the tumor was sonicated with HIFU (PWM ═ 2%) immediately after injection to trigger the release of oxygen and DOX. For groups (1) and (2) containing no DOX, the drugs were injected twice a day from day 0 to day 9, once in the morning and once in the evening for 10 days; for the other doxorubicin-containing groups, the injections were given once daily at 5mg/kg on days 0, 2, 4, 6, and 8, respectively. For group (9), "D-vPCs (O) were administered once daily on days 0, 2, 4, 6, 8 in the morning 2 ) + HIFU "given once a night" vPCs (O) 2 ) + HIFU "and" vPCs (O) twice daily on days 1, 3, 5, 7, 9 2 ) + HIFU "once in the morning and once in the evening, thus, it is ensured that the mice of group (9) and group (2) receive the same number of hyperoxic breaths. Tumor volumes were measured every two days. The tumor volume is calculated by V ═ L × W 2 ) L and W are the length and width of the tumor, respectively. At the end of treatment (day 16), the tumor growth inhibition rate was calculated by the formula: (tumor volume in PBS group-experimental group)/tumor volume in PBS group.
As shown in FIG. 11, first, vpCs (O) 2 ) The + HIFU had no significant effect on tumor growth, indicating to some extent the good biological safety of vPCs and HIFU. Free DOX and
Figure BDA0002745855000000165
with similar tumor growth inhibition rate (20.6% vs 25.77%). The tumor treatment effect of the D-vPCs + HIFU is far better than that of the D-vPCs, Free DOX,
Figure BDA0002745855000000166
The tumor growth inhibition rate is improved to 83.2 percent. D-vPCs + HIFU also showed better tumor growth inhibition than D-PCs + HIFU, D-vPLs + HIFU (P-PCs + HIFU)<0.05), because the targeting property of the tumor is increased compared with that of the D-PCs, and the D-vPCs have better drug-loading stability compared with that of the D-vPLs.
By improving tumor hypoxia, and relieving hypoxia-mediated multidrug resistance, D-vPCs (O) 2 ) + HIFU has a higher antitumor effect than D-vPCs + HIFU. With Free DOX,
Figure BDA0002745855000000171
D-vPCs + HIFU in comparison to D-vPCs (O) 2 ) The tumor growth inhibition rate of the + HIFU is as high as 94.6%, which is respectively improved by 3.59, 2.67 and 0.14 times (P) compared with the former three<0.0001)。
Example 15 evaluation of the Effect of the formulations on reducing tumor metastasis
4T1 cells (4T1-Luciferase) transfected with the Luciferase gene were prepared. BALB/c mice bearing 4T1-Luciferase subcutaneous tumors were prepared until the tumor volume grew to approximately 100mm 3 On the left and right, mice were randomly divided into 7 groups of 5 mice: (1) PBS; (2) vPCs; (3) vPCs (O) 2 )+HIFU;(4)Free DOX;(5)
Figure BDA0002745855000000172
(6)D-vPCs+HIFU;(7)D-vPCs(O 2 ) + HIFU. D-vPCs and vPCs are doxorubicin-entrapped and doxorubicin-unencapsulated formulations of modified tumor targeting molecule var7, respectively, prepared according to the method of example 3; "(O) 2 ) "means that the mice are given hyperoxic respiratory treatment. Mice were treated by the dosing method described in example 14. The transfer and distribution of 4T1 cells in vivo can be dynamically monitored by bioluminescence imaging of luciferase. At the end of the in vivo treatment experiment (day 16), IVIS was usedThe Spectrum small animal in vivo imaging system (PerkinElmer, usa) observed the degree of lung metastasis of different groups of tumors: mice were injected intraperitoneally with 150mg/kg of D-fluorescein potassium salt and imaged 15 minutes after injection.
Results as shown in figure 12, spontaneous lung metastases occurred in all five mice in the PBS group (group 1) at day 16 of the in vivo treatment experiment. By Free DOX (group 4) or
Figure BDA0002745855000000173
Five administrations (group 5) showed a significant increase in lung metastases, although the growth of orthotopic tumors was inhibited to some extent. Via vPCs (O) 2 ) Multiple treatment with + HIFU (group 3), the degree of lung metastasis was significantly reduced compared to PBS group (group 1), indicating that vPCs (O) 2 ) + HIFU can inhibit lung metastasis of the carcinoma in situ by reducing hypoxia of the carcinoma in situ. The D-vPCs + HIFU group (group 6) had significantly lower lung metastasis than the PBS group (group 1). D-vPCs (O) 2 ) The extent of lung metastasis with + HIFU (group 7) was lower than with D-vPCs + HIFU (group 6), and no significant lung metastasis was observed in all five mice, suggesting that this formulation may improve the hypoxic extent of the carcinoma in situ, inhibiting spontaneous lung metastasis of the tumor.
Example 16 evaluation of the therapeutic efficacy of formulations as a novel adjunctive chemotherapeutic approach
BALB/c mice bearing 4T1 subcutaneous tumors were prepared until the tumor volume grew to about 100mm 3 On the left and right, the mice were randomly divided into four groups of 6 mice each, which were treated as follows: (group 1) PBS → HT; (group 2) Free DOX → HT; (group 3)
Figure BDA0002745855000000174
→ HT; (group 4) D-vPCs (O) 2 ) + HIFU → HT. HT stands for hyperthermia: tumors were irradiated with HIFU (PWM ═ 8%) for 15 minutes. HT was given on day 0 and chemotherapy was given once on days minus 4 and minus 2, respectively. Four groups of mice were given PBS, Free DOX,
Figure BDA0002745855000000181
and D-vPCs (O) 2 ) + HIFU, the drug is injected via the tail vein, the drugThe dose was 5mg/kg doxorubicin and the injection volume was 200. mu.L, where D-vPCs (O) 2 ) The + HIFU processing method is as described in embodiment 14: mice were injected with D-vPCs (prepared from example 3) via tail vein with high oxygen inhalation and tumor sites were irradiated with HIFU (PWM ═ 2%) for 15 minutes.
As a result, as shown in FIG. 13, since H-HIFU-mediated hyperthermia itself failed to eradicate all tumor cells, tumors of all mice rapidly recurred through group 1 of PBS pretreatment (days-4 and-2) and hyperthermia treatment (day 0). With Free DOX or
Figure BDA0002745855000000182
In groups 2 and 3, which had been pretreated with chemotherapy (days-4 and-2) and heat treatment (day 0), the recurrence rate of the tumors decreased to 66.7% and 50.0%, respectively. By D-vPCs (O) 2 ) + HIFU group 4 with chemotherapy pretreatment and hyperthermia treatment, no tumor recurrence was observed for the next 60 days. Thus, with Free DOX and
Figure BDA0002745855000000183
in contrast, D-vPCs (O) 2 ) + HIFU is more suitable as a neoadjuvant chemotherapy (chemotherapy pretreatment) means to suppress tumor recurrence after hyperthermia.

Claims (23)

1. A silicon plastid entrapping a carbon fluoride compound, wherein the silicon plastid is a particle with a core-shell structure, the shell is a single-layer lipid molecule layer consisting of a silicon plastid-forming lipid and a phospholipid component without Si, and the silicon plastid-forming lipid forms a silicate network structure in a form of-Si-O-Si-on the outer surface of the shell; the core contains a carbon fluoride compound;
based on the total mole amount of the monolayer liposome layer, the mole percentage of the liposome forming liposome is 50-100%, and the mole percentage of the phospholipid component without Si is 0-50%;
the carbon fluoride compound has a boiling point of 40-80 ℃ under normal pressure;
the Si-free phospholipid component is selected from: polyethyethylene of distearoylphosphatidylethanolamineDiol derivative DSPE-mPEG 1000~20000 Maleimide-modified distearoylphosphatidylethanolamine, polyethylene glycol derivative DSPE-mPEG of Maleimide-modified distearoylphosphatidylethanolamine 1000~20000 One or more of dipalmitoylphosphatidylcholine, 1-myristoyl-2-stearoyl lecithin, hydrogenated soybean lecithin, distearoylphosphatidylcholine; the siliceous plastid forming lipid is selected from the group consisting of complex lipids a-D:
Figure FDA0003534536650000011
the preparation method of the silica body comprises the following steps: adding carbon fluoride compound, formed siliceous liposome and phospholipid component without Si or further containing medicine into buffer solution or water, homogenizing, and high-pressure extruding to obtain uniform particle size distribution.
2. The siliceous body of claim 1, wherein said non-Si containing phospholipid component is selected from the group consisting of DSPE-mPEG2000, DSPE-mPEG5000, DSPE-Maleimide, DSPE-mPEG 2000 -one or more of Maleimide, dipalmitoylphosphatidylcholine, 1-myristoyl-2-stearoyl lecithin, hydrogenated soy lecithin or distearoylphosphatidylcholine.
3. The silica body of claim 1 wherein the fluorocarbon-based compound is selected from the group consisting of perfluorohexane, perfluoroheptane, dodecafluorohexane, and bromododecafluorohexane.
4. The silica body according to any one of claims 1-3, wherein the surface of the silica body is modified with a tumor targeting molecule selected from any one or more of an antibody, a polypeptide, an aptamer, folic acid or a folic acid derivative for targeting a tumor.
5. The silica body of claim 4 wherein said tumor targeting molecule is var7 polypeptide having an amino acid sequence of ACEEQNPWARYLEWLFPTETLLLEL.
6. The silica body of claim 4, wherein the tumor targeting molecule is a thiol-polyethylene glycol 2000~6000 -folic acid.
7. The siliceous body according to any one of claims 1 to 3, which has a particle size of 10nm to 1 μm.
8. The silica body of any one of claims 1-3, wherein the core of the silica body comprises a water-insoluble or hydrophobic small molecule drug.
9. The silica plastid of claim 8, wherein said small molecule drug is selected from the group consisting of one or more chemotherapeutic drugs, protein kinase inhibitors, photodynamic drugs.
10. The siliceous body of claim 8, wherein said small molecule drug is selected from the group consisting of doxorubicin, cisplatin, paclitaxel, irinotecan, sorafenib, gefitinib, porfimer sodium, verteporfin, indocyanine green.
11. A method for the preparation of a silica body according to any of claims 1-10, characterized in that the fluorocarbon-based compound, the lipid forming silica body and the Si-free phospholipid component, or the further included drug, are dissolved in an organic solvent prior to the addition of the buffer solution or water.
12. The method of claim 11, wherein the organic solvent is selected from the group consisting of alcohols and dimethylsulfoxide.
13. The method according to any one of claims 11 to 12, wherein the pH of the buffer solution is 5.0 to 8.0.
14. The method according to any one of claims 11 to 12, wherein the ratio of the amount of the drug to the amount of the phospholipid component is 1:10 to 1:2 by mass.
15. The method of any one of claims 11-12, wherein the preparation of the silica bodies is modified with a tumor targeting molecule after the preparation is obtained.
16. Use of a silica body according to any one of claims 1 to 10 in the manufacture of a carrier or medicament for delivery of oxygen and/or a medicament.
17. The use according to claim 16, wherein the medicament is a tumor therapy medicament.
18. Use according to claim 16 or 17, wherein the medicament is in combination with HIFU.
19. Use according to claim 16 or 17, characterized in that the medicament is used as a neoadjuvant chemotherapy medicament for hyperthermia.
20. The use of claim 16 or 17, wherein the medicament ameliorates tumor hypoxia.
21. The use according to claim 16 or 17, wherein the medicament reduces tumor metastasis.
22. The use according to claim 17, wherein said tumor is breast cancer.
23. Use of a silica body as claimed in any one of claims 1 to 10 in the preparation of an ultrasound imaging agent.
CN202011166195.9A 2020-10-27 2020-10-27 Perfluorocarbon silicon plastid and preparation method and application thereof Active CN112546062B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011166195.9A CN112546062B (en) 2020-10-27 2020-10-27 Perfluorocarbon silicon plastid and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011166195.9A CN112546062B (en) 2020-10-27 2020-10-27 Perfluorocarbon silicon plastid and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN112546062A CN112546062A (en) 2021-03-26
CN112546062B true CN112546062B (en) 2022-08-19

Family

ID=75042612

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011166195.9A Active CN112546062B (en) 2020-10-27 2020-10-27 Perfluorocarbon silicon plastid and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN112546062B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113332619B (en) * 2021-05-28 2023-06-20 西安交通大学 Ultrasonic conformal activation and monitoring imaging system for phase-change nano-droplet drug carrier
CN114732796B (en) * 2022-02-18 2023-01-17 北京大学第三医院(北京大学第三临床医学院) Double-targeting drug-loaded microbubble and preparation method and application thereof
CN116603101B (en) * 2023-07-20 2023-10-03 北京大学第三医院(北京大学第三临床医学院) Gold-siliceous body-polycaprolactone scaffold system and preparation method and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105194678A (en) * 2014-06-20 2015-12-30 北京三星通信技术研究有限公司 Thermosensitive silica body and preparing method and application thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105194678A (en) * 2014-06-20 2015-12-30 北京三星通信技术研究有限公司 Thermosensitive silica body and preparing method and application thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Design and Synthesis of Lipidic Organoalkoxysilanes for the Self-Assembly of Liposomal Nanohybrid Cerasomes with Controlled Drug Release Properties;Xiaolong Liang等;《Chem. Eur. J.》;20131231;第19卷;摘要、第16114页Figure 1 *
Nanohybrid Liposomal Cerasomes with Good Physiological Stability and Rapid Temperature Responsiveness for High Intensity Focused Ultrasound Triggered Local Chemotherapy of Cancer;Xiaolong Liang等;《ACS NANO》;20151011;第9卷(第2期);第1281页右栏Figure 1、最后一段,第1282页左栏第1段,第1289页右栏第1段 *
蒋灵冬;蒋灵冬;《中国优秀硕士学位论文全文数据库 医药卫生科技辑》;20150215(第2期);E080-13 *

Also Published As

Publication number Publication date
CN112546062A (en) 2021-03-26

Similar Documents

Publication Publication Date Title
CN112546062B (en) Perfluorocarbon silicon plastid and preparation method and application thereof
Zhu et al. CaCO 3-assisted preparation of pH-responsive immune-modulating nanoparticles for augmented chemo-immunotherapy
He et al. Tumor hypoxia relief overcomes multidrug resistance and immune inhibition for self-enhanced photodynamic therapy
JP6389539B2 (en) Porphyrin nanovesicle
EP2328554B1 (en) Encapsulation of photosensitizers in nano-emulsions
CN108354901A (en) For chemotherapy of tumors and the pH/ of photo-thermal combination therapy reduction Dual Sensitive multifunctional nano micellas and its application
CN108653733B (en) Polymer vesicle of double-loaded anthracycline drug and photosensitizer with bubble generation function and preparation method thereof
CN110755613A (en) Preparation and application of light-triggered erythrocyte membrane-coated NO nano bionic donor material
CN111760024B (en) Permeation enhanced gold nanocluster drug-loaded targeting preparation and preparation method and application thereof
WO2019127297A1 (en) Tetravalent platinum compound-bicyclic double bond amphiphilic polymer prodrug, and nanomicelle, preparation method, and application thereof
Kv et al. Tumor microenvironment-responsive and oxygen self-sufficient oil droplet nanoparticles for enhanced photothermal/photodynamic combination therapy against hypoxic tumors
KR101686145B1 (en) Composition for Phototherapy of Cancer Comprising Complex of Liposome And Indocyanine Green
CN110665003A (en) Double-drug-loading carrier-free nanoparticle and preparation method thereof
Xie et al. Photosensitizer-loaded gold nanocages for immunogenic phototherapy of aggressive melanoma
CN112316138A (en) PCP (prestressed concrete Polymer) targeted modified black phosphorus nanoparticle and preparation method and application thereof
Huang et al. Synergistic strategy with hyperthermia therapy based immunotherapy and engineered exosomes− liposomes targeted chemotherapy prevents tumor recurrence and metastasis in advanced breast cancer
Li et al. An intelligent responsive macrophage cell membrane-camouflaged mesoporous silicon nanorod drug delivery system for precise targeted therapy of tumors
CN110179754A (en) With redox responsiveness and the multi-functional liposome of tissue infiltration can be enhanced
CN112618514B (en) Ammonia borane/hollow mesoporous polydopamine/polyethylene glycol nano composite particle and preparation and application thereof
CN114904014A (en) Self-oxygen-generating bionic photodynamic/iron death/immunosuppression microenvironment regulation nano platform and preparation and application thereof
CN106166141A (en) A kind of Multifunctional composite nanometer medicine for tumor imaging and treatment and preparation method thereof
Ramedani et al. Hybrid ultrasound-activated nanoparticles based on graphene quantum dots for cancer treatment
CN114209829B (en) Photothermal liposome loaded with fluorescent dye, and preparation method and application thereof
Tian et al. Programmed nanoreactors Boost Immune Response through ROS Cascade amplification along with RNS storm
Su et al. Photo-responsive NIR-II biomimetic nanomedicine for efficient cancer-targeted theranostics

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