CN113599525A - Anti-tumor nano-drug and preparation method and application thereof - Google Patents

Anti-tumor nano-drug and preparation method and application thereof Download PDF

Info

Publication number
CN113599525A
CN113599525A CN202110859262.3A CN202110859262A CN113599525A CN 113599525 A CN113599525 A CN 113599525A CN 202110859262 A CN202110859262 A CN 202110859262A CN 113599525 A CN113599525 A CN 113599525A
Authority
CN
China
Prior art keywords
drug
tumor
nano
tumor nano
preparation
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
CN202110859262.3A
Other languages
Chinese (zh)
Inventor
赵静
黄秀玉
邱妙娟
李宾宾
刘鹏
何裕隆
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.)
Seventh Affiliated Hospital Of Sun Yat Sen University Shenzhen
Original Assignee
Seventh Affiliated Hospital Of Sun Yat Sen University Shenzhen
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 Seventh Affiliated Hospital Of Sun Yat Sen University Shenzhen filed Critical Seventh Affiliated Hospital Of Sun Yat Sen University Shenzhen
Priority to CN202110859262.3A priority Critical patent/CN113599525A/en
Publication of CN113599525A publication Critical patent/CN113599525A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/662Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
    • A61K31/663Compounds having two or more phosphorus acid groups or esters thereof, e.g. clodronic acid, pamidronic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic 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
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/644Transferrin, e.g. a lactoferrin or ovotransferrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery

Abstract

The invention relates to the technical field of tumor chemotherapy, in particular to an anti-tumor nano-drug and a preparation method and application thereof. The anti-tumor nano-drug comprises a diphosphate, a tumor chemotherapeutic drug and at least one metal ion. The nano-particles are assembled by adding the diphosphate and another anti-tumor drug into a salt solution. The anti-tumor nano-drug can be prepared into nano-particle freeze-dried powder and suspension, and can be used for the administration methods of intraperitoneal perfusion chemotherapy, intravenous injection chemotherapy, oral administration, enema and the like of tumor patients. The anti-tumor nano-drug has the dual effects of anti-angiogenesis and promoting tumor apoptosis, and can be simultaneously administered with other chemotherapeutic drugs or antibodies.

Description

Anti-tumor nano-drug and preparation method and application thereof
Technical Field
The invention relates to the technical field of tumor chemotherapy, in particular to an anti-tumor nano-drug and a preparation method and application thereof.
Background
The chemotherapy medicine is a medicine for treating tumor. The chemotherapy medicine can kill tumor cells. The medicines can act on different links of growth and reproduction of tumor cells to inhibit or kill the tumor cells. Chemotherapy is one of the main methods for treating tumors at present. The existing defects of tumor chemotherapy are low bioavailability and nonspecific targeting, and various drug resistances are easy to appear in the treatment process, so that tumor patients are relapsed and move to the life end point. Therefore, the development of novel antitumor drugs has great clinical significance.
The drug factors with different anti-tumor mechanisms are combined and packaged into single nano particles, so that the anti-tumor effects of the two drugs can be exerted simultaneously, the drug effect is enhanced, the drug resistance of the drugs is reduced, and the strong anti-tumor effect is achieved. Recently, researchers at the national academy of labor and technology of Massachusetts have devised a novel nanoparticle that can deliver two drugs simultaneously to the brain tumor site to enhance the therapeutic effect on glioblastoma multiforme, and have been successful in animal experiments. The study was published in journal of Nature Communications in the uk, and researchers found that the addition of transferrin to liposome nanoparticles enabled the particles to pass through the blood-brain barrier smoothly and reach the tumor site accurately while avoiding normal cells. Liposomes are understood to be hollow artificial spherical microparticles, the shell being a lipid bilayer. Researchers load the chemotherapy drug temozolomide inside liposomes, responsible for destroying the DNA of tumor cells (deoxyribonucleic acid); the outer shell is loaded with an experimental drug named "JQ-1" which is responsible for preventing tumor cells from repairing DNA damage. The two components act together to reduce drug resistance.
However, in the prior art, two drugs are loaded through liposome or nanoparticle carriers, and the method causes low drug loading rate, and the carriers also bring biological safety and immunogenicity problems.
Disclosure of Invention
The invention aims to overcome the defects of the prior art and provides an anti-tumor nano-medicament and a preparation method and application thereof.
The invention forms nano particles by self-assembling the medicine in aqueous solution, does not need a carrier and has strong anti-tumor activity.
In order to achieve the purpose, the invention adopts the technical scheme that: an anti-tumor nano-drug is provided, comprising a bisphosphonate, a tumor chemotherapeutic and at least one metal ion.
The nano-particles are assembled by adding the diphosphate and another anti-tumor drug into a salt solution. The anti-tumor nano-drug can be prepared into nano-particle freeze-dried powder and suspension, and can be used for the administration methods of intraperitoneal perfusion chemotherapy, intravenous injection chemotherapy, oral administration, enema and the like of tumor patients. The anti-tumor nano-drug has the dual effects of anti-angiogenesis and promoting tumor apoptosis, and can be simultaneously administered with other chemotherapeutic drugs or antibodies.
As a preferred embodiment of the anti-tumor nano-drug of the present invention, the bisphosphonate is alendronate, etidronate, clodronate, ibandronate or zoledronic acid.
As a preferred embodiment of the anti-tumor nano-drug of the present invention, the tumor chemotherapeutic is an alkylating agent, an anti-metabolite, an anti-cancer antibiotic, a Chinese medicine drug or a hormone drug.
As a preferred embodiment of the anti-tumor nano-drug of the present invention, the preparation method of the anti-tumor nano-drug comprises: adding alendronate sodium water solution and THZ1 hydrochloride water solution into calcium chloride solution, adjusting pH value to neutral or alkaline, stirring, centrifuging to obtain nanometer medicinal preparation, and washing with water solution and ethanol to remove free medicinal preparation to obtain ALN-Ca-THZ1 nanometer medicinal preparation for treating tumor.
As a preferred embodiment of the anti-tumor nano-drug of the present invention, the preparation method of the anti-tumor nano-drug comprises: adding alendronate sodium water solution and 5 fluorouracil water solution into calcium chloride solution, adjusting pH value to be neutral or alkaline, stirring, centrifuging to obtain nano-drug, and washing free drug with water solution and ethanol to obtain ALN-Ca-5Fu anti-tumor nano-drug.
As a preferred embodiment of the anti-tumor nano-drug of the present invention, the anti-tumor nano-drug may further modify a group targeting a tumor or a group targeting a normal tissue on the surface of the nanoparticle.
The surface of the nano-particle of the invention is provided with amino and can react with a plurality of groups, thus various tumor or normal tissue targeted groups can be further modified on the surface of the nano-particle.
As a preferred embodiment of the anti-tumor nano-drug of the present invention, the tumor targeting group is RGD polypeptide, folic acid, or transferrin.
As a preferred embodiment of the anti-tumor nano-drug of the invention, the RGD polypeptide is NHS-PEG-RGD, and the molecular weight of the NHS-PEG-RGD is 500-15000.
The invention also provides the application of the anti-tumor nano-medicament in preparing a medicament for treating cancer.
As a preferred embodiment of the use of the invention, the cancer is ovarian cancer.
The invention has the beneficial effects that:
(1) compared with other inventions, the invention does not need a drug carrier, and the diphosphate and the chemotherapeutic drug are self-assembled to form the nano-drug. The diphosphate selected by the invention has the activity of inhibiting the migration of tumor cells and can inhibit the diffusion of tumors. The drug effect can be further improved by combining with other chemotherapeutic drugs.
(2) Compared with the nano-drugs prepared by other methods, the nano-drug prepared by the invention has a large amount of amino groups on the surface, and can react with various groups, so that other functional groups are modified on the surface of the nano-drug.
(3) The invention discloses a method for self-assembling two small molecular drugs to form nano particles, and the nano drug prepared by the method is used for anti-tumor treatment, and the method comprises the following steps: has the functions of targeting tumor and effectively inhibiting tumor proliferation and metastasis. The invention constructs a mouse human ovarian cancer abdominal cavity tumor model, after solid tumors are formed in the mouse abdominal cavity, nano-drugs are injected into the mouse abdominal cavity through the abdominal cavity, the tumor development condition is monitored through a mouse living body imager, and the antitumor effect is evaluated through the mouse survival curve and the tumor size.
Drawings
FIG. 1: transmission electron microscopy pictures of sample 2; from the transmission results, the prepared sample was uniformly dispersed, and the size of the single particle was about 20 nm.
FIG. 2: x-ray photoelectron spectroscopy (XPS) pictures of sample 2; from the X-ray photoelectron spectrum, the nano-drug consists of four elements of C, O, P and Ca.
FIG. 3: ALN-Ca-THZ1 inhibits ovarian cancer cell proliferation time-concentration-dependently (n-6).
FIG. 4: a, cell scratch test result (n ═ 3); b, cell scratch experimental mobility statistical plot (n-3) p < 0.001.
FIG. 5: bioluminescence images in SKOV3 tumor-bearing mice treated with different drug groups.
Detailed Description
To more clearly illustrate the technical solutions of the present invention, the following embodiments are further described, but the present invention is not limited thereto, and these embodiments are only some examples of the present invention.
Example 1
Dissolving 0.66g of alendronate sodium in 50mL of deionized water, and marking as a solution A; dissolving 0.2g of anhydrous calcium chloride in 50mL of deionized water, and marking as a solution B; 0.033g of doxorubicin hydrochloride was dissolved in 10mL of deionized water and labeled as solution C. The solutions A, B and C were added together and labeled as solution D, and the pH of the solution D was adjusted to 5.0 with NaOH and reacted at room temperature for 2 hours. Samples were collected by centrifugation, washed 1 time with deionized water, 2 times with alcohol (100% absolute ethanol), and lyophilized to give a powder labeled ALN-Ca-DOX (sample 1).
Example 2
Dissolving 0.66g of alendronate sodium in 50mL of deionized water, and marking as a solution A; dissolving 0.2g of anhydrous calcium chloride in 50mL of deionized water, and marking as a solution B; 0.022g of THZ1 hydrochloride was dissolved in 10mL of deionized water and labeled as solution C. The solutions A, B and C were added together and labeled as solution D, and the pH of the solution D was adjusted to 5.0 with NaOH and reacted at room temperature for 2 hours. Samples were collected by centrifugation, washed 1 time with deionized water and 2 times with alcohol. And made into lyophilized powder, labeled ALN-Ca-THZ1 (sample 2).
Example 3
Dissolving 0.66g of alendronate sodium in 50mL of deionized water, and marking as a solution A; dissolving 0.2g of anhydrous calcium chloride in 50mL of deionized water, and marking as a solution B; 0.03g of 5-fluorouracil (5FU) was dissolved in 10mL of deionized water and labeled as solution C. The solutions A, B and C were added together and labeled as solution D, and the pH of the solution D was adjusted to 5.0 with NaOH and reacted at room temperature for 2 hours. Samples were collected by centrifugation, washed 1 time with deionized water and 2 times with alcohol. And made into lyophilized powder, labeled ALN-Ca-5FU (sample 3).
Example 4
Dissolving 0.66g of alendronate sodium in 50mL of deionized water, and marking as a solution A; dissolving 0.2g of anhydrous calcium chloride in 50mL of deionized water, and marking as a solution B; 0.02g oxaliplatin was dissolved in 10mL deionized water and labeled solution C. The solutions A, B and C were added together and labeled as solution D, and the pH of the solution D was adjusted to 5.0 with NaOH and reacted at room temperature for 2 hours. Samples were collected by centrifugation, washed 1 time with deionized water and 2 times with alcohol. And made into lyophilized powder, labeled ALN-Ca-Oxa (sample 4).
Example 5
10mg of ALN-Ca-THZ1 lyophilized powder prepared in example 2 was dispersed in 10mL of anhydrous ethanol and labeled as solution A, 1mg of NHS-PEG-RGD was added to solution A, and reacted for 24 hours, and the sample was collected by centrifugation and lyophilized and labeled as ALN-Ca-THZ1-RGD (sample 5).
Example 6
10mg of RGD polypeptide-polyethylene glycol-N-hydroxysuccinimide (English name: RGD-PEG2000-NHS) is dissolved in 10mL of DMSO, then 100mg of PAMAM/CaPO is added and dispersed in the solution, and the reaction is carried out for 24 hours in the dark at normal temperature. Centrifuging, dialyzing and freeze-drying to obtain the RGD modified calcium phosphate nano material, namely RGD-PEG-CaPO.
100mg of 5-fluorouracil was dissolved in 10mL of DMSO, dissolved sufficiently, and 1mg of the above RDG-PEG-CaPO was dispersed in this solution. The reaction was carried out in the dark at room temperature for 48 hours. Namely, the RGD-PEG-CaPO nano-particles loaded with 5-Fu drugs are called RGD-PEG-CaPO/5Fu for short.
Example 7ALN-Ca-THZ1 cell killing experiment
Recovering and culturing human ovarian cancer cells SKOV3 to logarithmic phase, collecting cells, adjusting cell suspension concentration after cell counting, adding 100ul McCoys 5A complete culture medium (containing 10% FBS) containing cell suspension into each well of the middle area of a 96-well plate, and ensuring that the number of cells in each well is in the range of 5000-8000 (the marginal wells are filled with sterile double distilled water). Place 96-well plate in 5% CO2Incubating at 37 deg.C until cell monolayer is fully coated on the bottom of 96-well plate, adding ALN-Ca-THZ1 with concentration gradient, and concentratingThe degrees are respectively: 100. 50, 25, 12.5, 6.25, 3.13, 1.56 and 0.78 mg/L. After the treated cells are respectively incubated for 24 hours, 48 hours and 72 hours, 10uL of CCK8 solution is added into each hole, after the cells are kept still for 2 to 4 hours, the transmission light intensity of each hole under the wavelength of 470nm is measured by a microplate spectrophotometer, the survival rate of the cells in each hole is calculated according to the measurement result, and the corresponding ALN-Ca-THZ1 concentration is plotted.
The results are shown in figure 3, ovarian cancer cells are respectively treated with ALN-Ca-THZ1 at different concentrations for 24 hours, 48 hours and 72 hours, the proliferation activity of the cells is detected by a CCK8 method, and the inhibition rate of ALN-Ca-THZ1 on the cell proliferation is increased along with the increase of the concentrations under the condition of the same intervention time; the inhibition rate of ALN-Ca-THZ1 on cell proliferation increased with time at the same intervention concentration.
Example 8 inhibition of tumor cell migration assay
Collecting SKOV3 cells in logarithmic growth phase, digesting with pancreatin, and treating with 1 × 104The density per well was seeded in 12-well plates. At 37 ℃ and 5% CO2And (5) standing and culturing in a constant-temperature incubator with saturated humidity. When the cells are cultured in a conventional way until the cells are in a monolayer and the cell fusion degree reaches 85 percent, 2 longitudinal lines in a straight line shape with the same width are scribed at the bottom of the pore plate in parallel by a 100-mu-L sterile gun head, and the cells and cell fragments are washed by PBS for 3 times immediately so as to wash off the suspended cells and cell fragments. The relative distance of the scratched area was recorded by taking a photograph under a microscope (0 hour). After sucking all PBS under negative pressure, 1mL of 2% serum culture medium containing 2mg/L ALN-Ca-THZ1 was added, and the culture was continued in an incubator with the 2% serum culture medium as a control group without drug addition. Observing under a microscope, taking a picture for 24 hours, placing the picture in an incubator, and continuing culturing at a magnification of 200 x. Each scratch was photographed at 6 different locations. Measuring the widths of different grouped scratches at different times in Adobe Illustrator (AI) software; cell mobility ═ (0 hr intercellular space-24 hr intercellular space)/0 hr intercellular space 100%.
The result of the cell scratching experiment is shown in fig. 4, and shows that the scratching distance of the ALN-Ca-THZ1 group is obviously larger than that of the control group, which indicates that the cell activity and the migration capability are weakened, and the ALN-Ca-THZ1 plays a role in obviously inhibiting the migration of ovarian cancer cells.
Example 9 animal experiments
SKOV3 cells were dispersed in PBS and cell density was adjusted to 2X 107Per mL, 0.2mL of cell suspension is extracted by a 1mL syringe, 5-6 weeks old female nude mice abdominal skin is disinfected by 75% alcohol, and the cells with the number of 4 multiplied by 10 are injected into the nude mice abdominal cavity after the left lower abdomen is back-extracted without blood6And (4) respectively. After 5 days from the injection of tumor cells, nude mice were randomly divided into a control group and three treatment groups, the control group was intraperitoneally injected with PBS 0.2mL, and the treatment groups were intraperitoneally injected with NaALN 0.16 mg/mouse, THZ10.02mg/mouse, ALN-Ca-THZ10.2 mg/mouse, once every 7 days, and three consecutive injections were performed. The mental condition, skin, diet, activity and the like of the nude mice were observed every day, the abdominal circumference and body weight of the nude mice were measured from the seventh day after tumor transplantation, the growth of the abdominal cavity transplantation tumor was observed with a mouse living body imager, and the number of days of survival of each group of mice was calculated. The data processing is carried out by using SPSS12.0 statistical software, and the experimental data are all expressed by mean +/-standard deviation.
The results are shown in fig. 5, and the four groups of tumor-bearing nude mice all form transplantation tumor in the abdominal cavity, and the tumor formation rate is 100%. The fluorescence value of the abdominal cavity tumor of the nude mice of the ALN-Ca-THZ1 treatment group is smaller than that of the control group, and the number of the metastasis spots is not increased, which indicates that the ALN-Ca-THZ1 can inhibit the growth and the metastasis of the tumor in vivo, and has potential value for treating the abdominal cavity metastasis of the ovarian cancer.
Finally, it should be noted that the above embodiments are only used for illustrating the technical solutions of the present invention and not for limiting the protection scope of the present invention, and although the present invention is described in detail with reference to the preferred embodiments, it should be understood by those skilled in the art that modifications or equivalent substitutions can be made on the technical solutions of the present invention without departing from the spirit and scope of the technical solutions of the present invention.

Claims (10)

1. An anti-tumor nano-drug, which is characterized by comprising a bisphosphonate, a tumor chemotherapeutic drug and at least one metal ion.
2. The anti-tumor nano-drug according to claim 1, wherein the bisphosphonate is alendronate, etidronate, clodronate, ibandronate, or zoledronic acid.
3. The nanomedicine according to claim 1, wherein the chemotherapeutic agent is an alkylating agent, an antimetabolite, an anticancer antibiotic, a traditional Chinese medicine drug or a hormone drug.
4. The anti-tumor nano-drug according to claim 1, wherein the preparation method of the anti-tumor nano-drug comprises: adding alendronate sodium water solution and THZ1 hydrochloride water solution into calcium chloride solution, adjusting pH value to neutral or alkaline, stirring, centrifuging to obtain nanometer medicinal preparation, and washing with water solution and ethanol to remove free medicinal preparation to obtain ALN-Ca-THZ1 nanometer medicinal preparation for treating tumor.
5. The anti-tumor nano-drug according to claim 1, wherein the preparation method of the anti-tumor nano-drug comprises: adding alendronate sodium water solution and 5 fluorouracil water solution into calcium chloride solution, adjusting pH value to be neutral or alkaline, stirring, centrifuging to obtain nano-drug, and washing free drug with water solution and ethanol to obtain ALN-Ca-5Fu anti-tumor nano-drug.
6. The anti-tumor nano-drug according to claim 1, wherein the anti-tumor nano-drug can further modify a group targeting tumor or a group targeting normal tissue on the surface of the nanoparticle.
7. The anti-tumor nano-drug according to claim 6, wherein the tumor targeting group is RGD polypeptide, folic acid or transferrin.
8. The anti-tumor nano-drug according to claim 7, wherein the RGD polypeptide is NHS-PEG-RGD, and the molecular weight of the NHS-PEG-RGD is 500-15000.
9. The use of the anti-tumor nano-drug of any one of claims 1 to 8 in the preparation of a medicament for the treatment of cancer.
10. The use of claim 9, wherein the cancer is ovarian cancer.
CN202110859262.3A 2021-07-28 2021-07-28 Anti-tumor nano-drug and preparation method and application thereof Pending CN113599525A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110859262.3A CN113599525A (en) 2021-07-28 2021-07-28 Anti-tumor nano-drug and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110859262.3A CN113599525A (en) 2021-07-28 2021-07-28 Anti-tumor nano-drug and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN113599525A true CN113599525A (en) 2021-11-05

Family

ID=78305814

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110859262.3A Pending CN113599525A (en) 2021-07-28 2021-07-28 Anti-tumor nano-drug and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN113599525A (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114632078A (en) * 2022-02-16 2022-06-17 中山大学附属第七医院(深圳) Heat shock protein inhibitor and preparation method and application thereof
CN114712310A (en) * 2022-01-18 2022-07-08 中国人民解放军空军军医大学 Preparation method and application of intelligent bone-targeted delivery drug capable of efficiently entering cells
CN115054691A (en) * 2022-07-15 2022-09-16 中山大学附属第七医院(深圳) Ternary sulfur group metal nano particle and preparation method and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111973573A (en) * 2020-08-04 2020-11-24 中山大学附属第七医院(深圳) Calcium phosphate nano-particles and preparation method and application thereof
CN112826939A (en) * 2021-02-03 2021-05-25 中山大学附属第七医院(深圳) Abdominal perfusion nano-medicine and preparation method and application thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111973573A (en) * 2020-08-04 2020-11-24 中山大学附属第七医院(深圳) Calcium phosphate nano-particles and preparation method and application thereof
CN112826939A (en) * 2021-02-03 2021-05-25 中山大学附属第七医院(深圳) Abdominal perfusion nano-medicine and preparation method and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
彭慧昕等: "细胞周期蛋白依赖性激酶7基因沉默或敲除抑制卵巢癌细胞增殖及其机制探讨", 《肿瘤》 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114712310A (en) * 2022-01-18 2022-07-08 中国人民解放军空军军医大学 Preparation method and application of intelligent bone-targeted delivery drug capable of efficiently entering cells
CN114632078A (en) * 2022-02-16 2022-06-17 中山大学附属第七医院(深圳) Heat shock protein inhibitor and preparation method and application thereof
CN114632078B (en) * 2022-02-16 2023-12-19 中山大学附属第七医院(深圳) Heat shock protein inhibitor and preparation method and application thereof
CN115054691A (en) * 2022-07-15 2022-09-16 中山大学附属第七医院(深圳) Ternary sulfur group metal nano particle and preparation method and application thereof

Similar Documents

Publication Publication Date Title
CN113599525A (en) Anti-tumor nano-drug and preparation method and application thereof
Çırpanlı et al. Antitumoral activity of camptothecin-loaded nanoparticles in 9L rat glioma model
Zhang et al. Preparation and characterization of insulin-loaded bioadhesive PLGA nanoparticles for oral administration
Wang et al. RGD peptide conjugated liposomal drug delivery system for enhance therapeutic efficacy in treating bone metastasis from prostate cancer
Chen et al. Lipid insertion enables targeted functionalization of paclitaxel-loaded erythrocyte membrane nanosystem by tumor-penetrating bispecific recombinant protein
Jain Nanomedicine: application of nanobiotechnology in medical practice
Alphandéry Applications of magnetotactic bacteria and magnetosome for cancer treatment: a review emphasizing on practical and mechanistic aspects
CN108409756B (en) Camptothecin-based heterodimer multifunctional prodrug and preparation method and application thereof
US20140294983A1 (en) Stable nanocomposition comprising doxorubicin, process for the preparation thereof, its use and pharmaceutical compositions containing it
CN107233577A (en) A kind of pH responses and the double medicine-carried nano particles and preparation method of cancer target and application
CA3062624C (en) Quinic acid-modified nanoparticles and uses thereof
CN105407878A (en) Pharmaceutical composition, preparation and uses thereof
EP2978420A1 (en) Stable nanocomposition comprising paclitaxel, process for the preparation thereof, its use and pharmaceutical compositions containing it
CN110665003A (en) Double-drug-loading carrier-free nanoparticle and preparation method thereof
CN107308457A (en) A kind of deep layer degraded with tumor microenvironment response penetrates nanoscale medicine delivery system
CN107496901B (en) Cell autophagy inhibitor and preparation method and application thereof
CN110403916B (en) Nano therapeutic agent and preparation method and application thereof
CN114099674B (en) Copper or zinc carrier for carrying diethyl dithiocarbamic acid prodrug and preparation and application thereof
CN107823652B (en) Long-circulation self-assembly composite nano preparation, preparation method and application thereof
Pan et al. Study on the Effect and Mechanism of Paclitaxel-Succinic Acid Drug-Loaded Nanofibers in Treating Lung Cancer
CN115414492A (en) Nanometer medicinal preparation for treating pancreatic fibrosis, and its preparation method
Ulku et al. The importance of nanotechnology and drug carrier systems in pharmacology
Liao et al. Preparation of galactosyl nanoparticles and their targeting efficiency to hepatocellular carcinoma
CN105796495B (en) Irinotecan hydrochloride liposome pharmaceutical composition and preparation method thereof
CN113616620A (en) Arotinib albumin nanoparticles, preparation method and application thereof, and preparation containing same

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

Application publication date: 20211105

RJ01 Rejection of invention patent application after publication