CN114209847B - Secondary nanocrystallized albumin paclitaxel medicament and application thereof - Google Patents

Secondary nanocrystallized albumin paclitaxel medicament and application thereof Download PDF

Info

Publication number
CN114209847B
CN114209847B CN202111665880.0A CN202111665880A CN114209847B CN 114209847 B CN114209847 B CN 114209847B CN 202111665880 A CN202111665880 A CN 202111665880A CN 114209847 B CN114209847 B CN 114209847B
Authority
CN
China
Prior art keywords
cancer
cell
pharmaceutical composition
albumin
paclitaxel
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
CN202111665880.0A
Other languages
Chinese (zh)
Other versions
CN114209847A (en
Inventor
王刚
郑骏年
韩露露
陈玉鑫
李慧忠
柴大飞
邱小燕
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.)
Xuzhou Medical University
Original Assignee
Xuzhou Medical University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Xuzhou Medical University filed Critical Xuzhou Medical University
Priority to CN202111665880.0A priority Critical patent/CN114209847B/en
Publication of CN114209847A publication Critical patent/CN114209847A/en
Application granted granted Critical
Publication of CN114209847B publication Critical patent/CN114209847B/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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • 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
    • A61P35/02Antineoplastic agents specific for leukemia
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y40/00Manufacture or treatment of nanostructures
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery

Landscapes

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

Abstract

The invention belongs to the field of biomedicine, and particularly relates to a secondary nanocrystallized albumin paclitaxel preparation and application thereof. Specifically, the invention provides a preparation method of a medicine with low toxic and side effects, which comprises the steps of carrying out twice nano treatment on an active ingredient or carrying out nano treatment on a nano medicine again.

Description

Secondary nanocrystallized albumin paclitaxel medicament and application thereof
Technical Field
The invention belongs to the field of biomedicine, and particularly relates to a secondary nanocrystallized albumin paclitaxel medicament and application thereof.
Background
In the last 60 s, american scientists found anticancer active ingredients in the bark of the pacific yew tree: paclitaxel. Paclitaxel was approved for ovarian cancer treatment in 1992. However, the extraction of paclitaxel from yew tree alone is far from sufficient for the clinical needs of many patients, and scientists continue to conduct research and find abundant semi-finished products from european yew tree to semi-synthesize paclitaxel and solve the problem of paclitaxel supply. However, paclitaxel can only be dissolved by means of polyoxyethylated castor oil to ensure a certain solubility and stability, but in 20% to 40% of patients, injection of the solvent causes severe allergic reactions.
The new generation of taxoids is nanoparticle albumin-bound paclitaxel Nab-paclitaxel, and the latest albumin-bound nanoparticle technology is adopted, so that toxic solvents are removed, the problem of allergy is solved, and the treatment effect is improved. In vitro studies show that the concentration of albumin-bound paclitaxel in tumor tissues is 1.33 times that of the common solvent-based paclitaxel, while the drug concentration in normal tissues is 1/2 of that of the common paclitaxel. The existing guidelines recommend albumin-bound paclitaxel to be used in solid tumors such as metastatic breast cancer and pancreatic cancer.
In view of the good solubility of albumin-bound paclitaxel, it is common practice in clinical treatment to increase the dosage of the drug to achieve better therapeutic effect. However, high doses of albumin-bound paclitaxel also induce significant toxic side effects, such as myelosuppression, alopecia, and peripheral neurotoxicity. Therefore, it is a very worthwhile research direction to reduce the toxic and side effects of albumin-bound paclitaxel while maintaining its therapeutic effect, aiming at the problems of the prior albumin-bound paclitaxel in clinical application.
Disclosure of Invention
In order to reduce the toxic and side effects of albumin paclitaxel in treatment, the albumin paclitaxel nanoparticles are subjected to secondary nano-packaging, the obtained secondary nano-albumin paclitaxel avoids the problems of bone marrow suppression and limited hematopoiesis, and the albumin paclitaxel nanoparticles show a very good anti-tumor effect by combining radiotherapy, and the lung cancer experiment shows that the effect is multiplied.
Preparation method
In one aspect, the invention provides a method for preparing a low toxic side effect drug, the method comprising twice nanometering an active ingredient or again nanometering a nanometering drug.
Preferably, the nanocrystallization process includes any method of nanoparticle preparation. Specifically, for example: antisolvent method, emulsification method, thermal gel method, albumin nanometer combination technology, self-assembly technology, and nanometer spray drying technology.
Preferably, the two nano-treatments can adopt the same or different nano-particle preparation methods.
Preferably, the nanocrystallization treatment is performed by an anti-solvent method, and the anti-solvent method comprises the following steps: mixing serum albumin (HSA) with active component or nano-drug, slowly dripping ethanol or acetone, and finally adding cross-linking agent to obtain nano-particles.
Preferably, the step of the antisolvent process comprises the steps of:
1) Mixing HSA + active ingredient or nano-drug; preferably, stirring the mixed solution at a high rotation speed; preferably, a high rotational speed refers to 1000rpm;
2) Adding ethanol or acetone into the solution obtained in the step 1), and continuously stirring; preferably, stirring is carried out for at least 5 minutes (including in particular 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 minutes); more preferably, stirring for 10 minutes;
3) Adding a cross-linking agent to the solution of 2); preferably, after stirring uniformly, incubating in an environment of 25 ℃ or higher for at least 1 hour (specifically including 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours); more preferably, the incubation is for 4 hours.
Preferably, the cross-linking agent comprises formaldehyde, glutaraldehyde;
preferably, the cross-linking agent is glutaraldehyde; more preferably, it is 8% glutaraldehyde.
Preferably, the ethanol or acetone is slowly dropped; more preferably, the dropwise addition rate of the ethanol or acetone is 1ml/min;
preferably, the operation step further comprises the step of taking the nano solution for centrifugal resuspension. Specifically, the step of centrifugation resuspension is: after centrifugation at 12000rpm, the supernatant was discarded, air-dried, and resuspended in sterile water. Preferably, filtration through a 0.22nm filtration membrane is also required.
Secondary nano medicine
On the other hand, the invention provides a secondary nano-sized medicament, which is prepared by carrying out nano-sized treatment on active ingredients twice or carrying out nano-sized treatment on nano-sized medicaments again. That is, the present invention provides the product prepared by the foregoing preparation method.
Suitable "active ingredients" in the present invention illustratively include 5-aminosalicylic acid (salicylic acid), abacavir (abacayir), abacrelix (abarelix), abatacept (abatacept), acamprosate (acamprosate), acarbose (acarbose), aceclofenac (aceclofenac), acetylsalicylic acid, abamectin (acitretin), aclarubicin (aclarubicin), actinomycin (actinomycin), acyclovir (acyclovir), adalimumab (adalimumab), adefovir (adefovir), adefovir dipivoxil (adefovir dipivoxil), adenosine, ademetionine (adenosylmethionine), adrenalin, adriamycin (adriacin).
Preferably, the active ingredient is an anticancer active ingredient.
Preferably, the anticancer active ingredient includes, but is not limited to, any plant-derived ingredient having anticancer activity, including plant-derived active ingredients, specifically, sulphoraphane, paclitaxel (Paclitaxel), epipodophyllotoxin, vincristine (Vincristine), vinblastine (Vinblastine), vinorelbine (Vinorelbine), vindesine (Vindesine), vinblastine (Vinflunine), allicin (Pomiferin), epigallothechin-3-gallate, combrestatin A-4, sphastatin, roscovitine (Roscovitine), flavopirimidyl (Flavopiridol), noscapine (Noscapine), formononectin, cabazitaxel, colchicine (Colocine), combrestatin, docetaxel (docetaxel), duratoxin (Isotaxinol), potentillin (Potentillin-2, verticine.
Preferably, the anticancer active ingredient is paclitaxel.
Preferably, the nano-drug includes, but is not limited to, albumin-bound paclitaxel (paclitaxel: (paclitaxel)) (
Figure BDA0003451830490000031
For example as described in patents WO2014105644 and WO 2008057562) and liposomal daunorubicin (e.g., (r) ((r))
Figure BDA0003451830490000032
For example as described in patents EP0004467 and US 20070286897).
Preferably, the nano-drug is albumin-bound paclitaxel (also referred to herein simply as "albumin paclitaxel").
Preferably, the particle size range of the secondary nano-medicament provided by the invention is 50-150nm; preferably about 100nm.
Cells
In another aspect, the present invention provides a cell comprising the drug produced by the above-described production method or the above-described secondary nanometerized drug.
Preferably, the cells comprise animal cells;
preferably, the cell is a human cell;
more preferably, the cell is a neutrophil.
Pharmaceutical composition
In another aspect, the present invention provides a pharmaceutical composition comprising the drug prepared by the foregoing preparation method or the foregoing secondary nanosized drug.
Preferably, the pharmaceutical composition further comprises any active pharmaceutical substance capable of being an ingredient of a multiparticulate dosage form, a tablet comprising pellets, a mini-tablet, a capsule, a sachet, an effervescent tablet or a dry powder for oral suspension;
pharmaceutical compositions of the compounds of the present invention may be administered by any of the following means: oral, aerosol inhalation, rectal, nasal, buccal, topical, parenteral, such as subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intracardiac, intrasternal or intravenous administration.
Preferably, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier, diluent or excipient.
Preferably, the pharmaceutically acceptable carrier, diluent or excipient includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersant, suspending agent, stabilizer, isotonic agent, solvent, surfactant or emulsifier that has been approved by the U.S. food and drug administration or the national food and drug administration for use in humans or livestock.
Preferably, the pharmaceutical composition may be in the form of tablets, pills, powders, granules, capsules, lozenges, syrups, liquids, emulsions, suspensions, controlled release preparations, aerosols, films, injections, intravenous drip, transdermal preparations, ointments, lotions, adhesive preparations, suppositories, pellets, nasal preparations, pulmonary preparations, eye drops and the like, oral or parenteral preparations.
System for controlling a power supply
In another aspect, the invention provides a cancer treatment system combining chemoradiotherapy, the system comprising at least the following two modules:
1) A radiotherapy module to perform radiotherapy, in particular, the radiotherapy module comprises an instrument or reagent to perform radiotherapy;
2) The chemotherapy module for administering the medicine prepared by the preparation method or the secondary nano-medicine to the patient preferably also comprises other instrument devices required for administering the medicine.
Preferably, the radiotherapy module and the chemotherapy module are repeatedly administered alternately;
preferably, the chemotherapy module is administered 1-3 days prior to administration of the radiotherapy module; one radiotherapy and one chemotherapy can be called as one course of treatment, and a patient can be treated by a plurality of courses of treatment; specifically, it may be: a first-use radiotherapy module (abbreviated as "first radiotherapy") -a first-use chemotherapy module (abbreviated as "first chemotherapy") -a second-use radiotherapy module (abbreviated as "second radiotherapy") -a second-use chemotherapy module (abbreviated as "second chemotherapy");
the radiotherapy and the radiotherapy have the same meanings and can be replaced mutually. Radiation therapy is one of three cancer core therapies. Researchers have found that tumor-targeted radiation therapy can act as a "cancer vaccine" to induce neutrophil-mediated tumor cell death, stimulating the immune system against other metastatic cancer cells.
The chemotherapy and the chemical drug treatment are the same in meaning and can be replaced with each other. Specifically, the chemical drug used in the present invention is a drug prepared by the above-mentioned preparation method or the above-mentioned secondary nanocrystallized drug.
Applications of the invention
In another aspect, the invention provides a product prepared by the preparation method, a secondary nano-sized medicament, a cell, a medicinal composition and application in preparing a medicament for treating cancer.
Preferably, the medicament for treating cancer includes a medicament used in conjunction with radiotherapy.
Preferably, the radiation comprises ionizing radiation, particle beam radiation.
Preferably, the particle beam comprises electrons, protons, neutrons, heavy ions such as carbon ions or mesons.
Preferably, the ionizing radiation comprises x-ray radiation, ultraviolet radiation, infrared radiation, gamma ray radiation or microwave radiation.
In another aspect, the invention provides the use of a cancer treatment system as hereinbefore described in the manufacture of a product for the treatment of cancer.
Preferably, the cancer comprises: cervical cancer, seminoma, testicular lymphoma, prostate cancer, ovarian cancer, lung cancer, rectal cancer, breast cancer, cutaneous squamous cell carcinoma, colon cancer, liver cancer, pancreatic cancer, gastric cancer, esophageal cancer, thyroid cancer, transitional epithelial cancer of the bladder, leukemia, brain tumor, gastric cancer, peritoneal cancer, head and neck cancer, endometrial cancer, kidney cancer, cancer of the female reproductive tract, carcinoma in situ, neurofibroma, bone cancer, skin cancer, gastrointestinal stromal tumors, mast cell tumors, multiple myeloma, melanoma, glioma, and sarcoma.
Preferably, the cancer comprises any cancer that can be treated with paclitaxel. In particular, ovarian cancer, breast cancer, lung cancer, esophageal cancer, gastric cancer, non-Hodgkin's lymphoma, colorectal cancer, melanoma, head and neck cancer, lymphoma and brain tumor have certain curative effects.
Preferably, the cancer is lung cancer.
Preferably, the cancer treatment is a model organism constructed against LLC lung cancer cells.
Preferably, the model organism is a mouse.
Method of treatment
In another aspect, the present invention provides a method for treating cancer, which comprises administering any one of the secondary nanocrystallization drugs, the products, the cells and the pharmaceutical compositions prepared by the preparation method to a patient.
Preferably, the method further comprises subjecting the patient to radiation therapy.
Drawings
Fig. 1 is a graph showing the measurement result of the particle size of the double-nanocrystallized albumin paclitaxel, fig. 1A is a graph showing the detection result, and fig. 1B is a data statistical graph.
Fig. 2 is a graph showing the results of potential measurement of double nanocrystallized albumin paclitaxel, fig. 2A is a graph showing the results of detection, and fig. 2B is a data statistical graph.
FIG. 3 shows the stability test results of two-time nanocrystallized albumin paclitaxel.
FIG. 4 shows the results of particle size stability test of the double nanocrystallized albumin paclitaxel.
FIG. 5 is the morphology of two-time nanocrystallized albumin paclitaxel under electron microscope.
FIG. 6 is a graph showing the result of the measurement of the drug loading of neutrophils under the stimulation of radiotherapy.
FIG. 7 is a graph showing the result of detection of drug loading of neutrophils under PMA stimulation, FIG. 7A is a graph showing the result of flow cytometry, and FIG. 1B is a graph showing the result under a microscope.
FIG. 8 is a graph showing the results of a chemotaxis experiment, FIG. 8A is a graph showing the results of detection, and FIG. 8B is a data statistics graph.
FIG. 9 is a graph showing the results of ROS detection, FIG. 9A is a graph showing the results of detection, and FIG. 9B is a data statistics graph.
FIG. 10 is a graph showing the results of measurement of the expression levels of il-1. Beta. And il-6. Beta. In FIG. 10A, il-6. Beta. And il-6 in FIG. 10B.
FIG. 11 is a flow chart of a mouse experiment.
Fig. 12 is a tumor solid map.
Fig. 13 is a statistical graph of tumor volume and tumor weight change, fig. 13A is volume, and fig. 13B is weight.
FIG. 14 is a statistical graph of the change in body weight of mice.
Fig. 15 is a graph showing the percentage of drug-loaded neutrophils in the tumor, fig. 15A is a flow chart, and fig. 15B is a statistical chart.
Fig. 16 is a graph of the results of HE staining of the heart, liver, spleen, lung and kidney, fig. 16A: heart, fig. 16B: lung, fig. 16C: the liver of a human being is,
FIG. 16D: spleen, fig. 16E: kidneys.
FIG. 17 is a graph showing the results of blood routine and liver renal function tests.
Detailed Description
The present invention will be further described with reference to the following examples, which are intended to be illustrative only and not to be limiting of the invention in any way, and any person skilled in the art can modify the present invention by applying the teachings disclosed above and applying them to equivalent embodiments with equivalent modifications. Any simple modifications or equivalent changes made to the following embodiments according to the technical essence of the present invention, without departing from the technical spirit of the present invention, fall within the scope of the present invention.
Example 1 preparation of Secondary Nanometrized Albumin paclitaxel
The preparation method comprises the following steps:
1. placing 1ml of albumin-binding paclitaxel solution (trade name: aiyue; jiangsu Henry pharmaceutical Co., ltd.) with concentration of 20mg/ml into a 50ml conical flask containing a stirring rotor, and slowly adjusting the rotation speed (1000 rpm) for stirring;
2. slowly dripping 3.45ml of absolute ethyl alcohol into the conical flask at the speed of 1ml/min, sealing the conical flask opening by using a preservative film, and stirring for 10min at room temperature;
3. finally, 20 mul of 8% glutaraldehyde is added, the solution is transferred to a 15ml centrifuge tube after being stirred uniformly, and the solution is incubated for 4 hours at room temperature (> 25 ℃);
4. taking 500 mu L of the nano solution, subpackaging the nano solution into 1.5ml Ep tubes, centrifuging the tubes at 12000rpm for 15min, removing supernatant, and then airing the tubes at room temperature;
5. adding 1ml of sterilized water to resuspend the precipitate, filtering by a 0.22nm filter membrane, and storing at 4 ℃ in the dark.
Feature verification
And scanning the prepared product under an electron microscope, and measuring the particle size, potential, stability and particle size stability of the product. The scanograms and the results of the measurements are shown in FIGS. 1-5.
Example 2 Effect of Secondary Nanometrized Albumin paclitaxel on neutrophils
Considering that the therapeutic effect of the modified nano-drug is mediated by neutrophils, the influence of the modified nano-drug on neutrophils is judged.
The nano-drug prepared as shown in fig. 6 can be phagocytized by neutrophils normally in vivo and in vitro (flow and electron microscope detection), and is more obvious under the inflammatory stimulation state (fig. 7), and the maximum drug loading can reach 99.1%.
In order to determine the influence of the modified nano-drug on the migration characteristic, oxygen burst and inflammatory cytokine expression of the nano-drug, the migration characteristic of the drug-loaded neutrophilic granulocyte is evaluated through a Transwell experiment; secondly, evaluating the change of the intracellular ROS level of the neutrophilic granulocyte after drug loading through flow cytometry and fluorescence intensity detection; and the expression of neutrophil associated inflammatory cytokines, in particular il-1 β, il-6, after loading was assessed by RT-qPCR.
The chemotaxis experiment result is shown in FIG. 8 (in the figure, fMLP is fMLP, which is called N-formamyl-methionyl-leucyl-phenylalanine, chinese name: N-formyl-L-methionyl-L-leucyl-L-phenylalanine tripeptide; a leukocyte chemotactic peptide found in bacteria, has strong chemotaxis and inflammation-causing activity, and can rapidly activate leukocytes). The ROS detection results are shown in fig. 9 (H2 DCFDA, a probe conventionally used for detecting Reactive Oxygen Species (ROS) in cells). FIG. 10 shows the results of measurement of the expression levels of il-1. Beta. And il-6 of inflammatory cytokines.
The above results indicate that drug loading does not affect neutrophil migration itself, ROS production and expression of related inflammatory cytokines.
Example 3, in vivo experiments: lung cancer model
Constructing a mouse LLC lung cancer subcutaneous tumor model at the thigh part on the right side of the mouse; radiotherapy is carried out one day before the medicine treatment; mice body weight and tumor volume were monitored periodically. The specific flow is shown in fig. 11.
The tumor mass is shown in fig. 12, and the change in the statistical volume and weight of the data is shown in fig. 13. The mice were also monitored for weight change and statistics are shown in figure 14.
Whether the drug loading of neutrophils in the tumor was detected is shown in fig. 15, and about 89% of the drug loading of neutrophils was performed.
Example 4 judgment of drug safety
After the nano-drug can obviously inhibit tumors after being definitely modified, the toxic and side effects of the nano-drug after being definitely modified are definitely determined by the HE staining of the heart, the liver, the spleen, the lung and the kidney of a mouse, and the result is shown in figure 16; simultaneously, the liver and kidney functions and the peripheral blood cell changes of the mice were detected by a mouse biochemical analyzer and a blood conventional analyzer, respectively (fig. 17).
The results show that the modified nano-drug has no obvious toxic or side effect and can obviously reduce the bone marrow suppression effect of the albumin-bound paclitaxel.

Claims (18)

1. A method for preparing a low toxic and side effects drug, the method comprising the step of performing nanocrystallization treatment on albumin-bound paclitaxel again, wherein the nanocrystallization treatment adopts an anti-solvent method, and the anti-solvent method comprises the following steps:
1) Taking the paclitaxel combined with the albumin,
2) Adding ethanol or acetone into the solution of the step 1),
3) Adding a crosslinking agent to the solution of 2).
2. The method of claim 1, wherein the cross-linking agent comprises formaldehyde, glutaraldehyde.
3. The method of claim 2, wherein the cross-linking agent is glutaraldehyde.
4. The method of claim 3, wherein the glutaraldehyde content is 8%.
5. The production method according to claim 1, wherein the dropping rate of ethanol or acetone is 1 ml/min.
6. A medicament obtainable by the process of any one of claims 1 to 5.
7. A cell comprising the agent of claim 6.
8. The cell of claim 7, comprising an animal cell.
9. The cell of claim 8, which is a human cell.
10. The cell of claim 9, which is a neutrophil.
11. A pharmaceutical composition comprising the medicament of claim 6.
12. The pharmaceutical composition of claim 11, wherein the pharmaceutical composition is administered by any of the following means: oral, aerosol inhalation, rectal, nasal, buccal, subcutaneous, intravenous, intramuscular, intraperitoneal or intrathecal administration.
13. The pharmaceutical composition of claim 11, further comprising the pharmaceutically acceptable adjuvant, carrier, glidant, sweetener, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersant, suspending agent, isotonic agent, or solvent.
14. The pharmaceutical composition of claim 11, wherein the dosage form of the pharmaceutical composition comprises tablets, pills, powders, granules, capsules, lozenges, syrups, emulsions, suspensions, aerosols, films, injections, ointments, lotions, suppositories.
15. A chemoradiotherapy-combined cancer treatment system, comprising at least the following two modules:
1) A radiotherapy module for an instrument or reagent for performing radiotherapy;
2) A chemotherapy module for administering the drug of claim 6 to a patient.
16. Use of the medicament of claim 6, the cell of claim 7, the pharmaceutical composition of claim 11, the cancer therapy system of claim 15 in the manufacture of a product for the treatment of cancer.
17. The use of claim 16, wherein the cancer comprises: cervical cancer, seminoma, testicular lymphoma, prostate cancer, ovarian cancer, lung cancer, rectal cancer, breast cancer, cutaneous squamous cell carcinoma, colon cancer, liver cancer, pancreatic cancer, gastric cancer, esophageal cancer, thyroid cancer, transitional epithelial carcinoma of the bladder, leukemia, brain tumor, cancer of the peritoneum, cancer of the head and neck, endometrial cancer, kidney cancer, cancer of the female reproductive tract, carcinoma in situ, neurofibroma, bone cancer, skin cancer, gastrointestinal stromal tumors, mast cell tumors, multiple myeloma, melanoma, glioma, and sarcoma.
18. The use of claim 16, wherein the cancer is lung cancer.
CN202111665880.0A 2021-12-31 2021-12-31 Secondary nanocrystallized albumin paclitaxel medicament and application thereof Active CN114209847B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111665880.0A CN114209847B (en) 2021-12-31 2021-12-31 Secondary nanocrystallized albumin paclitaxel medicament and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111665880.0A CN114209847B (en) 2021-12-31 2021-12-31 Secondary nanocrystallized albumin paclitaxel medicament and application thereof

Publications (2)

Publication Number Publication Date
CN114209847A CN114209847A (en) 2022-03-22
CN114209847B true CN114209847B (en) 2022-12-13

Family

ID=80707384

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111665880.0A Active CN114209847B (en) 2021-12-31 2021-12-31 Secondary nanocrystallized albumin paclitaxel medicament and application thereof

Country Status (1)

Country Link
CN (1) CN114209847B (en)

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6749868B1 (en) * 1993-02-22 2004-06-15 American Bioscience, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
CN102784109B (en) * 2012-08-22 2014-08-06 复旦大学 Taxane medicines albumin nanoparticle preparation for injection and preparation method thereof
CA2949092A1 (en) * 2014-05-16 2015-11-19 Dana-Farber Cancer Institute, Inc. Protein-based particles for drug delivery
CN106540270A (en) * 2016-12-07 2017-03-29 中国科学技术大学 Paclitaxel transports albumin nano medicine altogether with all-trans-retinoic acid
CN107412172A (en) * 2017-08-04 2017-12-01 南京拉克森生物医药科技有限公司 A kind of suspension freeze-dried powder of taxol albumin nano and its preparation technology

Also Published As

Publication number Publication date
CN114209847A (en) 2022-03-22

Similar Documents

Publication Publication Date Title
Hu et al. The potentiated checkpoint blockade immunotherapy by ROS-responsive nanocarrier-mediated cascade chemo-photodynamic therapy
Caster et al. Effect of particle size on the biodistribution, toxicity, and efficacy of drug-loaded polymeric nanoparticles in chemoradiotherapy
US9814734B2 (en) Bufalin liposome, preparation method therefor and application thereof
US20190224238A1 (en) Tumor therapeutic drug
CN103209693B (en) Arctigenin prevents in preparation or treats the purposes in the medicine of cytopenia relevant disease
CN111084787A (en) Preparation method, medicament and application of anti-tumor microparticles
CN105997943B (en) A kind of nano particle and its preparation method and application of human serum albumins load camptothecine
Zhao et al. Biomimetic nanotherapy: core–shell structured nanocomplexes based on the neutrophil membrane for targeted therapy of lymphoma
CN104586775A (en) Astragalus polysaccharide sustained release microsphere for treating radiation pneumonitis and preparation method of astragalus polysaccharide sustained release microsphere
CN104548125A (en) Preparation and application of PEG-PTX-NCs (pegylation-paclitaxel-nanocrystals)
Sunil et al. Convection enhanced delivery of light responsive antigen capturing oxygen generators for chemo-phototherapy triggered adaptive immunity
CN106983719A (en) A kind of docetaxel polymer nano micelle injection, its preparation method and its application in tumor is prepared
CN114209847B (en) Secondary nanocrystallized albumin paclitaxel medicament and application thereof
CN113599532A (en) Drug and collagenase loaded albumin composite nanoparticles, preparation and application
JP2006525238A (en) Activated carbon infusion, method for its preparation and use thereof in the preparation of drugs for cancer treatment
CN105997942B (en) A kind of nano particle and its preparation method and application of human serum albumins load CHROMATOGRAPHIC FRACTIONATION AND MASS drug
KR102304705B1 (en) Immunocyte-based Microrobot for Medical use
CN110665009B (en) Nanometer gemcitabine for promoting normalization of tumor blood vessels and application thereof
CN113384698B (en) Self-assembled nano-medicament for synergetic chemotherapy/acousto-photodynamic therapy and application thereof
CN108392483B (en) A kind of preparation method and application of the albumin nano granular of paclitaxel plus 2ME2
CN114788811A (en) Gemcitabine hydrochloride chitosan micelle and preparation method thereof
CN104688722B (en) Purposes of the epimedium aglucone in preparation prevention or treatment bone marrow suppression drug
Miao et al. Preparation of DOX/BSANP and its antitumor effect on bel-7404 liver cancer cells in vitro and in vivo
CN115634211B (en) Nanoparticle for delivering lotus plumule alkaloid by taking human serum albumin as carrier and preparation method and application thereof
CN103638027B (en) Utilize method and the preparation of amphiphilic drug encapsulation hydrophobic anticancer drug

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