WO2021147598A1 - 含氟化合物修饰的阳离子聚合物作为药物载体的应用及制备方法 - Google Patents

含氟化合物修饰的阳离子聚合物作为药物载体的应用及制备方法 Download PDF

Info

Publication number
WO2021147598A1
WO2021147598A1 PCT/CN2020/138465 CN2020138465W WO2021147598A1 WO 2021147598 A1 WO2021147598 A1 WO 2021147598A1 CN 2020138465 W CN2020138465 W CN 2020138465W WO 2021147598 A1 WO2021147598 A1 WO 2021147598A1
Authority
WO
WIPO (PCT)
Prior art keywords
chitosan
drug
drugs
fluorine
acid
Prior art date
Application number
PCT/CN2020/138465
Other languages
English (en)
French (fr)
Inventor
刘庄
陈倩
金秋桐
赵琪
肖志晟
韦婷
沈菁菁
Original Assignee
苏州大学
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 苏州大学 filed Critical 苏州大学
Priority to US17/630,143 priority Critical patent/US20220273806A1/en
Priority to EP20915498.8A priority patent/EP4095162A4/en
Publication of WO2021147598A1 publication Critical patent/WO2021147598A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • 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
    • A61K47/61Medicinal 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 the organic macromolecular compound being a polysaccharide or a derivative thereof
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B37/00Preparation of polysaccharides not provided for in groups C08B1/00 - C08B35/00; Derivatives thereof
    • C08B37/0006Homoglycans, i.e. polysaccharides having a main chain consisting of one single sugar, e.g. colominic acid
    • C08B37/0024Homoglycans, i.e. polysaccharides having a main chain consisting of one single sugar, e.g. colominic acid beta-D-Glucans; (beta-1,3)-D-Glucans, e.g. paramylon, coriolan, sclerotan, pachyman, callose, scleroglucan, schizophyllan, laminaran, lentinan or curdlan; (beta-1,6)-D-Glucans, e.g. pustulan; (beta-1,4)-D-Glucans; (beta-1,3)(beta-1,4)-D-Glucans, e.g. lichenan; Derivatives thereof
    • C08B37/00272-Acetamido-2-deoxy-beta-glucans; Derivatives thereof
    • C08B37/003Chitin, i.e. 2-acetamido-2-deoxy-(beta-1,4)-D-glucan or N-acetyl-beta-1,4-D-glucosamine; Chitosan, i.e. deacetylated product of chitin or (beta-1,4)-D-glucosamine; Derivatives thereof
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • 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
    • 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
    • A61K47/542Carboxylic acids, e.g. a fatty acid or an amino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/40Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing nitrogen
    • A61K8/43Guanidines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/40Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing nitrogen
    • A61K8/44Aminocarboxylic acids or derivatives thereof, e.g. aminocarboxylic acids containing sulfur; Salts; Esters or N-acylated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/49Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds
    • A61K8/4906Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with one nitrogen as the only hetero atom
    • A61K8/4913Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with one nitrogen as the only hetero atom having five membered rings, e.g. pyrrolidone carboxylic acid
    • A61K8/492Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with one nitrogen as the only hetero atom having five membered rings, e.g. pyrrolidone carboxylic acid having condensed rings, e.g. indol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/60Sugars; Derivatives thereof
    • A61K8/606Nucleosides; Nucleotides; Nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/72Cosmetics or similar toiletry preparations characterised by the composition containing organic macromolecular compounds
    • A61K8/73Polysaccharides
    • A61K8/736Chitin; Chitosan; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • 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
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/02Preparations for care of the skin for chemically bleaching or whitening the skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q7/00Preparations for affecting hair growth
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L5/00Compositions of polysaccharides or of their derivatives not provided for in groups C08L1/00 or C08L3/00
    • C08L5/08Chitin; Chondroitin sulfate; Hyaluronic acid; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/10General cosmetic use
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/40Chemical, physico-chemical or functional or structural properties of particular ingredients
    • A61K2800/54Polymers characterized by specific structures/properties
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/40Chemical, physico-chemical or functional or structural properties of particular ingredients
    • A61K2800/56Compounds, absorbed onto or entrapped into a solid carrier, e.g. encapsulated perfumes, inclusion compounds, sustained release forms
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to the technical fields of polymer chemistry and medical biomaterials, in particular to a cationic polymer modified and modified based on fluoride as a drug carrier, and a preparation method and application thereof.
  • hydrophilic cationic polymer materials such as polyethyleneimine (PEI) and polylysine can combine with nucleic acid, polypeptide and protein molecules to form nanocomposites, which not only promote the entry of these macromolecular compounds into cells, but also their cationic properties. And it can protect the drug from being degraded by hydrolase in the microenvironment, and the internal tertiary ammonia structure promotes the escape of the drug in the cell endosomes through the action of proton sponge.
  • the cationic polymer material can weaken the tight junction of epithelial cells, increase the permeability of the epithelium, and promote the absorption efficiency of drug macromolecules in the epithelial cells.
  • the high cytotoxicity of cationic polymer materials during use ultimately limits their clinical applications.
  • Chitosan is a cationic polysaccharide after deacetylation of chitin. It has good biosafety properties and excellent mucosal adhesion properties. It has been widely used in the design of transmucosal dosage forms. It is reported in the literature that chitosan can produce mucosal adhesion through its own positive charge and the negative charge on the skin and mucosal surface and the hydrophobic effect of the hydrophobic group, effectively extending the biologically active substances (drugs, peptides, proteins, etc.) in the chitosan solution.
  • chitosan can significantly improve the bioavailability of infused drugs, high concentrations of chitosan may cause severe mucosal and epithelial damage, limiting its clinical application as a drug carrier.
  • Transdermal drug delivery refers to a drug delivery method in which the drug penetrates the skin, is absorbed through the capillaries and then enters the blood circulation to achieve an effective blood drug concentration and then produce a drug effect.
  • Transdermal administration can avoid the liver first-pass effect of oral administration, and avoid the inactivation of the drug in the gastrointestinal tract, especially for patients who need long-term administration, transdermal administration is a convenient and quick treatment method.
  • the skin can prevent the intrusion of most foreign substances. The rate of drug penetration through the skin is often slow, and the penetration amount is difficult to reach the concentration required for effective treatment, and the optimal therapeutic effect cannot be exerted.
  • the dose of transdermal drug preparations is usually related to the effective contact area between the drug delivery system and the skin. Increasing the area can increase the dose, but generally the drug delivery area is not greater than 60cm2, so a certain transdermal rate of the drug is required. . Except for a small number of drugs with small dosage requirements and suitable dissolution characteristics, most drugs are difficult to meet the treatment requirements.
  • transdermal dosage forms include patches or gels containing chemical penetration-enhancing components and patches or gel dosage forms that promote physical penetration. Since the launch of Transderm Scop, a transdermal patch for the treatment of motion sickness in 1981, patches and gels containing chemical penetration enhancing ingredients have been widely used to treat a variety of diseases including dementia, Parkinson’s disease and acute pain. So far, more than 20 transdermal patches have been approved by the US FDA for marketing. However, the existing technologies or products usually have one or more defects such as limited transdermal effect, low drug bioavailability, inability to integrate well with a variety of drugs universally, and high toxicity.
  • Mucosa refers to the membrane-like structure lining the lumen of the tubular or sac-like organs of the organism (digestive, respiratory, urinary, reproductive, etc.). It is composed of epithelial tissue and loose connective tissue, and some organs contain muscularis mucosa.
  • the connective tissue part is called the lamina propria, and the epithelial tissue part is called the epithelium, which contains blood vessels and nerves and can secrete mucus.
  • the color of normal mucosa can vary from light red to bright red, moist, with a certain degree of stretch due to the degree of blood filling, and wrinkles are often formed in an empty state.
  • epithelium is the main part of tubular organs for functional activities.
  • the type of epithelium varies depending on the location and function. There are different names according to different parts, such as nasal mucosa, lung mucosa, vaginal mucosa, oral mucosa, gastrointestinal mucosa, etc.
  • hydrophilic cationic polymer materials such as polyethyleneimine (PEI), polyamide-amine dendrimer (dendrimer), chitosan (chitosan), ⁇ -cyclodextrin, gelatin, cationic Peptides/amino acids, such as polylysine (PLL), cationic polyester, cationic polyphosphate, polyvinylpyridinium, poly(dimethylamino)ethyl methacrylate, can weaken the tight junctions of epithelial cells and increase The permeability of the epithelium can promote the penetration of the drug through the mucosa and improve the bioavailability of the drug.
  • PEI polyethyleneimine
  • dendrimer polyamide-amine dendrimer
  • chitosan chitosan
  • ⁇ -cyclodextrin gelatin
  • cationic Peptides/amino acids such as polylysine (PLL)
  • PLL polylysine
  • polyester cationic polyester
  • the purpose of the present invention is to provide a new type of drug carrier material with obvious effect of promoting drug absorption and low toxicity, and the fluorine-containing compound modified chitosan proposed by the present invention has a mature synthesis process, simple operation, high synthesis efficiency, short cycle, and no need
  • the cumbersome purification steps can obtain high-yield drug carriers, and its simple synthesis method provides a good basis for commercialization.
  • the fluorine-containing compound modified chitosan of the present invention can be used as a variety of drug carriers.
  • a fluorinated modified chitosan derivative used as a drug carrier has the following structure: a fluorine-containing compound is covalently attached to the chitosan backbone,
  • the molecular weight of the chitosan is in the range of 1000-5000000, the degree of deacetylation is not less than 55%, and the viscosity is in the range of 25-1000 centipoise,
  • the fluorine-containing compound is the following chemical formula (I) The fluorine-containing aliphatic chain shown, or formula (II) As shown in the aromatic ring functional group, the R1 is halogen (fluorine, chlorine, bromine, iodine), halogen-substituted alkanes, cycloalkanes, aldehyde groups, carboxyl groups, double bonds, alkyne bonds, hydroxyl groups, sulfonyl chlorides, sulfonic acids Reactive groups such as bonds or mercapto groups that can react with primary amino groups.
  • a fluorinated modified chitosan derivative used as a drug carrier has a chitosan molecular skeleton containing primary amino groups as shown in formula (IV):
  • the fluorine-containing functional group is a fluorine-containing aliphatic chain or aromatic ring functional group.
  • x is an integer from 0 to 3
  • y is an integer from 0 to 20
  • z is an integer from 0 to 8
  • R2 is CF 3 , CHF 2 , CH 2 F, or CH 3 (when y is not 0);
  • the fluorine-containing aliphatic chain compound refers to a fluorine-containing hydrocarbon group and its derivatives, including trifluoroacetic acid, pentafluoropropionic acid, heptafluorobutyric acid, nonafluorovaleric acid, undecafluorohexanoic acid, tridecafluoroheptanoic acid, ten Pentafluorooctanoic acid, heptafluorononanoic acid, nonadecafluoroquinic acid, perfluorobutyric acid anhydride, perfluoroheptanoic acid anhydride, perfluorodecanoic acid anhydride, 2,2,3,3,4,4,4-heptafluorobutyl acrylic acid Esters, 3-(1H, 1H, 5H octafluoropentyloxy)-1,2-epoxypropylene, nonafluorobutylsulfonamide anhydride and its derivatives.
  • R is H, CH 3 , OH, NO 2 , O, CF 3 , F, CH 2 OH, CN, NCO, or (CF 2 )aCF 3 (a is an integer of 1-20), etc., and at least one R is F;
  • the fluorine-containing aromatic ring compound includes 3-fluorobenzoic acid, 3,5-difluorobenzoic acid, 2,3,5,6-tetrafluoro-4-methylbenzoic acid, pentafluorobenzoic acid, 2-fluoro- 3-(Trifluoromethyl)benzoic acid and its derivatives.
  • the chitosan and a fluorine-containing compound are covalently connected, and the chitosan molecule is modified on the surface to form a drug carrier
  • formula (V) is as follows:
  • B is a linking group formed by a fluorine-containing functional group and a primary amino group of chitosan
  • C is a fluorine-containing aliphatic chain and aromatic ring functional group.
  • the fluorine-containing aliphatic chain is a type of fluorine-containing compound with an active group that can react with an amino group, including the formula (VI ) Shows:
  • A is -COOH, Reactive groups capable of reacting with primary amino groups, x is an integer of 0-3, and y is an integer of 0-8.
  • the fluorine-containing aromatic ring compound is a type of fluorine-containing compound with an active group that can react with an amino group, including the formula ( VII) Shown:
  • the fluorinated modified chitosan derivative used as a drug carrier is used as a drug carrier of the following drugs, the following drugs are small molecule drugs, polypeptides, Protein drugs, combination drugs of different drugs, and combination drugs of drugs and other pharmaceutical excipients.
  • fluorine-containing compound modified chitosan as a drug carrier
  • the fluorinated modified chitosan derivatives can be used as small molecule drugs, polypeptides, protein drugs, combination drugs of different drugs, and drugs and other drugs.
  • the drug carrier application of combined drugs with excipients can be used as small molecule drugs, polypeptides, protein drugs, combination drugs of different drugs, and drugs and other drugs.
  • a drug complex includes the fluorinated modified chitosan derivative used as a drug carrier and a drug, and the drug includes a small molecule drug, a polypeptide, a protein drug, a combination drug of different drugs, and a drug Combination medicine with other pharmaceutical excipients.
  • a transdermal drug delivery formulation prepared according to the fluorinated modified chitosan derivative used as a drug carrier includes a transdermal formulation component (a), and the component (a) is fluorine-containing A cationic polymer modified by a compound, the fluorine-containing compound modified cationic polymer is a fluorinated chitosan, the fluorine-containing compound is covalently attached to the chitosan backbone, and the molecular weight of the chitosan ranges from 1,000 to 5,000,000 , The degree of deacetylation is not less than 55% and the viscosity range is 25-1000 centipoise.
  • a transmucosal drug delivery formulation prepared according to the fluorinated modified chitosan derivative used as a drug carrier includes a transmucosal formulation component (a), and the component (a) contains A cationic polymer modified by a fluorine compound, the fluorine-containing compound modified cationic polymer is a fluorinated chitosan, the fluorine-containing compound is covalently attached to the chitosan backbone, and the molecular weight of the chitosan is in the range of 1000- 5000000, the degree of deacetylation is not less than 55% and the viscosity is in the range of 25-1000 centipoise.
  • the mucosa includes nasal mucosa, lung mucosa, vaginal mucosa, oral mucosa, gastrointestinal mucosa.
  • an ocular barrier-penetrating drug delivery formulation prepared according to the fluorinated modified chitosan derivative used as a drug carrier comprising component (a) of the ocular barrier-penetrating formulation, said group Part (a) is a cationic polymer modified by a fluorine-containing compound, the cationic polymer modified by a fluorine-containing compound is a fluorinated chitosan, and the fluorine-containing compound is covalently attached to the chitosan backbone.
  • the ocular barriers are tear fluid barrier, corneal/conjunctival barrier, blood humor barrier, blood retinal barrier.
  • a transdermal vaccine carrier prepared according to the fluorinated modified chitosan derivative used as a drug carrier includes a transdermal vaccine carrier (a), and the transdermal vaccine carrier (a) contains A cationic polymer modified by a fluorine compound, the fluorine-containing compound modified cationic polymer is a fluorinated chitosan, the fluorine-containing compound is covalently attached to the chitosan backbone, and the molecular weight of the chitosan is in the range of 1000- 5000000, the degree of deacetylation is not less than 55%, and the viscosity is in the range of 25-1000 centipoise.
  • the transdermal vaccine carrier has three antigen penetration pathways: intracellular penetration, intercellular penetration and hair follicle penetration.
  • a medical beauty and health care product carrier made according to the fluorinated modified chitosan derivative used as a drug carrier including the medical beauty and health care product carrier (a), the medical beauty and health care product
  • the carrier (a) is a cationic polymer modified by a fluorine-containing compound, and the cationic polymer modified by a fluorine-containing compound is a fluorinated chitosan, and the fluorine-containing compound is covalently attached to the chitosan backbone.
  • the molecular weight range is 1000-5000000, the deacetylation degree is not less than 55%, and the viscosity range is 25-1000 centipoise.
  • the medical beauty and health care product carrier can be suitable for hair growth drugs, hair care drugs, beauty drugs, and health care drugs.
  • a method for preparing a fluorinated modified chitosan derivative which includes the following steps: preparing a chitosan acetic acid aqueous solution, weighing the chitosan into the acetic acid aqueous solution, stirring to make it fully dissolved, and then adding sodium hydroxide dropwise. Stir until the solution is clear and the pH is 6.2-6.8;
  • the activated fluorine-containing compound solution is added dropwise to the rapidly stirring chitosan solution, and the reaction is fully reacted while avoiding light and stirring.
  • the method for preparing a fluorinated modified chitosan derivative further includes the following steps: slowly adding the fully reacted solution to the potassium hydroxide ethanol solution and stirring, filtering the precipitate, and rinsing with a large amount of absolute ethanol until the filtrate is Neutral, the precipitate is washed and dehydrated with methanol and ether, and dried in vacuum. The dried precipitate is dissolved in a hydrochloric acid solution and lyophilized to obtain fluorinated chitosan hydrochloride.
  • Figure 1-14 is a synthetic route diagram of a fluorine-containing carboxylic acid modified chitosan bladder infusion drug carrier.
  • a method for preparing 3-fluorobenzoic acid fluorinated chitosan which comprises the following steps:
  • the method for preparing 3-fluorobenzoic acid fluorinated chitosan further comprises the following steps:
  • the dried precipitate is dissolved in a hydrochloric acid solution and lyophilized to obtain a molecule of 3-fluorobenzoic acid fluorinated chitosan hydrochloride.
  • a method for preparing perfluoroheptanoic acid fluorinated chitosan which comprises the following steps:
  • the method for preparing perfluoroheptanoic acid fluorinated chitosan further comprises the following steps:
  • the fluorinated modified chitosan derivative used as a drug carrier is perfluoroheptanoic acid fluorinated chitosan hydrochloride molecule, and the perfluoroheptanoic acid
  • the degree of fluorination modification of fluorinated chitosan hydrochloride is 18%-25%, or 20%-22%.
  • a drug complex including a fluorinated modified chitosan derivative used as a drug carrier and a drug.
  • the drug includes a small molecule drug, a polypeptide, a protein drug, a combination drug of different drugs, and a combination of drugs and other pharmaceutical excipients. Combination drugs.
  • FCS fluorinated modified chitosan
  • FCS can significantly reduce the SV-HUC-1 monolayer cell membrane resistance, increase the permeation efficiency of fluorescent yellow, and regulate the tight junctions of cells by changing the structure and spatial distribution of tight junction proteins and E-cadherin proteins, and increase drugs Cellular bypass uptake efficiency of molecules.
  • FCS can effectively increase the gap between cells in biological tissue barriers (such as mucosal epithelial tissue, etc.), so that free drug molecules or drugs carried by FCS can more effectively traverse these biological tissue barriers.
  • FCS is selected as a new type of drug carrier for further research.
  • This patent designs and synthesizes a series of fluorinated modified chitosan derivatives, and its applications in the pharmaceutical field include but are not limited to the following disease models, such as bladder cancer perfusion (or other intracavity perfusion), lung inhalation administration , Transdermal administration, oral administration, ocular administration, vaccine administration, nasal administration.
  • Bladder cancer is one of the most common urinary tumors. Clinically, more than 75% of bladder cancers are non-muscular invasive bladder cancer (NMIBC). Among them, 30% to 80% of NMIBC patients undergo transurethral bladder tumor resection (TURBT). Recurrence within 5 years, and 10%-20% of NMIBC patients progress to muscular invasive bladder cancer. Therefore, adjuvant infusion chemotherapy or immunotherapy after TURBT to inhibit or delay tumor recurrence has become the first choice for clinical treatment guidelines for bladder cancer.
  • NMIBC non-muscular invasive bladder cancer
  • TURBT transurethral bladder tumor resection
  • the lung Compared with intravenous administration, pulmonary inhalation therapy locally delivers the drug to the tumor tissue, the required drug dose is significantly reduced, and the side effects are small.
  • the special physiological structure of the lung determines the characteristics and advantages of lung inhalation drug delivery: the lung has a large surface area, abundant capillaries, and thin alveolar epithelial cell layer, which makes lung drug delivery quicker; the distribution of biological metabolic enzymes in the lungs Concentration, low biological activity, thereby reducing the hydrolysis of proteins, making proteins and peptides easily absorbed through the surface of the alveoli, maintaining their biological activity; avoiding the liver first-pass effect.
  • the inhaled drugs are quickly cleared from the lungs, and the effective deposition of the drugs in the lungs cannot be guaranteed.
  • lung cancer although the drugs inhaled from the lungs can reach the alveoli, the efficiency of entering the lung tumors is generally very low, which seriously affects the efficacy of the lung inhaled administration mode for the treatment of lung cancer.
  • FCS transmucosal drug carrier
  • the transdermal drug delivery system refers to a preparation that is administered on the surface of the skin, and the drug passes through each layer of the skin at a certain rate and enters the systemic circulation to produce a systemic or local therapeutic effect.
  • transdermal drug delivery has many advantages such as easy operation and strong patient adaptability.
  • transdermal delivery is usually restricted by the stratum corneum barrier of the skin and the physical and chemical properties of the drug. How to improve the ability of the drug to enter the blood circulation or enter the subcutaneous lesions (such as skin cancer) through the transdermal delivery mode is the technology faced by this technology. An important challenge.
  • FCS transmucosal drug carrier
  • the present invention proposes a compound including chitosan modified by a fluorine-containing compound and a drug.
  • the drug includes a small molecule drug, a polypeptide, a protein drug, a combination drug of different drugs, and a combination drug of a drug and other pharmaceutical excipients, and Use in promoting drug absorption.
  • epirubicin THP
  • fluorine-containing compound-modified chitosan prepared by the present invention is used as a drug transport carrier to promote the drug to enter the bladder tissue.
  • the present invention has the following advantages: the present invention maintains good biocompatibility while maintaining significant bladder mucosal absorption efficiency of the perfusion-promoting drug.
  • the bladder perfusion drug carrier provided by the invention has the advantages of high efficiency, low toxicity, low price, simple synthesis and the like.
  • a pharmaceutical composition adopting the transdermal administration preparation comprising a transdermal preparation component (a) and a drug component (b), and the component (b) is a diabetes treatment drug and an anti-tumor drug , Immunomodulators, antiviral drugs, anti-inflammatory drugs, analgesic anesthetics.
  • a drug complex for the treatment of diabetes using the transdermal drug formulation is characterized in that it comprises fluorinated chitosan and insulin, and the mass ratio of the fluorinated chitosan to insulin is 1:0.25 -4.
  • the drug complex for treating diabetes using a transdermal drug delivery formulation is characterized in that: the fluorinated chitosan and insulin adsorb each other to form a complex, and the particle size range of the complex is less than 10 microns, Or the particle size range of the complex is not greater than 500 nanometers, and the mass ratio of the fluorinated chitosan to insulin is 1:0.5-2.
  • a method for preparing the drug complex for treating diabetes by adopting the transdermal administration mode characterized in that:
  • the method for preparing a drug complex for the treatment of diabetes by means of transdermal administration is characterized in that:
  • reaction mass ratio of the fluorinated chitosan and insulin is 1:0.25-4 or 1:0.5-2;
  • the pharmaceutical composition is taken out, the lyophilized protective agent is pre-added and then lyophilized to obtain the fluorinated chitosan-insulin lyophilized powder.
  • a transdermal application of a pharmaceutical composition for the treatment of diabetes using the transdermal drug formulation is characterized in that: the fluorinated chitosan and insulin are mutually adsorbed to form a complex, and the fluorinated chitosan is The mass ratio of sugar to insulin is 1:0.25-4, which is uniformly mixed with the hydrogel to obtain a transdermal patch.
  • a drug complex for treating melanoma using the transdermal drug delivery formulation is characterized in that: the fluorinated chitosan forms a complex with an apoptosis-ligand 1 antibody, and the complex
  • the particle size range is less than 10 microns, or the particle size range of the complex is not greater than 500 nanometers, and the reaction mass ratio of the fluorinated chitosan and the apoptosis-ligand 1 antibody is 1:0.25-4.
  • the drug complex for treating melanoma is characterized in that the reaction mass ratio of the fluorinated chitosan and the apoptosis-ligand 1 antibody is 1:0.25-4 or 1:1, so The fluorinated chitosan-apoptosis-ligand 1 antibody aqueous solution is mixed with petrolatum ointment to form a fluorinated chitosan-apoptosis-ligand 1 antibody transdermal ointment.
  • a transdermal drug delivery formulation comprising component (a), the component (a) is a cationic polymer modified with a fluorine-containing compound, and the cationic polymer modified with a fluorine-containing compound can be used as a medicine for treating diabetes , Transdermal drug delivery preparations for tumor disease drugs and anti-inflammatory drugs.
  • a preparation for transdermal administration comprising component (a), the component (a) is a cationic polymer modified with a fluorine-containing compound, and the cationic polymer modified with a fluorine-containing compound is used for transdermal administration
  • the application of the preparation in the preparation of medical cosmetology drugs, external drug preparations, medical device external preparations, and cosmetic skin care products.
  • a penetration enhancer to the patch to promote the penetration of the drug through the skin.
  • penetration enhancers include alcohols such as ethanol and butanol, dimethyl sulfoxide, laurin, pyrrolidone, etc. Derivatives, surfactants and fatty acid compounds, etc.
  • the penetration-promoting mechanism of alcohols and pyrrolidone derivatives is mainly through swelling lipids in the stratum corneum to increase the solubility of the drug; azepines by changing the compactness of the lipid bilayer, increasing the lipid profile Fluidity promotes drug penetration; when fatty acid compounds are inserted into the hydrophobic structure of the lipid bilayer through the cis structure of their unsaturated hydrophobic chains, the lipid bilayer is twisted and a very fine pore is formed, which makes the drug Be able to diffuse into.
  • the physical penetration enhancement methods mainly include iontophoresis, ultrasonic method, electroporation method and microneedle method.
  • the physical penetration enhancement method is mainly used for drugs that are difficult for chemical penetration enhancers, such as peptides, proteins and other macromolecular drugs and ionic drugs.
  • Iontophoresis is to increase the penetration rate of the drug by applying an appropriate electric field on the skin surface. Due to the existence of the electric field, the interaction of ions in the electric field, the convective movement of the solvent under the electric field and the increase in skin permeability caused by the electric current will all Promote the transdermal absorption of drugs.
  • Ultrasound is to promote the penetration of drug molecules into the skin under the action of ultrasound. The possible mechanisms are: 1. Local thermal effects increase the permeability of the drug; 2. Local radiation pressure causes the drug to move in the direction of sound waves to promote drug penetration; 3.
  • the microfluidic effect enables the drug to enter the skin through hair follicles and sweat glands; 4. Cavitation causes disordered arrangement of the skin stratum corneum, which promotes drug penetration.
  • the electroporation law promotes the absorption of drugs through the skin through a pulsed electric field.
  • the currently reported electroporation mechanism is that under a pulsed electric field, lipid molecules in the skin are re-arranged in an orderly manner to form a new channel to promote drug penetration. After the pulsed electric field is over, the lipid molecules recover their previous Arrange out of order, thereby closing the channel.
  • Microneedles use micron-level microneedles to form very small wounds on the skin to efficiently promote drug penetration.
  • the cationic polymer modified by the fluorine-containing compound described in this patent can increase the penetration ability of the drug in the skin, while reducing the administration area, thereby reducing the toxic and side effects that the drug may have on normal skin.
  • the technical solution described in this patent does not have strong volatility and irritation as an aqueous solution, has a fast action speed and a long duration, and can promote a series of drug molecules Through the skin, including small molecule drugs, peptides, protein macromolecular drugs, ionic drugs, etc.
  • the patented technical solution does not require an external electric field, ultrasound, etc., which greatly reduces the trauma, pain, inconvenience and safety hazards brought to the patient. Therefore, the fluorinated chitosan disclosed in the examples of this patent can be used as transdermal preparations and used in combination with other drugs.
  • the cationic polymers modified by fluorine-containing compounds have been modified by fluorine-containing compounds. It can be used as a transdermal preparation and used in combination with other drugs.
  • the fluorinated chitosan drug carrier provided by this patent has the advantages of obvious effect of promoting drug absorption, low toxicity, etc., and the fluorine-containing compound modified chitosan proposed by the present invention has a mature synthesis process, simple operation, high synthesis efficiency, and short cycle. , Without cumbersome purification steps, high-yield drug carriers can be obtained, and its simple synthesis method provides a good basis for commercialization.
  • the fluorine-containing compound modified chitosan of the present invention is useful as a variety of drug carriers. Uses, can effectively improve the therapeutic effect, have a wide range of uses, and low cost.
  • the skin is composed of the epidermis and the dermis.
  • the epidermis is the stratum corneum, the transparent layer, the granular layer and the germinal layer from shallow to deep.
  • the dermis is composed of dense connective tissue, from shallow to deep into the papillary layer and the reticular layer.
  • the papillary layer is connected to the germinal layer of the epidermis, which is rich in capillaries, lymphatic vessels, nerve endings, and tactile corpuscles.
  • the stratum corneum is the largest restrictive barrier for transdermal drug delivery.
  • stratum corneum consists of 5-25 layers of flat keratinocytes. These cells have no nucleus, organelles, and thicker cell membranes. They are inanimate and impermeable to prevent tissue fluids. Outflow, anti-friction and anti-infection functions. Cationic polymers modified with fluorine compounds can stimulate the distribution of tight junction proteins in these cells, reduce tight junctions between cells, and further stimulate actin phosphorylation, thereby promoting paracellular transport, opening intercellular spaces, forming channels, and carrying The drug penetrates the stratum corneum, further penetrates into the skin, and then enters the dermis, enters the skin capillaries and lymphatic circulation, and exerts the effect of the drug (see Figure 1).
  • the small intestine is the main part of drug absorption. Before drugs are absorbed from the small intestine into the blood circulation, they will inevitably face three main gastrointestinal physiological barriers, as shown in Figure 3-1, including protease barrier, mucus barrier and intestinal epithelial cell barrier. There are abundant proteases in the small intestine, including trypsin, chymotrypsin, elastase and carboxypeptidase. Protein peptide drugs will be rapidly degraded and inactivated under the action of various proteases. There is also a mucus barrier in the intestine.
  • the surface of the intestinal epithelial cells is covered with a layer of mucus of varying thickness, which is composed of 95% water and 5% electrolytes, lipids, proteins and glycoproteins.
  • the mucus layer has viscoelasticity and plays a protective role.
  • the mucus barrier is also one of the main barriers that affect the oral absorption of protein peptide drugs.
  • the intestinal epithelial cell layer mainly includes two types of cells: epithelial cells (intestinal epithelial cells) that can absorb substances and epithelial cells that are difficult to absorb substances. These cells are connected to each other through tight connections, forming a relatively impermeable barrier that severely limits the absorption of foreign substances.
  • fluorinated chitosan as a carrier can not only solve the above problems, but also increase the penetration of the drug, so that the drug can reach the blood or the treatment site efficiently, and the utilization of the drug can be improved.
  • fluorinated chitosan explains the mechanism of mucosal penetration.
  • fluorinated chitosan when fluorinated chitosan is used as a drug carrier to deliver insulin, it can open epithelial channels without disrupting the function of intestinal epithelial cells to secrete tight junction proteins, as shown in Figure 3-2.
  • fluorinated chitosan As the main body, it can be made into corresponding oral drugs such as solutions, syrups, granules, capsules, powders, pills, and tablets. After oral administration, the drug is absorbed into the blood by the gastrointestinal tract, and reaches the local or systemic tissues through blood circulation to achieve the purpose of curing diseases.
  • protein and peptide drugs are easily degraded by digestive enzymes in the gastrointestinal tract.
  • the molecular weight is large and the molecules have a strong tendency to polymerize before. It is difficult to play a sufficient role through the body barrier. If they are directly orally absorbed without treatment, they The bioavailability is only 0.5%.
  • fluorinated chitosan as a carrier can not only maintain the activity of protein and peptide drugs, and stabilize their configuration; reduce the formation of multimers, which facilitates increasing the absorption of the intestinal mucosa; reduce the degradation of protein and peptide drugs by digestive enzymes At the same time, it can also open the epithelial channel without destroying the function of intestinal epithelial cells to secrete tight junction proteins, which can increase the penetration of the drug in the small intestine, facilitate the drug to reach the blood or the treatment site efficiently, and improve the utilization of the drug.
  • take insulin and apoptosis-ligand 1 antibody as example drugs to realize the application of fluorine-containing compound modified cationic polymers as drug carriers for oral administration.
  • the drug may be diabetes drugs, anticolitis drugs, anesthetics, anti-inflammatory drugs, antibacterial drugs, antiviral drugs, antiparasitic drugs, and the like.
  • the diabetes drugs may be sulfonylureas, including but not limited to: sulfonamide, tolbutamide, chlorpropamide, chlorhexamide, gliclazide, glipidin, glimepiride, etc. And its derivatives.
  • the diabetes drugs may be non-sulfonylureas, including but not limited to: repaglinide, nateglinide and other drugs and their derivatives.
  • the diabetes drugs may be thiazolidinediones, including but not limited to: rosiglitazone, pioglitazone and other drugs and their derivatives.
  • the diabetes drug may be a biguanide, including but not limited to: phenformin, metformin and other drugs and their derivatives.
  • the diabetes drug may be an ⁇ -glucosidase inhibitor, including but not limited to: drugs such as acarbose, voglibose, miglitol, and derivatives thereof.
  • the diabetes drug may be dipeptidyl peptidase-IV including but not limited to: glucagon-like peptide, DPP-IV inhibitor sitagliptin, vildagliptin, saxagliptin.
  • anti-diabetic drugs include but are not limited to insulin.
  • fluorinated chitosan can be used as a drug carrier to deliver insulin and be administered as an oral drug for the treatment of diabetes.
  • the invention can solve the problem of low oral utilization rate of insulin.
  • Oral administration is the most easily accepted route of administration by patients.
  • insulin is easily degraded by various enzymes in the gastrointestinal tract, and the molecular weight of insulin itself is relatively large, and it is difficult to be absorbed by the epithelial cell membrane of the gastrointestinal tract.
  • the oral administration of insulin through the fluorinated chitosan drug carrier can not only protect the activity of insulin, but also promote the passage of insulin through the small intestinal mucosa and improve the availability of drugs.
  • Analgesics are drugs that act on the central nervous system and selectively inhibit the pain center, reducing or eliminating pain without affecting other sensations. They are mainly used clinically for trauma, burns, post-operation and cancer. The sharp pain relieves the pain. However, many analgesics cannot quickly relieve pain and are administered locally. The present invention can solve this problem.
  • the analgesic may be morphine and its derivatives including but not limited to: codeine, ethylmorphine, benzylmorphine, iscodeine, heroin, phenethylmorphine, hydromorphone, oxymorphone, propylene Morphine, nalorphine, naloxone, naltrexone and other drugs and their derivatives.
  • the analgesics may be synthetic analgesics including but not limited to: pethidine, aniridine, benperiidine, piminodine, afarotidine, betarotidine, fentanyl, alfenac Tanyl, Sufentanil, Remifentanil, Pethidine Hydrochloride, Methadone, Dexmorphine, Dexpropoxyphene, Methadone Hydrochloride, N-Methylmorphinan, Levophanol, Butorphanol, Morphine Nang, pentazocine, dezocine, sumatriptan, tramadol, fenthiidine, nefopam.
  • Anesthetics refer to drugs that can temporarily and reversibly lose consciousness and pain in the entire body or part of the body.
  • most of the anesthetic drugs are intravenous injection and intraspinal injection, which increase the patient's pain compared to oral administration.
  • the present invention can increase patient compliance through oral anesthetics.
  • the anesthetics may be intravenous anesthetics including but not limited to: ketamine hydrochloride, propofol, thiopental sodium, etomidate, midazolam and sodium gamma-hydroxybutyrate.
  • the anesthetics may be local anesthetics including but not limited to: aromatic acid esters, aromatic amides, amino ketones, amino ethers, carbamates, hydroxyprocaine, chloroprocaine, tetracaine Because, butacaine, thiocaine, procainamide, bupivacaine, articaine, eticaine, ropivacaine, mepivacaine, clonin hydrochloride and other drugs and Its derivatives.
  • Inflammation is closely related to the occurrence and development of many major diseases such as cardiovascular and cerebrovascular diseases, neurodegenerative diseases and even tumors. Therefore, early imaging, diagnosis and anti-inflammatory strategies for inflammation are important means to prevent and treat many diseases.
  • most anti-inflammatory drugs are administered through intravenous injection and oral administration.
  • intravenous anti-inflammatory drugs can have serious side effects, and the bioavailability of oral anti-inflammatory drugs is extremely low.
  • the invention can improve the oral utilization rate of anti-inflammatory drugs and make patients more comply.
  • the anti-inflammatory drug may be a non-steroidal anti-inflammatory drug including but not limited to: aspirin, acetaminophen, non-specific cyclooxygenase inhibitors, antipyrine, analgin, butazepine, oxybutazone , Mefenamic acid, indomethacin, sulindac, diclofenac sodium, ibuprofen, propanaxan, piroxicam, meloxicam and other non-specific cyclooxygenase inhibitor drugs and their derivatives.
  • a non-steroidal anti-inflammatory drug including but not limited to: aspirin, acetaminophen, non-specific cyclooxygenase inhibitors, antipyrine, analgin, butazepine, oxybutazone , Mefenamic acid, indomethacin, sulindac, diclofenac sodium, ibuprofen, propanaxan, piroxicam, meloxicam and
  • the anti-inflammatory drug may be a steroidal anti-inflammatory drug, including but not limited to: hydrocortisone, corticosterone, aldosterone, triamcinolone, prednisolone, dexamethasone acetate, prednisone acetonide and other drugs And its derivatives.
  • a steroidal anti-inflammatory drug including but not limited to: hydrocortisone, corticosterone, aldosterone, triamcinolone, prednisolone, dexamethasone acetate, prednisone acetonide and other drugs And its derivatives.
  • the embodiments of the present invention can be used to treat various inflammations, including but not limited to: myocarditis, arthritis, colitis, tonsillitis, gastritis, rhinitis, periodontitis, enteritis, pharyngitis, prostatitis, vaginitis , Cervicitis, frozen shoulder, cervical spondylitis, bursitis, dermatitis, conjunctivitis, otitis media and so on.
  • the colon is an important part of the digestive system, mainly involved in the absorption of water, vitamins and inorganic salts, and is also an important place for the formation of feces.
  • the intestinal wall of the colon can be divided into serosal layer, longitudinal muscle layer, circular muscle layer, submucosal layer and mucosal layer from the outside to the inside.
  • the mucosal layer serves as the body's first line of defense and can protect the intestine from harmful substances in the intestinal cavity such as bacteria and The invasion of toxins.
  • the normal colonic mucosal barrier consists of mechanical barriers, chemical barriers, immune barriers and biological barriers.
  • mechanical barriers include colonic epithelial cells and connections between epithelial cells, which are an important part of the mucosal barrier; chemical barriers mainly include mucosal epithelial secretion Antibacterial substances produced by the intestinal mucus, digestive juice and intestinal protobacteria;
  • the immune barrier is composed of the lymphatic tissue of the intestinal mucosa and the antibodies secreted by the intestinal tract.
  • the lymphatic tissue can protect the intestine from some immune cells through cellular and humoral immunity.
  • the invasion of pathogenic antigens; the biological barrier is the normal commensal flora of the intestinal tract, and an interdependent microecosystem is formed between the intestinal commensal flora and the host.
  • the colonic epithelial cell barrier is an important part of the intestinal mechanical barrier. It is composed of colonic epithelial cells and cell connections between epithelial cells. These cell connections include tight junctions, gap junctions, and adhesion junctions. They work together to seal cells.
  • the gap in which the role of tight junctions is very important, is composed of connecting adhesion molecules, Occludin, Claudin, and ZO-1. Cell connection can prevent macromolecular substances such as endotoxin in the intestinal cavity from entering the intestinal tissue.
  • fluorinated chitosan as a carrier can effectively overcome the colon-related mechanical barriers, chemical barriers, immune barriers and biological barriers, improve the permeability of drugs in the colon, and enhance the availability of drugs. Compared with intravenous injection or intraperitoneal injection, more drugs will penetrate the affected area.
  • fluorinated chitosan when fluorinated chitosan is used as a drug carrier to deliver insulin, it can open epithelial channels without disrupting the function of intestinal epithelial cells to secrete tight junction proteins, as shown in Figure 3-2.
  • fluorinated chitosan can be used as a drug carrier to deliver anti-tumor drugs such as immune checkpoint blocking antibodies, which can be administered in the form of oral drugs for tumor treatment.
  • Colon cancer is one of the most common gastrointestinal tumors, with high morbidity and mortality.
  • the apoptosis-ligand 1 antibody has been successful in the treatment of colorectal cancer, the apoptosis-ligand 1 antibody is administered by intravenous injection, which may cause serious adverse reactions such as gastrointestinal toxicity.
  • the drug is less enriched in the colorectal region.
  • Oral administration is the most acceptable method of administration by patients, which can deliver most of the drugs to the diseased site and reduce adverse reactions.
  • the apoptosis-ligand 1 antibody is easily inactivated due to the harsh environment in the digestive tract, and the molecular weight of the apoptosis-ligand 1 antibody is large, which makes it difficult to cross the intestinal epithelial cells.
  • Oral administration of the apoptosis-ligand 1 antibody through the fluorinated chitosan drug carrier can make the apoptosis-ligand 1 antibody pass through the colorectal mucosa and improve the availability of the drug.
  • IBD Inflammatory bowel disease
  • GI gastrointestinal tract
  • IBD consists of two main clinically defined forms, Crohn's disease (CD) and ulcerative colitis (UC).
  • CD usually affects the ileum and colon, but it can discontinuously affect any area of the gastrointestinal tract, and the inflammation is usually transmural.
  • UC ulcerative colitis
  • Both CD and UC are associated with high morbidity and decreased quality of life.
  • the incidence of IBD is increasing on a global scale, putting a heavy burden on public health care.
  • Oral administration as the most easily accepted method of administration by patients, can deliver most of the anticolitis drugs to the affected area and reduce the inconvenience caused by anal plugs.
  • the anticolitis drug may be a small molecule drug including but not limited to: budesonide, beclomethasone, 5-aminosalicylic acid, superoxide dismutase, 4-aminotempol, catalase, methotrexate Retinol, rolipram, adriamycin, vancomycin, colistine sulfate, suromycin, lamotulin, LFF-571, berberine, bilirubin, gallic acid, catechol and its derivatives Things.
  • the anti-colitis drug may be a protein drug including but not limited to: leukemia inhibitory factor, transforming growth factor ⁇ , ovalbumin and derivatives thereof.
  • the anti-colitis drug may be small interfering ribonucleic acid (siRNA) and antisense oligonucleotides including but not limited to: antisense oligonucleotides for tumor necrosis factor- ⁇ (TNF- ⁇ ), abciximab -TNF messenger ribonucleic acid-15 (miRNA-155) inhibitor, tumor necrosis factor- ⁇ siRNA, CD98 siRNA, cyclin D1 siRNA, prolyl hydroxylase 2 siRNA, Map4k4 siRNA, cleavage oligonucleotide, IL10-producing plasmid, NF -kB decoy oligonucleotide, pcDNA3-EGFP, peptide antigen (PeptAg) and its derivatives.
  • TNF- ⁇ tumor necrosis factor- ⁇
  • miRNA-155 abciximab -TNF messenger ribonucleic acid-15
  • cyclin D1 siRNA cyclin D1 siRNA
  • the anti-colitis drug may be probiotics including but not limited to: Lactobacillus casei (ATCC 39392), Lactococcus lactis, Bacillus ovale, Vibrio cholerae, probiotic VSL-3 or its isolated DNA, Escherichia coli .
  • Colorectal cancer is one of the most common malignant tumors in the world.
  • the treatment of colorectal cancer mainly relies on surgery, chemotherapy and immunotherapy.
  • chemotherapy and immunotherapy are usually administered by intravenous injection.
  • oral administration has the highest patient acceptance and compliance among different routes, clinically used chemotherapy products developed in oral formulations are rare. This is due to the limited and unstable absorption of drugs from the gastrointestinal tract (GI) and the liver's first-pass effect on drug metabolism, which is closely related to the reduction of the therapeutic anti-tumor effect of oral preparations.
  • GI gastrointestinal tract
  • liver's first-pass effect on drug metabolism which is closely related to the reduction of the therapeutic anti-tumor effect of oral preparations.
  • the invention can increase the oral availability of the medicine.
  • the anti-colorectal cancer drug may be a bioalkylating agent including, but not limited to: chlorambucil, chlorambucil, melphalan, prenimustine, cyclophosphamide, cetepa, carmustine , Baisulfan, cisplatin, carboplatin and other drugs and their derivatives.
  • the anti-colorectal cancer drug may be an anti-metabolic drug including but not limited to: fluorouracil, cytarabine, mercaptopurine, methotrexate and other drugs and their derivatives.
  • the anti-colorectal cancer drug may be an anti-tumor antibiotic including but not limited to drugs such as actinomycin D, doxorubicin (adriamycin), zorubicin, mitoxantrone and derivatives thereof.
  • the anti-colorectal cancer drug may be active ingredients of traditional Chinese medicines, including but not limited to: 10-hydroxycamptothecin, vinblastine sulfate, paclitaxel, docetaxel and other natural medicine active ingredients and derivatives thereof.
  • the anti-colorectal cancer drug can be an immune spot check inhibitor. See Table 3-1.
  • the anti-colorectal cancer drug can be a cytokine, including but not limited to: cytokine is produced by immune cells (such as monocytes, macrophages, T cells, B cells, NK cells, etc.) and certain non-immune cells (endothelial cells, epidermal cells, fibroblasts, etc.) are stimulated to synthesize and secrete a class of small molecular proteins with a wide range of biological activities.
  • immune cells such as monocytes, macrophages, T cells, B cells, NK cells, etc.
  • non-immune cells endothelial cells, epidermal cells, fibroblasts, etc.
  • Cytokines include but are not limited to interleukin (IL), interferon (IFN), tumor necrosis factor (TNF), colony stimulating factor (CSF), chemokine (chemokine family), growth factor (growth factor, GF), transforming growth factor- ⁇ family (transforming growth factor- ⁇ family, TGF- ⁇ family).
  • Interleukins include but are not limited to IL-1-IL-38.
  • Colony stimulating factors include but are not limited to G (granulocyte)-CSF, M (macrophage)-CSF, GM (granulocyte, macrophage)-CSF, Multi (multiple)-CSF (IL-3), SCF, EPO etc.
  • Interferons include, but are not limited to, including but not limited to IFN- ⁇ , IFN- ⁇ , and IFN- ⁇ .
  • Tumor necrosis factors include but are not limited to TNF- ⁇ and TNF- ⁇ .
  • the transforming growth factor- ⁇ family includes, but is not limited to, TGF- ⁇ 1, TGF- ⁇ 2, TGF- ⁇ 3, TGF ⁇ 1 ⁇ 2, and bone morphogenetic protein (BMP).
  • Growth factors include but are not limited to epidermal growth factor (EGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), insulin-like growth factor-I (IGF-1) , IGF-II, Leukemia Inhibitory Factor (LIF), Nerve Growth Factor (NGF), Oncostatin M (OSM), Platelet-derived Endothelial Growth Factor (PDECGF), Transforming Growth Factor- ⁇ (TGF- ⁇ ), Vascular Endothelial cell growth factor (VEGF).
  • EGF epidermal growth factor
  • PDGF platelet-derived growth factor
  • FGF fibroblast growth factor
  • HGF hepatocyte growth factor
  • IGF-II insulin-like growth factor-I
  • LIF Leukemia Inhibitory Factor
  • NGF Nerve Growth Factor
  • OSM Oncostatin M
  • PDECGF Platelet-derived Endothelial Growth Factor
  • TGF- ⁇ Vascular Endothelial
  • the chemokine family includes, but is not limited to, four subfamilies: (1) CXC/ ⁇ subfamily, mainly chemotactic neutrophils, the main members are IL-8, melanoma cell growth stimulating activity (GRO/MGSA) , Platelet factor-4 (PF-4), platelet basic protein, proteolytic products CTAP-III and ⁇ -thromboglobulin, inflammatory protein 10 (IP-10), ENA-78; (2) CC/ ⁇ subfamily , Mainly chemotactic monocytes, members of this subfamily include macrophage inflammatory protein 1 ⁇ (MIP-1 ⁇ ), MIP-1 ⁇ , RANTES, monocyte chemotactic protein-1 (MCP-1/MCAF), MCP- 2. MCP-3 and I-309.
  • CXC/ ⁇ subfamily mainly chemotactic neutrophils
  • the main members are IL-8, melanoma cell growth stimulating activity (GRO/MGSA) , Platelet factor-4 (PF-4), platelet basic protein, proteolytic products CTAP-
  • Cytokines include, but are not limited to, cytokines used to treat cancer and cytokines that reduce the side effects of cancer treatment. They play an important role in the normal immune response of the human body and the ability of the immune system to respond to cancer. Cytokines used to treat cancer include but are not limited to interferons and interleukins. Cytokines can also be hematopoietic growth factors, which reduce the side effects of cancer treatment by promoting the growth of blood cells destroyed by chemotherapy.
  • Cytokines that reduce the side effects of cancer treatment include but are not limited to erythropoietin, IL-11, granulocyte-macrophage colony stimulating factor (GM-CSF) and granulocyte-colony stimulating factor (G-CSF).
  • BCG Vaccine is a live vaccine made from a suspension of attenuated Mycobacterium bovis, which can enhance the activity of macrophages, enhance the body's cellular immunity, and can be used to treat bladder cancer.
  • Immunomodulatory drugs including but not limited to thalidomide Lenalidomide Pomali Imiquimod
  • the oral mucosal drug delivery system has many advantages compared with the traditional drug delivery system: 1. It can maintain a long-lasting, constant and controllable blood drug concentration, thereby reducing adverse reactions; 2. Avoiding the first pass effect of the liver and the interference of gastrointestinal factors And degradation, improve the bioavailability of the drug; 3. Relieve the pain of injection medication and improve patient compliance; 4. If there is a problem after the administration, the drug can be stopped in time, which is convenient to use; 5. Reduce the number of administrations and doses, and reduce the drug The incidence of adverse reactions. Therefore, oral mucosal drug delivery systems have received more and more attention.
  • the drug delivery system cannot be too large; involuntary saliva secretion and swallowing affect the efficacy of the oral mucosal pathway; the taste stimulation and foreign body sensation of the drug affect the compliance of this pathway; not all Substances can pass through the oral mucosa, and their absorption is affected by fat solubility, pH, and molecular weight.
  • Oral mucosa is a barrier during the transportation of drugs through the oral cavity.
  • the properties of membranes in different areas of the oral cavity are different.
  • the thickness and composition of the stratum corneum and non-stratum corneum tissues are also different.
  • the thickness of the membrane at the absorption site, blood flow, blood or lymph Perfusion, cell renewal, and enzyme content are all related to the oral mucosal absorption of drugs.
  • the oral mucosal barrier is composed of epithelial barrier, basement membrane barrier, and lamina intestinal. There is an epithelial barrier in the oral mucosal epithelium. The epithelial layer is the main barrier for drugs to pass through the mucosa.
  • the permeability of the keratinized epithelium is greater than that of the non-keratinized epithelium.
  • the main permeability barrier exists in the outermost epithelium from 1/4 to Up to 1/3, the thickness of the epithelium also has a great influence on the permeability, and the stratum corneum is the most important absorption barrier.
  • the basement membrane located at the junction of epithelial cells and lamina basement can also restrict the penetration of drugs.
  • the lipophilicity of the drug should not be too strong, the drug is difficult to dissolve in saliva, does not reach an effective level, and it is difficult to pass through the basement membrane, which affects the transmembrane absorption of the drug.
  • the lamina propria does not function as a permeability barrier. Blood vessels and nerves are in the lamina basement membrane, providing nutrition and innervation for the epithelium. However, some researchers believe that the lamina basement will also constitute a certain permeability barrier. Using fluorinated chitosan as a carrier can effectively overcome the epithelial barrier, stratum corneum barrier, and permeability barrier related to the oral mucosa, effectively increase the penetration rate of the drug, and facilitate the drug to actually reach the blood or the treatment site, and improve the utilization of the drug.
  • Preparations include mouthwashes, lozenges, sublingual tablets, films and other dosage forms.
  • it can treat diseases such as herpetic trigeminal/glossopharyngeal nerve pain, pharyngitis caused by inflammation, bacteria, viruses, oral ulcers, local anesthesia, etc.
  • sublingual venous plexus which can quickly enter the blood after penetrating the mucosa, avoiding the liver first-pass effect, and is mostly used for heart-related diseases.
  • Heart disease-related drugs may include, but are not limited to, specific drugs and their derivatives in Table 3-2 below.
  • Table 3-2 shows drugs related to heart disease
  • Analgesics are drugs that act on the central nervous system and selectively inhibit the pain center, reducing or eliminating pain without affecting other sensations. They are mainly used clinically for trauma, burns, post-operation and cancer. The sharp pain relieves the pain. However, many analgesics cannot quickly relieve pain and are administered locally. The present invention can solve this problem.
  • the analgesic may be morphine and its derivatives including but not limited to: codeine, ethylmorphine, benzylmorphine, iscodeine, heroin, phenethylmorphine, hydromorphone, oxymorphone, propylene Morphine, nalorphine, naloxone, naltrexone and other drugs and their derivatives.
  • the analgesics may be synthetic analgesics including but not limited to: pethidine, aniridine, benperiidine, piminodine, afarotidine, betarotidine, fentanyl, alfenac Tanyl, Sufentanil, Remifentanil, Pethidine Hydrochloride, Methadone, Dexmorphine, Dexpropoxyphene, Methadone Hydrochloride, N-Methylmorphinan, Levophanol, Butorphanol, Morphine Nang, pentazocine, dezocine, sumatriptan, tramadol, fenthiidine, nefopam.
  • Anesthetics refer to drugs that can temporarily and reversibly lose consciousness and pain in the entire body or part of the body.
  • most of the anesthetic drugs are intravenous injection and intraspinal injection, which increase the patient's pain compared to oral administration.
  • the present invention can increase patient compliance through oral anesthetics.
  • the anesthetics may be intravenous anesthetics including but not limited to: ketamine hydrochloride, propofol, thiopental sodium, etomidate, midazolam and sodium gamma-hydroxybutyrate.
  • the anesthetics may be local anesthetics including but not limited to: aromatic acid esters, aromatic amides, amino ketones, amino ethers, carbamates, hydroxyprocaine, chloroprocaine, tetracaine Because, butacaine, thiocaine, procainamide, bupivacaine, articaine, eticaine, ropivacaine, mepivacaine, clonin hydrochloride and other drugs and Its derivatives.
  • Inflammation is closely related to the occurrence and development of many major diseases such as cardiovascular and cerebrovascular diseases, neurodegenerative diseases and even tumors. Therefore, early imaging, diagnosis and anti-inflammatory strategies for inflammation are important means to prevent and treat many diseases.
  • most anti-inflammatory drugs are administered through intravenous injection and oral administration.
  • intravenous anti-inflammatory drugs can have serious side effects, and the bioavailability of oral anti-inflammatory drugs is extremely low.
  • the invention can improve the oral utilization rate of anti-inflammatory drugs and make patients more comply.
  • the anti-inflammatory drug may be a non-steroidal anti-inflammatory drug including but not limited to: aspirin, acetaminophen, non-specific cyclooxygenase inhibitors, antipyrine, analgin, butazepine, oxybutazone , Mefenamic acid, indomethacin, sulindac, diclofenac sodium, ibuprofen, propanaxan, piroxicam, meloxicam and other non-specific cyclooxygenase inhibitor drugs and their derivatives.
  • a non-steroidal anti-inflammatory drug including but not limited to: aspirin, acetaminophen, non-specific cyclooxygenase inhibitors, antipyrine, analgin, butazepine, oxybutazone , Mefenamic acid, indomethacin, sulindac, diclofenac sodium, ibuprofen, propanaxan, piroxicam, meloxicam and
  • the anti-inflammatory drug may be a steroidal anti-inflammatory drug, including but not limited to: hydrocortisone, corticosterone, aldosterone, triamcinolone, prednisolone, dexamethasone acetate, prednisone acetonide and other drugs And its derivatives.
  • a steroidal anti-inflammatory drug including but not limited to: hydrocortisone, corticosterone, aldosterone, triamcinolone, prednisolone, dexamethasone acetate, prednisone acetonide and other drugs And its derivatives.
  • the embodiments of the present invention can be used to treat various inflammations, including but not limited to: myocarditis, arthritis, colitis, tonsillitis, gastritis, rhinitis, periodontitis, enteritis, pharyngitis, prostatitis, vaginitis , Cervicitis, frozen shoulder, cervical spondylitis, bursitis, dermatitis, conjunctivitis, otitis media and so on.
  • the oral delivery vaccine can be coated with a protective shell to deliver antigens to the intestine to bind to intestinal-associated lymphoid tissues, to be captured and recognized by intestinal antigen-presenting cells and migrate to Peyer’s lymph nodes to trigger an immune response, or it can be directly contained in Oral vaccine delivery is performed in the mouth.
  • Oral vaccines include but are not limited to: live attenuated polio vaccine, rotavirus vaccine, typhoid vaccine, live dysentery vaccine, enteric typhoid vaccine, pertussis vaccine, rabies vaccine, diphtheria toxoid vaccine, tetanus toxoid vaccine, B chain Cholera toxin vaccine, E. coli enterotoxin vaccine, 3-deoxyacylated monophosphoryl lipid A vaccine, BCG polysaccharide nucleic acid vaccine, bacterial cell wall skeleton vaccine.
  • the technical solution of the patent can be used in an inhalation (pulmonary inhaler/nebulizer) drug delivery system, as follows.
  • fluorinated chitosan as the main body, combined with drugs can also be made into corresponding aerosol preparations/inhalation preparations to promote drug delivery across the lung mucosa.
  • Intravenous injection although as a conventional treatment route, is still difficult to effectively deliver drugs to the lung tissues. Intravenous drugs cannot be effectively deposited in the lungs and can easily cause side effects in other parts of the body.
  • the absorption area of the alveoli is large, and the permeability of the alveolar epithelial cells is high; direct pulmonary administration by means of aerosol preparations can increase the local drug concentration in the lung tissue, reduce drug loss, accurately quantify the drug dosage, and reduce toxic side effects.
  • large, hydrophilic, and negatively charged macromolecules, such as proteins and nucleic acids still have difficulty penetrating the lung barrier.
  • Nebulized formulations include, but are not limited to, aerosols, inhalation powder mists and sprays.
  • Inhalation preparations include liquids (such as inhalation aerosols and nebulized inhalation solutions) or solid preparations (such as inhalation powders) delivered to the lungs in the form of vapor or aerosol.
  • the drugs used include, but are not limited to, local therapeutic drugs, antibiotic drugs, antiviral drugs, antitumor drugs, protein peptide drugs, and the like.
  • the drug used in the aerosol preparation can be an anti-inflammatory drug, including but not limited to non-steroidal drugs and steroidal drugs.
  • Non-steroidal drugs include, but are not limited to, aspirin, acetaminophen, diclofenac, indomethacin (indomethacin), ibuprofen, fenbufen and other drugs and their derivatives in various dosage forms.
  • Steroid drugs include, but are not limited to, adrenocorticoid drugs, that is, various dosage forms of drugs such as glucocorticoids and their derivatives.
  • the drugs used in the aerosol preparation can be antibiotics.
  • Antibiotics include but are not limited to ⁇ -lactams, aminoglycosides, macrolides, polypeptides, lincosamides, tetracyclines, amido alcohols/chloramphenicols , Rifamycins and other antibiotics.
  • Beta-lactams include, but are not limited to, penicillin drugs (penicillin G, procaine penicillin, penicillin V, benzathine penicillin, oxacillin, cloxacillin, flucloxacillin, amoxicillin, ampicillin, piperazine Cillin, mezlocillin, azlocillin, amoxicillin and clavulanate potassium, piperacillin sodium, tazobactam, ampicillin sodium and sulbactam), cephalosporins (cephalothin, cefazolin, cefathiamidine) , Cephalexin, cefadroxil, cefradine, cefuroxime, cefmandol, cefotiam, cefonisi, cefuroxime axetil, cefaclor, cefprozil, cefotaxime, ceftazidime, cefoperazone, Cefazoxime, ceftriaxone, cefixime, cefd
  • Atypical ⁇ -lactams (cephalosporins such as cefoxitin, cefmetazole, cefotetan, cefmino, cefrazon, monocyclic ⁇ -lactams such as aztreonam, carlumonam, Carbapenems such as imipenem, meropenem, panipenem, faropenem, ertapenem, biapenem, donipenem, and oxycephalosporins such as laoxycephalosporin and fluoxefone)
  • ⁇ -lactams such as cefoxitin, cefmetazole, cefotetan, cefmino, cefrazon
  • monocyclic ⁇ -lactams such as aztreonam, carlumonam
  • Carbapenems such as imipenem, meropenem, panipenem, faropenem, ertapenem, biapenem, donipenem
  • Aminoglycosides include but are not limited to etimicin, streptomycin, gentamicin, kanamycin, amikacin, tobramycin, netilmicin, spectinomycin, isepamicin, Various dosage forms of neomycin, paromomycin, kasugamycin, paromycin, sisomicin, ribosomycin and its derivatives.
  • Macrolides include, but are not limited to, erythromycin, azithromycin, clarithromycin, roxithromycin, telithromycin, dirithromycin, medicamycin, acetylmedicamycin, kitasamycin, and acetylkiatamycin , Acetylspiramycin, spiramycin, oleandomycin, tylosmycin, josamycin, rosomycin, erythromycin ethylsuccinate, tilmicosin, kitorimycin and its derivatives Dosage form.
  • Polypeptides include, but are not limited to, various dosage forms of vancomycin, norvancomycin, teicoplanin, bleomycin, polymyxin B, polymyxin E, bacitracin and its derivatives.
  • Lincosamides include, but are not limited to, various dosage forms of lincomycin, clindamycin and their derivatives.
  • Tetracyclines include, but are not limited to, various dosage forms of doxycycline, minocycline, tetracycline, chlortetracycline, oxytetracycline, demecycline, doxycycline, tigecycline and its derivatives.
  • Amido alcohols/chloramphenicols include, but are not limited to, various dosage forms of chloramphenicol, thiamphenicol, palm chloramphenicol, chloramphenicol succinate, florfenicol and their derivatives.
  • Rifamycins include, but are not limited to, various dosage forms of rifampicin, rifapentin, rifabutin, rifamycin sodium and derivatives thereof.
  • Various dosage forms of other antibiotics such as fosfomycin, nystatin and its derivatives.
  • the drugs used in the aerosol preparation can be antiviral drugs, including but not limited to sulfonamides and antibacterial synergists, quinolone antibacterials, antituberculosis drugs, antifungal drugs, antiviral drugs, anti-AIDS drugs, and antiparasitic drugs.
  • Sulfa drugs include but are not limited to sulfamethoxazole, sulfasalazine, sulfadiazine, sulfadimethoxine, sulfadimethoxine, sulfadiazine sodium, sulfisoxazole, sulfamethazine , Sulfamethazine, sulfamethazine sodium, sulfamethoxazole, sulfamethoxine, sulfamethoxazole, sulfamethoxazine sodium, sulfamethoxazole, sulfamethoxazole, Various dosage forms of sulfachloropyridazine, sulfachlordazine sodium, sulfafenpyrazole, sulfachlorpyrazine sodium, sulfaquinoxaline, sulfaquinox
  • Antibacterial synergists include, but are not limited to, trimethoprim (TMP), dimethoprim (DVD), dimethoprim (OMP) and various formulations of drugs and their derivatives.
  • Quinolone antibacterial drugs include but are not limited to nalidixic acid, pipemidic acid, norfloxacin, ofloxacin, levofloxacin, pefloxacin, enoxacin, ciprofloxacin, lomefloxacin, fleroxacin, Various dosage forms of sparfloxacin, enrofloxacin, gatifloxacin, moxifloxacin, pazufloxacin and other drugs and their derivatives.
  • Anti-tuberculosis drugs include, but are not limited to, butadiene hydrazide, prothiazide, ethionamide, ethionamide, Yunnan Baiyao, rifapentine, rifampicin-isoniazide (weifenin), Rifampicin-Isoniazid-Pyrazinamide (Weifete), Rifampicin isoniazid, Rifampicin (Meperifampicin, Weifuxian, Xendoxin), Rifamycin Sodium (Fenglifu ), rifamycin sodium, pyrazinamide, pyrazinamide, furosemide, prunella vulgaris, allicin, Weifuxian, p-aminosalicylic acid isoniazid (nuclear), sodium p-aminosalicylate, Pasoniazid, Isoniazid, Isoniazid, Isoflurane, Isofolamide (Dy
  • Antifungal drugs include, but are not limited to, the following two types of drugs and their derivatives in various dosage forms: the first category is antibiotics, mainly griseofulvin, nystatin and amphotericin B; the other category is synthetic drugs , Including imidazole drugs (such as clotrimazole, econazole, miconazole and ketoconazole, etc.), flucytosine and allylamine derivatives.
  • antibiotics mainly griseofulvin, nystatin and amphotericin B
  • synthetic drugs Including imidazole drugs (such as clotrimazole, econazole, miconazole and ketoconazole, etc.), flucytosine and allylamine derivatives.
  • Antiviral drugs include but are not limited to iodoside (IDU), trifluorothymidine (TFT), adenosine arabinoside (Ara-A), ribavirin (RBV), acyclovir (ACV) , Propoxyuridine (DHPG), azidothymidine (AZT), dideoxyinosine (DDI), amantadine, rimantadine, morpholinoguanidine (viral), phthalamide, foscarnet (PFA) , Isoprinosine and other drugs and various dosage forms of their derivatives.
  • Anti-AIDS drugs include but are not limited to nucleoside reverse transcriptase inhibitors (NRTIS), non-nucleoside reverse transcriptase inhibitors (NNRTIS), protease inhibitors (PIS), integrase inhibitors (raltegravir), fusion Various dosage forms of drugs such as inhibitors (FIS) and CCRS inhibitors (maraviroc) and their derivatives.
  • Antiparasitic drugs include, but are not limited to, antiprotozoal drugs, antihelminth drugs, insecticides and other drugs and various formulations of their derivatives.
  • the drug used in the aerosol preparation can be an anti-tumor drug, as described in Table 3-3.
  • fluorinated chitosan can be used as a drug carrier to deliver antibody drugs such as apoptosis-ligand 1 antibody for pulmonary administration in the form of inhalation/spray.
  • fluorinated chitosan as the main body, combined with drugs can also be made into corresponding nasal drops to promote the transnasal mucosal administration of drugs.
  • Nasal mucosa covers the surface of the nasal cavity, underneath the mucosa is cartilage, bone or skeletal muscle. According to the difference of structure and function, the nasal mucosa is divided into three parts: vestibular part, respiratory part and olfactory part.
  • the vestibular part is the part adjacent to the outer nostril, which is rich in nasal hairs to block the inhalation of larger dust particles in the air.
  • the respiratory part occupies most of the nasal mucosa and has developed epithelial cilia, which can swing to the pharynx, and the mucus with dust particles and bacteria is discharged to the pharynx, and finally they are excreted from the body.
  • the olfactory mucosa is small, mainly located at the top of the nasal cavity. It contains olfactory cells and olfactory glands that specialize in the sense of smell. They secrete odor-containing particles that can dissolve the olfactory area and stimulate the olfactory hairs on the surface of the olfactory cells to produce the sense of smell. Moreover, because the olfactory cells have different receptors, they can Respectively accept the stimulation of different chemical molecules, so it can produce different sense of smell.
  • the total surface area of the human nasal mucosa is about 150cm 2.
  • the existing preparations for nasal administration include drops, odorants, ointments, nasal congestion, inhalants, sprays, powder mists and the like.
  • the mucosa in the upper part of the nasal cavity is thicker than the mucosa in the bottom of the nasal cavity and the inner sinuses, and the blood vessels are dense, which is the main place for drug absorption.
  • the product of this patent can increase the retention time of the drug in the nasal mucosa, reduce the entry of the drug into the oral cavity, increase the utilization rate, and accelerate the drug to penetrate the mucosa and enter the blood to reach the lesion site for treatment.
  • bypassing the blood-brain barrier and administering nasal mucosa to treat brain diseases is an example of this patent.
  • the nasal mucosa is rich in olfactory cells.
  • the central processes of olfactory cells form olfactory nerve fibers, which extend to the olfactory filaments.
  • the olfactory filaments terminate in the olfactory bulb of the olfactory neuron and reach the brain along the olfactory nerve of the olfactory bulb.
  • the drug carrier of the patent can extend the residence time of the drug, increase the bioavailability, and be delivered to the brain by the olfactory cells, bypassing the blood-brain barrier for drug delivery.
  • the trigeminal nerve in the nasal cavity is also considered to be one of the ways for nasal cavity administration to target the central nervous system. Therefore, this patent uses this technology to penetrate the nasal mucosa to treat brain diseases, trigeminal neuralgia, etc., and to prepare nasal mucosal vaccines. Nasal mucosa local anesthesia and other areas.
  • the treatment drugs for cerebral apoplexy may include, but are not limited to, the specific drugs and their derivatives in Table 3-4 below.
  • Anti-tumor drugs may include, but are not limited to, the specific drugs and their derivatives in Table 3-1 below.
  • Nasal mucosal administration via the trigeminal nerve route can also be used for the treatment of herpetic trigeminal/glossopharyngeal nerve pain.
  • Examples include, but are not limited to, ketamine, dixylamine, carbamazepine and derivatives thereof.
  • Nasal mucosal administration can also treat other diseases, such as anti-inflammatory drugs, antibiotics, and antiviral drugs to treat rhinitis, throat, lung and other inflammations. See the aerosol formulation section for example drugs.
  • Nasal mucosal administration penetrates the mucosa to exert a local effect or enters the blood to exert a systemic effect, such as applied to anesthesia.
  • general anesthesia include but are not limited to ketamine hydrochloride, propofol, thiopental, etomidate, midazolam and Sodium ⁇ -hydroxybutyrate.
  • Local anesthetics include but are not limited to aromatic acid esters, aromatic amides, amino ketones, amino ethers, carbamates, hydroxyprocaine, chloroprocaine, tetracaine, butacaine, Thiocaine, procainamide, bupivacaine, articaine, eticaine, ropivacaine, mepivacaine, clonin and other drugs and their derivatives.
  • Analgesics are drugs that act on the central nervous system and selectively inhibit the pain center, reducing or eliminating pain without affecting other sensations. They are mainly used clinically for trauma, burns, post-operation and cancer. The sharp pain relieves the pain.
  • the analgesic may be morphine and its derivatives including but not limited to: codeine, ethylmorphine, benzylmorphine, iscodeine, heroin, phenethylmorphine, hydromorphone, oxymorphone, propylene Morphine, nalorphine, naloxone, naltrexone and other drugs and their derivatives.
  • the analgesics may be synthetic analgesics including but not limited to: pethidine, aniridine, benperiidine, piminodine, afarotidine, betarotidine, fentanyl, alfenac Tanyl, Sufentanil, Remifentanil, Pethidine Hydrochloride, Methadone, Dexmorphine, Dexpropoxyphene, Methadone Hydrochloride, N-Methylmorphinan, Levophanol, Butorphanol, Morphine Nang, pentazocine, dezocine, sumatriptan, tramadol, fenthiidine, nefopam.
  • the drug may be a diabetes treatment drug including but not limited to sulfonylureas such as tolbutamide, tolbutamide, chlorpropamide, chlorhexamide, gliclazide, glipidin, glimepiride, etc.
  • sulfonylureas such as tolbutamide, tolbutamide, chlorpropamide, chlorhexamide, gliclazide, glipidin, glimepiride, etc.
  • Non-sulfonylurea drugs such as repaglinide and nateglinide and their derivatives
  • thiazolidinedione drugs such as rosiglitazone and piroglitazone and their derivatives
  • phenformin Metformin and other biguanide drugs and their derivatives
  • acarbose voglibose, miglitol and other ⁇ -glucosidase inhibitor drugs and their derivatives
  • glucagon-like peptide, DPP- IV inhibitors sitagliptin, vildagliptin, saxagliptin dipeptidyl peptidase-IV drugs and their derivatives, and insulin and other drugs and their derivatives.
  • Nasal vaccines include but are not limited to: influenza vaccine, pertussis vaccine, tuberculosis spray vaccine, influenza spray vaccine, measles spray vaccine, Burdella pertussis spray vaccine, Chlamydia pneumoniae spray vaccine, Streptococcus pneumoniae spray vaccine, Bacillus anthracis spray vaccine, pneumonia Coccal surface protein-nasal gel vaccine, botulinum neurotoxin type A BoHc/A gel vaccine, tetanus toxoid gel vaccine.
  • fluorinated chitosan can be used as a drug carrier to deliver drugs from the nasal mucosa to treat brain tumor diseases.
  • Intravaginal administration is the route of administration of drugs used in the vagina. Pharmacologically, compared with other routes of administration, it has the advantage of mainly acting on the vagina or nearby structures (such as the vaginal part of the cervix) and reducing systemic adverse reactions.
  • the vagina is an efficient site for drug delivery, especially in terms of women's health.
  • Vagina is usually the ideal route for drug administration, because compared with the oral route, the vaginal dose is lower, the drug level is stable, and the administration frequency is lower. Through vaginal administration, absorption is not affected by gastrointestinal diseases, and there is no first pass effect.
  • Methods of vaginal administration include vaginal tablets, vaginal creams, vaginal suppositories and vaginal rings. Vaginal diseases are mostly caused by bacterial and viral infections.
  • the example drugs are the same as antibacterial, antiviral drugs and anti-inflammatory drugs.
  • ocular mucosal infections mainly divided into three categories: ocular mucosal infections, corneal/conjunctival-related infections, and fundus-related diseases.
  • the first category, ocular mucosal diseases is mainly due to decreased ocular immunity, vitamin deficiency or accidental injury, which leads to damage and inflammation of the mucous membrane, or pathogenic microorganisms such as bacteria, viruses, fungi, parasites, amoeba and chlamydia invade the human eye Local tissue inflammation caused by the mucosa, such as eyelid infection, orbital infection, etc., if not treated in time, it will further infect the cornea and conjunctiva of the eye and make the condition worse.
  • clinical treatment methods such as pure medicated eye drops, eye gels, and eye ointments are commonly used.
  • the present invention uses the fluorinated chitosan as a carrier to deliver drugs for treating ocular surface diseases such as trachoma, blepharitis, meibomian gland cyst, blepharitis, meibomianitis, etc., to penetrate the mucosa and deliver them to the focal site for treatment.
  • the treatment drugs are shown in Table 4-1.
  • the second category of corneal/conjunctival-related infections mainly includes various microbial infections such as keratitis and conjunctivitis, corneal/conjunctival neovascular disease, and ocular allergies.
  • This site infection has the shortcomings of high pathogenicity, difficult treatment, troublesome treatment, prone to attacks, and poor patient compliance.
  • Corneal/conjunctival-related infections refer to local tissue inflammation caused by pathogenic microorganisms such as bacteria, viruses, fungi, parasites, amoeba and chlamydia, invading the cornea and conjunctiva of the human eye, such as conjunctival inflammation, keratitis and endophthalmitis Wait.
  • Common pathogenic fungi in the eye such as Fusarium, Aspergillus, Penicillium, Candida albicans, etc.
  • common viruses in the eye include adenovirus, rubella virus, herpes simplex virus, varicella-zoster virus, intestinal tract Viruses, etc.
  • the common chlamydia in the eye is Chlamydia trachomatis, which often causes trachoma, conjunctivitis, and lymphogranuloma. Chlamydia psittaci often causes psittacosis and so on.
  • the present invention uses fluorinated chitosan as a drug carrier. Because of the positive adsorption capacity of fluorinated chitosan, it can adhere to the surface of the cornea/conjunctiva, open the cornea/conjunctival epithelial cell channel, and deliver drugs related to the treatment of eye infections to the cornea. / Inhibition or removal of microorganisms at the site of conjunctival infection. It can also deliver ocular anti-inflammatory drugs to play anti-inflammatory and analgesic effects.
  • the third type of fundus disease is the most difficult. To reach the lesion site, the drug needs to penetrate the multiple barriers of the eyeball. At present, the methods of intravitreal injection and subconjunctival or conjunctival sac administration are mainly used. Causes irreparable scars and affects vision.
  • fundus macular degeneration the fundus macular area is an important area of the retina, which is related to visual functions such as fine vision and color vision. Once the macular area has lesions, vision loss, dark shadows in front of the eyes, or distortion of vision often occur.
  • Fundus macular degeneration can be caused by genetic disease, age-related disease, inflammatory disease, etc., and can also be affected by other fundus diseases, and treatment is difficult; age-related macular degeneration mainly includes age-related macular degeneration, age-related idiopathic epiretinal membrane and senile Changes such as macular holes can be improved or stabilized through early diagnosis and appropriate treatment; inflammatory macular degeneration is more common in various retinal choroiditis, such as toxoplasmosis, uveitis, etc.; in addition, retinal vein occlusion, Retinal vasculitis, diabetic retinopathy, high myopia, and traumatic choroidal rupture can cause damage to the macular area, which can cause edema or hemorrhage in the macular area and cause a certain degree of visual impairment.
  • age-related macular degeneration mainly includes age-related macular degeneration, age-related idiopathic epiretinal membrane and senile Changes such as macular holes
  • Surgical treatment mainly includes laser treatment, transpupillary thermotherapy, photodynamic therapy, surgical resection of neovascularization, macular displacement, and retinal transplantation.
  • medical treatment is mainly Intracavitary administration of anti-VEGF series monoclonal antibodies will still cause conjunctival and retinal scars to affect vision, and also make the patient bear greater pain and greater risk.
  • Eye diseases and eye tumors bring great pain to patients. Eye tumors are divided into internal eye tumors and external eye tumors. The internal eye tumors are accompanied by yellow and white reflections in the pupils (commonly known as cat eyes), vision disappears, and intraocular pressure increases.
  • Tumors of the external eye manifest as local induration in the early stage, and can invade all eyelids, orbits and paranasal sinuses in the late stage, forming severe local tissue defects.
  • infants and young children it is the most serious and most harmful malignant tumor. It occurs in the nuclear layer of the retina and has a family genetic tendency. It mostly occurs under 5 years of age. The disease is prone to intracranial and distant metastasis. The lives of children are endangered, so early detection, early diagnosis and early treatment are the keys to increase the cure rate and reduce the mortality rate.
  • the current treatment methods are mainly surgical treatment including tumor resection, eyeball enucleation, orbital content enucleation; radiotherapy, using deep X-ray, Co and other deep irradiation, or P, Sr and other shallow irradiation, according to the conventional treatment of radiotherapy; Comprehensive therapy, the comprehensive application of traditional Chinese medicine, western medicine, radiotherapy and surgery; immunotherapy, the use of immunosuppressive therapy to control tumor proliferation.
  • Tumor therapeutic drugs and derivatives are shown in Table 4-2.
  • Table 4-2 the same problem as the above-mentioned macular degeneration exists, and it is difficult for drugs to pass through the barrier for treatment.
  • the present invention provides an application of fluorinated chitosan in promoting the efficiency of drug absorption; a fluorinated chitosan and its use as a variety of drug carriers.
  • the skin is composed of the epidermis and the dermis.
  • the epidermis is the stratum corneum, the transparent layer, the granular layer and the germinal layer from shallow to deep.
  • the dermis is composed of dense connective tissue, from shallow to deep into the papillary layer and the reticular layer.
  • the papillary layer is connected to the germinal layer of the epidermis, which is rich in capillaries, lymphatic vessels, nerve endings, and tactile corpuscles.
  • the stratum corneum is the largest restrictive barrier for transdermal drug delivery.
  • stratum corneum consists of 5-25 layers of flat keratinocytes. These cells have no nucleus, organelles, and thicker cell membranes. They are inanimate and impermeable to prevent tissue fluids. Outflow, anti-friction and anti-infection functions. Cationic polymers modified with fluorine compounds can stimulate the distribution of tight junction proteins in these cells, reduce tight junctions between cells, and further stimulate actin phosphorylation, thereby promoting paracellular transport, opening intercellular spaces, forming channels, and carrying The drug penetrates the stratum corneum, further penetrates into the skin, and then enters the dermis, enters the skin capillaries and lymphatic circulation, and exerts the effect of the drug (see Figure 4-1).
  • the fluorine-containing compound modified cationic polymer provided by the present invention can be universally combined with a variety of drugs, promote drug absorption, improve drug bioavailability, and reduce toxicity. It has the advantages of good effect and wide application. , Has great commercial value, and the fluorine-containing compound modified cationic polymer proposed in the present invention is easy to produce and has a commercial basis.
  • fluorinated chitosan and medicine are prepared into eye drops for administration, which can penetrate the ocular barrier and partially deliver the medicine to the eye to play a therapeutic effect.
  • Exemplary drugs may include, but are not limited to, specific drugs and their derivatives for eye-related diseases in Table 4-1 below.
  • Anti-tumor drugs may include, but are not limited to, specific drugs and their derivatives in Table 4-2 below.
  • the present invention uses cationic polymers modified with fluorine-containing compounds, especially fluorinated chitosan, as a drug carrier to penetrate the most important corneal barrier of the eyeball to realize the application of drug local treatment but capable of treating intraocular or fundus diseases.
  • the schematic diagram of permeation is shown in Figure 4-2, and the black arrow represents the direction of drug permeation.
  • fluorinated chitosan When fluorinated chitosan is combined with drugs to prepare eye drops and dropped on the ocular surface, fluorinated chitosan adheres to the surface of the cornea and conjunctiva due to positive charges, increasing the retention time of the drug, and opening the intercellular channel protein to help the drug Penetrate the cornea and conjunctival barrier, penetrate the blood-retinal barrier, and treat related diseases.
  • Fluorinated chitosan can be used as a drug carrier to deliver therapeutic drugs into the eye and administer the cornea in the form of eye drops for the treatment of eye diseases, such as corneal/conjunctival related inflammation, age-related macular degeneration, retinal melanoma, etc. .
  • the present invention uses chitosan modified with fluorine-containing compounds as a drug carrier and a drug delivery body, which can increase the stagnation time of eye drops on the ocular surface, open the eye-related barriers and deliver drugs to the lesion site for enrichment.
  • the purpose of treatment is not only for ocular mucosal infections and corneal/conjunctival-related infections, but also for ocular fundus-related diseases.
  • drugs used to treat ocular neovascularization include bevacizumab, ranibizumab, sorafenib, and sunitinib.
  • the antibiotic eye drops of the present invention adopt the above-mentioned barrier-penetrating drug delivery formulation, and the compound formed by the medicine for treating corneal bacterial infection and the fluorinated chitosan, and the particle size range of the compound is less than 10 Micrometer, or the particle size range of the composite is not more than 500 nanometers, and the mass ratio of the fluorinated chitosan to the drug is 1:0.5-30.
  • drugs for the treatment of bacterial infections of the cornea and bacterial infections inside the eyeball include penicillin, erythromycin, cephalosporin, cephalosporin, streptomycin, gentamicin, kanamycin, azithromycin, clarithromycin, Roxithromycin, Telithromycin, Kitasamycin, Vancomycin, Norvancomycin, Teicoplanin, Bleomycin, Polymyxin B, Polymyxin E, Bacitracin, Linko Clindamycin, clindamycin, doxycycline, minocycline, tetracycline, chlortetracycline, oxytetracycline, demecycline, doxycycline, tigecycline.
  • the anti-viral eye drops of the present invention adopts the above-mentioned barrier-penetrating drug delivery formulation, a complex formed by a drug for treating corneal virus infection and the fluorinated chitosan, and the particle size range of the complex is less than 10 micrometers, or the particle size range of the composite is not greater than 500 nm, and the mass ratio of the fluorinated chitosan to the drug is 1:0.5-50.
  • drugs for the treatment of keratovirus infections and intraocular viral infections include iodoside, trifluorothymidine, adenosine arabinoside, ribavirin, acyclovir, propoxyuridine, azidothymidine, Dideoxyinosine, amantadine, rimantadine, morpholinoguanidine, phthalamide, foscarnet, and isopropinosine.
  • the anti-inflammatory drug eye drops of the present invention adopting the above-mentioned barrier-penetrating drug delivery formulation, the anti-inflammatory drug for treating bacterial virus and injury-induced eye inflammation and the complex formed by the fluorinated chitosan, the
  • the particle size range of the composite is less than 10 microns, preferably not greater than 500 nm, and the mass ratio of the fluorinated chitosan to the drug is 1:0.5-50.
  • anti-inflammatory drugs for the treatment of eye inflammation caused by bacterial viruses and injuries including aspirin, acetaminophen, non-specific cyclooxygenase inhibitors, antipyrine, analgin, phenylbutazone, and oxybutazone , Mefenamic acid, indomethacin, sulindac, diclofenac sodium, ibuprofen, propnasan, piroxicam, meloxicam, hydrocortisone, corticosterone, aldosterone, triamcinolone, prednisone Dragon, dexamethasone acetate, methylprednisolone vinegar.
  • a preparation for penetrating the eye barrier of the present invention comprising component (a), said component (a) is a cationic polymer modified with a fluorine-containing compound, and the cationic polymer modified with a fluorine-containing compound
  • the substance can be used as a preparation that penetrates the tear barrier, the cornea/conjunctival barrier, the blood-aqueous barrier, and the blood-retinal barrier.
  • a preparation for penetrating the eye barrier of the present invention comprising component (a), said component (a) is a cationic polymer modified with a fluorine-containing compound, and the cationic polymer modified with a fluorine-containing compound.
  • a preparation for penetrating the eye barrier of the present invention comprising component (a), said component (a) is a cationic polymer modified with a fluorine-containing compound, and the cationic polymer modified with a fluorine-containing compound
  • component (a) is a cationic polymer modified with a fluorine-containing compound
  • the cationic polymer modified with a fluorine-containing compound Application of the drug in the preparation of a medicine for the treatment of ocular mucosal infections, a medicine for the treatment of cornea/conjunctival related infections, and a medicine for the treatment of ocular fundus diseases.
  • the technical solution of the patent can be used in the skin medical beauty system, as follows.
  • the purpose of the present invention is to provide a transdermal carrier as a drug delivery formulation for medical beauty and health care products.
  • This patent uses chitosan modified with a fluorine-containing compound as the main body, which can be prepared
  • Corresponding transdermal preparations are used in drug delivery systems that require medical beauty and health care products through the epidermal layer or mucosal layer.
  • the method can improve the delivery effect of the drug, improve people's use experience, and has great application prospects.
  • the medicine may be a medical cosmetic medicine.
  • the medical beauty drugs include, but are not limited to, hair health and beauty drugs, skin beauty and health drugs.
  • the hair health and beauty medicine described in this patent can be hair growth medicine and hair care medicine.
  • chitosan modified with fluorine-containing compounds as the main body, it can be made into corresponding transdermal preparations for the treatment of various causes of hair loss, such as sprays, gels, ointments, lotions, etc. Therefore, using cationic polymers modified by such fluorine-containing compounds as a drug carrier to penetrate the epidermis, local treatment of drugs can reach the subcutaneous and hair follicles to treat hair loss-related diseases.
  • fluorine-containing compound-modified chitosan can be used as a drug carrier to deliver therapeutic drugs to the subcutaneous and hair follicles, and locally administered to the epidermis in the form of sprays for the treatment of alopecia diseases.
  • the skin beauty and health care drugs can also be moisturizing, skin rejuvenation, anti-wrinkle, freckle, scar removal drugs, or other traditional Chinese medicine beauty drugs. Drugs can accelerate blood circulation, promote the body's metabolism and tissue repair.
  • the drug may be dermatitis, eczema and other dermatological topical drugs, including compound dexamethasone acetate cream (Piyanping), compound miconazole nitrate cream, ketoconazole cream, triamcinolone acetonide and econazole cream, Paeonol ointment, tretinoin ointment, geranium ointment, compound Shuanghuateng blunt liniment, terbinafine hydrochloride cream, calcipotriol ointment, penciclovir cream, acyclovir gel, Mometasone furoate cream, triazin cream, ofloxacin gel, hydrocortisone butyrate cream, miconazole nitrate cream, triamcinolone acetonide and econazole ointment, metronidazole gel , Bifonazole cream, etc.
  • compound dexamethasone acetate cream Pieriyan
  • the drug can be hormone drugs or tretinoin drugs (also called retinoic acid drugs).
  • the hormone drugs may be glucocorticoids, such as Pikangwang (clobetasol), piyanping (dexamethasone), freckle cream (containing clobetasol), and so on.
  • the therapeutic effects of glucocorticoids mainly come from their anti-inflammatory effects, including inhibiting the release of lysosomal enzymes, inhibiting the stimulation of macrophages and capillary contraction, etc., but they are prone to side effects such as dermatitis and folliculitis.
  • the retinoic acid drugs include non-aromatic formic acid drugs (such as tretinoin and isotretinoin), monoaromatic formic acid drugs (such as isotretinoin and isotretinic acid), and polyaromatic retinoic acid drugs (such as aromatic ethyl methyl and adapalene).
  • Tretinoin drugs have the effects of anti-aging, reducing epidermal melanin, inhibiting sebum production, and immune regulation. They can protect skin, reduce wrinkles, fade chloasma, and treat skin problems such as acne, pigmentation, and abnormal keratosis.
  • oral retinoic acid drugs have large side effects, and the use of the epidermis can easily cause local irritation and cause burning sensation and slight pain. The clinical use has received certain restrictions.
  • the fluorine-containing compound modified polymer can improve the penetration efficiency, thereby reducing the dosage of the drug in the affected area, thereby achieving the effect of reducing side effects.
  • the present invention uses a cationic polymer modified with a fluorine-containing compound as a drug carrier to penetrate the epidermis to deliver polyinosinic acid below the stratum corneum, induce the production of endogenous retinoic acid, and achieve local drug treatment , In order to achieve the effect of improving scars and low side effects.
  • the drug can be an external antioxidant, including vitamin E, green tea polyphenols, acetylcysteine, coenzyme Q10, superoxide dismutase, etc., which can effectively respond to oxidative stress in cells and achieve the purpose of anti-aging .
  • the drug can be drugs such as omega-3 fatty acids, a-hydroxy acids, ⁇ -carotene, etc., which can increase skin elasticity and collagen synthesis.
  • the skin beauty and health care drugs can also be vitamins and trace elements, which can improve the nutritional status of the skin, beautify the skin, and delay aging. Deficiency of nutrients such as iron, iodine, zinc, vitamin A, vitamin B2, niacin, biotin, folic acid, vitamin B12, vitamin C and vitamin D may impair growth and cause disease, which is a health issue of widespread public concern. Although nutrient deficiencies can be effectively treated by fortifying the nutrition of food, there are still problems that the nutrients are not stable enough and the absorption efficiency is not high enough. Compared with free nutrients, the nutrients encapsulated in nanoparticles are more stable, but their bioavailability is lower. A nutrient preparation using perfluoroheptanoic acid modified chitosan as a carrier can effectively improve bioavailability and promote nutrient absorption.
  • the skin beauty and health care drug can also be a traditional Chinese medicine beauty drug, which can improve skin problems such as dull skin tone, pigmentation, sagging skin, enlarged pores, rough skin, etc., or repair skin barriers, and restore skin health.
  • the traditional Chinese medicine cosmetology drugs can be drugs for delaying cell aging (extracts of astragalus and ginseng); drugs for scavenging free radicals (schisandra, notoginseng, extracts, flavonoid drugs, salvianolic acid B and salvianolic acid C active substances); repair Collagen drugs (Poria cocos extract, Polygonatum and wolfberry, salvianolic acid B and salvianolic acid C active substances); immunomodulatory drugs (Schisandra extract, saponins, dodder extract); skin microcirculation drugs (Danshen extract, class Flavonoids) and other Chinese medicines and Chinese herbal medicines.
  • Antioxidant drugs or glucocorticoid drugs are often used in drug therapy, such as tranexamic acid, proanthocyanidins, glutathione, hydroquinone (also known as hydroquinone), tretinoin, cysteamine salts, oligopeptides, Jessner solution, salicylic acid, lactic acid.
  • tranexamic acid can inhibit ultraviolet-induced plasmin activity in keratinocytes, prevent plasminogen from binding to keratinocytes, reduce prostaglandin synthesis, and thereby inhibit pigment production.
  • Oral or local injection of tranexamic acid can treat melasma.
  • individuals and families need to be screened for the risk of thromboembolism before oral administration. The penetration effect of topical administration is not satisfactory. Therefore, in the present invention, the cationic polymer modified by the fluorine-containing compound is used as a drug carrier to penetrate the epidermis to achieve the effect of local drug treatment to subcutaneous treatment of melasma.
  • fluorine-containing compound-modified chitosan can be used as a drug carrier to deliver therapeutic drugs under the skin for topical administration to the epidermis for the treatment of melasma.
  • a drug complex of the present invention adopting the above-mentioned medical beauty and health care product carrier, including the medical beauty and health care product carrier (a) and a drug component (b), the drug component (b) is a hair growth drug and hair Nursing drugs; or moisturizing, skin rejuvenation, anti-wrinkle, freckle, scar removal drugs; or topical drugs for dermatitis and eczema skin diseases; or hormone drugs or tretinoin drugs; or vitamin E, green tea polyphenols, acetylcysteamine Acid, coenzyme Q10, superoxide dismutase.
  • the drug component (b) is a hair growth drug and hair Nursing drugs; or moisturizing, skin rejuvenation, anti-wrinkle, freckle, scar removal drugs; or topical drugs for dermatitis and eczema skin diseases; or hormone drugs or tretinoin drugs; or vitamin E, green tea polyphenols, acetylcysteamine Acid, coenzyme Q10, superoxide
  • a medical beauty and health care product carrier formulation of the present invention comprising component (a), said component (a) is a cationic polymer modified with a fluorine-containing compound, and the cationic polymer modified with a fluorine-containing compound It can be used as a preparation for administering hair growth medicine, hair care medicine, beauty medicine and health medicine.
  • An external preparation for hair growth prepared by the above-mentioned medical beauty and health care product carrier preparation of the present invention, including the effective inhibitor of mitochondrial pyruvate transporter UK5099, metformin, minoxidil, spironolactone, finasteride and fluoride
  • a complex formed by fluorinated chitosan, the particle size of the complex is less than 10 microns, or the particle size of the complex is not greater than 500 nanometers, and the mass ratio of the fluorinated chitosan to the hair growth drug is 1: 0.5-50.
  • a complex for removing scars prepared by using the above-mentioned medical beauty and health care product carrier formulations of the present invention includes complex particles formed mainly by perfluoroheptanoic acid-modified chitosan and polyinosinic acid.
  • the particle size range is less than 10 microns, or the composite particles are composites not greater than 500 nanometers, and the mass ratio of the fluorinated chitosan to the anti-freckle drug is 1:0.5-50.
  • the present invention is a medicine compound for treating melasma prepared by using the above-mentioned medical beauty and health care product carrier preparation.
  • the compound is perfluoroheptanoic acid-modified chitosan encapsulating tranexamic acid, and the fluorine
  • the mass ratio of chitosan to melasma treatment drug is 1:0.5-50.
  • the present invention provides the following technical solutions:
  • the skin is composed of the epidermis and the dermis.
  • the epidermis is the stratum corneum, the transparent layer, the granular layer and the germinal layer from shallow to deep.
  • the dermis is composed of dense connective tissue, from shallow to deep into the papillary layer and the reticular layer.
  • the papillary layer is connected to the germinal layer of the epidermis, which is rich in capillaries, lymphatic vessels, nerve endings, and tactile corpuscles.
  • the stratum corneum is the largest restrictive barrier for transdermal drug delivery.
  • stratum corneum consists of 5-25 layers of flat keratinocytes. These cells have no nucleus, organelles, and thicker cell membranes. They are inanimate and impermeable to prevent tissue fluids. Outflow, anti-friction and anti-infection functions. Cationic polymers modified with fluorine compounds can stimulate the distribution of tight junction proteins in these cells, reduce tight junctions between cells, and further stimulate actin phosphorylation, thereby promoting paracellular transport, opening intercellular spaces, forming channels, and carrying The drug penetrates the stratum corneum, further penetrates into the skin, then enters the dermis, enters the skin capillaries and lymphatic circulation, and exerts the drug effect
  • the fluorine-containing compound modified cationic polymer provided by the present invention can be universally combined with a variety of drugs, promote drug absorption, improve drug bioavailability, and reduce toxicity. It has the advantages of good effect and wide application. , Has great commercial value, and the fluorine-containing compound modified cationic polymer proposed in the present invention is easy to produce and has a commercial basis.
  • the invention also provides a cationic polymer modified with a fluorine compound, especially fluorinated chitosan, which promotes drug absorption across mucous membranes and has good biological safety.
  • a fluorine compound especially fluorinated chitosan
  • the cationic polymer modified by the fluorine-containing compound proposed in the present invention has a simple synthesis method and universal applicability; and has obvious advantages in transmucosal administration, mainly including: (1) It has good permeability and can span various mucosal membranes.
  • Nasal cavity mucosa, lung mucosa, vaginal mucosa, oral mucosa, gastrointestinal mucosa, etc. increase the concentration of drugs in blood and tissues; (2) It has good adhesion and can adhere to various mucosal surfaces to realize drugs (3) It has a wide range of applications. It can be combined with macromolecular drugs, can also adsorb small molecule drugs, and can also be combined with compound drugs with complex components for the treatment of various diseases and has great commercial value.
  • the fluorinated chitosan drug carrier provided by this patent has the advantages of obvious drug absorption promotion effect, low toxicity, etc., and the fluorinated chitosan proposed in the present invention has a mature synthesis process, simple operation, high synthesis efficiency, and short cycle. No tedious purification steps are needed to obtain high-yield drug carriers, and its simple synthesis method provides a good basis for commercialization.
  • the fluorinated chitosan of the present invention can be used as a variety of drug carriers. It can effectively improve the treatment effect, has a wide range of uses, and has a low cost.
  • the transmucosal effect produced by the technical solution of this patent is temporary. After the drug is removed, the intestinal mucosal cells will close the channel to protect the safety of the human body. (See Figure 3-1).
  • the fluorine-containing compound modified cationic polymer especially fluorinated chitosan, has the advantages of being able to be universally combined with a variety of drugs, promoting drug absorption, improving drug bioavailability, reducing toxicity, and having good effects.
  • the application is very wide and has great commercial value, and the cationic polymer modified by the fluorine-containing compound proposed in the present invention is easy to produce and has a commercial basis.
  • the application of the fluorinated modified cationic polymer designed in this patent in the fields of biochemistry and pharmacy includes but is not limited to the following modes of administration: oral drug delivery system, spray/inhalation, and nasal drops.
  • the cationic polymer modified by the fluorine-containing compound of the present invention is chitosan with a degree of deacetylation >95%.
  • the following application takes fluorinated modified chitosan as an example.
  • Other cationic polymers can also be fluorinated to achieve similar effects.
  • Figure 1-1 shows the effect of heptafluorobutyric acid-modified chitosan (7FCS) in Example 1-5 on the distribution and strength of THP in mouse bladder tissue.
  • THP epirubicin
  • CS chitosan
  • FCS fluorinated chitosan
  • the right picture is the relative fluorescence intensity analysis of THP corresponding to the left picture.
  • Figure 1-2 shows the effect of tridecafluoroheptanoic acid modified chitosan (13FCS) on the distribution and intensity of THP in mouse bladder tissue in Example 1-6, in which epirubicin is THP; CS, chitosan ; FCS, fluorinated chitosan; the right picture is the relative fluorescence intensity analysis of THP corresponding to the left picture.
  • 13FCS tridecafluoroheptanoic acid modified chitosan
  • Figure 1-3 shows the effect of different fluorine-containing fatty acid modified chitosan (7FCS, 13FCS, 19FCS) on the distribution and intensity of THP in mouse bladder tissues in Examples 1-7.
  • fluorine-containing fatty acid modified chitosan 7FCS, 13FCS, 19FCS
  • epirubicin, THP CS
  • Chitosan CS
  • FCS fluorinated chitosan
  • the right picture shows the relative fluorescence intensity analysis of THP corresponding to the left picture.
  • Figure 1-4a is a comparison diagram of the excellent in vitro cell safety of 13F-3 in Examples 1-8.
  • Figure 1-4b shows that there is no significant difference between the body weight of mice in the FCS group and the blank control group in Examples 1-8.
  • Figures 1-4c are comparison pictures of the bladder tissues and HE (hematoxylin-eosin) stained sections of the mouse bladder after perfusion in each group in Examples 1-8 and the mouse bladder of the blank control group.
  • Figure 1-5 is a comparison picture of the immunofluorescence results in Examples 1-8. It shows that the bladder of mice in the chitosan perfusion group has severe inflammatory stress and congestion and edema, but there is no significant difference between the FCS group and the blank control group.
  • the left picture Confocal pictures of bladder tissue slices. The right picture shows the CS, FCS treatment group and the blank group control.
  • Figures 1-6 are related pictures of MPI/FPEI measured by transmission electron microscopy in Examples 1-9.
  • Figures 1-7 are related pictures of MPI/PEI measured by transmission electron microscopy in Example 1-9.
  • Figure 1-8 shows that the mucosal permeability index of the polypeptides in the F-PEI group in Examples 1-9 is significantly higher than that of the PEI group and the blank control group.
  • the abscissa is the feed mass ratio of the polypeptide drug MPI to the material PEI or FPEI, and the ordinate is Is the permeability coefficient papp.
  • Figure 1-9 shows that the mucosal permeability index of the F-PEI histone drug in Example 1-9 is significantly higher than that of the PEI group and the blank control group.
  • the abscissa is the ratio of the feed quality of the polypeptide drug CAT-Ce6 to the material PEI or FPEI.
  • the ordinate is the permeability coefficient papp.
  • Figure 1-10 shows the different drug systems of MPI in Example 1-9.
  • the bladder was taken to prepare frozen sections and the drug fluorescence distribution and intensity control chart after infusion of drugs for different times.
  • the abscissa is the different perfusion time, and the ordinate is the peptide drug fluorescence. Relative value of intensity.
  • Figure 1-11 shows the different CAT drug systems in Example 1-9 after the bladder was perfused to prepare frozen sections and the drug fluorescence distribution and intensity control chart.
  • the left picture shows the fluorescence of the drug CAT-Ce6 different drug systems in the bladder tissue Distribution
  • the right picture shows the fluorescence intensity analysis of the drug.
  • Figures 1-12 are transmission electron microscope (TEM) imaging pictures in Example 1-10.
  • Figure 1-13 is the comparison chart of the frozen section of the bladder in Example 1-10 and the fluorescence intensity analysis of the fluorescent confocal microscope.
  • the left figure shows the fluorescence of the drug CAT-TCPP in the transverse and longitudinal sections of the bladder tissue from left to right. Distribution; from top to bottom, the right picture shows the fluorescence intensity of the drug in the bladder transverse and longitudinal section and bladder tissue homogenate.
  • Figure 1-14 is a synthetic route diagram of fluorine-containing carboxylic acid modified chitosan bladder infusion drug carrier.
  • Figure 2-1 Schematic diagram of the transdermal mechanism of cationic polymers modified by fluorine-containing compounds.
  • FIG. 2-2 The particle size changes of chitosan modified with perfluoroheptanoic acid in different proportions in aqueous solution before and after the reaction with the drug, taking perfluoroheptanoic acid modified chitosan-insulin (FCS-Insulin) as an example.
  • FCS-Insulin perfluoroheptanoic acid modified chitosan-insulin
  • FIG. 2-3 Potential changes of chitosan modified with perfluoroheptanoic acid in different proportions before and after the reaction with drugs in aqueous solution, taking perfluoroheptanoic acid modified chitosan-insulin (FCS-Insulin) as an example.
  • FCS-Insulin perfluoroheptanoic acid modified chitosan-insulin
  • FIG. 2-4 Different ratios of perfluoroheptanoic acid-modified chitosan and drug transdermal effect difference, the right picture is a schematic diagram of the transdermal diffusion pool, chitosan-insulin modified with perfluoroheptanoic acid (FCS-Insulin) Take for example.
  • FIG. 2-5 Photographs of the gel and scanning electron microscopy, taking perfluoroheptanoic acid modified chitosan-insulin (FCS-Insulin) as an example.
  • Figure 2-7 The therapeutic effect of the drug at the in vivo level, that is, the fluctuation of blood glucose after the application of the patch.
  • FCS-Insulin perfluoroheptanoic acid modified chitosan-insulin
  • FIG. 2-8 Different ratios of perfluoroheptanoic acid modified chitosan-immunoglobulin G (FCS-IgG) particle size and potential size in aqueous solution.
  • FCS-IgG perfluoroheptanoic acid modified chitosan-immunoglobulin G
  • FIG 2-9 The cumulative penetration of perfluoroheptanoic acid-modified chitosan-immunoglobulin G (FCS-IgG) to the skin at different time points, in which perfluoroheptanoic acid-modified chitosan-immunoglobulin
  • the ratios of protein G are 1:0.25, 1:0.5, 1:1, 1:2, 1:4 and pure immunoglobulin G (0:1).
  • FCS-IgG Perfluoroheptanoic acid-modified chitosan-immunoglobulin G
  • FCS-IgG Perfluoroheptanoic acid-modified chitosan-immunoglobulin G
  • Figure 2-11 In vivo tumor tissue penetration of perfluoroheptanoic acid-modified chitosan-apoptosis-ligand 1 antibody, chitosan-apoptosis-ligand 1 antibody and pure apoptosis-ligand 1 antibody Comparison of skin efficiency, from top to bottom are the pure apoptosis-ligand 1 antibody group (free aPDL1), chitosan-apoptosis-ligand 1 antibody (CS-aPDL1), and perfluoroheptanoic acid modified Chitosan-apoptosis-ligand 1 antibody (FCS-aPDL1) group, from left to right, DAPI channel (grey, indicating tumor tissue), FITC fluorescence channel (white, indicating apoptosis-ligand 1 antibody) ) And the fluorescence intensity of the mixed channel.
  • DAPI channel grey, indicating tumor tissue
  • FITC fluorescence channel white, indicating apoptosis-ligand 1 antibody
  • the left picture is a schematic diagram of the measurement method, and the right picture is the resistance change after adding perfluoroheptanoic acid modified chitosan-immunoglobulin G (FCS-IgG).
  • Perfluoroheptanoic acid modified chitosan-immunoglobulin G (FCS-IgG) transdermal mechanism: perfluoroheptanoic acid modified chitosan-immunoglobulin G (FCS-IgG) is dense Western Blotting analysis of the influence of related tight junction proteins in cell monolayers.
  • FIG. 2-15 Perfluoroheptanoic acid modified chitosan-apoptosis-ligand 1 antibody in vivo subcutaneous tumor treatment, divided into blank group (blank), pure apoptosis-ligand 1 antibody (free aPDL1), Chitosan-apoptosis-ligand 1 antibody (CS-aPDL1), and perfluoroheptanoic acid-modified chitosan-apoptosis-ligand 1 antibody (FCS-aPDL1) group, left picture mouse tumor growth The curve, the right figure is the survival rate curve of mice (definition that mouse tumors larger than 1500 cubic millimeters are dead).
  • Figure 3-1 is a schematic diagram of the intestinal mucosa.
  • Figure 3-2 is a schematic diagram of perfluoroheptanoic acid modified chitosan opening the intestinal mucosal epithelial cell barrier.
  • Figure 3-3 shows the particle size changes of chitosan modified with perfluoroheptanoic acid in different proportions in the aqueous solution before and after the reaction with the drug in Example 3-1, and the chitosan-insulin modified with perfluoroheptanoic acid (FCS-Insulin) ) As an example.
  • Figure 3-4 shows the potential changes of chitosan modified with perfluoroheptanoic acid in different proportions in the aqueous solution before and after the reaction with the drug in Example 3-1, and the chitosan-insulin modified with perfluoroheptanoic acid (FCS-Insulin) Take for example.
  • Figure 3-5 shows the change in particle size of the perfluoroheptanoic acid-modified chitosan drug complex before and after freeze-drying in Example 3-1, taking perfluoroheptanoic acid-modified chitosan-insulin (FCS-Insulin) as an example .
  • FCS-Insulin perfluoroheptanoic acid-modified chitosan-insulin
  • Figures 3-6 show the difference in the mucosal penetration effect between perfluoroheptanoic acid-modified chitosan and the drug in different proportions in Example 3-1, taking perfluoroheptanoic acid-modified chitosan-insulin (FCS-Insulin) as an example.
  • FCS-Insulin perfluoroheptanoic acid-modified chitosan-insulin
  • Figures 3-7 show the effect of transmucosal drug delivery of the drug-loaded capsules in gastric juice and intestinal juice in Example 3-1, taking perfluoroheptanoic acid modified chitosan-insulin (FCS-Insulin) as an example.
  • FCS-Insulin perfluoroheptanoic acid modified chitosan-insulin
  • Figure 3-8 shows the blood glucose fluctuations of mice after oral delivery of the drug-loaded capsule in Example 3-1, taking perfluoroheptanoic acid modified chitosan-insulin (FCS-Insulin) as an example.
  • FCS-Insulin perfluoroheptanoic acid modified chitosan-insulin
  • Figure 3-9 shows the cumulative permeation of perfluoroheptanoic acid-modified chitosan-immunoglobulin G (FCS-IgG) in different proportions to the intestinal mucosa at different time points in Example 3-2.
  • the abscissa is the injection time, and the ordinate is the transmittance of the cumulative penetration amount divided by the total injection amount.
  • chitosan-immunoglobulin G modified with different molecular weight perfluoroheptanoic acid can increase the efficiency of immunoglobulin G permeating rat intestinal mucosa.
  • a perfluoroheptanoic acid-modified chitosan-immunoglobulin G complex with a ratio of 1:1 is obtained.
  • Figure 3-10 shows the position of the perfluoroheptanoic acid modified chitosan-bovine serum albumin-cyanine dye Cy5.5 in the digestive tract at the 3rd hour and the 5th hour in Example 3-2, where white represents the cyanine dye Cy5 .5
  • the labeled bovine serum albumin indicates that the capsule can be released from the colorectal at a fixed point, so the capsule is used for subsequent experiments.
  • Figure 3-11 is a schematic diagram of the activity of the apoptosis ligand antibody 1 in Example 3-2 before and after lyophilization. After the lyophilization protectant is added and lyophilized, the apoptosis-ligand 1 antibody remains unchanged after it becomes a lyophilized powder. It shows that the perfluoroheptanoic acid modified chitosan-apoptosis-ligand 1 antibody particles can be freeze-dried and can be used for oral capsule filling.
  • Figure 3-12 shows the FCS/a PD-L1 and CT-26 cell surface PD-L1 binding activity before and after freeze-drying in Example 3-2.
  • Figure 3-13 shows the changes in the distribution of tight junction protein in human colorectal cancer epithelial cells before and after treatment with perfluoroheptanoic acid-modified chitosan and perfluoroheptanoic acid-modified chitosan/immunoglobulin G in Example 3-2.
  • Figure 3-14 shows the effect of perfluoroheptanoic acid-modified chitosan-apoptosis-ligand 1 antibody freeze-dried powder capsules in the treatment of Balb/c mouse colorectal cancer in Example 3-2, in which black indicates Bioluminescence intensity of mouse colorectal cancer cells.
  • Figure 3-15 shows the perfluoroheptanoic acid-modified chitosan/immunoglobulin G transmucosal effects of different molecular weights in Example 3-2.
  • Figure 3-16 is an immunofluorescence section of the lung of the mouse in Example 3-3.
  • the fluorescence signal indicates the retention of the drug in the lung.
  • Figures 3-17 are photographs of mouse brains in each experimental group in Example 3-4, in which the white arrows indicate the fluorescence signal of Cy5.5 in the mouse brain. .
  • Fig. 3-18 is a confocal laser confocal photograph of the mouse brain section in Example 3-4, in which the white arrow indicates the fluorescence signal of Cy5.5 at the mouse brain tumor site.
  • Figure 4-1 is a schematic diagram of the eyeball structure.
  • Figure 4-2 is a schematic diagram of the membrane permeation of fluorine-containing drug eye drops.
  • Figure 4-3 shows the cumulative permeation percentage of perfluoroheptanoic acid modified chitosan in Example 4-1 through cornea in vitro.
  • Figure 4-4 is the immunofluorescence staining image of the eyeball of the macromolecular drug bovine serum albumin in Example 4-1.
  • the left image is the perfluoroheptanoic acid-modified chitosan membrane immunofluorescence staining image, and the right image is the free drug permeation image.
  • Membrane immunofluorescence staining image is the immunofluorescence staining image.
  • Figure 4-5 shows the immunofluorescence staining image of the eyeball of the small and medium molecule drug Rhodamine B in Example 4-1.
  • the left image shows the perfluoroheptanoic acid-modified chitosan membrane immunofluorescence staining image, and the right image shows the free drug permeabilization immunofluorescence Fluorescence staining diagram.
  • Fig. 4-6 shows the content of ocular tissue of the perfluoroheptanoic acid-modified chitosan in Example 4-1 after eye drops.
  • Fig. 4-7 shows the immunofluorescence staining of the perfluoroheptanoic acid-modified chitosan partial cornea in Example 4-1 at different times after instillation.
  • Figures 4-8 are photos of perfluoroheptanoic acid-modified chitosan in Example 4-1 in the anterior segment of the corneal fluorescence staining of mice at various time points to detect the safety of perfluoroheptanoic acid-modified chitosan.
  • Fig. 4-9 is the bioluminescence image of in-situ choroidal malignant melanoma before treatment and one week after treatment in Example 4-2.
  • Fig. 4-10 is the bioluminescence quantitative analysis diagram of in-situ choroidal malignant melanoma in Example 4-2 after one week of treatment.
  • Figure 5-1 shows the percentage of mature dendritic cells in total dendritic cells after external stimulation of perfluoroheptanoic acid modified chitosan-chicken ovalbumin (FCS-OVA) complex in Example 5-1
  • FCS-OVA perfluoroheptanoic acid modified chitosan-chicken ovalbumin
  • FIG 5-2 shows that after external stimulation of the perfluoroheptanoic acid modified chitosan-chicken ovalbumin complex in Example 5-1, dendritic cells express and present the key protein of the antigen-the second type of histocompatibility Changes in the complex (MHC II) and differentiation cluster 40 protein (CD40).
  • MHC II antigen-the second type of histocompatibility Changes in the complex
  • CD40 differentiation cluster 40 protein
  • Figure 5-3 shows the fluorescent perfluoroheptanoic acid-modified chitosan-chicken ovalbumin complex and ointment in Example 5-1, mixed and applied on the back of mice, and perfluoroheptanoic acid-modified chitosan in the lymph nodes -In vivo imaging of the accumulation of chicken ovalbumin complex in lymph nodes over time.
  • the white arrow shows one of the lymph nodes in the mouse.
  • Figure 5-4 shows the use of transdermal application to plant the perfluoroheptanoic acid modified chitosan-chicken ovalbumin vaccine complex in Example 5-2 and the use of transdermal application to plant the chicken ovalbumin vaccine After vaccination, the growth of the tumor over time after a melanoma expressing chicken ovalbumin (B16-OVA) was planted on the back of the mouse.
  • B16-OVA melanoma expressing chicken ovalbumin
  • Figure 6-1 shows the in vitro transdermal delivery efficiency of different ratios of perfluoroheptanoic acid-modified chitosan and the small molecule drug UK5099 with hair growth treatment effect in Example 6-1.
  • Figure 6-2 shows the cumulative permeation of perfluoroheptanoic acid-modified chitosan in different proportions and the small molecule drug UK5099 with hair growth treatment effects in Example 6-1 at different times.
  • Figure 6-3 shows the hair growth on the back of mice at different time points treated with different treatment methods in Example 6-1.
  • FCS-Metformin is an experimental group in which perfluoroheptanoic acid modified chitosan is modified with metformin for transdermal transdermal, and metformin is a free drug group.
  • Figure 6-4 shows Example 6-2.
  • the left picture shows the efficiency of perfluoroheptanoic acid-modified chitosan and poly(I:C) to produce retinoic acid in mouse fibroblasts.
  • the right picture shows the efficiency of perfluoroheptanoic acid-modified chitosan and poly(I:C).
  • Figure 6-5 shows that in the diffusion cell penetration experiment of rat skin in Example 6-3, the mixture of perfluoroheptanoic acid-modified chitosan and tranexamic acid and pure tranexamic acid within three hours of tranexamic acid The cumulative percentage of the total permeation through rat skin [(cumulative permeation amount/theoretical total permeation amount) X 100%].
  • THP epirubicin
  • EDC 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • NHS N-hydroxysulfosuccinimide
  • DMSO dimethyl sulfoxide
  • MPI polypeptide drug Polybia-MPI
  • MPI-Cy5.5 fluorescence labeling of polypeptide drug MPI
  • PEI polyethylene imine
  • FPEI fluorinated polyethylene imine
  • CAT protein drug catalase
  • CAT-Ce6 composite protein drug labeled with photosensitizer Ce6
  • CAT-TCPP composite protein drug labeled with sonosensitizer TCPP
  • the experimental designers used adriamycin, epirubicin (THP) and the fluorescent dye rhodamine B as bladder perfusion drugs and chitosan (1% acetic acid aqueous solution) to prepare chitosan bladder perfusion drug system, 1h after bladder perfusion Frozen sections were taken from the bladder tissue, and the fluorescence intensity of the drug was analyzed with a confocal fluorescence microscope to investigate the distribution of the drug in the bladder tissue.
  • the experimental results show that chitosan (5mg/ml, 10mg/ml, 15mg/ml) solutions of equal concentrations of bladder infusion drugs have significantly better adhesion and penetration in the bladder mucosa than aqueous solutions perfused with pure drugs.
  • mice were kept under normal feeding conditions and recorded weight. The results of the experiment found that the weight of the mice dropped sharply on the second day after the chitosan was infused, and the mice were sluggish in activity. From the second day after treatment, the mice died, and within 3 days, all 8 mice in the experimental group died.
  • the bladder of the mice in the control group was compared with the bladder of the mice in the control group. It was found that the mice in the chitosan perfusion group Compared with the blank group, the bladder was more congested.
  • the results of HE staining and CD45 and Ki67 immunofluorescence showed that the bladder of the chitosan perfusion group had severe inflammatory stress and congestion and edema.
  • the above experimental results show that although chitosan can significantly improve the bioavailability of the infusion drug in the bladder mucosa, high concentrations of chitosan can also cause severe bladder mucosa and epithelial damage, which severely limits its clinical use as a drug carrier for bladder infusion application.
  • Example 1-1 Preparation of chitosan with different modification degrees of 3-fluorobenzoic acid (degree of deacetylation ⁇ 95%, viscosity 100-200mpa.s), wherein the molar ratio of 3-fluorobenzoic acid to N-glucosamine unit They are 1:1.1, 1:2.2, 1:4.4, 1:8.8.
  • Synthesis method (1) Preparation of chitosan acetic acid aqueous solution: Weigh 200mg of fully dried chitosan and add 10ml 1% acetic acid aqueous solution, of course, hydrochloric acid aqueous solution can also be used, stir for 30min to fully dissolve, then slowly add 1.6ml 0.5 0.5 M sodium hydroxide, stir until the solution is clear and the pH is about 6.5. From the perspective of simply considering the alkalization solution, sodium hydroxide can be replaced by ammonia, triethylamine and other alkalis, but from the product process perspective, the by-product of sodium hydroxide is sodium chloride, which is more suitable for industrialization.
  • the material obtained from the above reaction was tested by the ninhydrin reaction method to detect the degree of modification of the fluorinated aliphatic chain on the surface of the fluorinated modified chitosan (FCS) polymer.
  • the ninhydrin reaction method is a simple, fast, accurate and reliable method that can accurately detect the number of primary amino groups on the surface of FCS polymer in the aqueous solution, and then calculate the number of fluorinated groups on the surface of FCS.
  • Example 1-2 Preparation of chitosan with different modification degrees of heptafluorobutyric acid (degree of deacetylation ⁇ 95%, viscosity 100-200mpa.s), wherein the molar ratios of perfluoroheptanoic acid and N-glucosamine unit are respectively 1:1.1, 1:2.2, 1:4.4, 1:8.8.
  • Synthesis method (1) Preparation of chitosan acetic acid aqueous solution: Weigh 200mg of fully dried chitosan into 10ml 1% acetic acid aqueous solution, stir for 30min to fully dissolve, then slowly add 1.6ml 0.5M sodium hydroxide dropwise and stir Until the solution is clear, the pH is around 6.5. In this way, 4 parts of chitosan acetic acid aqueous solution were prepared.
  • the reaction was slowly added dropwise to 100ml 0.5M potassium hydroxide ethanol solution and stirred for 8h, the precipitate was filtered, washed with a large amount of absolute ethanol until the filtrate was neutral, the precipitate was washed with methanol and ether for dehydration, and dried under vacuum for 30 min.
  • the dried precipitate is dissolved in 10ml 0.1M hydrochloric acid solution, and freeze-dried to obtain white powder with different fluorination modification degree perfluoroheptanoic acid fluorinated chitosan hydrochloride molecules (the product is named 7FCS-1, 7FCS-2, 7FCS-3, 7FCS-4).
  • the material obtained from the above reaction was tested by the ninhydrin reaction method to detect the degree of modification of the FCS (fluorinated modified chitosan) polymer surface modified fluorinated aliphatic chain.
  • the ninhydrin reaction method is a simple, fast, accurate and reliable method that can accurately detect the number of primary amino groups on the surface of FCS polymer in the aqueous solution, and then calculate the number of fluorinated groups on the surface of FCS.
  • the ninhydrin reaction method calculates the degree of fluorination modification of FCS prepared above as follows: 7FCS-1, 6.9%; 7FCS-2, 10.4%; 7FCS-3, 23.5%; 7FCS-4, 42.3%.
  • Example 1-3 Preparation of chitosan with different modification degrees of perfluoroheptanoic acid (degree of deacetylation ⁇ 95%, viscosity 100-200mpa.s), wherein the molar ratios of perfluoroheptanoic acid to N-glucosamine unit are respectively 1:1.1, 1:2.2, 1:4.4, 1:8.8.
  • Synthesis method (1) Preparation of chitosan acetic acid aqueous solution: Weigh 200mg of fully dried chitosan into 10ml 1% acetic acid aqueous solution, stir for 30min to fully dissolve, then slowly add 1.6ml 0.5M sodium hydroxide dropwise and stir Until the solution is clear, the pH is around 6.5. In this way, 4 parts of chitosan acetic acid aqueous solution were prepared.
  • the reaction was slowly added dropwise to 100ml 0.5M potassium hydroxide ethanol solution and stirred for 8h, the precipitate was filtered, washed with a large amount of absolute ethanol until the filtrate was neutral, the precipitate was washed with methanol and ether for dehydration, and dried under vacuum for 30 minutes.
  • the dried precipitate was dissolved in 10ml 0.1M hydrochloric acid solution, and lyophilized to obtain white powder with different degrees of fluorination modification perfluoroheptanoic acid fluorinated chitosan hydrochloride molecules (products named 13FCS-1, 13FCS-2, 13FCS-3, 13FCS-4).
  • the ninhydrin reaction method calculates the degree of fluorination modification of FCS prepared above as follows: 13FCS-1, 5.2%; 13FCS-2, 11.3%; 13FCS-3, 21.4%; 13FCS-4, 42.5%.
  • the linking efficiency of 13FCS-1 ⁇ 13FCS-413 fluoroheptane carbonyl group is 5.2% ⁇ 42.5% with the increase of perfluoroheptanoic acid, that is to say, there are 5.2% ⁇ 42.5% glucose structural units in each chitosan molecule.
  • the fluorination modification was completed in, and the products were named 13FCS-1, 13FCS-2, 13FCS-3, 13FCS-4.
  • Example 1-4 Preparation of chitosan with different modification degrees of 19F decanoic acid (degree of deacetylation ⁇ 95%, viscosity 100-20 0mpa.s), wherein the molar ratio of 19F decanoic acid to N-glucosamine unit is 1 respectively : 1.1, 1:2.2.
  • Synthesis method (1) Preparation of chitosan acetic acid aqueous solution: Weigh 200mg of fully dried chitosan into 10ml 1% acetic acid aqueous solution, stir for 30min to fully dissolve, then slowly add 1.6ml 0.5M sodium hydroxide dropwise and stir Until the solution is clear, the pH is around 6.5. In this way, 2 parts of chitosan acetic acid aqueous solution were prepared.
  • (2) Activation of 19F decanoic acid Weigh 18 mg and 36.7 mg of 19F decanoic acid respectively, dissolve them in an appropriate amount of anhydrous dimethyl sulfoxide, add an appropriate amount of EDC, and stir for 1 hour with NHS protected from light.
  • the dried precipitate was dissolved in 10ml 0.1M hydrochloric acid solution, and lyophilized to obtain 19F decanoic acid fluorinated chitosan hydrochloride molecules with different fluorination modification degrees (the products were named 19FCS-1, 19FCS-2).
  • the water solubility of 19FCS-2 is relatively poor, and subsequent characterization and application evaluation cannot be performed. Therefore, the fluorination modification degree of 19FCS-1 prepared above is calculated by the ninhydrin reaction method as follows: 19FCS-1, 5.2%.
  • Example 1-5 Evaluation of bladder mucosal penetration enhancement effect of 7FCS: The 7FCS prepared in Example 1-2 and the THP aqueous solution were mixed, and infused into the bladder through the mouse urethra, and then frozen sections of the mouse bladder were prepared. Detect the distribution of THP fluorescence in the tissue to evaluate the drug carrier's absorption efficiency of the drug in the bladder mucosa.
  • the specific method is: 10-12 weeks of female C57BL/6 mice are anesthetized with pentobarbital solution, 0.2% THP solution is prepared with 0.5% FCS aqueous solution, and the mice are infused through a closed intravenous indwelling needle Bladder, 100 ⁇ l, clamp the urethra for 1h, then release the perfusate in the bladder, rinse the bladder with 1ml ultrapure water, take the bladder tissue into the tissue embedding machine and place it at -80°C, and then slice it with a fluorescent confocal microscope Detection.
  • a pure THP aqueous solution of equal concentration or the same prepared THP chitosan aqueous solution was used as a control.
  • FCS can significantly improve the penetration and absorption of the drug and enhance the absorption efficiency of the drug in the bladder mucosa, but excessive fluorinated substitution may not be conducive to the application of the material.
  • Example 1-6 The effect of 13FCS on promoting bladder mucosal absorption of the bladder perfusion drug: The 13FCS prepared in Example 1-3 and the THP aqueous solution were mixed, and the bladder was infused through the mouse urethra, and then frozen sections of the mouse bladder were prepared. By detecting the distribution of THP fluorescence in tissues, the drug carrier's absorption efficiency of the drug-promoting bladder mucosa was evaluated.
  • the specific method is: 10-12 weeks old female C57BL/6 mice are anesthetized with pentobarbital solution, and 0.2% THP solution is prepared with 0.5% 13FCS aqueous solution, and the mice are infused through a closed intravenous indwelling needle. Bladder, 100 ⁇ l, clamp the urethra for 1h, then release the perfusate in the bladder, rinse the bladder with 1ml ultrapure water, take the bladder tissue into the tissue embedding machine and place it at -80°C, and then slice it with a fluorescent confocal microscope Detection. The same prepared THP chitosan aqueous solution was used as a control.
  • Example 1-7 In order to screen the fluorinated chitosan with the best permeation effect on the bladder mucosa of the perfusion-promoting drug, mice were performed on 19FCS-1 in Examples 1-4 and the FCS with the best effect among the above-mentioned fluorinated modification types. In vivo evaluation.
  • the specific method is: 10-12 weeks of female C57BL/6 mice are anesthetized with pentobarbital solution, and 0.2% THP solution is prepared with 0.5% 7FCS-4, 13FCS-3, and 19FCS-1 aqueous solutions respectively.
  • a pure THP aqueous solution of equal concentration or the same prepared THP chitosan aqueous solution was used as a control.
  • Example 1-8 In vitro and in vivo safety evaluations of different types of fluorinated modified fluorinated chitosan in Examples 1-7 were carried out. The specific experimental schemes are as follows:
  • the CCK-8 (Cell Counting Kit-8) method (a mature in vitro evaluation method for evaluating cell activity) was used to evaluate the cytotoxic effect of fluorinated chitosan on SV-HUC-1 human normal bladder epithelial cells.
  • the in vitro biosafety of sugar the specific operation is as follows: inoculate 1x104 T24 cells per well into a 96-well plate, incubate overnight at 37°C, 5% CO 2 , and add different types of fluorinated chitosan (500ug/ml) serum-free The medium was cultured for 24 hours, and then an appropriate amount of cck-8 was added. Finally, the in vitro safety of fluorinated chitosan was evaluated by cell viability.
  • 13F-3 (abbreviation of 13FCS-3) has good cell safety in vitro. Combined with the above research results, it shows that 13F-3 has the most significant effect of promoting the absorption of the perfusion drug into the bladder mucosa. At the same time, it has better cell safety in vitro. In order to further evaluate the biological safety of FCS (13F-3), further safety evaluation experiments in mice were carried out.
  • mice The 10-12 weeks of healthy C57BL/6 mice were divided into three groups, each with 8 mice.
  • the experimental group was perfused with 15mg/ml fluorinated chitosan or chitosan 1% acetic acid aqueous solution for 1 hour, once a week for three weeks, and the blank control group was perfused with equal volume of double distilled water.
  • Example 1-9 Application of F-PEI bladder perfusion polypeptide protein drug carrier, taking polypeptide drug MPI and protein drug CAT-Ce6 as examples
  • MPI/F-PEI, CAT/F-PEI NPs can be obtained by mixing polypeptide (MPI), protein (CAT) drugs and F-PEI aqueous solution for 2 hours at room temperature.
  • the hydrated particle size measured by the dynamic light scattering instrument is about 200-300nm with a small amount of positive charge.
  • Transmission electron microscopy (TEM) imaging ( Figure 1-6, 1-7) characterizes it as a uniform spherical particle.
  • Ussing chamber (also called Ussing chamber) is a tool for studying transepithelial transport and can be used for research including ion transport, nutrient transport, and drug transport. Through the study of transepithelial transport, we can understand the absorption of epithelial drugs through the epithelium.
  • the Using chamber was used to evaluate the mucosal permeability of MPI-cy5.5/F-PEI NPs, CAT-ce6/F-PEI NPs prepared with different material ratios, respectively, using MPI-cy5.5/PEI NPs, CAT-cy5.5/PEI NPs, and CAT-ce6/F-PEI NPs.
  • ce6/PEI NPs was used as a control, the mice were anesthetized, the bladder was taken on ice and the bladder mucosa was peeled off, and it was fixed at the interface between the two chambers.
  • the experimental results showed that the mucosal permeability index papp of F-PEI group polypeptide (Figure 1-8) or protein drug (Figure 1-9) was significantly higher than that of PEI group and free group (blank group).
  • the experimental results show that the polypeptide and protein drugs in the F-PEI group have more significant bladder mucosal permeability compared with the PEI and free drug groups, that is, F-PEI can significantly improve the bladder mucosal permeability of polypeptide or protein drugs.
  • Example 1-10 Application of FCS Bladder Perfusion Protein Drug Carrier, taking the protein drug CAT-TCPP as an example.
  • CAT-TCPP/FCS NPs can be obtained by mixing protein drugs (CAT-TCPP) drugs and FCS aqueous solution at room temperature for 2 hours.
  • the hydrated particle size measured by a dynamic light scattering instrument is about 200-300nm with a small amount of positive charge.
  • Transmission electron microscopy (TEM) imaging Figure 1-12 characterizes it as a uniform spherical particle.
  • the perfluoroheptanoic acid modified chitosan used in all the examples of this patent is the molar ratio of perfluoroheptanoic acid to N-glucosamine unit in the examples 1-3 is divided into 1:4.2 fluorine Chitosan.
  • A is the molecular skeleton of chitosan containing primary amino groups, and the structural formula is as follows:
  • B is a linking group formed by a fluorine-containing functional group and a primary amino group of chitosan, where it is an amide bond.
  • C is a fluorine-containing aliphatic chain and aromatic ring functional group
  • perfluoroheptanoic acid is used in this patent, and the structural formula is as follows:
  • Example 2-1 Preparation of perfluoroheptanoic acid-modified chitosan as a carrier transdermal patch, transdermal delivery of insulin, and diabetes treatment.
  • fluorinated chitosan for the specific preparation process of fluorinated chitosan in this example, refer to Example 2-5.
  • FCS-Insulin perfluoroheptanoic acid
  • the ratios of chitosan-insulin modified by perfluoroheptanoic acid were 1:0.25, 1:0.5, 1:1, 1:2, 1:4 and pure immunoglobulin G (0:1), respectively.
  • the abscissa is the transmission time, and the ordinate is the cumulative transmission calculated from fluorescence. The results are shown in Figure 2-4, and the preferred ratio is 1:1.
  • the abscissa is time, and the ordinate is the cumulative amount of insulin permeation calculated based on Coomassie brilliant blue staining.
  • mice with perfluoroheptanoic acid modified chitosan drug-loaded transdermal patch had hyperglycemia Obviously inhibited and remained stable for a long time, indicating that perfluoroheptanoic acid modified chitosan can significantly improve the permeability of the drug in the skin, and promote the drug to enter the blood to maintain a stable blood drug concentration, and continue to play a role.
  • perfluoroheptanoic acid modified chitosan can significantly improve the permeability of the drug in the skin, and promote the drug to enter the blood to maintain a stable blood drug concentration, and continue to play a role.
  • the above results collectively indicate that cationic polymers modified by fluorine-containing compounds can successfully achieve transdermal drug delivery, and have greater medical value and transformational value.
  • Example 2-2 Preparation of perfluoroheptanoic acid-modified chitosan as a carrier transdermal ointment, transdermal delivery of apoptosis-ligand 1 antibody, for treatment of surface melanoma.
  • FCS-IgG perfluoroheptanoic acid-modified chitosan-immunoglobulin G complex
  • reaction mass ratio of perfluoroheptanoic acid-modified chitosan and immunoglobulin G is 1:0.25-4, and is further preferably 1:1 through dynamic light scattering particle size analysis and potential analysis.
  • the results of particle size distribution and potential distribution are shown in Figure 2-8.
  • Immunoglobulin refers to a globulin that has antibody activity or chemical structure and is similar to antibody molecules.
  • the immunoglobulin G used in the experiment of this patent is not specific.
  • the antibody is an immunoglobulin that can specifically bind to the antigen.
  • the apoptosis-ligand 1 antibody is also a type of immunoglobulin G, only the light chain end It has specificity, so immunoglobulin G can be used to simulate the behavior of apoptosis-ligand 1 antibody in non-therapeutic experiments.
  • FCS-IgG perfluoroheptanoic acid-modified chitosan-apoptosis-ligand 1 antibody transdermal kinetics analysis in vivo: using fluorescently labeled immunoglobulin G as a template to simulate the transdermal motility of apoptosis-ligand 1 antibody
  • FCS-IgG perfluoroheptanoic acid-modified chitosan-immunoglobulin G
  • the surface of the mouse tumor was smeared with a mixture of perfluoroheptane with a mass ratio of 1:1.
  • the acid-modified chitosan-immunoglobulin G ointment was fixed with a transdermal film to prevent it from falling.
  • the mice were killed at different time points, the tumor tissue was removed, the remaining ointment on the surface was wiped off and the epidermis was removed.
  • the tumor tissue was divided into two, half of which was lysed and the fluorescence intensity in the tissue was measured, and the other half was sliced for fluorescence imaging under a confocal microscope. The result is shown in Figure 2-10.
  • DAPI is a nuclear dye that can indicate the nucleus
  • FITC is a fluorescent secondary antibody labeled with apoptosis-ligand 1 antibody. It can be concluded that the perfluoroheptanoic acid modified chitosan-apoptosis-ligand 1 antibody group has the strongest transdermal efficiency under the same dose of apoptosis-ligand 1 antibody.
  • Perfluoroheptanoic acid-modified chitosan-cell apoptosis-ligand 1 antibody transdermal mechanism study using immunoglobulin G as a template to simulate cell apoptosis-ligand 1 antibody further analyzes the regulation of tight junctions between cells Protein changes after adding perfluoroheptanoic acid modified chitosan-immunoglobulin G (FCS-IgG). Incubate the human skin epithelial cells Hacat in a petri dish dedicated to a confocal microscope, and wait for them to grow into a dense cell monolayer.
  • FCS-IgG perfluoroheptanoic acid modified chitosan-immunoglobulin G
  • Perfluoroheptanoic acid-modified chitosan-cell apoptosis-ligand 1 antibody permeation mechanism using immunoglobulin G as a template to simulate cell apoptosis-ligand 1 antibody to further study the mechanism of tight junction protein distribution changes .
  • tight junction proteins are occludin (Occludin), transmembrane integral protein (Claudin-1), cadherin E (E-Cadherin) and tight junction protein-1 (ZO-1); actin is MLC, phosphoric acid The modified actin is p-MLC; GAPDH is glyceraldehyde-3-phosphate dehydrogenase, which has relatively stable expression in various tissues and is used here as an internal control. The result is shown in Figure 2-14.
  • the left picture is a graph of mouse tumor growth curve, and the right picture is a broken line graph of mouse survival rate.
  • the tumor size is 1500 cubic millimeters as the standard of mouse death. It can be found that due to the small initial volume, the meridian injection therapy routinely used in clinical practice also has a certain inhibitory effect on mouse tumors, but in contrast, perfluoroheptanoic acid modified chitosan-cell apoptosis-ligand 1 antibody Because of the antibody penetration rate of more than 50% in the group, the therapeutic effect is much higher than the other groups.
  • the cationic polymers modified by fluorine-containing compounds in this patent especially the drugs involved in fluorinated chitosan include, but are not limited to, diabetes treatment drugs, anti-tumor drugs (see Table 2-1 for details), immunomodulators, and antiviral drugs , Anti-inflammatory drugs, analgesic anesthetics, medical beauty drugs and other drugs and various dosage forms of their derivatives.
  • the drug may be an immunomodulator, including but not limited to cytokines, BCG, immune checkpoint blocking antibodies, and the like.
  • Cytokines are synthesized and secreted by immune cells (such as monocytes, macrophages, T cells, B cells, NK cells, etc.) and certain non-immune cells (endothelial cells, epidermal cells, fibroblasts, etc.) after stimulation
  • immune cells such as monocytes, macrophages, T cells, B cells, NK cells, etc.
  • non-immune cells endothelial cells, epidermal cells, fibroblasts, etc.
  • Cytokines include but are not limited to interleukin (IL), interferon (IFN), tumor necrosis factor (TNF), colony stimulating factor (CSF), chemokine (chemokine family), growth factor (growth factor, GF), transforming growth factor- ⁇ family (transforming growth factor- ⁇ family, TGF- ⁇ family).
  • Interleukins include but are not limited to IL-1-IL-38.
  • Colony stimulating factors include but are not limited to G (granulocyte)-CSF, M (macrophage)-CSF, GM (granulocyte, macrophage)-CSF, Multi (multiple)-CSF (IL-3), SCF, EPO etc.
  • Interferons include, but are not limited to, including but not limited to IFN- ⁇ , IFN- ⁇ , and IFN- ⁇ .
  • Tumor necrosis factors include but are not limited to TNF- ⁇ and TNF- ⁇ .
  • the transforming growth factor- ⁇ family includes but is not limited to TGF- ⁇ 1, TGF- ⁇ 2, TGF- ⁇ 3, TGF ⁇ 1 ⁇ 2, and bone morphogenetic protein (BMP).
  • Growth factors include, but are not limited to, epidermal growth factor (EGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), insulin-like growth factor-I (IGF-1) , IGF-II, Leukemia Inhibitory Factor (LIF), Nerve Growth Factor (NGF), Oncostatin M (OSM), Platelet Derived Endothelial Growth Factor (PDECGF), Transforming Growth Factor- ⁇ (TGF- ⁇ ), Vascular Endothelial cell growth factor (VEGF).
  • EGF epidermal growth factor
  • PDGF platelet-derived growth factor
  • FGF fibroblast growth factor
  • HGF hepatocyte growth factor
  • IGF-II insulin-like growth factor-I
  • LIF Leukemia Inhibitory Factor
  • NGF Nerve Growth Factor
  • OSM Oncostatin M
  • PDECGF Platelet Derived Endothelial Growth Factor
  • TGF- ⁇ Vascular End
  • the chemokine family includes, but is not limited to, four subfamilies: (1) CXC/ ⁇ subfamily, mainly chemotactic neutrophils, the main members are IL-8, melanoma cell growth stimulating activity (GRO/MGSA) , Platelet factor-4 (PF-4), platelet basic protein, proteolytic products CTAP-III and ⁇ -thromboglobulin, inflammatory protein 10 (IP-10), ENA-78; (2) CC/ ⁇ subfamily ,
  • the main chemotactic monocytes, members of this subfamily include macrophage inflammatory protein 1 ⁇ (MIP-1 ⁇ ), MIP-1 ⁇ , RANTES, monocyte chemotactic protein-1 (MCP-1/MCAF), MCP- 2. MCP-3 and I-309.
  • the representative of type C subfamily includes lymphocyte chemotactic protein.
  • CX3C subfamily, Fractalkine is a CX3C chemokine, which has a chemotactic effect on monocytes-macrophages, T cells and NK cells.
  • Cytokines include, but are not limited to, cytokines used to treat cancer and cytokines that reduce the side effects of cancer treatment. They play an important role in the normal immune response of the human body and the ability of the immune system to respond to cancer. Cytokines used to treat cancer include but are not limited to interferons and interleukins. Cytokines can also be hematopoietic growth factors, which reduce the side effects of cancer treatment by promoting the growth of blood cells destroyed by chemotherapy. Cytokines that reduce the side effects of cancer treatment include but are not limited to erythropoietin, IL-11, granulocyte-macrophage colony stimulating factor (GM-CSF) and granulocyte-colony stimulating factor (G-CSF).
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • G-CSF granulocyte-colony stimulating factor
  • BCG vaccine is a live vaccine made from the suspension of attenuated Mycobacterium bovis, which can enhance the activity of macrophages, enhance the body's cellular immunity, and can be used to treat skin cancer.
  • Immunomodulatory drugs including but not limited to thalidomide Lenalidomide Pomali Imiquimod Immune checkpoint blocking antibodies include but are not limited to CTLA4 monoclonal antibody PD-1 monoclonal antibody PD-L1 monoclonal antibody LAG-3 (lymphocyte activation gene 3) monoclonal antibody, TIM-3 (T cell immunoglobulin and mucin domain protein 3) monoclonal antibody, TIGIT (T cell immunoglobulin and ITIM domain protein) monoclonal Antibodies, co-stimulatory factors B7-H3, B7-H4 and B7-H5 monoclonal antibodies and other drugs and their derivatives.
  • the drug may be an anesthetic drug.
  • Example general anesthetics include, but are not limited to, ketamine hydrochloride, propofol, thiopental sodium, etomidate, midazolam, and sodium gamma-hydroxybutyrate.
  • Local anesthetics include but are not limited to aromatic acid esters, aromatic amides, amino ketones, amino ethers, carbamates, hydroxyprocaine, chloroprocaine, tetracaine, butacaine, Thiocaine, procainamide, bupivacaine, articaine, eticaine, ropivacaine, mepivacaine, clonin and other drugs and their derivatives.
  • the drug may be a diabetes treatment drug, including but not limited to sulfonylureas such as tolbutamide, tolbutamide, chlorpropamide, chlorhexamide, gliclazide, glipidin, glimepiride, etc.
  • sulfonylureas such as tolbutamide, tolbutamide, chlorpropamide, chlorhexamide, gliclazide, glipidin, glimepiride, etc.
  • Non-sulfonylurea drugs such as repaglinide and nateglinide and their derivatives
  • thiazolidinedione drugs such as rosiglitazone and piroglitazone and their derivatives
  • phenylethyl Biguanide metformin and other biguanide drugs and their derivatives
  • acarbose voglibose, miglitol and other ⁇ -glucosidase inhibitor drugs and their derivatives
  • glucagon-like peptide, DPP -IV inhibitors sitagliptin, vildagliptin, saxagliptin dipeptidyl peptidase-IV drugs and their derivatives, insulin and other drugs and their derivatives.
  • Diabetes is a metabolic endocrine disease characterized by high blood sugar.
  • the clinical dosage form is generally insulin injection.
  • Patients need to endure the pain of repeated injections, and long-term medication can also cause side effects such as inflammation and induration at the injection site.
  • Chitosan drugs modified by fluorine-containing compounds can penetrate the skin, carry hypoglycemic drugs into the blood, and improve the bioavailability of the drugs.
  • fluorine-containing compound-modified chitosan can be used as a drug carrier to deliver hypoglycemic drugs, which can be administered in the form of drug application for the treatment of diabetes.
  • the chitosan drug patch modified by fluorine-containing compound is administered in a transdermal manner.
  • the effective concentration of the drug is maintained for a long time.
  • the degree of action and the duration of the drug can be adjusted according to the area of application and the application time. It is flexible, The advantage of convenience. In addition, it can also be prepared into a more flexible lotion, liniment, smear and other dosage forms.
  • the drug may be an anti-tumor drug (see Table 2-1 for details).
  • transdermal delivery is a non-invasive way of delivery, although it brings great convenience, the stratum corneum barrier of the skin often prevents the drug from entering the subcutaneous lesion or even entering the blood vessel.
  • Melanoma is a malignant tumor that originates from cells that can produce melanin. It is easily metastatic, has strong drug resistance, has a poor prognosis, and has an extremely high mortality rate.
  • Melanoma chemotherapy drugs are mainly delivered by oral and injection methods, but this often causes many adverse reactions and even leads to organ damage. At the same time, it is impossible to deliver drugs efficiently, accurately and controllably.
  • Transdermal delivery methods have unique advantages for the treatment of subcutaneous melanoma, but still put forward higher requirements for the efficiency of transdermal delivery.
  • fluorine-containing compound-modified chitosan can be used as a drug carrier to deliver anti-tumor drugs, and be administered in the form of ointment for tumor treatment.
  • the fluorinated chitosan in each of the embodiments of the present patent 2-1 to 2-6 can be used as a transdermal preparation, and used as a transdermal drug preparation for treating diabetes, tumor disease, and anti-inflammatory drugs. At the same time, it can also be used as a transdermal drug delivery agent in the preparation of medical beauty drugs, external drug preparations, medical device external preparations, and cosmetic skin care products.
  • Example 3-1 Preparation of an oral drug with perfluoroheptanoic acid modified chitosan as a carrier, and oral delivery of insulin for diabetes treatment.
  • FCS-Insulin perfluoroheptanoic acid-modified chitosan-insulin
  • the drug-loaded capsule into simulated gastric juice or simulated intestinal juice, and characterize its release effect in simulated gastric juice or simulated intestinal juice by the fluorescence intensity of chitosan-insulin modified by perfluoroheptanoic acid released in the solution.
  • the left side is the release behavior in simulated gastric juice
  • the right side is the release behavior in intestinal juice.
  • the capsule can remain stable and not released for a long time in the simulated gastric juice, while the drug can be released in the simulated intestinal juice.
  • the abscissa is the time after the capsule is administered, and the ordinate is the blood glucose concentration.
  • Example 3-2 Preparation of oral capsules with perfluoroheptanoic acid-modified chitosan as a carrier, oral delivery of apoptosis-ligand 1 antibody, and observation of different ratios of perfluoroheptanoic acid-modified chitosan-apoptosis -The transmucosal effect of ligand 1 antibody particles.
  • Perfluoroheptanoic acid-modified chitosan-apoptosis-ligand 1 antibody permeates the intestinal mucosa kinetics in vitro: Take the rat intestine, remove the fascia layer, and fix it on the transdermal diffusion cell. Add different mass ratios of perfluoroheptanoic acid modified chitosan-FITC-immunoglobulin G particles on the transdermal diffusion cell, and add PBS solution under the transdermal diffusion cell at 0min, 15min, 30min, 45min, 60min, 90min, 120min, 150min, 180min, 210min take the liquid under the transdermal diffusion cell, measure the fluorescence intensity, and calculate the cumulative transmittance. The results are shown in 3-9.
  • Perfluoroheptanoic acid-modified chitosan-apoptosis-ligand 1 antibody particle freeze-dried powder before and after lyophilization activity determination use carbonate buffer to dilute the frozen cell apoptosis-ligand 1 to different concentrations , And added to the enzyme-linked immunosorbent method enzyme labeling plate, placed at 4 °C overnight. After the coating is finished, wash with PBST three times, add BSA solution, and block for 2h at room temperature. After blocking, wash with PBST three times, add the apoptosis-ligand 1 antibody before and after freeze-drying, and incubate at room temperature for two hours.
  • FCS/IgG FCS MEN serum-free medium solution
  • mice were anesthetized with 1% sodium pentobarbital, and each mouse was injected with a bioluminescent substrate. Ten minutes later, the mice were imaged to observe the tumor growth of the mice. The experimental results are shown in Figure 3-14.
  • perfluoroheptanoic acid modified chitosan with different molecular weights has different effects of increasing the drug's mucosal penetration. This indicates that perfluoroheptanoic acid modified chitosan can promote the drug to enter tumor tissues.
  • Example 3-3 Preparation of perfluoroheptanoic acid-modified chitosan-immunomodulator particles with perfluoroheptanoic acid-modified chitosan as a carrier, administered by spray/inhalation, and investigating drug delivery to the lungs Ability.
  • Example 3-4 Preparation of bovine serum albumin (BSA-Cy5.5) labeled with Cy5.5 fluorescent molecule as a model protein, and perfluoroheptanoic acid modified chitosan (FCS) as a transnasal mucosal carrier
  • BSA-Cy5.5 bovine serum albumin labeled with Cy5.5 fluorescent molecule
  • FCS perfluoroheptanoic acid modified chitosan
  • FCS perfluoroheptanoic acid modified chitosan
  • FCS/BSA-Cy5.5 nanoparticles Dissolve perfluoroheptanoic acid-modified chitosan in 1% acetic acid. After it is completely dissolved, add a weak base to adjust the pH of the solution to 6-7, and then Mix it with the BSA-Cy5.5 solution, and place it at 4°C for 1 hour with stirring to form stable nanoparticles as nasal drops.
  • the preferred reaction mass ratio of perfluoroheptanoic acid-modified chitosan and BSA is 1:1, and the final bovine serum albumin concentration is 2.5 mg/mL.
  • brain glioma model male C57BL/6 mice (about 20 g/mouse) at 7-8 weeks of age were injected with EGFP-expressing glioma cells (5000 cells/mouse) to construct brain glioma Tumor model.
  • Nasal administration Anesthetize the successfully modeled C57BL/6 mice and fix them on a heating pad in a supine position. Use a pipette to slowly drip FCS/BSA-Cy5 with a total volume of 20 ⁇ L into the mouse nostrils. .5 solution (free BSA-Cy5.5 solution as the control group), after the dripping, continue to maintain the supine position of the mouse for 30 minutes.
  • mice were anesthetized and perfused with formalin. Then the brain tissue was taken out, lyophilized, sectioned, and the fluorescence signal of Cy5.5 at the tumor tissue in the brain of the mouse was observed through a confocal microscope.
  • the left image represents the EGFP signal of brain tumors in mouse brain tissue
  • the right image represents the Cy5.5 signal of BSA delivered to the brain via the nose. It can be seen from the results on the right that: compared with the free BSA-Cy5.5 and CS/BSA-Cy5.5 in the control group, the brain tissue of the whole mouse taken out after the nasal drip operation in the experimental group FCS/BSA-Cy5.5 Cy5.5 has the highest fluorescence intensity, indicating that FCS/BSA-Cy5.5 has the best nasal brain delivery efficiency.
  • EGFP tumor tissue
  • Cy5.5 IgG
  • Example 4-1 Taking bovine serum albumin labeled with Cy5.5 fluorescent substance and small-molecule fluorescent substance rhodamine B as examples, fluorine-containing modified chitosan was used to encapsulate the drug, and the eye penetration time and penetration time were observed ⁇ depth.
  • mice were anesthetized and administered on the ocular surface with a metering device, 5 ⁇ L per eye, and the mice were treated in the dark for 12 hours.
  • the control group was bovine serum albumin labeled Cy5.5 without fluorine-containing chitosan, and the concentration and dose were the same as the experimental group.
  • the specific implementation method of the small molecule drug Rhodamine B is the same as that of the large molecule drug.
  • the membrane penetration effect of large molecule bovine serum albumin is shown in Figure 4-4, and the membrane penetration effect of small molecule Rhodamine B is shown in Figure 4-5.
  • perfluoroheptanoic acid modified chitosan can enter the inside of the eyeball, and the fluorescence intensity and small molecule fluorescence of the protein marker are much higher than that of free protein and free small molecule
  • perfluoroheptanoic acid modified chitosan can help a series of drugs penetrate the eye barrier and enter the eye to achieve the purpose of treatment.
  • the chitosan group modified with perfluoroheptanoic acid is significantly higher than the free protein group.
  • 6 hours of perfluoroheptanoic acid-modified chitosan protein group has higher fluorescence intensity than 3 hours. It can be seen that the perfluoroheptanoic acid modified chitosan may also adhere to the ocular surface. Slow release into the eyes.
  • the cornea was stained by immunofluorescence at different time points to observe the penetrating ability. Frozen sections were sampled 5 minutes, 15 minutes, 30 minutes, and 60 minutes after the drug containing perfluoroheptanoic acid-modified chitosan was applied, and the nuclei of the sections were stained and observed under a confocal microscope as shown in Figure 4-7. The red is the fluorescence of protein markers, and the blue is the nucleus of the eye tissue. It can be clearly seen from the figure that after 30 minutes, the chitosan group modified with perfluoroheptanoic acid has clearly penetrated into the corneal epithelial cells, while the free protein has hardly entered. It can be seen that chitosan modified with perfluoroheptanoic acid can open the corneal barrier and treat corneal-related diseases.
  • the defect area was reduced, and the defect area was less than 30%; after 48 hours, there were 2 mouse corneal epithelium defects, and the other 3 epithelium was completely healed; 72 hours later, 5 mice corneas The epithelium is completely healed.
  • Normal saline group 24 hours after the treatment, the defect area was reduced.
  • the corneal epithelial defect area of 2 mice was between 30% and 70%, and the other three mice were less than 30%.
  • One mouse had corneal epithelial defect after 48 hours.
  • the area of the defect was less than 30%, and the epithelium of 4 mice was completely healed; after 72 hours, there was an epithelial defect in the area of the epithelial defect of 1 mouse, and the area of the defect was less than 30%, and the corneal epithelium of 4 mice was completely healed.
  • 24 hours after PBS administration the defect area was reduced, and the defect area was less than 30%; after 48 hours, 1 mouse corneal epithelium had punctate staining, 4 epithelium was completely healed; 72 hours later, 5 mouse corneal epithelium was healed completely.
  • the defect area was reduced 24 hours after the treatment, and the defect area was less than 30%; after 48 hours, the corneal epithelium of 5 mice was completely healed.
  • chitosan modified with perfluoroheptanoic acid has extremely high safety to the eyes.
  • the cornea stained with sodium fluorescein under the slit lamp is stained if there is a corneal defect, and it should be gray in black and white mode.
  • Table 4-4 shows the evaluation results of Figure 4-8.
  • Example 4-2 Using anti-PDL1 as immunotherapeutic drug and preparing eye drops with perfluoroheptanoic acid-modified chitosan to treat choroidal malignant melanoma, proving that perfluoroheptanoic acid-modified chitosan can deliver effect.
  • Animal model B16 melanoma transfected with bioluminescence gene in logarithmic phase was injected into the right eye of Balb/c mouse eyeball choroid through the eye, each eye was injected with 1 ⁇ 10 5 cells, and the injection was nurtured. On 4 days, the bioluminescence intensity was used to express the tumor size through the bioluminescence imaging system. See Figure 4-9. The size of eye tumors in each group before administration.
  • mice that have been successfully modeled are divided into groups, the unadministered group is the control group, and the eye drops are the experimental group, with three mice in each group.
  • the treatment was started on the fourth day after the model was established.
  • the experimental group was instilled once a day, 2.5 ⁇ L each time, and the drug concentration was 2 mg/mL.
  • bioluminescence imaging was performed. The results are shown in Figure 4-9 and Figure 4-10.
  • the upper three mice in Figure 4-9 are the control group, and the lower three are the experimental group. It is obvious from the figure that the immune bioluminescence of mice containing perfluoroheptanoic acid-modified chitosan after one week of treatment is weaker than that of the control group.
  • the bioluminescence quantitative analysis of in situ choroidal malignant melanoma after one week of treatment shows that the autoluminescence after treatment The luminous intensity is one-fourth that of the control group, which has a significant therapeutic effect.
  • cy5.5-labeled chicken ovalbumin Weigh 10 mg of chicken ovalbumin, dissolve it with 1 mL of PBS, add 20 ⁇ L (20 mg/mL) of cy5.5, and place it overnight at 4°C. Remove free cy5.5 through G25-molecular sieve gel. It was quantified by BSA, and then OVA was concentrated by ultrafiltration to 20 mg/mL.
  • FCS-OVA vaccine ointment Take 12.5 ⁇ L of the above FCS-OVA complex solution and mix it into 12.5 ⁇ g ointment to prepare FCS-OVA vaccine ointment, and mix 12.5 ⁇ LOVA (2mg/mL) solution into 12.5 ⁇ g ointment to prepare OVA vaccine ointment.
  • the above ointment was applied to the back of C57 mice and fixed with an applicator for 12 hours. Repeat this operation twice a week for three weeks.
  • mice grown with perfluoroheptanoic acid-modified chitosan-chicken ovalbumin complex as a patch vaccine were compared with mice grown with chicken ovalbumin alone.
  • the tumor growth rate was slower, indicating that the perfluoroheptanoic acid modified chitosan-chicken ovalbumin complex as a patch has the effect of being a vaccine.
  • the fluorinated chitosan in each of Examples 1-1 to 1-4 of the present patent can be used as a vaccine preparation for various types of vaccination preparations.
  • Example 6-1 Taking the currently known small-molecule drug UK5099 with certain hair growth therapeutic effects as an example, fluorine-containing modified chitosan was used to encapsulate the drug to study different ratios of perfluoroheptanoic acid modified chitosan and The in vitro transdermal ability of the drug. Taking the small molecule drug metformin as an example, fluorine-containing modified chitosan was used to encapsulate the drug to study the actual effect of animal hair growth.
  • Preparation of perfluoroheptanoic acid-modified chitosan-metformin hair growth liquid first dissolve the perfluoroheptanoic acid-modified chitosan solid powder, add drop by drop the liquid that dissolves metformin under constant stirring to make the final metformin
  • concentration is 2mg/mL
  • concentration of perfluoroheptanoic acid modified chitosan is 2mg/mL.
  • mice choose Balb/c mice in the hair prohibition period at the sixth week, and remove the backs of the mice in the perfluoroheptanoic acid-modified chitosan-metformin group, free metformin group, and blank control group the day before the treatment. Area hair and take pictures as shown in the figure. Each mouse sprays the drug evenly according to its own treatment method, and the drug is given once every two days, and each dose is 100 microliters. The photos on the 11th, 13th, and 17th days after treatment are shown in Figure 6-3 and table 6-1.
  • mice in the chitosan group modified with perfluoroheptanoic acid have a clear trend of hair growth compared with the free drug and the control group.
  • Example 6-2 Preparation of perfluoroheptanoic acid-modified chitosan (FCS) and polyinosinic-polycytidylic acid (poly(I:C)) as the main body of scar removal cream to induce endogenous The production of retinoic acid, thereby using retinoic acid to improve scars.
  • FCS perfluoroheptanoic acid-modified chitosan
  • poly(I:C) polyinosinic-polycytidylic acid
  • HPLC High Performance Liquid Chromatography
  • Example 6-3 Taking tranexamic acid, a medicine for treating melasma as an example, fluorine-containing modified chitosan was used to encapsulate tranexamic acid, and the diffusion cell was used to explore its skin penetration ability.
  • Preparation of perfluoroheptanoic acid modified chitosan-tranexamic acid solution dissolve solid powder perfluoroheptanoic acid modified chitosan in PBS, and mix it with tranexamic acid dissolved in PBS 1:1, Vortex for 5 minutes to combine the two with electrostatic force.
  • the final concentration of perfluoroheptanoic acid-modified chitosan and tranexamic acid in the final solution are both 1 mg/mL.
  • Measurement of tranexamic acid transmission rate Take out 500 ⁇ L of PBS from the bottom diffusion cell at 0, 1, and 3 hours and add the same volume of PBS. Use methanol and 0.05mol/L KH2PO4-0.2% H3PO4 solution (volume ratio of 5:95) as mobile phase, flow rate of 1.0mL/min, detection wavelength of 210nm under HPLC conditions to detect tranexamic acid (tranexamic acid) content, And calculate the cumulative amount of penetration, the result is shown in the figure, the abscissa is time, and the ordinate means the cumulative amount of penetration.

Abstract

本发明公开了一种作为药物载体用的氟化修饰的壳聚糖衍生物,具有如下结构:含氟化合物共价连接在壳聚糖主链上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,所述含氟化合物为如下化学式(I) 所示的含氟脂肪链,或式(II) 所示的芳香环功能基团,所述R1为卤素(氟,氯,溴,碘)、卤素取代的烷烃、环烷烃、醛基、羧基、双键、炔键、羟基、磺酰氯、磺酸键或巯基这些能够与伯氨基反应的活性基团。本发明可以与多种药物进行普适性结合,促进药物吸收,提高药物的生物利用度,减少毒性的优点,效果好,应用十分广泛,具有巨大的商业价值,并本发明提出的含氟化合物修饰的阳离子聚合物生产简易,具备商业化的基础。

Description

含氟化合物修饰的阳离子聚合物作为药物载体的应用及制备方法 技术领域
本发明涉及高分子化学以及医药生物材料技术领域,具体涉及基于氟化物修饰改性的阳离子聚合物作为药物载体及其制备方法和应用。
背景技术
近年来亲水性阳离子高分子聚合材料如聚乙烯亚胺(PEI)、聚赖氨酸等其阳离子特性可以与核酸、多肽及蛋白分子结合形成纳米复合物,不仅促进这些大分子化合物进入细胞,而且能够保护药物不被微环境中的水解酶降解,其内部的三级氨结构通过质子海绵作用,促进药物在细胞内涵体的逃逸。同时,由于阳离子高分子聚合材料可通过削弱上皮细胞的紧密连接作用,增加上皮的渗透性,促进药物大分子在上皮细胞的吸收效率。但是,阳离子高分子聚合材料在使用过程中的高细胞毒性最终限制了其临床应用。
壳聚糖是甲壳质脱乙酰基后的阳离子多糖,具有良好的生物安全特性和优良的黏膜黏附性能,其已被广泛应用于经粘膜给药剂型的设计。文献报道壳聚糖可通过其自身正电荷与皮肤及黏膜表面的阴性电荷作用以及疏水基团的疏水效应产生黏膜粘附,有效的延长壳聚糖溶液中生物活性物质(药物、多肽、蛋白等)在病灶部位的滞留时间,在扩散或壳聚糖降解等后期作用的驱动下,使活性物质从壳聚糖溶液中缓慢释放,从而达到局部皮肤、黏膜的长效缓释效果。虽然壳聚糖可显著提高灌注药物的生物利用度,但高浓度的壳聚糖有可能会引起严重的黏膜、上皮损伤,限制了其作为药物载体的临床应用。
透皮给药是指药物透过皮肤,经毛细血管吸收后进入血液循环,达到有效的血药浓度进而产生药效的一种给药方式。透皮给药可以避免口服给药的肝脏首过效应,并避免药物在胃肠道失活,尤其是对于需要长期给药的病人来说,透皮给药是一种方便快捷的治疗方式。然而,皮肤作为人体的第一道屏障,可以阻碍大多数外源性物质的侵入。药物透过皮肤的速率往往较慢,透过量难以达到有效治疗所需的浓度,无法发挥最优的治疗效果。同时,透皮给药的药物制剂的剂量通常与给药系统与皮肤的有效接触面积有关,增加面积可以增加给药剂量,但一般给药面积不大于60cm2,因此要求药物有一定的透皮速率。除了小部分剂量需求小、具有适宜溶解特性的药物,大部分的药物难以满足治疗要求。
目前临床常用的透皮剂型包括含化学促渗透成分的贴片剂或凝胶剂以及通过物理促渗透的贴片或凝胶剂型。自1981年用于治疗晕动症的透皮贴片Transderm Scop上市以来,含化学促渗透成分的贴片剂和凝胶剂就被广泛应用于治疗多种疾病包括痴呆症、帕金森病和急性疼痛。迄今为止,已有超过20种透皮贴剂被美国FDA批准上市。但是现有技术或产品通常都存在透皮效果有限,药物生物利用度低、与多种药物无法很好的普适性结合,毒性较大等一种或多种缺陷。
寻求药物的非注射给药方式一直是药剂学、生物化学等领域的研究热点,涉及给药途径包括口服给药、肺部给药、鼻腔给药、直肠给药、阴道给药等方式。这些非注射给药方式,具有用药方便、减小患者用药痛苦、提高患者使用依从性等优点,成为外用制剂研究的 热点。但在实际应用中,仍存在大分子药物递送效率低下的问题。其中一个重要的原因是人体器官的各类黏膜(包括鼻腔黏膜、肺粘膜、阴道黏膜、口腔黏膜、胃肠黏膜等)是各类药物难以跨越的屏障之一。因此,开发跨黏膜载体提高药物的递送效率,实现药物在体内的跨黏膜高效递送具有十分重要的科学意义和巨大的实际应用价值。
黏膜是指衬于生物体(消化、呼吸、泌尿、生殖等)管状或囊状器官管壁腔面的膜状结构。由上皮组织和疏松结缔组织组成,有的器官并含黏膜肌层。其结缔组织部分被称为固有层,其上皮组织部分被称为上皮,内有血管和神经,能分泌黏液。正常黏膜的色泽因血液充盈度不同可由淡红到鲜红色,湿润,有一定伸展性,空虚状态下常形成皱褶。有保护、分泌和吸收等作用,其中上皮又是管状器官进行功能活动的主要部位。上皮类型因所在部位和功能不同而异。根据部位的不同而有不同的名称,如鼻腔黏膜、肺粘膜、阴道黏膜、口腔黏膜、胃肠黏膜等。
尽管近年来,系列亲水性阳离子高分子材料,如聚乙烯亚胺(PEI),聚酰胺-胺树形高分子(dendrimer),壳聚糖(chitosan),β-环糊精,明胶,阳离子多肽/氨基酸,如聚赖氨酸(PLL),阳离子聚酯,阳离子聚磷酸酯,聚乙烯基吡啶盐,聚(二甲氨基)乙基甲基丙烯酸酯,可以削弱上皮细胞的紧密连接,增加上皮的通透性,从而促进药物的黏膜渗透,提高药物的生物利用度。然而,这一传递体系的生物相容性较差,对机体有一定的毒副作用,高浓度下易引起黏膜、上皮损伤,其临床应用面临瓶颈。因此,此类阳离子高分子材料仍需进一步改进,以达到更好的促进药物吸收效果,和更低的毒性。
因此,如何发明一种促进药物吸收效果明显,同时低细胞毒性的新型药物载体材料是具有挑战性的方向。
发明内容
本发明的目的是提供一种促进药物吸收效果明显,低毒性的新型药物载体材料,并且本发明提出的含氟化合物修饰的壳聚糖合成工艺成熟、操作简易,合成效率高,周期短,无需繁琐的纯化步骤即可获得高产率的药物载体,其简易的合成方法为其提供了商业化的良好基础,本发明所述的含氟化合物修饰的壳聚糖具有作为多种药物载体的用途。
本专利提供了如下技术方案,一种作为药物载体用的氟化修饰的壳聚糖衍生物,具有如下结构:含氟化合物共价连接在壳聚糖主链上,
所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,
所述含氟化合物为如下化学式(I)
Figure PCTCN2020138465-appb-000001
所示的含氟脂肪链,或式(Ⅱ)
Figure PCTCN2020138465-appb-000002
所示的芳香环功能基团,所述R1为卤素(氟,氯,溴,碘)、卤素取代的烷烃、环烷烃、醛基、羧基、双键、炔键、羟基、磺酰氯、磺酸键或巯基这些能够与伯氨基反应的活性基团。
进一步的,一种作为药物载体用的氟化修饰的壳聚糖衍生物,具有如式(Ⅳ)所示的含有伯氨基的壳聚糖分子骨架:
Figure PCTCN2020138465-appb-000003
所述壳聚糖的伯氨基与含氟功能基团之间形成的连接基团为:-NH-、-N=C-、-NHCH 2CH(OH)-、-NHCH 2CH(OH)CH 2O-、
Figure PCTCN2020138465-appb-000004
Figure PCTCN2020138465-appb-000005
以及衍生基团;所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,
所述含氟功能基团为含氟脂肪链、芳香环功能基团。
进一步的,所述作为药物载体用的氟化修饰的壳聚糖衍生物,所述式(I)中x为0-3的整数,y为0-20的整数,z为0-8的整数,R2为CF 3、CHF 2、CH 2F、或CH 3(当y不为0);
所述含氟脂肪链化合物是指含氟烃基及其衍生物,包括三氟乙酸、五氟丙酸、七氟丁酸、九氟戊酸、十一氟己酸、十三氟庚酸、十五氟辛酸、十七氟壬酸、十九氟奎酸、全氟丁酸酐、全氟庚酸酐、全氟癸酸酐、2,2,3,3,4,4,4-七氟丁基丙烯酸酯、3-(1H,1H,5H八氟戊氧基)-1,2-环氧丙烯、九氟丁基璜酰胺酐及其衍生物。
进一步的,所述作为药物载体用的氟化修饰的壳聚糖衍生物,所述式(Ⅱ)中R为H,CH 3,OH,NO 2,O,CF 3,F,CH 2OH,CN,NCO,或(CF 2)aCF 3(a为1-20的整数)等,且至少一个R为F;
所述含氟芳香环化合物包括3-氟苯甲酸、3,5-二氟苯甲酸、2,3,5,6-四氟-4-甲基苯甲酸、五氟苯甲酸、2-氟-3-(三氟甲基)苯甲酸及其衍生物。
进一步的,所述的作为药物载体用的氟化修饰的壳聚糖衍生物,所述壳聚糖和含氟化合物共价连接,在所述壳聚糖分子表面进行修饰,构成一种药物载体,其结构如式(V)所示,b、c均为20-500的整数,式(V)如下:
Figure PCTCN2020138465-appb-000006
其中,B为含氟功能基团与壳聚糖伯氨基形成的连接基团,C为含氟脂肪链、芳香环功能基团。
进一步的,所述的作为药物载体用的氟化修饰的壳聚糖衍生物,所述含氟脂肪链是带有可以与氨基反应的活性基团的一类含氟化合物,包括如式(Ⅵ)所示:
Figure PCTCN2020138465-appb-000007
其中A为-COOH、
Figure PCTCN2020138465-appb-000008
能够与伯氨基反应的活性基团,x为0-3的整数,y为0-8的整数。
进一步的,所述的作为药物载体用的氟化修饰的壳聚糖衍生物,所述含氟芳香环化合物是带有可以与氨基反应的活性基团的一类含氟化合物,包括如式(Ⅶ)所示:
Figure PCTCN2020138465-appb-000009
进一步的,所述的作为药物载体用的氟化修饰的壳聚糖衍生物,所述氟化修饰的壳聚糖衍生物作为如下药物的药物载体,所述如下药物为小分子药物、多肽、蛋白药物、不同药物的组合药物以及药物与其它药用辅料的组合药物。
进一步的,含氟化合物修饰的壳聚糖作为药物载体的应用,所述的氟化修饰的壳聚糖衍生物可以作为小分子药物、多肽、蛋白药物、不同药物的组合药物以及药物与其它药用辅料的组合药物的药物载体应用。
进一步的,一种药物复合物,包括所述的作为药物载体用的氟化修饰的壳聚糖衍生物以及药物,所述药物包括小分子药物、多肽、蛋白药物、不同药物的组合药物以及药物与其它药用辅料的组合药物。
进一步的,一种根据所述作为药物载体用的氟化修饰的壳聚糖衍生物制作的透皮给药制剂,包括透皮制剂组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物为氟化壳聚糖,含氟化合物共价连接在壳聚糖主链上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊。
进一步的,一种根据所述的作为药物载体用的氟化修饰的壳聚糖衍生物制作的跨粘膜给药制剂,包括跨粘膜制剂组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物为氟化壳聚糖,含氟化合物共价连接在壳聚糖主链 上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,所述粘膜包括鼻腔黏膜、肺粘膜、阴道黏膜、口腔黏膜、胃肠黏膜。
进一步的,一种根据所述的作为药物载体用的氟化修饰的壳聚糖衍生物制作的穿透眼部屏障给药制剂,包括穿透眼部屏障制剂组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物为氟化壳聚糖,含氟化合物共价连接在壳聚糖主链上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,所述眼部屏障为泪液屏障、角膜/结膜屏障、血房水屏障、血视网膜屏障。
进一步的,一种根据所述的作为药物载体用的氟化修饰的壳聚糖衍生物制作的透皮疫苗载体,包括透皮疫苗载体(a),所述透皮疫苗载体(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物为氟化壳聚糖,含氟化合物共价连接在壳聚糖主链上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,所述透皮疫苗载体有胞内渗透、胞间渗透和毛囊渗透三种抗原透过途径。
进一步的,一种根据所述的作为药物载体用的氟化修饰的壳聚糖衍生物制作的医美及保健品载体,包括医美及保健品载体(a),所述医美及保健品载体(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物为氟化壳聚糖,含氟化合物共价连接在壳聚糖主链上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,所述医美及保健品载体能适用于生发药物和毛发护理药物、美容药物、保健药物。
一种制备氟化修饰的壳聚糖衍生物的方法,其包括如下步骤:制备壳聚糖醋酸水溶液,称取壳聚糖加入醋酸水溶液中,搅拌使其充分溶解,随后滴加氢氧化钠,搅拌至溶液澄清,pH在6.2-6.8;
含氟化合物的活化,称取含氟化合物将其溶于适量无水二甲基亚砜中,依次加入反应量EDC,NHS避光搅拌;
将所述活化好的含氟化合物溶液滴加到快速搅拌的壳聚糖溶液中,避光搅拌充分反应。
所述制备氟化修饰的壳聚糖衍生物的方法,进一步包括如下步骤:将充分反应的溶液缓慢滴加到氢氧化钾乙醇溶液中搅拌,过滤沉淀,用大量无水乙醇冲洗,至滤液呈中性,沉淀经甲醇、乙醚洗涤脱水,真空干燥,干燥后的沉淀物溶于盐酸溶液,冻干得氟化壳聚糖盐酸盐。
如图1-14所示,图1-14为含氟羧酸修饰的壳聚糖膀胱灌注药物载体的合成路线图。
一种制备3-氟苯甲酸氟化壳聚糖的方法,其包括如下步骤:
(1)制备壳聚糖醋酸水溶液:称取充分干燥的壳聚糖加入醋酸水溶液中,搅拌使充分溶解,随后缓慢滴加氢氧化钠,搅拌至溶液澄清,pH在6.2-6.8;
(2)3-氟苯甲酸的活化:称取3-氟苯甲酸,将其溶于适量无水二甲基亚砜中,依次加入反应量EDC,NHS避光充分搅拌;
(3)3-氟苯甲酰壳聚糖的制备:将上述活化好的3-氟苯甲酸溶液分别缓慢滴加到快速搅拌的壳聚糖溶液中,避光搅拌充分反应。
所述制备3-氟苯甲酸氟化壳聚糖的方法,进一步包括如下步骤:
将充分反应的溶液缓慢滴加到氢氧化钾乙醇溶液中充分搅拌,过滤沉淀,用大量无水乙醇冲洗,至滤液呈中性,沉淀经甲醇、乙醚洗涤脱水,真空干燥;
干燥后的沉淀物溶于盐酸溶液,冻干得3-氟苯甲酸氟化壳聚糖盐酸盐分子。
一种制备全氟庚酸氟化壳聚糖的方法,其包括如下步骤:
(1)制备壳聚糖醋酸水溶液:称取充分干燥的壳聚糖加入醋酸水溶液中,搅拌使充分溶解,随后缓慢滴加氢氧化钠,搅拌至溶液澄清,pH在6.2-6.8;
(2)全氟庚酸(13氟庚酸)的活化:称取全氟庚酸,将其溶于适量无水二甲基亚砜中,依次加入适量EDC,NHS避光充分搅拌;
(3)13F庚酸壳聚糖的制备:将上述活化好的全氟酸溶液缓慢滴加到快速搅拌的壳聚糖溶液中,避光搅拌充分反应。
所述制备全氟庚酸氟化壳聚糖的方法,其进一步包括如下步骤:
将充分反应的溶液缓慢滴加到氢氧化钾乙醇溶液中充分搅拌,过滤沉淀,用大量无水乙醇冲洗,至滤液呈中性,沉淀经甲醇、乙醚洗涤脱水,真空干燥,干燥后的沉淀物溶于盐酸溶液,冻干得全氟庚酸氟化壳聚糖盐酸盐。
所述的作为药物载体用的氟化修饰的壳聚糖衍生物:所述氟化修饰的壳聚糖衍生物为全氟庚酸氟化壳聚糖盐酸盐分子,所述全氟庚酸氟化壳聚糖盐酸盐的氟化修饰程度依次为18%~25%,或为20%~22%。
一种药物复合物:包括作为药物载体用的氟化修饰的壳聚糖衍生物以及药物,所述药物包括小分子药物、多肽、蛋白药物、不同药物的组合药物以及药物与其它药用辅料的组合药物。
鉴于,阳离子高分子聚合材料在使用过程中的高细胞毒性最终限制了其临床应用,以及壳聚糖虽然可显著提高灌注药物的生物利用度,但是高浓度的壳聚糖有可能会引起严重的黏膜、上皮损伤,限制了其作为药物载体的临床应用。近年来,发明人尝试了各种技术方案以改善生物安全性,例如在这些阳离子高分子聚合物表面连接了各种修饰基团,如聚乙二醇、环糊精、氨基酸、糖基、氟化修饰等,但几乎没有效果,在偶然的尝试实验中发现氟化修饰后的阳离子高分子材料其生物相容性及大分子转运效果都能有非常显著的改善,并且对核酸、多肽以及蛋白药物具有更强的结合保护作用。
进一步的发明人设计合成了一系列氟化修饰的壳聚糖衍生物,实验结果表明,氟化修饰的壳聚糖(FCS)具有比壳聚糖更显著的促药物渗透吸收的性能,且细胞以及小鼠体内安全评价试验结果表明,FCS具有很好的生物安全性,即使是高浓度的FCS也无明显的细胞毒性及黏膜上皮损伤作用,其生物毒性显著低于没有修饰的壳聚糖(CS)。
发明人通过选用SV-HUC-1人正常膀胱癌商品细胞建立体外膀胱黏膜屏障模型,通过考察FCS对SV-HUC-1单层细胞膜电阻、荧光黄渗透率、细胞紧密连接超微结构以及紧密连接蛋白的影响,简单阐述FCS促药物膀胱黏膜渗透吸收的作用机制。实验结果表明,FCS可显著降低SV-HUC-1单层细胞膜电阻值,增加荧光黄的渗透效率,通过改变紧密连接蛋白以及E-钙黏素蛋白的结构和空间分布调控细胞紧密连接,增加药物分子的细胞旁路摄取效率。也就是说,FCS可以有效提高生物组织屏障中(如粘膜上皮组织等)细胞与细胞之间的间隙,从而使得游离的药物分子或者被FCS载带的药物能够更有效地穿越这些生物组织屏障。
此外,通过大量实验数据表明随着壳聚糖骨架上含氟脂肪链长度以及取代度的增加,修饰产物的促药物渗透吸收能力出现先增加后降低的现象,表明对于壳聚糖的修饰不能过度氟化,氟化壳聚糖的促灌注药物黏膜渗透吸收的作用可能是壳聚糖正电荷分子骨架与含氟脂肪链共同作用的结果。
因此,本专利确定选择FCS作为一种新型的药物载体进行进一步的研究。本专利设计合成了一系列氟化修饰的壳聚糖衍生物,其在制药领域的应用包括但不限于以下疾病模型,例如膀胱癌灌注给药(或其他腔内灌注)、肺部吸入给药、透皮给药、口服给药、眼部给药、疫苗给药、鼻部给药。
膀胱癌是最常见的泌尿肿瘤之一,临床上75%以上的膀胱癌为非肌层浸润型膀胱癌(NMIBC),其中30%~80%的NMIBC患者行经尿道膀胱肿瘤切除术(TURBT)后5年内复发,10%~20%的NMIBC患者进展为肌层浸润型膀胱癌。因此,TURBT后辅助灌注化疗或免疫治疗抑制或延缓肿瘤复发已成为膀胱癌临床治疗指南首选方案。虽然TURBT后辅助化疗药物可延缓肿瘤复发,但由于膀胱的生理特性及其黏膜的生理屏障作用,使得传统方式膀胱灌注的药液在膀胱内滞留时间有限、作用时间短、生物利用度低,不能使时间、浓度依赖性的灌注药物发挥显著抗肿瘤作用,无法有效降低膀胱癌复发和进展绝对风险并有效改善预后。本专利的技术方案基于FCS作为新型跨粘膜药物载体,可改善灌注药物生物利用度,提高药物在腔内灌注后进入膀胱肿瘤内部结构的效率,从而提高膀胱灌注治疗的疗效。该技术方案也可以适用于其他腔内灌注治疗(如腹腔、盆腔、胸腔)。
与静脉给药治疗相比,肺部吸入给药治疗局部地将药物输送到肿瘤组织,所需药物剂量明显降低,毒副作用小。肺部的特殊生理结构决定了肺部吸入给药的特点和优势:肺部的表面积大,毛细血管丰富,而且肺泡上皮细胞层薄,肺部给药起效快;肺部的生物代谢酶分布集中,生物活性低从而减少了对蛋白质的水解,使得蛋白质和多肽容易通过肺泡表面被快速吸收,保持了其生物活性;避免了肝脏首过效应。但仍存在着一些缺点限制了其临床应用,如吸入的药物很快被肺部清除,无法保证药物在肺部的有效沉积。针对肺癌而言,尽管肺部吸入的药物能够到达肺泡,但是进入肺部肿瘤内部的效率一般而言是很低的,严重影响了肺部吸入给药模式用于治疗肺癌的疗效。本专利的技术方案基于FCS作为新型跨粘膜药物载体,可改善肺部吸入药物的生物利用度,提高药物在吸入后进入肺部肿瘤内部结构的效率,从而提高肺部吸入给药治疗的疗效。
透皮给药系统是指在皮肤表面给药,药物以一定速率通过皮肤各层,进入体循环,产生全身或局部治疗作用的制剂。透皮给药作为一种非侵入式的外用给药方式,具有操作简便,患者适应性强等诸多优势。然而,透皮给药通常受到皮肤角质层类酯屏障以及药物理化性质的制约,如何提高药物通过透皮给药模式进入血液循环或者进入皮下病灶(如皮肤癌)的能力是这项技术面临的一个重要挑战。本专利的技术方案基于FCS作为新型跨粘膜药物载体,可改善透皮药物的生物利用度,大幅提高药物穿透皮肤屏障的能力,可以使药物能够更有效地通过透皮给药模式进入血液循环或者进入皮下病灶(如皮肤癌),从而提高透皮给药治疗的疗效。
本发明提出一种复合物包括含氟化合物修饰的壳聚糖以及药物,所述药物包括小分子药物、多肽、蛋白药物、不同药物的组合药物以及药物与其它药用辅料的组合药物,以及其在促进药物吸收中的用途。
例如以表阿霉素(THP)作为膀胱灌注药物,利用本发明制备的含氟化合物修饰的壳聚糖作为药物运输载体,促使药物进入膀胱组织。实验表明本发明具有以下优点:本发明在保持显著促灌注药物膀胱黏膜吸收效率的同时,保持很好的生物相容性。通过小鼠体内灌注实验发现,含氟化合物修饰的壳聚糖改善THP在膀胱黏膜吸收的效率要显著高于THP的水溶液以及其壳聚糖溶液;同时含氟化合物修饰的壳聚糖在较低浓度下实现高效的促药物膀胱黏膜吸收。本发明提出的膀胱灌注药物载体兼具高效、低毒、价格低廉、合成简易等优点。
进一步地,一种采用所述透皮给药制剂的药物组合物,包括透皮制剂组分(a)和药物组分(b),所述组分(b)为糖尿病治疗药物、抗肿瘤药物、免疫调节剂、抗病毒药物、抗炎药物、镇痛麻醉药物。
进一步地,一种采用所述透皮给药制剂的治疗糖尿病的药物复合物,其特征在于:包括氟化壳聚糖与胰岛素,所述氟化壳聚糖与胰岛素的质量比为1:0.25-4。
进一步地,所述的采用透皮给药制剂的治疗糖尿病的药物复合物,其特征在于:所述氟化壳聚糖与胰岛素互相吸附形成复合物,所述复合物粒径范围小于10微米,或所述复合物粒径范围不大于500纳米,所述氟化壳聚糖与胰岛素的质量比为1:0.5-2。
进一步地,一种制备采用所述采用透皮给药方式的治疗糖尿病的药物复合物的方法,其特征在于:
将氟化壳聚糖与胰岛素分别溶于弱酸的溶液环境中使其溶解均匀;
将反应质量比为1:0.25-4的氟化壳聚糖与胰岛素混合均匀,混匀后在搅拌中滴加弱碱溶液,调节pH为6-7,在中性条件下氟化壳聚糖和胰岛素吸附结合在一起,形成稳定的纳米颗粒。
进一步地,所述制备采用透皮给药方式的治疗糖尿病的药物复合物方法,其特征在于:
所述氟化壳聚糖与胰岛素的反应质量比为1:0.25-4或1:0.5-2;
反应充分后取出药物组合物,预加冻干保护剂后冻干,得到氟化壳聚糖-胰岛素冻干粉。
进一步地,一种采用所述透皮给药制剂的治疗糖尿病的药物组合物透皮敷贴,其特征在于:所述氟化壳聚糖与胰岛素互相吸附形成复合物,所述氟化壳聚糖与胰岛素的质量比为1:0.25-4,与水凝胶混合均匀后得到透皮敷贴。
进一步地,一种采用所述透皮给药制剂的治疗黑色素瘤的药物复合物,其特征在于:所述氟化壳聚糖与细胞程式死亡-配体1抗体形成复合物,所述复合物粒径范围小于10微米,或所述复合物粒径范围不大于500纳米,所述氟化壳聚糖与细胞程式死亡-配体1抗体的反应质量比为1:0.25-4。
进一步地,所述的治疗黑色素瘤的药物复合物,其特征在于:所述氟化壳聚糖与细胞程式死亡-配体1抗体的反应质量比为1:0.25-4或1:1,所述氟化壳聚糖-细胞程式死亡-配体1抗体水溶液与凡士林软膏混合形成氟化壳聚糖-细胞程式死亡-配体1抗体透皮软膏。
一种透皮给药制剂,所述制剂包括组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物能作为治疗糖尿病药物、肿瘤疾病药物、消炎药物的透皮给药制剂。
一种透皮给药制剂,所述制剂包括组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物作为透皮给药制剂在医疗美容药物、药物外用制剂、医疗器械外用制剂、化妆护肤品制备中的应用。
常规的透皮贴剂通常需要在贴剂中加入促渗透剂以促进药物透过皮肤,常用的促渗透剂包括乙醇、丁醇等醇类、二甲亚砜、月桂氮卓酮类、吡咯酮类衍生物、表面活性剂和脂肪酸类化合物等。其中,醇类、吡咯酮类衍生物的促渗透机制主要是通过溶胀角质层中的类脂,增加药物的溶解度;月桂氮卓酮类通过改变脂质双分子层的致密性,增加脂质的流动性来促进药物透皮;脂肪酸类化合物通过其不饱和疏水链的顺式结构插入脂质双分子层的疏水结构时,脂质双分子层产生扭转,形成了一个很微细的孔道,使药物得以扩散进入。物理促渗透法主要有离子电渗法、超声波法、电致孔法以及微针法。物理促渗透法主要用于化学促渗透剂难以奏效的药物,如多肽、蛋白质等大分子药物和离子型药物。离子电渗法是通过在皮肤表面施加适当的电场来增加药物的透皮率,由于电场的存在,离子在电场中的相互作用,溶剂在电场下的对流运动以及电流引起的皮肤渗透性增加都会促进药物透皮吸收。超声波法是在超声作用下促进药物分子进入皮肤,其可能的机制有:1.局部热效应导致药物渗透性增加;2.局部辐射压作用使药物沿声波方向移动,促进药物渗透;3.局部声微流作用使药物能通过毛囊和汗腺进入皮肤;4.空化作用造成皮肤角质层无序排列,促进药物渗透。电致孔法则是通过脉冲电场来促进药物透皮吸收。目前报道的电致孔法的透皮机制为,在脉冲电场下,皮肤中类脂分子重新有序的排列从而形成新的通道以促进药物渗透,在脉冲电场结束后,类脂分子恢复之前的无序排列,从而关闭通道。微针则是利用微米级别微针,在皮肤上形成极小的创口,来高效地促进药物透皮。
本专利所述含氟化合物修饰的阳离子聚合物,尤其是氟化壳聚糖可以增加药物在皮肤中的渗透能力,同时减少给药面积,从而降低药物可能对正常皮肤产生的毒副作用。作为一种化学促渗透聚合物,与其他化学促渗剂相比,本专利所述技术方案作为水溶液不具有强挥发性和刺激性,作用速度快,持续时间长,且能够促进一系列药物分子透过皮肤,包括小分子药物,多肽、蛋白类大分子药物,离子型药物等。与物理促渗透法相比,本专利技术方案不需要外加电场,超声等,大幅降低给患者带来的创伤、痛苦、不便与安全隐患。因此,本专利实施例所披露的氟化壳聚糖均可以作为透皮制剂,以及和其他药物联合使用,同时,含氟化合物修饰的阳离子聚合物由于均经过了含氟化合物的修饰,因此也可以作为透皮制剂使用,以及与其他药物联合使用。
本专利提供的氟化壳聚糖药物载体,具有促进药物吸收效果明显,低毒性等优点,并且本发明提出的含氟化合物修饰的壳聚糖合成工艺成熟、操作简易,合成效率高,周期短,无需繁琐的纯化步骤即可获得高产率的药物载体,其简易的合成方法为其提供了商业化的良好基础,本发明所述的含氟化合物修饰的壳聚糖具有作为多种药物载体的用途,能够有效的提升治疗效果,具有广泛的用途,并且成本较低。
进一步的,本专利所述技术方案所产生的透皮效应是临时性的,在除去药物后,皮肤角质层细胞即会关闭通道,保护人体安全。皮肤由表皮和真皮组成,其中表皮由浅入深依次为角质层、透明层、颗粒层和生发层。真皮由致密结缔组织构成,由浅入深依次为乳头层和网状层。乳头层与表皮的生发层相连,其中有丰富的毛细血管、淋巴管、神经末梢和触觉小体等感受器。角质层是透皮给药的最大的限流屏障,大部分皮肤的角质层由5-25层扁平 角质细胞组成,这些细胞无细胞核,细胞器,细胞膜较厚,无生命,不透水,具有防止组织液外流,抗磨擦和防感染等功能。含氟化合物修饰的阳离子聚合物可以通过刺激这些细胞的紧密连接蛋白分布改变,降低细胞间紧密连接,并进一步刺激肌动蛋白磷酸化,从而促进细胞旁运,打开细胞间隙,形成通道,并携带药物透过角质层,进一步深入皮肤,而后进入真皮层,进入皮肤毛细血管与淋巴循环并发挥药物作用(见图1)。
本专利的技术方案可以用于口服给药系统,具体如下。
肠道粘膜:
小肠是药物吸收的主要部位,药物从小肠吸收进入血液循环之前,必然会面临三种主要的胃肠道生理屏障,如图3-1所示,包括蛋白酶屏障,黏液屏障和肠上皮细胞屏障。小肠中存在着丰富的蛋白酶,包括胰蛋白酶,糜蛋白酶,弹性蛋白酶和羧肽酶等。蛋白多肽类药物会在各种蛋白酶的作用下快速降解失活。肠道中还存在粘液屏障,肠道上皮细胞表面覆盖着一层厚度不一的粘液,其中由95%的水及5%的电解质,脂质,蛋白质和糖蛋白组成。粘液层具有粘弹性,起到保护作用,同时粘液屏障也是影响蛋白多肽类药物口服吸收的主要屏障之一。除了粘液屏障,肠道中还存在肠上皮细胞屏障。肠道上皮细胞层主要包括两类细胞:可以吸收物质的上皮细胞(肠上皮细胞)和难以吸收物质的上皮细胞。这些细胞通过紧密连接相互联系,形成一个相对不渗透的,严重限制外来物质吸收的屏障。
以氟化壳聚糖为载体,不仅可以解决如上问题,还可以增加药物的渗透,使药物高效到达血液或治疗部位,提升药物的利用度。
接下来,以胰岛素和细胞程式死亡-配体1抗体为示例药物,氟化壳聚糖解释透粘膜机理。
如实例3-1所述,氟化壳聚糖作为药物载体递送胰岛素时可以在不破坏肠道上皮细胞分泌紧密连接蛋白功能的情况下,打开上皮通道,如图3-2所示。
以氟化壳聚糖为主体,可以制成相应的口服药物如溶液剂、糖浆剂、颗粒剂、胶囊剂、散剂、丸剂、片剂。药物经口服后被胃肠道吸收入血,通过血液循环到达局部或全身组织,达到治疗疾病的目的。但蛋白质多肽类药物容易被胃肠道内的消化酶所降解,同时分子量较大、分子之前具有很强的聚合趋势,很难透过体内屏障发挥足够的作用,若不经过处理直接口服吸收,其生物利用度仅为0.5%。以氟化壳聚糖为载体,不仅可以保持蛋白质多肽类药物的活性,稳定其构型;减少多聚体的形成,便于增加肠黏膜处的吸收;减轻消化酶对于蛋白质多肽类药物的降解作用,同时还可以在不破坏肠道上皮细胞分泌紧密连接蛋白功能的情况下,打开上皮通道,可以增加药物在小肠部位的渗透,便于药物高效到达血液或治疗部位,提升药物的利用度。接下来,以胰岛素和细胞程式死亡-配体1抗体为示例药物,实现含氟化合物修饰的阳离子聚合物作为药物载体,在口服给药方面的应用。
所述药物可以是糖尿病药物、抗结肠炎药物、麻醉药物、抗炎药物、抗菌药物、抗病毒药物、抗寄生虫药物等。
所述糖尿病药物可以是磺酰脲类包括但不限于:氨苯磺丁脲、甲苯磺丁脲、氯磺丙脲、乙酸已脲、格列齐特、格列吡啶、格列美脲等药物及其衍生物。所述糖尿病药物可以是非磺酰脲类包括但不限于:瑞格列奈、那格列奈等药物及其衍生物。所述糖尿病药物可以是噻唑烷二酮类包括但不限于:罗格列酮、吡咯列酮等药物及其衍生物。所述糖尿病药物可以是二胍类包括但不限于:苯乙双胍、二甲双胍等药物及其衍生物。所述糖尿病药物可以是α-葡 萄糖苷酶抑制剂包括但不限于:阿卡波糖、伏格列波糖、米格列醇等药物及其衍生物。所述糖尿病药物可以是二肽基肽酶-Ⅳ包括但不限于:胰高血糖素样肽,DPP-Ⅳ抑制剂西格列汀、维格列汀、沙格列汀。在本发明的一个实施方案中,抗糖尿病药物包括但不限于胰岛素。
如实例3-1所述,氟化壳聚糖可以作为药物载体递送胰岛素,以口服药物的方式进行给药,用于糖尿病治疗。随着世界范围内糖尿病病例的增加以及患者对使用可注射胰岛素的血糖管理缺乏依从性,迫切需要开发有效的口服胰岛素制剂。然而,胃肠道对生物制剂的口服递送提出了巨大的障碍。本发明可以解决胰岛素口服利用率低的问题。口服给药是最容易被患者接受的给药途径。然而胰岛素容易被胃肠道的各种酶降解,而且胰岛素自身的分子量较大,难以被胃肠道上皮细胞膜吸收。通过氟化壳聚糖药物载体以口服方式进行胰岛素给药,不仅可以保护胰岛素的活性,还可以促进胰岛素穿过小肠黏膜,提升药物的利用度。
任何疾病或机能失调,特别是机体损伤,都可能使人产生疼痛的感觉,很多情况下需要进行镇痛治疗。镇痛药是一类作用于中枢神经系统,对痛觉中枢产生选择性抑制作用,使疼痛减轻或消除而不影响其他感觉的药物,临床上主要用于如创伤,烧伤,手术后和癌症等引起的剧烈锐疼的止疼。但是许多止疼药不能快速止痛,和局部给药,本发明能解决该问题。
所述镇痛药可以是吗啡及其衍生物包括但不限于:可待因,乙基吗啡,苄基吗啡,异可待因,海洛因,苯乙基吗啡,氢吗啡酮,羟吗啡酮,丙烯吗啡,纳不啡,纳洛酮,钠曲酮等药物及其衍生物。
所述镇痛药可以是合成镇痛药包括但不限于:哌替啶,阿尼利定,苯哌利定,匹米诺定,阿法罗定,倍他罗定,芬太尼,阿芬太尼,舒芬太尼,瑞芬太尼,盐酸哌替啶,美沙酮,右吗拉胺,右丙氧芬,盐酸美沙酮,N-甲基吗啡喃,左啡诺,布托啡诺,吗啡喃,喷他佐辛,地佐辛,舒马普坦,曲马多,苯噻啶,奈福泮。
麻醉药是指能使患者整个机体或机体局部暂时,可逆性失去知觉及痛觉的药物。但是麻醉药物大部分为静脉注射和脊髓内注射,这两种给药方式相对于口服给药增加了患者的疼痛。本发明可以通过口服麻醉药来增加患者依从性。
所述麻醉药可以是静脉注射麻醉药包括但不限于:盐酸氯胺酮,丙泊酚,硫喷妥钠,依托咪脂,咪达唑仑和γ-羟基丁酸钠。
所述麻醉药可以是局部麻醉药包括但不限于:芳酸酯类,芳酰胺类,氨基酮类,氨基醚类,氨基甲酸酯类,羟普鲁卡因,氯普鲁卡因,丁卡因,布他卡因,硫卡因,普鲁卡因胺,布比卡因,阿替卡因,依替卡因,罗哌卡因,甲哌卡因,盐酸克罗宁及等药物及其衍生物。
炎症与诸多重大疾病如心脑血管疾病和神经退行性疾病甚至肿瘤的发生、发展密切相关,因此对炎症的早期成像、诊断与抗炎策略是防治许多疾病的重要手段。现阶段,抗炎药物大部分通过静脉注射给药和口服给药。但是静脉注射抗炎药会有很严重的副反应,口服抗炎药的生物利用率极低。本发明能将提高抗炎药的口服利用率,使患者更加依从。
所述抗炎药可以是非甾体类抗炎药包括但不限于:阿司匹林、对乙酰氨基酚、非特异性环氧化酶抑制剂、安替比林、安乃近、保泰松、羟布宗、甲芬那酸、吲哚美欣、舒林酸、双氯芬酸钠、布洛芬、普奈生、吡罗昔康、美洛昔康等非特异性环氧化酶抑制剂药物及其衍生物。
所述抗炎药可以是甾体类抗炎药包括但不限于:氢化可的松、皮质酮、醛固酮、曲安西龙、泼尼松龙、乙酸地塞米松、醋丙甲泼尼松等药物及其衍生物。
本发明的实施方案可以用于治疗各类炎症,包括但不限于:心肌炎,关节炎,结肠炎,扁桃体炎,有胃炎,鼻炎,牙周炎,肠道炎,咽喉炎,前列腺炎,阴道炎,宫颈炎,肩周炎,颈椎炎,滑囊炎,皮肤炎,结膜炎,中耳炎等。
结肠粘膜:
结肠是构成消化系统的重要组成部分,主要参与水分,维生素和无机盐的吸收,也是形成粪便的重要场所。结肠肠壁从外到内可以分为浆膜层,纵肌层,环肌层,粘膜下层和粘膜层,粘膜层作为人体第一道防线,可以保护肠道免受肠腔有害物质如细菌和毒素的入侵。正常的结肠粘膜屏障由机械屏障,化学屏障,免疫屏障和生物屏障组成,其中其中,机械屏障包括结肠上皮细胞以及上皮细胞间的连接,是粘膜屏障的重要组成部分;化学屏障主要包括粘膜上皮分泌的黏液、消化液和肠道原生菌群产生的抑菌类物质;免疫屏障由肠粘膜的淋巴组织和肠道分泌的抗体组成,淋巴组织可以通过细胞免疫和体液免疫,保护肠道免受一些致病性抗原的入侵;生物屏障即肠道正常的共生菌群,肠道共生菌群与宿主之间形成一个相互依赖的微生态系统。在正常情况下,肠道菌群可以形成一个微生物屏障,保护机体免受外来微生物的影响。结肠上皮细胞屏障是肠道机械屏障的重要组成部分,由结肠上皮细胞和上皮细胞间的细胞连接共同构成,这些细胞间连接包括紧密连接、缝隙连接和粘附连接等,它们共同作用,封闭细胞间隙,其中紧密连接的作用至关重要,由连接粘附分子、咬合蛋白(Occludin)、闭合蛋白(Claudin)、ZO-1等组成。细胞连接可以阻止肠腔的内毒素等大分子物质进入肠组织。
由于存在上述屏障,对于结肠部位患病的患者,药物很难渗透到结肠。以氟化壳聚糖为载体,可以有效克服结肠相关的机械屏障,化学屏障,免疫屏障和生物屏障组成,提高药物在结肠中的渗透能力,提升药物的利用度。相对于静脉注射或者腹腔注射,会有更多的药物渗透到患病部位。
如实例3-2所述,氟化壳聚糖作为药物载体递送胰岛素时可以在不破坏肠道上皮细胞分泌紧密连接蛋白功能的情况下,打开上皮通道,如图3-2所示。
如实例3-2所述,氟化壳聚糖可以作为药物载体递送抗肿瘤药物如免疫检查点阻断抗体,以口服药物的方式进行给药,用于肿瘤治疗。结肠癌是最常见的消化道肿瘤之一,其发病率和死亡率都很高。尽管细胞程式死亡-配体1抗体在结直肠癌症治疗中取得成功,但是细胞程式死亡-配体1抗体都是通过静脉注射给药,这可能引起严重的不良反应,比如肠胃道毒性。此外由于全身给药,药物在结直肠部富集较少。口服给药作为最容易被患者接受的给药方式,能把大部分药物递送到患病部位,并且降低不良反应。然而细胞程式死亡-配体1抗体容易因消化道内严酷的环境而失活,而且细胞程式死亡-配体1抗体分子量较大,难以跨过肠道上皮细胞。通过氟化壳聚糖药物载体以口服方式进行细胞程式死亡-配体1抗体给药,可以使细胞程式死亡-配体1抗体穿过结直肠黏膜,提升药物的利用度。
炎症性肠病(IBD)是一种慢性特发性炎症,可影响整个胃肠道(GI),并增加了结直肠癌的风险。IBD由两种主要的临床定义形式组成,克罗恩病(CD)和溃疡性结肠炎(UC)。CD通常累及回肠和结肠,但它可以不连续地影响胃肠道的任何区域,并且炎症通常是透壁的。相反,UC局限于大肠,从直肠向近端延伸,可能以不间断的方式累及整个结肠,并伴有广泛 的浅表粘膜溃疡。CD和UC都与高发病率和生活质量下降。IBD的发病率在全球范围内正在增加,给公共医疗保健带来沉重负担。口服给药作为最容易被患者接受的给药方式,能把大部分抗结肠炎药物递送到患病部位,并且减少肛塞带来的不便。
所述抗结肠炎药物可以是小分子药物包括但不限于:布地奈德、倍氯米松、5-氨基水杨酸、超氧化物歧化酶、4-氨基tempol、过氧化氢酶、甲氨蝶呤、咯利普兰、阿霉素、万古霉素、硫酸共利斯汀、苏罗霉素、拉莫普林、LFF-571、黄连素、胆红素、没食子酸、儿茶酚及其衍生物。
所述抗结肠炎药物可以是蛋白类药物包括但不限于:白血病抑制因子、转化生长因子β、卵清蛋白及其衍生物。
所述抗结肠炎药物可以是小干扰核糖核酸(siRNA)和反义寡核苷酸包括但不限于:针对肿瘤坏死因子-α(TNF-α)的反义寡核苷酸、阿昔单抗-TNF信使核糖核酸-15(miRNA-155)抑制剂、肿瘤坏死因子-αsiRNA、CD98siRNA、细胞周期蛋白D1siRNA、脯氨酰羟化酶2siRNA、Map4k4siRNA、切割寡核苷酸、产生IL10的质粒、NF-kB诱饵寡核苷酸、pcDNA3-EGFP、肽抗原(PeptAg)及其衍生物。
所述抗结肠炎药物可以是益生菌包括但不限于:干酪乳杆菌(ATCC 39392)、乳酸乳球菌、卵形芽孢杆菌、霍乱弧菌、益生菌VSL-3或及其分离的DNA、大肠杆菌。
结直肠癌是全世界最普遍的恶性肿瘤之一。大肠癌的治疗主要依靠手术和化学疗法以及免疫疗法。当前,化疗和免疫疗法通常通过静脉注射给药。尽管口服给药在不同途径中具有最高的患者接受度和依从性,但在口服制剂中开发的临床使用的化学疗法产品很少见。这归因于胃肠道(GI)药物吸收有限且不稳定,以及肝脏对药物代谢的首过效应,与口服制剂的治疗性抗肿瘤作用降低密切相关。此外,由于在肿瘤部位的停留时间短且不稳定,癌细胞对小分子药物的吸收较差,口服制剂对全身大肠癌的全身吸收和转运作用可忽略不计,目前尚无临床可用的化学疗法。本发明可以增加药物的口服利用度。
所述抗结直肠癌药物可以是生物烷化剂包括但不限于:盐酸氮芥、本丁酸氮芥、美法仑、泼尼莫司汀、环磷酰胺、塞替派、卡莫司汀、百消安、顺铂、卡铂等药物及其衍生物。所述抗结直肠癌药物可以是抗代谢药物包括但不限于:氟尿嘧啶、阿糖胞苷、巯嘌呤、甲氨蝶呤等药物及其衍生物。所述抗结直肠癌药物可以是抗肿瘤抗生素包括但不限于:放线菌素D、多柔比星(阿霉素)、佐柔比星、米托蒽醌等药物及其衍生物。所述抗结直肠癌药物可以是中药有效成分包括但不限于:10-羟基喜树碱、硫酸长春碱、紫杉醇、多西紫杉醇等中药天然药物有效成分以及其衍生物。所述抗结直肠癌药物可以是免疫点检查抑制剂参见表3-1所述抗结直肠癌药物可以是细胞因子包括但不限于:细胞因子是由免疫细胞(如单核、巨噬细胞、T细胞、B细胞、NK细胞等)和某些非免疫细胞(内皮细胞、表皮细胞、纤维母细胞等)经刺激而合成、分泌的一类具有广泛生物学活性的小分子蛋白质。细胞因子包括但不限于白细胞介素(interleukin,IL)、干扰素(interferon,IFN)、肿瘤坏死因子超家族(tumor necrosis factor,TNF)、集落刺激因子(colony stimulating factor,CSF)、趋化因子(chemokine family)、生长因子(growth factor,GF)、转化生长因子-β家族(transforming growth factor-βfamily,TGF-βfamily)。白细胞介素包括但不限于IL-1―IL-38。集落刺激因子包括但不限于G(粒细胞)-CSF、M(巨噬细胞)-CSF、GM(粒细胞、巨噬细胞)-CSF、Multi(多重)-CSF(IL-3)、SCF、EPO等。干扰素包括但不限于包括但不限于IFN-α、IFN-β和IFN- γ。肿瘤坏死因子包括但不限于TNF-α和TNF-β。转化生长因子-β家族包括但不限于TGF-β1、TGF-β2、TGF-β3、TGFβ1β2以及骨形成蛋白(BMP)。生长因子包括但不限于表皮生长因子(EGF)、血小板衍生的生长因子(PDGF)、成纤维细胞生长因子(FGF)、肝细胞生长因子(HGF)、胰岛素样生长因子-I(IGF-1)、IGF-Ⅱ、白血病抑制因子(LIF)、神经生长因子(NGF)、抑瘤素M(OSM)、血小板衍生的内皮细胞生长因子(PDECGF)、转化生长因子-α(TGF-α)、血管内皮细胞生长因子(VEGF)。趋化因子家族包括但不限于四个亚族:(1)C-X-C/α亚族,主要趋化中性粒细胞,主要的成员有IL-8、黑素瘤细胞生长刺激活性(GRO/MGSA)、血小板因子-4(PF-4)、血小板碱性蛋白、蛋白水解来源的产物CTAP-III和β-thromboglobulin、炎症蛋白10(IP-10)、ENA-78;(2)C-C/β亚族,主要趋化单核细胞,这个亚族的成员包括巨噬细胞炎症蛋白1α(MIP-1α)、MIP-1β、RANTES、单核细胞趋化蛋白-1(MCP-1/MCAF)、MCP-2、MCP-3和I-309。(3)C型亚家族的代表有淋巴细胞趋化蛋白。(4)CX3C亚家族,Fractalkine是CX3C型趋化因子,对单核-巨噬细胞、T细胞及NK细胞有趋化作用。细胞因子包括但不限于用于治疗癌症的细胞因子和减轻癌症治疗副作用的细胞因子,它们在人体正常的免疫反应以及免疫系统对癌症的反应能力中起重要作用。用于治疗癌症的细胞因子包括但不限于干扰素、白介素。细胞因子还可以是造血生长因子,通过促进受化疗破坏的血细胞生长来减少癌症治疗的副作用。减少癌症治疗副作用的细胞因子包括但不限于促红细胞生成素、IL-11、粒细胞-巨噬细胞集落刺激因子(GM-CSF)和粒细胞-集落刺激因子(G-CSF)。卡介苗(BCG Vaccine)是由减毒牛型结核杆菌悬浮液制成的活菌苗,可以增强巨噬细胞活性,增强机体细胞免疫的功能,可以用于治疗膀胱癌等。免疫调节药物包括但不限于沙利度胺
Figure PCTCN2020138465-appb-000010
,来那度胺
Figure PCTCN2020138465-appb-000011
泊马利度
Figure PCTCN2020138465-appb-000012
咪喹莫特
Figure PCTCN2020138465-appb-000013
口腔粘膜
口腔粘膜给药系统与传统给药系统相比较有很多优势:1,可保持持久、恒定和可控的血药浓度,从而减轻不良反应;2,避免肝脏首过效应和胃肠道因素的干扰与降解,提高药物生物利用度;3,减轻注射用药的痛苦,提高患者的依从性;4,给药后出现问题时可及时停药,使用方便;5,减少给药次数和剂量,降低药物不良反应的发生率。因此,口腔粘膜给药系统受到了越来越多的关注。
然而由于受口腔可吸收黏膜的空间限制,药物释放系统不能体积过大;不自主的唾液分泌以及吞咽影响口腔黏膜途径的效能;药物的味觉刺激和异物感影响该途径的依从性;不是所有的物质都能通过口腔粘膜,其吸收受脂溶性、pH值、分子量等的影响。
口腔粘膜是药物通过口腔转运过程中的屏障,口腔不同区域膜的性质不同,角质层与非角质层组织的厚度及组成也不尽相同,另外,吸收部位膜的厚度、血流量、血液或淋巴灌流、细胞更新、酶含量等都与药物口腔粘膜吸收量有关。口腔粘膜屏障由上皮屏障,基底膜屏障,固有层组成。口腔粘膜上皮内存在一种上皮屏障,上皮层为药物透过粘膜的主要屏障,角化上皮大于非角化上皮的通透性,主要的渗透屏障存在于最外层上皮中1/4处至1/3至处,上皮厚度对渗透率也有很大的影响,角质层是最主要的吸收屏障。虽然口腔上皮细胞的浅表部分能阻碍外部物质进入,但位于上皮细胞和固有层连接处的基底膜也可以限制药物的透膜。药物的亲脂性不宜太强,药物在唾液中难以溶解,达不到有效水平,且难以通过基底膜,影响药物的透膜吸收。一般认为固有层不起渗透屏障作用。血管和神经在固有层之中,为上皮提供营养和神经支配。然而有些研究者认为固有层部分也会构成一定的渗透 屏障。以氟化壳聚糖为载体,可以有效克服口腔粘膜相关的上皮屏障、角质层屏障、渗透屏障等,有效增加药物的透过率,便于药物真正到达血液或治疗部位,提升药物的利用度。
口腔黏膜给药入血后部分治疗具有与鼻黏膜给药相似的作用。制剂包括漱口剂、含片、舌下片剂、膜剂等剂型。如可以治疗疱疹性三叉/舌咽神经疼痛等疾病、炎症、细菌、病毒导致的咽喉炎、口腔溃疡、局部麻醉等。此外,舌下静脉丛较多,穿透黏膜后能迅速入血,避免肝脏首过效应,多用于心脏相关疾病。心脏疾病相关药物可以包括但不限于下表3-2具体药物及其衍生物。
表3-2为心脏疾病相关药物
Figure PCTCN2020138465-appb-000014
Figure PCTCN2020138465-appb-000015
任何疾病或机能失调,特别是机体损伤,都可能使人产生疼痛的感觉,很多情况下需要进行镇痛治疗。镇痛药是一类作用于中枢神经系统,对痛觉中枢产生选择性抑制作用,使疼痛减轻或消除而不影响其他感觉的药物,临床上主要用于如创伤,烧伤,手术后和癌症等引起的剧烈锐疼的止疼。但是许多止疼药不能快速止痛,和局部给药,本发明能解决该问题。
所述镇痛药可以是吗啡及其衍生物包括但不限于:可待因,乙基吗啡,苄基吗啡,异可待因,海洛因,苯乙基吗啡,氢吗啡酮,羟吗啡酮,丙烯吗啡,纳不啡,纳洛酮,钠曲酮等药物及其衍生物。
所述镇痛药可以是合成镇痛药包括但不限于:哌替啶,阿尼利定,苯哌利定,匹米诺定,阿法罗定,倍他罗定,芬太尼,阿芬太尼,舒芬太尼,瑞芬太尼,盐酸哌替啶,美沙酮,右吗拉胺,右丙氧芬,盐酸美沙酮,N-甲基吗啡喃,左啡诺,布托啡诺,吗啡喃,喷他佐辛,地佐辛,舒马普坦,曲马多,苯噻啶,奈福泮。
麻醉药是指能使患者整个机体或机体局部暂时,可逆性失去知觉及痛觉的药物。但是麻醉药物大部分为静脉注射和脊髓内注射,这两种给药方式相对于口服给药增加了患者的疼痛。本发明可以通过口服麻醉药来增加患者依从性。
所述麻醉药可以是静脉注射麻醉药包括但不限于:盐酸氯胺酮,丙泊酚,硫喷妥钠,依托咪脂,咪达唑仑和γ-羟基丁酸钠。
所述麻醉药可以是局部麻醉药包括但不限于:芳酸酯类,芳酰胺类,氨基酮类,氨 基醚类,氨基甲酸酯类,羟普鲁卡因,氯普鲁卡因,丁卡因,布他卡因,硫卡因,普鲁卡因胺,布比卡因,阿替卡因,依替卡因,罗哌卡因,甲哌卡因,盐酸克罗宁及等药物及其衍生物。
炎症与诸多重大疾病如心脑血管疾病和神经退行性疾病甚至肿瘤的发生、发展密切相关,因此对炎症的早期成像、诊断与抗炎策略是防治许多疾病的重要手段。现阶段,抗炎药物大部分通过静脉注射给药和口服给药。但是静脉注射抗炎药会有很严重的副反应,口服抗炎药的生物利用率极低。本发明能将提高抗炎药的口服利用率,使患者更加依从。
所述抗炎药可以是非甾体类抗炎药包括但不限于:阿司匹林、对乙酰氨基酚、非特异性环氧化酶抑制剂、安替比林、安乃近、保泰松、羟布宗、甲芬那酸、吲哚美欣、舒林酸、双氯芬酸钠、布洛芬、普奈生、吡罗昔康、美洛昔康等非特异性环氧化酶抑制剂药物及其衍生物。
所述抗炎药可以是甾体类抗炎药包括但不限于:氢化可的松、皮质酮、醛固酮、曲安西龙、泼尼松龙、乙酸地塞米松、醋丙甲泼尼松等药物及其衍生物。
本发明的实施方案可以用于治疗各类炎症,包括但不限于:心肌炎,关节炎,结肠炎,扁桃体炎,有胃炎,鼻炎,牙周炎,肠道炎,咽喉炎,前列腺炎,阴道炎,宫颈炎,肩周炎,颈椎炎,滑囊炎,皮肤炎,结膜炎,中耳炎等。
同样口服递送疫苗即可以包被保护外壳递送抗原至肠道与肠道相关淋巴样组织结合,被肠道抗原呈递细胞捕获识别并迁移至派伊尔氏淋巴结,引发免疫应答,也可以直接含在口中进行口部疫苗递送。口服疫苗包括但不限于:脊髓灰质减毒活疫苗、轮状病毒疫苗、伤寒疫苗、痢疾活疫苗、肠伤寒菌疫苗,百日咳疫苗,狂犬病疫苗、白喉类毒素疫苗,破伤风类毒素疫苗、B链的霍乱毒素疫苗,E.coli肠毒素疫苗,3-脱氧酰化单磷酰基脂质A疫苗、卡介菌多糖核酸疫苗、细菌细胞壁骨架疫苗。
本专利技术方案可以用于吸入(肺部吸入剂/雾化剂)给药系统,具体如下。
以氟化壳聚糖为主体,结合药物还可以制成相应的雾化制剂/吸入制剂,促进药物的跨肺黏膜给药。
静脉注射,虽然作为一种常规的治疗途径,仍然很难将药物有效递送到肺部组织。静脉注射中的药物无法有效沉积在肺部,也很容易引起其他部位的副作用。肺泡的吸收面积大,肺泡上皮细胞的通透性高;通过雾化制剂的方式直接进行肺部给药,可以提高肺组织局部的药物浓度,减少药物损失,精确量化药物用量,减少毒副作用。然而,分子量大、亲水的、带负电荷的大分子,如蛋白质和核酸,仍然很难穿透肺部屏障。一些带负电的亲水大分子很容易被核酸酶降解,难以穿透生物膜到达作用部位。药物在肺部的滞留时间短、生物利用度低,无法有效控制肺部疾病的进展并改善预后。本专利的技术方案以全氟庚酸修饰的壳聚糖为例作为新型跨肺黏膜载体,全氟庚酸修饰的壳聚糖可以有效保护蛋白类药物的活性,提高药物在肺部的滞留时间,有效改善肺部药雾的生物利用度,从而提高肺部给药的治疗效果。雾化制剂包括但不限于气雾剂,吸入粉雾剂和喷雾剂。吸入制剂包括以蒸气或气溶胶形式递送至肺部的液体(如吸入气雾剂和雾化吸入溶液)或固体制剂(如吸入粉雾剂)。所用药物包括但不限于局部治疗药、抗生素药、抗病毒药、抗肿瘤药、蛋白质多肽药等。
雾化制剂中所用药物可以是消炎药,包括但不限于非甾体类药物和甾体类药物。非甾体类药物包括但不限于阿司匹林,对乙酰氨基酚,双氯芬酸,消炎痛(吲哚美辛),布洛芬,芬布芬等药物及其衍生物的各类剂型。甾体类药物包括但不限于肾上腺皮质激素类药 物,即糖皮质激素等药物及其衍生物的各类剂型。
雾化制剂中所用药物可以是抗生素,抗生素包括但不限于β-内酰胺类、氨基糖苷类、大环内酯类、多肽类、林可酰胺类、四环素类、酰胺醇类/氯霉素类、利福霉素类及其他抗生素。β-内酰胺类包括但不限于青霉素类药物(青霉素G、普鲁卡因青霉素、青霉素V、苄星青霉素、苯唑西林、氯唑西林、氟氯西林、阿莫西林、氨苄西林、哌拉西林、美洛西林、阿洛西林、阿莫西林克拉维酸钾、哌拉西林钠他唑巴坦、氨苄西林钠舒巴坦),头孢菌素类(头孢噻吩、头孢唑林、头孢硫脒、头孢氨苄、头孢羟氨苄、头孢拉定、头孢呋辛、头孢孟多、头孢替安、头孢尼西、头孢呋辛酯、头孢克洛、头孢丙烯、头孢噻肟、头孢他定、头孢哌酮、头孢唑肟、头孢曲松、头孢克肟、头孢地嗪、头孢甲肟、头孢特仑、头孢布烯、头孢地尼、头孢匹胺、头孢匹罗、头孢吡肟、头孢哌酮钠舒巴坦钠)药物及其衍生物的各类剂型。非典型β-内酰胺类(头霉素类如头孢西丁、头孢美唑、头孢替坦、头孢米诺、头孢拉宗,单环β-内酰胺类如氨曲南、卡芦莫南,碳青霉烯类如亚胺培南、美罗培南、帕尼培南、法罗培南、厄他培南、比阿培南、多尼培南,氧头孢烯类如拉氧头孢、氟氧头孢)药物及其衍生物的各类剂型。氨基糖苷类包括但不限于依替米星、链霉素、庆大霉素、卡那霉素、阿米卡星、妥布霉素、奈替米星、大观霉素、异帕米星、新霉素、巴龙霉素、春雷霉素、小诺霉素、西索米星、核糖霉素及其衍生物的各类剂型。大环内酯类包括但不限于红霉素、阿奇霉素、克拉霉素、罗红霉素、泰利霉素、地红霉素、麦迪霉素、乙酰麦迪霉素、吉他霉素、乙酰吉他霉素、乙酰螺旋霉素、螺旋霉素、竹桃霉素、泰乐霉素、交沙霉素、蔷薇霉素、琥乙红霉素、替米考星、北里霉素及其衍生物的各类剂型。多肽类包括但不限于万古霉素、去甲万古霉素、替考拉宁、博来霉素、多粘菌素B、多粘菌素E、杆菌肽及其衍生物的各类剂型。林可酰胺类包括但不限于林可霉素、克林霉素及其衍生物的各类剂型。四环素类包括但不限于多西环素、米诺环素、四环素、金霉素、土霉素、地美环素、强力霉素、替加环素及其衍生物的各类剂型。酰胺醇类/氯霉素类包括但不限于氯霉素、甲砜霉素、棕榈氯霉素、琥珀氯霉素、氟苯尼考及其衍生物的各类剂型。利福霉素类包括但不限于利福平、利福喷汀、利福布汀、利福霉素钠及其衍生物的各类剂型。其它抗生素如磷霉素、制霉菌素及其衍生物的各类剂型。
雾化制剂所用药物可以是抗病毒药,包括但不限于磺胺类药物及抗菌增效剂、喹诺酮类抗菌药、抗结核病药物、抗真菌药物、抗病毒药物、抗艾滋病药物及抗寄生虫药物。磺胺类药物包括但不限于磺胺甲氧基哒嗪、柳氮磺吡啶、磺胺嘧啶、磺胺间二甲氧嘧啶、磺胺邻二甲氧嘧啶、磺胺嘧啶钠、磺胺异恶唑、磺胺二甲基嘧啶、磺胺二甲异嘧啶、磺胺二甲基嘧啶钠、磺胺对甲氧嘧啶、磺胺间甲氧嘧啶、磺胺甲氧哒嗪、磺胺甲氧哒嗪钠、磺胺甲氧吡嗪、磺胺甲恶唑、磺胺氯哒嗪、磺胺氯达嗪钠、磺胺苯吡唑、磺胺氯吡嗪钠、磺胺喹恶啉、磺胺喹恶啉钠、磺胺噻唑等药物及其衍生物的各类剂型。抗菌增效剂包括但不限于三甲氧苄氨嘧啶(TMP)、二甲氧苄氨嘧啶(DVD)、二甲氧甲基苄氨嘧啶(OMP)等药物及其衍生物的各类剂型。喹诺酮类抗菌药包括但不限于萘啶酸、吡哌酸、诺氟沙星、氧氟沙星、左氧氟沙星、培氟沙星、依诺沙星、环丙沙星、洛美沙星、氟罗沙星、司帕沙星、恩诺沙星、加替沙星、莫昔沙星、帕珠沙星等药物及其衍生物的各类剂型。抗结核病药物包括但不限于丁烯烟肼、丙硫异烟胺、乙硫异烟胺、乙硫烟胺、云南白药、利福喷丁、利福平-异烟肼(卫非宁)、利福平-异烟肼-吡嗪酰胺(卫非特)、利福平异烟肼、利福平(甲哌利福霉、威福仙、仙道纶)、利福霉素钠(锋立复)、利福霉素钠、吡嗪酰胺、吡嗪酰胺、呋烟腙、夏枯草、大蒜素、威福仙、对氨基水杨酸异 烟肼(核清)、对氨基水杨酸钠、帕司烟肼、异烟肼、异烟腙、异福、异福酰胺(戴菲林)、氨硫脲、硫酸卷曲霉素、硫酸链霉素、派斯钠(对氨水杨酸、抗痨钠、派斯钠)、白及、盐酸乙胺丁醇、石吊兰素(岩豆素)、草分枝杆菌F.U.36(乌体林斯)、醋地砜钠、链霉素(硫酸链霉素)等药物及其衍生物的各类剂型。抗真菌药物包括但不限于以下两类药物及其衍生物的各类剂型:第一类为抗生素,主要有灰黄霉素、制霉菌素和二性霉素B等;另一类是合成药物,包括咪唑类药物(如克霉唑、益康唑、咪康唑和酮康唑等)、氟胞嘧啶和丙烯胺衍生物。抗病毒药物包括但不限于碘苷(疱疹净,IDU)、三氟胸苷(TFT)、阿糖腺苷(Ara-A)、三氮唑核苷(RBV)、无环鸟苷(ACV)、丙氧尿苷(DHPG)、叠氮胸苷(AZT)、双脱氧肌苷(DDI)、金刚烷胺、金刚乙胺、吗啉胍(病毒灵)、酞丁胺、膦甲酸(PFA)、异丙肌苷等药物及其衍生物的各类剂型。抗艾滋病药物包括但不限于核苷类反转录酶抑制剂(NRTI S)、非核苷类反转录酶抑制剂(NNRTIS)、蛋白酶抑制剂(PIS)、整合酶抑制剂(raltegravir)、融合抑制剂(FIS)及CCRS抑制剂(maraviroc)等药物及其衍生物的各类剂型。抗寄生虫药物包括但不限于抗原虫药、抗蠕虫药、杀虫药等药物及其衍生物的各类剂型。
雾化制剂中所用药物可以是抗肿瘤药,具体如表3-3所述。
如实例3-3所述,氟化壳聚糖可以作为药物载体递送抗体药物如细胞程式死亡-配体1抗体,以吸入剂/喷雾剂的方式进行肺部给药。
本专利技术方案可以用于鼻腔给药系统,具体如下。
以氟化壳聚糖为主体,结合药物还可以制成相应的滴鼻剂,促进药物的跨鼻腔黏膜给药。
鼻腔黏膜,简称鼻黏膜,覆盖于鼻腔表面,黏膜下方为软骨、骨或骨骼肌。根据结构与功能的不同,鼻黏膜又分为前庭部、呼吸部和嗅部三部分。前庭部是邻近外鼻孔的部分,有丰富的鼻毛可以阻挡空气中较大的尘粒吸入。呼吸部占鼻黏膜的大部,有发达的上皮纤毛,它们可向咽部摆动,黏着有尘粒、细菌的黏液排向咽部,最终将它们排出体外。此外,此部分有丰富的血管与腺体,对吸入的空气有加温和湿润作用。嗅部黏膜范围较小,主要位于鼻腔顶部。它含有一种专司嗅觉的嗅细胞及嗅腺,它们分泌能溶解到达嗅区的含气味的微粒,刺激嗅细胞表面上的嗅毛产生嗅觉,而且还由于嗅细胞具有不同的受体,可分别接受不同化学分子的刺激,因此可产生不同的嗅觉。人鼻黏膜总表面积约150cm 2,其上皮细胞上有许多微绒毛,分布着大量的毛细血管和淋巴管,增大了药物吸收面积,加速药物吸收。现有的经鼻给药制剂包括滴剂、嗅剂、膏剂、鼻塞、吸入剂、喷雾剂、粉雾剂等。鼻腔上部的黏膜比鼻腔底部和鼻窦内黏膜厚,血管密集,作为药物吸收的主要地方。本专利的产品作为新型跨黏膜载体可提高药物在鼻粘膜部位的滞留时间,减少药物进入口腔,提高利用率,加速药物穿透黏膜进入血液到达病灶部位起治疗作用。其中,绕过血脑屏障,通过鼻黏膜给药治疗脑部疾病作为本专利实例。鼻黏膜富含嗅细胞,嗅细胞的中枢突形成嗅神经纤维,延展出嗅丝,嗅丝终止于嗅神经元的嗅球,沿着嗅球的嗅神经直达脑部。鼻腔给药后,本专利药物载体能延长药物的驻留时间,增加生物利用度,被嗅细胞传递至脑部,绕过血脑屏障进行药物递送。鼻腔的三叉神经也被认为是鼻腔给药靶向中枢的途径之一,因此,本专利利用该技术可以将药物穿透鼻黏膜治疗脑部疾病,三叉神经痛等,用于制备鼻黏膜疫苗,鼻黏膜局部麻醉等领域。
脑中风治疗药物可以包括但不限于下表3-4具体药物及其衍生物。
表3-4
Figure PCTCN2020138465-appb-000016
抗肿瘤药物可以包括但不限于下表3-1具体药物及其衍生物。
表3-1
Figure PCTCN2020138465-appb-000017
Figure PCTCN2020138465-appb-000018
Figure PCTCN2020138465-appb-000019
鼻黏膜给药经过三叉神经途径还可以用于疱疹性三叉/舌咽神经疼痛等治疗,示例包括但不限于氯胺酮、地西拉明、卡马西平等药物及其衍生物。
鼻黏膜给药还能治疗其他疾病,例如消炎药、抗生素、抗病毒药物治疗鼻炎、咽喉、肺部等炎症。示例药物详见雾化制剂部分。
鼻黏膜给药穿透黏膜起局部作用或者进入血液发挥全身作用,如应用于麻醉,示例全身麻醉包括但不限于盐酸氯胺酮,丙泊酚,硫喷妥钠,依托咪脂,咪达唑仑和γ-羟基丁酸钠。局部麻醉药包括但不限于芳酸酯类,芳酰胺类,氨基酮类,氨基醚类,氨基甲酸酯类,羟普鲁卡因,氯普鲁卡因,丁卡因,布他卡因,硫卡因,普鲁卡因胺,布比卡因,阿替卡因,依替卡因,罗哌卡因,甲哌卡因,克罗宁等药物及其衍生物。
任何疾病或机能失调,特别是机体损伤,都可能使人产生疼痛的感觉,很多情况下需要进行镇痛治疗。镇痛药是一类作用于中枢神经系统,对痛觉中枢产生选择性抑制作用,使疼痛减轻或消除而不影响其他感觉的药物,临床上主要用于如创伤,烧伤,手术后和癌症等引起的剧烈锐疼的止疼。
所述镇痛药可以是吗啡及其衍生物包括但不限于:可待因,乙基吗啡,苄基吗啡,异可待因,海洛因,苯乙基吗啡,氢吗啡酮,羟吗啡酮,丙烯吗啡,纳不啡,纳洛酮,钠曲酮等药物及其衍生物。所述镇痛药可以是合成镇痛药包括但不限于:哌替啶,阿尼利定,苯哌利定,匹米诺定,阿法罗定,倍他罗定,芬太尼,阿芬太尼,舒芬太尼,瑞芬太尼,盐酸哌替啶,美沙酮,右吗拉胺,右丙氧芬,盐酸美沙酮,N-甲基吗啡喃,左啡诺,布托啡诺,吗啡喃,喷他佐辛,地佐辛,舒马普坦,曲马多,苯噻啶,奈福泮。所述药物可以是糖尿病治疗药物包括但不限于氨苯磺丁脲、甲苯磺丁脲、氯磺丙脲、乙酸已脲、格列齐特、格列吡啶、格列美脲等磺酰 脲类药物及其衍生物;瑞格列奈、那格列奈等非磺酰脲类药物及其衍生物;罗格列酮、吡咯列酮等噻唑烷二酮类药物及其衍生物;苯乙双胍、二甲双胍等二胍类药物及其衍生物;阿卡波糖、伏格列波糖、米格列醇等α-葡萄糖苷酶抑制剂药物及其衍生物;胰高血糖素样肽,DPP-Ⅳ抑制剂西格列汀、维格列汀、沙格列汀二肽基肽酶-Ⅳ药物及其衍生物以及胰岛素等药物及其衍生物。
同时,还能通过鼻腔吸入喷雾疫苗进行肺部疫苗递送。经鼻疫苗包括但不限于:流感疫苗,百日咳疫苗,结核病喷雾疫苗,流感喷雾疫苗,麻疹喷雾疫苗,百日咳伯德杆菌喷雾疫苗,肺炎衣原体喷雾疫苗,肺炎链球菌喷雾疫苗,炭疽杆菌喷雾疫苗,肺炎球菌表面蛋白-鼻凝胶疫苗,A型肉毒杆菌神经毒素BoHc/A凝胶疫苗,破伤风类毒素凝胶疫苗。
如实例3-4所述,氟化壳聚糖可以作为药物载体从鼻黏膜递送药物,治疗脑肿瘤疾病。
阴道内给药是在阴道内使用药物的给药途径。在药理学上,与其他给药途径相比,它具有主要在阴道或附近结构(例如子宫颈的阴道部分)产生作用,而降低全身不良反应的优势。阴道是药物输送的高效部位,特别是在女性健康方面。阴道通常是药物给药的理想途径,因为与口服途径相比,阴道给药的剂量较低,药物水平稳定且给药频率较低。通过阴道给药,吸收不受胃肠道疾病的影响,没有首过效应。阴道给药方法包括阴道片,阴道乳膏,阴道栓剂和阴道环。阴道疾病多为细菌、病毒感染所致,示例药物同抗细菌抗病毒类药物以及消炎药物。
本专利技术方案可以用于眼部给药系统,具体如下。
涉及需要穿透眼部屏障而使治疗受限的疾病主要分为三类:眼部粘膜感染、角膜/结膜相关感染以及眼底相关的疾病。
第一类,眼部粘膜疾病,主要因为眼部免疫能力下降、维生素缺乏或意外受伤导致粘膜受损发炎或者再经细菌、病毒、真菌、寄生虫、阿米巴和衣原体等病原微生物侵入人体眼部粘膜所引起的局部组织炎症反应,如眼睑感染、眼眶感染等,不进行及时治疗会进一步感染眼球角膜结膜等部位,使病情恶化。临床目前普遍采用单纯含药眼药水、眼凝胶、眼软膏等治疗方式,但存在药物利用率极低,凝胶与软膏对患者的舒适度差,透明可视度差等缺点,对此,本发明利用所述氟化壳聚糖作为载体,将治疗沙眼、睑腺炎、睑板腺囊肿、睑缘炎、睑板腺炎等眼表疾病的药物穿透粘膜递送至病灶部位进行治疗。治疗药物见表4-1。
第二类角膜/结膜相关的感染,主要有各种微生物感染的角膜炎和结膜炎,角膜/结膜新生血管疾病、眼部过敏等疾病。该部位感染具有致病率高、治疗难、治疗烦、易发作、患者顺应性差等缺点。角膜/结膜相关的感染指细菌、病毒、真菌、寄生虫、阿米巴和衣原体等病原微生物侵入人体眼部的角膜与结膜所引起的局部组织炎症反应,如结膜炎症、角膜炎和眼内炎等。感染原因很多,包括不注意眼部卫生;被已感染者直接或间接传染;灰尘等进入眼睛造成角膜异物引发感染;母婴传播等。眼部也经常暴露于病原微生物中如干燥杆菌、葡萄球菌、肺炎球菌、链球菌、绿脓杆菌、大肠杆菌等。在正常情况下这些细菌并不致病,因为结膜角膜屏障完好,细菌不易入侵。但当眼部出现抵抗力下降,角膜结膜上皮损伤等问题时,眼部会有不同部位的感染。眼部常见的致病性真菌如镰刀菌属、曲霉菌属、青霉菌属、白色念珠菌等;眼部常见的病毒有腺病毒、风疹病毒、单纯疱疹病毒、水痘带状疱疹病毒、肠道病毒等;眼部常见的衣原体有沙眼衣原体,常引起沙眼、结膜炎、淋巴肉芽肿,鹦鹉热衣原 体常引起鹦鹉热等。虽然已有眼药水治疗相关炎症以及抗病原体,但一旦炎症深入眼球内部,普通眼药水几乎无能为力。因此有待开发强有力的透屏障材料或者载体进行给药治疗,否则不仅耽误治疗时间,还可能使得细菌多药耐药,感染进一步深入,使治疗难度加大。治疗药物多用针对性药物,见表4-1。本发明利用氟化壳聚糖作为药物载体,因氟化壳聚糖的正电吸附能力能黏附在角膜/结膜表面,并且打开角膜/结膜上皮细胞通道,将治疗眼部感染相关药物递送至角膜/结膜感染部位抑制或清除微生物等。也能将眼部消炎作用的药物递送过去起到消炎止痛作用。
第三类眼底疾病最为棘手,药物若要到达病灶部位需要穿透眼球的重重屏障,目前主要采用玻璃体腔注射以及结膜下或结膜囊给药的方式,均需要使用注射器反复注射给药,对眼球造成不可修复的瘢痕,影响视力。如眼底黄斑病变,眼底黄斑区是视网膜的一个重要区域,与精细视觉及色觉等视功能有关。一旦黄斑区出现病变,常常出现视力下降、眼前黑影或视物变形。眼底黄斑病变可由遗传病变、老年性病变、炎症性病变等所引起,也可受其他眼底病变累及,治疗比较棘手;年龄性黄斑病变主要有老年黄斑变性、老年性特发性视网膜前膜和老年性黄斑洞等改变,通过早期诊断和适当的治疗,可以使病情改善或稳定;炎症性黄斑病变多见于各种视网膜脉络膜炎,如弓浆虫病、葡萄膜炎等;此外,视网膜静脉阻塞、视网膜血管炎、糖尿病性视网膜病变、高度近视和外伤性脉络破裂等可导致黄斑区发生损害,可造成黄斑区水肿或出血而有一定程度的视功能损害。当前治疗分为手术治疗以及药物治疗。手术治疗主要有激光治疗、经瞳温热疗法、光动力疗法,手术切除新生血管、黄斑移位术以及视网膜移植等方式,但手术过程病人承担较大痛苦以及较大风险;药物治疗则主要是以anti-VEGF的系列单抗进行玻腔内给药治疗,但仍然会引起结膜视网膜瘢痕影响视力,也同样使得病人承担较大痛苦和较大风险。眼部疾病还有眼肿瘤给病人带来极大痛苦,眼部肿瘤分为内眼肿瘤和外眼肿瘤,内眼肿瘤伴随瞳孔内有黄色白色反光(俗称猫眼),视力消失,眼压升高,前房出血等症状。外眼肿瘤,早期表现为局部硬结,晚期可侵犯全部眼睑,眼眶及副鼻窦,形成严重局部组织缺损。在婴幼儿眼病中,是性质最严重、危害性最大的一种恶性肿瘤,发生于视网膜核层,具有家族遗传倾向,多发生于5岁以下,本病易发生颅内及远处转移,常危及患儿生命,因此早期发现、早期诊断及早期治疗是提高治愈率、降低死亡率的关键。目前治疗方式主要是手术治疗包括肿瘤切除术,眼球摘除术,眶内容剜出术;放射治疗,用深部X线、Co等深部照射,或P、Sr等浅层照射,按放射治疗常规处理;综合疗法,综合应用中药、西药、放疗及手术等疗法;免疫疗法,采用免疫抑制剂治疗以控制肿瘤的增殖。肿瘤治疗药物极其衍生物见表4-2。但同样存在上述黄斑病变一样的问题,药物难以透过屏障进行治疗。
本发明提供一种氟化壳聚糖在促进药物吸收效率中的应用;提供一种氟化壳聚糖以及其作为多种药物载体的用途。
进一步的,本专利所述技术方案所产生的透皮效应是临时性的,在除去药物后,皮肤角质层细胞即会关闭通道,保护人体安全。皮肤由表皮和真皮组成,其中表皮由浅入深依次为角质层、透明层、颗粒层和生发层。真皮由致密结缔组织构成,由浅入深依次为乳头层和网状层。乳头层与表皮的生发层相连,其中有丰富的毛细血管、淋巴管、神经末梢和触觉小体等感受器。角质层是透皮给药的最大的限流屏障,大部分皮肤的角质层由5-25层扁平角质细胞组成,这些细胞无细胞核,细胞器,细胞膜较厚,无生命,不透水,具有防止组织液 外流,抗磨擦和防感染等功能。含氟化合物修饰的阳离子聚合物可以通过刺激这些细胞的紧密连接蛋白分布改变,降低细胞间紧密连接,并进一步刺激肌动蛋白磷酸化,从而促进细胞旁运,打开细胞间隙,形成通道,并携带药物透过角质层,进一步深入皮肤,而后进入真皮层,进入皮肤毛细血管与淋巴循环并发挥药物作用(见图4-1)。
综上,本发明提供的含氟化合物修饰的阳离子聚合物,具有可以与多种药物进行普适性结合,促进药物吸收,提高药物的生物利用度,减少毒性的优点,效果好,应用十分广泛,具有巨大的商业价值,并且本发明提出的含氟化合物修饰的阳离子聚合物生产简易,具备商业化的基础。
本发明则通过氟化壳聚糖与药物制备成眼药水的形式进行给药,能穿透眼部屏障,将药物部分递送至眼内起到治疗作用。
示例性药物可以包括但不限于下表4-1眼部相关疾病具体药物及其衍生物。
表4-1
Figure PCTCN2020138465-appb-000020
Figure PCTCN2020138465-appb-000021
抗肿瘤药物可以包括但不限于下表4-2具体药物及其衍生物。
表4-2
Figure PCTCN2020138465-appb-000022
Figure PCTCN2020138465-appb-000023
接下来,本发明以含氟化合物修饰的阳离子聚合物,尤其是氟化壳聚糖,作为药物载体透过眼球最主要的角膜屏障,实现药物局部治疗却能治疗眼内或眼底疾病的应用。透膜示意图见图4-2,黑色箭头代表药物的渗透方向。当氟化壳聚糖与药物结合制备成眼药水,滴在眼表时,氟化壳聚糖因带正电黏附在角膜、结膜表面,增加药物滞留时间,并且打开细胞间通道蛋白,帮助药物穿透角膜、结膜屏障,穿透血视网膜屏障,治疗相关疾病。
氟化壳聚糖可以作为药物载体向眼内递送治疗药物,以滴眼液的方式进行角膜给 药,用于治疗眼部疾病,如角膜/结膜相关炎症、老年性黄斑病变、视网膜黑色素瘤等。
综上所述,本发明利用含氟化合物修饰的壳聚糖作为药物载体以及药物递送体,能增加眼药水在眼表的停滞时间,打开眼部相关屏障将药物递送至病灶部位富集起到治疗目的,不仅对于眼部粘膜感染及角膜/结膜相关感染疾病具有较好的治疗效果,对于眼底相关的疾病也有较好的疗效。
进一步地,治疗眼部新生血管所用的药物,包括贝伐单抗、雷珠单抗、索拉菲尼、苏尼替尼。
本发明的一种采用上述穿透屏障的给药制剂的抗生素滴眼液,治疗角膜细菌感染的药物与所述氟化壳聚糖形成的复合物,所述复合物的粒径范围为小于10微米,或所述复合物的粒径范围为不大于500纳米,所述氟化壳聚糖与药物的质量比为1:0.5-30。
进一步地,治疗角膜细菌感染以及眼球内部细菌感染的药物,包括青霉素、红霉素、头孢菌素、头霉素、链霉素、庆大霉素、卡那霉素、阿奇霉素、克拉霉素、罗红霉素、泰利霉素、吉他霉素、万古霉素、去甲万古霉素、替考拉宁、博来霉素、多粘菌素B、多粘菌素E、杆菌肽、林可霉素、克林霉素、多西环素、米诺环素、四环素、金霉素、土霉素、地美环素、强力霉素、替加环素。
本发明的一种采用上述穿透屏障的给药制剂的抗病毒滴眼液,治疗角膜病毒感染的药物与所述氟化壳聚糖形成的复合物,所述复合物的粒径范围为小于10微米,或所述复合物的粒径范围为不大于500nm的复合物,所述氟化壳聚糖与药物的质量比为1:0.5-50。
进一步地,治疗角膜病毒感染以及眼球内部病毒感染的药物,包括碘苷、三氟胸苷、阿糖腺苷、三氮唑核苷、无环鸟苷、丙氧尿苷、叠氮胸苷、双脱氧肌苷、金刚烷胺、金刚乙胺、吗啉胍、酞丁胺、膦甲酸和异丙肌苷。
本发明的一种采用上述穿透屏障的给药制剂的抗炎药物滴眼液,治疗细菌病毒以及损伤引起的眼睛炎症的抗炎药物与所述氟化壳聚糖形成的复合物,所述复合物的粒径范围为小于10微米,优选为不大于500nm的复合物,所述氟化壳聚糖与药物的质量比为1:0.5-50。
进一步地,治疗细菌病毒以及损伤引起的眼睛炎症的抗炎药物,包括阿司匹林、对乙酰氨基酚、非特异性环氧化酶抑制剂、安替比林、安乃近、保泰松、羟布宗、甲芬那酸、吲哚美欣、舒林酸、双氯芬酸钠、布洛芬、普奈生、吡罗昔康、美洛昔康、氢化可的松、皮质酮、醛固酮、曲安西龙、泼尼松龙、乙酸地塞米松、醋丙甲泼尼松。
本发明的一种穿透眼部屏障给药制剂,所述制剂包括组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物能作为穿透泪液屏障、角膜/结膜屏障、血房水屏障、血视网膜屏障给药的制剂。
本发明的一种穿透眼部屏障给药制剂,所述制剂包括组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物在制备用于穿透泪液屏障、角膜/结膜屏障、血房水屏障、血视网膜屏障药物中的应用。
本发明的一种穿透眼部屏障给药制剂,所述制剂包括组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物在制备用于治疗眼部粘膜感染的药物、治疗角膜/结膜相关感染的药物、治疗眼底疾病的药物中的应用。
本专利技术方案可以用于皮肤医美系统,具体如下。
鉴于透皮给药载体存在上述种种技术问题,本发明的目的是提供一种透皮载体作为医疗美容及保健品的给药制剂,本专利以含氟化合物修饰的壳聚糖为主体,可以制成相应的透皮制剂用于需要透过表皮层或粘膜层进行医疗美容及保健品的给药体系。该方法能够提高药物的递送效果,改善人们的使用感受,有很大的应用前景。所述药物可以为医疗美容药物。所述医疗美容药物包括但不限于毛发保健及美容药物,皮肤美容及保健药物。
本专利所述毛发保健及美容药物可以是生发药物和毛发护理药物。以含氟化合物修饰的壳聚糖为主体,可以制成相应的透皮制剂用于治疗各种病因的脱发,制剂如喷雾剂、凝胶剂、膏剂、洗剂等。因此,以此类含氟化合物修饰的阳离子聚合物作为药物载体透过表皮,可实现药物局部治疗到达皮下以及毛囊治疗脱发相关疾病。如实例6-1所述,含氟化合物修饰的壳聚糖可以作为药物载体向皮下以及毛囊递送治疗药物,以喷雾剂的方式进行表皮局部给药,用于治疗脱发疾病。
所述皮肤美容及保健药物也可以是保湿、嫩肤、抗皱、祛斑、祛疤药物,或其他中医美容药物。药物可以达到加速血液循环,促进机体的新陈代谢及组织修复的作用。
所述药物可以是皮炎、湿疹等皮肤病外用药物,包括复方醋酸地塞米松乳膏(皮炎平)、复方硝酸咪康唑乳膏、酮康唑乳膏、曲安奈德益康唑乳膏、丹皮酚软膏、维A酸软膏、老鹳草软膏、复方双花藤直言擦剂、盐酸特比萘芬乳膏、卡泊三醇软膏、喷昔洛韦乳膏、阿昔洛韦凝胶、糠酸莫米松乳膏、曲嘧新乳膏、氧氟沙星凝胶、丁酸氢化可的松乳膏、硝酸咪康唑乳膏、醋酸曲安奈德益康唑软膏、甲硝唑凝胶、联苯苄唑乳膏等。
所述药物可以是激素类药物或维A酸类药物(又称维甲酸类药物)。所述激素类药物可以是糖皮质激素,如皮康王(氯倍他索)、皮炎平(地塞米松)、祛斑霜(含氯倍他索)、肤轻松等。糖皮质激素的治疗作用主要来自其抗炎作用,包括抑制溶酶体酶的释放、抑制对巨噬细胞的刺激及毛细血管的收缩作用等,但容易出现副作用如皮炎、毛囊炎等。所述维甲酸类药物包括非芳香甲酸类药物(如维A酸和异维A酸)、单芳香甲酸类药物(如异曲替酯和异曲替酸)、多芳香维A酸类药物(如芳香甲乙酯和阿达帕林)。维A酸类药物具有抗皮肤老化、减少表皮黑素、抑制皮脂产生、免疫调节的作用,可以护肤养颜、减轻皱纹、消退黄褐斑,治疗痤疮、色斑、角化异常等皮肤问题。但维A酸类药物口服副作用大,表皮使用易引发局部刺激导致灼热感和微痛,临床使用收到了一定的限制。
特别地,对于祛疤来说,含氟化合物修饰的高分子可以改善渗透效率,从而减少药物在患处的用量,从而达到降低副作用的效果。在实例6-2中,本发明以含氟化合物修饰的阳离子聚合物作为药物载体透过表皮,将聚肌胞苷酸递送到角质层以下,诱导内源性维甲酸的产生,实现药物局部治疗,以达到改善疤痕且低副作用的效果。
所述药物可以是外用抗氧化剂,包括维生素E、绿茶多酚类、乙酰半胱氨酸、辅酶Q10、超氧化物歧化酶等,可以有效对细胞中的氧化应激反应,达到抗衰老的目的。所述药物可以是ω-3脂肪酸、a-羟酸类、β-胡萝卜素等药物,达到增加皮肤弹性、胶原合成的作用。
所述皮肤美容及保健药物也可以是维生素与微量元素,改善肌肤的营养状态,美容养颜,延缓衰老。营养素如铁、碘、锌、维生素A、维生素B2、烟酸、生物素、叶酸、维生素B12、维生素C和维生素D的缺乏可能会损害生长并导致疾病,这是一个公众广泛关注的健康问题。尽管通过强化食物的营养可以有效地治疗营养素的缺乏,但仍存在营养素不够稳定,吸收效率不够高的问题。与游离的营养素相比,封装在纳米颗粒中的营养素更为稳定,但生物 利用度较低。采用全氟庚酸修饰的壳聚糖作为载体的营养素制剂可以有效地改善生物利用度,促进营养素的吸收。
所述皮肤美容及保健药物也可以是中医美容药物,改善肤色暗沉、色素沉着、皮肤松弛、毛孔粗大、皮肤粗糙等皮肤问题或修复皮肤屏障,恢复肌肤健康。所述中医美容药物可以是延缓细胞老化药物(黄芪和人参的提取物);清除自由基药物(五味子、三七、提取物,类黄酮药物,丹参酸乙和丹酚酸C活性物);修复胶原药物(茯苓提取物,黄精和枸杞子,丹参酸乙和丹酚酸C活性物);免疫调节药物(五味子提取物,皂苷,菟丝子提取物);促进皮肤微循环药物(丹参提取物,类黄酮)等中药以及中草药。
黄褐斑为面部的黄褐色色素沉着,其病因复杂,治疗苦难,容易复发,给患者带来了很大的烦恼。药物治疗中往往采用抗氧化剂药物或糖皮质激素药物,如氨甲环酸、原花青素、谷胱甘肽、氢醌(又名对苯二酚)、维A酸、半胱胺盐、寡肽、Jessner溶液、水杨酸、乳酸。其中氨甲环酸能抑制角质形成细胞中紫外线诱导的纤溶酶活性,阻止纤溶酶原与角质细胞结合,降低前列腺素的合成,从而抑制色素产生。口服或局部注射氨甲环酸均可治疗黄褐斑。但口服前需筛查个人及家族的血栓血塞风险,局部给药时渗透效果不尽如人意。因此本发明以含氟化合物修饰的阳离子聚合物作为药物载体透过表皮,实现药物局部治疗到达皮下治疗黄褐斑的效果。如实例6-3所述,含氟化合物修饰的壳聚糖可以作为药物载体向皮下递送治疗药物,进行表皮局部给药,用于治疗黄褐斑。
本发明的一种采用上述医美及保健品载体的药物复合物,包括医美及保健品载体(a)和药物组分(b),所述药物组分(b)为,生发药物和毛发护理药物;或保湿、嫩肤、抗皱、祛斑、祛疤药物;或皮炎、湿疹皮肤病外用药物;或激素类药物或维A酸类药物;或维生素E、绿茶多酚类、乙酰半胱氨酸、辅酶Q10、超氧化物歧化酶。
本发明的一种医美及保健品载体制剂,所述制剂包括组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物能作为生发药物和毛发护理药物、美容药物、保健药物给药的制剂。
本发明的一种采用上述的医美及保健品载体制剂制备的用于生发的外用制剂,包括线粒体丙酮酸转运蛋白有效抑制剂UK5099、二甲双胍、米诺地尔、螺内酯、非那雄安与氟化壳聚糖形成的复合物,所述复合物的粒径范围为小于10微米,或所述复合物的粒径范围不大于500纳米,所述氟化壳聚糖与生发药物的质量比为1:0.5-50。
本发明的一种采用上述的医美及保健品载体制剂制备的用于祛疤的复合物,包括以全氟庚酸修饰的壳聚糖和聚肌胞苷酸为主体形成的复合物颗粒,所述粒径范围为小于10微米,或所述复合物颗粒为不大于500纳米的复合物,所述氟化壳聚糖与祛斑类药物的质量比为1:0.5-50。
本发明的一种采用上述的医美及保健品载体制剂制备的治疗黄褐斑的药物复合物,所述复合物为全氟庚酸修饰的壳聚糖包载氨甲环酸,所述氟化壳聚糖与黄褐斑治疗药物的质量比为1:0.5-50。
为解决上述技术问题,本发明提供了如下技术方案:
进一步的,本专利所述技术方案所产生的透皮效应是临时性的,在除去药物后,皮肤角质层细胞即会关闭通道,保护人体安全。皮肤由表皮和真皮组成,其中表皮由浅入深依次为角质层、透明层、颗粒层和生发层。真皮由致密结缔组织构成,由浅入深依次为乳头层 和网状层。乳头层与表皮的生发层相连,其中有丰富的毛细血管、淋巴管、神经末梢和触觉小体等感受器。角质层是透皮给药的最大的限流屏障,大部分皮肤的角质层由5-25层扁平角质细胞组成,这些细胞无细胞核,细胞器,细胞膜较厚,无生命,不透水,具有防止组织液外流,抗磨擦和防感染等功能。含氟化合物修饰的阳离子聚合物可以通过刺激这些细胞的紧密连接蛋白分布改变,降低细胞间紧密连接,并进一步刺激肌动蛋白磷酸化,从而促进细胞旁运,打开细胞间隙,形成通道,并携带药物透过角质层,进一步深入皮肤,而后进入真皮层,进入皮肤毛细血管与淋巴循环并发挥药物作用
综上,本发明提供的含氟化合物修饰的阳离子聚合物,具有可以与多种药物进行普适性结合,促进药物吸收,提高药物的生物利用度,减少毒性的优点,效果好,应用十分广泛,具有巨大的商业价值,并且本发明提出的含氟化合物修饰的阳离子聚合物生产简易,具备商业化的基础。
本发明还提供一种促进药物跨黏膜吸收,生物安全性好的氟化合物修饰的阳离子聚合物,尤其是氟化壳聚糖。本发明提出的含氟化合物修饰的阳离子聚合物,合成方法简单,具备普适性;并且在跨黏膜给药方面具有明显优势,主要包括:(1)具有良好的渗透性,可以跨越各类黏膜(鼻腔黏膜、肺粘膜、阴道黏膜、口腔黏膜、胃肠黏膜等),增加药物在血液、组织中的浓度;(2)具有良好的粘附性,可以粘附在各类黏膜表面,实现药物的缓释;(3)应用广泛,既可与大分子药物结合,也可以吸附小分子药物,还可以与成分复杂的复方药物结合,用于治疗各类疾病,具备极大的商业价值。
综上,本专利提供的氟化壳聚糖药物载体,具有促进药物吸收效果明显,低毒性等优点,并且本发明提出的氟化壳聚糖合成工艺成熟、操作简易,合成效率高,周期短,无需繁琐的纯化步骤即可获得高产率的药物载体,其简易的合成方法为其提供了商业化的良好基础,本发明所述的氟化壳聚糖具有作为多种药物载体的用途,能够有效的提升治疗效果,具有广泛的用途,并且成本较低。
进一步的,本专利所述技术方案所产生的透粘膜效应是临时性的,在除去药物后,肠道粘膜细胞即会关闭通道,保护人体安全。(见图3-1)。
本发明提供的含氟化合物修饰的阳离子聚合物尤其是氟化壳聚糖,具有可以与多种药物进行普适性结合,促进药物吸收,提高药物的生物利用度,减少毒性的优点,效果好,应用十分广泛,具有巨大的商业价值,并且本发明提出的含氟化合物修饰的阳离子聚合物生产简易,具备商业化的基础。
本专利设计的氟化修饰的阳离子聚合物,其在生物化学和药剂学领域的应用包括但不限于以下给药方式:口服给药系统、喷雾剂/吸入剂、滴鼻剂。作为本发明所述的含氟化合物修饰的阳离子聚合物的一种优选方案:所述阳离子聚合物为壳聚糖,其脱乙酰度>95%。以下应用以氟化修饰的壳聚糖为例,其他阳离子聚合物也可以经过氟化修饰达到相似的效果。
附图说明
图1-1为实施例1-5中七氟丁酸修饰壳聚糖(7FCS)对THP在小鼠膀胱组织中分布及强度的影响,其中,THP,表阿霉素;CS,壳聚糖;FCS,氟化壳聚糖;右图为左图对应的THP相对荧光强度分析。
图1-2为实施例1-6中十三氟庚酸修饰壳聚糖(13FCS)对THP在小鼠膀胱组织中分布及强度的影响,其中表阿霉素为THP;CS,壳聚糖;FCS,氟化壳聚糖;右图为左图对应的THP相对荧光强度分析。
图1-3为实施例1-7中不同含氟脂肪酸修饰壳聚糖(7FCS,13FCS,19FCS)对THP在小鼠膀胱组织中分布及强度的影响,其中表阿霉素,THP;CS,壳聚糖;FCS,氟化壳聚糖;右图为左图对应的THP相对荧光强度分析。
图1-4a为实施例1-8中13F-3具有很好的体外细胞安全性的对比图。
图1-4b为实施例1-8中FCS组小鼠体重与空白对照组无明显差异。
图1-4c为实施例1-8中各组灌注后小鼠膀胱与空白对照组小鼠膀胱组织及HE(苏木精—伊红)染色切片对比图片。
图1-5为实施例1-8中免疫荧光结果对照图片,表明壳聚糖灌注组小鼠膀胱出现了严重得炎症应激与充血水肿,而FCS组与空白对照组无明显差异,左图为膀胱组织切片的荧光共聚焦图片,右图为CS,FCS处理组与空白组control。
图1-6为实施例1-9中透射电镜测得MPI/FPEI的相关图片。
图1-7为实施例1-9中透射电镜测得MPI/PEI的相关图片。
图1-8为实施例1-9中F-PEI组多肽的黏膜渗透指数要显著高于PEI组及空白对照组,其中横坐标为多肽药物MPI与材料PEI或FPEI的投料质量比,纵坐标为渗透系数papp。
图1-9为实施例1-9中F-PEI组蛋白药物的黏膜渗透指数要显著高于PEI组及空白对照组,横坐标为多肽药物CAT-Ce6与材料PEI或FPEI的投料质量比,纵坐标为渗透系数papp。
图1-10为实施例1-9中MPI不同药物体系在灌注药物不同时间后取膀胱制备冰冻切片及药物荧光在组织分布及强度对照图表,横坐标为不同灌注时间,纵坐标为多肽药物荧光强度的相对值。
图1-11为实施例1-9中CAT不同药物体系在灌注后取膀胱制备冰冻切片及药物荧光在组织分布及强度对照图表,左图为药物CAT-Ce6不同药物体系在膀胱组织中的荧光分布,右图为药物的荧光强度分析。
图1-12为实施例1-10中透射电子显微镜(TEM)成像图片。
图1-13为实施例1-10中膀胱冰冻切片以及荧光共聚焦显微镜分析荧光强度的对照图表,左图从左至右依次为药物CAT-TCPP在膀胱组织横切、纵切切片中的荧光分布;右图从上至下依次为药物在膀胱横切纵切已经膀胱组织匀浆中的荧光强度。
图1-14为含氟羧酸修饰的壳聚糖膀胱灌注药物载体的合成路线图。
图2-1含氟化合物修饰的阳离子聚合物的透皮机制示意图。
图2-2不同比例全氟庚酸修饰的壳聚糖与药物反应前后在水溶液中的粒径变化,以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图2-3不同比例全氟庚酸修饰的壳聚糖与药物反应前后在水溶液中的电位变化,以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图2-4不同比例的全氟庚酸修饰的壳聚糖与药物透皮效果差异,其中右图为透皮扩散池示意图,以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图2-5凝胶实物照片及扫描电镜照片,以全氟庚酸修饰的壳聚糖-胰岛素(FCS- Insulin)为例。
图2-6药物从凝胶中的释放,以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图2-7药物在活体水平的治疗效果即施用贴剂后血糖的波动情况以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图2-8不同比例的全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)在水溶液中的粒径及电位大小。
图2-9不同比例的全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)在不同时间点对皮肤的累计透过量,其中全氟庚酸修饰的壳聚糖-免疫球蛋白G的比例分别为1:0.25,1:0.5,1:1,1:2,1:4以及单纯的免疫球蛋白G(0:1)。
图2-10全氟庚酸修饰的壳聚糖-免疫球蛋白G(以下简称FCS-IgG)进行活体透皮动力学分析,其中,前5张图分别为不同时间点小鼠肿瘤组织(灰色)切片的荧光强度(白色),第6张图为小鼠不同时间点肿瘤组织裂解后检测的荧光强度定量分析。
图2-11全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体与壳聚糖-细胞程式死亡-配体1抗体以及纯细胞程式死亡-配体1抗体的体内肿瘤组织透皮效率对比,从上往下分别为纯细胞程式死亡-配体1抗体组(free aPDL1),壳聚糖-细胞程式死亡-配体1抗体(CS-aPDL1),以及全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体(FCS-aPDL1)组,从左往右分别外DAPI通道(灰色,表示肿瘤组织),FITC荧光通道(白色,表示细胞程式死亡-配体1抗体)以及混合通道的荧光强度。
图2-12全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)的透皮机制:全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)对致密的细胞单层的细胞电阻影响,左图为测量方法示意图,右图为加入全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)后的电阻变化。
图2-13全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)的透皮机制:全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)对致密的细胞单层的相关紧密连接蛋白的影响的免疫荧光染色图。右图为荧光强度的半定量分析图。
图2-14全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)的透皮机制:全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)对致密的细胞单层的相关紧密连接蛋白的影响的免疫印迹法(Western Blotting)分析。
图2-15全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体活体皮下肿瘤治疗,分组分别为空白组(blank),纯细胞程式死亡-配体1抗体(free aPDL1),壳聚糖-细胞程式死亡-配体1抗体(CS-aPDL1),以及全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体(FCS-aPDL1)组,左图小鼠肿瘤生长曲线,右图为小鼠存活率曲线(定义小鼠肿瘤大于 1500立方毫米即为死亡)。
图3-1为肠道黏膜示意图。
图3-2为全氟庚酸修饰的壳聚糖打开肠黏膜上皮细胞屏障的示意图。
图3-3为实施例3-1中不同比例全氟庚酸修饰的壳聚糖与药物反应前后在水溶液中的粒径变化,以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图3-4为实施例3-1中不同比例全氟庚酸修饰的壳聚糖与药物反应前后在水溶液中的电位变化,以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图3-5为实施例3-1中冻干前后全氟庚酸修饰的壳聚糖药物复合物的粒径变化,以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图3-6为实施例3-1中不同比例的全氟庚酸修饰的壳聚糖与药物透黏膜效果差异,以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图3-7为实施例3-1中载药胶囊在胃液和肠液中透黏膜递送药物的效果,以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图3-8为实施例3-1中口服递送载药胶囊后小鼠血糖波动情况,以全氟庚酸修饰的壳聚糖-胰岛素(FCS-Insulin)为例。
图3-9为实施例3-2中不同比例的全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)在不同时间点对肠粘膜的累计透过量。其中横坐标为进样时间,纵坐标为累计透过量除以总进样量的透过率。与单纯的大分子蛋白免疫球蛋白G相比,不同分子量全氟庚酸修饰的壳聚糖-免疫球蛋白G均可增加免疫球蛋白G透大鼠肠粘膜效率。优选得到全氟庚酸修饰的壳聚糖-免疫球蛋白G比例为1:1的复合物。
图3-10为实施例3-2中全氟庚酸修饰的壳聚糖-牛血清蛋白-菁染料Cy5.5在第3小时,第5小时在消化道的部位,其中白色代表菁染料Cy5.5标记的牛血清蛋白,说明该胶囊能定点到结直肠释放,从而选用该胶囊进行后续实验。
图3-11为实施例3-2中细胞程式死亡配体抗体1冻干前后活性示意图,加入冻干保护剂冻干之后细胞程式死亡-配体1抗体变成冻干粉之后活力不变。说明全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体颗粒可以进行冻干,可以用于口服胶囊装填。
图3-12为实施例3-2中冻干前后FCS/a PD-L1和CT-26细胞表面PD-L1结合活性。
图3-13为实施例3-2中全氟庚酸修饰的壳聚糖和全氟庚酸修饰的壳聚糖/免疫球蛋白G处理前后人结直肠癌上皮细胞紧密连接蛋白分布变化。
图3-14为实施例3-2中装有全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体冻干粉末胶囊治疗Balb/c小鼠结直肠癌的效果,其中黑色表示小鼠结直肠癌细胞生物发光强度。
图3-15为实施例3-2中不同分子量的全氟庚酸修饰的壳聚糖/免疫球蛋白G的透粘膜效果。
图3-16为实施例3-3中小鼠肺部免疫荧光切片,荧光信号表示药物在肺部的滞留。
图3-17为实施例3-4中各个实验组的小鼠脑部照片,其中白色箭头指示小鼠脑部Cy5.5的荧光信号。。
图3-18为实施例3-4中小鼠脑部切片激光共聚焦照片,其中白色箭头指示小鼠脑肿瘤部位Cy5.5的荧光信号。
图4-1为眼球结构示意图。
图4-2为含氟药物滴眼液透膜示意图。
图4-3为实施例4-1中不同比例全氟庚酸修饰的壳聚糖在体外穿透角膜的累积透过百分比。
图4-4为实施例4-1中大分子药物牛血清白蛋白眼球免疫荧光染色图,左图为含全氟庚酸修饰的壳聚糖透膜免疫荧光染色图,右图为游离药物透膜免疫荧光染色图。
图4-5为实施例4-1中小分子药物罗丹明B眼球免疫荧光染色图,左图为含全氟庚 酸修饰的壳聚糖透膜免疫荧光染色图,右图为游离药物透膜免疫荧光染色图。
图4-6为实施例4-1中全氟庚酸修饰的壳聚糖的白蛋白滴眼后眼部组织含量。
图4-7为实施例4-1中全氟庚酸修饰的壳聚糖局部角膜滴眼后不同时间的免疫荧光染色。
图4-8为实施例4-1中全氟庚酸修饰的壳聚糖在小鼠各时间点角膜荧光染色前节照片检测全氟庚酸修饰的壳聚糖的安全性。
图4-9为实施例4-2中治疗前与治疗一周后原位脉络膜恶性黑色素瘤的生物自发光图像。
图4-10为实施例4-2中治疗一周后原位脉络膜恶性黑色素瘤的生物自发光定量分析图。
图5-1为实施例5-1中全氟庚酸修饰的壳聚糖-鸡卵清白蛋白(FCS-OVA)复合物体外刺激后,成熟的树突状细胞占总树突状细胞的百分比的流式细胞仪处理图与统计图,成熟的树突状细胞能有效地呈递抗原,激活T细胞,启动免疫应答,空白为没有处理的自然成熟阴性对照组,脂多糖为加了脂多糖的阳性对照组,全氟庚酸修饰的壳聚糖-鸡卵清白蛋白为实验组。
图5-2为实施例5-1中全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物体外刺激后,树突状细胞表达呈递抗原的关键蛋白——第二类组织相容性复合体(MHC II)与分化集群40蛋白(CD40)的变化。别藻青蛋白染料的强度越强,表明CD40蛋白在树突状细胞表面表达越多,藻红蛋白染料的强度越强表明MHC II在树突状细胞表面表达越多。
图5-3为实施例5-1中带荧光的全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物与软膏混合敷在小鼠背部,淋巴结内全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物在淋巴结的聚集随时间变化的活体成像。白色箭头所示为小鼠其中一个淋巴结。
图5-4为实施例5-2中使用透皮敷贴种植全氟庚酸修饰的壳聚糖-鸡卵清白蛋白疫苗复合物的小鼠与使用透皮敷贴种植鸡卵清白蛋白疫苗的小鼠,在接种疫苗后,在小鼠背部种植细胞表面表达鸡卵清白蛋白的黑色素瘤(B16-OVA)后,肿瘤随时间的生长的情况。
图6-1为实施例6-1中不同比例的全氟庚酸修饰的壳聚糖与具有生发治疗效果的小分子药物UK5099不同时间体外透皮的递送效率。
图6-2为实施例6-1中不同比例的全氟庚酸修饰的壳聚糖与具有生发治疗效果的小分子药物UK5099不同时间体外透皮的累积透过量。
图6-3为实施例6-1经不同治疗方式治疗的不同时间点小鼠背部毛发生长情况。其中,FCS-二甲双胍为全氟庚酸修饰的壳聚糖修饰上二甲双胍进行透皮的实验组,二甲双胍为游离药物组。
图6-4为实施例6-2,左图为全氟庚酸修饰的壳聚糖和poly(I:C)的混合物对于小鼠成纤维细胞产生维甲酸的效率,右图为全氟庚酸修饰的壳聚糖和poly(I:C)的混合物涂抹于小鼠疤痕后的效果图。
图6-5为实施例6-3中在扩散池透大鼠皮实验中,全氟庚酸修饰的壳聚糖与氨甲环酸混合和单纯氨甲环酸在三小时内氨甲环酸的累计透过大鼠皮的累计百分比【(累计透过量/理论全部透过量)X 100%】。
具体实施方式
以下结合具体实施例和附图,对本发明做进一步的详细说明,本发明的保护内容不局限于以下实施例。在不背离发明构思的精神和范围下,本领域技术人员能够想到的变化和优点都被包括在本发明中,并且以所附的权利要求书为保护范围。
缩写注释:THP(表阿霉素);EDC(1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐),NHS(N-羟基硫代琥珀酰亚胺);DMSO(二甲基亚砜);MPI(多肽药物Polybia-MPI);MPI-Cy5.5(对多肽药物MPI进行荧光标记);PEI(聚乙烯亚胺);FPEI(氟化聚乙烯亚胺);CAT(蛋白药物过氧化氢酶);CAT-Ce6(标记光敏剂Ce6的复合蛋白药物);CAT-TCPP(标记声敏剂TCPP的复合蛋白药物)。
鉴于,本专利文件中的实施例及附图比较多,因此为了区分各组不同实施例,以及不同附图等,实施例的命名采取如下规则:第一个系列实施例为实施例1-1、实施例1-2、实施例1-3,以此类推,第二个系列实施例为实施例2-1、实施例2-2,以此类推。附图命名采用类似规则:第一个系列附图为附图1-1、附图1-2、附图1-3,以此类推,第二个系列附图为附图2-1、附图2-2,以此类推。
参照例
实验设计人员分别以阿霉素、表阿霉素(THP)以及荧光染料罗丹明B作为膀胱灌注药物与壳聚糖(1%醋酸水溶液)混合制备壳聚糖膀胱灌注药物体系,膀胱灌注1h后取膀胱组织制备冰冻切片,以共聚焦荧光显微镜进行药物荧光强度分析,考察药物在膀胱组织的分布情况。实验结果表明等浓度的膀胱灌注药物的壳聚糖(5mg/ml、10mg/ml、15mg/ml)溶液,在膀胱黏膜中的黏附、渗透能力显著优于灌注单纯药物的水溶液,且随着壳聚糖浓度的增加灌注体系的黏附、渗透能力越强。为了进一步考察壳聚糖作为灌注药物载体的生物安全性,项目设计者将15mg/ml的壳聚糖水溶液进行小鼠膀胱灌注处理,1h后停止灌注,正常饲养条件继续饲养小鼠,并记录其体重。实验结果发现,小鼠在灌注壳聚糖后第二天就出现体重骤降,小鼠活动萎靡的状况。从治疗后的第二天起小鼠出现死亡,并在陆续的3天内实验组8只小鼠全部死亡,经解剖取其膀胱与空白对照组小鼠膀胱对比发现,壳聚糖灌注组小鼠膀胱相对于空白组小鼠膀胱充血严重,HE染色以及CD45,Ki67免疫荧光结果表明壳聚糖灌注组小鼠膀胱出现了严重得炎症应激与充血水肿。以上实验结果表明虽然壳聚糖可显著提高灌注药物在膀胱黏膜的生物利用度,但高浓度的壳聚糖同时会引起严重的膀胱黏膜、上皮损伤,严重限制了其作为膀胱灌注药物载体的临床应用。
实施例1-1:制备3-氟苯甲酸不同修饰程度的壳聚糖(脱乙酰度≧95%,粘度100-200mpa.s),其中3-氟苯甲酸与N氨基葡萄糖单元的投料摩尔比例分别为1:1.1、1:2.2、1:4.4、1:8.8。
合成方法:(1)制备壳聚糖醋酸水溶液:称取200mg充分干燥的壳聚糖加入10ml1%的醋酸水溶液中,当然也可以采用盐酸水溶液,搅拌30min使充分溶解,随后缓慢滴加1.6ml 0.5M的氢氧化钠,搅拌至溶液澄清,pH在6.5左右。单纯考虑碱化溶液的角度氢氧化钠可以被氨水,三乙胺等碱替换,但是从产品工艺角度使用氢氧化钠的副产物是氯化钠,更适合工业化。如此方法制备4份壳聚糖醋酸水溶液。(2)3-氟苯甲酸的活化:分别称取5.0mg、9.8mg、19.7mg、40mg的3-氟苯甲酸,将其溶于适量无水二甲基亚砜中,依次加入反应量EDC,NHS避光搅拌1h。(3)3-氟苯甲酰壳聚糖的制备:将上述活化好的3-氟苯甲酸溶液分别缓慢 滴加到快速搅拌的壳聚糖溶液中,避光搅拌反应20h。反应结束,依次将前述反应后的溶液缓慢滴加到100ml 0.5M氢氧化钾乙醇溶液中搅拌8h,过滤沉淀,用大量无水乙醇冲洗,至滤液呈中性,沉淀经甲醇、乙醚洗涤脱水,真空干燥30min。干燥后的沉淀物溶于10ml 0.1M盐酸溶液,冻干得外观白色粉末的不同氟化修饰程度3-氟苯甲酸氟化壳聚糖盐酸盐分子(产物命名为1FCS-1,1FCS-2,1FCS-3,1FCS-4)。
以上反应所得材料以茚三酮反应法检测氟化修饰的壳聚糖(FCS)高分子表面修饰氟化脂肪链的修饰度。茚三酮反应法是一种简单、快速、准确、可靠的方法,可以准确检测水溶液中FCS高分子表面伯氨基团的数量,近而计算出FCS表面氟化基团的数量。
实施例1-2:制备七氟丁酸不同修饰程度的壳聚糖(脱乙酰度≧95%,粘度100-200mpa.s),其中全氟庚酸与N氨基葡萄糖单元的投料摩尔比例分别为1:1.1、1:2.2、1:4.4、1:8.8。
合成方法:(1)制备壳聚糖醋酸水溶液:称取200mg充分干燥的壳聚糖加入10ml1%的醋酸水溶液中,搅拌30min使充分溶解,随后缓慢滴加1.6ml 0.5M的氢氧化钠,搅拌至溶液澄清,pH在6.5左右。如此方法制备4份壳聚糖醋酸水溶液。(2)七氟丁酸的活化:分别称取7.6mg、15mg、30mg、61mg七氟丁酸,将其溶于适量无水二甲基亚砜中,依次加入反应量EDC,NHS避光搅拌1h。(3)七氟丁酸壳聚糖的制备:将上述活化好的七氟丁酸溶液分别缓慢滴加到快速搅拌的壳聚糖溶液中,避光搅拌反应20h。反应结束,依次将反应缓慢滴加到100ml0.5M氢氧化钾乙醇溶液中搅拌8h,过滤沉淀,用大量无水乙醇冲洗,至滤液呈中性,沉淀经甲醇、乙醚洗涤脱水,真空干燥30min。干燥后的沉淀物溶于10ml 0.1M盐酸溶液,冻干得外观白色粉末的不同氟化修饰程度全氟庚酸氟化壳聚糖盐酸盐分子(产物命名为7FCS-1,7FCS-2,7FCS-3,7FCS-4)。
以上反应所得材料以茚三酮反应法检测FCS(氟化修饰的壳聚糖)高分子表面修饰氟化脂肪链的修饰度。茚三酮反应法是一种简单、快速、准确、可靠的方法,可以准确检测水溶液中FCS高分子表面伯氨基团的数量,近而计算出FCS表面氟化基团的数量。茚三酮反应法计算以上制备FCS的氟化修饰程度依次为:7FCS-1,6.9%;7FCS-2,10.4%;7FCS-3,23.5%;7FCS-4,42.3%。
实施例1-3:制备全氟庚酸不同修饰程度的壳聚糖(脱乙酰度≧95%,粘度100-200mpa.s),其中全氟庚酸与N氨基葡萄糖单元的投料摩尔比例分别为1:1.1、1:2.2、1:4.4、1:8.8。
合成方法:(1)制备壳聚糖醋酸水溶液:称取200mg充分干燥的壳聚糖加入10ml1%的醋酸水溶液中,搅拌30min使充分溶解,随后缓慢滴加1.6ml 0.5M的氢氧化钠,搅拌至溶液澄清,pH在6.5左右。如此方法制备4份壳聚糖醋酸水溶液。(2)全氟庚酸(13氟庚酸)的活化:分别称取13mg、26mg、51.5mg、103mg全氟庚酸,将其溶于适量无水二甲基亚砜中,依次加入适量EDC,NHS避光搅拌1h。(3)13F庚酸壳聚糖的制备:将上述活化好的全氟酸溶液分别缓慢滴加到快速搅拌的壳聚糖溶液中,避光搅拌反应20h。反应结束,依次将反应缓慢滴加到100ml 0.5M氢氧化钾乙醇溶液中搅拌8h,过滤沉淀,用大量无水乙醇冲洗,至滤液呈中性,沉淀经甲醇、乙醚洗涤脱水,真空干燥30min。干燥后的沉淀物溶于10ml 0.1M盐酸溶液,冻干得外观白色粉末的不同氟化修饰程度全氟庚酸氟化壳聚糖盐酸盐分子(产物命名为13FCS-1,13FCS-2,13FCS-3,13FCS-4)。
茚三酮反应法计算以上制备FCS的氟化修饰程度依次为:13FCS-1,5.2%;13FCS-2,11.3%;13FCS-3,21.4%;13FCS-4,42.5%。13FCS-1~13FCS-413氟庚羰基基团的连接效率随着全氟庚酸投料的增加为5.2%~42.5%,即平均每个壳聚糖分子中有5.2%~42.5%的葡萄糖结构单元中完成了氟化修饰,产物命名为13FCS-1,13FCS-2,13FCS-3,13FCS-4。
实施例1-4:制备19F癸酸不同修饰程度的壳聚糖(脱乙酰度≧95%,粘度100-20 0mpa.s),其中19F癸酸与N氨基葡萄糖单元的投料摩尔比例分别为1:1.1、1:2.2。
合成方法:(1)制备壳聚糖醋酸水溶液:称取200mg充分干燥的壳聚糖加入10ml1%的醋酸水溶液中,搅拌30min使充分溶解,随后缓慢滴加1.6ml 0.5M的氢氧化钠,搅拌至溶液澄清,pH在6.5左右。如此方法制备2份壳聚糖醋酸水溶液。(2)19F癸酸的活化:分别称取18mg、36.7mg19F癸酸,将其溶于适量无水二甲基亚砜中,依次加入适量EDC,NHS避光搅拌1h。(3)19F癸酸壳聚糖的制备:将上述活化好的19F癸酸溶液分别缓慢滴加到快速搅拌的壳聚糖溶液中,避光搅拌反应20h。反应结束,依次将反应缓慢滴加到100ml 0.5M氢氧化钾乙醇溶液中搅拌8h,过滤沉淀,用大量无水乙醇冲洗,至滤液呈中性,沉淀经甲醇、乙醚洗涤脱水,真空干燥30min。干燥后的沉淀物溶于10ml 0.1M盐酸溶液,冻干得外观白色粉末的不同氟化修饰程度19F癸酸氟化壳聚糖盐酸盐分子(产物命名为19FCS-1,19FCS-2)。
19FCS-2的水溶性比较差,无法进行后续的表征及应用评价,故用茚三酮反应法计算以上制备19FCS-1的氟化修饰程度依次为:19FCS-1,5.2%。
实施例1-5:制备7FCS的膀胱黏膜促渗透作用评价:将实施例1-2中所制备的7FC S和THP水溶液混合,经小鼠尿道灌注到膀胱,后制备小鼠膀胱冰冻切片,通过检测THP荧光在组织中的分布来评估药物载体的促药物膀胱黏膜吸收效率。
具体方法为:将10-12周的雌性C57BL/6小鼠以戊苯巴比妥溶液麻醉,以0.5%的FCS水溶液配置0.2%的THP溶液,通过密闭式静脉留置针将其灌注到小鼠膀胱,100μl,夹闭尿道1h,随后放出膀胱内的灌注液,用1ml超纯水冲洗膀胱,取膀胱组织放入组织包埋机中置于-80℃,后进行切片用荧光共聚焦显微镜进行检测。以单纯的等浓度THP水溶液或同样配制的THP壳聚糖水溶液作为对照。
实验结果:参见图1-1,左侧为不同条件下显微镜对小鼠膀胱组织切片的图片,右侧为对应的数据统计,右侧的横坐标为不同药物体系,纵坐标为relative fluorescence intensity相对荧光强度。
如图1-1所示荧光共聚焦显微镜对小鼠膀胱组织切片进行观察,结果发现7FCS组小鼠膀胱横切面的药物荧光分布面积及强度均显著高于壳聚糖(CS)以及空白对照组(单纯THP水溶液),表明FCS可显著提高药物在膀胱黏膜的组织渗透性,且随着氟化程度的增高促渗透作用提高,但当氟化程度达到一定程度时(7FCS,42.3%)制备所得的FCS的促渗透作用最为显著,此时制备的7FCS溶解性较低氟化取代壳聚糖差,不易于临床应用。以上结果表明FCS可显著提高药物的渗透吸收性能,增强药物在膀胱黏膜的吸收效率,但过高的氟化取代可能不利于材料的应用。
实施例1-6:13FCS的促膀胱灌注药物膀胱黏膜吸收的作用:将实施例1-3中所制备的13FCS和THP水溶液混合,经小鼠尿道灌注到膀胱,后制备小鼠膀胱冰冻切片,通过检测THP荧光在组织中的分布来评估药物载体的促药物膀胱黏膜吸收效率。
具体方法为:将10-12周的雌性C57BL/6小鼠以戊苯巴比妥溶液麻醉,以0.5%的13FCS水溶液配置0.2%的THP水溶液,通过密闭式静脉留置针将其灌注到小鼠膀胱,100μl,夹闭尿道1h,随后放出膀胱内的灌注液,用1ml超纯水冲洗膀胱,取膀胱组织放入组织包埋机中置于-80℃,后进行切片用荧光共聚焦显微镜进行检测。以同样配制的THP壳聚糖水溶液作为对照。
实验结果如图1-2所示FCS组小鼠膀胱纵切面的药物荧光分布面积及强度均显著高于壳聚糖(CS),表明13FCS可显著提高药物在膀胱黏膜的渗透性;同时,13FCS-3的促渗透作用最强,其氟化修饰程度为21.4%。以上结果也表明FCS促灌注药物膀胱黏膜渗透作用可能是壳聚糖阳离子骨架与氟化脂肪链共同作用的结果,当然此种原因也可能是多元化的。
实施例1-7:为了筛选促灌注药物膀胱黏膜渗透效果最好的氟化壳聚糖,对实施例1-4中19FCS-1以及上述各氟化修饰种类中效果最好的FCS进行小鼠体内评价。
具体方法为:将10-12周的雌性C57BL/6小鼠以戊苯巴比妥溶液麻醉,分别以0.5%的7FCS-4,13FCS-3,19FCS-1水溶液配置0.2%的THP溶液,通过密闭式静脉留置针将其灌注到小鼠膀胱,100μl,夹闭尿道1h,随后放出膀胱内的灌注液,用1ml超纯水冲洗膀胱,取膀胱组织放入组织包埋机中置于-80℃,后进行切片用荧光共聚焦显微镜进行检测。以单纯的等浓度THP水溶液或同样配制的THP壳聚糖水溶液作为对照。
实验结果:如图1-3所示荧光共聚焦显微镜对小鼠膀胱组织切片进行观察,结果发现19FCS组小鼠膀胱横切面的药物荧光分布面积及强度均较THP与CS组具有显著差异,但13FCS-3的促灌注药物渗透性作用最为突出。
实施例1-8:对实施例1-7中不同种类氟化修饰氟化壳聚糖进行体外、体内安全性评价,具体实验方案如下:
以CCK-8(Cell Counting Kit-8)法(一种成熟体外评价细胞活性的评价方法)评价氟化壳聚糖对SV-HUC-1人正常膀胱上皮细胞的细胞毒作用考察氟化壳聚糖的体外生物安全性,具体操作如下:以1x104个/孔T24细胞接种到96孔板,37℃,5%CO 2培养过夜,加入不同种类氟化壳聚糖(500ug/ml)的无血清培养基继续培养24h,然后加入适量cck-8,最终以细胞存活率评价氟化壳聚糖的体外安全性。实验结果如图1-4a所示,13F-3(13FCS-3的简称)具有很好的体外细胞安全性,结合上述研究结果表明13F-3具有最为显著的促灌注药物膀胱黏膜吸收的作用,同时具有较好的体外细胞安全性。为了进一步评价FCS(13F-3)的生物安全性,对其进行进一步的小鼠体内安全性评价实验。
将10-12周健康C57BL/6小鼠分为三组,每组8只。实验组以15mg/ml氟化壳聚糖或壳聚糖1%醋酸水溶液灌注1h,每周灌注一次,共三周,空白对照组灌注等体积双蒸水,以小鼠的体重、生存率以及第一次给药后第28天小鼠膀胱切片的HE、免疫组化分析(CD45和Ki67)结果评价氟化壳聚糖的体内生物安全性。
实验结果发现,小鼠在灌注壳聚糖后第二天就出现体重骤降,小鼠活动萎靡的状况并且从治疗后的第二天起小鼠出现死亡,并在陆续的3天内实验组8只小鼠全部死亡,而氟化壳聚糖组小鼠无死亡出现,如图1-4b所示,FCS组小鼠体重与空白对照组无明显差异。同时,如图1-4c所示,取各组灌注后小鼠膀胱与空白对照组小鼠膀胱对比发现,壳聚糖灌注组小鼠膀胱相对于空白组小鼠膀胱充血严重,HE染色以及图1-5中CD45,Ki67免疫荧光结果表明壳聚糖灌注组小鼠膀胱出现了严重得炎症应激与充血水肿,而FCS组与空白对照组无 明显差异。以上实验结果表明氟化壳聚糖(13FCS-3)可显著提高灌注药物在膀胱黏膜的生物利用度,同时高浓度的氟化壳聚糖不会引起明显的膀胱黏膜、上皮损伤,具有作为膀胱灌注药物载体的可能性。
实施例1-9:F-PEI膀胱灌注多肽蛋白药物载体的应用,以多肽药物MPI,以及蛋白药物CAT-Ce6为例
(1)氟化聚醚酰亚胺(F-PEI)的合成
具体操作如下:适量3-(perfluorohex-1-yl)-1,2-propenoxide缓慢滴加到枝化聚醚酰亚胺(PEI)的甲醇溶液中,室温搅拌48h。反应粗产物分别用甲醇/双蒸水透析纯化(MWCO 3500Da),冷冻干燥得终产物。通过1H NMR对产物进行结构鉴定,利用氟元素分析计算分子中的平均氟取代数。
(2)MPI/F-PEI,CAT-Ce6/F-PEI纳米药物体系的制备及表征
通过将多肽(MPI)、蛋白(CAT)药物与F-PEI的水溶液室温混合2h即可得到MPI/F-PEI,CAT/F-PEI NPs。动态光散射仪测得其水合粒径约为200-300nm,带有少量正电荷,透射电子显微镜(TEM)成像(图1-6,1-7)表征其为均一的球形粒子。
(3)MPI/F-PEI NPs,CAT-Ce6/F-PEI NPs的膀胱黏膜渗透性评价
Ussing chamber(尤斯室,也叫尤斯灌流室)是研究跨上皮转运的工具,可用于包括离子转运、营养物质转运及药物转运等的研究。通过跨上皮转运的研究,可以了解上皮的药物透过上皮的吸收。本实例应用Ussing chamber评价以不同物料比制备的MPI-cy5.5/F-PEI NPs,CAT-ce6/F-PEI NPs的黏膜透过性,分别以MPI-cy5.5/PEI NPs,CAT-ce6/PEI NPs作为对照,小鼠麻醉,取膀胱于冰上剥离膀胱黏膜,固定在两小室中间的接口处。扩散室中加入3mlMPI-cy5.5/F-PEI NPs或CAT-ce6/F-PEI NPs的台式液,接收室加入等体积的空白台式液。每隔15分钟从接收室取0.5ml台式液,同时在接受室补充等体积的空白台式液,连续四次并通过荧光分光光度计检测其相应药物含量。实验结果显示F-PEI组多肽(图1-8)或蛋白药物的黏膜渗(图1-9)透指数papp要显著高于PEI组及free组(空白组)。
同时我们也在小鼠体内对其膀胱黏膜渗透性进行了考察。小鼠麻醉后,分别膀胱灌注含有等量荧光标记的多肽或蛋白药物溶液,MPI组不同药物体系(free MPI-cy5.5,MPI-cy5.5/PEI,MPI-cy5.5/F-PEI)在灌注药物不同时间后(15,30,60min)取膀胱制备冰冻切片(图1-10);CAT组不同药物体系(free CAT-ce6,CAT-ce6/PEI,CAT-ce6/F-PEI)灌注1h后取膀胱制备冰冻切片,并通过荧光共聚焦显微镜分析荧光强度(图1-11)。实验结果表明与PEI以及free药物组相比F-PEI组的多肽及蛋白药物具有更加显著的膀胱黏膜渗透性,即F-PEI可显著改善多肽或蛋白药物的膀胱黏膜渗透性。
实施例1-10:FCS膀胱灌注蛋白药物载体的应用,以蛋白药物CAT-TCPP为例。
(1)CAT-TCPP/FCS纳米药物体系的制备及表征
通过将蛋白药物(CAT-TCPP)药物与FCS的水溶液室温混合2h即可得到CAT-TCPP/FCS NPs。动态光散射仪测得其水合粒径约为200-300nm,带有少量正电荷,透射电子显微镜(TEM)成像(图1-12)表征其为均一的球形粒子。
(2)CAT-TCPP/FCS NPs的膀胱黏膜渗透性评价
我们在小鼠体内对其膀胱黏膜渗透性进行了考察。小鼠麻醉后,分别膀胱灌注含有等量荧光标记的蛋白药物溶液,CAT-TCPP组不同药物体系(free CAT-TCPP,CAT-TCPP/ CS,CAT-TCPP/FCS)在灌注药物相同时间后(60min)取膀胱制备冰冻切片(图1-13),并通过荧光共聚焦显微镜分析荧光强度。实验结果表明与壳聚糖(CS)以及free药物组相比FCS组的蛋白药物具有更加显著的膀胱黏膜滞留能力及渗透性,即FCS可显著改善蛋白药物的膀胱黏膜渗透性。
若无特殊标明,本专利所有实施例所使用的全氟庚酸修饰的壳聚糖均为实施例1-3中全氟庚酸与N氨基葡萄糖单元的投料摩尔比例分为1:4.2的氟化壳聚糖。
如下为实施例1-3中氟化壳聚糖结构示意。
Figure PCTCN2020138465-appb-000024
其中,A为含有伯氨基的壳聚糖分子骨架,结构式如下:
Figure PCTCN2020138465-appb-000025
其中,B为含氟功能基团与壳聚糖伯氨基形成的连接基团,此处为酰胺键即
Figure PCTCN2020138465-appb-000026
其中,C为含氟脂肪链、芳香环功能基团;此专利中采用全氟庚酸,结构式如下:
Figure PCTCN2020138465-appb-000027
实施例2-1:制备全氟庚酸修饰的壳聚糖为载体的透皮敷贴,透皮递送胰岛素,进行糖尿病治疗。本实施例的具体氟化壳聚糖制备工艺参见实施例2-5。
具体方法:
1.制备全氟庚酸修饰的壳聚糖-胰岛素复合物:将全氟庚酸修饰的壳聚糖与胰岛素分别溶于弱酸的溶液环境中使其溶解均匀,混匀后在搅拌中滴加弱碱溶液,调节pH为6-7,在中性条件下全氟庚酸修饰的壳聚糖和胰岛素由于静电吸附作用结合在一起,形成稳定的复合物,其中全氟庚酸修饰的壳聚糖与胰岛素优选的反应质量比为1:0.25-4,进一步优选为1:0.5-2。反应充分后取出,预加冻干保护剂后冻干,得到全氟庚酸修饰的壳聚糖-胰岛素冻干粉。通过动态光散射粒径(图2-2)及电位(图2-3)进行分析。
如图2-2所示,不同比例的全氟庚酸修饰的壳聚糖与胰岛素反应均可生成复合物。全氟庚酸修饰的壳聚糖与胰岛素的质量比为1:0.5-2时,粒径较为均匀。
如图2-3所示,全氟庚酸修饰的壳聚糖与胰岛素的不同比例影响着复合物整体的电位。
2.体外透皮动力学分析:以荧光标记的胰岛素代替步骤1中的胰岛素,对制备获得 的不同比例的全氟庚酸修饰的壳聚糖-胰岛素(以下简称FCS-Insulin)进行优选及透皮动力学分析。首先合成不同质量比的全氟庚酸修饰的壳聚糖-胰岛素并放入Franz立式扩散池的进样池中,而后通过检测通过夹层小鼠皮肤进入取样池的全氟庚酸修饰的壳聚糖-胰岛素的荧光强度来表征其在不同时间点的透皮效果。其中全氟庚酸修饰的壳聚糖-胰岛素的比例分别为1:0.25,1:0.5,1:1,1:2,1:4以及单纯的免疫球蛋白G(0:1)。横坐标为透过时间,纵坐标为根据荧光计算出的累计透过量。结果如图2-4所示,优选比例为1:1。
3.制备全氟庚酸修饰的壳聚糖-胰岛素复合物透皮敷贴:复溶全氟庚酸修饰的壳聚糖-胰岛素冻干粉,加入提前制备好的水凝胶基质并混匀,得到全氟庚酸修饰的壳聚糖-胰岛素复合物透皮敷贴如图2-5左所示。对凝胶进行扫描电镜表征如图2-5右所示。
4.测量药物从凝胶中的释放:复溶全氟庚酸修饰的壳聚糖-胰岛素冻干粉,加入提前制备好的不同浓度的水凝胶基质并混匀,得到全氟庚酸修饰的壳聚糖-胰岛素复合物透皮敷贴。凝胶的浓度影响着药物从凝胶中的释放行为,将凝胶敷贴浸入缓冲溶液中,在不同时间点取溶液上清,根据考马斯亮蓝染色计算出胰岛素累计透过量如图2-6所示。
其中横坐标为时间,纵坐标为根据考马斯亮蓝染色计算出的胰岛素累计透过量。
5.通过小鼠血糖变化对载药透皮敷贴的促渗透作用进行评价:将10-12周的雌性C57BL/6小鼠以异氟烷麻醉,将载药透皮敷贴贴到小鼠背部的脱毛皮肤上,并用弹力绷带进行固定,随后在不同时间点测量小鼠血糖波动,以空白敷贴作为对照。图2-7为小鼠血糖波动情况,横坐标为贴上敷贴后的作用时间,纵坐标为血糖浓度。
如图2-7所示,通过血糖仪对小鼠血糖进行监测,结果发现与空白对照组相比,贴有全氟庚酸修饰的壳聚糖载药透皮敷贴的小鼠其高血糖得到明显抑制,并在长时间内保持稳定,这表明全氟庚酸修饰的壳聚糖可以显著提高药物在皮肤的渗透性,并促进药物进入血液维持稳定的血药浓度,持续发挥作用。以上结果共同表明,含氟化合物修饰的阳离子聚合物可以成功实现药物的透皮递送,具备较大医学价值及转化价值。
实施例2-2:制备全氟庚酸修饰的壳聚糖为载体的透皮软膏,透皮递送细胞程式死亡-配体1抗体,进行体表黑色素瘤治疗。
具体方法:
1.制备全氟庚酸修饰的壳聚糖-免疫球蛋白G复合物(以下简称FCS-IgG):由于免疫球蛋白G与细胞程式死亡-配体1抗体具有相同的结构,在材料水平以免疫球蛋白G为模板研究全氟庚酸修饰的壳聚糖与细胞程式死亡-配体1抗体结合的优选比例。将不同量的免疫球蛋白G加入全氟庚酸修饰的壳聚糖水溶液中,常温搅拌1小时,形成稳定的复合物。其中,全氟庚酸修饰的壳聚糖与免疫球蛋白G的反应质量比为1:0.25-4,并通过动态光散射粒径分析及电位分析,并进一步优选为1:1。粒径分布与电位分布结果如图2-8所示。
免疫球蛋白是指具有抗体活性或化学结构,与抗体分子类似的球蛋白。本专利实验中采用的免疫球蛋白G是没有特异性的,抗体是能与抗原特异性结合的免疫球蛋白,细胞程式死亡-配体1抗体也是免疫球蛋白G的一种,只是轻链端具有特异性,所以在非治疗实验中可以用免疫球蛋白G来模拟细胞程式死亡-配体1抗体的行为。
实验结果:参见图2-8,左图为不同比例的全氟庚酸修饰的壳聚糖-免疫球蛋白G在水溶液中的粒径分布,右图为电位分布。其中优选具有较好的粒径并且保持较高的正电的1:1组。
2.制备全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体透皮软膏:按照相同方法,获得全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体复合物。将获得的全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体水溶液与空白软膏以1:1的质量比混合形成全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体透皮软膏,其中软膏的主要成分为凡士林。
3.全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体体外透皮动力学分析:以荧光标记的免疫球蛋白G为模板模拟细胞程式死亡-配体1抗体的透皮动力学,对制备获得的全氟庚酸修饰的壳聚糖-免疫球蛋白G(以下简称FCS-IgG)进行优选及透皮动力学分析。首先合成不同质量比的全氟庚酸修饰的壳聚糖-免疫球蛋白G并放入franz立式扩散池的进样池中,而后通过检测通过夹层小鼠皮肤进入取样池的全氟庚酸修饰的壳聚糖-免疫球蛋白G的荧光强度来表征其在不同时间点的透皮效果。其中全氟庚酸修饰的壳聚糖-免疫球蛋白G的比例分别为1:0.25,1:0.5,1:1,1:2,1:4以及单纯的免疫球蛋白G(0:1)。每组均在上样后2小时,4小时,8小时,12小时及24小时取样,计算累积透过量。结果如图2-9所示。
实验结果:参见图2-9,不同比例的全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)在不同时间点的累计透过量。其中横坐标为进样时间,纵坐标为累计透过量除以总进样量的透过率。与单纯的大分子蛋白免疫球蛋白G相比,不同比例的全氟庚酸修饰的壳聚糖-免疫球蛋白G均可以不同程度的透过小鼠皮肤。优选得到全氟庚酸修饰的壳聚糖-免疫球蛋白G比例为1:1的复合物,并用于后续实验。
4.全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体体内透皮动力学分析:以荧光标记的免疫球蛋白G为模板模拟细胞程式死亡-配体1抗体的透皮动力学,制备获得的全氟庚酸修饰的壳聚糖-免疫球蛋白G(以下简称FCS-IgG)进行活体透皮动力学分析。首先在C57小鼠皮下注射B16黑色素细胞悬液(约1*10^6个/只),在肿瘤体积约为60立方毫米时,在小鼠肿瘤表面涂抹质量比1:1混合的全氟庚酸修饰的壳聚糖-免疫球蛋白G软膏,并用透皮贴膜固定防止掉落,在不同时间点处死小鼠,取下肿瘤组织,擦干净表面剩余的软膏并取下表皮,将取下后的肿瘤组织一分为二,一半裂解后测量组织内的荧光强度,另一半切片后在共聚焦显微镜下进行荧光成像。结果如图2-10所示。
实验结果:参见图2-10,分别为在全氟庚酸修饰的壳聚糖-免疫球蛋白G软膏在0h,4h,8h,12h及24h时解剖的肿瘤组织,通过共聚焦荧光显微镜的荧光成像图以及裂解后肿瘤内荧光含量。可以明显发现软膏的透过率在12小时达到峰值,到24小时开始下降,这可能是由于进入肿瘤内的全氟庚酸修饰的壳聚糖-免疫球蛋白G被降解导致。
5.全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体体内透皮效率对比:通过动力学分析,优选软膏涂抹12小时的时间点,进行单纯细胞程式死亡-配体1抗体,壳聚糖-细胞程式死亡-配体1抗体以及全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体的透皮效率比较。在荷瘤小鼠的肿瘤部位涂抹相同量的行单纯细胞程式死亡-配体1抗体,壳聚糖-细胞程式死亡-配体1抗体以及全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体,并用透明贴膜固定。在12小时后处死小鼠取下肿瘤组织并进行免疫荧光染色及共聚焦显微镜荧光成像。结果如图2-4所述。
实验结果:参见图2-11。从左到右分别是DAPI,FITC以及综合通道荧光,其中DAPI为细胞核染料,可以表明细胞核,FITC为细胞程式死亡-配体1抗体的带荧光二抗标记。可以按出全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体组在同样剂量的细胞程式死亡-配 体1抗体下,具有最强的透皮效率。
7.全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体透皮机制研究:以免疫球蛋白G为模板模拟细胞程式死亡-配体1抗体进行全氟庚酸修饰的壳聚糖-免疫球蛋白G的透皮机制分析。首先是全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)对于致密的细胞单层的透皮效果验证。将人皮肤上皮细胞Hacat孵育在如图2-12所示的Transwell孔板中,每隔1天使用细胞电阻检测仪检测其电阻变化,直到生长至平台期,电阻不再变化开始,加入全氟庚酸修饰的壳聚糖-免疫球蛋白G,并继续检测其电阻变化。结果如图2-5所示。
实验结果:参见图2-12。在第10天观测到致密细胞单层形成,并在第11天加入FCS-IgG,24小时后检测电阻变化,可见电阻骤减,表明致密细胞单层被打开,随后可见电阻缓慢恢复至平台期,表明材料对于打开细胞间的紧密连接是暂时性的。
8.全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体透皮机制研究:以免疫球蛋白G为模板模拟细胞程式死亡-配体1抗体进一步分析调控细胞间紧密连接的相关蛋白在加入全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)之后的变化。将人皮肤上皮细胞Hacat孵育在共聚焦显微镜专用的培养皿中,等待其生长为致密的细胞单层,随后,在加入FCS-IgG的24小时后,对培养皿中细胞进行免疫荧光染色,分别检测紧密连接相关蛋白:闭合蛋白(Occludin)、跨膜整合蛋白(Claudin-1)、钙粘附蛋白E(E-Cadherin)以及紧密连接蛋白-1(ZO-1)。结果如图2-13所示。
实验结果:参见图2-13。在加入FCS-IgG后,各组蛋白荧光含量(图中白色细胞周围的白色条带)均有明显下降。表明FCS-IgG通过改变细胞紧密连接蛋白在细胞膜表明的分布来打开细胞紧密连接从而穿过细胞间隙透过皮肤。
9、全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体透皮机制研究:以免疫球蛋白G为模板模拟细胞程式死亡-配体1抗体进一步研究紧密连接蛋白分布改变的机制。将人皮肤上皮细胞Hacat孵育在25mm2的培养皿中,等待其生长为致密的细胞单层,随后,将细胞裂解后,取出细胞全蛋白进行蛋白质印迹法(Western Blotting)测定。首先检测了4种紧密连接蛋白的含量变化,随后检测了肌动蛋白的磷酸化水平变化。其中紧密连接蛋白为闭合蛋白(Occludin)、跨膜整合蛋白(Claudin-1)、钙粘附蛋白E(E-Cadherin)以及紧密连接蛋白-1(ZO-1);肌动蛋白为MLC,磷酸化的肌动蛋白为p-MLC;GAPDH为甘油醛-3-磷酸脱氢酶,该酶在各个组织中的表达量相对稳定,此处用作内参。结果如图2-14所示。
实验结果:参见图2-14,左图为4种紧密连接蛋白的含量变化,可见加入FCS-IgG后,蛋白含量无明显改变,表明FCS-IgG仅影响了紧密连接蛋白在细胞膜表明的分布,而非降低了其表达,进一步说明这种影响只是暂时性的。右图为肌动蛋白磷酸化水平变化,可见肌动蛋白有明显磷酸化,表明FCS-IgG通过刺激肌动蛋白磷酸化来刺激细胞的旁运,从而进一步打开细胞间隙,促进材料穿过细胞间隙透过皮肤。
10.全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体活体皮下肿瘤治疗:将C57小鼠平均分为4组,分别为空白组,单独细胞程式死亡-配体1抗体经脉注射组,壳聚糖-细胞程式死亡-配体1抗体以及全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体组,在小鼠皮下注射B16细胞悬液(约1*10^6个/只),每天监控肿瘤大小,肿瘤体积通过以下公式计算获得:体积=0.5*肿瘤长*肿瘤宽^2。在起始肿瘤大小约为10立方毫米时开始涂软膏或经脉给药治疗,每隔一天进行一次治疗,共治疗4次,并且每隔一天记录肿瘤大小。结果如图2-8所 示。
实验结果:参见图2-15。其中左图为小鼠肿瘤生长曲线图,右图为小鼠生存率折线图,以肿瘤大小至1500立方毫米作为小鼠死亡的标准。可以发现由于起始体积较小,临床常规使用的经脉注射疗法对于小鼠肿瘤也有一定的抑制作用,但相比之下,全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体组由于具有超过50%的抗体透过率,治疗效果大大高于其他组。
本专利中含氟化合物修饰的阳离子聚合物,尤其是氟化壳聚糖涉及的药物包括但不限于糖尿病治疗药物、抗肿瘤药物(详见如下表2-1)、免疫调节剂、抗病毒药物、抗炎药物、镇痛麻醉药物、医疗美容药物等各种药物及其衍生物的各类剂型。
Figure PCTCN2020138465-appb-000028
Figure PCTCN2020138465-appb-000029
表2-1
其中所述药物可以是免疫调节剂,包括但不限于细胞因子、卡介苗、免疫检查点阻断抗体等。细胞因子是由免疫细胞(如单核、巨噬细胞、T细胞、B细胞、NK细胞等)和某些非免疫细胞(内皮细胞、表皮细胞、纤维母细胞等)经刺激而合成、分泌的一类具有广泛生物学活性的小分子蛋白质。细胞因子包括但不限于白细胞介素(interleukin,IL)、干扰素(interferon,IFN)、肿瘤坏死因子超家族(tumor necrosis factor,TNF)、集落刺激因子(colony stimulating factor,CSF)、趋化因子(chemokine family)、生长因子(growth factor,GF)、转化生长因子-β家族(transforming growth factor-βfamily,TGF-βfamily)。白细胞介素包括但不限于IL-1―IL-38。集落刺激因子包括但不限于G(粒细胞)-CSF、M(巨噬细胞)-CSF、GM(粒细胞、巨噬细胞)-CSF、Multi(多重)-CSF(IL-3)、SCF、EPO等。干扰素包括但不限于包括但不限于IFN-α、IFN-β和IFN-γ。肿瘤坏死因子包括但不限于TNF-α和TNF-β。转化生长因子-β家族包括但不限于TGF-β1、TGF-β2、TGF-β3、TGFβ1β2以及骨形成蛋白(BMP)。生长因子包括但不限于表皮生长因子(EGF)、血小板衍生的生长因子(PDGF)、成纤维细胞生长因子(FGF)、肝细胞生长因子(HGF)、胰岛素样生长因子-I(IGF-1)、IGF-Ⅱ、白血病抑制因子(LIF)、神经生长因子(NGF)、抑瘤素M(OSM)、血小板衍生的内皮细胞生长因子(PDECGF)、转化生长因子-α(TGF-α)、血管内皮细胞生长因子(VEGF)。趋化因子家族包括但不限于四个亚族:(1)C-X-C/α亚族,主要趋化中性粒细胞,主要的成员有IL-8、黑素瘤细胞生长刺激活性(GRO/MGSA)、血小板因子-4(PF-4)、血小板碱性蛋白、蛋白水解来源的产物CTAP-III和β-thromboglobulin、炎症蛋白10(IP-10)、ENA-78;(2)C-C/β亚族,主要趋化单核细胞,这个亚族的成员包括巨噬细胞炎症蛋白1α(MIP-1α)、MIP-1β、RANTES、单核细胞趋化蛋白-1(MCP-1/MCAF)、MCP-2、MCP-3和I-309。(3)C型亚家族的代表有 淋巴细胞趋化蛋白。(4)CX3C亚家族,Fractalkine是CX3C型趋化因子,对单核-巨噬细胞、T细胞及NK细胞有趋化作用。
细胞因子包括但不限于用于治疗癌症的细胞因子和减轻癌症治疗副作用的细胞因子,它们在人体正常的免疫反应以及免疫系统对癌症的反应能力中起重要作用。用于治疗癌症的细胞因子包括但不限于干扰素、白介素。细胞因子还可以是造血生长因子,通过促进受化疗破坏的血细胞生长来减少癌症治疗的副作用。减少癌症治疗副作用的细胞因子包括但不限于促红细胞生成素、IL-11、粒细胞-巨噬细胞集落刺激因子(GM-CSF)和粒细胞-集落刺激因子(G-CSF)。卡介苗(BCG Vaccine)是由减毒牛型结核杆菌悬浮液制成的活菌苗,可以增强巨噬细胞活性,增强机体细胞免疫的功能,可以用于治疗皮肤癌等。免疫调节药物包括但不限于沙利度胺
Figure PCTCN2020138465-appb-000030
来那度胺
Figure PCTCN2020138465-appb-000031
泊马利度
Figure PCTCN2020138465-appb-000032
咪喹莫特
Figure PCTCN2020138465-appb-000033
免疫检查点阻断抗体包括但不限于CTLA4单克隆抗体
Figure PCTCN2020138465-appb-000034
PD-1单克隆抗体
Figure PCTCN2020138465-appb-000035
Figure PCTCN2020138465-appb-000036
PD-L1单克隆抗体
Figure PCTCN2020138465-appb-000037
Figure PCTCN2020138465-appb-000038
LAG-3(淋巴细胞激活基因3)单克隆抗体、TIM-3(T细胞免疫球蛋白和粘蛋白结构域蛋白3)单克隆抗体、TIGIT(T细胞免疫球蛋白和ITIM结构域蛋白)单克隆抗体、协同刺激因子B7-H3、B7-H4和B7-H5单克隆抗体等药物及其衍生物。
所述药物可以是麻醉药物。示例全身麻醉包括但不限于盐酸氯胺酮,丙泊酚,硫喷妥钠,依托咪脂,咪达唑仑和γ-羟基丁酸钠。局部麻醉药包括但不限于芳酸酯类,芳酰胺类,氨基酮类,氨基醚类,氨基甲酸酯类,羟普鲁卡因,氯普鲁卡因,丁卡因,布他卡因,硫卡因,普鲁卡因胺,布比卡因,阿替卡因,依替卡因,罗哌卡因,甲哌卡因,克罗宁等药物及其衍生物。
所述药物可以是糖尿病治疗药物,包括但不限于氨苯磺丁脲、甲苯磺丁脲、氯磺丙脲、乙酸已脲、格列齐特、格列吡啶、格列美脲等磺酰脲类药物及其衍生物;瑞格列奈、那格列奈等非磺酰脲类药物及其衍生物;罗格列酮、吡咯列酮等噻唑烷二酮类药物及其衍生物;苯乙双胍、二甲双胍等二胍类药物及其衍生物;阿卡波糖、伏格列波糖、米格列醇等α-葡萄糖苷酶抑制剂药物及其衍生物;胰高血糖素样肽,DPP-Ⅳ抑制剂西格列汀、维格列汀、沙格列汀二肽基肽酶-Ⅳ药物及其衍生物以及胰岛素等药物及其衍生物。
糖尿病是以高血糖为主要特征的代谢内分泌性疾病,临床使用剂型一般为胰岛素注射剂。患者需要忍受反复注射的治疗痛苦,长期用药还会引发注射部位的炎症和硬结等副作用。含氟化合物修饰的壳聚糖药物可以透过皮肤,携带降糖药物进入血液,提高药物的生物利用度。如实施例2-1所述,含氟化合物修饰的壳聚糖可以作为药物载体递送降糖药物,以药物敷贴的方式进行给药,用于糖尿病治疗。通过含氟化合物修饰的壳聚糖药物敷贴以透皮方式进行给药,药物的有效浓度维持时间长,作用程度与维持时间可以根据敷贴的面积和敷贴时间来进行调节,具有灵活、方便的优点。除此之外,还可以制备成更灵活的洗剂、擦剂、涂抹剂等剂型。
所述药物可以是抗肿瘤药物(详见表2-1)。透皮给药作为一种非侵入性的给药方式,虽然带来了很大的便捷,但皮肤的角质层屏障往往阻碍药物进入皮下病灶甚至进入血管。黑色素瘤是起源于能制造黑色素的细胞的恶性肿瘤,其易转移、耐药性强、预后差、死亡 率极高。黑色素瘤化疗药物主要通过口服和注射的方式递送,但这往往引起诸多不良反应,甚至导致器官损伤,同时无法高效、精准、可控地递送药物。透皮递送方式对于皮下黑色素瘤的治疗有着得天独厚的优势,但仍对透皮递送的效率提出了更高的要求。如b2-2所述,含氟化合物修饰的壳聚糖可以作为药物载体递送抗肿瘤药物,以软膏的方式进行给药,用于肿瘤治疗。
本专利2-1至2-6各个实施例中的氟化壳聚糖,均可以用作透皮制剂,用于作为治疗糖尿病药物、肿瘤疾病药物、消炎药物的透皮给药制剂。同时,也可以作为透皮给药制剂在医疗美容药物、药物外用制剂、医疗器械外用制剂、化妆护肤品制备中进行应用。
实施例3-1:制备全氟庚酸修饰的壳聚糖为载体的口服药物,口服递送胰岛素,进行糖尿病治疗。
具体方法:
1.制备全氟庚酸修饰的壳聚糖-胰岛素胶囊:将全氟庚酸修饰的壳聚糖与胰岛素分别溶于弱酸溶液中使其溶解均匀,混匀后在搅拌中滴加弱碱溶液,调节pH为6-7,在中性条件下全氟庚酸修饰的壳聚糖和胰岛素由于静电吸附作用结合在一起,形成稳定的纳米颗粒。反应充分后取出,预加冻干保护剂后冻干,得到全氟庚酸修饰的壳聚糖-胰岛素冻干粉,冻干粉装入胶囊中并以肠衣包裹。
2.复溶全氟庚酸修饰的壳聚糖-胰岛素冻干粉,通过动态光散射粒径(图3-3)及电位(图3-4)进行分析。
3.对冻干前后的全氟庚酸修饰的壳聚糖-胰岛素进行动态光散射粒径分析,如图3-5所示,冻干前后没有明显变化。
4.体外透皮动力学分析:以荧光标记的胰岛素代替步骤1中的胰岛素,对制备获得的不同比例的全氟庚酸修饰的壳聚糖-胰岛素(以下简称FCS-Insulin)进行优选及透皮动力学分析。首先合成不同质量比的全氟庚酸修饰的壳聚糖-胰岛素并放入Franz立式扩散池的进样池中,而后通过检测通过夹层小鼠黏膜进入取样池的全氟庚酸修饰的壳聚糖-胰岛素的荧光强度来表征其在不同时间点的透皮效果。结果如图3-6所示,优选比例为1:0.5-2。
5.将载药胶囊放入模拟胃液或模拟肠液中,通过溶液中释放出的全氟庚酸修饰的壳聚糖-胰岛素的荧光强度来表征其在模拟胃液或模拟肠液中的释放效果。如图3-7所示,其中左侧为模拟胃液中的释放行为,右侧为肠液中的释放行为。胶囊在模拟胃液中可以长时间保持稳定不释放,而在模拟肠液中实现药物的释放。
6.通过小鼠血糖变化对给药效果进行评价:灌服全氟庚酸修饰的壳聚糖-胰岛素胶囊,随后在不同时间点测量小鼠血糖波动,以空白胶囊作为对照。
参见图3-8为小鼠血糖波动情况,横坐标为灌服胶囊后的时间,纵坐标为血糖浓度。
如图3-8所示,通过血糖仪对小鼠血糖进行监测,结果发现与空白对照组相比,灌服全氟庚酸修饰的壳聚糖-胰岛素胶囊的小鼠其高血糖得到明显抑制,并在长时间内保持稳定,这表明全氟庚酸修饰的壳聚糖可以显著提高药物在肠道的滞留和渗透,并促进药物进入血液维持稳定的血药浓度,持续发挥作用。以上结果共同表明,含氟化合物修饰的阳离子聚合物可以成功实现药物的口服递送,具备良好的医用转化价值。
实施例3-2:制备全氟庚酸修饰的壳聚糖为载体的口服胶囊,口服递送细胞程式死亡-配体1抗体,观测不同比例的全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体颗粒的穿粘膜效果。
具体方法:
1.制备全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体颗粒:称取0.2mg全氟庚酸修饰的壳聚糖用0.5mL超纯水溶解得到0.4mg/mL的全氟庚酸修饰的壳聚糖,并梯度稀释,最后得到0.4mg/mL,0.2mg/mL,0.1mg/mL,0.05mg/mL,0.0025mg/mL的全氟庚酸修饰的壳聚糖溶液。以荧光标记的免疫球蛋白G为模板代替细胞程式死亡-配体1抗体,在不断搅拌的情况下滴入0.5mL 0.1mg/mL的FITC标记的的免疫球蛋白G的0.02M pH=7.2的磷酸钾缓冲液,并在室温搅拌30min。
2.全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体体外透肠粘膜动力学分析:取大鼠肠道,去除筋膜层,并固定在透皮扩散池上面。在透皮扩散池上面加入不同质量比的全氟庚酸修饰的壳聚糖-FITC-免疫球蛋白G颗粒,在透皮扩散池下面加入PBS溶液,在0min,15min,30min,45min,60min,90min,120min,150min,180min,210min取透皮扩散池下面液体,测荧光强度,并计算累计透过率。结果如3-9所示。
实验结果:参见图3-9,不同比例的全氟庚酸修饰的壳聚糖-免疫球蛋白G(FCS-IgG)在不同时间点的累计透过量。其中横坐标为进样时间,纵坐标为累计透过量除以总进样量的透过率。与单纯的大分子蛋白免疫球蛋白G相比,不同比例的全氟庚酸修饰的壳聚糖-免疫球蛋白G均可以不同程度的透过小鼠皮肤。优选得到全氟庚酸修饰的壳聚糖-免疫球蛋白G比例为1:1的纳米颗粒,并用于后续实验。
3.全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体颗粒冻干粉制备:称取0.1mg全氟庚酸修饰的壳聚糖用1mL超纯水溶解得到0.1mg/mL的全氟庚酸修饰的壳聚糖,0.1mg/mL的全氟庚酸修饰的壳聚糖溶液。,在不断搅拌的情况下滴入0.5mL 0.1mg/mL的cy5.5标记的的免疫球蛋白G的0.02M pH=7.2的磷酸钾缓冲液,并在室温搅拌30min。搅拌结束之后加入冻干保护剂,放在-20摄氏度冰箱预冷,然后放到冷冻干燥机干燥。
4.全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体胶囊制备及其定点释放性能:称取一定量冻干粉装填于小鼠专用胶囊,并用尤特齐s100包衣进行包衣。用投药器给老鼠饲喂上述胶囊,在第3小时,第5小时处死小鼠,取出肠道,用小动物成像拍摄肠道部位cy5.5荧光分布,从而确定胶囊分布。
实验结果:参见图3-10,在第3小时,第5小时可以看到在结直肠部位观测到荧光,说明该胶囊能定点到结直肠释放。从而选用该胶囊进行后续实验。
5、全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体颗粒冻干粉冻干前后活性测定:用碳酸盐缓冲液将冻存细胞程式死亡-配体1稀释到不同浓度,并加入到酶联免疫吸附法酶标板中,放置于4℃过夜。包被结束之后用PBST洗涤三遍,并加入BSA溶液,室温封闭2h。封闭结束之后,用PBST洗涤三遍,加入冻干前后细胞程式死亡-配体1抗体,室温孵育两小时。孵育结束之后用PBST洗涤三遍,加入辣根过氧化物酶标记的羊抗大鼠二抗,室温孵育一小时,孵育结束之后加入TMB显色剂,之后再加入终止液停止显色。在OD450处吸收酶联免疫吸附法酶标板的吸光度,从而计算出冻干前后抗体亲和力。
实验结果:参见图3-11,加入冻干保护剂冻干之后细胞程式死亡-配体1抗体变成 冻干粉之后活力不变。说明全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体颗粒可以进行冻干,可以用于口服胶囊装填。
6、冻干前后FCS/a PD-L1和CT-26细胞表面PD-L1结合活性
(1)、取10mL 1640培养基往其中加入500ng IFN-γ作为CT-26细胞培养基,和CT-26细胞共孵育36h。
(2)、用细胞挂刀将贴壁CT-26刮下来,用FACS buffer(50mL PBS+0.5mL血清)洗涤三次,倒去上清。
(3)、往每个1.5mL离心管里加1*10 6个细胞并加入FCS/a PD-L1,其中a PD-L1含量为1μg,室温孵育0.5h,倒去上清,并用FACS buffer洗涤三遍,再倒去上清。
(4)、每个1.5mL离心管中加入0.5μL PE anti PD-L1antibody室温孵育30min,倒上清,用FACs buffer洗涤3次。最后加入200μL FACS buffer。
(5)、用流式细胞仪测试冻干之后FCS/a PD-L1与CT-26细胞表面PD-L1结合能力。结果如图3-13所示。
实验结果参见图3-12,可以看到冻干之后FCS/a PD-L1与CT-26细胞表面PD-L1结合能力与冻干前FCS/a PD-L1结合活力相差不大。FCS/a PD-L1可以用于进一步制作胶囊。
7.全氟庚酸修饰的壳聚糖和全氟庚酸修饰的壳聚糖/免疫球蛋白G处理前后人结直肠癌上皮细胞紧密连接蛋白变化。
(1)、配置0.1mg/mL的FCS/IgG,FCS的MEN无血清培养基溶液,放在培养有CaCO-2单层细胞的共聚焦小皿中,并在37℃恒温培养箱孵育5h。
(2)、除去培养液,用PBS洗涤三次,每次5分钟。
(3)、每孔加入4%多聚甲醛溶液,冰上固定20分钟,用PBS洗涤三次,每次5分钟。
(4)、每孔加入0.1%Triton x-100溶液,放置15分钟,用PBS洗涤三次,每次5分钟。
(5)、每个小皿加入2%BSA溶液,室温封闭1h。
(6)、按照1:200加入ZO-1抗体溶液或者1:1000加入E-Caclherin抗体,4摄氏度过夜,取出之后用PBS洗涤三次,每次5分钟。
(7)、每孔加入FITC Goat anti rabbit,室温孵育1h,用PBS洗三遍。
(8)、加入DAPI溶液孵育5分钟,PBS洗涤三次,每次5分钟。
(9)、用共聚焦显微镜拍照,实验结果如图3-13所示。
实验结果参加图3-13.可以看到加入FCS/IgG之后,细胞紧密连接蛋白变松散,细胞骨架重排。
8.全氟庚酸修饰的壳聚糖/细胞程式死亡-配体1抗体治疗小鼠结直肠癌
(1)、称取1mg全氟庚酸修饰的壳聚糖用1mL超纯水溶解得到1mg/mL的全氟庚酸修饰的壳聚糖溶液。,在不断搅拌的情况下滴入1mL 1mg/mL的细胞程式死亡-配体1抗体,并在室温搅拌30min。搅拌结束之后加入冻干保护剂,放在-20摄氏度冰箱预冷,然后放到冷冻干燥机干燥。
(2)、称取冻干后的粉末装填与小鼠专用胶囊,并用尤特齐S100进行包衣。准备好的胶囊备用。
(3)、用1%的戊巴比妥钠麻醉Balb/c小鼠。将小鼠腹部朝上并固定,在腹部右侧开一个小口,取出盲肠,在盲肠壁上注射500000个转染有荧光素酶的CT-26细胞。之后放回盲 肠,缝合伤口,四天之后开始治疗。
(4)、用投药器给小鼠投喂步骤2准备的胶囊,之后再罐100μL的盐酸甲氧氯普胺促进小鼠胃排空。在再种下肿瘤的第4,7,12,16天投喂胶囊。
(5)、投药之后第三天,用1%的戊巴比妥钠麻醉Balb/c小鼠,给每只小鼠注射生物发光底物,十分钟之后成像,观测小鼠肿瘤生长情况。实验结果如图3-14所示。
实验结果如图3-14所示,可以看到用装有全氟庚酸修饰的壳聚糖细胞程式死亡-配体1抗体胶囊治疗之后,能明显压住肿瘤长势。这表明全氟庚酸修饰的壳聚糖可以显著提高药物在结肠的渗透,并促进药物进入肿瘤组织,持续发挥作用。
9:不同分子量全氟庚酸修饰的壳聚糖/细胞程式死亡-配体1抗体的透肠粘膜效果
(1)、制备不同全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体颗粒:称取分子量为10Kda,50Kda,100Kda,300Kda,400Kda的全氟庚酸修饰的壳聚糖用超纯水溶解得到0.4mg/mL的全氟庚酸修饰的壳聚糖,以荧光标记的免疫球蛋白G为模板代替细胞程式死亡-配体1抗体,在不断搅拌的情况下滴入0.5mL 0.1mg/mL的FITC标记的的免疫球蛋白G的0.02M pH=7.2的磷酸钾缓冲液,并在室温搅拌30min。
(2)、全氟庚酸修饰的壳聚糖-细胞程式死亡-配体1抗体体外透肠粘膜动力学分析:取大鼠肠道,去除筋膜层,并固定在透皮扩散池上面。在透皮扩散池上面加入不同质量比的全氟庚酸修饰的壳聚糖-FITC-免疫球蛋白G颗粒,在透皮扩散池下面加入PBS溶液,在0min,15min,30min,45min,60min,90min,120min,150min,180min,210min取透皮扩散池下面液体,测荧光强度,并计算累计透过率。结果如图2-7所示。
如图3-15所示,不同分子量的全氟庚酸修饰的壳聚糖具有不同的增加药物的透粘膜效果。这表明全氟庚酸修饰的壳聚糖能促进药物进入肿瘤组织。
实施例3-3:以全氟庚酸修饰的壳聚糖为载体制备全氟庚酸修饰的壳聚糖-免疫调节剂颗粒,以喷雾剂/吸入剂的方式给药,考察药物肺部递送的能力。
具体方法:
1.制备全氟庚酸修饰的壳聚糖-抗体颗粒:将细胞程式死亡-配体1抗体加入全氟庚酸修饰的壳聚糖水溶液中,常温搅拌1小时,形成稳定的纳米颗粒。
2.通过肺部给药针以气溶胶递送全氟庚酸修饰的壳聚糖-抗体颗粒,递送后24h进行组织切片,观察荧光信号在肺部组织的滞留。结果如图3-16所示。
实验结果参见图3-16,全氟庚酸修饰的壳聚糖-抗体颗粒可以在肺部停留。
实施例3-4:以标有Cy5.5荧光分子的牛血清白蛋白(BSA-Cy5.5)为模型蛋白,以全氟庚酸修饰的壳聚糖(FCS)为透鼻黏膜载体,制备全氟庚酸修饰的壳聚糖-牛血清白蛋白纳米颗粒,通过滴鼻给药的方式,通过Cy5.5的荧光信号考察BSA在脑胶质瘤小鼠模型中经鼻到脑的递送效率。
具体方法:
1.制备FCS/BSA-Cy5.5纳米颗粒:将全氟庚酸修饰的壳聚糖溶于1%醋酸,待其完全溶解后,滴加弱碱调节溶液pH为6-7,再将其与BSA-Cy5.5溶液混匀,并置于4℃下搅拌反应1小时,使其形成稳定的纳米颗粒作为滴鼻剂。其中,全氟庚酸修饰的壳聚糖与BSA优选的反应质量比为1:1,最终牛血清白蛋白浓度为2.5mg/mL。
2.构建脑胶质瘤模型:对7-8周的雄性C57BL/6小鼠(约20克/只)脑内注射表达 EGFP的脑胶质瘤细胞(5000细胞/只),构建脑胶质瘤模型。
3.滴鼻给药:将建模成功的C57BL/6小鼠麻醉,以仰卧的姿势固定于加热垫上,用移液器缓慢向小鼠鼻孔中滴加总体积为20μL的FCS/BSA-Cy5.5溶液(游离BSA-Cy5.5溶液为对照组),滴加完毕后,继续保持小鼠仰卧姿势30min。
4.滴鼻72h之后,麻醉小鼠,进行福尔马林灌注,随后取出脑组织,冻干,切片,并通过共聚焦显微镜观小鼠脑内肿瘤组织处的Cy5.5的荧光信号。
实验结果,参见图3-17,与对照组游离BSA-Cy5.5与CS/BSA-Cy5.5相比,实验组FCS/BSA-Cy5.5在各个时间点的小鼠脑部荧光强度均最佳,说明FCS辅助BSA穿鼻黏膜递送到脑的效果最佳。
参见图3-18,左图为代表小鼠脑组织内脑肿瘤的EGFP信号,右图为代表经鼻向脑递送的BSA的Cy5.5信号。通过右图结果可以看出:与对照组游离BSA-Cy5.5与CS/BSA-Cy5.5相比,实验组FCS/BSA-Cy5.5在滴鼻操作后取出的完整小鼠脑组织的Cy5.5荧光强度最高,说明FCS/BSA-Cy5.5的鼻脑递送效率最好。
参见图3-19,EGFP代表肿瘤组织,Cy5.5代表IgG。通过对比Cy5.5信号可以看出FC S组脑肿瘤部位的Cy5.5信号最强,说明FCS对BSA的鼻脑递送效率最高。
实施例4-1:以标有Cy5.5荧光物质的牛血清白蛋白以及小分子荧光物质罗丹明B为例,利用含氟修饰的壳聚糖包载药物,观测眼部穿透时长与穿透深度。
具体方法:
1.摘取实验兔眼睛进行解剖,分离兔眼角膜,将其固定在Franz扩散池上,与游离蛋白比较,监测蛋白与不同比例的全氟庚酸修饰的壳聚糖混合后不同时间点的透膜效果。其中,蛋白与全氟庚酸修饰的壳聚糖的比例为1:0.25,1:0.5,1:1,1:4。每组均在上药后5分钟,30分钟,1小时,3小时,6小时,12小时,24小时取样,最后通过检测蛋白上标志的荧光,计算累积透过量。结果如图4-3。最后选用蛋白与全氟庚酸修饰的壳聚糖为1:1的比例进行动物实验。
2.制备含氟壳聚糖滴眼液:泪液的正常pH值为6.4-7.7,故采用pH=7.4的磷酸盐缓冲溶液溶解含氟壳聚糖固体粉末,浓度为2mg/mL,在不断搅拌的情况下逐滴滴加标有Cy5.5的高浓度牛血清白蛋白形成稳定复合物,最终牛血清白蛋白浓度为2mg/mL。全氟庚酸修饰的壳聚糖与小分子罗丹明B的滴眼液与上述制备一致。
3.为检测全氟庚酸修饰的壳聚糖的穿透眼睛屏障的能力,比较含全氟庚酸修饰的壳聚糖蛋白与游离蛋白穿透进入眼睛内部的程度。将小鼠麻醉,用定量给药器在眼表给药,每只眼睛5μL,将小鼠避光处理12小时。对照组为不加含氟壳聚糖的标有Cy5.5的牛血清白蛋白,与实验组浓度以及给药量相同。12小时后将小鼠脱颈处死,用PH=7.4的PBS冲洗眼球表面,取出眼球,处理多余组织,做切片准备,拍摄眼球中央纵切面。小分子药物罗丹明B的具体实施方法同大分子药物。大分子牛血清白蛋白透膜效果如图4-4,小分子罗丹明B透膜效果如图4-5。由图明显可见,含有全氟庚酸修饰的壳聚糖的蛋白和小分子荧光物质罗丹明B能进入眼球内部,并且蛋白标志的荧光强度与小分子荧光远远高于游离蛋白与游离小分子在眼内的强度,得出结论,全氟庚酸修饰的壳聚糖能帮助系列药物穿透眼睛屏障进入眼内达到治疗目的。
4.为评价不同时间点眼部的蛋白药物浓度,取全氟庚酸修饰的壳聚糖与蛋白比例 一比一的情况下,比较游离蛋白3小时以及6小时给药后解剖眼球各部分的药物浓度(角膜,晶状体,玻璃体,视网膜)。解剖后经组织破碎仪破碎,再用裂解液裂解,离心除去破碎片,取上清对蛋白标志的荧光进行检测,计算荧光强度,如图4-6。首先比较含全氟庚酸修饰的壳聚糖药物与游离药物在眼睛四个组织部分的蛋白荧光强度,明显含全氟庚酸修饰的壳聚糖组要明显高于游离蛋白组。其次,根据时间的累积,6小时的含全氟庚酸修饰的壳聚糖蛋白组比3小时的荧光强度高,由此可见,全氟庚酸修饰的壳聚糖可能还具有黏附于眼表缓释进入眼内的作用。
5.为评价对角膜的穿透能力,在不同时间点将角膜进行免疫荧光染色,观察穿透能力。含全氟庚酸修饰的壳聚糖的药物上药后5分钟,15分钟,30分钟,60分钟取样冷冻切片,将切片的细胞核进行染色,于共聚焦显微镜下观察如图4-7。其中红色为蛋白标志的荧光,蓝色为眼组织细胞核。从图中明显可以看出当30分钟后含全氟庚酸修饰的壳聚糖组已经明显渗透入角膜上皮细胞,而游离的蛋白则基本没有进入。可见全氟庚酸修饰的壳聚糖能打开角膜屏障,对角膜相关疾病进行针对治疗。
6.为评价全氟庚酸修饰的壳聚糖的生物安全性,选择20只Balb/c小鼠,鼠龄6-8w,性别不限,实验分为全氟庚酸修饰的壳聚糖组、生理盐水组、PBS组、空白对照组,每组各5只小鼠。点眼频率为4次/天。分别在用药后24h、48h和72h裂隙灯生物显微镜拍照纪录角膜上皮荧光素钠染色,以表4-3临床评价为标准进行评价,评价图片见图4-8。全氟庚酸修饰的壳聚糖组用药后24h缺损面积缩小,缺损面积均<30%;48h后有2只小鼠角膜上皮存在缺损,另外3只上皮完全愈合;72h后5只小鼠角膜上皮完全愈合。生理盐水组:用药后24h缺损面积缩小,2只小鼠角膜上皮缺损面积在30%-70%之间,另外三只小鼠均<30%;48h后有1只小鼠角膜上皮存在缺损,缺损面积<30%,4只小鼠上皮完全愈合;72h后1只小鼠上皮缺损面积存在上皮缺损,缺损面积<30%,4只小鼠角膜上皮完全愈合。PBS用药后24h缺损面积缩小,缺损面积均<30%;48h后有1只小鼠角膜上皮存在点状染色,4只上皮完全愈合;72h后5只小鼠角膜上皮完全愈合。空白对照组则用药后24h缺损面积缩小,缺损面积均<30%;48h后5只小鼠角膜上皮完全愈合。该临床方法检测下,全氟庚酸修饰的壳聚糖对眼睛具有极高安全性。图8中裂隙灯下经荧光素钠染色的角膜,若出现角膜缺损则被染色,黑白模式下应为灰色。表4-4为图4-8评价的结果。
表4-3临床检验对照表
Figure PCTCN2020138465-appb-000039
注:痊愈与显效合计为有效,计算有效率;进步与无效合计为无效,计算无效率。
表4-4
Figure PCTCN2020138465-appb-000040
图4-8与表4-4的评价结果明显可以看出实验组与对照组眼球表面的角膜均无可见灰色区域显示角膜上皮缺损情况,由此可见,经全氟庚酸修饰的壳聚糖对眼角膜基本没有影响,安全性较高。
实施例4-2:以药物anti-PDL1为免疫治疗药物,与全氟庚酸修饰的壳聚糖制备成眼药水对脉络膜恶性黑色素瘤进行治疗,证明全氟庚酸修饰的壳聚糖具有递送作用。
具体方法:
1.动物模型:将处于对数期的转染有生物发光基因的B16黑色素瘤通过眼部注射至Balb/c小鼠眼球脉络膜右眼,每只眼睛注射1×10 5个细胞,注射后养育4天,通过生物自发光成像体系,通过生物发光强度表示肿瘤大小。见图4-9给药前各组眼瘤成型大小。
2.全氟庚酸修饰的壳聚糖/anti-PDL1眼药水制备:制备方式同实施例4-1中第2点。
3.评价方式:图4-9中最右侧为生物发光强度变化指示图,图例中光斑越大,颜色越深说明肿瘤越严重。
治疗方式:将造模成功的小鼠进行分组,未给药的为对照组,滴眼药水的为实验组,每组三只。自造模后第四天开始治疗,实验组每天滴一次,每次滴2.5μL,药物浓度为2mg/mL。治疗一周后进行生物发光成像,结果如图4-9与图4-10,其中,图4-9上面三只小鼠为对照组,下面三只为实验组。从图中明显看到治疗一周后含全氟庚酸修饰的壳聚糖的小鼠免疫生物发光比对照组弱,治疗一周后原位脉络膜恶性黑色素瘤的生物自发光定量分析,治疗后的自发光强度为对照组的四分之一,具有显著治疗作用。
实施例5-1:
一、以全氟庚酸修饰的壳聚糖为载体制备氟化壳聚糖-鸡卵清白蛋白复合物,与骨髓来源树突状细胞孵育,考察复合物刺激树突状细胞成熟的能力。
具体方法:
1.制备全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物:称取0.9mg全氟庚酸修饰的壳聚糖用900μL超纯水搅拌溶解,在不断搅拌的情况下滴入100μL(20mg/mL)的鸡卵清白蛋白,并持续搅拌一小时,得到全氟庚酸修饰的壳聚糖-鸡卵清白蛋白(FCS-OVA)复合物。
2.将10μL上述制备复合物加入24孔板中,加入含有100万个树突状的细胞悬液1mL。在37℃培养箱中孵育24h,用FITC-CD11c、PE-CD86、APC-CD80对树突状细胞进行染色,用流式细胞技术分析FITC的荧光信号,选定树突状细胞再分析树突状细胞PE与APC的荧光信号,确定其成熟度结果如图1-1所示。
实验结果:如图5-1所示,全氟庚酸修饰的壳聚糖-鸡卵清白蛋白(FCS-OVA)复合物相比于不作处理组,能够刺激树突状细胞的成熟。
二、以全氟庚酸修饰的壳聚糖为载体制备全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物,与骨髓来源树突状细胞孵育,考察复合物刺激树突状细胞表达第二类组织相容性复合体(MHC II)与CD40蛋白的情况。
具体方法:
1.制备全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物:称取0.9mg全氟庚酸修饰的壳聚糖用900μL超纯水搅拌溶解,在不断搅拌的情况下滴入100μL(20mg/mL)的鸡卵清白蛋白,并持续搅拌一小时,得到全氟庚酸修饰的壳聚糖-鸡卵清白蛋白(FCS-OVA)复合物。
2.将10μL上述制备复合物加入24孔板中,加入含有100万个树突状的细胞悬液1mL。在37℃培养箱中孵育24h,用FITC-CD11c、PE-MHC II、APC-CD40对树突状细胞进行染色,用流式细胞技术分析FITC的荧光信号,选定树突状细胞再分析树突状细胞PE与APC的荧光信号,确定第二类组织相容性复合体(MHC II)与CD40蛋白的变化。结果如图5-2所示。
实验结果:如图5-2所示,在全氟庚酸修饰的壳聚糖-鸡卵清白蛋白(FCS-OVA)复合物的刺激下,树突状细胞中第二类组织相容性复合体(MHC II)与CD40蛋白的表达量明显增加,说明该复合物能有效地刺激树突状细胞呈递抗原。
三、以全氟庚酸修饰的壳聚糖为载体制备全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物,用敷贴敷与小鼠背部考察复合物在淋巴结聚集的情况。
具体方法:
1.制备cy5.5标记的鸡卵清白蛋白:称取10mg的鸡卵清白蛋白中,用1mL PBS溶解,加入20μL(20mg/mL)的cy5.5,4℃放置过夜。通过G25-分子筛凝胶除去游离cy5.5。通过BSA定量,再用超滤将OVA浓缩20mg/mL。
2.备全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物:称取1mg全氟庚酸修饰的壳聚糖用450μLPBS搅拌溶解,在不断搅拌的情况下滴入50μL(20mg/mL)PBS溶解被cy5.5标记的鸡卵清白蛋白,并持续搅拌一小时,得到全氟庚酸修饰的壳聚糖-鸡卵清白蛋白(FCS-OVA-cy5.5)复合物。
3.取12.5μL上述FCS-OVA-cy5.5复合物溶液混入12.5μg软膏中,制备成FCS-OVA-cy5.5疫苗软膏,将上述软膏涂抹于C57小鼠背部。利用小动物成像在0h、3h、7h、11h、24h拍摄小鼠正面荧光分布情况,观察FCS-OVA-cy5.5在淋巴结的聚集情况。结果如图5-3所示。
4.实验结果:如图5-3所示,在11小时、24小时,小鼠淋巴结部位出现荧光,意味着利用敷贴固定在小鼠背上的全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物能够透过皮肤进入小鼠淋巴结。
四、以全氟庚酸修饰的壳聚糖为载体制备全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物,并用贴片敷于皮肤上作为疫苗种植,三周后,种植B16-OVA肿瘤,观察肿瘤生长情况。
具体方法:
1.制备全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物:称取1mg全氟庚酸修饰的壳聚糖用450μLPBS搅拌溶解,在不断搅拌的情况下滴入50μL(20mg/mL)PBS溶解的鸡卵清白蛋白,并持续搅拌一小时,得到全氟庚酸修饰的壳聚糖-鸡卵清白蛋白(FCS-OVA)复合物。
2.取12.5μL上述FCS-OVA复合物溶液混入12.5μg软膏中,制备成FCS-OVA疫苗软膏,12.5μLOVA(2mg/mL)溶液混入12.5μg软膏中,制备成OVA疫苗软膏。将上述软膏分别涂抹于C57小鼠背部,用敷贴固定12h。每周重复该操作两次,持续三周。
3.每只小鼠种植1x10 5个B16-OVA细胞,观察其肿瘤生长情况。结果如图5-4所示。
实验结果:如图5-4所示,以全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物作为贴片疫苗种植的小鼠相比于单纯以鸡卵清白蛋白种植的小鼠,肿瘤生长速度更缓慢,表明了全氟庚酸修饰的壳聚糖-鸡卵清白蛋白复合物作为贴片具有作为疫苗的效果。
本专利实施例1-1至1-4各个实施例中的氟化壳聚糖,均可以用作疫苗制剂,用于作为各类疫苗接种制剂。
实施例6-1:以目前已知的具有一定生发治疗效果的小分子药物UK5099为例,利用含氟修饰的壳聚糖包载药物,研究不同比例的全氟庚酸修饰的壳聚糖与药物的体外透皮能力。以小分子药物二甲双胍为例,利用含氟修饰的壳聚糖包载药物,研究动物生发实际效果。
具体方法:
1.为观测不同比例全氟庚酸修饰的壳聚糖的透膜效果,进行体外扩散实验。取小鼠背部皮肤,去脂肪,将皮固定在Franz扩散池上,其中,全氟庚酸修饰的壳聚糖与UK5099的比例为1:1,2.5:1,5:1,10:1。每组均在上药后5分钟,30分钟,1小时,2小时,3小时,4小时,7小时,10小时,24小时取样,检测不同时间点接受液中UK5099的浓度,计算累积透过百分比,比较透膜效果。实验结果如图6-1至6-2。最后选用蛋白与全氟庚酸修饰的壳聚糖为1:1的比例进行系列动物实验。
2.制备全氟庚酸修饰的壳聚糖-二甲双胍生发液:首先溶解含全氟庚酸修饰的壳聚糖固体粉末,在不断搅拌的情况下逐滴滴加溶解二甲双胍的液体,使得最终二甲双胍浓度为2mg/mL,全氟庚酸修饰的壳聚糖浓度为2mg/mL。
3.选用第6周的处于毛发禁止期的Balb/c小鼠,将全氟庚酸修饰的壳聚糖-二甲双胍组、游离二甲双胍组、空白对照组的小鼠于治疗前一天分别除去背部等面积毛发并拍照如图。每只小鼠按照各自的治疗方式均匀喷射药物,每两天给一次药,每次给药均为100微升,治疗后第11天、13天、17天的照片如图6-3以及表6-1。
实验结果:从图中可以看出,虽然存在小鼠个体差异,但含全氟庚酸修饰的壳聚糖组的小鼠对比游离药物以及对照组明显有生发趋势。
表6-1
Figure PCTCN2020138465-appb-000041
实施例6-2:制备以全氟庚酸修饰的壳聚糖(FCS)和聚肌胞苷酸(polyinosinic-polycytidylic acid,poly(I:C))为主体的祛疤膏,诱导内源性维甲酸的产生,从而利用维甲酸来改善疤痕。
具体方法:
1、制备全氟庚酸修饰的壳聚糖-胰岛素混合物:将全氟庚酸修饰的壳聚糖与poly(I:C)分别溶于超纯水中使其溶解均匀,两者混合后,全氟庚酸修饰的壳聚糖和胰岛素由于静电吸附作用结合在一起,其中以质量比为1:1的比例混合,形成稳定的纳米颗粒。
2、通过体外L929(小鼠成纤维细胞)的维甲酸刺激实验对动物实验的祛疤效果进行可行性分析:将L929铺于细胞培养6孔板当中,每孔培养基定为3mL体积。在培养基中加入poly(I:C),全氟庚酸修饰的壳聚糖和poly(I:C)以及全氟庚酸修饰的壳聚糖的混合物,使最终poly(I:C)和全氟庚酸修饰的壳聚糖的浓度均为1μg/mL。L929培养48h后,收集培养基和细胞,离心取上清。将上清以1:5的比例和五倍体积乙醚丙酮混合液(乙醚:丙酮=1:8)均匀混合,收集上层油相。用氮气浓缩油相体积,再用HPLC(高效液相色谱法)来检测细胞分泌的维甲酸含量。
3、制备全氟庚酸修饰的壳聚糖与poly(I:C)的祛疤膏:将poly(I:C)和全氟庚酸修饰的壳聚糖1:1预混合,再与等体积的Aquaphor @软膏混匀,得到全氟庚酸修饰的壳聚糖与poly(I:C)的祛疤膏。
4、通过小鼠疤痕的形貌变化来预判断全氟庚酸修饰的壳聚糖与poly(I:C)祛疤膏的祛疤效果:将10-12周的雌性Balb/c小鼠以异氟烷麻醉,背部剪去3mm×10mm的皮肤,涂上碘伏,制作成开放性伤口,一周内小鼠皮肤愈合脱痂,两个月后开始进行祛疤实验。将全氟庚酸修饰的壳聚糖与poly(I:C)祛疤膏的疤痕上,每天涂抹,以周为单位对小鼠疤痕的形貌进行观察和评价。
实验结果:参见图6-4,左图为全氟庚酸修饰的壳聚糖和poly(I:C)的混合物对于小鼠成纤维细胞产生维甲酸的效率(纵坐标Mass of RA含义为维甲酸产生效率),右图为全氟庚酸修饰的壳聚糖和poly(I:C)的混合物涂抹于小鼠疤痕后的效果图。
如图6-4所示,通过HPLC的检测和分析,发现与空白组、单独全氟庚酸修饰的壳聚糖和单独poly(I:C)相比,在体外全氟庚酸修饰的壳聚糖和poly(I:C)混合物能显著提高小鼠成纤维细胞的维甲酸生成量。而经过7天的涂抹,小鼠疤痕明显变平滑。以上结果共同表明,含利用全氟庚酸修饰的壳聚糖作为载体递送poly(I:C)能够成功实现体外细胞维甲酸 的生成量,且在活体疤痕模型中有明显效果,具备转化价值。
实施例6-3:以治疗黄褐斑药物氨甲环酸为例,以含氟修饰的壳聚糖包载氨甲环酸,利用扩散池探究其透皮能力。
具体方法:
1.制备全氟庚酸修饰的壳聚糖-氨甲环酸溶液:将固体粉末全氟庚酸修饰的壳聚糖溶于PBS中,与溶于PBS的氨甲环酸1:1混合,涡旋振荡5分钟,使两者以静电作用力结合,最终溶液中全氟庚酸修饰的壳聚糖与氨甲环酸的终浓度都为1mg/mL。
2.体外扩散池搭建:将大鼠腹部皮肤取下,剃去脂肪层,固定在扩散池上,在皮肤下部扩散池加入7.5mL PBS,在皮肤上部扩散池加入0.75mL全氟庚酸修饰的壳聚糖-氨甲环酸和同浓度单纯氨甲环酸溶液。
3.氨甲环酸透过率测定:在0,1,3h从底部扩散池取出500μL PBS并补入同样体积的PBS。以甲醇与0.05mol/LKH2PO4-0.2%H3PO4溶液(体积比为5∶95)为流动相,流速为1.0mL/min,检测波长为210nm的HPLC条件下检测氨甲环酸(tranexamic acid)含量,并计算累计透过量,结果如图所示,横坐标为时间,纵坐标含义为累计透过量。
实验结果:如图6-5所示,在前三个小时内,混合了全氟庚酸修饰的壳聚糖的氨甲环酸能够达到30%的累计透过量,而单纯的氨甲环酸分子基本没有透过。证明了全氟庚酸修饰的壳聚糖-氨甲环酸具有较好的透皮肤能力,能够用于皮肤病黄褐斑的治疗。
对所公开的实施例的上述说明,使得本技术领域专业技术人员能够实现或使用本发明。对这些实施例的多种修改对于本领域技术人员而言将是显而易见的。本发明将不会被限制于本文所示的这些实施例,而是要符合与本文所公开的原理与特点相一致的最宽的范围。

Claims (15)

  1. 一种作为药物载体用的氟化修饰的壳聚糖衍生物,具有如下结构:含氟化合物共价连接在壳聚糖主链上,
    所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,
    所述含氟化合物为如下化学式(Ⅰ)
    Figure PCTCN2020138465-appb-100001
    所示的含氟脂肪链,或式(Ⅱ)
    Figure PCTCN2020138465-appb-100002
    所示的芳香环功能基团,所述R1为卤素(氟,氯,溴,碘)、卤素取代的烷烃、环烷烃、醛基、羧基、双键、炔键、羟基、磺酰氯、磺酸键或巯基这些能够与伯氨基反应的活性基团。
  2. 一种作为药物载体用的氟化修饰的壳聚糖衍生物,具有如式(Ⅳ)所示的含有伯氨基的壳聚糖分子骨架:
    Figure PCTCN2020138465-appb-100003
    所述壳聚糖的伯氨基与含氟功能基团之间形成的连接基团为:-NH-、-N=C-、-NHCH 2CH(OH)-、-NHCH 2CH(OH)CH 2O-、
    Figure PCTCN2020138465-appb-100004
    以及衍生基团;所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,
    所述含氟功能基团为含氟脂肪链、芳香环功能基团。
  3. 根据权利要求1所述的作为药物载体用的氟化修饰的壳聚糖衍生物,其特征在于:所述式(Ⅰ)中x为0-3的整数,y为0-20的整数,z为0-8的整数,R2为CF 3、CHF 2、CH 2F、或CH 3(当y不为0);
    所述含氟脂肪链化合物是指含氟烃基及其衍生物,包括三氟乙酸、五氟丙酸、七氟丁酸、九氟戊酸、十一氟己酸、十三氟庚酸、十五氟辛酸、十七氟壬酸、十九氟奎酸、全氟丁酸酐、全氟庚酸酐、全氟癸酸酐、2,2,3,3,4,4,4-七氟丁基丙烯酸酯、3-(1H,1H,5H八氟戊氧基)-1,2-环氧丙烯、九氟丁基璜酰胺酐及其衍生物。
  4. 根据权利要求1所述的作为药物载体用的氟化修饰的壳聚糖衍生物,其特征在于:所述式(Ⅱ)中R为H,CH 3,OH,NO 2,O,CF 3,F,CH 2OH,CN,NCO,或(CF 2)aCF 3(a为1-20的整数)等,且至少一个R为F;
    所述含氟芳香环化合物包括3-氟苯甲酸、3,5-二氟苯甲酸、2,3,5,6-四氟-4-甲基苯甲酸、五氟苯甲酸、2-氟-3-(三氟甲基)苯甲酸及其衍生物。
  5. 根据权利要求1所述的作为药物载体用的氟化修饰的壳聚糖衍生物,其特征在于:所述壳聚糖和含氟化合物共价连接,在所述壳聚糖分子表面进行修饰,构成一种药物载体,其结构如式(V)所示,b、c均为20-500的整数,式(V)如下:
    Figure PCTCN2020138465-appb-100005
    其中,B为含氟功能基团与壳聚糖伯氨基形成的连接基团,C为含氟脂肪链、芳香环功能基团。
  6. 根据权利要求1所述的作为药物载体用的氟化修饰的壳聚糖衍生物,其特征在于:所述含氟脂肪链是带有可以与氨基反应的活性基团的一类含氟化合物,包括如式(Ⅵ)所示:
    Figure PCTCN2020138465-appb-100006
    Figure PCTCN2020138465-appb-100007
    其中A为-COOH、
    Figure PCTCN2020138465-appb-100008
    能够与伯氨基反应的活性基团,x为0-3的整数,y为0-8的整数。
  7. 根据权利要求1所述的作为药物载体用的氟化修饰的壳聚糖衍生物,其特征在于:所述含氟芳香环化合物是带有可以与氨基反应的活性基团的一类含氟化合物,包括如式(Ⅶ)所示:
    Figure PCTCN2020138465-appb-100009
  8. 根据权利要求1至7中任一权利要求所述的作为药物载体用的氟化修饰的壳聚糖衍生物,其特征在于:所述氟化修饰的壳聚糖衍生物作为如下药物的药物载体,所述如下药物为小分子药物、多肽、蛋白药物、不同药物的组合药物以及药物与其它药用辅料的组合药物。
  9. 含氟化合物修饰的壳聚糖作为药物载体的应用,其特征在于:权利要求1至7中任一权利要求所述的氟化修饰的壳聚糖衍生物可以作为小分子药物、多肽、蛋白药物、不同药物的组合药物以及药物与其它药用辅料的组合药物的药物载体应用。
  10. 一种药物复合物,其特征在于:包括根据权利要求1至7中任一权利要求所述的作为药物载体用的氟化修饰的壳聚糖衍生物以及药物,所述药物包括小分子药物、多肽、蛋白药物、不同药物的组合药物以及药物与其它药用辅料的组合药物。
  11. 一种根据权利要求1至7中任一权利要求所述的作为药物载体用的氟化修饰的壳聚糖衍生物制作的透皮给药制剂,包括透皮制剂组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物为氟化壳聚糖,含氟化合物共价连接在壳聚糖主链上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊。
  12. 一种根据权利要求1至7中任一权利要求所述的作为药物载体用的氟化修饰的壳聚糖衍生物制作的跨粘膜给药制剂,包括跨粘膜制剂组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物为氟化壳聚糖,含氟化合物共价连接在壳聚糖主链上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,所述粘膜包括鼻腔黏膜、肺粘膜、阴道黏膜、口腔黏膜、胃肠黏膜。
  13. 一种根据权利要求1至7中任一权利要求所述的作为药物载体用的氟化修饰的壳聚糖衍生物制作的穿透眼部屏障给药制剂,包括穿透眼部屏障制剂组分(a),所述组分(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物为氟化壳聚糖,含氟化合物共价连接在壳聚糖主链上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,所述眼部屏障为泪液屏障、角膜/结膜屏障、血房水屏障、血视网膜屏障。
  14. 一种根据权利要求1至7中任一权利要求所述的作为药物载体用的氟化修饰的壳聚糖衍生物制作的透皮疫苗载体,包括透皮疫苗载体(a),所述透皮疫苗载体(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物为氟化壳聚糖,含氟化合物共价连接在壳聚糖主链上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,所述透皮疫苗载体有胞内渗透、胞间渗透和毛囊渗透三种抗原透过途径。
  15. 一种根据权利要求1至7中任一权利要求所述的作为药物载体用的氟化修饰的壳聚糖衍生物制作的医美及保健品载体,包括医美及保健品载体(a),所述医美及保健品载体(a)为含氟化合物修饰的阳离子聚合物,所述含氟化合物修饰的阳离子聚合物为氟化壳聚糖,含氟化合物共价连接在壳聚糖主链上,所述壳聚糖的分子量范围在1000-5000000,脱乙酰度不小于55%且粘度范围为25-1000厘泊,所述医美及保健品载体能适用于生发药物和毛发护理药物、美容药物、保健药物。
PCT/CN2020/138465 2019-04-30 2020-12-23 含氟化合物修饰的阳离子聚合物作为药物载体的应用及制备方法 WO2021147598A1 (zh)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/630,143 US20220273806A1 (en) 2019-04-30 2020-12-23 Use of fluorine-containing compound-modified cationic polymer as drug carrier and preparation method
EP20915498.8A EP4095162A4 (en) 2019-04-30 2020-12-23 APPLICATION AND MANUFACTURING PROCESS FOR FLUORINE CONTAINING COMPOUND MODIFIED CATIONIC POLYMER AS DRUG CARRIER

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN201910361123 2019-04-30
CN202010061971.2A CN111848832B (zh) 2019-04-30 2020-01-20 含氟化合物修饰的阳离子聚合物作为药物载体的应用及制备方法
CN202010061971.2 2020-01-20

Publications (1)

Publication Number Publication Date
WO2021147598A1 true WO2021147598A1 (zh) 2021-07-29

Family

ID=72984860

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2020/137711 WO2021147585A1 (zh) 2019-04-30 2020-12-18 含氟化合物修饰的壳聚糖作为药物载体的用途及其制备方法
PCT/CN2020/138465 WO2021147598A1 (zh) 2019-04-30 2020-12-23 含氟化合物修饰的阳离子聚合物作为药物载体的应用及制备方法

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/137711 WO2021147585A1 (zh) 2019-04-30 2020-12-18 含氟化合物修饰的壳聚糖作为药物载体的用途及其制备方法

Country Status (4)

Country Link
US (1) US20220273806A1 (zh)
EP (1) EP4095162A4 (zh)
CN (7) CN111848835B (zh)
WO (2) WO2021147585A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116650477A (zh) * 2023-06-14 2023-08-29 黑龙江迪龙制药有限公司 用于抗血小板聚集的含奥扎格雷钠的药物组合物及其制备方法
WO2023154888A3 (en) * 2022-02-11 2023-11-16 The Trustees Of Columbia University In The City Of New York Method of treating obesity by selective targeting of visceral adiposity using polycation nanomedicine
WO2024067874A1 (zh) * 2022-09-30 2024-04-04 苏州百迈生物医药有限公司 一种改性壳聚糖及其在大分子活性成分递送中的应用

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111848835B (zh) * 2019-04-30 2023-02-03 苏州大学 含氟化合物修饰的壳聚糖在透皮给药制剂制备中的应用
CN110772635B (zh) * 2019-11-11 2023-01-31 扬州大学 流感病毒小体包被的仿生纳米疫苗及其制备方法
CN113081987B (zh) * 2021-04-29 2022-03-25 药大制药有限公司 一种氟胞嘧啶片剂及其制备方法
CN113730332B (zh) * 2021-08-02 2022-12-30 深圳市众循精准医学研究院 一种靶向肿瘤表面胶原的促进剂及其制备方法与应用
CN113930454A (zh) * 2021-11-26 2022-01-14 南通大学 壳聚糖-聚二甲双胍在作为基因转染试剂方面的应用
CN114796157B (zh) * 2022-05-12 2024-01-05 苏州大学 一种氟化纳米胶囊及其制备方法和应用
CN115778845B (zh) * 2023-02-07 2023-04-25 荷本世新(北京)生物科技有限公司 蛋白组合物及其制备方法和用途
CN117462653A (zh) * 2023-11-28 2024-01-30 山东第一医科大学附属眼科研究所(山东省眼科研究所、山东第一医科大学附属青岛眼科医院) 抑瘤素m在制备促进角膜上皮损伤修复的药物中的应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1813684A (zh) * 2005-11-15 2006-08-09 暨南大学 5-氟尿嘧啶/壳聚糖纳米载药微球的制备方法
CN101029091A (zh) * 2006-12-01 2007-09-05 中国科学院海洋研究所 壳聚糖的羟基苯二磺酰胺类衍生物及其制备方法
CN101029092A (zh) * 2007-01-19 2007-09-05 中国科学院海洋研究所 壳聚糖硫酸酯的羟基苯二磺酰胺类衍生物及制备方法
WO2008141452A1 (en) * 2007-05-18 2008-11-27 Rival S.E.C. Chitosan salts, methods of manufacture and uses thereof
CN111848832A (zh) * 2019-04-30 2020-10-30 苏州大学 含氟化合物修饰的阳离子聚合物作为药物载体的应用及制备方法

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5422116A (en) * 1994-02-18 1995-06-06 Ciba-Geigy Corporation Liquid ophthalmic sustained release delivery system
US5800832A (en) * 1996-10-18 1998-09-01 Virotex Corporation Bioerodable film for delivery of pharmaceutical compounds to mucosal surfaces
MXPA02000053A (es) * 1999-07-05 2003-07-21 Idea Ag Un metodo para mejorar el tratamiento a traves de barreras adaptables semipermeables.
JP4750265B2 (ja) * 2000-11-02 2011-08-17 ダイセル化学工業株式会社 置換基にフッ素を含有するキチン誘導体
GB0126923D0 (en) * 2001-11-09 2002-01-02 Procter & Gamble Chitosan compositions
JPWO2005005485A1 (ja) * 2003-05-07 2006-08-24 財団法人生産開発科学研究所 キチンオリゴマー組成物及び/又はキトサンオリゴマー組成物、並びにその製造方法
US20050136103A1 (en) * 2003-09-17 2005-06-23 Ben-Sasson Shmuel A. Compositions capable of facilitating penetration across a biological barrier
JP2008505978A (ja) * 2004-07-12 2008-02-28 アラーガン、インコーポレイテッド 眼病用組成物および眼病治療法
CN101437478A (zh) * 2004-10-04 2009-05-20 Qlt美国有限公司 聚合送递制剂的眼部送递
US7604818B2 (en) * 2004-12-22 2009-10-20 Advanced Cardiovascular Systems, Inc. Polymers of fluorinated monomers and hydrocarbon monomers
CN100341899C (zh) * 2005-11-01 2007-10-10 中国药科大学 新的壳聚糖衍生物、其制法及其用于制备眼科制剂的用途
CN101209241A (zh) * 2006-12-29 2008-07-02 天津医科大学眼科中心 药物靶向控释纳米粒滴眼液的制备方法
US8877170B2 (en) * 2009-02-21 2014-11-04 Sofradim Production Medical device with inflammatory response-reducing coating
CN101550200B (zh) * 2009-05-27 2011-06-15 北京化工大学 一种大环多胺偶联壳聚糖基因载体及其制备方法和应用
CN101987084B (zh) * 2009-07-31 2012-05-09 天津市医药科学研究所 一种含有壳聚糖和胶原缓释滴眼剂及其制备方法
CN101838345B (zh) * 2010-05-17 2012-05-30 北京师范大学 氟-18标记的半乳糖化壳聚糖化合物及其制备方法
CN102485772A (zh) * 2010-12-01 2012-06-06 吉林大学 壳聚糖微球作为基因疫苗载体的制备方法及应用
CN107522880B (zh) * 2012-01-25 2020-07-24 阿克伦大学 水凝胶及其制备方法
ES2658972T3 (es) * 2012-06-20 2018-03-13 University Of Waterloo Sistema de administración de nanopartículas mucoadhesivas
WO2014028796A2 (en) * 2012-08-17 2014-02-20 Smartek International Llc Preparation of desiccated liposomes for use in compressible delivery systems
CN104854134B (zh) * 2012-12-12 2018-01-02 索尔维特殊聚合物意大利有限公司 氟化壳聚糖衍生物
CN103290046A (zh) * 2013-05-27 2013-09-11 华东师范大学 基于氟化物修饰的树形高分子基因转染载体及其制备方法和应用
CN103316362B (zh) * 2013-06-28 2015-03-04 江苏大学 基于壳聚糖结构的含氟潜在双功能探针及其制备方法
WO2015131144A1 (en) * 2014-02-28 2015-09-03 The University Of Memphis Research Foundation Chitosan nanofiber compositions, compositions comprising modified chitosan, and methods of use
US20190002594A1 (en) * 2014-07-16 2019-01-03 Immunophotonics, Inc. Chitosan-Derived Compositions
CN108697803A (zh) * 2015-10-29 2018-10-23 索卢贝斯特有限公司 透粘膜给药的药物组合物
CN105769817A (zh) * 2016-03-09 2016-07-20 辽宁科技学院 一种巯基化壳聚糖表面修饰纳米脂质载体及其制备方法
CN107638405A (zh) * 2016-07-15 2018-01-30 复旦大学 一种眼内递药组合物及其制备方法
CN106474095B (zh) * 2016-11-15 2021-03-26 宁波百思佳医药科技有限公司 一种用于治疗口腔真菌的口腔粘膜粘贴剂及其制备方法
CN109125740B (zh) * 2017-06-28 2022-04-05 成都威斯克生物医药有限公司 一种新型的肿瘤疫苗及其用途
CN107903337A (zh) * 2017-11-10 2018-04-13 中国药科大学 具有粘膜粘附性和Pept-1靶向性的一种壳聚糖衍生物及其制备方法
CN107903339B (zh) * 2017-11-27 2020-04-14 暨南大学 一种含两性氨基酸基团的壳聚糖衍生物及制备方法与应用
CN108435140B (zh) * 2018-03-19 2020-07-28 武汉瑞法医疗器械有限公司 一种破伤风外毒素吸附剂及应用
CN108904804B (zh) * 2018-08-02 2021-04-06 苏州百迈生物医药有限公司 一种装载全氟碳的氟化共价有机聚合物及其制备方法和应用
CN109438600B (zh) * 2018-10-25 2020-12-11 暨南大学 氟化聚(酰胺胺)的制备方法及其作为疫苗免疫佐剂的应用
CN110585130A (zh) * 2019-09-06 2019-12-20 深圳市罗湖区人民医院 一种具有原位产氢的纳米胶束及其制备方法与应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1813684A (zh) * 2005-11-15 2006-08-09 暨南大学 5-氟尿嘧啶/壳聚糖纳米载药微球的制备方法
CN101029091A (zh) * 2006-12-01 2007-09-05 中国科学院海洋研究所 壳聚糖的羟基苯二磺酰胺类衍生物及其制备方法
CN101029092A (zh) * 2007-01-19 2007-09-05 中国科学院海洋研究所 壳聚糖硫酸酯的羟基苯二磺酰胺类衍生物及制备方法
WO2008141452A1 (en) * 2007-05-18 2008-11-27 Rival S.E.C. Chitosan salts, methods of manufacture and uses thereof
CN111848832A (zh) * 2019-04-30 2020-10-30 苏州大学 含氟化合物修饰的阳离子聚合物作为药物载体的应用及制备方法

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP4095162A4 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023154888A3 (en) * 2022-02-11 2023-11-16 The Trustees Of Columbia University In The City Of New York Method of treating obesity by selective targeting of visceral adiposity using polycation nanomedicine
WO2024067874A1 (zh) * 2022-09-30 2024-04-04 苏州百迈生物医药有限公司 一种改性壳聚糖及其在大分子活性成分递送中的应用
CN116650477A (zh) * 2023-06-14 2023-08-29 黑龙江迪龙制药有限公司 用于抗血小板聚集的含奥扎格雷钠的药物组合物及其制备方法
CN116650477B (zh) * 2023-06-14 2023-11-10 黑龙江迪龙制药有限公司 用于抗血小板聚集的含奥扎格雷钠的药物组合物及其制备方法

Also Published As

Publication number Publication date
CN111848834A (zh) 2020-10-30
EP4095162A4 (en) 2023-08-02
CN111892667A (zh) 2020-11-06
EP4095162A1 (en) 2022-11-30
CN111848832B (zh) 2024-02-20
CN111848832A (zh) 2020-10-30
CN111848831A (zh) 2020-10-30
CN111848835B (zh) 2023-02-03
CN111848835A (zh) 2020-10-30
CN111848831B (zh) 2023-06-13
CN111848833B (zh) 2023-01-24
US20220273806A1 (en) 2022-09-01
CN111848833A (zh) 2020-10-30
CN111892667B (zh) 2024-02-20
WO2021147585A1 (zh) 2021-07-29
CN111848834B (zh) 2023-01-24
CN111848830A (zh) 2020-10-30

Similar Documents

Publication Publication Date Title
WO2021147598A1 (zh) 含氟化合物修饰的阳离子聚合物作为药物载体的应用及制备方法
Parhi Drug delivery applications of chitin and chitosan: a review
US20190105261A1 (en) Methods and compositions for topical delivery
EP2729123B1 (en) Cannabinoid receptor binding agents, compositions and methods
US20190008795A1 (en) Compositions for topical application of compounds
CN108135967B (zh) 用于药物递送的具有渗透增强剂的组合物
Sábio et al. Exploiting mesoporous silica nanoparticles as versatile drug carriers for several routes of administration
Li et al. Novel engineered systems for oral, mucosal and transdermal drug delivery
JP2019506460A (ja) 浮遊病原体および刺激物に対して防御するための組成物および方法
Wang et al. Chitosan/hyaluronan nanogels co-delivering methotrexate and 5-aminolevulinic acid: A combined chemo-photodynamic therapy for psoriasis
CN108366984A (zh) 用于治疗眼部病症的化合物和组合物
Bharat et al. A review: Novel advances in semisolid dosage forms & patented technology in semisolid dosage forms
Wang et al. Microneedles with two-stage glucose-sensitive controlled release for long-term insulin delivery
Frijlink et al. Biopharmaceutics
Garg et al. In-situ gel: A smart carrier for Drug Delivery
TWI468184B (zh) 藥學組成物及使用其之藥用化妝品
RU2481834C2 (ru) Антимикробная композиция для лечения ран и ожогов
Bermejo et al. How and where are drugs absorbed?
WO2017114379A1 (zh) 一种治疗多发性硬化症的鼻用凝胶组合物
Salatin et al. Introduction: An overview of the non-parenteral delivery of nanomedicine
WO2021110064A1 (en) New multi-functional oligopeptides
Zhang et al. Recent development in mucosal drug delivery system
Torres-Suárez et al. Design of dosage forms: influences of anatomy and administration routes
Kolawole Developing novel mucoadhesive chitosan based formulations for drug delivery to the urinary bladder
Sharma et al. NASAL MUCOSA AS A NOVELISTIC PLATFORM FOR BRAIN TARGETTING

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20915498

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020915498

Country of ref document: EP

Effective date: 20220822