WO2019019091A1 - Curcumin pharmaceutical preparations - Google Patents

Curcumin pharmaceutical preparations Download PDF

Info

Publication number
WO2019019091A1
WO2019019091A1 PCT/CN2017/094665 CN2017094665W WO2019019091A1 WO 2019019091 A1 WO2019019091 A1 WO 2019019091A1 CN 2017094665 W CN2017094665 W CN 2017094665W WO 2019019091 A1 WO2019019091 A1 WO 2019019091A1
Authority
WO
WIPO (PCT)
Prior art keywords
drug delivery
delivery system
curcumin
group
lipid
Prior art date
Application number
PCT/CN2017/094665
Other languages
French (fr)
Chinese (zh)
Inventor
周新富
安基特·帕里克
桑贾伊·加尔
Original Assignee
周意
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 周意 filed Critical 周意
Priority to PCT/CN2017/094665 priority Critical patent/WO2019019091A1/en
Publication of WO2019019091A1 publication Critical patent/WO2019019091A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • 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
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the invention belongs to the field of medicine.
  • the invention relates to novel curcumin pharmaceutical formulations.
  • the novel preparation can more effectively utilize curcumin for anti-oxidation, anti-inflammatory, anti-cancer, induce apoptosis, anti-angiogenesis, neuroprotection, anti-microbial, liver and kidney protection, inhibit blood vessel formation, prevent myocardial infarction, lower blood sugar, Anti-rheumatic and other pharmacological effects.
  • Curcumin is undoubtedly one of the most biologically active molecules discovered in nature to date. Curcumin interacts with a large number of intracellular extracellular targets in a biphasic dose-dependent manner. It controls inflammatory, oxidative stress, cell survival, cell secretion, homeostasis and proliferation. The mechanism of action generally points to cells that exhibit disordered physiology or abnormal states of apparent mutation. It can easily cross all physiological barriers, including the blood-brain barrier.
  • curcumin did not have any side effects when administered orally at 12 g/day.
  • Curcumin and turmeric products have been identified as safe food additives by the US Food and Drug Administration (FDA), the Canadian Natural Health Products Agency, and the Joint FAO/WHO Expert Committee.
  • Curcumin is a natural polyphenolic compound that is commercially isolated from the rhizome of Curcuma longa Linn (Curcuma) (Zingiberaceae) and has a long history in Ayurvedic medicine. Many Asian countries, such as India and China, have been widely used as herbs for thousands of years.
  • the chemical name of curcumin is 1,7-bis-(4-hydroxy-3-methoxyphenyl)-hepta-1,6-diene-3,5-dione, the chemical formula is C 21 H 20 O 6 .
  • Curcumin-like is called diferuloyl Methane and is the main component of curcumin (77% by weight); the other two curcumin are demethoxycurcumin (17% by weight) and bis-methoxycurcumin (3 wt%).
  • Curcumin is a fat-soluble polyphenolic compound consisting structurally of two cyclomethoxyphenols attached to a diketone structure. Keto-enol tautomerization allows curcumin to act as a Michael acceptor 4 . Phenol groups and diketones are characteristic of antioxidant compounds and are key structures for the antioxidant action of curcumin.
  • Curcumin is safe and well tolerated. Due to its efficacy and safety, curcumin has been studied in a wide range of research fields, in vitro and in vivo, in animals and in humans. Low oral bioavailability (only 1% in rats) and very short biological half-life are limiting factors in the clinical development of curcumin. However, due to its lipophilic nature, curcumin can pass freely through the cell membrane (log P 1/42.5). The main reason for the low bioavailability of curcumin is its very low solubility in water (only 11 ng/ml in an aqueous solution of pH 5.0), acidity and physiological pH, and rapid hydrolysis under alkaline conditions.
  • the serum level of curcumin peaked after 1-2 hours after oral administration, and the serum concentration ranged from 0.51 ⁇ 0.11 ⁇ M at a dose of 4,000 mg/day. At 8,000 mg/day, the serum concentration ranged from 1.77 ⁇ 1.87 uM.
  • alleviation of the condition usually requires a tailored method of administration in order for the drug to reach the lesion in a therapeutic amount.
  • Optimizing prophylactic or therapeutic applications requires the provision of an appropriate amount of curcumin to impaired cellular targets.
  • the therapeutic effect can be accompanied by accidental toxicity to surrounding normal healthy cells and tissues.
  • a biologically active compound is its solubility in aqueous body fluids.
  • Lipophilic compounds such as curcumin lack water solubility, but retain significant cell membrane or intracellular activity. It is necessary to use new formulations for its application. Therefore, one of the main challenges of the pharmaceutical industry is the application of strategies to develop such problematic compounds into clinically orally, bioavailable and therapeutically effective drugs.
  • curcumin In order to develop curcumin as a clinical drug, the ideal formulation should address the following problems: poor solubility, chemical stability (hydrolysis, oxidation, heat and light) and pharmacokinetic properties, including absorption, distribution, metabolism and elimination in vivo, bioavailability Low degree, poor permeability, short half-life, local delivery of curcumin to the treatment target.
  • the current trend in curcumin research is to develop potential delivery systems to increase their water solubility and bioavailability because solubility acts as a rate limiting step for absorption.
  • the new method is to use other delivery vehicles, such as chelating strategies and bioconjugates of liposomes curcumin, curcumin nanoparticles, curcumin phospholipid complexes, nanoliposomes, nanoemulsions, nanolipids Granules and micelle nanosuspensions, glue Beams, nanoparticles, nanoemulsions, inclusion complexes with cyclodextrins, and curcumin structural analogs have all been confirmed.
  • the solubility in water can be increased by a few hundred times.
  • most of the reported methods only provide limited improvement in the bioavailability of curcumin.
  • cyclodextrin is used, the complexation process is slow, and the high molecular weight of the cyclodextrin and the pH of the treatment medium limit their practical utility.
  • Most delivery systems such as microemulsions, liposomes, and micelles, have a limited effect on powders because their stability can be affected when converted to a powder.
  • micelles, microemulsions, and liposome complexes may degrade in the stomach prior to reaching the target site, thereby impairing the bioavailability of the active ingredient.
  • Few researchers have focused on stability, solubility, and bioavailability at physiological pH. Because curcumin is prone to hydrolysis, stability in aqueous media is critical to the bioavailability of curcumin.
  • the present invention describes novel pharmaceutical formulations of curcumin based on different strategies, including:
  • the drug delivery system includes micelles, nanoparticles, nanofibers, nanosuspensions, and the like.
  • Nanotechnology is increasingly seen as a technology of the future.
  • nanoparticles are used to increase the bioavailability and solubility of lipophilic compounds, such as curcumin, in drug delivery systems.
  • lipophilic compounds such as curcumin
  • Solid dispersion technology is a science in which one or more active ingredients are dispersed in an inert matrix in a solid phase, intended to increase the solubility, dissolution rate, permeability, sustained release, altered solid state properties and stability of the drug. , thereby achieving improved bioavailability.
  • Lipid-based drug delivery systems have shown great potential for oral delivery of difficult-to-drug candidates, and there are several successfully marketed products.
  • the pre-dissolved drug is in a lipid, a surfactant, or a mixture of a lipid and a surfactant, omitting the dissolution/dissolution step, and the dissolution/dissolution step is a potential rate limiting factor for oral absorption of a poorly water-soluble drug.
  • the results improve bioavailability, bypassing the liver to reduce hepatotoxicity (via lymphatic absorption), and reducing nephrotoxicity (unknown mechanism).
  • the lipid-based drug delivery system includes a lipid solution, a lipid suspension, a self-emulsifying drug delivery system, a micelle, a nanoemulsion formulation.
  • the main object of the present invention is to effectively utilize curcumin by solving various problems such as poor solubility, chemical stability (hydrolysis, oxidation, heat and light) and pharmacokinetic properties including absorption, distribution, metabolism and elimination in vivo, and bioavailability.
  • curcumin Low degree, poor permeability, short half-life and local delivery of curcumin to the therapeutic target to achieve the best results, such as anti-oxidation, anti-inflammatory, anti-cancer, induced apoptosis, anti-angiogenesis, neuroprotection, anti- It has excellent pharmacological effects such as microbial agents, liver and kidney protection, inhibition of thrombosis, myocardial infarction, hypoglycemia and anti-rheumatic activities, and is effective in treating mammalian diseases.
  • the present invention provides a drug delivery system comprising the active ingredient curcumin or a derivative thereof or a pharmaceutically acceptable salt thereof and a polymer carrier Soluplus.
  • the weight ratio of curcumin to Soluplus is from 1:0.001 to 1:100.
  • the drug delivery system further comprises other polymeric carriers and/or surfactants.
  • the other polymeric carrier is a water soluble polymer selected from the group consisting of N-vinyl lactam homopolymers, N-vinyl lactam copolymers, cellulose esters, cellulose ethers, polyalkylene oxides , polyacrylate, polymethacrylate, homopolymer and copolymer of acrylic acid, homopolymer and copolymer of methacrylic acid, polyacrylamide, polyvinyl alcohol, vinyl acetate polymer, vinyl acetate copolymerization , carboxyvinyl polymers, oligosaccharides, polysaccharides, and mixtures thereof.
  • the other polymeric carrier is selected from the group consisting of alkyl cellulose, hydroxyalkyl cellulose, hydroxyalkyl alkyl cellulose, methyl cellulose (MC), ethyl cellulose (EC), hydroxyethyl Cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methylcellulose (HPMC), hydroxyethyl methylcellulose (HEMC), hydroxypropylmethylcellulose succinate, hydroxypropyl Methyl cellulose acetate succinate, carboxymethyl ethyl cellulose, sodium carboxymethyl cellulose, potassium carboxymethyl cellulose, cellulose acetate succinate, cellulose acetate phthalate, hydroxyl Propyl methylcellulose phthalate, polyacrylic acid copolymer, poly(meth)acrylic acid polymer, poly(hydroxyalkyl acrylate), poly(hydroxyalkyl methacrylate), polyethylene Pyrrolidone (PVP), vinylpyrrolidone homopolymer,
  • PVP
  • the other polymeric carrier is selected from the group consisting of hydroxypropyl methylcellulose (HPMC), polyethylene glycol (PEG), chitosan, PVP, PVP/VA, HPC, hydroxypropyl methylcellulose acetate (HPMCAS), eudragit E100, based on dimethylaminoethyl methacrylate a cationic copolymer of an ester, butyl methacrylate and methyl methacrylate.
  • HPMC hydroxypropyl methylcellulose
  • PEG polyethylene glycol
  • chitosan PVP
  • PVP/VA HPC
  • HPC hydroxypropyl methylcellulose acetate
  • HPC hydroxypropyl methylcellulose acetate
  • HPC hydroxypropyl methylcellulose acetate
  • HPC hydroxypropyl methylcellulose acetate
  • HPC hydroxypropyl methylcellulose acetate
  • HPC hydroxypropyl methylcellulose acetate
  • HPC hydroxypropy
  • the surfactant comprises a negative, positive or amphoteric surfactant and is selected from the group consisting of sodium dodecyl sulfate, sodium dodecyl sulfate (SDS), sodium lauryl sulfate (SLS), polyoxygen Ethylene sorbate long-chain fatty acid ester, vitamin E-TPGS, bile salt, sodium deoxycholate, sodium glycocholate, polyoxyethylene polyoxypropylene glycol, and combinations thereof.
  • SDS sodium dodecyl sulfate
  • SLS sodium lauryl sulfate
  • polyoxygen Ethylene sorbate long-chain fatty acid ester vitamin E-TPGS
  • bile salt sodium deoxycholate
  • sodium glycocholate sodium glycocholate
  • polyoxyethylene polyoxypropylene glycol polyoxyethylene polyoxypropylene glycol
  • the drug delivery system is selected from the group consisting of nanotechnology-based drug delivery systems, solid solution systems, and emulsion systems to provide additional stability for possible precipitation.
  • the nanotechnology-based drug delivery system is selected from the group consisting of micelles, nanoparticles, nanofibers, and nanosuspensions
  • the solid solution based system is selected from the group consisting of solid dispersions, extrudates, and solid support systems.
  • the drug delivery system is a solid dispersion formulation, preferably further comprising TPGS 1000.
  • the drug delivery system is a micelle formulation, preferably further comprising TPGS 1000.
  • the micelle formulation further comprises other polymeric carriers and water/buffering agents, and wherein an effective amount of curcumin is encapsulated in the micelles.
  • the micelles further comprise a surfactant, a solid phase adsorbent, an acidulant and/or an antioxidant.
  • the invention also provides a preparation method of a solid dispersion preparation, comprising the following steps:
  • the active ingredient curcumin or a derivative thereof, or a pharmaceutically acceptable salt thereof is dispersed in a polymeric carrier and optionally a surfactant.
  • a method of preparing a solid dispersion formulation comprising a further step selected from: melting ice bath with stirring, the film was cooled with liquid nitrogen, spray congealing, hot melt extrusion, Meltrex TM, melt agglomeration, or solvent evaporation (drying , vacuum drying, rotary evaporation, hot plate heating, spray drying, freeze drying, supercritical antisolvent, coprecipitation, electrospinning, spray lyophilization, ultra-fast lyophilization, fluid bed coating) and solvent melting.
  • the invention also provides a preparation method of micelles, comprising the following steps:
  • the active ingredient curcumin or a derivative thereof, or a pharmaceutically acceptable salt thereof, a polymer carrier and a surfactant are optionally dissolved in an organic solvent, and the organic solvent is removed by rotary evaporation. After the film is formed, it is vacuum dried and added to a buffer. The agent is hydrated and sonicated.
  • the present invention also provides a lipid-based drug delivery system comprising the active ingredient curcumin or a derivative thereof or a pharmaceutically acceptable salt thereof and a lipid.
  • the lipid is a triglyceride comprising long chain triglycerides (LCT), medium chain triglycerides (MCT) and short chain triglycerides (SCT), wherein the long chain triglycerides are selected from hydrogenation Soybean oil, hydrogenated vegetable oil, corn oil, olive oil, soybean oil, peanut oil and sesame oil, medium chain triglycerides are selected from caprylic/maronic acid triglycerides from cocoa butter or palm seed oil.
  • LCT long chain triglycerides
  • MCT medium chain triglycerides
  • SCT short chain triglycerides
  • the lipid-based drug delivery system further comprises an excipient selected from the group consisting of chemical triglycerides, partial triglycerides, semi-synthetic oily esters, and semi-synthetic nonionic surfactant esters.
  • the lipid-based drug delivery system further comprises a water-insoluble excipient selected from the group consisting of beeswax, oleic acid, soy fatty acid, vitamin E, corn oil mono-di-triglyceride, medium chain (C8/C10) glycerol Monoesters and diglycerides, as well as propylene glycol esters of fatty acids.
  • a water-insoluble excipient selected from the group consisting of beeswax, oleic acid, soy fatty acid, vitamin E, corn oil mono-di-triglyceride, medium chain (C8/C10) glycerol Monoesters and diglycerides, as well as propylene glycol esters of fatty acids.
  • the lipid is selected from Caproyl 90, Capmul MCM Caproyl TM and one or more of PGMC.
  • the lipid-based drug delivery system further comprises a water-soluble organic solvent, a surfactant, a co-surfactant, a polymer solubilizer, a phospholipid, and/or an additive.
  • the water-soluble organic solvent is selected from the group consisting of PEG 200-10,000, polyvinyl caprolactam (PCL), polyvinyl acetate (PVA) or a copolymer thereof, a water-soluble form of vitamin E and ethanol; and a surfactant in which the fatty acid is a derivative of an unsaturated or saturated dietary oil synthesized by reacting PEG with a hydrolyzed vegetable oil, reacting an alcohol with ethylene oxide to form an alkyl ether ethoxylate, or reacting a polysorbate-based vegetable oil with ethylene oxide;
  • the agent is based on polyethylene glycol, polypropylene glycol, ethanol and glycerin;
  • the polymer solubilizer is selected from the group consisting of Soluplus, chitosan, polyvinylpyrrolidone (PVP), PVP/VA, HPC, HPMC, HPMCAS, eudragit E100, based on methacrylic acid A cationic cop
  • PEG 200-10,000 is selected from the group consisting of PEG 300, PEG 400, PEG 1,000 and PEG 6,000; the surfactant is selected from the group consisting of Cremophor RH 40, Labrasol, TPGS 1000, Tween 20, Cremophor E1 and Tween 80; and co-surfactant selection From PEG 300, PEG 400, propylene glycol, glycerol, ethanol, Transcutol HP and Transcutol P.
  • the additive comprises a solid phase adsorbent, a water soluble and fat soluble antioxidant, an acidulant, a chelating agent, a preservative, a stabilizer and/or a buffer
  • the solid phase adsorbent comprises a silicon based adsorbent and a silicon-based adsorbent
  • the silicon-based adsorbent is selected from the group consisting of Aerosil 200 and magnesium aluminum metasilicate
  • the non-silicon-based adsorbent is selected from the group consisting of microcrystalline cellulose, talc, anhydrous dibasic calcium phosphate (DCPA), and alkyl cellulose.
  • DCPA dibasic calcium phosphate
  • a water-soluble polymer composed of a group such as a hydroxyalkyl cellulose or a hydroxyalkylalkyl cellulose sugar;
  • the chelating agent is selected from the group consisting of ethylenediamine, disodium edetate, and disodium edetate.
  • the acidifying agent is selected from the group consisting of citric acid, acetic acid, fumaric acid, hydrochloric acid and nitric acid
  • the buffering agent is selected from the group consisting of potassium metaphosphate, potassium dihydrogen phosphate, sodium acetate, sodium citrate
  • the water-soluble or fat-soluble antioxidant is selected from the group consisting of Ascorbic acid, ascorbyl palmitate, butylhydroxyanisole, butylhydroxytoluene, hypophosphorous acid, thioglycerol, propyl gallate, sodium ascorbate, sodium hydrogen sulfite, sodium formaldehyde sulfoxylate, sulfoxylate, sodium metabisulfite .
  • lipid-based drug delivery system further comprises Capryol TM PGMC, RH 40, Labrasol, TPGS 1000, Transcutol P and/or Aerosil 200.
  • the lipid-based drug delivery system is selected from the group consisting of a lipid solution, a liposome suspension, a surfactant or a polymer-lipid mixed micelle, a self-microemulsifying drug delivery system (SMEDDS) and a nanoemulsion formulation. .
  • the lipid based drug delivery system is a liquid phase or solid phase SMEDDS.
  • a solid phase adsorbent is also included, preferably Aerosil 200.
  • the lipid based drug delivery system is a nanoemulsion formulation which further comprises water and/or a buffer.
  • the invention also provides a method of preparing a lipid-based drug delivery system comprising the following steps:
  • the active ingredient curcumin or a pharmaceutically acceptable salt thereof is dissolved in a lipid, a surfactant, or a mixture of a lipid and a surfactant.
  • the drug delivery system is in the form of a solid dosage form selected from the group consisting of tablets, rings, patches, capsules, pills, granules, fine granules or powders, powders or strips, by oral, parenteral, inhalation. , local or percutaneous, nasal, intraocular, ear, rectal, vaginal route.
  • the drug delivery system is in the form of a liquid, selected from the group consisting of solutions, suspensions, emulsions, cosolvent-based systems, aerosols, by oral, parenteral, inhalation, topical or transdermal, intranasal, intraocular, aural, Rectal, vaginal route of administration.
  • the drug delivery system is a semi-solid dosage form selected from the group consisting of ointments, creams, gels, pastes, for topical or systemic purposes by topical or transdermal, rectal, vaginal routes.
  • the drug delivery system further comprises a pharmaceutically acceptable excipient selected from the group consisting of a disintegrant, a lubricant, a glidant, an anti-adherent, an inert filler, a wetting agent, a pH modifier. , binders, solubility modifiers, recrystallization inhibitors, diluents, and combinations thereof.
  • curcumin is contained in a liquid preparation of a drug delivery system in an amount of 0.001 to 1000 mg/ml, or 0.1 to 100 mg/ml, or 10 to 20 mg/ml, and in a solid preparation of a drug delivery system, curcumin
  • the dose is from 0.001 to 1000 mg/unit, or from 0.1 to 100 mg/unit, or from 10 to 20 mg/unit.
  • the drug delivery system of the present invention can be used to treat a variety of diseases and to select a suitable route of administration and dosage form according to the type of disease. Therefore, the present invention also provides a drug delivery system for preparing for anti-oxidation, anti-inflammatory, anti-cancer, inducing apoptosis, anti-angiogenesis, neuroprotection, anti-microbial, liver and kidney protection, inhibiting blood vessel formation, preventing myocardial infarction, Application in blood sugar lowering and anti-rheumatic drugs.
  • Figure 4 shows the DSC thermogram of curcumin, physical mixture (PM) and solid dispersion (Example 1).
  • Figure 5 shows the X-ray diffraction pattern of curcumin, Soluplus, physical mixture (PM) and solid dispersion (Example 1).
  • Figure 6 shows SEM micrographs of curcumin, Soluplus and solid dispersion (Example 1).
  • Figure 7 shows the measurement of the size distribution of curcumin micelles (Example 3) by dynamic light scattering.
  • Figure 12 shows fluorescent micrographs of SH-SY5Y-APP695 cells overexpressing APP treated with curcumin and NCF (Example 1), where A is native curcumin and B is NCF.
  • Example 9 Lipid-based nanoemulsion formulation
  • Example 10 Lipid-based nanoemulsion formulation
  • the required amount of curcumin, Soluplus and optionally TPGS 1000 were dissolved in ethanol.
  • the organic solvent was removed by a Buchi rotary evaporator II.
  • the formed film was dried overnight in a vacuum desiccator.
  • the dried sample was scraped from the flask and collected in a mortar.
  • the powder was crushed with a mortar and made into a uniform form.
  • curcumin, Soluplus and optionally TPGS 1000 were dissolved in ethanol according to Examples 3 and 4.
  • the organic solvent was removed by a Buchi rotary evaporator II.
  • the resulting membrane was dried overnight in a vacuum desiccator, then hydrated with 10 ml of 1 x PBS buffer (pH 7.4), incubated at 37 ° C for 30 minutes, and then sonicated for a few minutes.
  • the resulting mixture was filtered through a 0.45 ⁇ m syringe filter (PVDF).
  • Preparation 3 Preparation of liquid self-microemulsifying drug delivery system (SMEDDS) formulations (Examples 5 and 7)
  • the required amount of oil (Capmul PGMC), surfactant (Cremophor RH 40, Labrasol and TPGS 1000) and co-surfactant (Transcutol P) were accurately weighed into small glass vials. Then, the above components were mixed by gentle stirring and vortex mixing, and heated at 37 ° C in an incubator. Add the required amount of curcumin and vortex to mix until the curcumin is completely dissolved.
  • a liquid SMEDDS formulation was prepared as above. After the required amount of Aerosil 200 was added, it was diluted with a minimum amount of miliQ water and stirred at room temperature for 2 hours. The resulting mixture was allowed to stand for 15 minutes, equilibrated and filtered through a 0.45 ⁇ m syringe filter (PVDF). Prior to lyophilization, the solution was frozen at -80 °C for at least 6 hours and then lyophilized for at least 24 hours at -45 ° C and 7102 mbar pressure in Novalyphe-NL 500 (Savant Instruments Corp., Holbrook, NY). Finally, the solid phase SMEDDS is stored in a desiccator.
  • PVDF 0.45 ⁇ m syringe filter
  • the required amount of oil (Capmul PGMC), surfactant (Cremophor RH 40, Labrasol and TPGS 1000) and co-surfactant (Transcutol P) were accurately weighed into small glass vials. Then, the above components were mixed by gentle stirring and vortex mixing, and heated at 37 ° C in an incubator. Add the required amount of curcumin and vortex to mix until the curcumin is completely dissolved. The required amount of miliQ water was added dropwise until a clear, clear formulation was obtained.
  • Examples 1-10 provide a variety of different curcumin formulation formulations, including solid dispersions, micelles, SMEDDS, and nanoemulsion formulations, respectively. The advantages of these formulations are detailed below by way of effect examples.
  • Sample analysis was performed on a HPLC (Shimadzu, Kyoto, Japan) system equipped with a UV-VIS detector [SPD-20A], a DGU-20A3 online degasser, a CBM-20A system controller, and a SIL-20AHT automatic addition.
  • Sampler, and LC Chromopac data processor solution The column was analyzed using a Zorbax Eclipse XDB-C18 (4.6*150*3.5 mm 3 ).
  • the mobile phase of the sample analysis consisted of acetonitrile and 1% (w/v) citrate buffer at a ratio of 70:30 (v/v).
  • the injection volume was 20 ⁇ l, the flow rate was 1 ml/min, and the detection wavelength was 423 nm.
  • Simulated gastrointestinal fluid (no enzyme and bile components) was prepared according to the USP method.
  • curcumin significantly degrades under neutral to alkaline pH conditions and remains substantially stable at acidic pH. Curcumin is more stable at low pH 1.2 compared to pH 6.8 and 7.4. Curcumin exists in an equilibrium form between diketone and keto-enol, and easily forms intramolecular H-bonds. Curcumin hydrolysis begins with the attack of a nucleophilic OH - ion on the carbonyl group in the keto-enol moiety. Therefore, the greater the rate of degradation observed at high pH. In contrast, the solid dispersion of the invention (Example 1) protects curcumin from degradation in biological media of different pH.
  • curcumin was relatively stable in buffers containing solid dispersions at pH 1.2, 6.8 and 7.4. This was confirmed, encapsulated polymer micelle hydrolysis prevention curcumin, curcumin because keto - enol portion from nucleophilic OH - ions attack. Studies on the chemical stability of curcumin have inferred that the degradation of curcumin is a complex mechanism involving a variety of potential factors.
  • Table 2 shows the total degradation rate of curcumin in different samples according to the second-order kinetics of the solid dispersion of the present invention and natural curcumin at different pH values.
  • DSC Differential Scanning Calorimetry
  • the composition of the physical mixture (PM) was the same as that of the solid dispersion (SD) except that the drug (curcumin) and the polymer (Soluplus) were simply prepared in a ceramic mortar. The mixture was then sieved (250 ⁇ m) and stored in a container of amber glass lid.
  • X-ray diffraction confirmed the solid state characteristics of the sample.
  • the crystalline form of curcumin showed a sharp diffractive characteristic peak confirming the crystal form of the initial form.
  • a less intense peak was observed compared to the crystalline curcumin, suggesting that the crystal was partially converted to an amorphous state.
  • Soluplus no peaks were observed, indicating amorphous nature.
  • the XRD pattern of the solid dispersion did not show crystallinity, confirming the DSC results, in which no endothermic phenomenon corresponding to the melting of curcumin was recorded.
  • the Zeiss Microscopy Merlin with GEMINI II column is equipped with a field emission gun and operates at 0.7kV for secondary electron imaging.
  • Samples of curcumin, Soluplus, and solid dispersion (Example 1) were mounted on SEM scaffolds with conductive double-sided tape.
  • the particle size, polydispersity index (PDI) and zeta potential of the novel curcumin preparation (NCF) (Example 3) prepared were measured using a Malvern Zeta Sizer Nano ZS. For the test, a 1 mg/ml solution was prepared, followed by dilution with MiliQ water at 25 ° C (100 ⁇ l, up to 1 ml). Then, the particle size, PDI, and Zeta potential were triple measured by the above scheme.
  • the load capacity is defined as the weight ratio of curcumin to Soluplus, and the loading efficiency is defined as the ratio of the loaded curcumin to the initial amount of curcumin. Stability assessment was performed to check for turbidity, clarity, and precipitation at various time points after preparation. Curcumin was quantitatively determined by HPLC triple analysis. All tests were repeated three times and the results were expressed as mean and standard deviation.
  • a particle size of 63 nm was observed, indicating potential for use as a nanotechnology system with a PDI of 0.09, indicating a small variation in the size of the different particles, and a zeta potential of -8.65, indicating its potential to remain stable over time.
  • NCF showed high load capacity (9.15%) and load efficiency (98.23%).
  • the present application also confirmed stability by turbidity, transparency, and precipitation evaluation.
  • the size distribution of the micelle preparation of the present invention was determined by dynamic light scattering.
  • a universal buffer was prepared from a solution having a composition of boric acid, citric acid, and phosphoric acid (0.04 M each). The pH of the final solution was adjusted by the addition of 0.2 M sodium hydroxide. The concentration of the novel curcumin preparation (NCF) (Example 3) in different buffer solutions (pH 1.8-8) was 100 ⁇ g/mL. The solution was incubated at room temperature in the dark to avoid photolysis. Samples were taken at predetermined time intervals and filtered through a 0.45 ⁇ m PVDF syringe filter. All samples were subjected to triple analysis by HPLC method.
  • NCF novel curcumin preparation
  • Example 3 A solution of 0.02% H 2 O 2 and 3% H 2 O 2 of a novel curcumin preparation (NCF) (Example 3) at a concentration of 100 ⁇ g/mL was prepared. The solution was incubated at room temperature in the dark to avoid photolysis. Samples were taken at predetermined time intervals and filtered through a 0.45 ⁇ m PVDF syringe filter. All samples were subjected to triple analysis by HPLC method.
  • NCF novel curcumin preparation
  • Example 3 A solution of a novel curcumin preparation (NCF) (Example 3) at a concentration of 100 ⁇ g/mL was prepared. The solution was incubated in a stable chamber at 4 ° C, 25 ° C and 40 ° C. Samples were taken at predetermined time intervals and filtered through a 0.45 ⁇ m PVDF syringe filter. All samples were subjected to triple analysis by HPLC method.
  • NCF novel curcumin preparation
  • NCF novel curcumin preparation
  • Example 3 A solution of a novel curcumin preparation (NCF) (Example 3) at a concentration of 100 ⁇ g/mL was prepared. The solution was incubated in a light stable chamber according to the International Conference on Harmonization (ICH) guidelines. Samples were taken at predetermined time intervals and filtered through a 0.45 ⁇ m PVDF syringe filter. All samples were subjected to triple analysis by HPLC method.
  • ICH International Conference on Harmonization
  • DMEM medium Dulbecco's Modified Eagle Medium
  • FBS fetal bovine serum
  • penicillin-streptavidin Prime solution.
  • the cells were cultured in an incubator at 37 ° C under 5% CO 2 .
  • SH-5YSY cells were seeded at a density of 5 x 10 3 cells/well in 96-well plates. After 24 hours, the original medium was replaced with a medium containing 10 ⁇ g/mL equivalent of unprocessed curcumin or a novel curcumin preparation (NCF) (Example 1) and Soluplus. The preparation was prepared using sterile water. Cell viability was measured by MTT ([3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide], thiazolyl blue) method. After 20 hours, 20 ⁇ L of MTT (Sigma-Aldrich, USA, 5 mg/ml in PBS) was added to each well and incubated for 1 hour.
  • MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide]
  • DMSO dimethyl methacrylate
  • DMEM medium Dulbecco's Modified Eagle Medium
  • FBS fetal bovine serum
  • penicillin-streptavidin Prime solution.
  • the cells were cultured in an incubator at 37 ° C under 5% CO 2 .
  • SH-5YSY-695 cells were seeded at a density of 5 x 10 3 cells/well in 96-well plates. After 24 hours, the original medium was replaced with a medium containing 10 ⁇ g/mL of unprocessed curcumin or a novel curcumin preparation (NCF) (Example 1), CuSO 4 and H 2 O 2 to induce the representative of Alz. Cytotoxicity of Haimer's disease. The preparation was prepared using sterile water. Cell viability was measured by MTT ([3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide], thiazolyl blue) method.
  • MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide]
  • MTT Sigma-Aldrich, USA, 5 mg/ml PBS
  • 150 ⁇ L of DMSO was added to dissolve the insoluble purple formazan product to produce a colored solution.
  • the optical density (OD) was read at a wavelength of 600 nm on a multi-hole scanning spectrophotometer (BIO-RAD model 2550 EIA reader).
  • SH-SY5Y-APP695 cells were seeded at a density of 5 x 10 4 cells/well in 24-well plates (Corning, NY, USA). After 24 hours of incubation at 37 ° C, adherent cells were treated with or without natural curcumin at a concentration of 10 ⁇ g/ml and at an equal concentration of NCF and maintained at 37 ° C in a cell culture device (Hera Cell, Thermo Scientific, Waltham, MA). in. After 6 hours, the cells were washed twice with PBS (0.01 M, pH 7.4) and methanol was added for lysis.
  • PBS 0.01 M, pH 7.4
  • the cell lysate was centrifuged at 10,000 rpm for 10 minutes at 4 °C.
  • the concentration of curcumin in the collected supernatant was measured by LC/MS/MS. Each measurement was repeated three times and the obtained data was expressed as the average of three experiments.
  • Mobile phase A was an aqueous solution of 5% methanol and 0.1% formic acid
  • mobile phase B was an aqueous solution of 95% methanol and 0.1% formic acid.
  • the gradient set by the mobile phase schedule was: starting 10% MPB, 100% MPB at 1.5 minutes, maintaining 95% MPB for 6 minutes, then 10% MPB for 30 seconds, preparing the next sample.
  • the total run time for each sample analysis was 10 minutes.
  • the column eluate was introduced into negative ion mode electrospray (ESI) mass spectrometry.
  • the operational parameters of the ion source include analyte dependent parameters and source dependent parameters, optimized for optimal performance from mass spectrometer analysis.
  • the mass-to-charge ratio (m/z) was generated by monitoring the precursor ions for MRM analysis as follows: curcumin 367.6/132.20 and Fahualin 307.2/161.2. Zero air is used as the source gas, and nitrogen is used as both the curtain gas and the collision gas.
  • the peak area was obtained from the compound, and the internal standard (IS) and the calibrant of known concentration were used as a calibration curve for constructing the ratio of compound/IS area.
  • the limit of quantification is 5 ng/ml.
  • the intra- and inter-day variability of each compound was within 15%.
  • the SH-SY5Y-APP695 cells at a density of 15 ⁇ 10 4 cells / well were seeded in 35mm culture plates (Corning, NY, USA), for, for fluorescence microscopy. After incubation for 24 hours at 37 ° C, adherent cells were incubated with a constant concentration (10 ⁇ g/ml) of natural curcumin and an equal concentration of NCF (Example 1) at 37 ° C in a cell culture device (Hera Cell, Thermo Scientific, Waltham, MA). Processing in 2 hours. After the incubation, the cell monolayer was washed three times with 1 ml of PBS (0.01 M, pH 7.4) to remove excess solid dispersion (SD) or natural curcumin. Fresh PBS (0.01 M, pH 7.4) was added to the plate, and the cells were observed, and photos were photographed by exciting the curcumin with a blue light microscope.
  • PBS 0.01 M, pH 7.4
  • the unprocessed curcumin remained substantially insoluble for 2 h in the dissolution medium.
  • the dissolution of the drug in the PM is slightly higher due to the solubilization of the drug due to micellization.
  • the solid dispersion (SD) (Example 1) showed a significantly higher release rate than PM. This suggests that curcumin exists mainly in amorphous form in SD and thus has higher solubility. Within 120 minutes, the dissolution rate of SD was observed to be 100%.
  • Rats Male Sprague-Dawley rats (250 ⁇ 10 g) were obtained at least 1 week prior to the start of the trial to regulate the environment, food and water in the laboratory. Rats were anesthetized before surgery A longitudinal incision is made in the neck and closer to the jugular vein area. Subsequently, the catheter was filled with 20 units/ml of heparin saline and inserted into the jugular vein until the first silica gel plug. Stitch the rubber plug and muscle to secure it there. The other end of the catheter is subcutaneously through the neck, closer to the ears. Finally, the catheter was filled with 500 units/ml heparin saline and inserted into the free end of the catheter. After the surgery was completed, the rats were placed in different cages to recover. The next day, a pharmacokinetic study was performed on each rat. Prior to dosing, the animals were fasted for 12 hours with free access to drinking water.
  • the curcumin suspension (CS) was prepared by adding curcumin to a 0.5% sodium carboxymethylcellulose (CMC-Na) solution and then sonicating for a few minutes to obtain a homogeneous suspension.
  • the new curcumin formulation (NCF) (Example 1) was dissolved in mili Q water. Two groups of rats were orally administered with curcumin suspension and NCF at a dose equivalent to 50 mg/kg of curcumin. After administration of the drug and formulation by oral gavage, 0.2 ml of blood samples were taken at time intervals of 0, 15, 30, 45, 60, 90, 120, 180, 240, 300, 360, 420, 480 and 720 minutes.
  • the catheter was flushed with the same amount of heparin saline each time the blood sample was collected. After the blood sample was collected, it was centrifuged at 5000 rpm and 4 ° C for 5 minutes to separate the plasma from the blood. Plasma was stored at -20 °C after separation until analysis. 900 ⁇ l of ice-cold methanol containing warfarin was added as an internal standard (400 ng/ml) to 100 ⁇ L of plasma sample, followed by shaking for 10 minutes, centrifugation at 13,000 rpm for 5 minutes, and then drying with nitrogen. The extract was reconstituted with methanol/water (50:50) prior to injection into LC/MS/MS and filtered through a 0.22 [mu]m membrane filter. Using Phoenix WinNonlin (Pharsight, St. Louis, MO) Noncompartmental pharmacokinetics analysis of each concentration-time characteristic.

Abstract

Curcumin pharmaceutical preparations, including a solid dispersion, a micelle preparation, SMEDDS and a nanoemulsion. By increasing the solubility and bioavailability of curcumin, curcumin can be used more effectively for the pharmacological effects such as antioxidation, anti-inflammation, anti-cancer, inducing apoptosis, anti-angiogenesis, neuroprotection, antimicrobial, protecting liver and kidney, inhibiting vascularization, preventing myocardial infarction, reducing blood glucose level, anti-rheumatism and the like.

Description

姜黄素药物制剂Curcumin pharmaceutical preparation 技术领域Technical field
本发明属于药物领域。具体地,本发明涉及新型姜黄素药物制剂。该新型制剂能更有效地利用姜黄素抗氧化、抗炎、抗癌、诱导细胞凋亡、抗血管生成、神经保护、抗微生物、保肝护肾、抑制血管形成、预防心梗、降血糖、抗风湿等药理作用。The invention belongs to the field of medicine. In particular, the invention relates to novel curcumin pharmaceutical formulations. The novel preparation can more effectively utilize curcumin for anti-oxidation, anti-inflammatory, anti-cancer, induce apoptosis, anti-angiogenesis, neuroprotection, anti-microbial, liver and kidney protection, inhibit blood vessel formation, prevent myocardial infarction, lower blood sugar, Anti-rheumatic and other pharmacological effects.
背景技术Background technique
姜黄素(Curcumin),无疑是迄今为止自然界发现的最具生物活性的分子之一。姜黄素以双相剂量依赖的方式与大量的胞内胞外靶相互作用。它控制炎性、氧化应激、细胞存活、细胞分泌、体内平衡和增殖。其作用机理大致指向表现出无序生理学或明显突变的异常状态的细胞。它可以轻松地越过所有的生理屏障,包括血脑屏障。Curcumin is undoubtedly one of the most biologically active molecules discovered in nature to date. Curcumin interacts with a large number of intracellular extracellular targets in a biphasic dose-dependent manner. It controls inflammatory, oxidative stress, cell survival, cell secretion, homeostasis and proliferation. The mechanism of action generally points to cells that exhibit disordered physiology or abnormal states of apparent mutation. It can easily cross all physiological barriers, including the blood-brain barrier.
临床试验中,口服12g/天,姜黄素没有任何副作用。姜黄素和姜黄产品已被美国食品和药物管理局(FDA)、加拿大天然健康产品局和***粮农组织/世界卫生组织联合专家委员会定性为安全的食品添加剂。In clinical trials, curcumin did not have any side effects when administered orally at 12 g/day. Curcumin and turmeric products have been identified as safe food additives by the US Food and Drug Administration (FDA), the Canadian Natural Health Products Agency, and the Joint FAO/WHO Expert Committee.
姜黄素是一种天然多酚类化合物,商业上分离自姜黄属(Curcuma longa Linn)(姜黄)(姜科)的根茎,在阿育吠陀医药中有着悠久的历史。许多亚洲国家,如印度和中国,已广泛用作草药数千年。姜黄素的化学名称为1,7-双-(4-羟基-3-甲氧基苯基)-庚-1,6-二烯-3,5-二酮,化学式为C21H20O6。类姜黄素称为二阿魏酰基甲烷(Diferuloyl Methane),是姜黄素的主要成分(77重量%);其他两个姜黄素是脱甲氧姜黄素(17重量%)和双脱甲氧姜黄素(3重量%)。姜黄素是一种脂溶性多酚化合物,结构上由连接到二酮结构中的两个环甲氧基苯酚组成。酮-烯醇互变异构允许姜黄素充当迈克尔(Michael)受体4。 酚基和二酮属于抗氧化剂化合物的特性,并且是姜黄素抗氧化作用的关键结构。Curcumin is a natural polyphenolic compound that is commercially isolated from the rhizome of Curcuma longa Linn (Curcuma) (Zingiberaceae) and has a long history in Ayurvedic medicine. Many Asian countries, such as India and China, have been widely used as herbs for thousands of years. The chemical name of curcumin is 1,7-bis-(4-hydroxy-3-methoxyphenyl)-hepta-1,6-diene-3,5-dione, the chemical formula is C 21 H 20 O 6 . Curcumin-like is called diferuloyl Methane and is the main component of curcumin (77% by weight); the other two curcumin are demethoxycurcumin (17% by weight) and bis-methoxycurcumin (3 wt%). Curcumin is a fat-soluble polyphenolic compound consisting structurally of two cyclomethoxyphenols attached to a diketone structure. Keto-enol tautomerization allows curcumin to act as a Michael acceptor 4 . Phenol groups and diketones are characteristic of antioxidant compounds and are key structures for the antioxidant action of curcumin.
姜黄素安全、耐受性良好。由于其药效和安全性,姜黄素已在广泛的研究领域,体外和体内、动物和人中进行了研究。低的口服生物利用度(大鼠仅1%)和非常短的生物半衰期是姜黄素临床开发的限制因素。但由于其亲脂性,姜黄素可以自由地通过细胞膜(log P 1/42.5)。姜黄素生物利用度低的主要原因是其在水中(在pH 5.0的水溶液中,仅为11ng/ml)、酸性和生理pH值的溶解度极低,以及碱性条件下快速水解。在人类中,从药物动力学的角度来看,口服给药后,姜黄素的血清水平1-2小时后达到峰值,剂量为4,000mg/天时,血清浓度范围为0.51±0.11μM,而剂量为8,000mg/天时,血清浓度范围为1.77±1.87uM。环境因素,如光,也会快速分解姜黄素,导致其临床上难以处理。为了实现药物施用的最佳结果,减轻病症通常需要定制给药方法,才能使药物以治疗量到达病变部位。优化预防或治疗性应用需要提供适量的姜黄素至受损的细胞靶标。最后,治疗效果可以伴随意外毒性至周围正常健康的细胞和组织。生物活性化合物的理想特性是在含水的体液中溶解度。姜黄素等亲脂性化合物缺乏水溶性,但保留显著的细胞膜或细胞内活性。它的应用有必要采用新的制剂。因此,医药工业的主要挑战之一是应用策略来开发此类问题化合物,转化成临床上口服、生物可利用和治疗有效的药物。Curcumin is safe and well tolerated. Due to its efficacy and safety, curcumin has been studied in a wide range of research fields, in vitro and in vivo, in animals and in humans. Low oral bioavailability (only 1% in rats) and very short biological half-life are limiting factors in the clinical development of curcumin. However, due to its lipophilic nature, curcumin can pass freely through the cell membrane (log P 1/42.5). The main reason for the low bioavailability of curcumin is its very low solubility in water (only 11 ng/ml in an aqueous solution of pH 5.0), acidity and physiological pH, and rapid hydrolysis under alkaline conditions. In humans, from the point of pharmacokinetics, the serum level of curcumin peaked after 1-2 hours after oral administration, and the serum concentration ranged from 0.51 ± 0.11 μM at a dose of 4,000 mg/day. At 8,000 mg/day, the serum concentration ranged from 1.77 ± 1.87 uM. Environmental factors, such as light, can also rapidly break down curcumin, making it clinically difficult to handle. In order to achieve optimal results for drug administration, alleviation of the condition usually requires a tailored method of administration in order for the drug to reach the lesion in a therapeutic amount. Optimizing prophylactic or therapeutic applications requires the provision of an appropriate amount of curcumin to impaired cellular targets. Finally, the therapeutic effect can be accompanied by accidental toxicity to surrounding normal healthy cells and tissues. The desirable property of a biologically active compound is its solubility in aqueous body fluids. Lipophilic compounds such as curcumin lack water solubility, but retain significant cell membrane or intracellular activity. It is necessary to use new formulations for its application. Therefore, one of the main challenges of the pharmaceutical industry is the application of strategies to develop such problematic compounds into clinically orally, bioavailable and therapeutically effective drugs.
为了开发姜黄素作为临床药物,理想的制剂应解决如下问题:差的溶解度、化学稳定性(水解、氧化、热和光)和药代动力学特性,包括吸收、分布、代谢和体内消除,生物利用度低,渗透性差,半衰期短,局部递送姜黄素至治疗靶位差。当前姜黄素研究的趋势是开发潜在的递送***,以提高其水溶解度和生物利用度,因为溶解度充当了吸收的限速步骤。为此目的,新方法是使用其它递送载体,像螯合策略和生物缀合物的脂质体姜黄素,姜黄素纳米颗粒,姜黄素磷脂复合物,纳米脂质体,纳米乳剂,纳米脂质颗粒和胶束纳米混悬剂,胶 束,纳米颗粒,纳米乳液,与环糊精形成包合物,以及姜黄素结构类似物,都已被确证。使用这些方法,水中溶解度可以增加几百倍。但大多数报道的方法仅提供姜黄素的生物利用度以有限的改善。如采用环糊精,络合进程缓慢,环糊精的高分子量和处理介质的pH值都限制它们的实际效用。大多数递送***,例如微乳剂、脂质体和胶束对粉剂作用有限,这是因为转换成粉末时其稳定性会受到影响。而且,胶束、微乳剂和脂质体复合物在抵达靶位之前有可能先在胃中降解,从而损害活性成分的生物利用度。很少有研究者关注生理pH条件下的稳定性、溶解度和生物利用度,由于姜黄素易于水解,水性介质中的稳定性对姜黄素的生物利用度至关重要。还有少量研究涉及避光,对光非常敏感可能成为药物商业开发的主要问题之一。有效地向治疗靶位递送药物,并没有引起研究者太多的注意力,而靶向癌症或脑部相关疾病是必要的。迄今为止,只有少数研究人员开展研究,并且其中一些使用纳米技术得到了成功。纳米颗粒的潜力不仅通过增加其生物利用度提高了每个剂量的治疗或成像造影制剂的功效,而且也被修改为针对肿瘤细胞的靶向选择性,从而增加图像分辨率和/或减少与当前化疗相关的脱靶毒性。In order to develop curcumin as a clinical drug, the ideal formulation should address the following problems: poor solubility, chemical stability (hydrolysis, oxidation, heat and light) and pharmacokinetic properties, including absorption, distribution, metabolism and elimination in vivo, bioavailability Low degree, poor permeability, short half-life, local delivery of curcumin to the treatment target. The current trend in curcumin research is to develop potential delivery systems to increase their water solubility and bioavailability because solubility acts as a rate limiting step for absorption. For this purpose, the new method is to use other delivery vehicles, such as chelating strategies and bioconjugates of liposomes curcumin, curcumin nanoparticles, curcumin phospholipid complexes, nanoliposomes, nanoemulsions, nanolipids Granules and micelle nanosuspensions, glue Beams, nanoparticles, nanoemulsions, inclusion complexes with cyclodextrins, and curcumin structural analogs have all been confirmed. Using these methods, the solubility in water can be increased by a few hundred times. However, most of the reported methods only provide limited improvement in the bioavailability of curcumin. If cyclodextrin is used, the complexation process is slow, and the high molecular weight of the cyclodextrin and the pH of the treatment medium limit their practical utility. Most delivery systems, such as microemulsions, liposomes, and micelles, have a limited effect on powders because their stability can be affected when converted to a powder. Moreover, micelles, microemulsions, and liposome complexes may degrade in the stomach prior to reaching the target site, thereby impairing the bioavailability of the active ingredient. Few researchers have focused on stability, solubility, and bioavailability at physiological pH. Because curcumin is prone to hydrolysis, stability in aqueous media is critical to the bioavailability of curcumin. There are also a small number of studies involving darkness, which is very sensitive to light and may be one of the main problems in the commercial development of drugs. Effective delivery of drugs to therapeutic targets does not cause the researchers to pay too much attention, and targeting cancer or brain-related diseases is necessary. To date, only a few researchers have conducted research, and some of them have succeeded in using nanotechnology. The potential of nanoparticles not only increases the efficacy of each dose of therapeutic or imaging contrast preparation by increasing its bioavailability, but is also modified to target selectivity for tumor cells, thereby increasing image resolution and/or reducing current Chemotherapy-related off-target toxicity.
本领域仍存在迫切需求,通过选择合适的剂型,改进姜黄素的功效。There is still an urgent need in the art to improve the efficacy of curcumin by selecting a suitable dosage form.
发明概述Summary of invention
本发明描述了基于不同策略的姜黄素新型药物制剂,所述策略包括:The present invention describes novel pharmaceutical formulations of curcumin based on different strategies, including:
1.纳米技术药物递送***1. Nanotechnology drug delivery system
该药物递送***包括胶束、纳米颗粒、纳米纤维和纳米悬浮液等。纳米技术越来越被认为是未来的技术。在纳米技术的广泛应用中,将纳米颗粒用于提高亲脂性化合物,如姜黄素,在药物递送***中的生物利用度和溶解度。因此,纳米颗粒在过去十年受到巨大欢迎,原因是通过保护药物免受酶降解,提供药物控释和延长的血液循环,改变 药物动力学,降低药物毒性,并限制药物的非特异性吸收,从而潜在改善封装药物的疗效。The drug delivery system includes micelles, nanoparticles, nanofibers, nanosuspensions, and the like. Nanotechnology is increasingly seen as a technology of the future. In a wide range of applications in nanotechnology, nanoparticles are used to increase the bioavailability and solubility of lipophilic compounds, such as curcumin, in drug delivery systems. As a result, nanoparticles have been greatly welcomed over the past decade due to the protection of drugs from enzymatic degradation, drug controlled release and prolonged blood circulation, Pharmacokinetics, reduce drug toxicity, and limit non-specific absorption of drugs, potentially improving the efficacy of encapsulated drugs.
2.固体分散体的药物递送***2. Solid dispersion drug delivery system
固体分散体技术是在固体阶段将一种或多种活性成分分散在惰性基质中的一门科学,旨在通过增加药物的溶解度、溶出速率、渗透性、缓释、改变的固态性质和稳定性,进而实现改进的生物利用度。Solid dispersion technology is a science in which one or more active ingredients are dispersed in an inert matrix in a solid phase, intended to increase the solubility, dissolution rate, permeability, sustained release, altered solid state properties and stability of the drug. , thereby achieving improved bioavailability.
3.基于脂质的药物递送***3. Lipid-based drug delivery system
基于脂质的药物递送***已在口服递送难以成药的候选物方面显示出巨大的潜力,并有数个成功上市的产品。预溶解药物在脂质、表面活性剂,或脂质和表面活性剂的混合物,省略了溶解/溶出步骤,而溶解/溶出步骤对口服吸收水溶性差的药物而言是一个潜在的限速因素,其结果改善了生物利用度,绕道肝脏减少了肝毒(经淋巴吸收),并且能够减少肾毒(机制不明)。所述基于脂质的药物递送***包括脂质溶液、脂质悬浮液、自乳化药物递送***、胶束、纳米乳剂制剂。Lipid-based drug delivery systems have shown great potential for oral delivery of difficult-to-drug candidates, and there are several successfully marketed products. The pre-dissolved drug is in a lipid, a surfactant, or a mixture of a lipid and a surfactant, omitting the dissolution/dissolution step, and the dissolution/dissolution step is a potential rate limiting factor for oral absorption of a poorly water-soluble drug. The results improve bioavailability, bypassing the liver to reduce hepatotoxicity (via lymphatic absorption), and reducing nephrotoxicity (unknown mechanism). The lipid-based drug delivery system includes a lipid solution, a lipid suspension, a self-emulsifying drug delivery system, a micelle, a nanoemulsion formulation.
本发明的主要目的是有效利用姜黄素,通过解决如下诸多问题:差的溶解度、化学稳定性(水解、氧化、热和光)和药物动力学特性,包括吸收、分布、代谢和体内消除,生物利用度低,渗透性差,半衰期短和向治疗靶位局部递送姜黄素差,从而获得最好的结果,实现如抗氧化、抗炎、抗癌、诱导细胞凋亡、抗血管生成、神经保护、抗微生物剂、保肝护肾、抑制血栓形成、心梗保护、降血糖、抗风湿活动等优异的药理作用,有效治疗哺乳动物疾病。The main object of the present invention is to effectively utilize curcumin by solving various problems such as poor solubility, chemical stability (hydrolysis, oxidation, heat and light) and pharmacokinetic properties including absorption, distribution, metabolism and elimination in vivo, and bioavailability. Low degree, poor permeability, short half-life and local delivery of curcumin to the therapeutic target to achieve the best results, such as anti-oxidation, anti-inflammatory, anti-cancer, induced apoptosis, anti-angiogenesis, neuroprotection, anti- It has excellent pharmacological effects such as microbial agents, liver and kidney protection, inhibition of thrombosis, myocardial infarction, hypoglycemia and anti-rheumatic activities, and is effective in treating mammalian diseases.
具体而言,本发明提供一种药物递送***,其包含活性成分姜黄素或其衍生物或其药学上可接受的盐和聚合物载体Soluplus。In particular, the present invention provides a drug delivery system comprising the active ingredient curcumin or a derivative thereof or a pharmaceutically acceptable salt thereof and a polymer carrier Soluplus.
根据本发明,姜黄素与Soluplus的重量比为1∶0.001-1∶100。 According to the invention, the weight ratio of curcumin to Soluplus is from 1:0.001 to 1:100.
根据本发明,药物递送***还包含其他聚合物载体和/或表面活性剂。According to the invention, the drug delivery system further comprises other polymeric carriers and/or surfactants.
根据本发明,所述其他聚合物载体为水溶性聚合物,选自N-乙烯基内酰胺均聚物、N-乙烯基内酰胺共聚物、纤维素酯、纤维素醚、聚亚烷基氧化物、聚丙烯酸酯、聚甲基丙烯酸酯、丙烯酸的均聚物和共聚物、甲基丙烯酸的均聚物和共聚物、聚丙烯酰胺、聚乙烯醇、乙酸乙烯酯聚合物、乙酸乙烯酯共聚物、羧乙烯基聚合物、寡糖、多糖及其混合物。According to the invention, the other polymeric carrier is a water soluble polymer selected from the group consisting of N-vinyl lactam homopolymers, N-vinyl lactam copolymers, cellulose esters, cellulose ethers, polyalkylene oxides , polyacrylate, polymethacrylate, homopolymer and copolymer of acrylic acid, homopolymer and copolymer of methacrylic acid, polyacrylamide, polyvinyl alcohol, vinyl acetate polymer, vinyl acetate copolymerization , carboxyvinyl polymers, oligosaccharides, polysaccharides, and mixtures thereof.
根据本发明,所述其他聚合物载体选自烷基纤维素、羟烷基纤维素、羟烷基烷基纤维素、甲基纤维素(MC)、乙基纤维素(EC)、羟乙基纤维素(HEC)、羟丙基纤维素(HPC)、羟丙基甲基纤维素(HPMC)、羟乙基甲基纤维素(HEMC)、羟丙基甲基纤维素琥珀酸酯、羟丙基甲基纤维素乙酸琥珀酸酯、羧甲基乙基纤维素、羧甲基纤维素钠、羧甲基纤维素钾、纤维素乙酸琥珀酸酯、纤维素乙酸邻苯二甲酸酯、羟丙基甲基纤维素邻苯二甲酸酯、聚丙烯酸共聚合物、聚(甲基)丙烯酸聚合物、聚(羟烷基丙烯酸酯)、聚(羟烷基甲基丙烯酸酯)、聚乙烯吡咯烷酮(PVP)、乙烯基吡咯烷酮均聚物、乙烯基吡咯烷酮共聚物、聚维酮、乙烯基吡咯烷酮-乙烯基乙酸酯共聚物(共聚维酮)、乙酸乙烯酯的共聚合物、丙酸乙烯酯的共聚物、乙酸乙烯酯和巴豆酸的共聚物、聚乙二醇、聚乙烯醇、部分水解的聚乙酸乙烯酯、明胶、藻酸钠、可溶性淀粉、***胶、糊精、透明质酸、软骨素硫酸钠、藻酸丙二醇酯、琼脂、黄芪胶、黄原胶、氨基烷基甲基丙烯酸酯共聚物、聚乙酸乙烯酯-二乙基氨基乙酸酯、甲基丙烯酸酯共聚物、甲基丙烯酸共聚物L、甲基丙烯酸共聚物LD、甲基丙烯酸共聚物S、聚乙二醇(macrogol)、聚氧乙烯、聚氧丙烯、环氧乙烷(EO)和环氧丙烷(PO)的共聚物、卡拉胶、半乳甘露聚糖及其组合物。According to the invention, the other polymeric carrier is selected from the group consisting of alkyl cellulose, hydroxyalkyl cellulose, hydroxyalkyl alkyl cellulose, methyl cellulose (MC), ethyl cellulose (EC), hydroxyethyl Cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methylcellulose (HPMC), hydroxyethyl methylcellulose (HEMC), hydroxypropylmethylcellulose succinate, hydroxypropyl Methyl cellulose acetate succinate, carboxymethyl ethyl cellulose, sodium carboxymethyl cellulose, potassium carboxymethyl cellulose, cellulose acetate succinate, cellulose acetate phthalate, hydroxyl Propyl methylcellulose phthalate, polyacrylic acid copolymer, poly(meth)acrylic acid polymer, poly(hydroxyalkyl acrylate), poly(hydroxyalkyl methacrylate), polyethylene Pyrrolidone (PVP), vinylpyrrolidone homopolymer, vinylpyrrolidone copolymer, povidone, vinylpyrrolidone-vinyl acetate copolymer (copovidone), copolymer of vinyl acetate, vinyl propionate Copolymer of ester, copolymer of vinyl acetate and crotonic acid, polyethylene glycol, polyvinyl alcohol, partially hydrolyzed poly Acid vinyl ester, gelatin, sodium alginate, soluble starch, gum arabic, dextrin, hyaluronic acid, sodium chondroitin sulfate, propylene glycol alginate, agar, tragacanth, xanthan gum, aminoalkyl methacrylate copolymerization , polyvinyl acetate-diethylaminoacetate, methacrylate copolymer, methacrylic acid copolymer L, methacrylic acid copolymer LD, methacrylic acid copolymer S, polyethylene glycol (macrogol) Polyoxyethylene, polyoxypropylene, copolymers of ethylene oxide (EO) and propylene oxide (PO), carrageenan, galactomannan, and combinations thereof.
根据本发明,所述其他聚合物载体选自羟丙基甲基纤维素 (HPMC)、聚乙二醇(PEG)、壳聚糖、PVP、PVP/VA、HPC、羟丙基甲基纤维素乙酸酯(HPMCAS)、eudragit E100、基于甲基丙烯酸二甲基氨基乙酯、甲基丙烯酸丁酯和甲基丙烯酸甲酯的阳离子共聚物。According to the invention, the other polymeric carrier is selected from the group consisting of hydroxypropyl methylcellulose (HPMC), polyethylene glycol (PEG), chitosan, PVP, PVP/VA, HPC, hydroxypropyl methylcellulose acetate (HPMCAS), eudragit E100, based on dimethylaminoethyl methacrylate a cationic copolymer of an ester, butyl methacrylate and methyl methacrylate.
根据本发明,所述表面活性剂包括阴性、阳性或两性表面活性剂,并且选自十二烷基磺酸钠、十二烷基硫酸钠(SDS),月桂基硫酸钠(SLS)、聚氧乙烯山梨醇酐长链脂肪酸酯、维生素E-TPGS、胆盐、脱氧胆酸钠、甘胆酸钠、聚氧乙烯聚氧丙烯二醇及其组合。According to the invention, the surfactant comprises a negative, positive or amphoteric surfactant and is selected from the group consisting of sodium dodecyl sulfate, sodium dodecyl sulfate (SDS), sodium lauryl sulfate (SLS), polyoxygen Ethylene sorbate long-chain fatty acid ester, vitamin E-TPGS, bile salt, sodium deoxycholate, sodium glycocholate, polyoxyethylene polyoxypropylene glycol, and combinations thereof.
根据本发明,药物递送***选自基于纳米技术的药物递送***、固溶体***以及乳液***,以针对可能的沉淀提供额外的稳定性。In accordance with the present invention, the drug delivery system is selected from the group consisting of nanotechnology-based drug delivery systems, solid solution systems, and emulsion systems to provide additional stability for possible precipitation.
根据本发明,所述基于纳米技术的药物递送***选自胶束、纳米颗粒、纳米纤维和纳米悬浮液,而所述基于固溶体***选自固体分散体、挤出物和固体载体***。According to the present invention, the nanotechnology-based drug delivery system is selected from the group consisting of micelles, nanoparticles, nanofibers, and nanosuspensions, and the solid solution based system is selected from the group consisting of solid dispersions, extrudates, and solid support systems.
根据本发明,药物递送***为固体分散体制剂,优选还含有TPGS 1000。According to the invention, the drug delivery system is a solid dispersion formulation, preferably further comprising TPGS 1000.
根据本发明,药物递送***为胶束制剂,优选还含有TPGS 1000。According to the invention, the drug delivery system is a micelle formulation, preferably further comprising TPGS 1000.
根据本发明,所述胶束制剂进一步含有其他聚合物载体与水/缓冲剂,以及其中有效量的姜黄素包合在胶束中。According to the invention, the micelle formulation further comprises other polymeric carriers and water/buffering agents, and wherein an effective amount of curcumin is encapsulated in the micelles.
根据本发明,所述胶束进一步包含表面活性剂、固相吸附剂、酸化剂和/或抗氧化剂。According to the invention, the micelles further comprise a surfactant, a solid phase adsorbent, an acidulant and/or an antioxidant.
本发明还提供一种固体分散体制剂的制备方法,包括下列步骤:The invention also provides a preparation method of a solid dispersion preparation, comprising the following steps:
将活性成分姜黄素或其衍生物或其可药用盐分散在聚合物载体和任选的表面活性剂中。 The active ingredient curcumin or a derivative thereof, or a pharmaceutically acceptable salt thereof, is dispersed in a polymeric carrier and optionally a surfactant.
根据本发明,固体分散体制剂的制备方法进一步包括选自以下的步骤:融冰浴搅拌、薄膜冷却、液氮、喷雾凝结、热熔挤出、MeltrexTM、熔融凝聚、或溶剂蒸发(烘干、真空干燥、旋转蒸发、热板加热、喷雾干燥、冷冻干燥、超临界抗溶剂、共沉淀、静电纺丝、喷雾冷干、超快冷干、流体床涂布)和溶剂熔融。According to the present invention, a method of preparing a solid dispersion formulation comprising a further step selected from: melting ice bath with stirring, the film was cooled with liquid nitrogen, spray congealing, hot melt extrusion, Meltrex TM, melt agglomeration, or solvent evaporation (drying , vacuum drying, rotary evaporation, hot plate heating, spray drying, freeze drying, supercritical antisolvent, coprecipitation, electrospinning, spray lyophilization, ultra-fast lyophilization, fluid bed coating) and solvent melting.
本发明还提供一种胶束的制备方法,包括以下步骤:The invention also provides a preparation method of micelles, comprising the following steps:
将活性成分姜黄素或其衍生物或其药学上可接受的盐、聚合物载体和表面活性剂,任选溶解于有机溶剂中,通过旋转蒸发除去有机溶剂,膜形成后,真空干燥,加入缓冲剂水合,超声处理。The active ingredient curcumin or a derivative thereof, or a pharmaceutically acceptable salt thereof, a polymer carrier and a surfactant are optionally dissolved in an organic solvent, and the organic solvent is removed by rotary evaporation. After the film is formed, it is vacuum dried and added to a buffer. The agent is hydrated and sonicated.
本发明还提供一种基于脂质的药物递送***,其包含活性成分姜黄素或其衍生物或其药学上可接受的盐和脂质。The present invention also provides a lipid-based drug delivery system comprising the active ingredient curcumin or a derivative thereof or a pharmaceutically acceptable salt thereof and a lipid.
根据本发明,所述脂质为甘油三酯,包括长链甘油三酯(LCT)、中链甘油三酯(MCT)和短链甘油三酯(SCT),其中长链甘油三酯选自氢化大豆油、氢化植物油、玉米油、橄榄油、豆油、花生油和芝麻油,中链甘油三酯选自来自可可油或棕榈籽油的辛酸/葵酸甘油三酯。According to the invention, the lipid is a triglyceride comprising long chain triglycerides (LCT), medium chain triglycerides (MCT) and short chain triglycerides (SCT), wherein the long chain triglycerides are selected from hydrogenation Soybean oil, hydrogenated vegetable oil, corn oil, olive oil, soybean oil, peanut oil and sesame oil, medium chain triglycerides are selected from caprylic/maronic acid triglycerides from cocoa butter or palm seed oil.
根据本发明,基于脂质的药物递送***还包括赋形剂,选自化学甘油三酯、部分甘油三酯、半合成油性酯和半合成非离子表面活性剂酯。According to the present invention, the lipid-based drug delivery system further comprises an excipient selected from the group consisting of chemical triglycerides, partial triglycerides, semi-synthetic oily esters, and semi-synthetic nonionic surfactant esters.
根据本发明,基于脂质的药物递送***还包括水不溶性赋形剂,选自蜂蜡、油酸、大豆脂肪酸、维生素E、玉米油单-二-甘油三酯、中链(C8/C10)甘油单酯和甘油二酯,以及脂肪酸的丙二醇酯。According to the present invention, the lipid-based drug delivery system further comprises a water-insoluble excipient selected from the group consisting of beeswax, oleic acid, soy fatty acid, vitamin E, corn oil mono-di-triglyceride, medium chain (C8/C10) glycerol Monoesters and diglycerides, as well as propylene glycol esters of fatty acids.
根据本发明,所述脂质选自Caproyl 90,Capmul MCM和CaproylTM PGMC中的一种或多种。 According to the present invention, the lipid is selected from Caproyl 90, Capmul MCM Caproyl TM and one or more of PGMC.
根据本发明,基于脂质的药物递送***还包括水溶性有机溶剂、表面活性剂、辅助表面活性剂、聚合物增溶剂、磷脂和/或添加剂。According to the present invention, the lipid-based drug delivery system further comprises a water-soluble organic solvent, a surfactant, a co-surfactant, a polymer solubilizer, a phospholipid, and/or an additive.
根据本发明,水溶性有机溶剂选自PEG 200-10,000、聚乙烯己内酰胺(PCL)、聚乙酸乙烯酯(PVA)或其共聚物、水溶性形式的维生素E和乙醇;表面活性剂是其中脂肪酸为不饱和或饱和的膳食油的衍生物,通过PEG与水解植物油反应、醇与氧化乙烯反应生成烷基醚乙氧基化物、或者基于聚山梨醇酯的植物油与氧化乙烯反应而合成;辅助表面活性剂基于聚乙二醇、聚丙二醇、乙醇和甘油;聚合物增溶剂选自Soluplus、壳聚糖、聚乙烯吡咯烷酮(PVP)、PVP/VA、HPC、HPMC、HPMCAS、eudragit E100、基于甲基丙烯酸二甲基氨基乙酯、甲基丙烯酸丁酯和甲基丙烯酸甲酯的阳离子共聚物。优选的,PEG 200-10,000选自PEG 300,PEG 400,PEG 1,000和PEG 6,000;表面活性剂选自Cremophor RH 40、Labrasol、TPGS 1000、Tween 20、Cremophor E1和Tween 80;以及辅助表面活性剂选自PEG 300、PEG 400、丙二醇、甘油、乙醇、Transcutol HP和Transcutol P。According to the present invention, the water-soluble organic solvent is selected from the group consisting of PEG 200-10,000, polyvinyl caprolactam (PCL), polyvinyl acetate (PVA) or a copolymer thereof, a water-soluble form of vitamin E and ethanol; and a surfactant in which the fatty acid is a derivative of an unsaturated or saturated dietary oil synthesized by reacting PEG with a hydrolyzed vegetable oil, reacting an alcohol with ethylene oxide to form an alkyl ether ethoxylate, or reacting a polysorbate-based vegetable oil with ethylene oxide; The agent is based on polyethylene glycol, polypropylene glycol, ethanol and glycerin; the polymer solubilizer is selected from the group consisting of Soluplus, chitosan, polyvinylpyrrolidone (PVP), PVP/VA, HPC, HPMC, HPMCAS, eudragit E100, based on methacrylic acid A cationic copolymer of dimethylaminoethyl ester, butyl methacrylate and methyl methacrylate. Preferably, PEG 200-10,000 is selected from the group consisting of PEG 300, PEG 400, PEG 1,000 and PEG 6,000; the surfactant is selected from the group consisting of Cremophor RH 40, Labrasol, TPGS 1000, Tween 20, Cremophor E1 and Tween 80; and co-surfactant selection From PEG 300, PEG 400, propylene glycol, glycerol, ethanol, Transcutol HP and Transcutol P.
根据本发明,所述添加剂包括固相吸附剂、水溶性和脂溶性抗氧化剂、酸化剂、螯合剂、防腐剂、稳定剂和/或缓冲剂,其中固相吸附剂包括硅基吸附剂和非硅基吸附剂,硅基吸附剂选自Aerosil 200和偏硅酸镁铝,非硅基吸附剂选自微晶纤维素、滑石、无水磷酸氢二钙(DCPA)、由烷基纤维素、羟烷基纤维素、羟烷基烷基纤维素糖等基团组成的水溶性聚合物;螯合剂为选自乙二胺、乙二胺四乙酸二钠钙和乙二胺四乙酸二钠的至少一种;酸化剂选自柠檬酸、乙酸、富马酸、盐酸和硝酸;缓冲剂选自偏磷酸钾、磷酸二氢钾、醋酸钠、柠檬酸钠;水溶性或脂溶性抗氧化剂选自抗坏血酸、抗坏血酸棕榈酸酯、丁基羟基茴香醚、丁基羟基甲苯、次磷酸、硫代甘油、没食子酸丙酯、抗坏血酸钠、亚硫酸氢钠、甲醛次硫酸氢钠、次硫酸盐、焦亚硫酸钠。 According to the present invention, the additive comprises a solid phase adsorbent, a water soluble and fat soluble antioxidant, an acidulant, a chelating agent, a preservative, a stabilizer and/or a buffer, wherein the solid phase adsorbent comprises a silicon based adsorbent and a silicon-based adsorbent, the silicon-based adsorbent is selected from the group consisting of Aerosil 200 and magnesium aluminum metasilicate, and the non-silicon-based adsorbent is selected from the group consisting of microcrystalline cellulose, talc, anhydrous dibasic calcium phosphate (DCPA), and alkyl cellulose. a water-soluble polymer composed of a group such as a hydroxyalkyl cellulose or a hydroxyalkylalkyl cellulose sugar; the chelating agent is selected from the group consisting of ethylenediamine, disodium edetate, and disodium edetate. At least one; the acidifying agent is selected from the group consisting of citric acid, acetic acid, fumaric acid, hydrochloric acid and nitric acid; the buffering agent is selected from the group consisting of potassium metaphosphate, potassium dihydrogen phosphate, sodium acetate, sodium citrate; and the water-soluble or fat-soluble antioxidant is selected from the group consisting of Ascorbic acid, ascorbyl palmitate, butylhydroxyanisole, butylhydroxytoluene, hypophosphorous acid, thioglycerol, propyl gallate, sodium ascorbate, sodium hydrogen sulfite, sodium formaldehyde sulfoxylate, sulfoxylate, sodium metabisulfite .
根据本发明,除姜黄素之外,基于脂质的药物递送***还包括CapryolTM PGMC、
Figure PCTCN2017094665-appb-000001
RH 40、Labrasol、TPGS 1000、Transcutol P和/或Aerosil 200。
According to the present invention, in addition to curcumin, lipid-based drug delivery system further comprises Capryol TM PGMC,
Figure PCTCN2017094665-appb-000001
RH 40, Labrasol, TPGS 1000, Transcutol P and/or Aerosil 200.
根据本发明,基于脂质的药物递送***选自脂质溶液、脂质体悬浮液、表面活性剂或聚合物-脂质混合的胶束,自微乳化药物递送***(SMEDDS)和纳米乳液制剂。According to the present invention, the lipid-based drug delivery system is selected from the group consisting of a lipid solution, a liposome suspension, a surfactant or a polymer-lipid mixed micelle, a self-microemulsifying drug delivery system (SMEDDS) and a nanoemulsion formulation. .
根据本发明,基于脂质的药物递送***为液相或固相SMEDDS。如是固相,还包括固相吸附剂,优选为Aerosil 200。According to the invention, the lipid based drug delivery system is a liquid phase or solid phase SMEDDS. In the case of a solid phase, a solid phase adsorbent is also included, preferably Aerosil 200.
根据本发明,基于脂质的药物递送***为纳米乳液制剂,其还包括水和/或缓冲剂。According to the invention, the lipid based drug delivery system is a nanoemulsion formulation which further comprises water and/or a buffer.
本发明还提供基于脂质的药物递送***的制备方法,包括下列步骤:The invention also provides a method of preparing a lipid-based drug delivery system comprising the following steps:
将活性成分姜黄素或其药学上可接受的盐溶解在脂质,表面活性剂,或脂质与表面活性剂的混合物中。The active ingredient curcumin or a pharmaceutically acceptable salt thereof is dissolved in a lipid, a surfactant, or a mixture of a lipid and a surfactant.
根据本发明,药物递送***为固体剂型,选自片剂、环剂、贴剂、胶囊、丸剂、颗粒剂、细粒剂或粉剂、粉末或条带(strip),通过口服、肠胃外、吸入、局部或经皮、鼻、眼内、耳、直肠、***途径给药。According to the invention, the drug delivery system is in the form of a solid dosage form selected from the group consisting of tablets, rings, patches, capsules, pills, granules, fine granules or powders, powders or strips, by oral, parenteral, inhalation. , local or percutaneous, nasal, intraocular, ear, rectal, vaginal route.
根据本发明,药物递送***为液体剂型,选自溶液、悬浮液、乳液、基于共溶剂的***、气溶胶,通过口服、肠胃外、吸入、局部或经皮、鼻内、眼内、耳、直肠、***途径给药。According to the invention, the drug delivery system is in the form of a liquid, selected from the group consisting of solutions, suspensions, emulsions, cosolvent-based systems, aerosols, by oral, parenteral, inhalation, topical or transdermal, intranasal, intraocular, aural, Rectal, vaginal route of administration.
根据本发明,药物递送***为半固体剂型,选自选自药膏、霜剂、凝胶、糊剂,通过外用或经皮、直肠、***途径给药,用于局部或全身目的。 According to the present invention, the drug delivery system is a semi-solid dosage form selected from the group consisting of ointments, creams, gels, pastes, for topical or systemic purposes by topical or transdermal, rectal, vaginal routes.
根据本发明,药物递送***还包括可药用赋形剂,所述赋形剂选自崩解剂、润滑剂、助流剂、抗粘附剂、惰性填料、润湿剂、pH改性剂、粘合剂、溶解度改性剂、重结晶抑制剂、稀释剂及其组合。According to the present invention, the drug delivery system further comprises a pharmaceutically acceptable excipient selected from the group consisting of a disintegrant, a lubricant, a glidant, an anti-adherent, an inert filler, a wetting agent, a pH modifier. , binders, solubility modifiers, recrystallization inhibitors, diluents, and combinations thereof.
根据本发明,在药物递送***的液体制剂中,姜黄素的含量为0.001-1000mg/ml,或者0.1-100mg/ml,或者10-20mg/ml,而在药物递送***的固体制剂中,姜黄素的剂量为0.001-1000mg/单位,或者0.1-100mg/单位,或者10-20mg/单位。According to the present invention, curcumin is contained in a liquid preparation of a drug delivery system in an amount of 0.001 to 1000 mg/ml, or 0.1 to 100 mg/ml, or 10 to 20 mg/ml, and in a solid preparation of a drug delivery system, curcumin The dose is from 0.001 to 1000 mg/unit, or from 0.1 to 100 mg/unit, or from 10 to 20 mg/unit.
本发明的药物递送***可用于治疗多种疾病,并根据疾病类型选择本发明合适的给药途径和剂型。因此,本发明还提供药物递送***在制备用于抗氧化、抗炎、抗癌、诱导细胞凋亡、抗血管生成、神经保护、抗微生物、保肝护肾、抑制血管形成、预防心梗、降血糖、抗风湿的药物中的应用。The drug delivery system of the present invention can be used to treat a variety of diseases and to select a suitable route of administration and dosage form according to the type of disease. Therefore, the present invention also provides a drug delivery system for preparing for anti-oxidation, anti-inflammatory, anti-cancer, inducing apoptosis, anti-angiogenesis, neuroprotection, anti-microbial, liver and kidney protection, inhibiting blood vessel formation, preventing myocardial infarction, Application in blood sugar lowering and anti-rheumatic drugs.
附图简述BRIEF DESCRIPTION OF THE DRAWINGS
为了更清楚地描述本发明的技术方案,下面将结合附图作简要介绍。显而易见,这些附图仅是本申请记载的一些具体实施方式。本发明包括但不限于这些附图。In order to more clearly describe the technical solution of the present invention, a brief description will be made below with reference to the accompanying drawings. It is apparent that these drawings are only some of the specific embodiments described herein. The invention includes, but is not limited to, the drawings.
图1显示实施例1-2的姜黄素在pH 1.2的缓冲液中的溶解度(平均值±S.D.,N=3)。Figure 1 shows the solubility of curcumin of Example 1-2 in a buffer of pH 1.2 (mean ± S. D., N = 3).
图2显示实施例3-10的姜黄素在pH 1.2的缓冲液中的溶解度(平均值±S.D.,N=3)。Figure 2 shows the solubility of curcumin of Examples 3-10 in a buffer of pH 1.2 (mean ± S. D., N = 3).
图3显示姜黄素在A缓冲液和含固体分散体的B缓冲液中的化学稳定性(平均值±S.D.,N=3)。Figure 3 shows the chemical stability of curcumin in A buffer and B buffer containing solid dispersion (mean ± S. D., N = 3).
图4显示姜黄素、物理混合物(PM)和固体分散体(实施例1)的DSC温谱图。Figure 4 shows the DSC thermogram of curcumin, physical mixture (PM) and solid dispersion (Example 1).
图5显示姜黄素、Soluplus、物理混合物(PM)和固体分散体(实施例1)的X射线衍射图。 Figure 5 shows the X-ray diffraction pattern of curcumin, Soluplus, physical mixture (PM) and solid dispersion (Example 1).
图6显示姜黄素、Soluplus和固体分散体(实施例1)的SEM显微照片。Figure 6 shows SEM micrographs of curcumin, Soluplus and solid dispersion (Example 1).
图7显示通过动态光散射法测定姜黄素胶束(实施例3)的尺寸分布。Figure 7 shows the measurement of the size distribution of curcumin micelles (Example 3) by dynamic light scattering.
图8显示NCF制剂(实施例3)与姜黄素比较的物理稳定性(平均值±S.D.,N=3),其中图8A-8D分别显示姜黄素在本发明胶束制剂中的水解稳定性、氧化稳定性、热稳定性和光稳定性。Figure 8 shows the physical stability (mean ± SD, N = 3) of the NCF preparation (Example 3) compared to curcumin, wherein Figures 8A-8D respectively show the hydrolytic stability of curcumin in the micelle preparation of the present invention, Oxidative stability, thermal stability and light stability.
图9显示姜黄素、Soluplus和NCF(实施例1)对SH-SY5Y细胞生存力的影响(平均值±S.D.,N=3)。Figure 9 shows the effect of curcumin, Soluplus and NCF (Example 1) on the viability of SH-SY5Y cells (mean ± S. D., N = 3).
图10显示未加工姜黄素和NCF(实施例1)对通过CuSO4和H2O2诱导的过表达APP的SH-SY5Y-APP695细胞毒性的影响(平均值±S.D.,N=3)。Figure 10 shows the effect of unprocessed curcumin and NCF (Example 1) on the cytotoxicity of SH-SY5Y-APP695 overexpressing APP induced by CuSO 4 and H 2 O 2 (mean ± SD, N = 3).
图11显示姜黄素和NCF(实施例1)对过度表达APP的SH-SY5Y-APP695的定量细胞摄取研究(平均值±S.D.,N=3)。Figure 11 shows quantitative cell uptake studies of curcumin and NCF (Example 1) on SH-SY5Y-APP695 overexpressing APP (mean ± S. D., N = 3).
图12显示以姜黄素和NCF(实施例1)处理的过度表达APP的SH-SY5Y-APP695细胞的荧光显微照片,其中A为天然姜黄素,而B为NCF。Figure 12 shows fluorescent micrographs of SH-SY5Y-APP695 cells overexpressing APP treated with curcumin and NCF (Example 1), where A is native curcumin and B is NCF.
图13显示姜黄素、物理混合物(PM)和固体分散体(SD)(实施例1)的溶出性质(平均值±S.D.,N=3)。Figure 13 shows the dissolution properties of curcumin, physical mixture (PM) and solid dispersion (SD) (Example 1) (mean ± S. D., N = 3).
图14显示纯姜黄素以及优化的NCF(实施例1)的血浆分布(平均值±S.D.,N=3)。Figure 14 shows the plasma distribution of pure curcumin and optimized NCF (Example 1) (mean ± S. D., N = 3).
实施例Example
为了进一步理解本发明,下面将结合实施例对本发明的优选方案进行描述。这些描述只是举例说明本发明姜黄素新型药物制剂的特征和优点,而非限制本发明的保护范围。 In order to further understand the present invention, the preferred embodiments of the present invention will be described below in conjunction with the embodiments. These descriptions are merely illustrative of the features and advantages of the novel curcumin pharmaceutical formulations of the present invention and are not intended to limit the scope of the invention.
表1:本发明所用组分及其化学名称Table 1: Components used in the present invention and their chemical names
Figure PCTCN2017094665-appb-000002
Figure PCTCN2017094665-appb-000002
实施例1:固体分散体制剂Example 1: Solid Dispersion Formulation
成分ingredient 数量(mg)Quantity (mg)
姜黄素 Curcumin 100100
Soluplus Soluplus 10001000
实施例2:固体分散体制剂Example 2: Solid dispersion preparation
成分ingredient 数量(mg)Quantity (mg)
姜黄素 Curcumin 100100
Soluplus:TPGS 1000Soluplus: TPGS 1000 1000:2001000:200
实施例3:胶束制剂Example 3: Micelle preparation
成分ingredient 数量(mg)Quantity (mg)
姜黄素 Curcumin 100100
Soluplus Soluplus 10001000
PBS(7.4pH)PBS (7.4pH) 10mL10mL
实施例4:胶束制剂Example 4: Micelle preparation
成分ingredient 数量(mg)Quantity (mg)
姜黄素 Curcumin 100100
Soluplus:TPGS 1000Soluplus: TPGS 1000 1000:2001000:200
PBS(7.4pH)PBS (7.4pH) 10mL10mL
实施例5:基于脂质的SMEDDS制剂Example 5: Lipid-based SMEDDS formulation
成分ingredient 数量Quantity
姜黄素Curcumin 50mg/mL50mg/mL
Caproyl PGMCCaproyl PGMC 20%20%
Cremophor RH 40Cremophor RH 40 25%25%
Labrasol:TPGS 1000(4:1)Labrasol: TPGS 1000 (4:1) 25%25%
Transcutol PTranscutol P 30%30%
实施例6:基于脂质的SMEDDS制剂Example 6: Lipid-based SMEDDS formulation
成分ingredient 数量Quantity
姜黄素Curcumin 50mg/mL50mg/mL
Caproyl PGMCCaproyl PGMC 20%20%
Cremophor RH 40Cremophor RH 40 25%25%
Labrasol:TPGS 1000(4:1)Labrasol: TPGS 1000 (4:1) 25%25%
Transcutol PTranscutol P 30%30%
Aerosil 200(吸附剂)Aerosil 200 (adsorbent) 5%w/v5% w/v
实施例7:基于脂质的SMEDDS制剂Example 7: Lipid-based SMEDDS formulation
成分ingredient 数量Quantity
姜黄素Curcumin 50mg/mL50mg/mL
Caproyl PGMCCaproyl PGMC 20%20%
Cremophor RH 40Cremophor RH 40 25%25%
LabrasolLabrasol 25%25%
Transcutol PTranscutol P 30%30%
实施例8:基于脂质的SMEDDS制剂Example 8: Lipid-based SMEDDS formulation
成分ingredient 数量Quantity
姜黄素Curcumin 50mg/mL50mg/mL
Caproyl PGMCCaproyl PGMC 20%20%
Cremophor RH 40Cremophor RH 40 25%25%
LabrasolLabrasol 25%25%
Aerosil 200(吸附剂)Aerosil 200 (adsorbent) 5%w/v5% w/v
实施例9:基于脂质的纳米乳液制剂Example 9: Lipid-based nanoemulsion formulation
成分ingredient 数量Quantity
姜黄素Curcumin 50mg/mL50mg/mL
Caproyl PGMCCaproyl PGMC 20%20%
Cremophor RH 40Cremophor RH 40 25%25%
Labrasol:TPGS 1000(4:1)Labrasol: TPGS 1000 (4:1) 25%25%
Transcutol PTranscutol P 30%30%
water 足量Sufficient
实施例10:基于脂质的纳米乳剂制剂Example 10: Lipid-based nanoemulsion formulation
成分ingredient 数量Quantity
姜黄素Curcumin 50mg/mL50mg/mL
Caproyl PGMCCaproyl PGMC 20%20%
Cremophor RH 40Cremophor RH 40 25%25%
LabrasolLabrasol 25%25%
Transcutol PTranscutol P 30%30%
water 足量Sufficient
制备例1:固体分散体制剂(实施例1和2)的制备Preparation Example 1: Preparation of Solid Dispersion Formulations (Examples 1 and 2)
根据实施例1和2,将所需量的姜黄素、Soluplus和任选的TPGS 1000溶解在乙醇中。通过Buchi旋转蒸发仪II去除有机溶剂。形成的膜在真空干燥器中干燥过夜。干燥样品从烧瓶刮下,并收集在研钵中。用研杵压碎粉末,并制成均匀形式。According to Examples 1 and 2, the required amount of curcumin, Soluplus and optionally TPGS 1000 were dissolved in ethanol. The organic solvent was removed by a Buchi rotary evaporator II. The formed film was dried overnight in a vacuum desiccator. The dried sample was scraped from the flask and collected in a mortar. The powder was crushed with a mortar and made into a uniform form.
制备例2:胶束制剂(实施例3和4)的制备Preparation Example 2: Preparation of micelle preparations (Examples 3 and 4)
根据实施例3和4,将所需量的姜黄素、Soluplus和任选的TPGS 1000溶解在乙醇中。通过Buchi旋转蒸发仪II去除有机溶剂。形成的膜在真空干燥器中干燥过夜,然后以10ml 1×PBS缓冲剂(pH 7.4)水合,37℃下温育30分钟,接着超声处理几分钟。所得混合物通过0.45μm针头过滤器(PVDF)过滤。 The required amounts of curcumin, Soluplus and optionally TPGS 1000 were dissolved in ethanol according to Examples 3 and 4. The organic solvent was removed by a Buchi rotary evaporator II. The resulting membrane was dried overnight in a vacuum desiccator, then hydrated with 10 ml of 1 x PBS buffer (pH 7.4), incubated at 37 ° C for 30 minutes, and then sonicated for a few minutes. The resulting mixture was filtered through a 0.45 μm syringe filter (PVDF).
制备例3:液体自微乳化药物递送***(SMEDDS)制剂(实施例5和7)的制备Preparation 3: Preparation of liquid self-microemulsifying drug delivery system (SMEDDS) formulations (Examples 5 and 7)
根据实施例5和7,将所需量的油(Capmul PGMC),表面活性剂(Cremophor RH 40,Labrasol和TPGS 1000)和辅助表面活性剂(Transcutol P)准确称重至小玻璃瓶中。然后,通过轻轻搅拌和旋涡混合,将上述组分混合,并在保温箱中37℃下加热。加入所需量的姜黄素,旋涡混合,直至姜黄素完全溶解。According to Examples 5 and 7, the required amount of oil (Capmul PGMC), surfactant (Cremophor RH 40, Labrasol and TPGS 1000) and co-surfactant (Transcutol P) were accurately weighed into small glass vials. Then, the above components were mixed by gentle stirring and vortex mixing, and heated at 37 ° C in an incubator. Add the required amount of curcumin and vortex to mix until the curcumin is completely dissolved.
制备例4:固体自微乳化药物递送***的制剂(实施例6和8)的制备Preparation Example 4: Preparation of formulations of solid self-microemulsifying drug delivery systems (Examples 6 and 8)
如上制备液体SMEDDS制剂。加入所需量的Aerosil 200之后,以最少量的miliQ水稀释,室温搅拌2小时。所得混合物放置15分钟,平衡后通过0.45μm针头过滤器(PVDF)过滤。冷冻干燥之前,将溶液在-80℃下冷冻至少6小时,然后置于Novalyphe-NL 500(Savant Instruments Corp.,Holbrook,NY)中-45℃和7102mbar压力下冻干至少24小时。最后,将固相SMEDDS存储在干燥器中。A liquid SMEDDS formulation was prepared as above. After the required amount of Aerosil 200 was added, it was diluted with a minimum amount of miliQ water and stirred at room temperature for 2 hours. The resulting mixture was allowed to stand for 15 minutes, equilibrated and filtered through a 0.45 μm syringe filter (PVDF). Prior to lyophilization, the solution was frozen at -80 °C for at least 6 hours and then lyophilized for at least 24 hours at -45 ° C and 7102 mbar pressure in Novalyphe-NL 500 (Savant Instruments Corp., Holbrook, NY). Finally, the solid phase SMEDDS is stored in a desiccator.
制备例5:基于纳米乳液的***(实施例9和10)的制备Preparation Example 5: Preparation of a Nanoemulsion-Based System (Examples 9 and 10)
根据实施例9和10,将所需量的油(Capmul PGMC),表面活性剂(Cremophor RH 40,Labrasol和TPGS 1000)和辅助表面活性剂(Transcutol P)准确称重至小玻璃瓶中。然后,通过轻轻搅拌和旋涡混合,将上述组分混合,并在保温箱中37℃下加热。加入所需量的姜黄素,旋涡混合,直至姜黄素完全溶解。逐滴加入所需量的miliQ水,直至获得清亮透明制剂。According to Examples 9 and 10, the required amount of oil (Capmul PGMC), surfactant (Cremophor RH 40, Labrasol and TPGS 1000) and co-surfactant (Transcutol P) were accurately weighed into small glass vials. Then, the above components were mixed by gentle stirring and vortex mixing, and heated at 37 ° C in an incubator. Add the required amount of curcumin and vortex to mix until the curcumin is completely dissolved. The required amount of miliQ water was added dropwise until a clear, clear formulation was obtained.
实施例1-10提供了多种不同的姜黄素制剂配方,分别包括基于固体分散体、胶束、SMEDDS和纳米乳液制剂。下文通过效果例详细说明这些制剂的优点。 Examples 1-10 provide a variety of different curcumin formulation formulations, including solid dispersions, micelles, SMEDDS, and nanoemulsion formulations, respectively. The advantages of these formulations are detailed below by way of effect examples.
效果例1Effect example 1
姜黄素在不同类型的制剂中的溶解度Solubility of curcumin in different types of preparations
由于姜黄素在低pH时更稳定,选择缓冲液(pH 1.2)用于溶解度研究。Since curcumin is more stable at low pH, a buffer (pH 1.2) was chosen for solubility studies.
实施例1-2固体分散体制剂的溶解度研究Example 1-2 Solubility Study of Solid Dispersion Formulation
在分开的玻璃瓶中,各加入1ml miliQ水。向上述溶液分别加入过量的姜黄素和固相分散体。接着,在测试全程,利用机械振动器(Axyos Technologies,Brisbane,Australia)室温下连续旋转24小时。达到平衡后,各小瓶以3000rpm离心5分钟,通过0.45μm PVDF针头过滤器过滤,丢弃过量的不溶性姜黄素。随后,滤液以甲醇稀释。采用早先开发和验证的HPLC方法,进行三重溶解度分析。In a separate glass bottle, 1 ml of miliQ water was added. An excess of curcumin and a solid phase dispersion were separately added to the above solution. Then, throughout the test, a mechanical vibrator (Axyos Technologies, Brisbane, Australia) was continuously rotated at room temperature for 24 hours. After equilibration, each vial was centrifuged at 3000 rpm for 5 minutes, filtered through a 0.45 μm PVDF syringe filter, and excess insoluble curcumin was discarded. Subsequently, the filtrate was diluted with methanol. Triple solubility analysis was performed using an HPLC method previously developed and validated.
样品分析在HPLC(Shimadzu,Kyoto,Japan)***上操作,该***装配有UV-VIS检测器[SPD-20A],DGU-20A3在线脱气器,CBM-20A***控制器,SIL-20AHT自动加样器,和LC Chromopac数据处理器解决方案。采用Zorbax Eclipse XDB-C18(4.6*150*3.5mm3)分析柱。样品分析的移动相由乙腈和1%(w/v)柠檬酸缓冲液组成,比例为70∶30(v/v)。注射体积20μl,流速1ml/min,检测波长423nm。Sample analysis was performed on a HPLC (Shimadzu, Kyoto, Japan) system equipped with a UV-VIS detector [SPD-20A], a DGU-20A3 online degasser, a CBM-20A system controller, and a SIL-20AHT automatic addition. Sampler, and LC Chromopac data processor solution. The column was analyzed using a Zorbax Eclipse XDB-C18 (4.6*150*3.5 mm 3 ). The mobile phase of the sample analysis consisted of acetonitrile and 1% (w/v) citrate buffer at a ratio of 70:30 (v/v). The injection volume was 20 μl, the flow rate was 1 ml/min, and the detection wavelength was 423 nm.
结果发现(参见图1),与晶体姜黄素和无定形姜黄素相比,姜黄素在固体分散体制剂(实施例1和实施例2)中的溶解度显著改善。As a result, it was found (see Fig. 1) that the solubility of curcumin in the solid dispersion preparation (Example 1 and Example 2) was remarkably improved as compared with the crystalline curcumin and amorphous curcumin.
实施例3-4胶束制剂的溶解度研究Study on the solubility of the micelle preparation of Example 3-4
将过量的姜黄素和所需量的Soluplus和任选的TPGS 1000溶解在乙醇中。通过Buchi旋转蒸发仪II去除有机溶剂。形成的膜在真空干燥器中干燥过夜,然后以10ml 1×PBS缓冲剂(pH 7.4)水合,37℃下温育30分钟,接着超声处理几分钟。各样品以3000rpm离心5分钟。所得混合物通过0.45μm针头过滤器(PVDF)过滤。采用早先开发和验证的HPLC方法,进行三重溶解度分析。 Excess curcumin and the required amount of Soluplus and optionally TPGS 1000 were dissolved in ethanol. The organic solvent was removed by a Buchi rotary evaporator II. The resulting membrane was dried overnight in a vacuum desiccator, then hydrated with 10 ml of 1 x PBS buffer (pH 7.4), incubated at 37 ° C for 30 minutes, and then sonicated for a few minutes. Each sample was centrifuged at 3000 rpm for 5 minutes. The resulting mixture was filtered through a 0.45 μm syringe filter (PVDF). Triple solubility analysis was performed using an HPLC method previously developed and validated.
实施例5-8液体或固体SMEDDS制剂和实施例9-10纳米乳液制剂的溶解度研究Study on Solubility of Examples 5-8 Liquid or Solid SMEDDS Formulations and Examples 9-10 Nanoemulsion Formulations
在分开的玻璃瓶中,各加入1ml所述制剂。向上述溶液加入过量的姜黄素,接着在测试全程,利用机械振动器(Axyos Technologies,Brisbane,Australia)室温下连续旋转24小时。达到平衡后,各小瓶以3000rpm离心5分钟,通过0.45μm PVDF针头过滤器过滤,丢弃过量的不溶性姜黄素。随后,滤液以甲醇稀释。采用早先开发和验证的HPLC方法,进行三重溶解度分析。In a separate glass bottle, 1 ml of the formulation was added each. Excess curcumin was added to the above solution, followed by continuous rotation at room temperature for 24 hours using a mechanical shaker (Axyos Technologies, Brisbane, Australia) throughout the test. After equilibration, each vial was centrifuged at 3000 rpm for 5 minutes, filtered through a 0.45 μm PVDF syringe filter, and excess insoluble curcumin was discarded. Subsequently, the filtrate was diluted with methanol. Triple solubility analysis was performed using an HPLC method previously developed and validated.
结果发现(参见图2),与晶体姜黄素和无定形姜黄素相比,姜黄素在所有新型制剂(实施例3-10)中的溶解度显著改善。As a result, it was found (see Fig. 2) that the solubility of curcumin in all of the novel preparations (Examples 3-10) was significantly improved as compared with the crystalline curcumin and amorphous curcumin.
效果例2Effect example 2
姜黄素及其在固体分散体中的稳定性Curcumin and its stability in solid dispersion
依据USP方法制备模拟胃肠液(无酶和胆汁组分)。为了确定姜黄素的化学稳定性,制备和使用姜黄素(100μg/mL)和固体分散体(100μg/mL的等量姜黄素)的溶液。预定时间间隔采集样品,并且通过0.45μm PVDF针头过滤器过滤。所有样品通过HPLC方法进行三重分析。Simulated gastrointestinal fluid (no enzyme and bile components) was prepared according to the USP method. To determine the chemical stability of curcumin, a solution of curcumin (100 μg/mL) and a solid dispersion (100 μg/mL equivalent of curcumin) was prepared and used. Samples were taken at predetermined time intervals and filtered through a 0.45 μm PVDF syringe filter. All samples were subjected to triple analysis by HPLC method.
如图3A所示,姜黄素在中性至碱性pH条件下显著降解,而在酸性pH下基本保持稳定。与pH 6.8和7.4相比,姜黄素在低pH 1.2时更稳定。姜黄素以二酮和酮-烯醇之间的平衡形式存在,易形成分子内H-键。姜黄素水解开始于亲核OH-离子对酮-烯醇部分中的羰基的攻击。因此,高pH下观察到降解速率越大。与之相反,本发明的固体分散体(实施例1)在不同pH的生物介质中可保护姜黄素免受降解。如图3B所示,姜黄素在pH 1.2、6.8和7.4含固体分散体的缓冲液中相对稳定。该结果确认,聚合物胶束包封姜黄素预防水解,原因是姜黄素的酮-烯醇部分免遭亲核OH-离子的攻击。姜黄素的化学稳定性研究推断,姜黄素的降解是复杂机制,牵涉多种潜在因素。 As shown in Figure 3A, curcumin significantly degrades under neutral to alkaline pH conditions and remains substantially stable at acidic pH. Curcumin is more stable at low pH 1.2 compared to pH 6.8 and 7.4. Curcumin exists in an equilibrium form between diketone and keto-enol, and easily forms intramolecular H-bonds. Curcumin hydrolysis begins with the attack of a nucleophilic OH - ion on the carbonyl group in the keto-enol moiety. Therefore, the greater the rate of degradation observed at high pH. In contrast, the solid dispersion of the invention (Example 1) protects curcumin from degradation in biological media of different pH. As shown in Figure 3B, curcumin was relatively stable in buffers containing solid dispersions at pH 1.2, 6.8 and 7.4. This was confirmed, encapsulated polymer micelle hydrolysis prevention curcumin, curcumin because keto - enol portion from nucleophilic OH - ions attack. Studies on the chemical stability of curcumin have inferred that the degradation of curcumin is a complex mechanism involving a variety of potential factors.
图3显示姜黄素在A缓冲液和B缓冲液中的化学稳定性(平均值±S.D.,N=3),其中A为不含聚合物的空白缓冲液,而B为含有固体分散体制剂的缓冲液。Figure 3 shows the chemical stability of curcumin in A buffer and B buffer (mean ± SD, N = 3), where A is a polymer-free blank buffer and B is a solid dispersion-containing formulation. Buffer.
表2显示,本发明固体分散体和天然姜黄素在不同pH值下,根据二级动力学,不同样品中姜黄素的总降解率。Table 2 shows the total degradation rate of curcumin in different samples according to the second-order kinetics of the solid dispersion of the present invention and natural curcumin at different pH values.
表2:速率常数和半衰期(平均值±S.D.,N=3)Table 2: Rate constants and half-lives (mean ± S. D., N = 3)
Figure PCTCN2017094665-appb-000003
Figure PCTCN2017094665-appb-000003
效果例3Effect example 3
固体分散体的性质研究Study on the properties of solid dispersion
(1)获取姜黄素、物理混合物和固体分散体的DSC温谱图(1) Obtaining DSC thermograms of curcumin, physical mixture and solid dispersion
利用TA Instruments Discovery DSC(型号2920)进行差示扫描量热法(DSC)测量。实验运行之前,对仪器的基线和细胞常数校准。姜黄素、Soluplus和固体分散体(实施例1)等样品封闭在密闭铝盘中,用于DSC实验,空盘用作参照。实验的温度范围从室温至250℃,加热速率10℃/分,受控的氮气流速50ml/分。Differential Scanning Calorimetry (DSC) measurements were performed using a TA Instruments Discovery DSC (Model 2920). The baseline and cell constants of the instrument were calibrated prior to the experiment run. Samples of curcumin, Soluplus, and solid dispersion (Example 1) were enclosed in a closed aluminum pan for DSC experiments and empty plates were used as a reference. The experimental temperature ranged from room temperature to 250 ° C, a heating rate of 10 ° C / min, and a controlled nitrogen flow rate of 50 ml / min.
物理混合物(PM)的组成与固体分散体(SD)相同,只是将药物(姜黄素)和聚合物(Soluplus)在陶瓷研钵中简单研杵而制备。该混合物然后筛分(250μm),并存储在琥珀玻璃盖的容器中。 The composition of the physical mixture (PM) was the same as that of the solid dispersion (SD) except that the drug (curcumin) and the polymer (Soluplus) were simply prepared in a ceramic mortar. The mixture was then sieved (250 μm) and stored in a container of amber glass lid.
参见图4,通过DSC确定了固体分散体制剂中固态的变化。在姜黄素的温谱图中,观察到183℃的尖熔融峰。在物理混合物PM样品中,与晶体姜黄素相比,观察到强度较小的峰,提示晶体有部分转化为无定形态。在固体分散体的情形下,没有观察到与姜黄素的熔融相对应的吸热现象,表明姜黄素在固体分散体中是无定型化的。Referring to Figure 4, the change in solid state in the solid dispersion formulation was determined by DSC. In the thermogram of curcumin, a sharp melting peak at 183 ° C was observed. In the PM sample of the physical mixture, a less intense peak was observed compared to the crystalline curcumin, suggesting that the crystal was partially converted to an amorphous state. In the case of the solid dispersion, no endothermic phenomenon corresponding to the melting of curcumin was observed, indicating that curcumin was amorphous in the solid dispersion.
(2)获取姜黄素、Soluplus和固体分散体(实施例1)样品的X-射线衍射图(2) Obtaining X-ray diffraction patterns of samples of curcumin, Soluplus, and solid dispersion (Example 1)
利用PANalytical(帕那科)、Empyrean(锐影)X射线衍射仪上的CuKα辐射
Figure PCTCN2017094665-appb-000004
操作参数:40kV和40mA,2-50°2θ,步长0.013°,固定的0.25°发散狭缝和0.50°防散射狭缝。
Utilizing CuKα radiation on PANalytical and Empyrean X-ray diffractometers
Figure PCTCN2017094665-appb-000004
Operating parameters: 40kV and 40mA, 2-50°2θ, step size 0.013°, fixed 0.25° divergence slit and 0.50° anti-scatter slit.
参见图5,X-射线衍射(XRD)确认了样品的固态特征。如预期一样,姜黄素的晶体形式显示出尖的衍射特征峰,证实初始形式的结晶性。在物理混合物PM样品中,与晶体姜黄素相比,观察到强度较小的峰,提示晶体有部分转化为无定形态。在Soluplus中,没有观察到任何峰,表明无定形性质。固体分散体的XRD图没有显示结晶性,印证DSC结果,其中没有记录与姜黄素的熔融相对应的吸热现象。Referring to Figure 5, X-ray diffraction (XRD) confirmed the solid state characteristics of the sample. As expected, the crystalline form of curcumin showed a sharp diffractive characteristic peak confirming the crystal form of the initial form. In the PM sample of the physical mixture, a less intense peak was observed compared to the crystalline curcumin, suggesting that the crystal was partially converted to an amorphous state. In Soluplus, no peaks were observed, indicating amorphous nature. The XRD pattern of the solid dispersion did not show crystallinity, confirming the DSC results, in which no endothermic phenomenon corresponding to the melting of curcumin was recorded.
(3)获取姜黄素、Soluplus和固体分散体(实施例1)的SEM显微照片(3) SEM micrographs of curcumin, Soluplus, and solid dispersion (Example 1)
超高分辨率二次电子显微镜(Zeiss Microscopy Merlin with GEMINI II column)配备有场发射枪,0.7kV操作,获取二次电子成像。姜黄素、Soluplus和固体分散体(实施例1)样品以导电双面胶安装在SEM标本台(stub)上。The Zeiss Microscopy Merlin with GEMINI II column is equipped with a field emission gun and operates at 0.7kV for secondary electron imaging. Samples of curcumin, Soluplus, and solid dispersion (Example 1) were mounted on SEM scaffolds with conductive double-sided tape.
参见图6,SEM分析研究形态特征。姜黄素显示针形晶体。Soluplus聚合物显示大而不规则形状。而在固体分散体粉末的成像中,表面特征大为不同。固体分散体的SEM结果显示,与Soluplus聚合物相比, 表面积显著增加,这有助于固体分散体在水性介质中的快速溶出。固体分散体中没有观察到痕量的晶体姜黄素。Referring to Figure 6, SEM analysis studies morphological features. Curcumin shows needle crystals. Soluplus polymers show large irregular shapes. In the imaging of solid dispersion powders, the surface characteristics are quite different. The SEM results of the solid dispersion show that compared to the Soluplus polymer, The surface area is significantly increased, which contributes to the rapid dissolution of the solid dispersion in an aqueous medium. No traces of crystalline curcumin were observed in the solid dispersion.
效果例4Effect example 4
胶束制剂的大小分布Size distribution of micelle preparations
利用Malvern Zeta Sizer Nano ZS测量制备的新型姜黄素制剂(NCF)(实施例3)的粒径、多分散性指数(PDI)和Zeta电位。为了测试,制备1mg/ml溶液,接着以MiliQ水25℃下稀释(100μl,直至1ml)。然后,通过上述方案,三重测量粒径、PDI和Zeta电位。负载能力定义为姜黄素与Soluplus之重量比,而负载效率定义为负载的姜黄素与初始量的姜黄素之比。稳定性评估是制备后不同时间点检查浊度、透明度和沉淀。姜黄素通过HPLC三重分析定量测定。所有测试都是重复三次,结果以平均数和标准偏差表示。The particle size, polydispersity index (PDI) and zeta potential of the novel curcumin preparation (NCF) (Example 3) prepared were measured using a Malvern Zeta Sizer Nano ZS. For the test, a 1 mg/ml solution was prepared, followed by dilution with MiliQ water at 25 ° C (100 μl, up to 1 ml). Then, the particle size, PDI, and Zeta potential were triple measured by the above scheme. The load capacity is defined as the weight ratio of curcumin to Soluplus, and the loading efficiency is defined as the ratio of the loaded curcumin to the initial amount of curcumin. Stability assessment was performed to check for turbidity, clarity, and precipitation at various time points after preparation. Curcumin was quantitatively determined by HPLC triple analysis. All tests were repeated three times and the results were expressed as mean and standard deviation.
观察到粒径为63nm,显示用作纳米技术***的潜力,PDI为0.09,显示不同粒子的大小变动较小,而Zeta电位为-8.65,显示其长时间维持稳定的潜力。NCF显示出高的负载能力(9.15%)和负载效率(98.23%)。本申请通过浊度、透明度和沉淀评估,还确认了稳定性。A particle size of 63 nm was observed, indicating potential for use as a nanotechnology system with a PDI of 0.09, indicating a small variation in the size of the different particles, and a zeta potential of -8.65, indicating its potential to remain stable over time. NCF showed high load capacity (9.15%) and load efficiency (98.23%). The present application also confirmed stability by turbidity, transparency, and precipitation evaluation.
参见图7和表3,通过动态光散射法测定本发明胶束制剂(实施例3)的尺寸分布。Referring to Figure 7 and Table 3, the size distribution of the micelle preparation of the present invention (Example 3) was determined by dynamic light scattering.
表3:姜黄素胶束制剂(实施例3)的物化参数Table 3: Physicochemical parameters of curcumin micelle preparation (Example 3)
Figure PCTCN2017094665-appb-000005
Figure PCTCN2017094665-appb-000005
*稳定性通过浊度、透明度和沉淀校验确定 * Stability is determined by turbidity, transparency and precipitation check
效果例5Effect example 5
稳定性研究,包括水解稳定性、氧化稳定性、热稳定性和光稳定性Stability studies including hydrolytic stability, oxidative stability, thermal stability and photostability
(1)从组成为硼酸、柠檬酸和磷酸(各0.04M)的溶液制备通用缓冲液。加入0.2M氢氧化钠调节终溶液的pH。新型姜黄素制剂(NCF)(实施例3)在不同缓冲溶液(pH 1.8-8)中的浓度为100μg/mL。黑暗中室温下温育溶液,以避免光解。预定时间间隔采集样品,并且通过0.45μm PVDF针头过滤器过滤。所有样品通过HPLC方法进行三重分析。(1) A universal buffer was prepared from a solution having a composition of boric acid, citric acid, and phosphoric acid (0.04 M each). The pH of the final solution was adjusted by the addition of 0.2 M sodium hydroxide. The concentration of the novel curcumin preparation (NCF) (Example 3) in different buffer solutions (pH 1.8-8) was 100 μg/mL. The solution was incubated at room temperature in the dark to avoid photolysis. Samples were taken at predetermined time intervals and filtered through a 0.45 μm PVDF syringe filter. All samples were subjected to triple analysis by HPLC method.
参见图8A,姜黄素在胶束制剂(实施例3)中的稳定性不受从酸性到碱性所有pH范围的影响。该结果确认了通过聚合物胶束包封姜黄素能防止水解。Referring to Figure 8A, the stability of curcumin in the micelle formulation (Example 3) was not affected by all pH ranges from acidic to basic. This result confirmed that encapsulation of curcumin by polymer micelles prevented hydrolysis.
(2)制备浓度为100μg/mL的新型姜黄素制剂(NCF)(实施例3)的0.02%H2O2和3%H2O2的溶液。黑暗中室温下温育溶液,以避免光解。预定时间间隔采集样品,并且通过0.45μm PVDF针头过滤器过滤。所有样品通过HPLC方法进行三重分析。(2) A solution of 0.02% H 2 O 2 and 3% H 2 O 2 of a novel curcumin preparation (NCF) (Example 3) at a concentration of 100 μg/mL was prepared. The solution was incubated at room temperature in the dark to avoid photolysis. Samples were taken at predetermined time intervals and filtered through a 0.45 μm PVDF syringe filter. All samples were subjected to triple analysis by HPLC method.
参见图8B,姜黄素在胶束制剂(实施例3)中的稳定性不受过氧化氢溶液的影响。该结果确认了通过聚合物胶束包封姜黄素能防止氧化性降解。Referring to Figure 8B, the stability of curcumin in the micelle formulation (Example 3) was not affected by the hydrogen peroxide solution. This result confirmed that encapsulation of curcumin by polymer micelles can prevent oxidative degradation.
(3)制备浓度为100μg/mL的新型姜黄素制剂(NCF)(实施例3)的溶液。4℃、25℃和40℃稳定室中温育溶液。预定时间间隔采集样品,并且通过0.45μm PVDF针头过滤器过滤。所有样品通过HPLC方法进行三重分析。(3) A solution of a novel curcumin preparation (NCF) (Example 3) at a concentration of 100 μg/mL was prepared. The solution was incubated in a stable chamber at 4 ° C, 25 ° C and 40 ° C. Samples were taken at predetermined time intervals and filtered through a 0.45 μm PVDF syringe filter. All samples were subjected to triple analysis by HPLC method.
参见图8C,姜黄素在胶束制剂(实施例3)中的稳定性,在不同热应激条件下未受影响。该结果确认了通过聚合物胶束包封姜黄素能 防止热降解。Referring to Figure 8C, the stability of curcumin in the micelle formulation (Example 3) was unaffected under different heat stress conditions. This result confirms that the curcumin can be encapsulated by polymer micelles. Prevent thermal degradation.
(4)制备浓度为100μg/mL的新型姜黄素制剂(NCF)(实施例3)的溶液。根据国际协调会议(ICH)指南,在光稳定室中温育溶液。预定时间间隔采集样品,并且通过0.45μm PVDF针头过滤器过滤。所有样品通过HPLC方法进行三重分析。(4) A solution of a novel curcumin preparation (NCF) (Example 3) at a concentration of 100 μg/mL was prepared. The solution was incubated in a light stable chamber according to the International Conference on Harmonization (ICH) guidelines. Samples were taken at predetermined time intervals and filtered through a 0.45 μm PVDF syringe filter. All samples were subjected to triple analysis by HPLC method.
参见图8D,姜黄素在胶束制剂(实施例3)中的稳定性,在光应激条件下未受影响。该结果确认了通过聚合物胶束包封姜黄素能防止光解。Referring to Figure 8D, the stability of curcumin in the micelle formulation (Example 3) was unaffected under light stress conditions. This result confirmed that encapsulation of curcumin by polymer micelles can prevent photolysis.
效果例6Effect example 6
体外安全性研究In vitro safety study
(1)采用SH-5Y5Y细胞系对本发明制剂的胞毒研究(1) Study on cytotoxicity of the preparation of the invention by using SH-5Y5Y cell line
细胞培养物分析Cell culture analysis
采用SH-SY5Y细胞系操作细胞培养物。DMEM培养基(Dulbecco’s Modified Eagle Medium):营养混合物F12以1∶1之比在25ml细胞培养烧瓶中用于培养细胞,该营养混合物补充有10%胎牛血清(FBS)和1%青霉素-链霉素溶液。细胞在培养箱中在5%CO2条件下37℃进行培养。Cell cultures were manipulated using the SH-SY5Y cell line. DMEM medium (Dulbecco's Modified Eagle Medium): The nutrient mixture F12 was used to culture cells in a 1:1 ratio in a 25 ml cell culture flask supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptavidin. Prime solution. The cells were cultured in an incubator at 37 ° C under 5% CO 2 .
MTT分析SH-SY5Y的细胞存活性MTT analysis of cell viability of SH-SY5Y
在96孔板中,以5×103细胞/孔的密度接种SH-5YSY细胞。24小时后,用含有10μg/mL等量的未加工姜黄素或新型姜黄素制剂(NCF)(实施例1)和Soluplus的培养基替换原培养基。制剂的制备采用无菌水。细胞存活性通过MTT([3-(4,5-二甲基噻唑-2-基)-2,5-二苯基四氮唑溴盐],噻唑蓝)方法测量。20小时后,每孔加入20μL MTT(Sigma-Aldrich,USA,5mg/ml的PBS),温育1小时。加入150μL DMSO溶解不溶性紫色formazan产物,以生成有色溶液。在多孔扫描分光光度计(BIO-RAD型号2550EIA读出仪)上以600nm波长对光 密度(OD)读数。SH-5YSY cells were seeded at a density of 5 x 10 3 cells/well in 96-well plates. After 24 hours, the original medium was replaced with a medium containing 10 μg/mL equivalent of unprocessed curcumin or a novel curcumin preparation (NCF) (Example 1) and Soluplus. The preparation was prepared using sterile water. Cell viability was measured by MTT ([3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide], thiazolyl blue) method. After 20 hours, 20 μL of MTT (Sigma-Aldrich, USA, 5 mg/ml in PBS) was added to each well and incubated for 1 hour. 150 μL of DMSO was added to dissolve the insoluble purple formazan product to produce a colored solution. The optical density (OD) was read at a wavelength of 600 nm on a multi-hole scanning spectrophotometer (BIO-RAD model 2550 EIA reader).
参见图9,与空白Soluplus和姜黄素相比,对新型姜黄素制剂进行安全性研究,以评价初步安全性特性。对细胞未观察到任何毒性差异,确认是非胞毒的。Referring to Figure 9, a safety study of the novel curcumin formulation was performed to evaluate the initial safety profile compared to the blank Soluplus and curcumin. No differences in toxicity were observed for the cells, which were confirmed to be non-cytotoxic.
(2)采用SH-5Y5Y-APP695细胞系对本发明制剂的胞毒研究(2) Study on cytotoxicity of the preparation of the invention by using SH-5Y5Y-APP695 cell line
细胞培养物分析Cell culture analysis
采用SH-SY5Y-APP695细胞系操作细胞培养物。DMEM培养基(Dulbecco’s Modified Eagle Medium):营养混合物F12以1∶1之比在25ml细胞培养烧瓶中用于培养细胞,该营养混合物补充有10%胎牛血清(FBS)和1%青霉素-链霉素溶液。细胞在培养箱中在5%CO2条件下37℃进行培养。Cell cultures were manipulated using the SH-SY5Y-APP695 cell line. DMEM medium (Dulbecco's Modified Eagle Medium): The nutrient mixture F12 was used to culture cells in a 1:1 ratio in a 25 ml cell culture flask supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptavidin. Prime solution. The cells were cultured in an incubator at 37 ° C under 5% CO 2 .
MTT分析SH-SY5Y-APP695的细胞存活性MTT analysis of cell viability of SH-SY5Y-APP695
在96孔板中,以5×103细胞/孔的密度接种SH-5YSY-695细胞。24小时后,用含有10μg/mL等量的未加工姜黄素或新型姜黄素制剂(NCF)(实施例1)的培养基,CuSO4和H2O2替换原培养基,以诱导代表阿尔茨海默病的细胞毒性。制剂的制备采用无菌水。细胞存活性通过MTT([3-(4,5-二甲基噻唑-2-基)-2,5-二苯基四氮唑溴盐],噻唑蓝)方法测量。20小时后,每孔加入20μL MTT(Sigma-Aldrich,USA,5mg/ml PBS),温育1小时。加入150μL DMSO溶解不溶性紫色formazan产物,以生成有色溶液。在多孔扫描分光光度计(BIO-RAD型号2550EIA读出仪)上以600nm波长对光密度(OD)读数。SH-5YSY-695 cells were seeded at a density of 5 x 10 3 cells/well in 96-well plates. After 24 hours, the original medium was replaced with a medium containing 10 μg/mL of unprocessed curcumin or a novel curcumin preparation (NCF) (Example 1), CuSO 4 and H 2 O 2 to induce the representative of Alz. Cytotoxicity of Haimer's disease. The preparation was prepared using sterile water. Cell viability was measured by MTT ([3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide], thiazolyl blue) method. After 20 hours, 20 μL of MTT (Sigma-Aldrich, USA, 5 mg/ml PBS) was added to each well and incubated for 1 hour. 150 μL of DMSO was added to dissolve the insoluble purple formazan product to produce a colored solution. The optical density (OD) was read at a wavelength of 600 nm on a multi-hole scanning spectrophotometer (BIO-RAD model 2550 EIA reader).
参见图10,SH-SY5Y-APP695细胞在经由CuSO4和H2O2诱导胞毒后的存活性,通过MTT分析加以评价。相比未加工姜黄素的抗阿尔茨海默病效果,新型姜黄素制剂(NCF)(实施例1)显示显著更好的功效,提供优异的神经保护。 Referring to Figure 10, the viability of SH-SY5Y-APP695 cells after induction of cytotoxicity via CuSO 4 and H 2 O 2 was evaluated by MTT assay. The novel curcumin formulation (NCF) (Example 1) showed significantly better efficacy than the anti-Alzheimer's disease effect of unprocessed curcumin, providing excellent neuroprotection.
效果例7Effect example 7
(1)细胞摄取研究(1) Cellular uptake study
以SH-SY5Y-APP695细胞评价姜黄素和新型姜黄素制剂(NCF)(实施例1)的细胞摄取。简言之,将SH-SY5Y-APP695细胞以密度为5×104细胞/孔接种在24孔板(Corning,NY,USA)。37℃温育24小时后,附着细胞用或不用浓度为10μg/ml的天然姜黄素和等浓度的NCF处理,并在37℃下维持在细胞培养器(Hera Cell,Thermo Scientific,Waltham,MA)中。6小时后,细胞以PBS(0.01M,pH 7.4)洗两次,并加入甲醇裂解。细胞裂解液以10,000rpm、4℃离心10分钟。通过LC/MS/MS,测量收集的上清液中姜黄素的浓度。各测量重复三次,获得的数据以三次实验的平均值表示。Cellular uptake of curcumin and the novel curcumin preparation (NCF) (Example 1) was evaluated by SH-SY5Y-APP695 cells. Briefly, SH-SY5Y-APP695 cells were seeded at a density of 5 x 10 4 cells/well in 24-well plates (Corning, NY, USA). After 24 hours of incubation at 37 ° C, adherent cells were treated with or without natural curcumin at a concentration of 10 μg/ml and at an equal concentration of NCF and maintained at 37 ° C in a cell culture device (Hera Cell, Thermo Scientific, Waltham, MA). in. After 6 hours, the cells were washed twice with PBS (0.01 M, pH 7.4) and methanol was added for lysis. The cell lysate was centrifuged at 10,000 rpm for 10 minutes at 4 °C. The concentration of curcumin in the collected supernatant was measured by LC/MS/MS. Each measurement was repeated three times and the obtained data was expressed as the average of three experiments.
LC/MS/MS方法:LC/MS/MS method:
样品分析在Quadrapole LC/MS/MS(Shimadzu,Kyoto,Japan)***上操作,该***装配有API 3000质谱仪,Shimadzu SIL 20A自动加样器,Shimadzu LC20AD泵和分析1.6.2数据处理器。采用新开发和验证的LC/MS/MS方法,对血浆中姜黄素的浓度定量。抽提物复溶在甲醇/水(50∶50)中,注射至Shimadzu Nexera HPLC***,在Kinetex C18 2.6mm×50mm×3mm柱(Phenomenex)解析,移动相流速0.2ml/min,注射体积15μl。移动相A(MPA)为5%甲醇和0.1%甲酸的水溶液,而移动相B(MPB)为95%甲醇和0.1%甲酸的水溶液。移动相时间表设定的梯度为:起始10%MPB,至第1.5分钟时100%MPB,维持95%MPB6分钟,然后10%MPB 30秒,准备下一样品。各样品分析的总运行时间为10分钟。将柱洗脱液引入负离子模式电喷雾(ESI)质谱分析。离子源的操作参数包括分析物依赖性参数和源依赖性参数,优化得到质谱仪分析的最佳性能。通过监控前体离子进行MRM分析,产生质荷比(m/z)如下:姜黄素367.0/134.20和法华林307.2/161.2。以0空气作为源气,而氮既用作气帘气也用作碰撞气。从化合物得到峰面积,内标(IS)和已知浓度的校准物用作构建化合物/IS面积之比的校准曲线。定量限为5ng/ml。各化合物的日内和日间变异性在15%内。 Sample analysis was performed on a Quadrapole LC/MS/MS (Shimadzu, Kyoto, Japan) system equipped with an API 3000 mass spectrometer, a Shimadzu SIL 20A autoloader, a Shimadzu LC20AD pump and an analytical 1.6.2 data processor. The concentration of curcumin in plasma was quantified using a newly developed and validated LC/MS/MS method. The extract was reconstituted in methanol/water (50:50), injected into a Shimadzu Nexera HPLC system, and analyzed on a Kinetex C18 2.6 mm x 50 mm x 3 mm column (Phenomenex) with a mobile phase flow rate of 0.2 ml/min and an injection volume of 15 μl. Mobile phase A (MPA) was an aqueous solution of 5% methanol and 0.1% formic acid, while mobile phase B (MPB) was an aqueous solution of 95% methanol and 0.1% formic acid. The gradient set by the mobile phase schedule was: starting 10% MPB, 100% MPB at 1.5 minutes, maintaining 95% MPB for 6 minutes, then 10% MPB for 30 seconds, preparing the next sample. The total run time for each sample analysis was 10 minutes. The column eluate was introduced into negative ion mode electrospray (ESI) mass spectrometry. The operational parameters of the ion source include analyte dependent parameters and source dependent parameters, optimized for optimal performance from mass spectrometer analysis. The mass-to-charge ratio (m/z) was generated by monitoring the precursor ions for MRM analysis as follows: curcumin 367.6/132.20 and Fahualin 307.2/161.2. Zero air is used as the source gas, and nitrogen is used as both the curtain gas and the collision gas. The peak area was obtained from the compound, and the internal standard (IS) and the calibrant of known concentration were used as a calibration curve for constructing the ratio of compound/IS area. The limit of quantification is 5 ng/ml. The intra- and inter-day variability of each compound was within 15%.
细胞摄取是重要参数,需要该参数来解释本发明制剂如何成功递送至癌症组织。参见图11,在浓度为10μg/mL时,NCF的细胞摄取相比天然姜黄素,提高了87%。因此,细胞摄取研究证实NCF比天然姜黄素更好的摄取。该研究获得的结果支持了相比未加工姜黄素,NCF对细胞具有更为优异的神经保护作用。Cell uptake is an important parameter that is required to explain how the formulation of the invention is successfully delivered to cancerous tissue. Referring to Figure 11, at a concentration of 10 μg/mL, the cellular uptake of NCF was increased by 87% compared to natural curcumin. Therefore, cell uptake studies have confirmed that NCF is better than natural curcumin. The results obtained in this study support the superior neuroprotective effects of NCF on cells compared to unprocessed curcumin.
(2)荧光显微观察(2) Fluorescence microscopic observation
为了定量细胞摄取研究,将SH-SY5Y-APP695细胞以密度为15×104细胞/孔接种在35mm培养板(Corning,NY,USA),对于,用于荧光显微研究。37℃温育24小时后,附着细胞用恒定浓度(10μg/ml)的天然姜黄素和等浓度的NCF(实施例1)37℃下在细胞培养器(Hera Cell,Thermo Scientific,Waltham,MA)中处理2小时。温育后,细胞单层以1ml PBS(0.01M,pH 7.4)冲洗三次,以去除过量的固体分散体(SD)或天然姜黄素。板中加入新鲜PBS(0.01M,pH 7.4),观察细胞,以蓝色光镜通过激发姜黄素拍照。To quantify cell uptake studies, the SH-SY5Y-APP695 cells at a density of 15 × 10 4 cells / well were seeded in 35mm culture plates (Corning, NY, USA), for, for fluorescence microscopy. After incubation for 24 hours at 37 ° C, adherent cells were incubated with a constant concentration (10 μg/ml) of natural curcumin and an equal concentration of NCF (Example 1) at 37 ° C in a cell culture device (Hera Cell, Thermo Scientific, Waltham, MA). Processing in 2 hours. After the incubation, the cell monolayer was washed three times with 1 ml of PBS (0.01 M, pH 7.4) to remove excess solid dispersion (SD) or natural curcumin. Fresh PBS (0.01 M, pH 7.4) was added to the plate, and the cells were observed, and photos were photographed by exciting the curcumin with a blue light microscope.
利用姜黄素的光化学特性,通过荧光光度计,将NCF的胞内摄取与天然姜黄素比较。参见图12,通过测量姜黄素的荧光强度,与天然姜黄素相比,NCF显示显著更高的细胞摄取。Using the photochemical properties of curcumin, the intracellular uptake of NCF was compared to natural curcumin by a fluorophotometer. Referring to Figure 12, by measuring the fluorescence intensity of curcumin, NCF showed significantly higher cellular uptake compared to native curcumin.
效果例8Effect example 8
溶出研究Dissolution study
采用USP II型桨装置(AT 7 Smart,Sotax GmbH,Germany)进行纯姜黄素、物理混合物(PM)和固体分散体(SD)(实施例1)形式的姜黄素的溶出。操作参数:50rpm转速,37±0.5℃温度,SIF(模拟肠液)pH 6.8(USP)。将20mg姜黄素等量的制剂填入“2”号硬明胶胶囊。胶囊置于沉子内,并放入溶出容器中。不同时间间隔采集样品,并且每次用等量新鲜溶出介质替换。样品通过0.45μm PVDF针头过滤器过滤,并通过早先开发的HPLC方法分析。 The dissolution of curcumin in the form of pure curcumin, physical mixture (PM) and solid dispersion (SD) (Example 1) was carried out using a USP Type II paddle device (AT 7 Smart, Sotax GmbH, Germany). Operating parameters: 50 rpm, 37 ± 0.5 ° C temperature, SIF (simulated intestinal fluid) pH 6.8 (USP). An equal amount of 20 mg of curcumin was filled into a "2" hard gelatin capsule. The capsule is placed in a sink and placed in a dissolution vessel. Samples were taken at different time intervals and replaced each time with an equal amount of fresh dissolution medium. Samples were filtered through a 0.45 [mu]m PVDF syringe filter and analyzed by previously developed HPLC methods.
如图13所示,未加工姜黄素在溶出介质中实质上维持不溶2h。与未加工姜黄素相比,由于胶束化导致药物增溶,PM中的姜黄素溶出稍高。相比PM,固体分散体(SD)(实施例1)显示了显著高的释放速率。这暗示,姜黄素在SD主要以无定形态存在,因而具有更高的溶解度。120分钟内,观察到SD的溶出率为100%。As shown in Figure 13, the unprocessed curcumin remained substantially insoluble for 2 h in the dissolution medium. Compared to unprocessed curcumin, the dissolution of the drug in the PM is slightly higher due to the solubilization of the drug due to micellization. The solid dispersion (SD) (Example 1) showed a significantly higher release rate than PM. This suggests that curcumin exists mainly in amorphous form in SD and thus has higher solubility. Within 120 minutes, the dissolution rate of SD was observed to be 100%.
效果例9Effect Example 9
药代动力学研究Pharmacokinetic study
试验开始前至少1周获取雄性SD大鼠(250±10g),以便实验室中为其调节环境、食物和水。术前麻醉大鼠。在颈部和更接近颈静脉区域作纵向切口。随后,以20单位/ml肝素生理盐水填充导管,并***颈静脉,直至第一硅胶塞。缝合胶塞和肌肉将其固定在那儿。导管的另一端穿过颈部皮下,更接近双耳。最后,以500单位/ml肝素生理盐水填充导管,并塞进导管的游离端。手术完成后,将大鼠置于不同笼中恢复。第二天,对每只大鼠进行药代动力学研究。给药前,动物禁食12小时,随意饮用水。Male Sprague-Dawley rats (250 ± 10 g) were obtained at least 1 week prior to the start of the trial to regulate the environment, food and water in the laboratory. Rats were anesthetized before surgery A longitudinal incision is made in the neck and closer to the jugular vein area. Subsequently, the catheter was filled with 20 units/ml of heparin saline and inserted into the jugular vein until the first silica gel plug. Stitch the rubber plug and muscle to secure it there. The other end of the catheter is subcutaneously through the neck, closer to the ears. Finally, the catheter was filled with 500 units/ml heparin saline and inserted into the free end of the catheter. After the surgery was completed, the rats were placed in different cages to recover. The next day, a pharmacokinetic study was performed on each rat. Prior to dosing, the animals were fasted for 12 hours with free access to drinking water.
姜黄素悬浮液(CS)的制备是将姜黄素加入0.5%羧甲基纤维素钠(CMC-Na)溶液,然后超声处理几分钟,得到均匀悬浮液。新型姜黄素制剂(NCF)(实施例1)溶解于mili Q水中。两组大鼠口服给药姜黄素悬浮液和NCF,剂量等同于50mg/kg的姜黄素。经口饲施用药物和制剂之后,在0,15,30,45,60,90,120,180,240,300,360,420,480和720分钟的时间间隔,采集0.2ml血样。每次血样采集时,导管都用相同量的肝素生理盐水冲洗。血样采集后,5000rpm、4℃离心5分钟,将血浆与血液分离。血浆分离后存储在-20℃,直到分析。900μl含有华法林(warfarin)的冰冷甲醇,作为内标(400ng/ml)加至100μL血浆样品,接着旋转震荡10分钟,13,000rpm离心5分钟,然后氮气干燥。在注射至LC/MS/MS前,提取物以甲醇/水(50∶50)复溶,并通过0.22μm膜过滤器过滤。采用Phoenix WinNonlin(Pharsight,St.Louis, MO)对各个浓度-时间特性进行非房室药代动力学分析(noncompartmental pharmacokinetics analysis)。The curcumin suspension (CS) was prepared by adding curcumin to a 0.5% sodium carboxymethylcellulose (CMC-Na) solution and then sonicating for a few minutes to obtain a homogeneous suspension. The new curcumin formulation (NCF) (Example 1) was dissolved in mili Q water. Two groups of rats were orally administered with curcumin suspension and NCF at a dose equivalent to 50 mg/kg of curcumin. After administration of the drug and formulation by oral gavage, 0.2 ml of blood samples were taken at time intervals of 0, 15, 30, 45, 60, 90, 120, 180, 240, 300, 360, 420, 480 and 720 minutes. The catheter was flushed with the same amount of heparin saline each time the blood sample was collected. After the blood sample was collected, it was centrifuged at 5000 rpm and 4 ° C for 5 minutes to separate the plasma from the blood. Plasma was stored at -20 °C after separation until analysis. 900 μl of ice-cold methanol containing warfarin was added as an internal standard (400 ng/ml) to 100 μL of plasma sample, followed by shaking for 10 minutes, centrifugation at 13,000 rpm for 5 minutes, and then drying with nitrogen. The extract was reconstituted with methanol/water (50:50) prior to injection into LC/MS/MS and filtered through a 0.22 [mu]m membrane filter. Using Phoenix WinNonlin (Pharsight, St. Louis, MO) Noncompartmental pharmacokinetics analysis of each concentration-time characteristic.
参见图14和表4,纯姜黄素悬浮液(CS)和新型姜黄素制剂(NCF)(实施例1)的Cmax值之间存在显著差异。与纯姜黄素相比,NCF的生物利用度改善了近123倍。姜黄素在胃肠道中有效的增溶和抗降解是生物利用度改善的可能原因。而且,载体Soluplus是聚合物,也诱导了肠上皮渗透性增加。Referring to Figure 14 and Table 4, there was a significant difference between the Cmax values of the pure curcumin suspension (CS) and the novel curcumin formulation (NCF) (Example 1). Compared to pure curcumin, the bioavailability of NCF has improved by nearly 123 times. Effective solubilization and anti-degradation of curcumin in the gastrointestinal tract is a possible cause of improved bioavailability. Moreover, the carrier Soluplus is a polymer that also induces an increase in intestinal epithelial permeability.
表4:姜黄素和NCF(50mg/kg)的药代动力学参数(平均值±标准差)(N=3)Table 4: Pharmacokinetic parameters of curcumin and NCF (50 mg/kg) (mean ± standard deviation) (N=3)
参数parameter 姜黄素悬浮液Curcumin suspension NCFNCF
Cmax(ng/mL)Cmax(ng/mL) 38.7561238.75612 3657.5523,657.552
Tmax(mins)Tmax(mins) 6060 180180
AUC0-t(ng·min/mL)AUC0-t (ng·min/mL) 6511.45316511.4531 801873.35801873.35
AUC0-∞(ng·min/mL)AUC0-∞ (ng·min/mL) 6607.57796607.5779 806448.55806448.55
F0-t(%)F0-t (%) -- 123.1481418123.1481418
F0-∞(%)F0-∞ (%) -- 122.0490416122.0490416

Claims (39)

  1. 药物递送***,其包含活性成分姜黄素或其衍生物或其药学上可接受的盐和聚合物载体Soluplus。A drug delivery system comprising the active ingredient curcumin or a derivative thereof, or a pharmaceutically acceptable salt thereof, and a polymeric carrier, Soluplus.
  2. 权利要求1的药物递送***,其中姜黄素与Soluplus的重量比为1∶0.001-1∶100。The drug delivery system of claim 1 wherein the weight ratio of curcumin to Soluplus is from 1:0.001 to 1:100.
  3. 权利要求1的药物递送***,其还包含其他聚合物载体和/或表面活性剂。The drug delivery system of claim 1 further comprising other polymeric carriers and/or surfactants.
  4. 权利要求3的药物递送***,其中所述其他聚合物载体为水溶性聚合物,选自N-乙烯基内酰胺均聚物、N-乙烯基内酰胺共聚物、纤维素酯、纤维素醚、聚亚烷基氧化物、聚丙烯酸酯、聚甲基丙烯酸酯、丙烯酸的均聚物和共聚物、甲基丙烯酸的均聚物和共聚物、聚丙烯酰胺、聚乙烯醇、乙酸乙烯酯聚合物、乙酸乙烯酯共聚物、羧乙烯基聚合物、寡糖、多糖及其混合物。The drug delivery system of claim 3 wherein said other polymeric carrier is a water soluble polymer selected from the group consisting of N-vinyl lactam homopolymers, N-vinyl lactam copolymers, cellulose esters, cellulose ethers, Polyalkylene oxides, polyacrylates, polymethacrylates, homopolymers and copolymers of acrylic acid, homopolymers and copolymers of methacrylic acid, polyacrylamide, polyvinyl alcohol, vinyl acetate polymers , vinyl acetate copolymers, carboxyvinyl polymers, oligosaccharides, polysaccharides, and mixtures thereof.
  5. 权利要求3的药物递送***,其中所述其他聚合物载体选自烷基纤维素、羟烷基纤维素、羟烷基烷基纤维素、甲基纤维素(MC)、乙基纤维素(EC)、羟乙基纤维素(HEC)、羟丙基纤维素(HPC)、羟丙基甲基纤维素(HPMC)、羟乙基甲基纤维素(HEMC)、羟丙基甲基纤维素琥珀酸酯、羟丙基甲基纤维素乙酸琥珀酸酯、羧甲基乙基纤维素、羧甲基纤维素钠、羧甲基纤维素钾、纤维素乙酸琥珀酸酯、纤维素乙酸邻苯二甲酸酯、羟丙基甲基纤维素邻苯二甲酸酯、聚丙烯酸共聚合物、聚(甲基)丙烯酸聚合物、聚(羟烷基丙烯酸酯)、聚(羟烷基甲基丙烯酸酯)、聚乙烯吡咯烷酮(PVP)、乙烯基吡咯烷酮均聚物、乙烯基吡咯烷酮共聚物、聚维酮、乙烯基吡咯烷酮-乙烯基乙酸酯共聚物(共聚维酮)、乙酸乙烯酯的共聚合物、丙酸乙烯酯的共 聚物、乙酸乙烯酯和巴豆酸的共聚物、聚乙二醇、聚乙烯醇、部分水解的聚乙酸乙烯酯、明胶、藻酸钠、可溶性淀粉、***胶、糊精、透明质酸、软骨素硫酸钠、藻酸丙二醇酯、琼脂、黄芪胶、黄原胶、氨基烷基甲基丙烯酸酯共聚物、聚乙酸乙烯酯-二乙基氨基乙酸酯、甲基丙烯酸酯共聚物、甲基丙烯酸共聚物L、甲基丙烯酸共聚物LD、甲基丙烯酸共聚物S、聚乙二醇(macrogol)、聚氧乙烯、聚氧丙烯、环氧乙烷(EO)和环氧丙烷(PO)的共聚物、卡拉胶、半乳甘露聚糖及其组合物。The drug delivery system of claim 3 wherein said other polymeric carrier is selected from the group consisting of alkyl cellulose, hydroxyalkyl cellulose, hydroxyalkyl alkyl cellulose, methyl cellulose (MC), ethyl cellulose (EC) ), hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), hydroxyethyl methyl cellulose (HEMC), hydroxypropyl methyl cellulose amber Acid ester, hydroxypropyl methylcellulose acetate succinate, carboxymethylethyl cellulose, sodium carboxymethyl cellulose, potassium carboxymethyl cellulose, cellulose acetate succinate, cellulose acetate phthalate Formate, hydroxypropyl methylcellulose phthalate, polyacrylic acid copolymer, poly(meth)acrylic acid polymer, poly(hydroxyalkyl acrylate), poly(hydroxyalkyl methacrylate Ester), polyvinylpyrrolidone (PVP), vinylpyrrolidone homopolymer, vinylpyrrolidone copolymer, povidone, vinylpyrrolidone-vinyl acetate copolymer (copovidone), copolymerization of vinyl acetate Of a total of vinyl acetate Polymer, copolymer of vinyl acetate and crotonic acid, polyethylene glycol, polyvinyl alcohol, partially hydrolyzed polyvinyl acetate, gelatin, sodium alginate, soluble starch, gum arabic, dextrin, hyaluronic acid, cartilage Sodium sulphate, propylene glycol alginate, agar, tragacanth, xanthan gum, aminoalkyl methacrylate copolymer, polyvinyl acetate-diethylaminoacetate, methacrylate copolymer, methyl Acrylic copolymer L, methacrylic copolymer LD, methacrylic acid copolymer S, macrogol, polyoxyethylene, polyoxypropylene, ethylene oxide (EO) and propylene oxide (PO) Copolymers, carrageenan, galactomannans, and combinations thereof.
  6. 权利要求3的药物递送***,其中所述其他聚合物载体选自羟丙基甲基纤维素(HPMC)、聚乙二醇(PEG)、壳聚糖、PVP、PVP/VA、HPC、羟丙基甲基纤维素乙酸酯(HPMCAS)、eudragit E100、基于甲基丙烯酸二甲基氨基乙酯、甲基丙烯酸丁酯和甲基丙烯酸甲酯的阳离子共聚物。The drug delivery system of claim 3 wherein said other polymeric carrier is selected from the group consisting of hydroxypropyl methylcellulose (HPMC), polyethylene glycol (PEG), chitosan, PVP, PVP/VA, HPC, hydroxypropyl Methylcellulose acetate (HPMCAS), eudragit E100, a cationic copolymer based on dimethylaminoethyl methacrylate, butyl methacrylate and methyl methacrylate.
  7. 权利要求3的药物递送***,其中所述表面活性剂包括阴性、阳性或两性表面活性剂,并且选自十二烷基磺酸钠、十二烷基硫酸钠(SDS),月桂基硫酸钠(SLS)、聚氧乙烯山梨醇酐长链脂肪酸酯、维生素E-TPGS、胆盐、脱氧胆酸钠、甘胆酸钠、聚氧乙烯聚氧丙烯二醇及其组合。The drug delivery system of claim 3 wherein said surfactant comprises a negative, positive or amphoteric surfactant and is selected from the group consisting of sodium dodecyl sulfate, sodium dodecyl sulfate (SDS), sodium lauryl sulfate ( SLS), polyoxyethylene sorbitan long chain fatty acid ester, vitamin E-TPGS, bile salt, sodium deoxycholate, sodium glycocholate, polyoxyethylene polyoxypropylene glycol, and combinations thereof.
  8. 权利要求1的药物递送***,其选自基于纳米技术的药物递送***,固溶体***以及乳液***。The drug delivery system of claim 1 selected from the group consisting of nanotechnology based drug delivery systems, solid solution systems, and emulsion systems.
  9. 权利要求8的药物递送***,其中所述基于纳米技术的药物递送***选自胶束、纳米颗粒、纳米纤维和纳米悬浮液,而所述基于固溶体***选自固体分散体、挤出物和固体载体***。The drug delivery system of claim 8 wherein said nanotechnology-based drug delivery system is selected from the group consisting of micelles, nanoparticles, nanofibers, and nanosuspensions, and said solid solution based system is selected from the group consisting of solid dispersions, extrudates, and solids Carrier system.
  10. 权利要求1的药物递送***,其为固体分散体制剂。 The drug delivery system of claim 1 which is a solid dispersion formulation.
  11. 权利要求10的药物递送***,其中所述固体分散体还含有TPGS 1000。The drug delivery system of claim 10 wherein said solid dispersion further comprises TPGS 1000.
  12. 权利要求1的药物递送***,其为胶束制剂。The drug delivery system of claim 1 which is a micelle formulation.
  13. 权利要求12的药物递送***,其中所述胶束制剂还含有TPGS 1000。The drug delivery system of claim 12, wherein the micelle formulation further comprises TPGS 1000.
  14. 权利要求12或13的药物递送***,其中所述胶束进一步含有其他聚合物载体与水/缓冲剂,以及其中有效量的姜黄素包合在胶束中。The drug delivery system of claim 12 or 13, wherein the micelle further comprises other polymeric carriers and water/buffering agents, and wherein an effective amount of curcumin is encapsulated in the micelles.
  15. 权利要求12或13的药物递送***,其中所述胶束进一步包含表面活性剂、固相吸附剂、酸化剂和/或抗氧化剂。The drug delivery system of claim 12 or 13, wherein the micelle further comprises a surfactant, a solid phase adsorbent, an acidulant, and/or an antioxidant.
  16. 固体分散体制剂的制备方法,包括下列步骤:A method of preparing a solid dispersion formulation comprising the steps of:
    将活性成分姜黄素或其衍生物或其可药用盐分散在聚合物载体和任选的表面活性剂中。The active ingredient curcumin or a derivative thereof, or a pharmaceutically acceptable salt thereof, is dispersed in a polymeric carrier and optionally a surfactant.
  17. 权利要求16的制备方法,其进一步包括选自以下的步骤:融冰浴搅拌、薄膜冷却、液氮、喷雾凝结、热熔挤出、MeltrexTM、熔融凝聚、或溶剂蒸发(烘干、真空干燥、旋转蒸发、热板加热、喷雾干燥、冷冻干燥、超临界抗溶剂、共沉淀、静电纺丝、喷雾冷干、超快冷干、流体床涂布)和溶剂熔融。The method of preparation of claim 16, further comprising a step selected from: melting ice bath with stirring, the film was cooled with liquid nitrogen, spray congealing, hot melt extrusion, Meltrex TM, melt agglomeration, or solvent evaporation (drying, dried in vacuo , rotary evaporation, hot plate heating, spray drying, freeze drying, supercritical antisolvent, coprecipitation, electrospinning, spray lyophilization, ultra-fast lyophilization, fluid bed coating) and solvent melting.
  18. 胶束的制备方法,包括以下步骤:The preparation method of the micelle includes the following steps:
    将活性成分姜黄素或其衍生物或其药学上可接受的盐、聚合物载体和表面活性剂,任选溶解于有机溶剂中,通过旋转蒸发除去有机溶剂,膜形成后,真空干燥,加入缓冲剂水合,超声处理。 The active ingredient curcumin or a derivative thereof, or a pharmaceutically acceptable salt thereof, a polymer carrier and a surfactant are optionally dissolved in an organic solvent, and the organic solvent is removed by rotary evaporation. After the film is formed, it is vacuum dried and added to a buffer. The agent is hydrated and sonicated.
  19. 基于脂质的药物递送***,其包含活性成分姜黄素或其衍生物或其药学上可接受的盐和脂质。A lipid-based drug delivery system comprising the active ingredient curcumin or a derivative thereof, or a pharmaceutically acceptable salt thereof and a lipid.
  20. 权利要求19的基于脂质的药物递送***,其中所述脂质为甘油三酯,包括长链甘油三酯(LCT)、中链甘油三酯(MCT)和短链甘油三酯(SCT),其中长链甘油三酯选自氢化大豆油、氢化植物油、玉米油、橄榄油、豆油、花生油和芝麻油,中链甘油三酯选自来自可可油或棕榈籽油的辛酸/葵酸甘油三酯。The lipid-based drug delivery system of claim 19, wherein the lipid is a triglyceride, including long chain triglycerides (LCT), medium chain triglycerides (MCT), and short chain triglycerides (SCT), Wherein the long chain triglyceride is selected from the group consisting of hydrogenated soybean oil, hydrogenated vegetable oil, corn oil, olive oil, soybean oil, peanut oil and sesame oil, and the medium chain triglyceride is selected from the group consisting of caprylic/maronic acid triglycerides from cocoa butter or palm seed oil.
  21. 权利要求19的基于脂质的药物递送***,其还包括赋形剂,选自化学甘油三酯、部分甘油三酯、半合成油性酯和半合成非离子表面活性剂酯。The lipid-based drug delivery system of claim 19, further comprising an excipient selected from the group consisting of chemical triglycerides, partial triglycerides, semi-synthetic oily esters, and semi-synthetic nonionic surfactant esters.
  22. 权利要求19的基于脂质的药物递送***,其还包括水不溶性赋形剂,选自蜂蜡、油酸、大豆脂肪酸、维生素E、玉米油单-二-甘油三酯、中链(C8/C10)甘油单酯和甘油二酯,以及脂肪酸的丙二醇酯。The lipid-based drug delivery system of claim 19, further comprising a water-insoluble excipient selected from the group consisting of beeswax, oleic acid, soy fatty acid, vitamin E, corn oil mono-di-triglyceride, medium chain (C8/C10) Monoglycerides and diglycerides, and propylene glycol esters of fatty acids.
  23. 权利要求19的基于脂质的药物递送***,其中所述脂质选自Caproyl 90,Capmul MCM和CaproylTM PGMC中的一种或多种。Lipid-based drug delivery system as claimed in claim 19, wherein said lipid is selected from Caproyl 90, Capmul MCM, and Caproyl TM PGMC of one or more.
  24. 权利要求19的基于脂质的药物递送***,其还包括水溶性有机溶剂、表面活性剂、辅助表面活性剂、聚合物增溶剂、磷脂和/或一种或多种添加剂。The lipid-based drug delivery system of claim 19, further comprising a water-soluble organic solvent, a surfactant, a co-surfactant, a polymer solubilizer, a phospholipid, and/or one or more additives.
  25. 权利要求24的基于脂质的药物递送***,其中水溶性有机溶剂选自PEG 200-10,000、聚乙烯己内酰胺(PCL)、聚乙酸乙烯酯(PVA)或其共聚物、水溶性形式的维生素E和乙醇;表面活性剂是其中脂肪酸为不饱和或饱和的膳食油的衍生物,通过PEG与水解植物油反应、醇与氧化乙烯反应生成烷基醚乙氧基化物、或者基于聚山梨醇酯的植物油与氧化乙烯反应而合成;辅助表面活性剂基于聚乙二醇、聚丙二 醇、乙醇和甘油;聚合物增溶剂选自Soluplus、壳聚糖、聚乙烯吡咯烷酮(PVP)、PVP/VA、HPC、HPMC、HPMCAS、eudragit E100、基于甲基丙烯酸二甲基氨基乙酯、甲基丙烯酸丁酯和甲基丙烯酸甲酯的阳离子共聚物。The lipid-based drug delivery system of claim 24, wherein the water-soluble organic solvent is selected from the group consisting of PEG 200-10,000, polyvinyl caprolactam (PCL), polyvinyl acetate (PVA) or a copolymer thereof, and a water-soluble form of vitamin E and Ethanol; a surfactant is a derivative of a dietary oil in which the fatty acid is unsaturated or saturated, reacted with a hydrolyzed vegetable oil by PEG, reacted with an ethylene oxide to form an alkyl ether ethoxylate, or a polysorbate-based vegetable oil and Synthesis by ethylene oxide reaction; auxiliary surfactant based on polyethylene glycol, polypropylene Alcohol, ethanol and glycerol; polymer solubilizer is selected from the group consisting of Soluplus, chitosan, polyvinylpyrrolidone (PVP), PVP/VA, HPC, HPMC, HPMCAS, eudragit E100, dimethylaminoethyl methacrylate, A A cationic copolymer of butyl acrylate and methyl methacrylate.
  26. 权利要求25的基于脂质的药物递送***,其中PEG 200-10,000选自PEG 300,PEG 400,PEG 1,000和PEG 6,000;表面活性剂选自Cremophor RH 40、Labrasol、TPGS 1000、Tween 20、Cremophor E1和Tween 80;以及辅助表面活性剂选自PEG 300、PEG 400、丙二醇、甘油、乙醇、Transcutol HP和Transcutol P。The lipid-based drug delivery system of claim 25, wherein the PEG 200-10,000 is selected from the group consisting of PEG 300, PEG 400, PEG 1,000 and PEG 6,000; the surfactant is selected from the group consisting of Cremophor RH 40, Labrasol, TPGS 1000, Tween 20, Cremophor E1 And Tween 80; and the co-surfactant is selected from the group consisting of PEG 300, PEG 400, propylene glycol, glycerin, ethanol, Transcutol HP, and Transcutol P.
  27. 权利要求24的基于脂质的药物递送***,其中所述添加剂包括固相吸附剂、水溶性和脂溶性抗氧化剂、酸化剂、螯合剂、防腐剂、稳定剂和/或缓冲剂,其中固相吸附剂包括硅基吸附剂和非硅基吸附剂,硅基吸附剂选自Aerosil 200和偏硅酸镁铝,非硅基吸附剂选自微晶纤维素、滑石、无水磷酸氢二钙(DCPA)、由烷基纤维素、羟烷基纤维素、羟烷基烷基纤维素糖等基团组成的水溶性聚合物;螯合剂为选自乙二胺、乙二胺四乙酸二钠钙和乙二胺四乙酸二钠的至少一种;酸化剂选自柠檬酸、乙酸、富马酸、盐酸和硝酸;缓冲剂选自偏磷酸钾、磷酸二氢钾、醋酸钠、柠檬酸钠;水溶性或脂溶性抗氧化剂选自抗坏血酸、抗坏血酸棕榈酸酯、丁基羟基茴香醚、丁基羟基甲苯、次磷酸、硫代甘油、没食子酸丙酯、抗坏血酸钠、亚硫酸氢钠、甲醛次硫酸氢钠、次硫酸盐、焦亚硫酸钠。The lipid-based drug delivery system of claim 24, wherein said additive comprises a solid phase adsorbent, a water soluble and fat soluble antioxidant, an acidulant, a chelating agent, a preservative, a stabilizer, and/or a buffer, wherein the solid phase The adsorbent comprises a silicon-based adsorbent and a non-silicon-based adsorbent, the silicon-based adsorbent is selected from the group consisting of Aerosil 200 and magnesium aluminum metasilicate, and the non-silicon-based adsorbent is selected from the group consisting of microcrystalline cellulose, talc, and anhydrous dibasic calcium phosphate ( DCPA), a water-soluble polymer composed of a group such as an alkyl cellulose, a hydroxyalkyl cellulose, a hydroxyalkyl alkyl cellulose sugar; the chelating agent is selected from the group consisting of ethylenediamine, calcium disodium edetate And at least one of disodium edetate; the acidifying agent is selected from the group consisting of citric acid, acetic acid, fumaric acid, hydrochloric acid and nitric acid; the buffering agent is selected from the group consisting of potassium metaphosphate, potassium dihydrogen phosphate, sodium acetate, sodium citrate; The water-soluble or fat-soluble antioxidant is selected from the group consisting of ascorbic acid, ascorbyl palmitate, butylhydroxyanisole, butylhydroxytoluene, hypophosphorous acid, thioglycerol, propyl gallate, sodium ascorbate, sodium hydrogen sulfite, formaldehyde sulfuric acid Sodium hydrogen, hyposulfite, Sulfite.
  28. 权利要求19的基于脂质的药物递送***,除姜黄素之外,其还包括CapryolTM PGMC、
    Figure PCTCN2017094665-appb-100001
    Labrasol、TPGS 1000、Transcutol P和/或Aerosil 200。
    Lipid-based drug delivery system as claimed in claim 19, in addition to curcumin, further comprising Capryol TM PGMC,
    Figure PCTCN2017094665-appb-100001
    Labrasol, TPGS 1000, Transcutol P and/or Aerosil 200.
  29. 权利要求19的基于脂质的药物递送***,其选自脂质溶液、脂质体悬浮液、表面活性剂或聚合物-脂质混合的胶束,自微乳化药 物递送***(SMEDDS)和纳米乳液制剂。The lipid-based drug delivery system of claim 19, which is selected from the group consisting of a lipid solution, a liposome suspension, a surfactant, or a polymer-lipid mixed micelle, from a microemulsion Delivery system (SMEDDS) and nanoemulsion formulations.
  30. 权利要求29的基于脂质的药物递送***,其中SMEDDS为固相,还包括固相吸附剂。The lipid-based drug delivery system of claim 29, wherein the SMEDDS is a solid phase, further comprising a solid phase adsorbent.
  31. 权利要求30的基于脂质的药物递送***,其中所述固体吸附剂为Aerosil 200。The lipid-based drug delivery system of claim 30, wherein the solid adsorbent is Aerosil 200.
  32. 权利要求29的基于脂质的药物递送***,其中纳米乳液制剂还包括水和/或缓冲剂。The lipid-based drug delivery system of claim 29, wherein the nanoemulsion formulation further comprises water and/or a buffer.
  33. 权利要求19-32任一项的基于脂质的药物递送***的制备方法,包括下列步骤:A method of preparing a lipid-based drug delivery system according to any of claims 19-32, comprising the steps of:
    将活性成分姜黄素或其药学上可接受的盐溶解在脂质,表面活性剂,或脂质与表面活性剂的混合物中。The active ingredient curcumin or a pharmaceutically acceptable salt thereof is dissolved in a lipid, a surfactant, or a mixture of a lipid and a surfactant.
  34. 权利要求1-15和19-32任一项的药物递送***,其为固体剂型,选自片剂、环剂、贴剂、胶囊、丸剂、颗粒剂、细粒剂或粉剂、粉末或条带(strip),通过口服、肠胃外、吸入、局部或经皮、鼻、眼内、耳、直肠、***途径给药。The drug delivery system according to any one of claims 1 to 15 and 19 to 32, which is a solid dosage form selected from the group consisting of a tablet, a ring, a patch, a capsule, a pill, a granule, a fine granule or a powder, a powder or a strip. (strip), administered by oral, parenteral, inhalation, topical or transdermal, nasal, intraocular, aural, rectal, vaginal routes.
  35. 权利要求1-15和19-32任一项的药物递送***,其为液体剂型,选自溶液、悬浮液、乳液、基于共溶剂的***、气溶胶,通过口服、肠胃外、吸入、局部或经皮、鼻内、眼内、耳、直肠、***途径给药。The drug delivery system according to any one of claims 1 to 15 and 19 to 32, which is in a liquid dosage form, selected from the group consisting of a solution, a suspension, an emulsion, a cosolvent-based system, an aerosol, by oral, parenteral, inhalation, topical or Transdermal, intranasal, intraocular, ear, rectal, vaginal routes of administration.
  36. 权利要求1-15和19-32任一项的药物递送***,其为半固体剂型,选自选自药膏、霜剂、凝胶、糊剂,通过外用或经皮、直肠、***途径给药,用于局部或全身目的。 The drug delivery system according to any one of claims 1 to 15 and 19 to 32, which is a semisolid dosage form selected from the group consisting of ointments, creams, gels, pastes, for administration by external or transdermal, rectal or vaginal routes. For local or systemic purposes.
  37. 权利要求1-15和19-32任一项的药物递送***,其还包括可药用赋形剂,所述赋形剂选自崩解剂、润滑剂、助流剂、抗粘附剂、惰性填料、润湿剂、pH改性剂、粘合剂、溶解度改性剂、重结晶抑制剂、稀释剂及其组合。The drug delivery system of any of claims 1-15 and 19-32, further comprising a pharmaceutically acceptable excipient selected from the group consisting of a disintegrant, a lubricant, a glidant, an anti-adhesive agent, Inert fillers, wetting agents, pH modifiers, binders, solubility modifiers, recrystallization inhibitors, diluents, and combinations thereof.
  38. 权利要求1-15和19-32任一项的药物递送***,其中在液体制剂中,姜黄素的含量为0.001-1000mg/ml,或者0.1-100mg/ml,或者10-20mg/ml,而在固体制剂中,姜黄素的剂量为0.001-1000mg/单位,或者0.1-100mg/单位,或者10-20mg/单位。The drug delivery system according to any one of claims 1 to 15 and 19 to 32, wherein in the liquid preparation, the curcumin is contained in an amount of 0.001 to 1000 mg/ml, or 0.1 to 100 mg/ml, or 10 to 20 mg/ml, and In the solid preparation, the dose of curcumin is 0.001 to 1000 mg/unit, or 0.1 to 100 mg/unit, or 10-20 mg/unit.
  39. 权利要求1-15、19-32和34-38任一项的药物递送***在制备用于抗氧化、抗炎、抗癌、诱导细胞凋亡、抗血管生成、神经保护、抗微生物、保肝护肾、抑制血管形成、预防心梗、降血糖、抗风湿的药物中的应用。 The drug delivery system according to any one of claims 1-15, 19-32 and 34-38 for use in the preparation of anti-oxidation, anti-inflammatory, anti-cancer, induced apoptosis, anti-angiogenesis, neuroprotection, anti-microbial, liver protection Application in medicine for protecting kidney, inhibiting blood vessel formation, preventing myocardial infarction, lowering blood sugar and resisting rheumatism.
PCT/CN2017/094665 2017-07-27 2017-07-27 Curcumin pharmaceutical preparations WO2019019091A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/CN2017/094665 WO2019019091A1 (en) 2017-07-27 2017-07-27 Curcumin pharmaceutical preparations

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2017/094665 WO2019019091A1 (en) 2017-07-27 2017-07-27 Curcumin pharmaceutical preparations

Publications (1)

Publication Number Publication Date
WO2019019091A1 true WO2019019091A1 (en) 2019-01-31

Family

ID=65039426

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/094665 WO2019019091A1 (en) 2017-07-27 2017-07-27 Curcumin pharmaceutical preparations

Country Status (1)

Country Link
WO (1) WO2019019091A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020181256A1 (en) * 2019-03-06 2020-09-10 Renibus Therapeutics, Inc. Novel tetrahydrocurcumin compositions, methods of making, and methods of using the same
CN111956713A (en) * 2020-09-03 2020-11-20 华熙生物科技股份有限公司 Hangover-alleviating and liver-protecting composition containing hyaluronic acid and application thereof
CN111995307A (en) * 2020-08-31 2020-11-27 湖北工业大学 Preparation method of protective coating material for marine peripheral buildings
CN113332250A (en) * 2021-07-15 2021-09-03 诺言医药科技(上海)有限公司 Glutaminase inhibitor freeze-dried powder and preparation method and application thereof
WO2023024185A1 (en) * 2021-08-24 2023-03-02 江苏万邦生化医药集团有限责任公司 Suspension tablet

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101627969A (en) * 2009-07-30 2010-01-20 浙江工业大学 Novel curcumin self-emulsifying drug delivery system and preparation method thereof
WO2010010431A1 (en) * 2008-07-24 2010-01-28 National Institute Of Pharmaceutical Education And Research Self-nano-emulsifying curcuminoids composition with enhanced bioavailability
CN101869692A (en) * 2010-06-30 2010-10-27 姜运华 Curcumin self-microemulsion and preparation method thereof
CN103237458A (en) * 2010-10-14 2013-08-07 雅培股份有限两合公司 Curcuminoid solid dispersion formulation
CN104997727A (en) * 2015-07-06 2015-10-28 山东省眼科研究所 Curcumin micelle nasal solution and preparation method thereof
CN107308133A (en) * 2016-04-27 2017-11-03 周意 Curcumin pharmaceutical preparation

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010010431A1 (en) * 2008-07-24 2010-01-28 National Institute Of Pharmaceutical Education And Research Self-nano-emulsifying curcuminoids composition with enhanced bioavailability
CN101627969A (en) * 2009-07-30 2010-01-20 浙江工业大学 Novel curcumin self-emulsifying drug delivery system and preparation method thereof
CN101869692A (en) * 2010-06-30 2010-10-27 姜运华 Curcumin self-microemulsion and preparation method thereof
CN103237458A (en) * 2010-10-14 2013-08-07 雅培股份有限两合公司 Curcuminoid solid dispersion formulation
CN104997727A (en) * 2015-07-06 2015-10-28 山东省眼科研究所 Curcumin micelle nasal solution and preparation method thereof
CN107308133A (en) * 2016-04-27 2017-11-03 周意 Curcumin pharmaceutical preparation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LIN, XIAO ET AL.: "Preparation of curcumin-loaded polymeric micelles and in vitro evaluation", CHINESE JOURNAL OF HOSPITAL PHARMACY, 28 February 2015 (2015-02-28), pages 213 - 216 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020181256A1 (en) * 2019-03-06 2020-09-10 Renibus Therapeutics, Inc. Novel tetrahydrocurcumin compositions, methods of making, and methods of using the same
CN111995307A (en) * 2020-08-31 2020-11-27 湖北工业大学 Preparation method of protective coating material for marine peripheral buildings
CN111956713A (en) * 2020-09-03 2020-11-20 华熙生物科技股份有限公司 Hangover-alleviating and liver-protecting composition containing hyaluronic acid and application thereof
CN111956713B (en) * 2020-09-03 2021-10-15 华熙生物科技股份有限公司 Hangover-alleviating and liver-protecting composition containing hyaluronic acid and application thereof
CN113332250A (en) * 2021-07-15 2021-09-03 诺言医药科技(上海)有限公司 Glutaminase inhibitor freeze-dried powder and preparation method and application thereof
CN113332250B (en) * 2021-07-15 2022-11-04 诺言医药科技(上海)有限公司 Glutaminase inhibitor freeze-dried powder as well as preparation method and application thereof
WO2023024185A1 (en) * 2021-08-24 2023-03-02 江苏万邦生化医药集团有限责任公司 Suspension tablet

Similar Documents

Publication Publication Date Title
WO2017157350A1 (en) Edaravone dosage form
WO2019019091A1 (en) Curcumin pharmaceutical preparations
Dudhipala et al. Improved anti-hyperlipidemic activity of Rosuvastatin Calcium via lipid nanoparticles: Pharmacokinetic and pharmacodynamic evaluation
Yang et al. Improved dissolution and bioavailability of silymarin delivered by a solid dispersion prepared using supercritical fluids
CN107308133A (en) Curcumin pharmaceutical preparation
EP3104844B1 (en) Complexes of sirolimus and its derivatives, process for the preparation thereof and pharmaceutical compositions containing them
Han et al. Honokiol nanosuspensions: preparation, increased oral bioavailability and dramatically enhanced biodistribution in the cardio-cerebro-vascular system
Zhang et al. Enhancement of oral bioavailability of tripterine through lipid nanospheres: preparation, characterization, and absorption evaluation
Kumar et al. Guargum and Eudragit® coated curcumin liquid solid tablets for colon specific drug delivery
KR102055542B1 (en) Solid dispersion
Bhalekar et al. Fabrication and efficacy evaluation of chloroquine nanoparticles in CFA-induced arthritic rats using TNF-α ELISA
CN104507902A (en) Anthraquinone analogs and methods of making and using thereofanthraquinone analogs and methods of making and using thereof
JP2013505233A (en) Pharmaceutical composition for oral administration of 3,3'-diindolylmethane
US20150132388A1 (en) Complexes of fulvestrant and its derivatives, process for the preparation thereof and pharmaceutical compositions containing them
Elnaggar et al. Self-emulsifying preconcentrates of daidzein–phospholipid complex: design, in vitro and in vivo appraisal
Ding et al. Integration of phospholipid-drug complex into self-nanoemulsifying drug delivery system to facilitate oral delivery of paclitaxel
Liu et al. Enhanced dissolution and oral bioavailability of tanshinone IIA base by solid dispersion system with low-molecular-weight chitosan
Kansom et al. Fabrication and characterization of andrographolide analogue (3A. 1) nanosuspensions stabilized by amphiphilic chitosan derivatives for colorectal cancer therapy
El Said et al. Vilazodone-phospholipid mixed micelles for enhancing oral bioavailability and reducing pharmacokinetic variability between fed and fasted states
CN108601759A (en) A kind of taxol drug composition and its pharmaceutical preparation, preparation method and purposes
US20210260050A1 (en) Amorphous solid dispersion comprising 6-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)nicotinamide
Althobaiti et al. Formulation Development of Curcumin-piperine solid dispersion via hot-melt extrusion
Nguyen et al. Preparation of an oil suspension containing ondansetron hydrochloride as a sustained release parenteral formulation
CN105012234B (en) A kind of 1,7-bis(3,4-dimethoxyphenyl)-1,6-heptadiene-3,5-dione polymer micelle and preparation method thereof and medical usage
Yusuf et al. Formulation design and cell cytotoxicity of curcumin-loaded liposomal solid gels for anti-Hepatitis C virus

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: 17919135

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17919135

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17919135

Country of ref document: EP

Kind code of ref document: A1

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 05/08/2020)

122 Ep: pct application non-entry in european phase

Ref document number: 17919135

Country of ref document: EP

Kind code of ref document: A1