CN116688137A - Pharmaceutical composition based on core-shell structure and application - Google Patents

Pharmaceutical composition based on core-shell structure and application Download PDF

Info

Publication number
CN116688137A
CN116688137A CN202310902318.8A CN202310902318A CN116688137A CN 116688137 A CN116688137 A CN 116688137A CN 202310902318 A CN202310902318 A CN 202310902318A CN 116688137 A CN116688137 A CN 116688137A
Authority
CN
China
Prior art keywords
drug
dspe
peg
acid
plga
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202310902318.8A
Other languages
Chinese (zh)
Other versions
CN116688137B (en
Inventor
李默晗
黄宇光
季天骄
马超
贺渝淼
裴丽坚
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Peking Union Medical College Hospital Chinese Academy of Medical Sciences
Original Assignee
Peking Union Medical College Hospital Chinese Academy of Medical Sciences
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 Peking Union Medical College Hospital Chinese Academy of Medical Sciences filed Critical Peking Union Medical College Hospital Chinese Academy of Medical Sciences
Priority to CN202310902318.8A priority Critical patent/CN116688137B/en
Publication of CN116688137A publication Critical patent/CN116688137A/en
Application granted granted Critical
Publication of CN116688137B publication Critical patent/CN116688137B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular 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
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/28Steroids, e.g. cholesterol, bile acids or glycyrrhetinic acid
    • 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
    • A61K9/48Preparations 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
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • A61K9/5153Polyesters, e.g. poly(lactide-co-glycolide)
    • 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]
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Nanotechnology (AREA)
  • Molecular Biology (AREA)
  • Dispersion Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Inorganic Chemistry (AREA)
  • Optics & Photonics (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Preparation (AREA)

Abstract

The invention discloses a pharmaceutical composition based on a core-shell structure and application thereof. The sustained-release medicine prepared by the invention accords with the concept of clinical multi-mode analgesia, and the medicines with different mechanisms are used for combined analgesia, so that the analgesic effect can be improved, the analgesic time can be prolonged, the meloxicam can be released preferentially in the first 48 hours to relieve the pain caused by acute inflammation, and the bupivacaine can be released continuously for more than 7 days, so that the effective postoperative analgesia can be realized. In addition, the nano particles with the core-shell structure show good biological safety in vitro and in vivo, provide a concise strategy for postoperative multi-mode analgesia management, and have wide application prospects.

Description

Pharmaceutical composition based on core-shell structure and application
Technical Field
The invention belongs to the field of biological medicine, and in particular relates to a pharmaceutical composition based on a core-shell structure and application thereof.
Background
Pain is an unpleasant sensory and emotional experience caused by tissue damage or potential tissue damage. Acute pain and chronic pain can be classified according to duration. Pain tends to cause people to experience mood such as depression and anxiety, and the quality of life is reduced. Through years of research and development, the generation of pain is more deeply understood, and a plurality of pain treatment methods are applied to clinic, and pain treatment medicines are also greatly developed. However, due to the complexity of the pathogenesis of pain, difficulties are presented to the diagnosis and treatment of pain. The world pain will indicate in 2005 that "the world is still painful". "thus developing an analgesic that is safer, more effective, and more convenient to use remains a formidable task.
Over 60% of patients undergoing surgery experience postoperative pain, with 10% -50% of patients experiencing persistent pain, which places a significant economic burden on the social and public health systems. Effective perioperative pain management is a prerequisite for post-operative accelerated rehabilitation surgery (erat), which has been demonstrated to reduce incidence of perioperative complications and enhance postoperative functional recovery, thereby shortening hospitalization. Although the importance of perioperative pain management is well recognized, the treatment of postoperative pain remains a significant challenge.
The mechanism of postoperative pain involves a number of factors, complex inflammation and spontaneous electrical activity (SA) caused by peripheral tissue injury, resulting in postoperative pain. Previous studies have shown that SA begins to appear 1 day post-surgery, rather than immediately after surgery, and gradually decreases over time. While inflammatory factor expression begins to rise as early as 1 hour post-surgery and continues to be upregulated in skin and muscle for up to 2 days post-surgery. Most of the existing slow-release analgesic drugs only deliver one analgesic drug, and the analgesic effect is difficult to further improve due to the limited slow-release time, if the slow-release analgesic drugs can be developed to simultaneously carry two different types of analgesic drugs and realize sequential administration according to the pain occurrence mechanism, the slow-release analgesic can meet the principle of multi-mode analgesia while realizing long-acting analgesia, and an effective and convenient administration scheme is provided for postoperative pain management.
Disclosure of Invention
In order to make up the defects of the prior art, the invention provides a pharmaceutical composition based on a core-shell structure and application thereof.
In order to achieve the above purpose, the invention adopts the following technical scheme:
a first aspect of the present invention provides a drug delivery vehicle/system comprising liposomes loaded with a first drug and PLGA nanoparticles loaded with a second drug.
Further, the liposome comprises a phospholipid and a sterol.
Further, the phospholipid comprises DOPC, DSPE, DSPE-PEG 2000 、DSPE-PEG 3000 、DSPE-PEG 5000 HSPC, DSPC and/or DPPC.
Further, the phospholipid comprises DOPC, DSPE-PEG 2000 、DSPE-PEG 3000 And/or DSPE-PEG 5000
Further, the phospholipids include DOPC and DSPE-PEG 2000
Further, the molecular weight of the PLGA is 24,000-38,000Da.
Further, the molar ratio of LA to GA in PLGA was 50:50.
further, the sterol is cholesterol.
Further, DOPC, DSPE-PEG 2000 And cholesterol in a mass ratio of 15:1:5.
In a second aspect the present invention provides a sustained release pharmaceutical composition comprising a first drug, a second drug and a drug sustained release carrier/system according to the first aspect of the present invention.
Further, the first drug is a non-steroidal anti-inflammatory drug.
Further, the non-steroidal anti-inflammatory drugs include salicylic acid derivatives, indole and indenacetic acid, fenamate, heteroaryl acetic acid, aryl acetic acid and propionic acid derivatives, enolic acid, para-aminophenol derivatives, alkanones, nimesulide and/or praline Luo Kuizong, or pharmaceutically acceptable salts of these ingredients.
Further, the non-steroidal anti-inflammatory drug is selected from alkenols.
Further, the enolic acid includes oxicam derivatives and pyrazolone derivatives.
Further, the enolic acid is selected from oxicam derivatives.
Further, the oxicam derivatives include ampiroxicam, cinnoxicam, drooxicam, lornoxicam, meloxicam, piroxicam, sudoxicam, tenoxicam.
Further, the oxicam derivative is meloxicam.
Further, the second drug comprises a local anesthetic.
Further, the local anesthetics include bupivacaine, levobupivacaine, tetracaine, ropivacaine, lidocaine, prilocaine, mepivacaine, procaine, chloroprocaine, procaine, hexetidine, cyclomecaine, butoxyprocaine, bupivacaine, procaine, cocaine, finacaine, dibucaine, faricaine, dyclonine, pramoxine, and/or dimethylisoquinoline, or pharmaceutically acceptable salts of these ingredients.
Further, the local anesthetic is bupivacaine.
Further, the mass ratio of bupivacaine to PLGA was 1:4.
In a third aspect, the present invention provides a method of preparing a sustained release pharmaceutical composition, the method comprising:
(1) Mixing PLGA loaded with a second drug into liposomes loaded with a first drug;
(2) Hydrating the lipid film;
(3) Extruding the colloidal solution in the liposome.
Further, step (2) hydrates the lipid film by water bath sonication.
Further, step (3) extrudes the colloidal solution in the liposome through a liposome extruder.
Further, the preparation method of the liposome loaded with the first drug comprises the following steps: mixing the first medicine, DOPC, DSPE-PEG 2000 And cholesterol was dissolved in chloroform/methanol solution.
Further, the method also includes removing the organic solvent and forming a dry film.
Further, the volume ratio of the chloroform/methanol solution is 9:1.
Further, the organic solvent was removed using a rotary evaporator.
Further, the temperature for removing the organic solvent was 30 ℃.
Further, the preparation method of the PLGA loaded with the second drug comprises the following steps: and dripping the acetone containing the second drug and PLGA into the polyvinyl alcohol aqueous solution to obtain a mixture.
Further, the method includes stirring the mixture.
Further, the speed of stirring the mixture is 100-2000rpm.
Further, the speed of stirring the mixture is 500-1000rpm.
Further, the speed at which the mixture was stirred was 800rpm.
Further, the stirring speed was 800rpm for 1 to 10 hours.
Further, the stirring speed was 800rpm for 3 to 5 hours.
Further, the stirring speed was 800rpm for 4 hours.
Further, the step of stirring the mixture further comprises the step of increasing the stirring speed.
Further, the increase in stirring speed includes a gradual increase in speed from 0 to 2000rpm.
Further, the increase in stirring speed includes a gradual increase in speed from 0 to 1000rpm.
Further, the stirring speed was increased by gradually increasing the speed from 100rpm to 800rpm.
Further, the stirring speed was gradually increased from 100rpm to 800rpm using 1 to 30 minutes.
Further, the stirring speed was gradually increased from 100rpm to 800rpm using 5 to 20 minutes.
Further, the stirring speed was gradually increased from 100rpm to 800rpm over 10 minutes.
Further, the method includes removing the organic solvent.
Further, the organic solvent was removed using a rotary evaporator.
Further, the method includes removing the free second drug.
Further, the free second drug is removed by repeated washing.
In a fourth aspect, the invention provides the use of a drug delivery vehicle/system according to the first aspect of the invention for the preparation of a drug delivery vehicle having a plurality of active ingredients.
Further, the medicament also comprises a pharmaceutically acceptable carrier.
In a fifth aspect, the invention provides the use of a sustained release pharmaceutical composition according to the second aspect of the invention in the manufacture of a medicament for the treatment of a disease.
Further, the disease is selected from pain and inflammation.
Further, the pain includes post-operative analgesia.
Further, the postoperative analgesia is selected from postoperative acute pain.
Further, the medicament also comprises a pharmaceutically acceptable carrier.
In a sixth aspect, the invention provides the use of a method according to the third aspect of the invention for the preparation of a sustained release medicament having a plurality of active ingredients.
Further, the medicament also comprises a pharmaceutically acceptable carrier.
The invention has the advantages and beneficial effects that:
the sustained-release medicine prepared by the invention accords with the concept of clinical multi-mode analgesia, and the medicines with different mechanisms are used for combined analgesia, so that the analgesic effect can be improved, the analgesic time can be prolonged, the preferential release of meloxicam in the first 48 hours can be realized, and the bupivacaine can be continuously released for about 7 days. In addition, the nano particles with the core-shell structure show good biological safety in vitro and in vivo, provide a convenient administration strategy for postoperative multi-mode analgesia management, and have wide application prospects.
Drawings
FIG. 1 is a schematic representation of the release profile and analgesic mechanism of NPB@LPM for postoperative pain;
FIG. 2 is a schematic diagram of the synthesis and characterization of an NPB@LPM core-shell nanosystem, wherein 2A is a schematic diagram of the preparation process of the NPB@LPM, 2B is a transmission electron microscope image of NP, 2C is a transmission electron microscope image of NPB, 2D is a transmission electron microscope image of the NPB@LPM, 2E is a histogram of particle size and potential characterization of NP, NPB and NPB@LPM, 2F is a particle size stability evaluation image of the NPB and NPB@LPM using DLS, 2G is a potential stability evaluation image of the NPB and NPB@LPM using DLS, and 2H is a PDI stability evaluation image of the NPB and NPB@LPM using DLS;
FIG. 3 is a graph showing the drug release of BUP and MLX, wherein 3A is a graph showing the dispersibility of the drug in water and the effect of tyndall, 3B is a graph showing the drug release of MLX in seven days, and 3C is a graph showing the drug release of BUP in seven days;
FIG. 4 is a graph of cytotoxicity of BUP and NPB@LPM at various concentrations, wherein 4A is a graph of cell viability after 24h incubation of BUP and NPB@LPM in PC12 cells and 4B is a graph of cell viability after 24h incubation of BUP and NPB@LPM in C2C12 cells;
FIG. 5 is a graph of a study of pain behavioural in rats, wherein 5A is a schematic diagram of a pain behavioural test, 5B is a graph of PWT in rats, 5C is a graph of PWT AUC in rats, 5D is a graph of PWL in rats, and 5E is a graph of PWL AUC in rats;
FIG. 6 is a graph of tissue response and toxicity assessment;
FIG. 7 is a graph showing the concentration of inflammatory factors, wherein 7A is a graph showing the PGE2 level of the post-operation 24h plantar incision tissue, 7B is a graph showing the IL-1β level of the post-operation 24h plantar incision tissue, and 7C is a graph showing the TNF- α level of the post-operation 24h plantar incision tissue;
fig. 8 is a histological evaluation chart of important organs.
Detailed Description
The following provides definitions of some of the terms used in this specification. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
The invention provides a drug delivery vehicle/system comprising liposomes loaded with a first drug and PLGA loaded with a second drug.
In the present invention, liposomes refer to particles having an aqueous interior space that is isolated from an external medium by one or more membranes forming a bilayer membrane of vesicles. Bilayer membranes of liposomes are typically formed from one or more lipids, i.e., amphiphilic molecules of synthetic or natural origin comprising a spatially separated hydrophobic domain and hydrophilic domain. In general, liposomes comprise a lipid mixture, which typically includes one or more lipids selected from the group consisting of: a double fatty chain lipid (dialiphatic chain lipid), such as a phospholipid, diglycerol ester, a di-fatty glycolipid (dialiphatic glycolipid); single lipids such as sphingomyelin (sphingomyelin) and glycosphingolipids (lysosphingolipids); sterols such as cholesterol and derivatives thereof; and combinations thereof.
In an embodiment of the invention, the liposomes include a double fatty chain lipid and a sterol.
In a preferred embodiment of the invention, the liposomes comprise phospholipids and sterols.
Wherein the phospholipids include, but are not limited to, PC (phosphatidyl choline ), PG (phosphatidyl glycerol, phosphatidylglycerol), PE (phosphatidyl ethanolamine ), PS (phosphatidyl serine, phosphatidylserine), PA (phosphatidic acid ), PI (phosphatidyl inositol, phosphatidylinositol), EPC (egg phosphatidyl choline, phosphatidylcholine), EPG (egg phosphatidyl glycerol, phosphatidylglycerol), EPE (egg phosphatidyl ethanolamine, phosphatidylethanolamine), EPS (egg phosphatidyl serine, phosphatidylserine), EPA (egg phosphatidic acid, phosphatidic acid), EPI (egg phosphatidyl inositol, phosphatidylinositol), SPC (so phosphoha)tidyl choline, soyabean phosphatidylcholine), SPG (soy phosphatidyl glycerol, soyabean phosphatidylglycerol), SPE (soy phosphatidyl ethanolamine, soyabean phosphatidylethanolamine), SPS (soy phosphatidyl serine, soyabean phosphatidylserine), SPA (soy phosphatidic acid, soyabean phosphatidic acid), SPI (soy phosphatidyl inositol, soyabean phosphatidylinositol), DPPC (dipalmitoyl phosphatidyl choline, diperoxy phosphatidylcholine), DOPC (1, 2-dioleoyl-sn-glycero-3-phosphotidyline, 1, 2-dioleoyl-sn-glycero-3-phosphatidylcholine), DMPC (dimyristoylphos phatidyl choline ), DPPG (dipalmitoylphos phatidyl glycerol, diperoxy phosphatidylglycerol), DOPG (dioleoyl phos phatidyl glycerol ), DMPG (dimyristoylphos phatidyl glycerol, diperoxy phosphatidylglycerol), HEPC (hepidecylphosphatidylcholine), hexadecylphosphoric acid), HSPC (hydrogenated soy phosphatidyl choline, hydrogenated soyabean phosphatidylglycerol), stearoyl phosphatidylcholine (DSPC-distearoylphos phatidyl choline, stearoyl phosphatidylcholine), stearoyl phosphatidylcholine (dspe, stearoyl phosphatidylethanolamine), stearoyl phosphatidylcholine (37, stearoyl phosphatidylcholine), dpp (37, stearoyl phosphatidylcholine, and (37, 463, stearoyl phosphatidylcholine) POPC (1-palmitoyl-2-oleyl-sn-glycero-3-phosphatidyl choline, 1-soft acyl-2-oleoyl-sn-glycero-3-phosphatidylcholine); DSPE (distearoylphos phatidylethanolamine ), DSPE-PEG 2000 (1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -2000])、DSPE-PEG 3000 (1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -3000])、DSPE-PEG 5000 (1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -5000]) DPPS (dipalmitoyl phosphatidyl serine, disoft acyl phosphatidylserine), DOPS (1, 2-d)ioleoyl-sn-glycero-3-phosphatidyl serine,1, 2-dioleoyl-sn-glycero-3-phosphatidylserine), DMPS (dimyristoyl phosphatidyl serine ), DSPS (distearoyl phosphatidyl serine, distearoyl phosphatidylserine), DPPA (dipalmitoyl phosphatidic acid ), DOPA (1, 2-dioleoyl-sn-glycero-3-phosphatidic acid,1, 2-dioleoyl-sn-glycero-3-phosphatidic acid), DMPA (dimyristoyl phosphatidic acid ); DSPA (distearoyl phosphatidic acid, distearylphospholic acid), DMPI (dimyristoyl phosphatidyl inositol, dimyristoylphospholitides), DSPI (distearoyl phosphatidyl inositol, distearylphospholitides) and mixtures thereof.
In a preferred embodiment of the present invention, the phospholipid comprises DOPC (1, 2-dioleoyl-sn-glycero-3-phosphotidyline, 1, 2-dioleoyl-sn-glycero-3-phosphatidylcholine), DSPE (distearoylphos phatidylethanolamine ), DSPE-PEG 2000 (1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -2000])、DSPE-PEG 3000 (1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -3000])、DSPE-PEG 5000 (1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -5000]) HSPC (hydrogenated soy phosphatidyl choline ), DSPC (distearoylphos phatidyl choline, distearoyl phosphatidylcholine) and/or DPPC (dipalmitoyl phosphatidyl choline ).
In a more preferred embodiment of the present invention, the phospholipid comprises DOPC (1, 2-dioleoyl-sn-glycero-glycero-3-phosphotidyline, 1, 2-dioleoyl-sn-glycero-3-phosphatidylcholine), DSPE-PEG 2000 (1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -2000])、DSPE-PEG 3000 (1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -3000 ]) And/or DSPE-PEG 5000 (1, 2-distearoyl-sn-glycero-3-phosphate)ethanolamine-N- [ methoxy (polyethylene glycol) -5000])。
In a specific embodiment of the present invention, the phospholipid comprises DOPC (1, 2-dioleoyl-sn-glycero-glycero-3-phosphotidyline, 1, 2-dioleoyl-sn-glycero-3-phosphatidylcholine) and DSPE-PEG 2000 (1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -2000])。
In the present invention, PLGA refers to a polymer comprising lactide and glycolide. PLGA is a linear copolymer that can be prepared according to methods well known to the skilled person in different proportions between its constituent monomers Lactic Acid (LA) and Glycolic Acid (GA). Wherein, the molar ratio of LA to GA in PLGA can be 90:10, 80:20, 75:25, 60:40 and 50:50, and a ratio between any of the above molar ratios.
In a specific embodiment of the invention, the molar ratio of LA to GA in PLGA is 50:50.
sustained release, controlled release, extended release, sustained release, delayed release or slow release are used interchangeably in the present invention, sustained release being well known in the art of pharmaceutical sciences and are used by the present invention to mean controlled release of an active compound or active agent from a dosage form to the environment over an extended period of time (throughout or during), for example greater than or equal to 1 hour. The sustained release dosage form will release the drug at a substantially constant rate over an extended period of time or will release a substantially constant amount of the drug incrementally over an extended period of time.
The invention provides a sustained release pharmaceutical composition comprising a first drug, a second drug and a drug sustained release carrier/system as described above.
The first drug is a non-steroidal anti-inflammatory drug.
In the present invention, non-steroidal anti-inflammatory drugs (NASIDs), non-steroidal anti-inflammatory drugs, non-steroidal anti-inflammatory agents and grammatical variations thereof may be used interchangeably, including but not limited to salicylic acid derivatives (e.g., salicylic acid, acetylsalicylic acid, methyl salicylate, diflunisal, oxasalazine, bis-salicylates, sulfasalazine), indole and indene acetic acids (e.g., indomethacin, etodolac, sulindac), fenamates (e.g., meclofenamic acid, mefenamic acid, flufenamic acid, niflumic acid, and tolfenamic acid), heteroaryl acetic acids (e.g., acemetacin, alclofenac, cyclocloindenic acid, diclofenac, fenamic acid, furofenofenac, ibufenac, isoxaglic, thioplac, tolmetin, zidometacin, aryl acetic acid and propionic acid derivatives (e.g., alaminoprofen, benzoxprofen, bucc acid, carprofen, fenbufen, fenoprofen, flurbiprofen, ibuprofen, indoprofen, ketoprofen, imiprofen, naproxen sodium, oxaprozin, pirprofen, pranoprofen, suprofen, tiaprofenic acid, thiooxaprofen), enolic acids (e.g., the oxicam derivatives ampoxicam, cinnoxicam, drooxicam, lornoxicam, meloxicam, piroxicam, suldoxoxicam and tenoxicam, and pyrazolone derivatives aminopyrine, antipyrine, azaprofen, analgin, oxybbuprofen, phenylbutazone), p-aminophenol derivatives (e.g., acetaminophen), alkanones (e.g., nabumetone), nimesulide and/or praziram Luo Kuizong, or pharmaceutically acceptable salts of these ingredients.
In an embodiment of the invention, the non-steroidal anti-inflammatory drug is selected from alkenols.
In a preferred embodiment of the invention, the enolic acid is selected from oxicam derivatives.
In a specific embodiment of the invention, the oxicam derivative is meloxicam.
The second drug comprises a local anesthetic.
In the present invention, local anesthetics, or local anesthetics are used interchangeably and refer to agents that provide relief from local pain, including, but not limited to, bupivacaine, levobupivacaine, tetracaine, ropivacaine, lidocaine, prilocaine, mepivacaine, procaine, chloroprocaine, propoxycaine, hexucaine, cyclomecaine, butoxyprocaine, bupivacaine, procaine, cocaine, finacaine, dibucaine, farivacaine, dyclonine, pramoxine, and/or dimethylisoquinoline, or pharmaceutically acceptable salts of these ingredients.
In a specific embodiment of the invention, the local anesthetic is bupivacaine.
In the present invention, a pharmaceutically acceptable salt is any non-toxic salt that is capable of providing the drug or prodrug of the present invention directly or indirectly after administration to a recipient. Including acid salts or salts of basic nitrogen atoms of purine compounds. Exemplary salts include, but are not limited to, sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisate, fumarate, gluconate, sucrose, formate, benzoate, glutamate, mesylate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate. The pharmaceutically acceptable salt may also be camphorsulfonate. Pharmaceutically acceptable salts also refer to salts and bases of purine compounds having acid functionality, such as carboxylic acid functionality. Suitable salts include, but are not limited to, hydroxides of alkali metals such as sodium, potassium and lithium; hydroxides of alkaline earth metals such as calcium and magnesium; hydroxides of other metals such as aluminum and zinc; ammonia and organic amines such as unsubstituted or hydroxy-substituted mono-, di-or tri-alkylamines, dicyclohexylamine; tributylamine; pyridine; n-methylamine, N-ethylamine; diethylamine; triethylamine; mono-, di-or tri- (2-OH-lower alkylamines), such as mono-, di-or tri- (2-hydroxyethyl) amine, 2-hydroxy tert-butylamine or tris- (hydroxymethyl) methylamine, N-di-lower alkyl-N- (hydroxy-lower alkyl) -amines, such as N, N dimethyl-N- (2-hydroxyethyl) amine or tri- (2-hydroxyethyl) amine; N-methyl-D-glucamine; and amino acids such as arginine, lysine, and the like. Pharmaceutically acceptable salts also include hydrates of purine compounds.
The invention provides application of the slow-release pharmaceutical composition in preparing medicines for treating diseases.
The medicament also comprises a pharmaceutically acceptable carrier.
In the present invention, a pharmaceutically acceptable carrier means a non-toxic material that does not interfere with the effect of the drug of the present invention or the biological activity of the drug of the present invention. Formulating pharmaceutically active ingredients with pharmaceutically acceptable carriers is known in the art, for example Remington: science and practice of pharmacy (The Science and Practice of Pharmacy) (e.g. 21 st edition (2005), and any later versions). Non-limiting examples of pharmaceutically acceptable carriers include: salts (e.g., acid/anion salts, base/cation salts), excipients, buffers, diluents, solubilizers, tonicity adjusting agents, surfactants, preservatives, isotonicity agents, flavoring agents, colorants, stabilizers and chelating agents.
In one embodiment of the invention, the pharmaceutically acceptable carrier comprises an acid salt/anion salt. Non-limiting examples of acid salts/anion salts include, but are not limited to, acetates, benzenesulfonates, benzoates, bicarbonates, bitartrate, bromides, calcium edetate, camphorsulfonates, carbonates, chlorides, citrates, dihydrochloride, edetate, ethanedisulfonate, etoates, ethanesulfonates, fumarates, glucoheptonates, gluconates, glutamates, acetamidophenylarsonates, hexylresorcinol salts, hydrabamines, hydrobromides, hydrochlorides, hydroxynaphthoates, iodides, isethionates, lactates, lactonates, malates, maleates, mandelates, methanesulfonates, methyl bromides, methyl nitrates, methyl sulfates, mucinates, naphthalenesulfonates, nitrates, pamonates, pantothenates, phosphates/bisphosphates, polygalactonates, salicylates, stearates, hypoacetates, succinates, sulfates, tannates, tartrates, theaters, toluene sulfonates, and triethyliodides.
In one embodiment of the invention, the pharmaceutically acceptable carrier comprises an alkali salt/cation salt. Non-limiting examples of base salts/cation salts include, but are not limited to, aluminum, 2-amino-2-hydroxymethyl-propane-1, 3-diol (also known as TRIS (hydroxymethyl) aminomethane, tromethamine, or TRIS), ammonia, benzathine, t-butylamine, chloroprocaine, choline, cyclohexylamine, diethanolamine, ethylenediamine, lithium, L-lysine, magnesium, meglumine, N-methyl-D-glucamine, piperidine, potassium, procaine, quinine, sodium, triethanolamine, or zinc.
In one embodiment of the invention, the pharmaceutically acceptable carrier comprises a buffer. Non-limiting examples of buffering agents include, but are not limited to, arginine, aspartic acid, dihydroxyethyl glycine, citrate, disodium hydrogen phosphate, fumaric acid, glycine, glycylglycine, histidine, lysine, maleic acid, malic acid, sodium acetate, sodium carbonate, sodium dihydrogen phosphate, sodium phosphate, succinate, tartaric acid, triazine, and tris (hydroxymethyl) aminomethane, and mixtures thereof.
In one embodiment of the invention, the pharmaceutically acceptable carrier comprises a preservative. Non-limiting examples of preservatives include, but are not limited to, benzethonium chloride, benzoic acid, benzyl alcohol, bromonitropropylene glycol, butyl 4-hydroxybenzoate, chlorobutanol, chlorocresol, chlorohexidine, chlorobenzeneglycerol ether, o-cresol, m-cresol, p-cresol, ethyl 4-hydroxybenzoate, imidurea, methyl 4-hydroxybenzoate, phenol, 2-phenoxyethanol, 2-phenylethanol, propyl 4-hydroxybenzoate, sodium dehydroacetate, thimerosal, and mixtures thereof.
In one embodiment of the invention, the pharmaceutically acceptable carrier comprises an isotonic agent. Non-limiting examples of isotonic agents include, but are not limited to, amino acids (such as glycine, histidine, arginine, lysine, isoleucine, aspartic acid, tryptophan, and threonine), sugar alcohols (such as glycerol, 1, 2-propanediol, propylene glycol), 1, 3-propanediol, and 1, 3-butanediol), polyethylene glycols (e.g., PEG 400), and mixtures thereof. Another example of an isotonic agent includes a sugar. Non-limiting examples of sugars may be mono-, di-, or polysaccharides, or water-soluble glucans including, for example, fructose, glucose, mannose, sorbose, xylose, maltose, lactose, sucrose, trehalose, dextran, pullulan, dextrin, cyclodextrin, alpha and beta-HPCD, soluble starch, hydroxyethyl starch, and sodium carboxymethyl cellulose. Another example of an isotonic agent is a sugar alcohol, wherein the term "sugar alcohol" is defined as a C (4-8) hydrocarbon having at least one-OH group. Non-limiting examples of sugar alcohols include mannitol, sorbitol, inositol, galactitol, hexitol, xylitol, and arabitol. Medicaments comprising each of the isotonic agents listed in this paragraph constitute an alternative embodiment of the invention.
In one embodiment of the invention, the pharmaceutically acceptable carrier comprises a chelating agent. Non-limiting examples of chelating agents include, but are not limited to, salts of citric acid, aspartic acid, ethylenediamine tetraacetic acid (EDTA), and mixtures thereof.
In one embodiment of the invention, the pharmaceutically acceptable carrier comprises a stabilizer. Non-limiting examples of stabilizers include carboxy-/hydroxy-cellulose and its derivatives (such as HPC, HPC-SL, HPC-L and HPMC), cyclodextrin, 2-methylthioethanol, polyethylene glycol (such as PEG 3350), polyvinyl alcohol (PVA), polyvinylpyrrolidone, salts (such as sodium chloride), sulfur-containing substances such as thioglycerol or thioglycollic acid.
In one embodiment of the invention, the pharmaceutically acceptable carrier comprises a lubricant. Lubricants can be used to prevent sticking of the dosage form to the roll, die and punch surfaces and to reduce inter-particle friction. Lubricants may also facilitate ejection of the dosage form from the mold cavity and improve the pelletization flow rate during processing. Examples of suitable lubricants include, but are not limited to, magnesium stearate, glyceryl behenate, calcium stearate, zinc stearate, stearic acid, silicon dioxide, talc, polyethylene glycol, mineral oil, carnauba wax, palmitic acid, sodium stearyl fumarate, sodium lauryl sulfate, glyceryl palmitostearate, myristic acid and hydrogenated vegetable oils and fats, and other known lubricants, and/or mixtures of two or more thereof. In one embodiment, the lubricant for the granulation of the feedstock, if present, is magnesium stearate.
In one embodiment of the invention, the pharmaceutically acceptable carrier comprises one or more surfactants, preferably one surfactant, at least one surfactant or two different surfactants. The term surfactant refers to any molecule or ion consisting of a water-soluble moiety (hydrophilic) and a fat-soluble and moiety (lipophilic). For example, the surfactant is selected from: anionic surfactants, cationic surfactants, nonionic surfactants, and/or zwitterionic surfactants.
The medicaments of the present invention may be formulated in liquid or solid form for oral, rectal, intranasal, topical (including buccal and sublingual), transdermal, vaginal or parenteral (including intramuscular, subcutaneous and intravenous) administration or in a form suitable for administration by inhalation or insufflation.
The invention is further illustrated below in connection with specific embodiments. It should be understood that the particular embodiments described herein are presented by way of example and not limitation. The principal features of the invention may be used in various embodiments without departing from the scope of the invention.
Examples
1 materials and methods
1.1 Experimental materials
Bupivacaine (bupivacaine) and bupivacaine hydrochloride (bupivacaine HCl) were purchased from shanghai microphone Lin Shenghua, inc; meloxicam (MLX) was purchased from alas Ding Gongsi; PLGA (molecular weight (Mw))=24,000-38,000 da, la: ga=50: 50 Purchased from Sigma;1, 2-Dioleoyl-sn-propyltri-3-phosphorylcholine (DOPC), 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [ methoxy (polyethylene glycol) -2000]
(DSPE-PEG 2000 ) Purchased from sierra xi biotechnology limited; cholesterol was purchased from beijing ku-da-tikoku-da-tsu.
1.2 cells and animals
PC12 cells and C2C12 cells were purchased from a national laboratory cell resource sharing platform.
Animal protocols were approved by Beijing co-ordination Hospital (XHDW-2022-045). Male Sprague-Dawley rats (200-250 g) used for the experiments were purchased from Fukang Biotechnology Co., ltd. Rats were kept in transparent plastic cages every 3 cages for 12h light and dark periods at 23+ -1deg.C. Rats are free to obtain food and water.
1.3 Preparation of NPs
2mL of acetone containing 20mg of PLGA was gently added dropwise to 6mL of a 1% aqueous solution of polyvinyl alcohol. During the first 10 minutes, the stirring speed was gradually increased from 100 to 800rpm, and then maintained at 800rpm for 4 hours. The organic solvent was removed under reduced pressure using a rotary evaporator and PLGA Nanoparticles (NPs) were collected by centrifugation at 15,000g for 15 min. Finally, the NPs were resuspended in 1mL of physiological saline.
Preparation of BUP-loaded nanoparticles: 5mg BUP was added to 20mg PLGA acetone, and precipitated by dropwise addition to 6mL of a 1% polyvinyl alcohol solution. The solvent was evaporated and collected as described above, and the free BUP was removed by repeated washing steps to obtain BUP-supported Nanoparticles (NPB).
For PLGA nanoparticles loaded with MLX or BUP+MLX, the synthesis method was the same as above, using 2mg of MLX and 20mg of PLGA, MLX was removed by pre-centrifugation (centrifugation at 1000rpm for 3 min).
1.4 Synthesis of core-Shell nanosystems
Preparation of blank liposome membrane: first, 7.5mg DOPC, 2.5mg cholesterol and 0.5mg DSPE-PEG were added 2000 Is dissolved in 9:1 (v/v) chloroform/methanol solution, and removing the organic solvent at low pressure in a rotary evaporator at 30 ℃ to form a dry film, thereby obtaining a blank liposome membrane (LP).
Preparation of MLX-loaded liposome membranes: first, 7.5mg DOPC, 2.5mg cholesterol, 0.5mg DSPE-PEG 2000 Is dissolved in 9:1 (v/v) chloroform/methanol solution, removing the organic solvent at low pressure in a rotary evaporator at 30℃to form a dry film, and obtaining an MLX-loaded liposome film (LPM).
PLGA NP solution was added to the flask and the lipid film was hydrated by water bath sonication (100W) for 20min. Finally, extruding the colloid solution through a liposome extruder (Avanti Polar Lipids, U.S.) with filter membranes with different pore diameters to obtain the NPB@LPM nano system. NP@LPM and NPB@LP were also prepared.
1.5 qualitative analysis of nanoparticles
The hydrated particle size, zeta potential and polydispersity index (PDI) of the repetitive nanoparticles (n=3) were determined with a laser particle sizer (NanoZS, malvern instruments ltd, uk) Dynamic Light Scattering (DLS) at 20 ℃. The nanoparticles were suspended in physiological saline at 4 ℃ and the particle size, PDI and Zeta potential of the nanoparticles were measured with DLS at 0, 1, 2, 3 and 7d to determine the stability of the nanoparticles.
The nanometer morphology is characterized by adopting a Transmission Electron Microscope (TEM), 10 mu L of sample (1 mg/mL) is dripped on a copper mesh carbon support film, and the sample is naturally dried in the environment. Samples were negative stained with 1% uranium acetate for 10min and examined by TEM (HT 7700, hitachi, japan).
The sample was freeze-dried for 24h and weighed to give a mass (n=3). The sample was dissolved with 1mL of acetone and sonicated for 10min. The solution was then centrifuged at 15000g for 15min, and the supernatant was filtered through a 0.22 μm filter and sampled. The concentrations of MLX and BUP were measured by using an Inertsil ODS-3 column (4.6 mm,250mm,5 μm particle size, shimadzu corporation) high performance liquid chromatograph (HPLC, LC-20AT, shimadzu corporation).
1.6 in vitro drug Release
The amount of BUP or MLX released was measured by HPLC using physiological saline as a release medium. 3 parts of 400. Mu.L of NPB@LPM sample having the same mass as the free BUP.HCl or MLX drug was taken in a dialysis bag (3.5K molecular weight cut-off) (n=3), and 4mL of physiological saline was added to the external solution and incubated in a constant temperature shaker at 37.+ -. 0.5 ℃ and 100 rpm. Then 1mL of each tube (0.5, 1, 2, 3, 6, 12h,1, 2, 3, 5, 7 d) was collected at a set time, replaced with new physiological saline, and drug release was calculated by HPLC.
1.7 cytotoxicity
The viability of PC12 cells and C2C12 cells was determined using Cell Counting Kit-8 (CCK-8, CK04, japan Kogyo Co., ltd.). Cells were digested with 0.05% pancreatin-EDTA (25300-062, gibco, U.S.A.), and seeded into 96-well plates (Costar, corning, U.S.A.) at a cell density of 5000-10000 PC12 cells or 5000C 2C12 cells per well. After 24h of incubation, the cells were replenished with 100. Mu.L of fresh medium containing the predetermined concentration of drug. After 24h incubation of the drug, 100. Mu.L fresh medium containing 10. Mu.L CCK-8 reagent was added to each well. After sufficient reaction in an incubator at 37℃in the absence of light for 2 hours, the Optical Density (OD) of the cells was measured at 450nm using an Epoch microplate reader (Bio Tek, wenusky, america VT). Experiments were repeated 3 times. The percentage of viable cells reflecting the cytotoxicity of the drug was calculated using the following formula:
cell viability (%) = (OD drug group-OD blank)/(OD control group-OD blank) ×100%.
1.8 preparation of plantar cutting model
Anesthetized rats were inhaled with 4% isoflurane and maintained anesthetized by mask inhalation with 2% isoflurane. An incision of about 1cm in length was made longitudinally from 0.5cm from the rat heel, the flexor digitorum brevis was blunt isolated with bending forceps and forceps, and the abdomen was scored 5 times with the back of the knife. Also receive muscles and stop bleeding and suture. Then, 0.1mL of physiological saline or drug was subcutaneously injected around the incision. Finally, an erythromycin ointment is applied to the wound.
1.9 pain behavioural test
For mechanical pain threshold, rats were stimulated with an electronic Von Frey instrument (IITC company, usa) hind paws and the mechanical paw withdrawal reflex threshold (Paw Withdraw Mechanical Threshold, PWT) was recorded when the rats were silent. The experiment was repeated 3 times at 5min intervals, and the results were averaged. The protection threshold was 20g. Rats were acclimatized for 30min in a separate plastic box (11X 21X 25 cm) on stainless steel mesh prior to the experiment.
For thermal pain threshold, rats were placed in separate plastic boxes on glass plates for 30min at a constant room temperature of 24 ℃. Radiant heat devices (BME-410A, china biomedical engineering institute) were directly irradiated onto the plantar surface of the hindpaw of each rat, and the paw withdrawal reflex time (Paw Withdraw Thermal Latency, PWL) of the rats was measured. Experiments were repeated 3 times at intervals of 10 min, and the results were averaged. And 20s is taken as a protection threshold value to prevent tissue injury.
1.10 PGE in blood and paw tissues 2 Is (are) determined by
Rats were deeply anesthetized with 1% sodium pentobarbital on postoperative day 1, perfused with PBS hearts, and plantar skin and subdermal soft tissues were harvested and stored at-80 ℃. Skin and subcutaneous tissue were minced and ground, acetonitrile was used: water (1:1, v/v) extracts PGE from tissue 2 。PGE 2 Is passed through an Inertsil ODS-3 column (2.1 mm, 150mm, 5 μm particle size, shimadzu corporation) and chromatographed on LC-MS (Bio-LCMS 8050, shimadzu corporation). Gradient elution profiles were used for each sample at the following intervals: 0.0-3.0min, b phase concentration: 5% to 90%;3.0-3.1min, phase b concentration: 90% to 95%;3.1-5.0min, phase B concentration: is kept at 95%;5.0-5.1min, phase b concentration: 95% to 5%;5.1-6.0min, phase B concentration: is kept at 5%. The flow rate was 300. Mu.l/min, phase A was 0.1% aqueous formic acid and phase B was 0.1% acetonitrile formic acid. Negative ion detection mode for detecting PGE 2 . The mass spectrum multi-reaction monitoring is 351.3-271.2, and the collision energy is-18 eV. Results PGE per microgram of tissue 2 Quality representation based on PGE's from commercial standards 2 (MedChemExpress ltd., usa) a calibration curve was constructed.
1.11 histological analysis
On day 4 post-surgery rats were deeply anesthetized with 1% sodium pentobarbital and perfused with PBS and 4% (W/V) paraformaldehyde. The plantar skin of the NA, NS, B, NPB@LP, NP@LPM, NPB@LPM group, including wound and surrounding skin, subdermal soft tissue and muscle, was taken to assess inflammation and muscle toxicity. The heart, liver, spleen, lung, kidney and other organs were compared with NA group rats at day 7 post-operation to evaluate systemic toxicity. Tissues and organs were fixed, embedded and Hematoxylin and Eosin (HE) stained for histological analysis.
1.12 statistical analysis
Data are expressed as mean ± Standard Deviation (SD). The comparison between the two groups adopts Student t test, and the comparison between the multiple groups adopts single-factor analysis of variance and double-factor analysis of variance. PWT and PWL are direct measurement data and area under the time curve (AUC). Statistical analysis was performed using Graph Pad Prism 9.5.0 software. The difference of P <0.05 is statistically significant.
2 experimental results
2.1 Synthesis and characterization of NPB@LPM core-shell nanosystems
The release profile and analgesic mechanism of npb@lpm for postoperative pain is shown in fig. 1.
PLGA showed higher loading on BUP as shown in table 1. The BUP content can reach 19.27% of the total mass of the preparation. However, the loading rate of PLGA to MLX was relatively low (0.64% of the total mass). Furthermore, when we tried to encapsulate both drugs simultaneously into PLGA nanoparticles, the loading rate of BUP was drastically reduced, accounting for only 7.08% by weight. To overcome this problem, it was decided to construct a core-shell nanostructure based on PLGA-lipids, load MLX into the liposome shell, and load BUP into the PLGA-NP core. Since the MLX is located in the outer layer, this design can release the MLX relatively quickly, which enables the MLX to be used to inhibit acute inflammation after surgery.
PLGA NP diameter was about 119.7.+ -. 3.1nm. After BUP loading, the particle size of NPB is 123.0+ -3.2 nm, which is slightly larger than that of blank NP, and Zeta potential is slightly raised. Both NPs and NPBs are spherical and relatively monodisperse. NPB was then entrapped in MLX-containing Liposomes (LPM) to form a core-shell nanosystem (npb@lpm). The DLS result shows that the average particle size of NPB@LPM is 182.2+ -0.8 nm, and zeta potential is reduced to-27.8+ -0.2 mV. TEM confirmed that the increase in particle size was consistent with core-shell structure assembly, with a lipid shell and PLGA NP core (fig. 2).
NPB@LPM significantly increases the solubility of MLX. Npb@lpm was uniformly dispersed and the solution showed tyndall phenomenon at low concentration (fig. 3A). In contrast, MLX rapidly aggregates into blocks under the same conditions (fig. 3A). The drug release profile of npb@lpm was determined using HPLC. Npb@lpm gradually released MLX (34.3±0.85%) at a relatively fast rate over the first 24 hours compared to insoluble MLX which failed to release MLX (fig. 3B). This drug release behavior helps to suppress acute inflammation caused after surgery. BUP can also be released gradually from NPB@LPM, with more than 80% BUP released within 7 days. In contrast, bupivacaine hydrochloride was released over 80% within 24 hours (fig. 3C). These results indicate that npb@lpm can control the release of MLX and BUP, and that the release behavior can meet the needs of post-operative MMA. In addition, based on the pathophysiological mechanism of postoperative acute pain, other dosage forms cannot achieve the aim of sequential administration.
Table 1 drug loading for each dosage form
2.2 in vitro drug Release
For npb@lpm nanosystems, the release pattern of MLX appeared to be early burst on day 1, reaching 34.3% of the total drug amount. Npb@lpm had a higher bioavailability compared to 7.67% of the total 7 day release of free MLX. Meanwhile, NPB@LPM achieves a 7-day slow release effect of BUP. These results indicate that npb@lpm achieves the aim of sequential and long-acting drug release, indicating its great potential in targeting pain mechanisms for analgesia (fig. 3).
2.3 Cytotoxicity of NPB@LPM
In cytotoxicity studies, the C2C12 cell line was used to assess potential myotoxicity, as was the PC12 cell line (fig. 4). Since npb@lpm can slow release BUP, npb@lpm with the same BUP dose is less toxic to both cell lines, especially for the C2C12 cell line, compared to free BUP. For example, incubation of 200 μg/mL free BUP for 24h resulted in more than 50% C2C12 cell death, whereas no significant cytotoxicity was found in the npb@lpm group (P < 0.01). This result suggests that npb@lpm has the potential to reduce muscle toxicity and neurotoxicity.
2.4 pain behavior study
All groups of rats have good wound healing without dehiscence, and scars are visible on the right hind feet of all groups of rats. The PWT and PWL began to decline 30min post-surgery in NS group and continued until 7d post-surgery. The AUC of the npb@lpm groups PWT and PWL was significantly higher than the other groups (fig. 5).
2.5 anti-inflammatory Studies
The HE staining around the wound on day 3 post-surgery in each group of rats showed that the wound sites were characterized by an inflammatory response with an increase in inflammatory cells. Extensive inflammatory cells and scarring outside the wound were observed in all experimental groups. Whereas the inflammatory response was significantly reduced in the np@lpm and npb@lpm groups (fig. 6).
MLX as a COX-2 selective inhibitor for PGE reduction 2 Is a synthesis of (a). PGE (PGE) 2 Can promote the tissue inflow of immune cells, and can be used for histological observation. To further assess the anti-inflammatory mechanism of MLX, PGE in tissues was assessed by LC-MS method 2 Horizontal. NS group periwound tissue PGE 2 Elevated levels, local injection of NP@LAfter PM or NPB@LPM, PGE 2 The level was higher than that of the NS, B, NPB@LP group (P<0.05 A significant decrease (fig. 7).
2.6 local toxicity Studies
Removal ofOutside the groups, inflammatory cell accumulation was present near the surgical incision in all groups (fig. 6). However, in the MLX-containing groups (NP@LPM and NPB@LPM), the number of inflammatory cells was significantly less than in the BUP and NPB@LP groups. BUP exhibits strong muscle toxicity, and the BUP and npb@lp groups present significant atrophy of muscle fibers and nuclear centering. In contrast, the muscle toxicity of the NPB@LPM and NP@LPM groups was significantly reduced, probably due to the reduction of inflammatory stimuli in the muscle by MLX and the protection of cells by reduction of mitochondrial oxidative stress injury.
2.7 System toxicity Studies
Organ damage, such as heart toxicity and liver and kidney damage, is considered to be direct evidence of systemic toxicity of BUP and MLX. However, withIn comparison to the groups, no histological lesions were found in the histological observations of all groups. No rats developed dyspnea or cardiac arrest throughout the experiment (fig. 8).
The above description of the embodiments is only for the understanding of the method of the present invention and its core ideas. It should be noted that it will be apparent to those skilled in the art that several improvements and modifications can be made to the present invention without departing from the principle of the invention, and these improvements and modifications will fall within the scope of the claims of the invention.

Claims (10)

1. A drug delivery vehicle/system comprising a liposome loaded with a first drug and PLGA nanoparticles loaded with a second drug;
preferably, the liposome comprises a phospholipid and a sterol;
preferably, the phospholipid comprises DOPC, DSPE, DSPE-PEG 2000 、DSPE-PEG 3000 、DSPE-PEG 5000 HSPC, DSPC and/or DPPC;
preferably, the phospholipid comprises DOPC, DSPE-PEG 2000 、DSPE-PEG 3000 And/or DSPE-PEG 5000
Preferably, the phospholipids include DOPC and DSPE-PEG 2000
Preferably, the molecular weight of the PLGA is 24,000-38,000Da;
Preferably, the molar ratio of LA to GA in PLGA is 50:50;
preferably, the sterol is cholesterol;
preferably DOPC, DSPE-PEG 2000 And cholesterol in a mass ratio of 15:1:5.
2. A sustained release pharmaceutical composition comprising a first drug, a second drug and the drug sustained release carrier/system of claim 1.
3. The sustained release pharmaceutical composition of claim 2, wherein the first drug is a non-steroidal anti-inflammatory drug;
preferably, the non-steroidal anti-inflammatory drugs include salicylic acid derivatives, indole and indenacetic acid, fenamate, heteroaryl acetic acid, aryl acetic acid and propionic acid derivatives, enolic acid, para-aminophenol derivatives, alkanones, nimesulide and/or praline Luo Kuizong, or pharmaceutically acceptable salts of these ingredients;
preferably, the non-steroidal anti-inflammatory drug is selected from alkenols;
preferably, the enolic acid comprises an oxicam derivative, a pyrazolone derivative;
preferably, the enolic acid is selected from oxicam derivatives;
preferably, the oxicam derivatives comprise ampiroxicam, cinnoxicam, drooxicam, lornoxicam, meloxicam, piroxicam, sudoxicam, tenoxicam;
Preferably, the oxicam derivative is meloxicam.
4. The extended release pharmaceutical composition of claim 2, wherein the second drug comprises a local anesthetic;
preferably, the local anesthetic comprises bupivacaine, levobupivacaine, tetracaine, ropivacaine, lidocaine, prilocaine, mepivacaine, procaine, chloroprocaine, propoxycaine, hexetidine, cyclomecaine, butoxyprocaine, bupivacaine, procaine, cocaine, finacaine, dibucaine, farivacaine, dyclonine, pramoxine and/or dimethylisoquinoline, or pharmaceutically acceptable salts of these ingredients;
preferably, the local anesthetic is bupivacaine;
preferably, the mass ratio of bupivacaine to PLGA is 1:4.
5. A method of preparing a sustained release pharmaceutical composition, the method comprising:
(1) Mixing PLGA loaded with a second drug into liposomes loaded with a first drug;
(2) Hydrating the lipid film;
(3) Extruding the colloidal solution in the liposome;
preferably, step (2) hydrates the lipid film by water bath ultrasound;
preferably, step (3) extrudes the colloidal solution in the liposomes through a liposome extruder.
6. The method of claim 5, wherein the method of preparing the first drug-loaded liposome comprises: mixing the first medicine, DOPC, DSPE-PEG 2000 And cholesterol in chloroform/methanol solution;
preferably, the method further comprises removing the organic solvent and forming a dry film;
preferably, the volume ratio of the chloroform/methanol solution is 9:1;
preferably, the organic solvent is removed using a rotary evaporator;
preferably, the temperature at which the organic solvent is removed is 30 ℃.
7. The method of claim 5, wherein the method of preparing the second drug loaded PLGA comprises: dripping acetone containing a second drug and PLGA into a polyvinyl alcohol aqueous solution to obtain a mixture;
preferably, the method further comprises stirring the mixture;
preferably, the speed at which the mixture is stirred is from 100 to 2000rpm;
preferably, the speed at which the mixture is stirred is 500-1000rpm;
preferably, the speed at which the mixture is stirred is 800rpm;
preferably, the stirring speed is 800rpm for 1-10 hours;
preferably, the stirring speed is 800rpm for 3-5 hours;
preferably, the stirring speed is 800rpm for 4 hours;
preferably, the step of stirring the mixture further comprises the step of increasing the stirring speed;
Preferably, the increase in agitation speed comprises a gradual increase in speed from 0 to 2000 rpm;
preferably, the increase in agitation speed comprises a gradual increase in speed from 0 to 1000 rpm;
preferably, the increase in stirring speed means a gradual increase in speed from 100rpm to 800rpm;
preferably, the stirring speed is gradually increased from 100rpm to 800rpm over 1 to 30 minutes;
preferably, the stirring speed is gradually increased from 100rpm to 800rpm over 5-20 minutes;
preferably, the stirring speed is gradually increased from 100rpm to 800rpm over 10 minutes;
preferably, the method further comprises removing the organic solvent;
preferably, the organic solvent is removed using a rotary evaporator;
preferably, the method further comprises removing the free second drug;
preferably, the free second drug is removed by repeated washing.
8. Use of a drug delivery vehicle/system according to claim 1 for the preparation of a drug delivery having a plurality of active ingredients;
preferably, the medicament further comprises a pharmaceutically acceptable carrier.
9. Use of a sustained release pharmaceutical composition according to any one of claims 2-4 in the manufacture of a medicament for the treatment of a disease;
preferably, the disease is selected from pain and inflammation;
preferably, the pain comprises post-operative analgesia;
Preferably, the postoperative analgesia is selected from postoperative acute pain;
preferably, the medicament further comprises a pharmaceutically acceptable carrier.
10. Use of the method of any one of claims 5-7 for the preparation of a sustained release medicament having a plurality of active ingredients;
preferably, the medicament further comprises a pharmaceutically acceptable carrier.
CN202310902318.8A 2023-07-21 2023-07-21 Pharmaceutical composition based on core-shell structure and application Active CN116688137B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310902318.8A CN116688137B (en) 2023-07-21 2023-07-21 Pharmaceutical composition based on core-shell structure and application

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310902318.8A CN116688137B (en) 2023-07-21 2023-07-21 Pharmaceutical composition based on core-shell structure and application

Publications (2)

Publication Number Publication Date
CN116688137A true CN116688137A (en) 2023-09-05
CN116688137B CN116688137B (en) 2024-05-14

Family

ID=87837649

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310902318.8A Active CN116688137B (en) 2023-07-21 2023-07-21 Pharmaceutical composition based on core-shell structure and application

Country Status (1)

Country Link
CN (1) CN116688137B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116840378A (en) * 2023-07-11 2023-10-03 加立(深圳)生物科技有限公司 Method for detecting content of monohydrate of ropivacaine/meloxicam salt

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5922340A (en) * 1992-09-10 1999-07-13 Children's Medical Center Corporation High load formulations and methods for providing prolonged local anesthesia
WO2011043532A1 (en) * 2009-10-07 2011-04-14 건일제약 주식회사 Lipid nanoparticles for oral administration, and method for preparing same
CN106177986A (en) * 2016-08-16 2016-12-07 国家纳米科学中心 A kind of lipopolymer medicine-carried nano particles and its preparation method and application
CN112654348A (en) * 2018-09-13 2021-04-13 台湾微脂体股份有限公司 Slow-release pharmaceutical composition containing sedative and application thereof
CN114177133A (en) * 2021-12-17 2022-03-15 中国医学科学院北京协和医院 Drug sustained-release carrier, sustained-release drug composition and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5922340A (en) * 1992-09-10 1999-07-13 Children's Medical Center Corporation High load formulations and methods for providing prolonged local anesthesia
WO2011043532A1 (en) * 2009-10-07 2011-04-14 건일제약 주식회사 Lipid nanoparticles for oral administration, and method for preparing same
CN106177986A (en) * 2016-08-16 2016-12-07 国家纳米科学中心 A kind of lipopolymer medicine-carried nano particles and its preparation method and application
CN112654348A (en) * 2018-09-13 2021-04-13 台湾微脂体股份有限公司 Slow-release pharmaceutical composition containing sedative and application thereof
CN114177133A (en) * 2021-12-17 2022-03-15 中国医学科学院北京协和医院 Drug sustained-release carrier, sustained-release drug composition and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MOHAN LI ET AL.: "Construction of meloxicam and bupivacaine co-delivery nanosystem based on the pathophysiological environment of surgical injuries for enhanced postoperative analgesia", NANO RESEARCH, pages 12274 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116840378A (en) * 2023-07-11 2023-10-03 加立(深圳)生物科技有限公司 Method for detecting content of monohydrate of ropivacaine/meloxicam salt
CN116840378B (en) * 2023-07-11 2024-03-22 加立(深圳)生物科技有限公司 Method for detecting content of monohydrate of ropivacaine/meloxicam salt

Also Published As

Publication number Publication date
CN116688137B (en) 2024-05-14

Similar Documents

Publication Publication Date Title
US10596132B2 (en) Solid solution compositions and use in chronic inflammation
EP3578172B1 (en) Solid solution compositions and use in chronic inflammation
US11964050B2 (en) Liposome compositions comprising weak acid drugs and uses thereof
US20220040102A1 (en) Solid Solution Compositions and Use in Chronic Inflammation
JPWO2014046191A1 (en) Local anesthetic sustained-release liposome preparation
CN116688137B (en) Pharmaceutical composition based on core-shell structure and application
JP7140418B2 (en) Pharmaceutical composition for controlled release of treprostinil
JP2023123843A (en) Pharmaceutical composition for controlled release of weak acid drugs and uses thereof
RU2778886C2 (en) Liposomal compositions containing slightly acidic drugs, and their use
RU2796305C2 (en) Pharmaceutical composition for trepostinil controlled release
RU2810790C2 (en) Pharmaceutical composition for controlled release of weak acid drugs and its use

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant