WO2024109612A1 - Procédé de préparation de nanoparticule lipidique pour administration efficace d'un médicament à base d'acide nucléique et utilisation associée - Google Patents

Procédé de préparation de nanoparticule lipidique pour administration efficace d'un médicament à base d'acide nucléique et utilisation associée Download PDF

Info

Publication number
WO2024109612A1
WO2024109612A1 PCT/CN2023/131882 CN2023131882W WO2024109612A1 WO 2024109612 A1 WO2024109612 A1 WO 2024109612A1 CN 2023131882 W CN2023131882 W CN 2023131882W WO 2024109612 A1 WO2024109612 A1 WO 2024109612A1
Authority
WO
WIPO (PCT)
Prior art keywords
lipid
nucleic acid
ionizable
glyceryl
lnp
Prior art date
Application number
PCT/CN2023/131882
Other languages
English (en)
Chinese (zh)
Inventor
吕雪光
林佳奇
程星迪
Original Assignee
中国科学院化学研究所
深圳近邻生物科技有限公司
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 中国科学院化学研究所, 深圳近邻生物科技有限公司 filed Critical 中国科学院化学研究所
Publication of WO2024109612A1 publication Critical patent/WO2024109612A1/fr

Links

Classifications

    • 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
    • A61K9/5192Processes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • 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
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • 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
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • 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
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • 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
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • 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
    • A61K9/5123Organic compounds, e.g. fats, sugars
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y40/00Manufacture or treatment of nanostructures
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16711Varicellovirus, e.g. human herpesvirus 3, Varicella Zoster, pseudorabies
    • C12N2710/16734Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the technical field to which the present invention belongs is the field of medical technology, and specifically relates to a lipid nanoparticle (LNP) capable of efficiently delivering nucleic acid drugs, and a preparation method and application thereof.
  • LNP lipid nanoparticle
  • mRNA messenger RNA
  • mRNA vaccines have the advantages of high safety, short R&D cycle, high production efficiency, ability to encode multiple proteins, and no need for adjuvants.
  • mRNA itself is difficult to enter cells, and mRNA itself has poor stability and is easily degraded by nucleases in the body. Therefore, a suitable delivery vehicle is needed to assist mRNA in entering target cells to work.
  • Lipid nanoparticles are currently the only clinically approved mRNA vaccine carriers, which are composed of ionizable lipids, auxiliary phospholipids, cholesterol and pegylated lipids. LNP can effectively deliver mRNA into cells after intramuscular injection, activate cellular immunity and humoral immune responses, and produce effective immune protection.
  • the delivery efficiency of LNP is the key to the efficacy of mRNA vaccines, and the low lysosomal escape efficiency of LNP after entering the cell is the key to limiting the delivery efficiency of LNP. Therefore, it is urgent to develop new carriers that can increase the lysosomal escape efficiency of LNP and improve the delivery efficiency of LNP.
  • the present invention provides a lipid nanoparticle (hereinafter referred to as LNP), comprising a carrier and an encapsulated nucleic acid, wherein the carrier comprises a composite ionizable lipid, an auxiliary phospholipid, a cholesterol-like substance and a pegylated lipid; the nucleic acid comprises but is not limited to one or more of mRNA, circular RNA, siRNA, microRNA, antisense nucleic acid and a plasmid.
  • LNP lipid nanoparticle
  • the complex ionizable lipid accounts for 20 mol% to 70 mol% of the total lipids in the LNP, such as 30 mol%, 40 mol%, 50 mol%, 60 mol%.
  • the complex ionizable lipid comprises a first ionizable lipid and a second ionizable lipid.
  • the molar ratio of the first ionizable lipid to the second ionizable lipid is 1:99 to 99:1, preferably 2:8 to 8:2, such as 3:7, 4:6, 5:5, 6:4, 7:3, and more preferably 6:4.
  • the first ionizable lipid and the second ionizable lipid are different and are independently selected from 8-[(2-hydroxyethyl)(6-oxo-6-decyloxyhexyl)amino]octanoic acid (heptadecan-9-yl) ester (SM-102), [(4-hydroxybutyl)azepinediyl]bis(hexane-6,1-diyl)bis(2-hexyldecanoate) (ALC-0315), 4(N,N-dimethylamino)butyric acid (dilinoleyl) methyl ester (DLin MC3DMA), 3,6-bis ⁇ 4-[bis(2-hydroxydodecyl)amino]butyl ⁇ piperazine-2,5-dione (cKK-E12), 9-(4-(dimethylamino)butyryloxy)heptadecanedioic
  • the complex ionizable lipid includes 3,6-bis ⁇ 4-[bis(2-hydroxydodecyl)amino]butyl ⁇ piperazine-2,5-dione (cKK-E12) and 9-(4-(dimethylamino)butyryloxy)heptadecanedioic acid di((Z)-non-2-en-1-yl) ester (L319), and the molar ratio thereof is 1:99 to 99:1, preferably 2:8 to 8:2, for example 3:7, 4:6, 5:5, 6:4, 7:3.
  • the auxiliary phospholipid accounts for 2 mol% to 20 mol% of the total lipids in the LNP, for example 4 mol%, 8 mol%, 10 mol%, 12 mol%, 4 mol%, 16 mol%, 18 mol%.
  • the auxiliary phospholipid includes but is not limited to 1,2-distearoyl-sn-glyceryl-3-phosphatidylcholine (DSPC), 1,2-dioleoyl-sn-glyceryl-3-phosphatidylcholine (DOPC), 1,2-dipalmitoyl-sn-glyceryl-3-phosphatidylcholine (DPPC), 2-oleoyl-1-palmitoyl-sn-glyceryl-3-phosphatidylcholine (POPC), 1,2-dioleoyl-sn-glyceryl-3-phosphatidylethanolamine (DOPE), 2-oleoyl-1-palmitoyl-sn-glyceryl-3-phosphatidylethanolamine (POPE), 1,2-distearoyl-sn-glyceryl-3-phosphatidylethanolamine (DSPE), 1,2-dipalmitoyl-sn-glyceryl-3-phosphatidylcholine (DS
  • the cholesterol-like substance accounts for 10 mol% to 60 mol% of the total lipids in the LNP, for example, 20 mol%, 30 mol%, 40 mol%, 50 mol%.
  • the cholesterol-like substance is selected from cholesterol and its derivatives.
  • the cholesterol and its derivatives include but are not limited to one or more of cholesterol, ⁇ -sitosterol, cholestanol, cholestanone, cholestenone, 7 ⁇ -hydroxycholesterol, 7 ⁇ -hydroxycholesterol, preferably cholesterol.
  • the PEGylated lipid accounts for 0.3 mol% to 30 mol% of the total lipid in the LNP, preferably 0.5 mol% to 2.5 mol%, such as 1.0 mol%, 1.5 mol%, 2.0 mol%.
  • the PEGylated lipids include but are not limited to one or more of 1,2-dimyristoyl-rac-glyceryl-3-methoxypolyethylene glycol (DMG-PEG), 1,2-distearoyl-rac-glyceryl-3-methoxypolyethylene glycol (DSG-PEG), 1,2-dipalmitoyl-rac-glyceryl-3-methoxypolyethylene glycol (DPG-PEG), and 1,2-distearoyl-sn-glyceryl-3-phosphatidylethanolamine-methoxypolyethylene glycol (DSPE-PEG), preferably DMG-PEG.
  • DMG-PEG 1,2-dimyristoyl-rac-glyceryl-3-methoxypolyethylene glycol
  • DSG-PEG 1,2-distearoyl-rac-glyceryl-3-methoxypolyethylene glycol
  • DPG-PEG 1,2-dipalmitoyl-rac-glyceryl-3-me
  • total lipids refers to the sum of complex ionizable lipids, auxiliary phospholipids, cholesterol substances and PEGylated lipids.
  • the nucleic acid selected by the present invention includes at least one of CpG-FAM, mFLuc (mRNA encoding firefly luciferase), mOVA (mRNA encoding chicken ovalbumin), and mVZV (mRNA encoding herpes zoster gE protein).
  • the encapsulation rate of the nucleic acid is 30% to 99%, preferably 70%-90%, such as 70%, 75%, 80%, 85%, 90%.
  • the molar ratio of the nitrogen element contained in the complex ionizable lipid to the phosphorus element contained in the nucleic acid is (1-50):1; preferably (5-10):1, and exemplarily 5.67:1.
  • the hydrated particle size of the lipid nanoparticles is less than 200 nm, such as 80 to 180 nm.
  • the polydispersity coefficient of the lipid nanoparticles is less than 0.2, for example, 0.119, 0.102, or 0.093.
  • the molar ratio of the complex ionizable lipid: helper phospholipid: cholesterol-like substance: polyethylene glycol lipid is 50:10:38.5:1.5.
  • the composite ionizable lipid comprises a first ionizable lipid and a second ionizable lipid, the two being Examples are 8:2, 7:3, 6:4, 5:5, 4:6, 3:7, or 2:8.
  • the nucleic acid is CpG-FAM, mFLuc, mOVA or mVZV.
  • the present invention also provides a method for preparing the lipid nanoparticles, which comprises preparing the lipid nanoparticles by blending an aqueous phase containing nucleic acids and a lipid organic phase through a blending method or a microfluidics method.
  • the lipid organic phase is prepared as follows: the composite ionizable lipid, auxiliary phospholipid, cholesterol-like substance and pegylated lipid are dissolved in an organic solvent in proportion to obtain an organic phase.
  • the organic solvent is an organic solvent that can dissolve the above substances, such as anhydrous ethanol.
  • the preparation method of the aqueous phase containing nucleic acid is as follows: dissolving the nucleic acid in a buffer solution to obtain an aqueous phase.
  • the buffer solution can be a buffer solution with a pH of 4 to 6, for example, a sodium citrate buffer solution with a pH of 4 or 5.5.
  • the blending method includes the following steps: using a pipette to absorb the aqueous phase containing nucleic acids, quickly adding it to the lipid organic phase, and then quickly blowing the mixed liquid for more than 30 times, blowing it evenly and letting it stand at room temperature for 10 minutes, and dialyzing the resulting product after standing, for example, dialyzing it in a PBS buffer solution with a volume greater than 1000 times for more than 6 hours to obtain the lipid nanoparticles.
  • the microfluidic method includes the following steps: fixing syringes containing an aqueous phase and a lipid organic phase on a syringe pump respectively, injecting the aqueous phase: lipid organic phase into a microfluidic chip at a volume ratio of 3:1 and mixing thoroughly, collecting the mixed solution after mixing and standing at room temperature for 10 minutes, and dialyzing the resulting product with a PBS buffer solution after standing, for example, dialyzing in a PBS buffer solution with a volume greater than 1000 times for more than 6 hours to obtain the lipid nanoparticles.
  • the present invention also provides the use of the lipid nanoparticles in the preparation of pharmaceutical preparations or biological preparations.
  • the biologic is an injectable biologic.
  • the biologic is a vaccine, preferably an mRNA vaccine.
  • the administration method of the biological preparation is intramuscular injection.
  • the biological agent is used to prevent herpes zoster.
  • the present invention also provides a pharmaceutical preparation or a biological preparation having the meaning as described above.
  • the present invention also provides a method for preventing and/or treating a disease, comprising administering a preventively and/or therapeutically effective amount of the above-mentioned pharmaceutical preparation or biological product to a subject; for example, the disease is herpes zoster.
  • the term "effective amount" can be determined according to the method mastered by a physician with clinical practice qualifications in the field to determine the amount of the pharmaceutical preparation or biological product of the present invention that is sufficient to achieve the intended application (including but not limited to the treatment of diseases defined above). Determining the effective dose for prevention and/or treatment is within the capabilities of clinicians or researchers in the field and may be changed due to the following factors: the intended application (in vitro or in vivo), or the subject and disease condition to be treated, such as the subject's weight and age, general health, severity of the disease condition, mode of administration, and other factors that affect the efficacy, such as a history of drug allergies.
  • the specific dosage will vary depending on the following factors: the specific biological product selected, the dosage regimen based on, whether it is co-administered with other drugs, the timing of administration, the tissue to which it is administered, and the physical delivery system carried.
  • the "subject" of the present invention refers to a specific human or other warm-blooded mammal.
  • the human as “subject” of the present invention includes adults, infants, and children, and other warm-blooded mammals include but are not limited to non-human primates, such as chimpanzees, other apes or monkeys, and other zoo animals, domestic mammals or laboratory animals, such as cats, pigs, dogs, cows, sheep, mice, rats, and guinea pigs.
  • the LNP prepared by the present invention has uniform particle size, good dispersibility, high encapsulation efficiency, good biocompatibility, low toxicity and side effects, and can be highly
  • the LNP preparation method of the present invention has simple operation steps and is easy to mass produce.
  • the present invention combines a first ionizable lipid and a second ionizable lipid to form a composite ionizable lipid, which has an ionizable lipid with high efficiency in entering cells and an ionizable lipid with a long alkyl chain and high efficiency in membrane fusion ability.
  • the composite ionizable lipid of the present invention can significantly improve the mRNA expression efficiency of the commercial four-component LNP through synergistic effect.
  • the present invention uses composite ionizable lipids to prepare five-component LNPs, which have a higher activation efficiency on antigen-presenting cells in draining lymph nodes and spleen than commercial four-component LNPs after intramuscular injection.
  • the five-component LNP prepared by the composite ionizable lipids of the present invention can efficiently deliver the herpes zoster mRNA vaccine. After intramuscular injection, it significantly activates the cellular immunity and humoral immune response in the body, produces a large number of antigen-specific binding antibodies in the blood, and increases the antigen-specific T cell content in the spleen, which has an excellent protective effect. It is expected to achieve a breakthrough in the field of herpes zoster mRNA vaccines and has great application prospects.
  • FIG. 1 Chemical structures of the first ionizable lipid and the second ionizable lipid used in the examples of the present invention.
  • FIG. 2 Hydration particle size distribution curve of LNP obtained in Example 1B of the present invention.
  • FIG3 Cryo-TEM image of the five-component LNP obtained in Example 1A of the present invention.
  • FIG. 4 shows the bioluminescence intensity of DC2.4 cells transfected with LNPs loaded with mFluc in Example 3 of the present invention.
  • FIG5 The membrane fusion ability of the five-component LNP was detected using mouse red blood cells in Example 4 of the present invention.
  • Example 5 of the present invention in vivo imaging of small animals was used to verify the bioluminescence intensity of the five-component LNP loaded with mFLuc at the injection site of mice; (a) In vivo imaging image and statistical results of bioluminescence intensity at the injection site 4 hours after intramuscular injection; (b) In vivo imaging image and statistical results of bioluminescence intensity at the injection site 24 hours after intramuscular injection.
  • Figure 7 Flow chart of animal experiment in Example 6 of the present invention.
  • FIG8 Activation efficiency of different LNPs loaded with mOVA on DC cells in draining lymph nodes and spleen in Example 6 of the present invention.
  • Figure 9 Flow chart of animal experiment in Example 7 of the present invention.
  • Figure 10A Antigen-specific Total IgG antibody titers in the serum of mice on days 14, 28, and 35 after the first intramuscular injection of LNPs loaded with mVZV in Example 7 of the present invention.
  • Figure 11 The number of antigen-specific splenocytes secreting IFN- ⁇ in the spleen of mice on the 42nd day after the first intramuscular injection of LNPs loaded with mVZV in Example 7 of the present invention (a) ELISpot spot number statistics; (b) optical photograph of the ELISpot well.
  • Figure 12 The number of antigen-specific CD4 + T cells secreting IFN- ⁇ in the spleen of mice on day 42 after the first intramuscular injection of LNPs loaded with mVZV in Example 7 of the present invention.
  • Figure 13 Concentrations of IL-2 and IFN- ⁇ secreted by antigen-specific spleen cells on day 42 after the first intramuscular injection of LNPs encapsulating mVZV in mice in Example 7 of the present invention.
  • Figure 14 Body weight monitoring curve of mice during immunization in Example 8 of the present invention.
  • FIG. 15 H&E staining of tissue and organ sections on day 42 after the first intramuscular injection of LNPs loaded with mVZV in mice in Example 8 of the present invention.
  • Figure 16 Flow chart of animal experiment in Example 9 of the present invention.
  • Figure 17 Total serum levels at 2, 4 and 6 weeks after the first intramuscular injection of LNPs loaded with mVZV in mice in Example 9 of the present invention Antibody titers of IgG, IgG1, IgG2a, and IgG2c.
  • Figure 18 Total IgG antibody titer in the serum of mice during 8 months after the first intramuscular injection of LNPs loaded with mVZV in Example 9 of the present invention.
  • Figure 19 The number of specific splenocytes secreting IFN- ⁇ in the spleen of mice on the 42nd day after the first intramuscular injection of LNPs loaded with mVZV in Example 9 of the present invention (a) optical photograph of ELISpot wells; (b) statistical graph of the number of ELISpot spots.
  • Figure 20 Body weight monitoring curve of mice during immunization process in Example 9 of the present invention.
  • Figure 21 Schematic diagram of the deglycosylation site in Example 10 of the present invention.
  • Figure 22 Total IgG, IgG1, IgG2a and IgG2c antibody titers in the serum of mice at 2, 4 and 6 weeks after the first intramuscular injection of LNPs loaded with mVZV in Example 10 of the present invention.
  • Figure 23 Flow chart of animal experiments in Example 11 of the present invention.
  • Figure 24 A diagram of the skin changes on the back of a guinea pig after infection with a poison in Example 11 of the present invention.
  • Figure 25 Guinea pig skin lesion scoring table in Example 11 of the present invention.
  • Figure 26 Skin lesion scores on the back of guinea pigs after poison challenge in Example 11 of the present invention.
  • Figure 27 H&E staining of the skin tissue of the guinea pig's back 7 days after infection in Example 11 of the present invention.
  • Figure 28 Skin viral load on the back of guinea pigs 7 days after infection in Example 11 of the present invention.
  • Figure 29 Body weight monitoring curve of guinea pigs during immunization in Example 11 of the present invention.
  • MC in the above figures represents the product of Comparative Example 1, i.e., a four-component LNP with MC3 as the ionizable lipid;
  • cKK-E12 represents the product of Comparative Example 2, i.e., a four-component LNP with cKK-E12 as the ionizable lipid;
  • L319 represents the product of Comparative Example 3, i.e., a four-component LNP with L319 as the ionizable lipid.
  • the composite ionizable lipid includes a first ionizable lipid 3,6-bis ⁇ 4-[bis(2-hydroxydodecyl)amino]butyl ⁇ piperazine-2,5-dione (abbreviated as cKK-E12), and a second ionizable lipid 9-(4-(dimethylamino)butyryloxy)heptadecanedioic acid di((Z)-non-2-en-1-yl) ester (abbreviated as L319);
  • the auxiliary phospholipid is 1,2-distearoyl-sn-glyceryl-3-phosphocholine, abbreviated as DSPC;
  • the polyethylene glycol lipid is 1,2-dimyristoyl-sn-glyceryl-3-methoxypolyethylene glycol 2000, abbreviated as DMG-PEG2000.
  • the structural formulas of the first ionizable lipid and the second ionizable lipid
  • Five-component lipid nanoparticles (referred to as five-component LNPs) were prepared as follows:
  • the preparation method of the five-component lipid nanoparticles of this embodiment is basically the same as that of Example 1A, wherein the nucleic acid drug is mRNA encoding firefly luciferase (mFLuc), and the molar ratio of cKK-E12 and L319 is 2:8, 3:7, 4:6, 5:5, 6:4, 7:3 or 8:2.
  • the nucleic acid drug is mRNA encoding firefly luciferase (mFLuc)
  • molar ratio of cKK-E12 and L319 is 2:8, 3:7, 4:6, 5:5, 6:4, 7:3 or 8:2.
  • the preparation method of the five-component lipid nanoparticles of this example is basically the same as that of Example 1A, except that the nucleic acid drug is mRNA encoding chicken ovalbumin (mOVA).
  • the nucleic acid drug is mRNA encoding chicken ovalbumin (mOVA).
  • the preparation method of the five-component lipid nanoparticles of this embodiment is basically the same as that of Example 1A, except that the nucleic acid drug is mRNA encoding the herpes zoster virus gE protein (mVZV).
  • the nucleic acid drug is mRNA encoding the herpes zoster virus gE protein (mVZV).
  • Comparative Example 1 the composite ionizable lipid was replaced with MC3 (i.e., 4(N,N-dimethylamino)butyric acid (dilinoleyl) methyl ester), i.e., a four-component LNP with MC3 as the ionizable lipid;
  • MC3 i.e., 4(N,N-dimethylamino)butyric acid (dilinoleyl) methyl ester
  • Comparative Example 2 the composite ionizable lipid was replaced with cKK-E12 (i.e., the molar ratio of cKK-E12 to L319 was 1:0), i.e., a four-component LNP with cKK-E12 as the ionizable lipid;
  • the hydrated particle size and polydispersity index of the five-component LNP of Example 1A are 140 nm and 0.09, respectively, the hydrated particle size and polydispersity index of the five-component LNP of Example 1C are 110 nm and 0.1, respectively, and the hydrated particle size and polydispersity index of the five-component LNP of Example 1D are 130 nm and 0.1, respectively.
  • the hydrated particle sizes of the five-component lipid nanoparticles prepared in Examples 1A to 1D are all less than 200 nm, and the polydispersity coefficient is less than 0.2, indicating that the preparation method of the present invention can form stable lipid nanoparticles with uniform particle size.
  • the five-component LNPs loaded with mFLuc prepared in Example 1B were used, and the four-component LNPs of Comparative Examples 1 to 3 were selected as a control group to screen different ratios of cKK-E12 and L319 as ionizable lipids to act synergistically.
  • the experimental steps are as follows: DC2.4 cells were incubated overnight in a 96-well plate at 20,000 cells per well. When the cell confluence reached 50%, LNPs of Example 1B and Comparative Examples 1 to 3 were added respectively, and cultured in a saturated humidity incubator at 37°C and 5% CO2 concentration for 24 hours. An Untreated cells group (referring to cells without any treatment after plating) and a Free mRNA group (referring to mRNA without a carrier being added directly to the cell culture medium) were also set up. The culture medium was removed, cell lysate and firefly luciferase substrate were added, and after shaking for 10 minutes, the bioluminescence value was read using an ELISA reader. The test results are shown in Figure 4.
  • Example 1A The mFLuc-encapsulated five-component LNP prepared in Example 1A was used, and the four-component LNP of Comparative Examples 1 to 3 was selected as a control group to detect the membrane fusion ability of the five-component LNP.
  • mice erythrocytes are placed in a transparent 96-well plate, sodium citrate buffer solution of pH 5.5 and PBS buffer solution of pH 7.4 are added respectively, and then LNPs of Example 1 and Comparative Examples 1 to 3 are added correspondingly and incubated at 37°C for 1 hour.
  • a negative control group i.e., only buffer solution and erythrocytes are added to the well plate, recorded as Untreated cells
  • a positive control group i.e., buffer solution, erythrocytes and 0.5% Triton X-100 are added to the well plate, recorded as 0.5% Triton X-100 are incubated at 37°C for 1 hour.
  • Example 1A The mFLuc-encapsulated five-component LNP prepared in Example 1A was used, and the four-component LNP of Comparative Examples 1 to 3 were selected as a control group to detect the mRNA expression efficiency of the five-component LNP of the present invention in vivo.
  • mice Female C57BL/6 mice aged 6-8 weeks were selected, and 2 ⁇ g of LNP of Example 1A and Comparative Examples 1 to 3 were administered to each mouse in each group by intramuscular injection. 200 ⁇ L of 200 mg/mL firefly luciferase substrate was injected intraperitoneally into each mouse 4 hours and 24 hours after administration, respectively. After 15 minutes, the bioluminescence intensity in the mice was detected using small animal in vivo imaging.
  • the five-component LNP of the present invention has a high mRNA expression efficiency in vivo.
  • Example 1C The mOVA-encapsulated five-component LNP prepared in Example 1C was used, wherein the four-component LNP of Comparative Example 1 was selected as a control group, and the DC cell activation efficiency of the five-component LNP of the present invention in C57BL/6 mice was detected.
  • mice 8-week-old female C57BL/6 mice were selected, with 4 mice in each experimental group. Each group of mice was immunized by intramuscular injection on day 0 and day 7, and each mouse in each group was administered 10 ⁇ g of LNP of Example 1C and Comparative Example 1, respectively. A PBS group was also set up in which only PBS buffer solution was injected; on day 8, the mice were killed, and the draining lymph nodes and spleen at the injection site were removed in vitro. For the experimental process, see Figure 7.
  • lymph node cells and spleen cells were processed separately, a single cell suspension was obtained, and the number of activated DC cells (CD11c + CD80 + , CD11c + CD86 + ) in the spleen and draining lymph nodes was detected by flow cytometry, see Figure 8.
  • the mRNA-loaded five-component LNP of the present invention has a high DC cell activation efficiency in C57BL/6 mice in the draining lymph nodes and spleen of mice.
  • Example 7 Verification of the immune efficacy of five-component LNP in C57BL/6 mice
  • the five-component LNPs encapsulating mVZV prepared in Example 1D were used, wherein the four-component LNPs of Comparative Example 1, the attenuated VZV virus vaccine (hereinafter referred to as the attenuated vaccine) and the herpes zoster recombinant gE protein subunit vaccine plus AS01 adjuvant (hereinafter referred to as the subunit vaccine) were selected as the control groups of this experiment to detect the immune efficacy of the five-component LNPs of the present invention in C57BL/6 mice.
  • the attenuated vaccine the attenuated VZV virus vaccine
  • AS01 adjuvant hereinafter referred to as the subunit vaccine
  • mice 8-week-old female C57BL/6 mice were selected, with 4 mice in each experimental group. Each group of mice was immunized by intramuscular injection at week 0 and week 3. Each mouse in each group was administered 10 ⁇ g of LNP, attenuated vaccine and subunit vaccine of Example 1D and Comparative Example 1, respectively. A PBS group was also set up to be injected with only PBS buffer solution. Blood was collected from the mice at week 2, 4 and 5, and serum was separated. The mice were killed at week 6. The experimental process is shown in Figure 9. The ELISA was used to measure the 14th, 28th and 35th day of the mice. The test results of the antibody titers of herpes zoster-specific IgG, IgG1, and IgG2c in serum are shown in FIG. 10A to FIG. 10C .
  • the number of herpes zoster-specific T cells (CD3 + CD4 + CD45 + IFN- ⁇ + ) was detected by flow cytometry, and the test results are shown in Figure 12.
  • Splenocytes were then placed in a 96-well plate with 1 million cells per well, stimulated with the herpes zoster gE peptide library for 48 hours, centrifuged to obtain the cell supernatant, and the amount of IL-2 and IFN- ⁇ secreted by spleen cells in the cell supernatant was measured by ELISA, and the test results are shown in Figure 13.
  • the mRNA-loaded five-component LNP of the present invention has the ability to efficiently activate specific cellular immunity and humoral immunity in C57BL/6 mice.
  • Example 8 Verification of the toxicity of five-component LNP in vivo
  • the five-component LNPs loaded with mVZV prepared in Example 1D were used, and the four-component LNPs, attenuated vaccine and subunit vaccine of Comparative Example 1 were selected as the control groups of this experiment to detect the vaccine efficacy of the five-component LNPs in vivo.
  • mice 8-week-old female C57BL/6 mice were selected, with 4 mice in each experimental group. Each group of mice was immunized by intramuscular injection at week 0 and week 3. Each mouse in each group was administered 10 ⁇ g of the five-component LNP in Example 1D and the four-component LNP, attenuated vaccine and subunit vaccine in Comparative Example 1, respectively.
  • a PBS group was set up in which only PBS buffer solution was injected. The weight of the mice was monitored during the immunization period. See Figure 14 for the experimental process. The mice were killed in week 6, and the heart, liver, spleen, lung, kidney, brain and injection site muscles of the mice were embedded in paraffin for H&E staining sections. See Figure 15 for the test results.
  • the five-component LNPs loaded with mVZV prepared in Example 1D were used, wherein the four-component LNPs of Comparative Example 1, the attenuated vaccine and the subunit vaccine were selected as the control groups of this experiment to detect the vaccine efficacy of the five-component LNPs in BALB/c mice.
  • mice 8-week-old female BALB/c mice were selected, with 3 mice in each experimental group, and the mice were immunized by intramuscular injection at week 0 and week 3, respectively.
  • Each mouse was administered 10 ⁇ g of the five-component LNP in Example 1D and the four-component LNP, attenuated vaccine and subunit vaccine in Comparative Example 1, respectively.
  • Orbital blood was drawn from the mice at weeks 2, 4 and 6, the serum was separated, and the mice were killed at week 6. See Figure 16 for the experimental process.
  • ELISA was used to measure the antibody titers of Total IgG, IgG1, IgG2a and IgG2c specific to herpes zoster in the serum of mice at 2, 4 and 6 weeks, see Figure 17.
  • ELISA was used to measure the changes in the titers of Total IgG specific to herpes zoster in the serum of mice within 8 months after immunization, see Figure 18.
  • the spleen of the mice was processed to obtain a single cell suspension, and 300,000 cells were placed in each well of the ELISpot plate and stimulated with the herpes zoster gE peptide library for 24 hours, and then the number of spleen cells that secreted IFN- ⁇ specifically for herpes zoster was detected by color development, see Figure 19 (a) ELISpot well optical photograph, (b) ELISpot spot number statistical chart. The weight of the mice was monitored during the immunization period, see Figure 20.
  • Example 10 Verification of the effect of five-component LNPs loaded with mVZV of different deglycosylation designs on humoral immunity
  • mVZV in Example 1D was used to perform different deglycosylation site designs, including: two nitrogen-glycosylation site mutations (abbreviated as mMUTE-N), six oxygen-glycosylation site mutations (abbreviated as mMUTE-O), and a combination of two glycosylation site mutations (abbreviated as mMUTE-N+O), see Figure 21.
  • mMUTE-N nitrogen-glycosylation site mutations
  • mMUTE-O oxygen-glycosylation site mutations
  • mMUTE-N+O a combination of two glycosylation site mutations
  • Example 9 Referring to the immune experiment process and experimental steps in Example 9, specifically including: selecting 8-week-old female BALB/c mice, 6 mice in each experimental group, immunizing the mice by intramuscular injection at week 0 and week 3, and administering 10 ⁇ g of the five-component LNP in Example 1D and the five-component LNP containing mMUTE-N, the five-component LNP containing mMUTE-O, and the five-component LNP containing mMUTE-N+O prepared above, respectively, which are recorded as VZV, mMUTE-N, mMUTE-O, and mMUTE-N+O, respectively, and collecting blood from the eye sockets of the mice at weeks 2, 4, and 6 to separate the serum.
  • Example 11 Verification of the five-component LNP in guinea pigs against VZV virus invasion
  • the five-component LNPs containing mVZV prepared in Example 1D were used, and subunit vaccines and PBS were selected as the control group of this experiment to detect the efficacy of the five-component LNPs in guinea pigs against the virus.
  • the experimental process is shown in Figure 23. The experimental steps are specifically as follows: guinea pigs weighing 200-250g were selected, and the guinea pigs were immunized by intramuscular injection on days 0 and 14. The guinea pigs were administered 10 ⁇ g of the five-component LNPs, subunit vaccines and PBS in Example 1D, respectively. The guinea pigs were scratched on the back on days 21 and 22 for the challenge experiment.
  • Example 1D The body weight of guinea pigs was monitored during the immunization period, see Figure 29. It was verified that the five-component LNP encapsulated with mVZV in Example 1D can effectively protect guinea pigs from VZV virus invasion.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Nanotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Optics & Photonics (AREA)
  • Biophysics (AREA)
  • Communicable Diseases (AREA)
  • Condensed Matter Physics & Semiconductors (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Medical Informatics (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • General Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Manufacturing & Machinery (AREA)
  • General Physics & Mathematics (AREA)
  • Genetics & Genomics (AREA)
  • Dermatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Preparation (AREA)

Abstract

L'invention concerne une nanoparticule lipidique pour administrer efficacement un médicament à base d'acide nucléique, un procédé de préparation et une utilisation associée. La nanoparticule lipidique comprend un vecteur et un acide nucléique encapsulé. Le vecteur comprend un lipide ionisable composite, un phospholipide auxiliaire, une substance de cholestérol et un lipide pégylé. L'acide nucléique comprend un ou plusieurs éléments parmi l'ARNm, l'ARN circulaire, l'ARNsi, le miARN, un acide nucléique antisens et un plasmide. Le lipide ionisable composite comprend un lipide ionisable ayant la capacité d'entrer efficacement dans des cellules, et un lipide ionisable avec une longue chaîne alkyle et la capacité de fusion membranaire efficace. Le lipide ionisable composite peut améliorer significativement l'efficacité d'expression d'ARNm de LNPs à quatre composants commerciaux au moyen d'un synergisme. La nanoparticule lipidique facilite l'expression efficace du médicament à base d'acide nucléique piégé dans celle-ci après administration intramusculaire, activant ainsi efficacement des réponses immunitaires humorales et à médiation cellulaire.
PCT/CN2023/131882 2022-11-23 2023-11-15 Procédé de préparation de nanoparticule lipidique pour administration efficace d'un médicament à base d'acide nucléique et utilisation associée WO2024109612A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211477861.X 2022-11-23
CN202211477861.XA CN118059061A (zh) 2022-11-23 2022-11-23 一种用于高效递送核酸药物的脂质纳米颗粒的制备方法和应用

Publications (1)

Publication Number Publication Date
WO2024109612A1 true WO2024109612A1 (fr) 2024-05-30

Family

ID=91096100

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/131882 WO2024109612A1 (fr) 2022-11-23 2023-11-15 Procédé de préparation de nanoparticule lipidique pour administration efficace d'un médicament à base d'acide nucléique et utilisation associée

Country Status (2)

Country Link
CN (1) CN118059061A (fr)
WO (1) WO2024109612A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111467321A (zh) * 2020-03-26 2020-07-31 深圳市新合生物医疗科技有限公司 一种mRNA核酸类药物胞内递送系统、制备方法和应用
CN114099699A (zh) * 2021-11-22 2022-03-01 华南理工大学 一种纳米递送系统及其制备方法和应用
CN114306279A (zh) * 2021-12-30 2022-04-12 复旦大学 基于科罗索酸或其类似物的脂质纳米颗粒系统及其制备方法和应用
WO2022207862A2 (fr) * 2021-03-31 2022-10-06 Curevac Ag Seringues contenant des compositions pharmaceutiques comprenant de l'arn
CN116763756A (zh) * 2022-10-11 2023-09-19 中国科学院化学研究所 一种脂质纳米颗粒及其制备方法和应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111467321A (zh) * 2020-03-26 2020-07-31 深圳市新合生物医疗科技有限公司 一种mRNA核酸类药物胞内递送系统、制备方法和应用
WO2022207862A2 (fr) * 2021-03-31 2022-10-06 Curevac Ag Seringues contenant des compositions pharmaceutiques comprenant de l'arn
CN114099699A (zh) * 2021-11-22 2022-03-01 华南理工大学 一种纳米递送系统及其制备方法和应用
CN114306279A (zh) * 2021-12-30 2022-04-12 复旦大学 基于科罗索酸或其类似物的脂质纳米颗粒系统及其制备方法和应用
CN116763756A (zh) * 2022-10-11 2023-09-19 中国科学院化学研究所 一种脂质纳米颗粒及其制备方法和应用

Also Published As

Publication number Publication date
CN118059061A (zh) 2024-05-24

Similar Documents

Publication Publication Date Title
JP4800485B2 (ja) ポリヌクレオチドベースのワクチンに対する免疫応答を増強するためのアジュバント組成物および方法
US20200016274A1 (en) Messenger rna vaccines and uses thereof
CN110974954A (zh) 一种用于增强核酸疫苗免疫效果的脂质纳米颗粒及其制备方法
CN107980004A (zh) 用于医治疾病的外泌体的用途
US20230226169A1 (en) Multilamellar rna nanoparticle vaccine against sars-cov-2
WO2024078193A1 (fr) Nanoparticule lipidique, son procédé de préparation et son utilisation
CN116133640A (zh) 脂质纳米颗粒
CN115103687A (zh) Hpv疫苗
WO2023147092A9 (fr) Vaccin anti-coronavirus
CN115252582A (zh) 红细胞膜杂合pH脂质体包被溶瘤病毒制剂的制备及应用
EP3866769A1 (fr) Nanotransporteurs pour une thérapie d'inflammation pulmonaire
Zhou et al. Single-injection subunit vaccine for rabies prevention using lentinan as adjuvant
Austin et al. Split-dose administration enhances immune responses elicited by a mRNA/lipid nanoparticle vaccine expressing respiratory syncytial virus F protein
WO2024109612A1 (fr) Procédé de préparation de nanoparticule lipidique pour administration efficace d'un médicament à base d'acide nucléique et utilisation associée
JP2023181377A (ja) 免疫賦活剤
US20040005302A1 (en) Encapsulated cells to elicit immune responses
Oh et al. Preclinical safety assessment of the suction-assisted intradermal injection of the SARS-CoV-2 DNA vaccine candidate pGO-1002 in white rabbit
Colombani et al. Harnessing the Potential of Biomaterials for COVID-19 Therapeutic Strategies
CN118146318B (zh) 一种腺相关病毒衣壳蛋白及其用途
US20050232906A1 (en) Adjuvant preparation for the induction of specific immunity
US20230302112A1 (en) Respiratory synctial virus rna vaccine
WO2024108955A1 (fr) Vaccin contre l'hépatite b
WO2023169500A1 (fr) Vaccin à arnm pour le codage de la protéine s du sars-cov-2
WO2023183945A1 (fr) Compositions et procédés pour vaccins adjuvantés
CN118574637A (zh) Rna分子

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: 23893702

Country of ref document: EP

Kind code of ref document: A1