CN116271066A - Recombinant adenovirus vector vaccine inhalation administration delivery system - Google Patents

Recombinant adenovirus vector vaccine inhalation administration delivery system Download PDF

Info

Publication number
CN116271066A
CN116271066A CN202211589718.XA CN202211589718A CN116271066A CN 116271066 A CN116271066 A CN 116271066A CN 202211589718 A CN202211589718 A CN 202211589718A CN 116271066 A CN116271066 A CN 116271066A
Authority
CN
China
Prior art keywords
adenovirus
albumin
vector vaccine
formulation according
abd
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.)
Pending
Application number
CN202211589718.XA
Other languages
Chinese (zh)
Inventor
邵娟
隋秀文
周朝东
曹龙龙
苏喆
苗伟
吴丹
魏搏超
徐方
司伟雪
齐源远
奚志嫒
朱涛
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.)
CanSino Biologics Inc
Original Assignee
CanSino Biologics Inc
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 CanSino Biologics Inc filed Critical CanSino Biologics Inc
Publication of CN116271066A publication Critical patent/CN116271066A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • 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
    • A61K39/215Coronaviridae, e.g. avian infectious bronchitis 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • 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
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • 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
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0078Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a nebulizer such as a jet nebulizer, ultrasonic nebulizer, e.g. in the form of aqueous drug solutions or dispersions
    • 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
    • A61K9/12Aerosols; Foams
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M11/00Sprayers or atomisers specially adapted for therapeutic purposes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • C07K14/08RNA viruses
    • C07K14/165Coronaviridae, e.g. avian infectious bronchitis virus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/315Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/861Adenoviral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/544Mucosal route to the airways
    • 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
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • 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)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Genetics & Genomics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Communicable Diseases (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pulmonology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Mycology (AREA)
  • Dispersion Chemistry (AREA)
  • Immunology (AREA)
  • Otolaryngology (AREA)
  • Physics & Mathematics (AREA)
  • Inorganic Chemistry (AREA)
  • Plant Pathology (AREA)
  • Anesthesiology (AREA)

Abstract

The invention relates to a recombinant adenovirus vector vaccine inhalation administration delivery system, which is characterized in that the delivery system is used for improving adenovirus vectors and matching with a specific preparation prescription, and the vaccine can reach the lung through nasal cavity or oral inhalation after inhalation administration, so that protective immune response including mucosal immunity is generated, the effective utilization rate of the vaccine is enhanced and the effect of the vaccine is improved while the pre-existing immunity of adenovirus is solved.

Description

Recombinant adenovirus vector vaccine inhalation administration delivery system
Technical Field
The invention relates to the technical field of vaccines, in particular to a recombinant adenovirus vector vaccine inhalation administration delivery system for solving adenovirus pre-existing immunity.
Background
Adenoviruses belong to the non-enveloped double-stranded DNA viruses, and human adenoviruses have now identified 53 serotypes. Adenovirus consists of an outer capsid protein and an inner core protein. The core protein comprises: protein v, protein vii, and protein x, which bind to the viral genome. The terminal protein TP is covalently bound to the 5' end of the viral DNA; surrounding the viral core is the viral capsid, which is an icosahedral symmetrical structure formed by the non-covalent interactions of 7 proteins (II, III, IIIa, IV, VI, VIII and IX), of which 240 trimeric hexons (protein II) are the main constituent proteins of the 20-face.
Adenovirus has a spherical structure without envelope, virus particles are arranged in lattice form in infected cell nucleus, each virus particle contains 36kb linear double-stranded DNA, two ends of each virus particle have 100-600 bp inverted terminal repeat sequences (inverted terminal re-peat, ITR), and the inner side of ITR is a virus packaging signal, which is a cis-acting element required by virus packaging. The genome comprises early expressed E1-E4 genes associated with adenovirus replication and late expressed L1-L5 genes associated with adenovirus particle assembly. The linear double-stranded DNA and the core protein form a marrow core with the diameter of 60-65 nm, and the marrow core is wrapped in a capsid. The capsid is icosahedral and consists of 252 capsomers with diameters of 8-10 nm, the capsomers are arranged on the triangular surface, 6 on each side, wherein 240 are hexons (non-vertex capsomers), and the other 12 are penton bases (vertex capsomers). Each hexon is a homotrimer of hexon protein, the hexon molecule of the trimer has a triangular cone tip and a pentahedral base, the cone region is composed of 4 rings, namely loop1, loop2, loop3 and loop4, and the base comprises two regions P1 and P2.
The current problem with adenovirus vector vaccines is the presence of pre-existing immunity (pre-existing immunity) to adenovirus, e.g., adenovirus type 5 is more common in the environment, and immune responses against adenovirus are readily generated in the human population, which would reduce the neutralizing antibody levels of adenovirus vector vaccines in humans. The existing method for solving the pre-existing immunity of adenovirus mainly comprises a chemical method and a genetic engineering method. Chemical methods mainly use PEG to encapsulate adenovirus and shield its epitope, thereby escaping the immune function of the host, but such methods have difficulty in obtaining high-titer viruses.
Genetic engineering methods are modification of adenovirus, e.g., construction of chimeric modified adenovirus capsids, construction of chimeric adenoviruses. Adenovirus-based vaccines are completely replaced with other rare, e.g., adenovirus serotypes from humans or non-humans (Chen H, xiang Z Q, li Y, et al adenoviruses-based vaccines: comparison of vectors from three species of adenoviridae [ J ]. Virol,2010,84 (20): 10522-10532), but the specific antibody response elicited by foreign genes is significantly lower than that elicited by type 5 adenovirus vector carrying.
Recombinant coronavirus vaccine (adenovirus type 5 vector) for inhalation developed by the biological stock company Kang Xinuo, 9, 2022, was suggested by the national health commission, and the national administration of pharmaceutical administration demonstrated consent for inclusion as an enhanced needle for emergency use.
Prior art (WO 2015166082A1, entitled adenovirus comprising an albumin binding moiety) adenoviruses genetically modified on the outer surface of the capsid, in particular on the outer surface of the adenovirus hexon protein, enable an albumin protection shield to be obtained, enabling the virus to evade neutralizing antibodies and to extend its retention time in the blood after systemic administration. Albumin is widely used as a drug carrier in the field of intensive research and development, thanks to its long half-life in plasma, and non-covalent binding with the drug to be carried does not allow rapid clearance of the drug complex in the blood. However, this current route of technology is administered by injection, and there is still a detrimental effect on the exertion of the potency of albumin-binding recombinant adenovirus vaccines due to the specific cd4+ and cd8+ T cells present in the infected adenovirus type 5.
Disclosure of Invention
The invention relates to an adenovirus vector vaccine preparation, which contains recombinant adenovirus and auxiliary components; the recombinant adenovirus vector backbone genome sequence comprises a sequence encoding an albumin binding domain; the adjunct component includes albumin. The vaccine can effectively escape the pre-existing immunity of adenovirus vectors in vivo.
The present invention adds specific doses of human albumin to the formulation of an inhalation vaccine formulation prior to vaccine delivery to ensure that a protective barrier has been formed prior to administration that effectively evades adenovirus neutralizing antibodies.
As an inhalation vaccine with Ad-ABD as a platform, it is not easy to form a protective shell of an effective anti-neutralizing antibody after the vaccine is delivered to the targeted organ. Furthermore, even an adenovirus vector vaccine with an albumin binding domain may be rapidly recognized and cleared by specific neutralizing antibodies before binding to albumin in vivo; or the specific neutralizing antibodies and albumin compete with each other for binding to the adenovirus vector, resulting in insufficient binding of albumin to the virus vector and subsequent failure to form a complete and effective pre-existing immune barrier.
Hexon is the highest protein content in the adenovirus capsid and is also considered the primary target for neutralizing antibodies. The loop 1-L1 and loop 2-L2 of the hexon protein are located outside the adenovirus capsid structure. Loop1 of hexon protein has 6 HVRs, (HVR 1-HVR 6). There are 7 hypervariable regions (HVRs) in the cone region of the hexon protein trimer structure, i.e., L2 contains the 7 th hypervariable region (HVR 7), comprising a specific epitope.
The sequence encoding an albumin binding domain is inserted in the coding region of HVR-1 of a hexon protein, the albumin binding domain being located on the outer surface of the hexon protein.
In particular, the albumin binding domain is selected from the albumin binding domain of streptococcal protein G, or a functionally equivalent variant thereof, preferably the albumin binding domain is albumin binding domain 3 of streptococcal protein G.
More preferably, the albumin binding domain 3 of streptococcal protein G has the sequence of SEQ ID NO:1.
SEQ ID NO. 1 sequence is as follows:
Cys Glu Trp Asp Glu Ala Ala Thr Ala Leu Glu Ile Asn Leu Glu Glu Glu
Asp Asp Asp Asn Glu Asp Glu Val Asp Glu Gln Ala Glu Gln Gln Lys Thr His Val
the inserted albumin binding encoding sequence is the binding domain of the fusion protein included after the D150 amino acid of the hexon protein.
The sequence of the hinge region between the hexon protein and the albumin binding domain is called a linker sequence and serves primarily to provide space between the two elements so that the Albumin Binding Domain (ABD) portion does not affect the secondary structure of the hexon protein. Since the connectors generally maintain the three-position structural length of the elements and allow the elements to be independent of each other.
The N-terminus and/or the C-terminus of the albumin binding domain is linked to the hexon protein by a linker sequence.
The linker sequence comprises the sequence GSGS, as shown in SEQ ID NO: 2.
In particular, the adenovirus is a human adenovirus.
Preferably, the human adenovirus is selected from one or more of the human adenoviruses of serotypes 1 to 57. More preferably, the human adenovirus is selected from the group consisting of human adenoviruses of serotypes 1 to 57; more preferably, the human adenovirus is a serotype 5, 26 or 35 human adenovirus.
Specifically, the albumin added in the auxiliary component is one or more of human serum albumin or recombinant human serum albumin or bovine serum albumin. Albumin may bind to the outer surface capsid region of the adenovirus hexon protein.
Specifically, the molar ratio of albumin to adenovirus vector is 1-10 5 :1, a step of; preferably, 1-240:1.
specifically, the vaccine is a mucosal or injectable formulation, preferably selected from: a liquid, solid, semi-solid, or gaseous dosage form; more preferably, the mucous membrane administration preparation is nasal drops, aerosol, spray, powder fog, powder, liquid preparation, freeze-dried preparation, gel, microsphere preparation, liposome, film preparation and suspension;
further, the mucosal delivery formulation is an inhalation liquid formulation, an inhalation aerosol, and an inhalation spray.
More preferably, nasal inhalation or oral inhalation; more preferably, the mucosa comprises nasal mucosa, oral mucosa or pulmonary mucosa.
Specifically, the auxiliary component also comprises pharmaceutically acceptable auxiliary materials. The pharmaceutically acceptable excipients include, but are not limited to: one or more of buffers, protectants, stabilizers, surfactants, tonicity adjusting agents, adjuvants, preservatives, inactivating agents and the like.
In particular, the buffering agent includes, but is not limited to, one or more of HEPES, HIS, TRIS, PB, succinic acid, citric acid; the protective agent includes, but is not limited to, one or more of gelatin, ethanol, diethylaminotetraacetic acid (EDTA), disodium diethylaminotetraacetic acid (EDTA-2 Na), and magnesium chloride; the stabilizers include, but are not limited to, one or more of sucrose, mannitol, fucose and maltose; the surfactant includes, but is not limited to, one or more of tween, span, glycerol; the osmolality adjusting agent includes, but is not limited to, sodium chloride or is omitted.
Specifically, the auxiliary material components comprise mannitol, sucrose, sodium chloride, magnesium chloride, HEPES, polysorbate 80 and glycerin, preferably 10-150mg/ml mannitol, 10-150mg/ml sucrose, 10-150mM sodium chloride, 1-10mM magnesium chloride and 1-15mM HEPES, wherein the polysorbate 80 is 0.001% -0.5% by weight percent and the glycerin is 0.05-2% by weight percent.
Specifically, the formulation components comprise: 10-50mg/ml of sucrose, 15-75mg/ml of mannitol, 40-60mM of sodium chloride, 0.5-5mg/ml of glycerin, 1-5mM of magnesium chloride, 0.05-0.5mg/ml of Tween 80, 1-5mM of HEPES and 0.1% -1% of HAS in percentage by weight.
Specifically, the dosage form is an atomized inhalant, and the vaccine forms particles below 10 mu m after being atomized by an atomized administration device.
In particular, the albumin is present in the vaccine in a content of 0.001% -50% by weight, preferably 0.005% or 0.01% or 0.05% or 0.1% or 0.2% or 0.3% or 0.4% or 0.5% or 0.6% or 0.7% or 0.8% or 0.9% or 1% or 2% or 3% or 4% or 5% or 6% or 7% or 8% or 9% or 10% or 15% or 20% or 25% or 30% or 35% or 40% or 45% or 50%.
Specifically, the adenovirus vector also comprises viral or bacterial antigen proteins. Such as HIV, rabies, dengue, ebola, coronavirus, human papilloma, hepatitis c, hepatitis b, rotavirus, measles, respiratory Syncytial Virus (RSV), herpes zoster (VZV), cytomegalovirus, herpes simplex virus type 2, epstein barr, influenza, trypanosoma cruzi and plasmodium falciparum, tubercle bacillus.
Specifically, the packaging cell of choice is a HEK293 cell.
The invention provides a preparation method of an adenovirus vector vaccine preparation, which comprises the following preparation steps:
s1, introducing Albumin Binding Domain (ABD) of Streptococcus (Streptococcus) protein G into HVR region of adenovirus vector skeleton hexon gene to construct adenovirus-ABD vector;
s2, introducing a virus or bacterial antigen protein gene sequence into the modified adenovirus-ABD vector to construct an Ad-ABD vector vaccine;
s3, adding albumin as an auxiliary component into the preparation.
Specifically, the packaging cell of choice is a HEK293 cell.
The invention provides a drug delivery system of an adenovirus vector vaccine preparation, which is aerosol inhalation drug delivery, and the aerosol drug delivery device is a jet aerosol drug delivery device or an ultrasonic aerosol drug delivery device or a vibration aerosol drug delivery device.
Preferably, the aerosolized drug delivery device comprises an atomizer and an atomized vaccine particle collection member.
The invention provides an application of an adenovirus vector vaccine preparation in preparing a vaccine for preventing infectious diseases. Specifically, adenoviruses harbor viral or bacterial disease antigens.
In particular, the viral or bacterial antigens entrapped therein are one or more of HIV, rabies, dengue, ebola, coronavirus, human papilloma, hepatitis c, hepatitis b, rotavirus, measles, respiratory Syncytial Virus (RSV), herpes zoster (VZV), cytomegalovirus, herpes simplex type 2, epstein barr, influenza, trypanosoma cruzi and plasmodium falciparum, tubercle bacillus.
The present invention provides a method of preventing an infectious disease comprising administering an adenovirus vector vaccine to a subject.
The subject may be a human or mammal or a cell, tissue or organ thereof.
The beneficial effects of the invention include:
the invention provides a vaccine inhalation administration system through experimental verification, so that vaccine is administered in an aerosol inhalation mode, which can activate protective immune response including mucosal immunity, and simultaneously solve the problem of preexisting immunity of rAD, and through experimental verification, the influence of the preexisting immunity of Ad on vaccine immunogenicity can be effectively reduced
In order to coordinate with the transformation of adenovirus, the invention adds the matching component albumin into the preparation, and better combines with the adenovirus containing albumin binding domain to form an albumin coated capsid so as to avoid the pre-existing immunity. By inhalation administration, the problem that the independently administered ABD modified adenovirus cannot effectively stimulate humoral immunity and cellular immunity is solved, and mucous membrane immunity can be activated simultaneously to obtain higher titer.
Drawings
FIG. 1 is a schematic diagram showing the molecular design of a recombinant Ad-ABD vector;
FIG. 2Ad and Ad-ABD binding to HSA and BSA-ELISA results (noted with no corresponding histogram, i.e., undetectable);
FIG. 3 in vitro neutralization assay-quantitative assay IFU calculation;
FIG. 4 day 14 neutralizing titer of antigen-binding antibodies (1 og 10);
FIG. 5 day 28 neutralizing titer of anti-prototype binding antibodies (1 og 10);
FIG. 6 day 42 neutralizing titer of antigen-binding antibodies (1 og 10);
FIG. 7 anti-S-IgG antibody titers;
FIG. 8CD4+T cell immunization results;
FIG. 9CD8+T cell immunization results.
Detailed Description
Unless defined otherwise, all scientific and technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention relates.
The "adenovirus vector vaccine" refers to a vaccine prepared by recombining a target antigen gene into an adenovirus genome using adenovirus as a vector, and using a recombinant adenovirus capable of expressing the antigen gene. The technical solutions of the present invention will be clearly and completely described in conjunction with the embodiments of the present invention, and it is apparent that the described embodiments are only some embodiments of the present invention, but not all embodiments. All other embodiments, which can be made by those skilled in the art based on the embodiments of the invention without making any inventive effort, are intended to be within the scope of the invention.
Example 1: preparation of Ad-ABD adenovirus
Recombinant novel coronavirus vaccine using replication-defective adenovirus as vector was selected as evaluation model:
the target antigen of the recombinant novel coronavirus vaccine is the S protein of a novel coronavirus strain (Genebank number: NC_ 045512.2);
(1) Constructing a shuttle plasmid vector containing a polynucleotide encoding a novel coronavirus S protein;
backbone plasmid: the schematic of the molecular design of the recombinant Ad-ABD vector is shown in FIG. 1: using the prior art, an albumin binding domain (SEQ ID NO: 1) with two linkers (sequence: GSGS) at the flanking ends was inserted into HVR1 of adenovirus hexon (after D150 amino acids);
(2) Transfecting the shuttle plasmid vector of step (1) together with a backbone plasmid into a host cell HEK293;
(3) Culturing the host cell HEK293 of step (2);
(4) Harvesting the human replication-defective recombinant adenovirus (adenovirus type 5 vector) released from the cells of step (3);
(5) Performing amplification culture on the recombinant adenovirus in the step (4);
(6) Purifying the culture product in the step (5) to obtain the Ad capable of expressing the target gene.
HEK293 cells were cultured in medium and passaged. When HEK293 cell density is > 1.5X10 6 At cells/ml, virus amplification was performed by dilution with medium, and the multiplicity of virus infection (MOI) was 1,5 and 10, 37℃and 5% CO, respectively 2 Culturing at 125rpm, and harvesting for 42-48 h to obtain adenovirus seed harvest liquid. The titer IFU was measured by the Ad5-ABD virus harvest and supernatant from HEK293 cells using the Ad-FITC fluorescent antibody method. The results are shown in Table 1.
TABLE 1Ad-ABD adenovirus preparation Virus titre detection results
Figure BDA0003993537740000071
The adenovirus strain Ad-ABD harvest liquid is processed in HEK293 SF-3F6 cell density of 0.8-1.2X10 6 cells/ml, moi=3, amplified, harvested 48h later.
Example 2: ELISA detection of binding of Ad-ABD adenovirus to albumin
(1) Coating: bovine Serum Albumin (BSA) and Human Serum Albumin (HSA) were diluted to 2mg/ml with coating solution, added to 96-well ELISA plates and coated overnight at 2-8deg.C. Washing, and adding a sealing liquid.
(2) Sample adding: to the Ad-ABD stock solution in example 1, NCVA stock solution was diluted 10-fold and 100-fold respectively and added to the ELISA plate; control wells were added with PBS. After incubation for 1h at 37℃the plates were washed.
(3) Incubation: 1% BSA/PBST 1: adenovirus monoclonal antibody (invitrogen) at 1000 dilution, 100 μl/well, incubated at 37 ℃ for 1h and plate washed; 1% BSA/PBST 1: sheep anti-mice IgG (Proteintech) were diluted 1000, incubated at 37℃for 30min and plates were washed.
(4) And (3) measuring: after the color development liquid returns to the room temperature, the solution A and the solution B are 1:1, adding an enzyme label plate, developing color for 15min, and reading by an enzyme label instrument.
The result of the attached figure 2 shows that the binding capacity of the Ad-ABD stock solution and HSA of the same dilution is larger than that of the Ad5 stock solution and HSA; with dilution of the virus stock, the binding capacity to HSA gradually decreases. Ad-ABD adenovirus can bind to HSA in vitro, demonstrating the feasibility of Ad-ABD adenovirus vector engineering and binding by the addition of albumin.
Example 3: in vitro neutralization experiment-qualitative experiment for escape of Ad-ABD adenovirus from preexisting immunity
(1) Virus samples and antibody dilution: ad-ABD stock and NCVA stock were diluted to a virus titer of 1.5X10 with DMEM complete culture (DMEM+10% FBS+1% P/S) and DMEM complete medium+1 mg/ml HSA, respectively 6 IFU/ml. Ad5 rabbit polyclonal antibody (Source Kang Xinuo Biotechnology Co.) was diluted with DMEM complete medium, 10-fold and 100-fold antibody dilutions, respectively.
(2) Incubation: 100. Mu.L/well virus sample and 1. Mu.L diluted antibody were added to 96-well plates and incubated at 37℃for 1h, 4 replicates for each experimental group, and the experimental groups are shown in Table 2:
TABLE 2 in vitro neutralization assay-qualitative analysis grouping
Figure BDA0003993537740000081
Figure BDA0003993537740000091
(3) Preparation of HEK293 cell suspension: HEK293 cells were diluted to a cell density of 3X 10 with DMEM complete medium 5 cells/ml. After incubation of virus samples and diluted Ad rabbit polyclonal antibody for 1h at 37 ℃, 100. Mu.L HEK293 cells/well are added into a 96-well plate, and after uniform mixing, the mixture is incubated at 37 ℃ with 5% CO 2 Culturing for 4-5h. The plate was washed 3 times with PBS,
(4) Antibody incubation: ad-FITC fluorescent antibody was diluted with 1% BSA/1 XPBS, dilution ratio 1:500; after washing the plate, 50. Mu.L/well diluted fluorescent antibody was added to the 96-well plate, and after incubation for 1h at room temperature in the dark, the plate was washed 2 times with PBS.
(5) Color development: cells were observed under a fluorescence microscope.
It can be seen from figure 3 that Ad or Ad-ABD virus vaccine stock was incubated with serial dilutions of anti-Ad antibodies in the presence of HSA, and then HEK293 cells were infected. The extent of neutralization of Ad-ABD was lower than that of Ad vectors, indicating that ABD modification with the addition of albumin in the formulation could evade pre-immunization.
Example 4 in vivo immunogenicity evaluation-antibody level test
The experimental object: female BALB/c mice of 6-8 weeks of age, 8/group.
Establishment of pre-stored immunity: guinea pig blood thawed to a neutralization titer of 3000 against adenovirus was taken and 50ul per mouse was intraperitoneally injected. After 24h, the serum was tested for neutralizing antibodies against adenovirus type 5 vectors, and the neutralizing antibody titer was between 200-1000 in all mice.
NCVA group: the recombinant novel coronavirus vaccine (Ad) prepared in example 1 without ABD modification;
ad-abd+hsa group: the novel coronavirus vaccine stock solution of Ad-ABD prepared in example 1 is added with a Human Serum Albumin (HSA) solution, and the final concentration of HSA is 1mg/ml;
Ad-ABD+BSA group: adding Bovine Serum Albumin (BSA) solution into the novel coronavirus vaccine stock solution of Ad-ABD prepared in example 1, wherein the final concentration of BSA is 1mg/ml;
Ad-ABD group: new coronavirus vaccine stock of Ad-ABD prepared in example 1.
Mode of mucosal inoculation: immunization by inhalation using inhalation type exposure tower and aerosol inhalation immunization, wherein the immunization dose is 5×10 9 VP。
Blood is taken from the eyesockets 14 days and 28 days after atomization inhalation immunization, and neutralization titer of the serum sample aiming at novel coronavirus S strain is detected, and the detection results are shown in tables 3-5 and figures 4-5;
table 3 neutralizing titres of binding antibodies for 14 days (1 og 10)
Group of Rat 1 Rat 2 Mouse 3 Mouse 4 Mouse 5 Mouse 6 Mouse 7 Mouse 8
NCVA 5.51 5.19 5.81 5.74 5.51 5.52 5.20 5.01
Ad-ABD+BSA 5.86 5.72 5.72 5.66 5.99 5.39 5.87 6.10
Ad-ABD+HSA 5.89 5.81 5.89 6.51 5.81 6.11 6.32 6.11
Ad-ABD 4.02 4.60 4.00 5.20 4.60 4.60 4.60 4.90
Table 4 neutralizing titres of 28 days binding antibody (1 og 10)
Group of Rat 1 Rat 2 Mouse 3 Mouse 4 Mouse 5 Mouse 6 Mouse 7 Mouse 8
NCVA 3.90 3.82 4.20 3.99 4.46 4.58 4.24 3.90
Ad-ABD+BSA 4.90 4.17 4.83 4.09 5.15 4.90 4.22 4.80
Ad-ABD+HSA 5.22 5.11 4.81 4.20 4.81 4.93 5.75 4.97
Ad-ABD 3.60 4.20 3.90 3.81 4.81 3.76 3.54 3.30
Table 5 neutralizing titres of bound antibody for 42 days (1 og 10)
Group of Rat 1 Rat 2 Mouse 3 Mouse 4 Mouse 5 Mouse 6 Mouse 7 Mouse 8
NCVA 4.60 4.21 5.12 5.24 5.17 4.90 4.89 4.33
Ad-ABD+BSA 5.52 5.97 5.81 5.51 5.87 6.11 5.98 5.58
Ad-ABD+HSA 5.98 5.99 5.81 5.74 5.90 6.21 6.01 6.11
Ad-ABD 4.07 4.60 4.34 5.20 4.02 4.58 4.13 3.93
Performing a second immunization 28 days after the first immunization, and performing deratization and evaluation of the cellular immunity level and the total antibody titer of S after 14 days of the second immunization, wherein the detection result is shown in FIG. 6;
the experimental results show that: the experiment was performed at 2 doses and after 14 days of immunization, the total antibody level specific for S protein was detected, and the results showed that the level of antibody elicited by the Ad-ABD vector was lower than that elicited by the Ad without ABD modification by inhalation.
The possible reasons are: ABD engineered Ad, based on a specific inhaled administration regimen, while having reduced sensitivity to pre-existing adenovirus antibodies in vivo, also results in reduced host infection efficiency. While the positive effects of evading pre-existing immunity cannot counteract the negative effects of reduced immunostimulatory capacity. The antibody levels of the experimental groups Ad-ABD+HSA and Ad-ABD+BSA are obviously higher than those of other groups, namely, the technical scheme that albumin is added in advance in the preparation and then the albumin is combined with the Ad-ABD for co-inhalation administration can greatly improve the positive influence of escaping pre-existing immunity. As a result, it is possible that pulmonary albumin is difficult to bind to Ad containing ABD domains when Ad is delivered by inhalation administration, unlike in vitro and in an injection environment, and thus the desired escape from preexisting immunity is not achieved, and even the effect is inferior to that of unmodified Ad, and thus the drug cannot be formulated (mucosal administration). The invention fundamentally solves the problem of pharmacy of the Ad-ABD mucous membrane administration preparation for avoiding pre-existing immunity by adding albumin into the preparation, namely, by re-inhalation administration in a mode of in vitro pre-preparation combination.
Example 5 in vivo immunogenicity evaluation-cellular immune response.
The experimental object: female BALB/c mice of 6-8 weeks of age, 8/group.
The experimental method comprises the following steps: the same experimental procedure as in example 4 was used.
And (3) detection: at 21 days post immunization, orbital blood was taken and tested for S-IgG antibodies, the results are shown in Table 6; splenocytes were harvested and used as antigen-specific CD4 and CD 8T cell immune responses.
TABLE 6S-IgG antibody titre detection results
Figure BDA0003993537740000111
TNFα, IFNγ, IL2, IL5 were detected by flow cytometry using Anti-TNFα -FITC antibody, anti-IFNγ -PE, mo μse antibody, anti-IL2-APC, mo μse antibody, anti-IL5-AF700 antibody (tables 7-14).
TABLE 7CD4+T cell TNF alpha immune response assay results
Figure BDA0003993537740000112
TABLE 8CD4+T cell IFN gamma immune response detection results
Figure BDA0003993537740000113
TABLE 9CD4+T cell IL2 immune response test results
Figure BDA0003993537740000114
TABLE 10CD4+T cell IL5 immune response assay results
Figure BDA0003993537740000121
TABLE 11CD8+T cell TNF alpha immune response assay results
Figure BDA0003993537740000122
TABLE 12CD8+T cell IFN gamma immune response assay results
Figure BDA0003993537740000123
Table 13CD8+T cell IL2 immune response test results
Figure BDA0003993537740000124
TABLE 14CD8+T cell IL5 immune response assay results
Figure BDA0003993537740000125
The experimental results are shown in FIGS. 7-9.
The experimental results show that: mucosal immunization by inhalation (mucosal administration), NCVA group, ad-ABD+HSA group, ad-ABD+BSA group, ad-ABD group vectors induced S-IgG antibody titers in mouse serum. Can excite humoral immunity and cellular immunity in mice, mainly uses CD8+T cells, but the cellular immunity level of Ad-ABD group is obviously lower than that of control NCVA. The technical scheme of pre-adding albumin in the Ad-ABD preparation and then combining with Ad-ABD for co-inhalation can induce mice to generate the COVID-19S proteins THF alpha, IFN gamma and IL2 response level higher than NCVA group and Ad-ABD group for CD4+ T cells and CD8+ T cells. The trend of the results is consistent with the experimental results of 'in vivo immunogenicity evaluation-antibody level test'.
The foregoing description of the preferred embodiments of the invention is not intended to be limiting, but rather is to be construed as including any modifications, equivalents, and alternatives falling within the spirit and principles of the invention.
The foregoing embodiments and methods described in this invention may vary based on the capabilities, experience, and preferences of those skilled in the art.
The listing of the steps of a method in a certain order in the present invention does not constitute any limitation on the order of the steps of the method.

Claims (18)

1. An adenovirus vector vaccine formulation comprising a recombinant adenovirus and a helper component; the recombinant adenovirus vector backbone genome sequence comprises a sequence encoding an albumin binding domain; the adjunct component includes albumin.
2. The adenoviral vector vaccine formulation according to claim 1, wherein the adenovirus is a human adenovirus; preferably, the human adenovirus is selected from one or more of the human adenoviruses of serotypes 1 to 57; more preferably, the human adenovirus is a human adenovirus of serotype 5, 26 or 35.
3. The adenoviral vector vaccine formulation according to any one of claims 1-2, wherein the albumin added to the auxiliary component is one or more of human serum albumin, recombinant human serum albumin, bovine serum albumin.
4. An adenoviral vector vaccine preparation according to any one of claims 1-3, wherein the molar ratio of albumin to adenoviral vector is 1-10 5 :1, a step of; preferably, 1-240:1.
5. the adenoviral vector vaccine formulation according to any one of claims 1-4, wherein the sequence encoding the albumin binding domain is inserted in the coding region of hypervariable region 1 (HVR 1) of the hexon protein, said albumin binding domain being located on the outer surface of the hexon protein.
6. The adenoviral vector vaccine formulation according to any one of claims 1-5, wherein the albumin binding domain is the albumin binding domain of streptococcal protein G or a functionally equivalent variant thereof, preferably the albumin binding domain is albumin binding domain 3 of streptococcal protein G.
7. The adenoviral vector vaccine formulation according to any one of claims 1-6, wherein the vaccine is a mucosal or injectable formulation, preferably the mucosal formulation is selected from the group consisting of: a liquid, solid, semi-solid, or gaseous dosage form; more preferably, the mucous membrane administration preparation is nasal drops, aerosol, spray, powder fog, powder, liquid preparation, freeze-dried preparation, gel, microsphere, liposome, film agent and suspension; further preferably, the mucosally administered formulation is an inhalation liquid formulation, an inhalation aerosol, and an inhalation spray; more preferably, nasal inhalation or oral inhalation; more preferably, the mucosa comprises nasal mucosa, oral mucosa or pulmonary mucosa.
8. The adenoviral vector vaccine formulation according to any one of claims 1-7, wherein the adjunct component further comprises pharmaceutically acceptable excipients; preferably, the pharmaceutically acceptable excipients include, but are not limited to: one or more of buffers, protectants, stabilizers, surfactants, tonicity adjusting agents, adjuvants, preservatives, inactivating agents and the like.
9. The adenoviral vector vaccine formulation according to any one of claims 1-8, wherein the buffer comprises, but is not limited to, one or more of HEPES, HIS, TRIS, PB, succinic acid, citric acid; the protective agent includes, but is not limited to, one or more of gelatin, ethanol, diethylaminotetraacetic acid (EDTA), disodium diethylaminotetraacetic acid (EDTA-2 Na), and magnesium chloride; the stabilizers include, but are not limited to, one or more of sucrose, mannitol, fucose and maltose; the surfactant includes, but is not limited to, one or more of tween, span, glycerol; the osmolality adjusting agent includes, but is not limited to, sodium chloride or is omitted.
10. Adenovirus vector vaccine formulation according to any one of claims 1-9, wherein the adjuvant component comprises mannitol, sucrose, sodium chloride, magnesium chloride, HEPES, polysorbate 80, glycerol, preferably mannitol 10-150mg/ml, sucrose 10-150mg/ml, sodium chloride 10-150mM, magnesium chloride 1-10mM, HEPES 1-15mM, polysorbate 80.001% -0.5% in weight percent, glycerol 0.05-2% in weight percent.
11. The adenoviral vector vaccine formulation according to any one of claims 1-10, wherein the formulation components comprise: 10-50mg/ml of sucrose, 15-75mg/ml of mannitol, 40-60mM of sodium chloride, 0.5-5mg/ml of glycerin, 1-5mM of magnesium chloride, 0.05-0.5mg/ml of Tween 80, 1-5mM of HEPES and 0.1% -1% of HAS in percentage by weight.
12. The adenoviral vector vaccine formulation according to any one of claims 1-11, wherein the formulation is an aerosol inhalation, the vaccine forming particles below 10 μm after aerosol administration by an aerosol administration device.
13. Adenovirus vector vaccine formulation according to any one of claims 1-12, wherein the albumin is present in the vaccine in an amount of 0.001% -50%, preferably 0.005% or 0.01% or 0.05% or 0.1% or 0.2% or 0.3% or 0.4% or 0.5% or 0.6% or 0.7% or 0.8% or 0.9% or 1% or 2% or 3% or 4% or 5% or 6% or 7% or 8% or 9% or 10% or 15% or 20% or 25% or 30% or 35% or 40% or 45% or 50% by weight.
14. A method of preparing an adenovirus vector vaccine formulation according to any one of claims 1-13, comprising the steps of:
s1, introducing Albumin Binding Domain (ABD) of Streptococcus (Streptococcus) protein G into HVR region of adenovirus vector skeleton hexon gene to construct adenovirus-ABD vector;
s2, introducing a virus or bacterial antigen protein gene sequence into the modified adenovirus-ABD vector to construct an Ad-ABD vector vaccine;
s3, adding albumin as an auxiliary component into the preparation.
15. The method of claim 14, wherein the selected packaging cells are HEK293 cells.
16. The method of claim 14 or 15, wherein the viral or bacterial antigen is one or more of HIV, rabies, dengue, ebola, coronavirus, human papilloma, hepatitis c, hepatitis b, rotavirus, measles, respiratory Syncytial Virus (RSV), herpes zoster (VZV), cytomegalovirus, herpes simplex virus type 2, epstein barr, influenza, trypanosoma cruzi and plasmodium falciparum, tubercle bacillus.
17. A delivery system for an adenovirus vector vaccine formulation according to any one of claims 1-13, wherein the delivery system is an aerosol inhalation delivery, the aerosol delivery device being a jet aerosol delivery device or an ultrasonic aerosol delivery device or a vibratory aerosol delivery device, preferably the aerosol delivery device comprising an atomizer and an atomized vaccine particle collection means.
18. Use of an adenoviral vector vaccine formulation according to any one of claims 1-13 in the manufacture of a vaccine for preventing infectious diseases.
CN202211589718.XA 2021-12-13 2022-12-12 Recombinant adenovirus vector vaccine inhalation administration delivery system Pending CN116271066A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021115157890 2021-12-13
CN202111515789 2021-12-13

Publications (1)

Publication Number Publication Date
CN116271066A true CN116271066A (en) 2023-06-23

Family

ID=86774905

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211589718.XA Pending CN116271066A (en) 2021-12-13 2022-12-12 Recombinant adenovirus vector vaccine inhalation administration delivery system

Country Status (2)

Country Link
CN (1) CN116271066A (en)
WO (1) WO2023109740A1 (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2940128A1 (en) * 2014-04-30 2015-11-04 Institut d'Investigació Biomèdica de Bellvitge (IDIBELL) Adenovirus comprising an albumin-binding moiety
CN112451811A (en) * 2014-10-13 2021-03-09 欧米伽生命科学公司 Atomizer and use thereof
KR102601875B1 (en) * 2020-03-25 2023-11-14 (주)큐리진 Immunologically cloaking anti-tumor adenovirus
CN113750227A (en) * 2020-06-01 2021-12-07 康希诺生物股份公司 SARS-CoV-2 vaccine

Also Published As

Publication number Publication date
WO2023109740A1 (en) 2023-06-22

Similar Documents

Publication Publication Date Title
Funk et al. A snapshot of the global race for vaccines targeting SARS-CoV-2 and the COVID-19 pandemic
Batty et al. Vaccine formulations in clinical development for the prevention of severe acute respiratory syndrome coronavirus 2 infection
AU2018357912B2 (en) Adenovirus and uses thereof
Vu et al. Current and future nanoparticle vaccines for COVID-19
CA3167833A1 (en) Coronavirus immunogenic compositions and uses thereof
WO2022077593A1 (en) Sars-cov-2 coronavirus vaccine and preparation method therefor
Clark et al. Bacterial viruses as human vaccines?
Deal et al. Prospects for oral replicating adenovirus-vectored vaccines
US11931410B1 (en) SARS-CoV-2 mRNA vaccine and preparation method and use thereof
Graham et al. Mucosal delivery of human papillomavirus pseudovirus-encapsidated plasmids improves the potency of DNA vaccination
Huang et al. SARS-CoV-2 virus: vaccines in development
JP2015051018A (en) Chimeric adenovirus capsid proteins
Smith et al. Assembly of trans-encapsidated recombinant viral vectors engineered from Tobacco mosaic virus and Semliki Forest virus and their evaluation as immunogens
Feng et al. Shell-mediated phagocytosis to reshape viral-vectored vaccine-induced immunity
Narayanan et al. Recombinant helical plant virus-based nanoparticles for vaccination and immunotherapy
Luo et al. A self-biomineralized novel adenovirus vectored COVID-19 vaccine for boosting immunization of mice
Dowaidar Gene-free Viral-like particles (VLPs) offer a safer alternative to inactivating or weakening viral strains for traditional vaccines
WO2006028214A1 (en) Vaccine for oral administration
WO2022077591A1 (en) Use of recombinant adenovirus in preparation of drugs for prevention of virus
CN116271066A (en) Recombinant adenovirus vector vaccine inhalation administration delivery system
Sarangi et al. Success of nano-vaccines against COVID-19: a transformation in nanomedicine
Mitra New generation vaccine strategies against COVID-19
WO2023142283A1 (en) Sars-cov-2 mrna vaccine, and preparation method therefor and use thereof
So et al. Combined biolistic and cell penetrating peptide delivery for the development of scalable intradermal DNA vaccines
JP4535874B2 (en) Bacteriophage-mediated immunity against hepatitis

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