CN114213548A - Method for simultaneously inducing immune response against multiple viruses - Google Patents

Method for simultaneously inducing immune response against multiple viruses Download PDF

Info

Publication number
CN114213548A
CN114213548A CN202111597361.5A CN202111597361A CN114213548A CN 114213548 A CN114213548 A CN 114213548A CN 202111597361 A CN202111597361 A CN 202111597361A CN 114213548 A CN114213548 A CN 114213548A
Authority
CN
China
Prior art keywords
leu
virus
asn
gly
ser
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
CN202111597361.5A
Other languages
Chinese (zh)
Inventor
徐建青
张晓燕
周东明
曹康丽
王祥
胡杨洋
丁龙飞
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.)
SHANGHAI PUBLIC HEALTH CLINICAL CENTER
Original Assignee
SHANGHAI PUBLIC HEALTH CLINICAL CENTER
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 SHANGHAI PUBLIC HEALTH CLINICAL CENTER filed Critical SHANGHAI PUBLIC HEALTH CLINICAL CENTER
Publication of CN114213548A publication Critical patent/CN114213548A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • 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
    • 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
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • 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
    • A61K2039/525Virus
    • A61K2039/5256Virus expressing foreign proteins
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • C07K2319/42Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation containing a HA(hemagglutinin)-tag
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use 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
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Mycology (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pulmonology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present application relates to methods of simultaneously inducing immune responses against multiple viruses (e.g., influenza virus as well as new corona virus). In particular, the present application provides an immunogenic peptide comprising (a) an immunogenic stem: it comprises the HA2 region of influenza virus hemagglutinin HA; (b) immunogen crown: comprising a viral membrane protein or an immunogenic fragment thereof, wherein the viral membrane protein is derived from a virus other than the virus from which the HA2 region of (a) was derived; and (c) optionally, a further moiety fused to the aforementioned moiety. The vaccine of the application is safe, can continuously generate high-titer neutralizing antibodies and T cell responses, and can be used for preventing and treating influenza and new coronavirus.

Description

Method for simultaneously inducing immune response against multiple viruses
Technical Field
The present invention relates to the field of vaccines, and in particular to a method for simultaneously inducing an immune response against a plurality of viruses including influenza, such as coronaviruses, and in particular neocoronaviruses.
Background
Influenza, called influenza for short, is an acute respiratory infectious disease caused by influenza virus, and is mainly divided into seasonal and pandemic influenza. It is estimated that the adult accounts for 5-10% and the child accounts for 20-30% of the influenza infection patients all over the world. Influenza not only causes a severe disease burden, but also causes a huge direct and indirect economic burden.
The influenza virus genome is divided into 8 segments, and 10 proteins such as Hemagglutinin (HA), Neuraminidase (NA), matrix proteins 1 and 2(M1 and M2), nonstructural proteins 1 and 2(NS1 and NS2), Nucleocapsid Protein (NP), and three polymerase complexes (PB1, PB2, and PA) are encoded respectively. Influenza viruses can be classified into influenza A, influenza B and influenza C types, for example, Avian Influenza Virus (AIV) is influenza A virus (influenza A virus), based on the antigenic difference of nucleoprotein, M1 and M2. Influenza a can be classified into 18H subtypes and 9N subtypes according to antigenicity differences of HA and NA. These 18H subtypes can be classified into group I (H1, H2, H5, H6, H8, H9, H11, H12, H13, H16, H17, H18) and group II (H3, H4, H7, H10, H14, H15). Among the subtypes known to directly infect humans are subtypes H1, H2, H3, H5, H7, H9, and H10, and viruses causing highly pathogenic avian influenza are all subtypes H5 and H7.
Most protective antibodies raised against influenza virus target the HA protein, a trimeric surface glycoprotein consisting of HA1 and HA2 domains. Most of the HA1 chains formed a spherical crown comprising the receptor binding site, while HA2 formed a stem, supporting HA1 and formed a trimer. Although the HA crown (HA1) HAs significant immune advantages in inducing the production of neutralizing antibodies, it is highly susceptible to mutation by itself, leading to antigenic drift and antigenic variation of the virus, forming a new influenza virus subtype. The HA2 region is more conserved than HA1, and 3GBN of currently isolated neutralizing antibodies against H7N9 HA2 HAs broad protection against influenza a, 3SDY HAs broad protection against influenza a group II, and 4FQV HAs broad protection against influenza a and b. Therefore, HA2 of H7N9 is crucial as an influenza neutralizing antibody that induces a broad spectrum.
At present, although some vaccines have entered clinical trials, marketing, production and use of vaccines remain an urgent challenge.
Coronaviruses comprise four structural proteins, including the spike protein (S protein), the envelope protein, the membrane protein, and the nucleocapsid protein. Among them, the S protein plays the most important role in the attachment, fusion and entry processes of viruses, and is also a main target of antibodies, entry inhibitors and vaccines. The S protein mediates entry of the virus into the host cell by first binding to the host receptor via the Receptor Binding Domain (RBD) of the S1 subunit, and then fusing the virus and host cell membranes via the S2 subunit. According to WHO report: animals immunized with coronavirus vaccines may develop more severe symptoms when exposed to live virus again. Non-neutralizing antibodies or lower antibody levels produced by vaccine immunization may cause antibody-dependent enhancement (ADE) effects that enhance viral pathogenicity. Therefore, to reduce the side effects of ADE, the RBD region in the S protein of the novel coronavirus would be the most effective target for vaccine development.
Autumn and winter are high-incidence seasons of respiratory diseases, including influenza virus infection, and how to prevent the risk of overlapping epidemic of influenza and respiratory infectious diseases such as new crown becomes important. In the prevention of infectious diseases, vaccines have been the most effective and economical measure since the advent, and most of the currently used preventive vaccines are aimed at activating neutralizing antibodies. Therefore, designing an immunogen that can simultaneously activate neutralizing antibodies against multiple influenza viruses or against influenza viruses and other viruses (such as new coronaviruses) at the same time is a direction we need to explore and develop urgently.
Disclosure of Invention
Immunogenic peptides capable of simultaneously inducing immune responses against a variety of viruses, including influenza viruses, their encoding nucleotide molecules, vectors, host cells, vaccines and uses thereof are provided in the present disclosure.
In one aspect of the present disclosure, an immunogenic peptide is provided comprising the following moieties:
(a) immunogen stem: it comprises the HA2 region of influenza virus hemagglutinin HA;
(b) immunogen crown: comprising a viral membrane protein or an immunogenic fragment thereof, wherein the viral membrane protein is derived from a virus other than the virus from which the HA2 region of (a) was derived;
(c) optionally, other moieties attached to the foregoing moieties.
In some embodiments, the HA2 region is derived from a source selected from the group consisting of: any of H1 to H18, in particular from the widely prevalent human influenza H1, H2, H3 and the multiple-appearing human infections with avian influenza H5 and H7, for example from 2009 prevalent H1(H1N1) and 2013 prevalent H7(H7N 9).
In some embodiments, the amino acid sequence of the HA2 region is set forth in SEQ ID NO. 1, or is encoded by a nucleotide molecule having the sequence set forth in SEQ ID NO. 2.
In some embodiments, the viral membrane protein is an immunogenic membrane protein.
In some embodiments, the source of the immunogen crown is selected from the group consisting of: an influenza virus of a different origin from the HA2 region in (a); other viruses than influenza virus.
In some embodiments, the source of the immunogen crown is selected from the group consisting of: coronavirus (such as neocoronavirus), hiv, influenza virus (such as the HA1 region of influenza virus hemagglutinin HA), rabies virus, hog cholera virus, porcine reproductive and respiratory syndrome virus, measles virus, ebola virus, herpes virus, arbovirus (such as zika virus, epidemic encephalitis b virus, forest encephalitis virus, dengue virus, hantavirus, hemorrhagic fever virus).
In some embodiments, the source of the immunogen crown is selected from the group consisting of: coronavirus SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-229E, HCoV-OC43, HCoV-NL63, HCoV-HKU1, bat-CoV, e.g., an S1 protein comprising the receptor binding domain of a coronavirus, or a Receptor Binding Domain (RBD), or an engineered receptor binding domain (e.g., the RBD region modified with a terminal cysteine to form an sRBD region), or an immunogenic fragment thereof.
In some embodiments, the immunogenic crown is selected from S1 of coronavirus SARS-CoV-2, particularly the RBD region from S1 (e.g., the sequence shown in SEQ ID NO:3 or encoded by the nucleotide molecule of SEQ ID NO: 4) or a modified RBD region thereof (e.g., the terminal Cys-modified sRBD region, the peptide stretch shown in SEQ ID NO:5 or encoded by the nucleotide molecule shown in SEQ ID NO: 6), or an immunogenic fragment of an RBD region (e.g., the peptide stretch shown in SEQ ID NO:21 or encoded by the nucleotide molecule shown in SEQ ID NO: 22).
In some embodiments, the additional moiety is selected from: immune modulatory sequences, such as IL-2, IL-7, IL-12, IL-18, IL-21, GM-CSF, CD40L, CD40 stimulatory antibodies, PD-1 and PD-L1 antibodies, CTLA4 antibodies, chemokines CXCL9, CXCL10, CXCL11, CXCL12, CXCL3, XCL1, CCL4, CCL20, cholera toxin and subunits thereof, bacterial flagellin, FimH, HIV p24, HIV gp 41.
In some embodiments, the additional moiety is selected from: enabling the immunogenic peptide to form part of a nanoparticle, such as transferrin (Fn, e.g. a peptide molecule as shown in SEQ ID NO:7 or encoded by a nucleotide molecule as shown in SEQ ID NO: 8).
In some embodiments, the additional moiety is selected from: signal peptides, such as CD33, CD8, CD16, mouse IgG1 antibody.
In some embodiments, the additional moiety is selected from: a transmembrane region enabling the immunogenic peptide to be expressed on the surface of a viral vector, for example, the CD8 transmembrane region (CD8TM, for example, a peptide molecule represented by SEQ ID NO:17 or encoded by a nucleotide molecule represented by SEQ ID NO: 18), the HA2 transmembrane region, the CD4 transmembrane region, the gp41 transmembrane region.
In some embodiments, the additional moiety is selected from: linker peptides, e.g. (G4S)3、(G4S)n、 GSAGSAAGSGEF、(Gly)6、EFPKPSTPPGSSGGAP、KESGSVSSEQLAQFRSLD、 (Gly)8、EGKSSGSGSESKST。
In some embodiments, the additional moiety is selected from: tags such as His-tag, AviTag, Calmodulin tag, polyglutamate tag, E-tag, FLAG tag, HA-tag, Myc-tag, S-tag, SBP-tag, Sof-tag 1, Sof-tag3, Strep-tag, TC tag, V5 tag, T7 tag, VSV tag, Xpress tag, 3X FLAG tag, Isopep tag, Spytag, Snoop tag and PNE tag.
In some embodiments, the immunogenic peptide comprises: the RBD region or sRBD region or immunogenic fragments thereof linked to the HA2 region, and optionally other moieties linked to the foregoing.
In some embodiments, the immunogenic peptide comprises: an RBD region or an sRGB region (e.g., a peptide molecule represented by SEQ ID NO:5 or encoded by a nucleotide molecule represented by SEQ ID NO: 6) and an Fn region (e.g., a peptide molecule represented by SEQ ID NO:7 or encoded by a nucleotide molecule represented by SEQ ID NO: 8) linked to an HA2 region; an RBD region or sRBD region linked to the HA2 region, and a CD8 transmembrane region (e.g., a peptide molecule represented by SEQ ID NO:17 or encoded by a nucleotide molecule represented by SEQ ID NO: 18); an immunogenic fragment of the RBD region (e.g., the peptide molecule shown in SEQ ID NO:21 or encoded by the nucleotide molecule shown in SEQ ID NO: 22) linked to the HA2 region and the Fn region.
In some embodiments, the amino acid sequence of the immunogenic peptide is shown as SEQ ID No. 9 or 15 or 25, or the coding sequence of the immunogenic peptide is shown as SEQ ID No. 10 or 16 or 26.
In some embodiments, the present application uses the HA2 region of H7N9 virus linked to the RBD region of the new coronavirus, or an immunogenic fragment thereof, as an immunogen, while further modifying the immunogen, such as adding disulfide bonds, fusion proteins, fusion cytokines to the RBD region, thereby inducing neutralizing antibodies against both influenza virus and new coronavirus.
In some embodiments, the present application provides an immunogenic peptide directed against both influenza and neocorona, comprising the HA2 region of H7N9 viral hemagglutinin HA and the RBD region of SARS-CoV-2 viral spike protein S, which may be further modified with cysteine to form the sRBD region.
In some embodiments, the cysteine modification is the addition of a pair of cysteines at the root of the RBD domain to enable disulfide bond formation.
In one aspect of the disclosure, a nucleotide molecule is provided that encodes an immunogenic peptide herein.
In some embodiments, the coding sequence for the HA2 region comprises the nucleotide sequence set forth in SEQ ID NO. 2; the coding sequence of the immunogen crown comprises a nucleotide sequence shown as SEQ ID NO. 4 or a nucleotide sequence shown as SEQ ID NO. 6 or a nucleotide sequence shown as SEQ ID NO. 22; and/or the coding sequence of said further part comprises the nucleotide sequence shown in SEQ ID NO 8 or SEQ ID NO 18.
In some embodiments, the nucleotide molecule has the sequence shown in SEQ ID NO 10 or 16 or 26.
In one aspect of the present disclosure, a vector is provided comprising a nucleotide molecule herein.
In one aspect of the disclosure, a host cell is provided comprising a nucleotide molecule or vector herein or capable of expressing an immunogenic peptide described herein.
In some embodiments, the host cell is a mammalian cell or an insect cell, such as HEK293, HeLa, K562, CHO, NS0, SP2/0, PER.C6, Vero, RD, BHK, HT 1080, A549, Cos-7, ARPE-19, and MRC-5 cells; high Five, Sf9, Se301, SeIZD2109, SeUCR1, Sf9, Sf900+, Sf21, BTI-TN-5B1-4, MG-1, Tn368, HzAm1, BM-672302, Hz2E5 and Ao 38.
In one aspect of the present disclosure, there is provided a vaccine capable of simultaneously inducing an immune response against an influenza virus and another non-influenza virus or a non-alloinfluenza virus, comprising an immunogenic peptide, a nucleotide molecule, a vector and/or a host cell as described herein.
In some embodiments, the vaccine is a nucleic acid vaccine (DNA or RNA vaccine), a recombinant protein subunit vaccine, a recombinant viral vector vaccine, a recombinant bacterial vector vaccine, a virus-like particle vaccine, a nanoparticle vaccine, a cell vector vaccine.
In some embodiments, the vaccine is a viral vector vaccine, the viral vector selected from the group consisting of: poxviruses (e.g., Tiantan strain, North American vaccine strain, Whitman-derived strain, Listeria strain, Ankara-derived strain, Copenhagen strain, and New York strain), adenoviruses (Ad5, Ad11, Ad26, Ad35, AdC68, or modified variants thereof), adeno-associated viruses, herpes simplex viruses, measles viruses, reoviruses, rhabdoviruses, forest encephalitis viruses, influenza viruses, respiratory syncytial viruses, polioviruses.
In some embodiments, the vaccine comprises or is used in combination with an adjuvant, for example selected from the group consisting of: aluminum adjuvant, cholera toxin and its subunit, oligodeoxynucleotide, manganese ion adjuvant, colloidal manganese adjuvant, Freund's adjuvant, MF59 adjuvant, QS-21 adjuvant, Poly I: C and other TLR ligands, GM-CSF, IL-2, IL-3, IL-7, IL-11, IL-12, IL-18, IL-21.
In some embodiments, the vaccine is in a form suitable for intramuscular administration, intradermal administration, subcutaneous administration, nasal drip, aerosol inhalation, genital tract, rectal administration, oral administration, or a combination of the different modes of administration described above (e.g., intramuscular injection + nasal drip).
In some embodiments, the vaccine is in a form suitable for combined vaccination (e.g., combined vaccination or sequential vaccination) of 2 or more, such as sequential vaccination before and after with S or S1 vaccine of coronavirus (e.g., SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-229E, HCoV-OC43, HCoV-NL63, HCoV-HKU1, bat-CoV), or sequential vaccination before and after with HA or HA2 vaccine of influenza virus (e.g., HA or HA2 derived from any of H1-H18).
In one aspect of the present disclosure, there is provided the use of an immunogenic peptide, nucleotide molecule, vector and/or host cell as described herein in the manufacture of a medicament for the simultaneous prevention or treatment of influenza virus and another non-influenza virus.
In one aspect of the present disclosure, there is also provided an immunogenic peptide, nucleotide molecule, vector and/or host cell of the present disclosure for use in the simultaneous prevention or treatment of influenza and new coronavirus infection.
In one aspect of the present disclosure, there is also provided a method of preventing or treating influenza and new coronavirus infection simultaneously, the method comprising administering to a subject in need thereof an immunogenic peptide, nucleotide molecule, vector, host cell and/or vaccine of the present disclosure.
In some embodiments, the vaccine is a nucleic acid vaccine (DNA or RNA vaccine), a recombinant protein subunit vaccine, a recombinant viral vector vaccine, a recombinant bacterial vector vaccine, a virus-like particle vaccine, a nanoparticle vaccine, a cell vector vaccine.
In some embodiments, the vaccine comprises or is combined with an adjuvant including, but not limited to: aluminum adjuvant, cholera toxin and its subunit, oligodeoxynucleotide, manganese ion adjuvant, colloidal manganese adjuvant, Freund's adjuvant, SAS adjuvant, MF59 adjuvant, QS-21 adjuvant, Poly I: C and other TLR ligands, GM-CSF, IL-2, IL-3, IL-7, IL-11, IL-12, IL-18, IL-21, etc.
In some embodiments, the vaccine is in a form suitable for vaccination as follows: intramuscular inoculation, intradermal inoculation, subcutaneous inoculation, nasal drops, aerosol inhalation, reproductive tract, rectal, oral, or any combination thereof.
In some embodiments, vaccination is with one or more of the vaccines, e.g., combined vaccination or sequential vaccination.
In some embodiments, one or more of the vaccines are used to vaccinate against other vaccines against the novel coronavirus or influenza, e.g., the other vaccines include vaccines against coronavirus S or S1, e.g., S or S1 is from a group including, but not limited to, SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-229E, HCoV-OC43, HCoV-NL63, HCoV-HKU1, bat-CoV, etc.; such other vaccines include vaccines against influenza virus HA or HA2, for example HA or HA2 from the list including but not limited to H1-H18.
In some embodiments, the vaccine comprises a nucleic acid vaccine (DNA or RNA vaccine) in combination with an adenoviral vector vaccine.
In some embodiments, the vaccine comprises pcDNA3.1-S in combination with AdC 68-RHAF. In some embodiments, the components of the vaccine combination are administered sequentially, preferably first, before and after administration of the DNA vaccine.
In one aspect of the present disclosure, there is provided a method of preparing a vaccine against both influenza and new coronavirus infection, the method comprising:
(a) providing immunogenic peptides, nucleotide molecules and vectors of the present disclosure;
(b) combining the active substance provided in (a) with an immunologically or pharmaceutically acceptable carrier.
In one aspect of the disclosure, a method is provided for simultaneously inducing immune responses against influenza and other viruses, including coronaviruses, wherein a fusion protein consisting of the HA2 region of influenza virus hemagglutinin HA as an immunogen stem and the membrane proteins of the other viruses or strains as the corona is used as a vaccine immunogen to simultaneously induce immune responses against influenza and other viruses.
In some embodiments, HA2 immunogen is selected from the group including, but not limited to, H1-H18, particularly from the widely circulating human influenza H1, H2, H3 and multiple-occurring human infections with avian influenza H5 and H7;
in some embodiments, the HA2 immunogen is from H1, which was epidemic in 2009, H7, which was epidemic in 2013.
In some embodiments, the HA2 immunogen is from the HA2 sequence of an early strain of 2013 epidemic H7 and is more than 80% full-length or fragment homologous to that sequence. In some embodiments, the HA2 immunogen may comprise the amino acid sequence shown in SEQ ID No. 1 or may be encoded by a coding sequence having SEQ ID No. 2.
In some embodiments, the crown immunogen linked to HA2 is from a group including, but not limited to, coronavirus, hiv, influenza virus, rabies virus, hog cholera virus, porcine reproductive and respiratory syndrome virus, measles virus, ebola virus, herpes virus, arbovirus (zika virus, epidemic encephalitis b virus, forest encephalitis virus, dengue virus, hantavirus, hemorrhagic fever virus).
In some embodiments, the crown immunogen attached to HA2 is selected from coronaviruses including, but not limited to, SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-229E, HCoV-OC43, HCoV-NL63, HCoV-HKU1, bat-CoV, and the like, particularly the S1 protein comprising the Receptor binding domain of a coronavirus, or the Receptor Binding Domain (RBD), or an engineered Receptor binding domain.
In some embodiments, the crown immunogen linked to HA2 is selected from S1 of coronavirus SARS-CoV-2, particularly the RBD region from S1 or full length and fragments thereof having more than 90% homology thereto. In some embodiments, the RBD region immunogen can comprise the amino acid sequence shown in SEQ ID NO. 3 or can be encoded by a coding sequence having SEQ ID NO. 4. In some embodiments, the RBD region immunogen may comprise the amino acid sequence shown in SEQ ID NO. 5 or may be encoded by a coding sequence having SEQ ID NO. 6.
In one aspect of the disclosure, a method for simultaneously inducing immune responses against influenza and other viruses including coronaviruses is provided, which comprises co-expressing the immunogen of the present application with other sequences, either by fusion or in a single reading frame, to further increase the antiviral species, or to enhance the ability of the immunogen to induce an immune response, thereby preventing multiple viral infections.
In some embodiments, the sequence linked to an immunogen of the present application: from a source including, but not limited to, various coronaviruses, influenza viruses, aids viruses, rabies viruses, hog cholera viruses, blue ear disease viruses, measles viruses, ebola viruses, herpes viruses, arboviruses (zika virus, epidemic encephalitis b virus, forest encephalitis b virus, dengue virus, hantaviruses, hemorrhagic fever viruses), especially co-expressed with immunogens that activate T cell responses, to prepare composite vaccines that are capable of activating both neutralizing antibodies and T cell responses; (iii) are immune modulatory sequences including, but not limited to, IL-2, IL-7, IL-12, IL-18, IL-21, GM-CSF, CD40L, CD40 stimulatory antibodies, PD-1 and PD-L1 antibodies, CTLA4 antibodies, chemokines CXCL9, CXCL10, CXCL11, CXCL12, CXCL3, XCL1, CCL4, CCL20, cholera toxin and subunits thereof, bacterial flagellin, FimH, and the like; features are provided for forming nanoparticles including, but not limited to, transferrin (Ferritin).
In one aspect of the present disclosure, a method is provided for simultaneously inducing an immune response against influenza and other viruses, including coronaviruses, comprising combining immunogens described herein, alone or in combination with co-expressed molecules described herein, for the preparation of vaccines including, but not limited to, nucleic acid vaccines (DNA or RNA vaccines), recombinant protein subunit vaccines, recombinant viral vector vaccines, recombinant bacterial vector vaccines, virus-like particle vaccines, nanoparticle vaccines, cell vector vaccines, and the like.
In one aspect of the disclosure, a method of simultaneously inducing an immune response against influenza and other viruses, including coronaviruses, is provided, comprising administering an immunogenic peptide, its encoding nucleotide molecule, vector, host cell or vaccine as described herein.
In some embodiments, vaccination with a combination of 2 or more of the vaccines described above is performed, either in combination or sequentially. In some embodiments, the vaccines herein are sequentially inoculated with a vaccine prepared from coronavirus S or S1, said S or S1 being from a group including, but not limited to, SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-229E, HCoV-OC43, HCoV-NL63, HCoV-HKU1, bat-CoV, and the like; or the vaccines herein are inoculated sequentially, before and after, with a vaccine prepared from influenza virus HA or HA2, said HA or HA2 being from the group including, but not limited to, H1-H18.
Any combination of the foregoing aspects and features may be made by those skilled in the art without departing from the spirit and scope of the present disclosure. Other aspects of the disclosure will be apparent to those skilled in the art in view of the disclosure herein.
Drawings
The present disclosure is further described with reference to the accompanying drawings, wherein the showings are for the purpose of illustrating embodiments of the disclosure only and not for the purpose of limiting the scope of the disclosure.
FIG. 1: the construction of S protein eukaryotic expression vector and sRGB-HA 2-hFn (RHAF) adenovirus and the expression of fusion protein:
a: pcDNA3.1-S and pAdC68XY3-RHAF plasmid construction maps;
b: the pcDNA3.1-S protein is successfully expressed in 293T cells, and the adenovirus AdC68-RHAF can successfully express the RHAF protein.
FIG. 2 immunogenicity of AdC68-RHAF in C57BL/6 mice:
the mice used in the experiment are female C57BL/6 with the age of 6-8 weeks, and the immunogen is plasmid pcDNA3.1-S and adenovirus AdC 68-RHAF.
A: the ELISA method measures the titer of bound antibody in the mouse serum at week 2 after the end of immunization. The abscissa is the immune group and the ordinate is the titer of bound antibody, p <0.01
B: data for bound antibodies to RBD protein before and after second immunization, indicates p < 0.01.
C: 293T-ACE2 cells were tested for the titer of neutralizing antibodies in mouse sera at week 2 after the end of immunization. The abscissa is the immune group and the ordinate is the titer (ID) of neutralizing antibodies50) Denotes p<0.01。
D: neutralizing antibody titers against SARS-Cov-2 pseudovirus before and after immunization of the second needle.
E: the ELISPOT method measures T cell responses in mice at week 2 after completion of immunization. The abscissa is the immunization group and the ordinate is the number of IFN- γ secreting cells per million splenocytes, indicating p < 0.05.
FIG. 3: immunogenicity of AdC68-RHAF in hACE2+ transgenic mice:
the mice used in the experiment are 6-8 weeks old hACE2+ transgenic mice, and the immunogen is adenovirus AdC 68-RHAF.
A: the ELISA method measures the titer of bound antibody in the mouse serum at week 2 after the end of immunization. The abscissa is the immune group and the ordinate is the titer of bound antibody, indicates p < 0.0001.
B: 293T-ACE2 cells were tested for the titer of neutralizing antibodies in mouse sera at week 2 after the end of immunization. The abscissa is the immune group and the ordinate is the titer (ID) of neutralizing antibodies50) Denotes p<0.0001。
FIG. 4: immunogenicity of AdC68-RHAF and AdC68-RBD-HA2-CD8TM in BALB/c mice:
the mice used in the experiment are female BALB/c mice with the age of 6-8 weeks, and the immunogens are adenovirus AdC-RHAF and AdC68-RBD-HA2-CD8 TM.
A: the ELISA method measures the titer of bound antibody in mouse serum 1 week after the completion of immunization. The horizontal axis represents the immunization group, and the vertical axis represents the titer of the bound antibody.
B: neutralizing antibody titers against SARS-CoV-2 pseudovirus detected one week after the end of immunization.
FIG. 5: challenge experiment of new coronavirus:
the mice used in the experiment are 6-8 weeks old hACE2+ transgenic mice, and the immunogen is adenovirus AdC 68-RHAF.
A: viral load. The abscissa is the immunization group and the ordinate is the viral load.
B: lung tissue H & E staining.
C: body weight changes after challenge in mice. The abscissa is the number of days after challenge, and the ordinate is the weight percentage.
D: survival of mice after challenge. The abscissa is the number of days after challenge, and the ordinate is the percentage of survival.
FIG. 6: different influenza virus challenge experiments:
a: body weight change in mice after H7N9 challenge. The abscissa is the number of days after challenge, and the ordinate is the weight percentage.
B: survival of mice after H7N9 challenge. The abscissa represents days after challenge, and the ordinate represents percentage survival, wherein p is less than 0.05 and p is less than 0.01.
C: body weight change in mice after H3N2 challenge. The abscissa is the number of days after challenge, the ordinate is the weight percentage, p is <0.05, p is < 0.01.
FIG. 7: construction of R545-HA2-IntN-PAB-7XHis eukaryotic expression vector and expression of R545-HA2-IntN-PAB-7XHis protein:
a: constructing a map by pFastBac-Dual-R545-HA2-IntN-PAB-7XHis plasmid;
b: baculovirus successfully expressed the protein R545-HA2-IntN-PAB-7XHis in insect cell line Sf 9.
FIG. 8: purification of R545-HA2-IntN-PAB-7XHis protein and assembly of nanoparticles R545 HAF:
a: purifying R545-HA2-IntN-PAB-7XHis protein;
b: R545-HA2-IntN-PAB-7XHis linked to Intein from gb 1-IntC-Fn; as shown in the figure, the amount of the monomers gb1-IntC-Fn was significantly reduced and cleavage was seen to generate the byproducts IntN-PAB (17kDa) and gb1-IntC (11kDa), indicating the formation of the nanoparticle R545 HAF.
FIG. 9: immunogenicity of nanoparticle R545HAF in ICR mice:
the mice used in the experiment are 6-8 weeks old hACE2+ transgenic mice, and the immunogen is plasmid pcDNA3.1-S and nanoparticle R545 HAF.
A: the ELISA method measures the titer of bound antibody in mouse serum at week 4 after completion of immunization. The horizontal axis represents the immune groups and the vertical axis represents the titers of bound antibodies, indicating that p < 0.05.
B: data for bound antibodies to RBD protein after the first immunization and the third immunization, the left panel is overall boost, the right panel is the corresponding boost of a single mouse, indicating p < 0.05.
C: 293T-ACE2 cell detection of mice at week 4 after immunizationSerum neutralizing antibody titers. The abscissa is the immune group and the ordinate is the titer (ID) of neutralizing antibodies50) Denotes p<0.05。
D: neutralizing antibody titers against SARS-Cov-2 pseudovirus after the first immunization and the third immunization, the left panel is the overall boost, the right panel is the corresponding boost of a single mouse, indicating p < 0.05.
Detailed Description
The present disclosure relates to the field of vaccines, and in particular to a method of simultaneously inducing an immune response against influenza and other viruses, including coronaviruses. Animal experiment results prove that the vaccine disclosed by the invention is safe, can continuously generate high-titer neutralizing antibodies, and can be used for preventing and treating influenza and Xinguan.
All numerical ranges provided herein are intended to expressly include all numbers between the end points of the ranges and numerical ranges there between. The features mentioned in the present disclosure or the features mentioned in the embodiments can be combined. All the features disclosed in this specification may be combined in any combination, and each feature disclosed in this specification may be replaced by alternative features serving the same, equivalent or similar purpose. Thus, unless expressly stated otherwise, the features disclosed are merely generic examples of equivalent or similar features.
As used herein, "about" in the context of a value or range means ± 10% of the recited or claimed value or range.
It is to be understood that when ranges of parameters are provided, the invention likewise provides all integers and decimals thereof within the ranges. For example, "0.1-2.5 mg/day" includes 0.1 mg/day, 0.2 mg/day, 0.3 mg/day, etc. up to 2.5 mg/day.
As used herein, "comprising," having, "or" including "includes" comprising, "" consisting essentially of … …, "" consisting essentially of … …, "and" consisting of … …; "consisting essentially of … …", "consisting essentially of … …", and "consisting of … …" are subordinate concepts of "comprising", "having", or "including".
Immunogenic peptides and molecules encoding same
As used herein, the term "immunogenic peptide" refers to a peptide capable of simultaneously inducing an immune response against an influenza virus and another non-influenza virus, comprising the following moieties: (a) immunogen stem: it comprises the HA2 region of influenza virus hemagglutinin HA; (b) immunogen crown: comprising a viral membrane protein or an immunogenic fragment thereof, wherein the viral membrane protein is derived from a virus other than the virus from which the HA2 region of (a) was derived; (c) optionally, other moieties attached to the foregoing moieties.
As used herein, the term "linked" has its broadest meaning and can include, for example, fusion, co-expression, covalent linkage, coupling, and the like.
For example, as used herein, the terms "sRBD-HA 2 immunogenic peptide", "immunogenic peptide against influenza virus and novel coronavirus SARS-CoV-2", which are used interchangeably, refer to a peptide that includes the HA2 region of influenza hemagglutinin HA fused to the RBD region of the SARS-CoV-2 virus spike protein S and that HAs the ability to elicit the binding and neutralizing antibody effects and corresponding T cell responses against influenza virus and novel coronavirus SARS-CoV-2.
In some embodiments, the immunogenic peptide can be: (a) has the sequence shown in SEQ ID NO:1 and the amino acid sequence shown in SEQ ID NO 3 or 5 or 21; (b) (ii) a polypeptide which is homologous or similar in sequence to the polypeptide of (a) and has the same or similar immunogenicity, e.g. wherein each segment is identical to the sequence of SEQ ID NO:1 and the amino acid sequence shown in SEQ ID No. 3 or 5 or 21, respectively, have a homology or sequence identity higher than or equal to 80%, higher than or equal to 85%, higher than or equal to 90%, higher than or equal to 95%, higher than or equal to 96%, higher than or equal to 97%, higher than or equal to 98%, higher than or equal to 99%, or have a homology or sequence identity higher than or equal to 80%, higher than or equal to 85%, higher than or equal to 90%, higher than or equal to 95%, higher than or equal to 96%, higher than or equal to 97%, higher than or equal to 98%, higher than or equal to 99% with the polypeptide in (a); (c) and (b) protein or polypeptide which is derived from (a) or (b) and has the same or similar immunogenicity through substituting, deleting or adding one or more amino acids in the amino acid sequence defined by (a) or (b).
In some embodiments, the immunogenic peptide can be: (a') has the sequence of SEQ ID NO:9 or 15 or 25; (b') a polypeptide encoded by the nucleotide sequence shown in SEQ ID NO. 10 or 16 or 26; (c ') a polypeptide which is homologous or sequence-similar to the polypeptides described in (a ') and (b ') and has the same or similar immunogenicity, for example has a homology or sequence identity of greater than or equal to 80%, greater than or equal to 85%, greater than or equal to 90%, greater than or equal to 95%, greater than or equal to 96%, greater than or equal to 97%, greater than or equal to 98%, greater than or equal to 99% to the polypeptides described in (a ') and (b '); (d ') a protein or polypeptide derived from (a ') or (b ') or (c ') by substitution, deletion or addition of one or several amino acids in the amino acid sequence defined in (a ') or (b ') or (c ') and having the same or similar immunogenicity.
In some embodiments, the immunogenic peptide can include other moieties attached to the immunogen stem or crown, for example, to enhance the stability of the immunogen stem or crown or fusion protein, to enhance neutralizing antibody responses, to form multimers, to increase cellular responses, and the like. In some embodiments, other portions may include, but are not limited to: proteins derived from viruses or hosts, transferrin (Fn), IL-2, IL-7, IL-12, IL-18, IL-21, GM-CSF, CD40L, CD40 stimulating antibody, PD-1 and PD-L1 antibodies, CTLA4 antibodies, chemokines CXCL9, CXCL10, CXCL11, CXCL12, CXCL3, XCL1, CCL4, CCL20, cholera toxin and subunits thereof, bacterial flagellin, FimH, and the like.
In some embodiments, the immunogenic peptide may further include elements such as a signal peptide, a linker, a molecular tag, and the like. For example, a signal peptide element may refer to an amino acid sequence having the function of directing secretion, localization and/or transport of a protein, which is typically 5-30 amino acids in length. In some embodiments, the signal peptide element may be selected from: a protein self signal peptide, a CD33 protein signal peptide, a CD8 protein signal peptide, a CD16 protein signal peptide, a mouse IgG1 antibody signal peptide and an influenza HA protein signal peptide. For example, a linker peptide sequence (or linker) may be referred to as a fusion protein hereinThe length of the short peptide is usually 1-50 (e.g. 5-50, 5-40, 10-40) amino acids. In general, the linker peptide does not affect or seriously affects the formation of the correct fold and spatial conformation of the peptide of the invention. In some embodiments, the linker peptide element may be selected from: (G4S)3Joint (G4S)n、GSAGSAAGSGEF、 (Gly)6、EFPKPSTPPGSSGGAP、KESGSVSSEQLAQFRSLD、(Gly)8、 EGKSSGSGSESKST。
The immunogenic peptide may also include variants thereof, such as deletion, insertion and/or substitution of one or more (typically 1 to 50, preferably 1 to 30, more preferably 1 to 20, most preferably 1 to 10, e.g., 1, 2, 3, 4, 5,6, 7, 8, 9 or 10) amino acids, and addition of one or several (typically up to 20, preferably up to 10, more preferably up to 5) amino acids at the C-terminus and/or N-terminus. For example, in the art, substitutions with amino acids of similar or similar properties will not generally alter the function of the protein or polypeptide. Also, for example, the addition of one or several amino acids at the C-terminus and/or N-terminus does not generally alter the function of the protein or polypeptide.
Immunogenic peptides can be produced by recombinant expression under appropriate circumstances and conditions, e.g., produced by the coding nucleotide molecules, vectors, host cells of the disclosure; it can also be obtained by chemical synthesis or the like, so long as it has the desired amino acid sequence and immunogenicity and reactivity.
As used herein, the terms "immunogenic peptide-encoding molecule", "coding sequence", and the like, are used interchangeably and all refer to a nucleotide molecule that encodes an immunogenic peptide described in the present disclosure.
In some embodiments, the nucleotide molecule may be selected from, for example: (i) has the sequence shown in SEQ ID NO:2 and the nucleotide sequence shown in SEQ ID NO. 4 or 6 or 22; (ii) (ii) a molecule that hybridizes to (i) under stringent conditions; (iii) (ii) a nucleotide molecule homologous or similar to the sequence of (i) and capable of expressing a functional immunogenic peptide, e.g. wherein each segment is identical to the sequence of SEQ ID NO:2 and the nucleotide sequence shown in SEQ ID No. 4 or 6 or 22, respectively, have a homology or sequence identity higher than or equal to 80%, higher than or equal to 85%, higher than or equal to 90%, higher than or equal to 95%, higher than or equal to 96%, higher than or equal to 97%, higher than or equal to 98%, higher than or equal to 99%, or a nucleotide molecule having a homology or sequence identity higher than or equal to 80%, higher than or equal to 85%, higher than or equal to 90%, higher than or equal to 95%, higher than or equal to 96%, higher than or equal to 97%, higher than or equal to 98%, higher than or equal to 99% with the sequence in (i) or (ii) and capable of expressing a functional immunogenic peptide; (iv) a nucleotide molecule which is formed by substituting, deleting or adding one or more nucleotides in the nucleotide sequence defined by (i) or (ii) and can express a functional immunogenic peptide.
In some embodiments, the nucleotide molecule may be selected from, for example: (i) has the sequence shown in SEQ ID NO:10 or 16 or 26; (ii) (ii) a molecule that hybridizes to (i) under stringent conditions; (iii) (iii) a nucleotide molecule having greater than or equal to 80%, greater than or equal to 85%, greater than or equal to 90%, greater than or equal to 95%, greater than or equal to 96%, greater than or equal to 97%, greater than or equal to 98%, greater than or equal to 99% homology or sequence identity to the sequence in (i) or (ii) and capable of expressing a functional immunogenic peptide; (iv) a nucleotide molecule which is formed by substituting, deleting or adding one or more nucleotides in the nucleotide sequence defined by (i) or (ii) and can express a functional immunogenic peptide.
As used herein, the term "stringent conditions" refers to: (1) hybridization and elution at lower ionic strength and higher temperature, such as 0.2 XSSC, 0.1% SDS, 60 ℃; or (2) adding denaturant during hybridization, such as 50% (v/v) formamide, 0.1% calf serum/0.1% Ficoll, 42 deg.C, etc.; or (3) hybridization occurs only when the identity between two sequences is at least 50%, preferably 55% or more, 60% or more, 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, or 90% or more, more preferably 95% or more.
The full-length nucleotide sequence or a fragment thereof of the present disclosure can be obtained by PCR amplification, recombination, or artificial synthesis. For PCR amplification, primers can be designed based on the nucleotide sequences disclosed in the present disclosure, and the sequences can be amplified using a commercially available cDNA library or a cDNA library prepared by a conventional method known to those skilled in the art as a template. When the sequence is long, two or more PCR amplifications are often required, and then the amplified fragments are spliced together in the correct order.
Vectors and host cells
The disclosure also relates to vectors comprising the immunogenic peptide-encoding nucleotide molecules of the present application, and host cells genetically engineered with the vectors.
The coding sequences of the present disclosure can be used to express or produce recombinant immunogenic peptides by conventional recombinant DNA techniques (Science, 1984; 224: 1431). Generally, the following steps are performed:
(1) transferring the coding nucleotide molecules of the present disclosure, or the recombinant expression vectors containing the nucleotide molecules, into suitable host cells;
(2) a host cell cultured in a suitable medium;
(3) isolating and purifying the protein or polypeptide from the culture medium or the cells.
In the present disclosure, the terms "vector" and "recombinant expression vector" are used interchangeably and refer to a bacterial plasmid, phage, yeast plasmid, animal cell virus, mammalian cell virus or other vector well known in the art. An important feature of expression vectors is that they generally contain an origin of replication, a promoter, a marker gene and translation control elements.
Expression vectors containing the immunogenic peptide coding sequences and appropriate transcriptional/translational control signals can be constructed using methods conventional in the art. These methods include in vitro recombinant DNA techniques, DNA synthesis techniques, in vivo recombinant techniques, and the like. The DNA sequence may be operably linked to a suitable promoter in an expression vector to direct mRNA synthesis. The expression vector also includes a ribosome binding site for translation initiation and a transcription terminator. Expression systems such as pcDNA3.1 vector, pIRES2-EGFP vector, AdMaxTM, AdC68, and the like may be employed in the present disclosure.
In addition, the expression vector may contain one or more selectable marker genes to provide phenotypic traits useful for selection of transformed host cells, such as dihydrofolate reductase, neomycin resistance, and Green Fluorescent Protein (GFP) for eukaryotic cell culture, or tetracycline or ampicillin resistance for E.coli.
Vectors comprising the appropriate DNA sequences described above, together with appropriate promoter or control sequences, may be used to transform an appropriate host cell so that it can express the protein or polypeptide. The host cell may be a prokaryotic cell, such as a bacterial cell; or lower eukaryotic cells, such as yeast cells; or higher eukaryotic cells, such as animal cells. Representative examples are: escherichia coli, Streptomyces, Agrobacterium; fungal cells such as yeast; animal cells, and the like. In the present disclosure, for example, a host cell selected from the group consisting of: HEK293, HeLa, CHO, K562, NS0, SP2/0, PER.C6, Vero, RD, BHK, HT 1080, A549, Cos-7, ARPE-19 and MRC-5 cells; high Five, Sf9, Se301, SeIZD2109, SeUCR1, Sf9, Sf900+, Sf21, BTI-TN-5B1-4, MG-1, Tn368, HzAm1, BM-672302, Hz2E5 and Ao 38.
The nucleotide molecules of the present disclosure will provide enhanced transcription when expressed in higher eukaryotic cells if enhancer sequences are inserted into the vector. Enhancers are cis-acting elements of DNA, usually about 10 to 300 base pairs, that act on a promoter to increase transcription of a gene. It will be clear to one of ordinary skill in the art how to select appropriate vectors, promoters, enhancers and host cells.
The recombinant polypeptide in the above method may be expressed or secreted intracellularly or on the cell membrane to the outside of the cell. If necessary, the recombinant protein can be isolated and purified by various separation methods using its physical, chemical and other properties. These methods are well known to those skilled in the art. Examples of such methods include, but are not limited to: conventional renaturation treatment, treatment with a protein precipitant (such as salt precipitation), centrifugation, cell lysis by osmosis, sonication, ultracentrifugation, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, High Performance Liquid Chromatography (HPLC), and other various liquid chromatography techniques, and combinations thereof.
Vaccines and immunoconjugates
Also provided herein is a vaccine, or immunogenic composition, comprising the immunogenic peptides, nucleotide molecules, vectors, and/or host cells of the disclosure. The vaccine comprises a formulation of immunogenic peptides and/or nucleic acid molecules of the present disclosure in a form capable of being administered to a vertebrate, preferably a mammal, and which induces a protective immune response that enhances immunity to prevent and/or reduce influenza virus infection and other non-influenza virus infections (e.g., novel coronavirus infections) and/or at least one symptom thereof.
The term "protective immune response" or "protective response" refers to an immune response to an infectious agent or disease that is exhibited by a vertebrate (e.g., a human), which prevents or reduces infection or reduces at least one disease symptom thereof, mediated by an immunogen.
The term "vertebrate" or "subject" or "patient" refers to any member of the subphylum chordata, including, but not limited to: humans and other primates, including non-human primates such as chimpanzees and other apes and monkey species; livestock such as cattle, sheep, pigs, goats, and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs; birds include domesticated, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese. The terms "mammal" and "animal" are included in this definition and are intended to encompass adult, juvenile, and newborn individuals.
The vaccines herein can be recombinant protein vaccines, recombinant DNA vaccines, recombinant viral vector vaccines (e.g., adenoviral vectors, poxvirus vectors, adeno-associated viral vectors, herpes simplex viral vectors, cytomegalovirus vectors), recombinant bacterial vector vaccines, recombinant yeast vector vaccines, or recombinant virus-like particle vaccines. In some embodiments, the vaccine herein is selected from a recombinant DNA vaccine, a recombinant adenoviral vector, or a combination of one or both thereof.
In some embodiments, one or more vaccines or combinations thereof selected from the group consisting of: recombinant plasmid vaccines (DNA), such as DNA vaccines comprising the SARS-CoV2 virus spike protein S coding sequence of the RBD region (e.g., pcDNA3.1-S); recombinant adenoviral vector vaccines, such as AdC 68-RHAF; recombinant protein subunit vaccines, such as nanoparticles, R545 HAF.
An effective amount of the immunogen herein is included in the vaccine compositions herein. The vaccine compositions of the present disclosure comprise an immunogen in an amount sufficient to achieve the desired biological effect. The term "effective amount" generally refers to an amount of immunogen that can induce a protective immune response sufficient to induce immunity to prevent and/or alleviate an infection or disease and/or to reduce at least one symptom of an infection or disease.
Adjuvants may also be included in the vaccines herein. Adjuvants known to those of ordinary skill in the art may be employed, such as those described in Vogel et al, "A Complex of Vaccine Adjuvants and Excipients" (2 nd edition), which is incorporated herein by reference in its entirety. Examples of known adjuvants include, but are not limited to: aluminum adjuvant, cholera toxin and its subunit, oligodeoxynucleotide, manganese ion adjuvant, colloidal manganese adjuvant, Freund's adjuvant, MF59 adjuvant, QS-21 adjuvant, Poly I: C and other TLR ligands, GM-CSF, IL-2, IL-3, IL-7, IL-11, IL-12, IL-18, IL-21, etc.
The vaccine compositions herein may further comprise pharmaceutically acceptable carriers, diluents, preservatives, solubilizers, emulsifiers and like excipients. For example, pharmaceutically acceptable carriers are known and include, but are not limited to, water for injection, saline solution, buffered saline, dextrose, water, glycerol, sterile isotonic aqueous buffer, and combinations thereof. Pharmaceutically acceptable carriers, diluents and other excipients can be found, for example, in Remington's pharmaceutical Sciences.
The vaccine compositions herein may be in a form suitable for systemic or local (especially in the respiratory tract) administration. Methods of administering vaccine compositions include, but are not limited to: intramuscular inoculation, intradermal inoculation, subcutaneous inoculation, nasal drops, aerosol inhalation, reproductive tract, rectal, oral administration or any combination thereof, e.g., intramuscular injection followed by nasal drops.
In some embodiments, the vaccines herein prevent, eliminate or reduce influenza virus and novel coronavirus infections or at least one symptom thereof in a subject, such as respiratory symptoms (e.g., nasal congestion, sore throat, hoarseness), headache, cough, sputum, fever, rales, wheezing, dyspnea, pneumonia due to infection, severe acute respiratory syndrome, renal failure, and the like.
Also contemplated herein is an immunoconjugate (also referred to as an immunoconjugate) comprising the immunogen herein and other materials coupled thereto. The additional substance may be a targeting substance (e.g., a moiety that specifically recognizes a particular target), a therapeutic substance (e.g., a drug, a toxin, a cytotoxic agent), a labeling substance (e.g., a fluorescent label, a radioisotope label).
Also provided in the present disclosure is a combination product comprising an immunogenic peptide, nucleotide molecule, vector, host cell and/or vaccine of the present disclosure, and may further comprise one or more additional substances that contribute to better functioning or enhancing the stability of the aforementioned substances in preventing and/or treating influenza virus infections and other non-influenza virus infections (e.g., novel coronavirus infections) or symptoms thereof. For example, the other substances may include other vaccines against coronavirus S or S1, such as S or S1 vaccines from including, but not limited to SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-229E, HCoV-OC43, HCoV-NL63, HCoV-HKU1, bat-CoV; other vaccines against influenza virus HA or HA2, said HA or HA2 being from the group including but not limited to H1-H18; other active agents that benefit from T cell activation and/or memory immune response with T cells.
Immunization method
Also provided herein is a method for preventing and/or treating influenza virus infections and other non-influenza virus infections (e.g., novel coronavirus infections) and/or symptoms thereof, comprising: administering at least once a prophylactically and/or therapeutically effective amount of one or more vaccines of the present disclosure. Inoculation regimes that may be used include, but are not limited to: systemic immunization modes, such as intramuscular injection, subcutaneous injection, intradermal injection and the like; and (3) immunization in respiratory tract, such as atomization, nasal drip and the like. In some embodiments, the primary immunization employs systemic or intrarespiratory vaccination, preferably systemic vaccination.
In some embodiments of the disclosure, the interval between each two vaccinations is at least 1 week, e.g., 2 weeks, 4 weeks, 2 months, 3 months, 6 months, or longer intervals.
In some embodiments, the primary immunization is performed with a DNA vaccine and the one or more booster immunizations are performed with a recombinant viral vaccine. The methods of immunization of the present disclosure may be by "prime-boost" or "prime-boost-re-boost", by a single systemic or local immunization of the respiratory tract, or by a combination of both immunization modalities.
Depending on the characteristics of the different vector vaccines, in some preferred embodiments, a systemic prime is performed using a recombinant DNA vaccine to establish a systemic immune response, followed by one or more boosts using other vaccines (e.g., recombinant adenoviral vaccines or recombinant poxvirus vaccines), which may include at least one boost in the respiratory tract (e.g., using an adenoviral vaccine).
The vaccine-specific immune response that can be effectively established in the local and systemic respiratory tract systems using the immunization methods herein helps to enhance the effectiveness of vaccine protection.
Providing the combination product herein in the form of a pharmaceutical pack or kit may, for example, be packaged in one or more containers, for example sealed containers such as ampoules or sachets, indicating the amount of composition, for example, one or more of the vaccine compositions herein or one or more of its ingredients. The vaccine compositions may be provided as a liquid, sterile lyophilized powder, or anhydrous concentrate, etc., which may be diluted, reconstituted and/or formulated with an appropriate liquid (e.g., water, saline, etc.) immediately prior to use to obtain the appropriate concentration and form for administration to a subject.
The combination product of the present disclosure can be used for the characteristic of locally inducing high-level antigen-specific CD8+ T cell responses in the respiratory tract, making it promising for the prevention and treatment of respiratory tract pathogen infection, reduction of respiratory tract pathogen pathogenicity, and respiratory tract tumors.
Examples
The disclosure is further illustrated with reference to specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present disclosure. Appropriate modifications, variations and changes may be made by those skilled in the art to the present disclosure, which modifications and changes are within the scope of the present disclosure.
The experimental procedures for the conditions not specified in the examples below can be carried out by methods conventional in the art, for example, by referring to the molecular cloning, A Laboratory Manual, New York, Cold Spring Harbor Laboratory Press, 1989 or according to the conditions recommended by the supplier. Methods for sequencing DNA are conventional in the art and tests are also available from commercial companies.
Unless otherwise indicated, percentages and parts are by weight. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present disclosure. The preferred embodiments and materials described herein are intended to be exemplary only.
The experimental animals, immunization protocols, immunogens, pseudoviruses and detection methods involved in the following experiments were as follows:
I. experimental animals:
6-8 week old female C57/BL6 and BALB/C mice, purchased from Shanghai Jihui laboratory animal feeding Co., Ltd.);
6-8 week old female hACE2+ ICR mice, purchased from Beijing Vital rising Tech technologies, Inc.).
Immunization protocol:
intramuscular injection is respectively carried out on the left hind limb and the right hind limb of the mouse; or for nasal drip. Specific dosages are given in the examples.
Selection of immunogen:
the HA2 sequence is from Genebank AGI60292.1, and the S and RBD sequences are from Genebank NC-045512.2; the hFn sequence (human transferrin) is from Genebank: M97164.1, and the specific sequence is shown in a sequence table.
1. Recombinant plasmid vaccine (DNA): pcDNA3.1-S, pcDNA3.1 (empty);
2. recombinant adenoviral vector vaccines: AdC68, AdC 68-sRGB-HA 2-hFn (AdC68-RHAF), AdC68-RBD-HA2-CD8 TM.
3. Recombinant protein subunit vaccine (nanoparticle, protein): r545HAF
Immunogen preparation and immunization dose:
preparation of immunogens see example 1
The immunogen immunizing doses used in the examples are as follows:
1. recombinant plasmid vaccine (DNA): 100 mu g/mouse, 100 mu L, dissolved in sterile physiological saline;
2. recombinant adenoviral vector vaccines: 5E10 vp/mouse, 100 μ L (intramuscular injection); 5E10 vp/mouse, 40. mu.L (nasal drip).
3. Recombinant protein subunit vaccine (nanoparticle, protein): the protein (dissolved in sterile PBS) was mixed with aluminum adjuvant (aluminum, InvivoGen, cat # 5200) at a volume ratio of 1:1 and immunized with 20. mu.g/mouse, 100. mu.L;
v. immunization interval:
specific immunization intervals are shown in the table below.
SARS-CoV2 envelope Pseudovirus (Pseudovirus) packaging:
1. 293T cells were prepared the day before transfection and used for transfection and expression of packaging plasmids. Cells were diluted to 5X 10 with DMEM complete medium6one/mL of cells, 1mL of diluted cells were plated in a 10cm dish at 37 ℃ with 5% CO2Culturing overnight;
2. sucking SARS-CoV2 membrane protein plasmid pcDNA3.1-S4 μ g and pNL4-3 delta env skeleton plasmid 8 μ g (NIH AIDS Reagent Program,3418) into 500 μ L DMEM without double antibody (serum-free, double antibody is mixed solution of streptomycin), and incubating at room temperature for 5 min;
3. diluting 24 μ L TurboFect (Thermo Fisher Scientific) with DMEM-free medium to a final volume of 500 μ L/sample, and incubating at room temperature for 5 min;
4. the two samples 2 and 3 were mixed well, 1000. mu.L/final sample volume, incubated at room temperature for 20min, and added to 293T cells in a 10cm dish after incubation. After 6h, replacing fresh 15mL of complete culture medium, and continuously culturing in a cell culture box for 48 h;
5. after the culture is finished, collecting cell culture supernatant in a 10cm dish, centrifuging for 10min at 4000g and 4 ℃ in a 15mL centrifuge tube, filtering the cell culture supernatant into a new 15mL centrifuge tube by using a 0.45 mu m filter, freezing the cell culture supernatant at the temperature of minus 80 ℃, and titrating the cell culture supernatant for later use.
Construction of 293T cells stably expressing hACE2 receptor:
1. an artificial synthetic human ACE2(hACE2) sequence (Genebank # NCBI _ NP-001358344.1) as shown in SEQ ID NO:13 with an Age1 cleavage site at the 5 'end and an Xba1 cleavage site at the 3' end, the synthetic fragment cleaved with vector plasmid pHAGE-MCS-puro using Age1 (Thermo Scientific, cat # FD1464) with Xba1 (Thermo Scientific, cat # FD0685) and recovered by gel electrophoresis followed by gel cutting and recovery of the cleaved fragments using Sanprep column DNA gel recovery kit (Promega, cat # A9282).
2. The gene recovery product was ligated to the digested linearized vector using T4 DNA ligase (Thermo Scientific Co., Ltd., cat # 2011A): coli Stable was transformed with the ligation product and grown overnight on ampicillin-containing plates. On day 2, single colonies were randomly picked and sequenced, the mutation sites were corrected, and after confirming that the entire sequence was correct, a lentiviral expression plasmid (pHAGE-hACE2-puro) for the hACE2 gene was successfully cloned.
3. 10cm dishes were taken and inoculated about 5X 10 in each dish6293T cells, which ensures that the cell density reaches 90% when the transfection is carried out on the next day; three plasmids, namely pHAGE-hACE2-puro, a lentivirus packaging plasmid psPAX and VSVG are added according to the mass ratio of 1: 2: 1 ratio to transfect 293T cells.
Culturing at 4.37 deg.C in 5% incubator for about 48 hr, and collecting cell supernatant according to cell condition. The collected cell supernatant was filtered through a 0.45 μm filter and concentrated with PEG 8000 to obtain a purified hACE2 lentivirus.
5. Spread 5X 10 one day in advance5293T cells in one well of a 12-well plate in the next dayTo the plated cells, 500. mu.L of the virus concentrated in step 2 was added at 1000g, and centrifuged for 2 hours.
6. After completion of the centrifugal infection, the cells were further cultured at 37 ℃ for about 12 hours in a 5% incubator, the medium was changed to a cell culture medium supplemented with 1. mu.g/mL puromycin (puro), and the 293T cells which had the hACE2 gene integrated therein were finally survived, and 293T cells (capable of binding to S protein) which stably expressed hACE2 were selected by flow screening.
VIII, detection method:
blood collection:
at 2/4 weeks after the last immunization, the mice were decapped before death, whole blood was collected from the periphery of the mice by an eye picking method, collected in 1.5mL EP tubes, allowed to stand at room temperature for natural coagulation, and the coagulated serum was centrifuged at 7000g for 15 min. Mouse sera were transferred to new 1.5mL EP tubes. Samples were inactivated at 56 ℃ for 30min prior to the experiment to destroy complement activity in the serum. And the tube is centrifuged for a short time before inactivation, so that the residual samples on the tube wall and the bottle cap are avoided. The bath level should be below the sample level but not above the cap.
ELSIA detection of bound antibodies:
1. the detected antigenic protein (S1, available from Beijing Yiqiao Shenzhou technologies, Inc.; RBD, available from Shanghai, offshore Biotechnologies, Inc.) was diluted with a4 ℃ pre-chilled ELISA coating to a final concentration of 1. mu.g/mL. Add 100. mu.L of the coating antigen solution to each well of the ELISA plate, and keep it at 4 ℃ overnight;
2. the next day, the ELISA plate was removed, the coating solution was discarded, and the plate was washed 3 times with 0.05% PBST buffer, 220. mu.L each time;
3. after washing, patting the mixture on absorbent paper, sealing each hole by using 200 mu L of ELISA sealing solution (0.5% skimmed milk powder, dissolved in PBST), and sealing at room temperature for 2 h;
4. after blocking, wash the plate 3 times with 0.05% PBST, 220 μ L each time;
5. for serum or plasma, dilutions were performed with ELISA sample dilutions (0.5% skim milk powder, PBST lysis) starting from 1:100 and 2-fold dilutions were performed. A negative control was set with non-immunized mouse serum. Setting blank holes, only adding sample diluent, making 2 multiple holes for each sample, wherein the final volume of each hole is 100 mu L, and incubating for 3h at room temperature;
6. after the incubation of the sample is finished, the plate is continuously washed by PBST for 5 times, and each time is 220 mu L;
7. diluting the corresponding proportion of secondary antibody (goat anti-mouse, purchased from Beijing Zhonghua Ching Jinqiao biotechnology Co., Ltd., product No. ZB-2305) with ELISA blocking solution (0.5% skimmed milk powder, PBST dissolved), adding 100 μ L per well, and incubating at room temperature for 1-1.5 h;
8. after the secondary antibody incubation was completed, the plate was washed 5 times with 0.05% PBST, 220 μ L each time;
9. dissolving a pair of gold and silver sheet OPD substrates in 20mL of deionized water, then adding 100 mu L of gold and silver sheet OPD substrates into each hole, and reacting for 5min in a dark place;
10. after the color development was completed, 50. mu.L of 2nM H was used2SO4The termination is performed and the OD is read on the microplate reader492-OD630A value;
11. at the last dilution OD492The reciprocal of the serum dilution ratio corresponding to a value of (negative mean + SD) greater than 2-fold was taken as the antibody titer.
293T-ACE2 cells detect neutralizing antibodies:
1. a96-well transparent bottom blackboard is taken for carrying out a neutralization experiment, a Cell Control (CC) (150 mu L) is arranged in the first column, a Virus Control (VC) (100 mu L) is arranged in the second column, all the other wells are sample wells, and serum samples are diluted in a multiple proportion, so that the volume in the final wells is 100 mu L.
2. In addition to the cell control group, 50. mu.L of SARS-CoV-2 pseudovirus diluent was added to each well, so that each well finally contained pseudovirus at 200TCID50
3. Gently shaking and mixing, placing the blackboard with 96 holes bottom in a cell culture box, keeping the temperature at 37 ℃ and 5% CO2Incubate for 1 h.
4. When the incubation time reached 20min, 293T-hACE2 target cells were initially prepared and diluted to 10 with complete medium5Individual cells/mL.
5. When the incubation time is up to 1h, 100. mu.L of target cells are added to each well of a 96-well transparent bottom blackboard, so that the cells in each well are 104And (4) respectively.
6. Lightly in front, back, left and rightShaking 96-well transparent bottom blackboard to disperse the cells uniformly, and placing the blackboard in a cell culture box at 37 deg.C and 5% CO2Culturing for 48 h.
7. Culturing for 48h, taking out 96-well transparent bottom blackboard from the cell culture box, sucking off supernatant in the wells, adding 100 μ L PBS to each well for washing, sucking off PBS, adding 50 μ L of 1 × lysis buffer (from Cat # E153A of Promega corporation) to each well, and incubating on a horizontal shaker at room temperature for 30min to fully lyse the cells;
8. add 30. mu.L of luciferase substrate (available from Promega, Cat # E1501) to a 96-well blackboard and use the instrument
Figure BDA0003431737740000221
The luciferase activity is detected by a 96-microplate luminescence-detection instrument.
9. Reading values of fluorescein are derived, neutralization inhibition rates are calculated, and ID is calculated by utilizing Graphpad Prism 5.0 software according to the results of the neutralization inhibition rates50
Figure BDA0003431737740000222
ELISPOT detects T cell responses:
mouse spleen single cell isolation:
1) lying the mouse on the back, dissecting the skin of the right abdomen, opening the peritoneum, taking down the spleen of the mouse, and putting the mouse into a small plate added with 5mL of complete RPIM1640 medium;
2) wrapping spleen with sterile gauze with sterile forceps, clamping the gauze with forceps, and lightly grinding spleen to release splenocytes into culture medium;
3) then sucking the spleen cell suspension into a sterile 15mL centrifuge tube through a gauze by using a 5mL pipette, and centrifuging for 5min at 800 g;
4) discarding the centrifuged supernatant, tapping 15mL centrifuge tubes to resuspend cell precipitates, adding 3mL erythrocyte lysate into each centrifuge tube to lyse erythrocytes, reversing and uniformly mixing, and standing at room temperature for 5min to fully lyse erythrocytes without damaging splenocytes;
5) after completion of the flushing, the flushing was stopped with 5 mlpep im1640 medium. 800g, centrifuging for 5 min;
6) discarding the centrifuged supernatant, washing with 5mL of RPIM1640 medium for 1 time, and centrifuging at 800g for 5 min;
7) the supernatant after centrifugation was discarded, and the spleen cells were frozen in a freezing medium (90% FBS and 10% DMSO) for use.
ELISpot experimental procedures were performed according to the Mouse IFN-. gamma./Monkey IFN-. gamma.instructions.
1) Millipore plates provided in a kit (purchased from BD, cat # 551083) were coated with purified IFN- γ antibodies, ratio 1: coating at 250, 4 ℃ overnight;
2) spin off the coated antibody solution in the plate, wash the plate once with 200 μ L RPMI 1640 complete medium, then block the Millipore plate with 200 μ L RPMI 1640 complete medium blocking solution, incubate for 2h at room temperature;
3) the blocking solution was discarded from the well plate and, according to various experimental designs, a pool of stimulating peptides was added to the Millipore plates at 50. mu.L/well and each peptide was at a concentration of 5. mu.g/mL. Add 50. mu.L of RPMI 1640 complete medium to the negative control wells; add 50. mu.L phorbol ester polyclonal stimulator (PMA, purchased from Sigma, cat. FXP012) (final concentration 100ng/mL) and Ionomycin (Ionomycin, final concentration 2. mu.g/mL) RPMI 1640 complete medium to the positive control wells;
wherein, the stimulating peptide library is synthesized by Suzhou Qiangyao biotechnology, Inc., each single peptide is 15 amino acids, covers RBD sequence, and one peptide library is formed by every five single peptides, namely 13 peptide libraries in total. Wherein, the 1 st peptide is MPTESIVRFPNITNL (SEQ ID NO:19), the 2 nd peptide is aa 8-22 in the RBD peptide sequence of SEQ ID NO:3, the following peptides are each shifted backward by four positions compared with other peptide amino acids, namely, the 3 rd peptide is aa 12-26 in the RBD peptide sequence of SEQ ID NO:3, the 4 th peptide is aa 16-30 … … in the RBD peptide sequence of SEQ ID NO:3, and so on until the 64 th peptide is aa 256-270 in the RBD peptide sequence of SEQ ID NO:3, and the 65 th peptide is AVRDPQTLEILDITP (SEQ ID NO: 20).
4) Counting the mouse spleen cells, adjusting the cells to 4 × 106cells/mL, 50. mu.L of cells per well, resulting in a 2X 10 cell count per well5And (4) cells. Mixing MillipoThe re plates were placed in a wet box at 37 5% CO2Incubating in incubator for 20-22h without shaking the plate to avoid cell migration;
5) after the culture incubation is finished, taking out the Millipore plate from the incubator, discarding the liquid in the plate, washing twice with precooled deionized water, wherein each time is 220 mu L, and each time is washed and incubated for 3 min;
6) wash the plate 3 times with 0.05% PBST (PBS + 0.05% Tween-20), 200. mu.L each time;
7) the Biotinylated Detection antibody was diluted with 10% FBS in PBS antibody diluent (Biotinylated Detection antibody, ratio 1: 200) adding 100 mu L of the mixture into each hole, and incubating for 2 hours at room temperature;
8) after incubation, wash the plate 3 times with 0.05% PBST, 220 μ L each time;
9) the streptavidin-HRP conjugated antibody was diluted with an antibody diluent (ratio 1: 100) adding 100 mu L of the mixture into each hole, and incubating for 1h at room temperature;
10) after incubation, wash the plate 4 times with 0.05% PBST, 220 μ L each time;
11) washing the plate with PBS for 2 times, 220 μ L each time;
12) a substrate solution (1mL of substrate buffer plus 1 drop of substrate solution) was prepared and 100. mu.L of substrate solution was added to each well. The reaction is carried out for 5-60min, and the incubation time is determined according to the spot formation condition.
13) Washing with deionized water to terminate the reaction, air drying at room temperature, and counting;
14) counting of Spot Forming Cells (SFC) and QC were performed using a champshot type III enzyme linked spot image analyzer.
Example 1: construction of S protein eukaryotic expression vector and RHAF and RBD-HA2-CD8TM adenovirus expression vector and protein expression
To study the function of RBD, we constructed eukaryotic expression vectors for the S protein and expressed the mature protein in vitro in a cell line.
First, we artificially synthesized the S gene (the corresponding amino acid sequence is shown in SEQ ID NO:11, and the gene sequence is shown in SEQ ID NO: 12), the synthesized fragment was digested with vector plasmid pcDNA3.1 (purchased from Youbao, Inc., FD0054) and Not1 (Thermo Scientific Inc., FD0596) using BamHI digestion (Thermo Scientific Inc., FD0054), and recovered by gel electrophoresis followed by gel cutting, and the digested fragment was recovered using a Sanprep column DNA gel recovery kit (Promega, Cat.A 9282).
Subsequently, the gene recovery product was ligated with the digested linearized vector by the method of T4 DNA ligase (Thermo Scientific Co., Ltd., cat. No. 2011A): coli Stable was transformed with the ligation product and grown overnight on ampicillin-containing plates. On day 2, single colony was randomly selected for sequencing, mutation site correction, and after all sequences were verified to be correct, the eukaryotic expression vector pcDNA3.1-S for the S protein was successfully cloned, and the plasmid construction map is shown in FIG. 1A.
We further examined whether the S protein could be expressed in the eukaryotic cell line 293T.
First, 6-well plates were prepared and seeded at approximately 6X 10 in each well5293T cells, which ensures that the cell density reaches 90% when the transfection is carried out on the next day; 293T cells (transfection reagent TurboFect) were transfected with pcDNA3.1 and pcDNA3.1-S, respectively. Culturing at 37 deg.C in 5% incubator for about 48 hr, collecting cells according to cell condition, performing Western Blotting (WB) identification, and finding that S protein is expressed in cells and has a size of about 200 kD; whereas the expression of the S protein could not be detected in the pcDNA3.1 control transfected cells (FIG. 1B left panel).
To investigate the function of HA2, we constructed an adenoviral expression vector for RHAF (i.e., sRBD-HA2-hFn fusion protein), and adenovirus AdC68-RHAF, and verified the expression of RHAF in adenovirus.
First, we artificially synthesized the RHAF gene (the specific sequence is shown in SEQ ID NO:10, wherein the HA2 part is based on the sequence shown in SEQ ID NO:2, the sRBD part is based on the sequence shown in SEQ ID NO:6, and the hFn part is based on the sequence shown in SEQ ID NO: 8). sRGB-HA 2 was expressed at the N-terminus of hFn with G between4And connecting the S joint. The synthetic fragment was ligated with the vector plasmid pAdC68XY3 (constructed according to the method described in patent application No. 201910777937.2, in which the E4 portion of the E1/E3 deleted replication deficient AdC68 vector was replaced with the corresponding E4 portion of AdHu5In addition, the modification can increase the yield of recombinant chimpanzee adenovirus without changing the serological properties of adenovirus), using SrfI restriction enzyme (NEB corporation, # R0629S), and recovering the fragments by gel electrophoresis followed by gel cutting, and recovering the fragments using a Sanprep column DNA gel recovery kit (Promega corporation, cat. No. A9282).
Subsequently, the gene recovery product was ligated with the digested linearized vector by the method of T4 DNA ligase (Thermo Scientific Co., Ltd., cat. No. 2011A): coli Stable was transformed with the ligation product and grown overnight on ampicillin-containing plates. On day 2, single colonies were randomly picked for sequencing, mutation site correction, and after all sequences were verified to be correct, the RHAF adenovirus expression vector pAdC68XY3-RHAF was successfully cloned, and the plasmid construction map is shown in FIG. 1A.
In order to research whether RBD-HA2 and hFn are fused and secreted or RBD-HA2 is expressed on the surface of adenovirus through a CD8 transmembrane region (CD8TM) to have better effect, an adenovirus expression vector of RBD-HA2-CD8TM and adenovirus AdC68-RBD-HA2-CD8TM are constructed. The construction method is consistent with that of AdC68-RHAF, and the amino acid sequence is shown as SEQ ID NO: 15 and the coding sequence is shown in SEQ ID NO 16. The plasmid construction map is shown in FIG. 1A.
We further packaged and amplified the recombinant adenovirus.
The constructed recombinant plasmids pAdC68XY3-RHAF and pAdC68XY3-RBD-HA2-CD8TM are respectively linearized for 3.5h in a water bath at 37 ℃ by restriction endonuclease Pac 1, and the endonuclease is inactivated at 65 ℃. The method comprises the steps of paving a six-hole plate with 293A cells, transfecting linearized recombinant plasmids to the 293A cells in the amounts of 2.5 mu g, 2 mu g and 1.5 mu g per hole respectively, culturing for about 11 days to enable plaques to appear, collecting samples after about 14 days to enable the cells to be infected by viruses, collecting cell supernatants together, repeatedly freezing and thawing for three times at-80 ℃, taking a small amount of infected 293A cells, and verifying the expression of target proteins by Western blot to find the expression of proteins with correct sizes (figure 1B).
Infecting the collected adenovirus sample to 293A cells of 1T 175 cell culture bottle, collecting the sample after 24h, placing the sample in a culture bottle for repeated freeze thawing at minus 80 ℃ for three times, infecting the sample to 293A cells of 6T 175 cell culture bottles, carrying out mass amplification in the same way, collecting cell precipitates when the sample is amplified to 36T 175 cell culture bottles, abandoning supernatant, resuspending the cell precipitates by using about 10mL of serum-free and non-resistant DMED culture medium, purifying adenovirus by adopting a cesium chloride density gradient centrifugation method after repeated freeze thawing at minus 80 ℃ for three times, and subpackaging the adenovirus sample for preservation at minus 80 ℃.
Cesium chloride density gradient adenovirus step:
(1) centrifuging the adenovirus subjected to freeze thawing at 4 ℃ and 4000g for 30min, taking the supernatant, and filtering with a 0.8-micron filter membrane;
(2) adding 3mL of 1.2M CsC solution into an ultracentrifuge tube, gently and slowly injecting the 3mL of 1.4CsC solution from the bottom by using an injector, and paving a cesium chloride gradient;
(3) gently adding the filtered adenovirus solution into an ultracentrifuge tube, filling the liquid level with PBS, and placing the solution into an SW41 sleeve after balancing;
(4) selecting an SW41 rotor, centrifuging at 4 ℃, 25000rpm for 2.5h, and reducing the speed to be no brake natural speed reduction;
(5) after the centrifugation is finished, the ultracentrifuge tube is gently taken out, two white virus strips can be seen, the ultracentrifuge tube is poked into the ultrariser tube from the side by using an injector, the lower layer strip is sucked out, and the phenomenon that the upper layer strip is sucked is avoided;
(6) injecting Bio-Gel P-6PG Gel desalting glue into a Liquid chromatography column (Liquid chromatography columns), and adding PBS for washing twice when Liquid falls off and glue of about 2-3cm is left;
(7) adding the virus solution into desalting gel, adding PBS (2 mL) for elution after the virus solution completely falls off, and collecting 3-4 drops of eluent in each 1.5mL EP tube;
(8) nanogrop 2000 determination of OD of eluate from tube260Value, OD260Mixing the eluates above 2, adding 10% sterile glycerol, and packaging;
(9) nanogrop 2000 determination of OD of Adenoviral solution after subpackaging260Value, OD260 Value X1.1X 1012the/mL is the titer of the purified adenovirus;
(10) the final AdC68-RHAF titer was 1.76X 1013The titer of AdC68-RBD-HA2-CD8TM is 1.48X 1013/mL。
Example 2: immunogenicity of AdC68-RHAF in C57BL/6 mice
The C57BL/6 mice were immunized with DNA-pcDNA3.1-S and adenovirus AdC68-RHAF, respectively, and the immune combinations were evaluated for induction of the titers of binding antibodies against the RBD protein and H7 protein and neutralizing antibodies against the SARS-CoV-2 pseudovirus 2 after 2 weeks of immunization. The level of T cell responses to the RBD peptide pool was also assessed.
Mice were randomly divided into 2 groups, and named control group and AdC68-RHAF group, respectively, based on the immunogen. The specific immunization combination is shown in table 1, and the immunization mode is intramuscular injection.
The AdC68-RHAF group generated binding antibody titers against RBD protein and H7 protein as shown in FIG. 2A: the titers of binding antibodies to the RBD protein were all above 10,000, up to 50,000; the mean of bound antibody titers against H7 was around 6,000, with the highest being able to reach 10,000. Also, comparing the data of the binding antibodies to the RBD protein before and after the second immunization, it was found that there was a significant increase in the titer of the binding antibodies (fig. 2B).
The AdC68-RHAF group generated neutralizing antibody titers against SARS-Cov-2 pseudovirus as shown in FIG. 2C: most titers were above 1000, the mean was 1360, and there was a neutralizing antibody titer up to 12808 in one mouse, with individual variability between mice. Simultaneously comparing the data of the neutralizing antibody titer against SARS-Cov-2 pseudovirus before and after the second needle immunization, it was found that the neutralizing antibody titer was significantly increased (FIG. 2D).
The AdC68-RHAF group generated T cell responses against the RBD peptide library as shown in FIG. 2E: the number of IFN-gamma secreting cells per million splenocytes is around 3,000.
This experiment demonstrated that AdC68-RHAF was able to induce antibodies against influenza and new corona simultaneously, and could induce neutralizing antibodies against new corona virus and T cell responses against new corona virus in C57BL/6 mice.
TABLE 1 immunization protocol for the immunogenicity test of AdC68-RHAF in C57BL/6 mice
Figure BDA0003431737740000271
Example 3: immunogenicity of AdC68-RHAF in hACE2+ ICR mice
According to the conclusions drawn in example 2, in order to be able to subsequently verify the protection of mice against challenge with the new coronavirus, we verified the immunogenicity of AdC68-RHAF in hACE2+ ICR mice and evaluated the immunological combination to induce a binding antibody titer against RBD protein, H3 protein and H7 protein and a neutralizing antibody titer against SARS-CoV-2 pseudovirus 2 weeks after completion of the immunization.
The hACE2+ ICR mouse used in this example is a COVID-19 preclinical model, which is a SARS-CoV-2 susceptible human angiotensin converting enzyme 2(hACE2) transgenic mouse constructed in the ICR mouse background, and expresses hACE2 mainly in the lung, heart, kidney and small intestine. The mouse model is an important tool for researching and developing SARS-CoV-2 therapeutic drugs and vaccines.
The experimental procedure was as follows: mice were randomly divided into 2 groups, designated control group and AdC68-RHAF group, respectively, based on immunogen. Specific immunization combinations are shown in table 2, and the immunization regimen was intramuscular injection.
The AdC68-RHAF combination produced binding antibody titers against the RBD, H3 and H7 proteins as shown in fig. 3A: the titers of binding antibodies against RBD proteins are mostly above 10,000, and in part can reach 100,000; the titer of the binding antibody against the H3 protein is mostly above 100, and in part can reach 1000; the binding antibody titer against H7 protein was mostly around 6,400.
2 weeks after the end of the second needle immunization, the AdC68-RHAF group generated neutralizing antibody titers against SARS-CoV-2 pseudovirus as shown in FIG. 3B: most of them are about 1000, and the highest can reach 2253.
The experiment proves that AdC68-RHAF can simultaneously induce the combined antibodies aiming at the new corona and the influenza in an hACE2+ ICR mouse, and can simultaneously induce the combined antibodies aiming at the influenza H3 and H7, thereby verifying the broad spectrum of the HA2 antibody. Meanwhile, the AdC68-RHAF can effectively induce a neutralizing antibody aiming at the new corona, and has good prospect of being developed into anti-influenza and new corona vaccines.
TABLE 2 immunization protocol for immunogenicity testing of AdC68-RHAF in hACE2+ ICR mice
Figure BDA0003431737740000281
Example 4: immunogenicity of AdC68-RHAF and AdC68-RBD-HA2-CD8TM in BALB/c mice
BALB/c mice were immunized with AdC68-RHAF and AdC68-RBD-HA2-CD8TM, respectively, and the immune combinations were evaluated to induce the titers of binding antibodies against RBD protein and H7 protein and neutralizing antibodies against SARS-CoV-2 pseudovirus 2 after 2 weeks of immunization. The level of T cell responses to the RBD peptide pool was also assessed.
Mice were randomly divided into 3 groups, designated AdC68, AdC68-RHAF and AdC68-RBD-HA2-CD8TM, respectively, based on the immunogen. Specific combinations of immunizations are shown in table 3, and the immunization regimen was intramuscular injection.
Serum collected one week after the end of immunization detected the bound antibody titer to RBD protein as shown in figure 4A: the mean value of the binding antibody titer of the AdC68-RHAF group against the RBD protein is 4031, and the highest energy is 12,800; the mean binding antibody titer against RBD protein of group AdC68-RBD-HA2-CD8TM was 10,160, the highest being able to reach 25,600, when there was no significant difference between group AdC68-RHAF and group AdC68-RBD-HA2-CD8 TM.
Neutralizing antibody titers against SARS-CoV-2 pseudovirus were detected one week after the end of immunization, as shown in figure 4B: the mean value of neutralizing antibody titer of the AdC68-RHAF group is 128, and the highest energy is 274; the mean value of the neutralizing antibody titer of the AdC68-RBD-HA2-CD8TM group was 97, the highest of which was 348, when there was no significant difference between the AdC68-RHAF group and the AdC68-RBD-HA2-CD8 TM.
The titer of bound antibodies against RBD and the titer of neutralizing antibodies against SARS-CoV-2 pseudovirus were then continuously measured two weeks after the end of immunization.
Meanwhile, the experiment further proves that two adenoviruses AdC68-RHAF and AdC68-RBD-HA2-CD8 TM: effective to induce the production of binding antibodies against influenza virus needle H7; can effectively induce neutralizing antibody against SARS-CoV-2 pseudovirus; at the same time, T cell responses against the RBD peptide pool can be induced.
TABLE 3 immunogenicity testing protocol of AdC68-RHAF and AdC68-RBD-HA2-CD8TM in BALB/c mice
Figure BDA0003431737740000291
Example 5: immunogenicity of AdC68-RHAF in BALB/c mice by different vaccination modes
BALB/c mice were immunized with DNA-pcDNA3.1-S and adenovirus AdC68-RHAF, respectively, and after 2 weeks of immunization, the immune combinations were evaluated for the induction of the titers of binding antibodies against RBD protein and H7 protein and neutralizing antibodies against SARS-CoV-2 pseudovirus. The level of T cell responses to the RBD peptide pool was also assessed.
Mice were randomly divided into 4 groups, and named AdC68 muscle group, AdC68 nasal drip group, AdC68-RHAF muscle group and AdC68-RHAF nasal drip group, respectively, according to immunogen and immunization modes. Specific immunological combinations are shown in table 4.
Collecting serum and lung lavage fluid two weeks after immunization to detect binding antibodies including IgG and IgA against RBD protein and influenza virus H7 protein; neutralizing antibodies against SARS-CoV-2 pseudovirus and T cell responses against the RBD peptide library were simultaneously detected.
TABLE 4 immunogenicity testing protocol generated by different vaccination regimes of AdC68-RHAF in BALB/c mice
Figure BDA0003431737740000292
Figure BDA0003431737740000301
Example 6 challenge test for New coronavirus
Mice: nanamo organism ACE 2-transgenic mice (C57BL/6-Tgtn (CAG-human ACE2-IRES-Luciferase-WPRE-polyA) Smoc), females.
Strain: SARS-CoV-2/human/CHN/Shanghai _ CH-02/2020.
Committing second military medical university to challenge
Immunization procedure: mice were randomly divided into 2 groups, designated control group and AdC68-RHAF group, respectively, based on immunogen. Specific combinations of immunizations are shown in table 5, with the immunization being by intramuscular injection.
And (3) toxin counteracting program: performing challenge test on the mice 1-2 weeks after the immunization is finished, observing for 5 days after challenge, and recording the weight change; 3 mice are taken out of each group to be killed on the third day after the challenge, lung tissues are taken, the left half lung tissue (one big leaf) of each mouse is fixed by 4% paraformaldehyde for 48 hours, and pathological sections are made (H & E staining and organizing); right half lung tissue (4 leaflets) of each mouse was ground for viral load qPCR rnaviruses or viral titer PFU/ml; on the fifth day, if there are more mice alive, the mice are weighed out and then sacrificed altogether.
The results show that: from the viral load, the AdC68-RHAF group was 3.7 Logs lower than the control group, with no viral load detectable in two of the three AdC68-RHAF groups (FIG. 5A). Meanwhile, from the results of H & E staining, the lung tissue pathology was significantly better in the AdC68-RHAF group than in the control group, with less damage and less inflammatory infiltration (fig. 5B). From the body weight, the AdC68-RHAF group was unchanged, while the control group showed a sharp drop on the third day (FIG. 5C). From a survival perspective, the AdC68-RHAF group survived one hundred percent, while the control group all died (FIG. 5D). Overall, all data demonstrate excellent protection of the AdC68-RHAF vaccine against new corona.
TABLE 5 AdC68-RHAF immunization protocol in hACE2+ C57BL/6 mice in vivo
Figure BDA0003431737740000302
Example 7 challenge test for mouse influenza Virus
Mice: hACE2+ ICR mice and BALB/c mice, female.
Strain: H7N9(A/Shanghai/4664T/2013), H3N2(A/Hong Kong/8/68).
The public health clinic centers P2 and P3 in Shanghai city conducted challenge tests.
Immunization procedure: mice were randomly divided into 2 groups, designated control group and AdC68-RHAF group, respectively, based on immunogen. Specific combinations of immunizations are shown in table 6, and the immunization regimen was intramuscular injection.
And (3) toxin counteracting program: and (3) after the immunization is finished, carrying out nasal drop challenge on the mice, continuously observing for 14 days after challenge, weighing every day, recording the weight change, and obtaining a survival curve.
The results showed that for H7N9 challenge, mice in the AdC68-RHAF group had a weight gain at day seven, while the control group had been decreasing (fig. 6A). Whereas from the survival point of view, the AdC68-RHAF group was completely alive, the control group was totally dead on the tenth day (FIG. 6B). For H3N2 challenge, the dose was non-lethal to mice, so that looking at body weight changes alone, it was found that the AdC68-RHAF group had significantly less weight loss than the control group (fig. 6C). In general, AdC68-RHAF can effectively generate protective effect against each test strain, and has broad spectrum.
TABLE 6 AdC68-RHAF immunization protocol in hACE2+ C57BL/6 mice in vivo
Figure BDA0003431737740000311
Example 8 construction of R545-HA2-IntN-PAB-7XHis eukaryotic expression vector and expression of R545-HA2-IntN-PAB-7XHis protein
From the previous experiments, we found that the adenoviral vector vaccine AdC68-RHAF was able to induce antibodies against both new corona and influenza, and both had protective effects. In subsequent experiments, we tried to replace the carrier, i.e. directly using purified nanoparticles for testing. In order to improve the solubility and renaturation rate of protein, we select a shorter RBD peptide fragment (R545, the corresponding amino acid sequence is shown as SEQ ID NO:21, and the gene sequence is shown as SEQ ID NO: 22), which is more favorable for the inclusion body purification of protein than the RBD in the previous paragraph.
In order to express R545-HA2-IntN protein, donor plasmids of R545-HA2-IntN (R545HA-IntN) are constructed, recombinant baculovirus capable of efficiently expressing R545-HA2-IntN in insect cell line Sf9 is obtained through transposition, and mature protein is expressed in insect cell line Sf9 in a eukaryotic mode.
First, we performed codon optimization of insect Sf9 cells for R545-HA2-IntN gene and added lysotropic tags PAB (protein A B domin) and 7XHis tags, artificially synthesized R545-HA2-IntN-PAB-7XHis gene fragment (department of pronunciations, corresponding amino acid sequence of which is shown in SEQ ID NO:23, gene sequence of which is shown in SEQ ID NO: 24), and inserted between digestion sites BamH I (Thermo Scientific, FD0054) and Hind III (Thermo Scientific, FD0504) downstream of PH promoter of pFastBac-Dual plasmid (Sammer Thermo, 10712024) to obtain donor plasmid pFBD-R545HA-IntN-PAB-7XHIS (FIG. 7A).
We then transformed Bacmid-containing DH10Bac competent (exclusively) with donor plasmid pFBD-R545HA-IntN-PAB-7XHis, plated with X-gal, IPTG, kanamycin and gentamicin resistant plates, cultured for 2 days at 37 ℃, picked white colonies and verified by PCR with M13-F/R primer, and picked positive clones to get bR545HA-IntN-PAB-7 XHis.
We then transfected adherent Sf9(Spodoptera frugiperda clone 9, ATCC) cells with bR545HA-IntN-PAB-7XHis, as specified in the Cellffectin II Reagent (Invitrogen, Cat. 10362100) transfection procedure, to obtain P1 generation recombinant baculovirus vAcR545HA-IntN-PAB-7XHisSubstituting P1 for vAcR545HA -IntN-PAB-7XHis600mL of Sf9 cell line suspension-cultured in SF-SFM medium (Womei organism, SF10111-2) was infected at a volume ratio of 1:100, with a cell density of 2X 106The expression and distribution of R545HA-IntN-PAB-7XHis protein was examined by harvesting cell cultures after 5 days/mL (FIG. 7B). The results show that the fusion protein is mostly expressed in cells, so the subsequent purification is performed by adopting an inclusion body purification mode.
Example 9 purification of R545-HA2-IntN-PAB-7XHis protein
To obtain nanoparticles displaying R545-HA2, we first purified the R545HA-IntN-PAB-7XHis protein by the following steps:
(1) to convert vAcR545HA-IntN-PAB-7XHis 600mL Sf9 cells 5 days after infection 2000g, centrifuged 10min, and cells were harvestedPrecipitating;
(2) mu.L of 100 Xprotease complex inhibitor (Producer, cat # 600387) was added to 50mL of cell lysate (50mM Tris-HCl pH 8,0.5M NaCl, 1% NP40), sonicated cell disruptor (SCIENTZ-IID), beaten for 15s, stopped for 20s, and microscopic examination was performed until the cells were completely disrupted.
(3)8000g were centrifuged at 4 ℃ for 20min, the pellet was collected and resuspended in 50mL 50mM Tris-HCl pH 8.5, 50. mu.L of a super nuclease (Tiandi and, PE001B) was added and incubated at room temperature for 20 min.
(4)8000g were centrifuged at 4 ℃ for 20min, and the pellet was collected, resuspended in 50mL of 2M urea (50mM Tris-HCl pH 8,0.5M NaCl, 0.5% NP40, 0.05% Tween), and kept on ice for 5 min.
(5)8000g, centrifuging at 4 deg.C for 20min, collecting precipitate, and repeating step (4).
(6)8000g were centrifuged at 4 ℃ for 20min, and the pellet was resuspended in 50mL of 8M urea (50mM Tris-HCl pH 8,0.5M NaCl), and the pellet was thoroughly solubilized by rotating the freezer overnight.
(7)8000g, centrifuging at 4 deg.C for 20min, collecting supernatant, and filtering with 0.22 μm filter membrane.
(8) After 4mL of High Affinity Ni-Charged Resin FF (Kinsery, L00666) was mixed with 10mL of an equilibration solution (8M urea, 50mM Tris-HCl pH 8,0.5M NaCl), 800g was centrifuged for 5min, and Beads was collected and washed 3 times.
(9) The Beads were resuspended in 2mL of equilibration solution, added to 8M urea lysate, spun at 4 ℃ for 4h, centrifuged at 2000g at 4 ℃ for 10min, and the Beads were collected.
(10) The column efficiency was checked by collecting a portion of the flow stream after complete collection of the Beads on the gravity column with 10mL of equilibration fluid.
(11) The column was washed with 20mL of equilibration buffer until complete drainage.
(12) 20mL of column wash (80mM imidazole, 8M urea, 50mM Tris-HCl pH 8,0.5M NaCl) was added to complete run-off.
(13) 11mL of eluent (300mM imidazole, 8M urea, 50mM Tris-HCl pH 8,0.5M NaCl) was added, the first 1mL of flow was discarded, and 10mL of eluent was collected.
(14) dTT was added to a final concentration of 20mM and spun overnight at 4 ℃.
(15) The eluate was diluted 10-fold (protein concentration less than 50. mu.g/mL) and subjected to gradient renaturation as follows:
i.4m urea, 50mM Tris-HCl PH 8,0.5M NaCl, 0.4mM reduced glutathione, 0.2mM oxidised glutathione, 0.1% PEG6000, 5% glycerol, 4 h.
2M Urea, 50mM Tris-HCl pH 8,0.5M NaCl, 0.4mM reduced glutathione, 0.2mM oxidised glutathione, 0.1% PEG6000, 5% glycerol, 4 h.
1M Urea, 50mM Tris-HCl pH 8,0.5M NaCl, 0.4mM reduced glutathione, 0.2mM oxidised glutathione, 0.1% PEG6000, 5% glycerol, 4 h.
Iv.50mm Tris-HCl PH 8,0.5M NaCl, 2mM dTT, 0.1% PEG6000, 5% glycerol, 4 h.
V.50mm Tris-HCl PH 8,0.5M NaCl, 2mM dTT, 5% glycerol, 4 h.
Pbs PH 7.4, 2mM dTT, 5% glycerol 4 h.
(16) The concentration of the renatured protein was measured by BCA (Sermeflier, cat # 23235). Subpackaging and storing at 4 ℃ for later use (growing at-80 ℃).
The purified protein was stained with WB and Coomassie Brilliant blue to show bands of the correct size (about 70kDa) (FIG. 8A).
Example 10 Assembly of nanoparticles R545HAF
Intein (Intein) is a peptide fragment that can self-cleave. As previously described, the N-terminal of the C-terminal fusion intein of R545-HA2 forms the recombinant protein R545HA-IntN-PAB-7 XHis. Fusing the N end of Ferritin (Ferritin, Fn) and the C end of a joint gbl-intein (gb1 is B domin of Protein G and is used as a solubility promoting label) to form a recombinant Protein gbl-intC-fertin, and then self-assembling the recombinant Protein gbl-intC-fertin into 24 polymers to form nanoparticles; specifically recognizing and cutting ferritin exposed on the surface of the nanoparticle by the N-terminal of the intein; the foreign protein and ferritin are covalently cross-linked to obtain R545-HA2-hFn, which is also in the form of 24-mer nanoparticles.
To investigate the effectiveness of this ligation, we first purified 7XHIS-GB1-IntC-Fn (according to patent application No. 201910421408.9) in the following manner:
(1) pET28a 7XHIS-GB1-IntC-Fn was transformed into the BL21 (Prokineticidae organism, cat # CD901-02) expressing strain.
(2) The monoclonal strains were picked, inoculated into 20mL of kanamycin-chloramphenicol double-resistant LB medium, and shake-cultured overnight at 37 ℃.
(3) The next day, 1:25 was inoculated into 500mL of LB medium with kanamycin resistance, 37 ℃ and 250 rpm.
(4) When the culture was carried out until OD was 0.8-1.0, 0.2mM IPTG was added for induction. After induction at 25 ℃ and 250rpm for 5 hours, the cells were collected by centrifugation at 4000rpm and 4 ℃ for 30 minutes, and the supernatant was discarded.
(5) 500mL of LB-cultured bacteria were resuspended in 30mL of lysis buffer (50mM Tris, pH 7.5, 150mM NaCl, 1% Triton X-100) and then sonicated.
(6) After completion of sonication, the mixture was centrifuged at 12000rpm at 4 ℃ for 30 minutes, and the supernatant was collected.
(7) 4.5g of ammonium sulfate was added to each 30mL of the solution, and the mixture was mixed at 4 ℃ for 15 min.
(8) The resulting mixture was centrifuged at 12000rpm at 4 ℃ for 30 minutes, and the precipitate was collected.
(9) 30mL of Tris-HCl solution (0mM Tris, pH 7.5, 150mM NaCl) was added to dissolve overnight at 4 ℃.
(10) BCA (Sammerfei, cat # 23235) was used to measure the concentration of 7XHIS-GB1-IntC-Fn protein, which was dispensed and stored at 4 ℃ until use (stored at-80 ℃).
The purified 7XHIS-GB1-IntC-Fn was ligated to the purified R545HA-IntN-PAB-7XHis (purified in example 9) at a dTT concentration of 2mM, as shown in FIG. 8B. It can be seen that R545HA-IntN-PAB is able to form a ligation product R545HAF (70 kDa band in the left two lanes of FIG. 8B) of about 70kDa (67 kDa before glycosylation) by ligation of Fn and cleavage generates by-products IntN-PAB (17kDa) and gb1-IntC (11 kDa).
Example 10 immunogenicity of nanoparticle R545HAF in hACE2+ mice
We verified the immunogenicity of the nanoparticle vaccine R545HAF in hACE2+ ICR mice and evaluated the immune combination to induce binding antibody titers against the RBD protein and H7 protein and neutralizing antibody titers against SARS-CoV-2 pseudovirus 4 weeks after completion of the immunization.
The hACE2+ ICR mouse used in this example is a COVID-19 preclinical model, which is a SARS-CoV-2 susceptible human angiotensin converting enzyme 2(hACE2) transgenic mouse constructed in the ICR mouse background, and expresses hACE2 mainly in the lung, heart, kidney and small intestine. The mouse model is an important tool for researching and developing SARS-CoV-2 therapeutic drugs and vaccines.
The experimental procedure was as follows: mice were randomly divided into 2 groups, which were designated as control group and nanoparticle group, respectively, based on the immunogen. Specific combinations of immunizations are shown in table 7, with the immunization being by intramuscular injection.
The titer of the binding antibody against the RBD protein produced by the nanoparticle group, i.e., R545HAF, is as shown in fig. 9A, and the titer of the binding antibody against the RBD protein is mostly over 400,000 and can partially reach 3,000,000 in the fourth week after the completion of immunization; meanwhile, by comparing the data of the binding antibody to the RBD protein after the first immunization with the data of the binding antibody to the RBD protein after the third immunization, it can be found that the titer of the binding antibody is significantly increased (fig. 9B). At the same time, R545HAF was also shown to be effective in inducing binding antibodies against influenza virus H7.
4 weeks after the end of immunization, the nanoparticle group produced neutralizing antibody titers against SARS-CoV-2 pseudovirus as shown in FIG. 9C: most of the total content is about 10,000, and the highest content can reach 92,172; meanwhile, by comparing the data of the binding antibody against the RBD protein after the first immunization with the data of the binding antibody against the RBD protein after the third immunization, it can be found that the titer of the binding antibody is significantly increased (fig. 9D).
This experiment demonstrates that the nanoparticle vaccine R545HAF is able to simultaneously induce binding antibodies against neocorona and influenza in hACE2+ ICR mice. Meanwhile, the nanoparticle vaccine R545HAF can effectively induce a neutralizing antibody aiming at the new corona, and has good prospects in development of anti-influenza and new corona vaccines.
TABLE 7 immunization protocol for immunogenicity testing of R545HAF in hACE2+ ICR mice
Figure BDA0003431737740000351
All documents referred to in this disclosure are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes or modifications to the disclosure may be made by those skilled in the art after reading the above teachings of the disclosure, and such equivalents may fall within the scope of the disclosure as defined by the appended claims.
Appendix: sequence Listing information
Figure BDA0003431737740000361
Sequence listing
<110> Shanghai city public health clinic center
<120> method for simultaneously inducing immune responses against various viruses
<130> 21A805 1CNCN
<160> 26
<170> PatentIn version 3.3
<210> 1
<211> 186
<212> PRT
<213> Influenza Virus (Influenza virus)
<400> 1
Met Gly Leu Phe Gly Ala Ile Ala Gly Phe Ile Glu Asn Gly Trp Glu
1 5 10 15
Gly Leu Ile Asp Gly Trp Tyr Gly Phe Arg His Gln Asn Ala Gln Gly
20 25 30
Glu Gly Thr Ala Ala Asp Tyr Lys Ser Thr Gln Ser Ala Ile Asp Gln
35 40 45
Ile Thr Gly Lys Leu Asn Arg Leu Ile Glu Lys Thr Asn Gln Gln Phe
50 55 60
Glu Leu Ile Asp Asn Glu Phe Asn Glu Val Glu Lys Gln Ile Gly Asn
65 70 75 80
Val Ile Asn Trp Thr Arg Asp Ser Ile Thr Glu Val Trp Ser Tyr Asn
85 90 95
Ala Glu Leu Leu Val Ala Met Glu Asn Gln His Thr Ile Asp Leu Ala
100 105 110
Asp Ser Glu Met Asp Lys Leu Tyr Glu Arg Val Lys Arg Gln Leu Arg
115 120 125
Glu Asn Ala Glu Glu Asp Gly Thr Gly Cys Phe Glu Ile Phe His Lys
130 135 140
Cys Asp Asp Asp Cys Met Ala Ser Ile Arg Asn Asn Thr Tyr Asp His
145 150 155 160
Ser Lys Tyr Arg Glu Glu Ala Met Gln Asn Arg Ile Gln Ile Asp Pro
165 170 175
Val Lys Leu Ser Ser Gly Tyr Lys Asp Val
180 185
<210> 2
<211> 558
<212> DNA
<213> Influenza Virus (Influenza virus)
<400> 2
atgggcctgt tcggcgccat tgccggcttc atcgagaacg gctgggaggg cctcattgac 60
ggctggtacg gcttcaggca ccagaacgcc cagggcgagg gcacagccgc cgactacaag 120
tccacccagt ccgccattga ccagatcaca ggcaagctga acagactcat tgaaaaaaca 180
aaccagcagt tcgagctgat tgacaacgag ttcaacgagg tggagaagca gattggcaac 240
gtgattaact ggacacggga ctctattaca gaggtgtggt cttacaacgc cgagttactc 300
gtggcaatgg agaaccagca cacaattgac ctcgccgact ctgagatgga caagctgtac 360
gagcgggtga agagacagct cagagagaac gccgaggagg acggcacagg ctgcttcgag 420
atattccaca agtgcgacga cgactgcatg gccagcatcc ggaacaacac atacgaccac 480
tctaagtacc gggaggaggc catgcagaac cgcatccaga ttgacccagt gaagctgtct 540
agcggctaca aggacgtg 558
<210> 3
<211> 272
<212> PRT
<213> novel coronaviruses (COVID-19)
<400> 3
Met Arg Val Gln Pro Thr Glu Ser Ile Val Arg Phe Pro Asn Ile Thr
1 5 10 15
Asn Leu Cys Pro Phe Gly Glu Val Phe Asn Ala Thr Arg Phe Ala Ser
20 25 30
Val Tyr Ala Trp Asn Arg Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr
35 40 45
Ser Val Leu Tyr Asn Ser Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly
50 55 60
Val Ser Pro Thr Lys Leu Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala
65 70 75 80
Asp Ser Phe Val Ile Arg Gly Asp Glu Val Arg Gln Ile Ala Pro Gly
85 90 95
Gln Thr Gly Lys Ile Ala Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe
100 105 110
Thr Gly Cys Val Ile Ala Trp Asn Ser Asn Asn Leu Asp Ser Lys Val
115 120 125
Gly Gly Asn Tyr Asn Tyr Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu
130 135 140
Lys Pro Phe Glu Arg Asp Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser
145 150 155 160
Thr Pro Cys Asn Gly Val Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln
165 170 175
Ser Tyr Gly Phe Gln Pro Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg
180 185 190
Val Val Val Leu Ser Phe Glu Leu Leu His Ala Pro Ala Thr Val Cys
195 200 205
Gly Pro Lys Lys Ser Thr Asn Leu Val Lys Asn Lys Cys Val Asn Phe
210 215 220
Asn Phe Asn Gly Leu Thr Gly Thr Gly Val Leu Thr Glu Ser Asn Lys
225 230 235 240
Lys Phe Leu Pro Phe Gln Gln Phe Gly Arg Asp Ile Ala Asp Thr Thr
245 250 255
Asp Ala Val Arg Asp Pro Gln Thr Leu Glu Ile Leu Asp Ile Thr Pro
260 265 270
<210> 4
<211> 816
<212> DNA
<213> novel coronaviruses (COVID-19)
<400> 4
atgagggtgc agcctaccga gtccatcgtg cgctttccca atatcacaaa cctgtgccct 60
tttggcgagg tgttcaacgc aacccgcttc gcaagcgtgt acgcctggaa taggaagcgc 120
atctccaact gcgtggccga ctattctgtg ctgtacaaca gcgcctcctt ctctaccttt 180
aagtgctatg gcgtgagccc cacaaagctg aatgacctgt gctttaccaa cgtgtacgcc 240
gattccttcg tgatcagggg cgacgaggtg cgccagatcg caccaggaca gacaggcaag 300
atcgcagact acaattataa gctgcctgac gatttcaccg gctgcgtgat cgcctggaac 360
tctaacaatc tggatagcaa agtgggcggc aactacaatt atctgtaccg gctgtttaga 420
aagtctaatc tgaagccatt cgagagggac atctccacag aaatctacca ggccggctct 480
accccctgca atggcgtgga gggctttaac tgttatttcc ctctgcagag ctacggcttc 540
cagccaacaa acggcgtggg ctatcagccc taccgcgtgg tggtgctgtc ttttgagctg 600
ctgcacgcac ctgcaacagt gtgcggacca aagaagagca ccaatctggt gaagaacaag 660
tgcgtgaact tcaacttcaa cggactgacc ggcacaggcg tgctgaccga gtccaacaag 720
aagttcctgc cttttcagca gttcggcagg gacatcgcag ataccacaga cgccgtgcgc 780
gaccctcaga ccctggagat cctggatatc acacca 816
<210> 5
<211> 272
<212> PRT
<213> Artificial sequence
<400> 5
Met Val Cys Pro Thr Glu Ser Ile Val Arg Phe Pro Asn Ile Thr Asn
1 5 10 15
Leu Cys Pro Phe Gly Glu Val Phe Asn Ala Thr Arg Phe Ala Ser Val
20 25 30
Tyr Ala Trp Asn Arg Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser
35 40 45
Val Leu Tyr Asn Ser Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val
50 55 60
Ser Pro Thr Lys Leu Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp
65 70 75 80
Ser Phe Val Ile Arg Gly Asp Glu Val Arg Gln Ile Ala Pro Gly Gln
85 90 95
Thr Gly Lys Ile Ala Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr
100 105 110
Gly Cys Val Ile Ala Trp Asn Ser Asn Asn Leu Asp Ser Lys Val Gly
115 120 125
Gly Asn Tyr Asn Tyr Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys
130 135 140
Pro Phe Glu Arg Asp Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr
145 150 155 160
Pro Cys Asn Gly Val Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser
165 170 175
Tyr Gly Phe Gln Pro Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val
180 185 190
Val Val Leu Ser Phe Glu Leu Leu His Ala Pro Ala Thr Val Cys Gly
195 200 205
Pro Lys Lys Ser Thr Asn Leu Val Lys Asn Lys Cys Val Asn Phe Asn
210 215 220
Phe Asn Gly Leu Thr Gly Thr Gly Val Leu Thr Glu Ser Asn Lys Lys
225 230 235 240
Phe Leu Pro Phe Gln Gln Phe Gly Arg Asp Ile Ala Asp Thr Thr Asp
245 250 255
Ala Val Arg Asp Pro Gln Thr Leu Glu Ile Leu Asp Ile Thr Pro Cys
260 265 270
<210> 6
<211> 816
<212> DNA
<213> Artificial sequence
<400> 6
atggtgtgtc ctaccgagtc catcgtgcgc tttcccaata tcacaaacct gtgccctttt 60
ggcgaggtgt tcaacgcaac ccgcttcgca agcgtgtacg cctggaatag gaagcgcatc 120
tccaactgcg tggccgacta ttctgtgctg tacaacagcg cctccttctc tacctttaag 180
tgctatggcg tgagccccac aaagctgaat gacctgtgct ttaccaacgt gtacgccgat 240
tccttcgtga tcaggggcga cgaggtgcgc cagatcgcac caggacagac aggcaagatc 300
gcagactaca attataagct gcctgacgat ttcaccggct gcgtgatcgc ctggaactct 360
aacaatctgg atagcaaagt gggcggcaac tacaattatc tgtaccggct gtttagaaag 420
tctaatctga agccattcga gagggacatc tccacagaaa tctaccaggc cggctctacc 480
ccctgcaatg gcgtggaggg ctttaactgt tatttccctc tgcagagcta cggcttccag 540
ccaacaaacg gcgtgggcta tcagccctac cgcgtggtgg tgctgtcttt tgagctgctg 600
cacgcacctg caacagtgtg cggaccaaag aagagcacca atctggtgaa gaacaagtgc 660
gtgaacttca acttcaacgg actgaccggc acaggcgtgc tgaccgagtc caacaagaag 720
ttcctgcctt ttcagcagtt cggcagggac atcgcagata ccacagacgc cgtgcgcgac 780
cctcagaccc tggagatcct ggatatcaca ccatgc 816
<210> 7
<211> 183
<212> PRT
<213> Artificial sequence
<400> 7
Met Thr Thr Ala Ser Thr Ser Gln Val Arg Gln Asn Tyr His Gln Asp
1 5 10 15
Ser Glu Ala Ala Ile Asn Arg Gln Ile Asn Leu Glu Leu Tyr Ala Ser
20 25 30
Tyr Val Tyr Leu Ser Met Ser Tyr Tyr Phe Asp Arg Asp Asp Val Ala
35 40 45
Leu Lys Asn Phe Ala Lys Tyr Phe Leu His Gln Ser His Glu Glu Arg
50 55 60
Glu His Ala Glu Lys Leu Met Lys Leu Gln Asn Gln Arg Gly Gly Arg
65 70 75 80
Ile Phe Leu Gln Asp Ile Lys Lys Pro Asp Cys Asp Asp Trp Glu Ser
85 90 95
Gly Leu Asn Ala Met Glu Cys Ala Leu His Leu Glu Lys Asn Val Asn
100 105 110
Gln Ser Leu Leu Glu Leu His Lys Leu Ala Thr Asp Lys Asn Asp Pro
115 120 125
His Leu Cys Asp Phe Ile Glu Thr His Tyr Leu Asn Glu Gln Val Lys
130 135 140
Ala Ile Lys Glu Leu Gly Asp His Val Thr Asn Leu Arg Lys Met Gly
145 150 155 160
Ala Pro Glu Ser Gly Leu Ala Glu Tyr Leu Phe Asp Lys His Thr Leu
165 170 175
Gly Asp Ser Asp Asn Glu Ser
180
<210> 8
<211> 549
<212> DNA
<213> Artificial sequence
<400> 8
atgacgaccg cgtccacctc gcaggtgcgc cagaactacc accaggactc agaggccgcc 60
atcaaccgcc agatcaacct ggagctctac gcctcctacg tttacctgtc catgtcttac 120
tactttgacc gcgatgatgt ggctttgaag aactttgcca aatactttct tcaccaatct 180
catgaggaga gggaacatgc tgagaaactg atgaagctgc agaaccaacg aggtggccga 240
atcttccttc aggatatcaa gaaaccagac tgtgatgact gggagagcgg gctgaatgca 300
atggagtgtg cattacattt ggaaaaaaat gtgaatcagt cactactgga actgcacaaa 360
ctggccactg acaaaaatga cccccatttg tgtgacttca ttgagacaca ttacctgaat 420
gagcaggtga aagccatcaa agaattgggt gaccacgtga ccaacttgcg caagatggga 480
gcgcccgaat ctggcttggc ggaatatctc tttgacaagc acaccctggg agacagtgat 540
aatgaaagc 549
<210> 9
<211> 684
<212> PRT
<213> Artificial sequence
<400> 9
Met Val Phe Thr Pro Gln Ile Leu Gly Leu Met Leu Phe Trp Ile Ser
1 5 10 15
Ala Ser Arg Gly Ser Tyr Tyr His His His His His His Val Cys Pro
20 25 30
Thr Glu Ser Ile Val Arg Phe Pro Asn Ile Thr Asn Leu Cys Pro Phe
35 40 45
Gly Glu Val Phe Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala Trp Asn
50 55 60
Arg Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser Val Leu Tyr Asn
65 70 75 80
Ser Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro Thr Lys
85 90 95
Leu Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe Val Ile
100 105 110
Arg Gly Asp Glu Val Arg Gln Ile Ala Pro Gly Gln Thr Gly Lys Ile
115 120 125
Ala Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys Val Ile
130 135 140
Ala Trp Asn Ser Asn Asn Leu Asp Ser Lys Val Gly Gly Asn Tyr Asn
145 150 155 160
Tyr Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe Glu Arg
165 170 175
Asp Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys Asn Gly
180 185 190
Val Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly Phe Gln
195 200 205
Pro Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val Val Val Leu Ser
210 215 220
Phe Glu Leu Leu His Ala Pro Ala Thr Val Cys Gly Pro Lys Lys Ser
225 230 235 240
Thr Asn Leu Val Lys Asn Lys Cys Val Asn Phe Asn Phe Asn Gly Leu
245 250 255
Thr Gly Thr Gly Val Leu Thr Glu Ser Asn Lys Lys Phe Leu Pro Phe
260 265 270
Gln Gln Phe Gly Arg Asp Ile Ala Asp Thr Thr Asp Ala Val Arg Asp
275 280 285
Pro Gln Thr Leu Glu Ile Leu Asp Ile Thr Pro Cys Ser Val Asp Glu
290 295 300
Leu Thr Ser Arg Gly Arg Gly Leu Phe Gly Ala Ile Ala Gly Phe Ile
305 310 315 320
Glu Asn Gly Trp Glu Gly Leu Ile Asp Gly Trp Tyr Gly Phe Arg His
325 330 335
Gln Asn Ala Gln Gly Glu Gly Thr Ala Ala Asp Tyr Lys Ser Thr Gln
340 345 350
Ser Ala Ile Asp Gln Ile Thr Gly Lys Leu Asn Arg Leu Ile Glu Lys
355 360 365
Thr Asn Gln Gln Phe Glu Leu Ile Asp Asn Glu Phe Asn Glu Val Glu
370 375 380
Lys Gln Ile Gly Asn Val Ile Asn Trp Thr Arg Asp Ser Ile Thr Glu
385 390 395 400
Val Trp Ser Tyr Asn Ala Glu Leu Leu Val Ala Met Glu Asn Gln His
405 410 415
Thr Ile Asp Leu Ala Asp Ser Glu Met Asp Lys Leu Tyr Glu Arg Val
420 425 430
Lys Arg Gln Leu Arg Glu Asn Ala Glu Glu Asp Gly Thr Gly Cys Phe
435 440 445
Glu Ile Phe His Lys Cys Asp Asp Asp Cys Met Ala Ser Ile Arg Asn
450 455 460
Asn Thr Tyr Asp His Ser Lys Tyr Arg Glu Glu Ala Met Gln Asn Arg
465 470 475 480
Ile Gln Ile Asp Pro Val Lys Leu Ser Ser Gly Tyr Lys Asp Val Val
485 490 495
Asp Gly Gly Gly Gly Ser Thr Thr Ala Ser Thr Ser Gln Val Arg Gln
500 505 510
Asn Tyr His Gln Asp Ser Glu Ala Ala Ile Asn Arg Gln Ile Asn Leu
515 520 525
Glu Leu Tyr Ala Ser Tyr Val Tyr Leu Ser Met Ser Tyr Tyr Phe Asp
530 535 540
Arg Asp Asp Val Ala Leu Lys Asn Phe Ala Lys Tyr Phe Leu His Gln
545 550 555 560
Ser His Glu Glu Arg Glu His Ala Glu Lys Leu Met Lys Leu Gln Asn
565 570 575
Gln Arg Gly Gly Arg Ile Phe Leu Gln Asp Ile Lys Lys Pro Asp Cys
580 585 590
Asp Asp Trp Glu Ser Gly Leu Asn Ala Met Glu Cys Ala Leu His Leu
595 600 605
Glu Lys Asn Val Asn Gln Ser Leu Leu Glu Leu His Lys Leu Ala Thr
610 615 620
Asp Lys Asn Asp Pro His Leu Cys Asp Phe Ile Glu Thr His Tyr Leu
625 630 635 640
Asn Glu Gln Val Lys Ala Ile Lys Glu Leu Gly Asp His Val Thr Asn
645 650 655
Leu Arg Lys Met Gly Ala Pro Glu Ser Gly Leu Ala Glu Tyr Leu Phe
660 665 670
Asp Lys His Thr Leu Gly Asp Ser Asp Asn Glu Ser
675 680
<210> 10
<211> 2052
<212> DNA
<213> Artificial sequence
<400> 10
atggttttca cacctcagat acttggactt atgctttttt ggatttcagc ctccagaggt 60
tcgtactacc atcaccatca ccatcacgtg tgtcctaccg agtccatcgt gcgctttccc 120
aatatcacaa acctgtgccc ttttggcgag gtgttcaacg caacccgctt cgcaagcgtg 180
tacgcctgga ataggaagcg catctccaac tgcgtggccg actattctgt gctgtacaac 240
agcgcctcct tctctacctt taagtgctat ggcgtgagcc ccacaaagct gaatgacctg 300
tgctttacca acgtgtacgc cgattccttc gtgatcaggg gcgacgaggt gcgccagatc 360
gcaccaggac agacaggcaa gatcgcagac tacaattata agctgcctga cgatttcacc 420
ggctgcgtga tcgcctggaa ctctaacaat ctggatagca aagtgggcgg caactacaat 480
tatctgtacc ggctgtttag aaagtctaat ctgaagccat tcgagaggga catctccaca 540
gaaatctacc aggccggctc taccccctgc aatggcgtgg agggctttaa ctgttatttc 600
cctctgcaga gctacggctt ccagccaaca aacggcgtgg gctatcagcc ctaccgcgtg 660
gtggtgctgt cttttgagct gctgcacgca cctgcaacag tgtgcggacc aaagaagagc 720
accaatctgg tgaagaacaa gtgcgtgaac ttcaacttca acggactgac cggcacaggc 780
gtgctgaccg agtccaacaa gaagttcctg ccttttcagc agttcggcag ggacatcgca 840
gataccacag acgccgtgcg cgaccctcag accctggaga tcctggatat cacaccatgc 900
tccgtcgacg agctcactag tcgcggccgc ggcctgttcg gcgccattgc cggcttcatc 960
gagaacggct gggagggcct cattgacggc tggtacggct tcaggcacca gaacgcccag 1020
ggcgagggca cagccgccga ctacaagtcc acccagtccg ccattgacca gatcacaggc 1080
aagctgaaca gactcattga aaaaacaaac cagcagttcg agctgattga caacgagttc 1140
aacgaggtgg agaagcagat tggcaacgtg attaactgga cacgggactc tattacagag 1200
gtgtggtctt acaacgccga gttactcgtg gcaatggaga accagcacac aattgacctc 1260
gccgactctg agatggacaa gctgtacgag cgggtgaaga gacagctcag agagaacgcc 1320
gaggaggacg gcacaggctg cttcgagata ttccacaagt gcgacgacga ctgcatggcc 1380
agcatccgga acaacacata cgaccactct aagtaccggg aggaggccat gcagaaccgc 1440
atccagattg acccagtgaa gctgtctagc ggctacaagg acgtggtcga cggcggaggc 1500
gggagcacga ccgcgtccac ctcgcaggtg cgccagaact accaccagga ctcagaggcc 1560
gccatcaacc gccagatcaa cctggagctc tacgcctcct acgtttacct gtccatgtct 1620
tactactttg accgcgatga tgtggctttg aagaactttg ccaaatactt tcttcaccaa 1680
tctcatgagg agagggaaca tgctgagaaa ctgatgaagc tgcagaacca acgaggtggc 1740
cgaatcttcc ttcaggatat caagaaacca gactgtgatg actgggagag cgggctgaat 1800
gcaatggagt gtgcattaca tttggaaaaa aatgtgaatc agtcactact ggaactgcac 1860
aaactggcca ctgacaaaaa tgacccccat ttgtgtgact tcattgagac acattacctg 1920
aatgagcagg tgaaagccat caaagaattg ggtgaccacg tgaccaactt gcgcaagatg 1980
ggagcgcccg aatctggctt ggcggaatat ctctttgaca agcacaccct gggagacagt 2040
gataatgaaa gc 2052
<210> 11
<211> 1281
<212> PRT
<213> novel coronaviruses (COVID-19)
<400> 11
Met Phe Val Phe Leu Val Leu Leu Pro Leu Val Ser Ser Gln Cys Val
1 5 10 15
Asn Leu Thr Thr Arg Thr Gln Leu Pro Pro Ala Tyr Thr Asn Ser Phe
20 25 30
Thr Arg Gly Val Tyr Tyr Pro Asp Lys Val Phe Arg Ser Ser Val Leu
35 40 45
His Ser Thr Gln Asp Leu Phe Leu Pro Phe Phe Ser Asn Val Thr Trp
50 55 60
Phe His Ala Ile His Val Ser Gly Thr Asn Gly Thr Lys Arg Phe Asp
65 70 75 80
Asn Pro Val Leu Pro Phe Asn Asp Gly Val Tyr Phe Ala Ser Thr Glu
85 90 95
Lys Ser Asn Ile Ile Arg Gly Trp Ile Phe Gly Thr Thr Leu Asp Ser
100 105 110
Lys Thr Gln Ser Leu Leu Ile Val Asn Asn Ala Thr Asn Val Val Ile
115 120 125
Lys Val Cys Glu Phe Gln Phe Cys Asn Asp Pro Phe Leu Gly Val Tyr
130 135 140
Tyr His Lys Asn Asn Lys Ser Trp Met Glu Ser Glu Phe Arg Val Tyr
145 150 155 160
Ser Ser Ala Asn Asn Cys Thr Phe Glu Tyr Val Ser Gln Pro Phe Leu
165 170 175
Met Asp Leu Glu Gly Lys Gln Gly Asn Phe Lys Asn Leu Arg Glu Phe
180 185 190
Val Phe Lys Asn Ile Asp Gly Tyr Phe Lys Ile Tyr Ser Lys His Thr
195 200 205
Pro Ile Asn Leu Val Arg Asp Leu Pro Gln Gly Phe Ser Ala Leu Glu
210 215 220
Pro Leu Val Asp Leu Pro Ile Gly Ile Asn Ile Thr Arg Phe Gln Thr
225 230 235 240
Leu Leu Ala Leu His Arg Ser Tyr Leu Thr Pro Gly Asp Ser Ser Ser
245 250 255
Gly Trp Thr Ala Gly Ala Ala Ala Tyr Tyr Val Gly Tyr Leu Gln Pro
260 265 270
Arg Thr Phe Leu Leu Lys Tyr Asn Glu Asn Gly Thr Ile Thr Asp Ala
275 280 285
Val Asp Cys Ala Leu Asp Pro Leu Ser Glu Thr Lys Cys Thr Leu Lys
290 295 300
Ser Phe Thr Val Glu Lys Gly Ile Tyr Gln Thr Ser Asn Phe Arg Val
305 310 315 320
Gln Pro Thr Glu Ser Ile Val Arg Phe Pro Asn Ile Thr Asn Leu Cys
325 330 335
Pro Phe Gly Glu Val Phe Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala
340 345 350
Trp Asn Arg Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser Val Leu
355 360 365
Tyr Asn Ser Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro
370 375 380
Thr Lys Leu Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe
385 390 395 400
Val Ile Arg Gly Asp Glu Val Arg Gln Ile Ala Pro Gly Gln Thr Gly
405 410 415
Lys Ile Ala Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys
420 425 430
Val Ile Ala Trp Asn Ser Asn Asn Leu Asp Ser Lys Val Gly Gly Asn
435 440 445
Tyr Asn Tyr Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe
450 455 460
Glu Arg Asp Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys
465 470 475 480
Asn Gly Val Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly
485 490 495
Phe Gln Pro Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val Val Val
500 505 510
Leu Ser Phe Glu Leu Leu His Ala Pro Ala Thr Val Cys Gly Pro Lys
515 520 525
Lys Ser Thr Asn Leu Val Lys Asn Lys Cys Val Asn Phe Asn Phe Asn
530 535 540
Gly Leu Thr Gly Thr Gly Val Leu Thr Glu Ser Asn Lys Lys Phe Leu
545 550 555 560
Pro Phe Gln Gln Phe Gly Arg Asp Ile Ala Asp Thr Thr Asp Ala Val
565 570 575
Arg Asp Pro Gln Thr Leu Glu Ile Leu Asp Ile Thr Pro Cys Ser Phe
580 585 590
Gly Gly Val Ser Val Ile Thr Pro Gly Thr Asn Thr Ser Asn Gln Val
595 600 605
Ala Val Leu Tyr Gln Asp Val Asn Cys Thr Glu Val Pro Val Ala Ile
610 615 620
His Ala Asp Gln Leu Thr Pro Thr Trp Arg Val Tyr Ser Thr Gly Ser
625 630 635 640
Asn Val Phe Gln Thr Arg Ala Gly Cys Leu Ile Gly Ala Glu His Val
645 650 655
Asn Asn Ser Tyr Glu Cys Asp Ile Pro Ile Gly Ala Gly Ile Cys Ala
660 665 670
Ser Tyr Gln Thr Gln Thr Asn Ser Pro Arg Arg Ala Arg Ser Val Ala
675 680 685
Ser Gln Ser Ile Ile Ala Tyr Thr Met Ser Leu Gly Ala Glu Asn Ser
690 695 700
Val Ala Tyr Ser Asn Asn Ser Ile Ala Ile Pro Thr Asn Phe Thr Ile
705 710 715 720
Ser Val Thr Thr Glu Ile Leu Pro Val Ser Met Thr Lys Thr Ser Val
725 730 735
Asp Cys Thr Met Tyr Ile Cys Gly Asp Ser Thr Glu Cys Ser Asn Leu
740 745 750
Leu Leu Gln Tyr Gly Ser Phe Cys Thr Gln Leu Asn Arg Ala Leu Thr
755 760 765
Gly Ile Ala Val Glu Gln Asp Lys Asn Thr Gln Glu Val Phe Ala Gln
770 775 780
Val Lys Gln Ile Tyr Lys Thr Pro Pro Ile Lys Asp Phe Gly Gly Phe
785 790 795 800
Asn Phe Ser Gln Ile Leu Pro Asp Pro Ser Lys Pro Ser Lys Arg Ser
805 810 815
Phe Ile Glu Asp Leu Leu Phe Asn Lys Val Thr Leu Ala Asp Ala Gly
820 825 830
Phe Ile Lys Gln Tyr Gly Asp Cys Leu Gly Asp Ile Ala Ala Arg Asp
835 840 845
Leu Ile Cys Ala Gln Lys Phe Asn Gly Leu Thr Val Leu Pro Pro Leu
850 855 860
Leu Thr Asp Glu Met Ile Ala Gln Tyr Thr Ser Ala Leu Leu Ala Gly
865 870 875 880
Thr Ile Thr Ser Gly Trp Thr Phe Gly Ala Gly Ala Ala Leu Gln Ile
885 890 895
Pro Phe Ala Met Gln Met Ala Tyr Arg Phe Asn Gly Ile Gly Val Thr
900 905 910
Gln Asn Val Leu Tyr Glu Asn Gln Lys Leu Ile Ala Asn Gln Phe Asn
915 920 925
Ser Ala Ile Gly Lys Ile Gln Asp Ser Leu Ser Ser Thr Ala Ser Ala
930 935 940
Leu Gly Lys Leu Gln Asp Val Val Asn Gln Asn Ala Gln Ala Leu Asn
945 950 955 960
Thr Leu Val Lys Gln Leu Ser Ser Asn Phe Gly Ala Ile Ser Ser Val
965 970 975
Leu Asn Asp Ile Leu Ser Arg Leu Asp Lys Val Glu Ala Glu Val Gln
980 985 990
Ile Asp Arg Leu Ile Thr Gly Arg Leu Gln Ser Leu Gln Thr Tyr Val
995 1000 1005
Thr Gln Gln Leu Ile Arg Ala Ala Glu Ile Arg Ala Ser Ala Asn
1010 1015 1020
Leu Ala Ala Thr Lys Met Ser Glu Cys Val Leu Gly Gln Ser Lys
1025 1030 1035
Arg Val Asp Phe Cys Gly Lys Gly Tyr His Leu Met Ser Phe Pro
1040 1045 1050
Gln Ser Ala Pro His Gly Val Val Phe Leu His Val Thr Tyr Val
1055 1060 1065
Pro Ala Gln Glu Lys Asn Phe Thr Thr Ala Pro Ala Ile Cys His
1070 1075 1080
Asp Gly Lys Ala His Phe Pro Arg Glu Gly Val Phe Val Ser Asn
1085 1090 1095
Gly Thr His Trp Phe Val Thr Gln Arg Asn Phe Tyr Glu Pro Gln
1100 1105 1110
Ile Ile Thr Thr Asp Asn Thr Phe Val Ser Gly Asn Cys Asp Val
1115 1120 1125
Val Ile Gly Ile Val Asn Asn Thr Val Tyr Asp Pro Leu Gln Pro
1130 1135 1140
Glu Leu Asp Ser Phe Lys Glu Glu Leu Asp Lys Tyr Phe Lys Asn
1145 1150 1155
His Thr Ser Pro Asp Val Asp Leu Gly Asp Ile Ser Gly Ile Asn
1160 1165 1170
Ala Ser Val Val Asn Ile Gln Lys Glu Ile Asp Arg Leu Asn Glu
1175 1180 1185
Val Ala Lys Asn Leu Asn Glu Ser Leu Ile Asp Leu Gln Glu Leu
1190 1195 1200
Gly Lys Tyr Glu Gln Tyr Ile Lys Trp Pro Trp Tyr Ile Trp Leu
1205 1210 1215
Gly Phe Ile Ala Gly Leu Ile Ala Ile Val Met Val Thr Ile Met
1220 1225 1230
Leu Cys Cys Met Thr Ser Cys Cys Ser Cys Leu Lys Gly Cys Cys
1235 1240 1245
Ser Cys Gly Ser Cys Cys Lys Phe Asp Glu Asp Asp Ser Glu Pro
1250 1255 1260
Val Leu Lys Gly Val Lys Leu His Tyr Thr Thr Gly Leu Gln Leu
1265 1270 1275
Ala Arg Val
1280
<210> 12
<211> 3843
<212> DNA
<213> novel coronaviruses (COVID-19)
<400> 12
atgttcgtgt ttctggtgct gctgcctctg gtgagctccc agtgcgtgaa cctgaccaca 60
aggacccagc tgccccctgc ctataccaat tccttcacac ggggcgtgta ctatcccgac 120
aaggtgttcc ggagcagcgt gctgcactcc acacaggatc tgtttctgcc tttcttttct 180
aacgtgacct ggttccacgc catccacgtg agcggcacca atggcacaaa gcggttcgac 240
aatccagtgc tgccctttaa cgatggcgtg tacttcgcct ccaccgagaa gtctaacatc 300
atcagaggct ggatctttgg caccacactg gacagcaaga cacagtccct gctgatcgtg 360
aacaatgcca ccaacgtggt catcaaggtg tgcgagttcc agttttgtaa tgatccattc 420
ctgggcgtgt actatcacaa gaacaataag tcttggatgg agagcgagtt tcgcgtgtat 480
tcctctgcca acaattgcac atttgagtac gtgtcccagc ccttcctgat ggacctggag 540
ggcaagcagg gcaatttcaa gaacctgagg gagttcgtgt ttaagaatat cgatggctac 600
ttcaaaatct actccaagca caccccaatc aacctggtgc gcgacctgcc acagggcttc 660
tctgccctgg agccactggt ggatctgccc atcggcatca acatcacccg gtttcagaca 720
ctgctggccc tgcacagaag ctacctgaca ccaggcgaca gctcctctgg atggaccgca 780
ggagcagcag cctactatgt gggctatctg cagcccagga ccttcctgct gaagtacaac 840
gagaatggca ccatcacaga cgccgtggat tgcgccctgg atcccctgtc tgagaccaag 900
tgtacactga agagctttac cgtggagaag ggcatctatc agacaagcaa tttcagggtg 960
cagcctaccg agtccatcgt gcgctttccc aatatcacaa acctgtgccc ttttggcgag 1020
gtgttcaacg caacccgctt cgcaagcgtg tacgcctgga ataggaagcg catctccaac 1080
tgcgtggccg actattctgt gctgtacaac agcgcctcct tctctacctt taagtgctat 1140
ggcgtgagcc ccacaaagct gaatgacctg tgctttacca acgtgtacgc cgattccttc 1200
gtgatcaggg gcgacgaggt gcgccagatc gcaccaggac agacaggcaa gatcgcagac 1260
tacaattata agctgcctga cgatttcacc ggctgcgtga tcgcctggaa ctctaacaat 1320
ctggatagca aagtgggcgg caactacaat tatctgtacc ggctgtttag aaagtctaat 1380
ctgaagccat tcgagaggga catctccaca gaaatctacc aggccggctc taccccctgc 1440
aatggcgtgg agggctttaa ctgttatttc cctctgcaga gctacggctt ccagccaaca 1500
aacggcgtgg gctatcagcc ctaccgcgtg gtggtgctgt cttttgagct gctgcacgca 1560
cctgcaacag tgtgcggacc aaagaagagc accaatctgg tgaagaacaa gtgcgtgaac 1620
ttcaacttca acggactgac cggcacaggc gtgctgaccg agtccaacaa gaagttcctg 1680
ccttttcagc agttcggcag ggacatcgca gataccacag acgccgtgcg cgaccctcag 1740
accctggaga tcctggatat cacaccatgc tccttcggcg gcgtgtctgt gatcacacca 1800
ggcaccaata caagcaacca ggtggccgtg ctgtatcagg acgtgaattg taccgaggtg 1860
cccgtggcaa tccacgcaga tcagctgacc cctacatggc gggtgtactc taccggcagc 1920
aacgtgttcc agacaagagc aggatgcctg atcggagcag agcacgtgaa caatagctat 1980
gagtgcgaca tccctatcgg cgccggcatc tgtgcctcct accagaccca gacaaactcc 2040
ccaaggagag cacggtctgt ggcaagccag tccatcatcg cctataccat gagcctgggc 2100
gccgagaatt ccgtggccta ctccaacaat tctatcgcca tccctaccaa cttcacaatc 2160
tccgtgacca cagagatcct gccagtgagc atgaccaaga catccgtgga ctgcacaatg 2220
tatatctgtg gcgattccac cgagtgctct aacctgctgc tgcagtacgg ctctttttgt 2280
acccagctga atagagccct gacaggcatc gccgtggagc aggacaagaa cacacaggag 2340
gtgttcgccc aggtgaagca aatctacaag accccaccca tcaaggactt tggcggcttc 2400
aacttcagcc agatcctgcc cgatcctagc aagccatcca agcggtcttt tatcgaggac 2460
ctgctgttca acaaggtgac cctggccgat gccggcttca tcaagcagta tggcgattgc 2520
ctgggcgaca tcgccgccag agacctgatc tgtgcccaga agtttaatgg cctgaccgtg 2580
ctgcctccac tgctgacaga tgagatgatc gcccagtaca catctgccct gctggcaggc 2640
accatcacaa gcggatggac cttcggcgca ggagccgccc tgcagatccc ctttgccatg 2700
cagatggcct atcggttcaa cggcatcggc gtgacccaga atgtgctgta cgagaaccag 2760
aagctgatcg ccaatcagtt taactccgcc atcggcaaga tccaggactc tctgagctcc 2820
acagcaagcg ccctgggcaa gctgcaggat gtggtgaatc agaacgccca ggccctgaat 2880
accctggtga agcagctgtc tagcaacttc ggcgccatct cctctgtgct gaatgatatc 2940
ctgagcaggc tggacaaggt ggaggcagag gtgcagatcg accggctgat cacaggcaga 3000
ctgcagtccc tgcagaccta cgtgacacag cagctgatca gggcagcaga gatcagggca 3060
tctgccaatc tggccgccac caagatgagc gagtgcgtgc tgggccagtc caagagagtg 3120
gacttttgtg gcaagggcta tcacctgatg agcttcccac agtccgcccc tcacggagtg 3180
gtgtttctgc acgtgaccta cgtgccagcc caggagaaga acttcaccac agcaccagca 3240
atctgccacg atggcaaggc acactttcct agggagggcg tgttcgtgag caacggcacc 3300
cactggtttg tgacacagcg caatttctac gagccacaga tcatcaccac agacaataca 3360
ttcgtgtccg gcaactgtga cgtggtcatc ggcatcgtga acaataccgt gtatgatcct 3420
ctgcagccag agctggactc ttttaaggag gagctggata agtacttcaa gaatcacacc 3480
agccccgacg tggatctggg cgacatctct ggcatcaatg ccagcgtggt gaacatccag 3540
aaggagatcg acaggctgaa cgaggtggcc aagaatctga acgagtccct gatcgatctg 3600
caggagctgg gcaagtatga gcagtacatc aagtggccct ggtatatctg gctgggcttc 3660
atcgccggcc tgatcgccat cgtgatggtg accatcatgc tgtgctgtat gacaagctgc 3720
tgttcctgcc tgaagggctg ctgttcttgt ggcagctgct gtaagtttga tgaggacgat 3780
agcgagcctg tgctgaaggg cgtgaagctg cactacacca ccggtctgca gctagctcga 3840
gtc 3843
<210> 13
<211> 805
<212> PRT
<213> Artificial sequence
<400> 13
Met Ser Ser Ser Ser Trp Leu Leu Leu Ser Leu Val Ala Val Thr Ala
1 5 10 15
Ala Gln Ser Thr Ile Glu Glu Gln Ala Lys Thr Phe Leu Asp Lys Phe
20 25 30
Asn His Glu Ala Glu Asp Leu Phe Tyr Gln Ser Ser Leu Ala Ser Trp
35 40 45
Asn Tyr Asn Thr Asn Ile Thr Glu Glu Asn Val Gln Asn Met Asn Asn
50 55 60
Ala Gly Asp Lys Trp Ser Ala Phe Leu Lys Glu Gln Ser Thr Leu Ala
65 70 75 80
Gln Met Tyr Pro Leu Gln Glu Ile Gln Asn Leu Thr Val Lys Leu Gln
85 90 95
Leu Gln Ala Leu Gln Gln Asn Gly Ser Ser Val Leu Ser Glu Asp Lys
100 105 110
Ser Lys Arg Leu Asn Thr Ile Leu Asn Thr Met Ser Thr Ile Tyr Ser
115 120 125
Thr Gly Lys Val Cys Asn Pro Asp Asn Pro Gln Glu Cys Leu Leu Leu
130 135 140
Glu Pro Gly Leu Asn Glu Ile Met Ala Asn Ser Leu Asp Tyr Asn Glu
145 150 155 160
Arg Leu Trp Ala Trp Glu Ser Trp Arg Ser Glu Val Gly Lys Gln Leu
165 170 175
Arg Pro Leu Tyr Glu Glu Tyr Val Val Leu Lys Asn Glu Met Ala Arg
180 185 190
Ala Asn His Tyr Glu Asp Tyr Gly Asp Tyr Trp Arg Gly Asp Tyr Glu
195 200 205
Val Asn Gly Val Asp Gly Tyr Asp Tyr Ser Arg Gly Gln Leu Ile Glu
210 215 220
Asp Val Glu His Thr Phe Glu Glu Ile Lys Pro Leu Tyr Glu His Leu
225 230 235 240
His Ala Tyr Val Arg Ala Lys Leu Met Asn Ala Tyr Pro Ser Tyr Ile
245 250 255
Ser Pro Ile Gly Cys Leu Pro Ala His Leu Leu Gly Asp Met Trp Gly
260 265 270
Arg Phe Trp Thr Asn Leu Tyr Ser Leu Thr Val Pro Phe Gly Gln Lys
275 280 285
Pro Asn Ile Asp Val Thr Asp Ala Met Val Asp Gln Ala Trp Asp Ala
290 295 300
Gln Arg Ile Phe Lys Glu Ala Glu Lys Phe Phe Val Ser Val Gly Leu
305 310 315 320
Pro Asn Met Thr Gln Gly Phe Trp Glu Asn Ser Met Leu Thr Asp Pro
325 330 335
Gly Asn Val Gln Lys Ala Val Cys His Pro Thr Ala Trp Asp Leu Gly
340 345 350
Lys Gly Asp Phe Arg Ile Leu Met Cys Thr Lys Val Thr Met Asp Asp
355 360 365
Phe Leu Thr Ala His His Glu Met Gly His Ile Gln Tyr Asp Met Ala
370 375 380
Tyr Ala Ala Gln Pro Phe Leu Leu Arg Asn Gly Ala Asn Glu Gly Phe
385 390 395 400
His Glu Ala Val Gly Glu Ile Met Ser Leu Ser Ala Ala Thr Pro Lys
405 410 415
His Leu Lys Ser Ile Gly Leu Leu Ser Pro Asp Phe Gln Glu Asp Asn
420 425 430
Glu Thr Glu Ile Asn Phe Leu Leu Lys Gln Ala Leu Thr Ile Val Gly
435 440 445
Thr Leu Pro Phe Thr Tyr Met Leu Glu Lys Trp Arg Trp Met Val Phe
450 455 460
Lys Gly Glu Ile Pro Lys Asp Gln Trp Met Lys Lys Trp Trp Glu Met
465 470 475 480
Lys Arg Glu Ile Val Gly Val Val Glu Pro Val Pro His Asp Glu Thr
485 490 495
Tyr Cys Asp Pro Ala Ser Leu Phe His Val Ser Asn Asp Tyr Ser Phe
500 505 510
Ile Arg Tyr Tyr Thr Arg Thr Leu Tyr Gln Phe Gln Phe Gln Glu Ala
515 520 525
Leu Cys Gln Ala Ala Lys His Glu Gly Pro Leu His Lys Cys Asp Ile
530 535 540
Ser Asn Ser Thr Glu Ala Gly Gln Lys Leu Phe Asn Met Leu Arg Leu
545 550 555 560
Gly Lys Ser Glu Pro Trp Thr Leu Ala Leu Glu Asn Val Val Gly Ala
565 570 575
Lys Asn Met Asn Val Arg Pro Leu Leu Asn Tyr Phe Glu Pro Leu Phe
580 585 590
Thr Trp Leu Lys Asp Gln Asn Lys Asn Ser Phe Val Gly Trp Ser Thr
595 600 605
Asp Trp Ser Pro Tyr Ala Asp Gln Ser Ile Lys Val Arg Ile Ser Leu
610 615 620
Lys Ser Ala Leu Gly Asp Lys Ala Tyr Glu Trp Asn Asp Asn Glu Met
625 630 635 640
Tyr Leu Phe Arg Ser Ser Val Ala Tyr Ala Met Arg Gln Tyr Phe Leu
645 650 655
Lys Val Lys Asn Gln Met Ile Leu Phe Gly Glu Glu Asp Val Arg Val
660 665 670
Ala Asn Leu Lys Pro Arg Ile Ser Phe Asn Phe Phe Val Thr Ala Pro
675 680 685
Lys Asn Val Ser Asp Ile Ile Pro Arg Thr Glu Val Glu Lys Ala Ile
690 695 700
Arg Met Ser Arg Ser Arg Ile Asn Asp Ala Phe Arg Leu Asn Asp Asn
705 710 715 720
Ser Leu Glu Phe Leu Gly Ile Gln Pro Thr Leu Gly Pro Pro Asn Gln
725 730 735
Pro Pro Val Ser Ile Trp Leu Ile Val Phe Gly Val Val Met Gly Val
740 745 750
Ile Val Val Gly Ile Val Ile Leu Ile Phe Thr Gly Ile Arg Asp Arg
755 760 765
Lys Lys Lys Asn Lys Ala Arg Ser Gly Glu Asn Pro Tyr Ala Ser Ile
770 775 780
Asp Ile Ser Lys Gly Glu Asn Asn Pro Gly Phe Gln Asn Thr Asp Asp
785 790 795 800
Val Gln Thr Ser Phe
805
<210> 14
<211> 2415
<212> DNA
<213> Artificial sequence
<400> 14
atgtcaagct cttcctggct ccttctcagc cttgttgctg taactgctgc tcagtccacc 60
attgaggaac aggccaagac atttttggac aagtttaacc acgaagccga agacctgttc 120
tatcaaagtt cacttgcttc ttggaattat aacaccaata ttactgaaga gaatgtccaa 180
aacatgaata atgctgggga caaatggtct gcctttttaa aggaacagtc cacacttgcc 240
caaatgtatc cactacaaga aattcagaat ctcacagtca agcttcagct gcaggctctt 300
cagcaaaatg ggtcttcagt gctctcagaa gacaagagca aacggttgaa cacaattcta 360
aatacaatga gcaccatcta cagtactgga aaagtttgta acccagataa tccacaagaa 420
tgcttattac ttgaaccagg tttgaatgaa ataatggcaa acagtttaga ctacaatgag 480
aggctctggg cttgggaaag ctggagatct gaggtcggca agcagctgag gccattatat 540
gaagagtatg tggtcttgaa aaatgagatg gcaagagcaa atcattatga ggactatggg 600
gattattgga gaggagacta tgaagtaaat ggggtagatg gctatgacta cagccgcggc 660
cagttgattg aagatgtgga acataccttt gaagagatta aaccattata tgaacatctt 720
catgcctatg tgagggcaaa gttgatgaat gcctatcctt cctatatcag tccaattgga 780
tgcctccctg ctcatttgct tggtgatatg tggggtagat tttggacaaa tctgtactct 840
ttgacagttc cctttggaca gaaaccaaac atagatgtta ctgatgcaat ggtggaccag 900
gcctgggatg cacagagaat attcaaggag gccgagaagt tctttgtatc tgttggtctt 960
cctaatatga ctcaaggatt ctgggaaaat tccatgctaa cggacccagg aaatgttcag 1020
aaagcagtct gccatcccac agcttgggac ctggggaagg gcgacttcag gatccttatg 1080
tgcacaaagg tgacaatgga cgacttcctg acagctcatc atgagatggg gcatatccag 1140
tatgatatgg catatgctgc acaacctttt ctgctaagaa atggagctaa tgaaggattc 1200
catgaagctg ttggggaaat catgtcactt tctgcagcca cacctaagca tttaaaatcc 1260
attggtcttc tgtcacccga ttttcaagaa gacaatgaaa cagaaataaa cttcctgctc 1320
aaacaagcac tcacgattgt tgggactctg ccatttactt acatgttaga gaagtggagg 1380
tggatggtct ttaaagggga aattcccaaa gaccagtgga tgaaaaagtg gtgggagatg 1440
aagcgagaga tagttggggt ggtggaacct gtgccccatg atgaaacata ctgtgacccc 1500
gcatctctgt tccatgtttc taatgattac tcattcattc gatattacac aaggaccctt 1560
taccaattcc agtttcaaga agcactttgt caagcagcta aacatgaagg ccctctgcac 1620
aaatgtgaca tctcaaactc tacagaagct ggacagaaac tgttcaatat gctgaggctt 1680
ggaaaatcag aaccctggac cctagcattg gaaaatgttg taggagcaaa gaacatgaat 1740
gtaaggccac tgctcaacta ctttgagccc ttatttacct ggctgaaaga ccagaacaag 1800
aattcttttg tgggatggag taccgactgg agtccatatg cagaccaaag catcaaagtg 1860
aggataagcc taaaatcagc tcttggagat aaagcatatg aatggaacga caatgaaatg 1920
tacctgttcc gatcatctgt tgcatatgct atgaggcagt actttttaaa agtaaaaaat 1980
cagatgattc tttttgggga ggaggatgtg cgagtggcta atttgaaacc aagaatctcc 2040
tttaatttct ttgtcactgc acctaaaaat gtgtctgata tcattcctag aactgaagtt 2100
gaaaaggcca tcaggatgtc ccggagccgt atcaatgatg ctttccgtct gaatgacaac 2160
agcctagagt ttctggggat acagccaaca cttggacctc ctaaccagcc ccctgtttcc 2220
atatggctga ttgtttttgg agttgtgatg ggagtgatag tggttggcat tgtcatcctg 2280
atcttcactg ggatcagaga tcggaagaag aaaaataaag caagaagtgg agaaaatcct 2340
tatgcctcca tcgatattag caaaggagaa aataatccag gattccaaaa cactgatgat 2400
gttcagacct ccttt 2415
<210> 15
<211> 602
<212> PRT
<213> Artificial sequence
<400> 15
Met Lys Thr Ile Ile Ala Leu Ser Tyr Ile Phe Cys Leu Val Phe Ala
1 5 10 15
Asp Tyr Lys Asp Asp Asp Asp Lys Ser Leu Gln Val Cys Pro Thr Glu
20 25 30
Ser Ile Val Arg Phe Pro Asn Ile Thr Asn Leu Cys Pro Phe Gly Glu
35 40 45
Val Phe Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala Trp Asn Arg Lys
50 55 60
Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser Val Leu Tyr Asn Ser Ala
65 70 75 80
Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro Thr Lys Leu Asn
85 90 95
Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe Val Ile Arg Gly
100 105 110
Asp Glu Val Arg Gln Ile Ala Pro Gly Gln Thr Gly Lys Ile Ala Asp
115 120 125
Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys Val Ile Ala Trp
130 135 140
Asn Ser Asn Asn Leu Asp Ser Lys Val Gly Gly Asn Tyr Asn Tyr Leu
145 150 155 160
Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe Glu Arg Asp Ile
165 170 175
Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys Asn Gly Val Glu
180 185 190
Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly Phe Gln Pro Thr
195 200 205
Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val Val Val Leu Ser Phe Glu
210 215 220
Leu Leu His Ala Pro Ala Thr Val Cys Gly Pro Lys Lys Ser Thr Asn
225 230 235 240
Leu Val Lys Asn Lys Cys Val Asn Phe Asn Phe Asn Gly Leu Thr Gly
245 250 255
Thr Gly Val Leu Thr Glu Ser Asn Lys Lys Phe Leu Pro Phe Gln Gln
260 265 270
Phe Gly Arg Asp Ile Ala Asp Thr Thr Asp Ala Val Arg Asp Pro Gln
275 280 285
Thr Leu Glu Ile Leu Asp Ile Thr Pro Cys Ser Gly Gly Gly Gly Ser
290 295 300
Gly Leu Phe Gly Ala Ile Ala Gly Phe Ile Glu Asn Gly Trp Glu Gly
305 310 315 320
Leu Ile Asp Gly Trp Tyr Gly Phe Arg His Gln Asn Ala Gln Gly Glu
325 330 335
Gly Thr Ala Ala Asp Tyr Lys Ser Thr Gln Ser Ala Ile Asp Gln Ile
340 345 350
Thr Gly Lys Leu Asn Arg Leu Ile Glu Lys Thr Asn Gln Gln Phe Glu
355 360 365
Leu Ile Asp Asn Glu Phe Asn Glu Val Glu Lys Gln Ile Gly Asn Val
370 375 380
Ile Asn Trp Thr Arg Asp Ser Ile Thr Glu Val Trp Ser Tyr Asn Ala
385 390 395 400
Glu Leu Leu Val Ala Met Glu Asn Gln His Thr Ile Asp Leu Ala Asp
405 410 415
Ser Glu Met Asp Lys Leu Tyr Glu Arg Val Lys Arg Gln Leu Arg Glu
420 425 430
Asn Ala Glu Glu Asp Gly Thr Gly Cys Phe Glu Ile Phe His Lys Cys
435 440 445
Asp Asp Asp Cys Met Ala Ser Ile Arg Asn Asn Thr Tyr Asp His Ser
450 455 460
Lys Tyr Arg Glu Glu Ala Met Gln Asn Arg Ile Gln Ile Asp Pro Val
465 470 475 480
Lys Leu Ser Ser Gly Tyr Lys Asp Val Ala Ser Thr Thr Thr Pro Ala
485 490 495
Pro Arg Pro Pro Thr Pro Ala Pro Thr Ile Ala Ser Gln Pro Leu Ser
500 505 510
Leu Arg Pro Glu Ala Cys Arg Pro Ala Ala Gly Gly Ala Val His Thr
515 520 525
Arg Gly Leu Asp Phe Ala Cys Asp Ile Tyr Ile Trp Ala Pro Leu Ala
530 535 540
Gly Thr Cys Gly Val Leu Leu Leu Ser Leu Val Ile Thr Leu Tyr Cys
545 550 555 560
Lys Arg Gly Arg Lys Lys Leu Leu Tyr Ile Phe Lys Gln Pro Phe Met
565 570 575
Arg Pro Val Gln Thr Thr Gln Glu Glu Asp Gly Cys Ser Cys Arg Phe
580 585 590
Pro Glu Glu Glu Glu Gly Gly Cys Glu Leu
595 600
<210> 16
<211> 1806
<212> DNA
<213> Artificial sequence
<400> 16
atgaaaacaa ttattgccct gtcttacatt ttctgcctcg tgttcgccga ctacaaggac 60
gacgacgaca agtccctgca ggtgtgccct accgagtcta tcgtgcgctt cccaaacatc 120
acaaacctgt gccctttcgg cgaggtgttc aacgccaccc ggttcgcctc tgtgtacgcc 180
tggaaccgga agcggatttc taactgcgtg gccgactact ccgtgctgta caactctgcc 240
tctttctcca cattcaagtg ctacggcgtg tcccctacca agctgaacga cctgtgcttc 300
accaacgtgt acgccgactc tttcgtgatt aggggcgacg aggtgagaca gattgcccct 360
ggccagacag gcaagatcgc cgactacaac tacaagctcc ctgacgactt cacaggctgc 420
gtgattgcct ggaactctaa caacctggac tctaaggtgg gcggcaacta caactacctg 480
tacagactgt tccggaagtc taacctcaag ccattcgagc gcgacattag caccgagatt 540
taccaggccg gcagcacccc atgcaacggc gtggagggct tcaactgcta cttcccactt 600
caatcttacg gcttccagcc aacaaacggc gtgggctacc agccataccg ggtggtggtg 660
ctgtccttcg agctactcca cgccccagcc acagtgtgcg gcccaaagaa gagcaccaac 720
ctcgtgaaga acaagtgcgt gaacttcaac ttcaacggcc tgacaggcac aggcgtgctc 780
accgagtcta acaagaagtt cctccctttc cagcagttcg gcagggacat cgccgacacc 840
accgacgccg tgcgcgaccc tcagacactc gaaattctgg acatcacccc ttgctctggc 900
ggcggcggct ctggcctgtt cggcgccatt gccggcttca tcgagaacgg ctgggagggc 960
ctcattgacg gctggtacgg cttcaggcac cagaacgccc agggcgaggg cacagccgcc 1020
gactacaagt ccacccagtc cgccattgac cagatcacag gcaagctgaa cagactcatt 1080
gaaaaaacaa accagcagtt cgagctgatt gacaacgagt tcaacgaggt ggagaagcag 1140
attggcaacg tgattaactg gacacgggac tctattacag aggtgtggtc ttacaacgcc 1200
gagttactcg tggcaatgga gaaccagcac acaattgacc tcgccgactc tgagatggac 1260
aagctgtacg agcgggtgaa gagacagctc agagagaacg ccgaggagga cggcacaggc 1320
tgcttcgaga tattccacaa gtgcgacgac gactgcatgg ccagcatccg gaacaacaca 1380
tacgaccact ctaagtaccg ggaggaggcc atgcagaacc gcatccagat tgacccagtg 1440
aagctgtcta gcggctacaa ggacgtggct agcaccacca cacccgcccc tagacctcct 1500
acccccgccc caacaattgc ctctcagcca ctgtctctca gacctgaggc gtgcaggccc 1560
gccgccggcg gcgccgtgca cacacggggc ctcgacttcg cctgcgacat ttacatttgg 1620
gccccactcg ccggcacatg cggcgtgctc ctcctgtctc tggtgatcac actgtactgc 1680
aagcggggca gaaagaagct cctgtacatt ttcaagcagc ctttcatgag acccgtgcag 1740
accacccagg aggaggacgg ctgctcttgc aggttccctg aggaggagga gggcggctgc 1800
gaacta 1806
<210> 17
<211> 111
<212> PRT
<213> Artificial sequence
<400> 17
Thr Thr Thr Pro Ala Pro Arg Pro Pro Thr Pro Ala Pro Thr Ile Ala
1 5 10 15
Ser Gln Pro Leu Ser Leu Arg Pro Glu Ala Cys Arg Pro Ala Ala Gly
20 25 30
Gly Ala Val His Thr Arg Gly Leu Asp Phe Ala Cys Asp Ile Tyr Ile
35 40 45
Trp Ala Pro Leu Ala Gly Thr Cys Gly Val Leu Leu Leu Ser Leu Val
50 55 60
Ile Thr Leu Tyr Cys Lys Arg Gly Arg Lys Lys Leu Leu Tyr Ile Phe
65 70 75 80
Lys Gln Pro Phe Met Arg Pro Val Gln Thr Thr Gln Glu Glu Asp Gly
85 90 95
Cys Ser Cys Arg Phe Pro Glu Glu Glu Glu Gly Gly Cys Glu Leu
100 105 110
<210> 18
<211> 333
<212> DNA
<213> Artificial sequence
<400> 18
accaccacac ccgcccctag acctcctacc cccgccccaa caattgcctc tcagccactg 60
tctctcagac ctgaggcgtg caggcccgcc gccggcggcg ccgtgcacac acggggcctc 120
gacttcgcct gcgacattta catttgggcc ccactcgccg gcacatgcgg cgtgctcctc 180
ctgtctctgg tgatcacact gtactgcaag cggggcagaa agaagctcct gtacattttc 240
aagcagcctt tcatgagacc cgtgcagacc acccaggagg aggacggctg ctcttgcagg 300
ttccctgagg aggaggaggg cggctgcgaa cta 333
<210> 19
<211> 15
<212> PRT
<213> Artificial sequence
<400> 19
Met Pro Thr Glu Ser Ile Val Arg Phe Pro Asn Ile Thr Asn Leu
1 5 10 15
<210> 20
<211> 15
<212> PRT
<213> Artificial sequence
<400> 20
Ala Val Arg Asp Pro Gln Thr Leu Glu Ile Leu Asp Ile Thr Pro
1 5 10 15
<210> 21
<211> 227
<212> PRT
<213> Artificial sequence
<400> 21
Arg Val Gln Pro Thr Glu Ser Ile Val Arg Phe Pro Asn Ile Thr Asn
1 5 10 15
Leu Cys Pro Phe Gly Glu Val Phe Asn Ala Thr Arg Phe Ala Ser Val
20 25 30
Tyr Ala Trp Asn Arg Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser
35 40 45
Val Leu Tyr Asn Ser Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val
50 55 60
Ser Pro Thr Lys Leu Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp
65 70 75 80
Ser Phe Val Ile Arg Gly Asp Glu Val Arg Gln Ile Ala Pro Gly Gln
85 90 95
Thr Gly Lys Ile Ala Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr
100 105 110
Gly Cys Val Ile Ala Trp Asn Ser Asn Asn Leu Asp Ser Lys Val Gly
115 120 125
Gly Asn Tyr Asn Tyr Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys
130 135 140
Pro Phe Glu Arg Asp Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr
145 150 155 160
Pro Cys Asn Gly Val Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser
165 170 175
Tyr Gly Phe Gln Pro Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val
180 185 190
Val Val Leu Ser Phe Glu Leu Leu His Ala Pro Ala Thr Val Cys Gly
195 200 205
Pro Lys Lys Ser Thr Asn Leu Val Lys Asn Lys Cys Val Asn Phe Asn
210 215 220
Phe Asn Gly
225
<210> 22
<211> 681
<212> DNA
<213> Artificial sequence
<400> 22
cgtgtccaac ctaccgagtc catcgtgcgc ttccctaaca tcaccaacct gtgccctttc 60
ggtgaagtgt tcaacgctac ccgcttcgct tctgtgtacg cttggaaccg caagcgcatc 120
tccaactgcg tggctgacta ctctgtgctc tacaactccg cctccttctc caccttcaag 180
tgttacggcg tgtcccctac caagttgaac gatctgtgct tcaccaacgt ctacgctgac 240
tccttcgtga tccgtggcga cgaggtccgc caaatcgctc ctggtcagac cggtaagatc 300
gccgactaca actacaagct gcctgacgac ttcaccggtt gcgtgatcgc ttggaactcc 360
aacaacctgg actccaaggt gggtggtaac tacaactacc tgtacaggct gttccgcaag 420
agcaacctca agcccttcga aagggacatc tccactgaga tctaccaggc tggctccaca 480
ccctgcaacg gtgtggaagg tttcaactgc tacttccccc tccagtccta cggtttccag 540
cccaccaacg gtgtgggata ccagccctac cgcgtggtgg tgctctcctt cgagctgctg 600
cacgcccctg ctaccgtctg cggccctaag aagtccacca acctggtcaa gaacaagtgc 660
gtgaacttca acttcaacgg t 681
<210> 23
<211> 640
<212> PRT
<213> Artificial sequence
<400> 23
Met Leu Leu Val Asn Gln Ser His Gln Gly Phe Asn Lys Glu His Thr
1 5 10 15
Ser Lys Met Val Ser Ala Thr Val Leu Tyr Val Leu Leu Ala Ala Ala
20 25 30
Ala His Ser Ala Phe Ala Thr Ser Tyr Tyr Glu Phe Arg Val Gln Pro
35 40 45
Thr Glu Ser Ile Val Arg Phe Pro Asn Ile Thr Asn Leu Cys Pro Phe
50 55 60
Gly Glu Val Phe Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala Trp Asn
65 70 75 80
Arg Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser Val Leu Tyr Asn
85 90 95
Ser Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro Thr Lys
100 105 110
Leu Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe Val Ile
115 120 125
Arg Gly Asp Glu Val Arg Gln Ile Ala Pro Gly Gln Thr Gly Lys Ile
130 135 140
Ala Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys Val Ile
145 150 155 160
Ala Trp Asn Ser Asn Asn Leu Asp Ser Lys Val Gly Gly Asn Tyr Asn
165 170 175
Tyr Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe Glu Arg
180 185 190
Asp Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys Asn Gly
195 200 205
Val Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly Phe Gln
210 215 220
Pro Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val Val Val Leu Ser
225 230 235 240
Phe Glu Leu Leu His Ala Pro Ala Thr Val Cys Gly Pro Lys Lys Ser
245 250 255
Thr Asn Leu Val Lys Asn Lys Cys Val Asn Phe Asn Phe Asn Gly Gly
260 265 270
Gly Gly Ser Gly Gly Gly Gly Ser Gly Leu Phe Gly Ala Ile Ala Gly
275 280 285
Phe Ile Glu Asn Gly Trp Glu Gly Leu Ile Asp Gly Trp Tyr Gly Phe
290 295 300
Arg His Gln Asn Ala Gln Gly Glu Gly Thr Ala Ala Asp Tyr Lys Ser
305 310 315 320
Thr Gln Ser Ala Ile Asp Gln Ile Thr Gly Lys Leu Asn Arg Leu Ile
325 330 335
Glu Lys Thr Asn Gln Gln Phe Glu Leu Ile Asp Asn Glu Phe Asn Glu
340 345 350
Val Glu Lys Gln Ile Gly Asn Val Ile Asn Trp Thr Arg Asp Ser Ile
355 360 365
Thr Glu Val Trp Ser Tyr Asn Ala Glu Leu Leu Val Ala Met Glu Asn
370 375 380
Gln His Thr Ile Asp Leu Ala Asp Ser Glu Met Asp Lys Leu Tyr Glu
385 390 395 400
Arg Val Lys Arg Gln Leu Arg Glu Asn Ala Glu Glu Asp Gly Thr Gly
405 410 415
Cys Phe Glu Ile Phe His Lys Cys Asp Asp Asp Cys Met Ala Ser Ile
420 425 430
Arg Asn Asn Thr Tyr Asp His Ser Lys Tyr Arg Glu Glu Ala Met Gln
435 440 445
Asn Arg Ile Gln Ile Asp Pro Val Lys Leu Ser Ser Gly Tyr Lys Asp
450 455 460
Val Val Asp Gly Gly Gly Gly Ser Thr Arg Ser Gly Tyr Cys Leu Asp
465 470 475 480
Leu Lys Thr Gln Val Gln Thr Pro Gln Gly Met Lys Glu Ile Ser Asn
485 490 495
Ile Gln Val Gly Asp Leu Val Leu Ser Asn Thr Gly Tyr Asn Glu Val
500 505 510
Leu Asn Val Phe Pro Lys Ser Lys Lys Lys Ser Tyr Lys Ile Thr Leu
515 520 525
Glu Asp Gly Lys Glu Ile Ile Cys Ser Glu Glu His Leu Phe Pro Thr
530 535 540
Gln Thr Gly Glu Met Asn Ile Ser Gly Gly Leu Lys Glu Gly Met Cys
545 550 555 560
Leu Tyr Val Lys Glu Gly Ser Ser Arg Gly Ser Leu Met Thr Ala Asp
565 570 575
Asn Lys Phe Asn Lys Glu Gln Gln Asn Ala Phe Tyr Glu Ile Leu His
580 585 590
Leu Pro Asn Leu Asn Glu Glu Asn Arg Asn Gly Phe Ile Gln Ser Leu
595 600 605
Lys Asp Asp Pro Ser Gln Ser Ala Asn Leu Leu Ala Glu Ala Lys Lys
610 615 620
Leu Asn Asp Gly Gly Gly Gly Ser Gly His His His His His His His
625 630 635 640
<210> 24
<211> 1920
<212> DNA
<213> Artificial sequence
<400> 24
atgctgctgg tgaaccagtc ccaccagggt ttcaacaagg agcacacctc caagatggtc 60
tccgccaccg tgttgtacgt gctgttggct gccgccgccc actccgcttt cgctacctcc 120
tactacgaat tccgtgtcca acctaccgag tccatcgtgc gcttccctaa catcaccaac 180
ctgtgccctt tcggtgaagt gttcaacgct acccgcttcg cttctgtgta cgcttggaac 240
cgcaagcgca tctccaactg cgtggctgac tactctgtgc tctacaactc cgcctccttc 300
tccaccttca agtgttacgg cgtgtcccct accaagttga acgatctgtg cttcaccaac 360
gtctacgctg actccttcgt gatccgtggc gacgaggtcc gccaaatcgc tcctggtcag 420
accggtaaga tcgccgacta caactacaag ctgcctgacg acttcaccgg ttgcgtgatc 480
gcttggaact ccaacaacct ggactccaag gtgggtggta actacaacta cctgtacagg 540
ctgttccgca agagcaacct caagcccttc gaaagggaca tctccactga gatctaccag 600
gctggctcca caccctgcaa cggtgtggaa ggtttcaact gctacttccc cctccagtcc 660
tacggtttcc agcccaccaa cggtgtggga taccagccct accgcgtggt ggtgctctcc 720
ttcgagctgc tgcacgcccc tgctaccgtc tgcggcccta agaagtccac caacctggtc 780
aagaacaagt gcgtgaactt caacttcaac ggtggtggtg gtagtggtgg tggtggttct 840
ggtttgttcg gtgctatcgc tggtttcatc gagaacggtt gggagggtct gatcgatggt 900
tggtacggtt tccgtcacca gaacgctcag ggtgagggta ctgctgctga ttacaagtct 960
acccagagtg ctatcgacca gatcaccgga aagttgaaca ggctgatcga gaagactaac 1020
cagcagttcg aactcatcga caacgagttc aacgaggtgg agaagcagat cggcaacgtg 1080
atcaactgga ccagggacag catcacagag gtgtggtcct acaacgccga gctgctggtg 1140
gctatggaaa accagcacac catcgacctg gctgatagcg agatggacaa gctgtacgag 1200
cgcgtgaaga ggcagttgcg tgagaacgct gaggaggacg gaaccggttg cttcgaaatc 1260
ttccacaagt gcgacgacga ctgcatggct tccatcagga acaacacata cgaccactcc 1320
aagtacaggg aggaggctat gcagaacagg attcaaatcg accctgtgaa gctgtcatcc 1380
ggttacaagg acgtggtcga cggtggtggt ggttctactc gttctggtta ctgcttggat 1440
ttgaagactc aggtgcagac tcctcagggt atgaaggaga tctctaacat ccaggtgggt 1500
gacttggtgc tgagcaacac tggttacaac gaggtgctga acgtgttccc taagtccaag 1560
aagaagagct acaagatcac tctcgaggac ggcaaggaga tcatctgctc cgaggagcac 1620
ctgttcccca cacaaaccgg cgagatgaac atctccggcg gcctgaagga aggtatgtgc 1680
ctgtacgtga aggagggcag ctctagaggt tctttgatga ctgctgataa caagttcaac 1740
aaggagcagc agaacgcttt ctacgagatc ttgcacttgc ctaacttgaa cgaggagaac 1800
aggaacggtt tcatccagtc tctgaaggac gaccctagcc agtccgccaa cctgctggct 1860
gaggctaaga agctgaacga cggtggtggt ggttccggtc accaccacca ccaccaccac 1920
<210> 25
<211> 654
<212> PRT
<213> Artificial sequence
<400> 25
Met Leu Leu Val Asn Gln Ser His Gln Gly Phe Asn Lys Glu His Thr
1 5 10 15
Ser Lys Met Val Ser Ala Thr Val Leu Tyr Val Leu Leu Ala Ala Ala
20 25 30
Ala His Ser Ala Phe Ala Thr Ser Tyr Tyr Glu Phe Arg Val Gln Pro
35 40 45
Thr Glu Ser Ile Val Arg Phe Pro Asn Ile Thr Asn Leu Cys Pro Phe
50 55 60
Gly Glu Val Phe Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala Trp Asn
65 70 75 80
Arg Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser Val Leu Tyr Asn
85 90 95
Ser Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro Thr Lys
100 105 110
Leu Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe Val Ile
115 120 125
Arg Gly Asp Glu Val Arg Gln Ile Ala Pro Gly Gln Thr Gly Lys Ile
130 135 140
Ala Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys Val Ile
145 150 155 160
Ala Trp Asn Ser Asn Asn Leu Asp Ser Lys Val Gly Gly Asn Tyr Asn
165 170 175
Tyr Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe Glu Arg
180 185 190
Asp Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys Asn Gly
195 200 205
Val Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly Phe Gln
210 215 220
Pro Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val Val Val Leu Ser
225 230 235 240
Phe Glu Leu Leu His Ala Pro Ala Thr Val Cys Gly Pro Lys Lys Ser
245 250 255
Thr Asn Leu Val Lys Asn Lys Cys Val Asn Phe Asn Phe Asn Gly Gly
260 265 270
Gly Gly Ser Gly Gly Gly Gly Ser Gly Leu Phe Gly Ala Ile Ala Gly
275 280 285
Phe Ile Glu Asn Gly Trp Glu Gly Leu Ile Asp Gly Trp Tyr Gly Phe
290 295 300
Arg His Gln Asn Ala Gln Gly Glu Gly Thr Ala Ala Asp Tyr Lys Ser
305 310 315 320
Thr Gln Ser Ala Ile Asp Gln Ile Thr Gly Lys Leu Asn Arg Leu Ile
325 330 335
Glu Lys Thr Asn Gln Gln Phe Glu Leu Ile Asp Asn Glu Phe Asn Glu
340 345 350
Val Glu Lys Gln Ile Gly Asn Val Ile Asn Trp Thr Arg Asp Ser Ile
355 360 365
Thr Glu Val Trp Ser Tyr Asn Ala Glu Leu Leu Val Ala Met Glu Asn
370 375 380
Gln His Thr Ile Asp Leu Ala Asp Ser Glu Met Asp Lys Leu Tyr Glu
385 390 395 400
Arg Val Lys Arg Gln Leu Arg Glu Asn Ala Glu Glu Asp Gly Thr Gly
405 410 415
Cys Phe Glu Ile Phe His Lys Cys Asp Asp Asp Cys Met Ala Ser Ile
420 425 430
Arg Asn Asn Thr Tyr Asp His Ser Lys Tyr Arg Glu Glu Ala Met Gln
435 440 445
Asn Arg Ile Gln Ile Asp Pro Val Lys Leu Ser Ser Gly Tyr Lys Asp
450 455 460
Val Val Asp Gly Gly Gly Gly Ser Thr Thr Ala Ser Thr Ser Gln Val
465 470 475 480
Arg Gln Asn Tyr His Gln Asp Ser Glu Ala Ala Ile Asn Arg Gln Ile
485 490 495
Asn Leu Glu Leu Tyr Ala Ser Tyr Val Tyr Leu Ser Met Ser Tyr Tyr
500 505 510
Phe Asp Arg Asp Asp Val Ala Leu Lys Asn Phe Ala Lys Tyr Phe Leu
515 520 525
His Gln Ser His Glu Glu Arg Glu His Ala Glu Lys Leu Met Lys Leu
530 535 540
Gln Asn Gln Arg Gly Gly Arg Ile Phe Leu Gln Asp Ile Lys Lys Pro
545 550 555 560
Asp Cys Asp Asp Trp Glu Ser Gly Leu Asn Ala Met Glu Cys Ala Leu
565 570 575
His Leu Glu Lys Asn Val Asn Gln Ser Leu Leu Glu Leu His Lys Leu
580 585 590
Ala Thr Asp Lys Asn Asp Pro His Leu Cys Asp Phe Ile Glu Thr His
595 600 605
Tyr Leu Asn Glu Gln Val Lys Ala Ile Lys Glu Leu Gly Asp His Val
610 615 620
Thr Asn Leu Arg Lys Met Gly Ala Pro Glu Ser Gly Leu Ala Glu Tyr
625 630 635 640
Leu Phe Asp Lys His Thr Leu Gly Asp Ser Asp Asn Glu Ser
645 650
<210> 26
<211> 1962
<212> DNA
<213> Artificial sequence
<400> 26
atgctgctgg tgaaccagtc ccaccagggt ttcaacaagg agcacacctc caagatggtc 60
tccgccaccg tgttgtacgt gctgttggct gccgccgccc actccgcttt cgctacctcc 120
tactacgaat tccgtgtcca acctaccgag tccatcgtgc gcttccctaa catcaccaac 180
ctgtgccctt tcggtgaagt gttcaacgct acccgcttcg cttctgtgta cgcttggaac 240
cgcaagcgca tctccaactg cgtggctgac tactctgtgc tctacaactc cgcctccttc 300
tccaccttca agtgttacgg cgtgtcccct accaagttga acgatctgtg cttcaccaac 360
gtctacgctg actccttcgt gatccgtggc gacgaggtcc gccaaatcgc tcctggtcag 420
accggtaaga tcgccgacta caactacaag ctgcctgacg acttcaccgg ttgcgtgatc 480
gcttggaact ccaacaacct ggactccaag gtgggtggta actacaacta cctgtacagg 540
ctgttccgca agagcaacct caagcccttc gaaagggaca tctccactga gatctaccag 600
gctggctcca caccctgcaa cggtgtggaa ggtttcaact gctacttccc cctccagtcc 660
tacggtttcc agcccaccaa cggtgtggga taccagccct accgcgtggt ggtgctctcc 720
ttcgagctgc tgcacgcccc tgctaccgtc tgcggcccta agaagtccac caacctggtc 780
aagaacaagt gcgtgaactt caacttcaac ggtggtggtg gtagtggtgg tggtggttct 840
ggtttgttcg gtgctatcgc tggtttcatc gagaacggtt gggagggtct gatcgatggt 900
tggtacggtt tccgtcacca gaacgctcag ggtgagggta ctgctgctga ttacaagtct 960
acccagagtg ctatcgacca gatcaccgga aagttgaaca ggctgatcga gaagactaac 1020
cagcagttcg aactcatcga caacgagttc aacgaggtgg agaagcagat cggcaacgtg 1080
atcaactgga ccagggacag catcacagag gtgtggtcct acaacgccga gctgctggtg 1140
gctatggaaa accagcacac catcgacctg gctgatagcg agatggacaa gctgtacgag 1200
cgcgtgaaga ggcagttgcg tgagaacgct gaggaggacg gaaccggttg cttcgaaatc 1260
ttccacaagt gcgacgacga ctgcatggct tccatcagga acaacacata cgaccactcc 1320
aagtacaggg aggaggctat gcagaacagg attcaaatcg accctgtgaa gctgtcatcc 1380
ggttacaagg acgtggtcga cggtggtggt ggttctacga ccgcgtccac ctcgcaggtg 1440
cgccagaact accaccagga ctcagaggcc gccatcaacc gccagatcaa cctggagctc 1500
tacgcctcct acgtttacct gtccatgtct tactactttg accgcgatga tgtggctttg 1560
aagaactttg ccaaatactt tcttcaccaa tctcatgagg agagggaaca tgctgagaaa 1620
ctgatgaagc tgcagaacca acgaggtggc cgaatcttcc ttcaggatat caagaaacca 1680
gactgtgatg actgggagag cgggctgaat gcaatggagt gtgcattaca tttggaaaaa 1740
aatgtgaatc agtcactact ggaactgcac aaactggcca ctgacaaaaa tgacccccat 1800
ttgtgtgact tcattgagac acattacctg aatgagcagg tgaaagccat caaagaattg 1860
ggtgaccacg tgaccaactt gcgcaagatg ggagcgcccg aatctggctt ggcggaatat 1920
ctctttgaca agcacaccct gggagacagt gataatgaaa gc 1962

Claims (10)

1. An immunogenic peptide comprising the following moieties:
(a) immunogen stem: it comprises the HA2 region of influenza virus hemagglutinin HA;
(b) immunogen crown: comprising a viral membrane protein or an immunogenic fragment thereof, wherein the viral membrane protein is derived from a virus other than the virus from which the HA2 region of (a) was derived;
(c) optionally, other moieties attached to the foregoing moieties.
2. The immunogenic peptide of claim 1, wherein,
the source of the HA2 region is selected from the group consisting of: any of H1 to H18, particularly from the widely prevalent human influenza H1, H2, H3 and the multiple-appearing human infections with avian influenza H5 and H7, for example from 2009 pandemic H1(H1N1), 2013 pandemic H7(H7N 9);
for example, the amino acid sequence of the HA2 region is shown in SEQ ID NO. 1, or is encoded by a nucleotide molecule having the sequence shown in SEQ ID NO. 2.
3. The immunogenic peptide of claim 1, wherein the immunogenic crown comprises: an influenza virus of a different origin from the HA2 region in (a); a membrane protein of a non-influenza virus or other influenza strain or an immunogenic fragment thereof;
the source of the immunogen crown is selected from the group consisting of: coronavirus, aids virus, influenza virus (e.g., the HA1 region of influenza virus hemagglutinin HA), rabies virus, classical swine fever virus, porcine reproductive and respiratory syndrome virus, measles virus, ebola virus, herpes virus, arbovirus (e.g., zika virus, epidemic encephalitis b virus, forest encephalitis virus, dengue virus, hantavirus, hemorrhagic fever virus);
for example, the source of the immunogen crown is selected from: coronavirus SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-229E, HCoV-OC43, HCoV-NL63, HCoV-HKU1, bat-CoV, e.g., an S1 protein comprising the receptor binding domain of a coronavirus, or a Receptor Binding Domain (RBD), or an engineered receptor binding domain (e.g., the RBD region is modified with a terminal cysteine to form an sRBD region), or an immunogenic fragment thereof;
for example, the immunogenic crown is selected from S1 of coronavirus SARS-CoV-2, particularly the RBD region from S1 (e.g., the sequence of which is shown in SEQ ID NO:3 or encoded by the nucleotide molecule of SEQ ID NO: 4), or a modified RBD region thereof (e.g., the region of sRBD modified with a terminal Cys, the peptide stretch shown in SEQ ID NO:5 or encoded by the nucleotide molecule of SEQ ID NO: 6), or an immunogenic fragment of the RBD region (e.g., the peptide stretch shown in SEQ ID NO:21 or encoded by the nucleotide molecule of SEQ ID NO: 22).
4. The immunogenic peptide of claim 1, wherein the additional moiety is selected from the group consisting of:
immune modulatory sequences, such as IL-2, IL-7, IL-12, IL-18, IL-21, GM-CSF, CD40L, CD40 stimulatory antibodies, PD-1 and PD-L1 antibodies, CTLA4 antibodies, chemokines CXCL9, CXCL10, CXCL11, CXCL12, CXCL3, XCL1, CCL4, CCL20, cholera toxin and subunits thereof, bacterial flagellin, FimH, HIV p24, HIV gp 41;
a moiety that enables the immunogenic peptide to form a nanoparticle, such as transferrin (Fn, e.g., a peptide molecule as shown in SEQ ID NO:7 or encoded by a nucleotide molecule as shown in SEQ ID NO: 8);
signal peptides such as CD33, CD8, CD16, mouse IgG1 antibody;
a transmembrane region enabling the immunogenic peptide to be expressed on the surface of a viral vector, such as the CD8 transmembrane region (CD8TM, e.g., a peptide molecule as set forth in SEQ ID NO:17 or encoded by a nucleotide molecule as set forth in SEQ ID NO: 18), the HA2 transmembrane region, the CD4 transmembrane region, the gp41 transmembrane region;
linker peptides, e.g. (G4S)3、(G4S)n、GSAGSAAGSGEF、(Gly)6、EFPKPSTPPGSSGGAP、KESGSVSSEQLAQFRSLD、(Gly)8、EGKSSGSGSESKST;
Tags such as His-tag, AviTag, Calmodulin tag, polyglutamate tag, E-tag, FLAG tag, HA-tag, Myc-tag, S-tag, SBP-tag, Sof-tag 1, Sof-tag3, Strep-tag, TC tag, V5 tag, T7 tag, VSV tag, Xpress tag, 3X FLAG tag, Isopep tag, Spytag, Snoop tag and PNE tag.
5. The immunogenic peptide of any one of claims 1-4, wherein the immunogenic peptide comprises: the RBD region or sRBD region or immunogenic fragment thereof linked to the HA2 region, and optionally other moieties linked to the foregoing;
for example, the immunogenic peptide comprises: an RBD region or sRGB region (e.g., a peptide molecule represented by SEQ ID NO:5 or encoded by a nucleotide molecule represented by SEQ ID NO: 6) and an Fn region (e.g., a Fn peptide molecule represented by SEQ ID NO:7 or encoded by a nucleotide molecule represented by SEQ ID NO: 8) linked to the HA2 region; an RBD region or sRBD region linked to the HA2 region, and a CD8 transmembrane region (e.g., the CD8 transmembrane region peptide molecule shown in SEQ ID NO:17 or encoded by the nucleotide molecule shown in SEQ ID NO: 18); an immunogenic fragment of an RBD region (e.g., a peptide molecule represented by SEQ ID NO:21 or encoded by a nucleotide molecule represented by SEQ ID NO: 22) linked to the HA2 region and an Fn region;
for example, the amino acid sequence of the immunogenic peptide is shown as SEQ ID NO 9 or 15 or 25, or the coding sequence of the immunogenic peptide is shown as SEQ ID NO 10 or 16 or 26.
6.A nucleotide molecule encoding the immunogenic peptide of any one of claims 1-5;
for example, the coding sequence of the HA2 region comprises the nucleotide sequence shown in SEQ ID NO. 2; the coding sequence of the immunogen crown comprises a nucleotide sequence shown as SEQ ID NO. 4 or a nucleotide sequence shown as SEQ ID NO. 6 or a nucleotide sequence shown as SEQ ID NO. 22; and/or the coding sequence of said further part comprises the nucleotide sequence shown in SEQ ID NO 8 or SEQ ID NO 18;
for example, the nucleotide molecule has a sequence shown in SEQ ID NO 10 or 16 or 26.
7. A vector comprising the nucleotide molecule of claim 6.
8. A host cell comprising the nucleotide molecule of claim 6 or the vector of claim 7 or capable of expressing the immunogenic peptide of any one of claims 1-5,
for example, the host cell is a mammalian cell or an insect cell, such as HEK293, HeLa, K562, CHO, NS0, SP2/0, PER.C6, Vero, RD, BHK, HT 1080, A549, Cos-7, ARPE-19 and MRC-5 cells; high Five, Sf9, Se301, SeIZD2109, SeUCR1, Sf9, Sf900+, Sf21, BTI-TN-5B1-4, MG-1, Tn368, HzAm1, BM-672302, Hz2E5 and Ao 38.
9. A vaccine capable of simultaneously inducing an immune response against an influenza virus and another non-influenza virus, comprising the immunogenic peptide of any one of claims 1-5, the nucleotide molecule of claim 6, the vector of claim 7, and/or the host cell of claim 8;
for example, the vaccine is a nucleic acid vaccine (DNA or RNA vaccine), a recombinant protein subunit vaccine, a recombinant viral vector vaccine, a recombinant bacterial vector vaccine, a virus-like particle vaccine, a nanoparticle vaccine, a cell vector vaccine;
for example, the non-influenza virus is selected from: coronavirus, aids virus, rabies virus, hog cholera virus, porcine reproductive and respiratory syndrome virus, measles virus, ebola virus, herpes virus, arbovirus (e.g., zika virus, epidemic encephalitis b virus, forest encephalitis virus, dengue virus, hantavirus, hemorrhagic fever virus);
for example, the vaccine is a viral vector vaccine, the viral vector selected from the group consisting of: poxviruses (e.g., Tiantan strain, North American vaccine strain, Huishi derived strain, Listeria strain, Ankara derived strain, Copenhagen strain, and New York strain), adenoviruses (Ad5, Ad11, Ad26, Ad35, AdC68), adeno-associated viruses, herpes simplex viruses, measles viruses, reoviruses, rhabdoviruses, forest encephalitis viruses, influenza viruses, respiratory syncytial viruses, poliovirus;
for example, the vaccine comprises or is used in combination with an adjuvant, for example selected from: aluminum adjuvant, cholera toxin and subunits thereof, oligodeoxynucleotide, manganese ion adjuvant, colloidal manganese adjuvant, Freund's adjuvant, MF59 adjuvant, QS-21 adjuvant, PolyI C and other TLR ligands, GM-CSF, IL-2, IL-3, IL-7, IL-11, IL-12, IL-18, IL-21;
for example, the vaccine is in a form suitable for intramuscular administration, intradermal administration, subcutaneous administration, nasal drops, aerosol inhalation, genital tract, rectal administration, oral administration, or a combination of the different modes of administration described above (e.g. intramuscular injection + nasal drops);
for example, the vaccine is in a form suitable for combined vaccination (e.g., combined or sequential vaccination) of 2 or more species, such as sequential vaccination before and after an S or S1 vaccine against a coronavirus (e.g., SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-229E, HCoV-OC43, HCoV-NL63, HCoV-HKU1, bat-CoV), or sequential vaccination before and after an HA or HA2 vaccine against an influenza virus (e.g., HA or HA2 from any of H1-H18).
10. Use of the immunogenic peptide according to any one of claims 1 to 5, the nucleotide molecule according to claim 6, the vector according to claim 7 and/or the host cell according to claim 8 for the preparation of a medicament for the simultaneous prevention or treatment of an influenza virus and another non-influenza virus,
for example, the non-influenza virus is selected from: coronavirus, aids virus, rabies virus, hog cholera virus, porcine reproductive and respiratory syndrome virus, measles virus, ebola virus, herpes virus, arbovirus (e.g., zika virus, epidemic encephalitis b virus, forest encephalitis virus, dengue virus, hantavirus, hemorrhagic fever virus).
CN202111597361.5A 2020-12-24 2021-12-24 Method for simultaneously inducing immune response against multiple viruses Pending CN114213548A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2020115520540 2020-12-24
CN2020139026 2020-12-24
CN202011552054 2020-12-24
CNPCT/CN2020/139026 2020-12-24

Publications (1)

Publication Number Publication Date
CN114213548A true CN114213548A (en) 2022-03-22

Family

ID=80705538

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111597361.5A Pending CN114213548A (en) 2020-12-24 2021-12-24 Method for simultaneously inducing immune response against multiple viruses

Country Status (2)

Country Link
CN (1) CN114213548A (en)
WO (1) WO2022135563A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116987719A (en) * 2023-06-21 2023-11-03 华南农业大学 H9N2AIV multi-epitope recombinant baculovirus, preparation method and application thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116162156A (en) * 2022-11-21 2023-05-26 中国科学院微生物研究所 Preparation and application of influenza and new crown bigeminal polyclonal antibody

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040096458A1 (en) * 2001-03-28 2004-05-20 Winfried Weissenhorn Fusion protein construct and method for inducing HIV-specific serum IgG and secretory IgA antibodies in-vivo
CN103842374A (en) * 2011-05-13 2014-06-04 诺华股份有限公司 Pre-fusion rsv f antigens
CN104250304A (en) * 2013-10-31 2014-12-31 普莱柯生物工程股份有限公司 Fusion protein, coding vaccine composition and application thereof
KR101525180B1 (en) * 2014-10-06 2015-06-04 주식회사 바이오리더스 Cell Surface Expression Vector for Influenza Virus Antigen and Microorganisms Transformed Thereby
CN107245105A (en) * 2017-06-29 2017-10-13 河南科技大学 HN VP233 221aa fusion proteins and its preparation method and application
CN107427571A (en) * 2014-12-31 2017-12-01 美利坚合众国,由健康及人类服务部部长代表 Novel multivalent vaccine based on nano particle
CN109851664A (en) * 2017-11-30 2019-06-07 清华大学 A kind of protein based on the reversed epitope design of antibody and its in the application prepared in AIDS virus resisting vaccine
CN111363045A (en) * 2020-02-18 2020-07-03 厦门大学 Preparation method of influenza and HIV chimeric protein and chimeric virus vaccine

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110272473B (en) * 2019-07-10 2021-03-02 军事科学院军事医学研究院军事兽医研究所 Influenza A universal virus-like particle and preparation method and application thereof
CN111358953A (en) * 2020-03-25 2020-07-03 上海市公共卫生临床中心 Vaccine vector for efficiently inducing humoral immune response of organism, preparation method and application thereof
CN111514287A (en) * 2020-04-29 2020-08-11 河南大学 Influenza A universal DNA vaccine and preparation method and application thereof
CN111560354B (en) * 2020-05-22 2022-07-19 中国人民解放军总医院第五医学中心 Recombinant novel coronavirus, preparation method and application thereof
CN112076315B (en) * 2020-08-25 2023-09-01 中国农业科学院生物技术研究所 Nanometer antigen particle fused by novel coronavirus S protein and ferritin subunit, novel coronavirus vaccine, preparation method and application thereof
CN113666990A (en) * 2021-08-24 2021-11-19 复旦大学 T cell vaccine immunogen for inducing broad-spectrum anti-coronavirus and application thereof

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040096458A1 (en) * 2001-03-28 2004-05-20 Winfried Weissenhorn Fusion protein construct and method for inducing HIV-specific serum IgG and secretory IgA antibodies in-vivo
CN103842374A (en) * 2011-05-13 2014-06-04 诺华股份有限公司 Pre-fusion rsv f antigens
CN104250304A (en) * 2013-10-31 2014-12-31 普莱柯生物工程股份有限公司 Fusion protein, coding vaccine composition and application thereof
KR101525180B1 (en) * 2014-10-06 2015-06-04 주식회사 바이오리더스 Cell Surface Expression Vector for Influenza Virus Antigen and Microorganisms Transformed Thereby
CN107427571A (en) * 2014-12-31 2017-12-01 美利坚合众国,由健康及人类服务部部长代表 Novel multivalent vaccine based on nano particle
CN107245105A (en) * 2017-06-29 2017-10-13 河南科技大学 HN VP233 221aa fusion proteins and its preparation method and application
CN109851664A (en) * 2017-11-30 2019-06-07 清华大学 A kind of protein based on the reversed epitope design of antibody and its in the application prepared in AIDS virus resisting vaccine
CN111363045A (en) * 2020-02-18 2020-07-03 厦门大学 Preparation method of influenza and HIV chimeric protein and chimeric virus vaccine

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116987719A (en) * 2023-06-21 2023-11-03 华南农业大学 H9N2AIV multi-epitope recombinant baculovirus, preparation method and application thereof
CN116987719B (en) * 2023-06-21 2024-03-15 华南农业大学 H9N2AIV multi-epitope recombinant baculovirus, preparation method and application thereof

Also Published As

Publication number Publication date
WO2022135563A1 (en) 2022-06-30

Similar Documents

Publication Publication Date Title
JP7250878B2 (en) Vaccines based on novel multivalent nanoparticles
JP6294828B2 (en) Influenza virus vaccine and use thereof
JP6643239B2 (en) Immunogenic middle east respiratory syndrome coronavirus (MERS-CoV) compositions and methods
ES2535421T3 (en) Immunogenic compositions in particulate form and methods to produce them
CN113164586B (en) Immune composition and preparation method and application thereof
EP3758747A1 (en) Self-asssembling nanostructure vaccines
CN113151184B (en) Method for cell membrane-based display of coronavirus immunogens to induce neutralizing antibodies
CN113666990A (en) T cell vaccine immunogen for inducing broad-spectrum anti-coronavirus and application thereof
CN113186173A (en) Novel coronavirus pneumonia vaccine based on attenuated influenza virus vector
CN114213548A (en) Method for simultaneously inducing immune response against multiple viruses
KR20140132332A (en) Vesicular stomatitis virus for prime boost vaccines
WO2021254270A1 (en) Method for inducing neutralizing antibody based on cell membrane to display coronavirus immunogen
KR20230084478A (en) Immunogenic coronavirus fusion proteins and related methods
WO2023023940A1 (en) Immunogen for inducing broad-spectrum anti-coronavirus t cell vaccine and use thereof
US20230174588A1 (en) A vaccine against sars-cov-2 and preparation thereof
WO2021253172A1 (en) Method for inducing anti-novel coronavirus neutralizing antibody using receptor recognition domain
CN113801206A (en) Method for inducing anti-neocoronavirus neutralizing antibody by using receptor recognition domain
JP2018052953A (en) Influenza vaccines and uses thereof
WO2021039873A1 (en) Composite protein monomer having non-structural protein of virus supported thereon, aggregate of composite protein monomer, and component vaccine comprising aggregate as active ingredient
EP4232084A1 (en) Vaccine
RU2701953C1 (en) Method of producing a polyvalent influenza vaccine
WO2023236822A1 (en) Development and use of h5n6 avian influenza broad-spectrum vaccine
RU2706191C1 (en) Multivalent influenza vaccine
WO2023207717A1 (en) Development and use of broad-spectrum vaccine for h5n8 avian influenza
US11535651B2 (en) Hepatitis B nanoparticle-based vaccine for influenza virus

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