CN116375881A - Fusion protein vaccine - Google Patents

Fusion protein vaccine Download PDF

Info

Publication number
CN116375881A
CN116375881A CN202211743150.2A CN202211743150A CN116375881A CN 116375881 A CN116375881 A CN 116375881A CN 202211743150 A CN202211743150 A CN 202211743150A CN 116375881 A CN116375881 A CN 116375881A
Authority
CN
China
Prior art keywords
antigen
domain
linker
pan
csf2
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
CN202211743150.2A
Other languages
Chinese (zh)
Inventor
彭华
曹学智
王修业
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.)
Guangzhou National Laboratory
Original Assignee
Guangzhou National Laboratory
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 Guangzhou National Laboratory filed Critical Guangzhou National Laboratory
Publication of CN116375881A publication Critical patent/CN116375881A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material

Abstract

The present invention provides a fusion protein comprising an antigen domain and an immune cell targeting domain.

Description

Fusion protein vaccine
Technical Field
The invention belongs to the field of vaccines, and particularly relates to a protein vaccine and application thereof.
Background
Subunit vaccine is a vaccine that contains a purified pathogen antigen fraction, or a fraction necessary to elicit a protective immune response. Subunit vaccines do not contain all pathogens like live attenuated or inactivated vaccines, but only antigenic parts like proteins, polysaccharides or peptides. Since the vaccine does not contain the "live" component of the pathogen, there is no risk of introducing the disease and is safer and more stable than a vaccine containing the complete pathogen. Other advantages of subunit vaccines include mature technology and suitability for immunocompromised individuals. A protein vaccine is one of subunit vaccines, containing proteins isolated from a pathogen (virus or bacteria). The recombinant protein vaccine is prepared through integrating some key segment of pathogen into bacterial, yeast, animal or insect cell, culturing in vitro, collecting and purifying, and adding adjuvant. The recombinant protein subunit vaccine has only the main surface protein of the pathogen, and avoids the generation of a plurality of antibodies induced by irrelevant antigens, thereby reducing the side reaction of the vaccine and the related diseases caused by the vaccine.
One of the disadvantages of recombinant protein vaccines is their low immunogenicity. The specific antigens used in recombinant protein vaccines may lack pathogen-associated molecular structures common to pathogens. These molecular structures can be used by immune cells to recognize the danger, so without them, the immune response will be weaker. A second disadvantage of recombinant protein vaccines is the lack of cellular immune responses. The recombinant protein subunit vaccine does not infect cells, and the immune response induced by the recombinant protein subunit vaccine is mainly antibody-mediated humoral immune response, but lacks important cellular immune response, which is unfavorable for effectively eliminating pathogens and protecting immunity for a long time. In order to enhance the immunogenicity of recombinant protein vaccines, subunit vaccines need to be used with adjuvants. This not only increases the dependency of the recombinant protein vaccine on the adjuvant, but also increases the complexity of the vaccine immunization procedure and the commercial cost of the vaccine.
Disclosure of Invention
The present invention provides fusion protein vaccines with enhanced immune effects comprising an antigen domain and an immune cell targeting domain. The fusion protein vaccine with enhanced immune effect has enhanced immune effect, enhances immune response induced by the recombinant protein vaccine, and reduces the dependency of the recombinant protein vaccine on an adjuvant.
In a first aspect, the invention provides fusion proteins comprising an antigen domain and an immune cell targeting domain.
The immune cell targeting domain is one or more domains selected from the group consisting of:
domain a: an antibody or polypeptide or active fragment thereof capable of binding an immune cell surface protein;
domain B: a cytokine or active fragment thereof capable of activating an immune cell;
domain C: a Pan epitope (PADRE) or active fragment thereof capable of activating immune cells;
domain D: immunoglobulin Fc capable of binding immune cells.
In some embodiments, the fusion protein comprises an antigen domain and domain a; fusion proteins of antigen domain with domain a and domain B; fusion proteins of antigen domain with domain a, domain B and domain C; fusion proteins of antigen domains with domain a, domain B, domain C and domain D; fusion proteins of antigen domain and domain B; fusion proteins of antigen domain with domain B and domain C; fusion proteins of antigen domain with domain B, domain C and domain D; fusion proteins of antigen domain and domain C; fusion proteins of antigen domain with domain C and domain D; or fusion proteins of an antigen domain and domain D.
The fusion protein can be any arrangement of antigen domains and domain a and/or domain B and/or domain C and/or domain D, e.g., the antigen domains and/or domain a and/or domain B and/or domain C and/or domain D are C-terminal or N-terminal with respect to each other in the fusion protein.
The domain A includes, but is not limited to, antibodies to CD274 (PDL 1), PDCD1LG2 (PDL 2), CLEC9A, LY75 (DEC 205), CD40, TNFSF9 (4-1 BB-L) and/or TNFSF4 (OX 4 OL), or active fragments of its ligands.
The domain B includes, but is not limited to, interleukin (IL) and/or Colony stimulating factor (Coloney-stimulating factor, CSF), or active fragments thereof.
Preferably, the interleukin comprises IL2, IL12, IL15 and/or IL21, or active fragments thereof.
Preferably, the colony stimulating factor comprises CSF1, CSF2 and/or CSF3, or an active fragment thereof.
Preferably, said domain C has the amino acid sequence shown in akfvaaawtlkaaa.
The domain D may be an Fc from IgG, igM, igA, igE or IgD, or a mutant thereof, which is a mutant to form a heterodimeric protein. In some embodiments, the Fc is a modified Fc, e.g., two Fc domains of a dimer have Fc knob modifications and Fc Hole modifications, respectively.
In some embodiments, the Antigen domain (anti) may be an immunogenic protein or immunogenic fragment thereof capable of inducing an immune response against a pathogenic microorganism.
The pathogenic microorganism may be SARS-Cov-2, SARS, cytomegalovirus CMV, herpes virus, respiratory syncytial virus RSV, influenza virus, ebola virus, epstein-Barr virus EBV, dengue virus, zike virus, HIV virus, rabies virus, plasmodium gametophyte, herpes zoster virus HZV, hepatitis B virus HBV, hepatitis C virus HCV, hepatitis B virus HDV, HPV, mycobacterium tuberculosis, helicobacter pylori, etc.
The antigen domain may be an immunogenic protein, or an immunogenic fragment thereof, capable of inducing an immune response against cancer cells.
In some embodiments, the antigen domain may be a tumor antigen domain, such as MelanA/MART1, cancer-germ line antigen, gp100, tyrosinase, CEA, PSA, her-2/neu, survivin, telomerase, or an immunogenic fragment thereof.
In some embodiments, the antigen domain and immune cell targeting domain, and/or immune cell targeting domain between can be connected by a connecting fragment.
In some embodiments, the linker segment may be a flexible linker segment, a rigid linker segment, or an in vivo shear linker segment. Wherein the amino acid sequence of the flexible linker fragment may be (G) N ,(GS) N ,(GGS) N ,(GGGS) N Or (GGGGS) N
In some embodiments, the fusion protein is a homodimer or a heterodimer.
In some embodiments, the fusion protein comprises an antigen domain and domain D, the fusion protein constituting a homodimer or a heterodimer in a similar antibody format. The fusion proteins consist of a homodimer or a heterodimer of disulfide bonds of two Fc domains. Preferably, the fusion protein comprises a first polypeptide chain comprising an antigen domain and domain D and a second polypeptide chain comprising an antigen domain and domain D. The first polypeptide chain and the second polypeptide chain are linked via disulfide bonds of domain D of the first polypeptide chain and domain D of the second polypeptide chain to form a homodimer or a heterodimer. Preferably, the first polypeptide chain further comprises domain B and/or domain C, and the second polypeptide chain further comprises domain B and/or domain C. For example, the first polypeptide chain further comprises domain B, and the second polypeptide chain further comprises domain B, thereby constituting a homodimer. Alternatively, the first polypeptide chain further comprises domain B and the second polypeptide chain further comprises domain C, thereby constituting a heterodimer and vice versa. The antigen domain and/or domain B and/or domain C may be linked at the C-terminus or N-terminus of domain D. For example, the first polypeptide chain further comprises domain B and domain C, and the second polypeptide chain further comprises domain B and domain C, thereby constituting a homodimer.
Alternatively, the first polypeptide chain comprises domain D, antigen domain, domain a from C-terminus to N-terminus, and the second polypeptide chain comprises domain D, antigen domain, domain a from C-terminus to N-terminus, thereby constituting a homodimer and vice versa.
Alternatively, the first polypeptide chain comprises domain D, antigen domain, domain B from C-terminus to N-terminus, and the second polypeptide chain comprises domain D, antigen domain, domain B from C-terminus to N-terminus, thereby constituting a homodimer, and vice versa.
Alternatively, the first polypeptide chain comprises domain D, antigen domain, domain C, domain a from C-terminus to N-terminus, and the second polypeptide chain comprises domain D, antigen domain, domain C, domain a from C-terminus to N-terminus, thereby constituting a homodimer, and vice versa.
Alternatively, the first polypeptide chain comprises domain D, antigen domain, domain C, domain B from C-terminus to N-terminus, and the second polypeptide chain comprises domain D, antigen domain, domain C, domain B from C-terminus to N-terminus, thereby constituting a homodimer, and vice versa.
Alternatively, the first polypeptide chain comprises domain B, domain D, antigen domain a from C-terminus to N-terminus, and the second polypeptide chain comprises domain B, domain D, antigen domain, domain a from C-terminus to N-terminus, thereby constituting a homodimer.
Alternatively, the first polypeptide chain comprises from C-terminus to N-terminus domain B, domain D, antigen domain, domain C, domain a, and the second polypeptide chain comprises from C-terminus to N-terminus domain B, domain D, antigen domain, domain C, domain a, thereby constituting a homodimer, and vice versa.
Alternatively, the first polypeptide chain comprises domain D, antigen domain, domain a from C-terminus to N-terminus, and the second polypeptide chain comprises domain D, antigen domain, domain C, domain B from C-terminus to N-terminus, thereby constituting a heterodimer and vice versa.
Alternatively, the first polypeptide chain comprises domain D, antigen domain, domain C, domain a from C-terminus to N-terminus, and the second polypeptide chain comprises domain D, antigen domain, domain C, domain B from C-terminus to N-terminus, thereby constituting a heterodimer, and vice versa.
Preferably, in the case of heterodimers, domain D has an Fc knob modification and an Fc Hole modification, respectively.
The antigen domain and the immune cell targeting domain can be arranged and combined in any form. The antigen domain and the immune cell targeting domain A, domain B, domain C, domain D can be arranged and combined in any form.
The antigen and the immune cell targeting molecule A can be arranged and combined in any form. The antigen and the immune cell targeting molecule B can be arranged and combined in any form. The antigen and the immune cell targeting molecule C can be arranged and combined in any form.
In some embodiments, the fusion protein has a structure selected from the group consisting of:
αPDL1-Antigen-Fc;
CLEC9A binding peptide-Antigen-Fc;
αDEC205-Antigen-Fc;
Antigen-Fc-CLEC9A binding peptide;
αPDL1-linker-Antigen-linker-Fc;
αPDL1-(GGGGS) 3 -Antigen-(G) 3 -Fc;
CLEC9A binding peptide-linker-Antigen-linker-Fc;
CLEC9A binding peptide-(GGGGS) 3 -Antigen-(G) 3 -Fc;
αDEC205-linker-Antigen-linker-Fc;
αDEC205-(GGGGS) 3 -Antigen-(G) 3 -Fc;
Antigen-linker-Fc-linker-CLEC9A binding peptide;
Antigen-(G) 3 -Fc-(GS) 3 -CLEC9A binding peptide;
IL2-Antigen-Fc;
IL12-Antigen-Fc;
IL15-Antigen-Fc;
IL21-Antigen-Fc;
CSF2-Antigen-Fc;
Antigen-Fc-CSF2;
IL2-linker-Antigen-linker-Fc;
IL2-(GGGGS) 3 -Antigen-(G) 3 -Fc;
IL12-linker-Antigen-linker-Fc;
IL12-(GGGGS) 3 -Antigen-(G) 3 -Fc;
IL15-linker-Antigen-linker-Fc;
IL15-(GGGGS) 3 -Antigen-(G) 3 -Fc;
IL21-linker-Antigen-linker-Fc;
IL21-(GGGGS) 3 -Antigen-(G) 3 -Fc;
CSF2-linker-Antigen-linker-Fc;
CSF2-(GGGGS) 3 -Antigen-(G) 3 -Fc;
Antigen-linker-Fc-linker-CSF2;
Antigen-(GGGGS) 3 -Fc-(G) 3 -CSF2;
αPDL1-Pan-Antigen-Fc;
CLEC9A binding peptide-Pan-Antigen-Fc;
αDEC205-Pan-Antigen-Fc;
αPDL1-linker-Pan-Antigen-linker-Fc;
αPDL1-(GGGGS) 3 -Pan-Antigen-(G) 3 -Fc;
CLEC9A binding peptide-linker-Pan-Antigen-linker-Fc;
CLEC9A binding peptide-(GGGGS) 3 -Pan-Antigen-(G) 3 -Fc;
αDEC205-linker-Pan-Antigen-linker-Fc;
αDEC205-(GGGGS) 3 -Pan-Antigen-(G) 3 -Fc;
IL2-Pan-Antigen-Fc;
IL12-Pan-Antigen-Fc;
IL15-Pan-Antigen-Fc;
IL21-Pan-Antigen-Fc;
CSF2-Pan-Antigen-Fc;
Pan-Antigen-Fc-CSF2;
IL2-linker-Pan-linker-Antigen-(G) 3 -Fc;
IL2-(GGGGS) 3 -Pan-(GS) 3 -Antigen-linker-Fc;
IL12-linker-Pan-linker-Antigen-linker-Fc;
IL12-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc;
IL15-linker-Pan-linker-Antigen-linker-Fc;
IL15-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc;
IL21-linker-Pan-linker-Antigen-linker-Fc;
IL21-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc;
CSF2-linker-Pan-linker-Antigen-linker-Fc;
CSF2-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc;
Pan-linker-Antigen-linker-Fc-linker-CSF2;
Pan-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -CSF2;
αPDL1-Antigen-Fc-IL12;
αPDL1-Antigen-Fc-IL21;
αPDL1-Antigen-Fc-CSF2;
CLEC9A binding peptide-Antigen-Fc-IL12;
CLEC9A binding peptide-Antigen-Fc-IL21;
CLEC9A binding peptide-Antigen-Fc-CSF2;
αPDL1-linker-Antigen-linker-Fc-linker-IL12;
αPDL1-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL12;
αPDL1-linker-Antigen-linker-Fc-linker-IL21;
αPDL1-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL21;
αPDL1-linker-Antigen-linker-Fc-linker-CSF2;
αPDL1-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -CSF2;
CLEC9A binding peptide-linker-Antigen-linker-Fc-linker-IL12;
CLEC9A binding peptide-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL12;
CLEC9A binding peptide-linker-Antigen-linker-Fc-linker-IL21;
CLEC9A binding peptide-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL21;
CLEC9A binding peptide-linker-Antigen-linker-Fc-linker-CSF2;
CLEC9A binding peptide-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -CSF2;
αPDL1-Pan-Antigen-Fc-IL12;
αPDL1-Pan-Antigen-Fc-IL21;
αPDL1-Pan-Antigen-Fc-CSF2;
CLEC9A binding peptide-Pan-Antigen-Fc-IL12;
CLEC9A binding peptide-Pan-Antigen-Fc-IL21;
CLEC9A binding peptide-Pan-Antigen-Fc-CSF2;
αPDL1-CSF2-Pan-Antigen-Fc;
αPDL1-linker-Pan-linker-Antigen-linker-Fc-linker-IL12;
αPDL1-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL12;
αPDL1-linker-Pan-linker-Antigen-linker-Fc-linker-IL21;
αPDL1-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL21;
αPDL1-linker-Pan-linker-Antigen-linker-Fc-linker-CSF2;
αPDL1-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -CSF2;
CLEC9A binding peptide-linker-Pan-linker-Antigen-linker-Fc-linker-IL12;
CLEC9A binding peptide-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL12;CLEC9A binding peptide-linker-Pan-linker-Antigen-linker-Fc-linker-IL21;
CLEC9A binding peptide-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL21;CLEC9A binding peptide-linker-Pan-linker-Antigen-linker-Fc-linker-CSF2;
CLEC9A binding peptide-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -CSF2;αPDL1-linker-CSF2-linker-Pan-linker-Antigen-linker-Fc;
αPDL1-(GGGGS) 3 -CSF2-(GS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc;
αPDL1-Antigen-Fc knob/CSF2-Antigen-Fc hole;
CLEC9A binding peptide-Antigen-Fc knob/CSF2-Antigen-Fc hole;
αPDL1-linker-Antigen-linker-Fc knob/CSF2-linker-Antigen-linker-Fc hole;
αPDL1-(GGGGS) 3 -Antigen-(G) 3 -Fc knob/CSF2-(GGGGS) 3 -Antigen-(G) 3 -Fchole;
CLEC9A binding peptide-linker-Antigen-linker-Fc knob/
CSF2-linker-Antigen-linker-Fc hole;
CLEC9A binding peptide-(GGGGS) 3 -Antigen-(G) 3 -Fc knob/
CSF2-(GGGGS) 3 -Antigen-(G) 3 -Fc hole;
αPDL1-Pan-Antigen-Fc knob/CSF2-Pan-Antigen-Fc hole;
CLEC9A binding peptide-Pan-Antigen-Fc knob/CSF2-Pan-Antigen-Fc hole;αPDL1-linker-Pan-linker-Antigen-linker-Fc knob/
CSF2-linker-Pan-linker-Antigen-linker-Fc hole;
αPDL1-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc knob/
CSF2-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc hole;
CLEC9A binding peptide-linker-Pan-linker-Antigen-linker-Fc knob/CSF2-linker-Pan-linker-Antigen-linker-Fc hole;
CLEC9A binding peptide-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc knob/
CSF2-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc hole。
in some embodiments, the antigenic domain is the S protein of SARS-Cov-2 or a fragment thereof. Preferably, the S protein is a prefusion stable S protein. In some embodiments, the pre-fusion stable S protein comprises a biproline (S2P) mutation or a hexapro (S6P) mutation.
In some embodiments, the antigen domain is the RBD domain of the S protein of SARS-Cov-2.
The fusion protein of the invention can be used as a recombinant protein vaccine.
In a second aspect, the invention provides a nucleic acid molecule for encoding the fusion protein of the first aspect.
In a third aspect, the invention provides a vector for expressing the fusion protein of the first aspect or comprising the nucleic acid molecule of the second aspect.
The vector may be a plasmid vector, an adenovirus vector or a lentiviral vector.
In a fourth aspect, the invention provides a host cell for expressing the fusion protein of the first aspect or a nucleic acid molecule comprising the second aspect, or the vector of the third aspect.
The host cell may be, for example, chinese hamster ovary cells (CHO cells), baby hamster kidney cells (BHK cells), COS cells, mouse NSO thymoma cells, mouse myeloma cells (SP 2/0 cells), human embryonic kidney cells HEK293 cells.
In a fifth aspect, the invention provides a composition comprising the fusion protein of the first aspect, or the nucleic acid molecule of the second aspect, or the vector of the third aspect, or the host cell of the fourth aspect.
In some embodiments, the composition does not comprise a pharmaceutically acceptable adjuvant.
In some embodiments, the composition further comprises a pharmaceutically acceptable adjuvant. Such pharmaceutically acceptable adjuvants include, but are not limited to, aluminum adjuvants such as aluminum hydroxide, aluminum phosphate or sulfate, or CpG adjuvants.
In a sixth aspect, the present invention provides a method for preventing or treating infection with a pathogenic microorganism or a tumor, the method comprising the step of administering to a subject the fusion protein of the first aspect, the nucleic acid molecule of the second aspect, the vector of the third aspect, the host cell of the fourth aspect or the composition of the fifth aspect.
The subject is a human or animal. The animals include, but are not limited to, cattle, sheep, cats, dogs, horses, rabbits, monkeys, mice, rats, alpacas, camels, etc.
In some embodiments, the subject is a immunocompromised human or animal.
In some embodiments, the subject has chronic lung disease, e.g., chronic obstructive pulmonary disease or asthma.
In some embodiments, the patient has a basal disease, such as heart disease, diabetes, or lung disease.
In a seventh aspect, the invention provides the use of a fusion protein of the first aspect, a nucleic acid molecule of the second aspect, a vector of the third aspect, a host cell of the fourth aspect or a composition of the fifth aspect in the manufacture of a medicament for preventing or treating infection or tumour associated with a pathogenic microorganism in a subject.
The subject is a human or animal. The animals include, but are not limited to, cattle, sheep, cats, dogs, horses, rabbits, monkeys, mice, rats, alpacas, camels, chickens, ducks, geese, etc.
In some embodiments, the subject is a immunocompromised human or animal.
In some embodiments, the subject has chronic lung disease, e.g., chronic obstructive pulmonary disease or asthma.
In some embodiments, the patient has a basal disease, such as heart disease, diabetes, or lung disease.
The cancer may be prostate cancer, non-small cell lung cancer, renal cell carcinoma, brain cancer, melanoma, acute myelogenous leukemia, pancreatic cancer, colorectal cancer, head and neck squamous cell carcinoma, cutaneous squamous cell carcinoma, adenoid cystic carcinoma, glioblastoma, breast cancer, mesothelioma, ovarian cancer, glioma, bladder cancer, liver cancer, bone marrow cancer, gastric cancer, thyroid cancer, lymphoma, cervical cancer, endometrial cancer, laryngeal cancer, acute lymphoblastic leukemia, and the like.
In some embodiments, the antigen is a tumor antigen including, but not limited to 5T4,AIM2,AKAP4 2,Art-4, auraA1 (AURKA), aura B1 (AURKB), BAGE, BCANs, B-cyclin, BSG, CCND1, CD133, CDC45L, CDCA1 (TTK), CEA, CHI3L2 (chitinase 3-like 2), CSPG4, epCAM4, epha2, EPHX1, ezh2, FABP7, fosl1 (Fra-1), GAGE, galt-3, G250 (CA 9), gBK, glass, gnT-V, gp100, HB-EGF, HER2, HNPRL, HO-1, hTERT, IGF2BP3, IL13-Ra2, IMP-3, IQGAP1, ITGAV, KIF1C, KIF20A, KIF21B, KIFC3, KK-LC-1, LAGE-1, lck, LRRC8A, MAGE-1 (MAGEA 1), MAGE-2 (MAGEA 2B), MAGE-3, MAGE-4, MAGE-6, MAGE-10, MAGE-12, MAGE-C1 (CT 7), MAGE-C2, MAGE-C3, mart-1, MELK, MRP3, MUC1, NAPSA, NLGN4X, nam, NY-ESO-1 (CTAG 1B), NY-SAR-35, OFA/iLRP, PCNA, PIK R1, prame, PRKDC, PTH-rP, PTPRZ1, PTTG 12, PRKDC, RAN, RGS1, RGS5, RHAMM (RHAMM-3R), RPL19, sart-1, sart-2, sart-3, SEC61G, SGT-1, SOX2, sox10, sox11, SP17, SPANX-B, SQSTM1, S.S.X-2, STAT1, STAT3, survivin, TARA, TNC, trag-3, TRP-1, TRP2, tyrosinase, URLC10 (LY 6K), ube2V, WT1, XAGE-1B (GAGED 2 a), YKL-40 (CHI 3L 1), ACRBP, SCP-1, S.S.X-4, NY-TLU-57, CAIX, brachyury, NY-BR-1, erbB, mesothelin, EGFRvIII, IL-13Ra2, MSLN, GPC3, FR, PSMA, GD2, L1-CAM, VEGFR1, VEGFR2, KOC1, OFA, SL-701, mutant P53, DEPDC1, MPHOSPH1, ONT-10,GD2L,GD3L,TF,PAP,BRCA1 DLC1,XPO1,HIF1A,ADAM2,CALR3,SAGE1,SCP-1, ppMAPkkk, WHSC, mutant Ras, COX1, COX2, FOXP3, IDO1, IDO2, TDO, PDL1, PDL2 and PGE2.
In some embodiments, the antigen includes, but is not limited to, antigens associated with any tumor/cancer, such as lung cancer (MTFR 2D 326Y, CHTF 18L 769V, myom R30W, HERC 1P 3278S, FAM3C K193E, CSMD1G3446E, SLC26 A7R 117Q, ap 1Y 903F, HELB P987S, ANKRD K603T); melanoma (TMEM 48F169L, TKT R438W, SEC24A P469L, AKAP 13Q 285K, EXOC 8Q 656P, PABPC 1R 520Q, MRPS5P59L, ABCC 2S 1342F, SEC23A P L, SYTL 4S 363F, MAP3K 9E 689K, AKAP 6M 1482I, RPBMP42L, HCAPG 2P 333L, H3F3C T4I, GABPA E161K, SEPT 2Q 125R, SRPX P55L, WDR46T300I, PRDX 3P 101L, HELZ 2D 614N, GCN1L 1P 769L, AFMID A52V, CR4R247C, CENPL P79L, TPX2H458Y, SEC22C H Y, POLA 2L 420F, SLC24A5 mut); mesothelioma (NOTCH 2G 703D, PDE4DIPL288M, BAP 1V 523fs, ATP10B E K, NSD 1K 2482T); glioma/glioblastoma (IDH 1R 132H, hole L424V); breast cancer (mPALB 2, mrrobo 3, mzdhc 16, mPTPRS, RBPJ H204L); cholangiocarcinoma ((ERBB 2IPE 805G); and cervical carcinoma (MAPK 1E 322K, PIK3CA E545K, PIK3CA E542K, EP 300D 1399N, ERBB2S310F, ERBB 3V 104M, KRAS G12D).
Such pathogenic microorganism-related diseases include, but are not limited to: acquired immunodeficiency syndrome (AIDS) (human immunodeficiency virus (HIV)); argentina root-extracted heat (ArgentineTeagan fever) (Junin virus); astrovirus infection (astroviridae); BK viral infection (BK virus); bolivia hemorrhagic fever (Machupo virus); brazil hemorrhagic fever (sabia virus); varicella (varicella zoster virus (VZV)); chikungunya heat (Chikungunya) (alphavirus); colorado tick heat transfer (Colorado tick fever) (CTF) (Colorado tick heat transfer virus (Colorado tick fever virus) (CTFV)); common cold, acute viral nasopharyngitis, acute rhinitis (usually rhinovirus and coronavirus); cytomegalovirus infection (cytomegalovirus); dengue fever (dengue-1, DEN-2, DEN-3 and DEN-4) and other flaviviruses including, but not limited to, west Nile virus (West Nile fever), yellow fever virus (yellow fever), zika virus (Zika fever) and tick-borne encephalitis virus, ebola hemorrhagic fever (Ebola virus (EBOV)), enterovirus infection (enterovirus species), infectious erythema disease (fifth disease) (parvovirus B19), infant eruption (Exanthemsubum) (sixth disease) (human herpesvirus 6 (HHV-6) and human herpesvirus 7 (HHV-7)), hand-foot-mouth disease (HFMD) (enterovirus, mainly Coxsackie A virus and enterovirus 71 (EV 71)), hantaan virus pulmonary syndrome (HantavirusPulmonary Syndrome, HPS) (Sin nompre virus); hepatitis a (hepatitis a virus); hepatitis b (hepatitis b virus); hepatitis c (hepatitis c virus); hepatitis d (hepatitis d virus); hepatitis E (hepatitis E Virus); herpes simplex (herpes simplex virus 1 and 2 (HSV-1 and HSV-2)); human bocavirus infection (human bocavirus (HBoV)); human interstitial pneumovirus infection (human interstitial pneumovirus (hMPV)); human Papillomavirus (HPV) infection (human papillomavirus (HPV)); human parainfluenza virus infection (human parainfluenza virus (HPIV)); epstein-barr virus infectious mononucleosis (Mono) (epstein-barr virus (EBV)); human influenza viruses (influenza a, including but not limited to H1N1, H2N2, H3N2, H5N1, H7N9, influenza b and other members of the orthomyxoviridae family); lassa fever (lassa virus); lymphocytic choriomeningitis (lymphocytic choriomeningitis virus (LCMV)); marburg Hemorrhagic Fever (MHF) (Marburg virus); measles (measles virus); middle East Respiratory Syndrome (MERS) (middle east respiratory syndrome coronavirus); molluscum Contagiosum (MC) (molluscum contagiosum virus (MCV)); monkey pox (monkey pox virus); mumps (mumps virus); norovirus (Norovirus) (children and infants) (Norovirus); polio (poliovirus); progressive multifocal leukoencephalopathy (JC virus); rabies (rabies virus); respiratory syncytial virus infection (respiratory syncytial virus (RSV)); rhinovirus infection (rhinovirus); setaria valgus (RVF) (Setaria Virus); rotavirus infection (rotavirus); rubella (rubella virus); herpes zoster (shawl) (Herpes zoster (varicella zoter)) (varicella zoster virus (VZV)); smallpox (Smallpox) (Variola) (Smallpox) or Smallpox; subacute sclerotic panencephalitis (measles virus); venezuelan equine encephalitis (venezuelan equine encephalitis virus); venezuelan hemorrhagic fever (melon narcissus virus); viral pneumonia.
The recombinant protein vaccine provided by the invention has the advantages of improved immunogenicity, enhanced immune response and reduced dependence of the recombinant protein vaccine on an adjuvant.
The design of antigen fusion expression in the protein vaccine can be applied to other types of vaccines as well, including but not limited to nucleic acid vaccines (such as mRNA vaccines, circular RNA vaccines, DNA vaccines and the like), viral vector vaccines (such as adenovirus vector vaccines, influenza virus vector vaccines and the like), nanoparticle vaccines and the like.
The invention shows that:
(1) Free RBD proteins are less immunogenic, have increased immunogenicity when the Fc portion is added on RBD, have further increased immunogenicity when cytokines IL12 and Pan are added on RBD-Fc, have increased immunogenicity when cytokines IL21 and Pan are added on RBD-Fc, have increased immunogenicity when cytokines CSF2 and Pan are added on RBD-Fc, and have increased immunogenicity when alpha PDL1 antibodies and Pan capable of binding the immune cell surface protein PDL1 are added on RBD-Fc;
(2) Free RBD proteins are less immunogenic, and can be increased in antibody titer when the Fc moiety is added on RBD, further increased in antibody titer when cytokine CSF2 is added on RBD-Fc, and further increased in antibody titer when an alpha PDL1 antibody capable of binding immune cell surface protein PDL1 is added on RBD-Fc;
(3) The free gp350 protein vaccine is less immunogenic, and can increase antibody titer when the cytokine CSF2 is added on the basis of gp350-Fc, antibody titer when the alpha PDL1 antibody capable of binding to the immune cell surface protein PDL1 is added on the basis of gp350-Fc, antibody titer when the cytokines CSF2 and Pan are added on the basis of gp350-Fc, and antibody titer when the alpha PDL1 antibody and Pan capable of binding to the immune cell surface protein PDL1 are added on the basis of gp 350-Fc.
Drawings
FIG. 1 shows a schematic representation of a homodimeric version of the fusion protein vaccine. Wherein, the antigen-Fc and the immune cell targeting molecule A are arranged and combined according to the sequence A-antigen-Fc.
FIG. 2 shows a schematic representation of a homodimeric version of the fusion protein vaccine. Wherein, the antigen-Fc and the immune cell targeting molecule B are arranged and combined according to the sequence of B-antigen-Fc.
FIG. 3 shows a schematic representation of a homodimeric version of the fusion protein vaccine. Wherein, the antigen-Fc and the immune cell targeting molecules A and C are arranged and combined according to the sequence of A-C-antigen-Fc.
FIG. 4 shows a schematic representation of a homodimeric version of the fusion protein vaccine. Wherein, the antigen-Fc and the immune cell targeting molecules B and C are arranged and combined according to the sequence of B-C-antigen-Fc.
FIG. 5 shows a schematic representation of a homodimeric version of the fusion protein vaccine. Wherein, the antigen-Fc and the immune cell targeting molecules A and B are arranged and combined according to the sequence of A-antigen-Fc-B.
FIG. 6 shows a schematic representation of a homodimeric version of the fusion protein vaccine. Wherein, the antigen-Fc and the immune cell targeting molecules A, B and C are arranged and combined according to the sequence of A-C-antigen-Fc-B.
FIG. 7 shows a schematic representation of a fusion protein vaccine in heterodimeric form. Wherein, antigen-Fcknob and immune cell targeting molecule A are arranged and combined according to the sequence of A-antigen-Fcknob, and antigen-Fchole and immune cell targeting molecule B are arranged and combined according to the sequence of B-antigen-Fchole.
FIG. 8 shows a schematic representation of a fusion protein vaccine in heterodimeric form. Wherein the antigen-Fcknob and the immune cell targeting molecules A and C are arranged and combined according to the sequence of A-C-antigen-Fcknob, and the antigen-Fchole and the immune cell targeting molecules B and C are arranged and combined according to the sequence of B-C-antigen-Fchole.
FIG. 9 shows a polyacrylamide gel (SDS-PAGE) electrophoretic identification of SARS-CoV-2 spike protein RBD associated immune cell targeting fusion protein. Wherein the proteins corresponding to RBD, RBD-Fc, IL4-Pan-RBD-Fc, IL10-Pan-RBD-Fc, IL12-Pan-RBD-Fc, IL21-Pan-RBD-Fc, CSF2-Pan-RBD-Fc, PD1-Pan-RBD-Fc, PDL1-Pan-RBD-Fc are identified on SDS-PAGE, respectively.
FIG. 10 shows the protein yield of the SARS-CoV-2 spike protein RBD-associated immune cell targeted fusion protein vaccine transiently expressed in eukaryotic cells 293F.
FIG. 11 shows the results of an immune cell targeted fusion protein vaccine induced antibody response against SARS-CoV-2 spike protein RBD.
FIG. 12 shows SDS-PAGE electrophoretic identification of novel coronavirus SARS-CoV-2 spike protein RBD-associated fusion protein.
FIG. 13 shows the expression yield of the novel coronavirus SARS-CoV-2 spike protein RBD-associated fusion protein.
FIG. 14 shows that immune cell targeting of the novel coronavirus SARS-CoV-2 spike protein RBD associated protein vaccine can elicit a stronger antibody response in mice than a pure RBD protein vaccine.
FIG. 15 shows an SDS-PAGE electrophoretic identification of EB virus (Epstein-Barr virus, EBV) membrane protein gp 350-associated fusion protein.
FIG. 16 shows the expression yield of the EB virus (Epstein-Barr virus, EBV) membrane protein gp 350-associated fusion protein.
Figure 17 shows that immune cell-targeted EBV membrane protein gp 350-associated protein vaccines can elicit stronger antibody responses in mice than do gp 350-protein vaccines alone.
Sequence:
>PAN
AKFVAAWTLKAAA
>IgG1 Fc_HUMAN
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
>CSF2_HUMAN
APARSPSPSTQPWEHVNAIQEARRLLNLSRDTAAEMNETVEVISEMFDLQEPTCLQTRLELYKQGLRGSLTKLKGPLTMMASHYKQHCPPTPETSCATQIITFESFKENLKDFLLVIPFDCWEPVQE
>IL12A_HUMAN
RNLPVATPDPGMFPCLHHSQNLLRAVSNMLQKARQTLEFYPCTSEEIDHEDITKDKTSTVEACLPLELTKNESCLNSRETSFITNGSCLASRKTSFMMALCLSSIYEDLKMYQVEFKTMNAKLLMDPKRQ
IFLDQNMLAVIDELMQALNFNSETVPQKSSLEEPDFYKTKIKLCILLHAFRIRAVTIDRVMSYLN
AS
>IL12B_HUMAN
IWELKKDVYVVELDWYPDAPGEMVVLTCDTPEEDGITWTLDQSSEVLGSGKTLTIQVKEFGDAGQ
YTCHKGGEVLSHSLLLLHKKEDGIWSTDILKDQKEPKNKTFLRCEAKNYSGRFTCWWLTTISTDL
TFSVKSSRGSSDPQGVTCGAATLSAERVRGDNKEYEYSVECQEDSACPAAEESLPIEVMVDAVHK
LKYENYTSSFFIRDIIKPDPPKNLQLKPLKNSRQVEVSWEYPDTWSTPHSYFSLTFCVQVQGKSK
REKKDRVFTDKTSATVICRKNASISVRAQDRYYSSSWSEWASVPCS
>IL21_HUMAN
HKSSSQGQDRHMIRMRQLIDIVDQLKNYVNDLVPEFLPAPEDVETNCEWSAFSCFQKAQLKSANT
GNNERIINVSIKKLKRKPPSTNAGRRQKHRLTCPSCDSYEKKPPKEFLERFKSLLQKMIHQHLSS
RTHGSEDS
>SP-Human CSF2-(GGGGS) 3 -RBD-(G) 3 -Human IgG1 Fc
METDTLLLWVLLLWVPGSTGDAPARSPSPSTQPWEHVNAIQEARRLLNLSRDTAAEMNETVEVIS
EMFDLQEPTCLQTRLELYKQGLRGSLTKLKGPLTMMASHYKQHCPPTPETSCATQIITFESFKEN
LKDFLLVIPFDCWEPVQEFEGGGGSGGGGSGGGGSSRRVQPTESIVRFPNITNLCPFGEVFNATR
FASVYAWNRKRISNCVADYSVLYNSASFSTFKCYGVSPTKLNDLCFTNVYADSFVIRGDEVRQIA
PGQTGKIADYNYKLPDDFTGCVIAWNSNNLDSKVGGNYNYLYRLFRKSNLKPFERDISTEIYQAG
STPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVVVLSFELLHAPATVCGPKKSTNLVKNKCVNF
LEGGGDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVD
GVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE
PQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKL
TVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
>SP-Human IL12B-(GGGGS) 3 -Human IL12A-(GGGGS) 3 -RBD-(G) 3 -Human IgG1 FcMETDTLLLWVLLLWVPGSTGDIWELKKDVYVVELDWYPDAPGEMVVLTCDTPEEDGITWTLDQSSEVLGSGKTLTIQVKEFGDAGQYTCHKGGEVLSHSLLLLHKKEDGIWSTDILKDQKEPKNKTFLRCEAKNYSGRFTCWWLTTISTDLTFSVKSSRGSSDPQGVTCGAATLSAERVRGDNKEYEYSVECQEDSACPAAEESLPIEVMVDAVHKLKYENYTSSFFIRDIIKPDPPKNLQLKPLKNSRQVEVSWEYPDTWSTPHSYFSLTFCVQVQGKSKREKKDRVFTDKTSATVICRKNASISVRAQDRYYSSSWSEWASVPCSGGGGSGGGGSGGGGSRNLPVATPDPGMFPCLHHSQNLLRAVSNMLQKARQTLEFYPCTSEEIDHEDITKDKTSTVEACLPLELTKNESCLNSRETSFITNGSCLASRKTSFMMALCLSSIYEDLKMYQVEFKTMNAKLLMDPKRQIFLDQNMLAVIDELMQALNFNSETVPQKSSLEEPDFYKTKIKLCILLHAFRIRAVTIDRVMSYLNASFEGGGGSGGGGSGGGGSSRRVQPTESIVRFPNITNLCPFGEVFNATRFASVYAWNRKRISNCVADYSVLYNSASFSTFKCYGVSPTKLNDLCFTNVYADSFVIRGDEVRQIAPGQTGKIADYNYKLPDDFTGCVIAWNSNNLDSKVGGNYNYLYRLFRKSNLKPFERDISTEIYQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVVVLSFELLHAPATVCGPKKSTNLVKNKCVNFLEGGGDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
>SP-Human IL21-(GGGGS) 3 -RBD-(G) 3 -Human IgG1 Fc
METDTLLLWVLLLWVPGSTGDHKSSSQGQDRHMIRMRQLIDIVDQLKNYVNDLVPEFLPAPEDVE
TNCEWSAFSCFQKAQLKSANTGNNERIINVSIKKLKRKPPSTNAGRRQKHRLTCPSCDSYEKKPP
KEFLERFKSLLQKMIHQHLSSRTHGSEDSFEGGGGSGGGGSGGGGSSRRVQPTESIVRFPNITNL
CPFGEVFNATRFASVYAWNRKRISNCVADYSVLYNSASFSTFKCYGVSPTKLNDLCFTNVYADSF
VIRGDEVRQIAPGQTGKIADYNYKLPDDFTGCVIAWNSNNLDSKVGGNYNYLYRLFRKSNLKPFE
RDISTEIYQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVVVLSFELLHAPATVCGPKKS
TNLVKNKCVNFLEGGGDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
DPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEK
TISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD
SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
>SP-Human CSF2-(GGGGS) 3 -Pan-(GS) 3 -RBD-(G) 3 -Human IgG1 Fc
METDTLLLWVLLLWVPGSTGDAPARSPSPSTQPWEHVNAIQEARRLLNLSRDTAAEMNETVEVIS
EMFDLQEPTCLQTRLELYKQGLRGSLTKLKGPLTMMASHYKQHCPPTPETSCATQIITFESFKEN
LKDFLLVIPFDCWEPVQEFEGGGGSGGGGSGGGGSAKFVAAWTLKAAAGSGSGSSRRVQPTESIV
RFPNITNLCPFGEVFNATRFASVYAWNRKRISNCVADYSVLYNSASFSTFKCYGVSPTKLNDLCF
TNVYADSFVIRGDEVRQIAPGQTGKIADYNYKLPDDFTGCVIAWNSNNLDSKVGGNYNYLYRLFR
KSNLKPFERDISTEIYQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVVVLSFELLHAPA
TVCGPKKSTNLVKNKCVNFLEGGGDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTC
VVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNK
ALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY
KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
>SP-Human IL12B-(GGGGS) 3 -Human IL12A-(GGGGS) 3 -Pan-(GS) 3 -RBD-(G) 3 -Human IgG1
Fc
METDTLLLWVLLLWVPGSTGDIWELKKDVYVVELDWYPDAPGEMVVLTCDTPEEDGITWTLDQSS
EVLGSGKTLTIQVKEFGDAGQYTCHKGGEVLSHSLLLLHKKEDGIWSTDILKDQKEPKNKTFLRC
EAKNYSGRFTCWWLTTISTDLTFSVKSSRGSSDPQGVTCGAATLSAERVRGDNKEYEYSVECQED
SACPAAEESLPIEVMVDAVHKLKYENYTSSFFIRDIIKPDPPKNLQLKPLKNSRQVEVSWEYPDT
WSTPHSYFSLTFCVQVQGKSKREKKDRVFTDKTSATVICRKNASISVRAQDRYYSSSWSEWASVP
CSGGGGSGGGGSGGGGSRNLPVATPDPGMFPCLHHSQNLLRAVSNMLQKARQTLEFYPCTSEEID
HEDITKDKTSTVEACLPLELTKNESCLNSRETSFITNGSCLASRKTSFMMALCLSSIYEDLKMYQ
VEFKTMNAKLLMDPKRQIFLDQNMLAVIDELMQALNFNSETVPQKSSLEEPDFYKTKIKLCILLH
AFRIRAVTIDRVMSYLNASFEGGGGSGGGGSGGGGSAKFVAAWTLKAAAGSGSGSSRRVQPTESI
VRFPNITNLCPFGEVFNATRFASVYAWNRKRISNCVADYSVLYNSASFSTFKCYGVSPTKLNDLC
FTNVYADSFVIRGDEVRQIAPGQTGKIADYNYKLPDDFTGCVIAWNSNNLDSKVGGNYNYLYRLF
RKSNLKPFERDISTEIYQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVVVLSFELLHAP
ATVCGPKKSTNLVKNKCVNFLEGGGDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVT
CVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSN
KALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENN
YKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
>SP-Human IL21-(GGGGS) 3 -Pan-(GS) 3 -RBD-(G) 3 -Human IgG1 Fc
METDTLLLWVLLLWVPGSTGDHKSSSQGQDRHMIRMRQLIDIVDQLKNYVNDLVPEFLPAPEDVE
TNCEWSAFSCFQKAQLKSANTGNNERIINVSIKKLKRKPPSTNAGRRQKHRLTCPSCDSYEKKPP
KEFLERFKSLLQKMIHQHLSSRTHGSEDSFEGGGGSGGGGSGGGGSAKFVAAWTLKAAAGSGSGS
SRRVQPTESIVRFPNITNLCPFGEVFNATRFASVYAWNRKRISNCVADYSVLYNSASFSTFKCYG
VSPTKLNDLCFTNVYADSFVIRGDEVRQIAPGQTGKIADYNYKLPDDFTGCVIAWNSNNLDSKVG
GNYNYLYRLFRKSNLKPFERDISTEIYQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVV
VLSFELLHAPATVCGPKKSTNLVKNKCVNFLEGGGDKTHTCPPCPAPELLGGPSVFLFPPKPKDT
LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLN
GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVE
WESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSP
GK
Detailed Description
The following are preferred embodiments of the present invention, and the present invention is not limited to the following preferred embodiments. It should be noted that modifications and improvements made on the basis of the inventive concept will be within the scope of the present invention for those skilled in the art. The reagents used were conventional products commercially available without the manufacturer's knowledge.
Examples
Example 1 design of fusion protein vaccine platforms
The immune cell targeting fusion protein vaccine is obtained by fusing an immune cell targeting molecule on an antigen. The immune cell targeting molecule may include the following four components:
A. an antibody or polypeptide capable of binding an immune cell surface protein;
B. cytokines capable of activating immune cells;
C. pan epitope (PADRE) capable of activating immune cells;
D. immunoglobulin Fc capable of binding immune cells.
The antigen may be combined with two, three or four of these four components in any arrangement to form a fusion protein. The representative forms are as follows:
in the form of homodimers, antigen-Fc is combined with immune cell targeting molecule A in an ordered arrangement of A-antigen-Fc as shown in FIG. 1. The antigen-Fc and the immune cell targeting molecule A can be arranged and combined in any form. The method specifically comprises the following steps: alpha PDL 1-anti-gen-Fc, CLEC9A binding peptide-anti-gen-Fc, alpha DEC 205-anti-gen-Fc, anti-gen-Fc-CLEC 9A binding peptide.
In the form of homodimers, antigen-Fc is combined with immune cell targeting molecule B in a B-antigen-Fc sequence arrangement, as shown in figure 2. The antigen-Fc and the immune cell targeting molecule B can be arranged and combined in any form. The method specifically comprises the following steps: IL 2-anti-gen-Fc, IL 12-anti-gen-Fc, IL 15-anti-gen-Fc, IL 21-anti-gen-Fc, CSF 2-anti-gen-Fc, anti-gen-Fc-CSF 2.
In the form of homodimers, antigen-Fc is combined with immune cell targeting molecules A and C in an A-C-antigen-Fc sequence, as shown in FIG. 3. The antigen-Fc and the immune cell targeting molecules A and C can be arranged and combined in any form. The method specifically comprises the following steps: alpha PDL 1-Pan-anti-Fc, CLEC 9-antibody peptide-Pan-anti-Fc, alpha DEC 205-Pan-anti-Fc.
In the form of homodimers, antigen-Fc is combined with immune cell targeting molecules B and C in a B-C-antigen-Fc sequential arrangement, as shown in FIG. 4. The antigen-Fc and the immune cell targeting molecules B and C can be arranged and combined in any form. The method specifically comprises the following steps: IL 2-Pan-anti-gen-Fc, IL 12-Pan-anti-gen-Fc, IL 15-Pan-anti-gen-Fc, IL 21-Pan-anti-gen-Fc, CSF 2-Pan-anti-gen-Fc, pan-anti-gen-Fc-CSF 2.
In the form of homodimers, antigen-Fc is combined with immune cell targeting molecules A and B in an A-antigen-Fc-B sequence, as shown in FIG. 5. The antigen-Fc and the immune cell targeting molecules A and B can be arranged and combined in any form. The method specifically comprises the following steps: alpha PDL 1-anti-Fc-IL 12, alpha PDL 1-anti-Fc-IL 21, alpha PDL 1-anti-Fc-CSF 2, CLEC9Abinding peptide-anti-Fc-IL 12, CLEC9Abinding peptide-anti-Fc-IL 21, CLEC9Abinding peptide-anti-Fc-CSF 2.
In the form of homodimers, antigen-Fc is combined with immune cell targeting molecules A, B and C in an A-C-antigen-Fc-B sequence as shown in FIG. 6. The antigen-Fc and the immune cell targeting molecules A, B and C can be arranged and combined in any form. The method specifically comprises the following steps: alpha PDL 1-Pan-anti-Fc-IL 12, alpha PDL 1-Pan-anti-Fc-IL 21, alpha PDL 1-Pan-anti-Fc-CSF 2, CLEC9A binding peptide-Pan-anti-Fc-IL 12, CLEC9A binding peptide-Pan-anti-Fc-IL 21, CLEC 9-Abinding peptide-Pan-anti-Fc-CSF 2, PDL1-CSF 2-Pan-anti-Fc.
In the form of heterodimers, antigen-Fc knob and immune cell targeting molecule A are aligned and combined in the A-antigen-Fc knob order, and antigen-Fc hole and immune cell targeting molecule B are aligned and combined in the B-antigen-Fc hole order, as shown in FIG. 7. The antigen-Fc knob and the immune cell targeting molecule A can be arranged and combined in any form. The antigen-Fc hole and the immune cell targeting molecule B can be arranged and combined in any form. The method specifically comprises the following steps: alpha PDL 1-anti-gen-Fc knob/CSF 2-anti-gen-Fc hole, CLEC9A binding peptide-anti-gen-Fc knob/CSF 2-anti-gen-Fc hole.
In the form of heterodimers, antigen-Fc knob and immune cell targeting molecules A and C are aligned in the A-C-antigen-Fc knob order, and antigen-Fc hole and immune cell targeting molecules B and C are aligned in the B-C-antigen-Fc hole order, as shown in FIG. 8. The antigen-Fc knob and the immune cell targeting molecules A and C can be arranged and combined in any form. The antigen-Fc hole and the immune cell targeting molecules B and C can be arranged and combined in any form. The method specifically comprises the following steps: alpha PDL 1-Pan-anti-Fc knob/CSF 2-Pan-anti-Fc hole, CLEC9A binding peptide-Pan-anti-Fc knob/CSF 2-Pan-anti-Fc hole. EXAMPLE 2 construction purification and production of vaccine platforms
In this example, the expression production of the vaccine platform is described using the novel coronavirus SARS-CoV-2 spike protein RBD protein homodimer format as an example.
1. Construction of vectors
And (3) using PEE6.4 as a carrier, and constructing antigen and immune cell targeting molecules on the carrier in a molecular cloning mode, so as to obtain the plasmid capable of expressing the fusion protein. Wherein KOZAK is a KOZAK sequence and SP (signal peptide) is a signal peptide.
(1)RBD:
PEE6.4-HindIII-KOZAK-Start-SP-XbaI-RBD-XhoI-6His-EcoRI
(2)RBD-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-RBD-XhoI-G3-Fc-EcoRI
(3)IL4-Pan-RBD-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-IL4-SfuI-(G4S) 3 -Pan-(GS) 3 -XbaI-RBD-XhoI-G 3 -Fc-EcoRI
(4)IL10-Pan-RBD-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-IL10-SfuI-(G4S) 3 -Pan-(GS) 3 -XbaI-RBD-XhoI-G 3 -Fc-EcoRI
(5)IL12-Pan-RBD-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-IL12-SfuI-(G4S) 3 -Pan-(GS) 3 -XbaI-RBD-XhoI-G 3 -Fc-EcoRI
(6)IL21-Pan-RBD-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-IL21-SfuI-(G4S) 3 -Pan-(GS) 3 -XbaI-RBD-XhoI-G 3 -Fc-EcoRI
(7)CSF2-Pan-RBD-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-CSF2-SfuI-(G4S) 3 -Pan-(GS) 3 -XbaI-RBD-XhoI-G 3 -Fc-EcoRI
(8)PD1-Pan-RBD-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-PD1-SfuI-(G4S) 3 -Pan-(GS) 3 -XbaI-RBD-XhoI-G 3 -Fc-EcoRI
(9)αPDL1-Pan-RBD-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-aPDL1-SfuI-(G4S) 3 -Pan-(GS) 3 -XbaI-RBD-XhoI-G 3 -Fc-EcoRI。
2. Transient transfection expression fusion proteins
(1) Cell resuscitation: freestyle 293F cells at 3X 10 7 The individual cells/ml concentration was frozen in Freestyle 293Expression Medium (10% DMSO). After removal from liquid nitrogen, rapid solubilization in a 37℃water bath, washing with SMM 293-TII medium, resuspending the cells in 50ml SMM 293-TII medium, 37℃8% CO 2 ,120rpm。
(2) 293F cells were resuspended in 100ml Freestyle 293 medium at a cell density of 3X 10 6 cells/ml. (3) 100. Mu.g of plasmid was diluted with 2ml Freestyle 293 medium and 400. Mu.g PEI was diluted with 2ml Freestyle 293 medium. Immediately thereafter, 2mL of the plasmid and 2mL of PEI were mixed and allowed to stand at room temperature for 5 minutes. (4) The plasmid/PEI mixture was added to the cell suspension and left at 37℃with 8% CO 2 Culturing in an incubator at 85 rpm.
(5) After 4 hours, 100ml of EX-CELL293 medium was added and the culture was continued with a speed of 120 rpm. (6) After 24 hours, 3.8mM of the cell proliferation inhibitor VPA was added, and the supernatant was collected for purification after 6 days of transfection.
3. Purification of fusion proteins
(1) The supernatant of the suspension cell culture broth was centrifuged, the precipitate was discarded, and the impurities were removed by filtration through a 0.45 μm filter.
(2) The appropriate amount of Protein A agarose beads was added to the column, and the column was washed and equilibrated with 10 volumes of distilled water and PBS, respectively.
(3) And loading by using a constant flow pump, wherein the flow rate is 0.2mL/min.
(4) After washing the column with PBS 10 times the column volume, eluting with elution buffer (0.1M glycine, pH 2.7), and neutralizing the eluted protein with appropriate amount of 1M Tris, pH 9.0.
(5) Protein was concentrated using a concentration column, the protein solution was replaced into the required buffer using a Zeba desalting column, the protein concentration was determined using Nano-500, and the protein expression yield was calculated.
4. Purity identification of fusion proteins
Proteins were loaded onto polyacrylamide gels with reducing loading buffer (R) and non-reducing loading buffer (N-R), respectively, and after electrophoresis the gels were stained with coomassie brilliant blue, then imaged with a gel imager to see the integrity and purity of the fusion proteins. The SDS-PAGE electrophoresis identification of the fusion protein is shown in FIG. 9. The fusion protein expression yields are shown in FIG. 10.
Example 3 SARS-CoV-2 spike protein RBD immune cell targeting fusion protein vaccine can elicit stronger antibody responses in mice than a pure RBD protein vaccine.
1. Material
C57BL/6 male mice (6-8 weeks purchased from Jiangsu Jiuyaokang biotechnology Co., ltd; horseradish peroxidase (HRP) -labeled goat anti-mouse IgG was purchased from Jiangsu kang as century biotechnology, inc; horseradish peroxidase (HRP) -labeled goat anti-mouse IgG1, igG2b, and IgG2c were purchased from Jiangsu kang as century biotechnology, inc; 96-well ELISA assay plates were purchased from Bioland; ELISA color development solutions were purchased from Shanghai Biyun biotechnology Co., ltd; ELISA stop solution was purchased from Beijing Soy Bao technology Co., ltd; the microplate reader Multiskan FC was purchased from Thermo Fisher Scientific.
2. Method of
(1) Immune cell targeting fusion protein vaccine the mice are immunized. After dilution of the fusion protein vaccine in PBS, each mouse was vaccinated with 0.5. Mu.g RBD protein or other fusion proteins of the same molar number, and each mouse was intramuscular injected with 50. Mu.l. Mice were immunized on days 0 and 21 using two immunization programs. Mouse serum was collected by cheek bleed at day 14 post immunization for antibody detection.
(2) ELISA detects RBD specific antibodies in serum. RBD (2. Mu.g/ml) coating was added to the Elisa plate in a 100. Mu.l per well system and coated overnight at 4 ℃. Blocking solution at 5% (PBS of 5% FBS) was blocked at 37℃for 1 hour. PBST was washed 3 times, serum samples were diluted in 10-fold gradients, 100 μl per well was added to the blocked ELISA plate and incubated for 1 hour at 37 ℃. PBST was washed 3 times, 100. Mu.l of enzyme-labeled secondary antibody (1:5000) was added to each well, and incubated at 37℃for 1 hour. Washing with PBST for 5 times, adding 100 μl/well of substrate TMB, incubating at room temperature in dark place, and waiting for substrate color development; the development was stopped by adding 50. Mu.l ELISA stop solution to each well, plate read by ELISA, OD450-620.
3. Results
Free RBD proteins are less immunogenic, and have increased immunogenicity when the Fc moiety is added on RBD, further increased immunogenicity when cytokines IL12 and Pan are added on RBD-Fc, further increased immunogenicity when cytokines IL21 and Pan are added on RBD-Fc, further increased immunogenicity when cytokines CSF2 and Pan are added on RBD-Fc, and further increased immunogenicity when alpha PDL1 antibodies and Pan capable of binding immune cell surface protein PDL1 are added on RBD-Fc, and the level of antibodies after secondary immunization is generally higher than that after primary immunization, as shown in fig. 11 (a). CSF2 or αpdl1 together with Pan induced higher levels of IgG1 representing Th2 immune response on RBD-Fc basis as shown in fig. 11 (b). IL12 or CSF2 together with Pan induced higher levels of IgG2b and IgG2c representing Th1 immune responses on RBD-Fc basis, as shown in FIGS. 11 (c) and 11 (d). The ratio of (IgG 2b+IgG2 c)/IgG 1 shows that IL12-Pan-RBD-Fc induces a relatively balanced Th1 and Th2 immune response, while RBD-Fc, CSF2-Pan-RBD-Fc and aPDL1-Pan-RBD-Fc induce a Th2 biased immune response, as shown in FIG. 11 (e).
EXAMPLE 4 construction, production and identification of novel coronavirus SARS-CoV-2 spike protein RBD-associated protein vaccine
1. Construction of vectors
And (3) using PEE6.4 as a carrier, and constructing antigen and immune cell targeting molecules on the carrier in a molecular cloning mode, so as to obtain the plasmid capable of expressing the fusion protein. Wherein KOZAK is a KOZAK sequence and SP (signal peptide) is a signal peptide.
(1)CSF2-RBD-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-CSF2-SfuI-(G4S)3-XbaI-RBD-XhoI-G3-Fc-EcoRI
(2)αPDL1-RBD-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-αPDL1-SfuI-(G4S)3-XbaI-RBD-XhoI-G3-Fc-EcoRI。
2. Transient transfection expression fusion proteins
(1) Cell resuscitation: freestyle 293F cells at 3X 10 7 The individual cells/ml concentration was frozen in Freestyle 293Expression Medium (10% DMSO). After removal from liquid nitrogen, rapid solubilization in a 37℃water bath, washing with SMM 293-TII medium, resuspending the cells in 50ml SMM 293-TII medium, 37℃8% CO 2 ,120rpm。
(2) 293F cells were resuspended in 100ml Freestyle 293 medium at a cell density of 3X 10 6 cells/ml. (3) 100. Mu.g of plasmid was diluted with 2ml Freestyle 293 medium and 400. Mu.g PEI was diluted with 2ml Freestyle 293 medium. Immediately thereafter, 2mL of the plasmid and 2mL of PEI were mixed and allowed to stand at room temperature for 5 minutes. (4) The plasmid/PEI mixture was added to the cell suspension and left at 37℃with 8% CO 2 Culturing in an incubator at 85 rpm.
(5) After 4 hours, 100ml of EX-CELL293 medium was added and the culture was continued with a speed of 120 rpm. (6) After 24 hours, 3.8mM of the cell proliferation inhibitor VPA was added, and the supernatant was collected for purification after 6 days of transfection.
3. Purification of fusion proteins
(1) The supernatant of the suspension cell culture broth was centrifuged, the precipitate was discarded, and the impurities were removed by filtration through a 0.45 μm filter.
(2) The appropriate amount of Protein A agarose beads was added to the column, and the column was washed and equilibrated with 10 volumes of distilled water and PBS, respectively.
(3) And loading by using a constant flow pump, wherein the flow rate is 0.2mL/min.
(4) After washing the column with PBS 10 times the column volume, eluting with elution buffer (0.1M glycine, pH 2.7), and neutralizing the eluted protein with appropriate amount of 1M Tris, pH 9.0.
(5) Protein was concentrated using a concentration column, the protein solution was replaced into the required buffer using a Zeba desalting column, the protein concentration was determined using Nano-500, and the protein expression yield was calculated.
4. Purity identification of fusion proteins
Proteins were loaded onto polyacrylamide gels with reducing loading buffer (R) and non-reducing loading buffer (N-R), respectively, and after electrophoresis the gels were stained with coomassie brilliant blue, then imaged with a gel imager to see the integrity and purity of the fusion proteins. The SDS-PAGE electrophoresis identification of the fusion proteins is shown in FIG. 12. The fusion protein expression yields are shown in FIG. 13.
Example 5 immune cell targeting novel coronavirus SARS-CoV-2 spike protein RBD associated protein vaccine can elicit stronger antibody response in mice than pure RBD protein vaccine
1. Material
C57BL/6 male mice (6-8 weeks purchased from Jiangsu Jiuyaokang biotechnology Co., ltd; horseradish peroxidase (HRP) -labeled goat anti-mouse IgG was purchased from Jiangsu kang as century biotechnology, inc; 96-well ELISA assay plates were purchased from Bioland; ELISA color development solutions were purchased from Shanghai Biyun biotechnology Co., ltd; ELISA stop solution was purchased from Beijing Soy Bao technology Co., ltd; the microplate reader Multiskan FC was purchased from Thermo Fisher Scientific.
2. Method of
(1) Immune cell targeting fusion protein vaccine the mice are immunized. After dilution of the fusion protein vaccine in PBS, each mouse was vaccinated with 0.5. Mu.g RBD protein or other fusion proteins of the same molar number, and each mouse was intramuscular injected with 50. Mu.l. Mice were immunized on days 0 and 21 using two immunization programs. Mouse serum was collected by cheek bleed at day 14 post immunization for antibody detection.
(2) ELISA detects RBD specific antibodies in serum. RBD (2. Mu.g/ml) coating was added to the Elisa plate in a 100. Mu.l per well system and coated overnight at 4 ℃. Blocking solution at 5% (PBS of 5% FBS) was blocked at 37℃for 1 hour. PBST was washed 3 times, serum samples were diluted in 10-fold gradients, 100 μl per well was added to the blocked ELISA plate and incubated for 1 hour at 37 ℃. PBST was washed 3 times, 100. Mu.l of enzyme-labeled secondary antibody (1:5000) was added to each well, and incubated at 37℃for 1 hour. Washing with PBST for 5 times, adding 100 μl/well of substrate TMB, incubating at room temperature in dark place, and waiting for substrate color development; the development was stopped by adding 50. Mu.l ELISA stop solution to each well, plate read by ELISA, OD450-620.
3. Results
The free RBD protein was less immunogenic, increased antibody titres when the Fc moiety was added on RBD, further increased antibody titres when the cytokine CSF2 was added on RBD-Fc, and further increased antibody titres when the alpha PDL1 antibody capable of binding to the immune cell surface protein PDL1 was added on RBD-Fc, as shown in fig. 14. These results demonstrate that CSF2-RBD-Fc is more immunogenic than RBD and RBD-Fc, and alpha PDL1-RBD-Fc is more immunogenic than RBD and RBD-Fc.
Example 6 construction, production and identification of Epstein-Barr virus (EBV) membrane protein gp 350-associated protein vaccine
EBV, also known as human herpesvirus 4, is one of the nine human herpesvirus types known in the herpesfamily and is also one of the most common viruses in humans. EBV is most well known as the cause of infectious mononucleosis, and is also associated with various non-malignant, precancerous and malignant epstein-barr virus-associated lymphoproliferative diseases, with about 200,000 cancer cases being considered attributable to EBV annually.
1. Construction of vectors
And (3) using PEE6.4 as a carrier, and constructing antigen and immune cell targeting molecules on the carrier in a molecular cloning mode, so as to obtain the plasmid capable of expressing the fusion protein. Wherein KOZAK is a KOZAK sequence and SP (signal peptide) is a signal peptide.
(1)gp350:
PEE6.4-HindIII-KOZAK-Start-SP-XbaI-gp350-XhoI-6His-EcoRI
(2)gp350-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-gp350-XhoI-G 3 -Fc-EcoRI
(3)CSF2-gp350-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-CSF2-SfuI-(G 4 S) 3 -XbaI-gp350-XhoI-G 3 -Fc-EcoRI
(4)αPDL1-gp350-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-αPDL1-SfuI-(G 4 S) 3 -XbaI-gp350-XhoI-G 3 -Fc-EcoRI
(5)CSF2-Pan-gp350-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-CSF2-SfuI-(G 4 S) 3 -Pan-(GS) 3 -XbaI-gp350-XhoI-G 3 -Fc-EcoRI
(6)αPDL1-Pan-gp350-Fc:
PEE6.4-HindIII-KOZAK-Start-SP-BsiwI-αPDL1-SfuI-(G 4 S) 3 -Pan-(GS) 3 -XbaI-gp350-XhoI-G 3 -Fc-EcoRI
2. Transient transfection expression fusion proteins
(1) Cell resuscitation: freestyle 293F cells at 3X 10 7 The individual cells/ml concentration was frozen in Freestyle 293Expression Medium (10% DMSO). After removal from liquid nitrogen, rapid solubilization in a 37℃water bath, washing with SMM 293-TII medium, resuspending the cells in 50ml SMM 293-TII medium, 37℃8% CO 2 ,120rpm。
(2) 293F cells were resuspended in 100ml Freestyle 293 medium at a cell density of 3X 10 6 cells/ml. (3) 100. Mu.g of plasmid was diluted with 2ml Freestyle 293 medium and 400. Mu.g PEI was diluted with 2ml Freestyle 293 medium. Immediately thereafter, 2mL of the plasmid and 2mL of PEI were mixed and allowed to stand at room temperature for 5 minutes. (4) The plasmid/PEI mixture was added to the cell suspension and left at 37℃with 8% CO 2 Culturing in an incubator at 85 rpm.
(5) After 4 hours, 100ml of EX-CELL293 medium was added and the culture was continued with a speed of 120 rpm. (6) After 24 hours, 3.8mM of the cell proliferation inhibitor VPA was added, and the supernatant was collected for purification after 6 days of transfection.
3. Purification of fusion proteins
(1) The supernatant of the suspension cell culture broth was centrifuged, the precipitate was discarded, and the impurities were removed by filtration through a 0.45 μm filter.
(2) The appropriate amount of Protein A agarose beads was added to the column, and the column was washed and equilibrated with 10 volumes of distilled water and PBS, respectively.
(3) And loading by using a constant flow pump, wherein the flow rate is 0.2mL/min.
(4) After washing the column with PBS 10 times the column volume, eluting with elution buffer (0.1M glycine, pH 2.7), and neutralizing the eluted protein with appropriate amount of 1M Tris, pH 9.0.
(5) Protein was concentrated using a concentration column, the protein solution was replaced into the required buffer using a Zeba desalting column, the protein concentration was determined using Nano-500, and the protein expression yield was calculated.
4. Purity identification of fusion proteins
Proteins were loaded onto polyacrylamide gels with reducing loading buffer (R) and non-reducing loading buffer (N-R), respectively, and after electrophoresis the gels were stained with coomassie brilliant blue, then imaged with a gel imager to see the integrity and purity of the fusion proteins. The SDS-PAGE electrophoresis identification of the fusion proteins is shown in FIG. 15. The fusion protein expression yields are shown in FIG. 16.
Example 7 immune cell-targeted EBV Membrane protein gp 350-associated protein vaccine can elicit stronger antibody responses in mice than pure gp350 protein vaccine
1. Material
C57BL/6 male mice (6-8 weeks purchased from Jiangsu Jiuyaokang biotechnology Co., ltd; horseradish peroxidase (HRP) -labeled goat anti-mouse IgG was purchased from Jiangsu kang as century biotechnology, inc; 96-well ELISA assay plates were purchased from Bioland; ELISA color development solutions were purchased from Shanghai Biyun biotechnology Co., ltd; ELISA stop solution was purchased from Beijing Soy Bao technology Co., ltd; the microplate reader Multiskan FC was purchased from Thermo Fisher Scientific.
2. Method of
(1) Immune cell targeting fusion protein vaccine the mice are immunized. After dilution of the fusion protein vaccine in PBS, each mouse was vaccinated with 0.5. Mu.g gp350 protein or other fusion protein in the same molar number, and each mouse was intramuscular injected with 50. Mu.l. Mice were immunized on days 0 and 21 using two immunization programs. Mouse serum was collected by cheek bleed at day 14 post immunization for antibody detection.
(2) ELISA detects gp 350-specific antibodies in serum. Gp350 (2. Mu.g/ml) coating was added to the Elisa plate in a 100. Mu.l per well system and coated overnight at 4 ℃. Blocking solution at 5% (PBS of 5% FBS) was blocked at 37℃for 1 hour. PBST was washed 3 times, serum samples were diluted in 10-fold gradients, 100 μl per well was added to the blocked ELISA plate and incubated for 1 hour at 37 ℃. PBST was washed 3 times, 100. Mu.l of enzyme-labeled secondary antibody (1:5000) was added to each well, and incubated at 37℃for 1 hour. Washing with PBST for 5 times, adding 100 μl/well of substrate TMB, incubating at room temperature in dark place, and waiting for substrate color development; the development was stopped by adding 50. Mu.l ELISA stop solution to each well, plate read by ELISA, OD450-620.
3. Results
The free gp350 protein vaccine was less immunogenic, did not increase antibody titer when the Fc portion was added on gp350, increased antibody titer when the cytokine CSF2 was added on gp350-Fc, increased antibody titer when the alpha PDL1 antibody capable of binding to the immune cell surface protein PDL1 was added on gp350-Fc, increased antibody titer when the cytokines CSF2 and Pan were added on gp350-Fc, and increased antibody titer when the alpha PDL1 antibody capable of binding to the immune cell surface protein PDL1 and Pan were added on gp350-Fc, as shown in fig. 17. These results demonstrate that CSF2-gp350-Fc is more immunogenic than gp350 and gp350-Fc, alpha PDL1-gp350-Fc is more immunogenic than gp350 and gp350-Fc, CSF2-Pan-gp350-Fc is more immunogenic than gp350 and gp350-Fc, alpha PDL1-Pan-gp350-Fc is more immunogenic than gp350 and gp350-Fc.

Claims (16)

1. A fusion protein comprising an antigen domain and an immune cell targeting domain, the immune cell targeting domain being one or more domains selected from the group consisting of:
domain a: an antibody or polypeptide or active fragment thereof capable of binding an immune cell surface protein;
domain B: a cytokine or active fragment thereof capable of activating an immune cell;
domain C: a Pan epitope (PADRE) or active fragment thereof capable of activating immune cells;
domain D: immunoglobulin Fc capable of binding immune cells.
2. The fusion protein of claim 1, comprising an antigen domain and domain a; fusion proteins of antigen domain with domain a and domain B; fusion proteins of antigen domain with domain a, domain B and domain C; fusion proteins of antigen domains with domain a, domain B, domain C and domain D; fusion proteins of antigen domain with domain a and domain C; fusion proteins of antigen domain with domain a and domain D; fusion proteins of antigen domain with domain a, domain C and domain D; fusion proteins of antigen domain and domain B; fusion proteins of antigen domain with domain B and domain C; fusion proteins of antigen domain with domain B and domain D; fusion proteins of antigen domain with domain B, domain C and domain D; fusion proteins of antigen domain and domain C; fusion proteins of antigen domain with domain C and domain D; fusion proteins of antigen domain and domain C; fusion proteins of antigen domain with domain C and domain D; or a fusion protein of an antigen domain and domain D;
The domain a is selected from antibodies against CD274 (PDL 1), PDCD1LG2 (PDL 2), CLEC9A, LY75 (DEC 205), CD40, TNFSF9 (4-1 BB-L) and/or TNFSF4 (OX 4 OL), or active fragments of ligands thereof;
said domain B is selected from the group consisting of an Interleukin (IL), a Colony stimulating factor (Colony-stimulating factor, CSF), or an active fragment thereof, preferably said interleukin is selected from the group consisting of IL2, IL12, IL15 and/or IL21, or an active fragment thereof; preferably, the colony stimulating factor is selected from CSF1, CSF2 and/or CSF3, or an active fragment thereof;
preferably, said domain C has an amino acid sequence as shown in akfvaaawtlkaaa;
preferably, the domain D is selected from IgG, igM, igA, igE or Fc of IgD or a mutant thereof;
preferably, the Fc is a modified Fc, more preferably, the Fc has an Fc knob modification and an Fc Hole modification;
preferably, the fusion protein is any arrangement of antigen domains with domain a and/or domain B and/or domain C and/or domain D; or alternatively, the process may be performed,
the antigen domain and/or domain a and/or domain B and/or domain C and/or domain D in the fusion protein are C-terminal or N-terminal with respect to each other.
3. The fusion protein of any one of claims 1-2, which is a homodimer or a heterodimer; preferably, the fusion protein comprises an antigen domain and domain D, the fusion protein constituting a homodimer or a heterodimer in a similar antibody format;
Preferably, the fusion protein consists of a homodimer or a heterodimer of disulfide bonds of two Fc domains;
preferably, the fusion protein comprises a first polypeptide chain comprising an antigen domain and domain D and a second polypeptide chain comprising an antigen domain and domain D; or alternatively
Preferably, the first polypeptide chain further comprises domain B and/or domain C, and the second polypeptide chain further comprises domain B and/or domain C; or alternatively, the process may be performed,
preferably, the first polypeptide chain further comprises domain B and the second polypeptide chain further comprises domain C, thereby constituting a heterodimer; or alternatively
The first polypeptide chain comprises from C-terminus to N-terminus domain D, an antigen domain, domain a, and the second polypeptide chain comprises from C-terminus to N-terminus domain D, an antigen domain, domain a, thereby constituting a homodimer, and vice versa; or alternatively
The first polypeptide chain comprises from C-terminus to N-terminus domain D, an antigen domain, domain B, and the second polypeptide chain comprises from C-terminus to N-terminus domain D, an antigen domain, domain B, thereby constituting a homodimer, and vice versa; or alternatively
The first polypeptide chain comprises, from C-terminus to N-terminus, domain D, an antigen domain, domain C, domain a, and the second polypeptide chain comprises, from C-terminus to N-terminus, domain D, an antigen domain, domain C, domain a, thereby constituting a homodimer, and vice versa; or alternatively
The first polypeptide chain comprises from C-terminus to N-terminus domain D, antigen domain, domain C, domain B, and the second polypeptide chain comprises from C-terminus to N-terminus domain D, antigen domain, domain C, domain B, thereby constituting a homodimer, and vice versa; or alternatively
The first polypeptide chain comprises a domain B, a domain D, an antigen domain and a domain A from the C end to the N end, and the second polypeptide chain comprises a domain B, a domain D, an antigen domain and a domain A from the C end to the N end, so that a homodimer is formed; or alternatively
The first polypeptide chain comprises, from C-terminus to N-terminus, domain B, domain D, an antigen domain, domain C, domain a, and the second polypeptide chain comprises, from C-terminus to N-terminus, domain B, domain D, an antigen domain, domain C, domain a, thereby constituting a homodimer, and vice versa; or alternatively
The first polypeptide chain comprises from C-terminus to N-terminus domain D, an antigen domain, domain a, and the second polypeptide chain comprises from C-terminus to N-terminus domain D, an antigen domain, domain C, domain B, thereby constituting a heterodimer, and vice versa; or alternatively
The first polypeptide chain comprises from C-terminus to N-terminus domain D, antigen domain, domain C, domain a, and the second polypeptide chain comprises from C-terminus to N-terminus domain D, antigen domain, domain C, domain B, thereby constituting a heterodimer, and vice versa;
Preferably, the antigen domain and the immune cell targeting domain A, domain B, domain C, domain D can be arranged and combined in any form.
4. A fusion protein according to any one of claims 1-3, wherein the fusion protein antigen is linked to the immune cell targeting molecule and/or the immune cell targeting molecule by a linker fragment;
preferably, the linker fragment is a flexible linker fragment, a rigid linker fragment or an in vivo cleavage linker fragment, more preferably the amino acid sequence of the flexible linker fragment is (G) N ,(GS) N ,(GGS) N ,(GGGS) N Or (GGGGS) N
5. The fusion protein of any one of claims 1-4, having a structure selected from the group consisting of:
αPDL1-Antigen-Fc;
CLEC9A bindingpeptide-Antigen-Fc;
αDEC205-Antigen-Fc;
Antigen-Fc-CLEC9A bindingpeptide;
αPDL1-linker-Antigen-linker-Fc;
αPDL1-(GGGGS) 3 -Antigen-(G) 3 -Fc;
CLEC9A bindingpeptide-linker-Antigen-linker-Fc;
CLEC9A bindingpeptide-(GGGGS) 3 -Antigen-(G) 3 -Fc;
αDEC205-linker-Antigen-linker-Fc;
αDEC205-(GGGGS) 3 -Antigen-(G) 3 -Fc;
Antigen-linker-Fc-linker-CLEC9A bindingpeptide;
Antigen-(G) 3 -Fc-(GS) 3 -CLEC9A binding peptide;
IL2-Antigen-Fc;
IL12-Antigen-Fc;
IL15-Antigen-Fc;
IL21-Antigen-Fc;
CSF2-Antigen-Fc;
Antigen-Fc-CSF2;
IL2-linker-Antigen-linker-Fc;
IL2-(GGGGS) 3 -Antigen-(G) 3 -Fc;
IL12-linker-Antigen-linker-Fc;
IL12-(GGGGS) 3 -Antigen-(G) 3 -Fc;
IL15-linker-Antigen-linker-Fc;
IL15-(GGGGS) 3 -Antigen-(G) 3 -Fc;
IL21-linker-Antigen-linker-Fc;
IL21-(GGGGS) 3 -Antigen-(G) 3 -Fc;
CSF2-linker-Antigen-linker-Fc;
CSF2-(GGGGS) 3 -Antigen-(G) 3 -Fc;
Antigen-linker-Fc-linker-CSF2;
Antigen-(GGGGS) 3 -Fc-(G) 3 -CSF2;
αPDL1-Pan-Antigen-Fc;
CLEC9A binding peptide-Pan-Antigen-Fc;
αDEC205-Pan-Antigen-Fc;
αPDL1-linker-Pan-Antigen-linker-Fc;
αPDL1-(GGGGS) 3 -Pan-Antigen-(G) 3 -Fc;
CLEC9A binding peptide-linker-Pan-Antigen-linker-Fc;
CLEC9A binding peptide-(GGGGS) 3 -Pan-Antigen-(G) 3 -Fc;
αDEC205-linker-Pan-Antigen-linker-Fc;
αDEC205-(GGGGS) 3 -Pan-Antigen-(G) 3 -Fc;IL2-Pan-Antigen-Fc;IL12-Pan-Antigen-Fc;
IL15-Pan-Antigen-Fc;
IL21-Pan-Antigen-Fc;
CSF2-Pan-Antigen-Fc;
Pan-Antigen-Fc-CSF2;
IL2-linker-Pan-linker-Antigen-(G) 3 -Fc;
IL2-(GGGGS) 3 -Pan-(GS) 3 -Antigen-linker-Fc;
IL12-linker-Pan-linker-Antigen-linker-Fc;
IL12-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc;
IL15-linker-Pan-linker-Antigen-linker-Fc;
IL15-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc;
IL21-linker-Pan-linker-Antigen-linker-Fc;
IL21-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc;
CSF2-linker-Pan-linker-Antigen-linker-Fc;
CSF2-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc;
Pan-linker-Antigen-linker-Fc-linker-CSF2;
Pan-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -CSF2;
αPDL1-Antigen-Fc-IL12;
αPDL1-Antigen-Fc-IL21;
αPDL1-Antigen-Fc-CSF2;
CLEC9A binding peptide-Antigen-Fc-IL12;
CLEC9A binding peptide-Antigen-Fc-IL21;
CLEC9A binding peptide-Antigen-Fc-CSF2;
αPDL1-linker-Antigen-linker-Fc-linker-IL12;
αPDL1-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL12;
αPDL1-linker-Antigen-linker-Fc-linker-IL21;
αPDL1-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL21;
αPDL1-linker-Antigen-linker-Fc-linker-CSF2;
αPDL1-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -CSF2;
CLEC9A binding peptide-linker-Antigen-linker-Fc-linker-IL12;CLEC9A binding peptide-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL12;
CLEC9A binding peptide-linker-Antigen-linker-Fc-linker-IL21;
CLEC9A binding peptide-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL21;
CLEC9A binding peptide-linker-Antigen-linker-Fc-linker-CSF2;
CLEC9A binding peptide-(GGGGS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -CSF2;
αPDL1-Pan-Antigen-Fc-IL12;
αPDL1-Pan-Antigen-Fc-IL21;
αPDL1-Pan-Antigen-Fc-CSF2;
CLEC9A binding peptide-Pan-Antigen-Fc-IL12;
CLEC9A binding peptide-Pan-Antigen-Fc-IL21;
CLEC9A binding peptide-Pan-Antigen-Fc-CSF2;
αPDL1-CSF2-Pan-Antigen-Fc;
αPDL1-linker-Pan-linker-Antigen-linker-Fc-linker-IL12;
αPDL1-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL12;
αPDL1-linker-Pan-linker-Antigen-linker-Fc-linker-IL21;
αPDL1-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL21;
αPDL1-linker-Pan-linker-Antigen-linker-Fc-linker-CSF2;
αPDL1-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -CSF2;
CLEC9A binding peptide-linker-Pan-linker-Antigen-linker-Fc-linker-IL12;
CLEC9A binding peptide-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL12;CLEC9A binding peptide-linker-Pan-linker-Antigen-linker-Fc-linker-IL21;
CLEC9A binding peptide-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -IL21;CLEC9A binding peptide-linker-Pan-linker-Antigen-linker-Fc-linker-CSF2;
CLEC9A binding peptide-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc-(GS) 3 -CSF2;αPDL1-linker-CSF2-linker-Pan-linker-Antigen-linker-Fc;
αPDL1-(GGGGS) 3 -CSF2-(GS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc;
αPDL1-Antigen-Fc knob/CSF2-Antigen-Fc hole;
CLEC9A binding peptide-Antigen-Fc knob/CSF2-Antigen-Fc hole;
αPDL1-linker-Antigen-linker-Fc knob/CSF2-linker-Antigen-linker-Fc hole;αPDL1-(GGGGS) 3 -Antigen-(G) 3 -Fc knob/
CSF2-(GGGGS) 3 -Antigen-(G) 3 -Fc hole;
CLEC9A binding peptide-linker-Antigen-linker-Fc knob/
CSF2-linker-Antigen-linker-Fc hole;
CLEC9A binding peptide-(GGGGS) 3 -Antigen-(G) 3 -Fc knob/
CSF2-(GGGGS) 3 -Antigen-(G) 3 -Fc hole;
αPDL1-Pan-Antigen-Fc knob/CSF2-Pan-Antigen-Fc hole;
CLEC9A binding peptide-Pan-Antigen-Fc knob/CSF2-Pan-Antigen-Fc hole;
αPDL1-linker-Pan-linker-Antigen-linker-Fc knob/
CSF2-linker-Pan-linker-Antigen-linker-Fc hole;
αPDL1-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc knob/
CSF2-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc hole;
αPDL1-linker-Pan-linker-Antigen-linker-Fc knob/
CLEC9A binding peptide-linker-Pan-linker-Antigen-linker-Fc knob/
CSF2-linker-Pan-linker-Antigen-linker-Fc hole;
CLEC9A binding peptide-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc knob/
CSF2-(GGGGS) 3 -Pan-(GS) 3 -Antigen-(G) 3 -Fc hole。
6. the fusion protein of any one of claims 1-5, wherein the antigen domain is an immunogenic protein or immunogenic fragment thereof capable of inducing an immune response against a pathogenic microorganism;
preferably the pathogenic microorganism is SARS-Cov-2, SARS, cytomegalovirus CMV, herpes virus, respiratory syncytial virus RSV, influenza virus, ebola virus, epstein-Barr virus EBV, dengue virus, zike virus, HIV virus, rabies virus, plasmodium gametophyte, herpes zoster virus HZV, hepatitis B virus HBV, hepatitis C virus HCV, hepatitis D virus HDV, HPV, mycobacterium tuberculosis, or helicobacter pylori.
7. The fusion protein of claim 6, wherein the antigen domain is the S protein of SARS-Cov-2 or a fragment thereof, preferably the S protein is a pre-fusion stable S protein, more preferably the pre-fusion stable S protein comprises a biproline (S2P) mutation or a hexapro (S6P) mutation; or alternatively
The antigen is RBD domain of S protein of SARS-Cov-2.
8. The fusion protein of any one of claims 1-5, wherein the antigen domain is an immunogenic protein, or an immunogenic fragment thereof, capable of inducing an immune response against a cancer cell;
preferably, the antigen domain is a tumor antigen or immunogenic fragment thereof selected from the group consisting of: melanA/MART1, cancer-germ line antigen, gp100, tyrosinase, CEA, PSA, her-2/neu, survivin, telomerase, or an immunogenic fragment thereof;
preferably, the cancer may be prostate cancer, non-small cell lung cancer, renal cell carcinoma, brain cancer, melanoma, acute myelogenous leukemia, pancreatic cancer, colorectal cancer, head and neck squamous cell carcinoma, cutaneous squamous cell carcinoma, adenoid cystic carcinoma, glioblastoma, breast cancer, mesothelioma, ovarian cancer, glioma, bladder cancer, liver cancer, bone marrow cancer, gastric cancer, thyroid cancer, lymphoma, cervical cancer, endometrial cancer, laryngeal cancer, acute lymphoblastic leukemia.
9. A nucleic acid molecule encoding the fusion protein of any one of claims 1-8.
10. A vector expressing the fusion protein of any one of claims 1-9 or comprising the nucleic acid molecule of claim 9.
11. The vector of claim 10, which is a plasmid vector, an adenovirus vector or a lentiviral vector.
12. A host cell for expressing a fusion protein according to any one of claims 1 to 8 or comprising a nucleic acid molecule according to claim 9 or a vector according to claim 10 or 11,
preferably, the host cell is a chinese hamster ovary cell (CHO cell), a baby hamster kidney cell (BHK cell), a COS cell, a mouse NSO thymoma cell, a mouse myeloma cell (SP 2/0 cell), a human embryonic kidney cell HEK293 cell.
13. A composition comprising the fusion protein of any one of claims 1-8 or the nucleic acid molecule of claim 9, or the vector of claim 10 or 11, or the host cell of claim 12.
14. The composition of claim 13, which does not comprise a pharmaceutically acceptable adjuvant; or the composition further comprises a pharmaceutically acceptable adjuvant, preferably including but not limited to an aluminum adjuvant or CpG adjuvant, preferably the aluminum adjuvant is aluminum hydroxide, aluminum phosphate or aluminum sulfate.
15. A method for preventing or treating infection or tumor with a pathogenic microorganism, said method comprising the step of administering to a subject a fusion protein according to any one of claims 1-8 or a nucleic acid molecule according to claim 9, or a vector according to claim 10 or 11, a host cell according to claim 12, or a composition according to claim 13 or 14,
preferably, the subject is a human or animal,
preferably, the animal is a cow, sheep, cat, dog, horse, rabbit, monkey, mouse, rat, alpaca or camel;
preferably, the subject is a immunocompromised human or animal;
preferably, the subject suffers from chronic lung disease, chronic obstructive pulmonary disease or asthma;
preferably, the patient has a basal disease selected from heart disease, diabetes or lung disease.
16. Use of a fusion protein according to any one of claims 1 to 8 or a nucleic acid molecule according to claim 9, or a vector according to claim 10 or 11, a host cell according to claim 12, or a composition according to claim 13 or 14 for the preparation of a medicament or kit for preventing or treating a subject associated with an infection or tumor by a pathogenic microorganism,
the subject is a human being or an animal,
preferably, the animal is a cow, sheep, cat, dog, horse, rabbit, monkey, mouse, rat, alpaca or camel;
Preferably, the subject is a immunocompromised human or animal;
preferably, the subject suffers from chronic lung disease, chronic obstructive pulmonary disease or asthma;
preferably, the patient has a basal disease selected from heart disease, diabetes or lung disease.
CN202211743150.2A 2021-12-31 2022-12-30 Fusion protein vaccine Pending CN116375881A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111683236 2021-12-31
CN2021116832366 2021-12-31

Publications (1)

Publication Number Publication Date
CN116375881A true CN116375881A (en) 2023-07-04

Family

ID=86966224

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211743150.2A Pending CN116375881A (en) 2021-12-31 2022-12-30 Fusion protein vaccine

Country Status (2)

Country Link
CN (1) CN116375881A (en)
WO (1) WO2023125976A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117003895A (en) * 2023-08-09 2023-11-07 成都新诺明生物科技有限公司 gE fusion protein containing IL2, fc and PADRE, and preparation method and application thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SK782002A3 (en) * 1999-07-21 2003-08-05 Lexigen Pharm Corp FC fusion proteins for enhancing the immunogenicity of protein and peptide antigens
AU2005251738A1 (en) * 2004-06-02 2005-12-22 New York Blood Center, Inc. SARS vaccines and methods to produce highly potent antibodies
CN102884194B (en) * 2010-03-11 2015-07-22 健康研究股份有限公司 Methods and compositions containing Fc fusiong proteins for enhancing immune responses
CN107286245B (en) * 2016-04-11 2021-10-08 北京普纳生物科技有限公司 PD-L1 and PD-L2 recombinant proteins and uses thereof

Also Published As

Publication number Publication date
WO2023125976A1 (en) 2023-07-06

Similar Documents

Publication Publication Date Title
WO2021185310A1 (en) Mvsv virus vector and virus vector vaccine thereof, and novel coronavirus pneumonia vaccine based on mvsv mediation
Sun et al. Interferon-armed RBD dimer enhances the immunogenicity of RBD for sterilizing immunity against SARS-CoV-2
CN104039833B (en) For hpv vaccine
WO2021239147A1 (en) β-CORONAVIRUS ANTIGEN, β-CORONAVIRUS BIVALENT VACCINE, PREPARATION METHODS THEREFOR, AND APPLICATIONS THEREOF
KR101912330B1 (en) Influenza nucleic acid molecules and vaccines made therefrom
ES2858315T3 (en) Stabilized soluble RSV prefusion protein F for use in the prophylaxis of RSV infection
Stepanova et al. Flagellin-fused protein targeting M2e and HA2 induces potent humoral and T-cell responses and protects mice against various influenza viruses a subtypes
TWI605124B (en) Novel baculovirus vectors and methods of ?use
Fan et al. Targeting the HA2 subunit of influenza A virus hemagglutinin via CD40L provides universal protection against diverse subtypes
US8981057B2 (en) B-cell stimulating fusion proteins of an antigen with BAFF or APRIL
EP3758747A1 (en) Self-asssembling nanostructure vaccines
CN111358953A (en) Vaccine vector for efficiently inducing humoral immune response of organism, preparation method and application thereof
JP2013532971A5 (en)
WO2022002180A1 (en) Construction and application of fusion protein vaccine platform
CN116375881A (en) Fusion protein vaccine
WO2022135563A1 (en) Method for simultaneously inducing immune response against multiple viruses
Dong et al. Nanoparticles of conformation-stabilized canine distemper virus hemagglutinin are highly immunogenic and induce robust immunity
CN115137812A (en) Construction and application of fusion protein vaccine platform
WO2021253172A1 (en) Method for inducing anti-novel coronavirus neutralizing antibody using receptor recognition domain
Tang et al. Recombinant adenoviruses displaying matrix 2 ectodomain epitopes on their fiber proteins as universal influenza vaccines
Montgomerie et al. Incorporation of SARS-CoV-2 spike NTD to RBD protein vaccine improves immunity against viral variants
WO2022112790A1 (en) Virus-like particles and methods of production thereof
CN116376942A (en) mRNA vaccine
Kawai et al. Intranasal immunization with an RBD-hemagglutinin fusion protein harnesses preexisting immunity to enhance antigen-specific responses
CN113801206A (en) Method for inducing anti-neocoronavirus neutralizing antibody by using receptor recognition domain

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