WO2022216949A1 - Fusion protein for antigen presentation - Google Patents

Fusion protein for antigen presentation Download PDF

Info

Publication number
WO2022216949A1
WO2022216949A1 PCT/US2022/023864 US2022023864W WO2022216949A1 WO 2022216949 A1 WO2022216949 A1 WO 2022216949A1 US 2022023864 W US2022023864 W US 2022023864W WO 2022216949 A1 WO2022216949 A1 WO 2022216949A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
seq
cell
exosome
membrane preparation
Prior art date
Application number
PCT/US2022/023864
Other languages
French (fr)
Inventor
James Robert EDGAR
John Trowsdale
Lesley Young
Mathew M.S. LO
Hannah JACKSON
Original Assignee
Exosis, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Exosis, Inc. filed Critical Exosis, Inc.
Publication of WO2022216949A1 publication Critical patent/WO2022216949A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • C07K14/08RNA viruses
    • C07K14/165Coronaviridae, e.g. avian infectious bronchitis virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/215Coronaviridae, e.g. avian infectious bronchitis virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/70Multivalent vaccine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • C07K2319/43Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation containing a FLAG-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
    • 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

Definitions

  • the present invention relates to a fusion protein comprising an exosomal protein and one or more immunogenic protein, wherein the exosomal protein is a tetraspanin protein.
  • the present invention also relates to an exosome comprising said fusion protein.
  • the present invention relates to a vaccine composition comprising an exosome of the invention.
  • the present invention also relates to a nucleic acid molecule encoding said fusion protein, and an expression vector comprising said nucleic acid molecule.
  • the present invention further relates to a cell comprising said fusion protein, exosome, nucleic acid molecule, and/or expression vector.
  • the invention also relates to the exosome and the vaccine composition for use as a medicament.
  • the invention also relates to the exosome and and/or the vaccine composition for use in the prevention or amelioration of an infection in a subject.
  • the invention relates to a method of preventing or ameliorating an infection in a subject, the method comprising providing the subject in need thereof with a therapeutically effective amount of an exosome and/or a vaccine composition of the invention.
  • Background SARS-CoV-2 is a recently emerging coronavirus responsible for the Covid-19 epidemic. The virus causes significant pathology, in some cases death, in a fraction of patients, in particular those with associated comorbidities and has an unprecedented gradient of severity in relation to older age.
  • Severity may extend from extremely mild (or even asymptomatic) to life-threatening.
  • the virus binds to the ACE2 receptor in cells of the airways, including pneumocytes and alveolar macrophages. It infects and destroys alveoli in the lungs. Mild cases may make neutralising antibodies that stop the virus early in its development in the individual.
  • One way it becomes problematic is by interfering with the production of interferon. Disease may be due to the body’s response to the pathogen, specifically over-reaction of immunity. If inflammation and tissue damage become too severe the infected person may die or end up with significant organ damage, not only to the lungs. Multiple organs may be affected by mechanisms such as excessive clotting.
  • a fusion protein comprising: i) an exosomal protein, wherein the exosomal protein is a tetraspanin protein; and ii) one or more immunogenic protein.
  • the tetraspanin is selected from the group consisting of CD63, CD9, and CD81 or a fragment, or variant thereof.
  • the one or more immunogenic protein is capable of eliciting an immune response against a SARS-CoV-2 protein.
  • the SARS-CoV-2 protein is selected from the group consisting of spike protein (S), nucleoprotein (N), membrane protein (M), envelope protein (E), an open reading frame (ORF) 1ab, ORF 3a protein, ORF 3b protein, ORF 6 protein, ORF 7a protein, ORF 7b protein, ORF 8 protein, ORF 9b protein, ORF 9c protein, and ORF 10 protein, optionally wherein when the SARS-CoV-2 protein is spike protein (S) it is the extracellular domain of spike protein (S).
  • the one or more immunogenic protein is capable of eliciting a B- cell and a T-cell immune response. In an embodiment, the one or more immunogenic protein is capable of eliciting a T- cell immune response. In an embodiment, the protein comprises a first one or more immunogenic protein capable of eliciting a B-cell and a T-cell immune response and a second one or more immunogenic protein capable of eliciting a T-cell immune response. In an embodiment, the first one or more immunogenic protein capable of eliciting a B- cell and T-cell immune response is incorporated into a luminal loop of the exosomal protein.
  • the second one or more immunogenic protein capable of eliciting a T-cell immune response is incorporated near or at the N-terminus of the exosomal protein, and/or is incorporated into an intravesicular loop of the exosomal protein.
  • the first one or more immunogenic protein is capable of eliciting a B-cell and T-cell immune response against the SARS-CoV-2 spike protein (S), optionally against the extracellular domain of spike protein (S).
  • the second one or more immunogenic protein is capable of eliciting a T-cell immune response against the SARS-CoV-2 protein selected from the group consisting of nucleoprotein (N), membrane protein (M), envelope protein (E), ORF 1ab polyprotein, ORF 3a protein, ORF 3b protein, ORF 6 protein, ORF 7a protein, ORF 7b protein, ORF 8 protein, ORF 9b protein, ORF 9c protein, and ORF 10 protein, optionally wherein when the SARS-CoV-2 protein is spike protein (S) it is the extracellular domain of spike protein (S).
  • SARS-CoV-2 protein selected from the group consisting of nucleoprotein (N), membrane protein (M), envelope protein (E), ORF 1ab polyprotein, ORF 3a protein, ORF 3b protein, ORF 6 protein, ORF 7a protein, ORF 7b protein, ORF 8 protein, ORF 9b protein, ORF 9c protein, and ORF 10 protein, optionally wherein when the SARS-CoV-2 protein is spike
  • the first one or more immunogenic protein capable of eliciting a B- cell and a T-cell immune response is incorporated into the first and/or second luminal loop of the tetraspanin, optionally wherein the tetraspanin is CD63.
  • the second one or more immunogenic protein capable of eliciting a T-cell immune response is incorporated near or at the N-terminus of the tetraspanin, optionally wherein the tetraspanin is CD63.
  • the one or more immunogenic protein shares at least 75% identity with a SARS-CoV-2 protein or protein variant, or a fragment thereof.
  • the fragment when the SARS-CoV-2 protein is spike protein (S), the fragment is the receptor binding domain (RBD) (SEQ ID NO: 4).
  • the SARS-CoV-2 protein is membrane protein (M) the fragment is according to SEQ ID NO: 57 or 72.
  • the SARS-CoV-2 protein is nucleoprotein (N) the fragment is according to SEQ ID NO: 56 or SEQ ID NO: 71.
  • the SARS-CoV-2 protein is selected from the group consisting of the extracellular domain of spike protein (S) (SEQ ID NO: 1), nucleoprotein (N) (SEQ ID NO: 2), membrane protein (M) (SEQ ID NO: 3), envelope protein (E) (SEQ ID NO: 46), ORF 3a protein (SEQ ID NO: 47), ORF 3b protein (SEQ ID NO: 48), ORF 6 protein (SEQ ID NO: 49), ORF 7a protein (SEQ ID NO: 50), ORF 7b protein (SEQ ID NO: 51), ORF 8 protein (SEQ ID NO: 52), ORF 9b protein (SEQ ID NO: 53), ORF 9c protein (SEQ ID NO: 54), ORF 10 protein (SEQ ID NO: 55), and ORF1ab polyprotein (SEQ ID NO: 73).
  • the fusion protein shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO 65, SEQ ID NO: 66, SEQ ID NO: 68, or SEQ ID NO:69.
  • the fusion protein shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% with SEQ ID NO: 5 or SEQ ID NO: 6.
  • the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 5.
  • the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 6. More suitably, the fusion protein is according to SEQ ID NO: 5 or SEQ ID NO: 6. In one embodiment, the fusion protein shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% with SEQ ID NO: 59 or SEQ ID NO: 60.
  • the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 59.
  • the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 60. More suitably, the fusion protein is according to SEQ ID NO: 59 or SEQ ID NO: 60.
  • the fusion protein shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% with SEQ ID NO: 65 or SEQ ID NO: 66.
  • the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 65.
  • the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 66. More suitably, the fusion protein is according to SEQ ID NO: 65 or SEQ ID NO: 66. In one embodiment, the fusion protein shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% with SEQ ID NO: 68 or SEQ ID NO: 69.
  • the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 68.
  • the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 69. More suitably, the fusion protein is according to SEQ ID NO: 68 or SEQ ID NO: 69.
  • the exosomal protein and the one or more immunogenic protein are separated by a linker sequence.
  • an exosome comprising the fusion protein of the invention or a membrane preparation comprising the fusion protein of the invention.
  • the exosome or membrane preparation comprises a DC receptor ligand and/or an immune enhancing moiety, optionally wherein the immune enhancing moiety is selected from the group consisting of a molecule with a pathogen-associated molecular pattern (PAMP), DNA oligonucleotide containing an CpG motif, a STING agonist (such as cyclic GMP-AMP), and a TLR9 (Toll-like Receptor) agonist.
  • PAMP pathogen-associated molecular pattern
  • STING agonist such as cyclic GMP-AMP
  • TLR9 Toll-like Receptor
  • a vaccine composition comprising an exosome or membrane preparation of the invention.
  • nucleic acid molecule encoding a fusion protein of the invention.
  • an expression vector comprising the nucleic acid molecule of the invention.
  • the vector is a lentivirus expression vector.
  • a cell comprising a fusion protein, an exosome, a membrane preparation, a nucleic acid, and/or an expression vector if the invention, optionally wherein the cell is an isolated cell.
  • the transformed cell line is a HeLa cell or a LCL721 cell.
  • the cell is modified to enhance the production of exosomes.
  • the cell has reduced expression of tetherin (Bst2).
  • Bst2 tetherin
  • an exosome, a membrane preparation, or vaccine composition of the invention for use as a medicament.
  • an exosome, a membrane preparation, or a vaccine composition of the invention for use in the prevention or amelioration of an infection.
  • the infection is a viral, bacterial or fungal infection.
  • the viral infection is a SARS-CoV-2 infection.
  • a method of preventing or ameliorating an infection in a subject comprising providing the subject in need thereof with a therapeutically effective amount of an exosome, a membrane preparation, or a vaccine composition of the invention.
  • the infection may be a viral, bacterial or fungal infection.
  • the viral infection is a SARS-CoV-2 infection.
  • any consideration set out with reference to the fusion protein will be applicable to the exosome, nucleic acid molecule, expression vector, vaccine composition, cell, uses and treatment methods of the invention.
  • the words “comprise” and “contain” and variations of them mean “including but not limited to”, and they are not intended to (and do not) exclude other moieties, additives, components, integers or steps.
  • a schematic representation of exemplary fusion polypeptides of the invention (A). A schematic representation of the orientation of luminal and intravesicular loops for native CD63 protein and CD63 fusion protein (B). Schematic representation of CD63 fusion constructs (C).
  • Figure 3. A schematic representation of an exemplary expression vector of the invention.
  • Figure 4. Shows the results of a restriction digest. Minipreps 1+3 were both found to have the correct sized insert by test digest, and the CD63-FLAG-SpikeRBD was confirmed by sanger sequencing (SEQ ID NO: 19 to 22 - restriction sites highlighted in red. Start codon in bold and italics. CD63 sequence underlined. FLAG in bold text. Linkers are italics. SpikeRBD in underlined text).
  • FIG. 1 A photo showing immunofluorescence assay results.
  • pLVX-EF1alpha-CD63-FLAG- SpikeRBD-IRES-Puro DNA was transiently transfected to HeLa cells and subject to immunofluorescence.
  • Figure 6. An image showing immunofluorescence results. FLAG signal was confirmed to be in CD63-positive compartments as shown by co-localisation of signal, and CD63 localisation remained in punctate organelles in transfected cells (as is also observed in non-transfected cells).
  • Figure 7. An image showing immunofluorescence assay results.
  • FIG. 8 An image showing Western Blot results. Whole cell lysates were collected from untransduced and transduced WT and Bst2KO HeLa cells. Lysates were probed with antibodies against CD63, and both transduced cell lines displayed enhanced staining, and at a higher molecular weight – reflecting additional molecular mass given from the addition of FLAG-SpikeRBD (A).
  • Figure 13B Characterization of exosomes containing three different constructs (V1, V19, and V16).
  • a flow nanoanalyzer (NanoFCM), a cytometer specifically designed to analyse particles smaller than the wavelength of visible light, was used to measure size and concentrations of isolated exosomes. Overall, the particles had an average modal diameter ranging between 62-69 nm, which is within the accepted range for exosomes. It can be seen from this figure that, exosomes with the V16 construct containing both N and sRBD domains were the largest.
  • Figure 14 A schematic illustrating the principles of the assay described in Example 2.
  • Figure 15. Illustrates the results of the T-cell activation described in Example 2.
  • Fig 16B shows that the different T cell lines were specific for numerous epitopes within S (Fig 16B), M and N.
  • Autologous B cell derived lymphoblastoid cell lines (LCL) from the respective donors were established as antigen presenting cells (APC) that can be pulsed with either viral proteins, peptides, or exosome vaccines.
  • Fig 16C shows a dose-dependent stimulation of S-specific polyclonal T cells co-cultured with autologous LCL exposed to FLAG-SRBD-CD63 exosomes.
  • ELISA a >60% maximal IFN ⁇ response was induced, but no measurable response to control exosomes.
  • Fig 16D shows that exosomes with sRBD (V1 construct) stimulate 2 independent CD4 S specific clones.
  • Fig 16E shows that multiple clones with extraordinar specificity for defined epitopes within S, M and N have been isolated.
  • Several are restricted by common alleles such as DRB1*01:01 or DRB4* 01:03 expressed in ⁇ 10-30% of the UK population.
  • the present invention is based on the inventors’ development of fusion proteins that may be useful in the context of vaccines, in particular anti-SARS-CoV-2 vaccines. As shown in the examples section of the present description, the inventors have developed fusion proteins that may have the ability to strongly activate T-cells rendering the fusion proteins especially useful in the context of vaccines.
  • the fusion proteins of the invention comprise an exosomal protein, which enables it to be localised on exosomes.
  • the inventors have shown that the fusion proteins of the invention do not prevent exosomal biogenesis and/or processing, allowing for exosomes expressing the fusion proteins to be released by cells to which a vector encoding the fusion proteins has been provided.
  • Exosomes comprising the fusion proteins of the invention may be provided to a subject and elicit an immune response that may prevent or ameliorate the symptoms and disease associated with a pathogen infection, such as for example SARS-CoV-2.
  • exosomes may be associated with a number of advantages compared to other vaccination approaches currently investigated.
  • exosomes have the ability to taken up by antigen presenting cells or directly fuse to the plasma membrane of antigen presenting cells. This may be beneficial because it may enable efficient uptake of antigens to cells key to immune stimulation (such as dendritic cells).
  • Exosomes may be packed with a payload of multiple viral antigens allowing the stimulation of B-cell immune and/or T- cell immune responses.
  • fusion proteins of the invention comprise immunogenic proteins capable of eliciting B- cell immune and/or T-cell immune responses.
  • Fusion protein and “fusion polypeptide” are used herein interchangeably, and refer to an artificial protein comprising amino acid sequences of at least two different proteins or protein variants, or fragments thereof.
  • the fusion protein of the invention comprises an exosomal protein and one or more immunogenic protein.
  • exosomal protein refers to any polypeptide that is enriched in a vesicular structure. The exosomal protein may therefore be utilised to transport the fusion protein of the invention to the vesicular structure.
  • the vesicular structure is an exosome, suitably a human exosome.
  • the exosomal protein may be a human exosomal protein.
  • the exosomal protein is a tetraspanin.
  • the tetraspanin is selected from the group consisting of CD63 (SEQ ID NO: 43), CD9 (SEQ ID NO: 44) and CD81 (SEQ ID NO: 45) or a fragment, or variant thereof.
  • the tetraspanin variant may share at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, identity with SEQ ID NO: 43, SEQ ID NO: 44 or SEQ ID NO: 45.
  • the tetraspanin variant may share at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, identity with SEQ ID NO: 43, SEQ ID NO: 44 or SEQ ID NO: 45.
  • the tetraspanin variant may share at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, identity with SEQ ID NO: 43.
  • the tetraspanin fragment may comprise or consist of at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% contiguous amino acids of SEQ ID NO: 43, SEQ ID NO: 44 or SEQ ID NO: 45.
  • the fragment may comprise or consist of at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% contiguous amino acids of SEQ ID NO: 43, SEQ ID NO: 44 or SEQ ID NO: 45. More suitably, the fragment may comprise or consist of at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% contiguous amino acids of SEQ ID NO: 43.
  • the tetraspanin protein is CD63 (SEQ ID NO: 43).
  • CD63 protein has four transmembrane domains, and three loops.
  • Two of the loops are luminal loops, meaning that they extend outward from the vesicular structure (such as exosome).
  • One of the loops is intravesicular, meaning that it extends into the vesicular structure (such as exosome).
  • the luminal loop that is closer to the N-terminal of the protein is referred to as the first (I) luminal loop
  • the luminal loop that is closer to the C-terminal of the protein is referred to as the second (II) luminal loop.
  • amino acids 33 to 51 of the human CD63 protein define the I luminal loop.
  • amino acids 103 to 203 of the human CD63 protein define the II luminal loop.
  • immunogenic protein refers to a polypeptide that is capable of eliciting an immune response.
  • the immunogenic protein may be capable of eliciting a protective immune response in a host.
  • a protective immune response may establish or improve the host’s immunity to a pathogen.
  • the pathogen may be selected from the group consisting of a virus, a bacterium, and a fungus. More suitably the virus may be SARS-CoV-2.
  • the host may be a mammal, preferably human.
  • the immunogenic protein may elicit a B-cell and/or T-cell immune response.
  • the phrase "elicit a B-cell immune response” as used herein refers to the immunogenic protein’s ability to trigger a humoral immune response, meaning that the immunogenic protein may cause cells of the subject’s immune system to produce antibodies that bind specific protein targets.
  • the protein targets may correspond to the immunogenic protein.
  • the protein target to which the antibodies are produced is SARS-CoV-2 spike protein (S), variant, or fragment thereof
  • the immunogenic protein may have a sequence corresponding to that SARS-CoV-2 spike protein (S) or variant, or fragment thereof.
  • the phrase "elicit a B-cell immune response” as used herein refers to the immunogenic protein’s ability to trigger a cellular immune response, meaning that the immunogenic protein may trigger the selection, expansion and maturation of B-cells producing antibodies capable of binding the antigen. Some of these antibodies are neutralizing antibodies that are capable of blocking the infection of host cells by the SARS-CoV-2 virus.
  • the phrase “elicit a T-cell immune response” as used herein refers to the immunogenic protein’s ability to trigger a cellular immune response, meaning that the immunogenic protein may trigger the selection and expansion of specific helper and/or cytotoxic T- lymphocytes capable of directly eliminating the cells that contain the antigen.
  • a T-cell immune response may be initiated when the immunogenic protein is presented as an antigen on the surface of the exosome. It will be appreciated that whether the immunogenic protein elicits a B-cell and/or T- cell immune response may depend upon the immunogenic protein’s location with respect to the exosomal protein. As explained in more detail elsewhere in the present specification, by way of example, immunogenic proteins incorporated into a luminal loop of an exosomal protein, may elicit a B-cell and T-cell immune response. Notably, antigens designed to elicit a B-cell response are constructed in the luminal loop of the fusion protein in order to orient the antigen epitope on the outside (luminal) of the exosome.
  • Immunogenic proteins incorporated into intravesicular loops, or at or near the N-terminal of an immunogenic protein may elicit a T-cell immune response.
  • “At the N-terminal” as used herein means attached to the first amino acid of a sequence encoding a protein that would normally have a free amine group.
  • Near the N-terminal as used herein means attached to an amino acid that is several amino acids away from the first amino acid of a sequence encoding a protein.
  • the immunogenic protein may be referred to as being near the N-terminal if it is attached to an amino acid located 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more amino acids away from the first amino acid of a sequence encoding a protein.
  • the immunogenic protein may be capable of eliciting a B-cell and/or T-cell immune response against a SARS-CoV-2 protein, variant, or fragment thereof.
  • the SARS-CoV-2 protein against which the immunogenic protein elicits an immune response may be selected from the group consisting of spike protein (S), nucleoprotein (N), membrane protein (M), envelope protein (E), ORF 1ab polyprotein, ORF 3a, ORF 3b protein, ORF 6 protein, ORF 7a protein, ORF 7b protein, ORF 8 protein, ORF 9b protein, ORF 9c protein and ORF 10 protein.
  • the spike protein (S) plays a key role in the receptor recognition and cell membrane fusion process, and is composed of two subunits, S1 and S2.
  • the S1 subunit contains a receptor-binding domain (RBD) that recognises and binds to the host receptor angiotensin-converting enzyme 2 (ACE2), while the S2 subunit mediates viral cell membrane fusion by forming a six-helical bundle via the two-heptad repeat domain.
  • the nucleoprotein (N) also known as the nucleocapsid protein
  • RNP helical ribonucleocapsid
  • the membrane protein (M) is a component of the viral envelope that plays a central role in virus morphogenesis and assembly via its interactions with other viral proteins.
  • the envelope protein (E) plays a central role in virus morphogenesis and assembly.
  • the protein acts as a viroporin and self-assembles in host membranes forming pentameric protein-lipid pores that allow ion transport. It also plays a role in the induction of apoptosis.
  • the ORF 1ab gene encodes 16 structural proteins, which include an RNA polymerase co-factor non-structural protein 7 (NSP7), RNA polymerase (NSP12), and helicase NSP13. These proteins are highly conserved amongst beta coronaviruses due to their key roles in the viral life cycle.
  • ORF 1ab polyprotein includes proteins selected from the group consisting of: NSP7, NSP12, and NSP13, or fragments thereof.
  • the ORF 3a protein Forms homotetrameric potassium sensitive ion channels (viroporin) and may modulate virus release.
  • the ORF 3b protein is potent interferon antagonist, suppressing the induction of type I interferon.
  • the ORF6 protein disrupts cell nuclear import complex formation by tethering karyopherin alpha 2 and karyopherin beta 1 to the membrane.
  • the ORF 7a protein plays a role as antagonist of host tetherin (BST2), disrupting its antiviral effect. It acts by binding to BST2 thereby interfering with its glycosylation.
  • BST2 host tetherin
  • the ORF 7a protein may also facilitate the disassembly of the Golgi apparatus.
  • the ORF 7b protein is transmembrane protein postulated to have the potential to interfere with important cellular processes that involve leucine-zipper formation by multimerizing with phospholamban and with E-cadherins.
  • the ORF 8 protein is believed to play a role in modulating host immune response and may play a role in blocking host IL17 cytokine by its interaction with host IL17RA.
  • the ORF 9b protein plays a role in the inhibition of host innate immune response by targeting the mitochondrial-associated adapter MAVS.
  • the ORF 9c protein is postulated to enhance the host proteasome activity thereby affect immune evasion, virulence and pathogenesis of the SARS-CoV2 virus.
  • the ORF 10 protein is postulated to bind Collin ubiquitin ligase complex and hijack it for ubiquitination and degradation of restriction factors.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 protein may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 protein, variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 spike protein may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 spike protein (S), variant, or a fragment thereof.
  • the fragment may comprise or consist of the extracellular domain of the spike protein (S).
  • the immunogenic protein may have an amino acid sequence that shares at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with the extracellular domain of the spike protein (S) or variant, or fragment thereof.
  • the fragment may comprise or consist of the receptor binding domain (RBD).
  • the immunogenic protein may have an amino acid sequence that shares at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with the receptor binding domain (RBD) (SEQ ID NO: 4), or variant, or fragment thereof. More suitably, the fragment is according to SEQ ID NO: 4.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 nucleoprotein may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 nucleoprotein (N) (SEQ ID NO: 2), variant, or a fragment thereof.
  • the fragment may share at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with SEQ ID NO: 56 or SEQ ID NO: 71. More suitably, the fragment is according to SEQ ID NO: 56 or SEQ ID NO: 71.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 membrane protein may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 membrane protein (M) (SEQ ID NO: 3), variant, or a fragment thereof.
  • the fragment may share at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with SEQ ID NO:57 or 72. More suitably, the fragment is according to SEQ ID NO: 72.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 envelope protein (E) may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 envelope protein (E), variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF3a may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF3a, variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF3b may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF3b, variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF6 may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF6, variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF7a may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF7a, variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF7b may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF7b, variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF8 may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF8, variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF9b may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF9b, variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF9c may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF9c, variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF10 may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF10, variant, or a fragment thereof.
  • the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF1ab may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF1ab, variant, or a fragment thereof. Sequences of the above-mentioned proteins are provided under the section “Sequences” of the present disclosure.
  • the fusion protein may comprise one, two, three, or more immunogenic proteins.
  • the fusion protein may comprise one immunogenic protein.
  • the fusion protein may comprise two immunogenic proteins.
  • the fusion protein may comprise three immunogenic proteins.
  • the immunogenic protein may elicit a B-cell and a T-cell immune response, or a T-cell immune response.
  • the type of immune response elicited may depend on the localisation of the immunogenic protein in the exosome.
  • the immunogenic protein that elicits a B-cell and a T-cell immune response elicits a response against the SARS-CoV-2 spike protein (S).
  • S SARS-CoV-2 spike protein
  • the immunogenic protein that elicits a T-cell response elicits a response against the nucleoprotein (N) or membrane protein (M).
  • N nucleoprotein
  • M membrane protein
  • the fusion protein comprises two or more immunogenic proteins
  • some or all of the immunogenic proteins may be the same or different.
  • the immunogenic proteins may have a sequence corresponding to different proteins, or a sequence that corresponds to different parts of the same protein (the different parts may be overlapping or non- overlapping).
  • the first immunogenic protein may elicit a B-cell and a T-cell immune response
  • the second immunogenic protein may elicit a T-cell immune response.
  • the immunogenic protein that elicits a B-cell and a T-cell immune response may elicit a response against the SARS-CoV-2 spike protein (S).
  • S SARS-CoV-2 spike protein
  • the immunogenic protein that elicits a T-cell response may elicit a response against the nucleoprotein (N) or membrane protein (M).
  • N nucleoprotein
  • M membrane protein
  • SEQ ID NO: 17, 59, or 62 are exemplary amino acid sequences of such proteins.
  • each one of the immunogenic proteins may elicit a T-cell response.
  • the first immunogenic protein may elicit a response against the SARS-CoV-2 nucleoprotein (N), whereas the second immunogenic protein elicits a response against membrane protein (M).
  • SEQ ID NO: 13, 17, or 62 are exemplary amino acid sequences of such proteins.
  • fusion protein may comprise one or two immunogenic proteins that elicit a T-cell immune response, and one or two immunogenic proteins that elicit a B-cell and T-cell immune response.
  • the fusion protein may comprise two immunogenic proteins that elicit a T-cell immune response against the nucleoprotein (N) or membrane protein (M), and an immunogenic protein that elicits a B-cell and T-cell immune response against SARS-CoV-2 spike protein (S).
  • N nucleoprotein
  • M membrane protein
  • S SARS-CoV-2 spike protein
  • the type of immune response elicited by the immunogenic protein may be dictated by the immunogenic protein’s location with respect to the exosomal protein.
  • Immunogenic proteins incorporated into a luminal loop of an exosomal protein for example a tetraspanin, such as CD63, may elicit a B-cell and a T-cell immune response.
  • the immunogenic protein that is incorporated into the luminal loop may be one that elicits a response against SARS-CoV-2 spike protein (S).
  • S SARS-CoV-2 spike protein
  • the immunogenic protein may be incorporated into the first and/or second luminal loop.
  • the immunogenic protein may be incorporated between amino acids Q36 and L37 (thereby incorporated into loop I) and/or between amino acids R108-D109 (thereby incorporated into loop II).
  • Immunogenic proteins incorporated into intravesicular loops, or at or near the N- terminal of an exosomal protein for example a tetraspanin, such as CD63, may elicit a T-cell immune response.
  • an immunogenic protein that is incorporated into the intravesicular loop and/or at or near the N-terminal of an exosomal protein may be one that elicits a response against SARS-CoV-2 nucleoprotein (N) or membrane protein (M).
  • the fusion protein of the invention may comprise a linker.
  • the linker may be between the immunogenic protein and exosomal protein sequence.
  • the linker may be on one or both side of the immunogenic protein.
  • a linker may be present between some, or all, of the immunogenic proteins and exosomal protein sequence.
  • the linker may be 20 or fewer amino acids in length.
  • the linker may be about 18, about 16, about 14, about 12 or about 10 amino acids in length.
  • the linker may be about 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2 or 1 amino acid in length. More suitably the linker may be from 6 to 2 amino acids long.
  • the linker may be a flexible or a rigid linker. More suitably the liker is flexible.
  • a flexible linker may have the sequence GGSG, GGGSG, GGGGS, GGG, or GG.
  • the fusion protein comprises CD63 and the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof.
  • S SARS-CoV-2 spike protein
  • the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof is incorporated into the first luminal of CD63.
  • the extracellular domain of the spike protein (S), variant, or fragment thereof may be incorporated between amino acids Q36 and L37 of CD63.
  • the extracellular domain of the SARS- CoV-2 spike protein may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1.
  • the fragment may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4.
  • a linker may be incorporated between both sides of the immunogenic protein and exosomal protein.
  • the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7 or SEQ ID NO:8. More suitably, the fusion protein comprises or consists of SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7 or SEQ ID NO:8.
  • the fusion protein comprises CD63 and the extracellular domain of the SARS-CoV-2 spike (S) protein or a fragment thereof.
  • the extracellular domain of the SARS-CoV-2 spike (S) protein or a fragment thereof is incorporated into the second luminal of CD63.
  • the extracellular domain of the spike protein (S), variant, or fragment thereof may be incorporated between amino acids R108 and D109 of CD63.
  • the extracellular domain of the SARS-CoV-2 spike (S) protein have a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1.
  • the fragment may have a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4.
  • a linker may be incorporated between both sides of the immunogenic protein and exosomal protein.
  • the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:15 or SEQ ID NO:16. More suitably, the fusion protein comprises or consists of SEQ ID NO:15 or SEQ ID NO:16.
  • the fusion protein comprises CD9 and the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof.
  • the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof is incorporated into the first luminal of CD9.
  • the extracellular domain of the spike protein (S), variant, or fragment thereof may be incorporated between amino acids N50 and N51 of CD9.
  • the extracellular domain of the SARS-CoV- 2 spike protein may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1.
  • the fragment may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4.
  • a linker may be incorporated between both sides of the immunogenic protein and exosomal protein.
  • the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:23 or SEQ ID NO:24. More suitably, the fusion protein comprises or consists of SEQ ID NO:23 or SEQ ID NO:24.
  • the fusion protein comprises CD81 and the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof.
  • the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof is incorporated into the first luminal of CD81.
  • the extracellular domain of the spike protein (S), variant, or fragment thereof may be incorporated between amino acids L49 and G50 of CD81.
  • the extracellular domain of the SARS- CoV-2 spike protein may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1.
  • the fragment may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4.
  • a linker may be incorporated between both sides of the immunogenic protein and exosomal protein.
  • the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:25 or SEQ ID NO:26. More suitably, the fusion protein comprises or consists of SEQ ID NO:25 or SEQ ID NO:26.
  • the fusion protein comprises CD63 and SARS-CoV-2 nucleoprotein (N) and/or membrane protein (M), or fragment thereof.
  • the SARS-CoV-2 nucleoprotein (N) and/or membrane protein (M), or fragment thereof is incorporated near or at the N-terminal of CD63.
  • the SARS-CoV-2 nucleoprotein (N) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 2.
  • the SARS-CoV-2 membrane protein (M) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 3.
  • the fragment of nucleoprotein (N) and has a sequence that is sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 56 or SEQ ID NO: 71.
  • the fragment of membrane protein (M) has a sequence that is sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 57, or SEQ ID NO: 72, more suitably SEQ ID NO: 72.
  • a linker may be incorporated between the immunogenic protein and the exosomal protein.
  • the membrane protein (M) may be incorporated at the N-terminal of the exosomal protein, followed by the nucleoprotein (N).
  • the nucleoprotein (N) and membrane protein (M) may or may not be separated by a linker.
  • the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 68 or SEQ ID NO:69.
  • the fusion protein comprises or consists of SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 68 or SEQ ID NO:69.
  • the fusion protein comprises CD63 and SARS-CoV-2 nucleoprotein (N) or membrane protein (M), or fragment thereof.
  • SARS- CoV-2 nucleoprotein (N) or membrane protein (M), or fragment thereof is incorporated near or at the N-terminal of CD63.
  • the SARS-CoV-2 nucleoprotein (N) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 2.
  • the fragment of nucleoprotein (N) and has a sequence that is sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 56 or SEQ ID NO: 71.
  • the SARS-CoV-2 membrane protein (M) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 3.
  • the fragment of membrane protein (M) has a sequence that is sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 57 or SEQ ID NO: 72, more suitably SEQ ID NO: 72.
  • a linker may be incorporated between the immunogenic protein and the exosomal protein.
  • the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 68, or SEQ ID NO: 69.
  • the fusion protein comprises or consists of SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 68, or SEQ ID NO: 69.
  • the fusion protein comprises CD63 and the extracellular domain of the SARS-CoV-2 spike (S) protein or a fragment thereof, and nucleoprotein (N) and/or membrane protein (M), or fragment thereof.
  • the extracellular domain of the SARS-CoV-2 spike (S) protein or a fragment thereof is incorporated into the first or second luminal of CD63.
  • the extracellular domain of the SARS- CoV-2 nucleoprotein (N) and/or membrane protein (M), or fragment thereof is incorporated near or at the N-terminal of CD63.
  • the extracellular domain of the SARS-CoV-2 spike (S) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1.
  • the fragment may have a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4.
  • the SARS-CoV-2 nucleoprotein (N) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 2.
  • the fragment of nucleoprotein (N) and has a sequence that is sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 56 or SEQ ID NO: 71.
  • the SARS-CoV-2 membrane protein (M) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 3.
  • the membrane protein (M) fragment has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 57, or SEQ ID NO: 72.
  • the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, or SEQ ID NO: 63. More suitably, the fusion protein comprises or consists of SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, or SEQ ID NO: 63.
  • the fusion protein comprises CD63 and SARS-CoV-2 membrane protein (M), variant, or a fragment thereof, and extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof.
  • the fragment of the membrane protein (M) may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 57 or 72. More suitably, the fragment is according to SEQ ID NO: 72.
  • the membrane protein (M), variant or fragment is incorporated into a luminal loop of CD63.
  • the extracellular domain of the SARS-CoV-2 spike protein (S) may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1.
  • the fragment of the spike protein (S) may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4.
  • the fragment of the spike protein (S) is according to SEQ ID NO: 4.
  • a linker may be incorporated between both sides of the immunogenic protein and exosomal protein.
  • the terms “homology” and “identity” are used interchangeably. Calculations of sequence homology or identity between sequences may be performed as follows.
  • sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, and even more preferably at least 70%, 75%, 80%, 82%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% of the length of the reference sequence.
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid “homology”.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. In a preferred embodiment, the percent identity between two amino acid sequences is determined using the Needleman et al. (1970) J. Mol.
  • Biol.48:444-453 algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a BLOSUM 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
  • the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (available at http://www.gcg.com), using a NWSgapdna. CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6.
  • a particularly preferred set of parameters are a BLOSUM 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
  • the percent identity between two amino acid or nucleotide sequences can be determined using the algorithm of Meyers et al. (1989) CABIOS 4:11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • Other methods of determining sequence identity will be known to those skilled in the art.
  • the fusion proteins of the invention are artificial proteins comprising amino acid sequences of at least two different proteins or protein variants, or fragments thereof.
  • the fusion proteins of the invention comprise an exosomal protein or an exosomal protein variant, or a fragment thereof, and one or more immunogenic protein or immunogenic protein variant, or fragment thereof.
  • a “protein” for example an exosomal protein or immunogenic protein, this also refers to the protein variant, or fragment of the protein or protein variant.
  • variant refers to a polypeptide that shares a partial sequence identity with the amino acid encoding the exosomal protein or the immunogenic protein.
  • fragment refers to a polypeptide that shares 100% identity with the corresponding portion of the amino acid sequence of the exosomal or immunogenic protein or variant, but that consist of a truncation in the amino acid sequence encoding the exosomal protein (for example a tetraspanin, such as CD63) or immunogenic protein (for example a protein selected from the group consisting of spike protein (S), nucleoprotein (N), and membrane protein (M) of SARS- CoV-2).
  • exosomes for example a protein selected from the group consisting of spike protein (S), nucleoprotein (N), and membrane protein (M) of SARS- CoV-2).
  • S spike protein
  • N nucleoprotein
  • M membrane protein of SARS- CoV- 2
  • Exosomes in one aspect the present invention provides exosomes comprising the fusion proteins of the invention.
  • An “exosome”, as used herein, is a membrane-bound extracellular vesicle between 30-150 nanometers in size.
  • exosomes are shed into the blood, urine, cerebrospinal fluid and other bodily fluids from cells, and may contain some of the proteins and functional ribonucleic acid (RNA) molecules from their cells of origin.
  • exosomes may be shed into the cell culture media.
  • the cells of origin as per the present invention may be selected from a wide range of cells. Merely by way of example the cells may be HeLa cells. Methods of purifying exosomes from cell culture media are well known in the art. Merely by way of example, exosomes may be purified using the Total Exosome Isolation Reagent (Invitrogen TM ) following the manufacturer’s instructions.
  • exosomes may be advantageous because they can be efficiently taken up by dendritic cells (DCs).
  • DCs are proficient at antigen processing and presentation and promotion of an active and beneficial T-cell immune response.
  • Antigens such as the immunogenic proteins of the fusion protein of the invention
  • DCs as well as unprocessed antigens, travel through the lymph to the nearest draining node to stimulate antigen specific T-cells. This leads to a cellular response, invoking both CD8 cytotoxic and CD4 helper T-cells, as well as humoral responses leading to production of specific antibodies. In some cases, this process may be made even more efficient by enhancing targeting of the exosomes to DCs.
  • DCs express receptors on their surface, including C-type lectin receptor, mannose receptor, DEC-205 or Dectin-1 that trigger DC activation.
  • ligands for these receptors may be expressed on the exosomes.
  • the exosomes of the invention may further comprise a DC receptor ligand.
  • the ligand may be a natural biological ligand, or a nanobody or related structure designed to bind to the receptor. It will be appreciated that the ligand may be encoded by a polypeptide forming part of the fusion protein, or a separate polypeptide.
  • the fusion protein of the invention may further comprise a polypeptide encoding a DC receptor ligand.
  • the exosomes of the invention may comprise an immune enhancing moiety.
  • an immune enhancing moiety may be selected from the group consisting of a molecule with a pathogen-associated molecular pattern (PAMP), DNA oligonucleotides containing the CpG motif, a STING agonist (such as cyclic GMP-AMP) and a TLR9 (Toll-like Receptor) agonist.
  • PAMP pathogen-associated molecular pattern
  • a molecule with a pathogen-associated molecular pattern (PAMP) may be advantageous as it may help the exosomes induce a potent Th1 response.
  • adjuvants include synthetic CpG (a TLR9 agonist) or poly-ICLC (a TLR3 ligand).
  • DNA oligonucleotides containing the CpG motif may be advantageous as they may help stimulate cells of the immune system leading to the production of cytokines such as type-I interferon, IL-6, TNF and IL-12.
  • a STING agonist such as cyclic GMP-AMP
  • cytokine such as type I interferon.
  • Methods for producing exosomes are well known in the art.
  • Methods for producing exosomes expressing fusion proteins are also well known in the art.
  • the exosomes may be produced by cells modified (for example, genetically modified) to enhance the production of exosomes.
  • exosomes may be produced in cells which have reduced expression of proteins which retain exosomes, such as tetherin (bone marrow stromal antigen 2) protein.
  • reduced expression it is meant that the expression levels are lower (for example by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or 100%) as compared to a suitable control.
  • a suitable control may be, for example, a cell in which a gene encoding a protein that retains exosomes (such as tetherin) is not knocked out. Techniques for knocking out an undesirable gene, such as the gene encoding tetherin are also known in the art.
  • a cell that has reduced expression of tetherin may be isolated.
  • the exosome may be genetically modified.
  • such a genetically modified exosome may be the exosome of the invention.
  • such a genetically modified exosome may comprise the fusion protein of the invention.
  • Expression Vectors In one aspect the invention provides an expression vector comprising an expressible nucleic acid encoding the fusion proteins herein described.
  • the nucleic acid may share at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with a sequence selected from the group consisting of: SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:61, SEQ ID NO:64, SEQ ID NO:67 and SEQ ID NO:70.
  • the vector may be any vector capable of transferring DNA to a cell.
  • the vector is an integrating vector or an episomal vector.
  • Preferred integrating vectors include recombinant retroviral vectors.
  • a recombinant retroviral vector will include DNA of at least a portion of a retroviral genome which portion is capable of infecting the target cells.
  • the term “infection” is used to mean the process by which a virus transfers genetic material to its host or target cell.
  • the retrovirus used in the construction of a vector of the invention is also rendered replication-defective to remove the effect of viral replication of the target cells. In such cases, the replication-defective viral genome can be packaged by a helper virus in accordance with conventional techniques.
  • the retroviral vector may be a lentiviral vector.
  • Lentiviral vectors are well known in the art, and have been used to deliver genes to numerous cell types, including HeLa or B lymphoid cells.
  • Other vectors useful in the present invention include adenovirus, adeno-associated virus, SV40 virus, vaccinia virus, HSV and poxvirus vectors.
  • Preferred episomal vectors include transient non-replicating episomal vectors and self-replicating episomal vectors with functions derived from viral origins of replication such as those from EBV, human papovavirus (BK) and BPV-1.
  • the vector of the present invention is a plasmid.
  • the plasmid may be a non-replicating, non-integrating plasmid.
  • plasmid refers to any nucleic acid encoding an expressible gene and includes linear or circular nucleic acids and double or single stranded nucleic acids.
  • the nucleic acid can be DNA or RNA and may comprise modified nucleotides or ribonucleotides, and may be chemically modified by such means as methylation or the inclusion of protecting groups or cap- or tail structures.
  • a non-replicating, non-integrating plasmid is a nucleic acid which when transfected into a host cell does not replicate and does not specifically integrate into the host cell’s genome (i.e. does not integrate at high frequencies and does not integrate at specific sites).
  • Replicating plasmids can be identified using standard assays including the standard replication assay of Ustav et al (1991 EMBO J 10: 449–457).
  • the present invention also provides a cell transformed or transfected with (i.e. comprising) the expression vector of the present invention.
  • the present invention also provides a cell comprising the fusion protein of the invention or exosome of the invention.
  • the cell is an isolated cell.
  • the cell is a mammalian cell.
  • the cell is a HeLa cell.
  • the cell may be modified to produce increased amounts of exosomes as compared to control cells.
  • a vaccine composition Provided herein is a vaccine composition comprising the exosome of the invention or a membrane preparation comprising the fusion protein of the invention.
  • the vaccine is anti-SARS-CoV-2.
  • the term “vaccine composition” and “composition” are used herein interchangeably and refer to a composition comprising, in addition to comprising the exosome of the invention, a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier.
  • "vaccine” is defined broadly to refer to any type of biological agent in an administrable form capable of stimulating an immune response (for example a B-cell immune and/or T-cell immune response) in an animal (for example human) inoculated with the vaccine.
  • the vaccine prevents or ameliorates the symptoms of a disease, such as an infection (for example COVID-19).
  • membrane preparation refers to a composition comprising membrane fragments from cells which express the fusion protein (for example HeLa cells or LCL721 cells). Methods for obtaining a membrane preparation are well known in the art.
  • the fusion protein is attached to the membrane fragments in the membrane preparation.
  • the fusion protein is attached to the membrane fragments by the exosomal protein (such as tetraspanin).
  • the present invention provides a membrane preparation comprising the fusion protein of the invention.
  • compositions may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, supplementary immune potentiating agents such as adjuvants and cytokines and optionally other therapeutic agents or compounds.
  • pharmaceutically acceptable refers to a material that is not biologically or otherwise undesirable, i.e., the material may be administered to an individual along with the selected binding protein without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained.
  • Excipients are natural or synthetic substances formulated alongside an active ingredient (e.g.
  • fusion protein or exosome of the invention included for the purpose of bulking-up the formulation or to confer a therapeutic enhancement on the active ingredient in the final dosage form, such as facilitating drug absorption or solubility.
  • Excipients can also be useful in the manufacturing process, to aid in the handling of the active substance concerned such as by facilitating powder flowability or non-stick properties, in addition to aiding in vitro stability such as prevention of denaturation over the expected shelf life.
  • Pharmaceutically acceptable excipients are well known in the art. A suitable excipient is therefore easily identifiable by one of ordinary skill in the art.
  • suitable pharmaceutically acceptable excipients include water, saline, aqueous dextrose, glycerol, ethanol, and the like.
  • Diluents are diluting agents.
  • Pharmaceutically acceptable diluents are well known in the art. A suitable diluent is therefore easily identifiable by one of ordinary skill in the art.
  • Carriers are non-toxic to recipients at the dosages and concentrations employed and are compatible with other ingredients of the formulation.
  • carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • Pharmaceutically acceptable carriers are well known in the art. A suitable carrier is therefore easily identifiable by one of ordinary skill in the art.
  • Provided herein is the exosome or vaccine composition of the invention for use as a medicament. Said medicament may be used for human or animal therapy of various diseases or disorders in a therapeutically effective dose. Suitably the disease may be an infection.
  • the medicament can be formulated by various techniques dependent on the desired application purposes. Formulations described herein in connection to the vaccine composition of the invention are exemplary formulations of the medicament. Also provided herein is the exosome or vaccine composition of the invention for use in the prevention or amelioration of an infection in a subject.
  • the term “infection” as used herein refers to the undesirable presence of a microorganism in or on the subject’s body. Suitably, the microorganism is pathogenic. Such an undesirable presence of a microorganism may be symptomatic or asymptomatic. It will be appreciated that whether the infection is symptomatic or asymptomatic may depend on the subject and/or type of microorganism.
  • an infection is symptomatic
  • the type of symptoms and their severity may depend on the subject and/or type of microorganism.
  • the infection may be a viral, bacterial or fungal infection.
  • the symptoms may include fever, cough, and/or loss of taste and/or smell.
  • a method of eliciting an anti-SARS-CoV-2 immune response in a mammal comprising administering the exosome or a vaccine composition comprising the exosome of the invention to a subject.
  • Also provided herein is a method for preventing or ameliorating an infection in a subject comprising providing the subject in need thereof with a therapeutically effective amount of an exosome or vaccine composition of the invention.
  • “Therapeutically effective amount” as used herein refers to the amount of exosomes or vaccine composition of the invention that elicit a biological response in a tissue or system of a subject such that the infection is prevented or ameliorated. Methods of establishing a therapeutically effective amount will be known to those skilled in the art and merely by way of example can include performing a dose response assay.
  • subject will be understood to refer to a warm-blooded animal, particularly a mammal.
  • Non-limiting examples of animals within the scope and meaning of this term include humans, non-human primates, dogs, cats, guinea pigs, rats, mice, horses, goats, cattle, sheep, and zoo animals.
  • the present invention provides novel vaccine compositions comprising the exosomes of the invention for controlling SARS-CoV-2 infection in mammalian, most preferably human, subjects.
  • the present vaccine compositions are useful in providing immune resistance against the strain(s) of SARS-CoV-2 used for preparation of the fusion proteins of the invention, as well as against strains which are different from those used in the preparation of the fusion proteins.
  • vaccines of the invention are administered prophylactically.
  • a vaccine of the invention is administered therapeutically.
  • the vaccine is administered to a patient with an active SARS-CoV-2 infection to both treat the active infection and/or prevent recurrence of a SARS-CoV- 2.
  • SARS-CoV-2 genes are: - Tagged to the N terminus of CD63; - Inserted into luminal loops 1; and/or - Inserted into luminal loop 2. Location may depend on the antigen. For example, N and M will be N terminal, Spike or ACE2 constructs will be within the loops. For inserts to luminal loop 1 inserts are between Q36 and L37 of CD63.
  • inserts will be between R108 and D109 of CD63.
  • CD63-FLAG-SpikeRBD (V1), N-CD63-FLAG-SpikeRBD (V16), FLAG-N-CD63 (V19) and FLAG-M-CD63 (V18) were synthesised by GeneWiz and cDNA was flanked by a 5’ EcoRI and a 3’ BamHI site. These were ligated to pLVX-EF1alpha-IRES-Puro and transduced to competent cells. Minipreps were picked, analysed by test digests and potential colonies sequenced by Sanger sequencing (GeneWiz). The construct V18 showed stable expression (data not shown).
  • Minipreps 1+3 were both found to have the correct sized insert by test digest, and the CD63-FLAG-SpikeRBD was confirmed by sanger sequencing (Fig 4).
  • pLVX- EF1alpha-CD63-FLAG-SpikeRBD-IRES-Puro DNA was transiently transfected to HeLa cells and subject to immunofluorescence (Fig 5). FLAG signal was confirmed to be in CD63-positive compartments as shown by colocalisation of signal, and CD63 localisation remained in punctate organelles in transfected cells (as is also observed in non-transfected cells).
  • HEK293T cells were transfected with pLVX-EF1alpha-CD63- FLAG-SpikeRBD-IRES-Puro and the lentiviral packaging plasmids pCMVR8.91 (“199”) and pMD.G (VSV-G) (“200”) using TransIT-293 (Mirus Bio, USA).
  • Viral supernatants were collected 48 h after transfection, passed through 0.45 ⁇ m filters and both wild-type and tetherin KO HeLa cells transduced by ‘spinfection’—viral supernatants were centrifuged at 1800 rpm in a benchtop centrifuge at 37 °C for 3 h to enhance viral transduction. After 48 hours, transduced WT HeLa and Bst2KO HeLa cells were selected with 1ug/ml Puromycin for 5 days.
  • WT and Bst2KO stable HeLa cell lines were confirmed to express CD63-FLAG-SpikeRBD using immunofluorescence, again using anti-FLAG antibodies, but also using a human monoclonal antibody raised against SARS-CoV-2 Spike-RBD (P06DHu, ThermoFisher) (Fig 6).
  • FLAG staining colocalised to CD63, a marker for multivesicular endosomes – the compartment where intraluminal vesicles (and therefore exosomes) are generated.
  • the increase in CD63 staining in transduced cells reflects the additional, ectopic copy of CD63, rather than just the endogenous CD63.
  • Antibodies against FLAG were similarly used to blot whole cell lysates, and a positive band was observed at the correct molecular weight in transduced stable cell lines, and not in untransduced controls.
  • Exosome-enriched fractions were generated from cells and subject to Western blotting.
  • Exosomes from tetherin KO HeLa cells + CD63- FLAG-SpikeRBD displayed a band at the correct molecular weight as expected (Fig 8B). Further evidence of expression of FLAG-SpikeRBD in the CD63 fusion protein was obtained by mass spectrometry analysis.
  • CD63-FLAG-SpikeRBD sequence The sequence of the theoretical CD63-FLAG-SpikeRBD sequence is shown in SEQ ID NO: 5. Of the identified unique peptides identified, several were identified which originate from the SpikeRBD. The identified unique peptides, and their position within CD63-FLAG-SpikeRBD and amino acid position were defined as shown below: Finally, to confirm that exosomes from transduced cells contained FLAG-SpikeRBD, we performed surface labelling immunogold electron microscopy, making use of the FLAG epitope that is exposed on the outside of exosomes.
  • transduced and untransduced WT HeLa cells were treated with Bafilomycin A1 for 16 hours to induce exosome release as previously described (Edgar et al., eLife 2016;5:e17180 DOI: 10.7554/ELIFE.17180).
  • Cells were fixed before being labelled with antibodies against FLAG.10nm Protein-A gold was then used to detect anti-FLAG antibodies.
  • Cells were then re-fixed with glutaraldehyde before being processed for electron microscopy. Ultrathin sections were cut, stained with heavy metals and imaged. Immunogold labelling was only observed in transduced cells and was specifically seen on surface- associated exosomes as expected.
  • Example 2 The aim of the following example is to demonstrate that SARS-CoV2 spike protein containing exosomes (RBD region) elicit recognition by T-cells specific to epitopes with SARS-CoV-2 Spike. A polyclonal T-cell culture from a donor with reactivity to the RBD region of SARS-CoV-2 spike was used.
  • each tube of LCL cells were washed once with 8ml LCL media, resuspended in 200 ul of LCL medium and pipetted into appropriate wells of a 96U bottom plate containing T-cells. Outer wells of each plate were filled with saline to reduce evaporation and plates were incubated at 37°C 5% CO2. After 24 and 48hr incubation, 60 ul of supernatant from each well were dispensed into separate wells of a 96 well plate and stored at 4°C.
  • the interferon gamma (IFN- ⁇ ) concentration of each aliquot of supernatant was tested using an IFN- ⁇ capture ELISA, with doubling dilutions of recombinant IFN- ⁇ included for quantitation of IFN- ⁇ concentration in the samples.
  • IFN- ⁇ capture ELISA IFN- ⁇ capture ELISA
  • the polyclonal T-cell culture did not produce any interferon-gamma when co-cultured with the autologous LCL, but produced large quantities of the cytokine when co- cultured with the same LCL that had been exposed to SARS-CoV-2 spike RBD PepMix. This demonstrates that the polyclonal culture contains T-cells specific to epitopes within the RBD region. The lack of any recognition of the autologous LCL allows this polyclonal culture to be used to investigate exosome and membrane preparations. This polyclonal culture produced large quantities of interferon-gamma when co- cultured with autologous LCL exposed to the SARS-CoV-2 spike exosomes, but not the control exosomes.
  • Example 2 was repeated using exosomes containing constructs v16 (N- CD63-FLAG-SpikeRBD) or v19 (FLAG-N-CD63). For V16, T-cell activation was tested in both a polyclonal T-cell culture from a donor with reactivity to the RBD region of SARS-CoV-2 spike and N protein.
  • Cells were mounted on slides with mounting medium containing DAPI. Cells were imaged using a LSM700 confocal microscope. Flow cytometry Cells were gently trypsinised, and surface stained for flow cytometry in PBS with 0.5 % BSA + 1 mM EDTA (FACS buffer) for 30 min on ice. For intracellular staining, cells were first fixed then permeabilised with 0.1* saponin/PBS and subsequently stained in PBS with FACs buffer. Samples were acquired on a 4 laser Cytoflex S. Western blotting Cell lysates were mixed with 4x NuPage LDS sample buffer.
  • a fusion protein comprising: i) an exosomal protein, wherein the exosomal protein is a tetraspanin protein; and ii) one or more immunogenic protein.
  • the tetraspanin is selected from the group consisting of CD63, CD9, and CD81, or a fragment, or variant thereof.
  • the one or more immunogenic protein is capable of eliciting an immune response against a SARS-CoV-2 protein. 4.
  • the fusion protein of paragraph 3 wherein the SARS-CoV-2 protein is selected from the group consisting of spike protein (S), nucleoprotein (N), membrane protein (M), envelope protein (E), open reading frame (ORF) 1ab polyprotein, ORF 3a protein, ORF 3b protein, ORF 6 protein, ORF 7a protein, ORF 7b protein, ORF 8 protein, ORF 9b protein, ORF 9c protein, and ORF 10 protein, optionally wherein when the SARS- CoV-2 protein is spike protein (S) it is the extracellular domain of spike protein (S). 5.
  • the protein comprises a first one or more immunogenic protein capable of eliciting a B-cell and a T-cell immune response and a second one or more immunogenic protein capable of eliciting a T-cell immune response.
  • the first one or more immunogenic protein capable of eliciting a B-cell and T-cell immune response is incorporated into a luminal loop of the exosomal protein.
  • S spike protein
  • N nucleoprotein
  • M membrane protein
  • SEQ ID NO: 3 membrane protein
  • the fragment of the spike protein (S) is the receptor binding domain (RBD) (SEQ ID NO: 4)
  • the fragment of the membrane protein (M) is according to SEQ ID NO: 57, 58, or 72
  • the fragment of the nucleoprotein (N) is according to SEQ ID NO: 56 or 71. 17.
  • the fusion protein of any one of the preceding paragraphs sharing at least 75%, at least 80%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identity with SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 65, SEQ ID NO: 66,
  • the fusion protein of paragraphs 17, wherein the fusion protein comprises or consists of SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 59, SEQ ID NO:60, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO:68, or SEQ ID NO: 69. 19.
  • An exosome or membrane preparation comprising a fusion protein of any one of paragraphs 1 to 19. 21.
  • exosome or membrane preparation wherein the exosome further comprises a DC receptor ligand and/or an immune enhancing moiety, optionally wherein the immune enhancing moiety is selected from the group consisting of a molecule with a pathogen-associated molecular pattern (PAMP), DNA oligonucleotide containing an CpG motif, a STING agonist (such as cyclic GMP-AMP), and a TLR9 (Toll-like Receptor) agonist. 22.
  • a vaccine composition comprising an exosome or membrane preparation of paragraph 20 or 21.
  • An expression vector comprising the nucleic acid molecule of paragraph 23. 25.
  • a cell comprising a fusion protein of any one of paragraphs 1 to 19, an exosome or membrane preparation of paragraph 20 or 21, a nucleic acid of paragraph 23, and/or an expression vector of paragraph 24 or 25, optionally wherein the cell is an isolated cell.
  • the cell of paragraph 26, wherein the transformed cell line is a HeLa cell or a LCL721 cell.
  • 28. The cell of paragraph 26 or 27, wherein the cell is modified to enhance the production of exosomes.
  • 29. The cell of any one of paragraph 26 to 28, wherein the cell has reduced expression of tetherin.
  • 33. A method of preventing or ameliorating an infection in a subject, the method comprising providing the subject in need thereof with a therapeutically effective amount of an exosome or membrane preparation of paragraph 20 or 21, or a vaccine composition according to paragraph 22.
  • 34. wherein the infection is a viral, bacterial or fungal infection.
  • the method of paragraph 34, wherein the viral infection is a SARS-CoV-2 infection.

Landscapes

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

Abstract

The present invention relates to a fusion protein comprising an exosomal protein and one or more immunogenic protein, wherein the exosomal protein is a tetraspanin protein. The present invention also relates to an exosome comprising said fusion protein. Further, the present invention relates to a vaccine composition comprising an exosome of the invention. The present invention also relates to a nucleic acid molecule encoding said fusion protein, and an expression vector comprising said nucleic acid molecule. The present invention further relates to a cell comprising said fusion protein, exosome, nucleic acid molecule, and/or expression vector. The invention also relates to the exosome and the vaccine composition for use as a medicament. The invention also relates to the exosome and and/or the vaccine composition for use in the prevention or amelioration of an infection in a subject. Furthermore, the invention relates to a method of preventing or ameliorating an infection in a subject, the method comprising providing the subject in need thereof with a therapeutically effective amount of an exosome and/or a vaccine composition of the invention.

Description

FUSION PROTEIN FOR ANTIGEN PRESENTATION Field of the Invention The present invention relates to a fusion protein comprising an exosomal protein and one or more immunogenic protein, wherein the exosomal protein is a tetraspanin protein. The present invention also relates to an exosome comprising said fusion protein. Further, the present invention relates to a vaccine composition comprising an exosome of the invention. The present invention also relates to a nucleic acid molecule encoding said fusion protein, and an expression vector comprising said nucleic acid molecule. The present invention further relates to a cell comprising said fusion protein, exosome, nucleic acid molecule, and/or expression vector. The invention also relates to the exosome and the vaccine composition for use as a medicament. The invention also relates to the exosome and and/or the vaccine composition for use in the prevention or amelioration of an infection in a subject. Furthermore, the invention relates to a method of preventing or ameliorating an infection in a subject, the method comprising providing the subject in need thereof with a therapeutically effective amount of an exosome and/or a vaccine composition of the invention. Background SARS-CoV-2 is a recently emerging coronavirus responsible for the Covid-19 epidemic. The virus causes significant pathology, in some cases death, in a fraction of patients, in particular those with associated comorbidities and has an unprecedented gradient of severity in relation to older age. Severity may extend from extremely mild (or even asymptomatic) to life-threatening. The virus binds to the ACE2 receptor in cells of the airways, including pneumocytes and alveolar macrophages. It infects and destroys alveoli in the lungs. Mild cases may make neutralising antibodies that stop the virus early in its development in the individual. One way it becomes problematic is by interfering with the production of interferon. Disease may be due to the body’s response to the pathogen, specifically over-reaction of immunity. If inflammation and tissue damage become too severe the infected person may die or end up with significant organ damage, not only to the lungs. Multiple organs may be affected by mechanisms such as excessive clotting. The SARS-CoV-2 pandemic has resulted in a desperate need for a vaccine against the virus. However, it has also highlighted a desperate need for new vaccine approaches that may be useful in the context of other pathogens, not just SARS-CoV- 2. The present invention aims to address some of these problems. Summary of Invention In one aspect, provided herein is a fusion protein comprising: i) an exosomal protein, wherein the exosomal protein is a tetraspanin protein; and ii) one or more immunogenic protein. In an embodiment, the tetraspanin is selected from the group consisting of CD63, CD9, and CD81 or a fragment, or variant thereof. In an embodiment, the one or more immunogenic protein is capable of eliciting an immune response against a SARS-CoV-2 protein. In an embodiment, the SARS-CoV-2 protein is selected from the group consisting of spike protein (S), nucleoprotein (N), membrane protein (M), envelope protein (E), an open reading frame (ORF) 1ab, ORF 3a protein, ORF 3b protein, ORF 6 protein, ORF 7a protein, ORF 7b protein, ORF 8 protein, ORF 9b protein, ORF 9c protein, and ORF 10 protein, optionally wherein when the SARS-CoV-2 protein is spike protein (S) it is the extracellular domain of spike protein (S). In an embodiment, the one or more immunogenic protein is capable of eliciting a B- cell and a T-cell immune response. In an embodiment, the one or more immunogenic protein is capable of eliciting a T- cell immune response. In an embodiment, the protein comprises a first one or more immunogenic protein capable of eliciting a B-cell and a T-cell immune response and a second one or more immunogenic protein capable of eliciting a T-cell immune response. In an embodiment, the first one or more immunogenic protein capable of eliciting a B- cell and T-cell immune response is incorporated into a luminal loop of the exosomal protein. In an embodiment, the second one or more immunogenic protein capable of eliciting a T-cell immune response is incorporated near or at the N-terminus of the exosomal protein, and/or is incorporated into an intravesicular loop of the exosomal protein. In an embodiment, the first one or more immunogenic protein is capable of eliciting a B-cell and T-cell immune response against the SARS-CoV-2 spike protein (S), optionally against the extracellular domain of spike protein (S). In an embodiment, the second one or more immunogenic protein is capable of eliciting a T-cell immune response against the SARS-CoV-2 protein selected from the group consisting of nucleoprotein (N), membrane protein (M), envelope protein (E), ORF 1ab polyprotein, ORF 3a protein, ORF 3b protein, ORF 6 protein, ORF 7a protein, ORF 7b protein, ORF 8 protein, ORF 9b protein, ORF 9c protein, and ORF 10 protein, optionally wherein when the SARS-CoV-2 protein is spike protein (S) it is the extracellular domain of spike protein (S). In an embodiment, the first one or more immunogenic protein capable of eliciting a B- cell and a T-cell immune response is incorporated into the first and/or second luminal loop of the tetraspanin, optionally wherein the tetraspanin is CD63. In an embodiment, the second one or more immunogenic protein capable of eliciting a T-cell immune response is incorporated near or at the N-terminus of the tetraspanin, optionally wherein the tetraspanin is CD63. In an embodiment, the one or more immunogenic protein shares at least 75% identity with a SARS-CoV-2 protein or protein variant, or a fragment thereof. Optionally, when the SARS-CoV-2 protein is spike protein (S), the fragment is the receptor binding domain (RBD) (SEQ ID NO: 4). Optionally wherein the SARS-CoV-2 protein is membrane protein (M) the fragment is according to SEQ ID NO: 57 or 72. Optionally, wherein the SARS-CoV-2 protein is nucleoprotein (N) the fragment is according to SEQ ID NO: 56 or SEQ ID NO: 71. In an embodiment, the SARS-CoV-2 protein is selected from the group consisting of the extracellular domain of spike protein (S) (SEQ ID NO: 1), nucleoprotein (N) (SEQ ID NO: 2), membrane protein (M) (SEQ ID NO: 3), envelope protein (E) (SEQ ID NO: 46), ORF 3a protein (SEQ ID NO: 47), ORF 3b protein (SEQ ID NO: 48), ORF 6 protein (SEQ ID NO: 49), ORF 7a protein (SEQ ID NO: 50), ORF 7b protein (SEQ ID NO: 51), ORF 8 protein (SEQ ID NO: 52), ORF 9b protein (SEQ ID NO: 53), ORF 9c protein (SEQ ID NO: 54), ORF 10 protein (SEQ ID NO: 55), and ORF1ab polyprotein (SEQ ID NO: 73). In an embodiment, the fusion protein shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO 65, SEQ ID NO: 66, SEQ ID NO: 68, or SEQ ID NO:69. In one embodiment, the fusion protein shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% with SEQ ID NO: 5 or SEQ ID NO: 6. Suitably, the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 5. Suitably, the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 6. More suitably, the fusion protein is according to SEQ ID NO: 5 or SEQ ID NO: 6. In one embodiment, the fusion protein shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% with SEQ ID NO: 59 or SEQ ID NO: 60. Suitably, the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 59. Suitably, the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 60. More suitably, the fusion protein is according to SEQ ID NO: 59 or SEQ ID NO: 60. In one embodiment, the fusion protein shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% with SEQ ID NO: 65 or SEQ ID NO: 66. Suitably, the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 65. Suitably, the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 66. More suitably, the fusion protein is according to SEQ ID NO: 65 or SEQ ID NO: 66. In one embodiment, the fusion protein shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% with SEQ ID NO: 68 or SEQ ID NO: 69. Suitably, the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 68. Suitably, the fusion protein shares at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 69. More suitably, the fusion protein is according to SEQ ID NO: 68 or SEQ ID NO: 69. In an embodiment, the exosomal protein and the one or more immunogenic protein are separated by a linker sequence. In one aspect, provided herein is an exosome comprising the fusion protein of the invention or a membrane preparation comprising the fusion protein of the invention. In an embodiment, the exosome or membrane preparation comprises a DC receptor ligand and/or an immune enhancing moiety, optionally wherein the immune enhancing moiety is selected from the group consisting of a molecule with a pathogen-associated molecular pattern (PAMP), DNA oligonucleotide containing an CpG motif, a STING agonist (such as cyclic GMP-AMP), and a TLR9 (Toll-like Receptor) agonist. In one aspect, provided herein is a vaccine composition comprising an exosome or membrane preparation of the invention. In one aspect, provided herein is nucleic acid molecule encoding a fusion protein of the invention. In one aspect, provided herein is an expression vector comprising the nucleic acid molecule of the invention. In an embodiment, the vector is a lentivirus expression vector. In one aspect, provided herein is a cell comprising a fusion protein, an exosome, a membrane preparation, a nucleic acid, and/or an expression vector if the invention, optionally wherein the cell is an isolated cell. In an embodiment, the transformed cell line is a HeLa cell or a LCL721 cell. In an embodiment, the cell is modified to enhance the production of exosomes. In an embodiment, the cell has reduced expression of tetherin (Bst2). In one aspect, provided herein is an exosome, a membrane preparation, or vaccine composition of the invention, for use as a medicament. In one aspect, provided herein is an exosome, a membrane preparation, or a vaccine composition of the invention, for use in the prevention or amelioration of an infection. In an embodiment, the infection is a viral, bacterial or fungal infection. Optionally, wherein the viral infection is a SARS-CoV-2 infection. In one aspect, provided herein is a method of preventing or ameliorating an infection in a subject, the method comprising providing the subject in need thereof with a therapeutically effective amount of an exosome, a membrane preparation, or a vaccine composition of the invention. The infection may be a viral, bacterial or fungal infection. Optionally, the viral infection is a SARS-CoV-2 infection. Except for where the context requires otherwise, the considerations set out in this disclosure should be considered to be applicable to all aspects of the invention. Merely by way of example, any consideration set out with reference to the fusion protein will be applicable to the exosome, nucleic acid molecule, expression vector, vaccine composition, cell, uses and treatment methods of the invention. Throughout the description and claims of this specification, the words “comprise” and “contain” and variations of them mean “including but not limited to”, and they are not intended to (and do not) exclude other moieties, additives, components, integers or steps. Throughout the description and claims of this specification, the singular encompasses the plural unless the context otherwise requires. In particular, where the indefinite article is used, the specification is to be understood as contemplating plurality as well as singularity, unless the context requires otherwise. Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith. Various aspects of the invention are described in further detail below. Brief Description of the Figures The invention is described in detail with reference to the following figures. Figure 1. A schematic representation of a SARS-CoV-2 virus particle (A). A schematic representation of an engineered exosome (B). Figure 2. A schematic representation of exemplary fusion polypeptides of the invention (A). A schematic representation of the orientation of luminal and intravesicular loops for native CD63 protein and CD63 fusion protein (B). Schematic representation of CD63 fusion constructs (C). Figure 3. A schematic representation of an exemplary expression vector of the invention. Figure 4. Shows the results of a restriction digest. Minipreps 1+3 were both found to have the correct sized insert by test digest, and the CD63-FLAG-SpikeRBD was confirmed by sanger sequencing (SEQ ID NO: 19 to 22 - restriction sites highlighted in red. Start codon in bold and italics. CD63 sequence underlined. FLAG in bold text. Linkers are italics. SpikeRBD in underlined text). Figure 5. A photo showing immunofluorescence assay results. To check the FLAG- CD63Spike insert was not disrupting CD63 trafficking, pLVX-EF1alpha-CD63-FLAG- SpikeRBD-IRES-Puro DNA was transiently transfected to HeLa cells and subject to immunofluorescence. Figure 6. An image showing immunofluorescence results. FLAG signal was confirmed to be in CD63-positive compartments as shown by co-localisation of signal, and CD63 localisation remained in punctate organelles in transfected cells (as is also observed in non-transfected cells). Figure 7. An image showing immunofluorescence assay results. An antibody against SpikeRBD showed similar pattern of staining, and colocalised with FLAG – confirming that Spike and FLAG are together localised in CD63-positive, late endosomal compartments. Together, these data indicate that Spike RBD and FLAG are localised to late endocytic compartments as expected. Figure 8. An image showing Western Blot results. Whole cell lysates were collected from untransduced and transduced WT and Bst2KO HeLa cells. Lysates were probed with antibodies against CD63, and both transduced cell lines displayed enhanced staining, and at a higher molecular weight – reflecting additional molecular mass given from the addition of FLAG-SpikeRBD (A). Antibodies against FLAG were similarly used to blot whole cell lysates, and a positive band was observed at the correct molecular weight in transduced stable cell lines, and not in untransduced controls. Exosome-enriched fractions were generated from cells and subject to Western blotting. Exosomes from tetherin KO HeLa cells + CD63-FLAG-SpikeRBD displayed a band at the correct molecular weight as expected (B). Figure 9. Electron microscopy images confirming that exosomes from transduced cells contained FLAG-SpikeRBD. An antibody against FLAG displays labelling specifically to exosomes. Labelling was not present in untransduced cells. Figure 10. An image showing immunofluorescence assay results confirming successful expression of N and sRBD sequences. Stable HeLa cell lines were established through antibiotic selection using plasmids encoding FLAG-tagged- fusions to CD63 scaffold. As can be seen from the figure, the investigated fusion proteins did not disrupt CD63 trafficking as confirmed by localization of the FLAG staining with CD63-positive compartments in stable cells. Figure 11. An image showing immunofluorescence assay results further confirming successful expression of N and sRBD sequences on the inside and outside of CD63. Expression remained high for several weeks. Figure 12. An image showing flow cytometry results. Transduced and untransduced cell lines were collected and analysed with anti-FLAG and anti-CD63 antibodies. The fluorescence intensities of FLAG and CD63 were analysed by flow cytometry. Transduced cell lines (comprising v16 and v19) displayed enhanced CD63 levels compared to the mock untransduced cells. Additionally, surface FLAG staining was observed in transduced v16 cell lines where FLAG is located on the outside of CD63. In v19 transduced cell lines, cells were fixed and permeabilised to reveal internal FLAG, confirming correct topology in cell lines. Figure 13A. A graph showing the fold change of CD63 levels in exosomes containing the three different constructs (V1, V19, and V16). The highest levels of CD63 were found in exosomes comprising the fusion protein V19, which contains two immunogenic proteins. Figure 13B Characterization of exosomes containing three different constructs (V1, V19, and V16). A flow nanoanalyzer (NanoFCM), a cytometer specifically designed to analyse particles smaller than the wavelength of visible light, was used to measure size and concentrations of isolated exosomes. Overall, the particles had an average modal diameter ranging between 62-69 nm, which is within the accepted range for exosomes. It can be seen from this figure that, exosomes with the V16 construct containing both N and sRBD domains were the largest. Figure 14. A schematic illustrating the principles of the assay described in Example 2. Figure 15. Illustrates the results of the T-cell activation described in Example 2. The results show that exosomes and membrane preparations containing the constructs V1, 19, and V16 can elicit recognition by T-cells specific to epitopes with SARS-CoV-2 Spike or N. Figure 16. Graphs illustrating T cell responses to SARS-CoV2. Fig 16A shows that PBMCs isolated 2-6 months post infection from most symptomatic patients with confirmed SARS-CoV-2 infection respond to in vitro stimulation with SARS-CoV-2 peptides derived from the dominant viral antigens S, M and N 24 (shown by IFNγ ELISPOT). Fig 16B and F shows that the inventors have established polyclonal T cell cultures from these donors by expansion with peptide cocktails. Epitope mapping using 15-mers overlapping by 10 amino acids confirmed that the different T cell lines were specific for numerous epitopes within S (Fig 16B), M and N. Autologous B cell derived lymphoblastoid cell lines (LCL) from the respective donors were established as antigen presenting cells (APC) that can be pulsed with either viral proteins, peptides, or exosome vaccines. Fig 16C shows a dose-dependent stimulation of S-specific polyclonal T cells co-cultured with autologous LCL exposed to FLAG-SRBD-CD63 exosomes. By ELISA, a >60% maximal IFNγ response was induced, but no measurable response to control exosomes. Fig 16D shows that exosomes with sRBD (V1 construct) stimulate 2 independent CD4 S specific clones. Fig 16E shows that multiple clones with exquisite specificity for defined epitopes within S, M and N have been isolated. Several are restricted by common alleles such as DRB1*01:01 or DRB4* 01:03 expressed in ~10-30% of the UK population. Detailed Description of the Invention The present invention is based on the inventors’ development of fusion proteins that may be useful in the context of vaccines, in particular anti-SARS-CoV-2 vaccines. As shown in the examples section of the present description, the inventors have developed fusion proteins that may have the ability to strongly activate T-cells rendering the fusion proteins especially useful in the context of vaccines. The fusion proteins of the invention comprise an exosomal protein, which enables it to be localised on exosomes. Importantly, the inventors have shown that the fusion proteins of the invention do not prevent exosomal biogenesis and/or processing, allowing for exosomes expressing the fusion proteins to be released by cells to which a vector encoding the fusion proteins has been provided. Exosomes comprising the fusion proteins of the invention may be provided to a subject and elicit an immune response that may prevent or ameliorate the symptoms and disease associated with a pathogen infection, such as for example SARS-CoV-2. Without wishing to be bound by this hypothesis, the inventors believe that providing a subject with exosomes comprising the fusion proteins of the invention may be associated with a number of advantages compared to other vaccination approaches currently investigated. For example, exosomes have the ability to taken up by antigen presenting cells or directly fuse to the plasma membrane of antigen presenting cells. This may be beneficial because it may enable efficient uptake of antigens to cells key to immune stimulation (such as dendritic cells). Exosomes may be packed with a payload of multiple viral antigens allowing the stimulation of B-cell immune and/or T- cell immune responses. Indeed, as explained in more detail hereinbelow, some of the fusion proteins of the invention comprise immunogenic proteins capable of eliciting B- cell immune and/or T-cell immune responses. Fusion protein The terms “fusion protein” and “fusion polypeptide” are used herein interchangeably, and refer to an artificial protein comprising amino acid sequences of at least two different proteins or protein variants, or fragments thereof. The fusion protein of the invention comprises an exosomal protein and one or more immunogenic protein. The term “exosomal protein” as used herein refers to any polypeptide that is enriched in a vesicular structure. The exosomal protein may therefore be utilised to transport the fusion protein of the invention to the vesicular structure. Suitably, the vesicular structure is an exosome, suitably a human exosome. Accordingly, the exosomal protein may be a human exosomal protein. Suitably, the exosomal protein is a tetraspanin. Suitably, the tetraspanin is selected from the group consisting of CD63 (SEQ ID NO: 43), CD9 (SEQ ID NO: 44) and CD81 (SEQ ID NO: 45) or a fragment, or variant thereof. Suitably the tetraspanin variant may share at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, identity with SEQ ID NO: 43, SEQ ID NO: 44 or SEQ ID NO: 45. Suitably the tetraspanin variant may share at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, identity with SEQ ID NO: 43, SEQ ID NO: 44 or SEQ ID NO: 45. More suitably the tetraspanin variant may share at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, identity with SEQ ID NO: 43. Suitably the tetraspanin fragment may comprise or consist of at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% contiguous amino acids of SEQ ID NO: 43, SEQ ID NO: 44 or SEQ ID NO: 45. More suitably, the fragment may comprise or consist of at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% contiguous amino acids of SEQ ID NO: 43, SEQ ID NO: 44 or SEQ ID NO: 45. More suitably, the fragment may comprise or consist of at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% contiguous amino acids of SEQ ID NO: 43. Most suitably, the tetraspanin protein is CD63 (SEQ ID NO: 43). Similarly to other tetraspanins, CD63 protein has four transmembrane domains, and three loops. Two of the loops are luminal loops, meaning that they extend outward from the vesicular structure (such as exosome). One of the loops is intravesicular, meaning that it extends into the vesicular structure (such as exosome). In the present specification, in the context of tetraspanins, (for example CD63), the luminal loop that is closer to the N-terminal of the protein is referred to as the first (I) luminal loop, and the luminal loop that is closer to the C-terminal of the protein is referred to as the second (II) luminal loop. Suitably, amino acids 33 to 51 of the human CD63 protein (SEQ ID NO: 43) define the I luminal loop. Suitably, amino acids 103 to 203 of the human CD63 protein (SEQ ID NO: 43) define the II luminal loop. The term “immunogenic protein” as used herein refers to a polypeptide that is capable of eliciting an immune response. Suitably, the immunogenic protein may be capable of eliciting a protective immune response in a host. Such a protective immune response may establish or improve the host’s immunity to a pathogen. Suitably, the pathogen may be selected from the group consisting of a virus, a bacterium, and a fungus. More suitably the virus may be SARS-CoV-2. Suitably, the host may be a mammal, preferably human. The immunogenic protein may elicit a B-cell and/or T-cell immune response. The phrase "elicit a B-cell immune response” as used herein refers to the immunogenic protein’s ability to trigger a humoral immune response, meaning that the immunogenic protein may cause cells of the subject’s immune system to produce antibodies that bind specific protein targets. Suitably, the protein targets may correspond to the immunogenic protein. For example, if the protein target to which the antibodies are produced is SARS-CoV-2 spike protein (S), variant, or fragment thereof, the immunogenic protein may have a sequence corresponding to that SARS-CoV-2 spike protein (S) or variant, or fragment thereof. The phrase "elicit a B-cell immune response” as used herein refers to the immunogenic protein’s ability to trigger a cellular immune response, meaning that the immunogenic protein may trigger the selection, expansion and maturation of B-cells producing antibodies capable of binding the antigen. Some of these antibodies are neutralizing antibodies that are capable of blocking the infection of host cells by the SARS-CoV-2 virus. The phrase "elicit a T-cell immune response” as used herein refers to the immunogenic protein’s ability to trigger a cellular immune response, meaning that the immunogenic protein may trigger the selection and expansion of specific helper and/or cytotoxic T- lymphocytes capable of directly eliminating the cells that contain the antigen. A T-cell immune response may be initiated when the immunogenic protein is presented as an antigen on the surface of the exosome. It will be appreciated that whether the immunogenic protein elicits a B-cell and/or T- cell immune response may depend upon the immunogenic protein’s location with respect to the exosomal protein. As explained in more detail elsewhere in the present specification, by way of example, immunogenic proteins incorporated into a luminal loop of an exosomal protein, may elicit a B-cell and T-cell immune response. Notably, antigens designed to elicit a B-cell response are constructed in the luminal loop of the fusion protein in order to orient the antigen epitope on the outside (luminal) of the exosome. Immunogenic proteins incorporated into intravesicular loops, or at or near the N-terminal of an immunogenic protein may elicit a T-cell immune response. “At the N-terminal” as used herein means attached to the first amino acid of a sequence encoding a protein that would normally have a free amine group. “Near the N-terminal” as used herein means attached to an amino acid that is several amino acids away from the first amino acid of a sequence encoding a protein. For example, the immunogenic protein may be referred to as being near the N-terminal if it is attached to an amino acid located 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more amino acids away from the first amino acid of a sequence encoding a protein. Suitably, the immunogenic protein may be capable of eliciting a B-cell and/or T-cell immune response against a SARS-CoV-2 protein, variant, or fragment thereof. Suitably, the SARS-CoV-2 protein against which the immunogenic protein elicits an immune response may be selected from the group consisting of spike protein (S), nucleoprotein (N), membrane protein (M), envelope protein (E), ORF 1ab polyprotein, ORF 3a, ORF 3b protein, ORF 6 protein, ORF 7a protein, ORF 7b protein, ORF 8 protein, ORF 9b protein, ORF 9c protein and ORF 10 protein. The spike protein (S) plays a key role in the receptor recognition and cell membrane fusion process, and is composed of two subunits, S1 and S2. The S1 subunit contains a receptor-binding domain (RBD) that recognises and binds to the host receptor angiotensin-converting enzyme 2 (ACE2), while the S2 subunit mediates viral cell membrane fusion by forming a six-helical bundle via the two-heptad repeat domain. The nucleoprotein (N) (also known as the nucleocapsid protein) packages the positive strand viral genome RNA into a helical ribonucleocapsid (RNP) and plays a fundamental role during virion assembly through its interactions with the viral genome and membrane protein (M). The membrane protein (M) is a component of the viral envelope that plays a central role in virus morphogenesis and assembly via its interactions with other viral proteins. The envelope protein (E) plays a central role in virus morphogenesis and assembly. The protein acts as a viroporin and self-assembles in host membranes forming pentameric protein-lipid pores that allow ion transport. It also plays a role in the induction of apoptosis. The ORF 1ab gene encodes 16 structural proteins, which include an RNA polymerase co-factor non-structural protein 7 (NSP7), RNA polymerase (NSP12), and helicase NSP13. These proteins are highly conserved amongst beta coronaviruses due to their key roles in the viral life cycle. The term “ORF 1ab polyprotein” includes proteins selected from the group consisting of: NSP7, NSP12, and NSP13, or fragments thereof. The ORF 3a protein Forms homotetrameric potassium sensitive ion channels (viroporin) and may modulate virus release. The ORF 3b protein is potent interferon antagonist, suppressing the induction of type I interferon. The ORF6 protein disrupts cell nuclear import complex formation by tethering karyopherin alpha 2 and karyopherin beta 1 to the membrane. The ORF 7a protein plays a role as antagonist of host tetherin (BST2), disrupting its antiviral effect. It acts by binding to BST2 thereby interfering with its glycosylation. The ORF 7a protein may also facilitate the disassembly of the Golgi apparatus. The ORF 7b protein is transmembrane protein postulated to have the potential to interfere with important cellular processes that involve leucine-zipper formation by multimerizing with phospholamban and with E-cadherins. The ORF 8 protein is believed to play a role in modulating host immune response and may play a role in blocking host IL17 cytokine by its interaction with host IL17RA. The ORF 9b protein plays a role in the inhibition of host innate immune response by targeting the mitochondrial-associated adapter MAVS. The ORF 9c protein is postulated to enhance the host proteasome activity thereby affect immune evasion, virulence and pathogenesis of the SARS-CoV2 virus. The ORF 10 protein is postulated to bind Collin ubiquitin ligase complex and hijack it for ubiquitination and degradation of restriction factors. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 protein may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 protein, variant, or a fragment thereof. Thus, suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 spike protein (S) may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 spike protein (S), variant, or a fragment thereof. Suitably, in the context of the spike protein (S), the fragment may comprise or consist of the extracellular domain of the spike protein (S). Accordingly, the immunogenic protein may have an amino acid sequence that shares at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with the extracellular domain of the spike protein (S) or variant, or fragment thereof. Suitably, in the context of spike protein (S), the fragment may comprise or consist of the receptor binding domain (RBD). Accordingly, the immunogenic protein may have an amino acid sequence that shares at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with the receptor binding domain (RBD) (SEQ ID NO: 4), or variant, or fragment thereof. More suitably, the fragment is according to SEQ ID NO: 4. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 nucleoprotein (N) may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 nucleoprotein (N) (SEQ ID NO: 2), variant, or a fragment thereof. Suitably, the fragment may share at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with SEQ ID NO: 56 or SEQ ID NO: 71. More suitably, the fragment is according to SEQ ID NO: 56 or SEQ ID NO: 71. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 membrane protein (M) may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 membrane protein (M) (SEQ ID NO: 3), variant, or a fragment thereof. Suitably, the fragment may share at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with SEQ ID NO:57 or 72. More suitably, the fragment is according to SEQ ID NO: 72. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 envelope protein (E) may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 envelope protein (E), variant, or a fragment thereof. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF3a may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF3a, variant, or a fragment thereof. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF3b may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF3b, variant, or a fragment thereof. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF6 may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF6, variant, or a fragment thereof. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF7a may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF7a, variant, or a fragment thereof. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF7b may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF7b, variant, or a fragment thereof. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF8 may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF8, variant, or a fragment thereof. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF9b may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF9b, variant, or a fragment thereof. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF9c may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF9c, variant, or a fragment thereof. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF10 may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF10, variant, or a fragment thereof. Suitably, the immunogenic protein capable of eliciting an immune response against a SARS-CoV-2 ORF1ab may have an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with a SARS-CoV-2 ORF1ab, variant, or a fragment thereof. Sequences of the above-mentioned proteins are provided under the section “Sequences” of the present disclosure. Suitably, the fusion protein may comprise one, two, three, or more immunogenic proteins. Suitably, the fusion protein may comprise one immunogenic protein. Suitably, the fusion protein may comprise two immunogenic proteins. Suitably, the fusion protein may comprise three immunogenic proteins. In an embodiment where the fusion protein comprises one immunogenic protein, the immunogenic protein may elicit a B-cell and a T-cell immune response, or a T-cell immune response. As mentioned elsewhere in the present specification, the type of immune response elicited may depend on the localisation of the immunogenic protein in the exosome. Suitably, the immunogenic protein that elicits a B-cell and a T-cell immune response, elicits a response against the SARS-CoV-2 spike protein (S). Suitably, the immunogenic protein that elicits a T-cell response, elicits a response against the nucleoprotein (N) or membrane protein (M). In an embodiment where the fusion protein comprises two or more immunogenic proteins, it will be appreciated that some or all of the immunogenic proteins may be the same or different. By different it is meant that the immunogenic proteins may have a sequence corresponding to different proteins, or a sequence that corresponds to different parts of the same protein (the different parts may be overlapping or non- overlapping). Merely by way of example, in an embodiment where the fusion protein comprises two different immunogenic proteins, the first immunogenic protein may elicit a B-cell and a T-cell immune response, and the second immunogenic protein may elicit a T-cell immune response. In such an example, the immunogenic protein that elicits a B-cell and a T-cell immune response, may elicit a response against the SARS-CoV-2 spike protein (S). The immunogenic protein that elicits a T-cell response, may elicit a response against the nucleoprotein (N) or membrane protein (M). SEQ ID NO: 17, 59, or 62 are exemplary amino acid sequences of such proteins. Merely by way of another example, in an embodiment where the fusion protein comprises two different immunogenic proteins, each one of the immunogenic proteins may elicit a T-cell response. In such an example, the first immunogenic protein may elicit a response against the SARS-CoV-2 nucleoprotein (N), whereas the second immunogenic protein elicits a response against membrane protein (M). SEQ ID NO: 13, 17, or 62 are exemplary amino acid sequences of such proteins. Merely by way of another example, in an embodiment where fusion protein comprises three different immunogenic proteins, it may comprise one or two immunogenic proteins that elicit a T-cell immune response, and one or two immunogenic proteins that elicit a B-cell and T-cell immune response. For example, the fusion protein may comprise two immunogenic proteins that elicit a T-cell immune response against the nucleoprotein (N) or membrane protein (M), and an immunogenic protein that elicits a B-cell and T-cell immune response against SARS-CoV-2 spike protein (S). As mentioned elsewhere in the present specification, the type of immune response elicited by the immunogenic protein may be dictated by the immunogenic protein’s location with respect to the exosomal protein. Immunogenic proteins incorporated into a luminal loop of an exosomal protein (for example a tetraspanin, such as CD63), may elicit a B-cell and a T-cell immune response. Suitably, the immunogenic protein that is incorporated into the luminal loop may be one that elicits a response against SARS-CoV-2 spike protein (S). Such an immunogenic protein may be incorporated into the first and/or second luminal loop. Suitably, in the context of CD63, the immunogenic protein may be incorporated between amino acids Q36 and L37 (thereby incorporated into loop I) and/or between amino acids R108-D109 (thereby incorporated into loop II). Immunogenic proteins incorporated into intravesicular loops, or at or near the N- terminal of an exosomal protein (for example a tetraspanin, such as CD63), may elicit a T-cell immune response. Suitably, an immunogenic protein that is incorporated into the intravesicular loop and/or at or near the N-terminal of an exosomal protein may be one that elicits a response against SARS-CoV-2 nucleoprotein (N) or membrane protein (M). The fusion protein of the invention may comprise a linker. The linker may be between the immunogenic protein and exosomal protein sequence. Suitably, the linker may be on one or both side of the immunogenic protein. When the fusion protein comprises more than one immunogenic protein, a linker may be present between some, or all, of the immunogenic proteins and exosomal protein sequence. Suitably, when the fusion protein comprises two or more immunogenic proteins adjacent to one another, there may be a linker between some, or all, of the immunogenic proteins. Suitably the linker may be 20 or fewer amino acids in length. Suitably, the linker may be about 18, about 16, about 14, about 12 or about 10 amino acids in length. Suitably the linker may be about 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2 or 1 amino acid in length. More suitably the linker may be from 6 to 2 amino acids long. Suitably the linker may be a flexible or a rigid linker. More suitably the liker is flexible. Merely by way of example a flexible linker may have the sequence GGSG, GGGSG, GGGGS, GGG, or GG. Other suitable linkers will be known to those skilled in the art. It will be appreciated that the linker may be used to enable or enhance correct immunogenic protein folding and/or stability. Table 1 Exemplary fusion polypeptides of the invention
Figure imgf000023_0001
*it will be appreciated that the presence of FLAG is optional in each of these exemplary fusion protein arrangements. In a suitable embodiment, the fusion protein comprises CD63 and the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof. Suitably, the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof is incorporated into the first luminal of CD63. Suitably, the extracellular domain of the spike protein (S), variant, or fragment thereof, may be incorporated between amino acids Q36 and L37 of CD63. Suitably the extracellular domain of the SARS- CoV-2 spike protein (S) may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1. Suitably the fragment may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4. Suitably, a linker may be incorporated between both sides of the immunogenic protein and exosomal protein. In a suitable embodiment, the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7 or SEQ ID NO:8. More suitably, the fusion protein comprises or consists of SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7 or SEQ ID NO:8. In a suitable embodiment, the fusion protein comprises CD63 and the extracellular domain of the SARS-CoV-2 spike (S) protein or a fragment thereof. Suitably, the extracellular domain of the SARS-CoV-2 spike (S) protein or a fragment thereof is incorporated into the second luminal of CD63. Suitably, the extracellular domain of the spike protein (S), variant, or fragment thereof may be incorporated between amino acids R108 and D109 of CD63. Suitably the extracellular domain of the SARS-CoV-2 spike (S) protein have a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1. Suitably the fragment may have a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4. Suitably, a linker may be incorporated between both sides of the immunogenic protein and exosomal protein. In a suitable embodiment, the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:15 or SEQ ID NO:16. More suitably, the fusion protein comprises or consists of SEQ ID NO:15 or SEQ ID NO:16. In a suitable embodiment, the fusion protein comprises CD9 and the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof. Suitably, the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof is incorporated into the first luminal of CD9. Suitably, the extracellular domain of the spike protein (S), variant, or fragment thereof may be incorporated between amino acids N50 and N51 of CD9. Suitably the extracellular domain of the SARS-CoV- 2 spike protein (S) may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1. Suitably the fragment may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4. Suitably, a linker may be incorporated between both sides of the immunogenic protein and exosomal protein. In a suitable embodiment, the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:23 or SEQ ID NO:24. More suitably, the fusion protein comprises or consists of SEQ ID NO:23 or SEQ ID NO:24. In a suitable embodiment, the fusion protein comprises CD81 and the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof. Suitably, the extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof is incorporated into the first luminal of CD81. Suitably, the extracellular domain of the spike protein (S), variant, or fragment thereof may be incorporated between amino acids L49 and G50 of CD81. Suitably the extracellular domain of the SARS- CoV-2 spike protein (S) may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1. Suitably the fragment may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4. Suitably, a linker may be incorporated between both sides of the immunogenic protein and exosomal protein. In a suitable embodiment, the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:25 or SEQ ID NO:26. More suitably, the fusion protein comprises or consists of SEQ ID NO:25 or SEQ ID NO:26. In a suitable embodiment, the fusion protein comprises CD63 and SARS-CoV-2 nucleoprotein (N) and/or membrane protein (M), or fragment thereof. Suitably, the SARS-CoV-2 nucleoprotein (N) and/or membrane protein (M), or fragment thereof is incorporated near or at the N-terminal of CD63. Suitably the SARS-CoV-2 nucleoprotein (N) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 2. Suitably the SARS-CoV-2 membrane protein (M) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 3. Suitably, the fragment of nucleoprotein (N) and has a sequence that is sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 56 or SEQ ID NO: 71. Suitably, the fragment of membrane protein (M) has a sequence that is sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 57, or SEQ ID NO: 72, more suitably SEQ ID NO: 72. Suitably, a linker may be incorporated between the immunogenic protein and the exosomal protein. Suitably, when both the nucleoprotein (N) and membrane protein (M) are incorporated into the fusion protein the membrane protein (M) may be incorporated at the N-terminal of the exosomal protein, followed by the nucleoprotein (N). In such an embodiment, the nucleoprotein (N) and membrane protein (M) may or may not be separated by a linker. In a suitable embodiment, the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 68 or SEQ ID NO:69. More suitably, the fusion protein comprises or consists of SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 68 or SEQ ID NO:69. In a suitable embodiment, the fusion protein comprises CD63 and SARS-CoV-2 nucleoprotein (N) or membrane protein (M), or fragment thereof. Suitably, the SARS- CoV-2 nucleoprotein (N) or membrane protein (M), or fragment thereof is incorporated near or at the N-terminal of CD63. Suitably the SARS-CoV-2 nucleoprotein (N) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 2. Suitably, the fragment of nucleoprotein (N) and has a sequence that is sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 56 or SEQ ID NO: 71. Suitably the SARS-CoV-2 membrane protein (M) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 3. Suitably, the fragment of membrane protein (M) has a sequence that is sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 57 or SEQ ID NO: 72, more suitably SEQ ID NO: 72. Suitably, a linker may be incorporated between the immunogenic protein and the exosomal protein. In a suitable embodiment, the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 68, or SEQ ID NO: 69. More suitably, the fusion protein comprises or consists of SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 68, or SEQ ID NO: 69. In a suitable embodiment, the fusion protein comprises CD63 and the extracellular domain of the SARS-CoV-2 spike (S) protein or a fragment thereof, and nucleoprotein (N) and/or membrane protein (M), or fragment thereof. Suitably, the extracellular domain of the SARS-CoV-2 spike (S) protein or a fragment thereof is incorporated into the first or second luminal of CD63. Suitably, the extracellular domain of the SARS- CoV-2 nucleoprotein (N) and/or membrane protein (M), or fragment thereof is incorporated near or at the N-terminal of CD63. Suitably the extracellular domain of the SARS-CoV-2 spike (S) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1. Suitably the fragment may have a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4. Suitably the SARS-CoV-2 nucleoprotein (N) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 2. Suitably, the fragment of nucleoprotein (N) and has a sequence that is sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 56 or SEQ ID NO: 71. Suitably the SARS-CoV-2 membrane protein (M) protein has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 3. Suitably, the membrane protein (M) fragment has a sequence sharing at least a 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 57, or SEQ ID NO: 72. In a suitable embodiment, the fusion protein comprises or consists of a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, or SEQ ID NO: 63. More suitably, the fusion protein comprises or consists of SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, or SEQ ID NO: 63. In a suitable embodiment, the fusion protein comprises CD63 and SARS-CoV-2 membrane protein (M), variant, or a fragment thereof, and extracellular domain of the SARS-CoV-2 spike protein (S), variant, or a fragment thereof. Suitably, the fragment of the membrane protein (M) may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 57 or 72. More suitably, the fragment is according to SEQ ID NO: 72. Suitably, the membrane protein (M), variant or fragment is incorporated into a luminal loop of CD63. Suitably the extracellular domain of the SARS-CoV-2 spike protein (S) may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 1. Suitably the fragment of the spike protein (S) may have a sequence sharing at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with SEQ ID NO: 4. Suitably, the fragment of the spike protein (S) is according to SEQ ID NO: 4. Suitably, a linker may be incorporated between both sides of the immunogenic protein and exosomal protein. As used herein, the terms “homology” and “identity” are used interchangeably. Calculations of sequence homology or identity between sequences may be performed as follows. To determine the percent identity of two amino acid sequences, or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). In a preferred embodiment, the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, and even more preferably at least 70%, 75%, 80%, 82%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% of the length of the reference sequence. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid “homology”). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. In a preferred embodiment, the percent identity between two amino acid sequences is determined using the Needleman et al. (1970) J. Mol. Biol.48:444-453) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a BLOSUM 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6. In yet another preferred embodiment, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (available at http://www.gcg.com), using a NWSgapdna. CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6. A particularly preferred set of parameters (and the one that should be used if the practitioner is uncertain about what parameters should be applied to determine if a molecule is within a sequence identity or homology limitation of the invention) are a BLOSUM 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5. Alternatively, the percent identity between two amino acid or nucleotide sequences can be determined using the algorithm of Meyers et al. (1989) CABIOS 4:11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. Other methods of determining sequence identity will be known to those skilled in the art. As mentioned elsewhere in the present specification, the fusion proteins of the invention are artificial proteins comprising amino acid sequences of at least two different proteins or protein variants, or fragments thereof. Specifically, the fusion proteins of the invention comprise an exosomal protein or an exosomal protein variant, or a fragment thereof, and one or more immunogenic protein or immunogenic protein variant, or fragment thereof. It will be appreciated that in the context of the present speciation, when reference is made to a “protein”, for example an exosomal protein or immunogenic protein, this also refers to the protein variant, or fragment of the protein or protein variant. The term “variant” as used herein refers to a polypeptide that shares a partial sequence identity with the amino acid encoding the exosomal protein or the immunogenic protein. The term “fragment” as used herein refers to a polypeptide that shares 100% identity with the corresponding portion of the amino acid sequence of the exosomal or immunogenic protein or variant, but that consist of a truncation in the amino acid sequence encoding the exosomal protein (for example a tetraspanin, such as CD63) or immunogenic protein (for example a protein selected from the group consisting of spike protein (S), nucleoprotein (N), and membrane protein (M) of SARS- CoV-2). Exosomes In one aspect the present invention provides exosomes comprising the fusion proteins of the invention. An “exosome”, as used herein, is a membrane-bound extracellular vesicle between 30-150 nanometers in size. In vivo, exosomes are shed into the blood, urine, cerebrospinal fluid and other bodily fluids from cells, and may contain some of the proteins and functional ribonucleic acid (RNA) molecules from their cells of origin. In vitro, exosomes may be shed into the cell culture media. The cells of origin as per the present invention may be selected from a wide range of cells. Merely by way of example the cells may be HeLa cells. Methods of purifying exosomes from cell culture media are well known in the art. Merely by way of example, exosomes may be purified using the Total Exosome Isolation Reagent (InvitrogenTM) following the manufacturer’s instructions. In the context of vaccines, exosomes may be advantageous because they can be efficiently taken up by dendritic cells (DCs). DCs are proficient at antigen processing and presentation and promotion of an active and beneficial T-cell immune response. Antigens (such as the immunogenic proteins of the fusion protein of the invention) taken up by DCs, as well as unprocessed antigens, travel through the lymph to the nearest draining node to stimulate antigen specific T-cells. This leads to a cellular response, invoking both CD8 cytotoxic and CD4 helper T-cells, as well as humoral responses leading to production of specific antibodies. In some cases, this process may be made even more efficient by enhancing targeting of the exosomes to DCs. DCs express receptors on their surface, including C-type lectin receptor, mannose receptor, DEC-205 or Dectin-1 that trigger DC activation. To facilitate this process ligands for these receptors may be expressed on the exosomes. Accordingly, the exosomes of the invention may further comprise a DC receptor ligand. The ligand may be a natural biological ligand, or a nanobody or related structure designed to bind to the receptor. It will be appreciated that the ligand may be encoded by a polypeptide forming part of the fusion protein, or a separate polypeptide. Suitably, the fusion protein of the invention may further comprise a polypeptide encoding a DC receptor ligand. In a suitable embodiment, the exosomes of the invention may comprise an immune enhancing moiety. Merely by way of example, such an immune enhancing moiety may be selected from the group consisting of a molecule with a pathogen-associated molecular pattern (PAMP), DNA oligonucleotides containing the CpG motif, a STING agonist (such as cyclic GMP-AMP) and a TLR9 (Toll-like Receptor) agonist. A molecule with a pathogen-associated molecular pattern (PAMP) may be advantageous as it may help the exosomes induce a potent Th1 response. Examples of adjuvants include synthetic CpG (a TLR9 agonist) or poly-ICLC (a TLR3 ligand). DNA oligonucleotides containing the CpG motif may be advantageous as they may help stimulate cells of the immune system leading to the production of cytokines such as type-I interferon, IL-6, TNF and IL-12. A STING agonist (such as cyclic GMP-AMP) may be advantageous as it may help stimulate cells of the immune system leading to the production of cytokine such as type I interferon. Methods for producing exosomes are well known in the art. Methods for producing exosomes expressing fusion proteins are also well known in the art. Suitably the exosomes may be produced by cells modified (for example, genetically modified) to enhance the production of exosomes. Suitably, exosomes may be produced in cells which have reduced expression of proteins which retain exosomes, such as tetherin (bone marrow stromal antigen 2) protein. By reduced expression it is meant that the expression levels are lower (for example by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or 100%) as compared to a suitable control. A suitable control may be, for example, a cell in which a gene encoding a protein that retains exosomes (such as tetherin) is not knocked out. Techniques for knocking out an undesirable gene, such as the gene encoding tetherin are also known in the art. The inventors believe that a reduction of tetherin expression in a cell may increase exosomal production by about 4-fold. Accordingly, in a further aspect, provided herein is a cell that has reduced expression of tetherin. Suitably, the cell may be isolated. Provided herein is also the use of a cell with reduced expression of tetherin in the production of an exosome. Suitably, the exosome may be genetically modified. Suitably, such a genetically modified exosome may be the exosome of the invention. Suitably, such a genetically modified exosome may comprise the fusion protein of the invention. Expression Vectors In one aspect the invention provides an expression vector comprising an expressible nucleic acid encoding the fusion proteins herein described. Suitably, the nucleic acid may share at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with a sequence selected from the group consisting of: SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:61, SEQ ID NO:64, SEQ ID NO:67 and SEQ ID NO:70. In one embodiment the vector may be any vector capable of transferring DNA to a cell. Suitably, the vector is an integrating vector or an episomal vector. Preferred integrating vectors include recombinant retroviral vectors. A recombinant retroviral vector will include DNA of at least a portion of a retroviral genome which portion is capable of infecting the target cells. The term “infection” is used to mean the process by which a virus transfers genetic material to its host or target cell. Suitably, the retrovirus used in the construction of a vector of the invention is also rendered replication-defective to remove the effect of viral replication of the target cells. In such cases, the replication-defective viral genome can be packaged by a helper virus in accordance with conventional techniques. Generally, any retrovirus meeting the above criteria of infectiousness and capability of functional gene transfer can be employed in the practice of the invention. Suitably, the retroviral vector may be a lentiviral vector. Lentiviral vectors are well known in the art, and have been used to deliver genes to numerous cell types, including HeLa or B lymphoid cells. Other vectors useful in the present invention include adenovirus, adeno-associated virus, SV40 virus, vaccinia virus, HSV and poxvirus vectors. Preferred episomal vectors include transient non-replicating episomal vectors and self-replicating episomal vectors with functions derived from viral origins of replication such as those from EBV, human papovavirus (BK) and BPV-1. Such integrating and episomal vectors are well known to those skilled in the art and are fully described in the body of literature well known to those skilled in the art. In particular, suitable episomal vectors are described in WO98/07876. Mammalian artificial chromosomes can also be used as vectors in the present invention. The use of mammalian artificial chromosomes is discussed by Calos (1996 Trends in Genetics 12: 463-466). In a preferred embodiment, the vector of the present invention is a plasmid. The plasmid may be a non-replicating, non-integrating plasmid. The term “plasmid” as used herein refers to any nucleic acid encoding an expressible gene and includes linear or circular nucleic acids and double or single stranded nucleic acids. The nucleic acid can be DNA or RNA and may comprise modified nucleotides or ribonucleotides, and may be chemically modified by such means as methylation or the inclusion of protecting groups or cap- or tail structures. A non-replicating, non-integrating plasmid is a nucleic acid which when transfected into a host cell does not replicate and does not specifically integrate into the host cell’s genome (i.e. does not integrate at high frequencies and does not integrate at specific sites). Replicating plasmids can be identified using standard assays including the standard replication assay of Ustav et al (1991 EMBO J 10: 449–457). The present invention also provides a cell transformed or transfected with (i.e. comprising) the expression vector of the present invention. The present invention also provides a cell comprising the fusion protein of the invention or exosome of the invention. Suitably the cell is an isolated cell. Suitably the cell is a mammalian cell. Suitably, the cell is a HeLa cell. Suitably, the cell may be modified to produce increased amounts of exosomes as compared to control cells. Numerous techniques are known and are useful according to the invention for delivering the vectors described herein to cells, including the use of nucleic acid condensing agents, electroporation, complexing with asbestos, polybrene, DEAE cellulose, Dextran, liposomes, cationic liposomes, lipopolyamines, polyornithine, particle bombardment and direct microinjection (reviewed by Kucherlapati and Skoultchi (1984 Crit. Rev. Biochem 16: 349-379); Keown et al (1990 Methods Enzymol 185:527-37). A vaccine composition Provided herein is a vaccine composition comprising the exosome of the invention or a membrane preparation comprising the fusion protein of the invention. Suitably, the vaccine is anti-SARS-CoV-2. The term “vaccine composition” and “composition” are used herein interchangeably and refer to a composition comprising, in addition to comprising the exosome of the invention, a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier. As used herein, "vaccine" is defined broadly to refer to any type of biological agent in an administrable form capable of stimulating an immune response (for example a B-cell immune and/or T-cell immune response) in an animal (for example human) inoculated with the vaccine. Suitably, the vaccine prevents or ameliorates the symptoms of a disease, such as an infection (for example COVID-19). Thus, reduction in the incidence or severity of characteristic symptoms of COVID-19 in comparison to non-immunized subjects may be termed a vaccine for the purposes of this invention. The term “membrane preparation” as used herein refers to a composition comprising membrane fragments from cells which express the fusion protein (for example HeLa cells or LCL721 cells). Methods for obtaining a membrane preparation are well known in the art. Suitably, the fusion protein is attached to the membrane fragments in the membrane preparation. Suitably, the fusion protein is attached to the membrane fragments by the exosomal protein (such as tetraspanin). In the one aspect, the present invention provides a membrane preparation comprising the fusion protein of the invention. As used herein, the term “membrane preparation” and “fragment of a membrane” may be used interchangeably. Compositions may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, supplementary immune potentiating agents such as adjuvants and cytokines and optionally other therapeutic agents or compounds. As used herein, "pharmaceutically acceptable" refers to a material that is not biologically or otherwise undesirable, i.e., the material may be administered to an individual along with the selected binding protein without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained. Excipients are natural or synthetic substances formulated alongside an active ingredient (e.g. fusion protein or exosome of the invention), included for the purpose of bulking-up the formulation or to confer a therapeutic enhancement on the active ingredient in the final dosage form, such as facilitating drug absorption or solubility. Excipients can also be useful in the manufacturing process, to aid in the handling of the active substance concerned such as by facilitating powder flowability or non-stick properties, in addition to aiding in vitro stability such as prevention of denaturation over the expected shelf life. Pharmaceutically acceptable excipients are well known in the art. A suitable excipient is therefore easily identifiable by one of ordinary skill in the art. By way of example, suitable pharmaceutically acceptable excipients include water, saline, aqueous dextrose, glycerol, ethanol, and the like. Diluents are diluting agents. Pharmaceutically acceptable diluents are well known in the art. A suitable diluent is therefore easily identifiable by one of ordinary skill in the art. Carriers are non-toxic to recipients at the dosages and concentrations employed and are compatible with other ingredients of the formulation. The term “carrier” denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. Pharmaceutically acceptable carriers are well known in the art. A suitable carrier is therefore easily identifiable by one of ordinary skill in the art. Provided herein is the exosome or vaccine composition of the invention for use as a medicament. Said medicament may be used for human or animal therapy of various diseases or disorders in a therapeutically effective dose. Suitably the disease may be an infection. The medicament can be formulated by various techniques dependent on the desired application purposes. Formulations described herein in connection to the vaccine composition of the invention are exemplary formulations of the medicament. Also provided herein is the exosome or vaccine composition of the invention for use in the prevention or amelioration of an infection in a subject. The term “infection” as used herein refers to the undesirable presence of a microorganism in or on the subject’s body. Suitably, the microorganism is pathogenic. Such an undesirable presence of a microorganism may be symptomatic or asymptomatic. It will be appreciated that whether the infection is symptomatic or asymptomatic may depend on the subject and/or type of microorganism. It will also be appreciated where an infection is symptomatic, the type of symptoms and their severity may depend on the subject and/or type of microorganism. Suitably the infection may be a viral, bacterial or fungal infection. Merely by way of example, if the infection is caused by SARS-CoV- 2, the symptoms may include fever, cough, and/or loss of taste and/or smell. Also provided is a method of eliciting an anti-SARS-CoV-2 immune response in a mammal comprising administering the exosome or a vaccine composition comprising the exosome of the invention to a subject. Also provided herein is a method for preventing or ameliorating an infection in a subject, the method comprising providing the subject in need thereof with a therapeutically effective amount of an exosome or vaccine composition of the invention. "Therapeutically effective amount" as used herein refers to the amount of exosomes or vaccine composition of the invention that elicit a biological response in a tissue or system of a subject such that the infection is prevented or ameliorated. Methods of establishing a therapeutically effective amount will be known to those skilled in the art and merely by way of example can include performing a dose response assay. The term "subject" will be understood to refer to a warm-blooded animal, particularly a mammal. Non-limiting examples of animals within the scope and meaning of this term include humans, non-human primates, dogs, cats, guinea pigs, rats, mice, horses, goats, cattle, sheep, and zoo animals. The present invention provides novel vaccine compositions comprising the exosomes of the invention for controlling SARS-CoV-2 infection in mammalian, most preferably human, subjects. The present vaccine compositions are useful in providing immune resistance against the strain(s) of SARS-CoV-2 used for preparation of the fusion proteins of the invention, as well as against strains which are different from those used in the preparation of the fusion proteins. In certain embodiments vaccines of the invention are administered prophylactically. In certain embodiments, a vaccine of the invention is administered therapeutically. In an embodiment, the vaccine is administered to a patient with an active SARS-CoV-2 infection to both treat the active infection and/or prevent recurrence of a SARS-CoV- 2. EXAMPLES Example 1 Constructs Constructs are designed to integrate fragments, or whole parts of SARS-CoV-2 genes CD63. SARS-CoV-2 genes are: - Tagged to the N terminus of CD63; - Inserted into luminal loops 1; and/or - Inserted into luminal loop 2. Location may depend on the antigen. For example, N and M will be N terminal, Spike or ACE2 constructs will be within the loops. For inserts to luminal loop 1 inserts are between Q36 and L37 of CD63. For inserts to luminal loop 2, inserts will be between R108 and D109 of CD63. CD63-FLAG-SpikeRBD (V1), N-CD63-FLAG-SpikeRBD (V16), FLAG-N-CD63 (V19) and FLAG-M-CD63 (V18) were synthesised by GeneWiz and cDNA was flanked by a 5’ EcoRI and a 3’ BamHI site. These were ligated to pLVX-EF1alpha-IRES-Puro and transduced to competent cells. Minipreps were picked, analysed by test digests and potential colonies sequenced by Sanger sequencing (GeneWiz). The construct V18 showed stable expression (data not shown). Other constructs, specifically V4 and V12 containing the M protein did not show stable expression. Without wishing to be bound by this hypothesis, the inventors believe that the M fragments (SEQ ID NO: 58) in V4 and V12 were the reason for lack of stable expression. Results The data below have been performed for the construct v1 - CD63-FLAG-SpikeRBD. CD63-FLAG-SpikeRBD cDNA was synthetically synthesised and cloned to the lentiviral vector pLVX-EF1alpha-IRES-Puro. Miniprepped DNA was subject to test digest and potential clonal DNA selected and sanger sequenced (GeneWiz). Minipreps 1+3 were both found to have the correct sized insert by test digest, and the CD63-FLAG-SpikeRBD was confirmed by sanger sequencing (Fig 4). To check the FLAG-CD63-Spike insert was not disrupting CD63 trafficking, pLVX- EF1alpha-CD63-FLAG-SpikeRBD-IRES-Puro DNA was transiently transfected to HeLa cells and subject to immunofluorescence (Fig 5). FLAG signal was confirmed to be in CD63-positive compartments as shown by colocalisation of signal, and CD63 localisation remained in punctate organelles in transfected cells (as is also observed in non-transfected cells). After confirming expression of CD63-FLAG-SpikeRBD DNA was tolerated by cells, and localised to the correct intracellular compartments, stable cell lines were generated. Briefly, HEK293T cells were transfected with pLVX-EF1alpha-CD63- FLAG-SpikeRBD-IRES-Puro and the lentiviral packaging plasmids pCMVR8.91 (“199”) and pMD.G (VSV-G) (“200”) using TransIT-293 (Mirus Bio, USA). Viral supernatants were collected 48 h after transfection, passed through 0.45 μm filters and both wild-type and tetherin KO HeLa cells transduced by ‘spinfection’—viral supernatants were centrifuged at 1800 rpm in a benchtop centrifuge at 37 °C for 3 h to enhance viral transduction. After 48 hours, transduced WT HeLa and Bst2KO HeLa cells were selected with 1ug/ml Puromycin for 5 days. WT and Bst2KO stable HeLa cell lines were confirmed to express CD63-FLAG-SpikeRBD using immunofluorescence, again using anti-FLAG antibodies, but also using a human monoclonal antibody raised against SARS-CoV-2 Spike-RBD (P06DHu, ThermoFisher) (Fig 6). FLAG staining colocalised to CD63, a marker for multivesicular endosomes – the compartment where intraluminal vesicles (and therefore exosomes) are generated. The increase in CD63 staining in transduced cells reflects the additional, ectopic copy of CD63, rather than just the endogenous CD63. An antibody against SpikeRBD showed similar pattern of staining, and colocalised with FLAG – confirming that Spike and FLAG are together localised in CD63-positive, late endosomal compartments. Together, these data indicate that Spike RBD and FLAG are localised to late endocytic compartments as expected (Fig 7). Whole cell lysates were collected from untransduced and transduced WT and Bst2KO HeLa cells. Lysates were probed with antibodies against CD63, and both transduced cell lines displayed enhanced staining, and at a higher molecular weight – reflecting additional molecular mass given from the addition of FLAG-SpikeRBD (Fig 8A). Antibodies against FLAG were similarly used to blot whole cell lysates, and a positive band was observed at the correct molecular weight in transduced stable cell lines, and not in untransduced controls. Exosome-enriched fractions were generated from cells and subject to Western blotting. Exosomes from tetherin KO HeLa cells + CD63- FLAG-SpikeRBD displayed a band at the correct molecular weight as expected (Fig 8B). Further evidence of expression of FLAG-SpikeRBD in the CD63 fusion protein was obtained by mass spectrometry analysis. HeLa cells stably expressing CD63-FLAG- SpikeRBD were lysed, ran on a NuPage (Invitrogen) protein gel and stained with InstantBlue Commassie Stain. A gel band between 50 and 70 kDa was excised, digested with Trypsin and subject to proteomic analysis. Liquid chromatography-mass spectrometry identified a number of unique peptides within CD63-FLAG-SpikeRBD (SEQ ID NO: 5) as follows: Table 2
Figure imgf000040_0001
Figure imgf000041_0001
The sequence of the theoretical CD63-FLAG-SpikeRBD sequence is shown in SEQ ID NO: 5. Of the identified unique peptides identified, several were identified which originate from the SpikeRBD. The identified unique peptides, and their position within CD63-FLAG-SpikeRBD and amino acid position were defined as shown below:
Figure imgf000041_0002
Finally, to confirm that exosomes from transduced cells contained FLAG-SpikeRBD, we performed surface labelling immunogold electron microscopy, making use of the FLAG epitope that is exposed on the outside of exosomes. Briefly, transduced and untransduced WT HeLa cells were treated with Bafilomycin A1 for 16 hours to induce exosome release as previously described (Edgar et al., eLife 2016;5:e17180 DOI: 10.7554/ELIFE.17180). Cells were fixed before being labelled with antibodies against FLAG.10nm Protein-A gold was then used to detect anti-FLAG antibodies. Cells were then re-fixed with glutaraldehyde before being processed for electron microscopy. Ultrathin sections were cut, stained with heavy metals and imaged. Immunogold labelling was only observed in transduced cells and was specifically seen on surface- associated exosomes as expected. Together, these data confirm that CD63-FLAG-SpikeRBD is trafficked to late endosomes, and to exosomes (Fig 9A, B). Antibodies SARS-CoV-2 Spike-RBD (P06DHu, human monoclonal, ThermoFisher, 1:100 for IF) Anti-FLAG; (L5, rat monoclonal, BioLegend, 1:200 for IF), (anti-DYKDDDDK, D6W5B, rabbit monoclonal, Cell Signalling Technology, 1:200 for IF)), (M2, mouse monoclonal, Sigma Aldrich, 1:1000 for Wester blotting). CD63 (H5C6, mouse monoclonal, BDBiosciences, 1:200 for IF) GAPDH (14C10, rabbit monoclonal, Cell Signalling Technology, 1:1000 for Western blotting). Example 2 The aim of the following example is to demonstrate that SARS-CoV2 spike protein containing exosomes (RBD region) elicit recognition by T-cells specific to epitopes with SARS-CoV-2 Spike. A polyclonal T-cell culture from a donor with reactivity to the RBD region of SARS-CoV-2 spike was used. Reactivity was demonstrated by incubating some of the culture with overlapping peptides spanning the RBD region of the SARS-CoV-2 spike protein (commercially available PepMix™, JPT Peptide Technologies). Autologous Lymphoblastoid Cell Lines (LCL) were established from B cells isolated from the same donor and transformed with Epstein-Barr virus (EBV) into permanent cell lines. These LCLs serve as targets of T-cells in assays. Membranes prepared from HeLa cells expressing the SpikeRBD fusion protein were used as a positive control. Method The autologous LCL cells were washed once with serum-free AIM-V medium, then dispensed into 10 different 15 ml tubes (150,000 cells/tube in volume of 50ul). One tube was left untreated (negative control) and one had 0.5 ul RBD PepMix added (positive control). To the remaining tubes, 50 ul or 5 ul of the different exosome or membrane preparations was added. LCL cells were then incubated at 37°C 5% CO2, with intermittent agitation to keep cells suspended. During the incubation, polyclonal T-cells were counted, washed and resuspended to 250,000 T-cells per ml. Aliquots (100 ul) were added to a 96 well plate. After one hour each tube of LCL cells were washed once with 8ml LCL media, resuspended in 200 ul of LCL medium and pipetted into appropriate wells of a 96U bottom plate containing T-cells. Outer wells of each plate were filled with saline to reduce evaporation and plates were incubated at 37°C 5% CO2. After 24 and 48hr incubation, 60 ul of supernatant from each well were dispensed into separate wells of a 96 well plate and stored at 4°C. The interferon gamma (IFN-γ) concentration of each aliquot of supernatant was tested using an IFN-γ capture ELISA, with doubling dilutions of recombinant IFN-γ included for quantitation of IFN-γ concentration in the samples. Whole cell membranes from HeLa cells expressing CD63-FLAG-SpikeRBD were prepared in cold hypotonic buffer and mechanically lysed. The homogenate was centrifuged at 25,000 xg, 20mins, 4°C and the resulting cell membrane pellets were resuspended in PBS. An aliquot was used as a positive control in the T cell assay. Results The polyclonal T-cell culture did not produce any interferon-gamma when co-cultured with the autologous LCL, but produced large quantities of the cytokine when co- cultured with the same LCL that had been exposed to SARS-CoV-2 spike RBD PepMix. This demonstrates that the polyclonal culture contains T-cells specific to epitopes within the RBD region. The lack of any recognition of the autologous LCL allows this polyclonal culture to be used to investigate exosome and membrane preparations. This polyclonal culture produced large quantities of interferon-gamma when co- cultured with autologous LCL exposed to the SARS-CoV-2 spike exosomes, but not the control exosomes. The same pattern was seen for LCL exposed to the membrane fractions. These results are shown in Table 4. These results demonstrates that the Spike RBD containing exosomes elicit strong activation of RBD-specific T-cells. Control exosomes not containing Spike RBD did not non-specifically activate T-cells. Table 4 T-cell activation results
Figure imgf000044_0001
Further, Example 2 was repeated using exosomes containing constructs v16 (N- CD63-FLAG-SpikeRBD) or v19 (FLAG-N-CD63). For V16, T-cell activation was tested in both a polyclonal T-cell culture from a donor with reactivity to the RBD region of SARS-CoV-2 spike and N protein. For V19, T-cell activation was tested in both a polyclonal T-cell culture from a donor with reactivity to the N protein. The results are shown in Figure 15. As can be seen in Figure 15A, exosomes containing construct v16 results in even greater activation of T cells than construct V1. The same pattern was seen for LCL exposed to the membrane fractions. Example 3 Materials and methods (of experiments the results of which are shown in Figures 10 to 13). Immunofluorescence Cells were grown on glass coverslips and fixed using 4% PFA/PBS. Cells were quenched with 15 mM glycine/PBS and permeabilised with 0.1% saponin/PBS. Blocking and subsequent steps were performed with 1% BSA, 0.01% saponin in PBS. Cells were mounted on slides with mounting medium containing DAPI. Cells were imaged using a LSM700 confocal microscope. Flow cytometry Cells were gently trypsinised, and surface stained for flow cytometry in PBS with 0.5 % BSA + 1 mM EDTA (FACS buffer) for 30 min on ice. For intracellular staining, cells were first fixed then permeabilised with 0.1* saponin/PBS and subsequently stained in PBS with FACs buffer. Samples were acquired on a 4 laser Cytoflex S. Western blotting Cell lysates were mixed with 4x NuPage LDS sample buffer. Gels were loaded to NuPage 4–12% Bis-Tris precast gels and transferred to PVDF membranes before being blocked using 5% milk / PBS / 0.1% Tween. Primary antibodies and secondary antibodies were diluted in 5% milk/PBS-tween. Blots were imaged using a Odyseey CLx. SEQUENCES Throughout this section, FLAG sequences are shown in bold; linker sequences are shown in italic font SEQ ID NO: 1 amino acid sequence of S protein extracellular domain
Figure imgf000046_0001
SEQ ID NO: 2 N protein amino acid sequence
Figure imgf000046_0002
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
EXEMPLARY EMBODIMENTS OF THE INVENTION The below paragraphs provide exemplary embodiments of the fusion protein and other aspects of the invention described herein 1. A fusion protein comprising: i) an exosomal protein, wherein the exosomal protein is a tetraspanin protein; and ii) one or more immunogenic protein. 2. The fusion protein of paragraph 1, wherein the tetraspanin is selected from the group consisting of CD63, CD9, and CD81, or a fragment, or variant thereof. 3. The fusion protein of paragraphs 1 or 2, wherein the one or more immunogenic protein is capable of eliciting an immune response against a SARS-CoV-2 protein. 4. The fusion protein of paragraph 3, wherein the SARS-CoV-2 protein is selected from the group consisting of spike protein (S), nucleoprotein (N), membrane protein (M), envelope protein (E), open reading frame (ORF) 1ab polyprotein, ORF 3a protein, ORF 3b protein, ORF 6 protein, ORF 7a protein, ORF 7b protein, ORF 8 protein, ORF 9b protein, ORF 9c protein, and ORF 10 protein, optionally wherein when the SARS- CoV-2 protein is spike protein (S) it is the extracellular domain of spike protein (S). 5. The fusion protein of any one of the preceding paragraphs, wherein the one or more immunogenic protein is capable of eliciting a B-cell and a T-cell immune response. 6. The fusion protein of any one of the preceding paragraphs, wherein the one or more immunogenic protein is capable of eliciting a T-cell immune response. 7. The fusion protein of the preceding paragraphs, wherein the protein comprises a first one or more immunogenic protein capable of eliciting a B-cell and a T-cell immune response and a second one or more immunogenic protein capable of eliciting a T-cell immune response. 8. The fusion protein of any one of paragraphs 5 to 7, wherein the first one or more immunogenic protein capable of eliciting a B-cell and T-cell immune response is incorporated into a luminal loop of the exosomal protein. 9. The fusion protein of any one of paragraphs 5 to 8, wherein the second one or more immunogenic protein capable of eliciting a T-cell immune response is incorporated near or at the N-terminus of the exosomal protein, and/or is incorporated into an intravesicular loop of the exosomal protein. 10. The fusion protein of any one of paragraphs 5 to 9, wherein first the one or more immunogenic protein is capable of eliciting a B-cell and T-cell immune response against the SARS-CoV-2 spike protein (S), optionally the extracellular domain of spike protein (S). 11. The fusion protein of any one of paragraphs 5 to 10, wherein the second one or more immunogenic protein is capable of eliciting a T-cell immune response against the SARS-CoV-2 M and/or N protein, and/or ORF proteins, and/or E protein. 12. The fusion protein of any one of paragraphs 3 to 11, wherein the first one or more immunogenic protein capable of eliciting a B-cell and a T-cell immune response is incorporated into the first and/or second luminal loop of the tetraspanin, optionally wherein the tetraspanin is CD63, or CD9 or CD81. 13.The fusion protein of any one of paragraphs 2 to 12, wherein the second one or more immunogenic protein capable of eliciting a T-cell immune response is incorporated near or at the N-terminus of the tetraspanin, optionally wherein the tetraspanin is CD63, or CD9 or CD81. 14. The fusion protein of any one of the preceding paragraphs, wherein the one or more immunogenic protein shares at least 75% identity a SARS-CoV-2 protein or protein variant, or a fragment thereof. 15. The fusion protein of paragraph 14 wherein the SARS-CoV-2 protein is selected from the group consisting of the extracellular domain of spike protein (S) (SEQ ID NO: 1), nucleoprotein (N) (SEQ ID NO: 2), and membrane protein (M) (SEQ ID NO: 3). 16. The fusion protein of paragraph 15, wherein the fragment of the spike protein (S) is the receptor binding domain (RBD) (SEQ ID NO: 4), the fragment of the membrane protein (M) is according to SEQ ID NO: 57, 58, or 72 and/or the fragment of the nucleoprotein (N) is according to SEQ ID NO: 56 or 71. 17. The fusion protein of any one of the preceding paragraphs, sharing at least 75%, at least 80%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identity with SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 68, or SEQ ID NO:69. 18. The fusion protein of paragraphs 17, wherein the fusion protein comprises or consists of SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 59, SEQ ID NO:60, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO:68, or SEQ ID NO: 69. 19. The fusion protein of any one of the preceding paragraphs, wherein the exosomal protein and the one or more immunogenic protein is separated by a linker sequence. 20. An exosome or membrane preparation comprising a fusion protein of any one of paragraphs 1 to 19. 21. The exosome or membrane preparation according to paragraph 20, wherein the exosome further comprises a DC receptor ligand and/or an immune enhancing moiety, optionally wherein the immune enhancing moiety is selected from the group consisting of a molecule with a pathogen-associated molecular pattern (PAMP), DNA oligonucleotide containing an CpG motif, a STING agonist (such as cyclic GMP-AMP), and a TLR9 (Toll-like Receptor) agonist. 22. A vaccine composition comprising an exosome or membrane preparation of paragraph 20 or 21. 23. A nucleic acid molecule encoding a fusion protein of any one of paragraphs 1 to 19. 24. An expression vector comprising the nucleic acid molecule of paragraph 23. 25. The expression vector of paragraph 24, wherein it is a lentivirus expression vector. 26. A cell comprising a fusion protein of any one of paragraphs 1 to 19, an exosome or membrane preparation of paragraph 20 or 21, a nucleic acid of paragraph 23, and/or an expression vector of paragraph 24 or 25, optionally wherein the cell is an isolated cell. 27. The cell of paragraph 26, wherein the transformed cell line is a HeLa cell or a LCL721 cell. 28. The cell of paragraph 26 or 27, wherein the cell is modified to enhance the production of exosomes. 29. The cell of any one of paragraph 26 to 28, wherein the cell has reduced expression of tetherin. 30. The exosome or membrane preparation of paragraph 20 or 21, or the vaccine composition according to paragraph 22, for use as a medicament. 31. The exosome or membrane preparation of paragraph 20 or 21, or the vaccine composition according to paragraph 22, for use in the prevention or amelioration of an infection. 32. The exosome, membrane preparation, or vaccine composition for use according to paragraph 31, wherein the infection is a viral, bacterial or fungal infection. 33. A method of preventing or ameliorating an infection in a subject, the method comprising providing the subject in need thereof with a therapeutically effective amount of an exosome or membrane preparation of paragraph 20 or 21, or a vaccine composition according to paragraph 22. 34. The method of paragraph 33, wherein the infection is a viral, bacterial or fungal infection. 35. The method of paragraph 34, wherein the viral infection is a SARS-CoV-2 infection.

Claims

CLAIMS 1. An exosome or membrane preparation comprising a fusion protein, wherein the fusion protein comprises: i) an exosomal protein, wherein the exosomal protein is a tetraspanin protein; and ii) one or more immunogenic protein.
2. The exosome or membrane preparation of claim 1, wherein the tetraspanin is selected from the group consisting of CD63, CD9, and CD81, or a fragment, or variant thereof.
3. The exosome or membrane preparation of claims 1 or 2, wherein the one or more immunogenic protein is capable of eliciting an immune response against a SARS-CoV- 2 protein.
4. The exosome or membrane preparation of claim 3, wherein the SARS-CoV-2 protein is selected from the group consisting of spike protein (S), nucleoprotein (N), membrane protein (M), envelope protein (E), Open reading frame (ORF) 1ab polyproteins, ORF 3a protein, ORF 3b protein, ORF 6 protein, ORF 7a protein, ORF 7b protein, ORF 8 protein, ORF 9b protein, ORF 9c protein, and ORF 10 protein, optionally wherein when the SARS-CoV-2 protein is spike protein (S) it is the extracellular domain of spike protein (S).
5. The exosome or membrane preparation of any one of the preceding claims, wherein the one or more immunogenic protein is capable of eliciting a B-cell and a T-cell immune response.
6. The exosome or membrane preparation of any one of the preceding claims, wherein the one or more immunogenic protein is capable of eliciting a T-cell immune response.
7. The exosome or membrane preparation of the preceding claims, wherein the protein comprises a first one or more immunogenic protein capable of eliciting a B-cell and a T-cell immune response and a second one or more immunogenic protein capable of eliciting a T-cell immune response.
8. The exosome or membrane preparation of any one of claims 5 to 7, wherein the first one or more immunogenic protein capable of eliciting a B-cell and T-cell immune response is incorporated into a luminal loop of the exosomal protein.
9. The exosome or membrane preparation of any one of claims 5 to 8, wherein the second one or more immunogenic protein capable of eliciting a T-cell immune response is incorporated near or at the N-terminus of the exosomal protein, and/or is incorporated into an intravesicular loop of the exosomal protein.
10. The exosome or membrane preparation of any one of claims 5 to 9, wherein first the one or more immunogenic protein is capable of eliciting a B-cell and T-cell immune response against the SARS-CoV-2 spike protein (S), optionally the extracellular domain of spike protein (S).
11. The exosome or membrane preparation of any one of claims 5 to 10, wherein the second one or more immunogenic protein is capable of eliciting a T-cell immune response against the SARS-CoV-2 M and/or N protein, and/or ORF proteins, and/or E protein.
12. The exosome or membrane preparation of any one of claims 3 to 11, wherein the first one or more immunogenic protein capable of eliciting a B-cell and a T-cell immune response is incorporated into the first and/or second luminal loop of the tetraspanin, optionally wherein the tetraspanin is CD63, or CD9 or CD81.
13.The exosome or membrane preparation of any one of claims 2 to 12, wherein the second one or more immunogenic protein capable of eliciting a T-cell immune response is incorporated near or at the N-terminus of the tetraspanin, optionally wherein the tetraspanin is CD63, or CD9 or CD81.
14. The exosome or membrane preparation of any one of the preceding claims, wherein the one or more immunogenic protein shares at least 75% identity a SARS- CoV-2 protein or protein variant, or a fragment thereof.
15. The exosome or membrane preparation of claim 14, wherein the SARS-CoV-2 protein is selected from the group consisting of the extracellular domain of spike protein (S) (SEQ ID NO: 1), nucleoprotein (N) (SEQ ID NO: 2), and membrane protein (M) (SEQ ID NO: 3).
16. The exosome or membrane preparation of claim 15, wherein the fragment of the spike protein (S) is the receptor binding domain (RBD) (SEQ ID NO: 4), the fragment of the membrane protein (M) is according to SEQ ID NO: 57 or 72, and/or the fragment of the nucleoprotein (N) is according to SEQ ID NO: 56 or 71.
17. The exosome or membrane preparation of any one of the preceding claims, wherein the fusion protein shares at least 75%, at least 80%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identity with SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 68, or SEQ ID NO:69.
18. The exosome or membrane preparation of claim 17, wherein the fusion protein shares at least 75%, at least 80%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identity with SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 59, SEQ ID NO:60, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO:68, or SEQ ID NO: 69, optionally wherein the fusion protein comprises or consists of SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 59, SEQ ID NO:60, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO:68, or SEQ ID NO: 69.
19. The exosome or membrane preparation of any one of the preceding claims, wherein the exosomal protein and the one or more immunogenic protein is separated by a linker sequence.
20. The exosome or membrane preparation of any one of claims 1 to 19, wherein the exosome or membrane preparation further comprises a DC receptor ligand and/or an immune enhancing moiety, optionally wherein the immune enhancing moiety is selected from the group consisting of a molecule with a pathogen-associated molecular pattern (PAMP), DNA oligonucleotide containing an CpG motif, a STING agonist (such as cyclic GMP-AMP), and a TLR9 (Toll-like Receptor) agonist.
21. A vaccine composition comprising an exosome or membrane preparation according to any one of claim 1 to 20.
22. A nucleic acid molecule encoding a fusion protein as described in claims 1 to 19.
23. An expression vector comprising the nucleic acid molecule of claim 22.
24. The expression vector of claim 23, wherein it is a lentivirus expression vector.
25. A cell comprising an exosome or membrane preparation of any one of claim 1 to 20, a fusion protein as described in any one of claims 1 to 19, a nucleic acid of claim 22, and/or an expression vector of claim 23 or 24, optionally wherein the cell is an isolated cell.
26. The cell of claim 25, wherein the transformed cell line is a HeLa cell or a LCL721 cell.
27. The cell of claim 25 or 26, wherein the cell is modified to enhance the production of exosomes.
28. The cell of any one of claims 25 to 27, wherein the cell has reduced expression of tetherin.
29. The exosome or membrane preparation of any one of claims 1 to 20, or the vaccine composition according to claim 21, for use as a medicament.
30. The exosome or membrane preparation of any one of claims 1 to 20, or the vaccine composition according to claim 21, for use in the prevention or amelioration of an infection.
31. The exosome, membrane preparation, or vaccine composition for use according to claim 30, wherein the infection is a viral, bacterial or fungal infection.
32. A method of preventing or ameliorating an infection in a subject, the method comprising providing the subject in need thereof with a therapeutically effective amount of an exosome or membrane preparation of any one of claims 1 to 20, or a vaccine composition according to claim 21.
33. The method of claim 32, wherein the infection is a viral, bacterial or fungal infection.
34. The method of claim 33, wherein the viral infection is a SARS-CoV-2 infection.
PCT/US2022/023864 2021-04-08 2022-04-07 Fusion protein for antigen presentation WO2022216949A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB2105044.8A GB2606693A (en) 2021-04-08 2021-04-08 Fusion protein
GB2105044.8 2021-04-08

Publications (1)

Publication Number Publication Date
WO2022216949A1 true WO2022216949A1 (en) 2022-10-13

Family

ID=75949647

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/023864 WO2022216949A1 (en) 2021-04-08 2022-04-07 Fusion protein for antigen presentation

Country Status (2)

Country Link
GB (1) GB2606693A (en)
WO (1) WO2022216949A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998007876A2 (en) 1996-08-16 1998-02-26 Medical Research Council Self-replicating episomal expression vectors conferring tissue-specific gene expression
US20190112351A1 (en) * 2016-04-04 2019-04-18 National Institutes Of Biomedical Innovation, Health And Nutrition Exosome-targeted dna vaccine

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2569142A1 (en) * 2004-06-02 2005-12-22 New York Blood Center Sars vaccines and methods to produce highly potent antibodies
US20220184225A1 (en) * 2019-04-23 2022-06-16 Case Western Reserve University Fusogenic particles and related methods for delivering therapeutic agents to cells
CA3170538A1 (en) * 2020-02-11 2021-08-19 Cytonus Therapeutics, Inc. Rapid vaccine platform
CA3171092A1 (en) * 2020-03-13 2021-09-16 Stephen John GOULD Selectable marker proteins, expression vectors, for the production of virus-like particles for therapeutic and prophylactic applications
CN112226413B (en) * 2020-10-14 2022-04-15 苏州大学 Targeting exosome based on SARS-CoV-2S protein RBD region and preparation method thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998007876A2 (en) 1996-08-16 1998-02-26 Medical Research Council Self-replicating episomal expression vectors conferring tissue-specific gene expression
US20190112351A1 (en) * 2016-04-04 2019-04-18 National Institutes Of Biomedical Innovation, Health And Nutrition Exosome-targeted dna vaccine

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
CALOS, TRENDS IN GENETICS, vol. 12, 1996, pages 463 - 466
EDGAR ET AL., ELIFE, vol. 5, 2016, pages e17180
IVANUSIC DANIEL ET AL: "tANCHOR: a novel mammalian cell surface peptide display system", BIOTECHNIQUES, vol. 70, no. 1, 14 December 2020 (2020-12-14), US, pages 21 - 28, XP055945575, ISSN: 0736-6205, DOI: 10.2144/btn-2020-0073 *
KEOWN ET AL., METHODS ENZYMOL, vol. 185, 1990, pages 527 - 37
KUCHERLAPATISKOULTCHI, CRIT. REV. BIOCHEM, vol. 16, 1984, pages 349 - 379
MEYERS ET AL., CABIOS, vol. 4, 1989, pages 11 - 17
NEEDLEMAN ET AL., J. MOL. BIOL., vol. 48, 1970, pages 444 - 453
POLAK K. ET AL: "Extracellular vesicle-based vaccine platform displaying native viral envelope proteins elicits a robust anti-SARS-CoV-2 response in mice", BIORXIV, 28 October 2020 (2020-10-28), XP055785135, Retrieved from the Internet <URL:https://www.biorxiv.org/content/10.1101/2020.10.28.357137v1.full.pdf> [retrieved on 20210312], DOI: 10.1101/2020.10.28.357137 *
RICE ADRIAN ET AL: "A Next Generation Bivalent Human Ad5 COVID-19 Vaccine Delivering Both Spike and Nucleocapsid Antigens Elicits Th1 Dominant CD4+, CD8+ T-cell and Neutralizing Antibody Responses", BIORXIV, 30 July 2020 (2020-07-30), pages 1 - 36, XP055929959, DOI: 10.1101/2020.07.29.227595 *
SABANOVIC BERINA ET AL: "Promising Extracellular Vesicle-Based Vaccines against Viruses, Including SARS-CoV-2", BIOLOGY, vol. 10, no. 2, 27 January 2021 (2021-01-27), pages 94, XP055785141, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7912280/pdf/biology-10-00094.pdf> DOI: 10.3390/biology10020094 *
STEINBUCK MARTIN P. ET AL: "A lymph node-targeted Amphiphile vaccine induces potent cellular and humoral immunity to SARS-CoV-2", SCIENCE ADVANCES, 5 February 2021 (2021-02-05), XP055896177, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7864572/pdf/abe5819.pdf> *
TOMOHIRO KANUMA ET AL: "CD63-Mediated Antigen Delivery into Extracellular Vesicles via DNA Vaccination Results in Robust CD8 + T Cell Responses", THE JOURNAL OF IMMUNOLOGY, vol. 198, no. 12, 15 May 2017 (2017-05-15), US, pages 4707 - 4715, XP055541346, ISSN: 0022-1767, DOI: 10.4049/jimmunol.1600731 *
USTAV ET AL., EMBO J, vol. 10, 1991, pages 449 - 457

Also Published As

Publication number Publication date
GB2606693A (en) 2022-11-23
GB202105044D0 (en) 2021-05-26

Similar Documents

Publication Publication Date Title
JP3581366B2 (en) Lysosomal targeting of immunogens
CN111375055B (en) 2019-nCoV subunit vaccine composition and immunization method thereof
CN114096675A (en) Coronavirus immunogenic compositions and uses thereof
Toes et al. Enhancement of tumor outgrowth through CTL tolerization after peptide vaccination is avoided by peptide presentation on dendritic cells
Polyak et al. Impaired class II expression and antigen uptake in monocytic cells after HIV-1 infection.
Granadillo et al. A novel fusion protein-based vaccine comprising a cell penetrating and immunostimulatory peptide linked to human papillomavirus (HPV) type 16 E7 antigen generates potent immunologic and anti-tumor responses in mice
SG191830A1 (en) Use of toll-like receptor agonist for treating cancer
KR20140043348A (en) Liposomal formulations
CN113666990A (en) T cell vaccine immunogen for inducing broad-spectrum anti-coronavirus and application thereof
Zhao et al. TAT-mediated gp96 transduction to APCs enhances gp96-induced antiviral and antitumor T cell responses
JP4587566B2 (en) Methods for creating or enhancing T cell responses to target cells using complexes comprising HLA class I molecules and attachment means
US20220411481A1 (en) Artificial synapses
WO2023051701A1 (en) Mrna, protein and vaccine against sars-cov-2 infection
WO2023184861A1 (en) Hpv epitope and identification method therefor, and application thereof
AU2022201142A1 (en) Vaccine compositions and methods for restoring NKG2D pathway function against cancers
EA027803B1 (en) Immunogenic compounds comprising hiv gp41 peptide coupled to crm197 carrier protein
FR2757169A1 (en) FUSION PROTEIN COMPRISING ALL OR PART OF THE PP65 PROTEIN OF HUMAN CMV, ESPECIALLY USEFUL IN THE PREPARATION OF A VACCINE
Zong et al. Human HSP70 and modified HPV16 E7 fusion DNA vaccine induces enhanced specific CD8+ T cell responses and anti-tumor effects
US20230256082A1 (en) Vaccine against human-pathogenic coronaviruses
JP2021515588A (en) Peptide exchange protein
WO2023184862A1 (en) Hpv epitope, identification method therefor, and application thereof
WO2008069663A1 (en) Use of a varicellovirus tap-inhibitor for the induction of tumor- or virus-specific immunity against teipp
WO2022216949A1 (en) Fusion protein for antigen presentation
KR20220052292A (en) Vaccine or pharmaceutical composition for preventing or treating COVID-19 using exosomes
KR101570453B1 (en) Recombinant cell expressing recombinant spike 1 protein of porcine epidemic diarrhea virus

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22719714

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22719714

Country of ref document: EP

Kind code of ref document: A1