WO2023039535A1 - Outer membrane vesicle (omv)-based vaccines against sudden acute respiratory syndrome coronavirus 2 (sars-cov-2) spike protein - Google Patents

Outer membrane vesicle (omv)-based vaccines against sudden acute respiratory syndrome coronavirus 2 (sars-cov-2) spike protein Download PDF

Info

Publication number
WO2023039535A1
WO2023039535A1 PCT/US2022/076219 US2022076219W WO2023039535A1 WO 2023039535 A1 WO2023039535 A1 WO 2023039535A1 US 2022076219 W US2022076219 W US 2022076219W WO 2023039535 A1 WO2023039535 A1 WO 2023039535A1
Authority
WO
WIPO (PCT)
Prior art keywords
omvs
protein
cov
sars
plor
Prior art date
Application number
PCT/US2022/076219
Other languages
French (fr)
Inventor
Andrew Camilli
Stefan Schild
Himadri Bhadur THAPA
Anna Maria MÜLLER
Original Assignee
Trustees Of Tufts College
University Of Graz
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 Trustees Of Tufts College, University Of Graz filed Critical Trustees Of Tufts College
Publication of WO2023039535A1 publication Critical patent/WO2023039535A1/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/025Enterobacteriales, e.g. Enterobacter
    • A61K39/0258Escherichia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/107Vibrio
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/74Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/523Bacterial cells; Fungal cells; Protozoal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/543Mucosal route intranasal
    • 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 field of the invention relates to vaccines.
  • the field of the invention relates to outer membrane vesical (OMV)-based vaccines against sudden acute respiratory syndrome coronavirus-2 (SARS-CoV-2) or COVID 19.
  • OMV outer membrane vesical
  • SARS-CoV-2 sudden acute respiratory syndrome coronavirus-2
  • COVID 19 BACKGROUND
  • the prevailing pandemic of SARS-CoV-2 highlights the desperate need of alternative vaccine-platforms, which are safe, effective, and can be modified to carry antigens of emerging pathogens.
  • the current SARS-CoV-2 vaccines based on mRNA and adenoviral vector technology meet some of these criteria but still face limitations regarding administration route, mass production, stability, and storage.
  • SUMMARY The inventors have developed a novel vaccine to protect against SARS-CoV-2 (COVID 19).
  • the inventors added the SARS-CoV-2 Spike protein Receptor-Binding Domain (RBD) as a heterologous antigen to outer membrane vesicles (OMVs) produced by bacterial.
  • RBD SARS-CoV-2 Spike protein Receptor-Binding Domain
  • OmpA outer membrane vesicles
  • the OmpA protein in both of V. cholerae and E. coli is a major outer membrane protein, and thus is present in multiple copies in OMVs produced by the strains.
  • Each OmpA-RBD fusion was designed so that the RBD portion is exposed on the outer surface of the OMVs where it can interact with immune cells.
  • Advantages of the disclosed OMV-based vaccines are that they can be administered intranasally as opposed to by needle injection.
  • the disclosed OMV-based vaccines exhibit heat stability as opposed to, for example, RNA-based vaccines which require deep cold storage and transportation. As such, the disclosed OMV-based vaccines may be useful in low-income, resource-poor nations, where cold storage and transportation are limited.
  • compositions comprising a fusion protein, the fusion protein comprising from N-terminus to C-terminus: the N-terminal signal sequence of the E. coli Lpp protein fused in frame either directly or via a linker to a fragment of the E. coli OmpA protein, wherein the fragment of the E. coli OmpA protein comprises a N-terminal deletion and a C-terminal deletion, and the fragment of the E. coli OmpA protein is fused either directly or via linker to a fragment of the SARS CoV-2 Spike protein comprising the receptor binding domain (RBD).
  • the compositions comprises outer membrane vesicles (OMVs) and the fusion protein is present on the surface of the OMVs.
  • the OMVs are derived from V. cholerae. In some embodiments, the OMVs are derived from E. coli. Also disclosed herein are expression vectors that expresses the fusion proteins disclosed herein. In some embodiments, the fusion protein is encoded by a polynucleotide that comprises codons that are optimized for expression in V. cholerae. In some embodiments, the fusion protein is encoded by a polynucleotide that comprises codons that are optimized for expression in E. coli. Also disclosed herein are microorganisms transfected with expression vectors comprising fusion proteins. By way of example, in some embodiments, a V.
  • cholerae cell transfected with an expression vector that expresses the fusion protein described herein is provided.
  • an E. coli cell transfected with an expression vector that expresses the fusion protein described herein is provided.
  • methods for inducing an immune response against SARS CoV-2 Spike protein comprising administering the composition comprising the fusion protein to a subject in need thereof.
  • the immune response comprises an antibody response against SARS CoV-2 Spike protein.
  • the immune response comprises a neutralizing antibody response against SARS CoV-2 Spike protein.
  • the method induces a protective response against infection by SARS CoV-2.
  • composition comprising a fusion protein, the fusion protein comprising SEQ ID NO: 2 or SEQ ID NO: 4.
  • methods for inducing an immune response against SARS CoV-2 Spike protein comprising administering the composition comprising the fusion protein of SEQ ID NO: 2 or SEQ ID NO: 4 to a subject in need thereof.
  • the composition comprises SEQ ID NO: 2 and SEQ ID NO: 4.
  • FIG. 1 Schematic design of the Lpp-OmpA-RBD (LOR) fusion constructs (top) and expected localization in the OMVs (bottom). Relevant parts of the LOR include the Lpp fragment (light blue), linkers (green), OmpA fragment (red), theSARS-CoV-2 RBD fragment (yellow), and the FLAG-tag (pink).
  • Panel (B) shown is a representative immunoblot detecting the LOR fusion protein in OMVs derived from V111pLOR-V and V111 p, E111 pLOR-E, and E111 p. The commercially available anti-RBD antisera specifically detecting the SARS-CoV-2 Spike protein was used for this immunoblot.
  • Panel (C) shown is a representative immunoblot detecting the heavy chain (approx. 55 kDa) of the commercially available anti-RBD antisera after an immunoprecipitation (IP) with OMVs derived from V111 pLOR-V and V111 p, E111 pLOR-E, and E111 p.
  • OMVs of the respective strains were co-incubated with the anti-RBD antisera (IgG), subsequently pelleted and washed by centrifugation steps and finally separated by SDS-PAGE for immunoblot analyses (for details please see “Materials and Methods” section).
  • FIGURE 2A-2B Design of the immunization study.
  • A Timeline is given by a horizontal arrow from left to the right. Each vertical bar marks the day of a bleed (B) or fecal pellet (F) collection. Mice received three intranasal immunizations starting with the initial immunization on day 0, followed by boosts on days 14 and 28.
  • Immunizations are indicated by I-1, I-2, and I-3 and are highlighted by vertical arrows.
  • Immunization groups V111 pLOR-V, V111 p, E111 pLOR-E, or E111 p received the OMVs of the respective donor strain in all three immunizations (I-1 to I-3).
  • EVE pLOR-E/V and EVE p were immunized with alternating regimen and received OMVs from E111 pLOR-E or E111 p on days 0 (I-1) and 28 (I-3) as well as OMVs from V111 pLOR-V or V111 p on day 14 (I-2).
  • a sham-immunized group receiving saline at days 0, 14, and 28 served as control.
  • FIGURE 3A-3F Temporal immune responses to V. cholerae and ETEC OMVs.
  • FIGURE 8 Genetically engineered detoxified triple mutants show increased vesiculation compared to the parental wild type strain.
  • FIGURE 9A-9B DNA and amino acid sequences of the codon-optimized Lpp- OmpARBD (LOR) fusion constructs for V. cholerae (A)(SEQ ID NO: 1 (DNA) and SEQ ID NO: 2 (amino acid)) and E. coli (B) (SEQ ID NO: 3 (DNA) and SEQ ID NO:4 (amino acid)).
  • FIG. 1B Shown is a representative immunoblot detecting the LOR fusion protein in OMVs derived from V ⁇ pLORV and V ⁇ p, E ⁇ pLOR-E and E ⁇ p.
  • B SDS-PAGE and Kang staining was executed in parallel with the same samples used for the immunoblot analyses provided in Fig. 1B and 10A and serves as a loading control. Molecular mass standards (PageRuler Prestained Protein Ladder – Thermo Fisher Scientific) are indicated on the left.
  • FIGURE 11 Protein profiles of the immunoprecipitation (IP).
  • FIGURE 12 Size distribution and biomass quantification of OMVs used in this study. Mean and mode OMV diameter sizes were measured by Zetasizer. Biomass quantification of OMV preparations derived from equivalent OD600 units of the respective cultures were analyzed for total protein (Bradford) or lipopolysaccharide (Purpald). Median with maximum and minimum values are shown (n ⁇ 5 biological replicates).
  • FIGURE 13 Controls for the neutralization assay.
  • SARS-CoV-2 includes membrane (M), spike (S), envelope (E), and nucleocapsid (N) structural proteins.
  • M membrane
  • S spike
  • E envelope
  • N nucleocapsid
  • the membrane, spike and envelope proteins provide the structure of the exterior viral envelope.
  • the spike protein mediates binding to the human ACE2 receptor triggering cell entry.
  • Spike is processed into two subunits, S1 and S2, which remain associated.
  • the S1 subunit has a compact C-terminal domain that is largely comprised of the receptor binding domain (RBD).
  • Outer membrane vesicles which are naturally released by gram-negative bacteria during growth, are comprised largely of outer membrane, lipids, lipopolysaccharide (LPS), integral membrane proteins, and lipoproteins.
  • OMVs possess characteristics that make them ideal candidates as a vaccine platform, including immunostimulatory pathogen-associated molecular patterns (PAMPs) and a size of 20–200 nm which allows them to drain freely into lymph nodes to target locally residing immune cells.
  • PAMPs pathogen-associated molecular patterns
  • OMV production strains can also be genetically engineered to express and decorate their surface with antigens of interest.
  • a composition comprising- from N terminus to C-terminus- the N-terminal signal sequence of the E. coli Lpp protein fused in frame either directly or via a linker to a fragment of the E. coli OmpA protein, wherein the fragment of the E. coli OmpA protein comprises a N-terminal deletion and a C-terminal deletion, the fragment of the E. coli OmpA protein fused either directly or via linker to a fragment of the SARS CoV-2 Spike protein comprising the receptor binding domain (RBD).
  • RBD receptor binding domain
  • the composition further comprises outer membrane vesicles (OMVs) and the fusion protein is present on the surface of the OMVs.
  • OMVs outer membrane vesicles
  • the fusion protein may be a codon optimized polynucleotide and comprise SEQ ID NO: 1 or SEQ ID NO: 2.
  • OMVs are nanosized proteoliposomes derived from the outer membrane of Gram- negative bacteria. OMVs are secreted via vesiculation of the outer membrane as part of the normal growth process. OMVs may be derived from any gram-negative bacteria, for example Salmonella, Neisseria, Bordetella or Yersinia enterocolitica.
  • the OMVs of the present disclosure may be derived from Vibrio cholerae (V.
  • the present disclosure employs the Lpp-OmpA fusion strategy for surface targeting.
  • the Lpp domain serves to target and anchor in outer membrane and the OmpA domain is required for the surface expression.
  • the LOR fusion constructs of the present disclosure consist of a N-terminal signal sequence and next nine amino acids of the major outer membrane lipoprotein (Lpp), followed by a two-amino-acid linker, and then approximately 110 amino acids of OmpA, i.e., amino acids 46–146 of ETEC and amino acids 51–158 of V. cholerae OmpA.
  • the Lpp fragment directs the fusion protein to the outer membrane, with the acylated N-terminal end being anchored in the inner leaflet of the outer membrane.
  • the OmpA fragment contains five membrane-spanning segments with the C-terminus residing externally. Via an additional four-amino-acid linker the heterologous peptide consisting of the entire RBM and the majority of the RBD of SARS-CoV-2 is fused to the C-terminal end.
  • Species-specific fusion constructs using the individual lpp and ompA sequences as well as codon optimized sequences of the SARS-CoV-2 RBD for each species may also be used.
  • V. cholerae or E. coli specific lpp and ompA are disclosed.
  • each segment of the fusion construct described herein may further include modifications in length or sequence, for example, additional amino acids in the linkers, to optimize function.
  • the composition is a fusion protein.
  • Fusion proteins or chimeric proteins are proteins created through the joining of two or more genes that originally coded for separate proteins. Translation of this fusion gene results in a single or multiple polypeptides with functional properties derived from each of the original proteins.
  • the fusion protein may comprise SEQ ID NO: 2 or SEQ ID NO: 4 or sequences 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical thereto, a fragment thereof, or combinations thereof.
  • the amino acid sequences of variants, mutants, or derivatives as contemplated herein may include conservative amino acid substitutions relative to a reference amino acid sequence.
  • a variant, mutant, or derivative protein may include conservative amino acid substitutions relative to a reference molecule.
  • conservative amino acid substitutions are those substitutions that are a substitution of an amino acid for a different amino acid where the substitution is predicted to interfere least with the properties of the reference polypeptide. In other words, conservative amino acid substitutions substantially conserve the structure and the function of the reference polypeptide.
  • Conservative amino acid substitutions generally maintain (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a beta sheet or alpha helical conformation, (b) the charge or hydrophobicity of the molecule at the site of the substitution, and/or (c) the bulk of the side chain.
  • Non-conservative amino acids typically disrupt (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a beta sheet or alpha helical conformation, (b) the charge or hydrophobicity of the molecule at the site of the substitution, and/or (c) the bulk of the side chain.
  • Disclosed proteins, mutants, or variants, described herein may have one or more functional or biological activities exhibited by a reference polypeptide (e.g., one or more functional or biological activities exhibited by wild-type protein, or the unmodified fusion).
  • the activity of the variant or mutant protein may have an activity that is enhanced, as compared to a comparable wild-type or control protein, or may have an alternative or a modified activity as compared to a comparable or wild-type or control protein.
  • the fusion protein described herein may be expression by an expression vector.
  • An expression vector is any particle used as a vehicle to artificially carry a foreign nucleic sequence, typically DNA into another cell, where it can be replicated and/or expressed.
  • a vector containing foreign DNA is termed recombinant DNA.
  • the four major types of vectors are plasmids, viral vectors, cosmids, and artificial chromosomes.
  • Expression constructs comprise a heterologous promoter and the nucleic acid sequence encoding protein of interest (e.g., SEQ ID NO: 1 or SEQ ID NO: 3, sequences 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical thereto, or nucleic acid sequence encoding the amino acid sequence of SEQ ID NO: 2 and SEQ ID NO: 4) which is capable of expression in the cell in which it is introduced.
  • protein of interest e.g., SEQ ID NO: 1 or SEQ ID NO: 3, sequences 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical thereto, or nucleic acid sequence encoding the amino acid sequence of SEQ ID NO: 2 and SEQ ID
  • the expression constructs include vectors which are capable of directing the expression of exogenous genes to which they are operatively linked. Such vectors are referred to herein as “recombinant constructs,” “expression constructs,” “recombinant expression vectors” (or simply, “expression vectors” or “vectors”) and may be used interchangeably. Suitable vectors are known in the art and contain the necessary elements in order for the gene encoded within the vector to be expressed as a protein in the host cell.
  • vector'' refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • vector refers to a circular double stranded DNA loop into which additional DNA segments may be ligated, specifically exogenous DNA segments encoding the mutant ⁇ -gal protein.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • Viral vectors are incorporated into viral particles that are then used to transport the viral polynucleotide encoding the protein of interest into the target cells.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced.
  • Other vectors can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome (e.g., lentiviral vectors).
  • vectors are capable of directing the expression of exogenous genes to which they are operatively linked.
  • vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • Suitable vectors include, but are not limited to, plasmids, expression vectors, lentiviruses (lentiviral vectors), adeno- associated viral vectors (rAAV), among others and includes constructs that are able to express the protein of interest.
  • a preferred vector is a lentiviral vector or adeno-associated vector.
  • Suitable methods of making viral particles are known in the art to be able to transform cells in order to express the protein of interest described herein.
  • the fusion protein is encoded by a polynucleotide.
  • polynucleotide is used herein interchangeably with the term “nucleic acid” and refers to an organic polymer composed of two or more monomers including nucleotides, nucleosides or analogs thereof, including but not limited to single stranded or double stranded, sense or antisense deoxyribonucleic acid (DNA) of any length and, where appropriate, single stranded or double stranded, sense or antisense ribonucleic acid (RNA) of any length, including siRNA.
  • DNA single stranded or double stranded
  • RNA ribonucleic acid
  • nucleotide refers to any of several compounds that consist of a ribose or deoxyribose sugar joined to a purine or a pyrimidine base and to a phosphate group, and that are the basic structural units of nucleic acids.
  • nucleoside refers to a compound (as guanosine or adenosine) that consists of a purine or pyrimidine base combined with deoxyribose or ribose and is found especially in nucleic acids.
  • nucleotide analog or “nucleoside analog” refers, respectively, to a nucleotide or nucleoside in which one or more individual atoms have been replaced with a different atom or with a different functional group.
  • polynucleotide includes nucleic acids of any length, including DNA, RNA, ORFs, analogs and fragments thereof.
  • the polynucleotide is codon optimized. Codon optimization is a process to improve gene expression and increase translational efficiency by accommodating codon bias of the host organism.
  • an expression vector is introduced into a cell for purposes of expressing the fusion protein of the expression vector.
  • the cell is V. cholerae or E. coli.
  • the expression vector can be introduced into the cell by any means known in the art. These means may include transfection or transduction. Transfection is the process of introducing nucleic acids into cells by non-viral methods. Transduction is the process whereby foreign DNA is introduced into another cell via a viral vector. These are common tools to introduce a foreign gene into host cells. Among others, additional means include transformation, and conjugation. In some embodiments the OMVs may be administered to induce an immune response. As used herein, the term "administering" an agent, such as a therapeutic entity to an animal or cell, is intended to refer to dispensing, delivering or applying the substance to the intended target.
  • administering is intended to refer to contacting or dispensing, delivering or applying the therapeutic agent to a subject by any suitable route for delivery of the therapeutic agent to the desired location in the animal, including delivery by either the parenteral or oral route, intramuscular injection, subcutaneous/intradermal injection, intravenous injection, intrathecal administration, buccal administration, transdermal delivery, topical administration, and administration by the intranasal or respiratory tract route.
  • the composition may be administered intranasally.
  • the compound(s) can be conveniently delivered in the form of an aerosol spray from pressurized packs or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, fluorocarbons, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, fluorocarbons, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch
  • a powder mix of the compound and a suitable powder base such as lactose or starch
  • suitable powder base such as lactose or starch
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated and/or eradication or amelioration of one or more of the symptoms associated with the underlying disorder such that the patient reports an improvement in feeling or condition, notwithstanding that the patient may still be afflicted with the underlying disorder.
  • Therapeutic benefit also generally includes halting or slowing the progression of the disease, regardless of whether improvement is realized.
  • the amount of composition administered will depend upon a variety of factors, including, for example, the particular indication being treated, the mode of administration, whether the desired benefit is prophylactic or therapeutic, the severity of the indication being treated and the age and weight of the patient, the bioavailability of the particular composition, the conversion rate and efficiency of delivery under the selected route of administration, etc. Determination of an effective dosage for a particular use and mode of administration is well within the capabilities of those skilled in the art. Effective dosages may be estimated initially from in vitro activity and metabolism assays.
  • an initial dosage for use in animals may be formulated to achieve a circulating blood or serum concentration of the composition that is at or above an IC 50 of the particular composition as measured in an in vitro assay. Calculating dosages to achieve such circulating blood or serum concentrations taking into account the bioavailability of the particular composition via the desired route of administration is well within the capabilities of skilled artisans. Initial dosages can also be estimated from in vivo data, such as animal models. Animal models useful for testing the efficacy of the active metabolites to treat or prevent the various diseases described above are well-known in the art. Animal models suitable for testing the bioavailability and/or metabolism of compositions are also well-known. Ordinarily skilled artisans can routinely adapt such information to determine dosages suitable for human administration.
  • an immune response is induced.
  • An immune response involves many cells and proteins of the immune system and is a reaction which occurs within an organism for the purpose of defending against substances it sees as harmful or foreign.
  • An immune response may comprise the activation of T or B cells, phagocytes or natural killer cells, and may include the production of cytokines or antibodies.
  • the immune response comprises an antibody response against SARS CoV- 2 spike protein as part of a humoral immune response.
  • Antibody responses are generated by B lymphocytes and plasma cells and result in the appearance of immunoglobulin classes each of which bears identical antibody-combining sites.
  • the antibody response is a neutralizing antibody response.
  • a neutralizing antibody is an antibody that defends a cell from a pathogen or infectious particle by neutralizing any effect it has biologically. Neutralization renders the particle no longer infectious or pathogenic.
  • the terms “a”, “an”, and “the” mean “one or more.”
  • a molecule should be interpreted to mean “one or more molecules.”
  • “about”, “approximately,” “substantially,” and “significantly” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which they are used.
  • Example 1 An intranasal vaccine based on outer membrane vesicles against SARS- CoV-2 Reference is made to the manuscript: Thapa et al., "An intranasal vaccine based on outer membrane vesicles against SARS-CoV-2," Front.
  • OMV outer membrane vesicle
  • RBD receptor binding domain
  • S1 and S2 which remain associated.
  • the S1 subunit has a compact C-terminal domain that is largely composed of the RBD, which includes the receptor binding motif (RBM) directly binding the human ACE2 receptor.
  • OMVs possess characteristics that make them ideal candidates as a vaccine platform (Kulp and Kuehn, 2010; van der Pol et al., 2015; Pathirana and Kaparakis-Liaskos, 2016).
  • PAMPs pathogen-associated molecular patterns
  • lipid A lipid A
  • porins lipid A
  • lipoproteins primarily lipid A, porins, and lipoproteins.
  • PRRs pathogen recognition receptors
  • the OMV diameter of 20– 200 nm allows them to drain freely into lymph nodes to target locally residing immune cells (van der Pol et al., 2015; Pathirana and Kaparakis-Liaskos, 2016).
  • OMV production strains can be genetically engineered to express and decorate their surface with antigens of interest (Gerritzen et al., 2017).
  • cholerae and enterotoxigenic Escherichia coli ETEC
  • genetic modification of lipid A resulted in less endotoxicity without diminishing the immunogenic potential (Leitner et al., 2013, 2015).
  • both bacterial species have been successfully genetically engineered to produce OMVs loaded with antigens of interest (Leitner et al., 2015; Gnopo et al., 2017).
  • OMVs released by V. cholerae and ETEC could be efficiently decorated with the C-terminal part of the SARS- CoV-2 Spike protein S1 containing the RBD.
  • Mice immunized with OMVs decorated with Lpp-OmpA-RBD (LOR) fusion protein induced a robust immune response not only against the bacterial surface components, but also against the Spike protein.
  • SARS-CoV-2 neutralizing antibodies were confirmed in cell culture infection assays using the lentiviral SARS-CoV-2 pseudovirus in combination with 293T cells engineered to express the SARS- CoV-2 receptor ACE2.
  • Materials and Methods Bacterial Strains, Cell Lines and Growth Conditions Bacterial strains, cell lines and plasmids used in this study are listed in Table 1; oligonucleotides are listed in Table 2.
  • AC53 a spontaneous streptomycin (Sm)-resistant mutant of the clinical isolate E7946 (O1 El Tor Ogawa), or ETEC H10407-S, a Sm-resistant mutant of the clinical isolate H10407, were used as wild-type strains (V-WT and E-WT).
  • coli strain DH5 ⁇ pir and SM10 ⁇ pir were used for genetic manipulations. Unless stated otherwise, strains were cultivated in Lysogeny broth (LB) or on LB agar plates with aeration at 37°C. If required, antibiotics and other supplements were used in the following final concentrations: streptomycin (Sm), 100 ⁇ g/ml; ampicillin (Ap), 100 ⁇ g/ml or in combination with other antibiotics 50 ⁇ g/ml; kanamycin (Km), 50 ⁇ g/ml; IPTG, 0.1 mM; glucose (Gluc), 0.2%; and sucrose (Suc), 10%. Table 1. Bacterial strains, cell lines and plasmids used in this study Table 2.
  • Qiagen plasmid kits were used for isolation of plasmid DNA; QIAquick® Gel extraction and QIAquick® PCR Purification kits (Qiagen) were used for purifying DNA fragments. PCR reactions for subcloning were carried out using the Q5® High-Fidelity DNA Polymerase (NEB), while Taq DNA Polymerase (NEB) was used for all other PCRs. Construction of ompA in-frame deletion mutants in V. cholerae and ETEC were carried out as described by Donnenberg and Kaper (1991) using derivatives of pCVD442, i.e., p ⁇ ompA-V or p ⁇ ompA-E.
  • the suicide vector p ⁇ ompA-V was already available from a previous study (Song et al., 2008).
  • p ⁇ ompA-E ⁇ 800 bp PCR fragments located up- and downstream of the ompA were amplified using the oligonucleotide pairs ompA_E_SacI_1 and ompA_E_EcoRI_2 as well as ompA_E_EcoRI_3 and ompA_E_XbaI_4 with chromosomal DNA from E-WT as template (Table 2).
  • pCVD442 After digestion of the PCR fragments with the appropriate restriction enzyme (NEB) indicated by the name of the oligonucleotide, they were ligated into pCVD442, which was digested with the appropriate restriction enzymes. Unless noted otherwise, ligation products were transformed into DH5 ⁇ pir and ApR colonies were characterized for the correct constructs by PCR. To obtain deletion strains, generated derivatives of pCVD442 were transformed into E. coli Sm10 ⁇ pir and conjugated into V. cholerae or ETEC. Exconjugants were purified by SmR/ApR selection. Sucrose selection was used to obtain ApS colonies and chromosomal deletions were confirmed by PCR, respectively.
  • NEB restriction enzyme
  • Lpp-OmpA-RBDVch LORVch
  • Lpp-OmpA-RBDETEC LORETEC
  • fusion proteins were designed in silico and the corresponding sequences including a 5′ untranslated region harboring a unique KpnI restriction site and an optimal Shine-Dalgarno sequence (Fig.9) were synthesized and subcloned into the standard vector system pMK by the GeneArt Gene Synthesis platform (Thermo Fisher Scientific). Thus, constructs were provided as pMK-V-LOR and pMK-E-LOR.
  • the expression plasmid pLOR-V and pLOR-E were constructed using the oligonucleotides LOR_V_1 and LOR_V_BamHI_2 as well as LOR_E_1 and LOR_E_BamHI_2 for amplifying the fusion construct using the ordered plasmids as template. This added a C-terminal FLAG-tag epitope to the constructs.
  • the resulting PCR fragments were purified, digested with KpnI and BamHI, and ligated into pMMB67EH, which has been digested with the same enzymes. Ligation products were transformed into DH5 ⁇ pir and ApR colonies were characterized for the correct constructs by PCR.
  • the OMVs present in the supernatant were pelleted through subsequent ultracentrifugation (150,000 ⁇ g, 4°C, 4 h) and resuspended in saline to generate a 1000-fold concentrated OMV suspension compared to the original filter-sterilized supernatant. Protein concentration was determined using Bradford assay (Bio-Rad Laboratories, Protein Assay Dye Reagent) according to the manufacturer’s manual. To quantify the LPS content of OMVs, purpald assays were performed as described previously using 3-deoxy-D-mannooctulosonic acid (Kdo) (Sigma- Aldrich) as a standard (Roier et al., 2016).
  • Kdo 3-deoxy-D-mannooctulosonic acid
  • mice Female BALB/c mice (Charles River Laboratories) were used in all experiments in accordance with the rules of the ethics committee at the University of Graz and the corresponding animal protocol, which has been approved by the federal ministry BMBWF (protocol: 39/12/75ex2017/18). Mice were housed with food and water ad libitum and monitored under the care of full-time staff. All animals were acclimated for 1 week before any procedures were carried out and were approximately 8 weeks old at the start of the immunization.
  • Blood samples as well as fecal pellets were collected from immunized and sham- immunized adult mice throughout the immunization study and processed as previously described to monitor the induced immune response (Schild et al., 2008, 2009). Briefly, blood samples were collected from immunized and sham immunized mice on days 0, 14, 28, 42, and 60. The collected blood was allowed to clot at room temperature (RT) for 30 min after which serum was isolated by removing the blood clot using a sterile toothpick followed by centrifugation (15 min, 1,000 ⁇ g).
  • RT room temperature
  • the supernatant representing the serum of each sample was removed, diluted threefold in PBS/sodium azide (0.05%), and subsequently stored at ⁇ 80°C.
  • aliquots of the sera collected on day 60 were stored at ⁇ 80°C without any dilution or addition of sodium azide of to be used in cell culture neutralization assays.
  • freshly voided fecal pellets collected on days 42 and 60 were vacuum-dried for 10 min before their weight was recorded.
  • Immunoglobulins were extracted by adding 1 ml of extraction buffer [PBS, 0.01% sodium azide, 5% fetal calf serum, 1 tablet complete EDTA-free protease inhibitor cocktail (Roche) per ml] per 100 mg dry- weight feces. After vortexing the samples for 15 min at 4°C, solid material was separated by centrifugation (2 min, 13,000 ⁇ g) and the supernatants were stored at ⁇ 80°C.
  • extraction buffer [PBS, 0.01% sodium azide, 5% fetal calf serum, 1 tablet complete EDTA-free protease inhibitor cocktail (Roche) per ml] per 100 mg dry- weight feces. After vortexing the samples for 15 min at 4°C, solid material was separated by centrifugation (2 min, 13,000 ⁇ g) and the supernatants were stored at ⁇ 80°C.
  • the OMV pellet was washed once with 1 ml saline and centrifuged again (75,600 ⁇ g, 2 h, 4°C) before the sample was finally resuspended in 40 ⁇ l saline and subjected to immunoblot analyses.
  • Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis and Immunoblot Analysis To analyze the protein content of OMVs, the standard sodium dodecyl sulfate- polyacrylamide gel electrophoresis (SDS-PAGE) procedure in combination with 12% gels and the PageRuler Prestained Protein Ladder (Thermo Fisher Scientific) as a molecular mass standard were used.
  • Enzyme-Linked Immunosorbent Assay Temporal immune responses of different Igs, half-maximum total Ig titers and mucosal immune responses to V ⁇ or E ⁇ OMVs (5 ⁇ g/ml in PBS, pH 7.4) as well as the determination of the half-maximum total Ig titers to SARS-CoV-2 Spike Protein S1 (5 ⁇ g/ml in PBS, pH 7.4, RP-87679 Thermo Fisher Scientific) were carried out essentially as described previously (Roier et al., 2012; Leitner et al., 2013, 2015) using appropriate purified mouse Ig isotype standard (IgA, 553476; IgG1, 557273; or IgM, 550963, BD Biosciences) as well as horseradish peroxidase-conjugated goat anti-mouse antibodies (IgA, 62-6720; IgG1, A10551; IgM, 62-6820; IgG, IgM,
  • Optical densities were monitored at 450 nm with a FLUOstar Omega microplate reader (BMG Labtech). Starting dilutions of the mouse sera were 1:10 for quantification of the temporal immune responses and half-maximum total Ig titers against the Spike protein, 1:100 for quantification of the half-maximum total Ig titers against OMVs, or 1:400 for quantification of the temporal immune responses against OMVs. Starting dilutions of the fecal pellet extracts for quantification of the mucosal immune responses against OMVs were 1:10.
  • each plate was incubated in the dark for 2 min, followed by measuring relative light units (RLU) on a SynergyTM HT Microplate Reader using Gen5 data analysis software (BioTek). Background was initially assayed from uninfected cells, virus in the absence of cells, or DMEM medium alone, and all were equivalent. Therefore, in subsequent experiments, each plate contained at least two DMEM alone wells.
  • the maximum infection on each plate was measured by infecting at least four wells of HEK 293T-ACE2 cells with pseudotype virus that had not been neutralized with serum. The fraction infectivity was calculated by averaging technical replicates and dividing by the average of the maximum infectivity on that plate.
  • the inhibitory concentration 50% was calculated graphically.
  • Data Presentation and Statistical Analysis The data is generally presented as median with interquartile range as some data sets were not normally distributed. Consequently, data were analyzed using the Mann–Whitney U test or a Kruskal–Wallis test followed by post hoc Dunn’s multiple comparisons. Differences were considered significant at p-values of ⁇ 0.05.
  • GraphPad Prism version 8 was used for all statistical analyses. Results Generation of V. cholerae and Enterotoxigenic Escherichia coli Outer Membrane Vesicles Decorated With the SARS-CoV-2 Receptor Binding Domain Antigen We recently demonstrated that OMVs derived from ETEC or V.
  • cholerae lacking one functional secondary lipid A acyltransferase, MsbB (also referred to as LpxN) retain their potential to induce a high-titer, protective immune response but show a significantly reduced pro-inflammatory cytokine induction suggesting lower endotoxicity (Leitner et al., 2013, 2015).
  • cholera toxin subunits A and B in V. cholerae, and the heat-labile enterotoxin catalytic A subunit in E. coli were deleted. Thus, presence of these secreted toxins in OMVs is precluded (Leitner et al., 2015).
  • the LOR fusion constructs consist of a N-terminal signal sequence and next nine amino acids of the major outer membrane lipoprotein (Lpp), followed by a two-amino- acid linker, and then approximately 110 amino acids of OmpA, i.e., amino acids 46–146 of ETEC and amino acids 51–158 of V. cholerae OmpA.
  • Lpp major outer membrane lipoprotein
  • the Lpp fragment directs the fusion protein to the outer membrane, with the acylated N-terminal end being anchored in the inner leaflet of the outer membrane.
  • the OmpA fragment contains five membrane-spanning segments with the C-terminus residing externally (Francisco et al., 1992).
  • the heterologous peptide consisting of the entire RBM and the majority of the RBD of SARS-CoV-2 is fused to the C-terminal end.
  • Lpp and OmpA of V. cholerae and ETEC share only 38% identity
  • the amino acid sequence as well as the SARS-CoV-2 Spike protein is neither optimal for E. coli nor V. cholerae; we have used species-specific fusion constructs using the individual lpp and ompA sequences as well as codon optimized sequences (obtained via the IDT codon optimization tool) of the SARS- CoV-2 RBD for each species, namely, LOR-V and LOR-E, respectively ( Figure 9).
  • LOR fusion proteins in V ⁇ and E ⁇ should result in their localization on the bacterial cell surface and therefore also on the surface of OMVs, which bud from the outer membrane. Successful localization of LOR in OMVs was confirmed by immunoblot analyses ( Figure 1B and Figure 10). Detection of LOR fusion proteins with anti-FLAG and anti-RBD antibodies showed specific bands with the expected molecular weight of approximately 36 kDa for V. cholerae and 38 kDa for ETEC for the OMVs derived from V ⁇ pLOR-V and E ⁇ pLOR-E, but not for OMVs isolated from strains harboring the empty vector (V ⁇ p and E ⁇ p).
  • OMVs of both species can be decorated with significant amounts of the LOR. Furthermore, outward exposure of RBD part on OMVs was confirmed via an immunoprecipitation assay using OMVs derived from V ⁇ pLOR-V, V ⁇ p, E ⁇ pLOR-E, and E ⁇ p as antigen in combination with anti-RBD antibodies. After incubation of OMVs and anti-RBD antibodies, samples were subjected to centrifugation and washing steps to purify OMVs. Finally, immunoblot analyses with the immunoprecipitated samples were performed to detect anti-RBD antibody (raised in rabbits) bound to the OMV samples using an HRP-conjugated anti-rabbit antisera (Figure 1C).
  • mice were intranasally immunized on days 0, 14, and 28 receiving 25 ⁇ g OMVs (protein equivalent) per immunization.
  • OMVs protein equivalent
  • EVE pLOR-E/V and EVE p Two groups (EVE pLOR-E/V and EVE p) were immunized with alternating regimen and received OMVs from E ⁇ pLOR-E on days 0 and 28 as well as OMVs from V ⁇ pLOR-V on day 14 (EVE pLOR-E/V) or E ⁇ p on days 0 and 28 as well as V ⁇ p on day 14 (EVE p).
  • a change of the OMV donor species between the immunizations was thought to specifically boost the immune response against the RBD of SARS-CoV-2 being the common antigen on OMVs derived from V ⁇ pLOR-V and E ⁇ pLOR-E.
  • a sham-immunized group receiving saline at days 0, 14, and 28 served as control.
  • OMVs derived from V ⁇ pLOR-V or E ⁇ pLOR-E were used as coating material allowing the detection of temporal IgM, IgG1, and IgA responses to the respective OMVs ( Figure 3).
  • IgG1 the dominant IgG subclass induced after intranasal immunization with OMVs (Schild et al., 2008, 2009; Bishop et al., 2010; Leitner et al., 2013, 2015).
  • immunization with V. cholerae or ETEC OMVs induces a species-specific immune response without any significant cross-reactivity (Leitner et al., 2015).
  • IgM titers peaked at days 28 or 42, while IgG and IgA were slightly delayed and peaked on days 42 or 60.
  • Ig titers of the immunization groups receiving the same OMVs at all three immunization were overall higher and reached the Ig peak earlier than the immunization groups with alternating regimen.
  • IgG1 and IgA titers remained stable or even increased between days 42 and 60, which indicates the induction of a robust immune response until day 60, the designated endpoint of this study.
  • the OMV-specific responses were slightly lower in the immunization groups with alternating regimen (EVE pLOR-E/V and EVE p) compared to the immunization groups receiving the same OMVs at all three immunizations.
  • the EVE pLOR-E/V immunization group showed slightly, but not significantly higher Ig titers against the Spike protein than the V ⁇ pLOR-V and E ⁇ pLOR-E groups.
  • the immune response against the Spike protein remained undetectable or at low levels for immunization groups receiving OMVs from strains harboring the empty vector (V ⁇ p, E ⁇ p and EVE p).
  • isotype-specific and total Ig titers did not massively drop between days 42 and 60 in the respective immunization groups suggesting the induction of a stable immune response (Figures 3–5).
  • OMVs derived from V. cholerae and ETEC have been chosen as platform as both are highly immunogenic and have been successfully used in previous immunization studies (Schild et al., 2008, 2009; Bishop et al., 2010; Leitner et al., 2013, 2015).
  • Parental strains of V. cholerae and ETEC have been optimized to exhibit increased OMV release, lack the enterotoxins, and show reduced LPS reactivity.
  • the genetically engineered OMV production strains were successfully used for heterologous antigen expression to generate detoxified OMVs decorated with the RBD portion of the SARS- CoV-2 Spike protein.
  • intranasal immunization with OMVs induced a robust immune response against surface structures of the respective donor bacteria, but only mice immunized with RBD-decorated OMVs induced a significant Spike protein-specific immune response.
  • neutralizing activity was only observed for sera from mice immunized with RBD-decorated OMVs.
  • the results from the Spike-pseudotyped lentivirus neutralization assay indicate the induction of a neutralizing-antibody response upon intranasal immunization with RBD-decorated OMVs.
  • An interesting and potentially important finding was the increased efficacy of the alternating immunization regimen using RBD-decorated OMVs derived from ETEC and from V.
  • the alternating regimen reduces the immunization with OMVs derived from ETEC to two doses and OMVs derived from V. cholerae to one dose.
  • OMVs derived from ETEC to two doses
  • OMVs derived from V. cholerae to one dose.
  • the study raises several questions to be addressed in future studies, such as the longevity of the immune response, the optimal OMVs amount per immunization dose, and number of booster immunizations.
  • Previous reports demonstrated that a two-dose immunization schedule with V. cholerae OMVs is sufficient to induce a long-lasting, protective immune response against the bacterial pathogen (Bishop et al., 2010).
  • OMV-based SARS- CoV-2 vaccine candidate we focused on the original Wuhan Hu-1 isolate. Future studies need to address the neutralization activity against epidemiologically relevant SARS-CoV-2 variants with RBM mutations, which have been emerging in the last months. However, even if a reduced efficacy would be observed, the fusion strategy presented herein is adaptable to decorate OMVs with appropriate antigens upon emergence of new viral variants.
  • Some OMV-based vaccine technologies purify the OMVs and the heterologous antigens separately in a first step to subsequently coat or load the vesicles in a second step (Li and Liu, 2020; Zingl et al., 2021a).
  • heterologous antigen directly in the bacterial OMV donor species simplifies the production process and may reduce costs.
  • the decorated OMVs could be used for immunization without any further manipulation.
  • host-specific post-translational modifications are lacking along the bacterial expression system., e.g., glycosylation or disulfide bonds.
  • V. cholerae and ETEC encode periplasmic enzymes for catalyzing disulfide bond formation and isomerization, the incorrect formation of disulfide bonds cannot be excluded.
  • SARS- CoV-2 is a mucosal pathogen of the nasopharynx and respiratory tract. It is becoming increasingly evident that a mucosal IgA response could be valuable for protection against SARS-CoV-2, vaccine efficacy and clearance of the virus from the infection site (Quinti et al., 2021; Wang et al., 2021). Indeed, IgA antibodies have been shown to neutralize the virus via binding to the RBD of SARS-CoV-2 (Ma et al., 2020; Yu et al., 2020; Sterlin et al., 2021).
  • lung lavage might have been a better option to analyze mucosal immune responses and should be preferred in future studies. It must be noted that lung lavage was not approved by our current animal protocol and is an endpoint assay requiring killing of the mice, while fecal pellet extraction allowed us to maintain the animals and do temporal studies, i.e., collection on days 42 and 60. Thus, we cannot exclude that other administration routes, e.g., subcutaneous, intramuscular or oral, or combinations thereof would improve the immunization efficacy, which also needs to be addressed in future comparative studies. Several widely used vaccines have demonstrated high protective efficacy against COVID-19.
  • OMV vaccine lots have been demonstrated to be stable for months even at room temperature and do not require stabilization additives (Vyssokikh et al., 2002; Bishop et al., 2010).
  • OMVs also have advantages in terms of vaccine production.
  • donor bacteria are easy to cultivate and replicate fast.
  • mutations e.g., deletion of ompA, that result in hypervesiculating strains (Song et al., 2008; Schwechheimer and Kuehn, 2015), which in this study increased the yield of OMVs by approximately fivefold. Based on the yields presented herein, approximately 8–10 mg protein equivalent of OMVs can be isolated per 1 L of culture volume.
  • OMVs protein equivalent
  • the OMVs isolated from 1 L last for more than 100 animals.
  • OMVs are non-replicative and their isolation does not require treatment with inactivating agents, thus preserving the native state of antigens.
  • mice with Vibrio cholerae outer-membrane vesicles protects against hyperinfectious challenge and blocks transmission.
  • the coronavirus spike protein is a class I virus fusion protein: structural and functional characterization of the fusion core complex. J. Virol. 77, 8801–8811.
  • Vaccine profile of 4CMenB a four-component Neisseria meningitidis serogroup B vaccine. Exp. Rev. Vaccines 13, 193–202. doi: 10.1586/14760584.2014.874949 Evans, D. J. Jr., and Evans, D. G. (1973).

Abstract

Disclosed are outer membrane vesicle (OMV)-based vaccines against sudden acute respiratory syndrome coronavirus-2 (SARS-CoV-2) or COVID 19. The OMVs comprise a fusion protein comprising at least a fragment of the OmpA protein fused to at least a fragment of the SARS-CoV-2 Spike protein comprising the receptor binding domain (RBD).

Description

OUTER MEMBRANE VESICLE (OMV)-BASED VACCINES AGAINST SUDDEN ACUTE RESPIRATORY SYNDROME CORONAVIRUS 2 (SARS-COV- 2) SPIKE PROTEIN CROSS-REFERENCE TO RELATED APPLICATIONS This patent application claims the benefit of priority of United States Provisional Patent Application No.63/261,027 filed on September 9, 2021, which is incorporated herein by reference in its entirety. REFERENCE TO AN ELECTRONIC SEQUENCE LISTING The contents of the electronic sequence listing (166118.01217.xml; Size: 24,252 bytes; and Date of Creation: September 9, 2022) is herein incorporated by reference in its entirety. FIELD The field of the invention relates to vaccines. In particular, the field of the invention relates to outer membrane vesical (OMV)-based vaccines against sudden acute respiratory syndrome coronavirus-2 (SARS-CoV-2) or COVID 19. BACKGROUND The prevailing pandemic of SARS-CoV-2 highlights the desperate need of alternative vaccine-platforms, which are safe, effective, and can be modified to carry antigens of emerging pathogens. The current SARS-CoV-2 vaccines based on mRNA and adenoviral vector technology meet some of these criteria but still face limitations regarding administration route, mass production, stability, and storage. SUMMARY The inventors have developed a novel vaccine to protect against SARS-CoV-2 (COVID 19). The inventors added the SARS-CoV-2 Spike protein Receptor-Binding Domain (RBD) as a heterologous antigen to outer membrane vesicles (OMVs) produced by bacterial. The inventors showed that the vaccine elicits neutralizing antibodies in mice. Two types of vaccines were made using outer membrane vesicles (OMV) produced by bacteria, and both types were shown to elicit neutralizing antibodies in mice. One vaccine was made using OMVs produced by the bacterium Vibrio cholerae, and the second vaccine was made using OMVs produced by the bacterium Escherichia coli. The bacterial strains used to produce the OMVs express a protein fusion of the RBD and the protein OmpA. The OmpA protein in both of V. cholerae and E. coli is a major outer membrane protein, and thus is present in multiple copies in OMVs produced by the strains. Each OmpA-RBD fusion was designed so that the RBD portion is exposed on the outer surface of the OMVs where it can interact with immune cells. Advantages of the disclosed OMV-based vaccines are that they can be administered intranasally as opposed to by needle injection. In addition, the disclosed OMV-based vaccines exhibit heat stability as opposed to, for example, RNA-based vaccines which require deep cold storage and transportation. As such, the disclosed OMV-based vaccines may be useful in low-income, resource-poor nations, where cold storage and transportation are limited. Disclosed herein are composition comprising a fusion protein, the fusion protein comprising from N-terminus to C-terminus: the N-terminal signal sequence of the E. coli Lpp protein fused in frame either directly or via a linker to a fragment of the E. coli OmpA protein, wherein the fragment of the E. coli OmpA protein comprises a N-terminal deletion and a C-terminal deletion, and the fragment of the E. coli OmpA protein is fused either directly or via linker to a fragment of the SARS CoV-2 Spike protein comprising the receptor binding domain (RBD). In some embodiments, the compositions comprises outer membrane vesicles (OMVs) and the fusion protein is present on the surface of the OMVs. In some embodiments, the OMVs are derived from V. cholerae. In some embodiments, the OMVs are derived from E. coli. Also disclosed herein are expression vectors that expresses the fusion proteins disclosed herein. In some embodiments, the fusion protein is encoded by a polynucleotide that comprises codons that are optimized for expression in V. cholerae. In some embodiments, the fusion protein is encoded by a polynucleotide that comprises codons that are optimized for expression in E. coli. Also disclosed herein are microorganisms transfected with expression vectors comprising fusion proteins. By way of example, in some embodiments, a V. cholerae cell transfected with an expression vector that expresses the fusion protein described herein is provided. In some embodiments, an E. coli cell transfected with an expression vector that expresses the fusion protein described herein is provided. Also disclosed herein are methods for inducing an immune response against SARS CoV-2 Spike protein, the method comprising administering the composition comprising the fusion protein to a subject in need thereof. In some embodiments, the immune response comprises an antibody response against SARS CoV-2 Spike protein. In some embodiments, the immune response comprises a neutralizing antibody response against SARS CoV-2 Spike protein. In some embodiments, the method induces a protective response against infection by SARS CoV-2. Also disclosed herein are composition comprising a fusion protein, the fusion protein comprising SEQ ID NO: 2 or SEQ ID NO: 4. Also disclosed herein are methods for inducing an immune response against SARS CoV-2 Spike protein, the method comprising administering the composition comprising the fusion protein of SEQ ID NO: 2 or SEQ ID NO: 4 to a subject in need thereof. In some embodiments, the composition comprises SEQ ID NO: 2 and SEQ ID NO: 4. BRIEF DESCRIPTION OF THE DRAWINGS FIGURE 1A-1C. Decoration of OMVs with RBD antigens via a Lpp-OmpA fusion strategy. (A) Schematic design of the Lpp-OmpA-RBD (LOR) fusion constructs (top) and expected localization in the OMVs (bottom). Relevant parts of the LOR include the Lpp fragment (light blue), linkers (green), OmpA fragment (red), theSARS-CoV-2 RBD fragment (yellow), and the FLAG-tag (pink). Panel (B) shown is a representative immunoblot detecting the LOR fusion protein in OMVs derived from V111pLOR-V and V111 p, E111 pLOR-E, and E111 p. The commercially available anti-RBD antisera specifically detecting the SARS-CoV-2 Spike protein was used for this immunoblot. A Kang-stained SDS gel executed in parallel served as loading control (Figure 10). Panel (C) shown is a representative immunoblot detecting the heavy chain (approx. 55 kDa) of the commercially available anti-RBD antisera after an immunoprecipitation (IP) with OMVs derived from V111 pLOR-V and V111 p, E111 pLOR-E, and E111 p. OMVs of the respective strains were co-incubated with the anti-RBD antisera (IgG), subsequently pelleted and washed by centrifugation steps and finally separated by SDS-PAGE for immunoblot analyses (for details please see “Materials and Methods” section). The commercially available HRP-linked anti-rabbit antibody detecting the heavy chain of the anti-RBD IgG raised in rabbits was used for this immunoblot. A Kang-stained SDS gel executed in parallel served as loading control (Figure 11). (B,C) Molecular mass standards (Prestained Protein Marker Broad Range–New England Biolab) are indicated on the left. FIGURE 2A-2B. Design of the immunization study. (A) Timeline is given by a horizontal arrow from left to the right. Each vertical bar marks the day of a bleed (B) or fecal pellet (F) collection. Mice received three intranasal immunizations starting with the initial immunization on day 0, followed by boosts on days 14 and 28. Immunizations are indicated by I-1, I-2, and I-3 and are highlighted by vertical arrows. (B) The study comprised seven different immunization groups (n = 7 for each group) being named after the donor strains of the OMVs used for immunization, i.e., V111 pLOR-V, V111 p, E111 pLOR-E, E111 p, EVE pLOR-E/V, and EVE p. Immunization groups V111 pLOR-V, V111 p, E111 pLOR-E, or E111 p received the OMVs of the respective donor strain in all three immunizations (I-1 to I-3). The immunization groups EVE pLOR-E/V and EVE p were immunized with alternating regimen and received OMVs from E111 pLOR-E or E111 p on days 0 (I-1) and 28 (I-3) as well as OMVs from V111 pLOR-V or V111 p on day 14 (I-2). A sham-immunized group receiving saline at days 0, 14, and 28 served as control. FIGURE 3A-3F. Temporal immune responses to V. cholerae and ETEC OMVs. Shown are the median titers over time of IgM (A,D), IgG1 (B,E), and IgA (C,F) antibodies to OMVs derived from V111 pLOR-V (A–C) and E111 pLOR-E (D–F) in sera from mice intranasally immunized with OMVs derived from V111 pLOR-V (red solid line), V111 p (red dashed line), E111 pLOR-E (blue solid line), E111 p (blue dashed line), and from the sham-immunized control group (gray dotted line). Moreover, two groups were intranasally immunized with alternating regimen and received OMVs from E111 pLOR-E on days 0 and 28 as well as OMVs from V111 pLOR-V on day 14 (EVE pLOR-E/V, purple solid line) or OMVs from E111 p on days 0 and 28 as well as V111 p on day 14 (EVE p, purple dashed line). Samples which did not yield in any detectable signal at their highest concentration were set to the limit of detection (0.15 mg/ml). The error bars indicate the interquartile range of each data set for each time point (n = 7 for each group). FIGURE 4. Temporal immune responses against the SARS-CoV-2 Spike protein. Shown are the median titers over time of IgG1 antibodies to SARS-CoV-2 Spike protein in sera from mice intranasally immunized with OMVs derived from V111 pLOR-V (red solid line), V111 p (red dashed line), E111 pLOR-E (blue solid line), E111 p (blue dashed line), and from the sham-immunized control group (gray dotted line). Moreover, two groups were intranasally immunized with alternating regimen and received OMVs from E111 pLOR-E on days 0 and 28 as well as OMVs from V111 pLOR-V on day 14 (EVE pLOR-E/V, purple solid line) or OMVs from E111 p on days 0 and 28 as well as V111 p on day 14 (EVE p, purple dashed line). Samples which did not yield in any detectable signal at their highest concentration were set to the limit of detection (0.01 mg/ml). The error bars indicate the interquartile range of each data set for each time point (n = 7 for each group). FIGURE 5A-5F. Quantification of the total immune response against V. cholerae, ETEC and SARS-CoV-2 Spike protein induced in OMV-immunized mice. Shown are median half-maximum total Ig titers to OMVs derived from V111 pLOR-V (A,D) and E111 pLOR-E (B,E) as well as to SARS-CoV-2 Spike protein (C,F) in sera collected at day 42 (A–C) or day 60 (D–F) from mice of the respective immunization groups as indicated (n = 7 for each group). The error bars indicate the interquartile range of each data set and the hash key that the result of the data set was below the limit of detection, which was set to 1. Significant differences to the sham-immunized control group are marked by asterisks (p < 0.05; Kruskal–Wallis test and post hoc Dunn’s multiple comparisons). FIGURE 6A-6D. Mucosal immune responses induced in OMV-immunized mice. Shown are median IgA titers to OMVs derived from V111 pLOR-V (A,C) and E111 pLOR- E (B,D) extracted from fecal pellets collected at day 42 (A,B) or day 60 (C,D) from the respective immunization groups as indicated (n = 7 for each group). The error bars indicate the interquartile range of each data set and the hash key that the result of the data set was below the limit of detection (1 mg/g). Significant differences to the sham-immunized control group are marked by asterisks (p < 0.05; Kruskal–Wallis test and post hoc Dunn’s multiple comparisons). FIGURE 7. Neutralization assays with sera from OMV-immunized mice. Shown is the median fraction of infectivity using a Spike-pseudotyped lentivirus neutralization assay in combination with sera collected at day 60 from mice of the respective immunization groups as indicated (n = 7 for each group). Starting with a 1:4 dilution, 3.5-fold serial dilutions of each sera were tested. The error bars indicate the interquartile range of each data set. The limit of detection in these assays was a fraction of infectivity = 0.0001. Overlapping median values and error bars were slightly nudged on the x-axis to allow better visualization. Significant reductions of infectivity compared to the sham-immunized control group are marked by asterisks in the color of the respective data set and were observed for the following data sets: V111 pLOR-V and E111 pLOR-E for the 1:4 serum dilution as well as EVE pLOR-E/V for the 1:4, 1:14, and 1:49 dilution (p < 0.05; Kruskal–Wallis test and post hoc Dunn’s multiple comparisons). FIGURE 8. Genetically engineered detoxified triple mutants show increased vesiculation compared to the parental wild type strain. OMV quantification (Bradford) for V-WT, VΔΔΔ, E-WT and EΔΔΔ isolated from cultures grown for 8 h at 37°C with aeration. Shown is median ± IQR (n ≥ 4; *P < 0.05). FIGURE 9A-9B. DNA and amino acid sequences of the codon-optimized Lpp- OmpARBD (LOR) fusion constructs for V. cholerae (A)(SEQ ID NO: 1 (DNA) and SEQ ID NO: 2 (amino acid)) and E. coli (B) (SEQ ID NO: 3 (DNA) and SEQ ID NO:4 (amino acid)). Shown are the in silico designed, codon-optimized DNA sequences synthesized and subcloned into the standard vector system pMK by the GeneArt Gene Synthesis platform (ThermoFisher Scientific). The corresponding amino acid sequences (if applicable) are provided in 1-letter code centered below each nucleotide triplet. Original sequence information from V. cholerae N16961, ETEC H10407 or SARS-CoV-2 (isolate Wuhan Hu1) was retrieved from KEGG or NCBI databases (Kanehisa and Goto, 2000;Brister et al., 2015). Relevant features are highlighted as follows: underlined: binding of the oligonucleotides for amplification (LOR_E_1 and LOR_E_BamHI_2 as well as LOR_V_1 and LOR_V_BamHI_2); light grey and bold: restriction sites KpnI and BamHI for sublconing into pMMB67EH; dark grey: ShineDalgarno sequence; light blue: Lpp fragment; green: linker; red: OmpA fragment; yellow: SARS-CoV2 RBD fragment; pink: FLAG-tag FIGURE 10A-10B. Additional characterization of LOR decoration of OMVs. (A) Shown is a representative immunoblot detecting the LOR fusion protein in OMVs derived from VΔΔΔ pLORV and VΔΔΔ p, EΔΔΔ pLOR-E and EΔΔΔ p. The commercially available anti-FLAG antisera specifically detecting the C-terminal FLAG-tag of the LOR fusion protein was used for this immunoblot. (B) SDS-PAGE and Kang staining was executed in parallel with the same samples used for the immunoblot analyses provided in Fig. 1B and 10A and serves as a loading control. Molecular mass standards (PageRuler Prestained Protein Ladder – Thermo Fisher Scientific) are indicated on the left. FIGURE 11. Protein profiles of the immunoprecipitation (IP). SDS-PAGE and Kang staining was executed in parallel with the same samples used for the immunoblot analyses provided in Fig.1C and serves as a loading control. Molecular mass standards (PageRuler Prestained Protein Ladder – Thermo Fisher Scientific) are indicated on the left. FIGURE 12. Size distribution and biomass quantification of OMVs used in this study. Mean and mode OMV diameter sizes were measured by Zetasizer. Biomass quantification of OMV preparations derived from equivalent OD600 units of the respective cultures were analyzed for total protein (Bradford) or lipopolysaccharide (Purpald). Median with maximum and minimum values are shown (n ≥ 5 biological replicates). FIGURE 13. Controls for the neutralization assay. Shown is the median fraction of infectivity using a Spike-pseudotyped lentivirus neutralization assay in combination with commercially available anti-RBD antisera (PA5-114451, ThermoFisher Scientific). Starting with a 1:10 dilution, 2.5-fold serial dilutions of the anti-RBD antisera were tested in duplicate to proof functionality of the assay. Appropriate dilutions of PBS served as solvent (negative) control. DETAILED DESCRIPTION The present disclosure describes severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine compositions, as well as methods for making and using the same. In particular, the inventors disclose a novel SARS-CoV-2 vaccine candidate based on bacterial outer membrane vesicles (OMVs) as disclosed in U.S. Patent No.8,642,046 (the content of which is incorporated herein by reference in its entirety). SARS-CoV-2 includes membrane (M), spike (S), envelope (E), and nucleocapsid (N) structural proteins. The membrane, spike and envelope proteins provide the structure of the exterior viral envelope. The spike protein mediates binding to the human ACE2 receptor triggering cell entry. Spike is processed into two subunits, S1 and S2, which remain associated. The S1 subunit has a compact C-terminal domain that is largely comprised of the receptor binding domain (RBD). Which includes the receptor binding motif (RBM) directly binding the human ACE2 receptor. Antibodies which recognize epitopes with the RBM, or regions of the RBD outside of the RBM can effectively neutralize the virus. Outer membrane vesicles, which are naturally released by gram-negative bacteria during growth, are comprised largely of outer membrane, lipids, lipopolysaccharide (LPS), integral membrane proteins, and lipoproteins. OMVs possess characteristics that make them ideal candidates as a vaccine platform, including immunostimulatory pathogen-associated molecular patterns (PAMPs) and a size of 20–200 nm which allows them to drain freely into lymph nodes to target locally residing immune cells. Importantly for this disclosure, OMV production strains can also be genetically engineered to express and decorate their surface with antigens of interest. In some embodiment a composition is provided, the composition comprising- from N terminus to C-terminus- the N-terminal signal sequence of the E. coli Lpp protein fused in frame either directly or via a linker to a fragment of the E. coli OmpA protein, wherein the fragment of the E. coli OmpA protein comprises a N-terminal deletion and a C-terminal deletion, the fragment of the E. coli OmpA protein fused either directly or via linker to a fragment of the SARS CoV-2 Spike protein comprising the receptor binding domain (RBD). In some embodiments, the composition further comprises outer membrane vesicles (OMVs) and the fusion protein is present on the surface of the OMVs. The fusion protein may be a codon optimized polynucleotide and comprise SEQ ID NO: 1 or SEQ ID NO: 2. OMVs are nanosized proteoliposomes derived from the outer membrane of Gram- negative bacteria. OMVs are secreted via vesiculation of the outer membrane as part of the normal growth process. OMVs may be derived from any gram-negative bacteria, for example Salmonella, Neisseria, Bordetella or Yersinia enterocolitica. By way of example, and not limitation, the OMVs of the present disclosure may be derived from Vibrio cholerae (V. cholerae) or Escherichia coli (E. coli). The present disclosure employs the Lpp-OmpA fusion strategy for surface targeting. In the Lpp-OmpA vector, the Lpp domain serves to target and anchor in outer membrane and the OmpA domain is required for the surface expression. By way of example, but not limitation, the LOR fusion constructs of the present disclosure consist of a N-terminal signal sequence and next nine amino acids of the major outer membrane lipoprotein (Lpp), followed by a two-amino-acid linker, and then approximately 110 amino acids of OmpA, i.e., amino acids 46–146 of ETEC and amino acids 51–158 of V. cholerae OmpA. The Lpp fragment directs the fusion protein to the outer membrane, with the acylated N-terminal end being anchored in the inner leaflet of the outer membrane. The OmpA fragment contains five membrane-spanning segments with the C-terminus residing externally. Via an additional four-amino-acid linker the heterologous peptide consisting of the entire RBM and the majority of the RBD of SARS-CoV-2 is fused to the C-terminal end. Species-specific fusion constructs using the individual lpp and ompA sequences as well as codon optimized sequences of the SARS-CoV-2 RBD for each species may also be used. In the present disclosure V. cholerae or E. coli specific lpp and ompA are disclosed. Each segment of the fusion construct described herein may further include modifications in length or sequence, for example, additional amino acids in the linkers, to optimize function. In some embodiments, the composition is a fusion protein. Fusion proteins or chimeric proteins are proteins created through the joining of two or more genes that originally coded for separate proteins. Translation of this fusion gene results in a single or multiple polypeptides with functional properties derived from each of the original proteins. The fusion protein may comprise SEQ ID NO: 2 or SEQ ID NO: 4 or sequences 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical thereto, a fragment thereof, or combinations thereof. Regarding proteins, the amino acid sequences of variants, mutants, or derivatives as contemplated herein may include conservative amino acid substitutions relative to a reference amino acid sequence. For example, a variant, mutant, or derivative protein may include conservative amino acid substitutions relative to a reference molecule. “Conservative amino acid substitutions” are those substitutions that are a substitution of an amino acid for a different amino acid where the substitution is predicted to interfere least with the properties of the reference polypeptide. In other words, conservative amino acid substitutions substantially conserve the structure and the function of the reference polypeptide. The following table provides a list of exemplary conservative amino acid substitutions which are contemplated herein:
Figure imgf000012_0001
Conservative amino acid substitutions generally maintain (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a beta sheet or alpha helical conformation, (b) the charge or hydrophobicity of the molecule at the site of the substitution, and/or (c) the bulk of the side chain. Non-conservative amino acids typically disrupt (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a beta sheet or alpha helical conformation, (b) the charge or hydrophobicity of the molecule at the site of the substitution, and/or (c) the bulk of the side chain. Disclosed proteins, mutants, or variants, described herein may have one or more functional or biological activities exhibited by a reference polypeptide (e.g., one or more functional or biological activities exhibited by wild-type protein, or the unmodified fusion). In some embodiments, the activity of the variant or mutant protein may have an activity that is enhanced, as compared to a comparable wild-type or control protein, or may have an alternative or a modified activity as compared to a comparable or wild-type or control protein. In some embodiment, the fusion protein described herein may be expression by an expression vector. An expression vector is any particle used as a vehicle to artificially carry a foreign nucleic sequence, typically DNA into another cell, where it can be replicated and/or expressed. A vector containing foreign DNA is termed recombinant DNA. The four major types of vectors are plasmids, viral vectors, cosmids, and artificial chromosomes. Expression constructs comprise a heterologous promoter and the nucleic acid sequence encoding protein of interest (e.g., SEQ ID NO: 1 or SEQ ID NO: 3, sequences 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical thereto, or nucleic acid sequence encoding the amino acid sequence of SEQ ID NO: 2 and SEQ ID NO: 4) which is capable of expression in the cell in which it is introduced. The expression constructs include vectors which are capable of directing the expression of exogenous genes to which they are operatively linked. Such vectors are referred to herein as "recombinant constructs," "expression constructs," "recombinant expression vectors" (or simply, "expression vectors" or "vectors") and may be used interchangeably. Suitable vectors are known in the art and contain the necessary elements in order for the gene encoded within the vector to be expressed as a protein in the host cell. The term "vector'' refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated, specifically exogenous DNA segments encoding the mutant α-gal protein. Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. Viral vectors are incorporated into viral particles that are then used to transport the viral polynucleotide encoding the protein of interest into the target cells. Certain vectors are capable of autonomous replication in a host cell into which they are introduced. Other vectors can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome (e.g., lentiviral vectors). Moreover, certain vectors are capable of directing the expression of exogenous genes to which they are operatively linked. In general, vectors of utility in recombinant DNA techniques are often in the form of plasmids. Suitable vectors include, but are not limited to, plasmids, expression vectors, lentiviruses (lentiviral vectors), adeno- associated viral vectors (rAAV), among others and includes constructs that are able to express the protein of interest. A preferred vector is a lentiviral vector or adeno-associated vector. Suitable methods of making viral particles are known in the art to be able to transform cells in order to express the protein of interest described herein. In some embodiments the fusion protein is encoded by a polynucleotide. The term “polynucleotide” is used herein interchangeably with the term “nucleic acid” and refers to an organic polymer composed of two or more monomers including nucleotides, nucleosides or analogs thereof, including but not limited to single stranded or double stranded, sense or antisense deoxyribonucleic acid (DNA) of any length and, where appropriate, single stranded or double stranded, sense or antisense ribonucleic acid (RNA) of any length, including siRNA. The term “nucleotide” refers to any of several compounds that consist of a ribose or deoxyribose sugar joined to a purine or a pyrimidine base and to a phosphate group, and that are the basic structural units of nucleic acids. The term “nucleoside” refers to a compound (as guanosine or adenosine) that consists of a purine or pyrimidine base combined with deoxyribose or ribose and is found especially in nucleic acids. The term “nucleotide analog” or “nucleoside analog” refers, respectively, to a nucleotide or nucleoside in which one or more individual atoms have been replaced with a different atom or with a different functional group. Accordingly, the term polynucleotide includes nucleic acids of any length, including DNA, RNA, ORFs, analogs and fragments thereof. In some embodiments the polynucleotide is codon optimized. Codon optimization is a process to improve gene expression and increase translational efficiency by accommodating codon bias of the host organism. In some embodiments an expression vector is introduced into a cell for purposes of expressing the fusion protein of the expression vector. In particular embodiments the cell is V. cholerae or E. coli. The expression vector can be introduced into the cell by any means known in the art. These means may include transfection or transduction. Transfection is the process of introducing nucleic acids into cells by non-viral methods. Transduction is the process whereby foreign DNA is introduced into another cell via a viral vector. These are common tools to introduce a foreign gene into host cells. Among others, additional means include transformation, and conjugation. In some embodiments the OMVs may be administered to induce an immune response. As used herein, the term "administering" an agent, such as a therapeutic entity to an animal or cell, is intended to refer to dispensing, delivering or applying the substance to the intended target. In terms of the therapeutic agent, the term "administering" is intended to refer to contacting or dispensing, delivering or applying the therapeutic agent to a subject by any suitable route for delivery of the therapeutic agent to the desired location in the animal, including delivery by either the parenteral or oral route, intramuscular injection, subcutaneous/intradermal injection, intravenous injection, intrathecal administration, buccal administration, transdermal delivery, topical administration, and administration by the intranasal or respiratory tract route. In some embodiments, the composition may be administered intranasally. For intranasal administration or administration by inhalation or insufflation, the compound(s) can be conveniently delivered in the form of an aerosol spray from pressurized packs or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, fluorocarbons, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges for use in an inhaler or insufflator (for example capsules and cartridges comprised of gelatin) may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch The compositions described herein will generally be used in an amount effective to achieve the intended result, for example in an amount effective to treat or prevent the particular disease being treated. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated and/or eradication or amelioration of one or more of the symptoms associated with the underlying disorder such that the patient reports an improvement in feeling or condition, notwithstanding that the patient may still be afflicted with the underlying disorder. Therapeutic benefit also generally includes halting or slowing the progression of the disease, regardless of whether improvement is realized. The amount of composition administered will depend upon a variety of factors, including, for example, the particular indication being treated, the mode of administration, whether the desired benefit is prophylactic or therapeutic, the severity of the indication being treated and the age and weight of the patient, the bioavailability of the particular composition, the conversion rate and efficiency of delivery under the selected route of administration, etc. Determination of an effective dosage for a particular use and mode of administration is well within the capabilities of those skilled in the art. Effective dosages may be estimated initially from in vitro activity and metabolism assays. For example, an initial dosage for use in animals may be formulated to achieve a circulating blood or serum concentration of the composition that is at or above an IC50 of the particular composition as measured in an in vitro assay. Calculating dosages to achieve such circulating blood or serum concentrations taking into account the bioavailability of the particular composition via the desired route of administration is well within the capabilities of skilled artisans. Initial dosages can also be estimated from in vivo data, such as animal models. Animal models useful for testing the efficacy of the active metabolites to treat or prevent the various diseases described above are well-known in the art. Animal models suitable for testing the bioavailability and/or metabolism of compositions are also well-known. Ordinarily skilled artisans can routinely adapt such information to determine dosages suitable for human administration. In some embodiment, an immune response is induced. An immune response involves many cells and proteins of the immune system and is a reaction which occurs within an organism for the purpose of defending against substances it sees as harmful or foreign. An immune response may comprise the activation of T or B cells, phagocytes or natural killer cells, and may include the production of cytokines or antibodies. In particular embodiments, the immune response comprises an antibody response against SARS CoV- 2 spike protein as part of a humoral immune response. Antibody responses are generated by B lymphocytes and plasma cells and result in the appearance of immunoglobulin classes each of which bears identical antibody-combining sites. In some aspects the antibody response is a neutralizing antibody response. A neutralizing antibody is an antibody that defends a cell from a pathogen or infectious particle by neutralizing any effect it has biologically. Neutralization renders the particle no longer infectious or pathogenic. Unless otherwise specified or indicated by context, the terms “a”, “an”, and “the” mean “one or more.” For example, “a molecule” should be interpreted to mean “one or more molecules.” As used herein, “about”, “approximately,” “substantially,” and “significantly” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which they are used. If there are uses of the term which are not clear to persons of ordinary skill in the art given the context in which it is used, “about” and “approximately” will mean plus or minus ≤10% of the particular term and “substantially” and “significantly” will mean plus or minus >10% of the particular term. As used herein, the terms “include” and “including” have the same meaning as the terms “comprise” and “comprising.” The terms “comprise” and “comprising” should be interpreted as being “open” transitional terms that permit the inclusion of additional components further to those components recited in the claims. The terms “consist” and “consisting of” should be interpreted as being “closed” transitional terms that do not permit the inclusion additional components other than the components recited in the claims. The term “consisting essentially of” should be interpreted to be partially closed and allowing the inclusion only of additional components that do not fundamentally alter the nature of the claimed subject matter. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention. All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein. Preferred aspects of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred aspects may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect a person having ordinary skill in the art to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context. EXAMPLES The following Examples are illustrative and should not be interpreted to limit the scope of the claimed subject matter. Example 1 - An intranasal vaccine based on outer membrane vesicles against SARS- CoV-2 Reference is made to the manuscript: Thapa et al., "An intranasal vaccine based on outer membrane vesicles against SARS-CoV-2," Front. Microbiol.2021 Nov.5;12:752739, the content of which is incorporated by reference in its entirety. Vaccines were invented over 200 years ago, with the recombinant vaccine era beginning in 1981 (Plotkin, 2014). Despite this long experience, generation of effective vaccines against emerging pathogens remains exceedingly difficult and still retains a significant empirical component. The current pandemic of SARS-CoV-2 highlights the desperate need of versatile vaccine-platforms, which can be easily modified to carry antigens of emerging pathogens. Within a remarkable time-frame and substantial investment we now have a handful of available vaccines that elicit significant protection against COVID-19, and ongoing vaccination efforts are successful in many countries (Abdulla et al., 2021; Pormohammad et al., 2021). However, the vaccines still face limitations as they are currently relying on invasive immunization routes (intramuscular), depend on highly specialized custom-built production pipelines, and require a cold-chain. Hence, mass production and worldwide distribution will be a huge challenge. Thus, the current and future pandemic outbreaks still warrant that we take a broad approach in developing safe and effective vaccine candidates. Herein, we present an outer membrane vesicle (OMV)-based vaccine candidate that is decorated with the receptor binding domain (RBD) of the SARS- CoV-2 Spike protein. The Spike protein is present in multiple copies as protruding homotrimeric spikes on the viral envelope and mediates binding to the human ACE2 receptor triggering cell entry (Bosch et al., 2003). Spike is processed into two subunits, S1 and S2, which remain associated. The S1 subunit has a compact C-terminal domain that is largely composed of the RBD, which includes the receptor binding motif (RBM) directly binding the human ACE2 receptor. Importantly, antibodies recognizing epitopes within the RBM, or regions of the RBD outside of the RBM, can effectively neutralize the virus (Cao et al., 2020; Grifoni et al., 2020; Yi et al., 2020). For these reasons, current vaccine designs are mainly focused on the RBD of the SARS-CoV-2 Spike protein. Outer membrane vesicles, which are naturally released by gram-negative bacteria during growth, are comprised largely of outer membrane, lipids, lipopolysaccharide (LPS), integral membrane proteins, and lipoproteins. OMVs possess characteristics that make them ideal candidates as a vaccine platform (Kulp and Kuehn, 2010; van der Pol et al., 2015; Pathirana and Kaparakis-Liaskos, 2016). First, they have intrinsic immunostimulatory properties due to the presence of pathogen-associated molecular patterns (PAMPs), primarily lipid A, porins, and lipoproteins. Together these PAMPs stimulate multiple pathogen recognition receptors (PRRs) in and on host cells, which in turn stimulate innate and adaptive immune responses (Ellis and Kuehn, 2010). Second, the OMV diameter of 20– 200 nm allows them to drain freely into lymph nodes to target locally residing immune cells (van der Pol et al., 2015; Pathirana and Kaparakis-Liaskos, 2016). Thus, using the intranasal route of immunization will likely elicit a mucosal immune response in the airways to OMV antigens. Third, OMV production strains can be genetically engineered to express and decorate their surface with antigens of interest (Gerritzen et al., 2017). Among others, we have been studying OMVs as vaccine candidates against human mucosal pathogens of the intestinal and respiratory tracts, e.g., members of the Pasteurellaceae, pathogenic Escherichia coli, and Vibrio cholerae (Schild et al., 2008, 2009; Bishop et al., 2010; Roier et al., 2012, 2013; Leitner et al., 2013, 2015). Overall, our studies show that non-invasive intranasal immunization induces a specific, high-titer, protective antibody response in the murine model that is long-lasting. Genetic engineering of donor strains allowed a deeper characterization of OMVs derived from V. cholerae and enterotoxigenic Escherichia coli (ETEC). For example, genetic modification of lipid A resulted in less endotoxicity without diminishing the immunogenic potential (Leitner et al., 2013, 2015). Furthermore, both bacterial species have been successfully genetically engineered to produce OMVs loaded with antigens of interest (Leitner et al., 2015; Gnopo et al., 2017). Herein, we have genetically engineered detoxified ETEC and V. cholerae strains with increased OMV production. Using a Lpp-OmpA’ fusion strategy, previously used to express proteins of interest on the surface of E. coli K-12 bacteria (Francisco et al., 1992; Stathopoulos et al., 1996; Daugherty et al., 1998; Earhart, 2000), OMVs released by V. cholerae and ETEC could be efficiently decorated with the C-terminal part of the SARS- CoV-2 Spike protein S1 containing the RBD. Mice immunized with OMVs decorated with Lpp-OmpA-RBD (LOR) fusion protein induced a robust immune response not only against the bacterial surface components, but also against the Spike protein. SARS-CoV-2 neutralizing antibodies were confirmed in cell culture infection assays using the lentiviral SARS-CoV-2 pseudovirus in combination with 293T cells engineered to express the SARS- CoV-2 receptor ACE2. Materials and Methods Bacterial Strains, Cell Lines and Growth Conditions Bacterial strains, cell lines and plasmids used in this study are listed in Table 1; oligonucleotides are listed in Table 2. AC53, a spontaneous streptomycin (Sm)-resistant mutant of the clinical isolate E7946 (O1 El Tor Ogawa), or ETEC H10407-S, a Sm-resistant mutant of the clinical isolate H10407, were used as wild-type strains (V-WT and E-WT). E. coli strain DH5αλpir and SM10λpir were used for genetic manipulations. Unless stated otherwise, strains were cultivated in Lysogeny broth (LB) or on LB agar plates with aeration at 37°C. If required, antibiotics and other supplements were used in the following final concentrations: streptomycin (Sm), 100 μg/ml; ampicillin (Ap), 100 μg/ml or in combination with other antibiotics 50 μg/ml; kanamycin (Km), 50 μg/ml; IPTG, 0.1 mM; glucose (Gluc), 0.2%; and sucrose (Suc), 10%. Table 1. Bacterial strains, cell lines and plasmids used in this study
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Table 2. Oligonucleotides used in this study
Figure imgf000024_0002
Restriction sites are underlined. Construction of Deletion Mutants and Expression Plasmids The isolation of chromosomal DNA, PCR reactions, the purification of plasmids or PCR products, the construction of suicide and expression plasmids as well as the subsequent generation of deletion mutants were carried out as described previously using derivatives of pCVD442 or pMMB67EH (Seper et al., 2011; Leitner et al., 2015; Pressler et al., 2016). Qiagen plasmid kits were used for isolation of plasmid DNA; QIAquick® Gel extraction and QIAquick® PCR Purification kits (Qiagen) were used for purifying DNA fragments. PCR reactions for subcloning were carried out using the Q5® High-Fidelity DNA Polymerase (NEB), while Taq DNA Polymerase (NEB) was used for all other PCRs. Construction of ompA in-frame deletion mutants in V. cholerae and ETEC were carried out as described by Donnenberg and Kaper (1991) using derivatives of pCVD442, i.e., pΔompA-V or pΔompA-E. The suicide vector pΔompA-V was already available from a previous study (Song et al., 2008). For construction of pΔompA-E, ∼800 bp PCR fragments located up- and downstream of the ompA were amplified using the oligonucleotide pairs ompA_E_SacI_1 and ompA_E_EcoRI_2 as well as ompA_E_EcoRI_3 and ompA_E_XbaI_4 with chromosomal DNA from E-WT as template (Table 2). After digestion of the PCR fragments with the appropriate restriction enzyme (NEB) indicated by the name of the oligonucleotide, they were ligated into pCVD442, which was digested with the appropriate restriction enzymes. Unless noted otherwise, ligation products were transformed into DH5αλpir and ApR colonies were characterized for the correct constructs by PCR. To obtain deletion strains, generated derivatives of pCVD442 were transformed into E. coli Sm10λpir and conjugated into V. cholerae or ETEC. Exconjugants were purified by SmR/ApR selection. Sucrose selection was used to obtain ApS colonies and chromosomal deletions were confirmed by PCR, respectively. The Lpp-OmpA-RBDVch (LORVch) and Lpp-OmpA-RBDETEC (LORETEC) fusion proteins were designed in silico and the corresponding sequences including a 5′ untranslated region harboring a unique KpnI restriction site and an optimal Shine-Dalgarno sequence (Fig.9) were synthesized and subcloned into the standard vector system pMK by the GeneArt Gene Synthesis platform (Thermo Fisher Scientific). Thus, constructs were provided as pMK-V-LOR and pMK-E-LOR. Finally, the expression plasmid pLOR-V and pLOR-E were constructed using the oligonucleotides LOR_V_1 and LOR_V_BamHI_2 as well as LOR_E_1 and LOR_E_BamHI_2 for amplifying the fusion construct using the ordered plasmids as template. This added a C-terminal FLAG-tag epitope to the constructs. The resulting PCR fragments were purified, digested with KpnI and BamHI, and ligated into pMMB67EH, which has been digested with the same enzymes. Ligation products were transformed into DH5αλpir and ApR colonies were characterized for the correct constructs by PCR. Preparation of Outer Membrane Vesicles Vibrio cholerae as well as ETEC OMVs were isolated as previously described (Schild et al., 2008; Leitner et al., 2013, 2015). Briefly, overnight cultures grown in LB medium of the respective strains were diluted (1:100) in fresh LB and cultivated overnight at 24°C and 180 rpm. The cells were then removed from the supernatant by centrifugation (9,500 × g, 20 min, 4°C) and subsequent sterile filtration (0.22 μm). The OMVs present in the supernatant were pelleted through subsequent ultracentrifugation (150,000 × g, 4°C, 4 h) and resuspended in saline to generate a 1000-fold concentrated OMV suspension compared to the original filter-sterilized supernatant. Protein concentration was determined using Bradford assay (Bio-Rad Laboratories, Protein Assay Dye Reagent) according to the manufacturer’s manual. To quantify the LPS content of OMVs, purpald assays were performed as described previously using 3-deoxy-D-mannooctulosonic acid (Kdo) (Sigma- Aldrich) as a standard (Roier et al., 2016). Size Measurement To estimate the size distributions of the isolated OMVs, dynamic light scattering (DLS) was carried out using Zetasizer Nano ZS90 (Malvern, United Kingdom) as previously described (Zingl et al., 2021b). Samples were diluted 1:1000 in saline and processed at 25°C under standard settings [Dispersant Refractive Index = 1.331, viscosity (cP) = 0.89]. Three measurements were performed using a measurement angle of 173° (backscatter), auto measurement duration, and “seek for optimal position” as positioning setting. Animals Female BALB/c mice (Charles River Laboratories) were used in all experiments in accordance with the rules of the ethics committee at the University of Graz and the corresponding animal protocol, which has been approved by the federal ministry BMBWF (protocol: 39/12/75ex2017/18). Mice were housed with food and water ad libitum and monitored under the care of full-time staff. All animals were acclimated for 1 week before any procedures were carried out and were approximately 8 weeks old at the start of the immunization. Outer Membrane Vesicle Immunization Protocol Eight-week-old female mice were intranasally immunized with OMVs (25 μg) in saline at days 0, 14, and 28 as described previously (Schild et al., 2008; Leitner et al., 2013). A group of sham (saline)-immunized control mice were housed in parallel with the immunized mice for the duration of each experiment. Overall two independent immunization rounds for each immunization group were performed with at least three mice per group. Comparison of the results from the independent immunization rounds revealed no differences in the induction of a humoral immune response in the respective immunization group. Preparation of Blood and Stool Samples Blood samples as well as fecal pellets were collected from immunized and sham- immunized adult mice throughout the immunization study and processed as previously described to monitor the induced immune response (Schild et al., 2008, 2009). Briefly, blood samples were collected from immunized and sham immunized mice on days 0, 14, 28, 42, and 60. The collected blood was allowed to clot at room temperature (RT) for 30 min after which serum was isolated by removing the blood clot using a sterile toothpick followed by centrifugation (15 min, 1,000 × g). The supernatant representing the serum of each sample was removed, diluted threefold in PBS/sodium azide (0.05%), and subsequently stored at −80°C. In addition, aliquots of the sera collected on day 60 were stored at −80°C without any dilution or addition of sodium azide of to be used in cell culture neutralization assays. In case of stool samples, freshly voided fecal pellets collected on days 42 and 60 were vacuum-dried for 10 min before their weight was recorded. Immunoglobulins were extracted by adding 1 ml of extraction buffer [PBS, 0.01% sodium azide, 5% fetal calf serum, 1 tablet complete EDTA-free protease inhibitor cocktail (Roche) per ml] per 100 mg dry- weight feces. After vortexing the samples for 15 min at 4°C, solid material was separated by centrifugation (2 min, 13,000 × g) and the supernatants were stored at −80°C. Immunoprecipitation Using Outer Membrane Vesicles and Anti-receptor Binding Domain Antisera For immunoprecipitation 9 μl of SARS-CoV-2 Spike Protein (RBD) Antibody (PA5-114451, Thermo Fisher Scientific) were mixed with 100 μg OMVs (protein equivalent determined by Bradford) and adjusted to final volume of 200 μl using saline. Antibodies were allowed to bind to the OMVs for 30 min at RT under gentle rocking before the OMVs were pelleted by centrifugation (75,600 × g, 4 h, 4°C). The OMV pellet was washed once with 1 ml saline and centrifuged again (75,600 × g, 2 h, 4°C) before the sample was finally resuspended in 40 μl saline and subjected to immunoblot analyses. Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis and Immunoblot Analysis To analyze the protein content of OMVs, the standard sodium dodecyl sulfate- polyacrylamide gel electrophoresis (SDS-PAGE) procedure in combination with 12% gels and the PageRuler Prestained Protein Ladder (Thermo Fisher Scientific) as a molecular mass standard were used. Approximately 7.5 μg of each sample was loaded and either stained according to the procedure of Kang et al. (2002) or transferred to a nitrocellulose membrane (Amersham) for immunoblot analysis, which was essentially performed as described previously (Roier et al., 2012). Either the anti-FLAG® M2-HRP monoclonal antibody (A8592, Sigma Aldrich) was used as sole antibody or SARS-CoV-2 Spike Protein (RBD) Antibody (PA5-114451, Thermo Fisher Scientific) was used in combination with the HRP- linked anti-rabbit IgG (111-035-003, Jackson ImmunoResearch) were used as primary and secondary antibody, respectively. Enzyme-Linked Immunosorbent Assay Temporal immune responses of different Igs, half-maximum total Ig titers and mucosal immune responses to VΔΔΔ or EΔΔΔ OMVs (5 μg/ml in PBS, pH 7.4) as well as the determination of the half-maximum total Ig titers to SARS-CoV-2 Spike Protein S1 (5 μg/ml in PBS, pH 7.4, RP-87679 Thermo Fisher Scientific) were carried out essentially as described previously (Roier et al., 2012; Leitner et al., 2013, 2015) using appropriate purified mouse Ig isotype standard (IgA, 553476; IgG1, 557273; or IgM, 550963, BD Biosciences) as well as horseradish peroxidase-conjugated goat anti-mouse antibodies (IgA, 62-6720; IgG1, A10551; IgM, 62-6820; IgG, IgM, IgA, PA1-84388, Invitrogen) in combination with the TMB peroxidase substrate reagent set (BioLegend). Optical densities were monitored at 450 nm with a FLUOstar Omega microplate reader (BMG Labtech). Starting dilutions of the mouse sera were 1:10 for quantification of the temporal immune responses and half-maximum total Ig titers against the Spike protein, 1:100 for quantification of the half-maximum total Ig titers against OMVs, or 1:400 for quantification of the temporal immune responses against OMVs. Starting dilutions of the fecal pellet extracts for quantification of the mucosal immune responses against OMVs were 1:10. If the highest sample concentration (sera or fecal pellet extracts) used did not yield any detectable absorption the value was set to the respective limit of detection of the assay stated in each figure legend. Spike Pseudotyped Lentivirus Particles Plasmids pHAGE2-CMV-luciferase-IRES-ZsGreen, pHDM-Spike, pHDM-Hgpm2, pHDM-tat1b, and pRC-CMV-rev1b were transfected into HEK 293T cells as described (Crawford et al., 2020). The plasmids and HEK 239T-ACE2 cell line were obtained from BEI Resources (Manassas, VA, United States). Transfection was confirmed by scoring green fluorescence of the cells 24 h later. Viral particles were collected from cell culture supernatants after 48 h, as described (Crawford et al., 2020) and were stored in single-use aliquots at −80°C. The titer of thawed virus stock was 1.5 × 105 infectious units/ml. Serum Neutralization of Spike-Pseudotyped Lentivirus Neutralization assays were performed essentially as recently described (Crawford et al., 2020). Briefly, serial dilutions of each mouse serum were made in duplicate in 96-well plates (12565501, Fisher Scientific) in Dulbecco’s Modified Eagle Medium (DMEM) (MT10013CV, Corning) supplemented with 10% heat-inactivated fetal bovine serum (16140063, Gibco), and penicillin (100 U/ml) and streptomycin (100 μg/ml) (15140148, Gibco). A volume of 150 μl DMEM containing 3.1 × 103 infectious units of pseudotype virus was added to each well and incubated for 1.2 h at 37°C in a 5% CO2 incubator to allow antibodies to bind. This amount of virus gave a multiplicity of infection of approximately 0.08. Polybrene (TR1003E, Sigma) was added to each well to increase the efficiency of viral infection just before transferring the contents of each well to semi- confluent monolayers of HEK 293T-ACE2 cells in solid white 96-well plates (07-200-591, Fisher Scientific). The plates were incubated for 48 h at 37°C in a 5% CO2 incubator to allow for viral infection. Infection by the Spike-pseudotyped lentivirus expressing Luciferase-IRES-ZsGreen was quantified by measuring light production after removing spent medium and adding Bright-Glo Luciferase reagent (E2610, Promega). After mixing for 20 s by swirling, each plate was incubated in the dark for 2 min, followed by measuring relative light units (RLU) on a SynergyTM HT Microplate Reader using Gen5 data analysis software (BioTek). Background was initially assayed from uninfected cells, virus in the absence of cells, or DMEM medium alone, and all were equivalent. Therefore, in subsequent experiments, each plate contained at least two DMEM alone wells. The maximum infection on each plate was measured by infecting at least four wells of HEK 293T-ACE2 cells with pseudotype virus that had not been neutralized with serum. The fraction infectivity was calculated by averaging technical replicates and dividing by the average of the maximum infectivity on that plate. The inhibitory concentration 50% (IC50) was calculated graphically. Data Presentation and Statistical Analysis The data is generally presented as median with interquartile range as some data sets were not normally distributed. Consequently, data were analyzed using the Mann–Whitney U test or a Kruskal–Wallis test followed by post hoc Dunn’s multiple comparisons. Differences were considered significant at p-values of < 0.05. GraphPad Prism version 8 was used for all statistical analyses. Results Generation of V. cholerae and Enterotoxigenic Escherichia coli Outer Membrane Vesicles Decorated With the SARS-CoV-2 Receptor Binding Domain Antigen We recently demonstrated that OMVs derived from ETEC or V. cholerae lacking one functional secondary lipid A acyltransferase, MsbB (also referred to as LpxN) retain their potential to induce a high-titer, protective immune response but show a significantly reduced pro-inflammatory cytokine induction suggesting lower endotoxicity (Leitner et al., 2013, 2015). Moreover, cholera toxin subunits A and B in V. cholerae, and the heat-labile enterotoxin catalytic A subunit in E. coli were deleted. Thus, presence of these secreted toxins in OMVs is precluded (Leitner et al., 2015). In addition to these modifications resulting in detoxified OMVs, we also deleted ompA, encoding the outer membrane protein OmpA, which crosslinks the periplasmic peptidoglycan and the outer membrane. Deletion of ompA has been reported to result in elevated OMV release in various gram-negative bacteria (Song et al., 2008; Schwechheimer and Kuehn, 2015). Concordantly, the engineered triple mutants VΔΔΔ and EΔΔΔ showed a significant three- to six-fold increase in OMV production compared to the parental WT strains (V-WT and E-WT), respectively (Figure 8). Thus, the engineered triple mutants VΔΔΔ and EΔΔΔ are optimized V. cholerae and ETEC platform strains for production of OMVs to be used for vaccination. To ensure efficient translocation to the outer membrane and external exposure of the SARS-CoV-2 RBD domain, we adapted the Lpp-OmpA’ fusion strategy, which has been used previously to display a variety of heterologous proteins on the bacterial surface of E. coli (Francisco et al., 1992; Stathopoulos et al., 1996; Daugherty et al., 1998; Earhart, 2000). The general design of the LOR fusion constructs was similar for V. cholerae and ETEC (Figure 1A). The LOR fusion constructs consist of a N-terminal signal sequence and next nine amino acids of the major outer membrane lipoprotein (Lpp), followed by a two-amino- acid linker, and then approximately 110 amino acids of OmpA, i.e., amino acids 46–146 of ETEC and amino acids 51–158 of V. cholerae OmpA. The Lpp fragment directs the fusion protein to the outer membrane, with the acylated N-terminal end being anchored in the inner leaflet of the outer membrane. The OmpA fragment contains five membrane-spanning segments with the C-terminus residing externally (Francisco et al., 1992). Via an additional four-amino-acid linker the heterologous peptide consisting of the entire RBM and the majority of the RBD of SARS-CoV-2 is fused to the C-terminal end. As Lpp and OmpA of V. cholerae and ETEC share only 38% identity, the amino acid sequence as well as the SARS-CoV-2 Spike protein is neither optimal for E. coli nor V. cholerae; we have used species-specific fusion constructs using the individual lpp and ompA sequences as well as codon optimized sequences (obtained via the IDT codon optimization tool) of the SARS- CoV-2 RBD for each species, namely, LOR-V and LOR-E, respectively (Figure 9). Along the design, a unique KpnI restriction site for subsequent cloning into expression vectors and an optimized Shine Dalgarno sequence ensuring high translation efficiency were included in the 5′ untranslated region. Synthesized fragments were PCR-amplified, which added a C- terminal FLAG-tag epitope to the fusion proteins, and subcloned into the IPTG-inducible broad range vector pMMB67EH (see Figure 9 and “Materials and Methods” section for details) resulting in the LOR fusion expression constructs pLOR-V and pLOR-E. Expression of the LOR fusion proteins in VΔΔΔ and EΔΔΔ should result in their localization on the bacterial cell surface and therefore also on the surface of OMVs, which bud from the outer membrane. Successful localization of LOR in OMVs was confirmed by immunoblot analyses (Figure 1B and Figure 10). Detection of LOR fusion proteins with anti-FLAG and anti-RBD antibodies showed specific bands with the expected molecular weight of approximately 36 kDa for V. cholerae and 38 kDa for ETEC for the OMVs derived from VΔΔΔ pLOR-V and EΔΔΔ pLOR-E, but not for OMVs isolated from strains harboring the empty vector (VΔΔΔ p and EΔΔΔ p). Thus, OMVs of both species can be decorated with significant amounts of the LOR. Furthermore, outward exposure of RBD part on OMVs was confirmed via an immunoprecipitation assay using OMVs derived from VΔΔΔ pLOR-V, VΔΔΔ p, EΔΔΔ pLOR-E, and EΔΔΔ p as antigen in combination with anti-RBD antibodies. After incubation of OMVs and anti-RBD antibodies, samples were subjected to centrifugation and washing steps to purify OMVs. Finally, immunoblot analyses with the immunoprecipitated samples were performed to detect anti-RBD antibody (raised in rabbits) bound to the OMV samples using an HRP-conjugated anti-rabbit antisera (Figure 1C). A Kang-stained gel executed in parallel served as a loading control (Figure 11). Heavy chains of rabbit IgG, indicating bound anti-RBD antibodies to OMVs, were detected at the expected molecular weight of approximately 55 kDa in immunoprecipitations using OMVs derived from VΔΔΔ pLOR-V and EΔΔΔ pLOR-E, but not for OMVs isolated from strains harboring the empty vector (VΔΔΔ p and EΔΔΔ p). The accessibility of the LOR fusion proteins on OMVs by the anti-RBD antibody strongly suggests that the RBD portion is residing externally on the surface of the OMVs. Finally, we determined the size distribution and quantified the biomass of OMVs liberated by the parental WT strains (V-WT and E-WT), the triple mutants (VΔΔΔ and EΔΔΔ) as well as the donor strains for the OMV-vaccine candidates used herein (i.e., VΔΔΔ p, EΔΔΔ p, VΔΔΔ pLOR-V, and EΔΔΔ pLOR-E) (Figure 12). These analyses confirmed the increased OMV biomass amounts liberated by the triple mutants in both species. Intranasal Immunization of Lpp-OmpA-Receptor Binding Domain-Outer Membrane Vesicles Decorated With SARS-CoV-2 Receptor Binding Domain Induces a Robust Immune Response Against the Spike Protein In order to investigate the immune response upon immunization with LOR- decorated OMVs (LOR-OMVs), we intranasally immunized mice with OMVs derived from VΔΔΔ pLOR-V, VΔΔΔ p, EΔΔΔ pLOR-E, and EΔΔΔ p according to our standard immunization protocol (Schild et al., 2008; Figure 2A). Briefly, mice were intranasally immunized on days 0, 14, and 28 receiving 25 μg OMVs (protein equivalent) per immunization. In total, seven different immunization groups were tested (Figure 2B). Four groups received the same OMVs at days 0, 14, and 28 and were therefore only immunized with OMVs from either VΔΔΔ pLOR-V, VΔΔΔ p, EΔΔΔ pLOR-E, or EΔΔΔ p. Two groups (EVE pLOR-E/V and EVE p) were immunized with alternating regimen and received OMVs from EΔΔΔ pLOR-E on days 0 and 28 as well as OMVs from VΔΔΔ pLOR-V on day 14 (EVE pLOR-E/V) or EΔΔΔ p on days 0 and 28 as well as VΔΔΔ p on day 14 (EVE p). A change of the OMV donor species between the immunizations was thought to specifically boost the immune response against the RBD of SARS-CoV-2 being the common antigen on OMVs derived from VΔΔΔ pLOR-V and EΔΔΔ pLOR-E. Finally, a sham-immunized group receiving saline at days 0, 14, and 28 served as control. To monitor the immune responses in sera of immunized and sham-immunized control mice by enzyme-linked immunosorbent assay (ELISA), OMVs derived from VΔΔΔ pLOR-V or EΔΔΔ pLOR-E were used as coating material allowing the detection of temporal IgM, IgG1, and IgA responses to the respective OMVs (Figure 3). Herein we focused on IgG1 as previous studies revealed IgG1 to be the dominant IgG subclass induced after intranasal immunization with OMVs (Schild et al., 2008, 2009; Bishop et al., 2010; Leitner et al., 2013, 2015). As reported earlier, immunization with V. cholerae or ETEC OMVs induces a species-specific immune response without any significant cross-reactivity (Leitner et al., 2015). Thus, groups immunized with OMVs solely derived from one species were only tested for immune responses against OMVs derived from the same species, while immunization groups with alternating regimen and the sham-immunized control group were tested for immune responses against both, OMVs derived from V. cholerae and ETEC. The antibody titers of the sham-immunized control group were determined for days 0 and 60 and remained below the limit of detection or at very low levels for both time points against OMVs derived from V. cholerae and ETEC, respectively. In general, the induction of an immune response was characterized by an Ig titer increase during the vaccination period. Thus, IgM titers peaked at days 28 or 42, while IgG and IgA were slightly delayed and peaked on days 42 or 60. Not surprisingly, Ig titers of the immunization groups receiving the same OMVs at all three immunization were overall higher and reached the Ig peak earlier than the immunization groups with alternating regimen. Importantly, IgG1 and IgA titers remained stable or even increased between days 42 and 60, which indicates the induction of a robust immune response until day 60, the designated endpoint of this study. Next, we evaluated the induction of a humoral immune response against the SARS- CoV-2 Spike protein. Based on the high IgG1 titers in serum samples we focused on this isotype to analyze the temporal immune response against the Spike protein (Figure 4). IgG1 titers of the sham-immunized control group as well as the immunization groups VΔΔΔ p, EΔΔΔ p, and EVE p remained below the limit of detection or at very low levels at all time points, respectively. In contrast, all three immunization groups receiving OMVs decorated with LOR, i.e., VΔΔΔ pLOR-V, EΔΔΔ pLOR-E, and EVE pLOR-E/V showed detectable IgG1 titers against the Spike protein from days 28 onward, which peaked on days 42 or 60. We also tried to determine serum IgA responses against the Spike protein, but IgA titers were overall lower compared to the IgG1 titers and remained below the limit of detection (0.04 μg/μl) even for several serum samples of the immunization groups receiving OMVs decorated with LOR. Unfortunately, this analysis was hampered by observable cross- reactivity of the secondary antibody with serum components or the Spike protein combined with relatively low IgA titers in the samples. To further characterize the induced humoral immune response, sera from days 42 and 60 were used to determine the half-maximum total Ig titers against OMVs derived from VΔΔΔ pLOR-V or EΔΔΔ pLOR-E as well as against SARS-CoV-2 Spike protein (Figure 5). In comparison to the sham-immunized control group the immunization groups receiving OMVs showed a marked increase in total Ig titers for both time points against the respective OMVs derived from VΔΔΔ pLOR-V or EΔΔΔ pLOR-E (Figures 5A,B,D,E). Similar to the temporal immune response patterns, the OMV-specific responses were slightly lower in the immunization groups with alternating regimen (EVE pLOR-E/V and EVE p) compared to the immunization groups receiving the same OMVs at all three immunizations. In comparison to the sham-immunized control group, a significant increase of the total Ig titers against the Spike protein was observed at day 42 for the VΔΔΔ pLOR-V and EVE pLOR- E/V immunization group as well as for all immunization groups receiving OMVs decorated with LOR (i.e., VΔΔΔ pLOR-V, EΔΔΔ pLOR-E and EVE pLOR-E/V) at day 60 (Figures 5C,F). In contrast, to the OMV-specific immune response, the EVE pLOR-E/V immunization group showed slightly, but not significantly higher Ig titers against the Spike protein than the VΔΔΔ pLOR-V and EΔΔΔ pLOR-E groups. For both time points tested, the immune response against the Spike protein remained undetectable or at low levels for immunization groups receiving OMVs from strains harboring the empty vector (VΔΔΔ p, EΔΔΔ p and EVE p). In general, isotype-specific and total Ig titers did not massively drop between days 42 and 60 in the respective immunization groups suggesting the induction of a stable immune response (Figures 3–5). In addition to the humoral immune responses, we also analyzed the induced mucosal immune responses by measuring the secretory IgA titers in fecal pellet extracts collected at days 42 and 60 against OMVs derived from VΔΔΔ pLOR-V or EΔΔΔ pLOR-E (Figures 6A–D). Fecal pellet collection was preferred over other methods (e.g., saliva collection or lung lavage) as it seems less stressful and allowed us to keep the mice alive for later time points. Furthermore, it has been shown in earlier studies that secretory IgA antibodies reflecting the mucosal immune response can also be found in feces upon OMV immunization and that IgA titers in feces correlate by trend with those found in the respiratory tract, e.g., saliva (Vetvik et al., 1998; Hirano et al., 2006; Schild et al., 2009). Generally, the mucosal immune response on days 42 and 60 showed a similar pattern as the half-maximum total Ig titers. All immunization groups receiving OMVs showed a significant increase in secretory IgA titers against the respective OMVs compared to the sham-immunized control group (Figures 6A–D). In majority of the cases, the secretory IgA titers against OMVs were slightly lower in immunization groups with alternating regimen (EVE pLOR-E/V and EVE p) compared to the immunization groups receiving the same OMVs at all three immunizations. We also tried to determine secretory IgA levels against the Spike protein in fecal pellet extracts, but for the majority of samples the titers remained below or close to the limit of detection (1 μg/g). As described above, this analysis was hampered by observable cross-reactivity of the secondary antibody with components of the fecal pellet extract or the Spike protein combined with relatively low concentration of mucosal IgA in the samples. Immunization With V. cholerae and Enterotoxigenic Escherichia coli Lpp-OmpA- Receptor Binding Domain-Outer Membrane Vesicles Elicits Neutralizing Antibody Titers To determine if the vaccines were capable of eliciting neutralizing antibodies, a Spike-pseudotyped lentivirus neutralization assay was used as previously described (Crawford et al., 2020). Functionality of the assay was confirmed by using commercially available anti-RBD antisera as positive control and PBS as solvent (negative) control, which revealed a robust neutralizing activity of the anti-RBD antisera, but not for the solvent control (Figure 13). Briefly, a pseudotype lentivirus expressing the SARS-CoV-2 Spike protein on its envelope and, upon infection, luciferase in the host cell cytoplasm, was incubated with each mouse serum to allow antibody binding, followed by infection of HEK 293T cells expressing the ACE2 receptor. The fraction infectivity was determined by measuring RLU of the monolayers. An amount of virus was used to achieve a signal >1000 above background. Sera from the sham-immunized control group and immunization groups receiving OMVs from strains harboring the empty vector (VΔΔΔ p, EΔΔΔ p, and EVE p) showed no measurable neutralization activity. Using the highest sera concentration, i.e., 1:4 dilution, a significant neutralizing activity was observed for all three immunization groups receiving OMVs decorated with LOR, i.e., VΔΔΔ pLOR-V, EΔΔΔ pLOR-E and EVE pLOR-E/V, compared to the sham-immunized control group (Figure 7). In case of sera from the EVE pLOR-E/V group a significant neutralizing activity was also detectable for following two dilutions (1:14 and 1:49). The IC50 for the serum from the immunization group with alternating regimen (EVE pLOR-E/V) was approximately 1:142, which was about fourfold higher than for the repeating regimens (VΔΔΔ pLOR-V and EΔΔΔ pLOR- E). Discussion In this study, we focused on the development of a mucosally delivered, vesicle-based, subunit vaccine, which can stimulate a robust immune response directed against the SARS- CoV-2 Spike protein. Therefore, we introduce a bacterial OMV decoration strategy utilizing Lpp-OmpA’ fusions. OMVs derived from V. cholerae and ETEC have been chosen as platform as both are highly immunogenic and have been successfully used in previous immunization studies (Schild et al., 2008, 2009; Bishop et al., 2010; Leitner et al., 2013, 2015). Parental strains of V. cholerae and ETEC have been optimized to exhibit increased OMV release, lack the enterotoxins, and show reduced LPS reactivity. The genetically engineered OMV production strains were successfully used for heterologous antigen expression to generate detoxified OMVs decorated with the RBD portion of the SARS- CoV-2 Spike protein. In general, intranasal immunization with OMVs induced a robust immune response against surface structures of the respective donor bacteria, but only mice immunized with RBD-decorated OMVs induced a significant Spike protein-specific immune response. Concordantly, neutralizing activity was only observed for sera from mice immunized with RBD-decorated OMVs. The results from the Spike-pseudotyped lentivirus neutralization assay indicate the induction of a neutralizing-antibody response upon intranasal immunization with RBD-decorated OMVs. An interesting and potentially important finding was the increased efficacy of the alternating immunization regimen using RBD-decorated OMVs derived from ETEC and from V. cholerae (the EVE pLOR-E/V group). This group not only showed the highest titers against the Spike protein, but sera from that group also exhibited the strongest potency for neutralization in the cell culture infection assay. Thus, a change of the OMV donor species between immunization can specifically boost the immune response against the heterologously expressed common antigen of interest, i.e., RBD of the Spike protein. A similar approach was successfully applied along the development of the recombinant adenovirus-based vaccine (Sputnik V), which uses two different adenovirus vectors, with rAd26 in the first and rAd5 in the second immunization dose, respectively (Logunov et al., 2021). It should be noted that the alternating regimen reduces the immunization with OMVs derived from ETEC to two doses and OMVs derived from V. cholerae to one dose. Thus, it is not surprising that the immune response against the OMVs themselves is lowest in these groups compared to the groups receiving the same OMV type in all three immunizations. The study raises several questions to be addressed in future studies, such as the longevity of the immune response, the optimal OMVs amount per immunization dose, and number of booster immunizations. Previous reports demonstrated that a two-dose immunization schedule with V. cholerae OMVs is sufficient to induce a long-lasting, protective immune response against the bacterial pathogen (Bishop et al., 2010). Thus, three immunizations might not even be required. Along this first report of our OMV-based SARS- CoV-2 vaccine candidate we focused on the original Wuhan Hu-1 isolate. Future studies need to address the neutralization activity against epidemiologically relevant SARS-CoV-2 variants with RBM mutations, which have been emerging in the last months. However, even if a reduced efficacy would be observed, the fusion strategy presented herein is adaptable to decorate OMVs with appropriate antigens upon emergence of new viral variants. Some OMV-based vaccine technologies purify the OMVs and the heterologous antigens separately in a first step to subsequently coat or load the vesicles in a second step (Li and Liu, 2020; Zingl et al., 2021a). Expression of the heterologous antigen directly in the bacterial OMV donor species simplifies the production process and may reduce costs. Once purified, the decorated OMVs could be used for immunization without any further manipulation. However, it is likely that host-specific post-translational modifications are lacking along the bacterial expression system., e.g., glycosylation or disulfide bonds. Indeed, there are four predicted disulfide bonds within the CTD (Lan et al., 2020). Although V. cholerae and ETEC encode periplasmic enzymes for catalyzing disulfide bond formation and isomerization, the incorrect formation of disulfide bonds cannot be excluded. Two of the four disulfide bonds form between sequential, closely spaced cysteines (C336-C361 and C480-C488) are therefore likely to form correctly, while the remaining two (C379-C432 and C391-C525) are non-sequential. To reduce protein instability due to misfolding, 26 amino acids from the C-terminal end of the RBD were excluded from the LOR fusion protein, which consequently lacked the C525. In addition, the Spike protein is heavily glycosylated with two potential N-glycosylation sites (N331 and N343) in the RBD (Watanabe et al., 2020; Sanda et al., 2021). Notably, the lack of glycosylation in our bacterial system did not seem to affect the generation of a protective immune response. This could be attributed to their localization outside of the RBM, likely being the most important target antigen for neutralizing antibodies. Lack of such post-translational modifications may even be advantageous, since glycosylation of viral envelope proteins can interfere with the ability of the host to raise an adaptive immune response (Baum and Cobb, 2017). The intranasal administration route could have advantages compared to the currently approved vaccines relying on intramuscular injections. Consistent with previous observations, intranasal application of OMVs results in robust IgG and IgA titers representing mucosal and systemic immune responses, which may act synergistically for protection (Schild et al., 2008, 2009; Bishop et al., 2010; Roier et al., 2012, 2013; Leitner et al., 2013, 2015). Vaccine development against SARS-CoV-2 has mainly focused on injections, which predominantly induce an IgG and potentially cytotoxic T cell response but lack a mucosal immune response (Russell et al., 2020; Vabret et al., 2020). However, SARS- CoV-2 is a mucosal pathogen of the nasopharynx and respiratory tract. It is becoming increasingly evident that a mucosal IgA response could be valuable for protection against SARS-CoV-2, vaccine efficacy and clearance of the virus from the infection site (Quinti et al., 2021; Wang et al., 2021). Indeed, IgA antibodies have been shown to neutralize the virus via binding to the RBD of SARS-CoV-2 (Ma et al., 2020; Yu et al., 2020; Sterlin et al., 2021). Based on the low secretory IgA titers in fecal pellets, lung lavage might have been a better option to analyze mucosal immune responses and should be preferred in future studies. It must be noted that lung lavage was not approved by our current animal protocol and is an endpoint assay requiring killing of the mice, while fecal pellet extraction allowed us to maintain the animals and do temporal studies, i.e., collection on days 42 and 60. Thus, we cannot exclude that other administration routes, e.g., subcutaneous, intramuscular or oral, or combinations thereof would improve the immunization efficacy, which also needs to be addressed in future comparative studies. Several widely used vaccines have demonstrated high protective efficacy against COVID-19. However, knowledge on the longevity of antibody responses to current vaccines remains limited and additional booster vaccinations have already started in some countries, while several areas in the world do not even have sufficient access to vaccines yet. For example, less than 3% of the population are currently vaccinated on the African continent (CDC, 2021). Thus, there is still demand for development of additional vaccines to increase production volumes, facilitate accessibility as well as broaden and strengthen the immune response. In contrast to intramuscular injections used for currently approved vaccines, the non-invasive intranasal application allows an easy administration without the requirement of sterile equipment, e.g., syringes and needles. Moreover, OMV vaccine lots have been demonstrated to be stable for months even at room temperature and do not require stabilization additives (Vyssokikh et al., 2002; Bishop et al., 2010). OMVs also have advantages in terms of vaccine production. In general, donor bacteria are easy to cultivate and replicate fast. There are defined mutations, e.g., deletion of ompA, that result in hypervesiculating strains (Song et al., 2008; Schwechheimer and Kuehn, 2015), which in this study increased the yield of OMVs by approximately fivefold. Based on the yields presented herein, approximately 8–10 mg protein equivalent of OMVs can be isolated per 1 L of culture volume. Using a three dose regimen with 25 μg OMVs (protein equivalent) per immunization, the OMVs isolated from 1 L last for more than 100 animals. Additionally, OMVs are non-replicative and their isolation does not require treatment with inactivating agents, thus preserving the native state of antigens. These features are advantageous for mass vaccination campaigns in less developed areas of the world and could be executed even without trained health care staff. Not surprisingly, a significant amount of research has been recently dedicated to OMV-based vaccines, including the FDA-approved vesicle-based vaccine against meningitis serogroup B (Holst et al., 2009; Caron et al., 2011; Esposito and Principi, 2014; Gandhi et al., 2016). In comparison to currently approved mRNA-based vaccines against SARS-CoV-2, an OMV-based vaccine candidate warrants easier storage and administration, which could permit worldwide use even in areas with low infrastructure and limited access to cold chain devices and medical equipment. References Abdulla, Z. A., Al-Bashir, S. M., Al-Salih, N. S., Aldamen, A. A., and Abdulazeez, M. Z. (2021). A summary of the SARS-CoV-2 vaccines and technologies available or under development. Pathogens 10:788. doi: 10.3390/pathogens10070788 Baum, L. G., and Cobb, B. A. (2017). The direct and indirect effects of glycans on immune function. Glycobiology 27, 619–624. doi: 10.1093/glycob/cwx036 Bishop, A. L., Schild, S., Patimalla, B., Klein, B., and Camilli, A. (2010). Mucosal immunization with Vibrio cholerae outer membrane vesicles provides maternal protection mediated by antilipopolysaccharide antibodies that inhibit bacterial motility. Infect. Immun. 78, 4402–4420. doi: 10.1128/IAI.00398-10 Bishop, A. L., Tarique, A. A., Patimalla, B., Calderwood, S. B., Qadri, F., and Camilli, A. (2012). Immunization of mice with Vibrio cholerae outer-membrane vesicles protects against hyperinfectious challenge and blocks transmission. J. Infect. Dis.205, 412– 421. doi: 10.1093/infdis/jir756 Bosch, B. J., Van Der Zee, R., De Haan, C. A., and Rottier, P. J. (2003). The coronavirus spike protein is a class I virus fusion protein: structural and functional characterization of the fusion core complex. J. Virol. 77, 8801–8811. doi: 10.1128/jvi.77.16.8801-8811.2003 Cao, Y., Su, B., Guo, X., Sun, W., Deng, Y., Bao, L., et al. (2020). Potent neutralizing antibodies against SARS-CoV-2 identified by high-throughput single-cell sequencing of convalescent patients’. Br. Cells Cell. 182:e16. doi: 10.1016/j.cell.2020.05.025 Caron, F., Du Chatelet, I. P., Leroy, J. P., Ruckly, C., Blanchard, M., Bohic, N., et al. (2011). From tailor-made to ready-to-wear meningococcal B vaccines: longitudinal study of a clonal meningococcal B outbreak. Lancet Infect. Dis.11, 455–463. doi: 10.1016/S1473- 3099(11)70027-5 CDC, A. (2021). COVID-19 Vaccination. Available online at: https://africacdc.org/covid-19-vaccination/ (accessed September 8, 2021). Crawford, K. H. D., Eguia, R., Dingens, A. S., Loes, A. N., Malone, K. D., Wolf, C. R., et al. (2020). Protocol and reagents for pseudotyping lentiviral particles with SARS- CoV-2 spike protein for neutralization assays. Viruses 12:513. doi: 10.3390/v12050513 Daugherty, P. S., Chen, G., Olsen, M. J., Iverson, B. L., and Georgiou, G. (1998). Antibody affinity maturation using bacterial surface display. Protein Eng.11, 825–832. doi: 10.1093/protein/11.9.825 Donnenberg, M. S., and Kaper, J. B. (1991). Construction of an eae deletion mutant of enteropathogenic Escherichia coli by using a positive-selection suicide vector. Infect. Immun.59, 4310–4317. doi: 10.1128/iai.59.12.4310-4317.1991 Earhart, C. F. (2000). Use of an lpp-ompa fusion vehicle for bacterial surface display. Methods Enzymol.326, 506–516. doi: 10.1016/s0076-6879(00)26072-2 Ellis, T. N., and Kuehn, M. J. (2010). Virulence and immunomodulatory roles of bacterial outer membrane vesicles. Microbiol. Mol. Biol. Rev. 74, 81–94. doi: 10.1128/mmbr.00031-09 Esposito, S., and Principi, N. (2014). Vaccine profile of 4CMenB: a four-component Neisseria meningitidis serogroup B vaccine. Exp. Rev. Vaccines 13, 193–202. doi: 10.1586/14760584.2014.874949 Evans, D. J. Jr., and Evans, D. G. (1973). Three characteristics associated with enterotoxigenic Escherichia coli isolated from man. Infect. Immun. 8, 322–328. doi: 10.1128/iai.8.3.322-328.1973 Francisco, J. A., Earhart, C. F., and Georgiou, G. (1992). Transport and anchoring of beta-lactamase to the external surface of Escherichia coli. Proc. Natl. Acad. Sci. U.S.A. 89, 2713–2717. doi: 10.1073/pnas.89.7.2713 Gandhi, A., Balmer, P., and York, L. J. (2016). Characteristics of a new meningococcal serogroup B vaccine, bivalent rLP2086 (MenB-FHbp. Trumenba Postgrad Med.128, 548–556. doi: 10.1080/00325481.2016.1203238 Gerritzen, M. J. H., Martens, D. E., Wijffels, R. H., Van Der Pol, L., and Stork, M. (2017). Bioengineering bacterial outer membrane vesicles as vaccine platform. Biotechnol Adv 35, 565–574. Gnopo, Y. M. D., Watkins, H. C., Stevenson, T. C., Delisa, M. P., and Putnam, D. (2017). Designer outer membrane vesicles as immunomodulatory systemsreprogramming bacteria for vaccine delivery. Adv. Drug Deliv. Rev. 114, 132–142. doi: 10.1016/j.addr.2017.05.003 Grifoni, A., Sidney, J., Zhang, Y., Scheuermann, R. H., Peters, B., and Sette, A. (2020). A sequence homology and bioinformatic approach can predict candidate targets for immune responses to SARS-CoV-2. Cell Host. Microbe. 27, 671–680.e672. doi: 10.1016/j.chom.2020.03.002 Hanahan, D. (1983). Studies on transformation of Escherichia coli with plasmids. J. Mol. Biol.166, 557–580. Hirano, T., Jiao, X., Chen, Z., Van Waes, C., and Gu, X. X. (2006). Kinetics of mouse antibody and lymphocyte responses during intranasal vaccination with a lipooligosaccharide-based conjugate vaccine. Immunol. Lett. 107, 131–139. doi: 10.1016/j.imlet.2006.08.005 Holst, J., Martin, D., Arnold, R., Huergo, C. C., Oster, P., O’hallahan, J., et al. (2009). Properties and clinical performance of vaccines containing outer membrane vesicles from Neisseria meningitidis. Vaccine 27, B3–B12. doi: 10.1016/j.vaccine.2009.04.071 Kang, D., Gho, Y. S., Suh, M., and Kang, C. (2002). Highly sensitive and fast protein detection with coomassie brilliant blue in sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Bull. Kor. Chem. Soc.23, 1511–1512. doi: 10.5012/bkcs.2002.23.11.1511 Kulp, A., and Kuehn, M. J. (2010). Biological functions and biogenesis of secreted bacterial outer membrane vesicles. Annu Rev. Microbiol. 64, 163–184. doi: 10.1146/annurev.micro.091208.073413 Lan, J., Ge, J., Yu, J., Shan, S., Zhou, H., Fan, S., et al. (2020). Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 581, 215– 220. Leitner, D. R., Feichter, S., Schild-Prufert, K., Rechberger, G. N., Reidl, J., and Schild, S. (2013). Lipopolysaccharide modifications of a cholera vaccine candidate based on outer membrane vesicles reduce endotoxicity and reveal the major protective antigen. Infect. Immun.81, 2379–2393. doi: 10.1128/IAI.01382-12 Leitner, D. R., Lichtenegger, S., Temel, P., Zingl, F. G., Ratzberger, D., Roier, S., et al. (2015). A combined vaccine approach against Vibrio cholerae and ETEC based on outer membrane vesicles. Front. Microbiol.6:823. doi: 10.3389/fmicb.2015.00823 Li, R., and Liu, Q. (2020). Engineered bacterial outer membrane vesicles as multifunctional delivery platforms. Front. Mater. 7, 2150–2159. doi: 10.1016/j.bbamem.2009.08.001 Logunov, D. Y., Dolzhikova, I. V., Shcheblyakov, D. V., Tukhvatulin, A. I., Zubkova, O. V., Dzharullaeva, A. S., et al. (2021). Safety and efficacy of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine: an interim analysis of a randomised controlled phase 3 trial in Russia. Lancet 397, 671–681. doi: 10.1016/S0140- 6736(21)00234-8 Ma, H., Zeng, W. H., He, H. L., Zhao, D., Jiang, D. H., Zhou, P. G., et al. (2020). Serum IgA, IgM, and IgG responses in COVID-19. Cell. Mol. Immunol.17, 773–775. doi: 10.1038/s41423-020-0474-z Miller, V. L., and Mekalanos, J. J. (1988). A novel suicide vector and its use in construction of insertion mutations: osmoregulation of outer membrane proteins and virulence determinants in Vibrio cholerae requires toxR. J. Bacteriol.170, 2575–2583. doi: 10.1128/jb.170.6.2575-2583.1988 Miller, V. L., Dirita, V. J., and Mekalanos, J. J. (1989). Identification of toxS, a regulatory gene whose product enhances toxR-mediated activation of the cholera toxin promoter. J. Bacteriol.171, 1288–1293. doi: 10.1128/jb.171.3.1288-1293.1989 Morales, V. M., Backman, A., and Bagdasarian, M. (1991). A series of wide-host- range low-copy-number vectors that allow direct screening for recombinants. Gene 97, 39– 47. doi: 10.1016/0378-1119(91)90007-x Pathirana, R. D., and Kaparakis-Liaskos, M. (2016). Bacterial membrane vesicles: Biogenesis, immune regulation and pathogenesis. Cell Microbiol.18, 1518–1524. Plotkin, S. (2014). History of vaccination. Proc. Natl. Acad. Sci. U.S.A.111, 12283– 12287. Pormohammad, A., Zarei, M., Ghorbani, S., Mohammadi, M., Razizadeh, M. H., Turner, D. L., et al. (2021). Efficacy and safety of COVID-19 vaccines: a systematic review and meta-analysis of randomized clinical trials. Vaccines (Basel) 9:467. doi: 10.3390/vaccines9050467 Pressler, K., Vorkapic, D., Lichtenegger, S., Malli, G., Barilich, B. P., Cakar, F., et al. (2016). AAA+ proteases and their role in distinct stages along the Vibrio cholerae lifecycle. Int. J. Med. Microbiol.306, 452–462. doi: 10.1016/j.ijmm.2016.05.013 Quinti, I., Mortari, E. P., Fernandez Salinas, A., Milito, C., and Carsetti, R. (2021). IgA antibodies and IgA deficiency in SARS-CoV-2 infection. Front. Cell. Infect. Microbiol. 11:655896. Roier, S., Fenninger, J. C., Leitner, D. R., Rechberger, G. N., Reidl, J., and Schild, S. (2013). Immunogenicity of Pasteurella multocida and Mannheimia haemolytica outer membrane vesicles. Int. J. Med. Microbiol.303, 247–256. doi: 10.1016/j.ijmm.2013.05.001 Roier, S., Leitner, D. R., Iwashkiw, J., Schild-Prufert, K., Feldman, M. F., Krohne, G., et al. (2012). Intranasal immunization with nontypeable Haemophilus influenzae outer membrane vesicles induces cross-protective immunity in mice. PLoS One 7:e42664. doi: 10.1371/journal.pone.0042664 Roier, S., Zingl, F. G., Cakar, F., Durakovic, S., Kohl, P., Eichmann, T. O., et al. (2016). A novel mechanism for the biogenesis of outer membrane vesicles in gram-negative bacteria. Nat. Commun.7:10515. Russell, M. W., Moldoveanu, Z., Ogra, P. L., and Mestecky, J. (2020). Mucosal immunity in COVID-19: a neglected but critical aspect of SARS-CoV-2 infection. Front. Immunol.11:611337. doi: 10.3389/fimmu.2020.611337 Sanda, M., Morrison, L., and Goldman, R. (2021). N- and O-glycosylation of the SARS-CoV-2 spike protein. Anal. Chem.93, 2003–2009. Schild, S., Nelson, E. J., and Camilli, A. (2008). Immunization with Vibrio cholerae outer membrane vesicles induces protective immunity in mice. Infect. Immun. 76, 4554– 4563. doi: 10.1128/IAI.00532-08 Schild, S., Nelson, E. J., Bishop, A. L., and Camilli, A. (2009). Characterization of Vibrio cholerae outer membrane vesicles as a candidate vaccine for cholera. Infect. Immun. 77, 472–484. doi: 10.1128/IAI.01139-08 Schwechheimer, C., and Kuehn, M. J. (2015). Outer-membrane vesicles from Gram- negative bacteria: biogenesis and functions. Nat. Rev. Microbiol. 13, 605–619. doi: 10.1038/nrmicro3525 Seper, A., Fengler, V. H., Roier, S., Wolinski, H., Kohlwein, S. D., Bishop, A. L., et al. (2011). Extracellular nucleases and extracellular DNA play important roles in Vibrio cholerae biofilm formation. Mol. Microbiol. 82, 1015–1037. doi: 10.1111/j.1365- 2958.2011.07867.x Song, T., Mika, F., Lindmark, B., Liu, Z., Schild, S., Bishop, A., et al. (2008). A new Vibrio cholerae sRNA modulates colonization and affects release of outer membrane vesicles. Mol. Microbiol.70, 100–111. doi: 10.1111/j.1365-2958.2008.06392.x Stathopoulos, C., Georgiou, G., and Earhart, C. F. (1996). Characterization of Escherichia coli expressing an Lpp’OmpA(46-159)-PhoA fusion protein localized in the outer membrane. Appl. Microbiol. Biotechnol.45, 112–119. doi: 10.1007/s002530050657 Sterlin, D., Mathian, A., Miyara, M., Mohr, A., Anna, F., Claer, L., et al. (2021). IgA dominates the early neutralizing antibody response to SARS-CoV-2. Sci. Trans. Med. 13:eabd2223. doi: 10.1126/scitranslmed.abd2223 Vabret, N., Britton, G. J., Gruber, C., Hegde, S., Kim, J., Kuksin, M., et al. (2020). Immunology of COVID-19: current state of the science. Immunity 52, 910–941. van der Pol, L., Stork, M., and Van Der Ley, P. (2015). Outer membrane vesicles as platform vaccine technology. Biotechnol. J.10, 1689–1706. doi: 10.1002/biot.201400395 Vetvik, H., Grewal, H. M., Haugen, I. L., Ahren, C., and Haneberg, B. (1998). Mucosal antibodies can be measured in air-dried samples of saliva and feces. J. Immunol. Methods 215, 163–172. doi: 10.1016/s0022-1759(98)00089-1 Vyssokikh, M. Y., Zorova, L., Zorov, D., Heimlich, G., Jurgensmeier, J. J., and Brdiczka, D. (2002). Bax releases cytochrome c preferentially from a complex between porin and adenine nucleotide translocator. hexokinase activity suppresses this effect. Mol. Biol. Rep.29, 93–96. doi: 10.1023/a:1020383108620 Wang, Z., Lorenzi, J. C. C., Muecksch, F., Finkin, S., Viant, C., Gaebler, C., et al. (2021). Enhanced SARS-CoV-2 neutralization by dimeric IgA. Sci. Trans. Med. 13, eabf1555. doi: 10.1126/scitranslmed.abf1555 Watanabe, Y., Allen, J. D., Wrapp, D., Mclellan, J. S., and Crispin, M. (2020). Site- specific glycan analysis of the SARS-CoV-2 spike. Science 369, 330–333. doi: 10.1126/science.abb9983 Yi, C., Sun, X., Ye, J., Ding, L., Liu, M., Yang, Z., et al. (2020). Key residues of the receptor binding motif in the spike protein of SARS-CoV-2 that interact with ACE2 and neutralizing antibodies. Cell Mol. Immunol.17, 621–630. doi: 10.1038/s41423-020-0458-z Yu, H. Q., Sun, B. Q., Fang, Z. F., Zhao, J. C., Liu, X. Y., Li, Y. M., et al. (2020). Distinct features of SARS-CoV-2-specific IgA response in COVID-19 patients. Eur. Respir. J.56:2001526. doi: 10.1183/13993003.01526-2020 Zingl, F. G., Leitner, D., Thapa, H. B., and Schild, S. (2021a). Outer membrane vesicles as versatile tools for therapeutic approaches. microLife 2:uqab006. Zingl, F. G., Thapa, H. B., Scharf, M., Kohl, P., Muller, A. M., and Schild, S. (2021b). Outer membrane vesicles of Vibrio cholerae protect and deliver active cholera toxin to host cells via porin-dependent uptake. mBio 12:e0053421. doi: 10.1128/mBio.00534-21 In the foregoing description, it will be readily apparent to one skilled in the art that varying substitutions and modifications may be made to the invention disclosed herein without departing from the scope and spirit of the invention. The invention illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein. The terms and expressions which have been employed are used as terms of description and not of limitation, and there is no intention that in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention. Thus, it should be understood that although the present invention has been illustrated by specific embodiments and optional features, modification and/or variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention. Citations to a number of patent and non-patent references may be made herein. The cited references are incorporated by reference herein in their entireties. In the event that there is an inconsistency between a definition of a term in the specification as compared to a definition of the term in a cited reference, the term should be interpreted based on the definition in the specification.

Claims

CLAIMS 1. A composition comprising a fusion protein, the fusion protein comprising from N-terminus to C-terminus: (i) the N-terminal signal sequence of the E. coli Lpp protein fused in frame either directly or via a linker to (ii) a fragment of the E. coli OmpA protein, wherein the fragment of the E. coli OmpA protein comprises a N-terminal deletion and a C-terminal deletion, and the fragment of the E. coli OmpA protein is fused either directly or via linker to (iii) a fragment of the SARS CoV-2 Spike protein comprising the receptor binding domain (RBD). 2. The composition of claim 1, wherein the composition comprises outer membrane vesicles (OMVs) and the fusion protein is present on the surface of the OMVs. 3. The composition of claim 2, wherein the OMVs are derived from V. cholerae. 4. The composition of claim 2, wherein the OMVs are derived from E. coli. 5. A V. cholerae cell transfected with an expression vector that expresses the fusion protein of claim 1. 6. An E. coli cell transfected with an expression vector that expresses the fusion protein of claim 1. 7. An expression vector that expresses the fusion protein of claim 1. 8. The expression vector of claim 7, wherein the fusion protein is encoded by a polynucleotide that comprises codons that are optimized for expression in V. cholerae. 9. The expression vector of claim 7, wherein the fusion protein is encoded by a polynucleotide that comprises codons that are optimized for expression in E. coli. 10. A method for inducing an immune response against SARS CoV-2 Spike protein, the method comprising administering the composition of claim 1 to a subject in need thereof. 11. The method of claim 10, wherein the immune response comprises an antibody response against SARS CoV-2 Spike protein. 12. The method of claim 10, wherein the immune response comprises a neutralizing antibody response against SARS CoV-2 Spike protein. 13. The method of claim 10, wherein the method induces a protective response against infection by SARS CoV-2. 14. The composition of claim 1, wherein the fusion protein comprises SEQ ID NO: 2 or SEQ ID NO: 4 15. The method of claim 10, wherein the method comprises administering a composition comprising SEQ ID NO: 2 or SEQ ID NO: 4 to a subject in need thereof. 16. The method of claim 10, wherein the method comprises administering a composition comprising SEQ ID NO: 2 and SEQ ID NO: 4 to a subject in need thereof.
PCT/US2022/076219 2021-09-09 2022-09-09 Outer membrane vesicle (omv)-based vaccines against sudden acute respiratory syndrome coronavirus 2 (sars-cov-2) spike protein WO2023039535A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163261027P 2021-09-09 2021-09-09
US63/261,027 2021-09-09

Publications (1)

Publication Number Publication Date
WO2023039535A1 true WO2023039535A1 (en) 2023-03-16

Family

ID=85506955

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/076219 WO2023039535A1 (en) 2021-09-09 2022-09-09 Outer membrane vesicle (omv)-based vaccines against sudden acute respiratory syndrome coronavirus 2 (sars-cov-2) spike protein

Country Status (1)

Country Link
WO (1) WO2023039535A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040076976A1 (en) * 2000-11-02 2004-04-22 Pan Shen Quan Aopb gene, protein,homologs, fragments and variants thereof, and their use for cell surface display
US7189402B1 (en) * 1998-02-06 2007-03-13 Research Development Foundation Live vaccine for human immunodeficiency virus
US20170080080A1 (en) * 2014-05-19 2017-03-23 Board Of Regents, The University Of Texas System Combinatorial platform for the display of surface adjuvants and antigens
US20210262941A1 (en) * 2019-11-27 2021-08-26 Promega Corporation Multipartite luciferase peptides and polypeptides

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7189402B1 (en) * 1998-02-06 2007-03-13 Research Development Foundation Live vaccine for human immunodeficiency virus
US20040076976A1 (en) * 2000-11-02 2004-04-22 Pan Shen Quan Aopb gene, protein,homologs, fragments and variants thereof, and their use for cell surface display
US20170080080A1 (en) * 2014-05-19 2017-03-23 Board Of Regents, The University Of Texas System Combinatorial platform for the display of surface adjuvants and antigens
US20210262941A1 (en) * 2019-11-27 2021-08-26 Promega Corporation Multipartite luciferase peptides and polypeptides

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
FRANCISCO ET AL.: "Transport and anchoring of beta-lactamase to the external surface of Escherichia coli", PROC NATL ACAD SCI USA, vol. 89, April 1992 (1992-04-01), pages 2713 - 2717, XP001203586, DOI: 10.1073/pnas.89.7.2713 *
THAPA HIMADRI B., MÜLLER ANNA M., CAMILLI ANDREW, SCHILD STEFAN: "An Intranasal Vaccine Based on Outer Membrane Vesicles Against SARS-CoV-2", FRONTIERS IN MICROBIOLOGY, vol. 12, XP093047095, DOI: 10.3389/fmicb.2021.752739 *
VIJAYAKUMAR VIDHYA, VANHOVE AUDREY S., PICKERING BRADLEY S., LIAO JULIE, TIERNEY BRADEN T., ASARA JOHN M., BRONSON RODERICK, WATNI: "Removal of a Membrane Anchor Reveals the Opposing Regulatory Functions of Vibrio cholerae Glucose-Specific Enzyme IIA in Biofilms and the Mammalian Intestine", MBIO, AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 9, no. 5, 7 November 2018 (2018-11-07), US , XP093047097, ISSN: 2161-2129, DOI: 10.1128/mBio.00858-18 *
YANG ET AL.: "A vaccine targeting the RBD of the S protein of SARS-CoV-2 induces protective immunit y", NATURE, vol. 586, 29 July 2020 (2020-07-29), pages 572 - 577, XP037277111, DOI: 10.1038/s41586-020-2599-8 *

Similar Documents

Publication Publication Date Title
WO2021185310A1 (en) Mvsv virus vector and virus vector vaccine thereof, and novel coronavirus pneumonia vaccine based on mvsv mediation
EP2066339B1 (en) Compositions and methods of enhancing immune responses
Thapa et al. An intranasal vaccine based on outer membrane vesicles against SARS-CoV-2
JP6473086B2 (en) Recombinant Bordetella strain
Wei et al. Immunogenicity and protective efficacy of orally or intranasally administered recombinant Lactobacillus casei expressing ETEC K99
JP2010500399A (en) Immunogen from Urinary Pathogenic Escherichia coli
US20210338804A1 (en) Vaccine Compositions For Preventing Coronavirus Disease
AU2005294436A1 (en) Methods and compositions for immunizing against Pseudomonas infection
US9750793B2 (en) Multifunctional oral vaccine based on chromosome recombineering
US11744884B2 (en) Live Salmonella typhi vectors engineered to express heterologous outer membrane protein antigens and methods of use thereof
EA027981B1 (en) Chimeric protein vaccine against pneumonia caused by streptococcus pneumoniae
JP4653934B2 (en) Bacteriophage mediated immunization methods
KR20230154048A (en) Compositions containing SBI adjuvants and methods of using them
US9511135B2 (en) Immunogenic respiratory syncytial virus glycoprotein-containing VLPs and related compositions, constructs, and therapeutic methods
Moore et al. Enhanced protective immunity against pneumococcal infection with PspA DNA and protein
WO2023039535A1 (en) Outer membrane vesicle (omv)-based vaccines against sudden acute respiratory syndrome coronavirus 2 (sars-cov-2) spike protein
AU2021298886A1 (en) A dna plasmid SARS-Coronavirus-2/covid-19 vaccine
Xu et al. Intramuscular delivery of a cholera DNA vaccine primes both systemic and mucosal protective antibody responses against cholera
JP2002531135A (en) Two-step immunization for Chlamydia infection
US20150238590A1 (en) Use of the salmonella spp type iii secretion proteins as a protective vaccination
US20170035878A1 (en) Multi-Functional Mucosal Vaccine Platform
Ruth et al. DNA vaccination for the priming of neutralizing antibodies against non-immunogenic STa enterotoxin from enterotoxigenic Escherichia coli
JP4535874B2 (en) Bacteriophage-mediated immunity against hepatitis
US20240016916A1 (en) Bacterial outer membrane vesicles carrying coronavirus polypeptides, method of preparation, compositions and use thereof
Orne Vaccinogenic and Therapeutic Applications of Burkholderia Polysaccharides

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: 22868347

Country of ref document: EP

Kind code of ref document: A1