WO2023060220A1 - Methods of immunization against coronavirus - Google Patents

Methods of immunization against coronavirus Download PDF

Info

Publication number
WO2023060220A1
WO2023060220A1 PCT/US2022/077737 US2022077737W WO2023060220A1 WO 2023060220 A1 WO2023060220 A1 WO 2023060220A1 US 2022077737 W US2022077737 W US 2022077737W WO 2023060220 A1 WO2023060220 A1 WO 2023060220A1
Authority
WO
WIPO (PCT)
Prior art keywords
cov
sars
protein
human subject
fragment
Prior art date
Application number
PCT/US2022/077737
Other languages
French (fr)
Inventor
David Berd
James PASSIN
Original Assignee
BioVaxys Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by BioVaxys Inc. filed Critical BioVaxys Inc.
Publication of WO2023060220A1 publication Critical patent/WO2023060220A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • a computer readable text file entitled “SequenceListing.txt” created on October 6, 2021 with a file size of 32,216 bytes contains the sequence listing for this application and is hereby incorporated by reference in its entirety.
  • the invention described herein relates generally to method of immunizing a human subject against multiple species of sarbecovirus.
  • Coronaviruses are plus-strand RNA viruses that cause disease in animals and humans.
  • a novel zoonotic coronavirus outbreak started in Wuhan, China in 2019.
  • This pandemic disease has now been defined as novel coronavirus disease 2019 (Covid-19), and is sustained by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
  • SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
  • SARS-CoV-1 and SARS-CoV-2 belong to the species of SARS-related coronavirus (subgenus sarbecovirus, genus betacoronavirus). Antigenically, the two coronaviruses are placed in two distinct phylogenetic clades; convalescent serum specimens from patients with SARS or Covid-19 lack cross-neutralization, despite the majority of survivors of SARS-CoV-1 infection continuing to have detectable neutralizing antibodies against the homologous SARS-CoV-1 virus seventeen years after infection (Ten, New. Engl. J. Med. (2021) 10.1056/NEJMoa2108453).
  • coronavirus infections such as those found in humans
  • sarbecovirus such as SARS-CoV-1 infections or SARS-CoV- 2 infections resulting severe acute respiratory syndrome.
  • a need exists for identifying and developing immunogenic and vaccine compositions against sarbecovirus infections that can elicit a broadly cross-reactive neutralizing antibody immune response against multiple species of sarbecovirus.
  • immunogenic compositions and vaccine formulations are needed that can be delivered directly to or in close proximity to the site of infection to maximize therapeutic effectiveness.
  • Haptenized proteins have been widely used to provide defined epitopes for the measurement of antibody titers and affinities.
  • the present invention meets such needs and further provides other related advantages.
  • the present invention provides for cross-reactive immunogens for sarbecovirus and meets the need for immunogenic compositions and vaccine formulations that can be delivered directly or in close proximity to the site of infection to maximize a protective immune response in a human subject.
  • Embodiments disclosed herein are immunogenic compositions or vaccines comprising haptenized S proteins from coronavirus, including severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and methods of immunizing a human subject with the immunogenic compositions or vaccines.
  • SARS-CoV-1 severe acute respiratory syndrome coronavirus 1
  • SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
  • Figure 1 shows the anti-spike protein antibody response following subcutaneous administration of BVX-0320 in CF-1 mice.
  • an "immunogenic composition” or “vaccine” as used herein refers to any one or more compounds or agents or immunogens capable of priming, potentiating, activating, eliciting, stimulating, augmenting, boosting, amplifying, or enhancing an adaptive (specific) immune response, which may be cellular (T cell) or humoral (B cell), or a combination thereof.
  • the adaptive immune response is protective, which may include neutralization of a coronavirus (decreasing or eliminating virus infectivity).
  • a representative example of an immunogen is a viral antigen (such as one or more coronavirus antigens).
  • any concentration range, percentage range, ratio range, or integer range is understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer, etc.), unless otherwise indicated.
  • an effective amount refers to that amount of a compound or combination of compounds as described herein that is sufficient to effect the intended application including, but not limited to, disease treatment.
  • a therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated (e.g. the weight, age and gender of the subject), the severity of the disease condition, the manner of administration, etc. which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in target cells (e.g. the reduction of platelet adhesion and/or cell migration).
  • the specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether the compound is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which the compound is carried.
  • immunogenicity means the ability of an S protein immunogen to evoke an immune response directed to the coronavirus, including broadly cross-reactive neutralizing antibody-dependent immune responses against multiple species of sarbecovirus. Whether a S protein preparation is immunogenic can be tested by, for instance, a DTH-assay or an in vivo assay in an experimental animal model.
  • a prophylactic effect includes delaying or eliminating the appearance of a sarbecovirus infection, delaying or eliminating the onset of symptoms of a sarbecovirus infection, slowing, halting, or reversing the progression of a sarbecovirus infection, including reducing or eliminating any symptoms associated with SARS, or any combination thereof.
  • “Pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and inert ingredients.
  • the use of such pharmaceutically acceptable carriers or pharmaceutically acceptable excipients for active pharmaceutical ingredients is well known in the art. Except insofar as any conventional pharmaceutically acceptable carrier or pharmaceutically acceptable excipient is incompatible with the active pharmaceutical ingredient, its use in the therapeutic compositions of the invention is contemplated. Additional active pharmaceutical ingredients, such as other drugs, can also be incorporated into the described compositions and methods.
  • ranges are used herein to describe, for example, physical or chemical properties such as molecular weight or chemical formulae, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included.
  • Use of the term "about" when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary. The variation is typically from 0% to 15%, preferably from 0% to 10%, more preferably from 0% to 5% of the stated number or numerical range.
  • isotype refers to the antibody class (e.g. IgM or IgGl) that is encoded by the heavy chain constant region genes.
  • IgA antibody class
  • IgD antibody isotype
  • IgG immunoglobulin G
  • IgM immunoglobulin G
  • IgE immunoglobulin G
  • subclasses of the IgG isotype IgGl, lgG2, lgG3 and lgG4
  • two subclasses of the IgA isotype IgAl and lgA2.
  • sequence identity and “sequence percent identity” in the context of two or more nucleic acids or polypeptides, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity.
  • the percent identity can be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences. Suitable programs to determine percent sequence identity include for example the BLAST suite of programs available from the U.S.
  • BLASTN is used to compare nucleic acid sequences
  • BLASTP is used to compare amino acid sequences
  • ALIGN, ALIGN-2 (Genentech, South San Francisco, California) or MegAlign, available from DNASTAR are additional publicly available software programs that can be used to align sequences.
  • One skilled in the art can determine appropriate parameters for maximal alignment by particular alignment software. In certain embodiments, the default parameters of the alignment software are used.
  • haptenized vaccines using a single hapten reagent are known to the skilled artisan.
  • the invention provides for an haptenized S protein coronavirus vaccine, such a vaccine may be prepared comprising recombinantly producing an S protein or fragment thereof, and haptenizing with a haptenization reagent.
  • the step of haptenizing may be performed by modifying recombinant S protein with DNP by a 30-minute incubation with the haptenization reagent 2,4-difluoronitrobenzene (DNFB).
  • DNFB 2,4-difluoronitrobenzene
  • the haptenized S protein is washed with HBSS.
  • Preferred haptenization reagents may target the e-amino group of an amino acid.
  • the haptenization reagent is selected from trinitrochlorobenzene (TNCB), 2,4-difluoronitrobenzene (DNFB), N-iodoacetyl-N'-(5-sulfonic-l-naphthyl)ethylenediamine (AED), sulfanilic acid (SA), trinitrophenol (TNP), 2,4,6-trinitrobenzenesulfonic acid (TNBS) and combinations thereof.
  • TTCB trinitrochlorobenzene
  • DNFB 2,4-difluoronitrobenzene
  • AED N-iodoacetyl-N'-(5-sulfonic-l-naphthyl)ethylenediamine
  • SA sulfanilic acid
  • TNP trinitrophenol
  • TNBS 2,4,6-trinitrobenzenesulfonic acid
  • a haptenized S protein immunogen (and corresponding immunogenic epitopes) and fragments, and variants thereof may be produced synthetically or recombinantly.
  • a coronavirus S protein fragment that contains an epitope that induces an immune response against coronavirus may be synthesized by standard chemical methods, including synthesis by automated procedure.
  • immunogenic peptides are synthesized based on the standard solid-phase Fmoc protection strategy with HATU as the coupling agent.
  • the immunogenic peptide is cleaved from the solid-phase resin with trifluoroacetic acid containing appropriate scavengers, which also deprotects side chain functional groups.
  • the crude immunogenic peptide may be further purified using preparative reverse phase chromatography.
  • a synthetic S protein immunogen has an amino acid sequence that is identical to, or at least 85% identical (which includes at least 90% or 95% or any percent in between 85% and 100%) to SEQ ID NO :2.
  • the haptenized S protein immunogens may be recombinant, wherein desired S protein immunogens are individually or in combination expressed from a polynucleotide that is operably linked to an expression control sequence (e.g. a promoter) in a nucleic acid expression construct.
  • a recombinant S protein antigen will comprise an amino acid sequence that is identical to, or at least 85% identical (which includes at least 90% or 95% or any percent in between 80% and 100%) to SEQ. ID NO:2.
  • a recombinant S protein immunogen consists of an amino acid sequence as set forth in SEQ ID NO:1 or 2.
  • recombinant S protein immunogens and variants thereof are fragments of SEQ ID NO:1 or 2.
  • the variants are SARS-CoV-2 Spike protein variants found in different strains of SARS-CoV- 2. Exemplary variants of the Spike protein from these different strains are set forth in Table 1. Variants include, but are not limited to, Spike proteins from the alpha, beta or delta variant of SARS-CoV-2, B.1.1.7 strain, B.1.351 strain, P.l strain, CAL 20 strain or any combination thereof.
  • Table 1 List of amino acid positions and relative amino acid changes in the different variants in the Spike protein with respect to the ancestral Wuhan strain Spike protein (SEQ ID NO: 2). protein position Wuhan Bl.1.7 Bl.351 P.l CAL.20
  • the S proteins are haptenized.
  • a variety of haptens of quite different chemical structure have been shown to induce similar types of immune response, e.g. TNP (Kempkes, J. Immunol. (1991) 147:2467); phosphorylcholine (Jang, Eur. J. Immunol. (1991) 21:1303); nickel (Pistoor, J. Invest.
  • Conjugation of a hapten to a cell to elicit an immune response may preferably be accomplished by conjugation via e-amino groups of lysine or --COOH groups.
  • This group of haptens include a number of chemically diverse compounds: dinitrophenyl, trinitrophenyl, N-iodoacetyl-N'-(5-sulfonic 1-naphthyl) ethylene diamine, trinitrobenzene-sulfonic acid, dinitrobenzene sulfonic acid, fluorescein isothiocyanate, arsenic acid benzene isothiocyanate, and dinitrobenzene-S-mustard (Nahas, Cellular Immunol. (1980) 54:241).
  • haptens include a "recognition group" which is the group that interacts with an antibody.
  • the recognition group is irreversibly associated with the hapten reactive group.
  • the hapten recognition group is available for binding with antibody.
  • hapten recognition groups include without limitation to dinitriophenyl, trinitrophenyl, fluorescein, other aromatics, phosphorylcholine, peptides, advanced glycosylation endproducts (AGE), carbohydrates, etc.
  • Haptens also include a functional group for conjugation to a substituent on an amino acid side chain of a protein or polypeptide.
  • Amino acid side chain groups that can be conjugated to hapten include, e.g. free carboxylic acid groups in the aspartic acid or glutamic acid; the e-amino group of lysine; the thiol moiety of cysteine; the hydroxyl group of serine or tyrosine; the imidazole moiety of histidine; or the aryl groups of tryptophan, tyrosine, or phenylalanine.
  • Hapten functional groups capable of reacting with specific amino acid side chains are described as follows.
  • Functional groups reactive with primary amines Hapten reactive groups that would form a covalent bond with primary amines present on amino acid side chains would include, but not be limited to, acid chlorides, anhydrides, reactive esters, a,p-unsaturated ketones, imidoesters, and halonitrobenzenes. Various reactive esters with the capability of reacting with nucleophilic groups such as primary amines are available commercially. Functional groups reactive with carboxylic acids. Carboxylic acids in the presence of carbodiimides, such as EDC, can be activated, allowing for interaction with various nucleophiles, including primary and secondary amines. Alkylation of carboxylic acids to form stable esters can be achieved by interaction with sulfur or nitrogen mustards, or haptens containing either an alkyl or aryl aziridine moiety.
  • Functional groups reactive with sulfhydryl groups There are several reactive groups that can be coupled to sulfhydryl groups present on the side chains of amino acids. Haptens containing an a,p- unsaturated ketone or ester moiety, such as maleimide, provide a reactive functionality that can interact with sulfhydryl as well as amino groups. In addition, a reactive disulfide group, such as 2-pyridyldithio group or a 5,5'-dithio-bis-(2-nitrobenzoic acid) group is also applicable.
  • reagents containing reactive disulfide bonds include N-succinimidly 3-(2-pyridyl-dithio) propionate (Carlsson, Biochem J. (1978) 173:723-737), sodium S-4-succinimidyloxycarbonyl-alpha-methylbenzyl-thiosulfate, and 4 succinimidyloxycarbonyl-alpha-methyl-(2-pyridyldithio)-toluene.
  • Other functional molecules include succinimidyl 3-(maleimido)-propionate, sulfosuccinimidyl 4- (p-maleimido-phenyl)butyrate, sulfo-succinimidyl-4-(N-maleimidomethyl-cyclohexane)-l-carboxylate, maleimidobenzolyl-N-hydroxy-succinimide ester.
  • any hapten or combination of different haptens can be used in the compositions of the invention.
  • the same hapten recognition group is coupled to different amino acids through different functional groups on the S protein.
  • the reagents dinitrobenzene sulfonic acid, dinitro phenyldiazonium, and dinitrobenzene S mustard all form the dinitrophenyl hapten coupled to amino groups, aromatic groups, and carboxylic acid groups, respectively.
  • an arsonic acid hapten can be coupled by reacting arsonic acid benzene isothiocyanate to amino groups or azobenzenearsonate to aromatic groups.
  • Immunogenic compositions or vaccines as described herein useful for treating and/or preventing a coronavirus infection comprises immunogenic sarbecovirus polypeptides, such as S protein, fragments, and variants thereof, and also includes a fusion of a sarbecovirus immunogen to other peptides or polypeptides (e.g. a hydrophobic amino acid sequence or a histidine tag or a non-S protein coronavirus polypeptide or fragment thereof) or other modifications (e.g. glycosylation).
  • the immunogenic S polypeptides may comprise any portion of an S protein that has an epitope capable of eliciting a protective immune response (e.g.
  • Immunogenic polypeptides as described herein may be arranged, combined, or fused in a linear form, and each immunogen may or may not be reiterated, wherein the reiteration may occur once or multiple times, and may be located at the N-terminus, C-terminus, or internal to a linear sequence of immunogenic S or other sarbecovirus polypeptide immunogens.
  • a plurality of different coronavirus immunogenic polypeptides e.g.
  • S proteins from multiple sarbecovirus species can be selected and mixed or combined into a cocktail composition to provide a multivalent vaccine for use in eliciting a protective immune response without a harmful or otherwise unwanted associated immune responses or side effects.
  • methods for producing a haptenized synthetic or recombinant multivalent coronavirus polypeptide immunogens including fusion proteins.
  • host cells containing an S protein immunogen-encoding nucleic acid expression construct may be cultured to produce the recombinant S protein immunogen, or variants thereof (e.g.
  • deletion mutants or S polypeptide fragments lacking a C-terminal transmembrane domain are also contemplated. Also contemplated are methods for treating or preventing coronavirus infections or eliciting an immune response using an S protein immunogen or variant thereof, or a combination of polypeptides (including fusion proteins).
  • Coronavirus has a positive-sense, non-segmented, single-stranded RNA genome, which encodes at least 18 viral proteins (such as non-structural proteins (NSP) 1-13, structural proteins E, M, N, S), and an RNA-dependent RNA polymerase).
  • viral proteins such as non-structural proteins (NSP) 1-13, structural proteins E, M, N, S), and an RNA-dependent RNA polymerase.
  • Coronavirus has three major surface glycoproteins (designated S, E, and M), and some coronaviruses have another surface glycoprotein referred to as hemagglutinin esterase (HE), which is not found in the SARS virus, the N (nucleocapsid) protein is a basic phosphoprotein, which is generally associated with the genome and has been reported to be antigenic (Holmes, Fields Virology, Chapter 34, 2013).
  • HE hemagglutinin esterase
  • the S (spike) protein a major antigen of coronavirus, has multiple domains: SI which is believed to be involved in receptor binding, S2 believed to mediate membrane fusion between the virus and target cell, and the receptor binding domain (RBD) believed to mediate binding to the ACE2 receptor.
  • SI which is believed to be involved in receptor binding
  • S2 believed to mediate membrane fusion between the virus and target cell
  • RBD receptor binding domain
  • the S (spike) protein may form non-covalently linked homotrimers (oligomers), which may mediate receptor binding and virus infectivity. Homotrimers of S proteins are likely necessary for presenting the correct native conformation of receptor binding domains and for eliciting a neutralizing antibody response.
  • intracellular processing of S protein is associated with significant posttranslation oligosaccharide modification.
  • the posttranslation oligosaccharide modification (glycosylation) expected by N-glycan motif analysis indicates that the S protein has as many as 23 sites for such modification.
  • C-terminal cysteine residues may also participate in protein folding and preserving the native (functional) S protein conformation.
  • the S protein of some coronaviruses e.g.
  • strains of group II and III viruses can be proteolytically processed near the center of the S protein by a trypsin-like protease in the Golgi apparatus or by extracellularly localized enzymes into to a linked polypeptide, containing an N-terminal SI and a C-terminal S2 polypeptide.
  • Some members of the type II group of coronaviruses and group I viruses may not be so processed.
  • the coronaviruses were divided into three groups on the basis of serological and genetic properties, which groups were referred to as Group 1 , Group 2, and Group 3, which are also referred to in the art and herein as Group I, Group II, and Group III (see e.g. Holmes, Fields Virology, supra; Stadler, Nat. Rev. Microbiol. (2003) 209-18; Holmes, J Clin. Invest. (2003) 111 :1605-609).
  • the coronaviruses are subdivided into Group 1, Group 2, Group 3, and SARS- CoV (SARS-associated coronavirus including SARS-CoV-2).
  • An exemplary S protein is from SARS-CoV-1 and has 1,255 amino acids (see SEQ ID NO:4), with a 12 amino acid signal sequence, the SI domain between amino acids 13-679 and the S2 domain between amino acids 680-1195.
  • the SARS-CoV-1 S protein and variants thereof that have one or more epitopes i.e. are immunogens
  • that are capable of eliciting a neutralizing e.g. IgA or IgG antibody
  • cell-mediated immune response against multiple species of sarbecovirus e.g. a pan- sarbecovirus immune response
  • compositions for use in treating or preventing coronavirus infections are included in compositions for use in treating or preventing coronavirus infections.
  • SARS-CoV-1 S protein immunogens containing one or more immunogenic epitopes
  • the SARS-CoV-1 S protein immunogen is a portion or fragment of the full-length S protein. This region also corresponds to the region of the SI domain that is believed to be involved with cell receptor binding. Accordingly, a fragment comprising the receptor binding domain (RBD) amino acids at positions 318-510 of SEQ. ID NO:4, or a portion thereof, may be immunogenic and an immune response specific for one or more epitopes within this sequence may prevent entry of the coronavirus into a target cell.
  • RBD receptor binding domain
  • identification of such immunogenic fragments of the SARS-CoV-1 S protein that do not contain glycosylation sites provides the advantage that the fragments may be made and produced in cells, such as bacteria, that are not capable of glycosylating a protein in the same manner as a mammalian cell.
  • a SARS-CoV-1 S protein immunogen includes a fragment of S protein or a S protein variant (which may be a variant of a full-length SARS-CoV-1 S protein or fragment as described herein) that retains or that has at least one epitope contained within the full-length S protein or wildtype S protein, respectively, that elicits a protective immune response against coronavirus, preferably against SARS coronavirus, including multiple species of sarbecovirus.
  • a SARS-CoV-1 S protein fragment or an S protein variant has at least one biological activity or function of a full-length or wildtype (natural) SARS-CoV-1 S protein (such as receptor binding or cell fusion activity), or has multiple S protein-specific biological activities or functions.
  • a SARS-CoV-1 S protein variant may contain an epitope that induces an immune response (for example, induces production of an antibody that specifically binds to a wildtype or full-length S polypeptide) or may have S protein receptor binding activity.
  • a SARS-CoV-1 S-protein fragment is a truncated S-protein that comprises an amino acid set forth at positions 1-1195 of SEQ ID NO:4.
  • SARS-CoV-1 S protein immunogenic fragments also include smaller portions or fragments of the aforementioned amino acid fragments of an S protein.
  • a SARS-CoV-1 S protein fragment that comprises an epitope that stimulates, induces, or elicits an immune response may comprise a sequence of consecutive amino acids ranging from any number of amino acids between 8 amino acids and 150 amino acids (e.g. 8, 10, 12, 15, 18, 20, 25, 30, 35, 40, 50 or more amino acids) of SEQ. ID NO:4.
  • a SARS-CoV-1 S polypeptide variant has at least 85% to 100% amino acid sequence identity (that is, at least 85%, 90%, 95% or 99% sequence identity) to the amino acid sequence of the full length S protein as set forth in SEQ ID NO:4 (which is from SARS-CoV-4; SEQ ID NO:3 is the nucleic acid sequence that encodes the amino acid sequence of SEQ ID NO:4).
  • Such SARS- CoV-1 S protein variants and fragments retain at least one S protein-specific biological activity or function, such as (1) the capability to elicit a protective immune response (that is, the S polypeptide variant contains an epitope that induces or elicits a protective immune response), for example, a neutralizing response and/or a cell-mediated immune response against multiple species of sarbecovirus, such as SARS-CoV-1 and SARS-CoV-2; (2) the capability to mediate viral infection via receptor binding; and (3) the capability to mediate membrane fusion between a virion and the host cell. Additional examples of full-length SARS-CoV-1 S protein sequences are available in the art.
  • An exemplary S protein is from SARS-CoV-2 and has 1,255 amino acids (see SEQ ID NO:2), with a 12 amino acid signal sequence, the SI domain between amino acids 12-672 and the S2 domain between amino acids 673-1192.
  • SARS-CoV-2 S proteins and variants thereof that have one or more epitopes (i.e. are immunogens) and that are capable of eliciting a neutralizing (e.g. IgA or IgG antibody) or cell-mediated immune response are included in compositions for use in treating or preventing sabrecovirus infections.
  • the SARS-CoV-2 S protein immunogen is a portion or fragment of the full-length S protein.
  • a portion of the SARS-CoV-2 S protein immunogen that includes amino acids at positions 417-560 of SEQ. ID NO:2 does not contain an N-glycan substitution site and is a hydrophilic region. This region also corresponds to the region of the SI domain that is believed to be involved with cell receptor binding.
  • a fragment comprising amino acids at positions 417-560 of SEQ ID NO:2 corresponding to the receptor binding domain (RBD), or a portion thereof may be immunogenic and an immune response specific for one or more epitopes within this sequence may prevent entry of the coronavirus into a target cell.
  • identification of such immunogenic fragments of the SARS-CoV-2 S protein that do not contain glycosylation sites provides the advantage that the fragments may be made and produced in cells, such as bacteria, that are not capable of glycosylating a protein in the same manner as a mammalian cell.
  • a SARS-CoV-2 S protein immunogen includes a fragment of S protein or a S protein variant (which may be a variant of a full-length SARS-CoV-2 S protein or fragment as described herein) that retains or that has at least one epitope contained within the full-length S protein or wildtype SARS-CoV-2 S protein, respectively, that elicits a protective immune response against multiple species of sarbecovirus.
  • An SARS-CoV-2 S protein fragment or an S protein variant has at least one biological activity or function of a full-length or wildtype (natural) S protein (such as receptor binding or cell fusion activity), or has multiple S protein-specific biological activities or functions.
  • a SARS-CoV-2 S protein variant may contain an epitope that induces an immune response (for example, induces production of an antibody that specifically binds to a SARS-CoV-2 wildtype or full-length S protein) or may have S protein receptor binding activity.
  • an S-protein fragment is a truncated SARS-CoV-2 S protein that comprises an amino acid set forth at positions 1-1200 of SEQ ID NO:2. The portion of the SARS-CoV-2 S protein that is deleted is the transmembrane region.
  • SARS-CoV-2 S protein immunogenic fragments also include smaller portions or fragments of the aforementioned amino acid fragments of a SARS-CoV-2 S protein.
  • a SARS-CoV-2 S protein fragment that comprises an epitope that stimulates, induces, or elicits an immune response may comprise a sequence of consecutive amino acids ranging from any number of amino acids between 8 amino acids and 150 amino acids (e.g. 8, 10, 12, 15, 18, 20, 25, 30, 35, 40, 50 or more amino acids) of SEQ ID NO:2.
  • a SARS-CoV-2 S polypeptide variant has at least 85% to 100% amino acid sequence identity (that is, at least 85%, 90%, 95% or 99% sequence identity) to the amino acid sequence of the full length S protein as set forth in SEQ.
  • SARS-CoV-2 (which is from SARS-CoV-2; SEQ ID NO: 1 is the nucleic acid sequence that encodes the amino acid sequence of SEQ ID NO:2).
  • S protein variants and fragments retain at least one SARS-CoV-2 S protein-specific biological activity or function, such as (1) the capability to elicit a protective immune response (that is, the S polypeptide variant contains an epitope that induces or elicits a protective immune response), for example, a neutralizing response and/or a cell- mediated immune response against coronavirus, such as SARS-CoV-2; (2) the capability to mediate viral infection via receptor binding; and (3) the capability to mediate membrane fusion between a virion and the host cell.
  • a protective immune response that is, the S polypeptide variant contains an epitope that induces or elicits a protective immune response
  • coronavirus such as SARS-CoV-2
  • SARS-CoV-2 the capability to mediate viral infection via receptor binding
  • S protein immunogens, fragments, and variants thereof described herein contain a haptenized epitope that elicits or induces an immune response, preferably a protective immune response, which may be a humoral response and/or a cell-mediated immune response.
  • a protective immune response may be manifested by at least one of the following: preventing infection of a host by a coronavirus; modifying or limiting the infection; aiding, improving, enhancing, or stimulating recovery of the host from infection; and generating immunological memory that will prevent or limit a subsequent infection by a coronavirus.
  • a humoral response may include production of antibodies that neutralize infectivity, lyse the virus and/or infected cell, facilitate removal of the virus by host cells (for example, facilitate phagocytosis), and bind to and facilitate removal of viral antigenic material.
  • a humoral response may also include a mucosal response, which comprises eliciting or inducing a specific mucosal IgA response.
  • Induction of an immune response in a subject or host (human or non- human animal) by a haptenized S protein, fragment, or variant described herein may be determined and characterized by methods described herein and routinely practiced in the art. These methods include in vivo assays, such as animal immunization studies (e.g. using a rabbit, mouse or rhesus macaque model), and any one of a number of in vitro assays, such as immunochemistry methods for detection and analysis of antibodies, including Western immunoblot analysis, ELISA, immunoprecipitation, radioimmunoassay, and the like, and combinations thereof.
  • animal models may be used for determining the capability of a coronavirus antigen to elicit and induce an immune response that is protective in animals, which may be determined by endpoints relevant to the particular model.
  • neutralization assays such as a plaque reduction assay or an assay that measures cytopathic effect (CPE) or any other neutralization assay practiced by persons skilled in the art
  • CPE cytopathic effect
  • the haptenized S protein immunogens are provided in an isolated form, and in certain embodiments, are purified to homogeneity.
  • isolated means that the polypeptide is removed from its original or natural environment.
  • the invention provides a method of immunizing a human subject against a Coronavirus, the method comprising administering an effective amount of an haptenized S protein from Coronavirus, including SARS-CoV-2 Coronavirus.
  • the S protein is from SARS-CoV- 2 Coronavirus and has the amino acid sequence as set forth in SEQ ID NO:2.
  • the haptenized S protein is administered in a first dose followed by a second dose at three to four weeks weeks.
  • the method further comprising at least one booster injection of the haptenized S protein about six months after the first injection.
  • booster injections continue every six months or until an immunogenic response against a sarbecovirus occurs in the human subject.
  • the effective amount of haptenized S protein is administered until the delayed type hypersensitivity diagnostic test is positive, or a neutralizing antibody response is detected (e.g. anti-S protein antibodies are detected in the blood or serum of the human subject).
  • the invention provides a method of producing an immunogenic response in a human subject against a Coronavirus, including SARS-CoV-2 Coronavirus, the method comprising administering an effective amount of an haptenized S protein from Coronavirus, including SARS-CoV-2 Coronavirus.
  • the S protein is from SARS-CoV-2 Coronavirus and has the amino acid sequence as set forth in SEQ. ID NO:2.
  • the invention encompasses a method of immunizing a human subject against multiple species of sarbecovirus comprising: (a) immunizing a human subject against SARS-CoV- 1, and (b) immunizing a human subject against SARS-CoV-2, wherein the human subject has never been infected with, nor immunized against, SARS-CoV-1 and SARS-CoV-2.
  • the immunizing comprises: (i) administration to the human subject of an immunogenic composition comprising: (a) an S protein or fragment thereof from SARS-CoV-1 and at least one pharmaceutically acceptable carrier, or (b) a polynucleotide encoding an S protein or fragment thereof from SARS-CoV-1 and at least one pharmaceutically acceptable carrier; and (ii) administration to the human subject of an immunogenic composition comprising: (a) an S protein or fragment thereof from SARS-CoV-2 and at least one pharmaceutically acceptable carrier, or (b) a polynucleotide encoding an S protein or fragment thereof from SARS-CoV-2 and at least one pharmaceutically acceptable carrier.
  • the immunizing against SARS-CoV-1 occurs first, followed by a second immunizing against SARS-CoV-2. In some embodiments, the immunizing against SARS-CoV-2 occurs first, followed by a second immunizing against SARS-CoV-1.
  • the invention encompasses a method of immunizing a human subject against multiple species of sarbecovirus comprising: (i) administering to the human subject an immunogenic composition comprising: (a) an S protein or fragment thereof from SARS-CoV-1 and at least one pharmaceutically acceptable carrier, or (b) a polynucleotide or fragment thereof encoding an S protein from SARS-CoV-1, wherein the human subject has already been immunized against SARS-CoV-2 and/or been infected with SARS-CoV-2.
  • the S protein fragment comprises the receptor binding domain (RBD) of the S protein while in other embodiments, the S protein fragment comprises the SI domain of the S protein.
  • the S protein or fragment thereof is haptenized.
  • the second immunization occurs at least three weeks after the first immunization.
  • the method further comprises administering at least one booster injection of the immunogenic composition about six months after the first injection.
  • the immunogenic composition is administered until a cross-reactive neutralizing antibody response against multiple species of sarbecovirus is detected in the human subject.
  • the human subject has been previously infected with SARS-CoV-2 and recovered from Covid-19.
  • the human subject has previously been immunized against SARS-CoV-2.
  • the SARS-CoV-2 immunization is an mRNA vaccine, inactivated virus vaccine, viral vector vaccine, or protein subunit vaccine.
  • the mRNA vaccine is BNT162b2 or mRNA-
  • viral vector vaccine is Ad26.COV2.S COVID-19 or ChAdOxl.
  • the SARS-CoV-1 immunogenic composition is administered first, followed by administration of the SARS-CoV-2 immunogenic composition.
  • the SARS-CoV-2 immunogenic composition is administered first, followed by administration of the SARS- CoV-1 immunogenic composition.
  • the subject has not been infected with any species of sarbecovirus.
  • the SARS-CoV-1 S protein immunogen comprises an amino acid sequence that is identical to, or at least 85% identical to (which includes at least 90% or 95% or any percent in between 85% and 100%) SEQ ID NO: 4 and wherein the SARS-CoV-1 S protein immunogen has an epitope that elicits a protective immune response, which is a humoral immune response (including, for example, a mucosal IgA, systemic IgA, IgG, IgM response) and/or a cell-mediated immune response, and pharmaceutically acceptable carrier, diluent, or excipient.
  • a protective immune response which is a humoral immune response (including, for example, a mucosal IgA, systemic IgA, IgG, IgM response) and/or a cell-mediated immune response, and pharmaceutically acceptable carrier, diluent, or excipient.
  • the SARS-CoV-1 S protein immunogen composition is administered at a dose sufficient to elicit a pan-sarbecovirus immune response specific for multiple species of sarbecovirus S protein immunogen, immunogens or variants thereof.
  • an infection being prevented or treated may be caused by any type of sarbecovirus species, or a combination thereof.
  • the SARS-CoV-1 S protein immunogen is haptenized as described herein.
  • the SARS-CoV-2 S protein immunogen comprises an amino acid sequence that is identical to, or at least 85% identical to (which includes at least 90% or 95% or any percent in between 85% and 100%) SEQ. ID NO: 2 and wherein the SARS-CoV-2 S protein immunogen has an epitope that elicits a protective immune response, which is a humoral immune response (including, for example, a mucosal IgA, systemic IgA, IgG, IgM response) and/or a cell-mediated immune response, and pharmaceutically acceptable carrier, diluent, or excipient.
  • a protective immune response which is a humoral immune response (including, for example, a mucosal IgA, systemic IgA, IgG, IgM response) and/or a cell-mediated immune response, and pharmaceutically acceptable carrier, diluent, or excipient.
  • the SARS-CoV-2 S protein immunogen composition is administered at a dose sufficient to elicit a pan-sarbecovirus immune response specific for multiple species of soopcovirus S protein immunogen or immunogens or variants thereof.
  • an infection being prevented or treated may be caused by any type of sarbecovirus species, or a combination thereof.
  • the SARS-CoV-2 S protein immunogen is haptenized as described herein.
  • a human subject or host suitable for treatment with a coronavirus immunogen composition or formulation may be identified by well-established indicators of risk for developing a disease such as Covid-19 or by well-established hallmarks of an existing sarbecovirus disease.
  • indicators of an infection include fever, dry cough, dyspnea (shortness of breath), headache, hypoxemia (low blood oxygen concentration), lymphopenia (reduced lymphocyte numbers), mildly elevated aminotransferase levels (indicating liver damage), microorganism positive cultures, inflammation, and the like.
  • Infections that may be treated or prevented with a coronavirus S protein immunogen vaccine as described herein include those caused by or due to coronavirus, whether the infection is primary, secondary or opportunistic.
  • sarbecovirus examples include any species, strain, antigenic variant, and the like, of these viruses, including SARS coronavirus such as SARS-CoV-1, Bat CoV WIV17, Bat CoV RsSHC0147, SARS-CoV-2, SARS-CoV-2 Alpha variant, SARS-CoV-2 Beta variant, SARS-CoV-2 Delta variant, Bat CoV RaTG135, Pangolin CoV GD-16, Pangolin GX-P5L6.
  • SARS coronavirus such as SARS-CoV-1, Bat CoV WIV17, Bat CoV RsSHC0147
  • SARS-CoV-2 SARS-CoV-2 Alpha variant
  • SARS-CoV-2 Beta variant SARS-CoV-2 Delta variant
  • Bat CoV RaTG135, Pangolin CoV GD-16, Pangolin GX-P5L6 examples include any species, strain, antigenic variant, and the like, of these viruses, including SARS coronavirus such as SARS-CoV-1, Bat CoV WIV17
  • the immunogenic compositions that contain one or more haptenized coronavirus S protein immunogens of the invention may be in any form that allows for the composition to be administered to a subject, such as a human or non-human animal.
  • a haptenized S protein immunogen may be prepared and administered as a liquid solution or prepared as a solid form (e.g. lyophilized), which may be administered in solid form, or resuspended in a solution in conjunction with administration.
  • the hybrid polypeptide composition is prepared or formulated to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a subject or patient or to be bioavailable via slow release.
  • compositions that will be administered to a subject or patient take the form of one or more dosage units; for example, a tablet may be a single dosage unit, and a container of one or more compounds of the invention in aerosol form may hold a plurality of dosage units.
  • any of the aforementioned immunogenic compositions or vaccines comprising a haptenized S protein immunogen described herein are in a container, preferably in a sterile container.
  • the immunogenic composition or vaccine is administered nasally, wherein a haptenized coronavirus S protein immunogen can be taken up by cells, such as cells located in the nasal- associated lymphoid tissue.
  • a haptenized coronavirus S protein immunogen can be taken up by cells, such as cells located in the nasal- associated lymphoid tissue.
  • Other typical routes of administration include, without limitation, parenteral, transdermal/transmucosal, nasal, and inhalation.
  • parenteral describes administration routes that bypass the gastrointestinal tract, including intraarterial, intradermal, intramuscular, intranasal, intraocular, intraperitoneal, intravenous, subcutaneous, submucosal, and intravaginal injection or infusion techniques.
  • transdermal/transmucosal is a route of administration in which the immunogenic composition is administered through or by way of the skin, including topical.
  • nasal and “inhalation” encompass techniques of administration in which an immunogenic composition is introduced into the pulmonary tree, including intrapulmonary or transpulmonary.
  • the compositions of the present invention are administered nasally.
  • the immunogenic composition or vaccine contains an amount of haptenized S protein from about 30 pg to about 300 pg per dose. In some embodiments, the amount of haptenized coronavirus S protein administered per dose is from about 1 pg to about 240 pg.
  • the amount of haptenized S protein administered per dose is from about 1 pg, 3 pg, 5 pg, 10 pg, 25 pg, 30 pg, 40 pg, 50 pg, 60 pg, 70 pg, 80 pg, 90 pg, 100 pg, 110 pg, 120 pg, 130 pg, 140 pg, 150 pg, 160 pg, 170 pg, 180 pg, 190 pg, 200 pg, 210 pg, 220 pg, 230 pg, 240 pg, 250 pg or more.
  • the amount of haptenized coronavirus S protein varies depending on dosing schedule. For example, an initial dose may be the same as, lower or higher than any subsequent immunization dose, including a booster immunization dose.
  • the amount of haptenized S protein administered per dose to any human subject can be adjusted according to the age, weight and physical condition of the human subject.
  • the invention provides an immunogenic composition or vaccine comprising an haptenized S protein for injection further comprising a vaccine adjuvant.
  • a composition that is useful as an immunogenic composition for treating and/or preventing a coronavirus infection contains at least one coronavirus antigen (immunogen) as described herein capable of eliciting an immune response and protollin or proteosome adjuvant (see e.g. U.S. Patent No. 5,726,292).
  • an adjuvant may enhance or improve the immunogenicity of an immunogen (that is, act as an immunostimulant), and many antigens are poorly immunogenic unless combined or admixed or mixed with an adjuvant.
  • a variety of sources can be used as a source of antigen, such as live attenuated virus, killed virus, split antigen preparations, subunit antigens, recombinant or synthetic viral antigens, and combinations thereof.
  • the antigens can be combined with a potent immunostimulant or adjuvant.
  • Other exemplary adjuvants include alum (aluminum hydroxide, REHYDRAGEL); aluminum phosphate; virosomes; liposomes with and without Lipid A; or other oil in water emulsions type adjuvants such as MF59 (Novartis), also such as nanoemulsions (see e.g. U.S. Patent No. 5,716,637) or submicron emulsions (see e.g. U.S. Patent No. 5,961,970); and Freund's complete and incomplete adjuvant.
  • a proteosome-based adjuvant i.e. protollin or proteosome
  • a proteosome-based adjuvant can be used in vaccine compositions or formulations that may include any one or more of a variety of coronavirus antigen (immunogen) sources as described herein.
  • Proteosomes are comprised of outer membrane proteins (OMP) from Neisseria species typically, but can be derived from other Gram-negative bacteria (see e.g. U.S. Patent No. 5,726,292).
  • Proteosomes have the capability to auto-assemble into vesicle or vesicle-like OMP clusters of 20-800 nm, and to noncovalently incorporate, coordinate, associate, or otherwise cooperate with protein antigens, particularly antigens that have a hydrophobic moiety.
  • Proteosomes are hydrophobic, safe for human use, and comparable in size to certain viruses.
  • mixing proteosomes with an antigen such as a protein antigen provides a composition comprising non-covalent association or coordination between the antigen and Proteosomes, which association or coordination forms when solubilizing detergent is selectively removed or reduced in concentration, for example, by dialysis.
  • proteosomes are from Neisseria species, and from Neisseria meningitidis.
  • proteosomes may be an adjuvant and an antigen delivery composition.
  • an immunogenic composition comprises one or more coronavirus antigens and an adjuvant, wherein the adjuvant comprises Projuvant or Protollin.
  • a coronavirus antigen may be isolated from the virus particles, a cell infected by the coronavirus, or from a recombinant source.
  • an immunogenic composition further comprises a second immunostimulant, such as a liposaccharide.
  • the adjuvant may be prepared to include an additional immunostimulant.
  • the projuvant may be mixed with a liposaccharide to provide an OMP-LPS adjuvant.
  • the OMP-LPS (protollin) adjuvant can be comprised of two components.
  • the first component includes an outer membrane protein preparation of proteosomes (i.e.
  • Projuvant prepared from Gram-negative bacteria, such as Neisseria meningitidis, and the second component includes a preparation of liposaccharide. It is also contemplated that the second component may include lipids, glycolipids, glycoproteins, small molecules or the like, and combinations thereof. As described herein, the two components of an OMP-LPS adjuvant may be combined (admixed or formulated) at specific initial ratios to optimize interaction between the components, resulting in stable association and formulation of the components for use in the preparation of an immunogenic composition. The process generally involves the mixing of components in a selected detergent solution (e.g.
  • an immunogenic composition comprises one or more coronavirus haptenized S protein antigens and an adjuvant, wherein the adjuvant comprises a projuvant (i.e. proteosome) and liposaccharide.
  • a projuvant i.e. proteosome
  • the final liposaccharide content by weight as a percentage of the total proteosome protein can be in a range from about 1% to about 500%, also in range from about 10% to about 200%, or in a range from about 30% to about 150%.
  • Another embodiment includes an adjuvant wherein the proteosomes are prepared from Neisseria meningitidis and the liposaccharide is prepared from Shigella flexneri or Plesiomonas shigelloides, and the final liposaccharide content is between 50% to 150% of the total Proteosome protein by weight.
  • proteosomes are prepared with endogenous lipooligosaccharide (LOS) content ranging from about 0.5% up to about 5% of total OMP.
  • proteosomes have endogenous liposaccharide in a range from about 12% to about 25%, and in still another embodiment the endogenous liposaccharide is between about 15% and about 20% of total OMP.
  • the instant disclosure also provides an immunogenic composition containing liposaccharide derived from any Gram-negative bacterial species, which may be from the same Gram-negative bacterial species that is the source of proteosomes or may be from a different bacterial species.
  • the proteosome or protollin to coronavirus antigen ratio in the immunogenic composition is greater than 1:1, greater than 2:1, greater than 3:1 or greater than 4:1. In other embodiments, proteosome or protollin to haptenized coronavirus S protein antigen ratio in the immunogenic composition is about 1:1, 2:1, 3:1 or 4:1. The ratio can be 8:1 or higher. In other embodiments, the ratio of proteosome or protollin to haptenized coronavirus S protein antigen of the immunogenic composition ranges from about 1:1 to about 1:500, and is at least 1:5, at least 1:10, at least 1:20, at least 1:50, or at least 1:100, or at least 1:200.
  • An advantage of protollin to haptenized coronavirus S protein antigen ratios ranging from 1:2 to 1:200 is that the amount of proteosome-based adjuvant can be reduced dramatically with no significant effect on the ability of a coronavirus antigen to elicit an immune response.
  • an immunogenic composition comprises one or more haptenized coronavirus S protein immunogens combined (admixed or formulated) with proteosome or protollin, wherein the S protein immunogen comprises an amino acid sequence that is identical to, or at least 85% identical (which includes at least 90% or 95% or any percent in between 85% and 100%) to SEQ ID NO:2 or fragment thereof and wherein the haptenized S protein immunogen or fragment thereof has an epitope that elicits a protective immune response against coronavirus infection.
  • An exemplary haptenized S protein immunogen comprises an amino acid sequence as set forth in SEQ. ID NO:2 or 4, or consisting of SEQ ID NO:2 or 4.
  • an S protein immunogen is a fragment of SEQ ID NO:2 or 4, which fragment comprises an amino acid sequence that is identical to, or at least 85% identical (which includes at least 90% or 95% or any percent in between 85% and 100%) to an amino acid selected from SEQ ID NO:2 or 4.
  • any haptenized S protein immunogen as described herein can be combined (admixed or formulated) in an immunogenic composition with a liposome.
  • liposomes that contain one or more sarbecovirus immunogens further comprise Deinococcus radiodurans lipids or a- galactosylphosphotidylglycerolalkylamine.
  • the addition of such lipids in a liposome can enhance the efficacy of a sabrecovirus vaccine composition by increasing protective immunity.
  • Haptenized sabrecovirus S protein immunogens of the present invention may further include a covalently attached hydrophobic portion.
  • a hydrophobic portion may be, for example, an amino acid sequence or a lipid, as disclosed in U.S. Patent No. 5,726,292.
  • Naturally occurring sabrecovirus S protein and a recombinantly expressed S protein having the sequence set forth in SEQ ID NO:2 or 4 contains a hydrophobic transmembrane domain (from about amino acid 1195 to about 1240 of SEQ ID NO:2), which may function as a hydrophobic portion with an S protein immunogen fragment.
  • the haptenized S protein immunogen may further contain a second amino acid sequence to form a fusion protein, wherein the second amino acid sequence is a tag, carrier, or enzyme, as described herein.
  • immunogenic compositions may comprise (projuvant or protollin), or further comprise components (e.g. receptor ligands) capable of stimulating a host immune response by interacting with certain receptors (e.g. Toll-like receptors or "TLR") produced by one or more host cells of a vaccine recipient.
  • compositions comprising immunogenic epitopes of a sabrecovirus protein may contain polypeptide epitopes capable of interacting with Toll-like receptors, thereby promoting an innate immune response, which may or may not evoke a subsequent adaptive immune response.
  • An innate immune response is a nonspecific protective immune response that is not a specific antigen-dependent or antibody-dependent response (that is, does not involve clonal expansion of T cells and/or B cells) and may be elicited by any one of numerous antigens, immunogens, or sabrecovirus described herein.
  • An immunostimulatory composition described herein comprises proteosomes and liposaccharide (protollin), either one of which or both may elicit a nonspecific protective response.
  • proteosomes and liposaccharide protollin
  • one or more components of vaccine compositions or formulations disclosed herein may interact with Toll-like receptors associated with an innate or adaptive immune response of a vaccine recipient.
  • TLR8 and TLR10 Certain outer membrane proteins of Neisseria meningitidis, for example OMP2 (also referred to as PorB), interact with TLR2, while LPS of most but not all Gram-negative bacteria interacts with TLR4. Accordingly, one activity of vaccine compositions or formulations described herein, which may contribute to a biological effect, includes activation of one or both of TLR2 and TLR4.
  • An exemplary adjuvant is the TLR4 agonist MPL (3- O-desacyl-4'-monophosphoryl lipid A).
  • TLR Activation of other TLR may serve a similar function or further enhance the qualitative or quantitative profile of cytokines expressed, and may be associated with a host Thl/Th2 immune response. It is also contemplated that TLR ligands other than LPS and PorB may be used alone or in combination to activate TLR2 or TLR4. The qualitative or quantitative activation of one or more TLR is expected to elicit, effect, or influence a relative stimulation (balanced or unbalanced) of a Thl or Th2 immune response, with or without a concomitant humoral antibody response.
  • Ligands interacting with TLR other than TLR2 and TLR4 may also be used in vaccine compositions described herein. Such vaccine components may, alone or in combination, be used to influence the development of a host immune response sufficient to treat or protect from virus infection, as set forth herein.
  • haptenized S protein immunogen may further comprise adjuvants, such as Bacillus Calmette-Guerin (BCG), cytokines (for non-limiting example, granulocyte-macrophage colonystimulating (GM-CSF)), saponins (for non limiting example Q.S-21), squalenes (for non-limiting example M59), bisphosphonate, aluminum gels or aluminum salts (for non-limiting example, amorphous aluminum hydroxyphosphate sulfate (AAHS), aluminum hydroxide, aluminum phosphate, potassium aluminum sulfate (Alum)), AS01 (a liposome-based vaccine adjuvant system containing 3-O-desacyl-4'- monophosphoryl lipid A (MPL) and the saponin Q.S-21), AS03 (a-tocopherol, squalene and polysorbate 80 in an oil-in-water emulsion
  • adjuvants such as Bacillus Calmette-Guerin (BCG), cytokines
  • a haptenized S protein immunogenic composition or vaccine may further comprise preservatives known to the art, including without limitation, formaldehyde, antibiotics, monosodium glutamate, 2- phenoxyethanol, phenol, and benzethonium chloride.
  • a haptenized S protein immunogenic composition or vaccine may further comprise sterile water for injection, balanced salt solutions for injections.
  • DNP modification Modification of the prepared cells with DNP or another hapten may be performed by known methods, e.g. by the method of Miller, J. Immunol. (1976) 117:151 which involves a thirty minute incubation of S protein with DNFB under sterile conditions, followed by washing with sterile saline or HBSS-HSA.
  • DNFB Stemmett-B
  • HBSS-HSA sterile saline
  • DNFB Sigma Chemical
  • PBS PBS
  • the solution is stirred overnight in a 37°C water bath.
  • the shelf life of the solution is about four weeks.
  • the S protein is suspended in Hanks balanced salt solution. About 0.1 ml DNFB solution is added to each sample of S protein and incubated for about thirty minutes at room temperature.
  • SA modification Modification of the S protein with SA may be performed by known methods. For example, in one embodiment, sulfanilic acid (SA) is converted to a diazonium salt by adding a saturating amount of sodium nitrite. Ice-cold, sterile filtered (0.2 pm), 10% sodium nitrite solution is added, dropwise, to a SA solution of 100 mg of anhydrous SA dissolved in 10 ml of 0.1N NCI until saturation. (The saturation point corresponds approximately to a final concentration of a sulfanilic acid diazonium salt of about 40 mM).
  • the SA diazonium salt solution is then sterile filtered (0.2 pm membrane), and diluted 1:8 (v/v) in HBSS (without HSA). If needed, the pH is adjusted to 7.2 by dropwise addition of IN NaOH.
  • the SA diazonium salt-HBSS solution is then sterilized by filtration (0.2 pm membrane). S protein is suspended in diazonium salt-HBSS solution. The mixture is incubated for five minutes at room temperature. After the five minute incubation period, the haptenization reaction is stopped by the addition of 0.5 ml of a 25% HSA-HBSS solution to the mixture.
  • a human sarbecovirus vaccine consisting of recombinant S protein from SARS-CoV-2 modified with the hapten, dinitrophenyl (DNP) was prepared as per Example 1.
  • a human sarbecovirus vaccine consisting of the recombinant S protein from SARS-CoV-2 modified with hapten, DNP was prepared as per Example 1.
  • a phase I trial of the haptenized vaccine in patients is conducted, testing four dosage levels.
  • the major endpoints are the presence of a neutralizing antibody response to DNP-modified, SA-modified, and unmodified S protein from both SARS-CoV-1 and SARS-CoV-2.
  • the level of neutralizing antibodies to SARS-CoV-1 and SARS-CoV-2 is assessed over time.
  • a phase II trial using the lowest dose that is found to be immunologically effective in the phase I trial is conducted.
  • the following preparation is representative of the process used to prepare the haptenized S- Protein.
  • the scale of the previous prep is given parenthetically. Filtration steps were added to produce a sterile product.
  • Water should be low endotoxin water (nuclease free or WFI grade).
  • CF-1 Mice (Charles River) were administered up to 10 pg of BVX-0320 by subcutaneous injection. Mortality was an assessment of toxicity. All animals survived to the scheduled necropsy. There were no unusual BVX-0320-related clinical/veterinary observations. Additional clinical observations were within the range of normal findings for group-housed animals of this age, sex, and species, or were procedure-related and were not considered to be related to test article administration.
  • Body Weight and Body Weight Gains There were no BVX-0320-related body weight effects. Additional minor fluctuations among mean and individual body weight were considered sporadic, consistent with biologic variation, and/or negligible in magnitude and not related to test article administration. There were no BVX-0320-related food consumption effects. Minor fluctuations among mean and individual cage food consumption were considered sporadic, consistent with biologic variation, and/or negligible in magnitude and not related to test article administration. There were no unusual gross pathology findings.
  • Example 6 Immunology Study of BVX-0320 by Subcutaneous Injection in CF-1 Mice 1
  • CF-1 Mice (Charles River) were administered 0.3, 1, 3 or lOpg of BVX-0320 by subcutaneous injection followed by a second injection and endpoints were obtained at six weeks (see Figs. 1-2).
  • 1 pg group one mouse had baseline antibody titer 1:5.
  • 3 pg group the titre obtained was 1:120,000.

Abstract

In some embodiments, immunization methods are disclosed which encompass a pansarbecovirus vaccine which induces a broadly cross-reactive neutralizing antibody response to multiple species of sarbecovirus.

Description

METHODS OF IMMUNIZATION AGAINST CORONAVIRUS
Sequence Listing Submission via EFS-Web
A computer readable text file, entitled "SequenceListing.txt" created on October 6, 2021 with a file size of 32,216 bytes contains the sequence listing for this application and is hereby incorporated by reference in its entirety.
FIELD OF THE INVENTION
[001] The invention described herein relates generally to method of immunizing a human subject against multiple species of sarbecovirus.
BACKGROUND OF THE INVENTION
[002] Coronaviruses are plus-strand RNA viruses that cause disease in animals and humans. A novel zoonotic coronavirus outbreak started in Wuhan, China in 2019. This pandemic disease has now been defined as novel coronavirus disease 2019 (Covid-19), and is sustained by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The Covid-19 pandemic that started in December 2019 caused by SARS-CoV-2 shares an overall genome sequence identity of approximately 80% with SARS-CoV-1. This latter coronavirus was responsible for the SARS outbreak in 2002-2003, which included more than 8000 infections and more than 700 deaths worldwide. SARS-CoV-1 and SARS-CoV-2 belong to the species of SARS-related coronavirus (subgenus sarbecovirus, genus betacoronavirus). Antigenically, the two coronaviruses are placed in two distinct phylogenetic clades; convalescent serum specimens from patients with SARS or Covid-19 lack cross-neutralization, despite the majority of survivors of SARS-CoV-1 infection continuing to have detectable neutralizing antibodies against the homologous SARS-CoV-1 virus seventeen years after infection (Ten, New. Engl. J. Med. (2021) 10.1056/NEJMoa2108453).
[003] A need exists for alternative prophylactic strategies or therapies to treat or prevent coronavirus infections, such as those found in humans (e.g. sarbecovirus such as SARS-CoV-1 infections or SARS-CoV- 2 infections resulting severe acute respiratory syndrome). For example, a need exists for identifying and developing immunogenic and vaccine compositions against sarbecovirus infections that can elicit a broadly cross-reactive neutralizing antibody immune response against multiple species of sarbecovirus. Furthermore, immunogenic compositions and vaccine formulations are needed that can be delivered directly to or in close proximity to the site of infection to maximize therapeutic effectiveness. [004] Haptenized proteins have been widely used to provide defined epitopes for the measurement of antibody titers and affinities. While some protein-haptenization is thought to do little more than create epitopes for B cell recognition, others have shown that certain haptenized proteins can induce adaptive immune responses under conditions where native proteins fail to induce such responses; thus, haptenization does more than simply create epitopes for antigen receptor recognition (Palm, Proc. Natl. Acad. Sci. USA (2000) 106:4782). The present invention meets such needs and further provides other related advantages. The present invention provides for cross-reactive immunogens for sarbecovirus and meets the need for immunogenic compositions and vaccine formulations that can be delivered directly or in close proximity to the site of infection to maximize a protective immune response in a human subject.
SUMMARY OF THE INVENTION
[005] Embodiments disclosed herein are immunogenic compositions or vaccines comprising haptenized S proteins from coronavirus, including severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and methods of immunizing a human subject with the immunogenic compositions or vaccines.
BRIEF DESCRIPTION OF THE DRAWINGS
[006] The foregoing summary, as well as the following detailed description of the invention, will be better understood when read in conjunction with the appended drawings.
[007] Figure 1 shows the anti-spike protein antibody response following subcutaneous administration of BVX-0320 in CF-1 mice.
[008] Figure 2 shows the T cell (gamma interferon) response to BVX-0320 (X = spleen samples wth >7,000 spots/106 T cells).
DETAILED DESCRIPTION OF THE INVENTION
[009] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entireties. Definitions
[0010] An "immunogenic composition" or "vaccine" as used herein refers to any one or more compounds or agents or immunogens capable of priming, potentiating, activating, eliciting, stimulating, augmenting, boosting, amplifying, or enhancing an adaptive (specific) immune response, which may be cellular (T cell) or humoral (B cell), or a combination thereof. Preferably, the adaptive immune response is protective, which may include neutralization of a coronavirus (decreasing or eliminating virus infectivity). A representative example of an immunogen is a viral antigen (such as one or more coronavirus antigens). In the present description, any concentration range, percentage range, ratio range, or integer range is understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer, etc.), unless otherwise indicated.
[0011] As used herein, "about" or "comprising essentially of" means ± 10%. As used herein, the use of an indefinite article, such as "a" or "an," should be understood to refer to the singular and the plural of a noun or noun phrase (i.e. meaning "one or more" or "at least one" of the enumerated elements or components). The use of the alternative (e.g. "or") should be understood to mean either one, both or any combination thereof of the alternatives. In addition, it should be understood that the individual compounds, or groups of compounds, derived from the various combinations of the sequences, structures, and substituents described herein, are disclosed by the present application to the same extent as if each compound or group of compounds was set forth individually. Thus, selection of particular sequences, structures, or substituents is within the scope of the present invention.
[0012] The term "effective amount" or "therapeutically effective amount" refers to that amount of a compound or combination of compounds as described herein that is sufficient to effect the intended application including, but not limited to, disease treatment. A therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated (e.g. the weight, age and gender of the subject), the severity of the disease condition, the manner of administration, etc. which can readily be determined by one of ordinary skill in the art. The term also applies to a dose that will induce a particular response in target cells (e.g. the reduction of platelet adhesion and/or cell migration). The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether the compound is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which the compound is carried. [0013] The term "immunogenicity" means the ability of an S protein immunogen to evoke an immune response directed to the coronavirus, including broadly cross-reactive neutralizing antibody-dependent immune responses against multiple species of sarbecovirus. Whether a S protein preparation is immunogenic can be tested by, for instance, a DTH-assay or an in vivo assay in an experimental animal model.
[0014] A "therapeutic effect" as that term is used herein, encompasses a therapeutic benefit and/or a prophylactic benefit. A prophylactic effect includes delaying or eliminating the appearance of a sarbecovirus infection, delaying or eliminating the onset of symptoms of a sarbecovirus infection, slowing, halting, or reversing the progression of a sarbecovirus infection, including reducing or eliminating any symptoms associated with SARS, or any combination thereof.
[0015] "Pharmaceutically acceptable carrier" or "pharmaceutically acceptable excipient" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and inert ingredients. The use of such pharmaceutically acceptable carriers or pharmaceutically acceptable excipients for active pharmaceutical ingredients is well known in the art. Except insofar as any conventional pharmaceutically acceptable carrier or pharmaceutically acceptable excipient is incompatible with the active pharmaceutical ingredient, its use in the therapeutic compositions of the invention is contemplated. Additional active pharmaceutical ingredients, such as other drugs, can also be incorporated into the described compositions and methods.
[0016] When ranges are used herein to describe, for example, physical or chemical properties such as molecular weight or chemical formulae, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included. Use of the term "about" when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary. The variation is typically from 0% to 15%, preferably from 0% to 10%, more preferably from 0% to 5% of the stated number or numerical range. The term "comprising" (and related terms such as "comprise" or "comprises" or "having" or "including") includes those embodiments such as, for example, an embodiment of any composition of matter, method or process that "consist of" or "consist essentially of" the described features.
[0017] As used herein, "isotype" refers to the antibody class (e.g. IgM or IgGl) that is encoded by the heavy chain constant region genes. In mammals, there are five antibody isotypes: IgA, IgD, IgG, IgM and IgE. In humans, there are four subclasses of the IgG isotype: IgGl, lgG2, lgG3 and lgG4, and two subclasses of the IgA isotype: IgAl and lgA2. [0018] The terms "sequence identity" and "sequence percent identity" in the context of two or more nucleic acids or polypeptides, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity. The percent identity can be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences. Suitable programs to determine percent sequence identity include for example the BLAST suite of programs available from the U.S. Government's National Center for Biotechnology Information BLAST web site. Comparisons between two sequences can be carried using either the BLASTN or BLASTP algorithm. BLASTN is used to compare nucleic acid sequences, while BLASTP is used to compare amino acid sequences. ALIGN, ALIGN-2 (Genentech, South San Francisco, California) or MegAlign, available from DNASTAR, are additional publicly available software programs that can be used to align sequences. One skilled in the art can determine appropriate parameters for maximal alignment by particular alignment software. In certain embodiments, the default parameters of the alignment software are used.
[0019] For the avoidance of doubt, it is intended herein that particular features (for example integers, characteristics, values, uses, diseases, formulae, compounds or groups) described in conjunction with a particular aspect, embodiment or example of the invention are to be understood as applicable to any other aspect, embodiment or example described herein unless incompatible therewith. Thus such features may be used where appropriate in conjunction with any of the definition, claims or embodiments defined herein. All of the features disclosed in this specification (including any accompanying claims, abstract and drawings), and/or all of the steps of any method or process so disclosed, may be combined in any combination, except combinations where at least some of the features and/or steps are mutually exclusive. The invention is not restricted to any details of any disclosed embodiments. The invention extends to any novel one, or novel combination, of the features disclosed in this specification (including any accompanying claims, abstract and drawings), or to any novel one, or any novel combination, of the steps of any method or process so disclosed.
Methods of Preparing Haptenized S protein
[0020] The preparation of haptenized vaccines using a single hapten reagent are known to the skilled artisan. In an embodiment, the invention provides for an haptenized S protein coronavirus vaccine, such a vaccine may be prepared comprising recombinantly producing an S protein or fragment thereof, and haptenizing with a haptenization reagent.
[0021] The step of haptenizing may be performed by modifying recombinant S protein with DNP by a 30-minute incubation with the haptenization reagent 2,4-difluoronitrobenzene (DNFB). The haptenized S protein is washed with HBSS. Preferred haptenization reagents may target the e-amino group of an amino acid.
[0022] In some embodiments, the haptenization reagent is selected from trinitrochlorobenzene (TNCB), 2,4-difluoronitrobenzene (DNFB), N-iodoacetyl-N'-(5-sulfonic-l-naphthyl)ethylenediamine (AED), sulfanilic acid (SA), trinitrophenol (TNP), 2,4,6-trinitrobenzenesulfonic acid (TNBS) and combinations thereof.
[0023] A haptenized S protein immunogen (and corresponding immunogenic epitopes) and fragments, and variants thereof may be produced synthetically or recombinantly. A coronavirus S protein fragment that contains an epitope that induces an immune response against coronavirus may be synthesized by standard chemical methods, including synthesis by automated procedure. In general, immunogenic peptides are synthesized based on the standard solid-phase Fmoc protection strategy with HATU as the coupling agent. The immunogenic peptide is cleaved from the solid-phase resin with trifluoroacetic acid containing appropriate scavengers, which also deprotects side chain functional groups. The crude immunogenic peptide may be further purified using preparative reverse phase chromatography. Other purification methods, such as partition chromatography, gel filtration, gel electrophoresis, or ionexchange chromatography may be used. Other synthesis techniques known in the art may be employed to produce similar immunogenic peptides, such as the tBoc protection strategy, use of different coupling reagents, and the like. In addition, any naturally occurring amino acid or derivative thereof may be used, including D-amino acids or L-amino acids, and combinations thereof. In certain embodiments, a synthetic S protein immunogen has an amino acid sequence that is identical to, or at least 85% identical (which includes at least 90% or 95% or any percent in between 85% and 100%) to SEQ ID NO :2.
[0024] As described herein, the haptenized S protein immunogens may be recombinant, wherein desired S protein immunogens are individually or in combination expressed from a polynucleotide that is operably linked to an expression control sequence (e.g. a promoter) in a nucleic acid expression construct. In certain embodiments, a recombinant S protein antigen will comprise an amino acid sequence that is identical to, or at least 85% identical (which includes at least 90% or 95% or any percent in between 80% and 100%) to SEQ. ID NO:2. In another embodiment, a recombinant S protein immunogen consists of an amino acid sequence as set forth in SEQ ID NO:1 or 2. In other embodiments, recombinant S protein immunogens and variants thereof are fragments of SEQ ID NO:1 or 2. In some embodiments, the variants are SARS-CoV-2 Spike protein variants found in different strains of SARS-CoV- 2. Exemplary variants of the Spike protein from these different strains are set forth in Table 1. Variants include, but are not limited to, Spike proteins from the alpha, beta or delta variant of SARS-CoV-2, B.1.1.7 strain, B.1.351 strain, P.l strain, CAL 20 strain or any combination thereof.
[0025] Table 1. List of amino acid positions and relative amino acid changes in the different variants in the Spike protein with respect to the ancestral Wuhan strain Spike protein (SEQ ID NO: 2). protein position Wuhan Bl.1.7 Bl.351 P.l CAL.20
S 13 S 1
S 18 L F F
S 20 T N
S 26 P S
S 69 H Del
S 70 V Del
S 80 D A
S 138 D Y
S 145 Y Del
S 152 W C
S 190 R S
S 215 D G/H
S 241 L Del
S 242 L Del
S 243 A Del
S 417 K N T
S 452 L R
S 484 E K K
S 501 N Y Y Y
S 570 A D
S 614 D G G G G
S 655 H Y
S 681 P H
S 701 A V
S 716 T 1
S 938 L F
S 982 S A
S 1027 T 1
S 1118 D H
S 1176 V F
S 1191 K N [0026] In one embodiment, the S proteins are haptenized. For purposes of the present invention, virtually any small protein or other small molecule that fails to induce an immune response when administered alone, may function as a hapten. A variety of haptens of quite different chemical structure have been shown to induce similar types of immune response, e.g. TNP (Kempkes, J. Immunol. (1991) 147:2467); phosphorylcholine (Jang, Eur. J. Immunol. (1991) 21:1303); nickel (Pistoor, J. Invest.
Dermatol. (1995) 105:92) and arsenate (Nalefski, J. Immunol. (1993) 150:3806). Conjugation of a hapten to a cell to elicit an immune response may preferably be accomplished by conjugation via e-amino groups of lysine or --COOH groups. This group of haptens include a number of chemically diverse compounds: dinitrophenyl, trinitrophenyl, N-iodoacetyl-N'-(5-sulfonic 1-naphthyl) ethylene diamine, trinitrobenzene-sulfonic acid, dinitrobenzene sulfonic acid, fluorescein isothiocyanate, arsenic acid benzene isothiocyanate, and dinitrobenzene-S-mustard (Nahas, Cellular Immunol. (1980) 54:241). Once armed with the present disclosure, skilled artisans would be able to choose haptens for use in the present invention.
[0027] In general, haptens include a "recognition group" which is the group that interacts with an antibody. The recognition group is irreversibly associated with the hapten reactive group. Thus, when the hapten reactive group is conjugated to a functional group on the target molecule, the hapten recognition group is available for binding with antibody. Examples of different hapten recognition groups include without limitation to dinitriophenyl, trinitrophenyl, fluorescein, other aromatics, phosphorylcholine, peptides, advanced glycosylation endproducts (AGE), carbohydrates, etc.
[0028] Haptens also include a functional group for conjugation to a substituent on an amino acid side chain of a protein or polypeptide. Amino acid side chain groups that can be conjugated to hapten include, e.g. free carboxylic acid groups in the aspartic acid or glutamic acid; the e-amino group of lysine; the thiol moiety of cysteine; the hydroxyl group of serine or tyrosine; the imidazole moiety of histidine; or the aryl groups of tryptophan, tyrosine, or phenylalanine. Hapten functional groups capable of reacting with specific amino acid side chains are described as follows.
[0029] Functional groups reactive with primary amines. Hapten reactive groups that would form a covalent bond with primary amines present on amino acid side chains would include, but not be limited to, acid chlorides, anhydrides, reactive esters, a,p-unsaturated ketones, imidoesters, and halonitrobenzenes. Various reactive esters with the capability of reacting with nucleophilic groups such as primary amines are available commercially. Functional groups reactive with carboxylic acids. Carboxylic acids in the presence of carbodiimides, such as EDC, can be activated, allowing for interaction with various nucleophiles, including primary and secondary amines. Alkylation of carboxylic acids to form stable esters can be achieved by interaction with sulfur or nitrogen mustards, or haptens containing either an alkyl or aryl aziridine moiety.
[0030] Functional groups reactive with aromatic groups. Interaction of the aromatic moieties associated with certain amino acids can be accomplished by photoactivation of aryl diazonium compound in the presence of the protein or peptide. Thus, modification of the aryl side chains of histidine, tryptophan, tyrosine, and phenylalanine, particularly histidine and tryptophan, can be achieved by the use of such a reactive functionality.
[0031] Functional groups reactive with sulfhydryl groups. There are several reactive groups that can be coupled to sulfhydryl groups present on the side chains of amino acids. Haptens containing an a,p- unsaturated ketone or ester moiety, such as maleimide, provide a reactive functionality that can interact with sulfhydryl as well as amino groups. In addition, a reactive disulfide group, such as 2-pyridyldithio group or a 5,5'-dithio-bis-(2-nitrobenzoic acid) group is also applicable. Some examples of reagents containing reactive disulfide bonds include N-succinimidly 3-(2-pyridyl-dithio) propionate (Carlsson, Biochem J. (1978) 173:723-737), sodium S-4-succinimidyloxycarbonyl-alpha-methylbenzyl-thiosulfate, and 4 succinimidyloxycarbonyl-alpha-methyl-(2-pyridyldithio)-toluene. Some examples of reagents comprising reactive groups having a double bond that reacts with a thiol group include succinimidyl 4- (N-maleimidomethyl)cyclohexahe-l-carboxylate and succinimidyl m-maleimidobenzoate.
[0032] Other functional molecules include succinimidyl 3-(maleimido)-propionate, sulfosuccinimidyl 4- (p-maleimido-phenyl)butyrate, sulfo-succinimidyl-4-(N-maleimidomethyl-cyclohexane)-l-carboxylate, maleimidobenzolyl-N-hydroxy-succinimide ester.
[0033] Any hapten or combination of different haptens can be used in the compositions of the invention. For example in one embodiment, the same hapten recognition group is coupled to different amino acids through different functional groups on the S protein. For example, the reagents dinitrobenzene sulfonic acid, dinitro phenyldiazonium, and dinitrobenzene S mustard, all form the dinitrophenyl hapten coupled to amino groups, aromatic groups, and carboxylic acid groups, respectively. Similarly, an arsonic acid hapten can be coupled by reacting arsonic acid benzene isothiocyanate to amino groups or azobenzenearsonate to aromatic groups. Immunogenic Compositions & Vaccines
[0034] Immunogenic compositions or vaccines as described herein useful for treating and/or preventing a coronavirus infection comprises immunogenic sarbecovirus polypeptides, such as S protein, fragments, and variants thereof, and also includes a fusion of a sarbecovirus immunogen to other peptides or polypeptides (e.g. a hydrophobic amino acid sequence or a histidine tag or a non-S protein coronavirus polypeptide or fragment thereof) or other modifications (e.g. glycosylation). In certain embodiments, the immunogenic S polypeptides may comprise any portion of an S protein that has an epitope capable of eliciting a protective immune response (e.g. eliciting production of a neutralizing antibody and/or stimulating a cell-mediated immune response) against a sarbecovirus infection. Immunogenic polypeptides as described herein may be arranged, combined, or fused in a linear form, and each immunogen may or may not be reiterated, wherein the reiteration may occur once or multiple times, and may be located at the N-terminus, C-terminus, or internal to a linear sequence of immunogenic S or other sarbecovirus polypeptide immunogens. In addition, a plurality of different coronavirus immunogenic polypeptides (e.g. other S proteins from multiple sarbecovirus species, N proteins, M proteins, or other coronavirus polypeptides, and variants or fragments thereof) can be selected and mixed or combined into a cocktail composition to provide a multivalent vaccine for use in eliciting a protective immune response without a harmful or otherwise unwanted associated immune responses or side effects. Also provided herein are methods for producing a haptenized synthetic or recombinant multivalent coronavirus polypeptide immunogens, including fusion proteins. For example, host cells containing an S protein immunogen-encoding nucleic acid expression construct may be cultured to produce the recombinant S protein immunogen, or variants thereof (e.g. deletion mutants or S polypeptide fragments lacking a C-terminal transmembrane domain). Also contemplated are methods for treating or preventing coronavirus infections or eliciting an immune response using an S protein immunogen or variant thereof, or a combination of polypeptides (including fusion proteins).
[0035] Coronavirus has a positive-sense, non-segmented, single-stranded RNA genome, which encodes at least 18 viral proteins (such as non-structural proteins (NSP) 1-13, structural proteins E, M, N, S), and an RNA-dependent RNA polymerase). Coronavirus has three major surface glycoproteins (designated S, E, and M), and some coronaviruses have another surface glycoprotein referred to as hemagglutinin esterase (HE), which is not found in the SARS virus, the N (nucleocapsid) protein is a basic phosphoprotein, which is generally associated with the genome and has been reported to be antigenic (Holmes, Fields Virology, Chapter 34, 2013). The S (spike) protein, a major antigen of coronavirus, has multiple domains: SI which is believed to be involved in receptor binding, S2 believed to mediate membrane fusion between the virus and target cell, and the receptor binding domain (RBD) believed to mediate binding to the ACE2 receptor.
[0036] The S (spike) protein may form non-covalently linked homotrimers (oligomers), which may mediate receptor binding and virus infectivity. Homotrimers of S proteins are likely necessary for presenting the correct native conformation of receptor binding domains and for eliciting a neutralizing antibody response. In addition, intracellular processing of S protein is associated with significant posttranslation oligosaccharide modification. The posttranslation oligosaccharide modification (glycosylation) expected by N-glycan motif analysis indicates that the S protein has as many as 23 sites for such modification. In addition, C-terminal cysteine residues may also participate in protein folding and preserving the native (functional) S protein conformation. The S protein of some coronaviruses (e.g. some strains of group II and III viruses) can be proteolytically processed near the center of the S protein by a trypsin-like protease in the Golgi apparatus or by extracellularly localized enzymes into to a linked polypeptide, containing an N-terminal SI and a C-terminal S2 polypeptide. Some members of the type II group of coronaviruses and group I viruses may not be so processed. Until the characterization of the SARS-associated viral agent as a coronavirus, the coronaviruses were divided into three groups on the basis of serological and genetic properties, which groups were referred to as Group 1 , Group 2, and Group 3, which are also referred to in the art and herein as Group I, Group II, and Group III (see e.g. Holmes, Fields Virology, supra; Stadler, Nat. Rev. Microbiol. (2003) 209-18; Holmes, J Clin. Invest. (2003) 111 :1605-609). Presently, the coronaviruses are subdivided into Group 1, Group 2, Group 3, and SARS- CoV (SARS-associated coronavirus including SARS-CoV-2).
[0037] An exemplary S protein is from SARS-CoV-1 and has 1,255 amino acids (see SEQ ID NO:4), with a 12 amino acid signal sequence, the SI domain between amino acids 13-679 and the S2 domain between amino acids 680-1195. In certain embodiments, the SARS-CoV-1 S protein and variants thereof that have one or more epitopes (i.e. are immunogens) and that are capable of eliciting a neutralizing (e.g. IgA or IgG antibody) or cell-mediated immune response against multiple species of sarbecovirus (e.g. a pan- sarbecovirus immune response), are included in compositions for use in treating or preventing coronavirus infections. Also described herein is the identification of SARS-CoV-1 S protein immunogens (containing one or more immunogenic epitopes) that are not glycosylated and that are capable of eliciting a neutralizing immune response. In one embodiment, the SARS-CoV-1 S protein immunogen is a portion or fragment of the full-length S protein. This region also corresponds to the region of the SI domain that is believed to be involved with cell receptor binding. Accordingly, a fragment comprising the receptor binding domain (RBD) amino acids at positions 318-510 of SEQ. ID NO:4, or a portion thereof, may be immunogenic and an immune response specific for one or more epitopes within this sequence may prevent entry of the coronavirus into a target cell. In addition, identification of such immunogenic fragments of the SARS-CoV-1 S protein that do not contain glycosylation sites provides the advantage that the fragments may be made and produced in cells, such as bacteria, that are not capable of glycosylating a protein in the same manner as a mammalian cell.
[0038] As described herein, a SARS-CoV-1 S protein immunogen includes a fragment of S protein or a S protein variant (which may be a variant of a full-length SARS-CoV-1 S protein or fragment as described herein) that retains or that has at least one epitope contained within the full-length S protein or wildtype S protein, respectively, that elicits a protective immune response against coronavirus, preferably against SARS coronavirus, including multiple species of sarbecovirus. A SARS-CoV-1 S protein fragment or an S protein variant has at least one biological activity or function of a full-length or wildtype (natural) SARS-CoV-1 S protein (such as receptor binding or cell fusion activity), or has multiple S protein-specific biological activities or functions. For example, a SARS-CoV-1 S protein variant may contain an epitope that induces an immune response (for example, induces production of an antibody that specifically binds to a wildtype or full-length S polypeptide) or may have S protein receptor binding activity. In one embodiment, a SARS-CoV-1 S-protein fragment is a truncated S-protein that comprises an amino acid set forth at positions 1-1195 of SEQ ID NO:4. The portion of the S-protein that is deleted is the transmembrane region. SARS-CoV-1 S protein immunogenic fragments also include smaller portions or fragments of the aforementioned amino acid fragments of an S protein. A SARS-CoV-1 S protein fragment that comprises an epitope that stimulates, induces, or elicits an immune response may comprise a sequence of consecutive amino acids ranging from any number of amino acids between 8 amino acids and 150 amino acids (e.g. 8, 10, 12, 15, 18, 20, 25, 30, 35, 40, 50 or more amino acids) of SEQ. ID NO:4. In related embodiments, a SARS-CoV-1 S polypeptide variant has at least 85% to 100% amino acid sequence identity (that is, at least 85%, 90%, 95% or 99% sequence identity) to the amino acid sequence of the full length S protein as set forth in SEQ ID NO:4 (which is from SARS-CoV-4; SEQ ID NO:3 is the nucleic acid sequence that encodes the amino acid sequence of SEQ ID NO:4). Such SARS- CoV-1 S protein variants and fragments retain at least one S protein-specific biological activity or function, such as (1) the capability to elicit a protective immune response (that is, the S polypeptide variant contains an epitope that induces or elicits a protective immune response), for example, a neutralizing response and/or a cell-mediated immune response against multiple species of sarbecovirus, such as SARS-CoV-1 and SARS-CoV-2; (2) the capability to mediate viral infection via receptor binding; and (3) the capability to mediate membrane fusion between a virion and the host cell. Additional examples of full-length SARS-CoV-1 S protein sequences are available in the art.
[0039] An exemplary S protein is from SARS-CoV-2 and has 1,255 amino acids (see SEQ ID NO:2), with a 12 amino acid signal sequence, the SI domain between amino acids 12-672 and the S2 domain between amino acids 673-1192. In certain embodiments, SARS-CoV-2 S proteins and variants thereof that have one or more epitopes (i.e. are immunogens) and that are capable of eliciting a neutralizing (e.g. IgA or IgG antibody) or cell-mediated immune response, are included in compositions for use in treating or preventing sabrecovirus infections. Also described herein is the identification of S protein immunogens (containing one or more immunogenic epitopes) that are not glycosylated and that are capable of eliciting a neutralizing immune response. In one embodiment, the SARS-CoV-2 S protein immunogen is a portion or fragment of the full-length S protein. For example, a portion of the SARS-CoV-2 S protein immunogen that includes amino acids at positions 417-560 of SEQ. ID NO:2 does not contain an N-glycan substitution site and is a hydrophilic region. This region also corresponds to the region of the SI domain that is believed to be involved with cell receptor binding. Accordingly, a fragment comprising amino acids at positions 417-560 of SEQ ID NO:2 corresponding to the receptor binding domain (RBD), or a portion thereof, may be immunogenic and an immune response specific for one or more epitopes within this sequence may prevent entry of the coronavirus into a target cell. In addition, identification of such immunogenic fragments of the SARS-CoV-2 S protein that do not contain glycosylation sites provides the advantage that the fragments may be made and produced in cells, such as bacteria, that are not capable of glycosylating a protein in the same manner as a mammalian cell.
[0040] As described herein, a SARS-CoV-2 S protein immunogen includes a fragment of S protein or a S protein variant (which may be a variant of a full-length SARS-CoV-2 S protein or fragment as described herein) that retains or that has at least one epitope contained within the full-length S protein or wildtype SARS-CoV-2 S protein, respectively, that elicits a protective immune response against multiple species of sarbecovirus. An SARS-CoV-2 S protein fragment or an S protein variant has at least one biological activity or function of a full-length or wildtype (natural) S protein (such as receptor binding or cell fusion activity), or has multiple S protein-specific biological activities or functions. For example, a SARS-CoV-2 S protein variant may contain an epitope that induces an immune response (for example, induces production of an antibody that specifically binds to a SARS-CoV-2 wildtype or full-length S protein) or may have S protein receptor binding activity. In one embodiment, an S-protein fragment is a truncated SARS-CoV-2 S protein that comprises an amino acid set forth at positions 1-1200 of SEQ ID NO:2. The portion of the SARS-CoV-2 S protein that is deleted is the transmembrane region. SARS-CoV-2 S protein immunogenic fragments also include smaller portions or fragments of the aforementioned amino acid fragments of a SARS-CoV-2 S protein. A SARS-CoV-2 S protein fragment that comprises an epitope that stimulates, induces, or elicits an immune response may comprise a sequence of consecutive amino acids ranging from any number of amino acids between 8 amino acids and 150 amino acids (e.g. 8, 10, 12, 15, 18, 20, 25, 30, 35, 40, 50 or more amino acids) of SEQ ID NO:2. In related embodiments, a SARS-CoV-2 S polypeptide variant has at least 85% to 100% amino acid sequence identity (that is, at least 85%, 90%, 95% or 99% sequence identity) to the amino acid sequence of the full length S protein as set forth in SEQ. ID NO:2 (which is from SARS-CoV-2; SEQ ID NO: 1 is the nucleic acid sequence that encodes the amino acid sequence of SEQ ID NO:2). Such S protein variants and fragments retain at least one SARS-CoV-2 S protein-specific biological activity or function, such as (1) the capability to elicit a protective immune response (that is, the S polypeptide variant contains an epitope that induces or elicits a protective immune response), for example, a neutralizing response and/or a cell- mediated immune response against coronavirus, such as SARS-CoV-2; (2) the capability to mediate viral infection via receptor binding; and (3) the capability to mediate membrane fusion between a virion and the host cell. Additional examples of full-length SARS-CoV-2 S protein sequences are available in the art. [0041] As described herein, S protein immunogens, fragments, and variants thereof described herein contain a haptenized epitope that elicits or induces an immune response, preferably a protective immune response, which may be a humoral response and/or a cell-mediated immune response. A protective immune response may be manifested by at least one of the following: preventing infection of a host by a coronavirus; modifying or limiting the infection; aiding, improving, enhancing, or stimulating recovery of the host from infection; and generating immunological memory that will prevent or limit a subsequent infection by a coronavirus. A humoral response may include production of antibodies that neutralize infectivity, lyse the virus and/or infected cell, facilitate removal of the virus by host cells (for example, facilitate phagocytosis), and bind to and facilitate removal of viral antigenic material. A humoral response may also include a mucosal response, which comprises eliciting or inducing a specific mucosal IgA response.
[0042] Induction of an immune response in a subject or host (human or non- human animal) by a haptenized S protein, fragment, or variant described herein, may be determined and characterized by methods described herein and routinely practiced in the art. These methods include in vivo assays, such as animal immunization studies (e.g. using a rabbit, mouse or rhesus macaque model), and any one of a number of in vitro assays, such as immunochemistry methods for detection and analysis of antibodies, including Western immunoblot analysis, ELISA, immunoprecipitation, radioimmunoassay, and the like, and combinations thereof. By way of example, animal models may be used for determining the capability of a coronavirus antigen to elicit and induce an immune response that is protective in animals, which may be determined by endpoints relevant to the particular model.
[0043] Other methods and techniques that may be used to analyze and characterize an immune response include neutralization assays (such as a plaque reduction assay or an assay that measures cytopathic effect (CPE) or any other neutralization assay practiced by persons skilled in the art) to assess whether a haptenized S protein immunogen or variant thereof is capable of eliciting an immune response, particularly a neutralizing immune response
[0044] The haptenized S protein immunogens (full-length proteins, variants, fragments, and fusion proteins thereof) are provided in an isolated form, and in certain embodiments, are purified to homogeneity. As used herein, the term "isolated" means that the polypeptide is removed from its original or natural environment.
[0045] In an embodiment, the invention provides a method of immunizing a human subject against a Coronavirus, the method comprising administering an effective amount of an haptenized S protein from Coronavirus, including SARS-CoV-2 Coronavirus. In some embodiments, the S protein is from SARS-CoV- 2 Coronavirus and has the amino acid sequence as set forth in SEQ ID NO:2.
[0046] In some embodiments, the haptenized S protein is administered in a first dose followed by a second dose at three to four weeks weeks. In some embodiments, the method further comprising at least one booster injection of the haptenized S protein about six months after the first injection. In some embodiments, booster injections continue every six months or until an immunogenic response against a sarbecovirus occurs in the human subject.
[0047] In some embodiments, the effective amount of haptenized S protein is administered until the delayed type hypersensitivity diagnostic test is positive, or a neutralizing antibody response is detected (e.g. anti-S protein antibodies are detected in the blood or serum of the human subject).
[0048] In an embodiment, the invention provides a method of producing an immunogenic response in a human subject against a Coronavirus, including SARS-CoV-2 Coronavirus, the method comprising administering an effective amount of an haptenized S protein from Coronavirus, including SARS-CoV-2 Coronavirus. In some embodiments, the S protein is from SARS-CoV-2 Coronavirus and has the amino acid sequence as set forth in SEQ. ID NO:2. Methods of Immunization
[0049] In some embodiments, the invention encompasses a method of immunizing a human subject against multiple species of sarbecovirus comprising: (a) immunizing a human subject against SARS-CoV- 1, and (b) immunizing a human subject against SARS-CoV-2, wherein the human subject has never been infected with, nor immunized against, SARS-CoV-1 and SARS-CoV-2. In some embodiments, the immunizing comprises: (i) administration to the human subject of an immunogenic composition comprising: (a) an S protein or fragment thereof from SARS-CoV-1 and at least one pharmaceutically acceptable carrier, or (b) a polynucleotide encoding an S protein or fragment thereof from SARS-CoV-1 and at least one pharmaceutically acceptable carrier; and (ii) administration to the human subject of an immunogenic composition comprising: (a) an S protein or fragment thereof from SARS-CoV-2 and at least one pharmaceutically acceptable carrier, or (b) a polynucleotide encoding an S protein or fragment thereof from SARS-CoV-2 and at least one pharmaceutically acceptable carrier. In some embodiments, the immunizing against SARS-CoV-1 occurs first, followed by a second immunizing against SARS-CoV-2. In some embodiments, the immunizing against SARS-CoV-2 occurs first, followed by a second immunizing against SARS-CoV-1.
[0050] In some embodiments, the invention encompasses a method of immunizing a human subject against multiple species of sarbecovirus comprising: (i) administering to the human subject an immunogenic composition comprising: (a) an S protein or fragment thereof from SARS-CoV-1 and at least one pharmaceutically acceptable carrier, or (b) a polynucleotide or fragment thereof encoding an S protein from SARS-CoV-1, wherein the human subject has already been immunized against SARS-CoV-2 and/or been infected with SARS-CoV-2. In some embodiments, the S protein fragment comprises the receptor binding domain (RBD) of the S protein while in other embodiments, the S protein fragment comprises the SI domain of the S protein.
[0051] In some embodiments, the S protein or fragment thereof is haptenized. In some embodiments, the second immunization occurs at least three weeks after the first immunization. In some embodiments, the method further comprises administering at least one booster injection of the immunogenic composition about six months after the first injection. In some embodiments, the immunogenic composition is administered until a cross-reactive neutralizing antibody response against multiple species of sarbecovirus is detected in the human subject. In some embodiments, the human subject has been previously infected with SARS-CoV-2 and recovered from Covid-19. In some embodiments, the human subject has previously been immunized against SARS-CoV-2. In some embodiments, the SARS-CoV-2 immunization is an mRNA vaccine, inactivated virus vaccine, viral vector vaccine, or protein subunit vaccine. In some embodiments, the mRNA vaccine is BNT162b2 or mRNA-
1273. In some embodiments, viral vector vaccine is Ad26.COV2.S COVID-19 or ChAdOxl.
[0052] Also described herein are methods for treating and/or preventing a sarbecovirus infection in a subject previously not infected with SARS-CoV-1 or SARS-CoV-2, comprising administering to the subject in need thereof a first immunogenic composition comprising at least one SARS-CoV-1 S protein immunogen, and a second immunogenic composition comprising at one SARS-CoV-2 S protein immunogen. In some embodiments, the SARS-CoV-1 immunogenic composition is administered first, followed by administration of the SARS-CoV-2 immunogenic composition. In some embodiments, the SARS-CoV-2 immunogenic composition is administered first, followed by administration of the SARS- CoV-1 immunogenic composition. In some embodiments, the subject has not been infected with any species of sarbecovirus.
[0053] In some embodiments, the SARS-CoV-1 S protein immunogen comprises an amino acid sequence that is identical to, or at least 85% identical to (which includes at least 90% or 95% or any percent in between 85% and 100%) SEQ ID NO: 4 and wherein the SARS-CoV-1 S protein immunogen has an epitope that elicits a protective immune response, which is a humoral immune response (including, for example, a mucosal IgA, systemic IgA, IgG, IgM response) and/or a cell-mediated immune response, and pharmaceutically acceptable carrier, diluent, or excipient. The SARS-CoV-1 S protein immunogen composition is administered at a dose sufficient to elicit a pan-sarbecovirus immune response specific for multiple species of sarbecovirus S protein immunogen, immunogens or variants thereof. In certain embodiments, an infection being prevented or treated may be caused by any type of sarbecovirus species, or a combination thereof. In some embodiments, the SARS-CoV-1 S protein immunogen is haptenized as described herein.
[0054] In some embodiments, the SARS-CoV-2 S protein immunogen comprises an amino acid sequence that is identical to, or at least 85% identical to (which includes at least 90% or 95% or any percent in between 85% and 100%) SEQ. ID NO: 2 and wherein the SARS-CoV-2 S protein immunogen has an epitope that elicits a protective immune response, which is a humoral immune response (including, for example, a mucosal IgA, systemic IgA, IgG, IgM response) and/or a cell-mediated immune response, and pharmaceutically acceptable carrier, diluent, or excipient. The SARS-CoV-2 S protein immunogen composition is administered at a dose sufficient to elicit a pan-sarbecovirus immune response specific for multiple species of sarbrecovirus S protein immunogen or immunogens or variants thereof. In certain embodiments, an infection being prevented or treated may be caused by any type of sarbecovirus species, or a combination thereof. In some embodiments, the SARS-CoV-2 S protein immunogen is haptenized as described herein.
[0055] A human subject or host suitable for treatment with a coronavirus immunogen composition or formulation may be identified by well-established indicators of risk for developing a disease such as Covid-19 or by well-established hallmarks of an existing sarbecovirus disease. For example, indicators of an infection include fever, dry cough, dyspnea (shortness of breath), headache, hypoxemia (low blood oxygen concentration), lymphopenia (reduced lymphocyte numbers), mildly elevated aminotransferase levels (indicating liver damage), microorganism positive cultures, inflammation, and the like. Infections that may be treated or prevented with a coronavirus S protein immunogen vaccine as described herein include those caused by or due to coronavirus, whether the infection is primary, secondary or opportunistic. Examples of sarbecovirus include any species, strain, antigenic variant, and the like, of these viruses, including SARS coronavirus such as SARS-CoV-1, Bat CoV WIV17, Bat CoV RsSHC0147, SARS-CoV-2, SARS-CoV-2 Alpha variant, SARS-CoV-2 Beta variant, SARS-CoV-2 Delta variant, Bat CoV RaTG135, Pangolin CoV GD-16, Pangolin GX-P5L6. By way of example, sarbecovirus infections are characterized by flu-like symptoms, including high fever, myalgia, dry and non-productive dyspnea, lymphopenia, and infiltrate on chest radiography.
[0056] The immunogenic compositions that contain one or more haptenized coronavirus S protein immunogens of the invention may be in any form that allows for the composition to be administered to a subject, such as a human or non-human animal. For example, a haptenized S protein immunogen may be prepared and administered as a liquid solution or prepared as a solid form (e.g. lyophilized), which may be administered in solid form, or resuspended in a solution in conjunction with administration. The hybrid polypeptide composition is prepared or formulated to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a subject or patient or to be bioavailable via slow release. Compositions that will be administered to a subject or patient take the form of one or more dosage units; for example, a tablet may be a single dosage unit, and a container of one or more compounds of the invention in aerosol form may hold a plurality of dosage units. In certain preferred embodiments, any of the aforementioned immunogenic compositions or vaccines comprising a haptenized S protein immunogen described herein are in a container, preferably in a sterile container.
[0057] In one embodiment, the immunogenic composition or vaccine is administered nasally, wherein a haptenized coronavirus S protein immunogen can be taken up by cells, such as cells located in the nasal- associated lymphoid tissue. Other typical routes of administration include, without limitation, parenteral, transdermal/transmucosal, nasal, and inhalation. The term "parenteral" as used herein, describes administration routes that bypass the gastrointestinal tract, including intraarterial, intradermal, intramuscular, intranasal, intraocular, intraperitoneal, intravenous, subcutaneous, submucosal, and intravaginal injection or infusion techniques. The term "transdermal/transmucosal" as used herein, is a route of administration in which the immunogenic composition is administered through or by way of the skin, including topical. The terms "nasal" and "inhalation" encompass techniques of administration in which an immunogenic composition is introduced into the pulmonary tree, including intrapulmonary or transpulmonary. In one embodiment, the compositions of the present invention are administered nasally.
[0058] In some embodiments, the immunogenic composition or vaccine contains an amount of haptenized S protein from about 30 pg to about 300 pg per dose. In some embodiments, the amount of haptenized coronavirus S protein administered per dose is from about 1 pg to about 240 pg. In some embodiments, the amount of haptenized S protein administered per dose is from about 1 pg, 3 pg, 5 pg, 10 pg, 25 pg, 30 pg, 40 pg, 50 pg, 60 pg, 70 pg, 80 pg, 90 pg, 100 pg, 110 pg, 120 pg, 130 pg, 140 pg, 150 pg, 160 pg, 170 pg, 180 pg, 190 pg, 200 pg, 210 pg, 220 pg, 230 pg, 240 pg, 250 pg or more. In some embodiments, the amount of haptenized coronavirus S protein varies depending on dosing schedule. For example, an initial dose may be the same as, lower or higher than any subsequent immunization dose, including a booster immunization dose. The amount of haptenized S protein administered per dose to any human subject can be adjusted according to the age, weight and physical condition of the human subject.
Adjuvants
[0059] In some embodiments, the invention provides an immunogenic composition or vaccine comprising an haptenized S protein for injection further comprising a vaccine adjuvant.
[0060] In one embodiment, a composition that is useful as an immunogenic composition for treating and/or preventing a coronavirus infection contains at least one coronavirus antigen (immunogen) as described herein capable of eliciting an immune response and protollin or proteosome adjuvant (see e.g. U.S. Patent No. 5,726,292). As is understood in the art, an adjuvant may enhance or improve the immunogenicity of an immunogen (that is, act as an immunostimulant), and many antigens are poorly immunogenic unless combined or admixed or mixed with an adjuvant. A variety of sources can be used as a source of antigen, such as live attenuated virus, killed virus, split antigen preparations, subunit antigens, recombinant or synthetic viral antigens, and combinations thereof. To maximize the effectiveness of a subunit, recombinant, or synthetic vaccine, the antigens can be combined with a potent immunostimulant or adjuvant. Other exemplary adjuvants include alum (aluminum hydroxide, REHYDRAGEL); aluminum phosphate; virosomes; liposomes with and without Lipid A; or other oil in water emulsions type adjuvants such as MF59 (Novartis), also such as nanoemulsions (see e.g. U.S. Patent No. 5,716,637) or submicron emulsions (see e.g. U.S. Patent No. 5,961,970); and Freund's complete and incomplete adjuvant.
[0061] A proteosome-based adjuvant (i.e. protollin or proteosome) can be used in vaccine compositions or formulations that may include any one or more of a variety of coronavirus antigen (immunogen) sources as described herein. Proteosomes are comprised of outer membrane proteins (OMP) from Neisseria species typically, but can be derived from other Gram-negative bacteria (see e.g. U.S. Patent No. 5,726,292). Proteosomes have the capability to auto-assemble into vesicle or vesicle-like OMP clusters of 20-800 nm, and to noncovalently incorporate, coordinate, associate, or otherwise cooperate with protein antigens, particularly antigens that have a hydrophobic moiety. Proteosomes are hydrophobic, safe for human use, and comparable in size to certain viruses. By way of background, and not wishing to be bound by theory, mixing proteosomes with an antigen such as a protein antigen, provides a composition comprising non-covalent association or coordination between the antigen and Proteosomes, which association or coordination forms when solubilizing detergent is selectively removed or reduced in concentration, for example, by dialysis.
[0062] Any preparation method that results in the outer membrane protein component in vesicular or vesicle-like form, including molten globular-like OMP compositions of one or more OMP, is included within the scope of proteosome. In one embodiment, the proteosomes are from Neisseria species, and from Neisseria meningitidis. In certain other embodiments, proteosomes may be an adjuvant and an antigen delivery composition. In an embodiment, an immunogenic composition comprises one or more coronavirus antigens and an adjuvant, wherein the adjuvant comprises Projuvant or Protollin. As described herein, a coronavirus antigen may be isolated from the virus particles, a cell infected by the coronavirus, or from a recombinant source. In certain embodiments, an immunogenic composition further comprises a second immunostimulant, such as a liposaccharide. That is, the adjuvant may be prepared to include an additional immunostimulant. For example, the projuvant may be mixed with a liposaccharide to provide an OMP-LPS adjuvant. Thus, the OMP-LPS (protollin) adjuvant can be comprised of two components. The first component includes an outer membrane protein preparation of proteosomes (i.e. Projuvant) prepared from Gram-negative bacteria, such as Neisseria meningitidis, and the second component includes a preparation of liposaccharide. It is also contemplated that the second component may include lipids, glycolipids, glycoproteins, small molecules or the like, and combinations thereof. As described herein, the two components of an OMP-LPS adjuvant may be combined (admixed or formulated) at specific initial ratios to optimize interaction between the components, resulting in stable association and formulation of the components for use in the preparation of an immunogenic composition. The process generally involves the mixing of components in a selected detergent solution (e.g. Empigen BB, Triton x-100 or Mega-10) and then effecting complex formation of the OMP and LPS components while reducing the amount of detergent to a predetermined, preferred concentration by dialysis or by diaf iltration/ultraf iltration methodologies. Mixing, co-precipitation, or lyophilization of the two components may also be used to effect an adequate and stable association, composition, or formulation. In one embodiment, an immunogenic composition comprises one or more coronavirus haptenized S protein antigens and an adjuvant, wherein the adjuvant comprises a projuvant (i.e. proteosome) and liposaccharide.
[0063] In an embodiment, the final liposaccharide content by weight as a percentage of the total proteosome protein can be in a range from about 1% to about 500%, also in range from about 10% to about 200%, or in a range from about 30% to about 150%. Another embodiment includes an adjuvant wherein the proteosomes are prepared from Neisseria meningitidis and the liposaccharide is prepared from Shigella flexneri or Plesiomonas shigelloides, and the final liposaccharide content is between 50% to 150% of the total Proteosome protein by weight. In another embodiment, proteosomes are prepared with endogenous lipooligosaccharide (LOS) content ranging from about 0.5% up to about 5% of total OMP. In another embodiment proteosomes have endogenous liposaccharide in a range from about 12% to about 25%, and in still another embodiment the endogenous liposaccharide is between about 15% and about 20% of total OMP. The instant disclosure also provides an immunogenic composition containing liposaccharide derived from any Gram-negative bacterial species, which may be from the same Gram-negative bacterial species that is the source of proteosomes or may be from a different bacterial species. In certain embodiments, the proteosome or protollin to coronavirus antigen ratio in the immunogenic composition is greater than 1:1, greater than 2:1, greater than 3:1 or greater than 4:1. In other embodiments, proteosome or protollin to haptenized coronavirus S protein antigen ratio in the immunogenic composition is about 1:1, 2:1, 3:1 or 4:1. The ratio can be 8:1 or higher. In other embodiments, the ratio of proteosome or protollin to haptenized coronavirus S protein antigen of the immunogenic composition ranges from about 1:1 to about 1:500, and is at least 1:5, at least 1:10, at least 1:20, at least 1:50, or at least 1:100, or at least 1:200. An advantage of protollin to haptenized coronavirus S protein antigen ratios ranging from 1:2 to 1:200 is that the amount of proteosome-based adjuvant can be reduced dramatically with no significant effect on the ability of a coronavirus antigen to elicit an immune response. In another embodiment, an immunogenic composition comprises one or more haptenized coronavirus S protein immunogens combined (admixed or formulated) with proteosome or protollin, wherein the S protein immunogen comprises an amino acid sequence that is identical to, or at least 85% identical (which includes at least 90% or 95% or any percent in between 85% and 100%) to SEQ ID NO:2 or fragment thereof and wherein the haptenized S protein immunogen or fragment thereof has an epitope that elicits a protective immune response against coronavirus infection. An exemplary haptenized S protein immunogen comprises an amino acid sequence as set forth in SEQ. ID NO:2 or 4, or consisting of SEQ ID NO:2 or 4. In other embodiments, an S protein immunogen is a fragment of SEQ ID NO:2 or 4, which fragment comprises an amino acid sequence that is identical to, or at least 85% identical (which includes at least 90% or 95% or any percent in between 85% and 100%) to an amino acid selected from SEQ ID NO:2 or 4.
[0064] Alternatively, any haptenized S protein immunogen as described herein can be combined (admixed or formulated) in an immunogenic composition with a liposome. Preferably, liposomes that contain one or more sarbecovirus immunogens further comprise Deinococcus radiodurans lipids or a- galactosylphosphotidylglycerolalkylamine. The addition of such lipids in a liposome can enhance the efficacy of a sabrecovirus vaccine composition by increasing protective immunity. Haptenized sabrecovirus S protein immunogens of the present invention may further include a covalently attached hydrophobic portion. A hydrophobic portion may be, for example, an amino acid sequence or a lipid, as disclosed in U.S. Patent No. 5,726,292. Naturally occurring sabrecovirus S protein and a recombinantly expressed S protein having the sequence set forth in SEQ ID NO:2 or 4 contains a hydrophobic transmembrane domain (from about amino acid 1195 to about 1240 of SEQ ID NO:2), which may function as a hydrophobic portion with an S protein immunogen fragment. In certain other embodiments, the haptenized S protein immunogen, may further contain a second amino acid sequence to form a fusion protein, wherein the second amino acid sequence is a tag, carrier, or enzyme, as described herein.
[0065] In other embodiments, immunogenic compositions may comprise (projuvant or protollin), or further comprise components (e.g. receptor ligands) capable of stimulating a host immune response by interacting with certain receptors (e.g. Toll-like receptors or "TLR") produced by one or more host cells of a vaccine recipient. According to one embodiment, compositions comprising immunogenic epitopes of a sabrecovirus protein may contain polypeptide epitopes capable of interacting with Toll-like receptors, thereby promoting an innate immune response, which may or may not evoke a subsequent adaptive immune response.
[0066] An innate immune response is a nonspecific protective immune response that is not a specific antigen-dependent or antibody-dependent response (that is, does not involve clonal expansion of T cells and/or B cells) and may be elicited by any one of numerous antigens, immunogens, or sabrecovirus described herein. An immunostimulatory composition described herein comprises proteosomes and liposaccharide (protollin), either one of which or both may elicit a nonspecific protective response. Without wishing to be bound by theory, one or more components of vaccine compositions or formulations disclosed herein may interact with Toll-like receptors associated with an innate or adaptive immune response of a vaccine recipient. One or more ligands that interact with and subsequently activate certain TLR have been identified, with the exception of TLR8 and TLR10. Certain outer membrane proteins of Neisseria meningitidis, for example OMP2 (also referred to as PorB), interact with TLR2, while LPS of most but not all Gram-negative bacteria interacts with TLR4. Accordingly, one activity of vaccine compositions or formulations described herein, which may contribute to a biological effect, includes activation of one or both of TLR2 and TLR4. An exemplary adjuvant is the TLR4 agonist MPL (3- O-desacyl-4'-monophosphoryl lipid A). Activation of other TLR (other than TLR2 and TLR4) may serve a similar function or further enhance the qualitative or quantitative profile of cytokines expressed, and may be associated with a host Thl/Th2 immune response. It is also contemplated that TLR ligands other than LPS and PorB may be used alone or in combination to activate TLR2 or TLR4. The qualitative or quantitative activation of one or more TLR is expected to elicit, effect, or influence a relative stimulation (balanced or unbalanced) of a Thl or Th2 immune response, with or without a concomitant humoral antibody response. Ligands interacting with TLR other than TLR2 and TLR4 may also be used in vaccine compositions described herein. Such vaccine components may, alone or in combination, be used to influence the development of a host immune response sufficient to treat or protect from virus infection, as set forth herein.
[0067] Other components known to the art may be used in the compositions described herein. Some embodiments of the haptenized S protein immunogen may further comprise adjuvants, such as Bacillus Calmette-Guerin (BCG), cytokines (for non-limiting example, granulocyte-macrophage colonystimulating (GM-CSF)), saponins (for non limiting example Q.S-21), squalenes (for non-limiting example M59), bisphosphonate, aluminum gels or aluminum salts (for non-limiting example, amorphous aluminum hydroxyphosphate sulfate (AAHS), aluminum hydroxide, aluminum phosphate, potassium aluminum sulfate (Alum)), AS01 (a liposome-based vaccine adjuvant system containing 3-O-desacyl-4'- monophosphoryl lipid A (MPL) and the saponin Q.S-21), AS03 (a-tocopherol, squalene and polysorbate 80 in an oil-in-water emulsion), AS04 (3-O-desacyl-4'-monophosphoryl lipid A (MPL) and aluminum salt), and CpG-1018 (Dynavax). Other adjuvants known to the art to non-specifically stimulate immune response and enhance the efficacy of the immune response to the vaccine are also encompassed in the invention.
[0068] A haptenized S protein immunogenic composition or vaccine may further comprise preservatives known to the art, including without limitation, formaldehyde, antibiotics, monosodium glutamate, 2- phenoxyethanol, phenol, and benzethonium chloride. A haptenized S protein immunogenic composition or vaccine may further comprise sterile water for injection, balanced salt solutions for injections.
[0069] While some embodiments of the invention are shown and described herein, such embodiments are provided by way of example only and are not intended to otherwise limit the scope of the invention. Various alternatives to the described embodiments of the invention may be employed in practicing the invention. Therefore, the spirit and scope of the appended claims should not be limited to the description of the preferred versions contained herein.
[0070] The attention of the reader is directed to all papers and documents which are filed concurrently with this specification and which are open to public inspection with this specification, and the contents of all such papers and documents incorporated herein by reference. All the features disclosed in the specification (including any accompanying claims, abstract, and drawings) may be replaced by alternative features serving the same, equivalent or similar purpose, unless expressly stated otherwise. Thus, unless expressly stated otherwise, each feature disclosed is one example only of a generic series of equivalent or similar features.
EXAMPLES
[0071] The embodiments encompassed herein are now described with reference to the following examples. These examples are provided for the purpose of illustration only and the disclosure encompassed herein should in no way be construed as being limited to these examples, but rather should be construed to encompass any and all variations which become evident as a result of the teachings provided herein. Example 1. Preparing haptenized S protein
[0072] The following procedures illustrate exemplary haptenization procedures.
[0073] DNP modification. Modification of the prepared cells with DNP or another hapten may be performed by known methods, e.g. by the method of Miller, J. Immunol. (1976) 117:151 which involves a thirty minute incubation of S protein with DNFB under sterile conditions, followed by washing with sterile saline or HBSS-HSA. For example about 100 mg of DNFB (Sigma Chemical) can be dissolved in about 0.5 ml of 70% ethanol. About 99.5 ml of PBS is added. The solution is stirred overnight in a 37°C water bath. The shelf life of the solution is about four weeks. The S protein is suspended in Hanks balanced salt solution. About 0.1 ml DNFB solution is added to each sample of S protein and incubated for about thirty minutes at room temperature.
[0074] SA modification. Modification of the S protein with SA may be performed by known methods. For example, in one embodiment, sulfanilic acid (SA) is converted to a diazonium salt by adding a saturating amount of sodium nitrite. Ice-cold, sterile filtered (0.2 pm), 10% sodium nitrite solution is added, dropwise, to a SA solution of 100 mg of anhydrous SA dissolved in 10 ml of 0.1N NCI until saturation. (The saturation point corresponds approximately to a final concentration of a sulfanilic acid diazonium salt of about 40 mM). The SA diazonium salt solution is then sterile filtered (0.2 pm membrane), and diluted 1:8 (v/v) in HBSS (without HSA). If needed, the pH is adjusted to 7.2 by dropwise addition of IN NaOH. The SA diazonium salt-HBSS solution is then sterilized by filtration (0.2 pm membrane). S protein is suspended in diazonium salt-HBSS solution. The mixture is incubated for five minutes at room temperature. After the five minute incubation period, the haptenization reaction is stopped by the addition of 0.5 ml of a 25% HSA-HBSS solution to the mixture.
Example 2. Clinical Studies
[0075] This Example outlines clinical studies for immunization with haptenized S protein.
[0076] A human sarbecovirus vaccine, consisting of recombinant S protein from SARS-CoV-2 modified with the hapten, dinitrophenyl (DNP) was prepared as per Example 1. A human sarbecovirus vaccine, consisting of the recombinant S protein from SARS-CoV-2 modified with hapten, DNP was prepared as per Example 1.
[0077] A phase I trial of the haptenized vaccine in patients is conducted, testing four dosage levels. The major endpoints are the presence of a neutralizing antibody response to DNP-modified, SA-modified, and unmodified S protein from both SARS-CoV-1 and SARS-CoV-2. Also, the level of neutralizing antibodies to SARS-CoV-1 and SARS-CoV-2 is assessed over time. Subsequently, a phase II trial using the lowest dose that is found to be immunologically effective in the phase I trial is conducted.
Example 3. Administration Schedule
[0078] The tables below shows an exemplary administration schedule for haptenized S protein to human subject not previously infected with SARS-CoV-2.
Figure imgf000027_0002
Figure imgf000027_0003
Example 4. Preparation of SARS-COV-2 S Protein Modified with Dinitrophenyl (BVX-0320)
[0079]
Figure imgf000027_0001
The following preparation is representative of the process used to prepare the haptenized S- Protein. The scale of the previous prep is given parenthetically. Filtration steps were added to produce a sterile product. Water should be low endotoxin water (nuclease free or WFI grade).
Prepare 0.1M sodium bicarbonate pH 8.2 buffer in water (nuclease free or WFI grade). Filter through 0.2-micron filter (Sigma s6014).
1. Thaw the requisite amount of S-protein (5 mg) (If necessary, filter through 0.2-micron filter).
2. Buffer exchange the S-protein with the bicarbonate buffer.
3. Determine concentration of S-protein (A280, 1.14 ml/mg *cm, target concentration 1 mg/mL).
4. Prepare 0.35M dinitrobenzene sulfonate (Sigma 556971) in 0.1M sodium bicarbonate buffer.
Filter through 0.2-micron filter. 5. Combine S-protein (5 mL) and dinitrobenzene sulfonate (0.375 mL, 2000 equivalents) solutions in an appropriately sized vessel (original preparation split material into 10 vials) and stir gently for 18 hours at 30°C.
6. Ensure pH is greater or equal to 8.0
7. Purify reaction mixture using a Zeba column (equilibrated with 0.1M bicarbonate buffer) into PBS (pH 7.4). The purpose of this step is to both remove excess reagents as well as to perform the buffer exchange. The solution will be uncolored when all dinitrobenzene sulfonate is removed. Filter through 0.2-micron filter into sterile container.
8. Determine final concentration using BCA assay.
9. Analyze the final material for purity (SEC @ 280 nm), free hapten (RP-HPLC @ 235 nm), endotoxin by LAL, hapten loading (MS comparison with native protein).
Example 5. Toxicity Study of BVX-0320 by Subcutaneous Injection in CF-1 Mice
[0080] CF-1 Mice (Charles River) were administered up to 10 pg of BVX-0320 by subcutaneous injection. Mortality was an assessment of toxicity. All animals survived to the scheduled necropsy. There were no unusual BVX-0320-related clinical/veterinary observations. Additional clinical observations were within the range of normal findings for group-housed animals of this age, sex, and species, or were procedure-related and were not considered to be related to test article administration.
[0081] Body Weight and Body Weight Gains. There were no BVX-0320-related body weight effects. Additional minor fluctuations among mean and individual body weight were considered sporadic, consistent with biologic variation, and/or negligible in magnitude and not related to test article administration. There were no BVX-0320-related food consumption effects. Minor fluctuations among mean and individual cage food consumption were considered sporadic, consistent with biologic variation, and/or negligible in magnitude and not related to test article administration. There were no unusual gross pathology findings.
[0082] In conclusion, there were no BVX-0320-related effects on body weight, food consumption or clinical signs. Based on these findings, BVX-0320 was determined to be well tolerated up to 10 pg/dose, the highest tested dose.
Example 6. Immunology Study of BVX-0320 by Subcutaneous Injection in CF-1 Mice 1 [0083] CF-1 Mice (Charles River) were administered 0.3, 1, 3 or lOpg of BVX-0320 by subcutaneous injection followed by a second injection and endpoints were obtained at six weeks (see Figs. 1-2). For the 1 pg group: one mouse had baseline antibody titer 1:5. For the 3 pg group, the titre obtained was 1:120,000. Statistical analyses among the different groups yielded the following results: 0.3 pg group vs. 3 pg group, p=0.002, 1 pg group vs. 3 pg group, p=0.041, 3 pg group vs. 10 pg group, p=0.522.
[0084] Thus, two injections of BVX-0320 induced (1) Antibody against the SI subunit of the spike protein of SARS-Cov-2 (Figure 1). All 4 doses induced antibody, but titers were significantly higher in the two highest dose (3 pg, 10 pg groups). Adjuvant alone (dose = 0 pg) did not induce an antibody response. Pre-vaccine sera were negative with the exception of a very low titer detected in a single sample. (2) Splenic T cells that produced the type I cytokine, gamma interferon, upon restimulation in vitro with a pool of peptides derived from the SI subunit (Figure 2). All four doses were effective. Adjuvant alone (dose = 0 pg) did not induce a measurable response. (3) Splenic T cells, both CD4+ (Tables 2 and 4) and CD8+ (Tables 3 and 5) that became activated upon restimulation in vitro with the peptide pool. Activation was indicated by expression of CD25 (Tables 4 and 5) and CD69 (Tables 2 and 3). For three of the four parameters, only the two highest doses (3 pg, 10 pg) induced activation. Adjuvant alone (dose = 0 pg) did not induce T cell activation. These results demonstrate that immunization with BVX-0320, a hapten (DNP)-modified SI subunit of the spike protein, induced both antibody and T cell responses against the unmodified, native spike protein.
Table 2: Percent Activated Splenic CD4+, CD69+ T Cells After Two Doses of BVX-0320
Cells were left unstimulated or stimulated with a pool of peptides derived from the SARS-Cov-2 spike protein, SI subunit. The differences between unstimulated and peptide-stimulated spleen cells were analyzed by paired t-test.
Dose = 0: QS21 only Dose = 3 pg
Unstim Peptide Unstim Peptide
19.3% 19.5% 22.1% 25.0%
21.2% 22.2% 22.5% 24.8%
35.8% 39.9% 21.0% 39.5%
10.4% 10.3% 23.3% 27.1%
9.0% 10.6% 18.3% 22.0% p = NS 12.2% 15.9%
15.5% 19.8% p = 0.021
Dose = 0.3 pg Dose = 10 pg
Unstim Peptide Unstim Peptide
17.6% 16.5% 24.2% 27.6%
23.4% 25.4% 17.5% 25.4%
30.1% 23.7% 17.2% 26.3%
20.2% 33.2% 16.2% 21.7%
19.1% 19.5% 12.0% 19.0%
25.2% 33.3% 13.5% 14.2%
16.5% 21.5% p = 0.004 p = NS
Dose = 1 pg
Unstim Peptide
13.4% 14.3%
15.6% 16.1%
19.6% 29.6%
14.1% 28.1%
17.8% 17.5% p = NS Table 3: Percent Activated Splenic CD8+, CD69+ T Cells After Two Doses of BVX-0320
Cells were left unstimulated or stimulated with a pool of peptides derived from the SARS-Cov-2 spike protein, SI subunit. The differences between unstimulated and peptide-stimulated spleen cells were analyzed by paired t-test.
Dose = 0: QS21 only Dose = 3 pg
Unstim Unstim Unstim Peptide
26.9% 24.1% 24.1% 27.9%
38.5% 29.5% 29.5% 31.9%
63.3% 38.7% 38.7% 48.0%
18.6% 44.1% 44.1% 46.1%
9.8% 20.5% 20.5% 21.0%
19.4% 19.4% 20.1%
27.1% 27.1% 26.7%
Dose = 0.3 pg p = 0.04
Unstim Peptide Dose = 10 pg
23.6% 22.6% Unstim Peptide
31.9% 33.4% 24.8% 27.1%
47.8% 37.8% 27.3% 33.4%
31.5% 37.2% 20.9% 27.3%
24.1% 24.8% 20.6% 28.2%
32.2% 36.3% 22.7% 27.0%
32.5% 35.5% 17.7% 19.1% p = NS p = 0.003
Dose = 1 pg
Unstim Peptide
15.3% 16.8%
20.3% 21.1%
35.8% 47.1%
27.9% 35.2%
12.1% 11.7% p = NS
Table 4: Percent Activated Splenic CD4+, CD25+ T Cells After Two Doses of BVX-0320
Cells were left unstimulated or stimulated with a pool of peptides derived from the SARS-Cov-2 spike protein, SI subunit. The differences between unstimulated and peptide-stimulated spleen cells were analyzed by paired t-test.
Dose = 0 pg Dose = 3 pg
Unstim Peptide Unstim Peptide
4.7% 4.1% 4.0% 4.4%
3.5% 3.3% 4.3% 4.7%
2.3% 2.2% 1.4% 4.4%
1.9% 1.9% 2.4% 3.7%
3.1% 2.8% 1.9% 2.5% p = NS 0.9% 1.6%
1.1% 1.7%
Dose = 0.3 pg p = 0.016
Unstim Peptide
3.7% 2.9% Dose = 10 pg
2.8% 2.3% Unstim Peptide
2.5% 3.6% 3.0% 3.9%
1.1% 1.9% 2.5% 4.1%
3.1% 3.2% 2.5% 4.1%
1.4% 2.2% 2.3% 3.2%
1.3% 2.5% 1.0% 2.1% p = NS 1.3% 1.3% p = 0.005
Dose = 1 pg
Unstim Peptide
2.5% 2.5%
4.1% 4.0%
1.9% 4.0%
1.2% 4.1%
0.9% 0.8% p = NS
Table 5: Percent Activated Splenic CD8+, CD25+ T Cells After Two Doses of BVX-0320
Cells were left unstimulated or stimulated with a pool of peptides derived from the SARS-Cov-2 spike protein, SI subunit. The differences between unstimulated and peptide-stimulated spleen cells were analyzed by paired t-test.
Dose = 0 pg Dose = 3 pg
Unstim Peptide Unstim Peptide
0.16% 0.23% 0.19% 0.46%
0.22% 0.15% 0.17% 0.27%
0.57% 0.42% 0.06% 0.78%
0.17% 0.19% 0.00% 0.22%
0.14% 0.21% 0.04% 0.21% p = NS 0.12% 0.18%
0.27% 0.18%
Dose = 0.3 pg p = 0.038
Unstim Peptide
0.20% 0.19% Dose = 10 pg
0.18% 0.14% Unstim Peptide
0.00% 0.30% 0.20% 0.36%
0.13% 0.28% 0.28% 0.33%
0.20% 0.25% 0.35% 0.33%
0.26% 0.81% 0.18% 0.35%
0.24% 0.35% 0.21% 0.36% p = .044 0.09% 0.17% p = 0.012
Dose = 1 pg
Unstim Peptide
0.07% 0.10%
0.24% 0.32%
0.21% 0.55%
0.25% 0.50%
0.04% 0.08% p = .038
[0085] Although the present invention has been described in connection with specific selected embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in biochemistry and biotechnology or related fields are intended to be within the scope of the following claims.

Claims

1. A method of immunizing a human subject against multiple species of sarbecovirus comprising:
(a) immunizing a human subject against SARS-CoV-1, and
(b) immunizing a human subject against SARS-CoV-2, wherein the human subject has never been infected with, nor immunized against, SARS-CoV-1 and SARS-CoV-2.
2. The method of claim 1, wherein the immunizing comprises:
(i) administration to the human subject of an immunogenic composition comprising:
(a) an S protein or fragment thereof from SARS-CoV-1 and at least one pharmaceutically acceptable carrier, or
(b) a polynucleotide encoding an S protein or fragment thereof from SARS-CoV-1 and at least one pharmaceutically acceptable carrier; and
(ii) administration to the human subject of an immunogenic composition comprising:
(a) an S protein or fragment thereof from SARS-CoV-2 and at least one pharmaceutically acceptable carrier, or
(b) a polynucleotide encoding an S protein or fragment thereof from SARS-CoV-2 and at least one pharmaceutically acceptable carrier.
2. The method of claim 1, wherein the immunizing against SARS-CoV-1 occurs first, followed by a second immunizing against SARS-CoV-2.
3. The method of claim 1, wherein the immunizing against SARS-CoV-2 occurs first, followed by a second immunizing against SARS-CoV-1.
4. A method of immunizing a human subject against multiple species of sarbecovirus comprising:
(i) administering to the human subject an immunogenic composition comprising:
(a) an S protein or fragment thereof from SARS-CoV-1 and at least one pharmaceutically acceptable carrier, or
(b) a polynucleotide or fragment thereof encoding an S protein from SARS-CoV-1, wherein the human subject has already been immunized against SARS-CoV-2 and/or been infected with SARS-CoV-2.
33
5. The method of any previous claim, wherein the fragment thereof comprises the receptor binding domain (RBD) of the S protein.
6. The method of any previous claim, wherein the fragment thereof comprises the SI domain of the S protein.
7. The method of any previous claim, wherein the S protein from SARS-CoV-1 comprises the amino acid sequence of SEQ ID NO:4.
8. The method of any previous claim, wherein the S protein from SARS-CoV-2 comprises the amino acid sequence of SEQ. ID NO:2.
9. The method of any previous claim, wherein the species of sarbecovirus are selected from the group consisting of SARS-CoV-1, Bat CoV WIV17, Bat CoV RsSHC0147, SARS-CoV-2, SARS-CoV-2 Alpha variant, SARS-CoV-2 Beta variant, SARS-CoV-2 Delta variant, Bat CoV RaTG135, Pangolin CoV GD-16, Pangolin GX- P5L6.
10. The method of any preceding claim, wherein the S protein or fragment thereof is haptenized.
11. The method of claim 10, wherein the hapten is selected from the group consisting of trinitrochlorobenzene (TNCB), 2,4-difluoronitrobenzene (DNFB), Niodoacetyl-N'-(5-sulfonic-l- naphthyl)ethylenediamine (AED), sulfanilic acid (SA), trinitrophenol (TNP), and 2,4,6- trinitrobenzenesulfonic acid (TNBS).
12. The method of any preceding claim, wherein the immunogenic composition further comprises an adjuvant.
13. The method of claim 12, wherein the adjuvant is an oil in water emulsion.
14. The method of claim 13, wherein the oil is squalene oil (MF59).
15. The method of claim 12, wherein the adjuvant is selected from the group consisting of: AS01, AS03,
AS04, amorphous aluminum hydroxyphosphate sulfate (AAHS), aluminum hydroxide, aluminum phosphate, potassium aluminum sulfate (Alum), squalene (MF59), saponin (QS-21), biphosphanate, CpG-1018, and combinations thereof.
16. The method of any previous claim wherein the S protein or fragment thereof in the immunogenic composition is present in an amount from 30 to 300 pg.
34
17. The method of claim 2 or 3, wherein the second immunization occurs at least three weeks after the first immunization.
18. The method of any previous claim, the method further comprising administering at least one booster injection of the immunogenic composition about six months after the first injection.
19. The method of any of previous claim, wherein the immunogenic composition is administered until a cross-reactive neutralizing antibody response against multiple species of sarbecovirus is detected in the human subject.
20. The method of any of claims 4-19, wherein the human subject has been previously infected with SARS-CoV-2 and recovered from Covid-19.
21. The method of any of claims 4-19, wherein the human subject has previously been immunized against SARS-CoV-2.
22. The method of claim 21, wherein the SARS-CoV-2 immunization is an mRNA vaccine, inactivated virus vaccine, viral vector vaccine, or protein subunit vaccine.
23. The method of claim 22, wherein the mRNA vaccine is BNT162b2 or mRNA-1273.
24. The method of claim 18, wherein the viral vector vaccine is Ad26.COV2.S COVID-19 or ChAdOxl.
25. The method of any previous claim, wherein the human subject is at risk for a coronavirus infection.
PCT/US2022/077737 2021-10-07 2022-10-07 Methods of immunization against coronavirus WO2023060220A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163253149P 2021-10-07 2021-10-07
US63/253,149 2021-10-07

Publications (1)

Publication Number Publication Date
WO2023060220A1 true WO2023060220A1 (en) 2023-04-13

Family

ID=85803767

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/077737 WO2023060220A1 (en) 2021-10-07 2022-10-07 Methods of immunization against coronavirus

Country Status (1)

Country Link
WO (1) WO2023060220A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021019102A2 (en) * 2019-08-01 2021-02-04 Acm Biolabs Pte Ltd A method of eliciting an immune response by administering a population of polymersomes having an associated antigen together with a population of polymersomes having an associated adjuvant as well as compositions comprising the two populations of polymersomes
WO2021178971A1 (en) * 2020-03-06 2021-09-10 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Vaccines against sars-cov-2 and other coronaviruses

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021019102A2 (en) * 2019-08-01 2021-02-04 Acm Biolabs Pte Ltd A method of eliciting an immune response by administering a population of polymersomes having an associated antigen together with a population of polymersomes having an associated adjuvant as well as compositions comprising the two populations of polymersomes
WO2021178971A1 (en) * 2020-03-06 2021-09-10 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Vaccines against sars-cov-2 and other coronaviruses

Similar Documents

Publication Publication Date Title
JP7187237B2 (en) Vaccine against hepatitis B virus
AU570929B2 (en) Broad spectrum influenza antisera
AU2009259964B2 (en) Compositions and methods for treating influenza
Levi et al. Intranasal immunization of mice against influenza with synthetic peptides anchored to proteosomes
CA2593746A1 (en) Compositions of influenza viral proteins and methods of use thereof
US20120009212A1 (en) Conjugates utilizing platform technology for stimulating immune response
SK7762000A3 (en) Vaccines with an ltb adjuvant
US20090175903A1 (en) Measles subunit vaccine
CN104507496B (en) Including the immunogenic compound with the HIV GP41 peptides of CRM197 carrier protein couplets
CA2727866A1 (en) Novel peptide adjuvant for influenza vaccination
Glück Intranasal immunization against influenza
US20230149534A1 (en) Haptenized coronavirus spike proteins
WO2023060220A1 (en) Methods of immunization against coronavirus
US20230263881A1 (en) Vaccine compositions for SARS-related coronaviruses and methods of use
JPWO2004087767A1 (en) Polypeptide
TW202308685A (en) Coronavirus and influenza compositions and methods for using them
EP3981426A1 (en) Adjuvant based on peptide nucleic acid
Uchida et al. Application of surface-linked liposomal antigens to the development of vaccines that induce both humoral and cellular immunity
US20230146694A1 (en) Peptide immunogens targeting pituitary adenylate cyclase-activating peptide (pacap) and formulations thereof for prevention and treatment of migraine
US20110097418A1 (en) Compositions and methods for treating influenza
EA021191B1 (en) NOVEL gp41 ANTIGENS
CN117957017A (en) Vaccine formulations comprising recombinant overlapping peptides and native proteins
WIESMÜLLER et al. WG BESSLER, W. BAIER, M. HUBER, P. HOFFMANN, L. HEINEVETTER
US20140154282A1 (en) Conjugates utilizing platform technology for stimulating immune response

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: 22879510

Country of ref document: EP

Kind code of ref document: A1