WO2023159082A2 - Nanotechnology based intranasal vaccine for covid-19 comprising chitosan - Google Patents

Nanotechnology based intranasal vaccine for covid-19 comprising chitosan Download PDF

Info

Publication number
WO2023159082A2
WO2023159082A2 PCT/US2023/062681 US2023062681W WO2023159082A2 WO 2023159082 A2 WO2023159082 A2 WO 2023159082A2 US 2023062681 W US2023062681 W US 2023062681W WO 2023159082 A2 WO2023159082 A2 WO 2023159082A2
Authority
WO
WIPO (PCT)
Prior art keywords
vaccine
cov
sars
subject
antigen
Prior art date
Application number
PCT/US2023/062681
Other languages
French (fr)
Other versions
WO2023159082A3 (en
Inventor
Renukaradhya J. Gourapura
Ganesh Yadagiri
Scott Kenney
Jennifer SCHROCK
Patricia BOLEY
Kaissar TABYNOV
Kairat TABYNOV
Tlektes YESPOLOV
Original Assignee
Ohio State Innovation Foundation
Kazakh National Agrarian Research University
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 Ohio State Innovation Foundation, Kazakh National Agrarian Research University filed Critical Ohio State Innovation Foundation
Publication of WO2023159082A2 publication Critical patent/WO2023159082A2/en
Publication of WO2023159082A3 publication Critical patent/WO2023159082A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • A61K47/6939Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being a polysaccharide, e.g. starch, chitosan, chitin, cellulose or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/543Mucosal route intranasal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • 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
    • C12N2770/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • VN virus-neutralizing antibodies
  • SARS-CoV-2-specific central memory CD4 and effector memory CD8 T cells were detected in the peripheral blood of COVID-19 patients after 14 days of infection (Thevarajan 2020; Weiskopf 2020) leading to a proposition that detection of memory T cells may be the appropriate readout of effective COVID vaccines (Melgaco 2020).
  • protective immunity to viral infections in vaccinated and convalescent individuals with weak antibody production has been attributed to memory CD8 T cells (Bauer 2006).
  • COVID vaccines that can induce both antibodies and T cell responses.
  • Protective mucosal immune responses are most effectively induced by mucosal immunization through oral, nasal, rectal or vaginal routes, but the vast majority of vaccines in use today are administered by injection (Neutra 2006).
  • respiratory pathogens such as SARS-CoV-2
  • induction of mucosal antibodies is critical.
  • Mucosal vaccines have gained attention due to their ability to induce broad cross-reactive responses (Manocha 2005; Lisa Schnirring, 2018).
  • RNA viruses are characterized by a high mutation rate, up to a million times higher than that of their hosts.
  • SARS-CoV-2 being an RNA virus
  • S gene Seretti 2020; Harvey 20231
  • injectable vaccines elicit systemic IgG response and protect against homologous strains, they induce poor secretory (s) sIgA antibodies needed to prevent virus replication and load in the respiratory tract (Cox 2004).
  • vaccines delivered intranasally (IN) can induce sIgA in the respiratory tract that can neutralize homologous and genetic variants by binding to newly synthesized viral proteins in mucosal epithelial cells (Mazanec 1995; Suzuki 2015).
  • T cells as described above, play an indispensable role in the mediation of long-term protection, but such an effort requires methods to elicit antigen-specific T cell responses in vaccine settings.
  • Soluble and subunit antigens that are otherwise poor immunogens become highly immunogenic when delivered after entrapping in nanoparticle (NP) (Bacon 2000; Bertram 2010; Dhakal 2018).
  • nanoparticles ⁇ 500 nm
  • DCs dendritic cells
  • macrophages (0 ⁇ V)
  • B and T cells via cross-linking Wang 2012; Heit 2007; Schliehe 2011).
  • the virus antigens when entrapped or encapsulated within the NP are protected from degradation, especially when delivered to mucosal surfaces (Dhakal 2017; Hiremath 2016; Danhier 2012).
  • Advantages of intranasal immunization to avoid interference by preexisting antibodies in the host has been studied in rodents and pigs (Renu 2021; Zhang 2016).
  • Induction of cell mediated cross-protective immune response to killed/subunit antigens is possible when delivered through nanoparticles (Dhakal 2017; Zhang 2016).
  • Coadministration of antigen and adjuvant in the nanoparticle is possible which help to improve the immunity and vaccine dose sparing. What is needed in the art is delivery of a vaccine cargo targeting the immune cells using nanoparticles (Renu 2021; Han 2020).
  • S-RBD receptor binding domain of the spike protein
  • N nucleocapsid
  • M matrix
  • composition comprising SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof, wherein the nanoparticle comprises chitosan.
  • a vaccine comprising SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof in a pharmaceutically acceptable carrier, wherein the nanoparticle comprises chitosan.
  • the vaccine comprises a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof; wherein the vaccine further comprises a pharmaceutically acceptable carrier, wherein the nanoparticle comprises chitosan.
  • Disclosed further is a method of preventing or lessening the severity of symptoms associated with SARS-CoV-2 infection in a subject comprising administering to the subject a vaccine, wherein the vaccine comprises a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof; wherein the vaccine further comprises a pharmaceutically acceptable carrier, wherein the nanoparticle comprises chitosan.
  • S spike protein
  • N nucleocapsid protein
  • FIG. 1A-1D shows detection of replicating infectious SARS-CoV-2 load in the respiratory tract of vaccinated and virus challenged ferrets.
  • SARS-CoV-2 RBD spike
  • N nucleocapsid
  • Figure 2A-2D shows detection of SARS-CoV-2 RNA load in the respiratory tract of vaccinated and virus challenged ferrets.
  • Figure 3A-3F shows detection of mRNA of immune genes in the respiratory tract of vaccinated and virus challenged ferrets.
  • One-year-old male ferrets were vaccinated twice at three weeks interval intranasally with SARS-CoV-2 RBD (spike) and nucleocapsid (N) antigens and monosodium urate crystal adjuvant and challenged with SARS-CoV-2 intranasally.
  • SARS-CoV-2 RBD spike
  • N nucleocapsid
  • the samples of respiratory tract tissues expressing immune genes: IFNa in (3 A.) Lungs and (3B) Nasal turbinate at DPC 7; MCP1 in (3C) Lungs at DPC 7 and (3D & 3E) Nasal turbinate at DPC 7 and DPC 14; and (3F) IL-17 in Lungs at DPC 14 were measured by qRT-PCR.
  • the standard double delta. Ct values were normalized with housekeeping gene GAPDH and the values of mock uninfected ferret respective tissue values were subtracted from the experimental group values. Each bar is the mean of 3 animals ⁇ SEM. Asterisks refers to significant (*p ⁇ 0.05) difference between the indicated groups.
  • Figure 4A-4B shows scanning electron microscope analysis of (4A) mannose- conjugated chitosan nanoparticles and (4B) mannose-conjugated chitosan nanoparticles entrapped with RBD protein.
  • Figure 5A-5E shows antibody response in BALB/c mice after vaccination.
  • Antigen-specific serum (5A) and lung (5B) IgA antibodies in mice at 21 days after prime and booster intranasal immunization with RBD-based nanoparticle vaccine formulations and intramuscular immunization with Alum adjuvanted vaccine.
  • Mannose- conjugated chitosan-NP-based vaccine formulations, including those with CpG adjuvant (NP-CpG) were administered in doses containing 5, 2.5, and 1.25 ⁇ g RBD protein.
  • Viral neutralizing antibody levels with the wild-type of SARS-CoV-2 are presented as geometric mean titers with 95% confidence intervals. Differences in antibody levels between animal groups were assessed using Tukey's multiple comparisons test or t-test. P ⁇ 0.05 was considered statistically significant. *P ⁇ 0.05, **P ⁇ 0.01, ***P ⁇ 0.001, and ****p ⁇ 0.0001.
  • Figure 6A-6H shows antigen-specific cytokine production in the splenocyte suspension of BALB/c mice at day 21 post booster IN immunization with RBD-based NP-vaccine formulations and control IM immunization with Alum adjuvanted vaccine.
  • Mannose-conjugated chitosan-NP-based vaccine formulations with and without including CpG adjuvant were administered in doses containing 5, 2.5, and 1.25 ⁇ g RBD protein for (6 A) IFN- ⁇ ; (6B) IL-2; (6C) IL- 17 A; (6D) TNF-a; (6E) IL- 4; (6F) IL-6; (6G) IL-5; and (6H) IL-10.
  • Figure 7A-7G shows efficacy of RBD-based NP-vaccine in Syrian hamsters for protection against wild-type SARS-CoV-2 (D614G) infection and virus transmission.
  • Mannose-conjugated chitosan-NP-based vaccine formulations including the NP-CpG vaccine were administered with a dose containing 5 ⁇ g RBD protein.
  • an “immunogenic composition” is a composition of matter suitable for administration to a human or animal subject (e.g., in an experimental setting) that is capable of eliciting a specific immune response, e.g., against a pathogen, such as SARS-CoV-2.
  • an immunogenic composition includes one or more antigens (for example, whole purified virus or antigenic subunits, e.g., polypeptides, thereof) or antigenic epitopes.
  • -An immunogenic composition can also include one or more additional components capable of eliciting or enhancing an immune response, such as an excipient, carrier, and/or adjuvant.
  • immunogenic compositions are administered to elicit an immune response that protects the subject against symptoms or conditions induced by a pathogen.
  • symptoms or disease caused by a pathogen is prevented (or treated, e.g., reduced or ameliorated) by inhibiting replication of the pathogen following exposure of the subject to the pathogen.
  • immunogenic composition will be understood to encompass compositions that are intended for administration to a subj ect or population of subj ects for the purpose of eliciting a protective or palliative immune response against the virus (that is, vaccine compositions or vaccines).
  • an “antigen” is a compound, composition, or substance that can stimulate the production of antibodies and/or a T cell response in an animal, including compositions that are injected, absorbed or otherwise introduced into an animal.
  • the term “antigen” includes all related antigenic epitopes.
  • the term “epitope” or “antigenic determinant” refers to a site on an antigen to which B and/or T cells respond.
  • the “dominant antigenic epitopes” or “dominant epitope” are those epitopes to which a functionally significant host immune response, e.g., an antibody response or a T-cell response, is made.
  • the dominant antigenic epitopes are those antigenic moieties that when recognized by the host immune system result in protection from disease caused by the pathogen.
  • T-cell epitope refers to an epitope that when bound to an appropriate MHC molecule is specifically bound by a T cell (via a T cell receptor).
  • a “B-cell epitope” is an epitope that is specifically bound by an antibody (or B cell receptor molecule).
  • An antigen can also affect the innate immune response.
  • An “immune response” is a response of different cell types of the immune system, such as a B cell, T cell, or monocyte, to a stimulus.
  • An immune response can be a B cell response, which results in the production of specific antibodies, such as antigen specific neutralizing antibodies.
  • An immune response can also be a T cell response, such as a CD4+ T cell response and/or a CD8+ T cell response. In some cases, the response is specific for a particular antigen (that is, an “antigen-specific response”).
  • An immune response can also include the innate response. If the antigen is derived from a pathogen, the antigen-specific response is a “pathogen-specific response.”
  • a “protective immune response” is an immune response that inhibits a detrimental function or activity of a pathogen, reduces infection by a pathogen, or decreases symptoms (including death) that result from infection by the pathogen.
  • a protective immune response can be measured, for example, by the inhibition of viral replication or plaque formation in a plaque reduction assay or virus-neutralization assay, or by measuring resistance to pathogen challenge in vivo.
  • the immunogenic compositions disclosed herein are suitable for preventing, ameliorating and/or treating disease caused by infection of the virus.
  • the abbreviation “KAg” stands for killed antigen and represents the killed or inactivated virus.
  • the inactivated virus comprises one or more immunogenic viral proteins and therefore the inactivated virus can be considered a killed antigen.
  • NP-KAg stands for nanoparticle-killed antigen. This represents the nanoparticle encapsulated inactivated swine influenza virus.
  • virus-like particle refers to a non- replicating, viral shell.
  • VLPs are generally composed of one or more viral proteins associated with viral surface capsid structure. VLPs can form spontaneously upon recombinant expression of the protein in an appropriate expression system. VLPs, when administered to an animal, can be immunogenic and thus can cause a protective or therapeutic immune response in the animal. Methods for producing VLPs are generally known in the art and discussed more fully below. The presence of VLPs following recombinant expression of viral proteins can be detected using conventional techniques known in the art, such as by electron microscopy, biophysical characterization, and the like. See, e.g., Baker et al., Biophys. J.
  • VLPs can be isolated by density gradient centrifugation and/or identified by characteristic density banding.
  • treatment refers to the medical management of a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder.
  • This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder.
  • this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder.
  • palliative treatment that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder
  • preventative treatment that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder
  • supportive treatment that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder.
  • Optional or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
  • An "increase" can refer to any change that results in a greater amount of
  • An increase can be any individual, median, or average increase in a condition, symptom, activity, composition in a statistically significant amount.
  • the increase can be a 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% increase so long as the increase is statistically significant.
  • a "decrease” can refer to any change that results in a smaller amount of a symptom, disease, composition, condition, or activity.
  • a substance is also understood to decrease the genetic output of a gene when the genetic output of the gene product with the substance is less relative to the output of the gene product without the substance. Also for example, a decrease can be a change in the symptoms of a disorder such that the symptoms are less than previously observed.
  • a decrease can be any individual, median, or average decrease in a condition, symptom, activity, composition in a statistically significant amount.
  • the decrease can be a 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% decrease so long as the decrease is statistically significant.
  • “Inhibit,” “inhibiting,” and “inhibition” mean to decrease an activity, response, condition, disease, or other biological parameter. This can include but is not limited to the complete ablation of the activity, response, condition, or disease. This may also include, for example, a 10% reduction in the activity, response, condition, or disease as compared to the native or control level.
  • the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.
  • reduce or other forms of the word, such as “reducing” or “reduction,” is meant lowering of an event or characteristic (e.g., tumor growth). It is understood that this is typically in relation to some standard or expected value, in other words it is relative, but that it is not always necessary for the standard or relative value to be referred to.
  • “reduces tumor growth” means reducing the rate of growth of a tumor relative to a standard or a control.
  • prevent or other forms of the word, such as “preventing” or “prevention,” is meant to stop a particular event or characteristic, to stabilize or delay the development or progression of a particular event or characteristic, or to minimize the chances that a particular event or characteristic will occur. Prevent does not require comparison to a control as it is typically more absolute than, for example, reduce. As used herein, something could be reduced but not prevented, but something that is reduced could also be prevented. Likewise, something could be prevented but not reduced, but something that is prevented could also be reduced. It is understood that where reduce or prevent are used, unless specifically indicated otherwise, the use of the other word is also expressly disclosed.
  • subject refers to any individual who is the target of administration or treatment.
  • the subject can be a vertebrate, for example, a mammal.
  • the subject can be human, non-human primate, bovine, avian, equine, porcine, canine, or feline.
  • the subject can also be a guinea pig, rat, hamster, rabbit, mouse, or mole.
  • the subject can be a human or veterinary patient.
  • patient refers to a subject under the treatment of a clinician, e.g., physician.
  • the term “therapeutically effective” refers to the amount of the composition used is of sufficient quantity to ameliorate one or more causes or symptoms of a disease or disorder. Such amelioration only requires a reduction or alteration, not necessarily elimination.
  • Biocompatible generally refers to a material and any metabolites or degradation products thereof that are generally non-toxic to the recipient and do not cause significant adverse effects to the subject.
  • “Comprising” is intended to mean that the compositions, methods, etc. include the recited elements, but do not exclude others.
  • Consisting essentially of'' when used to define compositions and methods shall mean including the recited elements, but excluding other elements of any essential significance to the combination. Thus, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives, and the like.
  • Control is an alternative subject or sample used in an experiment for comparison purposes.
  • a control can be "positive” or “negative.”
  • a negative control can be an untreated or mock treated control.
  • a positive control can be a control with a known positive response.
  • a “pharmaceutically acceptable” component can refer to a component that is not biologically or otherwise undesirable, i.e., the component may be incorporated into a pharmaceutical formulation provided by the disclosure and administered to a subject as described herein without causing significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the formulation in which it is contained.
  • the term When used in reference to administration to a human, the term generally implies the component has met the required standards of toxicological and manufacturing testing or that it is included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug Administration.
  • “Pharmaceutically acceptable carrier” (sometimes referred to as a “carrier”) means a carrier or excipient that is useful in preparing a pharmaceutical or therapeutic composition that is generally safe and non-toxic and includes a carrier that is acceptable for veterinary and/or human pharmaceutical or therapeutic use.
  • carrier or “pharmaceutically acceptable carrier” can include, but are not limited to, phosphate buffered saline solution, water, emulsions (such as an oil/water or water/oil emulsion) and/or various types of wetting agents.
  • carrier encompasses, but is not limited to, any excipient, diluent, filler, salt, buffer, stabilizer, solubilizer, stabilizer, or other material well known in the art for use in pharmaceutical formulations and as described further herein.
  • “Pharmacologically active” (or simply “active”), as in a “pharmacologically active” derivative or analog, can refer to a derivative or analog (e.g., a salt, ester, amide, conjugate, metabolite, isomer, fragment, etc.) having the same type of pharmacological activity as the parent compound and approximately equivalent in degree.
  • “Therapeutic agent” refers to any composition that has a beneficial biological effect.
  • Beneficial biological effects include both therapeutic effects, e.g., treatment of a disorder or other undesirable physiological condition, and prophylactic effects, e.g., prevention of a disorder or other undesirable physiological condition (e.g., a non- immunogenic cancer).
  • the terms also encompass pharmaceutically acceptable, pharmacologically active derivatives of beneficial agents specifically mentioned herein, including, but not limited to, salts, esters, amides, proagents, active metabolites, isomers, fragments, analogs, and the like.
  • therapeutic agent refers to an amount that is effective to achieve a desired therapeutic result.
  • a desired therapeutic result is the control of type I diabetes.
  • a desired therapeutic result is the control of obesity.
  • Therapeutically effective amounts of a given therapeutic agent will typically vary with respect to factors such as the type and severity of the disorder or disease being treated and the age, gender, and weight of the subject.
  • the term can also refer to an amount of a therapeutic agent, or a rate of delivery of a therapeutic agent (e.g., amount over time), effective to facilitate a desired therapeutic effect, such as pain relief.
  • the precise desired therapeutic effect will vary according to the condition to be treated, the tolerance of the subject, the agent and/or agent formulation to be administered (e.g., the potency of the therapeutic agent, the concentration of agent in the formulation, and the like), and a variety of other factors that are appreciated by those of ordinary skill in the art.
  • a desired biological or medical response is achieved following administration of multiple dosages of the composition to the subject over a period of days, weeks, or years.
  • intranasal(ly) refers to application of the compositions of the present invention to the surface of the skin and mucosal cells and tissues of the nasal passages, e.g., nasal mucosa, sinus cavity, nasal turbinates, or other tissues and cells which line the nasal passages.
  • drug composition and “drug”, “vaccinal composition” and “vaccine” and “vaccine composition” and “drug-vaccine composition” and “drug-vaccine dual agent” and “therapeutic composition” and “therapeutic-immunologic composition” cover any composition that induces protection against a pathogen.
  • the protection may be due to an inhibition or prevention of infection by a pathogen.
  • the protection may be induced by an immune response against the antigen(s) of interest, or which efficaciously protects against the antigen; for instance, after administration or injection into the subject, elicits a protective immune response against the targeted antigen or immunogen or provides efficacious protection against the antigen or immunogen expressed from the inventive adenovirus vectors of the invention.
  • various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this pertains. The references disclosed are also individually and specifically incorporated by reference herein for the material contained in them that is discussed in the sentence in which the reference is relied upon.
  • composition comprising SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises at least a spike protein (S) or an antigenic fragment thereof, and at least a nucleocapsid protein (N) or an antigenic fragment thereof.
  • SARS-CoV-2 is a single-stranded RNA-enveloped virus.
  • the capsid consists of the nucleocapsid protein (N) and this is further surrounded by a membrane, that contains three proteins: the membrane protein (M) and the envelope protein (E), which are involved in the virus budding process, and the spike glycoprotein (S), which is a key player in binding host receptor and mediating membrane fusion and virus entry into host cells.
  • M membrane protein
  • E envelope protein
  • S spike glycoprotein
  • a large number of glycosylated S proteins cover the surface of SARS-CoV-2 and bind to the host cell receptor angiotensin-converting enzyme 2 (ACE2), mediating viral cell entry.
  • ACE2 angiotensin-converting enzyme 2
  • TM protease serine 2 (TMPRSS2), a type 2 TM serine protease located on the host cell membrane, promotes virus entry into the cell by activating the S protein.
  • TMPRSS2 TM protease serine 2
  • the viral RNA is released, polyproteins are translated from the RNA genome, and replication and transcription of the viral RNA genome occur via protein cleavage and assembly of the replicase–transcriptase complex.
  • Viral RNA is replicated, and structural proteins are synthesized, assembled, and packaged in the host cell, after which viral particles are released.
  • Antigens The present invention makes use of not only the spike protein, which is commonly used in vaccines, but also the N protein. This combination of proteins provides a powerful synergistic effect.
  • the S antigen alone is the basis for the Pfizer/BioNTek vaccine, Moderna vaccine, Johnson & Johnson’s (J&J) Janssen vaccine, AstraZeneca/Oxford vaccine, CanSino vaccine, Sputnik V vaccine, Novavax vaccine, and others. Utilizing both antigens provides better and broader protection against variant viruses coming out of SARS-CoV-2 in multiple tissues/organs/cells. Furthermore, because viral N protein contributes to forming helical ribonucleoproteins during the packaging of the RNA genome, regulating viral RNA synthesis during replication and transcription and modulating metabolism in infected subjects, it is a highly efficacious antigen when combined with the S protein.
  • antigen protein or antigenic fragment thereof is meant that either the entire antigen is included in the composition (such as the “S” or “N” protein), or a fragment of the antigen is included.
  • a fragment it is contemplated herein that the fragment will be of sufficient length to induce an immune response when the fragment is exposed to a subject who would have an immune response to the full-length protein.
  • the fragment can be 10, 20, 30, 40, 50, 60, 70, 80, 90, 91, 92, 93, 94, 95, 96, 97. 98, or 99% identical to the protein itself.
  • the fragment can be truncated at either the C terminal, N terminal, or both, and can comprise one or more variations with respect to the full-length antigen.
  • the antigen or antigenic fragment used in the compositions disclosed herein can be identical to a known antigen, or can be a derivative (variation) thereof.
  • the derivative can comprise substitutions, deletions, or insertions in the amino acid sequence in comparison to the known, or native, sequence of an antigen. These variations can render the amino acid sequence of the derivative antigen different than the known antigen, yet still capable of eliciting an immune response in a subject.
  • the derivative can have 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36 ,37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80,81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, or more mutated amino acid residues compared to the original, known, or native sequence.
  • the composition can comprise one or more antigenic proteins or fragments thereof of the spike (S) protein.
  • S protein is a glycosylated type I membrane protein that consists of two subunits, S1 and S2.
  • the S protein exists in a trimeric pre- fusion form that is later cleaved by a host furin protease into the two subunits S1 and S2.
  • the N-terminal S1 subunit contains the receptor-binding domain (RBD), which mediates binding to the host cell receptor, namely the angiotensin converting enzyme 2 (ACE2) for both SARS-CoV and SARS-CoV-2.
  • RBD receptor-binding domain
  • ACE2 angiotensin converting enzyme 2
  • the composition can be bivalent, or can be multivalent, meaning it can comprise two, three, four, five, or more antigens or fragments thereof. Multiple antigenic proteins or fragments thereof in the same composition can all come from SARS-CoV-2. These multiple antigenic proteins or fragments thereof can be from the same variant, or can be from different variants. Furthermore, the antigenic proteins or fragments thereof can be from different viruses. This is discussed in more detail below.
  • the antigenic protein or fragments of the composition comprises the S protein or an antigenic fragment thereof.
  • the spike protein antigen of the composition can comprise the S1 subunit, the S2 subunit, or both.
  • An example of the spike protein can be found in SEQ ID NO: 1.
  • the spike protein antigen can comprise the receptor-binding domain (RBD).
  • the RBD can be found, by way of example, in residues 336-516 of SEQ ID NO: 1.
  • the RBD is a short immunogenic fragment from a virus that binds to a specific endogenous receptor sequence to gain entry into host cells. Specifically, these refer to a part of the spike glycoprotein (S- domain) which is needed to interact with endogenous receptors to facilitate membrane fusion and delivery to the cytoplasm.
  • the S-domain is also the site of neutralizing antibodies.
  • the antigenic protein or fragments of the composition can also comprise the N protein or an antigenic fragment thereof.
  • the N protein plays a multifaceted role in the infection cycle.
  • SARS-CoV-2 the N protein binds to and packages the viral RNA into ribonucleoprotein RNP complexes.
  • the N protein is recruited at the replication-transcription complex by Nsp3 and thus, plays a role in viral genome replication.
  • this interaction involves the N-terminal domains of Nsp3 and the N protein and its function is to guide the viral genome to the newly assembled replication complex.
  • the antigenic protein or fragments thereof of the composition can also comprise the M protein or an antigenic fragment thereof.
  • the M protein is embedded in the viral membrane, through three predicted transmembrane helices. Its role is to drive the assembly of new virions within the host cells. Coronaviruses M proteins oligomerize at the membrane of Golgi-endoplasmic reticulum intermediary compartment and induce apoptosis. S, N and E proteins are then recruited through interaction with the M protein.
  • the antigenic protein or fragments of the composition can also comprise the E protein or an antigenic fragment thereof.
  • the E protein presents one trans-membrane domain and shows oligomerization properties. Interaction between the C-terminus of E and M proteins guides E recruitment to the Golgi-endoplasmic reticulum intermediary compartment, initiating virus budding into host.
  • the composition comprises at least one S protein antigen or fragment thereof, and at least one N protein or fragment thereof.
  • the S protein can comprise all or a fragment of the RBD.
  • the sequence and structure of RBD of S protein can be found in Lan et al. (2020), which is hereby incorporated by reference in its entirety for its disclosure concerning the RBD of S protein.
  • the composition can comprise either the S1 subunit, the S2 subunit, or both.
  • the composition comprises at least two different antigens or fragments thereof from S protein and at least one N protein or fragment thereof.
  • the S1 and S2 proteins can both be included in the composition as separate antigens, as well as the N protein.
  • the composition comprises one or more S proteins or fragments thereof, one or more N proteins or fragments thereof, and at least one other antigen from SARS-CoV-2.
  • This further antigen can be from the M protein, the E protein, or any other proteins associated with SARS-CoV-2.
  • the composition comprises one or more S proteins or fragments thereof, one or more N proteins or fragments thereof, and at least one other antigen from another virus.
  • the further antigen can be from influenza.
  • the composition can comprise the SARS-CoV-2 antigens described above, and can further comprise antigens to one, two, three, four, five, or more other antigens. When more than one additional antigen is provided from another virus, the antigens can be from the same strain or from multiple strains.
  • the multiple antigens can be to different hemagglutinin subtypes. Eighteen different phylogenetically distinct subtypes of HA have emerged in influenza A viruses (H1 to H18), and each can be used to as a distinct antigen in a vaccine.
  • antigens other than those derived from SARS-CoV-2 that can be used in a multivalent vaccine with SARS-CoV-2 antigens include, but are not limited to, influenza virus, cytomegalovirus, avian leukosis-sarcoma virus (ALV), Rous Sarcoma virus (RSV), Mammalian C-type Murine leukemia virus (MLV), Feline leukemia virus (FeLV), simian sarcoma virus (SIS), B-type viruses like Mouse mammary tumor virus (MMTV), D-type viruses like Mason-Pfizer monkey virus (MPMV), Simian AIDS viruses (SRVs), HTLV-BLV group such as Human T-cell leukemia virus (HTLV), Simian T-cell leukemia virus (STLV), bovine leukemia virus (BLV).
  • ABV avian leukosis-sarcoma virus
  • RSV Rous Sarcoma virus
  • MLV Mammalian C-type Murine leukemia virus
  • Lentivirinae comprise Human immunodeficiency virus (HIV-1 and HIV-2), Simian immunodeficiency virus (SIV), Feline immunodeficiency virus (FIV), Visna/maedi virus (MV), Equine infectious anemia virus (EIAV), Caprine arthritis- encephalitis virus (CAEV).
  • HIV-1 and HIV-2 Human immunodeficiency virus
  • SIV Simian immunodeficiency virus
  • FV Feline immunodeficiency virus
  • MV Visna/maedi virus
  • EIAV Equine infectious anemia virus
  • CAEV Caprine arthritis- encephalitis virus
  • Spumavirinae or “Foamy viruses” like Human (HSRV), Simian (SSRV), Feline (FSRV), Bovine (BSRV), Murine (MSRV), endogenous retroviruses (ERV), papilloma virus, respiratory syncytial virus, poliomyelitis virus, pox virus, measles virus, arbor virus, Coxsackie virus, herpes virus, hantavirus, hepatitis virus, Baculovirus, mumps virus, circovirus, vichaivirus, arenavirus, or rotavirus.
  • HSRV Human
  • SSRV Simian
  • Feline Feline
  • BSRV Bovine
  • MSRV Murine
  • ERV endogenous retroviruses
  • papilloma virus papilloma virus
  • respiratory syncytial virus poliomyelitis virus
  • pox virus measles virus
  • arbor virus Coxsackie virus
  • a bacteria may be a member of the genus Neisseria, Aerobacter, Pseudomonas, Porphyromonas, Salmonella, Escherichia, Pasteurella, Shigella, Bacillus, Helibacter, Corynebacterium, Clostridium, Mycobacterium, Yersinia, Staphylococcus; Bordetelia, Brucelia, Vibrio, Streptococcus, Plasmodium, Schisostoma, Candida.
  • Any microbial infections which are present and/or transmitted as Zoonoses, Cyclozoonoses, Metazoonoses, Saprozoonoses, Anthropozoonoses, Zooanthropozoonoses and Amphixenoses, are encompassed by the present invention.
  • the invention in addition to whole pathogens also encompasses a single antigen or a plurality of antigens from such pathogens, e.g., HIV antigens: gp160, gag, pol, Nef, Tat, and Rev; the malaria antigens: CS protein and Sporozoite surface protein 2; the Hepatitis B surface antigens: Pre-S1, Pre-S2, HBc Ag, and HBe Ag; the influenza antigens: HA, NP and NA; Hepatitis A surface antigens; the Herpes virus antigens: EBV gp340, EBV gp85, HSV gB, HSV gD, HSV gH, HSV early protein product, cytomegalovirus gB, cytomegalovirus gH, and IE protein gp72; the respiratory syncytial virus antigens: F protein, G protein, and N protein or fragments thereof.
  • HIV antigens gp160, gag, pol, Nef
  • Adjuvants Compositions of the invention can be administered in conjunction with other immunoregulatory agents, including adjuvants.
  • adjuvant refers to a compound or mixture that enhances an immune response.
  • the compositions disclosed herein can include an adjuvant.
  • the invention can also comprise combinations of aspects of one or more of the adjuvants identified herein.
  • Many adjuvants contain a substance designed to protect the antigen from rapid catabolism, such as aluminum hydroxide or mineral oil, and a stimulator of immune responses, such as lipid A, Bortadella pertussis or Mycobacterium tuberculosis derived proteins.
  • adjuvants are commercially available as, for example, Freund's Incomplete Adjuvant and Complete Adjuvant (Difco Laboratories, Detroit, Mich.); Merck Adjuvant 65 (Merck and Company, Rahway, N.J.); AS-2 (GlaxoSmithKline, Philadelphia, Pa.); aluminum salts such as aluminum hydroxide gel (alum) or aluminum phosphate; salts of calcium, iron or zinc; an insoluble suspension of acylated tyrosine; acylated sugars; cationically or anionically derivatized polysaccharides; polyphosphazenes; biodegradable microspheres; monophosphoryl lipid A and quil A.
  • Cytokines such as GM-CSF, interleukin-2, -7, - 12, and other like growth factors, may also be used as adjuvants.
  • the adjuvant composition can be a composition that induces an anti- inflammatory immune response (antibody or cell-mediated).
  • high levels of anti-inflammatory cytokines may include, but are not limited to, interleukin 4 (IL-4), interleukin 5 (IL-5), interleukin 10 (IL-10), and transforming growth factor beta (TGF ⁇ Optionally, an anti-inflammatory response would be mediated by CD4+ T helper cells.
  • Bacterial flagellin has been shown to have adjuvant activity (McSorley et al., J. Immunol.
  • the adjuvants used in conjunction with the disclosed compositions increase lipopolysaccharide (LPS) responsiveness.
  • Illustrative adjuvants include but are not limited to, monophosphoryl lipid A (MPL), aminoalkyl glucosaminide 4- phosphates (AGPs), including, but not limited to RC-512, RC-522, RC-527, RC-529, RC-544, and RC-560 (Corixa, Hamilton, Mont.).
  • the adjuvant composition can be one that induces an immune response predominantly of the Th1 type.
  • Th1-type cytokines tend to favor the induction of cell mediated immune responses to an administered antigen.
  • high levels of Th2-type cytokines e.g., IL-4, IL-5, IL-6 and IL-10
  • Th2-type cytokines tend to favor the induction of humoral immune responses.
  • a subject will support an immune response that includes Th1- and Th2-type responses.
  • the level of Th1-type cytokines will increase to a greater extent than the level of Th2-type cytokines.
  • the levels of these cytokines may be readily assessed using standard assays.
  • the level of Th2-type cytokines can increase to a greater extent than the level of Th1-type cytokines.
  • Certain adjuvants for eliciting a predominantly Th1-type response include, for example, a combination of monophosphoryl lipid A, preferably 3-de-O-acylated monophosphoryl lipid A, together with an aluminum salt adjuvants are available from Corixa Corporation (Seattle, Wash.; see, for example, U.S. Pat. Nos. 4,436,727; 4,877,611; 4,866,034 and 4,912,094, which are hereby incorporated by reference for their teaching of the same).
  • CpG-containing oligonucleotides in which the CpG dinucleotide is unmethylated also induce a predominantly Th1 response.
  • oligonucleotides are well known and are described, for example, in WO 96/02555, WO 99/33488 and U.S. Pat. Nos. 6,008,200 and 5,856,462. Immunostimulatory DNA sequences are also described, for example, by Sato et al., Science 273:352, 1996.
  • Another adjuvant comprises a saponin, such as Quil A, or derivatives thereof, including QS21 and QS7 (Aquila Biopharmaceuticals Inc., Framingham, Mass.); Escin; Digitonin; or Gypsophila or Chenopodium quinoa saponins.
  • compositions can include more than one saponin in the adjuvant combinations, for example combinations of at least two of the following group comprising QS21, QS7, Quil A, ⁇ -escin, or digitonin.
  • Saponin formulations can also be combined with vaccine vehicles composed of chitosan or other polycationic polymers, polylactide and polylactide-co-glycolide particles, poly-N-acetyl glucosamine-based polymer matrix, particles composed of polysaccharides or chemically modified polysaccharides, particles composed of glycerol monoesters, etc.
  • the saponins may be formulated together with a polyoxyethylene ether or ester, in either a non-particulate solution or suspension, or in a particulate structure.
  • the saponins can also be formulated with excipients such as CARBOPOLTM (Noveon, Cleveland, Ohio) to increase viscosity, or may be formulated in a dry powder form with a powder excipient such as lactose.
  • the adjuvant system includes the combination of a monophosphoryl lipid A and a saponin derivative, such as the combination of QS21 and 3D-MPL.
  • WO 94/00153 or a less reactogenic composition where the QS21 is quenched with cholesterol, as described in WO 96/33739.
  • Other formulations comprise an oil-in-water emulsion and tocopherol.
  • Another adjuvant formulation employing QS21, 3D-MPL.RTM. adjuvant and tocopherol in an oil-in-water emulsion is described in WO 95/17210.
  • Another enhanced adjuvant system involves the combination of a CpG- containing oligonucleotide and a saponin derivative particularly the combination of CpG and QS21 is disclosed in WO 00/09159.
  • the formulation additionally comprises an oil in water emulsion and tocopherol.
  • the disclosures of which are incorporated herein by reference in their entireties and polyoxyethylene ether adjuvants such as those described in WO 99/52549A1.
  • the adjuvant disclosed herein can be monosodium urate crystal (WO2004100984A1).
  • Other specific adjuvants contemplated herein include CpG- ODN, ADU-S100, and Poly(I:C).
  • ADU-S100 has increased stability and high affinity for mouse and human Stimulator of Interferon Genes (STING).
  • a TLR-3 adjuvant poly(I:C) is a potent mucosal adjuvant for intranasal delivery of vaccines shown for killed influenza virus antigens in a vaccine trial in pigs (Renu 2020).
  • a TLR-9 adjuvant CPG-ODN is known to induce antigen-specific sIgA antibody response in mucosal surfaces in intranasal vaccinated pigs against two respiratory viruses (Dhakal 2019; Zhang 2007).
  • Uric acid crystals activate innate host defense mechanisms and trigger robust inflammation and immune activation through NLRP3 inflammasome pathway (Braga 2017).
  • the innate immune activation by monosodium urate (MSU) crystal potentiates the adaptive immune response, such as the antibody response (Ng 2010).
  • MSU crystals are shown to be safe after intradermal injection (2-2000 ⁇ g) in humans and non-toxic (Sakamaki 2011; Cheng 2014).
  • the combination adjuvant targets multiple signaling pathways resulting in synergistic activation of immune cells and a balanced immune response.
  • CpG55.2 (Vaxine Pty Ltd) is a proprietary 24mer class B CpG oligonucleotide that is a potent activator of both human and mouse TLR9.
  • TLR9 agonists alone may be only weakly effective and Papagona et al.
  • TLR9 ligand CpG ODN 2006 is a poor adjuvant for the induction of de novo CD8+ T-cell responses in vitro. Scientific Reports, 2020. 10(1): p. 11620.) reported CpG ODN2006, a synthetic oligonucleotide TLR9 ligand, promoted antigen-driven expansion and functional maturation of naive CD8+ T cells ineffectively compared to other TLR adjuvants.
  • TLR9 adjuvants may work optimally when co-formulated with particulate adjuvant delivery systems such as alum or delta inulin.
  • Advax-CpG adjuvant (Vaxine Pty Ltd) is a combination of delta inulin polysaccharide (Gordon D.L., Sajkov D., Woodman R.J., Hyundai-Okubo Y., Cox M.M.J., Heinzel S., et al. Randomized clinical trial of immunogenicity and safety of a recombinant H1N1/2009 pandemic influenza vaccine containing AdvaxTM polysaccharide adjuvant. Vaccine.
  • AdvaxTM a polysaccharide adjuvant derived from delta inulin, provides improved influenza vaccine protection through broad-based enhancement of adaptive immune responses.
  • Vaccine. 2021;39:5940–5953 Li L., Honda-Okubo Y., Huang Y., Jang H., Carlock M.A., Baldwin J., et al. Immunisation of ferrets and mice with recombinant SARS-CoV-2 spike protein formulated with Advax-SM adjuvant protects against COVID-19 infection.
  • Advax delta inulin adjuvant overcomes immune immaturity in neonatal mice thereby allowing single-dose influenza vaccine protection.
  • a M2 protein-based universal influenza vaccine containing Advax-SM adjuvant provides newborn protection via maternal or neonatal immunization. Vaccine.
  • Advax-CpG adjuvant efficacy has been demonstrated to be effective in vaccines against coronavirus disease 2019 (COVID)-19 (Li L, Honda-Okubo Y, Huang Y, Jang H, Carlock MA, Baldwin J, et al.. Immunisation of Ferrets and Mice With Recombinant SARS-CoV-2 Spike Protein Formulated With Advax-SM Adjuvant Protects Against COVID-19 Infection. Vaccine (2021) 39(40):5940–53.
  • Nanoparticles The compositions, immunogenic compositions and vaccines described herein can comprise one or more nanoparticles.
  • nanoparticles include, but are not limited to, nanocarriers composed of one or more polymers.
  • the one or more polymers is a water soluble, non-adhesive polymer.
  • polymer is polyethylene glycol (PEG) or polyethylene oxide (PEO).
  • PEG polyethylene glycol
  • PEO polyethylene oxide
  • the polymer is polyalkylene glycol or polyalkylene oxide.
  • the one or more polymers is a biodegradable polymer.
  • the one or more polymers is a biocompatible polymer that is a conjugate of a water soluble, non-adhesive polymer and a biodegradable polymer.
  • the biodegradable polymer is polylactic acid (PLA), poly(glycolic acid) (PGA), or poly(lactic acid/glycolic acid) (PLGA).
  • the nanocarrier is composed of PEG-PLGA polymers.
  • the nanocarrier is formed by self-assembly. Self- assembly refers to the process of the formation of a nanocarrier using components that will orient themselves in a predictable manner forming nanocarriers predictably and reproducibly.
  • the nanocarriers are formed using amphiphillic biomaterials which orient themselves with respect to one another to form nanocarriers of predictable dimension, constituents, and placement of constituents.
  • the nanocarrier is a microparticle, nanoparticle, or picoparticle.
  • the microparticle, nanoparticle, or picoparticle is self-assembled.
  • the nanocarrier has a positive zeta potential.
  • the nanocarrier has a net positive charge at neutral pH.
  • the nanocarrier comprises one or more amine moieties at its surface.
  • the amine moiety is a primary, secondary, tertiary, or quaternary amine.
  • the amine moiety is an aliphatic amine.
  • the nanocarrier comprises an amine-containing polymer.
  • the nanocarrier comprises a protein or a peptide that is positively charged at neutral pH.
  • the nanocarrier is a latex particle.
  • the nanocarrier with the one or more amine moieties on its surface has a net positive charge at neutral pH.
  • Nanoparticles can aid the delivery of the antigens disclosed herien, and/or can also be immunogenic. Delivery can be to a particular site of interest, e.g. the mucosa.
  • the nanoparticle can create a timed release of antigens to enhance and/or extend the immune response.
  • the nanoparticle is associated with the antigens such that the composition can elicit an immune response.
  • the association can be, for example, wherein the nanoparticle is entrapped or encapsulated with the SARS-CoV-2 antigens and/or other antigens.
  • entrapped is meant that there is a physical encasing the antigens in nanoparticles.
  • the antigens are entrapped within the nanoparticle by a water/oil/water emulsion method.
  • the nanoparticle is poly(lactide co- glycolide) (PLGA). Depending on the ratio of lactide to glycolide used for the polymerization, different forms of PLGA can be obtained and utilized.
  • PLGA 75:25 identifies a copolymer whose composition is 75% lactic acid and 25% glycolic acid.
  • Different ratios can be used in this invention, e.g. 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80, 10:90, and numbers above and in between these ratios.
  • Additional examples of suitable nanoparticles include chitosin, calcium phosphate, lipids of various bacteria like E. coli, mycobactera, leptospira and mixtures thereof.
  • the composition can be derived mixing about 180 mg of PLGA to about 5 mg of antigenic material (or about 36 mg PLGA to 1mg antigenic material).
  • the entrapment (encapsulation) efficiency of antigens can vary.
  • the nanoparticle were 50-55% entrapped/encapsulated, calculated based on amount of total antigens used in the entrapment.
  • Entrapped antigens can be administered as mixtures of entrapped/encapsulated and unentrapped/unencapsulated antigens or the entrapped/encapsulated antigens can be further purified.
  • Additional compounds suitable for use with the compositions of the invention include, but are not limited to, one or more solvents, such as an organic phosphate- based solvent, bulking agents, coloring agents, pharmaceutically acceptable excipients, a preservative, pH adjuster, buffer, chelating agent, etc.
  • the additional compounds can be admixed into a previously formed composition comprising a nanoparticle, or the additional compounds can be added to the original mixture to be emulsified.
  • one or more additional compounds are admixed into an existing compositions immediately prior to its use.
  • Suitable preservatives for use with the compositions of the invention include, but are not limited to, cetylpyridinium chloride, benzalkonium chloride, benzyl alcohol, chlorhexidine, imidazolidinyl urea, phenol, potassium sorbate, benzoic acid, bronopol, chlorocresol, paraben esters, phenoxyethanol, sorbic acid, alpha- tocophernol, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, sodium ascorbate, sodium metabisulphite, citric acid, edetic acid, semi-synthetic derivatives thereof, and combinations thereof.
  • Suitable preservatives include, but are not limited to, benzyl alcohol, chlorhexidine (bis (p- chlorophenyldiguanido) hexane), chlorphenesin (3-(-4-chloropheoxy)-propane-1,2- diol), Kathon CG (methyl and methylchloroisothiazolinone), parabens (methyl, ethyl, propyl, butyl hydrobenzoates), phenoxyethanol (2-phenoxyethanol), sorbic acid (potassium sorbate, sorbic acid), Phenonip (phenoxyethanol, methyl, ethyl, butyl, propyl parabens), Phenoroc (phenoxyethanol 0.73%, methyl paraben 0.2%, propyl paraben 0.07%), Liquipar Oil (isopropyl, isobutyl, butylparabens), Liquipar PE (70% phenoxyethanol, 30% liquipar oil), Nipaguard
  • compositions disclosed herein can further comprise at least one pH adjuster.
  • pH adjusters include, but are not limited to, diethyanolamine, lactic acid, monoethanolamine, triethylanolamine, sodium hydroxide, sodium phosphate, semi-synthetic derivatives thereof, and combinations thereof.
  • the nanoemulsion vaccine can comprise a chelating agent.
  • the chelating agent is present in an amount of about 0.0005% to about 1%.
  • chelating agents include, but are not limited to, ethylenediamine, ethylenediaminetetraacetic acid (EDTA), phytic acid, polyphosphoric acid, citric acid, gluconic acid, acetic acid, lactic acid, and dimercaprol, and a preferred chelating agent is ethylenediaminetetraacetic acid.
  • the compositions disclosed herein can further comprise a buffering agent, such as a pharmaceutically acceptable buffering agent. Examples of buffering agents are known to those of skill in the art.
  • the vaccine can also comprise one or more emulsifying agents to aid in the formation of emulsions. Emulsifying agents include compounds that aggregate at the oil/water interface to form a kind of continuous membrane that prevents direct contact between two adjacent droplets.
  • compositions disclosed herein can be readily diluted with water or another aqueous phase to a desired concentration without impairing their desired properties.
  • Vaccines Specifically disclosed herein is a vaccine comprising the compositions disclosed herein, wherein said composition is a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof in a pharmaceutically acceptable carrier.
  • Intranasal administration includes administration via the nose, either with or without concomitant inhalation during administration. Such administration is typically through contact by the composition comprising the vaccine with the nasal mucosa, nasal turbinates or sinus cavity.
  • Administration by inhalation comprises intranasal administration, or may include oral inhalation. Such administration may also include contact with the oral mucosa, bronchial mucosa, and other epithelia.
  • Exemplary dosage forms for pharmaceutical administration are described herein. Examples include but are not limited to liquids, ointments, creams, emulsions, lotions, gels, bioadhesive gels, sprays, aerosols, pastes, foams, sunscreens, capsules, microcapsules, suspensions, pessary, powder, semi-solid dosage form, etc.
  • the pharmaceutical compositions may be formulated for immediate release, sustained release, controlled release, delayed release, or any combinations thereof, into the epidermis or dermis.
  • the formulations may comprise a penetration-enhancing agent.
  • Suitable penetration-enhancing agents include, but are not limited to, alcohols such as ethanol, triglycerides and aloe compositions.
  • the amount of the penetration-enhancing agent may comprise from about 0.5% to about 40% by weight of the formulation.
  • the vaccines of the invention can be applied and/or delivered utilizing electrophoretic delivery/electrophoresis.
  • the composition may be a transdermal delivery system such as a patch or administered by a pressurized or pneumatic device (i.e., “gene gun”). Such methods, which comprise applying an electrical current, are well known in the art.
  • the pharmaceutical compositions for administration may be applied in a single administration or in multiple administrations.
  • one dose can be placed in each nostril during vaccination.
  • bi-dose delivery can be used with the vaccines according to the invention.
  • Bi-dose devices contain two sub-doses of a single vaccine dose, one sub-dose for administration to each nostril.
  • the two sub-doses are present in a single chamber and the construction of the device allows the efficient delivery of a single sub-dose at a time.
  • a mono-dose device may be used for administering the vaccines according to the invention.
  • the vaccine can be given in one, two, three, four, or more doses, so that the subject is given a first dose (which can be a bi-dose or mono-dose, as described above), and then a second dose is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26,27, 28, 29, or 30 days, or 4 ,5, 6, 7, 8, 9, 10, 11, or 12 weeks, or 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months, or 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more years apart.
  • the subject can also receive a booster dose, which can be given at any time after the initial dose or doses.
  • the booster dose can comprise the same, or different, antigens than the initial vaccination(s).
  • the vaccine can be updated with more recent variant antigens.
  • the vaccine is administered locally to the nasopharyngeal area.
  • Preferred devices for intranasal administration of the vaccines according to the invention are spray devices. Suitable commercially available nasal spray devices include AccusprayTM (Becton Dickinson). Nebulizers produce a very fine spray (such as a mist) which can be easily inhaled and are also contemplated herein.
  • Preferred spray devices for intranasal use are devices for which the performance of the device is not dependent upon the pressure applied by the user. These devices are known as pressure threshold devices. Liquid is released from the nozzle only when a threshold pressure is applied.
  • the invention provides in a further aspect a pharmaceutical kit comprising an intranasal administration device as described herein containing a vaccine formulation according to the invention.
  • the invention is not necessarily limited to spray delivery of liquid formulations.
  • Vaccines according to the invention may be administered in other forms e.g. as a powder.
  • the subject, or patient, being given the vaccine can be a mammal, such as a human.
  • the subject may have previously been vaccinated against SARS-CoV-2, for example by intramuscular injection.
  • Such vaccination does not preclude the patient from also receiving the vaccine disclosed herein.
  • the vaccine that was initially given can be against a different variant, or the same variant.
  • the subject may have been exposed to one, or all, of the antigens in the vaccine disclosed herein, previous to the vaccine of the present invention being administered.
  • the subject may have previously been vaccinated, or may have natural immunity through exposure.
  • the subject may be given a smaller dose, or fewer separate doses, than a subject who does not have natural immunity.
  • One of skill in the art can readily determine the proper dosage of the vaccine to be given to the subject.
  • the dosage can be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, or 600 ⁇ g per dose, or more, less, or any amount in between these values.
  • Also disclosed is a method of preventing or lessening the severity of symptoms or markers of disease associated with SARS-CoV-2 infection in a subject comprising administering to the subject a vaccine, wherein the vaccine comprises a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS- CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof; wherein the vaccine further comprises a pharmaceutically acceptable carrier.
  • the vaccine comprises a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS- CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof; wherein the vaccine further comprises a pharmaceutically acceptable carrier.
  • preventing is meant reducing the chance that the subject will be infected with SARS-CoV-2 by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36 ,37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80,81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% compared with a subject who has not been vaccinated with the vaccine disclosed herein.
  • the subject contracts SARS-CoV-2, but has less severe symptoms as compared to what symptoms the subject would have had, had they not been vaccinated.
  • the subject can have a “mild” infection instead of a “moderate” infection.
  • “mild” infection refers to patients diagnosed with COVID-19 by a standardized RT-PCR assay exhibiting fever, rhinorrhea, mild cough, sore throat, malaise, headache, muscle pain, malaise, or any combination thereof, but with no shortness of breath. Patients with “mild” infection present no signs of a more serious lower airway disease and have a respiratory rate of less than 20 breaths per minute, a heart rate of less than 90 beats per minute, and oxygen saturation (pulse oximetry) greater than 93% on room air.
  • “moderate” infection refers to patients diagnosed with COVID-19 by a standardized RT-PCR assay exhibiting symptoms in the mild category and additional symptoms. These include more significant lower respiratory symptoms, including shortness of breath (at rest or with exertion) or signs of moderate pneumonia, including a respiratory rate of ⁇ 20 but ⁇ 30 breaths per minute, a heart rate of ⁇ 90 but ⁇ 125 beats per minute and oxygen saturation (pulse oximetry ) greater than 93% on room air. If some embodiments, subjects with moderate infection further exhibit lung infiltrates based on X-ray or CT scan that are ⁇ 50% present.
  • millild-to-moderate infection collectively refers to mild and moderate infections, as defined herein.
  • critical infection refers to a severe infection in which the patient has at least one of the following: (1) respiratory failure requiring at least one of the following: Endotracheal intubation and mechanical venti lation, oxygen delivered by high-flow nasal cannula, noninvasive positive pressure ventilation, or ECMO; (2) a clinical diagnosis of respiratory failure (in setting of resource limitation); (3) Septic shock (defined by SBP ⁇ 90 mm Hg, or Diastolic BP ⁇ 60 mm Hg); and (4) Multiple organ dysfunction/failure.
  • vaccination of a subject according to the methods described herein results in reduction of one or more inflammatory cytokines and/or chemokines.
  • the inflammatory cytokine or chemokine is a cytokine or chemokine listed in FIGS.
  • 9A-9J (e.g., sCD40L, EGF, Eotaxin (CCL11), FGF-2, Flt-3 ligand, Fractalkine, G-CSF, GM-CSF, GRO ⁇ (CXCL1), IFN- ⁇ 2, IFN- ⁇ , IL-1 ⁇ , IL-1 ⁇ , IL-1RA, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (CXCL8), IL-9, IL-10, IL-12 (p40), IL-12 (p70), IL-13, IL-15, IL-17A, IL-17E/IL-25, IL-17F, IL-18, IL-22, IL-27, IP-10 (CXCL10), MCP-1 (CCL2), MCP-3, M-CSF, MDC (CCL22), MIG (CXCL9), MIP-1 ⁇ (CCL3), MIP-1 ⁇ (CCL4), PDGF-AA, PDGF-AB/BB
  • FIGS. 17-19 CCL5, IL-5, IL-13, IL-2, IL-6, IL-10, IL-9, IFN- ⁇ , TNF- ⁇ .
  • the inflammatory cytokine and/or chemokine may comprise a cytokine or chemokine selected from one of CCL5, sCD40L, EGF, Eotaxin, FGF-2, Flt-3 ligand, Fractalkine, G-CSF, GM-CSF, GRO ⁇ , IFN- ⁇ 2, IFN- ⁇ , IL-1 ⁇ , IL-1 ⁇ , IL-1ra, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12 (p40), IL-12 (p70), IL-13, IL-15, IL-17A, IL-17E/IL-25, IL-17F, IL-18, IL-21, IL-22, IL-27, IP-10, MCP-1, MCP-3, M-CSF, MDC (CCL22), MIG, MIP-1 ⁇ , MIP-1 ⁇ , PDGF-AA, PDGF-AB/BB, RAN
  • the one or more inflammatory cytokines and/or chemokines comprises CCL5, IL-6, IL-8, IL-1 ⁇ , IL-10, TNF- ⁇ , or any combination thereof. In some embodiments, the one or more inflammatory cytokines and/or chemokines comprises CCL5, IL-6, TNF- ⁇ , or a combination thereof. In some embodiments, the one or more inflammatory cytokines and/or chemokines comprises CCL5, IL-6, IL-1 ⁇ , IL-8, or any combination thereof.
  • the level of the inflammatory cytokine and/or chemokines may be measured in blood plasma, e.g., by enzyme- linked immunosorbent assays (ELISA), bead-based immunoassays and other immunoassays.
  • the transcriptional level of inflammatory cytokines and/or chemokines may also be measured by RNA sequencing.
  • vaccination of a subject according to the methods described herein results in reduction of a symptom associated hyperinflammation.
  • hyperinflammation is cytokine release syndrome, hemophagocytic lymphohistiocytosis (HLH), macrophage activation syndrome, or acute respiratory distress syndrome (ARDS).
  • HHLH hemophagocytic lymphohistiocytosis
  • ARDS acute respiratory distress syndrome
  • vaccination of a subject according to the methods described herein reduces migration of CCR5+ immune cells.
  • the CCR5+ immune cells comprise macrophages, T cells, or both.
  • vaccination of a subject according to the methods described herein improves at least one respiratory parameter in the subject.
  • vaccination of a subject according to the methods described herein increases oxygen saturation in the subject.
  • vaccination of a subject according to the methods described herein results in reduced occurrence or risk of developing liver toxicity, kidney failure or a coagulation event.
  • the coagulation event comprises a blood clot, stroke, or pulmonary embolism.
  • treatment results in more normalization of kidney function.
  • Kidney function may be measured by measuring blood levels of creatine, BUN, sodium, or any combination thereof in the subject blood. In some embodiments, vaccination results in more normalization of liver function. Liver function may be measured by measuring blood levels of bilirubin, alanine transaminase (ALT), aspartate aminotransferase (AST), or any combination thereof. In some embodiments, vaccination of a subject according to the methods described herein results in reduction of SARS-CoV-2 viral load in the subject. In some embodiments, vaccination of a subject according to the methods described herein results in reduced duration or occurrence of hospitalization, ventilation or dialysis of the subject. In some embodiments, vaccination of a subject according to the methods described herein results in reduced lung damage to the subject.
  • ALT alanine transaminase
  • AST aspartate aminotransferase
  • vaccination of a subject according to the methods described herein results in the subject having a faster and/or more extensive recovery than a subject with similar symptoms who has not been vaccinated with the vaccine disclosed herein.
  • a number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims.
  • RBD receptor binding domain
  • MSU monosodium urate crystal
  • SARS-CoV-2 proteins Synthetic nucleic acids encoding the SARS-CoV-2 S-RBD (amino acids 319- 516) or full length nucleocapsid (N) were codon optimized for expression in E. coli and ordered as gblock gene fragments (IDTDNA). Fragments were cloned into a pRSET A bacterial expression plasmid (Invitrogen) utilizing BamHI/HindIII.
  • Plasmids were verified by restriction digest and Sanger sequencing. Plasmids were used to transform BL21(DE3) E. coli (NEB). Protein was induced by autoinduction (54). Bacteria were lysed using bacterial protein extraction reagent (BPER) (Thermo Fisher) with HALT protease inhibitors (Thermo Fisher). Insoluble material including protein of interest was pelleted spinning at 10,000 x G for 10 minutes.
  • BPER bacterial protein extraction reagent
  • HALT protease inhibitors Thermo Fisher
  • Insoluble pellets were washed twice in inclusion body wash buffer (20mM Tris-HCl, pH7.5, 10mM EDTA, 1% triton X-100), resolubilized using 50 mM CAPS, pH 11.0, 1% N- lauroylsarcosine, and 1mM dithiothreitol (DTT) with end-over-end mixing at room temperature for 30 minutes.
  • Soluble protein was dialyzed 3x against 20mM Tris-HCl, pH 8.5 with 0.1mM DTT [2]. Dialyzed proteins were purified by nickel affinity chromatography (Hispur kit, Thermo Fisher 88229).
  • MSU crystal adjuvant MSU crystals were synthesized by following the procedure described previously (25, 56, 58), which yielded the crystals with similar morphology and birefringence to those found in gout patients. Briefly, 1.68 g of solid uric acid was added to 400 mL sodium hydroxide solution (0.4 g of NaOH, 25 mM). The resultant opaque solution was allowed to remain overnight at 80 qC and the filtrate was rinsed with cold distilled water three times and air dried in the fume hood for 2 days.
  • the dried MSU particles were sieved into a size range of 1-5 ⁇ m in length and were nano- sized in diameter. They were divided into 5 mg aliquots, dispensed into individual vials, and sterilized by ethylene trioxide. The MSU crystals were then entrapped.
  • Experimental Animals Neutered male ferrets, 12-months-old, were obtained (Triple F Farms, PA) for use in this study. Ferrets were seronegative for influenza A viruses, MERS-CoV, and SARS-CoV-2. All ferrets were housed (3 per cage, 2 cages per experimental group) with a 12 h light/dark cycle and allowed access to food and water ad libitum.
  • Blood, oropharyngeal swab, nasal swab, and rectal swab were collected at days post vaccination (DPV) 0 and 21, and days post challenge (DPC) 0, 2, 4, 7, 9, 11 and 14 dpi.
  • DPC days post challenge
  • Half of the ferrets (3 per group, total 18) were necropsied at 7 DPC, the rest at 14 DPC.
  • Tissues and specimens collected at necropsy were bronchoalveolar lavage (BAL) fluid, blood, urine, intestine, lung, trachea, and nasal turbinates. Collected ferret secretions were resuspended in cold PBS containing 1% bovine serum albumin and antibiotics (5% penicillin/streptomycin).
  • Tissue samples were weighed and collected in viral transport media (TCID 50 ) or RNA later (RNA extraction). Tissue samples were homogenized, centrifuged, and the supernatant was aliquoted and stored at -80 °C for further testing. Titration of SARS-CoV-2 in samples To investigate whether collected specimens contain infectious live virus, the samples were inoculated onto confluent Vero E6 cells in 96 well plates, 100 ⁇ l/well serial diluted 10-fold beginning with 1 : 10 dilution. Plates were incubated at 37°C and 5% CO 2 for 1 hour for viral adsorption and an additional 100 ⁇ l of infection media was added to each well. After 5 days of incubation the plates were read for cytopathic effect and TCID 50 values were calculated using the Spearman-Karber algorithm (Ramakrishnan 2016).
  • a cDNA synthesis kit was used to synthesize single strand cDNA using total viral RNA.
  • SYBR Green supermix kit Bio-Rad, Hercules, CA
  • the number of viral RNA copies was calculated and compared to the number of copies of the standard control.
  • Primers used were either designed in house or previously published (Carolan 2014).
  • the generation of oligonucleotide dimers for each TaqMan primer pair was assessed using Power SYBR® Green PCR MasterMix with melting curve analysis, according to the manufacturer’s instructions. Primers which resulted in oligonucleotide dimer generation were redesigned and retested.
  • a comparison between primer pairs was also performed using Power SYBR® Green PCR MasterMix without a melting curve, according to the manufacturer’s instructions.
  • One to two microliter cDNA sample was assayed per reaction. Each reaction consisted of 1 cycle of 50 °C for 2 min, 1 cycle of 95 °C for 2 min, followed by 40 cycles of 95 °C for 15 s and 60 °C for 1 min.
  • Real time PCR runs for each gene included cDNA standards (10-fold and 2-fold dilutions, in triplicate).
  • the efficiency of each gene amplification was calculated by plotting the average Ct (y-axis) against the logarithm of the input amount of RNA/ ⁇ l cDNA (x- axis). A 10-fold dilution series was used for each gene.
  • the geometric mean of the efficiencies for the indicated genes was used for the housekeeping gene efficiency.
  • the fold change of expression of a gene was calculated using Double delta CT calculations using the housekeeping gene GAPDH for normalization (Livak 2001).
  • the viral RNA load was also substantially reduced in nasal turbinate, trachea, bronchoalveolar lavage and lungs of ferrets vaccinated with SARS- CoV-2 subunit vaccine compared to only viral proteins and adjuvant formulation in the control group (Fig. 2A, B, C, & D).
  • Enhanced immunological responses in terms of expression of important immune cytokines genes in lungs and nasal turbinates was noticed supporting the reduced viral load in the respiratory tract of ferrets received SARS-CoV-2 subunit vaccine (Fig. 3). This include IFN ⁇ and MCP-1 in lungs and nasal turbinate (Fig. 3A, B, C, D & E) and IL-17A in lungs (Fig. 3F).
  • Example 2 SARS-CoV-2 Spike receptor-binding domain entrapped chitosan nanoparticle intranasal vaccine elicits local and systemic Th1 and Th2 response in mice and antiviral immunity in Syrian hamsters Summary Given the ongoing COVID-19 pandemic and the need to build sustainable herd immunity in the population, the search for novel and safe vaccines is ongoing.
  • Disclosed herein is a novel intranasal subunit vaccine platform called NARUVAX- C19/Nano based on the SARS-CoV-2 spike protein receptor-binding domain (RBD) entrapped in mannose-conjugated chitosan nanoparticles (NP).
  • RBD SARS-CoV-2 spike protein receptor-binding domain
  • the adjuvant CpG55.2, a toll-like receptor 9 antagonist was included in the formulation. Both the resulting vaccines were assessed for immunogenicity and protective efficacy, as well as protection against virus transmission; soluble RBD mixed with alum adjuvant administered intramuscular was included as a control.
  • soluble RBD mixed with alum adjuvant administered intramuscular was included as a control.
  • This vaccine contain Spike protein receptor-binding domain (RBD) entrapped in mannose-conjugated chitosan nanoparticle called NARUVAX-C19/Nano, which has the potential to induce not only systemic but also local mucosal immunity.
  • RBD Spike protein receptor-binding domain
  • Anti-RBD antibodies block the interaction of the virus with the angiotensin-converting enzyme 2 (ACE2) cell receptor, and thus neutralize the virus and prevent infection (Buchholz 2004; Jiang 2020).
  • ACE2 angiotensin-converting enzyme 2
  • Intranasal delivery of vaccine induces mucosal immunity including production of secretory IgA antibodies which acts as the first line of defense against respiratory pathogens, including SARS- CoV-2 virus (Russell 2020).
  • intranasal vaccination is a needle-free noninvasive method which eliminates several issues (local pain and discomfort at injection site, increased cost of vaccine, need of trained person for vaccination, and fear of injection (Zheng 2018).
  • Intranasal immunization unlike other mucosal routes of administration, requires lower doses and does not expose antigens to extreme pH and has a larger absorption area (Riese 2014).
  • Nanoparticle (NP)-based protein subunit delivery helps protect the vaccine antigens from premature degradation, increases its stability, and ensures targeted delivery of immunogen to antigen- presenting cells (APC) (Pati 2018; Means 2003).
  • the NP-forming a natural carbohydrate polymer chitosan is biocompatible and bioavailable, and its positively charged amino groups electrostatically interact with negatively charged sialic acid mucus and epithelial surfaces, becoming a mucoadhesive vaccine vehicle (Renu 2020).
  • Surface labeling NP with mannose, a calcium-dependent (type C) mannose receptor of the lectin family binds readily to dendritic cells and macrophages [18], providing a pronounced adjuvant effect to the vaccine (Dhakal 2018; Han 2020).
  • the adjuvant CpG55.2 (Toll-like receptor 9 antagonist (Bode 2011); Vaxine Pty Ltd, Sydney, Australia) was additionally included in its formulation. Therefore we developed a novel candidate chitosan NP based S-RBD plus CpG containing vaccine formulation and evaluated for immunogenicity and protective efficacy in appropriate preclinical animal models. Further, elucidated the ability of vaccine for mitigating the SARS- CoV-2 transmission and compared its performance with an injectable RBD plus aluminum hydroxide (Alum) adjuvant vaccine.
  • Alum aluminum hydroxide
  • the virus strain hCoV-19/Kazakhstan/KazNAU-NSCEDI481/2020 of wildtype SARS-CoV-2 with D614G mutation in spike protein was used.
  • This virus was isolated at the Aikimbayev National Research Center for Especially Dangerous Infections (NSCEDI) in June 2020 from nasopharyngeal swab of a 45-year-old COVID-19 patient in Almaty, Ukraine (GISAID, #EPI_ISL_514093).
  • the virus was grown as described previously (Tabynov 2022).
  • Vaccine preparation Spike Protein RBD [Gln321-Ser591] purchased from a commercial source (ABP Biosciences, USA) was expressed in HEK293 cells, purity > 95% as determined by SDS-PAGE; Endotoxin - ⁇ 1.0 EU per ⁇ g protein as determined by the LAL method.
  • the SARS-CoV-2 vaccine formulation was prepared using mannose- conjugated chitosan nanoparticles (NP-vaccine) by a standard ionic gelation method as described previously (Tabynov 2022).
  • NP-CpG vaccine an adjuvanted NP-vaccine formulation
  • injectable vaccine used for comparative analysis was prepared by mixing S- RBD with aluminum hydroxide adjuvant (Alhydrogel® adjuvant 2%, InvivoGen, CA, USA; Alum vaccine). Both the vaccine formulations was sterile and contained less than 2 EU bacterial endotoxins per dose. The details of the vaccines content and their administration are provided in Table 1. Table 1. Vaccine formulations and routes of administration
  • DLS dynamic light scattering
  • APD avalanche photodiode detector
  • NP suspension (5 ⁇ L) was placed on the cleaned silicon wafer chip (SPI Supplies, USA) (Cat no. 4136SC-AB) and then on aluminum stubs, air dried in fume hood for 60 min and kept overnight under vacuum. Samples were coated with platinum for up to 30 nm thickness in the Q150T plus sputter coater (Quorum Technologies, UK) and imaging was done on the Hitachi SU5000 Field emission scanning electron microscope.
  • mice Four to-six-week-old SPF (specific pathogen-free) female BALB/c mice obtained from the NSCEDI Laboratory Animal Breeding Facility were used. Animals were placed in ventilated cages with HEPA filters (Allentown, USA) for 7 days prior to the experiment for acclimatization. Mice were immunized with NP-vaccine formulations, antigen alone, and PBS IN under ketamine (100 mg/kg) and xylazine (10 mg/kg) anesthesia in a. volume of 100 ⁇ l twice at 21 -day intervals. The Alum vaccine was administered in a similar manner but using an intramuscular route of administration.
  • mice were euthanized by cervical dislocation under ketamine-xylazine anesthesia, and their spleens were collected under aseptic conditions to evaluate the cellular immune response and lungs for determining the antigen-specific IgA antibodies.
  • the levels of anti-RBD specific secretory IgA, IgG, IgG1, and IgG2a antibodies were determined by enzyme linked immunoassay (ELISA) as previously described [22], SARS-CoV-2 Surrogate Virus Neutralization Test (sVNT) Kit (L00847; GenScript, Piscataway, USA) was used to detect antibodies that inhibit RBD binding to the ACE2 cell receptor according to the manufacturer's instructions.
  • the inhibition percentage of the sample was calculated as (1-Average OD of the sample/ Average OD of the negative control) ⁇ 100%.
  • a sample with an inhibition percentage ⁇ 30% was considered “negative” and ⁇ 30% was considered “positive” for SARS-CoV-2 neutralizing antibodies.
  • the neutralizing antibody values were determined according to the level of inhibition : low (30-59%), medium (60-89), high (90 ⁇ ). Determination of viral neutralizing antibodies was performed as previously described [22] using 1000 TCID 50 of wild-type SARS-CoV-2 (D614G). The neutralizing antibody titer was the highest dilution of serum that inhibited the cytopathic effect in 100% of wells.
  • IL-2 (#B320273), IFN- ⁇ (#B307222), IL-4 (#B320413), IL-10 (#B311304), IL-5 (#B317463), IL-6 (#B321215), IL-17A (#B303513), TNF- ⁇ (#B306271), using ELISA MAXTM Deluxe Set Mouse kits (BioLegend), in a suspension of splenocytes in response to restimulation with 5 ⁇ g RBD protein as previously described [22]. Cytokine production data were presented as the difference (Delta) of cytokine concentrations (pg/mL) between samples with and without RBD protein stimulation.
  • an Alum-based vaccine containing 5 ⁇ g/100 ⁇ l of RBD protein was administered IM.
  • hamsters of all groups were IN challenged with SARS-CoV-2 at a dose of 1 x 10 4 TCID 50 under ketamine-xylazine anesthesia and observed for 7 days post infection with daily measurement of body weight.
  • half of the animals (3/6) from each group were euthanized and collected nasal turbinates and lung samples.
  • Three lobes of the right lung from each animal were fixed in 10% formaldehyde for histopathological studies.
  • Two lobes of the left lung were homogenized in 1 mL DMEM using a TissueLyser II instrument (QIAGEN) at 300 vibrations/min for 60 seconds, the supernatant after centrifugation (5000 g for 15 min at 4°C) was collected and stored at -70°C for determination of viral titer.
  • Virus Transmission Assessment In all groups of hamsters, on the 2 nd day of the challenge, two na ⁇ ve animals were comingled for one day and then separated into clean cages, where they were kept in isolation for additional 4 days.
  • the sentinel animals were euthanized to assess viral load in the nasal turbinates and lungs, as well as pathological changes in the lungs through histological analysis.
  • the weight of the comingled animals for 5 days was also recorded.
  • Analysis of viral titers and histological evaluation Virus titers in the tissue homogenates were determined as previously described (Tabynov 2022).
  • the virus titer was calculated by using the Reed and Mench method and expressed in log10 TCID 50 /0.2 mL. Histological analysis of hamster lungs was performed as previously described (Tabynov 2022).
  • Each slide was quantified based on the severity of histologic changes, including interstitial pneumonia, alveolitis, bronchiolitis, alveolar destruction, interstitial infiltration, pulmonary hemorrhage, and peribronchiolar inflammation. Based on the previously described method (Tabynov 2022; Lu 2021) the assessment score was as follows: 4 points - extremely severe pathological lung changes; 3 points - severe pathological lung changes; 2 points - moderate pathological lung changes; 1 point - mild pathological lung changes; 0 points - no pathological changes.
  • Statistical analysis The GraphPad Prism 9.0.0 (San Diego, USA) was used for graphing and statistical analysis of experimental data.
  • NP-vaccine formulation without CpG elicited IgA antibody at levels significantly higher than mock and antigen only control groups.
  • NP-vaccine formulations with all tested doses in immunized mice did not elicit detectable levels of IgG, IgG1, IgG2a, and RBD-ACE2 blocking and virus-neutralizing antibodies.
  • Alum based vaccine induced significant levels of IgG, IgG1, IgG2a (Th2 polarization, Figure 5C) as well as RBD-ACE2 blocking (predominantly at medium and high levels, Figure 5D) and viral neutralizing antibody titers (GMT 15.7, Figure 5E) after booster immunization compared to control group.
  • NP-vaccines adjuvanted with the CpG provided significant production of IL-17A compered to control and corresponding groups of NP-vaccine formulations groups without CpG.
  • the Alum adjuvanted vaccine distinguished itself by the production of Th2 cytokines IL-4 and IL-6, whose levels were significant compared to controls, antigen alone and NP-vaccine groups.
  • Intranasal NP-vaccines provide protection against wild-type SARS-CoV-2 (D614G) infection in hamsters, but did not block the viral transmission Protective efficacy was evaluated clinically by recording changes in body weight gain for 7 days post challenge infection (Figure 7A).
  • Virus load was determined by measuring the infectious viral titer in oropharyngeal swabs of hamsters at day 2 post challenge (Figure 7C), as well as in nasal turbinates and lungs of euthanized animals at day 3 ( Figure 7D) and day 7 ( Figure 7E) after challenge infection.
  • the virus load in the oropharyngeal swabs of vaccinated hamsters was lower than in the control group, and this difference was significant for the NP-vaccine group.
  • Virus was detected in the respiratory organs of all hamsters, with the highest titers on day 3 after challenge.
  • NP-vaccine formulations elicited a different immunogenicity profile from that of IM alum-adjuvanted RBD vaccine. Both of the NP-vaccine formulations in rodent models did not induce measurable serum neutralizing antibodies, while induced mainly secretory IgA and cell mediated immune responses.
  • This feature of this NP vaccine is a cardinal difference from other intranasal subunit RBD- or Spike-based vaccines conjugated with Diphtheria toxoid (EcoCRM®) (Wong 2022) or outer membrane vesicles (OMVs) from Neisseria meningitidis (van der Ley 2021), respectively, which were shown to induce both systemic neutralizing antibodies and local IgA responses.
  • EcoCRM® Diphtheria toxoid
  • OMVs outer membrane vesicles
  • the NP-vaccine significantly reduced the challenge virus load in both the upper and lower respiratory tract, and, most importantly, reduced the degree of lung damage in hamsters after challenge to a greater extent than the IM vaccine group. It appears likely that protection with NP-vaccine formulations was due to both anti-RBD IgA plus the Th1 cellular immune responses that is consistent with findings of others (Weiskopf 2020; Moss 2022; Aleebrahim-Dehkordi 2022). Although the addition of CpG to the NP-vaccine formulation induced production of IL-17, a Th17 cytokine involved in protective immunity against many pathogens (Mills 2022), this did not translate to increased protective efficacy of the intranasal NP vaccine.
  • Alum is a highly Th2 biased adjuvant and in an earlier study (Tabynov 2022) where a veterinary COVID-19 vaccine called NARUVAX-C19 (pets) was studied in juvenile cats, the alum adjuvant did not enhance neutralizing antibody titers compared to antigen alone. Another important part of the present research was evaluation of the vaccines in protecting against virus transmission from vaccinated challenged animals to na ⁇ ve sentinels.
  • Mucosal vaccines the promise and the challenge. Nat Rev Immunol 6:148-58. 9. Manocha M, Pal PC, Chitralekha KT, Thomas BE, Tripathi V, Gupta SD, Paranjape R, Kulkarni S, Rao DN. 2005. Enhanced mucosal and systemic immune response with intranasal immunization of mice with HIV peptides entrapped in PLG microparticles in combination with Ulex Europaeus-I lectin as M cell target. Vaccine 23:5599-617. 10. Lisa Schnirring CN, Feb 21, 2018. 2018. CDC vaccine panel brings back FluMist for 2018-19 season. 11.
  • SARS-CoV-2 mutation hot spots include a novel RNA-dependent-RNA polymerase variant. J Transl Med 18:179. 12. Harvey WT, Carabelli AM, Jackson B, Gupta RK, Thomson EC, Harrison EM, Ludden C, Reeve R, Rambaut A, Consortium C-GU, Peacock SJ, Robertson DL. 2021. SARS-CoV-2 variants, spike mutations and immune escape.
  • Intranasal vaccination of recombinant adeno-associated virus encoding receptor-binding domain of severe acute respiratory syndrome coronavirus (SARS-CoV) spike protein induces strong mucosal immune responses and provides long-term protection against SARS-CoV infection.
  • SARS-CoV severe acute respiratory syndrome coronavirus
  • CD8- dendritic cells and macrophages cross-present poly(D,L-lactate-co-glycolate) acid microsphere- encapsulated antigen in vivo.
  • Swine influenza H1N1 virus induces acute inflammatory immune responses in pig lungs: a potential animal model for human H1N1 influenza virus.
  • NKT-cell adjuvant differentially induces the expression of B-cell activation factors in mice and swine.
  • Bacterial c-di-GMP is an immunostimulatory molecule. J Immunol 178:2171-81.
  • Ebensen T Schulze K, Riese P, Morr M, Guzman CA.2007.
  • Intranasal administration of CpG oligonucleotides induces mucosal and systemic Type 1 immune responses and adjuvant activity to porcine reproductive and respiratory syndrome killed virus vaccine in piglets in vivo.
  • Tabynov K Turebekov N, Babayeva M, Fomin G, Yerubayev T, Yespolov T, Li L, Renukaradhya GJ, Petrovsky N, Tabynov K.
  • An adjuvanted subunit SARS-CoV-2 spike protein vaccine provides protection against Covid-19 infection and transmission. NPJ Vaccines. 2022 Feb 23;7(1):24. doi: 10.1038/s41541- 022-00450-8.
  • Tabynov K Babayeva M, Nurpeisov T, Fomin G, Nurpeisov T, Saltabayeva U, Renu S, Renukaradhya GJ, Petrovsky N, Tabynov K.
  • Ad26 vaccine protects against SARS-CoV-2 severe clinical disease in hamsters. Nat Med. 2020 Nov;26(11):1694-1700. doi: 10.1038/s41591-020-1070-6 103. Kalnin KV, Plitnik T, Kishko M, Zhang J, Zhang D, Beauvais A, Anosova NG, Tibbitts T, DiNapoli J, Ulinski G, Piepenhagen P, Cummings SM, Bangari DS, Ryan S, Huang PD, Huleatt J, Vincent D, Fries K, Karve S, Goldman R, Gopani H, Dias A, Tran K, Zacharia M, Gu X, Boeglin L, Abysalh J, Vargas J, Beaulieu A, Shah M, Jeannotte T, Gillis K, Chivukula S, Swearingen R, Landolfi V, Fu TM, DeRosa F, Casimiro D.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Nanotechnology (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Biochemistry (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)

Abstract

Disclosed herein are compositions comprising SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof, wherein the nanoparticle comprises chitosan. These compositions can be present in the form of a vaccine for administration. The vaccine can be present in a kit, for example. The composition can be administered to a subject in need thereof in order to prevent, or lessen the severity of, SARS-CoV-2 infection in the subject.

Description

NANOTECHNOLOGY BASED INTRANASAL VACCINE FOR COVID-19 COMPRISING CHITOSAN CROSS-REFERENCE TO RELATED APPLICATIONS This application claims benefit of U.S. Provisional Application Nos. 63/310,215, filed February 15, 2022, and 63/417,817, filed October 20, 2022, both of which are hereby incorporated by reference in their entirety. BACKGROUND The traditional view that antibodies prevent viral infection - but in their absence, virus-infected cells are eliminated only with the participation of T cells - is also highly relevant to SARS-CoV-2 infection. However, the route of entry for SARS- CoV-2 being the respiratory tract provides definition as to the type and quality of antibodies needed to prevent infection and droplet transmission. Available data indicate that virus-neutralizing (VN) antibodies may appear in COVID-19 patients, but not consistently (Prompetchara et al. 2020), and recovered patients may have low or none of the VN antibody titers (Huang 2020; Melgaco 2020; Wu 2020). Conversely, SARS-CoV-2-specific central memory CD4 and effector memory CD8 T cells were detected in the peripheral blood of COVID-19 patients after 14 days of infection (Thevarajan 2020; Weiskopf 2020) leading to a proposition that detection of memory T cells may be the appropriate readout of effective COVID vaccines (Melgaco 2020). Additionally, protective immunity to viral infections in vaccinated and convalescent individuals with weak antibody production has been attributed to memory CD8 T cells (Bauer 2006). Thus, what is needed in the art are COVID vaccines that can induce both antibodies and T cell responses. Protective mucosal immune responses are most effectively induced by mucosal immunization through oral, nasal, rectal or vaginal routes, but the vast majority of vaccines in use today are administered by injection (Neutra 2006). For respiratory pathogens such as SARS-CoV-2, induction of mucosal antibodies is critical. Mucosal vaccines have gained attention due to their ability to induce broad cross-reactive responses (Manocha 2005; Lisa Schnirring, 2018). RNA viruses are characterized by a high mutation rate, up to a million times higher than that of their hosts. SARS-CoV-2, being an RNA virus, is prone to mutation, and multiple mutations were already noted in the S gene (Pachetti 2020; Harvey 2021). While injectable vaccines elicit systemic IgG response and protect against homologous strains, they induce poor secretory (s) sIgA antibodies needed to prevent virus replication and load in the respiratory tract (Cox 2004). In contrast, vaccines delivered intranasally (IN) can induce sIgA in the respiratory tract that can neutralize homologous and genetic variants by binding to newly synthesized viral proteins in mucosal epithelial cells (Mazanec 1995; Suzuki 2015). This property of sIgA is important to prevent dissemination of virus to lungs and to prevent virus shedding (Shim 2010; Du 2008; Hu 2007). In addition, mucosal vaccines can also induce IgG response as shown with influenza and tuberculosis (Eickhoff 2019; Gallorini 2014; Renu 2021). Several SARS-CoV and MERS-CoV vaccine studies revealed better correlates of protection with mucosal vaccines as compared with parenteral vaccination (Moreno-Fierros 2020). Thus, what is needed in the art are COVID vaccines for intranasal (IN) delivery, because respiratory tract is the main port of entry identified for SARS-CoV-2. T cells, as described above, play an indispensable role in the mediation of long-term protection, but such an effort requires methods to elicit antigen-specific T cell responses in vaccine settings. Soluble and subunit antigens (Ags) that are otherwise poor immunogens become highly immunogenic when delivered after entrapping in nanoparticle (NP) (Bacon 2000; Bertram 2010; Dhakal 2018). Importantly, nanoparticles (<500 nm) readily traffic to lymphoid tissues, and processed efficiently by dendritic cells (DCs), macrophages (0ĭV), and activate B and T cells via cross-linking (Woodrow 2012; Heit 2007; Schliehe 2011). The virus antigens when entrapped or encapsulated within the NP are protected from degradation, especially when delivered to mucosal surfaces (Dhakal 2017; Hiremath 2016; Danhier 2012). Advantages of intranasal immunization to avoid interference by preexisting antibodies in the host has been studied in rodents and pigs (Renu 2021; Zhang 2016). Induction of cell mediated cross-protective immune response to killed/subunit antigens is possible when delivered through nanoparticles (Dhakal 2017; Zhang 2016). Coadministration of antigen and adjuvant in the nanoparticle is possible which help to improve the immunity and vaccine dose sparing. What is needed in the art is delivery of a vaccine cargo targeting the immune cells using nanoparticles (Renu 2021; Han 2020). Several types of polymer-based nanoparticle vaccine delivery platforms have been developed for influenza virus intranasal vaccine (Renu 2021; Dhakal 2018; Dhakal 2017; Products 2016; Makadia 2011; Menon 2014; Dhakal 2018). It was shown that nano vaccines made of influenza virus peptides and inactivated virus antigens, chitosan, and PLGA administered intranasally led to induction of robust mucosal B and/or T cell responses, including virus neutralizing sIgA and IgG antibodies both locally (respiratory tract) and systemically (serum), accompanied with reduction in flu signs and virus load in the airways of pigs (Renu 2021; Dhakal 2018; Dhakal 2017; Hiremath 2016; Dhakal 2018; Khatri 2010; Dhakal 2017; Renu 2020; Dhakal 2019; Dhakal 2019 (2); Run 2018). What is needed in the art is a nanoparticle vaccine for SARS-CoV-2. Three subunits of structural proteins of SARS-CoV-2, spike (S) protein, receptor binding domain (RBD) of the spike protein (S-RBD), nucleocapsid (N), and matrix (M) proteins are known to encompass large numbers of immunogenic antigenic epitopes. What is needed in the art is a combination of these proteins to induce better protective immunity as opposed to the use of only S protein in a vaccine, as it is prone for frequent genetic changes. In summary, what is needed in the art is a safe, effective SARS-CoV-2 nanoparticle vaccine that can be administered intranasally. SUMMARY Disclosed herein is a composition comprising SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof, wherein the nanoparticle comprises chitosan. Further disclosed is a vaccine comprising SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof in a pharmaceutically acceptable carrier, wherein the nanoparticle comprises chitosan. Also disclosed is a method of eliciting or enhancing an immune response to SARS-CoV-2 in a subject, the method comprising administering to the subject a vaccine, wherein the vaccine comprises a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof; wherein the vaccine further comprises a pharmaceutically acceptable carrier, wherein the nanoparticle comprises chitosan. Disclosed further is a method of preventing or lessening the severity of symptoms associated with SARS-CoV-2 infection in a subject, the method comprising administering to the subject a vaccine, wherein the vaccine comprises a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof; wherein the vaccine further comprises a pharmaceutically acceptable carrier, wherein the nanoparticle comprises chitosan. The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims. DESCRIPTION OF DRAWINGS Figure 1A-1D shows detection of replicating infectious SARS-CoV-2 load in the respiratory tract of vaccinated and virus challenged ferrets. One-year-old male ferrets were vaccinated twice at three weeks interval intranasally with SARS-CoV-2 RBD (spike) and nucleocapsid (N) antigens and monosodium urate crystal adjuvant and challenged with SARS-CoV-2 intranasally. The swab samples collected from: (1A) Nostrils and (1B) Oro-pharynx at day post challenge infection (DPC) 2, 4, 7, 9, 11, and 14; and samples of (1C) nasal turbinate and (1D) lung tissues collected at DPC 7 were measured by cell culture method for the infectious viral load, expressed in log10 TCID50 per ml of swab fluid or per g tissue. Each bar is the mean of 3 to 6 animals ± SEM. Figure 2A-2D shows detection of SARS-CoV-2 RNA load in the respiratory tract of vaccinated and virus challenged ferrets. One-year-old male ferrets were vaccinated twice at three week intervals intranasally with SARS-CoV-2 RBD (spike) and nucleocapsid (N) antigens and monosodium urate crystal adjuvant and challenged with SARS-CoV-2 intranasally. The samples of respiratory tract tissue/fluid: (2A) Nasal turbinate, (2B) Trachea, (2C) Bronchoalveolar lavage fluid, and (2D) Lungs collected at DPC 14 were measured for viral RNA load expressed in log10 RNA copies per g tissue or per ml fluid. Each bar is the mean of 3 animals ± SEM.
Figure 3A-3F shows detection of mRNA of immune genes in the respiratory tract of vaccinated and virus challenged ferrets. One-year-old male ferrets were vaccinated twice at three weeks interval intranasally with SARS-CoV-2 RBD (spike) and nucleocapsid (N) antigens and monosodium urate crystal adjuvant and challenged with SARS-CoV-2 intranasally. The samples of respiratory tract tissues expressing immune genes: IFNa in (3 A.) Lungs and (3B) Nasal turbinate at DPC 7; MCP1 in (3C) Lungs at DPC 7 and (3D & 3E) Nasal turbinate at DPC 7 and DPC 14; and (3F) IL-17 in Lungs at DPC 14 were measured by qRT-PCR. The standard double delta. Ct values were normalized with housekeeping gene GAPDH and the values of mock uninfected ferret respective tissue values were subtracted from the experimental group values. Each bar is the mean of 3 animals ± SEM. Asterisks refers to significant (*p<0.05) difference between the indicated groups.
Figure 4A-4B shows scanning electron microscope analysis of (4A) mannose- conjugated chitosan nanoparticles and (4B) mannose-conjugated chitosan nanoparticles entrapped with RBD protein.
Figure 5A-5E shows antibody response in BALB/c mice after vaccination. Antigen-specific serum (5A) and lung (5B) IgA antibodies in mice at 21 days after prime and booster intranasal immunization with RBD-based nanoparticle vaccine formulations and intramuscular immunization with Alum adjuvanted vaccine. Levels of IgG, IgG1, IgG2a (5C) and RBD-ACE2 blocking antibodies (5D) as well as viral neutralizing antibody titers (5E) in mice after booster immunization. Mannose- conjugated chitosan-NP-based vaccine formulations, including those with CpG adjuvant (NP-CpG), were administered in doses containing 5, 2.5, and 1.25 μg RBD protein. For comparison, studies included antigen alone group ( Ag) with an intranasal immunization of 5 μg RBD protein/dose, an Alum adjuvanted. (5 μg RBD/dose) vaccine group with intramuscular injection, and control group (PBS). Levels of IgA, IgG, IgG1, and IgG2a. antibodies are presented as optical density at 450 nm. A sample with an inhibition percentage <30% was considered "negative" and ≥30% was considered “positive" for SARS-CoV-2 neutralizing antibodies. The following neutralizing antibody values were determined according to the level of inhibition: low (30-59%), medium (60-89), high (90≤). Viral neutralizing antibody levels with the wild-type of SARS-CoV-2 (D614G) are presented as geometric mean titers with 95% confidence intervals. Differences in antibody levels between animal groups were assessed using Tukey's multiple comparisons test or t-test. P < 0.05 was considered statistically significant. *P < 0.05, **P < 0.01, ***P < 0.001, and ****p < 0.0001.
Figure 6A-6H shows antigen-specific cytokine production in the splenocyte suspension of BALB/c mice at day 21 post booster IN immunization with RBD-based NP-vaccine formulations and control IM immunization with Alum adjuvanted vaccine. Mannose-conjugated chitosan-NP-based vaccine formulations with and without including CpG adjuvant were administered in doses containing 5, 2.5, and 1.25 μg RBD protein for (6 A) IFN-γ; (6B) IL-2; (6C) IL- 17 A; (6D) TNF-a; (6E) IL- 4; (6F) IL-6; (6G) IL-5; and (6H) IL-10. For comparison included antigen alone group (Ag) delivered IN with 5 μg RBD protein/dose, an Alum adjuvanted (5 μg RBD/dose) vaccine group administered. IM, and a control group (PBS). Cytokine production data were presented as the difference (Delta) of cytokine concentrations (pg/mL) between samples with and without RBD stimulation. Differences between animal groups were assessed using Tukey's multiple comparisons test. P < 0.05 was considered statistically significant. *P < 0.05, **P < 0.01, ***p < 0.001.
Figure 7A-7G shows efficacy of RBD-based NP-vaccine in Syrian hamsters for protection against wild-type SARS-CoV-2 (D614G) infection and virus transmission. Mannose-conjugated chitosan-NP-based vaccine formulations, including the NP-CpG vaccine were administered with a dose containing 5 μg RBD protein. For comparison included IM alum-adjuvanted (5 μg RBD/dose) group, and control group (PBS). Animals were IN challenged with SARS-CoV-2 and the following parameters were studied: Changes in body weight (7 A); Viral load in oropharyngeal swabs (expressed as log10 TCID50/0.2 mL) on day 2 (7C) after challenge; Viral load in nasal turbinates and lungs on day 3 (7D) and. day 7 (7E) after challenge; Viral load in nasal turbinates and lungs of sentinel animals on day 5 (7F) post co-mingled with infected animals; Pathological changes in the lungs of animals on days 3 and 7 after challenge, as well as sentinels on day 5 after co-mingling with challenged animals by histological analysis (7B, 7G). Differences between the animal groups were assessed using Tukey's multiple comparisons test. P < 0.05 was considered, statistically significant. *P < 0.05, **P < 0.01. † P=0.03 - <0.0001 compared to the control group.
DETAILED DESCRIPTION
Definitions
Disclosed herein are methods and compositions for treating or preventing COVID-19 in a subject that involve combining SARS-CoV-2 antigens with a nanoparticle.
An “immunogenic composition” is a composition of matter suitable for administration to a human or animal subject (e.g., in an experimental setting) that is capable of eliciting a specific immune response, e.g., against a pathogen, such as SARS-CoV-2. As such, an immunogenic composition includes one or more antigens (for example, whole purified virus or antigenic subunits, e.g., polypeptides, thereof) or antigenic epitopes. -An immunogenic composition can also include one or more additional components capable of eliciting or enhancing an immune response, such as an excipient, carrier, and/or adjuvant. In certain instances, immunogenic compositions are administered to elicit an immune response that protects the subject against symptoms or conditions induced by a pathogen. In some cases, symptoms or disease caused by a pathogen is prevented (or treated, e.g., reduced or ameliorated) by inhibiting replication of the pathogen following exposure of the subject to the pathogen. In the context of this disclosure, the term immunogenic composition will be understood to encompass compositions that are intended for administration to a subj ect or population of subj ects for the purpose of eliciting a protective or palliative immune response against the virus (that is, vaccine compositions or vaccines).
An “antigen” is a compound, composition, or substance that can stimulate the production of antibodies and/or a T cell response in an animal, including compositions that are injected, absorbed or otherwise introduced into an animal. The term “antigen” includes all related antigenic epitopes. The term “epitope” or “antigenic determinant” refers to a site on an antigen to which B and/or T cells respond. The “dominant antigenic epitopes” or “dominant epitope” are those epitopes to which a functionally significant host immune response, e.g., an antibody response or a T-cell response, is made. Thus, with respect to a protective immune response against a pathogen, the dominant antigenic epitopes are those antigenic moieties that when recognized by the host immune system result in protection from disease caused by the pathogen. The term “T-cell epitope” refers to an epitope that when bound to an appropriate MHC molecule is specifically bound by a T cell (via a T cell receptor). A “B-cell epitope” is an epitope that is specifically bound by an antibody (or B cell receptor molecule). An antigen can also affect the innate immune response. An “immune response” is a response of different cell types of the immune system, such as a B cell, T cell, or monocyte, to a stimulus. An immune response can be a B cell response, which results in the production of specific antibodies, such as antigen specific neutralizing antibodies. An immune response can also be a T cell response, such as a CD4+ T cell response and/or a CD8+ T cell response. In some cases, the response is specific for a particular antigen (that is, an “antigen-specific response”). An immune response can also include the innate response. If the antigen is derived from a pathogen, the antigen-specific response is a “pathogen-specific response.” A “protective immune response” is an immune response that inhibits a detrimental function or activity of a pathogen, reduces infection by a pathogen, or decreases symptoms (including death) that result from infection by the pathogen. A protective immune response can be measured, for example, by the inhibition of viral replication or plaque formation in a plaque reduction assay or virus-neutralization assay, or by measuring resistance to pathogen challenge in vivo. The immunogenic compositions disclosed herein are suitable for preventing, ameliorating and/or treating disease caused by infection of the virus. The abbreviation “KAg” stands for killed antigen and represents the killed or inactivated virus. The inactivated virus comprises one or more immunogenic viral proteins and therefore the inactivated virus can be considered a killed antigen. The abbreviation “NP-KAg” stands for nanoparticle-killed antigen. This represents the nanoparticle encapsulated inactivated swine influenza virus. As used herein, the terms “virus-like particle” or “VLP” refer to a non- replicating, viral shell. VLPs are generally composed of one or more viral proteins associated with viral surface capsid structure. VLPs can form spontaneously upon recombinant expression of the protein in an appropriate expression system. VLPs, when administered to an animal, can be immunogenic and thus can cause a protective or therapeutic immune response in the animal. Methods for producing VLPs are generally known in the art and discussed more fully below. The presence of VLPs following recombinant expression of viral proteins can be detected using conventional techniques known in the art, such as by electron microscopy, biophysical characterization, and the like. See, e.g., Baker et al., Biophys. J. (1991) 60:1445-1456; Hagensee et al., J. Virol. (1994) 68:4503-4505. For example, VLPs can be isolated by density gradient centrifugation and/or identified by characteristic density banding. The term “treatment” refers to the medical management of a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder. This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder. Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. An "increase" can refer to any change that results in a greater amount of a symptom, disease, composition, condition or activity. An increase can be any individual, median, or average increase in a condition, symptom, activity, composition in a statistically significant amount. Thus, the increase can be a 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% increase so long as the increase is statistically significant. A "decrease" can refer to any change that results in a smaller amount of a symptom, disease, composition, condition, or activity. A substance is also understood to decrease the genetic output of a gene when the genetic output of the gene product with the substance is less relative to the output of the gene product without the substance. Also for example, a decrease can be a change in the symptoms of a disorder such that the symptoms are less than previously observed. A decrease can be any individual, median, or average decrease in a condition, symptom, activity, composition in a statistically significant amount. Thus, the decrease can be a 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% decrease so long as the decrease is statistically significant. "Inhibit," "inhibiting," and "inhibition" mean to decrease an activity, response, condition, disease, or other biological parameter. This can include but is not limited to the complete ablation of the activity, response, condition, or disease. This may also include, for example, a 10% reduction in the activity, response, condition, or disease as compared to the native or control level. Thus, the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels. By “reduce” or other forms of the word, such as “reducing” or “reduction,” is meant lowering of an event or characteristic (e.g., tumor growth). It is understood that this is typically in relation to some standard or expected value, in other words it is relative, but that it is not always necessary for the standard or relative value to be referred to. For example, “reduces tumor growth” means reducing the rate of growth of a tumor relative to a standard or a control. By “prevent” or other forms of the word, such as “preventing” or “prevention,” is meant to stop a particular event or characteristic, to stabilize or delay the development or progression of a particular event or characteristic, or to minimize the chances that a particular event or characteristic will occur. Prevent does not require comparison to a control as it is typically more absolute than, for example, reduce. As used herein, something could be reduced but not prevented, but something that is reduced could also be prevented. Likewise, something could be prevented but not reduced, but something that is prevented could also be reduced. It is understood that where reduce or prevent are used, unless specifically indicated otherwise, the use of the other word is also expressly disclosed. The term “subject” refers to any individual who is the target of administration or treatment. The subject can be a vertebrate, for example, a mammal. In one aspect, the subject can be human, non-human primate, bovine, avian, equine, porcine, canine, or feline. The subject can also be a guinea pig, rat, hamster, rabbit, mouse, or mole. Thus, the subject can be a human or veterinary patient. The term “patient” refers to a subject under the treatment of a clinician, e.g., physician. The term “therapeutically effective” refers to the amount of the composition used is of sufficient quantity to ameliorate one or more causes or symptoms of a disease or disorder. Such amelioration only requires a reduction or alteration, not necessarily elimination. "Biocompatible" generally refers to a material and any metabolites or degradation products thereof that are generally non-toxic to the recipient and do not cause significant adverse effects to the subject. "Comprising" is intended to mean that the compositions, methods, etc. include the recited elements, but do not exclude others. "Consisting essentially of'' when used to define compositions and methods, shall mean including the recited elements, but excluding other elements of any essential significance to the combination. Thus, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives, and the like. "Consisting of'' shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the compositions provided and/or claimed in this disclosure. Embodiments defined by each of these transition terms are within the scope of this disclosure. A “control” is an alternative subject or sample used in an experiment for comparison purposes. A control can be "positive" or "negative." For example, a negative control can be an untreated or mock treated control. A positive control, can be a control with a known positive response. A "pharmaceutically acceptable" component can refer to a component that is not biologically or otherwise undesirable, i.e., the component may be incorporated into a pharmaceutical formulation provided by the disclosure and administered to a subject as described herein without causing significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the formulation in which it is contained. When used in reference to administration to a human, the term generally implies the component has met the required standards of toxicological and manufacturing testing or that it is included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug Administration. "Pharmaceutically acceptable carrier" (sometimes referred to as a “carrier”) means a carrier or excipient that is useful in preparing a pharmaceutical or therapeutic composition that is generally safe and non-toxic and includes a carrier that is acceptable for veterinary and/or human pharmaceutical or therapeutic use. The terms "carrier" or "pharmaceutically acceptable carrier" can include, but are not limited to, phosphate buffered saline solution, water, emulsions (such as an oil/water or water/oil emulsion) and/or various types of wetting agents. As used herein, the term "carrier" encompasses, but is not limited to, any excipient, diluent, filler, salt, buffer, stabilizer, solubilizer, stabilizer, or other material well known in the art for use in pharmaceutical formulations and as described further herein. “Pharmacologically active” (or simply “active”), as in a “pharmacologically active” derivative or analog, can refer to a derivative or analog (e.g., a salt, ester, amide, conjugate, metabolite, isomer, fragment, etc.) having the same type of pharmacological activity as the parent compound and approximately equivalent in degree. “Therapeutic agent” refers to any composition that has a beneficial biological effect. Beneficial biological effects include both therapeutic effects, e.g., treatment of a disorder or other undesirable physiological condition, and prophylactic effects, e.g., prevention of a disorder or other undesirable physiological condition (e.g., a non- immunogenic cancer). The terms also encompass pharmaceutically acceptable, pharmacologically active derivatives of beneficial agents specifically mentioned herein, including, but not limited to, salts, esters, amides, proagents, active metabolites, isomers, fragments, analogs, and the like. When the terms “therapeutic agent” is used, then, or when a particular agent is specifically identified, it is to be understood that the term includes the agent per se as well as pharmaceutically acceptable, pharmacologically active salts, esters, amides, proagents, conjugates, active metabolites, isomers, fragments, analogs, etc. “Therapeutically effective amount” or “therapeutically effective dose” of a composition (e.g. a composition comprising an agent) refers to an amount that is effective to achieve a desired therapeutic result. In some embodiments, a desired therapeutic result is the control of type I diabetes. In some embodiments, a desired therapeutic result is the control of obesity. Therapeutically effective amounts of a given therapeutic agent will typically vary with respect to factors such as the type and severity of the disorder or disease being treated and the age, gender, and weight of the subject. The term can also refer to an amount of a therapeutic agent, or a rate of delivery of a therapeutic agent (e.g., amount over time), effective to facilitate a desired therapeutic effect, such as pain relief. The precise desired therapeutic effect will vary according to the condition to be treated, the tolerance of the subject, the agent and/or agent formulation to be administered (e.g., the potency of the therapeutic agent, the concentration of agent in the formulation, and the like), and a variety of other factors that are appreciated by those of ordinary skill in the art. In some instances, a desired biological or medical response is achieved following administration of multiple dosages of the composition to the subject over a period of days, weeks, or years. As used herein, the term “intranasal(ly)” refers to application of the compositions of the present invention to the surface of the skin and mucosal cells and tissues of the nasal passages, e.g., nasal mucosa, sinus cavity, nasal turbinates, or other tissues and cells which line the nasal passages. As used herein, the terms "drug composition" and "drug", "vaccinal composition" and "vaccine" and "vaccine composition" and "drug-vaccine composition" and "drug-vaccine dual agent" and "therapeutic composition" and "therapeutic-immunologic composition" cover any composition that induces protection against a pathogen. In some embodiments, the protection may be due to an inhibition or prevention of infection by a pathogen. In other embodiments, the protection may be induced by an immune response against the antigen(s) of interest, or which efficaciously protects against the antigen; for instance, after administration or injection into the subject, elicits a protective immune response against the targeted antigen or immunogen or provides efficacious protection against the antigen or immunogen expressed from the inventive adenovirus vectors of the invention. Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this pertains. The references disclosed are also individually and specifically incorporated by reference herein for the material contained in them that is discussed in the sentence in which the reference is relied upon. General Description Disclosed herein is a composition comprising SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises at least a spike protein (S) or an antigenic fragment thereof, and at least a nucleocapsid protein (N) or an antigenic fragment thereof. The composition can be used as a vaccine, such as an intranasal vaccine. Vaccines are discussed in more detail below. SARS-CoV-2 is a single-stranded RNA-enveloped virus. The capsid consists of the nucleocapsid protein (N) and this is further surrounded by a membrane, that contains three proteins: the membrane protein (M) and the envelope protein (E), which are involved in the virus budding process, and the spike glycoprotein (S), which is a key player in binding host receptor and mediating membrane fusion and virus entry into host cells. A large number of glycosylated S proteins cover the surface of SARS-CoV-2 and bind to the host cell receptor angiotensin-converting enzyme 2 (ACE2), mediating viral cell entry. When the S protein binds to the receptor, TM protease serine 2 (TMPRSS2), a type 2 TM serine protease located on the host cell membrane, promotes virus entry into the cell by activating the S protein. Once the virus enters the cell, the viral RNA is released, polyproteins are translated from the RNA genome, and replication and transcription of the viral RNA genome occur via protein cleavage and assembly of the replicase–transcriptase complex. Viral RNA is replicated, and structural proteins are synthesized, assembled, and packaged in the host cell, after which viral particles are released. Antigens The present invention makes use of not only the spike protein, which is commonly used in vaccines, but also the N protein. This combination of proteins provides a powerful synergistic effect. The S antigen alone is the basis for the Pfizer/BioNTek vaccine, Moderna vaccine, Johnson & Johnson’s (J&J) Janssen vaccine, AstraZeneca/Oxford vaccine, CanSino vaccine, Sputnik V vaccine, Novavax vaccine, and others. Utilizing both antigens provides better and broader protection against variant viruses coming out of SARS-CoV-2 in multiple tissues/organs/cells. Furthermore, because viral N protein contributes to forming helical ribonucleoproteins during the packaging of the RNA genome, regulating viral RNA synthesis during replication and transcription and modulating metabolism in infected subjects, it is a highly efficacious antigen when combined with the S protein. By “antigen protein or antigenic fragment thereof” is meant that either the entire antigen is included in the composition (such as the “S” or “N” protein), or a fragment of the antigen is included. When a fragment is used, it is contemplated herein that the fragment will be of sufficient length to induce an immune response when the fragment is exposed to a subject who would have an immune response to the full-length protein. For example, the fragment can be 10, 20, 30, 40, 50, 60, 70, 80, 90, 91, 92, 93, 94, 95, 96, 97. 98, or 99% identical to the protein itself. The fragment can be truncated at either the C terminal, N terminal, or both, and can comprise one or more variations with respect to the full-length antigen. One of skill in the art will understand how to identify antigenic fragments that are capable of eliciting an immune response. Furthermore, the antigen or antigenic fragment used in the compositions disclosed herein can be identical to a known antigen, or can be a derivative (variation) thereof. When a derivative is used, the derivative can comprise substitutions, deletions, or insertions in the amino acid sequence in comparison to the known, or native, sequence of an antigen. These variations can render the amino acid sequence of the derivative antigen different than the known antigen, yet still capable of eliciting an immune response in a subject. For example, the derivative can have 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36 ,37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80,81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, or more mutated amino acid residues compared to the original, known, or native sequence. These differences can be engineered, of can have occurred naturally through evolution of the virus into different variants. The composition can comprise one or more antigenic proteins or fragments thereof of the spike (S) protein. The S protein is a glycosylated type I membrane protein that consists of two subunits, S1 and S2. The S protein exists in a trimeric pre- fusion form that is later cleaved by a host furin protease into the two subunits S1 and S2. The N-terminal S1 subunit contains the receptor-binding domain (RBD), which mediates binding to the host cell receptor, namely the angiotensin converting enzyme 2 (ACE2) for both SARS-CoV and SARS-CoV-2. Binding of RBD to ACE2, followed by additional cleavage of the S2 subunit at a second specific site by the host serine protease TMPRSS2, are fundamental to trigger the disassociation between S1 and S2, leading to the conformational changes in S2 that are responsible for the fusion of viral and host membranes and virus entry. The composition can be bivalent, or can be multivalent, meaning it can comprise two, three, four, five, or more antigens or fragments thereof. Multiple antigenic proteins or fragments thereof in the same composition can all come from SARS-CoV-2. These multiple antigenic proteins or fragments thereof can be from the same variant, or can be from different variants. Furthermore, the antigenic proteins or fragments thereof can be from different viruses. This is discussed in more detail below. In one specific example, the antigenic protein or fragments of the composition comprises the S protein or an antigenic fragment thereof. The spike protein antigen of the composition can comprise the S1 subunit, the S2 subunit, or both. An example of the spike protein can be found in SEQ ID NO: 1. The spike protein antigen can comprise the receptor-binding domain (RBD). The RBD can be found, by way of example, in residues 336-516 of SEQ ID NO: 1. The RBD is a short immunogenic fragment from a virus that binds to a specific endogenous receptor sequence to gain entry into host cells. Specifically, these refer to a part of the spike glycoprotein (S- domain) which is needed to interact with endogenous receptors to facilitate membrane fusion and delivery to the cytoplasm. Typically, the S-domain is also the site of neutralizing antibodies. The antigenic protein or fragments of the composition can also comprise the N protein or an antigenic fragment thereof. The N protein plays a multifaceted role in the infection cycle. In SARS-CoV-2, the N protein binds to and packages the viral RNA into ribonucleoprotein RNP complexes. The N protein is recruited at the replication-transcription complex by Nsp3 and thus, plays a role in viral genome replication. In particular, this interaction involves the N-terminal domains of Nsp3 and the N protein and its function is to guide the viral genome to the newly assembled replication complex. The antigenic protein or fragments thereof of the composition can also comprise the M protein or an antigenic fragment thereof. The M protein is embedded in the viral membrane, through three predicted transmembrane helices. Its role is to drive the assembly of new virions within the host cells. Coronaviruses M proteins oligomerize at the membrane of Golgi-endoplasmic reticulum intermediary compartment and induce apoptosis. S, N and E proteins are then recruited through interaction with the M protein. The antigenic protein or fragments of the composition can also comprise the E protein or an antigenic fragment thereof. The E protein presents one trans-membrane domain and shows oligomerization properties. Interaction between the C-terminus of E and M proteins guides E recruitment to the Golgi-endoplasmic reticulum intermediary compartment, initiating virus budding into host. In one embodiment, the composition comprises at least one S protein antigen or fragment thereof, and at least one N protein or fragment thereof. For example, the S protein can comprise all or a fragment of the RBD. The sequence and structure of RBD of S protein can be found in Lan et al. (2020), which is hereby incorporated by reference in its entirety for its disclosure concerning the RBD of S protein. The composition can comprise either the S1 subunit, the S2 subunit, or both. In another embodiment, the composition comprises at least two different antigens or fragments thereof from S protein and at least one N protein or fragment thereof. For example, the S1 and S2 proteins can both be included in the composition as separate antigens, as well as the N protein. In another embodiment, the composition comprises one or more S proteins or fragments thereof, one or more N proteins or fragments thereof, and at least one other antigen from SARS-CoV-2. This further antigen can be from the M protein, the E protein, or any other proteins associated with SARS-CoV-2. In yet another embodiment, the composition comprises one or more S proteins or fragments thereof, one or more N proteins or fragments thereof, and at least one other antigen from another virus. For example, the further antigen can be from influenza. The composition can comprise the SARS-CoV-2 antigens described above, and can further comprise antigens to one, two, three, four, five, or more other antigens. When more than one additional antigen is provided from another virus, the antigens can be from the same strain or from multiple strains. Again, using the example of influenza, the multiple antigens can be to different hemagglutinin subtypes. Eighteen different phylogenetically distinct subtypes of HA have emerged in influenza A viruses (H1 to H18), and each can be used to as a distinct antigen in a vaccine. Examples of antigens other than those derived from SARS-CoV-2 that can be used in a multivalent vaccine with SARS-CoV-2 antigens include, but are not limited to, influenza virus, cytomegalovirus, avian leukosis-sarcoma virus (ALV), Rous Sarcoma virus (RSV), Mammalian C-type Murine leukemia virus (MLV), Feline leukemia virus (FeLV), simian sarcoma virus (SIS), B-type viruses like Mouse mammary tumor virus (MMTV), D-type viruses like Mason-Pfizer monkey virus (MPMV), Simian AIDS viruses (SRVs), HTLV-BLV group such as Human T-cell leukemia virus (HTLV), Simian T-cell leukemia virus (STLV), bovine leukemia virus (BLV). Lentivirinae comprise Human immunodeficiency virus (HIV-1 and HIV-2), Simian immunodeficiency virus (SIV), Feline immunodeficiency virus (FIV), Visna/maedi virus (MV), Equine infectious anemia virus (EIAV), Caprine arthritis- encephalitis virus (CAEV). Spumavirinae or “Foamy viruses” like Human (HSRV), Simian (SSRV), Feline (FSRV), Bovine (BSRV), Murine (MSRV), endogenous retroviruses (ERV), papilloma virus, respiratory syncytial virus, poliomyelitis virus, pox virus, measles virus, arbor virus, Coxsackie virus, herpes virus, hantavirus, hepatitis virus, Baculovirus, mumps virus, circovirus, vichaivirus, arenavirus, or rotavirus. A bacteria may be a member of the genus Neisseria, Aerobacter, Pseudomonas, Porphyromonas, Salmonella, Escherichia, Pasteurella, Shigella, Bacillus, Helibacter, Corynebacterium, Clostridium, Mycobacterium, Yersinia, Staphylococcus; Bordetelia, Brucelia, Vibrio, Streptococcus, Plasmodium, Schisostoma, Candida. Any microbial infections, which are present and/or transmitted as Zoonoses, Cyclozoonoses, Metazoonoses, Saprozoonoses, Anthropozoonoses, Zooanthropozoonoses and Amphixenoses, are encompassed by the present invention. The invention in addition to whole pathogens also encompasses a single antigen or a plurality of antigens from such pathogens, e.g., HIV antigens: gp160, gag, pol, Nef, Tat, and Rev; the malaria antigens: CS protein and Sporozoite surface protein 2; the Hepatitis B surface antigens: Pre-S1, Pre-S2, HBc Ag, and HBe Ag; the influenza antigens: HA, NP and NA; Hepatitis A surface antigens; the Herpes virus antigens: EBV gp340, EBV gp85, HSV gB, HSV gD, HSV gH, HSV early protein product, cytomegalovirus gB, cytomegalovirus gH, and IE protein gp72; the respiratory syncytial virus antigens: F protein, G protein, and N protein or fragments thereof. Adjuvants Compositions of the invention can be administered in conjunction with other immunoregulatory agents, including adjuvants. As used herein, the term "adjuvant" refers to a compound or mixture that enhances an immune response. In particular, the compositions disclosed herein can include an adjuvant. The invention can also comprise combinations of aspects of one or more of the adjuvants identified herein. Many adjuvants contain a substance designed to protect the antigen from rapid catabolism, such as aluminum hydroxide or mineral oil, and a stimulator of immune responses, such as lipid A, Bortadella pertussis or Mycobacterium tuberculosis derived proteins. Certain adjuvants are commercially available as, for example, Freund's Incomplete Adjuvant and Complete Adjuvant (Difco Laboratories, Detroit, Mich.); Merck Adjuvant 65 (Merck and Company, Rahway, N.J.); AS-2 (GlaxoSmithKline, Philadelphia, Pa.); aluminum salts such as aluminum hydroxide gel (alum) or aluminum phosphate; salts of calcium, iron or zinc; an insoluble suspension of acylated tyrosine; acylated sugars; cationically or anionically derivatized polysaccharides; polyphosphazenes; biodegradable microspheres; monophosphoryl lipid A and quil A. Cytokines, such as GM-CSF, interleukin-2, -7, - 12, and other like growth factors, may also be used as adjuvants. The adjuvant composition can be a composition that induces an anti- inflammatory immune response (antibody or cell-mediated). Accordingly, high levels of anti-inflammatory cytokines (anti-inflammatory cytokines may include, but are not limited to, interleukin 4 (IL-4), interleukin 5 (IL-5), interleukin 10 (IL-10), and transforming growth factor beta (TGFβ^^^Optionally, an anti-inflammatory response would be mediated by CD4+ T helper cells. Bacterial flagellin has been shown to have adjuvant activity (McSorley et al., J. Immunol. 169:3914-19, 2002). Also disclosed are polypeptide sequences that encode flagellin proteins that can be used in adjuvant compositions. Optionally, the adjuvants used in conjunction with the disclosed compositions increase lipopolysaccharide (LPS) responsiveness. Illustrative adjuvants include but are not limited to, monophosphoryl lipid A (MPL), aminoalkyl glucosaminide 4- phosphates (AGPs), including, but not limited to RC-512, RC-522, RC-527, RC-529, RC-544, and RC-560 (Corixa, Hamilton, Mont.). In addition, the adjuvant composition can be one that induces an immune response predominantly of the Th1 type. High levels of Th1-type cytokines tend to favor the induction of cell mediated immune responses to an administered antigen. In contrast, high levels of Th2-type cytokines (e.g., IL-4, IL-5, IL-6 and IL-10) tend to favor the induction of humoral immune responses. Following application of a vaccine as provided herein, a subject will support an immune response that includes Th1- and Th2-type responses. Optionally, the level of Th1-type cytokines will increase to a greater extent than the level of Th2-type cytokines. The levels of these cytokines may be readily assessed using standard assays. The level of Th2-type cytokines can increase to a greater extent than the level of Th1-type cytokines. Certain adjuvants for eliciting a predominantly Th1-type response include, for example, a combination of monophosphoryl lipid A, preferably 3-de-O-acylated monophosphoryl lipid A, together with an aluminum salt adjuvants are available from Corixa Corporation (Seattle, Wash.; see, for example, U.S. Pat. Nos. 4,436,727; 4,877,611; 4,866,034 and 4,912,094, which are hereby incorporated by reference for their teaching of the same). CpG-containing oligonucleotides (in which the CpG dinucleotide is unmethylated) also induce a predominantly Th1 response. Such oligonucleotides are well known and are described, for example, in WO 96/02555, WO 99/33488 and U.S. Pat. Nos. 6,008,200 and 5,856,462. Immunostimulatory DNA sequences are also described, for example, by Sato et al., Science 273:352, 1996. Another adjuvant comprises a saponin, such as Quil A, or derivatives thereof, including QS21 and QS7 (Aquila Biopharmaceuticals Inc., Framingham, Mass.); Escin; Digitonin; or Gypsophila or Chenopodium quinoa saponins. Other formulations can include more than one saponin in the adjuvant combinations, for example combinations of at least two of the following group comprising QS21, QS7, Quil A, β-escin, or digitonin. Saponin formulations can also be combined with vaccine vehicles composed of chitosan or other polycationic polymers, polylactide and polylactide-co-glycolide particles, poly-N-acetyl glucosamine-based polymer matrix, particles composed of polysaccharides or chemically modified polysaccharides, particles composed of glycerol monoesters, etc. Furthermore, the saponins may be formulated together with a polyoxyethylene ether or ester, in either a non-particulate solution or suspension, or in a particulate structure. The saponins can also be formulated with excipients such as CARBOPOLTM (Noveon, Cleveland, Ohio) to increase viscosity, or may be formulated in a dry powder form with a powder excipient such as lactose. Optionally, the adjuvant system includes the combination of a monophosphoryl lipid A and a saponin derivative, such as the combination of QS21 and 3D-MPL. adjuvant, as described in WO 94/00153, or a less reactogenic composition where the QS21 is quenched with cholesterol, as described in WO 96/33739. Other formulations comprise an oil-in-water emulsion and tocopherol. Another adjuvant formulation employing QS21, 3D-MPL.RTM. adjuvant and tocopherol in an oil-in-water emulsion is described in WO 95/17210. Another enhanced adjuvant system involves the combination of a CpG- containing oligonucleotide and a saponin derivative particularly the combination of CpG and QS21 is disclosed in WO 00/09159. Optionally the formulation additionally comprises an oil in water emulsion and tocopherol. Additional illustrative adjuvants for use in the disclosed compositions Montamide ISA 720 (Seppic, France), SAF (Chiron, Calif., United States), ISCOMS (CSL), MF-59 (Sequirus), the SBAS series of adjuvants (e.g., SBAS-2 or SBAS-4, available from GlaxoSmithKline, Philadelphia, Pa.), Detox (EnhanzynTM) (Corixa, Hamilton, Mont.), RC-529 (Corixa, Hamilton, Mont.) and other aminoalkyl glucosaminide 4-phosphates (AGPs), such as those described in pending U.S. patent application Ser. Nos. 08/853,826 and 09/074,720, the disclosures of which are incorporated herein by reference in their entireties, and polyoxyethylene ether adjuvants such as those described in WO 99/52549A1. Specifically, the adjuvant disclosed herein can be monosodium urate crystal (WO2004100984A1). Other specific adjuvants contemplated herein include CpG- ODN, ADU-S100, and Poly(I:C). ADU-S100 has increased stability and high affinity for mouse and human Stimulator of Interferon Genes (STING). A TLR-3 adjuvant poly(I:C) is a potent mucosal adjuvant for intranasal delivery of vaccines shown for killed influenza virus antigens in a vaccine trial in pigs (Renu 2020). A TLR-9 adjuvant CPG-ODN is known to induce antigen-specific sIgA antibody response in mucosal surfaces in intranasal vaccinated pigs against two respiratory viruses (Dhakal 2019; Zhang 2007). Uric acid crystals activate innate host defense mechanisms and trigger robust inflammation and immune activation through NLRP3 inflammasome pathway (Braga 2017). The innate immune activation by monosodium urate (MSU) crystal potentiates the adaptive immune response, such as the antibody response (Ng 2010). MSU crystals are shown to be safe after intradermal injection (2-2000μg) in humans and non-toxic (Sakamaki 2011; Cheng 2014). The combination adjuvant targets multiple signaling pathways resulting in synergistic activation of immune cells and a balanced immune response. CpG55.2 (Vaxine Pty Ltd) is a proprietary 24mer class B CpG oligonucleotide that is a potent activator of both human and mouse TLR9. However, TLR9 agonists alone may be only weakly effective and Papagona et al. (Papagno, L., et al., The TLR9 ligand CpG ODN 2006 is a poor adjuvant for the induction of de novo CD8+ T-cell responses in vitro. Scientific Reports, 2020. 10(1): p. 11620.) reported CpG ODN2006, a synthetic oligonucleotide TLR9 ligand, promoted antigen-driven expansion and functional maturation of naive CD8+ T cells ineffectively compared to other TLR adjuvants. TLR9 adjuvants may work optimally when co-formulated with particulate adjuvant delivery systems such as alum or delta inulin. Advax-CpG adjuvant (Vaxine Pty Ltd) is a combination of delta inulin polysaccharide (Gordon D.L., Sajkov D., Woodman R.J., Honda-Okubo Y., Cox M.M.J., Heinzel S., et al. Randomized clinical trial of immunogenicity and safety of a recombinant H1N1/2009 pandemic influenza vaccine containing Advax™ polysaccharide adjuvant. Vaccine. 2012;30:5407–5416) formulated with CpG oligonucleotide, a toll- like receptor 9 (TLR9) agonist that has been shown to enhance humoral and T cell responses in a broad range of animal species as well as humans, having low reactogenicity and a strong safety profile (Gordon D., Kelley P., Heinzel S., Cooper P., Petrovsky N. Immunogenicity and safety of Advax™, a novel polysaccharide adjuvant based on delta inulin, when formulated with hepatitis B surface antigen: a randomized controlled Phase 1 study. Vaccine. 2014;32:6469–6477; Honda-Okubo Y., Saade F., Petrovsky N. Advax™, a polysaccharide adjuvant derived from delta inulin, provides improved influenza vaccine protection through broad-based enhancement of adaptive immune responses. Vaccine. 2012;30:5373–5381; Li L., Honda-Okubo Y., Huang Y., Jang H., Carlock M.A., Baldwin J., et al. Immunisation of ferrets and mice with recombinant SARS-CoV-2 spike protein formulated with Advax-SM adjuvant protects against COVID-19 infection. Vaccine. 2021;39:5940–5953). It is effective and well-tolerated in humans and has been shown to be highly effective and safe in newborn (Honda-Okubo Y., Ong C.H., Petrovsky N. Advax delta inulin adjuvant overcomes immune immaturity in neonatal mice thereby allowing single-dose influenza vaccine protection. Vaccine. 2015;33:4892–4900; Sakala I.G., Honda- Okubo Y., Li L., Baldwin J., Petrovsky N. A M2 protein-based universal influenza vaccine containing Advax-SM adjuvant provides newborn protection via maternal or neonatal immunization. Vaccine. 2021;39:5162–5172) and pregnant mice (Honda- Okubo Y., Kolpe A., Li L., Petrovsky N. A single immunization with inactivated H1N1 influenza vaccine formulated with delta inulin adjuvant (Advax™) overcomes pregnancy-associated immune suppression and enhances passive neonatal protection. Vaccine. 2014;32:4651–4659; Eichinger K.M., Kosanovich J.L., Lipp M.A., Perkins T.N., Petrovsky N., Marshall C., et al. Maternal immunization with adjuvanted RSV prefusion F protein effectively protects offspring from RSV challenge and alters innate and T cell immunity. Vaccine. 2020;38:7885–7891). Advax-CpG adjuvant efficacy has been demonstrated to be effective in vaccines against coronavirus disease 2019 (COVID)-19 (Li L, Honda-Okubo Y, Huang Y, Jang H, Carlock MA, Baldwin J, et al.. Immunisation of Ferrets and Mice With Recombinant SARS-CoV-2 Spike Protein Formulated With Advax-SM Adjuvant Protects Against COVID-19 Infection. Vaccine (2021) 39(40):5940–53. doi: 10.1016/j.vaccine.2021.07.087; Tabarsi P, Anjidani N, Shahpari R, Mardani M, Sabzvari A, Yazdani B, Roshanzamir K, Bayatani B, Taheri A, Petrovsky N, Li L, Barati S. Safety and immunogenicity of SpikoGen®, an Advax-CpG55.2-adjuvanted SARS-CoV-2 spike protein vaccine: a phase 2 randomized placebo-controlled trial in both seropositive and seronegative populations. Clin Microbiol Infect. 2022 Sep;28(9):1263-1271. doi: 10.1016/j.cmi.2022.04.004), among others. Nanoparticles The compositions, immunogenic compositions and vaccines described herein can comprise one or more nanoparticles. Examples of nanoparticles (used interchangeably with the term “nanocarrier”) include, but are not limited to, nanocarriers composed of one or more polymers. In some embodiments, the one or more polymers is a water soluble, non-adhesive polymer. In some embodiments, polymer is polyethylene glycol (PEG) or polyethylene oxide (PEO). In some embodiments, the polymer is polyalkylene glycol or polyalkylene oxide. In some embodiments, the one or more polymers is a biodegradable polymer. In some embodiments, the one or more polymers is a biocompatible polymer that is a conjugate of a water soluble, non-adhesive polymer and a biodegradable polymer. In some embodiments, the biodegradable polymer is polylactic acid (PLA), poly(glycolic acid) (PGA), or poly(lactic acid/glycolic acid) (PLGA). In some embodiments, the nanocarrier is composed of PEG-PLGA polymers. In some embodiments, the nanocarrier is formed by self-assembly. Self- assembly refers to the process of the formation of a nanocarrier using components that will orient themselves in a predictable manner forming nanocarriers predictably and reproducibly. In some embodiments, the nanocarriers are formed using amphiphillic biomaterials which orient themselves with respect to one another to form nanocarriers of predictable dimension, constituents, and placement of constituents. In some embodiments, the nanocarrier is a microparticle, nanoparticle, or picoparticle. In some embodiments, the microparticle, nanoparticle, or picoparticle is self-assembled. In some embodiments, the nanocarrier has a positive zeta potential. In some embodiments, the nanocarrier has a net positive charge at neutral pH. In some embodiments, the nanocarrier comprises one or more amine moieties at its surface. In some embodiments, the amine moiety is a primary, secondary, tertiary, or quaternary amine. In some embodiments, the amine moiety is an aliphatic amine. In some embodiments, the nanocarrier comprises an amine-containing polymer. In some embodiments, the nanocarrier comprises a protein or a peptide that is positively charged at neutral pH. In some embodiments, the nanocarrier is a latex particle. In some embodiments, the nanocarrier with the one or more amine moieties on its surface has a net positive charge at neutral pH. Nanoparticles can aid the delivery of the antigens disclosed herien, and/or can also be immunogenic. Delivery can be to a particular site of interest, e.g. the mucosa. In some embodiments, the nanoparticle can create a timed release of antigens to enhance and/or extend the immune response. In some embodiments, the nanoparticle is associated with the antigens such that the composition can elicit an immune response. The association can be, for example, wherein the nanoparticle is entrapped or encapsulated with the SARS-CoV-2 antigens and/or other antigens. By entrapped is meant that there is a physical encasing the antigens in nanoparticles. In some embodiments, the antigens are entrapped within the nanoparticle by a water/oil/water emulsion method. In some embodiments, the nanoparticle is poly(lactide co- glycolide) (PLGA). Depending on the ratio of lactide to glycolide used for the polymerization, different forms of PLGA can be obtained and utilized. These forms are typically identified in regard to the monomers' ratio used (e.g. PLGA 75:25 identifies a copolymer whose composition is 75% lactic acid and 25% glycolic acid). Different ratios can be used in this invention, e.g. 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80, 10:90, and numbers above and in between these ratios. Additional examples of suitable nanoparticles include chitosin, calcium phosphate, lipids of various bacteria like E. coli, mycobactera, leptospira and mixtures thereof. In one example, the composition can be derived mixing about 180 mg of PLGA to about 5 mg of antigenic material (or about 36 mg PLGA to 1mg antigenic material). The entrapment (encapsulation) efficiency of antigens can vary. In one embodiment the nanoparticle were 50-55% entrapped/encapsulated, calculated based on amount of total antigens used in the entrapment. Entrapped antigens can be administered as mixtures of entrapped/encapsulated and unentrapped/unencapsulated antigens or the entrapped/encapsulated antigens can be further purified. Additional Ingredients Additional compounds suitable for use with the compositions of the invention include, but are not limited to, one or more solvents, such as an organic phosphate- based solvent, bulking agents, coloring agents, pharmaceutically acceptable excipients, a preservative, pH adjuster, buffer, chelating agent, etc. The additional compounds can be admixed into a previously formed composition comprising a nanoparticle, or the additional compounds can be added to the original mixture to be emulsified. In certain of these embodiments, one or more additional compounds are admixed into an existing compositions immediately prior to its use. Suitable preservatives for use with the compositions of the invention include, but are not limited to, cetylpyridinium chloride, benzalkonium chloride, benzyl alcohol, chlorhexidine, imidazolidinyl urea, phenol, potassium sorbate, benzoic acid, bronopol, chlorocresol, paraben esters, phenoxyethanol, sorbic acid, alpha- tocophernol, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, sodium ascorbate, sodium metabisulphite, citric acid, edetic acid, semi-synthetic derivatives thereof, and combinations thereof. Other suitable preservatives include, but are not limited to, benzyl alcohol, chlorhexidine (bis (p- chlorophenyldiguanido) hexane), chlorphenesin (3-(-4-chloropheoxy)-propane-1,2- diol), Kathon CG (methyl and methylchloroisothiazolinone), parabens (methyl, ethyl, propyl, butyl hydrobenzoates), phenoxyethanol (2-phenoxyethanol), sorbic acid (potassium sorbate, sorbic acid), Phenonip (phenoxyethanol, methyl, ethyl, butyl, propyl parabens), Phenoroc (phenoxyethanol 0.73%, methyl paraben 0.2%, propyl paraben 0.07%), Liquipar Oil (isopropyl, isobutyl, butylparabens), Liquipar PE (70% phenoxyethanol, 30% liquipar oil), Nipaguard MPA (benzyl alcohol (70%), methyl & propyl parabens), Nipaguard MPS (propylene glycol, methyl & propyl parabens), Nipasept (methyl, ethyl and propyl parabens), Nipastat (methyl, butyl, ethyl and propyel parabens), Elestab 388 (phenoxyethanol in propylene glycol plus chlorphenesin and methylparaben), and Killitol (7.5% chlorphenesin and 7.5% methyl parabens). The compositions disclosed herein can further comprise at least one pH adjuster. Suitable pH adjusters include, but are not limited to, diethyanolamine, lactic acid, monoethanolamine, triethylanolamine, sodium hydroxide, sodium phosphate, semi-synthetic derivatives thereof, and combinations thereof. In addition, the nanoemulsion vaccine can comprise a chelating agent. In one embodiment of the invention, the chelating agent is present in an amount of about 0.0005% to about 1%. Examples of chelating agents include, but are not limited to, ethylenediamine, ethylenediaminetetraacetic acid (EDTA), phytic acid, polyphosphoric acid, citric acid, gluconic acid, acetic acid, lactic acid, and dimercaprol, and a preferred chelating agent is ethylenediaminetetraacetic acid. The compositions disclosed herein can further comprise a buffering agent, such as a pharmaceutically acceptable buffering agent. Examples of buffering agents are known to those of skill in the art. The vaccine can also comprise one or more emulsifying agents to aid in the formation of emulsions. Emulsifying agents include compounds that aggregate at the oil/water interface to form a kind of continuous membrane that prevents direct contact between two adjacent droplets. The compositions disclosed herein can be readily diluted with water or another aqueous phase to a desired concentration without impairing their desired properties. Vaccines Specifically disclosed herein is a vaccine comprising the compositions disclosed herein, wherein said composition is a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof in a pharmaceutically acceptable carrier. Intranasal administration includes administration via the nose, either with or without concomitant inhalation during administration. Such administration is typically through contact by the composition comprising the vaccine with the nasal mucosa, nasal turbinates or sinus cavity. Administration by inhalation comprises intranasal administration, or may include oral inhalation. Such administration may also include contact with the oral mucosa, bronchial mucosa, and other epithelia. Exemplary dosage forms for pharmaceutical administration are described herein. Examples include but are not limited to liquids, ointments, creams, emulsions, lotions, gels, bioadhesive gels, sprays, aerosols, pastes, foams, sunscreens, capsules, microcapsules, suspensions, pessary, powder, semi-solid dosage form, etc. The pharmaceutical compositions may be formulated for immediate release, sustained release, controlled release, delayed release, or any combinations thereof, into the epidermis or dermis. In some embodiments, the formulations may comprise a penetration-enhancing agent. Suitable penetration-enhancing agents include, but are not limited to, alcohols such as ethanol, triglycerides and aloe compositions. The amount of the penetration-enhancing agent may comprise from about 0.5% to about 40% by weight of the formulation. The vaccines of the invention can be applied and/or delivered utilizing electrophoretic delivery/electrophoresis. Further, the composition may be a transdermal delivery system such as a patch or administered by a pressurized or pneumatic device (i.e., “gene gun”). Such methods, which comprise applying an electrical current, are well known in the art. The pharmaceutical compositions for administration may be applied in a single administration or in multiple administrations. For example, one dose can be placed in each nostril during vaccination. For example, bi-dose delivery can be used with the vaccines according to the invention. Bi-dose devices contain two sub-doses of a single vaccine dose, one sub-dose for administration to each nostril. Generally, the two sub-doses are present in a single chamber and the construction of the device allows the efficient delivery of a single sub-dose at a time. Alternatively, a mono-dose device may be used for administering the vaccines according to the invention. The vaccine can be given in one, two, three, four, or more doses, so that the subject is given a first dose (which can be a bi-dose or mono-dose, as described above), and then a second dose is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26,27, 28, 29, or 30 days, or 4 ,5, 6, 7, 8, 9, 10, 11, or 12 weeks, or 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months, or 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more years apart. The subject can also receive a booster dose, which can be given at any time after the initial dose or doses. The booster dose can comprise the same, or different, antigens than the initial vaccination(s). For example, the vaccine can be updated with more recent variant antigens. Most preferably, the vaccine is administered locally to the nasopharyngeal area. Preferred devices for intranasal administration of the vaccines according to the invention are spray devices. Suitable commercially available nasal spray devices include Accuspray™ (Becton Dickinson). Nebulizers produce a very fine spray (such as a mist) which can be easily inhaled and are also contemplated herein. Preferred spray devices for intranasal use are devices for which the performance of the device is not dependent upon the pressure applied by the user. These devices are known as pressure threshold devices. Liquid is released from the nozzle only when a threshold pressure is applied. These devices make it easier to achieve a spray with a regular droplet size. Pressure threshold devices suitable for use with the present invention are known in the art The invention provides in a further aspect a pharmaceutical kit comprising an intranasal administration device as described herein containing a vaccine formulation according to the invention. The invention is not necessarily limited to spray delivery of liquid formulations. Vaccines according to the invention may be administered in other forms e.g. as a powder. Methods Disclosed herein are methods of eliciting or enhancing an immune response to SARS-CoV-2 in a subject, the method comprising administering to the subject a vaccine, wherein the vaccine comprises a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof; wherein the vaccine further comprises a pharmaceutically acceptable carrier. The subject, or patient, being given the vaccine can be a mammal, such as a human. The subject may have previously been vaccinated against SARS-CoV-2, for example by intramuscular injection. Such vaccination does not preclude the patient from also receiving the vaccine disclosed herein. For example, the vaccine that was initially given can be against a different variant, or the same variant. In the case of a multivalent vaccine, the subject may have been exposed to one, or all, of the antigens in the vaccine disclosed herein, previous to the vaccine of the present invention being administered. For example, the subject may have previously been vaccinated, or may have natural immunity through exposure. In one example, if the patient has antibodies to SARS-CoV-2 either through natural immunity or vaccination, the subject may be given a smaller dose, or fewer separate doses, than a subject who does not have natural immunity. One of skill in the art can readily determine the proper dosage of the vaccine to be given to the subject. For example, the dosage can be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, or 600 μg per dose, or more, less, or any amount in between these values. Also disclosed is a method of preventing or lessening the severity of symptoms or markers of disease associated with SARS-CoV-2 infection in a subject, the method comprising administering to the subject a vaccine, wherein the vaccine comprises a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS- CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof; wherein the vaccine further comprises a pharmaceutically acceptable carrier. By “preventing” is meant reducing the chance that the subject will be infected with SARS-CoV-2 by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36 ,37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80,81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% compared with a subject who has not been vaccinated with the vaccine disclosed herein. By “lessening severity of symptoms” is meant that the subject contracts SARS-CoV-2, but has less severe symptoms as compared to what symptoms the subject would have had, had they not been vaccinated. For example, the subject can have a “mild” infection instead of a “moderate” infection.
As used herein, “mild” infection refers to patients diagnosed with COVID-19 by a standardized RT-PCR assay exhibiting fever, rhinorrhea, mild cough, sore throat, malaise, headache, muscle pain, malaise, or any combination thereof, but with no shortness of breath. Patients with “mild” infection present no signs of a more serious lower airway disease and have a respiratory rate of less than 20 breaths per minute, a heart rate of less than 90 beats per minute, and oxygen saturation (pulse oximetry) greater than 93% on room air.
As used herein, “moderate” infection refers to patients diagnosed with COVID-19 by a standardized RT-PCR assay exhibiting symptoms in the mild category and additional symptoms. These include more significant lower respiratory symptoms, including shortness of breath (at rest or with exertion) or signs of moderate pneumonia, including a respiratory rate of ≥ 20 but <30 breaths per minute, a heart rate of ≥ 90 but <125 beats per minute and oxygen saturation (pulse oximetry ) greater than 93% on room air. If some embodiments, subjects with moderate infection further exhibit lung infiltrates based on X-ray or CT scan that are <50% present.
As used herein “mild-to-moderate” infection collectively refers to mild and moderate infections, as defined herein.
As used herein, “severe” infection refers to patients diagnosed with COVID- 19 by a standardized RT-PCR. assay having significant lower respiratory symptoms, including difficulty in breathing or shortness of breath at rest or one or more of the following signs of severe pneumonia: a. respiratory rate ≥ 30 breaths per minute, oxygen saturation (pulse oximetry) ≤93% on room air, partial pressure of oxygen/fraction of inspired oxygen (PaO2/FiO2) ≤300 mmHg (1 mmHg=0.133 kPa). Additionally, clinical assessment shows evidence of rales/ crackles on exam or if available, radiographic evidence of pulmonary' infiltrates (chest x-ray, CT scan, etc.).
As used herein “critical” infection refers to a severe infection in which the patient has at least one of the following: (1) respiratory failure requiring at least one of the following: Endotracheal intubation and mechanical venti lation, oxygen delivered by high-flow nasal cannula, noninvasive positive pressure ventilation, or ECMO; (2) a clinical diagnosis of respiratory failure (in setting of resource limitation); (3) Septic shock (defined by SBP <90 mm Hg, or Diastolic BP<60 mm Hg); and (4) Multiple organ dysfunction/failure.
In some embodiments, vaccination of a subject according to the methods described herein results in reduction of one or more inflammatory cytokines and/or chemokines. In some embodiments, the inflammatory cytokine or chemokine is a cytokine or chemokine listed in FIGS. 9A-9J (e.g., sCD40L, EGF, Eotaxin (CCL11), FGF-2, Flt-3 ligand, Fractalkine, G-CSF, GM-CSF, GROα (CXCL1), IFN-α2, IFN-γ, IL-1α, IL-1β, IL-1RA, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (CXCL8), IL-9, IL-10, IL-12 (p40), IL-12 (p70), IL-13, IL-15, IL-17A, IL-17E/IL-25, IL-17F, IL-18, IL-22, IL-27, IP-10 (CXCL10), MCP-1 (CCL2), MCP-3, M-CSF, MDC (CCL22), MIG (CXCL9), MIP-1α (CCL3), MIP-1β (CCL4), PDGF-AA, PDGF-AB/BB, RANTES (CCL5), TGF-α, TNF-α, TNF-β, TNF-r1, or VEGF-A), in FIG. 12A-D (CCL5, IL-6, IL-8, IL-1β), in FIGS. 17-19, CCL5, IL-5, IL-13, IL-2, IL-6, IL-10, IL-9, IFN-γ, TNF-α. IL-17A, IL-17F, IL-4, IL-21, IL-22, or any combination thereof. In some embodiments, the inflammatory cytokine and/or chemokine may comprise a cytokine or chemokine selected from one of CCL5, sCD40L, EGF, Eotaxin, FGF-2, Flt-3 ligand, Fractalkine, G-CSF, GM-CSF, GROα, IFN-α2, IFN-γ, IL-1α, IL-1β, IL-1ra, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12 (p40), IL-12 (p70), IL-13, IL-15, IL-17A, IL-17E/IL-25, IL-17F, IL-18, IL-21, IL-22, IL-27, IP-10, MCP-1, MCP-3, M-CSF, MDC (CCL22), MIG, MIP-1α, MIP-1β, PDGF-AA, PDGF-AB/BB, RANTES, TGF-α, TNF-α, TNF-β, TNF-r1, C reactive protein (CRP), VEGF-A or any combination thereof. In some embodiments, the one or more inflammatory cytokines and/or chemokines comprises CCL5, IL-6, IL-8, IL-1β, IL-10, TNF-α, or any combination thereof. In some embodiments, the one or more inflammatory cytokines and/or chemokines comprises CCL5, IL-6, TNF-α, or a combination thereof. In some embodiments, the one or more inflammatory cytokines and/or chemokines comprises CCL5, IL-6, IL-1β, IL-8, or any combination thereof. The level of the inflammatory cytokine and/or chemokines may be measured in blood plasma, e.g., by enzyme- linked immunosorbent assays (ELISA), bead-based immunoassays and other immunoassays. The transcriptional level of inflammatory cytokines and/or chemokines may also be measured by RNA sequencing. In some embodiments, vaccination of a subject according to the methods described herein results in reduction of a symptom associated hyperinflammation. In some embodiments, hyperinflammation is cytokine release syndrome, hemophagocytic lymphohistiocytosis (HLH), macrophage activation syndrome, or acute respiratory distress syndrome (ARDS). In some embodiments, vaccination of a subject according to the methods described herein reduces migration of CCR5+ immune cells. In some embodiments, the CCR5+ immune cells comprise macrophages, T cells, or both. In some embodiments, vaccination of a subject according to the methods described herein improves at least one respiratory parameter in the subject. In some embodiments, vaccination of a subject according to the methods described herein increases oxygen saturation in the subject. In some embodiments, vaccination of a subject according to the methods described herein results in reduced occurrence or risk of developing liver toxicity, kidney failure or a coagulation event. In some embodiments, the coagulation event comprises a blood clot, stroke, or pulmonary embolism. In some embodiments, treatment results in more normalization of kidney function. Kidney function may be measured by measuring blood levels of creatine, BUN, sodium, or any combination thereof in the subject blood. In some embodiments, vaccination results in more normalization of liver function. Liver function may be measured by measuring blood levels of bilirubin, alanine transaminase (ALT), aspartate aminotransferase (AST), or any combination thereof. In some embodiments, vaccination of a subject according to the methods described herein results in reduction of SARS-CoV-2 viral load in the subject. In some embodiments, vaccination of a subject according to the methods described herein results in reduced duration or occurrence of hospitalization, ventilation or dialysis of the subject. In some embodiments, vaccination of a subject according to the methods described herein results in reduced lung damage to the subject. In some embodiments, vaccination of a subject according to the methods described herein results in the subject having a faster and/or more extensive recovery than a subject with similar symptoms who has not been vaccinated with the vaccine disclosed herein. A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims. EXAMPLES Example 1: Nanotechnology Based Intranasal Vaccine for COVID-19 One-year-old male ferrets were used in this study. Ferrets (n=6 per group) were inoculated intranasally twice at 3 weeks interval with lipid nanoparticle entrapped subunit SARS-CoV-2 vaccine containing receptor binding domain (RBD) of the spike protein and nucleocapsid protein (N) along with monosodium urate crystal (MSU) adjuvant, control proteins with adjuvant or empty liposomes and then challenged with the SARS-CoV-2 (1 x106 TCID50) via intranasal route. Ferrets were sampled at the indicated timepoints before necropsy. Three animals in each group were euthanized at 7- and 14-days post challenge infection and collected nasal swab, nasal turbinate, trachea, lung, urine and intestine for analysis of infectious virus and RNA load and for immune gene expression analyses (Figure 1). Materials and Methods Production SARS-CoV-2 proteins Synthetic nucleic acids encoding the SARS-CoV-2 S-RBD (amino acids 319- 516) or full length nucleocapsid (N) were codon optimized for expression in E. coli and ordered as gblock gene fragments (IDTDNA). Fragments were cloned into a pRSET A bacterial expression plasmid (Invitrogen) utilizing BamHI/HindIII. Plasmids were verified by restriction digest and Sanger sequencing. Plasmids were used to transform BL21(DE3) E. coli (NEB). Protein was induced by autoinduction (54). Bacteria were lysed using bacterial protein extraction reagent (BPER) (Thermo Fisher) with HALT protease inhibitors (Thermo Fisher). Insoluble material including protein of interest was pelleted spinning at 10,000 x G for 10 minutes. Insoluble pellets were washed twice in inclusion body wash buffer (20mM Tris-HCl, pH7.5, 10mM EDTA, 1% triton X-100), resolubilized using 50 mM CAPS, pH 11.0, 1% N- lauroylsarcosine, and 1mM dithiothreitol (DTT) with end-over-end mixing at room temperature for 30 minutes. Soluble protein was dialyzed 3x against 20mM Tris-HCl, pH 8.5 with 0.1mM DTT [2]. Dialyzed proteins were purified by nickel affinity chromatography (Hispur kit, Thermo Fisher 88229). Purified proteins were denatured by boiling in Laemmli sample buffer, separated by SDS polyacrylamide gel electrophoresis followed by Coomassie blue staining. Proteins were stored at -80 until use. Synthesis of MSU crystal adjuvant MSU crystals were synthesized by following the procedure described previously (25, 56, 58), which yielded the crystals with similar morphology and birefringence to those found in gout patients. Briefly, 1.68 g of solid uric acid was added to 400 mL sodium hydroxide solution (0.4 g of NaOH, 25 mM). The resultant opaque solution was allowed to remain overnight at 80 qC and the filtrate was rinsed with cold distilled water three times and air dried in the fume hood for 2 days. The dried MSU particles were sieved into a size range of 1-5 μm in length and were nano- sized in diameter. They were divided into 5 mg aliquots, dispensed into individual vials, and sterilized by ethylene trioxide. The MSU crystals were then entrapped. Experimental Animals Neutered male ferrets, 12-months-old, were obtained (Triple F Farms, PA) for use in this study. Ferrets were seronegative for influenza A viruses, MERS-CoV, and SARS-CoV-2. All ferrets were housed (3 per cage, 2 cages per experimental group) with a 12 h light/dark cycle and allowed access to food and water ad libitum. All animal studies were carried out in accordance with protocols approved by the Institutional Animal Care and Use Committee (IACUC) at OSU. Baseline body weights and temperatures were measured before vaccination and challenge infection. Ferrets were housed at a BSL-1 facility for approximately 7 weeks during the immunization period and moved to BSL-3 facility before challenge infection. Growing the SARS-CoV-2 SARS-CoV-2 was obtained from a Covid-19 confirmed patient in Washington State (BEI Resources). To infect ferrets, virus was propagated on Vero E6 cells in Dulbelco’s modified eagle medium (DMEM) supplemented with 10% Fetal Bovine Serum (FBS) and 1% penicillin/streptomycin at 37°C for 72 h. Propagated viruses were stored at -80°C freezer for future use. Virus was titrated using Vero E6 cells by measuring the cytopathic effect after 5 days of infection in 96-well plate and titers were expressed as tissue culture infection dose assay (TCID50) using the standard method (59). Vaccination and challenge After animals were shipped (n =36) they were allowed for 7 days of initial acclimation. Sampling and vaccination were performed under isoflurane anesthesia. Ferrets (6 per group) were vaccinated intranasally with either mock saline, empty liposomes, N and S-RBD/MSU adjuvant or liposomes entrapped N and S-RBD/MSU adjuvant. Animals were given a vaccine booster in the same manner three weeks later. The amount of N and S-RBD soluble protein per dose was 60 μg each and in entrapped liposomes it was 40 μg each administered to ferrets. The MSU adjuvant used was 20 μg entrapped in liposomes per dose in all the three experimental groups. Ferrets were infected through intranasal route with 1 x 106 TCID50 dose per ferrets. Mock saline group of animals (n=6) remained at the BSL-1 facility. Blood, oropharyngeal swab, nasal swab, and rectal swab were collected at days post vaccination (DPV) 0 and 21, and days post challenge (DPC) 0, 2, 4, 7, 9, 11 and 14 dpi. Half of the ferrets (3 per group, total 18) were necropsied at 7 DPC, the rest at 14 DPC. Tissues and specimens collected at necropsy were bronchoalveolar lavage (BAL) fluid, blood, urine, intestine, lung, trachea, and nasal turbinates. Collected ferret secretions were resuspended in cold PBS containing 1% bovine serum albumin and antibiotics (5% penicillin/streptomycin). Tissue samples were weighed and collected in viral transport media (TCID50) or RNA later (RNA extraction). Tissue samples were homogenized, centrifuged, and the supernatant was aliquoted and stored at -80 °C for further testing. Titration of SARS-CoV-2 in samples To investigate whether collected specimens contain infectious live virus, the samples were inoculated onto confluent Vero E6 cells in 96 well plates, 100 μl/well serial diluted 10-fold beginning with 1 : 10 dilution. Plates were incubated at 37°C and 5% CO2 for 1 hour for viral adsorption and an additional 100 μl of infection media was added to each well. After 5 days of incubation the plates were read for cytopathic effect and TCID50 values were calculated using the Spearman-Karber algorithm (Ramakrishnan 2016).
Real-Time RT-PCR to detect SARS-CoV-2
For virus RNA titration, total RNA was extracted from the collected samples using the QIAmp Viral RNA extraction kit. according to the manufacturer’s instructions. RT-PCR was conducted using CDC N1 primers and probe using TAQman polymerase. Program followed was: 50 °C for 5 min, 95 °C for 20 sec, followed by 45 cycles: 95°C for 3 sec, 55°C for 30 sec. qPCR to detect the immune gene expression
A cDNA synthesis kit was used to synthesize single strand cDNA using total viral RNA. SYBR Green supermix kit (Bio-Rad, Hercules, CA), and the number of viral RNA copies was calculated and compared to the number of copies of the standard control. Primers used were either designed in house or previously published (Carolan 2014). The generation of oligonucleotide dimers for each TaqMan primer pair was assessed using Power SYBR® Green PCR MasterMix with melting curve analysis, according to the manufacturer’s instructions. Primers which resulted in oligonucleotide dimer generation were redesigned and retested. A comparison between primer pairs was also performed using Power SYBR® Green PCR MasterMix without a melting curve, according to the manufacturer’s instructions. One to two microliter cDNA sample was assayed per reaction. Each reaction consisted of 1 cycle of 50 °C for 2 min, 1 cycle of 95 °C for 2 min, followed by 40 cycles of 95 °C for 15 s and 60 °C for 1 min. Real time PCR runs for each gene included cDNA standards (10-fold and 2-fold dilutions, in triplicate).
Calculations of reaction efficiency and fold change of gene expression
The efficiency of each gene amplification was calculated by plotting the average Ct (y-axis) against the logarithm of the input amount of RNA/μl cDNA (x- axis). A 10-fold dilution series was used for each gene. Real time PCR efficiency (E) = (10-1/slope) for 10-fold dilution series (Pfaffl 2004) - % real time PCR efficiency = (E - 1) × 100, if the standard deviation for the efficiencies determined using 10-fold dilution. The geometric mean of the efficiencies for the indicated genes was used for the housekeeping gene efficiency. The fold change of expression of a gene was calculated using Double delta CT calculations using the housekeeping gene GAPDH for normalization (Livak 2001).
Statistical Analysis
The statistical significance of infected and contact samples compared with naive sample was assessed by one-way ANOVA with Tukey’s multiple comparisons test. Data plotting, interpolation and statistical analysis were performed using GraphPad Prism 9.2. Statistical details of experiments are described in the figure legends. A p value less than 0.05 is considered statistically significant.
Results
Intranasal delivered SARS-CoV-2 subunit vaccine reduces the nasal viral load:
In SARS-CoV-2 subunit vaccine administered ferrets subjected to challenge with the SARS-CoV-2 observed complete absence of any detectable replicating virus from day 8 in nostrils compared to control groups (Fig. 1A). In oro-pharynx also substantially reduced infectious virus load was noticed (Fig. 1B). In nasal turbinate and lungs at DPC 7 the replicating virus was undetectable, while only protein and adjuvant vaccine group still had high virus load (Fig. 1C&D). This data suggests that these vaccines have the ability to reduce and clear the SARS-CoV-2 load rapidly from the entire respiratory tract of intranasal vaccinates.
Similarly, the viral RNA load was also substantially reduced in nasal turbinate, trachea, bronchoalveolar lavage and lungs of ferrets vaccinated with SARS- CoV-2 subunit vaccine compared to only viral proteins and adjuvant formulation in the control group (Fig. 2A, B, C, & D).
Immune responses in intranasal SARS-CoV-2 subunit vaccine received animals:
Enhanced immunological responses in terms of expression of important immune cytokines genes in lungs and nasal turbinates was noticed supporting the reduced viral load in the respiratory tract of ferrets received SARS-CoV-2 subunit vaccine (Fig. 3). This include IFNα and MCP-1 in lungs and nasal turbinate (Fig. 3A, B, C, D & E) and IL-17A in lungs (Fig. 3F). Example 2: SARS-CoV-2 Spike receptor-binding domain entrapped chitosan nanoparticle intranasal vaccine elicits local and systemic Th1 and Th2 response in mice and antiviral immunity in Syrian hamsters Summary Given the ongoing COVID-19 pandemic and the need to build sustainable herd immunity in the population, the search for novel and safe vaccines is ongoing. Disclosed herein is a novel intranasal subunit vaccine platform called NARUVAX- C19/Nano based on the SARS-CoV-2 spike protein receptor-binding domain (RBD) entrapped in mannose-conjugated chitosan nanoparticles (NP). To enhance the T-cell immune response of the NP-vaccine formulation, the adjuvant CpG55.2, a toll-like receptor 9 antagonist, was included in the formulation. Both the resulting vaccines were assessed for immunogenicity and protective efficacy, as well as protection against virus transmission; soluble RBD mixed with alum adjuvant administered intramuscular was included as a control. In BALB/c mice administered with both the NP vaccines by intranasal route twice detected induced secretory IgA antibodies and a pronounced Th1-cell response, which was absent in intramuscular alum-adjuvanted RBD control vaccine group. In Syrian hamsters delivered with similar NP formulations provided protection against a wild-type SARS-CoV-2 (D614G) challenge infection, indicated by significantly rescued weight loss, reduced lung viral load and lung pathology. However, despite significantly reduced virus titers in nasal turbinates and oropharyngeal swabs in NP vaccinates the virus transmission to naïve sentinel animals was present. In conclusion, intranasal delivered RBD-based NP vaccine formulations induced mucosal immune responses in mice and protected against SARS-CoV-2 infection in Syrian hamsters. These findings are encouraging and supporting the further development of NP-based intranasal vaccine platform to mitigate SARS-CoV- 2 infection. Disclosed herein is a new subunit SARS-CoV-2 vaccine delivery platform for intranasal administration. This vaccine contain Spike protein receptor-binding domain (RBD) entrapped in mannose-conjugated chitosan nanoparticle called NARUVAX-C19/Nano, which has the potential to induce not only systemic but also local mucosal immunity. Anti-RBD antibodies block the interaction of the virus with the angiotensin-converting enzyme 2 (ACE2) cell receptor, and thus neutralize the virus and prevent infection (Buchholz 2004; Jiang 2020). Intranasal delivery of vaccine induces mucosal immunity including production of secretory IgA antibodies which acts as the first line of defense against respiratory pathogens, including SARS- CoV-2 virus (Russell 2020). In addition, intranasal vaccination is a needle-free noninvasive method which eliminates several issues (local pain and discomfort at injection site, increased cost of vaccine, need of trained person for vaccination, and fear of injection (Zheng 2018). Intranasal immunization, unlike other mucosal routes of administration, requires lower doses and does not expose antigens to extreme pH and has a larger absorption area (Riese 2014). Nanoparticle (NP)-based protein subunit delivery helps protect the vaccine antigens from premature degradation, increases its stability, and ensures targeted delivery of immunogen to antigen- presenting cells (APC) (Pati 2018; Means 2003). The NP-forming a natural carbohydrate polymer chitosan is biocompatible and bioavailable, and its positively charged amino groups electrostatically interact with negatively charged sialic acid mucus and epithelial surfaces, becoming a mucoadhesive vaccine vehicle (Renu 2020). Surface labeling NP with mannose, a calcium-dependent (type C) mannose receptor of the lectin family binds readily to dendritic cells and macrophages [18], providing a pronounced adjuvant effect to the vaccine (Dhakal 2018; Han 2020). To enhance the T-cell immune response induced by the NP-formulation, the adjuvant CpG55.2 (Toll-like receptor 9 antagonist (Bode 2011); Vaxine Pty Ltd, Adelaide, Australia) was additionally included in its formulation. Therefore we developed a novel candidate chitosan NP based S-RBD plus CpG containing vaccine formulation and evaluated for immunogenicity and protective efficacy in appropriate preclinical animal models. Further, elucidated the ability of vaccine for mitigating the SARS- CoV-2 transmission and compared its performance with an injectable RBD plus aluminum hydroxide (Alum) adjuvant vaccine. Materials and methods Virus, Biosafety and Bioethics The virus strain hCoV-19/Kazakhstan/KazNAU-NSCEDI481/2020 of wildtype SARS-CoV-2 with D614G mutation in spike protein was used. This virus was isolated at the Aikimbayev National Research Center for Especially Dangerous Infections (NSCEDI) in June 2020 from nasopharyngeal swab of a 45-year-old COVID-19 patient in Almaty, Kazakhstan (GISAID, #EPI_ISL_514093). The virus was grown as described previously (Tabynov 2022). In this study, used the 3rd passage virus which had an infectious titer of 6.2 log10 TCID50/mL. Vaccine preparation Spike Protein RBD [Gln321-Ser591] purchased from a commercial source (ABP Biosciences, USA) was expressed in HEK293 cells, purity > 95% as determined by SDS-PAGE; Endotoxin - < 1.0 EU per μg protein as determined by the LAL method. The SARS-CoV-2 vaccine formulation was prepared using mannose- conjugated chitosan nanoparticles (NP-vaccine) by a standard ionic gelation method as described previously (Tabynov 2022). The particle size, the antigen loading efficiency, and size distribution and morphology analyses of NP-vaccine was determined by scanning electron microscopy. The vaccine formulation was lyophilized and stored at -20°C until use. Resuspension of the NP-vaccine was carried out with PBS to the desired volume. The CpG (Vaxine Pty Ltd, Australia) was added while entrapping S-RBD to obtain an adjuvanted NP-vaccine formulation (NP-CpG vaccine). Injectable vaccine used for comparative analysis was prepared by mixing S- RBD with aluminum hydroxide adjuvant (Alhydrogel® adjuvant 2%, InvivoGen, CA, USA; Alum vaccine). Both the vaccine formulations was sterile and contained less than 2 EU bacterial endotoxins per dose. The details of the vaccines content and their administration are provided in Table 1. Table 1. Vaccine formulations and routes of administration
Figure imgf000042_0001
Figure imgf000043_0001
Particle Size Determination
The Particle size distribution and mean diameter of NP-formulations were assessed in aqueous dispersions with proper dilution by using the dynamic light scattering (DLS) technique or photon correlation spectroscopy (DLS Zetasizer Nano ZSP; Model-ZEN5600; Malvern Instruments Ltd., Worcestershire, UK) in disposable polystyrene cuvettes (Model DTS0012; Malvern) at 25 °C. All the readings were taken in triplicate at different time intervals and for independent experiments with He - Ne laser 63.3 nrn and with avalanche photodiode detector (APD). The average of 3 readings (each reading = 30 runs) was reported as the actual particle size.
Scanning electron microscopy
The morphology of the nanoparticles was determined by using scanning electron microscopy. NP suspension (5 μL) was placed on the cleaned silicon wafer chip (SPI Supplies, USA) (Cat no. 4136SC-AB) and then on aluminum stubs, air dried in fume hood for 60 min and kept overnight under vacuum. Samples were coated with platinum for up to 30 nm thickness in the Q150T plus sputter coater (Quorum Technologies, UK) and imaging was done on the Hitachi SU5000 Field emission scanning electron microscope.
Entrapment efficiency
The protein entrapment efficiency in mannose conjugated chitosan NP was estimated by an indirect method by determining difference between protein amount found in the vaccine formulation supernatant and initial amount used. The amount of protein present in the supernatant was measured using the micro-BCA protein assay kit (Biorad, USA). Entrapment efficiency (%) = [(RBD protein added - Free "unentrapped RBD protein")/RBD protein added] *100.
Vaccination and Immune Response Analysis in Mice Four to-six-week-old SPF (specific pathogen-free) female BALB/c mice obtained from the NSCEDI Laboratory Animal Breeding Facility were used. Animals were placed in ventilated cages with HEPA filters (Allentown, USA) for 7 days prior to the experiment for acclimatization. Mice were immunized with NP-vaccine formulations, antigen alone, and PBS IN under ketamine (100 mg/kg) and xylazine (10 mg/kg) anesthesia in a. volume of 100 μl twice at 21 -day intervals. The Alum vaccine was administered in a similar manner but using an intramuscular route of administration. At 21 days post prime (n=7/group) and booster (n=7/group) vaccination, blood samples were taken from the orbital venous sinus to determine antigen-specific IgA, IgG antibodies and its isotypes (IgG1 and IgG2a), RBD-ACE2 blocking and virus neutralizing antibody titers against a wild-type SARS-CoV-2 (D614G).
At day 21 after booster (n=3/group) vaccination, mice were euthanized by cervical dislocation under ketamine-xylazine anesthesia, and their spleens were collected under aseptic conditions to evaluate the cellular immune response and lungs for determining the antigen-specific IgA antibodies.
Determination of humoral and cellular immune responses
The levels of anti-RBD specific secretory IgA, IgG, IgG1, and IgG2a antibodies were determined by enzyme linked immunoassay (ELISA) as previously described [22], SARS-CoV-2 Surrogate Virus Neutralization Test (sVNT) Kit (L00847; GenScript, Piscataway, USA) was used to detect antibodies that inhibit RBD binding to the ACE2 cell receptor according to the manufacturer's instructions. The inhibition percentage of the sample was calculated as (1-Average OD of the sample/ Average OD of the negative control) × 100%. A sample with an inhibition percentage <30% was considered "negative" and ≥ 30% was considered "positive" for SARS-CoV-2 neutralizing antibodies. The following neutralizing antibody values were determined according to the level of inhibition : low (30-59%), medium (60-89), high (90≤). Determination of viral neutralizing antibodies was performed as previously described [22] using 1000 TCID50 of wild-type SARS-CoV-2 (D614G). The neutralizing antibody titer was the highest dilution of serum that inhibited the cytopathic effect in 100% of wells. Cellular immunity was determined by cytokine production of IL-2 (#B320273), IFN-γ (#B307222), IL-4 (#B320413), IL-10 (#B311304), IL-5 (#B317463), IL-6 (#B321215), IL-17A (#B303513), TNF-α (#B306271), using ELISA MAX™ Deluxe Set Mouse kits (BioLegend), in a suspension of splenocytes in response to restimulation with 5 μg RBD protein as previously described [22]. Cytokine production data were presented as the difference (Delta) of cytokine concentrations (pg/mL) between samples with and without RBD protein stimulation. Hamster vaccination and protective efficacy evaluation of vaccines Male Syrian hamsters 6-8 week-old obtained from the NSCEDI Laboratory Animal Breeding Facility were used. Animals were placed in ventilated cages with HEPA filters for 7 days prior to the experiment for acclimatization. Hamsters were immunized with NP-vaccine formulations (NP vaccine and NP-CpG vaccine, n=6/group) containing RBD protein 5 pg/dose or PBS (control group, n=6) IN in a 100 μl volume under ketamine (80 mg/kg) and xylazine (8 mg/kg) anesthesia twice at 21-day intervals. For comparison, an Alum-based vaccine containing 5 μg/100 μl of RBD protein was administered IM. At day 21 after booster vaccination, hamsters of all groups were IN challenged with SARS-CoV-2 at a dose of 1 x 104 TCID50 under ketamine-xylazine anesthesia and observed for 7 days post infection with daily measurement of body weight. On days 3 and 7 after challenge, half of the animals (3/6) from each group were euthanized and collected nasal turbinates and lung samples. Three lobes of the right lung from each animal were fixed in 10% formaldehyde for histopathological studies. Two lobes of the left lung were homogenized in 1 mL DMEM using a TissueLyser II instrument (QIAGEN) at 300 vibrations/min for 60 seconds, the supernatant after centrifugation (5000 g for 15 min at 4°C) was collected and stored at -70°C for determination of viral titer. Virus Transmission Assessment In all groups of hamsters, on the 2nd day of the challenge, two naïve animals were comingled for one day and then separated into clean cages, where they were kept in isolation for additional 4 days. On day 5 after co-housing, the sentinel animals were euthanized to assess viral load in the nasal turbinates and lungs, as well as pathological changes in the lungs through histological analysis. The weight of the comingled animals for 5 days was also recorded. Analysis of viral titers and histological evaluation Virus titers in the tissue homogenates were determined as previously described (Tabynov 2022). The virus titer was calculated by using the Reed and Mench method and expressed in log10 TCID50/0.2 mL. Histological analysis of hamster lungs was performed as previously described (Tabynov 2022). Each slide was quantified based on the severity of histologic changes, including interstitial pneumonia, alveolitis, bronchiolitis, alveolar destruction, interstitial infiltration, pulmonary hemorrhage, and peribronchiolar inflammation. Based on the previously described method (Tabynov 2022; Lu 2021) the assessment score was as follows: 4 points - extremely severe pathological lung changes; 3 points - severe pathological lung changes; 2 points - moderate pathological lung changes; 1 point - mild pathological lung changes; 0 points - no pathological changes. Statistical analysis The GraphPad Prism 9.0.0 (San Diego, USA) was used for graphing and statistical analysis of experimental data. Differences in antibody levels, cytokine production, viral load in respiratory organs and oropharyngeal swabs, weight dynamics, and pathological changes in the lungs between animal groups were assessed using Tukey's multiple comparisons test. The limit of viral titer detection was 0.7 log10 TCID50/0.2 mL. The limit of detection for neutralizing antibodies was 3.0 log2. Geometric mean titers with 95% confidence interval were calculated for neutralizing antibody analysis. For all comparisons, P<0.05 was considered significant. All error bars in the graphs represent the standard error mean. Results Nanoparticle Characteristics The average size of mannose-conjugated chitosan particles and RBD protein entrapped mannose-conjugated chitosan NPs was 180±12 nm and 290±18 nm, respectively. Scanning electron microscopy showed that the particles are discrete and spherical in shape (Figure 4). The entrapment efficiency of RBD protein in mannose- conjugated chitosan NPs was 66.78%. Intranasal NP-vaccine induced antigen-specific IgA antibodies in mice NP-vaccine formulations in mice after both prime and booster IN immunization induced antigen-specific IgA antibodies in serum (Figure 4A) and lungs (Figure 5B). The NP-vaccine formulation without CpG elicited IgA antibody at levels significantly higher than mock and antigen only control groups. Notably, NP-vaccine formulations with all tested doses in immunized mice did not elicit detectable levels of IgG, IgG1, IgG2a, and RBD-ACE2 blocking and virus-neutralizing antibodies. In contrast, Alum based vaccine induced significant levels of IgG, IgG1, IgG2a (Th2 polarization, Figure 5C) as well as RBD-ACE2 blocking (predominantly at medium and high levels, Figure 5D) and viral neutralizing antibody titers (GMT 15.7, Figure 5E) after booster immunization compared to control group. However, IgA antibody formation was not observed in this group of mice. Intranasal NP-vaccine formulations in mice elicits a pronounced Th1- polarized cellular immune response Evaluation of eight types of cytokines production in response to restimulation of mouse splenocytes with RBD protein showed that NP-vaccine formulations at most of the tested doses induced significant production of Th1 cytokines IFN-γ, IL-2, TNF- α, and Th17-type IL-17A compared to controls, and in some cases (for IFN-γ, IL- 17A) compared to antigen alone group (Figure 6). Notably, only the NP-vaccines adjuvanted with the CpG provided significant production of IL-17A compered to control and corresponding groups of NP-vaccine formulations groups without CpG. In contrast, the Alum adjuvanted vaccine distinguished itself by the production of Th2 cytokines IL-4 and IL-6, whose levels were significant compared to controls, antigen alone and NP-vaccine groups. Intranasal NP-vaccines provide protection against wild-type SARS-CoV-2 (D614G) infection in hamsters, but did not block the viral transmission Protective efficacy was evaluated clinically by recording changes in body weight gain for 7 days post challenge infection (Figure 7A). All hamsters, both vaccinated and unvaccinated, had a steadily decreasing body weight up to 6 days post challenge. However, NP-vaccine immunized animals had significantly lower weight loss over the entire observation period compared to the control group. Weight loss at peak was 9 - 10.5% in vaccinated animals and 15% in control animals. In the alum adjuvanted IM vaccine group the body weight loss was slightly higher than in the NP- vaccine groups, and it was significantly better compared to control group at 2-5 days after challenge. Virus load was determined by measuring the infectious viral titer in oropharyngeal swabs of hamsters at day 2 post challenge (Figure 7C), as well as in nasal turbinates and lungs of euthanized animals at day 3 (Figure 7D) and day 7 (Figure 7E) after challenge infection. The virus load in the oropharyngeal swabs of vaccinated hamsters was lower than in the control group, and this difference was significant for the NP-vaccine group. Virus was detected in the respiratory organs of all hamsters, with the highest titers on day 3 after challenge. However, the viral load in NP-vaccine immunized animals in nasal turbinates and lungs was significantly lower in comparison not only to the control group, but also to the alum-adjuvanted vaccine group. During the period of observation, the overall virus titers in nasal turbinates was much higher than in the lungs. In the lungs of all infected hamsters, including vaccinated ones, the presence of classical signs of acute respiratory distress syndrome (ARDS) manifested by SARS-CoV-2 infection was confirmed. On day 3, the lungs of hamsters had signs of the exudative phase of ARDS, which by day 7 changed to the fibroproliferative phase of ARDS (Figure 7G). Comparative morphological characterization of lungs in terms of lesions was significantly lower in both NP-vaccine groups compared to control and alum-adjuvanted vaccine groups (Figure 7B). Interestingly, in the alum vaccine group, the amount of lung damage was as high as in the control group. None of the vaccinated animals blocked the virus transmission to naïve sentinels. Sentinel animals of all groups showed negative weight dynamics (3.9-4.8% at 5 days post co-housing, data not shown) and the presence of virus (Figure 7F) in the nasal turbinates (in 2/2 of the group) and lungs (in 1/2 of the group). All the sentinels had similar level of lung damage as per histological analysis (Figure 7B). Discussion In this study, intranasal administered NP-vaccine formulations elicited a different immunogenicity profile from that of IM alum-adjuvanted RBD vaccine. Both of the NP-vaccine formulations in rodent models did not induce measurable serum neutralizing antibodies, while induced mainly secretory IgA and cell mediated immune responses. This feature of this NP vaccine is a cardinal difference from other intranasal subunit RBD- or Spike-based vaccines conjugated with Diphtheria toxoid (EcoCRM®) (Wong 2022) or outer membrane vesicles (OMVs) from Neisseria meningitidis (van der Ley 2021), respectively, which were shown to induce both systemic neutralizing antibodies and local IgA responses. However, despite the absence of serum neutralizing antibodies, a crucial component of anti-SARS-CoV-2 infection immunity (Buchholz 2004; Jiang 2020) this NP-vaccine formulations in vaccinated hamsters provided significant protection against SARS-CoV-2 infection. The NP-vaccine significantly reduced the challenge virus load in both the upper and lower respiratory tract, and, most importantly, reduced the degree of lung damage in hamsters after challenge to a greater extent than the IM vaccine group. It appears likely that protection with NP-vaccine formulations was due to both anti-RBD IgA plus the Th1 cellular immune responses that is consistent with findings of others (Weiskopf 2020; Moss 2022; Aleebrahim-Dehkordi 2022). Although the addition of CpG to the NP-vaccine formulation induced production of IL-17, a Th17 cytokine involved in protective immunity against many pathogens (Mills 2022), this did not translate to increased protective efficacy of the intranasal NP vaccine. The IM administered alum-adjuvanted RBD-based vaccine, despite inducing a systemic humoral and Th2 cellular immune responses, failed to provide protection against SARS-CoV-2 infection. This is attributed to the use of monomeric RBD protein, which when compared to the full-length Spike trimer induces significantly lower titers of neutralizing antibodies (Tabynov 2022). In general, the use of RBD protein in COVID-19 vaccine requires large doses of antigen and multiple immunizations to achieve protection (Yang 2021). Alum is a highly Th2 biased adjuvant and in an earlier study (Tabynov 2022) where a veterinary COVID-19 vaccine called NARUVAX-C19 (pets) was studied in juvenile cats, the alum adjuvant did not enhance neutralizing antibody titers compared to antigen alone. Another important part of the present research was evaluation of the vaccines in protecting against virus transmission from vaccinated challenged animals to naïve sentinels. Despite significantly lower titers of SARS-CoV-2 than control group in NP- vaccinates in nasal turbinates and oropharyngeal swabs of hamsters which was not been seen with other intranasal vaccine candidates (Wong 2022; van der Ley 2021) the infected NP vaccinates still transmitted infection to sentinel animals. This finding is consistent with others showing hamsters or humanized mice immunized with viral vector (Bricker 2021; Tostanoski 2020) or genetic vaccines (Kalnin 2021) continued to transmit infectious challenge virus to sentinels. Previously, an IM subunit spike- trimer based squalene emulsion-adjuvanted NARUVAX-C19 vaccine protected against SARS-CoV-2 virus transmission in a hamster model, showing that transmission blocking is achievable with some vaccines (Tabynov 2022). Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of skill in the art to which the disclosed invention belongs. Publications cited herein and the materials for which they are cited are specifically incorporated by reference. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
REFERENCES 1. Prompetchara E, Ketloy C, Palaga T. 2020. Immune responses in COVID-19 and potential vaccines: Lessons learned from SARS and MERS epidemic. Asian Pac J Allergy Immunol 38:1-9. 2. Huang AT, Garcia-Carreras B, Hitchings MDT, Yang B, Katzelnick L, Rattigan SM, Borgert B, Moreno C, Solomon BD, Rodriguez-Barraquer I, Lessler J, Salje H, Burke DS, Wesolowski A, Cummings DAT. 2020. A systematic review of antibody mediated immunity to coronaviruses: antibody kinetics, correlates of protection, and association of antibody responses with severity of disease, Infectious Diseases (except HIV/AIDS). 3. Melgaco JG, Azamor T, Ano Bom APD. 2020. Protective immunity after COVID-19 has been questioned: What can we do without SARS-CoV-2-IgG detection? Cell Immunol 353:104114. 4. Wu F, Wang A, Liu M, Wang Q, Chen J, Xia S, Ling Y, Zhang Y, Xun J, Lu L, Jiang S, Lu H, Wen Y, Huang J.2020. Neutralizing antibody responses to SARS-CoV-2 in a COVID-19 recovered patient cohort and their implications, Infectious Diseases (except HIV/AIDS). 5. Thevarajan I, Nguyen TH, Koutsakos M, Druce J, Caly L, van de Sandt CE, Jia X, Nicholson S, Catton M, Cowie B, Tong S, Lewin S, Kedzierska K. 2020. Breadth of concomitant immune responses underpinning viral clearance and patient recovery in a non-severe case of COVID-19, Infectious Diseases (except HIV/AIDS). 6. Weiskopf D, Schmitz KS, Raadsen MP, Grifoni A, Okba NMA, Endeman H, van den Akker JPC, Molenkamp R, Koopmans MPG, van Gorp ECM, Haagmans BL, de Swart RL, Sette A, de Vries RD. 2020. Phenotype of SARS-CoV- 2-specific T-cells in COVID-19 patients with acute respiratory distress syndrome, Infectious Diseases (except HIV/AIDS). doi: 7. Bauer T, Jilg W. 2006. Hepatitis B surface antigen-specific T and B cell memory in individuals who had lost protective antibodies after hepatitis B vaccination. Vaccine 24:572-7. 8. Neutra MR, Kozlowski PA.2006. Mucosal vaccines: the promise and the challenge. Nat Rev Immunol 6:148-58. 9. Manocha M, Pal PC, Chitralekha KT, Thomas BE, Tripathi V, Gupta SD, Paranjape R, Kulkarni S, Rao DN. 2005. Enhanced mucosal and systemic immune response with intranasal immunization of mice with HIV peptides entrapped in PLG microparticles in combination with Ulex Europaeus-I lectin as M cell target. Vaccine 23:5599-617. 10. Lisa Schnirring CN, Feb 21, 2018. 2018. CDC vaccine panel brings back FluMist for 2018-19 season. 11. Pachetti M, Marini B, Benedetti F, Giudici F, Mauro E, Storici P, Masciovecchio C, Angeletti S, Ciccozzi M, Gallo RC, Zella D, Ippodrino R. 2020. Emerging SARS-CoV-2 mutation hot spots include a novel RNA-dependent-RNA polymerase variant. J Transl Med 18:179. 12. Harvey WT, Carabelli AM, Jackson B, Gupta RK, Thomson EC, Harrison EM, Ludden C, Reeve R, Rambaut A, Consortium C-GU, Peacock SJ, Robertson DL. 2021. SARS-CoV-2 variants, spike mutations and immune escape. Nat Rev Microbiol 19:409-424. 13. Cox RJ, Brokstad KA, Ogra P. 2004. Influenza virus: immunity and vaccination strategies. Comparison of the immune response to inactivated and live, attenuated influenza vaccines. Scand J Immunol 59:1-15. 14. Mazanec MB, Coudret CL, Fletcher DR. 1995. Intracellular neutralization of influenza virus by immunoglobulin A anti-hemagglutinin monoclonal antibodies. J Virol 69:1339-43. 15. Suzuki T, Kawaguchi A, Ainai A, Tamura S, Ito R, Multihartina P, Setiawaty V, Pangesti KN, Odagiri T, Tashiro M, Hasegawa H. 2015. Relationship of the quaternary structure of human secretory IgA to neutralization of influenza virus. Proc Natl Acad Sci U S A 112:7809-14. 16. Shim BS, Park SM, Quan JS, Jere D, Chu H, Song MK, Kim DW, Jang YS, Yang MS, Han SH, Park YH, Cho CS, Yun CH. 2010. Intranasal immunization with plasmid DNA encoding spike protein of SARS- coronavirus/polyethylenimine nanoparticles elicits antigen-specific humoral and cellular immune responses. BMC Immunol 11:65. 17. Du L, Zhao G, Lin Y, Sui H, Chan C, Ma S, He Y, Jiang S, Wu C, Yuen KY, Jin DY, Zhou Y, Zheng BJ. 2008. Intranasal vaccination of recombinant adeno-associated virus encoding receptor-binding domain of severe acute respiratory syndrome coronavirus (SARS-CoV) spike protein induces strong mucosal immune responses and provides long-term protection against SARS-CoV infection. J Immunol 180:948-56. 18. Hu MC, Jones T, Kenney RT, Barnard DL, Burt DS, Lowell GH. 2007. Intranasal Protollin-formulated recombinant SARS S-protein elicits respiratory and serum neutralizing antibodies and protection in mice. Vaccine 25:6334-40. 19. Eickhoff CS, Blazevic A, Killoran EA, Morris MS, Hoft DF. 2019. Induction of mycobacterial protective immunity by sublingual BCG vaccination. Vaccine 37:5364-5370. 20. Gallorini S, Taccone M, Bonci A, Nardelli F, Casini D, Bonificio A, Kommareddy S, Bertholet S, O'Hagan DT, Baudner BC. 2014. Sublingual immunization with a subunit influenza vaccine elicits comparable systemic immune response as intramuscular immunization, but also induces local IgA and TH17 responses. Vaccine 32:2382-8. 21. Renu S, Feliciano-Ruiz N, Patil V, Schrock J, Han Y, Ramesh A, Dhakal S, Hanson J, Krakowka S, Renukaradhya GJ. 2021. Immunity and Protective Efficacy of Mannose Conjugated Chitosan-Based Influenza Nanovaccine in Maternal Antibody Positive Pigs. Front Immunol 12:584299. 22. Moreno-Fierros L, Garcia-Silva I, Rosales-Mendoza S. 2020. Development of SARS-CoV-2 vaccines: should we focus on mucosal immunity? Expert Opin Biol Ther doi:10.1080/14712598.2020.1767062. 23. Bacon A, Makin J, Sizer PJ, Jabbal-Gill I, Hinchcliffe M, Illum L, Chatfield S, Roberts M. 2000. Carbohydrate biopolymers enhance antibody responses to mucosally delivered vaccine antigens. Infect Immun 68:5764-70. 24. Bertram U, Bernard MC, Haensler J, Maincent P, Bodmeier R. 2010. In situ gelling nasal inserts for influenza vaccine delivery. Drug Dev Ind Pharm 36:581-93. 25. Dhakal S, Cheng X, Salcido J, Renu S, Bondra K, Lakshmanappa YS, Misch C, Ghimire S, Feliciano-Ruiz N, Hogshead B, Krakowka S, Carson K, McDonough J, Lee CW, Renukaradhya GJ. 2018. Liposomal nanoparticle-based conserved peptide influenza vaccine and monosodium urate crystal adjuvant elicit protective immune response in pigs. Int J Nanomedicine 13:6699-6715. 26. Woodrow KA, Bennett KM, Lo DD. 2012. Mucosal vaccine design and delivery. Annu Rev Biomed Eng 14:17-46. 27. Heit A, Schmitz F, Haas T, Busch DH, Wagner H. 2007. Antigen co- encapsulated with adjuvants efficiently drive protective T cell immunity. Eur J Immunol 37:2063-74. 28. Schliehe C, Redaelli C, Engelhardt S, Fehlings M, Mueller M, van Rooijen N, Thiry M, Hildner K, Weller H, Groettrup M.2011. CD8- dendritic cells and macrophages cross-present poly(D,L-lactate-co-glycolate) acid microsphere- encapsulated antigen in vivo. J Immunol 187:2112-21. 29. Foged C, Brodin B, Frokjaer S, Sundblad A. 2005. Particle size and surface charge affect particle uptake by human dendritic cells in an in vitro model. Int J Pharm 298:315-22. 30. Dhakal S, Hiremath J, Bondra K, Lakshmanappa YS, Shyu DL, Ouyang K, Kang KI, Binjawadagi B, Goodman J, Tabynov K, Krakowka S, Narasimhan B, Lee CW, Renukaradhya GJ. 2017. Biodegradable nanoparticle delivery of inactivated swine influenza virus vaccine provides heterologous cell- mediated immune response in pigs. J Control Release 247:194-205. 31. Hiremath J, Kang KI, Xia M, Elaish M, Binjawadagi B, Ouyang K, Dhakal S, Arcos J, Torrelles JB, Jiang X, Lee CW, Renukaradhya GJ.2016. Entrapment of H1N1 Influenza Virus Derived Conserved Peptides in PLGA Nanoparticles Enhances T Cell Response and Vaccine Efficacy in Pigs. PLoS One 11:e0151922. 32. Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Preat V. 2012. PLGA-based nanoparticles: an overview of biomedical applications. J Control Release 161:505-22. 33. Zhang F, Peng B, Chang H, Zhang R, Lu F, Wang F, Fang F, Chen Z. 2016. Intranasal Immunization of Mice to Avoid Interference of Maternal Antibody against H5N1 Infection. PLoS One 11:e0157041. 34. Kanekiyo M, Wei CJ, Yassine HM, McTamney PM, Boyington JC, Whittle JR, Rao SS, Kong WP, Wang L, Nabel GJ. 2013. Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies. Nature 499:102-6. 35. Han Y, Renu S, Patil V, Schrock J, Feliciano-Ruiz N, Selvaraj R, Renukaradhya GJ. 2020. Immune Response to Salmonella Enteritidis Infection in Broilers Immunized Orally With Chitosan-Based Salmonella Subunit Nanoparticle Vaccine. Front Immunol 11:935. 36. Products FsRSPfGPP-BD. 2016. 37. Makadia HK, Siegel SJ.2011. Poly Lactic-co-Glycolic Acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier. Polymers (Basel) 3:1377-1397. 38. Menon JU, Ravikumar P, Pise A, Gyawali D, Hsia CC, Nguyen KT. 2014. Polymeric nanoparticles for pulmonary protein and DNA delivery. Acta Biomater 10:2643-52. 39. Dhakal S, Renu S, Ghimire S, Shaan Lakshmanappa Y, Hogshead BT, Feliciano-Ruiz N, Lu F, HogenEsch H, Krakowka S, Lee CW, Renukaradhya GJ. 2018. Mucosal Immunity and Protective Efficacy of Intranasal Inactivated Influenza Vaccine Is Improved by Chitosan Nanoparticle Delivery in Pigs. Front Immunol 9:934. 40. Khatri M, Dwivedi V, Krakowka S, Manickam C, Ali A, Wang L, Qin Z, Renukaradhya GJ, Lee CW. 2010. Swine influenza H1N1 virus induces acute inflammatory immune responses in pig lungs: a potential animal model for human H1N1 influenza virus. J Virol 84:11210-8. 41. Dhakal S, Goodman J, Bondra K, Lakshmanappa YS, Hiremath J, Shyu DL, Ouyang K, Kang KI, Krakowka S, Wannemuehler MJ, Won Lee C, Narasimhan B, Renukaradhya GJ. 2017. Polyanhydride nanovaccine against swine influenza virus in pigs. Vaccine doi:S0264-410X(17)30038-5 [pii] 10.1016/j.vaccine.2017.01.019. 42. Renu S, Feliciano-Ruiz N, Ghimire S, Han Y, Schrock J, Dhakal S, Patil V, Krakowka S, Renukaradhya GJ. 2020. Poly(I:C) augments inactivated influenza virus-chitosan nanovaccine induced cell mediated immune response in pigs vaccinated intranasally. Vet Microbiol 242:108611. 43. Dhakal S, Renukaradhya GJ. 2019. Nanoparticle-based vaccine development and evaluation against viral infections in pigs. Vet Res 50:90. 44. Dhakal S, Ghimire S, Renu S, Ross KA, Lakshmanappa YS, Hogshead BT, Bernardo P, Lee CW, Wannemuehler MJ, Narasimhan B, Renukaradhya GJ. 2019. Evaluation of CpG-ODN-adjuvanted polyanhydride-based intranasal influenza nanovaccine in pigs. Vet Microbiol 237:108401. 45. Renu S, Dhakal S, Kim E, Goodman J, Lakshmanappa YS, Wannemuehler MJ, Narasimhan B, Boyaka PN, Renukaradhya GJ. 2018. Intranasal delivery of influenza antigen by nanoparticles, but not NKT-cell adjuvant differentially induces the expression of B-cell activation factors in mice and swine. Cell Immunol 329:27-30. 46. Karaolis DK, Means TK, Yang D, Takahashi M, Yoshimura T, Muraille E, Philpott D, Schroeder JT, Hyodo M, Hayakawa Y, Talbot BG, Brouillette E, Malouin F. 2007. Bacterial c-di-GMP is an immunostimulatory molecule. J Immunol 178:2171-81. 47. Ebensen T, Schulze K, Riese P, Morr M, Guzman CA.2007. The bacterial second messenger cdiGMP exhibits promising activity as a mucosal adjuvant. Clin Vaccine Immunol 14:952-8. 48. Renu S, Feliciano-Ruiz N, Lu F, Ghimire S, Han Y, Schrock J, Dhakal S, Patil V, Krakowka S, HogenEsch H, Renukaradhya GJ. 2020. A Nanoparticle- Poly(I:C) Combination Adjuvant Enhances the Breadth of the Immune Response to Inactivated Influenza Virus Vaccine in Pigs. Vaccines (Basel) 8. 49. Zhang L, Tian X, Zhou F. 2007. Intranasal administration of CpG oligonucleotides induces mucosal and systemic Type 1 immune responses and adjuvant activity to porcine reproductive and respiratory syndrome killed virus vaccine in piglets in vivo. Int Immunopharmacol 7:1732-40. 50. Braga TT, Forni MF, Correa-Costa M, Ramos RN, Barbuto JA, Branco P, Castoldi A, Hiyane MI, Davanso MR, Latz E, Franklin BS, Kowaltowski AJ, Camara NO. 2017. Soluble Uric Acid Activates the NLRP3 Inflammasome. Sci Rep 7:39884. 51. Ng G, Chau EM, Shi Y. 2010. Recent developments in immune activation by uric acid crystals. Arch Immunol Ther Exp (Warsz) 58:273-7. 52. Sakamaki I, Inai K, Tsutani H. 2011. Safety of intradermal injection of monosodium urate crystals as a vaccine carrier in volunteers. Nucleosides Nucleotides Nucleic Acids 30:1077-84. 53. Cheng XG, Zhong GM, Mcdonough J, MacNaughton M. 2014. In vivo evaluation of MSU crystals as an adjuvant, abstr Vaccine Development Center of San Antonio Conference, Nov 13-14, 2014, 54. Studier FW.2005. Protein production by auto-induction in high density shaking cultures. Protein Expr Purif 41:207-34. 55. Cheng X, Tsao C, M. Saul J, Sylvia V, Cornet D, Christy R. 2013. Comparison of Two Nanoparticle Formulations for Localized Delivery of Platelet- Derived Growth Factor (PDGF) from Aligned Collagen Fibers. Pharmaceutical Nanotechnology 1:105-114. 56. Cheng X, Carson K, Mcdonough J, Gourapura RG, Lee CW, Dhakal S. 2017. A liposomal subunit flu vaccine formulation. US Patent application (pending), SwRI invention disclosure docket # 3928 57. Fry DW, White JC, Goldman ID. 1978. Rapid separation of low molecular weight solutes from liposomes without dilution. Anal Biochem 90:809-15. 58. Cheng X, Haggins DG, York RH, Yeni YN, Akkus O. 2009. Analysis of crystals leading to joint arthropathies by Raman spectroscopy: comparison with compensated polarized imaging. Appl Spectrosc 63:381-6. 59. Ramakrishnan MA. 2016. Determination of 50% endpoint titer using a simple formula. World J Virol 5:85-6. 60. Carolan LA, Butler J, Rockman S, Guarnaccia T, Hurt AC, Reading P, Kelso A, Barr I, Laurie KL. 2014. TaqMan real time RT-PCR assays for detecting ferret innate and adaptive immune responses. J Virol Methods 205:38-52. 61. Pfaffl MW, Tichopad A, Prgomet C, Neuvians TP. 2004. Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper--Excel-based tool using pair-wise correlations. Biotechnol Lett 26:509-15. 62. Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25:402-8. 63. Lan, J., Ge, J., Yu, J. et al. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 581, 215–220 (2020). 64. World Health Organization. COVID-19 vaccine tracker and landscape. [accessed 2022 Aug 12]. 65. COVID-19 Vaccines with WHO Emergency Use Listing. 66. World Health Organization. Coronavirus disease (COVID-19) pandemic. 2021. [accessed 2022 Aug 12]. 67. Planas D, Veyer D, Baidaliuk A, Staropoli I, Guivel-Benhassine F, Rajah MM, Planchais C, Porrot F, Robillard N, Puech J, Prot M, Gallais F, Gantner P, Velay A, Le Guen J, Kassis-Chikhani N, Edriss D, Belec L, Seve A, Courtellemont L, Péré H, Hocqueloux L, Fafi-Kremer S, Prazuck T, Mouquet H, Bruel T, Simon- Lorière E, Rey FA, Schwartz O. Reduced sensitivity of SARS-CoV-2 variant Delta to antibody neutralization. Nature. 2021 Jul 8. doi: 10.1038/s41586-021-03777-9. 68. Cao, Y., Wang, J., Jian, F. et al. Omicron escapes the majority of existing SARS-CoV-2 neutralizing antibodies. Nature 602, 657–663 (2022). https://doi.org/10.1038/s41586-021-04385-3. 69. Grewal R, Kitchen S A, Nguyen L, Buchan S A, Wilson S E, Costa A P et al. Effectiveness of a fourth dose of covid-19 mRNA vaccine against the omicron variant among long term care residents in Ontario, Canada: test negative design study BMJ 2022; 378 :e071502 doi:10.1136/bmj-2022-071502 70. Kuehn BM. mRNA Booster Improves a COVID-19 Vaccine’s Effectiveness. JAMA. 2022;327(18):1749. doi:10.1001/jama.2022.6891 71. Rashedi R, Samieefar N, Masoumi N, Mohseni S, Rezaei N. COVID- 19 vaccines mix-and-match: The concept, the efficacy and the doubts. J Med Virol. 2022 Apr;94(4):1294-1299. doi: 10.1002/jmv.27463. 72. Beatty AL, Peyser ND, Butcher XE, et al. Analysis of COVID-19 Vaccine Type and Adverse Effects Following Vaccination. JAMA Netw Open. 2021;4(12):e2140364. doi:10.1001/jamanetworkopen.2021.40364 73. Buchholz UJ, Bukreyev A, Yang L, Lamirande EW, Murphy BR, Subbarao K, Collins PL. Contributions of the structural proteins of severe acute respiratory syndrome coronavirus to protective immunity. Proc Natl Acad Sci U S A. 2004;101(26):9804-9. 74. Jiang S, Hillyer C, Du L. Neutralizing Antibodies against SARS-CoV- 2 and Other Human Coronaviruses. Trends Immunol. 2020;41(5):355-359. 75. Russell MW, Moldoveanu Z, Ogra PL and Mestecky J (2020) Mucosal Immunity in COVID-19: A Neglected but Critical Aspect of SARS-CoV-2 Infection. Front. Immunol. 11:611337. doi: 10.3389/fimmu.2020.611337 76. Zheng, Z., Diaz-Arévalo, D., Guan, H., & Zeng, M. Noninvasive vaccination against infectious diseases. Human vaccines & immunotherapeutics, 2018; 14(7), 1717–1733. 77. Riese P, Sakthivel P, Trittel S, Guzman CA. Intranasal formulations: promising strategy to deliver vaccines. Exp Opin Drug Delivery. 2014;11:1619–34. 78. Pati, R., Shevtsov, M., & Sonawane, A. Nanoparticle Vaccines Against Infectious Diseases. Frontiers in immunology, 2018;9, 2224. 79. Means TK, Hayashi F, Smith KD, Aderem A, Luster AD. The Toll- like receptor 5 stimulus bacterial flagellin induces maturation and chemokine production in human dendritic cells. J Immunol. 2003; 170:5165–75. 80. Renu, S., Markazi, A.D., Dhakal, S., Lakshmanappa, Y.S., Shanmugasundaram, R., Selvaraj, R.K., and Renukaradhya, G.J. Oral Deliverable Mucoadhesive Chitosan Salmonella Subunit Nanovaccine for Layer Chickens. Int J Nanomedicine 2020; 15, 761-777. 81. Apostolopoulos, V., Thalhammer, T., Tzakos, A.G., and Stojanovska, L. Targeting antigens to dendritic cell receptors for vaccine development. Journal of drug delivery 2013; 2013:869718. 82. Dhakal, S., Renu, S., Ghimire, S., Shaan Lakshmanappa, Y., Hogshead, B. T., Feliciano-Ruiz, N., Lu, F., HogenEsch, H., Krakowka, S., Lee, C. W., & Renukaradhya, G. J. Mucosal Immunity and Protective Efficacy of Intranasal Inactivated Influenza Vaccine Is Improved by Chitosan Nanoparticle Delivery in Pigs. Frontiers in immunology 2018; 9, 934. 83. Han, Y., Renu, S., Patil, V., Schrock, J., Feliciano-Ruiz, N., Selvaraj, R., & Renukaradhya, G. J. Mannose-Modified Chitosan-Nanoparticle-Based Salmonella Subunit OralVaccine-Induced Immune Response and Efficacy in a Challenge Trial in Broilers. Vaccines, 2020; 8(2), 299. 84. Bode C, Zhao G, Steinhagen F, Kinjo T, Klinman DM. CpG DNA as a vaccine adjuvant. Expert Rev Vaccines. 2011;10(4):499-511. 85. Tabynov K, Turebekov N, Babayeva M, Fomin G, Yerubayev T, Yespolov T, Li L, Renukaradhya GJ, Petrovsky N, Tabynov K. An adjuvanted subunit SARS-CoV-2 spike protein vaccine provides protection against Covid-19 infection and transmission. NPJ Vaccines. 2022 Feb 23;7(1):24. doi: 10.1038/s41541- 022-00450-8. 86. Tabynov K, Babayeva M, Nurpeisov T, Fomin G, Nurpeisov T, Saltabayeva U, Renu S, Renukaradhya GJ, Petrovsky N, Tabynov K. Evaluation of a Novel Adjuvanted Vaccine for Ultrashort Regimen Therapy of Artemisia Pollen- Induced Allergic Bronchial Asthma in a Mouse Model. Front Immunol. 2022 Mar 15;13:828690. doi: 10.3389/fimmu.2022.828690. 87. Lu M, et al. A safe and highly efficacious measles virus-based vaccine expressing SARS-CoV-2 stabilized prefusion spike. Proc. Natl Acad. Sci. USA. 2021;118:e2026153118. doi: 10.1073/pnas.2026153118. 88. Borges O, Cordeiro-da-Silva A, Tavares J, Santarém N, de Sousa A, Borchard G, Junginger HE. Immune response by nasal delivery of hepatitis B surface antigen and codelivery of a CpG ODN in alginate coated chitosan nanoparticles. Eur J Pharm Biopharm. 2008 Jun;69(2):405-16. doi: 10.1016/j.ejpb.2008.01.019. 89. Zuo Z, Zou Y, Li Q, Guo Y, Zhang T, Wu J, He C, Eko FO. Intranasal immunization with inactivated chlamydial elementary bodies formulated in VCG- chitosan nanoparticles induces robust immunity against intranasal Chlamydia psittaci challenge. Sci Rep. 2021 May 17;11(1):10389. doi: 10.1038/s41598-021-89940-8. 90. Malik A, Gupta M, Mani R, Gogoi H and Bhatnagar R (2018) Trimethyl Chitosan Nanoparticles Encapsulated Protective Antigen Protects the Mice Against Anthrax. Front. Immunol. 9:562. doi: 10.3389/fimmu.2018.00562 91. Li L, Honda-Okubo Y, Huang Y, Jang H, Carlock MA, Baldwin J, Piplani S, Bebin-Blackwell AG, Forgacs D, Sakamoto K, Stella A, Turville S, Chataway T, Colella A, Triccas J, Ross TM, Petrovsky N. Immunisation of ferrets and mice with recombinant SARS-CoV-2 spike protein formulated with Advax-SM adjuvant protects against COVID-19 infection. Vaccine. 2021 Sep 24;39(40):5940- 5953. doi: 10.1016/j.vaccine.2021.07.087. 92. Li L, Honda-Okubo Y, Baldwin J, Bowen R, Bielefeldt-Ohmann H, Petrovsky N. Covax-19/Spikogen® vaccine based on recombinant spike protein extracellular domain with Advax-CpG55.2 adjuvant provides single dose protection against SARS-CoV-2 infection in hamsters. Vaccine. 2022 May 20;40(23):3182- 3192. doi: 10.1016/j.vaccine.2022.04.041. 93. Tabarsi P, Anjidani N, Shahpari R, Mardani M, Sabzvari A, Yazdani B, Roshanzamir K, Bayatani B, Taheri A, Petrovsky N, Li L, Barati S. Safety and immunogenicity of SpikoGen®, an Advax-CpG55.2-adjuvanted SARS-CoV-2 spike protein vaccine: a phase 2 randomized placebo-controlled trial in both seropositive and seronegative populations. Clin Microbiol Infect. 2022 Apr 15:S1198- 743X(22)00207-5. doi: 10.1016/j.cmi.2022.04.004. 94. Wong, T.Y., Lee, K.S., Russ, B.P. et al. Intranasal administration of BReC-CoV-2 COVID-19 vaccine protects K18-hACE2 mice against lethal SARS- CoV-2 challenge. npj Vaccines 7, 36 (2022). 95. van der Ley PA, Zariri A, van Riet E, Oosterhoff D, Kruiswijk CP. An Intranasal OMV-Based Vaccine Induces High Mucosal and Systemic Protecting Immunity Against a SARS-CoV-2 Infection. Front Immunol. 2021 Dec 17;12:781280. doi: 10.3389/fimmu.2021.781280. 96. Moss, P. The T cell immune response against SARS-CoV-2. Nat Immunol 23, 186–193 (2022). 97. Aleebrahim-Dehkordi E, Molavi B, Mokhtari M, Deravi N, Fathi M, Fazel T, Mohebalizadeh M, Koochaki P, Shobeiri P, Hasanpour-Dehkordi A. T helper type (Th1/Th2) responses to SARS-CoV-2 and influenza A (H1N1) virus: From cytokines produced to immune responses. Transpl Immunol. 2022 Feb;70:101495. doi: 10.1016/j.trim.2021.101495. 98. Mills, K.H.G. IL-17 and IL-17-producing cells in protection versus pathology. Nat Rev Immunol (2022). 99. Yang S, Li Y, Dai L, Wang J, He P, Li C, Fang X, Wang C, Zhao X, Huang E, Wu C, Zhong Z, Wang F, Duan X, Tian S, Wu L, Liu Y, Luo Y, Chen Z, Li F, Li J, Yu X, Ren H, Liu L, Meng S, Yan J, Hu Z, Gao L, Gao GF. Safety and immunogenicity of a recombinant tandem-repeat dimeric RBD-based protein subunit vaccine (ZF2001) against COVID-19 in adults: two randomised, double-blind, placebo-controlled, phase 1 and 2 trials. Lancet Infect Dis. 2021 Aug;21(8):1107- 1119. doi: 10.1016/S1473-3099(21)00127-4. 100. Tabynov K, Orynbassar M, Yelchibayeva L, Turebekov N, Yerubayev T, Matikhan N, Yespolov T, Petrovsky N, Tabynov K. A Spike Protein-Based Subunit SARS-CoV-2 Vaccine for Pets: Safety, Immunogenicity, and Protective Efficacy in Juvenile Cats. Front Vet Sci. 2022 Mar 14;9:815978. doi: 10.3389/fvets.2022.815978. 101. Bricker TL, Darling TL, Hassan AO, Harastani HH, Soung A, Jiang X, Dai YN, Zhao H, Adams LJ, Holtzman MJ, Bailey AL, Case JB, Fremont DH, Klein R, Diamond MS, Boon ACM. A single intranasal or intramuscular immunization with chimpanzee adenovirus-vectored SARS-CoV-2 vaccine protects against pneumonia in hamsters. Cell Rep. 2021 Jul 20;36(3):109400. doi: 10.1016/j.celrep.2021.109400. 102. Tostanoski LH, Wegmann F, Martinot AJ, Loos C, McMahan K, Mercado NB, Yu J, Chan CN, Bondoc S, Starke CE, Nekorchuk M, Busman-Sahay K, Piedra-Mora C, Wrijil LM, Ducat S, Custers J, Atyeo C, Fischinger S, Burke JS, Feldman J, Hauser BM, Caradonna TM, Bondzie EA, Dagotto G, Gebre MS, Jacob- Dolan C, Lin Z, Mahrokhian SH, Nampanya F, Nityanandam R, Pessaint L, Porto M, Ali V, Benetiene D, Tevi K, Andersen H, Lewis MG, Schmidt AG, Lauffenburger DA, Alter G, Estes JD, Schuitemaker H, Zahn R, Barouch DH. Ad26 vaccine protects against SARS-CoV-2 severe clinical disease in hamsters. Nat Med. 2020 Nov;26(11):1694-1700. doi: 10.1038/s41591-020-1070-6 103. Kalnin KV, Plitnik T, Kishko M, Zhang J, Zhang D, Beauvais A, Anosova NG, Tibbitts T, DiNapoli J, Ulinski G, Piepenhagen P, Cummings SM, Bangari DS, Ryan S, Huang PD, Huleatt J, Vincent D, Fries K, Karve S, Goldman R, Gopani H, Dias A, Tran K, Zacharia M, Gu X, Boeglin L, Abysalh J, Vargas J, Beaulieu A, Shah M, Jeannotte T, Gillis K, Chivukula S, Swearingen R, Landolfi V, Fu TM, DeRosa F, Casimiro D. Immunogenicity and efficacy of mRNA COVID-19 vaccine MRT5500 in preclinical animal models. NPJ Vaccines. 2021 Apr 19;6(1):61. doi: 10.1038/s41541-021-00324-5. SEQUENCES SEQ ID NO: 1 Spike Protein (S) of SARS-CoV-2 (GenBank: QIH45093.1)
Figure imgf000063_0001

Claims

WHAT IS CLAIMED IS: 1. A composition comprising SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof; and further wherein said nanoparticle comprises chitosan.
2. The composition of claim 1, wherein the antigenic fragment is a receptor binding domain (RBD) of the S protein.
3. The composition of claim 1 or 2, wherein the composition further comprises at least one additional SARS-CoV-2 antigen.
4. The composition of claim 3, wherein the additional SARS-CoV-2 antigen comprises an S1 subunit of the S protein or an antigenic fragment thereof.
5. The composition of claim 3, wherein the additional SARS-CoV-2 antigen comprises an S2 subunit of the S protein or an antigenic fragment thereof.
6. The composition of claim 3, wherein the additional SARS-CoV-2 antigen comprises envelope protein (E) or an antigenic fragment thereof.
7. The composition of claim 3, wherein the additional SARS-CoV-2 antigen comprises membrane protein (M) or an antigenic fragment thereof.
8. The composition of any one of claims 1-7, wherein the SARS-CoV-2 antigen further comprises antigens from one or more SARS-CoV-2 variants.
9. The composition of claim 8, wherein the SARS-CoV-2 antigen comprises antigens from two or more SARS-CoV-2 variants.
10. The composition of any one of claims 1-9, wherein the composition further comprises at least one antigen which is not derived from SARS-CoV-2.
11. The composition of claim 10, wherein said antigen which is not derived from SARS-CoV-2 is an influenza antigen.
12. The composition of any one of claims 1-11, wherein the composition further comprises an adjuvant.
13. The composition of claim 12, wherein said adjuvant is monosodium urate crystal.
14. The composition of claim 12, wherein said adjuvant is a CpG oligodeoxynucleotide (CpG ODN).
15. The composition of claim 14, wherein said CpG ODN is CpG55.2.
16. The composition of claim 1, wherein the nanoparticle is made of mannose conjugated chitosan.
17. The composition of any one of claims 1-16, wherein the nanoparticle is conjugated with the SARS-CoV-2 antigen.
18. The composition of any one of claims 1-16, wherein the nanoparticle is entrapped within the SARS-CoV-2 antigen.
19. The composition of claim 18, wherein the SARS-CoV-2 antigen is entrapped within the nanoparticle by a water/oil emulsion.
20. A vaccine comprising the composition of any one of claims 1-19 in a pharmaceutically acceptable carrier.
21. The vaccine of claim 20, wherein the vaccine is formulated for intranasal delivery.
22. The vaccine of claim 20 or 21, wherein the vaccine is formulated for delivery as a drop.
23. The vaccine of claim 20 or 21, wherein the vaccine is formulated for delivery as a mist.
24. A method of eliciting or enhancing an immune response to SARS-CoV-2 in a subject, the method comprising administering to the subject a vaccine, wherein the vaccine comprises a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof, wherein said nanoparticle comprises chitosan; further wherein the vaccine further comprises a pharmaceutically acceptable carrier.
25. The method of claim 24, wherein the subject is a human.
26. The method of claim 24 or 25, wherein the vaccine is administered intranasally.
27. The method of any one of claims 24-26, wherein the vaccine comprises at least one additional antigen other than the first spike protein (S) or an antigenic fragment thereof, and the first nucleocapsid protein (N) or antigenic fragment thereof.
28. The method of claim 27, wherein the vaccine comprises antigens for two or more different variants of SARS-CoV-2.
29. The method of any one of claims 24-28, wherein the vaccine also comprises antigens for a non-SARS-CoV-2 virus.
30. The method of claim 29, wherein the non-SARS-CoV-2 virus is an influenza virus.
31. The method of any one of claims 24-30, wherein the subject has previously been vaccinated against SARS-CoV-2 with an intramuscular injection.
32. The method of claim 31, wherein the subject was vaccinated against a different variant.
33. The method of claim 31, wherein the subject was vaccinated against the same variant.
34. The method of any one of claims 24-33, wherein the subject has previously been infected with SARS-CoV-2.
35. The method of claim 34, wherein the subject was infected with the same variant.
36. The method of claim 34, wherein the subject was infected with a different variant.
37. The method of any one of claims 24-36, wherein the vaccine is administered in droplet form.
38. The method of any one of claims 24-36, wherein the vaccine is administered in mist form.
39. The method of any one of claims 24-38, wherein the vaccine is administered to the subject more than once.
40. The method of claim 39, wherein the vaccine is administered to the subject at least twice, with each dose being at least 30 days apart, wherein the subject has not previously been exposed to SARS-CoV-2 or a vaccine thereof.
41. The method of claim 39 or 40, wherein a booster vaccine is given at least 30 days after the initial vaccine is given.
42. The method of any one of claims 24-41, wherein the vaccine is given in an amount of 50 to 500 ug per dose.
43. A method of preventing or lessening the severity of symptoms associated with SARS-CoV-2 infection in a subject, the method comprising administering to the subject a vaccine, wherein the vaccine comprises a SARS-CoV-2 antigen associated with a nanoparticle, wherein the SARS-CoV-2 antigen comprises spike protein (S) or an antigenic fragment thereof, and a nucleocapsid protein (N) or an antigenic fragment thereof, wherein the nanoparticle comprises chitosan; and further wherein the vaccine further comprises a pharmaceutically acceptable carrier.
44. The method of claim 43, wherein the subject is a human.
45. The method of claim 43 or 44, wherein the vaccine is administered intranasally.
46. The method of any one of claims 43-45, wherein the vaccine comprises at least one additional antigen other than the spike protein (S) or an antigenic fragment thereof, and the nucleocapsid protein (N) or antigenic fragment thereof.
47. The method of claim 46, wherein the vaccine comprises antigens for two or more different variants of SARS-CoV-2.
48. The method of claim 46, wherein the vaccine also comprises antigens for a non-SARS-CoV- 2 virus.
49. The method of claim 48, wherein the non-SARS-CoV-2 virus is an influenza virus.
50. The method of any one of claims 43-49, wherein the subject has previously been vaccinated against SARS-CoV-2 with an intramuscular injection.
51. The method of claim 50, wherein the subject was vaccinated against a different variant.
52. The method of claim 50, wherein the subject was vaccinated against the same variant.
53. The method of any one of claims 43-52, wherein the subject has previously been infected with SARS-CoV-2.
54. The method of claim 53, wherein the subject was infected with the same variant.
55. The method of claim 53, wherein the subject was infected with a different variant.
56. The method of any one of claims 43-55, wherein the vaccine is administered in droplet form.
57. The method of any one of claims 43-55, wherein the vaccine is administered in mist form.
58. The method of any one of claims 43-57, wherein the vaccine is administered to the subject more than once.
59. The method of claim 58, wherein the vaccine is administered to the subject at least twice, with each dose being at least 21 days apart, wherein the subject has not previously been exposed to SARS-CoV-2 or a vaccine thereof.
60. The method of claim 58 or 59, wherein a booster vaccine is given at least 21 days after the initial vaccine is given.
PCT/US2023/062681 2022-02-15 2023-02-15 Nanotechnology based intranasal vaccine for covid-19 comprising chitosan WO2023159082A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263310215P 2022-02-15 2022-02-15
US63/310,215 2022-02-15
US202263417817P 2022-10-20 2022-10-20
US63/417,817 2022-10-20

Publications (2)

Publication Number Publication Date
WO2023159082A2 true WO2023159082A2 (en) 2023-08-24
WO2023159082A3 WO2023159082A3 (en) 2023-10-05

Family

ID=87579113

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/062681 WO2023159082A2 (en) 2022-02-15 2023-02-15 Nanotechnology based intranasal vaccine for covid-19 comprising chitosan

Country Status (1)

Country Link
WO (1) WO2023159082A2 (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200306364A1 (en) * 2017-11-30 2020-10-01 Ohio State Innovation Foundation Mucoadhesive nanoparticle entrapped influenza virus vaccine delivery system
AU2021254768A1 (en) * 2020-04-14 2022-11-17 The Regents Of The University Of California Large sequence pan-Coronavirus vaccine compositions
EP4178627A1 (en) * 2020-07-13 2023-05-17 Oregon Health & Science University Immunogenic constructs, compositions, and methods for inducing immune response

Also Published As

Publication number Publication date
WO2023159082A3 (en) 2023-10-05

Similar Documents

Publication Publication Date Title
Chavda et al. Intranasal vaccines for SARS-CoV-2: From challenges to potential in COVID-19 management
KR100764678B1 (en) A vaccine composition comprising alpha-galactosylceramide as an adjuvatnt for intranasal administration
TWI737584B (en) Hand, foot, and mouth vaccines and methods of manufacture and use thereof
Garlapati et al. Enhanced immune responses and protection by vaccination with respiratory syncytial virus fusion protein formulated with CpG oligodeoxynucleotide and innate defense regulator peptide in polyphosphazene microparticles
KR102483033B1 (en) PEGylated Liposomes and Uses Thereof
JP5809560B2 (en) Vaccine composition for use against influenza
Cho et al. Enhanced humoral and cellular immune responses after sublingual immunization against human papillomavirus 16 L1 protein with adjuvants
EP0889736B1 (en) Iscom or iscom-matrix comprising a mucus targetting substance and an antigen
EP2066332A1 (en) Flu vaccine admixture of mannan and flu antigen
JP2023549815A (en) Protein-based nanoparticle vaccine for metapneumovirus
US10682314B2 (en) Nanoparticle based vaccine strategy against swine influenza virus
US9585954B2 (en) Mucosal immunization
JP2023091085A (en) mucosal adjuvant
S Almeida et al. Nasal vaccines against hepatitis B: an update
Silveira et al. Quillaja brasiliensis nanoparticle adjuvant formulation improves the efficacy of an inactivated trivalent influenza vaccine in mice
WO2023159082A2 (en) Nanotechnology based intranasal vaccine for covid-19 comprising chitosan
WO2023159081A2 (en) Nanotechnology based intranasal vaccine for covid-19
JPWO2013085021A1 (en) Composition for enhancing antibody production
WO2008037033A1 (en) Flu vaccine admixture of mannan and flu antigen
JP2005082581A (en) SECRETORY IgA ANTIBODY INDUCER
Morein et al. New ISCOMs meet unsettled vaccine demands
Nath et al. A Pervasive Review on New Advancements of Nano Vaccines on Covid-19 Pandemic
US20220257752A1 (en) New use of cyclic dinucleotides
KR20240052044A (en) Virus-like particles for treating or preventing infection by coronaviruses
Eze et al. Vaccine Delivery Using Nanoparticles: A Critical Look at ISCOMs 4 Decades but 2 and Still Counting.

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: 23757062

Country of ref document: EP

Kind code of ref document: A2