WO2021207281A2 - Vaccines, adjuvants, and methods of generating an immune response - Google Patents

Vaccines, adjuvants, and methods of generating an immune response Download PDF

Info

Publication number
WO2021207281A2
WO2021207281A2 PCT/US2021/026058 US2021026058W WO2021207281A2 WO 2021207281 A2 WO2021207281 A2 WO 2021207281A2 US 2021026058 W US2021026058 W US 2021026058W WO 2021207281 A2 WO2021207281 A2 WO 2021207281A2
Authority
WO
WIPO (PCT)
Prior art keywords
vaccine
antigen
adjuvant
virus
cov
Prior art date
Application number
PCT/US2021/026058
Other languages
French (fr)
Other versions
WO2021207281A3 (en
Inventor
Fabian Gusovsky
Lynn Hawkins
Sally Ishizaka
David Evander Anderson
Anne-Catherine Fluckiger
Original Assignee
Eisai R&D Management Co., Ltd.
Variation Biotechnologies Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eisai R&D Management Co., Ltd., Variation Biotechnologies Inc. filed Critical Eisai R&D Management Co., Ltd.
Priority to IL297093A priority Critical patent/IL297093A/en
Priority to MX2022012527A priority patent/MX2022012527A/en
Priority to EP21722639.8A priority patent/EP4132577A2/en
Priority to BR112022020298A priority patent/BR112022020298A2/en
Priority to CN202180040867.8A priority patent/CN115916254A/en
Priority to US17/995,546 priority patent/US20230149537A1/en
Priority to JP2022560944A priority patent/JP2023520603A/en
Priority to KR1020227038812A priority patent/KR20230034936A/en
Priority to AU2021252972A priority patent/AU2021252972A1/en
Priority to CA3179739A priority patent/CA3179739A1/en
Publication of WO2021207281A2 publication Critical patent/WO2021207281A2/en
Publication of WO2021207281A3 publication Critical patent/WO2021207281A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/215Coronaviridae, e.g. avian infectious bronchitis virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • 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
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5258Virus-like particles
    • 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
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/70Multivalent vaccine
    • CCHEMISTRY; METALLURGY
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13023Virus like particles [VLP]
    • 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
    • C12N2770/10011Arteriviridae
    • C12N2770/10034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • Embodiments of the present disclosure relate to vaccines including adjuvants and antigens, in addition to methods of generating an immune response using those vaccines for the mitigation of infection by a coronavirus.
  • Vaccines have proven to be successful methods for the mitigation of infectious diseases. Generally, they are cost effective, and do not induce antibiotic resistance to the target pathogen or affect normal flora present in the host. In many cases, such as when inducing anti-viral immunity, vaccines can prevent a disease for which there are no viable curative or ameliorative treatments available.
  • Vaccines function by triggering the immune system to mount a response to an agent, or antigen, typically an infectious organism or a portion thereof that is introduced into the body in a non-infectious or non-pathogenic form.
  • an agent typically an infectious organism or a portion thereof that is introduced into the body in a non-infectious or non-pathogenic form.
  • later exposure of the immune system to this organism as an infectious pathogen results in a rapid and robust immune response that destroys the pathogen before it can multiply and infect enough cells in the host organism to cause disease symptoms.
  • the agent, or antigen, used to prime the immune system can be the entire organism in a less infectious state, known as an attenuated organism, or in some cases, components of the organism such as carbohydrates, proteins or peptides representing various structural components of the organism.
  • Freund's complete adjuvant consists of a mixture of mycobacteria in an oil/water emulsion. Freund's adjuvant works in two ways: first, by enhancing cell and humoral-mediated immunity, and second, by blocking rapid dispersal of the antigen challenge (the “depot effect”). However, due to frequent toxic physiological and immunological reactions to this material, Freund's adjuvant cannot be used in humans.
  • LPS lipopolysaccharide
  • TLR-4 human toll-like receptor-4
  • Aluminum-based adjuvants have been used in human vaccines since 1932 and, although they have a long record of safety, their mode of action is not completely understood. It is generally believed that aluminum-based adjuvants enhance immune response by activation of dendritic cells.
  • the two most frequently used aluminum- based adjuvants are referred to as “aluminum phosphate” and “aluminum hydroxide”, with aluminum hydroxide adjuvant being the most widely used commercially.
  • Aluminum hydroxide adjuvant is not Al(OH)3, but rather crystalline aluminum oxyhydroxide (AIOOH) which has a larger surface area than crystalline aluminum hydroxide.
  • Aluminum phosphate adjuvant is actually amorphous aluminum hydroxy phosphate (Al(OH) x (P04) y ) in which some of the hydroxyl groups of aluminum hydroxide adjuvant are replaced by phosphate groups.
  • the surface of aluminum phosphate adjuvant is composed of Al-OH and AI-OPO3 groups. Although they are chemically similar, the two adjuvants have different chemical properties. They are often both referred to simply as “alum” adjuvants.
  • E6020 is a potent TLR-4 receptor agonist, and thus is useful as an immunological adjuvant when co-administered with antigens in vaccines.
  • Toll-like receptors (TLRs) belong to the family of innate immune receptors, which play an important role in the activation of innate immunity, regulation of cytokine expression, indirect activation of the adaptive immune system, and the recognition of pathogen-associated molecular patterns (PAMPs).
  • TLRs Toll-like receptors
  • E6020 has been reported for use in combination with antigen or vaccine components, e.g ., an antigenic agent selected from the group consisting of antigens from pathogenic and non-pathogenic organisms, viruses, and fungi.
  • E6020 has been reported for use as an adjuvant in combination with proteins, peptides, antigens and vaccines that are pharmacologically active for disease states and conditions including staphylococcus aureus, pertussis toxin, tetanus, influenza, Chagas disease, meningococcus, HIV, cancer, chlamydia, cytomegalovirus, Leishmaniasis, and whooping cough (caused by pertussis toxin).
  • E6020 and the antigen are each present in an amount effective to elicit an immune response when administered to a host animal, embryo, or ovum being vaccinated therewith.
  • Coronaviruses are a genus in the Coronaviridae family and are pleomorphic, enveloped viruses. See S. Perlman et al, Nature Reviews Microbiology, 7:439-450 (2009). Coronaviruses contain a single stranded, 5 '-capped, positive strand RNA molecule that ranges from 26-32 kb and contain at least 6 open reading frames. Coronaviruses use host proteins as part of their replication strategies. Immune, metabolic stress, cell cycling, and other cellular pathways are activated by infection. See Tang Y. et al, Front. Immunol. 11 : 1708 (2020).
  • Coronaviruses usually cause mild to moderate upper-respiratory tract illnesses, like the common cold, in people. See “Coronaviruses,” National Institute of Allergy and Infectious Disease, https://www.niaid.nih.gov/diseases-conditions/coronaviruses (accessed on April 1,
  • coronaviruses that affect animal species. Seven coronaviruses are known to cause human disease. Four of these coronaviruses are mild: viruses 229E, OC43, NL63 and HKU1; three of the coronaviruses can have more serious outcomes in people: SARS (severe acute respiratory syndrome), which emerged in late 2002 and disappeared by 2004; MERS (Middle East respiratory syndrome), which emerged in 2012 and remains in circulation in camels; and COVID-19, which emerged in December 2019 (a global effort is under way to contain its spread).
  • SARS severe acute respiratory syndrome
  • MERS Middle East respiratory syndrome
  • COVID-19 which emerged in December 2019 (a global effort is under way to contain its spread).
  • COVID-19 is caused by the coronavirus known as SARS-CoV-2 (also known as 2019-nCoV).
  • SARS-CoV-2 has been shown to cause mild to fatal symptoms in the human population. See Hantoushzadeh S. et al ., Arch. Med. Res. 51: 347-348 (2020) and Ingraham N. E. et al., Lancet Respir. Med. 8: 544-546 (2020).
  • SARS-CoV-2 also known as 2019-nCoV
  • SARS-CoV-2 has been shown to cause mild to fatal symptoms in the human population. See Hantoushzadeh S. et al ., Arch. Med. Res. 51: 347-348 (2020) and Ingraham N. E. et al., Lancet Respir. Med. 8: 544-546 (2020).
  • SARS - CoV - 2 viral spike protein (S) binds with the extracellular domain of various TLRs including TLR1, TLR4, and TLR6, with the strongest binding with TLR4.
  • Embodiments provided herein include vaccines comprising E6020 as an adjuvant and methods of generating an immune response using those vaccines for the mitigation of infection by coronavirus.
  • E6020 may be used as an adjuvant in vaccines directed to mitigation of nidovirales infection. Further embodiments relate to use as an adjuvant in vaccines directed to mitigation of coronavirus infection. Yet further embodiments relate to use of E6020 as an adjuvant in vaccines directed to mitigation of infection caused by the virus SAR-CoV-2 (aka COVID-19). [0015] Embodiments include, for example, a vaccine including E6020 and an antigen related to coronavirus. Yet further embodiments relate to a vaccine including a virus like particle containing a SARS-CoV-2 spike protein and E6020.
  • Vaccines prepared according to embodiments presented herein may include one or more adjuvants in addition to E6020 to form an adjuvant system.
  • an embodiment of the invention includes a vaccine comprising a virus like particle containing a SARS-CoV-2 spike protein, E6020 and aluminum phosphate adjuvant.
  • Vaccines may be formulated in a number of ways. For example, they may be formulated as a buffered solution, as an emulsion, as microparticles, or as nanoparticles (for example, as gene nanoparticles).
  • Vaccines prepared according to embodiments as reported herein may also include pharmaceutically acceptable additives. These include, for example, polymer additives and/or surfactant additives. Polymer and surfactant additives may be particularly useful, for example, in emulsion formulations.
  • FIG. 1 For purposes of clarity, a subject is an unvaccinated human or a human who has undergone some but not all of the recommended number of doses of vaccine.
  • a subject may be a person already exposed or infected by a coronavirus, in whom a boosted immune response following vaccination is sought.
  • FIG. 1 depicts the detection of SARS-CoV-2 native S eVLP vaccines formulated with different adjuvants.
  • FIG. 2 depicts the neutralizing antibody titers from the pooled sera of C75BL/6 mice administered a SARS-CoV-2 eVLP vaccine.
  • FIG. 3 depicts the antibody and T cell responses of SARS-CoV-2 native S eVLP vaccines combined with different adjuvants in C75BL/6 mice.
  • FIG. 4 depicts the structures of SARS-CoV-2 S protein constructs and a Western blot analysis of the protein expression for each SARS-CoV-2 S protein construct.
  • FIG. 5 depicts the antibody titers in the sera from 20 COVID-19 confirmed convalescent patients, wherein the sera samples were separated into groups with either high or low levels of Ab binding activity to recombinant SARS-CoV-2 S.
  • FIG. 6 depicts humoral responses from various types of SARS-CoV-2 eVLP vaccines in C75BL/6 mice.
  • FIG. 7 depicts the antibody and T cell responses of different monovalent constructs of SARS-CoV-2 SPG vaccines in C75BL/6 mice.
  • FIG. 8 depicts the neutralizing antibody response in mice vaccinated with VBI-2902 and VBI-2901 with and without E6020.
  • FIG. 9 depicts the body weight changes of Syrian golden hamsters vaccinated with VBI- 2902 vaccines with and without E6020.
  • FIG 10 depicts the serum antibody titers of Syrian golden hamsters vaccinated with VBI- 2902 vaccines with and without E6020.
  • FIG. 11 depicts the viral RNA levels of Syrian golden hamsters vaccinated with VBI-
  • Vaccines including E6020 and methods for treatment of coronavirus infection using those vaccines will be described in detail.
  • E6020 is present in these vaccines as an adjuvant, which is a compound that is included in a vaccine to increase the vaccine’s ability to provoke, increase, and/or extend an immune response, or drive a favorable type of immune mechanism(s).
  • E6020 is a di sodium salt of ER-804057 and is shown below:
  • the vaccines described herein include from 0.1 ⁇ g to 100 ⁇ gs, 0.5 ⁇ g to 100 ⁇ gs, 1 ⁇ g to 50 ⁇ g, 1 ⁇ g to 25 ⁇ g, 1 ⁇ g to 20 ⁇ g, 5 ⁇ g to 30 ⁇ g, 0.5 ⁇ g to 10 ⁇ g, 10 ⁇ g to 20 ⁇ g or 20 ⁇ g to 50 ⁇ g of E6020.
  • the vaccine comprises 10 ⁇ gs of E6Q20.
  • Antigens for Inclusion are molecules that may be recognized by the immune system of a patient to generate an immune response and/or cell-mediated immunity.
  • E6020 is useful as an adjuvant in a vaccine where the antigen is a nidovirales antigen, a coronavirus antigen, or a SARS-CoV-2 antigen.
  • Antigens may be present in a vaccine in various forms.
  • an antigen may be present as purified antigen molecules (which may be, for example, proteins, multimerized proteins, protein subunits (including subunit trimers), peptides (including Ii-key peptides and locked peptides), peptides conjugated to a protein carrier, oligonucleotides, RNA (including mRNA), DNA, plasmid DNA, or polysaccharides, conjugated to a carrier or not), on live-attenuated, recombinant or inactivated-whole viruses, as dendritic cells, as antigen presenting cell vectors, as recombinant viral vectors, as adenoviral vectors, through liposomal delivery vehicles including lipoproteins or lipopolyplexes, or by a composition that includes a nucleic acid that encodes antigen.
  • purified antigen molecules which may be, for example, proteins, multimerized proteins, protein subunits (including subunit trimers), peptides (including Ii-key peptide
  • the antigen is derived from a viral or bacterial pathogen, such as an influenza or corona virus.
  • Vaccines described herein may comprise antigens, wherein the antigens are presented on an enveloped virus like particle (“eVLP”).
  • eVLPs may comprise retroviral vectors that lack a retrovirus-derived genome and are therefore non-replicating.
  • Retroviruses are enveloped RNA viruses that belong to the family Retroviridae. After infection of a host cell by a retrovirus, RNA is transcribed into DNA via the enzyme reverse transcriptase. DNA is then incorporated into the host cell’s genome by an integrase enzyme and thereafter replicates as part of the host cell’s DNA.
  • eVLPs may be comprised of a structural polyprotein from a Moloney Murine Leukemia Virus (MMLV) known as a Gag protein (SEQ ID NO: 5). Expression of Gag in some host cells can result in self-assembly of the expression product into eVLPs.
  • MMLV Moloney Murine Leukemia Virus
  • SEQ ID NO: 5 Moloney Murine Leukemia Virus
  • the antigen may be derived from a viral or bacterial pathogen, such as an influenza virus or a corona-virus.
  • the antigen is derived from a Nidovirales virus.
  • the antigen present within the vaccine resembles a SARS-CoV-2 spike (S) protein.
  • SARS-CoV-2 spike (S) protein plays a crucial role in host cell receptor binding and fusion properties leading to virus entry. See Patra R. et al., J Med Virol. 93:615-617
  • SARS-CoV-2 S proteins resemble features of class I viral proteins, wherein they are constituted of 2 subunits, SI, containing the receptor binding domain (RBD) and S2, containing the fusion entry domain. Binding of the RBD to the host cell receptor induces conformational changes resulting in activation of the protease cleavage site upstream of the fusion domain followed by release and activation of the S2 fusogenic domain.
  • SARS-CoV-2 S protein contains a furin cleavage site located at the boundary of SI and S2 enabling rapid processing of the S protein during biosynthesis in host cells.
  • the antigen present within the vaccine can be a native SARS-CoV-2 S protein, a stabilized prefusion form of a SARS-CoV-2 S protein, a modified SARS-CoV-2 S wherein the TMCTD domain has been replaced with a transmembrane and cytoplasmic terminal domain of vesicular stomatitis virus G (VSV-G) or any combination thereof.
  • VSV-G vesicular stomatitis virus G
  • the SARS-CoV-2 spike antigen present within the vaccine may possess, for example, a protein sequence provided by SEQ ID NOs: 1-4.
  • the vaccine may comprise from 0.1 ⁇ g to 100 ⁇ g, 0.1 ⁇ g to 50 ⁇ g, 1 ⁇ g to 25 ⁇ g, 5 ⁇ g to 25 ⁇ g or 5 ⁇ g to 10 ⁇ g of an antigen.
  • E6020 as an adjuvant. These include, for example, but are not limited to those listed in Table 1
  • the vaccine may comprise additional adjuvants in combination with E6020.
  • adjuvants such as aluminum hydroxide adjuvant (crystalline aluminum oxyhydroxide (AIOOH)) or aluminum phosphate adjuvant (amorphous aluminum hydroxy phosphate (for example Adju-Phos®)), an aluminum salt, chemokines, cytokines, nucleic acid sequences (particularly bacterial nucleic acid systems), lipoprotein, lipopolysaccharide (LPS), monophosphoryl lipid A, lipoteichoic acid, imiquimod, reiquimod, QS-21 or any combination thereof.
  • aluminum hydroxide adjuvant crystalline aluminum oxyhydroxide (AIOOH)
  • aluminum phosphate adjuvant amorphous aluminum hydroxy phosphate (for example Adju-Phos®)
  • an aluminum salt chemokines, cytokines, nucleic acid sequences (particularly bacterial nucleic acid systems), lipoprotein, lipopolysaccharide (LPS), monophospho
  • TLR agonists may be useful in adjuvant systems with E6020.
  • other adjuvants may be agonists that activate TLR 2, 3, 5, 7, 8, 9 or a combination thereof.
  • Use of E6020 in combination with other adjuvants may include simultaneous or sequential administration of the adjuvants in the system.
  • the vaccine further comprises from 50 ⁇ g to 50 mg, 0.1 mg to 20 mg, 1 mg to 5 mg, 3 mg to 5 mg or 0.1 mg to 1 mg of at least one additional adjuvant. In other embodiments, the vaccine further comprises 150 ⁇ g to 180 ⁇ g, such as 165 ⁇ g, of at least one additional adjuvant.
  • the vaccine further comprises from 0.1 mg/mL to 1 mg/mL, from 0.05 mg/mL to 0.9 mg/mL, 0.5 mg/mL to 1.0 mg/mL, 0.1 mg/mL to 0.5 mg/mL, 0.1 mg/mL to 5.0 mg/mL or 0.165 mg/mL to 0.33 mg/mL of at least one additional adjuvant.
  • the vaccine further comprises 0.165 mg/mL or 0.33 mg/mL of at least one additional adjuvant.
  • the vaccine may also comprise up to 800 mg/mL of at least one additional adjuvant.
  • the amount of the at least one additional adjuvant is based on the aluminum content within the at least one adjuvant.
  • eVLP SARS-CoV-2 vaccines formulated with alum (US 17/218148) which induced a strong immune response against the SARS-CoV-2 spike protein in mice studies. These vaccines were shown to be strongly protective against COVID-19 infection in Golden Hamsters challenged with SARS-CoV-2. As described in US 17/218148, the eVLP SARS-CoV-2 vaccines can be formulated with a variety of different adjuvants, several of which (including E6020) were tested in mice as described in Example 4.
  • Example 4 the use of E6020 as an additional adjuvant to the eVLP/alum formulation resulted in a significantly stronger immune response than the use of other additional adjuvants. Furthermore, and quite surprisingly, the use of E6020 as an additional adjuvant significantly enhanced Thl-type T cell responses and a change of IgG profile with induction of significant higher IgG2 responses than seen in the formulation with the eVLP SARS-CoV-2 vaccine with alum alone or with other additional adjuvants such as mimics of AS03 and AS04. This enhanced Thl response represents a shift in immune response toward a Thl from Th2. The Thl response is correlated with immunity against viral infection. Accordingly, the unexpected and significant enhancement of the Thl response induced by the combination of the eVLP SARS-CoV-2 vaccine formulated with alum and E6020 indicates that this combination is a highly potent immunogenic composition against COVID-19.
  • Vaccines including E6020 may be delivered using multiple modes of administration and multiple formulations.
  • formulations typically include at least one pharmaceutically acceptable carrier.
  • a carrier may be, for example, a diluent, excipient, or vehicle used in administration of the E6020.
  • Carriers may include, for example, water or saline.
  • Methods of administration include, but are not limited to, parenteral, e.g., intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal and intrathecal. Administration can also occur systemically.
  • Vaccines may be formulated for parenteral administration by injection. This may include, for example, bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers. They may include a preservative. Injectable dosage forms include suspensions, solutions or emulsions. They may be in oily or aqueous vehicles. They may contain further additives including suspending, stabilizing and/or dispersing agents. The vaccine may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen free water, before use.
  • a suitable vehicle e.g., sterile pyrogen free water
  • Vaccines may be delivered, for example, as an aerosol spray presentation from pressurized packs or a nebulizer.
  • a propellant is also used.
  • a suitable propellant may be dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other gas.
  • a nasal spray which is not pressurized or which is pressurized mechanically rather than chemically, can be used for intranasal administration.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges may be made for use in an inhaler or insufflater. These would contain a powder mix of the compound and a suitable powder base such as lactose or starch.
  • Embodiments may be delivered using virus-like particles (VLPs), microparticles or nanoparticles.
  • a nanoparticle may include a peptide nucleic acid oligomer conjugated to a lipid.
  • the oligomer complexes with an antigen and/or an adjuvant, forming a nanoparticle for delivery according to one of the methods reported herein.
  • Particle-based delivery including microparticle or nanoparticle based delivery, may be, for example, protein- based scaffolds or matrices, lipid-based scaffolds or matrices, or polymer-based scaffolds or matrices.
  • E6020 is formulated with enveloped virus-like particles (eVLPs) that possess a stable core of Gag protein and a lipid bilayer.
  • eVLPs structurally resemble viruses but are much safer to administer because they lack the genetic material needed to replicate within the host.
  • eVLPs enable repeating, array-like presentation of antigens which is a preferred means of activating B cells and eliciting high affinity antibodies.
  • eVLPs that can be used as vaccines can be, but are not limited to, MLV-Gag eVLPs and those disclosed in US Patent 9,765,304 and US Patent Application No. 17/218,148.
  • the vaccine comprises eVLP particles comprising 0.05 mgs to 0.50 mgs, 1 ⁇ g to 50 mg, 10 ⁇ g to 10 mg, 50 ⁇ g to 5 mg or 100 ⁇ g to 500 ⁇ g of Gag protein.
  • the vaccine comprises eVLP particles comprising an amount of antigen and an amount of Gag protein, wherein the amount of antigen is from 0.1 % to 4.0 % relative to the amount of Gag protein.
  • the vaccine comprises eVLP particles comprising a of murine leukemia virus (MMLV) Gag protein.
  • MMLV murine leukemia virus
  • the Gag protein is a MMLV-Gag protein according to SEQ ID NO: 5.
  • a typical treatment regimen includes administration of an amount effective or likely to lead to mitigation of an infectious agent over a period of time. That may range from a few hours to a few days, or to a few months.
  • Mitigation may include prevention of a disease, minimization of symptoms, or alleviation of symptoms.
  • An effective treatment may provide mitigation for a lifetime, for decades, for years, for months, or for even a single month.
  • the amount of mitigation may increase, decrease, or both over the time that the mitigation is in effect.
  • non-antigen, non-adjuvant, non-carrier compounds may be administered in or in conjunction with vaccines reported herein.
  • vaccines may include or may be administered with a separate antiviral agent, an antibacterial agent, antifungal agent, or other antibiotic. These may include, for example, oseltamivir, azithromycin, chloroqine, hydroxychloroquine, or zanamivir.
  • the vaccines described herein may be administered in doses ranging from 0.1 mL to 10 mL, 0.2 mL to 5 mL or 0.3 mL to 3 mL.
  • Example 1 An experiment to demonstrate adjuvant activity would examine in vivo mouse immune response, comparing the strength and characteristics of response to antigen administered with or without the candidate adjuvant.
  • Example l is a prophetic example.
  • mice are immunized with a SARS-Coactivated SARS-Coactivated virus
  • CoV-2 antigen in vaccine formulations with or without E6020, in order to assess the effect of E6020 on the response to immunization.
  • Antigens may be chosen for their potential to raise protective antibodies, cytotoxic T cell responses, or both (Grifoni A, Sidney J, Zhang Y, Scheuermann RH, Peters B, Sette A. Cell Host Microbe. 2020 Mar 12. pii: S1931- 3128(20)30166-9. doi: 10.1016/j.chom.2020.03.002.).
  • the SARS-CoV-2 antigen is recombinantly produced S protein.
  • the S protein, or the receptor-binding domain of the S protein can be produced by introduction of an appropriate vector into HEK cells.
  • the receptor-binding domain of the S protein S331-524 (Tai etal ., Cellular and Molecular Immunology; https://doi.org/10.1038/s41423-020-0400-4) is expressed and fused to a carrier such as the human antibody Fc to allow secretion and purification.
  • a carrier such as the human antibody Fc
  • Other approaches might include expression of full-length spike ectodomain or other subdomains of the S protein (Wang etal ., https://doi.org/10.1101/2020.03.ll.987958 doi: bioRxiv preprint).
  • an inactivated whole virus preparation might be used as antigen.
  • S protein sequences may be produced as monomeric subunits, or produced as fused sequences containing multimerization sequences, purification tags or sequences that allow appropriate presentation of antigenic epitopes.
  • Various linkers or carriers of the antigenic domain can be utilized.
  • the Fc fusion is produced by inserting an appropriate S protein sequence into the pFUSE-hIgGl-Fc2 expression vector (InvivoGen, San Diego, CA) and expressing the resultant fusion protein by transfecting the vector into the human HEK-293 cell line.
  • secreted protein is recovered from the cell culture supernatant and isolated by an affinity method that selects for either the S protein (as for example with a specific anti-spike protein antibody) or the Fc sequence (e.g. using a protein A column) (ref. Tai et al. as above).
  • mice are immunized subcutaneously with between 10 and 100 micrograms of SARS-CoV-2 S protein. Three immunizations are given at three week intervals.
  • SARS-CoV-2 antigen is given in PBS, as an unadjuvanted control, or with a test adjuvant.
  • E6020 is tested at doses known to augment antibody responses to other antigens, for example 1.0, 3.0 or 10 micrograms.
  • Other adjuvants such as alum at 2.7 mg/dose, or appropriate doses of other commercially available adjuvant substances, are included as positive controls.
  • Blood samples are taken from immunized animals at two weeks after the second and/or the third immunization.
  • Mucosal samples are taken by appropriate lavage methods. Serum or lavage are isolated and tested for anti-antigen titers of immunoglobulin using standard ELISA methods.
  • an S protein construct might be expressed that includes the sequences used to immunize, and used to coat an ELISA plate. It is important that the ELISA antigen construct not contain carrier protein sequences that are the same as those used to immunize the mice, in order to avoid measuring spurious reactivities to carrier protein that are unrelated to anti-virus responses. Examples of ELISA methods for SARS-CoV-2 S protein are given in (Nisreen M.A.
  • An effective adjuvant is expected to increase the amount of anti-antigen antibody raised, cause titers to rise earlier or persist longer, or cause a shift in isotype as compared to antigen given alone. Changes in isotype response reflect differences in the cytokine patterns elicited by the adjuvant, which in turn is associated with effective protection against different types of infection. For example, IgG2a in mice is associated with an interferon-driven Thl response that might support cytotoxic T cells and give particularly efficient anti-viral responses.
  • Example 2 An experiment to demonstrate adjuvant-enhanced elicitation of neutralizing antibodies against the coronavirus, comparing the strength and characteristics of virus neutralizing response to antigen administered with or without E6020.
  • Example 2 is a prophetic example. Because ELISA titers may not correlate with the ability of serum antibodies to effectively protect against infection, it is helpful to measure neutralization titers elicited by immunization.
  • mice are immunized as in Example 1, one or more times. After immunization, serum or lavage are taken as described, and are used in neutralization assays.
  • a neutralization assay is performed by measuring the infectious potential of a pseudotyped virus which carries SARS-CoV-2 surface proteins but genetic material derived from a crippled HIV construct that cannot replicate but does express a tracking marker such as luciferase.
  • pseudotyped virus is harvested from HEK293 cells cotransfected with a plasmid encoding Env-defective, luciferase- expressing HIV-1 (pNL4-3.1uc.RE) and another encoding SARS-CoV-2 S protein, and pseudovirus-containing supernatants harvested subsequently.
  • Neutralization is assessed by incubation of pseudovirus with serially diluted mouse sera from vaccinated mice for 1 h at 37 °C, followed by addition of the mixture into hACE2- expressing HEK293 cells. After an appropriate incubation the cells are lysed and the resulting supernatant is mixed with luciferase substrate and tested for relative luciferase activity, typically using a luminometer to measure light output. This assay measures production of spike-specific antibodies capable of inhibiting viral interaction with cells.
  • neutralization by antibody binding to other viral proteins is measured by a classic plaque assay in which dilutions of infectious SARS-CoV-2 are preincubated with test sera before application to a confluent culture of Vero cells. Appearance of plaques, where cells have been lysed by viral replication, will occur in the absence of neutralizing antibody. The reduction of plaque frequency in the presence of neutralization is quantified by standard means (Okba et al., medRxivhttps://doi.org/10.1101 /2020.03.18.20038059).
  • Example 3 An experiment to demonstrate adjuvant-enhanced elicitation of neutralizing antibodies against the coronavirus, using an in vivo challenge of mice or another species in a viral infection model. Groups receiving vaccine with or without E6020 will be compared for their resistance to infection.
  • Example 3 is a prophetic example.
  • animals are immunized with a SARS-CoV-2 vaccine as described above and then exposed to live infectious virus in a challenge model.
  • the animals are mice that have been engineered to allow infection by SARS-CoV-2, for example through expression of the human ACE2 receptor protein in appropriate tissues (McCray et al. JOURNAL OF VIROLOGY, Jan. 2007, p. 813-821).
  • animal species that are susceptible to infection with SARS-CoV-2 even in the absence of transgene introduction such as ferrets or cats (Shi et al.
  • animals are challenged by an appropriate route, e.g. intranasally, with a dose of SARS-CoV-2 that is known to produce infection and replication.
  • Immunization groups are compared by measuring physical symptoms (for example body temperature, oxygenation or mortality), or alternatively suppression of in vivo viral replication by the vaccine- induced immune response may be assessed by measuring viral titers in target tissues such as the lungs. This is performed by PCR, measurement of viral load in tissue homogenates (Stadler et al., Emerging Infectious Diseases ⁇ www.cdc.gov/eid ⁇ Vol. 11, No. 8, August 2005, p. 1312), or other measures such as immuno-staining for viral antigens in tissue sections.
  • a decreased presence of virus in animals receiving vaccine adjuvanted with E6020 indicates the superior protective effect of this adjuvant.
  • Example 4 Influence of different adjuvants on antibody and T cell responses induced by monovalent SARS-CoV-2 eVLP vaccines.
  • Th2-type response has been suggested to contribute to the “cytokine storm” associated with severe lung pathologies (Peeples, PNAS 2020;
  • S eVLP vaccines express the native S protein of SARS-CoV-2 (SEQ ID NO: 1). For this purpose, mimics of MF59, the adjuvant systems AS03 and AS04, and E6020 co-formulated with aluminum phosphate (Adju-Phos) were evaluated.
  • the formulations of the native SARS-CoV-2 S eVLP vaccines with the various adjuvants are provided in Table 1.
  • SARS-CoV-2 native S eVLPs were used to compare the effects of the adjuvants as they were less immunogenic than eVLPs expressing the prefusion form of the S protein and might better enable differences in the adjuvants to be observed.
  • Mice received 2 IP injections of the native SARS-CoV-2 eVLPs formulated with the various adjuvants. The IgG binding titers, neutralizing antibody titers and Ab and T cell responses was assessed 14 days after the second injection.
  • FIG. 1 Western blot analysis of native SARS-CoV-2 eVLPs vaccines (FIG. 1) was performed using a primary antibody that is a SARS-CoV-2 (2019-nCoV) Spike RBD Rabbit Antibody (Cat# 40592-T62 Sinobiological) (1/5,000 dilution).
  • the secondary antibody is a goat anti-rabbit IgG-Fc HRP-conjugated (Bethyl, Cat# A120-11 IP- 18) lmg/mL - 1/10,000 Precision Protein Streptactin HRP conjugate (Bio-Rad, cat # 161-0381) (1/10,000 dilution).
  • TA 1 is the AS03 vaccine (see Group 1 in Table 1)
  • TA 2 is the E6020 + aluminum phosphate adjuvant vaccine (see Group 2 is Table 1)
  • TA 3 is the AS04 modified vaccine (see Group 3 in Table 1)
  • TA 4 is the AS04 vaccine (see Group 4 in Table 1)
  • TA 5 is the ASOIB vaccine (see Group 5 of Table 1)
  • TA 6 is the MF59 vaccine (see Group 6 of Table 1)
  • the control lane is a Native Monovalent SARS-CoV-2 vaccine from 29CH07- Post TFF/UC Pellet Filtered BDS (Lot#V20200501- nCOVID)
  • Lane 9 is 0.025 ⁇ g of a recombinant SARS-CoV-2 (Srn T2 #DL2020APR30 0.46/mL) vaccine
  • Lane 10 is 3.65 ⁇ g of 19CH102-Psot TFF/UC Pellet Filtered BDS (V20200409-EG).
  • Table 3 Native SARS-CoV-2 S eVLP vaccine virus-neutralizing titers
  • the neutralizing anti-body titers from the pooled sera of the treatment groups are depicted in FIG. 2.
  • the neutralizing anti-body titers at P1VD14 and at P2VD14 were measured.
  • the neutralizing anti-body titers of Group 2 were similar to those of Group 1 and higher than those of Groups 3-4.
  • the data presented within FIG. 2 demonstrates that the use of E6020 as an additional adjuvant significantly improved the effectiveness of the native monovalent SARS- CoV-2 vaccine formulated with the aluminum phosphate adjuvant.
  • MF59 enhanced Thl-type T cell responses compared to alum (FIG. 3A) but induced similar Ab responses (FIG. 3B-C) and comparable, balanced IgG2/IgGl ratio (FIG. 3D).
  • E6020 to the eVLP+aluminum phosphate adjuvant significantly enhanced Thl-type T cell responses and a change of IgG profile with induction of significant higher IgG2 than with Alum alone, demonstrating Thl polarization of both T cell and antibody responses.
  • Mimics of AS03 and AS04 adjuvants also biased responses towards a Thl-type response, though to a lesser extent than E6020.
  • Example 5 Contruction of SARS-CoV-2 eVLP antigens and vaccines
  • S SARS-CoV-2 S protein
  • SP SEQ ID NO: 2
  • the furin cleavage site of S was inhibited by mutation of the RRAR into GSAS and 2 proline substitutions were introduced at successive residues K986 and V987.
  • TM-CTD transmembrane and cytoplasmic terminal domains
  • FIG. 4B lane 2-3
  • Expression of S was slighly improved by the VSV-G swap in SG, and more dramatically enhanced by the inhibition of the cleavage sites in SP and SPG (FIG. 4B, lane 4-5).
  • Overexpression of S in the prefusion forms showed a major band at 180 Kda, the size commonly described for uncleaved SI 80 Kda and an additional band around 150 Kda.
  • the additional band around 150 Kda is reproducibly seen upon overexpression of uncleaved S, and most likely represents the S protein deprived of N-Glycosylation (Sun, 2020) that would occur because of overloading of the host cell machinery.
  • Example 6 Impact of SARS-CoV-2 S antigen design on neutralizing antibody responses.
  • Comparison to convalescent serum is commonly used as a benchmark to help evaluate immunogenicity and potential efficacy of Covid-19 candidate vaccines.
  • a wide spectrum of Ab responses can be observed in recovering patients, ranging from barely detectable to very high levels, likely influenced by time since infection and severity of disease.
  • a cohort of 20 sera from COVID-19 confirmed convalescent patients with moderate COVID-19 symptoms who all recovered without specific treatment intervention or hospitalization were obtained.
  • the cohort was separated into two groups of 10 samples according to high or low levels of Ab binding activity to recombinant SARS-CoV-2 S (FIG. 5A). Sera from each group were then pooled and tested for neutralizing activity (FIG. 5B).
  • Humoral responses of the various types of SARS-CoV-2 eVLPs were evaluated in C75BL/6 mice that had received 2 intraperitoneal injections at 3 week intervals (FIG. 6).
  • the first injection of unmodified S presented on eVLPs induced levels of anti-SARS-COV-2 S Ab binding titers similar to those in mice that received a recombinant trimerized prefusion S protein, but they were not associated with significant (90% or greater) neutralization activity as measured in a PRNT assay (FIG. 6A-B).
  • nAb response was induced by a single injection of eVLPs expressing prefusion SP or SPG, with PRNT90 EPTs of 80 and 160 respectively that were higher than that observed with the human convalescent control pool (PRNT90 EPT of 50). All nAb responses were greatly enhanced by the second injection and reflected the responses that were observed prior to the boosting dose. Dilutions of 1/640 and 1/2560 of the pooled sera from animals immunized with native or prefusion forms of S eVLPs, respectively, neutralized 90% of the cytopathic effect of the virus. Notably, all forms of SARS- CoV-2 S presented on eVLPs induced higher antibody titers than recombinant prefusion S protein, both in the levels of total IgG and neutralization activity, after one or two injections.
  • mice sera were performed 14 days after the second injection of eVLPs to evaluate the Ab responses against the whole S1+S2 protein or the RBD (FIG. 6C-D). All immunized mice that received eVLPs showed robust anti-SARS-Cov-2 Ab responses either against a full length S1+S2 protein (FIG. 6C) or against the RBD protein (FIG. 6D). A more homogenous response was observed in mice that received the SPG eVLPs, with all Ab EPTs above (5.6Log10) 400,000 against S, and above (5.8Logl0) 650,000 against RBD.
  • Example 7 eVLP expression of a stabilized prefusion form of the SARS-CoV-2 spike protein with E6020 elicits potent immunity after a single dose.
  • mice sera contained total anti-Spike IgG EPTs reaching geometric means of (4.8 Log 10) 54,891 with VBI-2902a and (5.2 Log10) 258,865 with VBI- 2902e that were associated with neutralizing PRNT90 titers of 365 (2.6 Log10) for VBI-2902a and 651 (2.8 Log10) for VBI-2902e.
  • VBI-2902e induced a clear enhancement in the number of IFN-g- producing T cells in response to SI peptides.
  • Evaluation of IgGl and IgG2 Ab binding titers confirmed that formulation with the TLR4 agonist E6020 resulted in an increase in IgG2 production, suggesting a Thl polarized response (FIG. 7D).
  • Example 8 Comparison of Neutralizing Antibody Responses in Monovalent and Trivalent eVLP vaccines formulated with and without E6020.
  • each of the blood samples was tested for neutralizing antibodies at 14 days after first injection and 14 days after the second vaccination.
  • human serum from patients who had previously had confimed cases of COVID-19 was also tested for neutralizing antibodies.
  • Neutralizing antibodies were tested as follows. Vero cells were seeded in 6-well plates 48 h prior to infection. Sera was heat-inactivated at 56°C for 30 min then quickly transfer on ice. Serum was diluted 1:10 with virus infection media then make serial dilutions of 1:2 for 8 subsequent dilutions. Equal volumes of diluted serum and virus (100 pfu per serum dilution) were mixed and incubated at 37°C for 1 h. No sera and no virus controls were included.
  • the results are shown in FIG. 8.
  • the trivalent eVLP containing a pre-fusion version SARS-CoV-2 spike protein formulated with alum provided similar antibody titre to the monovalent eVLP vaccine containing the same prefusion spike protein formulated with alum (relative to non-pre-fusion prototypes). These results were significantly higher than the antibody titres observice in the serum from human convalescent patients. Specifically, after one dose of the alum-only eVLP vaccines, antibody titres were 3-6X higher than human convalescent serum levels. After two doses, the antibody titres were 15-20X higher than human convalescent levels. As is shown in Fig.
  • a single dose of the E6020 adjuvanted vaccines formulated with the trivalent and monovalent eVLP SARS-CoV-2 constructs produce 10-15X higher antibody titres when compared to the titres observed in human convalescent serum.
  • Two doses of the E6020 adjuvanted eVLP vaccines produce -100 times higher antibody titres when compared to the antibody titres seen in human convalescent serum.
  • one dose of E6020 adjuvanted vaccines produced antibody titres observed after two doses of alum-only vaccines.
  • Example 9 Vaccination of Syrian golden hamsters with VBI-2902a and VBI-2902-e
  • the monovalent eVLP vaccines contained the pre-fusion, stabilized version of the SARS-CoV-2 spike protein.
  • the three components of the TriAdj per dose were 10 ⁇ g PCEP-3, 10 ⁇ g poly I:C and 20 ⁇ g IDR-1002.
  • PCEP-3 was manufactured by Idaho National Laboratory.
  • PolyTC was purchased from Invivogen (Cat# +lrl-picw; Lot# PIW-11-03).
  • IDR- 1002 was synthesized by Peptide CPC Scientific (Cat# 818360; Lot# CN-11-00590).
  • Table 6 Vaccine formulations and groupings for the vaccination of Syrian gdden hamsters
  • the study duration was 56 days. Animals were acclimatized for 7 days before immunization. One day before immunization (Day -1), under anesthesia, a temperature transponder was implanted. The vaccines were administered twice at a 3 -week interval, on Day 0, and Day 21, respectively. The vaccines were given via the intramuscular (IM) route at one side of the thighs, as specified in Table 5. The injection volume adminiserted was 100 ⁇ L. On Day 42 (3 weeks after the boosting immunization), all animals were challenged with the SARS- CoV-2 virus intranasally with 50 ⁇ L/nare, via both nares. The challenge viral dose was 1 ⁇ 105 TCID50 per animal.
  • Group B or C animals produced antibody median litres of 2.9 ⁇ 103, and Group D at a median titre of 7.1 ⁇ 102 (FIG. 10).
  • medians of antibody titres increased to 4.0 ⁇ 104 (Group B), 5.3 ⁇ 104 (Group C) and 1 ,7 ⁇ 104 (Group D).
  • Antibodies for pre-bleed were at background levels for all animals.
  • Group A animals did not increase antibody production.
  • Viral RNA levels were at the peak at 2 day post-challenge (FIG. 11). Compared to Group A (Saline control), it seems that the levels of viral RNA were lower in Group B, C and D. Viral RNA levels were similar between Group B, C and D.

Abstract

Provided herein are vaccines including coronavirus antigens and E6020, as well as methods of mitigation of coronavirus infection by administering those vaccines.

Description

VACCINES, ADJUVANTS, AND METHODS OF GENERATING AN IMMUNE
RESPONSE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of priority of United States Provisional Patent Application No. 63/005,908, filed on April 06, 2020. That application is incorporated by reference as if fully rewritten herein.
BACKGROUND
Field
[0002] Embodiments of the present disclosure relate to vaccines including adjuvants and antigens, in addition to methods of generating an immune response using those vaccines for the mitigation of infection by a coronavirus.
Background of the Related Art
[0003] Vaccines have proven to be successful methods for the mitigation of infectious diseases. Generally, they are cost effective, and do not induce antibiotic resistance to the target pathogen or affect normal flora present in the host. In many cases, such as when inducing anti-viral immunity, vaccines can prevent a disease for which there are no viable curative or ameliorative treatments available.
[0004] Vaccines function by triggering the immune system to mount a response to an agent, or antigen, typically an infectious organism or a portion thereof that is introduced into the body in a non-infectious or non-pathogenic form. Once the immune system has been “primed” or sensitized to the organism, later exposure of the immune system to this organism as an infectious pathogen results in a rapid and robust immune response that destroys the pathogen before it can multiply and infect enough cells in the host organism to cause disease symptoms. The agent, or antigen, used to prime the immune system can be the entire organism in a less infectious state, known as an attenuated organism, or in some cases, components of the organism such as carbohydrates, proteins or peptides representing various structural components of the organism. [0005] In many cases, it is useful to enhance the immune response to the antigens present in a vaccine in order to stimulate the immune system to a sufficient extent to make a vaccine effective, i.e., to confer immunity. Many protein and most peptide and carbohydrate antigens, administered alone, do not elicit a sufficient antibody response to confer immunity. Such antigens need to be presented to the immune system in such a way that they will be recognized as foreign and will elicit an immune response. To this end, additives (adjuvants) have been devised which stimulate, enhance and/or direct the immune response toward a selected antigen.
[0006] One historical example of an adjuvant, “Freund's complete adjuvant,” consists of a mixture of mycobacteria in an oil/water emulsion. Freund's adjuvant works in two ways: first, by enhancing cell and humoral-mediated immunity, and second, by blocking rapid dispersal of the antigen challenge (the “depot effect”). However, due to frequent toxic physiological and immunological reactions to this material, Freund's adjuvant cannot be used in humans.
[0007] Another immunostimulant with adjuvant activity that has been shown to have immunostimulatory or adjuvant activity is endotoxin, also known as lipopolysaccharide (LPS). LPS stimulates the immune system by triggering an “innate” immune response, a response that has evolved to enable an organism to recognize endotoxin (and the invading bacteria of which it is a component) without the need for the organism to have been previously exposed. While LPS is too toxic to be a viable adjuvant, molecules that are structurally related to endotoxin, such as monophosphoryl lipid A (“MPL”) have been tested as adjuvants in clinical trials. Both LPS and MPL have been demonstrated to be agonists to the human toll-like receptor-4 (TLR-4). One FDA-approved adjuvant for use in humans is the aluminum persulfate salt, which is used to “depot” antigens by precipitation of the antigens. Aluminum-based adjuvants have been used in human vaccines since 1932 and, although they have a long record of safety, their mode of action is not completely understood. It is generally believed that aluminum-based adjuvants enhance immune response by activation of dendritic cells. The two most frequently used aluminum- based adjuvants are referred to as “aluminum phosphate” and “aluminum hydroxide”, with aluminum hydroxide adjuvant being the most widely used commercially. Aluminum hydroxide adjuvant is not Al(OH)3, but rather crystalline aluminum oxyhydroxide (AIOOH) which has a larger surface area than crystalline aluminum hydroxide. Aluminum phosphate adjuvant is actually amorphous aluminum hydroxy phosphate (Al(OH)x(P04)y) in which some of the hydroxyl groups of aluminum hydroxide adjuvant are replaced by phosphate groups. The surface of aluminum phosphate adjuvant is composed of Al-OH and AI-OPO3 groups. Although they are chemically similar, the two adjuvants have different chemical properties. They are often both referred to simply as “alum” adjuvants.
[0008] E6020 is a potent TLR-4 receptor agonist, and thus is useful as an immunological adjuvant when co-administered with antigens in vaccines. Toll-like receptors (TLRs) belong to the family of innate immune receptors, which play an important role in the activation of innate immunity, regulation of cytokine expression, indirect activation of the adaptive immune system, and the recognition of pathogen-associated molecular patterns (PAMPs). E6020 has been reported for use in combination with antigen or vaccine components, e.g ., an antigenic agent selected from the group consisting of antigens from pathogenic and non-pathogenic organisms, viruses, and fungi. As a further example, E6020 has been reported for use as an adjuvant in combination with proteins, peptides, antigens and vaccines that are pharmacologically active for disease states and conditions including staphylococcus aureus, pertussis toxin, tetanus, influenza, Chagas disease, meningococcus, HIV, cancer, chlamydia, cytomegalovirus, Leishmaniasis, and whooping cough (caused by pertussis toxin). When used as a component in a vaccine, E6020 and the antigen are each present in an amount effective to elicit an immune response when administered to a host animal, embryo, or ovum being vaccinated therewith.
Figure imgf000004_0001
E6020
[0009] Coronaviruses are a genus in the Coronaviridae family and are pleomorphic, enveloped viruses. See S. Perlman et al, Nature Reviews Microbiology, 7:439-450 (2009). Coronaviruses contain a single stranded, 5 '-capped, positive strand RNA molecule that ranges from 26-32 kb and contain at least 6 open reading frames. Coronaviruses use host proteins as part of their replication strategies. Immune, metabolic stress, cell cycling, and other cellular pathways are activated by infection. See Tang Y. et al, Front. Immunol. 11 : 1708 (2020).
[0010] Coronaviruses usually cause mild to moderate upper-respiratory tract illnesses, like the common cold, in people. See “Coronaviruses,” National Institute of Allergy and Infectious Disease, https://www.niaid.nih.gov/diseases-conditions/coronaviruses (accessed on April 1,
2020) and Lee J. S. et al, Sci. Immunol. 5 (2020). There are hundreds of coronaviruses that affect animal species. Seven coronaviruses are known to cause human disease. Four of these coronaviruses are mild: viruses 229E, OC43, NL63 and HKU1; three of the coronaviruses can have more serious outcomes in people: SARS (severe acute respiratory syndrome), which emerged in late 2002 and disappeared by 2004; MERS (Middle East respiratory syndrome), which emerged in 2012 and remains in circulation in camels; and COVID-19, which emerged in December 2019 (a global effort is under way to contain its spread). COVID-19 is caused by the coronavirus known as SARS-CoV-2 (also known as 2019-nCoV). SARS-CoV-2 has been shown to cause mild to fatal symptoms in the human population. See Hantoushzadeh S. et al ., Arch. Med. Res. 51: 347-348 (2020) and Ingraham N. E. et al., Lancet Respir. Med. 8: 544-546 (2020). [0011] Activation of the human innate immune cells (macrophages, dendritic cells) through the binding of PAMP from SARS - Cov - 2 to cell surface TLRs has been demonstrated to be a vital mediator of COVID - 19 immunopathogenesis. Particularly in SARS - CoV - 2 infection, major immunopathological consequences leading to death have resulted from the interaction of the SARS - CoV - 2 antigens and human TLRs. Precisely, SARS - CoV - 2 viral spike protein (S) binds with the extracellular domain of various TLRs including TLR1, TLR4, and TLR6, with the strongest binding with TLR4.
[0012] While treatment of specific symptoms improves survival, there are currently only a few vaccines which have been licensed for emergency use for COVID-19 prophylaxis. Thus, a need exists for new vaccines and vaccine adjuvants that are useful in provoking an immune response against COVID-19.
BRIEF SUMMARY
[0013] Embodiments provided herein include vaccines comprising E6020 as an adjuvant and methods of generating an immune response using those vaccines for the mitigation of infection by coronavirus.
[0014] E6020 may be used as an adjuvant in vaccines directed to mitigation of nidovirales infection. Further embodiments relate to use as an adjuvant in vaccines directed to mitigation of coronavirus infection. Yet further embodiments relate to use of E6020 as an adjuvant in vaccines directed to mitigation of infection caused by the virus SAR-CoV-2 (aka COVID-19). [0015] Embodiments include, for example, a vaccine including E6020 and an antigen related to coronavirus. Yet further embodiments relate to a vaccine including a virus like particle containing a SARS-CoV-2 spike protein and E6020. [0016] Vaccines prepared according to embodiments presented herein may include one or more adjuvants in addition to E6020 to form an adjuvant system. For example, an embodiment of the invention includes a vaccine comprising a virus like particle containing a SARS-CoV-2 spike protein, E6020 and aluminum phosphate adjuvant. Vaccines may be formulated in a number of ways. For example, they may be formulated as a buffered solution, as an emulsion, as microparticles, or as nanoparticles (for example, as gene nanoparticles).
[0017] Vaccines prepared according to embodiments as reported herein may also include pharmaceutically acceptable additives. These include, for example, polymer additives and/or surfactant additives. Polymer and surfactant additives may be particularly useful, for example, in emulsion formulations.
[0018] Further embodiments provide methods of generating an immune response in a subject in whom an immune response to coronavirus is desired. Typically a subject is an unvaccinated human or a human who has undergone some but not all of the recommended number of doses of vaccine. In some embodiments, a subject may be a person already exposed or infected by a coronavirus, in whom a boosted immune response following vaccination is sought.
BRIEF DESCRIPTION OF DRAWINGS
[0019] FIG. 1 depicts the detection of SARS-CoV-2 native S eVLP vaccines formulated with different adjuvants.
[0020] FIG. 2 depicts the neutralizing antibody titers from the pooled sera of C75BL/6 mice administered a SARS-CoV-2 eVLP vaccine.
[0021] FIG. 3 depicts the antibody and T cell responses of SARS-CoV-2 native S eVLP vaccines combined with different adjuvants in C75BL/6 mice.
[0022] FIG. 4 depicts the structures of SARS-CoV-2 S protein constructs and a Western blot analysis of the protein expression for each SARS-CoV-2 S protein construct.
[0023] FIG. 5 depicts the antibody titers in the sera from 20 COVID-19 confirmed convalescent patients, wherein the sera samples were separated into groups with either high or low levels of Ab binding activity to recombinant SARS-CoV-2 S.
[0024] FIG. 6 depicts humoral responses from various types of SARS-CoV-2 eVLP vaccines in C75BL/6 mice.
[0025] FIG. 7 depicts the antibody and T cell responses of different monovalent constructs of SARS-CoV-2 SPG vaccines in C75BL/6 mice.
[0026] FIG. 8 depicts the neutralizing antibody response in mice vaccinated with VBI-2902 and VBI-2901 with and without E6020. [0027] FIG. 9 depicts the body weight changes of Syrian golden hamsters vaccinated with VBI- 2902 vaccines with and without E6020.
[0028] FIG 10 depicts the serum antibody titers of Syrian golden hamsters vaccinated with VBI- 2902 vaccines with and without E6020.
[0029] FIG. 11 depicts the viral RNA levels of Syrian golden hamsters vaccinated with VBI-
2902 vaccines with and without E6020.
DETAILED DESCRIPTION
[0030] Vaccines including E6020 and methods for treatment of coronavirus infection using those vaccines will be described in detail. E6020 is present in these vaccines as an adjuvant, which is a compound that is included in a vaccine to increase the vaccine’s ability to provoke, increase, and/or extend an immune response, or drive a favorable type of immune mechanism(s).
[0031] E6020
[0032] E6020 is a di sodium salt of ER-804057 and is shown below:
Figure imgf000007_0001
E6020
[0033] In some embodiments, the vaccines described herein include from 0.1 μg to 100 μgs, 0.5 μg to 100 μgs, 1 μg to 50 μg, 1 μg to 25 μg, 1 μg to 20 μg, 5 μg to 30 μg, 0.5 μg to 10 μg, 10 μg to 20 μg or 20 μg to 50 μg of E6020. In other embodiments, the vaccine comprises 10 μgs of E6Q20.
[0034] Antigens for Inclusion [0035] Antigens are molecules that may be recognized by the immune system of a patient to generate an immune response and/or cell-mediated immunity. In embodiments reported herein, E6020 is useful as an adjuvant in a vaccine where the antigen is a nidovirales antigen, a coronavirus antigen, or a SARS-CoV-2 antigen. Antigens may be present in a vaccine in various forms. For example, an antigen may be present as purified antigen molecules (which may be, for example, proteins, multimerized proteins, protein subunits (including subunit trimers), peptides (including Ii-key peptides and locked peptides), peptides conjugated to a protein carrier, oligonucleotides, RNA (including mRNA), DNA, plasmid DNA, or polysaccharides, conjugated to a carrier or not), on live-attenuated, recombinant or inactivated-whole viruses, as dendritic cells, as antigen presenting cell vectors, as recombinant viral vectors, as adenoviral vectors, through liposomal delivery vehicles including lipoproteins or lipopolyplexes, or by a composition that includes a nucleic acid that encodes antigen.
[0036] In some embodiments, the antigen is derived from a viral or bacterial pathogen, such as an influenza or corona virus.
[0037] Vaccines described herein may comprise antigens, wherein the antigens are presented on an enveloped virus like particle (“eVLP”). eVLPs may comprise retroviral vectors that lack a retrovirus-derived genome and are therefore non-replicating. Retroviruses are enveloped RNA viruses that belong to the family Retroviridae. After infection of a host cell by a retrovirus, RNA is transcribed into DNA via the enzyme reverse transcriptase. DNA is then incorporated into the host cell’s genome by an integrase enzyme and thereafter replicates as part of the host cell’s DNA. eVLPs may be comprised of a structural polyprotein from a Moloney Murine Leukemia Virus (MMLV) known as a Gag protein (SEQ ID NO: 5). Expression of Gag in some host cells can result in self-assembly of the expression product into eVLPs.
[0038] The antigen may be derived from a viral or bacterial pathogen, such as an influenza virus or a corona-virus. In some embodiments, the antigen is derived from a Nidovirales virus.
[0039] In some embodiments, the antigen present within the vaccine resembles a SARS-CoV-2 spike (S) protein. The SARS-CoV-2 spike (S) protein plays a crucial role in host cell receptor binding and fusion properties leading to virus entry. See Patra R. et al., J Med Virol. 93:615-617
(2021); Aboudounya M. M. et al., Mediators Inflamm. 2021: ID 8874339; Bhattacharya, M. et al., Infect. Genet. Evol. 85: 104587 (2020). SARS-CoV-2 S proteins resemble features of class I viral proteins, wherein they are constituted of 2 subunits, SI, containing the receptor binding domain (RBD) and S2, containing the fusion entry domain. Binding of the RBD to the host cell receptor induces conformational changes resulting in activation of the protease cleavage site upstream of the fusion domain followed by release and activation of the S2 fusogenic domain.
These conformational changes allow SARS-CoV-2 to infiltrate a cell and begin replication. See Khanmohammadi S. et al., J Med Virol. 1-5 (2021). SARS-CoV-2 S protein contains a furin cleavage site located at the boundary of SI and S2 enabling rapid processing of the S protein during biosynthesis in host cells.
[0040] The antigen present within the vaccine can be a native SARS-CoV-2 S protein, a stabilized prefusion form of a SARS-CoV-2 S protein, a modified SARS-CoV-2 S wherein the TMCTD domain has been replaced with a transmembrane and cytoplasmic terminal domain of vesicular stomatitis virus G (VSV-G) or any combination thereof.
[0041] The SARS-CoV-2 spike antigen present within the vaccine may possess, for example, a protein sequence provided by SEQ ID NOs: 1-4.
[0042] In some embodiments, the vaccine may comprise from 0.1 μg to 100 μg, 0.1 μg to 50 μg, 1 μg to 25 μg, 5 μg to 25 μg or 5 μg to 10 μg of an antigen.
[0043] Vaccines That May Benefit from an E6020 Adjuvant
[0044] A number of existing or in development vaccine platforms could benefit from inclusion of E6020 as an adjuvant. These include, for example, but are not limited to those listed in Table 1
Table 1
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
[0045] Other Adjuvants for Use in Adjuvant Systems with E6020
[0046] In some embodiments, the vaccine may comprise additional adjuvants in combination with E6020. Non-limiting examples of other adjuvants that may be useful in combination with E6020 include aluminum-based adjuvants such as aluminum hydroxide adjuvant (crystalline aluminum oxyhydroxide (AIOOH)) or aluminum phosphate adjuvant (amorphous aluminum hydroxy phosphate (for example Adju-Phos®)), an aluminum salt, chemokines, cytokines, nucleic acid sequences (particularly bacterial nucleic acid systems), lipoprotein, lipopolysaccharide (LPS), monophosphoryl lipid A, lipoteichoic acid, imiquimod, reiquimod, QS-21 or any combination thereof. Other TLR agonists may be useful in adjuvant systems with E6020. For example, other adjuvants may be agonists that activate TLR 2, 3, 5, 7, 8, 9 or a combination thereof. Use of E6020 in combination with other adjuvants may include simultaneous or sequential administration of the adjuvants in the system.
[0047] In some embodiments the vaccine further comprises from 50 μg to 50 mg, 0.1 mg to 20 mg, 1 mg to 5 mg, 3 mg to 5 mg or 0.1 mg to 1 mg of at least one additional adjuvant. In other embodiments, the vaccine further comprises 150 μg to 180 μg, such as 165 μg, of at least one additional adjuvant.
[0048] In other embodiments, the vaccine further comprises from 0.1 mg/mL to 1 mg/mL, from 0.05 mg/mL to 0.9 mg/mL, 0.5 mg/mL to 1.0 mg/mL, 0.1 mg/mL to 0.5 mg/mL, 0.1 mg/mL to 5.0 mg/mL or 0.165 mg/mL to 0.33 mg/mL of at least one additional adjuvant. In other embodiments, the vaccine further comprises 0.165 mg/mL or 0.33 mg/mL of at least one additional adjuvant. The vaccine may also comprise up to 800 mg/mL of at least one additional adjuvant. In some embodiments, the amount of the at least one additional adjuvant is based on the aluminum content within the at least one adjuvant. [0049] Inventors of the present disclosure previously described monovalent and multivalent eVLP SARS-CoV-2 vaccines formulated with alum (US 17/218148) which induced a strong immune response against the SARS-CoV-2 spike protein in mice studies. These vaccines were shown to be strongly protective against COVID-19 infection in Golden Hamsters challenged with SARS-CoV-2. As described in US 17/218148, the eVLP SARS-CoV-2 vaccines can be formulated with a variety of different adjuvants, several of which (including E6020) were tested in mice as described in Example 4. As is shown in Example 4, the use of E6020 as an additional adjuvant to the eVLP/alum formulation resulted in a significantly stronger immune response than the use of other additional adjuvants. Furthermore, and quite surprisingly, the use of E6020 as an additional adjuvant significantly enhanced Thl-type T cell responses and a change of IgG profile with induction of significant higher IgG2 responses than seen in the formulation with the eVLP SARS-CoV-2 vaccine with alum alone or with other additional adjuvants such as mimics of AS03 and AS04. This enhanced Thl response represents a shift in immune response toward a Thl from Th2. The Thl response is correlated with immunity against viral infection. Accordingly, the unexpected and significant enhancement of the Thl response induced by the combination of the eVLP SARS-CoV-2 vaccine formulated with alum and E6020 indicates that this combination is a highly potent immunogenic composition against COVID-19.
[0050] Formulations and Modes of Administration
[0051] Vaccines including E6020 may be delivered using multiple modes of administration and multiple formulations. In addition to including coronavirus antigen and E6020, formulations typically include at least one pharmaceutically acceptable carrier.
[0052] A carrier may be, for example, a diluent, excipient, or vehicle used in administration of the E6020. Carriers may include, for example, water or saline.
[0053] Methods of administration include, but are not limited to, parenteral, e.g., intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal and intrathecal. Administration can also occur systemically.
[0054] Vaccines may be formulated for parenteral administration by injection. This may include, for example, bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers. They may include a preservative. Injectable dosage forms include suspensions, solutions or emulsions. They may be in oily or aqueous vehicles. They may contain further additives including suspending, stabilizing and/or dispersing agents. The vaccine may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen free water, before use.
[0055] Administration by inhalation is also possible. Vaccines may be delivered, for example, as an aerosol spray presentation from pressurized packs or a nebulizer. A propellant is also used. A suitable propellant may be dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other gas. A nasal spray, which is not pressurized or which is pressurized mechanically rather than chemically, can be used for intranasal administration. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges may be made for use in an inhaler or insufflater. These would contain a powder mix of the compound and a suitable powder base such as lactose or starch.
[0056] Embodiments may be delivered using virus-like particles (VLPs), microparticles or nanoparticles. For example a nanoparticle may include a peptide nucleic acid oligomer conjugated to a lipid. The oligomer complexes with an antigen and/or an adjuvant, forming a nanoparticle for delivery according to one of the methods reported herein. Particle-based delivery, including microparticle or nanoparticle based delivery, may be, for example, protein- based scaffolds or matrices, lipid-based scaffolds or matrices, or polymer-based scaffolds or matrices.
[0057] In some embodiments, E6020 is formulated with enveloped virus-like particles (eVLPs) that possess a stable core of Gag protein and a lipid bilayer. eVLPs structurally resemble viruses but are much safer to administer because they lack the genetic material needed to replicate within the host. eVLPs enable repeating, array-like presentation of antigens which is a preferred means of activating B cells and eliciting high affinity antibodies. eVLPs that can be used as vaccines can be, but are not limited to, MLV-Gag eVLPs and those disclosed in US Patent 9,765,304 and US Patent Application No. 17/218,148.
[0058] In some embodiments, the vaccine comprises eVLP particles comprising 0.05 mgs to 0.50 mgs, 1 μg to 50 mg, 10 μg to 10 mg, 50 μg to 5 mg or 100 μg to 500 μg of Gag protein.
[0059] In other embodiments, the vaccine comprises eVLP particles comprising an amount of antigen and an amount of Gag protein, wherein the amount of antigen is from 0.1 % to 4.0 % relative to the amount of Gag protein.
[0060] In some embodiments, the vaccine comprises eVLP particles comprising a of murine leukemia virus (MMLV) Gag protein. In other embodiments, the Gag protein is a MMLV-Gag protein according to SEQ ID NO: 5.
[0061] A typical treatment regimen includes administration of an amount effective or likely to lead to mitigation of an infectious agent over a period of time. That may range from a few hours to a few days, or to a few months.
[0062] Mitigation may include prevention of a disease, minimization of symptoms, or alleviation of symptoms. An effective treatment may provide mitigation for a lifetime, for decades, for years, for months, or for even a single month. The amount of mitigation may increase, decrease, or both over the time that the mitigation is in effect.
[0063] Other Additives
[0064] Other non-antigen, non-adjuvant, non-carrier compounds may be administered in or in conjunction with vaccines reported herein. For example, vaccines may include or may be administered with a separate antiviral agent, an antibacterial agent, antifungal agent, or other antibiotic. These may include, for example, oseltamivir, azithromycin, chloroqine, hydroxychloroquine, or zanamivir.
[0065] Vaccine Doses
[0066] The vaccines described herein may be administered in doses ranging from 0.1 mL to 10 mL, 0.2 mL to 5 mL or 0.3 mL to 3 mL.
[0067] Examples
[0068] Example 1 : An experiment to demonstrate adjuvant activity would examine in vivo mouse immune response, comparing the strength and characteristics of response to antigen administered with or without the candidate adjuvant.
[0069] Example l is a prophetic example. In this example mice are immunized with a SARS-
CoV-2 antigen, in vaccine formulations with or without E6020, in order to assess the effect of E6020 on the response to immunization. Antigens may be chosen for their potential to raise protective antibodies, cytotoxic T cell responses, or both (Grifoni A, Sidney J, Zhang Y, Scheuermann RH, Peters B, Sette A. Cell Host Microbe. 2020 Mar 12. pii: S1931- 3128(20)30166-9. doi: 10.1016/j.chom.2020.03.002.). In this example the SARS-CoV-2 antigen is recombinantly produced S protein. The S protein, or the receptor-binding domain of the S protein, can be produced by introduction of an appropriate vector into HEK cells. In this example the receptor-binding domain of the S protein, S331-524 (Tai etal ., Cellular and Molecular Immunology; https://doi.org/10.1038/s41423-020-0400-4) is expressed and fused to a carrier such as the human antibody Fc to allow secretion and purification. Other approaches might include expression of full-length spike ectodomain or other subdomains of the S protein (Wang etal ., https://doi.org/10.1101/2020.03.ll.987958 doi: bioRxiv preprint). Alternatively an inactivated whole virus preparation might be used as antigen.
[0070] S protein sequences may be produced as monomeric subunits, or produced as fused sequences containing multimerization sequences, purification tags or sequences that allow appropriate presentation of antigenic epitopes. Various linkers or carriers of the antigenic domain can be utilized. In this example the Fc fusion is produced by inserting an appropriate S protein sequence into the pFUSE-hIgGl-Fc2 expression vector (InvivoGen, San Diego, CA) and expressing the resultant fusion protein by transfecting the vector into the human HEK-293 cell line. After transfection, secreted protein is recovered from the cell culture supernatant and isolated by an affinity method that selects for either the S protein (as for example with a specific anti-spike protein antibody) or the Fc sequence (e.g. using a protein A column) (ref. Tai et al. as above).
[0071] To raise a protective antibody response, groups of six to eight BALB/c mice are immunized subcutaneously with between 10 and 100 micrograms of SARS-CoV-2 S protein. Three immunizations are given at three week intervals. SARS-CoV-2 antigen is given in PBS, as an unadjuvanted control, or with a test adjuvant. E6020 is tested at doses known to augment antibody responses to other antigens, for example 1.0, 3.0 or 10 micrograms. Other adjuvants such as alum at 2.7 mg/dose, or appropriate doses of other commercially available adjuvant substances, are included as positive controls.
[0072] In all cases, responses by the groups receiving E6020 or alum or other commercially available adjuvant substances are compared to unadjuvanted groups. Control groups receiving adjuvants or carrier alone are included to confirm antigen dependence of any observed response. Similar experiments, with volumes of administration adjusted appropriately, are done looking at other routes of administration such as intranasal or intradermal. Antibody titers or neutralization titers are measured in blood, and on mucosal surfaces such as the lung or vaginal lining.
[0073] Blood samples are taken from immunized animals at two weeks after the second and/or the third immunization. Mucosal samples are taken by appropriate lavage methods. Serum or lavage are isolated and tested for anti-antigen titers of immunoglobulin using standard ELISA methods. For example, an S protein construct might be expressed that includes the sequences used to immunize, and used to coat an ELISA plate. It is important that the ELISA antigen construct not contain carrier protein sequences that are the same as those used to immunize the mice, in order to avoid measuring spurious reactivities to carrier protein that are unrelated to anti-virus responses. Examples of ELISA methods for SARS-CoV-2 S protein are given in (Nisreen M.A. Okba, Marcel A. Miiller, Wentao Li, et al., medRxivhttps://doi.org/10.1101/2020.03.18.20038059) and include potential use of commercial ELISA kits. In all cases the standard ELISA procedures of coating plates with the relevant antigen, blocking the plate surface, reacting the coated plate with test serum or lavage and developing with a labeled anti-mouse immunoglobulin antibody are used. Raw ODs are plotted, or titers derived by appropriate methods.
[0074] An effective adjuvant is expected to increase the amount of anti-antigen antibody raised, cause titers to rise earlier or persist longer, or cause a shift in isotype as compared to antigen given alone. Changes in isotype response reflect differences in the cytokine patterns elicited by the adjuvant, which in turn is associated with effective protection against different types of infection. For example, IgG2a in mice is associated with an interferon-driven Thl response that might support cytotoxic T cells and give particularly efficient anti-viral responses.
[0075] Example 2: An experiment to demonstrate adjuvant-enhanced elicitation of neutralizing antibodies against the coronavirus, comparing the strength and characteristics of virus neutralizing response to antigen administered with or without E6020.
[0076] Example 2 is a prophetic example. Because ELISA titers may not correlate with the ability of serum antibodies to effectively protect against infection, it is helpful to measure neutralization titers elicited by immunization. In this example, mice are immunized as in Example 1, one or more times. After immunization, serum or lavage are taken as described, and are used in neutralization assays. A neutralization assay is performed by measuring the infectious potential of a pseudotyped virus which carries SARS-CoV-2 surface proteins but genetic material derived from a crippled HIV construct that cannot replicate but does express a tracking marker such as luciferase. When applied to human ACE2-expressing HEK293 cells the SARS-CoV-2 receptor binding domain allows infection and expression of luciferase by the cells. If the pseudotyped virus is exposed to neutralizing antibodies the infection is blocked and luciferase is not expressed. Such an assay is described in (Tai et al, Cellular and Molecular Immunology; https://doi.org/10.1038/s41423-020-0400-4). Specifically, pseudotyped virus is harvested from HEK293 cells cotransfected with a plasmid encoding Env-defective, luciferase- expressing HIV-1 (pNL4-3.1uc.RE) and another encoding SARS-CoV-2 S protein, and pseudovirus-containing supernatants harvested subsequently.
[0077] Neutralization is assessed by incubation of pseudovirus with serially diluted mouse sera from vaccinated mice for 1 h at 37 °C, followed by addition of the mixture into hACE2- expressing HEK293 cells. After an appropriate incubation the cells are lysed and the resulting supernatant is mixed with luciferase substrate and tested for relative luciferase activity, typically using a luminometer to measure light output. This assay measures production of spike-specific antibodies capable of inhibiting viral interaction with cells.
[0078] In another approach, neutralization by antibody binding to other viral proteins (as might be induced by a whole inactivated viral vaccine) is measured by a classic plaque assay in which dilutions of infectious SARS-CoV-2 are preincubated with test sera before application to a confluent culture of Vero cells. Appearance of plaques, where cells have been lysed by viral replication, will occur in the absence of neutralizing antibody. The reduction of plaque frequency in the presence of neutralization is quantified by standard means (Okba et al., medRxivhttps://doi.org/10.1101 /2020.03.18.20038059). Infectious centers may also be detected and enumerated after staining the Vero culture with enzyme-tagged anti -viral specific antibody reagents. [0079] Example 3: An experiment to demonstrate adjuvant-enhanced elicitation of neutralizing antibodies against the coronavirus, using an in vivo challenge of mice or another species in a viral infection model. Groups receiving vaccine with or without E6020 will be compared for their resistance to infection.
[0080] Example 3 is a prophetic example. To confirm authentic protective efficacy of a vaccine in the context of an in vivo infection, animals are immunized with a SARS-CoV-2 vaccine as described above and then exposed to live infectious virus in a challenge model. The animals are mice that have been engineered to allow infection by SARS-CoV-2, for example through expression of the human ACE2 receptor protein in appropriate tissues (McCray et al. JOURNAL OF VIROLOGY, Jan. 2007, p. 813-821). Alternatively, animal species that are susceptible to infection with SARS-CoV-2 even in the absence of transgene introduction, such as ferrets or cats (Shi et al. bioRxiv preprint doi: https://doi.org/10.1101/2020.03.30.015347), are immunized with vaccine. After immunization, animals are challenged by an appropriate route, e.g. intranasally, with a dose of SARS-CoV-2 that is known to produce infection and replication. Immunization groups are compared by measuring physical symptoms (for example body temperature, oxygenation or mortality), or alternatively suppression of in vivo viral replication by the vaccine- induced immune response may be assessed by measuring viral titers in target tissues such as the lungs. This is performed by PCR, measurement of viral load in tissue homogenates (Stadler et al., Emerging Infectious Diseases · www.cdc.gov/eid · Vol. 11, No. 8, August 2005, p. 1312), or other measures such as immuno-staining for viral antigens in tissue sections. A decreased presence of virus in animals receiving vaccine adjuvanted with E6020 indicates the superior protective effect of this adjuvant.
[0081] Example 4: Influence of different adjuvants on antibody and T cell responses induced by monovalent SARS-CoV-2 eVLP vaccines.
[0082] Direct correlations between neutralizing antibody titers and T cell activity have been observed among COVID-19 patients (Ni, Immunity, 2020), suggesting that strong T cell inducing adjuvants may improve protection against SARS-CoV-2 infection. After SARS-Cov-2 infection, a preferential Thl-type response has been observed in acute and recovering patients
(Weiskopf, 2020; Griffoni, Cell 2020), associated with higher levels of IgGl Ab against the S protein (Ateyo, Immunity 2020). In contrast, a Th2-type response has been suggested to contribute to the “cytokine storm” associated with severe lung pathologies (Peeples, PNAS 2020;
Roncati, 2020). In light of these results, a variety of adjuvants and adjuvant combinations were tested in combination with a native SARS-CoV-2 S eVLP vaccine for their ability to enhance neutralizing Ab production while also promoting a Thl-type response. The native SARS-CoV-2
S eVLP vaccines express the native S protein of SARS-CoV-2 (SEQ ID NO: 1). For this purpose, mimics of MF59, the adjuvant systems AS03 and AS04, and E6020 co-formulated with aluminum phosphate (Adju-Phos) were evaluated. The formulations of the native SARS-CoV-2 S eVLP vaccines with the various adjuvants are provided in Table 1.
Table 1: Native SARS-CoV-2 S eVLP vaccine formulations for each sample group
Figure imgf000020_0001
* = proposed mouse doses of adjuvant constituents, human doses are approximately 4 to 5 times higher.
[0083] SARS-CoV-2 native S eVLPs were used to compare the effects of the adjuvants as they were less immunogenic than eVLPs expressing the prefusion form of the S protein and might better enable differences in the adjuvants to be observed. Mice received 2 IP injections of the native SARS-CoV-2 eVLPs formulated with the various adjuvants. The IgG binding titers, neutralizing antibody titers and Ab and T cell responses was assessed 14 days after the second injection.
[0084] Western blot analysis of native SARS-CoV-2 eVLPs vaccines (FIG. 1) was performed using a primary antibody that is a SARS-CoV-2 (2019-nCoV) Spike RBD Rabbit Antibody (Cat# 40592-T62 Sinobiological) (1/5,000 dilution). The secondary antibody is a goat anti-rabbit IgG-Fc HRP-conjugated (Bethyl, Cat# A120-11 IP- 18) lmg/mL - 1/10,000 Precision Protein Streptactin HRP conjugate (Bio-Rad, cat # 161-0381) (1/10,000 dilution). TA 1 is the AS03 vaccine (see Group 1 in Table 1), TA 2 is the E6020 + aluminum phosphate adjuvant vaccine (see Group 2 is Table 1), TA 3 is the AS04 modified vaccine (see Group 3 in Table 1), TA 4 is the AS04 vaccine (see Group 4 in Table 1), TA 5 is the ASOIB vaccine (see Group 5 of Table 1), TA 6 is the MF59 vaccine (see Group 6 of Table 1), the control lane is a Native Monovalent SARS-CoV-2 vaccine from 29CH07- Post TFF/UC Pellet Filtered BDS (Lot#V20200501- nCOVID), Lane 9 is 0.025 μg of a recombinant SARS-CoV-2 (Srn T2 #DL2020APR30 0.46/mL) vaccine, and Lane 10 is 3.65 μg of 19CH102-Psot TFF/UC Pellet Filtered BDS (V20200409-EG).
[0085] The IgG binding titers of the vaccine constructs are presented in Table 2 below.
Table 2: IgG Binding Titers of the Native SARS-CoV-2 S eVLP vaccines
Figure imgf000021_0001
Figure imgf000022_0001
[0086] The virus-neutralizing titers of the vaccine constructs are presented in Table 3 below.
Table 3: Native SARS-CoV-2 S eVLP vaccine virus-neutralizing titers
Figure imgf000022_0002
[0087] The neutralizing anti-body titers from the pooled sera of the treatment groups are depicted in FIG. 2. The neutralizing anti-body titers at P1VD14 and at P2VD14 were measured. The neutralizing anti-body titers of Group 2 were similar to those of Group 1 and higher than those of Groups 3-4. The data presented within FIG. 2 demonstrates that the use of E6020 as an additional adjuvant significantly improved the effectiveness of the native monovalent SARS- CoV-2 vaccine formulated with the aluminum phosphate adjuvant.
[0088] MF59 enhanced Thl-type T cell responses compared to alum (FIG. 3A) but induced similar Ab responses (FIG. 3B-C) and comparable, balanced IgG2/IgGl ratio (FIG. 3D). In contrast, the addition of E6020 to the eVLP+aluminum phosphate adjuvant significantly enhanced Thl-type T cell responses and a change of IgG profile with induction of significant higher IgG2 than with Alum alone, demonstrating Thl polarization of both T cell and antibody responses. Mimics of AS03 and AS04 adjuvants also biased responses towards a Thl-type response, though to a lesser extent than E6020.
[0089] Example 5: Contruction of SARS-CoV-2 eVLP antigens and vaccines
[0090] Four constructs were designed based on the S protein sequence of the SARS-CoV-2 Wuhan-Hul isolate and subcloned into expression plasmids for the production of eVLPs (FIG.
4) as described in US Patent Application No. 17/218,148. One of the constructs expressed the native form of the SARS-CoV-2 S protein (S, SEQ ID NO: 1). Briefly, to obtain a stabilized prefusion form of the S protein (SP, SEQ ID NO: 2), the furin cleavage site of S was inhibited by mutation of the RRAR into GSAS and 2 proline substitutions were introduced at successive residues K986 and V987. Previous work has demonstrated that the swap of the transmembrane and cytoplasmic terminal domains (TM-CTD) of CMV gB resulted in enhanced yields and immunogenicity of the gB glycoprotein presented on eVLPs. Based on this data, two additional constructs, Native-VSVg (SG, SEQ ID NO: 3) and Stabilized Prefusion-VSVg (SPG, SEQ ID NO: 4) were designed by swapping the TM-CTD of S with that of VSV-G (FIG. 4A).
[0091] Western blot analysis of eVLPs using a polyclonal Ab directed against the SARS-CoV-2 S receptor biding domain confirmed the processing of SARS-CoV-2 S during biosynthesis in HEK293 cells as expected by the presence of the furin cleavage site in S1/S2 (Walls, 2020)
(FIG. 4B, lane 2-3). Expression of S was slighly improved by the VSV-G swap in SG, and more dramatically enhanced by the inhibition of the cleavage sites in SP and SPG (FIG. 4B, lane 4-5). Overexpression of S in the prefusion forms showed a major band at 180 Kda, the size commonly described for uncleaved SI 80 Kda and an additional band around 150 Kda. The additional band around 150 Kda is reproducibly seen upon overexpression of uncleaved S, and most likely represents the S protein deprived of N-Glycosylation (Sun, 2020) that would occur because of overloading of the host cell machinery. [0092] Quantitative analysis of protein content in eVLP preparations showed that for a similar numbers of particles, corresponding to comparable amounts of Gag protein, the amount of SARS-CoV-2 S protein was increased substantially with replacement of the TM-CTD and by use of the stabilized prefusion construct, suggesting that the density of the S protein was enhanced using the VSV-G constructs (see Table 4 below). The best yield was reproducibly obtained when producing the eVLP expressing the prefusion-VSV-G form of S, with an increase up to 40-fold more than in eVLPs expressing native S.
Table 4: Quantitative analysis of SARS-CoV-2 S protein content in eVLP vaccine constructs
Figure imgf000024_0001
[0093] Example 6: Impact of SARS-CoV-2 S antigen design on neutralizing antibody responses.
[0094] Comparison to convalescent serum is commonly used as a benchmark to help evaluate immunogenicity and potential efficacy of Covid-19 candidate vaccines. However, a wide spectrum of Ab responses can be observed in recovering patients, ranging from barely detectable to very high levels, likely influenced by time since infection and severity of disease. To enable comparison across experiments, a cohort of 20 sera from COVID-19 confirmed convalescent patients with moderate COVID-19 symptoms who all recovered without specific treatment intervention or hospitalization were obtained. The cohort was separated into two groups of 10 samples according to high or low levels of Ab binding activity to recombinant SARS-CoV-2 S (FIG. 5A). Sera from each group were then pooled and tested for neutralizing activity (FIG. 5B). As expected, the pool showing higher levels of IgG titers against SARS-CoV-2 S had the highest neutralizing activity, which was consistent with previous observations (Ni, Immunity 2020). To provide a robust benchmark with which to assess the immunogenicity of the vaccine candidates, only the high titer pooled sera was used to assess vaccine-induced animal sera.
[0095] Humoral responses of the various types of SARS-CoV-2 eVLPs were evaluated in C75BL/6 mice that had received 2 intraperitoneal injections at 3 week intervals (FIG. 6). The first injection of unmodified S presented on eVLPs induced levels of anti-SARS-COV-2 S Ab binding titers similar to those in mice that received a recombinant trimerized prefusion S protein, but they were not associated with significant (90% or greater) neutralization activity as measured in a PRNT assay (FIG. 6A-B). In contrast, a significant nAb response was induced by a single injection of eVLPs expressing prefusion SP or SPG, with PRNT90 EPTs of 80 and 160 respectively that were higher than that observed with the human convalescent control pool (PRNT90 EPT of 50). All nAb responses were greatly enhanced by the second injection and reflected the responses that were observed prior to the boosting dose. Dilutions of 1/640 and 1/2560 of the pooled sera from animals immunized with native or prefusion forms of S eVLPs, respectively, neutralized 90% of the cytopathic effect of the virus. Notably, all forms of SARS- CoV-2 S presented on eVLPs induced higher antibody titers than recombinant prefusion S protein, both in the levels of total IgG and neutralization activity, after one or two injections.
[0096] Analysis of individual mice sera was performed 14 days after the second injection of eVLPs to evaluate the Ab responses against the whole S1+S2 protein or the RBD (FIG. 6C-D). All immunized mice that received eVLPs showed robust anti-SARS-Cov-2 Ab responses either against a full length S1+S2 protein (FIG. 6C) or against the RBD protein (FIG. 6D). A more homogenous response was observed in mice that received the SPG eVLPs, with all Ab EPTs above (5.6Log10) 400,000 against S, and above (5.8Logl0) 650,000 against RBD.
[0097] Example 7: eVLP expression of a stabilized prefusion form of the SARS-CoV-2 spike protein with E6020 elicits potent immunity after a single dose.
[0098] To assess the immunogenicity and potential efficacy of the vaccines disclosed herein, two embodiments possessing eVLPs expressing the stabilized prefusion VSV-G form of S (SPG from FIG. 4) were chosen because of their significantly higher production yields and greater potency. One vaccine was formulated with aluminum phosphate adjuvant (“alum”) alone, due to its extensive safety record in several approved prophylactic vaccines (“VBI-2902a”), and the other vaccine was formulated with aluminum phosphate adjuvant and E6020 (“VBI-2902-e”). Fourteen days after a single injection, mouse sera contained total anti-Spike IgG EPTs reaching geometric means of (4.8 Log 10) 54,891 with VBI-2902a and (5.2 Log10) 258,865 with VBI- 2902e that were associated with neutralizing PRNT90 titers of 365 (2.6 Log10) for VBI-2902a and 651 (2.8 Log10) for VBI-2902e. A second injection boosted Ab binding titers to 228,374 (5.4 Log10) and 1,400,285 (6.2 Log10), respectively, for VBI-2902a and VBI-2902e and nAb titers to 1,079 (3.0 Log10) and 5,178 (3.7 Log10) (FIG. 7A-B).
[0099] Ex vivo stimulation of splenocytes (FIG. 7C) demonstrated preferential T cell response against the SI domain of the spike protein rather than the S2 domain. After the first injection, no differences were observed between groups that were adjuvanted with alum or with E6020+alum.
The second injection of VBI-2902e induced a clear enhancement in the number of IFN-g- producing T cells in response to SI peptides. Evaluation of IgGl and IgG2 Ab binding titers confirmed that formulation with the TLR4 agonist E6020 resulted in an increase in IgG2 production, suggesting a Thl polarized response (FIG. 7D).
[0100] Example 8: Comparison of Neutralizing Antibody Responses in Monovalent and Trivalent eVLP vaccines formulated with and without E6020.
[0101] A preclinical study was conducted to evaluate the efficacy of monovalent SARS-CoV-2 vs trivalent (eVLP constructs, with Adjuphos® or Adjuphos® and E6020 adjuvanted vaccine candidates in C57BL/6 mice. Both the trivalent and the monovalent eVLP constructs were the pre-fusion stabilized form described above. Mice were randomly assigned to 4 experimental groups as shown in Table 5 below and immunized intraperitoneally on week 0 (day 0) and week 3 (day 21) with 0.5 mL vaccine. Blood was collected on day 14 after the 1st and 2nd immunization.
Table 5: Vaccine formulations administered to each of the study groups
Figure imgf000026_0001
[0102] Each of the blood samples was tested for neutralizing antibodies at 14 days after first injection and 14 days after the second vaccination. As well, human serum from patients who had previously had confimed cases of COVID-19 was also tested for neutralizing antibodies. Neutralizing antibodies were tested as follows. Vero cells were seeded in 6-well plates 48 h prior to infection. Sera was heat-inactivated at 56°C for 30 min then quickly transfer on ice. Serum was diluted 1:10 with virus infection media then make serial dilutions of 1:2 for 8 subsequent dilutions. Equal volumes of diluted serum and virus (100 pfu per serum dilution) were mixed and incubated at 37°C for 1 h. No sera and no virus controls were included. Cells were washed with PBS and each virus/serum were transferred and mixed to each well containing cells, and incubated at 37°C for 1 h, with interval rocking of the plates. After the 1 h adsorption, excess inoculum was removed and a 2 ml virus infection media/agarose mix were overlaid onto the cells. The overlay was allowed to solidify and plates were incubated at 37°C for 72 h. Cells were stained with crystal violet at 72 h post-infection. Plaques were quantified for all dilutions and PRNT titre was calculated. The % plaque reduction for all the dilutions based on the no serum control, was calculate using the Reed-Muench formula to determine the PRNT titre 90.
[0103] The results are shown in FIG. 8. The trivalent eVLP containing a pre-fusion version SARS-CoV-2 spike protein formulated with alum provided similar antibody titre to the monovalent eVLP vaccine containing the same prefusion spike protein formulated with alum (relative to non-pre-fusion prototypes). These results were significantly higher than the antibody titres observice in the serum from human convalescent patients. Specifically, after one dose of the alum-only eVLP vaccines, antibody titres were 3-6X higher than human convalescent serum levels. After two doses, the antibody titres were 15-20X higher than human convalescent levels. As is shown in Fig. 8, a single dose of the E6020 adjuvanted vaccines formulated with the trivalent and monovalent eVLP SARS-CoV-2 constructs produce 10-15X higher antibody titres when compared to the titres observed in human convalescent serum. Two doses of the E6020 adjuvanted eVLP vaccines produce -100 times higher antibody titres when compared to the antibody titres seen in human convalescent serum. Thus, one dose of E6020 adjuvanted vaccines produced antibody titres observed after two doses of alum-only vaccines.
[0104] Example 9: Vaccination of Syrian golden hamsters with VBI-2902a and VBI-2902-e
[0105] Forty-eight (48) Syrian golden hamsters (males, aged approximately 5-6 weeks old) were purchased from Charles River Laboratories and were randomly assigned to Groups A, B, C and D (n=12/group, see Table 6). The randomly assigned group were administered with A. VBI- 2902a (monovalent containing alum) vaccine, B. the VBI-2902e (monovalent containing alum and E6020) vaccine, C. a placebo, or D. a SARS-CoV-2 vaccine comprising the monovalent eVLP containing formulated with a proprietary adjuvant provided by the University of Saskatchewan (“TriAdj”). The monovalent eVLP vaccines contained the pre-fusion, stabilized version of the SARS-CoV-2 spike protein. The three components of the TriAdj per dose were 10 μg PCEP-3, 10 μg poly I:C and 20 μg IDR-1002. PCEP-3 was manufactured by Idaho National Laboratory. PolyTC was purchased from Invivogen (Cat# +lrl-picw; Lot# PIW-11-03). IDR- 1002 was synthesized by Peptide CPC Scientific (Cat# 818360; Lot# CN-11-00590). Table 6: Vaccine formulations and groupings for the vaccination of Syrian gdden hamsters
Figure imgf000028_0001
[0106] The study duration was 56 days. Animals were acclimatized for 7 days before immunization. One day before immunization (Day -1), under anesthesia, a temperature transponder was implanted. The vaccines were administered twice at a 3 -week interval, on Day 0, and Day 21, respectively. The vaccines were given via the intramuscular (IM) route at one side of the thighs, as specified in Table 5. The injection volume adminiserted was 100 μL. On Day 42 (3 weeks after the boosting immunization), all animals were challenged with the SARS- CoV-2 virus intranasally with 50 μL/nare, via both nares. The challenge viral dose was 1×105 TCID50 per animal.
[0107] During the immunization phase, compared to the start day (DayO), hamsters gained approximately 10% weight at week 1, 20% at week 2 or 30% at week 3. All groups had similar patterns of body weight growth. After challenge, Group A animals (Saline controls) had body weight reduction and they lost body weights peaking at 6-8 days post-challenge (dpc), approximately 15% of the initial weights (FIG. 9). The medians of % body weight changes of the Group B, C or D animals were only about 1-2% and peaked at 2dpc (FIG. 9).
[0108] At Day 14, Group B or C animals produced antibody median litres of 2.9× 103, and Group D at a median titre of 7.1×102 (FIG. 10). At Day 35, medians of antibody titres increased to 4.0× 104 (Group B), 5.3× 104 (Group C) and 1 ,7× 104 (Group D). Antibodies for pre-bleed (prior to priming immunization) were at background levels for all animals. At Day 14 or Day 35, Group A animals did not increase antibody production.
27 [0109] Viral RNA levels were at the peak at 2 day post-challenge (FIG. 11). Compared to Group A (Saline control), it seems that the levels of viral RNA were lower in Group B, C and D. Viral RNA levels were similar between Group B, C and D.
[0110] The above results from the Syrian golden hamster study further demonstrate that the vaccines of the present application are effective in preventing a SARS-CoV-2 infection.
[0111] All documents referenced in this disclosure are incorporated by reference herein, though if any incorporated document contradicts this written specification, then this written specification shall control. Those of skill in the will recognize that various changes and modifications may be made to the material provided herein, and that that material is within the scope and spirit of the disclosure.

Claims

We claim:
1. A vaccine comprising E6020 and virus-like particles comprising a coronavirus virus antigen and at least one additional adjuvant, wherein the at least one additional adjuvant is an aluminum-based adjuvant.
2. The vaccine according to claim 1, wherein the antigen is a SARS-CoV-2 spike protein possessing an amino acid sequence according to any one of SEQ ID NO: 1-4.
3. The vaccine according to claim 1, wherein the vaccine contains 0.1 μg to 100 μg of the antigen.
4. The vaccine according to claim 1, wherein the vaccine contains 1 μg to 25 μg of the antigen.
5. The vaccine according to claim 1, wherein the vaccine contains 5 μg to 10 μg of the antigen.
6. The vaccine according to claim 1, wherein the vaccine contains 1 μg to 50 μg of
E6020.
7. The vaccine of claim 1, wherein the vaccine contains 0.5 μg to 20 μg of E6020.
8. The vaccine according to claim 1, wherein the virus-like particles comprise 0.05 mg to 0.5 mg of Gag protein.
9. The vaccine according to claim 1, wherein the vaccine contains 0.01 mg to 5 mg of the at least one additional adjuvant.
10. The vaccine according to claim 1, wherein the vaccine contains 0.05 mg to 1.0 mg of the at least one additional adjuvant.
11. The vaccine according to claim 1, wherein the vaccine contains 150 μg to 180 μg of the at least one additional adjuvant.
12. The vaccine according to claim 1, wherein the vaccine comprises 0.1 mg/mL to 0.9 mg/mL of the at least one additional adjuvant.
13. The vaccine according to claim 1, wherein the vaccine comprises 0.1 mg/mL to 0.5 mg/mL of the at least one additional adjuvant.
14. The vaccine according to claim 1, wherein the vaccine comprises 0.165 mg/mL to 0.33 mg/mL of the at least one additional adjuvant.
15. The vaccine according to claim 1, wherein the at least one additional adjuvant is selected from the group consisting of an aluminum hydroxide adjuvant, an aluminum phosphate adjuvant, an aluminum salt and any combination thereof.
16. The vaccine according to claim 1, wherein the at least one additional adjuvant is an aluminum phosphate adjuvant.
17. The vaccine according to claim 1, wherein said vaccine is formulated as a buffered solution.
18. The vaccine according to claim 1, wherein said vaccine is formulated as an emulsion.
19. The vaccine according to claim 18, wherein said emulsion comprises surfactants.
20. The vaccine according to claim 18, wherein said emulsion comprises polymers.
21. The vaccine according to claim 1, wherein the vaccine is formulated as microparticles or nanoparticles.
22. A method of generating an immune response in a subject in need of mitigation of coronavirus infection comprising administering to the subject a vaccine according to any one of claims 1-21.
23. The method according to claim 22, wherein said vaccine is administered intranasally, intravenously, intradermally, intramuscularly or subcutaneously.
24. The method according to claim 22, wherein the vaccine is administered in a dose ranging from 0.1 mL to 10 mL.
25. The method according to claim 22, wherein the vaccine is administered in a dose ranging from 0.2 mL to 5 mL.
26. The method according to claim 22, wherein the vaccine is administered in a dose ranging from 0.3 mL to 3.0 mL.
27. A vaccine comprising E6020 and virus-like particles comprising an antigen.
28. The vaccine according to claim 27, wherein the antigen is derived from an influenza or corona virus.
29. The vaccine according to claim 27, wherein the antigen is a SARS-CoV-2 spike protein possessing an amino acid sequence according to any one of SEQ ID NO: 1-4.
30. The vaccine according to claim 27, wherein the vaccine contains 0.1 μg to 100 μg of the antigen.
31. The vaccine according to claim 27, wherein the vaccine contains 1 μg to 25 μg of the antigen.
32. The vaccine according to claim 27, wherein the vaccine contains 5 μg to 10 μg of the antigen.
33. The vaccine according to claim 27, wherein the vaccine contains 1 μg to 50 μg of
E6020.
34. The vaccine according to claim 27, wherein the vaccine contains 0.5 μg to 10 μg of E6020.
35. The vaccine according to claim 27, wherein the vaccine contains 0.1 μg to 100 μg of E6020.
36. The vaccine according to claim 27, wherein the vaccine contains 0.5 μg to 50 μg μg of E6020.
37. The vaccine according to claim 27, wherein the virus-like particles comprise a Gag protein of murine leukemia virus.
38. The vaccine according to claim 27, wherein the vaccine comprises at least one additional adjuvant selected from the group consisting of an aluminum hydroxide adjuvant, an aluminum phosphate adjuvant, an aluminum salt, chemokines, cytokines, nucleic acid sequences, lipoprotein, lipopolysaccharide, monophosphoryl lipid A, lipoteichoic acid, imiquimod, reiquimod, QS-21 and any combination thereof.
39. The vaccine according to claim 38, wherein the vaccine comprises 50 μg to 50 mg of the at least one additional adjuvant.
40. The vaccine according to claim 38, wherein the vaccine comprises 0.1 mg to 20.0 mg of the at least one additional adjuvant.
41. The vaccine according to claim 38, wherein the vaccine comprises 1.0 mg to 5.0 mg of the at least one additional adjuvant.
42. The vaccine according to claim 38, wherein the at least one additional adjuvant is an aluminum hydroxide adjuvant or an aluminum phosphate adjuvant.
43. The vaccine according to claim 27, wherein the virus-like particles comprise 10 μg to 10 mg of Gag protein.
44. The vaccine according to claim 27, wherein the virus-like particles contain 50 μg to 5 mg of Gag protein.
45. The vaccine according to claim 27, wherein the virus-like particles contain 100 μg to 500 μg of Gag protein.
46. The vaccine according to claim 27, wherein the virus-like particles contain 1 μg to 50 mg of Gag protein.
47. The vaccine according to claim 27, wherein the vaccine is formulated as a buffered solution.
48. The vaccine according to claim 27, wherein the vaccine is formulated as an emulsion.
49. The vaccine according to claim 48, wherein said emulsion comprises surfactants.
50. The vaccine according to claim 48, wherein said emulsion comprises polymers.
51. A method of generating an immune response in a subject in need of mitigation of coronavirus infection comprising administering to the subject a vaccine according to any one of claims 27-50.
52. The method according to claim 51, wherein said vaccine is administered intranasally, intravenously, intradermally, intramuscularly or subcutaneously.
53. The method according to claim 51, wherein the vaccine is administered in a dose ranging from 0.1 mL to 10 mL.
54. The method according to claim 51, wherein the vaccine is administered in a dose ranging from 0.2 mL to 5 mL.
55. The method according to claim 51, wherein the vaccine is administered in a dose ranging from 0.3 mL to 3.0 mL.
56. A vaccine comprising E6020 and a Nidovirales virus antigen.
57. The vaccine of claim 56, wherein the Nidovirales virus antigen is a coronavirus antigen.
58. The vaccine of claim 56, wherein the coronavirus antigen is a SARS-CoV-2 antigen.
59. The vaccine according to claim 56, further comprising one or more additional adjuvants.
60. The vaccine according to claim 56, wherein said vaccine is formulated as a buffered solution.
61. The vaccine according to claim 56, wherein said vaccine is formulated as an emulsion.
62. The vaccine according to claim 61, wherein said emulsion comprises surfactants.
63. The vaccine according to claim 61, wherein said emulsion comprises polymers.
64. The vaccine according to claim 56, wherein said vaccine is formulated as microparticles or nanoparticles.
65. A method of generating an immune response in a subject in need of mitigation of coronavirus infection comprising administering to the subject a vaccine according to any one of claims 56-64.
66. The method according to claim 65, wherein said vaccine is administered intranasally, intravenously, intradermally, intramuscularly or subcutaneously.
67. The method according to claim 65, wherein the vaccine is administered in a dose ranging from 0.1 mL to 10 mL.
68. A method of generating an immune response in a subject in need of mitigation of coronavirus infection comprising administering to the subject a vaccine comprising E6020 and an antigen.
69. The method according to claim 68, wherein the antigen is derived from a viral or bacterial pathogen.
70. The method according to claim 68, wherein the antigen is derived from the influenza virus.
71. The method according to claim 68, wherein the antigen is derived from the corona virus.
72. The method according to claim 68, wherein the antigen is a SARS-CoV-2 spike protein.
73. The method according to claim 68, wherein the vaccine further comprises one or more additional adjuvants.
74. The method according to claim 68, wherein the vaccine is formulated as a buffered solution.
75. The method according to claim 68, wherein said vaccine is formulated as an emulsion.
76. The method according to claim 75, wherein said emulsion comprises surfactants.
77. The method according to claim 75, wherein said emulsion comprises polymers.
78. The method according to claim 69, wherein said vaccine is formulated as microparticles or nanoparticles.
79. The method according to claim 69, wherein said vaccine is administered intranasally, intravenously, intramuscularly or subcutaneously.
80. The method according to claim 69, wherein the vaccine is administered in a dose ranging from 0.1 mL to 10 mL.
PCT/US2021/026058 2020-04-06 2021-04-06 Vaccines, adjuvants, and methods of generating an immune response WO2021207281A2 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
IL297093A IL297093A (en) 2020-04-06 2021-04-06 Vaccines, adjuvants, and methods of generating an immune response
MX2022012527A MX2022012527A (en) 2020-04-06 2021-04-06 Vaccines, adjuvants, and methods of generating an immune response.
EP21722639.8A EP4132577A2 (en) 2020-04-06 2021-04-06 Vaccines, adjuvants, and methods of generating an immune response
BR112022020298A BR112022020298A2 (en) 2020-04-06 2021-04-06 VACCINES, ADJUVANTS AND METHODS TO GENERATE AN IMMUNE RESPONSE
CN202180040867.8A CN115916254A (en) 2020-04-06 2021-04-06 Vaccines, adjuvants and methods for generating immune responses
US17/995,546 US20230149537A1 (en) 2020-04-06 2021-04-06 Vaccines, adjuvants, and methods of generating an immune response
JP2022560944A JP2023520603A (en) 2020-04-06 2021-04-06 Vaccines, adjuvants, and methods for eliciting an immune response
KR1020227038812A KR20230034936A (en) 2020-04-06 2021-04-06 Vaccines, adjuvants and methods for generating an immune response
AU2021252972A AU2021252972A1 (en) 2020-04-06 2021-04-06 Vaccines, adjuvants, and methods of generating an immune response
CA3179739A CA3179739A1 (en) 2020-04-06 2021-04-06 Vaccines, adjuvants, and methods of generating an immune response

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063005908P 2020-04-06 2020-04-06
US63/005,908 2020-04-06

Publications (2)

Publication Number Publication Date
WO2021207281A2 true WO2021207281A2 (en) 2021-10-14
WO2021207281A3 WO2021207281A3 (en) 2021-12-16

Family

ID=75747059

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/026058 WO2021207281A2 (en) 2020-04-06 2021-04-06 Vaccines, adjuvants, and methods of generating an immune response

Country Status (11)

Country Link
US (1) US20230149537A1 (en)
EP (1) EP4132577A2 (en)
JP (1) JP2023520603A (en)
KR (1) KR20230034936A (en)
CN (1) CN115916254A (en)
AU (1) AU2021252972A1 (en)
BR (1) BR112022020298A2 (en)
CA (1) CA3179739A1 (en)
IL (1) IL297093A (en)
MX (1) MX2022012527A (en)
WO (1) WO2021207281A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022051859A1 (en) * 2020-09-11 2022-03-17 Manuel Caruso Pseudotyped retroviral particles for inducing immunity against coronavirus infections
WO2022127946A1 (en) * 2020-12-16 2022-06-23 Instituto Finlay De Vacunas Use of vaccine compositions based on sars-cov-2 receptor binding domain in delivering protective immunity

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9765304B2 (en) 2000-10-26 2017-09-19 L'universite Pierre Et Marie Curie Synthetic viruses and uses thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1910387B1 (en) * 2005-06-30 2016-11-16 Eisai R&D Management Co., Ltd. Compounds for preparing immunological adjuvant
TR201807340T4 (en) * 2013-02-01 2018-06-21 Glaxosmithkline Biologicals Sa Intradermal administration of immunological compositions comprising bell-like receptor agonists.

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9765304B2 (en) 2000-10-26 2017-09-19 L'universite Pierre Et Marie Curie Synthetic viruses and uses thereof

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
"Coronaviruses", 1 April 2020, NATIONAL INSTITUTE OF ALLERGY AND INFECTIOUS DISEASE
ABOUDOUNYA M. M. ET AL., MEDIATORS INFLAMM., 2021
ATEYO, IMMUNITY, 2020
BHATTACHARYA, M. ET AL., INFECT. GENET. EVOL., vol. 85, 2020, pages 104587
GRIFFONI, CELL, 2020
GRIFONI ASIDNEY JZHANG YSCHEUERMANN RHPETERS BSETTE A, CELL HOST MICROBE, 12 March 2020 (2020-03-12)
HANTOUSHZADEH S. ET AL., ARCH. MED. RES., vol. 51, 2020, pages 347 - 348
INGRAHAM N. E. ET AL., LANCET RESPIR. MED., vol. 8, 2020, pages 544 - 546
KHANMOHAMMADI S. ET AL., J MED VIROL., vol. 93, 2021, pages 615 - 617
LEE J. S. ET AL., SCI. IMMUNOL., vol. 5, 2020
MCCRAY ET AL., JOURNAL OF VIROLOGY, January 2007 (2007-01-01), pages 813 - 821
NISREEN M.A. OKBAMARCEL A. MIILLERWENTAO LI ET AL., MEDRXIV, Retrieved from the Internet <URL:https://doi.org/10.1101/2020.03.18.20038059>
PEEPLES, PNAS, 2020
S. PERLMAN ET AL., NATURE REVIEWS MICROBIOLOGY, vol. 7, 2009, pages 439 - 450
SHI ET AL., BIORXIV PREPRINT, Retrieved from the Internet <URL:https://doi.org/10.1101/2020.03.30.015347>
STADLER ET AL., EMERGING INFECTIOUS DISEASES, vol. 11, no. 8, August 2005 (2005-08-01), pages 1312
TAI ET AL., CELLULAR AND MOLECULAR IMMUNOLOGY, Retrieved from the Internet <URL:https://doi.org/10.1038/s41423-020-0400-4>
TANG Y., FRONT. IMMUNOL., vol. 11, 2020, pages 1708

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022051859A1 (en) * 2020-09-11 2022-03-17 Manuel Caruso Pseudotyped retroviral particles for inducing immunity against coronavirus infections
WO2022127946A1 (en) * 2020-12-16 2022-06-23 Instituto Finlay De Vacunas Use of vaccine compositions based on sars-cov-2 receptor binding domain in delivering protective immunity

Also Published As

Publication number Publication date
US20230149537A1 (en) 2023-05-18
IL297093A (en) 2022-12-01
WO2021207281A3 (en) 2021-12-16
KR20230034936A (en) 2023-03-10
CA3179739A1 (en) 2021-10-14
JP2023520603A (en) 2023-05-17
BR112022020298A2 (en) 2022-12-20
MX2022012527A (en) 2023-03-13
CN115916254A (en) 2023-04-04
EP4132577A2 (en) 2023-02-15
AU2021252972A1 (en) 2022-11-10

Similar Documents

Publication Publication Date Title
US11759516B2 (en) Nucleic acid vaccine against the SARS-CoV-2 coronavirus
WO2022110099A1 (en) Coronavirus vaccines and uses thereof
US20230346916A1 (en) Immunogenic compositions against severe acute respiratory syndrome coronavirus 2
US20230149537A1 (en) Vaccines, adjuvants, and methods of generating an immune response
US20230372466A1 (en) Universal mammalian influenza vaccine
US11253587B2 (en) Vaccine compositions for the treatment of coronavirus
AU2021298886A1 (en) A dna plasmid SARS-Coronavirus-2/covid-19 vaccine
EP4144752A1 (en) Viral-like particles for the treatment or prevention of an infection by a coronaviridae virus
US11964013B2 (en) Nucleic acid vaccine against the SARS-CoV-2 coronavirus
US20220233682A1 (en) Vaccine compositions for the treatment of coronavirus
KR20240052044A (en) Virus-like particles for treating or preventing infection by coronaviruses
AU2022289462A9 (en) Virus-like particle vaccine for coronavirus
AU2022336142A1 (en) Viral-like particles for the treatment or prevention of an infection by a coronaviridae virus
Özcengiz et al. Two Formulations of a SARS-CoV-2 Recombinant Subunit Vaccine Candidate Made Up of S1 Fragment Protein (P1), S2 Fragment Protein (P2) and Nucleocapsid Protein (N) Elicit Strong Immunogenicity in Mice
CN117203243A (en) Compositions and methods for preventing RSV and PIV3 infection

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: 21722639

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2022560944

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3179739

Country of ref document: CA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112022020298

Country of ref document: BR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021252972

Country of ref document: AU

Date of ref document: 20210406

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2021722639

Country of ref document: EP

Effective date: 20221107

ENP Entry into the national phase

Ref document number: 112022020298

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20221006