WO2017070735A1 - Vaccin à base de liposomes - Google Patents

Vaccin à base de liposomes Download PDF

Info

Publication number
WO2017070735A1
WO2017070735A1 PCT/AU2016/051005 AU2016051005W WO2017070735A1 WO 2017070735 A1 WO2017070735 A1 WO 2017070735A1 AU 2016051005 W AU2016051005 W AU 2016051005W WO 2017070735 A1 WO2017070735 A1 WO 2017070735A1
Authority
WO
WIPO (PCT)
Prior art keywords
immunogenic
immunogenic agent
protein
fragment
agent
Prior art date
Application number
PCT/AU2016/051005
Other languages
English (en)
Inventor
Michael Good
Mehfuz ZAMAN
Original Assignee
Griffith University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2015904403A external-priority patent/AU2015904403A0/en
Application filed by Griffith University filed Critical Griffith University
Priority to CN201680074917.3A priority Critical patent/CN109069424B/zh
Publication of WO2017070735A1 publication Critical patent/WO2017070735A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • A61K39/092Streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/543Mucosal route intranasal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; 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
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55583Polysaccharides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6037Bacterial toxins, e.g. diphteria toxoid [DT], tetanus toxoid [TT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6062Muramyl peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/62Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16211Influenzavirus B, i.e. influenza B virus
    • C12N2760/16234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • LIPOSOMAL VACCINE TECHNICAL FIELD relates to prevention and treatment of infectious diseases. More particularly, this invention relates to a liposomal vaccine for treating or preventing infectious diseases and conditions by inducing a mucosal immune response.
  • Systemic immunity has proven effective in preventing diseases caused by a variety of different pathogens through serum immunoglobulin (Ig) at systemic sites, but not in preventing colonisation of mucosal sites and thus person-to- person transmission. Therefore, for some diseases, systemic vaccination is not the optimal approach for inducing immunity.
  • mucosal vaccines against various organisms administered nasally are effective in inducing antigen-specific immune responses in both systemic and mucosal compartments. Due to this two- layered protective immunity, mucosal vaccination is an ideal strategy for combating both systemic and mucosal infections with the added benefit that prevention of mucosal colonization will also inhibit transmission by droplets and aerosols from the upper respiratory tract. Mucosal vaccination is also economically advantageous, an important consideration for vaccine development. Owing to the ease of vaccine administration by the nasal route, use of needles can be avoided. Pain-free, delivery will aid in greater patient compliance.
  • An objective of the present invention is to provide an immunogenic agent and delivery system that elicits a mucosal immune response to a pathogen.
  • the invention relates to facilitating or inducing mucosal immunity to a by delivery of an immunogenic protein, fragment or variant, by way of a lipid vesicle that further comprises a carrier protein such as diptheria toxoid (DT).
  • DT diptheria toxoid
  • the carrier protein is located in an intravesicular space.
  • a single immunogenic agent comprises a plurality of different immunogenic proteins, fragments or variants from a plurality of different pathogens.
  • An aspect of the invention provides an immunogenic agent suitable for administration to a mammal, said immunogenic agent comprising one or a plurality of immunogenic proteins, fragmentd, variants or derivatives thereof, a lipid vesicle and a carrier protein or a fragment or variant thereof.
  • the carrier protein is diptheria toxoid (DT).
  • the immunogenic agent is a lipid vesicle comprising a plurality of immunogenic proteins, fragments, variants or derivatives thereof from different pathogens.
  • Another aspect of the invention provides a composition comprising the immunogenic agent of the aforementioned aspect.
  • the composition comprises an immunogenic agent that includes a lipid vesicle comprising a plurality of immunogenic proteins, fragments, variants or derivatives thereof, of or from the same or single pathogen.
  • the composition comprises a plurality of different immunogenic agents that respectively comprise one or plurality of immunogenic proteins, fragments, variants or derivatives thereof, of or from different pathogens.
  • the composition comprises a single immunogenic agent that includes a lipid vesicle comprising a plurality of immunogenic proteins, fragments, variants or derivatives thereof, of or from different pathogens.
  • the composition comprises a plurality of different immunogenic agents that respectively comprise one or plurality of immunogenic proteins, fragments, variants or derivatives thereof, of or from different pathogens.
  • Another aspect of the invention provides a method of eliciting an immune response to one or a plurality of pathogens in a mammal, said method including the step of administering to the mammal an immunogenic agent comprising one or a plurality of immunogenic proteins, fragments, variants or derivatives thereof, a lipid vesicle and diptheria toxoid (DT) or a fragment or variant thereof, or a composition comprising same, to thereby elicit an immune response to the one or plurality of pathogens in the mammal.
  • an immunogenic agent comprising one or a plurality of immunogenic proteins, fragments, variants or derivatives thereof, a lipid vesicle and diptheria toxoid (DT) or a fragment or variant thereof, or a composition comprising same, to thereby elicit an immune response to the one or plurality of pathogens in the mammal.
  • DT diptheria toxoid
  • Another aspect of the invention provides a method of immunizing a mammal against one or a plurality of pathogens, said method including the step of administering to the mammal an immunogenic agent comprising one or a plurality of immunogenic proteins, fragments, variants or derivatives thereof, a lipid vesicle and diptheria toxoid (DT) or a fragment or variant thereof, or a composition comprising same, to thereby immunize the mammal against the one or plurality of pathogens.
  • an immunogenic agent comprising one or a plurality of immunogenic proteins, fragments, variants or derivatives thereof, a lipid vesicle and diptheria toxoid (DT) or a fragment or variant thereof, or a composition comprising same, to thereby immunize the mammal against the one or plurality of pathogens.
  • DT diptheria toxoid
  • Yet another aspect of the invention provides a method of treating or preventing infection by one or a plurality of pathogens in a mammal, said method including the step of administering to the mammal an immunogenic agent comprising one or a plurality of immunogenic proteins, fragments, variants or derivatives thereof, a lipid vesicle and diptheria toxoid (DT) or a fragment or variant thereof, or a composition comprising same, to thereby treat or prevent the infection by the one or plurality of pathogens in the mammal.
  • an immunogenic agent comprising one or a plurality of immunogenic proteins, fragments, variants or derivatives thereof, a lipid vesicle and diptheria toxoid (DT) or a fragment or variant thereof, or a composition comprising same, to thereby treat or prevent the infection by the one or plurality of pathogens in the mammal.
  • the immunogenic agent elicits a mucosal immune response.
  • the mucosal immune response includes the production of IgA.
  • the immunogenic agent is administered intranasally.
  • the immunogenic protein, fragment or variant is displayed on the surface of the lipid vesicle.
  • the diptheria toxoid (DT) or a fragment or variant thereof is located in an intravesicular space within the vesicle.
  • the lipid vesicle is a liposome.
  • the immunogenic protein fragment or variant is lipidated.
  • a lysine (K) residue at the N-terminus of the immunogenic protein fragment or variant is lipidated.
  • an N-terminal lysine (K) residue is lipidated via a and ⁇ amino groups.
  • the or each lipid is a C 16 fatty acid such as palmitic acid.
  • the N-terminal lysine (K) residue is in a spacer amino acid sequence at the N-terminus of the immunogenic protein fragment or variant.
  • the immunogenic protein, fragment or variant is a group A streptococcus M protein fragment, variant or derivative thereof.
  • the immunogenic protein is an agent that facilitates restoring or enhancing neutrophil activity.
  • the M protein fragment is or comprises a conserved region of the M protein.
  • the fragment is an immunogenic fragment that comprises, or is contained within a pi 45 peptide.
  • the immunogenic fragment is within, or comprises, a J8 peptide or variant thereof.
  • the J8 peptide comprises, or consists essentially of, the amino acid sequence Q AEDKVKQ SRE AKKQ VEK ALKQLEDKVQ (SEQ ID NO: l).
  • the agent that facilitates restoring or enhancing neutrophil activity is the protein is SpyCEP, or a fragment thereof.
  • the SpyCEP fragment comprises, or consists essentially of, the amino acid sequence NSDNIKENQFEDFDEDWENF (SEQ ID NO:2).
  • the SpyCEP fragment and the M protein fragment may be fused to form a single, chimeric peptide.
  • the chimeric peptide is, may comprise, or consist essentially of, the amino acid sequence NSDNIKENQFEDFDEDWE FQAEDKVKQSREAKKQVEKALKQLEDKVQ
  • the immunogenic protein, fragment or variant is of an influenza virus.
  • the influenza virus may be influenza A.
  • a non-limiting example is, comprises, or consists essentially of, the amino acid sequence M SLLTE VETPIRNEWGCRC D S SD (SEQ ID NO:4).
  • the influenza virus may be influenza B.
  • a non-limiting example is, or comprises, or consists essentially of, the amino acid sequence PAKLLKERGFFGAIAGFLE (SEQ ID NO: 5).
  • the immunogenic protein, fragment or variant is of a rhinovirus.
  • a non-limiting example is, comprises, or consists essentially of, the amino acid sequence
  • GAQVSTQKSGSHENQNILTNGSNQTFTVINY (SEQ ID NO: 6).
  • Another non- limiting example is, comprises, or consists essentially of, the amino acid sequence GAQVSRQNVGTHSTQNMVSNGSSL (SEQ ID NO: 7).
  • the immunogenic protein, fragment or variant is of a worm, such as a hookworm.
  • a non-limiting example is, comprises, or consists essentially of, the amino acid sequence
  • the immunogenic agent may further comprise an activator of innate immunity.
  • the activator of innate immunity may target a C- type lectin such as macrophage inducible Ca 2+ -dependent (C-type) lectin ("Mincle").
  • the activatopr of innate imunity may be a glycolipid.
  • Non-limiting examples include a glycolipid such as the mycobacterial cord factor trehalose- 6,6'-dimycolate (TDM) and/or its synthetic analogue trehalose-6,6'-dibehenate (TDB) or a lipid A glycolipid adjuvant.
  • the immunogenic agent may further comprise a bile salt such as sodium deoxycholate.
  • FIG. 1 Idealized structure of J8-Lipo-DT. Liposome encapsulates DT while J8 attached to the spacer KSS at the N-terminus is covalently coupled to two palmitic acid molecules, facilitating the insertion of J8 into the liposome membrane.
  • FIG. 1 J8-specific antibody response for individual BALB/c mice. Mean antibody titer are represented as a bar. A) Salivary IgA titer. B) Fecal IgA titer. C) Serum IgG titer. Statistical analysis was performed using a one-way ANOVA followed by the Tukey post hoc test (ns, p> 0.05; *, p ⁇ 0.05; **, p ⁇ 0.01; ***, p ⁇ 0.001).
  • FIG. 3 Bacterial burden after intranasal challenge with Ml GAS strain in BALB/C mice.
  • Results are represented as the mean CFU + SEM for 10 mice/group on days 1-3 for throat swabs, nasal shedding and day 3 for NALT.
  • Statistical analysis was performed using a nonparametric, unpaired Mann- Whitney U test to compare test groups to the PBS control group (ns, p> 0.05; *, p ⁇ 0.05; **, p ⁇ 0.01; ***, p ⁇ 0.001).
  • FIG. 1 URT GAS challenge model assessing bacterial burden after intranasal challenge with Ml strain.
  • Statistical analysis was performed using a nonparametric, unpaired Mann- Whitney U test to compare test groups to the PBS control group (ns, p> 0.05; *, p ⁇ 0.05).
  • FIG. 6 J8-specific antibody response for individual BALB/c mice. Mean antibody titer are represented as a bar. A) Salivary IgA titer. B) Serum IgG titer. Statistical analysis was performed using a one-way ANOVA followed by the Tukey post hoc test (ns, p> 0.05; *, p ⁇ 0.05; **, /? ⁇ 0.01; ***, /? ⁇ 0.001).
  • FIG. 7 Antigen specific secreted chemokines and cytokines in immunized mice. Splenocytes were plated out and the following stimuli were added as indicated: LPS (2 ⁇ g/mL), J8 (10 ⁇ g/mL) or media alone. 72 hours post stimulation, supematants were isolated and levels of secreted chemokines or cytokines were assayed using a cytometric bead array (see Materials and Methods). Statistical analysis was performed using a Student's t test (ns, p> 0.05; *, p ⁇ 0.05; **, /? ⁇ 0.01).
  • FIG. 8 Levels of surface markers on human DC subsets with and without treatment with reagents. The following stimuli were added as indicated: Polyinosinic:polycytidylic acid (pIC, 10 ⁇ g/mL), J8-Lipo-DT (150 ⁇ g/mL) or media alone. Cell surface markers were measured by a flow cytometer 24 hours post stimulation. A) CD123+ plasmacytoid DCs. B) CD141+ classical type 1 DCs. C) CDlc+ classical type 2 DCs. Values are expressed as the median fluorescence intensities (MFI) for pooled data from three individual donors ⁇ SEM. Statistical analysis was performed using a nonparametric, unpaired Mann- Whitney U test to compare test groups to the media control group (ns, p> 0.05; *, p ⁇ 0.05; **, p ⁇ 0.01; ***, ⁇ 0.001).
  • MFI median fluorescence intensities
  • FIG. 9 J8-Lipo-DT induced secreted chemokines and cytokines in human dendritic cells.
  • Dendritic cells were plated out and the following stimuli were added as indicated: pIC (10 ⁇ g/mL), J8-Lipo-DT (150 ⁇ g/mL) or media alone.
  • Supematants were isolated 24 hours post stimulation and levels of secreted chemokines or cytokines were assayed using a cytometric bead array (see Materials and Methods).
  • Statistical analysis was performed using a Student's t test (ns, p> 0.05; *, p ⁇ 0.05; **, p ⁇ 0.01; ***, p ⁇ 0.001).
  • SpyCEP peptide S2; SEQ ID NO:2
  • liposomal delivery agent elicits a mucosal IgA response.
  • Liposomes displaying palmitic acid lipidated S2 peptide or an S2-J8 chimera (SEQ ID NO: 3), together with intravesicular DT were administered intranasally to mice and S2-specific IgA titers measured.
  • J8-Lipo-DT immunogenic agents can be extruded to form nano- to micro-sized particles.
  • Liposome size measurement was by Nanosizer (Dynamic light scattering or DLS).
  • Figure 16 Reconstituted, freeze-dried, J8-Lipo-DT liposomal immunogenic agents induced J8-specific systemic response without additional adjuvant.
  • FIG. 18 Schematic depiction of a liposomal J8-Lipo-DT immunogenic agent comprising glycolipid adjuvants trehalose 6,6'-dibehenate (TDB).
  • TDB glycolipid adjuvants trehalose 6,6'-dibehenate
  • TDB enhances mucosal IgA response induced by J8-Lipo-DT.
  • Figure 20 Schematic depiction of a liposomal immunogenic agent comprising the bile salt sodium deoxycholate.
  • Figure 21 (A) Schematic depiction of a single immunogenic agent comprising a lipid vesicle, intravesicular DT and immunogenic proteins of, or from, a plurality of different pathogens, namely influenza A, influenza B and group A streptococcus; (B) Schematic depiction of a single immunogenic agent comprising a lipid vesicle, intravesicular DT and immunogenic proteins of, or from, a plurality of different pathogens, namely influenza A, influenza B and group A streptococcus and glycolipid adjuvants trehalose 6,6'-dibehenate (TDB) and Monophosphoryl 3-Deacyl Lipid A (3D-PHAD®).
  • TDB Monophosphoryl 3-Deacyl Lipid A
  • FIG 22 Immunogenicity of a single immunogenic agent ("Multivax”) against influenza A, influenza B and group A streptococcus compared to individual vaccination with separate immunogenic agenst against each of influenza A, influenza B and group A streptococcus ("Single Antigen-vax”), as measured by antigen-specific salivary IgA titre.
  • Multivax single immunogenic agent
  • Single Antigen-vax separate immunogenic agenst against each of influenza A, influenza B and group A streptococcus
  • the present invention is at least partly predicated on the discovery that intranasal vaccination of mice with a liposomal immunogenic agent comprising an immunogenic peptide displayed on the liposome surface together with an intravesicular carrier protein such as diptheria toxoid (DT) resulted in mucosal and systemic antibody responses which were comparable to those induced by the established non- human compatible adjuvant, CTB.
  • DT diptheria toxoid
  • the liposomal immunogenic agent may comprise a SpyCEP peptide or other fragment thereof, alone or together with the J8 peptide.
  • the liposomal immunogenic agent may be suitable for treating or preventing infections by particularly virulent strains or isolates of Group A streptococci that are resistant to the typical antibiotic treatments used for group A streptococcal infections.
  • an embodiment of the invention provides a single immunogenic agent that includes one or a plurality of immunogenic proteins, fragments, variants or derivatives of, or from, a plurality of different pathogens.
  • isolated material that has been removed from its natural state or otherwise been subjected to human manipulation. Isolated material may be substantially or essentially free from components that normally accompany it in its natural state, or may be manipulated so as to be in an artificial state together with components that normally accompany it in its natural state. Isolated material may be in native, chemical synthetic or recombinant form.
  • protein is meant an amino acid polymer.
  • the amino acids may be natural or non-natural amino acids, D- or L-amino acids as are well understood in the art.
  • protein includes and encompasses "peptide”, which is typically used to describe a protein having no more than fifty (50) amino acids and "polypeptide”, which is typically used to describe a protein having more than fifty (50) amino acids.
  • a protein is referred to as being "of or "from” a pathogen is meant that the protein comprises an amino acid sequence which is at least partly, or entirely, present in a protein of the pathogen.
  • a “fragment” is a segment, domain, portion or region of a protein, which constitutes less than 100% of the amino acid sequence of the protein. It will be appreciated that the fragment may be a single fragment or may be repeated alone or with other fragments.
  • fragments may comprise, consist essentially of or consist of up to 5, 6, 7, 8, 9, 10, 12, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 100, 1050, 1100, 1150, 1200, 1250, 1300, 1350, 1400, 1450, 1500, 1550 or 1600 amino acids of the full length protein.
  • a protein "variant” shares a definable nucleotide or amino acid sequence relationship with a reference amino acid sequence.
  • the "variant” protein may have one or a plurality of amino acids of the reference amino acid sequence deleted or substituted by different amino acids. It is well understood in the art that some amino acids may be substituted or deleted without changing the activity of the immunogenic fragment and/or protein (conservative substitutions).
  • protein variants share at least 70% or 75%, preferably at least 80%> or 85% or more preferably at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%) or 99% sequence identity with a reference amino acid sequence.
  • sequence comparisons are typically performed by comparing sequences over a “comparison window” to identify and compare local regions of sequence similarity.
  • a “comparison window” refers to a conceptual segment of typically 6, 9 or 12 contiguous residues that is compared to a reference sequence.
  • the comparison window may comprise additions or deletions (i.e., gaps) of about 20% or less as compared to the reference sequence for optimal alignment of the respective sequences.
  • Optimal alignment of sequences for aligning a comparison window may be conducted by computerised implementations of algorithms (Geneworks program by Intelligenetics; GAP, BESTFIT, FAST A, and TFASTA in the Wisconsin Genetics Software Package Release 7.0, Genetics Computer Group, 575 Science Drive Madison, WI, USA, incorporated herein by reference) or by inspection and the best alignment (i.e. resulting in the highest percentage homology over the comparison window) generated by any of the various methods selected.
  • sequence identity is used herein in its broadest sense to include the number of exact nucleotide or amino acid matches having regard to an appropriate alignment using a standard algorithm, having regard to the extent that sequences are identical over a window of comparison.
  • a “percentage of sequence identity” is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical nucleic acid base ⁇ e.g., A, T, C, G, I) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison ⁇ i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity.
  • sequence identity ' may be understood to mean the "match percentage” calculated by the DNASIS computer program (Version 2.5 for windows; available from Hitachi Software engineering Co., Ltd., South San Francisco, California, USA).
  • derivatives ' are molecules such as proteins, fragments or variants thereof that have been altered, for example by conjugation or complexing with other chemical moieties, by post-translational modification ⁇ e.g. phosphorylation, acetylation and the like), modification of glycosylation ⁇ e.g. adding, removing or altering glycosylation), lipidation and/or inclusion of additional amino acid sequences as would be understood in the art.
  • an additional amino acid sequence may comprise one or a plurality of lysine residues at an N and/or C-terminus thereof.
  • the plurality of lysine residues may be a linear sequence of lysine residues or may be branched chain sequences of lysine residues. These additional lysine residues may facilitate increased peptide solubility.
  • Additional amino acid sequences may include fusion partner amino acid sequences which create a fusion protein.
  • fusion partner amino acid sequences may assist in detection and/or purification of the isolated fusion protein.
  • Non-limiting examples include metal-binding ⁇ e.g. polyhistidine) fusion partners, maltose binding protein (MBP), Protein A, glutathione S-transferase (GST), fluorescent protein sequences ⁇ e.g. GFP), epitope tags such as myc, FLAG and haemagglutinin tags.
  • Other additional amino acid sequences include spacer sequences.
  • spacer sequence is an amino acid sequence at the N- or C-terminus of an amino acid sequence of an immunogenic protein fragment or variant that includes a lysine (K) residue suitable for lipidation.
  • the spacer amino acid sequence comprises two (2) to ten (10) amino acids, such as the three (3) amino acid sequence KSS.
  • Other derivatives contemplated by the invention include, but are not limited to, modification to side chains, incorporation of unnatural amino acids and/or their derivatives during peptide, or protein synthesis and the use of crosslinkers and other methods which impose conformational constraints on the immunogenic proteins, fragments and variants of the invention.
  • immunogenic proteins, fragments and variants disclosed herein may be displayed on the surface of a lipid vesicle individually or as a chimera or fusion protein comprising multiple copies of the same peptide or multiple different peptides.
  • a non-limiting example is the chimeric peptide of SEQ ID NO:3, as will be described in more detail hereinafter.
  • immunogenic indicates the ability or potential to generate or elicit an immune response, such as to a pathogen or molecular components thereof, upon administration of the immunogenic agent to a mammal or other animal.
  • the one or more elements of the immune system include B lymphocytes, antibodies, neutrophils, dendritic cells inclusive of plasmacytoid dendritic cells, cytokines and/or chemokines.
  • cytokines include pro- inflammatory cytokines such as TNF-a, IL-6 and IL-1 (e.g IL-1 ⁇ ).
  • a chemokine is the neutrophil chemo-attractant IL-8.
  • the immune response is, or comprises, a mucosal immune response, such as including IgA production.
  • the immune response that is elicited by the immunogenic agent is protective.
  • pathogen refers to any living or non-living entity that is capable of causing a disease, disorder or condition in an animal, such as an avian or mammal.
  • the pathogen may be a virus, bacterium, protozoan or worm, although without limitation thereto.
  • pathogens include group A streptococcus bacteria, respiratory viruses such as influenza virus and rhinovirus and nematodes such as hookworms.
  • the invention provides a lipid vesicle formulation that comprises an immunogenic protein fragment, variant or derivative and a carrier protein formulated in a lipid vesicle.
  • the lipid vesicle may be a liposome, minicell, multilamellar vesicle, micelle, vacuole or other vesicular structure comprising a lipid bilayer,
  • the immunogenic protein fragment, variant or derivative is derivatized to comprise one or more lipids that facilitate anchoring to the lipid bilayer so that the immunogenic protein fragment, variant or derivative is displayed on the surface of the lipid vesicle.
  • a lysine (K) residue is lipidated via a and/or ⁇ amino groups.
  • the immunogenic protein fragment, variant or derivative may further comprise an N-terminal spacer comprising a lysine (K) residue that is lipidated.
  • the spacer may typically comprise 2, 3, 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids.
  • An embodiment is the three (3) amino acid spacer KSS.
  • the lipid may comprise a saturated or unsaturated fatty acid (e.g mono-unsaturated or polyunsaturated).
  • the or each lipid is a Ci6 fatty acid such as palmitic acid.
  • lipids such as saturated or unsaturated (e.g mono-unsaturated or polyunsaturated) fatty acids having C 12 , C 13 C 14 , C 15 C 17 , C 18; C19, C20, C 21 or C22 carbon chains may be useful according to the invention.
  • saturated or unsaturated (e.g mono-unsaturated or polyunsaturated) fatty acids having C 12 , C 13 C 14 , C 15 C 17 , C 18; C19, C20, C 21 or C22 carbon chains may be useful according to the invention.
  • the lipid vesicle suitably comprises any lipid or mixture of lipids capable of forming a lipid bilayer structure.
  • lipids capable of forming a lipid bilayer structure.
  • phospholipids include phosphatidylcholine (PC) (lecithin), phosphatidic acid, phosphatidylethanolamine (PE) (cephalin), phosphatidylglycerol (PG), phosphatidyl serine (PS), phosphatidylinositol (PI) and sphingomyelin (SM) or natural or synthetic derivatives thereof.
  • PC phosphatidylcholine
  • PE phosphatidylethanolamine
  • PG phosphatidylglycerol
  • PS phosphatidyl serine
  • PI phosphatidylinositol
  • SM sphingomyelin
  • Natural derivatives include egg PC, egg PG, soy bean PC, hydrogenated soy bean PC, soy bean PG, brain PS, sphingolipids, brain SM, galactocerebroside, gangliosides, cerebrosides, cephalin, cardiolipin, and dicetylphosphate.
  • Synthetic derivatives include dipalmitoylphosphatidylcholine (DPPC), didecanoylphosphatidylcholine (DDPC), dierucoylphosphatidylcholine (DEPC), dimyristoylphosphatidylcholine (DMPC), distearoylphosphatidylcholine (DSPC), dilaurylphosphatidylcholine (DLPC), palmitoyloleoylphosphatidylcholine (POPC), palmitoylmyristoylphosphatidylcholine (PMPC), palmitoylstearoylphosphatidylcholine (PSPC), dioleoylphosphatidylcholine (DOPC), dioleoylphosphatidylethanolamine (DOPE), dilauroylphosphatidylglycerol (DLPG), distearoylphosphatidylglycerol (DSPG), dimyristoylphosphatidylglycerol (DMPG), dip
  • a mixture of lipids may comprise each lipid at a desired molar or wt% ratio.
  • the ratio between respective lipids may be between 20: 1 to 1 : 1, including 15: 1 12: 1, 10: 1, 7: 1, 5: 1, 4: 1, 3 : 1 and 2: 1 or any ratio between these stated values.
  • a liposome may be formed using a molar ratio of 7 dipalmitoyl-sn-glycero-3-phosphocholine (DPPC): 2 Cholesterol (CHOL): 1 L-a-phosphatidylglycerol (PG).
  • DPPC dipalmitoyl-sn-glycero-3-phosphocholine
  • PG 1 L-a-phosphatidylglycerol
  • the lipid vesicle further comprises a carrier protein.
  • the carrier protein is immunogenic, or at least partly facilitates or enhances the immunogenicity of the immieuxic agent.
  • the carrier protein is formulated with the lipid vesicle so that the carrier protein is located inside the lipid vesicle within the internal aqueous space.
  • the carrier protein is separate to the immunogenic protein fragment, variant or derivative thereof. According this embodiment, the immunogenic protein fragment, variant or derivative thereof may be displayed on the vesicle surface.
  • the carrier protein may be fused, conjugated or complexed with said immunogenic protein fragment, variant or derivative thereof.
  • said immunogenic protein fragment, variant or derivative thereof is located inside the lipid vesicle within the internal aqueous space, fused, conjugated or complexed with the carrier protein.
  • This embodiment may be particularly suitable for oral delivery of the immunogenic agent, such as in liposomes comprising a bile salt, as will be described in more detail hereinafter.
  • Non-limiting examples of carrier proteins include diptheria toxoid (DT), tetanus toxoid (TT), CRM proteins such as CRM197 and Pertussis toxin mutant, although without limitation thereto. Also contemplated are fragments and variants of carrier proteins. In one particular embodiment, the carrier protein is diptheria toxoid (DT), or a fragment thereof.
  • the lipid vesicle further comprise an activator of innate immunity.
  • the activator of innate immunity may target a C-type lectin expressed by one or more cells that are associated with innate immunity.
  • a preferred C-type lectin is macrophage inducible Ca 2+ -dependent (C-type) lectin ("Mincle").
  • Non-limiting examples include a glycolipid such as the mycobacterial cord factor trehalose-6,6'-dimycolate (TDM) and/or its synthetic analogue trehalose-6,6'-dibehenate (TDB) and/or lipid A glycolipid adjuvants such as monophosphoryl 3-deacyl lipid A which mnay be in the form of PHAD®, 3D- PHAD® and 3D (6-acyl) PHAD®.
  • TDM mycobacterial cord factor trehalose-6,6'-dimycolate
  • TDB synthetic analogue trehalose-6,6'-dibehenate
  • lipid A glycolipid adjuvants such as monophosphoryl 3-deacyl lipid A which mnay be in the form of PHAD®, 3D- PHAD® and 3D (6-acyl) PHAD®.
  • activators of innate immunity such as described above may enhance or improve mucosal immunity elicited by the immuno
  • the lipid vesicle may further comprise a bile acid or bile salt.
  • Bile acids are typically dihydroxylated or trihydroxylated steroids (in some embodiments comprising 24 carbons), including cholic acid, deoxycholic acid, chenodeoxycholic acid and ursodeoxycholic acid.
  • the lipid vesicle comprises a bile salt such as a cholate, deoxycholate, chenodeoxycholate or ursodeoxycholate salt.
  • a preferred bile salt is sodium deoxycholate.
  • immunogenic agents comprising liposomes may be produced at a particular, selected or desired particle size or size range.
  • larger particle size liposomes may elicit a stronger mucosal immune response.
  • immunogenic agents comprising liposomes may be freeze-dried or lyophilized to facilitate longer term storage. Reconstituted freeze- dried liposomal immunogenic agents elicited an immune response comparable to that of "fresh" liposomal immunogenic agents
  • the pathogen is group A streptococcus.
  • group A streptococcus As used herein the terms "group A streptococcus”, “Group A Streptococci”, “Group A “Group A Strep” and the abbreviation “GAS” refer to streptococcal bacteria of Lancefield serogroup A which are gram positive ⁇ -hemolytic bacteria of the species Streptococcus pyogenes.
  • An important virulence factor of GAS is M protein, which is strongly anti-phagocytic and binds to serum factor H, destroying C3-convertase and preventing opsonization by C3b.
  • M protein An important virulence factor of GAS is M protein, which is strongly anti-phagocytic and binds to serum factor H, destroying C3-convertase and preventing opsonization by C3b.
  • Group A streptococci Diseases and conditions caused by group A streptococci include cellulitis, erysipelas, impetigo, scarlet fever, throat infections such as acute pharyngitis ("strep throat"), bacteremia, toxic shock syndrome, necrotizing fasciitis, acute rheumatic fever and acute glomerulonephritis, although without limitation thereto.
  • strep throat acute pharyngitis
  • bacteremia bacteremia
  • toxic shock syndrome necrotizing fasciitis
  • acute rheumatic fever acute glomerulonephritis
  • neutrophils or neutrophil granulocytes are cells that form part of the polymorphonuclear cell family (PMNs) together with basophils and eosinophils.
  • Neutrophils are relatively short-lived phagocytic cells formed from bone marrow stem cells and typically constitute 40% to 75% of white blood cells in mammals.
  • phagocytic neutrophils release soluble anti- microbials (e.g granule proteins) and generate neutrophil extracellular traps.
  • Neutrophils are responsive to molecules such as interleukin-8 (IL-8), C5a, fMLP and leukotriene B4 which promote neutrophil chemotaxis to sites of injury and/or acute inflammation.
  • the immunogenic protein may be an M protein, fragment or variant thereof.
  • an "protein fragment” is any fragment of a GAS M protein that is immunogenic and/or is capable of being bound by an antibody or antibody fragment.
  • the fragment is, comprises, or is contained within an amino acid sequence of a C-repeat region of a GAS M protein, or a fragment thereof.
  • Non-limiting examples include pi 45 which is a 20mer with the amino acid sequence with the amino acid sequence LRRDLDASREAKKQVEKALE (SEQ ID NO:4).
  • fragments of the pl45 amino acid sequence may be present in a J8 peptide.
  • J8 peptide is a peptide which comprises an amino acid sequence at least partly derived from, or corresponding to, a GAS M protein C- region peptide.
  • J8 peptide suitably comprises a conformational B-cell epitope and lacks potentially deleterious T-cell autoepitopes.
  • a preferred J8 peptide amino acid sequence is QAEDKVKQSREAKKQVEKALKQLEDKVQ (SEQ ID NO: 1) or a fragment or variant thereof, wherein the bolded residues correspond to residues 344 to 355 of the GAS M protein.
  • J8 is a chimeric peptide that further comprises flanking GCN4 DNA-binding protein sequences which assist maintaining the correct helical folding and conformational structure of the J8 peptide.
  • the immunogenic protein may be an agent that facilitates restoring or enhancing neutrophil activity.
  • an "agent that facilitates restoring or enhancing neutrophil activity” is a molecule that directly or indirectly at least partly increases, enhances or restores the production, migration and/or chemotaxis of neutrophils and/or one or more immunological activities of neutrophils.
  • the agent elicits an immune response to a neutrophil inhibitor.
  • the agent binds and at least partly inactivates the neutrophil inhibitor.
  • the neutrophil inhibitor may be a molecule derived or originating from Group A Streptococcal bacteria.
  • the neutrophil inhibitor is a serine protease, or a fragment thereof, that proteolytically cleaves interleukin 8.
  • the neutrophil inhibitor is SpyCEP or a fragment thereof.
  • SpyCEP is a 170-kDa multidomain serine protease expressed on the surface of the human pathogen Streptococcus pyogenes, which plays an important role in infection by catalyzing cleavage and inactivation of the neutrophil chemoattractant interleukin-8.
  • Non-limiting examples of SpyCEP amino acid sequences may be found under accession numbers YP597949.1 and (S. pyogenes MGAS10270) and YP596076.1 (S. pyogenes MGAS9429).
  • the SpyCEP fragment is, or comprises, the amino acid sequence set forth in SEQ ID NO:2 (NSDNIKENQFEDFDED WENF) . It is proposed that SEQ ID NO: 2 is, or comprises, the dominant epitope on SpyCEP that can induce functional antibodies.
  • a chimeric peptide comprising an M-protein amino acid sequence and a SpyCEP amino acid sequence that form a single, contiguous amino acid sequence.
  • the M-protein amino acid sequence may be located C- terminal to the SpyCEP amino acid sequence, or vice versa.
  • the chimeric peptide may comprise the amino acid sequence NSDNIKENQFEDFDED WENFQAEDKVKQSREAKKQVEKALKQLEDKVQ
  • respective liposomes comprising an M- protein amino acid sequence and a SpyCEP amino acid sequence may be produced for administration as an "admixture".
  • a variant M protein or peptide may comprise one or a plurality of lysine residues at an N and/or C-terminus thereof.
  • the plurality of lysine residues e.g polylysine
  • J8 peptide variants include:
  • a model a-helical coiled coil peptide has been based on the structure of the GCN4 leucine zipper (O'Shea et al., 1991).
  • the first heptad contains the sequence MKQLEDK (SEQ ID NO:9), which includes several of the features found in a stable coiled coil heptad repeat motif (a-b-c-d-e-f-g)n (Cohen & Parry, 1990).
  • the GCN4 peptide also contains a consensus valine in the a position. It has also been noted that when positions a and d are occupied by V and L a coiled coil dimer is favoured (Harbury et al, 1994).
  • a model heptad repeat was derived from these consensus features of the GCN4 leucine zipper peptide: (VKQLEDK; SEQ ID NO: 10) with the potential to form a a-helical coiled coil.
  • This peptide became the framework peptide.
  • Overlapping fragments of a conformational epitope under study were embedded within the model coiled coil peptide to give a chimeric peptide.
  • Amino acid substitutions, designed to ensure correct helical coiled coil conformations (Cohen & Parry, 1990) were incorporated into the chimeric peptides whenever an identical residue was found in both the helical model peptide and the epitope sequence.
  • lipid core peptide may comprise a plurality of J8 peptides (e.g four J8 peptides) synthesized directly onto two amino groups of each lysine of a branched polylysine core coupled to a lipophilic anchor.
  • the M protein fragment or variant and/or the SpyCEP fragment or variant may be derivatized to comprise one or more lipids that facilitate anchoring to the lipid bilayer as hereinbefore described.
  • a chimeric peptide comprising an M-protein amino acid sequence and a SpyCEP amino acid sequence (e.g. SEQ ID NO:3) may comprise a spacer amino acid sequence at the N-terminus thereof.
  • a SpyCEP fragment or variant may be separately lipidated along with the M protein fragment or variant or may be present as a chimeric peptide which is lipidated.
  • the pathogen is influenza virus.
  • the immunogenic protein, fragment or variant is of influenza A virus.
  • the immunogenic protein or fragment may be matrix protein 2, or a fragment thereof.
  • a non-limiting example is, or comprises, the amino acid sequence M SLLTE VETPIRNEWGCRCND S SD (SEQ ID NO:4).
  • the immunogenic protein, fragment or variant is of influenza B virus.
  • the immunogenic protein or fragment may be a haemagglutinin protein, or a fragment thereof.
  • a non-limiting example is, or comprises, the amino acid sequence PAKLLKERGFFGAIAGFLE (SEQ II) NO.5).
  • the pathogen is rhinovirus.
  • the immunogenic protein, fragment or variant is of a rhinovirus B protein, such as a capsid protein.
  • a non-limiting example is, or comprises, the amino acid sequence GAQVSTQKSGSHENQNILTNGSNQTFTVINY (SEQ ID NO: 6).
  • the immunogenic protein, fragment or variant is of a rhinovirus A protein, such as a capsid protein.
  • Another non- limiting example is, or comprises, the amino acid sequence GAQVSRQNVGTHSTQNMVSNGSSL (SEQ ID NO: 7).
  • the pathogen is a worm, such as a hookworm.
  • the immunogenic protein, fragment or variant is of Necator americanus.
  • a non-limiting example is, or comprises, the amino acid sequence T SLI AGLK AQ VE AIQKYIGAEL (SEQ ID NO:8).
  • the isolated immunogenic proteins, fragments and/or derivatives of the present invention may be produced by any means known in the art, including but not limited to, chemical synthesis, recombinant DNA technology and proteolytic cleavage to produce peptide fragments.
  • Chemical synthesis is inclusive of solid phase and solution phase synthesis. Such methods are well known in the art, although reference is made to examples of chemical synthesis techniques as provided in Chapter 9 of SYNTHETIC VACCINES Ed. Nicholson (Blackwell Scientific Publications) and Chapter 15 of CURRENT PROTOCOLS IN PROTEIN SCIENCE Eds. Coligan et al, (John Wiley & Sons, Inc. NY USA 1995-2008). In this regard, reference is also made to International Publication WO 99/02550 and International Publication WO 97/45444.
  • Recombinant proteins may be conveniently prepared by a person skilled in the art using standard protocols as for example described in Sambrook et al, MOLECULAR CLONING. A Laboratory Manual (Cold Spring Harbor Press, 1989), in particular Sections 16 and 17; CURRENT PROTOCOLS IN MOLECULAR BIOLOGY Eds. Ausubel et al., (John Wiley & Sons, Inc. NY USA 1995-2015), in particular Chapters 10 and 16; and CURRENT PROTOCOLS IN PROTEIN SCIENCE Eds. Coligan et al., (John Wiley & Sons, Inc. NY USA 1995-2015), in particular Chapters 1, 5 and 6.
  • recombinant protein preparation includes expression of a nucleic acid encoding the protein in a suitable host cell.
  • the invention provides immunogenic agents and/or use thereof for preventing or treating a pathogen-associated disease, disorder or condition in a mammal or other animal.
  • treating refers to a therapeutic intervention that at least partly ameliorates, eliminates or reduces a symptom or pathological sign of a pathogen-associated disease, disorder or condition after it has begun to develop. Treatment need not be absolute to be beneficial to the subject. The beneficial effect can be determined using any methods or standards known to the ordinarily skilled artisan.
  • preventing refers to a course of action initiated prior to infection by, or exposure to, a pathogen or molecular components thereof and/or before the onset of a symptom or pathological sign of the disease, disorder or condition, so as to prevent infection and/or reduce the symptom or pathological sign. It is to be understood that such preventing need not be absolute to be beneficial to a subject.
  • a “prophylactic” treatment is a treatment administered to a subject who does not exhibit signs of the disease, disorder or condition, or exhibits only early signs for the purpose of decreasing the risk of developing a symptom or pathological sign of the disease, disorder or condition.
  • the disease, disorder or condition may be a group A- strep-associated disease, disorder or condition.
  • group A-strep-associated disease, disorder or condition is meant any clinical pathology resulting from infection by group A strep and includes cellulitis, erysipelas, impetigo, scarlet fever, throat infections such as acute pharyngitis ("strep throat"), bacteremia, toxic shock syndrome, necrotizing fasciitis, acute rheumatic fever and acute glomerulonephritis, although without limitation thereto.
  • the uses for treatment and/or immunization disclosed herein include administration of the immunogenic agent comprising an M protein fragment, variant or derivative, a lipid vesicle, carrier protein and/or a SpyCEP peptide or other fragment that facilitates restoring or enhancing neutrophil activity to a mammal.
  • treatment and/or immunization may include, in addition, administration of antibodies or antibody fragments to therapeutically treat GAS infections, such as by targeting SpyCEP at the site of infection ⁇ e.g. the skin) and/or antibodies or antibody fragments that bind an M protein, fragment or variant thereof.
  • Antibodies and antibody fragments may be polyclonal or monoclonal, native or recombinant.
  • Antibody fragments include Fc, Fab or F(ab)2 fragments and/or may comprise single chain Fv antibodies (scFvs).
  • scFvs may be prepared, for example, in accordance with the methods described respectively in United States Patent No 5,091,513, European Patent No 239,400 or the article by Winter & Milstein, 1991, Nature 349:293.
  • Antibodies may also include multivalent recombinant antibody fragments, such as diabodies, triabodies and/or tetrabodies, comprising a plurality of scFvs, as well as dimerisation-activated demibodies ⁇ e.g.
  • Such antibodies may be prepared in accordance with the methods described in Holliger et al, 1993 Proc Natl Acad Sci USA 90 6444; or in Kipriyanov, 2009 Methods Mol Biol 562 177.
  • Well-known protocols applicable to antibody production, purification and use may be found, for example, in Chapter 2 of Coligan et al, CURRENT PROTOCOLS IN IMMUNOLOGY (John Wiley & Sons NY, 1991-1994) and Harlow, E. & Lane, D. Antibodies: A Laboratory Manual, Cold Spring Harbor, Cold Spring Harbor Laboratory, 1988.
  • polyclonal antibodies are well known to those skilled in the art. Exemplary protocols which may be used are described for example in Coligan et al, CURRENT PROTOCOLS IN IMMUNOLOGY, supra, and in Harlow & Lane, 1988, supra.
  • anti-SpyCEP polyclonal antibodies may be obtained or purified from human sera from individuals exposed to, or infected by, Group A strep.
  • polyclonal antibodies may be raised against purified or recombinant SpyCEP, or an immunogenic fragment thereof, in production species such as horses and then subsequently purified prior to administration.
  • Monoclonal antibodies may be produced using the standard method as for example, originally described in an article by Kohler & Milstein, 1975, Nature 256, 495, or by more recent modifications thereof as for example, described in Coligan et al, CURRENT PROTOCOLS IN IMMUNOLOGY, supra by immortalizing spleen or other antibody producing cells derived from a production species which has been inoculated with one or more of the isolated proteins, fragments, variants or derivatives of the invention. Accordingly, monoclonal antibodies may be raised against an M protein fragment, variant or derivative and/or the agent that facilitates restoring or enhancing neutrophil activity (e.g SpyCEP) for use according to the invention.
  • an M protein fragment, variant or derivative and/or the agent that facilitates restoring or enhancing neutrophil activity e.g SpyCEP
  • the monoclonal antibody or fragment thereof may be in recombinant form. This may be particularly advantageous for "humanizing" the monoclonal antibody or fragment if the monoclonal antibody is initially produced by spleen cells of a non- human mammal.
  • a preferred M protein fragment may be a p 145 peptide.
  • a preferred fragment of SpyCEP for antibody production may comprise or consist of the amino acid sequence NSDNIKENQFEDFDEDWENF (SEQ ID NO:2).
  • the disease, disorder or condition may be an influenza virus-associated disease, disorder or condition.
  • Influenza virus may cause a transmissable or otherwise infectious disease known as the "flu”. Typical symptoms include fever, headache, coughing, lethargy, respiratory and nasopharyngeal mucous build-up and secretionm muscular pain, nausea and vomiting. Symptoms may last for a few days or persist for several weeks. In some cases, secondary respiratory bacterial infections may arise, in some cases causing severe conditions such as pneumonia. Accordingly, the immunogenic agents and/or methods of the invention may treat or prevent influenza virus-associated disease, disorder or conditions such as described above.
  • the disease, disorder or condition may be a rhinovirus-associated disease, disorder or condition.
  • Rhinoviruses e.g. Rhinovirus A and Rhinovirus B
  • Rhinoviruses are species in the genus Enterovirus of the Picornaviridae family of viruses. Rhinoviruses are typically the causative agents of the common cold, the symptoms of which are similar to influenza but generally less severe and with a lower probability of secondary bacterial infections such as pneumonia.
  • the immunogenic agents and/or methods of the invention may treat or prevent rhinovirus-associated disease, disorder or conditions such as described above.
  • the disease, disorder or condition may be a hookworm-associated disease, disorder or condition.
  • Hookworms are nematode worms that infest a variety of different animals. Hookworms that typically infect humans may include Necator americanus and Ancylostoma duodenalis. Hookworms have hook-like mouthparts with attach the hookworm to the wall of the gut, puncturing the blood vessels and feeding on blood, leading in some cases to severe anaemia. Hookworm infection in pregnancy can cause retarded growth of the fetus, premature birth and a low birth weight. Hookworms in children can cause intellectual, cognitive and growth problems.
  • the immunogenic agents and/or methods of the invention may treat or prevent worm-associated disease, disorder or conditions such as described above.
  • the aforementioned methods may be performed as follows:
  • the immunogenic agent may be administered in the form of a composition.
  • composition may comprise:
  • an immunogenic agent comprising one or a plurality of different proteins, fragments, variants or derivatives of a same or single pathogen
  • composition comprises an acceptable carrier, diluent or excipient.
  • acceptable carrier diluent or excipient
  • a solid or liquid filler diluent or encapsulating substance that may be safely used in systemic administration.
  • a variety of carriers, diluent and excipients well known in the art may be used.
  • These may be selected from a group including sugars, starches, cellulose and its derivatives, malt, gelatine, talc, calcium sulfate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline and salts such as mineral acid salts including hydrochlorides, bromides and sulfates, organic acids such as acetates, propionates and malonates, water and pyrogen- free water.
  • immunostimulant includes within its scope carriers, delivery agents, immunostimulants and/or adjuvants as are well known in the art.
  • immunostimulants and adjuvants refer to or include one or more substances that enhance the immunogenicity and/or efficacy of a formulation.
  • Non-limiting examples of suitable immunostimulants and adjuvants include squalane and squalene (or other oils of plant or animal origin); block copolymers; detergents such as Tween®-80; Quil® A, mineral oils such as Drakeol or Marcol, vegetable oils such as peanut oil; Corynebacterium-derived adjuvants such as Corynebacterium parvum Propionibacterium-denwed adjuvants such as Propionibacterium acne] Mycobacterium bovis (Bacille Calmette and Guerin or BCG); Bordetella pertussis antigens; tetanus toxoid; diphtheria toxoid; surface active substances such as hexadecylamine, octadecylamine, octadecyl amino acid esters, lysolecithin, dimethyldioctadecylammonium bromide, N,N-dicoctadecy
  • Immunological agents may include carrier proteins such as thyroglobulin; albumins such as human serum albumin; toxins, toxoids or any mutant crossreactive material (CRM) of the toxin from tetanus, diphtheria, pertussis, Pseudomonas, E.
  • carrier proteins such as thyroglobulin; albumins such as human serum albumin; toxins, toxoids or any mutant crossreactive material (CRM) of the toxin from tetanus, diphtheria, pertussis, Pseudomonas, E.
  • coli coli, Staphylococcus, and Streptococcus; polyamino acids such as poly(lysine: glutamic acid); influenza; Rotavirus VP6, Parvovirus VP1 and VP2; hepatitis B virus core protein; hepatitis B virus recombinant vaccine and the like.
  • a fragment or epitope of a carrier protein or other immunogenic protein may be used.
  • a T cell epitope of a bacterial toxin, toxoid or CRM may be used.
  • U.S. Patent No 5,785,973 which is incorporated herein by reference.
  • Any suitable procedure is contemplated for producing vaccine formulations.
  • Exemplary procedures include, for example, those described in New Generation Vaccines (1997, Levine et al, Marcel Dekker, Inc. New York, Basel, Hong Kong), which is incorporated herein by reference.
  • Any safe route of administration may be employed, including intranasal, oral, rectal, parenteral, sublingual, buccal, intravenous, intra-articular, intramuscular, intra-dermal, subcutaneous, inhalational, intraocular, intraperitoneal, intracerebroventricular, topical, mucosal and transdermal administration, although without limitation thereto.
  • Dosage forms include tablets, dispersions, suspensions, injections, solutions, syrups, troches, capsules, nasal sprays, suppositories, aerosols, transdermal patches and the like. These dosage forms may also include injecting or implanting controlled releasing devices designed specifically for this purpose or other forms of implants modified to act additionally in this fashion. Controlled release may be effected by coating with hydrophobic polymers including acrylic resins, waxes, higher aliphatic alcohols, polylactic and polyglycolic acids and certain cellulose derivatives such as hydroxypropylmethyl cellulose.
  • compositions may be presented as discrete units such as capsules, sachets, functional foods/feeds or tablets each containing a pre-determined amount of one or more therapeutic agents of the invention, as a powder or granules or as a solution or a suspension in an aqueous liquid, a non-aqueous liquid, an oil-in- water emulsion or a water-in-oil liquid emulsion.
  • Such formulations may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association one or more agents as described above with the carrier which constitutes one or more necessary ingredients.
  • the formulations are prepared by uniformly and intimately admixing the agents of the invention with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
  • the above formulations may be administered in a manner compatible with the dosage formulation, and in such amount as effective.
  • the dose administered to a patient in the context of the present invention, should be sufficient to effect a beneficial response in a patient over an appropriate period of time.
  • the quantity of agent(s) to be administered may depend on the subject to be treated inclusive of the age, sex, weight and general health condition thereof, factors that will depend on the judgement of the practitioner.
  • the composition is suitable for intranasal administration to a subject.
  • the terms "patient”, “individuaF and “subject” are used in the context of any mammalian recipient of a treatment or formulation disclosed herein. Accordingly, the methods and formulations disclosed herein may have medical and/or veterinary applications. In a preferred form, the mammal is a human.
  • GAS Group A streptococcus
  • URT ulcerative colitis
  • RF rheumatic fever
  • RHD rheumatic heart disease
  • GAS vaccine candidates can be divided into M protein and non M protein- based vaccines (5).
  • the cell surface M protein a coiled-coil protein consisting of 3 major domains, is a major virulence determinant (6).
  • This protein consists of a hypervariable amino-terminal and A-repeat domains used for epidemiologic molecular typing (emm or M typing); a B-repeat domain and a conserved C-repeat domain (6).
  • the leading subunit vaccines under clinical investigations are the amino-terminal M protein-based multivalent vaccines and conserved C-repeat M protein peptide vaccines (5).
  • These GAS vaccine candidates have entered into clinical trials based on their success in inducing systemic immunity (7).
  • J8 a vaccine candidate peptide, J8, based on a minimal B cell epitope from the conserved C3 -repeat domain of the M protein (12).
  • the J8 peptide (QAEDKVKQSREAKKQVEKALKQLEDKVQ; SEQ ID NO: l) is a chimeric peptide that contains 12 amino acids from the C-region (shown in bold) flanked by GCN4 DNA-binding protein sequences to maintain the correct helical conformational structure (13).
  • DT diphtheria toxoid
  • J8 induces IgG antibodies that protect mice from systemic and skin challenge with multiple GAS strains (4, 13).
  • Liposomes are spherical vesicles composed of biocompatible phospholipid bilayers and can be loaded with and deliver both hydrophilic and hydrophobic molecules (19). Liposomes have been administered safely to humans via the intranasal route (20, 21). However, liposomes that present peptide antigens are not ideal platforms to induce peptide-specific antibody responses. Peptides can only contain limited antigenic epitopes capable of activating helper T cells necessary for an antibody response by B cells. They require conjugation to 'carrier' proteins to render them immunogenic in an outbred population and as such are not ideally suited for presentation by liposomes.
  • particulates such as liposome are unsurprising, as natural pathogens are also particulates and are well-recognised by the immune system (22).
  • the natural tendency of liposomes to interact with antigen presenting cells has served as the primary rationale for using liposomes to present antigens to the immune system (23).
  • the aim of this study was to develop a J8-based liposomal formulation (in the absence of an adjuvant) where a lipophilic J8 construct was incorporated within the lipid bilayers and a hydrophilic carrier protein (DT) was encapsulated in the inner aqueous core.
  • DT hydrophilic carrier protein
  • mice All animal protocols used were approved by the Griffith University Research Ethics Review Board for Animal-Based Work, GU Ref No: GLY/1914/AEC. This study was carried out in strict accordance with the National Health and Medical Research Council (NHMRC) of Australia guidelines for laboratory animals. Methods were chosen to minimize pain and distress to the mice and animals were observed daily by trained animal care staff. Mice were terminated using a C0 2 inhalation chamber.
  • Human blood With written informed consent, blood was obtained from donors by a phlebotomist at the Griffith university health centre. Approval of the study was granted by the Griffith University Human Research Ethics Committee (GU HREC, Protocol # GLY/03/14/HREC). Samples were de-identified prior to handling by laboratory personnel.
  • J8-Lipo-DT formulation To promote noncovalent complexing of J8 to liposome bilayer, a hydrophobic anchor consisting of two palmitic acid (CI 6) was added to the epsilon and primary amine group of the lysine in a tripeptide spacer (consisting of Lys Ser Ser) present in J8 amino-terminus (C16-C16-KSSJ8).
  • This construct was manufactured by Chinapeptides Co., Ltd. (Shanghai, China).
  • the expected molecular weight of the construct (MW 4061.97 g/mol) was confirmed by ESI-MS, and the product obtained at greater than 95% purity (by analytical RP-HPLC area under the curve analysis).
  • Liposomes were prepared using the thin film hydration method (42). Lipids from Avanti Polar Lipids, Inc. (Alabaster, United States) were used at a molar ratio of 7 dipalmitoyl-sn-glycero-3- phosphocholine (DPPC): 2 Cholesterol (CHOL): 1 L-a-phosphatidylglycerol (PG). Lipids in chloroform (CHC1 3 ) solution were coated onto round-bottom flasks using a rotary evaporator along with predetermined amount of C16-C16- KSSJ8.
  • DPPC dipalmitoyl-sn-glycero-3- phosphocholine
  • PG 1 L-a-phosphatidylglycerol
  • the volumes used were 0.7 ml of DPPC (10 mg/ml) in CHC1 3 , 0.2 ml of CHOL (5 mg/ml) in CHC1 3 , and 0.1 ml of PG (10 mg/ml) in CHC1 3 .
  • the lipid thin film was then hydrated and dispersed in 1 mL of phosphate buffer saline (PBS) containing a predetermined amount of DT by vigorous mixing at room temperature.
  • PBS phosphate buffer saline
  • the resultant liposomal suspension was centrifuged at 16, 162 g for 10 min, the supernatant removed and the liposome pellet resuspended in an appropriate volume of PBS to be administered in mice.
  • the supernatant was collected and the amount of unentrapped DT in supernatant was determined using a NanoDrop 2000 UV-Vis Spectrophotometer (Thermo Scientific, Massachusetts, United States). Subtraction of supernatant DT concentration from starting DT concentration in PBS used for rehydration of lipids to produce liposomes allowed quantification of encapsulation efficiency.
  • the average particle size (nm) of liposome was measured at 25°C using a Nanosizer (Zetasizer Nano Series ZS, Malvern Instruments, United Kingdom) with disposable capillary cuvettes. Size was analysed using a noninvasive backscatter system and measurements taken with a 173° scattering angle.
  • mice Intranasal immunization of mice.
  • B10.BR and BALB/c mice (Animal Resources Centre, Western Australia, Australia) to be immunized were anesthetized by use of a mixture of xylazine and ketamine (1 : 1 : 10 mixture of xylazine: ketamine: H 2 0).
  • Mice were administered 30 ⁇ g of J8-Lipo-DT alone in a total volume of 20 ⁇ _, PBS (10 ⁇ ) whilst control mice were administered 20 ⁇ _, of PBS (10 ⁇ ).
  • Positive control mice received 30 ⁇ g of J8 conjugated to DT, co-administered with 10 ⁇ g of CTB (Sigma Aldrich, St.
  • mice received 2 booster immunizations 21 days apart in the same fashion as the primary immunization.
  • Other control groups received equivalent amounts of J8, DT or liposome alone as described above.
  • Serum, saliva and fecal sample collection Serum was collected on days 20, 40, and 60 after primary immunization to determine the level of J8-specific systemic antibodies. Blood was collected from mice via the tail artery and allowed to clot for at least 30 min at 37 °C. Serum was collected after centrifugation for 10 min at
  • mice were administered 50 ⁇ ⁇ of a 0.1% solution of pilocarpine to induce salivation. Saliva was then collected in eppendorf tubes containing 2 ⁇ _, of 50 mmol/L phenylmethylsulfonyl fluoride (PMSF) protease inhibitor (Sigma).
  • PMSF phenylmethylsulfonyl fluoride
  • ELISA enzyme linked immunosorbent assay
  • Samples were serially diluted down the plate in 0.5% skim milk PBS-Tween 20 buffer, starting at an initial dilution of 1 : 100 to a final dilution of 1 : 12,800 for sera and 1 :2 to 1 :256 for saliva/ fecal samples. Each sample was diluted to a final volume of 100 ⁇ and incubated for 1.5 h at 37°C. The plates were washed 5 times and peroxidase conjugated goat anti-mouse IgG or IgA (Invivogen, San Diego, United States) were added at a dilution of 1 :3000 or 1 : 1000 respectively in 0.5% skim milk PBS-Tween20 for 1.5 h at 37°C.
  • OPD substrate Sigma Aldrich
  • 100 ⁇ of OPD substrate was added according to the manufacturer's instructions and incubated at room temperature for 30 min in the dark.
  • the absorbance was measured at 450 nm in a Victor 3 1420 multilabel counter (Perkin Elmer Life and Analytical Sciences, Shelton, United States).
  • the titer was described as the lowest dilution that gave an absorbance of >3 standard deviation (SD) above the mean absorbance of negative control wells (containing normal mouse serum immunized with PBS).
  • SD standard deviation
  • Statistical significance p ⁇ 0.05 was determined using a one-way analysis of variance (ANOVA) with Tukey post hoc test using GraphPad Prism 5 software (GraphPad, California, United States). Procedure for GAS challenge.
  • mice Immunized and control mice were challenged intranasally with a predetermined dose of the GAS strain Ml on day 63 after primary immunization.
  • the GAS strain Ml had been serially passaged in mouse spleen to enhance virulence, and made streptomycin-resistant to enable GAS to be distinguished in throat swabs from normal murine bacterial flora (44).
  • throat swabs were obtained from mice on days 1-3 after challenge. The throat swabs were streaked out on Todd-Hewitt agar plates containing 2% defibrinated horse blood and incubated overnight at 37°C.
  • the resultant DC population was resuspended in RPMI 1640 (Gibco, Gaithersburg, United States) complete media (with 2 mM 1- glutamine, 1% non-essential amino acids, 1 % Pen-strep, 10 mM HEPES) supplemented with 10% FCS (Gibco).
  • DCs (2 X 10 6 ) in a total volume of 0.2 mL was plated out and the following stimuli were added as indicated: pIC (Invivogen, San Diego, United States) at 10 ⁇ g/mL, J8-Lipo-DT (150 ⁇ g/mL) or complete media alone for 24 hours. Supernatants were collected after 24 h and stored at - 20°C.
  • the resultant populations were assessed by flow cytometric analysis using the following antibodies (Becton Dickinson): anti-HL A-DR- V450, -CDlc-APC, -CD80-PE- Cy7, -CD83-PE-TexasRed, -CD86-PE, -CD123-Percp-5.5, -CD141-APC-Cy7.
  • Anti-HL A-DR- V450, -CDlc-APC, -CD80-PE- Cy7, -CD83-PE-TexasRed, -CD86-PE, -CD123-Percp-5.5, -CD141-APC-Cy7 After staining with the appropriate antibodies in the dark at 4°C for 30 min, cells were washed twice in PBS, and fixed in 1% paraformaldehyde. Gating was on large granular cells, and 2000-5000 gated events were collected from each sample.
  • ULA-DR positive cells were gated to define human DCs and further subdivided into the CD141+ conventional DCs type 1, CDlc+ DCs conventional (myeloid) DCs type 2 and CD123+ plasmacytoid DCs according to previously established methods (45).
  • Mean fluorescence intensity (MFI) values were determined on the gated population. The data are reported as means + SEM, and differences were analysed with GraphPad Prism 5 software (GraphPad, California, United States) using Student's t test. P values under 0.05 were considered as significant.
  • CBA cytometric bead array
  • flow cytometry analysis were used to quantify the pro- inflammatory response produced by splenocytes after stimulation with the J8 peptide.
  • CBA cytometric bead array
  • Single-cell suspensions of spleens from J8-Lipo-DT immunized mice free of erythrocytes were prepared in RPMI 1640 media.
  • Splenocytes (4 x 10 5 )in a total volume of 0.1 mL was plated out and the following stimuli were added as indicated: LPS (Sigma Aldrich) at 2 ⁇ g/mL, J8 (10 ⁇ g/mL) or RPMI 1640 media alone for 72 h.
  • Supernatants were isolated After 72 h and stored at - 80°C for CBA flow cytometric analysis.
  • Liposomes with surface-associated J8 peptide and containing diphtheria toxoid were constructed as described in Materials and Methods.
  • the administered formulation contained 30 ⁇ g of J8 per dose linked to the liposome surface using a palmitic acid-based moiety.
  • liposomes contained 30 ⁇ g of DT per dose.
  • mice Using a primary and 2-boost regimen, BALB/c mice (10 per group) were immunized intranasally with J8-Lipo-DT and various controls: liposomes alone (Lipo); liposomes encapsulating DT (Lipo-DT); J8 embedded on the surface of liposomes but without encapsulated DT (J8-Lipo); J8-DT+CTB; PBS+CTB; and PBS.
  • mice vaccinated intranasally were then challenged intra-nasally with the pharyngeal isolate Ml GAS strain obtained from a patient with scarlet fever (16).
  • J8-Lipo-DT induced higher J8-specific IgA (fecal and salivary) and serum IgG titers than J8-Lipo.
  • J8-Lipo-DT was observed to be superior for the salivary IgA response, the fecal IgA response and the serum IgG response (Fig.
  • J8-DT+CTB-immunized mice were not protected from colonization of the throat or NALT whereas J8-Lipo-DT-immunized mice showed significant protection against colonization in both compartments (Fig. 3 B and C). Protection due to J8-Lipo-DT was significantly better than that induced by J8-Lipo.
  • Murine NALT is a portal of entry for persistent GAS infection (24), and is a functional homologue of human tonsils (25).
  • J8-Lipo-DT and J8-DT+CTB immunized mice had undetectable bio-burden in nasal shedding (Fig. 5A).
  • the data also demonstrated an absence of bacteria in J8-Lipo-DT immunized mice by day 2 post-challenge for throat swabs, whereas J8-DT+CTB immunised mice still had detectable level of GAS in throat swabs on day 3 (Fig. 5 ⁇ ).
  • the cytokine response of spleen cells from intranasally immunized mice was measured to determine whether intranasal immunization induced a systemic cellular immune response that may explain the self adjuvanticity of J8-Lipo-DT and the switch of the isotype of antibodies to IgA.
  • Pro-inflammatory cytokines gamma interferon [IFN- ⁇ ], interleukin 1 [IL-1], IL-6, IL-12p70, monocyte chemotactic protein 1 [MCP-1], and tumor necrosis factor alpha [TNF-a]
  • mice immunized with J8-Lipo-DT were sacrificed and splenocytes were stimulated with J8, LPS or media.
  • Other cytokines assessed were not detected.
  • IL-6 is known to be responsible for the switch of the antibody response to IgA (27).
  • the chemo-attractant MCP-1 is known to play a major role in GAS defence mechanisms (28).
  • dendritic cell subsets were isolated from the blood of three healthy volunteers and stimulated with J8-Lipo- DT.
  • Mature DCs are potent antigen presenting cells, expressing high levels of cell surface molecules involved in antigen presentation and co-stimulation that facilitate antigen recognition and cell-cell interactions.
  • the modulation of various cell surface molecules in response to J8- Lipo-DT was examined by flow cytometry (Fig. 8A-C).
  • the synthetic double- stranded RNA adjuvant, polyriboinosinic-polyribocytidylic acid (pIC) was used as a control (29).
  • Levels of the costimulatory molecules CD80, CD83 and CD86 were significantly higher on CD123+ plasmacytoid DCs (pDCs) cultured with J8- Lipo-DT (Fig. 8A).
  • Expression of CD80 was also increased in the two subsets of classical DCs (cDC), CD 141+ classical type 1 DCs and CDlc+ classical type 2 DCs (Fig. 8fi-C).
  • CD86 expression was also increased for CD141+ DCs (Fig. 8fi).
  • pro-inflammatory cytokines post stimulation were evaluated using the cytometric bead array.
  • Neutrophils are known to be crucial to IgA control of GAS infections (30).
  • Elevated levels of anti-inflammatory cytokine, IL-10 were also observed (Fig. 9). This is possibly due to the regulatory effects of IL-10 in DC maturation steps and counterbalancing host pro-inflammatory responses (31, 32).
  • J8+S2-Lipo-DT induced antigen specific IgA, IgG responses. Comparable immune responses were observed in response to J8-Lipo- DTand S2-Lipo-DT.
  • Different formulation strategies employing either (i) both epitopes in a liposome (J8+S2-Lipo-DT) or (ii) an admixture of J8/S2S2-Lipo-DT liposomes.
  • Mucosal immunization as a means of eliciting protective immunity against infectious diseases, has attracted much interest.
  • In practice it has often proven difficult to stimulate strong mucosal IgA immune responses and the progress in mucosal vaccination efforts using subunit peptide antigens has been disappointing. This is due in part to difficulty in stimulating potent immune responses in comparison to traditional whole-organism based approaches. Addition of an adjuvant and conjugation of subunit antigens to protein carriers as sources of T cell help have proven effective for systemic immunity.
  • induction of mucosal immunity requires novel strategies.
  • the topographical position of liposome associated antigens affects antigen processing and presentation to B and helper T cells (33). It has been demonstrated that antigens exposed on the liposome surface are preferentially processed and presented by B cells whilst liposome encapsulated antigen are more effectively processed and presented to T cells by antigen presenting cells (34).
  • the vaccine candidate J8-Lipo-DT represents a rational subunit liposomal vaccine design, ensuring B cell epitopes are liposome bilayer-associated to be exposed to bind to the Ig receptor of a B cell whilst encapsulation of DT allows effective delivery, processing and presentation to T cells.
  • liposomes have previously been reported to deliver encapsulated peptides to induce a cellular immune response, such liposomes do not induce an IgA nor an IgG response except in the presence of a potent adjuvant such as Lipid A (26, 35). It is possible that the lipid tail on J8 provided adjuvant activity thus contributing to the immunogenicity of J8-Lipo-DT; however, the J8 peptide with the lipid tail on its own was not immunogenic demonstrating the need for the liposome formulation. Self-adjuvanting immunostimulatory activities of liposomes have been previously reported and shown to be due to interaction with antigen presenting cells and induction of a pro-inflammatory response (36).
  • MCP-1 is a chemoattractant for lymphocytes, monocytes, and antigen presenting cells (37). Previously implicated in mediating mucosal inflammation, it has been reported as a potential mucosal adjuvant due to significantly increasing mucosal IgA secretion (37).
  • DCs are key elements bridging the innate and adaptive immune responses to infection (39). Mature DCs produce inflammatory cytokines, up-regulate costimulatory and antigen presenting molecules, and migrate to lymph nodes where they function as potent antigen presenting cells for naive T lymphocytes to initiate adaptive immune responses.
  • IL-6 plays a critical role in B cell terminal differentiation, and in mucosal sites it stimulates proliferation and secretion of IgA in mucosal sites (27).
  • IgA specific immunity against GAS requires the presence of neutrophils (30), IL-8 has a key role in the recruitment and activation of neutrophils.
  • human pDCs increased both maturation and costimulatory markers upon stimulation with J8-Lipo-DT.
  • Human pDCs readily phagocytose and process antigens entrapped in particulate delivery systems (40), indicating that particulate delivery systems can be used to facilitate efficient delivery of antigens to pDCs.
  • our results show for the first time the ability to stimulate human pDCs with a liposome-based particulate delivery system.
  • Human pDCs initially identified in the blood have subsequently been detected in the spleen, lymph nodes and mucosal sites including tonsils (41). Therefore, liposome-based vaccine delivery could potentially be exploited to target this DC subset for desired mucosal immune responses in humans.
  • Liposome extrusion was done with a heat block, 1 mL syringe mini- extruder (Avanti Polar Lipids). The rehydrated solution was passed eleven times through a 50 nm, 400 nm, 1000 mm filter (Avanti Polar Lipids) while the heat block was set at ⁇ 40°C. Liposome size measurement was performed by Nanosizer (Dynamic light scattering or DLS).
  • J8-Lipo-DT can be extruded to form nano- to micro-sized particles The majority of particle sizes had a narrow molecular- weight distribution (low polydispersity index of ⁇ 0.3).
  • the data shown in Figure 13 suggest that J8-Lipo-DT size does not influence systemic IgG response.
  • larger sized liposomes induced a J8-specific mucosal response.
  • Liposome thin films were rehydrated with milliQ water containing 10% Trehalose and then lyophilized. Post 1, 4 and 7 weeks lyophilization, J8-Lipo-DT powder was reconstituted in PBS.
  • Figure 15 shows the size results of liposome size measurement by Nanosizer (Dynamic light scattering or DLS). The majority of particle sizes had a narrow molecular-weight distribution (low polydispersity index of ⁇ 0.3).
  • Figure 16 shows that reconstituted, freeze-dried, J8-Lipo-DT liposomes induced J8- specific systemic response without additional adjuvant This was a comparable immune response to freshly made J8-Lipo-DT. Trehalose was important for the immunogenicity of freeze-dryed J8-Lipo-DT.
  • Figure 17 demonstrates that reconstituted, freeze-dried J8-Lipo-DT liposomes induced a J8-specific mucosal response
  • TDB glycolipid activators of innate immunity
  • 3D-PHAD® glycolipid activators of innate immunity
  • TDB is formulated with liposomes based on % of total phospholipids in liposomes. 9 mg of phospholipids are used and TDB is used at radio of 20% of this (1.8 mg).
  • Efficacy is measured by antibody titers post-immunization (IgA and IgG antibodies) and skin challenge experiment with a GAS strain. Data are shown in FIG. 19.
  • bile salt such as sodium deoxycholate
  • FIG. 20 An example of a liposome comprising the bile salt sodium deoxycholate is shown schematically in FIG. 20.
  • the immunogenic agent in this case J8 peptide
  • the carreier protein e.g. DT
  • Bile salt will be formulated in liposomes when phospholipids are hydrated to produce liposomes (bile salt containing liposomes are referred to as "bilosomes"). Preparation of bilosomes is as follows.
  • Sorbitan tristearate (150 mmol), cholesterol and dicetyl phosphate (DCP) in a molar ratio of 7:3 : 1 is dissolved in 10 mL chloroform in a round-bottomed flaskalong with 150 ⁇ g of J8 modified with palmitic acid moiety. Solvent is removed by rotary evaporator to form a thin film on the glass surface of a round- bottomed flask. The film is then hydrated with 3.5 mL PBS (pH 7.4), containing 100 mg of sodium deoxycholate (bile salt) along with 150 ⁇ g of diphtheria toxoid.
  • PBS pH 7.4
  • FIG. 21 shows a schematic depiction of the immunogenic agent comprising a single lipid vesicle having respective immunogens from each of influenza A, influenza B and group A streptococcus.
  • the results shown in FIG. 22 show that a lipid vesicle comprising respective immunogens from each of influenza A, influenza B and group A streptococcus (as shown in FIG 21 A) induced immunity against each of these pathogens in mice.
  • FIG. 2 IB shows the immunogenicity of an immunogenic agen comprising glycolipid adjuvants such as shown schematically in FIG. 2 IB.
  • a liposome-based, mucosally active GAS vaccine candidate is the first to report on a liposome-based, mucosally active GAS vaccine candidate.
  • Our findings are an important step toward overcoming current obstacles in the development of a GAS vaccine to prevent infection at mucosal sites and community dissemination.
  • the study provides important mechanistic insights into how liposomal particulate delivery systems can collectively induce the desired mucosal immune responses to combat GAS infection.
  • the strategy reported here is relevant to the development of subunit mucosal vaccines against other pathogenic organisms. Non-limiting examples include influenza virus, rhinovirus and hookworms as hereinbefore described.
  • a single lipid vesicle may comprise immunogens against a plurality of different pathogens.
  • Childers NK, Tong G, & Michalek SM (1997) Nasal immunization of humans with dehydrated liposomes containing Streptococcus mutans antigen. Oral microbiology and immunology 12(6):329-335.
  • Dutertre CA Wang LF, & Ginhoux F (2014) Aligning bona fide dendritic cell populations across species.
  • mice Protection of mice from group A streptococcal infection by intranasal immunisation with a peptide vaccine that contains a conserved M protein B cell epitope and lacks a T cell autoepitope.

Landscapes

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

Abstract

L'invention concerne un agent immunogène approprié pour la prévention, au traitement ou à l'immunisation contre un ou plusieurs pathogènes différents, qui comprend un agent immunogène comprenant une ou plusieurs protéines dérivées de pathogènes, ou fragments, variants ou dérivés de ces dernières, présentées en surface d'une vésicule lipidique, ainsi qu'une protéine à rôle de véhicule telle que l'anatoxine diphtérique, située dans un espace intravésiculaire. L'agent immunogène peut être approprié pour une administration intranasale et peut être apte à induire une réponse immunitaire muqueuse. L'agent immunogène peut de plus comprendre un activateur de l'immunité innée tel que le tréhalose-6,6'-dibéhénate et/ou un sel biliaire tel que le désoxycholate de sodium. Le ou les agents pathogènes pouvent être des streptocoques du groupe A, des virus ou des nématodes responsables d'ankylostomes.
PCT/AU2016/051005 2015-10-27 2016-10-26 Vaccin à base de liposomes WO2017070735A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201680074917.3A CN109069424B (zh) 2015-10-27 2016-10-26 脂质体疫苗

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
AU2015904403 2015-10-27
AU2015904403A AU2015904403A0 (en) 2015-10-27 Liposomal vaccine
AU2016901026 2016-03-18
AU2016901026A AU2016901026A0 (en) 2016-03-18 Liposomal vaccine
CN201610222100.8 2016-04-11
CN201610222100.8A CN106606775A (zh) 2015-10-27 2016-04-11 脂质体a群链球菌疫苗

Publications (1)

Publication Number Publication Date
WO2017070735A1 true WO2017070735A1 (fr) 2017-05-04

Family

ID=58615051

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2016/051005 WO2017070735A1 (fr) 2015-10-27 2016-10-26 Vaccin à base de liposomes

Country Status (2)

Country Link
CN (2) CN106606775A (fr)
WO (1) WO2017070735A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019036761A1 (fr) * 2017-08-23 2019-02-28 Griffith University Peptide immunogène dirigé contre le streptocoque de groupe a
WO2021041891A1 (fr) * 2019-08-30 2021-03-04 Emory University Utilisation d'acide désoxycholique, de dérivés ou de sels de ceux-ci dans la gestion d'infections bactériennes et compositions associées
WO2021102505A1 (fr) 2019-11-25 2021-06-03 Griffith University Protéine immunogène contre infection gonococcique
RU2775621C2 (ru) * 2017-08-23 2022-07-05 Гриффит Юниверсити (Griffith University) Иммуногенный пептид против стрептококков группы а
US11679141B2 (en) 2019-12-20 2023-06-20 Nammi Therapeutics, Inc. Formulated and/or co-formulated liposome compositions containing toll-like receptor (“TLR”) agonist prodrugs useful in the treatment of cancer and methods thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070298093A1 (en) * 2003-12-23 2007-12-27 Abdo Konur Synergistic Liposomal Adjuvants
US20120039994A1 (en) * 2006-09-26 2012-02-16 Infectious Disease Research Institute Vaccine composition containing synthetic adjuvant

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ239643A (en) * 1990-09-17 1996-05-28 North American Vaccine Inc Vaccine containing bacterial polysaccharide protein conjugate and adjuvant (c-nd-che-a-co-b-r) with a long chain alkyl group.
US5866135A (en) * 1994-04-21 1999-02-02 North American Vaccine, Inc. Group A streptococcal polysaccharide immunogenic compositions and methods
ES2185762T3 (es) * 1995-03-13 2003-05-01 Secr Defence Vacunas contra la peste.
EP1506007B1 (fr) * 2002-05-14 2011-06-22 Novartis Vaccines and Diagnostics S.r.l. Vaccins combines destines a etre administres aux muqueuses pour l'immunisation contre la meningite bacterienne
US7255867B2 (en) * 2002-11-15 2007-08-14 Id Biomedical Corporation Of Quebec Vaccine
CN101140276A (zh) * 2007-10-26 2008-03-12 兰州生物制品研究所 肺炎结合疫苗原液中结合物含量的测定方法
CN101642434B (zh) * 2009-06-08 2011-07-20 邓菊娟 乳酸左氧氟沙星脂质体氯化钠注射液及其制备方法
KR101998431B1 (ko) * 2011-04-26 2019-07-09 몰레큘라 익스프레스 인코포레이티드 리포솜 제제
WO2013004234A2 (fr) * 2011-07-04 2013-01-10 Statens Serum Institut Procédé de production de liposomes
WO2013020090A2 (fr) * 2011-08-04 2013-02-07 The Regents Of The University Of California Glycopolypeptides dépourvus de glcnac streptococcique, glucides de paroi cellulaire, vaccins streptococciques et procédés pour les fabriquer et les utiliser
JP6240077B2 (ja) * 2011-10-06 2017-11-29 イムノバクシーン・テクノロジーズ・インコーポレイテッドImmunovaccine Technologies Inc. Tlr2を活性化するか、またはその活性を増加させるアジュバントを含むリポソーム組成物およびその使用
CN103157108B (zh) * 2011-12-13 2016-01-20 宁云山 免疫调节剂组合物及其药物组合物和应用
MY191217A (en) * 2014-04-15 2022-06-09 Univ Griffith Group a streptococcus vaccine
CN104055736A (zh) * 2014-07-08 2014-09-24 安徽医科大学 包封纳米铝的载体及其应用

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070298093A1 (en) * 2003-12-23 2007-12-27 Abdo Konur Synergistic Liposomal Adjuvants
US20120039994A1 (en) * 2006-09-26 2012-02-16 Infectious Disease Research Institute Vaccine composition containing synthetic adjuvant

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
HAROKOPAKIS E. ET AL.: "Effectiveness of Liposomes Possessing Surface-Linked Recombinant B Subunit of Cholera Toxin as an Oral Antigen Delivery System", INFECTION AND IMMUNITY, vol. 66, no. 9, 1998, pages 4299 - 4304, XP055378636 *
OZBERK V. ET AL.: "Understanding the disease pathogenesis of group A streptococcus upper respiratory tract infection to help improve vaccine and vaccination strategies with J8-DT", BACPATH 13: MOLECULAR ANALYSIS OF BACTERIAL PATHOGENS CONFERENCE, 28 September 2015 (2015-09-28), XP055378640 *
PIERCE N. ET AL.: "Enhancement by Lipid A of Mucosal lmmunogenicity of Liposome- Associated Cholera Toxin", REVIEWS OF INFECTIOUS DISEASES, vol. 6, no. 4, 1984, pages 563 - 566 *
SCHWENDENER R.: "Liposomes as vaccine delivery systems: a review of the recent advances", THERAPEUTIC ADVANCES IN VACCINES, vol. 2, no. 6, 2014, pages 159 - 182, XP055206678 *
WATSON D. ET AL.: "Design considerations for liposomal vaccines: Influence of formulation parameters on antibody and cell -mediated immune responses to liposome associated antigens", VACCINE, vol. 30, no. 13, 2012, pages 2256 - 2272, XP028464639 *
ZAMAN M. ET AL.: "Liposomal mucosal vaccine delivery system: Immunogenicity, inflammatory response and protection from group A Streptococcus challenge", JOURNAL OF CLINICAL AND CELLULAR IMMUNOLOGY, vol. 7, no. 4, 28 July 2016 (2016-07-28), XP055378641 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019036761A1 (fr) * 2017-08-23 2019-02-28 Griffith University Peptide immunogène dirigé contre le streptocoque de groupe a
RU2775621C2 (ru) * 2017-08-23 2022-07-05 Гриффит Юниверсити (Griffith University) Иммуногенный пептид против стрептококков группы а
US11732033B2 (en) 2017-08-23 2023-08-22 Griffith University Immunogenic peptide against group a Streptococcus
AU2018322485B2 (en) * 2017-08-23 2024-02-01 Griffith University Immunogenic peptide against group A streptococcus
WO2021041891A1 (fr) * 2019-08-30 2021-03-04 Emory University Utilisation d'acide désoxycholique, de dérivés ou de sels de ceux-ci dans la gestion d'infections bactériennes et compositions associées
WO2021102505A1 (fr) 2019-11-25 2021-06-03 Griffith University Protéine immunogène contre infection gonococcique
US11679141B2 (en) 2019-12-20 2023-06-20 Nammi Therapeutics, Inc. Formulated and/or co-formulated liposome compositions containing toll-like receptor (“TLR”) agonist prodrugs useful in the treatment of cancer and methods thereof
US11744874B2 (en) 2019-12-20 2023-09-05 Nammi Therapeutics, Inc. Formulated and/or co-formulated liposome compositions containing toll-like receptor (“TLR”) agonist prodrugs useful in the treatment of cancer and methods thereof
US11896646B2 (en) 2019-12-20 2024-02-13 Nammi Therapeutics, Inc. Formulated and/or co-formulated liposome compositions containing toll-like receptor (“TLR”) agonist prodrugs useful in the treatment of cancer and methods thereof

Also Published As

Publication number Publication date
CN109069424A (zh) 2018-12-21
CN106606775A (zh) 2017-05-03
CN109069424B (zh) 2021-12-31

Similar Documents

Publication Publication Date Title
JP2022078317A (ja) Staphylococcus aureusに対して免疫化するための組成物
JP6494527B2 (ja) 肺炎球菌のコロニー形成および/または疾患からの保護を提供するタンパク質抗原
JP2009515831A (ja) ペスト菌(Yersiniapestis)抗原を含む組成物
JP2011512152A (ja) 改善された溶解度を有するEscherichiacoli免疫原
JP2012502073A (ja) H因子結合タンパク質免疫原
JP2012501959A (ja) Yersiniapestis抗原を含む組成物
US10513544B2 (en) Group A streptococcus vaccine
CN109069424B (zh) 脂质体疫苗
AU2012257771C1 (en) Vaccine against Streptococcus pneumoniae
JP2013520487A (ja) 免疫原性タンパク質および組成物
JP2012532600A (ja) 保存された大腸菌免疫原
JP2018135388A (ja) シュードモナス抗原および抗原の組み合わせ
AU2021205866A1 (en) Self-assembling, self-adjuvanting system for delivery of vaccines
Tarahomjoo Utilizing bacterial flagellins against infectious diseases and cancers
US9119803B2 (en) Carious tooth vaccine and preparation method
TW201726164A (zh) 脂質體疫苗
RU2775621C2 (ru) Иммуногенный пептид против стрептококков группы а
JP2013510188A (ja) 黄色ブドウ球菌に由来する菌血症関連抗原

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: 16858499

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16858499

Country of ref document: EP

Kind code of ref document: A1