NZ616804B2 - Vaccine delivery method - Google Patents

Vaccine delivery method Download PDF

Info

Publication number
NZ616804B2
NZ616804B2 NZ616804A NZ61680412A NZ616804B2 NZ 616804 B2 NZ616804 B2 NZ 616804B2 NZ 616804 A NZ616804 A NZ 616804A NZ 61680412 A NZ61680412 A NZ 61680412A NZ 616804 B2 NZ616804 B2 NZ 616804B2
Authority
NZ
New Zealand
Prior art keywords
composition
antigen
vaccine
schistosome
manufacture
Prior art date
Application number
NZ616804A
Other versions
NZ616804A (en
Inventor
Donald A Harn
Rafaella Queiroz
Lisa Shollenberger
Original Assignee
University Of Georgia Research Foundation Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Georgia Research Foundation Inc filed Critical University Of Georgia Research Foundation Inc
Priority claimed from PCT/US2012/034012 external-priority patent/WO2012145355A1/en
Publication of NZ616804A publication Critical patent/NZ616804A/en
Publication of NZ616804B2 publication Critical patent/NZ616804B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5252Virus inactivated (killed)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • A61K2039/552Veterinary vaccine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • 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
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0003Invertebrate antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/002Protozoa antigens
    • A61K39/012Coccidia antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/002Protozoa antigens
    • A61K39/015Hemosporidia antigens, e.g. Plasmodium antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/0208Specific bacteria not otherwise provided for
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/025Enterobacteriales, e.g. Enterobacter
    • A61K39/0258Escherichia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/025Enterobacteriales, e.g. Enterobacter
    • A61K39/0275Salmonella
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/04Mycobacterium, e.g. Mycobacterium tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/05Actinobacteria, e.g. Actinomyces, Streptomyces, Nocardia, Bifidobacterium, Gardnerella, Corynebacterium; Propionibacterium
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/08Clostridium, e.g. Clostridium tetani
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/085Staphylococcus
    • 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
    • A61K39/02Bacterial antigens
    • A61K39/099Bordetella
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/102Pasteurellales, e.g. Actinobacillus, Pasteurella; Haemophilus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/104Pseudomonadales, e.g. Pseudomonas
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/104Pseudomonadales, e.g. Pseudomonas
    • A61K39/1045Moraxella
    • 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
    • A61K39/12Viral antigens
    • A61K39/145Orthomyxoviridae, e.g. influenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/15Reoviridae, e.g. calf diarrhea virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/155Paramyxoviridae, e.g. parainfluenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/155Paramyxoviridae, e.g. parainfluenza virus
    • A61K39/17Newcastle disease virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/215Coronaviridae, e.g. avian infectious bronchitis virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • A61K39/255Marek's disease virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • A61K39/265Infectious rhinotracheitis virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/29Hepatitis virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/29Hepatitis virus
    • A61K39/292Serum hepatitis virus, hepatitis B virus, e.g. Australia antigen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • 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
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/10Anthelmintics
    • A61P33/12Schistosomicides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16311Mardivirus, e.g. Gallid herpesvirus 2, Marek-like viruses, turkey HV
    • C12N2710/16334Use 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16711Varicellovirus, e.g. human herpesvirus 3, Varicella Zoster, pseudorabies
    • C12N2710/16734Use 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24034Use 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
    • C12N2720/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsRNA viruses
    • C12N2720/00011Details
    • C12N2720/10011Birnaviridae
    • C12N2720/10034Use 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
    • C12N2720/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsRNA viruses
    • C12N2720/00011Details
    • C12N2720/12011Reoviridae
    • C12N2720/12311Rotavirus, e.g. rotavirus A
    • C12N2720/12334Use 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
    • C12N2730/00Reverse transcribing DNA viruses
    • C12N2730/00011Details
    • C12N2730/10011Hepadnaviridae
    • C12N2730/10111Orthohepadnavirus, e.g. hepatitis B virus
    • C12N2730/10134Use 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/10011Circoviridae
    • C12N2750/10034Use 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14311Parvovirus, e.g. minute virus of mice
    • C12N2750/14334Use 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
    • C12N2760/16111Influenzavirus 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
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16171Demonstrated in vivo effect
    • 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
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18034Use 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
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18111Avulavirus, e.g. Newcastle disease virus
    • C12N2760/18134Use 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/10011Arteriviridae
    • C12N2770/10034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24311Pestivirus, e.g. bovine viral diarrhea virus
    • C12N2770/24334Use 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/32011Picornaviridae
    • C12N2770/32311Enterovirus
    • C12N2770/32334Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

Disclosed is a composition comprising: -a self assembling peptide that forms a stable hydrogel at physiological salt concentrations; -an adjuvant, wherein the adjuvant comprises a Toll-Like Receptor (TLR) agonist; and -one or more antigens. Also disclosed is the use of the above-described composition in the manufacture of a medicament for immunising a subject. osition in the manufacture of a medicament for immunising a subject.

Description

VACCINE DELIVERY METHOD CONTINUING APPLICATION DATA This application claims the benefit ofUS. Provisional Application Serial No. 61/476,431, filed April 18, 2011, which is incorporated by reference herein.
GOVERNMENT FUNDING This invention was made with government support under Grant Nos. AIO7l883 and A1036657, awarded by that National utes of Health. The Government has certain rights in the invention.
BACKGROUND Vaccines remain the single greatest public health asset for combating infectious es.
The goal of vaccine delivery is to present vaccine antigens in a manner that enhances antigen presenting cell tion, uptake of antigen and processing. An additional goal is to reduce the number of vaccinations required to induce an ive, vaccine-specific response, especially if a single effective dose of a vaccine is ble. Current, conventional vaccine delivery s use alum. um salts, such as alum, were first licensed for use as adjuvants in human vaccines in the 1920's. There is a need for improved delivery modes and adjuvants that are safe for use in vaccine formulations.
SUMMARY OF THE INVENTION The present invention includes a ition including as one component a slurry matrix that is a liquid at room temperature and a gel at physiological salt concentrations and/or physiological temperatures and as another component one or more antigens. In some aspects of the composition, the slurry matrix is a peptide hydrogel. In some aspects, the peptide hydrogel includes PURAMATRIX, or a derivative thereof. In some aspects, the peptide hydrogel includes the peptide scaffold RADARADARADARADA, or a derivative f. In some aspects of the composition, the slurry matrix es MATRIGEL, or a derivative thereof In some aspects of the composition, the composition further includes one of more nts. In some aspects, an adjuvant includes a Toll-Like Receptor (TLR) agonist and/or a cytokine. In some s, a TLR agonist includes a TLR4 agonist. In some aspects, a TLR agonist includes a TLR9 agonist. In some aspects, a TLR9 agonist includes a CpG oligodeoxynucleotide (ODN).
In some aspects of the composition, the composition further includes a Toll-Like Receptor (TLR) agonist and/or a cytokine. In some aspects, a TLR agonist includes a TLR4 agonist. In some aspects, a TLR agonist includes a TLR9 agonist. In some aspects, a TLR9 t includes a CpG oligodeoxynucleotide (ODN).
In some aspects of the composition, the antigen includes a hepatitis antigen, an influenza antigen, a schistosomiasis antigen, and/or a burkolderia antigen, or an nic fragment thereof The present invention includes a method of producing an immune response in a subject, the method including administering a composition as described herein to the subject.
The present invention includes a method of immunizing a subject, the method including administering a composition as described herein to the subject.
The present invention includes a method of delivering one or more immunogenic antigens to a subject, the method including administering a composition as described herein to the subject.
The present invention includes a method of delivering one or more therapeutic antigens to a t, the method including administering a composition as described herein to the t.
In some aspects of the methods of the present invention, the subject is a domestic livestock or a companion animal. In some aspects of the methods of the present invention, the subject is y. In some aspects of the methods of the present invention, the t is human.
In some aspects of the methods of the present invention, stration of the composition es subcutaneous (sc) injection or intramuscular (im) injection.
In some aspects of the methods of the present ion, administration of the ition es administration as a primary and/or booster vaccination.
In some aspects of the methods of the t invention, administration of the composition includes administration as a booster vaccination after a primary vaccination with a polypeptide vaccine or a plasmid DNA vaccine.
The t invention includes a method of producing an anti-schistosome immune response in a bovoid, the method including stering a composition including as one component a slurry matrix that is a liquid at room temperature and is a gel at physiological conditions and as another component one or more schistosome antigens to the bovoid.
The present invention includes a method of producing an anti-schistosome immune response in a bovoid, the method including administering a composition as described herein to the bovoid, wherein one or more antigen includes a schistosome antigen.
The present invention includes a method of schistosomiasis vaccination in a bovoid, the method including stering a composition including as one component a slurry matrix that is a liquid at room temperature and is a gel at physiological ions and as another component one or more schistosome antigens.
The present invention includes a method of schistosomiasis vaccination in a bovoid, the method including administering a composition as described herein to the bovoid, wherein one or more antigen includes a schistosome antigen.
In some aspects of the methods, the composition fiarther es one or more nts.
In some aspects, an adjuvant includes a Toll-Like Receptor (TLR) agonist and/or a cytokine. In some aspects, a TLR agonist includes a TLR4 and/or a TLR9 t. In some aspects, a TLR9 agonist includes a CpG oligodeoxynucleotide (ODN). In some aspects, a CpG ODN includes bovine CpG. In some aspects, the nt includes the cytokine IL-12.
In some aspects of the methods, the composition further includes a Toll-Like Receptor (TLR) agonist and/or a cytokine. In some s, a TLR agonist includes a TLR4 and/or a TLR9 agonist. In some aspects, a TLR9 agonist includes a CpG oligodeoxynucleotide (ODN).
In some aspects of the s, a CpG ODN includes bovine CpG.
In some aspects of the methods, the osome n es a Schistosoma japonz'cum antigen, or an antigenic fragment thereof.
In some aspects of the s, the schistosome antigen includes is a SjCTPI polypeptide, a SjCTPI-Hsp70 polypeptide, a SjC23 polypeptide, and/or a SjC23-Hsp70 polypeptide, or an antigenic fragment thereof.
In some s of the methods, administration of the composition includes administration as a primary and/or a booster vaccination.
In some aspects of the methods, administration of the composition includes administration as booster ation after a primary vaccination with a SjCTPI-Hsp70 plasmid DNA vaccine.
In some aspects of the s, the method r includes administration of one or more anti-schistosome chemotherapeutic agents.
In some aspects of the methods, the method trates at least 45% efficacy in the prevention of infection with a schistosome parasite.
The present invention includes methods of making the composition described herein.
The term "and/or" means one or all of the listed elements or a combination of any two or more of the listed elements.
The words "preferred" and "preferably" refer to ments of the invention that may afford certain benefits, under certain circumstances. However, other embodiments may also be preferred, under the same or other circumstances. rmore, the recitation of one or more preferred embodiments does not imply that other embodiments are not , and is not intended to exclude other embodiments from the scope of the invention.
The terms "comprises" and variations f do not have a limiting meaning where these terms appear in the description and claims.
Unless otherwise specified, "a, an," "the," and "at least one" are used interchangeably and mean one or more than one.
Also herein, the recitations of numerical ranges by endpoints include all numbers subsumed within that range (e.g., l to 5 includes 1, 1.5, 2, 2.75, 3, 3.80, 4, 5, etc.).
Where a range of values is ed, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.
For any method disclosed herein that includes discrete steps, the steps may be conducted in any feasible order. And, as appropriate, any combination of two or more steps may be conducted simultaneously.
The above summary of the present invention is not ed to describe each disclosed embodiment or every implementation of the present invention. The description that follows more particularly ifies illustrative embodiments. In several places hout the application, guidance is ed through lists of examples, which examples can be used in various combinations. In each instance, the recited list serves only as a representative group and should not be interpreted as an exclusive list.
BRIEF DESCRIPTION OF THE FIGURES Figure 1 shows the kinetics of IgA anti-HBsAg antibody titers. Data shown is pooled from two independent experiments for total n=l 0.
Figure 2 shows the kinetics of IgM anti-HBsAg antibody titers. Data shown is pooled from two independent experiments for total n=l 0.
Figure 3 shows the cs of IgG BsAg antibody titers. Data shown is pooled from two independent ments for total n=l 0.
Figure 4 shows the kinetics of IgG1 anti-HBsAg antibody . Data shown is pooled from two independent experiments for total n=l 0.
Figure 5 shows the kinetics of Inga anti-HBsAg antibody titers. Data shown is pooled from two independent experiments for total n=l 0.
Figure 6 shows higher anti-HBsAg antibody titers after single vaccination (21 days).
Data shown is pooled from two independent experiments for total n=lO; *p<0.05, **p<0.0l, **p<0.00l, ****p<0.0001 compared to antigen alone using two-way ANOVA with Bonferroni post-test.
Figure 7 shows higher anti-HBsAg antibody titers after single vaccination (35 days).
Data shown is pooled from two independent ments for total n=lO; *p<0.05 compared to n alone using two-way ANOVA with Bonferroni post-test.
Figure 8 shows cytokine profile twenty-four hours after HBsAg mulation of splenocytes. Data shown is representative of one experiment, n=5; *p<0.05, ****p<0.0001 compared to antigen alone using two-way ANOVA with Bonferroni post-test.
Figure 9 shows cytokine profile forty-eight hours after HBsAg re-stimulation of splenocytes. Data shown is pooled from two independent experiments for total n=10 (n=5 for adjuvants only); no statistical differences seen (p<0.05) ed to antigen alone using two- way ANOVA with Bonferroni post-test.
Figure 10 shows cytokine profile seventy-two hours after HBsAg re-stimulation of splenocytes. Data shown is pooled from two independent experiments for total n=10 (n=5 for nts only); **p<0.01 compared to antigen alone using two-way ANOVA with Bonferroni post-test.
Figure ll shows increased HBsAg-specific cell-mediated immunity by ELISpot in HbsAg-specific T cell responses. Data shown is pooled from two independent experiments for total n=lO; *p<0.05 compared to antigen alone using two-way ANOVA with Bonferroni post- test.
Figure 12 shows increased specific T cell-mediated ty by flow try.
Data shown is pooled from two independent experiments for total n=lO; 01 compared to antigen alone using two-way ANOVA with Bonferroni post-test.
Figure 13 is a radar graph representing the prevalence of ex vivo cytokine balance in a range of cell subsets from immunized mice (n=lO/group) 24 hours post-primary inoculation.
The values of the axis can be joined to form the central polygon area which represents the general cytokine balance. The analysis of the radar chart axes highlights the contribution of leukocyte for the overall cytokine balance.
Figures 14A and 14B are radar graphs representing the prevalence of ex vivo ne balance in a range of cell subsets from immunized mice group) 48 hours post-primary ation. Fig. 14A presents TNF and IFN gamma. Fig. 14B presents IL-5 and IlL-4. Each axis displays the proportion of each cytokine balance category. The values of each axis can be joined to form the central polygon area which represents the general cytokine balance. sing or decreasing central polygon areas reflect higher or lower contribution of inflammatory or regulatory cytokine balance in each group.
Figure 15 is a radar graph representing the prevalence of ex vivo cytokine balance in a range of cell subsets from immunized mice group) 72 hours rimary inoculation.
The values of the axis can be joined to form the central polygon area which represents the general cytokine balance. The analysis of the radar chart axes highlights the contribution of leukocyte for the overall cytokine balance. s 16A to 16D are comparisons of rHepBag-specific antibodies induced in immunized mice (n=10/group) between 14 and 35 days rime inoculation. The boost was performed 28 days after prime immunization and g-specific antibodies levels were determined by ELISA assay. Fig. 16A presents data from 14 days, Fig. 16B ts data from 21 days, Fig. 16C presents data from 28 days, and Fig. 16D presents data from 35 days.
Statistical significance at P S 0.05 are represented by superscript letters 'a', 'b' and 'c' for comparisons with rHepBag, rHepBag in Alhydrogel ® and rHepBag in Freund, respectively.
Figures 17A to 17D show IgGl :IgG2a ratio after immunization of mice (n=10/group) with rHepBag plus adjuvants between 14 and 35 days. The boost was performed 28 days after prime immunization and rHepBag-specific antibodies levels were determined by ELISA assay.
Fig. 17A presents data from 14 days, Fig. 17B presents data from 21 days, Fig. 17C ts data from 28 days, and Fig. 17D ts data from 35 days.
Figures 18A and 18B show increased anti-NP IgG titers in two strains of mice vaccinated with rNP. Fig. 18A shows titers in C57BL/6 mice. Fig. 18B shows titers in Balb/c mice. Sera were collected four weeks post-last vaccination (4wplv).
Figures 19A and 19B show increased anti-influenza IgG titers in in C57Bl/6 mice vaccinated with PR8 WIV. Fig. 19A and Fig. 19B represent results from two independent experiments. Sera were collected four weeks post-last vaccination (4wplv).
Figures 20A and 20B show increased tion from lethal challenge in 6 mice vaccinated with PR8 WIV. Fig. 20A shows results from an independent experiment with lethal challenge at 30 LD50. Fig. 20B shows results from an independent experiment with lethal challenge at 1000 LD50.
Figures 2lA-2lC show increased urkholderia IgG titers in mice zed with a cocktail of three recombinant burkholderia protein antigens with three different adjuvants (alum, CFA, or PURAMTRIX gel). Fig. 21A shows anti-burkholderia protein 4-9 IgG titers in zed mice. Fig. 21B shows anti-burkholderia protein 22-ll IgG titers in immunized mice.
Fig. 21C shows anti-burkholderia protein 42 IgG titers in immunized mice.
Figures 22A and 22B show IgG anti-schistosome CCA protein antibody titers in mice immunized with CCA in complete Freund’s adjuvant (Fig. 22A) and CCA in MATRIGEL plus CpG (Fig. 22B).
DETAILED DESCRIPTION OF RATIVE EMBODIMENTS OF THE PRESENT INVENTION The present invention provides compositions and methods that provide for improved vaccine ry, providing for the improved focal and sustained delivery of antigen at the site of vaccine administration; enhancing antigen presenting cell activation and antigen uptake and processing. With the t invention, one or more immunogenic agents is administered in a composition ing a slurry matrix component that is a liquid at room temperature and/or low salt concentrations and a gel at physiological salt concentrations and/or physiological temperatures. Gelling may be induced by the physiological body ature of a vertebrate, such as a mammal or bird. Such a temperature may be, for example, at least about 25° Celsius (C), at least about 30° Celsius, at least about 32° Celsius, at least about 35° Celsius, at least about 37°Celsius, at least about 39° Celsius, or at least about 40°. Gelling may be induced by the physiological salt concentrations. In some embodiments, gelling may in the presence of millimolar concentrations of salt, for e by a salt concentration of r than about 0.05 molar (M).
Thus, the vaccine composition gels or rizes after administration to a subject, localizing the vaccine antigens to a single site where innate n presenting cells can home and begin taking up vaccine antigens. Ideally, the slurry matrix is a biocompatible material will not induce rable reactions in the body as a result of contact with bodily fluids or tissue, such as tissue death, tumor formation, allergic reaction, foreign body reaction (rejection), atory reaction, antibody se, or blood ng, for example. A slurry matrix may also be referred to herein as a “biomedical polymer hydrogel,” a “biomedical hydrogel,” “biomedical polymer,” a “polymer hydrogel,” a “biocompatible polymer hydrogel,” a “biocompatible hydrogel,” a mpatible polymer,” or a “hydrogel.” As used herein, a “hydrogel” is a 3-dimensional network of cross-linked, hydrophilic macromolecules capable of being swelled and incorporating about 20 percent to about 95 percent water by weight. A hydrogel is a gel in which the liquid constituent is water. As used herein, a gel is a solid, j elly- like material that can have properties ranging from soft and weak to hard and tough. A gel is a ntially dilute cross-linked system, which exhibits no flow when in the steady-state. By weight, gels are mostly liquid, yet they behave like solids due to a three-dimensional cross-linked network within the liquid. It is the crosslinks within the fluid that give a gel its structure (hardness). In this way gels are a dispersion of molecules of a liquid within a solid in which the solid is the continuous phase and the liquid is the discontinuous phase. Hydrogel is a network of polymer chains that are hydrophilic, sometimes found as a colloidal gel in which water is the dispersion medium. Hydrogels are highly absorbent (they can contain over 99.9% water) natural or synthetic polymers. Hydrogels also possess a degree of flexibility very similar to natural tissue, due to their cant water content.
Any of a wide variety of biomedical polymer els available for use in medical technologies may be used with the methods and itions described herein.
In some ments, the slurry matrix is lly occurring, such as for example, fibrin, collagen, n, agarose, methylcellulose, hyaluronan, and other lly derived polymers. In some embodiments, the slurry matrix is MATRIGEL, or a derivative thereof.
EL is a basal membrane extract from mouse cells and includes laminin, collagen IV, entactin, nidogen, and proteoglycans. The invasion of tumor cells into MATRIGEL has been used to study the involvement of extracellular matrix receptors and matrix degrading enzymes in tumor progression and invasion and MATRIGEL has been used also as an in vitro and in vivo angiogenesis model (MATRIGEL plug assay) to study the activity of enic and anti- angiogenic cytokines and other substances. MATRIGEL is commercially available as BD MatrigelTM Matrix. See the world wide web at bdbiosciences.com/cellculture/ecm/ ecmtypes/index.j sp.
In some embodiments, a slurry matrix is synthetic, such as for example, a synthetic e el or a self-assembling peptide (sapeptide) scaffold. The sapeptide scaffolds are formed h the neous assembly of ionic self-complementary beta-sheet oligopeptides under physiological conditions, producing a hydrogel material. These short peptides (typically about 8, about 12, about 16, about 24, or about 32 amino acid residues with intemally-repeating sequences) self-assemble in aqueous salt solution into three-dimensional matrices. The peptides are characterized as being amphiphilic, having alternating hydrophobic and hydrophilic amino acid residues; greater than 12 amino acids, and preferably at least 16 amino acids; complementary and structurally compatible. Complementary refers to the ability of the peptides to interact through ionized pairs and/or hydrogen bonds which form between their hydrophilic side-chains, and structurally compatible refers to the ability of complementary peptides to maintain a constant distance between their peptide backbones. Peptides having these properties participate in intermolecular interactions which result in the formation and ization of betasheets at the secondary structure level and interwoven filaments at the tertiary structure level.
Examples include, but are not d to, peptide family members, RAD 16-1 ((RADA)(4); SEQ ID NO:2) RAD 16-11 ((RARADADA)(2); SEQ ID NO:3), KFE-8 ((FKFE)(2); SEQ ID NO:4), or KLD-12 ((KLDL)(3); SEQ ID NO:5). See, for example, U.S. Patent 483; Holmes et al, 2000, Proc Natl Acad Sci USA; 97(12):6728-33; Yokoi et al, 2005, Proc Natl Acad Sci USA; 102(24):8414-9; Liu et al, 2012, Nanoscale; 4(8):2720-7, and BD PuraMatrix™ Peptide Hydrogel, Guidelines for Use, Catalog Number 354250 (SPCG rev 2.0; BD Biosciences, Bedford, MA). In some aspects, a peptide hydrogel includes the peptide scaffold self-assembling building blocks of arginine-alanine-aspartate-alanine (RADA; SEQ ID NO:1). In some aspects, a e hydrogel includes DARADARADA (SEQ ID NO:2), or a derivative thereof.
In some aspects, the peptide el includes PURAMATRIX, or a derivative thereof. See, for example, U.S. Patent 5,670,483; Holmes et al, 2000, Proc Natl Acad Sci USA; 97(12):6728-33; Yokoi et al, 2005, Proc Natl Acad Sci USA; 102(24):8414-9; Liu et al, 2012, Nanoscale; 4(8):2720-7, and BD PuraMatrix™ Peptide Hydrogel, ines for Use, Catalog Number 354250 (SPCG rev 2.0; BD Biosciences, Bedford, MA), each of which are incorporated herein in their entireties.
In some embodiments, a biomedical polymer hydrogel may be a polyethylene glycol (PEG) hydrogel that rizes spontaneously in vivo.
In some embodiments, a slurry matrix, in on to gelling at vertebrate or ian body temperature and/or logical salt concentrations, is a bioresorbable synthetic polymer that degrades and ves with time. Such compounds are naturally degraded in the body by hydrolysis and absorbed as water-soluble monomers. Examples include, polylactic acid, polylactide (PLA), poly (L-lactic acid), poly-D-lactide, polyglycolic acid (PGA), polyglycolide and its copolymers (poly(lactic-co-glycolic acid) with lactic acid, homo- and copolymers of lactic acid and glycolic acid, poly (DL-lactic lycine) copolymers, poly (DL-lacticco-glycolic acid) (PLGA), poly (DL-lacticco-glycolic acid) , porous poly(DL-lactic-co-glycolic acid) foams, poly(amino acids) poly[ox(l-oxo-l,2-ethanediyl)] 02)n; Biovek), poly(glycolide-co-caprolactone), poly(glycolide-co-trimethylene ate), polydioxanone (PDO, PDS), poly-p-dioxanone, caprolactone (also referred to as 2-oxepanone), epsiloncaprolactone , 6-hexanolactone, hexanolactone, l-oxaoxocycloheptane polyglactin 910, polyanhydrides, and polyorthoester films formed from poly (D,L-lactic-co-glycolic acid, 88: 12) (PLGA) or from a 50/50 (W/W) blend of PLGA and poly (L-lactic acid) (PLLA). See, for example, Schakenraad and Dijkstra, 1991, Clin Mater; 53-69; Mooney et al., 1997, J Biomed Mater Res; 4 1 3-20; and Lu et al., 2000, Biomaterials; 21(18): 1837-45.
Compositions of the present invention e one or more antigenic agents (also referred to herein as an gen). An antigenic agent may be any of the great variety of agents that are administered to a subject to elicit an immune response in the subject. An nic agent may be an immunogen derived from a pathogen. The antigenic agent may be, for example, a peptide or protein antigen, a viral antigen or polypeptide, an inactivated virus, a recombinant virus, a bacterial or parasitic antigen, an inactivated bacteria or parasite, a Whole cell, a genetically modified cell, a tumor associated antigen or tumor cell, or a carbohydrate antigen. In some applications an antigen is not a living cell. In some embodiments, an antigenic agent is a soluble antigen.
A composition as described herein may include as an antigenic agent any of the great variety of immunogenic agents available as vaccine components. Such vaccines may include, but are not limited to, antigenic vaccines components directed against various infectious, viral, and tic diseases and anti-tumor vaccine components. Antitumor vaccines include, but are not limited to, peptide vaccines, whole cell vaccines, genetically modified Whole cell vaccines, recombinant protein vaccines or vaccines based on expression of tumor associated antigens by recombinant viral s. In some embodiments, an antigenic agent is a soluble n.
An antigenic agent includes a bacterial antigen from, for example, Haemopholis influenza, za virus types A or B, Streptococcus pneumonia, Staphylococcus aureus, us cis antigen (such as, for example, PA).
An antigenic agent includes a parasite antigen from, for example, a malaria parasite or a schistosome parasite. Malaria antigens e, but are not d to antigens from the plasmodium species Plasmoclium vivax, Plasmodiumfalciparum, and Plasmoclium knowlesi, Plasmoclium ovaleancl, and Plasmodium malariae. Schistosome parasites e, but are not limited to, Schistosomajaponicum, Schistosoma i, and Schistosoma haematobium. A schistosome antigen may be a osome triose phosphate isomerase (CTPI) protein, or antigenic fragment or derivative thereof, including, but not limited to a S. japom'cum, S. monsoni, or S. haematobium CTPI protein, or antigenic nt or derivative thereof. A schistosome antigen may be a schistosome tetraspin 23 kDa integral membrane protein (C23), or antigenic fragment or tive thereof, including, but not limited to a S. japom'cum, S. monsoni, or S. haematobium C23 protein, or antigenic fragment or derivative thereof Such a schistosome antigen may be a chimeric polypeptide, fused to one or more additional antigenic determinants, such as for e, a heat shock protein, or nic fragment or derivative thereof, including, but not limited to, bovine heat shock protein 70 (Hsp70). Schistosome antigens include, for example, SjCTPI, SjCTPI-Hsp70, SjC23, and SjC23-Hsp70 polypeptides. Antigens may be form any of a variety of other parasites, including, but not limited to, kinetoplastid protozoa, such as for example, protozoa of the Blastocrz'thz'dz'a, Crithz'dz'a, Endotrypanum, Herpetomonas, Leishmam’a, Leptomonas, Phytomonas, Trypanosoma, and Wallaceina genera. In preferred embodiments, the protozoan is of the genus Trypanosoma, including, but not limited to, T. cruzi, T. brucez’, T.b. gambiense, and T.b. rhodesz'ense. In some ments, the protozoan is of the genus am'a, including, for example, Leishmania major. Notable trypanosomal es include trypanosomiasis an Sleeping Sickness and South American Chagas Disease, caused by species of osoma) and leishmaniasis (caused by species of Leishmania).
An antigenic agent includes a viral antigen from, for e, hepatitis, such as for example, hepatitis C, hepatitis B, or tis A, influenza, for example, the M2, hemaglutinin, and/or neuraminidase ns of an influenza virus, including, for example, influenza A (including, but not d to, the H5N1 and HlNl subtypes), influenza B, and za C, respiratory syncytial virus (RSV), rabies, papilloma virus, measles, a, varicella, rotavirus, polio, variscella zoster virus (VZV), and negative stranded RNA viruses, such as for example, a virus of the family Paramyxoviridae. Examples of a virus of the family Paramyxoviridae include, but are not limited to, human parainfluenza virus 1, human parainfluenza virus 2, human parainfluenza virus 3, human parainfluenza virus 4, parainfluenza virus 5, mumps virus, measles virus, human metapneumovirus, human respiratory syncytial virus, bovine respiratory syncytial virus rinderpest virus, canine distemper virus, phocine distemper virus, Newcastle disease virus, avian pneumovirus, Peste des Petits Ruminants virus (PPRV), Sendai virus, le virus, Tupaia paramyxovirus, Tioman virus, Tuhokovirus l, Tuhokovirus 2, Tuhokovirus 3, Hendravirus, Nipahvirus, Fer-de-Lance virus, Nariva virus, Salem virus, J virus, Mossman virus, and Beilong virus.
An antigenic agent may include one or more gens derived from pathogens infectious to poultry. Such immunogens may be derived from, for example, infectious bronchitis virus (IBV), Newcastle disease virus (NDV), Marek's disease (MDV), infectious bursal disease (IBD) virus, infectious laryngotracheitis (1LT), avian reovirus, cholera, fowl pox, mycoplasmosis, turkey and n Coryza, avian influenza, avian alomyelitis (AE), avian rhinotracheitis (ART), duck virus hepatitis, haemorrhagic enteritis, goose parvovirus, Paramyxovirus 3, chicken a virus (CAV), E. coli, Erysipelas, Reimerella, Mycoplasma gallisepticam, Pastearella maltocida, Salmonella enteritidis, Salmonella zjiphimariam, coccidiosis, egg drop syndrome (EDS) virus, turkey rhinotracheitis virus (TRTV), and poxvirus.
An nic agent may include one or more immunogens derived from pathogens infectious to bovoids, including, but not limited to, domestic , water buffalo, African buffalo, bison, and yaks. Such immunogens may be derived from, for example, bovine respiratory disease (BRD) vaccine, including, but not limited to BVDV types I and 11, bovine herpes virus 1 (BHV-l) vaccine, including, but not limited to, subunit vaccines that would not result in latent virus, Haemophilus somnas e, Mannheimia haemolytica vaccine, Mycoplasma bovis vaccine, bovine rus vaccine, Escherichia coli K99 e, bovine coronavirus (BCV) vaccine, Clostridiam ei (black leg) vaccine, Clostridiam am vaccine, Clostridiam li (malignant edema) vaccine, Clostridiam novyi (black disease) vaccine, Clostridiam perfringens otoxemia) vaccine, infectious bovine keratoconjunctivitis (pink eye) vaccine, including, but not limited to, lla bovis, chlamydia, mycoplasma, acholeplasma, or infectious bovine rhinotracheitis (1BR) virus vaccines, is vaccines, including, but not limited to, Escherichia coli J5 vaccine.
An antigenic agent may include one or more immunogens derived from pathogens infectious to swine, including, but are not limited to, porcine circovirus type 2 (PCV2), porcine uctive and respiratory syndrome (PRRSV), respiratory mycoplasma, Streptococcus sais, porcine coronavirus, rotavirus, enterotoxigenic Escherichia coli (K88), Actinobacillus plearopneamonia (APP), and swine influenza.
An nic agent may include one or more immunogens derived from the Barkholderia genus, a group of virtually ubiquitous gram-negative, motile, obligatory aerobic rod-shaped bacteria including both animal/human and plant pathogens as well as some environmentally important s. Burkholderz’a is best known for its pathogenic members. Burkholderz’a mallez' is responsible for glanders, a disease that occurs mostly in horses and related animals.
Burkholderia mallei is the causative agent of melioidosis (also called Whitmore's e), an infectious disease predominately of tropical climates that can infect humans or animals, especially in Southeast Asia and rn Australia. Burkholderz'a cepacia is an important en of pulmonary infections in people with cystic fibrosis. Due to their antibiotic resistance and the high mortality rate from their associated diseases Burkholderz’a mallez' and Burkholderia pseudomallei are considered to be potential biological warfare agents, targeting livestock and humans. Burkholderia antigens include, for example, any of three burkholderia recombinant proteins (the burkholderia 4-9 n, the burkholderia 22-11 protein, and the burkholderia 42 protein). Such deria antigens may be administered separately or as a cocktail of any two or three antigens.
An antigenic agent may be one or more of those currently used in the combination measles-mumps-rubella (MMR) and measles-mumps-rubella-varicella (MMRV) vaccines.
In some embodiments, the antigenic agent is a polynucleotide vaccine, that is, the antigenic agent is delivered as a vector construct, such as a plasmid, that results in the expression of a polypeptide antigen upon delivery to a subject. As used , the term "polynucleotide" refers to a polymeric form of nucleotides of any length, either ribonucleotides or deoxynucleotides, and es both double- and single-stranded RNA and DNA. A polynucleotide can be obtained directly from a natural , or can be ed with the aid of recombinant, enzymatic, or chemical techniques. A cleotide can be linear or circular in topology. A polynucleotide may be, for example, a portion of a vector, such as an expression or g vector, or a fragment. A polynucleotide may include nucleotide sequences having different fianctions, including, for ce, coding regions, and ding regions such as regulatory regions. Any suitable vector or delivery vehicle may be utilized. Various vectors are publicly available. The vector may, for example, be in the form of a plasmid, cosmid, viral particle, or phage. A vector may be an expression vector. The appropriate nucleic acid sequence may be inserted into the vector by a variety of ures. In general, DNA is inserted into an appropriate restriction endonuclease site(s) using techniques known in the art. Vector components generally include, but are not limited to, one or more of a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a ription termination sequence. Construction of le vectors containing one or more of these components employs standard ligation techniques which are known to those of skill in the art.
A composition as described herein may be useful as a vaccine. The vaccine can be a prophylactic or protective vaccine.
A composition of the present invention may include one or more compounds with adjuvant activity. Such an adjuvant stimulates the immune system and increases the response to a vaccine antigen, without having any specific antigenic effect in itself. An adjuvant acts to accelerate, prolong, or enhance antigen-specific immune responses when used in ation with specific vaccine antigens. le compounds or compositions for this e include, but are not limited to, an aluminum based adjuvant, such as, for example, aluminum phosphate, aluminum hydroxide (also referred to as alum), aluminum hydroxyl-phosphate, and aluminum hydroxyl-phosphate-sulfate, and non-aluminum adjuvants, such as, for example, QSZl, MF59, Lipid-A, neutral lipsomes, microparticles, a ne such as, for example, IL-l2, plant oils, animal oils, oil-in-water or water-in-oil emulsion based on, for example a mineral oil, such as Bayol FTM or Marcol 52““, Complete Freund's adjuvant, incomplete Freund's adjuvant, a vegetable oil such as, for e, vitamin E acetate, a saponin, squalene, a lipidated amino acid (“LAA”), and/or a TLR agonist.
In some embodiments, an adjuvant component is a toll-like receptor (TLR) ligand. TLRs in mammals were f1rstidentif1edin 1997. They constitute the first line of defense against many pathogens and play a crucial role in the function of the innate immune system. There are many known subclasses of Toll-like receptors, including, TLRl, TLR2, TLR3, TLR4, TLRS, TLR6, TLR7, TLR8, TLR9, TLRlO, TLRl l, TLRlZ, TLRl3, TLRl4, TLRlS, and TLRl6, and their ligands exhibit significant ural variation. A TLR t is a molecular ligand for one of the s Toll-like receptors (TLRs). Known TLRs include: TLRl (TLRl s include triacyl oteins); TLR2 (TLR2 ligands include oteins, gram positive peptidoglycan, lipoteichoic acids, fungi, and viral glycoproteins); TLR3 (TLR3 ligands include -stranded RNA, as found in certain viruses, and poly I:C); TLR4 (TLR4 ligands include lipopolysaccharide and viral glycoproteins); TLRS (TLRS ligands include flagellin); TLR6 (TLR6 ligands e diacyl lipoproteins); TLR7 (TLR7 ligands include small tic immune modifiers (such as imiquimod, R-848, loxoribine, and bropirimine) and single-stranded RNA); TLR8 (TLR8 ligands include small synthetic compounds and -stranded RNA); and TLR9 (TLR9 ligands include unmethylated CpG DNA motifs). Some TLR ligands are described herein, but it should be understood that such listings do not limit the invention in any way. TLR ligands are widely available commercially.
Preferred TLR agonists include TLR2 agonists, TLR4 agonists, TLR7 agonists, TLR8 agonists, and TLR9 agonists. LR2 is involved in the recognition of a wide array of microbial molecules from Gram-positive and Gram-negative ia, as well as mycoplasma and yeast.
TLR2 ligands include lipoglycans, lipopolysaccharides, ichoic acids and peptidoglycans.
TLR4 recognizes Gram-negative lipopolysaccharide (LPS) and lipid A, its toxic moiety.
TLR4 agonists include, but are not d to, lipopolysaccharide (LPS), Viral glycoproteins, monophosphoryl lipid A (MPL) (Anderson et al., 2010, Colloids SurfB Bioz'nterfaces; 75(1): 123-32), Glucopyranosyl Lipid nt—Stable on (GLA-SE) (Coler et al., 2010, PL0S One; 5(10):e13677), and the synthetic hexaacylated lipid A derivative, denoted as glucopyranosyl lipid adjuvant (GLA) (Coler et al., 2011, PLOS One; 6(1):e16333).
TLR9 is activated by unmethylated ntaining sequences, including those found in bacterial DNA or synthetic oligonucleotides (ODNs). Such ylated CpG containing sequences are present at high frequency in bacterial DNA, but are rare in mammalian DNA.
Thus, unmethylated CpG sequences distinguish microbial DNA from ian DNA. A TLR9 agonist may be a ation of microbial DNA, including, but not limited to, E. 6011' DNA, endotoxin free E. coli DNA, or endotoxin-free bacterial DNA from E. coli K12. A TLR9 agonist may be a synthetic oligonucleotide containing unmethylated CpG motifs, also referred to herein as “a igodeoxynucleotide,” “CpGODNs,” “ODN,” or “CpG.” CpG ODNs are short, single stranded, DNA molecules that contain a ne (“C” nucleotide) followed by a guanine (“G” nucleotide). The “p” typically refers to the phosphodiester backbone of DNA. A TLR9 agonist of the present invention may include any of the at least three types of stimulatory ODNs have been described, type A, type B, and type C. igodeoxynucleotides may be produced by standard methods for chemical synthesis of polynucleotides or purchased commercially. For example, CPG ODNs can be purchased through oGen (San Diego, CA).
The compositions as described herein may include one or more cytokines. Cytokines may include, but are not limited to, IL-lot, IL-lB, IL-2, IL-3, IL-4, IL-6, IL-8, IL-9, IL-10, IL-12, IL- 1 3 IL- 1 5 IL- 1 9, IL-20, IFN-(x, IFN—B, IFN—y, tumor necrosis factor (TNF), , , IL-18, transforming growth factor-B (TGF-B), granulocyte colony stimulating factor (G-CSF), macrophage colony stimulating factor (M-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), and or Flt-3 ligand. In some applications, one or more cytokines may serve as an adjuvant.
The present invention also includes methods of inducing an immune response in a subject by administering a composition as described herein to the t. The immune response may or may not confer protective immunity. An immune response may include, for example, a humoral response and/or a cell mediated immune response. A humoral immune response may include an IgG ding IgGl, IgG2 (including IgG2a and/or , IgG3, and/or IgG4), IgM, IgA, IgD, IgE, and/or IgY response. A ar immune response may include T cell activation and/or cytokine production. The determination of a humoral or cellular immune response may be determined by any of a variety of methods known in the immunological arts, including, but not limited to, any of those described herein. The induction of an immune response may include the priming and/or the stimulation of the immune system to a filture nge with an infectious agent, providing immunity to future infections. The ion of such an immune response may serve as a protective response, generally resulting in a reduction of the ms. The immune response may enhance an innate and/or adaptive immune response. The immune response may demonstrate higher concentrations of antibodies with a single, primary immunization. The immune se may show altered immunoglobulin ratios and/or altered induction of inflammatory cytokines, type I interferons, and/or chemokines, compared to immunization without the slurry matrix. Such alteration may be an increase or a decrease. For example, a higher ratio of one isotype of immunoglobulin compared to another immunoglobulin isotype (for example, any one of IgM, IgA, IgD, IgG, or IgE compared to any one of IgM, IgA, IgD, IgG, or IgE) or a higher ratio of one IgG subclass compared to r IgG subclass (for example, any one of IgG1, IgG2a, IgG2b, IgG3, or IgG4 compared to any one of IgG1, IgG2a, IgG2b, IgG3, or IgG4) may be obtained.
The present invention also includes methods of vaccinating a subject by administering a ition as described herein to the subject. Such vaccination may result in a reduction or mitigation of the symptoms of filture infection and may prevent a filture ion. The compositions described herein may have therapeutic and/or lactic applications as immunogenic compositions in preventing and/or ameliorating infection, such that resistance to new infection will be enhanced and/or the clinical severity of the e reduced. Such protection may be demonstrated by either a reduction or lack of the symptoms associated with RSS, including, but not limited to, any of those described herein. Any of a wide variety of available assays may be used to determine the effectiveness of the vaccination method of the t ion, including, but not d to, any of those described herein In some applications, an immunologically effective amount of at least one immunogen is employed in such amount to cause a substantial reduction in the course of the normal ion.
Immunogenicity and effectiveness may be assayed in any of a variety ofknown experimental systems, including, but not limited to, any of those described herein.
The compositions and methods described herein may be administered to a subject for the treatment and/or prevention of viral diseases, infectious diseases, including, but not limited to bacterial, fungal and parasitic infections, cancer, and other diseases in which the administration of one or more immunogens is therapeutically desired. With the methods of the present disclosure, the efficacy of the administration of one or more agents may be assessed by any of a variety of parameters known in the art, including, but not limited to, any of those described herein. This includes, for example, determinations of an increase in the delayed type hypersensitivity reaction to tumor antigen, determinations of a delay in the time to relapse of the post-treatment malignancy, determinations of an increase in relapse-free survival time, determinations of an increase in post-treatment survival, determination of tumor size, determination of the number of reactive T cells that are activated upon exposure to the vaccinating antigens by a number of methods including ELISPOT, FACS analysis, cytokine release, or T cell proliferation assays.
For example, the compositions and methods described herein may be stered to a patient for the treatment of cancer. mor vaccines include, but are not limited to, peptide vaccines, whole cell vaccines, genetically modified whole cell es, recombinant n es or vaccines based on expression of tumor associated antigens by recombinant viral vectors. Cancers to be treated include, but are not limited to, melanoma, basal cell oma, colorectal cancer, atic , breast cancer, prostate cancer, lung cancer (including small- cell lung carcinoma and non-small-cell carcinoma), leukemia, lymphoma, sarcoma, ovarian cancer, 's a, n's Disease, Non-Hodgkin's Lymphoma, multiple myeloma, neuroblastoma, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, small-cell lung tumors, primary brain tumors, stomach cancer, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, lymphomas, thyroid cancer, neuroblastoma, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, cervical cancer, endometrial cancer, glioblastoma, and adrenal cortical cancer. In some aspects, the cancer is a primary cancer. In some aspects, the cancer is metastatic. As used herein, “tumor” refers to all types of cancers, neoplasms, or malignant tumors found in mammals.
The efficacy of treatment of a cancer may be assessed by any of various parameters well known in the art. This includes, but is not limited to, inations of a reduction in tumor size, determinations of the tion of the growth, spread, veness, vascularization, angiogenesis, and/or metastasis of a tumor, determinations of the inhibition of the growth, spread, veness and/or vascularization of any atic lesions, determinations of tumor infiltrations by immune system cells, and/or determinations of an increased delayed type hypersensitivity reaction to tumor antigen. The efficacy of treatment may also be ed by the determination of a delay in relapse or a delay in tumor progression in the subject or by a determination of survival rate of the subject, for example, an increased al rate at one or five years post ent. As used , a relapse is the return of a tumor or neoplasm after its apparent cessation.
As used herein, unless the context makes clear otherwise, “treatment,” and similar word such as “treated,” “treating,” etc., is an approach for obtaining beneficial or desired results, including and preferably clinical results. A ent may include therapeutic and/or prophylactic treatments. Desirable effects of treatment e preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of one or more direct or indirect pathological consequences of the e, decreasing the rate of e progression, amelioration or palliation of the disease state, and ion or improved prognosis. In some ations, a composition as described may demonstrate an improvement in one or more desired results of at least about 10%, at least about 15%, at least about 20%, or at least about %, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, or at least about 95%.
As used herein, an “effective amount” or a peutically ive amount” of a substance is that amount sufficient to affect a desired biological , such as beneficial results, including clinical results. Therapeutically effective concentrations and amounts may be determined for each application herein empirically by testing the compounds in known in vitro and in vivo systems, ing any of those described herein. Dosages for humans or other animals may then be extrapolated therefrom. With the methods of the present invention, the efficacy of the administration of one or more interventions may be assessed by any of a variety of parameters well known in the art.
In some embodiments, an “effective amount” is an amount that results in a reduction of at least one pathological parameter. Thus, for example, an amount that is effective to achieve a reduction of at least about 10%, at least about 15%, at least about 20%, or at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, or at least about 95%, compared to the expected ion in the parameter in an individual not receiving ent.
The present invention also includes s of making and using the vaccine compositions described herein. The compositions of the present sure may be formulated in pharmaceutical preparations in a variety of forms adapted to the chosen route of administration.
Any of a variety of modes of administration may be used. For example, administration may be intravenous, topical, oral, intranasal, subcutaneous, intraperitoneal, intramuscular, or intratumor.
A vaccine ition as described herein may take the form of an implant. Such an implant may be implanted within the tumor. Delivery may be lished by any of a variety of available technologies, including, for example, injection, infilsion, instillation, l application, delivery by a , and/or delivery by a catheter. Delivery may be by the use of a delivery device or tool, such as a needle or catheter. Such delivery devices are included in the present invention.
A composition of the present invention may be administered with one or more additional therapeutic interventions. Additional therapeutic treatments include, but are not limited to, surgical ion, radiation therapy, chemotherapy, hormone therapy, anti-tumor vaccines, antibody based therapies, whole body irradiation, bone marrow transplantation, eral blood stem cell lantation, the administration of cytokines, antibiotics, antimicrobial agents, ral agents, such as AZT, ddI or ddC, the administration of herapeutic agents, such as, for example, cyclophosphamide, methotrexate, 5-fluorouracil, doxorubicin, vincristine, ifosfamide, cisplatin, gemcitabine, busulfan, ara-C, adriamycin, mitomycin, cytoxan, methotrexate, and combinations thereof. Such stration may take place before, during, and/or after the administration of a vaccine ition as described.
As used herein, the term “subject” includes, but is not limited to, humans and non-human vertebrates. In some embodiments, a subject is a mammal, ularly a human. A subject may be an “individual,” “patient,” or “host.” A subject may include, for example, a human, a higher primate, a non-human primate, domestic livestock and domestic pets (such as dogs, cats, cattle, horses, pigs, sheep, goats, mules, donkeys, mink, and poultry), laboratory s (such as for example, mice, rats, hamsters, guinea pigs, and rabbits), and wild life. In some embodiments, a vaccine composition, as described herein is administered to a bovine, including, but not limited to, domestic cattle, water o, African o, bison, and yaks.
The vaccine compositions described herein may be administered to poultry, including, for example, chickens, turkeys, guinea fowl, partridges, and water fowl, such as, for example, ducks and geese. Chickens include, but are not limited to, hens, roosters, broilers, rs, r, the offspring of breeder hens, and layers. The vaccine of the present invention may be stered to poultry before or after hatching. Poultry may receive a vaccine at a variety of ages. For example, broilers may be vaccinated in ovo, at one-day-old, in ovo, or at 2-3 weeks of age.
Laying stock or reproduction stock may be vaccinated, for example, at about 6-12 weeks of age and boosted at about 16-20 weeks of age. Such laying stock or reproduction stock may be ated at about 6, at about 7, at about 8, at about 9, at about 10, at about 11, or at about 12 weeks of age. Such laying stock or reproduction stock may be boosted at about 16, at about 17, at about 18, at about 19, or at about 20 weeks of age. The offspring of such laying stock or reproduction stock may demonstrate an antibody titer to the administered immunogen(s), which may prevent or mitigate the ms of an infection in the offspring.
The compositions of the present invention may be formulated according to methods known and used in the art. A vaccine composition of the present invention may include salts, buffers, preservatives, or other substances designed to improve or stabilize the composition. A vaccine composition may include a pharmaceutically acceptable excipient or carrier. As used herein, the term “pharmaceutically acceptable carrier” refers to a substance suitable for administration to a human or other vertebrate animal. For administration, a composition as described herein may be ly buffered if necessary and the composition rendered isotonic with sufficient saline or glucose. In this connection, sterile aqueous media that can be employed will be known to those of skill in the art. Moreover, for human stration, preparations should meet sterility, pyrogenicity, and general safety and purity standards as ed by the FDA. Such preparation may be pyrogen-free, may be sterile, and/or endotoxin-free.
A composition of the present invention may also n one or more stabilizers. Any suitable stabilizer can be used including carbohydrates such as sorbitol, mannitol, starch, e, dextrin, or e; proteins such as albumin or ; and buffers such as alkaline metal phosphate and the like. A stabilizer is particularly advantageous when a dry vaccine preparation is prepared by lyophilization. Such a composition may include pharmaceutically acceptable carriers or diluents. Carriers include, for example, stabilizers, preservatives and buffers. Suitable stabilizers include, for example, SPGA, ydrates (such as sorbitol, mannitol, starch, sucrose, dextran, glutamate or glucose), proteins (such as dried milk serum, n or casein) or degradation products thereof. Suitable buffers e, for example, alkali metal phosphates.
Suitable preservatives include, for example, thimerosal, merthiolate and gentamicin. ts, include, but are not limited to, water, aqueous buffer (such as buffered saline), alcohols, and s (such as ol).
Any of a wide variety of modulating agents may be included in the methods and compositions described herein. As used herein, a "modulating agen " is an agent that has a therapeutic effect on living tissue. Modulatory agents include, for example, therapeutic agents which are effective to prevent and/or overcome disease and/or promote recovery.
The vaccine of the present ion may be administered to a subject by any ofmany ent routes. For example, the e may be administered intravenously, intraperitonealy, aneously, intranasally, orally, transdermally, and/or intramuscularly. Suitable dosing s may be determined by taking into account factors well known in the art including, for example, the age, weight, sex, and medical condition of the subject; the route of administration; the desired effect; and the particular conjugate and formulation employed. The vaccine may be administered as either a single does or multiple doses. When administered in a multi-dose vaccination format, the timing of the doses may follow schedules known in the art. For example, after an initial stration, one or more booster doses may subsequently me administered to in antibody titers and/or immunologic memory.
The methods of the t invention may e in vitro, ex vivo, or in vivo methods.
As used herein “in vitro” is in cell culture and “in vivo” is within the body of a subject. With the present invention, an isolated immunogen or agent may be delivered. As used herein, “isolated” refers to material that has been either removed from its natural nment, produced using recombinant techniques, or chemically or enzymatically synthesized, and thus is altered “by the hand ofman” from its natural state.
The present invention includes kits employing one or more of the compositions bed herein. Such kits may provide for the administration of an immunogen to a subject in order to elicit an immune response. Kits of the present invention may include other reagents such as buffers and solutions needed to ce the invention are also included. Optionally associated with such container(s) can be a notice or printed instructions. As used herein, the phrase “packaging material” refers to one or more al structures used to house the contents of the kit. The packaging material is constructed by known methods, preferably to provide a sterile, contaminant-free environment. As used herein, the term “package” refers to a solid matrix or al such as glass, c, paper, foil, and the like, capable of holding within fixed limits a polypeptide. Kits of the t invention may also e ctions for use. Instructions for use typically include a tangible expression describing the reagent concentration or at least one assay method parameter, such as the relative amounts of reagent and sample to be admixed, maintenance time periods for reagent/sample admixtures, temperature, buffer ions, and the like.
Unless otherwise indicated, all numbers expressing quantities of components, molecular weights, and so forth used in the specification and claims are to be understood as being modified in all instances by the term “about.” Accordingly, unless otherwise indicated to the contrary, the numerical parameters set forth in the specification and claims are approximations that may vary depending upon the desired properties sought to be obtained by the present ion. At the very least, and not as an attempt to limit the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.
Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. All numerical values, however, inherently contain a range necessarily resulting from the standard ion found in their respective testing measurements.
The ption exemplifies illustrative embodiments. In several places throughout the application, guidance is provided through lists of examples, which examples can be used in various combinations. In each instance, the recited list serves only as a entative group and should not be reted as an ive list.
The present invention is illustrated by the following examples. It is to be understood that the particular examples, als, amounts, and procedures are to be interpreted broadly in accordance with the scope and spirit of the invention as set forth herein. All headings are for the ience of the reader and should not be used to limit the g of the text that follows the heading, unless so specified.
EXAMPLES Example 1 New vaccination delivery regimen drives enhanced, vaccine-specific immune ses To enhance vaccine delivery over that seen when conventional delivery methods are used, this example focused on providing antigen, plus or minus CpG adjuvant, along with a component which is in liquid state at room temperature, but that forms a gel-depot under physiological conditions, after ion at 35°C. This allows for the antigen and adjuvant to be delivered in a concentrated form, enhancing antigen presenting cell activity, and leading to pro- inflammatory, vaccine-specific responses. Recombinant hepatitis B antigen (rHepBag) was used as the antigen for vaccination, and the delivery method evaluated along with seven different vaccine delivery s for the ability to induce Hepatitis B specific antibodies and cytokines.
Mice were vaccinated with rHepBag in two different types of gel slurries (PURAMATRIX, also referred to herein as “P1,” and MATRIGEL, also referred to herein as “P2”), or with rHepBag in ALHYDROGEL (aluminum salt) or rHepBAg mixed with Complete Freund's adjuvant (CFA).
Gel slurries and ALHYDROGEL were mixed +/- with the murine CpG ODN 1826.
Results showed that mice vaccinated with either gel slurry plus ODN had significantly higher TNF production 24 to 48 hours after primary ation, while P1 was significantly superior to ALHYDROGEL at 24 hours. Adjuvant P2 presented a promising Th2 inhibition after 48 hours with the reduction of IL-4, IL-5 and IL-10 levels coincident with increased antigen-specific IgG2a production in serum.
The is of vaccine-specific dies showed that P1 drove high vaccine-specific IgA, IgM and IgG titers 14 days post-prime with or without using ODN and the high IgA and IgG titers was maintained for 35 days. As both of the gel slurry systems tested in this study was superior to the conventional adjuvants, this new gel slurry vaccine delivery system will have broad utility for enhancing responses to numerous current vaccines that are currently marginally fianctional. The use of this new vaccine ry system will be further investigated in the development of es for any of a wide variety of infectious diseases, fiom parasitic to viral infection.
Material and Methods Vaccines and Route of Administration. The experimental vaccine used in this study was produced from a recombinant Hepatitis B antigen, , rHepBag (Fitzgerald Industries, Inc.
Massachusetts, USA). 90 6- to 8-week-old female BALB/c mice were evenly divided into 9 groups and respectively received a prime subcutaneous injection (sc) on the back and, a boost 4 weeks later, of 0.1 ml solution containing 5 ug of rHepBag, 0.1 ml of 50 ug ODN 1826 (InvivoGen, Inc. California, USA) with 5 ug of rHepBag, 0.4 ml solution of PURAMATRIX (P1) and 5 ug of g with or without ODN 1826, 0.4 ml of MATRIGEL (P2) and 5 ug of rHepBag with or without ODN 1826, 0.1 ml solution of 250 ug alum (Thermo Fisher Scientific, Inc. Pennsylvania, USA) and 5 ug of g with or without ODN 1826 and, 0.1 ml of te Freund's Adjuvant (Sigma-Aldrich Co. Missouri, USA) with 5 ug of rHepBag (1 :2).
To prepare one dose of the slurry, 5 ug of rHBsAg antigen, with or without prior mixing with 50 ug CpG, was brought to a final volume of 400 ul with MATRIGEL or PURAMATRIX and mixed thoroughly before subcutaneous injection. Amounts were scaled up depending on the number of doses needed. MATRIGEL and PURAMATRIX were sed from BD (Franklin Lakes, NJ).
Cytokines and antibody evaluation. Splenocytes were isolated one week after boost for cytolcines evaluation. Single cell suspensions (1.5 10 6/ml) were prepared and suspended in 1640 medium (RPMI 1640 Thermo Scientific Hyclone, Utah, USA) with penicillin—streptomycin (final trations of 100 U/ml and 100 ug/ml respectively) (Sigma—Aldrich. St. Louis, MO, USA). 0.5 ml ofthe single cell suspension was added to 48—well plates (Sigma-Aldrich. St.
Louis, MO, USA) with 0.5 ml ofmedia, 0.5 ml of l rig/ml of Concanavalin A (ConA) or 0.5 ml of 5 rig/ml ofrHepBag and cultured at 37°C with 5% C02. The levels ofTNF were quantified after 24 and 48 hours culture, IL~4 and IL-5 after 48 hours, IL—4 and lL-lO after 72 hours, each in triplicate. The percentages of cytokine-positive mice were finther transformed using a two—step platform that consisted of (l) to calculate the global median for each cytokine considering the whole range of values obtained for each group; and (2) to establish for each group the concept of 'low' and 'high'—cytokine producers using the global median percentage of cytokine—positive cells as the cut-off edge to ate the individuals into two categories named as 'low’ and 'high'— cytokine ers. It is important to highlight that the overall cytokine profile for each group was constructed by giving the same weight to all cytokines and producing cell populations.
IFN gamma ELISpot was also performed with 3 X 105 and 1.5 x 105 splenocytes after 24 hours of culture, as described by the manufacturer (BD Biosciences (San Francisco, CA, USA) using 1 rig/ml of ConA as positive control. The orming unit (SFU) value was expressed as mean ofthe triplicate cultures minus the mean value of its individual background.
Blood samples from mice were collected weekly from 1 to 6 weeks, including the day prior to the primary zation. Sera collected from these bleeds were used in ULISA assays for the detection and quantification of antibodies.
Synthetic Peptides for T—cell analysis for flow cytometry. Synthetic peptides Were synthesized by Biosynthesis, Inc, and were ed based on relevant literature. The S 228—39 peptide (IPQSLDSWWTSL; SEQ ID NO:6) is H2~Ld—restricted and the dominant e in Balb/c mice. The splenocytes from five mice per group were individually ated with 5 uM peptide and 40 U/ml IL-2 for flow cytometry.
Statistical analyses. For antibody evaluation, comparisons were ed by Mann— Whitney or t's t test using ad PRISM software, version 4.0 (GraphPad Software, California, USA), after Kolmogorov-Smimov normality test. A difference was considered as statistically significant when a P-value was 5 0.05. Chi squared-test was used for comparisons of 'low' and 'high'-cytokine producers frequencies among groups and cance considered at P S 0.05. Comparison of radar graphs axes and polygon areas were considered significant for ratios two times lower in magnitude. Data is for the results presented in the radar chart format was performed by ing the central polygon areas among cytokine-producers categories intra and inter . Significant differences were considered for ratios indicating axes and polygon areas two times lower or higher in size.
Results Initial results showed mice vaccinated with either gel slurry plus Cst had significantly higher vaccine-specific IgG2a 14 days after the prime, and IgA, IgM at 28 days post inoculation than mice vaccinated with alum or CFA. One gel slurry delivery drove significantly higher vaccine-specific IgG titers 14 days post-prime than the other delivery methods did oost, suggesting that the boost was unnecessary. Recall assays showed upregulated IL-lO and IL-4 from cytes of mice vaccinated with OGEL or CFA compared to cells from gel- slurry + CpG vaccinated mice. CpG use reduced levels of IL-5 to background in all groups compared to elevated levels in CFA. No ences in levels of IFN or TNF were seen.
Figure 1 shows the kinetics of IgA anti-HBsAg antibody titers. Data shown is pooled from two independent experiments for total n=l 0.
Figure 2 shows the kinetics of IgM anti-HBsAg antibody titers. Data shown is pooled from two independent experiments for total n=l 0.
Figure 3 shows the kinetics of IgG anti-HBsAg antibody titers. Data shown is pooled from two independent experiments for total n=l 0.
Figure 4 shows the kinetics of IgG1 anti-HBsAg antibody titers. Data shown is pooled from two independent experiments for total n=l 0.
Figure 5 shows the kinetics of Inga anti-HBsAg antibody . Data shown is pooled from two independent experiments for total n=l 0.
Figure 6 shows higher anti-HBsAg antibody titers after single vaccination (21 days).
Data shown is pooled from two independent experiments for total n=lO; 5, **p<0.0l, **p<0.00l, ****p<0.0001 compared to n alone using two-way ANOVA with Bonferroni post-test.
Figure 7 shows higher anti-HBsAg antibody titers after single vaccination (35 days).
Data shown is pooled fiom two ndent ments for total n=lO; *p<0.05 compared to antigen alone using two-way ANOVA with Bonferroni post-test.
Figure 8 shows cytokine profile twenty-four hours after HBsAg re-stimulation of splenocytes. Data shown is representative of one experiment, n=5; *p<0.05, ****p<0.0001 compared to antigen alone using two-way ANOVA with Bonferroni post-test.
Figure 9 shows ne profile forty-eight hours after HBsAg mulation of splenocytes. Data shown is pooled from two independent experiments for total n=lO (n=5 for adjuvants only); no statistical differences seen (p<0.05) compared to antigen alone using two- way ANOVA with Bonferroni post-test.
Figure 10 shows cytokine profile seventy-two hours after HBsAg re-stimulation of splenocytes. Data shown is pooled from two independent experiments for total n=lO (n=5 for adjuvants only); 0l compared to antigen alone using two-way ANOVA with Bonferroni post-test.
Figure ll shows increased HBsAg-specific cell-mediated immunity by ELISpot in specific T cell responses. Data shown is pooled from two independent experiments for total n=lO; *p<0.05 compared to antigen alone using two-way ANOVA with Bonferroni post- test.
Figure 12 shows increased HBsAg-specific T cell-mediated immunity by flow cytometry.
Data shown is pooled from two independent experiments for total n=lO; **p<0.0l compared to antigen alone using two-way ANOVA with Bonferroni post-test.
Increase of cytokines secretion by splenocytes with stimulation of rHepBag. In order to measure the cellular immune response induced by the vaccines, mice were sacrificed one week after boost and splenocytes were ed with or without 5 ug/ml final concentration of rHepBag. nes levels were measured by ELISA. The concept of low and high ers was applied to igate a broader range of cytokines and assess ex vivo cytokine profiles of circulating ytes. For this purpose, the global median for each cytokine-positive cell subset was calculated, taking the whole range of values obtained for each group, as described elsewhere (Vitelli-Avelar et al., 2008, Scand J Immunol; 68(5):5 16-25). The global median percentage of each cytokine-positive cell population was used as the cut-off edge to segregate the individuals into two categories named 'low' and 'high'-cytokine producers, as shown in Table l.
The data demonstrated that none of the mice inoculated with ODN 1826 showed TNF production in 24 hours, but when ODN 1826 was associated with adjuvants P1 or P2, a significant proportion of mice displayed values of TNF above the cut-off edge, as shown in Fig. 13 (P < 0.011). gh ALHYDROGEL, when associated or not to ODN 1826, induced a significant amount of 'high'-TNF producers, all of the mice inoculated with adjuvant Pl without CpG displayed values above the cut-off, and this value was cantly higher than all Alhydrogel groups (P < 0.001). This same result was not observed after 48 hours when adjuvants Pl, P2, ALHYDROGEL and Freund showed a comparable number of 'high'-TNF ers (Fig. 14A and 14B) (P < 0.001).
Moreover, interesting data was found for IL-4 and IL-5 production after 48 hours when most of the mice immunized with adjuvant P2 fell into a region of 'low'-IL-4 and IL-5 producers (P < 0.05). Differently, most of the mice inoculated with adjuvant Pl presented a high production of both cytokines (P < 0.002). As shown in Fig. 15, the adjuvant Pl continued to present 'high'-IL-4 ers after 72 hours, in association or not with ODN 1826 and this same result was found after ALHYDROGEL or Freund immunization (P < 0.001). Also, the data demonstrated that most of the mice previously zed with adjuvant P2 or adjuvant P2 plus ODN l826 displayed values of IL-lO below the cut-off edge, cantly lower than mice immunized with rHepBag, associated or not to ODN 1826 (P < 0.05). 95:50 mammumg >9 E on . ESEdo 9“ Now om mom Now Now ow $0-30 685338 mammomh E mammumg S @0832 ZOO “flow 38 om 0 0 0 cm :2on 630329 + 95 JomoHPEfia $5 mammumg uwo 392% $0-30 mod E .W mm» NS mm “mm NS N3 on % 2: W mammow: .va :2 :33 m 25330 mammumg + zoo Ho E gov om 8 O 0 Dom om E “BEE nZOO SEES cosmomawfim SUEZ: mammomg E 322 93 mom 9“ New Now 38 cm a mzoo 392% m mammowfi 2e 2: mammumg +mm 32 3% cm 0 0 0 a: wage“? B zoo mo .mzoo no “mammomh 393 RX. mammumg S 9“ mom om NS mom cm mom SE xEZEEE a bfiqsfififi £8.33 285on owfigobm mammomg zoo o 9“ om 0 2 2 9“ + magmaflu 959mg mnemtmmfig .boéoame $8383-:me monfio m 08 Mom on mm a 9“ ON om ON 2“ 2“ E nou E DEM 8%: ESE, 655$ mo was a .82on EV 8 up 25330 be €4-38 aw so ma am n Sam. .
ELSE So m ou *mmmmomh cm 02%. mamaomLH is souflsmom 8 8 0-0%“ 2%“ 80.0 2 0%“. omfiqoohom 39: o.8 How mo mm :8 23 >25:on SE 3E0 mafimw 750 :08 a SS Mom @532 m5 m7: «S 9d EH 338me + U 2w: 632585 :3 a? as fl 95 2an San? 632528 Ecombmsm 38332 Sustained humoral response in mice vaccinated with rHepBag and adjuvants Pl and P2.
Sera were collected weekly before and after zation to test c isotypes antibody titer by ELISA. Mice vaccinated with both adjuvants Pl and P2 developed an anti-rHepBag IgG antibody in 2 weeks after prime inoculation (P < 0.002). The highest anti-rHepBag antibody titers were reached in mice vaccinated with Pl associated or not to ODN, and these responses were significantly greater than immunization with ODN, Alum or Freund's adjuvants (P < 0.001). The IgGl :IgG2a ratio elicited in vivo represents different patterns for both adjuvants.
Adjuvant Pl drives to a Th2 response with high levels of IgGl in 14 to 35 days post-prime inoculation. Whereas, the response elicited by P2 plus ODN is a mixed system rather than a pure Thl or Th2 response, wherein the IgGl and IgG2a levels were upregulated during all the ne. See Fig. l7A-l7D.
The combination of adjuvant Pl +/- ODN d the upregulation of IgA and IgM titers as soon as 14 days post-prime inoculation and this humoral response was maintained until 35 and 21 days, respectively (P < 0.04). This adjuvant was superior to Freund's during 14 and 35 days and to ALHYDROGEL for the first 3 weeks for the production of IgA, IgM and IgG (P < 0.02).
Additionally, both adjuvants Pl and P2 demonstrated to be superior to Freund's adjuvant for the production of all Ig after boost (P < 0.02). See Figs.l6A-l6D.
Discussion This e showed that mice ated with either a Pl or P2 gel slurry plus ODN had significantly higher TNF tion 24 to 48 hours after primary inoculation, while Pl was significantly superior to ALHYDROGEL at 24 hours. nt P2 presented a promising Th2 inhibition after 48 hours with the reduction of IL-4, IL-5 and IL-lO levels coincident with increased antigen-specific IgG2a production in serum.
The analysis of vaccine-specific antibodies showed that Pl drove high vaccine-specific IgA, IgM and IgG titers 14 days post-prime with or without using ODN and the high IgA and IgG titers was maintained for 35 days. As both of the gel slurry s tested in this study were superior to the conventional adjuvants, this new gel slurry e delivery system will have broad utility for enhancing responses to numerous current vaccines that are currently marginally fianctional. The use of this new vaccine ry system will be r investigated in the development of vaccines for any kind of infectious diseases, from parasitic to viral infection.
Example 2 Influenza Vaccination Following methods described in more detail in the previous examples, C57/BL and Balb/c mice were immunized with the recombinant nucleoprotein (rNP) influenza virus antigen administered with alum, CpG, or a slurry of PURAMATRIX and CpG. To e one dose of the slurry, 10 ug rNP antigen, with or without prior mixing with 50 ug CpG, was t to a final volume of 200 ul with PURAMATRIX and mixed thoroughly before subcutaneous injection. Amounts were scaled up depending on the number of doses needed. As shown in Fig. 18B, anti-influenza IgG titers were increased Balb/c mice vaccinated with rNP administered as a slurry with PURAMATRIX and CpG, as compared to mice vaccinated with rNP administered with the adjuvant alum or CpG. Fig. 18A shows anti-influenza IgG titers in C57BL/6 mice.
Anti-influenza IgG titers were determined four weeks post-last vaccination (wplv).
Again, ing methods described in more detail in the previous examples, C57Bl/6 mice were immunized with whole inactivated influenza A virus (HlNl) strain PR8 administered with alum, as a PURAMATRIX slurry, or as a slurry ofPURAMATRIX and CpG. As a control, additional mice were zed with PR8 whole inactivated virus (WIV) only. To prepare one dose of the , 15 ug of PR8 antigen, with or without prior mixing with 50 ug CpG, was brought to a final volume of 200 ul with PURAMATRIX and mixed thoroughly before subcutaneous injection. Amounts were scaled up ing on the number of doses needed.
PR8 (WIV) is formalin-inactivated influenza /34 (HlNl) from Charles River rNP is inant human Influenza A (A/PlV8/34/Mount Sinai (HlNl) segment 5) nuclear protein NP from Imgenex.
As shown in Fig. 19, serum anti-influenza IgG titers were increased in C57Bl/6 mice vaccinated with PR8 WIV administered with as a slurry of PURAMATRIX and CpG, as compared to mice vaccinated with PR8 WIV administered with the adjuvant alum or as a slurry with PURAMATRIX alone, without CpG. Anti-influenza IgG titers were determined four weeks post-last vaccination (wplv). Fig. 19 shows results from two independent experiments.
As show in Fig. 20, enhanced tion from lethal challenge was observed in C57Bl/6 mice vaccinated with PR8 WIV. Fig. 20A shows results from an ndent experiment with lethal challenge at 30 LD50 and Fig. 20B shows results from an independent experiment with lethal nge at 1000 LD50.
Example 3 Burkholderia Vaccination Following methods described in more detail in the previous e, mice were immunized with a cocktail of three different burkholderia recombinant ns (burkholderia 4- 9 protein, the burkholderia 22-ll protein, and the burkholderia 42 protein) administered with one of three ent adjuvants preparation, alum, Complete Freund’s Adjuvant (CFA), or slurry of PURAMATRIX and CpG. To prepare one dose of the PURAMATRIX + CpG slurry, 75 ug of Burkholderia proteins antigen, with or without prior mixing with 50 ug CpG, was brought to a final volume of 200 ul PURAMATRIX and mixed thoroughly before subcutaneous injection. s were scaled up ing on the number of doses needed. As controls, additional mice were immunized with alum only, CFA only, or PURAMATRIX + CpG slurry, without antigen.
The Burkholderz’a (previously part ofPseudomonas) genus name refers to a group of virtually tous gram-negative, motile, obligatory aerobic rod-shaped bacteria including both animal/human and plant pathogens as well as some environmentally important species.
Burkholderia is best known for its pathogenic members. Burkholderz’a mallez' is responsible for rs, a disease that occurs mostly in horses and related animals. Burkholderia pseudomallei is the causative agent of dosis (also called re's disease), an infectious disease predominately of tropical climates that can infect humans or animals, especially in Southeast Asia and northern Australia. Burkholderz’a cepacz’a is an important pathogen of pulmonary infections in people with cystic fibrosis. Due to their otic ance and the high mortality rate from their associated diseases Burkholderz’a mallez' and Burkholderia pseudomallei are considered to be potential biological warfare agents, targeting livestock and humans.
Fig. 21 shows anti-burkholderia IgG titers in mice immunized with a cocktail of three burkholderia recombinant proteins (burkholderia 4-9 protein, the burkholderia 22-ll protein, and the burkholderia 42 protein). Fig. 21B shows anti-burkholderia protein 4-9 IgG titers in immunized mice. Fig. 21A shows anti-burkholderia protein 22-ll IgG titers in immunized mice.
Fig. 21C shows anti-burkholderia protein 42 IgG titers in zed mice. Antibody titers after immunization with alum only, alum + protein cocktail, complete Freund’s adjuvant (CFA) only, CFA + protein cocktail, TRIX + CpG slurry only, and PURAMATRIX gel + protein il are shown. An increased immune response as ed by specific serum antibody levels were observed against two of the three proteins administered as a gel vaccine ition with puramatrix compared with vaccination with alum or CFA. Specifically, increased anti- burkholderia IgG titers were observed t the burkholderia 4-9 protein (Fig. 21B) and the burkholderia 22-11 protein (Fig. ZlAB) when administered as a gel vaccine with PURAMATRIX, compared with vaccination with alum or CFA. Challenge data is being Example 4 Veterinary Vaccines ing procedures described in more detail in the previous examples, the present invention may be used with any of a variety of veterinary vaccines. The vaccine compositions, delivery methods, and delivery system of the present invention will provide many advantages and better value for the commercial livestock ry, ing, but not limited to, improved efficacy, delivery early in the production cycle, and efficacy with the administration of only a single dose to provide protection throughout the production cycle.
The compositions, delivery methods, and delivery systems of the present invention may be used in the zation of swine. Vaccines that may be administered to swine using the compositions, methods and systems of the present invention include, but are not limited to, porcine circovirus type 2 (PCV2) vaccine, porcine reproductive and respiratory syndrome (PRRSV), respiratory mycoplasma vaccine, Streptococcus suis vaccine, porcine coronavirus vaccine, rotavirus vaccine, enterotoxigenic Escherichia coli (K88) vaccine, bacz'llus pleurapneumom’a (APP) vaccine, and swine influenza vaccine. See, for example, the world wide web at merck—animal—health.com/s ecies/ igsx’vaccinesas x, “Vaccinations for the Swine Herd,” Alabama Cooperative Extension System Publication 2, Alabama A&M and Auburn Universities (available on the world wide web at aces.edu/pubs/docs/A/ANR-0902/ ANR- 0902.pdf), and “Pig vaccination ms,” PRIME FACT publication 944, September 2009 (available on the world wide web at dpi.nsw.gov.au/_data/assets/pdf_file/0009 /301500/Pig- vaccination-programs.pdf) for more detailed information on available vaccines and the administration of such vaccines to swine.
The compositions, delivery methods, and delivery s of the present invention may be used in the immunization of bovine, including, but not limited to, domestic cattle, water buffalo, African buffalo, bison, and yaks. Vaccines that may be administered to bovines, using the compositions, methods and s of the present invention include, but are not limited to, bovine respiratory disease (BRD) vaccine, including, but not limited to BVDV types I and 11, bovine herpes virus 1 (BHV-l) vaccine, including, but not limited to, subunit vaccines that would not result in latent virus, Haemophilus somnus vaccine, Mannheimia haemolytica vaccine, Mycoplasma bovis vaccine, bovine rus vaccine, Escherichia coli K99 vaccine, bovine coronavirus (BCV) e, Clostridium chauvoei (black leg) vaccine, Clostridium septicum vaccine, Clostridium sordelli (malignant edema) vaccine, Clostridium novyi (black disease) vaccine, Clostridium perfringens (enterotoxemia) vaccine, ious bovine keratoconjunctivitis (pink eye) vaccine, including, but not limited to, Moraxella bovis, chlamydia, mycoplasma, acholeplasma, or infectious bovine rhinotracheitis (1BR) virus vaccines, mastitis vaccines, including, but not limited to, Escherichia coli J5 e.
In some applications, the compositions, delivery s, and ry systems of the present invention may be used for the administration of one or more schistosomiasis antigens to a bovoid. Such a schistosome n may be derived from, for example, Schistosoma japonicum, Schistosoma monsoni, or Schistosoma haematobium. A schistosome antigen may be a schistosome triose phosphate isomerase (CTPI) protein, or nic nt or derivative thereof, including, but not limited to a S. japonicum, S. monsoni, or S. haematobium CTPI protein, or antigenic fragment or derivative thereof. A schistosome antigen may be a schistosome tetraspin 23 kDa integral membrane protein (C23), or antigenic fragment or derivative thereof, including, but not limited to a S. japonicum, S. monsoni, or S. haematobium C23 protein, or antigenic fragment or tive thereof Such a schistosome antigen may be a chimeric polypeptide, fiased to one or more onal antigenic determinants, such as for example, a heat shock n, or antigenic fragment or tive thereof, including, but not limited to, bovine heat shock protein 70 (Hsp70).
See, for example, “Beef Cattle Herd Health Vaccination le” Powell et al., sity of Arkansas, Division of lture, Agriculture and Natural Resources publication FSA3009 (available on the world wide web at uaex.edu/Other_Areas/publications/PDF/FSA- 3009.pdf), “How to ate,” ma Cooperative Extension Service, Division of Agricultural Sciences and Natural Resources, Publication No. 350 (available on the world wide web at ansci.colostate.edu/pdf_files/YLE/Dairy7_vaccinate.pdf), and “Cattle Vaccines and Their Use,” Beef Cattle Handbook publication BCH-3015 (available on the world wide web at iowabeefcenter.org/Beef%20Cattle%20Handbook/Vaccines_Cattle.pdf) for more detailed information on available vaccines and the stration of such vaccines to cattle.
The vaccine compositions, delivery methods, and delivery system of the present invention may also be used in the vaccination of companion animals, including, but not limited to cats and dogs, such as, for example, for the immunization of dogs with irus vaccine for transfer of maternal immunity.
Routes of administration include, but are not limited to, subcutaneous (sc) or intramuscular (im) ion. Vaccination with the composition, methods, and ry systems of the present invention will yield rapid, longer g protection after administration of a single dose.
Example 5 Poultry Vaccines ing procedures described in more detail in the previous examples, the present invention may be used as a delivery system for any of a variety of poultry vaccines, including, but not limited to, vaccines for infectious bronchitis (1B), Newcastle disease (ND), Marek’s diseases, infectious bursal disease (IBD) virus, infectious laryngotracheitis (1LT), avian reovirus, cholera, fowl pox, asmosis, turkey and n Coryza, avian influenza, avian encephalomyelitis (AE), avian rhinotracheitis (ART), duck virus hepatitis, haemorrhagic enteritis, goose parvovirus, Paramyxovirus 3, chicken anaemia virus (CAV), E. coli, Erysipelas, Reimerella, asma galliseptz'cam, Pastearella maltocz'da, Salmonella enteritidis, Salmonella typhz'marz’am, and coccidiosis.
Routes of administration include, but are not limited to, subcutaneous (sc) or intramuscular (im) injection. With subcutaneous ion, vaccine is injected into the space between the skin and underlying tissues. Typically the site of application used in poultry in the loose skin at the back of the neck. With intramuscular injection, the vaccine preparation is deposited within a mass of muscle. Typically, either the breast muscle or the thigh muscle is used for this purpose. Vaccination may include the vaccination of day old hatchlings, s, layers, breeders, broilers, and/or show birds. lly layers and breeders get vaccinated prior to the laying , and then every 6 to 8 weeks with inactivated vaccines during the laying period. This not only protects them from disease but passes on maternal antibodies to the progeny. Poultry that may be vaccinated include, but are not limited to, chickens, turkeys, and waterfowl, such as, for example, ducks and geese.
Example 6 Schistosomiasis Immunization Mice were immunized with the schistosome protein antigen CCA in either complete Freund’s adjuvant or in MATRIGEL plus Cst. Anti-CCA IgG antibody titers were ined by ELISA The data is shown in Fig. 22A (CCA in complete Freund’s adjuvant) and Fig. 22B (CCA in EL plus Cst). Two mice were immunized for each n preparation.
Sera samples were collected at 0, 8, l6, and 19 days post immunization for mice immunized with CCA in te ’s adjuvant and at 0, 8, 16, 24, 32, and 40 days post immunization for mice immunized with CCA in MATRIGEL plus Cst. The ELISA data clearly shows higher titers of CCA specific antibodies in mice immunized with MATRIGEL plus Cst.
Example 7 Schistosomiasis Vaccination of Water Buffalo Schistosomiasis is a parasitic disease affecting more than 200 million people worldwide.
Reassessment of schistosomiasis-related disability, combined with recent ation on the global prevalence of schistosome infection indicates that the true burden of osomiasis is substantially greater than previously appreciated. In Asia, ularly China, the causative agent is Schistosomajaponicum. Unlike the African species, S. mansoni and S. haematobium, S. japom'cum is a zoonotic parasite, with bovines, particularly water buffaloes accounting for about 75% of schistosome transmission to humans in China. Interventions that reduce schistosome infection in water buffaloes will enhance their health simultaneously reducing disease transmission to humans. Current control programs in many areas of China include simultaneous uantel (PZQ) ent of humans and water buffaloes; while this has shown a reduction in the overall prevalence, it requires continued mass ents that are both time consuming and expensive. A more sustainable option would be development of a vaccine which reduces transmission of S. japanicum from bovines to replace bovine herapy. Indeed mathematical modeling (Williams et al., 2002, Acta Trap; 82(2):253-262) has demonstrated that reducing S. japanicum infection in bovine reservoirs using prophylactic es with 45% cy alone or in combination with PZQ should over time reduce the equilibrium prevalence and potentially lead to long-term sustainable control of osomiasis. This two-pronged base intervention would significantly reduce ission of schistosomiasis for the long term, increase bovine health and growth and would likely reduce l ity in village populations. See Da’Dara et al., 2008, Vaccine; 26(29-30):36l7-3625, which is herein incorporated by reference in its entirety.
Antigen-PURAMATRIX compositions as described herein will be used to immunize livestock with S. japonicum antigens. For these trials, all animals will be given a primary vaccination with a SjCTPI-Hsp70 plasmid DNA vaccine (as described in more detail in Da’Dara et al., 2008, Vaccine; 26(29-30):36l7-3625). Water buffalo will be boosted with a composition of recombinant SjCTPI protein, PURAMATRIX and bovine CpG. Specifically, 100 ug of recombinant SjCTPI plus bovine CpG will be mixed in PURAMATRIX for a total injection volume of approximately 0.50 ml/animal. This will be injected into the shoulder of buffalo/and cattle. Only a single booster vaccination will be stered to an .
The elicitation of a l and cellular immune responses, including anti-SjCTPI IgG antibody response, will be determined. After boosting, animals will be challenged were challenged with cercariae and vaccine efficacy determined by measuring the reduction in the number of eggs per gram feces, reduction in eggs in liver tissues, reduction in miracidial hatching, and reduction in worm burden. Methods for these determinations are described in more detail in a et al., 2008, Vaccine; 26(29-30):36l7-3625.
A first trial in the pines is already in Year 2 in the Philippines, with 400 water buffalo or cattle having been boosted, as described above. A second trial in Samar, Philippines, will include 1500 water buffalo or cattle. A third trail in China will include 600 water buffalo or cattle.
Immunization of water buffalo and cattle with compositions of schistosome polypeptides, such as for e, SjCTPI, SjCTPI-Hsp70, SjC23, or SjC23-Hsp70 polypeptides, in a puramatrix composition, with or t adjuvants, such as for example, CpG or IL-l2, may serve as the basis for new control programs for schistosomiasis in Asia. Such a program, in addition to treatment with uantel (PZQ), would include vaccination of water buffaloes with partially protective vaccines such SjC23-Hsp70 and SjCTPI-Hsp70 as a means to reduce numbers of egg-laying parasites in ock, g to measurable declines in prevalence, intensity and transmission of S. japom'cum.
The complete disclosure of all patents, patent applications, and publications, and electronically available material (including, for instance, nucleotide sequence submissions in, e. g., GenBank and RefSeq, and amino acid sequence submissions in, e.g., rot, PIR, PRF, PDB, and translations from annotated coding regions in GenBank and RefSeq) cited herein are incorporated by reference. In the event that any inconsistency exists between the disclosure of the present application and the disclosure(s) of any document incorporated herein by reference, the sure of the present ation shall govern. The foregoing detailed description and examples have been given for clarity of understanding only. No unnecessary limitations are to be tood therefrom. The ion is not limited to the exact details shown and described, for variations obvious to one skilled in the art will be included within the invention defined by the . All headings are for the convenience of the reader and should not be used to limit the meaning of the text that follows the heading, unless so specified.
Claims

Claims (31)

What is claimed
1. A composition comprising: a self assembling peptide that forms a stable hydrogel at physiological salt concentrations; 10 an adjuvant, wherein the adjuvant comprises a Toll-Like Receptor (TLR) agonist; and one or more antigens.
2. The composition of claim 1, wherein the self assembling peptide is selected from the group consisting of RADARADARADARADA (SEQ ID NO:2), RA16-II (SEQ ID NO:3), 15 KFE-8 (SEQ ID NO:4), and KLD-12 (SEQ ID NO:5).
3. The composition of claim 2, wherein the self assembling peptide comprises RADARADARADARADA (SEQ ID NO:2). 20
4. The composition of any one of claims 1 to 3, wherein the antigen comprises a bacterial antigen, a viral n, a parasitic antigen, a toxin, and/or an anti-tumor vaccine component.
5. The composition of any one of claims 1 to 4, wherein the TLR agonist comprises a TLR4 and/or TLR9 agonist.
6. The composition of claim 5, n the TLR9 agonist comprises a CpG oligodeoxynucleotide (ODN).
7. The ition of any one of claims 1 to 6, wherein the n comprises a tis 30 antigen, an influenza antigen, a schistosomiasis antigen, and/or a lderia antigen, or an antigenic fragment thereof.
8. Use of a ition of any one of claims 1 to 7 in the manufacture of a ment for producing an immune response in a subject.
9. Use of a ition of any one of claims 1 to 7 in the manufacture of a medicament for 5 immunizing a subject.
10. Use of a composition of any one of claims 1 to 7 in the manufacture of a medicament for treating a subject with one or more immunogenic antigens. 10
11. Use of a composition of any one of claims 1 to 7 in the manufacture of a medicament for treating a subject with one or more therapeutic antigens.
12. A method of delivering one or more immunogenic antigens to domestic livestock or a companion animal, the method sing administering a composition of any one of claims 1 to 15 7 to the domestic livestock or the companion animal.
13. A method of delivering one or more immunogenic antigens to poultry, the method comprising administering a composition of any one of claims 1 to 7 to the poultry. 20
14. Use of a composition of any one of claims 1 to 7 in the manufacture of a medicament for treatment involving subcutaneous (sc) injection and/or uscular (im) injection.
15. Use of a composition of any one of claims 1 to 7 in the manufacture of a medicament for a primary and/or booster ation.
16. Use of a composition of any one of claims 1 to 7 in the manufacture of a medicament for a booster vaccination.
17. Use of a composition of any one of claims 1 to 7 in the manufacture of a medicament for 30 a booster vaccination after a primary vaccination with a polypeptide vaccine or a d DNA vaccine.
18. A method of producing an anti-schistosome immune response in a bovoid, the method comprising stering a composition of any one of claims 1 to 10 to the bovoid, wherein one or more antigen comprises a schistosome antigen.
19. A method of osomiasis vaccination in a , the method comprising administering a composition of any one of claims 1 to 10 to the bovoid, wherein the one or more antigen ses a schistosome antigen. 10
20. The method of any one of claims 18 or 19, wherein the composition further comprises one or more adjuvants.
21. The method of any one of claims 18 to 20, wherein the composition further ses a Toll-Like Receptor (TLR) agonist and/or a cytokine.
22. The method of claim 21, wherein the TLR agonist comprises a TLR4 and/or TLR9 agonist.
23. The method of claim 22, wherein the TLR9 agonist comprises a CpG 20 oligodeoxynucleotide (ODN).
24. The method of claim 23, wherein the CpG ODN comprises bovine CpG.
25. The method of any one of claims 20 to 24, n the adjuvant comprises IL-12.
26. The method of any one of claims 18 to 25, wherein the schistosome antigen comprises a Schistosoma japonicum antigen, or an antigenic fragment thereof.
27. The method of any one of claims 18 to 26, wherein the schistosome antigen comprises a 30 SjCTPI polypeptide, a SjCTPI-Hsp70 polypeptide, a SjC23 polypeptide, and/or a SjC23-Hsp70 polypeptide, or an antigenic fragment thereof.
28. The method of any one of claims 18 to 27, wherein administration of the composition comprises a primary and/or a booster vaccination. 5
29. The method of any one of claims 18 to 28, wherein administration of the composition comprises a r administered after a primary vaccination with a SjCTPI-Hsp70 plasmid DNA vaccine.
30. The method of any one of claims 18 to 29, r comprising the administration of one 10 or more anti-schistosome chemotherapeutic agents.
31. The method of any one of claims 18 to 30, demonstrating at least 45% efficacy in the prevention of infection with a osome parasite.
NZ616804A 2011-04-18 2012-04-18 Vaccine delivery method NZ616804B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161476431P 2011-04-18 2011-04-18
US61/476,431 2011-04-18
PCT/US2012/034012 WO2012145355A1 (en) 2011-04-18 2012-04-18 Vaccine delivery method

Publications (2)

Publication Number Publication Date
NZ616804A NZ616804A (en) 2015-12-24
NZ616804B2 true NZ616804B2 (en) 2016-03-30

Family

ID=

Similar Documents

Publication Publication Date Title
US11524072B2 (en) Vaccine delivery method
Burakova et al. Adjuvants for animal vaccines
JP5824538B2 (en) Novel adjuvant composition
TWI507214B (en) Emulsion vaccine compositions comprising antigen and adjuvant in the aqueous phase
EP2004220B1 (en) Methods and compositions for vaccination of poultry
EP2331128B1 (en) Composition comprising chitosan for ocular administration of vaccine(s) to avians
JP2015535259A (en) Novel mucosal adjuvant and delivery system
Akache et al. Adjuvants: Engineering protective immune responses in human and veterinary vaccines
Makwana et al. Role of adjuvants in vaccine preparation: A review
NZ616804B2 (en) Vaccine delivery method