US20040043032A1 - Vaccine - Google Patents

Vaccine Download PDF

Info

Publication number
US20040043032A1
US20040043032A1 US10/297,256 US29725603A US2004043032A1 US 20040043032 A1 US20040043032 A1 US 20040043032A1 US 29725603 A US29725603 A US 29725603A US 2004043032 A1 US2004043032 A1 US 2004043032A1
Authority
US
United States
Prior art keywords
antigen
immune response
mice
llo
mucosal
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/297,256
Other languages
English (en)
Inventor
lan McKenzie
Geoffrey Pietersz
Christina Cheers
John Stambas
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Austin Research Institute
University of Melbourne
Original Assignee
Individual
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 Individual filed Critical Individual
Assigned to STAMBAS, JOHN, UNIVERSITY OF MELBOURNE, THE, AUSTIN RESEARCH INSTITUTE, THE reassignment STAMBAS, JOHN ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEERS, CHRISTINA, STAMBAS, JOHN, MCKENZIE, IAN F. C., PIETERSZ, GEOFFREY A.
Publication of US20040043032A1 publication Critical patent/US20040043032A1/en
Assigned to STAMBAS, JOHN (24.5%), CHEERS, CHRISTINA (0.5%) reassignment STAMBAS, JOHN (24.5%) RECORDAL OF AN INTEREST Assignors: STAMBAS, JOHN
Assigned to MELBOURNE, UNIVERSITY OF, THE (25.0%), AUSTIN RESEARCH INSTITUTE, THE (50.0%) reassignment MELBOURNE, UNIVERSITY OF, THE (25.0%) RECORDAL OF AN INTEREST Assignors: AUSTIN RESEARCH INSTITUTE, THE, CHEERS, CHRISTINA, MELBOURNE, UNIVERSITY OF, THE, STAMBAS, JOHN
Priority to US12/813,982 priority Critical patent/US20110033493A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • 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
    • A61K39/04Mycobacterium, 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
    • A61K39/12Viral 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
    • A61K39/15Reoviridae, 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
    • A61K39/385Haptens or antigens, bound to carriers
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/18Feminine contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/543Mucosal route intranasal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55544Bacterial toxins
    • 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
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55583Polysaccharides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6087Polysaccharides; Lipopolysaccharides [LPS]
    • 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
    • C12N2710/20011Papillomaviridae
    • C12N2710/20034Use 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
    • 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
    • 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

Definitions

  • the present invention relates to a method of vaccination comprising the step of administering at a mucosal site a composition comprising a carbohydrate polymer and an antigen. It also relates to a composition used in the vaccination and to a vaccine adapted for administration at a mucosal site.
  • the oral polio vaccine remains the only successful large-scale vaccine that gives protection at the mucosal level. It is a live vaccine whose attenuation was achieved empirically with the attendant risks of back-mutation. In contrast, immunisation with defined protein antigens would have many advantages, including safety in immunosuppressed individuals. However, before such vaccines can be developed there is a need for adjuvants and delivery systems to overcome the problems associated with mucosal vaccination using defined protein antigens, including poor immunogenicity or the induction of tolerance.
  • Bacterial enterotoxins such as cholera toxin (CT) produced by Vibrio cholerae , and labile toxins from Esherichia coli have been co-administered with antigens and act as adjuvants with antigens [5,6].
  • CT cholera toxin
  • these adjuvants pose health risks due to their toxic properties, and for the most part, cannot be used in human trials, and have limited usefulness for human vaccination.
  • cholera toxin Wiedermann, John-Schmid and Lindblad et al [7] showed that the tolerogenic or immunogenic properties for the B subunit of cholera toxin depended on the nature of the antigen/allergen to which it is coupled.
  • CT in addition to its toxicity, CT suffers from a major disadvantage in that it may not be effective as an adjuvant, but rather, lead to tolerance.
  • Vaccines containing DNA encoding the antigen and low-viscosity carboxymethylcellulose sodium salt (CMCS-L) as a vehicle to carry the DNA to its site of action have been studied (9].
  • CMPL monophosphoryl lipid A
  • Adjuvants which have been administered with immunogens via the mucosa have therefore consisted of potentially dangerous bacterial or viral derivatives and display variable degrees of adjuvanticity.
  • the development of efficacious mucosal vaccines has further been hindered by incomplete understanding of mechanisms of pathogen transmission, and the immune responses specific to each pathogen.
  • development of a potent mucosal HIV vaccine has been affected by lack of understanding of mucosal HIV infection, and the immune responses that control the infection.
  • passive administration of IgG can confer protection against HIV infection at a mucosal site. However, the protection is limited, in that it appears unlikely that the protection is totally via neutralisation of the initial cell infection.
  • Mannan a polymannose or polysaccharide derived from the cell wall of yeast, when oxidised and conjugated to human Mucin 1 (an overexpressed cancer antigen) has been used in mice to induce immune responses [12, 13]. Intraperitoneal immunisation resulted in the induction of cellular immune responses as shown by production of CTLs and their precursors, Th1 cytokines IFN- ⁇ and IL-12. Antibody production was usually low. In tests in more than 100 patients, mannan has not shown any obvious toxicity or autoimmunity [14].
  • Mannose receptors which bind mannan have so far been identified on macrophages and dendritic cells. Oxidised mannan has been shown to stimulate production of interleukin 12 in macrophages, and also to stimulate T-cells, and to cause rapid trafficking of antigens to the class I pathway to produce cytotoxic T-cells in mice [15].
  • the invention relates to a method of immunising a subject, comprising the step of administering a composition comprising:
  • a carbohydrate polymer comprising mannose to a mucosal site.
  • the immunisation preferably results in a humoral or cellular immune response. Preferably, it results in a humoral response. More preferably, it results in a humoral response wherein one or more of IgA, IgG, IgM and optionally, IgE antibody production is stimulated. In some instances, stimulation of IgE production may be beneficial, eg to immunise against worm infections.
  • a reduction in total IgE production, or a reduction in the level of IgE relative to other antibody classes can be beneficial, e.g. to prevent or reduce type I hypersensitivity or atopy, e.g.hayfever, asthma attacks or food and other allergies.
  • IgE binding to its receptor and subsequent cross-linking with allergen is responsible for triggering immune responses underlying conditions such as asthma including atopic asthma, allergic rhinitis and atopic dermatitis, which are health problems of epidemic proportions.
  • the immune response is such that IgE production is reduced.
  • the IgE titre relative to one or more of IgA, IgG, IgM or subclasses of these is reduced.
  • IgE production may be unchanged, or be substantially unchanged, whilst the production of one or more of the other antibodies is increased upon immunization with the composition.
  • IgA production is stimulated, and the titre of IgA at one or more mucosal areas, and/or in the serum, is increased.
  • IgA production upon immunization is greater when compared with production of IgG, IgM and IgE.
  • the immunisation selectively stimulates production of one or more of IgA, IgG and IgM over IgE.
  • the immunoglobulins may include one or more of the subclasses within each class of antibody, for instance IgG2a and IgG1.
  • immunization results in greater production of IgA relative to the increase in IgG1 and/or IgG2a production.
  • the levels of antibody may be increased in serum or at mucosal sites, or both.
  • the subject immunised may be a human or an animal.
  • the invention may be used to prevent and/or treat animal diseases.
  • the antigen may be any pathogen known to cause diseases or infections in any animal species that include but are not limited to farm animals such as pigs, cattle, sheep, horses, and domestic animals and/or household pets such as cats and dogs, exotic animals such as zoo animals and feral animals.
  • the antigen may also be an antigenic part of any of the pathogens.
  • infections or diseases that may be prevented or treated in accordance with the invention include but are not limited to those described in “Hagen and Bruner's Microbiology and Infectious Diseases of Domestic animals: with reference to etiology, epizootiology, pathogenisis, immunity, diagnosis and antimicrobial susceptibility”, WA Hagen, Comstock Pub. Associates, 1988, which is incorporated herein in its entirety by this reference.
  • the invention relates to a method of immunising a subject comprising the step of administering a composition comprising:
  • a carbohydrate polymer comprising mannose to a mucosal site thereby to stimulate a secretory immune response.
  • the secretory immune response is preferably IgA immune response at one or more mucosal sites. Stimulation of secretory immune responses at these sites is particularly advantageous, as it can assist in neutralisation of pathogens or infectious agents upon their entry into the body through mucosal membranes.
  • the mucosal site may be a region or any area, from any mucosal surfaces, e.g. those lining the oral cavity or tissues, including the teeth and gingivae, those lining the gastrointestinal tract, or those lining the nasal passages and lungs, and the reproductive tract/tissues.
  • the conjunctiva of the eyes also provides a suitable surface for administration of the composition.
  • mucosal sites or surfaces include but are not limited to the respiratory tract such as the nasal region (e.g. the nose), the trachea, bronchi and the lungs, the buccal or oral tissues including the oral (e.g. the mouth and gingivae) and oro-pharyngeal cavities, the throat including the tonsils, the gastrointestinal tract (e.g.
  • An increase in antibody production in the male and female urinary and reproductive tracts is also contemplated and includes but is not limited to the bladder, ureter, urethra and associated tissues, the penis, the vulva/vagina and cervico-vaginal tissues, as well as the uterus and fallopian tubes.
  • antibody production is increased in the respiratory tract such as the nasal region (e.g. the nose), the trachea, bronchi and the lungs, the buccal or oral tissues including the oral (e.g. the mouth and gingivae) and oro-pharyngeal cavities, the throat including the tonsils, the gastrointestinal tract (e.g. oesophagus, stomach, duodenum, small and large intestine, colon and rectum).
  • the respiratory tract such as the nasal region (e.g. the nose), the trachea, bronchi and the lungs, the buccal or oral tissues including the oral (e.g. the mouth and gingivae) and oro-pharyngeal cavities, the throat including the tonsils, the gastrointestinal tract (e.g. oesophagus, stomach, duodenum, small and large intestine, colon and rectum).
  • an increase in antibody production in the male and female urinary and reproductive tracts is also contemplated and these include but is not limited to the bladder, ureter, urethra and associated tissues, the penis, the vulva/vagina and cervico-vaginal tissues, as well as the uterus and fallopian tubes.
  • the titre of IgA upon immunization is increased in a mucosal region selected from the group consisting of the lungs, nasal region, throat, gut, and male and female urinary and reproductive tracts.
  • the titre of one or more of IgA, IgG, IgM and, optionally, IgE is increased.
  • the level of one or more of IgA, IgG or IgM is increased to a greater degree than IgE.
  • the immunoglobulins may include one or more sub-classes within each class of antibody.
  • the titre of IgA is increased upon immunization, to a greater degree than increases in IgG and IgM.
  • the increase in serum IgA upon immunization is greater than the increase in IgG1 and/or IgG2a.
  • the invention relates to a method of modulating an immune response at a mucosal site in a subject, comprising the step of administering a composition comprising:—
  • a carbohydrate polymer comprising mannose to a mucosal site.
  • the immune response is preferably modulated so that antibody production, in response to administration of the composition, is selected from the group consisting of IgA production, IgG production, IgM production and IgE production.
  • the immune response may also be modulated by selectively inducing a desired type of antibody response, for example, by increasing its level of production so that a greater amount is produced when compared with another class or subclass of antibody.
  • the immune response is modulated by inducing an increase in production of one or more of IgA, IgG and IgM relative to that of IgE. More preferably, the immune response is modulated by increasing production of IgA relative to that of IgG, IgM and/or IgE.
  • the immune response is modulated such that IgA production is increased relative to the production of IgG, particularly IgG1 and/or IgG2a.
  • the immune response may also be modulated by inducing production of antibodies whose presence in turn modulates response to other classes of antibodies.
  • the immune response is modulated such that production of one or more of IgA, IgG1, IgG2a or IgM attenuates or inhibits the action of IgE.
  • the production of one type of antibody stimulates the action of another class of antibody.
  • the immune response is preferably modulated so that mediators of cellular immunity are stimulated, such as Th1 and/or Th2.
  • the mucosal or secretory immune response is stronger than the systemic immune response.
  • the secretory immune response is an IgA response
  • the systemic immune response is IgG response.
  • the antigen which evokes the immune response may include and is not limited to all or immunogenic portions of pollens, allergens especially those that induce asthma, bacteria, viruses, yeasts, fungi, protozoa and other microorganisms, or pathogens of human, animal or plant origin. Derivatives of these antigens are also contemplated and may be homologues, analogues, conjugates or salts of such entities.
  • the antigen is selected from the group consisting of influenza virus, haemagglutinin of influenza, Porphyromonas gingivalis (which causes gum disease and cardiovascular disease), proteinase and adhesin epitopes of Porphyromona gingivalis, Helicobacter pylori, urease of Helicobacter pylori, rotavirus, recombinant outer capsid proteins of rotavirus, HIV, gp120 of HIV, RSV, surface proteins of RSV, antigens from microorganisms which cause venereal disease and ovalbumin peptides.
  • antigens or antigenic portions or derivatives thereof may also be used in accordance with the invention. These include but are not limited to Listeria monocytogenes, Mycobacterium tuberculosis, BCG, Mycobacterium avium, M.
  • avium hsp65 influenza nucleoprotein
  • Respiratory Syncyticial virus (RSV) F or G proteins HIV
  • Candida especially Candida albicans and Chlamydia trachomatis or outer membrane proteins thereof
  • Neisseria meningitidis class 1 outer protein Herpes simplex virus type I glycoprotein G or gp D or CP27
  • Human Papilloma Virus antigen E7 Porphyromonas gingivalis Cpx
  • Murray valley encephalitis virus E glycoprotein antigenic portions thereof and derivatives thereof.
  • Antigens selected from the group consisting of those pathogens which cause the common cold, those which cause chlamydia, from Streptococcus, from Staphylococcus may also be used in accordance with the invention.
  • the antigen is selected from the group consisting of Listeria monocytogenes, Mycobacterium tuberculosis, Chlamydia trachomatis, Chlamydia pneumoniae, outer membrane proteins thereof, antigenic portions thereof, derivatives thereof, and synthetic peptides based on epitopes from these organisms.
  • the antigen is selected from the group comprising the 19 kDa lipoprotein of M. tuberculosis , hsp-65 of M. avium, VP5 of rotavirus, E7 of HPV and Cpx of P. gingivalis.
  • a selected antigen may form part of a fusion protein in order to facilitate expression and purification on production of the fusion protein in recombinant host cells.
  • the non-antigen portion of the fusion protein would generally represent the N-terminal region of the fusion polypeptide with the carboxy terminal sequences comprising antigen sequences.
  • Fusion proteins may be selected from glutathione-S-transferase, ⁇ -galactosidase, or any other protein or part thereof, particularly those which enable affinity purification utilising the binding or other affinity characteristics of the protein to purify the resultant fusion protein.
  • the protein may also be fused to the C-terminal or N-terminal of the carrier protein. The nature of the fusion protein will depend upon the vector system in which fusion proteins are produced.
  • a bacterial expression vector is pGEX which on subcloning of a gene of interest into this vector produces a fusion protein consisting of glutathione-S-transferase with the protein of interest.
  • pGEX which on subcloning of a gene of interest into this vector produces a fusion protein consisting of glutathione-S-transferase with the protein of interest.
  • Examples of other vector systems which give rise to fusion proteins with a protein of interest are described in Sambrook et al, “Molecular Cloning: A Laboratory Manual”, Cold Spring Harbor Press, Cold Spring Harbor, USA, 1989, incorporated herein in its entirety by this reference.
  • Synthetic peptides or epitopes may be produced in accordance with standard methods.
  • the carbohydrate polymer comprising mannose is most preferably a carbohydrate polymer containing mannan.
  • the antigen is conjugated to oxidised mannan in a similar manner to that described in WO 95/18145.
  • the mannan is preferably isolated from cell wall of yeast, and may be oxidised using reagents such as sodium periodate to produce a polyaldehyde which is then directly reacted with a selected antigen.
  • Reduced mannan may also be used, and a composition containing this may be prepared by adding sodium borohydride to an oxidised mannan-antigen conjugate.
  • polysaccharide chains may be first activated with cyanogen bromide and the activated polysaccharide then reacted with a diamine, followed by conjugation to the antigen to form a conjugate which may optionally then be oxidized.
  • the carbohydrate and polypeptide may be derivatized with bifunctional agents in order to cross-link the carbohydrate and polypeptide.
  • crosslinking agents include 1,1-bis (diazoacetyl)-2-phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4-azidosalicyclic acid, homobifunctional imidoesters including disuccinimidyl esters such as 3,3′-dithiobis(succinimidyl-propionate), and bi functional maleimides such as bis-N-maleimido-1,8-octane.
  • Derivatizing agents such as methyl-3-[(p-azido-phenyl)dithio] propioimidate yield photactivitable intermediates which are capable of forming cross-links in the presence of light.
  • Oxidised carbohydrates may be reacted with hydrazine derivatives of antigens to give a conjugate.
  • carbohydrates may be reacted with reagents such as carbonyl diimidazole, which after oxidation gives the desired conjugate.
  • the coupling of antigens to carbohydrate involves converting any or all of the functional groups on the carbohydrate to reactive groups and thereafter reacting the reactive groups on the carbohydrate with reactive groups on the polypeptide.
  • Carbohydrate polymers are replete with hydroxide groups, and in some instances, carboxyl groups (such as in iurodinate), ester groups (such as methylgalacturonate) and the like. These groups may be activated according to standard chemical procedures. For example, hydroxyl groups may be reacted with hydrogen halides, such as hydrogen iodide, hydrogen bromide and hydrogen chloride to give the reactive halogenated polysaccharide.
  • Hydroxy groups may be activated with phosphorous trihalides, active metals (such as sodium ethoxide, aluminium isopropoxide and potassium tert-butoxide), or esterified (with groups such as tosyl chloride or acetic acid) to form reactive groups which can be then be reacted with reactive groups on the polypeptide to form one or more bonds.
  • active metals such as sodium ethoxide, aluminium isopropoxide and potassium tert-butoxide
  • esterified with groups such as tosyl chloride or acetic acid
  • Other functional groups on carbohydrates apart from hydroxyl groups may be activated to give reactive groups according to well known procedures in the art.
  • the mannan not only acts as an adjuvant, but is also a potent mucosal adjuvant by virtue of increased or efficient uptake via the mucosa.
  • the mucosa is as described above, and includes the mucosal surfaces of the respiratory tract such as the nasal region and the lungs, the GI tract such as the buccal or oral and oro-pharyngeal cavities, throat, tonsils and gut, the rectal tissues, and the male and female urinary and reproductive tracts including the cervico-vaginal tissues.
  • the invention provides a composition adapted to be administered at a mucosal site thereby to generate an immune response, the composition comprising an antigen and a carbohydrate polymer comprising mannose.
  • the antigen, carbohydrate polymer and immune response are as described above.
  • These antigens and carbohydrates are also preferably treated in the manner described above to form the composition.
  • the composition is preferably formulated for administration at a mucosal site by inhalation of the composition which is sprayed into the nasal region.
  • it may also be administered by absorption of droplets or fluid placed on or applied to, one or more mucosal sites such as regions of the mouth, tongue, throat, the gut including the stomach, the nasal and respiratory passages including the lungs, the reproductive tract including the vagina and cervix, and the rectum.
  • Application of the composition may also be by rubbing or massaging it onto one or more accessible mucosal areas.
  • the composition may also be placed in a slow- or time-release device such as a capsule or a suppository (e.g.
  • compositions may also be formulated in accordance with the relevant methods set out in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., USA, incorporated herein by this reference.
  • the composition is most preferably used to vaccinate a subject in need thereof via a mucosal site.
  • the immune response is preferably a response as described above.
  • the invention relates to a vaccine to be administered at a mucosal site, comprising a composition as described above.
  • the invention relates to a method of vaccinating a subject, comprising the step of administering at a mucosal site a vaccine comprising a composition as described above.
  • composition or vaccine in accordance with the invention is preferably administered via a mucosal site to human or animal patients to protect against various disease states including diseases or infection of the eyes such as trachoma or conjunctivitis, listeriosis, tuberculosis, influenza, colds, respiratory diseases, sexually transmitted diseases or infections by viruses, bacteria, fungi, protozoa, or other microorganisms or pathogens.
  • diseases or infection of the eyes such as trachoma or conjunctivitis, listeriosis, tuberculosis, influenza, colds, respiratory diseases, sexually transmitted diseases or infections by viruses, bacteria, fungi, protozoa, or other microorganisms or pathogens.
  • Administration or vaccination may be as described above, or may be a single or multiple event, or may be part of a prime-boost protocol, a combination of these, or each of these with other, conventional methods of vaccination.
  • the prime-boost protocol may, for example, comprise priming by intramuscular administration, and boosting by intranasal administration.
  • the prime-boost may, in addition to the composition of the invention, include DNA, viruses (eg vaccinia) or other immunogenic peptides or molecules, including the antigens described above.
  • One or both of the priming and boosting composition may include the composition of the invention, namely, an antigen and a carbohydrate polymer comprising mannose.
  • one of the events may include the composition in accordance with the invention, while the other may omit the carbohydrate polymer containing mannose.
  • Other methods that may be used include but are not limited to injection via the rectum, the oral cavity, or rubbing of the vaccine onto a mucosal surface or site.
  • the amount and frequency may be determined by an attending physician or veterinarian.
  • 1 ⁇ g to 10,000 ⁇ g/kg may be administered to a subject, preferably 5 ⁇ g to 5000 ⁇ g/kg, more preferably 8-1000 ⁇ g/kg and most preferably 400-600 ⁇ g/kg. Even more preferably a dose of 100-200 ⁇ g/kg is contemplated particularly for humans.
  • the compounds or vaccines of the invention may also be administered to subjects in conjunction with other immune regulators that lend themselves to efficacious administration via the mucosa.
  • composition or vaccine of the invention may also be included therein, or be co-administered therewith.
  • lipofectamine may be co-administered with the composition or vaccine of the invention.
  • composition or vaccine may be formulated in accordance with the manner in which it is to be administered.
  • it may be formulated for inclusion in a spray-container, aerosol can or nebuliser for administration by inhalation.
  • it may be formulated as a solution or liquid to be added dropwise or as droplets to a mucosal site, e.g. the oral cavity or throat for absorption.
  • Nose-drop formulations, liposomal formulations, slow-release formulations, capsules or devices, or formulations comprising DNA are also contemplated.
  • the invention may be used to protect or treat animal and/or human subjects.
  • the invention provides a method of vaccinating a subject against Chlamydia comprising the step of administering a composition as described above.
  • the antigen is outer membrane protein of C. trachomatis and the method is used in the treatment or prophylaxis of trachoma.
  • the invention provides a method of immunising a subject with BCG, comprising the step of administering a composition as described above, wherein the antigen is BCG, or purified or recombinant preparations thereof.
  • the subject is an animal such as a possum.
  • the invention provides a method of sterilising a non-human subject, comprising the step of administering a composition comprising:
  • a carbohydrate polymer comprising mannose to a mucosal site.
  • the antigen is derived from sperm and administration of the composition results in production of antibody against sperm thereby to prevent fertilisation of an egg.
  • the antibody is preferably IgA.
  • the antigen is derived from the egg and induces production of antibodies against the zona pellucida preventing maturation of an embryo.
  • the antibody is preferably IgA.
  • Such methods may be used in controlling fertility of subjects such as animals that kill native wild life, e.g. foxes, or animals that destroy natural vegetation, or that compete with livestock for food, e.g. rabbits.
  • populations of animals that are a reservoir for undesirable diseases, e.g. possums, which carry or act as a reservoir for tuberculosis may also be controlled using the invention.
  • the present invention is advantageous in that it stimulates an effective immune response in a non-invasive manner.
  • animals such as native species in danger of extinction due to diseases may be vaccinated by administering nasal droplets of an immunogenic composition in accordance with the invention.
  • Non-invasive administration could also be achieved by use of aerosols or nebulisers, and are also likely to increase compliance in humans, particularly children.
  • the invention also extends to use of the composition described above in the manufacture of a medicament for modulating an immune response in a subject and use of the composition in the manufacture of a vaccine suitable for administration to a mucosal site.
  • FIG. 1 shows the results of intranasal versus intraperitoneal immunisation: (CBA ⁇ BALB/c) F1 mice were immunised on days 0, 10, 17 with 12 ⁇ g of M-LLOFP intranasally or intraperitoneally, and serum was obtained from individual mice on day 24. Antigen specific IgA (FIG. 1a) and IgG1 (FIG. 1 b ) were detected by ELISA and optical density was measured at 450 nm. Closed squares ( ⁇ ) indicate intranasally immunised mice, open circles ( ⁇ ) intraperitoneally immunised mice and open triangles ( ⁇ ) the unimmunised controls. Results for individual mice are shown.
  • FIG. 2 shows subclasses of antibody in the serum of M-LLO.FP immunised mice.
  • CBA ⁇ BALB/c mice were immunised intranasally with 12 ⁇ g of M-LLO.FP or unconjugated LLO.FP on day 0, 10 and 17 and serum from 5 individual mice obtained on day 24.
  • Antigen specific IgA (a), IgG1 (b) and IgG2a (c) was detected by ELISA. Individual titres are shown as dot plots, with corresponding symbols for different antibody classes from the same mouse. In addition, titres were converted to geometric means (heavy bar) and standard deviations (vertical bar) for display. Differences between groups receiving M-LLO.FP or unconjugated LLO.FP were significant for all classes of antibody.
  • FIG. 3 shows the levels of antigen specific IgA in the serum and distant mucosal sites over a 41 day period: (CBA ⁇ BALB/c) F1 mice were immunised on day 0,10 and 17 with 12 ⁇ g of M-LLOFP, with 1pg of CT mixed with 12 ⁇ g of LLOFP or with 12 ⁇ g of LLOFP alone. Serum (a), vaginal washings (b), and mouth washings (c) were collected from 5 individual mice on days 7, 20, 27 and 41. Antigen specific IgA titres were determined by ELISA and the progress in individual mice shown.
  • FIG. 4 shows the antigen specific IgA at distant mucosal sites over a 112 day period: (CBA ⁇ BALB/c) F1 mice were immunised on day 0, 28 and 56 with 12 ⁇ g of M-LLOFP, with 1 ⁇ g of CT mixed with 12 ⁇ g of LLOFP or with 12 ⁇ g of LLOFP alone. Serum (a) and, vaginal washings (b) were collected from 4 individual mice on day 31, 74,and 112. Antigen specific IgA titres were determined by ELISA and the progress in individual mice shown.
  • FIG. 5 shows the antigen specific IgA in the tears and in the lung washings.
  • CBA ⁇ BALB/c F1 mice were immunised on day 0, 10 and 17 with 121 g of M-LLO.FP or with 12 ⁇ g of LLO.FP alone. Mice were anaesthetised on day 24 of the experiment and tears (111) collected from 5 mice per group (a). Mice were euthanased on day 35 of the experiment and lung washings (0.5 ml) collected (b).
  • Antigen specific IgA titres were determined by ELISA and individual titres are shown as dot plots. In addition, titres were converted to geometric means (heavy bar) and standard deviations (vertical bar) for display. Differences between groups receiving M-LLO.FP or unconjugated LLO.FP were significant (p ⁇ 0.01) in tear samples and (p ⁇ 0.05) for lung washings.
  • FIG. 6 shows the effects of the M-LLOFP conjugate when in the oxidised form: (CBA ⁇ BALB/c) mice were immunised intranasally with 12 ⁇ g of M-LLOFP in the oxidised or reduced form on day 0, 10 and 17. Serum from 5 individual mice was collected on day 24 and antigen specific IgA (a), IgG1 (b) and IgG2a (c) was detected by ELISA. Individual titres are shown as dot plots, with corresponding symbols for different antibody classes from the same mouse. In addition, titres were converted to log 10 and geometric means (heavy bar) and standard deviations (vertical bar) were derived and converted back to arithmetic numbers for display. Differences between groups receiving oxidised or reduced M-LLO.FP were significant (p ⁇ 0.02) for all classes of antibody (Student's t test).
  • FIG. 7( a ) shows the effects of oxidised mannan as adjuvant for mycobacterial 19 kDa FP: Groups of 4 C57/B. 10 mice were immunised intranasally with 12 ⁇ g of M-19FP or 12 ⁇ g of 19 kDaFP on day 0,10 and 17. Serum was collected on day 24 and antigen specific IgA determined by ELISA. Individual titres are shown as dot plots. In addition, titres were converted to log 10 and geometric means (heavy bar) and standard deviations (vertical bar) were derived and converted back to arithmetic numbers for display. Differences between groups receiving M-19FP and 19FP alone were significant by Student's t test (p ⁇ 0.02).
  • FIG. 7( b ) shows ELISA readings for IgA titres for mice immunised with hsp65—mannan conjugate.
  • FIG. 8 Comparison of intranasal and intraperitoneal immunisation with M-LLO.FP.
  • CBA ⁇ BALB/c F1 mice (4 per group) were immunised with 12 ⁇ g of M-LLO.FP i.n. or i.p. or unconjugated control on day 0, 10 and 17 and mice sacrificed on day 24.
  • IL-2 production Spleen cells from all groups were cultured in the presence of antigen (45 ⁇ g/ml LLO (215-226) ( ⁇ ) or LLO.FP ( )) or without stimulus ( ⁇ ). Data represent the means and standard deviations of triplicate cultures. * p ⁇ 0.01 ** p ⁇ 0.002 compared M-LLO.FP i.p.
  • IFN- ⁇ producing cells Spleen cells from all groups were cultured in the presence of antigen (45 ⁇ g/ml of LLO.FP ( ⁇ )) or without stimulus ( ⁇ ). Data represent the means and standard deviations of triplicate cultures. * p ⁇ 0.05 compared with MLLO.FP i.p.
  • FIG. 9 IFN- ⁇ and IL4 production following i.n. immunisation with oxidised or reduced M-LLO.FP. (CBA ⁇ BALB/c) Fl mice (5 per group) were immunised i.n. with 12 ⁇ g of M-LLO.FP (oxidised) or with M-LLO.FP (reduced) on day 0, 10 and 17 or infected i.v. with 1 ⁇ 10 3 Listeria on day 17. Mice were sacrificed on day 24. Spleen cells from all groups were cultured in the presence of antigen (45 ⁇ g/ml of LLO.FP ( ⁇ )) or without stimulus ( ⁇ ) and the frequency of (a) IFN- ⁇ and (b) IL-4 producing cells determined. Data represent the means and standard deviations of triplicate cultures. *p ⁇ 0.02, **p ⁇ 0.05 compared with the reduced group for IFN- ⁇ and ILA respectively. Note different scales.
  • FIG. 10 IFN- ⁇ production following i.n. immunisation with M-19.FP.
  • C57BU10 mice (4 per group) were immunised i.n. with 12 ⁇ g of M-19.FP or unconjugated control on day 0, 10 and 17 or infected i.n. with M. evium 6 weeks prior to immunisation.
  • Mice were sacrificed on day 24 and spleen cells from all groups cultured in the presence of antigen ( ⁇ ) (19 kDa.FP 23 ⁇ g/ml), 10 7 live M. avium ( ) or without stimulus ( ⁇ ).
  • Data represent the means and standard deviations of triplicate wells. * p ⁇ 0.05 compared with the 19.kDa.FP group.
  • FIG. 11 Antibody titres in serum following immunisation with VP5 246-274 FP with and without mannan adjuvant: Mice were immunised intranasally with 6 ⁇ g VP5 246-274 -mannan of VP5 246-274 alone on days 0,10 and 17, and bled from the tail vein on days 24 and 35. Titres are expressed as log 10 geometric means plus or minus standard deviation for groups of 5 mice. Differences p ⁇ 0.05 are considered significant. **p ⁇ 0.02, *p ⁇ 0.05.
  • FIG. 12 IgA titres on mucosal surfaces following immunisation with VP5 246-274 FP with and without mannan adjuvant: Mice were immunised intranasally with 6 ⁇ g VP5 246-274 -mannan of VP5 246-274 alone on days 0,10 and 17. Titres are expressed as log 10 geometric means plus or minus standard deviation for groups of 5 mice.
  • FIG. 13 Protection against Porphyromonas gingivalis lesions: Groups of 5 C57B1/10 mice were immunised with Cpx-mannan given intranasally (FIG. 13( a )) or Cpx in incomplete Freund's adjuvant (IFA) subcutaneously (FIG. 13( b )). Control mice received the respective adjuvants alone. Mice given Cpx-mannan intranasally showed no lesions—complete protection (p ⁇ 0.001), while those given Cpx in IFA showed incomplete protection (p ⁇ 0.05).
  • FIG. 14 Serum antibody titres: Groups of 5 C57B1/10 mice were immunised with Cpx-mannan given intranasally or Cpx in incomplete Freund's adjuvants (IFA) subcutaneously. Control mice received the respective adjuvants alone. The mice were bled from the retro-orbital plexus after 2 or three doses and serum antibody assayed by ELISA. Titres on pooled sera are shown.
  • IFA incomplete Freund's adjuvants
  • FIG. 15 Antibody titres in serum following immunisation with E7.FP coupled to mannan under oxidising or reducing conditions or without mannan adjuvant (E7 alone): Mice were immunised intranasally with 12 ⁇ g antigen (with or without mannan) on days 0, 10 and 17, and bled from the tail vein on days 24. Titres are expressed as log 10 geometric means plus or minus standard deviation for groups of 5 mice. Titres of all classes of antibody in groups immunised E7.FP plus either oxidised or reduced mannan were significantly different from the group receiving E7.FP, alone p ⁇ 0.01.
  • mice Female 6-8 week old (CBA ⁇ BALB/c)F1 and C57B1/10 mice were bred and maintained under conventional but infection-free conditions in the animal house of the Department of Microbiology and Immunology, University of Melbourne.
  • a p. bluescript plasmid containing the gene for Listeriolysin O (LLO) from Listeria monocytogenes (without its leader sequence) was obtained from Richard S scholarnell (University of Melbourne, Australia) and subcloned into the Escherichia coli expression vector pGEX-2T [16] in the correct reading frame and orientation.
  • the expression of an 84 kDa LLO glutathione-s-transferase (GST) fusion protein (LLO.FP) was induced with 0.1 mM IPTG (Sigma Chemical Co., MO, USA) at 37° C. for 5 hours.
  • Bacteria were collected by centrifugation (1,500 ⁇ g) for 5 minutes, washed and lysed by sonication. The pellet was collected after centrifugation and solubilised in 8M urea (Eastman Kodak Co., Rochester, N.Y., USA) overnight at 4° C. After further centrifugation (26,000 ⁇ g) for 15 minutes the supernatant was dialysed in 0.01 M Tris (Sigma Chemical Co., MO, USA) pH 8.0/1 M urea and the BIOCAD perfusion chromatography system (Perceptive Biosystems, Framingham, Mass., USA) used to purify the protein by anion exchange.
  • the 19 kDa lipoprotein from Mycobacterium tuberculosis was produced using a recombinant 19 kDa-plasmid construct was obtained from Professor Douglas Young (Imperial College School of Medicine, London, U.K) and the expression of a 19 kDa HIS tag fusion protein (19FP) induced with 0.1 mM IPTG at 37° C. for 5 hours. Bacteria were collected by centrifugation (1,500 ⁇ g), washed and lysed by sonication.
  • the supernatant was collected after further centrifugation (26,000 ⁇ g) for 15 minutes and dialysed in 0.01 M Tris pH 8.0/300 mM NaCl/20 mM imidazole and purified under native conditions on a Nickel chelate column (Qiagen, Hilden, Germany) using the BIOCAD perfusion chromatography system.
  • the fragment containing the coding sequence the hsp65 was moved from the M13tg130B derivative into p2GEX-2T, so that the hsp65 reading frame was fused in-frame to the 3′ end of one of the two copies of the glutathione-S-transferase (GST) of Schistosoma japonicum.
  • GST glutathione-S-transferase
  • E. coli strain BL21 transformed with the recombinant plasmid p2GEXhsp65. Expression of the GST fusion was induced by the addition of 0.1 mM Isopropylthiogalactoside (IPTG) (Sigma). Bacteria were harvested, washed and lysed.
  • the cell lysate was cleared by centrifugation and the separated cell pellet and supernatant analysed on a 12% SDS-PAGE.
  • the supernatant which contained the majority of the fusion protein (GST-Hsp65) was filtered and the GST fusion protein purified by binding to glutathione agarose beads according to manufacturers' instructions (Sigma).
  • GST-Hsp65 the majority of the fusion protein
  • GST fusion protein purified by binding to glutathione agarose beads according to manufacturers' instructions (Sigma).
  • the beads were incubated overnight at room temperature with thrombin (Pharmacia) (10 units per milligram of protein) in 10 ml of PBS, then pelleted, the supernatant collected and the protein content estimated spectrophotometrically.
  • the E7 antigen of Human Papilloma Virus was produced by recombinant means.
  • the DNA sequence for the E7 antigen from HPV was cloned into the pGEX-2T vectors. Protein expression was induced by IPTG, and purified using glutathione-sepharose column chromatography. Elution of the bound protein was with a solution of 5 mM glutathione in Tris 0.05M, pH 8.0.
  • Mannan (Sigma Chemical Co., St Louis, Mo., USA) was coupled to the antigens under oxidising conditions. Mannan at 14 mg/ml in 0.1 M phosphate buffer pH 6.0 was oxidised with 0.01 M sodium periodate for 60 min at 4° C. Ten microlitres of ethandiol (Sigma Chemical Co., St Louis, Mo., USA) was added to quench the reaction and the mixture incubated for 30 min at 4° C. This mixture was then passed through a PD-10 column (Pharmacia Biotech, Uppsala, Sweden)-equilibrated with bicarbonate buffer pH 9.0.
  • the oxidised mannan eluted in the 2 ml void volume, was mixed with 700 ⁇ g of the antigen, incubated overnight at room temperature and used without any further purification. Conjugation was confirmed when the conjugates were separated using 12.5% sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) and a heterogeneous smear (compared with the single band of uncoupled protein) was observed using Comassie Blue stain. For comparison in some experiments, the oxidised conjugates were reduced with sodium borohydride (NaBH 4 ) (Aldrich, Castle Hill, NSW, Australia) 1 mg/ml overnight at room temperature and used without further purification.
  • NaBH 4 sodium borohydride
  • mice were lightly anaesthetised with penthrane and 50pl of mannan-antigen conjugate (12 ⁇ g antigen/mouse in bicarbonate buffer pH 9.0, unless otherwise specified), placed onto the nares to be inhaled by the mouse. The same amount of non-conjugated antigen was similarly applied. Unless stated otherwise, this procedure was performed on days 0, 10 and 17 of the experiments. In some experiments mice were given 12 ⁇ g of mannan-LLO.FP (M-LLO.FP) conjugate intraperitoneally in 0.2 ml bicarbonate buffer pH 9.0 on days 0, 10 and 17.
  • M-LLO.FP mannan-LLO.FP
  • Cholera toxin (Sigma Chemical Co., St Louis, Mo., USA) was also used for comparative purposes as an adjuvant to act as a positive control in some experiments.
  • One microgram of CT was mixed with 12 ⁇ g of LLO.FP and administered intranasally (i.n) in a 50 ⁇ l volume on days 0, 10 and 17.
  • mice Serum samples were collected after mice were bled by cardiac puncture following euthanasia at the end of each experiment. For time-course experiments mice were placed on a heat box, a small incision made in a lateral vein and 200 ⁇ l of blood collected with a micropippeter. The serum was subsequently separated by centrifugation. Mouth and vaginal washings were collected from anaesthetised mice, by washing with 50 or 100 ⁇ l of phosphate buffered saline (PBS) respectively, using a micropippetor. For the collection of faecal samples, mice were placed in cages without sawdust and 2-5 fresh stools were collected per mouse.
  • PBS phosphate buffered saline
  • PBS/SBTI PBS containing 0.1 mg/ml soybean trypsin inhibitor (Sigma Chemical Co., St Louis, Mo., USA)
  • PMSF Phenyl sulphonyl fluoride
  • antigen-specific IgA was detected by the addition of an anti-mouse IgA affinity purified horse radish peroxidase (HRP) conjugate (Southern Biotechnology Associates Inc. Birmingham, USA) diluted ⁇ fraction (1/1000) ⁇ in 0.1% bovine serum albumin (BSA) (CSL, Melbourne, Australia) for 1 hour at room temperature.
  • HRP horse radish peroxidase
  • BSA bovine serum albumin
  • Antigen specific IgG1 or IgG2a was detected with the addition of a biotinylated anti-mouse IgG1 or IgG2a conjugate (Caltag Laboratories, Burlinggame, Ca, USA) diluted ⁇ fraction (1/1000) ⁇ in 0.1% BSA.
  • the plates were washed twice more and a streptavidin peroxidase conjugate (Boehringer Mannheim, Mannheim, Germany) added at a ⁇ fraction (1/1000) ⁇ dilution in 0.1% BSA. Following a further two washes, the antibody titres of all the subclasses tested were determined when the substrates containing either 0.4 g/l 3, 3′, 5, 5′-tetramethylbenzidine (TMB) (Kirkgaard and Perry Laboratories, Gaithersburg, Md., USA) and 0.02% H 2 O 2 or 2,2′-azino-bis(3ethylbenthiazoline-6-sulphonic acid) (ABTS) (Sigma Chemical Co., St Louis, Mo., USA) and 0.03% H 2 O 2 were added (50 ⁇ l/well).
  • TMB trimethylbenzidine
  • ABTS 2,2′-azino-bis(3ethylbenthiazoline-6-sulphonic acid
  • OD at 450 nm (TMB) or 405 nm (ABTS) was read in an ELISA reader (Labsystems, Helsinki, Finland).
  • Antibody titres were presented as the highest dilution which yielded an optical density at 450 nm or 405 nm>0.1 OD units higher than normal serum at ⁇ fraction (1/100) ⁇ dilution. For calculation of means, the titre was converted to log 10 and a geometric mean was derived.
  • Spleen cell proliferation and cytokine production Mice were euthanased with CO 2 and spleen cells prepared by passage through an 80 gauge wire mesh sieve. Red blood cells were lysed using Tris-NH 4 Cl buffer [20]. After thorough washing, suspensions of spleen cells were cultured in flat-bottomed 96 well plates at a concentration of 2 ⁇ 10 6 /ml in 200 ⁇ l volumes with and without the MHC class 11 restricted epitope of LLO (215-226) (45 ⁇ g/ml) for 3 days at 37° C., 5% CO 2 .
  • the cells were pulsed with 0.25 ⁇ Ci [ 3 H]-TdR (Amersham, Buckinghamshire, U.K) before being harvested onto glass fibre filters (Packard, Meridan, Conn., USA) using the Micro 96 harvester (Skatron Instruments, Wokingham, UK) and ⁇ emissions counted using a Packard Matrix 9600 (Packard).
  • IL-2 bioassay supernatants from similarly-cultured cells were collected after 24 hour culture and IL-2 was assayed by its ability to cause the proliferation of CTLL cells [21].
  • Elispot Spleen cells were assayed for the frequency of IFN- ⁇ and IL-4 producing cells. Maxisorp 96 well plates (Nunc) were coated overnight at 4° C. with 50 ⁇ l of 10 ⁇ g/ml anti-IFN- ⁇ monoclonal antibody HB170 [22] or 100 ⁇ l of anti-1 L-4 monoclonal antibody 11B11 [23] in carbonate buffer pH 9.6. The plates were washed with PBS and blocked with culture medium at 37° C. for 1 hr.
  • Cells (2 ⁇ 10 5 , 1 ⁇ 10 5 , 5 ⁇ 10 4 , 2.5 ⁇ 10 4 per well) were added to the wells in conjunction with the appropriate antigen (45 ⁇ g/ml) and incubated for 72 hrs at 37° C. in 5% CO 2 .
  • the cells were removed and cytokines detected with the addition of a biotinylated secondary rat anti-mouse anti-IFN- ⁇ antibody XMG 1.2 [24] or rat anti-mouse anti-IL4 antibody BVD6 [25] at a ⁇ fraction (1/1000) ⁇ dilution.
  • spots were developed using 5-bromo-4-chloro-3-indyl phosphate/nitrobluetetrazolium (BCIP/NBT) (Sigma) tablets. The spots were counted using a dissecting microscope and the frequency determined by averaging the number of spots for triplicate wells.
  • BCIP/NBT 5-bromo-4-chloro-3-indyl phosphate/nitrobluetetrazolium
  • a Listeriolysin-mannan (M-LLO) conjugate was prepared as described in Example 1.
  • IgA antibody was not detectable after the standard i.p. immunisation. It should be noted that i.p. immunisation could lead to antibody production, as IgG1 titres of ⁇ fraction (1/640) ⁇ were detected (FIG. 1 b ). However, even with this isotype i.n immunisation was superior, inducing titres in excess of ⁇ fraction (1/1280) ⁇ .
  • Oxidised mannan was a consistently better mucosal adjuvant than CT when administered with LLO.FP.
  • Immunisation with M-LLO.FP induced a peak geometric mean IgA titre of ⁇ fraction (1/667) ⁇ in serum with some titres in excess of ⁇ fraction (1/1280) ⁇ . This compared with ⁇ fraction (1/165) ⁇ for CT+LLO.FP (FIG. 3 a ).
  • LLO.FP alone induced a mean titre of only ⁇ fraction (1/69) ⁇ .
  • IgA titres were examined over a considerable timecourse in these experiments. In general, they did not reach substantial levels until after three injections, whether of M-LLO.FP or CT+LLO.FP. IgA persisted at peak titre in the serum for more than three weeks after the last immunisation, particularly of M-LLO.FP (FIG. 3 a , 4 a ). IgA production in the vagina (FIG. 3 b , 4 b ) also required three immunisations and these titres were more variable than in-serum, and generally less sustained.
  • mice were immunised i.n. with M-LLO.FP as before and tears and lung washings collected on days 24 and 35 respectively.
  • Mice immunised with M-LLO.FP conjugate produced significantly higher titres of IgA (p ⁇ 0.01) in tears when compared with the unconjugated controls (geometric means titres: M-LLO.FP ⁇ fraction (1/121) ⁇ compared with ⁇ fraction (1/25) ⁇ for LLO.FP alone) (FIG. 5 a ).
  • the same trend was observed in the lungs at day 35 (FIG. 5 b ).
  • Significantly higher titres of IgA were detected in mice immunised with M-LLO.FP (1/975) compared with LLO.FP alone (1/38) (p ⁇ 0.05).
  • mice were immunised with the M-LLO.FP conjugate or with a mannan+LLO.FP mixture (mannan and 12 ⁇ g of LLO.FP mixed together).
  • CBA ⁇ BALB/c)F1 mice were immunised i.n on days 0, 10 and 17.
  • Serum and vaginal washings were taken from individual mice on day 24 of the experiment.
  • IgA titres were subsequently determined by ELISA (Table 1). The results showed that a conjugate provides a better adjuvant effect.
  • the geometric mean antibody titre in the serum for conjugated LLO.FP was > ⁇ fraction (1/1000) ⁇ . This was significantly higher than the mannan+LLO.FP mixture which produced an average titre of ⁇ fraction (1/34) ⁇ . Notwithstanding this some antibody is obtained from the mannan antigen admixture. A similar pattern was seen in the antibody response at a distant mucosal site, namely the vagina, with titres of ⁇ fraction (1/195) ⁇ for the conjugate and ⁇ fraction (1/45) ⁇ for the mixture. All negative controls produced undetectable levels of IgA with small titres of antibody observed in vaginal washings for the LLO.FP alone group ( ⁇ fraction (1/14) ⁇ ).
  • mice immunised with M-LLO.FP had higher titres than the corresponding four mice given the reduced form of M-LLO.FP, with the exception of one high responder mouse in the group given reduced mannan.
  • mannan was conjugated to the 19 kDa lipoprotein of Mycobacterium tuberculosis and to Mycobacterium avium antigen hsp65 as set forth in Example 1.
  • C57B1/10 mice were immunised i.n. with 12 ⁇ g of M-19FP conjugate on days 0, 10 and 17.
  • the mice were euthanased and bled by cardiac puncture.
  • Mice were similarly immunised with mannan hsp65. Serum was separated and IgA titres determined by ELISA (FIG. 7 a ).
  • mice immunised with M-19FP produced significantly higher titres of IgA (geometric mean ⁇ fraction (1/1530) ⁇ ) when compared with mice given 12 ⁇ g of 19 kDaFP ( rates of IgA ⁇ fraction (1/100) ⁇ ).
  • the results for mice immunised with mannan-hsp65 are shown in FIG. 7( b ). IgA titres for the immunised mice were well above that of the normal mice.
  • mannan as a novel mucosal adjuvant which, when administered intranasally, induced high mucosal and serum titres of IgA, IgG1 and IgG2a specific for recombinant protein antigens with which it was administered.
  • Oxidised mannan was previously used conjugated to the breast cancer antigen MUC1 and injected i.p. into mice where it induced CTL and a Th1 cytokines, protecting against tumours expressing MUC1 [12, 13]. In that context it induced a poor antibody response, dominated by IgG2a. It was the change to intranasal administration in the current experiments which resulted production of IgA and other classes of antibody in serum and mucosal secretions.
  • IgA responses on mucosal surfaces is a highly desirable outcome in immunisation against a wide spectrum of diseases.
  • mucosal antibodies and particularly IgA have been shown to protect against influenza in the lung [29], against Helicobacter pylori in the stomach [30], against Haemophilis influenzae in the ear [31] and against Chlamidia trachomatis in the genital tract [32].
  • Induction of IgA and other mucosal antibodies following intramuscular or subcutaneous immunisation with conventional vaccines is poor [33, 33a]. Therefore oxidised mannan may be a valuable adjuvant if coupled to protective antigens from a wide variety of infectious agents.
  • Mannan has the further advantage over CT in being non-toxic and already approved for human use and is currently involved in human trails for cancer therapy [14].
  • CTB B subunit of CT
  • antigen resulted in higher titres of IgA in the serum and mucosa when given to mice intranasally rather than intraperitoneally [33]. It is widely believed that this influence of the route of exposure is a function of the mucosal antigen presenting cells.
  • mannan act as an adjuvant? It has been shown that antigens bearing mannose residues are bound to the mannose receptors on antigen presenting cells (APC), facilitating uptake into the MHC Class II pathway for efficient antigen presentation [35, 36, 37]. Oxidised mannan-MUCI induced both Class I restricted CTL and a Th1 response [12]. It has been suggested that oxidised mannan-MUC1, by virtue of the aldehyde residues created by the oxidation process, escapes the phagocytic pathway and is transported into the MHC Class I pathway inducing CTL [15].
  • M cells membranous cells (specialised epithelial cells) on the mucosal surface. These cells either process and present antigen to underlying T-Cells or B-cells themselves or transport antigen to parenchymal macrophages, dendritic cells and B-cells.
  • APC antigen presenting cell
  • Immune responses generally involve antibody production, with IgA the predominant antibody isotype.
  • Antigen sensitised immune cells are then circulated to other systemic and mucosal sites for expansion of effector mechanisms [40]. The fact that there is a preferential circulation of T cells activated at mucosal sites to return to the same or other mucosal sites accounts for the induction of IgA at remote mucosal surfaces.
  • mice production of antigens, conjugates and immunisation were the same as Example 1.
  • Spleen cell proliferation, cytokine production and Elispot assays were as described in Example 1.
  • FIG. 8 b The ability of spleen cells to produce IL-2 is shown in FIG. 8 b .
  • Spleen cells were cultured in the presence of LLO (215-226) or LLO.FP (45 ⁇ g/ml) and supernatants harvested at 24 hours.
  • the ability of the supernatants to maintain the IL-2 dependent CTL cell line was determined by the incorporation of [ 3 H]-TdR. Mice immunised with MLLO.FP intranasally produced counts of 1219 ⁇ 268 (LLO (215-226 )) and 4523 ⁇ 689 (LLO.FP).
  • IFN- ⁇ producing cells/10 6 spleen cells for the 12 ⁇ g dose was significantly higher than for the 6 ⁇ g (p ⁇ 0.01) and 3 ⁇ g (p ⁇ 0.01) dose of M-LLO.FP (97 ⁇ 20, 32 ⁇ 8 and 12 ⁇ 6 respectively).
  • Mice given the unconjugated control antigen all produced fewer than 50 IFN- ⁇ producing cells/10 6 spleen cells (results not shown).
  • oxidised mannan as an adjuvant which, when administered intranasally, induced proliferative responses, IL-2 production, IFN- ⁇ production and IL-4 production by cultured lymphocytes. Comparison of intranasal and intraperitoneal routes of immunisation revealed the superior efficacy for induction of these immune responses by the intranasal route. As most proteins administered to the mucosal surface without adjuvant fail to induce systemic or mucosal immune responses, oxidised mannan may be used as an adjuvant to induce both Th1 and Th2 immune responses. It offers the great advantage as an adjuvant of being non-toxic and already approved for human use in the context of breast-cancer therapy.
  • oxidised mannan administered intranasally can be an effective adjuvant for the induction of Th1 and Th2 responses as seen with the two model antigens used in this Example.
  • This ability is of particular interest with infections such as C. trachomatis or H. pylon where both cell mediated and humoral responses appear necessary for control of infection.
  • C. trachomatis cell mediated responses alone appear to contribute to immunopathology whereas antibody responses are not sufficient to control infection [42, 43].
  • Th1 and Th2 responses are required at different stages for control of infection. The early response is dominated by Th1 and the late by Th2 [44].
  • Candidate vaccines such as those contemplated by the present invention with the ability to produce both Th1 and Th2 responses might therefore prove to be effective against both pathogens.
  • mice and immunisation thereof were as described in Example 1. Samples of serum, tears and/or stools were collected as described in Example 1 at days 24 and 34 after immunisation. On day 35 the mice were sacrificed and lung washings were collected as shown in Example 1.
  • FIG. 11 shows that in mice immunised intranasally with 6ug VP5 246-274 -mannan, titres of serum IgA, IgG1 and IgG2a were significantly higher than in mice immunised intranasally with the antigen alone, at 24 and 35 days after immunisation.
  • FIG. 12 shows the presence of IgA in tears collected 24 days after immunisation with the antigen-mannan conjugate, and in the stools collected at day 34. No antibody was detected in mice receiving antigen alone. IgA in lung washings was also significantly higher than in samples from animals injected with the antigen alone.
  • the antigen used was the major virulence factor of Porphyromonas gingivalis, the extracellular complex of Arg- and Lysspecific proteases and adhesions designated the RpgA-Kgp complex (formerly the PrtR-PrtK complex and abbreviated Cpx), prepared and conjugated with mannan as described in Example 1.
  • Cpx antigen (12 ⁇ g) was administered intranasally on days 0, 10 and 17.
  • the Cpx antigen was administered subcutaneously in incomplete Freunds adjuvant as previously described [18].
  • Mice were infected with 8 ⁇ 10 9 cells of Porphyromonas gingivalis subcutaneously in the abdomen on day 30 and lesion sizes measured over 14 days. Lesion sizes were statistically analysed using the Kruskal-Wallis test and the Mann-Whitney U-Wilcoxon rank sum test with a Bonferroni correction.
  • mice Groups of 5 C57B1/10 mice were immunised with Cpx-mannan given intranasally or Cpx in incomplete Freund's adjuvant (IFA) subcutaneously. Control mice received the respective adjuvants alone. Results of protection against challenge with Porphyromonas gingivalis are shown in FIG. 13. Cpx linked to mannan afforded complete protection. IFA, the standard adjuvant used in studies of Porphyromonas gingivalis vaccination afforded only partial protection. Interestingly, the serum antibody titres, although raised in the mice immunised with Cpx-mannan, was higher in the mice immunised with Cpx in IFA. Both had significant adjuvant effects (p ⁇ 0.05). This emphasises the point that, particularly where inflammatory lesions are critical to disease, antibody titre may not be an accurate predictor of protection.
  • IFA incomplete Freund's adjuvant
  • mice were immunised with HPV-E7 conjugated to oxidised mannan, E7 conjugated to reduced mannan and with E7 alone, as per Example 1.
  • the results show no substantial difference between the oxidised and reduced forms although this may have been due to dose levels.
  • Antigens used in accordance with the invention may also be produced by conventional peptide synthesis after selecting putative immunogenic peptides.
  • the putative immunogenic peptides may be selected from published studies or may be peptides suspected of being immunogenic based on a skilled person's knowledge of the art.
  • the putative immunogenic peptides are then coupled to a carrier protein which is, in turn, conjugated to mannan.
  • plasmids containing genes encoding selected antigens are expressed in E. coli .
  • the proteins coupled to mannan are administered and the antibody response at the site most relevant to protection is examined. Examples of proteins that can be tested are as follows: PROTECTIVE SITE OF ORGANISM DISEASE ANTIGEN ANTIBODY Influenza Flu Haemagglutinin Lungs virus Helicobacter Stomach ulcers, Recombinant Stomach pylori cancer proteins, urease Rotavirus Gastroenteritis Recombinant VP4 Intestine in infants protein and its fragment VP5 and other outer capsid proteins Chlamydia Sexually transmitted Major outer Vagina, Eye trachomatis disease, pelvic membrane protein inflammatory disease, non- specific urethritis, trachoma
  • Mannan-antigen complexes are encased in various timeddelivery capsules and delivered intranasally, orally or rectally. At intervals, serum, saliva, lung or vaginal washings will be collected, and ELISA are used to measure IgG1, IgG2a, IgE and IgA.
  • mice and monkeys were administered to humans, mice and monkeys mostly IgG1 antibodies were found. There was no IgA response. Given this background it was extremely surprising that intranasal administration of mannan-antigen gave rise to secreted IgA.
  • intranasal administration of mannan and antigen may advance vaccine development for sexually transmitted diseases such as chlamydia or human immunodeficiency virus, gum disease, eye diseases, other mucosally acquired infections like influenza and rotavirus or even have veterinary application with uses in animal infections or pest control.
  • sexually transmitted diseases such as chlamydia or human immunodeficiency virus, gum disease, eye diseases, other mucosally acquired infections like influenza and rotavirus or even have veterinary application with uses in animal infections or pest control.
  • this is a novel, non-toxic adjuvant that induces systemic and mucosal antibody responses superior to the standard mucosal adjuvant CT.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Communicable Diseases (AREA)
  • Molecular Biology (AREA)
  • Pulmonology (AREA)
  • Oncology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
US10/297,256 2000-06-06 2001-06-06 Vaccine Abandoned US20040043032A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/813,982 US20110033493A1 (en) 2000-06-06 2010-06-11 Vaccine

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
AUPQ7977 2000-06-06
AUPQ7977A AUPQ797700A0 (en) 2000-06-06 2000-06-06 Vaccine
PCT/AU2001/000669 WO2001093912A1 (en) 2000-06-06 2001-06-06 Vaccine

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2001/000669 Continuation WO2001093912A1 (en) 2000-06-06 2001-06-06 Vaccine

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/813,982 Continuation US20110033493A1 (en) 2000-06-06 2010-06-11 Vaccine

Publications (1)

Publication Number Publication Date
US20040043032A1 true US20040043032A1 (en) 2004-03-04

Family

ID=3822049

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/297,256 Abandoned US20040043032A1 (en) 2000-06-06 2001-06-06 Vaccine
US12/443,048 Expired - Fee Related US8182821B2 (en) 2000-06-06 2007-10-02 Flu vaccine admixture of mannan and flu antigen
US12/813,982 Abandoned US20110033493A1 (en) 2000-06-06 2010-06-11 Vaccine

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/443,048 Expired - Fee Related US8182821B2 (en) 2000-06-06 2007-10-02 Flu vaccine admixture of mannan and flu antigen
US12/813,982 Abandoned US20110033493A1 (en) 2000-06-06 2010-06-11 Vaccine

Country Status (7)

Country Link
US (3) US20040043032A1 (de)
EP (2) EP1301208B1 (de)
AT (1) ATE456958T1 (de)
AU (4) AUPQ797700A0 (de)
DE (1) DE60141243D1 (de)
HK (1) HK1056830A1 (de)
WO (1) WO2001093912A1 (de)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050147607A1 (en) * 2003-04-11 2005-07-07 Reed Jennifer L. Methods of preventing or treating respiratory conditions
US20080031872A1 (en) * 2006-08-02 2008-02-07 Karp Nelson M HIV immunogenic composition and method of administration
US20090130131A1 (en) * 2005-08-05 2009-05-21 Hiroshi Kido Antigen-Drug Vehicle Enabling Switch From Selective Production of IgA Antibody to Production of Both of IgA and IgG Antibodies and Transnasal/Mucosal Vaccine Using the Same
US20100008951A1 (en) * 2000-06-06 2010-01-14 The Macfarlane Burnet Institute For Medical Research And Public Health Limited Flu vaccine admixture of mannan and flu antigen
US20100119609A1 (en) * 2006-10-17 2010-05-13 John Daniel Dobak Methods, compositions, and formulations for the treatment of thyroid eye disease
US20110105446A1 (en) * 2005-07-14 2011-05-05 Lithera, Inc. Sustained Release Enhanced Lipolytic Formulation for Regional Adipose Tissue Treatment
US20110130373A1 (en) * 2009-05-27 2011-06-02 Lithera, Inc. Methods for administration and formulations for the treatment of regional adipose tissue
US20110224176A1 (en) * 2010-01-15 2011-09-15 Lithera, Inc. Lyophilized Cake Formulations
US9597392B2 (en) 2010-05-10 2017-03-21 Ascend Biopharmaceuticals Pty Ltd. Use of high molecular weight mannan for inducing and/or enhancing an immune response
US9597531B2 (en) 2010-11-24 2017-03-21 Neothetics, Inc. Selective, lipophilic, and long-acting beta agonist monotherapeutic formulations and methods for the cosmetic treatment of adiposity and contour bulging

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008037033A1 (en) * 2006-09-29 2008-04-03 The Macfarlane Burnet Institute For Medical Research And Public Health Limited Flu vaccine admixture of mannan and flu antigen
JP2003286191A (ja) * 2002-03-27 2003-10-07 Univ Nihon 歯周病用粘膜免疫ワクチン
US10344261B2 (en) * 2011-11-09 2019-07-09 Ascend Biopharmaceuticals Ltd Immunomodulatory conjugates
EP4304606A1 (de) * 2021-03-12 2024-01-17 Children's Medical Center Corporation Polysaccharid-adjuvantien für virusimpfstoffe

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5597807A (en) * 1994-08-01 1997-01-28 University Of Saskatchewan Quinoa saponin compositions and methods of use
US5804200A (en) * 1991-02-12 1998-09-08 Colorado State University Research Foundation Parasitic nematode proteins and vaccines
US5989552A (en) * 1993-12-24 1999-11-23 Austin Research Institute Antigen carbohydrate compounds and their use in immunotherapy
US6548643B1 (en) * 1994-11-16 2003-04-15 Austin Research Institute Antigen carbohydrate compounds and their use in immunotherapy
US6555115B1 (en) * 1998-12-08 2003-04-29 Corixa Corporation Compounds and methods for treatment and diagnosis of chlamydial infection
US6573245B1 (en) * 1998-04-28 2003-06-03 Galenica Pharmaceuticals, Inc. Modified polysaccharide adjuvant-protein antigen conjugates, the preparation thereof and the use thereof
US7329408B2 (en) * 1999-12-01 2008-02-12 Novartis Vaccines And Diagnostics, Inc. Eliciting HCV-specific antibodies

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU620149B2 (en) 1988-08-18 1992-02-13 Vaxine Pty Ltd Gamma inulin compositions
AUPN568095A0 (en) 1995-09-27 1995-10-26 Austin Research Institute, The Anti-Galalpha(1,3)Gal antibody binding peptides
CA2275259C (en) * 1996-12-20 2008-02-12 Merck & Co., Inc. Formulations of recombinant papillomavirus vaccines
DE69840578D1 (de) * 1997-09-29 2009-04-02 Macfarlane Burnet Inst For Med Mannose-rezeptor tragende zellinie und antigen zusammensetzung
ES2245805T3 (es) * 1997-10-03 2006-01-16 Galenica Pharmaceuticals, Inc. Polisacaridos que forman iminas, preparacion de los mismos y el uso de los mismos como adyuvantes de inmunoestimulantes.
ATE481108T1 (de) 1999-02-26 2010-10-15 Novartis Vaccines & Diagnostic Verwendung von bioadhaesiven und adjuvantien für die mukosale anwendung von antigenen
JP2003533181A (ja) 2000-02-01 2003-11-11 ジ・オースティン・リサーチ・インスティテュート ムチン−1誘導抗原および免疫療法におけるその使用
AU2001263653B2 (en) * 2000-06-06 2006-06-29 Igavax Pty Ltd Vaccine
AUPQ797700A0 (en) * 2000-06-06 2000-06-29 Austin Research Institute, The Vaccine

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5804200A (en) * 1991-02-12 1998-09-08 Colorado State University Research Foundation Parasitic nematode proteins and vaccines
US5989552A (en) * 1993-12-24 1999-11-23 Austin Research Institute Antigen carbohydrate compounds and their use in immunotherapy
US6177256B1 (en) * 1993-12-24 2001-01-23 Austin Research Institute Antigen carbohydrate compounds and their use in immunotherapy
US5597807A (en) * 1994-08-01 1997-01-28 University Of Saskatchewan Quinoa saponin compositions and methods of use
US6548643B1 (en) * 1994-11-16 2003-04-15 Austin Research Institute Antigen carbohydrate compounds and their use in immunotherapy
US6573245B1 (en) * 1998-04-28 2003-06-03 Galenica Pharmaceuticals, Inc. Modified polysaccharide adjuvant-protein antigen conjugates, the preparation thereof and the use thereof
US6555115B1 (en) * 1998-12-08 2003-04-29 Corixa Corporation Compounds and methods for treatment and diagnosis of chlamydial infection
US7329408B2 (en) * 1999-12-01 2008-02-12 Novartis Vaccines And Diagnostics, Inc. Eliciting HCV-specific antibodies

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8182821B2 (en) * 2000-06-06 2012-05-22 The Macfarlane Burnet Institute For Medical Research And Public Health Ltd. Flu vaccine admixture of mannan and flu antigen
US20100008951A1 (en) * 2000-06-06 2010-01-14 The Macfarlane Burnet Institute For Medical Research And Public Health Limited Flu vaccine admixture of mannan and flu antigen
WO2004091519A3 (en) * 2003-04-11 2007-08-02 Medimmune Inc Methods of preventing or treating respiratory conditions
US20050147607A1 (en) * 2003-04-11 2005-07-07 Reed Jennifer L. Methods of preventing or treating respiratory conditions
US7582297B2 (en) 2003-04-11 2009-09-01 Medimmune, Llc Methods of treating respiratory conditions
US9198885B2 (en) 2005-07-14 2015-12-01 Neothetics, Inc. Lipolytic methods for regional adiposity comprising salmeterol or formoterol
US9370498B2 (en) 2005-07-14 2016-06-21 Neothetics, Inc. Methods of using lipolytic formulations for regional adipose tissue treatment
US9707192B2 (en) 2005-07-14 2017-07-18 Neothetics, Inc. Lipolytic methods
US9452147B2 (en) 2005-07-14 2016-09-27 Neothetics, Inc. Lipolytic methods
US20110105446A1 (en) * 2005-07-14 2011-05-05 Lithera, Inc. Sustained Release Enhanced Lipolytic Formulation for Regional Adipose Tissue Treatment
US20090130131A1 (en) * 2005-08-05 2009-05-21 Hiroshi Kido Antigen-Drug Vehicle Enabling Switch From Selective Production of IgA Antibody to Production of Both of IgA and IgG Antibodies and Transnasal/Mucosal Vaccine Using the Same
US8211442B2 (en) * 2005-08-05 2012-07-03 The University Of Tokushima Mucosal vaccine enabling switching from production of IgA antibody to production of both of IgA and IgG antibodies
US20080031872A1 (en) * 2006-08-02 2008-02-07 Karp Nelson M HIV immunogenic composition and method of administration
US20100119609A1 (en) * 2006-10-17 2010-05-13 John Daniel Dobak Methods, compositions, and formulations for the treatment of thyroid eye disease
US9452132B2 (en) 2009-05-27 2016-09-27 Neothetics, Inc. Methods for administration and formulations for the treatment of regional adipose tissue
US9132084B2 (en) 2009-05-27 2015-09-15 Neothetics, Inc. Methods for administration and formulations for the treatment of regional adipose tissue
US20110130373A1 (en) * 2009-05-27 2011-06-02 Lithera, Inc. Methods for administration and formulations for the treatment of regional adipose tissue
US20110224176A1 (en) * 2010-01-15 2011-09-15 Lithera, Inc. Lyophilized Cake Formulations
US9597392B2 (en) 2010-05-10 2017-03-21 Ascend Biopharmaceuticals Pty Ltd. Use of high molecular weight mannan for inducing and/or enhancing an immune response
US9597531B2 (en) 2010-11-24 2017-03-21 Neothetics, Inc. Selective, lipophilic, and long-acting beta agonist monotherapeutic formulations and methods for the cosmetic treatment of adiposity and contour bulging

Also Published As

Publication number Publication date
US20110033493A1 (en) 2011-02-10
WO2001093912A1 (en) 2001-12-13
US8182821B2 (en) 2012-05-22
EP1301208A4 (de) 2004-11-17
AU2006222717A1 (en) 2006-10-19
ATE456958T1 (de) 2010-02-15
EP2066332A1 (de) 2009-06-10
EP2066332A4 (de) 2012-01-18
EP1301208B1 (de) 2010-02-03
AUPQ797700A0 (en) 2000-06-29
US20100008951A1 (en) 2010-01-14
HK1056830A1 (en) 2004-03-05
AU6365301A (en) 2001-12-17
EP1301208A1 (de) 2003-04-16
DE60141243D1 (de) 2010-03-25
AU2007302633A1 (en) 2008-04-03

Similar Documents

Publication Publication Date Title
US20110033493A1 (en) Vaccine
Su et al. Induction of mucosal immunity through systemic immunization: Phantom or reality?
Holmgren et al. Mucosal immunisation and adjuvants: a brief overview of recent advances and challenges
Hu et al. Immunostimulating complexes (ISCOMs) for nasal vaccination
Rhee et al. Mucosal vaccine adjuvants update
JP2001509491A (ja) アジュバントとしてモノグリセリドまたはジグリセリドを含む抗原送達系
Po et al. Design and delivery of non-parenteral vaccines.
WO2003051394A2 (en) Calcium phosphate particles as mucosal adjuvants
McGhee et al. The common mucosal immune system: from basic principles to enteric vaccines with relevance for the female reproductive tract
CN102711815B (zh) 用于疫苗的佐剂、包含所述佐剂的疫苗及其用途
Anggraeni et al. Development of mucosal vaccine delivery: an overview on the mucosal vaccines and their adjuvants
Stambas et al. Oxidised mannan as a novel adjuvant inducing mucosal IgA production
McCluskie et al. Mucosal immunization with DNA vaccines
AU2001263653B2 (en) Vaccine
AU2001263653A1 (en) Vaccine
US20220257752A1 (en) New use of cyclic dinucleotides
WO2008037033A1 (en) Flu vaccine admixture of mannan and flu antigen
Meeusen et al. Advances in mucosal vaccination
Peter et al. Development and use of mucosal vaccines: Potential and limitations
US6946448B2 (en) In utero oral nucleic acid immunization
Tian et al. The development of a human Brucella mucosal vaccine: What should be considered?
US20060029618A1 (en) Vaccine against dental caries based on virulence-associated immunomodulatory extracellular proteins produced by the cariogenic bacteria streptococcus sobrinus and streptococcus mutans
Li Exploring mucosal vaccination as an alternative to conventional vaccines
KR20240043865A (ko) 점막에서 항원 특이적 면역 반응을 증진시키는 신규한 펩타이드 및 상기 펩타이드를 유효성분으로 포함하는 점막 백신 보조제
Lavelle et al. Mucosal Vaccines, Adjuvants and Delivery 2016-A Meetings Management Conference, Lausanne, Switzerland, September 14-16, 2016

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITY OF MELBOURNE, THE, AUSTRALIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MCKENZIE, IAN F. C.;PIETERSZ, GEOFFREY A.;CHEERS, CHRISTINA;AND OTHERS;REEL/FRAME:014063/0566;SIGNING DATES FROM 20030226 TO 20030401

Owner name: STAMBAS, JOHN, AUSTRALIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MCKENZIE, IAN F. C.;PIETERSZ, GEOFFREY A.;CHEERS, CHRISTINA;AND OTHERS;REEL/FRAME:014063/0566;SIGNING DATES FROM 20030226 TO 20030401

Owner name: AUSTIN RESEARCH INSTITUTE, THE, AUSTRALIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MCKENZIE, IAN F. C.;PIETERSZ, GEOFFREY A.;CHEERS, CHRISTINA;AND OTHERS;REEL/FRAME:014063/0566;SIGNING DATES FROM 20030226 TO 20030401

AS Assignment

Owner name: CHEERS, CHRISTINA (0.5%), AUSTRALIA

Free format text: RECORDAL OF AN INTEREST;ASSIGNOR:STAMBAS, JOHN;REEL/FRAME:016010/0976

Effective date: 20040516

Owner name: STAMBAS, JOHN (24.5%), AUSTRALIA

Free format text: RECORDAL OF AN INTEREST;ASSIGNOR:STAMBAS, JOHN;REEL/FRAME:016010/0976

Effective date: 20040516

AS Assignment

Owner name: MELBOURNE, UNIVERSITY OF, THE (25.0%), AUSTRALIA

Free format text: RECORDAL OF AN INTEREST;ASSIGNORS:MELBOURNE, UNIVERSITY OF, THE;AUSTIN RESEARCH INSTITUTE, THE;STAMBAS, JOHN;AND OTHERS;REEL/FRAME:017306/0479

Effective date: 20030630

Owner name: AUSTIN RESEARCH INSTITUTE, THE (50.0%), AUSTRALIA

Free format text: RECORDAL OF AN INTEREST;ASSIGNORS:MELBOURNE, UNIVERSITY OF, THE;AUSTIN RESEARCH INSTITUTE, THE;STAMBAS, JOHN;AND OTHERS;REEL/FRAME:017306/0479

Effective date: 20030630

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION