US20100183667A1 - Vaccine Comprising an Oil in Water Emulsion Adjuvant - Google Patents

Vaccine Comprising an Oil in Water Emulsion Adjuvant Download PDF

Info

Publication number
US20100183667A1
US20100183667A1 US12/445,090 US44509007A US2010183667A1 US 20100183667 A1 US20100183667 A1 US 20100183667A1 US 44509007 A US44509007 A US 44509007A US 2010183667 A1 US2010183667 A1 US 2010183667A1
Authority
US
United States
Prior art keywords
oil
spp
dose
vaccine
composition according
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
US12/445,090
Other languages
English (en)
Inventor
William Ripley Ballou, JR.
Emmanuel Jules Hanon
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.)
GlaxoSmithKline Biologicals SA
Original Assignee
GlaxoSmithKline Biologicals SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB0620336.8A external-priority patent/GB0620336D0/en
Priority claimed from GB0620337A external-priority patent/GB0620337D0/en
Priority claimed from GB0620815A external-priority patent/GB0620815D0/en
Priority claimed from GB0620816A external-priority patent/GB0620816D0/en
Priority claimed from PCT/EP2006/069979 external-priority patent/WO2007071711A2/en
Priority claimed from GB0707697A external-priority patent/GB0707697D0/en
Priority claimed from GB0711357A external-priority patent/GB0711357D0/en
Priority claimed from GB0712062A external-priority patent/GB0712062D0/en
Application filed by GlaxoSmithKline Biologicals SA filed Critical GlaxoSmithKline Biologicals SA
Assigned to GLAXOSMITHKLINE BIOLOGICALS SA reassignment GLAXOSMITHKLINE BIOLOGICALS SA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BALLOU, WILLIAM RIPLEY, JR, HANON, EMMANUEL JULES
Publication of US20100183667A1 publication Critical patent/US20100183667A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K39/12Viral 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
    • A61K39/295Polyvalent viral antigens; Mixtures of viral and bacterial antigens
    • 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
    • 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
    • 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
    • A61P35/00Antineoplastic agents
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • 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
    • A61K2039/555Medicinal 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
    • A61K2039/70Multivalent vaccine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16211Influenzavirus B, i.e. influenza B virus
    • C12N2760/16234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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 improved vaccine and immunogenic compositions and their use in medicine.
  • the invention relates to vaccine or immunogenic formulations comprising an oil-in-water emulsion adjuvant and their use in medicine, in particular their use in augmenting immune responses to various antigens, and to methods of preparation, wherein the oil in water emulsion comprises a tocol, a metabolisable oil and an emulsifying agent.
  • compositions or vaccines with an improved immunogenicity are always needed.
  • adjuvants have been used to try and improve the immune response raised to any given antigen and/or reduce reactogenicity/toxicity in the host.
  • Oil in water emulsions per se are well known in the art, and have been suggested to be useful as adjuvant compositions (EP 399843; WO 95/17210).
  • WO95/17210 discloses oil in water emulsions comprising from 2 to 10% squalene, from 2 to 10% alpha tocopherol and from 0.3 to 3% tween 80 and their use alone or in combination with QS21 and/or 3D-MPL.
  • WO99/12565 discloses oil in water emulsion compositions comprising a metabolisable oil, a saponin and a sterol.
  • the oil in water emulsions further comprise 3D-MPL.
  • WO99/11241 discloses oil in water emulsions comprising metabolisable oil and a saponin, wherein the oil and saponin are present in a ratio of between 1:1 and 200:1.
  • vaccine or immunogenic compositions comprising lower amounts of each component of the oil in water emulsion may be used whilst still maintaining a comparable immune response against an antigen or antigenic composition within said composition.
  • This carries the advantage maintaining the level of immunogenicity against an antigen whilst the reactogenicity within the host recipient is reduced.
  • an immunogenic composition comprising an antigen or antigenic composition, and an adjuvant composition comprising an oil-in-water emulsion, wherein said oil-in-water emulsion comprises 0.5-10 mg metabolisable oil, 0.5-11 mg tocol and 0.4-4 mg emulsifying agent, per human dose.
  • a vaccine composition comprising an antigen or antigenic composition, and an adjuvant composition comprising an oil-in-water emulsion, wherein said oil-in-water emulsion comprises 0.5-10 mg metabolisable oil, 0.5-11 mg tocol and 0.4-4 mg emulsifying agent, per human dose.
  • a vaccine or immunogenic composition comprising an antigen or antigenic composition, and an adjuvant composition comprising an oil-in-water emulsion wherein said oil-in-water emulsion comprises 0.5-10 mg metabolisable oil, 0.5-11 mg tocol and 0.4-4 mg emulsifying agent in the manufacture of an immunogenic composition for the prevention of infection and/or disease.
  • a method or use as hereinabove defined, for protection against infection or disease caused by a pathogen which is a variant of the pathogen from which the antigen in the immunogenic composition is derived in another embodiment, there is provided a method or use as hereinabove defined for protection against infections or disease caused by a pathogen which comprises an antigen which is a variant of that antigen in the immunogenic composition.
  • FIG. 1 Clinical trial: geometric mean titers (GMTs) for anti-HA antibody at different timepoints (ATP cohort for immunogenicity).
  • FIG. 2 Clinical trial: seroprotection rate (SPR) for HI antibody titer with 95% confidence interval at day 0 and day 21 (ATP cohort for immunogenicity).
  • SPR seroprotection rate
  • FIG. 3 Clinical trial: seroconversion rate (SCR) for HI antibody titer with 95% confidence interval at day 21 (ATP cohort for immunogenicity).
  • FIG. 4 Clinical trial: seroconversion factor (SCF) for HI antibody titer with 95% confidence interval at day 21 (ATP cohort for immunogenicity).
  • SCF seroconversion factor
  • FIG. 5 Mice study: Haemagglutinin Inhibition test (GMT+/ ⁇ IC95) in BALB/c mice primed with heterosubtypic strains (dose range AS03).
  • FIG. 5A Anti-A/New Calcdonia/20/99 HI titers;
  • FIG. 5B Anti-A/Panama/2007/99 HI titers.
  • FIG. 5C Anti-B/Shandong/7/97 HI titers.
  • FIG. 6 Mice study: Haemagglutinin Inhibition test (GMT+/ ⁇ IC95) in C57Bl/6 mice primed with heterosubtypic strains (dose range AS03).
  • FIG. 7 Mice study: Cellular immune response (CD4+ T cell) in PBMC from C57Bl/6 mice primed with heterosubtypic strains (dose range AS03).
  • FIG. 8 Mice study: Cellular immune response (CD4+ T cell) in PBMC from C57Bl/6 mice primed with heterosubtypic strains and immunized with low dose antigen (0.5 ⁇ g) adjuvanted with dose range AS03.
  • FIG. 9 Mice study: H5N1-specific serum Ig ELISA titers (A and B) and anti-H5N1 IgG1 (C and D) and IgG2b (E and F) isotypic responses on day 14 post-immunization (GMT+/ ⁇ IC95) for two different antigen dose: 1.5 ⁇ g (A, C and E) or 0.38 ⁇ g (B, D and F)
  • FIG. 10 Mice study: Hemagglutination inhibition test (GMT+/ ⁇ IC95) on day 21 post-immunization (GMT+/ ⁇ IC95) for two different antigen dose: 1.5 ⁇ g (A) or 0.38 ⁇ g (B).
  • FIG. 11 Mice study: Cellular immune response (CD4+ T cell) in na ⁇ ve C57Bl/6 mice immunized with different dose of H5N1 vaccine (1.5 or 0.38 ⁇ g) adjuvanted with dose range AS03: (A) 1.5 ⁇ g HA Ag (antigen) or (B) 0.38 ⁇ g HA Ag (antigen).
  • FIG. 12 Pigs study. Haemagglutinin Inhibition test (GMT+/ ⁇ IC95) in pigs primed with homologous strains (dose range AS03).
  • Embodiments herein relating to “vaccine compositions” of the invention are also applicable to embodiments relating to “immunogenic compositions” of the invention, and vice versa.
  • a vaccine or immunogenic composition comprising an antigen or antigen composition and an adjuvant composition consisting of an oil in water emulsion, wherein said oil in water emulsion comprises 0.5-10 mg metabolisable oil, 0.5-11 mg tocol and 0.4-4 mg emulsifying agent, per human dose.
  • a vaccine or immunogenic composition comprising an antigen or antigen composition and an adjuvant composition comprising an oil in water emulsion, wherein the oil in water emulsion comprises 0.5-10 mg metabolisable oil, (such as squalene), 0.5-11 mg tocol (such as alpha-tocopherol and 0.4-4 mg emulsifying agent (such as polyoxyethylene sorbitan monooleate), per human dose.
  • the oil in water emulsion comprises 0.5-10 mg metabolisable oil, (such as squalene), 0.5-11 mg tocol (such as alpha-tocopherol and 0.4-4 mg emulsifying agent (such as polyoxyethylene sorbitan monooleate), per human dose.
  • the adjuvant composition of the invention comprises an oil-in-water emulsion adjuvant, preferably said emulsion comprises a metabolisable oil in an amount of 0.5-10 mg, a tocol in an amount of 0.5-11 mg and an emulsifying agent in an amount of 0.4-4 mg and having oil droplets of which at least 70% by intensity have diameters of less than 1 ⁇ m.
  • the oil phase of the emulsion system has to comprise a metabolisable oil.
  • metabolisable oil is well known in the art. Metabolisable can be defined as ‘being capable of being transformed by metabolism’ (Dorland's Illustrated Medical Dictionary, W.B. Sanders Company, 25th edition (1974)).
  • the oil may be any vegetable oil, fish oil, animal oil or synthetic oil, which is not toxic to the recipient and is capable of being transformed by metabolism. Nuts, seeds, and grains are common sources of vegetable oils. Synthetic oils are also part of this invention and can include commercially available oils such as NEOBEE® and others. A particularly suitable metabolisable oil is squalene.
  • Squalene (2,6,10,15,19,23-Hexamethyl-2,6,10,14,18,22-tetracosahexaene) is an unsaturated oil which is found in large quantities in shark-liver oil, and in lower quantities in olive oil, wheat germ oil, rice bran oil, and yeast, and is a particularly preferred oil for use in this invention.
  • Squalene is a metabolisable oil by virtue of the fact that it is an intermediate in the biosynthesis of cholesterol (Merck index, 10th Edition, entry no. 8619).
  • the metabolisable oil is present in the adjuvant composition in an amount of 0.5-10 mg, preferably 1-10, 2-10, 3-9, 4-8, 5-7, or 5-6 mg (e.g.
  • the metabolisable oil is present in the vaccine (or immunogenic) composition in an amount of 0.5-10 mg, preferably 1-10, 2-10, 3-9, 4-8, 5-7, or 5-6 mg (e.g. 2-3, 5-6, or 9-10 mg), specifically 5.35 mg or 2.14 mg.
  • the amount of metabolisable oil in vaccine or immunogenic composition may be expressed as a percentage of the total composition.
  • the metabolisable oil is present in the vaccine composition in an amount of 0.5% to 2%, preferably 0.25-2, or 0.25-1.75, or 0.5-1.65, or 0.6-1.5, or 0.8-1.4 or 1-1.25% (v/v) oil of the total composition volume.
  • the metabolisable oil is present in a final amount of about 1.25% of the total volume of the vaccine (or immunogenic) composition. In another specific embodiment, the metabolisable oil is present in a final amount of 0.25% (v/v) of the total composition volume.
  • concentrations given in v/v can be converted into concentration in w/v by applying the following conversion factor: a 5% (v/v) squalene concentration is equivalent to a 4.28% (w/v) squalene concentration.
  • the oil in water emulsion comprises a tocol.
  • Tocols are well known in the art and are described in EP0382271.
  • the tocol is alpha-tocopherol or a derivative thereof such as alpha-tocopherol succinate (also known as vitamin E succinate).
  • Said tocol is suitably present in the adjuvant composition in an amount of 0.5-11 mg, preferably 1-11, 2-10, 3-9, 4-8, 5-7, 5-6 (e.g. 10-11, 5-6, 2.5-3.5 or 1-3 mg).
  • the tocol is present in an amount of 5.94 mg or 2.38 mg.
  • said tocol is suitably present in the vaccine (or immunogenic) composition in an amount of 0.5-11 mg, preferably 1-11, 2-10, 3-9, 4-8, 5-7, 5-6 (e.g. 10-11, 5-6, 2.5-3.5 or 1-3 mg). In a specific embodiment the tocol is present in an amount of 5.94 mg or 2.38 mg.
  • the amount of tocol may be expressed as a percentage of the total vaccine or immunogenic composition volume.
  • Suitably tocol is present in the vaccine composition in an amount 0.25% to 2% (v/v) of the total volume of the immunogenic composition, preferably at 0.25-2 comprises 0.25-2, or 0.25-1.75, or 0.5-1.65, or 0.6-1.5, or 0.8-1.4 or 1-1.25% (v/v) tocol of the total volume.
  • tocol is present in an amount of between 0.2% and 2% (v/v) of the total volume of the vaccine (or immunogenic) composition, more preferably at an amount of 1.25% (v/v) in a 0.5 ml dose volume.
  • the tocol is present in a final amount of about 1.25% of the total volume of the vaccine or immunogenic composition. In another specific embodiment, the tocol is present in a final amount of 0.25% (v/v) of the total volume or 1.25% (v/v) in 0.5 ml dose volume or 0.9% (v/v), in 0.7 ml dose volume, or 0.5% (v/v) in 0.5 ml dose or 0.35-0.37%, preferably 0.36% in 0.7 ml vaccine or immunogenic dose.
  • concentrations given in v/v can be converted into concentration in w/v by applying the following conversion factor: a 5% (v/v) alpha-tocopherol concentration is equivalent to a 4.8% (w/v) alpha-tocopherol concentration.
  • the oil in water emulsion further comprises an emulsifying agent.
  • the emulsifying agent may suitably be polyoxyethylene sorbitan monooleate.
  • the emulsifying agent may be selected from the group comprising: Polysorbate® 80 or Tween® 80.
  • Said emulsifying agent is suitably present in the adjuvant composition in an amount of 0.1-5, 0.2-5, 0.3-4, 0.4-3 or 2-3 mg (e.g. 0.4-1.2, 2-3 or 4-5 mg) emulsifying agent.
  • the emulsifying agent is present in an amount of 0.97 mg or 2.425 mg.
  • said emulsifying agent is suitably present in the vaccine or immunogenic composition in an amount of 0.1-5, 0.2-5, 0.3-4, 0.4-3 or 2-3 mg (e.g. 0.4-1.2, 2-3 or 4-5 mg) emulsifying agent.
  • the emulsifying agent is present in an amount of 0.97 mg or 2.425 mg.
  • the amount of emulsifying agent may be expressed as a percentage of the total vaccine or immunogenic composition volume.
  • the emulsifying agent is present in the vaccine (or immunogenic) composition in an amount 0.125-0.8% (v/v) of the total volume of the composition, preferably at 0.08-0.05, or 0.1-0.7, or 0.2-0.6, or 0.25-0.55, or 0.3-0.52 or 0.4-0.5% (v/v) of the total volume.
  • the emulsifying agent is present in an amount of 1%, 0.5% or 0.2% (v/v) of the total vaccine or immunogenic composition volume.
  • concentrations given in v/v can be converted into concentration in w/v by applying the following conversion factor: a 1.8% (v/v) polysorbate 80 concentration is equivalent to a 1.91% (w/v) polysorbate 80 concentration.
  • a 0.5 ml vaccine or immunogenic dose volume contains 0.45% (v/v) Tween 80, and a 0.7 ml dose volume contains 0.315% (v/v) Tween 80.
  • a 0.5 ml dose contains 0.18% (v/v) emulsifying agent and a 0.7 ml vaccine or immunogenic composition dose contains 0.126% (v/v) emulsifying agent.
  • human dose is meant a dose which is in a volume suitable for human use. Generally this is between 0.25 and 1.5 ml. In one embodiment, a human dose is 0.5 ml. In a further embodiment, a human dose is higher than 0.5 ml, for example 0.6, 0.7, 0.8, 0.9 or 1 ml. In a further embodiment, a human dose is between 1 ml and 1.5 ml. In another embodiment, in particular when the immunogenic composition is for the paediatric population, a human dose may be less than 0.5 ml such as between 0.25 and 0.5 ml.
  • compositions comprise the following adjuvant components in the following amounts are in a final volume of human dose of 0.5 ml:
  • the invention further provides an adjuvant composition comprising the individual components as defined herein above and in the amount defined above, for example but not exclusively as illustrated in Table 1.
  • an adjuvant composition will be in a human dose suitable volume.
  • the adjuvant is in a liquid form to be combined with a liquid form of an antigenic composition
  • the adjuvant composition will be in a human dose suitable volume which is a fraction of the intended final volume of the human dose, such as for example approximately half of the intended final volume of the human dose, for example a 350 ⁇ l volume for an intended human dose of 0.7 ml, or a 250 ⁇ l volume for an intended human dose of 0.5 ml.
  • the adjuvant composition is diluted when combined with the antigen composition to provide the final human dose of vaccine.
  • the final volume of such dose will of course vary dependent on the initial volume of the adjuvant composition and the volume of antigen composition added to the adjuvant composition.
  • liquid adjuvant is used to reconstitute a lyophilised antigen composition.
  • the human dose suitable volume of the adjuvant composition is approximately equal to the final volume of the human dose.
  • the liquid adjuvant composition is added to the vial containing the lyophilised antigen composition.
  • the final human dose can vary between 0.5 and 1.5 ml.
  • the method of producing oil-in-water emulsions is well known to the person skilled in the art.
  • the method comprises mixing the tocol-containing oil phase with a surfactant such as a PBS/TWEEN80TM solution, followed by homogenisation using a homogenizer, it would be clear to a man skilled in the art that a method comprising passing the mixture twice through a syringe needle would be suitable for homogenising small volumes of liquid.
  • microfluidiser M110S Microfluidics machine, maximum of 50 passes, for a period of 2 minutes at maximum pressure input of 6 bar (output pressure of about 850 bar)
  • output pressure of about 850 bar
  • the adaptation could be achieved by routine experimentation comprising the measurement of the resultant emulsion until a preparation was achieved with oil droplets of the required diameter.
  • the oil and emulsifier should be in an aqueous carrier.
  • the aqueous carrier may be, for example, phosphate buffered saline.
  • the oil-in-water emulsion systems of the present invention have a small oil droplet size in the sub-micron range.
  • the droplet sizes will be in the range 120 to 750 nm, more preferably sizes from 120 to 600 nm in diameter.
  • the oil-in water emulsion contains oil droplets of which at least 70% by intensity are less than 500 nm in diameter, more preferably at least 80% by intensity are less than 300 nm in diameter, more preferably at least 90% by intensity are in the range of 120 to 200 nm in diameter.
  • the oil droplet size i.e. diameter
  • the oil droplet size is given by intensity.
  • Intensity is measured by use of a sizing instrument, suitably by dynamic light scattering such as the Malvern Zetasizer 4000 or preferably the Malvern Zetasizer 3000HS.
  • a detailed procedure is given in Example II.2.
  • a first possibility is to determine the z average diameter ZAD by dynamic light scattering (PCS-Photon correlation spectroscopy); this method additionally give the polydispersity index (PDI), and both the ZAD and PDI are calculated with the cumulants algorithm. These values do not require the knowledge of the particle refractive index.
  • a second mean is to calculate the diameter of the oil droplet by determining the whole particle size distribution by another algorithm, either the Contin, or NNLS, or the automatic “Malvern” one (the default algorithm provided for by the sizing instrument). Most of the time, as the particle refractive index of a complex composition is unknown, only the intensity distribution is taken into consideration, and if necessary the intensity mean originating from this distribution.
  • a vaccine or immunogenic composition comprising an antigen or antigen composition and an adjuvant composition comprising an oil in water emulsion and optionally one or more further immunostimulants, wherein said oil in water emulsion comprises 0.5-10 mg metabolisable oil, 0.5-11 mg tocol and 0.4-4 mg emulsifying agent.
  • the adjuvant composition comprises an oil and water emulsion as described herein.
  • the adjuvant composition may further comprise one or more additional adjuvants or immunostimulants.
  • the adjuvant composition optionally comprises one or more additional adjuvants or immunostimulants other than QS21 and/or MPL.
  • the optional additional adjuvant is selected from the group: a saponin, lipid A or a derivative thereof, an immunostimulatory oligonucleotide, an alkyl glucosaminide phosphate, a metal salt, a toll-like receptor agonist or combinations thereof. It is preferred that the adjuvant is a Toll like receptor agonist in particular an agonist of a Toll like receptor 2, 3, 4, 7, 8 or 9, or a saponin. It is further preferred that the adjuvant system comprises two or more adjuvants from the above list.
  • Combinations preferably contain a saponin (in particular QS21) adjuvant and/or a Toll like receptor 4 against such as 3D-MPL or a Toll like receptor 9 agonist such as a CpG containing immunostimulatory oligonucleotide.
  • a saponin (in particular QS21) and a Toll like receptor 4 agonist such as a saponin (in particular QS21) and a Toll like receptor 4 ligand such as 3D-MPL or an alkyl glucosaminide phosphate.
  • the additional adjuvant is a Toll like receptor (TLR) 4 ligand, preferably an agonist such as a lipid A derivative particularly monophosphoryl lipid A or more particularly 3 Deacylated monophoshoryl lipid A (3 D-MPL).
  • TLR Toll like receptor
  • 3D-MPL is available under the trademark MPL® by GlaxoSmithKline Biologicals North America and primarily promotes CD4+ T cell responses with an IFN-g (Th1) phenotype. It can be produced according to the methods disclosed in GB 2 220 211 A. Chemically it is a mixture of 3-deacylated monophosphoryl lipid A with 3, 4, 5 or 6 acylated chains. Preferably in the compositions of the present invention small particle 3 D-MPL is used. Small particle 3 D-MPL has a particle size such that it may be sterile-filtered through a 0.22 ⁇ m filter. Such preparations are described in International Patent Application No. WO 94/21292. Synthetic derivatives of lipid A are known and thought to be TLR 4 agonists including, but not limited to:
  • TLR4 ligands which may be used are alkyl Glucosaminide phosphates (AGPs) such as those disclosed in WO9850399 or U.S. Pat. No. 6,303,347 (processes for preparation of AGPs are also disclosed), or pharmaceutically acceptable salts of AGPs as disclosed in U.S. Pat. No. 6,764,840.
  • AGPs alkyl Glucosaminide phosphates
  • Some AGPs are TLR4 agonists, and some are TLR4 antagonists. Both are thought to be useful as adjuvants.
  • TLR-4 ligands capable of causing a signalling response through TLR-4 (Sabroe et al, JI 2003 p1630-5) are, for example, lipopolysaccharide from gram-negative bacteria and its derivatives, or fragments thereof, in particular a non-toxic derivative of LPS (such as 3D-MPL).
  • TLR agonist examples include heat shock protein (HSP) 10, 60, 65, 70, 75 or 90; surfactant Protein A, hyaluronan oligosaccharides, heparan sulphate fragments, fibronectin fragments, fibrinogen peptides and b-defensin-2, muramyl dipeptide (MDP) or F protein of respiratory syncitial virus.
  • HSP heat shock protein
  • surfactant Protein A hyaluronan oligosaccharides, heparan sulphate fragments, fibronectin fragments, fibrinogen peptides and b-defensin-2
  • MDP muramyl dipeptide
  • F protein of respiratory syncitial virus amyl dipeptide
  • TLRs Toll-like receptors
  • TLRs are type I transmembrane receptors, evolutionarily conserved between insects and humans. Ten TLRs have so far been established (TLRs 1-10) (Sabroe et al, JI 2003 p1630-5). Members of the TLR family have similar extracellular and intracellular domains; their extracellular domains have been shown to have leucine-rich repeating sequences, and their intracellular domains are similar to the intracellular region of the interleukin-1 receptor (IL-1R). TLR cells are expressed differentially among immune cells and other cells (including vascular epithelial cells, adipocytes, cardiac myocytes and intestinal epithelial cells).
  • IL-1R interleukin-1 receptor
  • the intracellular domain of the TLRs can interact with the adaptor protein Myd88, which also posses the IL-1R domain in its cytoplasmic region, leading to NF-KB activation of cytokines; this Myd88 pathway is one way by which cytokine release is effected by TLR activation.
  • the main expression of TLRs is in cell types such as antigen presenting cells (eg dendritic cells, macrophages etc).
  • TLRs recognise different types of agonists, although some agonists are common to several TLRs.
  • TLR agonists are predominantly derived from bacteria or viruses, and include molecules such as flagellin or bacterial lipopolysaccharide (LPS).
  • TLR agonist it is meant a component which is capable of causing a signalling response through a TLR signalling pathway, either as a direct ligand or indirectly through generation of endogenous or exogenous ligand (Sabroe et al, JI 2003 p1630-5).
  • TLR molecules are used as optional additional immunostimulants. These could include, but are not limited to agonists for TLR2, TLR3, TLR7, TLR8 and TLR9.
  • a TLR agonist is used that is capable of causing a signalling response through TLR-1 (Sabroe et al, JI 2003 p1630-5).
  • the TLR agonist capable of causing a signalling response through TLR-1 is selected from: Tri-acylated lipopeptides (LPs); phenol-soluble modulin; Mycobacterium tuberculosis LP; S-(2,3-bis(palmitoyloxy)-(2-RS)-propyl)-N-palmitoyl-(R)-Cys-(S)-Ser-(S)-Lys(4)-OH, trihydrochloride (Pam 3 Cys) LP which mimics the acetylated amino terminus of a bacterial lipoprotein and OspA LP from Borrelia burgdorfei.
  • a TLR agonist is used that is capable of causing a signalling response through TLR-2 (Sabroe et al, JI 2003 p1630-5).
  • the TLR agonist capable of causing a signalling response through TLR-2 is one or more of a lipoprotein, a peptidoglycan, a bacterial lipopeptide from M tuberculosis, B burgdorferi.
  • T pallidum peptidoglycans from species including Staphylococcus aureus ; lipoteichoic acids, mannuronic acids, Neisseria porins , bacterial fimbriae, Yersina virulence factors, CMV virions, measles haemagglutinin, and zymosan from yeast.
  • a TLR agonist is used that is capable of causing a signalling response through TLR-3 (Sabroe et al, JI 2003 p1630-5).
  • the TLR agonist capable of causing a signalling response through TLR-3 is double stranded RNA (dsRNA), or polyinosinic-polycytidylic acid (Poly IC), a molecular nucleic acid pattern associated with viral infection.
  • dsRNA double stranded RNA
  • Poly IC polyinosinic-polycytidylic acid
  • a TLR agonist is used that is capable of causing a signalling response through TLR-5 (Sabroe et al, JI 2003 p1630-5).
  • the TLR agonist capable of causing a signalling response through TLR-5 is bacterial flagellin.
  • a TLR agonist is used that is capable of causing a signalling response through TLR-6 (Sabroe et al, JI 2003 p1630-5).
  • the TLR agonist capable of causing a signalling response through TLR-6 is mycobacterial lipoprotein, di-acylated LP, and phenol-soluble modulin.
  • TLR6 agonists are described in WO2003043572.
  • a TLR agonist is used that is capable of causing a signalling response through TLR-7 (Sabroe et al, JI 2003 p1630-5).
  • the TLR agonist capable of causing a signalling response through TLR-7 is a single stranded RNA (ssRNA), loxoribine, a guanosine analogue at positions N7 and C8, or an imidazoquinoline compound, or derivative thereof.
  • the TLR agonist is imiquimod.
  • TLR7 agonists are described in WO02085905.
  • a TLR agonist is used that is capable of causing a signalling response through TLR-8 (Sabroe et al, JI 2003 p1630-5).
  • the TLR agonist capable of causing a signalling response through TLR-8 is a single stranded RNA (ssRNA), an imidazoquinoline molecule with anti-viral activity, for example resiquimod (R848); resiquimod is also capable of recognition by TLR-7.
  • ssRNA single stranded RNA
  • R848 imidazoquinoline molecule with anti-viral activity
  • resiquimod resiquimod
  • Other TLR-8 agonists which may be used include those described in WO2004071459.
  • Immunostimulatory oligonucleotides or any other Toll-like receptor (TLR) 9 agonist may also be used.
  • the preferred oligonucleotides for use in adjuvants or vaccines or immunogenic compositions of the present invention are CpG containing oligonucleotides, preferably containing two or more dinucleotide CpG motifs separated by at least three, more preferably at least six or more nucleotides.
  • a CpG motif is a Cytosine nucleotide followed by a Guanine nucleotide.
  • the CpG oligonucleotides of the present invention are typically deoxynucleotides.
  • the internucleotide in the oligonucleotide is phosphorodithioate, or more preferably a phosphorothioate bond, although phosphodiester and other internucleotide bonds are within the scope of the invention.
  • oligonucleotides with mixed internucleotide linkages are included within the scope of the invention. Methods for producing phosphorothioate oligonucleotides or phosphorodithioate are described in U.S. Pat. No. 5,666,153, U.S. Pat. No. 5,278,302 and WO95/26204.
  • oligonucleotides have the following sequences.
  • the sequences preferably contain phosphorothioate modified internucleotide linkages.
  • OLIGO 1 (SEQ ID NO: 1): TCC ATG ACG TTC CTG ACG TT (CpG 1826)
  • OLIGO 2 (SEQ ID NO: 2): TCT CCC AGC GTG CGC CAT (CpG 1758)
  • OLIGO 3 (SEQ ID NO: 3): ACC GAT GAC GTC GCC GGT GAC GGC ACC ACG
  • OLIGO 4 (SEQ ID NO: 4): TCG TCG TTT TGT CGT TTT GTC GTT (CpG 2006)
  • OLIGO 5 (SEQ ID NO: 5): TCC ATG ACG TTC CTG ATG CT (CpG 1668)
  • OLIGO 6 (SEQ ID NO: 6): TCG ACG TTT TCG GCG CGC GCC G (CpG 5456)
  • CpG oligonucleotides may comprise the preferred sequences above in that they have inconsequential deletions or additions thereto.
  • the CpG oligonucleotides utilised in the present invention may be synthesized by any method known in the art (for example see EP 468520). Conveniently, such oligonucleotides may be synthesized utilising an automated synthesizer.
  • the adjuvant composition further comprises an additional immunostimulant which is selected from the group consisting of: a TLR-1 agonist, a TLR-2 agonist, TLR-3 agonist, a TLR-4 agonist, TLR-5 agonist, a TLR-6 agonist, TLR-7 agonist, a TLR-8 agonist, TLR-9 agonist, or a combination thereof.
  • an additional immunostimulant which is selected from the group consisting of: a TLR-1 agonist, a TLR-2 agonist, TLR-3 agonist, a TLR-4 agonist, TLR-5 agonist, a TLR-6 agonist, TLR-7 agonist, a TLR-8 agonist, TLR-9 agonist, or a combination thereof.
  • Quil A is a saponin preparation isolated from the South American tree Quilaja Saponaria Molina and was first described as having adjuvant activity by Dalsgaard et al. in 1974 (“Saponin adjuvants”, Archiv. für diedorf Virusforschung, Vol. 44, Springer Verlag, Berlin, p243-254). Purified fragments of Quil A have been isolated by HPLC which retain adjuvant activity without the toxicity associated with Quil A (EP 0 362 278), for example QS7 and QS21 (also known as QA7 and QA21).
  • QS-21 is a natural saponin derived from the bark of Quillaja saponaria Molina which induces CD8+ cytotoxic T cells (CTLs), Th1 cells and a predominant IgG2a antibody response and is a preferred saponin in the context of the present invention.
  • CTLs cytotoxic T cells
  • Th1 cells Th1 cells
  • IgG2a antibody response is a preferred saponin in the context of the present invention.
  • these formulations further comprise a sterol (WO96/33739).
  • a sterol WO96/33739
  • squalene and a saponin are included, it is of benefit to also include a sterol (optionally cholesterol) to the formulation as this allows a reduction in the total level of oil in the emulsion. This leads to a reduced cost of manufacture, improvement of the overall comfort of the vaccination, and also qualitative and quantitative improvements of the resultant immune responses, such as improved IFN- ⁇ production.
  • the vaccine or immunogenic formulations will contain an antigen or antigenic composition capable of eliciting an immune response against a human or animal pathogen.
  • said antigen or antigenic composition is derived from one or more of the following: HIV-1, (such as gag or fragments thereof such as p24, tat, nef, gp120 or gp160 or fragments of any of these), human herpes viruses, such as gD or derivatives thereof or Immediate Early protein such as ICP27 from HSV1 or HSV2, cytomegalovirus ((esp Human) (such as gB or derivatives thereof), Rotavirus (including live-attenuated viruses), Epstein Barr virus (such as gp350 or derivatives thereof), Varicella Zoster Virus (such as gpl, II and IE63), or from a hepatitis virus such as hepatitis B virus (for example Hepatitis B Surface antigen or a derivative thereof), hepatitis A virus, hepatitis C virus and hepatitis E virus, or from other viral pathogens, such as paramyxoviruses: Respiratory ra virus
  • Influenza virus whole live or inactivated virus, split influenza virus, grown in eggs or MDCK cells, or whole flu virosomes (as described by R. Gluck, Vaccine, 1992, 10, 915-920) or purified or recombinant proteins thereof, such as HA, NP, NA, or M proteins, or combinations thereof), or derived from bacterial pathogens such as Neisseria spp, including N. gonorrhea and N. meningitidis (for example capsular saccharides and conjugates thereof, transferrin-binding proteins, lactoferrin binding proteins, PilC, adhesins); S.
  • pyogenes for example M proteins or fragments thereof, C5A protease, lipoteichoic acids), S. agalactiae, S. mutans; H. ducreyi; Moraxella spp, including M catarrhalis , also known as Branhamella catarrhalis (for example high and low molecular weight adhesins and invasins); Bordetella spp, including B. pertussis (for example pertactin, pertussis toxin or derivatives thereof, filamenteous hemagglutinin, adenylate cyclase, fimbriae), B. parapertussis and B.
  • B. pertussis for example pertactin, pertussis toxin or derivatives thereof, filamenteous hemagglutinin, adenylate cyclase, fimbriae
  • Mycobacterium spp. including M. tuberculosis (for example ESAT6, Antigen 85A, -B or -C), M. bovis, M. leprae, M. avium, M. paratuberculosis, M. smegmatis; Legionella spp, including L. pneumophila; Escherichia spp, including enterotoxic E. coli (for example colonization factors, heat-labile toxin or derivatives thereof, heat-stable toxin or derivatives thereof), enterohemorragic E. coli , enteropathogenic E. coli (for example shiga toxin-like toxin or derivatives thereof); Vibrio spp, including V.
  • M. tuberculosis for example ESAT6, Antigen 85A, -B or -C
  • M. bovis for example ESAT6, Antigen 85A, -B or -C
  • M. bovis for example ESAT6, Antigen 85A, -B or -
  • cholera for example cholera toxin or derivatives thereof
  • Shigella spp including S. sonnei, S. dysenteriae, S. flexnerii
  • Yersinia spp including Y. enterocolitica (for example a Yop protein), Y. pestis, Y. pseudotuberculosis
  • Campylobacter spp including C. jejuni (for example toxins, adhesins and invasins) and C. coli
  • Salmonella spp including S. typhi, S. paratyphi, S. choleraesuis, S. enteritidis
  • Listeria spp. including L.
  • H. pylori for example urease, catalase, vacuolating toxin
  • Pseudomonas spp including P. aeruginosa
  • Staphylococcus spp. including S. aureus, S. epidermidis
  • Enterococcus spp. including E. faecalis, E. faecium
  • Clostridium spp. including C. tetani (for example tetanus toxin and derivative thereof), C. botulinum (for example botulinum toxin and derivative thereof), C.
  • Bacillus spp. including B. anthracis (for example botulinum toxin and derivatives thereof); Corynebacterium spp., including C. diphtheriae (for example diphtheria toxin and derivatives thereof); Borrelia spp., including B. burgdorferi (for example OspA, OspC, DbpA, DbpB), B. garinii (for example OspA, OspC, DbpA, DbpB), B. afzelii (for example OspA, OspC, DbpA, DbpB), B.
  • B. burgdorferi for example OspA, OspC, DbpA, DbpB
  • B. garinii for example OspA, OspC, DbpA, DbpB
  • B. afzelii for example OspA, OspC, DbpA, DbpB
  • pallidum for example the rare outer membrane proteins
  • T. denticola for example the rare outer membrane proteins
  • T. hyodysenteriae or derived from parasites such as Plasmodium spp., including P. falciparum; Toxoplasma spp., including T. gondii (for example SAG2, SAG3, Tg34); Entamoeba spp., including E. histolytica; Babesia spp., including B. microti; Trypanosoma spp., including T. cruzi; Giardia spp., including G. lamblia; Leshmania spp., including L. major; Pneumocystis spp., including P.
  • Plasmodium spp. including P. falciparum
  • Toxoplasma spp. including T. gondii (for example SAG2, SAG3, Tg34)
  • Entamoeba spp. including E. his
  • Trichomonas spp. including T. vaginalis
  • Schisostoma spp. including S. mansoni
  • yeast such as Candida spp., including C. albicans
  • Cryptococcus spp. including C. neoformans.
  • compositions of the present invention may be used for the prophylaxis or therapy of allergy.
  • Such vaccines would comprise allergen specific and allergen non-specific antigens.
  • M. tuberculosis are for example Tb Ra12, Tb H9, Tb Ra35, Tb38-1, Erd 14, DPV, MTI, MSL, mTTC2 and hTCC1 (WO 99/51748).
  • Proteins for M. tuberculosis also include fusion proteins and variants thereof where at least two, preferably three polypeptides of M. tuberculosis are fused into a larger protein.
  • Preferred fusions include Ra12-TbH9-Ra35, Erd14-DPV-MTI, DPV-MTI-MSL, Erd14-DPV-MTI-MSL-mTCC2, Erd14-DPV-MTI-MSL, DPV-MTI-MSL-mTCC2, TbH9-DPV-MTI (WO 99/51748).
  • Chlamydia antigens for Chlamydia include for example the High Molecular Weight Protein (HMW) (WO 99/17741), ORF3 (EP 366 412), and putative membrane proteins (Pmps).
  • HMW High Molecular Weight Protein
  • ORF3 ORF3
  • Pmps putative membrane proteins
  • Other Chlamydia antigens of the vaccine formulation can be selected from the group described in WO 99/28475.
  • Preferred bacterial vaccines comprise antigens derived from Streptococcus spp, including S. pneumoniae (for example, PsaA, PspA, streptolysin, choline-binding proteins) and the protein antigen Pneumolysin (Biochem Biophys Acta, 1989, 67, 1007; Rubins et al., Microbial Pathogenesis, 25, 337-342), and mutant detoxified derivatives thereof (WO 90/06951; WO 99/03884).
  • Other preferred bacterial vaccines comprise antigens derived from Haemophilus spp., including H. influenzae type B, non typeable H.
  • influenzae for example OMP26, high molecular weight adhesins, P5, P6, protein D and lipoprotein D, and fimbrin and fimbrin derived peptides (U.S. Pat. No. 5,843,464) or multiple copy variants or fusion proteins thereof.
  • the vaccine formulation of the invention comprises the HIV-1 antigen, gp120, especially when expressed in CHO cells.
  • the vaccine formulation of the invention comprises gD2t as hereinabove defined.
  • vaccines containing the claimed adjuvant comprise antigen derived from the Human Papilloma Virus (HPV) considered to be responsible for genital warts (HPV 6 or HPV 11 and others), and the HPV viruses responsible for cervical cancer (HPV16, HPV18 and others).
  • HPV Human Papilloma Virus
  • Particularly preferred forms of genital wart prophylactic, or therapeutic, vaccine comprise L1 protein, and fusion proteins comprising one or more antigens selected from the HPV proteins E1, E2, E5, E6, E7, L1, and L2.
  • fusion protein L2E7 as disclosed in WO 96/26277, and protein D(1/3)-E7 disclosed in WO99/10375.
  • a preferred HPV cervical infection or cancer, prophylaxis or therapeutic vaccine, composition may comprise HPV 16 or 18 antigens.
  • HPV 16 antigens comprise the early proteins E6 or E7 in fusion with a protein D carrier to form Protein D-E6 or E7 fusions from HPV 16, or combinations thereof; or combinations of E6 or E7 with L2 (WO 96/26277).
  • HPV 16 or 18 early proteins E6 and E7 may be presented in a single molecule, preferably a Protein D-E6/E7 fusion.
  • Such vaccine may optionally contain either or both E6 and E7 proteins from HPV 18, preferably in the form of a Protein D-E6 or Protein D-E7 fusion protein or Protein D E6/E7 fusion protein.
  • the vaccine of the present invention may additionally comprise antigens from other HPV strains, preferably from strains HPV 31 or 33.
  • Vaccines or immunogenic compositions of the present invention further comprise antigens derived from parasites that cause Malaria, for example, antigens from Plasmodia falciparum including circumsporozoite protein (CS protein), RTS,S, MSP1, MSP3, LSA1, LSA3, AMA1 and TRAP.
  • RTS is a hybrid protein comprising substantially all the C-terminal portion of the circumsporozoite (CS) protein of P. falciparum linked via four amino acids of the preS2 portion of Hepatitis B surface antigen to the surface (S) antigen of hepatitis B virus. Its full structure is disclosed in International Patent Application No. PCT/EP92/02591, published under Number WO 93/10152 claiming priority from UK patent application No.
  • One embodiment of the present invention is a malaria vaccine wherein the antigen preparation comprises RTS,S or CS protein or a fragment thereof such as the CS portion of RTS,S, in combination with one or more further malarial antigens, either or both of which may be attached to the Shiga toxin B subunit in accordance with the invention.
  • the one or more further malarial antigens may be selected for example from the group consisting of MPS1, MSP3, AMA1, LSA1 or LSA3.
  • the formulations may also contain an anti-tumour antigen and be useful for the immunotherapeutic treatment of cancers.
  • the adjuvant formulation finds utility with tumour rejection antigens such as those for prostrate, breast, colorectal, lung, pancreatic, renal or melanoma cancers.
  • Exemplary antigens include MAGE 1 and MAGE 3 or other MAGE antigens (for the treatment of melanoma), PRAME, BAGE, or GAGE (Robbins and Kawakami, 1996, Current Opinions in Immunology 8, pps 628-636; Van den Eynde et al., International Journal of Clinical & Laboratory Research (submitted 1997); Correale et al. (1997), Journal of the National Cancer Institute 89, p293.
  • tumour-specific antigens are suitable for use with the adjuvants of the present invention and include, but are not restricted to tumour-specific gangliosides, Prostate specific antigen (PSA) or Her-2/neu, KSA (GA733), PAP, mammaglobin, MUC-1, carcinoembryonic antigen (CEA), p501S (prostein).
  • PSA Prostate specific antigen
  • KSA Her-2/neu
  • PAP mammaglobin
  • MUC-1 mammaglobin
  • CEA carcinoembryonic antigen
  • p501S prostein
  • a vaccine comprising an adjuvant composition according to the invention and a tumour rejection antigen.
  • the tumour antigen is Her-2/neu.
  • the vaccines comprise a tumour antigen such as prostrate, breast, colorectal, lung, pancreatic, renal, ovarian or melanoma cancers.
  • the formulations may contain tumour-associated antigen, as well as antigens associated with tumour-support mechanisms (e.g. angiogenesis, tumour invasion).
  • antigens particularly relevant for vaccines in the therapy of cancer also comprise Prostate-specific membrane antigen (PSMA), Prostate Stem Cell Antigen (PSCA), p501S (prostein), tyrosinase, survivin, NY-ESO1, prostase, PS108 (WO 98/50567), RAGE, LAGE, HAGE.
  • said antigen may be a self peptide hormone such as whole length Gonadotrophin hormone releasing hormone (GnRH, WO 95/20600), a short 10 amino acid long peptide, useful in the treatment of many cancers, or in immunocastration.
  • the vaccine preparations containing immunogenic compositions of the present invention may be used to protect or treat a mammal susceptible to infection, by means of administering said vaccine via systemic or mucosal route. These administrations may include injection via the intramuscular, intraperitoneal, intradermal or subcutaneous routes; or via mucosal administration to the oral/alimentary, respiratory, genitourinary tracts.
  • the vaccine of the invention may be administered as a single dose, components thereof may also be co-administered together at the same time or at different times (for instance pneumococcal saccharide conjugates could be administered separately, at the same time or 1-2 weeks after the administration of the any bacterial protein component of the vaccine for optimal coordination of the immune responses with respect to each other).
  • the vaccines of the invention may be administered IM for priming doses and IN for booster doses.
  • the content of protein antigens in the vaccine will typically be in the range 1-100 ⁇ g, preferably 5-50 ⁇ g, most typically in the range 5-25 ⁇ g. Following an initial vaccination, subjects may receive one or several booster immunizations adequately spaced.
  • Vaccine preparation is generally described in Vaccine Design (“The subunit and adjuvant approach” (eds Powell M. F. & Newman M. J.) (1995) Plenum Press New York). Encapsulation within liposomes is described by Fullerton, U.S. Pat. No. 4,235,877.
  • the vaccines of the present invention may be stored in solution or lyophilized.
  • the solution is lyophilized in the presence of a sugar such as sucrose or lactose. It is still further preferable that they are lyophilized and extemporaneously reconstituted prior to use.
  • a vaccine kit comprising a vial containing an immunogenic composition of the invention, optionally in lyophilised form, and further comprising a vial containing an adjuvant as described herein. It is envisioned that in this aspect of the invention, the adjuvant will be used to reconstitute the lyophilised immunogenic composition.
  • the vaccines of the present invention may be administered by any route, administration of the described vaccines into the skin (ID) forms one embodiment of the present invention.
  • Human skin comprises an outer “horny” cuticle, called the stratum corneum, which overlays the epidermis. Underneath this epidermis is a layer called the dermis, which in turn overlays the subcutaneous tissue.
  • the dermis which in turn overlays the subcutaneous tissue.
  • Intradermal vaccination with the vaccines described herein forms a preferred feature of the present invention.
  • the conventional technique of intradermal injection comprises steps of cleaning the skin, and then stretching with one hand, and with the bevel of a narrow gauge needle (26-31 gauge) facing upwards the needle is inserted at an angle of between 10-15°.
  • the barrel of the needle is lowered and further advanced whilst providing a slight pressure to elevate it under the skin.
  • the liquid is then injected very slowly thereby forming a bleb or bump on the skin surface, followed by slow withdrawal of the needle.
  • Alternative methods of intradermal administration of the vaccine preparations may include conventional syringes and needles, or devices designed for ballistic delivery of solid vaccines (WO 99/27961), or transdermal patches (WO 97/48440; WO 98/28037); or applied to the surface of the skin (transdermal or transcutaneous delivery WO 98/20734; WO 98/28037).
  • the vaccine is in a low liquid volume, particularly a volume of between about 0.05 ml and 0.2 ml.
  • the content of antigens in the skin or intradermal vaccines of the present invention may be similar to conventional doses as found in intramuscular vaccines (see above). However, it is a feature of skin or intradermal vaccines that the formulations may be “low dose”. Accordingly the protein antigens in “low dose” vaccines are preferably present in as little as 0.1 to 10 ⁇ g, preferably 0.1 to 5 ⁇ g per dose; and the saccharide (preferably conjugated) antigens may be present in the range of 0.01-1 ⁇ g, and preferably between 0.01 to 0.5 ⁇ g of saccharide per dose.
  • the term “intradermal delivery” means delivery of the vaccine to the region of the dermis in the skin.
  • the vaccine will not necessarily be located exclusively in the dermis.
  • the dermis is the layer in the skin located between about 1.0 and about 2.0 mm from the surface in human skin, but there is a certain amount of variation between individuals and in different parts of the body. In general, it can be expected to reach the dermis by going 1.5 mm below the surface of the skin.
  • the dermis is located between the stratum corneum and the epidermis at the surface and the subcutaneous layer below.
  • the vaccine may ultimately be located solely or primarily within the dermis, or it may ultimately be distributed within the epidermis and the dermis.
  • the amount of each antigen in each vaccine dose is selected as an amount which induces an immunoprotective response without significant, adverse side effects in typical vaccinees. Such amount will vary depending upon which specific immunogen is employed and how it is presented.
  • Also provided is a method to prevent an individual from contracting a disease selected from the group comprising infectious bacterial and viral diseases, parasitic diseases, particularly intracellular pathogenic disease, proliferative diseases such as prostate, breast, colorectal, lung, pancreatic, renal, ovarian or melanoma cancers; non-cancer chronic disorders, allergy comprising the administration of a composition as substantially described herein to said individual.
  • a vaccine composition for use in the prophylactic therapy or therapy of a condition or disease wherein the vaccine composition comprises an antigen or antigen composition and an adjuvant composition consisting of an oil in water emulsion comprising 0.5-10 mg metabolisable oil, 0.5-11 mg tocol and 0.1-4 mg emulsifying agent, per human dose.
  • a vaccine composition in the manufacture of a medicament for use in prophylactic therapy or therapy of a condition or disease wherein the vaccine composition comprises an antigen or antigen composition and an adjuvant composition consisting of an oil in water emulsion comprising 0.5-10 mg metabolisable oil, 0.5-11 mg tocol and 0.1-4 mg emulsifying agent, per human dose.
  • Example I describes immunological read-out methods used in mice, ferrets, pigs and human studies.
  • Example II describes the preparation of the oil in water emulsion and adjuvant formulations used in the studies exemplified.
  • Example III shows a clinical trial in an adult population aged 18-59 years with a vaccine containing a split influenza antigen preparation and various doses of AS03 adjuvant
  • Example IV shows a preclinical evaluation of adjuvanted and non-adjuvanted split influenza vaccines (comprising various doses of AS03 adjuvant) in primed BALB/c mice
  • Example V shows a preclinical evaluation of adjuvanted and non-adjuvanted split influenza vaccines (comprising various doses of AS03 adjuvant) in primed C57Bl/6 mice
  • Example VI shows a preclinical evaluation of adjuvanted and non-adjuvanted split influenza vaccines (comprising various doses of AS03 adjuvant and low dose antigen) in primed C57Bl/6 mice
  • Example VII shows a preclinical evaluation of adjuvanted and non-adjuvanted split H5N1 vaccines (comprising various doses of AS03 adjuvant and antigen) in na ⁇ ve C57Bl/6 mice
  • Example VIII shows a preclinical evaluation of adjuvanted and non-adjuvanted influenza vaccines in primed Large White pigs
  • Anti-Hemagglutinin antibody titers to the three (seasonal) influenza virus strains are determined using the hemagglutination inhibition test (HI).
  • the principle of the HI test is based on the ability of specific anti-Influenza antibodies to inhibit hemagglutination of red blood cells (RBC) by influenza virus hemagglutinin (HA). Heat inactivated sera are treated by Kaolin and RBC to remove non-specific inhibitors. After pretreatment, two-fold dilutions of sera are incubated with 4 hemagglutination units of each influenza strain. Red blood cells are then added and the inhibition of agglutination is scored.
  • the titers are expressed as the reciprocal of the highest dilution of serum that completely inhibited hemagglutination. As the first dilution of sera is 1:20, an undetectable level is scored as a titer equal to 10.
  • Erythrocytes of horses are used for the H5N1 Pandemic strains. 0.5% (end concentration) horse red blood cell suspension in phosphate buffer containing 0.5% BSA (bovine serum albumin, end concentration). This suspension is prepared every day by washing red blood cell with the same phosphate buffer and a subsequent centrifugation step (10 min, 2000 rpm). This washing step has to be repeated once. After the addition of the horse red blood cells to the reaction mix of sera and virus suspension; the plates have to be incubated at room temperature (RT, 20° C. +/ ⁇ 2° C.) for two hours due to the low sedimentation rate of the horse red blood cells.
  • RT room temperature
  • effector T cells and/or effector-memory T cells produce IFN- ⁇ and/or central memory T cells produce IL-2.
  • PBMCs are harvested at day 7 post-immunization.
  • Lymphoid cells are re-stimulated in vitro in the presence of secretion inhibitor (Brefeldine). These cells are then processed by conventional immunofluorescent procedure using fluorescent antibodies (CD4, CD8, IFN- ⁇ and IL-2). Results are expressed as a frequency of cytokine positive cell within CD4/CD8 T cells. Intracellular staining of cytokines of T cells was performed on PBMC 7 days after the second immunization. Blood was collected from mice and pooled in heparinated medium RPMI+Add. For blood, RPMI+Add-diluted PBL suspensions were layered onto a Lympholyte-Mammal gradient according to the recommended protocol (centrifuge 20 min at 2500 rpm and R.T.). The mononuclear cells at the interface were removed, washed 2 ⁇ in RPMI+Add and PBMCs suspensions were adjusted to 2 ⁇ 10 6 cells/ml in RPMI 5% fetal calf serum.
  • PBMC are incubated overnight at 37° C. in presence of Brefeldin (1 ⁇ g/ml) at 37° C. to inhibit cytokine secretion.
  • IFN- ⁇ /IL-2/CD4/CD8 staining was performed as follows: Cell suspensions were washed, resuspended in 50 ⁇ l of PBS 1% FCS containing 2% Fc blocking reagent (1/50; 2.4G2). After 10 min incubation at 4° C., 50 ⁇ l of a mixture of anti-CD4-PE (2/50) and anti-CD8 perCp (3/50) was added and incubated 30 min at 4° C. After a washing in PBS 1% FCS, cells were permeabilized by resuspending in 200 ⁇ l of Cytofix-Cytoperm (Kit BD) and incubated 20 min at 4° C.
  • Cytofix-Cytoperm Kerat BD
  • split H5N1 Quantitation of anti-H5N1 Ig, IgG1 and IgG2b antibody titers was performed by ELISA using split H5N1 as coating. Virus and antibody solutions were used at 100 ⁇ l per well. Split virus H5N1 was diluted at a final concentration of 1 ⁇ g/ml in PBS and was adsorbed overnight at 4° C. to the wells of 96 wells microtiter plates (Maxisorb Immunoplate Nunc 439454). The plates were then incubated for 1 hour at 37° C. with 200 ⁇ l per well of PBS containing 1% BSA and 0.1% Tween 20 (saturation buffer).
  • Anti-Hemagglutinin antibody titers to the three influenza virus strains were determined using the hemagglutination inhibition test (HI).
  • the principle of the HI test is based on the ability of specific anti-Influenza antibodies to inhibit hemagglutination of chicken red blood cells (RBC) by influenza virus hemagglutinin (HA).
  • Sera were first treated with a 25% neuraminidase solution (RDE) and were heat-inactivated to remove non-specific inhibitors. After pre-treatment, two-fold dilutions of sera were incubated with 4 hemagglutination units of each influenza strain. Chicken red blood cells were then added and the inhibition of agglutination was scored. The titers were expressed as the reciprocal of the highest dilution of serum that completely inhibited hemagglutination. As the first dilution of sera was 1:10, an undetectable level was scored as a titer equal to 5.
  • the nasal washes were performed by administration of 5 ml of PBS in both nostrils in awoke animals.
  • the inoculum was collected in a Petri dish and placed into sample containers on dry ice.
  • the culture medium is gently removed and 100 ⁇ l of a 1/20 WST-1 containing medium is added and incubated for another 18 hrs.
  • the intensity of the yellow formazan dye produced upon reduction of WST-1 by viable cells is proportional to the number of viable cells present in the well at the end of the viral titration assay and is quantified by measuring the absorbance of each well at the appropriate wavelength (450 nanometers).
  • the cut-off is defined as the OD average of uninfected control cells-0.3 OD (0.3 OD correspond to +/ ⁇ 3 StDev of OD of uninfected control cells).
  • a positive score is defined when OD is ⁇ cut-off and in contrast a negative score is defined when OD is >cut-off.
  • Viral shedding titers were determined by “Reed and Muench” and expressed as Log TCID50/ml.
  • Anti-Hemagglutinin antibody titers to the three influenza virus strains were determined using the hemagglutination inhibition test (HI).
  • the principle of the HI test is based on the ability of specific anti-Influenza antibodies to inhibit hemagglutination of chicken red blood cells (RBC) by influenza virus hemagglutinin (HA).
  • Sera were first treated with a 25% neuraminidase solution (RDE) and were heat-inactivated to remove non-specific inhibitors. After pre-treatment, two-fold dilutions of sera were incubated with 4 hemagglutination units of each influenza strain. Chicken red blood cells were then added and the inhibition of agglutination was scored. The titers were expressed as the reciprocal of the highest dilution of serum that completely inhibited hemagglutination. As the first dilution of sera was 1:10, an undetectable level was scored as a titer equal to 5.
  • the immune response was determined by measuring HI antibodies using the method described by the WHO Collaborating Centre for influenza, Centres for Disease Control, Atlanta, USA (1991).
  • Antibody titre measurements were conducted on thawed frozen serum samples with a standardised and comprehensively validated micromethod using 4 hemagglutination-inhibiting units (4 HIU) of the appropriate antigens and a 0.5% fowl erythrocyte suspension. Non-specific serum inhibitors were removed by heat treatment and receptor-destroying enzyme.
  • the sera obtained were evaluated for HI antibody levels.
  • a dilution series (by a factor of 2) was prepared up to an end dilution of 1:20480.
  • the titration end-point was taken as the highest dilution step that showed complete inhibition (100%) of hemagglutination. All assays were performed in duplicate.
  • the assay was performed in fetuin-coated microtitre plates.
  • a 2-fold dilution series of the antiserum was prepared and mixed with a standardised amount of influenza A H3N2, H1N1 or influenza B virus.
  • the test was based on the biological activity of the neuraminidase which enzymatically releases neuraminic acid from fetuin. After cleavage of the terminal neuraminic acid ⁇ -D-glactose-N-acetyl-galactosamin was unmasked.
  • HRP horseradish peroxidase
  • Virus neutralisation by antibodies contained in the serum was determined in a microneutralization assay. The sera were used without further treatment in the assay. Each serum was tested in triplicate. A standardised amount of virus was mixed with serial dilutions of serum and incubated to allow binding of the antibodies to the virus. A cell suspension, containing a defined amount of MDCK cells was then added to the mixture of virus and antiserum and incubated at 33° C. After the incubation period, virus replication was visualised by hemagglutination of chicken red blood cells. The 50% neutralisation titre of a serum was calculated by the method of Reed and Muench.
  • Peripheral blood antigen-specific CD4 and CD8 T cells can be restimulated in vitro to produce IL-2, CD40L, TNF-alpha and IFN if incubated with their corresponding antigen. Consequently, antigen-specific CD4 and CD8 T cells can be enumerated by flow cytometry following conventional immunofluorescence labelling of cellular phenotype as well as intracellular cytokines production.
  • Influenza vaccine antigen as well as peptides derived from specific influenza protein were used as antigen to restimulate Influenza-specific T cells. Results were expressed as a frequency of cytokine(s)-positive CD4 or CD8 T cell within the CD4 or CD8 T cell sub-population.
  • reactogenicity/safety may be secondary endpoints, and immunogenicity may be the primary endpoint.
  • CMI Cell Mediated Immune
  • the immunogenicity analysis was based on the total vaccinated cohort. For each treatment group, the following parameters (with 95% confidence intervals) were calculated:
  • the oil/water emulsion used in the subsequent examples is composed an organic phase made of 2 oils (alpha-tocopherol and squalene), and an aqueous phase of PBS containing Tween 80 as emulsifying agent.
  • the oil in water emulsion adjuvant formulations used in the subsequent examples were made comprising the following oil in water emulsion component (final concentrations given): 2.5% squalene (v/v), 2.5% alpha-tocopherol (v/v), 0.9% polyoxyethylene sorbitan monooleate (v/v) (Tween 80), see WO 95/17210.
  • the preparation of the SB62 emulsion is made by mixing under strong agitation of an oil phase composed of hydrophobic components (DL- ⁇ -tocopherol and squalene) and an aqueous phase containing the water soluble components (the anionic detergent Tween 80 and PBS mod (modified), pH 6.8). While stirring, the oil phase (1/10 total volume) is transferred to the aqueous phase (9/10 total volume), and the mixture is stirred for 15 minutes at room temperature.
  • an oil phase composed of hydrophobic components (DL- ⁇ -tocopherol and squalene) and an aqueous phase containing the water soluble components (the anionic detergent Tween 80 and PBS mod (modified), pH 6.8). While stirring, the oil phase (1/10 total volume) is transferred to the aqueous phase (9/10 total volume), and the mixture is stirred for 15 minutes at room temperature.
  • the resulting mixture then subjected to shear, impact and cavitation forces in the interaction chamber of a microfluidizer (15000 PSI-8 cycles, or 3 cycles in the adjuvant used in the clinical trial reported in Example III) to produce submicron droplets (distribution between 100 and 200 nm).
  • the resulting pH is between 6.8 ⁇ 0.1.
  • the SB62 emulsion is then sterilised by filtration through a 0.22 ⁇ m membrane and the sterile bulk emulsion is stored refrigerated in Cupac containers at 2 to 8° C. Sterile inert gas (nitrogen or argon) is flushed into the dead volume of the SB62 emulsion final bulk container for at least 15 seconds.
  • the final composition of the SB62 emulsion is as follows:
  • Tween 80 1.8% (v/v) 19.4 mg/ml; Squalene: 5% (v/v) 42.8 mg/ml; ⁇ -tocopherol: 5% (v/v) 47.5 mg/ml; PBS-mod: NaCl 121 mM, KCl 2.38 mM, Na2HPO4 7.14 mM, KH2PO4 1.3 mM; pH 6.8 ⁇ 0.1.
  • a phase II, controlled, randomized, single blind study was conducted in an adult population aged 18-59 years old in 2006 in order to evaluate the immunogenicity, safety and reactogenicity of the GlaxoSmithKline Biologicals low dose influenza candidate vaccine (i.e. containing 5 ⁇ g HA per strain) with two doses of AS03 adjuvant.
  • the humoral immune response i.e. anti-hemagglutinin
  • FluarixTM was used as reference.
  • the standard trivalent split influenza vaccine—FluarixTM used in this study, is a commercial vaccine from the year 2006 developed and manufactured by GlaxoSmithKline Biologicals.
  • the non-adjuvanted influenza vaccine is a trivalent split virion, inactivated influenza vaccine consisting of three monovalent viral antigen bulks (prepared from respectively influenza strains A/H1N1, A/H3N2 and B).
  • the antigens present in this vaccine are the same as in the licensed FluarixTM vaccine which is available on the market as FluarixTM ( ⁇ -Rix®) since 1992 and contain 15 ⁇ g HA/strain per dose.
  • the influenza strains included in the FluLD clinical lots are the strains that were chosen for the 2006/2007 Northern Hemisphere:
  • the antigens are derived from egg-grown viruses. Splitting is carried out with sodium deoxycholate prior to the inactivation step, which is performed through the subsequent action of sodium deoxycholate and formaldehyde.
  • the AS03 adjuvanted low dose influenza (FluLD) vaccine (clinical lots) is based on the commercially available FluarixTM vaccine (prepared from respectively influenza strains A/H1N1, A/H3N2 and B), but with a lower antigen content and adjuvanted with GSK adjuvant system AS03.
  • AS03 consists of an oil-in-water emulsion (SB62) that contains two biodegradable oils, squalene and ⁇ -tocopherol (Vitamin E), and a surfactant, polysorbate 80 (Tween 80). Influenza antigens are incorporated in the aqueous phase of the adjuvant system by simple mixing with the emulsion.
  • the adjuvanted vaccines contain 5 ⁇ g haemagglutinin (HA) of each influenza virus strain per dose, combined with a full dose (AS03) or half a dose (AS03 1 ⁇ 2) of the adjuvant system AS03.
  • the excipients are the following: polysorbate 80 (Tween 80), octoxynol 10 (Triton X-100), alpha-tocopheryl hydrogen succinate, sodium chloride, disodium hydrogen phosphate, potassium dihydrogen phosphate, potassium chloride, water for injection.
  • the AS03 adjuvanted low dose influenza vaccines (FluLD, full or half dose of AS03) are preservative-free vaccines. However, they contain trace amounts of thiomersal ( ⁇ 1.25 ⁇ g of Hg per dose) from the early stages of the manufacturing process. They are both presented as monodose vaccines in glass (Type I) pre-filled syringes at a volume of 0.5 ml/dose.
  • FluLD full or half dose of AS03
  • the composition is provided in Table 3.
  • the HA content per dose is 5 ⁇ g for both formulations, the sole difference being the amount of AS03 present in the final containers.
  • the influenza antigens are identical to those included in FluarixTM (Influenza Virus Vaccine).
  • the monovalent bulks consist of purified inactivated split viruses that are prepared from working seeds of the three strains of influenza virus, type A (H1N1 and H3N2) and type B, which are grown individually in embryonated hens' eggs. These working seeds are derived from strains that are received from a WHO collaborating center following the annual WHO recommendations.
  • the volumes of the three monovalent bulks are based on the HA content measured in each monovalent bulk prior to the formulation and on the target manufacturing volume.
  • a 10-times concentrated phosphate buffered saline (pH 7.4 when 1 time concentrated) and a pre-mixture of Tween 80 and ⁇ -tocopheryl hydrogen succinate are diluted in water for injection, followed by stirring during 5-30 minutes at room temperature.
  • the three concentrated monovalent bulks are then successively diluted in the resulting phosphate buffered saline/Tween 80- ⁇ -tocopheryl hydrogen succinate solution to a concentration of
  • pre-pool can be held at +2-+8° C. or further processed to the final formulation step on the same day.
  • the final volume of pre-pool is 250 ⁇ l per dose.
  • Adjuvanted Vaccine LD AS03 1/1 (Table 4)
  • PBS mod 10 fold concentrated pH 7.4 when one fold concentrated; 137 mM NaCl, 2.7 mM KCl, 8.1 mM Na 2 HPO 4 , 1.47 mM KH 2 PO 4 , pH 7.4) as well as a mixture containing Tween80, Triton X-100 and VES (quantities taking into account the detergent present in the strains) are added to water for injection. After 5 to 30 minutes stirring, 20 ⁇ g HA per ml of each strain H1N1 and H3N2 and 23.32 ⁇ g HA per ml of B strain are added with 10 to 30 minutes stirring between each addition.
  • intermediate bulk After 15 to 30 minutes stirring, a small volume of the so called “intermediate bulk” are discarded for analysis and stored between +2 and +8° C.
  • the intermediate bulk is in PBS mod 1 fold concentrated.
  • the target's detergents concentration are 488 ⁇ g Tween 80 per ml, 73.6 ⁇ g Triton X-100 per ml and 66.6 ⁇ g VES per ml.
  • the final formulation is then prepared: an equal volume of SB62 (see preparation in Example II) is added to each 250 ⁇ l of pre-pool intermediate bulk and mixed during 30 to 60 minutes at room temperature. pH is checked to range between 6.8 and 7.5. Formulation is flushed with nitrogen and then stored between +2 and 8° C. prior to filling.
  • PBS mod 10 fold concentrated (pH 7.4 when one fold concentrated—see composition above) as well as a mixture containing Tween 80, Triton X-100 and VES (quantities taking into account the detergent present in the strains) are added to water for injection. After 5 to 30 minutes stirring, 20 ⁇ g HA per ml of each strain H1N1 and H3N2 and 23.32 ⁇ g HA per ml of B strain are added with 10 to 30 minutes stirring between each addition. After 15 to 30 minutes stirring, a small volume of the so called “intermediate bulk” are discarded for analysis and stored between +2 and +8° C. PBS mod is 1 fold concentrated in the intermediate bulk. The target's detergents concentration are 488 ⁇ g Tween 80 per ml, 73.6 ⁇ g Triton X-100 per ml and 66.6 ⁇ g VES per ml
  • SB62 is first diluted with the PBS mod buffer and stirred for 15-30 minutes at RT. An equal volume of this diluted SB62 is then added to each 250 ⁇ l of pre-pool of intermediate bulk. After 30 to 60 minutes stirring at RT, pH is checked to range between 6.8 and 7.5. Formulation is flushed with nitrogen and then stored between +2 and 8° C. prior to filling.
  • the final volume of both formulation is 500 ⁇ l per dose and the final HA concentration is 10 ⁇ g of each A monovalent bulk and 11.66 ⁇ g of B monovalent bulk per ml of trivalent final bulk.
  • Final Tween 80, Triton X-100 (residual from H3N2 monobulk manufacturing) and ⁇ -tocopheryl hydrogen succinate ( ⁇ -tocopheryl hydrogen succinate is an ester form of RRR (D isomer)- ⁇ -tocopherol) target concentrations are 244 ⁇ g/ml, 58.6 ⁇ g/ml and 33.3 ⁇ g/ml, respectively.
  • the vaccine is filled into 1.25-ml sterile Type I (Ph. Eur.) glass syringes. Each syringe is filled to a target of 0.57 ml (range: 0.54-0.60 ml).
  • the vaccines were administered intramuscularly in the deltoid region of the non-dominant arm. All vaccines were presented as pre-filled syringes (0.5 ml). In order to ensure proper IM injection of the vaccine, a needle of at least 25 G and at least 2.5 cm in length was used.
  • the mean age of the total vaccinated cohort at the time of vaccination was 36.7 years with a standard deviation of 13.67 years.
  • the GMTs for HI antibodies with 95% CI are shown in Table 10 and FIG. 1 . Adjusted GMT ratios between groups are shown in Table 11.
  • Pre-vaccination GMTs of HI antibodies for all 3 vaccine strains were within the same range in the 3 treatment groups.
  • the observed GMTs at day 21 for adjuvanted groups tends to be higher than Fluarix group for all 3 strains with a statistical difference (no overlapping of 95% CIs and adjusted GMT ratio did not contain the value 1) between FluLD1/1 and Fluarix for the A/Wisconsin vaccine strain.
  • the SCR at day 21 for the FluLD1/1 group tended to be higher compared to the Fluarix group.
  • the SCR at day 21 for the FluLD1/2 group was within the same range compared to the Fluarix group.
  • the SCR at day 21 for the FluLD1/2 group tended to be higher compared to the Fluarix group.
  • the SCR at day 21 for the FluLD1/1 group was within the same range compared to the Fluarix group.
  • the SCF at day 21 for the FluLD1/1 group tended to be higher compared to the Fluarix group.
  • the SCF at day 21 for the FluLD1/2 group was within the same range compared to Fluarix group.
  • the SCF at day 21 for the two adjuvanted groups tended to be higher compared to the Fluarix group.
  • a reduction of the AS03 content in the adjuvanted vaccine has a significant impact on all the general and on the local grade 3 symptoms.
  • the primary objective of this study was to assess humoral immune response (anti-HI antibody titres) elicited by low dose influenza vaccine with two different concentrations of AS03 adjuvant, and by Fluarix.
  • the administration of the low dose influenza candidate vaccine adjuvanted with AS03 was safe and clinically well tolerated in the study population, i.e. adult people aged between 18 and 59 years.
  • the half dose adjuvanted vaccine showed a marked decrease in the incidence of solicited local and general symptoms, compared to the full dose adjuvanted vaccine.
  • influenza-primed mice were performed in order to evaluate the increase in humoral responses by AS03 induced by influenza vaccines formulated with this oil-in-water adjuvant. To simulate the human situation, an experiment was conducted using mice primed with heterosubtypic strains.
  • mice Groups of 27 adult female BALB/c mice were primed intranasally (20 ⁇ l volume) on day 0 with trivalent whole, formalin-inactivated influenza virus (5 ⁇ g HA for each strain). Priming strains consisted of earlier drift variants (5 ⁇ g HA whole inactivated H1N1 A/Johannesburg/82/96, H3N2 A/Sydney/5/97, B/Harbin/7/94) to those included in the vaccine. Twenty-eight days later, the mice were vaccinated with a single dose of the vaccine candidate intramuscularly in a total volume of 50 ⁇ l.
  • mice were immunized with formulations containing split antigens alone (trivalent split plain) or formulations containing split antigens adjuvanted with two doses of AS03 (full or 1 ⁇ 5).
  • the strains used for the immunizations included H1N1 A/New Calcdonia/20/99, H3N2 A/Panama/2007/99, B/Shangdong/7/97 viral antigens (1.5 ⁇ g/strain, 1/10 th of the human dose).
  • a Premix of Tween 80, Triton X100 and Vitamin E Succinate (VES) is prepared in order to reach a final concentration into the vaccine of 750 ⁇ g/ml of Tween 80, 110 ⁇ g/ml of Triton X100 and 100 ⁇ g/ml of VES.
  • the quantities used in the premix are calculated taking into account the quantities of detergent and VES already present in the strains.
  • the formulation of one 50 ⁇ l dose is prepared extemporaneously according the following sequence: Water For Injection+Saline Buffer (10 fold concentrated PBS pH 7.4)+Premix, 5 min magnetic stirring at room temperature, +1.5 ⁇ g HA H1N1 strain, 10 min magnetic stirring at room temperature, +1.5 ⁇ g HA H3N2 strain, 10 min magnetic stirring at room temperature, +1.5 ⁇ g HA B strain, 15 min magnetic stirring at room temperature.
  • the formulations are injected within the hour following the end of their preparation.
  • a Premix of Tween 80, Triton X100 and Vitamin E Succinate (VES) is prepared in order to reach a final concentration into the vaccine of 750 ⁇ g/ml of Tween 80, 110 ⁇ g/ml of Triton X100 and 100 ⁇ g/ml of VES.
  • the quantities used in the premix are calculated taking into account the quantities of detergent and VES already present in the strains.
  • the formulation of one 50 ⁇ l dose is prepared extemporaneously according the following sequence: Water For Injection+Saline Buffer (10 fold concentrated PBS pH 7.4)+Premix, 5 min magnetic stirring at room temperature, +1.5 ⁇ g HA H1N1 strain, 10 min magnetic stirring at room temperature, +1.5 ⁇ g HA H3N2 strain, 10 min magnetic stirring at room temperature, +1.5 ⁇ gHA B strain, 15 min magnetic stirring at room temperature, +25 ⁇ l SB62 emulsion for the full dose AS03 or 5 ⁇ l SB62 emulsion for the 1 ⁇ 5 dose AS03, min magnetic stirring at room temperature.
  • the formulations are injected within the hour following the end of their preparation.
  • the humoral immune response to vaccination was measured before immunization (day 28) and 14 days after immunization (27 mice/group). Serum samples were tested by the hemagglutination inhibition (HI) test.
  • Results are presented in FIG. 5 .
  • AS03 and dilutions thereof were shown to induce higher HI titres compared to the plain vaccine.
  • For all influenza A strains a statistically significant increase of HI titres was observed (p ⁇ 0.05).
  • For the H1N1 strain a significant difference in HI titres was also observed between AS03 and AS03 1 ⁇ 5 (p ⁇ 0 . 05 ).
  • a reduced dose of AS03 failed to increase HI titres for all three strains compared to the plain vaccine. Very low responses were observed against the B strain (B/Shangdong); this is likely to be due to the significant antigenic drift between the B strains used for the priming and the vaccine.
  • mice primed with heterosubtypic strains were primed with heterosubtypic strains.
  • mice Groups of 25 adult female C57Bl/6 mice were primed intranasally (20 ⁇ l volume) on day 0 with trivalent whole, formalin-inactivated influenza virus (5 ⁇ g HA for each strain). Priming strains consisted of earlier drift variants (5 ⁇ g HA whole inactivated H1N1 A/Beijing/262/95, H3N2 A/Panama/2007/99, B/Shangdong/7/97) to those included in the vaccine. Twenty-eight days later, the mice were vaccinated with a single dose of the vaccine candidate intramuscularly in a total volume of 100 ⁇ l.
  • mice were immunized with formulations containing split antigens alone (trivalent split plain) or formulations containing split antigens adjuvanted with three doses of AS03 (full, 1 ⁇ 2 or 1 ⁇ 5).
  • the strains used for the immunizations included H1N1 A/New Calcdonia/20/99, H3N2 A/New York/55/2004, B/Jiangsu/10/2003 viral antigens (1.5 ⁇ g/strain, 1/10 th of the human dose).
  • the formulations for a 100 ⁇ l dose are prepared extemporaneously according the following sequence: Water For Injection+Saline Buffer (10 fold concentrated PBS pH 7.4 prepared as taught in example IV)+Fluarix Clinical Lot DFLUA014 (1.5 ⁇ g per strain in the final dose).
  • the formulations for a 100 ⁇ l dose are prepared extemporaneously according the following sequence: Water For Injection+Saline Buffer (10 fold concentrated PBS pH 7.4 prepared as taught in example IV)+Fluarix Clinical Lot DFLUA014 (1.5 ⁇ g per strain in the final dose)+25 ⁇ l SB62 emulsion for the full dose or 12.5 ⁇ l SB 62 emulsion for the 1 ⁇ 2 dose or 5 ⁇ l SB62 emulsion for the 1 ⁇ 5 dose.
  • the formulations are injected within the hour following the end of the preparation.
  • the humoral immune response to vaccination was measured 21 days after immunization (10 mice/group) and the serum samples were tested by the haemagglutination inhibition (HI) test.
  • the cellular immune response was tested 7 days post-immunization by intracellular cytokine staining (ICS).
  • mice immunized with AS03-adjuvanted trivalent split vaccine compared to mice immunized with trivalent split plain.
  • mice immunized with trivalent split adjuvanted with a full dose AS03 Compared to the response induced in mice immunized with trivalent split adjuvanted with a full dose AS03, a trend for lower cellular responses was observed when mice were immunized with trivalent split adjuvanted with lower doses of AS03.
  • influenza-primed mice were performed in order to evaluate the increase in cellular immune responses by AS03 induced by influenza vaccines containing low dose antigen (0.5 ⁇ g/strain, 1/30 th human dose) and formulated with this oil-in-water adjuvant. To simulate the human situation, an experiment was conducted using mice primed with heterosubtypic strains.
  • mice Groups of 15 adult female C57Bl/6 mice were primed intranasally (20 ⁇ l volume) on day 0 with trivalent whole, formalin-inactivated influenza virus (5 ⁇ g HA for each strain). Priming strains consisted of earlier drift variants (5 ⁇ g HA whole inactivated H1N1 A/Beijing/262/95, H3N2 N Panama/2007/99, B/Shangdong/7/97) to those included in the vaccine. Twenty-eight days later, the mice were vaccinated with a single dose of the vaccine candidate intramuscularly in a total volume of 50 ⁇ l.
  • mice were immunized with formulations containing split antigens alone (trivalent split plain) or formulations containing split antigens adjuvanted with three doses of AS03 (full, 1 ⁇ 2 or 1 ⁇ 5).
  • the strains used for the immunizations included H1N1 A/New Calcdonia/20/99, H3N2 A/New York/55/2004, B/Jiangsu/10/2003 viral antigens (0.5 ⁇ g/strain, 1/30 th of the human dose).
  • the formulations for a 50 ⁇ l dose are prepared extemporaneously according the following sequence: Water For Injection+Saline Buffer (10 fold concentrated PBS pH 7.4 prepared as taught in example IV)+Fluarix Clinical Lot DFLUA014 (0.5 ⁇ g per strain in the final dose).
  • the formulations for a 50 ⁇ l dose are prepared extemporaneously according the following sequence: Water For Injection+Saline Buffer (10 fold concentrated PBS pH 7.4 prepared as taught in example IV)+Fluarix Clinical Lot DFLUA014 (0.5 ⁇ g per strain in the final dose)+25 ⁇ l SB62 emulsion for the full dose or 12.5 ⁇ l SB 62 emulsion for the 1 ⁇ 2 dose or 5 ⁇ l SB62 emulsion for the 1 ⁇ 5 dose.
  • the formulations are injected within the hour following the end of the preparation.
  • the cellular immune response was tested 7 days post-immunization by intracellular cytokine staining.
  • mice immunized with trivalent split vaccine adjuvanted with AS03 full or 1 ⁇ 2 dose
  • mice immunized with trivalent split plain Compared to the response induced in mice immunized with trivalent split plain or adjuvanted with a full dose or a half dose of AS03, higher cellular responses were observed when mice were immunized with trivalent split adjuvanted with 1 ⁇ 5 of AS03 dose.
  • H5N1-naive mice were performed in order to evaluate the increase in humoral and cellular immune responses by AS03 induced by H5N1 split vaccines formulated with this oil-in-water adjuvant.
  • a pandemic it is expected that the entire world population will be immunologically naive to the newly circulating pandemic influenza strain. Due to this naive immune status a pandemic vaccine will likely require two vaccine doses to protect individuals from infection and severe illness caused by a new influenza strain. To represent this lack of previous exposure a na ⁇ ve mouse model was developed to assess vaccine immunogenicity.
  • mice Groups of 15 adult female na ⁇ ve C57Bl/6 mice were immunized on days 0 and 28 with pandemic H5N1 vaccine candidate intramuscularly in a total volume of 50 ⁇ l. Mice were immunized with formulations containing split H5N1 antigens alone (H5N1 split plain) or formulations containing split antigens adjuvanted with different doses of AS03 (double, full, 1 ⁇ 2 or 1 ⁇ 5). The strains used for the immunizations included H5N1 A/Vietnam/1194/04 viral antigen (1.5 or 0.38 ⁇ g/strain corresponding to 1/10 th of the human dose).
  • Thiomersal (quantities taking into account its concentration in the strain) and Triton X100 are added to the Final Bulk Buffer.
  • Tween 80 is not added as the content target in the formulation is reach by the Tween concentration of the strain.
  • the final concentrations are of 10 ⁇ g/ml for Thiomersal, 368 ⁇ g/ml for Tween 80 and 35 ⁇ g/ml for Triton X100 in the 1.5 ⁇ g formulation dose. They are of 10 ⁇ g/ml for Thiomersal, 93 ⁇ g/ml for Tween80 and 8.9 ⁇ g/ml for Triton X100 in the 0.38 ⁇ g formulation dose.
  • After 5-30 min magnetic stirring 1.5 or 0.38 ⁇ g of HA (H5N1 strain) are added.
  • the formulations are stirred for 30-60 minutes.
  • the pH is checked. Injections occur within the hour following the end of the formulation.
  • Thiomersal (quantities taking into account its concentration in the strain) and Triton X100 are added to the Final Bulk Buffer.
  • Tween 80 is not added as the content target in the formulation is reach by the Tween concentration of the strain.
  • the final concentrations are of 10 ⁇ g/ml for Thiomersal, 368 ⁇ g/ml for Tween 80 and 35 ⁇ g/ml for Triton X100 in the 1.5 ⁇ g formulation dose. They are of 10 ⁇ g/ml for Thiomersal, 93 ⁇ g/ml for Tween80 and 8.9 ⁇ g/ml for Triton X100 in the 0.38 ⁇ g formulation dose.
  • the humoral immune response was measured 14 days after immunization (10 mice/group) by anti-Ig, IgG1 and IgG2b antibody titers ( FIG. 9 , A-F).
  • the humoral immune response was also measured 21 days after immunization (10 mice/group) by anti-H5N1 hemagglutination inhibition assay ( FIG. 10 , A-B).
  • the cellular immune response was tested 6 days post-immunization (5 pools of 3 mice per group) by intracellular cytokine staining (ICS) of antigen-specific CD4+ T cells numerated by flow cytometry ( FIG. 11 , A-B).
  • ICS cytokine staining
  • Results are presented in FIG. 9 .
  • mice immunized with AS03-adjuvanted H5N1 split vaccine induced higher HI titers compared to the response obtained in mice immunized with the non-adjuvanted H5N1 split vaccine ( FIG. 10-A ).
  • mice immunized with AS03-adjuvanted H5N1 split vaccine induced higher HI titers compared to the response obtained in mice immunized with the non-adjuvanted H5N1 split vaccine ( FIG. 10B ).
  • mice immunized with H5N1 split vaccine adjuvanted with 2 ⁇ full dose AS03 or a full dose AS03 or between mice immunized with H5N1 split vaccine adjuvanted with AS03/2 or AS03/5 ( FIG. 10B ).
  • Results are presented in FIG. 11 .
  • mice immunized with H5N1 split vaccine at either antigen dose
  • H5N1 split vaccine adjuvanted with a full dose AS03 or with AS03/2.
  • Some enhancement in the immune response was detected when 2 ⁇ full dose AS03 was used in the vaccine formulation and accordingly a decrease in the immune response was detected when AS03/5 was used in the vaccine formulation.
  • Pigs were used in order to evaluate a dose range of AS03 in an animal model close to humans. Pigs show a long list of biological analogies that establish this animal as physiologically the closest to man with very few exceptions (Douglas R., 1972). Moreover, the manifestation of influenza infection in pigs is commonly observed.
  • Groups of 10 adult Large White female pigs were primed on day 0 with trivalent whole, formalin-inactivated influenza virus (25 ⁇ g HA for each strain) intranasally in a total volume of 200 ⁇ l.
  • Priming strains consisted of strains homologous to vaccine strains (25 ⁇ g HA whole inactivated H1N1 A/New Calcdonia/20/99, H3N2 A/Panama/2007/99 and B/Shangdong/7/97). Twenty-eight days later, pigs were vaccinated with a single dose of the vaccine candidate intramuscularly in a total volume of 500 ⁇ l.
  • Pigs were immunized with formulations containing split antigens alone (trivalent split plain) or formulations containing split antigens adjuvanted with a dose range of AS03 (full, 1 ⁇ 2 or 1 ⁇ 5).
  • the strains used for the immunizations included H1N1 A/New Calcdonia/20/99, H3N2 A/Panama/2007/99 and B/Shangdong/7/97 viral antigens (15 ⁇ g HA for H1N1 A/New Calcdonia/20/99, H3N2 A/Panama/2007/99 strains and 17.5 ⁇ g B/Shangdong/7/97 strain as in one human dose).
  • a Premix of Tween 80, Triton X100 and Vitamin E Succinate (VES) is prepared in order to reach a final concentration into the vaccine of 750 ⁇ g/ml of Tween 80, 110 ⁇ g/ml of Triton X100 and 100 ⁇ g/ml of VES.
  • the quantities used in the premix take into account their content into the strains.
  • the formulation of one 500 ⁇ l dose is prepared extemporaneously according the following sequence: Water For Injection+Saline Buffer (10 fold concentrated PBS pH 7.4 prepared as taught in example IV)+Premix, 5 min magnetic stirring at room temperature, +15 ⁇ g HA H1N1 strain, 10 min magnetic stirring at room temperature, +15 ⁇ g HA H3N2 strain, 10 min magnetic stirring at room temperature, +17.5 ⁇ g HA B strain, 15 min magnetic stirring at room temperature.
  • the formulations are injected within the hour following the end of their preparation.
  • a Premix of Tween 80, Triton X100 and Vitamin E Succinate (VES) is prepared in order to reach a final concentration into the vaccine of 750 ⁇ g/ml of Tween 80, 110 ⁇ g/ml of Triton X100 and 100 ⁇ g/ml of VES.
  • the quantities used in the premix take into account their content into the strains.
  • the formulation of one 500 ⁇ l dose is prepared extemporaneously according the following sequence: Water For Injection+Saline Buffer (10 fold concentrated PBS pH 7.4 prepared as taught in example IV)+Premix, 5 min magnetic stirring at room temperature, +15 ⁇ g HA H1N1 strain, 10 min magnetic stirring at room temperature, +15 ⁇ g HA H3N2 strain, 10 min magnetic stirring at room temperature, +17.5 ⁇ g HA B strain, 15 min magnetic stirring at room temperature, +250 ⁇ l SB62 emulsion for the full dose AS03 or 125 ⁇ l SB62 emulsion for the 1 ⁇ 2 dose AS03 or 50 ⁇ l SB62 emulsion for the 1 ⁇ 5 dose AS03, 15 min magnetic stirring at room temperature.
  • the formulations are injected within the hour following the end of their preparation.
  • the humoral immune response to vaccination was measured before intranasal priming (day 0), before immunization (day 28) and 14 days after immunization (10 pigs/group). Serum samples were tested by the haemagglutination inhibition (HI) test.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pulmonology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
US12/445,090 2006-10-12 2007-10-10 Vaccine Comprising an Oil in Water Emulsion Adjuvant Abandoned US20100183667A1 (en)

Applications Claiming Priority (19)

Application Number Priority Date Filing Date Title
GBGB0620336.8A GB0620336D0 (en) 2006-10-12 2006-10-12 Vaccine
GB0620337.6 2006-10-12
GB0620337A GB0620337D0 (en) 2006-10-12 2006-10-12 Vaccine
GB0620336.8 2006-10-12
GB0620815.1 2006-10-19
GB0620815A GB0620815D0 (en) 2006-10-19 2006-10-19 Vaccine
GB0620816.9 2006-10-19
GB0620816A GB0620816D0 (en) 2006-10-19 2006-10-19 vaccine
PCT/EP2006/069979 WO2007071711A2 (en) 2005-12-22 2006-12-20 Vaccine
PCT/EP2006/069977 WO2007071710A2 (en) 2005-12-22 2006-12-20 Vaccine comprising streptococcus pneumoniae capsular polysaccharide conjugates
EPPCTEP2006069977 2006-12-20
EPPCTEP2006069979 2006-12-20
GB0707697.9 2007-04-20
GB0707697A GB0707697D0 (en) 2007-04-20 2007-04-20 Vaccine
GB0711357A GB0711357D0 (en) 2007-06-12 2007-06-12 Vaccine
GB0711357.4 2007-06-12
GB0712062.9 2007-06-21
GB0712062A GB0712062D0 (en) 2007-06-21 2007-06-21 Vaccine
PCT/EP2007/060743 WO2008043774A1 (en) 2006-10-12 2007-10-10 Vaccine comprising an oil in water emulsion adjuvant

Publications (1)

Publication Number Publication Date
US20100183667A1 true US20100183667A1 (en) 2010-07-22

Family

ID=40551582

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/445,090 Abandoned US20100183667A1 (en) 2006-10-12 2007-10-10 Vaccine Comprising an Oil in Water Emulsion Adjuvant
US14/593,911 Active US9700605B2 (en) 2006-10-12 2015-01-09 Vaccine comprising an oil in water emulsion

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/593,911 Active US9700605B2 (en) 2006-10-12 2015-01-09 Vaccine comprising an oil in water emulsion

Country Status (18)

Country Link
US (2) US20100183667A1 (no)
JP (2) JP5230632B2 (no)
KR (1) KR101151202B1 (no)
CN (1) CN101522218B (no)
AU (1) AU2007306381B2 (no)
BR (1) BRPI0717219B8 (no)
CA (1) CA2664619C (no)
CR (1) CR10726A (no)
DK (1) DK2086582T3 (no)
EA (1) EA015817B1 (no)
ES (1) ES2397714T3 (no)
HR (1) HRP20130023T1 (no)
IL (1) IL197512A (no)
MA (1) MA30866B1 (no)
MX (1) MX2009003325A (no)
NO (1) NO20091022L (no)
PT (1) PT2086582E (no)
WO (1) WO2008043774A1 (no)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080181911A1 (en) * 2005-03-23 2008-07-31 Emmanuel Jules Hanon Use Of An Influenza Virus, An Oil-In-Water Emulsion Adjuvant, To Induce Cd4 T-Cell And/Or Improved B-Memory Cell Response
US20090136543A1 (en) * 2007-04-20 2009-05-28 William Ripley Ballou Vaccine
US20120164178A1 (en) * 2008-04-16 2012-06-28 Ballou Jr William Ripley Vaccine
US8221761B1 (en) * 1999-02-26 2012-07-17 Novartis Ag Enhancement of bactericidal activity of neisseria antigens with oligonucleotides containing CG motifs
US9278127B2 (en) 2006-07-17 2016-03-08 Glaxosmithkline Biologicals, Sa Influenza vaccine

Families Citing this family (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10144906B4 (de) 2001-09-12 2013-11-28 Novartis Vaccines And Diagnostics Gmbh Verfahren zur großtechnischen Herstellung von Impfstoffen
CN101522218B (zh) 2006-10-12 2012-09-26 葛兰素史密丝克莱恩生物有限公司 包含水包油乳液佐剂的疫苗
UA99719C2 (ru) 2006-11-03 2012-09-25 Шеринг-Плау Лтд. Вакцина для собак против болезни лайма
GB0714963D0 (en) 2007-08-01 2007-09-12 Novartis Ag Compositions comprising antigens
EP2268309B1 (en) 2008-03-18 2015-01-21 Novartis AG Improvements in preparation of influenza virus vaccine antigens
EA201001478A1 (ru) * 2008-04-16 2011-06-30 Глаксосмитклайн Байолоджикалс С.А. Вакцина
US11236366B2 (en) 2008-08-28 2022-02-01 Novartis Ag Production of squalene from hyper-producing yeasts
CN102159242A (zh) 2008-09-18 2011-08-17 诺华有限公司 疫苗佐剂组合
US20120093859A1 (en) 2009-02-10 2012-04-19 Novartis Ag Influenza vaccine regimens for pandemic-associated strains
DK2396032T3 (en) 2009-02-10 2016-12-19 Seqirus Uk Ltd Influenza vaccines with reduced amounts of squalene
JP2012517416A (ja) 2009-02-10 2012-08-02 ノバルティス アーゲー 増加した量のh3抗原を含むインフルエンザワクチン
AR075437A1 (es) 2009-02-17 2011-03-30 Glaxosmithkline Biolog Sa Composicion inmunogenica que comprende al menos un antigeno del virus del dengue inactivado y un adyuvante sin aluminio, metodo para producir dicha vacuna y su uso para preparar un medicamento
CA2756533A1 (en) 2009-03-24 2010-09-30 Novartis Ag Combinations of meningococcal factor h binding protein and pneumococcal saccharide conjugates
PL2510947T3 (pl) 2009-04-14 2016-09-30 Kompozycje do immunizacji przeciwko Staphylococcus aureus
SI2401384T1 (sl) 2009-05-21 2013-01-31 Novartis Ag Reverzna genetika z uporabo neendogenih pol i promotorjev
CA2769673A1 (en) 2009-07-31 2011-02-03 Novartis Ag Reverse genetics systems
AU2010309404B2 (en) 2009-10-20 2015-07-09 Seqirus UK Limited Improved reverse genetics methods for virus rescue
GB0918392D0 (en) 2009-10-20 2009-12-02 Novartis Ag Diagnostic and therapeutic methods
GB0919117D0 (en) 2009-10-30 2009-12-16 Glaxosmithkline Biolog Sa Process
GB0919690D0 (en) 2009-11-10 2009-12-23 Guy S And St Thomas S Nhs Foun compositions for immunising against staphylococcus aureus
DK2528621T3 (da) 2010-01-27 2017-01-02 Glaxosmithkline Biologicals Sa Modificerede tuberkuloseantigener
CN102802662A (zh) 2010-03-18 2012-11-28 诺华有限公司 用于脑膜炎球菌血清组b的含佐剂疫苗
EA027142B1 (ru) 2010-05-12 2017-06-30 Новартис Аг Улучшенные способы получения сквалена
EP2571520B1 (en) 2010-05-21 2018-04-04 Seqirus UK Limited Influenza virus reassortment method
GB201009861D0 (en) 2010-06-11 2010-07-21 Novartis Ag OMV vaccines
EP2680885B8 (en) 2011-03-02 2018-07-25 GlaxoSmithKline Biologicals SA Combination vaccines with lower doses of antigen and/or adjuvant
WO2013057715A1 (en) 2011-10-20 2013-04-25 Novartis Ag Adjuvanted influenza b virus vaccines for pediatric priming
EP2791680B1 (en) 2011-12-12 2015-12-09 Novartis AG Assay for influenza virus hemagglutinins
CN104023744A (zh) 2011-12-23 2014-09-03 诺华股份有限公司 用于针对金黄色葡萄球菌免疫的稳定组合物
AU2013205478B9 (en) 2012-03-02 2014-11-20 Seqirus UK Limited Influenza virus reassortment
RU2014140521A (ru) 2012-03-08 2016-04-27 Новартис Аг Адъювантные составы бустерных вакцин
JP2015528456A (ja) 2012-08-31 2015-09-28 ノバルティス アーゲー Staphylococcusaureusに対する免疫化のための安定化されたタンパク質
US9926344B2 (en) 2012-08-31 2018-03-27 Glaxosmithkline Biologicals Sa Stabilised proteins for immunising against Staphylococcus aureus
RU2015106930A (ru) 2012-09-06 2016-10-20 Новартис Аг Комбинированные вакцины с менингококком серогруппы в и к/д/с
GB201218195D0 (en) 2012-10-10 2012-11-21 Istituto Zooprofilattico Sperimentale Delle Venezie Composition
BR112015008040A2 (pt) 2012-10-12 2017-07-04 Glaxosmithkline Biologicals Sa composição de vacina, vacina de combinação, e, processos para preparar um componente ap e para a fabricação de uma composição de vacina
ES2826555T3 (es) 2012-11-30 2021-05-18 Glaxosmithkline Biologicals Sa Antígenos de Pseudomonas y combinación de antígenos
AU2013354219A1 (en) 2012-12-03 2015-07-02 Novartis Ag Reassortant influenza a viren
CN104884081A (zh) 2012-12-05 2015-09-02 葛兰素史密丝克莱恩生物有限公司 免疫原性组合物
EP2934574A1 (en) 2012-12-18 2015-10-28 GlaxoSmithKline Biologicals SA Conjugates for protecting against diphtheria and/or tetanus
WO2014141125A2 (en) 2013-03-13 2014-09-18 Novartis Ag Influenza virus reassortment
CA2903345A1 (en) * 2013-03-19 2014-09-25 Biotech Tools S.A. Allergen preparation
EP2801372A3 (en) 2013-05-10 2015-02-18 Novartis AG Avoiding narcolepsy risk in influenza vaccines
PT3542819T (pt) * 2013-05-14 2021-10-14 Zoetis Services Llc Composições de vacina inovadoras que compreendem oligonucleótidos imunoestimulantes
DE202013005130U1 (de) 2013-06-05 2013-09-10 Novartis Ag Influenza Virus Reassortierung
DE202013005100U1 (de) 2013-06-05 2013-08-26 Novartis Ag Influenza Virus Reassortierung
MX2015016755A (es) 2013-06-06 2016-04-13 Novartis Ag Reagrupamiento del virus de influenza.
RU2710393C2 (ru) 2014-01-21 2019-12-26 Пфайзер Инк. Способ получения иммуногенного конъюгата капсульный полисахарид Streptococcus pneumoniae-белок-носитель
LT3122378T (lt) 2014-03-26 2020-02-10 Glaxosmithkline Biologicals S.A. Mutantų stafilokokiniai antigenai
WO2015181173A1 (en) * 2014-05-26 2015-12-03 Becton Dickinson France Method for storing an emulsion-adjuvanted vaccine in a lubricated medical injection device
ES2721306T3 (es) 2014-12-02 2019-07-30 Novartis Ag Fabricación de composiciones que contienen tensioactivo
HUE051783T2 (hu) 2015-07-07 2021-03-29 Seqirus Uk Ltd Eljárás immunogén hemagglutinin mennyiségi meghatározására
BE1024160B9 (fr) 2015-12-22 2017-12-06 Glaxosmithkline Biologicals Sa Formulation immunogène
EP3518963A4 (en) * 2016-09-27 2020-06-10 ImmunoVaccine Technologies Inc. METHOD FOR USE OF B-CELL EPITOP COMPOSITIONS WITH LOW DOSAGE VOLUME FOR INDUCING AN ANTIBODY IMMUNE REACTION IN PERSONS
CN106511996B (zh) * 2016-11-04 2019-09-10 江苏省农业科学院 用于口蹄疫疫苗的复乳型佐剂及其制备方法
US20200222550A1 (en) 2017-01-31 2020-07-16 Merck Sharp & Dohme Corp. Methods for production of capsular polysaccharide protein conjugates from streptococcus pneumoniae serotype 19f
CN109652384B (zh) * 2019-02-21 2021-10-26 昆明理工大学 一种体外培养戊型肝炎病毒的方法
IL293031A (en) 2019-11-18 2022-07-01 Seqirus Pty Ltd A method for producing reassortant influenza viruses
KR20210117832A (ko) * 2020-03-20 2021-09-29 강원대학교산학협력단 Gla-se를 포함하는 수족구병 백신 조성물
EP4171629A1 (en) 2020-06-29 2023-05-03 GlaxoSmithKline Biologicals S.A. Adjuvants
CA3190375A1 (en) 2020-08-24 2022-03-03 Natalie ANOSOVA Covid-19 vaccines with tocopherol-containing squalene emulsion adjuvants
US20240165224A1 (en) 2021-03-26 2024-05-23 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2023020994A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023020993A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023020992A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
KR20240033309A (ko) * 2022-09-05 2024-03-12 주식회사 차백신연구소 에멀전 제형의 면역증강제 조성물 및 이의 제조 방법

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4652518A (en) * 1982-07-02 1987-03-24 Orion Corporation, Ltd. Diagnosing chlamydia infections with Re-lipopolysaccharide complexed to carrier or antibody thereto
US20090136543A1 (en) * 2007-04-20 2009-05-28 William Ripley Ballou Vaccine

Family Cites Families (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2905287A1 (de) 1979-02-12 1980-08-21 Siemens Ag Verfahren zur automatischen elektrochemischen endpunktsbestimmung einer titration
US4235877A (en) 1979-06-27 1980-11-25 Merck & Co., Inc. Liposome particle containing viral or bacterial antigenic subunit
US4454119A (en) 1981-06-29 1984-06-12 Mitsubishi Chemical Industries Limited Therapeutic agents
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
FI861417A0 (fi) 1985-04-15 1986-04-01 Endotronics Inc Hepatitis b ytantigen framstaelld med rekombinant-dna-teknik, vaccin, diagnostiskt medel och cellinjer samt foerfaranden foer framstaellning daerav.
JP2851288B2 (ja) 1987-06-05 1999-01-27 アメリカ合衆国 癌診断および管理における自己分泌運動性因子
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4940460A (en) 1987-06-19 1990-07-10 Bioject, Inc. Patient-fillable and non-invasive hypodermic injection device assembly
EP0304578B1 (en) 1987-06-22 2001-10-24 Medeva Holdings Bv Peptide comprising hepatitis B surface antigen
US5339163A (en) 1988-03-16 1994-08-16 Canon Kabushiki Kaisha Automatic exposure control device using plural image plane detection areas
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
GB8819209D0 (en) 1988-08-12 1988-09-14 Research Corp Ltd Polypeptide & dna encoding same
US4999403A (en) 1988-10-28 1991-03-12 Exxon Chemical Patents Inc. Graft polymers of functionalized ethylene-alpha-olefin copolymer with polypropylene, methods of preparation, and use in polypropylene compositions
JP3237842B2 (ja) 1988-12-16 2001-12-10 オランダ国 ニューモリシンミュータント及びそれから製造されたニューモコッカルワクチン
EP0382271B1 (en) 1989-02-04 1994-12-21 Akzo Nobel N.V. Tocols as adjuvant in vaccine
CA2017507C (en) 1989-05-25 1996-11-12 Gary Van Nest Adjuvant formulation comprising a submicron oil droplet emulsion
EP0414374B1 (en) 1989-07-25 1997-10-08 Smithkline Biologicals S.A. Novel antigens and methods for their preparation
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
EP0468520A3 (en) 1990-07-27 1992-07-01 Mitsui Toatsu Chemicals, Inc. Immunostimulatory remedies containing palindromic dna sequences
GB9118204D0 (en) 1991-08-23 1991-10-09 Weston Terence E Needle-less injector
DK0614465T3 (da) 1991-11-16 1999-09-27 Smithkline Beecham Biolog Hybridprotein mellem CS fra Plasmodium og HBsAg
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
US5569189A (en) 1992-09-28 1996-10-29 Equidyne Systems, Inc. hypodermic jet injector
US5334144A (en) 1992-10-30 1994-08-02 Becton, Dickinson And Company Single use disposable needleless injector
HU219056B (hu) 1993-03-23 2001-02-28 Smithkline Beecham Biologicals Sa 3-O-Dezacilezett monofoszforil-lipid A-t tartalmazó vakcinakészítmény
GB9326253D0 (en) * 1993-12-23 1994-02-23 Smithkline Beecham Biolog Vaccines
US5688506A (en) 1994-01-27 1997-11-18 Aphton Corp. Immunogens against gonadotropin releasing hormone
WO1995024176A1 (en) 1994-03-07 1995-09-14 Bioject, Inc. Ampule filling device
US5466220A (en) 1994-03-08 1995-11-14 Bioject, Inc. Drug vial mixing and transfer device
WO1995026204A1 (en) 1994-03-25 1995-10-05 Isis Pharmaceuticals, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
US5599302A (en) 1995-01-09 1997-02-04 Medi-Ject Corporation Medical injection system and method, gas spring thereof and launching device using gas spring
JP3958360B2 (ja) 1995-02-24 2007-08-15 キャンタブ ファーマシューティカルズ リサーチ リミティド 免疫治療剤として役立つポリペプチド及びポリペプチド調製の方法
UA56132C2 (uk) 1995-04-25 2003-05-15 Смітклайн Бічем Байолоджікалс С.А. Композиція вакцини (варіанти), спосіб стабілізації qs21 відносно гідролізу (варіанти), спосіб приготування композиції вакцини
US5730723A (en) 1995-10-10 1998-03-24 Visionary Medical Products Corporation, Inc. Gas pressured needle-less injection device and method
US5843464A (en) 1995-06-02 1998-12-01 The Ohio State University Synthetic chimeric fimbrin peptides
US5666153A (en) 1995-10-03 1997-09-09 Virtual Shopping, Inc. Retractable teleconferencing apparatus
US5893397A (en) 1996-01-12 1999-04-13 Bioject Inc. Medication vial/syringe liquid-transfer apparatus
GB9607549D0 (en) 1996-04-11 1996-06-12 Weston Medical Ltd Spring-powered dispensing device
AR008242A1 (es) 1996-06-18 1999-12-29 Alza Corp Dispositivo para atravesar el estrato corneo de una superficie corporal
US20030133944A1 (en) * 2001-04-05 2003-07-17 Smithkline Beecham Biologicals S.A. Vaccine composition against malaria
US5980898A (en) 1996-11-14 1999-11-09 The United States Of America As Represented By The U.S. Army Medical Research & Material Command Adjuvant for transcutaneous immunization
AU722574B2 (en) 1996-12-20 2000-08-10 Alza Corporation Device and method for enhancing transdermal agent flux
US6130043A (en) 1997-05-02 2000-10-10 Abbott Laboratories Reagents and methods useful for detecting diseases of the prostate
US5993412A (en) 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
KR100619350B1 (ko) 1997-07-21 2006-09-05 박스터 헬쓰케어 에스.에이. 변형 면역성 뉴멀리신 백신조성물
GB9717953D0 (en) 1997-08-22 1997-10-29 Smithkline Beecham Biolog Vaccine
DE69838992T2 (de) 1997-09-05 2008-12-24 Glaxosmithkline Biologicals S.A., Rixensart Öl-in-Wasser Emulsionen mit Saponinen
GB9718901D0 (en) 1997-09-05 1997-11-12 Smithkline Beecham Biolog Vaccine
US7459524B1 (en) 1997-10-02 2008-12-02 Emergent Product Development Gaithersburg Inc. Chlamydia protein, sequence and uses thereof
KR100735651B1 (ko) 1997-11-28 2007-07-06 세로노 제네틱스 인스티튜트 에스.에이. 클라미디아 트라코마티스 게놈 서열과 폴리펩티드, 이의단편 및 이의 용도, 특히, 감염의 진단, 예방 및 치료 용도
AU756828B2 (en) 1997-12-02 2003-01-23 Powderject Vaccines, Inc. Transdermal delivery of particulate vaccine compositions
IT1298087B1 (it) 1998-01-08 1999-12-20 Fiderm S R L Dispositivo per il controllo della profondita' di penetrazione di un ago, in particolare applicabile ad una siringa per iniezioni
KR100692227B1 (ko) 1998-04-07 2007-03-09 코릭사 코포레이션 마이코박테리움 튜베르쿨로시스 항원의 융합 단백질 및이의 용도
US6319224B1 (en) 1999-08-20 2001-11-20 Bioject Medical Technologies Inc. Intradermal injection system for injecting DNA-based injectables into humans
GB9923176D0 (en) * 1999-09-30 1999-12-01 Smithkline Beecham Biolog Novel composition
US6494865B1 (en) 1999-10-14 2002-12-17 Becton Dickinson And Company Intradermal delivery device including a needle assembly
JP2001316251A (ja) * 2000-02-24 2001-11-13 Fuairudo Kk 炭素燃焼残査の超微粒子を含むスクワラン及びその製造方法
GB0025577D0 (en) * 2000-10-18 2000-12-06 Smithkline Beecham Biolog Vaccine
JP2005513021A (ja) 2001-11-16 2005-05-12 スリーエム イノベイティブ プロパティズ カンパニー Irm化合物およびトール様受容体経路に関する方法および組成物
JP5869744B2 (ja) * 2005-03-23 2016-02-24 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム Cd4t細胞および/または改善された記憶b細胞応答を誘導するためのインフルエンザウイルスおよび水中油型エマルジョンアジュバントの使用
WO2007052155A2 (en) * 2005-11-04 2007-05-10 Novartis Vaccines And Diagnostics Srl Influenza vaccine with reduced amount of oil-in-water emulsion as adjuvant
CN101522218B (zh) * 2006-10-12 2012-09-26 葛兰素史密丝克莱恩生物有限公司 包含水包油乳液佐剂的疫苗

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4652518A (en) * 1982-07-02 1987-03-24 Orion Corporation, Ltd. Diagnosing chlamydia infections with Re-lipopolysaccharide complexed to carrier or antibody thereto
US20090136543A1 (en) * 2007-04-20 2009-05-28 William Ripley Ballou Vaccine
US20100189741A1 (en) * 2007-04-20 2010-07-29 William Ripley Ballou Oil-in-water emulsion influenza vaccine

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Fluarix(TM) product information sheet, Fluarix SH 2005 English, 2005 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8221761B1 (en) * 1999-02-26 2012-07-17 Novartis Ag Enhancement of bactericidal activity of neisseria antigens with oligonucleotides containing CG motifs
US20080181911A1 (en) * 2005-03-23 2008-07-31 Emmanuel Jules Hanon Use Of An Influenza Virus, An Oil-In-Water Emulsion Adjuvant, To Induce Cd4 T-Cell And/Or Improved B-Memory Cell Response
US9730999B2 (en) 2005-03-23 2017-08-15 Glaxosmithkline Biologicals Sa Adjuvanted influenza virus compositions
US11564984B2 (en) 2006-07-17 2023-01-31 Glaxosmithkline Biologicals Sa Influenza vaccine
US9943588B2 (en) 2006-07-17 2018-04-17 Glaxosmithkline Biologicals S.A. Influenza vaccine
US9278127B2 (en) 2006-07-17 2016-03-08 Glaxosmithkline Biologicals, Sa Influenza vaccine
US9452209B2 (en) * 2007-04-20 2016-09-27 Glaxosmithkline Biologicals Sa Influenza vaccine
US9597389B2 (en) 2007-04-20 2017-03-21 Glaxosmithkline Biologicals Sa Oil-in-water emulsion influenza vaccine
US20100189741A1 (en) * 2007-04-20 2010-07-29 William Ripley Ballou Oil-in-water emulsion influenza vaccine
US10016495B2 (en) 2007-04-20 2018-07-10 Glaxosmithkline Biologicals S.A. Oil-in-water emulsion influenza vaccine
US10548969B2 (en) 2007-04-20 2020-02-04 Glaxosmithkline Biologicals Sa Oil-in-water emulsion influenza vaccine
US20090136543A1 (en) * 2007-04-20 2009-05-28 William Ripley Ballou Vaccine
US20120164178A1 (en) * 2008-04-16 2012-06-28 Ballou Jr William Ripley Vaccine

Also Published As

Publication number Publication date
BRPI0717219A2 (pt) 2015-05-26
JP5230632B2 (ja) 2013-07-10
MA30866B1 (fr) 2009-11-02
CN101522218B (zh) 2012-09-26
EA200900350A1 (ru) 2009-10-30
HRP20130023T1 (hr) 2013-02-28
BRPI0717219B8 (pt) 2021-05-25
IL197512A (en) 2013-10-31
ES2397714T3 (es) 2013-03-08
CN101522218A (zh) 2009-09-02
CR10726A (es) 2009-05-25
MX2009003325A (es) 2009-04-09
CA2664619C (en) 2012-12-11
AU2007306381A1 (en) 2008-04-17
PT2086582E (pt) 2013-01-25
JP2013067646A (ja) 2013-04-18
KR101151202B1 (ko) 2012-06-11
BRPI0717219B1 (pt) 2021-01-19
AU2007306381B2 (en) 2011-12-08
US9700605B2 (en) 2017-07-11
NO20091022L (no) 2009-04-27
KR20090066323A (ko) 2009-06-23
US20150359863A1 (en) 2015-12-17
JP5767201B2 (ja) 2015-08-19
EA015817B1 (ru) 2011-12-30
IL197512A0 (en) 2011-08-01
CA2664619A1 (en) 2008-04-17
WO2008043774A1 (en) 2008-04-17
JP2010505907A (ja) 2010-02-25
DK2086582T3 (da) 2013-02-04

Similar Documents

Publication Publication Date Title
US9700605B2 (en) Vaccine comprising an oil in water emulsion
JP6009007B2 (ja) ワクチン
US20090081253A1 (en) Composition
AU2010202974A1 (en) Vaccine compositions comprising virosomes and a saponin adjuvant
US20110189223A1 (en) Vaccine
EP2433648A2 (en) Vaccine comprising an oil in water emulsion adjuvant
NZ575471A (en) Vaccine comprising an oil in water emulsion adjuvant

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLAXOSMITHKLINE BIOLOGICALS SA, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BALLOU, WILLIAM RIPLEY, JR;HANON, EMMANUEL JULES;SIGNING DATES FROM 20090225 TO 20090316;REEL/FRAME:023749/0815

STCB Information on status: application discontinuation

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