US20090081253A1 - Composition - Google Patents

Composition Download PDF

Info

Publication number
US20090081253A1
US20090081253A1 US11/909,414 US90941406A US2009081253A1 US 20090081253 A1 US20090081253 A1 US 20090081253A1 US 90941406 A US90941406 A US 90941406A US 2009081253 A1 US2009081253 A1 US 2009081253A1
Authority
US
United States
Prior art keywords
antigen
influenza
composition
oil
immunogenic composition
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
US11/909,414
Other languages
English (en)
Inventor
Emmanuel Jules Hanon
Jean Stephenne
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=36441219&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20090081253(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from GB0505998A external-priority patent/GB0505998D0/en
Priority claimed from GB0506004A external-priority patent/GB0506004D0/en
Priority claimed from GB0506000A external-priority patent/GB0506000D0/en
Priority claimed from GB0505989A external-priority patent/GB0505989D0/en
Priority claimed from GB0506001A external-priority patent/GB0506001D0/en
Priority claimed from GB0510598A external-priority patent/GB0510598D0/en
Priority claimed from GB0510591A external-priority patent/GB0510591D0/en
Priority claimed from GB0510593A external-priority patent/GB0510593D0/en
Priority claimed from GB0510589A external-priority patent/GB0510589D0/en
Priority claimed from GB0510596A external-priority patent/GB0510596D0/en
Priority claimed from GB0603790A external-priority patent/GB0603790D0/en
Priority claimed from GB0603788A external-priority patent/GB0603788D0/en
Priority claimed from GB0603789A external-priority patent/GB0603789D0/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: HANON, EMMANUEL JULES, STEPHENNE, JEAN
Publication of US20090081253A1 publication Critical patent/US20090081253A1/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
    • A61K39/12Viral antigens
    • A61K39/155Paramyxoviridae, e.g. parainfluenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/145Orthomyxoviridae, 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
    • 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
    • A61P31/12Antivirals
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • C07K14/08RNA viruses
    • C07K14/11Orthomyxoviridae, e.g. influenza virus
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • 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
    • C12N2760/16034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16211Influenzavirus B, i.e. influenza B virus
    • C12N2760/16234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the present invention relates to influenza vaccine formulations and vaccination regimes for immunising against various diseases.
  • the invention relates to vaccine formulations comprising an oil-in-water emulsion adjuvant and 3D-MPL, 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 sterol, 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.
  • influenza vaccines and vaccines against human papilloma virus have been developed with adjuvants.
  • Influenza viruses are one of the most ubiquitous viruses present in the world, affecting both humans and livestock. Influenza results in an economic burden, morbidity and even mortality, which are significant.
  • the influenza virus is an RNA enveloped virus with a particle size of about 125 nm in diameter. It consists basically of an internal nucleocapsid or core of ribonucleic acid (RNA) associated with nucleoprotein, surrounded by a viral envelope with a lipid bilayer structure and external glycoproteins. The inner layer of the viral envelope is composed predominantly of matrix proteins and the outer layer mostly of host-derived lipid material.
  • Influenza virus comprises two surface antigens, glycoproteins neuraminidase (NA) and haemagglutinin (HA), which appear as spikes, 10 to 12 nm long, at the surface of the particles. It is these surface proteins, particularly the haemagglutinin that determine the antigenic specificity of the influenza subtypes.
  • drifts and ‘shifts’ are unpredictable and may have a dramatic impact from an immunological point of view as they eventually lead to the emergence of new influenza strains and that enable the virus to escape the immune system causing the well known, almost annual, epidemics.
  • influenza virus strains to be incorporated into influenza vaccine each season are determined by the World Health Organisation in collaboration with national health authorities and vaccine manufacturers.
  • HA is the most important antigen in defining the serological specificity of the different influenza strains.
  • This 75-80 kD protein contains numerous antigenic determinants, several of which are in regions that undergo sequence changes in different strains (strain-specific determinants) and others in regions which are common to many HA molecules (common to determinants).
  • Influenza viruses cause epidemics almost every winter, with infection rates for type A or B virus as high as 40% over a six-week period. Influenza infection results in various disease states, from a sub-clinical infection through mild upper respiratory infection to a severe viral pneumonia. Typical influenza epidemics cause increases in incidence of pneumonia and lower respiratory disease as witnessed by increased rates of hospitalization or mortality. The severity of the disease is primarily determined by the age of the host, his immune status and the site of infection.
  • influenza vaccines are either inactivated or live attenuated influenza vaccine.
  • Inactivated flu vaccines are composed of three possible forms of antigen preparation: inactivated whole virus, sub-virions where purified virus particles are disrupted with detergents or other reagents to solubilise the lipid envelope (so-called “split” vaccine) or purified HA and NA (subunit vaccine). These inactivated vaccines are given intramuscularly (i.m.) or intranasaly (i.n.).
  • Influenza vaccines of all kinds, are usually trivalent vaccines. They generally contain antigens derived from two influenza A virus strains and one influenza B strain. A standard 0.5 ml injectable dose in most cases contains 15 ⁇ g of haemagglutinin antigen component from each strain, as measured by single radial immunodiffusion (SRD) (J. M. Wood et al.: An improved single radial immunodiffusion technique for the assay of influenza haemagglutinin antigen: adaptation for potency determination of inactivated whole virus and subunit vaccines. J. Biol. Stand. 5 (1977) 237-247; J. M. Wood et al., International collaborative study of single radial diffusion and immunoelectrophoresis techniques for the assay of haemagglutinin antigen of influenza virus. J. Biol. Stand. 9 (1981) 317-330).
  • SRD single radial immunodiffusion
  • Influenza vaccines currently available are considered safe in all age groups (De Donato et al. 1999, Vaccine, 17, 3094-3101). However, there is little evidence that current influenza vaccines work in small children under two years of age. Furthermore, reported rates of vaccine efficacy for prevention of typical confirmed influenza illness are 23-72% for the elderly, which are significantly lower than the 60-90% efficacy rates reported for younger adults (Govaert, 1994, J. Am. Med. Assoc., 21, 166-1665; Gross, 1995, Ann Intern. Med. 123, 523-527).
  • influenza vaccine has been shown to correlate with serum titres of hemagglutination inhibition (HI) antibodies to the viral strain, and several studies have found that older adults exhibit lower HI titres after influenza immunisation than do younger adults (Murasko, 2002, Experimental gerontology, 37, 427-439).
  • HI hemagglutination inhibition
  • a sub-unit influenza vaccine adjuvanted with the adjuvant MF59, in the form of an oil-in-water emulsion is commercially available, and has demonstrated its ability to induce a higher antibody titer than that obtained with the non-adjuvanted sub-unit vaccine (De Donato et al. 1999, Vaccine, 17, 3094-3101). However, in a later publication, the same vaccine has not demonstrated its improved profile compared to a non-adjuvanted split vaccine (Puig-Barbera et al., 2004, Vaccine 23, 283-289).
  • Papillomaviruses are small DNA tumour viruses, which are highly species specific. So far, over 100 individual human papillomavirus (HPV) genotypes have been described. HPVs are generally specific either for the skin (e.g. HPV-1 and -2) or mucosal surfaces (e.g. HPV-6 and -11) and usually cause benign tumours (warts) that persist for several months or years. Such benign tumours may be distressing for the individuals concerned but tend not to be life threatening, with a few exceptions.
  • HPVs human papillomavirus
  • HPVs are also associated with cancers.
  • the strongest positive association between an HPV and human cancer is that which exists between HPV-16 and HPV-18 and cervical carcinoma. Cervical cancer is the most common malignancy in developing countries, with about 500,000 new cases occurring in the world each year. It is now technically feasible to actively combat primary HPV-16 infections, and even established HPV-16-containing cancers, using vaccines.
  • For a review on the prospects for prophylactic and therapeutic vaccination against HPV-16 see Cason J., Clin. Immunother. 1994; 1(4) 293-306 and Hagenesee M. E., Infections in Medicine 1997 14(7) 555-556, 559-564.
  • HPVs genomes described have at least eight early genes, E1 to E8 and two late genes L1 and L2.
  • an upstream regulatory region harbors the regulatory sequences which appear to control most transcriptional events of the HPV genome.
  • HPV L1 based vaccines are disclosed in WO94/00152, WO94/20137, WO93/02184 and WO94/05792.
  • a vaccine can comprise the L1 antigen as a monomer, a capsomer or a virus like particle.
  • Methods for the preparation of VLPs are well known in the art, and include VLP disassembly-reassembly approaches to provide enhanced homogeneity, for example as described in WO9913056 and U.S. Pat. No. 6,245,568.
  • Such particles may additionally comprise L2 proteins.
  • L2 based vaccines are described, for example, in WO93/00436.
  • Other HPV vaccine approaches are based on the early proteins, such as E7 or fusion proteins such as L2-E7.
  • an immunogenic composition comprising:
  • the invention also relates to use of a composition
  • a composition comprising:
  • the invention also relates to a method of vaccination comprising delivery of an antigen, an oil in water emulsion adjuvant as defined herein and 3D-MPL.
  • the invention also relates to a method for the preparation of an immunogenic composition comprising combining an oil in water emulsion as defined herein with an antigen and 3D-MPL.
  • FIG. 1 Oil droplet particle size distribution in SB62 oil-in-water emulsion as measured by PCS.
  • FIG. 1B shows a schematic illustration of record 22 (upper part) and record 23 (lower part) by intensity.
  • FIG. 2 Schematic illustration of the preparation of MPL bulk.
  • FIG. 3 Schematic illustration of the preparation of AS03+MPL adjuvant.
  • FIG. 6 Explo Flu-001 clinical trial. Cross-reactive CD4 T-cell response to split influenza virus antigen after vaccination with Fluarix+AS03.
  • FIG. 7 Explo Flu-001 clinical trial. B cell memory response post vaccination.
  • FIG. 8 Explo Flu-002 clinical trial. CD4 T cell response against split influenza antigen following revaccination.
  • FIG. 9 Explo Flu-002 clinical trial. Anti-HI titers following revaccination.
  • FIG. 10 Ferret study 1. Temperature monitoring (priming and challenge). FIG. 10A is priming, FIG. 10B is challenge.
  • FIG. 11 Ferret study 1. Viral shedding.
  • FIG. 12 Ferret study II. Temperature monitoring (priming and challenge). FIG. 12A is priming, FIG. 12B is challenge.
  • FIG. 13 Ferret study II. Viral shedding.
  • FIG. 14 Ferret study II. HI titers to H3N2 A/Panama (vaccine strain) ( FIG. 14A ) and to H3N2 A/Wyoming (challenge strain) ( FIG. 14B ).
  • FIG. 15 Mice study. Frequencies of CD4 T cells in C57BI/6 primed mice using whole inactivated virus as re-stimulating antigen (day 7 post-immunisation).
  • FIG. 16 Mice study. Frequencies of CD8 T cells in C57BI/6 primed mice using whole inactivated virus as re-stimulating antigen (day 7 post-immunisation).
  • FIG. 17 Mice study. Frequencies of CD4 (upper part) and CD8 (lower part) T cells in C57BI/6 mice primed with heterologous strains, using whole inactivated virus as re-stimulating antigen (day 7 post-immunisation).
  • FIG. 18 Human clinical trial. B cell memory response post-vaccination of elderly with Fluarix, Fluarix+AS03, Fluarix+AS03+MPL (difference between pre- and post-).
  • FIG. 19 Ferret study III. Temperature monitoring before and after challenge.
  • FIG. 20 Ferret study III. Viral shedding before and after challenge.
  • FIG. 21 Ferret study II. HI titers to H3N2 A/Woming (vaccine strain).
  • FIG. 22 Ferret study III. HI titers to H3N2 A/Panama (challenge strain).
  • FIG. 23 Human clinical trial. HI titers (GMTs) at days 21, 90 and 180 post vaccination (persistence).
  • FIG. 24 Human clinical trial.
  • CD4 response all double test—Pool antigen at days 21, 90 and 180 post vaccination (persistence).
  • FIG. 25 Human clinical trial. HI titers in a revaccination clinical trial with AS03+MPL compared to Fluarix.
  • FIG. 26 Human clinical trial.
  • CMI for CD4 response all double test—Pool antigen at days 0 and 21.
  • FIG. 27 Human clinical trial with AS03+MPL at two concentrations. HI titers at days 0 and 21.
  • FIG. 28 Human clinical trial with AS03+MPL at two concentrations. Reactogenicity.
  • an influenza formulation comprising a sub-unit or split influenza virus or antigenic preparation thereof together with an oil-in-water emulsion adjuvant and 3D-MPL is capable of improving the CD4 T-cell immune response and/or the B cell memory response, against said antigen or antigenic composition in a human compared to that obtained with the un-adjuvanted sub-unit or split virus or split virus antigenic preparation thereof.
  • compositions thus provide improved influenza vaccines.
  • the claimed formulations may be used to induce anti-influenza CD4-T cell responses capable of detection of influenza epitopes presented by MHC class II molecules.
  • the present Applicant has now found that it is effective to target the cell-mediated immune system in order to increase responsiveness against homologous and drift influenza strains (upon vaccination and infection).
  • an influenza formulation comprising a split influenza virus or split virus antigenic preparation thereof together with an oil-in-water emulsion adjuvant as defined herein and 3D MPL is capable of inducing at least a trend for a higher B cell memory response following the first vaccination of a human subject, compared to the un-adjuvanted composition.
  • an oil-in-water emulsion adjuvant as defined herein +3D MPL demonstrates immunogenicity results for both antibody production and B cell memory which are equivalent to, or sometimes greater than, those generated with an adjuvant devoid of the oil-in-water emulsion component.
  • Antigens that may be used in the present invention include:
  • Streptococcal antigens such as from Group A Streptococcus , or Group B Streptococcus , but is most preferably from Streptococcus pneumoniae .
  • a protein and/or saccharide antigen is most preferably used.
  • the Streptococcus pneumoniae saccharide antigen and/or at least one Streptococcus pneumoniae protein antigen(s) is most preferably selected from the group consisting of: pneumolysin, PspA or transmembrane deletion variants thereof, PspC or transmembrane deletion variants thereof, PsaA or transmembrane deletion variants thereof, glyceraldehyde-3-phosphate dehydrogenase, CbpA or transmembrane deletion variants thereof, PhtA, PhtD, PhtB, PhtE, SpsA, LytB, LytC, LytA, Sp125, Sp101, Sp128, Sp130 and Sp133, or immunologically functional equivalent thereof.
  • Streptococcus pneumoniae saccharide antigen(s) and/or Streptococcus pneumoniae protein antigen preferably selected from the group of protein antigens listed above.
  • compositions are described in WO 00/56359 and WO 02/22167 and WO 02/22168 (incorporated by reference herein).
  • the antigen may comprise capsular saccharide antigens (preferably conjugated to a carrier protein), wherein the saccharides (most preferably polysaccharides) are derived from at least four serotypes of pneumococcus.
  • the four serotypes include 6B, 14, 19F and 23F. More preferably, at least 7 serotypes are included in the composition, for example those derived from serotypes 4, 6B, 9V, 14, 18C, 19F, and 23F.
  • At least 11 serotypes are included in the composition, for example the composition in one embodiment includes capsular saccharides derived from serotypes 1, 3, 4, 5, 6B, 7F, 9V, 14, 18C, 19F and 23F (preferably conjugated to a carrier protein).
  • at least 13 saccharide antigens are included, although further saccharide antigens, for example 23 valent (such as serotypes 1, 2, 3, 4, 5, 6B, 7F, 8, 9N, 9V, 10A, 11A, 12F, 14, 15B, 17F, 18C, 19A, 19F, 20, 22F, 23F and 33F), are also contemplated by the invention.
  • serotypes 8 and 12F are advantageously included to form a 15 valent composition
  • serotypes 6A and 19A are advantageously included to form a 13 valent composition.
  • a multivalent Streptococcus pneumonia saccharide as herein described with a Streptococcus pneumoniae protein preferably selected from the group of proteins listed above.
  • a combination of pneumococcal proteins may also be advantageously utilised as described below.
  • Streptococcus pneumoniae antigens are preferably selected from the group consisting of: a protein from the polyhistidine triad family (Pht), a protein from the Lyt family, a choline binding protein, proteins having an LPXTG motif (where X is any amino acid), proteins having a Type II Signal sequence motif of LXXC (where X is any amino acid), and proteins having a Type I Signal sequence motif.
  • Pht polyhistidine triad family
  • Lyt Lyt
  • a choline binding protein proteins having an LPXTG motif (where X is any amino acid)
  • proteins having a Type II Signal sequence motif of LXXC where X is any amino acid
  • proteins having a Type I Signal sequence motif are the following proteins (or truncate or immunologically functional equivalent thereof):
  • the Pht (Poly Histidine Triad) family comprises proteins PhtA, PhtB, PhtD, and PhtE.
  • the family is characterised by a lipidation sequence, two domains separated by a proline-rich region and several histidine triads, possibly involved in metal or nucleoside binding or enzymatic activity, (3-5) coiled-coil regions, a conserved N-terminus and a heterogeneous C terminus. It is present in all strains of pneumococci tested. Homologous proteins have also been found in other Streptococci and Neisseria .
  • Preferred members of the family comprise PhtA, PhtB and PhtD. More preferably, it comprises PhtA or PhtD.
  • phrases Pht A, B, D, and E refer to proteins having sequences disclosed in the citations below as well as naturally-occurring (and man-made) variants thereof that have a sequence homology that is at least 90% identical to the referenced proteins. Preferably it is at least 95% identical and most preferably it is 97% identical.
  • PhtA is disclosed in WO 98/18930, and is also referred to Sp36. As noted above, it is a protein from the polyhistidine triad family and has the type II signal motif of LXXC.
  • PhtD is disclosed in WO 00/37105, and is also referred to Sp036D. As noted above, it also is a protein from the polyhistidine triad family and has the type II LXXC signal motif.
  • PhtB is disclosed in WO 00/37105, and is also referred to Sp036B.
  • Another member of the PhtB family is the C3-Degrading Polypeptide, as disclosed in WO 00/17370. This protein also is from the polyhistidine triad family and has the type II LXXC signal motif.
  • a preferred immunologically functional equivalent is the protein Sp42 disclosed in WO 98/18930.
  • a PhtB truncate (approximately 79 kD) is disclosed in WO99/15675 which is also considered a member of the PhtX family.
  • PhtE is disclosed in WO00/30299 and is referred to as BVH-3.
  • SpsA is a Choline binding protein (Cbp) disclosed in WO 98/39450.
  • the Lyt family is membrane associated proteins associated with cell lysis.
  • the N-terminal domain comprises choline binding domain(s), however the Lyt family does not have all the features found in the choline binding protein family (Cbp) family noted below and thus for the present invention, the Lyt family is considered distinct from the Cbp family.
  • the C-terminal domain contains the catalytic domain of the Lyt protein family.
  • the family comprises LytA, B and C.
  • LytA is disclosed in Ronda et al., Eur J Biochem, 164:621-624 (1987).
  • LytB is disclosed in WO 98/18930, and is also referred to as Sp46.
  • LytC is also disclosed in WO 98/18930, and is also referred to as Sp91.
  • a preferred member of that family is LytC.
  • Lyt family particularly LytA truncates wherein “Lyt” is defined above and “truncates” refers to proteins lacking 50% or more of the Choline binding region. Preferably such proteins lack the entire choline binding region.
  • Sp125 is an example of a pneumococcal surface protein with the Cell Wall Anchored motif of LPXTG (where X is any amino acid). Any protein within this class of pneumococcal surface protein with this motif has been found to be useful within the context of this invention, and is therefore considered a further protein of the invention. Sp125 itself is disclosed in WO 98/18930, and is also known as ZmpB—a zinc metalloproteinase.
  • Sp101 is disclosed in WO 98/06734 (where it has the reference # y85993. It is characterised by a Type I signal sequence.
  • Sp133 is disclosed in WO 98/06734 (where it has the reference # y85992. It is also characterised by a Type I signal sequence.
  • Sp128 and Sp130 are disclosed in WO 00/76540.
  • the proteins used in the present invention are preferably selected from the group PhtD, PhtA and PhtE, or a combination of 2 or all 3 of these proteins (i.e. PhtA+D, A+E, D+E or A+D+E).
  • pneumococcal protein antigens that may be included are one or more from the group consisting of: pneumolysin (also referred to as Ply; preferably detoxified by chemical treatment or mutation) [WO 96/05859, WO 90/06951, WO 99/03884], PsaA and transmembrane deletion variants thereof (Berry & Paton, Infect Immun 1996 December; 64(12):5255-62), PspA and transmembrane deletion variants thereof (U.S. Pat. No.
  • pneumolysin also referred to as Ply; preferably detoxified by chemical treatment or mutation
  • PsaA and transmembrane deletion variants thereof Berry & Paton, Infect Immun 1996 December; 64(12):5255-62
  • PspA and transmembrane deletion variants thereof U.S. Pat. No.
  • Choline Binding Protein family members of that family were originally identified as pneumococcal proteins that could be purified by choline-affininty chromatography. All of the choline-binding proteins are non-covalently bound to phosphorylcholine moieties of cell wall teichoic acid and membrane-associated lipoteichoic acid. Structurally, they have several regions in common over the entire family, although the exact nature of the proteins (amino acid sequence, length, etc.) can vary.
  • choline binding proteins comprise an N terminal region (N), conserved repeat regions (R1 and/or R2), a proline rich region (P) and a conserved choline binding region (C), made up of multiple repeats, that comprises approximately one half of the protein.
  • Cbp Choline Binding Protein family
  • CbpA is disclosed in WO 97/41151.
  • CbpD and CbpG are disclosed in WO 00/29434.
  • PspC is disclosed in WO 97/09994.
  • PbcA is disclosed in WO 98/21337.
  • the Choline Binding Proteins are selected from the group consisting of CbpA, PbcA, SpsA and PspC.
  • a Cbp is the further protein utilised it may be a Cbp truncate wherein “Cbp” is defined above and “truncate” refers to proteins lacking 50% or more of the Choline binding region (C).
  • truncate refers to proteins lacking 50% or more of the Choline binding region (C).
  • such proteins lack the entire choline binding region.
  • the such protein truncates lack (i) the choline binding region and (ii) a portion of the N-terminal half of the protein as well, yet retain at least one repeat region (R1 or R2). More preferably still, the truncate has 2 repeat regions (R1 and R2).
  • NR1 ⁇ R2, R1 ⁇ R2, NR1 ⁇ R2P and R1 ⁇ R2P are also contemplated within the scope of this invention.
  • Cbp truncate-Lyt truncate chimeric proteins (or fusions) may also be used in the composition of the invention.
  • this comprises NR1 ⁇ R2 (or R1 ⁇ R2 or NR1 ⁇ R2P or R1 ⁇ R2P) of Cbp and the C-terminal portion (Cterm, i.e., lacking the choline binding domains) of Lyt (e.g., LytCCterm or Sp91Cterm). More preferably Cbp is selected from the group consisting of CbpA, PbcA, SpsA and PspC. More preferably still, it is CbpA.
  • Lyt is LytC (also referred to as Sp91).
  • a PspA or PsaA truncate lacking the choline binding domain (C) and expressed as a fusion protein with Lyt may also be used.
  • Lyt is LytC.
  • pneumococcal composition it is possible to combine different pneumococcal proteins of the invention.
  • the combination of proteins of the invention are selected from 2 or more (3 or 4) different categories such as proteins having a Type II Signal sequence motif of LXXC (where X is any amino acid, e.g., the polyhistidine triad family (Pht)), choline binding proteins (Cbp), proteins having a Type I Signal sequence motif (e.g., Sp101), proteins having a LPXTG motif (where X is any amino acid, e.g., Sp128, Sp130), toxins (e.g., Ply), etc.
  • Preferred examples within these categories (or motifs) are the proteins mentioned above, or immunologically functional equivalents thereof.
  • Toxin+Pht, toxin+Cbp, Pht+Cbp, and toxin+Pht+Cbp are preferred category combinations.
  • Preferred beneficial combinations include, but are not limited to, PhtD+NR1 ⁇ R2, PhtD+NR1 ⁇ R2 ⁇ Sp91Cterm chimeric or fusion proteins, PhtD+Ply, PhtD+Sp128, PhtD+PsaA, PhtD+PspA, PhtA+NR1 ⁇ R2, PhtA+NR1 ⁇ R2 ⁇ Sp91Cterm chimeric or fusion proteins, PhtA+Ply, PhtA+Sp128, PhtA+PsaA, PhtA+PspA, NR1 ⁇ R2+LytC, NR1 ⁇ R2+PspA, NR1 ⁇ R2+PsaA, NR1 ⁇ R2+Sp128, R1 ⁇ R2+LytC, R1 ⁇ R2+PspA, R1 ⁇ R2+PsaA, R1 ⁇ R2+PsaA, R1 ⁇ R2+Sp128, R1 ⁇ R2+LytC,
  • a particularly preferred combination of pneumococcal proteins comprises Ply (or a truncate or immunologically functional equivalent thereof)+PhtD (or a truncate or immunologically functional equivalent thereof) optionally with NR1 ⁇ R2 (or R1 ⁇ R2 or NR1 ⁇ R2P or R1 ⁇ R2P).
  • NR1 ⁇ R2 or R1 ⁇ R2 or NR1 ⁇ R2P or R1 ⁇ R2P
  • NR1 ⁇ R2 is from CbpA or PspC. More preferably it is from CbpA.
  • the antigen may be a pneumococcus saccharide conjugate comprising polysaccharide antigens derived from at least four serotypes, preferably at least seven serotypes, more preferably at least eleven serotypes, and at least one, but preferably 2, 3, or 4, Streptococcus pneumoniae proteins preferably selected from the group of proteins described above.
  • one of the proteins is PhtD (or an immunologically functional equivalent thereof) and/or Ply (or an immunologically functional equivalent thereof).
  • a problem associated with the polysaccharide approach to vaccination is the fact that polysaccharides per se are poor immunogens.
  • saccharides may be conjugated to protein carriers, which provide bystander T-cell help.
  • the saccharides utilised in the invention are linked to such a protein carrier.
  • a protein carrier which are currently commonly used for the production of saccharide immunogens include the Diphtheria and Tetanus toxoids (DT, DT CRM197 and TT respectively), Keyhole Limpet Haemocyanin (KLH), OMPC from N. meningitidis , and the purified protein derivative of Tuberculin (PPD).
  • a preferred carrier for the pneumococcal saccharide based immunogenic compositions is protein D from Haemophilus influenzae (EP 594610-B), or fragments thereof. Fragments suitable for use include fragments encompassing T-helper epitopes. In particular a protein D fragment will preferably contain the N-terminal 1 ⁇ 3 of the protein.
  • a protein D carrier is useful as a carrier in compositions where multiple pneumococcal saccharide antigens are conjugated. One or more pneumococcal saccharides in a combination may be advantageously conjugated onto protein D.
  • a further preferred carrier for the pneumococcal saccharide is the pneumococcal protein itself (as defined above in section “Pneumococcal Proteins of the invention”).
  • the saccharide may be linked to the carrier protein by any known method (for example, by Likhite, U.S. Pat. No. 4,372,945 and by Armor et al., U.S. Pat. No. 4,474,757).
  • CDAP conjugation is carried out (WO 95/08348).
  • the protein:saccharide (weight:weight) ratio of the conjugates is 0.3:1 to 1:1, more preferably 0.6:1 to 0.8:1, and most preferably about 0.7:1.
  • compositions of the invention comprise one or more conjugated pneumococcal saccharides, and one or more pneumococcal proteins of the invention.
  • pneumococcal saccharides and proteins can be stably stored as bulk components adsorbed onto aluminium phosphate in a liquid form.
  • antigens are suitably derived from 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 gpI, 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 Syncytial virus (such as F, N, M
  • 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, PiIC, 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.
  • 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 (HWMP) (WO 99/17741), ORF3 (EP 366 412), and putative membrane proteins (Pmps).
  • HWMP High Molecular Weight Protein
  • ORF3 ORF3
  • Pmps putative membrane proteins
  • Other Chlamydia antigens of the composition can be selected from the group described in WO 99/28475.
  • Preferred bacterial compositions comprise antigens derived from Haemophilus spp., including H. influenzae type B (for example PRP and conjugates thereof), 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 varients or fusion proteins thereof.
  • H. influenzae type B for example PRP and conjugates thereof
  • non typeable H. influenzae for example OMP26
  • high molecular weight adhesins for example P5, P6, protein D and lipoprotein D
  • fimbrin and fimbrin derived peptides U.S. Pat. No. 5,843,464
  • the vaccine formulation of the invention comprises the HIV-1 antigen, gp120, especially when expressed in CHO cells.
  • the composition of the invention comprises gD2t as hereinabove defined.
  • compositions contain an 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
  • compositions comprise L1 particles or capsomers, 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 proteinD(1 ⁇ 3)-E7 disclosed in GB 9717953.5 (PCT/EP98/05285).
  • a preferred HPV cervical infection or cancer, prophylaxis or therapeutic compositions may comprise HPV 16 or 18 antigens.
  • VLP virus like particle
  • Such antigens, virus like particles and capsomer are per se known. See for example WO94/00152, WO94/20137, WO94/05792, and WO93/02184.
  • Additional early proteins may be included alone or as fusion proteins such as E7, E2 or preferably E5 for example; particularly preferred embodiments of this includes a VLP comprising L1E7 fusion proteins (WO 96/11272).
  • 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 a composition 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.
  • composition of the present invention may additionally comprise antigens from other HPV strains, preferably from strains HPV 31 or 33.
  • compositions of the present invention further comprise antigens derived from parasites that cause Malaria.
  • preferred antigens from Plasmodia falciparum include 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 the International Patent Application No. PCT/EP92/02591, published under Number WO 93/10152 claiming priority from UK patent application No. 9124390.7.
  • RTS When expressed in yeast RTS is produced as a lipoprotein particle, and when it is co-expressed with the S antigen from HBV it produces a mixed particle known as RTS,S.
  • TRAP antigens are described in the International Patent Application No. PCT/GB89/00895, published under WO 90/01496.
  • a preferred embodiment of the present invention is a Malaria vaccine wherein the antigenic preparation comprises a combination of the RTS,S and TRAP antigens.
  • Other plasmodia antigens that are likely candidates to be components of a multistage Malaria vaccine are P.
  • One embodiment of the present invention is a composition comprising 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 which may be selected for example from the group consisting of MPS1, MSP3, AMA1, LSA1 or LSA3.
  • compositions may also contain an anti-tumour antigen and be useful for the immunotherapeutic treatment of cancers.
  • the antigen may be a tumour rejection antigens such as those for prostrate, breast, colorectal, lung, pancreatic, renal or melanoma cancers.
  • Exemplary antigens include MAGE 1, 3 and MAGE 4 or other MAGE antigens such as disclosed in WO99/40188, PRAME, BAGE, Lü (also known as NY Eos 1) SAGE and HAGE (WO 99/53061) 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. Indeed these antigens are expressed in a wide range of tumour types such as melanoma, lung carcinoma, sarcoma and bladder carcinoma.
  • MAGE antigens for use in the present invention may be expressed as a fusion protein with an expression enhancer or an Immunological fusion partner.
  • the Mage protein may be fused to Protein D from Heamophilus infuenzae B or a lipidated derivative thereof.
  • the fusion partner may comprise the first 1 ⁇ 3 of Protein D.
  • tumour-specific antigens include, but are not restricted to KSA (GA733) tumour-specific gangliosides such as GM 2, and GM3 or conjugates thereof to carrier proteins; or 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.
  • KSA tumour-specific gangliosides
  • GM 2 tumour-specific gangliosides
  • GM3 or conjugates thereof to carrier proteins or 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.
  • GnRH Gonadotrophin hormone releasing hormone
  • prostate antigens are utilised, such as Prostate specific antigen (PSA), PAP, PSCA (PNAS 95(4) 1735-1740 1998), PSMA or antigen known as Prostase.
  • PSA Prostate specific antigen
  • PAP PAP
  • PSCA PSCA
  • PSMA antigen known as Prostase.
  • Prostase is a prostate-specific serine protease (trypsin-like), 254 amino acid-long, with a conserved serine protease catalytic triad H-D-S and a amino-terminal pre-propeptide sequence, indicating a potential secretory function (P. Nelson, Lu Gan; C. Ferguson, P. Moss, R. Gelinas, L. Hood & K. Wand, “Molecular cloning and characterisation of prostase, an androgen-regulated serine protease with prostate restricted expression, In Proc. Natl. Acad. Sci. USA (1999) 96, 3114-3119). A putative glycosylation site has been described. The predicted structure is very similar to other known serine proteases, showing that the mature polypeptide folds into a single domain. The mature protein is 224 amino acids-long, with one A2 epitope shown to be naturally processed.
  • Prostase nucleotide sequence and deduced polypeptide sequence and homologs are disclosed in Ferguson, et al. (Proc. Natl. Acad. Sci. USA 1999, 96, 3114-3119) and in International Patent Applications No. WO 98/12302 (and also the corresponding granted patent U.S. Pat. No. 5,955,306), WO 98/20117 (and also the corresponding granted patents U.S. Pat. No. 5,840,871 and U.S. Pat. No. 5,786,148) (prostate-specific kallikrein) and WO 00/04149 (P703P).
  • compositions comprising prostase protein fusions based on prostase protein and fragments and homologues thereof (“derivatives”).
  • derivatives are suitable for use in therapeutic vaccine formulations which are suitable for the treatment of a prostate tumours.
  • the fragment will contain at least 20, preferably 50, more preferably 100 contiguous amino acids as disclosed in the above referenced patent and patent applications.
  • a further preferred prostate antigen is known as P501S, sequence ID no 113 of Wo98/37814. Immunogenic fragments and portions thereof comprising at least 20, preferably 50, more preferably 100 contiguous amino acids as disclosed in the above referenced patent application. See for example PS108 (WO 98/50567). Other prostate specific antigens are known from Wo98/37418, and WO/004149. Another is STEAP PNAS 96 14523 14528 7-12 1999.
  • tumour associated antigens useful in the context of the present invention include: Plu-1 J. Biol. Chem. 274 (22) 15633-15645, 1999, HASH-1, HasH-2, Cripto (Salomon et al Bioessays 199, 21 61-70,U.S. Pat. No. 5,654,140) Criptin U.S. Pat. No. 5,981,215, Additionally, antigens particularly relevant for therapy of cancer also comprise tyrosinase and survivin.
  • Mucin dervied peptides such as Muc1 see for example U.S. Pat. No. 5,744,144 U.S. Pat. No. 5,827,666 WO 8805054, U.S. Pat. No. 4,963,484.
  • Muc 1 derived peptides that comprise at least one repeat unit of the Muc 1 peptide, preferably at least two such repeats and which is recognised by the SM3 antibody (U.S. Pat. No. 6,054,438).
  • Other mucin derived peptides include peptide from Muc 5.
  • the antigen of the invention may be a breast cancer antigens such as her 2/Neu, mammaglobin (U.S. Pat. No. 5,668,267) or those disclosed in WO/00 52165, WO99/33869, WO99/19479, WO 98/45328.
  • Her 2 neu antigens are disclosed inter alia, in U.S. Pat. No. 5,801,005.
  • the Her 2 neu comprises the entire extracellular domain (comprising approximately amino acid 1-645) or fragments thereof and at least an immunogenic portion of or the entire intracellular domain approximately the C terminal 580 amino acids.
  • the intracellular portion should comprise the phosphorylation domain or fragments thereof.
  • Such constructs are disclosed in WO00/44899.
  • a particularly preferred construct is known as ECD PD a second is known as ECD PD See Wo/00/44899.
  • the her 2 neu as used herein can be derived from rat, mouse or human.
  • compositions may contain antigens associated with tumour-support mechanisms (e.g. angiogenesis, tumour invasion) for example tie 2, VEGF.
  • antigens may include nucleic acid, pathogen derived antigen or antigenic preparations, recombinantly produced protein or peptides, and chimeric fusion proteins.
  • the antigen is OspA.
  • the OspA may be a full mature protein in a lipidated form virtue of the host cell ( E. Coli ) termed (Lipo-OspA) or a non-lipidated derivative.
  • non-lipidated derivatives include the non-lipidated NS1-OspA fusion protein which has the first 81 N-terminal amino acids of the non-structural protein (NS1) of the influenza virus, and the complete OspA protein, and another, MDP-OspA is a non-lipidated form of OspA carrying 3 additional N-terminal amino acids.
  • compositions of the present invention may be used for the prophylaxis or therapy of allergy.
  • vaccines would comprise allergen specific (for example Der p1) and allergen non-specific antigens (for example peptides derived from human IgE, including but not restricted to the stanworth decapeptide (EP 0 477 231 B1)).
  • compositions of the present invention may also be used for the prophylaxis or therapy of chronic disorders others than allergy, cancer or infectious diseases.
  • chronic disorders are diseases such as atherosclerosis, and Alzheimer.
  • compositions of the present invention are particularly suited for the immunotherapeutic treatment of diseases, such as chronic conditions and cancers, but also for the therapy of persistent infections. Accordingly the compositions of the present invention are particularly suitable for the immunotherapy of infectious diseases, such as Tuberculosis (TB), AIDS and Hepatitis B (HepB) virus infections.
  • infectious diseases such as Tuberculosis (TB), AIDS and Hepatitis B (HepB) virus infections.
  • a method of treatment of an individual susceptible to or suffering from AIDS comprising the administration of a vaccine of the present invention to the individual, thereby reducing the amount of CD4+ T-cell decline caused by subsequent HIV infection, or slowing or halting the CD4+ T-cell decline in an individual already infected with HIV.
  • antigens include bacterial (preferably capsular) saccharides other than (or in addition to) those pneumococcal antigens described above.
  • Polysaccharide antigens are conveniently stored in liquid bulk adsorbed onto aluminium phosphate—it is therefore straightforward to generate vaccine compositions of the invention by admixing said liquid bulk with the oil emulsions of the invention extemporaneously.
  • the other bacterial saccharides are selected from a group consisting of: N. meningitidis serogroup A capsular saccharide (MenA), N. meningitidis serogroup C capsular saccharide (MenC), N. meningitidis serogroup Y capsular saccharide (MenY), N.
  • meningitidis serogroup W-135 capsular saccharide (MenW), Group B Streptococcus group I capsular saccharide, Group B Streptococcus group II capsular saccharide, Group B Streptococcus group III capsular saccharide, Group B Streptococcus group IV capsular saccharide, Group B Streptococcus group V capsular saccharide, Staphylococcus aureus type 5 capsular saccharide, Staphylococcus aureus type 8 capsular saccharide, Vi saccharide from Salmonella typhi, N. meningitidis LPS, M. catarrhalis LPS, and H. influenzae LPS.
  • LPS lipo-polysaccharide
  • lipo-oligosaccharide lipo-polysaccharide where the lipid A portion has been detoxified by any of a number of known methods (see for example WO 97/18837 or WO 98/33923), or any molecule comprising the O-polysaccharide derived from said LPS.
  • N. meningitidis LPS it is meant one or more of the 12 known immunotypes (L1, L2, L3, L4, L5, L6, L7, L8, L9, L10, L11 or L12).
  • compositions comprising: 1) conjugated Hib, conjugated MenA and conjugated MenC; 2) conjugated Hib, conjugated MenY and conjugated MenC; 3) conjugated Hib and conjugated MenC; and 4) conjugated MenA, conjugated MenC, conjugated MenY and conjugated MenW-135.
  • the amount of PS in each of the above conjugates may be 5 or 10 ⁇ g each per 0.5 mL human dose.
  • Hib, MenA, MenC, MenW-135 and MenY are TT conjugates.
  • saccharides of the invention may be conjugated to protein carriers, which provide bystander T-cell help. It is preferred, therefore, that the saccharides utilised in the invention are linked to such a protein carrier.
  • Examples of such carriers which are currently commonly used for the production of saccharide immunogens include the Diphtheria and Tetanus toxoids (DT, DT CRM197 and TT respectively), Keyhole Limpet Haemocyanin (KLH), protein D from Haemophilus influenzae (EP 594610-B), OMPC from N. meningitidis , and the purified protein derivative of Tuberculin (PPD).
  • the saccharide may be linked to the carrier protein by any known method (for example, by Likhite, U.S. Pat. No. 4,372,945 and by Armor et al., U.S. Pat. No. 4,474,757).
  • CDAP conjugation is carried out (WO 95/08348).
  • the protein:saccharide (weight:weight) ratio of the conjugates is 0.3:1 to 1:1, more preferably 0.6:1 to 0.8:1, and most preferably about 0.7:1.
  • Combinations of antigens which provide protection against pneumococcus and a different pathogen are included in the present invention.
  • Many Paediatric vaccines are now given as a combination vaccine so as to reduce the number of injections a child has to receive.
  • other antigens from other pathogens may be formulated with the pneumococcal vaccines of the invention.
  • the vaccines of the invention can be formulated with (or administered separately but at the same time) the well known ‘trivalent’ combination vaccine comprising Diphtheria toxoid (DT), tetanus toxoid (TT), and pertussis components [typically detoxified Pertussis toxoid (PT) and filamentous haemagglutinin (FHA) with optional pertactin (PRN) and/or agglutinin 1+2], for example the marketed vaccine INFANRIX-DTPaTM (SmithKlineBeecham Biologicals) which contains DT, TT, PT, FHA and PRN antigens, or with a whole cell pertussis component for example as marketed by SmithKlineBeecham Biologicals s.a., as TritanrixTM.
  • DT Diphtheria toxoid
  • TT tetanus toxoid
  • pertussis components typically detoxified Pertussis toxoid (PT
  • the combined vaccine may also comprise other antigen, such as Hepatitis B surface antigen (HBsAg), Polio virus antigens (for instance inactivated trivalent polio virus—IPV), Moraxella catarrhalis outer membrane proteins, non-typeable Haemophilus influenzae proteins, N. meningitidis B outer membrane proteins.
  • HsAg Hepatitis B surface antigen
  • Polio virus antigens for instance inactivated trivalent polio virus—IPV
  • Moraxella catarrhalis outer membrane proteins non-typeable Haemophilus influenzae proteins
  • N. meningitidis B outer membrane proteins such as HBsAg
  • Polio virus antigens for instance inactivated trivalent polio virus—IPV
  • Moraxella catarrhalis outer membrane proteins such as non-typeable Haemophilus influenzae proteins, N. meningitidis B outer membrane proteins.
  • Moraxella catarrhalis protein antigens which can be included in a combination vaccine (especially for the prevention of otitis media) are: OMP106 [WO 97/41731 (Antex) & WO 96/34960 (PMC)]; OMP21; LbpA &/or LbpB [WO 98/55606 (PMC)]; TbpA &/or TbpB [WO 97/13785 & WO 97/32980 (PMC)]; CopB [Helminen M E, et al. (1993) Infect. Immun.
  • non-typeable Haemophilus influenzae antigens which can be included in a combination vaccine (especially for the prevention of otitis media) include: Fimbrin protein [(U.S. Pat. No. 5,766,608—Ohio State Research Foundation)] and fusions comprising peptides therefrom [eg LB1 (f) peptide fusions; U.S. Pat. No.
  • pneumococcal saccharide & protein of the invention in combination with viral antigens, for example, from influenza (attenuated, split, or subunit [e.g., surface glycoproteins neuraminidase (NA) and haemagglutinin (HA). See, e.g., Chaloupka I. et al, Eur. Journal Clin. Microbiol. Infect. Dis. 1996, 15:121-127], RSV (e.g., F and G antigens or F/G fusions, see, eg, Schmidt A. C.
  • influenza attenuated, split, or subunit
  • NA surface glycoproteins neuraminidase
  • HA haemagglutinin
  • RSV e.g., F and G antigens or F/G fusions, see, eg, Schmidt A. C.
  • a preferred Paediatric combination vaccine contemplated by the present invention for global treatment or prevention of otitis media comprises: one or more Streptococcus pneumoniae saccharide antigen(s) (preferably conjugated to protein D), one or more pneumococcal proteins (preferably those described above), and one or more surface-exposed antigen from Moraxella catarrhalis and/or non-typeable Haemophilus influenzae .
  • Protein D can advantageously be used as a protein carrier for the pneumococcal saccharides (as mentioned above), and because it is in itself an immunogen capable of producing B-cell mediated protection against non-typeable H. influenzae (ntHi).
  • the Moraxella catarrhalis or non-typeable Haemophilus influenzae antigens can be included in the vaccine in a sub-unit form, or may be added as antigens present on the surface of outer membrane vesicles (blebs) made from the bacteria.
  • the invention relates to use of a composition
  • a composition comprising:
  • composition of the invention is thus used for infections and/or diseases which are capable of being prevented or ameliorated by that composition, and suitably in which the antigen is derived from or associated with a pathogen (such as a bacteria or virus) which is associated with the disease.
  • a pathogen such as a bacteria or virus
  • Antigens from influenza virus A and B, HPV antigens, RSV A and B, SARS, streptococcus , VZV, rhinovirus, parainfluenza virus are preferred for use in the present invention, such as split influenza, VZV gE, VZV IE63, and PhtD from Streptococcus pneumonia .
  • any suitable antigen may be used.
  • composition used in the invention does not comprise an influenza subunit antigen with the MF59TM adjuvant.
  • antigens comprise a CD4 T cell epitope, or a B cell epitope or suitably both.
  • influenza virus or antigenic preparation thereof for use according to the present invention may be a split influenza virus or split virus antigenic preparation thereof.
  • the influenza preparation may contain another type of inactivated influenza antigen, such as inactivated whole virus or purified HA and NA (subunit vaccine), or an influenza virosome.
  • influenza virus may be a live attenuated influenza preparation.
  • split influenza virus or split virus antigenic preparation thereof for use according to the present invention is suitably an inactivated virus preparation where virus particles are disrupted with detergents or other reagents to solubilise the lipid envelope.
  • Split virus or split virus antigenic preparations thereof are suitably prepared by fragmentation of whole influenza virus, either infectious or inactivated, with solubilising concentrations of organic solvents or detergents and subsequent removal of all or the majority of the solubilising agent and some or most of the viral lipid material.
  • split virus antigenic preparation thereof is meant a split virus preparation which may have undergone some degree of purification compared to the split virus whilst retaining most of the antigenic properties of the split virus components.
  • split virus antigenic preparation may comprise split virus antigenic components of more than one viral strain.
  • Vaccines containing split virus called ‘influenza split vaccine’
  • split virus antigenic preparations generally contain residual matrix protein and nucleoprotein and sometimes lipid, as well as the membrane envelope proteins.
  • split virus vaccines will usually contain most or all of the virus structural proteins although not necessarily in the same proportions as they occur in the whole virus.
  • influenza virus preparation is in the form of a purified sub-unit influenza.
  • Sub-unit influenza vaccines generally contain the two major envelope proteins, HA and NA, and may have an additional advantage over whole virion vaccines as they are generally less reactogenic, particularly in young vaccinees.
  • Sub-unit vaccines can be produced either recombinantly or purified from disrupted viral particles.
  • influenza virus preparation is in the form of a virosome.
  • Virosomes are spherical, unilamellar vesicles which retain the functional viral envelope glycoproteins HA and NA in authentic conformation, intercalated in the virosomes' phospholipids bilayer membrane.
  • Said influenza virus or antigenic preparation thereof may be egg-derived or tissue-culture derived.
  • influenza virus antigen or antigenic preparations thereof according to the invention may be derived from the conventional embryonated egg method, by growing influenza virus in eggs and purifying the harvested allantoic fluid. Eggs can be accumulated in large numbers at short notice. Alternatively, they may be derived from any of the new generation methods using tissue culture to grow the virus or express recombinant influenza virus surface antigens.
  • Suitable cell substrates for growing the virus include for example dog kidney cells such as MDCK or cells from a clone of MDCK, MDCK-like cells, monkey kidney cells such as AGMK cells including Vero cells, suitable pig cell lines, or any other mammalian cell type suitable for the production of influenza virus for vaccine purposes. Suitable cell substrates also include human cells e.g. MRC-5 cells. Suitable cell substrates are not limited to cell lines; for example primary cells such as chicken embryo fibroblasts and avian cell lines are also included.
  • the influenza virus antigen or antigenic preparation thereof may be produced by any of a number of commercially applicable processes, for example the split flu process described in patent no. DD 300 833 and DD 211 444, incorporated herein by reference.
  • split flu was produced using a solvent/detergent treatment, such as tri-n-butyl phosphate, or diethylether in combination with TweenTM (known as “Tween-ether” splitting) and this process is still used in some production facilities.
  • Other splitting agents now employed include detergents or proteolytic enzymes or bile salts, for example sodium deoxycholate as described in patent no. DD 155 875, incorporated herein by reference.
  • Detergents that can be used as splitting agents include cationic detergents e.g.
  • cetyl trimethyl ammonium bromide CAB
  • other ionic detergents e.g. laurylsulfate, taurodeoxycholate, or non-ionic detergents such as the ones described above including Triton X-100 (for example in a process described in Lina et al, 2000, Biologicals 28, 95-103) and Triton N-101, or combinations of any two or more detergents.
  • Triton X-100 for example in a process described in Lina et al, 2000, Biologicals 28, 95-103
  • Triton N-101 Triton N-101
  • the preparation process for a split vaccine may include a number of different filtration and/or other separation steps such as ultracentrifugation, ultrafiltration, zonal centrifugation and chromatography (e.g. ion exchange) steps in a variety of combinations, and optionally an inactivation step eg with heat, formaldehyde or ⁇ -propiolactone or U.V. which may be carried out before or after splitting.
  • the splitting process may be carried out as a batch, continuous or semi-continuous process.
  • a preferred splitting and purification process for a split immunogenic composition is described in WO 02/097072.
  • Preferred split flu vaccine antigen preparations according to the invention comprise a residual amount of Tween 80 and/or Triton X-100 remaining from the production process, although these may be added or their concentrations adjusted after preparation of the split antigen.
  • Tween 80 and Triton X-100 are present.
  • the preferred ranges for the final concentrations of these non-ionic surfactants in the vaccine dose are:
  • Tween 80 0.01 to 1%, more preferably about 0.1% (v/v) Triton X-100:0.001 to 0.1 (% w/v), more preferably 0.005 to 0.02% (w/v).
  • the final concentration for Tween 80 ranges from 0.045%-0.09% w/v.
  • the antigen is provided as a 2 fold concentrated mixture, which has a Tween 80 concentration ranging from 0.045%-0.2% (w/v) and has to be diluted two times upon final formulation with the adjuvanted (or the buffer in the control formulation).
  • the final concentration for Triton X-100 ranges from 0.005%-0.017% w/v.
  • the antigen is provided as a 2 fold concentrated mixture, which has a Triton X-100 concentration ranging from 0.005%-0.034% (w/v) and has to be diluted two times upon final formulation with the adjuvanted (or the buffer in the control formulation).
  • influenza preparation is prepared in the presence of low level of thiomersal, or preferably in the absence of thiomersal.
  • the resulting influenza preparation is stable in the absence of organomercurial preservatives, in particular the preparation contains no residual thiomersal.
  • influenza virus preparation comprises a haemagglutinin antigen stabilised in the absence of thiomersal, or at low levels of thiomersal (generally 5 ⁇ g/ml or less).
  • the stabilization of B influenza strain is performed by a derivative of alpha tocopherol, such as alpha tocopherol succinate (also known as vitamin E succinate, i.e. VES).
  • alpha tocopherol succinate also known as vitamin E succinate, i.e. VES
  • a preferred composition contains three inactivated split virion antigens prepared from the WHO recommended strains of the appropriate influenza season.
  • influenza virus or antigenic preparation thereof and the oil-in-water emulsion adjuvant are contained in the same container. It is referred to as ‘one vial approach’.
  • the vial is a pre-filled syringe.
  • influenza virus or antigenic preparation thereof and the oil-in-water emulsion adjuvant are contained in separate containers or vials and admixed shortly before or upon administration into the subject. It is referred to as ‘two vials approach’.
  • the concentrated antigens for example the concentrated trivalent inactivated split virion antigens
  • the concentrated antigens are presented in one vial (335111) (antigen container) and a pre-filled syringe contains the adjuvant (360 ⁇ l) (adjuvant container).
  • the content of the vial containing the concentrated trivalent inactivated split virion antigens is removed from the vial by using the syringe containing the adjuvant followed by gentle mixing of the syringe.
  • the used needle Prior to injection, the used needle is replaced by an intramuscular needle and the volume is corrected to 530 ⁇ l.
  • One dose of the reconstituted adjuvanted influenza vaccine candidate corresponds to 530 ⁇ l.
  • compositions of the invention comprise antigens having CD4 T cell epitopes and optionally B cell epitopes.
  • compositions contain an antigen derived from the Human Papilloma Virus (HPV), for example from a virus considered to be responsible for genital warts (HPV 6 or HPV 11 and others), or the HPV viruses responsible for cervical cancer (HPV16, HPV18 and others).
  • HPV Human Papilloma Virus
  • prophylactic or therapeutic compositions comprise HPV 16 or 18 antigens. Infection by HPV 16 and HPV 18 is related to development of cancer.
  • Combinations of antigens from different HPV genotypes may be employed in the invention, such as a combination of HPV 16 and HPV 18 antigens, suitably in the form of VLP.
  • Antigens from additional VLP types that may be included with 16 and/or 18 include antigens from other known cancer-causing types such as HPV 31, 45, 33, 58 and 52.
  • the HPV antigens are L1 or L2 antigen monomers.
  • the invention relates to a combination of HPV L1 and L2 antigens from the same genotype presented together as a capsomer or virus like particle (VLP).
  • the HPV antigen is an L1 protein (absent an L2 antigen) in the form of a VLP or capsomer structure.
  • Such antigens, virus like particles and capsomer are per se known. See, for example, WO94/00152, WO94/20137, WO94/05792, and WO93/02184.
  • a truncated L1 protein may be used in the invention, for example as disclosed in WO 96/11272.
  • a C-terminal truncation of L1 is used, for example a 34 amino acid C-terminal truncation of HPV 16, or an equivalent truncation from other HPV type.
  • a composition comprises a combination of HPV virus like particles or capsomers from HPV 16 and HPV 18, together with HPV 31 and/or HPV 45. In one aspect of the invention a composition comprises a combination of HPV virus like particles or capsomers from HPV 16 and HPV 18, together with HPV 33 and/or HPV 58. In one aspect of the invention a composition comprises a combination of HPV virus like particles or capsomers from HPV 16 and HPV 18, together with VLPs or capsomers from one or more cancer causing HPV types, such as one, two, three, four or all of HPV 31, 33, 45, 52, and 58.
  • the invention thus relates in one aspect to a composition
  • a composition comprising virus like particles from HPV 16, 18, 31 and 45 in combination with an adjuvant comprising 3D MPL and an oil in water emulsion as described herein.
  • a composition comprises a mixture of HPV 16, 18, 31, 33, 45, 52, and 58 L1-only virus like particles or capsomers.
  • L1 or L2 proteins may be provided in the form of fusion proteins.
  • compositions comprise L1 particles or capsomers, and fusion proteins comprising one or more antigens selected from the HPV 6 and HPV 11 proteins, for example E6, E7, L1, and L2.
  • HPV antigens from cancer types may be combined with antigens from genital warts types, such as HPV 16 and/or 18 with HPV 6 and/or 11.
  • a composition comprising HPV 16, 18, 6 and 11 is contemplated.
  • a combination of HPV 16, 18, 31, 33, 45, 52, and 58 L1-only virus like particle or capsomers may be used in combination with virus like particles or capsomers from HPV 6 and/or HPV 11.
  • early proteins such as E7, E2 or E5 for example may be included alone, in combinations, or may be fusion proteins; an embodiment of this includes a VLP comprising L1E7 fusion proteins (WO 96/11272).
  • the fusion protein is L2E7 as disclosed in WO 96/26277, or proteinD (1 ⁇ 3)-E7 disclosed in GB 9717953.5 (PCT/EP98/05285).
  • 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, such as a Protein D-E6/E7 fusion.
  • a composition may optionally contain either or both E6 and E7 proteins from HPV 18, such as in the form of a Protein D-E6 or Protein D-E7 fusion protein or Protein D E6/E7 fusion protein.
  • the adjuvant composition of the invention contains an oil-in-water emulsion adjuvant, preferably said emulsion comprises a metabolisable oil in an amount of 0.5% to 20% of the total volume, 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).
  • 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).
  • the metabolisable oil is present in an amount of 0.5% to 20% (final concentration) of the total volume of the immunogenic composition, preferably an amount of 1.0% to 10% of the total volume, preferably in an amount of 2.0% to 6.0% of the total volume.
  • the metabolisable oil is present in a final amount of about 0.5%, 1%, 3.5% or 5% of the total volume of the immunogenic composition. In another specific embodiment, the metabolisable oil is present in a final amount of 0.5%, 1%, 3.57% or 5% of the total volume of the immunogenic composition.
  • 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.
  • the oil in water emulsion comprises a sterol.
  • Sterols are well known in the art, for example cholesterol is well known and is, for example, disclosed in the Merck Index, 11th Edn., page 341, as a naturally occurring sterol found in animal fat.
  • Other suitable sterols include ⁇ -sitosterol, stigmasterol, ergosterol, alpha-tocopherol and ergocalciferol.
  • Said sterol is suitably present in an amount of 0.01% to 20% (w/v) of the total volume of the immunogenic composition, preferably at an amount of 0.1% to 5% (w/v).
  • the sterol when it is cholesterol, it is present in an amount of between 0.02% and 0.2% (w/v) of the total volume of the immunogenic composition, more preferably at an amount of 0.02% (w/v) in a 0.5 ml vaccine dose volume, or 0.07% (w/v) in 0.5 ml vaccine dose volume or 0.1% (w/v) in 0.7 ml vaccine dose volume.
  • the sterol is alpha-tocopherol or a derivative thereof such as alpha-tocopherol succinate (also known as vitamin E succinate).
  • alpha-tocopherol is present in an amount of between 0.2% and 5.0% (v/v) of the total volume of the immunogenic composition, more preferably at an amount of 2.5% (v/v) in a 0.5 ml vaccine dose volume, or 0.5% (v/v) in 0.5 ml vaccine dose volume or 1.7-1.9% (v/v), preferably 1.8% in 0.7 ml vaccine dose volume.
  • 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 comprise an emulsifying agent.
  • the emulsifying agent may be present at an amount of 0.01 to 5.0% by weight of the immunogenic composition (w/w), preferably present at an amount of 0.1 to 2.0% by weight (w/w). Preferred concentration are 0.5 to 1.5% by weight (w/w) of the total composition.
  • the emulsifying agent may suitably be polyoxyethylene sorbitan monooleate (Tween 80).
  • Tween 80 polyoxyethylene sorbitan monooleate
  • a 0.5 ml vaccine dose volume contains 1% (w/w) Tween 80
  • a 0.7 ml vaccine dose volume contains 0.7% (w/w) Tween 80.
  • the concentration of Tween 80 is 0.2% (w/w).
  • the oil in water emulsion adjuvant may be utilised with other adjuvants or immuno-stimulants and therefore an important embodiment of the invention is an oil in water formulation comprising squalene or another metabolisable oil, alpha tocopherol, and tween 80.
  • the oil in water emulsion may also contain span 85 and/or Lecithin.
  • the oil in water will comprise from 2 to 10% squalene of the total volume of the immunogenic composition, from 2 to 10% alpha tocopherol and from 0.3 to 3% Tween 80, and may be produced according to the procedure described in WO 95/17210.
  • the ratio of squalene: alpha tocopherol is equal or less than 1 as this provides a more stable emulsion.
  • Span 85 polyoxyethylene sorbitan trioleate
  • Span 85 polyoxyethylene sorbitan trioleate
  • composition comprise an additional adjuvant, 3 de-O-acylated monophosphoryl lipid A (3D-MPL).
  • 3D MPL is a TRL-4 ligand adjuvant, a non-toxic derivative of lipid A.
  • 3D-MPL is sold under the trademark MPL® by Corixa corporation and is referred throughout the document as MPL. It primarily promotes CD4+ T cell responses with an IFN- ⁇ (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 3D-MPL has a particle size such that it may be sterile-filtered through a 0.22 ⁇ m filter. Such preparations are described in WO 94/21292 and in Example II.
  • 3D-MPL can be used, for example, at an amount of 1 to 100 ⁇ g (w/v) per composition dose, preferably in an amount of 10 to 50 ⁇ g (w/v) per composition dose.
  • a suitable amount of 3D-MPL is for example any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 ⁇ g (w/v) per composition dose. More preferably, 3D-MPL amount ranges from 25 to 75 ⁇ g (w/v) per composition dose.
  • composition dose will be ranging from about 0.5 ml to about 1 ml.
  • a typical vaccine dose are 0.5 ml, 0.6 ml, 0.7 ml, 0.8 ml, 0.9 ml or 1 ml.
  • a final concentration of 50 ⁇ g of 3D-MPL is contained per ml of vaccine composition, or 25 ⁇ g per 0.5 ml vaccine dose.
  • a final concentration of 35.7 ⁇ g or 71.4 ⁇ g of 3D-MPL is contained per ml of vaccine composition.
  • a 0.5 ml vaccine dose volume contains 25 ⁇ g or 50 ⁇ g of 3D-MPL per dose.
  • the dose of MPL is suitably able to enhance an immune response to an antigen in a human.
  • a suitable MPL amount is that which improves the immunological potential of the composition compared to the unadjuvanted composition, or compared to the composition adjuvanted with another MPL amount, whilst being acceptable from a reactogenicity profile.
  • 3D MPL is a TRL-4 ligand, a non-toxic derivative of lipid A.
  • the present invention contemplates use of other suitable TLR-4 ligands in place of 3D-MPL, including lipopolysaccharide (LPS) and derivatives, MDP (muramyl dipeptide) and F protein of RSV.
  • LPS lipopolysaccharide
  • MDP muramyl dipeptide
  • F protein of RSV F protein of RSV.
  • Non-toxic derivatives of lipid A, particularly monophosphoryl lipid A are also contemplated.
  • Synthetic derivatives of lipid A are known, some being described as TLR-4 agonists, which might be suitable for use in the present invention and include, but are not limited to:
  • composition of the invention suitably induces an improved CD4 T-cell immune response against at least one of the component antigen(s) or antigenic composition compared to the CD4 T-cell immune response obtained with the corresponding composition which is un-adjuvanted, i.e. does not contain any exogeneous adjuvant (herein also referred to as ‘plain composition’) or at least lacks one of both components (either 3D-MPL or the oil-in-water emulsion adjuvant) of the adjuvant composition.
  • a higher CD4 T-cell immune response is meant that a higher CD4 response is obtained in a human patient after administration of the adjuvanted immunogenic composition than that obtained after administration of the same composition without adjuvant or lacking one of the two components of the adjuvant composition.
  • a higher CD4 T-cell response is obtained in a human patient upon administration of an immunogenic composition comprising a split influenza virus or split virus antigenic preparation thereof together with an oil-in-water emulsion adjuvant as herein defined, compared to the response induced after administration of an immunogenic composition comprising a split influenza virus or split virus antigenic preparation thereof.
  • Such formulation will advantageously be used to induce anti-influenza CD4-T cell response capable of detection of influenza epitopes presented by MHC class II molecules.
  • said immunological response induced by an adjuvanted split influenza composition of the present invention is higher than the immunological response induced by any other un-adjuvanted influenza conventional vaccine, such as sub-unit influenza vaccine or whole influenza virus vaccine.
  • said ‘improved CD4 T-cell immune response’ is suitably obtained in an immunologically “unprimed” patient, i.e. a patient who is seronegative to the antigen.
  • This seronegativity may be the result of said patient having never faced such an antigen (for example, not having been infected with a virus or bacteria containing said antigen—a so-called ‘naive’ patient) or, alternatively, having failed to respond to said antigen once encountered.
  • An improved CD4 T-cell immune response may be assessed by measuring the number of cells producing any of the following cytokines:
  • CD4 T-cell immune response when cells producing any of the above cytokines will be in a higher amount following administration of the adjuvanted composition compared to the administration of the un-adjuvanted composition. Typically at least one, preferably two of the five conditions mentioned herein above will be fulfilled. In a particular embodiment, the cells producing all four cytokines will be present at a higher amount in the adjuvanted group compared to the un-adjuvanted group.
  • An improved CD4 T-cell immune response conferred by an adjuvanted composition of the present invention may be ideally obtained after one single administration.
  • the single dose approach will be extremely relevant for example in a rapidly evolving outbreak situation.
  • the second dose of said same composition (still considered as ‘composition for first vaccination’) may be administered during the on-going primary immune response and is adequately spaced.
  • the second dose of the composition is given a few weeks, or about one month, e.g. 2 weeks, 3 weeks, 4 weeks, 5 weeks, or 6 weeks after the first dose, to help prime the immune system in unresponsive or poorly responsive individuals.
  • the present invention also relates to use of a composition
  • a composition comprising:
  • the invention also relates to a method of inducing an improved CD4 T-cell response against an antigen comprising delivery of a composition comprising:
  • the administration of said immunogenic composition alternatively or additionally induces an improved B-memory cell response in patients administered with the adjuvanted immunogenic composition compared to the B-memory cell response induced in individuals immunized with the un-adjuvanted composition or with a composition lacking one of the two components of the adjuvant composition.
  • An improved B-memory cell response is intended to mean an increased frequency of peripheral blood B lymphocytes capable of differentiation into antibody-secreting plasma cells upon antigen encounter as measured by stimulation of in-vitro differentiation (see Example sections, e.g. methods of Elispot B cells memory).
  • the vaccination with the composition for the first vaccination, adjuvanted has no measurable impact on the CD8 response.
  • said improved CD4 T-cell immune response is obtained in an immunocompromised subject such as an elderly individual, typically at least 50, 55, 60 or 65 years of age or above, or an adult younger than 55 years of age with a high risk medical condition (‘high risk’ adult), or a child under the age of two.
  • an immunocompromised subject such as an elderly individual, typically at least 50, 55, 60 or 65 years of age or above, or an adult younger than 55 years of age with a high risk medical condition (‘high risk’ adult), or a child under the age of two.
  • an oil-in-water emulsion adjuvant comprising a metabolisable oil, a sterol and an emulsifying agent, and 3D MPL is effective in promoting T cell responses in an immuno-compromised human population.
  • the administration of a single dose of the immunogenic composition for first vaccination, as described in the invention is capable of providing better sero-protection, as assessed by the correlates of protection for influenza vaccines, following re-vaccination against influenza in a human elderly population, than does the vaccination with an un-adjuvanted split vaccine.
  • the claimed adjuvanted formulation has also been able to induce an improved CD4 T-cell immune response against influenza virus compared to that obtained with the un-adjuvanted formulation.
  • This finding can be associated with an increased responsiveness upon vaccination or infection vis-á-vis influenza antigenic exposure.
  • this may also be associated with a cross-responsiveness, i.e. a higher ability to respond against variant influenza strains.
  • This improved response may be especially beneficial in an immuno-compromised human population such as the elderly population (eg 50, 55, 60, 65 years of age and above) and in particular the high risk elderly population. This may result in reducing the overall morbidity and mortality rate and preventing emergency admissions to hospital for pneumonia and other influenza-like illness.
  • the Inventors have also been capable of demonstrating that the claimed adjuvanted composition was able to not only induce but also maintain protective levels of antibodies against all three strains present in the vaccine, in more individuals than those obtained with the un-advanted composition (see Table 43 for example).
  • the claimed composition is capable of ensuring a persistent immune response against influenza related disease.
  • persistence it is meant an HI antibody immune response which is capable of meeting regulatory criteria after at least three months, preferably after at least 6 months after the vaccination.
  • the claimed composition is able to induce protective levels of antibodies in >70% of individuals, suitably in >80% of individuals or suitably in >90% of individuals for at least one influenza strain, preferably for all strains present in the vaccine, after at least three months.
  • protective levels of antibodies of >90% are obtained at least 6 months post-vaccination against at least one, suitably two, or all strains present in the vaccine composition.
  • the present invention also relates to the use of the composition of the invention in an immunocompromised human individual or population such as high risk adults or elderly, and in the manufacture of an immunogenic composition for vaccination of a human immuno-compromised individual or population, such as a high risk adult or a elderly population.
  • the CD4 T-cell immune response such as the improved CD4 T-cell immune response obtained in an unprimed subject, involves the induction of a cross-reactive CD4 T helper response.
  • the amount of cross-reactive CD4 T cells is increased.
  • cross-reactive CD4 response is meant CD4 T-cell targeting shared epitopes between influenza strains.
  • influenza vaccines are usually effective only against infecting strains of influenza virus that have haemaglutinins of similar antigenic characteristics.
  • the infecting (circulating) influenza virus has undergone minor changes (such as a point mutation or an accumulation of point mutations resulting in amino acid changes in the for example) in the surface glycoproteins in particular haemagglutinin (antigenic drift variant virus strain) the vaccine may still provide some protection, although it may only provide limited protection as the newly created variants may escape immunity induced by prior influenza infection or vaccination.
  • Antigenic drift is responsible for annual epidemics that occur during interpandemic periods (Wiley & Skehel, 1987, Ann. Rev. Biochem. 56, 365-394).
  • cross-reactive CD4 T cells provides an additional advantage to the composition of the invention, in that it may provide also cross-protection, in other words protection against heterologous infections, i.e. infections caused by a circulating influenza strain which is a variant (e.g. a drift) of the influenza strain contained in the immunogenic composition.
  • This may be advantageous when the circulating strain is difficult to propagate in eggs or to produce in tissue culture, rendering the use of a drifted strain a working alternative.
  • This may also be advantageous when the subject received a first and a second vaccination several months or a year apart, and the influenza strain in the immunogenic composition used for a second immunization is a drift variant strain of the strain used in the composition used for the first vaccination.
  • composition of the invention for protection against infections or disease caused by a pathogen which is a variant of the pathogen from which the antigen in the first composition is derived. Also provided issue of the composition of the invention for protection against infection or disease caused by a pathogen which comprises an antigen which is a variant of that in the composition of the invention.
  • Variant pathogens and/or antigens suitably have antigens with common CD4 T cell epitopes and/or B cell epitopes with the first pathogen or antigen, but which are not identical.
  • peripheral blood CD4 T-cells responding to antigenic strain preparation (whole virus or split antigen) that is homologous to the one present in the vaccine (H3N2: A/Panama/2007/99, H1N1: A/New Calcdonia/20/99, B: B/Shangdong/7/97) (see Example III).
  • H3N2 A/Panama/2007/99
  • H1N1 A/New Calcdonia/20/99
  • B B/Shangdong/7/97
  • a comparable increase in frequency can be seen if peripheral blood CD4 T-cells are restimulated with influenza strains classified as drifted strains (H3N2: A/Sydney/5/97, H1N1: A/Beijing/262/95, B: B/Yamanashi/166/98).
  • peripheral blood CD4 T-cells are restimulated with influenza strains classified as shift strains (H2N2: A/Singapore/1/57, H9N2: A/Hongkong/1073/99) by expert in the field, there is no observable increase following vaccination.
  • H2N2 A/Singapore/1/57
  • H9N2 A/Hongkong/1073/99
  • CD4 T-cells that are able to recognize both homologous and drifted Influenza strains have been named in the present document “cross-reactive”.
  • CD4 T-cell epitopes shared by different Influenza strains have been identified in human (Gelder C et al. 1998, Int Immunol. 10(2):211-22; Gelder C M et al. 1996 J. Virol. 70(7):4787-90; Gelder C M et al. 1995 J. Virol. 1995 69(12):7497-506).
  • the adjuvanted composition may offer the additional benefit of providing better protection against circulating strains which have undergone a major change (such as gene recombination for example, between two different species) in the haemagglutinin (antigenic shift) against which currently available vaccines have no efficacy.
  • composition may comprise additional adjuvants, suitably such as TRL-4 ligand adjuvants or a non-toxic derivative of lipid A.
  • TRL-4 ligands are lipopolysaccharide (LPS) and derivatives, MDP (muramyl dipeptide) and F protein of RSV.
  • Synthetic derivatives of lipid A are known, some being described as TLR-4 agonists, and include, but are not limited to:
  • TLR-4 ligands are, for example, lipopolysaccharide and its derivatives, muramyl dipeptide (MDP) or F protein of respiratory syncitial virus.
  • MDP muramyl dipeptide
  • Quil A is a saponin preparation isolated from the South American tree Quilaja Saponaria Molina and was first described by Dalsgaard et al. in 1974 (“Saponin adjuvants”, Archiv. für dienati Virusforschung, Vol. 44, Springer Verlag, Berlin, p243-254) to have adjuvant activity. 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.
  • compositions of QS21 have been described which are particularly preferred, these formulations further comprise a sterol (WO96/33739).
  • the saponins forming part of the present invention may be in the form of an oil in water emulsion (WO 95/17210).
  • An aspect of the present invention provides the use of an antigen in the manufacture of an immunogenic composition for revaccination of humans previously vaccinated with the antigen or fragment or variant thereof with 3D MPL and an oil-in-water emulsion adjuvant as herein defined.
  • revaccination is made at least 6 months after the first vaccination(s), preferably 8 to 14 months after, more preferably at around 10 to 12 months after.
  • an antigen (2) an oil-in-water emulsion adjuvant in the manufacture of an immunogenic composition for revaccination of humans previously vaccinated with the antigen, or fragment or variant thereof, 3D MPL and an oil-in-water emulsion adjuvant as herein defined.
  • the invention provides for the use of:
  • the composition for revaccination suitably shares common CD4 T-cell epitopes with the composition used for the first vaccination. In this respect it is a considered a variant of the antigen used in first vaccination.
  • the immunogenic composition for re-vaccination may contain the same type of antigen preparation—eg subunit/split/whole inactivated virus—as the immonogenic composition used for the first vaccination.
  • the boosting composition may contain another type of antigen preparation,
  • influenza the first vaccination may be with a split preparation and the booster vaccination with another inactivated influenza antigen, such as inactivated whole virus or purified HA and NA (subunit vaccine).
  • the booster composition may be adjuvanted or un-adjuvanted.
  • influenza the un-adjuvanted booster composition may be FluarixTM/ ⁇ -Rix® given intramuscularly.
  • the formulation contains three inactivated split virion antigens prepared from the WHO recommended strains of the appropriate influenza season.
  • a variant may be an antigen which shares common CD4 T-cell epitopes (generally considered as antigenic determinants recognized and bound by the T-cell receptor) with the antigenic composition used for the first vaccination, but which is not identical to that antigenic composition.
  • a variant may be an antigen which shares common B-cell epitopes (generally considered as antigenic determinants recognized and bound by the B-cell receptor) with the antigenic composition used for the first vaccination, but which is not identical to that antigenic composition.
  • T cell and B cell epitopes may be predicted using techniques well known in the art or inferred from immune responses using techniques as described herein.
  • Said oil-in-water emulsion adjuvant preferably comprises at least one metabolisable oil in an amount of 0.5% to 20% of the total volume, and has oil droplets of which at least 70% by intensity have diameters of less than 1 ⁇ m.
  • the immunogenic composition for revaccination (also called herein below the ‘boosting composition’) contains a split influenza virus or split virus antigenic preparation thereof which shares a common CD4 T-cell epitope with the split influenza virus or split virus antigenic preparation thereof used for the first vaccination.
  • a ‘common CD4 T cell epitope is intended to mean peptides/sequences/epitopes from different antigens which can be recognised by the same CD4 cell (see examples of described epitopes in: Gelder C et al. 1998, Int Immunol. 10(2):211-22; Gelder C M et al. 1996 J. Virol. 70(7):4787-90; Gelder C M et al. 1995 J. Virol. 1995 69(12):7497-506).
  • influenza strain may be associated with a pandemic outbreak or have the potential to be associated with a pandemic outbreak.
  • the vaccine is a multivalent vaccine such as a bivalent or a trivalent vaccine
  • at least one strain is associated with a pandemic outbreak or has the potential to be associated with a pandemic outbreak.
  • Suitable strains are, but not limited to: H5N1, H9N2, H7N7, H2N2 and H1N1.
  • a booster composition where used, is given at the next season, e.g. approximately one year after the first immunogenic composition.
  • the booster composition may also be given every subsequent year (third, fourth, fifth vaccination and so forth).
  • the boosting composition may be the same as the first composition.
  • the boosting composition contains an strain or antigenic preparation therefrom which is a variant of the strain or antigen used for the first vaccination.
  • the influenza antigen or antigenic composition used in revaccination preferably comprises an adjuvant or an oil-in-water emulsion, suitably as described above.
  • the adjuvant may be an oil-in-water emulsion adjuvant as herein above described, which is preferred, optionally containing an additional adjuvant such as TLR-4 ligand such as 3D-MPL or a saponin, or may be another suitable adjuvant such as alum for example.
  • re-vaccination suitably induces any, preferably two or all, of the following: (i) an improved CD4 response against the influenza virus or antigenic preparation thereof, or (ii) an improved B cell memory response or (iii) an improved humoral response, compared to the equivalent response induced after a first vaccination with the un-adjuvanted split influenza virus or split virus antigenic preparation thereof.
  • the immunological responses induced after re-vaccination with the adjuvanted split influenza virus or split virus antigenic preparation thereof as herein defined are higher than the corresponding response induced after the re-vaccination with the un-adjuvanted composition.
  • the immunological responses induced after re-vaccination with an un-adjuvanted, preferably split, influenza virus are higher in the population first vaccinated with the adjuvanted split influenza composition than the corresponding response in the population first vaccinated with the un-adjuvanted split influenza composition.
  • the adjuvanted composition of the invention is capable of inducing a better protection against drifted strain (the influenza strain from the next influenza season) compared to the protection conferred by the control vaccine.
  • compositions comprising a split influenza virus or split virus antigenic preparation thereof
  • the composition is suitably monovalent or multivalent such as bivalent or trivalent or quadrivalent.
  • the split influenza virus or split virus antigenic preparation thereof is trivalent or quadrivalent, having an antigen from three different influenza strains.
  • At least one strain is associated with a pandemic outbreak or has the potential to be associated with a pandemic outbreak.
  • influenza viruses circulate that are related to those from the preceding epidemic.
  • the viruses spread among people with varying levels of immunity from infections earlier in life.
  • Such circulation over a period of usually 2-3 years, promotes the selection of new strains that have changed enough to cause an epidemic again among the general population; this process is termed ‘antigenic drift’.
  • drift variants may have different impacts in different communities, regions, countries or continents in any one year, although over several years their overall impact is often similar.
  • an influenza pandemics occurs when a new influenza virus appears against which the human population has no immunity.
  • Typical influenza epidemics cause increases in incidence of pneumonia and lower respiratory disease as witnessed by increased rates of hospitalisation or mortality.
  • the elderly or those with underlying chronic diseases are most likely to experience such complications, but young infants also may suffer severe disease.
  • novel influenza viruses emerge with a key surface antigen, the haemagglutinin, of a totally different subtype from strains circulating the season before.
  • the resulting antigens can vary from 20% to 50% from the corresponding protein of strains that were previously circulating in humans. This can result in virus escaping ‘herd immunity’ and establishing pandemics. This phenomenon is called ‘antigenic shift’. It is thought that at least in the past pandemics have occurred when an influenza virus from a different species, such as an avian or a porcine influenza virus, has crossed the species barrier. If such viruses have the potential to spread from person to person, they may spread worldwide within a few months to a year, resulting in a pandemic.
  • viruses of the H2N2 subtype replaced H1N1 viruses that had been circulating in the human population since at least 1918 when the virus was first isolated.
  • the H2 HA and N2 NA underwent antigenic drift between 1957 and 1968 until the HA was replaced in 1968 (Hong-Kong Flu pandemic) by the emergence of the H3N2 influenza subtype, after which the N2 NA continued to drift along with the H3 HA (Nakajima et al., 1991, Epidemiol. Infect. 106, 383-395).
  • an influenza virus strain that give it the potential to cause a pandemic outbreak are: it contains a new haemagglutinin compared to the haemagglutinin in the currently circulating strains, which may or not be accompanied by a change in neuraminidase subtype; it is capable of being transmitted horizontally in the human population; and it is pathogenic for humans.
  • a new haemagglutinin may be one which has not been evident in the human population for an extended period of time, probably a number of decades, such as H2. Or it may be a haemagglutinin that has not been circulating in the human population before, for example H5, H9, H7 or H6 which are found in birds. In either case the majority, or at least a large proportion of, or even the entire population has not previously encountered the antigen and is immunologically na ⁇ ve to it.
  • Certain parties are generally at an increased risk of becoming infected with influenza in a pandemic situation.
  • the elderly, the chronically ill and small children are particularly susceptible but many young and apparently healthy people are also at risk.
  • H2 influenza the part of the population born after 1968 is at an increased risk. It is important for these groups to be protected effectively as soon as possible and in a simple way.
  • Suitable strains are, but not limited to: H5N1, H9N2, H7N7, H2N2 and H1N1.
  • composition may contain more than three valencies, for example two non pandemic strains plus a pandemic strain.
  • the composition may contain three pandemic strains.
  • the invention relates to a vaccination regime in which the first vaccination is made with a split influenza composition containing at least one influenza strain that could potentially cause a pandemic outbreak and the re-vaccination is made with a circulating strain, either a pandemic strain or a classical strain.
  • This antigenic drift mainly resides in epitope regions of the viral surface proteins haemagglutinin (HA) and neuraminidase (NA). It is known that any difference in CD4 and B cell epitopes between different influenza strains, being used by the virus to evade the adaptive response of the host immune system, will play a major role in influenza vaccination and is.
  • HA haemagglutinin
  • NA neuraminidase
  • CD4 T-cell epitopes shared by different Influenza strains have been identified in human (see for example: Gelder C et al. 1998, Int Immunol. 10(2):211-22; Gelder C M et al. 1996 J Virol. 70(7):4787-90; and Gelder C M et al. 1995 J Virol. 1995 69(12):7497-506).
  • the re-vaccination is made by using a booster composition which contain an influenza virus or antigenic preparation thereof which shares common CD4 T-cell epitopes with the split influenza virus antigen or split virus antigenic preparation thereof used for the first vaccination.
  • the invention thus relates to the use of the immunogenic composition comprising a split influenza virus or split virus antigenic preparation thereof, an oil-in-water emulsion adjuvant as herein defined and 3D MPL in the manufacture of a first vaccination-component of a multi-dose vaccine, the multi-dose vaccine further comprising, as a boosting dose, an influenza virus or antigenic preparation thereof which shares common CD4 T-cell epitopes with the split influenza virus antigen or split virus antigenic preparation thereof of the dose given at the first vaccination.
  • composition of the invention may be administered by any suitable delivery route, such as intradermal, mucosal e.g. intranasal, oral, intramuscular or subcutaneous.
  • suitable delivery route such as intradermal, mucosal e.g. intranasal, oral, intramuscular or subcutaneous.
  • Other delivery routes are well known in the art.
  • the intramuscular delivery route is preferred for the adjuvanted influenza composition.
  • Intradermal delivery is another suitable route.
  • Any suitable device may be used for intradermal delivery, for example short needle devices such as those described in U.S. Pat. No. 4,886,499, U.S. Pat. No. 5,190,521, U.S. Pat. No. 5,328,483, U.S. Pat. No. 5,527,288, U.S. Pat. No. 4,270,537, U.S. Pat. No. 5,015,235, U.S. Pat. No. 5,141,496, U.S. Pat. No. 5,417,662.
  • Intradermal vaccines may also be administered by devices which limit the effective penetration length of a needle into the skin, such as those described in WO99/34850 and EP1092444, incorporated herein by reference, and functional equivalents thereof.
  • jet injection devices which deliver liquid vaccines to the dermis via a liquid jet injector or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis. Jet injection devices are described for example in U.S. Pat. No. 5,480,381, U.S. Pat. No. 5,599,302, U.S. Pat. No. 5,334,144, U.S. Pat. No. 5,993,412, U.S. Pat. No. 5,649,912, U.S. Pat. No.
  • Another suitable administration route is the subcutaneous route.
  • Any suitable device may be used for subcutaneous delivery, for example classical needle.
  • a needle-free jet injector service is used, such as that published in WO 01/05453, WO 01/05452, WO 01/05451, WO 01/32243, WO 01/41840, WO 01/41839, WO 01/47585, WO 01/56637, WO 01/58512, WO 01/64269, WO 01/78810, WO 01/91835, WO 01/97884, WO 02/09796, WO 02/34317. More preferably said device is pre-filled with the liquid vaccine formulation.
  • the vaccine is administered intranasally.
  • the vaccine is administered locally to the nasopharyngeal area, preferably without being inhaled into the lungs. It is desirable to use an intranasal delivery device which delivers the vaccine formulation to the nasopharyngeal area, without or substantially without it entering the lungs.
  • Preferred devices for intranasal administration of the vaccines according to the invention are spray devices.
  • Suitable commercially available nasal spray devices include AccusprayTM (Becton Dickinson).
  • Nebulisers produce a very fine spray which can be easily inhaled into the lungs and therefore does not efficiently reach the nasal mucosa. Nebulisers are therefore not preferred.
  • Preferred spray devices for intranasal use are devices for which the performance of the device is not dependent upon the pressure applied by the user. These devices are known as pressure threshold devices. Liquid is released from the nozzle only when a threshold pressure is applied. These devices make it easier to achieve a spray with a regular droplet size.
  • Pressure threshold devices suitable for use with the present invention are known in the art and are described for example in WO 91/13281 and EP 311 863 B and EP 516 636, incorporated herein by reference. Such devices are commercially available from Pfeiffer GmbH and are also described in Bommer, R. Pharmaceutical Technology Europe, September 1999.
  • Preferred intranasal devices produce droplets (measured using water as the liquid) in the range 1 to 200 ⁇ m, preferably 10 to 120 ⁇ m. Below 10 ⁇ m there is a risk of inhalation, therefore it is desirable to have no more than about 5% of droplets below 10 ⁇ m. Droplets above 120 ⁇ m do not spread as well as smaller droplets, so it is desirable to have no more than about 5% of droplets exceeding 120 ⁇ m.
  • Bi-dose delivery is a further preferred feature of an intranasal delivery system for use with the vaccines according to the invention.
  • Bi-dose devices contain two sub-doses of a single vaccine dose, one sub-dose for administration to each nostril. Generally, the two sub-doses are present in a single chamber and the construction of the device allows the efficient delivery of a single sub-dose at a time. Alternatively, a monodose device may be used for administering the vaccines according to the invention.
  • epidermal or transdermal vaccination route is also contemplated in the present invention.
  • the adjuvanted immunogenic composition for the first administration may be given intramuscularly, and the booster composition, either adjuvanted or not, may be administered through a different route, for example intradermal, subcutaneous or intranasal.
  • the composition for the first administration may contain a standard HA content of 15 ⁇ g per influenza strain, and the booster composition may contain a low dose of HA, i.e. below 15 ⁇ g, and depending on the administration route, may be given in a smaller volume.
  • the target population is preferably a human population or individual.
  • the target population to vaccinate may be immuno-compromised human.
  • Immuno-compromised humans generally are less well able to respond to an antigen, in particular to an influenza antigen, in comparison to healthy adults.
  • the target population is a population which is unprimed against influenza, either being na ⁇ ve (such as vis à vis a pandemic strain), or having failed to respond previously to infection or vaccination.
  • the target population is elderly persons suitably aged 55 years and over, younger high-risk adults (i.e. between 18 and 54 years of age) such as people working in health institutions, or those young adults with a risk factor such as cardiovascular and pulmonary disease, or diabetes.
  • Another target population is all children 6 months of age and over, especially children 6-23 months of age who experience a relatively high influenza-related hospitalization rate.
  • the target population is elderly above 65 years of age.
  • each vaccine dose is selected as an amount which induces an immunoprotective response without significant, adverse side effects in typical vaccines. Such amount will vary depending upon which specific immunogen is employed and how it is presented. Generally, it is expected that each dose will comprise 0.1-100 ⁇ g of saccharide, preferably 0.1-50 ⁇ g, preferably 0.1-10 ⁇ g, of which 1 to 5 ⁇ g is the most preferable range.
  • 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.
  • Optimal amounts of components for a particular vaccine can be ascertained by standard studies involving observation of appropriate immune responses in subjects. Following an initial vaccination, subjects may receive one or several booster immunisations 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 immunogenic compositions according to the present invention are a standard 0.5 ml injectable dose in most cases, and contains 15 ⁇ g of haemagglutinin antigen component from the or each influenza strain, as measured by single radial immunodiffusion (SRD) (J. M. Wood et al.: J. Biol. Stand. 5 (1977) 237-247; J. M. Wood et al., J. Biol. Stand. 9 (1981) 317-330).
  • the vaccine dose volume will be between 0.5 ml and 1 ml, in particular a standard 0.5 ml, or 0.7 ml vaccine dose volume. Slight adaptation of the dose volume will be made routinely depending on the HA concentration in the original bulk sample.
  • said immunogenic composition contains a low dose of HA antigen—e.g any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 ⁇ g of HA per influenza strain.
  • a suitable low dose of HA is between 1 to 7.5 ⁇ g of HA per influenza strain, suitably between 3.5 to 5 ⁇ g such as 3.75 ⁇ g of HA per influenza strain, typically about 5 ⁇ g of HA per influenza strain.
  • a vaccine dose according to the invention in particular a low dose vaccine, may be provided in a smaller volume than the conventional injected split flu vaccines, which are generally around 0.5, 0.7 or 1 ml per dose.
  • the low volume doses according to the invention are preferably below 500 ⁇ l, more preferably below 300 ⁇ l and most preferably not more than about 200 ⁇ l or less per dose.
  • a preferred low volume vaccine dose is a dose with a low antigen dose in a low volume, e.g. about 15 ⁇ g or about 7.5 ⁇ g HA or about 3.0 ⁇ g HA (per strain) in a volume of about 200 ⁇ l.
  • the influenza medicament of the invention preferably meets certain international criteria for vaccines.
  • At least two or all three of the criteria will need to be met for all strains, particularly for a new vaccine such as a new vaccine for delivery via a different route. Under some circumstances two criteria may be sufficient. For example, it may be acceptable for two of the three criteria to be met by all strains while the third criterion is met by some but not all strains (e.g. two out of three strains). The requirements are different for adult populations (18-60 years) and elderly populations (>60 years).
  • the invention provides a method of designing a vaccine for diseases known to be cured or treated through a CD4+ T cell activation, comprising
  • Example I describes immunological read-out methods used in mice, ferret and human studies.
  • Example II describes the preparation and characterization of the oil in water emulsion and adjuvant formulations used in the studies exemplified.
  • Example III describes a clinical trial in an elderly population aged over 65 years with a vaccine containing a split influenza antigen preparation and AS03 adjuvant
  • Example IV describes a second clinical trial—revaccination trial—in an elderly population aged over 65 years with a vaccine containing a split influenza antigen preparation and AS03 adjuvant.
  • Example V shows a pre-clinical evaluation of adjuvanted and un-adjuvanted influenza vaccines in ferrets (study I and study II). The temperature monitoring, viral shedding and CD4 T-cell response were measured.
  • Example VI shows a pre-clinical evaluation of adjuvanted and un-adjuvanted influenza vaccines in C57BI/6 na ⁇ ve and primed mice.
  • Example VII shows a pre-clinical evaluation of adjuvanted and un-adjuvanted split and sub-unit influenza vaccines in C57BI/6 mice primed with heterologous strains.
  • Example VIII describes a clinical trial in an elderly population aged over 65 years with a vaccine containing a split influenza antigen preparation containing AS03 adjuvant, AS03+MPL adjuvant, or no exogeneous adjuvant.
  • Example IX shows a pre-clinical evaluation of adjuvanted and un-adjuvanted influenza vaccines in ferrets (study III). The temperature monitoring, viral shedding and HI titers were measured.
  • Example X shows a clinical trial in an elderly population aged over 65 years with a vaccine containing a split influenza antigen preparation containing AS03 with or without MPL adjuvant: immunogenicity persistence data at day 90 and day 180.
  • Example XI shows a clinical trial in an elderly population aged over 65 years with a vaccine containing a split influenza antigen preparation containing AS03 with MPL adjuvant.
  • Example XII shows a clinical trial in an elderly population aged over 65 years with a vaccine containing a split influenza antigen preparation containing AS03 with MPL adjuvant at two concentrations.
  • Example XIII shows a pre-clinical evaluation of two adjuvanted HPV vaccines in mice. Antibody and B cell memory responses were measured.
  • 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). Heat inactivated sera were previously treated by Kaolin and chicken RBC to remove non-specific inhibitors. After pretreatment, 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 1:20, an undetectable level was scored as a titer equal to 10.
  • 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
  • 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.
  • 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.
  • 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.
  • Tween 80 is dissolved in phosphate buffered saline (PBS) to give a 2% solution in the PBS.
  • PBS phosphate buffered saline
  • To provide 100 ml two-fold concentrate emulsion 5 g of DL alpha tocopherol and 5 ml of squalene are vortexed to mix thoroughly. 90 ml of PBS/Tween solution is added and mixed thoroughly.
  • the resulting emulsion is then passed through a syringe and finally microfluidised by using an M110S microfluidics machine.
  • the resulting oil droplets have a size of approximately 120-180 nm (expressed as Z average measured by PCS).
  • the other adjuvants/antigen components are added to the emulsion in simple admixture.
  • the preparation of the SB62 emulsion is made by mixing under strong agitation of an oil phase composed of hydrophobic components ( ⁇ -tocopherol and squalene) and an aqueous phase containing the water soluble components (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) 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
  • 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.
  • the size of the diameter of the oil droplets is determined according to the following procedure and under the following experimental conditions.
  • the droplet size measure is given as an intensity measure and expressed as z average measured by PCS.
  • PL-Nanocal Particle size standards 100 nm (cat n° 6011-1015) was diluted in 10 mM NaCl.
  • the Automatic Malvern algorithm uses a combination of cumulants, Contin and non negative least squares (NNLS) algorithms.
  • the intensity distribution may be converted into volume distribution thanks to the Mie theory.
  • the z-average diameter (ZAD) size is weighed by the amount of light scattered by each size of particles in the sample. This value is related to a monomodal analysis of the sample and is mainly used for reproducibility purposes.
  • the count rate (CR) is a measure of scattered light: it corresponds to thousands of photons per second.
  • the polydispersity (Poly) index is the width of the distribution. This is a dimensionless measure of the distribution broadness.
  • FIG. 1 A schematic representation of these results is shown in FIG. 1 for formulation 1023.
  • the great majority of the particles e.g. at least 80%
  • SB62 formulation was measured at different dilutions with the Malvern Zetasizer 3000HS and two optical models.
  • the particle size ZAD i.e. intensity mean by cumulant analysis
  • MPL (as used throughout the document it is an abbreviation for 3D-MPL, i.e. 3-O-deacylated monophosphoryl lipid A) liquid bulk is prepared from MPL® lyophilized powder.
  • MPL liquid bulk is a stable concentrated (around 1 mg/ml) aqueous dispersion of the raw material, which is ready-to-use for vaccine or adjuvant formulation.
  • FIG. 2 A schematic representation of the preparation process is given in FIG. 2 .
  • MPL liquid bulk preparation is carried over in sterile glass containers.
  • the dispersion of MPL consists of the following steps:
  • MPL powder is lyophilized by microfluidisation resulting in a stable colloidal aqueous dispersion (MPL particle size smaller than 200 nm).
  • the MPL lyophilized powder is dispersed in water for injection in order to obtain a coarse 10 mg/ml suspension.
  • the suspension then undergoes a thermal treatment under stirring. After cooling to room temperature, the microfluidization process is started in order to decrease the particle size.
  • Microfluidization is conducted using Microfluidics apparatus M110EH, by continuously circulating the dispersion through a microfluidization interaction chamber, at a defined pressure for a minimum amount of passages (number of cycles: n min ).
  • the microfluidization duration representing the number of cycles, is calculated on basis of the measured flow rate and the dispersion volume.
  • the resulting flow rate may vary from one interaction chamber to another, and throughout the lifecycle of a particular interaction chamber.
  • the interaction chamber used is of the type F20Y Microfluidics.
  • the processing time may vary from one batch to another.
  • the time required for 1 cycle is calculated on basis of the flow rate.
  • the flow rate to be considered is the flow rate measured with water for injection just before introduction of MPL into the apparatus.
  • One cycle is defined as the time (in minutes) needed for the total volume of MPL to pass once through the apparatus.
  • the time needed to obtain n cycles is calculated as follows:
  • the number of cycles is thus adapted accordingly.
  • Minimum amount of cycles to perform (n min ) are described for the preferred equipment and interaction chambers used.
  • the total amount of cycles to run is determined by the result of a particle size measurement performed after n min cycles.
  • a particle size limit (d lim ) is defined, based on historical data. The measurement is realized by photon correlation spectroscopy (PCS) technique, and d lim is expressed as an unimodal result (Z average ). Under this limit, the microfluidization can be stopped after n min cycles. Above this limit, microfluidization is continued until satisfactory size reduction is obtained, for maximum another 50 cycles.
  • the dispersed MPL is stored at +2 to +8° C. awaiting transfer to the filtration area.
  • the dispersion is diluted with water for injection, and sterile filtered through a 0.22 ⁇ m filter under laminal flow.
  • the final MPL concentration is 1 mg/ml (0.80-1.20 mg/ml).
  • MPL is added at a final concentration of between 10 and 50 ⁇ g per vaccine dose.
  • PBS 10 fold concentrated (pH 7.4 when one fold concentrated) as well as a SB62 mixture containing Tween, Triton X-100 and VES (vitamin E succinate, i.e. alpha-tocopherol succinate) is added to water for injection.
  • the quantities take into account the detergent present in the influenza strains so as to reach a target final concentration of 750 ⁇ g/ml Tween 80, 110 ⁇ g/ml Triton X-100 and 100 ⁇ g/ml VES.
  • 15 ⁇ g of each influenza strain of interest for example strain H1N1, H3N2 and B in a classical tri-valent vaccine
  • 250 ⁇ l of SB62 emulsion is added and then 25 ⁇ g or 50 ⁇ g of MPL.
  • the same formulation can be prepared from a 2 vials approach by mixing 2 fold concentrated antigen or antigenic preparation with the AS03 (SB62 250 ⁇ l) or the AS03+MPL (SB62 250 ⁇ g+25 ⁇ g or 50 ⁇ g MPL) adjuvant. In this instance it is proceeded as follows.
  • the manufacturing of the AS25-adjuvanted influenza vaccine consists of three main steps:
  • the volumes of the three monovalent bulks are based on the HA content measured in each monovalent bulk prior to the formulation and on a target volume of 1100 ml.
  • Concentrated phosphate buffered saline and a pre-mixture of Tween 80, Triton X-100 and ⁇ -tocopheryl hydrogen succinate are diluted in water for injection.
  • the three concentrated monobulks (A/New Calcdonia, A/New York, B/Jiangsu) are then successively diluted in the resulting phosphate buffered saline/Tween 80-Triton X-100- ⁇ -tocopheryl hydrogen succinate solution (pH 7.4, 137 mM NaCl, 2.7 mM KCl, 8.1 mM Na2HPO4, 1.47 mM KH2PO4, 990 ⁇ g/ml Tween 80, 150 ⁇ g/ml Triton X-100 and 130 ⁇ g/ml ⁇ -tocopheryl hydrogen succinate) in order to have a final concentration of 39.47 ⁇ g HA of A strains (H1N1, H3N2) per ml of trivalent final bulk (15 ⁇ g HA/A strain/380 ⁇ l trivalent final bulk) and 46 ⁇ g HA of B strain (17.5 ⁇ g HA/B strain/380 ⁇ l trivalent final bulk). Between addition of each
  • the trivalent final bulk of antigens is aseptically filled into 3-ml sterile Type I (Ph. Eur.) glass vials. Each vial contains a volume of 470 ⁇ l (380 ⁇ l+90 ⁇ l overfill).
  • the adjuvant AS03/MPL is prepared by mixing of two components: SB62 emulsion (method in section II.1.2) and MPL (method in section II.3.1).
  • One-fold concentrated PBS mod prepared by diluting 10 ⁇ concentrated PBS mod in water for injection
  • SB62 bulk and MPL liquid bulk at 1 mg/ml.
  • MPL concentration will be determined so as to reach a final content of between 10 to 50 ⁇ g, suitably around 25 ⁇ g per final human vaccine dose.
  • the mixture is stirred for 5-30 minutes at room temperature, and the pH is adjusted to 6.8 ⁇ 0.1 with NAOH (0.05 or 0.5 M)/HCl (0.03 M or 0.3 M).
  • the mixture is sterilised by filtration through a 0.22 ⁇ m membrane. Sterile inert gas (nitrogen) flushing is performed to produce inert head space in the filled containers during minimum 1 minute.
  • the sterile AS03+MPL adjuvant is stored at +2-8° C. until aseptical filling into 1.25-ml sterile Type I (Ph. Eur.) glass syringes. Each syringe contains a volume overage of 80 ⁇ l (320 ⁇ l+80 ⁇ l overfill).
  • the content of the prefilled syringe containing the adjuvant is injected into the vial that contains the concentrated trivalent inactivated split virion antigens. After mixing the content is withdrawn into the syringe and the needle is replaced by an intramuscular needle.
  • One dose of the reconstituted the AS25-adjuvanted influenza candidate vaccine corresponds to 0.7 mL.
  • a phase I, open, randomised study was conducted in an elderly population aged over 65 years in 2003 in order to evaluate the reactogenicity and the immunogenicity of GlaxoSmithKline Biologicals influenza candidate vaccine containing the adjuvant AS03.
  • the humoral immune response i.e. anti-hemagglutinin, neutralising and anti-neuraminidase antibody titres
  • cell mediated immune response CD4 and/or CD8 T cell responses
  • Vaccination schedule one injection of influenza vaccine at day 0, blood sample collection, read-out analysis at day 21 (HI antibody determination, NI antibody determination, determination of neutralising antibodies, and CMI analysis) and study conclusion.
  • the standard trivalent split influenza vaccine—FluarixTM used in this study, is a commercial vaccine from the year 2003 developed and manufactured by GlaxoSmithKline Biologicals.
  • the AS03-adjuvanted influenza vaccine candidate is a 2 components vaccine consisting of a concentrated trivalent inactivated split virion antigens presented in a type I glass vial (335 ⁇ l) (antigen container) and of a pre-filled type I glass syringe containing the SB62 emulsion (335 ⁇ l) (adjuvant container).
  • the content of the antigen container is removed from the with the help of the SB62 emulsion pre-filled syringe, followed by gently mixing of the syringe. Mixing of the SB62 emulsion with the vaccine antigens reconstitute the AS03 adjuvant.
  • the used needle Prior to injection, the used needle is replaced by an intramuscular needle and the volume is corrected to 500 ⁇ l.
  • One dose of the reconstituted AS03-adjuvanted influenza vaccine corresponds to 0.5 ml, contains 15 ⁇ g HA of each influenza virus strain as in the registered FluarixTM/ ⁇ -Rix® vaccine and contains 10.68 mg squalene, 11.86 mg DL-alpha tocopherol, and 4.85 mg polysorbate 80 (Tween 80).
  • the manufacturing of the AS03-adjuvanted influenza vaccine consists of three main steps:
  • the volumes of the three monovalent bulks are based on the HA content measured in each monovalent bulk prior to the formulation and on a target volume of 800 ml.
  • Concentrated phosphate buffered saline and a pre-mixture of Tween 80, Triton X-100 and ⁇ -tocopheryl hydrogen succinate are diluted in water for injection.
  • the three concentrated monobulks (strain A/New Calcdonia-, strain A/Panama- and strain B/Shangdong-) are then successively diluted in the resulting phosphate buffered saline/Tween 80-Triton X-100- ⁇ -tocopheryl hydrogen succinate solution (pH 7.4, 137 mM NaCl, 2.7 mM KCl, 8.1 mM Na 2 HPO 4 , 1.47 mM KH 2 PO 4 , 1500 ⁇ g/ml Tween 80, 220 ⁇ g/ml Triton X-100 and 200 ⁇ g/ml ⁇ -tocopheryl hydrogen succinate) in order to have a final concentration of 60 ⁇ g HA of A strains per ml of trivalent final bulk (15 ⁇ g HA/A strain/250 ⁇ l trivalent final bulk) and 70 ⁇ g HA of B strain (17.5 ⁇ g HA/B strain/250 ⁇ l trivalent final bulk). Between addition of
  • the trivalent final bulk of antigens is aseptically filled into 3-ml sterile Type I glass vials. Each vial contains a 34% volume overage (335 ⁇ l total volume).
  • Emulsification the resulting mixture is subjected to shear, impact and cavitation forces in the interaction chamber of a microfluidizer (15000 PSI ⁇ 8 cycles) to produce submicron droplets (distribution between 100 and 200 nm).
  • the resulting pH is between 6.8 ⁇ 0.1.
  • the final composition of the SB62 emulsion is as follows:
  • the sterile SB62 bulk emulsion is then aseptically filled into 1.25-ml sterile Type I glass syringes. Each syringe contains a 34% volume overage (335 ⁇ l total volume).
  • the content of the vial containing the concentrated trivalent inactivated split virion antigens is removed from the vial with the help the syringe containing the SB62 emulsion followed by gently mixing of the syringe. Mixing of the SB62 emulsion with the vaccine antigens reconstitutes the AS03 adjuvant.
  • the vaccines were administered intramuscularly in the deltoid region of the non-dominant arm.
  • the vaccinees were observed closely for at least 30 minutes, with appropriate medical treatment readily available in case of a rare anaphylactic reaction following the administration of vaccine.
  • the mean age of the total vaccinated cohort at the time of vaccination was 71.8 years with a standard deviation of 6.0 years.
  • influenza vaccine adjuvanted with AS03 was safe and clinically well tolerated in the study population, i.e. elderly people aged over 65 years.
  • the GMTs for HI antibodies with 95% CI are shown in Table 7 (GMT for anti-HI antibody).
  • Pre-vaccination GMTs of antibodies for all vaccine strains were within the same range in the three groups. After vaccination, anti-haemagglutinin antibody levels increased significantly. Post vaccination, there was a trend for higher GMTs of HI antibody for all three vaccine strains in the FluAS03 and Fluarix groups although there was some overlap of 95% CI between the Fluarix group and the FluWVV group.
  • the conversion factors represent the fold increase in serum HI GMTs for each vaccine strain on day 21 compared to day 0.
  • the conversion factor varies from 6.1 to 13.6 according to the virus strain and the vaccine. This conversion factor is largely superior to the 2.0 fold increase in GMT required by the European authorities.
  • the seroprotection rates represent the proportion of subjects with a serum HI titre ⁇ 40 on day 21. At the outset of the study, half of the subjects (range 34.0%-69.4%) in all groups had protective levels of antibodies for all strains At day 21, the seroprotection rates in the three groups ranged from 88.0% to 100% for the different virus strains. In terms of protection, this means that more than 88% of the subjects had a serum HI titre 240 after vaccination and were deemed to be protected against the three strains. This rate is largely superior to the seroprotection rate of 60% required in the ⁇ 60 years old population, by the European authorities.
  • the GMTs and seroconversion rates for NI antibodies with 95% CI are shown in Table 11 (anti-NA antibody GMT) and Table 12 (Seroconversion rates of NA at post-vaccination (day 21) (4-fold-increase)).
  • 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 are presented for the CD4 and CD8 T-cell response in Tables 13 to 18.
  • Results were also expressed as a frequency of cytokine(s)-positive CD4 or CD8 T cell within the CD4 or CD8 T cell sub-population and presented in FIG. 4 and FIG. 5 .
  • the cross-reactive CD4 T-cells response was evaluated using influenza antigen from drifted strains (A/H1N1/Beijing/262/95 (H1N1d), A/H3N2/Sydney/5/97 (H3N2d), B/Yamanashi/166/98 (Bd)) or shift strains (A/Singapore/1/57 (H2N2), A/Hongkong/1073/99 (H9N2)). Results expressed as a frequency of cytokine(s)-positive CD4 T cells are presented in FIG. 6 .
  • Vaccination with Fluarix only induces low levels of cross-reactive CD4 T-cell response ( FIG. 6 ).
  • Vaccination with FluAS03 induces a strong CD4 T-cell response against drifted influenza strains and this is statistically significant ( FIG. 6 ). A little response was detected against shift strains.
  • the B-cells Elispot memory response induced to differentiate into plasma cells in vitro using influenza vaccine strains or anti-human immunoglobulin was evaluate in order to enumerate anti-influenza or IgG secreting plasma. The results are described in Table 19 and Table 20 and in FIG. 7 .
  • a subset of 22 first subjects having received one dose of FluAS03 vaccine and 21 first subjects having received one dose of Fluarix vaccine was selected to evaluate the impact of vaccination on influenza-specific memory B-cells using the B-cell memory Elispot technology. The following endpoints were determined
  • Descriptive statistics for each vaccination group at days 0 and day 21 expressed as a frequency of Influenza specific-antibody forming cells per million (10 6 ) of antibody forming cells. Descriptive statistics in individual difference between day 21 and day 0 (Post ⁇ Pre) as a frequency of Influenza specific-antibody forming cells per million (10 6 ) of antibody forming cells.
  • B-cells Memory descriptive statistics on pre (Day 0) and post (Day 21) and inferential statistics of post (Day 21) frequency of antigen- plasma within a 10 6 of IgG-producing plasma cells (subset of subjects) Time- STRAIN Group point N Mean SD Min A/NEW 1 Day 0 22 9751.58 6630.335 0.00 CALEDONIA 1 Day 21 22 22001.65 11308.261 3981.90 2 Day 0 21 9193.61 4339.421 1300.81 2 Day 21 21 12263.08 7285.698 789.47 A/PANAMA 1 Day 0 22 4329.17 2923.497 0.00 1 Day 21 22 18066.69 14604.842 714.29 2 Day 0 21 4860.41 3392.373 0.00 2 Day 21 21 13872.95 12052.163 0.00 B/SHANDONG 1 Day 0 22 3722.80 2347.315 0.00 1 Day 21 22 15949.60 12385.965 0.00 2 Day 0
  • the three vaccines exceeded the requirements of the European authorities for annual registration of split virion influenza vaccines (“Note for Guidance on Harmonisation of Requirements for influenza Vaccines” for the immuno-logical assessment of the annual strain changes -CPMP/BWP/214/96).
  • the three influenza vaccines tested in this study were immunogenic in the healthy elderly, who developed a good antibody response to influenza hemagglutinin and neutralising antigens (Table 21).
  • CMI cell-mediated immunity
  • a phase I/II, open, controlled study has been conducted in order to evaluate the reactogenicity and the immunogenicity of the GlaxoSmithKline Biologicals influenza candidate vaccine containing the adjuvant AS03, in an elderly population aged over 65 years and previously vaccinated in 2003 with the candidate vaccine in the Explo-Flu-001 clinical trial.
  • FluarixTM vaccine (known as ⁇ -rixTM in Belgium) has been used as reference.
  • the humoral immune response i.e. anti-hemagglutinin antibody titres
  • cell mediated immune response CD4 and/or CD8 T cell responses
  • B memory cell response were measured 21 days after intramuscular administration of one dose of an AS03 adjuvanted vaccine. FluarixTM was used as reference.
  • the objectives were:
  • the vaccine composition is similar to that used for the study Explo-Flu-001 except for the influenza strains included in the vaccine (year 2004 vaccine).
  • the strains are as follows:
  • cells producing at least two different cytokines CD40L, IL-2, IFN ⁇ , TNF ⁇
  • cells producing at least CD40L and another cytokine IL-2, TNF ⁇ , IFN ⁇
  • cells producing at least IL-2 and another cytokine CD40L, TNF ⁇ , IFN ⁇
  • cells producing at least IFN ⁇ and another cytokine IL-2, TNF ⁇ , CD40L
  • cells producing at least TNF ⁇ and another cytokine IL-2, CD40L, IFN ⁇
  • Results were expressed as a frequency of cytokine(s)-positive CD4 or CD8 T cell within the CD4 or CD8 T cell sub-population.
  • the frequency of antigen-specific CD4 T-lymphocytes secreting in response was summarised by descriptive statistics for each antigen, for each cytokine, for each vaccine group and at each timepoint (pre- and post-vaccination).
  • Vaccine-induced CD4 T-cells are shown to be able to persist at least for one year since there is an observable difference in prevaccination levels of CD4 T-cell responses between individuals vaccinated with Fluarix has compared to those vaccinated with Fluarix/AS03 the year before.
  • the results are also shown in FIG. 8 , showing the CD4 T-cell response to split Flu antigen before and after revaccination. D0 corresponds to 12 months after first year vaccination and thus indicates persistence.
  • the frequency of antigen-specific CD8 T-lymphocytes secreting in response was summarised by descriptive statistics for each antigen, for each cytokine, for each vaccine group and at each timepoint (pre- and post-vaccination), similarly to the procedure followed in respect of CD4 T cell response.
  • the frequency of antigen-specific CD4 T-lymphocytes secreting in response at prevaccination was summarised by descriptive statistics for each cytokine and for each vaccine group and for each of the two studies in Table 25, for each of the two studies study and for each vaccine group in Table 27. Inferential statistics are given in Table 26 and Table 28.
  • the frequency of antigen-specific CD4 T-lymphocytes secreting in response at (post-pre) timepoint was summarised by descriptive statistics for each cytokine and for each vaccine group and for each study in Table 29, for each study and for each vaccine group in Table 31. Inferential statistics are given in Table 30 and Table 32.
  • the adjuvanted vaccine Flu-AS03 is superior to the equivalent unadjuvated vaccine Fluarix in terms of frequency of influenza specific CD4 T cells, and also in terms of persistence of the immune response elicited by the first Flu-AS03 vaccination (primo-vaccination in Explo Flu 001) until D0 of the revaccination study (Explo Flu 002 i.e. +/ ⁇ 1 year later). Furthermore this response is capable to recognise drifted influenza strains present in the new vaccine and to recognise the strains of the 2004 influenza vaccine.
  • Influenza infection in the ferret model closely mimics human influenza, with regards both to the sensitivity to infection and the clinical response.
  • the ferret is extremely sensitive to infection with both influenza A and B viruses without prior adaptation of viral strains. Therefore, it provides an excellent model system for studies of protection conferred by administered influenza vaccines.
  • the objective of this experiment was to demonstrate the efficacy of an adjuvanted influenza vaccine compared to the plain (un-adjuvanted) vaccine.
  • ferrets Mustela putorius furo (6 ferrets/group) aged 14-20 weeks were obtained from MISAY Consultancy (Hampshire, UK). Ferrets were primed on day 0 with heterosubtypic strain H1N1 A/Stockholm/24/90 (4 Log TCID 50 /ml). On day 21, ferrets were injected intramuscularly with a full human dose (500 ⁇ g vaccine dose, 15 ⁇ g HA/strain) of a combination of H1N1 A/New Calcdonia/20/99, H3N2A/Panama/2007/99 and B/Shangdong/7/97. Ferrets were then challenged on day 41 by intranasal route with an homotypic strain H3N2 A/Panama/2007/99 (4.51 Log TCID 50 /ml).
  • Trivalent Full HD 15 ⁇ g IM; Day 21 Priming H1N1 Plain HA/strain (A/Stockolm/24/ 90) Day 0 2 Trivalent Full HD: 15 ⁇ g IM; Day 21 Priming H1N1 AS03 HA/strain (A/Stockolm/24/90) Day 0 3 Trivalent Full HD: 15 ⁇ g IM; Day 21 Priming H1N1 AS03 + MPL HA/strain (A/Stockolm/24/ 90) Day 0 4 PBS IM; Day 21 Priming H1N1 (A/Stockolm/24/ 90) Day 0
  • Formulation 1 Trivalent Plain (Un-Adjuvanted) Formulation (500 ⁇ l):
  • PBS 10 fold concentrated (pH 7.4 when one fold concentrated) as well as a mixture containing Tween 80, Triton X-100 and VES (quantities taking into account the detergents present in the strains) are added to water for injection.
  • the detergents quantities reached are the following: 750 ⁇ g Tween 80, 110 ⁇ g Triton X-100 and 100 ⁇ g VES per 1 ml.
  • 15 ⁇ g of each strain H1N1, H3N2 and 17.5 ⁇ g of B strain are added in sequence with 10 min stirring between each addition. The formulation is stirred for 15 minutes at room temperature and stored at 4° C. if not administered directly.
  • Formulation 2 Trivalent Split Influenza Adjuvanted with AS03 (500 ⁇ L):
  • PBS 10 fold concentrated (pH 7.4 when one fold concentrated) as well as a mixture containing Tween 80, Triton X-100 and VES (quantities taking into account the detergents present in the strains) is added to water for injection.
  • the detergents quantities reached are the following: 750 ⁇ g Tween 80, 110 ⁇ g Triton X-100 and 100 ⁇ g VES per 1 ml.
  • 15 ⁇ g of each strain H1N1, H3N2 and 17.5 ⁇ g of B strain are added with 10 min stirring between each addition.
  • 250 ⁇ l of SB62 emulsion (prepared as in taught in Example 11.1) is added. The formulation is stirred for 15 minutes at room temperature and stored at 4° C. if not administered directly.
  • PBS 10 fold concentrated (pH 7.4 when one fold concentrated) as well as a mixture containing Tween 80, Triton X-100 and VES (quantities taking into account the detergents present in the strains) is added to water for injection.
  • the detergents quantities reached are the following: 750 ⁇ g Tween 80, 110 ⁇ g Triton X-100 and 100 ⁇ g VES per 1 ml.
  • 15 ⁇ g of each strain H1N1, H3N2 and 17.5 ⁇ g of B strain are added with 10 min stirring between each addition.
  • 250 ⁇ l of SB62 emulsion (prepared as in taught in Example II.1) is added.
  • the mixture is stirred again for 15 min just prior addition of 25 ⁇ g of MPL from a suspension prepared as detailed in Example II.3.1.
  • the formulation is stirred for 15 minutes at room temperature and stored at 4° C. if not administered directly.
  • FIG. 10 A schematic representation of the results is given in FIG. 10 and FIG. 11 .
  • Viral titration of nasal washes was performed on 6 animals per group.
  • the nasal washes were performed by administration of 5 ml of PBS in both nostrils in awake animals.
  • the inoculation was collected in a Petri dish and placed into sample containers at ⁇ 80° C. (dry ice).
  • 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.
  • Trivalent Split adjuvanted with AS03 or AS03+MPL were observed with Trivalent Split adjuvanted with AS03 or AS03+MPL compared to the Trivalent Split Plain for all 3 strains (at least 2-fold for 2 out of 3 strains, i.e. H3N2 and B strains).
  • AS03 and AS03+MPL formulations showed added benefit in terms of protective efficacy in ferrets (lower viral shedding and temperature) ( FIGS. 10 and 11 ).
  • FIG. 12 A schematic representation of the results is given in FIG. 12 and in FIG. 13 .
  • the nasal washes were performed by administration of 5 ml of PBS in both nostrils in awake animals.
  • the inoculation was collected in a Petri dish and placed into sample containers at ⁇ 80° C. (dry ice).
  • 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 corresponds to +/ ⁇ 3 St Dev 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.
  • Viral shedding was measured for 12 ferrets from Day 1 Pre-priming- to Day 7 Post-priming. Results are expressed in pool.
  • Viral shedding was measured for 6 ferrets/group from Day 1 Pre-challenge to Day 7 Post-challenge.
  • Anti-Hemagglutinin antibody titers to the H3N2 influenza virus 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.
  • results are shown in FIGS. 14A and 14B .
  • H3N2 A/Panama After immunization with H3N2 A/Panama, higher humoral responses (HI titers) were observed in ferrets immunized with the trivalent split vaccine adjuvanted with AS03 or AS03+MPL, as compared to the humoral response observed after immunization of ferrets with the un-adjuvanted (plain) trivalent split vaccine (FluarixTM).
  • a boost of A/Wyoming-specific HI titers was observed in ferrets immunized with the heterologous strain A/Panama H3N2 and challenged with A/Wyoming H3N2.
  • the heterologous challenge resulted in an increase of A/Panama-specific HI titers in ferrets immunized with A/Panama H3N2 adjuvanted with AS03 and AS03+MPL.
  • the purpose was to select readouts to induce in mice similar CMI responses than observed in humans. Particularly, the purpose was to show higher CMI responses in mice by using Split AS03 or split AS03+MPL compared to Split plain.
  • mice/group mice Female C57BI/6 mice (15 mice/group) aged 6-8 weeks were obtained from Harlan Horst, Netherland. The groups tested were:
  • mice were primed on day 0 with heterosubtypic strains (5 ⁇ g HA whole inactivated H1N1 A/Johnannesburg/82/96, H3N2 A/Sydney/5/97, B/Harbin/7/94). On day 28, mice were injected intramuscularly with 1.5 ⁇ g HA Trivalent split (A/New Calcdonia/20/99, A/Panama/2007/99, B/Shangdong/7/97) plain or adjuvanted (see groups below).
  • PBS 10 fold concentrated is added to reach isotonicity and is 1 fold concentrated in the final volume.
  • H 2 O volume is calculated to reach the targeted volume.
  • Formulation 1 (for 500 ⁇ l): PBS 10 fold concentrated (pH 7.4 when one fold concentrated) as well as a mixture containing Tween 80, Triton X-100 and VES (quantities taking into account the detergents present in the strains) are added to water for injection.
  • the detergents quantities reached are the following: 750 ⁇ g Tween 80, 110 ⁇ g Triton X-100 and 100 ⁇ g VES per 1 ml
  • 15 ⁇ g of each strain H1N1, H3N2 and B are added with 10 min stirring between each addition.
  • the formulation is stirred for 15 minutes at room temperature and stored at 4° C. if not administered directly.
  • PBS 10 fold concentrated (pH 7.4 when one fold concentrated) as well as a mixture containing Tween 80, Triton X-100 and VES (quantities taking into account the detergents present in the strains) is added to water for injection.
  • the detergents quantities reached are the following: 750 ⁇ g Tween 80, 110 ⁇ g Triton X-100 and 100 ⁇ g VES per 1 ml.
  • 15 ⁇ g of each strain H1N1, H3N2 and B are added with 10 min stirring between each addition.
  • 250 ⁇ l of SB62 emulsion (prepared as taught in Example II.1) is added. The formulation is stirred for 15 minutes at room temperature and stored at 4° C. if not administered directly.
  • PBS 10 fold concentrated (pH 7.4 when one fold concentrated) as well as a mixture containing Tween 80, Triton X-100 and VES (quantities taking into account the detergents present in the strains) is added to water for injection.
  • the detergents quantities reached are the following: 750 ⁇ g Tween 80, 110 ⁇ g Triton X-100 and 100 ⁇ g VES per 1 ml
  • 15 ⁇ g of each strain H1N1, H 3 N 2 and B are added with 10 min stirring between each addition.
  • 250 ⁇ l of SB62 emulsion (prepared as taught in Example II.1) is added. The mixture is stirred again for 15 min just prior addition of 25 ⁇ g of MPL.
  • the formulation is stirred for 15 minutes at room temperature and stored at 4° C. if not administered directly.
  • PBMCs from primed mice were harvested 7 days post-immunization. They were tested in pools/group.
  • the purpose was to compare the CMI response induced by a GlaxoSmithKline commercially available split vaccine (FluarixTM) versus a subunit vaccine (Chiron's vaccine FluadTM) as well as the CMI response obtained with these vaccines adjuvanted with AS03, or AS03+MPL or another oil-in-water emulsion adjuvant (OW).
  • mice Female C57BI/6 mice (24 mice/group) aged 6-8 weeks were obtained from Harlan Horst, Netherland. Mice were primed intranasally on day 0 with heterosubtypic strains (5 ⁇ g HA whole formaldehyde inactivated H1N1 A/Johnannesburg/82/96, H3N2 A/Sydney/5/97, B/Harbin/7/94). On day 29, mice were injected intramuscularly with 1.5 ⁇ g HA Trivalent split (A/New Calcdonia/20/99, A/Wyoming/3/2003, B/Jiangsu/10/2003) plain or adjuvanted (see groups in Table 39 below).
  • An oil-in-water emulsion called OW is prepared following the recipe published in the instruction booklet contained in Chiron Behring FluAd vaccine.
  • Equal volume of PBS and FluAdTM/GripguardTM(commercial vaccine) vaccine are mixed.
  • the formulation is stirred for 15 minutes and stored at 4° C. if not administered directly.
  • PBS mod pH 7.4 (to reach a final volume of 1 ml) is added to a 500 ⁇ l dose of AggripalTM (commercial vaccine). After 15 min stirring, 250 ⁇ l of SB62 is added (prepared according to the methodoly detailed for the scaled-up production). 25 ⁇ g of MPL are then added. The formulation is stirred for 15 minutes and stored at 4° C. if not administered directly.
  • 250 ⁇ l of PBS mod pH 7.4 are added to a 500 ⁇ l dose of Aggripal. After 15 min stirring, 250 ⁇ l of OW as prepared for group 2 is added and the formulation is stirred 15 min and stored at 4° C. if not administered directly.
  • Equal volume of PBS mod pH 7.4 and Aggripal are mixed. The formulation is stirred for 15 minutes and stored at 4° C. if not administered directly.
  • IHA/neutralization assay 21 Days Post-immunization.
  • mice/group PBMCs from 24 mice/group were harvested 7 days post-immunization and tested in pools/group.
  • Haemagglutination inhibition activity against the 3 vaccine strains was detected in sera from 24 animals per group at Day 14 after intranasal heterologous priming and at Day 16 Post-immunization.
  • mice from 24 mice per group were harvested at Day 7 Post-immunization and tested in one pool/group. Inactivated trivalent whole viruses (1 ⁇ g/ml) were used as re-stimulating antigen. Results are shown in FIG. 17 upper part.
  • PBMCs from 24 mice per group were harvested at Day 7 Post-immunization and tested in one pool/group. Inactivated trivalent whole viruses (1 ⁇ g/ml) were used as re-stimulating antigen.
  • the strains used in the three vaccines were the ones that had been recommended by the WHO for the 2004-2005 Northern Hemisphere season, i.e. A/New Calcdonia/20/99 (H1N1), A/New California/3/2003 (H 3 N 2 ) and B/Jiangsu/10/2003.
  • the commercially available vaccine used as a comparator the adjuvanted vaccines (AS03, or AS03+MPL) contain 15 ⁇ g haemagglutinin (HA) of each influenza virus strain per dose.
  • the adjuvanted influenza candidate vaccines are 2 component vaccines consisting of a concentrated trivalent inactivated split virion antigens presented in a type I glass vial and of a pre-filled type I glass syringe containing the adjuvant (AS03 or AS03+MPL). They have been prepared as detailed in Example II.
  • the three inactivated split virion antigens (monovalent bulks) used in formulation of the adjuvanted influenza candidate vaccines, are exactly the same as the active ingredients used in formulation of the commercial FluarixTM/ ⁇ -Rix.
  • the AS03-adjuvanted influenza candidate vaccine is a 2 components vaccine consisting of a concentrated trivalent inactivated split virion antigens presented in a type I glass vial (335 ⁇ l) (antigen container) and of a pre-filled type I glass syringe containing the SB62 emulsion (335 ⁇ l) (adjuvant container). Description and composition of the AS03 candidate vaccine is explained in Example III.
  • the AS03+MPL-adjuvanted influenza candidate vaccine is a 2 components vaccine consisting of a concentrated trivalent inactivated split virion antigens presented in a type I glass vial (335 ⁇ l) (antigen container) and of a pre-filled type I glass syringe containing the AS03+MPL adjuvant (360 ⁇ l) (adjuvant container).
  • the content of the antigen container is removed from the vial by using the syringe containing the AS03+MPL adjuvant, followed by gently mixing of the syringe.
  • the used needle Prior to injection, the used needle is replaced by an intramuscular needle and the volume is corrected to 530 ⁇ l.
  • One dose of the reconstituted the AS03+MPL—adjuvanted influenza candidate vaccine corresponds to 530 ⁇ l.
  • the inactivated split virion antigen are concentrated two-fold in the antigen container (i.e. 60 ⁇ g HA/ml) as compared to FluarixTM (i.e. 30 ⁇ g HA/ml).
  • composition of one dose of the reconstituted adjuvanted influenza vaccine is identical to that reported in Table 45 (see Example XI) except for the influenza strains. Both vaccines were given intramuscularly.
  • the CMI objectives were to determine which immunogenic composition between the formulation adjuvanted with AS03, or AS03+MPL versus the composition without any adjuvant has the strongest immunostimulating activity on CD4- and CD8-mediated immunity of individuals vaccinated with influenza antigens.
  • the CMI analysis was based on the Total vaccinated cohort.
  • Results were expressed as a frequency of cytokine(s)-positive CD4 or CD8 T cell within the CD4 or CD8 T cell sub-population.
  • the objective of the study was to investigate whether the frequency of memory B cell specific to Flu Antigen are significantly induced upon one intramuscular vaccination with the Flu candidate vaccine containing the Adjuvant AS03+MPL or AS03, as compared to Fluarix in elderly population.
  • the frequency of memory B cell has been assessed by B cell Elispot assay.
  • the end points are:
  • AgrippalTM Chiron un-adjuvanted commercial sub-unit vaccine, which was in the present study adjuvanted with AS03 adjuvant.
  • the objective of this experiment was to evaluate the ability of these vaccines to reduce disease symptoms (body temperature and viral shedding) in nasal secretions of ferrets challenged with heterologous strains.
  • ferrets Mustela putorius furo ) aged 14-20 weeks were obtained from MISAY Consultancy (Hampshire, UK). Ferrets were primed intranasally on day 0 with the heterosubtypic strain H1N1 A/Stockholm/24/90 (4 Log TCID 50 /ml). On day 21, ferrets were injected intramuscularly with a full human dose (1 ml vaccine dose, 15 ⁇ g HA/strain) of a combination of H1N1 A/New Calcdonia/20/99, H3N2 A/Wyoming/3/2003 and B/Jiangsu/10/2003.
  • PBS 10 fold concentrated (pH 7.4 when one fold concentrated) as well as a mixture containing Tween 80, Triton X-100 and VES (quantities taking into account the detergents present in the strains) are added to water for injection.
  • the detergents quantities reached are the following: 375 ⁇ g Tween 80, 55 ⁇ g Triton X-100 and 50 ⁇ g VES per 1 ml.
  • 15 ⁇ g of each strain H1N1, H3N2 and 17.5 ⁇ g of B strain are added with 10 min stirring between each addition.
  • the formulation is stirred for 15 minutes at room temperature and stored at 4° C. if not administered directly.
  • PBS 10 fold concentrated (pH 7.4 when one fold concentrated) as well as a mixture containing Tween 80, Triton X-100 and VES (quantities taking into account the detergents present in the strains) is added to water for injection.
  • the detergents quantities reached are the following: 375 ⁇ g Tween 80, 55 ⁇ g Triton X-100 and 50 ⁇ g VES per 1 ml.
  • 15 ⁇ g of each strain H1N1, H3N2 and B are added with 10 min stirring between each addition.
  • 250 ⁇ l of SB62 emulsion (prepared as detailed in Example II.1) is added. The mixture is stirred again for 15 minutes just prior addition of 25 ⁇ g of MPL.
  • the formulation is stirred for 15 minutes at room temperature and stored at 4° C. if not administered directly.
  • Viral titration of nasal washes was performed on 6 animals per group.
  • the nasal washes were performed by the administration of 5 ml of PBS in both nostrils in awake animals.
  • the inoculation was collected in a Petri dish and placed into sample containers on dry ice ( ⁇ 80° C.).
  • 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 corresponds to +/ ⁇ 3 St Dev 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.
  • Results are shown in FIG. 20 .
  • Lower viral shedding was observed post-challenge with the trivalent split vaccine adjuvanted with AS03+MPL, or with the AgrippalTM sub-unit vaccine adjuvanted with AS03, as compared to the very low viral shedding reduction observed after immunization of ferrets with the un-adjuvanted (plain) trivalent split vaccine (FluarixTM) or with FluadTM sub-unit vaccine.
  • Anti-Hemagglutinin antibody titers to the H3N2 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 GMTs for HI antibodies with 95% CI are shown in FIG. 23 .
  • Pre-vaccination GMTs of antibodies for all 3 vaccine-strains were within the same range in the 3 groups. After vaccinations, anti-haemagglutinin antibody levels increased significantly.
  • Post-vaccination GMTs of antibodies for the 3 vaccine strains remained however within the same ranges for all vaccines.
  • influenza vaccines fulfilled the requirements of the European authorities for annual registration of influenza inactivated vaccines [“Note for Guidance on Harmonisation of Requirements for Influenza Vaccines for the immunological assessment of the annual strain changes” (CPMP/BWP/214/96)] in subjects aged over 60 years.
  • IL-2 cells producing at least IL-2 and another cytokine (CD40L, IFN- ⁇ , TNF- ⁇ ).
  • TNF- ⁇ cells producing at least TNF- ⁇ and another cytokine (CD40L, IFN- ⁇ , IL-2).
  • a phase I/II, open, controlled study was conducted in order to evaluate the reactogenicity and the immunogenicity of GlaxoSmithKline Biologicals influenza candidate vaccine containing the AS03+MPL adjuvant in an elderly population aged over 65 years (>65 years-old) previously vaccinated in 2004 with the same candidate vaccine.
  • Fluarix (known as ⁇ -RixTM in Belgium) vaccine was used as reference.
  • One objective of this study was to evaluate the humoral immune response (anti-haemagglutinin and anti-MPL titres) of the revaccination with the adjuvanted influenza vaccine Flu AS03+MPL administered about one year after administration of the first dose.
  • subjects who had already received FluarixTM in the previous trial received a dose of commercial vaccine and formed the control group of this trial.
  • the strains used in the three vaccines were the ones that had been recommended by the WHO for the 2005-2006 Northern Hemisphere season, i.e. A/New Calcdonia/20/99 (H1N1), A/New California/7/2004 (H3N2) and B/Jiangsu/10/2003.
  • the commercially available vaccine used as a comparator the (AS03+MPL—adjuvanted vaccine, hereinafter in short “the adjuvanted vaccine”) contains 15 ⁇ g haemagglutinin (HA) of each influenza virus strain per dose.
  • the adjuvanted influenza candidate vaccine is a 2 component vaccine consisting of a concentrated trivalent inactivated split virion antigens presented in a type I glass vial and of a pre-filled type I glass syringe containing the AS03+MPL adjuvant. It has been prepared according the method detailed in Example II.
  • the content of the prefilled syringe containing the adjuvant is injected into the vial that contains the concentrated trivalent inactivated split virion antigens. After mixing the content is withdrawn into the syringe and the needle is replaced by an intramuscular needle.
  • One dose of the reconstituted the adjuvanted influenza candidate vaccine corresponds to 0.7 mL.
  • the adjuvanted influenza candidate vaccine is a preservative-free vaccine.
  • composition of one dose of the reconstituted adjuvanted influenza vaccine is given in Table 45. Both vaccines were given intramuscularly.
  • influenza vaccines fulfilled the requirements of the European authorities for annual registration of influenza inactivated vaccines [“Note for Guidance on Harmonisation of Requirements for Influenza Vaccines for the immunological assessment of the annual strain changes” (CPMP/BWP/214/96)] in subjects aged over 60 years (Table 46).
  • the primary objective is to demonstrate the non inferiority 21 days post-vaccination of the influenza adjuvanted vaccines administered in elderly subjects (aged 65 years and older) as compared to FluarixTM administered in adults (aged 18-40 years) in terms of frequency of influenza-specific CD4 T-lymphocytes producing at least two different cytokines (CD40L, IL-2, TNF- ⁇ , IFN- ⁇ ).
  • the secondary objectives are:
  • the tertiary objective is to evaluate the cell mediated immune response (production of IFN- ⁇ , IL-2, CD40L, and TNF- ⁇ and memory B-cell response) 21, 90 and 180 days after vaccination with adjuvanted influenza-vaccines. FluarixTM is used as reference.
  • influenza vaccine adjuvanted with AS03+MPL(25 ⁇ g per dose) system is also used in study illustrated in Example XI.
  • the influenza vaccine adjuvanted with AS03+MPL(50 ⁇ g per dose) system is of identical composition except that the concentration of MPL is doubled.
  • the process is the same as the one described in Example VIII for the influenza vaccine adjuvanted with AS03+MPL, with as only difference that the concentration of MPL is doubled.
  • the GM ratio in term of influenza-specific CD4 frequency between groups vaccinated with adjuvanted vaccines and Flu YNG is obtained using an ANCOVA model on the logarithm-transformed titres.
  • the ANCOVA model includes the vaccine group as fixed effect and the pre-vaccination log-transformed titre as regressor.
  • the GM ratio and their 98.75% CI are derived as exponential-transformation of the corresponding group contrast in the model.
  • the 98.75% CI for the adjusted GM is obtained by exponential-transformation of the 98.75% CI for the group least square mean of the above ANCOVA model.
  • the adjusted GM and GM ratios (with their 98.75% CI) of influenza-specific CD4 T-lymphocyte producing at least two cytokines (IL-2, IFN- ⁇ , TNF- ⁇ and CD40L) at day 21, after in vitro restimulation with “pooled antigens II”, are presented in Table 47.
  • the upper limit of two-sided 98.75% CI of GM ratio is far below the clinical limit of 2.0. This shows the non-inferiority of both adjuvanted influenza vaccines administered to elderly subjects compared to the FluarixTM vaccine administered in adults aged between 18 and 40 years in term of post-vaccination frequency of influenza-specific CD4.
  • the frequency of influenza-specific CD4/CD8 T-lymphocytes and memory B-cells were measured at days 0, 21, 90 and 180.
  • the frequency of influenza-specific cytokine-positive CD4/CD8 T-lymphocytes was summarised (descriptive statistics) for each vaccination group at days 0 and 21, for each antigen.
  • Non-parametric test (Wilcoxon test) was used to compare the location of difference between the two groups (influenza adjuvanted vaccine versus FluarixTM) and the statistical p-value is calculated for each antigen at each different test.
  • Descriptive statistics in individual difference between day 21/day 0 (Post-/Pre-vaccination) responses is calculated for each vaccination group and each antigen at each different test.
  • Non-parametric test (Wilcoxon test) is used to compare the individual difference Post-/Pre-vaccination) and the statistical p-value will be calculated for each antigen at each different test.
  • HI antibody titres serum haemagglutination-inhibition (HI) antibody titres, tested separately against each of the three influenza virus strains represented in the vaccine (anti-H1N1, anti-H3N2 & anti-B-antibodies).
  • the cut-off value for HI antibody against all vaccine antigens was defined by the laboratory before the analysis (and equals 1:10).
  • a seronegative subject is a subject whose antibody titre is below the cut-off value.
  • a seropositive subject is a subject whose antibody titre is greater than or equal to the cut-off value.
  • Antibody titre below the cut-off of the assay is given an arbitrary value of half the cut-off.
  • the 95% CI for GM is obtained within each group separately.
  • the 95% CI for the mean of log-transformed titre is first obtained assuming that log-transformed titres are normally distributed with unknown variance.
  • the 95% CI for the GM is then obtained by exponential-transformation of the 95% CI for the mean of log-transformed titre.
  • Missing serological result for a particular antibody measurement is not replaced. Therefore a subject without serological result at a given time point do not contribute to the analysis of the assay for that time point.
  • Pre-vaccination GMTs of HI antibodies for all 3 vaccine strains were within the same range in the 4 treatment groups. After vaccination, there is clear impact of the 2 adjuvants which increase the humoral response in elderly, compared to standard Fluarix in the same population.
  • the adjuvanted influenza vaccines exceeded the requirements of the European authorities for annual registration of split virion influenza vaccines [“Note for Guidance on Harmonization of Requirements for Influenza Vaccines for the immunological assessment of the annual strain changes” (CPMP/BWP/214/96)] in subjects aged over 60 years.
  • the seroprotection rates for the 2 influenza adjuvanted vaccine groups are in the same range compared to Fluarix (18-40 years) group.
  • the seroconversion rates for the 2 influenza adjuvanted vaccine groups are in the same range compared to Fluarix (18-40 years) group excepted for New Calcdonia strain.
  • the maximum intensity of local injection site redness/swelling is scored as follows:
  • the maximum intensity of fever is scored as follows:
  • the investigator makes an assessment of intensity for all other AEs, i.e. unsolicited symptoms, including SAEs reported during the study. The assessment is based on the investigator's clinical judgement. The intensity of each AE recorded is assigned to one of the following categories:
  • Grade 3 symptoms showed a trend to be higher in the group who received the vaccine adjuvanted with the highest MPL concentration compared to the group who received the adjuvanted vaccine wherein the MPL is at a lower concentration. In all cases, symptoms however resolved rapidly.
  • mice were injected with an adjuvanted mixture containing 2.5 ⁇ g of each of 4 different antigens, HPV 16 L1, HPV 18 L1, HPV 31 L1, and HPV 45 L1, in the form of virus like particles.
  • Each L1 protein was a C terminal truncate removing, in the case of HPV 16, 34 amino acids (or the equivalent region in the other sequences).
  • HPV proteins were expressed and purified in baculovirus expression systems, for example as described in WO 03/077942.
  • the tetravalent VLP combination was combined with one of 2 different adjuvants.
  • the adjuvants tested were:
  • the adjuvant AS04 was used in a vaccine comprising HPV 16 and HPV 18 L1-only virus like particles, tested in phase II clinical trials as described in The Lancet, vol 364, issue 947, 13 Nov. 2004, 1757-1765. It thus provides a good basis for comparison with AS03+3D MPL.
  • This adjuvant can be made as described in, for example, WO 01/17751 and WO 00/23105.
  • HPV-16/18/31/45 L1 antibody Quantitation of anti-HPV-16/18/31/45 L1 antibody was performed by ELISA using HPV-16 L1 (lot E16L1P093), HPV-18 L1 (lot E18L1P079), HPV-31 L1 (lot EA31 L1P329) and HPV-45 L1 (lot EA45L1P328) as coating antigen.
  • HPV-16/18/31/45 L1 and antibody solutions were used at 50 ⁇ l per well; only the saturation solution was used at 100 ⁇ l per well.
  • HPV-16/18/31/45 L1 were diluted at a final concentration of 0.5 ⁇ g/ml in PBS and were adsorbed overnight at 4° C.
  • mice sera in the dilution buffer (saturation buffer+0.1% Tween20) were added to the coated plates after removal of saturation solution and incubated for 1 hr 30 min at 37° C.
  • optical densities were measured using a microplate reader connected to a computer. Data were captured with the SoftMaxPro software. In order to titrate each sample, a standard is included on each plate. A four parameters logistic log function is used to calculate the standard curve. Antibody concentrations were calculated at each dilution of the test sample by interpolation of the standard curves.
  • the antibody titers were obtained by averaging the values from all dilutions that fall within the working range (20-80% OD) of the standard curve. ELISA titers are expressed in EU/ml.
  • mice Thirty-three or seventy-five days after the second immunization, mice were sacrificed; spleen cells were separated by a lymphoprep gradient. PBMCs were then resuspended in RPMI 1640 medium (Gibco) containing additives (sodium pyruvate 1 mM, MEM non-essential amino acids, Pen/Strep, Glutamine and ⁇ -2 mercaptoethanol), 5% fetal calf serum, 50 U/ml rhlL-2 (eBioscience) and 3 ⁇ g/ml CpG (phosphothioated CpG ODN-7909-5′-TCG TCG TTT TGT CGT TTT GTC GTT-3′-SEQ ID NO.1).
  • additives sodium pyruvate 1 mM, MEM non-essential amino acids, Pen/Strep, Glutamine and ⁇ -2 mercaptoethanol
  • 5% fetal calf serum 50 U/ml rh
  • CpG sequences are also suitable for use in this B memory evaluation method.
  • Cells were cultured five days at a final concentration of 10 6 cells/ml, in 5 ml per flat-bottomed 6 wells.
  • nitrocellulose plates Multiscreen-IP; Millipore
  • GAM goat anti-mouse Ig
  • 100 ⁇ l of 2.10 6 cells/ml were added to HPV-16/18 L1 coated plates and 100 ⁇ l of 10 6 and 5.10 5 cells/ml were added to GAM plates.
  • the percentage of B memory cells specific for HPV-16/18 L1 corresponds to the ratio of HPV-16/18 L1 positive spots compared to the total IgG spots.
  • HPV16 HPV18 Frequency of HPV specific memory B cells in total IgG memory B cells at day 33 post II VLPs 2.5 ⁇ g/AS04 0.4% 0.2% VLPs 2.5 ⁇ g/AS03 + 3D MPL 0.7% 0.4% Frequency of HPV specific memory B cells in total lgG memory B cells at day 75 post II VLPs 2.5 ⁇ g/AS04 2.0% 0.6% VLPs 2.5 ⁇ g/AS03 + 3D MPL 3.8% 0.5%
  • AS03+3D-MPL adjuvant demonstrates immunogenicity results for both antibody production and B cell memory which are equivalent to, or sometimes greater than, those generated with AS04, depending upon the HPV type being assessed.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pulmonology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US11/909,414 2005-03-23 2006-03-21 Composition Abandoned US20090081253A1 (en)

Applications Claiming Priority (27)

Application Number Priority Date Filing Date Title
GB0506004A GB0506004D0 (en) 2005-03-23 2005-03-23 Compositions
GB0505998.5 2005-03-23
GB0506000A GB0506000D0 (en) 2005-03-23 2005-03-23 Novel use
GB0505989.4 2005-03-23
GB0505989A GB0505989D0 (en) 2005-03-23 2005-03-23 Compositions
GB0506001A GB0506001D0 (en) 2005-03-23 2005-03-23 Novel use
GB0505998A GB0505998D0 (en) 2005-03-23 2005-03-23 Novel compositions
GB0506000.9 2005-03-23
GB0506001.7 2005-03-23
GB0506004.1 2005-03-23
GB0510596.0 2005-05-24
GB0510591A GB0510591D0 (en) 2005-05-24 2005-05-24 Novel use
GB0510598A GB0510598D0 (en) 2005-05-24 2005-05-24 Composition
GB0510589.5 2005-05-24
GB0510598.6 2005-05-24
GB0510591.1 2005-05-24
GB0510593A GB0510593D0 (en) 2005-05-24 2005-05-24 Novel compositions
GB0510593.7 2005-05-24
GB0510589A GB0510589D0 (en) 2005-05-24 2005-05-24 Novel use
GB0510596A GB0510596D0 (en) 2005-05-24 2005-05-24 Composition
GB0603788.1 2006-02-24
GB0603790.7 2006-02-24
GB0603790A GB0603790D0 (en) 2006-02-24 2006-02-24 Composition
GB0603789.9 2006-02-24
GB0603788A GB0603788D0 (en) 2006-02-24 2006-02-24 Novel composition
GB0603789A GB0603789D0 (en) 2006-02-24 2006-02-24 Novel use
PCT/EP2006/002838 WO2006100111A1 (en) 2005-03-23 2006-03-21 Composition

Publications (1)

Publication Number Publication Date
US20090081253A1 true US20090081253A1 (en) 2009-03-26

Family

ID=36441219

Family Applications (5)

Application Number Title Priority Date Filing Date
US11/909,414 Abandoned US20090081253A1 (en) 2005-03-23 2006-03-21 Composition
US11/909,388 Abandoned US20080171063A1 (en) 2005-03-23 2006-03-21 Novel Composition
US11/909,351 Abandoned US20080181911A1 (en) 2005-03-23 2006-03-21 Use Of An Influenza Virus, An Oil-In-Water Emulsion Adjuvant, To Induce Cd4 T-Cell And/Or Improved B-Memory Cell Response
US13/096,180 Abandoned US20110287054A1 (en) 2005-03-23 2011-04-28 Multivalent adjuvanted influenza virus compositions
US14/465,055 Active US9730999B2 (en) 2005-03-23 2014-08-21 Adjuvanted influenza virus compositions

Family Applications After (4)

Application Number Title Priority Date Filing Date
US11/909,388 Abandoned US20080171063A1 (en) 2005-03-23 2006-03-21 Novel Composition
US11/909,351 Abandoned US20080181911A1 (en) 2005-03-23 2006-03-21 Use Of An Influenza Virus, An Oil-In-Water Emulsion Adjuvant, To Induce Cd4 T-Cell And/Or Improved B-Memory Cell Response
US13/096,180 Abandoned US20110287054A1 (en) 2005-03-23 2011-04-28 Multivalent adjuvanted influenza virus compositions
US14/465,055 Active US9730999B2 (en) 2005-03-23 2014-08-21 Adjuvanted influenza virus compositions

Country Status (24)

Country Link
US (5) US20090081253A1 (hr)
EP (4) EP1861120B1 (hr)
JP (4) JP5770414B2 (hr)
KR (5) KR20070116650A (hr)
AR (3) AR053833A1 (hr)
AU (3) AU2006226459A1 (hr)
CA (3) CA2601022C (hr)
CY (1) CY1117874T1 (hr)
DK (1) DK1861120T3 (hr)
EA (3) EA011393B1 (hr)
ES (1) ES2585810T3 (hr)
HR (1) HRP20160816T1 (hr)
HU (1) HUE027837T2 (hr)
IL (3) IL185901A0 (hr)
MA (3) MA29342B1 (hr)
MX (3) MX2007011756A (hr)
NO (3) NO20074635L (hr)
NZ (2) NZ561823A (hr)
PE (3) PE20061428A1 (hr)
PL (1) PL1861120T3 (hr)
PT (1) PT1861120T (hr)
SI (1) SI1861120T1 (hr)
TW (3) TW200700079A (hr)
WO (3) WO2006100109A1 (hr)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070141078A1 (en) * 1999-09-30 2007-06-21 D Hondt Erik Influenza vaccine
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
US20090176273A1 (en) * 2007-12-07 2009-07-09 National Health Research Institutes Production of Lipidated Proteins In E. coli
US20100166785A1 (en) * 2007-12-07 2010-07-01 National Health Research Institutes Adjuvants
US20100303849A1 (en) * 2009-06-02 2010-12-02 National Health Research Institutes Lipidated Vaccine against Dengue Virus Infection
US20100322953A1 (en) * 2009-06-22 2010-12-23 National Health Research Institutes Lipidated Tumor-Associated Antigens and Immunotherapeutic Compositions
US20110034996A1 (en) * 2009-08-06 2011-02-10 Alexander Borck Medical implant containing an antioxidative substance
WO2011040811A1 (en) * 2009-10-02 2011-04-07 Mucosis B.V. Adjuvanted vaccine formulations
DE102009056871A1 (de) * 2009-12-03 2011-06-22 Novartis AG, 4056 Impfstoff-Adjuvantien und verbesserte Verfahren zur Herstellung derselben
WO2011088451A1 (en) * 2010-01-15 2011-07-21 Novavax, Inc. Uses of influenza virus-like particles (vlps) for characterization of neuraminidase and hemagglutinin activity
WO2012066420A1 (en) * 2010-11-15 2012-05-24 National Health Research Institutes Lipidated polyepitope vaccines
EP2464341A2 (en) * 2009-08-12 2012-06-20 Sigmoid Pharma Limited Immunomodulatory compositions comprising a polymer matrix and an oil phase
US8771990B2 (en) 2010-11-15 2014-07-08 National Health Research Institutes Method of producing lipidated polypeptides
US9278127B2 (en) 2006-07-17 2016-03-08 Glaxosmithkline Biologicals, Sa Influenza vaccine

Families Citing this family (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2371994C (en) * 1999-02-26 2010-09-28 Guido Grandi Enhancement of bactericidal activity of neisseria antigens with oligonucleotides containing cg motifs
US20100221284A1 (en) * 2001-05-30 2010-09-02 Saech-Sisches Serumwerk Dresden Novel vaccine composition
DE10144906B4 (de) 2001-09-12 2013-11-28 Novartis Vaccines And Diagnostics Gmbh Verfahren zur großtechnischen Herstellung von Impfstoffen
US11707520B2 (en) 2005-11-03 2023-07-25 Seqirus UK Limited Adjuvanted vaccines with non-virion antigens prepared from influenza viruses grown in cell culture
DE202006021242U1 (de) * 2005-11-04 2014-01-29 Novartis Vaccines And Diagnostics S.R.L. Emulsionen mit freiem wässrigen Phasen Tensid als Adjuvans für Spalt-Grippeimpfstoffe
JP2009514838A (ja) * 2005-11-04 2009-04-09 ノバルティス ヴァクシンズ アンド ダイアグノスティクス エスアールエル 細胞培養物において増殖されたインフルエンザウイルスから調製された非ビリオン抗原を含むアジュバントワクチン
ATE513561T1 (de) 2005-11-04 2011-07-15 Novartis Vaccines & Diagnostic Veränderung des th1/th2-gleichgewichts in spalt- grippeimpfstoffen mit adjuvantien
CN102755645A (zh) * 2005-11-04 2012-10-31 诺华疫苗和诊断有限公司 佐剂配制的包含细胞因子诱导剂的流感疫苗
FR2896162B1 (fr) * 2006-01-13 2008-02-15 Sanofi Pasteur Sa Emulsion huile dans eau thermoreversible
TW200745158A (en) 2006-03-07 2007-12-16 Vaxinnate Corp Compositions that include hemagglutinin, methods of making and methods of use thereof
CA2646349A1 (en) * 2006-03-24 2007-10-04 Novartis Vaccines And Diagnostics Gmbh & Co Kg Storage of influenza vaccines without refrigeration
CN101500603A (zh) * 2006-06-15 2009-08-05 诺华有限公司 节省佐剂的多剂量流感疫苗接种方案
CA2583555C (en) * 2006-07-17 2020-01-07 Glaxosmithkline Biologicals S.A. Influenza vaccine
SI2422810T1 (sl) * 2006-07-17 2015-01-30 Glaxosmithkline Biologicals S.A. Influenčno cepivo
ES2397714T3 (es) * 2006-10-12 2013-03-08 Glaxosmithkline Biologicals S.A. Vacuna que comprende un adyuvante de emulsión de aceite en agua
SI2086582T1 (sl) * 2006-10-12 2013-02-28 Glaxosmithkline Biologicals S.A. Cepivo, ki obsega emulzijo olja v vodi kot adjuvans
GB0622282D0 (en) 2006-11-08 2006-12-20 Novartis Ag Quality control methods
EP1923070A1 (en) * 2006-11-15 2008-05-21 Intervet International BV Vaccine for vaccinating feline against influenza virus
SG177141A1 (en) * 2006-12-06 2012-01-30 Novartis Ag Vaccines including antigen from four strains of influenza virus
EP1938835A1 (en) * 2006-12-29 2008-07-02 Pevion Biotech AG Non-specific immunostimulating agents
AU2008231242B2 (en) * 2007-02-23 2013-01-31 Baylor Research Institute Therapeutic applications of activation of human antigen-presenting cells through Dectin-1
EP2195019A4 (en) * 2007-10-12 2012-02-15 Csl Ltd METHOD FOR TRIGGERING AN IMMUNE RESPONSE AGAINST THE PANDEMIC INFLUENZA VIRUS
SI2200642T1 (sl) * 2007-10-19 2012-06-29 Novartis Ag Formulacije meningokoknega cepiva
GB0810305D0 (en) 2008-06-05 2008-07-09 Novartis Ag Influenza vaccination
EP2227251A1 (en) 2007-12-06 2010-09-15 GlaxoSmithKline Biologicals SA Influenza composition
CN101998990B (zh) * 2008-03-18 2013-11-27 诺华股份有限公司 流感病毒疫苗抗原制备方法的改进
WO2009128949A2 (en) 2008-04-18 2009-10-22 Vaxinnate Corporation Compositions of dengue viral proteins and methods of use
GB0905570D0 (en) * 2009-03-31 2009-05-13 Novartis Ag Combined vaccines
US9421251B2 (en) 2008-06-25 2016-08-23 Novartis Ag Rapid responses to delayed booster immunisations
JP5642712B2 (ja) 2009-02-10 2014-12-17 ノバルティス アーゲー 少ない量のスクアレンを含むインフルエンザワクチン
EA201171032A1 (ru) 2009-02-10 2012-02-28 Новартис Аг Схемы лечения с помощью вакцины против гриппа, связанного с пандемическими штаммами
AU2015203072B2 (en) * 2009-02-10 2017-05-25 Seqirus UK Limited Influenza vaccine regimens for pandemic-associated strains
GB0918830D0 (en) 2009-10-27 2009-12-09 Glaxosmithkline Biolog Niederl Process
GB0919117D0 (en) 2009-10-30 2009-12-16 Glaxosmithkline Biolog Sa Process
AU2013202690B2 (en) * 2009-12-03 2016-02-11 Novartis Ag Hydrophilic filtration during manufacture of vaccine adjuvants
CA2782511C (en) 2009-12-03 2013-11-12 Novartis Ag Circulation of components during homogenization of emulsions
CL2012001399A1 (es) 2009-12-03 2013-03-08 Novartis Ag Metodo para fabricar adyuvante para vacuna (emulsion aceite/agua con escualeno, polisorbato 80 y trioleato de sorbitan), que comprende (i) formar primera emulsion en homogenizador desde un contendor a otro para formar segunda emulsion, (ii) y microfluidizar primera emulsion para formar segunda emulsion.
CN102740832B (zh) * 2009-12-03 2016-01-13 诺华股份有限公司 就微流化排列交互作用和背压室
DE102009056883B4 (de) 2009-12-03 2012-08-16 Novartis Ag Impfstoff-Adjuvantien und verbesserte Verfahren zur Herstellung derselben
EA025622B1 (ru) * 2009-12-03 2017-01-30 Новартис Аг Способ получения содержащей сквален эмульсии масла
DE102009056884B4 (de) 2009-12-03 2021-03-18 Novartis Ag Impfstoff-Adjuvantien und verbesserte Verfahren zur Herstellung derselben
JP5894083B2 (ja) * 2009-12-22 2016-03-23 サノフィ パストゥール リミテッドSanofi Pasteur Limited 免疫原性組成物
SG182305A1 (en) * 2010-01-06 2012-08-30 Vaxinnate Corp Methods and compositions for providing protective immunity in the elderly
RU2527688C2 (ru) * 2010-02-27 2014-09-10 Общество С Ограниченной Ответственностью "Научно-Производственная Фирма "Материа Медика Холдинг" Способ лечения инфекционных заболеваний, вызванных вирусом гриппа с типом поверхностого антигена н1n1 и лекарственное средство для лечения инфекционных заболеваний, вызванных вирусом гриппа с типом поверхностного антигена н1n1
GB201009671D0 (en) 2010-06-10 2010-07-21 Glaxosmithkline Biolog Sa Novel process
GB201009676D0 (en) 2010-06-10 2010-07-21 Glaxosmithkline Biolog Sa Novel process
GB201009673D0 (en) 2010-06-10 2010-07-21 Glaxosmithkline Biolog Sa Novel process
US9821051B1 (en) 2010-10-28 2017-11-21 Seqirus UK Limited Reducing hospitalization in elderly influenza vaccine recipients
WO2012116714A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in elderly patients
US20160015800A1 (en) * 2012-12-28 2016-01-21 Japan As Represented By Director General Of National Institute Of Infectious Diseases Nasal influenza vaccine composition
JP2016521282A (ja) * 2013-05-10 2016-07-21 ノバルティス アーゲー インフルエンザワクチンにおけるナルコレプシーのリスクの回避
US10080792B2 (en) * 2013-09-23 2018-09-25 Engen Bio, Inc. Influenza vaccine and therapy
CN104436157A (zh) * 2013-09-23 2015-03-25 恩金生物有限公司 流感疫苗和治疗
US20170304429A1 (en) 2014-09-26 2017-10-26 Seqirus UK Limited Vaccination of immunocompromised subjects
WO2019108928A1 (en) * 2017-11-30 2019-06-06 Ohio State Innovation Foundation Mucoadhesive nanoparticle entrapped influenza virus vaccine delivery system
CN113993542A (zh) * 2019-01-30 2022-01-28 葛兰素史密丝克莱恩生物有限公司 用于自乳化的油/表面活性剂混合物
MX2022007691A (es) * 2019-12-20 2022-07-19 Fresenius Kabi Austria Gmbh Metodo para producir emulsiones aceite en agua.
RU2741003C1 (ru) * 2020-03-27 2021-01-22 Евгений Дмитриевич Некрасов Способ производства четырехвалентной субъединичной вакцины против гриппа без адъювантов
US20230256090A1 (en) 2020-06-29 2023-08-17 Glaxosmithkline Biologicals Sa Adjuvants
MX2023002339A (es) 2020-08-24 2023-03-22 Sanofi Pasteur Inc Vacunas contra infecciones por sars-cov-2.
CA3190375A1 (en) 2020-08-24 2022-03-03 Natalie ANOSOVA Covid-19 vaccines with tocopherol-containing squalene emulsion adjuvants
KR102270165B1 (ko) 2020-10-22 2021-06-28 한국화학연구원 세정제 조성물
WO2022200582A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2023274860A1 (en) 2021-06-28 2023-01-05 Glaxosmithkline Biologicals Sa Novel influenza antigens
RU2766292C1 (ru) * 2021-08-05 2022-03-14 Федеральное государственное унитарное предприятие "Санкт-Петербургский научно-исследовательский институт вакцин и сывороток и предприятие по производству бактерийных препаратов" Федерального медико-биологического агентства (ФГУП СПбНИИВС ФМБА России) Состав вакцины против covid-19
WO2023020992A1 (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
WO2023020994A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023061993A1 (en) 2021-10-13 2023-04-20 Glaxosmithkline Biologicals Sa Polypeptides

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376369A (en) * 1987-11-03 1994-12-27 Syntex (U.S.A.) Inc. Vaccine adjuvant
US6451320B1 (en) * 1998-03-09 2002-09-17 Smithkline Beecham Biologicals S.A. Combined hepatitis and herpesvirus antigen compositions
US20030095974A1 (en) * 1997-09-05 2003-05-22 Smithkline Beecham Biologicals S.A. Oil in water emulsions containing saponins

Family Cites Families (113)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3095974A (en) * 1961-10-27 1963-07-02 Anthony R Perini Collapsible book rack
US4047869A (en) * 1973-02-23 1977-09-13 Mulvany Jr R F Apparatus for forming plastic articles
FR2333610A1 (fr) * 1975-12-03 1977-07-01 Zhdanovsky Z Tyazhelogo Mash Bande-electrode a ame en poudre pour rechargement a l'aide d'un alliage resistant a l'usure
US4270537A (en) 1979-11-19 1981-06-02 Romaine Richard A Automatic hypodermic syringe
DD155875A1 (de) 1980-12-31 1982-07-14 Willy Nordheim Verfahren zur herstellung eines ballaststoffarmen inaktivierten influenzaimpfstoffes
US4436727A (en) * 1982-05-26 1984-03-13 Ribi Immunochem Research, Inc. Refined detoxified endotoxin product
US4866034A (en) * 1982-05-26 1989-09-12 Ribi Immunochem Research Inc. Refined detoxified endotoxin
DD211444A3 (de) 1982-08-19 1984-07-11 Saechsisches Serumwerk Verfahren zur herstellung von influenza-impfstoffen
GB8300467D0 (en) 1983-01-08 1983-02-09 Wellcome Found Equine influenza
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
DD300833A7 (de) 1985-10-28 1992-08-13 Saechsische Landesgewerbefoerd Verfahren zur herstellung von inaktivierten influenza-vollvirusimpfstoffen
CA1283827C (en) 1986-12-18 1991-05-07 Giorgio Cirelli Appliance for injection of liquid formulations
GB8704027D0 (en) 1987-02-20 1987-03-25 Owen Mumford Ltd Syringe needle combination
AU614755B2 (en) 1987-06-05 1991-09-12 United States of America, as represented by the Secretary, U.S. Department of Commerce, The Autocrine motility factors in cancer diagnosis and management
US4940460A (en) 1987-06-19 1990-07-10 Bioject, Inc. Patient-fillable and non-invasive hypodermic injection device assembly
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
DE3734306A1 (de) 1987-10-10 1989-04-27 Pfeiffer Erich Gmbh & Co Kg Austragvorrichtung fuer fliessfaehige medien
US5339163A (en) 1988-03-16 1994-08-16 Canon Kabushiki Kaisha Automatic exposure control device using plural image plane detection areas
US4912094B1 (en) 1988-06-29 1994-02-15 Ribi Immunochem Research Inc. Modified lipopolysaccharides and process of preparation
GB8821049D0 (en) 1988-09-08 1988-10-05 Health Lab Service Board Method & composition for treatment & prevention of viral infections
FR2638359A1 (fr) 1988-11-03 1990-05-04 Tino Dalto Guide de seringue avec reglage de la profondeur de penetration de l'aiguille dans la peau
DK0382271T3 (da) * 1989-02-04 1995-05-01 Akzo Nobel Nv Tocoler som adjuvanser i vacciner
US5092235A (en) 1989-05-24 1992-03-03 Tektronix, Inc. Pressure fixing and developing apparatus
HU212924B (en) 1989-05-25 1996-12-30 Chiron Corp Adjuvant formulation comprising a submicron oil droplet emulsion
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
DE4005528C2 (de) 1990-02-22 1998-01-15 Pfeiffer Erich Gmbh & Co Kg Austragvorrichtung für Medien
US5969109A (en) * 1990-02-28 1999-10-19 Bona; Constantin Chimeric antibodies comprising antigen binding sites and B and T cell epitopes
WO1991013261A1 (en) 1990-03-02 1991-09-05 Experimentalny Nauchno-Issledovatelsky Institut Metallorezhuschikh Stankov Digital servo drive
US5149531A (en) * 1990-06-27 1992-09-22 University Of Pittsburgh Of The Commonwealth System Of Higher Education Method of using cold-adapted live influenza virus vaccine as an antiviral agent against influenza
US5190521A (en) 1990-08-22 1993-03-02 Tecnol Medical Products, Inc. Apparatus and method for raising a skin wheal and anesthetizing skin
US5527288A (en) 1990-12-13 1996-06-18 Elan Medical Technologies Limited Intradermal drug delivery device and method for intradermal delivery of drugs
GB9105992D0 (en) 1991-03-21 1991-05-08 Smithkline Beecham Biolog Vaccine
GB9118204D0 (en) 1991-08-23 1991-10-09 Weston Terence E Needle-less injector
SE9102652D0 (sv) 1991-09-13 1991-09-13 Kabi Pharmacia Ab Injection needle arrangement
US5328483A (en) 1992-02-27 1994-07-12 Jacoby Richard M Intradermal injection device with medication and needle guard
MY111880A (en) 1992-03-27 2001-02-28 Smithkline Beecham Biologicals S A Hepatitis vaccines containing 3-0 deacylated monophosphoryl lipid a
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
WO1994019013A1 (en) 1993-02-19 1994-09-01 Smithkline Beecham Corporation Influenza vaccine compositions containing 3-o-deacylated monophosphoryl lipid a
JPH08506592A (ja) * 1993-02-19 1996-07-16 スミスクライン・ビーチャム・コーポレイション 3−o−脱アシル化モノホスホリル脂質A含有のインフルエンザワクチン組成物
CN1087176C (zh) 1993-03-23 2002-07-10 史密斯克莱·比奇曼生物公司 含有3-o脱酰基单磷酰脂a的疫苗制剂
US5762939A (en) * 1993-09-13 1998-06-09 Mg-Pmc, Llc Method for producing influenza hemagglutinin multivalent vaccines using baculovirus
WO1995011700A1 (en) 1993-10-29 1995-05-04 Pharmos Corp. Submicron emulsions as vaccine adjuvants
WO1995014026A1 (en) 1993-11-17 1995-05-26 Laboratoires Om S.A. Glucosamine disaccharides, method for their preparation, pharmaceutical composition comprising same, and their use
GB9326253D0 (en) * 1993-12-23 1994-02-23 Smithkline Beecham Biolog Vaccines
BR9506885A (pt) 1994-02-24 1997-08-19 Micro Pak Inc Vacinas contendo vesículas de lipidio paucilamelares como adjuvantes imunológicos
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
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
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
US5916879A (en) 1996-11-12 1999-06-29 St. Jude Children's Research Hospital DNA transcription unit vaccines that protect against avian influenza viruses and methods of use thereof
TW570803B (en) 1997-04-09 2004-01-11 Duphar Int Res Influenza vaccine
US5993412A (en) 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
GB9711990D0 (en) 1997-06-11 1997-08-06 Smithkline Beecham Biolog Vaccine
GB9718901D0 (en) 1997-09-05 1997-11-12 Smithkline Beecham Biolog Vaccine
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
US6838269B1 (en) * 1998-04-15 2005-01-04 Genencor International, Inc. Proteins producing an altered immunogenic response and methods of making and using the same
JP2002513773A (ja) 1998-05-07 2002-05-14 コリクサ コーポレイション アジュバント組成物及びその使用法
SK18602000A3 (sk) 1998-06-08 2001-07-10 Sca Emballage France Obal umožňujúci rýchle vyrovnanie
HUP0102475A3 (en) 1998-06-30 2001-12-28 Om Pharma Novel acyl pseudodipeptides, preparation method and pharmaceutical compositions containing same
AT408615B (de) 1998-09-15 2002-01-25 Immuno Ag Neue influenzavirus-impfstoffzusammensetzung
US20040006242A1 (en) 1999-02-01 2004-01-08 Hawkins Lynn D. Immunomodulatory compounds and method of use thereof
US6551600B2 (en) 1999-02-01 2003-04-22 Eisai Co., Ltd. Immunological adjuvant compounds compositions and methods of use thereof
AT407958B (de) 1999-02-11 2001-07-25 Immuno Ag Inaktivierte influenza-virus-vakzine zur nasalen oder oralen applikation
AU2875700A (en) 1999-02-26 2000-09-14 Chiron Corporation Microemulsions with adsorbed macromoelecules and microparticles
FR2796290B1 (fr) 1999-07-16 2001-09-14 Cross Site Technologies Seringue sans aiguille fonctionnant avec un generateur d'onde de choc a travers une paroi
FR2796291B1 (fr) 1999-07-16 2001-09-21 Cross Site Technologies Seringue sans aiguille munie d'un systeme de declenchement piezo-electrique
FR2796289B1 (fr) 1999-07-16 2001-08-10 Cross Site Technologies Seringue sans aiguille avec injecteur a elements superposes
GB9921146D0 (en) * 1999-09-07 1999-11-10 Smithkline Beecham Biolog Novel composition
GB9921147D0 (en) * 1999-09-07 1999-11-10 Smithkline Beecham Biolog Novel composition
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
FR2800619B1 (fr) 1999-11-05 2002-02-08 Cross Site Technologies Seringue sans aiguille avec un moyen de poussee temporairement retenu
FR2802102B1 (fr) 1999-12-08 2002-07-12 Poudres & Explosifs Ste Nale Seringue sans aiguille munie d'un tube d'ejection a section constante
FR2802103B1 (fr) 1999-12-08 2003-10-03 Poudres & Explosifs Ste Nale Seringue sans aiguille fonctionnant avec entrainement du principe actif par effet tube a choc
AU1581400A (en) 1999-12-22 2001-07-03 Om Pharma Acyl pseudopeptides bearing a functionalised auxiliary spacer
FR2802820B1 (fr) 1999-12-27 2002-10-18 Poudres & Explosifs Ste Nale Seringue sans aiguille fonctionnant par effet tube a choc, avec maintien prealable du principe actif sur le cote
CZ20022643A3 (cs) 2000-01-31 2003-02-12 Smithkline Beecham Biologicals S. A. Farmaceutický prostředek
FR2804329B1 (fr) 2000-02-02 2002-12-13 Poudres & Explosifs Ste Nale Seringue sans aiguille munie d'un opercule contenant le principe actif
FR2804869B1 (fr) 2000-02-11 2002-05-17 Poudres & Explosifs Ste Nale Seringue sans aiguille pour l'injection d'un liquide contenu dans une ampoule pre-remplie
FR2805749B1 (fr) 2000-03-01 2002-05-17 Poudres & Explosifs Ste Nale Seringue sans aiguille a deux niveaux de vitesse d'injection
FR2807946B1 (fr) 2000-04-19 2002-06-07 Poudres & Explosifs Ste Nale Seringue sans aiguille fonctionnant avec un chargement pyrotechnique bicomposition
FR2809626B1 (fr) 2000-05-30 2003-03-07 Poudres & Explosifs Ste Nale Seringue sans aiguille avec membrane d'isolation d'un ejecteur multiconduit
FR2810554B1 (fr) 2000-06-22 2003-05-16 Poudres & Explosifs Ste Nale Seringue sans aiguille munie d'un reservoir modulable
FR2812202B1 (fr) 2000-07-28 2002-09-13 Poudres & Explosifs Ste Nale Seringue sans aiguille fonctionnant par mise en compression du reservoir contenant le principe actif liquide
GB0025577D0 (en) * 2000-10-18 2000-12-06 Smithkline Beecham Biolog Vaccine
FR2815544B1 (fr) 2000-10-23 2003-02-14 Poudres & Explosifs Ste Nale Seringue sans aiguille securisee a architecture compacte
EP1201250A1 (en) 2000-10-25 2002-05-02 SMITHKLINE BEECHAM BIOLOGICALS s.a. Immunogenic compositions comprising liver stage malarial antigens
PT1361890E (pt) 2001-02-23 2011-06-07 Glaxosmithkline Biolog Sa Formulações vacinais de influenza para distribuição intradérmica
MY134424A (en) * 2001-05-30 2007-12-31 Saechsisches Serumwerk Stable influenza virus preparations with low or no amount of thiomersal
US7361352B2 (en) * 2001-08-15 2008-04-22 Acambis, Inc. Influenza immunogen and vaccine
US6861410B1 (en) 2002-03-21 2005-03-01 Chiron Corporation Immunological adjuvant compositions
CA2478930A1 (en) * 2002-04-01 2003-10-16 Euro-Celtique, S.A. Epitope constructs comprising antigen presenting cell targeting mechanisms
EP1594536B1 (en) 2003-01-30 2009-03-25 Novartis Vaccines and Diagnostics, Inc. Adjuvanted influenza vaccine
WO2005107797A1 (en) * 2004-03-09 2005-11-17 Chiron Corporation Influenza virus vaccines
KR20070116650A (ko) * 2005-03-23 2007-12-10 글락소스미스클라인 바이오로지칼즈 에스.에이. 조성물
US20090028903A1 (en) * 2005-03-23 2009-01-29 Glaxosmithkline Biologicals, S.A. Novel use
AR054822A1 (es) 2005-07-07 2007-07-18 Sanofi Pasteur Emulsion inmuno adyuvante
AU2006310163B2 (en) 2005-11-04 2011-09-15 Seqirus UK Limited Influenza vaccine with reduced amount of oil-in-water emulsion as adjuvant
FR2896162B1 (fr) 2006-01-13 2008-02-15 Sanofi Pasteur Sa Emulsion huile dans eau thermoreversible
US9101578B2 (en) 2006-05-01 2015-08-11 Technovax, Inc. Polyvalent influenza virus-like particle (VLP) compositions
SI2422810T1 (sl) * 2006-07-17 2015-01-30 Glaxosmithkline Biologicals S.A. Influenčno cepivo
WO2008009309A1 (en) 2006-07-17 2008-01-24 Glaxosmithkline Biologicals S.A. Influenza vaccine
ES2397714T3 (es) * 2006-10-12 2013-03-08 Glaxosmithkline Biologicals S.A. Vacuna que comprende un adyuvante de emulsión de aceite en agua
PE20090146A1 (es) * 2007-04-20 2009-03-23 Glaxosmithkline Biolog Sa Composicion inmunogenica contra el virus influenza
EP2227251A1 (en) * 2007-12-06 2010-09-15 GlaxoSmithKline Biologicals SA Influenza composition

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376369A (en) * 1987-11-03 1994-12-27 Syntex (U.S.A.) Inc. Vaccine adjuvant
US20030095974A1 (en) * 1997-09-05 2003-05-22 Smithkline Beecham Biologicals S.A. Oil in water emulsions containing saponins
US7323182B2 (en) * 1997-09-05 2008-01-29 Smithkline Beecham Biologicals Sg Oil in water emulsions containing saponins
US6451320B1 (en) * 1998-03-09 2002-09-17 Smithkline Beecham Biologicals S.A. Combined hepatitis and herpesvirus antigen compositions

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070141078A1 (en) * 1999-09-30 2007-06-21 D Hondt Erik Influenza vaccine
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
US9278127B2 (en) 2006-07-17 2016-03-08 Glaxosmithkline Biologicals, Sa Influenza vaccine
US9943588B2 (en) 2006-07-17 2018-04-17 Glaxosmithkline Biologicals S.A. Influenza vaccine
US9452209B2 (en) * 2007-04-20 2016-09-27 Glaxosmithkline Biologicals Sa Influenza vaccine
US10548969B2 (en) 2007-04-20 2020-02-04 Glaxosmithkline Biologicals Sa Oil-in-water emulsion influenza vaccine
US10016495B2 (en) 2007-04-20 2018-07-10 Glaxosmithkline Biologicals S.A. Oil-in-water emulsion influenza vaccine
US20100189741A1 (en) * 2007-04-20 2010-07-29 William Ripley Ballou Oil-in-water emulsion influenza vaccine
US9597389B2 (en) 2007-04-20 2017-03-21 Glaxosmithkline Biologicals Sa Oil-in-water emulsion influenza vaccine
US20090136543A1 (en) * 2007-04-20 2009-05-28 William Ripley Ballou Vaccine
US20100166785A1 (en) * 2007-12-07 2010-07-01 National Health Research Institutes Adjuvants
US20090176273A1 (en) * 2007-12-07 2009-07-09 National Health Research Institutes Production of Lipidated Proteins In E. coli
US8466259B2 (en) 2007-12-07 2013-06-18 National Health Research Institutes Adjuvants
US8426163B2 (en) 2007-12-07 2013-04-23 National Health Research Institutes Production of lipidated proteins in E. coli
US8287880B2 (en) 2009-06-02 2012-10-16 National Health Research Institutes Lipidated vaccine against dengue virus infection
US20100303849A1 (en) * 2009-06-02 2010-12-02 National Health Research Institutes Lipidated Vaccine against Dengue Virus Infection
US8658176B2 (en) 2009-06-22 2014-02-25 National Health Research Institutes Lipidated tumor-associated antigens and immunotherapeutic compositions
US20100322953A1 (en) * 2009-06-22 2010-12-23 National Health Research Institutes Lipidated Tumor-Associated Antigens and Immunotherapeutic Compositions
US20110034996A1 (en) * 2009-08-06 2011-02-10 Alexander Borck Medical implant containing an antioxidative substance
US8486434B2 (en) * 2009-08-06 2013-07-16 Biotronik Vi Patent Ag Medical implant containing an antioxidative substance
EP2464341A2 (en) * 2009-08-12 2012-06-20 Sigmoid Pharma Limited Immunomodulatory compositions comprising a polymer matrix and an oil phase
EP2464341B1 (en) * 2009-08-12 2022-07-06 Sublimity Therapeutics Limited Immunomodulatory compositions comprising a polymer matrix and an oil phase
US9878034B2 (en) 2009-10-02 2018-01-30 Mucosis B.V. Adjuvanted vaccine formulations
CN105617374A (zh) * 2009-10-02 2016-06-01 缪科西斯私人有限公司 含佐剂疫苗制剂
EP2308506A1 (en) * 2009-10-02 2011-04-13 Mucosis B.V. Adjuvanted intranasal vaccine formulations
CN102648001A (zh) * 2009-10-02 2012-08-22 缪科西斯私人有限公司 含佐剂疫苗制剂
WO2011040811A1 (en) * 2009-10-02 2011-04-07 Mucosis B.V. Adjuvanted vaccine formulations
AU2010301213B2 (en) * 2009-10-02 2013-11-07 Mucosis B.V. Adjuvanted vaccine formulations
DE102009056871A1 (de) * 2009-12-03 2011-06-22 Novartis AG, 4056 Impfstoff-Adjuvantien und verbesserte Verfahren zur Herstellung derselben
WO2011088451A1 (en) * 2010-01-15 2011-07-21 Novavax, Inc. Uses of influenza virus-like particles (vlps) for characterization of neuraminidase and hemagglutinin activity
WO2012066420A1 (en) * 2010-11-15 2012-05-24 National Health Research Institutes Lipidated polyepitope vaccines
US8771990B2 (en) 2010-11-15 2014-07-08 National Health Research Institutes Method of producing lipidated polypeptides
US8658767B2 (en) 2010-11-15 2014-02-25 National Health Research Institutes Lipidated polyepitope vaccines

Also Published As

Publication number Publication date
EP1861122A1 (en) 2007-12-05
MA29342B1 (fr) 2008-03-03
IL185906A0 (en) 2008-01-06
JP2008534465A (ja) 2008-08-28
HUE027837T2 (en) 2016-11-28
JP2014129359A (ja) 2014-07-10
MX2007011748A (es) 2008-03-11
PE20061300A1 (es) 2006-12-23
CA2603180A1 (en) 2006-09-28
EP1861120A1 (en) 2007-12-05
MX2007011775A (es) 2008-03-11
MA29715B1 (fr) 2008-09-01
NZ561823A (en) 2010-04-30
AR053833A1 (es) 2007-05-23
SI1861120T1 (sl) 2016-09-30
JP2008534466A (ja) 2008-08-28
EA200701787A1 (ru) 2008-08-29
TW200700079A (en) 2007-01-01
AU2006226458B2 (en) 2012-08-30
AR052625A1 (es) 2007-03-21
US20110287054A1 (en) 2011-11-24
KR20140004815A (ko) 2014-01-13
KR101916787B1 (ko) 2019-01-24
TW200722101A (en) 2007-06-16
MA30298B1 (fr) 2009-04-01
AR054020A1 (es) 2007-05-30
KR20070116650A (ko) 2007-12-10
JP5770414B2 (ja) 2015-08-26
IL185901A0 (en) 2008-01-06
IL185897A0 (en) 2008-01-06
KR20070116651A (ko) 2007-12-10
WO2006100111A1 (en) 2006-09-28
EA200701785A1 (ru) 2008-04-28
WO2006100109A1 (en) 2006-09-28
NZ561822A (en) 2010-04-30
PL1861120T3 (pl) 2016-11-30
EA011393B1 (ru) 2009-02-27
NO20074635L (no) 2007-12-19
AU2006226543A1 (en) 2006-09-28
HRP20160816T1 (hr) 2016-08-12
EA011419B1 (ru) 2009-02-27
US20080181911A1 (en) 2008-07-31
PE20061428A1 (es) 2007-01-16
AU2006226543B2 (en) 2011-10-06
CA2601022C (en) 2023-03-07
TW200700078A (en) 2007-01-01
WO2006100110A1 (en) 2006-09-28
AU2006226459A1 (en) 2006-09-28
NO20074638L (no) 2007-12-19
EP1863529A1 (en) 2007-12-12
KR20070116652A (ko) 2007-12-10
KR20160064249A (ko) 2016-06-07
CY1117874T1 (el) 2017-05-17
PE20061387A1 (es) 2007-01-19
ES2585810T3 (es) 2016-10-10
US20140363474A1 (en) 2014-12-11
CA2602456A1 (en) 2006-09-28
EP1861120B1 (en) 2016-05-25
EA200701786A1 (ru) 2008-04-28
AU2006226458A1 (en) 2006-09-28
US9730999B2 (en) 2017-08-15
CA2603180C (en) 2015-10-13
US20080171063A1 (en) 2008-07-17
DK1861120T3 (en) 2016-07-25
MX2007011756A (es) 2008-03-11
IL185897A (en) 2015-11-30
NO20074930L (no) 2007-12-07
PT1861120T (pt) 2016-08-18
JP2008534467A (ja) 2008-08-28
EP2397153A1 (en) 2011-12-21
JP5869744B2 (ja) 2016-02-24
CA2601022A1 (en) 2006-09-28

Similar Documents

Publication Publication Date Title
US10143745B2 (en) Vaccine compositions comprising a saponin adjuvant
US20090081253A1 (en) Composition
US20090028903A1 (en) Novel use
BRPI0609519A2 (pt) composição, uso de uma composição, método de vacinação, uso de um antìgeno, e, método para a preparação de uma composição imunogênica

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLAXOSMITHKLINE BIOLOGICALS SA, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HANON, EMMANUEL JULES;STEPHENNE, JEAN;REEL/FRAME:019937/0694;SIGNING DATES FROM 20060418 TO 20060419

STCB Information on status: application discontinuation

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