WO2009071613A2 - Vaccine - Google Patents

Vaccine Download PDF

Info

Publication number
WO2009071613A2
WO2009071613A2 PCT/EP2008/066762 EP2008066762W WO2009071613A2 WO 2009071613 A2 WO2009071613 A2 WO 2009071613A2 EP 2008066762 W EP2008066762 W EP 2008066762W WO 2009071613 A2 WO2009071613 A2 WO 2009071613A2
Authority
WO
WIPO (PCT)
Prior art keywords
protein
seq
vaccine
viral vector
ade2
Prior art date
Application number
PCT/EP2008/066762
Other languages
French (fr)
Other versions
WO2009071613A3 (en
Inventor
Joseph D. Cohen
Martine Marchand
Original Assignee
Glaxosmithkline Biologicals S.A.
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=40591828&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2009071613(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to MX2010006207A priority Critical patent/MX2010006207A/en
Priority to CA2707245A priority patent/CA2707245A1/en
Priority to AU2008333208A priority patent/AU2008333208A1/en
Priority to CN2008801267101A priority patent/CN101939438A/en
Priority to BRPI0819889-6A priority patent/BRPI0819889A2/en
Application filed by Glaxosmithkline Biologicals S.A. filed Critical Glaxosmithkline Biologicals S.A.
Priority to AP2010005295A priority patent/AP2010005295A0/en
Priority to JP2010536451A priority patent/JP2011505796A/en
Priority to EP08857977A priority patent/EP2227550A2/en
Publication of WO2009071613A2 publication Critical patent/WO2009071613A2/en
Publication of WO2009071613A3 publication Critical patent/WO2009071613A3/en
Priority to IL205953A priority patent/IL205953A0/en
Priority to ZA2010/03851A priority patent/ZA201003851B/en
Priority to MA32991A priority patent/MA32003B1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/002Protozoa antigens
    • A61K39/015Hemosporidia antigens, e.g. Plasmodium antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5256Virus expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55577Saponins; Quil A; QS21; ISCOMS
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a simian derived adenoviral vector particularly encoding a new malaria antigen derived from the circumsporozoite protein of Plasmodium falciparum.
  • the invention further relates to processes of preparing said viral vector and use of same in the treatment/prevention of malaria infection.
  • Malaria is one of the world's major health problems with more than 2 to 4 million people dying from the disease each year.
  • One of the most acute forms of the disease is caused by the protozoan parasite,
  • Plasmodium falciparum (P '. falciparum) which is responsible for most of the mortality attributable to malaria.
  • the life cycle of P. falciparum is complex, requiring two hosts, man and mosquito for completion.
  • the infection of man is initiated by the inoculation of sporozoites in the bloodstream through the bite of an infected mosquito.
  • the sporozoites migrate to the liver and there infect hepatocytes where they differentiate, via the exoerythrocytic intracellular stage, into the merozoite stage which infects red blood cells (RBC) to initiate cyclical replication in the asexual blood stage.
  • RBC red blood cells
  • the cycle is completed by the differentiation of a number of merozoites in the RBC into sexual stage gametocytes, which are ingested by the mosquito, where they develop through a series of stages in the midgut to produce sporozoites which migrate to the salivary gland.
  • the sporozoite stage of Plasmodium has been identified as a potential target of a malaria vaccine.
  • Vaccination with deactivated (irradiated) sporozoite has been shown to induce protection against experimental human malaria (Am. J, Trap. Med. Hyg 24: 297-402, 1975).
  • CS protein circumsporozoite protein
  • the CS protein of Plasmodia species is characterized by a central repetitive domain (repeat region) flanked by non-repetitive amino (N-terminus) and carboxy (C- terminus) fragments.
  • RTS lipoprotein particle
  • S This particle contains a portion of the CS protein of P. falciparum substantially as corresponding to amino acids 207-395 of the CS protein of P. falciparum (strain NF54[3D7]) fused to the N-terminal of the S antigen from Hepatitis B.
  • the S antigen may comprise a portion of the preS2.
  • adeno virus The RTS, S particle is usually delivered along with a strong adjuvant.
  • malaria vaccines have been proposed employing recombinant adenoviral vectors, for example WO 2004/055187 describes certain viral vectors including specific adeno 5 (Ad5) and adeno 35 (Ad 35) vectors, both derived from human adeno viruses, encoding CS protein.
  • Ad5 specific adeno 5
  • Ad 35 adeno 35 vectors
  • Ad5 is associated with mild respiratory infections in children
  • Ad4 and Ad7 are thought to be associated with respiratory infections in adults
  • Ad40 is thought to cause diarrhoea in infants. Immunity to adenovirus infections is thought to be life-long following infection.
  • Ad5 and Ad35 may result in the neutralisation of therapeutic adenoviral vectors based on human adeno viruses. This may reduce the therapeutic effectiveness of the vector as the vector is prevented from entering cells and manufacturing the relevant antigen in vivo.
  • the present invention is thought to reduce the issues of pre-existing immunity by providing a vaccine for prevention and/or treatment of malaria comprising: a replication deficient simian adenoviral vector C7 (also referred to as Pan 7 or CV- 33) encoding a protein comprising CS protein from P. falciparum or a fragment thereof, for example as shown in Seq ID No: 1 or Seq ID No: 3.
  • a replication deficient simian adenoviral vector C7 also referred to as Pan 7 or CV- 33
  • a protein comprising CS protein from P. falciparum or a fragment thereof for example as shown in Seq ID No: 1 or Seq ID No: 3.
  • Seq ID No: 1 An amino acid sequence of a protein/antigen derived from CS protein of P '. falciparum (referred to herein as Ade2 protein)
  • Seq ID No: 2 A nucleic acid sequence encoding the protein of Seq ID
  • Ade2 gene No: 1 (referred herein as Ade2 gene)
  • Seq ID No: 3 An alternative amino acid sequence of a protein/antigen derived from CS protein of P. falciparum (referred herein as Adel protein)
  • Seq ID No: 4 A nucleic acid sequence encoding the protein of Seq ID
  • Seq ID No: 5 Capsid protein sequence from Chimp Adeno 7(seq ID No 17 from WO 03/046124)
  • Seq ID No: 6 An amino acid sequence from Chimp Adeno 7(seq ID No 20 from WO 03/046124)
  • Seq ID No: 7 An amino acid sequence from P. falciparum CS protein
  • Seq ID No: 8 An amino acid sequence from P. falciparum CS protein
  • Seq ID No: 9 An amino acid sequence from P. falciparum CS protein
  • Seq ID No: 10 An amino acid sequence from P. falciparum CS protein
  • Seq ID No: 17 Shows the nucleotide sequence of an alternative expression cassette to Ade2 expression cassette and cloned into C7 adenoviral vector
  • Seq ID No: 18 Shows the nucleotide sequence of the Ade2 expression cassette, cloned into C7 adenoviral vector
  • Seq ID No: 19 Shows the complete nucleotide sequence of the synthetic recombinant vector C7 -Ade2.
  • Figure 1 Shows a plasmid map for pCR2.1-Ade2
  • Figure 2 Shows the plasmid map for pShuttle6-Ade2
  • Figure 3 Shows plasmid maps for pC7000-CMV Ade2
  • 4 to 7 Show comparison between the CS-specif ⁇ c T cell responses induced by C7 Adel & C7 Ade2 in C57B1/6 mice.
  • Figures 8 to 11 Show comparison between the CS-specif ⁇ c T cell responses induced by C7 Ade2 and Ad5 Ade2 in C57B1/6 mice.
  • Figures 12 to 15 Show comparison between the CS-specif ⁇ c T cell responses induced by C7 Adel and Ad5 Adel in C57B1/6 mice.
  • Figure 16 Shows anti-CS antibody responses determined by ELISA in C57B1/6 mice.
  • Figures 17 and 18 Show kinetics of the CS-specif ⁇ c CD8 T cell responses induced by C7-Ade2 in CB6F1 mice.
  • Figures 19 and 20 Show kinetics of the CS-specif ⁇ c CD4 T cell responses induced by C7-Ade2 in CB6F1 mice.
  • Figures 21 and 22 Show cytokine profile of the CS-specific CD8 T cell responses induced by C7-Ade2 in CB6F1 mice.
  • Figures 23 and 24 Show cytokine profile of the CS-specific CD4 T cell responses induced by C7-Ade2 in CB6F1 mice.
  • Figures 25 and 26 Show kinetics of the CS-specific CD8 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
  • Figures 27 and 28 Show kinetics of the CS-specific CD4 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
  • Figure 29 Shows kinetics of the HBs-specific CD8 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
  • Figurre 30 Shows kinetics of the HBs-specif ⁇ c CD4 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
  • Figures 31 and 32 Show cytokine profile of the CS-specific CD8 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
  • Figures 34 and 35 Cytokine profile of the CS-specific CD4 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
  • FIG. 36 Cytokine profile of the HBs-specific CD4 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
  • Figures 37 and 38 Show antibody responses antibody responses induced by C7- Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
  • Figures 39 to 41 Show kinetics of the CS- & HBs-specific CD8 T cell responses induced by co-formulation C7-Ade2+ RTS,S/AS01B in CB6F1 mice.
  • Figures 42 to 44 Show kinetics of the CS- & HBs-specific CD4 T cell responses induced by co-formulation C7-Ade2+ RTS,S/AS01B in CB6F1 mice.
  • Figures 45 to 47 Show cytokine profile of the CS- & HBs-specific CD8 T cell responses induced by co-formulation C7-Ade2+ RTS,S/AS01B in CB6F1 mice.
  • Figures 48 to 50 Show cytokine profile of the CS- & HBs-specific CD4 T cell responses induced by co-formulation C7-Ade2+ RTS,S/AS01B in CB6F1 mice.
  • Figures 51 and 52 Show antibody responses induced by co-formulation C7- Ade2+ RTS,S/AS01B in CB6F1 mice.
  • Figures 53 to 55 Show kinetics of the CS- & HBs-specific CD8 T cell responses induced by C7-Ade2, RTS,S and ASOlB responses in CB6F1 mice.
  • Figures 56 to 58 Show kinetics of the CS- & HBs-specific CD4 T cell responses induced by C7-Ade2, RTS,S and ASOlB responses in CB6F1 mice.
  • Figures 59 to 61 Show cytokine profile of the CS- & HBs-specific CD8 T cell responses induced by C7-Ade2, RTS, S and ASOlB responses in CB6F1 mice.
  • Figures 62 to 64 Show cytokine profile of the CS- & HBs-specific CD4 T cell responses induced by C7-Ade2, RTS, S and ASOlB responses in CB6F1 mice.
  • Figures 65 and 66 Show antibody responses induced by C7-Ade2, RTS, S and ASOlB responses in CB6F1 mice.
  • Figures 67 and 68 Show kinetics of the CS-specific CD8 T cell responses induced by synthetic C7 Ade2 in CB6F1 mice.
  • Figures 69 and 70 Show kinetics of the CS-specific CD4 T cell responses induced by synthetic C7 Ade2 in CB6F1 mice.
  • Figures 71 and 72 Show cytokine profile of the CS-specific CD8 T cell responses induced by synthetic C7 Ade2 in CB6F1 mice.
  • Figures 73 and 74 Show cytokine profile of the CS-specific CD4 T cell responses induced by synthetic C7 Ade2 in CB6F1 mice.
  • adenoviral vectors The characteristics and properties of any given adenoviral vector are often individual, although there is a hypothesis that vectors may be grouped into families and that adenoviral vectors within a given family may have similar characteristics.
  • C7 is thought to be particularly advantageous as it seems to be more stable once the protein encoding gene is inserted than certain other known vectors, for example C6 also described in WO 2003/046124. That is to say C7 is thought to be less prone to re-organisation.
  • any adenoviral vector employed in a vaccine is stable because pharmaceutical products need to be well characterised and shown to be stable and safe before they can be marketed.
  • Pre-existing immunity to C7 is thought to be very low and thus the risk of neutralisation of the viral vector after the first administration to a patient is low.
  • the invention employs a synthetic C7 viral vector, which may be particularly suitable for gaining regulatory approval for administration to humans.
  • the malaria antigen component from the CS protein has the last 12 to 14 amino acids removed.
  • the malaria antigen encoded by the adenoviral vector is modified to remove potential glycosylation sites, for example the amino acid alanine may replace a serine, such as shown in position about 379 of Seq ID No: 1.
  • the protein/antigen employed comprises the following amino acids; NNGDNGREGKDEDKRDGNN [Seq ID No: 7] optionally located at about amino acid 81 to 99.
  • protein/antigen encoded comprises the amino acids:
  • AIGL (Seq ID No: 8] for example at the C terminus.
  • the invention employs a protein comprising the following amino acids:
  • the invention employs a protein comprising the following amino acids:
  • the protein/antigen employed comprises the sequences of Seq ID No. 7 and/or Seq ID No. 8 and/or Seq ID No 9.
  • the protein/antigen employed comprises the sequences of Seq ID No. 7 and/or Seq ID No. 8 and/or Seq ID No 10.
  • the protein/antigen encoded is Seq ID No: 1 or 3.
  • the protein sequence given in Seq ID No: 1 is new and forms an aspect of the invention.
  • Polynucleotide encoding the protein sequence of Seq ID No:l also forms an aspect of the invention, in particular the polynucleotide sequence of Seq ID No: 2.
  • This polynucleotide sequence (ID No: 2) is already codon-optimized for expression in, humans.
  • a polynucleotide sequence encoding the protein of Seq ID No: 1 may be codon-optimized.
  • the invention also extends to vectors/plasmids/hosts employed in the preparation of the novel hybrid fusion protein of Seq ID No: 1 or employed in the preparation of a viral vector according to the invention.
  • a suitable plasmid can be employed to insert the sequence encoding for the protein into a suitable host for synthesis.
  • An example of a suitable plasmid is pRIT 15546 a 2 micron-based vector for carrying a suitable expression cassette.
  • the plasmid will generally contain an inbuilt marker to assist selection, for example a gene encoding for antibiotic resistance or LEU2 or HIS auxotrophy.
  • Host cells can be prokaryotic or eukaryotic but preferably, are yeast, for example Saccharomyces (for example Saccharomyces cerevisiae such as DC5 in ATCC data base (accession number 20820), under the name RIT DC5 cir(o). Depositor: Smith Kline-RITj and non- Saccharomyces yeasts.
  • Saccharomyces for example Saccharomyces cerevisiae such as DC5 in ATCC data base (accession number 20820), under the name RIT DC5 cir(o).
  • Depositor Smith Kline-RITj and non- Saccharomyces yeasts.
  • Schizosaccharomyces eg Schizosaccharomyces pombe
  • Kluyveromyces eg Kluyveromyces lactis
  • Pichia eg Pichiapastoris
  • Hansenula eg Hansenula polymorpha
  • Yarrowia eg Yarrowia lipolytica
  • Schwanniomyces eg Schwanniomyces occidentalis
  • the invention provides use of the vectors according to the invention or a protein of Seq ID No 1 for the treatment or prevention of malaria.
  • the invention provides a pharmaceutical formulation comprising a viral vector according to the invention and an excipient such as an isotonic carrier suitable for injection. Suitable excipients are discussed in more detail below.
  • a formulation comprises: • an adeno viral vector according to the invention,
  • a malaria antigen such as a lipoprotein particle particularly RTS, S, and
  • an adjuvant for example comprising a saponin and/or 3D-MPL.
  • the vector When the vector encodes the sequence of Seq ID No: 1 the vector is particularly suitable for use in a treatment regime with the protein known as RTS, S. This is because the protein encoded by the adenoviral vector corresponds as closely as possible to the "RT" component in RTS, S. Use of the vector in a regime with RTS, S is thought to have the ability to reinforce efficiently the efficacy of RTS, S.
  • the viral vectors described herein are suitable for use as component for a malaria vaccine.
  • the viral vectors of the invention may need to be used in combination with other components including other antigens to provide adequate protection against infection. Nevertheless the vectors of the present invention are suitable for use at least as a component of vaccine or treatment regime.
  • RTS RTS
  • S can be prepared as described in WO 93/10152 (eg from P. falciparum NF54/3D7 strain).
  • the nucleotide sequence for the RTS expression cassette and predicted translation product is provided in Figure 9 of WO 93/10152 (referred to therein as RTS*).
  • excipient refers to a component in a pharmaceutical formulation with no therapeutic effect in its own right.
  • a diluent or carrier falls within the definition of an excipient. Suitable carriers include PBS, saline and the like.
  • Adjuvants are also within this definition of excipient because whilst adjuvants may have a physiological effect in vivo this effect is general and in the absence of a therapeutic component is not a specific therapeutic effect.
  • Adjuvants are those selected from the group of metal salts, oil in water emulsions, Toll like receptors agonist, (in particular Toll like receptor 2 agonist, Toll like receptor 3 agonist, Toll like receptor 4 agonist, Toll like receptor 7 agonist, Toll like receptor 8 agonist and Toll like receptor 9 agonist), saponins or combinations thereof.
  • the adjuvant is a Toll like receptor (TLR) 4 ligand, for example an agonist such as a lipid A derivative particularly monophosphoryl lipid A or more particularly 3-deacylated monophoshoryl lipid A (3D - MPL).
  • TLR Toll like receptor
  • 3-Deacylated monophosphoryl lipid A is known from US patent No. 4,912,094 and UK patent application No. 2,220,211 (Ribi) and is available from Ribi Immunochem, Montana, USA.
  • 3D-MPL is sold under the trademark MPL® by Corixa corporation and primarily promotes CD4+ T cell responses with an IFN-g (ThI) 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. Generally in the compositions of the present invention small particle 3D-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. Synthetic derivatives of lipid A are known and thought to be TLR 4 agonists including, but not limited to:
  • OM 294 DP (3 S, 9 R) -3-[(R)-dodecanoyloxytetradecanoylamino]-4-oxo-5-aza-9(R)- [(R)-3-hydroxytetradecanoyl amino] decan- 1 , 10-diol, 1 , 10-bis(dihydrogenophosphate) (WO99 /64301 and WO 00/0462 ), OM 197 MP-Ac DP ( 3S-, 9R) -3-[(R) -dodecanoyloxytetradecanoylamino]-4-oxo-5- aza-9-[(R)-3-hydroxytetradecanoylamino]decan- 1 , 10-diol, 1 -dihydrogenophosphate 10-(6-aminohexanoate) (WO 01/46127).
  • 3D-MPL typically when 3D-MPL is used the antigen and 3D-MPL are delivered in an oil in water emulsion or multiple oil in water emulsions.
  • the incorporation of 3D-MPL is advantageous since it is a stimulator of effector T-cells responses.
  • TLR4 ligands which may be used are alkyl Glucosaminide phosphates (AGPs) such as those disclosed in WO 9850399 or US 6303347 (processes for preparation of AGPs are also disclosed), or pharmaceutically acceptable salts of AGPs as disclosed in US 6764840.
  • AGPs alkyl Glucosaminide phosphates
  • Some AGPs are TLR4 agonists, and some are TLR4 antagonists. Both are thought to be useful as adjuvants.
  • Another immunostimulant for use in the present invention is Quil A and its derivatives.
  • Quil A is a saponin preparation isolated from the South American tree Quilaja Saponaria Molina and was first described as having adjuvant activity by Dalsgaard et al.
  • QS7 and QS21 are natural saponin derived from the bark of Quillaja saponaria Molina which induces CD8+ cytotoxic T cells (CTLs), ThI cells and a predominant IgG2a antibody response.
  • QS21 which further comprise a sterol (WO 96/33739).
  • the ratio of QS21 : sterol will typically be in the order of 1 :100 to 1 : 1 weight to weight. Generally an excess of sterol is present, the ratio of QS21 : sterol being at least 1 : 2 w/w.
  • QS21 and sterol will be present in a vaccine in the range of about 1 ⁇ g to about 100 ⁇ g, such as about 10 ⁇ g to about 50 ⁇ g per dose.
  • Liposomal formulations generally contain a neutral lipid, for example phosphatidylcholine, which is usually non-crystalline at room temperature, for example eggyolk phosphatidylcholine, dioleoyl phosphatidylcholine or dilauryl phosphatidylcholine.
  • the liposomes may also contain a charged lipid which increases the stability of the lipsome-QS21 structure for liposomes composed of saturated lipids. In these cases the amount of charged lipid is often 1-20% w/w, such as 5-10%.
  • the ratio of sterol to phospholipid is 1-50% (mol/mol), such as 20-25%.
  • compositions may contain MPL (3-deacylated mono-phosphoryl lipid A, also known as 3D-MPL).
  • 3D-MPL is known from GB 2 220 211 (Ribi) as a mixture of 3 types of de-O-acylated monophosphoryl lipid A with 4, 5 or 6 acylated chains and is manufactured by Ribi Immunochem, Montana.
  • the saponins may in the form of micelles, mixed micelles (generally, but not exclusively with bile salts) or may be in the form of ISCOM matrices (EP 0 109 942), liposomes or related colloidal structures such as worm-like or ring-like multimeric complexes or lipidic/layered structures and lamellae when formulated with cholesterol and lipid, or in the form of an oil in water emulsion (for example as in WO 95/17210).
  • the saponin is presented in the form of a liposomal formulation, ISCOM or an oil in water emulsion.
  • Immunostimulatory oligonucleotides may also be used.
  • oligonucleotides for use in adjuvants or vaccines of the present invention include CpG containing oligonucleotides, generally containing two or more dinucleotide CpG motifs separated by at least three, more preferably at least six or more nucleotides.
  • a CpG motif is a Cytosine nucleotide followed by a Guanine nucleotide.
  • the CpG oligonucleotides are typically deoxynucleotides.
  • the internucleotide in the oligonucleotide is phosphorodithioate, or more preferably a phosphorothioate bond, although phosphodiester and other internucleotide bonds are within the scope of the invention.
  • oligonucleotides with mixed internucleotide linkages are included within the scope of the invention. Methods for producing phosphorothioate oligonucleotides or phosphorodithioate are described in US 5,666,153, US 5,278,302 and WO 95/26204.
  • oligonucleotides are as follows:
  • Alternative CpG oligonucleotides may comprise one or more sequences above in that they have inconsequential deletions or additions thereto.
  • the CpG oligonucleotides may be synthesized by any method known in the art (for example see EP 468520). Conveniently, such oligonucleotides may be synthesized utilising an automated synthesizer.
  • TLR 2 agonist examples include peptidoglycan or lipoprotein.
  • Imidazoquinolines such as Imiquimod and Resiquimod are known TLR7 agonists.
  • Single stranded RNA is also a known TLR agonist (TLR8 in humans and TLR7 in mice), whereas double stranded RNA and poly IC (polyinosinic-polycytidylic acid - a commercial synthetic mimetic of viral RNA) are exemplary of TLR 3 agonists.
  • 3D- MPL is an example of a TLR4 agonist whilst CpG is an example of a TLR9 agonist.
  • An immunostimulant may alternatively or in addition be included. In a one embodiment this immunostimulant will be 3-deacylated monophosphoryl lipid A (3D- MPL).
  • the adjuvant comprises 3D-MPL.
  • the adjuvant comprises QS21.
  • the adjuvant comprises CpG. In one aspect the adjuvant is formulated as an oil in water emulsion.
  • the adjuvant is formulated as liposomes.
  • Adjuvants combinations include 3D-MPL and QS21 (EP 0 671 948 Bl) oil in water emulsions or liposomal formulations comprising 3D-MPL and QS21 or 3D-MPL formulated with other carriers (EP 0 689 454 Bl).
  • Other preferred adjuvant systems comprise a combination of 3D-MPL, QS21 and a CpG oligonucleotide as described in US 6558670 and US 6544518.
  • Vaccine preparation is generally described in New Trends and Developments in Vaccines, edited by Voller et al., University Park Press, Baltimore, Maryland, U.S.A., 1978. Encapsulation within liposomes is described, for example, by Fullerton, U.S. Patent 4,235,877.
  • the formulations of the present invention may be used for both prophylactic and therapeutic purposes. Accordingly the invention provides a vaccine composition as described herein for use in medicine, for example, for the treatment and/or prophylaxis of malaria.
  • the invention provides a composition
  • a composition comprising a C7 adenoviral vector according to the invention and a malaria antigen such as RTS, S or the novel antigen of Seq ID No: 1 or virus like particles of the same and an excipient, optionally in the presence of an adjuvant.
  • a malaria antigen such as RTS, S or the novel antigen of Seq ID No: 1 or virus like particles of the same and an excipient, optionally in the presence of an adjuvant.
  • Immunogenic in the context of this specification is intended to refer to the ability to elicit an immune response, wherein said response is specific to a malaria component in the relevant formulation.
  • This response may require the presence of a suitable adjuvant and/or boosting.
  • a booster for example, comprising a dose similar or less than the original dose, may be required to obtain an appropriate immunogenic response.
  • composition/pharmaceutical formulations according to the invention may also include in admixture one or more further antigens such as those derived from P. falciparium and/or P. vivax, for example wherein the antigen is selected from DBP, PvTRAP, PvMSP2, PvMSP4, PvMSP5, PvMSP ⁇ , PvMSP7, PvMSP8, PvMSP9, PvAMAl and RBP or fragment thereof.
  • the antigen is selected from DBP, PvTRAP, PvMSP2, PvMSP4, PvMSP5, PvMSP ⁇ , PvMSP7, PvMSP8, PvMSP9, PvAMAl and RBP or fragment thereof.
  • antigens derived from P falciparum include, PfEMP-I, Pfs 16 antigen, MSP-I, MSP-3, LSA-I, LSA-3, AMA-I and TRAP.
  • Other Plasmodium antigens include P. falciparum EBA, GLURP, RAPl, RAP2, Sequestrin, PO32, STARP,
  • the invention also relates to use of C7 for encoding a malaria antigen, for example particularly as described herein for the treatment and/or prevention of malaria, or for the manufacture of a medicament for same.
  • the invention also includes a method of treatment comprising administering a therapeutically effective amount of one or more aspects of the invention.
  • the C7 viral vector according to the invention may be co-administered or co-formulated with a malaria antigen such as RTS, S or the antigen of Seq ID No. 1, optionally in the presence of an adjuvant for example comprising 3D-MPL and/or a saponin such as QS21.
  • the C7 vector may also be co-administered or co-formulated with another adenoviral vector of a different serotype and/or origin, encoding the same of different antigens.
  • the invention also extends to use of any aspect defined herein in a prime boost regime, for example wherein the priming dose or doses is/are given at a timpoint zero (and subsequent primes within for example 3 months) and a boost is given, for example at about 4, 5, 6, 7, 8, 9, 10, 11 or 12 weeks after the last priming dose, optionally with a further boosting shot or shots given up to one year after said first boosting shot.
  • a prime boost regime for example wherein the priming dose or doses is/are given at a timpoint zero (and subsequent primes within for example 3 months) and a boost is given, for example at about 4, 5, 6, 7, 8, 9, 10, 11 or 12 weeks after the last priming dose, optionally with a further boosting shot or shots given up to one year after said first boosting shot.
  • a further boosting shot or shots given up to one year after said first boosting shot.
  • one or more aspects of the invention including the combination vaccine described above, stimulate specific humoral (that is antibody responses) and/or cellular immune responses (such
  • This type of balanced immune response may be required to give so called sterile protection against malarial infection.
  • antibody responses for combinations may be augmented in relation to antibody responses to adjuvanted protein only regime schemes.
  • the invention provides use of C7 as the prime or boost in a prime boost regime with: • a C7 adenoviral vector encoding the same or different malaria antigen,
  • a human adenoviral vector such as Ad5 or Ad35 encoding a malaria antigen such as a CS protein from P. falciparum or simian adenoviral vector of a different serotype (ie not C7), and/or • a malaria antigen such as RTS, S and an adjuvant, for example comprising a saponin and/or 3D-MPL, as the complementary component of the regime.
  • a malaria antigen such as a CS protein from P. falciparum or simian adenoviral vector of a different serotype (ie not C7)
  • a malaria antigen such as RTS, S and an adjuvant, for example comprising a saponin and/or 3D-MPL, as the complementary component of the regime.
  • the invention also provides any of the aspects herein described for the manufacture of a medicament for the treatment and/or prevention of malarial infection.
  • the amount of 3D-MPL used is generally small, but depending on the vaccine formulation may be in the region of l-1000 ⁇ g per dose, for example l-500 ⁇ g per dose, and such as in the range 1 to lOO ⁇ g per dose, such as 50 or 25 ⁇ g per dose.
  • the amount of CpG or immunostimulatory oligonucleotides in the adjuvants or vaccines of the present invention is generally small, but depending on the vaccine formulation may be in the region of l-1000 ⁇ g per dose, for example l-500 ⁇ g per dose, and such as in the range 1 to lOO ⁇ g per dose.
  • the amount of saponin for use in the adjuvants of the present invention may be in the region of l-1000 ⁇ g per dose, for example l-500 ⁇ g per dose, such as l-250 ⁇ g per dose, and particularly in the range 1 to lOO ⁇ g per dose such as 50 or 25 ⁇ g per dose.
  • the dose may, for example be 1 to 500 ⁇ g such as 10 to 100 ⁇ g, particularly 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75 or 80 ⁇ g per dose.
  • the dose may, for example be 10 3 to 10 16 vpu such as 10 6 to 10 10 vpu.
  • the amounts employed for each component of the combination may correspond to the dose given for that component alone.
  • the invention also extends to kits comprising the elements employed in combinations according to the invention.
  • the invention further relates to a process for preparing an adenoviral vector according to the invention and formulations comprising the same.
  • the invention also relates to a method of producing the protein of Seq ID No 1.
  • the synthetic gene was prepared by a company Medigenomix.
  • the gene was cloned into pCR2.1 -TOPO-TA cloning vector (Invitrogen see Figure 1).
  • This vector was digested with Notl and BamHI and a recombinant shuttle plasmid vector (-Ade2) was created.
  • the map of the shuttle plasmid is shown in Figure 2.
  • the expression cassette contains the cytomegalovirus (CMV) early promoter and first exon, an intron derived from the plasmid pCI (purchased from Promega) the DNA encoding Ade2, and the rabbit globin polyadenylation signal.
  • the complete cassette is flanked by recognition sites for the restriction enzymes I-Ceul and PI-SceI respectively.
  • the expression cassettes were excised from the shuttle plasmid using I- Ceul and PI-SceI and introduced into a plasmid molecular clone of an El deleted genome of SAdV-24 (ie C7) - pC7 000 pkGFP as described (Roy et al. Hum Gene Ther. (2004) 5 :519-530) to obtain the plasmid shown in Figure 3
  • the plasmid molecular clone DNA was linearized by digesting with the restriction enzyme Pad and transfected into HEK 293 cells to rescue recombinant adenovirus.
  • the adenoviruses were propagated, amplified and purified using standard techniques.
  • C57B1/6 mice were immunized once intramuscularly with a dose range (1OeIO, 10e9, 10e8 viral particles) of the C7 chimpadenoviruses expressing either of the construct Adel or Ade2.
  • some mice were immunized with the human adenovirus 5 (at the dose of 10e9 and 10e8) expressing either of the construct Adel or Ade2.
  • negative controls some mice were immunized with empty C7 & empty Ad5 viral vectors.
  • Peripheral blood was collected and pooled on days 14, 28, 34 and 49 post- immunization and the Ag-specific CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest, i.e. the N-terminal region (N-term) or C-terminal region (C-term) of the CS protein. As negative controls, some cells were also cultured overnight in vitro in culture medium (unstimulated). The Ag-specific responses were calculated by subtracting the average cytokine response produced by unstimulated cells from the average cytokine response produced by the peptide-stimulated cells.
  • CB6F1 mice were immunized once intramuscularly with a dose range (1OeIO, 10e9, 10e8 viral particles) of the C7 chimpadeno virus expressing the Ade2 construct (5 pools of mice/group).
  • Peripheral blood was collected and pooled on days 21, 28 and 35 post-immunization and the CS C-term and CS N-term specific CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest, i.e. the N-terminal region (N-term) or C-terminal region (C-term) of the CS protein.
  • N-term N-terminal region
  • C-term C-terminal region
  • the profile displayed on d28 post-immunization is shown in Figures 21 to 24 and is representative of the other tested timepoints.
  • the CS-specific CD8 T cell responses are mostly composed of CD8 T cells producing IFNg only ( Figures 21 & 22).
  • the CS-specific CD4 T cell responses are also composed of IFNg producing CD4 T cells but also and to a lesser extent of CD4 T cells producing IL2 only or both IL2 & IFNg ( Figures 23 & 24).
  • ASOlB is an adjuvant system containing 3D- MPL and QS21 formulated with liposomes. Mice were immunized intramuscularly on d0, 14 and 28 as follows:
  • Peripheral blood was collected and pooled on days 21 (7d pll), 35 (7d pill), 49(2 Id pill), 63 (35d pIII),77 (49d pill) post-immunization and the CS C-term, CS N-term and HBs specific CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest (CS N-term, CS C-term or HBs). As negative controls, some cells were also cultured overnight in vitro in culture medium (unstimulated). The Ag-specific responses were calculated by subtracting the average cytokine response produced by unstimulated cells from the average cytokine response produced by the peptide-stimulated cells.
  • N-term specific CD8 T cell responses are observed in the CCC group, followed by the APPgroup.
  • 2 Id pill, 35d pill & 49d pill these response are & remain of similar intensity in the APP, PPA and CCC groups ( Figure 25).
  • N-term specific CD4 T cell responses are mainly detected in mice immunized with APP or CCC with higher intensities of such responses in the APP group ( Figure 27)
  • the HBs-specific CD4 and CD8T cell responses are higher in the animals immunized with PPP than with APP, PPA or CCC ( Figures 29 and 30).
  • the CCC treatment regimen is the only one that is associated with the simultaneous induction of CS and HBs-specific CD4 and CD8 T cell responses.
  • the cytokine profiles of the CS- and HBs- specific CD4 and CD8 T cell responses were also determined and were similar across the timepoints tested. The ones from the 21 d pill timepoint are shown below as representative of all timepoints tested ( Figures 31 to 36).
  • the Ag-specif ⁇ c CD8 T cell responses are mostly composed of CD8 T cells producing IFNg ( Figures 31 to 33).
  • the Ag-specif ⁇ c CD4 T cell responses are composed of a mixture of CD4 T cells producing IFNg, IFNg and IL-2 and to a lesser extent CD4 T cells producing IL-2 ( Figures 34 to 36).
  • the Ag-specif ⁇ c antibody responses were determined by ELISA on sera collected 14 and 42 days post-3 rd immunization.
  • the total Ig responses against the R32LR polypeptide i.e. which covers the middle portion of P '. falciparum CSP
  • HBs HBs-specif ⁇ c antibody responses
  • Peripheral blood was collected and pooled on days 35, 42, 49, 63 and 98 and the CS C-term, CS N-term and HBs specific CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest, i.e. the N-terminal region (N-term), the C-terminal region (C-term) of the CS protein or HBs. As negative controls, some cells were also cultured overnight in vitro in culture medium (unstimulated). The Ag-specific responses were calculated by subtracting the average cytokine response produced by unstimulated cells from the average cytokine response produced by the peptide-stimulated cells.
  • the cytokine profiles of the CS- and HBs- specific CD4 and CD8 T cell responses were also determined and were similar across the timepoints tested. The ones from the day 42 of the study are shown below as representative of all timepoints tested ( Figures 42 to 47). Briefly, the Ag-specific CD8 T cell responses are mostly composed of CD8 T cells producing IFNg ( Figures 45 to 47). In contrast, the Ag-specific CD4 T cell responses are composed of a mixture of CD4 T cells producing IFNg, IFNg and IL-2 and to a lesser extent CD4 T cells producing IL-2 ( Figures 48 to 50).
  • the Ag-specific antibody responses were determined by ELISA on sera collected on day 56 and 99 of the study.
  • the total Ig responses against the R32LR polypeptide i.e. which covers the middle portion of P. falciparum CSP
  • All immunization regimens did elicit R32LR and HBs-specific antibody responses: within each group, these responses were of similar intensity at both timepoints tested.
  • the groups immunized with the combo were compared, there was a trend for higher responses in groups immunized with 3 doses of the combo ( Figures 51 & 52).
  • Peripheral blood was collected and pooled on days 14, 28, 70, 91 & 112 and the CS C-term, CS N-term and HBs specific CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest, i.e. the
  • N-term N-terminal region (N-term), the C-terminal region (C-term) of the CS protein or HBs.
  • the Ag-specific antibody responses were determined by ELISA on sera collected on day 42 and 84 of the study.
  • the total Ig responses against the R32LR polypeptide i.e. which covers the middle portion of P. falciparum CSP
  • HBs HBs
  • the average cytokine profiles of the CS- and HBs- specific CD4 and CD8 T cell responses were also determined at each timepoint of the study and these are shown in Figures 59 to 64.
  • the Ag-specific CD8 T cell responses are mostly composed of CD8 T cells producing IFNg ( Figures 59 to 61).
  • the Ag-specific CD4 T cell responses are composed of a mixture of CD4 T cells producing IFNg, IFNg and IL-2 and to a lesser extent CD4 T cells producing IL-2 ( Figures 62 to 64).
  • CB6F1 mice (6 pools of mice/group) were immunized with 10e9 vp of the original C7 Ade2 or its synthetic counterpart.
  • Peripheral blood was collected and pooled on days 21, 28 & 35 post-immunization and the CS C-term and CS N-term CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest, i.e. the N-terminal region (N-term), the C-terminal region (C-term) of the CS protein. As negative controls, some cells were also cultured overnight in vitro in culture medium (unstimulated). The Ag-specific responses were calculated by subtracting the average cytokine response produced by unstimulated cells from the average cytokine response produced by the peptide-stimulated cells.
  • N-term specific ( Figures 67 and 68).
  • the N-term and C-term specific CD4 T cell responses were of similar intensity regardless of the viral vector used ( Figures 69 and
  • the cytokine profiles of the CS-specific CD4 and CD8 T cell responses were also determined and are shown in Figures 71 to 74. Briefly, the Ag-specific CD8 T cell responses were mostly composed of CD8 T cells producing IFNg ( Figures 71 & 72).
  • the Ag-specific CD4 T cell responses were composed of a mixture of CD4
  • GIy lie Ser lie Thr Lys Asp GIy lie GIn Leu GIy Thr Asp Ser Asp
  • GIy GIn Ala lie Tyr Ala Asp GIu Thr Tyr GIn Pro GIu Pro GIn VaI
  • GIu Thr GIy GIy Thr Lys GIu Tyr Asp lie Asp Met Ala Phe Phe Asp
  • Ade2 expression cassette Sequence of Ade2 expression cassette is below. The protein encoding region is underlined.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biotechnology (AREA)
  • Public Health (AREA)
  • Microbiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Molecular Biology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)

Abstract

The invention relates to a replication deficient simian adenoviral vector C7 encoding a protein comprising CS protein from P. falciparum or a fragment thereof, for example as shown in Seq ID No: 1 or Seq ID No: 3. The invention also relates to processes of preparing said viral vector and use of the viral vector in the treatment/prevention of malaria infection. Compositions, vaccines and kits comprising said viral vector are also described. In one aspect the invention employs a synthetic C7 viral vector. The C7 viral vector according to the invention may be co-administered or co-formulated with a malaria antigen such as RTS,S optionally in the presence of an adjuvant for example comprising 3D-MPL and/or a saponin such as QS21.

Description

Vaccine
The present invention relates to a simian derived adenoviral vector particularly encoding a new malaria antigen derived from the circumsporozoite protein of Plasmodium falciparum. The invention further relates to processes of preparing said viral vector and use of same in the treatment/prevention of malaria infection.
Malaria, is one of the world's major health problems with more than 2 to 4 million people dying from the disease each year. One of the most acute forms of the disease is caused by the protozoan parasite,
Plasmodium falciparum (P '. falciparum) which is responsible for most of the mortality attributable to malaria.
The life cycle of P. falciparum is complex, requiring two hosts, man and mosquito for completion. The infection of man is initiated by the inoculation of sporozoites in the bloodstream through the bite of an infected mosquito. The sporozoites migrate to the liver and there infect hepatocytes where they differentiate, via the exoerythrocytic intracellular stage, into the merozoite stage which infects red blood cells (RBC) to initiate cyclical replication in the asexual blood stage. The cycle is completed by the differentiation of a number of merozoites in the RBC into sexual stage gametocytes, which are ingested by the mosquito, where they develop through a series of stages in the midgut to produce sporozoites which migrate to the salivary gland.
The sporozoite stage of Plasmodium has been identified as a potential target of a malaria vaccine. Vaccination with deactivated (irradiated) sporozoite has been shown to induce protection against experimental human malaria (Am. J, Trap. Med. Hyg 24: 297-402, 1975). However, it is has not been possible practically and logistically to manufacture a vaccine for malaria for the general population based on this methodology, employing irradiated sporozoites.
The major surface protein of the sporozoite is known as circumsporozoite protein (CS protein). It is thought to be involved in the motility and invasion of the sporozoite during its passage from the initial site of inoculation by the mosquito into the circulation, where it migrates to the liver.
The CS protein of Plasmodia species is characterized by a central repetitive domain (repeat region) flanked by non-repetitive amino (N-terminus) and carboxy (C- terminus) fragments. To date the most advanced malaria vaccine in the clinic is based on a lipoprotein particle (also known as a virus like particle) referred to as RTS, S. This particle contains a portion of the CS protein of P. falciparum substantially as corresponding to amino acids 207-395 of the CS protein of P. falciparum (strain NF54[3D7]) fused to the N-terminal of the S antigen from Hepatitis B. The S antigen may comprise a portion of the preS2.
The RTS, S particle is usually delivered along with a strong adjuvant. Nevertheless malaria vaccines have been proposed employing recombinant adenoviral vectors, for example WO 2004/055187 describes certain viral vectors including specific adeno 5 (Ad5) and adeno 35 (Ad 35) vectors, both derived from human adeno viruses, encoding CS protein. There are more than 40 different serotypes of human adeno viruses, which vary in their pathogenicity, for example Ad5 is associated with mild respiratory infections in children, Ad4 and Ad7 are thought to be associated with respiratory infections in adults, and Ad40 is thought to cause diarrhoea in infants. Immunity to adenovirus infections is thought to be life-long following infection. It is thought that pre-existing immunity to particularly Ad5 and Ad35 may result in the neutralisation of therapeutic adenoviral vectors based on human adeno viruses. This may reduce the therapeutic effectiveness of the vector as the vector is prevented from entering cells and manufacturing the relevant antigen in vivo.
The present invention is thought to reduce the issues of pre-existing immunity by providing a vaccine for prevention and/or treatment of malaria comprising: a replication deficient simian adenoviral vector C7 (also referred to as Pan 7 or CV- 33) encoding a protein comprising CS protein from P. falciparum or a fragment thereof, for example as shown in Seq ID No: 1 or Seq ID No: 3.
Brief description of the Sequences
Seq ID No: 1 An amino acid sequence of a protein/antigen derived from CS protein of P '. falciparum (referred to herein as Ade2 protein)
Seq ID No: 2 A nucleic acid sequence encoding the protein of Seq ID
No: 1 (referred herein as Ade2 gene)
Seq ID No: 3 An alternative amino acid sequence of a protein/antigen derived from CS protein of P. falciparum (referred herein as Adel protein)
Seq ID No: 4 A nucleic acid sequence encoding the protein of Seq ID
No: 3 (referred herein as Adel gene)
Seq ID No: 5 Capsid protein sequence from Chimp Adeno 7(seq ID No 17 from WO 03/046124)
Seq ID No: 6 An amino acid sequence from Chimp Adeno 7(seq ID No 20 from WO 03/046124)
Seq ID No: 7 An amino acid sequence from P. falciparum CS protein
Seq ID No: 8 An amino acid sequence from P. falciparum CS protein
Seq ID No: 9 An amino acid sequence from P. falciparum CS protein
Seq ID No: 10 An amino acid sequence from P. falciparum CS protein
Seq ID No: 11 Nucleotide sequence for CpG 1826
Seq ID No: 12 Nucleotide sequence for CpG 1758
Seq ID No: 13 Nucleotide sequence for a CpG
Seq ID No: 14 Nucleotide sequence for CpG 2006
Seq ID No: 15 Nucleotide sequence for CpG 1668
Seq ID No: 16 Nucleotide sequence for CpG 5456
Seq ID No: 17 Shows the nucleotide sequence of an alternative expression cassette to Ade2 expression cassette and cloned into C7 adenoviral vector Seq ID No: 18 Shows the nucleotide sequence of the Ade2 expression cassette, cloned into C7 adenoviral vector
Seq ID No: 19 Shows the complete nucleotide sequence of the synthetic recombinant vector C7 -Ade2.
Brief description of the Figures.
Figure 1 Shows a plasmid map for pCR2.1-Ade2 Figure 2 Shows the plasmid map for pShuttle6-Ade2 Figure 3 Shows plasmid maps for pC7000-CMV Ade2 Figures 4 to 7 Show comparison between the CS-specifϊc T cell responses induced by C7 Adel & C7 Ade2 in C57B1/6 mice.
Figures 8 to 11 Show comparison between the CS-specifϊc T cell responses induced by C7 Ade2 and Ad5 Ade2 in C57B1/6 mice. Figures 12 to 15 Show comparison between the CS-specifϊc T cell responses induced by C7 Adel and Ad5 Adel in C57B1/6 mice. Figure 16 Shows anti-CS antibody responses determined by ELISA in C57B1/6 mice. Figures 17 and 18 Show kinetics of the CS-specifϊc CD8 T cell responses induced by C7-Ade2 in CB6F1 mice. Figures 19 and 20 Show kinetics of the CS-specifϊc CD4 T cell responses induced by C7-Ade2 in CB6F1 mice. Figures 21 and 22 Show cytokine profile of the CS-specific CD8 T cell responses induced by C7-Ade2 in CB6F1 mice. Figures 23 and 24 Show cytokine profile of the CS-specific CD4 T cell responses induced by C7-Ade2 in CB6F1 mice. Figures 25 and 26 Show kinetics of the CS-specific CD8 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
Figures 27 and 28 Show kinetics of the CS-specific CD4 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice. Figure 29 Shows kinetics of the HBs-specific CD8 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice. Figurre 30 Shows kinetics of the HBs-specifϊc CD4 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
Figures 31 and 32 Show cytokine profile of the CS-specific CD8 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
Figure 33 Cytokine profile of the HBs-specific CD8 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
Figures 34 and 35 Cytokine profile of the CS-specific CD4 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
Figure 36 Cytokine profile of the HBs-specific CD4 T cell responses induced by C7-Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice. Figures 37 and 38 Show antibody responses antibody responses induced by C7- Ade2 in prime/boost or co-formulation with RTS,S/AS01B in CB6F1 mice.
Figures 39 to 41 Show kinetics of the CS- & HBs-specific CD8 T cell responses induced by co-formulation C7-Ade2+ RTS,S/AS01B in CB6F1 mice.
Figures 42 to 44 Show kinetics of the CS- & HBs-specific CD4 T cell responses induced by co-formulation C7-Ade2+ RTS,S/AS01B in CB6F1 mice.
Figures 45 to 47 Show cytokine profile of the CS- & HBs-specific CD8 T cell responses induced by co-formulation C7-Ade2+ RTS,S/AS01B in CB6F1 mice.
Figures 48 to 50 Show cytokine profile of the CS- & HBs-specific CD4 T cell responses induced by co-formulation C7-Ade2+ RTS,S/AS01B in CB6F1 mice. Figures 51 and 52 Show antibody responses induced by co-formulation C7- Ade2+ RTS,S/AS01B in CB6F1 mice.
Figures 53 to 55 Show kinetics of the CS- & HBs-specific CD8 T cell responses induced by C7-Ade2, RTS,S and ASOlB responses in CB6F1 mice.
Figures 56 to 58 Show kinetics of the CS- & HBs-specific CD4 T cell responses induced by C7-Ade2, RTS,S and ASOlB responses in CB6F1 mice. Figures 59 to 61 Show cytokine profile of the CS- & HBs-specific CD8 T cell responses induced by C7-Ade2, RTS, S and ASOlB responses in CB6F1 mice.
Figures 62 to 64 Show cytokine profile of the CS- & HBs-specific CD4 T cell responses induced by C7-Ade2, RTS, S and ASOlB responses in CB6F1 mice.
Figures 65 and 66 Show antibody responses induced by C7-Ade2, RTS, S and ASOlB responses in CB6F1 mice.
Figures 67 and 68 Show kinetics of the CS-specific CD8 T cell responses induced by synthetic C7 Ade2 in CB6F1 mice.
Figures 69 and 70 Show kinetics of the CS-specific CD4 T cell responses induced by synthetic C7 Ade2 in CB6F1 mice.
Figures 71 and 72 Show cytokine profile of the CS-specific CD8 T cell responses induced by synthetic C7 Ade2 in CB6F1 mice.
Figures 73 and 74 Show cytokine profile of the CS-specific CD4 T cell responses induced by synthetic C7 Ade2 in CB6F1 mice.
The sequence and preparation of C7 is described in WO 2003/046124. Sequence ID Nos: 6 (penton sequence), 9 (nucleic acid sequence), 10 & 11 (hexon sequence), & 12 (fibre protein) of WO 2003/046124 are incorporated by reference. The deposit number for C7 is [ATCC VR-593].
The characteristics and properties of any given adenoviral vector are often individual, although there is a hypothesis that vectors may be grouped into families and that adenoviral vectors within a given family may have similar characteristics.
Employing C7 is thought to be particularly advantageous as it seems to be more stable once the protein encoding gene is inserted than certain other known vectors, for example C6 also described in WO 2003/046124. That is to say C7 is thought to be less prone to re-organisation. Of course it is very important that any adenoviral vector employed in a vaccine is stable because pharmaceutical products need to be well characterised and shown to be stable and safe before they can be marketed.
Pre-existing immunity to C7 is thought to be very low and thus the risk of neutralisation of the viral vector after the first administration to a patient is low.
Furthermore, there are thought to be one or more other properties of C7 that are likely to make it particularly suitable for administration to humans and/or for generating a favourable immune response in vivo.
In one aspect the invention employs a synthetic C7 viral vector, which may be particularly suitable for gaining regulatory approval for administration to humans. In one aspect the malaria antigen component from the CS protein has the last 12 to 14 amino acids removed.
In one aspect the malaria antigen encoded by the adenoviral vector is modified to remove potential glycosylation sites, for example the amino acid alanine may replace a serine, such as shown in position about 379 of Seq ID No: 1.
In one aspect the invention the protein/antigen employed comprises the following amino acids; NNGDNGREGKDEDKRDGNN [Seq ID No: 7] optionally located at about amino acid 81 to 99.
In one aspect the protein/antigen encoded comprises the amino acids:
AIGL [Seq ID No: 8] for example at the C terminus. In one aspect the invention employs a protein comprising the following amino acids:
PNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNVDPN ANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNAN PNANPNANPNANPNANPNANPN
[Seq ID No: 9]
In one aspect the invention employs a protein comprising the following amino acids:
NANP NVDP NANP NVDP NANP NVDP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NVDP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP
[Seq ID No: 10] In a further aspect the protein/antigen employed comprises the sequences of Seq ID No. 7 and/or Seq ID No. 8 and/or Seq ID No 9.
In a further aspect the protein/antigen employed comprises the sequences of Seq ID No. 7 and/or Seq ID No. 8 and/or Seq ID No 10.
In one aspect the protein/antigen encoded is Seq ID No: 1 or 3.
The protein sequence given in Seq ID No: 1 is new and forms an aspect of the invention.
Polynucleotide encoding the protein sequence of Seq ID No:l also forms an aspect of the invention, in particular the polynucleotide sequence of Seq ID No: 2. This polynucleotide sequence (ID No: 2) is already codon-optimized for expression in, humans. Optionally a polynucleotide sequence encoding the protein of Seq ID No: 1 may be codon-optimized.
The invention also extends to vectors/plasmids/hosts employed in the preparation of the novel hybrid fusion protein of Seq ID No: 1 or employed in the preparation of a viral vector according to the invention.
When preparation and isolation of the protein is required a suitable plasmid can be employed to insert the sequence encoding for the protein into a suitable host for synthesis. An example of a suitable plasmid is pRIT 15546 a 2 micron-based vector for carrying a suitable expression cassette. The plasmid will generally contain an inbuilt marker to assist selection, for example a gene encoding for antibiotic resistance or LEU2 or HIS auxotrophy.
Host cells can be prokaryotic or eukaryotic but preferably, are yeast, for example Saccharomyces (for example Saccharomyces cerevisiae such as DC5 in ATCC data base (accession number 20820), under the name RIT DC5 cir(o). Depositor: Smith Kline-RITj and non- Saccharomyces yeasts. These include Schizosaccharomyces (eg Schizosaccharomyces pombe) Kluyveromyces (eg Kluyveromyces lactis), Pichia (eg Pichiapastoris), Hansenula (eg Hansenula polymorpha), Yarrowia (eg Yarrowia lipolytica) and Schwanniomyces (eg Schwanniomyces occidentalis).
In one aspect the invention provides use of the vectors according to the invention or a protein of Seq ID No 1 for the treatment or prevention of malaria. In one aspect the invention provides a pharmaceutical formulation comprising a viral vector according to the invention and an excipient such as an isotonic carrier suitable for injection. Suitable excipients are discussed in more detail below.
In one embodiment a formulation comprises: • an adeno viral vector according to the invention,
• a malaria antigen such as a lipoprotein particle particularly RTS, S, and
• optionally an adjuvant for example comprising a saponin and/or 3D-MPL.
When the vector encodes the sequence of Seq ID No: 1 the vector is particularly suitable for use in a treatment regime with the protein known as RTS, S. This is because the protein encoded by the adenoviral vector corresponds as closely as possible to the "RT" component in RTS, S. Use of the vector in a regime with RTS, S is thought to have the ability to reinforce efficiently the efficacy of RTS, S. The viral vectors described herein are suitable for use as component for a malaria vaccine. The viral vectors of the invention may need to be used in combination with other components including other antigens to provide adequate protection against infection. Nevertheless the vectors of the present invention are suitable for use at least as a component of vaccine or treatment regime. RTS9S
RTS, S can be prepared as described in WO 93/10152 (eg from P. falciparum NF54/3D7 strain). The nucleotide sequence for the RTS expression cassette and predicted translation product is provided in Figure 9 of WO 93/10152 (referred to therein as RTS*).
In the context of this specification excipient, refers to a component in a pharmaceutical formulation with no therapeutic effect in its own right. A diluent or carrier falls within the definition of an excipient. Suitable carriers include PBS, saline and the like. Adjuvants are also within this definition of excipient because whilst adjuvants may have a physiological effect in vivo this effect is general and in the absence of a therapeutic component is not a specific therapeutic effect.
Adjuvants Particular adjuvants are those selected from the group of metal salts, oil in water emulsions, Toll like receptors agonist, (in particular Toll like receptor 2 agonist, Toll like receptor 3 agonist, Toll like receptor 4 agonist, Toll like receptor 7 agonist, Toll like receptor 8 agonist and Toll like receptor 9 agonist), saponins or combinations thereof.
In an embodiment the adjuvant is a Toll like receptor (TLR) 4 ligand, for example an agonist such as a lipid A derivative particularly monophosphoryl lipid A or more particularly 3-deacylated monophoshoryl lipid A (3D - MPL).
3-Deacylated monophosphoryl lipid A is known from US patent No. 4,912,094 and UK patent application No. 2,220,211 (Ribi) and is available from Ribi Immunochem, Montana, USA.
3D-MPL is sold under the trademark MPL® by Corixa corporation and primarily promotes CD4+ T cell responses with an IFN-g (ThI) 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. Generally in the compositions of the present invention small particle 3D-MPL is used. Small particle 3D-MPL has a particle size such that it may be sterile-filtered through a 0.22μm filter. Such preparations are described in WO 94/21292. Synthetic derivatives of lipid A are known and thought to be TLR 4 agonists including, but not limited to:
OM174 (2-deoxy-6-O-[2-deoxy-2-[(R)-3-dodecanoyloxytetra-decanoylamino]-4-o- phosphono-β-D-glucopyranosyl]-2-[(R)-3-hydroxytetradecanoylamino]-α-D- glucopyranosyldihydrogenphosphate), (WO 95/14026)
OM 294 DP (3 S, 9 R) -3-[(R)-dodecanoyloxytetradecanoylamino]-4-oxo-5-aza-9(R)- [(R)-3-hydroxytetradecanoyl amino] decan- 1 , 10-diol, 1 , 10-bis(dihydrogenophosphate) (WO99 /64301 and WO 00/0462 ), OM 197 MP-Ac DP ( 3S-, 9R) -3-[(R) -dodecanoyloxytetradecanoylamino]-4-oxo-5- aza-9-[(R)-3-hydroxytetradecanoylamino]decan- 1 , 10-diol, 1 -dihydrogenophosphate 10-(6-aminohexanoate) (WO 01/46127). Typically when 3D-MPL is used the antigen and 3D-MPL are delivered in an oil in water emulsion or multiple oil in water emulsions. The incorporation of 3D-MPL is advantageous since it is a stimulator of effector T-cells responses.
Other TLR4 ligands which may be used are alkyl Glucosaminide phosphates (AGPs) such as those disclosed in WO 9850399 or US 6303347 (processes for preparation of AGPs are also disclosed), or pharmaceutically acceptable salts of AGPs as disclosed in US 6764840. Some AGPs are TLR4 agonists, and some are TLR4 antagonists. Both are thought to be useful as adjuvants. Another immunostimulant for use in the present invention is Quil A and its derivatives. Quil A is a saponin preparation isolated from the South American tree Quilaja Saponaria Molina and was first described as having adjuvant activity by Dalsgaard et al. in 1974 ("Saponin adjuvants", Archiv. fur die gesamte Virusforschung, Vol. 44, Springer Verlag, Berlin, p243-254). Purified fragments of Quil A have been isolated by HPLC which retain adjuvant activity without the toxicity associated with Quil A (EP 0 362 278), for example QS7 and QS21 (also known as QA7 and QA21). QS21 is a natural saponin derived from the bark of Quillaja saponaria Molina which induces CD8+ cytotoxic T cells (CTLs), ThI cells and a predominant IgG2a antibody response.
Particular formulations of QS21 have been described which further comprise a sterol (WO 96/33739). The ratio of QS21 : sterol will typically be in the order of 1 :100 to 1 : 1 weight to weight. Generally an excess of sterol is present, the ratio of QS21 : sterol being at least 1 : 2 w/w. Typically for human administration QS21 and sterol will be present in a vaccine in the range of about 1 μg to about 100 μg, such as about 10 μg to about 50 μg per dose.
Liposomal formulations generally contain a neutral lipid, for example phosphatidylcholine, which is usually non-crystalline at room temperature, for example eggyolk phosphatidylcholine, dioleoyl phosphatidylcholine or dilauryl phosphatidylcholine. The liposomes may also contain a charged lipid which increases the stability of the lipsome-QS21 structure for liposomes composed of saturated lipids. In these cases the amount of charged lipid is often 1-20% w/w, such as 5-10%. The ratio of sterol to phospholipid is 1-50% (mol/mol), such as 20-25%.
These compositions may contain MPL (3-deacylated mono-phosphoryl lipid A, also known as 3D-MPL). 3D-MPL is known from GB 2 220 211 (Ribi) as a mixture of 3 types of de-O-acylated monophosphoryl lipid A with 4, 5 or 6 acylated chains and is manufactured by Ribi Immunochem, Montana. The saponins may in the form of micelles, mixed micelles (generally, but not exclusively with bile salts) or may be in the form of ISCOM matrices (EP 0 109 942), liposomes or related colloidal structures such as worm-like or ring-like multimeric complexes or lipidic/layered structures and lamellae when formulated with cholesterol and lipid, or in the form of an oil in water emulsion (for example as in WO 95/17210).
Usually, the saponin is presented in the form of a liposomal formulation, ISCOM or an oil in water emulsion.
Immunostimulatory oligonucleotides may also be used. Examples oligonucleotides for use in adjuvants or vaccines of the present invention include CpG containing oligonucleotides, generally containing two or more dinucleotide CpG motifs separated by at least three, more preferably at least six or more nucleotides. A CpG motif is a Cytosine nucleotide followed by a Guanine nucleotide. The CpG oligonucleotides are typically deoxynucleotides. In one embodiment the internucleotide in the oligonucleotide is phosphorodithioate, or more preferably a phosphorothioate bond, although phosphodiester and other internucleotide bonds are within the scope of the invention. Also included within the scope of the invention are oligonucleotides with mixed internucleotide linkages. Methods for producing phosphorothioate oligonucleotides or phosphorodithioate are described in US 5,666,153, US 5,278,302 and WO 95/26204.
Examples of oligonucleotides are as follows:
TCC ATG ACG TTC CTG ACG TT (CpG 1826) [Seq ID No: 11]
TCT CCC AGC GTG CGC CAT (CpG 1758) [Seq ID No: 12] ACC GAT GAC GTC GCC GGT GAC GGC ACC ACG [Seq ID No: 13]
TCG TCG TTT TGT CGT TTT GTC GTT (CpG 2006) [Seq ID No: 14]
TCC ATG ACG TTC CTG ATG CT (CpG 1668) [Seq ID No: 15]
TCG ACG TTT TCG GCG CGC GCC G (CpG 5456), [Seq ID No: 16] the sequences may contain phosphorothioate modified internucleotide linkages.
Alternative CpG oligonucleotides may comprise one or more sequences above in that they have inconsequential deletions or additions thereto.
The CpG oligonucleotides may be synthesized by any method known in the art (for example see EP 468520). Conveniently, such oligonucleotides may be synthesized utilising an automated synthesizer.
Examples of a TLR 2 agonist include peptidoglycan or lipoprotein.
Imidazoquinolines, such as Imiquimod and Resiquimod are known TLR7 agonists. Single stranded RNA is also a known TLR agonist (TLR8 in humans and TLR7 in mice), whereas double stranded RNA and poly IC (polyinosinic-polycytidylic acid - a commercial synthetic mimetic of viral RNA) are exemplary of TLR 3 agonists. 3D- MPL is an example of a TLR4 agonist whilst CpG is an example of a TLR9 agonist. An immunostimulant may alternatively or in addition be included. In a one embodiment this immunostimulant will be 3-deacylated monophosphoryl lipid A (3D- MPL).
In one aspect the adjuvant comprises 3D-MPL.
In one aspect the adjuvant comprises QS21.
In one aspect the adjuvant comprises CpG. In one aspect the adjuvant is formulated as an oil in water emulsion.
In one aspect the adjuvant is formulated as liposomes.
Adjuvants combinations include 3D-MPL and QS21 (EP 0 671 948 Bl) oil in water emulsions or liposomal formulations comprising 3D-MPL and QS21 or 3D-MPL formulated with other carriers (EP 0 689 454 Bl). Other preferred adjuvant systems comprise a combination of 3D-MPL, QS21 and a CpG oligonucleotide as described in US 6558670 and US 6544518.
Formulations
Vaccine preparation is generally described in New Trends and Developments in Vaccines, edited by Voller et al., University Park Press, Baltimore, Maryland, U.S.A., 1978. Encapsulation within liposomes is described, for example, by Fullerton, U.S. Patent 4,235,877.
The formulations of the present invention may be used for both prophylactic and therapeutic purposes. Accordingly the invention provides a vaccine composition as described herein for use in medicine, for example, for the treatment and/or prophylaxis of malaria.
In one aspect the invention provides a composition comprising a C7 adenoviral vector according to the invention and a malaria antigen such as RTS, S or the novel antigen of Seq ID No: 1 or virus like particles of the same and an excipient, optionally in the presence of an adjuvant.
Immunogenic in the context of this specification is intended to refer to the ability to elicit an immune response, wherein said response is specific to a malaria component in the relevant formulation. This response may require the presence of a suitable adjuvant and/or boosting. A booster, for example, comprising a dose similar or less than the original dose, may be required to obtain an appropriate immunogenic response.
The composition/pharmaceutical formulations according to the invention may also include in admixture one or more further antigens such as those derived from P. falciparium and/or P. vivax, for example wherein the antigen is selected from DBP, PvTRAP, PvMSP2, PvMSP4, PvMSP5, PvMSPβ, PvMSP7, PvMSP8, PvMSP9, PvAMAl and RBP or fragment thereof.
Other example, antigens derived from P falciparum include, PfEMP-I, Pfs 16 antigen, MSP-I, MSP-3, LSA-I, LSA-3, AMA-I and TRAP. Other Plasmodium antigens include P. falciparum EBA, GLURP, RAPl, RAP2, Sequestrin, PO32, STARP,
SALSA, PfEXPl, Pfs25, Pfs28, PFS27/25, Pfs48/45, Pfs230 and their analogues in other Plasmodium spp.
The invention also relates to use of C7 for encoding a malaria antigen, for example particularly as described herein for the treatment and/or prevention of malaria, or for the manufacture of a medicament for same.
The invention also includes a method of treatment comprising administering a therapeutically effective amount of one or more aspects of the invention. Optionally the C7 viral vector according to the invention may be co-administered or co-formulated with a malaria antigen such as RTS, S or the antigen of Seq ID No. 1, optionally in the presence of an adjuvant for example comprising 3D-MPL and/or a saponin such as QS21. The C7 vector may also be co-administered or co-formulated with another adenoviral vector of a different serotype and/or origin, encoding the same of different antigens.
The invention also extends to use of any aspect defined herein in a prime boost regime, for example wherein the priming dose or doses is/are given at a timpoint zero (and subsequent primes within for example 3 months) and a boost is given, for example at about 4, 5, 6, 7, 8, 9, 10, 11 or 12 weeks after the last priming dose, optionally with a further boosting shot or shots given up to one year after said first boosting shot. Advantageously one or more aspects of the invention, including the combination vaccine described above, stimulate specific humoral (that is antibody responses) and/or cellular immune responses (such as CD8+ and/or CD4+) such as antibody responses and CD 8+ and/or CD4+ responses, particularly CD8+ and antibody responses. That is to say responses specific to the CS protein and/or S antigen (as appropriate).
This type of balanced immune response may be required to give so called sterile protection against malarial infection. Furthermore antibody responses for combinations may be augmented in relation to antibody responses to adjuvanted protein only regime schemes.
In one embodiment the invention provides use of C7 as the prime or boost in a prime boost regime with: • a C7 adenoviral vector encoding the same or different malaria antigen,
• another viral vector such as a human adenoviral vector such as Ad5 or Ad35 encoding a malaria antigen such as a CS protein from P. falciparum or simian adenoviral vector of a different serotype (ie not C7), and/or • a malaria antigen such as RTS, S and an adjuvant, for example comprising a saponin and/or 3D-MPL, as the complementary component of the regime.
The invention also provides any of the aspects herein described for the manufacture of a medicament for the treatment and/or prevention of malarial infection.
Quantities
The amount of 3D-MPL used is generally small, but depending on the vaccine formulation may be in the region of l-1000μg per dose, for example l-500μg per dose, and such as in the range 1 to lOOμg per dose, such as 50 or 25μg per dose.
The amount of CpG or immunostimulatory oligonucleotides in the adjuvants or vaccines of the present invention is generally small, but depending on the vaccine formulation may be in the region of l-1000μg per dose, for example l-500μg per dose, and such as in the range 1 to lOOμg per dose.
The amount of saponin for use in the adjuvants of the present invention may be in the region of l-1000μg per dose, for example l-500μg per dose, such as l-250μg per dose, and particularly in the range 1 to lOOμg per dose such as 50 or 25μg per dose.
When protein is administered the dose may, for example be 1 to 500 μg such as 10 to 100 μg, particularly 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75 or 80 μg per dose.
When adenoviral vectors are administered the dose may, for example be 103 to 1016 vpu such as 106 to 1010 vpu.
When a combination is employed then the amounts employed for each component of the combination may correspond to the dose given for that component alone. The invention also extends to kits comprising the elements employed in combinations according to the invention.
The invention further relates to a process for preparing an adenoviral vector according to the invention and formulations comprising the same.
The invention also relates to a method of producing the protein of Seq ID No 1.
In the context of this specification comprising is to be interpreted as including. The invention extends to embodiments which correspond to embodiments described herein as comprising certain element but consisting or consisting essentially of the relevant elements.
Discussion in the background section of this specification is provided for the purpose of putting the invention in context. It is not to be taken as admission about what is known in the art and in particular is not an admission of what constitutes common general knowledge. The examples below are shown to illustrate the methodology, which may be employed in the invention.
EXAMPLES
Example 1
The synthetic gene was prepared by a company Medigenomix. The gene was cloned into pCR2.1 -TOPO-TA cloning vector (Invitrogen see Figure 1). This vector was digested with Notl and BamHI and a recombinant shuttle plasmid vector (-Ade2) was created. The map of the shuttle plasmid is shown in Figure 2.
Description of the expression cassette and methodology for virus rescue
The expression cassette contains the cytomegalovirus (CMV) early promoter and first exon, an intron derived from the plasmid pCI (purchased from Promega) the DNA encoding Ade2, and the rabbit globin polyadenylation signal. The complete cassette is flanked by recognition sites for the restriction enzymes I-Ceul and PI-SceI respectively. The expression cassettes were excised from the shuttle plasmid using I- Ceul and PI-SceI and introduced into a plasmid molecular clone of an El deleted genome of SAdV-24 (ie C7) - pC7 000 pkGFP as described (Roy et al. Hum Gene Ther. (2004) 5 :519-530) to obtain the plasmid shown in Figure 3
The plasmid molecular clone DNA was linearized by digesting with the restriction enzyme Pad and transfected into HEK 293 cells to rescue recombinant adenovirus. The adenoviruses were propagated, amplified and purified using standard techniques.
The sequences of the expression cassette for (Ade2) from the I-Ceul to the PI-Sce recognition sites is shown in Seq ID Nos: 18.
Example 2 Immunogenicitv of the C7-Adel & C7-Ade2 in C57B1/6 mice
C57B1/6 mice were immunized once intramuscularly with a dose range (1OeIO, 10e9, 10e8 viral particles) of the C7 chimpadenoviruses expressing either of the construct Adel or Ade2. As positive controls, some mice were immunized with the human adenovirus 5 (at the dose of 10e9 and 10e8) expressing either of the construct Adel or Ade2. As negative controls, some mice were immunized with empty C7 & empty Ad5 viral vectors.
Peripheral blood was collected and pooled on days 14, 28, 34 and 49 post- immunization and the Ag-specific CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest, i.e. the N-terminal region (N-term) or C-terminal region (C-term) of the CS protein. As negative controls, some cells were also cultured overnight in vitro in culture medium (unstimulated). The Ag-specific responses were calculated by subtracting the average cytokine response produced by unstimulated cells from the average cytokine response produced by the peptide-stimulated cells.
The results indicate that under the experimental conditions described above, both constructs induced CS-specifϊc CD4 and CD8 T cell responses (Figures 4 to 15). Of note, C-term specific CD8 T cell responses were only detected in mice immunized with adenoviruses carrying the Ade2 insert (Figure 4 & 8). In particular, these C-term specific CD8 T cell responses were similar in mice immunized with either lOelOvp of C7 Ade2 or 10e9vp of Ad5 Ade2 (Figure 8). In addition, the anti-CS antibody responses were determined by ELISA on sera collected 48 days post-immunization. In particular, it is the total Ig response against the R32LR polypeptide (i.e. which covers the middle portion of P '. falciparum CSP) that was measured (Mettens et al., Vaccine 2008). The results indicate that a single immunization with C7 Adel or C7 Ade2 induces low levels of R32LR-specific antibody response. The intensity of this response correlates with the number of viral particles used for immunization (dose range effect).
Example 3
Immunogenicity of the C7-Ade2 in CB6F1 mice
CB6F1 mice were immunized once intramuscularly with a dose range (1OeIO, 10e9, 10e8 viral particles) of the C7 chimpadeno virus expressing the Ade2 construct (5 pools of mice/group). Peripheral blood was collected and pooled on days 21, 28 and 35 post-immunization and the CS C-term and CS N-term specific CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest, i.e. the N-terminal region (N-term) or C-terminal region (C-term) of the CS protein. As negative controls, some cells were also cultured overnight in vitro in culture medium (unstimulated). The Ag-specifϊc responses were calculated by subtracting the average cytokine response produced by unstimulated cells from the average cytokine response produced by the peptide-stimulated cells.
The results indicate that in this mouse strain, a single immunization of either 10e9vp or lOelOvp of C7 Ade2 induce C-term and N-term specific CD4 and CD8 T cell responses (Figures 17 to 20). In particular, the intensity of the average observed response is equal if not higher when the 10e9 vp dose is used. The CS-specific CD8 T cell responses are mainly N term-specific while the CS-specific CD4 T cell responses are equally targeting the N-term and C-term region of the CS protein. The cytokine profile of the CS-specific CD4 and CD8 T cell response was also determined and it was similar across all the tested timepoints. The profile displayed on d28 post-immunization is shown in Figures 21 to 24 and is representative of the other tested timepoints. Briefly, the CS-specific CD8 T cell responses are mostly composed of CD8 T cells producing IFNg only (Figures 21 & 22). The CS-specific CD4 T cell responses are also composed of IFNg producing CD4 T cells but also and to a lesser extent of CD4 T cells producing IL2 only or both IL2 & IFNg (Figures 23 & 24). Example 4
Immunogenicity of the C7-Ade2 in prime/boost or co-formulation with
RTS,S/AS01B in CB6F1 mice
We have tested the immunogenicity of the C7 chimpadeno virus expressing the Ade2 construct in either prime -boost or co-formulation (combo) with RTS,S/AS01B in CB6F1 mice (4 pools of mice/group). ASOlB is an adjuvant system containing 3D- MPL and QS21 formulated with liposomes. Mice were immunized intramuscularly on d0, 14 and 28 as follows:
Figure imgf000017_0001
Where,
A = 10e9 vp of C7 Ade2
P = 5μg RTS,S/50μl AS01B
C = lOelO vp of C7 Ade2 + 5μg RTS,S/50μl ASOlB
Peripheral blood was collected and pooled on days 21 (7d pll), 35 (7d pill), 49(2 Id pill), 63 (35d pIII),77 (49d pill) post-immunization and the CS C-term, CS N-term and HBs specific CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest (CS N-term, CS C-term or HBs). As negative controls, some cells were also cultured overnight in vitro in culture medium (unstimulated). The Ag-specific responses were calculated by subtracting the average cytokine response produced by unstimulated cells from the average cytokine response produced by the peptide-stimulated cells.
The results indicate that:
- APP, PPA and CCC all induce N-term specific CD8 T cell responses. At the 7d pll timepoint, N-term specific CD8 T cell responses are observed in the CCC group, followed by the APPgroup. At the later timepoints tested, (2 Id pill, 35d pill & 49d pill), these response are & remain of similar intensity in the APP, PPA and CCC groups (Figure 25).
- APP, PPA and CCC induce C-term specific CD8 T cell responses that still persist 49 days post 3rd immunization (Figure 26)
N-term specific CD4 T cell responses are mainly detected in mice immunized with APP or CCC with higher intensities of such responses in the APP group (Figure 27)
- All groups display C-term specific CD4 T cell responses. However, APP and CCC induce similar higher levels of C-term specific CD4 T cell responses and these about 2 to 3 times higher than the ones induced by PPA and PPP at all the timepoints tested (Figure 28).
The HBs-specific CD4 and CD8T cell responses are higher in the animals immunized with PPP than with APP, PPA or CCC (Figures 29 and 30). - The CCC treatment regimen is the only one that is associated with the simultaneous induction of CS and HBs-specific CD4 and CD8 T cell responses.
The cytokine profiles of the CS- and HBs- specific CD4 and CD8 T cell responses were also determined and were similar across the timepoints tested. The ones from the 21 d pill timepoint are shown below as representative of all timepoints tested (Figures 31 to 36). Briefly, the Ag-specifϊc CD8 T cell responses are mostly composed of CD8 T cells producing IFNg (Figures 31 to 33). In contrast, the Ag-specifϊc CD4 T cell responses are composed of a mixture of CD4 T cells producing IFNg, IFNg and IL-2 and to a lesser extent CD4 T cells producing IL-2 (Figures 34 to 36).
In addition, the Ag-specifϊc antibody responses were determined by ELISA on sera collected 14 and 42 days post-3rd immunization. In particular, the total Ig responses against the R32LR polypeptide (i.e. which covers the middle portion of P '. falciparum CSP) and against HBs were measured (Mettens et al., Vaccine 2008). All immunization regimens did elicit R32LR and HBs-specifϊc antibody responses that persisted up to the last timepoint tested, i.e. 42 days post 3rd immunization (Figures 37 & 38).
Example 5
Immunogenicity of co-formulation C7-Ade2+ RTS,S/AS01B in CB6F1 mice
In this experiment, we compared the immunogenicity of the chimpadenovirus C7 Ade2 co-formulated (Combo) with RTS,S/AS01B. In particular, we compared the immune response elicited by 1, 2 or 3 injections of the combo in CB6F1 mice (4 pools of mice/group). In addition, different intervals between the 2 injections of the combo were evaluated (i.e. 14 & 21 days). Finally, a group of mice immunized with A-P-P served as control in the experiment. The experimental design can be summarized as follows:
Figure imgf000018_0001
Where,
A = 10e9 vp of C7 Ade2
P = 5μg RTS,S/50μl AS01B
C = 10e9 vp of C7 Ade2 + 5μg RTS,S/50μl ASOlB
Peripheral blood was collected and pooled on days 35, 42, 49, 63 and 98 and the CS C-term, CS N-term and HBs specific CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest, i.e. the N-terminal region (N-term), the C-terminal region (C-term) of the CS protein or HBs. As negative controls, some cells were also cultured overnight in vitro in culture medium (unstimulated). The Ag-specific responses were calculated by subtracting the average cytokine response produced by unstimulated cells from the average cytokine response produced by the peptide-stimulated cells.
The results indicate that under these experimental conditions, 2 immunizations of the combo were required to simultaneously induce CS C-term, CS N-term and HBs- specific CD4 and CD8 T cell responses. The interval between the 2 immunizations of the combo did not seem to significantly impact the levels of Ag-specific T cell responses detected. There was a trend for higher HBs-specific CD4 and CD8 T cell responses in mice immunized 3 times with the combo. The kinetics of the Ag-specific T cell responses are shown in Figures 39 to 44.
The cytokine profiles of the CS- and HBs- specific CD4 and CD8 T cell responses were also determined and were similar across the timepoints tested. The ones from the day 42 of the study are shown below as representative of all timepoints tested (Figures 42 to 47). Briefly, the Ag-specific CD8 T cell responses are mostly composed of CD8 T cells producing IFNg (Figures 45 to 47). In contrast, the Ag-specific CD4 T cell responses are composed of a mixture of CD4 T cells producing IFNg, IFNg and IL-2 and to a lesser extent CD4 T cells producing IL-2 (Figures 48 to 50).
In addition, the Ag-specific antibody responses were determined by ELISA on sera collected on day 56 and 99 of the study. In particular, the total Ig responses against the R32LR polypeptide (i.e. which covers the middle portion of P. falciparum CSP) and against HBs were measured (Mettens et al., Vaccine 2008). All immunization regimens did elicit R32LR and HBs-specific antibody responses: within each group, these responses were of similar intensity at both timepoints tested. In addition, when the groups immunized with the combo were compared, there was a trend for higher responses in groups immunized with 3 doses of the combo (Figures 51 & 52).
Example 6
Evaluation of the need for each component of the combo, i.e. C7-Ade2, RTS9S and ASOlB to elicit both CS & HBs T cell and Ab responses simultaneously in CB6F1 mice
In this experiment, we evaluated the need for each component of the combo (i.e. C7 Ade2, RTS,S and ASOlB) to elicit CS and HBs-specific CD4 and CD8 T cell responses simultaneously. In this experiment, CB6F1 mice (6 pools of mice/group) were immunized twice intramuscularly (on days 0 & 14) with the combo or components thereof as shown below:
Figure imgf000019_0001
Where: A = 10e9 vp of C7 Ade2 P = 5μg RTS,S/50μl AS01B
C = 10e9 vp of C7 Ade2 + 5μg RTS,S/50μl ASOlB C7 empty = 10e9 vp of C7 empty vector (no insert) RTS,S = 5μg ofRTS,S
ASOlB = GSK Proprietary Adjuvant System IB Buffer = ASOlB buffer (no immunostimulant)
Peripheral blood was collected and pooled on days 14, 28, 70, 91 & 112 and the CS C-term, CS N-term and HBs specific CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest, i.e. the
N-terminal region (N-term), the C-terminal region (C-term) of the CS protein or HBs.
As negative controls, some cells were also cultured overnight in vitro in culture medium (unstimulated). The Ag-specific responses were calculated by subtracting the average cytokine response produced by unstimulated cells from the average cytokine response produced by the peptide-stimulated cells.
In addition, the Ag-specific antibody responses were determined by ELISA on sera collected on day 42 and 84 of the study. In particular, the total Ig responses against the R32LR polypeptide (i.e. which covers the middle portion of P. falciparum CSP) and against HBs were measured (Mettens et al., Vaccine 2008).
The results indicate that each component of the combo is required to simultaneously elicit CS (N-term & C-term) CD4 and CD8 T cell responses (Figures 53 to 58) as well as R32LR and HBs antibody responses (Figures 65 and 66).
The average cytokine profiles of the CS- and HBs- specific CD4 and CD8 T cell responses were also determined at each timepoint of the study and these are shown in Figures 59 to 64. Briefly, the Ag-specific CD8 T cell responses are mostly composed of CD8 T cells producing IFNg (Figures 59 to 61). In contrast, the Ag-specific CD4 T cell responses are composed of a mixture of CD4 T cells producing IFNg, IFNg and IL-2 and to a lesser extent CD4 T cells producing IL-2 (Figures 62 to 64).
Example 7
Immunogenicity of a synthetic C7 Ade2 in CB6F1 mice
A synthetic C7 chimpadenovirus expressing the Ade2 construct was made available and its immunogenicity in mice was compared to the one of the original C7 Ade2. In this experiment, CB6F1 mice (6 pools of mice/group) were immunized with 10e9 vp of the original C7 Ade2 or its synthetic counterpart.
Peripheral blood was collected and pooled on days 21, 28 & 35 post-immunization and the CS C-term and CS N-term CD4 & CD8 T cell responses producing IL-2 and/or IFN-gamma were measured by flow cytometry, after overnight in vitro restimulation with pools of 15mer peptides covering the sequences of interest, i.e. the N-terminal region (N-term), the C-terminal region (C-term) of the CS protein. As negative controls, some cells were also cultured overnight in vitro in culture medium (unstimulated). The Ag-specific responses were calculated by subtracting the average cytokine response produced by unstimulated cells from the average cytokine response produced by the peptide-stimulated cells.
The results indicate that both C7 adenoviruses elicited similar levels of N-term and C- term specific CD4 and CD8 T cell responses. In particular, and regardless of the viral vector used (original or synthetic), the Ag-specific CD8 T cell responses were mainly
N-term specific (Figures 67 and 68). The N-term and C-term specific CD4 T cell responses were of similar intensity regardless of the viral vector used (Figures 69 and
70).
The cytokine profiles of the CS-specific CD4 and CD8 T cell responses were also determined and are shown in Figures 71 to 74. Briefly, the Ag-specific CD8 T cell responses were mostly composed of CD8 T cells producing IFNg (Figures 71 & 72).
In contrast, the Ag-specific CD4 T cell responses were composed of a mixture of CD4
T cells producing IFNg, IFNg and IL-2 and to a lesser extent CD4 T cells producing
IL-2 (Figures 73 to 74).
SEQ ID NO 1
Amino acid sequence
MMRKLAILSVSSFLFVEALFQEYQCYGSSSNTRVLNELNYDNAGTNLYNE 50
LEMNYYGKQENWYSLKKNSRSLGENDDGNNNNGDNGREGKDEDKRDGNNE 100
DNEKLRKPKHKKLKQPADGNPDPNANPNVDPNANPNVDPNANPNVDPNAN 150
PNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNVDPNANPN 200
ANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNAN 250
PNANPNANPNANPNANPNKNNQGNGQGHNMPNDPNRNVDENANANSAVKN 300
NNNEEPSDKHIKEYLNKIQNSLSTEWSPCSVTCGNGIQVRIKPGSANKPK 350
DELDYANDIEKKICKMEKCSSVFNVVNSAIGL 382
SEQ ID NO 2
Nucleotide sequence
ATGATGAGAAAACTTGCCATCCTCAGCGTCAGCTCTTTCCTGTTCGTGGA 50
GGCCCTCTTCCAGGAGTATCAGTGCTACGGAAGCAGCAGCAATACAAGGG 100
TCCTGAACGAGCTCAACTATGACAACGCTGGAACGAACCTGTATAACGAG 150
CTGGAGATGAACTACTATGGCAAGCAGGAGAACTGGTATAGCCTGAAGAA 200
GAACAGCCGGTCCCTGGGCGAGAACGACGACGGCAACAACAACAACGGCG 250
ACAACGGCAGGGAGGGCAAAGATGAGGACAAGAGGGACGGGAACAACGAG 300
GATAACGAGAAGCTGCGGAAGCCCAAGCACAAGAAACTCAAGCAGCCCGC 350
CGACGGGAACCCGGACCCCAATGCAAATCCCAACGTCGACCCAAACGCAA 400
ACCCTAACGTGGACCCCAACGCCAATCCCAACGTCGATCCTAATGCCAAT 450
CCAAATGCCAACCCTAACGCAAATCCTAATGCAAACCCCAACGCCAATCC 500
TAACGCCAACCCAAATGCCAACCCAAACGCTAACCCCAACGCTAACCCAA 550
ATGCAAATCCCAATGCTAACCCAAACGTGGACCCTAACGCTAACCCCAAC 600
GCAAACCCTAACGCCAATCCTAACGCAAACCCCAATGCAAACCCAAACGC 650
AAATCCCAACGCTAACCCTAACGCAAACCCCAACGCCAACCCTAATGCCA 700
ACCCCAATGCTAACCCCAACGCCAATCCAAACGCAAATCCAAACGCCAAC 750
CCAAATGCAAACCCCAACGCTAATCCCAACGCCAACCCAAACGCCAATCC 800
TAACAAGAACAATCAGGGCAACGGGCAGGGCCATAACATGCCGAACGACC 850
CTAATCGGAATGTGGACGAGAACGCCAACGCCAACAGCGCCGTGAAGAAC 900
AACAACAACGAGGAGCCCTCCGACAAGCACATCAAGGAATACCTGAACAA 950
GATCCAGAACAGTCTGAGCACCGAGTGGTCCCCCTGCTCCGTGACCTGCG 1000
GCAACGGCATCCAGGTGAGGATCAAGCCCGGCTCCGCCAACAAGCCCAAG 1050
GACGAGCTGGACTACGCCAACGACATCGAGAAGAAGATCTGCAAGATGGA 1100
GAAATGCAGCTCTGTGTTCAACGTCGTGAACTCCGCCATCGGCCTGTGA 1149 Seq ID NO 3
MMRKLAILSVSSFLFVEALFQEYQCYGSSSNTRVLNELNYDNAGTNLYNE 50
LEMNYYGKQENWYSLKKNSRSLGENDDGNNNNGDNGREGKDEDKRDGNNE 100
DNEKLRKPKHKKLKQPADGNPDPNANPNVDPNANPNVDPNANPNVDPNAN 150
PNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNVDPNANPN 200
ANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNAN 250
PNANPNANPNANKNNQGNGQGHNMPNDPNRNVDENANANSAVKNNNNEEP 300
SDKHIKEYLNKIQNSLSTEWSPCSVTCGNGIQVRIKPGSANKPKDELDYA 350
NDIEKKICKMEKCSSVFNVVNS 372
Seq ID NO 4
Atgatgaggaaactggccatcctgagcgtgagcagcttcctgttcgtgga 50 Ggccctgtttcaggagtaccagtgctacggcagcagcagcaacacccggg 100 Tgctgaacgagctgaactacgacaacgccggcaccaacctgtacaacgag 150 Ctggagatgaactactacggcaagcaggagaactggtacagcctgaagaa 200 Gaacagccggtctctgggcgagaacgacgacggcaacaacaacaacggcg 250 Acaacggccgggagggcaaggacgaggacaagcgggacggcaacaacgag 300 Gacaacgagaagctgcggaagcccaagcacaagaaacttaagcagcccgc 350 Cgacggcaaccccgaccccaacgccaaccccaacgtggaccccaacgcca 400 Atcctaatgtcgaccccaatgccaatccgaacgttgatcccaatgcgaat 450 Cctaacgctaaccccaatgccaacccaaatgccaatccaaatgcaaatcc 500 Caacgccaatccaaacgcaaaccctaatgctaatccaaacgctaatccta 550 Atgccaatcccaatgctaacccaaacgtcgatcctaacgcaaatccgaac 600 Gctaaccccaacgcaaatcccaacgctaacccgaacgcaaaccctaacgc 650 Caatccgaatgccaacccaaacgccaacccgaacgctaatccgaatgcta 700 Acccgaatgctaatcctaacgcaaacccaaatgcaaaccccaatgcaaac 750 Ccgaacgccaatcccaacgccaatcctaatgccaacaagaacaatcaggg 800 Caacggccagggccacaacatgcccaacgaccccaaccggaacgtggacg 850 Agaacgccaacgccaacagcgccgtgaagaacaacaacaacgaggagccc 900 Agcgacaagcacatcaaggagtacctgaacaagatccagaacagcctgag 950 Caccgagtggagcccctgcagcgtgacctgcggcaacggcattcaggtgc 1000 Ggatcaagcccggcagcgccaacaagcccaaggacgagctggactacgcc 1050 Aatgacatcgagaagaagatctgcaagatggagaagtgcagcagcgtgtt 1100 Caacgtggtgaactcctga 1119
Seq ID No. 5
Chimp Adeno 7 (seq ID No 17 from WO 03/046124)
Ala Pro Lys GIy Ala Pro Asn Thr Cys GIn Trp Thr Tyr Lys Ala GIy
1 5 10 15
Asp Thr Asp Thr GIu Lys Thr Tyr Thr Tyr GIy Asn Ala Pro VaI GIn
20 25 30
GIy lie Ser lie Thr Lys Asp GIy lie GIn Leu GIy Thr Asp Ser Asp
35 40 45
GIy GIn Ala lie Tyr Ala Asp GIu Thr Tyr GIn Pro GIu Pro GIn VaI
50 55 60
GIy Asp Ala GIu Trp His Asp lie Thr GIy Thr Asp GIu Lys Tyr GIy 65 70 75 80
GIy Arg Ala Leu Lys Pro Asp Thr Lys Met Lys Pro Cys Tyr GIy Ser 85 90 95 Phe Ala Lys Pro Thr Asn Lys GIu GIy GIy GIn Ala Asn VaI Lys Thr
100 105 110
GIu Thr GIy GIy Thr Lys GIu Tyr Asp lie Asp Met Ala Phe Phe Asp
115 120 125
Asn Arg Ser Ala Ala Ala Ala GIy Leu Ala Pro GIu lie VaI Leu Tyr
130 135 140
Thr GIu Asn VaI Asp Leu GIu Thr Pro Asp Thr His lie VaI Tyr Lys 145 150 155 160
Ala GIy Thr Asp Asp Ser Ser Ser Ser lie Asn Leu GIy GIn GIn Ser
165 170 175
Met Pro Asn Arg Pro Asn Tyr lie GIy Phe Arg Asp Asn Phe lie GIy
180 185 190
Leu Met Tyr Tyr Asn Ser Thr GIy Asn Met GIy VaI Leu Ala GIy GIn
195 200 205
Ala Ser GIn Leu Asn Ala VaI VaI Asp Leu GIn Asp Arg Asn Thr GIu
210 215 220
Leu Ser Tyr GIn Leu Leu Leu Asp Ser Leu GIy Asp Arg Thr Arg Tyr 225 230 235 240
Phe Ser Met Trp Asn GIn Ala VaI Asp Ser Tyr Asp Pro Asp VaI Arg
245 250 255 lie lie GIu Asn His GIy VaI GIu Asp GIu Leu Pro Asn Tyr Cys Phe
260 265 270
Pro Leu Asp Ala VaI GIy Arg Thr Asp Thr Tyr GIn GIy lie Lys Ala
275 280 285
Asn GIy Asp Asn GIn Thr Thr Trp Thr Lys Asp Asp Thr VaI Asn Asp
290 295 300
Ala Asn GIu Leu GIy Lys GIy Asn Pro Phe 305 310
Seq ID No. 6
Chimp Adeno 7 (seq ID No 20 from WO 03/046124)
Thr Leu Trp Thr Thr Ala Asp Pro Ser Pro Asn Cys Lys lie Tyr Ser
1 5 10 15
GIu Lys Asp Ala Lys Leu Thr Leu Cys Leu Thr Lys Cys GIy Ser GIn
20 25 30 lie Leu GIy Thr VaI Thr VaI Leu Ala VaI Asn Asn GIy Ser Leu Asn
35 40 45
Pro lie Thr Asn Thr VaI Ser Thr Ala Leu VaI Ser Leu Lys Phe Asp
50 55 60
Ala Ser GIy VaI Leu Leu Ser Ser Ser Thr Leu Asp Lys GIu Tyr Trp 65 70 75 80
Asn Phe Arg Lys GIy Asp VaI Thr Pro Ala GIu Pro Tyr Thr Asn Ala
85 90 95 lie GIy Phe Met Pro Asn lie Lys Ala Tyr Pro Lys Asn Thr Ser Ala
100 105 110
Ala Ser Lys Ser His lie VaI Ser GIn VaI Tyr Leu Asn GIy Asp GIu
115 120 125
Ala Lys Pro Leu Met Leu lie lie Thr Phe Asn GIu Thr GIu Asp Ala
130 135 140
Thr Cys Thr Tyr Ser lie Thr Phe GIn Trp Lys Trp Asp Ser Thr Lys 145 150 155 160
Tyr Thr GIy GIu Thr Leu Ala Thr Ser Ser Phe Thr Phe Ser Tyr lie
165 170 175
Ala GIn GIu
NNGDNGREGKDEDKRDGNN [Seq ID No: 7]
AIGL [Seq ID No: 8] PNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNVDPN ANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNANPNAN PNANPNANPNANPNANPNANPN
[Seq ID No: 9]
NANP NVDP NANP NVDP NANP NVDP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NVDP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP NANP
[Seq ID No: 10]
TCC ATG ACG TTC CTG ACG TT (CpG 1826) [Seq ID No: 11]
TCT CCC AGC GTG CGC CAT (CpG 1758) [Seq ID No: 12]
ACC GAT GAC GTC GCC GGT GAC GGC ACC ACG [Seq ID No: 13]
TCG TCG TTT TGT CGT TTT GTC GTT (CpG 2006) [Seq ID No: 14]
TCC ATG ACG TTC CTG ATG CT (CpG 1668) [Seq ID No: 15]
TCG ACG TTT TCG GCG CGC GCC G (CpG 5456), [Seq ID No: 16]
Seq ID No: 17
Sequence of a expression cassette for a protein of Seq ID No. 3. The protein encoding region is underlined.
TAACTATAACGGTCCTAAGGTAGCGAAAGCTCAGATCGGCTGACCGCCCAACGACCC CCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGGGACTTT CCATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCCCACTTGGCAGTACATCA AGTGTATCATATGCCAAGTCCGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGC CTGGCATTATGCCCAGTACATGACCTTACGGGACTTTCCTACTTGGCAGTACATCTA CGTATTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGTACACCAATGGGCG TGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCATTGACGTCAATGG GAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCAAAATGTCGTAATAACCCCGC CCCGTTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGAGGTCTATATAAGCAGAGC TCGTTTAGTGAACCGTCAGATCGCCTGGAGACGCCATCCACGCTGTTTTGACCTCCA TAGAAGACACCGGGACCGATCCAGCCTCCGCGGCCGGGAACGGTGCATTGGAACGCG GATTCCCCGTGCCAAGAGTGCGGCCAGCTTTATTGCGGTAGTTTATCACAGTTAAAT TGCTAACGCAGTCAGTGCTTCTGACACAACAGTCTCGAACTTAAGCTGCAGAAGTTG GTCGTGAGGCACTGGGCAGGTAAGTATCAAGGTTACAAGACAGGTTTAAGGAGACCA ATAGAAACTGGGCTTGTCGAGACAGAGAAGACTCTTGCGTTTCTGATAGGCACCTAT TGGTCTTACTGACATCCACTTTGCCTTTCTCTCCACAGGTGTCCACTCCCAGTTCAA TTACAGCTCTTAAGGCTAGAGTGGCCGCACCATGATGAGGAAACTGGCCATCCTGAG CGTGAGCAGCTTCCTGTTCGTGGAGGCCCTGTTTCAGGAGTACCAGTGCTACGGCAG CAGCAGCAACACCCGGGTGCTGAACGAGCTGAACTACGACAACGCCGGCACCAACCT GTACAACGAGCTGGAGATGAACTACTACGGCAAGCAGGAGAACTGGTACAGCCTGAA GAAGAACAGCCGGTCTCTGGGCGAGAACGACGACGGCAACAACAACAACGGCGACAA CGGCCGGGAGGGCAAGGACGAGGACAAGCGGGACGGCAACAACGAGGACAACGAGAA GCTGCGGAAGCCCAAGCACAAGAAACTTAAGCAGCCCGCCGACGGCAACCCCGACCC CAACGCCAACCCCAACGTGGACCCCAACGCCAATCCTAATGTCGACCCCAATGCCAA TCCGAACGTTGATCCCAATGCGAATCCTAACGCTAACCCCAATGCCAACCCAAATGC CAATCCAAATGCAAATCCCAACGCCAATCCAAACGCAAACCCTAATGCTAATCCAAA CGCTAATCCTAATGCCAATCCCAATGCTAACCCAAACGTCGATCCTAACGCAAATCC GAACGCTAACCCCAACGCAAATCCCAACGCTAACCCGAACGCAAACCCTAACGCCAA TCCGAATGCCAACCCAAACGCCAACCCGAACGCTAATCCGAATGCTAACCCGAATGC TAATCCTAACGCAAACCCAAATGCAAACCCCAATGCAAACCCGAACGCCAATCCCAA CGCCAATCCTAATGCCAACAAGAACAATCAGGGCAACGGCCAGGGCCACAACATGCC CAACGACCCCAACCGGAACGTGGACGAGAACGCCAACGCCAACAGCGCCGTGAAGAA CAACAACAACGAGGAGCCCAGCGACAAGCACATCAAGGAGTACCTGAACAAGATCCA GAACAGCCTGAGCACCGAGTGGAGCCCCTGCAGCGTGACCTGCGGCAACGGCATTCA GGTGCGGATCAAGCCCGGCAGCGCCAACAAGCCCAAGGACGAGCTGGACTACGCCAA TGACATCGAGAAGAAGATCTGCAAGATGGAGAAGTGCAGCAGCGTGTTCAACGTGGT GAACTCCTGAGGATCCGATCTTTTTCCCTCTGCCAAAAATTATGGGGACATCATGAA GCCCCTTGAGCATCTGACTTCTGGCTAATAAAGGAAATTTATTTTCATTGCAATAGT GTGTTGGAATTTTTTGTGTCTCTCACTCGGAAGCGATCTGAATTCATCTATGTCGGG TGCGGAGAAAGAGGTAATGAAATGGCA
Seq ID No: 18
Sequence of Ade2 expression cassette is below. The protein encoding region is underlined.
TAACTATAACGGTCCTAAGGTAGCGAAAGCTCAGATCGGCTGACCGCCCAACGACCC CCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGGGACTTT CCATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCCCACTTGGCAGTACATCA AGTGTATCATATGCCAAGTCCGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGC CTGGCATTATGCCCAGTACATGACCTTACGGGACTTTCCTACTTGGCAGTACATCTA CGTATTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGTACACCAATGGGCG TGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCATTGACGTCAATGG GAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCAAAATGTCGTAATAACCCCGC CCCGTTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGAGGTCTATATAAGCAGAGC TCGTTTAGTGAACCGTCAGATCGCCTGGAGACGCCATCCACGCTGTTTTGACCTCCA TAGAAGACACCGGGACCGATCCAGCCTCCGCGGCCGGGAACGGTGCATTGGAACGCG GATTCCCCGTGCCAAGAGTGCGGCCAGCTTTATTGCGGTAGTTTATCACAGTTAAAT TGCTAACGCAGTCAGTGCTTCTGACACAACAGTCTCGAACTTAAGCTGCAGAAGTTG GTCGTGAGGCACTGGGCAGGTAAGTATCAAGGTTACAAGACAGGTTTAAGGAGACCA ATAGAAACTGGGCTTGTCGAGACAGAGAAGACTCTTGCGTTTCTGATAGGCACCTAT TGGTCTTACTGACATCCACTTTGCCTTTCTCTCCACAGGTGTCCACTCCCAGTTCAA TTACAGCTCTTAAGGCTAGAGTGGCCGCACCATGATGAGAAAACTTGCCATCCTCAG CGTCAGCTCTTTCCTGTTCGTGGAGGCCCTCTTCCAGGAGTATCAGTGCTACGGAAG CAGCAGCAATACAAGGGTCCTGAACGAGCTCAACTATGACAACGCTGGAACGAACCT GTATAACGAGCTGGAGATGAACTACTATGGCAAGCAGGAGAACTGGTATAGCCTGAA GAAGAACAGCCGGTCCCTGGGCGAGAACGACGACGGCAACAACAACAACGGCGACAA CGGCAGGGAGGGCAAAGATGAGGACAAGAGGGACGGGAACAACGAGGATAACGAGAA GCTGCGGAAGCCCAAGCACAAGAAACTCAAGCAGCCCGCCGACGGGAACCCGGACCC CAATGCAAATCCCAACGTCGACCCAAACGCAAACCCTAACGTGGACCCCAACGCCAA TCCCAACGTCGATCCTAATGCCAATCCAAATGCCAACCCTAACGCAAATCCTAATGC AAACCCCAACGCCAATCCTAACGCCAACCCAAATGCCAACCCAAACGCTAACCCCAA CGCTAACCCAAATGCAAATCCCAATGCTAACCCAAACGTGGACCCTAACGCTAACCC CAACGCAAACCCTAACGCCAATCCTAACGCAAACCCCAATGCAAACCCAAACGCAAA TCCCAACGCTAACCCTAACGCAAACCCCAACGCCAACCCTAATGCCAACCCCAATGC TAACCCCAACGCCAATCCAAACGCAAATCCAAACGCCAACCCAAATGCAAACCCCAA CGCTAATCCCAACGCCAACCCAAACGCCAATCCTAACAAGAACAATCAGGGCAACGG GCAGGGCCATAACATGCCGAACGACCCTAACCGGAATGTGGACGAGAACGCCAACGC CAACAGCGCCGTGAAGAACAACAACAACGAGGAGCCCTCCGACAAGCACATCAAGGA ATACCTGAACAAGATCCAGAACAGTCTGAGCACCGAGTGGTCCCCCTGCTCCGTGAC CTGCGGCAACGGCATCCAGGTGAGGATCAAGCCCGGCTCCGCCAACAAGCCCAAGGA CGAGCTGGACTACGCCAACGACATCGAGAAGAAGATCTGCAAGATGGAGAAATGCAG CTCTGTGTTCAACGTCGTGAACTCCGCCATCGGCCTGTGAGGATCCGATCTTTTTCC CTCTGCCAAAAATTATGGGGACATCATGAAGCCCCTTGAGCATCTGACTTCTGGCTA ATAAAGGAAATTTATTTTCATTGCAATAGTGTGTTGGAATTTTTTGTGTCTCTCACT CGGAAGCGATCTGAATTCATCTATGTCGGGTGCGGAGAAAGAGGTAATGAAATGGCA T

Claims

Claims
1. A replication deficient simian adenoviral vector C7 encoding a protein comprising CS protein from P. falciparum or a fragment thereof.
2. A viral vector according to claim 1, wherein the protein encoded comprises the sequence of Seq ID No: 7.
3. A viral vector according to claim 1 or claim 2, where the protein encoded comprises the sequence of Seq ID No: 8.
4. A viral vector according to any one of claims 1 to 3, wherein the protein encoded has the sequence of Seq ID No: 1.
5. A viral vector according to claim 1 or claim 2, wherein the protein encoded has the sequence of Seq ID No: 3.
6. A composition comprising a viral vector as defined in any one of claims 1 to 5 and an excipient.
7. A vaccine composition for the treatment or prophylaxis of malaria comprising a viral vector as defined in any one of claims 1 to 5 and an adjuvant.
8. A vaccine as defined in claim 7, which further comprises a protein.
9. A vaccine as defined in claim 8, wherein the protein is RTS, S.
10. A vaccine composition comprising (1) a replication deficient simian adenoviral vector C7 encoding a protein comprising CS protein from P. falciparum or a fragment thereof (2) a malaria antigen, and (3) an adjuvant.
11. A kit comprising (1) a replication deficient simian adenoviral vector C7 encoding a protein comprising CS protein from P. falciparum or a fragment thereof (2) a malaria antigen, and (3) an adjuvant.
12. A vaccine or kit as claimed in claim 10 or claim 11, wherein the protein encoded by the adenoviral vector C7 has the sequence of Seq ID No: 1.
13. A vaccine or kit as claimed in any of claims 7 to 12, wherein the malaria antigen is RT S, S.
14. A vaccine or kit according to any of claims 7 to 13, wherein the adjuvant comprises 3D-MPL.
15. A vaccine or kit according to any of claims 7 to 13, wherein the adjuvant comprises a saponin.
16. A vaccine or kit according to claim 15, wherein the saponin is QS21.
17. A vaccine or kit according to any of claims 7 to 13, wherein the adjuvant comprises 3D-MPL and QS21.
18. A vaccine or kit as defined in any one of claims 7 to 17, wherein the formulation is provided as an oil-in-water emulsion.
19. A vaccine as defined in any one of claims 7 to 10 or 12 to 17, wherein the vaccine is a liposomal formulation.
20. A vaccine composition as claimed in claim 10 comprising (1) a replication deficient simian adenoviral vector C7 encoding a protein comprising CS protein from P. falciparum having the sequence of Seq ID No: 1 (2) malaria antigen RTS, S, and (3) an adjuvant comprising 3D-MPL and QS21.
21. A process for the preparation of a viral vector as defined in any one of claims 1 to 5 comprising the steps of: a. propagating the vector on a suitable cell line, and b. recovering the vector.
22. A process for the preparation of a composition of claim 6 or a vaccine as defined in any one of claims 7 to 10 or 12 to 20 comprising the step of admixing the viral vector of claims 1 to 5 with at least one excipient/carrier.
23. A viral vector as defined in any one of claims 1 to 5, for the treatment or prophylaxis of malaria.
24. A viral vector as claimed in any of claims 1 to 5 for priming or boosting in a prime boost regime for the treatment or prophylaxis of malaria.
25. Use of a viral vector as defined in any one of claims 1 to 5 in the manufacture of a medicament for the treatment or prophylaxis of malaria.
26. A method of treatment comprising administering a therapeutically effective amount of: a. a viral vector as defined in any one of claims 1 to 5, b. a composition as defined in claim 6, or c. a vaccine as defined in any one of claims 7 to 10 or 12 to 20.
PCT/EP2008/066762 2007-12-06 2008-12-04 Vaccine WO2009071613A2 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
EP08857977A EP2227550A2 (en) 2007-12-06 2008-12-04 Vaccine
JP2010536451A JP2011505796A (en) 2007-12-06 2008-12-04 vaccine
AU2008333208A AU2008333208A1 (en) 2007-12-06 2008-12-04 Vaccine
CN2008801267101A CN101939438A (en) 2007-12-06 2008-12-04 Vaccine
BRPI0819889-6A BRPI0819889A2 (en) 2007-12-06 2008-12-04 Viral vector, composition, vaccine, vaccine composition, kit, processes for the preparation of a viral vector and for the preparation of a composition, use of a viral vector, and method of treatment
MX2010006207A MX2010006207A (en) 2007-12-06 2008-12-04 Vaccine.
AP2010005295A AP2010005295A0 (en) 2008-12-04 2008-12-04 Vaccine
CA2707245A CA2707245A1 (en) 2007-12-06 2008-12-04 Vaccine
IL205953A IL205953A0 (en) 2007-12-06 2010-05-25 Vaccine
ZA2010/03851A ZA201003851B (en) 2007-12-06 2010-05-28 Vaccine
MA32991A MA32003B1 (en) 2007-12-06 2010-07-05 Vaccine

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US99280207P 2007-12-06 2007-12-06
US60/992,802 2007-12-06

Publications (2)

Publication Number Publication Date
WO2009071613A2 true WO2009071613A2 (en) 2009-06-11
WO2009071613A3 WO2009071613A3 (en) 2009-08-13

Family

ID=40591828

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/066762 WO2009071613A2 (en) 2007-12-06 2008-12-04 Vaccine

Country Status (20)

Country Link
EP (1) EP2227550A2 (en)
JP (1) JP2011505796A (en)
KR (1) KR20100108544A (en)
CN (1) CN101939438A (en)
AR (1) AR069568A1 (en)
AU (1) AU2008333208A1 (en)
BR (1) BRPI0819889A2 (en)
CA (1) CA2707245A1 (en)
CL (1) CL2008003614A1 (en)
CO (1) CO6300795A2 (en)
CR (1) CR11537A (en)
DO (1) DOP2010000164A (en)
IL (1) IL205953A0 (en)
MA (1) MA32003B1 (en)
MX (1) MX2010006207A (en)
PE (1) PE20091106A1 (en)
TW (1) TW200938633A (en)
UY (1) UY31510A1 (en)
WO (1) WO2009071613A2 (en)
ZA (1) ZA201003851B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010023260A1 (en) * 2008-09-01 2010-03-04 Glaxosmithkline Biologicals S.A. Vaccine compositions
WO2012042279A3 (en) * 2010-09-30 2012-07-26 Isis Innovation Limited Viral vector immunogenic compositions
WO2012141984A1 (en) * 2011-04-08 2012-10-18 Immune Design Corp. Immunogenic compositions and methods of using the compositions for inducing humoral and cellular immune responses

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110687289B (en) * 2019-10-17 2023-04-18 中国人民解放军陆军军医大学 Application of FGL2 protein as malaria infection marker

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040136963A1 (en) * 2001-06-22 2004-07-15 The Trustees Of The University Of Pennsylvania Simian adenovirus vectors and methods of use
WO2005063805A1 (en) * 2003-12-23 2005-07-14 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibodies against the amino terminus region of circumsporozoite protein prevent the onset of malaria infection
WO2005071093A2 (en) * 2004-01-23 2005-08-04 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Chimpanzee adenovirus vaccine carriers
WO2006040334A1 (en) * 2004-10-14 2006-04-20 Crucell Holland B.V. Malaria prime/boost vaccines
US20060188527A1 (en) * 2002-10-23 2006-08-24 Hoffman Stephen L Methods for vaccinating against malaria
WO2007003384A1 (en) * 2005-06-30 2007-01-11 Glaxosmithkline Biologicals Sa Anti-malaria vaccine
US20070071726A1 (en) * 2002-12-17 2007-03-29 Crucell Holland B.V. Recombinant viral-based malaria vaccines
WO2007110409A1 (en) * 2006-03-27 2007-10-04 Crucell Holland B.V. Compositions comprising a recombinant adenovirus and an adjuvant
WO2008009650A2 (en) * 2006-07-18 2008-01-24 Glaxosmithkline Biologicals S.A. Vaccines for malaria
WO2008107370A1 (en) * 2007-03-02 2008-09-12 Glaxosmithkline Biologicals S.A. Novel method and compositions
WO2008122811A2 (en) * 2007-04-10 2008-10-16 Isis Innovation Ltd Adenoviral vectors encoding a pathogen or tumour antigen

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040136963A1 (en) * 2001-06-22 2004-07-15 The Trustees Of The University Of Pennsylvania Simian adenovirus vectors and methods of use
US20060188527A1 (en) * 2002-10-23 2006-08-24 Hoffman Stephen L Methods for vaccinating against malaria
US20070071726A1 (en) * 2002-12-17 2007-03-29 Crucell Holland B.V. Recombinant viral-based malaria vaccines
WO2005063805A1 (en) * 2003-12-23 2005-07-14 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibodies against the amino terminus region of circumsporozoite protein prevent the onset of malaria infection
WO2005071093A2 (en) * 2004-01-23 2005-08-04 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Chimpanzee adenovirus vaccine carriers
WO2006040334A1 (en) * 2004-10-14 2006-04-20 Crucell Holland B.V. Malaria prime/boost vaccines
WO2007003384A1 (en) * 2005-06-30 2007-01-11 Glaxosmithkline Biologicals Sa Anti-malaria vaccine
WO2007110409A1 (en) * 2006-03-27 2007-10-04 Crucell Holland B.V. Compositions comprising a recombinant adenovirus and an adjuvant
WO2008009650A2 (en) * 2006-07-18 2008-01-24 Glaxosmithkline Biologicals S.A. Vaccines for malaria
WO2008107370A1 (en) * 2007-03-02 2008-09-12 Glaxosmithkline Biologicals S.A. Novel method and compositions
WO2008122811A2 (en) * 2007-04-10 2008-10-16 Isis Innovation Ltd Adenoviral vectors encoding a pathogen or tumour antigen

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
DAME J B ET AL: "STRUCTURE OF THE GENE ENCODING THE IMMUNODOMINANT SURFACE ANTIGEN ON THE SPOROZOITE OF THE HUMAN MALARIA PARASITE PLASMODIUM FALCIPARUM" SCIENCE, AMERICAN ASSOCIATION FOR THE ADVANCEMENT OF SCIENCE, US, WASHINGTON, DC, vol. 225, 1 January 1984 (1984-01-01), pages 593-599, XP000907559 ISSN: 0036-8075 *
SCHNEIDER J ET AL: "ENHANCED IMMUNOGENICITY FOR CD8+ T CELL INDUCTION AND COMPLETE PROTECTIVE EFFICACY OF MALARIA DNA VACCINATION BY BOOSTING WITH MODIEFIED VACCINIA VIRUS ANKARA" NATURE MEDICINE, NATURE PUBLISHING GROUP, NEW YORK, NY, US, vol. 4, no. 4, 1 April 1998 (1998-04-01), pages 397-402, XP000739989 ISSN: 1078-8956 *
See also references of EP2227550A2 *
SHOTT J P ET AL: "Adenovirus 5 and 35 vectors expressing Plasmodium falciparum circumsporozoite surface protein elicit potent antigen-specific cellular IFN-gamma and antibody responses in mice" VACCINE, BUTTERWORTH SCIENTIFIC. GUILDFORD, GB, vol. 26, no. 23, 2 June 2008 (2008-06-02), pages 2818-2823, XP022666955 ISSN: 0264-410X [retrieved on 2008-04-16] *
STEWART V ANN ET AL: "Priming with an adenovirus 35-circumsporozoite protein (CS) vaccine followed by RTS,S/AS01B boosting significantly improves immunogenicity to Plasmodium falciparum CS compared to that with either malaria vaccine alone." INFECTION AND IMMUNITY MAY 2007, vol. 75, no. 5, May 2007 (2007-05), pages 2283-2290, XP002527574 ISSN: 0019-9567 *
TATSIS N ET AL: "Chimpanzee-origin adenovirus vectors as vaccine carriers." GENE THERAPY MAR 2006, vol. 13, no. 5, March 2006 (2006-03), pages 421-429, XP002527576 ISSN: 0969-7128 *
ZHI YAN ET AL: "Efficacy of severe acute respiratory syndrome vaccine based on a nonhuman primate adenovirus in the presence of immunity against human adenovirus." HUMAN GENE THERAPY MAY 2006, vol. 17, no. 5, May 2006 (2006-05), pages 500-506, XP002527575 ISSN: 1043-0342 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010023260A1 (en) * 2008-09-01 2010-03-04 Glaxosmithkline Biologicals S.A. Vaccine compositions
JP2012508160A (en) * 2008-09-01 2012-04-05 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム Vaccine composition
WO2012042279A3 (en) * 2010-09-30 2012-07-26 Isis Innovation Limited Viral vector immunogenic compositions
WO2012141984A1 (en) * 2011-04-08 2012-10-18 Immune Design Corp. Immunogenic compositions and methods of using the compositions for inducing humoral and cellular immune responses
US9044420B2 (en) 2011-04-08 2015-06-02 Immune Design Corp. Immunogenic compositions and methods of using the compositions for inducing humoral and cellular immune responses
AU2012243039B2 (en) * 2011-04-08 2017-07-13 Immune Design Corp. Immunogenic compositions and methods of using the compositions for inducing humoral and cellular immune responses
EA027236B1 (en) * 2011-04-08 2017-07-31 Иммьюн Дизайн Корп. Immunogenic compositions and methods of using the compositions for inducing humoral and cellular immune responses
EP3632463A1 (en) * 2011-04-08 2020-04-08 Immune Design Corp. Immunogenic compositions and methods of using the compositions for inducing humoral and cellular immune responses

Also Published As

Publication number Publication date
CR11537A (en) 2010-08-18
UY31510A1 (en) 2009-08-03
CO6300795A2 (en) 2011-07-21
PE20091106A1 (en) 2009-08-24
MX2010006207A (en) 2010-10-04
BRPI0819889A2 (en) 2015-06-16
MA32003B1 (en) 2011-01-03
CA2707245A1 (en) 2009-06-11
CN101939438A (en) 2011-01-05
JP2011505796A (en) 2011-03-03
EP2227550A2 (en) 2010-09-15
CL2008003614A1 (en) 2010-01-15
ZA201003851B (en) 2012-11-28
IL205953A0 (en) 2010-11-30
AR069568A1 (en) 2010-02-03
KR20100108544A (en) 2010-10-07
DOP2010000164A (en) 2010-07-31
TW200938633A (en) 2009-09-16
WO2009071613A3 (en) 2009-08-13
AU2008333208A1 (en) 2009-06-11

Similar Documents

Publication Publication Date Title
US20100272786A1 (en) Vaccine
EP2040743B1 (en) Vaccines for malaria
US20100272745A1 (en) Vaccines for malaria
WO2009071613A2 (en) Vaccine
US8999347B2 (en) Vaccines for malaria
CN101553249A (en) Vaccines for malaria

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880126710.1

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08857977

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 205953

Country of ref document: IL

Ref document number: 1897/KOLNP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2707245

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 585894

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2010536451

Country of ref document: JP

Ref document number: 12010501294

Country of ref document: PH

Ref document number: MX/A/2010/006207

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 10070274

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 2008333208

Country of ref document: AU

Date of ref document: 20081204

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2008857977

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 201011537

Country of ref document: CR

Ref document number: CR2010-011537

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: DZP2010000406

Country of ref document: DZ

ENP Entry into the national phase

Ref document number: 20107014859

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PI 2010002585

Country of ref document: MY

ENP Entry into the national phase

Ref document number: PI0819889

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20100604