WO2007062656A2 - A nucleotide vaccine - Google Patents

A nucleotide vaccine Download PDF

Info

Publication number
WO2007062656A2
WO2007062656A2 PCT/DK2006/000675 DK2006000675W WO2007062656A2 WO 2007062656 A2 WO2007062656 A2 WO 2007062656A2 DK 2006000675 W DK2006000675 W DK 2006000675W WO 2007062656 A2 WO2007062656 A2 WO 2007062656A2
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
protein
vaccine
nucleic acid
antigenic
Prior art date
Application number
PCT/DK2006/000675
Other languages
French (fr)
Other versions
WO2007062656A3 (en
Inventor
Peter Johannes Holst
Allan Randrup Thomsen
Jan Pravsgaard Christensen
Original Assignee
Copenhagen University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Copenhagen University filed Critical Copenhagen University
Priority to PL06818135T priority Critical patent/PL1957528T3/en
Priority to DK06818135.3T priority patent/DK1957528T3/en
Priority to US12/095,730 priority patent/US20100278904A1/en
Priority to EP06818135A priority patent/EP1957528B1/en
Priority to CN200680045006.4A priority patent/CN101321781B/en
Priority to SI200631493T priority patent/SI1957528T1/en
Priority to ES06818135T priority patent/ES2396437T3/en
Publication of WO2007062656A2 publication Critical patent/WO2007062656A2/en
Publication of WO2007062656A3 publication Critical patent/WO2007062656A3/en
Priority to US14/817,631 priority patent/US20150368672A1/en
Priority to US17/166,419 priority patent/US20210230633A1/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • 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
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/605MHC molecules or ligands thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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
    • C12N2710/10034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • C12N2710/10043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/10011Arenaviridae
    • C12N2760/10022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/10011Arenaviridae
    • C12N2760/10034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/20011Rhabdoviridae
    • C12N2760/20211Vesiculovirus, e.g. vesicular stomatitis Indiana virus
    • C12N2760/20234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a technology and method whereby a faster, broader and stronger immune response is obtained when using viral and DNA-based vaccines.
  • Vaccines can be prophylactic: they are given before the actual infection occurs, or therapeutic: where they elicit or accelerate an immune response to a pathogen already in the body. Both methods of vaccination require the establishment of a solid immune response.
  • the immune response that is activated by infection or vaccination depends on the interaction of several cell types, such as T-, B- and antigen presenting cells as well as several different molecules, primarily antigens, MHC molecules, T- and B-cells receptors and many more.
  • Antigens are peptide fragments presented on the surface of antigen presenting cells by MHC molecules. Antigens can be of foreign, i.e. pathogenic origin, or stem from the organism itself, so called self or auto antigens.
  • the MHC molecules are representatives of a polymorphous gene family encoded by a specific chromosomal region known as the "major histocompatibility complex", hence MHC.
  • MHC-I MHC class I
  • MHC-II MHC class Il
  • T-helper cells are stimulated by antigens presented by MHC class Il (MHC-II) molecules residing on the surface of antigen presenting cells.
  • the MHC-II molecules are synthesized in the endoplasmatic reticulum.
  • invariant chain (Ii) in a manner preventing the MHC-II molecules from being loaded with self- or auto-antigens.
  • the MHC-II molecule is by signal sequences in the invariant chain transported to the cell surface in a specific cellular compartment. As the compartment matures by the processing of its contents it progresses from being a lysosome, to a late endosome (after fusion with endocytotic vesicles) to an MHC class Il compartment (MIIC).
  • the endocytotic vesicle contains foreign antigen i.e. proteolytically cleaved bacterial peptide fragments. These fragments are by their degradation prepared to be loaded onto the MHC-II molecule.
  • the MHC-II molecule is released by the invariant chain in a two part process when the invariant chain first is degraded proteolytically leaving only a peptide termed CLIP in the MHC-II binding domain, secondly by the removal of CLIP by an HLA-DM molecule.
  • the MHC-II molecule is then free to bind the foreign antigens and present these on the cell surface after fusion of the MIIC vesicle to the plasma membrane. This initiates the humoral immune response as the presented antigen stimulates activation of a T- helper cell which in turn by several means activates a B cell, which ultimately differentiates into an antibody secreting cell.
  • the cellular immune response is initiated when the T-cell receptor of T-cytotoxic cells recognizes antigen bound to the MHC class I molecule on an antigen presenting cell.
  • MHC-I molecules are not associated with a molecule of a functionality like the invariant chain that associates with MHC-II.
  • the processing of MHC-I into an antigen presenting molecule furthermore differs from that of MHC-II molecules in that the MHC-I molecule is loaded with antigen already in the endoplasmatic reticulum.
  • the antigens presented by the MHC-I molecule are typically peptide fragments cleaved by the proteasome of proteins that have been synthesized by the antigen presenting cell itself. These proteins may be abnormal proteins encoded in the cells own DNA or proteins derived from viruses or other pathogens that have infected the cell and parasitize its protein synthesis machinery.
  • the MHC class l-related proteolytic system is present in virtually all cells.
  • Cytotoxic T cells eradicate intracellular pathogens and tumors by direct lysis of cells and by secreting cytokines such as ⁇ -interferon.
  • the predominant cytotoxic T cell is the CD8 + T cell, which also is antigen specific.
  • Helper T cells also can lyse cells, but their primary function is to secrete cytokines that promote the activities of B cells (antibody-producing cells) and other T cells and thus they broadly enhance the immune response to foreign antigens, including antibody-mediated and cytotoxic T cell-mediated response mechanisms.
  • CD4 + T cells are the major helper T cell phenotype in the immune response.
  • DNA vaccines are new and promising candidates for the development of both prophylactic and therapeutic vaccines.
  • the strength of the ensuing immune response is determined through a combination of the potency of the vector (i.e. naked DNA, viral vectors, live attenuated viruses etc.), the expression level of the antigen, and the recombinant antigen it self (i.e. high or low affinity MHC binders, structural determinants selecting for more or less limited T-or B-cell repertoire etc.). It is generally held to be true, that efficient induction of immunological memory requires or benefits from the interactions of CD4 + (helper cell) T-cells with CD8 + (cytotoxic) T-cells and B-cells that mediate many of the effects of immune memory.
  • CD4 + (helper cell) T-cells with CD8 + (cytotoxic) T-cells and B-cells that mediate many of the effects of immune memory.
  • CD4 + helper cell
  • CD8 + cytotoxic
  • the present invention has solved the problem of stimulating the immune response in a manner that increases the kinetics of the response, simultaneously with both broadening and improving the response, while avoiding, among other things, the above mentioned disadvantages of the vaccination methods described in the state of the art.
  • a novel system for a directed, specific and fast stimulation of the immune system is hereby made available in order to improve the vaccination of all animals.
  • nucleic acid construct comprising sequences encoding at least one invariant chain operatively linked to at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide.
  • an adenoviral vector comprising a nucleotide construct encoding at least one antigen and at least one protein or peptide or fragment of a protein or peptide which stimulates an immune response.
  • an aspect of the present invention to provide means of stimulating an MHC-I mediated immune response by an adenoviral vector comprising a nucleotide construct encoding at least one antigen and at least one protein or peptide or fragment of a protein or peptide.
  • Another aspect includes stimulating an MHC-II response by an adenoviral vector comprising a nucleotide construct encoding at least one antigen and at least one protein or peptide or fragment of a protein or peptide.
  • a further aspect provides means of stimulating intercellular spreading of the nucleic , acid construct, the adenoviral vector, the proteins encoded within any of these or any parts of any of these.
  • a vaccine comprising the nucleic acid construct or the adenoviral vector as detailed herein.
  • a vaccine where at least one invariant chain is operatively linked to at least one protein or peptide or fragment of a protein or peptide which stimulates an MHC-I response. This may be done by operatively linking at least one invariant chain with at least one at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide.
  • a vaccine comprising the chimeric protein encoded within the nucleic acid construct or the adenoviral vector as detailed herein.
  • kit in parts comprising either a vaccine composition comprising an adenoviral vector or a nucleic acid construct as described herein together with a medical instrument or other means of administering said vaccine and furthermore instructions on how to use the kit in parts.
  • the present invention provides means for inducing an immune response in an animal, by administering to the animal a vaccine comprising the nucleic acid construct or the adenoviral vector as detailed herein below.
  • Figure 1 Schematic drawing of inserts in the adenovirus vector.
  • Figure 2 CD8 + and CD4 + T-cell responses to adenovirus encoded epitopes.
  • FIG. 3 CD8 + and CD4 + T-cell responses to adenovirus encoded epitopes in F 1 hybrid mice.
  • Figure 4 Ad-IiGP exerts CD8 + T-cell stimulatory effects that are independent of
  • CD4 + T-cells Figure 5 Ad-IiGP confers rapid and superior protection against lethal LCMV infection.
  • FIG. 7 Ad-IiGP confers superior protection to lethal LCMV variants with mutations in immunodominant epitopes.
  • Figure 8 Frequencies of CD8 + or CD4 + T cells reacting to specific LCMV epitopes after Ad-IiGP vaccination and challenge with LCMV variants with mutations in immunodominant epitopes.
  • FIG. 9 CD8 + and CD4 + T cell responses to vaccination with naked DNA-IiGP and DNA-GP
  • Figure 10 Prophylactic vaccination with Ad-Ii-GP increases tumor rejection
  • Figure 11 Therapeutic vaccination with Ad-Ii-GP increases average life span in tumor carrying mice.
  • Figure 12 Survival rate following vaccination with either Ad-Ii-VSVGP or Ad- VSVGP.
  • Figure 13 CD8 + and CD4 + T-cell responses to more adenovirus encoded epitopes
  • Figure 14 Efficiency of Ad-Ii-GP constructs compared to Ad-GP-Lamp-1 constructs measured by CD8 + T-cell responses to adenovirus encoded epitopes.
  • Figure 15 Vector based on in-frame polylinkers.
  • FIG. 16 Vectors with IRES2 sites
  • Adenovirus A group of double-stranded DNA containing viruses. Adenoviruses can be genetically modified making them replication incompetent or conditionally replication incompetent. In this form, as adenoviral constructs or adenovectors, they can be used as gene delivery vehicles for vaccination or gene therapy.
  • Adjuvant Any substance whose admixture with an administered immunogenic determinant / antigen / nucleic acid construct increases or otherwise modifies the immune response to said determinant.
  • Amino acid Any synthetic or naturally occurring amino carboxylic acid, including any amino acid occurring in peptides and polypeptides including proteins and enzymes synthesized in vivo thus including modifications of the amino acids.
  • the term amino acid is herein used synonymously with the term "amino acid residue" which is meant to encompass amino acids as stated which have been reacted with at least one other species, such as 2, for example 3, such as more than 3 other species.
  • the generic term amino acid comprises both natural and non-natural amino acids any of which may be in the "D" or "L” isomeric form.
  • Antibody Immunoglobulin molecules and active portions of immunoglobulin molecules. Antibodies are for example intact immunoglobulin molecules or fragments thereof retaining the immunologic activity.
  • Antigen Any substance that can bind to a clonally distributed immune receptor (T- cell or B-cell receptor). Usually a peptide, polypeptide or a multimeric polypeptide.
  • Antigens are preferably capable of eliciting an immune response.
  • Boost To boost by a booster shot or dose is to give an additional dose of an immunizing agent, such as a vaccine, given at a time after the initial dose to sustain the immune response elicited by the previous dose of the same agent.
  • Carrier Entity or compound to which antigens are coupled to aid in the induction of an immune response.
  • Chimeric protein A genetically engineered protein that is encoded by a nucleotide sequence made by a splicing together of two or more complete or partial genes or a series of (non)random nucleic acids.
  • Complement A complex series of blood proteins whose action "complements" the work of antibodies. Complement destroys bacteria, produces inflammation, and regulates immune reactions.
  • Cytokine Growth or differentiation modulator, used non-determinative herein, and should not limit the interpretation of the present invention and claims.
  • cytokines adhesion or accessory molecules, or any combination thereof, may be employed alone or in combination with the cytokines.
  • CTL Cytotoxic T lymphocytes. A sub group of T-cells expressing CD8 along with the T-cell receptor and therefore able to respond to antigens presented by class I molecules.
  • Delivery vehicle An entity whereby a nucleotide sequence or polypeptide or both can be transported from at least one media to another.
  • Fragment is used to indicate a non-full length part of a nucleic acid or polypeptide.
  • a fragment is itself also a nucleic acid or polypeptide, respectively.
  • Invariant chain an integral membrane protein glycoprotein that associates with and stabilizes MHC Il molecules in the endoplasmatic reticulum and subsequent cellular compartments.
  • invariant chain covers all naturally occurring or artificially generated full length or fragmented homologous genes and proteins of a certain similarity to human invariant chain. Invariant chain is herein abbreviated Ii.
  • Isolated used in connection with nucleic acids, polypeptides, and antibodies disclosed herein 'isolated' refers to these having been identified and separated and/or recovered from a component of their natural, typically cellular, environment.
  • Nucleic acids, polypeptides, and antibodies of the invention are preferably isolated, and vaccines and other compositions of the invention preferably comprise isolated nucleic acids, polypeptides or isolated antibodies.
  • MHC Major histocompatibility complex, two main subclasses of MHC, Class I and Class Il exist.
  • Nucleic acid A chain or sequence of nucleotides that convey genetic information.
  • the nucleic acid is a deoxyribonucleic acid (DNA).
  • Nucleic acid construct A genetically engineered nucleic acid. Typically comprising several elements such as genes or fragments of same, promoters, enhancers, terminators, polyA tails, linkers, polylinkers, operative linkers, multiple cloning sites (MCS), markers, STOP codons, other regulatory elements, internal ribosomal entry sites (IRES) or others.
  • Operative linker A sequence of nucleotides or amino acid residues that bind together two parts of a nucleic acid construct or (chimeric) polypeptide in a manner securing the biological processing of the nucleic acid or polypeptide.
  • Pathogen a specific causative agent of disease, especially a biological agent such as a virus, bacteria, prion or parasite that can cause disease to its host, also referred to as an infective agent.
  • Peptide Plurality of covalently linked amino acid residues defining a sequence and linked by amide bonds.
  • the term is used analogously with oligopeptide and polypeptide.
  • the natural and/or non-natural amino acids may be linked by peptide bonds or by non-peptide bonds.
  • the term peptide also embraces post-translational modifications introduced by chemical or enzyme-catalyzed reactions, as are known in the art.
  • the term can refer to a variant or fragment of a polypeptide.
  • Pharmaceutical carriers also termed excipients, or stabilizers are non-toxic to the cell or individual being exposed thereto at the dosages and concentrations employed. Often the physiologically acceptable carrier is an aqueous pH buffered solution.
  • physiologically acceptable carriers include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEEN.TM., polyethylene glycol (PEG), and PLURONICS.TM.
  • buffers such as phosphate, citrate, and other organic acids
  • antioxidants including ascorbic acid
  • proteins such as serum
  • Promoter A binding site in a DNA chain at which RNA polymerase binds to initiate transcription of messenger RNA by one or more nearby structural genes.
  • Signal peptide A short sequence of amino acids that determine the eventual location of a protein in the cell, also referred to as sorting peptide.
  • siRNA Small interfering RNAs (siRNAs), which target (in a sequence-specific manner) endogenous RNAs for degradation, thereby reducing the amount of a gene product.
  • Surfactant A surface active agent capable of reducing the surface tension of a liquid in which it is dissolved.
  • a surfactant is a compound containing a polar group which is hydrophilic and a non polar group which is hydrophobic and often composed of a fatty chain.
  • Vaccine A substance or composition capable of inducing an immune response in an animal. Also referred to as an immunogenic composition in the present text.
  • An immune response being an immune response (humoral/antibody and/or cellular) inducing memory in an organism, resulting in the infectious agent, being met by a secondary rather than a primary response, thus reducing its impact on the host organism.
  • a vaccine of the present invention may be given as or prophylactic and/or therapeutic medicament.
  • the composition may comprise one or more of the following: antigen(s), nucleic acid constructs comprising one or more antigens operatively linked to Ii, carriers, adjuvants and pharmaceutical carriers..
  • a 'variant' of a given reference nucleic acid or polypeptide refers to a nucleic acid or polypeptide that displays a certain degree of sequence homology/identity to said reference nucleic acid or polypeptide but is not identical to said reference nucleic acid or polypeptide.
  • the present invention relates to a vaccine comprising a nucleic acid construct such as a DNA construct especially a nucleic acid construct comprising sequences encoding invariant chain operatively linked to antigenic protein or peptide encoding sequences.
  • the vaccine stimulates an immune response, especially an immune response in an MHC-I dependent, but CD4 + T-cell independent manner.
  • An aspect of the present invention relates to nucleic acid constructs comprising sequences encoding at least one invariant chain operatively linked to at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide, in short an antigen.
  • nucleic acid construct is understood a genetically engineered nucleic acid.
  • the nucleic acid construct may be a non-replicating and linear nucleic acid, a circular expression vector, an autonomously replicating plasmid or viral expression vector.
  • a nucleic acid construct may comprise several elements such as, but not limited to genes or fragments of same, promoters, enhancers, terminators, poly-A tails, linkers, polylinkers, operative linkers, multiple cloning sites (MCS), markers, STOP codons, internal ribosomal entry sites (IRES) and host homologous sequences for integration or other defined elements.
  • nucleic acid constructs examples are given in figures 1 , 15 and 16, as well as in the sequences identified as SEQ ID NO: 5, 6, 7, 8, 9, and 10.
  • the partial vector sequences of SEQ ID NO: 5, 6, 7, 8, 9, and 10 have been generated by subcloning various elements as described in the above and as illustrated in figures 1, 15 and 16. These partial sequences are all inserted into the pAC-CMVpLpARS(+) vector (Becker et al., 1994, Methods Cell Biol. 43 Pt A:161-189), see GenBank accession number AY590429.1.
  • invariant chain (Ii) or CD74, is a non-polymorphic type Il integral membrane protein, see SEQ ID NOs: 2 and 4 for the amino acid sequences of human and mouse Ii, respectively, and likewise SEQ ID NOs: 1 and 3 for the nucleic acid sequences of human and mouse Ii, respectively.
  • Invariant chain has multiple functions in lymphocyte maturation and in adaptive immune responses, in particular targeting to lysosomal compartments were the Ii CLIP sequence can occupy MHC class Il molecules until these are fused with endosomal compartments (Pieters J. 1997, Curr. Opin. Immunol., 9:8996).
  • Ii has been shown to function as an MHC class I chaperone (Morris et al, 2004, Immunol. Res. 30:171-179) and by its endosomal targeting sequence, to facilitate stimulation of CD4 + , but not CD8 + T-cells directed against covalently linked antigen (Diebold et al., 2001 , Gene Ther. 8:487- 493).
  • the invariant chain protein comprises several domains: a cytosolic domain which includes a signal or sorting peptide (also known as the lysosomal targeting sequence), a transmembrane domain, and a luminal domain which in itself comprises a CLIP region, KEY region, core domain and trimerization domain. Both of these domains are flanked by highly flexible regions (Strumptner-Cuvelette & Benaroch, 2002, Biochem. Biophys. Acta., 1542:1-13). Invariant chain has been characterized in several organisms, including vertebrates (e.g. chicken), mammals (e.g. cow, dog, mouse and rat) and human.
  • vertebrates e.g. chicken
  • mammals e.g. cow, dog, mouse and rat
  • the present invention relates to nucleic acid constructs comprising sequences wherein at least one invariant chain is organism specific or can be related to a specific organism.
  • at least one invariant chain is of vertebrate origin, more preferably of mammalian origin and most preferably of human origin.
  • sequence defined by SEQ ID NO: 1 is the nucleic acid sequence of the invariant chain from human.
  • the employed invariant chain is preferably the invariant chain of the organism that is to receive the vaccination. It is an object of the present invention that the invariant chain and the host organisms or receivers of the treatment are of the same species.
  • the present invention also relates to a nucleic acid construct wherein the encoded at least one invariant chain is a fragment of the sequence identified in SEQ ID NO: 2 of at least 40 amino acids and of at least 85% identity to the same fragment of SEQ ID NO: 2.
  • the fragment is a fragment of at least 40 amino acids from any part of the invariant chain as set forth in SEQ ID NO: 2. This includes a fragment including residues 1 to 40, 10 to 50, 20 to 60, 25 to 65, 30 to 70, 35 to 75, 40 to 80, 45 to 85, 50 to 90, 55 to 95, 60 to 100, 65 to 105, 70 to 110, 75 to 115, 80 to 120, 85 to 125, 90 to 130, 95 to 135, 100 to 140, 105 to 145, 110 to 150, 115 to 155, 120 to 160, 125 to 165, 130 to 170, 135 to 175, 140 to 180, 145 to 185, 150 to 190, 155 to 195, 160 to 200,165 to 205, 170 to 210 and 175 to 216.
  • fragments as any of the above listed expanding up to 5 residues to either side hereof. It further includes fragment of at least 50 residues, of at least 60 residues, of at least 70 residues, of at least 80 residues, of at least 90 residues, of at least 100 residues, of at least 110 residues, of at least 120 residues, of at least 130 residues, of at least 140 residues, of at least 150 residues, of at least 160 residues, of at least 170 residues, of at least 180 residues of at least 190 residues, of at least 200 residues and of at least 210 residues.
  • any of the above described fragments of at least 85 % sequence identity, for example at least 90 % sequence identity, for example at least 91% sequence identity, such as at least 92 % sequence identity, for example at least 93 % sequence identity, such as at least 94 % sequence identity, for example at least 95 % sequence identity, such as at least 96 % sequence identity, for example at least 97% sequence identity, such as at least 98 % sequence identity, for example 99% sequence identity with SEQ ID NO: 2 are included within the scope of the present invention.
  • the identity/homology between amino acid sequences may be calculated using well known scoring matrices such as any one of BLOSUM 30, BLOSUM 40, BLOSUM 45, BLOSUM 50, BLOSUM 55, BLOSUM 60, BLOSUM 62, BLOSUM 65, BLOSUM 70, BLOSUM 75, BLOSUM 80, BLOSUM 85, and BLOSUM 90.
  • scoring matrices such as any one of BLOSUM 30, BLOSUM 40, BLOSUM 45, BLOSUM 50, BLOSUM 55, BLOSUM 60, BLOSUM 62, BLOSUM 65, BLOSUM 70, BLOSUM 75, BLOSUM 80, BLOSUM 85, and BLOSUM 90.
  • the present invention is a nucleic acid construct wherein the encoded at least one invariant chain is a fragment of the SEQ ID NO: 2 of at least 186 amino acids. This includes any of the fragments as defined above, and which thus share identity with the sequence of the invariant chain of SEQ ID NO: 2.
  • the present invention furthermore relates to a nucleic acid construct wherein the encoded at least one invariant chain is at least 85% identical to SEQ ID NO: 2.
  • the present invention relates to a nucleic acid construct wherein the encoded at least one invariant chain is identical to SEQ ID NO: 2.
  • a variant of invariant chain from any organism may be a variant according to the above, i.e. that the variant may be a fragment of the invariant chain of an organism and/or be at least 85% identical to said invariant chain either over all the sequence of the invariant chain or within the fragment of same.
  • the invariant chain may also be from a related species of organism or be from a distantly related species.
  • Another aspect of the present invention relates to the addition, removal or substitution of regions, peptides or domains of the at least one invariant chain as encoded by the nucleic acid construct.
  • the removal of one or more of these regions, peptides or domains will truncate the resulting invariant chain.
  • the addition or replacement of a region, peptide or domain includes the options of choosing these sequences from known sources such as naturally occurring proteins or polypeptides or from artificially synthesized polypeptides or nucleic acids encoding the same.
  • the addition of regions, domains or peptides includes the option of adding one, two or more of each type or of different types of regions, domains, peptides and one, two, three or more of the nucleic acids encoding these regions, domains and peptides. These may be identical or differ from one another based on the sequence.
  • the regions, peptides and domains need not arise from the same organism as the scaffold invariant chain. It is well known in the art to perform additions, deletions and substitutions of individual as well as stretches of nucleotides which will encode the resulting polypeptide.
  • Aligning nucleic acid and especially protein sequences of homologous genes or proteins from different organisms can be of great assistance when determining which substitutions, deletions, rearrangements or other alterations it would be beneficial to construct.
  • Aligning human and murine invariant chain sequences as illustrated below gives an indication of which amino acid residues may be of importance for the structure and function of the invariant chain in these organisms - these are the residues which are conserved between the two sequences. Likewise, the presumably less important residues are the ones in which the sequences differ. It is of interest in regard to the present invention to perform substitutions and/or deletions of the variant residues / regions. When attempting to mutate or delete or otherwise alter the sequence of e.g.
  • the human invariant chain in order to improve its immune response stimulating capacity, it may also be relevant to examine the conserved residues and make e.g. homologous substitutions (i.e. substitutions where the amino acids are considered to be of e.g. same structural quality, polarity, hydrophobicity or other).
  • homologous substitutions i.e. substitutions where the amino acids are considered to be of e.g. same structural quality, polarity, hydrophobicity or other).
  • a preferred embodiment of the present invention relates to the at least one invariant chain wherein the signal peptide is removed, replaced or added onto the sequence encoding the invariant chain.
  • a signal peptide is a short sequence of amino acids that determine the eventual location of a protein in the cell, also referred to as a sorting peptide.
  • Signal peptides that determine the location of proteins to subcellular compartments such as the endoplasmatic reticulum, golgi apparatus and the various compartments comprising the golgi apparatus, the nucleus, the plasma membrane, mitochondria and the various spaces and membranes herein, peroxisomes, lysosomes, endosomes and secretory vesicles among others are all included within the scope of the present invention.
  • a preferred embodiment comprises alone the lysosomal targeting sequence of invariant chain.
  • Another preferred embodiment comprises alone the KEY region of invariant chain.
  • Another preferred embodiment of the present invention relates to the removal, addition, or replacement of the CLIP region of the at least one invariant chain.
  • the addition or replacement of the CLIP region includes the options of adding or replacing the existing CLIP region in the variant of the invariant chain or chains chosen, with CLIP regions from invariant chains of the same or other organisms or of variants of CLIP regions form the same or other organisms.
  • the variant CLIP regions may, as follows from the above, be specifically generated mutant versions of the CLIP region, generated by single or multiple nucleic acid substitutions, deletions or additions.
  • a preferred embodiment comprises the CLIP region alone, or the CLIP region together with the N-terminally adjacent sequence or the C-terminally adjacent sequence without any other regions or domains of invariant chain.
  • Other preferred embodiments comprise alone the N-terminally or C- terminally adjacent sequences to the CLIP region but without the CLIP region itself.
  • adjacent is meant any amino acids within 10 residues of the CLIP region, within 20 residues, within 30 residues, within 40 residues, within 50 residues, within 75 residues or within 100 residues of the CLIP region.
  • An embodiment of the present invention relates to fragments of invariant chain as described above without the CLIP region. These fragments may be at least 5 amino acid residues long, at least 10 residues, at least 15 residues, at least 20 residues, at least 25 residues, at least 30 residues or at least 35 residues in length.
  • Another embodiment relates to fragments of invariant chain wherein the signal peptide is removed and the invariant chain fragment is at least 10 amino acid residues long, at least 15 residues, at least 20 residues, at least 25 residues, at least 30 residues, at least 35 residues, at least 50 residues at least 60 residues, at least 70 residues at least 80 residues, at least 90 residues, at least 100 residues, at least 110 residues at least 120 residues at least 130 residues, at least 140 residues, at least 150 residues, at least 160 residues, at least 170 residues, or at least 180 residues in length.
  • any of the above variants of invariant chain are encompassed in the present invention in the form wherein at least one of said variants is operatively linked to at least one antigen such as an antigenic protein or peptide or an antigenic fragment of said protein or peptide.
  • an object of the present invention to include an antigen originating from any of the following types of pathogens: virus, micro organisms and parasites.
  • At least one antigen may originate from, but is not limited to any of the following families of virus: Adenovirus, arenaviridae, astroviridae, bunyaviridae, caliciviridae, coronaviridae, flaviviridae, herpesviridae, orthomyxoviridae, paramyxoviridae, picomaviridae, poxviridae, reoviridae, retroviridae, rhabdoviridae and togaviridae.
  • Adenovirus arenaviridae, astroviridae, bunyaviridae, caliciviridae, coronaviridae, flaviviridae, herpesviridae, orthomyxoviridae, paramyxoviridae, picomaviridae, poxviridae, reoviridae, retroviridae, rhabdoviridae and togaviridae.
  • At least one antigen or antigenic sequence may be derived from any of the following virus: Influenza A such as H1N1, H1N2, H3N2 and H5N1 (bird flu), Influenza B, Influenza C virus, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus, Hepatitis D virus, Hepatitis E virus, Rotavirus, any virus of the Norwalk virus group, enteric adenoviruses, parvovirus, Dengue fever virus, Monkey pox, Mononegavirales, Lyssavirus such as rabies virus, Lagos bat virus, Mokola virus, Duvenhage virus, European bat virus 1 & 2 and Australian bat virus, Ephemerovirus, Vesiculovirus, Vesicular Stomatitis Virus (VSV), Herpesviruses such as Herpes simplex virus types 1 and 2, varicella zoster, cytomegalovirus, Epstein-Bar virus (EBV), human herpesvirusses (HHV), human herpes
  • the at least one antigenic protein or peptide is from a virus selected from the group of: HIV, Hepatitis C virus, influenza virus, herpes virus, Lassa, Ebola, smallpox, Bird flu, filovirus, Marburg, and papilloma virus.
  • the at least one antigenic protein or peptide is selected from the group of and/or may be at least one antigenic fragment of any of the following: vesicular stomatitis virus glycoprotein (VSV-GP), Influenza A NS-1 (non-structural protein 1 ), Influenza A M1 (matrix protein 1), Influenza A NP (nucleoprotein), LCMV NP, LCMV GP, Ebola GP, Ebola NP, murine gammaherpesvirus M2, M3 and ORF73 (such as MHV-68 M2, M3 and ORF73), chicken Ovalbumin (OVA), or a helper T-cell epitope. It is within the scope of the invention to combine two or more of any of the herein mentioned antigens.
  • VSV-GP vesicular stomatitis virus glycoprotein
  • Influenza A NS-1 non-structural protein 1
  • Influenza A M1 matrix protein 1
  • Influenza A NP nucleoprotein
  • An embodiment of the present invention includes at least one antigenic protein or peptide or fragment of an antigenic protein or peptide from a micro organism. More specifically at least one antigen may be derived from the one of the following from a non-exhaustive list: Anthrax (Bacillus anthracis), Mycobacterium tuberculosis, Salmonella (Salmonella gallinarum, S. pullorum, S. typhi, S. enteridtidis, S. paratyphi, S. dublin, S.
  • Clostridium botulinum Clostridium perfringens, Corynebacterium diphtheriae, Bordetella pertussis, Campylobacter such as Campylobacter jejuni, Crytococcus neoformans, Yersinia pestis, Yersinia enterocolitica, Yersinia pseudotuberculosis, Listeria monocytogenes, Leptospira species, Legionella pneumophila, Borrelia burgdorferi, Streptococcus species such as Streptococcus pneumoniae, Neisseria meningitides, Haemophilus influenzae, Vibrio species such as Vibrio cholerae 01, V.
  • Aeromonas sobria & Aeromonas veronii Plesiomonas shigelloides, Shigella species such as Shigella sonnei, S. boydii, S. flexneri, and S. dysenteriae, Enterovirulent Escherichia coli EEC (Escherichia coli - enterotoxigenic (ETEC), Escherichia coli - enteropathogenic (EPEC), Escherichia coli O157:H7 enterohemorrhagic (EHEC), Escherichia coli - enteroinvasive (EIEC)), Staphylococcus species, such as S.
  • ETEC Escherichia coli - enterotoxigenic
  • EPEC Escherichia coli O157:H7 enterohemorrhagic
  • EIEC Escherichia coli - enteroinvasive
  • Staphylococcus species such as S.
  • VRSA vancomycin intermediate/resistant species
  • MRSA multidrug resistant species
  • Shigella species such as S. flexneri, S. sonnei, S. dysenteriae, Cryptosporidium parvum
  • Brucella species such as B. abortus, B.
  • the at least one antigenic protein or peptide is from a micro-organism selected from the group of: Mycobacterium tuberculosis, Bacillus anthracis, Staphylococcus species, and Vibrio species.
  • An embodiment of the invention relates to a nucleic acid construct, wherein the at least one antigenic protein or peptide encoded is from a parasite.
  • Another embodiment of the present invention relates to a nucleic acid construct comprising combinations of at least two antigenic proteins or peptides from any of the abovementioned pathogens.
  • the antigen is derived from, but not limited to, a parasite selected from the group of: Plasmodium species such as Plasmodium malariae, Plasmodium ovale, Plasmodium vivax, Plasmodium falciparum, Endolimax nana, Giardia lamblia, Entamoeba histolytica, Cryptosporidum parvum, Blastocystis hominis, Trichomonas vaginalis, Toxoplasma gondii, Cyclospora cayetanensis, Cryptosporidium muris, Pneumocystis carinii, Leishmania donovani, Leishmania tropica, Leishmania braziliensis, Leishmania mexicana, Acanthamoeba species such as Acanthamoeba castellanii, and A.
  • Plasmodium species such as Plasmodium malariae, Plasmodium ovale, Plasmodium vivax, Plasmodium fal
  • the at least one antigenic protein or peptide is from a parasite selected from the group of: Plasmodium species, Leishmania species, and Trypanosoma species.
  • An aspect of the present invention relates antigens and/or antigenic sequences derived from diseases or agents that infect domestic animals, especially commercially relevant animals such as pigs, cows, horses, sheep, goats, llamas, rabbits, mink, mice, rats, dogs, cats, poultry such as chicken, turkeys, pheasants and others, fish such as trout, salmon and other farmed species.
  • diseases or agents here of from which at least one antigen or antigenic sequence may be derived include, but are not limited to: Multiple species diseases such as: Anthrax, Aujeszky's disease, Bluetongue, Brucellosis such as: Brucella abortus, Brucella melitensis or Brucella suis; Crimean Congo haemorrhagic fever,
  • Echinococcosis/hydatidosis virus of the family Picornaviridae, genus Aphthovirus causing Foot and Mouth disease especially any of the seven immunologically distinct serotypes: A, O, C, SAT1 , SAT2, SAT3, Asial , or Heartwater, Japanese encephalitis, Leptospirosis, New world screwworm (Cochliomyia hominivorax), Old world screwworm (Chrysomya bezziana), Paratuberculosis, Q fever, Rabies, Rift Valley fever, Rinderpest, Trichinellosis, Tularemia, Vesicular stomatitis or West Nile fever; Cattle diseases such as: Bovine anaplasmosis, Bovine babesiosis, Bovine genital campylobacteriosis, Bovine spongiform encephalopathy, Bovine tuberculosis, Bovine viral diarrhoea, Contagious bovine
  • Equine diseases such as: African horse sickness, Contagious equine metritis, Dourine, Equine encephalomyelitis (Eastern), Equine encephalomyelitis (Western), Equine infectious anaemia, Equine influenza, Equine piroplasmosis, Equine rhinopneumonitis, Equine viral arteritis, Glanders, Surra (Trypanosoma evansi) or Venezuelan equine encephalomyelitis; Swine diseases such as: African swine fever, Classical swine fever, Nipah virus encephalitis, Porcine cysticercosis, Porcine reproductive and respiratory syndrome, Swine vesicular disease or Transmissible gastroenteritis; Avian diseases such as: Avian chlamydiosis, Avian infectious bronchitis, Avian infectious laryngotracheitis, Avian mycoplasmosis
  • avian mycoplasmosis M. synoviae
  • Duck virus hepatitis Fowl cholera
  • Fowl typhoid Highly pathogenic avian influenza this being any Influenzavirus A or B and especially H5N1 , Infectious bursal disease (Gumboro disease), Marek's disease, Newcastle disease, Pullorum disease or Turkey rhinotracheitis; Lagomorph and rodent diseases such as: Virus enteritis,
  • Myxomatosis or Rabbit haemorrhagic disease Fish diseases such as: Epizootic haematopoietic necrosis, Infectious haematopoietic necrosis, Spring viraemia of carp, Viral haemorrhagic septicaemia, Infectious pancreatic necrosis, Infectious salmon anaemia, Epizootic ulcerative syndrome, Bacterial kidney disease (Renibacterium salmoninarum), Gyrodactylosis (Gyrodactylus salaris), Red sea bream iridoviral disease; or other diseases such as Camelpox or Leishmaniosis.
  • Fish diseases such as: Epizootic haematopoietic necrosis, Infectious haematopoietic necrosis, Spring viraemia of carp, Viral haemorrhagic septicaemia, Infectious pancreatic necrosis, Infectious salmon anaemia, Epizootic ulcerative syndrome, Bacterial
  • the at least one antigenic protein or peptide is from Aujeszky's disease, Foot and mouth disease, Vesicular stomatitis virus, Avian influenza or Newcastle disease.
  • the at least one antigenic protein or peptide or fragment of said antigenic protein or peptide being an antigenic peptide or protein with at least 85% identity to any of the above described antigens.
  • the homology or identity between amino acids may be calculated by any of the previously mentioned BLOSUM scoring matrices.
  • An embodiment of the invention relates to a nucleic acid construct, wherein the at least one antigenic protein or peptide or fragment of an antigenic protein or peptide is from a cancer-specific polypeptide or cancer antigen.
  • cancer specific polypeptides tumor-associated antigens or cancer antigens.
  • any antigen that is found to be associated with cancer tumors may be used.
  • subtraction analyses such as various micro array analyses, such as DNA microarray analysis.
  • the gene expression pattern (as seen in the level of RNA or protein encoded by said genes) between healthy and cancerous patients, between groups of cancerous patients or between healthy and cancerous tissue in the same patient is compared.
  • the genes that have approximately equal expression levels are "subtracted” from each other leaving the genes / gene products that differ between the healthy and cancerous tissue.
  • the at least one antigen of the present invention is derived from, but not limited to, a cancer specific polypeptide selected from the group of: MAGE-3, MAGE-1 , gplOO, gp75, TRP-2, tyrosinase, MART-1 , CEA, Ras, p53, B-Catenin, gp43, GAGE-1 , BAGE-1 , PSA, MUC-1 , 2, 3, and HSP-70, TRP-1 , gp100/pmel17, .beta.
  • a cancer specific polypeptide selected from the group of: MAGE-3, MAGE-1 , gplOO, gp75, TRP-2, tyrosinase, MART-1 , CEA, Ras, p53, B-Catenin, gp43, GAGE-1 , BAGE-1 , PSA, MUC-1 , 2, 3, and HSP-70, TRP-1 , gp100/pmel17, .bet
  • -HCG Ras mutants, p53 mutants, HMW melanoma antigen, MUC-18, HOJ-1 , cyclin-dependent kinase 4 (Cdk4), Caspase 8, HER-2/neu, Human papilloma virus HPV type 6, 11 , 16, 18, 31 and 33, Bcr-Abl tyrosine kinase, carcinoembryonic antigen (CEA), telomerase, and SV40 Large T.
  • the at least one antigenic protein or peptide or fragment of an antigenic protein or peptide is from a cancer-specific polypeptide selected from the group of: p53, HER-2/neu, telomerase, and melanoma antigen.
  • An embodiment of the invention relates to a nucleic acid construct, wherein the at least one antigenic protein or peptide or fragment of an antigenic protein or peptide is from a polypeptide associated with an abnormal physiological response.
  • an abnormal physiological response includes, but is not limited to autoimmune diseases, allergic reactions, cancers and congenital diseases.
  • a non-exhaustive list of examples of hereof includes diseases such rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, psoriasis and Crohn's disease.
  • An aspect of the present invention relates to the nucleic acid construct wherein the operative link between the invariant chain and the antigenic protein or peptide or fragment of antigenic protein or peptide either is a direct link or a link mediated by a spacer region.
  • operative linker is understood a sequence of nucleotides or amino acid residues that bind together two parts of a nucleic acid construct or chimeric polypeptide in a manner securing the biological processing of the nucleic acid or polypeptide. If the operative linker is a direct link, the two nucleic acids each encoding either an open reading frame or a fragment of an open reading frame are placed immediately adjacent to each other and thereby also in frame.
  • the operative linker is mediated by a spacer region
  • a series of nucleotides are inserted between the nucleotides encoding the at least one invariant chain and the at least one antigenic peptide, respectively. It is within the scope of the present invention having a spacer region wherein the spacer region merely is a series of nucleotides linking the at least two elements of the present invention in a manner retaining the open reading frames, or the spacer region may encode one or more signals or separate elements as defined herein below.
  • the invention comprises an operative linker, wherein the operative linker is a spacer region.
  • the invention comprises a spacer region encoding at least one helper epitope for class Il MHC molecules.
  • An example of a helper epitope is an immunogenic determinant such as Diphtheria toxin. Especially Diphtheria toxin B fragment COOH-terminal region has been shown to be immunogenic in mice.
  • HSP70 in part or in whole, as well as other immunogenic peptides, such as influenza viral or immunogenic sequences or peptides with an anchoring motif to HLA class I and class Il molecules, also may be encoded in the spacer region of the nucleic acid construct.
  • the spacer region of the nucleic acid construct encodes at least one protease cleavage site.
  • Cleavage sites of lysosomal proteases such as cathepsins, aspartate proteases and zinc proteases as well as other intracellular proteases fall within the scope of the present invention.
  • the operative linker of the nucleic acid construct may comprise at least one siRNA or miRNA encoding sequence.
  • siRNAs small interfering RNAs
  • miRNAs miRNAs
  • An siRNA or miRNA encoded within the nucleic acid construct of the present invention may thus be chosen to target an undesirable gene product.
  • the operative linker comprises at least one polylinker or multiple cloning site (MCS).
  • Polylinkers and MCS's are series of nucleotides comprising restriction enzyme recognition sequences, i.e. sites where a restriction enzyme cut the DNA in blunt or staggered manner facilitating the subcloning of other fragments / sequences of DNA into the nucleic acid construct.
  • the recognition sequences of the polylinkers / MCS's are typically unique meaning that they are not found elsewhere on the nucleic acid construct.
  • the operative linker may furthermore comprise one or more stop or termination codons that signal the release of the nascent polypeptide from the ribosome.
  • the operative linker may also comprise at least one IRES (Internal Ribosomal Entry Site) and / or at least one promoter.
  • IRES Internal Ribosomal Entry Site
  • An IRES is a nucleotide sequence that allows for translation initiation in the middle of a messenger RNA (mRNA) sequence as part of the greater process of protein synthesis.
  • a promoter is a DNA sequence that enables a gene to be transcribed. The promoter is recognized by RNA polymerase, which then initiates transcription, see in the below.
  • the promoter may be single or bidirectional.
  • the operative linker spanning the region between the invariant chain and the at least one antigen is an operative linker comprising at least one polylinker, and at least one promoter, and optionally also at least one IRES. These elements may be placed in any order.
  • the STOP codon of the invariant chain has been deleted, and the polylinker has been cloned into the vector in a manner conserving the open reading frame allowing for in frame reading of the at least one antigen that is inserted into the polylinker.
  • a STOP codon may be inserted after the polylinker for translation termination.
  • This embodiemtn may be combined with any of the above helper epitopes, mi/siRNAs or any of the other elements herein described.
  • An embodiment of the present invention relates to the placement of the operative linker in relations to the at least one invariant chain and the at least one antigenic protein or peptide or fragment of said protein or peptide, wherein the at antigenic peptide encoding sequences are placed: within the invariant chain sequence, at the front end of the invariant chain sequence, at the terminal part of the invariant chain sequence. This is done in a manner ensuring the readability of the open reading frame of the construct, so that the antigenic peptide is: preceded, surrounded or rounded off by, at least one operative linker.
  • a preferred embodiment of the present invention further relates to the placement of the operative linker in relations to the at least one invariant chain and the at least one antigenic protein or peptide or fragment of said protein or peptide, wherein the at least one antigenic peptide encoding sequence preferably is placed at the terminal part of the invariant chain and an operative linker is inserted herein between.
  • the terminal part being the first or last residue of the invariant chain or fragment hereof.
  • the nucleic acid construct encodes a plurality of elements.
  • the elements being the at least one invariant chain and the at least one antigenic protein or peptide or fragment of said protein or peptide. It therefore falls within the scope of the present invention to have a plurality of invariant chains each of these being operatively linked to each other and to a plurality of antigenic proteins or peptides or fragments of antigenic proteins or peptides, wherein these also are operatively linked.
  • the elements of the nucleic acid construct must thus be operatively linked to each other.
  • Several series of invariant chains each operatively linked to one antigenic protein or peptide or fragment of said protein or peptide, each of these series being operatively linked to each other are encompassed within the present invention.
  • Advantages and very important aspects of the present invention relate to the fact that any type of immune response e.g. T cell mediated and antibody mediated responses, can be initiated, both with epitopes known to be weak antigens, with polypeptides of unknown antigenic properties, and with multiple epitopes/antigens simultaneously.
  • any type of immune response e.g. T cell mediated and antibody mediated responses
  • a preferred embodiment is a nucleic acid construct encoding at least one invariant chain operatively linked to a plurality of antigenic proteins or peptides or fragment of proteins or peptides, such as two, three, four, five, six, eight, ten, twelve or more antigenic proteins or peptides or fragment of proteins or peptides.
  • the nucleic acid construct may comprise additional elements.
  • IVS internal ribosomal entry sites
  • genes encoding proteins related to antigen presentation such as LAMP, calreticulin and Hsp70
  • genes encoding proteins that are related to intracellular spreading such as VP22, HIV Tat, Cx43 or other connexins and intercellular gap-junction constituents
  • genes encoding natural killer cell (NK-cell) activation molecules such as H60 and cytokines, chicken ovalbumin, or any T-helper cell epitope.
  • the nucleic acid construct comprises at least one gene encoding a protein related to antigen presentation such as LAMP, LIMP, calreticulin or Hsp70.
  • the nucleic acid construct comprises at least one gene encoding a protein related to intracellular spreading such as VP22, Cx43, HlV Tat, other connexins or intercellular gap-junction constituents.
  • promoter will be used here to refer to a group of transcriptional control modules that are clustered around the initiation site for RNA polymerase II.
  • Much of the thinking about how promoters are organized derives from analyses of several viral promoters, including those for the HSV thymidine kinase (tk) and SV40 early transcription units. These studies, augmented by more recent work, have shown that promoters are composed of discrete functional modules, each consisting of approximately 7-20 bp of DNA, and containing one or more recognition sites for transcriptional activator proteins. At least one module in each promoter functions to position the start site for RNA synthesis.
  • TATA box in some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV 40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation.
  • Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well.
  • the spacing between elements is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the tk promoter, the spacing between elements can be increased to 50 bp apart before activity begins to decline.
  • individual elements can function either cooperatively or independently to activate transcription. Any promoter that can direct transcription initiation of the sequences encoded by the nucleic acid construct may be used in the invention.
  • An aspect of the present invention comprises the nucleic acid construct wherein the at least one operatively linked invariant chain and antigenic protein or peptide encoding sequence is preceded by a promoter enabling expression of the construct.
  • the promoter is selected from the group of constitutive promoters, inducible promoters, organism specific promoters, tissue specific promoters and cell type specific promoters.
  • promoters include, but are not limited to: constitutive promoters such as: simian virus 40 (SV40) early promoter, a mouse mammary tumor virus promoter, a human immunodeficiency virus long terminal repeat promoter, a Moloney virus promoter, an avian leukaemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus (RSV) promoter, a human actin promoter, a human myosin promoter, a human haemoglobin promoter, cytomegalovirus (CMV) promoter and a human muscle creatine promoter, inducible promoters such as: a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline promoter (tet-on or tet-off), tissue specific promoters such as: HER-2 promoter and PSA associated promoter and bidirectional promoters
  • an inducible promoter includes the option of providing a "dormant" vaccine that can be activated at will. This may be of use if the vaccination preferably only is induced locally vs. systemically within a body (e.g. in cases involving cancer), or the vaccine is detrimental to the health of the recipient at the time of vaccination.
  • the nucleic acid construct comprises a promoter selected from the group of: CMV promoter, SV40 promoter and RSV promoter.
  • An aspect of the present invention comprises the nucleic acid construct as described in any of the above, comprised within a delivery vehicle.
  • a delivery vehicle is an entity whereby a nucleotide sequence or polypeptide or both can be transported from at least one media to another. Delivery vehicles are generally used for expression of the sequences encoded within the nucleic acid construct and/or for the intracellular delivery of the construct or the polypeptide encoded therein. It is within the scope of the present invention that the delivery vehicle is a vehicle selected from the group of: RNA based vehicles, DNA based vehicles/ vectors, lipid based vehicles, virally based vehicles and cell based vehicles.
  • delivery vehicles include, but are not limited to: biodegradable polymer microspheres, lipid based formulations such as liposome carriers, coating the construct onto colloidal gold particles, lipopolysaccharides, polypeptides, polysaccharides, and pegylation of viral vehicles.
  • a preferred embodiment of the present invention regards delivery of the nucleic acid construct as naked DNA by mechanical or electrical techniques. Especially the coating of the nucleic acid construct upon gold particles is a favoured embodiment.
  • the delivery of the nucleic acid construct upon gold particles is done by ballistic transfer using particle bombardment equipment such as a gene gun.
  • a more preferred embodiment of the present invention comprises a virus as a delivery vehicle, where the virus is selected from the non-exhaustive group of: adenoviruses, retroviruses, Antiviruses, adeno-associated viruses, herpesviruses, vaccinia viruses, foamy viruses, cytomegaloviruses, Semliki forest virus, poxviruses, RNA virus vector and DNA virus vector.
  • viruses are well known in the art.
  • Viral vectors are often made up of two components, a modified viral genome and a coat structure surrounding it, although sometimes viral vectors are introduced in naked form or coated with proteins other than viral proteins. Most current vectors have coat structures similar to a wild-type virus. This structure packages and protects the viral nucleic acid and provides the means to bind and enter target cells.
  • viral vectors are modified from wild-type viral genomes to disable the growth of the virus in a target cell while enabling the virus to grow in a host cell (e.g. such as a packaging or helper cell) used to prepare infectious particles.
  • a host cell e.g. such as a packaging or helper cell
  • Vector nucleic acids generally essential cis-acting viral sequences for replication and packaging in a helper line and expression control sequences for regulating the expression of a polynucleotide being delivered to a target cell.
  • Other viral functions are expressed in trans in specific packaging or helper cell lines as known in the art.
  • Adenovirus Adenovirus
  • the vehicle comprising the nucleic acid construct as described herein is an adenovirus.
  • the adenoviral genome consists of a linear double-stranded DNA molecule of approximately 36 kb carrying more than about thirty genes necessary to complete the viral replication cycle.
  • the early genes are divided into 4 regions (E 1 to E4) that are essential for viral replication with the exception of the E3 region, which is believed to modulate the anti-viral host immune response.
  • the E1 region (EIA and EIB) encodes proteins responsible for the regulation of transcription of the viral genome. Expression of the E2 region genes (E2A and E2B) leads to the synthesis of the polypeptides needed for viral replication.
  • the proteins encoded by the E3 region prevent cytolysis by cytotoxic T cells and tumor necrosis factor.
  • the proteins encoded by the E4 region are involved in DNA replication, late gene expression and splicing and host cell shut off.
  • the late genes generally encode structural proteins contributing to the viral capsid.
  • the adenoviral genome carries at cis-acting 5' and 3' ITRs (Inverted
  • the ITRs harbor origins of DNA replication while the encapsidation region is required for the packaging of adenoviral DNA into infectious particles (see for example US 2004/0157307).
  • the vehicle comprising the nucleic acid construct as described herein is a replication defective adenovirus or a conditionally replication deficient adenovirus.
  • Adenoviral vectors can be engineered to be conditionally replicative (CRAd vectors) in order to replicate selectively in specific cells (e.g., such as proliferative cells).
  • CRAd vectors conditionally replicative
  • an adenoviral vector is replication-defective for the E1 function (e.g., by total or partial deletion or mutagenesis of E1).
  • the adenoviral backbone of the vector may comprise additional modifications (deletions, insertions or mutations in one or more viral genes).
  • thermosensitive mutation localized on the DBP (DNA Binding Protein) encoding gene An example of an E2 modification is illustrated by the thermosensitive mutation localized on the DBP (DNA Binding Protein) encoding gene.
  • the adenoviral sequence may also be deleted of all or part of the E4 region. Additional deletions within the non-essential E3 region may allow the size of the polynucleotide being delivered to be increased. However, it may be advantageous to retain all or part of the E3 sequences coding for polypeptides (e.g., such as gp19k) allowing the virus to escape the immune system or inflammatory reactions.
  • Second generation vectors retaining the ITRs and packaging sequences and comprising substantial genetic modifications to abolish the residual synthesis of the viral antigens also may be used in order to improve long-term expression of the expressed gene in the transduced cells.
  • the nucleic acid construct being introduced into the cell may be inserted in any location of the viral genome, with the exception of the cis-acting sequences (
  • Adenoviruses can be derived from any human or animal source, in particular canine, avian, bovine, murine, ovine, feline, porcine or simian sources or alternatively, may be a hybrid virus. Any serotype can be employed. However, the human adenoviruses are preferred and such viruses are available, for example, from the ATCC (American Type Culture Collection).
  • a preferred embodiment of the present invention comprises an adenovirus such as: Ovine adenovirus, Canine adenovirus type II, Modified vaccinia Ankara (MVA) or MVA-BN.
  • adenovirus such as: Ovine adenovirus, Canine adenovirus type II, Modified vaccinia Ankara (MVA) or MVA-BN.
  • Adenoviral particles or empty adenoviral capsids also can be used to transfer nucleic acid constructs or nucleic acid based delivery vectors by a virus-mediated co-internalization process. This process can be accomplished in the presence of cationic agent(s) such as polycarbenes or lipid vesicles comprising one or more lipid layers.
  • Adenoviral particles may be prepared and propagated according to any conventional technique in the field of the art using a complementation cell line or a helper virus, which supplies in trans the missing viral genes necessary for viral replication.
  • the adenoviral particles can be recovered from the culture supernatant but also from the cells after lysis and optionally further purified according to standard techniques (e.g. chromatography and ultracentrifugation).
  • Cell-type specific targeting may be achieved with vectors derived from adenoviruses having a broad host range by the modification of viral surface proteins.
  • the specificity of infection of adenoviruses is determined by the attachment to cellular receptors present at the surface of permissive cells.
  • the fiber and penton present at the surface of the adenoviral capsid play a critical role in cellular attachment.
  • cell targeting of adenoviruses can be carried out by genetic modification of the viral gene encoding fiber and/or penton, to generate modified fiber and/or penton capable of specific interaction with unique cell surface receptors.
  • An aspect of the present invention relates to an adenoviral vector comprising a nucleotide construct encoding at least one antigen and at least one protein or peptide or fragment of a protein or peptide which stimulates an MHC-I response.
  • a further aspect of the present invention relates to an adenoviral vector, wherein the nucleotide construct encodes at least one protein or peptide or fragment of a protein or peptide which stimulates an MHC-II response.
  • the adenoviral vector comprises sequences, wherein the at least one antigen is operatively linked to the at least one MHC response stimulating protein or peptide or fragment of an MHC response stimulating protein or peptide.
  • the MHC stimulating protein or peptide or fragment of protein or peptide is preferably an MHC associated protein or peptide.
  • Such an MHC associated peptide can be but is not limited being selected from the group of: ER localizing peptide, Golgi localizing peptide, endosomal peptide loading compartment localizing peptide, lysosomal, MIIC, CIIV, melanosomes, secretory granules, Birbeck granules.
  • the adenoviral vector comprises an endosomal peptide loading compartment localizing peptide.
  • an endosomal peptide loading compartment localizing peptide can be, but is not limited to being, selected from the group of: sorting signal peptides, LAMP, LIMP and invariant chain.
  • the adenoviral vector comprises at least one MHC response stimulating protein or peptide or fragment of protein or peptide and said MHC response stimulating protein or peptide or fragment of protein or peptide is invariant chain.
  • the adenoviral vector may furthermore comprise proteins that assist in the spreading of the virus or the construct comprised therein.
  • proteins include connexins, gap-junction related proteins and pore-forming proteins.
  • a preferred embodiment of the present invention comprises an adenoviral vector encoding or otherwise comprising any one or more of the following proteins related to intercellular spreading: VP22, Cx43 and HIV Tat.
  • An aspect of the present invention relates to a cell comprising the nucleic acid construct as defined in any of the above.
  • a recombinant cell can be used a tool for in vitro research, as a delivery vehicle for the nucleic acid construct or as part of a gene therapy regime.
  • the nucleic acid construct and nucleic acid based vectors according to the invention can be introduced into cells by techniques well known in the art and which include microinjection of DNA into the nucleus of a cell, transfection, electroporation, lipofection/liposome fusion and particle bombardment.
  • Suitable cells include autologous and non-autologous cells, and may include xenogenic cells.
  • the nucleic acid construct of the present invention is comprised within an antigen presenting cell (APC).
  • APC antigen presenting cell
  • Any cell that presents antigens on its surface in association with an MHC molecule is considered an antigen presenting cell.
  • Such cells include but are not limited to macrophages, dendritic cells, B cells, hybrid APCs 1 and foster APCs. Methods of making hybrid APCs are well known in the art.
  • the APC is a professional antigen presenting cell and most preferably the APC is an MHC-I and/or MHC-II expressing cell.
  • the APC according to any of the above may be a stem cell obtained from a patient. After introducing the nucleic acid construct of the invention, the stem cell may be reintroduced into the patient in an attempt to treat the patient of a medical condition.
  • the cell isolated from the patient is a stem cell capable of differentiating into an antigen presenting cell.
  • the antigen presenting cell comprising the nucleic acid construct of the present invention does not express any co-stimulatory signals and the antigenic protein or peptide or antigenic fragment of said protein or peptide is an auto-antigen.
  • An object of the present invention is the chimeric protein encoded by the nucleic acid constructs as described herein above, comprising at least one operatively linked invariant chain and at least one antigenic protein or peptide or fragment of said antigenic protein or peptide.
  • chimeric protein is understood a genetically engineered protein that is encoded by a nucleotide sequence made by a splicing together of two or more complete or partial genes or a series of (non)random nucleic acids.
  • An aspect of the present invention relates to an antibody that can recognize the chimeric protein as defined herein above.
  • antibody is understood immunoglobulin molecules and active portions of immunoglobulin molecules.
  • Antibodies are for example intact immunoglobulin molecules or fragments thereof retaining the immunologic activity. Such antibodies can be used for the passive immunization of an animal, or for use in an assay for detecting proteins to which the antibody binds.
  • An aspect of the present invention relates to a vaccine comprising a nucleic acid sequence encoding at least one invariant chain operatively linked to at least one antigenic protein or peptide or fragment of said antigenic protein or peptide.
  • the vaccine may thus comprise a nucleic acid construct as defined in any of the above.
  • the vaccine may furthermore be used as a medicament.
  • the vaccine composition according to the invention can be formulated according to known methods such as by the admixture of one or more pharmaceutically acceptable carriers, also known as excipients or stabilizers with the active agent.
  • pharmaceutically acceptable carriers also known as excipients or stabilizers with the active agent.
  • excipients may be acceptable for administration to any individual / animal, preferably to vertebrates and more preferably to humans as they are non-toxic to the cell or individual being exposed thereto at the dosages and concentrations employed.
  • the physiologically acceptable carrier is an aqueous pH buffered solution. Examples of such excipients, carriers and methods of formulation may be found e.g. in Remington's Pharmaceutical Sciences (Maack Publishing Co, Easton, PA).
  • physiologically acceptable carriers include but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEEN.TM., polyethylene glycol (PEG), and PLURONICS.TM.
  • buffers such as phosphate, citrate, and other organic acids
  • antioxidants including ascorbic acid
  • compositions will according to the invention contain an effective amount of the nucleic acid construct, the nucleic acid construct comprised within a delivery vehicle or the chimeric protein encoded within the nucleic acid construct as described herein.
  • a carrier will be used as a scaffold by coupling the proteins or peptides hereto and thus aiding in the induction of an immune response.
  • the carrier protein may be any conventional carrier including any protein suitable for presenting immunogenic determinants.
  • Suitable carriers are typically large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplets or liposomes), and inactive virus particles. Such carriers are well known to those of ordinary skill in the art. Additionally, these carriers may function as immunostimulating agents ("adjuvants"). Immunisation of the animal may be carried out with adjuvants and/or pharmaceutical carriers.
  • Conventional carrier proteins include, but are not limited to, keyhole limpet hemocyanin, serum proteins such as transferrin, bovine serum albumin, or human serum albumin, an ovalbumin, immunoglobulins, or hormones, such as insulin. The carrier may be present together with an adjuvant or independently here from.
  • vaccine compositions are meant to encompass compositions useful for prophylactic and therapeutic use, including stimulating an immune response in a patient. It is further contemplated that the vaccine composition of the invention does not induce any systemic or local toxicity reactions or any other side effects.
  • the nucleic acid construct of the vaccine is packaged.
  • Packaging means for the nucleic acid construct include means selected from, but not limited to the group of: RNA based or DNA based vectors, lipid based carriers, viral expression vectors, viral delivery vectors, cell based vehicles, coating of colloidal gold particles, biodegradable polymer microspheres. Any of the previously mentioned delivery means may thus be used for packing purposes for use in a vaccine composition.
  • the packaging means of the nucleic acid construct for the vaccine is a viral expression vector selected from, but not limited to the group of: adenovirus, retrovirus, lentivirus, adeno-associated virus, herpes virus, vaccinia virus and DNA virus vector.
  • nucleic acid construct of the vaccine is packaged into an adenoviral vector.
  • nucleic acids construct as described in any of the herein above of the vaccine is packaged in a replication deficient or conditionally replication deficient adenoviral vector.
  • Adenoviral vectors are described in detail in the above.
  • An aspect of the invention relates to a vaccine comprising at least two vectors. This encompasses that any one or two different nucleic acid constructs as described may be packed into at least two vectors, these vectors being of a type as described in any of the above.
  • the invention furthermore relates to a vaccine comprising three, four, five or six vectors. Again, these vectors may differ from one another or not, and may carry identical or different nucleic acid constructs as described herein above.
  • a further aspect of the present invention relates to a vaccine comprising at least one chimeric protein as encoded by any of the nucleic acid constructs described herein.
  • a chimeric protein or polypeptide When a chimeric protein or polypeptide is to be used as an immunogen, it may be produced by expression of any one or more of the DNA constructs described above in a recombinant cell or it may be prepared by chemical synthesis by methods known in the art. As described in the above, such chimeric proteins and / or peptides may be coupled to carriers to increase the immunologic response to the proteins / peptides and may be administered with or without an adjuvant and/or excipient.
  • Adjuvants may be included in the vaccine composition to enhance the specific immune response.
  • an adjuvant that when combined with the antigen(s) / nucleic acid constructs and / or delivery vehicles such as adenoviral vehicles (any of which may also be referred to as immunogenic determinant), results in a vaccine composition capable of inducing a strong specific immunological response.
  • the immunogenic determinant may also be mixed with two or more different adjuvants prior to immunisation.
  • Vaccine compositions are also referred to as immunogenic compositions in the present text.
  • adjuvants have been described and used for the generation of antibodies in laboratory animals, such as mouse, rats and rabbits. In such setting the tolerance of side effect is rather high as the main aim is to obtain a strong antibody response.
  • the components of the vaccine composition, including the adjuvant are well characterized. It is further required that the composition has minimal risk of any adverse reaction, such as granuloma, abscesses or fever.
  • An embodiment of the present invention relates to a vaccine comprising an adjuvant.
  • the vaccine composition is suitable for administration to a mammal, and most preferably to a human subject, therefore the preferred adjuvant is suitable for administration to a mammal and most preferably is suitable for administration to a human subject.
  • the choice of adjuvant may further be selected by its ability to stimulate the type of immune response desired, B-cell or/and T-cell activation and the vaccine composition may be formulated to optimize distribution and presentation to the relevant lymphatic tissues.
  • Adjuvants pertaining to the present invention may be grouped according to their origin, be it mineral, bacterial, plant, synthetic, or host product.
  • the first group under this classification is the mineral adjuvants, such as aluminum compounds.
  • Antigens precipitated with aluminum salts or antigens mixed with or adsorbed to performed aluminum compounds have been used extensively to augment immune responses in animals and humans. Aluminium particles have been demonstrated in regional lymph nodes of rabbits seven days following immunization, and it may be that another significant function is to direct antigen to T cell containing areas in the nodes themselves.
  • Adjuvant potency has been shown to correlate with intimation of the draining lymph nodes.
  • Aluminium hydroxide has also been described as activating the complement pathway. This mechanism may play a role in the local inflammatory response as well as immunoglobulin production and B cell memory. Furthermore, aluminum hydroxide can protect the antigen from rapid catabolism. Primarily because of their excellent record of safety, aluminum compounds are presently the only adjuvants used in humans.
  • Adjuvants with bacterial origins can be purified and synthesized (e.g. muramyl dipeptides, lipid A) and host mediators have been cloned (Interleukin 1 and 2).
  • the last decade has brought significant progress in the chemical purification of several adjuvants of active components of bacterial origin: Bordetella pertussis, Mycobacterium tuberculosis, lipopoly-saccharide, Freund's Complete Adjuvant (FCA) and Freund's Incomplete Adjuvant (Difco Laboratories, Detroit, Mich.) and Merck Adjuvant 65 (Merck and Company, Inc., Rahway, NJ. ).
  • suitable adjuvants in accordance with the present invention are e.g. Titermax Classical adjuvant (SIGMA-ALDRICH), ISCOMS, Quil A, ALUN, see US 58767 and 5,554,372, Lipid A derivatives, choleratoxin derivatives, HSP derivatives, LPS derivatives, synthetic peptide matrixes, GMDP, and other as well as combined with immunostimulants (US 5,876,735).
  • B. pertussis is of interest as an adjuvant in the context of the present invention due to its ability to modulate cell-mediated immunity through action on T- lymphocyte populations.
  • adjuvant active moieties have been identified and synthesized which permit study of structure-function relationships. These are also considered for inclusion in immunogenic compositions according to the present invention.
  • immunogenic compositions include plant products such as saponin, animal products such as chitin and numerous synthetic chemicals.
  • Adjuvants according to the present invention can also been categorized by their proposed mechanisms of action. This type of classification is necessarily somewhat arbitrary because most adjuvants appear to function by more than one mechanism. Adjuvants may act through antigen localization and delivery, or by direct effects on cells making up the immune system, such as macrophages and lymphocytes. Another mechanism by which adjuvants according to the invention enhance the immune response is by creation of an antigen depot. This appears to contribute to the adjuvant activity of aluminum compounds, oil emulsions, liposomes, and synthetic polymers. The adjuvant activity of lipopolysaccharides and muramyl dipeptides appears to be mainly mediated through activation of the macrophage, whereas B. pertussis affects both macrophages and lymphocytes. Further examples of adjuvants that may be useful when incorporated into immunogenic compositions according to the present invention are described in US 5,554,372.
  • Adjuvants useful in both prophylactic and therapeutic vaccines according to the present invention may thus be mineral salts, such as aluminium hydroxide and aluminium or calcium phosphates gels, oil emulsions and surfactant based formulations such as MF59 (microfluidized detergent stabilized oil in water emulsion), QS21 (purified saponin), AS02 (SBAS2, oil-in-water emulsion + monophosphoryl lipid A (MPL) + QS21), Montanide ISA 51 and ISA-720 (stabilized water in oil emulsion), Adjuvant 65 (containing peanut oil, mannide monooleate and aluminum monostearate), RIBI ImmunoChem Research Inc., Hamilton, Utah), particulate adjuvants, such as virosomes (unilamellar liposomal cehicles incorporating influenza haemagglutinin), AS04 (Al salt with MPL), ISCOMS (structured complex of saponins and lipids (such as cholesterol),
  • Phlei cell wall skeleton Phlei cell wall skeleton
  • AGP RC- 529 (synthetic acylated monosaccharide)
  • DC_chol lipoidal immunostimulators able to self organise into liposomes
  • OM-174 lipid A derivative
  • CpG motifs synthetic oligonucleotides containing immunostimulatory CpG motifs
  • modified bacterial toxins LT and CT, with non-toxic adjuvant effects
  • Endogenous human immunomodulators e.g., hGM-CSF or hlL-12 or lmmudaptin (C3d tandem array), inert vehicles such as gold particles.
  • adjuvants comprise: Immunostimulatory oil emulsions (for example, water-in-oil, oil-in-water, water-in-oil-in-water such as e.g. Freund's incomplete adjuvant such as Montainde®, Specol, mineral salts such e.g. as AI(OH) 3 , AIPO 4 , microbial products, Saponins such as Qual A, synthetic products, as well as adjuvant formulations, and immune stimulatory complexes (ISCOMs) and cytokines, heat-inactivated bacteria/components, nanobeads, LPS, LTA.
  • Immunostimulatory oil emulsions for example, water-in-oil, oil-in-water, water-in-oil-in-water such as e.g. Freund's incomplete adjuvant such as Montainde®, Specol, mineral salts such e.g. as AI(OH) 3 , AIPO 4 , microbial products, Saponins
  • Immunogenic compositions according to the invention may also contain diluents such as buffers, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, sucrose or dextrins, chelating agents such as EDTA, glutathione and other stabilizers and excipients.
  • diluents such as buffers, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, sucrose or dextrins, chelating agents such as EDTA, glutathione and other stabilizers and excipients.
  • Neutral buffered saline or saline mixed with non-specific serum albumin are exemplary appropriate diluents.
  • Adjuvants are generally included in the immunogenic compositions in an amount according to the instructions of the manufacturer.
  • Vaccine compositions according to the invention may be administered to an individual in therapeutically effective amounts.
  • the effective amount may vary according to a variety of factors such as the individual's condition, weight, sex and age. Other factors include the mode of administration.
  • compositions may be provided to the individual by a variety of routes such as subcutaneous, topical, oral and intramuscular. Administration of pharmaceutical compositions is accomplished orally or parenterally. Methods of parenteral delivery include topical, intra-arterial (directly to the tissue), intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, or intranasal administration.
  • the present invention also has the objective of providing suitable topical, oral, systemic and parenteral pharmaceutical formulations for use in the methods of prophylaxis and treatment with the vaccine composition.
  • the vaccine compositions can be administered in such oral dosage forms as tablets, capsules (each including timed release and sustained release formulations), pills, powders, granules, elixirs, tinctures, solutions, suspensions, syrups and emulsions, or by injection.
  • they may also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous, topical with or without occlusion, or intramuscular form, all using forms well known to those of ordinary skill in the pharmaceutical arts.
  • An effective but non-toxic amount of the vaccine, comprising any of the herein described compounds can be employed as a prophylactic or therapeutic agent.
  • any and all conventional dosage forms that are known in the art to be appropriate for formulating injectable immunogenic peptide composition are encompassed, such as lyophilized forms and solutions, suspensions or emulsion forms containing, if required, conventional pharmaceutically acceptable carriers, diluents, preservatives, adjuvants, buffer components, etc.
  • Preferred modes of administration of the vaccine composition according to the invention include, but are not limited to systemic administration, such as intravenous or subcutaneous administration, intradermal administration, intramuscular administration, intranasal administration, oral administration, rectal administration, vaginal administration, pulmonary administration and generally any form of mucosal administration. Furthermore, it is within the scope of the present invention that the means for any of the administration forms mentioned in the herein are included in the present invention.
  • a vaccine according to the present invention can be administered once, or any number of times such as two, three, four or five times. Administering the vaccine more than once has the effect of boosting the resulting immune response.
  • the vaccine can further be boosted by administering the vaccine in a form or body part different from the previous administration.
  • the booster shot is either a homologous or a heterologous booster shot.
  • a homologous booster shot is a where the first and subsequent vaccinations comprise the same constructs and more specifically the same delivery vehicle especially the same viral vector.
  • a heterologous booster shot is where identical constructs are comprised within different viral vectors. This is especially of interest when employing adenoviral delivery as the human body raises an immune response against a given adenovirus if it has previously been exposed thereto.
  • a preferred embodiment of the present invention therefore relates to the pegylation of the adenoviral vector providing the option of boosting with the same (homologous) adenoviral vector.
  • An alternative and preferred embodiment relates to the sequential boosting of a vaccine with different adenoviral vectors comprising the same constructs.
  • a preferred administration form of the vaccine according to the present invention is administering the vaccine to the body area, inside or out, most likely to be the receptacle of a given infection.
  • the receptacle of infection is the body area that the infection is received by, e.g. regarding influenza, the receptacle of infection is the lungs.
  • the vaccine of the present invention can be administered to any organism to which it may be beneficial, especially any animal such as a vertebrate animal. It falls within the scope of the present invention that the means and modes of administration of the vaccine are adapted to the recipient.
  • a preferred recipient of the vaccine is a mammal and the mammal is in a more preferred embodiment of the present invention selected from the group of: cows, pigs, horses, sheep, goats, llamas, mice, rats, monkeys, dogs, cats and humans. In the most preferred embodiment the mammal is a human.
  • An embodiment of the present invention includes a vaccine composition further comprising a second active ingredient.
  • the second active ingredient is selected from, but not limited the group of antibiotics, chemotherapeutics, anti-allergenics, cytokines, complement factors and co-stimulatory molecules of the immune system.
  • kits of parts comprising at least one vaccine composition according to any of the above, a means for administering said vaccine and the instruction on how to do so. It is within the scope of the present invention to include multiple dosages of the same vaccine or several different vaccines.
  • the kit of parts further comprises a second active ingredient.
  • the present invention further comprises a method for inducing an immune response in an animal, comprising administering to the animal a vaccine according to any of the above.
  • the immune response is, but is not limited to any of the following types of responses: an MHC-I dependent response, an MHC-I and/ or MHC-II dependent response, a T-cell dependent response, a CD4 T-cell dependent response, a CD4 + T cell independent response, a CD8 + T-cell dependent response and a B cell dependent immune response.
  • Another method falling within the scope of the present invention is the method of providing at least one vaccine according to any of the above and administering said at least one vaccine to a subject at least once for treatment or prophylaxis of an animal.
  • the invention also encompasses the nucleic acid construct according the herein described for the preparation of a composition for the production of a vaccine.
  • Said vaccine can be but is not limited being used for genetic immunization of an animal, or to treat a clinical condition in an individual in need thereof.
  • FIG. 1 Schematic drawing of inserts in the adenovirus vector.
  • CMV Cytomegalovirus promoter
  • Ii Invariant chain
  • LCMV GP 1-498 Glycoprotein from lymfocytic choriomeningitis virus
  • STOP Stop codon
  • PoIyA SV40 polyadenylation signal
  • FIG. 2 CD8 + and CD4 + T-cell responses to adenovirus encoded epitopes.
  • C57BL/6 mice were vaccinated with 2x10 7 infectious unit (IFU) of Ad-GP or Ad-IiGP in the right hind footpad.
  • IFU infectious unit
  • the number of epitope specific CD8 + or CD4 + T cells were determined by intracellular staining for peptide-induced IFN- ⁇ of spleen cells. Bars represent Average (Avg) ⁇ standard deviation (SD) of 3-5 animals.
  • FIG. 3 CD8 + and CD4 + T-cell responses to adenovirus encoded epitopes in F 1 hybrid mice.
  • the number of epitope specific CD8 + or CD4 + T cells were determined by intracellular staining for peptide-induced IFN- ⁇ of spleen cells. Bars represent Avg ⁇ SD of 4-5 animals
  • Figure 4 Ad-IiGP exerts CD8 + T-cell stimulatory effects that are independent of CD4 + T-cells.
  • MHC-II + or C57BL/6 mice were vaccinated with 2x10 7 IFU of Ad-GP or Ad-IiGP in the right hind footpad.
  • the number of epitope specific CD8 + or CD4 + T cells were determined by intracellular staining for peptide-induced IFN- ⁇ of spleen cells. Bars represent Avg ⁇ SD of 4 animals.
  • Figure 5 Ad-IiGP confers rapid and superior protection against lethal LCMV infection.
  • C57BL/6 mice were vaccinated with 2x10 7 IFU of Ad-GP or Ad-IiGP in the right hind footpad.
  • FIG. 7 Dashed line represent detection limit of the assay.
  • Figure 7 Ad-IiGP confers superior protection to lethal LCMV variants with mutations in immunodominant epitopes.
  • C57BL/6 mice were vaccinated with 2x10 7 IFU of Ad- GP, Ad-IiGP or sham infected in the right hind footpad.
  • On day 90 post vaccination animals were challenged with 20 pfu LCMV Arm 53b i.e. carrying mutations in gp33, gp276 or both epitopes. Mortality was recorded for 14 days.
  • gp33 nil and gp276 nil each group consisted of 5 animals, with gp33/gp276 double nil, the groups were 10 animals.
  • Figure 8 Frequencies of CD8 + or CD4 + T cells reacting to specific LCMV epitopes after Ad-IiGP vaccination and challenge with LCMV variants with mutations in immunodominant epitopes.
  • Surviving animals from the experiment depicted in figure 7 were analysed for epitope specific CD8 + or CD4 + T cells by intracellular staining for peptide-induced IFN- ⁇ of spleen cells. Bars represent Avg ⁇ SD of 3-5 animals.
  • FIG. 9 CD8 + and CD4 + T cell responses to vaccination with naked DNA-IiGP and DNA-GP.
  • C57BL/6 mice were vaccinated with DNA coated onto 1.6-nm gold particles in a concentration of 2 ⁇ g DNA/mg gold, and the DNA-gold complex was coated onto plastic tubes such that 0.5 mg gold was delivered to the mouse per shot (1 ⁇ g DNA per shot).
  • Mice were immunized at the abdominal skin using a hand-held gene gun device employing compressed helium (400 psi) as the particle motive force. Mice were inoculated four times with an interval of 1 week and then allowed to rest for 1 week before investigation.
  • the number of epitope specific CD8 + or CD4 + T cells was determined by intracellular staining for peptide-induced IFN- ⁇ of spleen cells. Bars represent Avg ⁇ SD of 4-5 animals.
  • FIG. 10 Prophylactic vaccination with Ad-Ii-GP increases tumor rejection.
  • C57BL/6 mice were vaccinated in the right hind foot-pad with 2 x 10 7 IFU of adenovirus encoding either full-length glycoprotein of LCMV (Ad-GP) or glycoprotein linked to invariant chain (Ad-Ii-GP).
  • Controls were infected with either adenovirus encoding full-length ⁇ -galactosidase or live LCMV (10 3 PFU of LCMV Armstrong 53b). About 3 months later all mice were challenged by subcutaneous injection of 10 6 B16.F10 melanoma cells expressing the LCMV derived GP33 epitope.
  • mice When tumors were palpable in all mice (day 5 after tumor injection), the animals were vaccinated in the right hind foot-pad using 2 x 10 7 IFU of adenovirus encoding either full-length glycoprotein of LCMV (Ad-GP) or full-length glycoprotein of LCMV linked to invariant chain (Ad-Ii-GP); controls were vaccinated with either adenovirus encoding full-length ⁇ -galactosidase or live LCMV (10 3 PFU of LCMV Armstrong 53b). Mice were sacrificed when the tumor exceeded 12 mm in length or ulceration was observed. The numbers in bold in the center of the figure represents the mean day of death following the tumor challenge. Each group consisted of 7-10 animals.
  • Figure 12 Survival rate following vaccination with either Ad-Ii-VSVGP or Ad- VSVGP.
  • C57BL/6 mice were vaccinated in the right hind foot-pad with 2 x 10 7 IFU of adenovirus encoding either full-length glycoprotein of vesicular stomatitis virus (Ad- VSVGP) or full-length glycoprotein of vesicular stomatitis virus linked to invariant chain (Ad-Ii-VSVGP).
  • Ad- VSVGP full-length glycoprotein of vesicular stomatitis virus
  • Ad-Ii-VSVGP full-length glycoprotein of vesicular stomatitis virus linked to invariant chain
  • Figure 14 Efficiency of Ad-Ii-GP constructs compared to Ad-GP-Lamp-1 constructs measured by CD8 + T-cell responses to adenovirus encoded epitopes.
  • C57BL/6 mice were vaccinated with 2x10 7 IFU of the indicated construct in the right hind footpad.
  • the number of epitope specific CD8+ cells was determined by intracellular staining for peptide-induced IFN- ⁇ of spleen cells. Bars represent Avg ⁇ SD of 3 animals.
  • Figure 15 Vectors based on in-frame polylinkers.
  • Ii without stop-codon was amplified by PCR with the sequence for the AsiSI, Swal, Ascl, Pmel, Fsel and a stop site included in the 3' primer and cloned into the pacCMV vector.
  • the resulting vector was numbered 770 (see partial sequence hereof in SEQ ID NO: 7).
  • a corresponding vector termed 768 (see partial sequence hereof in SEQ ID NO: 8) without the Ii sequence incorporated was also constructed.
  • Figure 16 Vectors with IRES2 sites.
  • pacCMV Ii MCS IRES2 The vector termed "pacCMV Ii MCS IRES2" (and numbered 1163, see partial sequence hereof in SEQ ID NO: 9) was constructed by cloning the IRES2 from pLP-IRES2-EGFP into the 770 vector by PCR and restriction enzyme digestion. The sequences for l-scel and Srfl were included in the 3' primer. The first ATG site after the IRES sequence initiates expression of a second protein. A corresponding vector termed "pacCMV MCS
  • Example 1 The invention will now be further illustrated with reference to the following examples. It will be appreciated that what follows is by way of example only and that modifications in detail may be made while still falling within the scope of the invention.
  • Example 1 Example 1 :
  • CD8 + and CD4 + T-cell responses to adenovirus encoded epitopes CD8 + and CD4 + T-cell responses to adenovirus encoded epitopes.
  • mice C57BL/6 (H-2b), C57BL/6 x BALB/c (H-2bxd) F1 hybrids and MHC H-/- mice (B6.129-H2-Ab1 tm1Glm N12 (H-2b)) were obtained from Taconic M&B (Ry, Denmark). All mice used were between 7-10 weeks old and housed in a specific germ free facility. All experimental procedures were performed according to local experimental guidelines.
  • Adenovirus vectors For construction of E1 and E3 deleted adenovirus-expressing, LCMV derived antigen fused to invariant chain we performed 2-step PCR. First we obtained overlapping PCR products containing the full-length mouse invariant chain and full-length LCMV glycoprotein and these were joined by secondary PCR with invariant chain 5' and glycoprotein 3' primers. Adenovirus expressing full-length GP was amplified in single step PCR. The obtained fragments were cloned into the pacCMV shuttle vector. The obtained plasmid was co-transfected with pJM17 plasmid into HEK293 cells and viral lysates were obtained.
  • mice to be vaccinated were anaesthetized and injected with 2x10 7 infectious units in the right hind footpad in a volume of 0.03 ml.
  • Virus infection Mice were infected i.e. with 20 pfu of LCMV Armstrong clone 53b in a volume of 0.03 ml or i.v. with 10 6 pfu of LCMV clone 13 in 0.3 ml. I.e. infection induces a fatal CD8 + T cell-mediated meningitis from which immunocompetent mice succumb on days 7 to 10 p.i. (post infection) (Christensen et al., Scandinavian Journal of Immunology 40:373-382).
  • Organ virus titers To determine virus titers in organs, these were first homogenized in PBS to yield 10% (v/w) organ suspensions, and serial 10-fold dilutions were prepared. Each dilution was then plated in duplicates on MC57G cells.
  • infected cell clusters Forty-eight hours after infection, infected cell clusters were detected using monoclonal rat anti- LCMV (VL-4) antibody, peroxidase-labeled goat anti-rat antibody and o- phenylendiamin (substrate) (Battegay et al., 1991 , Journal of Virological Methods 33:191-198). The numbers of pfu were counted, and results expressed as pfu/g tissue.
  • Cell preparations Single cell suspensions of spleen cells were obtained by pressing the organs through a fine steel mesh.
  • Abs for flow cytometric The following monoclonal antibodies (mAbs) were purchased from BD PharMingen (San Diego, CA) as rat anti-mouse antibody: Cy- chrome-conjugated anti-CD8, FITC-conjugated anti-CD44, phycoerythrin (PE)- conjugated anti-IFN- ⁇ and PE-conjugated IgGi isotype standard.
  • splenocytes were incubated in vitro for 5 h at 37 0 C in 5% CO 2 with relevant peptide (0.1 ⁇ g/ml) in the presence of monensin (3 ⁇ M, Sigma Chemicals co., St. Louis, MO) and murine recombinant IL-2 (10 units/well, R&D Systems Europe Ltd, Abingdon, UK). After incubation cells were surface stained, washed, fixed and permeabilized using 0.5% saponin. Cells were then stained with anti-IFN- ⁇ or IgGi isotype control for 20 min at 4 0 C.
  • Samples were analyzed using a Becton Dickinson FACSCalibur, and at least 10 4 mononuclear cells were gated using a combination of low angle and side scatter to exclude dead cells and debris. Data analysis was conducted using Cell Quest Pro (B&D Biosciences).
  • Ad-GP lymphocytic choriomeningitis virus full-length glycoprotein
  • Ad-IiGP lymphocytic choriomeningitis virus full- length glycoprotein N-terminally linked to murine invariant chain
  • Ad-GP and Ad-IiGP could also induce an immune response in C57BL/6 x BALB/c Fi mice that express both the H-2 b and H2 d haplotypes. These mice resemble an out bred population, but with defined haplotypes. The experiments were performed as above, but testing was limited to day 21 after vaccination. As can be seen from figure 3, Ad-IiGP efficiently induces CD8 + T-cell responses towards a multitude of epitopes while Ad-GP seemed to perform worse than in homozygous C57BL/6 mice.
  • Ad-IiGP exerts CD8 + T-cell stimulatory effects that are independent of CD4 + T-cells:
  • Ii function in the enhanced stimulation of CD8 + T-cells is the ability to traffic to endosomal and lysosomal compartments and stimulate CD4 + T-cells (Diebold et al., 2001, Gene Ther. 8:487-493) through MHC class II, we performed vaccination of MHC class Il deficient mice. To this effect MHC-I I ' ' " or C57BL/6 mice were vaccinated with 2x10 7 IFU of Ad-GP or Ad-IiGP in the right hind footpad.
  • Ad-IiGP confers rapid, superior and sustained protection against lethal LCMV infection: As we observed an accelerated response to Ad-IiGP compared to Ad-GP we investigated the ability of vaccination to confer protection both at 21 days post vaccination (peak of Ad-GP) and at 3, 5, 7, 14, 60, 90, 180 and 360 days post vaccination (Figure 5). Remarkably, we found that Ad-IiGP vaccinated animals vaccinated as little as 3 days previously were protected against intracerebral LCMV infection. Protection conferred by Ad-GP was only partial at 14 and 21 days post infection and no protection were seen at day 60 or later. Furthermore, the Ad-IiGP conferred protection was sustained for 360 days, at which point Ad-GP no longer protected against intracerebral LCMV infection.
  • Ad-IiGP efficiently protects against high-dose, intravenous LCMV infection: Since we found that Ad-IiGP protected mice against an acute localised infection, we wanted to investigate whether the same held true for a high-dose systemic infection.
  • mice were vaccinated with Ad-GP, Ad-IiGP or sham (PBS), and challenged 21 days later by intravenous injection of 10 6 plaque-forming units of the fast replicating LCMV clone 13 strain. 5 days later animals were sacrificed and infectious titers in the lungs were determined. Although Ad-GP conferred significant protection upon vaccinated animals, Ad-IiGP was superior and reduced titers to a level at or below the detection limit of the assay ( Figure 6).
  • Example 6 Comparative analyses of novel constructs with respect to kinetics, magnitude of response, long-term immunity and virus dose needed for immunity.
  • adenovirus-encoded GP is fused N-terminally to: 1. the lysosomal targeting sequence of Ii 2. the CLIP sequence of Ii
  • constructs are furthermore used as the starting point for a series of constructs in which the GP-fusions are followed by an internal ribosomal entry site (IRES) and a gene encoding VP22, HIV tat or Cx43.
  • IRS internal ribosomal entry site
  • constructs are prepared where adenovirus-encoded GP is followed by an internal ribosomal entry site (IRES) and a gene encoding VP22, HIV tat or Cx43 or other connexins and intercellular gap-junctions constituents.
  • IRS internal ribosomal entry site
  • All of the above constructs are furthermore altered in any of the following ways: 1. All of the above constructs followed by an IRES site and a gene encoding an NK-cell (natural killer cell) activation molecule, for example H60. This alteration gives enhanced delivery of co-stimulatory signals and cytokine help. 2. All of the above constructs involving IRES sites, where the downstream gene is placed under control of a separate promoter. 3. All of the above constructs involving IRES sites, where the downstream gene is instead encoded on a separate vector.
  • Ad-Ii-GP pg33 and gp276 constructs See figure 14 for an example of a comparison of the efficiency of Ad-Ii-GP pg33 and gp276 constructs compared to Ad-GP-Lamp-1 pg33 and gp276 constructs as measured by CD8+ T-cell responses.
  • the Ad-Ii-GP constructs are superior to the Ad-GP-Lamp-1 constructs in their capability of evoking a CD8+ T-cell response.
  • Example 6 All constructs described in Example 6 are comparatively analysed among each other and to Ad-GP and Ad-IiGP following alternative administration methods. The comparisons are done with respect to the kinetics and magnitude of immune response e.g. as described in Examples 1 , 2 and 3, long-term immunity e.g. as described in Examples 1 , 4 and 5, and virus dose needed for immunity.
  • Alternative administration methods include:
  • Alterations with regard to enhanced delivery of co-stimulatory signals and cytokine help in which all of the in Example 6 described constructs are co- injected with adenovirus encoded type 1 interferon, for example tetracycline inducible IFN- ⁇ . Furthermore, all the in Example 6 described constructs are co-injected with adenovirus encoded cytokine, for example IL-15.
  • Ad-IiGP simultaneously with Ad-Tet-onGP at separate sites of the body (Ad-Tet-onGP encodes GP under control of a tetracycline inducible promoter).
  • Ad-IiGP confers rapid and superior protection against lethal LCMV infection in absence of major epitopes.
  • Lymphocytic choriomeningitis virus full-length glycoprotein (GP) comprises four CD8 + specific epitopes of varying antigenicity, measured by the percentile of CD8 + cells with specificity for the individual epitope against all the CD8 + cells raised in response to GP vaccination.
  • the predominant population of CD8 + cells raised against GP is specific for the gp33 epitope, a somewhat smaller population is specific for gp276, and minor populations are specific for gp118 and for gp92.
  • gp33 and gp276 are the major / immunodominant epitopes
  • C57BL/6 mice were vaccinated with 2x10 7 IFU Ad-GP and Ad-IiGP or sham infected in the right hind footpad. Each group consisted of 5 animals. On day 90 post vaccination, the animals were challenged with 20 pfu LCMV Arm 53b i.e. (intra cerebral) constructs carrying gp33 nil mutations, gp276 nil mutations or gp33/gp276 double nil mutations. Mortality was recorded for 14 days.
  • Ad-IiGP conferred superior protection against lethal LCMV infection despite the gp33 or gp276 nil mutations.
  • the double nil mutation of gp33/gp276 lead to the survival of 70% of the animals compared to no surviving animals in the Ad-GP and Sham vaccinated groups.
  • CD8 + and CD4 + T cell responses to vaccination with naked DNA-IiGP and DNA-GP were tested the ability of IiGP and GP as naked DNA to raise GP- epitope specific CD8 + and CD4 + T cells.
  • the naked DNA comprised the GP and Ii- GP fragments from the Adenoviral vectors illustrated in figure 1.
  • C57BL/6 mice were vaccinated with DNA coated onto 1.6-nm gold particles in a concentration of 2 ⁇ g DNA/mg gold, and the DNA-gold complex was coated onto plastic tubes such that 0.5 mg gold (1 ⁇ g DNA per shot) was delivered to the mouse per shot.
  • Mice were immunized at the abdominal skin using a hand-held gene gun device employing compressed helium (400 psi) as the particle motive force. Mice were inoculated four times with an interval of 1 week and then allowed to rest for 1 week before investigation.
  • the number of epitope specific CD8 + or CD4 + T cells were determined by intracellular staining for peptide-induced IFN-y of spleen cells. As can be seen from figure 9, DNA-IiGP efficiently induces CD8 + T-cell responses directed against several epitopes.
  • Ad-IiGP confers superior protection against challenge with tumor cells expressing the gp33 epitope from LCMV. Under normal circumstances, tumor cells express several different antigens recognized by T cells. To determine the efficiency of this response, B16.F10 melanoma cells expressing gp33 from LCMV were used to challenge vaccinated animals with.
  • C57BL/6 mice were vaccinated with 2x10 7 IFU Ad-GP or Ad-IiGP in the right hind footpad. As a control some animals were vaccinated with Ad- ⁇ -galactosidase (negative control) or infected with LCMV (positive control). On day 90 after vaccination/infection animals were challenged with 10 6 tumor cells subcutaneously and the tumor growth was followed by measuring the size of the tumor. Initially a tumor will form in all animals, but eventually the immune response directed towards gp33 will eliminate the tumor cells. Each group consisted of 7-10 animals. Prophylactic vaccination with Ad-Ii-GP resulted in tumor free mice in 70% of the cases compared to only 10% in Ad-GP vaccinated animals, as can be seen from figure 10.
  • mice were injected with 10 6 B16.F10 tumor cells subcutaneously. After 5 days the tumors were palpably recognizable and could be measured. At this time point animals were vaccinated with 2x10 7 IFU Ad-GP or Ad- IiGP in the right hind footpad. As a control some animals were vaccinated with Ad- ⁇ - galactosidase (negative control) or infected with LCMV (positive control). Tumor growth was followed by measuring the size of the tumor, and once the size was greater that 12 mm in any dimension the animals were sacrificed. As can be seen from figure 11 , in mice given the therapeutic Ad-IiGP vaccine, the speed of tumor development was approximately half of that seen in mice given Ad-GP as therapeutic vaccine, as measured by number of days passing prior to reaching a 12 mm tumor size and animal sacrifice.
  • VSV Vesicular stomatitis virus
  • Indiana serotype Indiana serotype was used throughout this study. Stocks of virus were propagated in L929 cells (ATCC CCL 1 ) and stored at -70 0 C until use. Virus quantitation was performed by plaque assay on monolayers of L929 cells. In brief, serial 10-fold dilutions of virus were prepared in Eagle ' s minimal essential medium (F11 ) containing 1% L- glutamine, 1% penicillin/streptomycin, 5% NaHCO 3 and 10% fetal calf serum. One ml of each dilution was added in duplicate to monolayers of L929 cells in petri dishes plated 48 hours earlier.
  • F11 Eagle ' s minimal essential medium
  • Serum neutralizations test Serial two-fold dilutions of serum in F11 were mixed with equal volumes of virus diluted to contain approximately 100 PFU/ml. After 1 h of incubation at room temperature, 1 ml of each serum-virus mixture was added in duplicate to monolayers of L929 cells in petri dishes and assayed for the presence of residual virus by plaque assay (see virus and virus quantitation). The highest serum dilution that reduced the number of plaques by at least 50% was taken as the neutralizing titer.

Abstract

The present invention relates to a vaccine comprising a nucleic acid construct such as a DNA construct especially a nucleic acid construct comprising sequences encoding invariant chain operatively linked to antigenic protein or peptide encoding sequences. The vaccine stimulates an immune response, especially an immune response in an MHC-I dependent, but CD4+ T-cell independent manner.

Description

A NUCLEOTIDE VACCINE
All patent and non-patent references cited in the application, or in the present application, are also hereby incorporated by reference in their entirety.
Field of the invention
The present invention relates to a technology and method whereby a faster, broader and stronger immune response is obtained when using viral and DNA-based vaccines.
Background of the invention
Despite current knowledge in the field of immunology especially regarding vaccine technologies no suitable vaccines are available against numerous pathogens. Widespread pandemics of HIV (Human Immunodeficiency Virus), HTLV (Human T- cell Lymphotropic Virus), tuberculosis and HCV (Hepatitis C virus) remain out of reach of effective vaccination, while bird flu and other emerging pathogens threaten to overwhelm our healthcare systems. Similarly, the burst in world wide terrorism has expanded the potential epidemics to include exotic and lethal pathogens such as Ebola, Lassa and Marburg.
Vaccines can be prophylactic: they are given before the actual infection occurs, or therapeutic: where they elicit or accelerate an immune response to a pathogen already in the body. Both methods of vaccination require the establishment of a solid immune response. The immune response that is activated by infection or vaccination depends on the interaction of several cell types, such as T-, B- and antigen presenting cells as well as several different molecules, primarily antigens, MHC molecules, T- and B-cells receptors and many more.
Antigens are peptide fragments presented on the surface of antigen presenting cells by MHC molecules. Antigens can be of foreign, i.e. pathogenic origin, or stem from the organism itself, so called self or auto antigens. The MHC molecules are representatives of a polymorphous gene family encoded by a specific chromosomal region known as the "major histocompatibility complex", hence MHC. Two classes of MHC molecules exist, MHC class I (MHC-I) and MHC class Il (MHC-II). T-helper cells are stimulated by antigens presented by MHC class Il (MHC-II) molecules residing on the surface of antigen presenting cells. The MHC-II molecules are synthesized in the endoplasmatic reticulum. During synthesis, they combine with invariant chain (Ii) in a manner preventing the MHC-II molecules from being loaded with self- or auto-antigens. The MHC-II molecule is by signal sequences in the invariant chain transported to the cell surface in a specific cellular compartment. As the compartment matures by the processing of its contents it progresses from being a lysosome, to a late endosome (after fusion with endocytotic vesicles) to an MHC class Il compartment (MIIC). The endocytotic vesicle contains foreign antigen i.e. proteolytically cleaved bacterial peptide fragments. These fragments are by their degradation prepared to be loaded onto the MHC-II molecule. The MHC-II molecule is released by the invariant chain in a two part process when the invariant chain first is degraded proteolytically leaving only a peptide termed CLIP in the MHC-II binding domain, secondly by the removal of CLIP by an HLA-DM molecule. The MHC-II molecule is then free to bind the foreign antigens and present these on the cell surface after fusion of the MIIC vesicle to the plasma membrane. This initiates the humoral immune response as the presented antigen stimulates activation of a T- helper cell which in turn by several means activates a B cell, which ultimately differentiates into an antibody secreting cell.
The cellular immune response is initiated when the T-cell receptor of T-cytotoxic cells recognizes antigen bound to the MHC class I molecule on an antigen presenting cell. MHC-I molecules are not associated with a molecule of a functionality like the invariant chain that associates with MHC-II. The processing of MHC-I into an antigen presenting molecule furthermore differs from that of MHC-II molecules in that the MHC-I molecule is loaded with antigen already in the endoplasmatic reticulum. The antigens presented by the MHC-I molecule are typically peptide fragments cleaved by the proteasome of proteins that have been synthesized by the antigen presenting cell itself. These proteins may be abnormal proteins encoded in the cells own DNA or proteins derived from viruses or other pathogens that have infected the cell and parasitize its protein synthesis machinery. The MHC class l-related proteolytic system is present in virtually all cells.
The functions of the two types of T cells are significantly different, as implied by their names. Cytotoxic T cells eradicate intracellular pathogens and tumors by direct lysis of cells and by secreting cytokines such as γ-interferon. The predominant cytotoxic T cell is the CD8+ T cell, which also is antigen specific. Helper T cells also can lyse cells, but their primary function is to secrete cytokines that promote the activities of B cells (antibody-producing cells) and other T cells and thus they broadly enhance the immune response to foreign antigens, including antibody-mediated and cytotoxic T cell-mediated response mechanisms. CD4+ T cells are the major helper T cell phenotype in the immune response.
Traditional vaccines rely on whole organisms, either pathogenic strains that have been killed or strains with attenuated pathogenicity. On the one hand, these vaccines run the risk of introducing the disease they are designed to prevent if the attenuation is insufficient or if enough organisms survive the killing step during vaccine preparation. On the other hand, such vaccines have reduced infectivity and are often insufficiently immunogenic, resulting in inadequate protection from the vaccination.
Recently, molecular biological techniques have been used in an attempt to develop new vaccines based on individual antigenic proteins from the pathogenic organisms. Conceptually, use of antigenic peptides rather than whole organisms would avoid pathogenicity while providing a vaccine containing the most immunogenic antigens. However, it has proven difficult to select the optimal antigen of a given protein or polypeptide and furthermore it has been found that pure peptides or carbohydrates tend to be weak immunogens.
Genetic (DNA) vaccines are new and promising candidates for the development of both prophylactic and therapeutic vaccines. The strength of the ensuing immune response is determined through a combination of the potency of the vector (i.e. naked DNA, viral vectors, live attenuated viruses etc.), the expression level of the antigen, and the recombinant antigen it self (i.e. high or low affinity MHC binders, structural determinants selecting for more or less limited T-or B-cell repertoire etc.). It is generally held to be true, that efficient induction of immunological memory requires or benefits from the interactions of CD4+ (helper cell) T-cells with CD8+ (cytotoxic) T-cells and B-cells that mediate many of the effects of immune memory. However, one potential disadvantage of conventional DNA vaccines is their low immunogenicity in humans. One likely cause of this low immunogenicity is the restricted access of antigens formed within cells to the MHC Il pathway for antigen processing and presentation to T helper cells.
Summary of invention Thus, the present invention has solved the problem of stimulating the immune response in a manner that increases the kinetics of the response, simultaneously with both broadening and improving the response, while avoiding, among other things, the above mentioned disadvantages of the vaccination methods described in the state of the art. In particular, a novel system for a directed, specific and fast stimulation of the immune system is hereby made available in order to improve the vaccination of all animals.
This problem is solved by the embodiments of the present invention characterized in the claims. By the present invention it was found that fusion of an antigen to the invariant chain dramatically enhanced the ensuing antiviral CD4+ and CD8+ T-cell responses. Additionally, and surprisingly, it was found that this effect is obtained through a CD4+ T-cell independent mechanism. It was further found that the protection is both accelerated and enhanced in an acute localized and lethal infection, and enhanced in a high-dose systemic infection.
It is thus an object of the present invention to provide a nucleic acid construct comprising sequences encoding at least one invariant chain operatively linked to at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide.
It is likewise an aspect of the present invention to provide an adenoviral vector comprising a nucleotide construct encoding at least one antigen and at least one protein or peptide or fragment of a protein or peptide which stimulates an immune response.
Thus it is an aspect of the present invention to provide means of stimulating an MHC-I mediated immune response by an adenoviral vector comprising a nucleotide construct encoding at least one antigen and at least one protein or peptide or fragment of a protein or peptide. Another aspect includes stimulating an MHC-II response by an adenoviral vector comprising a nucleotide construct encoding at least one antigen and at least one protein or peptide or fragment of a protein or peptide.
A further aspect provides means of stimulating intercellular spreading of the nucleic , acid construct, the adenoviral vector, the proteins encoded within any of these or any parts of any of these.
It is further an object of the present invention to provide a delivery vehicle comprising the nucleic acid construct as detailed herein, especially a delivery vehicle such as a replication deficient adenoviral vector is relevant to the present invention.
It is yet an object and an aspect of the present invention to provide a cell comprising the nucleic acid construct or the adenoviral vector according to the present invention.
It is yet an object of the present invention to provide a chimeric protein as encoded by the nucleic acid construct described herein or encoded by the adenoviral vector described herein.
It is further an aspect of the present invention to provide an antibody that recognizes the chimeric protein encoded by either the nucleic acid construct or the adenoviral vector described herein.
It is an aspect of the present invention to provide a vaccine comprising the nucleic acid construct or the adenoviral vector as detailed herein. Especially relevant to the present invention is a vaccine, where at least one invariant chain is operatively linked to at least one protein or peptide or fragment of a protein or peptide which stimulates an MHC-I response. This may be done by operatively linking at least one invariant chain with at least one at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide. It is further an aspect of the present invention to provide a vaccine comprising the chimeric protein encoded within the nucleic acid construct or the adenoviral vector as detailed herein.
It is yet an aspect of the present invention to provide a kit in parts, said kit comprising either a vaccine composition comprising an adenoviral vector or a nucleic acid construct as described herein together with a medical instrument or other means of administering said vaccine and furthermore instructions on how to use the kit in parts.
It follows that the present invention provides means for inducing an immune response in an animal, by administering to the animal a vaccine comprising the nucleic acid construct or the adenoviral vector as detailed herein below.
Description of Drawings
Figure 1 : Schematic drawing of inserts in the adenovirus vector.
Figure 2: CD8+ and CD4+ T-cell responses to adenovirus encoded epitopes.
Figure 3: CD8+ and CD4+ T-cell responses to adenovirus encoded epitopes in F1 hybrid mice. Figure 4: Ad-IiGP exerts CD8+ T-cell stimulatory effects that are independent of
CD4+ T-cells Figure 5: Ad-IiGP confers rapid and superior protection against lethal LCMV infection.
Figure 6: Ad-IiGP efficiently protects against high-dose, intravenous LCMV infection.
Figure 7: Ad-IiGP confers superior protection to lethal LCMV variants with mutations in immunodominant epitopes. Figure 8: Frequencies of CD8+ or CD4+ T cells reacting to specific LCMV epitopes after Ad-IiGP vaccination and challenge with LCMV variants with mutations in immunodominant epitopes.
Figure 9: CD8+ and CD4+ T cell responses to vaccination with naked DNA-IiGP and DNA-GP
Figure 10: Prophylactic vaccination with Ad-Ii-GP increases tumor rejection Figure 11 : Therapeutic vaccination with Ad-Ii-GP increases average life span in tumor carrying mice. Figure 12: Survival rate following vaccination with either Ad-Ii-VSVGP or Ad- VSVGP.
Figure 13: CD8+ and CD4+ T-cell responses to more adenovirus encoded epitopes Figure 14: Efficiency of Ad-Ii-GP constructs compared to Ad-GP-Lamp-1 constructs measured by CD8+ T-cell responses to adenovirus encoded epitopes.
Figure 15: Vector based on in-frame polylinkers.
Figure 16: Vectors with IRES2 sites
Detailed description of the invention
Definitions:
Adenovirus: A group of double-stranded DNA containing viruses. Adenoviruses can be genetically modified making them replication incompetent or conditionally replication incompetent. In this form, as adenoviral constructs or adenovectors, they can be used as gene delivery vehicles for vaccination or gene therapy.
Adjuvant: Any substance whose admixture with an administered immunogenic determinant / antigen / nucleic acid construct increases or otherwise modifies the immune response to said determinant. Amino acid: Any synthetic or naturally occurring amino carboxylic acid, including any amino acid occurring in peptides and polypeptides including proteins and enzymes synthesized in vivo thus including modifications of the amino acids. The term amino acid is herein used synonymously with the term "amino acid residue" which is meant to encompass amino acids as stated which have been reacted with at least one other species, such as 2, for example 3, such as more than 3 other species. The generic term amino acid comprises both natural and non-natural amino acids any of which may be in the "D" or "L" isomeric form.
Antibody: Immunoglobulin molecules and active portions of immunoglobulin molecules. Antibodies are for example intact immunoglobulin molecules or fragments thereof retaining the immunologic activity.
Antigen: Any substance that can bind to a clonally distributed immune receptor (T- cell or B-cell receptor). Usually a peptide, polypeptide or a multimeric polypeptide.
Antigens are preferably capable of eliciting an immune response. Boost: To boost by a booster shot or dose is to give an additional dose of an immunizing agent, such as a vaccine, given at a time after the initial dose to sustain the immune response elicited by the previous dose of the same agent.
Carrier: Entity or compound to which antigens are coupled to aid in the induction of an immune response.
Chimeric protein: A genetically engineered protein that is encoded by a nucleotide sequence made by a splicing together of two or more complete or partial genes or a series of (non)random nucleic acids.
Complement: A complex series of blood proteins whose action "complements" the work of antibodies. Complement destroys bacteria, produces inflammation, and regulates immune reactions.
Cytokine: Growth or differentiation modulator, used non-determinative herein, and should not limit the interpretation of the present invention and claims. In addition to the cytokines, adhesion or accessory molecules, or any combination thereof, may be employed alone or in combination with the cytokines.
CTL: Cytotoxic T lymphocytes. A sub group of T-cells expressing CD8 along with the T-cell receptor and therefore able to respond to antigens presented by class I molecules.
Delivery vehicle: An entity whereby a nucleotide sequence or polypeptide or both can be transported from at least one media to another.
Fragment: is used to indicate a non-full length part of a nucleic acid or polypeptide.
Thus, a fragment is itself also a nucleic acid or polypeptide, respectively.
Individual: Any species or subspecies of bird, mammal, fish, amphibian, or reptile.
Invariant chain: an integral membrane protein glycoprotein that associates with and stabilizes MHC Il molecules in the endoplasmatic reticulum and subsequent cellular compartments. Here the term invariant chain covers all naturally occurring or artificially generated full length or fragmented homologous genes and proteins of a certain similarity to human invariant chain. Invariant chain is herein abbreviated Ii.
Isolated: used in connection with nucleic acids, polypeptides, and antibodies disclosed herein 'isolated' refers to these having been identified and separated and/or recovered from a component of their natural, typically cellular, environment.
Nucleic acids, polypeptides, and antibodies of the invention are preferably isolated, and vaccines and other compositions of the invention preferably comprise isolated nucleic acids, polypeptides or isolated antibodies. MHC: Major histocompatibility complex, two main subclasses of MHC, Class I and Class Il exist.
Nucleic acid: A chain or sequence of nucleotides that convey genetic information. In regards to the present invention the nucleic acid is a deoxyribonucleic acid (DNA). Nucleic acid construct: A genetically engineered nucleic acid. Typically comprising several elements such as genes or fragments of same, promoters, enhancers, terminators, polyA tails, linkers, polylinkers, operative linkers, multiple cloning sites (MCS), markers, STOP codons, other regulatory elements, internal ribosomal entry sites (IRES) or others. Operative linker: A sequence of nucleotides or amino acid residues that bind together two parts of a nucleic acid construct or (chimeric) polypeptide in a manner securing the biological processing of the nucleic acid or polypeptide. Pathogen: a specific causative agent of disease, especially a biological agent such as a virus, bacteria, prion or parasite that can cause disease to its host, also referred to as an infective agent.
Peptide: Plurality of covalently linked amino acid residues defining a sequence and linked by amide bonds. The term is used analogously with oligopeptide and polypeptide. The natural and/or non-natural amino acids may be linked by peptide bonds or by non-peptide bonds. The term peptide also embraces post-translational modifications introduced by chemical or enzyme-catalyzed reactions, as are known in the art. The term can refer to a variant or fragment of a polypeptide. Pharmaceutical carriers: also termed excipients, or stabilizers are non-toxic to the cell or individual being exposed thereto at the dosages and concentrations employed. Often the physiologically acceptable carrier is an aqueous pH buffered solution. Examples of physiologically acceptable carriers include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEEN.TM., polyethylene glycol (PEG), and PLURONICS.TM. Plurality: At least two. Promoter: A binding site in a DNA chain at which RNA polymerase binds to initiate transcription of messenger RNA by one or more nearby structural genes. Signal peptide: A short sequence of amino acids that determine the eventual location of a protein in the cell, also referred to as sorting peptide. siRNA: Small interfering RNAs (siRNAs), which target (in a sequence-specific manner) endogenous RNAs for degradation, thereby reducing the amount of a gene product.
Surfactant: A surface active agent capable of reducing the surface tension of a liquid in which it is dissolved. A surfactant is a compound containing a polar group which is hydrophilic and a non polar group which is hydrophobic and often composed of a fatty chain.
Vaccine: A substance or composition capable of inducing an immune response in an animal. Also referred to as an immunogenic composition in the present text. An immune response being an immune response (humoral/antibody and/or cellular) inducing memory in an organism, resulting in the infectious agent, being met by a secondary rather than a primary response, thus reducing its impact on the host organism. A vaccine of the present invention may be given as or prophylactic and/or therapeutic medicament. The composition may comprise one or more of the following: antigen(s), nucleic acid constructs comprising one or more antigens operatively linked to Ii, carriers, adjuvants and pharmaceutical carriers..
Variant: a 'variant' of a given reference nucleic acid or polypeptide refers to a nucleic acid or polypeptide that displays a certain degree of sequence homology/identity to said reference nucleic acid or polypeptide but is not identical to said reference nucleic acid or polypeptide.
The present invention relates to a vaccine comprising a nucleic acid construct such as a DNA construct especially a nucleic acid construct comprising sequences encoding invariant chain operatively linked to antigenic protein or peptide encoding sequences. The vaccine stimulates an immune response, especially an immune response in an MHC-I dependent, but CD4+ T-cell independent manner.
Nucleic acid construct
An aspect of the present invention relates to nucleic acid constructs comprising sequences encoding at least one invariant chain operatively linked to at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide, in short an antigen.
By nucleic acid construct is understood a genetically engineered nucleic acid. The nucleic acid construct may be a non-replicating and linear nucleic acid, a circular expression vector, an autonomously replicating plasmid or viral expression vector. A nucleic acid construct may comprise several elements such as, but not limited to genes or fragments of same, promoters, enhancers, terminators, poly-A tails, linkers, polylinkers, operative linkers, multiple cloning sites (MCS), markers, STOP codons, internal ribosomal entry sites (IRES) and host homologous sequences for integration or other defined elements. Methods for engineering nucleic acid constructs are well known in the art (see, e.g., Molecular Cloning: A Laboratory Manual, Sambrook et al., eds., Cold Spring Harbor Laboratory, 2nd Edition, Cold Spring Harbor, N.Y., 1989).
Examples of parts of nucleic acid constructs are given in figures 1 , 15 and 16, as well as in the sequences identified as SEQ ID NO: 5, 6, 7, 8, 9, and 10. The partial vector sequences of SEQ ID NO: 5, 6, 7, 8, 9, and 10 have been generated by subcloning various elements as described in the above and as illustrated in figures 1, 15 and 16. These partial sequences are all inserted into the pAC-CMVpLpARS(+) vector (Becker et al., 1994, Methods Cell Biol. 43 Pt A:161-189), see GenBank accession number AY590429.1.
Invariant chain The invariant chain (Ii) or CD74, is a non-polymorphic type Il integral membrane protein, see SEQ ID NOs: 2 and 4 for the amino acid sequences of human and mouse Ii, respectively, and likewise SEQ ID NOs: 1 and 3 for the nucleic acid sequences of human and mouse Ii, respectively. Invariant chain has multiple functions in lymphocyte maturation and in adaptive immune responses, in particular targeting to lysosomal compartments were the Ii CLIP sequence can occupy MHC class Il molecules until these are fused with endosomal compartments (Pieters J. 1997, Curr. Opin. Immunol., 9:8996). Additionally Ii has been shown to function as an MHC class I chaperone (Morris et al, 2004, Immunol. Res. 30:171-179) and by its endosomal targeting sequence, to facilitate stimulation of CD4+, but not CD8+ T-cells directed against covalently linked antigen (Diebold et al., 2001 , Gene Ther. 8:487- 493).
The invariant chain protein comprises several domains: a cytosolic domain which includes a signal or sorting peptide (also known as the lysosomal targeting sequence), a transmembrane domain, and a luminal domain which in itself comprises a CLIP region, KEY region, core domain and trimerization domain. Both of these domains are flanked by highly flexible regions (Strumptner-Cuvelette & Benaroch, 2002, Biochem. Biophys. Acta., 1542:1-13). Invariant chain has been characterized in several organisms, including vertebrates (e.g. chicken), mammals (e.g. cow, dog, mouse and rat) and human.
The present invention relates to nucleic acid constructs comprising sequences wherein at least one invariant chain is organism specific or can be related to a specific organism. Preferably, at least one invariant chain is of vertebrate origin, more preferably of mammalian origin and most preferably of human origin. In relation hereto the sequence defined by SEQ ID NO: 1 is the nucleic acid sequence of the invariant chain from human. The employed invariant chain is preferably the invariant chain of the organism that is to receive the vaccination. It is an object of the present invention that the invariant chain and the host organisms or receivers of the treatment are of the same species.
The present invention also relates to a nucleic acid construct wherein the encoded at least one invariant chain is a fragment of the sequence identified in SEQ ID NO: 2 of at least 40 amino acids and of at least 85% identity to the same fragment of SEQ ID NO: 2.
The fragment is a fragment of at least 40 amino acids from any part of the invariant chain as set forth in SEQ ID NO: 2. This includes a fragment including residues 1 to 40, 10 to 50, 20 to 60, 25 to 65, 30 to 70, 35 to 75, 40 to 80, 45 to 85, 50 to 90, 55 to 95, 60 to 100, 65 to 105, 70 to 110, 75 to 115, 80 to 120, 85 to 125, 90 to 130, 95 to 135, 100 to 140, 105 to 145, 110 to 150, 115 to 155, 120 to 160, 125 to 165, 130 to 170, 135 to 175, 140 to 180, 145 to 185, 150 to 190, 155 to 195, 160 to 200,165 to 205, 170 to 210 and 175 to 216. It also includes fragments as any of the above listed expanding up to 5 residues to either side hereof. It further includes fragment of at least 50 residues, of at least 60 residues, of at least 70 residues, of at least 80 residues, of at least 90 residues, of at least 100 residues, of at least 110 residues, of at least 120 residues, of at least 130 residues, of at least 140 residues, of at least 150 residues, of at least 160 residues, of at least 170 residues, of at least 180 residues of at least 190 residues, of at least 200 residues and of at least 210 residues.
Any of the above described fragments of at least 85 % sequence identity, for example at least 90 % sequence identity, for example at least 91% sequence identity, such as at least 92 % sequence identity, for example at least 93 % sequence identity, such as at least 94 % sequence identity, for example at least 95 % sequence identity, such as at least 96 % sequence identity, for example at least 97% sequence identity, such as at least 98 % sequence identity, for example 99% sequence identity with SEQ ID NO: 2 are included within the scope of the present invention.
The identity/homology between amino acid sequences may be calculated using well known scoring matrices such as any one of BLOSUM 30, BLOSUM 40, BLOSUM 45, BLOSUM 50, BLOSUM 55, BLOSUM 60, BLOSUM 62, BLOSUM 65, BLOSUM 70, BLOSUM 75, BLOSUM 80, BLOSUM 85, and BLOSUM 90.
Preferably, the present invention is a nucleic acid construct wherein the encoded at least one invariant chain is a fragment of the SEQ ID NO: 2 of at least 186 amino acids. This includes any of the fragments as defined above, and which thus share identity with the sequence of the invariant chain of SEQ ID NO: 2.
The present invention furthermore relates to a nucleic acid construct wherein the encoded at least one invariant chain is at least 85% identical to SEQ ID NO: 2.
This encompasses that any sequence derived from the invariant chain as put forward in SEQ ID NO: 2 of at least 85 % sequence identity, for example at least 90 % sequence identity, for example at least 91% sequence identity, such as at least 92 % sequence identity, for example at least 93 % sequence identity, such as at least 94 % sequence identity, for example at least 95 % sequence identity, such as at least 96 % sequence identity, for example at least 97% sequence identity, such as at least 98 % sequence identity, for example 99% sequence homology with SEQ ID NO: 2 are included within the scope of the present invention. This includes sequences that are either longer or shorter than the sequence described in SEQ ID NO: 2.
Most preferably, the present invention relates to a nucleic acid construct wherein the encoded at least one invariant chain is identical to SEQ ID NO: 2.
Any of the above described sequences regardless of origin, sequence identity or length are from hereon termed variants of invariant chain.
It follows, that it is within the scope of the present invention that a variant of invariant chain from any organism may be a variant according to the above, i.e. that the variant may be a fragment of the invariant chain of an organism and/or be at least 85% identical to said invariant chain either over all the sequence of the invariant chain or within the fragment of same. The invariant chain may also be from a related species of organism or be from a distantly related species.
Another aspect of the present invention relates to the addition, removal or substitution of regions, peptides or domains of the at least one invariant chain as encoded by the nucleic acid construct. The removal of one or more of these regions, peptides or domains will truncate the resulting invariant chain. The addition or replacement of a region, peptide or domain includes the options of choosing these sequences from known sources such as naturally occurring proteins or polypeptides or from artificially synthesized polypeptides or nucleic acids encoding the same. The addition of regions, domains or peptides includes the option of adding one, two or more of each type or of different types of regions, domains, peptides and one, two, three or more of the nucleic acids encoding these regions, domains and peptides. These may be identical or differ from one another based on the sequence. The regions, peptides and domains need not arise from the same organism as the scaffold invariant chain. It is well known in the art to perform additions, deletions and substitutions of individual as well as stretches of nucleotides which will encode the resulting polypeptide. Aligning nucleic acid and especially protein sequences of homologous genes or proteins from different organisms can be of great assistance when determining which substitutions, deletions, rearrangements or other alterations it would be beneficial to construct. Aligning human and murine invariant chain sequences as illustrated below, gives an indication of which amino acid residues may be of importance for the structure and function of the invariant chain in these organisms - these are the residues which are conserved between the two sequences. Likewise, the presumably less important residues are the ones in which the sequences differ. It is of interest in regard to the present invention to perform substitutions and/or deletions of the variant residues / regions. When attempting to mutate or delete or otherwise alter the sequence of e.g. the human invariant chain in order to improve its immune response stimulating capacity, it may also be relevant to examine the conserved residues and make e.g. homologous substitutions (i.e. substitutions where the amino acids are considered to be of e.g. same structural quality, polarity, hydrophobicity or other).
human 1 MDDQRDLISNNEQLPMLGRRPGAPESKCSRGALYTGFSILVTLLLAGQATTAYFLYQQQG murine 1 MDDQRDLISNHEQLPILGNRPREPE-RCSRGALYTGVSVLVALL]-AGQATTAYFLYQQQG human 61 RLDKLTVTSQNLQLENLRMKLPKPPKPVSKMRMATPLLMQALPMGALPQGPMQNATKYGN murine 60 RLDKLTITSQNLQLESLRMKLPKSAKPVSQMRMATPLLMRPMSMDNMLLGPVKNVTKYGN human 121 MTEDHVMHLLQNADPLKVYPPLKGSFPENLRHLKNTMETIDWKVFESWMHHWLLFEMSRH murine 120 MTQDHVMHLLTRSGPLE-YPQLKGTFPENLKHLKNSMDGWWKIFESWMKQWLLFEMSKN human 181 SLEQK-PTDAPPKESLELEDPSSGLGVTKQDLGPVPM murine 179 SLEEKKPTEAPPKEPLDMEDLSSGLGVTRQELGQVTL
A preferred embodiment of the present invention relates to the at least one invariant chain wherein the signal peptide is removed, replaced or added onto the sequence encoding the invariant chain. A signal peptide is a short sequence of amino acids that determine the eventual location of a protein in the cell, also referred to as a sorting peptide. Signal peptides that determine the location of proteins to subcellular compartments such as the endoplasmatic reticulum, golgi apparatus and the various compartments comprising the golgi apparatus, the nucleus, the plasma membrane, mitochondria and the various spaces and membranes herein, peroxisomes, lysosomes, endosomes and secretory vesicles among others are all included within the scope of the present invention. A preferred embodiment comprises alone the lysosomal targeting sequence of invariant chain. Another preferred embodiment comprises alone the KEY region of invariant chain. Another preferred embodiment of the present invention relates to the removal, addition, or replacement of the CLIP region of the at least one invariant chain. As described above, the addition or replacement of the CLIP region includes the options of adding or replacing the existing CLIP region in the variant of the invariant chain or chains chosen, with CLIP regions from invariant chains of the same or other organisms or of variants of CLIP regions form the same or other organisms. The variant CLIP regions may, as follows from the above, be specifically generated mutant versions of the CLIP region, generated by single or multiple nucleic acid substitutions, deletions or additions. A preferred embodiment comprises the CLIP region alone, or the CLIP region together with the N-terminally adjacent sequence or the C-terminally adjacent sequence without any other regions or domains of invariant chain. Other preferred embodiments comprise alone the N-terminally or C- terminally adjacent sequences to the CLIP region but without the CLIP region itself. By adjacent is meant any amino acids within 10 residues of the CLIP region, within 20 residues, within 30 residues, within 40 residues, within 50 residues, within 75 residues or within 100 residues of the CLIP region.
An embodiment of the present invention relates to fragments of invariant chain as described above without the CLIP region. These fragments may be at least 5 amino acid residues long, at least 10 residues, at least 15 residues, at least 20 residues, at least 25 residues, at least 30 residues or at least 35 residues in length. Another embodiment relates to fragments of invariant chain wherein the signal peptide is removed and the invariant chain fragment is at least 10 amino acid residues long, at least 15 residues, at least 20 residues, at least 25 residues, at least 30 residues, at least 35 residues, at least 50 residues at least 60 residues, at least 70 residues at least 80 residues, at least 90 residues, at least 100 residues, at least 110 residues at least 120 residues at least 130 residues, at least 140 residues, at least 150 residues, at least 160 residues, at least 170 residues, or at least 180 residues in length.
Antigen
Any of the above variants of invariant chain are encompassed in the present invention in the form wherein at least one of said variants is operatively linked to at least one antigen such as an antigenic protein or peptide or an antigenic fragment of said protein or peptide.
It is an object of the present invention to include but not limit the antigenic proteins or peptides or fragments of said proteins or peptides to stem from pathogenic organisms, cancer-specific polypeptides and antigens, and proteins or peptides associated with an abnormal physiological response.
More preferably it is an object of the present invention to include an antigen originating from any of the following types of pathogens: virus, micro organisms and parasites. This includes pathogens of any animal known. It is preferable to have an antigen from a mammalian pathogen i.e. a pathogen that specifically targets mammalian animals. It is more preferred to have an antigen from a human pathogen. In general, any antigen that is found to be associated with a human pathogen may be used.
In a preferred embodiment at least one antigen may originate from, but is not limited to any of the following families of virus: Adenovirus, arenaviridae, astroviridae, bunyaviridae, caliciviridae, coronaviridae, flaviviridae, herpesviridae, orthomyxoviridae, paramyxoviridae, picomaviridae, poxviridae, reoviridae, retroviridae, rhabdoviridae and togaviridae.
More specifically at least one antigen or antigenic sequence may be derived from any of the following virus: Influenza A such as H1N1, H1N2, H3N2 and H5N1 (bird flu), Influenza B, Influenza C virus, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus, Hepatitis D virus, Hepatitis E virus, Rotavirus, any virus of the Norwalk virus group, enteric adenoviruses, parvovirus, Dengue fever virus, Monkey pox, Mononegavirales, Lyssavirus such as rabies virus, Lagos bat virus, Mokola virus, Duvenhage virus, European bat virus 1 & 2 and Australian bat virus, Ephemerovirus, Vesiculovirus, Vesicular Stomatitis Virus (VSV), Herpesviruses such as Herpes simplex virus types 1 and 2, varicella zoster, cytomegalovirus, Epstein-Bar virus (EBV), human herpesvirusses (HHV), human herpesvirus type 6 and 8, Human immunodeficiency virus (HIV), papilloma virus, murine gammaherpesvirus, Arenaviruses such as Argentine hemorrhagic fever virus, Bolivian hemorrhagic fever virus, Sabia-associated hemorrhagic fever virus, Venezuelan hemorrhagic fever virus, Lassa fever virus, Machupo virus, Lymphocytic choriomeningitis virus (LCMV), Bunyaviridiae such as Crimean-Congo hemorrhagic fever virus, Hantavirus, hemorrhagic fever with renal syndrome causing virus, Rift Valley fever virus, Filoviridae (filovirus) including Ebola hemorrhagic fever and Marburg hemorrhagic fever, Flaviviridae including Kaysanur Forest disease virus, Omsk hemorrhagic fever virus, Tick-borne encephalitis causing virus and Paramyxoviridae such as Hendra virus and Nipah virus, variola major and variola minor (smallpox), alphaviruses such as Venezuelan equine encephalitis virus, eastern equine encephalitis virus, western equine encephalitis virus, SARS-associated coronavirus (SARS-CoV), West Nile virus, any encephaliltis causing virus.
In a preferred embodiment of the invention the at least one antigenic protein or peptide is from a virus selected from the group of: HIV, Hepatitis C virus, influenza virus, herpes virus, Lassa, Ebola, smallpox, Bird flu, filovirus, Marburg, and papilloma virus.
In a more preferred embodiment of the invention the at least one antigenic protein or peptide is selected from the group of and/or may be at least one antigenic fragment of any of the following: vesicular stomatitis virus glycoprotein (VSV-GP), Influenza A NS-1 (non-structural protein 1 ), Influenza A M1 (matrix protein 1), Influenza A NP (nucleoprotein), LCMV NP, LCMV GP, Ebola GP, Ebola NP, murine gammaherpesvirus M2, M3 and ORF73 (such as MHV-68 M2, M3 and ORF73), chicken Ovalbumin (OVA), or a helper T-cell epitope. It is within the scope of the invention to combine two or more of any of the herein mentioned antigens.
An embodiment of the present invention includes at least one antigenic protein or peptide or fragment of an antigenic protein or peptide from a micro organism. More specifically at least one antigen may be derived from the one of the following from a non-exhaustive list: Anthrax (Bacillus anthracis), Mycobacterium tuberculosis, Salmonella (Salmonella gallinarum, S. pullorum, S. typhi, S. enteridtidis, S. paratyphi, S. dublin, S. typhimurium), Clostridium botulinum, Clostridium perfringens, Corynebacterium diphtheriae, Bordetella pertussis, Campylobacter such as Campylobacter jejuni, Crytococcus neoformans, Yersinia pestis, Yersinia enterocolitica, Yersinia pseudotuberculosis, Listeria monocytogenes, Leptospira species, Legionella pneumophila, Borrelia burgdorferi, Streptococcus species such as Streptococcus pneumoniae, Neisseria meningitides, Haemophilus influenzae, Vibrio species such as Vibrio cholerae 01, V. cholerae non-O1 , V. parahaemolyticus, V. parahemolyticus, V. alginolyticus, V. furnissii, V. carchariae, V. hollisae, V. cincinnatiensis, V. metschnikovii, V. damsela, V. mimicus, V. fluvialis, V. vulnificus, Bacillus cereus, Aeromonas hydrophila, Aeromonas caviae,
Aeromonas sobria & Aeromonas veronii, Plesiomonas shigelloides, Shigella species such as Shigella sonnei, S. boydii, S. flexneri, and S. dysenteriae, Enterovirulent Escherichia coli EEC (Escherichia coli - enterotoxigenic (ETEC), Escherichia coli - enteropathogenic (EPEC), Escherichia coli O157:H7 enterohemorrhagic (EHEC), Escherichia coli - enteroinvasive (EIEC)), Staphylococcus species, such as S. aureus and especially the vancomycin intermediate/resistant species (VISA/VRSA) or the multidrug resistant species (MRSA), Shigella species, such as S. flexneri, S. sonnei, S. dysenteriae, Cryptosporidium parvum, Brucella species such as B. abortus, B. melitensis, B.ovis, B. suis, and B. canis, Burkholderia mallei and Burkholderia pseudomallei, Chlamydia psittaci, Coxiella burnetii, Francisella tularensis, Rickettsia prowazekii, Histoplasma capsulatum, Coccidioides immitis.
In a preferred embodiment of the invention the at least one antigenic protein or peptide is from a micro-organism selected from the group of: Mycobacterium tuberculosis, Bacillus anthracis, Staphylococcus species, and Vibrio species.
An embodiment of the invention relates to a nucleic acid construct, wherein the at least one antigenic protein or peptide encoded is from a parasite.
Another embodiment of the present invention relates to a nucleic acid construct comprising combinations of at least two antigenic proteins or peptides from any of the abovementioned pathogens.
Preferably the antigen is derived from, but not limited to, a parasite selected from the group of: Plasmodium species such as Plasmodium malariae, Plasmodium ovale, Plasmodium vivax, Plasmodium falciparum, Endolimax nana, Giardia lamblia, Entamoeba histolytica, Cryptosporidum parvum, Blastocystis hominis, Trichomonas vaginalis, Toxoplasma gondii, Cyclospora cayetanensis, Cryptosporidium muris, Pneumocystis carinii, Leishmania donovani, Leishmania tropica, Leishmania braziliensis, Leishmania mexicana, Acanthamoeba species such as Acanthamoeba castellanii, and A. culbertsoni, Naegleria fowleri, Trypanosoma cruzi, Trypanosoma brucei rhodesiense, Trypanosoma brucei gambiense, lsospora belli, Balantidium coli, Roundworm (Ascaris lumbricoides), Hookworm (Necator Americanus, Ancylostoma duodenal), Pinworm (Enterobius vermicularis), Roundworm (Toxocara canis, Toxocara cati), Heart worm (Dirofilaria immitis), Strongyloides (Stronglyoides stercoralis), Trichinella (Trichinella spiralis), Filaria (Wuchereria bancrofti, Brugia malayi, Onchocerca volvulus, Loa loa, Mansonella streptocerca, Mansonella perstans, Mansonella ozzardij, and Anisakine larvae (Anisakis simplex (herring worm), Pseudoterranova (Phocanema, Terranova) decipiens (cod or seal worm), Contracaecum species, and Hysterothylacium (Thynnascaris species) Trichuris trichiura, Beef tapeworm (Taenia saginata), Pork tapeworm (Taenia solium), Fish tapeworm (Diphyllobothrium latum), and Dog tapeworm (Dipylidium caninum), Intestinal fluke (Fasciolopsis buski), Blood fluke (Schistosoma japonicum, Schistosoma mansoni) Schistosoma haematobium), Liver fluke (Clonorchis sinensis), Oriental lung fluke (Paragonimus westermani), and Sheep liver fluke (Fasciola hepatica), Nanophyetus salmincola and N. schikhobalowi.
In a preferred embodiment of the invention the at least one antigenic protein or peptide is from a parasite selected from the group of: Plasmodium species, Leishmania species, and Trypanosoma species.
An aspect of the present invention relates antigens and/or antigenic sequences derived from diseases or agents that infect domestic animals, especially commercially relevant animals such as pigs, cows, horses, sheep, goats, llamas, rabbits, mink, mice, rats, dogs, cats, poultry such as chicken, turkeys, pheasants and others, fish such as trout, salmon and other farmed species. Examples of diseases or agents here of from which at least one antigen or antigenic sequence may be derived include, but are not limited to: Multiple species diseases such as: Anthrax, Aujeszky's disease, Bluetongue, Brucellosis such as: Brucella abortus, Brucella melitensis or Brucella suis; Crimean Congo haemorrhagic fever,
Echinococcosis/hydatidosis, virus of the family Picornaviridae, genus Aphthovirus causing Foot and Mouth disease especially any of the seven immunologically distinct serotypes: A, O, C, SAT1 , SAT2, SAT3, Asial , or Heartwater, Japanese encephalitis, Leptospirosis, New world screwworm (Cochliomyia hominivorax), Old world screwworm (Chrysomya bezziana), Paratuberculosis, Q fever, Rabies, Rift Valley fever, Rinderpest, Trichinellosis, Tularemia, Vesicular stomatitis or West Nile fever; Cattle diseases such as: Bovine anaplasmosis, Bovine babesiosis, Bovine genital campylobacteriosis, Bovine spongiform encephalopathy, Bovine tuberculosis, Bovine viral diarrhoea, Contagious bovine pleuropneumonia, Enzootic bovine leukosis, Haemorrhagic septicaemia, Infectious bovine rhinotracheitis / infectious pustular vulvovaginitis, Lumpky skin disease, Malignant catarrhal fever, Theileriosis, Trichomonosis or Trypanosomosis (tsetse-transmitted); Sheep and goat diseases such as: Caprine arthritis / encephalitis, Contagious agalactia, Contagious caprine pleuropneumonia, Enzootic abortion of ewes (ovine chlamydiosis), Maedi-visna, Nairobi sheep disease, Ovine epididymitis (Brucella ovis), Peste des petits ruminants, Salmonellosis (S. abortusovis), Scrapie, Sheep pox and goat pox; Equine diseases such as: African horse sickness, Contagious equine metritis, Dourine, Equine encephalomyelitis (Eastern), Equine encephalomyelitis (Western), Equine infectious anaemia, Equine influenza, Equine piroplasmosis, Equine rhinopneumonitis, Equine viral arteritis, Glanders, Surra (Trypanosoma evansi) or Venezuelan equine encephalomyelitis; Swine diseases such as: African swine fever, Classical swine fever, Nipah virus encephalitis, Porcine cysticercosis, Porcine reproductive and respiratory syndrome, Swine vesicular disease or Transmissible gastroenteritis; Avian diseases such as: Avian chlamydiosis, Avian infectious bronchitis, Avian infectious laryngotracheitis, Avian mycoplasmosis (M. gallisepticum), Avian mycoplasmosis (M. synoviae), Duck virus hepatitis, Fowl cholera, Fowl typhoid, Highly pathogenic avian influenza this being any Influenzavirus A or B and especially H5N1 , Infectious bursal disease (Gumboro disease), Marek's disease, Newcastle disease, Pullorum disease or Turkey rhinotracheitis; Lagomorph and rodent diseases such as: Virus enteritis,
Myxomatosis or Rabbit haemorrhagic disease; Fish diseases such as: Epizootic haematopoietic necrosis, Infectious haematopoietic necrosis, Spring viraemia of carp, Viral haemorrhagic septicaemia, Infectious pancreatic necrosis, Infectious salmon anaemia, Epizootic ulcerative syndrome, Bacterial kidney disease (Renibacterium salmoninarum), Gyrodactylosis (Gyrodactylus salaris), Red sea bream iridoviral disease; or other diseases such as Camelpox or Leishmaniosis.
In a preferred embodiment of the invention the at least one antigenic protein or peptide is from Aujeszky's disease, Foot and mouth disease, Vesicular stomatitis virus, Avian influenza or Newcastle disease. Yet a preferred embodiment of the present invention relates to the at least one antigenic protein or peptide or fragment of said antigenic protein or peptide being an antigenic peptide or protein with at least 85% identity to any of the above described antigens. The homology or identity between amino acids may be calculated by any of the previously mentioned BLOSUM scoring matrices.
An embodiment of the invention relates to a nucleic acid construct, wherein the at least one antigenic protein or peptide or fragment of an antigenic protein or peptide is from a cancer-specific polypeptide or cancer antigen.
Many protein/glycoproteins have been identified and linked to certain types of cancer; these are referred to as cancer specific polypeptides, tumor-associated antigens or cancer antigens. In general, any antigen that is found to be associated with cancer tumors may be used. One way in which cancer specific antigens may be found is by subtraction analyses such as various micro array analyses, such as DNA microarray analysis. Herein the gene expression pattern (as seen in the level of RNA or protein encoded by said genes) between healthy and cancerous patients, between groups of cancerous patients or between healthy and cancerous tissue in the same patient is compared. The genes that have approximately equal expression levels are "subtracted" from each other leaving the genes / gene products that differ between the healthy and cancerous tissue. This approach is known in the art and may be used as a method of identifying novel cancer antigens or to create a gene expression profile specific for a given patient or group of patients. Antigens this identified, both single antigen and the combinations in which they may have been found fall within the scope of the present invention.
Preferably the at least one antigen of the present invention is derived from, but not limited to, a cancer specific polypeptide selected from the group of: MAGE-3, MAGE-1 , gplOO, gp75, TRP-2, tyrosinase, MART-1 , CEA, Ras, p53, B-Catenin, gp43, GAGE-1 , BAGE-1 , PSA, MUC-1 , 2, 3, and HSP-70, TRP-1 , gp100/pmel17, .beta. -HCG, Ras mutants, p53 mutants, HMW melanoma antigen, MUC-18, HOJ-1 , cyclin-dependent kinase 4 (Cdk4), Caspase 8, HER-2/neu, Human papilloma virus HPV type 6, 11 , 16, 18, 31 and 33, Bcr-Abl tyrosine kinase, carcinoembryonic antigen (CEA), telomerase, and SV40 Large T. A preferred embodiment of the invention, the at least one antigenic protein or peptide or fragment of an antigenic protein or peptide is from a cancer-specific polypeptide selected from the group of: p53, HER-2/neu, telomerase, and melanoma antigen.
An embodiment of the invention relates to a nucleic acid construct, wherein the at least one antigenic protein or peptide or fragment of an antigenic protein or peptide is from a polypeptide associated with an abnormal physiological response. Such an abnormal physiological response includes, but is not limited to autoimmune diseases, allergic reactions, cancers and congenital diseases. A non-exhaustive list of examples of hereof includes diseases such rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, psoriasis and Crohn's disease.
Operative linker
An aspect of the present invention relates to the nucleic acid construct wherein the operative link between the invariant chain and the antigenic protein or peptide or fragment of antigenic protein or peptide either is a direct link or a link mediated by a spacer region. By the term operative linker is understood a sequence of nucleotides or amino acid residues that bind together two parts of a nucleic acid construct or chimeric polypeptide in a manner securing the biological processing of the nucleic acid or polypeptide. If the operative linker is a direct link, the two nucleic acids each encoding either an open reading frame or a fragment of an open reading frame are placed immediately adjacent to each other and thereby also in frame. If the operative linker is mediated by a spacer region, a series of nucleotides are inserted between the nucleotides encoding the at least one invariant chain and the at least one antigenic peptide, respectively. It is within the scope of the present invention having a spacer region wherein the spacer region merely is a series of nucleotides linking the at least two elements of the present invention in a manner retaining the open reading frames, or the spacer region may encode one or more signals or separate elements as defined herein below.
In a preferred embodiment the invention comprises an operative linker, wherein the operative linker is a spacer region. In a more preferred embodiment the invention comprises a spacer region encoding at least one helper epitope for class Il MHC molecules. An example of a helper epitope is an immunogenic determinant such as Diphtheria toxin. Especially Diphtheria toxin B fragment COOH-terminal region has been shown to be immunogenic in mice. Furthermore, HSP70, in part or in whole, as well as other immunogenic peptides, such as influenza viral or immunogenic sequences or peptides with an anchoring motif to HLA class I and class Il molecules, also may be encoded in the spacer region of the nucleic acid construct.
In another preferred embodiment the spacer region of the nucleic acid construct encodes at least one protease cleavage site. Cleavage sites of lysosomal proteases such as cathepsins, aspartate proteases and zinc proteases as well as other intracellular proteases fall within the scope of the present invention.
In yet a preferred embodiment the operative linker of the nucleic acid construct may comprise at least one siRNA or miRNA encoding sequence. siRNAs (small interfering RNAs) and miRNAs (microRNAs) target endogenous RNAs, in a sequence-specific manner, for degradation. An siRNA or miRNA encoded within the nucleic acid construct of the present invention may thus be chosen to target an undesirable gene product.
In a more preferred embodiment the operative linker comprises at least one polylinker or multiple cloning site (MCS). Polylinkers and MCS's are series of nucleotides comprising restriction enzyme recognition sequences, i.e. sites where a restriction enzyme cut the DNA in blunt or staggered manner facilitating the subcloning of other fragments / sequences of DNA into the nucleic acid construct. The recognition sequences of the polylinkers / MCS's are typically unique meaning that they are not found elsewhere on the nucleic acid construct. The operative linker may furthermore comprise one or more stop or termination codons that signal the release of the nascent polypeptide from the ribosome. The operative linker may also comprise at least one IRES (Internal Ribosomal Entry Site) and / or at least one promoter. An IRES is a nucleotide sequence that allows for translation initiation in the middle of a messenger RNA (mRNA) sequence as part of the greater process of protein synthesis. A promoter is a DNA sequence that enables a gene to be transcribed. The promoter is recognized by RNA polymerase, which then initiates transcription, see in the below. The promoter may be single or bidirectional.
In a very preferred embodiment the operative linker spanning the region between the invariant chain and the at least one antigen is an operative linker comprising at least one polylinker, and at least one promoter, and optionally also at least one IRES. These elements may be placed in any order. In a further preferred embodiment, the STOP codon of the invariant chain has been deleted, and the polylinker has been cloned into the vector in a manner conserving the open reading frame allowing for in frame reading of the at least one antigen that is inserted into the polylinker. This has the advantage of facilitating subcloning of multiple antigens into the same construct in one step or in multiple cloning steps and allowing for the simultaneous expression of multiple antigens in the same frame as the invariant chain. A STOP codon may be inserted after the polylinker for translation termination. This embodiemtn may be combined with any of the above helper epitopes, mi/siRNAs or any of the other elements herein described.
An embodiment of the present invention relates to the placement of the operative linker in relations to the at least one invariant chain and the at least one antigenic protein or peptide or fragment of said protein or peptide, wherein the at antigenic peptide encoding sequences are placed: within the invariant chain sequence, at the front end of the invariant chain sequence, at the terminal part of the invariant chain sequence. This is done in a manner ensuring the readability of the open reading frame of the construct, so that the antigenic peptide is: preceded, surrounded or rounded off by, at least one operative linker.
A preferred embodiment of the present invention further relates to the placement of the operative linker in relations to the at least one invariant chain and the at least one antigenic protein or peptide or fragment of said protein or peptide, wherein the at least one antigenic peptide encoding sequence preferably is placed at the terminal part of the invariant chain and an operative linker is inserted herein between. The terminal part being the first or last residue of the invariant chain or fragment hereof. Combinations
It is within the scope of the present invention that the nucleic acid construct encodes a plurality of elements. The elements being the at least one invariant chain and the at least one antigenic protein or peptide or fragment of said protein or peptide. It therefore falls within the scope of the present invention to have a plurality of invariant chains each of these being operatively linked to each other and to a plurality of antigenic proteins or peptides or fragments of antigenic proteins or peptides, wherein these also are operatively linked. The elements of the nucleic acid construct must thus be operatively linked to each other. Several series of invariant chains each operatively linked to one antigenic protein or peptide or fragment of said protein or peptide, each of these series being operatively linked to each other are encompassed within the present invention.
Advantages and very important aspects of the present invention relate to the fact that any type of immune response e.g. T cell mediated and antibody mediated responses, can be initiated, both with epitopes known to be weak antigens, with polypeptides of unknown antigenic properties, and with multiple epitopes/antigens simultaneously.
It is therefore also within the scope of the present invention that a preferred embodiment is a nucleic acid construct encoding at least one invariant chain operatively linked to a plurality of antigenic proteins or peptides or fragment of proteins or peptides, such as two, three, four, five, six, eight, ten, twelve or more antigenic proteins or peptides or fragment of proteins or peptides. The nucleic acid construct may comprise additional elements. These include but are not limited to: internal ribosomal entry sites (IRES), genes encoding proteins related to antigen presentation such as LAMP, calreticulin and Hsp70, genes encoding proteins that are related to intracellular spreading such as VP22, HIV Tat, Cx43 or other connexins and intercellular gap-junction constituents, genes encoding natural killer cell (NK-cell) activation molecules such as H60 and cytokines, chicken ovalbumin, or any T-helper cell epitope.
In a preferred embodiment of the present invention the nucleic acid construct comprises at least one gene encoding a protein related to antigen presentation such as LAMP, LIMP, calreticulin or Hsp70. In yet a preferred embodiment of the present invention the nucleic acid construct comprises at least one gene encoding a protein related to intracellular spreading such as VP22, Cx43, HlV Tat, other connexins or intercellular gap-junction constituents.
Promoter
The term promoter will be used here to refer to a group of transcriptional control modules that are clustered around the initiation site for RNA polymerase II. Much of the thinking about how promoters are organized derives from analyses of several viral promoters, including those for the HSV thymidine kinase (tk) and SV40 early transcription units. These studies, augmented by more recent work, have shown that promoters are composed of discrete functional modules, each consisting of approximately 7-20 bp of DNA, and containing one or more recognition sites for transcriptional activator proteins. At least one module in each promoter functions to position the start site for RNA synthesis. The best known example of this is the TATA box, but in some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV 40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation.
Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well. The spacing between elements is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the tk promoter, the spacing between elements can be increased to 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription. Any promoter that can direct transcription initiation of the sequences encoded by the nucleic acid construct may be used in the invention. An aspect of the present invention comprises the nucleic acid construct wherein the at least one operatively linked invariant chain and antigenic protein or peptide encoding sequence is preceded by a promoter enabling expression of the construct.
It is a further aspect that the promoter is selected from the group of constitutive promoters, inducible promoters, organism specific promoters, tissue specific promoters and cell type specific promoters.
Examples of promoters include, but are not limited to: constitutive promoters such as: simian virus 40 (SV40) early promoter, a mouse mammary tumor virus promoter, a human immunodeficiency virus long terminal repeat promoter, a Moloney virus promoter, an avian leukaemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus (RSV) promoter, a human actin promoter, a human myosin promoter, a human haemoglobin promoter, cytomegalovirus (CMV) promoter and a human muscle creatine promoter, inducible promoters such as: a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline promoter (tet-on or tet-off), tissue specific promoters such as: HER-2 promoter and PSA associated promoter and bidirectional promoters, that are capable of initiating transcription in either direction from the promoter.
Advantages of using an inducible promoter includes the option of providing a "dormant" vaccine that can be activated at will. This may be of use if the vaccination preferably only is induced locally vs. systemically within a body (e.g. in cases involving cancer), or the vaccine is detrimental to the health of the recipient at the time of vaccination.
In a preferred embodiment the nucleic acid construct comprises a promoter selected from the group of: CMV promoter, SV40 promoter and RSV promoter.
Delivery vehicle
An aspect of the present invention comprises the nucleic acid construct as described in any of the above, comprised within a delivery vehicle. A delivery vehicle is an entity whereby a nucleotide sequence or polypeptide or both can be transported from at least one media to another. Delivery vehicles are generally used for expression of the sequences encoded within the nucleic acid construct and/or for the intracellular delivery of the construct or the polypeptide encoded therein. It is within the scope of the present invention that the delivery vehicle is a vehicle selected from the group of: RNA based vehicles, DNA based vehicles/ vectors, lipid based vehicles, virally based vehicles and cell based vehicles. Examples of such delivery vehicles include, but are not limited to: biodegradable polymer microspheres, lipid based formulations such as liposome carriers, coating the construct onto colloidal gold particles, lipopolysaccharides, polypeptides, polysaccharides, and pegylation of viral vehicles.
A preferred embodiment of the present invention regards delivery of the nucleic acid construct as naked DNA by mechanical or electrical techniques. Especially the coating of the nucleic acid construct upon gold particles is a favoured embodiment. The delivery of the nucleic acid construct upon gold particles is done by ballistic transfer using particle bombardment equipment such as a gene gun.
A more preferred embodiment of the present invention comprises a virus as a delivery vehicle, where the virus is selected from the non-exhaustive group of: adenoviruses, retroviruses, Antiviruses, adeno-associated viruses, herpesviruses, vaccinia viruses, foamy viruses, cytomegaloviruses, Semliki forest virus, poxviruses, RNA virus vector and DNA virus vector. Such viral vectors are well known in the art.
Viral vectors are often made up of two components, a modified viral genome and a coat structure surrounding it, although sometimes viral vectors are introduced in naked form or coated with proteins other than viral proteins. Most current vectors have coat structures similar to a wild-type virus. This structure packages and protects the viral nucleic acid and provides the means to bind and enter target cells.
Preferably, viral vectors are modified from wild-type viral genomes to disable the growth of the virus in a target cell while enabling the virus to grow in a host cell (e.g. such as a packaging or helper cell) used to prepare infectious particles. Vector nucleic acids generally essential cis-acting viral sequences for replication and packaging in a helper line and expression control sequences for regulating the expression of a polynucleotide being delivered to a target cell. Other viral functions are expressed in trans in specific packaging or helper cell lines as known in the art. Adenovirus
In a more preferred embodiment the vehicle comprising the nucleic acid construct as described herein is an adenovirus. The adenoviral genome consists of a linear double-stranded DNA molecule of approximately 36 kb carrying more than about thirty genes necessary to complete the viral replication cycle. The early genes are divided into 4 regions (E 1 to E4) that are essential for viral replication with the exception of the E3 region, which is believed to modulate the anti-viral host immune response. The E1 region (EIA and EIB) encodes proteins responsible for the regulation of transcription of the viral genome. Expression of the E2 region genes (E2A and E2B) leads to the synthesis of the polypeptides needed for viral replication. The proteins encoded by the E3 region prevent cytolysis by cytotoxic T cells and tumor necrosis factor. The proteins encoded by the E4 region are involved in DNA replication, late gene expression and splicing and host cell shut off. The late genes generally encode structural proteins contributing to the viral capsid. In addition, the adenoviral genome carries at cis-acting 5' and 3' ITRs (Inverted
Terminal Repeat) and packaging sequences essential for DNA replication. The ITRs harbor origins of DNA replication while the encapsidation region is required for the packaging of adenoviral DNA into infectious particles (see for example US 2004/0157307).
In the most preferred embodiment of the present invention the vehicle comprising the nucleic acid construct as described herein is a replication defective adenovirus or a conditionally replication deficient adenovirus. Adenoviral vectors can be engineered to be conditionally replicative (CRAd vectors) in order to replicate selectively in specific cells (e.g., such as proliferative cells). In another aspect, an adenoviral vector is replication-defective for the E1 function (e.g., by total or partial deletion or mutagenesis of E1). The adenoviral backbone of the vector may comprise additional modifications (deletions, insertions or mutations in one or more viral genes). An example of an E2 modification is illustrated by the thermosensitive mutation localized on the DBP (DNA Binding Protein) encoding gene. The adenoviral sequence may also be deleted of all or part of the E4 region. Additional deletions within the non-essential E3 region may allow the size of the polynucleotide being delivered to be increased. However, it may be advantageous to retain all or part of the E3 sequences coding for polypeptides (e.g., such as gp19k) allowing the virus to escape the immune system or inflammatory reactions. Second generation vectors retaining the ITRs and packaging sequences and comprising substantial genetic modifications to abolish the residual synthesis of the viral antigens also may be used in order to improve long-term expression of the expressed gene in the transduced cells. The nucleic acid construct being introduced into the cell may be inserted in any location of the viral genome, with the exception of the cis-acting sequences (see for example US 2004/0157307).
Adenoviruses can be derived from any human or animal source, in particular canine, avian, bovine, murine, ovine, feline, porcine or simian sources or alternatively, may be a hybrid virus. Any serotype can be employed. However, the human adenoviruses are preferred and such viruses are available, for example, from the ATCC (American Type Culture Collection).
A preferred embodiment of the present invention comprises an adenovirus such as: Ovine adenovirus, Canine adenovirus type II, Modified vaccinia Ankara (MVA) or MVA-BN.
Adenoviral particles or empty adenoviral capsids also can be used to transfer nucleic acid constructs or nucleic acid based delivery vectors by a virus-mediated co-internalization process. This process can be accomplished in the presence of cationic agent(s) such as polycarbenes or lipid vesicles comprising one or more lipid layers.
Adenoviral particles may be prepared and propagated according to any conventional technique in the field of the art using a complementation cell line or a helper virus, which supplies in trans the missing viral genes necessary for viral replication. The adenoviral particles can be recovered from the culture supernatant but also from the cells after lysis and optionally further purified according to standard techniques (e.g. chromatography and ultracentrifugation).
Cell-type specific targeting may be achieved with vectors derived from adenoviruses having a broad host range by the modification of viral surface proteins. For example, the specificity of infection of adenoviruses is determined by the attachment to cellular receptors present at the surface of permissive cells. In this regard, the fiber and penton present at the surface of the adenoviral capsid play a critical role in cellular attachment. Thus, cell targeting of adenoviruses can be carried out by genetic modification of the viral gene encoding fiber and/or penton, to generate modified fiber and/or penton capable of specific interaction with unique cell surface receptors.
An aspect of the present invention relates to an adenoviral vector comprising a nucleotide construct encoding at least one antigen and at least one protein or peptide or fragment of a protein or peptide which stimulates an MHC-I response.
A further aspect of the present invention relates to an adenoviral vector, wherein the nucleotide construct encodes at least one protein or peptide or fragment of a protein or peptide which stimulates an MHC-II response.
Preferably, the adenoviral vector comprises sequences, wherein the at least one antigen is operatively linked to the at least one MHC response stimulating protein or peptide or fragment of an MHC response stimulating protein or peptide. The MHC stimulating protein or peptide or fragment of protein or peptide is preferably an MHC associated protein or peptide. Such an MHC associated peptide can be but is not limited being selected from the group of: ER localizing peptide, Golgi localizing peptide, endosomal peptide loading compartment localizing peptide, lysosomal, MIIC, CIIV, melanosomes, secretory granules, Birbeck granules.
More preferably the adenoviral vector comprises an endosomal peptide loading compartment localizing peptide. Such an endosomal peptide loading compartment localizing peptide can be, but is not limited to being, selected from the group of: sorting signal peptides, LAMP, LIMP and invariant chain.
Most preferably the adenoviral vector comprises at least one MHC response stimulating protein or peptide or fragment of protein or peptide and said MHC response stimulating protein or peptide or fragment of protein or peptide is invariant chain.
The adenoviral vector may furthermore comprise proteins that assist in the spreading of the virus or the construct comprised therein. Such proteins include connexins, gap-junction related proteins and pore-forming proteins. A preferred embodiment of the present invention comprises an adenoviral vector encoding or otherwise comprising any one or more of the following proteins related to intercellular spreading: VP22, Cx43 and HIV Tat.
Recombinant cell
An aspect of the present invention relates to a cell comprising the nucleic acid construct as defined in any of the above. Such a recombinant cell can be used a tool for in vitro research, as a delivery vehicle for the nucleic acid construct or as part of a gene therapy regime. The nucleic acid construct and nucleic acid based vectors according to the invention can be introduced into cells by techniques well known in the art and which include microinjection of DNA into the nucleus of a cell, transfection, electroporation, lipofection/liposome fusion and particle bombardment. Suitable cells include autologous and non-autologous cells, and may include xenogenic cells.
In a preferred embodiment the nucleic acid construct of the present invention is comprised within an antigen presenting cell (APC). Any cell that presents antigens on its surface in association with an MHC molecule is considered an antigen presenting cell. Such cells include but are not limited to macrophages, dendritic cells, B cells, hybrid APCs1 and foster APCs. Methods of making hybrid APCs are well known in the art.
In a more preferred embodiment the APC is a professional antigen presenting cell and most preferably the APC is an MHC-I and/or MHC-II expressing cell.
The APC according to any of the above may be a stem cell obtained from a patient. After introducing the nucleic acid construct of the invention, the stem cell may be reintroduced into the patient in an attempt to treat the patient of a medical condition. Preferably, the cell isolated from the patient is a stem cell capable of differentiating into an antigen presenting cell.
It is furthermore included within the scope of the present invention to that the antigen presenting cell comprising the nucleic acid construct of the present invention does not express any co-stimulatory signals and the antigenic protein or peptide or antigenic fragment of said protein or peptide is an auto-antigen. Chimeric proteins and antibodies
An object of the present invention is the chimeric protein encoded by the nucleic acid constructs as described herein above, comprising at least one operatively linked invariant chain and at least one antigenic protein or peptide or fragment of said antigenic protein or peptide. By chimeric protein is understood a genetically engineered protein that is encoded by a nucleotide sequence made by a splicing together of two or more complete or partial genes or a series of (non)random nucleic acids.
An aspect of the present invention relates to an antibody that can recognize the chimeric protein as defined herein above. By the term antibody is understood immunoglobulin molecules and active portions of immunoglobulin molecules. Antibodies are for example intact immunoglobulin molecules or fragments thereof retaining the immunologic activity. Such antibodies can be used for the passive immunization of an animal, or for use in an assay for detecting proteins to which the antibody binds.
Vaccine compositions
An aspect of the present invention relates to a vaccine comprising a nucleic acid sequence encoding at least one invariant chain operatively linked to at least one antigenic protein or peptide or fragment of said antigenic protein or peptide. The vaccine may thus comprise a nucleic acid construct as defined in any of the above. The vaccine may furthermore be used as a medicament.
The vaccine composition according to the invention can be formulated according to known methods such as by the admixture of one or more pharmaceutically acceptable carriers, also known as excipients or stabilizers with the active agent. These excipients may be acceptable for administration to any individual / animal, preferably to vertebrates and more preferably to humans as they are non-toxic to the cell or individual being exposed thereto at the dosages and concentrations employed. Often the physiologically acceptable carrier is an aqueous pH buffered solution. Examples of such excipients, carriers and methods of formulation may be found e.g. in Remington's Pharmaceutical Sciences (Maack Publishing Co, Easton, PA). Examples of physiologically acceptable carriers include but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEEN.TM., polyethylene glycol (PEG), and PLURONICS.TM.
To formulate a pharmaceutically acceptable composition suitable for effective administration, such compositions will according to the invention contain an effective amount of the nucleic acid construct, the nucleic acid construct comprised within a delivery vehicle or the chimeric protein encoded within the nucleic acid construct as described herein. Often, if vaccinating with protein or polypeptides as encoded by the nucleic acid construct of the present invention, a carrier will be used as a scaffold by coupling the proteins or peptides hereto and thus aiding in the induction of an immune response. The carrier protein may be any conventional carrier including any protein suitable for presenting immunogenic determinants. Suitable carriers are typically large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplets or liposomes), and inactive virus particles. Such carriers are well known to those of ordinary skill in the art. Additionally, these carriers may function as immunostimulating agents ("adjuvants"). Immunisation of the animal may be carried out with adjuvants and/or pharmaceutical carriers. Conventional carrier proteins include, but are not limited to, keyhole limpet hemocyanin, serum proteins such as transferrin, bovine serum albumin, or human serum albumin, an ovalbumin, immunoglobulins, or hormones, such as insulin. The carrier may be present together with an adjuvant or independently here from.
In the following vaccine compositions are meant to encompass compositions useful for prophylactic and therapeutic use, including stimulating an immune response in a patient. It is further contemplated that the vaccine composition of the invention does not induce any systemic or local toxicity reactions or any other side effects.
In a preferred embodiment the nucleic acid construct of the vaccine is packaged. Packaging means for the nucleic acid construct include means selected from, but not limited to the group of: RNA based or DNA based vectors, lipid based carriers, viral expression vectors, viral delivery vectors, cell based vehicles, coating of colloidal gold particles, biodegradable polymer microspheres. Any of the previously mentioned delivery means may thus be used for packing purposes for use in a vaccine composition.
In a more preferred embodiment the packaging means of the nucleic acid construct for the vaccine is a viral expression vector selected from, but not limited to the group of: adenovirus, retrovirus, lentivirus, adeno-associated virus, herpes virus, vaccinia virus and DNA virus vector.
In an even more preferred embodiment the nucleic acid construct of the vaccine is packaged into an adenoviral vector. In the most preferred embodiment the nucleic acids construct as described in any of the herein above of the vaccine is packaged in a replication deficient or conditionally replication deficient adenoviral vector. Adenoviral vectors are described in detail in the above.
An aspect of the invention relates to a vaccine comprising at least two vectors. This encompasses that any one or two different nucleic acid constructs as described may be packed into at least two vectors, these vectors being of a type as described in any of the above. The invention furthermore relates to a vaccine comprising three, four, five or six vectors. Again, these vectors may differ from one another or not, and may carry identical or different nucleic acid constructs as described herein above. A further aspect of the present invention relates to a vaccine comprising at least one chimeric protein as encoded by any of the nucleic acid constructs described herein. When a chimeric protein or polypeptide is to be used as an immunogen, it may be produced by expression of any one or more of the DNA constructs described above in a recombinant cell or it may be prepared by chemical synthesis by methods known in the art. As described in the above, such chimeric proteins and / or peptides may be coupled to carriers to increase the immunologic response to the proteins / peptides and may be administered with or without an adjuvant and/or excipient.
Adjuvant
Adjuvants may be included in the vaccine composition to enhance the specific immune response. Thus, it is particular important to identify an adjuvant that when combined with the antigen(s) / nucleic acid constructs and / or delivery vehicles such as adenoviral vehicles (any of which may also be referred to as immunogenic determinant), results in a vaccine composition capable of inducing a strong specific immunological response. The immunogenic determinant may also be mixed with two or more different adjuvants prior to immunisation. Vaccine compositions are also referred to as immunogenic compositions in the present text.
A large number of adjuvants have been described and used for the generation of antibodies in laboratory animals, such as mouse, rats and rabbits. In such setting the tolerance of side effect is rather high as the main aim is to obtain a strong antibody response. For use and for approval for use in pharmaceuticals, and especially for use in humans it is required that the components of the vaccine composition, including the adjuvant, are well characterized. It is further required that the composition has minimal risk of any adverse reaction, such as granuloma, abscesses or fever.
An embodiment of the present invention relates to a vaccine comprising an adjuvant. In a preferred embodiment the vaccine composition is suitable for administration to a mammal, and most preferably to a human subject, therefore the preferred adjuvant is suitable for administration to a mammal and most preferably is suitable for administration to a human subject.
The choice of adjuvant may further be selected by its ability to stimulate the type of immune response desired, B-cell or/and T-cell activation and the vaccine composition may be formulated to optimize distribution and presentation to the relevant lymphatic tissues.
Adjuvants pertaining to the present invention may be grouped according to their origin, be it mineral, bacterial, plant, synthetic, or host product. The first group under this classification is the mineral adjuvants, such as aluminum compounds. Antigens precipitated with aluminum salts or antigens mixed with or adsorbed to performed aluminum compounds have been used extensively to augment immune responses in animals and humans. Aluminium particles have been demonstrated in regional lymph nodes of rabbits seven days following immunization, and it may be that another significant function is to direct antigen to T cell containing areas in the nodes themselves. Adjuvant potency has been shown to correlate with intimation of the draining lymph nodes. While many studies have confirmed that antigens administered with aluminium salts lead to increased humoral immunity, cell mediated immunity appears to be only slightly increased, as measured by delayed- type hypersensitivity. Aluminium hydroxide has also been described as activating the complement pathway. This mechanism may play a role in the local inflammatory response as well as immunoglobulin production and B cell memory. Furthermore, aluminum hydroxide can protect the antigen from rapid catabolism. Primarily because of their excellent record of safety, aluminum compounds are presently the only adjuvants used in humans.
Another large group of adjuvants is those of bacterial origin. Adjuvants with bacterial origins can be purified and synthesized (e.g. muramyl dipeptides, lipid A) and host mediators have been cloned (Interleukin 1 and 2). The last decade has brought significant progress in the chemical purification of several adjuvants of active components of bacterial origin: Bordetella pertussis, Mycobacterium tuberculosis, lipopoly-saccharide, Freund's Complete Adjuvant (FCA) and Freund's Incomplete Adjuvant (Difco Laboratories, Detroit, Mich.) and Merck Adjuvant 65 (Merck and Company, Inc., Rahway, NJ. ). Additionally suitable adjuvants in accordance with the present invention are e.g. Titermax Classical adjuvant (SIGMA-ALDRICH), ISCOMS, Quil A, ALUN, see US 58767 and 5,554,372, Lipid A derivatives, choleratoxin derivatives, HSP derivatives, LPS derivatives, synthetic peptide matrixes, GMDP, and other as well as combined with immunostimulants (US 5,876,735). B. pertussis is of interest as an adjuvant in the context of the present invention due to its ability to modulate cell-mediated immunity through action on T- lymphocyte populations. For lipopolysaccharide and Freund's Complete Adjuvant, adjuvant active moieties have been identified and synthesized which permit study of structure-function relationships. These are also considered for inclusion in immunogenic compositions according to the present invention.
Lipopolysaccharide and its various derivatives, including lipid A, have been found to be powerful adjuvants in combination with liposomes or other lipid emulsions. It is not yet certain whether derivatives with sufficiently low toxicity for general use in humans can be produced. Freund's Complete Adjuvant is the standard in most experimental studies. Mineral oil may be added to the immunogenic composition in order to protect the antigen from rapid catabolism.
Many other types of materials can be used as adjuvants in immunogenic compositions according to the present invention. They include plant products such as saponin, animal products such as chitin and numerous synthetic chemicals.
Adjuvants according to the present invention can also been categorized by their proposed mechanisms of action. This type of classification is necessarily somewhat arbitrary because most adjuvants appear to function by more than one mechanism. Adjuvants may act through antigen localization and delivery, or by direct effects on cells making up the immune system, such as macrophages and lymphocytes. Another mechanism by which adjuvants according to the invention enhance the immune response is by creation of an antigen depot. This appears to contribute to the adjuvant activity of aluminum compounds, oil emulsions, liposomes, and synthetic polymers. The adjuvant activity of lipopolysaccharides and muramyl dipeptides appears to be mainly mediated through activation of the macrophage, whereas B. pertussis affects both macrophages and lymphocytes. Further examples of adjuvants that may be useful when incorporated into immunogenic compositions according to the present invention are described in US 5,554,372.
Adjuvants useful in both prophylactic and therapeutic vaccines according to the present invention may thus be mineral salts, such as aluminium hydroxide and aluminium or calcium phosphates gels, oil emulsions and surfactant based formulations such as MF59 (microfluidized detergent stabilized oil in water emulsion), QS21 (purified saponin), AS02 (SBAS2, oil-in-water emulsion + monophosphoryl lipid A (MPL) + QS21), Montanide ISA 51 and ISA-720 (stabilized water in oil emulsion), Adjuvant 65 (containing peanut oil, mannide monooleate and aluminum monostearate), RIBI ImmunoChem Research Inc., Hamilton, Utah), particulate adjuvants, such as virosomes (unilamellar liposomal cehicles incorporating influenza haemagglutinin), AS04 (Al salt with MPL), ISCOMS (structured complex of saponins and lipids (such as cholesterol), polyactide co- glycolide (PLG), microbial derivatives (natural and synthetic) such as monophosphoryl lipid A (MPL), Detox (MPL + M. Phlei cell wall skeleton), AGP (RC- 529 (synthetic acylated monosaccharide)), DC_chol (lipoidal immunostimulators able to self organise into liposomes), OM-174 (lipid A derivative), CpG motifs (synthetic oligonucleotides containing immunostimulatory CpG motifs), modified bacterial toxins, LT and CT, with non-toxic adjuvant effects, Endogenous human immunomodulators, e.g., hGM-CSF or hlL-12 or lmmudaptin (C3d tandem array), inert vehicles such as gold particles.
Additional examples of adjuvants comprise: Immunostimulatory oil emulsions (for example, water-in-oil, oil-in-water, water-in-oil-in-water such as e.g. Freund's incomplete adjuvant such as Montainde®, Specol, mineral salts such e.g. as AI(OH)3, AIPO4, microbial products, Saponins such as Qual A, synthetic products, as well as adjuvant formulations, and immune stimulatory complexes (ISCOMs) and cytokines, heat-inactivated bacteria/components, nanobeads, LPS, LTA. A list of other commonly used adjuvants is disclosed on pages 6-8 in WO 03089471 , the list being hereby incorporated by reference.
Immunogenic compositions according to the invention may also contain diluents such as buffers, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, sucrose or dextrins, chelating agents such as EDTA, glutathione and other stabilizers and excipients. Neutral buffered saline or saline mixed with non-specific serum albumin are exemplary appropriate diluents.
Adjuvants are generally included in the immunogenic compositions in an amount according to the instructions of the manufacturer.
Administration
Vaccine compositions according to the invention may be administered to an individual in therapeutically effective amounts. The effective amount may vary according to a variety of factors such as the individual's condition, weight, sex and age. Other factors include the mode of administration.
The pharmaceutical or veterinary compositions may be provided to the individual by a variety of routes such as subcutaneous, topical, oral and intramuscular. Administration of pharmaceutical compositions is accomplished orally or parenterally. Methods of parenteral delivery include topical, intra-arterial (directly to the tissue), intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, or intranasal administration. The present invention also has the objective of providing suitable topical, oral, systemic and parenteral pharmaceutical formulations for use in the methods of prophylaxis and treatment with the vaccine composition.
For example, the vaccine compositions can be administered in such oral dosage forms as tablets, capsules (each including timed release and sustained release formulations), pills, powders, granules, elixirs, tinctures, solutions, suspensions, syrups and emulsions, or by injection. Likewise, they may also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous, topical with or without occlusion, or intramuscular form, all using forms well known to those of ordinary skill in the pharmaceutical arts. An effective but non-toxic amount of the vaccine, comprising any of the herein described compounds can be employed as a prophylactic or therapeutic agent. Also any and all conventional dosage forms that are known in the art to be appropriate for formulating injectable immunogenic peptide composition are encompassed, such as lyophilized forms and solutions, suspensions or emulsion forms containing, if required, conventional pharmaceutically acceptable carriers, diluents, preservatives, adjuvants, buffer components, etc.
Preferred modes of administration of the vaccine composition according to the invention include, but are not limited to systemic administration, such as intravenous or subcutaneous administration, intradermal administration, intramuscular administration, intranasal administration, oral administration, rectal administration, vaginal administration, pulmonary administration and generally any form of mucosal administration. Furthermore, it is within the scope of the present invention that the means for any of the administration forms mentioned in the herein are included in the present invention.
A vaccine according to the present invention can be administered once, or any number of times such as two, three, four or five times. Administering the vaccine more than once has the effect of boosting the resulting immune response. The vaccine can further be boosted by administering the vaccine in a form or body part different from the previous administration. The booster shot is either a homologous or a heterologous booster shot. A homologous booster shot is a where the first and subsequent vaccinations comprise the same constructs and more specifically the same delivery vehicle especially the same viral vector. A heterologous booster shot is where identical constructs are comprised within different viral vectors. This is especially of interest when employing adenoviral delivery as the human body raises an immune response against a given adenovirus if it has previously been exposed thereto. A preferred embodiment of the present invention therefore relates to the pegylation of the adenoviral vector providing the option of boosting with the same (homologous) adenoviral vector. An alternative and preferred embodiment relates to the sequential boosting of a vaccine with different adenoviral vectors comprising the same constructs.
A preferred administration form of the vaccine according to the present invention is administering the vaccine to the body area, inside or out, most likely to be the receptacle of a given infection. The receptacle of infection is the body area that the infection is received by, e.g. regarding influenza, the receptacle of infection is the lungs.
The vaccine of the present invention can be administered to any organism to which it may be beneficial, especially any animal such as a vertebrate animal. It falls within the scope of the present invention that the means and modes of administration of the vaccine are adapted to the recipient. A preferred recipient of the vaccine is a mammal and the mammal is in a more preferred embodiment of the present invention selected from the group of: cows, pigs, horses, sheep, goats, llamas, mice, rats, monkeys, dogs, cats and humans. In the most preferred embodiment the mammal is a human.
An embodiment of the present invention includes a vaccine composition further comprising a second active ingredient. The second active ingredient is selected from, but not limited the group of antibiotics, chemotherapeutics, anti-allergenics, cytokines, complement factors and co-stimulatory molecules of the immune system.
Another embodiment of the present invention comprises a kit of parts, wherein the kit includes at least one vaccine composition according to any of the above, a means for administering said vaccine and the instruction on how to do so. It is within the scope of the present invention to include multiple dosages of the same vaccine or several different vaccines. In a preferred embodiment the kit of parts further comprises a second active ingredient.
The present invention further comprises a method for inducing an immune response in an animal, comprising administering to the animal a vaccine according to any of the above. The immune response is, but is not limited to any of the following types of responses: an MHC-I dependent response, an MHC-I and/ or MHC-II dependent response, a T-cell dependent response, a CD4 T-cell dependent response, a CD4+ T cell independent response, a CD8+ T-cell dependent response and a B cell dependent immune response. Another method falling within the scope of the present invention is the method of providing at least one vaccine according to any of the above and administering said at least one vaccine to a subject at least once for treatment or prophylaxis of an animal. The invention also encompasses the nucleic acid construct according the herein described for the preparation of a composition for the production of a vaccine. Said vaccine can be but is not limited being used for genetic immunization of an animal, or to treat a clinical condition in an individual in need thereof.
Detailed description of the drawings Figure 1 : Schematic drawing of inserts in the adenovirus vector. A) Schematic drawing of Ad-GP expression cassette, B) Schematic drawing of Ad-IiGP expression cassette. Shown is also the situation of various LCMV GP epitopes. Ad-GP: adenoviral-glycoprotein, CMV: Cytomegalovirus promoter, Ii : Invariant chain, LCMV GP 1-498 : Glycoprotein from lymfocytic choriomeningitis virus, STOP : Stop codon, PoIyA : SV40 polyadenylation signal
Figure 2: CD8+ and CD4+ T-cell responses to adenovirus encoded epitopes. C57BL/6 mice were vaccinated with 2x107 infectious unit (IFU) of Ad-GP or Ad-IiGP in the right hind footpad. On the indicated days post vaccination the number of epitope specific CD8+ or CD4+ T cells were determined by intracellular staining for peptide-induced IFN-γ of spleen cells. Bars represent Average (Avg) ± standard deviation (SD) of 3-5 animals.
Figure 3: CD8+ and CD4+ T-cell responses to adenovirus encoded epitopes in F1 hybrid mice. C57BL/6 x BALB/c (H-2bxd) F1 mice were vaccinated with 2x107 IFU of Ad-GP or Ad-IiGP in the right hind footpad. On day 21 post vaccination the number of epitope specific CD8+ or CD4+ T cells were determined by intracellular staining for peptide-induced IFN-γ of spleen cells. Bars represent Avg ± SD of 4-5 animals Figure 4: Ad-IiGP exerts CD8+ T-cell stimulatory effects that are independent of CD4+ T-cells. MHC-II+ or C57BL/6 mice were vaccinated with 2x107 IFU of Ad-GP or Ad-IiGP in the right hind footpad. On day 21 or 90 post vaccination the number of epitope specific CD8+ or CD4+ T cells were determined by intracellular staining for peptide-induced IFN-γ of spleen cells. Bars represent Avg ± SD of 4 animals. Figure 5: Ad-IiGP confers rapid and superior protection against lethal LCMV infection. C57BL/6 mice were vaccinated with 2x107 IFU of Ad-GP or Ad-IiGP in the right hind footpad. On the indicated days post vaccination animals were challenged with 20 pfu (plaque forming units) LCMV Arm 53b i.e. (intra cerebral). Mortality was recorded for 14 days. Each group consisted of 5 to 18 animals. ND means no data. Figure 6: Ad-IiGP efficiently protects against high-dose, intravenous LCMV infection. C57BL/6 mice were vaccinated with 2x107 IFU of Ad-GP or Ad-IiGP in the right hind footpad. On day 21 post vaccination animals were challenged with 1x106 pfu (plaque forming units) LCMV Arm clone13 i.v. (intra venous). 8 days after virus challenge organ virus titer was determined. Points represent individual animals. Dashed line represent detection limit of the assay. Figure 7: Ad-IiGP confers superior protection to lethal LCMV variants with mutations in immunodominant epitopes. C57BL/6 mice were vaccinated with 2x107 IFU of Ad- GP, Ad-IiGP or sham infected in the right hind footpad. On day 90 post vaccination animals were challenged with 20 pfu LCMV Arm 53b i.e. carrying mutations in gp33, gp276 or both epitopes. Mortality was recorded for 14 days. For gp33 nil and gp276 nil, each group consisted of 5 animals, with gp33/gp276 double nil, the groups were 10 animals.
Figure 8: Frequencies of CD8+ or CD4+T cells reacting to specific LCMV epitopes after Ad-IiGP vaccination and challenge with LCMV variants with mutations in immunodominant epitopes. Surviving animals from the experiment depicted in figure 7 were analysed for epitope specific CD8+ or CD4+T cells by intracellular staining for peptide-induced IFN-γ of spleen cells. Bars represent Avg ± SD of 3-5 animals.
Figure 9: CD8+ and CD4+ T cell responses to vaccination with naked DNA-IiGP and DNA-GP. C57BL/6 mice were vaccinated with DNA coated onto 1.6-nm gold particles in a concentration of 2 μg DNA/mg gold, and the DNA-gold complex was coated onto plastic tubes such that 0.5 mg gold was delivered to the mouse per shot (1 μg DNA per shot). Mice were immunized at the abdominal skin using a hand-held gene gun device employing compressed helium (400 psi) as the particle motive force. Mice were inoculated four times with an interval of 1 week and then allowed to rest for 1 week before investigation. The number of epitope specific CD8+ or CD4+ T cells was determined by intracellular staining for peptide-induced IFN-γ of spleen cells. Bars represent Avg ± SD of 4-5 animals.
Figure 10: Prophylactic vaccination with Ad-Ii-GP increases tumor rejection. C57BL/6 mice were vaccinated in the right hind foot-pad with 2 x 107 IFU of adenovirus encoding either full-length glycoprotein of LCMV (Ad-GP) or glycoprotein linked to invariant chain (Ad-Ii-GP). Controls were infected with either adenovirus encoding full-length β-galactosidase or live LCMV (103 PFU of LCMV Armstrong 53b). About 3 months later all mice were challenged by subcutaneous injection of 106 B16.F10 melanoma cells expressing the LCMV derived GP33 epitope. Initially a tumor was formed in all animals, but the majority of Ad-Ii-GP and LCMV primed mice eventually rejected the tumor. Each group consisted of 7-10 animals. Figure 11 : Therapeutic vaccination with Ad-Ii-GP increases average life span in tumor carrying mice. C57BL/6 mice were challenged subcutaneously by injection of 106 B16.F10 melanoma cells expressing the LCMV derived GP33 epitope. When tumors were palpable in all mice (day 5 after tumor injection), the animals were vaccinated in the right hind foot-pad using 2 x 107 IFU of adenovirus encoding either full-length glycoprotein of LCMV (Ad-GP) or full-length glycoprotein of LCMV linked to invariant chain (Ad-Ii-GP); controls were vaccinated with either adenovirus encoding full-length β-galactosidase or live LCMV (103 PFU of LCMV Armstrong 53b). Mice were sacrificed when the tumor exceeded 12 mm in length or ulceration was observed. The numbers in bold in the center of the figure represents the mean day of death following the tumor challenge. Each group consisted of 7-10 animals. Figure 12: Survival rate following vaccination with either Ad-Ii-VSVGP or Ad- VSVGP. C57BL/6 mice were vaccinated in the right hind foot-pad with 2 x 107 IFU of adenovirus encoding either full-length glycoprotein of vesicular stomatitis virus (Ad- VSVGP) or full-length glycoprotein of vesicular stomatitis virus linked to invariant chain (Ad-Ii-VSVGP). (A) On the indicated days serum samples were collected and in vitro neutralizing antibody titers were determined in a plaque-reduction assay, dots represent individual animals. (B) On the indicated days vaccinated mice were challenged with 105 PFU of VSV intranasally, and mortality was registered over the next 14 days. Survival of control (unvaccinated) mice has been included for comparison. Each group consisted of 5-10 animals Figure 13: CD8+ and CD4+ T-cell responses to more adenovirus encoded epitopes. C57BL/6 mice (Influenza and OVA) or B6D2 F1 mice (MHV-68 M2 and M3) were vaccinated with 2x107 IFU of the indicated construct in the right hind footpad. On the indicated days the number of epitope specific CD8+ or CD4+ T cells was determined by intracellular staining for peptide-induced IFN-γ of spleen cells, lso is the isotype control which determined the background. Bars represent Avg ± SD of 4- 5 animals.
Figure 14: Efficiency of Ad-Ii-GP constructs compared to Ad-GP-Lamp-1 constructs measured by CD8+ T-cell responses to adenovirus encoded epitopes. C57BL/6 mice were vaccinated with 2x107 IFU of the indicated construct in the right hind footpad. On day 21 the number of epitope specific CD8+ cells was determined by intracellular staining for peptide-induced IFN-γ of spleen cells. Bars represent Avg ± SD of 3 animals. Figure 15: Vectors based on in-frame polylinkers. Ii without stop-codon was amplified by PCR with the sequence for the AsiSI, Swal, Ascl, Pmel, Fsel and a stop site included in the 3' primer and cloned into the pacCMV vector. The resulting vector was numbered 770 (see partial sequence hereof in SEQ ID NO: 7). A corresponding vector termed 768 (see partial sequence hereof in SEQ ID NO: 8) without the Ii sequence incorporated was also constructed. Figure 16: Vectors with IRES2 sites. The vector termed "pacCMV Ii MCS IRES2" (and numbered 1163, see partial sequence hereof in SEQ ID NO: 9) was constructed by cloning the IRES2 from pLP-IRES2-EGFP into the 770 vector by PCR and restriction enzyme digestion. The sequences for l-scel and Srfl were included in the 3' primer. The first ATG site after the IRES sequence initiates expression of a second protein. A corresponding vector termed "pacCMV MCS
IRES2" and numbered 1165 (see partial sequence hereof in SEQ ID NO: 10) without the Ii sequence incorporated was also constructed
Examples The invention will now be further illustrated with reference to the following examples. It will be appreciated that what follows is by way of example only and that modifications in detail may be made while still falling within the scope of the invention. Example 1 :
CD8+ and CD4+ T-cell responses to adenovirus encoded epitopes.
Mice: C57BL/6 (H-2b), C57BL/6 x BALB/c (H-2bxd) F1 hybrids and MHC H-/- mice (B6.129-H2-Ab1tm1GlmN12 (H-2b)) were obtained from Taconic M&B (Ry, Denmark). All mice used were between 7-10 weeks old and housed in a specific germ free facility. All experimental procedures were performed according to local experimental guidelines.
Adenovirus vectors: For construction of E1 and E3 deleted adenovirus-expressing, LCMV derived antigen fused to invariant chain we performed 2-step PCR. First we obtained overlapping PCR products containing the full-length mouse invariant chain and full-length LCMV glycoprotein and these were joined by secondary PCR with invariant chain 5' and glycoprotein 3' primers. Adenovirus expressing full-length GP was amplified in single step PCR. The obtained fragments were cloned into the pacCMV shuttle vector. The obtained plasmid was co-transfected with pJM17 plasmid into HEK293 cells and viral lysates were obtained. These were cloned by plaque purification before sequencing, large-scale production and purification by CsCI gradient centrifugation as described (Becker et al., 1994, Methods Cell Biol. 43 Pt A: 161 -189). Infectivity of adenovirus stocks was determined with the Adeno-X Rapid Titer Kit (Clontech). All unmodified virus stocks had particle/IFU ratios between 46 and 201.
Vaccinations: In all studies, mice to be vaccinated were anaesthetized and injected with 2x107 infectious units in the right hind footpad in a volume of 0.03 ml.
Virus infection: Mice were infected i.e. with 20 pfu of LCMV Armstrong clone 53b in a volume of 0.03 ml or i.v. with 106 pfu of LCMV clone 13 in 0.3 ml. I.e. infection induces a fatal CD8+ T cell-mediated meningitis from which immunocompetent mice succumb on days 7 to 10 p.i. (post infection) (Christensen et al., Scandinavian Journal of Immunology 40:373-382).
Survival study: Mortality was used to evaluate the clinical severity of acute LCMV- induced meningitis. Mice were checked twice daily for a minimum of 2 weeks after i.e. inoculation; deaths occurring less than 3 days after infection were excluded from analysis. Organ virus titers: To determine virus titers in organs, these were first homogenized in PBS to yield 10% (v/w) organ suspensions, and serial 10-fold dilutions were prepared. Each dilution was then plated in duplicates on MC57G cells. Forty-eight hours after infection, infected cell clusters were detected using monoclonal rat anti- LCMV (VL-4) antibody, peroxidase-labeled goat anti-rat antibody and o- phenylendiamin (substrate) (Battegay et al., 1991 , Journal of Virological Methods 33:191-198). The numbers of pfu were counted, and results expressed as pfu/g tissue.
Cell preparations: Single cell suspensions of spleen cells were obtained by pressing the organs through a fine steel mesh.
Abs for flow cytometric: The following monoclonal antibodies (mAbs) were purchased from BD PharMingen (San Diego, CA) as rat anti-mouse antibody: Cy- chrome-conjugated anti-CD8, FITC-conjugated anti-CD44, phycoerythrin (PE)- conjugated anti-IFN-γ and PE-conjugated IgGi isotype standard.
Flow cytometri analysis: Staining of cells for flow cytometry was performed according to standard laboratory procedure (Andersson et al., 1994, Journal of Immunology 152:1237-1245; Andreasen et al., 1999, International Immunology
11 : 1463-1473). For enumeration of LCMV-specific CD8+ T cells, splenocytes were incubated in vitro for 5 h at 37 0C in 5% CO2 with relevant peptide (0.1 μg/ml) in the presence of monensin (3 μM, Sigma Chemicals co., St. Louis, MO) and murine recombinant IL-2 (10 units/well, R&D Systems Europe Ltd, Abingdon, UK). After incubation cells were surface stained, washed, fixed and permeabilized using 0.5% saponin. Cells were then stained with anti-IFN-γ or IgGi isotype control for 20 min at 4 0C. Samples were analyzed using a Becton Dickinson FACSCalibur, and at least 104 mononuclear cells were gated using a combination of low angle and side scatter to exclude dead cells and debris. Data analysis was conducted using Cell Quest Pro (B&D Biosciences).
Replication deficient adenovirus vectors expressing lymphocytic choriomeningitis virus full-length glycoprotein (Ad-GP), or lymphocytic choriomeningitis virus full- length glycoprotein N-terminally linked to murine invariant chain ((Ad-IiGP) for schematic representation of the expression cassette see figure 1 ), were generated through standard methods (Becker et al., Methods Cell Biol. 43 Pt A:161-189). C57BL/6 mice were then vaccinated in the right hind paw with 2x107 infectious units of Ad-GP or Ad-IiGP and mice were sacrificed 5, 7, 11, 14, 21, 28, 90, 180 or 360 days later. The generation of LCMV glycoprotein specific T-cells were then analysed on splenic cells. Evidently (see figure 2), at all time points tested, Ad-IiGP induced numerically superior T-cell responses compared to Ad-GP, and these were accelerated and included both CD4+ and CD8+ T-cell responses. Thus peak numbers of T-cells generated after Ad-IiGP vaccination were obtained at 7-14 days after vaccination depending on the epitope, with responses after Ad-GP peaking at day 21 after vaccination.
Example 2:
CD8+ and CD4+ T-cell responses to adenovirus encoded epitopes in F-i hybrid mice:
As C57BL/6 mice are homozygous with regard to both MHC class I and MHC class Il molecules on all loci, we tested whether Ad-GP and Ad-IiGP could also induce an immune response in C57BL/6 x BALB/c Fi mice that express both the H-2b and H2d haplotypes. These mice resemble an out bred population, but with defined haplotypes. The experiments were performed as above, but testing was limited to day 21 after vaccination. As can be seen from figure 3, Ad-IiGP efficiently induces CD8+ T-cell responses towards a multitude of epitopes while Ad-GP seemed to perform worse than in homozygous C57BL/6 mice.
Example 3:
Ad-IiGP exerts CD8+ T-cell stimulatory effects that are independent of CD4+ T-cells: As a potential mechanism of Ii function in the enhanced stimulation of CD8+ T-cells is the ability to traffic to endosomal and lysosomal compartments and stimulate CD4+ T-cells (Diebold et al., 2001, Gene Ther. 8:487-493) through MHC class II, we performed vaccination of MHC class Il deficient mice. To this effect MHC-I I''" or C57BL/6 mice were vaccinated with 2x107 IFU of Ad-GP or Ad-IiGP in the right hind footpad. On day 21 or 90 post vaccination the number of epitope specific CD8+ or CD4+ T cells were determined by intracellular staining for peptide-induced IFN-γ of spleen cells. As can be seen from figure 4, Ad-IiGP efficiently induces CD8+ T-cell responses directed against several epitopes; in the absence of CD4+ T cell help however, some responses were lower than what is seen in wild type mice. Example 4:
Ad-IiGP confers rapid, superior and sustained protection against lethal LCMV infection: As we observed an accelerated response to Ad-IiGP compared to Ad-GP we investigated the ability of vaccination to confer protection both at 21 days post vaccination (peak of Ad-GP) and at 3, 5, 7, 14, 60, 90, 180 and 360 days post vaccination (Figure 5). Remarkably, we found that Ad-IiGP vaccinated animals vaccinated as little as 3 days previously were protected against intracerebral LCMV infection. Protection conferred by Ad-GP was only partial at 14 and 21 days post infection and no protection were seen at day 60 or later. Furthermore, the Ad-IiGP conferred protection was sustained for 360 days, at which point Ad-GP no longer protected against intracerebral LCMV infection.
Example 5:
Ad-IiGP efficiently protects against high-dose, intravenous LCMV infection: Since we found that Ad-IiGP protected mice against an acute localised infection, we wanted to investigate whether the same held true for a high-dose systemic infection.
Accordingly, mice were vaccinated with Ad-GP, Ad-IiGP or sham (PBS), and challenged 21 days later by intravenous injection of 106 plaque-forming units of the fast replicating LCMV clone 13 strain. 5 days later animals were sacrificed and infectious titers in the lungs were determined. Although Ad-GP conferred significant protection upon vaccinated animals, Ad-IiGP was superior and reduced titers to a level at or below the detection limit of the assay (Figure 6).
Example 6: Comparative analyses of novel constructs with respect to kinetics, magnitude of response, long-term immunity and virus dose needed for immunity.
All constructs described below are comparatively analysed among each other and to Ad-GP and Ad-IiGP with respect to the kinetics and magnitude of immune response e.g. as described in Examples 1 , 2 and 3, long-term immunity e.g. as described in Examples 1 , 4 and 5, and virus dose needed for immunity.
Novel constructs based upon alterations to the Ii fusions. In each construct adenovirus-encoded GP is fused N-terminally to: 1. the lysosomal targeting sequence of Ii 2. the CLIP sequence of Ii
3. the KEY sequence of Ii
4. the CLIP sequence and the sequence N-terminally adjacent to the CLIP sequence 5. the CLIP sequence and the sequence C-terminally adjacent to the CLIP sequence
6. the sequence N-terminally adjacent to the CLIP sequence
7. the sequence C-terminally adjacent to the CLIP sequence
Alterations of antigen presentation context. In each construct adenovirus-encoded GP is fused:
1. C-terminally to LAMP
2. N-terminally to the N-domain of calreticulin
3. C-terminally to the N-domain of calreticulin 4. C-terminally to Hsp70.
5. N-terminally to Ii and C-terminally to the N-domain of calreticulin (AdliGPCrt)
6. N-terminally to Ii and C-terminally to Hsp70 (AdliGPHsp70)
All of these constructs are furthermore used as the starting point for a series of constructs in which the GP-fusions are followed by an internal ribosomal entry site (IRES) and a gene encoding VP22, HIV tat or Cx43.
Alterations with regard to intercellular spreading. In each construct adenovirus- encoded GP is fused:
1. N-terminally to herpes simplex virus encoded VP22 2. N-terminally to HIV encoded tat
3. N-terminally to connexin 43 (Cx43)
4. N-terminally to other connexins and intercellular gap-junctions constituents. Furthermore constructs are prepared where adenovirus-encoded GP is followed by an internal ribosomal entry site (IRES) and a gene encoding VP22, HIV tat or Cx43 or other connexins and intercellular gap-junctions constituents.
All of the above constructs are furthermore altered in any of the following ways: 1. All of the above constructs followed by an IRES site and a gene encoding an NK-cell (natural killer cell) activation molecule, for example H60. This alteration gives enhanced delivery of co-stimulatory signals and cytokine help. 2. All of the above constructs involving IRES sites, where the downstream gene is placed under control of a separate promoter. 3. All of the above constructs involving IRES sites, where the downstream gene is instead encoded on a separate vector.
4. All of the above constructs placed under inducible promoter systems.
5. All of the above constructs placed under cell type specific and/or inducible promoter systems. 6. All of the above constructs where the GP antigenic sequence is replaced with a sequence encoding any of the following antigens, several of which comprise multiple antigens, see examples of specific antigens in figures 12 and 13: VSV-GP, Influenza A NS-1 , Influenza A M1 , Influenza A NP, LCMV NP, LCMV GP, Ebola GP Ebola NP, murine gammaherpesvirus (MHV-68) M2, M3 (this corresponds to the human EBV and HHV8 viruses) and ORF73, chicken Ovalbumin (OVA), or a helper T-cell epitope. These antigenic sequences will furthermore be combined so at least 2 or more are encoded in the same vector.
See figure 14 for an example of a comparison of the efficiency of Ad-Ii-GP pg33 and gp276 constructs compared to Ad-GP-Lamp-1 pg33 and gp276 constructs as measured by CD8+ T-cell responses. As can be seen, the Ad-Ii-GP constructs are superior to the Ad-GP-Lamp-1 constructs in their capability of evoking a CD8+ T-cell response.
Example 7:
Comparative analyses of novel constructs and alternative administration methods with respect to kinetics, magnitude of response, long-term immunity and virus dose needed for immunity.
All constructs described in Example 6 are comparatively analysed among each other and to Ad-GP and Ad-IiGP following alternative administration methods. The comparisons are done with respect to the kinetics and magnitude of immune response e.g. as described in Examples 1 , 2 and 3, long-term immunity e.g. as described in Examples 1 , 4 and 5, and virus dose needed for immunity. Alternative administration methods include:
1. Alterations with regard to enhanced delivery of co-stimulatory signals and cytokine help in which all of the in Example 6 described constructs are co- injected with adenovirus encoded type 1 interferon, for example tetracycline inducible IFN-β. Furthermore, all the in Example 6 described constructs are co-injected with adenovirus encoded cytokine, for example IL-15.
2. Administration of Ad-IiGP simultaneously with Ad-Tet-onGP at separate sites of the body (Ad-Tet-onGP encodes GP under control of a tetracycline inducible promoter).
3. Adenovirally delivery of any one of the in Example 6 described inserts/constructs followed by homologous viral vector boosting with the same insert/construct or followed by heterologous viral vector boosting with lentivirus encoded or other adenovirus-encoded delivery of the same insert/construct.
Example 8:
Ad-IiGP confers rapid and superior protection against lethal LCMV infection in absence of major epitopes. Lymphocytic choriomeningitis virus full-length glycoprotein (GP) comprises four CD8+ specific epitopes of varying antigenicity, measured by the percentile of CD8+ cells with specificity for the individual epitope against all the CD8+ cells raised in response to GP vaccination. The predominant population of CD8+ cells raised against GP is specific for the gp33 epitope, a somewhat smaller population is specific for gp276, and minor populations are specific for gp118 and for gp92. As gp33 and gp276 are the major / immunodominant epitopes, we investigated how nil mutations of either epitope independently or both simultaneously would effect the efficiency of the protection offered by the Ad-IiGP fusion construct. C57BL/6 mice were vaccinated with 2x107 IFU Ad-GP and Ad-IiGP or sham infected in the right hind footpad. Each group consisted of 5 animals. On day 90 post vaccination, the animals were challenged with 20 pfu LCMV Arm 53b i.e. (intra cerebral) constructs carrying gp33 nil mutations, gp276 nil mutations or gp33/gp276 double nil mutations. Mortality was recorded for 14 days. As can be seen from figure 7, Ad-IiGP conferred superior protection against lethal LCMV infection despite the gp33 or gp276 nil mutations. The double nil mutation of gp33/gp276 lead to the survival of 70% of the animals compared to no surviving animals in the Ad-GP and Sham vaccinated groups.
Surviving animals from the experiment above were analyzed for epitope specific CD8+ or CD4+T cells by intracellular staining for peptide-induced IFN-γ of spleen cells. Expectedly, as can be seen from figure 8, in the absence of either gp33 or gp276, the major epitope specificity of the CD8+ or CD4+T cells is gp276 or gp33, respectively. In the absence of both gp33/gp276, the major epitope specificity of the CD8+ or CD4+T cells is gp92.
Example 9:
CD8+ and CD4+ T cell responses to vaccination with naked DNA-IiGP and DNA-GP. To investigate an alternative platform for invariant chain fusion vaccines other than adenoviral delivery, we tested the ability of IiGP and GP as naked DNA to raise GP- epitope specific CD8+ and CD4+ T cells. The naked DNA comprised the GP and Ii- GP fragments from the Adenoviral vectors illustrated in figure 1.
C57BL/6 mice were vaccinated with DNA coated onto 1.6-nm gold particles in a concentration of 2 μg DNA/mg gold, and the DNA-gold complex was coated onto plastic tubes such that 0.5 mg gold (1 μg DNA per shot) was delivered to the mouse per shot. Mice were immunized at the abdominal skin using a hand-held gene gun device employing compressed helium (400 psi) as the particle motive force. Mice were inoculated four times with an interval of 1 week and then allowed to rest for 1 week before investigation. The number of epitope specific CD8+ or CD4+ T cells were determined by intracellular staining for peptide-induced IFN-y of spleen cells. As can be seen from figure 9, DNA-IiGP efficiently induces CD8+ T-cell responses directed against several epitopes.
Example 10:
Ad-IiGP confers superior protection against challenge with tumor cells expressing the gp33 epitope from LCMV. Under normal circumstances, tumor cells express several different antigens recognized by T cells. To determine the efficiency of this response, B16.F10 melanoma cells expressing gp33 from LCMV were used to challenge vaccinated animals with.
C57BL/6 mice were vaccinated with 2x107 IFU Ad-GP or Ad-IiGP in the right hind footpad. As a control some animals were vaccinated with Ad-β-galactosidase (negative control) or infected with LCMV (positive control). On day 90 after vaccination/infection animals were challenged with 106 tumor cells subcutaneously and the tumor growth was followed by measuring the size of the tumor. Initially a tumor will form in all animals, but eventually the immune response directed towards gp33 will eliminate the tumor cells. Each group consisted of 7-10 animals. Prophylactic vaccination with Ad-Ii-GP resulted in tumor free mice in 70% of the cases compared to only 10% in Ad-GP vaccinated animals, as can be seen from figure 10.
In many cases the tumor has already formed when a physician sees the patient. To mimic this situation, C57BL/6 mice were injected with 106 B16.F10 tumor cells subcutaneously. After 5 days the tumors were palpably recognizable and could be measured. At this time point animals were vaccinated with 2x107 IFU Ad-GP or Ad- IiGP in the right hind footpad. As a control some animals were vaccinated with Ad-β- galactosidase (negative control) or infected with LCMV (positive control). Tumor growth was followed by measuring the size of the tumor, and once the size was greater that 12 mm in any dimension the animals were sacrificed. As can be seen from figure 11 , in mice given the therapeutic Ad-IiGP vaccine, the speed of tumor development was approximately half of that seen in mice given Ad-GP as therapeutic vaccine, as measured by number of days passing prior to reaching a 12 mm tumor size and animal sacrifice.
Example 11 :
To demonstrate that the vaccine construct also can induce protecting antibody response the VSV infection was used.
Virus and virus quantitation: Vesicular stomatitis virus (VSV) of the Indiana serotype was used throughout this study. Stocks of virus were propagated in L929 cells (ATCC CCL 1 ) and stored at -70 0C until use. Virus quantitation was performed by plaque assay on monolayers of L929 cells. In brief, serial 10-fold dilutions of virus were prepared in Eagle's minimal essential medium (F11 ) containing 1% L- glutamine, 1% penicillin/streptomycin, 5% NaHCO3 and 10% fetal calf serum. One ml of each dilution was added in duplicate to monolayers of L929 cells in petri dishes plated 48 hours earlier. After incubation for 90 min at 370C in 5% CO2, medium containing the virus dilutions was aspirated, and the monolayers were overlaid with a mixture of 2.5 ml 1 % agarose and 2.5 ml 2 x F11. Monolayers were then incubated for 24 hours at 370C in 5% CO2 before staining with a mixture of 1 ml of 1% agarose and 1 ml of 2 x F11 containing 1% neutralred. After further 24 hours of incubation, the numbers of PFU were counted.
Survival study: Mortality was used as parameter for the severity of VSV infection, based on previous findings that virus titer in CNS correlate strongly with clinical symptoms (Thomsen et al. 1997, Int. Immunol. 9:1757-1766.). Mice were inspected daily for signs of VSV-induced paralysis, and sacrificed when severe paralysis was noted and the animals expected to succumb within the next 24 hours.
Serum neutralizations test: Serial two-fold dilutions of serum in F11 were mixed with equal volumes of virus diluted to contain approximately 100 PFU/ml. After 1 h of incubation at room temperature, 1 ml of each serum-virus mixture was added in duplicate to monolayers of L929 cells in petri dishes and assayed for the presence of residual virus by plaque assay (see virus and virus quantitation). The highest serum dilution that reduced the number of plaques by at least 50% was taken as the neutralizing titer.
C57BL/6 mice were vaccinated in the right hind foot-pad with 2 x 107 IFU of adenovirus encoding either full-length glycoprotein of vesicular stomatitis virus (Ad- VSVGP) or glycoprotein linked to invariant chain (Ad-Ii-VSVGP). On day 7, 14, 21 and 110 after vaccination serum samples were collected and in vitro neutralizing antibody titers were determined in a plaque-reduction assay, figure 12a. On day 3, 7, 14, 21 and 110 after vaccination, animals were challenged with 105 PFU of VSV intranasally, and mortality was registered over the next 14 days, figure 12b. As seen in figure 12 almost identical responses are seen in the two groups, suggesting that, although not an advantage, the invariant chain allows for antibody production and does not have a detrimental effect on the production.
Reference List
Andersson, E. C, J. P. Christensen, O. Marker and A. R. Thomsen. 1994. Journal of Immunology 152:1237-1245. Changes in Cell Adhesion Molecule Expression on T Cells Associated with Systemic Virus Infection.
Andreasen, S. 0., J. P. Christensen, O. Marker and A. R. Thomsen. 1999.
International Immunology 11 :1463-1473. Virus induced non-specific signals cause cell cycle progression of primed CD8+ T cells but do not induce cell differentiation.
Battegay M., S. Cooper, A. Althage, J. Banziger, H. Hengartner, and R. M. Zinckernagel.1991. Journal of Virological Methods 33:191-198. Quantification of lymphocytic choriomenigitis virus with an immunological focus assay in 24- or 96- well plates.
Becker, T- C, R. J. Noel, W. S. Coats, A. M. Gomez-Foix, T. Alam, R. D. Gerard, and C. B. Newgard. 1994. Use of recombinant adenovirus for metabolic engineering
Christensen, J. P., O. Marker, A. R. Thomsen. 1994. Scandinavian Journal of Immunology 40:373-382. The role of CD4+ T cells in cell-mediated immunity to LCMV: studies in MHC class I and class Il deficient mice.
Diebold, S. S., M. Gotten, N. Koch, and M. Zenke. 2001. MHC class Il presentation of endogenously expressed antigens by transfected dendritic cells. Gene Ther. 8:487-493.
Morris, C. R., A. J. Reber, J. L. Petersen, S. E. Vargas, and J. C. Solheim. 2004. Association of intracellular proteins with folded major histocompatibility complex class I molecules. Immunol. Res. 30:171-179.
Pieters, J. 1997. MHC class Il restricted antigen presentation. Curr.Opin. Immunol. 9:89-96.
Sambrook et al., eds., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, 2nd Edition, Cold Spring Harbor, N.Y., 1989. Strumptner-Cuvelette, P., and P. Benaroch. 2002. Biochem. Biophys. Acta., 1542:1- 13. Multiple roles of the invariant chain in MHC class Il function.
Thomsen, R. A., A. Nansen, C. Andersen, J. Johansen, O. Marker, and J. P. Christensen. 1997. International Immunology 9:1757-1766. Cooperation of B cells and T cells is required for survival of mice infected with vesicular stomatitis virus.

Claims

Claims
1. A nucleic acid construct comprising sequences encoding: a) at least one invariant chain operatively linked to b) at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide.
2. The nucleic acid construct according to ciaim 1 , wherein at least one invariant chain is organism specific.
3. The nucleic acid construct according any of the claims 1 or 2, wherein at least one invariant chain is of vertebrate origin.
4. The nucleic acid construct according to any of the preceding claims, wherein at least one invariant chain is of mammalian origin.
5. The nucleic acid construct according to any of the preceding claims, wherein at least one invariant chain is of human origin.
6. The nucleic acid construct according to any of the preceding claims, wherein the encoded at least one invariant chain is a fragment of the SEQ ID NO: 2 of at least 40 amino acids and of 85% homology to the same fragment of SEQ ID NO: 2.
7. The nucleic acid construct according to any of the preceding claims, wherein the encoded at least one invariant chain is a fragment of the SEQ ID NO: 2 of at least 186 amino acids.
8. The nucleic acid construct according to any of the preceding claims, wherein the encoded at least one invariant chain is 85% identical to SEQ ID NO: 2.
9. The nucleic acid construct according to any of the preceding claims, wherein the encoded at least one invariant chain is identical to SEQ ID NO: 2.
10. The nucleic acid construct according to any of the preceding claims, wherein at least one region, peptide or domain of the at least one invariant chain is added to, removed from or substitutes a region, peptide or domain of the at least one invariant chain.
11. The nucleic acid construct according to any of the preceding claims, wherein at least one signal peptide is added to, removed from or replaces the signal peptide of the at least one invariant chain.
12. The nucleic acid construct according to any of the preceding claims, at least one CLIP region is added to, removed from or replaces the CLIP region of the at least one invariant chain.
13. The nucleic acid construct according to any of the preceding claims wherein the at least one invariant chain or fragments of same elicit an MHC-I and/or an
MHC-II dependent immune response in a CD4+ T cell dependent and/or independent manner.
14. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is selected from the group of: pathogenic organisms, cancer-specific polypeptides, and proteins or peptides associated with an abnormal physiological response.
15. The nucleic acid construct according to any of the preceding claims, wherein the at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide from a pathogenic organism is selected from the group of pathogens comprising: virus, micro organisms and parasites.
16. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a mammalian organism, preferably human pathogen.
17. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a virus.
18. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a virus selected from the group of: HIV, Hepatitis C virus, influenza virus, herpes virus, Lassa, Ebola, smallpox, Bird flu, filovirus, Marburg, papillomavirus
19. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a micro organism selected from the group of: Mycobacterium tuberculosis, Bacillus anthracis, Staphylococcus species, and Vibrio species.
20. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a parasite selected from the group of: Plasmodium species, Leishmania species, and Trypanosoma species.
21. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is an antigenic peptide or protein at least 85% identical to any of the in claims 14, 15 or 16 described antigens.
22. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a cancer-specific polypeptide.
23. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a cancer-specific polypeptide selected from the group of: p53, telomerase and melanoma antigen.
24. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a polypeptide associated with an abnormal physiological response.
25. The nucleic acid construct according to any of the preceding claims, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a polypeptide associated with an abnormal physiological response, wherein the abnormal physiological response is an autoimmune disease, an allergic reaction, cancer or a congenital disease.
26. The nucleic acid construct according to any of the preceding claims, wherein the operative link between the invariant chain and the antigenic protein or peptide or an antigenic fragment of said protein or peptide is selected from the group of: a direct link or a link mediated by a spacer region.
27. The nucleic acid construct according to claim 26, wherein the operative linker is a spacer region.
28. The nucleic acid construct according to claim 27, wherein the spacer region encodes at least one helper epitope for class Il MHC molecules.
29. The nucleic acid construct according to claim 28, wherein the spacer region encodes at least one protease cleavage site.
30. The nucleic acid construct according to claim 26, comprising at least one siRNA encoding sequence.
31. The nucleic acid construct according to claim any of the preceding claims, wherein at least one invariant chain is operatively linked to at least two, antigenic proteins or peptides or an antigenic fragment of said protein or peptide.
32. The nucleic acid construct according to claim 31, wherein the number of antigenic proteins or peptides or antigenic fragments of said protein or peptide is three, four, five six, eight or ten or more.
33. The nucleic acid construct according to claim 31 or 32, wherein each invariant chain element and each antigenic element is operatively linked to each other as defined in claims 26 to 30.
34. The nucleic acid construct according to any of the preceding claims, wherein the at least one operatively linked invariant chain and at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide encoding sequence is preceded by a promoter enabling expression of the construct.
35. The nucleic acid construct according to any of the preceding claims, wherein the promoter is selected from the group of constitutive promoters, inducible promoters, organism specific promoters, tissue specific promoters and cell type specific promoters, CMV promoter, SV40 promoter, and RSV promoter.
36. The nucleic acid construct according to any of the preceding claims, comprising genes related to antigen presentation and/or intercellular spreading.
37. A delivery vehicle comprising the nucleic acid construct according to any of the preceding claims
38. The delivery vehicle according to claim 37, wherein the vehicle is selected from the group of: RNA based vehicles, DNA based vehicles/ vectors, lipid based vehicles, virally based vehicles and cell based vehicles.
39. The vehicle according to claim 37 and 38, wherein the vehicle is a virus selected from the group of adenoviruses, retroviruses, lentiviruses, adeno- associated viruses, herpesviruses, vaccinia viruses, foamy viruses, cytomegaloviruses, Semliki forest virus, poxviruses, RNA virus vector, DNA virus vector.
40. The vehicle according to claims 37 to 39, wherein the vehicle is an adenovirus.
41. The vehicle according to claim 40, wherein the vehicle is a replication defective adenovirus or a conditionally replication deficient adenovirus.
42. A cell comprising the nucleic acid construct as defined in any of the claims 1 to 41.
43. The cell according to claim 42, wherein the cell is an antigen presenting cell
44. The cell according to claims 42 and 43, wherein the antigen presenting cell is a professional antigen presenting cell.
45. The cell according to any of the claims 42 to 44, wherein the cell is an MHC-I expressing cell.
46. The cell according to any of the claims 42 to 45, wherein the cell is a stem cell obtained from the patient.
47. The cell according to claim 46, wherein the stem cell is capable of differentiating into an antigen presenting cell.
48. The cell according to any of the claims 42 to 47, wherein the antigen presenting cell does not express any co-stimulatory signals and the antigenic protein or peptide or antigenic fragment of said protein or peptide is an auto- antigen.
49. A chimeric protein comprising at least one operatively linked invariant chain and at least one antigenic protein or peptide encoding sequence, as defined by any of the nucleic acid constructs in any of the claims 1 to 36.
50. An antibody that can recognize the chimeric protein as defined in claim 49.
51. Use of the antibody according to claim 50, for passive immunization of an animal.
52. Use of the antibody according to claim 50, in an assay for detecting proteins to which the antibody binds.
53. A vaccine comprising a nucleic acid sequence encoding: a) at least one invariant chain operatively linked to b) at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide.
54. A vaccine composition comprising a nucleic acid sequence encoding: a) at least one invariant chain operatively linked to b) at least one polypeptide for use as a medicament.
55. The vaccine according to any of the claims 53 and 54, comprising a nucleic acid construct as defined in any of the claims 1 to 36.
56. The vaccine according to claim 55, wherein the nucleic acid is packaged
57. The vaccine according to any of the claims 53 to 56, wherein the nucleic acid construct is packaged by a packaging means selected from the group of: liposomes, coated onto colloidal gold particle, viral expression vector, viral delivery vector, pegylated vector.
58. The vaccine according to any of the claims 53 to 57, wherein the packaging means is a viral expression vector selected from the group of: adenovirus, retrovirus, lentivirus, adeno-associated virus, herpes virus, vaccinia virus, DNA virus vector.
59. The vaccine according to any of the claims 53 to 58, wherein the nucleic acid construct is packaged into an adenoviral vector.
60. The vaccine according to claim 59, wherein the adenoviral vector is replication deficient or conditionally replication deficient.
61. The vaccine according to any of the claims 53 to 60, wherein the vaccine comprises at least two vectors.
62. The vaccine according to any of the claims 53 to 61 , wherein the vaccine comprises at three, four five or six vectors.
63. A vaccine comprising at least one chimeric protein according to claim 49 as defined by any of the nucleic acid constructs in any of the claims 1 to 36.
64. The vaccine according to any of the claims 53 to 63, wherein the vaccine comprises an adjuvant.
65. The vaccine according to any of the claims 53 to 64, wherein the adjuvant is selected from the group of: Freund adjuvant, Montanide ISA 52, Montanide ISA 720 and alum.
66. The vaccine according to any of the claims 53 to 64, wherein the vaccine comprises means for intramuscular, intravenous or subcutaneous administration.
67. The vaccine according to any of the claims 53 to 64, wherein the vaccine comprises means for oral, rectal or vaginal administration.
68. The vaccine according to any of the claims 53 to 64, wherein the vaccine comprises means for intranasal/pulmonary administration.
69. The vaccine composition according to any of the claims 53 to 68, comprising a second active ingredient.
70. The vaccine composition according to claim 69, wherein the second active ingredient is selected from the group of: antibiotics, chemotherapeutics, anti- allergenics, cytokines and co-stimulatory molecules of the immunesystem.
71. A kit in parts comprising: - a vaccine composition comprising a nucleotide construct according to any of the claims 53 to 70
- a medical instrument or other means for administering the vaccine
- instructions on how to use the kit in parts.
72. The kit in parts according to claim 71, comprising a second active ingredient.
73. A method for inducing an immune response in an animal, comprising administering to the animal a vaccine according to any of the claims 53 to 70.
74. The method according to claim 73, wherein the immune response is MHC-I dependent.
75. The method according to any of the claims 73 or 74 , wherein the immune response is MHC-I and/ or MHC-II dependent.
76. The method according to any of the claims 73 to 75, wherein the immune response is T-cell dependent.
77. The method according to any of the claims 73 to 76, wherein the immune response is CD4+ T-cell dependent.
78. The method according to any of the claims 73 to 76, wherein the immune response is CD4+ T-cell independent.
79. The method according to any of the claims 73 to 78, wherein the immune response is CD8+ T-cell dependent.
80. The method according to any of the claims 73 to 79, wherein the immune response is B cell dependent.
81. The method, according to any of the claims 73 to 80, comprising the steps of:
- providing at least one vaccine according to any of the claims 53 to 70
- administering said at least one vaccine to a subject at least once for treatment or prophylaxis of an animal.
82. The method according to claim 81 , wherein at least one vaccine is administered any number of times from two, three, four or five times.
83. The method according to claim 82, wherein the vaccine is boosted by administration in a form or body part different from the previous administration.
84. The method according to any of the claims 73 to 83, wherein the vaccine is administered to the area most likely to be the receptacle of a given infection.
85. The method according to any of the claims 73 to 84, wherein the animal is a vertebrate.
86. The method according to any of the claims 73 to 85, wherein the animal is a mammal.
87. The method according to any of the claims 73 to 86, wherein the mammal is selected from the group of: cows, pigs, horses, sheep, goats, llamas, mice, rats, monkeys and humans
88. The method according to any of the claims 73 to 87, wherein the mammal is a human.
89. A method for preparing a vaccine for genetic immunization comprising the steps of:
- preparing a nucleic acid construct according to any of the claims 1 to 36,
- administering said nucleic acid construct.
90. A method of treatment comprising administering the vaccine according to claims 53 to 70, to treat a clinical condition in an individual in need thereof.
91. Use of the nucleic acid construct according to claims 1 to 36, for the production of a vaccine.
92. An adenoviral vector comprising a nucleotide construct encoding: a) at least one antigen and b) at least one protein or peptide or fragment of a protein or peptide which stimulates an MHC-I response and/or an MHC-II response and/or intercellular spreading.
93. The adenoviral vector according to claim 92, wherein the nucleotide construct encodes at least one protein or peptide or fragment of a protein or peptide which stimulates an MHC-I response.
94. The adenoviral vector according to any of the claims 92 and 93, wherein the at least one antigen is operatively linked to the at least one MHC response stimulating protein or peptide or fragment of an MHC response stimulating protein or peptide.
95. The adenoviral vector according to claim 94, wherein the MHC stimulating protein or peptide or fragment of protein or peptide is an MHC associated protein or peptide.
96. The adenoviral vector according to claim 95, wherein the MHC associated peptide is selected from the group of: ER localizing peptide, Golgi localizing peptide, Endosomal peptide loading compartment localizing peptide, lysosomal, MIIC, CIIV, melanosomes, secretory granules, Birbeck granules.
97. The adenoviral vector according to claim 96, wherein the MHC associated protein or peptide or fragment of a protein or peptide is selected from the group of sorting signal peptides, LAMP, LIMP, Hsp70, calreticulin and invariant chain.
98. The adenoviral vector according to any of the claims 92 to 97, comprising proteins or peptides related to intercellular spreading such as VP22, Cx43, HIV Tat, other connexins or gap-junction constituents.
99. The adenoviral vector according to any of the claims 92 to 98, wherein the at least one MHC response stimulating protein or peptide or fragment of protein or peptide is invariant chain.
100. The adenoviral vector according to claim 99, wherein at least one invariant chain is organism specific.
101. The adenoviral vector according any of the claims 99 to 100, wherein at least one invariant chain is of vertebrate origin.
102. The adenoviral vector according to any of the claims 99 to 101 , wherein at least one invariant chain is of mammalian origin.
103. The adenoviral vector according to any of the claims 99 to 102, wherein at least one invariant chain is of human origin.
104. The adenoviral vector according to any of the claims 99 to 103, wherein the encoded at least one invariant chain is a fragment of the SEQ ID NO: 2 of at least 40 amino acids and of 85% homology to the same fragment of SEQ ID NO: 2
105. The adenoviral vector according to any of the claims 99 to 104, wherein the encoded at least one invariant chain is a fragment of the SEQ ID NO: 2 of at least 186 amino acids.
106. The adenoviral vector according to any of the claims 99 to 105, wherein the encoded at least one invariant chain is 85% identical to SEQ ID NO: 2.
107. The adenoviral vector according to any of the claims 99 to 106, wherein the encoded at least one invariant chain is identical to SEQ ID NO: 2.
108. The adenoviral vector according to any of the claims 99 to 107, wherein at least one region, peptide or domain of the at least one invariant chain is added to, removed from or substitutes a region, peptide or domain of the at least one invariant chain.
109. The adenoviral vector according to any of the claims 99 to 108, wherein at least one signal peptide is added to, removed from or replaces the signal peptide of the at least one invariant chain.
110. The adenoviral vector according to any of the claims 99 to 109, at least one CLIP region is added to, removed from or replaces the CLIP region of the at least one invariant chain.
111. The adenoviral vector according any of the claims 92 to 110, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is selected from the group of: pathogenic organisms, cancer-specific polypeptides, and proteins or peptides associated with an abnormal physiological response.
112. The adenoviral vector according to any of the claims 92 to 111 , wherein the at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide from a pathogenic organism is selected from the group of pathogens comprising: virus, micro organisms and parasites.
113. The adenoviral vector according to any of the claims 92 to 112, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a mammalian pathogen.
114. The adenoviral vector according to any of the claims 92 to 113, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a human pathogen.
115. The adenoviral vector according to any of the claims 92 to 114, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a virus.
116. The adenoviral vector according to any of the claims 92 to 115, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a virus selected from the group of: HIV, Hepatitis C virus, influenza virus, herpes virus, Lassa, Ebola, smallpox, Bird flu, filovirus, Marburg, papillomavirus
117. The adenoviral vector according to any of the claims 92 to 116, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a micro organism selected from the group of: Mycobacterium tuberculosis, Bacillus anthracis, Staphylococcus species, and Vibrio species.
118. The adenoviral vector according to any of the claims 92 to 117, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a parasite selected from the group of: Plasmodium species, Leishmania species, and Trypanosoma species.
119. The adenoviral vector according to any of the claims 92 to 118, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is an antigenic peptide or protein at least 85% identical to any of the in claims 14, 15 or 16 described antigens.
120. The adenoviral vector according to any of the claims 92 to 119, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a cancer-specific polypeptide.
121. The adenoviral vector according to any of the claims 92 to 120, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a cancer-specific polypeptide selected from the group of: p53, telomerase and melanoma antigen.
122. The adenoviral vector according to any of the claims 92 to 121 , wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a polypeptide associated with an abnormal physiological response.
123. The adenoviral vector according to any of the claims 92 to 122, wherein at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide is from a polypeptide associated with an abnormal physiological response, wherein the abnormal physiological response is an autoimmune disease, an allergic reaction, cancer or a congenital disease.
124. The adenoviral vector according to any of the claims 92 to 123, wherein the operative link between the invariant chain and the antigenic protein or peptide or an antigenic fragment of said protein or peptide is selected from the group of: a direct link or a link mediated by a spacer region.
125. The adenoviral vector according to claim 124, wherein the operative linker is a spacer region.
126. The adenoviral vector according to claim 125, wherein the spacer region encodes at least one helper epitope for class Il MHC molecules.
127. The adenoviral vector according to claim 124, wherein the spacer region encodes at least one protease cleavage site.
128. The adenoviral vector according to claim 124, comprising at least one siRNA encoding sequence.
129. The adenoviral vector according to claim any of the claims 92 to 128, wherein at least one invariant chain is operatively linked to at least two, antigenic proteins or peptides or an antigenic fragment of said protein or peptide.
130. The adenoviral vector according to claim 129, wherein the number of antigenic proteins or peptides or antigenic fragments of said protein or peptide is three, four, five six, eight or ten or more.
131. The adenoviral vector according to any of the claims 129 and 130, wherein each invariant chain element and each antigenic element is operatively linked to each other as defined in claims 124 to 128.
132. The adenoviral vector according to any of the claims 92 to 131 , wherein the at least one operatively linked invariant chain and antigenic protein or peptide or an antigenic fragment of said protein or peptide encoding sequence is preceded by a promoter enabling expression of the construct.
133. The adenoviral vector according to any of the claims 92 to 132, wherein the promoter is selected from the group of constitutive promoters, inducible promoters, organism specific promoters, tissue specific promoters and cell type specific promoters.
134. The adenoviral vector according to any of the claims 92 to 133, wherein the promoter is selected from the group of: CMV promoter, SV40 promoter and the
RSV promoter.
135. The adenoviral vector according to any of the vehicle according to any of the claims 92 to 134, wherein the vector is a replication defective adenovirus or a conditionally replication deficient adenovirus.
136. A cell comprising the adenoviral vector as defined in any of the claims 92 to 135.
137. The cell according to claim 136, wherein the cell is an antigen presenting cell
138. The cell according to any of the claims 136 and 137, wherein the antigen presenting cell is a professional antigen presenting cell.
139. The cell according to any of the claims 136 to 138, wherein the cell is an MHC- I expressing cell.
140. The cell according to any of the claims 136 to 139, wherein the cell is a stem cell obtained from the patient.
141. The cell according to claim 140, wherein the stem cell is capable of differentiating into an antigen presenting cell.
142. The cell according to any of the claims 136 to 141 , wherein the antigen presenting cell does not express any co-stimulatory signals and the antigenic protein or peptide or antigenic fragment of said protein or peptide is an auto- antigen.
143. A chimeric protein comprising at least one operatively linked protein or peptide or fragment of a protein or peptide which stimulates an MHC-I response, as defined by any of the adenoviral vectors in any of the claims 92 to 135.
144. An antibody that can recognize the chimeric protein as defined in claim 143.
145. Use of the antibody according to claim 144, for passive immunization of an animal.
146. Use of the antibody according to claim 144, in an assay for detecting proteins to which the antibody binds.
147. A vaccine comprising a nucleic acid sequence encoding: a) at least one operatively linked protein or peptide or fragment of a protein or peptide which stimulates an MHC-I response. b) at least one antigenic protein or peptide or an antigenic fragment of said protein or peptide.
148. A vaccine composition comprising a nucleic acid sequence encoding: a) at least one at least one operatively linked protein or peptide or fragment of a protein or peptide which stimulates an MHC-I response operatively linked to b) at least one polypeptide for use as a medicament.
149. The vaccine according to any of the claims 147 and 148, comprising an adenoviral vector as defined in any of the claims 92 to 135.
150. The vaccine according to claim 149, wherein the adenoviral vector is packaged.
151. The vaccine according to any of the claims 147 to 150, wherein the adenoviral vector is packaged by a packaging means selected from the group of: liposomes, coated onto colloidal gold particle, viral expression vector, viral delivery vector
152. The vaccine according to any of the claims 147 to 151, wherein the packaging means is a viral expression vector selected from the group of: adenovirus, retrovirus, lentivirus, adeno-associated virus, herpes virus, vaccinia virus, DNA virus vector.
153. The vaccine according to any of the claims 147 to 152, wherein the adenoviral vector is packaged into an adenoviral vector.
154. The vaccine according to any of the claims 147 to 153, wherein the adenoviral vector is replication deficient or conditionally replication deficient.
155. The vaccine according to any of the claims 147 to 154, wherein the vaccine comprises at least two vectors.
156. The vaccine according to any of the claims 147 to 155, wherein the vaccine comprises at three, four five or six vectors.
157. A vaccine comprising at least one chimeric protein according to claim 143 as defined by any of the adenoviral vectors in any of the claims 92 to 135.
158. The vaccine according to any of the claims 147 to 157, wherein the vaccine comprises an adjuvant.
159. The vaccine according to any of the claims 147 to 158, wherein the adjuvant is selected from the group of: Freund adjuvant, Montanide ISA 52, Montanide
ISA 720 and alum.
160. The vaccine according to any of the claims 147 to 159, wherein the vaccine comprises means for intramuscular, intravenous or subcutaneous administration.
161. The vaccine according to any of the claims 147 to 160, wherein the vaccine comprises means for oral, rectal or vaginal administration.
162. The vaccine according to any of the claims 147 to 161 , wherein the vaccine comprises means for intranasal/pulmonary administration.
163. The vaccine composition according to any of the claims 147 to 162, comprising a second active ingredient.
164. The vaccine composition according to claim 163, wherein the second active ingredient is selected from the group of: antibiotics, chemotherapeutics, anti- allergenics, cytokines and co-stimulatory molecules of the immunesystem.
165. A kit in parts comprising:
- a vaccine composition comprising an adenoviral vector according to any of the claims 92 to 135,
- a medical instrument or other means of administering said vaccine,
- instructions on how to use the kit in parts.
166. The kit in parts according to claim 165, comprising a second active ingredient.
167. A method for inducing an immune response in an animal, comprising administering to the animal a vaccine according to any of the claims 147 to 164.
168. The method according to claim 167, wherein the immune response is MHC-I dependent.
169. The method according to any of the claims 167 to 168, wherein the immune response is MHC-I and/ or MHC-II dependent.
170. The method according to any of the claims 167 to 169, wherein the immune response is T-cell dependent.
171. The method according to any of the claims 167 to 170, wherein the immune response is CD4+ T-cell dependent.
172. The method according to any of the claims 167 to 171, wherein the immune response is CD4+ T-cell independent.
173. The method according to any of the claims 167 to 172, wherein the immune response is CD8+ T-cell dependent.
174. The method according to any of the claims 167 to 173, wherein the immune response is B cell dependent.
175. The method according to any of the claims 167 to 174, comprising the steps of:
- providing at least one vaccine according to any of the claims 147 to 164
- administering said at least one vaccine to a subject at least once for treatment or prophylaxis of an animal.
176. The method according to claim 175, wherein at least one vaccine is administered any number of times from two, three, four or five times.
177. The method according to claim 175, wherein the vaccine is boosted by administration in a form or body part different from the previous administration.
178. The method according to any of the claims 167 to 177, wherein the vaccine is administered to the area most likely to be the receptacle of a given infection.
179. The method according to any of the claims 167 to 178, wherein the animal is a vertebrate.
180. The method according to any of the claims 167 to 179, wherein the animal is a mammal.
181. The method according to any of the claims 167 to 180, wherein the mammal is selected from the group of: cows, pigs, horses, sheep, goats, llamas, mice, rats, monkeys and humans
182. The method according to any of the claims 167 to 181 , wherein the mammal is a human.
183. A method for preparing a vaccine for genetic immunization comprising the steps of: - preparing a adenoviral vector according to any of the claims 92 to
135 - administering said adenoviral vector.
184. A method of treatment comprising administering the vaccine according to claims 147 to 164, to treat a clinical condition in an individual in need thereof.
185. Use of the adenoviral vector according to claims 92 to 135, for the production of a vaccine.
PCT/DK2006/000675 2005-11-30 2006-11-30 A nucleotide vaccine WO2007062656A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
PL06818135T PL1957528T3 (en) 2005-11-30 2006-11-30 A nucleotide vaccine
DK06818135.3T DK1957528T3 (en) 2005-11-30 2006-11-30 The nucleotide VACCINE
US12/095,730 US20100278904A1 (en) 2005-11-30 2006-11-30 Nucleotide vaccine
EP06818135A EP1957528B1 (en) 2005-11-30 2006-11-30 A nucleotide vaccine
CN200680045006.4A CN101321781B (en) 2005-11-30 2006-11-30 A kind of nucleotide vaccine
SI200631493T SI1957528T1 (en) 2005-11-30 2006-11-30 A nucleotide vaccine
ES06818135T ES2396437T3 (en) 2005-11-30 2006-11-30 Nucleotide vaccine
US14/817,631 US20150368672A1 (en) 2005-11-30 2015-08-04 Nucleotide vaccine
US17/166,419 US20210230633A1 (en) 2005-11-30 2021-02-03 Nucleotide vaccine

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
DKPA200501697 2005-11-30
DKPA200501697 2005-11-30
US75571205P 2005-12-30 2005-12-30
DKPA200501857 2005-12-30
DKPA200501857 2005-12-30
US60/755,712 2005-12-30
DKPA200600431 2006-03-27
DKPA200600431 2006-03-27

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/095,730 A-371-Of-International US20100278904A1 (en) 2005-11-30 2006-11-30 Nucleotide vaccine
US14/817,631 Continuation US20150368672A1 (en) 2005-11-30 2015-08-04 Nucleotide vaccine

Publications (2)

Publication Number Publication Date
WO2007062656A2 true WO2007062656A2 (en) 2007-06-07
WO2007062656A3 WO2007062656A3 (en) 2008-05-22

Family

ID=43030525

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2006/000675 WO2007062656A2 (en) 2005-11-30 2006-11-30 A nucleotide vaccine

Country Status (10)

Country Link
US (3) US20100278904A1 (en)
EP (1) EP1957528B1 (en)
CN (2) CN101321781B (en)
DK (1) DK1957528T3 (en)
ES (1) ES2396437T3 (en)
HK (1) HK1215052A1 (en)
PL (1) PL1957528T3 (en)
PT (1) PT1957528E (en)
SI (1) SI1957528T1 (en)
WO (1) WO2007062656A2 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009014702A2 (en) * 2007-07-25 2009-01-29 Board Of Regents, The University Of Texas System Cd74 for use in the treatment of cancer and liver toxicity
WO2010057501A1 (en) * 2008-11-21 2010-05-27 Københavns Universitet (University Of Copenhagen) Priming of an immune response
WO2011114346A1 (en) 2010-03-18 2011-09-22 Chetan Balar Chitin and related compounds for use in treating bacterial and viral infections
CN103966219A (en) * 2014-05-26 2014-08-06 武汉大学 Construction and application of human foamy virus vector mediated shRNA for inhibiting HIV-1 replication
WO2014139587A1 (en) 2013-03-15 2014-09-18 Okairòs Ag Improved poxviral vaccines
WO2017025782A1 (en) 2014-09-17 2017-02-16 Glaxosmithkline Biologicals Sa Improved poxviral vaccines
AU2015227479B2 (en) * 2008-11-21 2017-05-04 Københavns Universitet (University Of Copenhagen) Priming of an immune response
WO2018037045A1 (en) 2016-08-23 2018-03-01 Glaxosmithkline Biologicals Sa Fusion peptides with antigens linked to short fragments of invariant chain (cd74)
WO2018060288A1 (en) 2016-09-29 2018-04-05 Glaxosmithkline Biologicals S.A. Compositions and methods of treatment of persistent hpv infection
WO2018104911A1 (en) 2016-12-09 2018-06-14 Glaxosmithkline Biologicals Sa Adenovirus polynucleotides and polypeptides
WO2019012091A1 (en) * 2017-07-12 2019-01-17 Nouscom Ag Neoantigen vaccine composition for treatment of cancer
WO2019115816A1 (en) 2017-12-15 2019-06-20 Glaxosmithkline Biologicals Sa Hepatitis b immunisation regimen and compositions
WO2019115817A2 (en) 2017-12-15 2019-06-20 Glaxosmithkline Biologicals Sa Hepatitis b immunisation regimen and compositions
WO2019239311A1 (en) 2018-06-12 2019-12-19 Glaxosmithkline Biologicals Sa Adenovirus polynucleotides and polypeptides
US10576143B2 (en) * 2013-03-15 2020-03-03 Glaxosmithkline Biologicals Sa Poxviral vaccines
WO2020128012A1 (en) 2018-12-21 2020-06-25 Glaxosmithkline Biologicals Sa Methods of inducing an immune response
WO2020178359A1 (en) 2019-03-05 2020-09-10 Glaxosmithkline Biologicals Sa Hepatitis b immunisation regimen and compositions
US20210008195A1 (en) * 2017-12-20 2021-01-14 Zoetis Services Llc Vaccines against hendra and nipah virus infection
US11179461B2 (en) 2017-03-20 2021-11-23 University Of Copenhagen Ii vaccine adjuvant
EP3737411A4 (en) * 2018-08-10 2022-01-05 NantBio, Inc. Cellular adjuvants for viral infection
WO2022051866A1 (en) * 2020-09-11 2022-03-17 The University Of British Columbia Vaccine for viral pathogens
EP4137153A1 (en) 2021-08-18 2023-02-22 Sirion Biotech GmbH Therapeutic papilloma virus vaccines

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0918154D0 (en) * 2009-10-16 2009-12-02 Isis Innovation Mycobacterial vaccines
CN101991863B (en) * 2010-04-28 2012-11-07 中国人民解放军军事医学科学院野战输血研究所 Double-target DNA vaccine and constructing method thereof
CN104350070A (en) * 2012-01-27 2015-02-11 生物防护科技公司 Compositions and methods of modulating immune response
CN103819562B (en) * 2013-06-03 2017-02-08 安徽农业大学 Construction and application of vaccine vector based on chicken invariant chain fragments
CN104306995B (en) * 2014-10-29 2017-04-12 山东农业大学 Epitope vaccine for resisting subgroup J avian leukosis virus infection as well as preparation method and application of epitope vaccine
CN106511991B (en) * 2016-12-12 2019-11-08 南京农业大学 Application of the Ii gene order in Porcine epidemic diarrhea virus gene vaccine
RU2725493C2 (en) * 2018-12-28 2020-07-02 Федеральное государственное бюджетное учреждение науки институт биоорганической химии им. академиков М.М. Шемякина и Ю.А. Овчинникова Российской академии наук (ИБХ РАН) System for stable expression of tumor-associated antigens based on lentiviral vector
CN112430625B (en) * 2020-11-23 2022-08-05 武汉枢密脑科学技术有限公司 Recombinant adeno-associated virus transfer vector containing variant porcine pseudorabies virus gD protein gene, virus, preparation method and application thereof
CN116981777A (en) * 2021-03-12 2023-10-31 巴斯德研究所 Lentiviral vectors targeting MHC-II pathway antigens and inducing protective CD8+ and CD4+ T cell immunity in hosts

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997010335A1 (en) * 1995-09-14 1997-03-20 Universität Tübingen Recombinant polypeptide based on the primary sequence of the invariant chain with at least one primary sequence of a specific t-cell epitope or a protein derivative and nucleic acids coding for this recombinant polypeptide
DE10228218A1 (en) * 2002-06-25 2004-02-05 Norbert Prof. Koch New nucleic acid for polypeptide containing trimerization domain and epitopes, useful for vaccination against e.g. infectious disease or tumors, improves peptide antigenicity

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1548118A2 (en) * 1994-06-10 2005-06-29 Genvec, Inc. Complementary adenoviral vector systems and cell lines
WO1998004720A1 (en) * 1996-07-26 1998-02-05 Sloan-Kettering Institute For Cancer Research Method and reagents for genetic immunization
AU6336398A (en) * 1997-02-24 1998-09-09 Johns Hopkins University, The Immunomodulatory polypeptides derived from the invariant chain of mhc class ii
CN1290244A (en) * 1998-02-09 2001-04-04 Rpc公司 Process for treating cobalt catalyst in oxidation mixtures of hydrocarbons to dibasic acids
US6534482B1 (en) * 1998-05-13 2003-03-18 Epimmune, Inc. Expression vectors for stimulating an immune response and methods of using the same
US6432409B1 (en) * 1999-09-14 2002-08-13 Antigen Express, Inc. Hybrid peptides modulate the immune response
ES2404689T3 (en) * 2001-04-05 2013-05-28 The Johns Hopkins University Chimeric vaccines comprising the lumenal domain of LAMP-1 or LAMP-2
US7179645B2 (en) * 2002-09-24 2007-02-20 Antigen Express, Inc. Ii-Key/antigenic epitope hybrid peptide vaccines

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997010335A1 (en) * 1995-09-14 1997-03-20 Universität Tübingen Recombinant polypeptide based on the primary sequence of the invariant chain with at least one primary sequence of a specific t-cell epitope or a protein derivative and nucleic acids coding for this recombinant polypeptide
DE10228218A1 (en) * 2002-06-25 2004-02-05 Norbert Prof. Koch New nucleic acid for polypeptide containing trimerization domain and epitopes, useful for vaccination against e.g. infectious disease or tumors, improves peptide antigenicity

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
ANTOINE J-C ET AL: "LOCALIZATION OF MAJOR HISTOCOMPATIBILITY COMPLEX CLASS II MOLECULES IN PHAGOLYSOSOMES OF MURINE MACROPHAGES INFECTED WITH LEISHMANIA-AMAZONENSIS" INFECTION AND IMMUNITY, vol. 59, no. 3, 1991, pages 764-775, XP002434457 ISSN: 0019-9567 *
DATABASE Geneseq [Online] 29 January 2003 (2003-01-29), "Human expressed protein tag (EPT) #1935." XP002434464 retrieved from EBI accession no. GSP:ABU05269 Database accession no. ABU05269 *
DE MARCO FEDERICO ET AL: "DNA vaccines against HPV-16 E7-expressing tumour cells." ANTICANCER RESEARCH, vol. 23, no. 2B, March 2003 (2003-03), pages 1449-1454, XP009084130 ISSN: 0250-7005 *
HOLST P J ET AL: "'Molecular modification of antigens delivered by replication deficient adenovirus greatly improves immunogenicity'" IMMUNOLOGY, vol. 120, no. Suppl. 1, March 2007 (2007-03), page 13, XP009084090 & ANNUAL CONGRESS OF THE BRITISH-SOCIETY-OF-IMMUNOLOGY; GLASGOW, UK; FEBRUARY 20 -23, 2007 ISSN: 0019-2805 *
HUMBERT MARTINE ET AL: "The invariant chain induces compact forms of class II molecules localized in late endosomal compartments" EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 23, no. 12, 1993, pages 3158-3166, XP002434456 ISSN: 0014-2980 *
MALCHEREK G ET AL: "MHC CLASS II-ASSOCIATED INVARIANT CHAIN PEPTIDE REPLACEMENT BY T CELL EPITOPES: ENGINEERED INVARIANT CHAIN AS A VEHICLE FOR DIRECTED AND ENHANCED MHC CLASS II ANTIGEN PROCESSING AND PRESENTATION" EUROPEAN JOURNAL OF IMMUNOLOGY, WEINHEIM, DE, vol. 28, no. 5, May 1998 (1998-05), pages 1524-1533, XP001206094 ISSN: 0014-2980 *
NAGATA T ET AL: "Induction of a single helper T-cell epitope-specific T cells by Th-oriented minigene DNA vaccine against Listeria monocytogenes." ABSTRACTS OF THE GENERAL MEETING OF THE AMERICAN SOCIETY FOR MICROBIOLOGY, vol. 103, 2003, pages E-022 URL, XP009084146 & 103RD AMERICAN SOCIETY FOR MICROBIOLOGY GENERAL MEETING; WASHINGTON, DC, USA; MAY 18-22, 2003 ISSN: 1060-2011 *
VAN TIENHOVEN E A E ET AL: "Induction of antigen specific CD4+ T cell responses by invariant chain based DNA vaccines" VACCINE, BUTTERWORTH SCIENTIFIC. GUILDFORD, GB, vol. 19, no. 11-12, 8 December 2001 (2001-12-08), pages 1515-1519, XP004313967 ISSN: 0264-410X *

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009014702A2 (en) * 2007-07-25 2009-01-29 Board Of Regents, The University Of Texas System Cd74 for use in the treatment of cancer and liver toxicity
WO2009014702A3 (en) * 2007-07-25 2009-04-16 Univ Texas Cd74 for use in the treatment of cancer and liver toxicity
EP2865387A2 (en) 2008-11-21 2015-04-29 Københavns Universitet (University Of Copenhagen) Priming of an immune response
EP3552622A3 (en) * 2008-11-21 2019-12-04 Københavns Universitet (University Of Copenhagen) Priming of an immune response
CN102292102A (en) * 2008-11-21 2011-12-21 哥本哈根大学 Priming of an immune response
EP3552622A2 (en) 2008-11-21 2019-10-16 Københavns Universitet (University Of Copenhagen) Priming of an immune response
CN107090472A (en) * 2008-11-21 2017-08-25 哥本哈根大学 The initiation of immune response
WO2010057501A1 (en) * 2008-11-21 2010-05-27 Københavns Universitet (University Of Copenhagen) Priming of an immune response
JP2017131220A (en) * 2008-11-21 2017-08-03 ケベンハウン ユニバーシテッド(ユニバーシティー オフ コペンハーゲン) Priming of immune response
EP2865387A3 (en) * 2008-11-21 2016-02-24 Københavns Universitet (University Of Copenhagen) Priming of an immune response
AU2015227479B2 (en) * 2008-11-21 2017-05-04 Københavns Universitet (University Of Copenhagen) Priming of an immune response
WO2011114346A1 (en) 2010-03-18 2011-09-22 Chetan Balar Chitin and related compounds for use in treating bacterial and viral infections
WO2014141176A1 (en) 2013-03-15 2014-09-18 Okairos Ag Improved poxviral vaccines
US10588961B2 (en) 2013-03-15 2020-03-17 Glaxosmithkline Biologicals Sa Poxviral vaccines
JP2016510989A (en) * 2013-03-15 2016-04-14 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 Improved poxvirus vaccine
US20160000904A1 (en) * 2013-03-15 2016-01-07 Glaxosmithkline Biologicals Sa Improved poxviral vaccines
WO2014139587A1 (en) 2013-03-15 2014-09-18 Okairòs Ag Improved poxviral vaccines
US10576143B2 (en) * 2013-03-15 2020-03-03 Glaxosmithkline Biologicals Sa Poxviral vaccines
US11278614B2 (en) 2013-03-15 2022-03-22 Glaxosmithkline Biologicals Sa Poxviral vaccines
CN103966219B (en) * 2014-05-26 2016-08-10 武汉大学 The structure of the shRNA of the Inhibit the replication of HIV-1 that Human foamy spumavirus is carrier mediated and application
CN103966219A (en) * 2014-05-26 2014-08-06 武汉大学 Construction and application of human foamy virus vector mediated shRNA for inhibiting HIV-1 replication
WO2017025782A1 (en) 2014-09-17 2017-02-16 Glaxosmithkline Biologicals Sa Improved poxviral vaccines
WO2018037045A1 (en) 2016-08-23 2018-03-01 Glaxosmithkline Biologicals Sa Fusion peptides with antigens linked to short fragments of invariant chain (cd74)
WO2018060288A1 (en) 2016-09-29 2018-04-05 Glaxosmithkline Biologicals S.A. Compositions and methods of treatment of persistent hpv infection
WO2018104911A1 (en) 2016-12-09 2018-06-14 Glaxosmithkline Biologicals Sa Adenovirus polynucleotides and polypeptides
US11813327B2 (en) 2017-03-20 2023-11-14 University Of Copenhagen Ii vaccine adjuvant
US11179461B2 (en) 2017-03-20 2021-11-23 University Of Copenhagen Ii vaccine adjuvant
WO2019012091A1 (en) * 2017-07-12 2019-01-17 Nouscom Ag Neoantigen vaccine composition for treatment of cancer
CN111093699A (en) * 2017-07-12 2020-05-01 Nouscom股份公司 Novel antigenic vaccine compositions for the treatment of cancer
WO2019115817A2 (en) 2017-12-15 2019-06-20 Glaxosmithkline Biologicals Sa Hepatitis b immunisation regimen and compositions
WO2019115816A1 (en) 2017-12-15 2019-06-20 Glaxosmithkline Biologicals Sa Hepatitis b immunisation regimen and compositions
US20210008195A1 (en) * 2017-12-20 2021-01-14 Zoetis Services Llc Vaccines against hendra and nipah virus infection
WO2019239311A1 (en) 2018-06-12 2019-12-19 Glaxosmithkline Biologicals Sa Adenovirus polynucleotides and polypeptides
EP3737411A4 (en) * 2018-08-10 2022-01-05 NantBio, Inc. Cellular adjuvants for viral infection
US11298414B2 (en) 2018-08-10 2022-04-12 Nantbio, Inc. Cellular adjuvants for viral infection
WO2020128012A1 (en) 2018-12-21 2020-06-25 Glaxosmithkline Biologicals Sa Methods of inducing an immune response
WO2020178359A1 (en) 2019-03-05 2020-09-10 Glaxosmithkline Biologicals Sa Hepatitis b immunisation regimen and compositions
WO2022051866A1 (en) * 2020-09-11 2022-03-17 The University Of British Columbia Vaccine for viral pathogens
EP4137153A1 (en) 2021-08-18 2023-02-22 Sirion Biotech GmbH Therapeutic papilloma virus vaccines
WO2023021116A1 (en) 2021-08-18 2023-02-23 Sirion Biotech Gmbh Therapeutic papilloma virus vaccines

Also Published As

Publication number Publication date
PT1957528E (en) 2013-01-09
EP1957528B1 (en) 2012-11-07
ES2396437T3 (en) 2013-02-21
WO2007062656A3 (en) 2008-05-22
DK1957528T3 (en) 2012-12-17
CN105039372A (en) 2015-11-11
US20100278904A1 (en) 2010-11-04
SI1957528T1 (en) 2013-02-28
US20210230633A1 (en) 2021-07-29
US20150368672A1 (en) 2015-12-24
EP1957528A2 (en) 2008-08-20
PL1957528T3 (en) 2013-03-29
CN101321781A (en) 2008-12-10
CN101321781B (en) 2018-06-29
HK1215052A1 (en) 2016-08-12

Similar Documents

Publication Publication Date Title
US20210230633A1 (en) Nucleotide vaccine
US20210386842A1 (en) Priming of an immune response
US8470560B2 (en) CR-2 binding peptide P28 as molecular adjuvant for DNA vaccines
US20160304582A1 (en) Molecular adjuvant
US11813327B2 (en) Ii vaccine adjuvant
Neukirch et al. The potential of adenoviral vaccine vectors with altered antigen presentation capabilities
AU2015227479B2 (en) Priming of an immune response

Legal Events

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

Ref document number: 200680045006.4

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2006818135

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 4965/DELNP/2008

Country of ref document: IN

WWP Wipo information: published in national office

Ref document number: 2006818135

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12095730

Country of ref document: US