WO2004052930A2 - Epitopes uniques de ciblage - Google Patents

Epitopes uniques de ciblage Download PDF

Info

Publication number
WO2004052930A2
WO2004052930A2 PCT/DK2003/000859 DK0300859W WO2004052930A2 WO 2004052930 A2 WO2004052930 A2 WO 2004052930A2 DK 0300859 W DK0300859 W DK 0300859W WO 2004052930 A2 WO2004052930 A2 WO 2004052930A2
Authority
WO
WIPO (PCT)
Prior art keywords
binding protein
chimeric binding
receptor
animal
protein
Prior art date
Application number
PCT/DK2003/000859
Other languages
English (en)
Other versions
WO2004052930A3 (fr
Inventor
Søren MOURITSEN
Original Assignee
Pharmexa A/S
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 Pharmexa A/S filed Critical Pharmexa A/S
Priority to EA200500943A priority Critical patent/EA200500943A1/ru
Priority to CA002505870A priority patent/CA2505870A1/fr
Priority to MXPA05006113A priority patent/MXPA05006113A/es
Priority to AU2003285280A priority patent/AU2003285280A1/en
Priority to EP03778264A priority patent/EP1572740A2/fr
Publication of WO2004052930A2 publication Critical patent/WO2004052930A2/fr
Publication of WO2004052930A3 publication Critical patent/WO2004052930A3/fr
Priority to NO20053363A priority patent/NO20053363L/no

Links

Classifications

    • 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
    • C07K14/001Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof by chemical synthesis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/33Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Clostridium (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/34Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Corynebacterium (G)
    • 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
    • C07K14/44Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from protozoa
    • C07K14/445Plasmodium
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/525Tumour necrosis factor [TNF]
    • 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
    • C07K14/70546Integrin superfamily
    • C07K14/70553Integrin beta2-subunit-containing molecules, e.g. CD11, CD18
    • 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
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • 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
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • 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
    • C07K14/76Albumins
    • 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
    • C07K14/79Transferrins, e.g. lactoferrins, ovotransferrins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/64Medicinal preparations containing antigens or antibodies characterised by the architecture of the carrier-antigen complex, e.g. repetition of carrier-antigen units
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the present invention relates to the field of active specific immunotherapy ("therapeutic vaccination").
  • the present invention relates to novel immunogenic agents that are capable of inducing specific B-cell immunity that is directed against one single epitope present in a self-antigen.
  • the invention further relates to methods of preparing such immunogens, methods of using the immunogens in therapy.
  • the invention relates to proteins, nucleic acid fragments, recombinantly modified host cells and virus that are useful in the practice of the methods of the invention.
  • Active specific immunotherapy also known as therapeutic vaccination
  • targets weak antigens and self-proteins has attained increased interest in the last two decades.
  • the idea of stimulating the immune system to target antigens relevant for a pathological state includes many attractive aspects in terms of patient compliance, safety and efficacy.
  • therapeutic vaccines have been prepared as chemical conjugates between an antigen and a large foreign carrier protein, where the latter provides for T-cell epitopes that are recognised by the immune system as being foreign.
  • a refinement of this approach utilises fusion molecules where the fusion partners are constituted by the antigen (normally proteina- ceous) and the foreign carrier.
  • a related approach has been to couple a single defined supposed B-cell epitope (in the form of a short peptide) to either a single, broadly recognized, T helper epitope or to a carrier protein (the "hapten-carrier” approach).
  • These approaches both suffer the drawback that the immune response against the single B-cell epitope is very hard to control, since the native conforma- tion of a peptide stretch derived from a larger protein is very difficult to maintain.
  • such peptide vaccines have met a lot of problems due to the weak immune responses induced.
  • Another approach has been to prepare anti-idiotypic antibodies raised against antibodies that recognize an antigen it is of interest to down-regulate. To a large extent, this approach resembles the hapten-carrier approach where the non-idiotypic regions of the anti-idiotypic antibody serves as a carrier moiety, and where the idiotype of the anti-idiotypic antibody is a 3- dimensional mimic of an epitope of the antigen of interest.
  • the anti-idiotypic antibody is either foreign to the vaccinated animal, so the immune response will over time be directed towards the non-essential parts of the anti-idiotypic antibody, or the antibody is self and therefore the immune response will be very weak.
  • WO 99/25378 it has been suggested to graft one member of a binding pair into the CDR (complementarity determining region) of an antibody and use this engineered antibody as an immunization agent - for all practical purposes, such an engineered antibody will share the same features as does a anti-idiotypic antibody.
  • WO 99/25378 mentions no specifics that relate to the problems associated with using autologous vs. heterologous antibodies as recipients for the grafted molecule, i.e. there is no discussion of the problems of carrier suppression when using a heterologous antibody for vaccination or lack of immunogenicity when using an autologous antibody.
  • the antigen may be a membrane protein where only minor parts are exposed to the extracellular phase and thereby accessible to antibody binding. Also, if it is of great concern to avoid activation of secondary immune mechanisms (complement, NK activity etc.) it is not suitable to direct a polyclonal antibody response against the antigen.
  • certain antigens must for safety reasons be targeted in specific epitopic regions to avoid safety issues - for instance, IgE can be safely targeted in the Fc ⁇ R binding region, whereas it is controversial whether other parts of the molecule can be targeted in a clinical setting.
  • Such antigens can be targeted with specific passive immunotherapy, where e.g. monoclonal antibodies are administered to the individual in need thereof - monoclonal antibodies only recognize one single epitope, and they are not capable of triggering secondary immune responses.
  • monoclonal antibodies are administered to the individual in need thereof - monoclonal antibodies only recognize one single epitope, and they are not capable of triggering secondary immune responses.
  • infusion of monoclonals has to be done during hospitalisation, and the amounts administered are often in the order of grams of protein per treatment. The costs involved in treatment with monoclonal antibodies are thus a considerable concern.
  • abundant self-proteins such as autologous immunoglobulins
  • carriers that are able to display single 3D structures that correspond to or are identical to B-cell epitopes of antigens. Since such modified, abundant self-proteins are not normally capable of exhibiting sufficient immu- nogenicity so as to give rise to an effective immune response towards the B-cell epitope introduced in the scaffold structure, they are according to the invention engineered to also include foreign T helper epitopes that stimulate the necessary T-cell help for B-cells producing autoreactive antibodies.
  • the present invention relates to a chimeric binding protein that is immunogenic in an animal, said chimeric binding protein being one that binds specifically to a first receptor, said first receptor being one that binds a second receptor present in an antigen of said animal, wherein said chimeric binding protein comprises: - a B-cell epitope in the form of a binding site that specifically binds the first receptor and which competes with the second receptor for binding to the first receptor,
  • scaffold protein structure that stabilises the 3D conformation of the binding site, said scaffold protein structure being autologous in said mammal, and
  • the present invention also relates to a nucleic acid fragment that encodes the chimeric protein of the present invention.
  • a third aspect of the present invention relates to a method of preparing the chimeric binding protein of the present invention.
  • Yet another aspect of the present invention relates to a vector that is capable of expressing the nucleic acid fragment of the invention.
  • a further aspect of the present invention relates to a method of immunizing against a self- antigen by administering an immunogenic agent of the invention so as to provoke an immune response against an epitope of said antigen.
  • Fig. 1 Schematic depiction of a filamentous phage.
  • the figure shows a filamentous phage with its protein sheath that is made up of five different coating proteins (pill, pVI, pVII, pVIII and pIX).
  • Fig. 2 Schematic depiction of phage display technology.
  • One round of panning consists of: Superinfection of the E. coli antibody library, production of recombinant phage particles, incubation of phage with the antigen, removal of non-binding phage by a washing procedure, elution of bound phage particles and transduction of fresh E. coli cells with the eluted phage.
  • a "chimeric binding protein” is in the present context intended to mean a protein/polypeptide that includes a combination of amino acid sequences not found in association in the same molecule in nature. More specifically, the chimeric binding protein includes at least an amino acid sequence from a mammalian protein (the scaffold or carrier) and a foreign T H epitope (foreign in the sense that it is not naturally associated with the remainder of the chimeric binding protein). Further, the term implies that the chimeric binding protein includes a struc- ture that functions as a binding site. This binding site may be a natural part of the scaffold (in the case of an anti-idiotypic antibody) or it may be a binding site which is grafted onto or introduced into the scaffold.
  • immunogen in the present context refers to an agent (a substance or a composition of matter) that induces an immune response. It will be understood that certain molecules (e.g. traditional small haptens or self-proteins that are tolerated in the autologous host) are incapable of inducing an immune response.
  • T-lymphocyte and "T-cell” will be used interchangeably for lymphocytes of thymic origin which are responsible for various cell mediated immune responses as well as for helper activity in the humeral immune response.
  • B-lymphocyte and “B-cell” will be used interchangeably for antibody-producing lymphocytes.
  • polypeptide is in the present context intended to mean both short peptides of from 2 to 10 amino acid residues, oligopeptides of from 11 to 100 amino acid residues, and polypeptides of more than 100 amino acid residues. Furthermore, the term is also intended to include proteins, i.e. functional biomolecules comprising at least one polypeptide; when com- prising at least two polypeptides, these may form complexes, be covalently linked, or may be non-covalently linked.
  • the polypeptide(s) in a protein can be glycosylated and/or lipidated and/or comprise prosthetic groups. Also, the term "polyamino acid” is an equivalent to the term "polypeptide".
  • sequence means any consecutive stretch of at least 3 amino acids or, when relevant, of at least 3 nucleotides, derived directly from a naturally occurring amino acid sequence or nucleic acid sequence, respectively.
  • animal is in the present context in general intended to denote an animal species (preferably mammalian), such as Homo sapiens, Cam ' s domesticus, etc. and not just one single animal. However, the term also denotes a population of such an animal species, since it is important that the individuals immunized according to the method of the invention all harbour substantially the same a allowing for immunization of the animals with the same immuno- gen(s).
  • an animal in the present context is a living being which has an im- mune system. It is preferred that the animal is a vertebrate, such as a mammal.
  • ⁇ vivo down-regulation of antigen activity is herein meant reduction in the living organism of the number of interactions between the antigen in question and its recep- tors/ligands or down-regulation of any other measurable physiological activity exerted by the antigen.
  • the down-regulation can be obtained by means of several mechanisms: Of these, simple interference with the active site in an antigen by antibody binding is the most simple. However, it is also within the scope of the present invention that the antibody binding results in removal of antigen by scavenger cells (such as macrophages and other phagocytic cells). Another possibility is binding of antibodies to the antigen that are capable of interfering with the normal cleavage of a propeptide into a mature polypeptide.
  • effecting presentation ... to the immune system is intended to denote that the animal's immune system is subjected to an immunogenic challenge in a controlled manner.
  • challenge of the immune system can be effected in a number of ways of which the most important are vaccination with polypeptide containing "pharmaccines” (i.e. a vaccine which is administered to treat or ameliorate ongoing disease) or nucleic acid "pharmaccine” vaccination, but also live and viral vaccines are within the scope of the present invention.
  • the important result to achieve is that immune competent cells in the animal are confronted with the antigen in an immunologically effective manner, whereas the precise mode of achieving this result is of less importance to the inventive idea underlying the present invention.
  • immunologically effective amount has its usual meaning in the art of immunology, i.e. an amount of an immunogen which is capable of inducing an immune response which significantly engages molecules which share immunological features with the immunogen.
  • a protein has been "modified" is herein meant a chemical modification of the polypeptide which constitutes the backbone of the relevant protein that forms the scaffold part of a chimeric binding molecule of the present invention.
  • a modifi- cation can e.g. be derivatization (e.g. alkylation, acylation, esterification etc.) of certain amino acid residues in the scaffold's polypeptide sequence, but as will be appreciated from the disclosure below, the preferred modifications comprise changes of (or additions to) the primary structure of the amino acid sequence.
  • the animal's tolerance towards the scaffold protein structure is at a higher level than the tolerance towards the B-cell epitope part of the chimeric binding protein - in fact, the tolerance against the scaffold protein part is so high that no significant immunological effects are brought about by antibodies cross-reacting with the animal's naturally occurring scaffold protein.
  • a “foreign T-cell epitope” is a peptide which is able to bind to an MHC molecule and which stimulates T-cells in an animal species.
  • Preferred foreign T-cell epitopes in the invention are "promiscuous" epitopes, i.e. epitopes which bind to a substantial fraction of a particular class of MHC molecules in an animal species or population. Only a very limited number of such promiscuous T-cell epitopes are known, and they will be discussed in detail below.
  • a "foreign T helper lymphocyte epitope” (a foreign T H epitope) is a foreign T cell epitope which binds an MHC Class II molecule and can be presented on the surface of an antigen presenting cell (APC) bound to the MHC Class II molecule.
  • APC antigen presenting cell
  • the term “foreign” means that the T H epitope is foreign to the immunized animal but also that the T H epitope is not naturally associated with the scaffold protein structure that forms part of the chimeric binding protein of he invention. It will be understood that a “tolerance breaking amino acid sequence" includes at least one such foreign T H epitope.
  • a "functional part" of a (bio)molecule is in the present context intended to mean the part of the molecule which is responsible for at least one of the biochemical or physiological effects exerted by the molecule. It is well-known in the art that many enzymes and other effector molecules have an active site which is responsible for the effects exerted by the molecule in question. Other parts of the molecule may serve a stabilizing or solubility enhancing purpose and can therefore be left out if these purposes are not of relevance in the context of a certain embodiment of the present invention. For instance it is possible to use certain other cytokines as a modifying moiety in the chimeric binding protein (cf.
  • the issue of stability may be irrelevant since the incorporation into the chimeric binding protein provides the stability necessary.
  • the functional part is herein intended to be a fragment of the antibody, which is capable of exerting binding to the relevant epitope for that antibody.
  • adjuvant has its usual meaning in the art of vaccine technology, i.e. a substance or a composition of matter which is 1) not in itself capable of mounting a specific immune response against the immunogen of the vaccine, but which is 2) nevertheless capable of enhancing the immune response against the immunogen. Or, in other words, vaccination with the adjuvant alone does not provide an immune response against the immunogen, vaccina- tion with the immunogen may or may not give rise to an immune response against the immunogen, but the combination of vaccination with immunogen and adjuvant induces an immune response against the immunogen which is stronger than that induced by the immunogen alone.
  • Targeting of a molecule is in the present context intended to denote the situation where a molecule upon introduction in the animal will appear preferentially in certain tissue(s) or will be preferentially associated with certain cells or cell types.
  • the effect can be accomplished in a number of ways including formulation of the molecule in composition facilitating targeting or by introduction in the molecule of groups which facilitates targeting.
  • “Stimulation of the immune system” means that a substance or composition of matter exhibits a general, non-specific immunostimulatory effect. A number of adjuvants and putative adjuvants (such as certain cytokines) share the ability to stimulate the immune system. The result of using an immunostimulating agent is an increased "alertness" of the immune system meaning that simultaneous or subsequent immunization with an immunogen induces a signifi- cantly more effective immune response compared to isolated use of the immunogen. "Productive binding” means binding of a peptide to the MHC molecule (Class I or II) so as to be able to stimulate T-cells that engage a cell that present the peptide bound to the MHC molecule. For instance, a peptide bound to an MHC Class II molecule on the surface of an APC is said to be productively bound if this APC will stimulate a T H cell that binds to the presented peptide-MHC Class II complex.
  • the present invention has been conceived based on the knowledge that a number of specific binding agents (monoclonal antibodies, soluble receptors, small molecule inhibitors etc.) are accepted in the art of therapy as being safe and efficacious.
  • these agents are Remi- cade® (Infliximab, an anti-TNF mAB), Herceptin® (Trastuzumab, an anti-HER-2 mAB), Xolair® (Omalizumab, an anti-IgE mAB), and Retuximab (an anti-CD20 mAB).
  • Remi- cade® Infliximab, an anti-TNF mAB
  • Herceptin® Trastuzumab, an anti-HER-2 mAB
  • Xolair® Opmalizumab, an anti-IgE mAB
  • Retuximab an anti-CD20 mAB.
  • most of these specific binding agents need to be administered frequently and under medical supervision and control in order
  • the preferred antigens against which it is desired to immunize are of human origin, i.e. that the B-cell epitope against which an immune response is directed is a B-cell epitope of a human antigen.
  • the technology is exercised by means of identification/isolation of 3D structures that precisely mimic (or are identical to) the structures that bind the clinical safe agent and incorporation into immunogenic variants of abundant, otherwise non-immunogenic self-proteins.
  • the present invention discloses protein constructs that are specialised variations of the immunogenic self-proteins of WO 95/05849 but an important difference is that the immune response against the self-protein part is insignificant, cf. below.
  • the present chimeric binding proteins are effective in raising immune responses against single B-cell epitopes of self-antigens for the following reasons:
  • an anti-idiotypic IgG antibody it is ob- served that an immune response is generated against the B-cell epitope constituted by the idiotype of the anti-idiotypic antibody.
  • the remainder of the anti-idiotypic antibody is not autologous in the vaccinated animal, that part of the molecule will act as a traditional carrier protein, and hence a strong immune response will be generated against that part of the molecule.
  • this autologous self-protein (IgG) is selected so as to be an abundant self-protein in the serum of the vaccinated individual.
  • the immune system will therefore not be capable of raising a physiologically relevant response against the antibody, either because the immune system is completely incapable of raising a B-cell response against such proteins due to B-cell anergy or, alternatively, antibody production is actually induced, but due to the abundance of the autologous antibody structure, the induced antibodies that do not react with the idiotype are "absorbed" due to binding to the large amounts of IgG in serum.
  • the only relevant and effective immune re- sponse will be that directed against the idiotype of the anti-idiotypic antibody, since this part of the antibody response is not "diluted" by reactivity with the abundant serum protein.
  • the present approach is not limited to the use of such immunoge- nized autologous anti-idiotypic antibodies.
  • Other abundant self-proteins may serve as a carrier structure for the B-cell epitope it is of interest to immunize against - the importance is here that either no antibodies can be induced against the abundant self-protein, or, if antibodies are induced against the abundant self-protein, these should not be capable of exerting adverse effects due to their binding to the abundant self-protein in vivo - in other words, the concentration of the self-protein should be so high that it can absorb antibodies that cross- react with regions outside the B-cell epitope of interest.
  • the scaffold protein structure serves two purposes. First, the scaffold protein has to stabilise the native 3D conformation of the B-cell epitope against which the relevant immune response is intended to be raised. Second, when immunising with the chimeric binding protein, the antibodies induced against the scaffold protein structure (if any are raised at all) should be incapable of giving rise to adverse effects.
  • the scaffold protein structure It will normally be feasi- ble to utilise an exposed loop structure or an active site in the scaffold protein structure.
  • the complementarity determining regions (CDRs) of an antibody are kept in their correct conformation by parts of invariable parts of the heavy and light chains of an antibody, and the same holds true for active sites in enzymes and other molecules having active or binding sites.
  • the scaffold protein structure is preferred to use such ones that are derived from abundant proteins, meaning that if any antibodies against such scaffold protein structures are produced, these antibodies will be absorbed by the large amounts of scaffold protein that is naturally occurring in the immunized individual.
  • the scaffold protein structure is derived from an abun- dant serum protein.
  • the normal serum concentration of an abundant serum protein is at least 0.1 g per 100 ml serum. It is even more preferred that the normal serum concentration of the abundant serum protein is at least 0.3 g per 100 ml serum, such as at least 0.4, at least 0.6, at least 0.8, at least 1.0, at least 1.2, at least 1.4, and at least 1.5 g per 100 ml serum. Even higher values are preferred when the abundant serum protein is albumin: at least 2.0 g per 100 ml, at least 2.5, at least 3.0, at least 3.5, and at least 4.0 g per 100 ml serum.
  • especially interesting scaffold protein structures are derived from albumin, an immu- noglobulin (such as IgG and IgA), transferrin, ⁇ 2-macroglobulin, haptaglobin, ⁇ i-lipoprotein, ⁇ i-lipoprotein, all of which are proteins having a high serum concentration.
  • an immu- noglobulin such as IgG and IgA
  • transferrin ⁇ 2-macroglobulin
  • haptaglobin ⁇ i-lipoprotein
  • ⁇ i-lipoprotein proteins having a high serum concentration.
  • the chimeric binding protein is derived from an antibody molecule such as IgG.
  • the scaffold protein structure will then typically be derived from the non- idiotypic region of the molecule, La. from the non-idiotypic region of a complete antibody or from a fragment thereof such as an F(ab')2 fragment, an Fab fragment, and an scFv.
  • the scaffold protein structure comprises a substantially complete amino acid sequence of a polypeptide autologous in said animal, e.g.
  • the scaffold protein structure comprises a substantial number of B-cell epitopes found in the autologous scaffold protein structure in the animal. It is most preferred that the scaffold protein structure has substantially the same tertiary (and, if relevant, quaternary) structure of a polypeptide autologous in said animal. Again, if the scaffold protein structure is derived from an antibody, this will entail that the chimeric binding protein includes a substantially complete antibody molecule within its structure.
  • the scaffold protein structure is an IgG molecule
  • This is e.g. of interest when the target for immunization with the chimeric binding protein is IgE.
  • this precaution is of less relevance, since the clinically relevant antibodies induced by the chimeric binding proteins of the present invention will never bind simultaneously to the same antigen (unless the epitope is of repetitive nature as in certain carbohydrates) - only one single epitope is bound. Since complement fixation and activation requires binding of more than one antibody, this and other secondary effector mechanisms are not believed to be an issue.
  • the scaffold protein structure serves the purpose of presenting the B-cell epitope of interest in a spatial conformation that is quasi-identical or identical to an antigenic determinant in an antigen found in the animal to be vaccinated.
  • One way of achieving this goal is to graft an identified B-cell epitope of an antigen into a known presentation structure or scaffold.
  • CDRs complementarity determining regions
  • these can e.g. be humanized using a variety of techniques including CDR- grafting (EP 0 239 400; WO 91/09967; US 5,530,101; and US 5,585,089), veneering or resurfacing (EP 0 592 106; EP 0 519 596; Padlan E.
  • the B-cell epitope may be any amino acid sequence that can be isolated according to the methods of the invention, cf. below.
  • the invention relies in part on the use of anti-idiotypic antibodies, where the idiotype constitutes the B-cell epitope part of the chimeric binding protein, but other abundant serum proteins can be randomly modified in an exposed loop structure and screened according to the invention so as to isolate suitable binding species.
  • the inventive screening methods of the invention it is ensured that the B-cell epitope part of the chimeric binding protein is indeed presented in a suitable conformation, and it is hence not necessary to rely on introduction of the B-cell epitope in a scaffold that may or may not be capable of presenting the B-cell epitope in a satis- factory manner.
  • the chimeric binding protein of the invention preferably contains the B-cell epitope which is constituted by the idiotype of an antibody.
  • an idiotype of an antibody has a defined 3D conformation that is known to bind to an epitope. Further, if the idiotype of the antibody is reactive with a different antibody's idiotype, then the antibody's idiotype will mimic the antigenic epitope against which the different antibody reacts. Hence, there is no need to test whether or not the epitope in question will be presented sufficiently well in a chosen scaffold protein structure, and it does not matter whether the epitope is regional or assembled topographic in nature.
  • anti-idiotypic antibodies have already been demonstrated to be capable of functioning as vaccination agents so as to raise an immune response against the idiotype.
  • the chimeric binding protein of the invention is one wherein said first receptor is the idiotype of an antibody or a specific binding region of a ligand that binds the second receptor in the animal to be immunized.
  • the chimeric binding protein of the invention is one, wherein said first receptor is the idiotype of a monoclonal antibody.
  • the tolerance breaking amino acid sequence may be situated in any suitable position in the chimeric binding protein.
  • it may be introduced in the form of a simple fusion partner or conjugation partner to the scaffold protein structure and in these embodiments the tolerance breaking amino acid sequence may constitute a complete protein. Technologies relevant for this are well-known in the art and include chemical coupling of carrier proteins to antigens by means of glutaraldehyde..
  • the tolerance breaking amino acid sequence is introduced by means of amino acid insertion or substitution in the amino acid sequence of the scaffold protein structure.
  • An especially preferred chimeric binding protein of the invention is an anti- idiotypic antibody or an effectively binding fragment thereof that is modified so as to include said tolerance breaking amino acid sequence, where said anti-idiotypic antibody is autologous in the animal to be immunized.
  • an tolerance breaking amino acid sequence can be accomplished by introduction of at least one amino acid insertion, addition, deletion, or substitution in the scaffold protein structure.
  • the normal situation will be the introduc- tion of more than one change in the amino acid sequence (e.g. insertion of or substitution by a complete T-cell epitope) but the important goal to reach is that the chimeric binding protein, when processed by an antigen presenting cell (APC), will give rise to a tolerance breaking amino acid sequence being presented in context of an MCH Class II molecule on the surface of the APC.
  • APC antigen presenting cell
  • the amino acid sequence of the scaffold protein structure in appropriate po- sitions comprises a number of amino acid residues which can also be found in a tolerance breaking amino acid sequence
  • the introduction of such a tolerance breaking amino acid sequence can be accomplished by providing the remaining amino acids of the foreign epitope by means of amino acid insertion, addition, deletion and substitution.
  • the number of amino acid insertions, deletions, substitutions or additions is at least 2, such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 and 25 insertions, substitutions, additions or deletions. It is furthermore preferred that the number of amino acid insertions, substitutions, additions or deletions is not in excess of 150, such as at most 100, at most 90, at most 80, and at most 70. It is especially preferred that the number of substitutions, insertions, deletions, or additions does not exceed 60, and in particular the number should not exceed 50 or even 40. Most preferred is a number of not more than 30. With respect to amino acid additions, it should be noted that these, when the resulting construct is in the form of a fusion polypeptide, is often considerably higher than 150.
  • Preferred embodiments of the invention include introducing at least one immunodominant tolerance breaking amino acid sequence (in this specification also denoted "a foreign immune dominant T H epitope").
  • a foreign immune dominant T H epitope a foreign immune dominant T H epitope
  • immunodomi- nance simply refers to epitopes which in the vaccinated individual/population gives rise to a significant immune response, but it is a well-known fact that a T H epitope which is immunodominant in one individual/population is not necessarily immunodominant in another individual of the same species, even though it may be capable of binding MHC-II molecules in the latter individual.
  • an immune dominant T H epitope is a T H epitope which will be effective in providing T-cell help when present in an antigen.
  • immune dominant T H epitopes have as an inherent feature that they will substantially always be presented bound to an MHC Class II molecule, irrespective of the polypeptide wherein they appear.
  • T H epitopes Another important point is the issue of MHC restriction of T H epitopes.
  • naturally occurring T H epitopes are MHC restricted, i.e. a certain peptide constituting a T H epitope will only bind effectively to a subset of MHC Class II molecules. This in turn has the effect that in most cases the use of one specific T H epitope will result in a vaccine component which is only effective in a fraction of the population, and depending on the size of that fraction, it can be necessary to include more T H epitopes in the same molecule, or alternatively prepare a multi- component vaccine wherein the components are variants chimeric binding protein which are distinguished from each other by the nature of the T H epitope introduced.
  • the fraction of the population covered by a specific vaccine composition can be determined by means of the following formula
  • n is the total number of foreign T H epitopes in the vaccine composition.
  • ⁇ ⁇ is the sum of frequencies in the population of allelic haplotypes encoding MHC molecules which bind any one of the T H epitopes in the vaccine and which belong to the h of the 3 known HLA loci (DP, DR and DQ); in practice, it is first determined which MHC molecules will recognize each T H epitope in the vaccine and thereafter these are listed by type (DP, DR and DQ) - then, the individual frequencies of the different listed allelic haplotypes are summed for each type, thereby yielding ⁇ _, ⁇ 2 , and ⁇ 3 .
  • u 3 is the sum of frequencies in the population of allelic haplotype encoding MHC molecules which bind the / th T H epitope in the vaccine and which belong to the h of the 3 known HLA loci (DP, DR and DQ).
  • DP allelic haplotype encoding MHC molecules which bind the / th T H epitope in the vaccine and which belong to the h of the 3 known HLA loci (DP, DR and DQ).
  • tolerance breaking amino acid sequences to be introduced in the chimeric binding protein it is important to include all knowledge of the epitopes which is available: 1) The frequency of responders in the population to each epitope, 2) MHC restriction data, and 3) frequency in the population of the relevant haplotypes.
  • T H epitopes which are active in a large proportion of individuals of an animal species or an animal population and these are preferably introduced in the vaccine thereby reducing the need for a very large number of different analogues in the same vaccine.
  • the promiscuous epitope can according to the invention be a naturally occurring human T H epitope such as epitopes from tetanus toxoid (e.g. the P2 and P30 epitopes), diphtheria toxo- id, Influenza virus hemagluttinin (HA), and P. falclparum CS antigen.
  • tetanus toxoid e.g. the P2 and P30 epitopes
  • diphtheria toxo- id e.g. the P2 and P30 epitopes
  • HA Influenza virus hemagluttinin
  • P. falclparum CS antigen a naturally occurring human T H epitope
  • sequences of the P2 and P30 epitopes are Gln-Tyr-Ile-Lys-Ala-Asn-Ser-Lys-Phe-Ile-Gly-Ile-Thr-Glu-Leu (SEQ ID NO: 1) and Phe-Asn-Asn-Phe-Thr-Val-Ser-Phe-Trp-Leu-Arg-Val-Pro-Lys-Val-Ser-Ala-Ser-His- Leu-Glu (SEQ ID NO: 2), respetively.
  • T H epitopes Over the years a number of other promiscuous T H epitopes have been identified. Especially peptides capable of binding a large proportion of HLA-DR molecules encoded by the different HLA-DR alleles have been identified and these are all possible T-cell epitopes to be introduced in the analogues used according to the present invention. Cf. also the epitopes discussed in the following references which are hereby all incorporated by reference herein: WO 98/23635 (Frazer IH et al., assigned to The University of Queensland); Southwood S et al., 1998, J. Immunol.
  • the tolerance breaking amino acid sequence can be any artificial T H epitope, which is capable of binding a large proportion of MHC Class II molecules.
  • the pan DR epitope peptides PADRE
  • PADRE pan DR epitope peptides
  • WO 95/07707 and in the corresponding paper Alexander J et al., 1994, Immunity 1: 751-761 (both disclosures are incorporated by reference herein) are interesting candidates for tolerance breaking amino acid sequences to be used according to the present invention. It should be noted that the most effective PADRE peptides disclosed in these papers carry D-amino acids in the C- and N-termini in order to improve stability when administered.
  • the present invention primarily aims at incorporating the relevant epitopes as part of the chimeric binding protein, which should then subsequently be broken down enzymatically inside the lysosomal compartment of APCs to allow subsequent presentation in the context of an MHC-II molecule and therefore it is not expedient to incorporate D-amino acids in the epitopes used in the present invention.
  • One especially preferred PADRE peptide is the one having the amino acid sequence
  • AKFVAAWTLKAAA SEQ ID NO: 3
  • SEQ ID NO: 3 an immunologically effective subsequence thereof.
  • This, and other epitopes having the same lack of MHC restriction are preferred tolerance breaking amino acid sequences which should be present in the chimeric binding proteins of the invention.
  • Such super-promiscuous epitopes will allow for the simplest embodiments of the inven- tion wherein only one single chimeric binding protein species is presented to the vaccinated animal's immune system.
  • Chimeric bin- ding proteins which react to the same extent with the antiserum as does the autologous scaffold protein structure must be regarded as having the same 3D structure as the autologous scaffold protein structure whereas chimeric molecules exhibiting a limited (but still significant and specific) reactivity with such an antiserum is regarded as having maintained a substantial fraction of the original B-cell epitopes.
  • a selection of monoclonal antibodies reactive with distinct epitopes on the autologous scaffold protein can be prepared and used as a test panel. This approach has the advantage of allowing 1) an epitope mapping of the autologous scaffold molecule and 2) a mapping of the epitopes which are maintained in the chimeric binding protein.
  • a third approach would be resolve the 3-dimensional structure of the autologous scaffold protein or of a biologically active truncate thereof (cf. above) and compare this to the resolved three-dimensional structure of the chimeric binding protein.
  • Three-dimensional structure can be resolved by the aid of X-ray diffraction studies and NMR-spectroscopy.
  • the present invention relies on the identification of especially well-suited regions for introduction in the scaffold protein structure of the foreign element that must provide for the necessary T H epitopes.
  • Especially preferred regions are flexible loop regions (which do not contribute directly to tertiary structure) as well as flexible linker regions and N or C termini.
  • the introduction of the T H epitope can be made in a region that has a secondary structure that has a high degree of similarity with the secondary structure of the epitope (an ⁇ -helical region may be substituted with an epitope that can form part of an ⁇ -helix, a ⁇ - strand region may be substituted with an epitope that can form part of a ⁇ -strand region).
  • the chimeric binding protein of the invention may also include a number of other features.
  • the chimeric binding protein includes
  • They can be present in the form of side groups attached covalently or non-covalently to suit- able chemical groups in the amino acid sequence of the scaffold protein structure. This is to mean that stretches of amino acid residues derived from the autologous scaffold protein are derivatized without altering the primary amino acid sequence, or at least without introducing changes in the peptide bonds between the individual amino acids in the chain.
  • the moieties can also be in the form of fusion partners to the amino acid sequence derived from the autologous scaffold protein and/or the tolerance breaking amino acid sequence.
  • both possibilities include the option of conjugating the amino acid sequence to a carrier, cf. the discussion of these below.
  • fusion protein is not merely restricted to a fusion construct prepared by means of expression of a DNA fragment encoding the construct but also to a conju- gate between two proteins which are joined by means of a peptide bond in a subsequent chemical reaction.
  • the chimeric binding protein can also include the introduction of a first moiety which targets the chimeric binding protein to an APC or a B-lymphocyte.
  • the first moiety can be a specific binding partner for a B-lymphocyte specific surface antigen or for an APC specific surface antigen.
  • the moiety can be a carbohydrate for which there is a receptor on the B- lymphocyte or the APC (e.g. mannan or mannose).
  • the second moiety can be a hapten.
  • an antibody fragment which specifically recognizes a surface molecule on APCs or lymphocytes can be used as a first moiety (the surface molecule can e.g.
  • FC ⁇ RI FC ⁇ RI
  • CTLA-1 CTLA-4
  • FC ⁇ RI FC ⁇ RI
  • CD40 or CTLA-4 any other specific surface marker
  • use of chimeric binding proteins of the invention that are anti-idiotypic IgG provides for such a first moiety in the form of the Fc ⁇ receptor binding region of IgG.
  • all these exemplary targeting molecules can be used as part of an adjuvant, cf. below.
  • CD40 ligand, antibodies against CD40, or variants thereof which bind CD40 will target the chimeric binding protein to dendritic cells.
  • CD40 binding molecules as the first moiety (or as adjuvants, cf. below) will enhance the CTL response considerably; in fact, the use of such CD40 binding molecules as adjuvants and "first moieties" in the meaning of the present invention is believed to be inventive in its own right.
  • chimeric binding protein As an alternative or supplement to targeting the chimeric binding protein to a certain cell type in order to achieve an enhanced immune response, it is possible to increase the level of responsiveness of the immune system by including the above-mentioned second moiety which stimulates the immune system.
  • second moieties are cytokines, heat- shock proteins, and hormones, as well as effective parts thereof.
  • Suitable cytokines to be used according to the invention are those which will normally also function as adjuvants in a vaccine composition, e.g. interferon ⁇ (IFN- ⁇ ), Flt3 ligand (Flt3L), interleukin 1 (IL-1), interleukin 2 (IL-2), interleukin 4 (IL-4), interleukin 6 (IL-6), interleukin 12 (IL-12), interleukin 13 (IL-13), interleukin 15 (IL-15), and granulocyte-macrophage colony stimulating factor (GM-CSF); alternatively, the functional part of the cytokine molecule may suffice as the second moiety.
  • IFN- ⁇ interferon ⁇
  • Flt3L Flt3 ligand
  • IL-1 interleukin 1
  • IL-2 interleukin 2
  • IL-4 interleukin 4
  • IL-6 interleukin 6
  • IL-12 interleukin 12
  • IL-13 interleukin 13
  • the second moiety can be a toxin, such as listeriolycin (LLO), lipid A and heat- labile enterotoxin.
  • LLO listeriolycin
  • lipid A lipid A
  • heat- labile enterotoxin lipid A
  • mycobacterial derivatives such as MDP (muramyl dipep- tide), CFA (complete Freund's adjuvant) and the trehalose diesters TDM and TDE are interesting possibilities.
  • suitable heat shock proteins used as the second moiety can be HSP70, HSP90, HSC70 (a heat shock cognate), GRP94, and calreticulin (CRT).
  • the possibility of introducing a third moiety that enhances the presentation of the chimeric binding protein to the immune system is an important embodiment of the invention.
  • the art has shown several examples of this principle. For instance, it is known that the palmitoyl lipidation anchor in the Borrelia burgdorferi protein OspA can be utilised so as to provide self-adjuvating polypeptides (cf. e.g. WO 96/40718). It seems that the lipidated proteins form up micelle-like structures with a core consisting of the lipidation anchor parts of the polypeptides and the remaining parts of the molecule protruding therefrom, resulting in multiple presentations of the antigenic determinants.
  • lipidation anchors e.g. a myristyl group, a famesyl group, a geranyl- geranyl group, a GPI-anchor, and an N-acyl diglyceride group
  • lipidation anchors e.g. a myristyl group, a famesyl group, a geranyl- geranyl group, a GPI-anchor, and an N-acyl diglyceride group
  • C3d fragment of complement factor C3 or C3 itself cf. Dempsey et al., 1996, Science 271, 348-350 and Lou & Kohler, 1998, Nature Biotechnology 16, 458-462).
  • the chimeric binding proteins of the present invention are especially suited for active specific immunotherapy that targets antigens that already are known as suitable targets for monoclonal antibody therapy.
  • immunoglobulin E, CD20, CDlla, beta amyloid, HER-2, and TNF ⁇ are all suitable targets for the present invention, meaning that the above-discussed second receptor is present in one of these antigens.
  • Such targets can be ones where it is desirable to only target a very specific "safe" region of the molecule (as in the case of IgE, where it is desirable to target the FC ⁇ R binding region to avoid problems with anaphylaxis induced by cross-linking anti-IgE antibodies), or it may be a defined exposed region of an antigen which is otherwise not accessible to antibodies (as with CD20, where only a minor portion is exposed to the extracellular phase.
  • Interleukin 15 / IL15 Azimi N. et al. 2001.
  • IL-15 plays a major role in the persistence of Tax-specific CD8 cells in HAM/TSP patients. Proc. Natl. Acad. Sci. USA. 98 (25): 14559- 14564.
  • Interleukin 17 / IL17 Nakae S. et al. 2003. Suppression of immune induction of collagen-induced arthritis in IL-17-deficient mice. 171(11): 6173-6177.
  • Interleukin 18 / IL18 Banda N., K. et al. 2003. Mechanisms of inhibition of collagen-induced arthritis by murine IL-18 binding protein. J. Immunol. 170: 2100-2105.
  • Interleukin 20 / IL20 Conti Conti, P. et al. 2003.
  • IL-10 subfamily members IL- 19, IL-20, IL-22, IL-24 and IL-26. Immunol. Lett. 88(3): 171-174
  • Eotaxin Ponath P., D. 1996. Cloning of the human eosinophil chemoattractant, eotaxin. Expression, receptor binding, and functional properties suggest a mechanism for the selective recruitment of eosinophils. J Clin Invest. 97(3): 604-612. Eotaxin-2 Ying, S. et al. 1999. Eosinophil chemotactic chemo- kines (eotaxin, eotaxin-2, RANTES, monocyte chemoattractant protein-3 (MCP-3), and MCP-4), and C-C chemokine receptor 3 expression in bronchial biopsies from atopic and nonatopic (Intrinsic) asthmatics. J. Immunol. 163(11): 6321-6329.
  • Monocyte chemoattractant prote- Pype J., L. et al. 1999. Expression of monocyte in-2 / MCP-2 chemotactic protein (MCP)-l, MCP-2, and MCP-3 by human airway smooth-muscle cells. Modulation by corticosteroids and T-helper 2 cytokines. Am. J. Res- pir. Cell Mol. Biol. 21(4):528-36.
  • Eosinophil chemotactic chemoki- nes eotaxin, eotaxin-2, RANTES, monocyte chemoattractant protein-3 (MCP-3), and MCP-4
  • CX3CL1 neutralrotactin; CX3C memMcDermott, D., L. et al. 2001.
  • EGF retor / EGFR ceptor family as targets for cancer therapy.
  • CD55 Spendlove, L. et al. 2000. A therapeutic human anti- idiotypic antibody mimics CD55 in three distinct regions. Eur J Immunol, 30(10): 2944-2953.
  • Tumor-associated antigen mucin-1 Carr-Brendel, V. 2000. Immunity to murine breast / MUC-1 cancer cells modified to express MUC-1, a human breast cancer antigen, in transgenic mice tolerant to human MUC-1. Cancer Res. 60(9): 2435-2443.
  • GM2 Tsuchida T. et al. 1987. Gangliosides of human melanoma. J Natl Cancer Inst. 78(1): 45-54.
  • GM3 Tsuchida T. et al. 1987. Gangliosides of human melanoma. J Natl Cancer Inst. 78(1): 45-54.
  • GD2 T suchida, T. et al. 1987. Gangliosides of human melanoma. J. Natl. Cancer Inst. 78(1): 45-54.
  • GD3 T suchia, T. et al. 1987. Gangliosides of human melanoma. J Natl Cancer Inst. 78(1): 45-54. Sialyl-Tn Miles, D. and Papazisis, K. 2003. Rationale for the clinical development of STn-KLH (Theratope®) and anti-MUC-1 vaccines in breast cancer. Clin. Breast Cancer 3(suppl.4): S134-S138.
  • EpCa m/KSA/GA73- 3/CO 17- 1 A Herlyn, M. et al. 1979. Colorectal carcinoma-specific antigen detection by means of monoclonal antibodies. Proc Natl Acad Sci 76: 1438-1452.
  • Phencyclidine Laurenzana, E., M. et al. 2003. Treatment of adverse effects of excessive phencyclidine exposure in rats with a minimal dose of monoclonal antibody. J. Pharmacol. Exp. Ther. 306(3): 1092-1098.
  • Myostatin a transforming growth factor-beta superfamily member, is expressed in heart muscle and is upregulated in cardiomyocytes after infa ret. J. Cell Physiol. 180(1): 1-9.
  • Monoclonal antibodies inhibit prion replication and delay the development of prion disease. Nature 422: 80-83.
  • Soluble IAPP Islet Amlyloid polyH ⁇ ppener, J., W. 2000. Islet amyloid and type 2 diapeptide; amylin) betes mellitus. N. Engl. J. Med. 343: 411-419.
  • Soluble amyloid oligomers of A ⁇ of Kayed, R. et al. 2003.
  • Common structure of soluble IAPP, of PrP and of polyglutamine amyloid oligomers implies common mechanism of pathogenesis. Science 300:486-489.
  • the invention also relates to a nucleic acid fragment that encodes a chimeric binding protein of the invention.
  • nucleic acid fragments are useful for recombinant preparation of the chimeric binding proteins of the invention but also as parts of immunizing agents for nucleic acid vaccination, cf. below.
  • the chimeric binding proteins can be prepared by means of recombinant gene technology but also by means of chemical synthesis or semisynthesis; the latter two options are especially relevant when the modification consists in coupling to protein carriers (such as KLH, diphtheria toxoid, tetanus toxoid, and BSA) and non-proteinaceous molecules such as carbohydrate polymers and of course also when the modification comprises addition of side chains or side groups to an polypeptide-derived peptide chain.
  • protein carriers such as KLH, diphtheria toxoid, tetanus toxoid, and BSA
  • non-proteinaceous molecules such as carbohydrate polymers
  • the modification comprises addition of side chains or side groups to an polypeptide-derived peptide chain.
  • nucleic acid fragments encoding the chimeric binding proteins or polypeptides are important chemical products.
  • an important part of the invention pertains to a nucleic acid fragment which encodes chimeric binging protein described above, preferably a polypeptide wherein has been introduced a foreign T H -cell epitope in the scaffold protein structure by means of insertion and/or addition, preferably by means of substitution and/or deletion.
  • the nucleic acid fragments of the invention are either DNA or RNA fragments. Also included within the scope of the present invention are nucleic acid fragments complementary to a nucleic acid fragment encoding a chimeric binding protein of the invention.
  • the nucleic acid fragments of the invention will normally be inserted in suitable vectors to form cloning or expression vectors carrying the nucleic acid fragments of the invention; such novel vectors are also part of the invention. Details concerning the construction of these vectors of the invention will be discussed in context of transformed cells and microorganisms below.
  • the vectors can, depending on purpose and type of application, be in the form of plasmids, phages, cosmids, mini-chromosomes, or virus, but also naked DNA which is only expressed transiently in certain cells is an important vector.
  • Preferred cloning and expression vectors of the invention are capable of autonomous replication, thereby enabling high copy- numbers for the purposes of high-level expression or high-level replication for subsequent cloning.
  • the general outline of a vector of the invention comprises the following features in the 5'- 3' direction and in operable linkage: a promoter for driving expression of the nucleic acid fragment of the invention, optionally a nucleic acid sequence encoding a leader peptide enabling secretion of or integration into the membrane of the polypeptide fragment, the nucleic acid fragment of the invention, and a nucleic acid sequence encoding a terminator.
  • a promoter for driving expression of the nucleic acid fragment of the invention optionally a nucleic acid sequence encoding a leader peptide enabling secretion of or integration into the membrane of the polypeptide fragment, the nucleic acid fragment of the invention, and a nucleic acid sequence encoding a terminator.
  • a promoter for driving expression of the nucleic acid fragment of the invention optionally a nucleic acid sequence encoding a leader peptide enabling secretion of or integration into the membrane of the polypeptide fragment, the nucleic acid fragment of the invention
  • the vectors of the invention are used to transform host cells to produce the chimeric binding protein of the invention.
  • Such transformed cells which are also part of the invention, can be cultured cells or cell lines used for propagation of the nucleic acid fragments and vectors of the invention, or used for recombinant production of the chimeric binding protein of the invention.
  • the transformed cells can be suitable live vaccine strains wherein the nucleic acid fragment (one single or multiple copies) have been inserted so as to effect secretion or integration into the bacterial membrane or cell-wall of the chimeric binding protein.
  • Preferred transformed cells of the invention are microorganisms such as bacteria (such as the species Escherichia [e.g. E. coli], Bacillus [e.g. Bacillus subtilis], Salmonella, or Mycobacte- rium [preferably non-pathogenic, e.g. M. bovis BCG]), yeasts (such as Saccharomyces cere- visiae), and protozoans.
  • the transformed cells are derived from a multicellular organism such as a fungus, an insect cell, a plant cell, or a mammalian cell. Most preferred are cells derived from a human being, cf. the discussion of cell lines and vectors below.
  • the transformed cell is capable of replicating the nucleic acid fragment of the invention.
  • Cells expressing the nucleic fragment are preferred useful embodiments of the invention; they can be used for small-scale or large-scale preparation of the chimeric binding proteins or, in the case of non- pathogenic bacteria, as vaccine constituents in a live vaccine.
  • the expression product is either exported out into the culture medium or carried on the surface of the transformed cell.
  • this stable cell line which carries the vector of the invention and which expresses the nucleic acid fragment encoding the chimeric binding protein.
  • this stable cell line secretes or carries the chimeric binding protein of the invention, thereby facilitating purification thereof.
  • plasmid vectors containing replicon and control sequences which are derived from species compatible with the host cell are used in connection with the hosts.
  • the vector ordinarily carries a replication site, as well as marking sequences which are capable of providing phenotypic selection in transformed cells.
  • E. coli is typically transformed using pBR322, a plasmid derived from an E. coli species (see, e.g., Bolivar et al., 1977).
  • the pBR322 plasmid contains genes for ampicillin and tetracycline resistance and thus provides easy means for identifying transformed cells.
  • the pBR plasmid, or other microbial plasmid or phage must also contain, or be modified to contain, promoters which can be used by the pro- karyotic microorganism for expression.
  • promoters most commonly used in recombinant DNA construction include the B-lac- tamase (penicillinase) and lactose promoter systems (Chang et al., 1978; Itakura et al., 1977; Goeddel et al., 1979) and a tryptophan (trp) promoter system (Goeddel et al., 1979; EP-A-0 036 776). While these are the most commonly used, other microbial promoters have been discovered and utilized, and details concerning their nucleotide sequences have been published, enabling a skilled worker to ligate them functionally with plasmid vectors (Sieb- wenlist et al., 1980). Certain genes from prokaryotes may be expressed efficiently in E. coli from their own promoter sequences, precluding the need for addition of another promoter by artificial means.
  • eukaryqtic microbes such as yeast cultures may also be used, and here the promoter should be capable of driving expression.
  • Saccharomyces cerevisiase, or common baker's yeast is the most commonly used among eukaryotic microorganisms, al- though a number of other strains are commonly available such as Pichla pastoris.
  • the plasmid YRp7 for example, is commonly used (Stinchcomb et al., 1979; Kingsman et al., 1979; Tschemper et al., 1980).
  • This plasmid already contains the trpl gene which provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan for example ATCC No. 44076 or PEP4-1 (Jones, 1977).
  • the presence of the trpl lesion as a characteristic of the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
  • Suitable promoting sequences in yeast vectors include the promoters for 3-phosphoglycerate kinase (Hitzman et al., 1980) or other glycolytic enzymes (Hess et al., 1968; Holland et al., 1978), such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glu- cokinase.
  • the termination sequences associated with these genes are also ligated into the expression vector 3' of the sequence desired to be expressed to provide polyadenylation of the mRNA and termination.
  • promoters which have the additional advantage of transcription controlled by growth conditions are the promoter region for alcohol dehydrogenase 2, isocytochrome C, acid phos- phatase, degradative enzymes associated with nitrogen metabolism, and the aforementioned glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and ga- lactose utilization.
  • Any plasmid vector containing a yeast-compatible promoter, origin of repli- cation and termination sequences is suitable.
  • cultures of cells derived from multicellular organisms may also be used as hosts.
  • any such cell culture is workable, whether from vertebrate or invertebrate culture.
  • interest has been greatest in vertebrate cells, and propagation of vertebrate in culture (tissue culture) has become a routine procedure in recent years (Tis- sue Culture, 1973).
  • useful host cell lines are VERO and HeLa cells, Chinese hamster ovary (CHO) cell lines, and W138, BHK, COS-7 293 and MDCK cell lines.
  • Expression vectors for such cells ordinarily include (if necessary) an origin of replication, a promoter located in front of the gene to be expressed, along with any necessary ribosome binding sites, RNA splice sites, polyadenylation site, and transcriptional terminator sequences.
  • control functions on the expression vectors are often provided by viral material.
  • promoters are derived from polyoma, Adenovirus 2, and most frequently Simian Virus 40 (SV40).
  • the early and late promoters of SV40 virus are particularly useful because both are obtained easily from the virus as a frag- ment which also contains the SV40 viral origin of replication (Fiers et al., 1978).
  • Smaller or larger SV40 fragments may also be used, provided there is included the approximately 250 bp sequence extending from the Hind ⁇ l ⁇ site toward the Bgll site located in the viral origin of replication.
  • promoter or control sequences normally associated with the desired gene sequence provided such control sequen- ces are compatible with the host cell systems.
  • An origin of replication may be provided either by construction of the vector to include an exogenous origin, such as may be derived from SV40 or other viral (e.g., Polyoma, Adeno, VSV, BPV) or may be provided by the host cell chromosomal replication mechanism. If the vector is integrated into the host cell chromosome, the latter is often sufficient.
  • an exogenous origin such as may be derived from SV40 or other viral (e.g., Polyoma, Adeno, VSV, BPV) or may be provided by the host cell chromosomal replication mechanism. If the vector is integrated into the host cell chromosome, the latter is often sufficient.
  • compositions of the invention are provided.
  • immunogenic compositions that include chimeric binding proteins of the invention, nucleic acid fragments of the invention or non-pathogenic virus or bacteria that are capable of functioning as immunogenic agents because they express the chimeric binding proteins of the invention.
  • the invention also relates to a composition for inducing production of antibodies against an antigen in the autologous host, the composition comprising
  • a chimeric binding proteinor the invention or a nucleic acid fragment of the invention or a vector of the invention including such vectors as non-pathogenic virus or microorganisms, and - a pharmaceutically and immunologically acceptable carrier and/or vehicle and/or adjuvant
  • the invention relates to a method for down-regulating a self-antigen or a cell that displays epitopes of said self-antigen in an animal, the method comprising presenting the animal's immune system with an immunogenicaily effective amount of a chimeric binding protein of the invention.
  • One preferred way of achieving this goal entails administration to the animal in question of an immunogenicaily effective amount of the chimeric binding protein.
  • the chimeric binding protein is formulated together with a pharmaceutically and immunologically acceptable carrier and/or vehicle and, optionally an adjuvant.
  • the formulation of the polypeptide follows the principles generally acknowledged in the art.
  • vaccines which contain peptide sequences as active ingredients are generally well understood in the art, as exemplified by US Patents 4,608,251; 4,601,903; 4,599,231; 4,599,230; 4,596,792; and 4,578,770, all incorporated herein by reference.
  • such vaccines are prepared as injectables either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection may also be prepared.
  • the preparation may also be emulsified.
  • the active immunogenic ingredient is often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient.
  • Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof.
  • the vaccine may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, or adjuvants which enhance the effectiveness of the vaccines; cf. the detailed discussion of adjuvants below.
  • the vaccines are conventionally administered parenterally, by injection, for example, either subcutaneously, intracutaneously, or intramuscularly.
  • Additional formulations which are suitable for other modes of administration include suppositories and, in some cases, oral, buccal, sublinqual, intraperitoneal, intravaginal, anal and intracranial formulations.
  • suppositories traditional binders and carriers may include, for example, polyalkalene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in the range of 0.5% to 10%, preferably 1-2%.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and contain 10-95% of active ingredient, preferably 25-70%.
  • cholera toxin is an interesting formulation partner (and also a possible conjugation partner).
  • the chimeric binding protein may be formulated into the vaccine as neutral or salt forms.
  • Pharmaceutically acceptable salts include acid addition salts (formed with the free amino groups of the peptide) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • the vaccines are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective and immunogenic.
  • the quantity to be administered depends on the subject to be treated, including, e.g., the capacity of the individual's immune system to mount an immune response, and the degree of protection desired.
  • Suit- able dosage ranges are of the order of several hundred micrograms active ingredient per vaccination with a preferred range from about 0.1 ⁇ g to 2000 ⁇ g (even though higher amounts in the 1-10 mg range are contemplated), such as in the range from about 0.5 ⁇ g to 1000 ⁇ g, preferably in the range from 1 ⁇ g to 500 ⁇ g and especially in the range from about 10 ⁇ g to 100 ⁇ g.
  • Suitable regimens for initial administration and booster shots are also variable but are typified by an initial administration followed by subsequent inoculations or other administrations.
  • the manner of application may be varied widely. Any of the conventional methods for administration of a vaccine are applicable. These include oral application on a solid physiologically acceptable base or in a physiologically acceptable dispersion, parenterally, by injection or the like.
  • the dosage of the vaccine will depend on the route of administration and will vary according to the age of the person to be vaccinated and the formulation of the antigen.
  • chimeric binding proteins of the vaccine are sufficiently immunogenic in a vaccine, but for some of the others the immune response will be enhanced if the vaccine further comprises an adjuvant substance. It is especially preferred to use an adjuvant which can be demonstrated to facilitate breaking of the autotolerance to autoantigens.
  • One group of preferred adjuvants facilitate uptake of the vaccine molecules by APCs, such as dendritic cells, and activate these.
  • APCs such as dendritic cells
  • Non-limiting examples are selected from the group consis- ting of an immune targeting adjuvant; an immune modulating adjuvant such as a toxin, a cytokine, and a mycobacterial derivative; an oil formulation; a polymer; a micelle forming adjuvant; a saponin; an immunostimulating complex matrix (ISCOM matrix); a particle; DDA; aluminium adjuvants; DNA adjuvants; ⁇ -inulin; and an encapsulating adjuvant.
  • an immune targeting adjuvant an immune modulating adjuvant such as a toxin, a cytokine, and a mycobacterial derivative
  • an oil formulation a polymer
  • a micelle forming adjuvant such as a toxin, a cytokine, and a mycobacterial derivative
  • adjuvants include use of agents such as aluminium hydroxide or phosphate (alum), commonly used as 0.05 to 0.1 percent solution in buffered saline, admixture with synthetic polymers of sugars (e.g. Carbopol®) used as 0.25 percent solution, aggregation of the protein in the vaccine by heat treatment with temperatures ranging between 70° to 101°C for 30 second to 2 minute periods respectively and also aggregation by means of cross-linking agents are possible. Aggregation by reactivation with pepsin treated antibodies (Fab fragments) to albumin, mixture with bacterial cells such as C.
  • agents such as aluminium hydroxide or phosphate (alum), commonly used as 0.05 to 0.1 percent solution in buffered saline, admixture with synthetic polymers of sugars (e.g. Carbopol®) used as 0.25 percent solution, aggregation of the protein in the vaccine by heat treatment with temperatures ranging between 70° to 101°C for 30 second to 2 minute periods respectively and also
  • parvum or endotoxins or lipopoly- saccharide components of gram-negative bacteria emulsion in physiologically acceptable oil vehicles such as mannide mono-oleate (Aracel A) or emulsion with 20 percent solution of a perfluorocarbon (Fluosol-DA) used as a block substitute may also be employed. Admixture with oils such as squalene and IFA is also preferred.
  • DDA dimethyldioctadecylammonium bromide
  • DNA DNA, MF59, and ⁇ -inulin
  • Freund's complete and in- complete adjuvants as well as quillaja saponins such as QuilA and QS21 are interesting.
  • MPL monophosphoryl lipid A
  • Liposome formulations are also known to confer adjuvant effects, and therefore liposome adjuvants are preferred according to the invention.
  • immunostimulating complex matrix type (ISCOM® matrix) adjuvants are preferred choices according to the invention, especially since it has been shown that this type of adjuvants are capable of up-regulating MHC Class II expression by APCs.
  • An ISCOM® matrix consists of (optionally fractionated) saponins (triterpenoids) from Quillaja saponaria, cholesterol, and phospholipid. When admixed with the immunogenic protein, the resulting particulate formulation is what is known as an ISCOM particle where the saponin constitutes 60-70% w/w, the cholesterol and phospholipid 10-15% w/w, and the protein 10-15% w/w. Details relating to composition and use of immunostimulating complexes can e.g.
  • a relevant antigen such as a modified CEA poly- peptide of the present invention
  • the presentation of a relevant antigen can be enhanced by conjugating the antigen to antibodies (or antigen binding antibody fragments) against the Fc ⁇ receptors on monocytes/macrophages.
  • conjugates including anti-Fc ⁇ RI or Fc ⁇ RI binding structures are believed to enhance immunogenicity for the purposes of vaccination.
  • cytokines targeting and immune modulating substances (i.a. cytokines) mentioned above as candidates for the first and second moieties in the chimeric binding protein.
  • synthetic inducers of cytokines like poly I:C are possibilities.
  • Suitable mycobacterial derivatives are selected from the group consisting of muramyl dipep- tide, complete Freund's adjuvant, RIBI, and a diester of trehalose such as TDM and TDE.
  • Suitable immune targeting adjuvants are selected from the group consisting of CD40 ligand and CD40 antibodies or specifically binding fragments thereof (cf. the discussion above), mannose, a Fab fragment, and CTLA-4.
  • Suitable polymer adjuvants are selected from the group consisting of a carbohydrate such as dextran, PEG, starch, mannan, and mannose; a plastic polymer; and latex such as latex beads.
  • VLN virtual lymph node
  • the VLN (a thin tubular device) mimics the structure and function of a lymph node. Insertion of a VLN under the skin creates a site of sterile inflammation with an upsurge of cytokines and chemokines. T- and B-cells as well as APCs rapidly respond to the danger signals, home to the inflamed site and accumulate inside the porous matrix of the VLN.
  • the vaccine should be administered at least once a year, such as at least 1, 2, 3, 4, 5, 6, and 12 times a year. More specifically, 1-12 times per year is expected, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 times a year to an individual in need thereof. It has previously been shown that the memory immunity induced by the use of the preferred auto- vaccines according to the invention is not permanent, and therefore the immune system needs to be periodically challenged with the chimeric binding protein.
  • the vaccine according to the invention may comprise several different chimeric binding proteins polypeptides in order to increase the immune response, cf. also the discussion above concerning the choice of foreign T H epitope introductions.
  • the vaccine may comprise two or more polypeptides, where all of the polypeptides are as defined above.
  • the vaccine may consequently comprise 3-20 different modified polypeptides, such as 3-10 different polypeptides. However, normally the number of peptides will be sought kept to a minimum such as 1 or 2 peptides.
  • the second alternative for effecting presentation to the immune system is the use of live vac- cine technology.
  • presentation to the immune system is effected by administering, to the animal, a non-pathogenic microorganism which has been transformed with a nucleic acid fragment encoding the chimeric binding protein.
  • the microorganism is transformed with a vector incorporating such a nucleic acid fragment.
  • the non-pathogenic microorganism can be any suitable attenuated bacterial strain (attenuated by means of pas- saging or by means of removal of pathogenic expression products by recombinant DNA technology), e.g. Mycobacterium bovis BCG., non-pathogenic Streptococcus spp., E.
  • BCG has been used extensively as a live bacterial vaccine:
  • the BCG vaccine has been successfully used to prevent tuberculosis around the world. Vaccination can be given after birth and results in few severe complications, even in individuals who are infected with human immunodeficiency virus type 1.
  • BCG possesses strong immune adjuvant activity, and has been used extensively in the treatment of superficial bladder cancers.
  • the T H epitope and/or the first and/or second and/or third moieties can, if present, be in the form of fusion partners to the amino acid sequence derived from the scaffold protein structure.
  • the nucleic acid fragment of the invention discussed below can be incorporated in a non-virulent viral vaccine vector.
  • Feasible viral vectors are selected from a pox virus such as vaccinia, MVA (modified Vaccinia virus), canary pox, avi-pox, and chicken pox etc.
  • a herpes simplex virus variant can be used.
  • the non-pathogenic microorganism or virus is administered only once to the animal, but in certain cases it may be necessary to administer the microorganism more than once in a lifetime.
  • the microorganism can be transformed with nucleic acid(s) containing regions encoding the 1 st , 2 nd and/or 3 rd moieties, e.g. in the form of the immunomodulating substances descri- bed above such as the cytokines discussed as useful adjuvants.
  • a preferred version of this embodiment encompasses having the coding region for the chimeric binding protein and the coding region for the immunomodulator in different open reading frames or at least under the control of different promoters. Thereby it is avoided that the chimeric binding protein are produced as fusion partners to the immunomodulator, and effect that may sometimes provide for advantages.
  • two distinct nucleotide fragments can be used as transforming agents.
  • nucleic acid vaccination also known as “nucleic acid immunisation”, “genetic immunisation”, “gene immunisation” and “DNA vaccination
  • nucleic acid immunisation also known as “nucleic acid immunisation”, “genetic immunisation”, “gene immunisation” and “DNA vaccination
  • nucleic acid vaccination does not require resource consuming large-scale production of the immunogenic agent (e.g. in the form of industrial scale fermentation of microorganisms producing the chimeric binding proteins necessary in polypeptide vaccination).
  • the immunogenic agent e.g. in the form of industrial scale fermentation of microorganisms producing the chimeric binding proteins necessary in polypeptide vaccination.
  • nucleic acid vaccination relies on the biochemical apparatus of the vaccinated individual in order to produce the expression product of the nucleic acid introduced, the optimum posttranslational processing of the expression product is expected to occur; this is especially important in the case of autovaccination, since, as mentioned above, a significant fraction of the original B-cell epitopes should be preserved in the chimeric binding proteins derived from extracellularly exposed polypeptide sequences, and since B-cell epitopes in principle can be constituted by parts of any (bio)molecule (e.g. carbohydrate, lipid, protein etc.). Therefore, native glycosylation and lipidation patterns of the immunogen may very well be of importance for the overall immunogenicity and this is best ensured by having the host producing the immunogen.
  • bio bio
  • an important embodiment of the method of the invention involves that presentation is effected by administering a nucleic acid fragment encoding and expressing a chimeric binding protein of the invention.
  • the nucleic acid vaccination can be combined with in vivo introduction, into the APC, of at least one nucleic acid fragment encoding and ex- pressing the second analogue.
  • in vivo introduction into the APC, of at least one nucleic acid fragment encoding and ex- pressing the second analogue.
  • the introduced nucleic acid is preferably DNA which can be in the form of naked DNA, DNA formulated with charged or uncharged lipids, DNA formulated in liposomes, emulsified DNA, DNA included in a viral vector, DNA formulated with a transfection-facilitating protein or polypeptide, DNA formulated with a targeting protein or polypeptide, DNA formulated with Calcium precipitating agents, DNA coupled to an inert carrier molecule, and DNA formulated with an adjuvant.
  • DNA can be in the form of naked DNA, DNA formulated with charged or uncharged lipids, DNA formulated in liposomes, emulsified DNA, DNA included in a viral vector, DNA formulated with a transfection-facilitating protein or polypeptide, DNA formulated with a targeting protein or polypeptide, DNA formulated with Calcium precipitating agents, DNA coupled to an inert carrier molecule, and DNA formulated with an adjuvant.
  • nucleic acid vaccination technology all disclosures herein which relate to use of adjuvants in the context of polypeptide based vaccines apply mutatis mutandis to their use in nucleic acid vaccination technology.
  • considerations relating to formulation and mode and route of administration and, hence, also these considerations discussed above in connection with a traditional vaccine apply mutatis mutandis to their use in nucleic acid vaccination technology.
  • One especially preferred type of formulation of nucleic acid vaccines are microparticles containing the DNA. Suitable microparticles are e.g. described in WO 98/31398.
  • the nucleic acid(s) used as an immunization agent can- contain regions encoding the 1 st , 2 nd and/or 3 rd moieties, e.g. in the form of the immunomodulating substances descri- bed above such as the cytokines discussed as useful adjuvants.
  • a preferred version of this embodiment encompasses having the coding region for the chimeric binding protein and the coding region for the immunomodulator in different open reading frames or at least under the control of different promoters. Thereby it is avoided that the chimeric binding protein is produced as a fusion partner to the immunomodulator, an effect that is believed to provide ad- vantages in terms of preservation of structure of the scaffold protein structure.
  • two distinct nucleotide fragments can be used, but this is less preferred because of the advantage of ensured co-expression when having both coding regions included in the same molecule.
  • nucleic acid of the vaccine is introduced in the form of a vector wherein expression is under control of a viral promoter.
  • vectors according to the invention cf. the discussion below.
  • detailed disclosures relating to the formulation and use of nucleic acid vaccines are available, cf. Donnelly JJ et al, 1997, Annu. Rev. Immunol. 15: 617-648 and Donnelly JJ et al., 1997, Life Sciences 60: 163- 172. Both of these references are incorporated by reference herein.
  • the expression cassette in the nucleic acid vaccine can be constructed so as to ensure that no export of the expression product takes place (e.g. by omitting signal sequences that would result in membrane integration or secretion). In this way, only minute amounts of expression product will be exported, whereas the remainder will be processed and presented as peptide fragments in the context of MHC molecules.
  • Prime-boost strategies have proven effective in facilitating an improved immune response.
  • use of any combination of nucleic acid vaccination, live vaccination and polypeptide vaccination may be utilised.
  • the most important features of the chimeric binding proteins of the invention are their ability to elicit an immune response that is only effective in targeting the B-cell epitope of interest even though they might be able to induce antibodies reactive with both the scaffold protein structure and the tolerance breaking amino acid sequence.
  • B-cell epitopes may have very varying structures, the scaffold protein structure that is suitable for one B-cell epitope and its presentation is almost with certainty not suitable for a different B-cell epitope, so the present invention is best put into practice when at least an effective combination of B-cell epitope and scaffold protein structure has been identified.
  • the method for identifying the chimeric binding protein of the invention will in its broadest scope include the following steps:
  • the chimeric binding protein that contains at least a) the binding site of a member isolated in step 3, b) a scaffold protein structure autologous in the animal that stabilises the native 3D structure of said binding site, and c) a non-human MHC Class II binding amino acid sequence;
  • a chimeric binding protein containing 1) the second receptor or a mimotope thereof in correct, native 3D conformation, 2) a scaffold protein structure autologous in the animal, said scaffold protein structure stabilising said 3D conformation and being derived from another molecule in the animal than the second receptor, and 3) the tolerance breaking amino acid sequence.
  • the B-cell epitope part can simply be the (already identified) epitope of interest in the relevant self-antigen that is targeted by the vaccines of the present invention - this is normally easiest to achieve when the epitope is a linear peptidic epitope.
  • the B-cell epitope can be inserted in a suitable abundant autologous protein structure, or it can be introduced randomly in a putative scaffold structure and subsequently the thus obtained library can be screened for those mem- bers that selectively bind to the first receptor.
  • the above mentioned library consists of a known putative scaffold protein structure (where the tolerance breaking amino acid sequence may already be present) where the B-cell epitope or a mimotope thereof is shuffled around in various random positions - from this library, only those members that provide a suitable binding to the first receptor (for instance a suitable mAB) will be selected.
  • the position for introduction of the B-cell epitope can be fixed, whereas the B- cell epitope itself is identified from a library of random sequences inserted in this position.
  • This can preferably be achieved by screening a library of antibodies of the same class and subclass for members that bind to the first receptor.
  • the above- mentioned library consists of a known putative scaffold protein structure (where the tolerance breaking amino acid sequence may already be present) where the B-cell epitope is a member of a random sequence library introduced in the scaffold protein structure at a fixed position. From this library, only those members that provide a suitable binding to the first receptor (for instance a suitable mAB) will be selected.
  • a third possibility, that does not necessarily require the above-mentioned screening, is to isolate naturally occurring anti-idiotypic antibodies that mimic an epitope of an antigen of interest. For instance, if an animal harbours antibodies against an autologous antigen, then the same animal also harbours anti-idiotypic antibodies reactive with the idiotype of these antibodies. Such anti-idiotypic antibodies can be isolated, sequenced and subsequently a modified version wherein a tolerance breaking amino acid sequence is introduced can be prepared. Again, the resulting molecule will include a B-cell epitope (the idiotype of the anti- idiotypic antibody), a scaffold protein structure (the non-idiotypic region of the anti-idiotypic antibody) and a tolerance breaking amino acid sequence.
  • the first mole- cule including the first receptor, is used as an immunogen to immunize a transgenic animal that expresses antibodies autologous in the host harbouring the antigen with the second receptor - if necessary the first molecule may be coupled to an immunogenic carrier molecule such as tetanus toxoid, KLH, and diphtheria toxoid (this is the case if the first molecule is autologous in the transgenic animal).
  • the transgenic animal may for instance be a non-hu- man animal that expresses human immunoglobulin (e.g.
  • the first molecule will in this situation induce production of human antibodies that react with the first molecule.
  • standard technology for preparing hybridomas followsing the classical teachings of K ⁇ hler and Millstein, a selection of hybridomas are prepared that produce monoclonal antibodies reacting with the first molecule, cf. below.
  • human monoclonal antibodies are thereafter screened for their specific binding to the, first receptor so as to exclude those mABs that bind non-specifically to other parts of the first molecule.
  • the hybridomas producing the specifically binding mABs are then isolated and by use of standard cloning and molecular biology technology, the DNA encoding these antibodies (or fragments thereof) is used to transform suitable production cell lines (of course, DNA having the same sequences but provided by means of e.g. synthesis or PCR may also be used to transform).
  • this can either be introduced as part of the cloning steps when the DNA is transformed into the production cell line, but an alternative is to provide a transgenic animal where the IgG molecules already include an amino acid se- quence that will be tolerance breaking in the animal to ultimately be vaccinated with the mABs.
  • the first molecule is preferably an antibody and even more preferably a monoclonal antibody.
  • this allows for preparation of e.g. an immunogenized anti-idiotypic antibody that will only raise a relevant immune response against the idiotype.
  • the first receptor is the idiotype of the antibody.
  • steps 2 and 3 of the method provides for isolation of those members of the library that interacts specifically with the first receptor.
  • the receptor is part of a larger molecule (e.g. when the receptor is the antigen binding part of an antibody) then the screening will also give rise to a number of isolated/identified members that react with the remainder of the larger molecule.
  • screening in step 3 includes an exclusion step that allows identification of members of the library that bind the first molecule outside the first receptor so as to exclude such members from subsequent steps.
  • Such an exclu- sion step involves a) a test of the library members' ability to bind to the region outside the first receptor in the first molecule, so as to allow exclusion of library members that exhibit such binding, or b) a test of the library members' ability to compete with the second receptor for binding to the first receptor that allows exclusion of library members that do not exhibit such ability.
  • option (a) relies on capture of members of the library that bind to the scaffold protein structure and/or the tolerance breaking amino acid sequence, e.g. in a chroma- tographic step where the first molecule lacking the first receptor (such as use of an irrelevant antibody of the same subclass as the antibody used as the first molecule) is fixed to a chro- matographic medium in a column and where the library is passed through said column: only members that are not retained by the column will be those that bind the first receptor.
  • a chroma- tographic step where the first molecule lacking the first receptor (such as use of an irrelevant antibody of the same subclass as the antibody used as the first molecule) is fixed to a chro- matographic medium in a column and where the library is passed through said column: only members that are not retained by the column will be those that bind the first receptor.
  • Option (b) relies on competition assays, where only those members of the library are selected that are capable of competing with the relevant second receptor for binding to the first receptor.
  • This can also be done chromatographically, where the first molecule (including the first receptor) is fixed to a chromatographic medium in a column, a molecule including the second receptor is applied to the column in excess so as to block all first receptors, and finally the library members are passed through the column. Only those that flow through will be those that bind the first receptor.
  • One suitable method for presenting the library of second molecules is to utilise phage display, cf. the example below, but also the technologies of ribosome display, mRNA-display, and yeast surface display are possibilities.
  • the present invention is illustrated by means of phage display technology as an example - it will be understood, however that any art-recognized method of screening a library of binding partners may be utilised according to the present invention.
  • the phage display technology is used to provide a direct physical link between phenotype and genotype by utilising the ability to display peptides and proteins on the surface of the filamentous phage.
  • the technology takes advantage of the infectious ability of the filamentous bacteriophage (like fl and M13), which infects bacteria by interaction with the F-pilus on the surface of the bac- teria. Unlike most bacterial viruses the filamentous phage are not released by cell lysis but is secreted from the cell while the phage is packaged and assembled in the bacterial membrane. The growth of the bacterial cell is only slightly reduced by the infection and it is therefore possible to achieve very high phage titers in culture (Markowski and Russel 1997).
  • the filamentous phage protein sheath is made up of five different coating proteins (pill, pVI, pVII, pVIII and pIX) as illustrated in Fig. 1.
  • the coding sequence of the peptide/protein has to be joined with the coding sequence of a coating protein of the phage.
  • George Smith was the first who cloned small protein-encoding DNA-fragments into gill (the coding sequence of the minor coat protein, pill) of the phage genome and showed that the resulting phage carried polypeptides that could be recognised by a specific antibody (Smith GP, 1985).
  • Phagemids are plasmids, which bear the integenic region of a filamentous phage. This region contains a DNA replication origin and a DNA packaging signal.
  • phagemids are propagated in cells su- perinfected with a helper phage, the phagemid DNA is packaged in phage particles in a fa- shion identical to the phage genome itself.
  • the phagemid carries the coding sequence of a coating protein of the phage (usually pill or part of pill) to which the antibody-fragment is fused. Upon superinfection with helper phage, antibody fragments are displayed on the surface of the phage, fused to the coating protein. (Hoogenboom et al.1991, ⁇ rum et al. 1993).
  • Antibody libraries for phage display are available (e.g human phage antibody libraries from Dyax). Methods for creating libraries have also been described (Engberg et al. 1996 and Krebber et al. 1997). Construction of an antibody library usually sets out with isolation of mRNA from a source containing antibody-producing cells. (Winter et al. 1994). The mRNA is subsequently used as template for cDNA synthesis by RT-PCR. The PCR amplification of the antibody repertoire is done by use of specific primers designed for amplification of the heavy and light chain genes of the antibody fragment. Subsequently the amplified heavy and light chain genes are cloned into a vector and transformed into an E. coli strain that expresses F-pili on the surface. The transformed cells are propagated in liquid culture or on plates, and the propagated cells thereby constitute the antibody library. (Engberg et al. 1996, Krebber et al. 1997).
  • the most preferred library to display would be a human antibody library, but also antibody libraries from other species would be useful.
  • the hypervariable regions of the identified antibodies will be identified and substituted into the hypervariable region of a human antibody scaffold.
  • the antibody library In order to prapare the phage display antibody library, the antibody library needs to be su- perinfected by a helper phage. Following the superinfection the infected bacteria produce re- combinant phage particles where each phage represents a unique antibody encoded by genes in the same phage particle.
  • the display of antibody fragments allows for selection and purification of antigen-specific phage by repeated rounds of selection (biopanning or panning).
  • one round of selection consists of: superinfection of the E. coli antibody library, production of recombinant phage particles, incubation of phage with the antigen (for instance a monoclonal antibody of choice), removal of non-binding phage by a washing procedure, Elution of bound phage particles and transduction fresh E. coli cells with the eluted phage. Elution of the bound phage is generally achieved with low pH buffers, but it is also possible to elute with high pH buffers, soluble antigen or by proteolytic cleavage. In the latter case, a specific protease recognition site has been inserted in the vector between the se- quence of the coating protein and the antibody-fragment. (Johansen et al. 1995). After several panning-rounds individual clones are isolated and analysed for binding affinity and specificity for the selected antigen.
  • the antigen (which can be any of the above-discussed monoclonal antibodies that have proven useful in a clinical setting, i.a. Herceptin or Retuximab) can be immobilized onto different solid phases such as ELISA wells, latex- or magnetic beads, immunotubes or columns.
  • phage could be directly selected on cells, or antigen-displaying cells may be sorted by virtue of a marker unique for these cells e.g. using fluorescence activated cell sorting (FACS) (Hoogenboom, 1997). Combinations and alterations between these me- thods may be done in order to avoid background binders (phage binding to the immobilization medium) e.g.
  • Binders can usually be isolated after three to four rounds of panning dependent of the panning procedure.
  • the present invention aims at isolating only the phage that bind the interesting region interacting with a known receptor structure in the animal to be vaccinated, it is expedient to also remove phage that bind to other regions in the antigen. For instance, if the antigen is a monoclonal antibody, and it is only of relevance to isolate phage that bind to the idiotype of the monoclonal, then a second round of selection can be carried out, where only phage are selected that compete with the monoclonal antibody's target ligand.
  • phage that have already passed the step of binding to the antigen can be applied to a column where the antigen has been pre-incubated with its known ligand. Phage that run through the column must be assumed to bind the site occupied by the known ligand.
  • the steps for providing the chimeric binding protein will involve simple isolation and purification of the positive antibodies (in cases where the library screened includes antibodies or fragments that also include the tolerance breaking amino acid sequence), genetic engineering of the DNA enco- ding the positive antibodies so as to introduce the tolerance breaking amino acid sequence and/or necessary parts of the scaffold protein structure (in cases where only fragments of antibody has been screened and it is necessary to supply all or some part of the remaining scaffold protein structure), and genetic engineering of the DNA encoding the positive antibodies so as to move the antigen binding region to a suitable scaffold protein structure with a tolerance breaking amino acid sequence.
  • the monoclonal antibody Xolair is known to bind IgE without being able to cross-link IgE when bound to the Fc ⁇ RI receptor on mast cells and basophils.
  • the following simple steps could be carried out:
  • - Generating/obtaining a fully human IgG phage display library (potentially containing suitable epitopes such as the P2 and P30 epitopes from tetanus toxoid or the above-reference PADRE epitope), - Probing in the phage display with Xolair for antibodies that: 1) compete with human IgE's binding to Xolair (positive selection) and/or 2) does not recognise non-specific IgG (negative selection)
  • Phage-enzymes expression and affinity chromatography of functional alkaline phosphatase on the surface of the bacteriophage. Protein eng. 4(8) : 955-61

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Virology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L'invention concerne de nouveaux agents immunogènes qui sont capables d'induire une immunité des cellules B spécifique, dirigée contre un seul épitope présente dans un auto-antigène. L'agent immunogène est une protéine de liaison chimère qui se lie spécifiquement à un premier récepteur, ledit premier récepteur étant un récepteur qui se lie à un second récepteur présent dans un antigène d'un animal. L'invention est caractérisée en ce que ladite protéine de liaison chimère comprend : (a) un épitope des cellules B sous la forme d'un site de combinaison avec l'antigène liant spécifiquement le premier récepteur et qui entre en compétition avec le second récepteur en vue de la liaison avec le premier récepteur, (b) une structure protéine support stabilisant la conformation 3D du site de combinaison, ladite structure protéine support étant autologue dans ledit mammifère, et (c) au moins une séquence aminoacide de rupture de tolérance, qui est hétérologue dans ledit animal et qui se lie à une molécule MHC classe II dans ledit animal. Selon des formes d'exécution préférées, la protéine de liaison chimère est sous la forme d'un anticorps anti-idiotypique ayant une séquence aminoacide de rupture de tolérance introduite. L'invention concerne en outre des procédés de préparation d'immunogènes et des procédés d'utilisation d'immunogènes en thérapie. L'invention concerne par ailleurs des protéines, des fragments d'acide nucléique, des cellules hôtes et des virus modifiés de façon recombinante qui sont utilisés pour la mise en oeuvre des procédés de l'invention.
PCT/DK2003/000859 2002-12-11 2003-12-11 Epitopes uniques de ciblage WO2004052930A2 (fr)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EA200500943A EA200500943A1 (ru) 2002-12-11 2003-12-11 Направленное действие на еденичные эпитопы
CA002505870A CA2505870A1 (fr) 2002-12-11 2003-12-11 Epitopes uniques de ciblage
MXPA05006113A MXPA05006113A (es) 2002-12-11 2003-12-11 Epitopos sencillos objetivo.
AU2003285280A AU2003285280A1 (en) 2002-12-11 2003-12-11 Targeting single epitopes
EP03778264A EP1572740A2 (fr) 2002-12-11 2003-12-11 Epitopes uniques de ciblage
NO20053363A NO20053363L (no) 2002-12-11 2005-07-11 Malrettede enkle epitoper.

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US43253202P 2002-12-11 2002-12-11
US60/432,532 2002-12-11
DKPA200201893 2002-12-11
DKPA200201893 2002-12-11
US44670703P 2003-02-12 2003-02-12
DKPA200300198 2003-02-12
DKPA200300198 2003-02-12
US60/446,707 2003-02-12

Publications (2)

Publication Number Publication Date
WO2004052930A2 true WO2004052930A2 (fr) 2004-06-24
WO2004052930A3 WO2004052930A3 (fr) 2004-07-29

Family

ID=32512419

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2003/000859 WO2004052930A2 (fr) 2002-12-11 2003-12-11 Epitopes uniques de ciblage

Country Status (6)

Country Link
EP (1) EP1572740A2 (fr)
AU (1) AU2003285280A1 (fr)
CA (1) CA2505870A1 (fr)
MX (1) MXPA05006113A (fr)
NO (1) NO20053363L (fr)
WO (1) WO2004052930A2 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7371726B2 (en) 2003-12-31 2008-05-13 Schering-Plough Animal Health Corporation Neutralizing GDF8 epitope-based growth enhancing vaccine
US7432079B2 (en) 2004-12-30 2008-10-07 Schering-Plough Animal Health Corporation Plant virus coat fusion proteins with GDF8 epitopes and vaccines thereof
EP2344184A2 (fr) * 2008-10-07 2011-07-20 Biovest International, Inc. Procédés d'induction d'une réponse immunitaire prolongée contre un idiotype de cellules b, au moyen de vaccins autologues anti-idiotypiques
CN102184348A (zh) * 2011-01-12 2011-09-14 湖北大学 基于自然生殖过程的昆虫遗传群体的模拟方法
US9725768B2 (en) 2012-08-31 2017-08-08 Biovest International, Inc. Methods for producing high-fidelity autologous idiotype vaccines
EP2771349B1 (fr) * 2011-09-16 2020-02-26 Iogenetics, LLC. Procédés bio-informatiques de détermination de liaisons peptidiques
WO2021163239A1 (fr) * 2020-02-11 2021-08-19 Ubi Ip Holdings Immunogènes peptidiques ciblant un polypeptide amyloïde d'îlots (iapp) pour la prévention et le traitement de troubles liés à un iapp agrégé
CN113383006A (zh) * 2018-02-19 2021-09-10 高丽大学校产学协力团 包含热休克蛋白表位的疫苗及其用途

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995005849A1 (fr) * 1993-08-26 1995-03-02 Mouritsen & Elsner A/S Procede d'induction de reactions immunitaires contre les proteines endogenes a l'aide d'epitopes de lymphocytes t exogenes
WO1995007707A1 (fr) * 1993-09-14 1995-03-23 Cytel Corporation Alteration de la reponse immunitaire a l'aide de peptides se liant a des alleles pan dr
WO1995017212A1 (fr) * 1993-12-23 1995-06-29 Boehringer Mannheim Gmbh Conjugues constitues d'antigenes de lymphocytes t peptidiques ainsi que de partenaires de liaison cellulaire et leur utilisation en therapie
WO1997026784A1 (fr) * 1996-01-24 1997-07-31 Cytel Corporation Induction d'une reaction immune contre des determinants souhaites
WO1999058658A2 (fr) * 1998-05-13 1999-11-18 Epimmune, Inc. Vecteurs d'expression destines a stimuler une reponse immunitaire et procedes de leur utilisation
WO2001041741A1 (fr) * 1999-12-13 2001-06-14 Epimmune Inc. Compositions de vaccins et peptides d'antigene associes a une tumeur de hla-a2 de classe i
WO2003042244A2 (fr) * 2001-11-16 2003-05-22 Pharmexa A/S Mimetiques immunogenes de proteines multimeres
US20030185845A1 (en) * 2001-11-16 2003-10-02 Steen Klysner Novel immunogenic mimetics of multimer proteins
US20030228324A1 (en) * 1999-05-06 2003-12-11 Malcolm Andrew J. Peptide compositions and methods of producing and using same

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995005849A1 (fr) * 1993-08-26 1995-03-02 Mouritsen & Elsner A/S Procede d'induction de reactions immunitaires contre les proteines endogenes a l'aide d'epitopes de lymphocytes t exogenes
WO1995007707A1 (fr) * 1993-09-14 1995-03-23 Cytel Corporation Alteration de la reponse immunitaire a l'aide de peptides se liant a des alleles pan dr
WO1995017212A1 (fr) * 1993-12-23 1995-06-29 Boehringer Mannheim Gmbh Conjugues constitues d'antigenes de lymphocytes t peptidiques ainsi que de partenaires de liaison cellulaire et leur utilisation en therapie
WO1997026784A1 (fr) * 1996-01-24 1997-07-31 Cytel Corporation Induction d'une reaction immune contre des determinants souhaites
WO1999058658A2 (fr) * 1998-05-13 1999-11-18 Epimmune, Inc. Vecteurs d'expression destines a stimuler une reponse immunitaire et procedes de leur utilisation
US20030228324A1 (en) * 1999-05-06 2003-12-11 Malcolm Andrew J. Peptide compositions and methods of producing and using same
WO2001041741A1 (fr) * 1999-12-13 2001-06-14 Epimmune Inc. Compositions de vaccins et peptides d'antigene associes a une tumeur de hla-a2 de classe i
WO2003042244A2 (fr) * 2001-11-16 2003-05-22 Pharmexa A/S Mimetiques immunogenes de proteines multimeres
US20030185845A1 (en) * 2001-11-16 2003-10-02 Steen Klysner Novel immunogenic mimetics of multimer proteins

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
LA ROSA CORINNA ET AL: "Preclinical development of an adjuvant-free peptide vaccine with activity against CMV pp65 in HLA transgenic mice." BLOOD. UNITED STATES 15 NOV 2002, vol. 100, no. 10, 15 November 2002 (2002-11-15), pages 3681-3689, XP002277230 ISSN: 0006-4971 *
VOGEL M ET AL: "Mimicry of human IgE epitopes by anti-idiotypic antibodies." JOURNAL OF MOLECULAR BIOLOGY. ENGLAND 19 MAY 2000, vol. 298, no. 5, 19 May 2000 (2000-05-19), pages 729-735, XP002277231 ISSN: 0022-2836 *
WEATHINGTON NATHANIEL M ET AL: "Rational design of peptide vaccines for autoimmune disease: harnessing molecular recognition to fix a broken network." EXPERT REVIEW OF VACCINES. ENGLAND FEB 2003, vol. 2, no. 1, February 2003 (2003-02), pages 61-73, XP002277232 ISSN: 1476-0584 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7371726B2 (en) 2003-12-31 2008-05-13 Schering-Plough Animal Health Corporation Neutralizing GDF8 epitope-based growth enhancing vaccine
US7585648B2 (en) 2003-12-31 2009-09-08 Schering-Plough Animal Health Corporation Nucleic acids encoding neutralizing GDF8 epitope-based peptides and fusion proteins
US7892561B2 (en) 2003-12-31 2011-02-22 Schering-Plough Animal Health Corporation Neutralizing GDF8 epitope-based growth enhancing vaccine
US7432079B2 (en) 2004-12-30 2008-10-07 Schering-Plough Animal Health Corporation Plant virus coat fusion proteins with GDF8 epitopes and vaccines thereof
EP2344184A2 (fr) * 2008-10-07 2011-07-20 Biovest International, Inc. Procédés d'induction d'une réponse immunitaire prolongée contre un idiotype de cellules b, au moyen de vaccins autologues anti-idiotypiques
EP2344184A4 (fr) * 2008-10-07 2012-08-29 Biovest Int Inc Procédés d'induction d'une réponse immunitaire prolongée contre un idiotype de cellules b, au moyen de vaccins autologues anti-idiotypiques
CN102184348A (zh) * 2011-01-12 2011-09-14 湖北大学 基于自然生殖过程的昆虫遗传群体的模拟方法
CN102184348B (zh) * 2011-01-12 2013-10-02 湖北大学 基于自然生殖过程的昆虫遗传群体的模拟方法
EP2771349B1 (fr) * 2011-09-16 2020-02-26 Iogenetics, LLC. Procédés bio-informatiques de détermination de liaisons peptidiques
US9725768B2 (en) 2012-08-31 2017-08-08 Biovest International, Inc. Methods for producing high-fidelity autologous idiotype vaccines
CN113383006A (zh) * 2018-02-19 2021-09-10 高丽大学校产学协力团 包含热休克蛋白表位的疫苗及其用途
WO2021163239A1 (fr) * 2020-02-11 2021-08-19 Ubi Ip Holdings Immunogènes peptidiques ciblant un polypeptide amyloïde d'îlots (iapp) pour la prévention et le traitement de troubles liés à un iapp agrégé

Also Published As

Publication number Publication date
WO2004052930A3 (fr) 2004-07-29
MXPA05006113A (es) 2005-09-30
CA2505870A1 (fr) 2004-06-24
EP1572740A2 (fr) 2005-09-14
NO20053363D0 (no) 2005-07-11
AU2003285280A1 (en) 2004-06-30
NO20053363L (no) 2005-07-11

Similar Documents

Publication Publication Date Title
US7070784B1 (en) Method for down-regulating GDF-8 activity using immunogenic GDF-8 analogues
JP4446601B2 (ja) B群連鎖球菌の糖質を免疫学的に模倣する抗イディオタイプ抗体を含むワクチン処方物
JP2002525060A (ja) オステオプロテゲリンリガンド活性をダウンレギュレートする方法
AU2006312847A1 (en) Therapeutic vaccines targeting HMGB1
TW202039587A (zh) 供合成胜肽免疫原作為免疫刺激劑的人工混雜t輔助細胞抗原決定位
EP1687333A2 (fr) Procede de regulation negative du facteur de croissance endotheliale vasculaire
US6746669B1 (en) Method for down-regulating IL5 activity
JP4875497B2 (ja) バクテリオファージナノ粒子を含む方法および組成物
EP1572740A2 (fr) Epitopes uniques de ciblage
ZA200302628B (en) Method for down-regulating IgE.
EP1448598A2 (fr) Mimetiques immunogenes de proteines multimeres
US20080253993A1 (en) Immunogenic Egfr Peptides Comprising Foreign T Cell Stimulating Epitope
US20040037840A1 (en) Novel therapeutic vaccine formulations
ZA200509031B (en) Immunogenic human TNF alpha analogues with reduced cytotoxicity and methods of their preparation
US20060222624A1 (en) Detoxified TNF and method of preparing
CA2440197A1 (fr) Agents de vaccin synthetiques
AU2002233166A1 (en) Synthetic vaccines comprising polyhydroxypolymer carriers
KR20050085605A (ko) 단일 에피토프의 표적화
ZA200502927B (en) Single chain recombinant T cell receptors.
Klein et al. Applications of immunochemistry in human health: advances in vaccinology and antibody design (IUPAC Technical Report)
CN101260152A (zh) 负调节Osteoprotegerin配体活性的方法
AU2002342596A1 (en) Immunogenic mimetics of multimer proteins with promiscuous T cell epitope inserts

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 168533

Country of ref document: IL

Ref document number: 2505870

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2003285280

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 973/KOLNP/2005

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 200504414

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: PA/a/2005/006113

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2003778264

Country of ref document: EP

Ref document number: 2005502302

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 1020057010743

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 20038A60230

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 541097

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 200500943

Country of ref document: EA

WWP Wipo information: published in national office

Ref document number: 1020057010743

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2003778264

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2003778264

Country of ref document: EP