EP1370661A2 - Epitopes lymphocytaires t des proteines l1 et e7 de papillomavirus et leur utilisation a des fins de diagnostic et de therapie - Google Patents

Epitopes lymphocytaires t des proteines l1 et e7 de papillomavirus et leur utilisation a des fins de diagnostic et de therapie

Info

Publication number
EP1370661A2
EP1370661A2 EP01998642A EP01998642A EP1370661A2 EP 1370661 A2 EP1370661 A2 EP 1370661A2 EP 01998642 A EP01998642 A EP 01998642A EP 01998642 A EP01998642 A EP 01998642A EP 1370661 A2 EP1370661 A2 EP 1370661A2
Authority
EP
European Patent Office
Prior art keywords
cells
cell
cell epitope
peptide
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP01998642A
Other languages
German (de)
English (en)
Inventor
John Nieland
Andreas Kaufmann
Manuela Schinz
Angela Kather
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Medigene AG
Original Assignee
Medigene AG
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 Medigene AG filed Critical Medigene AG
Publication of EP1370661A2 publication Critical patent/EP1370661A2/fr
Withdrawn legal-status Critical Current

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants

Definitions

  • T cell epitopes of the papillomavirus Ll and E7 proteins and their use in diagnostics and therapy
  • the present invention relates to a papilloma virus T-cell epitope having an amino acid sequence YLPPVPVSKWSTDEY ART, STDEYVARTNIYYHAGTSRL, VGHPYFPIKKPNNNKILVPK, GLQYRVFRIHLPDPNKFGFP, WACVGVEVGRGQPLGVGISG, QPLGVGISGHPLLNKLDDTE, QLCLIGCKPPIGEHWGKGSP, LELINTVIQDGDMVDTGFGA, DMVDTGFGAMDFTTLQANKS, VTVVDTTRSTNMSLCAAIST, TTYKNTNFKEYLRHGEEYDL, IFQLCKITLTADVMTYIHSM, PPPGGTLEDTYRFVTSQAIA, RFVTSQAIACQKHTPPAPKE, LKKYTFWEV LKEKFSADLD, PLGRKFLLQAGMHGDTPTLH, YCYEQLNDSSEEEDEIDGPA, VGNPYFRVPAGGG
  • NNGVCWHNQLFVTVVDTTRS PPPPTTSLVDTYRFVQSVAI, YRFVQSVAITCQKDAAPAEN, PYDKLKFWNVDLKEKFSLDL, YPLGRKFLVQAGMHGPKATL, MHGPKATLQDIVLHLEPQNE, VDLLCHEQLSDSEEENDEID, SEEENDEIDGVNHQHLPARR, SSADDLRAFQQLFLNTLSFV, NTDDYVTRTSIFYHAGSSRL, FYHAGSSRLLTVGNPYFRVP, PQRHTMLCMCCKCEARIKLV, GMHGPKATL, HGPKATLQDI, MHGPKATL or FQQLFLNTL
  • the papilloma viruses also called wart viruses, are double-stranded DNA viruses with a genome size of about 8000 base pairs and an icosahedral capsid with a diameter of approx. 55 nm.
  • HPV human pathogenic papilloma virus
  • the genome of the papillomaviruses can be divided into three areas:
  • the first area concerns a non-coding region that contains regulatory elements for the transcription and replication of the virus.
  • the second region so-called E (early) region, contains various protein-coding sections E1-E7, of which, for example, the E6 and E7 proteins are responsible for the transformation of epithelial cells and the E1 protein controls the DNA copy number.
  • the E6 and E7 regions are so-called oncogenes, which are also expressed in malignant cells.
  • the third region also known as the L (late) region, contains two protein-coding sections L1 and L2 which code for structural components of the virus capsid.
  • L1 protein is understood to mean the main capsid protein of the papillomaviruses (Baker T. et al. (1991) Biophys. J. 60, 1445).
  • HPV-16 has been linked to cervical cancer (cervical cancer).
  • HPV-16 is the main risk factor for the formation of cervical neoplasia.
  • the immune system plays an important role in the progression of the disease. Cellular immune responses and, in particular, antigen-specific T-lymphocytes are presumably important for the defense mechanism.
  • the E7 gene is constitutive in highly malignant cervical intra-epithelial neoplasia (CIN II / III) and cervical tumors is expressed in all layers of the infected epithelium. Therefore, the E7 protein in particular is regarded as a potential tumor antigen and as a target molecule for activated T cells (see, for example, WO 93/20844).
  • the E7-induced cellular immune response in the patient does not appear to be strong enough to influence the course of the disease.
  • the immune response may be boosted by appropriate vaccines.
  • the expression of the Ll gene or the coexpression of the Ll and L2 gene can lead to the formation of capsomers, stable capsomers, capsids or virus-like particles (VLPs for virus-like particles) (see for example WO 93/02184, WO 94/20137 or WO 94/05792).
  • Capsomers are understood to be an oligomeric configuration that is made up of five Ll proteins.
  • the capsomer is the basic building block from which viral capsids are built.
  • Stable capsomers are capsomers that are not able to form capsids.
  • Capsids are understood to mean the shell of the papillomavirus, which is composed, for example, of 72 capsomers (Baker T. et al. (1991) Biophys. J. 60, 1445).
  • VLP is a capsid that is morphologically and antigenically similar to an intact virus.
  • the VLPs could be used in various animal systems to elicit a humoral immune response, which is characterized by the formation of neutralizing antibodies.
  • the formation of virus-neutralizing antibodies against Ll and / or L2 protein is of less clinical importance if the viral infection has already taken place, since for the elimination of virus-infected cells no antibodies, but a virus-specific cytotoxic T cell - (CTL) response appears to be necessary.
  • CTL cytotoxic T cell -
  • CVLPs for chimeric virus-like particles
  • the CVLPs only triggered a minor humoral immune response against the E7 protein (Müller, M. et al. (1997), supra).
  • some of the CVLPs tested actually induce the desired E7-specific cytotoxic T cell response in mice (see also Peng S. et al. (1998) Virology 240, 147-57).
  • CVLPs are therefore This is of interest both for the development of a vaccine and for the treatment of already existing infections and resulting tumors, since the E7 peptides of tumor cells presented via MHC molecules of class I would represent target molecules of cytotoxic T cells.
  • a vaccine consisting of CVLPs is based on the principle of pseudo-infection of the cells by the CVLPs. This means that the CVLPs enter the cell like viruses, are processed there to form peptides, the peptides are then loaded onto MHC class I and II molecules and ultimately CD8 or CD4 positive T cells are presented.
  • CD8 cells can differentiate into cytotoxic T cells and then produce a cellular immune response
  • CD4 cells develop into T helper cells and stimulate B cells into a humoral or CD8 positive T cells into a cytotoxic immune response and can induce the lysis of infected cells themselves.
  • Small peptides can bind to MHC class I molecules already on the cell surface and then stimulate CD8 or CD4 positive cells to a cellular immune response without further processing.
  • a particular peptide can only be bound by certain MHC molecules. Due to the large polymorphism of the MHC molecules in natural populations, a certain peptide can therefore only be bound and presented by a small part of a population.
  • Presentation within the meaning of the present invention is understood to mean when a peptide or protein fragment binds to an MHC molecule, this binding taking place, for example, in the endoplasmic reticulum, in the extracellular space, the endosomes, proendosomes, lysosomes or protysosomes, and if then this MHC molecule-peptide complex is bound on the extracellular side of the cell membrane, so that it can be specifically recognized by immunoassays. Since CVLPs trigger both a cellular and humoral immune response and are not restricted to MHC, these particles are generally suitable for the development of vaccines, in that the ability to form particles is provided by an Ll component and an additional antigen component in this Ll component is merged.
  • CVLPs When developing such CVLPs, it is absolutely essential to have a functional test system available with which one can directly investigate the immunogenicity of CVLPs. Such a test system should have the property that CVLPs with different antigen contents can be examined with the same test system. Since the cellular immune response is of crucial importance for immunological therapy methods of tumors or viral diseases, the task was to make the cellular immune response caused by type 16 or 18 CVLPs measurable.
  • HPV-16 and HPV-18 T-cell epitopes which, in conjunction with MHC molecules, trigger a cytotoxic T-cell response in vivo and in vitro, for example.
  • the peptides according to the invention therefore have the sequence
  • T cells which recognize a peptide, measurable by the fact that they can be induced to give a T cell response by the peptide, do not necessarily also recognize cells which contain whole proteins - containing the corresponding peptide - were loaded. This can be explained by the fact that peptides often contain protease interfaces, within which the peptides are cut during the processing of the whole proteins in the cell and are therefore destroyed and thus can no longer be recognized by T cells. This problem is, for example, in Feltkamp et al. (1993), Eur. J. Immunol. 23: 2242-2249 confirmed.
  • the present invention therefore relates to a T cell epitope with an amino acid sequence
  • a functionally active variant of the T-cell epitope according to the invention is understood to mean a T-cell epitope which, in a T-cell cytotoxicity test system (see, for example, Examples 2-5 of the present invention), has an effect on the cytotoxicity of the T according to the invention
  • Cell epitope has measured cytotoxicity which corresponds at least to the sum of the mean of the negative controls and three times the standard deviation, preferably of at least approximately 30%, in particular at least approximately 50% and in a particularly preferred manner of at least approximately 80%.
  • a preferred variant is, for example, a T cell epitope with a sequence homology to the T cell epitopes according to the invention of at least approximately 65%, preferably at least approximately 15% and in particular at least approximately 85%) at the amino acid level.
  • Other preferred variants are also T cell epitopes which have a structural homology to the T cell epitopes according to the invention.
  • Such epitopes can be found by generating specific T cells against the T cell epitopes according to the invention (DeBruijn ML et al. (1991) Eur. J. Immunol. 21, 2963-70; and DeBruijn ML (1992) Eur. J. Immunol.
  • T cell epitopes are understood to mean cytotoxic T cell epitopes or T helper cell epitopes (T H , T HI or T H2 ).
  • non-cytotoxic T cells are also known which can also recognize MHC I molecules, so that non-cytotoxic T cell epitopes are also included as a variant of the present invention.
  • Another embodiment of the present invention is a T cell epitope that is part of a compound, which compound is not a naturally occurring Ll protein of a papillomavirus and in the case of an HPV-16 T cell epitope is not an exclusively N-terminal or exclusively C-terminal deletion mutant of a naturally occurring Ll protein of a papillomavirus.
  • the compound can be a fusion of the same or different T cell epitopes according to the invention.
  • a T-cell epitope according to the invention and / or a functionally active variant can be contained in an L1 protein of another papillomavirus or in a chimeric L1 protein, for example an HPV18L1E7 or HPV16LlE7 fusion protein.
  • a compound of the invention may have the ability to form CVLPs.
  • the T cell epitope mentioned can preferably be part of a compound, a polypeptide which preferably contains further amino acid sequences, in particular a fusion protein.
  • the compound can be a polypeptide of at least approximately 50 amino acids, preferably of at least approximately 35 amino acids, in particular of at least approximately 20 amino acids, and in a particularly preferred manner of at least approximately 9-12 amino acids in length.
  • the compound In order to detect the compound or to modify its binding activity to T cells, it can contain a chemical, radioactive isotope, non-radioactive isotope, and / or fluorescent label of the T cell epitope and / or of the fusion protein mentioned.
  • Examples of chemical substances known to the person skilled in the art which are suitable for a chemical labeling according to the invention are: biotin, FITC (fluorescein isothiocyanate) or streptavidin.
  • biotin FITC (fluorescein isothiocyanate) or streptavidin.
  • FITC fluorescein isothiocyanate
  • streptavidin streptavidin.
  • a possible embodiment is that a peptide is modified such that it contains at least one lysine.
  • Biotin or FITC (fluorescein isothiocyanate) is coupled to this lysine in the manner known to the person skilled in the art.
  • the modified peptide is bound to a corresponding MHC molecule or to a cell with corresponding MHC molecules.
  • the peptide can then be detected via labeled avidin or streptavidin or directly via the fluorescence of the FITC.
  • isotopes known to the person skilled in the art which are suitable for radioactive isotope labeling according to the invention are: H, I, I, P, P or
  • isotopes known to the person skilled in the art which are suitable for non-radioactive isotope labeling according to the invention are: 2 H or 13 C.
  • fluorescent substances known to the person skilled in the art which are suitable for a fluorescent label according to the invention are: 152 Eu, fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phaldehyde or fluorescamine.
  • Polypeptides according to the invention with an amino acid length of approximately 50 can be produced, for example, by chemical peptide synthesis. Longer polypeptides are preferably genetically engineered.
  • the present invention therefore furthermore relates to a nucleic acid, for example for the expression of the T cell epitope or the compounds mentioned, which contains, for example, the following components: (a) at least one regulatory element and (b) at least one nucleic acid which is responsible for an amino acid sequence encodes the compound of the invention.
  • the nucleic acid construct mentioned is preferably made of DNA or RNA.
  • Suitable regulatory elements allow, for example, the constitutive, regulatable, tissue-specific, cell cycle-specific or metabolically specific expression in eukaryotic cells or the constitutive, metabolically specific or regulatable expression in prokaryotic cells.
  • Adjustable elements according to the present invention are promoters, activator sequences, enhancers, silencers, and / or repressor sequences.
  • Suitable regulatable elements which enable constitutive expression in eukaryotes are promoters which are recognized by RNA polymerase III or viral promoters such as CMV enhancers, CMV promoters, SV40 promoters and viral promoter and activator sequences derived from, for example, HBV , HCV, HSV, HPV, EBV HTLV or HIV.
  • regulatable elements which enable regulatable expression in eukaryotes are the tetracycline operator in combination with a corresponding repressor (Gossen M. et al (1994) Curr. Opin. Biotechnol. 5, 516-20).
  • controllable elements which enable cell cycle-specific expression in eukaryytes are the promoters of the following genes: cdc25C, cyclin A, cyclin E, cdc2, E2F, B-myb or DHFR (Zwicker J. and Müller R. (1997) Trends Genet 13, 3-6).
  • the nucleic acid according to the invention can also be used, for example, for the production of a DNA vaccine.
  • the coding sequences for these epitopes as well as for other already known epitopes of papillomaviruses can be joined together in any order directly or with several nucleotides in between in an open reading frame.
  • DNA sequences foreign to papillomavirus can be added, which are used, for example, for intracellular targeting of the resulting polypeptide chain, in particular into the endoplasmic reticulum, into the endosomes or the lysomes.
  • the nucleic acid can be present as a plasmid, as part of a viral or non-viral vector.
  • Another object of the present invention is therefore a vector, in particular an expression vector, which contains a nucleic acid according to the invention. holds.
  • the following are particularly suitable as viral vectors: baculoviruses, vaccinia viruses, adenoviruses, adeno-associated viruses and herpes viruses.
  • baculoviruses baculoviruses, vaccinia viruses, adenoviruses, adeno-associated viruses and herpes viruses.
  • virosomes virosomes, liposomes, cationic lipids, or poly-lysine conjugated DNA.
  • Another object of the present invention is also preferably a cell that contains at least one T cell epitope.
  • the cell is transfected, transformed or infected by one of the vectors according to the invention.
  • this cell expresses the polypeptide according to the invention under conditions known to the person skilled in the art which lead to the activation of the regulatable elements used in each case.
  • the polypeptide can then be isolated from this cell and e.g. be cleaned using one of the above markers.
  • Procaryotic and eukaryotic cells in particular bacterial cells such as E. coli, yeast cells such as S.
  • insect cells such as Spodoptera frugiperda cells (Sf-9) or Trichoplusia ni cells or mammalian cells such as COS cells are suitable for genetic engineering or HeLa cells.
  • the expressed compounds according to the invention can then be purified by standard methods.
  • a preferred embodiment is to use the cell itself which expresses the polypeptide according to the invention, in a particularly preferred embodiment the cell presenting parts of the polypeptide according to the invention via MHC-1 molecules on the cell surface.
  • antigen-presenting cells such as B cells, macrophages, dendritic cells, fibroblasts or other HLA A2.01 positive cells, in a preferred embodiment JY, T2, CaSki cells or EBV-transformed B cells Cell lines (BLCL).
  • the invention Cells according to the invention which present a polypeptide containing a T cell epitope can be used as target cells for restimulating immune cells, in particular T cells and / or for measuring the activation of T cells.
  • Target cell in the sense of the present invention is to be understood as a cell which
  • a T cell epitope is presented via MHC molecules and thus specifically induces T cell activation, in particular a cytotoxic T cell reaction against the cell.
  • the compound containing a T cell epitope can be part of a complex which is characterized in that the compound is covalent or via hydrophobic interactions, ion binding or hydrogen bonding with at least one further species such as peptides, proteins, peptoids, linear or branched oligo - Or polysaccharides and nucleic acids is connected.
  • the present invention therefore relates to a complex containing a T cell epitope or a compound and at least one further compound.
  • a preferred embodiment is that the polypeptide is present in connection with MHC class I molecules, for example as HLA A2.01 (or also HLA AI, HLA A24 etc.) tetramer. Human MHC class I molecules are particularly preferred.
  • HLA A2.01 tetramers can be prepared with the corresponding bound peptides, which are able to bind to the T cell receptors of peptide-specific cytotoxic T cells.
  • a further embodiment is the immobilization of the compound according to the invention or of the complex mentioned on carrier materials.
  • Suitable carrier materials are, for example, ceramic, metal, in particular noble metal, glasses, plastics, crystalline materials or thin layers of the carrier, in particular of the materials mentioned, or (bio) molecular filaments such as cellulose or framework proteins.
  • a component of the complex can additionally contain a protein tag.
  • Protein tags according to the invention allow, for example, high affinity absorption to a matrix, stringent washing with suitable buffers without eluting the complex to any appreciable extent, and subsequent targeted elution of the absorbed complex.
  • Examples of protein tags known to the person skilled in the art are a (HIS) 6 tag, a Myc tag, a FL AG tag, a hemaglutenin tag, glutathione transferase (GST) tag, intein with an affinity chitin tag binding tag or maltose binding protein (MBP) tag.
  • the protein tags according to the invention can be N-, C-terminal, and / or internal.
  • Another object of the present invention is a method for in vitro detection of the activation of T cells by at least one compound containing a T cell epitope.
  • a method for in vitro detection of the activation of T cells by at least one compound containing a T cell epitope preferably consists of three steps: a) In a first step, cells are stimulated with at least one T cell epitope according to the invention, preferably with at least one compound containing a T cell epitope according to the invention.
  • This compound can contain at least one compound according to the invention containing a T cell epitope, at least one complex according to the invention containing a T cell epitope, at least one capsomer, at least one stable capsomer, at least one VLP, at least one CVLP, and / or at least mean a virus.
  • immune cells are stimulated by incubation with CVLPs.
  • This stimulation can take place, for example, in the form of a vaccination or by incubating immune cells with CVLPs in vitro.
  • Immune cells stimulated in this way are, for example, after a vaccination or in a tumor patient obtained from the blood, from tumors or from lymph nodes, and / or are cultivated.
  • a) cells with at least one T cell epitope according to the invention, at least one compound according to the invention containing a T cell epitope, at least one target cell presenting a T cell epitope and / or with at least one incubated according to the invention.
  • the activation of T cells is determined. Appropriate methods for this are, for example, the detection of
  • cytokine assay Chokine assay (Chapters 6.2 to 6.24 in Current Protocols in Immunology (1999), edited by Coligan J.E., Kruisbeek A.M., Margulies
  • DH Shevach EM and Strober W., John Wiley & Sons
  • ELISPOT Chapter 6.19 in Current Protocols in Immunology, supra
  • a 5I Cr release test Chapter 3.11 in Current Protocols in Immunology, supra
  • evidence of proliferation Chapter 3.12 in Current Protocols in Immunology, supra.
  • immune cells such as cytotoxic T cells, T helper cells, B cells, NK cells and other cells.
  • the use of compounds, complexes and / or cells according to the invention which contain the markings according to the invention allows the detection of T cells which recognize the T cell epitope by detecting the
  • Binding of labeled compounds, complexes and / or cells to the T cells In a preferred embodiment, the binding of MHC-polypeptide complexes according to the invention to the surface of the T cells is detected. This can be carried out in such a way that the MHC complexes themselves are labeled, for example fluorescence-labeled, or that in a further step an MHC-specific, Kiert, for example fluorescence-labeled antibodies used to in turn detect the MHC complexes. The fluorescent labeling of the T cells can then be activated, for example, in a luorescens
  • T cells cytokine assay, Elispot, 5I Cr release test, proliferation, see above.
  • coreceptors eg CD28
  • anti-CD28 coreceptor-specific antibodies
  • IL-2 non-specific activators
  • the present invention also relates to a method which contains an additional step a ') which is introduced after step a).
  • a ') In this additional step a) following step a)), the isolated or cultured cells are loaded with at least one target cell with at least one T cell epitope according to the invention, with at least one compound according to the invention containing a T cell epitope, at least a complex according to the invention containing a T cell epitope, at least one capsomer, at least one stable capsomer, at least one VLP, at least one CVLP and / or at least one virus, with at least one complex according to the invention containing a T cell epitope, and / or at least one target cell presenting a T cell epitope co-cultivates for at least about 8 weeks, in particular for at least about 1 week, before step b) follows.
  • Another object of the present invention is a method for producing a target cell presenting a T cell epitope. It is possible to load the target cell with combinations of different T cell epitopes.
  • the target cell is incubated with at least one T cell epitope according to the invention, with at least one compound containing a T cell epitope and / or at least one complex containing a T cell epitope.
  • the target cell is incubated in growth medium which contains polypeptides according to the invention or with MHC class I complexes with bound polypeptides according to the invention.
  • the MHC class I complexes can be present, for example, as HLA A2.01 tetramers.
  • a tetramer usually binds four peptides. These can either be identical or represent different species of peptides.
  • the target cell is transfected, transformed and / or infected with a nucleic acid according to the invention and / or a vector according to the invention.
  • the target cell is infected with a vaccinia virus vector.
  • the method according to the invention is carried out with antigen-presenting cells, for example with B cells, macrophages, dendritic cells, embryonic cells or fibroblasts or other HLA-positive cells, in one embodiment. form carried out with JY, T2, CaSki cells or EBV-transformed B cell lines.
  • the CVLPs used contain a Papillomavirus L1 protein or variants thereof, in particular HPV16 L1 protein and, but not necessarily, a protein heterologous to L1 or variants thereof.
  • the two proteins can be bound directly or indirectly.
  • directly bound means that there is a covalent bond between the two proteins, for example a peptide bond or a disulfide bond.
  • Bound indirectly means that the proteins are bound via non-covalent bonds, for example hydrophobic interactions, ionic bonds or hydrogen bonds.
  • the CVLPs contain a papillomavirus L2 protein in addition to L1 protein or variants thereof.
  • a preferred embodiment of the L1 protein of the present invention is, for example, L1 proteins with one or more deletions, in particular a C-terminal deletion.
  • a C-terminal deletion has the advantage that the efficiency of the formation of virus-like particles can be increased, since the nuclear localization signal located at the C-terminus is deleted.
  • the C-terminal deletion is therefore preferably up to approximately 35 amino acids, in particular approximately 25 to approximately 35 amino acids, especially approximately 32 to approximately 34 amino acids.
  • a 32 amino acid C-terminal deletion of the HPV16 L1 protein is sufficient to be able to increase the formation of virus-like particles by at least about ten times.
  • the Ll protein can carry one or more mutations or the Ll portion can be composed of Ll proteins from different papillomaviruses.
  • a common characteristic of the L1 proteins according to the invention is that they allow the formation of VLPs or CVLPs and that they contain at least one T cell epitope according to the invention.
  • the L1 protein or variants thereof and the protein heterologous to L1 is a fusion protein. Also included are heterologous proteins that are composed of several different proteins or parts thereof. Epitopes, for example, can also do this
  • Epitopes in the sense of the invention can also be part of a synthetic polypeptide with a length of approximately 50 amino acids, preferably of at least approximately 35 amino acids, in particular of at least approximately 20 amino acids and in a particularly preferred manner of at least approximately 9 amino acids.
  • proteins which are heterologous to L1 and which are derived from a viral protein for example derived from HIV, HBV or HCV, preferably from papillomaviruses, in particular from human papillomaviruses.
  • this is an E protein of a papilloma virus, preferably an E6 and / or E7 protein. It is particularly preferred if the E protein is a deleted E protein, preferably a C-terminally deleted, in particular a C-terminally deleted E7 protein, since these constructs in connection with deleted L1 protein preferably virus-like particles can train. Deletions of up to 55 amino acids are particularly preferred, preferably approximately 5 to approximately 55 amino acids, in particular approximately 38 to approximately 55 amino acids.
  • the protein heterologous to L1 can be derived from antigens that are not viral pathogens. They can also be derived from autoimmune antigens such as thyroglobulin, myelin basic protein or zona pellucida glycoprotein 3 (ZP 3 ), which are associated with certain autoimmune diseases such as thyroiditis, multiple sclerosis, oophoritis or rheumatoid arthritis.
  • autoimmune antigens such as thyroglobulin, myelin basic protein or zona pellucida glycoprotein 3 (ZP 3 ), which are associated with certain autoimmune diseases such as thyroiditis, multiple sclerosis, oophoritis or rheumatoid arthritis.
  • the protein heterologous to L1 originates from tumor antigens, preferably melanoma antigens such as MART, ovarian carcinoma antigens such as Her2 new (c-erbB2), BCRA-1 or CA125, colon carcinoma antigens such as CA125 or breast carcinoma antigens such as Her2 new (c-erbB2), BCRA-1, BCRA-2 ,
  • tumor antigens preferably melanoma antigens such as MART, ovarian carcinoma antigens such as Her2 new (c-erbB2), BCRA-1 or CA125, colon carcinoma antigens such as CA125 or breast carcinoma antigens such as Her2 new (c-erbB2), BCRA-1, BCRA-2 ,
  • Another object of this invention is a method for the in vitro detection of the activation of T cells, which are obtained by preparation from samples.
  • This method makes it possible to determine whether papillomavirus L1 protein-specific cytotoxic T cells are present in a sample, for example a blood sample from a patient or in tumors or lymph nodes of a tumor patient.
  • a detection method contains the following steps: a ") In a first step, cells are obtained, for example by taking blood from a patient or by preparing, for example, tumors or lymph nodes. The cells are then taken up in growth medium and cultured.
  • Suitable methods include the detection of the production or secretion of cytokines by the T cells, the expression of surface molecules on T cells, the lysis of target cells or the proliferation of cells.
  • cytokine assay Chapters 6.2 to 6.24 in Current Protocols in Immunolog y (1999), edited by Coligan JE, Kruisbeek AM, Margulies DH, Shevach EM and Strober W., John Wiley & Sons
  • ELISPOT Chapter 6.19 in Current Protocols in Immunology, supra
  • a 51 Cr release test Chapter 3.11 in Current Protocols in Immunology, supra
  • the proof of proliferation Chapter 3.12 in Current Protocols in Immunology, supra.
  • mun cells such as cytotoxic T cells, T helper cells, B cells, NK cells and other cells can be distinguished.
  • mun cells such as cytotoxic T cells, T helper cells, B cells, NK cells and other cells can be distinguished.
  • the use of compounds, complexes and / or cells according to the invention which contain labels allows the detection of T cells which recognize the T cell epitope.
  • the binding of MHC-polypeptide complexes according to the invention to the surface of the T cells is detected.
  • This can be carried out in such a way that the MHC complexes themselves are labeled, for example fluorescence-labeled, or that in a further step an MHC-specific, labeled, for example fluorescence-labeled antibody is used in order to in turn detect the MHC complexes.
  • the fluorescence labeling of the T cells can then be measured and evaluated, for example, in a luorescent activated cell sorter (FACS).
  • FACS luorescent activated cell sorter
  • cytokine assay Elispot, Cr release test, proliferation, see above.
  • coreceptors e.g. CD28
  • anti-CD28 coreceptor-specific antibodies
  • IL-2 non-specific activators
  • the present invention also relates to a process which contains an additional step a ') which is introduced after step a ").
  • the isolated or cultured cells are included at least one target cell loaded with at least one T cell epitope according to the invention, with a compound according to the invention containing a T cell epitope, at least one complex according to the invention containing a T cell epitope, at least one capsomer, at least one stable capsomer, at least a VLP, at least one CVLP and / or at least one virus, with at least one complex according to the invention containing a T cell epitope, and / or at least one target cell presenting a T cell epitope, cocultivated for at least about 8 weeks, in particular for at least about 1 week, before step b) followed.
  • Epitope at least one capsomer, at least one stable capsomer, at least one VLP, at least one CVLP, and / or at least one virus,
  • the invention further relates to a test system (kit) for in vitro detection of the activation of T cells, comprising: a) at least one T cell epitope according to the invention, at least one compound according to the invention, at least one vector according to the invention, at least one cell according to the invention, and / or at least one complex according to the invention, and b) effector cells of the immune system, preferably T cells, in particular cytotoxic T cells or T helper cells.
  • the test system is used to determine the Ll protein-specific cytotoxic T cells present, for example, in a patient's blood sample or in tumors or lymph nodes of a tumor patient. In this case, the cells described in b) are in the test
  • control cells contained in the system the activation of which by the first kit component, the substances mentioned under a), serves as the standard.
  • the activation observed in this reaction is compared with the T cell activation of cells isolated from patients by the kit component a).
  • the test system is used, for example, to determine the L1 protein-specific antigenicity of a compound containing a T cell epitope, a complex containing a T cell epitope, a capsomer, a stable capsomer, a VLP, a CVLP , and / or a virus is used.
  • the substances described in a) are control substances whose activating effect on the second kit component, the cells mentioned under b), serves as the standard.
  • the activation observed in this reaction is associated with the activating action of a compound containing a T cell epitope, a complex containing a T cell epitope, a capsomer, a stable capsomer, a VLP, a CVLP, and / or a virus the kit component b) compared.
  • Another object of the invention is the use of at least one T cell epitope according to the invention, at least one compound according to the invention containing a T cell epitope, at least one vector according to the invention containing a nucleic acid coding for a compound containing a T cell epitope, at least a cell according to the invention containing a T-cell epitope for, and / or at least one complex according to the invention containing a T-cell epitope for triggering or for detecting an immune response.
  • Cells which present at least one of the molecules according to the invention via their MHC class I molecules are particularly suitable for stimulating immune cells in vitro and in vivo. Suitable cells for antigen presentation are e.g. B. B cells, dendritic cells, macrophages, fibroblasts or other HLA cells
  • a compound according to the invention for example an HPV18 L1E7 fusion protein, which additionally contains a T cell epitope according to the invention, can be used to detect an immune response.
  • a compound of the invention may have the ability to form CVLPs.
  • Another object of the invention is a medicament or diagnostic agent containing at least one T cell epitope according to the invention, at least one compound according to the invention containing a T cell epitope, at least one vector containing a nucleic acid coding for a T cell epitope Compound, at least one cell according to the invention containing a T cell epitope, and / or at least one complex according to the invention containing a T cell epitope and optionally a pharmaceutically acceptable carrier.
  • a medicament or diagnostic agent according to the invention can be in solution, bound to a solid matrix, and / or admixed with an adjuvant.
  • the drug or diagnostic agent can be administered in various ways.
  • administration forms known to the person skilled in the art are parenteral, local and / or systemic administration by, for. B. oral, intranasal,
  • intravenous, intramuscular, and / or topical application The preferred form of application is influenced, for example, by the natural route of infection of the respective papillomavirus infection.
  • the amount administered depends on the age, weight, general health of the patient and the type of papillomavirus infection.
  • the drug or diagnostic agent can be administered in the form of capsules, solution, suspension, elixir (for oral administration) or sterile solutions or suspensions (for parenteral or intranasal administration).
  • saline or phosphate-buffered saline can be used as the inert and immunologically acceptable carrier.
  • the drug is administered in therapeutically effective amounts. That means amounts sufficient to elicit a protective immunological response.
  • a compound according to the invention for example an HPV 18 L1E7 fusion protein which additionally contains a T cell epitope according to the invention, can be used as a medicament or diagnostic agent.
  • a compound of the invention may have the ability to form CVLPs.
  • FIG. 1 shows the graphic evaluation of two FACScan experiments after restimulation of specific murine T cells with JA WS cells, which present different peptides.
  • the name of the respective peptide is indicated on the X axis, JAWS cells without peptide were only incubated with buffer and served as a negative control.
  • the proportion of CD8-positive T cells which were classified as reactive on the basis of IFN ⁇ expression in the FACScan experiment is plotted on the Y axis.
  • FIG. 2 shows the graphical evaluation of a 51 Cr release experiment after loading RMA cells with the peptide P33 (target cells).
  • the target cells were lysed by T cells (effector cells) stimulated with the peptides P1 to P43, the ratio of the effector cells used to the target cells used being 20.
  • RMA cells incubated with buffer (negative control) or RMA cells incubated with the P33 peptide are plotted on the X axis.
  • The%> of the specifically lysed target cells, determined by the release of 51 Cr from the target cells, are plotted on the Y axis. The% values were calculated using the formula given in Example 4.
  • FIG. 3 shows the graphical evaluation of two FACScan experiments after restimulation of specific murine T cells with LKK cells which present different peptides.
  • the name of the respective peptide is indicated on the X axis, LKK cells without peptide were only incubated with buffer and served as a negative control.
  • the proportion of CD8-positive T cells which were classified as reactive on the basis of IFN ⁇ expression in the FACScan experiment is plotted on the Y axis.
  • FIG. 5 shows the graphical evaluation of a FACScan experiment after restimulation of specific human T cells with donor-identical BLCL which present different HPV 18 peptide pools.
  • the name of the respective peptide pool is indicated on the X axis, "without” stands for BLCL (negative control) incubated only with buffer, L1 or E7 stands for BLCL incubated with HPV18 Ll or HPV18 E7 peptide pool (positive- The proportion of CD4-positive T cells which were classified as reactive on the basis of IFN ⁇ expression in the FACScan experiment is plotted on the Y axis.
  • FIG. 6 shows the graphical evaluation of a FACScan experiment after restimulation of specific human T cells with donor-identical BLCL which present different HPV18 peptide pools.
  • the name of the respective peptide pool is indicated on the X axis, "without” stands for BLCL (negative control) incubated only with buffer, L1 or E7 stands for BLCL incubated with HPV18 Ll or HPV18 E7 peptide pool (positive- The proportion of CD4-positive T cells which were classified as reactive on the basis of IFN ⁇ expression in the FACScan experiment is plotted on the Y axis.
  • FIG. 7 shows the graphic evaluation of a FACScan experiment after restimulation of specific human T cells with donor-identical BLCL, which present the Q9 peptide.
  • the name of the respective peptide is indicated on the X axis, BLCL stands for BLCL (negative control) incubated only with buffer.
  • the proportion of CD8-positive T cells which were classified as reactive on the basis of IFN ⁇ expression in the FACScan experiment is plotted on the Y axis.
  • 8 shows the graphical evaluation of two FACScan experiments after restimulation of specific human T cells with donor-identical BLCL, which present different HPV 16 peptide pools or the P39 peptide.
  • the name of the respective peptide pool (A to H, 1 to 7) or for the respective peptide (P39) is indicated on the X axis, "without” stands for BLCL (negative control) incubated only with buffer, L1 stands for BLCL (positive control) incubated with HPV 16 Ll peptide pool
  • the proportion of CD4-positive T cells is plotted on the Y axis of FIG. 8 which is based on IFN ⁇ expression in the FACScan experiment as reactive and thus were classified as T-helper-1 cells (T HI ).
  • T helper 2 cells T helper 2 cells
  • FIG. 9 shows the graphical evaluation of a FACScan experiment after restimulation of specific human T cells with donor-identical BLCL which present different HPV16 peptide pools.
  • the name of the respective peptide pool is indicated on the X axis, "without” stands for BLCL (negative control) incubated only with buffer, L1 stands for BLCL (positive controls) incubated with HPV16 L1 peptide pool.
  • L1 stands for BLCL (positive controls) incubated with HPV16 L1 peptide pool.
  • On the Y axis the proportion of CD8-positive T cells is plotted which were classified as reactive on the basis of IFN ⁇ expression in the FACScan experiment.
  • FIG. 10 shows the graphical evaluation of a FACScan experiment after re-stimulation of specific human T cells with donor-identical BLCL which present the P33 peptide.
  • the name of the peptide is given on the X-axis, "without” stands for BLCL (negative control) incubated only with buffer, L1 stands for BLCL (positive controls) incubated with HPV16 L1 peptide pool.
  • On the Y-axis is the proportion of CD8-positive T cells which were classified as reactive on the basis of IFN ⁇ expression in the FACScan experiment.
  • 11 shows the graphic evaluation of two FACScan experiments after restimulation of specific human T cells with donor-identical BLCL, which present different HPV 18 peptide pools.
  • the name of the respective peptide pool (A to H, 1 to 7) is indicated on the X axis, "without” stands for BLCL (negative control) incubated only with buffer, L1 or E7 stands for with HPV18 Ll or HPV18 E7 peptide pool incubated BLCL (positive control)
  • the proportion of CD4-positive T cells is plotted on the Y axis of FIG. 11 above, which is based on IFN ⁇ expression in the FACScan experiment as reactive and thus as T -Helfer-1 cells (T HI )
  • T HI T -Helfer-1 cells
  • FIG. 11 below shows the proportion of CD4-positive T cells which were classified as T-HeIfer-2 lines (T H 2) on the basis of IL-4 expression in the FACScan experiment.
  • FIG. 12 shows the graphical evaluation of four FACScan experiments after restimulation of specific human T cells with donor-identical BLCL, which present different HPV 18 peptides or peptide pools.
  • the name of the respective peptide (Q38, Q39, Q46 or Q47) is indicated on the X axis, "without” stands for BLCL (negative control) incubated only with buffer, Ll or E7 stands for with HPV18 Ll or HPV18 E7 peptide pool incubated BLCL (positive control).
  • the proportion of reactive T cells is shown on the Y axis.
  • FIG. 12 top left
  • the proportion of CD4 positive T cells is plotted, which is based on IFN ⁇ expression in the FACScan experiment were classified as reactive and thus as T-helper-1 cells (T H ⁇ ).
  • FIG. 12 shows the proportion of CD 8 positive T cells that were classified as reactive cytotoxic T cells based on IFN ⁇ expression in the FACScan experiment.
  • FIG. 12 shows the proportion of CD4-positive T cells which were classified as T helper 2 cells (T ⁇ _) on the basis of IL-4 expression in the FACScan experiment.
  • FIG. 13 shows the graphical evaluation of a FACScan experiment after restimulation of specific human T cells with donor-identical BLCL which present different HPV16 peptide pools.
  • the name of the respective peptide pool (A to H, 1 to 7) is indicated on the X axis, "without” stands for BLCL (negative control) incubated only with buffer, L1 or E7 stands for with HPV16 Ll or HPV16 E7 peptide pool incubated BLCL (positive control), the proportion of CD8-positive T cells is plotted on the Y axis, which were classified as reactive and thus as cytotoxic T cells based on IFN ⁇ expression in the FACScan experiment ,
  • FIG. 14 shows the graphical evaluation of two FACScan experiments after restimulation of specific human T cells with donor-identical BLCL, which present different HPV 18 peptide pools or a peptide.
  • the name of the respective peptide pool (A to H, 1 to 7) or of the peptide Q30 is indicated on the X axis, "without” stands for BLCL (negative control) incubated only with buffer, L1 or E7 stands for with HPV18 Ll or HPV18 E7 peptide pool inhibited BLCL (positive control).
  • T HI T-helper-1 cells
  • T H2 T-helper 2 cells
  • 15 shows the graphical evaluation of two FACScan experiments after restimulation of specific murine T cells with RMA cells which present different peptides. The name of the respective peptide is indicated on the X axis, RMA cells without peptide were only incubated with buffer and served as a negative control.
  • the proportion of CD8-positive T cells which were classified as reactive on the basis of IFN ⁇ expression in the FACScan experiment is plotted on the Y axis.
  • FIG. 16 shows the graphical evaluation of a 51 Cr release experiment after loading RMA (top) or LKK cells (bottom) with the peptides Q43, Q44 or Q49 (target cells).
  • the target cells were lysed by T cells (effector cells) stimulated with the HPV18 L1 and E7 peptide pools, the ratio of the effector cells used to the target cells used being 20.
  • the cell type and the peptide are indicated on the X axis, the cell acting as a negative control without a peptide.
  • The% of the specifically lysed target cells, determined by the release of 51 Cr from the target cells, are plotted on the Y axis. The% values were calculated using the formula given in Example 4.
  • a T cell line was generated by in vitro vaccination with H7.1-A2 cells. In the ELISpot assay, it was tested against autologous PBMC containing the HPV18 E7 86 peptide. 9 were loaded, tested.
  • T co stands for a negative control with unspecific T cells
  • PBMC co stands for a negative control with PBMC that were not loaded with peptide. 17 below: Cytolytic activity of HPV18E7 86 . 9 specific T cells. T cell lines from two healthy blood donors were raised against the HPV18 E7 86 epitope.
  • cytotoxicity test 51 chromium release assay
  • BLCL co stands for a negative control of BLCL that were not pulsed with peptide.
  • Fig. 18 above Natural processing rank of the HP VI 8 E7 86 epitope. 9 by dendritic cells. T cell lines against HPV 18 E7 were generated by in vitro vaccination with protein or peptide pool-loaded autologous dendritic cells. In the ELISpot assay the stimulation of HPV18E7 was 86 . 94 specific T cells measured by epitope-loaded dendritic cells (DC).
  • DC epitope-loaded dendritic cells
  • T co stands for a negative control with unspecific T cells
  • DC co stands for a negative control of DCs that were not loaded with peptide.
  • Fig 18 below: Presence of HPV 18 E7 86 . 94 specific T cells in populations of tumor infiltrating lymphocytes. TIL of an HPV 18 positive and HLA-A2 positive patient were expanded in vitro and in the ELISpot assay for reactivity with the HPV 18 E7 86 . 9 epitope tested.
  • T co stands for a negative control with unspecific T cells, K-A2 for a negative control with K-A2 cells that were not loaded with peptide.
  • PBMC peripheral blood mononuclear cells
  • BLCL means B cell lines transformed with the help of the Epstein-Barr virus, which were produced individually for each blood donor (obtained from Dr. Andreas Kaufmann, Jena, Germany).
  • CVLP-stimulated murine T cells were obtained as follows: 354Multiple C57BL / 6 or C 3 H mice were injected twice with 20 ⁇ g each
  • mice The spleens from non-vaccinated mice were removed and the spleen cells were resuspended according to standard methods.
  • APC is related to cells for antigen presenting cells.
  • JA WS cells were purchased from ATCC (CRL-11904).
  • RMA cells come from a thymoma of a C57BL / 6 mouse (see Ljunggren H.G. and Karre K. (1985) J. Exp. Med. 162, 1745-59).
  • ⁇ -mouse CD8 / PE means a monoclonal antibody which is directed against the extracellular part of the murine CDS and contains the fluorescence marker phycoerithrin (Pharmingen, Heidelberg, Germany).
  • ⁇ -mouse CD4 / Cychrome means a monoclonal antibody which is directed against the extracellular part of the murine CD4 and contains the fluorescent marker Cychrome (Pharmingen, Heidelberg, Germany).
  • ⁇ -mouse IFN ⁇ / FITC means a monoclonal antibody which is directed against the murine interferon ⁇ and contains the fluorescent marker FITC (Caltag, Hamburg, Germany).
  • ⁇ -human CD8 / APC means a monoclonal mouse antibody which is directed against the extracellular part of human CD8 and contains the fluorescent marker APC (Caltag, Hamburg, Germany).
  • ⁇ -human CD4 / PerCP means a monoclonal mouse antibody which is directed against the extracellular part of human CD4 and contains the fluorescent marker PerCP (Becton Dickinson, Hamburg, Germany).
  • ⁇ -human IFN ⁇ / FITC means a monoclonal mouse antibody that is directed against human interferon ⁇ and contains the fluorescent marker FITC (Caltag, Hamburg, Germany).
  • ⁇ -human IL4 / PE means a monoclonal mouse antibody which is directed against human interleukin 4 and contains the fluorescent marker phycoerithrin (Caltag, Hamburg, Germany).
  • Human GM-CSF (Leukomax) was obtained from Novartis Pharma GmbH (Nuremberg, Germany).
  • Human IL4 was obtained from Becton Dickinson (Hamburg, Germany).
  • Human IL2 was obtained from Becton Dickinson (Hamburg, Germany).
  • FACScan calibur means 'fluorescens activated cell sorter'; the equipment was purchased from Becton Dickenson (Hamburg, Germany).
  • HPV16 L1 peptide pools mean the mixture of peptides P1 to P43, HPV16 E7 peptide pools the mixture of peptides P44 to P51.
  • HPV 18 L1 peptide pools are understood to mean the mixture of peptides Q1 to Q43, and HPV18 E7 peptide pools are the mixture of peptides Q44 to Q52.
  • the genome of HPV 18 is under GenBank Accession No. X05015 is generally available and has been published by Cole and Danos ⁇ J. Mol. Biol. 1987, 193 (4), 599-608).
  • the sequence used for the construction of the fusion proteins according to the invention had the following changes in the Ll gene: At positions 89, 848, 1013 and 1230 of the Ll gene, a C was replaced by a G at the DNA level. At the protein level, the first three changes result in an exchange from Pro to Arg, while the last mutation does not result in a change at the protein level.
  • the E7 gene corresponds to the published sequence.
  • Another method of obtaining the desired nucleic acids is to isolate the papilloma virus-specific genes directly from warts or tumors by means of PCR. Suitable primers for the E6 and E7 genes from HP VI 6 and HPV 18 are e.g. in WO93 / 21958. Further references for the desired nucleic acids are, for example, Kirnbaum, R. et al. (1994), J. Virol., 67, 6929- 6936 and the clones mentioned above in the EMBL database.
  • the first primer has the sequence 5 '-ACC AGA CTC GAG ATG GCT TTG TGG CGG CCT AGT GAC-3'
  • the first primer 5 encodes an Xhol restriction enzyme site.
  • the second primer 5 'encodes an EcoRV restriction enzyme site.
  • a TAA translation stop codon follows the EcoRV site to delete the last 35 amino acids of the HPV18L1 ORF.
  • the PCR product was digested with XhoI / EcoRV and ligated into the vector pBluescript ®, also digested with XhoI / EcoRV.
  • HPV18L1 ⁇ C D I pKS was used to generate the ORF of HPV18E7I.53DI and HPV18E7 1 . 60D ⁇ to clone in the EcoRV site.
  • the PCR products were cleaved with EcoRV and inserted into the EcoRV site of the modified Ll gene.
  • Sf9 Spodoptera frugiperda
  • TNM-FH insect medium Life Technologies, Düsseldorf
  • Recombinant baculoviruses were generated by cotransfection of 5 ug of the recombinant plasmids and 1 ug of linearized Baculo-Gold ® sêtm DNA (Pharmingen, San Diego, CA) into Sf9 cells.
  • Recombinant viruses were purified by end point dilution and / or plaque separation.
  • 10 6 Sf9 cells were infected with recombinant baculovirus and a moi (“multiplicity of infection”) of 0.5 and 1 for 48 h and.
  • the medium was removed and the cells with PBS (140 mM NaCl, 2.7 mM Kcl, 8.1 mM Na 2 PO 4 , 1.5 mM KH 2 PO 4 , pH 7.2).
  • PBS 140 mM NaCl, 2.7 mM Kcl, 8.1 mM Na 2 PO 4 , 1.5 mM KH 2 PO 4 , pH 7.2.
  • the cells were then analyzed by FACS measurement. tested or lysed in SDS sample buffer and tested by SDS gel chromatography and immunoblot assay.
  • the homogenate was clarified by centrifugation at 10,000 rpm in the Sorvall SS34 rotor.
  • Purification of the L1E7 protein from the clarified crude extract for the immunological assay was carried out by ammonium sulfate precipitation at 35-40% saturation and subsequent anion-exchange chromatography on Fractogel TMAE ® (Merck, Darmstadt), said CVLPs in linear salt gradient at 300- 400mM NaCl be eluted.
  • the protein content of the purified fractions was determined by the Bradford method using bovine serum albumin as the standard.
  • Murine T cells (4x10 5 ) from HPV16Ll ⁇ C * E7 ⁇ .
  • 55 CVLP-inoculated C57BL / 6 mice were stimulated and harvested for 5 weeks with HPV16 Ll peptide pools at 37 ° C. with weekly addition of 1 ⁇ g / ml per individual peptide and 10 5 antigen-presenting cells (irradiated splenocytes).
  • the cells were then in 100 ⁇ l medium at 37 ° C. with 1 ⁇ g / ml of the peptides indicated on the X-axis of FIG. 1 and 10 5 antigen-presenting cells (JAWS) restimulated in the presence of 10 IU / ml IL2. Only cells incubated with buffer served as a negative control.
  • JAWS antigen-presenting cells
  • FIG. 1 above for peptides P18, P19, P43 and FIG. 1 below for peptides P35 and P3 shows that the JA WS cells incubated with these peptides restimulate peptide-stimulated CD8-positive murine T- Cells.
  • the peptides P3, P18, P19, P35 and P43 thus contain H2b-restricted cytotoxic T cell epitopes.
  • Target cells which were incubated with culture medium, were used for spontaneous lysis. the.
  • target cells were lysed by adding 0.5% Triton X100.
  • the batches were incubated for 5 hours at 37 ° C. 50 ⁇ l of the supernatant of the batches were placed on luma plates and dried overnight at room temperature. The next morning, the amount of radioactive 51 Cr was determined using a Canberra Packerd B-PIate Counter (counts) and related to the maximally lysed cells of the Triton approach. The% specific lysis was carried out according to the formula:
  • FIG. 2 shows that the RMA cells loaded with the P33 peptide could be effectively lysed by the T cells, but the non-loaded RMA cells could not.
  • the P33 peptide is thus an H2b restricted cytotoxic T cell epitope.
  • Murine T cells (4xl0 5 ) from HPV18 Ll ⁇ CD .E7 ⁇ - 53D ⁇ CVLP and HPV18 Ll cD.E7 ⁇ - 60 Di CVLP-vaccinated C 3 H mice were used for 5 weeks with HPV 18 Ll or E7 peptide pools at 37 ° C stimulated and harvested with weekly addition of 1 ⁇ g / ml per individual peptide and 10 5 antigen-presenting cells (irradiated splenocytes). The cells were then restimulated in 100 ⁇ l medium at 37 ° C. with 1 ⁇ g / ml of the peptides indicated on the X axis of FIG.
  • FIG. 3 shows for peptides Q22, Q23, Q51, Q43 and Q44 and FIG. 4 for peptides Q41 and Q5 that the LKK cells incubated with these peptides restimulate peptide-stimulated murine CD8-positive T Cells.
  • the peptides Q5, Q22, Q23, Q41, Q43, Q44 and Q51 thus contain H2k-restricted cytotoxic T cell epitopes.
  • Human T cells (4 ⁇ 10 5 ) from a non-HLA-typed blood donor were treated with HPN18 L1 ⁇ C DIE7I for 1 week.
  • the 20mer peptides Ql to 52 were according to the matrix
  • the T cells were then restimulated in 100 ⁇ l medium at 37 ° C. with the peptide pools and 10 5 antigen-presenting cells (donor-identical BLCL) in the presence of 10 IU / ml IL2.
  • the amounts of the peptide pools used were such that 1 ⁇ g / ml was added for each individual peptide.
  • FIG. 5 shows that in particular the BLCL incubated with the peptide pools F and 1 brought about a restimulation of CVLP-stimulated human CD4-positive T cells. Furthermore, peptide pools A and 6 showed a restimulation that was clearly above the negative control.
  • the BLCL incubated with the other peptide pools or the negative control or the BLCL incubated with the E7 peptide pool had a small proportion of reactive CD4-positive T cells.
  • the peptide pools F and 1 together contain the peptide Q6, which is therefore responsible for the restimulation of the CVLP-stimulated T cells.
  • the peptide Q6 thus contains a T helper epitope.
  • the peptide pools A and 1 contain Q1, A and 6 Q41 as well as F and 6 Q46 as a common peptide.
  • the peptides Ql, Q41 and Q46 thus contain T helper epitopes.
  • the 20mer peptides P1 to 51 were according to the matrix
  • T cells were then restimulated, fixed, permeabilized and stained as described in Example 6.
  • the evaluation was carried out as described in Example 6.
  • FIG. 6 shows for peptide pools G and 3 that the BLCL incubated with these peptide pools brought about a restimulation of CVLP-stimulated T cells. Furthermore, peptide pools B and C as well as 2 and 4 showed a restimulation that was clearly above the negative control.
  • the PBMC or the negative control incubated with the other peptide pools had a small proportion of reactive CD4-positive T cells, but not the BLCL or the negative control incubated with the other peptide pools or the PBMC incubated with the E7 peptide pool ,
  • the peptide pools G and 3 together contain the peptide P23, B and 2 contain PIO, B and 3 Pl 8, B and 4 P26, C and 2 Pl l, C and 3 P19, C and 4 P27, G and 2 P15 and G and 4 P31. These peptides are therefore each for the restimulation of the CVLP-stimulated T cells
  • the peptides PIO, P1, P15, P18, P19, P23, P26, P27 and P31 thus each contain a T helper epitope.
  • the cells were then restimulated in 100 ⁇ l medium at 37 ° C. with 10 ⁇ g / ml of the HPV 18 Ll peptide Q9 and 10 5 antigen-presenting cells (donor-identical BLCL) in the presence of 10 IU / ml IL2. Only cells incubated with buffer served as a negative control.
  • FIG. 7 shows that the BLCL cells incubated with the peptide Q9 brought about a restimulation of HPV18 L1 peptide pool-stimulated CD8-positive T cells.
  • the peptide Q9 thus contains an HLA A24-restricted cytotoxic T cell epitope.
  • the peptide lYNPETQRL binds to MHC class I molecules of the haplotype HLA A24.
  • the peptide lYNPETQRL is therefore responsible for the restimulation of the T cells by the BLCL cells incubated with the Q9 peptide.
  • the peptide IYNPETQRL contained in peptide Q9 of the overlapping 20mers is an HLA A24 restricted cytotoxic T cell epitope.
  • Example 6 human T cells from a non-HLA-typed blood donor were stimulated and harvested with HPV16Ll ⁇ C * E7 ⁇ - 5 5 CVLPs.
  • T cells were then restimulated in 100 ⁇ l medium at 37 ° C. with the peptide pools from Example 7 and 10 5 antigen-presenting cells (donor-identical BLCL) in the presence of 10 IU / ml IL2.
  • the amounts of the peptide pools used were such that 1 ⁇ g / ml was added for each individual peptide. Only cells incubated with buffer served as negative controls, and cells incubated with HPV 16 L1 peptide pool as positive controls.
  • FIG. 8 shows that the proportion of CD4 and IFN ⁇ positive cells is very high for the BLCL incubated with the peptide pools G and 5 as well as for the BLCL incubated with P39. Since the peptide pools G and 5 as a common Containing peptide P39, which by itself also caused residual simulation of CVLP-stimulated T cells, the peptide P39 contains a T helper epitope. Since CD4 and IFN ⁇ -positive cells are generally T H ⁇ cells, it is therefore a T H i epitope.
  • CD4 and IL4 positive cells are usually T H2 cells.
  • the peptide P39 contained in G and 5 additionally also contains a Tm epitope or is identical to the T ⁇ i epitope and that the peptide 38 contained in F and 5 additionally contains a T ⁇ epitope.
  • Example 6 human T cells from a non-HLA-typed blood donor were stimulated and harvested with HPV16Ll c * E7 ⁇ - 5 CVLPs.
  • T cells were then restimulated in 100 ⁇ l medium at 37 ° C. with the peptide pools from Example 7 and 10 5 antigen-presenting cells (donor-identical BLCL) in the presence of 10 IU / ml IL2.
  • the amounts of the peptide pools used were such that 1 ⁇ g / ml was added for each individual peptide. Only cells incubated with buffer served as negative controls, and cells incubated with HPV 16 Ll pool as positive controls.
  • FIG. 9 shows that the proportion of CD8-positive T cells is high for the BLCL incubated with the peptide pools B, E, G and 1. Since peptide pools B and 1 contain P2 as a common peptide, peptide pools E and 1 contain P5 and G and 1 P7, peptides P2, P5 and P7 each contain a cytotoxic T cell epitope.
  • Example 6 Analogously to Example 6 were human T-cells of a non-HLA-typed blood donor with HPN 16L 1 ⁇ c * E7 1 - stimulated 55 CVLPs and harvested.
  • T cells were then in 100 ⁇ l medium at 37 ° C. with 1 ⁇ g / ml peptide P33 and 10 5 antigen-presenting cells (donor-identical BLCL)
  • FIG. 10 shows that the proportion of CD8-positive cells for the BLCL incubated with the P33 peptide is significantly higher than in the negative control. P33 thus contains a cytotoxic T cell epitope.
  • Example 6 human T cells from a non-HLA-typed blood donor with HPV18L1 ⁇ CDIE7I-53DI CVLPs and HPV18L1 ⁇ CDIE7 1 . 60 DI CVLPs stimulated and harvested at 37 ° C.
  • T cells were then restimulated in 100 ⁇ l medium at 37 ° C. with the HPV18 peptide pools from Example 6 and 10 5 antigen-presenting cells (donor-identical BLCL) in the presence of 10 IU / ml IL2.
  • the amounts of the peptide pools used were such that 1 ⁇ g ml was added for each individual peptide. Only cells incubated with buffer served as negative control, and cells incubated with HPV 18 Ll peptide pool as positive control.
  • FIG. 11 shows that the proportion of CD4 and IFN ⁇ -positive cells is high for the BLCL incubated with the peptide pools F, G, 5 and 6.
  • the peptide pools F and 5 have the peptide Q38, F and 6 the peptide Q46, G and 5 the peptide Q39 and G and 6 the peptide Q47.
  • the peptides Q38, Q39, Q46 and Q47 each contain a T H ⁇ epitope.
  • Fig. 11 shows that the proportion of CD4 and IL4 positive cells for peptide pool G is particularly high, but the proportion of reactive cells for pools 3, 4, 5 and 6 are approximately the same, so that no clear conclusion about the T ⁇ 2 epitopes could be drawn from this experiment.
  • the peptides Q38, Q39, Q46 and Q47 were tested individually in the example below.
  • Example 6 human T cells (4 ⁇ 10 5 ) from a non-HLA-typed blood donor with HPV18L1 ⁇ C DIE7I. S 3DI CVLPs and HPV18L1 ⁇ DIE7I- 6 OD I CVLPs stimulated and harvested.
  • T cells were then in 100 ⁇ l medium at 37 ° C. with 1 ⁇ g / ml peptide Q38, Q39, Q46 or Q47 and 10 5 antigen-presenting cells (donor-identical BLCL) in the presence of 10 IU / ml IL2 restimulated. Only cells incubated with buffer served as negative controls, cells incubated with HPV 18 L 1 or E7 peptide pool as positive controls.
  • FIG. 12 shows that the proportion of reactive CD4 and IFN ⁇ positive T cells is particularly high for the BLCL incubated with the peptides Q38 and Q39, but also significantly higher for the peptides Q46 and Q47 is than in the negative control.
  • Q38, Q39, Q46 and Q47 each contain a T H ⁇ epitope.
  • FIG. 12 shows that the proportion of reactive CD4 and IL4 positive T cells for the peptides Q38 and Q39 is particularly high, so that Q38 and Q39 also contain T H2 epitopes in addition to the T ⁇ i epitopes or that the T HI epitopes are also T H2 epitopes.
  • Fig. 12 shows that the proportion of reactive CD8 and IFN ⁇ positive T cells for peptides Q38, Q39 and Q47 is particularly high, so that peptides Q38, Q39 and Q47 furthermore cytotoxic T cell epitopes contain.
  • the proportion of CD8 and IL4 positive cells for the peptides Q38, Q39, Q46 and Q47 is low and comparable to the negative control, which can be explained by the fact that CD8 positive cells generally do not express IL4.
  • Example 6 Analogously to Example 6 were human T-cells of a non-HLA-typed blood donor with HPN16Ll ⁇ c * E7 ⁇ - stimulated CVLPs 55 and harvested. The T cells were then restimulated in 100 ⁇ l medium at 37 ° C. with the peptide pools from Example 7 and 10 5 antigen-presenting cells (donor-identical BLCL) in the presence of 10 IU / ml IL2. The amounts of the peptide pools used were such that 1 ⁇ g / ml was added for each individual peptide. Only cells incubated with buffer served as negative controls and cells incubated with HPV 16 L1 or E7 peptide pool as positive controls.
  • FIG. 13 shows that the proportion of CD8 and IFN ⁇ -positive T cells is high for the BLCL incubated with the peptide pools A, C, E, F, 1 and 6.
  • the peptide pools A and 1 is the peptide P1,
  • the peptide P46 together.
  • the peptides P1, P3, P5, P6, P41, P43, P45 and P46 each contain a cytotoxic T cell epitope.
  • the T cells were then restimulated in 100 ⁇ l medium at 37 ° C. with the peptide pools from Example 6 and 10 5 antigen-presenting cells (donor-identical BLCL) in the presence of 10 lU / ml IL2.
  • the amounts of the peptide pools used were such that 1 ⁇ g / ml was added for each individual peptide.
  • Only cells incubated with buffer served as negative controls, and cells incubated with HPV 18 L1 or E7 peptide pool as positive controls (FIG. 14 above).
  • the T cells were in 100 ⁇ l medium at 37 ° C. with the peptide pools F and 4 and with 1 ⁇ g / ml peptide Q30 and 10 5 antigen-presenting cells (donor-identical BLCL) in the presence of 10 IU / ml IL2 restimulated.
  • FIG. 14 shows that the proportion of CD4 and IFN ⁇ -positive T cells is high for the BLCL incubated with the peptide pools F and 4. Peptide pools F and 4 share peptide Q30. The peptide Q30 thus contains a T H ⁇ epitope.
  • Example 5 murine T cells from CVLP-vaccinated mice were stimulated and harvested with L1 or E7 peptide pools with the addition of antigen-presenting cells. Subsequently, the cells were incubated with the on the X-axis of FIG. 15 indicated peptides (Q3 and Q4 in Fig. 15 above, L1 7 E7. 1 8, E7 2 - ⁇ , E7 ⁇ - 8 in Fig. 15 below), as well as antigen - presenting cells (RMA) restimulated. Only cells incubated with buffer served as a negative control.
  • RMA antigen - presenting cells
  • Example 5 The experiment was also evaluated analogously to Example 5, in that the reactive CD8-positive cells were determined by antibody staining and FACScan.
  • FIG. 15 above shows for the peptides Q3 and Q4 and FIG. 15 below for the peptides L1 4 4 E7 [. 8 , E7 2 -n and E7 ! - 8 that the RMA cells incubated with these peptides caused restimulation of peptide-stimulated murine CD8 T cells.
  • the peptides Q3, Q4, Ll 47 E7 ! - 8 , E7 2 - u and E7! - 8 thus each contain a H2b restricted cytotoxic T-cell titer. 17. Lysis of HPV18 peptide-loaded RMA / LKK cells
  • RMA or LKK cells were radioactively labeled with 51 Cr and incubated with the peptides plotted on the X axis (Q43 and Q44 in FIG. 16 above, Q49 in FIG. 16 below). Cells without a peptide served as negative controls. T cells which had previously been stimulated with the peptide pool were then added.
  • the lysis of the RMA or LKK cells by the T cells was measured on the basis of the release of the 51 Cr and calculated as in Example 4.
  • FIG. 16 shows that the RMA cells loaded with the Q43 or Q44 (FIG. 16 top) or the LKK cells loaded with the Q49 (FIG. 16 bottom) could be effectively lysed by the T cells, the non-loaded RMA or LKK cells, however, are not.
  • the Q43 and Q44 peptides are thus H2b-restricted cytotoxic T-cell epitopes
  • the Q49 is thus an H2k-restricted cytotoxic T-cell epitope.
  • the HLA binding prediction program of the NIH was used for the prediction of 9-mer sequences with high binding strength to HLA-A * 0201 in the E7 protein of HPV type 18 (address: http: // www- bimas.dcrt. nih.gov/molbio/hla ⁇ _bind/). 9-mers were examined because they are the optimal ligands for HLA class I molecules.
  • To predict de the complete amino acid sequence of the HPV 18 E7 protein was fed into the program and determined that 9-mers for the HLA allele A * 0201 should be predicted.
  • the program provides a list in which the 9-mers found are arranged according to binding strength.
  • the binding strength value is given as the half-life in minutes of dissociation of a peptide of the corresponding amino acid sequence (Tj / 2 ).
  • the predicted peptides were synthesized using standard methods (F moc method). Peptide-specific T cell lines have been established against peptides with high predicted HL A-A2 binding strength. For this purpose, each hole of a 96-well plate of lxlO 5 PBMC from healthy HLA-A2 positive donors was sown in 100 ⁇ l medium (RPMI 1640, 10% heat-inactivated AB plasma). 20 ⁇ g / ml peptide was added to the culture medium. The cells were incubated at 37 ° C. and restimulated once a week with irradiated peptide-loaded autologous PBMC.
  • the stimulator PBMC were incubated for 4 hours in medium with 20 ⁇ g / ml peptide. After the irradiation, they were diluted to 100 lxl0 4 / ul with medium and added to the peptide-stimulated PBMC. Half of the medium was aspirated from each well and 100 ⁇ l of the stimulator-PBMC suspension was added. From the 3rd stimulation, 10 U / ml IL-2 and 10 U / ml IL-7 were added. From the 5th stimulation, restimulation was carried out in the same way with peptide-loaded autologous B-lymphoblastic cell lines (BLCL).
  • BLCL peptide-loaded autologous B-lymphoblastic cell lines
  • HeLa cells were transfected with CD80 and HLA-A2 (H7.1-A2). These cells were irradiated and used to stimulate PBMC at Ixl0 4 / hole. It was restimulated 3 times analogously to peptide stimulation.
  • K-A2 - these are K-562 cells (ATCC CCL-243) were transfected with an HLA A2 expression vector, PBMC or dendritic cells) were in 60 ⁇ l medium (RPMI 1640, 0.4 % »Human albumin) are sown in a well of a 96-well round-bottom cell culture plate and loaded with 50 ⁇ g / ml peptide overnight. The next day, 5 ⁇ 10 4 T cells / well in 60 ⁇ l medium (each with 10 U / ml IL-2 and IL-7) were added and incubated for 4 hours. A Millipore nitrocellulose HA plate was coated overnight at 4 ° C.
  • the antibody Ab-7-B6-l-biotin was added (2 ⁇ g / ml in PBS / BSA, 60 ⁇ l well, Hölzel Diagnostika) and incubated overnight at 4 ° C. The next day, the filter plate was washed again 6 times for 2 minutes with PBS / Tween 20 (0.05%) and streptavidin-AP (50 ng / ml in PBS, Sigma (Deisenhofen), 100 ⁇ l well) was added. After 2 hours of incubation at room temperature, the filter plate was washed 3 times 2 minutes with PBS / Tween 20 (0.05%) and 3 times 2 minutes with PBS.
  • the cytotoxicity of HPV 18 E7 86 . 94 specific T cells were determined in the chromium release assay.
  • Autologous BLCL were used as target cells. 5 ⁇ 10 5 cells were taken up in 100 ⁇ l medium (RPMI 1640, 10% FCS) and 20 ⁇ l 51 chromium (NEN) were added. 50 ⁇ g / ml of the corresponding peptide was added to the peptide loading of the BLCL. This was followed by incubation at room temperature for 2 hours, with the cells being carefully resuspended every 20-30 minutes. The cells were washed 3 times with 5 ml of medium. To sediment the cells, they were centrifuged at 1500 rpm for 5 min and carefully resuspended.
  • the targets were taken up in medium and adjusted to 1 ⁇ 10 5 cells / ml. 40 .mu.l of this cell suspension were added to the effectors and K-562 cells CCL-243, which had already been sown in the wells of a 96-well pointed-bottom cell culture plate.
  • the effectors were harvested and taken up in medium (RPMI 1640, 10% FCS, 10 U / ml IL-2 and IL-7). 80 ⁇ l each of this cell suspension were pipetted into a well of a 96-well pointed-bottom cell culture plate (duplicates). The cell number / ml was set so that the desired effector-target ratio was achieved. Unlabeled K-562 cells (40 ⁇ l, 20-fold excess to target cells) were added to the effectors in order to completely block the activity of NK cells which may be present in the cell lines. The effectors were incubated with the K-562 cells for at least 30 minutes before the labeled target cells were added.
  • target cells were incubated with K-562 and medium (low-release control) and target cells with K-562, medium and Tween 20 (high-release control).
  • effectors were also incubated with unlabeled and labeled K-562.
  • the batches were incubated for 4 hours at 37 ° C., 98% atmospheric humidity and 5% CO 2 .
  • the culture plates were then centrifuged at 700 rpm for 5 min in order to sediment the cells. Without stirring up the cell pellet, 100 ⁇ l of the supernatant were pipetted from each well onto an Opti-Plate scintillation counter plate and left overnight at room temperature.
  • TIL Tumor Infiltrating Lymphocytes
  • TIL were obtained from the biopsies of tumors from patients with HPV 18 and HLA-A2 positivity by culture in AIM V medium (Gibco-Invitrogen, Düsseldorf), each with 100 U / ml IL-2 and IL-7 and 0J25 ⁇ l / well Dynal beads T cell expander (Dynal, Hamburg) brought out. This TIL were directly in the ELISpot assay for specificity for the HPV 18 E7 86 - examined 94 peptide epitope.
  • H7.1-A2 restricted peptides of the HPV 18 E7 protein are processed and presented by tumor cells
  • H7.1-A2 transfectants H7.1-A2 transfectants
  • the IFN- ⁇ ELISpot analysis showed a specific reaction against the predicted synthetic peptide FQQLFLNTL (see FIG. 17 above).
  • This peptide has a relatively low binding affinity for HLA-A2 compared to other predicted peptides. Nevertheless, specific T cells were detected.
  • HPV 16 E7 was 28 - 36 used, the induced no IFN- ⁇ secretion. This leads to the conclusion that this peptide is presented by H7.1-A2 cells.
  • T cell lines were produced against the synthetic peptide.
  • the T cells were tested in the chromium release assay.
  • Autologous peptide-loaded BLCL were used as target cells. With an effector to target ratio of 30: 1, a specific lysis of approximately 20% was observed (see FIG. 17 below). No specific lysis was measured against the ControUpeptides HPV 18 E7 7 - ⁇ 5 and HPV 16 E6 28 -36.
  • T cell lines were produced by stimulation with antigen-loaded autologous dendritic cells (DC).
  • the DC were loaded with recombinant HPV 18 E7 protein or a pool of overlapping 20-mer peptides, which represent the entire HPV 18 E7 (see FIG. 18 above: “E7 protein induced” or “E7 peptide pool inducted”).
  • the induced T cells were determined in the ELISpot assay for specificity for the HPV 18 E7 6 . 9 epitope tested.
  • HPV 18 E7 86 were found in both T cell lines. 94 specific T cells were detected (see Fig. 18 above). This means that the immunoproteasome processes the epitope.
  • TIL The biological relevance of HPV 18 E7 86 . 9 epitops were examined on TIL populations. TIL were isolated from a tumor biopsy of an HPV 18 and HLA-A2 positive patient and expanded linearly in vitro without antigen-specific Stimulation. They were confronted in the ELISpot assay with HPVl 8 E7 S 6. 9 loaded K-A2 (HLA-A2 transfected K-562 cells as stimulators).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Biotechnology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne un épitope lymphocytaire T de papillomavirus, qui possède la séquence d'acides aminés (I), et/ou une variante fonctionnellement active de cet épitope, ainsi que leur utilisation à des fins de diagnostic et de thérapie.
EP01998642A 2000-12-01 2001-11-30 Epitopes lymphocytaires t des proteines l1 et e7 de papillomavirus et leur utilisation a des fins de diagnostic et de therapie Withdrawn EP1370661A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
DE10059631A DE10059631A1 (de) 2000-12-01 2000-12-01 T-Zellepitope des Papillomavirus L1-und E7-Proteins und ihre Verwendung in Diagnostik und Therapie
DE10059631 2000-12-01
PCT/EP2001/014037 WO2002044384A2 (fr) 2000-12-01 2001-11-30 Epitopes lymphocytaires t des proteines l1 et e7 de papillomavirus et leur utilisation a des fins de diagnostic et de therapie

Publications (1)

Publication Number Publication Date
EP1370661A2 true EP1370661A2 (fr) 2003-12-17

Family

ID=7665350

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01998642A Withdrawn EP1370661A2 (fr) 2000-12-01 2001-11-30 Epitopes lymphocytaires t des proteines l1 et e7 de papillomavirus et leur utilisation a des fins de diagnostic et de therapie

Country Status (7)

Country Link
US (1) US20040091479A1 (fr)
EP (1) EP1370661A2 (fr)
JP (1) JP2004514449A (fr)
AU (1) AU2002220744A1 (fr)
CA (1) CA2430526A1 (fr)
DE (1) DE10059631A1 (fr)
WO (1) WO2002044384A2 (fr)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL105554A (en) * 1992-05-05 1999-08-17 Univ Leiden Peptides of human papillomavirus for use in preparations elicit a human T cell response
DE10137102A1 (de) 2001-07-30 2003-02-27 Deutsches Krebsforsch Polyvalente Vakzine gegen durch Papillomaviren verursachte Erkrankungen, Verfahren zu deren Herstellung und deren Verwendung
JP6035503B2 (ja) 2007-05-31 2016-11-30 アカデミシュ ジーケンハウス ライデン ハー.オー.デー.エン. ルムク ワクチンで使用するための、子宮頸部悪性腫瘍に浸潤するt細胞によって標的とされるhpvエピトープ
AU2015213420B2 (en) * 2007-05-31 2017-02-16 Isa Pharmaceuticals B.V. HPV epitopes targeted by T cells infiltrating cervical malignancies for use in vaccines
CA2703066A1 (fr) * 2007-10-22 2009-04-30 University Of Rochester Vaccin contre le virus syncytial respiratoire base sur des capsomeres ou des particules chimeriques semblables au virus du papillomavirus
US20120045413A1 (en) * 2010-04-09 2012-02-23 Mayumi Nakagawa Human papilloma virus peptide-specific T-cell response for treatment of warts
WO2013111448A1 (fr) * 2012-01-25 2013-08-01 学校法人 久留米大学 Peptide épitopique de lymphocyte b dérivé du papillomavirus humain de type 16l1
KR101680567B1 (ko) * 2014-07-09 2016-11-29 연세대학교 산학협력단 인유두종바이러스 펩타이드의 자궁경부암 진단 및 치료 용도
US10702598B2 (en) 2016-06-20 2020-07-07 Isa Pharmaceuticals B.V. Formulation of a peptide vaccine
CN114901799A (zh) * 2019-10-21 2022-08-12 弗拉斯沃克斯有限责任公司 用于细胞培养的系统和方法

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE8803870D0 (sv) * 1988-10-28 1988-10-28 Medscand Ab Method for detection of human papillomavirus (hpv) for diagnostic purposes
GB2279651A (en) * 1993-07-01 1995-01-11 British Tech Group Synthetic peptides of human papillomavirus
SE9501512D0 (sv) * 1995-04-24 1995-04-24 Euro Diagnostica Ab Synthetic peptide-defined eptopes useful for vaccination against papillomavirus
DE19925199A1 (de) * 1999-06-01 2000-12-07 Medigene Ag Zytotoxische T-Zellepitope des Papillomavirus L1-Proteins und ihre Verwendung in Diagnostik und Therapie

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0244384A2 *

Also Published As

Publication number Publication date
US20040091479A1 (en) 2004-05-13
JP2004514449A (ja) 2004-05-20
CA2430526A1 (fr) 2002-06-06
WO2002044384A2 (fr) 2002-06-06
WO2002044384A3 (fr) 2003-10-16
DE10059631A1 (de) 2002-07-18
AU2002220744A1 (en) 2002-06-11

Similar Documents

Publication Publication Date Title
DE69532532T2 (de) Chimerische papillomavirus-ahnliche partikel
DE19543553B4 (de) VP-Antigene des JC-Virus
EP0809700A1 (fr) Particules semblables au virus du papillome, proteines de fusion et leur procede de production
CN102497880A (zh) 新的人乳头状瘤病毒(hpv)蛋白构建体及其在预防hpv疾病中的用途
DE60126737T2 (de) Modifizierte HPV E6- und E7-Gene und -Proteine als Impfstoff
WO2000075180A2 (fr) Peptides servant a la vaccination contre le cytomegalovirus humain
DE69728289T2 (de) Immunreaktive ctl-peptidepitope von humanem cytomegalovirus
EP1370661A2 (fr) Epitopes lymphocytaires t des proteines l1 et e7 de papillomavirus et leur utilisation a des fins de diagnostic et de therapie
DE69531308T2 (de) Verfahren zur Herstellung von gereinigten papillomavirus Proteinen
DE69631586T9 (de) Für menschliches papillomavirus stamm 18 kodierende dns
DE69734387T2 (de) Ein kombiniertes antigen vom menschlichen cytomegalievirus und seine anwendung
EP1181312A1 (fr) Epitopes cytotoxiques de cellule t de la proteine l1 du papillomavirus et leur application en diagnostic et therapie
DE69933875T2 (de) Protein-verabreichungssystem, das dem menschlichen papillomavirus ähnliche partikel benützt.
WO2002043757A2 (fr) Medicament pour la prophylaxie ou le traitement de tumeurs induites par le virus du papillome humain de type 18
CN109715203A (zh) 用于治疗hsv的手段和方法
DE69635825T2 (de) Rekombinante virale pseudopartikel und deren verwendung als impfstoff oder als antitumorale wirkstoffe
EP1183367B1 (fr) Epitope cytotoxiques des lymphocytes t, de proteine l1 du papillomavirus, et utilisation dans les domaines du diagnostic et de la therapie
DE60035695T2 (de) Schutzantigen des epstein-barr-virus
JPH08512045A (ja) ヒトパピローマウイルスの合成ペプチド
EP1003877A2 (fr) Papillomavirus, agents permettant de les mettre en evidence et de traiter des affections induites par lesdits papillomavirus
WO2000073790A1 (fr) Systeme de test pour la detection in vitro d'une reaction immunitaire antigene specifique
DE19526752A1 (de) Hocheffiziente Bildung von Papillomavirusähnlichen Partikeln
DE10154831A1 (de) PNA-Konjugat oder PNA-Konjugat-Gemisch zur Therapie von mit HPV in Zusammenhang stehenden Erkrankungen
DE19943787A1 (de) Mittel zur Prävention und/oder Behandlung von Gewebeveränderungen mesenchymalen Ursprungs

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20030620

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20050601