WO2008035350A1 - Antigen specific multi epitope vaccines - Google Patents

Antigen specific multi epitope vaccines Download PDF

Info

Publication number
WO2008035350A1
WO2008035350A1 PCT/IL2007/001168 IL2007001168W WO2008035350A1 WO 2008035350 A1 WO2008035350 A1 WO 2008035350A1 IL 2007001168 W IL2007001168 W IL 2007001168W WO 2008035350 A1 WO2008035350 A1 WO 2008035350A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
cancer
cell
vaccine
epitopes
Prior art date
Application number
PCT/IL2007/001168
Other languages
French (fr)
Inventor
Lior Carmon
Original Assignee
Vaxil Biotherapeutics Ltd.
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 Vaxil Biotherapeutics Ltd. filed Critical Vaxil Biotherapeutics Ltd.
Priority to US12/442,495 priority Critical patent/US9487574B2/en
Priority to AU2007298494A priority patent/AU2007298494B2/en
Priority to CA2665816A priority patent/CA2665816C/en
Priority to EP07827143.4A priority patent/EP2089423B1/en
Publication of WO2008035350A1 publication Critical patent/WO2008035350A1/en
Priority to IL197737A priority patent/IL197737A/en
Priority to US15/345,036 priority patent/US11179452B2/en
Priority to US17/531,753 priority patent/US20220072113A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/00117Mucins, e.g. MUC-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4615Dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464469Tumor associated carbohydrates
    • A61K39/46447Mucins, e.g. MUC-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K14/4727Mucins, e.g. human intestinal mucin
    • 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
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • the present invention relates to cancer peptide vaccines with pan HLA class I and class II binding properties, as well as to pharmaceutical compositions containing the peptide vaccines and methods for treating or preventing cancer.
  • therapeutic vaccines such as anti-cancer vaccines and prophylactic (preventive or "conventional”) anti-infective vaccines.
  • therapeutic vaccines are generally expected to treat sick individuals, suggesting that a broader and stronger immune response is required.
  • prophylactic vaccines are generally induced against highly immunogenic "foreign" epitopes derived from viruses or bacteria and thus easily induce a strong response with a high number of T cell specific clones.
  • therapeutic vaccines in particular cancer vaccines composed of self derived TAAs are less immunogenic and therefore are frequently associated with low or minimal induction of activated T cell clones.
  • a desired therapeutic vaccine would need to prime a robust cellular reaction, which will involve multiple clones of T cell lymphocytes predominantly T killer (CD8 + ) and T Helper (CD4 + ).
  • Vaccines consisting of selected MHC class I-restricted sequences from a certain TAA/s. These vaccines consist of high purity sequence/s resembling immunogenic epitopes of a given TAA/s
  • Vaccines consisting of the entire sequence of a certain TAA/s.
  • the vaccine is composed of a mixture (at various levels of purity) of immunogenic and non-immunogenic MHC class I-restricted epitopes of a given TAA/s.
  • MHC class I-restricted TAA peptides are the targets of Cytotoxic T lymphocytes (CTL), which constitute one of the powerful effectors of the immune system against tumors (Townsend et al., 1989).
  • CTL Cytotoxic T lymphocytes
  • These peptide vaccines are usually 8 to 10 amino acids (AA) long, with 2 to 3 primary anchor residues that interact with the Major Histocompatibility complex (MHC) class I molecules and 2 to 3 AA residues that engage the T-cell receptor on CD8 + cells (Rammensee et al., 1993).
  • MHC Major Histocompatibility complex
  • CD8 + epitopes subsequent to the search for MHC-binding motifs in known putative TAAs, (Kast et al., 1994) as was shown in the case of the breast- carcinoma-associated HER-2/neu receptor (Fisk et al, 1995) or the colorectal tumor associated Carcino-Embryonic Antigen (CEA) (Ras et al, 1997).
  • CD4 + T cell activation is mediated via MHC class II-binding epitopes and is critical for the initiation (priming) and long lasting memory of most immune responses.
  • CD4 + T cells have a key role as effector cells with anti-tumor properties.
  • CD4 + T-cell responses are essential to promote the accumulation of Antigen-Presenting Cells (APC) for effective immune priming (Hung et ⁇ /.1998) and also for extending the life of antitumor CD8 + T cells i.e. memory response vs. short living response.
  • APC Antigen-Presenting Cells
  • MHC class I epitopes led in many cases to the administration of MHC class I epitopes with universal non-specific MHC class II- restricted epitopes such as the pan-class II epitope peptide PADRE (Weber et al., 1999). Although response against the universal MHC class Il-restricted epitopes was increased, elevation in CD8 + T-cell effectors specific to the MHC class I-restricted epitope have been limited (Weber et al., 1999).
  • CD4 + T cells Another important feature of CD4 + T cells is their role as effector cells with direct anti-tumor activity (Pardoll and Topalian 1998, Christopher et al., 2000).
  • MHC class II peptide ligands do not have restricted binding properties, their isolation is more complicated.
  • successful attempts in this direction were limited and arrived only more recently along with the development of sophisticated in-silico class II prediction software and class II transgenic mice (Chaux et al, 1999; Manici et al, 1999). Nevertheless, selected publications e.g.
  • Vaccines consisting of the entire TAA or (Non-defined) epitopes
  • the other strategy used to overcome the limited repertoire of anti-tumor CD8 + T cell clones is the use of the entire TAA rather then selecting and defining the only relevant immunodominant epitopes.
  • This strategy is more straightforward, as one does not need to isolate the immunogenic epitopes within a given TAA. However, it may very well lead to the "dilution" of the immunogenic epitopes with less immunogenic epitopes, hence decreasing the level of specific immunity or reduce the repertoire of anti-tumor CD8 + T cell clones. Furthermore, some of these less immunogenic epitopes could potentially induce a status of "Immune Anergy" (non responsiveness) which can potentially lead to a decrease in the intensity of the specific immunity or even to status of autoimmunity.
  • the peptide-specific T cells were able to lyse tumors. More importantly, the responses lasted for a long time and were detectable for more than a year after the final vaccination in select patients. This study suggested an improved anti-cancer immunity via combination of class-I and class-II epitopes derived from the same TAA.
  • cancer therapeutic vaccines are required to be: o Antigen specific to avoid potential Anergy and autoimmunity. o Highly potent in inducing a strong, comprehensive and long lasting response involving CD4 + plus CD8 + T cells. o Applicable in the majority of the target population.
  • SP signal peptides
  • Signal peptides generally consist of three parts: an N-terminal region of differing length, which usually comprises positively charged amino acids; a hydrophobic domain; and a short carboxy-terminal peptide segment.
  • pre-protein a nascent precursor protein
  • ER Endoplasmic Reticulum
  • the signal peptide directs the pre- protein to the cytoplasmic membrane.
  • the signal peptide is not responsible for the final destination of the mature protein; secretory proteins devoid of further address tags in their sequence are by default secreted to the external environment.
  • Signal peptides are cleaved from precursor proteins by an endoplasmic reticulum (ER)-resident signal peptidase or they remain uncleaved and function as a membrane anchor.
  • ER endoplasmic reticulum
  • Signal peptides are cleaved from precursor proteins by an endoplasmic reticulum (ER)-resident signal peptidase or they remain uncleaved and function as a membrane anchor.
  • ER endoplasmic reticulum
  • the signal peptide enhances the ability of the epitope to attract and activate CD8+ T cells (Minev BR, et al, 2000).
  • TAA Transporter for Antigen Presentation
  • the signal peptide merely acts as a chaperon or tag for selected epitopes/Vaccines.
  • These vaccines suffer from several drawbacks as they utilize selected immunogenic epitopes, MHC-class I-restricted peptides with limited repertoire for activation, or non-defined immunogenic epitopes with nonspecific activity.
  • MUCl is one of the most promising TAAs known today.
  • This polymorphic epithelial mucin, encoded by the MUCl gene is a high-molecular- weight glycoprotein with few alternative-splicing variants encoding for both a transmembranal (i.e. across the cell membrane) and a secreted (i.e. circulating) product both expressed in a broad range of tumors (Graham et al., 1996; Ho et al., 1993).
  • MUCl is one of the few known targets that are expressed by more than 90 per cent of common solid tumor cancers including Colon, Gastric, Lung, Renal Cell (RC), Transitional Cell (TC), Prostate, Pancreas, Breast, Ovary and Thyroid.
  • MUCl Lymphoma, Leukemia, and Multiple Myeloma
  • MM Multiple Myeloma
  • MUCl molecule A major feature of the MUCl molecule (in both the transmembranal and the secreted variants) is the presence of a highly immunogenic extracellular tandem repeat array (TRA) heavily O-glycosylated at serine and threonine residues. It was shown that this extracellular TRA domain can be recognized by monoclonal antibodies (MAbs), as well as MHC-restricted CD8 + T cells (CTLs) such as HLA-AI l and HLA-A2.1- CTLs. In spite of the high immunogenicity of the TRA, its role as a potential target/vaccine is ambiguous.
  • MAbs monoclonal antibodies
  • CTLs MHC-restricted CD8 + T cells
  • BAGE Another example of a tumor associated antigen is BAGE.
  • BAGE codes for a putative protein of 43 amino acids and seems to belong to a family of several genes.
  • Gene BAGE is expressed in 22% of melanomas, 30% of infiltrating bladder carcinomas, 10% of mammary carcinomas, 8% of head and neck squamous cell carcinomas, and 6% of non-small cell lung carcinomas. It is silent in normal tissues with the exception of testis (Boel et al., 1995).
  • ARMET Arginine rich, mutated in early stage of tumors
  • ARP Arginine-rich protein
  • ARP Arginine-rich protein
  • the present invention relates to promiscuous peptide vaccines comprising multiple MHC class I, and MHC class II epitopes of a given protein antigen. More particularly, the present invention relates to promiscuous peptide vaccines comprising multiple MHC class I and MHC class II epitopes with the specificity of a given antigen derived from the entire signal peptide domain of that protein antigen. These MHC class I and MHC class II epitopes have a high frequency in the population and thus the vaccine is effective in a large portion of the population.
  • the present invention thus provides a peptide vaccine which is able to induce strong, comprehensive response in the majority of the target population against said antigen. More specifically, but without wishing to be limited to a single hypothesis, such a vaccine preferably combines activation of both CD4 + and CD8 + T cells via multiple CD4 + and CD8 + -restricted epitopes which are present within the internal sequences of the vaccine and are derived from the same antigen.
  • the present invention relates to such peptide vaccines comprising the signal peptide domain of tumor associated antigens (TAA) or the signal peptide domain of proteins which are over-expressed in tumor cells.
  • TAA tumor associated antigens
  • the present invention relates to peptide vaccines comprising the signal peptide of a protein which is either a TAA or is over-expressed in tumor cells, wherein said peptides are recognized and presented by more than 50% of the MHC class I and MHC Class II alleles in the population.
  • said peptide is not longer than 50 amino acids, more preferably, not longer than 25 amino acids.
  • the peptide vaccines of the invention comprise the signal peptide of proteins selected from the group consisting of Armet, HSP60, CANX, MTHFD2, FAP, MMP6, BAGE-I, GNTV, Q5H943, MUCl, CEA, Pmel, Kallikrein- 4, Mammaglobin-1, MART-I, GPR143-OA1, PSA, TRPl, Tyrosinase, FGF-5, NEU proto-oncogene, Aft, MMP-2, PSMA, Telomerase-associated protein 2, PAP, Uroplakin II and Proteinase 3, i.e. SEQ ID Nos. 1-28, respectively (Table 1).
  • the present invention relates to tumor associated antigen peptides comprising the signal peptide domain of the polymorphic epithelial mucin, encoded by the MUCl gene.
  • the present invention thus provides a promiscuous peptide vaccine comprising the MUCl signal peptide domain which is able to induce strong, comprehensive response in the majority of the target population against any MUCl positive tumor.
  • the MUCl signal pepti de-derived peptide vaccines are able to bind to the majority of MHC Class I alleles in the population and thus induce CD8+ T-cell mediated cell lysis, and are also able to bind to bind to the majority of MHC Class II alleles in the population and thus prime an effective CD4+ T-cell mediated immune response.
  • the MUCl signal peptide-derived peptide vaccine comprises the amino acid sequence MTPGTQSPFFLLLLLTVLTW (SEQ ID NO. 10).
  • the peptide vaccine of the invention comprises a mixture of at least two short peptides of preferably about nine amino acid residues in length derived from the signal peptide domain of the MUCl protein.
  • These peptides represent various MHC Class I and Class II epitopes which are included in the MUCl signal peptide. Their combination results in effective binding of the vaccine composition to various alleles of MHC class I and MHC class II molecules, and thus to the induction of an immune response to tumors expressing the MUCl protein.
  • This response may include inducing "help" for priming a strong T cell activity via CD4+ T cell activation, combined with induction of CD 8+ T cell activation, and potent cellular activity (CTL) against MUCl expressing tumors.
  • CTL potent cellular activity
  • the mixture of short peptides comprises at least two peptides selected from the group consisting of SEQ ID NO: 29-39.
  • the mixture of short peptides comprises VXLOl (SEQ ID NO 29), VXL02 (SEQ ID NO 30), VXL04 (SEQ ID NO 31) and VXL05 (SEQ ID NO 32).
  • the present invention relates to peptide vaccines derived from the signal peptide domain of the BAGE-I gene.
  • the BAGE-I signal peptide-derived peptide vaccine comprises the amino acid sequence MAARA VFLAL SAQLLQA (SEQ ID NO. 7).
  • the present invention relates to peptide vaccines derived from the signal peptide domain of the Armet gene.
  • the Armet signal peptide-derived peptide vaccine comprises the amino acid sequence MWATQGLAVA LALS VLPGSR A (SEQ ID NO. 1).
  • the present invention also concerns use of the peptide vaccines described above in the preparation of pharmaceutical compositions for treating or inhibiting cancer.
  • the invention further concerns pharmaceutical compositions comprising said peptide vaccines and the use of said peptide vaccines or said pharmaceutical compositions as anti-tumor vaccines to treat or inhibit the development of cancer.
  • pharmaceutical compositions comprising said peptide vaccines and the use of said peptide vaccines or said pharmaceutical compositions as anti-tumor vaccines to treat or inhibit the development of cancer.
  • tumors which over-expresses the protein from which the signal peptide vaccine was derived, for example, MUCl -expressing cancer, BAGE-I -expressing cancer, or Armet-expressing cancer.
  • the invention further concerns nucleic acid molecules encoding said peptides, and antigen presenting cells (APC), e.g. dendritic cells, presenting said peptides, as well as pharmaceutical compositions comprising said nucleic acid molecules, or said cells.
  • APC antigen presenting cells
  • the invention also concerns use of the peptide vaccines for enrichment of T cell populations in vitro. Thus obtaining a peptide-specific enriched T cell population.
  • the invention further concerns the use of said nucleic acid molecules, cells, or pharmaceutical compositions comprising same as anti-tumor vaccines to treat or inhibit the development of cancer.
  • tumors which over-expresses the protein from which the signal peptide vaccine was derived, for example, MUCl -expressing cancer, BAGE-I -expressing cancer, or Armet- expressing cancer.
  • compositions of the present invention are directed to a method for treating or for inhibiting the development of cancer by administering the pharmaceutical compositions of the present invention to a patient in need thereof.
  • the pharmaceutical compositions of the invention may be adapted for use in combination with other anti neoplastic agents.
  • Figure 1 is a graph showing results of an ELISA quantitative assay measuring cytokine secretion profiles of a specific T cell subpopulation developed via repeated stimulation with ImMucin. The results represent one out of two experiments using four different donors.
  • Figure 2 is a graph showing FACS analysis of T cell phenotype evaluation during consistent stimulation with ImMucin. The Results represent one out of two experiments using four different donors.
  • the present invention provides antigen specific vaccines which are capable of inducing a robust T-cell immunity and which are applicable to the majority of the population.
  • SP signal peptides
  • the present invention is based on the surprising finding that SP-derived vaccines are able to bind simultaneously to multiple alleles of both MHC class I and MHC class II i.e. CDA + and CD8 + -restricted epitopes.
  • a signal peptide vaccine although containing just one sequence, could thus be compared to a large number of single Class I and Class II epitopes, used in a mixture.
  • This newly discovered feature of SP-vaccines facilitates the generation of a robust immune response in the majority of the target population.
  • signal peptide-based vaccines bare the ability to independently penetrate the ER and thus, at least partially, avoid immune escape mechanisms such as TAP deficiency. ⁇
  • the objective of the predictive algorithm is to identify and obtain signal peptide (SP) targets with a potential role as cancer vaccines.
  • SP signal peptide
  • Tumor antigens that are tissue-specific (differentiation) antigens httpV/www.cancerimmunitv-org/peptidedatabase/differentiation.htm
  • Tumor antigens that are overexpressed in cancer cells vs. the respective normal tissue (http://www.cancerimmunitv.org/peptidedatabase/overexpressed.htm)
  • Proteins with the following attributes are removed from the list of putative targets as being non eligible for an immune assault:
  • ⁇ Proteins that are located sub-cellularly e.g. in organelles or in any other location that does not require transport from the ER-Golgi, and thus have no signal peptides (e.g. purely cytoplasmic proteins, such as ATP-citrate synthase).
  • ⁇ Proteins which function in basic or homeostatic functions in all cells e.g. purine synthesis, for example phosphoribosylaminoimidazole carboxylase, phosphoribosylaminoimidazole succinocarboxamide synthetase (PAICS), succinate dehydrogenase, cytochrome b556 subunit (SDHC), cell division cycle 2 (CDC2).
  • ⁇ Proteins that are ubiquitously expressed in many tissues e.g. tubulin, beta TUBB, RNA binding motif protein 4 (RBM4).
  • Immune-related proteins e.g. proteasome (prosome, macropain) activator subunit 2 PSME2, CD213a2, Macrophage colony-stimulating factor (M-CSF).
  • Proteins that are found to be eligible targets for an immune assault are next examined for the presence of a signal peptide. This may be done by using appropriate computer software, e.g. the Signal P 3.0.
  • the Signal P 3.0 program uses both a neural network (NN) algorithm and a Hidden Markov models (HMM) algorithm for selection of the signal (http://www.cbs.dtu. dk/services/SignalPA).
  • N neural network
  • HMM Hidden Markov models
  • cancer protein targets eligible for an immune assault and having an identified signal peptide sequence of 17-50 amino acids are selected for further examination of predicted binding to MHC alleles.
  • HLA-A, B, C HLA class I
  • HLA-DRBl HLA class II
  • the binding strength of the previously identified signal peptides to the HLA alleles is predicted using any of numerous available software programs.
  • the following is a non-limiting list of available prediction programs:
  • ⁇ BIMAS http://www-bimas.cit.nih.gov/molbio/hla bind/) may be used for the prediction of HLA class I alleles.
  • ⁇ Propred http://bic.uams.edu/mirror/propred/ may be used to predict most DRBl genotypes.
  • ⁇ MHC2Pred http://www.imtech.res.in/raghava/mhc2pred/index.html
  • ⁇ MHC2Pred http://www.imtech.res.in/raghava/mhc2pred/index.html
  • the probability that the patient has one or more of the binding alleles is 1 minus the probability that he would have none of the binding alleles:
  • the most suitable SP vaccine candidates are chosen according to the following criteria:
  • Length of SP Peptide of up to 50 AA, preferably up to 30 AA, most preferably up to 25 AA.
  • TAA tumor associate antigen
  • a MUCl SP vaccine (hereinafter termed "VXLlOO” or “ImMucin”) was prepared.
  • the ImMucin vaccine of the invention is composed of a 21 amino acid (AA)-long peptide derived from the signal peptide domain of the MUCl protein and comprises the amino acid sequence MTPGTQSPFFLLLLLTVLTVV (SEQ ID NO 10).
  • This peptide vaccine is processed in the antigen-presenting cell (APC) and presented to immune effector cells by MHC class I, and II molecules.
  • APC antigen-presenting cell
  • ImMucin harbors a set of unique characteristics as listed below: o ImMucin targets the mucin MUCl expressed on the surface of Multiple
  • ImMucin does not contain any non-specific epitopes that could dilute and disturb specific anti-cancer immunity. o ImMucin was selected due to its ability to bind multiple MHC Class I and Class II alleles. This would potentially offer:
  • ImMucin has superior immunogenicity which may, at least partially, circumvent immune escape mechanisms such as TAP-deficiency of cancer cells.
  • the present invention concerns a vaccine comprising a mixture of short peptides comprising MHC Class I and II epitopes within the MUCl signal domain.
  • These short peptides include:
  • SEQ ID NO 29 LLLTVLTVV (designated VXLOl) SEQ ID NO 30: LLLLTVLTV (designated VXL02) SEQ ID NO 31 : TQSPFFLLL (designated VXL04) SEQ ID NO 32: SPFFLLLLL (designated VXL05) SEQ ID NO 33: FLLLLLTVL SEQ ID NO 34: LLLLLTVLT SEQ ID NO 35: GTQSPFFLL SEQ ID NO 36: TPGTQSPFF SEQ ID NO 37: FFLLLLLTV SEQ ID NO 38: MTPGTQSPF SEQ ID NO 39: QSPFFLLLLLL
  • tumor associated antigen or "TAA” refers to antigens or proteins that are highly correlated with certain tumor cells. They are not usually expressed in normal cells, or are expressed at a higher extent in tumor cells than in normal cells.
  • peptide refers to a molecular chain of amino acids, which, if required, can be modified in vivo or in viti'o, for example by manosylation, glycosylation, amidation (specifically C-terminal amides), carboxylation or phosphorylation with the stipulation that these modifications must preserve the biological activity of the original molecule.
  • peptides can be part of a chimeric protein.
  • Functional derivatives of the peptides are also included in the present invention.
  • Functional derivatives are meant to include peptides which differ in one or more amino acids in the overall sequence, which have deletions, substitutions, inversions or additions.
  • Amino acid substitutions which can be expected not to essentially alter biological and immunological activities have been described.
  • Amino acid replacements between related amino acids or replacements which have occurred frequently in evolution are, inter alia Ser/Ala, Ser/Gly, Asp/Gly, Asp/Asn, Ile/Val see Dayhof M.D (1978). Based on this information, Lipman and Pearson (1985) developed a method for rapid and sensitive protein comparison and determining the functional similarity between homologous polypeptides.
  • the peptides according to the invention can be produced synthetically, by recombinant DNA technology. Methods for producing synthetic peptides are well known in the art. - 1 * 2 * -
  • the organic chemical methods for peptide synthesis are considered to include the coupling of the required amino acids by means of a condensation reaction, either in homogenous phase or with the aid of a so-called solid phase.
  • the condensation reaction can be carried out as follows:
  • Activation of the carboxyl group can take place, inter alia, by converting the carboxyl group to an acid halide, azide, anhydride, imidazolide or an activated ester, such as the N-hydroxy-succinimide, N-hydroxy-benzotriazole or p-nitrophenyl ester.
  • polypeptide to be expressed is coded for by a nucleic acid sequence.
  • nucleic acid sequence comprising the sequence encoding the peptides according to the present invention.
  • the degeneracy of the genetic code permits substitution of bases in a codon to result in another codon still coding for the same amino acid, e.g., the codon for the amino acid glutamic acid is both GAT and GAA. Consequently, it is clear that for the expression of a polypeptide with an amino acid sequence as shown in any of SEQ ID NO: 1-28 use can be made of a derivate nucleic acid sequence with such an alternative codon composition thereby different nucleic acid sequences can be used.
  • Nucleotide sequence refers to a polymeric form of nucleotides of any length, both to ribonucleic acid (RNA) sequences and to deoxyribonucleic acid (DNA) sequences. In principle, this term refers to the primary structure of the molecule. Thus, this term includes double and single stranded DNA, as well as double and single stranded RNA, and modifications thereof.
  • nucleotide sequences encoding the peptide vaccines of the invention can be used for the production of the peptides using recombinant DNA techniques.
  • the nucleotide sequence must be comprised in a cloning vehicle which can be used to transform or transfect a suitable host cell.
  • useful cloning vehicles may include chromosomal, non-chromosomal and synthetic DNA sequences such as various known bacterial plasmids, and wider host range plasmids such as pBR 322, the various pUC, pGEM and pBluescript plasmids, bacteriophages, e.g. lambda-gt- Wes, Charon 28 and the Ml 3 derived phages and vectors derived from combinations of plasmids and phage or virus DNA, such as SV40, adenovirus or polyoma virus DNA.
  • chromosomal, non-chromosomal and synthetic DNA sequences such as various known bacterial plasmids, and wider host range plasmids such as pBR 322, the various pUC, pGEM and pBluescript plasmids, bacteriophages, e.g. lambda-gt- Wes, Charon 28 and the Ml 3 derived phages and vector
  • Useful hosts may include bacterial hosts, yeasts and other fungi, plant or animal hosts, such as Chinese Hamster Ovary (CHO) cells, melanoma cells, dendritic cells, monkey cells and other hosts.
  • bacterial hosts such as Chinese Hamster Ovary (CHO) cells, melanoma cells, dendritic cells, monkey cells and other hosts.
  • yeasts and other fungi such as Chinese Hamster Ovary (CHO) cells, melanoma cells, dendritic cells, monkey cells and other hosts.
  • CHO Chinese Hamster Ovary
  • Vehicles for use in expression of the peptides may further comprise control sequences operably linked to the nucleic acid sequence coding for the peptide.
  • control sequences generally comprise a promoter sequence and sequences which regulate and/or enhance expression levels.
  • an origin of replication and/or a dominant selection marker are often present in such vehicles.
  • control and other sequences can vary depending on the host cell selected.
  • the present invention also provides a polynucleotide encoding the signal peptide vaccine of the invention as part of a pharmaceutical composition preferably for targeted treatment of a tumor.
  • Further aspects of the present invention are directed to a method for treating or for inhibiting the development of cancer by administering the pharmaceutical compositions of the present invention to a patient in need thereof.
  • the present invention describes a method for treating or inhibiting the development of solid tumors for example, Colon, Gastric, Lung, Renal Cell (RC), Transitional Cell (TC), Prostate, Pancreas, Breast, Ovary or Thyroid cancers, as well as non-solid tumors such as Lymphoma, Leukemia, and Multiple Myeloma.
  • solid tumors for example, Colon, Gastric, Lung, Renal Cell (RC), Transitional Cell (TC), Prostate, Pancreas, Breast, Ovary or Thyroid cancers, as well as non-solid tumors such as Lymphoma, Leukemia, and Multiple Myeloma.
  • the present invention provides a method for treating or for inhibiting the development of MUCl -expressing cancers by administering the MUCl signal peptide-derived peptide vaccine of the present invention to a patient in need thereof.
  • the present invention provides a method for treating or for inhibiting the development of BAGEl -expressing cancers by administering the BAGEl signal peptide-derived peptide vaccine of the present invention to a patient in need thereof.
  • cancers include melanoma, bladder carcinoma, mammary carcinoma, head and neck squamous cell carcinoma, and non-small cell lung carcinomas.
  • the present provides a method for treating or for inhibiting the development of Armet-expressing cancers by administering the Armet signal peptide-derived peptide vaccine of the present invention to a patient in need thereof.
  • Such cancers include renal cell carcinomas, lung, breast, prostate, squamous cell carcinoma, head and neck carcinoma, pancreatic carcinoma.
  • the peptide vaccine of the invention is administered in an immunogenically effective amount with or without a co-stimulatory molecule.
  • the peptide vaccine may be administrated to a subject in need of such treatment for a time and under condition sufficient to prevent, and/or ameliorate the condition of cancer being treated.
  • the antigen and co-stimulatory molecule are formulated, separately or as a "chimeric vaccine" formulation, with a pharmaceutically acceptable carrier and administered in an amount sufficient to elicit a T lymphocyte-mediated immune response.
  • the peptide may be administered to subjects by a variety of administration modes, including by intradermal, intramuscular, subcutaneous, intravenous, intra-atrial, intra-articular, intraperitoneal, parenteral, oral, rectal, intranasal, intrapulmonary, and transdermal delivery, or topically to the eyes, ears, skin or mucous membranes.
  • the antigen may be administered ex-vivo by direct exposure to cells, tissues or organs originating from a subject (Autologus) or other subject (Allogeneic), optionally in a biologically suitable, liquid or solid carrier.
  • the peptides or pharmaceutical composition with or without a co-stimulatory molecule are delivered to a common or adjacent target site in the subject, for example to a specific target tissue or cell population in which the vaccine formulation is intended to elicit an immune response.
  • the peptide or pharmaceutical composition and the optional co- stimulatory molecule are administered separately, they are delivered to the same or closely proximate site(s), for example to a single target tissue or to adjacent sites that are structurally or fluidly connected with one another (e.g., to allow direct exposure of the same cells, e.g., fluid flow transfer, dissipation or diffusion through a fluid or extracellular matrix of both vaccine agents).
  • a shared target site for delivery of antigen and co-stimulatory molecule can be a common surface (e.g., a mucosal, basal or lunenal surface) of a particular target tissue or cell population, or an extracellular space, lumen, cavity, or structure that borders, surrounds or infiltrates the target tissue or cell population.
  • a common surface e.g., a mucosal, basal or lunenal surface
  • the peptide antigen with or without a co- stimulatory molecule may be administered to the subject separately or together, in a single bolus delivery, via continuous delivery (e.g., continuous intravenous or transdermal delivery) over an extended time period, or in a repeated administration protocol (e.g., on an hourly, daily or weekly basis).
  • continuous delivery e.g., continuous intravenous or transdermal delivery
  • a repeated administration protocol e.g., on an hourly, daily or weekly basis.
  • the various dosages and delivery protocols contemplated for administration of peptide and co-stimulatory molecule, in simultaneous or sequential combination are immunogenically effective to inhibit the occurrence or alleviate one or more symptoms of the target cancer in the subject.
  • an “immunogenically effective amount” of the antigen thus refers to an amount that is, in combination, effective, at dosages and for periods of time necessary, to elicit a specific T lymphocyte mediated immune response.
  • This response can be determined by conventional assays for T-cell activation, including but not limited to assays to detect proliferation, specific cytokine activation and/or cytolytic activity.
  • the amount of peptide vaccine is immunogenically effective to achieve a desired cancer inhibitory effect in the subject.
  • an immunogenically effective amount of the peptide depending on the selected mode, frequency and duration of administration, will effectively prevent cancer, or will inhibit progression of a cancerous condition in the subject.
  • an immunogenically effective dosage of the antigen which may include repeated doses within an ongoing prophylaxis or treatment regimen, will alleviate one or more symptoms or detectable conditions associated with a cancerous disorder. This includes any detectable symptom or condition amenable to prophylaxis and/or treatment with the vaccines of the invention, for example symptoms or conditions associated with breast cancer, cervical cancer, prostate cancer, colon cancer, melanoma and other cancerous conditions.
  • peptide antigens might be formulated with a “pharmaceutical acceptable carrier".
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption enhancing or delaying agents, and other excipients or additives that are physiologically compatible.
  • the carrier is suitable for intranasal, intravenous, intramuscular, intradermal, subcutaneous, parenteral, oral, transmucosal or _ _
  • the active compound may be coated in a material to protect the compound from the action of acids and other natural conditions which may inactivate the compound.
  • Peptide vaccine may be administered to the subject in the form of a peptide solution per se or a combination of a peptide with an appropriate auxiliary agent using an injector.
  • the peptide vaccine may be percutaneously administered through mucous membrane by, for instance, spraying the solution.
  • the unit dose of the peptide typically ranges from about 0.01 mg to 100 mg, more typically between about 100 micrograms to about 5 mg, which may be administered, one time or repeatedly, to a patient.
  • auxiliary agents which can be formulated with or conjugated to peptide or protein antigens and/or vectors for expressing co-stimulatory molecules to enhance their immunogenicity for use within the invention include cytokines (e.g. GM-CSF), bacterial cell components such as BCG bacterial cell components, imnunostimulating complex (ISCOM), extracted from the tree bark called QuillA (Morein et al., 1984 incorporated herein by reference), QS-21, a saponin-type auxiliary agent (Wu et al., (1992), incorporated herein by reference), Montanide ISA 51VG, liposomes, aluminum hydroxide (alum), bovine serum albumin (BSA), tetanus toxoid (TT) (Green et al., (1982) incorporated herein by reference) and keyhole limpet hemocyanin (KLH).
  • cytokines e.g. GM-CSF
  • bacterial cell components such as BCG bacterial cell components
  • compositions of the present invention it may be desirable to modify the peptide antigen, or to combine or conjugate the peptide with other agents, to alter pharmacokinetics and biodistribution.
  • a number of methods for altering pharmacokinetics and biodistribution are known to persons of ordinary skill in the art. Examples of such methods include protection of the proteins, protein complexes and polynucleotides in vesicles composed of other proteins, lipids (for example, liposomes), carbohydrates, or synthetic polymers.
  • the vaccine agents of the invention can be incorporated into liposomes in order to enhance pharmacokinetics and biodistribution characteristics.
  • liposome delivery vehicles peptides are typically entrapped within the liposome, or lipid vesicle, or are bound to the outside of the vesicle.
  • peptide antigens are associated with liposomes, such as lecithin liposomes or other liposomes known in the art, as a DNA-liposome mixture, or the DNA may be associated with an adjuvant known in the art to boost immune responses, such as a protein or other carrier.
  • Additional agents which assist in the cellular uptake of DNA such as, but not limited to, calcium ions, viral proteins and other transfection facilitating agents and methods may also be used to advantage (Wigler et al., Cell 14:725, 1978; Corsaro and Pearson, Somatic Cell Genetics 7:603, 1981; Graham and Van der Eb, Virology 52:456, 1973; Neumann et al., EMBO J. 1:841-845, 1982; and Hawley-Nelson et al., Focus 15:73- 79, 1993, each incorporated herein by reference).
  • Human PBMC were separated from buffy coat samples of naive donors using Ficoll UNI-SEPmaxi tube. Separated cells were suspended in RPMI medium supplemented with 10% FCS, L-Glutamine, Sodium Pyruvate, MEM-EAGLE non-essential amino- acids, HEPES and Gentamycin Sulphate (Bet-Haemek Industries, IL) and cultivated for 4h at 37 0 C in a culture dish (Corning 150mmx25mm).
  • Adherent cells were collected and cultured for 7 days in serum free DCCM-I medium supplemented with L-Glutamine, huIL-4 (1000IU/ml) (Cytolab IL) and GM-CSF-80 ug/ml (Cytolab IL). On day seven floating cells were collected and loaded with 50ug/ml of examined peptide for 18h at 37 0 C.
  • T cell medium contained RPMI medium supplemented with 10% FCS, L-Glutamine, Sodium Pyruvate, MEM-EAGLE non-essential amino-acids, HEPES and Gentamycin _ _
  • Cytokine release was studied during the culture of stimulated T-cells. The evaluated samples were collected from the T cell culture medium on day 2, 3, and 5 and stored until use at-20°C.
  • the selected capture antibody e.g. anti -Hu IFN-Gamma, or anti-Hu TNF- alfa or anti IL-2
  • 100 ml of the selected capture antibody were added to each well at the final concentration of 5 ⁇ g/ml in PBS. Plates were incubated for 2h at R.T. or over night at 4°C. Next, 200 ml of blocking solution were added for an incubation of 2h at R.T. lOO ⁇ g of evaluated samples were added in duplicate into 2 wells for 2 hours at RT. After 2h of incubation at R.T plates were washed X4 times in washing solution.
  • the selected capture antibody e.g. anti -Hu IFN-Gamma, or anti-Hu TNF- alfa or anti IL-2
  • a Biotin-conjugated detection antibody (relevant to coated antibody) was added at a final concentration of lO ⁇ g/ml in total volume of lOO ⁇ l for Ih at R.T.
  • lOO ⁇ l/well working dilution (1: 10000 in Blocker solution) of Streptavidin- HRP were added to each well and incubated for Ih at R.T.
  • PBL direct presentation In the 2nd method, lOO ⁇ l of 2x10 6 PBL were placed in Flat Bottom 96 well plates (Corning) and stimulated with different concentrations (0.05ug/ml-2ug/ml) of peptides. In the next step the cells underwent proliferation analysis as described above.
  • PBMC direct presentation In third method PNMC were used under similar conditions as described for the second method.
  • CTL In Vitro cytotoxic assay
  • Effector cells were ImMucin specific enriched T-CeIl clone.
  • Viable lymphocytes effector cells
  • CTL assays were performed in U-shaped microtiter wells, at 37 C, 5% CO2 for 5 hours. Cultures were terminated by centrifugation at 1000 rpm for 10 min at 4O 0 C. A total of 100 ⁇ l of the supernatants was mixed with scintillation fluid and measured in a ⁇ counter (Becton Dickinson Canberra Australia).
  • % lysis (cpm in experimental well -cpm spontaneous release)/(cpm maximal release - cpm spontaneous release) x 100.
  • Spontaneous release was determined by incubation of 100 ⁇ l-labeled target cells with 100 ⁇ l of medium. Maximal release was determined by lysis of target cells in lOO ⁇ l 10% TrytonX-100.
  • FACS analysis PBLs were suspended at a final concentration of 20x10 and 50 ⁇ l were transfer into FACS tube (Falcon) in Blocker solution (PBS with 3% FCS and 0.1% Sodium Azide). 10 ⁇ l of fluorochrome-conjugate anti-CD4, anti-CD8, and anti- CD45RO (eBioscience) were added for 30min on ice at O 0 C in the Dark. After the - -
  • Table 3 describes ImMucin (VLX-100) and other various VXL epitopes used in the experiments.
  • the CTL epitopes VXLl (D6) and VXL2 (Ml.2) were used as positive controls for MUC l's SP and class I epitopes.
  • MUC l's class I epitopes which is not derived from the SP domain, we have used the previously identified CTL epitopes VXL6 (M 1.1).
  • M 1.1 CTL epitopes derived from the SP domains of other non MUCl TAA like Her2/neu (VXL-8) or Tyrosinase (VXL- 11).
  • As a positive control for class II epitope we used the universal pan-class II epitope peptide PADRE (VXL- 14).
  • the most immunogenic antigen is the 21mer ImMucin which manifested a SI of 4 and 2 respectively for peptide presentation via DC and PBMC (see table 4).
  • SI of > 2 is considered to be a strong specific activation.
  • the high SI of ImMucin suggests polyclonal T cell activation via binding to multi MHC epitopes. In other words, different epitopes are used in different donors.
  • the index of stimulation for ImMucin's 9mer epitopes was analyzed. Like ImMucin, the class I and/or class II 9mer epitopes VXL-4, VXL-I and VXL-5 (see Table 4 and 3) manifested high stimulation index of SI>3.
  • Table 4 Proliferation analysis of PBL using ImMucin and other VXL target epitopes indicated by Index of stimulation (SI). SI is calculated by dividing the CPM obtained in an analyzed sample to the CPM obtained in a control sample. Results are representative of 5 similar experiments. Analyzing the time for maximal peak of PBL activation
  • a different parameter for assessing the properties of ImMucin and the other VXL- peptide epitopes is by analyzing the time until a maximal peak of activity occurs and the optimal dose for maximal stimulation. Maximal peak of PBL activation is determined when ⁇ 50% of the PBLs appear in clumps. In these experiments, the kinetic (time) in the proliferation of lymphocytes from the six naive donors stimulated at a fix dose of 0.05- lug/ml by ImMucin and other VXL-Peptides, was observed.
  • Results in the five experiments show a more rapid peak of proliferation to VXL-4 and VXL-5 with a peak at 48-72 hours (Table 5) and lowest concentration needed for activation 0.2-0.05ug/ml (Table 6) while ImMucin and other SP-derived epitopes or non SP epitope gave slower peak of proliferation at the range of 96 hours and a slightly higher dose for stimulation 0.05-lug/ml (Table 6).
  • SP-associated sequences/epitopes were noted, e.g. the MUCl VXLl, 2, and 4 epitopes, but also other SP epitopes such as VXL 8 and VXLI l, which contain antigen specific properties for CD4 + and/or CD8 + activation and other sequences (such as VXL-4 and VXL-5) which in addition to CD4 + and/or CD8 + activation also have an "adjuvant like" activity.
  • the adjuvant-like property of signal peptides was already shown in the past by attaching SP to other non-SP epitopes in order to increase their immunity (Sherritt et al., 2001).
  • TNF- Alfa is a proinflamatory cytokine which is secreted at early stage of activation by both CD4 + and CD8 + .
  • Interleukin-2 is one of the key cytokines which enhance the proliferation of CD4 + T lymphocytes. It induces a secondary immune response of CD8 + cells and the development of memory CD8 + cells following primary activation with antigen. Therefore, IL-2 is usually associated with late secretion.
  • IFN-gamma is produced mainly by CD8 + T cells following IL-2 secretion (late secretion) and is correlates with CD8 + specific activation and function.
  • Table 7 ELISA quantitative assay for Cytokine secretion profiles of PBL stimulated once with ImMucin or other VXL 9mer epitopes DC at the ratio of 16:1. The results represent one out of five experiments using six different donors. SI of the peptides is also indicated.
  • IFN-Gamma by CD8 T cells The results showing IFN-Gamma and IL- 2 secretion emphasize the "super activation" properties of ImMucin which can induce a polyclonal activation via a combination of multiple epitopes for both CD4 + and CD8 + on one sequence. This property seems to compensate in this experiment for the lack of repeated activation. Additional support for this assumption is provided by the high level (ng/ml) of cytokine release from the naive T cells, which are usually if at all, at the range of pg/ml (i.e. three folds lower).
  • an enriched ImMucin-specific T cell subpopulation was produced via repeated stimulation with ImMucin.
  • the specificity of the subpopulation for ImMucin, as well as to the other VXL epitopes, was examined using proliferation assays, cytokine release assays, FACS analysis in which the percentage and type of cells enriched during the enrichment process was analyzed, and using a CTL assay against selected target cells.
  • PBL For producing the specific T cell subpopulation PBL were stimulated three times for 7, 5 and 2 days with ImMucin presented via DC and PBMC. Following the 3 rd stimulation the cells were evaluated against the different VXL epitopes. _ _
  • each one of the MUCl SP epitopes VXLl, VXL2, VXL4 and VXL5 induced a specific proliferation with an average IS ranging from 2.66 to 3.5 which was comparable to the SI achieved using ImMucin itself.
  • VXL-Il the SP epitope which is not deduced from the MUCl antigen
  • VXL-6 which is a MUCl epitope which is not deduced form the SP domain manifested lower SI with an average SI of 1.14-1.125 or 2-5 times lower than the MUCl SP epitopes.
  • the cytokine profile obtained from the specific T cell subpopulation stimulated with the MUCl SP epitopes VXLl, VXL2, VXL4 and VXL5 conform to the proliferation results, showing again that antigen specificity is positively correlated with cytokine release (Table 9).
  • ImMucin and its SP epitopes VXLl, VXL2, and VXL4 manifested high IL-2 and IFN-gamma secretion compared to low to moderate levels induced by the other epitopes.
  • the secretion of TNF- Alfa which is less specific and appears in an early stage is high in all the epitopes tested.
  • the positive results achieved in this experiment are unique compared with results obtained with class I peptide vaccines since in this experiment ImMucin was not matched with the relevant HLA alleles of the donors and still received a positive proliferation.
  • Table 8 Proliferation of a specific T-cell subpopulation which underwent three stimulations with ImMucin or different VXL-peptides as assessed in a proliferation assay. Results represent one out of two experiments using four donors. - -
  • Table 9 ELISA quantitative assay for Cytokine secretion profiles of T-CeIl specific subpopulation (Clones) stimulated with ImMucin and various VXL-Peptides. The results represented summary of two experiments using four donors.
  • a breast tumor cell such as MDA-MB-231 which expresses both MUCl and HLA- A2.1 was lysed effectively (23%), while another breast tumor cell line MDA-MB-468 which expresses only MUCl but not the HLA- 2.1 was not lysed al all.
  • Other control cell-lines didn't manifest any lysis (see table 10).
  • ImMucin is able to induce antigen specific CD4 + as well as CD8 + T cell activation which can lead to effective anti-tumor vaccine properties including lysis, cytokine release and memory in the majority of the population.
  • Table 10 CTL assay of HLA-A2.1 positive T-CeIl specific sub-population (Clone) stimulated with ImMucin. MUCl expression (+ represents low, +++ represents high) was measured by the H23 anti-MUCl mAb. HLA- A21 expression was measured using the BB7.2 mAb. The results represent two separate experiments using two different donors.
  • VXL102 SEQ ID NO. 7
  • VXLlOl SEQ ID NO 1
  • ARMET novel protein
  • VXL-102 manifested lower SI than VXL-101 and ImMucin but still higher than the non-SP epitope VLX-6.
  • a similar pattern of results was obtained in ELISA assay analyzing the cytokine profile of the peptide-activated T cells (Table 12). While all peptides induced high secretion levels of TNF- Alfa ( ⁇ 10ng/ml), only ImMucin induced IL-2 and VXL-101 induced INF-Gamma suggesting that enrichment of CD4+ and CD8+ T cells occurred.
  • VXL-102 didn't induced IL-2 or IFN-gamma production. This moderate immunogenic profile could be associated with the HLA match of the two donors used in this experiment.
  • Table 11 Index of stimulation for PBL by VXL-Peptides. Results represent an average of two donors. - -
  • Table 12 ELISA quantitative assay for Cytokine secretion profiles of PBL stimulated once with ImMucin and two additional tumor associated SP vaccinesVXL-101 and VXL- 102 The results represent stimulation with peptide pulsed DC in ratio 16:1 one out of two experiments using two different donors.
  • Treon SP Maimonis P, Bua D, Young G, Raje N, Mollick J, Chauhan D, Tai YT, Hideshima T, Shima Y, Hilgers J, von Mensdorff-Pouilly S, Belch AR, Pilarski LM, Anderson KC. Elevated soluble MUCl levels and decreased anti-MUCl antibody levels in patients with multiple myeloma. Blood 96;3147-53 (2000)

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to cancer vaccines composed of the signal peptide domain of tumor associated antigens or proteins. The peptide vaccines of the invention are characterized by having multiple MHC class I and class II epitopes which are highly abundant in the population. Therefore, these vaccines are likely to induce a strong, comprehensive immune response against the target proteins in the majority of the vaccinated population, and thereby induce an immune reaction against tumors expressing such target proteins. Specifically, the invention relates to peptide vaccines composed of the signal peptide domain of Mucin (MUC1), BAGE-1 or ARMET, and their use for the treatment of cancers which express Mucin (MUC1), BAGE-1 or ARMET.

Description

ANTIGEN SPECIFIC MULTI EPITOPE VACCINES
FIELD OF THE INVENTION
The present invention relates to cancer peptide vaccines with pan HLA class I and class II binding properties, as well as to pharmaceutical compositions containing the peptide vaccines and methods for treating or preventing cancer.
INTRODUCTION
Cellular active immunotherapy
Widespread metastatic disease is considered by the medical establishment to be incurable, since surgery and radiation are not viable treatments, and response rates with current chemotherapy regimens are low. Also, the toxic side effects of nonspecific chemotherapeutic agents often limit the dose that can be administered.
Recent Meta Analysis compared the incidence of cancers in people with HIV/ AIDS to these with immunosuppressed transplant recipient and showed similarity of the pattern of increased risk of cancer in the two populations (Grulich et al, 2007) This finding suggests that it is the immune deficiency, rather than other risk factors for cancer, that is responsible for the increased risk in these two patient populations.
Active immunotherapy approaches such as cancer vaccines offer hope for cancer specific therapy that could eradicate metastatic tumor cells from the body to achieve a complete cure (Kedar et al., 1995). The rational behind this strategy is based on the following: o It is now well established that immune cells, predominantly T lymphocytes, can recognize Tumor Associated Antigens (TAA) and kill tumor cells. o It is also well appreciated, that cancer cells appear to be fully sensitive to tumor-specific T lymphocytes, suggesting that an anti-tumor vaccination is attainable. (Gilboa E. 2004).
Nevertheless, there are few major differences between therapeutic vaccines such as anti-cancer vaccines and prophylactic (preventive or "conventional") anti-infective vaccines. First, unlike prophylactic vaccines, therapeutic vaccines are generally expected to treat sick individuals, suggesting that a broader and stronger immune response is required. Second, prophylactic vaccines are generally induced against highly immunogenic "foreign" epitopes derived from viruses or bacteria and thus easily induce a strong response with a high number of T cell specific clones. In contrast, therapeutic vaccines in particular cancer vaccines composed of self derived TAAs are less immunogenic and therefore are frequently associated with low or minimal induction of activated T cell clones. Lastly, it is more difficult to induce an effective response in cancer patients, which are, at least temporarily immune- suppressed, than in healthy immune-competent individuals which are the target market of prophylactic vaccines.
For that extent, a desired therapeutic vaccine would need to prime a robust cellular reaction, which will involve multiple clones of T cell lymphocytes predominantly T killer (CD8+) and T Helper (CD4+).
Cancer vaccines
Strategies in cancer immunization have taken many forms based on the antigen used as the immunogenic determinant. These approaches include, among others, the use of whole tumor cell, tumor cell extraction, purified peptide, protein or DNA of selected TAA/s. The products in development can be divided into two main groups: 1. Vaccines consisting of selected MHC class I-restricted sequences from a certain TAA/s. These vaccines consist of high purity sequence/s resembling immunogenic epitopes of a given TAA/s
2. Vaccines consisting of the entire sequence of a certain TAA/s. In this approach the vaccine is composed of a mixture (at various levels of purity) of immunogenic and non-immunogenic MHC class I-restricted epitopes of a given TAA/s.
Vaccines with defined Class I -restricted epitopes
MHC class I-restricted TAA peptides are the targets of Cytotoxic T lymphocytes (CTL), which constitute one of the powerful effectors of the immune system against tumors (Townsend et al., 1989). These peptide vaccines are usually 8 to 10 amino acids (AA) long, with 2 to 3 primary anchor residues that interact with the Major Histocompatibility complex (MHC) class I molecules and 2 to 3 AA residues that engage the T-cell receptor on CD8+ cells (Rammensee et al., 1993). Several methods have been employed to identify tumor associated epitopes. One such method is the identification of CD8+ epitopes subsequent to the search for MHC-binding motifs in known putative TAAs, (Kast et al., 1994) as was shown in the case of the breast- carcinoma-associated HER-2/neu receptor (Fisk et al, 1995) or the colorectal tumor associated Carcino-Embryonic Antigen (CEA) (Ras et al, 1997).
Preclinical evaluation of the isolated MHC class I-restricted TAA peptides manifested promising results both in vitro and in vivo (Mandelboim et al, 1994; Mandelboim et al, 1995). Yet, in spite of the vast preclinical experience gathered over the last two decades, the clinical benefit of these MHC class I- restricted peptide vaccines, (most of which are HLA-2.1 -restricted), manifested a low level of response. That is to say, there was moderated improvement in clinical parameters beyond the induction of a measurable immune response (Marchand et al, 1995; Rosenberg et al, 1998; Jaeger et al, 1996 Rosenberg et al, 2004). The main explanations for this outcome are the following:
A. The limited repertoire of anti-tumor CD8+ T cell clones that could be induced against a single MHC-class I restricted immunogenic epitope. Namely, it is most likely that these vaccines are inducing an overall weak response via T cell clones that are restricted merely to a single immunogenic epitope on one MHC class I- restricted allele. Since the abundance of one epitope on tumor cell is limited, the chance of effective immunotherapy is low. In this regards, one also needs to appreciate that vaccines restricted to a single MHC class I allele are applicable only to the specific patients who are positive for the selected allele (up to 35% of the population in case of the most frequent alleles).
B. The lack of CD4+ T cells activation. CD4+ T cell activation is mediated via MHC class II-binding epitopes and is critical for the initiation (priming) and long lasting memory of most immune responses. CD4+ T cells have a key role as effector cells with anti-tumor properties.
The need for CD4+ signal for mounting an effective immunity of CD 8+ T cells is a well-documented process known as "immune priming". CD4+ T-cell responses are essential to promote the accumulation of Antigen-Presenting Cells (APC) for effective immune priming (Hung et α/.1998) and also for extending the life of antitumor CD8+ T cells i.e. memory response vs. short living response. The limited number of known antigen specific MHC class II epitopes, led in many cases to the administration of MHC class I epitopes with universal non-specific MHC class II- restricted epitopes such as the pan-class II epitope peptide PADRE (Weber et al., 1999). Although response against the universal MHC class Il-restricted epitopes was increased, elevation in CD8+ T-cell effectors specific to the MHC class I-restricted epitope have been limited (Weber et al., 1999).
Another important feature of CD4+ T cells is their role as effector cells with direct anti-tumor activity (Pardoll and Topalian 1998, Christopher et al., 2000). However, since, unlike MHC class I peptide, MHC class II peptide ligands do not have restricted binding properties, their isolation is more complicated. Thus, successful attempts in this direction were limited and arrived only more recently along with the development of sophisticated in-silico class II prediction software and class II transgenic mice (Chaux et al, 1999; Manici et al, 1999). Nevertheless, selected publications e.g. the isolation of class II epitopes like the HLA-DRl 3, melanoma- associated MAGE-3 epitope (Chaux et al., 1999) and HLA-DRB 1*0401 -Restricted Human gplOO (Pardoll and Topalian 1998, Christopher et al., 2000), emphasized the importance of these epitopes for more effective cancer vaccines.
Vaccines consisting of the entire TAA or (Non-defined) epitopes
The other strategy used to overcome the limited repertoire of anti-tumor CD8+ T cell clones is the use of the entire TAA rather then selecting and defining the only relevant immunodominant epitopes. This strategy is more straightforward, as one does not need to isolate the immunogenic epitopes within a given TAA. However, it may very well lead to the "dilution" of the immunogenic epitopes with less immunogenic epitopes, hence decreasing the level of specific immunity or reduce the repertoire of anti-tumor CD8+ T cell clones. Furthermore, some of these less immunogenic epitopes could potentially induce a status of "Immune Anergy" (non responsiveness) which can potentially lead to a decrease in the intensity of the specific immunity or even to status of autoimmunity.
As for the induction of CD4+ specific response, the majority of these vaccines where not designed with the ability to induce CD4+ response via specific MHC class II epitopes.
Knutson and her colleagues (Knutson et al., 2001) prepared antigen specific MHC class II "helper" peptides which contain encompassed class I binding motifs. In a set of studies, the researchers raised the question whether HER-2/neu-specific CD8+ T- cell immunity could be elicited using HER-2/neu-derived MHC class II "helper" peptides, which contain encompassed HLA-A2— binding motifs. The study was performed on nineteen HLA- A2 patients with HER-2/neu-over expressing cancers. After vaccination, the frequency of peptide-specific T-cell precursors specific to the HLA-A2 peptides increased in the majority of patients. In addition, the peptide- specific T cells were able to lyse tumors. More importantly, the responses lasted for a long time and were detectable for more than a year after the final vaccination in select patients. This study suggested an improved anti-cancer immunity via combination of class-I and class-II epitopes derived from the same TAA.
In summary, cancer therapeutic vaccines are required to be: o Antigen specific to avoid potential Anergy and autoimmunity. o Highly potent in inducing a strong, comprehensive and long lasting response involving CD4+ plus CD8+ T cells. o Applicable in the majority of the target population.
Signal peptides
Almost half of the proteins of an average cell are translocated across membranes. Proteins directed into the secretory pathway use amino-terminal signal peptides to interact with the translation machinery. The translocation of secretory proteins across intracellular membranes and final localization are mediated by signal peptides (SP) which are 'address tags' contained within their amino acid sequences. Signal peptides, comprising the N-terminal 15-60 amino acids of proteins, are necessary for the translocation across the membrane on the secretory pathway and thus universally control the entry of all proteins both in eukaryotes and prokaryotes to the secretory pathway. Signal peptides generally consist of three parts: an N-terminal region of differing length, which usually comprises positively charged amino acids; a hydrophobic domain; and a short carboxy-terminal peptide segment. In eukaryotes, the signal peptide of a nascent precursor protein (pre-protein) directs the ribosome to the rough Endoplasmic Reticulum (ER) membrane and initiates the transport of the growing peptide chain across it. hi prokaryotes, the signal peptide directs the pre- protein to the cytoplasmic membrane. However, the signal peptide is not responsible for the final destination of the mature protein; secretory proteins devoid of further address tags in their sequence are by default secreted to the external environment. Signal peptides are cleaved from precursor proteins by an endoplasmic reticulum (ER)-resident signal peptidase or they remain uncleaved and function as a membrane anchor. During recent years, a more advanced view of signal peptides has evolved, showing that the functions and irnmunodorninance of certain signal peptides are much more versatile than previously anticipated.
Signal peptides and elevated immunity
Selected reports have revealed the advantages of linking signal peptide sequences derived from various origins, to selected immunogenic epitopes, hi the context of cancer immunotherapy, research by Sherritt and her colleagues has shown that the linking of signal peptide from the adenovirus E3/19 kDa protein to several murine melanoma associate epitopes results in a superior production of CTL as well as in better CTL mediated immune protection (Sherritt et al., 2001). Minev and her colleagues demonstrated that linking signal peptide to a defined MHC-class I- restricted epitope, as in the case of the Melanoma MART-I TAA, enhances the epitope presentation i.e. the signal peptide enhances the ability of the epitope to attract and activate CD8+ T cells (Minev BR, et al, 2000). In both of these reports, immunity to the TAA epitopes was increased in both Transporter for Antigen Presentation (TAP) -deficient and TAP-expressing cells, suggesting that signal peptides can facilities TAP independent presentation of TAA epitopes. According to these reports the signal peptide merely acts as a chaperon or tag for selected epitopes/Vaccines. These vaccines, however, suffer from several drawbacks as they utilize selected immunogenic epitopes, MHC-class I-restricted peptides with limited repertoire for activation, or non-defined immunogenic epitopes with nonspecific activity.
The MUCl TAA and its role in cancer
MUCl is one of the most promising TAAs known today. This polymorphic epithelial mucin, encoded by the MUCl gene, is a high-molecular- weight glycoprotein with few alternative-splicing variants encoding for both a transmembranal (i.e. across the cell membrane) and a secreted (i.e. circulating) product both expressed in a broad range of tumors (Graham et al., 1996; Ho et al., 1993). MUCl is one of the few known targets that are expressed by more than 90 per cent of common solid tumor cancers including Colon, Gastric, Lung, Renal Cell (RC), Transitional Cell (TC), Prostate, Pancreas, Breast, Ovary and Thyroid. It is also associated with many non-solid tumors among which: Lymphoma, Leukemia, and Multiple Myeloma (MM). The association of MUCl with cancer progression was well documented in the literature (McGuckin et al., 1995). Many groups have shown that MUCl is expressed on the cell surface of most MM cell lines, MM patient plasma cells and circulating B cells, and plasmacytomas (Treon SP et al., 2000). In addition, soluble MUCl has also been detected in peripheral blood plasma of MM patients by the use of a noncommercial enzyme-linked immunosorbent assay (ELISA). Treon and his colleagues also determined elevation in soluble MUCl levels in MM patients using an immunoassay that recognizes the CA27.29 MUCl epitope. He further demonstrate that MUCl levels are elevated in both bone marrow (BM) and peripheral blood plasma of MM patients in comparison to healthy donors, and that BM MUCl levels are associated with tumor burden in MM patients.
A major feature of the MUCl molecule (in both the transmembranal and the secreted variants) is the presence of a highly immunogenic extracellular tandem repeat array (TRA) heavily O-glycosylated at serine and threonine residues. It was shown that this extracellular TRA domain can be recognized by monoclonal antibodies (MAbs), as well as MHC-restricted CD8+ T cells (CTLs) such as HLA-AI l and HLA-A2.1- CTLs. In spite of the high immunogenicity of the TRA, its role as a potential target/vaccine is ambiguous. One of the major drawbacks for targeting MUC l's TRA stems from the finding that most of the immunogenic epitope in the TRA domain exists both in the extracellular as well as in the secreted products. That is to say, that the secreted variant is acting as decoy that negatively interferes or competes with any potential drugs/vaccines. In addition, reports have indicated that synthetic peptides derived from MUC l's TRA cause suppression of human T-cell proliferative responses (Fung and Longenecker, 1991).
More recently, reports have showed Class I (HLA-A2.1 -restricted) CD 8+ T cell epitopes which were deduced from other domains on the MUCl protein (Cannon et al., 2000, Brossart et al., 1999; WO 00/06723; WO 00/63363). These epitopes (termed D6 or VXLl and Ml .2 or VXL2) were shown to be restricted only to a single class I epitope.
Another example of a tumor associated antigen is BAGE. BAGE, codes for a putative protein of 43 amino acids and seems to belong to a family of several genes. Gene BAGE is expressed in 22% of melanomas, 30% of infiltrating bladder carcinomas, 10% of mammary carcinomas, 8% of head and neck squamous cell carcinomas, and 6% of non-small cell lung carcinomas. It is silent in normal tissues with the exception of testis (Boel et al., 1995).
Another example, Arginine rich, mutated in early stage of tumors (ARMET), also designated Arginine-rich protein (ARP), is a highly conserved gene that maps to human chromosomal band 3p21.1. This gene contains an imperfect trinucleotide repeat which encodes a string of arginines. A specific mutation (ATG50~>AGG) was detected within this region of the gene in a high percentage of sporadic renal cell carcinomas, squamous cell carcinomas of the head and neck, small cell lung cancer cell lines, non-small cell lung carcinomas, breast tumors, and prostate tumors (Shridhar et al., 1996).
SUMMARY OF THE INVENTION
The present invention relates to promiscuous peptide vaccines comprising multiple MHC class I, and MHC class II epitopes of a given protein antigen. More particularly, the present invention relates to promiscuous peptide vaccines comprising multiple MHC class I and MHC class II epitopes with the specificity of a given antigen derived from the entire signal peptide domain of that protein antigen. These MHC class I and MHC class II epitopes have a high frequency in the population and thus the vaccine is effective in a large portion of the population.
The present invention thus provides a peptide vaccine which is able to induce strong, comprehensive response in the majority of the target population against said antigen. More specifically, but without wishing to be limited to a single hypothesis, such a vaccine preferably combines activation of both CD4+ and CD8+ T cells via multiple CD4+ and CD8+ -restricted epitopes which are present within the internal sequences of the vaccine and are derived from the same antigen.
In one aspect, the present invention relates to such peptide vaccines comprising the signal peptide domain of tumor associated antigens (TAA) or the signal peptide domain of proteins which are over-expressed in tumor cells.
In one embodiment, the present invention relates to peptide vaccines comprising the signal peptide of a protein which is either a TAA or is over-expressed in tumor cells, wherein said peptides are recognized and presented by more than 50% of the MHC class I and MHC Class II alleles in the population. Preferably, said peptide is not longer than 50 amino acids, more preferably, not longer than 25 amino acids.
In one embodiment the peptide vaccines of the invention comprise the signal peptide of proteins selected from the group consisting of Armet, HSP60, CANX, MTHFD2, FAP, MMP6, BAGE-I, GNTV, Q5H943, MUCl, CEA, Pmel, Kallikrein- 4, Mammaglobin-1, MART-I, GPR143-OA1, PSA, TRPl, Tyrosinase, FGF-5, NEU proto-oncogene, Aft, MMP-2, PSMA, Telomerase-associated protein 2, PAP, Uroplakin II and Proteinase 3, i.e. SEQ ID Nos. 1-28, respectively (Table 1).
According to one specific embodiment, the present invention relates to tumor associated antigen peptides comprising the signal peptide domain of the polymorphic epithelial mucin, encoded by the MUCl gene.
The present invention thus provides a promiscuous peptide vaccine comprising the MUCl signal peptide domain which is able to induce strong, comprehensive response in the majority of the target population against any MUCl positive tumor.
The MUCl signal pepti de-derived peptide vaccines are able to bind to the majority of MHC Class I alleles in the population and thus induce CD8+ T-cell mediated cell lysis, and are also able to bind to bind to the majority of MHC Class II alleles in the population and thus prime an effective CD4+ T-cell mediated immune response.
In one embodiment the MUCl signal peptide-derived peptide vaccine comprises the amino acid sequence MTPGTQSPFFLLLLLTVLTW (SEQ ID NO. 10).
In another embodiment, the peptide vaccine of the invention comprises a mixture of at least two short peptides of preferably about nine amino acid residues in length derived from the signal peptide domain of the MUCl protein. These peptides represent various MHC Class I and Class II epitopes which are included in the MUCl signal peptide. Their combination results in effective binding of the vaccine composition to various alleles of MHC class I and MHC class II molecules, and thus to the induction of an immune response to tumors expressing the MUCl protein. This response may include inducing "help" for priming a strong T cell activity via CD4+ T cell activation, combined with induction of CD 8+ T cell activation, and potent cellular activity (CTL) against MUCl expressing tumors.
Specifically, the mixture of short peptides comprises at least two peptides selected from the group consisting of SEQ ID NO: 29-39.
In one embodiment the mixture of short peptides comprises VXLOl (SEQ ID NO 29), VXL02 (SEQ ID NO 30), VXL04 (SEQ ID NO 31) and VXL05 (SEQ ID NO 32). _
In another specific embodiment, the present invention relates to peptide vaccines derived from the signal peptide domain of the BAGE-I gene.
In one embodiment the BAGE-I signal peptide-derived peptide vaccine comprises the amino acid sequence MAARA VFLAL SAQLLQA (SEQ ID NO. 7).
In another specific embodiment, the present invention relates to peptide vaccines derived from the signal peptide domain of the Armet gene.
In one embodiment the Armet signal peptide-derived peptide vaccine comprises the amino acid sequence MWATQGLAVA LALS VLPGSR A (SEQ ID NO. 1).
The present invention also concerns use of the peptide vaccines described above in the preparation of pharmaceutical compositions for treating or inhibiting cancer.
The invention further concerns pharmaceutical compositions comprising said peptide vaccines and the use of said peptide vaccines or said pharmaceutical compositions as anti-tumor vaccines to treat or inhibit the development of cancer. Specifically, for the treatment of tumors which over-expresses the protein from which the signal peptide vaccine was derived, for example, MUCl -expressing cancer, BAGE-I -expressing cancer, or Armet-expressing cancer.
The invention further concerns nucleic acid molecules encoding said peptides, and antigen presenting cells (APC), e.g. dendritic cells, presenting said peptides, as well as pharmaceutical compositions comprising said nucleic acid molecules, or said cells.
The invention also concerns use of the peptide vaccines for enrichment of T cell populations in vitro. Thus obtaining a peptide-specific enriched T cell population.
The invention further concerns the use of said nucleic acid molecules, cells, or pharmaceutical compositions comprising same as anti-tumor vaccines to treat or inhibit the development of cancer. Specifically, for the treatment of tumors which over-expresses the protein from which the signal peptide vaccine was derived, for example, MUCl -expressing cancer, BAGE-I -expressing cancer, or Armet- expressing cancer.
Further aspects of the present invention are directed to a method for treating or for inhibiting the development of cancer by administering the pharmaceutical compositions of the present invention to a patient in need thereof. The pharmaceutical compositions of the invention may be adapted for use in combination with other anti neoplastic agents.
Figure imgf000014_0001
Table 1: List of signal peptide vaccines
_ _
BRIEF DESCRIPTION OF THE DRAWINGS
In order to understand the invention and to see how it may be carried out in practice, embodiments will now be described, by way of non-limiting example only, with reference to the accompanying drawings, in which:
Figure 1 is a graph showing results of an ELISA quantitative assay measuring cytokine secretion profiles of a specific T cell subpopulation developed via repeated stimulation with ImMucin. The results represent one out of two experiments using four different donors.
Figure 2 is a graph showing FACS analysis of T cell phenotype evaluation during consistent stimulation with ImMucin. The Results represent one out of two experiments using four different donors.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides antigen specific vaccines which are capable of inducing a robust T-cell immunity and which are applicable to the majority of the population.
Whilst signal peptides (SP) have been used as non-specific immune stimulators for elevating the immunity of antigenic epitopes to which they were linked, their use as a source for antigens/epitopes with independent immunogenic properties, in addition to their stimulatory properties was not disclosed previously.
The present invention is based on the surprising finding that SP-derived vaccines are able to bind simultaneously to multiple alleles of both MHC class I and MHC class II i.e. CDA+ and CD8+-restricted epitopes. A signal peptide vaccine, although containing just one sequence, could thus be compared to a large number of single Class I and Class II epitopes, used in a mixture. This newly discovered feature of SP-vaccines facilitates the generation of a robust immune response in the majority of the target population. Moreover, but without wishing to be bound by theory, it should be emphasized that signal peptide-based vaccines bare the ability to independently penetrate the ER and thus, at least partially, avoid immune escape mechanisms such as TAP deficiency. ^
One non-limiting procedure/algorithm for selecting candidate vaccines in accordance with the present invention is described below. This procedure combines in-silico analysis and reverse immunology.
Specifically, the objective of the predictive algorithm is to identify and obtain signal peptide (SP) targets with a potential role as cancer vaccines. The putative targets should have the following characteristics:
Differentially expressed in tumor cells; Eligible targets for an immune assault; and
- Predicted as binding MHC Class I and Class II alleles (and therefore likely to be immunogenic) in the majority of the population.
Providing a list of putative cancer protein targets
A person versed in the art may find many information sources in the literature, providing data on tumor associated proteins. As a non-limiting example, see Rhodes DR and Chinnaiyan AM. (2005), who performed a meta-analysis of many microarray gene expression studies, checking differential gene expression in normal vs. cancer tissues of the most prominently appearing cancers. In addition, many web available databases provide lists of tumor-specific antigens. Non-limiting examples include: a. Tumor specific (unique) antigens (http://www.cancerirnmunitv.org/peptidedatabase/tumorspecific.htm) b. Tumor antigens that are tissue-specific (differentiation) antigens (httpV/www.cancerimmunitv-org/peptidedatabase/differentiation.htm) c. Tumor antigens that are overexpressed in cancer cells vs. the respective normal tissue (http://www.cancerimmunitv.org/peptidedatabase/overexpressed.htm)
Exclusion of non eligible targets for an immune assault
Proteins with the following attributes are removed from the list of putative targets as being non eligible for an immune assault:
Proteins that are located sub-cellularly, e.g. in organelles or in any other location that does not require transport from the ER-Golgi, and thus have no signal peptides (e.g. purely cytoplasmic proteins, such as ATP-citrate synthase). Proteins which function in basic or homeostatic functions in all cells (e.g. purine synthesis, for example phosphoribosylaminoimidazole carboxylase, phosphoribosylaminoimidazole succinocarboxamide synthetase (PAICS), succinate dehydrogenase, cytochrome b556 subunit (SDHC), cell division cycle 2 (CDC2).
Proteins that are ubiquitously expressed in many tissues (e.g. tubulin, beta TUBB, RNA binding motif protein 4 (RBM4).
Immune-related proteins (e.g. proteasome (prosome, macropain) activator subunit 2 PSME2, CD213a2, Macrophage colony-stimulating factor (M-CSF).
Identifying Signal Peptide sequences
Proteins that are found to be eligible targets for an immune assault are next examined for the presence of a signal peptide. This may be done by using appropriate computer software, e.g. the Signal P 3.0. The Signal P 3.0 program uses both a neural network (NN) algorithm and a Hidden Markov models (HMM) algorithm for selection of the signal (http://www.cbs.dtu. dk/services/SignalPA). A sequence was considered to be a signal peptide whenever a score of over 0.2 was received in one or more of the algorithms. Sequences having a score of above 0.7 are preferred. Sequences having a score of above 0.8 are most preferred.
Preferably, cancer protein targets, eligible for an immune assault and having an identified signal peptide sequence of 17-50 amino acids are selected for further examination of predicted binding to MHC alleles.
Predicting MHC class I and II binding
A prediction of putative binding of the selected candidate signal peptide sequences to frequently occurring HLA haplotypes is made based on information concerning HLA allele frequency (class I and II) which may be obtained, for example, from the dbMHC site belonging to the NCBI. (http://www.ncbi.nlm.nih.gov/projects/mhc/ihwg.cgi?cmd=PRJOV&ID:=9').
Alleles of HLA class I (HLA-A, B, C) and HLA class II (HLA-DRBl) which most frequently appear in the population are listed in Table 2. The table specifies the most frequent alleles (appearing in over 5% of the population) for which prediction methods exist.
Figure imgf000018_0001
Table 2: Alleles of HLA class I (HLA-A, B, C) and HLA class II (HLA-DRBl) which most frequently appear in the population
Subsequently, the binding strength of the previously identified signal peptides to the HLA alleles is predicted using any of numerous available software programs. The following is a non-limiting list of available prediction programs:
BIMAS (http://www-bimas.cit.nih.gov/molbio/hla bind/) may be used for the prediction of HLA class I alleles.
■ Propred (http://bic.uams.edu/mirror/propred/) may be used to predict most DRBl genotypes.
Immune Epitope
(http://tools.immuneepitope.org/tools/matrix/iedb input?matrixClass=II) may be used for the prediction of the HLA-DRB 1-0901 genotype that is not predicted by Propred.
■ MHC2Pred (http://www.imtech.res.in/raghava/mhc2pred/index.html) may be used to predict various DRBl serotypes.
Defining differential strength of binding In each of the programs used, various differential strength of binding are defined:
BIMAS: Strong= peptide score of 100+, Medium=10-100, Weak=5-10.
Propred: Strong=top 1% of binders, Medium= 1-2% of binders, Weak=2-3% of binders.
Immune Epitope: Strong=IC50 of 0.0 InM- 1OnM, Medium=10-100nM, Weak=100-10,000 nM.
MHC2Pred: Strong=cutoff 1.0, medium=cutoff 0.5, Weak =cutoff 0. As serotype prediction is expected to be less accurate than genotype prediction, only high and medium binders were predicted with MHC2Pred.
Determining the predicted percentage of population that has alleles having predicted binding peptides within a specific signal peptide
To calculate the probability that a patient (or a population) has one or more alleles predicted to bind a certain signal peptide, a statistic calculation using complementary probabilities is performed. Independent distribution of alleles in the population was assumed.
Explanatory calculation: if peptide X was predicted as a peptide that binds to only four HLA-class I alleles: HLA-Al (frequency 0.1), HLA-B2 (freq=0.2), HLA-B3 (freq.=0.3), and HLA-C4 (freq. 0.4) then the probability that it would bind neither of these alleles is the product of the probabilities that it would bind neither HLA-Al (1- 0.1), nor HLA-B2 (1-0.2), nor HLA-B3 (1-0.3), nor HLA-C4 (1-0.4) therefore the probability is: (l-0.1)(l-0.2)(l-0.3)(l-0.4)= 0.3024.
The probability that the patient has one or more of the binding alleles is 1 minus the probability that he would have none of the binding alleles:
1-0.3024=0.6976
The calculation was done separately for the HLA class I alleles, the HLA-class II alleles (genotypes), and the HLA class II alleles (serotypes). Each list contained no overlapping alleles (e.g. HLA-A02 and HLA-A0201).
Peptides that would bind in the majority (>50%) of the population (both in the HLA class I and in the HLA class II alleles) were further followed. In addition, targets that have an abundance of at least 40% in their class II serotypes were also included. This is due to the fact that only about 75% of class II serotypes were covered in the screen, assuming that with inclusion of the remaining 25%, the threshold of 50% will be achieved.
Selecting appropriate vaccine candidates
Preferably, the most suitable SP vaccine candidates are chosen according to the following criteria:
1. Having a high score for SP of more than 0.7 in the Signal P software, as measured by at least one but preferably by both tests, NN and HMM. Having a higher score than 0.8 in the Signal P software is preferred.
2. >50% allele frequency for both Class I, and Class II molecules.
3. Length of SP Peptide of up to 50 AA, preferably up to 30 AA, most preferably up to 25 AA.
4. Known role as a tumor associate antigen (TAA) or being over expressed in tumor vs. normal tissues.
5. Broad Expression of the TAA in various cancer tissues.
Utilizing this novel procedure, the entire sequence of signal peptide domains from over 30 potential targets, predominantly TAA was scanned in order to identify vaccines having both immune enhancing and immunogenic properties. Preferred candidates conforming to the above-defined criteria were identified and are listed in Table 1.
In one specific example, a MUCl SP vaccine (hereinafter termed "VXLlOO" or "ImMucin") was prepared. The ImMucin vaccine of the invention is composed of a 21 amino acid (AA)-long peptide derived from the signal peptide domain of the MUCl protein and comprises the amino acid sequence MTPGTQSPFFLLLLLTVLTVV (SEQ ID NO 10).
This peptide vaccine is processed in the antigen-presenting cell (APC) and presented to immune effector cells by MHC class I, and II molecules.
ImMucin harbors a set of unique characteristics as listed below: o ImMucin targets the mucin MUCl expressed on the surface of Multiple
Myeloma and other solid and non-solid cancers. o Unlike other MUCl vaccines which focus on the entire protein, ImMucin does not contain any non-specific epitopes that could dilute and disturb specific anti-cancer immunity. o ImMucin was selected due to its ability to bind multiple MHC Class I and Class II alleles. This would potentially offer:
A broad activation of multiple T cell clones both CD8+ and CD4+ T cells, by use of a single 21 AA peptide. This could actually be considered equal to a selected mixture of 11 different immunogenic epitopes.
A positive respond is expected in the majority of the target patients i.e. covering most of the allelic repertoire among the Caucasian and other populations. o Due to the unique sequence of Signal peptides, ImMucin has superior immunogenicity which may, at least partially, circumvent immune escape mechanisms such as TAP-deficiency of cancer cells.
According to one embodiment, the present invention concerns a vaccine comprising a mixture of short peptides comprising MHC Class I and II epitopes within the MUCl signal domain. These short peptides include:
SEQ ID NO 29: LLLTVLTVV (designated VXLOl) SEQ ID NO 30: LLLLTVLTV (designated VXL02) SEQ ID NO 31 : TQSPFFLLL (designated VXL04) SEQ ID NO 32: SPFFLLLLL (designated VXL05) SEQ ID NO 33: FLLLLLTVL SEQ ID NO 34: LLLLLTVLT SEQ ID NO 35: GTQSPFFLL SEQ ID NO 36: TPGTQSPFF SEQ ID NO 37: FFLLLLLTV SEQ ID NO 38: MTPGTQSPF SEQ ID NO 39: QSPFFLLLL
As used herein, the term "tumor associated antigen " or "TAA " refers to antigens or proteins that are highly correlated with certain tumor cells. They are not usually expressed in normal cells, or are expressed at a higher extent in tumor cells than in normal cells.
The nomenclature used to describe peptide and/or polynucleotide compounds of the invention follows the conventional practice wherein the amino group (N- terminus) and/or the 5' are presented to the left and the carboxyl group (C-terminus) and/or 3' to the right.
As used herein, the term "peptide" refers to a molecular chain of amino acids, which, if required, can be modified in vivo or in viti'o, for example by manosylation, glycosylation, amidation (specifically C-terminal amides), carboxylation or phosphorylation with the stipulation that these modifications must preserve the biological activity of the original molecule. In addition, peptides can be part of a chimeric protein.
Functional derivatives of the peptides are also included in the present invention. Functional derivatives are meant to include peptides which differ in one or more amino acids in the overall sequence, which have deletions, substitutions, inversions or additions. Amino acid substitutions which can be expected not to essentially alter biological and immunological activities have been described. Amino acid replacements between related amino acids or replacements which have occurred frequently in evolution are, inter alia Ser/Ala, Ser/Gly, Asp/Gly, Asp/Asn, Ile/Val see Dayhof M.D (1978). Based on this information, Lipman and Pearson (1985) developed a method for rapid and sensitive protein comparison and determining the functional similarity between homologous polypeptides.
The peptides according to the invention can be produced synthetically, by recombinant DNA technology. Methods for producing synthetic peptides are well known in the art. - 1*2* -
The organic chemical methods for peptide synthesis are considered to include the coupling of the required amino acids by means of a condensation reaction, either in homogenous phase or with the aid of a so-called solid phase. The condensation reaction can be carried out as follows:
Condensation of a compound (amino acid, peptide) with a free carboxyl group and protected other reactive groups with a compound (amino acid, peptide) with a free amino group and protected other reactive groups, in the presence of a condensation agent;
Condensation of a compound (amino acid, peptide) with an activated carboxyl group and free or protected other reaction groups with a compound (amino acid, peptide) with a free amino group and free or protected other reactive groups.
Activation of the carboxyl group can take place, inter alia, by converting the carboxyl group to an acid halide, azide, anhydride, imidazolide or an activated ester, such as the N-hydroxy-succinimide, N-hydroxy-benzotriazole or p-nitrophenyl ester.
The most common methods for the above condensation reactions are: the carbodiimide method, the azide method, the mixed anhydride method and the method using activated esters, such as described in The Peptides, Analysis, Synthesis, Biology Vol. 1-3 (Ed. Gross, E. and Meienhofer, J.) 1979, 1980, 1981 (Academic Press, Inc.).
Production of peptides by recombinant DNA techniques is a general method which is known, but which has a lot of possibilities all leading to somewhat different results. The polypeptide to be expressed is coded for by a nucleic acid sequence.
Also part of the invention is the nucleic acid sequence comprising the sequence encoding the peptides according to the present invention.
As is well known in the art, the degeneracy of the genetic code permits substitution of bases in a codon to result in another codon still coding for the same amino acid, e.g., the codon for the amino acid glutamic acid is both GAT and GAA. Consequently, it is clear that for the expression of a polypeptide with an amino acid sequence as shown in any of SEQ ID NO: 1-28 use can be made of a derivate nucleic acid sequence with such an alternative codon composition thereby different nucleic acid sequences can be used.
"Nucleotide sequence" as used herein refers to a polymeric form of nucleotides of any length, both to ribonucleic acid (RNA) sequences and to deoxyribonucleic acid (DNA) sequences. In principle, this term refers to the primary structure of the molecule. Thus, this term includes double and single stranded DNA, as well as double and single stranded RNA, and modifications thereof.
The nucleotide sequences encoding the peptide vaccines of the invention can be used for the production of the peptides using recombinant DNA techniques. For this, the nucleotide sequence must be comprised in a cloning vehicle which can be used to transform or transfect a suitable host cell.
A wide variety of host cell and cloning vehicle combinations may be usefully employed in cloning the nucleic acid sequence. For example, useful cloning vehicles may include chromosomal, non-chromosomal and synthetic DNA sequences such as various known bacterial plasmids, and wider host range plasmids such as pBR 322, the various pUC, pGEM and pBluescript plasmids, bacteriophages, e.g. lambda-gt- Wes, Charon 28 and the Ml 3 derived phages and vectors derived from combinations of plasmids and phage or virus DNA, such as SV40, adenovirus or polyoma virus DNA.
Useful hosts may include bacterial hosts, yeasts and other fungi, plant or animal hosts, such as Chinese Hamster Ovary (CHO) cells, melanoma cells, dendritic cells, monkey cells and other hosts.
Vehicles for use in expression of the peptides may further comprise control sequences operably linked to the nucleic acid sequence coding for the peptide. Such control sequences generally comprise a promoter sequence and sequences which regulate and/or enhance expression levels. Furthermore, an origin of replication and/or a dominant selection marker are often present in such vehicles. Of course, control and other sequences can vary depending on the host cell selected. _ _
Techniques for transforming or transfecting host cells are quite known in the art (for instance, Maniatis et al., 1982/1989, Molecular cloning: A laboratory Manual, Cold Spring Harbor Lab.).
The present invention also provides a polynucleotide encoding the signal peptide vaccine of the invention as part of a pharmaceutical composition preferably for targeted treatment of a tumor.
Further aspects of the present invention are directed to a method for treating or for inhibiting the development of cancer by administering the pharmaceutical compositions of the present invention to a patient in need thereof.
The present invention describes a method for treating or inhibiting the development of solid tumors for example, Colon, Gastric, Lung, Renal Cell (RC), Transitional Cell (TC), Prostate, Pancreas, Breast, Ovary or Thyroid cancers, as well as non-solid tumors such as Lymphoma, Leukemia, and Multiple Myeloma.
Specifically, the present invention provides a method for treating or for inhibiting the development of MUCl -expressing cancers by administering the MUCl signal peptide-derived peptide vaccine of the present invention to a patient in need thereof.
In another embodiment the present invention provides a method for treating or for inhibiting the development of BAGEl -expressing cancers by administering the BAGEl signal peptide-derived peptide vaccine of the present invention to a patient in need thereof. Such cancers include melanoma, bladder carcinoma, mammary carcinoma, head and neck squamous cell carcinoma, and non-small cell lung carcinomas.
In another embodiment the present provides a method for treating or for inhibiting the development of Armet-expressing cancers by administering the Armet signal peptide-derived peptide vaccine of the present invention to a patient in need thereof. Such cancers include renal cell carcinomas, lung, breast, prostate, squamous cell carcinoma, head and neck carcinoma, pancreatic carcinoma.
The peptide vaccine of the invention is administered in an immunogenically effective amount with or without a co-stimulatory molecule. According to the method of the invention, the peptide vaccine may be administrated to a subject in need of such treatment for a time and under condition sufficient to prevent, and/or ameliorate the condition of cancer being treated.
The antigen and co-stimulatory molecule, if used, are formulated, separately or as a "chimeric vaccine" formulation, with a pharmaceutically acceptable carrier and administered in an amount sufficient to elicit a T lymphocyte-mediated immune response.
According to the methods of the invention, the peptide may be administered to subjects by a variety of administration modes, including by intradermal, intramuscular, subcutaneous, intravenous, intra-atrial, intra-articular, intraperitoneal, parenteral, oral, rectal, intranasal, intrapulmonary, and transdermal delivery, or topically to the eyes, ears, skin or mucous membranes. Alternatively, the antigen may be administered ex-vivo by direct exposure to cells, tissues or organs originating from a subject (Autologus) or other subject (Allogeneic), optionally in a biologically suitable, liquid or solid carrier.
In certain embodiments of the invention, the peptides or pharmaceutical composition with or without a co-stimulatory molecule are delivered to a common or adjacent target site in the subject, for example to a specific target tissue or cell population in which the vaccine formulation is intended to elicit an immune response. Typically, when the peptide or pharmaceutical composition and the optional co- stimulatory molecule are administered separately, they are delivered to the same or closely proximate site(s), for example to a single target tissue or to adjacent sites that are structurally or fluidly connected with one another (e.g., to allow direct exposure of the same cells, e.g., fluid flow transfer, dissipation or diffusion through a fluid or extracellular matrix of both vaccine agents). Thus, a shared target site for delivery of antigen and co-stimulatory molecule can be a common surface (e.g., a mucosal, basal or lunenal surface) of a particular target tissue or cell population, or an extracellular space, lumen, cavity, or structure that borders, surrounds or infiltrates the target tissue or cell population.
For prophylactic and treatment purposes, the peptide antigen with or without a co- stimulatory molecule may be administered to the subject separately or together, in a single bolus delivery, via continuous delivery (e.g., continuous intravenous or transdermal delivery) over an extended time period, or in a repeated administration protocol (e.g., on an hourly, daily or weekly basis). The various dosages and delivery protocols contemplated for administration of peptide and co-stimulatory molecule, in simultaneous or sequential combination, are immunogenically effective to inhibit the occurrence or alleviate one or more symptoms of the target cancer in the subject. An "immunogenically effective amount" of the antigen thus refers to an amount that is, in combination, effective, at dosages and for periods of time necessary, to elicit a specific T lymphocyte mediated immune response. This response can be determined by conventional assays for T-cell activation, including but not limited to assays to detect proliferation, specific cytokine activation and/or cytolytic activity.
In more detailed aspects of the invention, the amount of peptide vaccine is immunogenically effective to achieve a desired cancer inhibitory effect in the subject. In specific embodiments, an immunogenically effective amount of the peptide, depending on the selected mode, frequency and duration of administration, will effectively prevent cancer, or will inhibit progression of a cancerous condition in the subject. Alternatively or in addition to these effects, an immunogenically effective dosage of the antigen, which may include repeated doses within an ongoing prophylaxis or treatment regimen, will alleviate one or more symptoms or detectable conditions associated with a cancerous disorder. This includes any detectable symptom or condition amenable to prophylaxis and/or treatment with the vaccines of the invention, for example symptoms or conditions associated with breast cancer, cervical cancer, prostate cancer, colon cancer, melanoma and other cancerous conditions.
For prophylactic and therapeutic use, peptide antigens might be formulated with a "pharmaceutical acceptable carrier". As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption enhancing or delaying agents, and other excipients or additives that are physiologically compatible. In specific embodiments, the carrier is suitable for intranasal, intravenous, intramuscular, intradermal, subcutaneous, parenteral, oral, transmucosal or _ _
transdermal administration. Depending on the route of administration, the active compound may be coated in a material to protect the compound from the action of acids and other natural conditions which may inactivate the compound.
Peptide vaccine may be administered to the subject in the form of a peptide solution per se or a combination of a peptide with an appropriate auxiliary agent using an injector. Alternatively, the peptide vaccine may be percutaneously administered through mucous membrane by, for instance, spraying the solution. The unit dose of the peptide typically ranges from about 0.01 mg to 100 mg, more typically between about 100 micrograms to about 5 mg, which may be administered, one time or repeatedly, to a patient.
Examples of auxiliary agents which can be formulated with or conjugated to peptide or protein antigens and/or vectors for expressing co-stimulatory molecules to enhance their immunogenicity for use within the invention include cytokines (e.g. GM-CSF), bacterial cell components such as BCG bacterial cell components, imnunostimulating complex (ISCOM), extracted from the tree bark called QuillA (Morein et al., 1984 incorporated herein by reference), QS-21, a saponin-type auxiliary agent (Wu et al., (1992), incorporated herein by reference), Montanide ISA 51VG, liposomes, aluminum hydroxide (alum), bovine serum albumin (BSA), tetanus toxoid (TT) (Green et al., (1982) incorporated herein by reference) and keyhole limpet hemocyanin (KLH).
In preparing pharmaceutical compositions of the present invention, it may be desirable to modify the peptide antigen, or to combine or conjugate the peptide with other agents, to alter pharmacokinetics and biodistribution. A number of methods for altering pharmacokinetics and biodistribution are known to persons of ordinary skill in the art. Examples of such methods include protection of the proteins, protein complexes and polynucleotides in vesicles composed of other proteins, lipids (for example, liposomes), carbohydrates, or synthetic polymers. For example, the vaccine agents of the invention can be incorporated into liposomes in order to enhance pharmacokinetics and biodistribution characteristics. A variety of methods are available for preparing liposomes, as described in, e.g., U.S. Pat. Nos. 4,235,871, 4,501,728 and 4,837,028, each incorporated herein by reference. For use with liposome delivery vehicles, peptides are typically entrapped within the liposome, or lipid vesicle, or are bound to the outside of the vesicle.
Within certain embodiments of the invention, peptide antigens are associated with liposomes, such as lecithin liposomes or other liposomes known in the art, as a DNA-liposome mixture, or the DNA may be associated with an adjuvant known in the art to boost immune responses, such as a protein or other carrier. Additional agents which assist in the cellular uptake of DNA, such as, but not limited to, calcium ions, viral proteins and other transfection facilitating agents and methods may also be used to advantage (Wigler et al., Cell 14:725, 1978; Corsaro and Pearson, Somatic Cell Genetics 7:603, 1981; Graham and Van der Eb, Virology 52:456, 1973; Neumann et al., EMBO J. 1:841-845, 1982; and Hawley-Nelson et al., Focus 15:73- 79, 1993, each incorporated herein by reference).
EXAMPLES
MATERIALS AND METHODS
Preparation of peptide-pulsed DC
Human PBMC were separated from buffy coat samples of naive donors using Ficoll UNI-SEPmaxi tube. Separated cells were suspended in RPMI medium supplemented with 10% FCS, L-Glutamine, Sodium Pyruvate, MEM-EAGLE non-essential amino- acids, HEPES and Gentamycin Sulphate (Bet-Haemek Industries, IL) and cultivated for 4h at 370C in a culture dish (Corning 150mmx25mm). Adherent cells were collected and cultured for 7 days in serum free DCCM-I medium supplemented with L-Glutamine, huIL-4 (1000IU/ml) (Cytolab IL) and GM-CSF-80 ug/ml (Cytolab IL). On day seven floating cells were collected and loaded with 50ug/ml of examined peptide for 18h at 370C.
Development of ImMucin specific enriched T-CeII clone
Thawed PBLs underwent 1st stimulation with ImMucin pulsed Autologues DC at a ratio of 20:1 and cultured for 7 days in dedicated T cell Medium. T cell medium contained RPMI medium supplemented with 10% FCS, L-Glutamine, Sodium Pyruvate, MEM-EAGLE non-essential amino-acids, HEPES and Gentamycin _ _
Sulphate (Bet-Haemek Industries, IL) as well as with 50IU/ml of recombinant IL-7 (Cytolab IL). For the second stimulation, PBL were transferred to adherent autologues ImMucin- pulsed PBMC and cultured for additional 5 days in T cell Medium. At the end of the 5 days, medium was partial replaced with fresh medium containing lug/ml of ImMucin and 50ILVmI IL-2 for a third stimulation of 48 h at 370C. Following the last stimulation ImMucin T cell line was checked for immune activation properties.
ELISA for evaluation of cytokine secretion by stimulated T-cells
Cytokine release was studied during the culture of stimulated T-cells. The evaluated samples were collected from the T cell culture medium on day 2, 3, and 5 and stored until use at-20°C.
100 ml of the selected capture antibody (e.g. anti -Hu IFN-Gamma, or anti-Hu TNF- alfa or anti IL-2) were added to each well at the final concentration of 5μg/ml in PBS. Plates were incubated for 2h at R.T. or over night at 4°C. Next, 200 ml of blocking solution were added for an incubation of 2h at R.T. lOOμg of evaluated samples were added in duplicate into 2 wells for 2 hours at RT. After 2h of incubation at R.T plates were washed X4 times in washing solution. In the next step, a Biotin-conjugated detection antibody (relevant to coated antibody) was added at a final concentration of lOμg/ml in total volume of lOOμl for Ih at R.T. Finally, lOOμl/well working dilution (1: 10000 in Blocker solution) of Streptavidin- HRP were added to each well and incubated for Ih at R.T.
Plates were washed X6 times with PBS-Tween (0.4%) and lOOμl/well of TMB/E Solution (CHEMICON, Catalog #ES001) was added. The plates were then exposed until blue color appeared in the wells of the positive control and no color was detected in the blank wells. To stop the color reaction, 50μl/well of 10% of Sulfuric acid was added. The plates were counted in ELISA-Reader using a 450nm filter.
Proliferation of stimulated T-Iymphocytes
Proliferation analysis was conducted in three different methods. In all three methods, plates were cultured for 3-5 days under visual control and then 0.5 μCu/well of 3[H] (Amersham) was added for additional culture of to 18h at 370C. In the last step, plates were harvested and the radioactive counts were measured in a β counter. 1. DC indirect presentation: In the first method, lOOμl of 2x106 thawed PBL were placed in U-shape Bottom 96 well plate (Nunc) with RPMI medium supplemented with 5% H AB serum, L-Glutamine, Sodium Pyruvate, MEM-EAGLE nonessential amino-acids, HEPES and Gentamycin. At the next stage lOOμl of 25x104 peptide-pulsed DC were added to each three wells of PBL and underwent proliferation analysis as described above.
2. PBL direct presentation: In the 2nd method, lOOμl of 2x106 PBL were placed in Flat Bottom 96 well plates (Corning) and stimulated with different concentrations (0.05ug/ml-2ug/ml) of peptides. In the next step the cells underwent proliferation analysis as described above.
3. PBMC direct presentation: In third method PNMC were used under similar conditions as described for the second method.
In Vitro cytotoxic assay (CTL)
Effector cells were ImMucin specific enriched T-CeIl clone. Viable lymphocytes (effector cells), were centrifugation, resuspended in RPMI-HEPES medium, and admixed at different ratios with 5xlO3 35S methionine-labeled, tumor cells. CTL assays were performed in U-shaped microtiter wells, at 37 C, 5% CO2 for 5 hours. Cultures were terminated by centrifugation at 1000 rpm for 10 min at 4O0C. A total of 100 μl of the supernatants was mixed with scintillation fluid and measured in a β counter (Becton Dickinson Canberra Australia). Percentage of specific lysis was calculated as follows: % lysis = (cpm in experimental well -cpm spontaneous release)/(cpm maximal release - cpm spontaneous release) x 100. Spontaneous release was determined by incubation of 100 μl-labeled target cells with 100 μl of medium. Maximal release was determined by lysis of target cells in lOOμl 10% TrytonX-100.
FACS analysis of T-lymphocytes
For FACS analysis, PBLs were suspended at a final concentration of 20x10 and 50μl were transfer into FACS tube (Falcon) in Blocker solution (PBS with 3% FCS and 0.1% Sodium Azide). 10 μl of fluorochrome-conjugate anti-CD4, anti-CD8, and anti- CD45RO (eBioscience) were added for 30min on ice at O0C in the Dark. After the - -
incubation cell were washed with 2ml of the blocker solution and re suspended in 0.5ml of Phosphate buffer saline (PBS). Samples were analyzed in a FACS-sort machine (BD) for positive florescence.
RESULTS
SP TECHNOLOGY
Table 3 describes ImMucin (VLX-100) and other various VXL epitopes used in the experiments. The CTL epitopes VXLl (D6) and VXL2 (Ml.2) were used as positive controls for MUC l's SP and class I epitopes. As a control for MUC l's class I epitopes which is not derived from the SP domain, we have used the previously identified CTL epitopes VXL6 (M 1.1). We also used CTL epitopes derived from the SP domains of other non MUCl TAA like Her2/neu (VXL-8) or Tyrosinase (VXL- 11). As a positive control for class II epitope we used the universal pan-class II epitope peptide PADRE (VXL- 14).
Figure imgf000032_0001
Table 3: ImMucin and other target epitopes used in the experiments _ _
Proliferation properties indicated by Stimulation Index (SI)
In the first set of experiments, the 21mer ImMucin, its different 9mer epitopes and other control epitopes (Table 3) were evaluated in proliferation assays with the aim of finding their optimal mode of presentation, stimulated dose as well as their peak of activity (time wise).
To evaluate the most efficient method for priming of PBLs with the different peptides, the following methods were compared: a. In-direct presentation of the peptide via Autologues- pulsed DC b. Direct presentation of the peptides via PBMC i.e. monocytes c. Direct presentation of peptides to PBL without any APC Proliferation analysis is usually indicating the existence of specific T cells activation mainly CD4+ but could also be associated with CD8+ activation.
Five experiments were performed according to the protocol described in material and methods with buffy-coat enriched PBLs obtained from six different naive donors (obtained from the Israeli blood bank).
The most immunogenic antigen is the 21mer ImMucin which manifested a SI of 4 and 2 respectively for peptide presentation via DC and PBMC (see table 4). In general, SI of > 2 is considered to be a strong specific activation. The high SI of ImMucin obtained with naive PBLs which didn't have any prior priming by ImMucin in vivo, i.e. the first and only antigen-T cell interaction occurred in-vitro, is a positive indication of the high immunodominant properties of IrriMucin. There was no activation of PBL when ImMucin (or other epitopes) were presented directly to PBL without APC, suggesting that ImMucin's activity is not mediated via a non-specific mitogen-like mechanism of action but rather it needs a specific intracellular process for efficient presentation via a proper APC. Moreover, it is clear that presentation via professional APC i.e. Dendritic Cells is better than presentation via monocytes from PBMC since stimulation of PBMC with peptides, gave a weaker effect (Table 4).
Since the experiments were carried out on six different donors, each one of them having an individual repertoire of Class I and II alleles, the high SI of ImMucin suggests polyclonal T cell activation via binding to multi MHC epitopes. In other words, different epitopes are used in different donors. To further examine this issue, the index of stimulation for ImMucin's 9mer epitopes was analyzed. Like ImMucin, the class I and/or class II 9mer epitopes VXL-4, VXL-I and VXL-5 (see Table 4 and 3) manifested high stimulation index of SI>3. This activity is probably not restricted to a single class I or II allele, since these peptides do not share the same MHC binding properties as can be seen in the case of XVL-5 which does not bind the HLA- A2.1 allele like the epitopes VXL-4, VXL-I. Another ImMucin epitope, the 9mer VXL-2, manifested moderate SI scores of 2.2 although it was shown in the past to be a highly potent class I, HLA- A2.1 -restricted CTL epitope. It is possible that in this assay the activation of CD4+ rather than CD8+ was more dominant. Other SP derived epitopes like the Her2/Neu or Tyrosinase, and the MUCl non SP- epitope showed moderate activation of 1.5<SI<2.
Figure imgf000034_0001
Table 4: Proliferation analysis of PBL using ImMucin and other VXL target epitopes indicated by Index of stimulation (SI). SI is calculated by dividing the CPM obtained in an analyzed sample to the CPM obtained in a control sample. Results are representative of 5 similar experiments. Analyzing the time for maximal peak of PBL activation
Figure imgf000035_0001
Table 5: Analyzing the maximal peak of PBL activation (in a proliferation assay), following stimulated with ImMucin and other VXL-peptides. Observation was done daily during 5 days by visual inspection using light microscopy. Maximal peak of PBL activation is determined when <50% of the PBLs appear in clumps. Results are the average of 5 experiments.
A different parameter for assessing the properties of ImMucin and the other VXL- peptide epitopes is by analyzing the time until a maximal peak of activity occurs and the optimal dose for maximal stimulation. Maximal peak of PBL activation is determined when <50% of the PBLs appear in clumps. In these experiments, the kinetic (time) in the proliferation of lymphocytes from the six naive donors stimulated at a fix dose of 0.05- lug/ml by ImMucin and other VXL-Peptides, was observed.
A different profile for maximal peak of proliferation (i.e. stimulation property) and optimal dose for proliferation was found when comparing different peptide epitopes. _ _
Results in the five experiments show a more rapid peak of proliferation to VXL-4 and VXL-5 with a peak at 48-72 hours (Table 5) and lowest concentration needed for activation 0.2-0.05ug/ml (Table 6) while ImMucin and other SP-derived epitopes or non SP epitope gave slower peak of proliferation at the range of 96 hours and a slightly higher dose for stimulation 0.05-lug/ml (Table 6). Interestingly, the ImMucin CTL epitopes VXLl and VXL2 as well as the Her2/Neu CTL epitope manifested in these experiments peak for maximum stimulation at 120 hours with a slightly higher dose 0.05-lug/ml although their MHC binding affinity are as high as those of VXL-4 and VXL-5 (data is not presented). Nevertheless, when comparing the results in this set of experiments (Table 5, 6) to those obtained in the proliferation assay (Table 4), it appears that the rapid peak of activation of VXL-4 and VXL-5 does not always positively correlate with their high SI. It is therefore assumed that at least part of this phenomenon is associated with the unique properties of sequences within the signal peptides (SP). In particular, SP-associated sequences/epitopes were noted, e.g. the MUCl VXLl, 2, and 4 epitopes, but also other SP epitopes such as VXL 8 and VXLI l, which contain antigen specific properties for CD4+ and/or CD8+ activation and other sequences (such as VXL-4 and VXL-5) which in addition to CD4+ and/or CD8+ activation also have an "adjuvant like" activity. The adjuvant-like property of signal peptides was already shown in the past by attaching SP to other non-SP epitopes in order to increase their immunity (Sherritt et al., 2001). The relatively slower peak of activity of ImMucin and VXL-I, 2, 6, and 8 can potentially be associated with the time needed for them to enter the APC and move from the Class II compartment into the Class I compartment in a process known as "cross presentation" or "cross priming". It is known that exogenous antigens can gain entry into the so- called endogenous pathway using the cross-presentation mechanism which is known to be very effective for class I— restricted cytotoxic T lymphocyte (CTL) epitopes. - -
The effect of dose on PBL activation
Figure imgf000037_0001
Table 6: Analyzing the optimal dose of ImMucin and other VXL-peptides required for PBL activation (in a proliferation assay). Observation was done daily during 5 days by visual inspection using light microscopy. Results are the average of 5 experiments using six different donors.
ELISA Assay for studying cytokine release by stimulated PBL
One of the important parameters of activated T cells is their secretion of key cytokines like TNF- Alfa, IL-2 and IFN-gamma as an outcome of antigen stimulation. Secretion of a given cytokine is highly dependent on the type of secretory cell, CD4+ vs. CD8+ and on the stage of activation, in particular early vs. late activation. TNF- Alfa is a proinflamatory cytokine which is secreted at early stage of activation by both CD4+ and CD8+. Interleukin-2 is one of the key cytokines which enhance the proliferation of CD4+ T lymphocytes. It induces a secondary immune response of CD8+ cells and the development of memory CD8+ cells following primary activation with antigen. Therefore, IL-2 is usually associated with late secretion. IFN-gamma is produced mainly by CD8+ T cells following IL-2 secretion (late secretion) and is correlates with CD8+ specific activation and function.
Hence, the cytokine profile released by T-Lymphocytes stimulated once (Table 7) or several times (Figure 1) with ImMucin or other VXL 9mer epitopes was studied using an ELISA assay according to the protocol described in material and methods.
Figure imgf000038_0001
Table 7: ELISA quantitative assay for Cytokine secretion profiles of PBL stimulated once with ImMucin or other VXL 9mer epitopes DC at the ratio of 16:1. The results represent one out of five experiments using six different donors. SI of the peptides is also indicated.
As envisaged, a single stimulation resulted in the early secretion of TNF-Alfa, produced by both CD4+ and CD8+. The antigens which induced the highest cytokine release are the 9mers VXL4 and VXL5; the 21mer ImMucin also revealed a high level of TNF-Alfa secretion of 11.6ng/ml. Also, VXL-Il which didn't perform well in the proliferation analysis also secreted high amounts of TNF-Alfa. The results of ImMucin's cytokine secretion are directly correlated with the high SI of ImMucin, SI=4 (see table 7). IL-2 secretion of 3.62ng/ml was also observed. This secretion is probably performed by CD4+ T cells activated with the ImMucin vaccine, as this phenomenon is usually associated with multiple activations and with the induction of memory. Lastly, ImMucin was the only antigen which could induce the secretion of _ _
IFN-Gamma by CD8 T cells. The results showing IFN-Gamma and IL- 2 secretion emphasize the "super activation" properties of ImMucin which can induce a polyclonal activation via a combination of multiple epitopes for both CD4+ and CD8+ on one sequence. This property seems to compensate in this experiment for the lack of repeated activation. Additional support for this assumption is provided by the high level (ng/ml) of cytokine release from the naive T cells, which are usually if at all, at the range of pg/ml (i.e. three folds lower). Other ImMucin SP epitopes like VXL-I, VXL-4 and VXL-5 as well as other SP epitopes like VXL-11 showed also high TNF- Alfa release which mostly correlates with the high SI of these epitopes. The high IL- 2 secretion by VXL-5 together with the other results obtained for this epitope suggest that it may work as a CDA+ epitope which may be responsible for some of the IL-2 secretion induced by ImMucin.
Cloning of specific T-cells
In the first set of experiments the immunogenic properties of ImMucin and its VXL epitopes were demonstrated with respect to CD4+ and CD8+ T cells. In the following set of experiments the ability of ImMucin to undergo a process of cross priming is demonstrated. During this process, the 21mer is entering the APC via the class II compartment, processed into the class I compartment and finally presents its different VXL 9mer epitopes, namely 1, 2, 4 and 5 by different MHC class I and/or II molecules. Also demonstrated is the ability of ImMucin specific clones to lyse MUCl expressing tumor cells.
For that purpose, an enriched ImMucin-specific T cell subpopulation was produced via repeated stimulation with ImMucin. The specificity of the subpopulation for ImMucin, as well as to the other VXL epitopes, was examined using proliferation assays, cytokine release assays, FACS analysis in which the percentage and type of cells enriched during the enrichment process was analyzed, and using a CTL assay against selected target cells.
For producing the specific T cell subpopulation PBL were stimulated three times for 7, 5 and 2 days with ImMucin presented via DC and PBMC. Following the 3rd stimulation the cells were evaluated against the different VXL epitopes. _ _
Analyzing the levels of cytokine release during the development process of the T cell clone revealed, as expected, a high peak of TNF-Alfa secretion (5ng/ml) during the first stimulation with ImMucin (Fig 1). Following a second stimulation with ImMucin, the levels of TNF-Alfa secretion decreased and TNF-Alfa secretion completely disappeared after the 3rd stimulation on day 14. On the contrary, the secretion levels of IL-2 from CD4+ T cells and INF-gamma mainly from CD8+ T cells have increased dramatically after the second and third stimulations with ImMucin suggesting an enrichment of CD4+ and CD8+ cells with potentially higher specificity as killer cells (high IFN-gamma secretion) with memory (IL-2 secretion). This unique and novel dual specificity of CD4+ and CD8+ subpopulations to ImMucin, may be explained by the nature of this peptide vaccine harboring both class I and II immunodominant epitopes. Similar to the results obtained in the initial experiment (Table 7) also in this experiment the levels of secretion are high at the range of ng/ml.
The ability of ImMucin to propagate a subpopulation of CD4+ and CD8+ T cells was further analyzed, and in particular the ability of ImMucin to transform naive CD4+ and CD8+ T cells into memory cells. This property was evaluated in a FACS analysis measuring the CD45RO+ marker for memory T cells on CD4+ and CD8+ subpopulations (Fig 2).
As can be seen in this particular experiment, repeated stimulation with ImMucin transformed PBLs into CD4+ and CD8+ T cells. In both cases, a two fold increase, from 43% to 85% in the case of CD4+ and from 15% to 36% in the case of CD8+ T cells. These numbers represent the normal CD4+ CD8+ percentage ratio and suggest a dual activation of both T cell subpopulations. ImMucin activation also increases by two fold the percentage of memory CD45RO+ cells which are the cells that were effectively primed with ImMucin and will remain as memory cells. It is important to note that although the percentage of memory cells did not increase after the first stimulation (Fig 2) their total numbers do increase (data is not presented).
Next, the ability of ImMucin to undergo cross priming and present its different epitopes for inducing T cell proliferation was explored. As can be seen in Table 8 each one of the MUCl SP epitopes VXLl, VXL2, VXL4 and VXL5 induced a specific proliferation with an average IS ranging from 2.66 to 3.5 which was comparable to the SI achieved using ImMucin itself. VXL-Il, the SP epitope which is not deduced from the MUCl antigen and VXL-6 which is a MUCl epitope which is not deduced form the SP domain manifested lower SI with an average SI of 1.14-1.125 or 2-5 times lower than the MUCl SP epitopes. Some background was expected for VXL6 and VXL-I l as they do harbor epitopes from other SP-sequences or from MUCl sequences.
The cytokine profile obtained from the specific T cell subpopulation stimulated with the MUCl SP epitopes VXLl, VXL2, VXL4 and VXL5 conform to the proliferation results, showing again that antigen specificity is positively correlated with cytokine release (Table 9). ImMucin and its SP epitopes VXLl, VXL2, and VXL4 manifested high IL-2 and IFN-gamma secretion compared to low to moderate levels induced by the other epitopes. The secretion of TNF- Alfa which is less specific and appears in an early stage is high in all the epitopes tested. The positive results achieved in this experiment are unique compared with results obtained with class I peptide vaccines since in this experiment ImMucin was not matched with the relevant HLA alleles of the donors and still received a positive proliferation.
Figure imgf000041_0001
Table 8: Proliferation of a specific T-cell subpopulation which underwent three stimulations with ImMucin or different VXL-peptides as assessed in a proliferation assay. Results represent one out of two experiments using four donors. - -
Figure imgf000042_0001
Table 9: ELISA quantitative assay for Cytokine secretion profiles of T-CeIl specific subpopulation (Clones) stimulated with ImMucin and various VXL-Peptides. The results represented summary of two experiments using four donors.
In order to further evaluate the class II / class I cross presentation as well as the specificity and function of the T-cell clones, their class I restricted lysis potential was analyzed. For this assay the clones were screened for Class I HLA-A2.1 expression and their potential to lyse MUCl and HLA- A2.1 positive and negative cell-lines was evaluated. Based on this evaluation two clones which were found to express the HLA- A2.1 allele was selected. The results summarized in Table 10 show a MUCl as well as HLA- A2.1 -restricted lysis. A breast tumor cell such as MDA-MB-231 which expresses both MUCl and HLA- A2.1 was lysed effectively (23%), while another breast tumor cell line MDA-MB-468 which expresses only MUCl but not the HLA- 2.1 was not lysed al all. Other control cell-lines didn't manifest any lysis (see table 10).
These results demonstrate several important properties of ImMucin which are important for an effective vaccine:
1. Cross presentation of ImMucin by host APC in the correct manner. Processing, expression, and presentation of ImMucin's epitopes by APC to lymphocytes.
2. The processing, expression, and presentation of ImMucin's epitopes on MUCl positive tumor cells. - -
3. The immunodominance of ImMucin as an effective inducers of CTL activation via CD8+.
In summary, out of 14 different blood samples derived from naive donors, in 12 ImMucin specific activation could be observed. It is therefore clear that ImMucin is able to induce antigen specific CD4+ as well as CD8+ T cell activation which can lead to effective anti-tumor vaccine properties including lysis, cytokine release and memory in the majority of the population.
Figure imgf000043_0001
Table 10: CTL assay of HLA-A2.1 positive T-CeIl specific sub-population (Clone) stimulated with ImMucin. MUCl expression (+ represents low, +++ represents high) was measured by the H23 anti-MUCl mAb. HLA- A21 expression was measured using the BB7.2 mAb. The results represent two separate experiments using two different donors.
The immunodominance of other SP vaccine
Immunogenic properties of SP-derived sequences were observed in all the experiments conducted using MUCl and non-MUCl derived peptides. Part of the immunity is antigen specific while part of it is more SP specific. These observations were supported by the in-silico algorithm which predicted the existence of immunodominant SP epitopes derived from novel as well as known TAA which manifested potential binding to both Class I and Class II epitope in over 50% of the population (cross HLA).
In order to verify the in-silico predictions, two additional tumor associated SP vaccines were synthesized. The first one VXL102 (SEQ ID NO. 7) is derived from the known melanoma associated TAA BAGE while the second VXLlOl (SEQ ID NO 1) is derived from a novel protein, ARMET which is known to be overexpressed in certain tumors. The immunodominant properties of these two peptides and in particular their ability to stimulate T cells in a proliferation assay, were examined.
PBL from two healthy donors were stimulated with VXL-102 or VXL-101 peptides. As a positive control for class II activation the universal pan-class II epitope peptide PADRE (VXL- 14) was used. As can be seen ImMucin and Armet manifested the highest SI in this experiment which was equivalent to the SI obtained with the universal PAN DR epitope VXL- 14 (Table 11). Since the later is considered to be a very immunogenic peptide which is added to many vaccines for PAN CD4+ T cell activation, the comparable SI obtained in the experiment is an important finding which stresses the high immunodominant properties of both ImMucin and ARMET. In this experiment VXL-102 manifested lower SI than VXL-101 and ImMucin but still higher than the non-SP epitope VLX-6. A similar pattern of results was obtained in ELISA assay analyzing the cytokine profile of the peptide-activated T cells (Table 12). While all peptides induced high secretion levels of TNF- Alfa (<10ng/ml), only ImMucin induced IL-2 and VXL-101 induced INF-Gamma suggesting that enrichment of CD4+ and CD8+ T cells occurred. As in the proliferation experiment VXL-102 didn't induced IL-2 or IFN-gamma production. This moderate immunogenic profile could be associated with the HLA match of the two donors used in this experiment.
Figure imgf000044_0001
Table 11: Index of stimulation for PBL by VXL-Peptides. Results represent an average of two donors. - -
Figure imgf000045_0001
Table 12: ELISA quantitative assay for Cytokine secretion profiles of PBL stimulated once with ImMucin and two additional tumor associated SP vaccinesVXL-101 and VXL- 102 The results represent stimulation with peptide pulsed DC in ratio 16:1 one out of two experiments using two different donors.
REFERENCES
Boel P. et al Immunity, 2 (2): 167-175 (1995)
Brossart P, Heinrich KS, Stuhler G, Behnke L, Reichardt VL, Stevanovic S, Muhm A, Rammensee HG, Kanz L, Brugger W. Identification of HLA-A2- restricted T-cell epitopes derived from the MUCl tumor antigen for broadly applicable vaccine therapies. Blood. 93(12): 4309-17 (1999).
Cannon L, El-Shami KM, Paz A, Pascolo S, Tzehoval E, Tirosh B, Koren R, Feldman M, Fridkin M, Lemonnier FA, Eisenbach L. Novel breast-tumor- associated muci-derived peptides: characterization in D ~ xB2 microglobulin (B2m) null mice transgenic for a chimeric HLA-A2.1/Db-β2 microglobulin single chain. Int. J. Cancer. 85,391-397 (2000).
Chaux, P., V. Vantomme, V. Stroobant, K. Thielemans, J. Corthals, R. Luiten, A. M. Eggermont, T. Boon, and P. Van der Bruggen. Identification of MAGE- 3 epitopes presented by HLA-DR molecules to CD4(+) T lymphocytes. J. Exp. Med. 189, 767-78 (1999).
Christopher E, Touloukian, Wolfgang W. Leitner, Suzanne L. Topalian, Yong F. Li, Paul F. Robbins, Steven A. Rosenberg and Nicholas P. Restifo. Identification of a MHC Class II-Restricted Human gplOO Epitope Using DR4-IE Transgenic Mice. The Journal of Immunology., 164, 3535-3542 (2000).
Dayhof, M. D., Atlas of protein sequence and structure, Nat. Biomed. Res. Found. 5, suppl. 3 T1978)
Fisk, B., Blevins, T.L., Tylor-Wharton, J. and Ionnides, C. G., Identification of an immunodominant peptide of HER2/neu protooncogene recognized by ovarian tumor specific cytotoxic T-lymphocyte lines. J exp. Med. 181, 2109- 2117 (1995).
Fung, P. Y. and Longenecker, B.M., Specific immunosuppressive activity of epiglycanin, a mucin-like glycoprotein secreted by a murine mammary adenocarcinoma (TA3-HA). Cancer Res. 51, 1170-1176 (1991).
Gilboa Eli., The promise of cancer vaccines Nature Reviews 4, 401-411 (2004).
Graham, R.A., Burchell, J.M. and Taylor-Papadimitriou, J., The polymorphic epithelial mucin: potential as an immunogen for a cancer vaccine. Cancer Immunol. Immunother. 42, 71-80 (1996).
Grulich et al., Lancet 370, 59 67 (2007)
Green et al., Cell 28:477, 1982
He X, Tsang TC, Luo P, Zhang T, Harris DT. Enhanced tumor immunogenicity through coupling cytokine expression with antigen presentation. Cancer Gene Ther. 10(9), 669-77 (2003).
Ho, S.B., Niehans, G.A., Lyftogt, C, Yan, P.S., Cherwitz, D.L., Gum, E.T., Dahiya, R. and Kim, Y.S., Heterogeneity of mucin gene expression in normal and neoplastic tissues. Cancer Res. 53, 641-651 (1993).
Hung K, Hayashi R, Lafond- Walker A, Lowenstein C, Pardoll D, Levitsky H. The central role of CD4(+) T cells in the antitumor immune response. J. Exp. Med. 188, 2357-2368 (1998).
Jaeger E, Bernhard H, Romero P, Ringhoffer M, Arand M, Karbach J, Ilsemann C, Hagedorn M, Knuth A. Generation of cytotoxic T-cell responses with synthetic melanoma-associated peptides in vivo: implications for tumor vaccines with melanoma-associated antigens. Int. J. Cancer. 66, 162-169 (1996).
Kast, W.M., Brandt, R.M.P., Sidney, J., Drijfhout, J. W., Kubo, R.T., Melief, C.J.M. and Sette, A. Role of HLA-A motifs in identification of potential CTL epitopes in human papillomavirus type 16 E6 and E7 proteins. J Immunol 152: 3904-3912 (1994)
Kedar, E. and Klein, E. Cancer Immunotherapy: Are the results discouraging? Can they be improved? Adv Immunol 58:245-322 (1995).
Knutson KL, Schiffman K, Disis ML. Immunization with a HER-2/neu helper peptide vaccine generates HER-2/neu CD8 T-cell immunity in cancer patients. J Clin Invest. 107(4): 477-84 (2001)
Lenstra et al., Arch. Virol., 110, 1-24 (1990)
Lipman and Pearson, Science 227, 1435-1441 (1985).
Mandelboim, O., G. Berke, M. Fridkin, M. Feldman, M. Eisenstein, and L. Eisenbach. CTL induction by a tumor associated antigen octapeptide derived from a murine lung carcinoma Nature. 369, 67-71 (1994).
Mandelboim, O., E. Vadai, M. Fridkin, A. Katz-Hillel, M. Feldman, G. Berke, and L. Eisenbach. Regression of established murine carcinoma metastases following vaccination with tumor-associated antigen peptides Nat. Med. 11, 1179-83 (1995).
Manici, S., T. Sturniolo, M. A.Imro, J. Hammer, F. Sinigaglia, C. Noppen, G. Spagnoli, B. Mazzi, M. Bellone, P. Dellabona, and M. P. Protti. Melanoma cells present a MAGE-3 epitope to CD4(+) cytotoxic T cells in association with histocompatibility leukocyte antigen DRI l. J. Exp. Med. 189, 871-6 (1999). Marchand, M., Weynants, P., Rankin, E., Arienti, F., Belli, F., Parmiani, G., Cascinelli, N., Bourland, A., Wanwijck, R., Humblet, Y., Canon, J.L., Naeyaert, J.M., Plagne, R., Deraemaeker, R., Knuth, A., Jager, E., Brasseur, F., Herman, J., Coulie, P.G. and Boon, T. Int. J. Cancer 63:883-885 (1995)
McGuckin, M. A., M. D. Walsh, B. G. Hohn, B. G. Ward, and R. G. Wright. Prognostic significance of Muc-1 epithelial mucin expression in breast cancer. Hum Pathol 26, 432-439 (1995).
Minev BR, Chavez FL, Dudouet BM and Mitchell MS. Synthetic insertion signal sequences enhance MHC class I presentation of a peptide from the melanoma antigen MART-I. Eur J Immunol. (8):2115-24. (2000)
Morein et al., Nature 308:457 (1984)
Pardoll, D. M., and S. L. Topalian. The role of CD4+ T cell responses in antitumor immunity. Curr. Opin. Immunol. 10, 588-94 (1998).
Rammensee, H.-G., FaIk, K. and Rotzschke, O. Peptides naturally presented by MHC class I molecules. Annu Rev Immunol 11 :213-244 (1993)
Ras, E., van der Burg, S.H., Zegveld, S.T., Brandt, R.M., Kuppen, P.J., Offringa, R., Warnarr, S.O., van de Velde, CJ. and Melief, C.J., Identification of potential HLA-A *0201 restricted CTL epitopes derived from the epithelial cell adhesion molecule (Ep-CAM) and the carcinoembryonic antigen (CEA). Hum. Immunol. 53, 81-89 (1997).
Rhodes DR and Chinnaiyan AM. Integrative analysis of the cancer transcriptome. Nature genetics. 37 Suppl: S31-7 (2005)
Rosenberg SA, Yang JC, Schwartzentruber DJ, Hwu P, Marincola FM, Topalian SL, Restifo NP, Dudley ME, Schwarz SL, Spiess PJ, Wunderlich JR, Parkhurst MR, Kawakami Y, Seipp CA, Einhorn JH, White DE. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat. Med. 4,321-327 (1998).
Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 10 (9):909-15 (2004)
Sherritt M, Cooper L, Moss DJ, Kienzle N, Altaian J and Khanna R. Immunization with tumor-associated epitopes fused to an endoplasmic reticulum translocation signal sequence affords protection against tumors with down-regulated expression of MHC and peptide transporters. Int. Immunol. (3):265-71 (2001)
Shridhar R. et al., Cancer Res. 56 (24) 5576-5578
Townsend, A. and Bodmer, H. Antigen recognition by class I-restricted T lymphocytes. Annu Rev Immunol 7: 601-624 (1989)
Treon SP, Maimonis P, Bua D, Young G, Raje N, Mollick J, Chauhan D, Tai YT, Hideshima T, Shima Y, Hilgers J, von Mensdorff-Pouilly S, Belch AR, Pilarski LM, Anderson KC. Elevated soluble MUCl levels and decreased anti-MUCl antibody levels in patients with multiple myeloma. Blood 96;3147-53 (2000)
Weber JS, Hua FL, Spears L, Marty V, Kuniyoshi C, Celis E. A phase I trial of an HLA-Al restricted MAGE-3 epitope peptide with incomplete Freund's adjuvant in patients with resected high-risk melanoma. J. Immunotherapy. 22:431-440 (1999).
Wu et al., J. Immunol. 148:1519, (1992)

Claims

1. A peptide vaccine comprising a signal peptide domain of a protein wherein said signal peptide domain comprises multiple MHC Class I and multiple MHC Class II epitopes which are recognizable by the majority of the MHC alleles of a vaccinated human population.
2. A peptide vaccine according to claim 1, wherein said protein is a Tumor Associated Antigen (TAA) or a protein which is over-expressed in tumor cells.
3. A peptide vaccine according to claim 1 or claim 2 wherein said peptide vaccine comprises up to about 50 amino acids.
4. A peptide vaccine according to claim 3 wherein said peptide vaccine comprises up to 25 amino acids.
5. A peptide vaccine according to claim 1 wherein said protein is selected from the group consisting of Armet, HSP60, CANX, MTHFD2, FAP, MMP6, BAGE-I, GNTV, Q5H943, MUCl, CEA, Pmel, Kallikrein-4, Mammaglobin- 1, MART-I, GPR143-OA1, PSA, TRPl, Tyrosinase, FGF-5, NEU proto- oncogene, Aft, MMP-2, PSMA, Telomerase-associated protein-2, PAP, Uroplakin II and Proteinase 3.
6. A peptide vaccine according to claim 1, wherein said signal peptide domain comprises a sequence selected from the group consisting of SEQ ID Nos. 1- 28.
7. A peptide vaccine comprising a signal peptide domain of MUC 1.
8. A peptide vaccine according to claim 7, wherein said signal peptide domain comprises the amino acid sequence MTPGTQSPFFLLLLLTVLTVV (SEQ ID No. 10).
9. A peptide vaccine comprising a signal peptide domain of BAGE- 1.
10. A peptide vaccine according to claim 9, wherein said signal peptide domain comprises the amino acid sequence MAARAVFLALSAQLLQA (SEQ ID NO. 7).
11. A peptide vaccine comprising a signal peptide domain of Armet.
12. A peptide vaccine according to claim 11, wherein said signal peptide domain comprises the amino acid sequence MWATQGLAV ALALSVLPGSRA (SEQ ID NO. 1). ^
13. An isolated nucleic acid molecule comprising a nucleotide sequence encoding a peptide according to any of claims 1-12.
14. A nucleic acid molecule according to claim 13, wherein said nucleic acid molecule is contained in an expression vector.
15. A nucleic acid vaccine comprising a nucleic acid molecule according to claim 13 or 14.
16. An isolated antigen-presenting cell preloaded with the peptide according to any of claims 1-12.
17. A pharmaceutical composition comprising the peptide vaccine according to any of claims 1-12 or the nucleic acid vaccine according to claim 15, or the antigen presenting cell according to claim 16, and a pharmaceutically acceptable carrier or diluent.
18. A pharmaceutical composition according to claim 17 for treating or inhibiting cancer.
19. A pharmaceutical composition according to claim 18 adapted for coadministration with other anti neo-plastic agents.
20. A pharmaceutical composition according to claim 7 for treating or inhibiting MUCl expressing cancers.
21. A pharmaceutical composition according to claim 20 wherein said MUCl expressing cancers are selected from the group consisting of colon cancer, gastric cancer, lung cancer, renal cell (RC) cancer, transitional cell (TC) cancer, prostate cancer, pancreatic cancer, breast cancer, ovary cancer, thyroid cancer, lymphoma, leukemia and multiple myeloma (MM).
22. A pharmaceutical composition according to claim 9 for treating or inhibiting BAGE-I expressing cancers.
23. A pharmaceutical composition according to claim 22 wherein said BAGE-I expressing cancers are selected from the group consisting of melanoma, soft tissue sarcoma (STS), non-small cell lung cancer, head and neck cancer, bladder cancer and breast carcinoma.
24. A pharmaceutical composition according to claim 1 1 for treating or inhibiting Armet expressing cancers.
25. A pharmaceutical composition according to claim 24 wherein said Armet expressing cancers are selected from the group consisting of renal cell _ _
carcinomas, lung cancer, breast cancer, prostate cancer, squamous cell carcinoma, head and neck carcinoma and pancreatic carcinoma.
26. Use of the peptide vaccines according to any of claims 1-12, or the nucleic acid vaccine according to claim 15, or the antigen presenting cell according to claim 16 for treating or inhibiting cancer.
27. Use according to claim 26, in combination with other anti neoplastic agents.
28. Use of the peptide vaccines according to any of claims 1-12, or the nucleic acid vaccine according to claim 15, or the antigen presenting cell according to claim 16 in the preparation of a pharmaceutical composition for treating or inhibiting cancer.
29. A method of treating or inhibiting cancer comprising administering the pharmaceutical compositions according to any of claims 17-25 to a cancer patient in need thereof.
30. Use of the peptide vaccines according to any of claims 1-12 for enriching a T cell population in vitro; whereby an enriched T cell population responsive to the peptide vaccine is obtained.
31. A method of treating or inhibiting cancer comprising administering the enriched T cell population according to claim 30 to a cancer patient in need thereof.
32. An isolated peptide selected from the group consisting of SEQ ID NO: 31-39.
33. A nucleic acid encoding for said isolated peptide according to claim 32.
34. A nucleic acid molecule according to claim 33, wherein said nucleic acid molecule is contained in an expression vector.
35. A pharmaceutical composition comprising the isolated peptide of claim 32, or the nucleic acid molecule of claim 33 or 34 and a pharmaceutically acceptable carrier or diluent.
36. A pharmaceutical composition comprising a mixture of at least 2 peptides selected from the group consisting of SEQ ID NO: 29-39, and a pharmaceutically acceptable carrier or diluent, whereby said mixture of peptides comprises MHC class I and MHC class II epitopes.
37. A pharmaceutical composition according to claim 36 comprising a mixture of at least 4 peptides selected from the group consisting of SEQ ID NO: 29-39.
38. A pharmaceutical composition according to claim 37 comprising SEQ ID NO: 29-32.
PCT/IL2007/001168 2006-09-21 2007-09-23 Antigen specific multi epitope vaccines WO2008035350A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US12/442,495 US9487574B2 (en) 2006-09-21 2007-09-23 Antigen specific multi epitope vaccines
AU2007298494A AU2007298494B2 (en) 2006-09-21 2007-09-23 Antigen specific multi epitope vaccines
CA2665816A CA2665816C (en) 2006-09-21 2007-09-23 Antigen specific multi epitope vaccines
EP07827143.4A EP2089423B1 (en) 2006-09-21 2007-09-23 Antigen specific multi epitope vaccines
IL197737A IL197737A (en) 2006-09-21 2009-03-22 Peptide vaccine consisting of a signal peptide, pharmaceutical compositions comprising it and uses thereof
US15/345,036 US11179452B2 (en) 2006-09-21 2016-11-07 Antigen specific multi epitope vaccines
US17/531,753 US20220072113A1 (en) 2006-09-21 2021-11-21 Antigen specific multi epitope vaccines

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US84608706P 2006-09-21 2006-09-21
US60/846,087 2006-09-21

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/442,495 A-371-Of-International US9487574B2 (en) 2006-09-21 2007-09-23 Antigen specific multi epitope vaccines
US15/345,036 Continuation US11179452B2 (en) 2006-09-21 2016-11-07 Antigen specific multi epitope vaccines

Publications (1)

Publication Number Publication Date
WO2008035350A1 true WO2008035350A1 (en) 2008-03-27

Family

ID=38913206

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2007/001168 WO2008035350A1 (en) 2006-09-21 2007-09-23 Antigen specific multi epitope vaccines

Country Status (5)

Country Link
US (3) US9487574B2 (en)
EP (1) EP2089423B1 (en)
AU (1) AU2007298494B2 (en)
CA (1) CA2665816C (en)
WO (1) WO2008035350A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011007359A2 (en) 2009-07-16 2011-01-20 Vaxil Biotherapeutics Ltd. Antigen specific multi epitope -based anti-infective vaccines
WO2011009173A1 (en) * 2009-07-23 2011-01-27 Mater Medical Research Institute Cancer immunotherapy
US20110195045A1 (en) * 2010-01-12 2011-08-11 Emmanuelle Godefroy Methods, agents and peptides for inducing an immune response to matrix metalloproteinase-2 expressing tumors
JP2012504563A (en) * 2008-10-01 2012-02-23 イマティクス バイオテクノロジーズ ゲーエムベーハー Tumor-related peptide compositions and related anti-cancer vaccines for the treatment of glioblastoma (GBM) and other cancers
WO2013006050A1 (en) * 2011-07-06 2013-01-10 Isa Pharmaceuticals B.V. Peptides inducing or enhancing an immune response against prostate-specific membrane protein (PSMA)
WO2013038412A2 (en) 2011-09-15 2013-03-21 Vaxil Bio Therapeutics Ltd. Antibodies directed against signal peptides, methods and uses thereof
CN103570821A (en) * 2012-07-27 2014-02-12 北京智飞绿竹生物制药有限公司 Mucin-1 antigenic polypeptide and application thereof as tumor vaccine
CN103570818A (en) * 2012-07-27 2014-02-12 北京智飞绿竹生物制药有限公司 Tumor antigenic polypeptide and application thereof as tumor vaccine
WO2015014820A1 (en) * 2013-07-29 2015-02-05 Eberhard Karls Universitaet Tuebingen Medizinische Fakultaet Prostate cancer immunotherapy
CN104387453A (en) * 2014-12-08 2015-03-04 深圳市同康生物医药有限公司 Dendritic cell targeted peptide, coding gene and application
EP2662389A4 (en) * 2011-01-07 2015-08-12 Toagosei Co Ltd Method for preparing antigen to obtain antihydrophobic peptide antibody
US9370182B2 (en) 2012-05-28 2016-06-21 Toagosei Co., Ltd. Antimicrobial peptide and use thereof
US9480727B2 (en) 2012-10-18 2016-11-01 Toagosei Co. Ltd. Synthetic peptide for inhibiting expression of type 2 TNF receptor and use thereof
JP2017158561A (en) * 2012-07-20 2017-09-14 ジェムバックス アンド カエル カンパニー,リミティド Peptides having anti-inflammatory activity and compositions comprising the same
US10568948B2 (en) 2015-05-13 2020-02-25 Agenus Inc. Vaccines for treatment and prevention of cancer
WO2020048990A1 (en) * 2018-09-04 2020-03-12 Treos Bio Zrt Peptide vaccines
WO2020243787A1 (en) * 2019-06-06 2020-12-10 The University Of Sydney Anti-inflammatory agents
US11065317B2 (en) 2018-04-26 2021-07-20 Agenus Inc. Heat shock protein-binding peptide compositions and methods of use thereof
US11213578B2 (en) 2017-03-03 2022-01-04 Treos Bio Limited Vaccine

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070202186A1 (en) 2006-02-22 2007-08-30 Iscience Interventional Corporation Apparatus and formulations for suprachoroidal drug delivery
US9238796B2 (en) 2010-06-04 2016-01-19 Toagosei Co. Ltd. Cell growth-promoting peptide and use thereof
US20170157243A1 (en) * 2014-06-24 2017-06-08 University Of Massachusetts MANF as a Regulator of Immune System Function
WO2017040309A1 (en) * 2015-08-28 2017-03-09 The Medical College Of Wisconsin, Inc. Peptide inhibitors of telomerase translocation and therapeutic uses thereof
EP3369431A1 (en) * 2017-03-03 2018-09-05 Treos Bio Kft Vaccine
US11844824B2 (en) * 2018-03-06 2023-12-19 Pepvax, Inc. Nucleic acid molecules and methods of using the same
GB201814361D0 (en) * 2018-09-04 2018-10-17 Treos Bio Zrt Immunogenetic cancer screening test
CN111138522B (en) * 2018-11-06 2023-01-24 香雪生命科学技术(广东)有限公司 Tumor antigen short peptides derived from AFP
CN111138521B (en) * 2018-11-06 2022-10-28 香雪生命科学技术(广东)有限公司 Short peptides derived from AFP antigen
GB202004974D0 (en) 2020-04-03 2020-05-20 Treos Bio Ltd Coronavirus vaccine
CN111875712A (en) * 2020-07-31 2020-11-03 广东昭泰体内生物医药科技有限公司 Enhanced MUC 1-targeted chimeric antigen receptor and application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995000159A1 (en) * 1993-06-17 1995-01-05 Ludwig Institute For Cancer Research Isolated peptides which form complexes with mhc molecule hla-c-clone 10 and uses thereof
WO2000006723A1 (en) * 1998-07-30 2000-02-10 Yeda Research And Development Company Ltd At The Weizmann Institute Of Science Tumor associated antigen peptides and use of same as anti-tumor vaccines
WO2001018035A2 (en) * 1999-09-08 2001-03-15 Transgene S.A. Muc-1 derived peptides
WO2005025612A1 (en) * 2003-09-15 2005-03-24 Glaxo Group Limited Vaccines

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US6969609B1 (en) * 1998-12-09 2005-11-29 The United States Of America As Represented By The Department Of Health And Human Serivces Recombinant vector expressing multiple costimulatory molecules and uses thereof
DE19917195B4 (en) 1999-04-16 2006-09-28 Immatics Biotechnologies Gmbh Peptide for triggering an immune reaction against tumor cells, pharmaceutical compositions containing them, their uses, nucleic acid coding therefor and expression vector containing said nucleic acid
EP2248910A1 (en) * 2000-04-28 2010-11-10 Mannkind Corporation Epitope synchronization in antigen presenting cells
AUPQ776100A0 (en) * 2000-05-26 2000-06-15 Australian National University, The Synthetic molecules and uses therefor
CN1691964A (en) 2002-09-06 2005-11-02 曼康公司 Epitope sequences
EP1660535A2 (en) 2002-11-27 2006-05-31 Minerva Biotechnologies Corporation Techniques and compositions for the diagnosis and treatment of cancer (muc1)
EP1609107A4 (en) * 2003-03-28 2006-08-30 Idm Pharma Inc Methods of identifying optimal variants of peptide epitopes
EP1697399B1 (en) * 2003-12-12 2016-11-23 GOVERNMENT OF THE UNITED STATES OF AMERICA, as repr. by THE SECR. OF THE DEPT. OF HEALTH AND HUMAN SERVICES AND HIS SUCCESSORS A human cytotoxic t-lymphocyte epitope and its agonist epitope from the non-variable number of tandem repeat sequence of muc-1
EP1848804B1 (en) * 2005-01-28 2011-10-05 Ramot at Tel Aviv University, Ltd. Anti-muc1 alpha beta antibodies
US8859495B2 (en) * 2005-03-30 2014-10-14 Minerva Biotechnologies Corporation Methods for stimulating or enhancing proliferation of non-tumorous cells expressing MUC1 receptors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995000159A1 (en) * 1993-06-17 1995-01-05 Ludwig Institute For Cancer Research Isolated peptides which form complexes with mhc molecule hla-c-clone 10 and uses thereof
WO2000006723A1 (en) * 1998-07-30 2000-02-10 Yeda Research And Development Company Ltd At The Weizmann Institute Of Science Tumor associated antigen peptides and use of same as anti-tumor vaccines
WO2001018035A2 (en) * 1999-09-08 2001-03-15 Transgene S.A. Muc-1 derived peptides
WO2005025612A1 (en) * 2003-09-15 2005-03-24 Glaxo Group Limited Vaccines

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BROSSART P ET AL: "Identification of HLA-A2-restricted T-cell epitopes derived from the MUC1 tumor antigen for broadly applicable vaccine therapies", BLOOD, W.B.SAUNDERS COMPANY, ORLANDO, FL, US, vol. 93, no. 12, 15 June 1999 (1999-06-15), pages 4309 - 4317, XP002147432, ISSN: 0006-4971 *
PINCHUK IRINA ET AL: "A CD8(+) T cell heptaepitope minigene vaccine induces protective immunity against Chlamydia pneumoniae", JOURNAL OF IMMUNOLOGY, vol. 174, no. 9, May 2005 (2005-05-01), pages 5729 - 5739, XP002464681, ISSN: 0022-1767 *
VON HEIJNE GUNNAR: "Signals for protein targeting into and across membranes", SUBCELLULAR BIOCHEMISTRY, XX, XX, 1994, pages 1 - 19, XP009094493 *
WIERECKY JAN ET AL: "Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients", CANCER RESEARCH, vol. 66, no. 11, June 2006 (2006-06-01), pages 5910 - 5918, XP002464680, ISSN: 0008-5472 *

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2012504563A (en) * 2008-10-01 2012-02-23 イマティクス バイオテクノロジーズ ゲーエムベーハー Tumor-related peptide compositions and related anti-cancer vaccines for the treatment of glioblastoma (GBM) and other cancers
US11135277B2 (en) 2009-07-16 2021-10-05 Vaxil Biotherapeutics Ltd. Antigen specific multi epitope-based anti-infective vaccines
US10245309B2 (en) 2009-07-16 2019-04-02 Lior Carmon Antigen specific multi epitope-based anti-infective vaccines
WO2011007359A2 (en) 2009-07-16 2011-01-20 Vaxil Biotherapeutics Ltd. Antigen specific multi epitope -based anti-infective vaccines
US10350284B1 (en) 2009-07-16 2019-07-16 Lior Carmon Antigen specific multi epitope-based anti-infective vaccines
US20120177677A1 (en) * 2009-07-16 2012-07-12 Vaxil Biotherapeutics Ltd. Antigen specific multi epitope-based anti-infective vaccines
WO2011007359A3 (en) * 2009-07-16 2011-07-07 Vaxil Biotherapeutics Ltd. Antigen specific multi epitope -based anti-infective vaccines
EP2453914B1 (en) * 2009-07-16 2018-09-05 Vaxil Biotherapeutics Ltd. Antigen specific multi epitope -based anti-infective vaccines
US9642903B2 (en) 2009-07-16 2017-05-09 Lior Carmon Antigen specific multi epitope-based anti-infective vaccines
WO2011009173A1 (en) * 2009-07-23 2011-01-27 Mater Medical Research Institute Cancer immunotherapy
US20110195045A1 (en) * 2010-01-12 2011-08-11 Emmanuelle Godefroy Methods, agents and peptides for inducing an immune response to matrix metalloproteinase-2 expressing tumors
US8481477B2 (en) * 2010-01-12 2013-07-09 New York University Methods, agents and peptides for inducing an immune response to matrix metalloproteinase-2 expressing tumors
US9395368B2 (en) 2010-01-12 2016-07-19 New York University Methods, agents and peptides for inducing an immune response to matrix metalloproteinase-2 expressing tumors
US9506933B2 (en) 2011-01-07 2016-11-29 Toagosei Co., Ltd. Method of preparing antigen for acquiring anti-hydrophobic peptide antibody
EP2662389A4 (en) * 2011-01-07 2015-08-12 Toagosei Co Ltd Method for preparing antigen to obtain antihydrophobic peptide antibody
WO2013006050A1 (en) * 2011-07-06 2013-01-10 Isa Pharmaceuticals B.V. Peptides inducing or enhancing an immune response against prostate-specific membrane protein (PSMA)
WO2013038412A2 (en) 2011-09-15 2013-03-21 Vaxil Bio Therapeutics Ltd. Antibodies directed against signal peptides, methods and uses thereof
US9732153B2 (en) 2011-09-15 2017-08-15 Vaxil Biotherapeutics Ltd. Antibodies directed against signal peptides, methods and uses thereof
US9370182B2 (en) 2012-05-28 2016-06-21 Toagosei Co., Ltd. Antimicrobial peptide and use thereof
JP2017158561A (en) * 2012-07-20 2017-09-14 ジェムバックス アンド カエル カンパニー,リミティド Peptides having anti-inflammatory activity and compositions comprising the same
CN103570821A (en) * 2012-07-27 2014-02-12 北京智飞绿竹生物制药有限公司 Mucin-1 antigenic polypeptide and application thereof as tumor vaccine
CN103570818A (en) * 2012-07-27 2014-02-12 北京智飞绿竹生物制药有限公司 Tumor antigenic polypeptide and application thereof as tumor vaccine
CN103570818B (en) * 2012-07-27 2016-06-29 北京智飞绿竹生物制药有限公司 Tumor antigenic polypeptide and the purposes as tumor vaccine thereof
US9480727B2 (en) 2012-10-18 2016-11-01 Toagosei Co. Ltd. Synthetic peptide for inhibiting expression of type 2 TNF receptor and use thereof
WO2015014820A1 (en) * 2013-07-29 2015-02-05 Eberhard Karls Universitaet Tuebingen Medizinische Fakultaet Prostate cancer immunotherapy
CN104387453A (en) * 2014-12-08 2015-03-04 深圳市同康生物医药有限公司 Dendritic cell targeted peptide, coding gene and application
US10568948B2 (en) 2015-05-13 2020-02-25 Agenus Inc. Vaccines for treatment and prevention of cancer
US11213578B2 (en) 2017-03-03 2022-01-04 Treos Bio Limited Vaccine
US11426452B2 (en) 2017-03-03 2022-08-30 Treos Bio Limited Vaccine
US11628211B2 (en) 2017-03-03 2023-04-18 Treos Bio Limited Vaccine
US11065317B2 (en) 2018-04-26 2021-07-20 Agenus Inc. Heat shock protein-binding peptide compositions and methods of use thereof
WO2020048990A1 (en) * 2018-09-04 2020-03-12 Treos Bio Zrt Peptide vaccines
US11666644B2 (en) 2018-09-04 2023-06-06 Treos Bio Limited Peptide vaccines
WO2020243787A1 (en) * 2019-06-06 2020-12-10 The University Of Sydney Anti-inflammatory agents
CN114555624A (en) * 2019-06-06 2022-05-27 悉尼大学 Anti-inflammatory agents
EP3980440A4 (en) * 2019-06-06 2023-07-05 The University Of Sydney Anti-inflammatory agents

Also Published As

Publication number Publication date
CA2665816C (en) 2016-07-12
AU2007298494A1 (en) 2008-03-27
CA2665816A1 (en) 2008-03-27
US9487574B2 (en) 2016-11-08
US20220072113A1 (en) 2022-03-10
EP2089423A1 (en) 2009-08-19
US11179452B2 (en) 2021-11-23
US20100074925A1 (en) 2010-03-25
AU2007298494B2 (en) 2013-09-26
US20170072036A1 (en) 2017-03-16
EP2089423B1 (en) 2016-10-26
AU2007298494A8 (en) 2009-05-28

Similar Documents

Publication Publication Date Title
US20220072113A1 (en) Antigen specific multi epitope vaccines
US9907842B2 (en) Cytotoxic T lymphocyte inducing immunogens for prevention treatment and diagnosis of cancer
JP5156882B2 (en) HLA-A2 tumor-associated antigenic peptide and composition
US20130011424A1 (en) Polyepitope constructs and methods for their preparation and use
EP1239866A1 (en) INDUCING CELLULAR IMMUNE RESPONSES TO HER2/neu USING PEPTIDE AND NUCLEIC ACID COMPOSITIONS
WO2007150077A2 (en) Cytotoxic t-lymphocyte-inducing immunogens for prevention, treatment, and diagnosis of cancer
EP1244465A1 (en) Inducing cellular immune responses to prostate cancer antigens using peptide and nucleic acid compositions
WO2001041741A9 (en) Hla class i a2 tumor associated antigen peptides and vaccine compositions
US20090208518A1 (en) Immunogenic peptides for the treatment of prostate and breast cancer
US20040146519A1 (en) Inducing cellular immune responses to carcinoembryonic antigen using peptide and nucleic acid compositions
CZ20022874A3 (en) Immunogenic composition
US20030224036A1 (en) Hla class I a2 tumor associated antigen peptides and vaccine compositions
AU764550B2 (en) Isolated peptides which bind to HLA-B35 molecules
JP2003517310A (en) Induction of a cellular immune response against MAGE2 / 3 using peptide and nucleic acid compositions
US20060009393A1 (en) Immunogenic epitopes for fibroblast growth factors 5 (FGF-5)
IL197737A (en) Peptide vaccine consisting of a signal peptide, pharmaceutical compositions comprising it and uses thereof
WO2004045555A2 (en) Immunogenic epitopes for fibroblast growth factor 5 (fgf-5) presented by hla-a3 and hla-a2

Legal Events

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

Ref document number: 07827143

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2665816

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007298494

Country of ref document: AU

REEP Request for entry into the european phase

Ref document number: 2007827143

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2595/DELNP/2009

Country of ref document: IN

Ref document number: 2007827143

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2007298494

Country of ref document: AU

Date of ref document: 20070923

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12442495

Country of ref document: US