PROTEIN ANTIGENS AND USES THEREOF

Information

  • Patent Application
  • 20230241207
  • Publication Number
    20230241207
  • Date Filed
    April 03, 2018
    6 years ago
  • Date Published
    August 03, 2023
    a year ago
Abstract
The field of the present invention relates to immunotherapeutic peptides, peptide binding agents, and their use, for example, in the immunotherapy of cancer.
Description
FIELD

The field of the present invention relates to immunotherapeutic peptides, nucleic acids encoding the peptides, peptide binding agents, and their use, for example, in the immunotherapy of cancer. In one aspect, the invention provides non-mutated protein epitopes expressed in cancer cells, useful alone or in combination with other tumor-associated peptides, anti-cancer, or immunomodulatory agents to treat cancer.


BACKGROUND

Tumor vaccines are typically composed of tumor antigens and immunostimulatory molecules (e.g., adjuvants, cytokines or TLR ligands) that work together to induce antigen-specific cytotoxic T cells (CTLs) that recognize and lyse tumor cells. Such vaccines contain either shared tissue restricted tumor antigens or a mixture of shared and patient-specific antigens in the form of whole tumor cell preparations. The shared tissue restricted tumor antigens are ideally immunogenic proteins with selective expression in tumors across many individuals and are commonly delivered to patients as synthetic peptides or recombinant proteins. In contrast, whole tumor cell preparations are delivered to patients as autologous irradiated cells, cell lysates, cell fusions, heat-shock protein preparations or total mRNA. Since whole tumor cells are isolated from the autologous patient, the cells may include patient-specific tumor antigens as well as shared tumor antigens. Finally, there is a third class of tumor antigens, neoantigens, which consists of proteins with tumor-specific mutations (which can be patient-specific or shared) that result in altered amino acid sequences. Accordingly, there is still a need for developing additional cancer therapeutics.


SUMMARY

Provided herein an isolated antigenic peptide comprising an epitope from a sequence in Table 1 or 2. The present disclosure is also directed to an isolated antigenic peptide 100 amino acids or less in length which comprises an epitope from a sequence in Table 1 or 2. The present disclosure is also directed to an isolated antigenic peptide comprising an epitope from a sequence in Table 3 or 4. The present disclosure is also directed to an isolated antigenic peptide 100 amino acids or less in length which comprises an epitope from a sequence in Table 3 or 4. The present disclosure is also directed to an isolated antigenic peptide comprising an epitope from a sequence in Table 5 or 6. The present disclosure is also directed to an isolated antigenic peptide 100 amino acids or less in length which comprises an epitope from a sequence in Table 5 or 6.


In one embodiment, the isolated antigenic peptide is a retroviral antigen. In another embodiment, the isolated antigenic peptide is a non-mutated overexpressed antigen. In another embodiment, the isolated antigenic peptide is a viral antigen.


In one embodiment, the isolated antigenic peptide is between about 5 to about 50 amino acids in length. In another embodiment, the isolated antigenic peptide is between about 15 to about 35 amino acids in length. In another embodiment, the isolated antigenic peptide is about 15 amino acids or less in length. In another embodiment, the isolated antigenic peptide is between about 8 and about 11 amino acids in length. In another embodiment, the isolated antigenic peptide is 9 or 10 amino acids in length. In one embodiment, the isolated antigenic peptide binds major histocompatibility complex (MHC) class I. In another embodiment, the isolated antigenic peptide binds MHC class I with a binding affinity of less than about 500 nM.


In one embodiment, the isolated antigenic peptide is about 30 amino acids or less in length. In another embodiment, the isolated antigenic peptide is between about 6 and about 25 amino acids in length. In another embodiment, the isolated antigenic peptide is between about 15 and about 24 amino acids in length. In another embodiment, the isolated antigenic peptide is between about 9 and about 15 amino acids in length. In one embodiment, the isolated antigenic peptide binds MHC class II. In another embodiment, the isolated antigenic peptide binds MHC class II with a binding affinity of less than about 1000 nM.


In one embodiment, the isolated antigenic peptide further comprises flanking amino acids. In another embodiment, the flanking amino acids are not native flanking amino acids. In one embodiment, the isolated antigenic peptide is linked to at least a second antigenic peptide. In another embodiment, the peptides are linked using a poly-glycine or poly-serine linker. In another embodiment, the second antigenic peptide binds MHC class I or class II with a binding affinity of less than about 1000 nM. In another embodiment, the second antigenic peptide binds MHC class I or class II with a binding affinity of less than about 500 nM. In another embodiment, both of the epitopes bind to human leukocyte antigen (HLA)-A, -B, -C, -DP, -DQ, or -DR. In another embodiment, the isolated antigenic peptide binds a class I HLA and the second antigenic peptide binds a class II HLA. In another embodiment, the isolated antigenic peptide binds a class II HLA and the second antigenic peptide binds a class I HLA.


In one embodiment, the isolated antigenic peptide further comprises modifications which increase in vivo half-life, cellular targeting, antigen uptake, antigen processing, MHC affinity, MHC stability, or antigen presentation. In another embodiment, the modification is conjugation to a carrier protein, conjugation to a ligand, conjugation to an antibody, PEGylation, polysialylation HESylation, recombinant PEG mimetics, Fc fusion, albumin fusion, nanoparticle attachment, nanoparticulate encapsulation, cholesterol fusion, iron fusion, acylation, amidation, glycosylation, side chain oxidation, phosphorylation, biotinylation, the addition of a surface active material, the addition of amino acid mimetics, or the addition of unnatural amino acids. In one embodiment, the cells that are targeted are antigen presenting cells. In another embodiment, the antigen presenting cells are dendritic cells. In another embodiment, the dendritic cells are targeted using DEC205, XCR1, CD197, CD80, CD86, CD123, CD209, CD273, CD283, CD289, CD184, CD85h, CD85j, CD85k, CD85d, CD85g, CD85a, CD141, CD11c, CD83, TSLP receptor, or CD1a marker. In another embodiment, the dendritic cells are targeted using the CD141, DEC205, or XCR1 marker.


In one embodiment, provided herein is an in vivo delivery system comprising an isolated antigenic peptide described herein. In another embodiment, the delivery system includes cell-penetrating peptides, nanoparticulate encapsulation, virus like particles, or liposomes. In another embodiment, the cell-penetrating peptide is TAT peptide, herpes simplex virus VP22, transportan, or Antp.


In one embodiment, provided herein is a cell comprising an isolated antigenic peptide described herein. In another embodiment, the cell is an antigen presenting cell. In another embodiment, the cell is a dendritic cell.


In one embodiment, provided herein is a composition comprising an isolated antigenic peptide described herein. In another embodiment, the composition comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 of the isolated antigenic peptides comprising a tumor-specific epitope defined in Table 1 or 2. In another embodiment, the composition comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 of the isolated antigenic peptides comprising a tumor-specific epitope defined in Table 3 or 4. In another embodiment, the composition comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 of the isolated antigenic peptides comprising a tumor-specific epitope defined in Table 5 or 6. In another embodiment, the composition comprises between 2 and 20 antigenic peptides. In another embodiment, the composition further comprises at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 additional antigenic peptides. In another embodiment, the composition comprises between about 4 and about 20 additional antigenic peptides. In another embodiment, the additional antigenic peptide is specific for an individual patient's tumor. In another embodiment, an antigenic peptide is selected by identifying differences in expression between the transcriptome or proteome of the patient's tumor sample and the transcriptome or proteome of a non-tumor sample. In another embodiment, the samples are fresh or formalin-fixed paraffin embedded tumor tissues, freshly isolated cells, or circulating tumor cells. In some embodiments, the sequences of the antigenic peptides are determined by Next Generation Sequencing.


In one embodiment, provided herein is an isolated polynucleotide encoding the isolated antigenic peptide described herein. In another embodiment, the isolated polynucleotide is RNA, optionally a self-amplifying RNA. In another embodiment, the RNA is modified to increase stability, increase cellular targeting, increase translation efficiency, adjuvanticity, cytosol accessibility, and/or decrease cytotoxicity. In another embodiment, the modification is conjugation to a carrier protein, conjugation to a ligand, conjugation to an antibody, codon optimization, increased GC-content, incorporation of modified nucleosides, incorporation of 5′-cap or cap analog, and/or incorporation of an unmasked poly-A sequence.


In one embodiment, provided herein is a cell comprising a polynucleotide described herein.


In one embodiment, provided herein is a vector comprising a polynucleotide described herein. In another embodiment, the polynucleotide is operably linked to a promoter. In another embodiment, the vector is a self-amplifying RNA replicon, plasmid, phage, transposon, cosmid, virus, or virion. In another embodiment, the vector is an adeno-associated virus, herpesvirus, lentivirus, or pseudotypes thereof.


In one embodiment, provided herein is an in vivo delivery system comprising an isolated polynucleotide described herein. In another embodiment, the delivery system includes spherical nucleic acids, viruses, virus-like particles, plasmids, bacterial plasmids, or nanoparticles.


In one embodiment, provided herein is a cell comprising a vector or delivery system described herein. In another embodiment, the cell is an antigen presenting cell. In another embodiment, the cell is a dendritic cell. In another embodiment, the cell is an immature dendritic cell.


In one embodiment, provided herein is a composition comprising at least one polynucleotide described herein. In another embodiment, the composition comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 of the isolated polynucleotides. In another embodiment, the composition comprises between about 2 and about 20 polynucleotides. In another embodiment, the composition further comprises at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 additional antigenic polynucleotides encoding for additional antigenic peptides. In another embodiment, the composition comprises between about 4 and about 20 additional antigenic polynucleotides. In another embodiment, the isolated polynucleotides and the additional antigenic polynucleotides are linked. In another embodiment, the polynucleotides are linked using nucleic acids that encode a poly-glycine or poly-serine linker. In another embodiment, at least one of the additional antigenic peptide is specific for an individual patient's tumor. In another embodiment, an antigenic peptide is selected by identifying differences in expression between the transcriptome or proteome of the patient's tumor sample and the transcriptome or proteome of a non-tumor sample. In another embodiment, the samples are fresh or formalin-fixed paraffin embedded tumor tissues, freshly isolated cells, or circulating tumor cells. In some embodiments, the sequences of the antigenic peptides are determined by Next Generation Sequencing.


In one embodiment, provided herein is a T cell receptor (TCR) capable of binding at least one antigenic peptide described herein. In another embodiment, the TCR is capable of binding the isolated antigenic peptide in the context of MHC class I or class II.


In one embodiment, provided herein is a chimeric antigen receptor comprising: (i) a T cell activation molecule; (ii) a transmembrane region; and (iii) an antigen recognition moiety capable of binding an isolated antigenic peptide described herein. In another embodiment, CD3-zeta is the T cell activation molecule. In another embodiment, the chimeric antigen receptor further comprises at least one costimulatory signaling domain. In another embodiment, the signaling domain is CD28, 4-1BB, ICOS, OX40, ITAM, or Fc epsilon RI-gamma. In another embodiment, the antigen recognition moiety is capable of binding the isolated antigenic peptide in the context of MHC class I or class II. In another embodiment, the chimeric antigen receptor comprises the CD3-zeta, CD28, CTLA-4, ICOS, BTLA, KIR, LAG3, CD137, OX40, CD27, CD40L, Tim-3, A2aR, or PD-1 transmembrane region. In another embodiment, the tumor-specific epitope is located in the extracellular domain of a tumor associated polypeptide.


In one embodiment, provided herein is a T cell comprising the T cell receptor or chimeric antigen receptor described herein. In one embodiment, the T cell is a helper or cytotoxic T cell.


In one embodiment, provided herein is a nucleic acid comprising a promoter operably linked to a polynucleotide encoding a T cell receptor described herein. In another embodiment, the TCR is capable of binding the at least one antigenic peptide in the context of major histocompatibility complex (MHC) class I or class II. In one embodiment, the nucleic acid comprises a promoter operably linked to a polynucleotide encoding a chimeric antigen receptor described herein. In another embodiment, the antigen recognition moiety is capable of binding the at least one antigenic peptide in the context of major histocompatibility complex (MHC) class I or class II. In another embodiment, the tumor-specific epitope is located in the extracellular domain of a tumor associated polypeptide. In another embodiment, the nucleic acid comprises the CD3-zeta, CD28, CTLA-4, ICOS, BTLA, KIR, LAG3, CD137, OX40, CD27, CD40L, Tim-3, A2aR, or PD-1 transmembrane region.


In one embodiment, provided herein is an antibody capable of binding at least one antigenic peptide listed in Table 1 or 2. In another embodiment, provided herein is an antibody capable of binding at least one antigenic peptide listed in Table 3 or 4. In another embodiment, provided herein is an antibody capable of binding at least one antigenic peptide listed in Table 5 or 6. In another embodiment, the at least one antigenic peptide listed in Table 1 or 2 is a retroviral antigenic peptide. In another embodiment, the at least one antigenic peptide listed in Table 3 or 4 is a non-mutated overexpressed antigenic peptide. In another embodiment, the at least one antigenic peptide listed in Table 5 or 6 is a viral antigenic peptide.


In one embodiment, provided herein is a modified cell transfected or transduced with a nucleic acid described herein. In one embodiment, the modified cell is a T cell, tumor infiltrating lymphocyte, NK-T cell, TCR-expressing cell, CD4+ T cell, CD8+ T cell, or NK cell.


In one embodiment, provided herein is a composition comprising a T cell receptor or chimeric antigen receptor described herein. In another embodiment, a composition comprises autologous patient T cells containing a T cell receptor or chimeric antigen receptor described herein. In another embodiment, the composition further comprises an immune checkpoint inhibitor. In another embodiment, the composition further comprises at least two immune checkpoint inhibitors. In another embodiment, each of the immune checkpoint inhibitors inhibits a checkpoint protein selected from the group consisting of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, and B-7 family ligands or a combination thereof. In another embodiment, each of the immune checkpoint inhibitors interacts with a ligand of a checkpoint protein selected from the group consisting of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, and B-7 family ligands or a combination thereof.


In one embodiment, the composition further comprises an immune modulator or adjuvant. In another embodiment, the immune modulator is a co-stimulatory ligand, a TNF ligand, an Ig superfamily ligand, CD28, CD80, CD86, ICOS, CD40L, OX40, CD27, GITR, CD30, DR3, CD69, or 4-1BB. In another embodiment, the immune modulator is at least one cancer cell or cancer cell extract. In another embodiment, the cancer cell is autologous to the subject in need of the composition. In another embodiment, the cancer cell has undergone lysis or been exposed to UV radiation. In another embodiment, the composition further comprises an adjuvant. In another embodiment, the adjuvant is selected from the group consisting of: Poly(I:C), Poly-ICLC, STING agonist, 1018 ISS, aluminum salts, Amplivax, AS15, BCG, CP-870,893, CpG7909, CyaA, dSLIM, GM-CSF, IC30, IC31, Imiquimod, ImuFact IMP321, IS Patch, ISS, ISCOMATRIX, JuvImmune, LipoVac, MF59, monophosphoryl lipid A, Montanide IMS 1312 VG, Montanide ISA 206 VG, Montanide ISA 50 V2, Montanide ISA 51 VG, OK-432, OM-174, OM-197-MP-EC, ISA-TLR2 agonist, ONTAK, PepTel®. vector system, PLG microparticles, resiquimod, SRL172, virosomes and other virus-like particles, YF-17D, VEGF trap, R848, beta-glucan, Pam3Cys, Pam3CSK4, acrylic or methacrylic polymers, copolymers of maleic anhydride, and QS21 stimulon. In another embodiment, the adjuvant induces a humoral when administered to a subject. In another embodiment, the adjuvant induces a T helper cell type 1 when administered to a subject.


In one embodiment, provided herein is a method of inhibiting growth of a tumor cell expressing a tumor-specific epitope defined in Table 1 or 2, comprising contacting a tumor cell with a peptide, polynucleotide, delivery system, vector, composition, antibody, or cells of the invention. In another embodiment, provided herein is a method of inhibiting growth of a tumor cell expressing a tumor-specific epitope defined in Table 3 or 4, comprising contacting the tumor cell with the peptide, polynucleotide, delivery system, vector, composition, antibody, or cells of the invention. In another embodiment, provided herein is a method of inhibiting growth of a tumor cell expressing a tumor-specific epitope defined in Table 5 or 6, comprising contacting the tumor cell with the peptide, polynucleotide, delivery system, vector, composition, antibody, or cells of the invention.


In one embodiment, provided herein is a method of treating cancer or initiating, enhancing, or prolonging an anti-tumor response in a subject in need thereof comprising administering to the subject the peptide, polynucleotide, vector, composition, antibody, or cells described herein. In one embodiment, the cancer is selected from the group consisting of CRC, head and neck, stomach, lung squamous, lung adeno, Prostate, Bladder. stomach, renal cell carcinoma, and uterine. In one embodiment, the cancer is selected from the group consisting of melanoma, lung squamous, DLBCL, uterine, head and neck, uterine, liver, and CRC. In one embodiment, the cancer is selected from the group consisting of cervical, head and neck, anal, stomach, Burkitt's lymphoma, and nasopharyngeal carcinoma.


In one embodiment, the subject is a human. In another embodiment, the subject has cancer. In another embodiment, the cancer is selected from the group consisting of urogenital, gynecological, lung, gastrointestinal, head and neck cancer, malignant glioblastoma, malignant mesothelioma, non-metastatic or metastatic breast cancer, malignant melanoma, triple-negative breast cancer (TNBC), smoldering myeloma (SMM), Merkel Cell Carcinoma or bone and soft tissue sarcomas, hematologic neoplasias, multiple myeloma, acute myelogenous leukemia, chronic myelogenous leukemia, myelodysplastic syndrome and acute lymphoblastic leukemia, non-small cell lung cancer (NSCLC), breast cancer, metastatic colorectal cancers, hormone sensitive or hormone refractory prostate cancer, colorectal cancer, ovarian cancer, hepatocellular cancer, renal cell cancer, pancreatic cancer, gastric cancer, esophageal cancers, hepatocellular cancers, cholangiocellular cancers, head and neck squamous cell cancer soft tissue sarcoma, and small cell lung cancer. In another embodiment, the subject has undergone surgical removal of the tumor. In another embodiment, the peptide, polynucleotide, vector, composition, or cells is administered via intravenous, intraperitoneal, intratumoral, intradermal, or subcutaneous administration. In another embodiment, the peptide, polynucleotide, vector, composition, or cells is administered into an anatomic site that drains into a lymph node basin. In another embodiment, the administration is into multiple lymph node basins. In another embodiment, the administration is by a subcutaneous or intradermal route.


In one embodiment of the method, a peptide is administered. In another embodiment, the administration is intratumorally. In another embodiment of the method, a polynucleotide, optionally RNA, is administered. In another embodiment, the polynucleotide is administered intravenously. In one embodiment of the method, a cell is administered. In another embodiment, the cell is a T cell or dendritic cell. In another embodiment, the peptide or polynucleotide comprises an antigen presenting cell targeting moiety.


One embodiment of the method further comprises administering at least one immune checkpoint inhibitor to a subject. In another embodiment, the checkpoint inhibitor is a biologic therapeutic or a small molecule. In another embodiment, the checkpoint inhibitor is selected from the group consisting of a monoclonal antibody, a humanized antibody, a fully human antibody and a fusion protein or a combination thereof. In another embodiment, the checkpoint inhibitor inhibits a checkpoint protein selected from the group consisting of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, and B-7 family ligands or a combination thereof. In another embodiment, the checkpoint inhibitor interacts with a ligand of a checkpoint protein selected from the group consisting of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, and B-7 family ligands or a combination thereof. In another embodiment, two or more checkpoint inhibitors are administered. In another embodiment, the checkpoint inhibitors are: (i) ipilimumab or tremelimumab, and (ii) nivolumab. In another embodiment, the checkpoint inhibitor and the composition are administered simultaneously or sequentially in any order. In another embodiment, the peptide, polynucleotide, vector, composition, or cells is administered prior to the checkpoint inhibitor. In another embodiment, the peptide, polynucleotide, vector, composition, or cells is administered after the checkpoint inhibitor. In another embodiment, administration of the checkpoint inhibitor is continued throughout antigen peptide, polynucleotide, vector, composition, or cell therapy. In another embodiment, the antigen peptide, polynucleotide, vector, composition, or cell therapy is administered to subjects that only partially respond or do not respond to checkpoint inhibitor therapy. In another embodiment, the composition is administered intravenously or subcutaneously. In another embodiment, the checkpoint inhibitor is administered intravenously or subcutaneously. In another embodiment, the checkpoint inhibitor is administered subcutaneously within about 2 cm of the site of administration of the composition. In another embodiment, the composition is administered into the same draining lymph node as the checkpoint inhibitor.


In one embodiment of the method, an additional agent is administered. In another embodiment, the agent is a chemotherapeutic agent, an immunomodulatory drug, an immune metabolism modifying drug, a targeted therapy, radiation an anti-angiogenesis agent, or an agent that reduces immune-suppression. In another embodiment, the chemotherapeutic agent is an alkylating agent, a topoisomerase inhibitor, an anti-metabolite, or an anti-mitotic agent. In another embodiment, the additional agent is an anti-glucocorticoid induced tumor necrosis factor family receptor (GITR) agonistic antibody or antibody fragment, ibrutinib, docetaxeol, cisplatin, or cyclophosphamide. In another embodiment, the administration elicits a CD4+ T cell immune response. In another embodiment, the administration elicits a CD4+ T cell immune response and a CD8+ T cell immune response.


In one embodiment, provided herein is a method for stimulating an immune response in a subject, comprising administering an effective amount of modified cells or composition described herein. In another embodiment, the immune response is cytotoxic and/or humoral immune response. In another embodiment, the method stimulates a T cell-mediated immune response in a subject. In another embodiment, the T cell-mediated immune response is directed against a target cell. In another embodiment, the target cell is a tumor cell. In another embodiment, the modified cells are transfected or transduced in vivo. In another embodiment, the modified cells are transfected or transduced ex vivo. In another embodiment, the modified cells are autologous patient T cells. In another embodiment, the autologous patient T cells are obtained from a patient that has received an antigen peptide or nucleic acid vaccine. In another embodiment, the antigen peptide or nucleic acid vaccine comprises at least one personalized antigen. In another embodiment, the antigen peptide or nucleic acid vaccine comprises at least one additional antigenic peptide listed in Table 1 or 2. In another embodiment, the antigen peptide or nucleic acid vaccine comprises at least one additional antigenic peptide listed in Table 3 or 4. In another embodiment, the antigen peptide or nucleic acid vaccine comprises at least one additional antigenic peptide listed in Table 5 or 6. In another embodiment, the at least one additional antigenic peptide listed in Table 1 or 2 is a retroviral antigenic peptide. In another embodiment, the at least one additional antigenic peptide listed in Table 3 or 4 is a non-mutated overexpressed antigenic peptide. In another embodiment, the at least one additional antigenic peptide listed in Table 5 or 6 is a viral antigenic peptide. In another embodiment, the patient received a chemotherapeutic agent, an immunomodulatory drug, an immune metabolism modifying drug, targeted therapy or radiation prior to and/or during receipt of the antigen peptide or nucleic acid vaccine. In another embodiment, the patient receives treatment with at least one checkpoint inhibitor. In another embodiment, the autologous T cells are obtained from a patient that has already received at least one round of T cell therapy containing an antigen. In another embodiment, the method further comprises adoptive T cell therapy. In another embodiment, the adoptive T cell therapy comprises autologous T-cells. In another embodiment, the autologous T-cells are targeted against tumor antigens. In another embodiment, the adoptive T cell therapy further comprises allogenic T-cells. In another embodiment, the allogenic T-cells are targeted against tumor antigens. In another embodiment, the adoptive T cell therapy is administered before the checkpoint inhibitor.


In one embodiment, provided herein is a method for evaluating the efficacy of treatment comprising: (i) measuring the number or concentration of target cells in a first sample obtained from the subject before administering the modified cell, (ii) measuring the number concentration of target cells in a second sample obtained from the subject after administration of the modified cell, and (iii) determining an increase or decrease of the number or concentration of target cells in the second sample compared to the number or concentration of target cells in the first sample. In another embodiment, the treatment efficacy is determined by monitoring a clinical outcome; an increase, enhancement or prolongation of anti-tumor activity by T cells; an increase in the number of anti-tumor T cells or activated T cells as compared with the number prior to treatment; B cell activity; CD4 T cell activity; or a combination thereof. In another embodiment, the treatment efficacy is determined by monitoring a biomarker. In another embodiment, the biomarker is selected from the group consisting of CEA, Her-2/neu, bladder tumor antigen, thyroglobulin, alpha-fetoprotein, PSA, CA 125, CA19.9, CA 15.3, leptin, prolactin, osteopontin, IGF-II, CD98, fascin, sPIgR, 14-3-3 eta, troponin I, and b-type natriuretic peptide. In another embodiment, the clinical outcome is selected from the group consisting of tumor regression; tumor shrinkage; tumor necrosis; anti-tumor response by the immune system; tumor expansion, recurrence or spread; or a combination thereof. In another embodiment, the treatment effect is predicted by presence of T cells or by presence of a gene signature indicating T cell inflammation or a combination thereof.


In one embodiment, provided herein is a method of treating cancer or initiating, enhancing, or prolonging an anti-tumor response in a subject in need thereof comprising administering to the subject: (a) the peptide, polynucleotide, vector, composition, antibody, or cells described herein; and (b) at least one checkpoint inhibitor. In another embodiment, the method further comprises administration of an immunomodulator or adjuvant. In another embodiment, the immunomodulator or adjuvant is selected from the group consisting of Poly(I:C), Poly-ICLC, STING agonist, 1018 ISS, aluminum salts, Amplivax, AS15, BCG, CP-870,893, CpG7909, CyaA, dSLIM, GM-CSF, IC30, IC31, Imiquimod, ImuFact IMP321, IS Patch, ISS, ISCOMATRIX, Juvlmmune, LipoVac, MF59, monophosphoryl lipid A, Montanide IMS 1312 VG, Montanide ISA 206 VG, Montanide ISA 50 V2, Montanide ISA 51 VG, OK-432, OM-174, OM-197-MP-EC, ISA-TLR2 agonist, ONTAK, PepTel® vector system, PLG microparticles, resiquimod, SRL172, virosomes and other virus-like particles, YF-17D, VEGF trap, R848, beta-glucan, Pam3Cys, Pam3CSK4, acrylic or methacrylic polymers, copolymers of maleic anhydride, and QS21 stimulon. a co-stimulatory ligand, a TNF ligand, an Ig superfamily ligand, CD28, CD80, CD86, ICOS, CD40L, OX40, CD27, GITR, CD30, DR3, CD69, or 4-1BB. In another embodiment, the immunomodulator or adjuvant is Poly-ICLC. In another embodiment, the checkpoint inhibitor is an anti-PD1 antibody or antibody fragment. In another embodiment, the inhibitor of the PD-1 pathway is nivolumab. In another embodiment, the checkpoint inhibitor is an anti-CTLA4 antibody or antibody fragment. In another embodiment, the anti-CTLA4 antibody is ipilimumab or tremelimumab. In another embodiment, the method comprises administering both an anti-PD1 antibody and an anti-CTLA4 antibody. In another embodiment, the administration of the checkpoint inhibitor is initiated before initiation of administration of the peptide, polynucleotide, vector, composition, antibody, or cell. In another embodiment, the administration of the checkpoint inhibitor is initiated after initiation of administration of the peptide, polynucleotide, vector, composition, antibody, or cell. In another embodiment, the administration of the checkpoint inhibitor is initiated simultaneously with the initiation of administration of the peptide, polynucleotide, vector, composition, antibody, or cell. In another embodiment, the peptide, polynucleotide, vector, composition, antibody, or cell is administered intravenously or subcutaneously. In another embodiment, the checkpoint inhibitor is administered intravenously or subcutaneously. In another embodiment, the checkpoint inhibitor is administered subcutaneously within about 2 cm of the site of administration of the peptide, polynucleotide, vector, composition, antibody, or cell. In another embodiment, the peptide, polynucleotide, vector, composition, antibody, or cell is administered into the same draining lymph node as the checkpoint inhibitor.


In one embodiment of the therapeutic methods, the additional therapeutic agent is for example, a chemotherapeutic or biotherapeutic agent, radiation, or immunotherapy. Any suitable therapeutic treatment for a particular cancer may be administered. Examples of chemotherapeutic and biotherapeutic agents include, but are not limited to, an angiogenesis inhibitor, such as hydroxy angiostatin K 1-3, DL-a-Difluoromethyl-ornithine, endostatin, fumagillin, genistein, minocycline, staurosporine, and thalidomide; a DNA intercalator/cross-linker, such as Bleomycin, Carboplatin, Carmustine, Chlorambucil, Cyclophosphamide, cis-Diammineplatinum (II) dichloride (Cisplatin), Melphalan, Mitoxantrone, and Oxaliplatin; a DNA synthesis inhibitor, such as (±)-Amethopterin (Methotrexate), 3-Amino-1,2,4-benzotriazine 1,4-dioxide, Aminopterin, Cytosine β-D-arabinofuranoside, 5-Fluoro-5′-deoxyuridine, 5-Fluorouracil, Ganciclovir, Hydroxyurea, and Mitomycin C; a DNA-RNA transcription regulator, such as Actinomycin D, Daunorubicin, Doxorubicin, Homoharringtonine, and Idarubicin; an enzyme inhibitor, such as S(-+-)-Camptothecin, Curcumin, (−)-Deguelin, 5,6-Dichlorobenzimidazole 1-β-D-ribofuranoside, Etoposide, Formestane, Fostriecin, Hispidin, 2-Immo-1-imidazoli-dineacetic acid (Cyclocreatine), Mevinolin, Trichostatin A, Tyrphostin AG 34, and Tyrphostin AG 879; a gene regulator, such as 5-Aza-2′-deoxycytidine, 5-Azacytidine, Cholecalciferol (Vitamin D3), 4-Hydroxytamoxifen, Melatonin, Mifepristone, Raloxifene, all trans-Retinal (Vitamin A aldehyde), Retinoic acid all trans (Vitamin A acid), 9-cis-Retinoic Acid, 13-cis-Retinoic acid, Retinol (Vitamin A), Tamoxifen, and Troglitazone; a microtubule inhibitor, such as Colchicine, docetaxel, Dolastatin15, Nocodazole, Paclitaxel, Podophyllotoxin, Rhizoxin, Vinblastine, Vincristine, Vindesine, and Vinorelbine (Navelbine); and an unclassified therapeutic agent, such as 17-(Allylamino)-17-demethoxygeldanamycin, 4-Amino-1,8-naphthalimide, Apigenin, Brefeldin A, Cimetidine, Dichioromethylene-diphosphonic acid, Leuprolide (Leuprorelin), Luteinizing Hormone-Releasing Hormone, Pifithrin-a, Rapamycin, Sex hormone-binding globulin, Thapsigargin, and Urinary trypsin inhibitor fragment (Bikunin). The therapeutic agent may be altretamine, amifostine, asparaginase, capecitabine, cladribine, cisapride, cyiarahirse, dacarbazine (DT1C), dactinomycin, dronabinol, epoetin alpha, “filgrastim, fludarabine, gemcitabine, granisetron, ifosfamide, irinotecan, lansoprazole, levamisole, leucovorin, megestrol, mesna, metoclopramide, mitotane, omeprazole, ondansetron, pilocarpine, prochloroperazine, or topotecan hydrochloride. The therapeutic agent may be a monoclonal antibody such as rituximab (Rituxan®), alemtuzumab (Campath®), Bevacizumab (Avastin®), Cetuximab (Erbitux®), panitumumab (Vectibix®), and trastuzumab (Herceptin®), Vemurafenib (Zelboraf®) imatinib mesylate (Gleevec®), erlotinib (Tarceva®), gefitinib (Iressa®), Vismodegib (Erivedge™), 90Y-ibritumomab tiuxetan, 1311-tosit.umomab, ado-trastuzumab emtansine, lapatinib (Tykerb®), pertuzumab (Perjeta™), ado-trastuzumab emtansine (Adcyla™), regorafenib (Stivarga®), sunitinib (Sutent®), Denosumab (Xgeva®), sorafenib (Nexavar®), pazopanib (Votrient®), axitinib (Inita®), dasatinib (Sprycel®), nilotinib (Tasigna®), bosutinib (Bosulif®), ofatumumab (Arzerra®), obinutuzumab (Gazyva™), ibrutinib (Imbruvica™), idelalisib (Zydelig®), crizotinib (Xalkori®), erlotinib (Tarceva®), afatimb dimaleate (Giiotrif®), ceritinib (LDK378/Zykadia), Tositumomab and 1311-tositumomab (Bexxar®), ibritumomab tiuxetan (Zevalin®), brentuximab vedotin (Adcetris®), bortezomib (Velcade®), siltuximab (Sylvant™), trametinib (Ekinist®), dabrafenib (Tafmlar®), pembrolizimiab (Keytruda®), carfilzomib (Kyprolis®), Ramucirumab (Cyramza™), Cabozantinib (Cometriq™), vandetanib (Caprelsa®), Optionally, the therapeutic agent is a neoantigen. The therapeutic agent may be a cytokine such as interferons (INFs), interlcukins (ILs), or hematopoietic growth factors. The therapeutic agent may be INF-α, IL-2, Aldesleukin, IL-2, Erythropoietin, Granulocyte-macrophage colony-stimulating factor (GM-CSF) or granulocyte colony-stimulating factor. The therapeutic agent may be a targeted therapy such as toremifene (Fareston®), fulvestrant (Faslodex®), anastrozole (Arimidex®), exemestane (Aromasin®), letrozole (Femara®), ziv-aflibercept (Zaltrap®), Aiitretinoin (Panretin®), temsirolimus (Torisel®), Tretinoin (Vesanoid®), denileukin diftitox (Ontak®), vorinostat (Zoiinza®), romidepsin (Istodax®), bexarotene (Targretin®), pralatrexate (Foiotyn®), lenaliomide (Revlimid®), belinostat (Beleodag™) lenaliomide (Revlimid®), pomalidomide (Pomalyst®), Cabazitaxel (Jevtana®), enzaluiamide (Xtandi®), abiraterone acetate (Zytiga®), radium 223 chloride (Xofigo®), or everolimus (Afiniior®). Aditionally, the therapeutic agent may be an epigenetic targeted drug such as HDAC inhibitors, kinase inhibitors, DNA methyltransferase inhibitors, histone demethylase inhibitors, or histone methylation inhibitors. The epigenetic drugs may be Azacitidine (Vidaza), Decitabine (Dacogen), Vorinostat (Zoiinza), Romidepsin (Istodax), or Ruxolitinib (Jakafi). For prostate cancer treatment, a preferred chemotherapeutic agent with which anti-CTLA-4 can be combined is paclitaxel (TAXOL).


In one embodiment, provided herein is a kit comprising any antigen therapeutic described herein.


Where aspects or embodiments of the invention are described in terms of a Markush group or other grouping of alternatives, the present invention encompasses not only the entire group listed as a whole, but also each member of the group individually and all possible subgroups of the main group, and also the main group absent one or more of the group members. The present invention also envisages the explicit exclusion of one or more of any of the group members in the claimed invention.







DETAILED DESCRIPTION

Described herein are novel immunotherapeutic agents and uses thereof based on the discovery of non-mutated protein epitopes expressed in cancer cells. Accordingly, the invention described herein provides peptides, polynucleotides encoding the peptides, and peptide binding agents, that can be used, for example, to stimulate an immune response to a tumor associated antigen, to create an immunogenic composition or cancer vaccine for use in treating disease.


I. Definitions

To facilitate an understanding of the present invention, a number of terms and phrases are defined below.


“Non-mutated protein antigens” refer to antigens expressed in cancers either specifically or at a level higher than in non-cancer tissue. They include, but are not limited to, antigens of exogenous viruses, antigens of endogenous retroviruses and overexpressed antigens that do not comprise somatic mutations.


“Viral antigens” refer to antigens encoded by a exogenous virus.


“Retroviral antigens” refer to antigens encoded by an endogenous retroviral sequence.


“Non-mutated overexpressed antigens” refer to non-mutated antigens encoded by a genome of a cancer cell that are expressed at a level higher than in non-cancer tissue.


A “tumor-specific epitope” refers to an epitope that is either not expressed in non-cancer or germline cells but is found expressed in cancer cells, or that is expressed at a higher level in cancer cells than in non-cancer cells.


A “reference” can be used to correlate and compare the results obtained in the methods of the invention from a tumor specimen. Typically the “reference” may be obtained on the basis of one or more normal specimens, in particular specimens which are not affected by a cancer disease, either obtained from a patient or one or more different individuals, for example, healthy individuals, in particular individuals of the same species. A “reference” can be determined empirically by testing a sufficiently large number of normal specimens.


The term “mutation” refers to a change of or difference in the nucleic acid sequence (nucleotide substitution, addition or deletion) compared to a reference. A “somatic mutation” can occur in any of the cells of the body except the germ cells (sperm and egg) and therefore are not passed on to children. These alterations can (but do not always) cause cancer or other diseases. In some embodiments, a mutation is a non-synonymous mutation. The term “non-synonymous mutation” refers to a mutation, for example, a nucleotide substitution, which does result in an amino acid change such as an amino acid substitution in the translation product.


Throughout this disclosure, “binding data” results can be expressed in terms of “IC50.” IC50 is the concentration of the tested peptide in a binding assay at which 50% inhibition of binding of a labeled reference peptide is observed. Given the conditions in which the assays are run (i.e., limiting HLA protein and labeled reference peptide concentrations), these values approximate KD values. Assays for determining binding are well known in the art and are described in detail, for example, in PCT publications WO 94/20127 and WO 94/03205, and other publications such Sidney et al., Current Protocols in Immunology 18.3.1 (1998); Sidney, et al., J. Immunol. 154:247 (1995); and Sette, et al., Mol. Immunol. 31:813 (1994). Alternatively, binding can be expressed relative to binding by a reference standard peptide. For example, can be based on its IC50, relative to the IC50 of a reference standard peptide.


Binding can also be determined using other assay systems including those using: live cells (e.g., Ceppellini et al., Nature 339:392 (1989); Christnick et al., Nature 352:67 (1991); Busch et al., Int. Immunol. 2:443 (1990); Hill et al., J. Immunol. 147:189 (1991); del Guercio et al., J. Immunol. 154:685 (1995)), cell free systems using detergent lysates (e.g., Cerundolo et al., J. Immunol. 21:2069 (1991)), immobilized purified MHC (e.g., Hill et al., J. Immunol. 152, 2890 (1994); Marshall et al., J. Immunol. 152:4946 (1994)), ELISA systems (e.g., Reay et al., EMBO J. 11:2829 (1992)), surface plasmon resonance (e.g., Khilko et al., J. Biol. Chem. 268:15425 (1993)); high flux soluble phase assays (Hammer et al., J. Exp. Med. 180:2353 (1994)), and measurement of class I MHC stabilization or assembly (e.g., Ljunggren et al., Nature 346:476 (1990); Schumacher et al., Cell 62:563 (1990); Townsend et al., Cell 62:285 (1990); Parker et al., J. Immunol. 149:1896 (1992)).


“Cross-reactive binding” indicates that a peptide is bound by more than one HLA molecule; a synonym is degenerate binding.


The term “derived” when used to discuss an epitope is a synonym for “prepared.” A derived epitope can be isolated from a natural source, or it can be synthesized according to standard protocols in the art. Synthetic epitopes can comprise artificial amino acid residues “amino acid mimetics,” such as D isomers of natural occurring L amino acid residues or non-natural amino acid residues such as cyclohexylalanine. A derived or prepared epitope can be an analog of a native epitope.


A “diluent” includes sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is also a diluent for pharmaceutical compositions. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as diluents, for example, in injectable solutions.


An “epitope” is the collective features of a molecule, such as primary, secondary and tertiary peptide structure, and charge, that together form a site recognized by, for example, an immunoglobulin, T cell receptor, HLA molecule, or chimeric antigen receptor. Alternatively, an epitope can be defined as a set of amino acid residues which is involved in recognition by a particular immunoglobulin, or in the context of T cells, those residues necessary for recognition by T cell receptor proteins, chimeric antigen receptors, and/or Major Histocompatibility Complex (MHC) receptors. Epitopes can be prepared by isolation from a natural source, or they can be synthesized according to standard protocols in the art. Synthetic epitopes can comprise artificial amino acid residues, “amino acid mimetics,” such as D isomers of naturally-occurring L amino acid residues or non-naturally-occurring amino acid residues such as cyclohexylalanine. Throughout this disclosure, epitopes may be referred to in some cases as peptides or peptide epitopes.


It is to be appreciated that proteins or peptides that comprise an epitope or an analog described herein as well as additional amino acid(s) are still within the bounds of the invention. In certain embodiments, the peptide comprises a fragment of an antigen.


In certain embodiments, there is a limitation on the length of a peptide of the invention. The embodiment that is length-limited occurs when the protein or peptide comprising an epitope described herein comprises a region (i.e., a contiguous series of amino acid residues) having 100% identity with a native sequence. In order to avoid the definition of epitope from reading, e.g., on whole natural molecules, there is a limitation on the length of any region that has 100% identity with a native peptide sequence. Thus, for a peptide comprising an epitope described herein and a region with 100% identity with a native peptide sequence, the region with 100% identity to a native sequence generally has a length of: less than or equal to 600 amino acid residues, less than or equal to 500 amino acid residues, less than or equal to 400 amino acid residues, less than or equal to 250 amino acid residues, less than or equal to 100 amino acid residues, less than or equal to 85 amino acid residues, less than or equal to 75 amino acid residues, less than or equal to 65 amino acid residues, and less than or equal to 50 amino acid residues. In certain embodiments, an “epitope” described herein is comprised by a peptide having a region with less than 51 amino acid residues that has 100% identity to a native peptide sequence, in any increment down to 5 amino acid residues; for example 50, 49, 48, 47, 46, 45, 44, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid residues.


“Human Leukocyte Antigen” or “HLA” is a human class I or class II Major Histocompatibility Complex (MHC) protein (see, e.g., Stites, et al., IMMUNOLOGY, 8TH ED., Lange Publishing, Los Altos, Calif. (1994).


An “HLA supertype or HLA family”, as used herein, describes sets of HLA molecules grouped on the basis of shared peptide-binding specificities. HLA class I molecules that share somewhat similar binding affinity for peptides bearing certain amino acid motifs are grouped into such HLA supertypes. The terms HLA superfamily, HLA supertype family, HLA family, and HLA xx-like molecules (where “xx” denotes a particular HLA type), are synonyms.


The terms “identical” or percent “identity,” in the context of two or more peptide sequences or antigen fragments, refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues that are the same, when compared and aligned for maximum correspondence over a comparison window, as measured using a sequence comparison algorithm or by manual alignment and visual inspection.


An “immunogenic” peptide or an “immunogenic” epitope or “peptide epitope” is a peptide that comprises an allele-specific motif such that the peptide will bind an HLA molecule and induce a cell-mediated or humoral response, for example, cytotoxic T lymphocyte (CTL), helper T lymphocyte (HTL) and/or B lymphocyte response. Thus, immunogenic peptides described herein are capable of binding to an appropriate HLA molecule and thereafter inducing a CTL (cytotoxic) response, or a HTL (and humoral) response, to the peptide.


As used herein, a “chimeric antigen receptor” or “CAR” refers to an antigen binding protein in that includes an immunoglobulin antigen binding domain (e.g., an immunoglobulin variable domain) and a T cell receptor (TCR) constant domain. As used herein, a “constant domain” of a TCR polypeptide includes a membrane-proximal TCR constant domain, and may also include a TCR transmembrane domain and/or a TCR cytoplasmic tail. For example, in some embodiments, the CAR is a dimer that includes a first polypeptide comprising a immunoglobulin heavy chain variable domain linked to a TCR-beta constant domain and a second polypeptide comprising an immunoglobulin light chain variable domain (e.g., a κ or λ variable domain) linked to a TCRα constant domain. In some embodiments, the CAR is a dimer that includes a first polypeptide comprising a immunoglobulin heavy chain variable domain linked to a TCRα constant domain and a second polypeptide comprising an immunoglobulin light chain variable domain (e.g., a κ or λ variable domain) linked to a TCRβ constant domain.


The phrases “isolated” or “biologically pure” refer to material which is substantially or essentially free from components which normally accompany the material as it is found in its native state. Thus, isolated peptides described herein do not contain some or all of the materials normally associated with the peptides in their in situ environment. An “isolated” epitope refers to an epitope that does not include the whole sequence of the antigen from which the epitope was derived. Typically the “isolated” epitope does not have attached thereto additional amino acid residues that result in a sequence that has 100% identity over the entire length of a native sequence. The native sequence can be a sequence such as a tumor-associated antigen from which the epitope is derived. Thus, the term “isolated” means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide or peptide present in a living animal is not isolated, but the same polynucleotide or peptide, separated from some or all of the coexisting materials in the natural system, is isolated. Such a polynucleotide could be part of a vector, and/or such a polynucleotide or peptide could be part of a composition, and still be “isolated” in that such vector or composition is not part of its natural environment. Isolated RNA molecules include in vivo or in vitro RNA transcripts of the DNA molecules described herein, and further include such molecules produced synthetically.


“Major Histocompatibility Complex” or “MHC” is a cluster of genes that plays a role in control of the cellular interactions responsible for physiologic immune responses. In humans, the MHC complex is also known as the human leukocyte antigen (HLA) complex. For a detailed description of the MHC and HLA complexes, see, Paul, FUNDAMENTAL IMMUNOLOGY, 3.sup.RD ED., Raven Press, New York (1993).


A “native” or a “wild type” sequence refers to a sequence found in nature. Such a sequence can comprise a longer sequence in nature.


A “T-cell epitope” is to be understood as meaning a peptide sequence which can be bound by the MHC molecules of class I or II in the form of a peptide-presenting MHC molecule or MHC complex and then, in this form, be recognized and bound by cytotoxic T-lymphocytes or T-helper cells, respectively.


A “receptor” is to be understood as meaning a biological molecule or a molecule grouping capable of binding a ligand. A receptor may serve, to transmit information in a cell, a cell formation or an organism. The receptor comprises at least one receptor unit, for example, where each receptor unit may consist of a protein molecule. The receptor has a structure which complements that of a ligand and may complex the ligand as a binding partner. The information is transmitted in particular by conformational changes of the receptor following complexation of the ligand on the surface of a cell. In some embodiments, a receptor is to be understood as meaning in particular proteins of MHC classes I and II capable of forming a receptor/ligand complex with a ligand, in particular a peptide or peptide fragment of suitable length.


A “ligand” is to be understood as meaning a molecule which has a structure complementary to that of a receptor and is capable of forming a complex with this receptor. In some embodiments, a ligand is to be understood as meaning a peptide or peptide fragment which has a suitable length and suitable binding motifs in its amino acid sequence, so that the peptide or peptide fragment is capable of forming a complex with proteins of MHC class I or MHC class II.


In some embodiments, a “receptor/ligand complex” is also to be understood as meaning a “receptor/peptide complex” or “receptor/peptide fragment complex”, including a peptide- or peptide fragment-presenting MHC molecule of class I or of class II.


“Proteins or molecules of the major histocompatibility complex (MHC)”, “MHC molecules”, “MHC proteins” or “HLA proteins” are to be understood as meaning proteins capable of binding peptides resulting from the proteolytic cleavage of protein antigens and representing potential lymphocyte epitopes, (e.g., T cell epitope and B cell epitope) transporting them to the cell surface and presenting them there to specific cells, in particular cytotoxic T-lymphocytes, T-helper cells, or B cells. The major histocompatibility complex in the genome comprises the genetic region whose gene products expressed on the cell surface are important for binding and presenting endogenous and/or foreign antigens and thus for regulating immunological processes. The major histocompatibility complex is classified into two gene groups coding for different proteins, namely molecules of MHC class I and molecules of MHC class II. The cellular biology and the expression patterns of the two MHC classes are adapted to these different roles.


The terms “peptide” and “peptide epitope” are used interchangeably with “oligopeptide” in the present specification to designate a series of residues connected one to the other, typically by peptide bonds between the α-amino and carboxyl groups of adjacent amino acid residues.


“Synthetic peptide” refers to a peptide that is obtained from a non-natural source, e.g., is man-made. Such peptides can be produced using such methods as chemical synthesis or recombinant DNA technology. “Synthetic peptides” include “fusion proteins.”


A “PanDR binding” peptide, a “PanDR binding epitope” is a member of a family of molecules that binds more than one HLA class II DR molecule.


“Pharmaceutically acceptable” refers to a generally non-toxic, inert, and/or physiologically compatible composition or component of a composition.


A “pharmaceutical excipient” or “excipient” comprises a material such as an adjuvant, a carrier, pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservatives, and the like. A “pharmaceutical excipient” is an excipient which is pharmaceutically acceptable.


The term “motif” refers to a pattern of residues in an amino acid sequence of defined length, for example, a peptide of less than about 15 amino acid residues in length, or less than about 13 amino acid residues in length, for example, from about 8 to about 13 amino acid residues (e.g., 8, 9, 10, 11, 12, or 13) for a class I HLA motif and from about 6 to about 25 amino acid residues (e.g., 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) for a class II HLA motif, which is recognized by a particular HLA molecule. Motifs are typically different for each HLA protein encoded by a given human HLA allele. These motifs differ in their pattern of the primary and secondary anchor residues. In some embodiments, an MHC class I motif identifies a peptide of 9, 10, or 11 amino acid residues in length.


A “supermotif” is a peptide binding specificity shared by HLA molecules encoded by two or more HLA alleles. In some embodiments, a supermotif-bearing peptide described herein is recognized with high or intermediate affinity (as defined herein) by two or more HLA antigens.


The term “naturally occurring” as used herein refers to the fact that an object can be found in nature. For example, a peptide or nucleic acid that is present in an organism (including viruses) and can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally occurring.


According to the invention, the term “vaccine” relates to a pharmaceutical preparation (pharmaceutical composition) or product that upon administration induces an immune response, for example, a cellular or humoral immune response, which recognizes and attacks a pathogen or a diseased cell such as a cancer cell. A vaccine may be used for the prevention or treatment of a disease. The term “individualized cancer vaccine” or “personalized cancer vaccine” concerns a particular cancer patient and means that a cancer vaccine is adapted to the needs or special circumstances of an individual cancer patient.


A “protective immune response” or “therapeutic immune response” refers to a CTL and/or an HTL response to an antigen derived from an pathogenic antigen (e.g., a tumor antigen), which in some way prevents or at least partially arrests disease symptoms, side effects or progression. The immune response can also include an antibody response which has been facilitated by the stimulation of helper T cells.


“Antigen processing” or “processing” refers to the degradation of a polypeptide or antigen into procession products, which are fragments of said polypeptide or antigen (e.g., the degradation of a polypeptide into peptides) and the association of one or more of these fragments (e.g., via binding) with MHC molecules for presentation by cells, for example, antigen presenting cells, to specific T cells.


“Antigen presenting cells” (APC) are cells which present peptide fragments of protein antigens in association with MHC molecules on their cell surface. Some APCs may activate antigen specific T cells. Professional antigen-presenting cells are very efficient at internalizing antigen, either by phagocytosis or by receptor-mediated endocytosis, and then displaying a fragment of the antigen, bound to a class II MHC molecule, on their membrane. The T cell recognizes and interacts with the antigen-class II MHC molecule complex on the membrane of the antigen presenting cell. An additional co-stimulatory signal is then produced by the antigen presenting cell, leading to activation of the T cell. The expression of co-stimulatory molecules is a defining feature of professional antigen-presenting cells.


The main types of professional antigen-presenting cells are dendritic cells, which have the broadest range of antigen presentation, and are probably the most important antigen presenting cells, macrophages, B-cells, and certain activated epithelial cells.


Dendritic cells (DCs) are leukocyte populations that present antigens captured in peripheral tissues to T cells via both MHC class II and I antigen presentation pathways. It is well known that dendritic cells are potent inducers of immune responses and the activation of these cells is a critical step for the induction of antitumoral immunity.


Dendritic cells are conveniently categorized as “immature” and “mature” cells, which can be used as a simple way to discriminate between two well characterized phenotypes. However, this nomenclature should not be construed to exclude all possible intermediate stages of differentiation.


Immature dendritic cells are characterized as antigen presenting cells with a high capacity for antigen uptake and processing, which correlates with the high expression of Fey receptor and mannose receptor. The mature phenotype is typically characterized by a lower expression of these markers, but a high expression of cell surface molecules responsible for T cell activation such as class I and class II MHC, adhesion molecules (e. g. CD54 and CD1 1) and costimulatory molecules (e. g., CD40, CD80, CD86 and 4-1 BB).


The term “residue” refers to an amino acid residue or amino acid mimetic residue incorporated into a peptide or protein by an amide bond or amide bond mimetic, or nucleic acid (DNA or RNA) that encodes the amino acid or amino acid mimetic.


The nomenclature used to describe peptides or proteins follows the conventional practice wherein the amino group is presented to the left (the amino- or N-terminus) and the carboxyl group to the right (the carboxy- or C-terminus) of each amino acid residue. When amino acid residue positions are referred to in a peptide epitope they are numbered in an amino to carboxyl direction with position one being the residue located at the amino terminal end of the epitope, or the peptide or protein of which it can be a part.


In the formulae representing selected specific embodiments of the present invention, the amino- and carboxyl-terminal groups, although not specifically shown, are in the form they would assume at physiologic pH values, unless otherwise specified. In the amino acid structure formulae, each residue is generally represented by standard three letter or single letter designations. The L-form of an amino acid residue is represented by a capital single letter or a capital first letter of a three-letter symbol, and the D-form for those amino acid residues having D-forms is represented by a lower case single letter or a lower case three letter symbol. However, when three letter symbols or full names are used without capitals, they can refer to L amino acid residues. Glycine has no asymmetric carbon atom and is simply referred to as “Gly” or “G”. The amino acid sequences of peptides set forth herein are generally designated using the standard single letter symbol. (A, Alanine; C, Cysteine; D, Aspartic Acid; E, Glutamic Acid; F, Phenylalanine; G, Glycine; H, Histidine; I, Isoleucine; K, Lysine; L, Leucine; M, Methionine; N, Asparagine; P, Proline; Q, Glutamine; R, Arginine; S, Serine; T, Threonine; V, Valine; W, Tryptophan; and Y, Tyrosine.)


The terms “polynucleotide” and “nucleic acid” are used interchangeably herein and refer to polymers of nucleotides of any length, and include DNA and RNA, for example, mRNA. The nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase. In some embodiments, the polynucleotide and nucleic acid can be in vitro transcribed mRNA. In some embodiments, the polynucleotide that is administered is mRNA.


The terms “identical” or percent “identity” in the context of two or more nucleic acids or polypeptides, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity. The percent identity can be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software that can be used to obtain alignments of amino acid or nucleotide sequences are well-known in the art. These include, but are not limited to, BLAST, ALIGN, Megalign, BestFit, GCG Wisconsin Package, and variations thereof. In some embodiments, two nucleic acids or polypeptides described herein are substantially identical, meaning they have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and in some embodiments at least 95%, 96%, 97%, 98%, 99% nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using a sequence comparison algorithm or by visual inspection. In some embodiments, identity exists over a region of the sequences that is at least about 10, at least about 20, at least about 40-60 residues, at least about 60-80 residues in length or any integral value 2 between. In some embodiments, identity exists over a longer region than 60-80 residues, such as at least about 80-100 residues, and in some embodiments the sequences are substantially identical over the full length of the sequences being compared, such as the coding region of a nucleotide sequence.


A “conservative amino acid substitution” is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). For example, substitution of a phenylalanine for a tyrosine is a conservative substitution. Methods of identifying nucleotide and amino acid conservative substitutions which do not eliminate peptide function are well-known in the art.


The term “vector” as used herein means a construct, which is capable of delivering, and usually expressing, one or more gene(s) or sequence(s) of interest in a host cell. Examples of vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid, or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, and DNA or RNA expression vectors encapsulated in liposomes.


A polypeptide, antibody, polynucleotide, vector, cell, or composition which is “isolated” is a polypeptide, antibody, polynucleotide, vector, cell, or composition which is in a form not found in nature. Isolated polypeptides, antibodies, polynucleotides, vectors, cells, or compositions include those which have been purified to a degree that they are no longer in a form in which they are found in nature. In some embodiments, a polypeptide, antibody, polynucleotide, vector, cell, or composition which is isolated is substantially pure. In one embodiment, an “isolated polynucleotide” encompasses a PCR or quantitative PCR reaction comprising the polynucleotide amplified in the PCR or quantitative PCR reaction.


The term “substantially pure” as used herein refers to material which is at least 50% pure (i.e., free from contaminants), at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure.


The term “subject” refers to any animal (e.g., a mammal), including, but not limited to, humans, non-human primates, canines, felines, rodents, and the like, which is to be the recipient of a particular treatment. Typically, the terms “subject” and “patient” are used interchangeably herein in reference to a human subject.


The terms “effective amount” or “therapeutically effective amount” or “therapeutic effect” refer to an amount of a therapeutic effective to “treat” a disease or disorder in a subject or mammal. The therapeutically effective amount of a drug has a therapeutic effect and as such can prevent the development of a disease or disorder; slow down the development of a disease or disorder; slow down the progression of a disease or disorder; relieve to some extent one or more of the symptoms associated with a disease or disorder; reduce morbidity and mortality; improve quality of life; or a combination of such effects.


The terms “treating” or “treatment” or “to treat” or “alleviating” or “to alleviate” refer to both 1) therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder and 2) prophylactic or preventative measures that prevent or slow the development of a targeted pathologic condition or disorder. Thus those in need of treatment include those already with the disorder; those prone to have the disorder; and those in whom the disorder is to be prevented.


As used in the present disclosure and claims, the singular forms “a”, “an” and “the” include plural forms unless the context clearly dictates otherwise.


It is understood that terms such as “comprises”, “comprised”, “comprising” and the like can have the meaning attributed to it in U.S. Patent law; e.g., they can mean “includes”, “included”, “including”, and the like; and that terms such as “consisting essentially of” and “consists essentially of” have the meaning ascribed to them in U.S. Patent law, e.g., they allow for elements not explicitly recited, but exclude elements that are found in the prior art or that affect a basic or novel characteristic of the invention. Nothing herein is intended as a promise.


The term “and/or” as used in a phrase such as “A and/or B” herein is intended to include both A and B; A or B; A (alone); and B (alone). Likewise, the term “and/or” as used in a phrase such as “A, B, and/or C” is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).


II. Non-Mutated Protein Antigens Expressed in Cancer Cells

Applicants have discovered antigens expressed by cancer cells encoded by the following genes: ERVH-2 matrix protein, ERVH-2 gag, ERVH48-1 coat protein, ERVH48-1 syncytin, ERVE-4 reverse transcriptase, ERVK-5 gag, env, pol protein, and ERVI-1 envelope protein.


Applicants have discovered antigens expressed by cancer encoded by the following genes: TYR, MAGEC1, MAGEA10, MAGEB17, MAGEA4, MABEB16, MAGEA1, MAGEA8, MAGEB4, CT45A5, ALPPL2, MMP13, CTAG1B, DCT, CLDN6, MLANA, AFP, DKK4, ASCL2, GAGE1, GAGE10, SLC45A2, PAGES, PAGE2, and PMEL.


Applicants have discovered antigens expressed by cancer encoded by the following genes: HPV-16, E6, HPV-16 E7, EBV LF2, EBV BALF5, EBV RPMS1, EBV A73, EBV BALF4, EBV BALF3, and EBV BARF0.


Non-Mutated Protein Epitope Polypeptides

In aspects, the invention provides isolated peptides that comprise a non-mutated protein epitope expressed in a cancer cell. In some embodiments, the non-mutated protein epitope is a retroviral antigen. In some embodiments, the non-mutated protein epitope is a non-mutated overexpressed antigen. In some embodiments, the non-mutated protein epitope is a viral antigen.


In aspects, the invention provides an isolated peptide that comprises a peptide from Tables 1-6. The term “peptide” is used in the present specification to designate a series of residues, typically L-amino acids, connected one to the other, typically by peptide bonds between the α-amino and carboxyl groups of adjacent amino acids. Similarly, the term “polypeptide” is used in the present specification to designate a series of residues, e.g., L-amino acids, connected one to the other, typically by peptide bonds between the α-amino and carboxyl groups of adjacent amino acids. The polypeptides or peptides can be a variety of lengths, either in their neutral (uncharged) forms or in forms which are salts, and either free of modifications such as glycosylation, side chain oxidation, or phosphorylation or containing these modifications, subject to the condition that the modification not destroy the biological activity of the polypeptides as herein described.


In some embodiments, sequencing methods are used to identify tumor specific epitopes. Any suitable sequencing method can be used according to the invention, for example, Next Generation Sequencing (NGS) technologies. Third Generation Sequencing methods might substitute for the NGS technology in the future to speed up the sequencing step of the method. For clarification purposes: the terms “Next Generation Sequencing” or “NGS” in the context of the present invention mean all novel high throughput sequencing technologies which, in contrast to the “conventional” sequencing methodology known as Sanger chemistry, read nucleic acid templates randomly in parallel along the entire genome by breaking the entire genome into small pieces. Such NGS technologies (also known as massively parallel sequencing technologies) are able to deliver nucleic acid sequence information of a whole genome, exome, transcriptome (all transcribed sequences of a genome) or methylome (all methylated sequences of a genome) in very short time periods, e.g. within 1-2 weeks, for example, within 1-7 days or within less than 24 hours and allow, in principle, single cell sequencing approaches. Multiple NGS platforms which are commercially available or which are mentioned in the literature can be used in the context of the invention e.g. those described in detail in WO 2012/159643.


In certain embodiments a non-mutated protein epitope peptide described herein molecule can comprise, but is not limited to, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39, about 40, about 41, about 42, about 43, about 44, about 45, about 46, about 47, about 48, about 49, about 50, about 60, about 70, about 80, about 90, about 100, about 110, about 120 or greater amino acid residues, and any range derivable therein. In specific embodiments, a non-mutated protein epitope peptide molecule is equal to or less than 100 amino acids.


In some embodiments, non-mutated protein epitope peptides and polypeptides described herein for MHC Class I are 13 residues or less in length and usually consist of between about 8 and about 11 residues, particularly 9 or 10 residues. In some embodiments, non-mutated protein epitope peptides and polypeptides described herein for MHC Class II are 9-24 residues in length.


A longer non-mutated protein epitope peptide can be designed in several ways. In some embodiments, when HLA-binding peptides are predicted or known, a longer non-mutated protein epitope peptide could consist of (1) individual binding peptides with extensions of 2-5 amino acids toward the N- and C-terminus of each corresponding peptide; or (2) a concatenation of some or all of the binding peptides with extended sequences for each. In some embodiments, use of a longer peptide is presumed to allow for endogenous processing by patient cells and can lead to more effective antigen presentation and induction of T cell responses. In some embodiments, two or more peptides can be used, where the peptides overlap and are tiled over the long non-mutated protein epitope peptide.


In some embodiments, the non-mutated protein epitope peptides and polypeptides bind an HLA protein (e.g., HLA class I or HLA class II). In specific embodiments the non-mutated protein epitope peptide or polypeptide has an IC50 of at least less than 5000 nM, at least less than 500 nM, at least less than 100 nM, at least less than 50 nM or less.


In some embodiments, a non-mutated protein epitope peptide described herein can comprise carriers such as those well known in the art, e.g., thyroglobulin, albumins such as human serum albumin, tetanus toxoid, polyamino acid residues such as poly L-lysine, poly L-glutamic acid, influenza virus proteins, hepatitis B virus core protein, and the like.


In some embodiments, a non-mutated protein epitope peptide described herein can be modified by terminal-NH2 acylation, e.g., by alkanoyl (C1-C20) or thioglycolyl acetylation, terminal-carboxyl amidation, e.g., ammonia, methylamine, etc. In some embodiments these modifications can provide sites for linking to a support or other molecule.


In some embodiments, a non-mutated protein epitope peptide described herein can contain modifications such as but not limited to glycosylation, side chain oxidation, biotinylation, phosphorylation, addition of a surface active material, e.g. a lipid, or can be chemically modified, e.g., acetylation, etc. Moreover, bonds in the peptide can be other than peptide bonds, e.g., covalent bonds, ester or ether bonds, disulfide bonds, hydrogen bonds, ionic bonds, etc.


In some embodiments, a non-mutated protein epitope peptide described herein can contain substitutions to modify a physical property (e.g., stability or solubility) of the resulting peptide. For example, non-mutated protein epitope peptides can be modified by the substitution of a cysteine (C) with α-amino butyric acid (“B”). Due to its chemical nature, cysteine has the propensity to form disulfide bridges and sufficiently alter the peptide structurally so as to reduce binding capacity. Substituting α-amino butyric acid for C not only alleviates this problem, but actually improves binding and crossbinding capability in certain instances. Substitution of cysteine with α-amino butyric acid can occur at any residue of a non-mutated protein epitope peptide, e.g., at either anchor or non-anchor positions of an epitope or analog within a peptide, or at other positions of a peptide.


In some embodiments, a non-mutated protein epitope peptide described herein can comprise amino acid mimetics or unnatural amino acid residues, e.g. D- or L-naphylalanine; D- or L-phenylglycine; D- or L-2-thieneylalanine; D- or L-1, -2, 3-, or 4-pyreneylalanine; D- or L-3 thieneylalanine; D- or L-(2-pyridinyl)-alanine; D- or L-(3-pyridinyl)-alanine; D- or L-(2-pyrazinyl)-alanine; D- or L-(4-isopropyl)-phenylglycine; D-(trifluoromethyl)-phenylglycine; D-(trifluoro-methyl)-phenylalanine; D-.rho.-fluorophenylalanine; D- or L-.rho.-biphenyl-phenylalanine; D- or L-p-methoxybiphenylphenylalanine; D- or L-2-indole(allyl)alanines; and, D- or L-alkylalanines, where the alkyl group can be a substituted or unsubstituted methyl, ethyl, propyl, hexyl, butyl, pentyl, isopropyl, iso-butyl, sec-isotyl, iso-pentyl, or a non-acidic amino acid residues. Aromatic rings of a non-natural amino acid include, e.g., thiazolyl, thiophenyl, pyrazolyl, benzimidazolyl, naphthyl, furanyl, pyrrolyl, and pyridyl aromatic rings. Modified peptides that have various amino acid mimetics or unnatural amino acid residues are particularly useful, as they tend to manifest increased stability in vivo. Such peptides can also possess improved shelf-life or manufacturing properties.


Peptide stability can be assayed in a number of ways. For instance, peptidases and various biological media, such as human plasma and serum, have been used to test stability. See, e.g., Verhoef, et al., Eur. J. Drug Metab. Pharmacokinetics 11:291 (1986). Half-life of the peptides described herein is conveniently determined using a 25% human serum (v/v) assay. The protocol is as follows: pooled human serum (Type AB, non-heat inactivated) is dilapidated by centrifugation before use. The serum is then diluted to 25% with RPMI-1640 or another suitable tissue culture medium. At predetermined time intervals, a small amount of reaction solution is removed and added to either 6% aqueous trichloroacetic acid (TCA) or ethanol. The cloudy reaction sample is cooled (4° C.) for 15 minutes and then spun to pellet the precipitated serum proteins. The presence of the peptides is then determined by reversed-phase HPLC using stability-specific chromatography conditions.


In some embodiments, a non-mutated protein epitope peptide described herein can be in solution, lyophilized, or can be in crystal form.


In some embodiments, a non-mutated protein epitope peptide described herein can be prepared synthetically, by recombinant DNA technology or chemical synthesis, or can be isolated from natural sources such as native tumors or pathogenic organisms. Epitopes can be synthesized individually or joined directly or indirectly in a peptide. Although a non-mutated protein epitope peptide described herein will be substantially free of other naturally occurring host cell proteins and fragments thereof, in some embodiments the peptide can be synthetically conjugated to be joined to native fragments or particles.


In some embodiments, a non-mutated protein epitope peptide described herein can be prepared in a wide variety of ways. In some embodiments, the peptides can be synthesized in solution or on a solid support according to conventional techniques. Various automatic synthesizers are commercially available and can be used according to known protocols. (See, for example, Stewart & Young, SOLID PHASE PEPTIDE SYNTHESIS, 2D. ED., Pierce Chemical Co., 1984). Further, individual peptides can be joined using chemical ligation to produce larger peptides that are still within the bounds of the invention.


Alternatively, recombinant DNA technology can be employed wherein a nucleotide sequence which encodes a peptide inserted into an expression vector, transformed or transfected into an appropriate host cell and cultivated under conditions suitable for expression. These procedures are generally known in the art, as described generally in Sambrook et al., MOLECULAR CLONING, A LABORATORY MANUAL, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (1989). Thus, recombinant peptides, which comprise or consist of one or more epitopes described herein, can be used to present the appropriate T cell epitope.


In one aspect, the invention described herein also provides compositions comprising one, at least two, or more than two non-mutated protein epitope peptides. In some embodiments a composition described herein contains at least two distinct peptides. In some embodiments, the at least two distinct peptides are derived from the same polypeptide. By distinct polypeptides is meant that the peptide vary by length, amino acid sequence or both. The peptides are derived from any polypeptide known to or have been found to contain a tumor specific epitope.


Non-Mutated Protein Epitope Polynucleotides

Polynucleotides encoding each of the peptides described herein are also part of the invention. As appreciated by one of ordinary skill in the art, various nucleic acids will encode the same peptide due to the redundancy of the genetic code. Each of these nucleic acids falls within the scope of the present invention. This embodiment of the invention comprises DNA and RNA, for example, mRNA, and in certain embodiments a combination of DNA and RNA. In one embodiment, the mRNA is a self-amplifying mRNA. (Brito et al., Adv. Genet. 2015; 89:179-233). It is to be appreciated that any polynucleotide that encodes a peptide described herein falls within the scope of this invention.


The term “RNA” includes and in some embodiments relates to “mRNA”. The term “mRNA” means “messenger-RNA” and relates to a “transcript” which is generated by using a DNA template and encodes a peptide or polypeptide. Typically, an mRNA comprises a 5′-UTR, a protein coding region, and a 3′-UTR. mRNA only possesses limited half-life in cells and in vitro. In one embodiment, the mRNA is self-amplifying mRNA. In the context of the present invention, mRNA may be generated by in vitro transcription from a DNA template. The in vitro transcription methodology is known to the skilled person. For example, there is a variety of in vitro transcription kits commercially available.


The stability and translation efficiency of RNA may be modified as required. For example, RNA may be stabilized and its translation increased by one or more modifications having a stabilizing effects and/or increasing translation efficiency of RNA. Such modifications are described, for example, in PCT/EP2006/009448 incorporated herein by reference. In order to increase expression of the RNA used according to the present invention, it may be modified within the coding region, i.e. the sequence encoding the expressed peptide or protein, without altering the sequence of the expressed peptide or protein, so as to increase the GC-content to increase mRNA stability and to perform a codon optimization and, thus, enhance translation in cells.


The term “modification” in the context of the RNA used in the present invention includes any modification of an RNA which is not naturally present in said RNA. In one embodiment of the invention, the RNA used according to the invention does not have uncapped 5′-triphosphates. Removal of such uncapped 5′-triphosphates can be achieved by treating RNA with a phosphatase. The RNA according to the invention may have modified ribonucleotides in order to increase its stability and/or decrease cytotoxicity. For example, in one embodiment, in the RNA used according to the invention 5-methylcytidine is substituted partially or completely, for example, completely, for cytidine. Alternatively or additionally, in one embodiment, in the RNA used according to the invention pseudouridine is substituted partially or completely, for example, completely, for uridine.


In one embodiment the term “modification” relates to providing an RNA with a 5′-cap or 5′-cap analog. The term “5′-cap” refers to a cap structure found on the 5′-end of an mRNA molecule and generally consists of a guanosine nucleotide connected to the mRNA via an unusual 5′ to 5′ triphosphate linkage. In one embodiment, this guanosine is methylated at the 7-position. The term “conventional 5′-cap” refers to a naturally occurring RNA 5′-cap, to the 7-methylguanosine cap (m G). In the context of the present invention, the term “5′-cap” includes a 5′-cap analog that resembles the RNA cap structure and is modified to possess the ability to stabilize RNA and/or enhance translation of RNA if attached thereto, in vivo and/or in a cell.


In certain embodiments, an mRNA encoding a non-mutated protein epitope is administered to a subject in need thereof. In one embodiment, the invention provides RNA, oligoribonucleotide, and polyribonucleotide molecules comprising a modified nucleoside, gene therapy vectors comprising same, gene therapy methods and gene transcription silencing methods comprising same. In one embodiment, the mRNA to be administered comprises at least one modified nucleoside.


The polynucleotides encoding peptides described herein can be synthesized by chemical techniques, for example, the phosphotriester method of Matteucci, et al., J. Am. Chem. Soc. 103:3185 (1981). Polynucleotides encoding peptides comprising or consisting of an analog can be made simply by substituting the appropriate and desired nucleic acid base(s) for those that encode the native epitope.


A large number of vectors and host systems suitable for producing and administering a non-mutated protein epitope peptide described herein are known to those of skill in the art, and are commercially available. The following vectors are provided by way of example. Bacterial: pQE70, pQE60, pQE-9 (Qiagen), pBS, pD10, phagescript, psiX174, pBluescript SK, pbsks, pNH8A, pNH16a, pNH18A, pNH46A (Stratagene); ptrc99a, pKK223-3, pKK233-3, pDR540, pRITS (Pharmacia); pCR (Invitrogen). Eukaryotic: pWLNEO, pSV2CAT, pOG44, pXT1, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia); p75.6 (Valentis); pCEP (Invitrogen); pCEI (Epimmune). However, any other plasmid or vector can be used as long as it is replicable and viable in the host.


As representative examples of appropriate hosts, there can be mentioned: bacterial cells, such as E. coli, Bacillus subtilis, Salmonella typhimurium and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus; fungal cells, such as yeast; insect cells such as Drosophila and Sf9; animal cells such as COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell 23:175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CHO, HeLa and BHK cell lines or Bowes melanoma; plant cells, etc. The selection of an appropriate host is deemed to be within the scope of those skilled in the art from the teachings herein.


Thus, the present disclosure is also directed to vectors, and expression vectors useful for the production and administration of the non-mutated protein epitope peptides described herein, and to host cells comprising such vectors.


Host cells are genetically engineered (transduced or transformed or transfected) with the vectors which can be, for example, a cloning vector or an expression vector. The vector can be, for example, in the form of a plasmid, a viral particle, a phage, etc. The engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the polynucleotides. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.


For expression of the non-mutated protein epitope peptides described herein, the coding sequence will be provided operably linked start and stop codons, promoter and terminator regions, and in some embodiments, and a replication system to provide an expression vector for expression in the desired cellular host. For example, promoter sequences compatible with bacterial hosts are provided in plasmids containing convenient restriction sites for insertion of the desired coding sequence. The resulting expression vectors are transformed into suitable bacterial hosts.


Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae TRP1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence. Such promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), acid phosphatase, or heat shock proteins, among others. The heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and in some embodiments, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium. Optionally, the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.


Yeast, insect or mammalian cell hosts can also be used, employing suitable vectors and control sequences. Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell 23:175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CHO, HeLa and BHK cell lines. Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5′ flanking nontranscribed sequences. Such promoters can also be derived from viral sources, such as, e.g., human cytomegalovirus (CMV-IE promoter) or herpes simplex virus type-1 (HSV TK promoter). Nucleic acid sequences derived from the SV40 splice, and polyadenylation sites can be used to provide the nontranscribed genetic elements.


Polynucleotides encoding non-mutated protein epitope peptides described herein can also comprise a ubiquitination signal sequence, and/or a targeting sequence such as an endoplasmic reticulum (ER) signal sequence to facilitate movement of the resulting peptide into the endoplasmic reticulum.


Polynucleotides described herein can be administered and expressed in human cells (e.g., immune cells, including dendritic cells). A human codon usage table can be used to guide the codon choice for each amino acid. Such polynucleotides comprise spacer amino acid residues between epitopes and/or analogs, such as those described above, or can comprise naturally-occurring flanking sequences adjacent to the epitopes and/or analogs (and/or CTL, HTL, and B cell epitopes).


In some embodiments, a non-mutated protein epitope peptide described herein can also be administered/expressed by viral or bacterial vectors. Examples of expression vectors include attenuated viral hosts, such as vaccinia or fowlpox. As an example of this approach, vaccinia virus is used as a vector to express nucleotide sequences that encode the non-mutated protein epitope peptides described herein. Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Pat. No. 4,722,848. Another vector is BCG (Bacille Calmette Guerin). BCG vectors are described by Stover et al., Nature 351:456-460 (1991). A wide variety of other vectors useful for therapeutic administration or immunization of the non-mutated protein epitope polypeptides described herein, e.g. adeno and adeno-associated virus vectors, retroviral vectors, Salmonella typhi vectors, detoxified anthrax toxin vectors, Sendai virus vectors, poxvirus vectors, canarypox vectors, and fowlpox vectors, and the like, will be apparent to those skilled in the art from the description herein. In some embodiments, the vector is Modified Vaccinia Ankara (VA) (e.g. Bavarian Noridic (MVA-BN)).


Standard regulatory sequences well known to those of skill in the art can be included in the vector to ensure expression in the human target cells. Several vector elements are desirable: a promoter with a downstream cloning site for polynucleotide, e.g., minigene insertion; a polyadenylation signal for efficient transcription termination; an E. coli origin of replication; and an E. coli selectable marker (e.g. ampicillin or kanamycin resistance). Numerous promoters can be used for this purpose, e.g., the human cytomegalovirus (hCMV) promoter. See, e.g., U.S. Pat. Nos. 5,580,859 and 5,589,466 for other suitable promoter sequences. In some embodiments, the promoter is the CMV-IE promoter.


Polynucleotides described herein can comprise one or more synthetic or naturally-occurring introns in the transcribed region. The inclusion of mRNA stabilization sequences and sequences for replication in mammalian cells can also be considered for increasing polynucleotide expression.


In addition, a polynucleotide described herein can comprise immunostimulatory sequences (ISSs or CpGs). These sequences can be included in the vector, outside the polynucleotide coding sequence to enhance immunogenicity.


Non-Mutated Protein Epitope Binding Peptides

In certain embodiments, the present invention provides a binding protein (e.g., an antibody or antigen-binding fragment thereof), or a T cell receptor (TCR), or a chimeric antigen receptor (CAR) capable of binding with a high affinity to a non-mutated protein epitope peptide:human leukocyte antigen (HLA) complex. In some embodiments, the present invention provides a CAR that is capable of binding with a high affinity to a non-mutated protein epitope peptide derived from the extracellular domain of a protein. In certain embodiments, an antigen-specific binding protein or TCR or CAR as described herein includes variant polypeptide species that have one or more amino acid substitutions, insertions, or deletions in the native amino acid sequence, provided that the binding protein retains or substantially retains its specific binding function. Conservative substitutions of amino acids are well known and may occur naturally or may be introduced when the binding protein or TCR is recombinantly produced. Amino acid substitutions, deletions, and additions may be introduced into a protein using mutagenesis methods known in the art (see, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, 3d ed., Cold Spring Harbor Laboratory Press, N Y, 2001). Oligonucleotide-directed site-specific (or segment specific) mutagenesis procedures may be employed to provide an altered polynucleotide that has particular codons altered according to the substitution, deletion, or insertion desired. Alternatively, random or saturation mutagenesis techniques, such as alanine scanning mutagenesis, error prone polymerase chain reaction mutagenesis, and oligonucleotide-directed mutagenesis may be used to prepare immunogen polypeptide variants (see, e.g., Sambrook et al., supra).


A variety of criteria known to persons skilled in the art indicate whether an amino acid that is substituted at a particular position in a peptide or polypeptide is conservative (or similar). For example, a similar amino acid or a conservative amino acid substitution is one in which an amino acid residue is replaced with an amino acid residue having a similar side chain. Similar amino acids may be included in the following categories: amino acids with basic side chains (e.g., lysine, arginine, histidine); amino acids with acidic side chains (e.g., aspartic acid, glutamic acid); amino acids with uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, histidine); amino acids with nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan); amino acids with beta-branched side chains (e.g., threonine, valine, isoleucine), and amino acids with aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan). Proline, which is considered more difficult to classify, shares properties with amino acids that have aliphatic side chains (e.g., leucine, valine, isoleucine, and alanine) In certain circumstances, substitution of glutamine for glutamic acid or asparagine for aspartic acid may be considered a similar substitution in that glutamine and asparagine are amide derivatives of glutamic acid and aspartic acid, respectively. As understood in the art “similarity” between two polypeptides is determined by comparing the amino acid sequence and conserved amino acid substitutes thereto of the polypeptide to the sequence of a second polypeptide (e.g., using GENEWORKS, Align, the BLAST algorithm, or other algorithms described herein and practiced in the art).


In certain embodiments, a non-mutated protein epitope specific binding protein, TCR or CAR is capable of (a) specifically binding to an antigen:HLA complex on a cell surface independent or in the absence of CD8. In certain embodiments, a non-mutated protein epitope specific binding protein is a T cell receptor (TCR), a chimeric antigen receptor or an antigen-binding fragment of a TCR, any of which can be chimeric, humanized or human. In further embodiments, an antigen-binding fragment of the TCR comprises a single chain TCR (scTCR).


In certain embodiments, there is provided a composition comprising a non-mutated protein epitope-specific binding protein or high affinity recombinant TCR according to any one of the above embodiments and a pharmaceutically acceptable carrier, diluent, or excipient.


Methods useful for isolating and purifying recombinantly produced soluble TCR, by way of example, can include obtaining supernatants from suitable host cell/vector systems that secrete the recombinant soluble TCR into culture media and then concentrating the media using a commercially available filter. Following concentration, the concentrate can be applied to a single suitable purification matrix or to a series of suitable matrices, such as an affinity matrix or an ion exchange resin. One or more reverse phase HPLC steps may be employed to further purify a recombinant polypeptide. These purification methods can also be employed when isolating an immunogen from its natural environment. Methods for large scale production of one or more of the isolated/recombinant soluble TCR described herein include batch cell culture, which is monitored and controlled to maintain appropriate culture conditions. Purification of the soluble TCR may be performed according to methods described herein and known in the art.


III. Immunogenic and Vaccine Compositions

In one embodiment, provided herein is an immunogenic composition, e.g., a vaccine composition capable of raising a non-mutated protein epitope-specific response (e.g., a humoral or cell-mediated immune response). In some embodiments, the immunogenic composition comprises non-mutated protein epitope therapeutics (e.g., peptides, polynucleotides, TCR, CAR, cells containing TCR or CAR, dendritic cell containing polypeptide, dendritic cell containing polynucleotide, antibody, etc.) described herein corresponding to tumor specific non-mutated protein epitope identified herein.


A person skilled in the art will be able to select non-mutated protein epitope therapeutics by testing, for example, the generation of T-cells in vitro as well as their efficiency and overall presence, the proliferation, affinity and expansion of certain T-cells for certain peptides, and the functionality of the T-cells, e.g. by analyzing the IFN-γ production or tumor killing by T-cells. The most efficient peptides can then combined as an immunogenic composition.


In one embodiment of the present invention the different non-mutated protein epitope peptides and/or polypeptides are selected so that one immunogenic composition comprises non-mutated protein epitope peptides and/or polypeptides capable of associating with different MHC molecules, such as different MHC class I molecule. In some embodiments, an immunogenic composition comprises non-mutated protein epitope peptides and/or polypeptides capable of associating with the most frequently occurring MHC class I molecules. Hence immunogenic compositions described herein comprise different peptides capable of associating with at least 2, at least 3, or at least 4 MHC class I or class II molecules.


In one embodiment, an immunogenic composition described herein is capable of raising a specific cytotoxic T-cells response, specific helper T-cell response, or a B cell response.


In some embodiments, an immunogenic composition described herein can further comprise an adjuvant and/or a carrier. Examples of useful adjuvants and carriers are given herein below. Polypeptides and/or polynucleotides in the composition can be associated with a carrier such as e.g. a protein or an antigen-presenting cell such as e.g. a dendritic cell (DC) capable of presenting the peptide to a T-cell or a B cell. In further embodiments, DC-binding peptides are used as carriers to target the non-mutated protein epitope peptides and polynucleotides encoding the non-mutated protein epitope peptides to dendritic cells (Sioud et al. FASEB J 27: 3272-3283 (2013)).


In embodiments, the non-mutated protein epitope polypeptides or polynucleotides can be provided as antigen presenting cells (e.g., dendritic cells) containing such polypeptides or polynucleotides. In other embodiments, such antigen presenting cells are used to stimulate T cells for use in patients.


In some embodiments, the antigen presenting cells are dendritic cells. In related embodiments, the dendritic cells are autologous dendritic cells that are pulsed with the non-mutated protein epitope peptide or nucleic acid. The non-mutated protein epitope peptide can be any suitable peptide that gives rise to an appropriate T-cell response. T-cell therapy using autologous dendritic cells pulsed with peptides from a tumor associated antigen is disclosed in Murphy et al. (1996) The Prostate 29, 371-380 and Tjua et al. (1997) The Prostate 32, 272-278. In some embodiments, the T cell is a CTL. In some embodiments, the T cell is a HTL.


Thus, one embodiment of the present invention an immunogenic composition containing at least one antigen presenting cell (e.g., a dendritic cell) that is pulsed or loaded with one or more non-mutated protein epitope polypeptides or polynucleotides described herein. In embodiments, such APCs are autologous (e.g., autologous dendritic cells). Alternatively, peripheral blood mononuclear cells (PBMCs) isolated from a patient can be loaded with non-mutated protein epitope peptides or polynucleotides ex vivo. In related embodiments, such APCs or PBMCs are injected back into the patient.


The polynucleotide can be any suitable polynucleotide that is capable of transducing the dendritic cell, thus resulting in the presentation of a non-mutated protein epitope peptide and induction of immunity. In one embodiment, the polynucleotide can be naked DNA that is taken up by the cells by passive loading. In another embodiment, the polynucleotide is part of a delivery vehicle, for example, a liposome, virus like particle, plasmid, or expression vector. In another embodiment, the polynucleotide is delivered by a vector-free delivery system, for example, high performance electroporation and high-speed cell deformation). In embodiments, such antigen presenting cells (APCs) (e.g., dendritic cells) or peripheral blood mononuclear cells (PBMCs) are used to stimulate a T cell (e.g., an autologous T cell). In related embodiments, the T cell is a CTL. In other related embodiments, the T cell is an HTL. Such T cells are then injected into the patient. In some embodiments, CTL is injected into the patient. In some embodiments, HTL is injected into the patient. In some embodiments, both CTL and HTL are injected into the patient. Administration of either therapeutic can be performed simultaneously or sequentially and in any order.


The pharmaceutical compositions (e.g., immunogenic compositions) described herein for therapeutic treatment are intended for parenteral, topical, nasal, oral or local administration. In some embodiments, the pharmaceutical compositions described herein are administered parenterally, e.g., intravenously, subcutaneously, intradermally, or intramuscularly. In embodiments, the composition can be administered intratumorally. The compositions can be administered at the site of surgical excision to induce a local immune response to the tumor. In some embodiments, described herein are compositions for parenteral administration which comprise a solution of the non-mutated protein epitope peptides and immunogenic compositions are dissolved or suspended in an acceptable carrier, for example, an aqueous carrier. A variety of aqueous carriers can be used, e.g., water, buffered water, 0.9% saline, 0.3% glycine, hyaluronic acid and the like. These compositions can be sterilized by conventional, well known sterilization techniques, or can be sterile filtered. The resulting aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration. The compositions can contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.


The concentration of non-mutated protein epitope peptides and polynucleotides described herein in the pharmaceutical formulations can vary widely, i.e., from less than about 0.1%, usually at or at least about 2% to as much as 20% to 50% or more by weight, and will be selected by fluid volumes, viscosities, etc., according to the particular mode of administration selected.


The non-mutated protein epitope peptides and polynucleotides described herein can also be administered via liposomes, which target the peptides to a particular cells tissue, such as lymphoid tissue. Liposomes are also useful in increasing the half-life of the peptides. Liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like. In these preparations the peptide to be delivered is incorporated as part of a liposome, alone or in conjunction with a molecule which binds to, e.g., a receptor prevalent among lymphoid cells, such as monoclonal antibodies which bind to the DEC205 antigen, or with other therapeutic or immunogenic compositions. Thus, liposomes filled with a desired peptide or polynucleotide described herein can be directed to the site of lymphoid cells, where the liposomes then deliver the selected therapeutic/immunogenic polypeptide/polynucleotide compositions. Liposomes can be formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, for example, cholesterol. The selection of lipids is generally guided by consideration of, e.g., liposome size, acid lability and stability of the liposomes in the blood stream. A variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al., Ann. Rev. Biophys. Bioeng. 9; 467 (1980), U.S. Pat. Nos. 4,235,871, 4,501,728, 4,501,728, 4,837,028, and 5,019,369.


For targeting to the immune cells, a non-mutated protein epitope polypeptides or polynucleotides to be incorporated into the liposome for cell surface determinants of the desired immune system cells. A liposome suspension containing a peptide can be administered intravenously, locally, topically, etc. in a dose which varies according to, inter alia, the manner of administration, the polypeptide or polynucleotide being delivered, and the stage of the disease being treated.


In some embodiments, non-mutated protein epitope polypeptides and polynucleotides are targeted to dendritic cells. In one embodiment, the non-mutated protein epitope polypeptides and polynucleotides are target to dendritic cells using the markers DEC205, XCR1, CD197, CD80, CD86, CD123, CD209, CD273, CD283, CD289, CD184, CD85h, CD85j, CD85k, CD85d, CD85g, CD85a, TSLP receptor, or CD1a.


For solid compositions, conventional or nanoparticle nontoxic solid carriers can be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. For oral administration, a pharmaceutically acceptable nontoxic composition is formed by incorporating any of the normally employed excipients, such as those carriers previously listed, and generally 10-95% of active ingredient, that is, one or more non-mutated protein epitope polypeptides or polynucleotides described herein at a concentration of 25%-75%.


For aerosol administration, the non-mutated protein epitope polypeptides or polynucleotides can be supplied in finely divided form along with a surfactant and propellant. Representative of such agents are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride. Mixed esters, such as mixed or natural glycerides can be employed. The surfactant can constitute 0.1%-20% by weight of the composition, or 0.25-5%. The balance of the composition can be propellant. A carrier can also be included as desired, as with, e.g., lecithin for intranasal delivery.


Additional methods for delivering the non-mutated protein epitope polynucleotides described herein are also known in the art. For instance, the nucleic acid can be delivered directly, as “naked DNA”. This approach is described, for instance, in Wolff et al., Science 247: 1465-1468 (1990) as well as U.S. Pat. Nos. 5,580,859 and 5,589,466. The nucleic acids can also be administered using ballistic delivery as described, for instance, in U.S. Pat. No. 5,204,253. Particles comprised solely of DNA can be administered. Alternatively, DNA can be adhered to particles, such as gold particles.


For therapeutic or immunization purposes, mRNA encoding the non-mutated protein epitope peptides, or peptide binding agents can also be administered to the patient. In one embodiment, the mRNA is self-amplifying RNA. In a further embodiment, the self-amplifying RNA is a part of a synthetic lipid nanoparticle formulation (Geall et al., Proc Natl Acad Sci USA. 109: 14604-14609 (2012)).


The nucleic acids can also be delivered complexed to cationic compounds, such as cationic lipids. Lipid-mediated gene delivery methods are described, for instance, in WO 96/18372, WO 93/24640; Mannino & Gould-Fogerite, BioTechniques 6(7): 682-691 (1988); U.S. Pat. No. 5,279,833; WO 91/06309; and Felgner et al., Proc. Natl. Acad. Sci. USA 84: 7413-7414 (1987).


The non-mutated protein epitope peptides and polypeptides described herein can also be expressed by attenuated viruses, such as vaccinia or fowlpox. This approach involves the use of vaccinia virus as a vector to express nucleotide sequences that encode the peptide described herein. Upon introduction into an acutely or chronically infected host or into a noninfected host, the recombinant vaccinia virus expresses the immunogenic peptide, and thereby elicits a host CTL response. Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Pat. No. 4,722,848. Another vector is BCG (Bacille Calmette Guerin). BCG vectors are described in Stover et al. (Nature 351:456-460 (1991)). A wide variety of other vectors useful for therapeutic administration or immunization of the peptides described herein will be apparent to those skilled in the art from the description herein.


Adjuvants are any substance whose admixture into the immunogenic composition increases or otherwise modifies the immune response to the therapeutic agent. Carriers are scaffold structures, for example a polypeptide or a polysaccharide, to which a non-mutated protein epitope polypeptide or polynucleotide, is capable of being associated. Optionally, adjuvants are conjugated covalently or non-covalently to the polypeptides or polynucleotides described herein.


The ability of an adjuvant to increase the immune response to an antigen is typically manifested by a significant increase in immune-mediated reaction, or reduction in disease symptoms. For example, an increase in humoral immunity can be manifested by a significant increase in the titer of antibodies raised to the antigen, and an increase in T-cell activity can be manifested in increased cell proliferation, or cellular cytotoxicity, or cytokine secretion. An adjuvant can also alter an immune response, for example, by changing a primarily humoral or T helper 2 response into a primarily cellular, or T helper 1 response.


Suitable adjuvants are known in the art (see, WO 2015/095811) and include, but are not limited to poly(I:C), poly-ICLC, STING agonist, 1018 ISS, aluminum salts, Amplivax, AS15, BCG, CP-870,893, CpG7909, CyaA, dSLIM, GM-CSF, IC30, IC31, Imiquimod, ImuFact IMP321, IS Patch, ISS, ISCOMATRIX, JuvImmune, LipoVac, MF59, monophosphoryl lipid A, Montanide IMS 1312, Montanide ISA 206, Montanide ISA 50V, Montanide ISA-51, OK-432, OM-174, OM-197-MP-EC, ONTAK, PepTel®. vector system, PLG microparticles, resiquimod, SRL172, virosomes and other virus-like particles, YF-17D, VEGF trap, R848, beta-glucan, Pam3Cys, Pam3CSK4, Aquila's QS21 stimulon (Aquila Biotech, Worcester, Mass., USA) which is derived from saponin, mycobacterial extracts and synthetic bacterial cell wall mimics, and other proprietary adjuvants such as Ribi's Detox. Quil or Superfos. Adjuvants also include incomplete Freund's or GM-CSF. Several immunological adjuvants (e.g., MF59) specific for dendritic cells and their preparation have been described previously (Dupuis M, et al., Cell Immunol. 1998; 186(1):18-27; Allison A C; Dev Biol Stand. 1998; 92:3-11) (Mosca et al. Frontiers in Bioscience, 2007; 12:4050-4060) (Gamvrellis et al. Immunol & Cell Biol. 2004; 82: 506-516). Also cytokines can be used. Several cytokines have been directly linked to influencing dendritic cell migration to lymphoid tissues (e.g., TNF-alpha), accelerating the maturation of dendritic cells into efficient antigen-presenting cells for T-lymphocytes (e.g., GM-CSF, PGE1, PGE2, IL-1, IL-1b, IL-4, IL-6 and CD40L) (U.S. Pat. No. 5,849,589 incorporated herein by reference in its entirety) and acting as immunoadjuvants (e.g., IL-12) (Gabrilovich D I, et al., J Immunother Emphasis Tumor Immunol. 1996 (6):414-418).


CpG immunostimulatory oligonucleotides have also been reported to enhance the effects of adjuvants in a vaccine setting. Without being bound by theory, CpG oligonucleotides act by activating the innate (non-adaptive) immune system via Toll-like receptors (TLR), mainly TLR9. CpG triggered TLR9 activation enhances antigen-specific humoral and cellular responses to a wide variety of antigens, including peptide or protein antigens, live or killed viruses, dendritic cell vaccines, autologous cellular vaccines and polysaccharide conjugates in both prophylactic and therapeutic vaccines. Importantly, it enhances dendritic cell maturation and differentiation, resulting in enhanced activation of TH1 cells and strong cytotoxic T-lymphocyte (CTL) generation, even in the absence of CD4 T-cell help. The TH1 bias induced by TLR9 stimulation is maintained even in the presence of vaccine adjuvants such as alum or incomplete Freund's adjuvant (IFA) that normally promote a TH2 bias. CpG oligonucleotides show even greater adjuvant activity when formulated or co-administered with other adjuvants or in formulations such as microparticles, nanoparticles, lipid emulsions or similar formulations, which are especially necessary for inducing a strong response when the antigen is relatively weak. They also accelerate the immune response and enabled the antigen doses to be reduced with comparable antibody responses to the full-dose vaccine without CpG in some experiments (Arthur M. Krieg, Nature Reviews, Drug Discovery, 5, June 2006, 471-484). U.S. Pat. No. 6,406,705 B1 describes the combined use of CpG oligonucleotides, non-nucleic acid adjuvants and an antigen to induce an antigen-specific immune response. A commercially available CpG TLR9 antagonist is dSLIM (double Stem Loop Immunomodulator) by Mologen (Berlin, GERMANY), which is a component of the pharmaceutical composition described herein. Other TLR binding molecules such as RNA binding TLR 7, TLR 8 and/or TLR 9 can also be used.


Other examples of useful adjuvants include, but are not limited to, chemically modified CpGs (e.g. CpR, Idera), Poly(I:C)(e.g. polyi:CI2U), non-CpG bacterial DNA or RNA, ssRNA40 for TLR8, as well as immunoactive small molecules and antibodies such as cyclophosphamide, sunitinib, bevacizumab, celebrex, NCX-4016, sildenafil, tadalafil, vardenafil, sorafinib, XL-999, CP-547632, pazopanib, ZD2171, AZD2171, ipilimumab, tremelimumab, and SC58175, which can act therapeutically and/or as an adjuvant. The amounts and concentrations of adjuvants and additives useful in the context of the present invention can readily be determined by the skilled artisan without undue experimentation. Additional adjuvants include colony-stimulating factors, such as Granulocyte Macrophage Colony Stimulating Factor (GM-CSF, sargramostim).


In some embodiments, an immunogenic composition according to the present invention can comprise more than one different adjuvants. Furthermore, the invention encompasses a therapeutic composition comprising any adjuvant substance including any of the above or combinations thereof. It is also contemplated that the non-mutated protein epitope therapeutic (e.g., a humoral or cell-mediated immune response). In some embodiments, the immunogenic composition comprises non-mutated protein epitope therapeutics (e.g., peptides, polynucleotides, TCR, CAR, cells containing TCR or CAR, dendritic cell containing polypeptide, dendritic cell containing polynucleotide, antibody, etc.) and the adjuvant can be administered separately in any appropriate sequence.


A carrier can be present independently of an adjuvant. The function of a carrier can for example be to increase the molecular weight of in particular mutant in order to increase their activity or immunogenicity, to confer stability, to increase the biological activity, or to increase serum half-life. Furthermore, a carrier can aid presenting peptides to T-cells. The carrier can be any suitable carrier known to the person skilled in the art, for example a protein or an antigen presenting cell. A carrier protein could be but is not limited to keyhole limpet hemocyanin, serum proteins such as transferrin, bovine serum albumin, human serum albumin, thyroglobulin or ovalbumin, immunoglobulins, or hormones, such as insulin or palmitic acid. In one embodiment, the carrier comprises a human fibronectin type III domain (Koide et al. Methods Enzymol. 2012; 503:135-56). For immunization of humans, the carrier must be a physiologically acceptable carrier acceptable to humans and safe. However, tetanus toxoid and/or diptheria toxoid are suitable carriers in one embodiment of the invention. Alternatively, the carrier can be dextrans for example sepharose.


In some embodiments, the polypeptides can be synthesized as multiply linked peptides as an alternative to coupling a polypeptide to a carrier to increase immunogenicity. Such molecules are also known as multiple antigenic peptides (MAPS).


IV. Combinations of CTL Peptides and HTL Peptides

Immunogenic or vaccine compositions comprising the non-mutated protein epitope polypeptides and polynucleotides described herein, or analogs thereof, which have immunostimulatory activity can be modified to provide desired attributes, such as improved serum half-life, or to enhance immunogenicity.


For instance, the ability of the non-mutated protein epitope peptides to induce CTL activity can be enhanced by linking the peptide to a sequence which contains at least one epitope that is capable of inducing a T helper cell response. In one embodiment, CTL epitope/HTL epitope conjugates are linked by a spacer molecule. The spacer is typically comprised of relatively small, neutral molecules, such as amino acids or amino acid mimetics, which are substantially uncharged under physiological conditions. The spacers are typically selected from, e.g., Ala, Gly, or other neutral spacers of nonpolar amino acids or neutral polar amino acids. It will be understood that the optionally present spacer need not be comprised of the same residues and thus can be a hetero- or homo-oligomer. When present, the spacer will usually be at least one or two residues, more usually three to six residues. Alternatively, the CTL peptide can be linked to the T helper peptide without a spacer.


Although the CTL peptide epitope can be linked directly to the T helper peptide epitope, CTL epitope/HTL epitope conjugates can be linked by a spacer molecule. The spacer is typically comprised of relatively small, neutral molecules, such as amino acids or amino acid mimetics, which are substantially uncharged under physiological conditions. The spacers are typically selected from, e.g., Ala, Gly, or other neutral spacers of nonpolar amino acids or neutral polar amino acids. It will be understood that the optionally present spacer need not be comprised of the same residues and thus can be a hetero- or homo-oligomer. When present, the spacer will usually be at least one or two residues, more usually three to six residues. The CTL peptide epitope can be linked to the T helper peptide epitope either directly or via a spacer either at the amino or carboxy terminus of the CTL peptide. The amino terminus of either the immunogenic peptide or the T helper peptide can be acylated.


HTL peptide epitopes can also be modified to alter their biological properties. For example, peptides comprising HTL epitopes can contain D-amino acids to increase their resistance to proteases and thus extend their serum half-life. Also, the epitope peptides can be conjugated to other molecules such as lipids, proteins or sugars, or any other synthetic compounds, to increase their biological activity. For example, the T helper peptide can be conjugated to one or more palmitic acid chains at either the amino or carboxyl termini.


In certain embodiments, the T helper peptide is one that is recognized by T helper cells present in the majority of the population. This can be accomplished by selecting amino acid sequences that bind to many, most, or all of the HLA class II molecules. These are known as “loosely HLA-restricted” or “promiscuous” T helper sequences. Examples of amino acid sequences that are promiscuous include sequences from antigens such as tetanus toxoid at positions 830-843 (QYIKANSKFIGITE), Plasmodium falciparum CS protein at positions 378-398 (DIEKKIAKMEKASSVFNVVNS), and Streptococcus 18 kD protein at positions 116 (GAVDSILGGVATYGAA). Other examples include peptides bearing a DR 1-4-7 supermotif, or either of the DR3 motifs.


Alternatively, it is possible to prepare synthetic peptides capable of stimulating T helper lymphocytes, in a loosely HLA-restricted fashion, using amino acid sequences not found in nature (see, e.g., PCT publication WO 95/07707). These synthetic compounds called Pan-DR-binding epitopes (e.g., PADRE, Epimmune, Inc., San Diego, Calif.) are designed to bind most HLA-DR (human HLA class II) molecules. For instance, a pan-DR-binding epitope peptide having the formula: aKXVWANTLKAAa, where “X” is either cyclohexylalanine, phenylalanine, or tyrosine, and a is either D-alanine or L-alanine, has been found to bind to most HLA-DR alleles, and to stimulate the response of T helper lymphocytes from most individuals, regardless of their HLA type. An alternative of a pan-DR binding epitope comprises all “L” natural amino acids and can be provided in the form of nucleic acids that encode the epitope.


In some embodiments it can be desirable to include in a non-mutated protein epitope therapeutic (e.g., peptides, polynucleotides, TCR, CAR, cells containing TCR or CAR, dendritic cell containing polypeptide, dendritic cell containing polynucleotide, antibody, etc.) in pharmaceutical compositions (e.g., immunogenic compositions) at least one component of which primes cytotoxic T lymphocytes. Lipids have been identified as agents capable of priming CTL in vivo against viral antigens. For example, palmitic acid residues can be attached to the ε- and α-amino groups of a lysine residue and then linked, e.g., via one or more linking residues such as Gly, Gly-Gly-, Ser, Ser-Ser, or the like, to an immunogenic non-mutated protein epitope peptide. The lipidated peptide can then be administered either directly in a micelle or particle, incorporated into a liposome, or emulsified in an adjuvant. In one embodiment, a particularly effective immunogenic construct comprises palmitic acid attached to ε- and α-amino groups of Lys, which is attached via linkage, e.g., Ser-Ser, to the amino terminus of the immunogenic peptide.


As another example of lipid priming of CTL responses, E. coli lipoproteins, such as tripalmitoyl-S-glycerylcysteinlyseryl-serine (P3CSS) can be used to prime virus specific CTL when covalently attached to an appropriate peptide. (See, e.g., Deres, et al., Nature 342:561, 1989). Non-mutated protein epitope peptides described herein can be coupled to P3CSS, for example, and the lipopeptide administered to an individual to specifically prime a CTL response to the target antigen. Moreover, because the induction of neutralizing antibodies can also be primed with P3CSS-conjugated epitopes, two such compositions can be combined to more effectively elicit both humoral and cell-mediated responses to infection.


As noted herein, additional amino acids can be added to the termini of a non-mutated protein epitope peptide to provide for ease of linking peptides one to another, for coupling to a carrier support or larger peptide, for modifying the physical or chemical properties of the peptide or oligopeptide, or the like. Amino acids such as tyrosine, cysteine, lysine, glutamic or aspartic acid, or the like, can be introduced at the C- or N-terminus of the peptide or oligopeptide. However, it is to be noted that modification at the carboxyl terminus of a T cell epitope can, in some cases, alter binding characteristics of the peptide. In addition, the peptide or oligopeptide sequences can differ from the natural sequence by being modified by terminal-NH2 acylation, e.g., by alkanoyl (C1-C20) or thioglycolyl acetylation, terminal-carboxyl amidation, e.g., ammonia, methylamine, etc. In some instances these modifications can provide sites for linking to a support or other molecule.


An embodiment of an immunogenic composition described herein comprises ex vivo administration of a cocktail of epitope-bearing non-mutated protein epitope polypeptide or polynucleotides to PBMC, or isolated DC therefrom, from the patient's blood. A pharmaceutical to facilitate harvesting of dendritic cells (DCs) can be used, including GM-CSF, IL-4, IL-6, IL-1β, and TNFα. After pulsing the DCs with peptides or polynucleotides encoding the peptides, and prior to reinfusion into patients, the DC are washed to remove unbound peptides. In this embodiment, a vaccine or immunogenic composition comprises peptide-pulsed DCs which present the pulsed peptide epitopes complexed with HLA molecules on their surfaces. The composition is then administered to the patient. In other embodiments, such pulsed DCs are used to stimulate T cells suitable for use in T cell therapy.


V. Multi-Epitope Immunogenic Compositions

A number of different approaches are available which allow simultaneous delivery of multiple epitopes. Nucleic acids encoding the non-mutated protein epitope peptides described herein are a particularly useful embodiment of the invention. In one embodiment, the nucleic acid is RNA. In some embodiments, minigene constructs encoding a non-mutated protein epitope peptide comprising one or multiple epitopes described herein are used to administer nucleic acids encoding the non-mutated protein epitope peptides described herein uses.


The use of multi-epitope minigenes is described An, L. and Whitton, J. L., J. Virol. 71:2292, 1997; Thomson, S. A. et al., J. Immunol. 157:822, 1996; Whitton, J. L. et al., J. Virol. 67:348, 1993; Hanke, R. et al., Vaccine 16:426, 1998. For example, a multi-epitope DNA plasmid encoding supermotif- and/or motif-bearing antigen peptides, a universal helper T cell epitope (or multiple tumor associated antigen HTL epitopes), and an endoplasmic reticulum-translocating signal sequence can be engineered.


The immunogenicity of a multi-epitopic minigene can be tested in transgenic mice to evaluate the magnitude of immune response induced against the epitopes tested. Further, the immunogenicity of DNA-encoded epitopes in vivo can be correlated with the in vitro responses of specific CTL lines against target cells transfected with the DNA plasmid. Thus, these experiments can show that the minigene serves to both: 1.) generate a cell mediated and/or humoral response and 2.) that the induced immune cells recognized cells expressing the encoded epitopes.


For example, to create a DNA sequence encoding the selected non-mutated protein epitope (minigene) for expression in human cells, the amino acid sequences of the epitopes can be reverse translated. A human codon usage table can be used to guide the codon choice for each amino acid. These non-mutated protein epitope-encoding DNA sequences can be directly adjoined, so that when translated, a continuous polypeptide sequence is created. To optimize expression and/or immunogenicity, additional elements can be incorporated into the minigene design. Examples of amino acid sequences that can be reverse translated and included in the minigene sequence include: HLA class I epitopes, HLA class II epitopes, a ubiquitination signal sequence, and/or an endoplasmic reticulum targeting signal. In addition, HLA presentation of CTL and HTL epitopes can be improved by including synthetic (e.g. poly-alanine) or naturally-occurring flanking sequences adjacent to the CTL or HTL epitopes; these larger peptides comprising the epitope(s) are within the scope of the invention.


The minigene sequence can be converted to DNA by assembling oligonucleotides that encode the plus and minus strands of the minigene. Overlapping oligonucleotides (30-100 bases long) can be synthesized, phosphorylated, purified and annealed under appropriate conditions using well known techniques. The ends of the oligonucleotides can be joined, for example, using T4 DNA ligase. This synthetic minigene, encoding the epitope polypeptide, can then be cloned into a desired expression vector.


Standard regulatory sequences well known to those of skill in the art can be included in the vector to ensure expression in the target cells. For example, a promoter with a down-stream cloning site for minigene insertion; a polyadenylation signal for efficient transcription termination; an E. coli origin of replication; and an E. coli selectable marker (e.g. ampicillin or kanamycin resistance). Numerous promoters can be used for this purpose, e.g., the human cytomegalovirus (hCMV) promoter. See, e.g., U.S. Pat. Nos. 5,580,859 and 5,589,466 for other suitable promoter sequences.


Additional vector modifications can be used to optimize minigene expression and immunogenicity. In some cases, introns are utilized for efficient gene expression, and one or more synthetic or naturally-occurring introns could be incorporated into the transcribed region of the minigene. The inclusion of mRNA stabilization sequences and sequences for replication in mammalian cells can also be considered for increasing minigene expression.


Once an expression vector is selected, the minigene can be cloned into the polylinker region downstream of the promoter. This plasmid is transformed into an appropriate E. coli strain, and DNA is prepared using standard techniques. The orientation and DNA sequence of the minigene, as well as all other elements included in the vector, can be confirmed using restriction mapping and DNA sequence analysis. Bacterial cells harboring the correct plasmid can be stored as a master cell bank and a working cell bank.


In addition, immunomodulatory sequences appear to play a role in the immunogenicity of DNA vaccines. These sequences can be included in the vector, outside the minigene coding sequence, if desired to enhance immunogenicity. In one embodiment, the sequences are immunostimulatory. In another embodiment, the sequences are ISSs or CpGs.


In some embodiments, a bi-cistronic expression vector which allows production of both the minigene-encoded epitopes and a second protein (included to enhance or decrease immunogenicity) can be used. Examples of proteins or polypeptides that could beneficially enhance the immune response if co-expressed include cytokines (e.g., IL-2, IL-12, GM-CSF), cytokine-inducing molecules (e.g., LeIF), costimulatory molecules, or for HTL responses, pan-DR binding proteins. Helper (HTL) epitopes can be joined to intracellular targeting signals and expressed separately from expressed CTL epitopes; this allows direction of the HTL epitopes to a cell compartment different than that of the CTL epitopes. If required, this could facilitate more efficient entry of HTL epitopes into the HLA class II pathway, thereby improving HTL induction. In contrast to HTL or CTL induction, specifically decreasing the immune response by co-expression of immunosuppressive molecules (e.g. TGF-β) can be beneficial in certain diseases.


Therapeutic quantities of plasmid DNA can be produced for example, by fermentation in E. coli, followed by purification. Aliquots from the working cell bank are used to inoculate growth medium, and grown to saturation in shaker flasks or a bioreactor according to well-known techniques. Plasmid DNA can be purified using standard bioseparation technologies such as solid phase anion-exchange resins supplied by QIAGEN, Inc. (Valencia, Calif.). If required, supercoiled DNA can be isolated from the open circular and linear forms using gel electrophoresis or other methods.


Purified plasmid DNA can be prepared for injection using a variety of formulations. The simplest of these is reconstitution of lyophilized DNA in sterile phosphate-buffer saline (PBS). This approach, known as “naked DNA,” is currently being used for intramuscular (IM) administration in clinical trials. To maximize the immunotherapeutic effects of minigene DNA vaccines, an alternative method for formulating purified plasmid DNA can be used. A variety of methods have been described, and new techniques can become available. Cationic lipids can also be used in the formulation (see, e.g., as described by WO 93/24640; Mannino & Gould-Fogerite, BioTechniques 6(7): 682 (1988); U.S. Pat. No. 5,279,833; WO 91/06309; and Feigner, et al., Proc. Nat'l Acad. Sci. USA 84:7413 (1987). In addition, glycolipids, fusogenic liposomes, peptides and compounds referred to collectively as protective, interactive, non-condensing compounds (PINC) could also be complexed to purified plasmid DNA to influence variables such as stability, intramuscular dispersion, or trafficking to specific organs or cell types.


In another embodiment, the nucleic acid is introduced into cells by use of high-speed cell deformation. During high-speed deformation, cells are squeezed such that temporary disruptions occur in the cell membrane, thus allowing the nucleic acid to enter the cell. Alternatively, protein can be produced from expression vectors—in a bacterial expression vector, for example, and the proteins can then be delivered to the cell.


Target cell sensitization can be used as a functional assay for expression and HLA class I presentation of minigene-encoded CTL epitopes. For example, the plasmid DNA is introduced into a mammalian cell line that is suitable as a target for standard CTL chromium release assays. The transfection method used will be dependent on the final formulation. Electroporation can be used for “naked” DNA, whereas cationic lipids allow direct in vitro transfection. A plasmid expressing green fluorescent protein (GFP) can be co-transfected to allow enrichment of transfected cells using fluorescence activated cell sorting (FACS). These cells are then chromium-51 (51Cr) labeled and used as target cells for epitope-specific CTL lines; cytolysis, detected by 51Cr release, indicates both production of, and HLA presentation of, minigene-encoded CTL epitopes. Expression of HTL epitopes can be evaluated in an analogous manner using assays to assess HTL activity.


In vivo immunogenicity is a second approach for functional testing of minigene DNA formulations. Transgenic mice expressing appropriate human HLA proteins are immunized with the DNA product. The dose and route of administration are formulation dependent (e.g., IM for DNA in PBS, intraperitoneal (IP) for lipid-complexed DNA). An exemplary protocol is twenty-one days after immunization, splenocytes are harvested and restimulated for 1 week in the presence of peptides encoding each epitope being tested. Thereafter, for CTL effector cells, assays are conducted for cytolysis of peptide-loaded, 51Cr-labeled target cells using standard techniques. Lysis of target cells that were sensitized by HLA loaded with peptide epitopes, corresponding to minigene-encoded epitopes, demonstrates DNA vaccine function for in vivo induction of CTLs. Immunogenicity of HTL epitopes is evaluated in transgenic mice in an analogous manner.


Alternatively, the nucleic acids can be administered using ballistic delivery as described, for instance, in U.S. Pat. No. 5,204,253. Using this technique, particles comprised solely of DNA are administered. In a further alternative embodiment, DNA can be adhered to particles, such as gold particles.


VI. Cells

In one aspect, the present invention also provides cells expressing a non-mutated protein epitope-recognizing receptor that activates an immunoresponsive cell (e.g., T cell receptor (TCR) or chimeric antigen receptor (CAR)), and methods of using such cells for the treatment of a disease that requires an enhanced immune response.


Such cells include genetically modified immunoresponsive cells (e.g., T cells, Natural Killer (NK) cells, cytotoxic T lymphocytes (CTL) cells, helper T lymphocyte (HTL) cells) expressing an antigen-recognizing receptor (e.g., TCR or CAR) that binds one of the non-mutated protein epitope peptides described herein, and methods of use therefore for the treatment of neoplasia and other pathologies where an increase in an antigen-specific immune response is desired. T cell activation is mediated by a TCR or a CAR targeted to an antigen.


The present invention provides cells expressing a combination of an antigen-recognizing receptor that activates an immunoresponsive cell (e.g., TCR, CAR) and a chimeric co-stimulating receptor (CCR), and methods of using such cells for the treatment of a disease that requires an enhanced immune response. In one embodiment, tumor antigen-specific T cells, NK cells, CTL cells or other immunoresponsive cells are used as shuttles for the selective enrichment of one or more co-stimulatory ligands for the treatment or prevention of neoplasia. Such cells are administered to a human subject in need thereof for the treatment or prevention of a particular cancer.


In one embodiment, the tumor antigen-specific human lymphocytes that can be used in the methods of the invention include, without limitation, peripheral donor lymphocytes genetically modified to express chimeric antigen receptors (CARs) (Sadelain, M., et al. 2003 Nat Rev Cancer 3:35-45), peripheral donor lymphocytes genetically modified to express a full-length tumor antigen-recognizing T cell receptor complex comprising the a and p heterodimer (Morgan, R. A., et al. 2006 Science 314:126-129), lymphocyte cultures derived from tumor infiltrating lymphocytes (TILs) in tumor biopsies (Panelli, M. C., et al. 2000 J Immunol 164:495-504; Panelli, M. C., et al. 2000 J Immunol 164:4382-4392), and selectively in vitro-expanded antigen-specific peripheral blood leukocytes employing artificial antigen-presenting cells (AAPCs) or pulsed dendritic cells (Dupont, J., et al. 2005 Cancer Res 65:5417-5427; Papanicolaou, G. A., et al. 2003 Blood 102:2498-2505). The T cells may be autologous, allogeneic, or derived in vitro from engineered progenitor or stem cells.


Co-Stimulatory Ligands

In one embodiment, the cells of the invention are provided with at least one co-stimulatory ligand which is a non-antigen specific signal important for full activation of an immune cell. Co-stimulatory ligands include, without limitation, tumor necrosis factor (TNF) ligands, cytokines (such as IL-2, IL-12, IL-15 or IL21), and immunoglobulin (Ig) superfamily ligands.


Tumor necrosis factor (TNF) is a cytokine involved in systemic inflammation and stimulates the acute phase reaction. Its primary role is in the regulation of immune cells. Tumor necrosis factor (TNF) ligands share a number of common features. The majority of the ligands are synthesized as type II transmembrane proteins containing a short cytoplasmic segment and a relatively long extracellular region. TNF ligands include, without limitation, nerve growth factor (NGF), CD40L (CD40L)/CD154, CD137L/4-1BBL, tumor necrosis factor alpha (TNFα), CD134L/OX4OL/CD252, CD27L/CD70, Fas ligand (FasL), CD3OL/CD153, tumor necrosis factor β (TNF(3)/lymphotoxin-alpha (LTa), lymphotoxin-beta (ur(3), CD257/B cell-activating factor (BAFF)/Blys/THANK/Ta11-1, glucocorticoid-induced TNF Receptor ligand (GITRL), and TNF-related apoptosis-inducing ligand (TRAIL), LIGHT (TNFSF14). The immunoglobulin (Ig) superfamily is a large group of cell surface and soluble proteins that are involved in the recognition, binding, or adhesion processes of cells. These proteins share structural features with immunoglobulins—they possess an immunoglobulin domain (fold). Immunoglobulin superfamily ligands include, without limitation, CD80 and CD86, both ligands for CD28.


Compositions comprising genetically modified immunoresponsive cells of the invention can be provided systemically or directly to a subject for the treatment of a neoplasia. In one embodiment, cells of the invention are directly injected into an organ of interest (e.g., an organ affected by a tumor). Alternatively, compositions comprising genetically modified immunoresponsive cells are provided indirectly to the organ of interest, for example, by administration into the circulatory system (e.g., the tumor vasculature). Expansion and differentiation agents can be provided prior to, during or after administration of the cells to increase production of T cells, NK cells, or CTL cells in vitro or in vivo.


The modified cells can be administered in any physiologically acceptable vehicle, normally intravascularly, although they may also be introduced into bone or other convenient site where the cells may find an appropriate site for regeneration and differentiation (e.g., thymus). Genetically modified immunoresponsive cells of the invention can comprise a purified population of cells. Those skilled in the art can readily determine the percentage of genetically modified immunoresponsive cells in a population using various well-known methods, such as fluorescence activated cell sorting (FACS). Dosages can be readily adjusted by those skilled in the art (e.g., a decrease in purity may require an increase in dosage). The cells can be introduced by injection, catheter, or the like. If desired, factors can also be included, including, but not limited to, interleukins, e.g. IL-2, IL-3, IL-6, and IL-11, as well as the other interleukins, the colony stimulating factors, such as G-, M- and GM-CSF, interferons, e.g. y-interferon and erythropoietin.


Compositions of the invention include pharmaceutical compositions comprising genetically modified immunoresponsive cells or their progenitors and a pharmaceutically acceptable carrier. Administration can be autologous or heterologous. For example, immunoresponsive cells, or progenitors can be obtained from one subject, and administered to the same subject or a different, compatible subject. Peripheral blood derived immunoresponsive cells of the invention or their progeny (e.g., in vivo, ex vivo or in vitro derived) can be administered via localized injection, including catheter administration, systemic injection, localized injection, intravenous injection, or parenteral administration. When administering a therapeutic composition of the present invention (e.g., a pharmaceutical composition containing a genetically modified immunoresponsive cell), it will generally be formulated in a unit dosage injectable form (solution, suspension, emulsion).


VII. Methods of Use and Pharmaceutical Compositions

The non-mutated protein epitope therapeutics (e.g., peptides, polynucleotides, TCR, CAR, cells containing TCR or CAR, dendritic cell containing polypeptide, dendritic cell containing polynucleotide, antibody, etc.) described herein are useful in a variety of applications including, but not limited to, therapeutic treatment methods, such as the treatment of cancer. In some embodiments, the therapeutic treatment methods comprise immunotherapy. In certain embodiments, a non-mutated protein epitope peptide is useful for activating, promoting, increasing, and/or enhancing an immune response, redirecting an existing immune response to a new target, increasing the immunogenicity of a tumor, inhibiting tumor growth, reducing tumor volume, increasing tumor cell apoptosis, and/or reducing the tumorigenicity of a tumor. The methods of use can be in vitro, ex vivo, or in vivo methods.


In some aspects, the present invention provides methods for activating an immune response in a subject using a non-mutated protein epitope therapeutic described herein. In some embodiments, the invention provides methods for promoting an immune response in a subject using a non-mutated protein epitope therapeutic described herein. In some embodiments, the invention provides methods for increasing an immune response in a subject using a non-mutated protein epitope peptide described herein. In some embodiments, the invention provides methods for enhancing an immune response using a non-mutated protein epitope peptide. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing cell-mediated immunity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing T-cell activity or humoral immunity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing CTL or HTL activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing NK cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing T-cell activity and increasing NK cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing CTL activity and increasing NK cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises inhibiting or decreasing the suppressive activity of Tregs. In some embodiments, the immune response is a result of antigenic stimulation. In some embodiments, the antigenic stimulation is a tumor cell. In some embodiments, the antigenic stimulation is cancer.


In some embodiments, the invention provides methods of activating, promoting, increasing, and/or enhancing of an immune response using a non-mutated protein epitope therapeutic described herein. In some embodiments, a method comprises administering to a subject in need thereof a therapeutically effective amount of a non-mutated protein epitope therapeutic that delivers a non-mutated protein epitope polypeptide or polynucleotide to a tumor cell. In some embodiments, a method comprises administering to a subject in need thereof a therapeutically effective amount of a non-mutated protein epitope therapeutic that binds the tumor associated antigen and is internalized by the tumor cell. In some embodiments, a method comprises administering to a subject in need thereof a therapeutically effective amount of a non-mutated protein epitope polypeptide that is internalized by a tumor cell, and the non-mutated protein epitope peptide is processed by the cell. In some embodiments, a method comprises administering to a subject in need thereof a therapeutically effective amount of a non-mutated protein epitope polypeptide that is internalized by a tumor cell, and an antigenic peptide is presented on the surface of the tumor cell. In some embodiments, a method comprises administering to a subject in need thereof a therapeutically effective amount of a non-mutated protein epitope polypeptide that is internalized by the tumor cell, is processed by the cell, and an antigenic peptide is presented on the surface of the tumor cell.


In some embodiments, a method comprises administering to a subject in need thereof a therapeutically effective amount of a non-mutated protein epitope polypeptide or polynucleotide described herein that delivers an exogenous polypeptide comprising at least one antigenic peptide to a tumor cell, wherein the antigenic peptide is presented on the surface of the tumor cell. In some embodiments, the antigenic peptide is presented on the surface of the tumor cell in complex with a MHC class I molecule. In some embodiments, the antigenic peptide is presented on the surface of the tumor cell in complex with a MHC class II molecule.


In some embodiments, a method comprises contacting a tumor cell with a non-mutated protein epitope polypeptide or polynucleotide described herein that delivers an exogenous polypeptide comprising at least one antigenic peptide to the tumor cell, wherein the antigenic peptide is presented on the surface of the tumor cell. In some embodiments, the antigenic peptide is presented on the surface of the tumor cell in complex with a MHC class I molecule. In some embodiments, the antigenic peptide is presented on the surface of the tumor cell in complex with a MHC class II molecule.


In some embodiments, a method comprises administering to a subject in need thereof a therapeutically effective amount of a non-mutated protein epitope polypeptide or polynucleotide described herein that delivers an exogenous polypeptide comprising at least one antigenic peptide to a tumor cell, wherein the antigenic peptide is presented on the surface of the tumor cell, and an immune response against the tumor cell is induced. In some embodiments, the immune response against the tumor cell is increased. In some embodiments, the non-mutated protein epitope polypeptide or polynucleotide delivers an exogenous polypeptide comprising at least one antigenic peptide to a tumor cell, wherein the antigenic peptide is presented on the surface of the tumor cell, and tumor growth is inhibited.


In some embodiments, a method comprises administering to a subject in need thereof a therapeutically effective amount of a non-mutated protein epitope polypeptide or polynucleotide described herein that delivers an exogenous polypeptide comprising at least one antigenic peptide to a tumor cell, wherein the antigenic peptide is presented on the surface of the tumor cell, and T-cell killing directed against the tumor cell is induced. In some embodiments, T-cell killing directed against the tumor cell is enhanced. In some embodiments, T-cell killing directed against the tumor cell is increased.


In some embodiments, a method of increasing an immune response in a subject comprises administering to the subject a therapeutically effective amount of a non-mutated protein epitope therapeutic described herein, wherein the agent is an antibody that specifically binds the non-mutated protein epitope described herein. In some embodiments, a method of increasing an immune response in a subject comprises administering to the subject a therapeutically effective amount of the antibody.


The present invention provides methods of redirecting an existing immune response to a tumor. In some embodiments, a method of redirecting an existing immune response to a tumor comprises administering to a subject a therapeutically effective amount of a non-mutated protein epitope therapeutic described herein. In some embodiments, the existing immune response is against a virus. In some embodiments, the virus is selected from the group consisting of: measles virus, varicella-zoster virus (VZV; chickenpox virus), influenza virus, mumps virus, poliovirus, rubella virus, rotavirus, hepatitis A virus (HAV), hepatitis B virus (HBV), Epstein Barr virus (EBV), and cytomegalovirus (CMV). In some embodiments, the virus is varicella-zoster virus. In some embodiments, the virus is cytomegalovirus. In some embodiments, the virus is measles virus. In some embodiments, the existing immune response has been acquired after a natural viral infection. In some embodiments, the existing immune response has been acquired after vaccination against a virus. In some embodiments, the existing immune response is a cell-mediated response. In some embodiments, the existing immune response comprises cytotoxic T-cells (CTLs) or HTLs.


In some embodiments, a method of redirecting an existing immune response to a tumor in a subject comprises administering a fusion protein comprising (i) an antibody that specifically binds a non-mutated protein epitope and (ii) at least one non-mutated protein epitope peptide described herein, wherein (a) the fusion protein is internalized by a tumor cell after binding to the tumor-associated antigen; (b) the non-mutated protein epitope peptide is processed and presented on the surface of the tumor cell associated with a MHC class I molecule; and (c) the non-mutated protein epitope peptide/MHC Class I complex is recognized by cytotoxic T-cells. In some embodiments, the cytotoxic T-cells are memory T-cells. In some embodiments, the memory T-cells are the result of a vaccination with the non-mutated protein epitope peptide.


The present invention provides methods of increasing the immunogenicity of a tumor. In some embodiments, a method of increasing the immunogenicity of a tumor comprises contacting the tumor or tumor cells with an effective amount of a non-mutated protein epitope therapeutic described herein. In some embodiments, a method of increasing the immunogenicity of a tumor comprises administering to a subject a therapeutically effective amount of a non-mutated protein epitope therapeutic described herein.


The present invention also provides methods for inhibiting growth of a tumor using a non-mutated protein epitope therapeutic described herein. In certain embodiments, a method of inhibiting growth of a tumor comprises contacting a cell mixture with a non-mutated protein epitope therapeutic in vitro. For example, an immortalized cell line or a cancer cell line mixed with immune cells (e.g., T-cells) is cultured in medium to which a non-mutated protein epitope peptide is added. In some embodiments, tumor cells are isolated from a patient sample such as, for example, a tissue biopsy, pleural effusion, or blood sample, mixed with immune cells (e.g., T-cells), and cultured in medium to which an antigen therapeutic is added. In some embodiments, a non-mutated protein epitope therapeutic increases, promotes, and/or enhances the activity of the immune cells. In some embodiments, a non-mutated protein epitope therapeutic inhibits tumor cell growth. In some embodiments, a non-mutated protein epitope therapeutic activates killing of the tumor cells.


In certain embodiments, the subject is a human. In certain embodiments, the subject has a tumor or the subject had a tumor which was at least partially removed.


In some embodiments, a method of inhibiting growth of a tumor comprises redirecting an existing immune response to a new target, comprising administering to a subject a therapeutically effective amount of a non-mutated protein epitope therapeutic, wherein the existing immune response is against an antigenic peptide delivered to the tumor cell by the non-mutated protein epitope peptide.


In certain embodiments, the tumor comprises cancer stem cells. In certain embodiments, the frequency of cancer stem cells in the tumor is reduced by administration of the non-mutated protein epitope therapeutic. In some embodiments, a method of reducing the frequency of cancer stem cells in a tumor in a subject, comprising administering to the subject a therapeutically effective amount of a non-mutated protein epitope therapeutic is provided.


In addition, in some aspects the invention provides a method of reducing the tumorigenicity of a tumor in a subject, comprising administering to the subject a therapeutically effective amount of a non-mutated protein epitope therapeutic described herein. In certain embodiments, the tumor comprises cancer stem cells. In some embodiments, the tumorigenicity of a tumor is reduced by reducing the frequency of cancer stem cells in the tumor. In some embodiments, the methods comprise using the non-mutated protein epitope therapeutic described herein. In certain embodiments, the frequency of cancer stem cells in the tumor is reduced by administration of a non-mutated protein epitope therapeutic described herein.


In some embodiments, the tumor is a solid tumor. In certain embodiments, the tumor is a tumor selected from the group consisting of: colorectal tumor, pancreatic tumor, lung tumor, ovarian tumor, liver tumor, breast tumor, kidney tumor, prostate tumor, neuroendocrine tumor, gastrointestinal tumor, melanoma, cervical tumor, bladder tumor, glioblastoma, and head and neck tumor. In certain embodiments, the tumor is a colorectal tumor. In certain embodiments, the tumor is an ovarian tumor. In some embodiments, the tumor is a breast tumor. In some embodiments, the tumor is a lung tumor. In certain embodiments, the tumor is a pancreatic tumor. In certain embodiments, the tumor is a melanoma tumor. In some embodiments, the tumor is a solid tumor.


The present invention further provides methods for treating cancer in a subject comprising administering to the subject a therapeutically effective amount of a non-mutated protein epitope therapeutic described herein.


In some embodiments, a method of treating cancer comprises redirecting an existing immune response to a new target, the method comprising administering to a subject a therapeutically effective amount of non-mutated protein epitope therapeutic, wherein the existing immune response is against an antigenic peptide delivered to the cancer cell by the non-mutated protein epitope peptide.


The present invention provides for methods of treating cancer comprising administering to a subject a therapeutically effective amount of a non-mutated protein epitope therapeutic described herein (e.g., a subject in need of treatment). In certain embodiments, the subject is a human. In certain embodiments, the subject has a cancerous tumor. In certain embodiments, the subject has had a tumor at least partially removed.


In certain embodiments, the cancer is a cancer selected from the group consisting of colorectal cancer, renal cancer, pancreatic cancer, lung cancer, ovarian cancer, liver cancer, breast cancer, kidney cancer, prostate cancer, gastrointestinal cancer, melanoma, cervical cancer, neuroendocrine cancer, bladder cancer, glioblastoma, triple-negative breast cancer (TNBC), smoldering myeloma (SMM), and head and neck cancer. In certain embodiments, the cancer is pancreatic cancer. In certain embodiments, the cancer is ovarian cancer. In certain embodiments, the cancer is colorectal cancer. In certain embodiments, the cancer is breast cancer. In certain embodiments, the cancer is prostate cancer. In certain embodiments, the cancer is lung cancer. In certain embodiments, the cancer is melanoma. In some embodiments, the cancer is a solid cancer. In some embodiments, the cancer comprises a solid tumor.


In some embodiments, the cancer is a hematologic cancer. In some embodiment, the cancer is selected from the group consisting of: acute myelogenous leukemia (AML), Hodgkin lymphoma, multiple myeloma, T-cell acute lymphoblastic leukemia (T-ALL), chronic lymphocytic leukemia (CLL), hairy cell leukemia, chronic myelogenous leukemia (CML), non-Hodgkin lymphoma, diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), and cutaneous T-cell lymphoma (CTCL).


In some embodiments, the non-mutated protein epitope therapeutic is administered as a combination therapy. Combination therapy with two or more therapeutic agents uses agents that work by different mechanisms of action, although this is not required. Combination therapy using agents with different mechanisms of action can result in additive or synergetic effects. Combination therapy can allow for a lower dose of each agent than is used in monotherapy, thereby reducing toxic side effects and/or increasing the therapeutic index of the agent(s). Combination therapy can decrease the likelihood that resistant cancer cells will develop. In some embodiments, combination therapy comprises a therapeutic agent that affects the immune response (e.g., enhances or activates the response) and a therapeutic agent that affects (e.g., inhibits or kills) the tumor/cancer cells.


In some embodiments, the combination of an agent described herein and at least one additional therapeutic agent results in additive or synergistic results. In some embodiments, the combination therapy results in an increase in the therapeutic index of the agent. In some embodiments, the combination therapy results in an increase in the therapeutic index of the additional therapeutic agent(s). In some embodiments, the combination therapy results in a decrease in the toxicity and/or side effects of the agent. In some embodiments, the combination therapy results in a decrease in the toxicity and/or side effects of the additional therapeutic agent(s).


In certain embodiments, in addition to administering a non-mutated protein epitope therapeutic described herein, the method or treatment further comprises administering at least one additional therapeutic agent. An additional therapeutic agent can be administered prior to, concurrently with, and/or subsequently to, administration of the agent. In some embodiments, the at least one additional therapeutic agent comprises 1, 2, 3, or more additional therapeutic agents.


Therapeutic agents that can be administered in combination with the non-mutated protein epitope therapeutic described herein include chemotherapeutic agents. Thus, in some embodiments, the method or treatment involves the administration of an agent described herein in combination with a chemotherapeutic agent or in combination with a cocktail of chemotherapeutic agents. Treatment with an agent can occur prior to, concurrently with, or subsequent to administration of chemotherapies. Combined administration can include co-administration, either in a single pharmaceutical formulation or using separate formulations, or consecutive administration in either order but generally within a time period such that all active agents can exert their biological activities simultaneously. Preparation and dosing schedules for such chemotherapeutic agents can be used according to manufacturers' instructions or as determined empirically by the skilled practitioner. Preparation and dosing schedules for such chemotherapy are also described in The Chemotherapy Source Book, 4th Edition, 2008, M. C. Perry, Editor, Lippincott, Williams & Wilkins, Philadelphia, Pa.


Useful classes of chemotherapeutic agents include, for example, anti-tubulin agents, auristatins, DNA minor groove binders, DNA replication inhibitors, alkylating agents (e.g., platinum complexes such as cisplatin, mono(platinum), bis(platinum) and tri-nuclear platinum complexes and carboplatin), anthracyclines, antibiotics, anti-folates, antimetabolites, chemotherapy sensitizers, duocarmycins, etoposides, fluorinated pyrimidines, ionophores, lexitropsins, nitrosoureas, platinols, purine antimetabolites, puromycins, radiation sensitizers, steroids, taxanes, topoisomerase inhibitors, vinca alkaloids, or the like. In certain embodiments, the second therapeutic agent is an alkylating agent, an antimetabolite, an antimitotic, a topoisomerase inhibitor, or an angiogenesis inhibitor.


Chemotherapeutic agents useful in the instant invention include, but are not limited to, alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamime; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, caminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytosine arabinoside, dideoxyuridine, doxifluridine, enocitabine, floxuridine, 5-FU; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenishers such as folinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK; razoxane; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethylamine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (Ara-C); taxoids, e.g. paclitaxel (TAXOL) and docetaxel (TAXOTERE); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; ibandronate; CPT11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; esperamicins; capecitabine (XELODA); and pharmaceutically acceptable salts, acids or derivatives of any of the above. Chemotherapeutic agents also include anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4 hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (FARESTON); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above. In certain embodiments, the additional therapeutic agent is cisplatin. In certain embodiments, the additional therapeutic agent is carboplatin.


In certain embodiments, the chemotherapeutic agent is a topoisomerase inhibitor. Topoisomerase inhibitors are chemotherapy agents that interfere with the action of a topoisomerase enzyme (e.g., topoisomerase I or II). Topoisomerase inhibitors include, but are not limited to, doxorubicin HCl, daunorubicin citrate, mitoxantrone HCl, actinomycin D, etoposide, topotecan HCl, teniposide (VM-26), and irinotecan, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these. In some embodiments, the additional therapeutic agent is irinotecan.


In certain embodiments, the chemotherapeutic agent is an anti-metabolite. An anti-metabolite is a chemical with a structure that is similar to a metabolite utilized for normal biochemical reactions, yet different enough to interfere with one or more normal functions of cells, such as cell division. Anti-metabolites include, but are not limited to, gemcitabine, fluorouracil, capecitabine, methotrexate sodium, ralitrexed, pemetrexed, tegafur, cytosine arabinoside, thioguanine, 5-azacytidine, 6 mercaptopurine, azathioprine, 6-thioguanine, pentostatin, fludarabine phosphate, and cladribine, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these. In certain embodiments, the additional therapeutic agent is gemcitabine.


In certain embodiments, the chemotherapeutic agent is an antimitotic agent, including, but not limited to, agents that bind tubulin. In some embodiments, the agent is a taxane. In certain embodiments, the agent is paclitaxel or docetaxel, or a pharmaceutically acceptable salt, acid, or derivative of paclitaxel or docetaxel. In certain embodiments, the agent is paclitaxel (TAXOL), docetaxel (TAXOTERE), albumin-bound paclitaxel (ABRAXANE), DHA-paclitaxel, or PG-paclitaxel. In certain alternative embodiments, the antimitotic agent comprises a vinca alkaloid, such as vincristine, vinblastine, vinorelbine, or vindesine, or pharmaceutically acceptable salts, acids, or derivatives thereof. In some embodiments, the antimitotic agent is an inhibitor of kinesin Eg5 or an inhibitor of a mitotic kinase such as Aurora A or Plk1. In certain embodiments, the additional therapeutic agent is paclitaxel. In some embodiments, the additional therapeutic agent is albumin-bound paclitaxel.


In some embodiments, an additional therapeutic agent comprises an agent such as a small molecule. For example, treatment can involve the combined administration of an agent of the present invention with a small molecule that acts as an inhibitor against tumor-associated antigens including, but not limited to, EGFR, HER2 (ErbB2), and/or VEGF. In some embodiments, an agent is administered in combination with a protein kinase inhibitor selected from the group consisting of: gefitinib (IRESSA), erlotinib (TARCEVA), sunitinib (SUTENT), lapatanib, vandetanib (ZACTIMA), AEE788, CI-1033, cediranib (RECENTIN), sorafenib (NEXAVAR), and pazopanib (GW786034B). In some embodiments, an additional therapeutic agent comprises an mTOR inhibitor. In another embodiment, the additional therapeutic agent is chemotherapy or other inhibitors that reduce the number of Treg cells. In certain embodiments, the therapeutic agent is cyclophosphamide or an anti-CTLA4 antibody. In another embodiment, the additional therapeutic reduces the presence of myeloid-derived suppressor cells. In a further embodiment, the additional therapeutic is carbotaxol. In another embodiment, the additional therapeutic agent shifts cells to a T helper 1 response. In a further embodiment, the additional therapeutic agent is ibrutinib.


In some embodiments, an additional therapeutic agent comprises a biological molecule, such as an antibody. For example, treatment can involve the combined administration of an agent of the present invention with antibodies against tumor-associated antigens including, but not limited to, antibodies that bind EGFR, HER2/ErbB2, and/or VEGF. In certain embodiments, the additional therapeutic agent is an antibody specific for a cancer stem cell marker. In certain embodiments, the additional therapeutic agent is an antibody that is an angiogenesis inhibitor (e.g., an anti-VEGF or VEGF receptor antibody). In certain embodiments, the additional therapeutic agent is bevacizumab (AVASTIN), ramucirumab, trastuzumab (HERCEPTIN), pertuzumab (OMNITARG), panitumumab (VECTIBIX), nimotuzumab, zalutumumab, or cetuximab (ERBITUX).


In certain embodiments, an additional therapeutic agent comprises a second immunotherapeutic agent. In some embodiments, the additional immunotherapeutic agent includes, but is not limited to, a colony stimulating factor, an interleukin, an antibody that blocks immunosuppressive functions (e.g., an anti-CTLA-4 antibody, anti-CD28 antibody, anti-CD3 antibody, anti-PD-1 antibody, anti-PD-L1 antibody, anti-TIGIT antibody), an antibody that enhances immune cell functions (e.g., an anti-GITR antibody, an anti-OX-40 antibody, an anti-CD40 antibody, or an anti-4-1BB antibody), a toll-like receptor (e.g., TLR4, TLR7, TLR9), a soluble ligand (e.g., GITRL, GITRL-Fc, OX-40L, OX-40L-Fc, CD40L, CD40L-Fc, 4-1BB ligand, or 4-1BB ligand-Fc), or a member of the B7 family (e.g., CD80, CD86). In some embodiments, the additional immunotherapeutic agent targets CTLA-4, CD28, CD3, PD-1, PD-L1, TIGIT, GITR, OX-40, CD-40, or 4-1BB.


In some embodiments, the additional therapeutic agent is an immune checkpoint inhibitor. In some embodiments, the immune checkpoint inhibitor is an anti-PD-1 antibody, an anti-PD-Ll antibody, an anti-CTLA-4 antibody, an anti-CD28 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, an anti-TIM3 antibody, an anti-GITR antibody, an anti-4-1BB antibody, or an anti-OX-40 antibody. In some embodiments, the additional therapeutic agent is an anti-TIGIT antibody. In some embodiments, the additional therapeutic agent is an anti-PD-1 antibody selected from the group consisting of: nivolumab (OPDIVO), pembrolizumab (KEYTRUDA), pidilzumab, MEDI0680, REGN2810, BGB-A317, and PDR001. In some embodiments, the additional therapeutic agent is an anti-PD-L1 antibody selected from the group consisting of: BMS935559 (MDX-1105), atexolizumab (MPDL3280A), durvalumab (MEDI4736), and avelumab (MSB0010718C). In some embodiments, the additional therapeutic agent is an anti-CTLA-4 antibody selected from the group consisting of: ipilimumab (YERVOY) and tremelimumab. In some embodiments, the additional therapeutic agent is an anti-LAG-3 antibody selected from the group consisting of: BMS-986016 and LAG525. In some embodiments, the additional therapeutic agent is an anti-OX-40 antibody selected from the group consisting of: MEDI6469, MEDI0562, and MOXR0916. In some embodiments, the additional therapeutic agent is an anti-4-1BB antibody selected from the group consisting of: PF-05082566.


In some embodiments, the non-mutated protein epitope therapeutic can be administered in combination with a biologic molecule selected from the group consisting of: adrenomedullin (AM), angiopoietin (Ang), BMPs, BDNF, EGF, erythropoietin (EPO), FGF, GDNF, granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), stem cell factor (SCF), GDF9, HGF, HDGF, IGF, migration-stimulating factor, myostatin (GDF-8), NGF, neurotrophins, PDGF, thrombopoietin, TGF-α, TGF-β, TNF-α, VEGF, P1GF, gamma-IFN, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-12, IL-15, and IL-18.


In some embodiments, treatment with a non-mutated protein epitope therapeutic described herein can be accompanied by surgical removal of tumors, removal of cancer cells, or any other surgical therapy deemed necessary by a treating physician.


In certain embodiments, treatment involves the administration of a non-mutated protein epitope therapeutic described herein in combination with radiation therapy. Treatment with an agent can occur prior to, concurrently with, or subsequent to administration of radiation therapy. Dosing schedules for such radiation therapy can be determined by the skilled medical practitioner.


Combined administration can include co-administration, either in a single pharmaceutical formulation or using separate formulations, or consecutive administration in either order but generally within a time period such that all active agents can exert their biological activities simultaneously.


It will be appreciated that the combination of a non-mutated protein epitope therapeutic described herein and at least one additional therapeutic agent can be administered in any order or concurrently. In some embodiments, the agent will be administered to patients that have previously undergone treatment with a second therapeutic agent. In certain other embodiments, the non-mutated protein epitope therapeutic and a second therapeutic agent will be administered substantially simultaneously or concurrently. For example, a subject can be given an agent while undergoing a course of treatment with a second therapeutic agent (e.g., chemotherapy). In certain embodiments, a non-mutated protein epitope therapeutic will be administered within 1 year of the treatment with a second therapeutic agent. It will further be appreciated that the two (or more) agents or treatments can be administered to the subject within a matter of hours or minutes (i.e., substantially simultaneously).


For the treatment of a disease, the appropriate dosage of a non-mutated protein epitope therapeutic described herein depends on the type of disease to be treated, the severity and course of the disease, the responsiveness of the disease, whether the agent is administered for therapeutic or preventative purposes, previous therapy, the patient's clinical history, and so on, all at the discretion of the treating physician. The non-mutated protein epitope therapeutic can be administered one time or over a series of treatments lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved (e.g., reduction in tumor size). Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient and will vary depending on the relative potency of an individual agent. The administering physician can determine optimum dosages, dosing methodologies, and repetition rates.


In some embodiments, a non-mutated protein epitope therapeutic can be administered at an initial higher “loading” dose, followed by one or more lower doses. In some embodiments, the frequency of administration can also change. In some embodiments, a dosing regimen can comprise administering an initial dose, followed by additional doses (or “maintenance” doses) once a week, once every two weeks, once every three weeks, or once every month. For example, a dosing regimen can comprise administering an initial loading dose, followed by a weekly maintenance dose of, for example, one-half of the initial dose. Or a dosing regimen can comprise administering an initial loading dose, followed by maintenance doses of, for example one-half of the initial dose every other week. Or a dosing regimen can comprise administering three initial doses for 3 weeks, followed by maintenance doses of, for example, the same amount every other week.


As is known to those of skill in the art, administration of any therapeutic agent can lead to side effects and/or toxicities. In some cases, the side effects and/or toxicities are so severe as to preclude administration of the particular agent at a therapeutically effective dose. In some cases, therapy must be discontinued, and other agents can be tried. However, many agents in the same therapeutic class display similar side effects and/or toxicities, meaning that the patient either has to stop therapy, or if possible, suffer from the unpleasant side effects associated with the therapeutic agent.


In some embodiments, the dosing schedule can be limited to a specific number of administrations or “cycles”. In some embodiments, the agent is administered for 3, 4, 5, 6, 7, 8, or more cycles. For example, the agent is administered every 2 weeks for 6 cycles, the agent is administered every 3 weeks for 6 cycles, the agent is administered every 2 weeks for 4 cycles, the agent is administered every 3 weeks for 4 cycles, etc. Dosing schedules can be decided upon and subsequently modified by those skilled in the art.


The present invention provides methods of administering to a subject a non-mutated protein epitope therapeutic described herein comprising using an intermittent dosing strategy for administering one or more agents, which can reduce side effects and/or toxicities associated with administration of an agent, chemotherapeutic agent, etc. In some embodiments, a method for treating cancer in a human subject comprises administering to the subject a therapeutically effective dose of a non-mutated protein epitope therapeutic in combination with a therapeutically effective dose of a chemotherapeutic agent, wherein one or both of the agents are administered according to an intermittent dosing strategy. In some embodiments, a method for treating cancer in a human subject comprises administering to the subject a therapeutically effective dose of a non-mutated protein epitope therapeutic in combination with a therapeutically effective dose of a second immunotherapeutic agent, wherein one or both of the agents are administered according to an intermittent dosing strategy. In some embodiments, the intermittent dosing strategy comprises administering an initial dose of a non-mutated protein epitope therapeutic to the subject, and administering subsequent doses of the agent about once every 2 weeks. In some embodiments, the intermittent dosing strategy comprises administering an initial dose of a non-mutated protein epitope therapeutic to the subject, and administering subsequent doses of the agent about once every 3 weeks. In some embodiments, the intermittent dosing strategy comprises administering an initial dose of a non-mutated protein epitope therapeutic to the subject, and administering subsequent doses of the agent about once every 4 weeks. In some embodiments, the agent is administered using an intermittent dosing strategy and the additional therapeutic agent is administered weekly.


The present invention provides compositions comprising the non-mutated protein epitope therapeutic described herein. The present invention also provides pharmaceutical compositions comprising a non-mutated protein epitope therapeutic described herein and a pharmaceutically acceptable vehicle. In some embodiments, the pharmaceutical compositions find use in immunotherapy. In some embodiments, the compositions find use in inhibiting tumor growth. In some embodiments, the pharmaceutical compositions find use in inhibiting tumor growth in a subject (e.g., a human patient). In some embodiments, the compositions find use in treating cancer. In some embodiments, the pharmaceutical compositions find use in treating cancer in a subject (e.g., a human patient).


Formulations are prepared for storage and use by combining an antigen therapeutic of the present invention with a pharmaceutically acceptable vehicle (e.g., a carrier or excipient). Those of skill in the art generally consider pharmaceutically acceptable carriers, excipients, and/or stabilizers to be inactive ingredients of a formulation or pharmaceutical composition. Exemplary formulations are listed in WO 2015/095811.


Suitable pharmaceutically acceptable vehicles include, but are not limited to, nontoxic buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine; preservatives such as octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butyl or benzyl alcohol, alkyl parabens, such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol; low molecular weight polypeptides (e.g., less than about 10 amino acid residues); proteins such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; carbohydrates such as monosaccharides, disaccharides, glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes such as Zn-protein complexes; and non-ionic surfactants such as TWEEN or polyethylene glycol (PEG). (Remington: The Science and Practice of Pharmacy, 22st Edition, 2012, Pharmaceutical Press, London.). In one embodiment, the vehicle is 5% dextrose in water.


The pharmaceutical compositions described herein can be administered in any number of ways for either local or systemic treatment. Administration can be topical by epidermal or transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders; pulmonary by inhalation or insufflation of powders or aerosols, including by nebulizer, intratracheal, and intranasal; oral; or parenteral including intravenous, intraarterial, intratumoral, subcutaneous, intraperitoneal, intramuscular (e.g., injection or infusion), or intracranial (e.g., intrathecal or intraventricular).


The therapeutic formulation can be in unit dosage form. Such formulations include tablets, pills, capsules, powders, granules, solutions or suspensions in water or non-aqueous media, or suppositories.


The non-mutated protein epitope peptides described herein can also be entrapped in microcapsules. Such microcapsules are prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions as described in Remington: The Science and Practice of Pharmacy, 22st Edition, 2012, Pharmaceutical Press, London.


In certain embodiments, pharmaceutical formulations include a non-mutated protein epitope therapeutic described herein complexed with liposomes. Methods to produce liposomes are known to those of skill in the art. For example, some liposomes can be generated by reverse phase evaporation with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes can be extruded through filters of defined pore size to yield liposomes with the desired diameter.


In certain embodiments, sustained-release preparations comprising the non-mutated protein epitope peptides described herein can be produced. Suitable examples of sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing an agent, where the matrices are in the form of shaped articles (e.g., films or microcapsules). Examples of sustained-release matrices include polyesters, hydrogels such as poly(2-hydroxyethyl-methacrylate) or poly(vinyl alcohol), polylactides, copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(−)-3-hydroxybutyric acid.


VIII. Kits

The non-mutated protein epitope therapeutic described herein can be provided in kit form together with instructions for administration. Typically the kit would include the desired antigen therapeutic in a container, in unit dosage form and instructions for administration. Additional therapeutics, for example, cytokines, lymphokines, checkpoint inhibitors, antibodies, can also be included in the kit. Other kit components that can also be desirable include, for example, a sterile syringe, booster dosages, and other desired excipients.


The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of non-critical parameters that can be changed or modified to yield alternative embodiments according to the invention. All patents, patent applications, and printed publications listed herein are incorporated herein by reference in their entirety.


EXAMPLES
Example 1: Identification of Mutant Sequences with Immunogenic Potential

Applicants have discovered that the following epitopes are recurrent in cancer patients.










TABLE 1





ERV element



(nomenclature see:



ncbi.nlm.nih.gov/



pmc/articles/



PMC3113919/)
Translated Sequence







ERVH-2:
MGNLPPSIPPSPLACVLKNLKPLQLTPDLKPKCLIFFCNTAWPQYKLDN


retroviral matrix
GSKWPENGTFDFSILQDLNNSCRKMGKWSEVPDVQAFFYTSVPS





ERVH48-1: coat
MPNRAIRLQAVLEIITNQTASALEMLAQQQNQMRAAIYQNRLALDYLL


protein
AEEGAGCGKFNISNCCLNIGNNGEEVLEIASNIRKVARVPVQTWEGWD



PANLLGGWFSNLGGFKMLVGTVIFITGVLLFLPCGIPLKLLLKLQLTS





ERVH48-1:
MACIYPTTFYTSLPTKSLNMGISLTTILILSVAVLLSTAAPPSCRECYQSL


syncytin
HYRGEMQQYFTYHTHIERSCYGNLIEECVESGKSYYKVKNLGVCGSRN



GAICPRGKQWLCFTKIGQWGVNTQVLEDIKREQIIAKAKASKPTTPPEN



RPRHFHSFIQKL





ERVH-2: gag
MARSAATLRRFTALDPKRSKGRLILNIHFITQSAPDIK





ERVE-4: reverse
RLFLTKPGKEIGPALAQWWPKVCAEDNPPGLAVNQAPVLREVKPEAQ


transcriptase
PVRQNQYPVPREALEGIQVHLKHLRTFGIIVPCQSPWNTPLLPVPKPGT



KDYRPVQDLRLVNQATVTFHPTVPNPYTLLGLLPAKDSWFTCLDLKD



AFFSIRLAPESQKLFAFQWEDPGSGVTTHYTWTRLPQGFKNFPHHLWG



GTGSRPPKVSCQRPRLRVVPVHRQPPAGTPHGSRVRQRNRRPASAPGG



LWV





ERVE-4: reverse
MAVGCVKGTDALLQHLEDYGYKVSKKKAQICRQQVRYLGFTIRQREC


transcriptase
SLGSERKQVICNLLEPKTRRQLRELLGAVGFCRLWIPNFAVLAKPLVPS



YKGG





ERVE-4: protease
NADLLAAAIRGVPLKGQGNGGSRKNTQSDRPRLQRNQCAYCKETGH



WKDKCPQLKEKQGGSEQKTPDKDEGALFNLAEGLLDRRGPGSRAPKE



PMVRMTVGGKDIKFLVNTGAEHSVVTTPVAPLSKKAIDIIGATGVLTK



QAFCLPRTCSVGGHEVIHQFLYIPDCPLPLLGRDLLSKLRAIFLYQARLF



TTEVAWNRSYHGPDSSPRGRVATLPNQTRQRDRASSGPVVAKSMRRR



QPSWIGSQSSSCTQGS





ERVE-4: unknown
SLFLHKTSVREVLSATIPATFLGSLTWKRGD





ERVK-5: gag, env,
MQNEAIEQVRAICLRAWGKIQDPGTAFPINSIRQGSKEPYPDFVARLQD


pol
AAQKSITDDNARKVIVELMAYENANPECQSAIKPLKGKVPAGVDVITE



YVKACDGIGGAMHKAMLMAQAMRGLTLGGQVRTFGKKCYNCGQIG



HLKRSCPVLNKQNIINQAITAKNKKPSGLCPKCGKGKHWANQCHSKFD



KDGQPLSGNRKRGQPQAPQQTGAFPVQLFVPQGFQGQQPLQKIPPLQG



VSQLQQSNSCPAPQQAAPQ





ERVI-1: envelope
MEWIKYSICTLNKSNCYACAHGRPEAQIVPFPLRWSSSRPSMGCMVAL



FQDSTAWGNISCQALSLLYPEVQHPAGQPPRAIQLPSPNVSFISCLS









For each epitope, the full-length amino acid sequence of the non-mutated protein epitope was derived. Any constituent 9mer or 10mer not found in the germline protein sequence was flagged and scored for binding potential on six common HLA alleles (HLA-A01:01, HLA-A02:01. HLA-A03:01, HLA-A24:02, HLA-B07:02, and HLA-B08:01) using available algorithms. Any peptide scoring better than 1000 nM was nominated.










TABLE 2





ERV element



(nomenclature see:



http://www.ncbi.nlm.



nih.gov/pmc/articles/



PMC3113919/)
Translated Sequence







ERVH-2: retroviral
MGNLPPSIPPSPLACVLKNLKPLQLTPDLKPKCLIFFCNTAWPQYKLD


matrix
NGSKWPENGTFDFSILQDLNNSCRKMGKWSEVPDVQAFFYTSVPS





ERVH48-1: coat
MPNRAIRLQAVLEIITNQTASALEMLAQQQNQMRAAIYQNRLALDY


protein
LLAEEGAGCGKFNISNCCLNIGNNGEEVLEIASNIRKVARVPVQTWE



GWDPANLLGGWFSNLGGFKMLVGTVIFITGVLLFLPCGIPLKLLLKL



QLTS





ERVH48-1: syncytin
MACIYPTTFYTSLPTKSLNMGISLTTILILSVAVLLSTAAPPSCRECYQS



LHYRGEMQQYFTYHTHIERSCYGNLIEECVESGKSYYKVKNLGVCG



SRNGAICPRGKQWLCFTKIGQWGVNTQVLEDIKREQIIAKAKASKPT



TPPENRPRHFHSFIQKL





ERVH-2: gag
MARSAATLRRFTALDPKRSKGRLILNIHFITQSAPDIK





ERVE-4: reverse
RLFLTKPGKEIGPALAQWWPKVCAEDNPPGLAVNQAPVLREVKPEA


transcriptase
QPVRQNQYPVPREALEGIQVHLKHLRTFGIIVPCQSPWNTPLLPVPKP



GTKDYRPVQDLRLVNQATVTFHPTVPNPYTLLGLLPAKDSWFTCLD



LKDAFFSIRLAPESQKLFAFQWEDPGSGVTTHYTWTRLPQGFKNFPH



HLWGGTGSRPPKVSCQRPRLRVVPVHRQPPAGTPHGSRVRQRNRRP



ASAPGGLWV





ERVE-4: reverse
MAVGCVKGTDALLQHLEDYGYKVSKKKAQICRQQVRYLGFTIRQR


transcriptase
ECSLGSERKQVICNLLEPKTRRQLRELLGAVGFCRLWIPNFAVLAKPL



VPSYKGG





ERVE-4: protease
NADLLAAAIRGVPLKGQGNGGSRKNTQSDRPRLQRNQCAYCKETGH



WKDKCPQLKEKQGGSEQKTPDKDEGALFNLAEGLLDRRGPGSRAPK



EPMVRMTVGGKDIKFLVNTGAEHSVVTTPVAPLSKKAIDIIGATGVL



TKQAFCLPRTCSVGGHEVIHQFLYIPDCPLPLLGRDLLSKLRAIFLYQ



ARLFTTEVAWNRSYHGPDSSPRGRVATLPNQTRQRDRASSGPVVAK



SMRRRQPSWIGSQSSSCTQGS





ERVE-4: unknown
SLFLHKTSVREVLSATIPATFLGSLTWKRGD





ERVK-5: gag, env,
MQNEAIEQVRAICLRAWGKIQDPGTAFPINSIRQGSKEPYPDFVARLQ


pol
DAAQKSITDDNARKVIVELMAYENANPECQSAIKPLKGKVPAGVDVI



TEYVKACDGIGGAMHKAMLMAQAMRGLTLGGQVRTFGKKCYNCG



QIGHLKRSCPVLNKQNIINQAITAKNKKPSGLCPKCGKGKHWANQC



HSKFDKDGQPLSGNRKRGQPQAPQQTGAFPVQLFVPQGFQGQQPLQ



KIPPLQGVSQLQQSNSCPAPQQAAPQ





ERVI-1: envelope
MEWIKYSICTLNKSNCYACAHGRPEAQIVPFPLRWSSSRPSMGCMVA



LFQDSTAWGNISCQALSLLYPEVQHPAGQPPRAIQLPSPNVSFISCLS


















TABLE 3





Over-




expressed
UCSC



gene
ID
Full Sequence







TYR
uc001pcs.3
MLLAVLYCLLWSFQTSAGHFPRACVSSKNLMEKECCPPWSGDRSP




CGQLSGRGSCQNILLSNAPLGPQFPFTGVDDRESWPSVFYNRTCQC




SGNFMGFNCGNCKFGFWGPNCTERRLLVRRNIFDLSAPEKDKFFAY




LTLAKHTISSDYVIPIGTYGQMKNGSTPMFNDINIYDLFVWMHYYV




SMDALLGGSEIWRDIDFAHEAPAFLPWHRLFLLRWEQEIQKLTGDE




NFTIPYWDWRDAEKCDICTDEYMGGQHPTNPNLLSPASFFSSWQIV




CSRLEEYNSHQSLCNGTPEGPLRRNPGNHDKSRTPRLPSSADVEFCL




SLTQYESGSMDKAANFSFRNTLEGFASPLTGIADASQSSMHNALHI




YMNGTMSQVQGSANDPIFLLHHAFVDSIFEQWLRRHRPLQEVYPE




ANAPIGHNRESYMVPFIPLYRNGDFFISSKDLGYDYSYLQDSDPDSF




QDYIKSYLEQASRIWSWLLGAAMVGAVLTALLAGLVSLLCRHKRK




QLPEEKQPLLMEKEDYHSLYQSHL





MAGEC1
uc004fbt.3
MGDKDMPTAGMPSLLQSSSESPQSCPEGEDSQSPLQIPQSSPESDDT




LYPLQSPQSRSEGEDSSDPLQRPPEGKDSQSPLQIPQSSPEGDDTQSP




LQNSQSSPEGKDSLSPLEISQSPPEGEDVQSPLQNPASSFFSSALLSIF




QSSPESTQSPFEGFPQSVLQIPVSAASSSTLVSIFQSSPESTQSPFEGFP




QSPLQIPVSRSFSSTLLSIFQSSPERTQSTFEGFAQSPLQIPVSPSSSSTL




LSLFQSFSERTQSTFEGFAQSSLQIPVSPSFSSTLVSLFQSSPERTQSTF




EGFPQSPLQIPVSSSSSSTLLSLFQSSPERTHSTFEGFPQSLLQIPMTSS




FSSTLLSIFQSSPESAQSTFEGFPQSPLQIPGSPSFSSTLLSLFQSSPERT




HSTFEGFPQSPLQIPMTSSFSSTLLSILQSSPESAQSAFEGFPQSPLQIP




VSSSFSYTLLSLFQSSPERTHSTFEGFPQSPLQIPVSSSSSSSTLLSLFQ




SSPECTQSTFEGFPQSPLQIPQSPPEGENTHSPLQIVPSLPEWEDSLSP




HYFPQSPPQGEDSLSPHYFPQSPPQGEDSLSPHYFPQSPQGEDSLSPH




YFPQSPPQGEDSMSPLYFPQSPLQGEEFQSSLQSPVSICSSSTPSSLPQ




SFPESSQSPPEGPVQSPLHSPQSPPEGMHSQSPLQSPESAPEGEDSLSP




LQIPQSPLEGEDSLSSLHFPQSPPEWEDSLSPLHFPQFPPQGEDFQSSL




QSPVSICSSSTSLSLPQSFPESPQSPPEGPAQSPLQRPVSSFFSYTLASL




LQSSHESPQSPPEGPAQSPLQSPVSSFPSSTSSSLSQSSPVSSFPSSTSS




SLSKSSPESPLQSPVISFSSSTSLSPFSEESSSPVDEYTSSSDTLLESDSL




TDSESLIESEPLFTYTLDEKVDELARFLLLKYQVKQPITKAEMLTNVI




SRYTGYFPVIFRKAREFIEILFGISLREVDPDDSYVFVNTLDLTSEGCL




SDEQGMSQNRLLILILSIIFIKGTYASEEVIWDVLSGIGVRAGREHFA




FGEPRELLTKVWVQEHYLEYREVPNSSPPRYEFLWGPRAHSEVIKR




KVVEFLAMLKNTVPITFPSSYKDALKDVEERAQAIIDTTDDSTATES




ASSSVMSPSFSSE





MAGEA10
uc022cgz.1
MPRAPKRQRCMPEEDLQSQSETQGLEGAQAPLAVEEDASSSTSTSS




SFPSSFPSSSSSSSSSCYPLIPSTPEEVSADDETPNPPQSAQIACSSPSV




VASLPLDQSDEGSSSQKEESPSTLQVLPDSESLPRSEIDEKVTDLVQF




LLFKYQMKEPITKAEILESVIRNYEDHFPLLFSEASECMLLVFGIDVK




EVDPTGHSFVLVTSLGLTYDGMLSDVQSMPKTGILILILSIVFIEGYC




TPEEVIWEALNMMGLYDGMEHLIYGEPRKLLTQDWVQENYLEYR




QVPGSDPARYEFLWGPRAHAEIRKMSLLKFLAKVNGSDPRSFPLW




YEEALKDEEERAQDRIATTDDTTAMASASSSATGSFSYPE





MAGEB17
uc031tgu.1
MPRGQASKRRAREKRRQARGEDQCLGGAQATAAEKEKLPSSSSPA




CQSPPQSFPNAGIPQESQRASYPSSPASAVSLTSSDEGAKGQKGESP




NSFHGPSSSESTGRDLLNTKTGELVQFLLNKYIRKEPITREAMLKVI




NRKYKQHFPEILRRSTENVEVVFGLYLKEMDPSRQSYVLVGKLDFP




NQGSLSDGGGFPLSGLLMVLLSTIFMHGNRATEEEMWECLNALGM




YKGRKHFIYGEPQELVTKDLVREGYLEYQQVPSSDPPRYEFLWGPR




ARAETSKMKVLEFVAKLNDTVASTYKSRYEEALREEEEQARARAV




ARDSARARASRSFQP





MAGEA4
uc022cgu.1
MLPLSVGLWVPIAQLLPALLPAALTRVIMSSEQKSQHCKPEEGVEA




QEEALGLVGAQAPTTEEQEAAVSSSSPLVPGTLEEVPAAESAGPPQS




PQGASALPTTISFTCWRQPNEGSSSQEEEGPSTSPDAESLFREALSNK




VDELAHFLLRKYRAKELVTKAEMLERVIKNYKRCFPVIFGKASESL




KMIFGIDVKEVDPASNTYTLVTCLGLSYDGLLGNNQIFPKTGLLIIVL




GTIAMEGDSASEEEIWEELGVMGVYDGREHTVYGEPRKLLTQDWV




QENYLEYRQVPGSNPARYEFLWGPRALAETSYVKVLEHVVRVNAR




VRIAYPSLREAALLEEEEGV





MABEB16
uc022bus.1
MSQDQESPRCTHDQHLQTFSETQSLEVAQVSKALEKTLLSSSHPLV




PGKLKEAPAAKAESPLEVPQSFCSSSIAVTTTSSSESDEASSNQEEED




SPSSSEDTSDPRNVPADALDQKVAFLVNFMLHKCQMKKPITKADM




LKIIIKDDESHFSEILLRASEHLEMIFGLDVVEVDPTTHCYGLFIKLGL




TYDGMLSGEKGVPKTGLLIIVLGVIFMKGNRATEEEVWEVLNLTGV




YSGKKHFIFGEPRMLITKDFVKEKYLEYQQVANSDPARYEFLWGPR




AKAETSKMKVLEFVAKVHGSYPHSFPSQYAEALKEEEERARARI





MAGEA1
uc022chs.1
MSLEQRSLHCKPEEALEAQQEALGLVCVQAATSSSSPLVLGTLEEV




PTAGSTDPPQSPQGASAFPTTINFTRQRQPSEGSSSREEEGPSTSCILE




SLFRAVITKKVADLVGFLLLKYRAREPVTKAEMLESVIKNYKHCFP




EIFGKASESLQLVFGIDVKEADPTGHSYVLVTCLGLSYDGLLGDNQI




MPKTGFLIIVLVMIAMEGGHAPEEEIWEELSVMEVYDGREHSAYGE




PRKLLTQDLVQEKYLEYRQVPDSDPARYEFLWGPRALAETSYVKV




LEYVIKVSARVRFFFPSLREAALREEEEGV





MAGEA8
uc022cgo.1
MLLGQKSQRYKAEEGLQAQGEAPGLMDVQIPTAEEQKAASSSSTLI




MGTLEEVTDSGSPSPPQSPEGASSSLTVTDSTLWSQSDEGSSSNEEE




GPSTSPDPAHLESLFREALDEKVAELVRFLLRKYQIKEPVTKAEMLE




SVIKNYKNHFPDIFSKASECMQVIFGIDVKEVDPAGHSYILVTCLGL




SYDGLLGDDQSTPKTGLLIIVLGMILMEGSRAPEEAIWEALSVMGL




YDGREHSVYWKLRKLLTQEWVQENYLEYRQAPGSDPVRYEFLWG




PRALAETSYVKVLEHVVRVNARVRISYPSLHEEALGEEKGV





MAGEB4
uc004dcb.3
MPRGQKSKLRAREKRQRTRGQTQDLKVGQPTAAEKEESPSSSSSVL




RDTASSSLAFGIPQEPQREPPTTSAAAAMSCTGSDKGDESQDEENAS




SSQASTSTERSLKDSLTRKTKMLVQFLLYKYKMKEPTTKAEMLKIIS




KKYKEHFPEIFRKVSQRTELVFGLALKEVNPTTHSYILVSMLGPNDG




NQSSAWTLPRNGLLMPLLSVIFLNGNCAREEEIWEFLNMLGIYDGK




RHLIFGEPRKLITQDLVQEKYLEYQQVPNSDPPRYQFLWGPRAHAE




TSKMKVLEFLAKVNDTTPNNFPLLYEEALRDEEERAGARPRVAAR




RGTTAMTSAYSRATSSSSSQPM





CT45A5
uc011mvu.2
MTDKTEKVAVDPETVFKRPRECDSPSYQKRQRMALLARKQGAGD




SLIAGSAMSKEKKLMTGHAIPPSQLDSQIDDFTGFSKDGMMQKPGS




NAPVGGNVTSNFSGDDLECRGIASSPKSQQEINADIKCQVVKEIRCL




GRKYEKIFEMLEGVQGPTAVRKRFFESIIKEAARCMRRDFVKHLKK




KLKRMI





ALPPL2
uc002vss.4
MQGPWVLLLLGLRLQLSLGIIPVEEENPDFWNRQAAEALGAAKKL




QPAQTAAKNLIIFLGDGMGVSTVTAARILKGQKKDKLGPETFLAMD




RFPYVALSKTYSVDKHVPDSGATATAYLCGVKGNFQTIGLSAAARF




NQCNTTRGNEVISVMNRAKKAGKSVGVVTTTRVQHASPAGAYAH




TVNRNWYSDADVPASARQEGCQDIATQLISNMDIDVILGGGRKYM




FPMGTPDPEYPDDYSQGGTRLDGKNLVQEWLAKHQGARYVWNRT




ELLQASLDPSVTHLMGLFEPGDMKYEIHRDSTLDPSLMEMTEAALL




LLSRNPRGFFLFVEGGRIDHGHHESRAYRALTETIMFDDAIERAGQL




TSEEDTLSLVTADHSHVFSFGGYPLRGSSIFGLAPGKARDRKAYTVL




LYGNGPGYVLKDGARPDVTESESGSPEYRQQSAVPLDGETHAGED




VAVFARGPQAHLVHGVQEQTFIAHVMAFAACLEPYTACDLAPRAG




TTDAAHPGPSVVPALLPLLAGTLLLLGTATAP





MMP13
uc001ph1.3
MHPGVLAAFLFLSWTHCRALPLPSGGDEDDLSEEDLQFAERYLRSY




YHPTNLAGILKENAASSMTERLREMQSFFGLEVTGKLDDNTLDVM




KKPRCGVPDVGEYNVFPRTLKWSKMNLTYRIVNYTPDMTHSEVEK




AFKKAFKVWSDVTPLNFTRLHDGIADIMISFGIKEHGDFYPFDGPSG




LLAHAFPPGPNYGGDAHFDDDETWTSSSKGYNLFLVAAHEFGHSL




GLDHSKDPGALMFPIYTYTGKSHFMLPDDDVQGIQSLYGPGDEDPN




PKHPKTPDKCDPSLSLDAITSLRGETMIFKDRFFWRLHPQQVDAELF




LTKSFWPELPNRIDAAYEHPSHDLIFIFRGRKFWALNGYDILEGYPK




KISELGLPKEVKKISAAVHFEDTGKTLLFSGNQVWRYDDTNHIMDK




DYPRLIEEDFPGIGDKVDAVYEKNGYIYFFNGPIQFEYSIWSNRIVRV




MPANSILWC





CTAG1B
uc004fmf.1
MQAEGRGTGGSTGDADGPGGPGIPDGPGGNAGGPGEAGATGGRG




PRGAGAARASGPGGGAPRGPHGGAASGLNGCCRCGARGPESRLLE




FYLAMPFATPMEAELARRSLAQDAPPLPVPGVLLKEFTVSGNILTIR




LTAADHRQLQLSISSCLQQLSLLMWITQCFLPVFLAQPPSGQRR





DCT
uc010afh.3
MSPLWWGFLLSCLGCKILPGAQGQFPRVCMTVDSLVNKECCPRLG




AESANVCGSQQGRGQCTEVRADTRPWSGPYILRNQDDRELWPRKF




FHRTCKCTGNFAGYNCGDCKFGWTGPNCERKKPPVIRQNIHSLSPQ




EREQFLGALDLAKKRVHPDYVITTQHWLGLLGPNGTQPQFANCSV




YDFFVWLHYYSVRDTLLGPGRPYRAIDFSHQGPAFVTWHRYHLLC




LERDLQRLIGNESFALPYWNFATGRNECDVCTDQLFGAARPDDPTL




ISRNSRFSSWETVCDSLDDYNHLVTLCNGTYEGLLRRNQMGRNSM




KLPTLKDIRDCLSLQKFDNPPFFQNSTFSFRNALEGFDKADGTLDSQ




VMSLHNLVHSFLNGTNALPHSAANDPIFVVISNRLLYNATTNILEHV




RKEKATKELPSLHVLVLHSFTDAIFDEWMKRFNPPADAWPQELAPI




GHNRMYNMVPFFPPVTNEELFLTSDQLGYSYAIDLPVSVEETPGWP




TTLLVVMGTLVALVGLFVLLAFLQYRRLRKGYTPLMETHLSSKRY




TEEA





CLDN6
uc021tbb.1
MASAGMQILGVVLTLLGWVNGLVSCALPMWKVTAFIGNSIVVAQ




VVWEGLWMSCVVQSTGQMQCKVYDSLLALPQDLQAARALCVIAL




LVALFGLLVYLAGAKCTTCVEEKDSKARLVLTSGIVFVISGVLTLIP




VCWTAHAIIRDFYNPLVAEAQKRELGASLYLGWAASGLLLLGGGL




LCCTCPSGGSQGPSHYMARYSTSAPAISRGPSEYPTKNYV





MLANA
uc003zjo.1
MPREDAHFIYGYPKKGHGHSYTTAEEAAGIGILTVILGVLLLIGCWY




CRRRNGYRALMDKSLHVGTQCALTRRCPQEGFDHRDSKVSLQEK




NCEPVVPNAPPAYEKLSAEQSPPPYSP





AFP
uc003hgz.1
MKWVESIFLIFLLNFTESRTLHRNEYGIASILDSYQCTAEISLADLATI




FFAQFVQEATYKEVSKMVKDALTAIEKPTGDEQSSGCLENQLPAFL




EELCHEKEILEKYGHSDCCSQSEEGRHNCFLAHKKPTPASIPLFQVP




EPVTSCEAYEEDRETFMNKFIYEIARRHPFLYAPTILLWAARYDKIIP




SCCKAENAVECFQTKAATVTKELRESSLLNQHACAVMKNFGTRTF




QAITVTKLSQKFTKVNFTEIQKLVLDVAHVHEHCCRGDVLDCLQD




GEKIMSYICSQQDTLSNKITECCKLTTLERGQCIIHAENDEKPEGLSP




NLNRFLGDRDFNQFSSGEKNIFLASFVHEYSRRHPQLAVSVILRVAK




GYQELLEKCFQTENPLECQDKGEEELQKYIQESQALAKRSCGLFQK




LGEYYLQNAFLVAYTKKAPQLTSSELMAITRKMAATAATCCQLSE




DKLLACGEGAADIIIGHLCIRHEMTPVNPGVGQCCTSSYANRRPCFS




SLVVDETYVPPAFSDDKFIFHKDLCQAQGVALQTMKQEFLINLVKQ




KPQITEEQLEAVIADFSGLLEKCCQGQEQEVCFAEEGQKLISKTRAA




LGV





DKK4
uc003xpb.3
MVAAVLLGLSWLCSPLGALVLDFNNIRSSADLHGARKGSQCLSDT




DCNTRKFCLQPRDEKPFCATCRGLRRRCQRDAMCCPGTLCVNDVC




TTMEDATPILERQLDEQDGTHAEGTTGHPVQENQPKRKPSIKKSQG




RKGQEGESCLRTFDCGPGLCCARHFWTKICKPVLLEGQVCSRRGH




KDTAQAPEIFQRCDCGPGLLCRSQLTSNRQHARLRVCQKIEKL





ASCL2
uc021qcf.1
MDGGTLPRSAPPAPPVPVGCAARRRPASPELLRCSRRRRPATAETG




GGAAAVARRNERERNRVKLVNLGFQALRQHVPHGGASKKLSKVE




TLRSAVEYIRALQRLLAEHDAVRNALAGGLRPQAVRPSAPRGPPGT




TPVAASPSRASSSPGRGGSSEPGSPRSAYSSDDSGCEGALSPAERELL




DFSSWLGGY





GAGE1
uc004dok.2
MSWRGRSTYYWPRPRRYVQPPEMIGPMRPEQFSDEVEPATPEEGEP




ATQRQDPAAAQEGEDEGASAGQGPKPEADSQEQGHPQTGCECEDG




PDGQEMDPPNPEEVKTPEEEMRSHYVAQTGILWLLMNNCFLNLSP




RKP





GAGE10
uc010nir.1
MSWRGRSTYRSRPRLYVEPPEMIGPMLPEQFSDEVEPATPEEGEPA




TQRQDPAAAQEGEDEGASAGQGPKPEADSQEQVHPKTGCECGDGP




DGQEMGLPNPEEVKRPEEGEKQSQC





SLC45A2
uc003jid.3
MGSNSGQAGRHIYKSLADDGPFDSVEPPKRPTSRLIMHSMAMFGRE




FCYAVEAAYVTPVLLSVGLPSSLYSIVWFLSPILGFLLQPVVGSASD




HCRSRWGRRRPYILTLGVMMLVGMALYLNGATVVAALIANPRRK




LVWAISVTMIGVVLFDFAADFIDGPIKAYLFDVCSHQDKEKGLHYH




ALFTGFGGALGYLLGAIDWAHLELGRLLGTEFQVMFFFSALVLTLC




FTVHLCSISEAPLTEVAKGIPPQQTPQDPPLSSDGMYEYGSIEKVKN




GYVNPELAMQGAKNKNHAEQTRRAMTLKSLLRALVNMPPHYRYL




CISHLIGWTAFLSNMLFFTDFMGQIVYRGDPYSAHNSTEFLIYERGV




EVGCWGFCINSVFSSLYSYFQKVLVSYIGLKGLYFTGYLLFGLGTGF




IGLFPNVYSTLVLCSLFGVMSSTLYTVPFNLITEYHREEEKERQQAP




GGDPDNSVRGKGMDCATLTCMVQLAQILVGGGLGFLVNTAGTVV




VVVITASAVALIGCCFVALFVRYVD





PAGE5
uc004duj.3
MQAPWAGNRGWAGTREEVRDMSEHVTRSQSSERGNDQESSQPVG




PVIVQQPTEEKRQEEEPPTDNQGIAPSGEIKNEGAPAVQGTDVEAFQ




QELALLKIEDAPGDGPDVREGTLPTFDPTKVLEAGEGQL





PAGE2
uc004duf.1
MSELLRARSQSSERGNDQESSQPVGSVIVQEPTEEKRQEEEPPTDNQ




GIAPSGEIENQAVPAFQGPDMEAFQQELALLKIEDEPGDGPDVREGI




MPTFDLTKVLEAGDAQP





PMEL
uc001siq.3
MDLVLKRCLLHLAVIGALLAVGATKVPRNQDWLGVSRQLRTKAW




NRQLYPEWTEAQRLDCWRGGQVSLKVSNDGPTLIGANASFSIALNF




PGSQKVLPDGQVIWVNNTIINGSQVWGGQPVYPQETDDACIFPDGG




PCPSGSWSQKRSFVYVWKTWGQYWQVLGGPVSGLSIGTGRAMLG




THTMEVTVYHRRGSRSYVPLAHSSSAFTITDQVPFSVSVSQLRALD




GGNKHFLRNQPLTFALQLHDPSGYLAEADLSYTWDFGDSSGTLISR




ALVVTHTYLEPGPVTAQVVLQAAIPLTSCGSSPVPGTTDGHRPTAE




APNTTAGQVPTTEVVGTTPGQAPTAEPSGTTSVQVPTTEVISTAPVQ




MPTAESTGMTPEKVPVSEVMGTTLAEMSTPEATGMTPAEVSIVVLS




GTTAAQVTTTEWVETTARELPIPEPEGPDASSIMSTESITGSLGPLLD




GTATLRLVKRQVPLDCVLYRYGSFSVTLDIVQGIESAEILQAVPSGE




GDAFELTVSCQGGLPKEACMEISSPGCQPPAQRLCQPVLPSPACQL




VLHQILKGGSGTYCLNVSLADTNSLAVVSTQLIMPVPGILLTGQEA




GLGQVPLIVGILLVLMAVVLASLIYRRRLMKQDFSVPQLPHSSSHW




LRLPRIFCSCPIGENSPLLSGQQV









For each epitope, the full-length amino acid sequence of the non-mutated protein epitope was derived. Any constituent 9mer or 10mer not found in the germline protein sequence was flagged and scored for binding potential on six common HLA alleles (HLA-A01:01, HLA-A02:01. HLA-A03:01, HLA-A24:02, HLA-B07:02, and HLA-B08:01) using available algorithms. Any peptide scoring better than 1000 nM was nominated.











TABLE 4





Over-




expressed




gene
UCSC ID
Full Sequence







TYR
uc001pcs.3
MLLAVLYCLLWSFQTSAGHFPRACVSSKNLMEKECCPPWSGDRS




PCGQLSGRGSCQNILLSNAPLGPQFPFTGVDDRESWPSVFYNRTC




QCSGNFMGFNCGNCKFGFWGPNCTERRLLVRRNIFDLSAPEKDK




FFAYLTLAKHTISSDYVIPIGTYGQMKNGSTPMFNDINIYDLFVW




MHYYVSMDALLGGSEIWRDIDFAHEAPAFLPWHRLFLLRWEQEI




QKLTGDENFTIPYWDWRDAEKCDICTDEYMGGQHPTNPNLLSPA




SFFSSWQIVCSRLEEYNSHQSLCNGTPEGPLRRNPGNHDKSRTPRL




PSSADVEFCLSLTQYESGSMDKAANFSFRNTLEGFASPLTGIADAS




QSSMHNALHIYMNGTMSQVQGSANDPIFLLHHAFVDSIFEQWLR




RHRPLQEVYPEANAPIGHNRESYMVPFIPLYRNGDFFISSKDLGYD




YSYLQDSDPDSFQDYIKSYLEQASRIWSWLLGAAMVGAVLTALL




AGLVSLLCRHKRKQLPEEKQPLLMEKEDYHSLYQSHL





MAGEC1
uc004fbt.3
MGDKDMPTAGMPSLLQSSSESPQSCPEGEDSQSPLQIPQSSPESDD




TLYPLQSPQSRSEGEDSSDPLQRPPEGKDSQSPLQIPQSSPEGDDTQ




SPLQNSQSSPEGKDSLSPLEISQSPPEGEDVQSPLQNPASSFFSSALL




SIFQSSPESTQSPFEGFPQSVLQIPVSAASSSTLVSIFQSSPESTQSPFE




GFPQSPLQIPVSRSFSSTLLSIFQSSPERTQSTFEGFAQSPLQIPVSPS




SSSTLLSLFQSFSERTQSTFEGFAQSSLQIPVSPSFSSTLVSLFQSSPE




RTQSTFEGFPQSPLQIPVSSSSSSTLLSLFQSSPERTHSTFEGFPQSLL




QIPMTSSFSSTLLSIFQSSPESAQSTFEGFPQSPLQIPGSPSFSSTLLSL




FQSSPERTHSTFEGFPQSPLQIPMTSSFSSTLLSILQSSPESAQSAFEG




FPQSPLQIPVSSSFSYTLLSLFQSSPERTHSTFEGFPQSPLQIPVSSSS




SSSTLLSLFQSSPECTQSTFEGFPQSPLQIPQSPPEGENTHSPLQIVPS




LPEWEDSLSPHYFPQSPPQGEDSLSPHYFPQSPPQGEDSLSPHYFPQ




SPQGEDSLSPHYFPQSPPQGEDSMSPLYFPQSPLQGEEFQSSLQSPV




SICSSSTPSSLPQSFPESSQSPPEGPVQSPLHSPQSPPEGMHSQSPLQ




SPESAPEGEDSLSPLQIPQSPLEGEDSLSSLHFPQSPPEWEDSLSPLH




FPQFPPQGEDFQSSLQSPVSICSSSTSLSLPQSFPESPQSPPEGPAQSP




LQRPVSSFFSYTLASLLQSSHESPQSPPEGPAQSPLQSPVSSFPSSTS




SSLSQSSPVSSFPSSTSSSLSKSSPESPLQSPVISFSSSTSLSPFSEESSS




PVDEYTSSSDTLLESDSLTDSESLIESEPLFTYTLDEKVDELARFLL




LKYQVKQPITKAEMLTNVISRYTGYFPVIFRKAREFIEILFGISLRE




VDPDDSYVFVNTLDLTSEGCLSDEQGMSQNRLLILILSIIFIKGTYA




SEEVIWDVLSGIGVRAGREHFAFGEPRELLTKVWVQEHYLEYRE




VPNSSPPRYEFLWGPRAHSEVIKRKVVEFLAMLKNTVPITFPSSYK




DALKDVEERAQAIIDTTDDSTATESASSSVMSPSFSSE





MAGEA10
uc022cgz.1
MPRAPKRQRCMPEEDLQSQSETQGLEGAQAPLAVEEDASSSTSTS




SSFPSSFPSSSSSSSSSCYPLIPSTPEEVSADDETPNPPQSAQIACSSPS




VVASLPLDQSDEGSSSQKEESPSTLQVLPDSESLPRSEIDEKVTDLV




QFLLFKYQMKEPITKAEILESVIRNYEDHFPLLFSEASECMLLVFGI




DVKEVDPTGHSFVLVTSLGLTYDGMLSDVQSMPKTGILILILSIVFI




EGYCTPEEVIWEALNMMGLYDGMEHLIYGEPRKLLTQDWVQEN




YLEYRQVPGSDPARYEFLWGPRAHAEIRKMSLLKFLAKVNGSDP




RSFPLWYEEALKDEEERAQDRIATTDDTTAMASASSSATGSFSYP




E





MAGEB17
uc031tgu.1
MPRGQASKRRAREKRRQARGEDQCLGGAQATAAEKEKLPSSSSP




ACQSPPQSFPNAGIPQESQRASYPSSPASAVSLTSSDEGAKGQKGE




SPNSFHGPSSSESTGRDLLNTKTGELVQFLLNKYIRKEPITREAML




KVINRKYKQHFPEILRRSTENVEVVFGLYLKEMDPSRQSYVLVGK




LDFPNQGSLSDGGGFPLSGLLMVLLSTIFMHGNRATEEEMWECL




NALGMYKGRKHFIYGEPQELVTKDLVREGYLEYQQVPSSDPPRY




EFLWGPRARAETSKMKVLEFVAKLNDTVASTYKSRYEEALREEE




EQARARAVARDSARARASRSFQP





MAGEA4
uc022cgu.1
MLPLSVGLWVPIAQLLPALLPAALTRVIMSSEQKSQHCKPEEGVE




AQEEALGLVGAQAPTTEEQEAAVSSSSPLVPGTLEEVPAAESAGP




PQSPQGASALPTTISFTCWRQPNEGSSSQEEEGPSTSPDAESLFREA




LSNKVDELAHFLLRKYRAKELVTKAEMLERVIKNYKRCFPVIFGK




ASESLKMIFGIDVKEVDPASNTYTLVTCLGLSYDGLLGNNQIFPKT




GLLIIVLGTIAMEGDSASEEEIWEELGVMGVYDGREHTVYGEPRK




LLTQDWVQENYLEYRQVPGSNPARYEFLWGPRALAETSYVKVLE




HVVRVNARVRIAYPSLREAALLEEEEGV





MABEB16
uc022bus.1
MSQDQESPRCTHDQHLQTFSETQSLEVAQVSKALEKTLLSSSHPL




VPGKLKEAPAAKAESPLEVPQSFCSSSIAVTTTSSSESDEASSNQEE




EDSPSSSEDTSDPRNVPADALDQKVAFLVNFMLHKCQMKKPITK




ADMLKIIIKDDESHFSEILLRASEHLEMIFGLDVVEVDPTTHCYGLF




IKLGLTYDGMLSGEKGVPKTGLLIIVLGVIFMKGNRATEEEVWEV




LNLTGVYSGKKHFIFGEPRMLITKDFVKEKYLEYQQVANSDPARY




EFLWGPRAKAETSKMKVLEFVAKVHGSYPHSFPSQYAEALKEEE




ERARARI





MAGEA1
uc022chs.1
MSLEQRSLHCKPEEALEAQQEALGLVCVQAATSSSSPLVLGTLEE




VPTAGSTDPPQSPQGASAFPTTINFTRQRQPSEGSSSREEEGPSTSCI




LESLFRAVITKKVADLVGFLLLKYRAREPVTKAEMLESVIKNYKH




CFPEIFGKASESLQLVFGIDVKEADPTGHSYVLVTCLGLSYDGLLG




DNQIMPKTGFLIIVLVMIAMEGGHAPEEEIWEELSVMEVYDGREH




SAYGEPRKLLTQDLVQEKYLEYRQVPDSDPARYEFLWGPRALAE




TSYVKVLEYVIKVSARVRFFFPSLREAALREEEEGV





MAGEA8
uc022cgo.1
MLLGQKSQRYKAEEGLQAQGEAPGLMDVQIPTAEEQKAASSSST




LIMGTLEEVTDSGSPSPPQSPEGASSSLTVTDSTLWSQSDEGSSSNE




EEGPSTSPDPAHLESLFREALDEKVAELVRFLLRKYQIKEPVTKAE




MLESVIKNYKNHFPDIFSKASECMQVIFGIDVKEVDPAGHSYILVT




CLGLSYDGLLGDDQSTPKTGLLIIVLGMILMEGSRAPEEAIWEALS




VMGLYDGREHSVYWKLRKLLTQEWVQENYLEYRQAPGSDPVR




YEFLWGPRALAETSYVKVLEHVVRVNARVRISYPSLHEEALGEE




KGV





MAGEB4
uc004dcb.3
MPRGQKSKLRAREKRQRTRGQTQDLKVGQPTAAEKEESPSSSSS




VLRDTASSSLAFGIPQEPQREPPTTSAAAAMSCTGSDKGDESQDEE




NASSSQASTSTERSLKDSLTRKTKMLVQFLLYKYKMKEPTTKAE




MLKIISKKYKEHFPEIFRKVSQRTELVFGLALKEVNPTTHSYILVS




MLGPNDGNQSSAWTLPRNGLLMPLLSVIFLNGNCAREEEIWEFLN




MLGIYDGKRHLIFGEPRKLITQDLVQEKYLEYQQVPNSDPPRYQF




LWGPRAHAETSKMKVLEFLAKVNDTTPNNFPLLYEEALRDEEER




AGARPRVAARRGTTAMTSAYSRATSSSSSQPM





CT45A5
uc011mvu.2
MTDKTEKVAVDPETVFKRPRECDSPSYQKRQRMALLARKQGAG




DSLIAGSAMSKEKKLMTGHAIPPSQLDSQIDDFTGFSKDGMMQKP




GSNAPVGGNVTSNFSGDDLECRGIASSPKSQQEINADIKCQVVKEI




RCLGRKYEKIFEMLEGVQGPTAVRKRFFESIIKEAARCMRRDFVK




HLKKKLKRMI





ALPPL2
uc002vss.4
MQGPWVLLLLGLRLQLSLGIIPVEEENPDFWNRQAAEALGAAKK




LQPAQTAAKNLIIFLGDGMGVSTVTAARILKGQKKDKLGPETFLA




MDRFPYVALSKTYSVDKHVPDSGATATAYLCGVKGNFQTIGLSA




AARFNQCNTTRGNEVISVMNRAKKAGKSVGVVTTTRVQHASPA




GAYAHTVNRNWYSDADVPASARQEGCQDIATQLISNMDIDVILG




GGRKYMFPMGTPDPEYPDDYSQGGTRLDGKNLVQEWLAKHQG




ARYVWNRTELLQASLDPSVTHLMGLFEPGDMKYEIHRDSTLDPS




LMEMTEAALLLLSRNPRGFFLFVEGGRIDHGHHESRAYRALTETI




MFDDAIERAGQLTSEEDTLSLVTADHSHVFSFGGYPLRGSSIFGLA




PGKARDRKAYTVLLYGNGPGYVLKDGARPDVTESESGSPEYRQQ




SAVPLDGETHAGEDVAVFARGPQAHLVHGVQEQTFIAHVMAFA




ACLEPYTACDLAPRAGTTDAAHPGPSVVPALLPLLAGTLLLLGTA




TAP





MMP13
uc001ph1.3
MHPGVLAAFLFLSWTHCRALPLPSGGDEDDLSEEDLQFAERYLRS




YYHPTNLAGILKENAASSMTERLREMQSFFGLEVTGKLDDNTLD




VMKKPRCGVPDVGEYNVFPRTLKWSKMNLTYRIVNYTPDMTHS




EVEKAFKKAFKVWSDVTPLNFTRLHDGIADIMISFGIKEHGDFYPF




DGPSGLLAHAFPPGPNYGGDAHFDDDETWTSSSKGYNLFLVAAH




EFGHSLGLDHSKDPGALMFPIYTYTGKSHFMLPDDDVQGIQSLYG




PGDEDPNPKHPKTPDKCDPSLSLDAITSLRGETMIFKDRFFWRLHP




QQVDAELFLTKSFWPELPNRIDAAYEHPSHDLIFIFRGRKFWALN




GYDILEGYPKKISELGLPKEVKKISAAVHFEDTGKTLLFSGNQVW




RYDDTNHIMDKDYPRLIEEDFPGIGDKVDAVYEKNGYIYFFNGPI




QFEYSIWSNRIVRVMPANSILWC





CTAG1B
uc004fmf.1
MQAEGRGTGGSTGDADGPGGPGIPDGPGGNAGGPGEAGATGGR




GPRGAGAARASGPGGGAPRGPHGGAASGLNGCCRCGARGPESRL




LEFYLAMPFATPMEAELARRSLAQDAPPLPVPGVLLKEFTVSGNI




LTIRLTAADHRQLQLSISSCLQQLSLLMWITQCFLPVFLAQPPSGQ




RR





DCT
uc010afh.3
MSPLWWGFLLSCLGCKILPGAQGQFPRVCMTVDSLVNKECCPRL




GAESANVCGSQQGRGQCTEVRADTRPWSGPYILRNQDDRELWPR




KFFHRTCKCTGNFAGYNCGDCKFGWTGPNCERKKPPVIRQNIHSL




SPQEREQFLGALDLAKKRVHPDYVITTQHWLGLLGPNGTQPQFA




NCSVYDFFVWLHYYSVRDTLLGPGRPYRAIDFSHQGPAFVTWHR




YHLLCLERDLQRLIGNESFALPYWNFATGRNECDVCTDQLFGAA




RPDDPTLISRNSRFSSWETVCDSLDDYNHLVTLCNGTYEGLLRRN




QMGRNSMKLPTLKDIRDCLSLQKFDNPPFFQNSTFSFRNALEGFD




KADGTLDSQVMSLHNLVHSFLNGTNALPHSAANDPIFVVISNRLL




YNATTNILEHVRKEKATKELPSLHVLVLHSFTDAIFDEWMKRFNP




PADAWPQELAPIGHNRMYNMVPFFPPVTNEELFLTSDQLGYSYAI




DLPVSVEETPGWPTTLLVVMGTLVALVGLFVLLAFLQYRRLRKG




YTPLMETHLSSKRYTEEA





CLDN6
uc021tbb.1
MASAGMQILGVVLTLLGWVNGLVSCALPMWKVTAFIGNSIVVA




QVVWEGLWMSCVVQSTGQMQCKVYDSLLALPQDLQAARALCV




IALLVALFGLLVYLAGAKCTTCVEEKDSKARLVLTSGIVFVISGVL




TLIPVCWTAHAIIRDFYNPLVAEAQKRELGASLYLGWAASGLLLL




GGGLLCCTCPSGGSQGPSHYMARYSTSAPAISRGPSEYPTKNYV





MLANA
uc003zjo.1
MPREDAHFIYGYPKKGHGHSYTTAEEAAGIGILTVILGVLLLIGC




WYCRRRNGYRALMDKSLHVGTQCALTRRCPQEGFDHRDSKVSL




QEKNCEPVVPNAPPAYEKLSAEQSPPPYSP





AFP
uc003hgz.1
MKWVESIFLIFLLNFTESRTLHRNEYGIASILDSYQCTAEISLADLA




TIFFAQFVQEATYKEVSKMVKDALTAIEKPTGDEQSSGCLENQLP




AFLEELCHEKEILEKYGHSDCCSQSEEGRHNCFLAHKKPTPASIPL




FQVPEPVTSCEAYEEDRETFMNKFIYEIARRHPFLYAPTILLWAAR




YDKIIPSCCKAENAVECFQTKAATVTKELRESSLLNQHACAVMK




NFGTRTFQAITVTKLSQKFTKVNFTEIQKLVLDVAHVHEHCCRGD




VLDCLQDGEKIMSYICSQQDTLSNKITECCKLTTLERGQCIIHAEN




DEKPEGLSPNLNRFLGDRDFNQFSSGEKNIFLASFVHEYSRRHPQL




AVSVILRVAKGYQELLEKCFQTENPLECQDKGEEELQKYIQESQA




LAKRSCGLFQKLGEYYLQNAFLVAYTKKAPQLTSSELMAITRKM




AATAATCCQLSEDKLLACGEGAADIIIGHLCIRHEMTPVNPGVGQ




CCTSSYANRRPCFSSLVVDETYVPPAFSDDKFIFHKDLCQAQGVA




LQTMKQEFLINLVKQKPQITEEQLEAVIADFSGLLEKCCQGQEQE




VCFAEEGQKLISKTRAALGV





DKK4
uc003xpb.3
MVAAVLLGLSWLCSPLGALVLDFNNIRSSADLHGARKGSQCLSD




TDCNTRKFCLQPRDEKPFCATCRGLRRRCQRDAMCCPGTLCVND




VCTTMEDATPILERQLDEQDGTHAEGTTGHPVQENQPKRKPSIKK




SQGRKGQEGESCLRTFDCGPGLCCARHFWTKICKPVLLEGQVCSR




RGHKDTAQAPEIFQRCDCGPGLLCRSQLTSNRQHARLRVCQKIEK




L





ASCL2
uc021qcf.1
MDGGTLPRSAPPAPPVPVGCAARRRPASPELLRCSRRRRPATAET




GGGAAAVARRNERERNRVKLVNLGFQALRQHVPHGGASKKLSK




VETLRSAVEYIRALQRLLAEHDAVRNALAGGLRPQAVRPSAPRGP




PGTTPVAASPSRASSSPGRGGSSEPGSPRSAYSSDDSGCEGALSPA




ERELLDFSSWLGGY





GAGE1
uc004dok.2
MSWRGRSTYYWPRPRRYVQPPEMIGPMRPEQFSDEVEPATPEEG




EPATQRQDPAAAQEGEDEGASAGQGPKPEADSQEQGHPQTGCEC




EDGPDGQEMDPPNPEEVKTPEEEMRSHYVAQTGILWLLMNNCFL




NLSPRKP





GAGE10
uc010nir.1
MSWRGRSTYRSRPRLYVEPPEMIGPMLPEQFSDEVEPATPEEGEP




ATQRQDPAAAQEGEDEGASAGQGPKPEADSQEQVHPKTGCECG




DGPDGQEMGLPNPEEVKRPEEGEKQSQC





SLC45A2
uc003jid.3
MGSNSGQAGRHIYKSLADDGPFDSVEPPKRPTSRLIMHSMAMFG




REFCYAVEAAYVTPVLLSVGLPSSLYSIVWFLSPILGFLLQPVVGS




ASDHCRSRWGRRRPYILTLGVMMLVGMALYLNGATVVAALIAN




PRRKLVWAISVTMIGVVLFDFAADFIDGPIKAYLFDVCSHQDKEK




GLHYHALFTGFGGALGYLLGAIDWAHLELGRLLGTEFQVMFFFS




ALVLTLCFTVHLCSISEAPLTEVAKGIPPQQTPQDPPLSSDGMYEY




GSIEKVKNGYVNPELAMQGAKNKNHAEQTRRAMTLKSLLRALV




NMPPHYRYLCISHLIGWTAFLSNMLFFTDFMGQIVYRGDPYSAHN




STEFLIYERGVEVGCWGFCINSVFSSLYSYFQKVLVSYIGLKGLYF




TGYLLFGLGTGFIGLFPNVYSTLVLCSLFGVMSSTLYTVPFNLITE




YHREEEKERQQAPGGDPDNSVRGKGMDCATLTCMVQLAQILVG




GGLGFLVNTAGTVVVVVITASAVALIGCCFVALFVRYVD





PAGE5
uc004duj.3
MQAPWAGNRGWAGTREEVRDMSEHVTRSQSSERGNDQESSQPV




GPVIVQQPTEEKRQEEEPPTDNQGIAPSGEIKNEGAPAVQGTDVEA




FQQELALLKIEDAPGDGPDVREGTLPTFDPTKVLEAGEGQL





PAGE2
uc004duf.1
MSELLRARSQSSERGNDQESSQPVGSVIVQEPTEEKRQEEEPPTDN




QGIAPSGEIENQAVPAFQGPDMEAFQQELALLKIEDEPGDGPDVR




EGIMPTFDLTKVLEAGDAQP





PMEL
uc001siq.3
MDLVLKRCLLHLAVIGALLAVGATKVPRNQDWLGVSRQLRTKA




WNRQLYPEWTEAQRLDCWRGGQVSLKVSNDGPTLIGANASFSIA




LNFPGSQKVLPDGQVIWVNNTIINGSQVWGGQPVYPQETDDACIF




PDGGPCPSGSWSQKRSFVYVWKTWGQYWQVLGGPVSGLSIGTG




RAMLGTHTMEVTVYHRRGSRSYVPLAHSSSAFTITDQVPFSVSVS




QLRALDGGNKHFLRNQPLTFALQLHDPSGYLAEADLSYTWDFGD




SSGTLISRALVVTHTYLEPGPVTAQVVLQAAIPLTSCGSSPVPGTT




DGHRPTAEAPNTTAGQVPTTEVVGTTPGQAPTAEPSGTTSVQVPT




TEVISTAPVQMPTAESTGMTPEKVPVSEVMGTTLAEMSTPEATG




MTPAEVSIVVLSGTTAAQVTTTEWVETTARELPIPEPEGPDASSIM




STESITGSLGPLLDGTATLRLVKRQVPLDCVLYRYGSFSVTLDIVQ




GIESAEILQAVPSGEGDAFELTVSCQGGLPKEACMEISSPGCQPPA




QRLCQPVLPSPACQLVLHQILKGGSGTYCLNVSLADTNSLAVVST




QLIMPVPGILLTGQEAGLGQVPLIVGILLVLMAVVLASLIYRRRLM




KQDFSVPQLPHSSSHWLRLPRIFCSCPIGENSPLLSGQQV


















TABLE 5





Virus,
Genbank



Gene
Accession
Sequence







HPV-16,
NC_001526
MHQKRTAMFQDPQERPRKLPQLCTELQTTIHDIILECVYCKQQLL


E6

RREVYDFAFRDLCIVYRDGNPYAVCDKCLKFYSKISEYRHYCYS




LYGTTLEQQYNKPLCDLLIRCINCQKPLCPEEKQRHLDKKQRFH




NIRGRWTGRCMSCCRSSRTRRETQL





HPV-16,
NC_001526
MHGDTPTLHEYMLDLQPETTDLYCYEQLNDSSEEEDEIDGPAGQ


E7

AEPDRAHYNIVTFCCKCDSTLRLCVQSTHVDIRTLEDLLMGTLGI




VCPICSQKP





EBV, LF2
KC207813
MAEAYPGGAHAALASRRSSFRNSLRRLRPTEKPDTSFMRGVWK




YEIFPSYVRVTNKQVLQLDAQCQELPPCPSVGQILSFKLPSFSFNT




TTYGSRYFTVAFLFFGAEDNEVFLKPFFVMHSDQDIVLSVLNPRS




LFIEKGKFTWYIVPIRLVKNPYLYLQILPGQSDIQLTRSCTQSGDK




LNTSEPQIFLSGSPVTSQDECLPYLLAQHTPPFLKSYARIHTFPGK




VCPVNAIRRGKGYVRVSVDTPDLKREGPLNVKVGMTLLDDVIIA




FRYNPYPKSHWRWDGESTDIRYFGSPVIIPPNFITELEYNNTYEAP




LSSKITAIVVSHSSNPVFYVYPQEWKPGQTLKLTVRNISNNPITIV




TGQSMAQAFFIYAGDPSISTIMRRYIQRQGCALTLPGNIVVESSSL




PTFERINKTFNGNIVASEGTL





EBV,
KC207813
MSGGLFYNPFLRPNKGLLKKPDKEYLRLIPKCFQTPGAAGVVDV


BALF5

RGPQPPLCFYQDSLTVVGGDEDGKGMWWRQRAQEGTARPEAD




THGSPLDFHVYDILETVYTHEKCAVIPSDKQGYVVPCGIVIKLLG




RRKADGASVCVNVFGQQAYFYASAPQGLDVEFAVLSALKASTF




DRRTPCRVSVEKVTRRSIMGYGNHAGDYHKITLSHPNSVCHVAT




WLQDKHGCRIFEANVDATRRFVLDNDFVTFGWYSCRRAIPRLQ




HRDSYAELEYDCEVGDLSVRREDSSWPSYQALAFDIECLGEEGF




PTATNEADLILQISCVLWSTGEEAGRYRRILLTLGTCEDIEGVEVY




EFPSELDMLYAFFQLIRDLSVEIVTGYNVANFDWPYILDRARHIY




SINPASLGKIRAGGVCEVRRPHDAGKGFLRANTKVRITGLIPIDM




YAVCRDKLSLSDYKLDTVARHLLGAKKEDVHYKEIPRLFAAGPE




GRRRLGMYCVQDSALVMDLLNHFVIHVEVAEIAKIAHIPCRRVL




DDGQQIRVFSCLLAAAQKENFILPMPSASDRDGYQGATVIQPLSG




FYNSPVLVVDFASLYPSIIQAHNLCYSTMITPGEEHRLAGLRPGED




YESFRLTGGVYHFVKKHVHESFLASLLTSWLAKRKAIKKLLAAC




EDPRQRTILDKQQLAIKCTCNAVYGFTGVANGLFPCLSIAETVTL




QGRTMLERAKAFVEALSPANLQALAPSPDAWAPLNPEGQLRVIY




GDTDSLFIECRGFSESETLRFAEALAAHTTRSLFVAPISLEAEKTFS




CLMLITKKRYVGVLTDGKTLMKGVELVRKTACKFVQTRCRRVL




DLVLADARVKEAASLLSHRPFQESFTQGLPVGFLPVIDILNQAYT




DLREGRVPMGELCFSTELSRKLSAYKSTQMPHLAVYQKFVERNE




ELPQIHDRIQYVFVEPKGGVKGARKTEMAEDPAYAERHGVPVA




VDHYFDKLLQGAANILQCLFDNNSGAALSVLQNFTARPPF





EBV,
KC207813
MAGARRRARCPASAGCAYSARPPPLSTRGRRISAGSGQPRWWP


RPMS1

WGSPPPLDTRYRRPGPGRRARSCLHAGPRGRPPHSRTRARRTSPG




AGGGGWRGGSCTSQR





EBV, A73
KC207813
MSMPPKGFLKKEMKPETRLLNKPPTVLTRPAMFCAWKLYSRKM




PSRSKTLEARCSSRPPCDSPACQTRDTGCPRRSGTGRRGWRARRL




GKESWFADAWRMARYWGCAVKAAAQSAFSASTASPEEL





EBV,
KC207813
MTRRRVLSVVVLLAALACRLGAQTPEQPAPPATTVQPTATRQQT


BALF4

SFPFRVCELSSHGDLFRFSSDIQCPSFGTRENHTEGLLMVFKDNIIP




YSFKVRSYTKIVTNILIYNGWYADSVTNRHEEKFSVDSYETDQM




DTIYQCYNAVKMTKDGLTRVYVDRDGVNITVNLKPTGGLANGV




RRYASQTELYDAPGWLIWTYRTRTTVNCLITDMMAKSNSPFDFF




VTTTGQTVEMSPFYDGKNKETFHERADSFHVRTNYKIVDYDNR




GTNPQGERRAFLDKGTYTLSWKLENRTAYCPLQHWQTFDSTIAT




ETGKSIHFVTDEGTSSFVTNTTVGIELPDAFKCIEEQVNKTMHEK




YEAVQDRYTKGQEAITYFITSGGLLLAWLPLTPRSLATVKNLTEL




TTPTSSPPSSPSPPAPPAARGSTSAAVLRRRRRDAGNATTPVPPAA




PGKSLGTLNNPATVQIQFAYDSLRRQINRMLGDLARAWCLEQKR




QNMVLRELTKINPTTVMSSIYGKAVAAKRLGDVISVSQCVPVNQ




ATVTLRKSMRVPGSETMCYSRPLVSFSFINDTKTYEGQLGTDNEI




FLTKKMTEVCQATSQYYFQSGNEIHVYNDYHHFKTIELDGIATL




QTFISLNTSLIENIDFASLELYSRDEQRASNVFDLEGIFREYNFQAQ




NIAGLRKDLDNAVSNGRNQFVDGLGELMDSLGSVGQSITNLVST




VGGLFSSLVSGFISFFKNPFGGMLILVLVAGVVILVISLTRRTRQM




SQQPVQMLYPGIDELAQQHASGEGPGINPISKTELQAIMLALHEQ




NQEQKRAAQRAAGPSVASRALQAARDRFPGLRRRRYHDPETAA




ALLGEAETEF





EBV,
KC207813
MSGLLAAAYSQVYALAVELSVCARLDPRSLDVAAVVRNAGLLA


BALF3

ELEAILLPRLRRQNDRACSALSLELVHLLENSREASAALLAPGRK




GTRVPPLRTPSVAYSVEFYGGHKVDVSLCLINDIEILMKRINSVFY




CMSHTMGLESLERALDLLGRFRGVSPIPDPRLYITSVPCWRCVGE




LMVLPNHGNPSTAEGTHVSCNHLAVPVNPEPVSGLFENEVRQAG




LGHLLEAEEKARPGGPEEGAVPGPGRPEAEGATRALDTYNVFST




VPPEVAELSELLYWNSGGHAIGATGQGEGGGHSRLSALFARERR




LALVRRACEEALAGARLTHLFDAVAPGATERLFCGGVYSSSGDA




VEALKADCAAAFTAHPQYRAILQKRNELYTRLNRAMQRLGRGE




EEASRESPEVPRPAGAREPGPSGALSDALKRKEQYLRQVATEGL




AKLQSCLAQQSETLTETLCLRVWGDVVYWELARMRNHFLYRR




AFVSGPWEDRRAGEGAAFENSKYIKTHLFTQTLSSEHLHALTHSL




YTFITGPLAEESGLFPPPSNVALARCCDAAGTLPHQKAFLTSLIWP




GIEPSDWIETSFNSFYSVPGGSLASSQQILCRALREAVLTVSLYNK




TWGRSLILRRADAVSPGQALPPDGLYLTYDSDRPLILLYKGRGW




VFKDLYALLYLHLQMRDDSA





EBV,
KC207813
APGYAVEAVEGGLYPVARLDAWPYQGSQERLLVRQRTCGVTA


BARF0

ASQGHVAGWGKEPALLRQGPRDEGVQAVRQRVQVLRAQGLGK




QVCFDVLGILKGGTLAGAPVLPGTRDEGPSVEEVVAHAGQLPVD




HVPPDAQAQGLGQGLALLRQAGLQLGQTLGGHLAQVLLLALER




VREGAGRAGLSCPSRPGHLRALPGRLLLASAQPLHGSVEPRVEL




VPLLQDGPVLGVRREGGGAVRLQRLHRVARGAVDPAAEEPLCG




PGSHGIKQVSQPCPRQRLLAGPPHQGQATLPGKQGREAGMSATL




PLPRCTDSMAARVPIEELREFRHLRGHCREDVVGVQRSGRPLCL




RPPRARDRALLWAARPRLLLSLQQVPEPSLPDFILKQSRDRLRIH




RHRQVVTGDVGPLCRGRVAVVGQNHQLAHTAPAGHRGDVEAR




VWDGTYAPKAAQQIQGPFQALQPHGVRHAIKHAIDSLH









For each epitope, the full-length amino acid sequence of the non-mutated protein epitope was derived. Any constituent 9mer or 10mer not found in the germline protein sequence was flagged and scored for binding potential on six common HLA alleles (HLA-A01:01, HLA-A02:01. HLA-A03:01, HLA-A24:02, HLA-B07:02, and HLA-B08:01) using available algorithms. Any peptide scoring better than 1000 nM was nominated.











TABLE 6





Peptide
Virus, Gene
Affinity







CMSCCRSSR
HPV-16, E6
A03.01 = 610





CPEEKQRHL
HPV-16, E6
B07.02 = 720





CVYCKQQLL
HPV-16, E6
B08.01 = 880





CVYCKQQLLR
HPV-16, E6
A03.01 = 250





CYSLYGTTL
HPV-16, E6
A24.02 = 290





DKKQRFHNI
HPV-16, E6
B08.01 = 200





EYRHYCYSL
HPV-16, E6
A24.02 = 350; B08.01 = 360





FAFRDLCIV
HPV-16, E6
A02.01 = 150





IILECVYCK
HPV-16, E6
A03.01 = 150





ISEYRHYCY
HPV-16, E6
A01.01 = 81





IVYRDGNPY
HPV-16, E6
A03.01 = 700





IVYRDGNPYA
HPV-16, E6
A02.01 = 760





KFYSKISEY
HPV-16, E6
A03.01 = 670





KISEYRHYCY
HPV-16, E6
A03.01 = 570





KLPQLCTEL
HPV-16, E6
A02.01 = 130





LIRCINCQK
HPV-16, E6
A03.01 = 230





LLIRCINCQK
HPV-16, E6
A03.01 = 130





MHQKRTAMF
HPV-16, E6
A24.02 = 980; B08.01 = 580





NPYAVCDKCL
HPV-16, E6
B07.02 = 550





QYNKPLCDLL
HPV-16, E6
A24.02 = 520





RFHNIRGRW
HPV-16, E6
A24.02 = 620





RGRWTGRCM
HPV-16, E6
B07.02 = 720





RPRKLPQLC
HPV-16, E6
B07.02 = 310





RPRKLPQLCT
HPV-16, E6
B07.02 = 79





SEYRHYCYSL
HPV-16, E6
B08.01 = 390





SSRTRRETQL
HPV-16, E6
B08.01 = 230





TIHDIILECV
HPV-16, E6
A02.01 = 140





TTLEQQYNK
HPV-16, E6
A03.01 = 520





VYDFAFRDL
HPV-16, E6
A24.02 = 600





GIVCPICSQK
HPV-16, E7
A03.01 = 200





GTLGIVCPI
HPV-16, E7
A02.01 = 120





HGDTPTLHEY
HPV-16, E7
A01.01 = 270





IVCPICSQK
HPV-16, E7
A03.01 = 200





LLMGTLGIV
HPV-16, E7
A02.01 = 20





RAHYNIVTF
HPV-16, E7
A24.02 = 330





RLCVQSTHV
HPV-16, E7
A02.01 = 770





TLEDLLMGTL
HPV-16, E7
A02.01 = 480





TLHEYMLDL
HPV-16, E7
A02.01 = 95





TPTLHEYML
HPV-16, E7
B07.02 = 490





YMLDLQPET
HPV-16, E7
A02.01 = 7





YMLDLQPETT
HPV-16, E7
A02.01 = 25





AALASRRSSF
EBV, LF2
B07.02 = 160





ALASRRSSF
EBV, LF2
B07.02 = 290; B08.01 = 41





ALASRRSSFR
EBV, LF2
A03.01 = 160





ALTLPGNIVV
EBV, LF2
A02.01 = 470





APLSSKITA
EBV, LF2
B07.02 = 490





APLSSKITAI
EBV, LF2
B07.02 = 31





AQHTPPFLK
EBV, LF2
A03.01 = 140





AYPGGAHAAL
EBV, LF2
A24.02 = 470





CPSVGQILSF
EBV, LF2
B07.02 = 190





EVFLKPFFV
EBV, LF2
A02.01 = 210





FFGAEDNEVF
EBV, LF2
A24.02 = 910





FIEKGKFTWY
EBV, LF2
A01.01 = 450





FIYAGDPSI
EBV, LF2
A02.01 = 13





FLSGSPVTS
EBV, LF2
A02.01 = 630





FMRGVWKYEI
EBV, LF2
A02.01 = 140; B08.01 = 340





FTVAFLFFGA
EBV, LF2
A02.01 = 17





FTWYIVPIRL
EBV, LF2
A02.01 = 63





FVMHSDQDIV
EBV, LF2
A02.01 = 130





GPLNVKVGM
EBV, LF2
B07.02 = 610





GQSMAQAFFI
EBV, LF2
A02.01 = 510





HSDQDIVLSV
EBV, LF2
A01.01 = 860





HSSNPVFYV
EBV, LF2
A02.01 = 800





HSSNPVFYVY
EBV, LF2
A01.01 = 170





HTFPGKVCPV
EBV, LF2
A02.01 = 200





IAFRYNPYPK
EBV, LF2
A03.01 = 58





IIPPNFITEL
EBV, LF2
A02.01 = 350





ILPGQSDIQL
EBV, LF2
A02.01 = 350





IPPNFITEL
EBV, LF2
B07.02 = 200





ITELEYNNTY
EBV, LF2
A01.01 = 61





IVVSHSSNPV
EBV, LF2
A02.01 = 160





KFTWYIVPI
EBV, LF2
A24.02 = 200





KLNTSEPQI
EBV, LF2
A02.01 = 270





KLPSFSFNT
EBV, LF2
A02.01 = 63





KLPSFSFNTT
EBV, LF2
A02.01 = 520





KNPYLYLQI
EBV, LF2
A24.02 = 900





KPDTSFMRGV
EBV, LF2
B07.02 = 920





KPGQTLKLTV
EBV, LF2
B07.02 = 320





KSYARIHTF
EBV, LF2
A24.02 = 210





KVCPVNAIRR
EBV, LF2
A03.01 = 850





KYEIFPSYV
EBV, LF2
A24.02 = 740





LAQHTPPFLK
EBV, LF2
A03.01 = 440





LDDVIIAFRY
EBV, LF2
A01.01 = 860





LFIEKGKFTW
EBV, LF2
A24.02 = 230





LLAQHTPPF
EBV, LF2
A02.01 = 540; B07.02 = 750; B08.01 = 190





LLAQHTPPFL
EBV, LF2
A02.01 = 9.6; B08.01 = 480





LLDDVIIAF
EBV, LF2
A02.01 = 120





LPGQSDIQL
EBV, LF2
B07.02 = 470





LPPCPSVGQI
EBV, LF2
B07.02 = 930





LPSFSFNTT
EBV, LF2
B07.02 = 490





LPSFSFNTTT
EBV, LF2
B07.02 = 560





LQLDAQCQEL
EBV, LF2
A02.01 = 220





LTLPGNIVV
EBV, LF2
A02.01 = 880





MAQAFFIYA
EBV, LF2
A02.01 = 450





NPYLYLQIL
EBV, LF2
B07.02 = 170; B08.01 = 150





NSLRRLRPT
EBV, LF2
B08.01 = 370





NTTTYGSRY
EBV, LF2
A01.01 = 210





NTYEAPLSSK
EBV, LF2
A03.01 = 120





PFLKSYARI
EBV, LF2
A24.02 = 390





PSYVRVTNK
EBV, LF2
A03.01 = 620





PYPKSHWRW
EBV, LF2
A24.02 = 100





QIFLSGSPV
EBV, LF2
A02.01 = 380





QLDAQCQEL
EBV, LF2
A02.01 = 370





QSMAQAFFI
EBV, LF2
A02.01 = 890





QSMAQAFFIY
EBV, LF2
A01.01 = 190





RLVKNPYLY
EBV, LF2
A03.01 = 410





RLVKNPYLYL
EBV, LF2
A02.01 = 120





RPTEKPDTSF
EBV, LF2
B07.02 = 39





RSLFIEKGK
EBV, LF2
A03.01 = 430





RSSFRNSLR
EBV, LF2
A03.01 = 230





RSSFRNSLRR
EBV, LF2
A03.01 = 490





RVSVDTPDLK
EBV, LF2
A03.01 = 600





RYFGSPVII
EBV, LF2
A24.02 = 25





RYFTVAFLF
EBV, LF2
A24.02 = 3.7





RYFTVAFLFF
EBV, LF2
A24.02 = 5.2





RYIQRQGCAL
EBV, LF2
A24.02 = 450





RYNPYPKSHW
EBV, LF2
A24.02 = 530





SFKLPSFSF
EBV, LF2
A24.02 = 98





SFMRGVWKY
EBV, LF2
A24.02 = 460





SLPTFERINK
EBV, LF2
A03.01 = 420





SLRRLRPTEK
EBV, LF2
A03.01 = 120





SMAQAFFIY
EBV, LF2
A0l.01 = 870; A03.01 = 340





SMAQAFFIYA
EBV, LF2
A02.01 = 18





SPVTSQDECL
EBV, LF2
B07.02 = 540





SQDECLPYL
EBV, LF2
A02.01 = 18





SQDECLPYLL
EBV, LF2
A02.01 = 45





SRRSSFRNSL
EBV, LF2
B07.02 = 620; B08.01 = 150





SSFRNSLRR
EBV, LF2
A03.01 = 350





SSNPVFYVY
EBV, LF2
A01.01 = 110





SVGQILSFK
EBV, LF2
A03.01 = 73





TFERINKTF
EBV, LF2
A24.02 = 540





TIMRRYIQR
EBV, LF2
A03.01 = 280





TLLDDVIIA
EBV, LF2
A02.01 = 51





TLLDDVIIAF
EBV, LF2
A02.01 = 650





TPDLKREGPL
EBV, LF2
B07.02 = 100





TSFMRGVWK
EBV, LF2
A03.01 = 83





TSFMRGVWKY
EBV, LF2
A01.01 = 730





TSQDECLPY
EBV, LF2
A01.01 = 74





TTYGSRYFTV
EBV, LF2
A02.01 = 130





TVAFLFFGA
EBV, LF2
A02.01 = 290





TVRNISNNPI
EBV, LF2
B07.02 = 450





TWYIVPIRL
EBV, LF2
A24.02 = 680





TYEAPLSSKI
EBV, LF2
A24.02 = 420





TYGSRYFTV
EBV, LF2
A24.02 = 19





VFLKPFFVM
EBV, LF2
A24.02 = 600





VFYVYPQEW
EBV, LF2
A24.02 = 110





VFYVYPQEWK
EBV, LF2
A03.01 = 650





VLNPRSLFI
EBV, LF2
A02.01 = 110





VLSVLNPRSL
EBV, LF2
A02.01 = 680





VMHSDQDIV
EBV, LF2
A02.01 = 440





VMHSDQDIVL
EBV, LF2
A02.01 = 910





VSHSSNPVFY
EBV, LF2
A01.01 = 330





VTSQDECLPY
EBV, LF2
A01.01 = 26





VVSHSSNPV
EBV, LF2
A02.01 = 690





WYIVPIRLV
EBV, LF2
A24.02 = 700





YARIHTFPG
EBV, LF2
B08.01 = 430





YARIHTFPGK
EBV, LF2
A03.01 = 660





YFTVAFLFF
EBV, LF2
A24.02 = 62





YIQRQGCAL
EBV, LF2
B07.02 = 500; B08.01 = 890





YIVPIRLVK
EBV, LF2
A03.01 = 300





YLLAQHTPPF
EBV, LF2
A02.01 = 69; A24.02 = 550; B08.01 = 310





YLYLQILPG
EBV, LF2
A02.01 = 320





YLYLQILPGQ
EBV, LF2
A02.01 = 800





YPGGAHAAL
EBV, LF2
B07.02 = 7; B08.01 = 700





YPGGAHAALA
EBV, LF2
B07.02 = 410





YVRVTNKQVL
EBV, LF2
B07.02 = 70; B08.01 = 470





AGRYRRILL
EBV, BALF5
B08.01 = 810





AIKCTCNAV
EBV, BALF5
B08.01 = 760





ALAAHTTRSL
EBV, BALF5
A02.01 = 93; B07.02 = 200; B08.01 = 340





ALAFDIECL
EBV, BALF5
A02.01 = 70





ALAPSPDAWA
EBV, BALF5
A02.01 = 280





ALKASTFDR
EBV, BALF5
A03.01 = 1000





ALSPANLQA
EBV, BALF5
A02.01 = 530





ALSPANLQAL
EBV, BALF5
A02.01 = 65





ALSVLQNFTA
EBV, BALF5
A02.01 = 260





APLNPEGQL
EBV, BALF5
B07.02 = 85





APQGLDVEF
EBV, BALF5
B07.02 = 280





APSPDAWAPL
EBV, BALF5
B07.02 = 18





ASLLTSWLAK
EBV, BALF5
A03.01 = 140





AVYGFTGVA
EBV, BALF5
A02.01 = 830





AVYQKFVER
EBV, BALF5
A03.01 = 340





AYKSTQMPHL
EBV, BALF5
A24.02 = 770





CLFDNNSGA
EBV, BALF5
A02.01 = 130





CLFDNNSGAA
EBV, BALF5
A02.01 = 390





CLGEEGFPT
EBV, BALF5
A02.01 = 680





CLGEEGFPTA
EBV, BALF5
A02.01 = 120





CLSIAETVTL
EBV, BALF5
A02.01 = 580





CVNVFGQQAY
EBV, BALF5
A01.01 = 750





DARVKEAASL
EBV, BALF5
B08.01 = 330





DLLNHFVIHV
EBV, BALF5
A02.01 = 290





DLREGRVPM
EBV, BALF5
B07.02 = 690; B08.01 = 600





DMLYAFFQL
EBV, BALF5
A02.01 = 340





DNDFVTFGWY
EBV, BALF5
A01.01 = 820





DRARHIYSI
EBV, BALF5
B08.01 = 350





DVRGPQPPL
EBV, BALF5
B07.02 = 870





EAGRYRRIL
EBV, BALF5
B08.01 = 770





ELSRKLSAYK
EBV, BALF5
A03.01 = 450





EMAEDPAYA
EBV, BALF5
A02.01 = 480





EYLRLIPKCF
EBV, BALF5
A24.02 = 310





FLASLLTSW
EBV, BALF5
A02.01 = 150





FLASLLTSWL
EBV, BALF5
A02.01 = 4.1





FLRANTKVRI
EBV, BALF5
B08.01 = 260





FLRPNKGLL
EBV, BALF5
B08.01 = 250





FLRPNKGLLK
EBV, BALF5
A03.01 = 96





FPTATNEADL
EBV, BALF5
B07.02 = 180





FQESFTQGL
EBV, BALF5
A02.01 = 920





FQLIRDLSV
EBV, BALF5
A02.01 = 210; B08.01 = 390





FQTPGAAGV
EBV, BALF5
A02.01 = 48





FQTPGAAGVV
EBV, BALF5
A02.01 = 640





FSESETLRF
EBV, BALF5
A01.01 = 180





FTQGLPVGFL
EBV, BALF5
A02.01 = 960





FVAPISLEA
EBV, BALF5
A02.01 = 26





FVEALSPANL
EBV, BALF5
A02.01 = 860





FVIHVEVAEI
EBV, BALF5
A02.01 = 64





FVKKHVHESF
EBV, BALF5
B08.01 = 130





FVLDNDFVT
EBV, BALF5
A02.01 = 650





FVLDNDFVTF
EBV, BALF5
A24.02 = 750





FVQTRCRRV
EBV, BALF5
B08.01 = 340





FVQTRCRRVL
EBV, BALF5
B07.02 = 330; B08.01 = 100





FYASAPQGL
EBV, BALF5
A24.02 = 100





GLDVEFAVL
EBV, BALF5
A02.01 = 640





GLFPCLSIA
EBV, BALF5
A02.01 = 33





GLFYNPFLR
EBV, BALF5
A03.01 = 100





GLIPIDMYA
EBV, BALF5
A02.01 = 28





GLIPIDMYAV
EBV, BALF5
A02.01 = 5.9





GLPVGFLPV
EBV, BALF5
A02.01 = 68





GLPVGFLPVI
EBV, BALF5
A02.01 = 330





GMYCVQDSA
EBV, BALF5
A02.01 = 100





GMYCVQDSAL
EBV, BALF5
A02.01 = 64





GPEGRRRLGM
EBV, BALF5
B07.02 = 47





GQQAYFYASA
EBV, BALF5
A02.01 = 800





GVANGLFPCL
EBV, BALF5
A02.01 = 340





GVYHFVKKH
EBV, BALF5
A03.01 = 990





GYNVANFDW
EBV, BALF5
A24.02 = 270





HIYSINPASL
EBV, BALF5
A02.01 = 360





HLAVYQKFV
EBV, BALF5
A02.01 = 140





HPNSVCHVA
EBV, BALF5
B07.02 = 530





HPNSVCHVAT
EBV, BALF5
B07.02 = 380





HVATWLQDK
EBV, BALF5
A03.01 = 380





HVHESFLASL
EBV, BALF5
A02.01 = 670; B07.02 = 260; B08.01 = 990





HVYDILETV
EBV, BALF5
A02.01 = 12





HVYDILETVY
EBV, BALF5
A03.01 = 990





HYKEIPRLF
EBV, BALF5
A24.02 = 71





IAHIPCRRVL
EBV, BALF5
B07.02 = 620





IIQAHNLCY
EBV, BALF5
A0l.01 = 600; A03.01 = 790





ILDKQQLAI
EBV, BALF5
A02.01 = 180





ILDKQQLAIK
EBV, BALF5
A03.01 = 310





ILDRARHIY
EBV, BALF5
A0l.01 = 160





ILETVYTHEK
EBV, BALF5
A03.01 = 290





ILNQAYTDL
EBV, BALF5
A02.01 = 750





ILQISCVLW
EBV, BALF5
A24.02 = 990





IPRLFAAGPE
EBV, BALF5
B07.02 = 640





IPRLQHRDSY
EBV, BALF5
B07.02 = 360





IPSDKQGYV
EBV, BALF5
B07.02 = 610





IPSDKQGYVV
EBV, BALF5
B07.02 = 150





IQYVFVEPK
EBV, BALF5
A03.01 = 78





ITGLIPIDMY
EBV, BALF5
A0l.01 = 340





ITKKRYVGV
EBV, BALF5
B08.01 = 520





ITKKRYVGVL
EBV, BALF5
B08.01 = 240





ITLSHPNSV
EBV, BALF5
A02.01 = 610





IVIKLLGRRK
EBV, BALF5
A03.01 = 350





IYGDTDSLF
EBV, BALF5
A24.02 = 37





IYGDTDSLFI
EBV, BALF5
A24.02 = 95





IYSINPASL
EBV, BALF5
A24.02 = 110





KAIKKLLAA
EBV, BALF5
B08.01 = 790





KEYLRLIPK
EBV, BALF5
A03.01 = 810





KGFLRANTK
EBV, BALF5
A03.01 = 170





KIAHIPCRR
EBV, BALF5
A03.01 = 340





KITLSHPNSV
EBV, BALF5
A02.01 = 920





KLDTVARHL
EBV, BALF5
A02.01 = 900





KLDTVARHLL
EBV, BALF5
A02.01 = 170





KLLQGAANI
EBV, BALF5
A02.01 = 25





KLLQGAANIL
EBV, BALF5
A02.01 = 63





KLSAYKSTQM
EBV, BALF5
A02.01 = 620





KLSLSDYKL
EBV, BALF5
A02.01 = 160





KPDKEYLRL
EBV, BALF5
B07.02 = 810





KTACKFVQTR
EBV, BALF5
A03.01 = 740





KTEMAEDPAY
EBV, BALF5
A01.01 = 200





KTFSCLMLI
EBV, BALF5
A02.01 = 46; A24.02 = 400





KTLMKGVELV
EBV, BALF5
A02.01 = 730





KVRITGLIPI
EBV, BALF5
B07.02 = 260





KVTRRSIMGY
EBV, BALF5
A03.01 = 140





LAAHTTRSL
EBV, BALF5
B07.02 = 76





LAKRKAIKKL
EBV, BALF5
B08.01 = 180





LDRARHIYSI
EBV, BALF5
B08.01 = 410





LILQISCVL
EBV, BALF5
A02.01 = 810





LIPIDMYAV
EBV, BALF5
A02.01 = 150





LITKKRYVGV
EBV, BALF5
B08.01 = 860





LLNHFVIHV
EBV, BALF5
A02.01 = 7.3





LLQGAANIL
EBV, BALF5
A02.01 = 360





LLTSWLAKRK
EBV, BALF5
A03.01 = 240





LMKGVELVRK
EBV, BALF5
A03.01 = 130





LMLITKKRYV
EBV, BALF5
A02.01 = 910





LPVGFLPVI
EBV, BALF5
B07.02 = 530





LQISCVLWST
EBV, BALF5
A02.01 = 220





LSRKLSAYK
EBV, BALF5
A03.01 = 94





LTDGKTLMK
EBV, BALF5
A0l.01 = 410; A03.01 = 610





LTGGVYHFV
EBV, BALF5
A02.01 = 200





LTGGVYHFVK
EBV, BALF5
A03.01 = 330





LTSWLAKRK
EBV, BALF5
A03.01 = 560





LVMDLLNHFV
EBV, BALF5
A02.01 = 5.3





LVVDFASLY
EBV, BALF5
A0l.01 = 460





MLERAKAFV
EBV, BALF5
A02.01 = 400





MLITKKRYV
EBV, BALF5
A02.01 = 190; B08.01 = 130





MLITKKRYVG
EBV, BALF5
B08.01 = 560





MLYAFFQLI
EBV, BALF5
A02.01 = 6.6; A24.02 = 800; B08.01 = 640





MLYAFFQLIR
EBV, BALF5
A03.01 = 40





MPHLAVYQKF
EBV, BALF5
B07.02 = 150





MSGGLFYNPF
EBV, BALF5
A24.02 = 490





MYAVCRDKL
EBV, BALF5
A24.02 = 210





NPEGQLRVI
EBV, BALF5
B07.02 = 650





NPFLRPNKGL
EBV, BALF5
B07.02 = 140; B08.01 = 630





NTKVRITGL
EBV, BALF5
B08.01 = 120





NTKVRITGLI
EBV, BALF5
B08.01 = 940





NVANFDWPY
EBV, BALF5
A0l.01 = 300





NVANFDWPYI
EBV, BALF5
A02.01 = 320





PLSGFYNSPV
EBV, BALF5
A02.01 = 520





QIHDRIQYV
EBV, BALF5
A02.01 = 180





QIRVFSCLL
EBV, BALF5
B08.01 = 960





QLIRDLSVEI
EBV, BALF5
A02.01 = 94





QMPHLAVYQK
EBV, BALF5
A03.01 = 900





QQIRVFSCLL
EBV, BALF5
A02.01 = 980





QTRCRRVLDL
EBV, BALF5
B08.01 = 200





RAKAFVEAL
EBV, BALF5
B07.02 = 270; B08.01 = 990





RIFEANVDA
EBV, BALF5
A02.01 = 870





RIQYVFVEPK
EBV, BALF5
A03.01 = 85





RLFAAGPEGR
EBV, BALF5
A03.01 = 260





RLIPKCFQT
EBV, BALF5
A02.01 = 130





RLTGGVYHF
EBV, BALF5
A24.02 = 340





RLTGGVYHFV
EBV, BALF5
A02.01 = 8.1





RPGEDYESF
EBV, BALF5
B07.02 = 630





RPHDAGKGF
EBV, BALF5
B07.02 = 21





RPHDAGKGFL
EBV, BALF5
B07.02 = 9.1





RTMLERAKAF
EBV, BALF5
B07.02 = 920





RTPCRVSVEK
EBV, BALF5
A03.01 = 430





RVFSCLLAA
EBV, BALF5
A02.01 = 69; A03.01 = 810





RVFSCLLAAA
EBV, BALF5
A02.01 = 58





RVIYGDTDSL
EBV, BALF5
B07.02 = 560





RVKEAASLL
EBV, BALF5
B07.02 = 380





RVLDLVLADA
EBV, BALF5
A02.01 = 670





RYRRILLTL
EBV, BALF5
A24.02 = 23





SFLASLLTSW
EBV, BALF5
A24.02 = 56





SFTQGLPVGF
EBV, BALF5
A24.02 = 350





SIMGYGNHA
EBV, BALF5
A02.01 = 760





SINPASLGK
EBV, BALF5
A03.01 = 63





SLFVAPISL
EBV, BALF5
A02.01 = 30





SLLTSWLAK
EBV, BALF5
A03.01 = 23





SLLTSWLAKR
EBV, BALF5
A03.01 = 490





SLSDYKLDTV
EBV, BALF5
A02.01 = 22





SLYPSIIQA
EBV, BALF5
A02.01 = 23





SLYPSIIQAH
EBV, BALF5
A03.01 = 240





SPANLQALA
EBV, BALF5
B07.02 = 500





SPLDFHVYDI
EBV, BALF5
B07.02 = 570





STFDRRTPCR
EBV, BALF5
A03.01 = 600





STGEEAGRY
EBV, BALF5
A01.01 = 130





STQMPHLAVY
EBV, BALF5
A01.01 = 130





SWLAKRKAI
EBV, BALF5
B08.01 = 340





SWPSYQALAF
EBV, BALF5
A24.02 = 49





SYQALAFDI
EBV, BALF5
A24.02 = 13





TKKRYVGVL
EBV, BALF5
B08.01 = 460





TLMKGVELV
EBV, BALF5
A02.01 = 7.9





TMLERAKAF
EBV, BALF5
B08.01 = 100





TMLERAKAFV
EBV, BALF5
A02.01 = 33; B08.01 = 840





TVARHLLGAK
EBV, BALF5
A03.01 = 90





VARHLLGAK
EBV, BALF5
A03.01 = 600





VARHLLGAKK
EBV, BALF5
A03.01 = 820





VIDILNQAY
EBV, BALF5
A01.01 = 34





VIKLLGRRK
EBV, BALF5
A03.01 = 720





VIQPLSGFY
EBV, BALF5
A0l.01 = 990; A03.01 = 920





VIYGDTDSL
EBV, BALF5
A02.01 = 680





VLADARVKEA
EBV, BALF5
A02.01 = 450





VLDDGQQIRV
EBV, BALF5
A02.01 = 17





VLDLVLADA
EBV, BALF5
A02.01 = 540





VLTDGKTLMK
EBV, BALF5
A03.01 = 200





VLVVDFASL
EBV, BALF5
A02.01 = 190





VLWSTGEEA
EBV, BALF5
A02.01 = 210





VMDLLNHFV
EBV, BALF5
A01.01 = 780; A02.01 = 9.7





VMDLLNHFVI
EBV, BALF5
A02.01 = 79





VTFGWYSCR
EBV, BALF5
A03.01 = 290





VTFGWYSCRR
EBV, BALF5
A03.01 = 230





VTRRSIMGY
EBV, BALF5
A03.01 = 250





VYTHEKCAVI
EBV, BALF5
A24.02 = 490





WLAKRKAIK
EBV, BALF5
A03.01 = 470





WLAKRKAIKK
EBV, BALF5
A03.01 = 190





WLQDKHGCRI
EBV, BALF5
A02.01 = 790; B08.01 = 810





WPSYQALAF
EBV, BALF5
B07.02 = 31





WSTGEEAGRY
EBV, BALF5
A0l.01 = 370





YAERHGVPV
EBV, BALF5
B08.01 = 700





YAVCRDKLSL
EBV, BALF5
B08.01 = 330





YFYASAPQGL
EBV, BALF5
A24.02 = 650





YILDRARHI
EBV, BALF5
A02.01 = 740; B08.01 = 770





YLRLIPKCF
EBV, BALF5
B08.01 = 330





YPSIIQAHNL
EBV, BALF5
B07.02 = 76





YQGATVIQPL
EBV, BALF5
A02.01 = 62





YSINPASLGK
EBV, BALF5
A03.01 = 230





YTDLREGRV
EBV, BALF5
A0l.01 = 510





YVFVEPKGGV
EBV, BALF5
A02.01 = 650





AYSARPPPL
EBV, RPMS1
A24.02 = 550





CAYSARPPPL
EBV, RPMS1
B07.02 = 420; B08.01 = 770





GARRRARCPA
EBV, RPMS1
B08.01 = 240





GPGRRARSCL
EBV, RPMS1
B07.02 = 52; B08.01 = 650





MAGARRRARC
EBV, RPMS1
B08.01 = 460





RPGPGRRARS
EBV, RPMS1
B07.02 = 520





RPPHSRTRA
EBV, RPMS1
B07.02 = 93





RRRARCPASA
EBV, RPMS1
B08.01 = 610





SGQPRWWPW
EBV, RPMS1
A24.02 = 350





STRGRRISA
EBV, RPMS1
B07.02 = 990; B08.01 = 240





WPWGSPPPL
EBV, RPMS1
B07.02 = 7.8





WWPWGSPPPL
EBV, RPMS1
A24.02 = 650





AMFCAWKLY
EBV, A73
A03.01 = 120





AMFCAWKLYS
EBV, A73
A02.01 = 700





AVKAAAQSAF
EBV, A73
B07.02 = 750





CAWKLYSRK
EBV, A73
A03.01 = 350





FADAWRMARY
EBV, A73
A01.01 = 11





KLYSRKMPS
EBV, A73
A03.01 = 400





KLYSRKMPSR
EBV, A73
A03.01 = 19





KPPTVLTRPA
EBV, A73
B07.02 = 540





KTLEARCSSR
EBV, A73
A03.01 = 770





MARYWGCAV
EBV, A73
B07.02 = 37; B08.01 = 51





MARYWGCAVK
EBV, A73
A03.01 = 420





MPSRSKTLEA
EBV, A73
B07.02 = 92; B08.01 = 910





MSMPPKGFLK
EBV, A73
A03.01 = 29





PPTVLTRPAM
EBV, A73
B07.02 = 350





RGWRARRLGK
EBV, A73
A03.01 = 290





RKMPSRSKTL
EBV, A73
B07.02 = 580





RLGKESWFA
EBV, A73
A02.01 = 56





RLLNKPPTV
EBV, A73
A02.01 = 21





RLLNKPPTVL
EBV, A73
A02.01 = 290





RMARYWGCAV
EBV, A73
A02.01 = 41; B08.01 = 740





RPAMFCAWKL
EBV, A73
B07.02 = 40





SMPPKGFLK
EBV, A73
A03.01 = 200





SMPPKGFLKK
EBV, A73
A03.01 = 230





SPACQTRDT
EBV, A73
B07.02 = 440





SWFADAWRM
EBV, A73
A24.02 = 530





VLTRPAMFCA
EBV, A73
A02.01 = 650





WRMARYWGC
EBV, A73
B08.01 = 880





WRMARYWGCA
EBV, A73
B08.01 = 190





YSRKMPSRSK
EBV, A73
A03.01 = 680





AARDRFPGL
EBV, BALF4
B07.02 = 720; B08.01 = 270





AARGSTSAA
EBV, BALF4
B07.02 = 110





AARGSTSAAV
EBV, BALF4
B07.02 = 61





AFLDKGTYTL
EBV, BALF4
A24.02 = 960





ALHEQNQEQK
EBV, BALF4
A03.01 = 730





APGKSLGTL
EBV, BALF4
B07.02 = 16





APPAARGST
EBV, BALF4
B07.02 = 170





AQNIAGLRK
EBV, BALF4
A03.01 = 570





ATLQTFISL
EBV, BALF4
A02.01 = 980





ATRQQTSFPF
EBV, BALF4
B07.02 = 720





ATVQIQFAY
EBV, BALF4
A0l.01 = 170





CLEQKRQNM
EBV, BALF4
B08.01 = 650





CLITDMMAK
EBV, BALF4
A03.01 = 74





CPLQHWQTF
EBV, BALF4
B07.02 = 88; B08.01 = 630





CQATSQYYF
EBV, BALF4
A24.02 = 620





CYSRPLVSF
EBV, BALF4
A24.02 = 12





DMMAKSNSPF
EBV, BALF4
B08.01 = 190





DSFHVRTNYK
EBV, BALF4
A03.01 = 850





ELMDSLGSV
EBV, BALF4
A02.01 = 21





ELYDAPGWLI
EBV, BALF4
A02.01 = 400





ENRTAYCPL
EBV, BALF4
B08.01 = 680





EQKRQNMVL
EBV, BALF4
B08.01 = 470





ETDQMDTIY
EBV, BALF4
A01.01 = 13





ETMCYSRPL
EBV, BALF4
B08.01 = 160





FFKNPFGGML
EBV, BALF4
B08.01 = 980





FISLNTSLI
EBV, BALF4
A02.01 = 140





FITSGGLLL
EBV, BALF4
A02.01 = 350





FITSGGLLLA
EBV, BALF4
A02.01 = 330





FLDKGTYTL
EBV, BALF4
A02.01 = 3





FLDKGTYTLS
EBV, BALF4
A02.01 = 200





FLTKKMTEV
EBV, BALF4
A02.01 = 10; B08.01 = 980





FPGLRRRRY
EBV, BALF4
B07.02 = 850





FQAQNIAGL
EBV, BALF4
A02.01 = 25





FQSGNEIHV
EBV, BALF4
A02.01 = 79





FSFINDTKTY
EBV, BALF4
A01.01 = 740





FVDGLGELM
EBV, BALF4
A01.01 = 760





FVTNTTVGI
EBV, BALF4
A02.01 = 340





FYDGKNKETF
EBV, BALF4
A24.02 = 190





GFISFFKNPF
EBV, BALF4
A24.02 = 160





GIATLQTFI
EBV, BALF4
A02.01 = 490





GIFREYNFQA
EBV, BALF4
A02.01 = 310





GLFSSLVSG
EBV, BALF4
A02.01 = 870





GLFSSLVSGF
EBV, BALF4
A02.01 = 850





GLGELMDSL
EBV, BALF4
A02.01 = 73





GLLLAWLPL
EBV, BALF4
A02.01 = 85





GLLLAWLPLT
EBV, BALF4
A02.01 = 280





GLLMVFKDNI
EBV, BALF4
A02.01 = 610





GLRKDLDNAV
EBV, BALF4
A02.01 = 680





GMLILVLVA
EBV, BALF4
A02.01 = 570





GPSVASRAL
EBV, BALF4
B07.02 = 7.9





GQEAITYFI
EBV, BALF4
A02.01 = 430





GTDNEIFLTK
EBV, BALF4
A03.01 = 690





GTLNNPATV
EBV, BALF4
A02.01 = 940





GTYTLSWKL
EBV, BALF4
A02.01 = 410





GVNITVNLK
EBV, BALF4
A03.01 = 120





HTEGLLMVFK
EBV, BALF4
A03.01 = 450





HVYNDYHHFK
EBV, BALF4
A03.01 = 22





HWQTFDSTI
EBV, BALF4
A24.02 = 490





ILIYNGWYA
EBV, BALF4
A02.01 = 15





IQFAYDSLR
EBV, BALF4
A03.01 = 920





IQFAYDSLRR
EBV, BALF4
A03.01 = 610





IYNGWYADSV
EBV, BALF4
A24.02 = 290





IYQCYNAVKM
EBV, BALF4
A24.02 = 140





KGTYTLSWK
EBV, BALF4
A03.01 = 570





KIVTNILIY
EBV, BALF4
A03.01 = 460





KMTEVCQAT
EBV, BALF4
A02.01 = 400





KMTKDGLTRV
EBV, BALF4
A02.01 = 96





KPTGGLANGV
EBV, BALF4
B07.02 = 580





KSNSPFDFFV
EBV, BALF4
A02.01 = 570





KTMHEKYEAV
EBV, BALF4
A02.01 = 250





LARAWCLEQK
EBV, BALF4
A03.01 = 920





LFSSLVSGF
EBV, BALF4
A24.02 = 490





LILVLVAGV
EBV, BALF4
A02.01 = 58





LILVLVAGVV
EBV, BALF4
A02.01 = 940





LLAALACRL
EBV, BALF4
A02.01 = 37





LLAWLPLTPR
EBV, BALF4
A03.01 = 440





LLLAWLPLT
EBV, BALF4
A02.01 = 21





LLMVFKDNI
EBV, BALF4
A02.01 = 130





LLMVFKDNII
EBV, BALF4
A02.01 = 320; B08.01 = 700





LPLTPRSLA
EBV, BALF4
B07.02 = 250





LPLTPRSLAT
EBV, BALF4
B07.02 = 110





LRRQINRML
EBV, BALF4
B08.01 = 770





LTPRSLATV
EBV, BALF4
A02.01 = 900





LVAGVVILV
EBV, BALF4
A02.01 = 46





LVSGFISFFK
EBV, BALF4
A03.01 = 27





LYDAPGWLI
EBV, BALF4
A24.02 = 110





LYDAPGWLIW
EBV, BALF4
A24.02 = 170





MCYSRPLVSF
EBV, BALF4
B08.01 = 760





MLILVLVAGV
EBV, BALF4
A02.01 = 7.6





MLYPGIDEL
EBV, BALF4
A02.01 = 4.2





MLYPGIDELA
EBV, BALF4
A02.01 = 16





MMAKSNSPF
EBV, BALF4
A02.01 = 920; A24.02 = 240; B07.02 = 290; B08.01 = 630





MSQQPVQMLY
EBV, BALF4
A01.01 = 67





MTRRRVLSV
EBV, BALF4
B07.02 = 200; B08.01 = 7.4





MTRRRVLSVV
EBV, BALF4
B07.02 = 490; B08.01 = 39





MVFKDNIIPY
EBV, BALF4
A03.01 = 360





MVLRELTKI
EBV, BALF4
A02.01 = 580





NIDFASLELY
EBV, BALF4
A01.01 = 24





NIIPYSFKV
EBV, BALF4
A02.01 = ll





NILIYNGWYA
EBV, BALF4
A02.01 = 470





NLTELTTPT
EBV, BALF4
A02.01 = 1000





NMVLRELTK
EBV, BALF4
A03.01 = 840





NPATVQIQF
EBV, BALF4
B07.02 = 810





NPFGGMLIL
EBV, BALF4
B07.02 = 110





NPQGERRAF
EBV, BALF4
B07.02 = 48





NPQGERRAFL
EBV, BALF4
B07.02 = 73





NPTTVMSSI
EBV, BALF4
B07.02 = 240





PPAAPGKSL
EBV, BALF4
B07.02 = 84





PPAARGSTSA
EBV, BALF4
B07.02 = 800





QMDTIYQCY
EBV, BALF4
A01.01 = 75





QMLYPGIDEL
EBV, BALF4
A02.01 = 210





QPAPPATTV
EBV, BALF4
B07.02 = 95





QQTSFPFRV
EBV, BALF4
A02.01 = 400





QTVEMSPFY
EBV, BALF4
A01.01 = 320





QVNKTMHEK
EBV, BALF4
A03.01 = 520





QYYFQSGNEI
EBV, BALF4
A24.02 = 320





RMLGDLARA
EBV, BALF4
A02.01 = 47





RMLGDLARAW
EBV, BALF4
A24.02 = 1000





RPLVSFSFI
EBV, BALF4
B07.02 = 87





RQQTSFPFR
EBV, BALF4
A03.01 = 790





RQQTSFPFRV
EBV, BALF4
A02.01 = 250





RTAYCPLQH
EBV, BALF4
A03.01 = 420





RTNYKIVDY
EBV, BALF4
A03.01 = 890





RTRQMSQQPV
EBV, BALF4
B07.02 = 160





RTRTTVNCL
EBV, BALF4
B07.02 = 220





RVLSVVVLL
EBV, BALF4
A02.01 = 150





RVLSVVVLLA
EBV, BALF4
A02.01 = 500





RYASQTELY
EBV, BALF4
A24.02 = 870





RYTKGQEAI
EBV, BALF4
A24.02 = 350





SFHVRTNYK
EBV, BALF4
A03.01 = 1000





SFHVRTNYKI
EBV, BALF4
A24.02 = 580





SFKVRSYTKI
EBV, BALF4
B08.01 = 410





SFPFRVCEL
EBV, BALF4
A24.02 = 670; B08.01 = 970





SITNLVSTV
EBV, BALF4
A02.01 = 370





SIYGKAVAA
EBV, BALF4
A02.01 = 490





SIYGKAVAAK
EBV, BALF4
A03.01 = 17





SLGSVGQSI
EBV, BALF4
A02.01 = 790





SLIENIDFA
EBV, BALF4
A02.01 = 7.7





SLIENIDFAS
EBV, BALF4
A02.01 = 320





SLNTSLIENI
EBV, BALF4
A02.01 = 76





SLRRQINRM
EBV, BALF4
B08.01 = 77





SLRRQINRML
EBV, BALF4
B07.02 = 560; B08.01 = 80





SLTRRTRQM
EBV, BALF4
B08.01 = 55





SLVSGFISF
EBV, BALF4
A02.01 = 990





SLVSGFISFF
EBV, BALF4
A02.01 = 770





SMRVPGSETM
EBV, BALF4
B07.02 = 270; B08.01 = 780





SPPSSPSPPA
EBV, BALF4
B07.02 = 260





SPSPPAPPA
EBV, BALF4
B07.02 = 55





SPSPPAPPAA
EBV, BALF4
B07.02 = 58





STIATETGK
EBV, BALF4
A03.01 = 320





STVGGLFSSL
EBV, BALF4
A02.01 = 490





SVGQSITNLV
EBV, BALF4
A02.01 = 810





SVVVLLAAL
EBV, BALF4
A02.01 = 980





SYTKIVTNI
EBV, BALF4
A24.02 = 62





SYTKIVTNIL
EBV, BALF4
A24.02 = 120





TFHERADSF
EBV, BALF4
A24.02 = 860





TFISLNTSLI
EBV, BALF4
A24.02 = 200





TIYQCYNAV
EBV, BALF4
A02.01 = 160





TIYQCYNAVK
EBV, BALF4
A03.01 = 43





TMCYSRPLV
EBV, BALF4
A02.01 = 630





TMHEKYEAV
EBV, BALF4
A02.01 = 61; B08.01 = 300





TTVMSSIYGK
EBV, BALF4
A03.01 = 220





TVGGLFSSLV
EBV, BALF4
A02.01 = 690





TVMSSIYGK
EBV, BALF4
A03.01 = 44





TYFITSGGLL
EBV, BALF4
A24.02 = 480





VILVISLTR
EBV, BALF4
A03.01 = 750





VILVISLTRR
EBV, BALF4
A03.01 = 850





VISVSQCVPV
EBV, BALF4
A02.01 = 510





VLLAALACR
EBV, BALF4
A03.01 = 980





VLLAALACRL
EBV, BALF4
A02.01 = 31





VLRRRRRDA
EBV, BALF4
B08.01 = 190





VLRRRRRDAG
EBV, BALF4
B08.01 = 490





VLSVVVLLA
EBV, BALF4
A02.01 = 340





VLSVVVLLAA
EBV, BALF4
A02.01 = 250





VLVAGVVIL
EBV, BALF4
A02.01 = 150





VLVAGVVILV
EBV, BALF4
A02.01 = 21





VMSSIYGKA
EBV, BALF4
A02.01 = 910





VMSSIYGKAV
EBV, BALF4
A02.01 = 140





VPPAAPGKSL
EBV, BALF4
B07.02 = 82





VPVNQATVTL
EBV, BALF4
B07.02 = 42





VSFSFINDTK
EBV, BALF4
A03.01 = 220





VSGFISFFK
EBV, BALF4
A03.01 = 65





VTDEGTSSF
EBV, BALF4
A01.01 = 110





VTDEGTSSFV
EBV, BALF4
A01.01 = 220; A02.01 = 150





VYVDRDGVNI
EBV, BALF4
A24.02 = 690





YADSVTNRH
EBV, BALF4
A01.01 = 950





YCPLQHWQTF
EBV, BALF4
A24.02 = 690





YFITSGGLLL
EBV, BALF4
A24.02 = 630





YSFKVRSYTK
EBV, BALF4
A03.01 = 52





YSRPLVSFSF
EBV, BALF4
A24.02 = 900





YTKGQEAITY
EBV, BALF4
A01.01 = 940





YYFQSGNEI
EBV, BALF4
A24.02 = 38





AAAYSQVYAL
EBV, BALF3
B07.02 = 440





AAFENSKYIK
EBV, BALF3
A03.01 = 430





AAFTAHPQYR
EBV, BALF3
A03.01 = 790





AAYSQVYAL
EBV, BALF3
A02.01 = 610; B07.02 = 490; B08.01 = 980





AGARLTHLF
EBV, BALF3
A24.02 = 640





AILLPRLRR
EBV, BALF3
A03.01 = 490





AILQKRNEL
EBV, BALF3
B08.01 = 250





ALAGARLTH
EBV, BALF3
A03.01 = 800





ALAGARLTHL
EBV, BALF3
A02.01 = 120





ALARCCDAA
EBV, BALF3
A02.01 = 570





ALAVELSVCA
EBV, BALF3
A02.01 = 170





ALDTYNVFST
EBV, BALF3
A02.01 = 150





ALFARERRL
EBV, BALF3
B08.01 = 800





ALKRKEQYL
EBV, BALF3
B08.01 = 100





ALLYLHLQM
EBV, BALF3
A02.01 = 540





ALPPDGLYL
EBV, BALF3
A02.01 = 330





ALPPDGLYLT
EBV, BALF3
A02.01 = 940





ALREAVLTV
EBV, BALF3
A02.01 = 40





ALSDALKRK
EBV, BALF3
A03.01 = 320





ALSLELVHL
EBV, BALF3
A02.01 = 130





ALSLELVHLL
EBV, BALF3
A02.01 = 50





ALTHSLYTF
EBV, BALF3
A24.02 = 540





ALTHSLYTFI
EBV, BALF3
A02.01 = 35





APGATERLF
EBV, BALF3
B07.02 = 990





APGRKGTRV
EBV, BALF3
B07.02 = 210





AVLTVSLYNK
EBV, BALF3
A03.01 = 60





AYSQVYALA
EBV, BALF3
A24.02 = 540





AYSQVYALAV
EBV, BALF3
A24.02 = 330





CARLDPRSL
EBV, BALF3
B07.02 = 530; B08.01 = 940





CLAQQSETL
EBV, BALF3
A02.01 = 570





CLINDIEIL
EBV, BALF3
A02.01 = 220





CLINDIEILM
EBV, BALF3
A02.01 = 540





DLLGRFRGV
EBV, BALF3
B08.01 = 620





DLYALLYLHL
EBV, BALF3
A02.01 = 620





DPRLYITSV
EBV, BALF3
B07.02 = 430; B08.01 = 220





DPRSLDVAAV
EBV, BALF3
B07.02 = 580





DSDRPLILLY
EBV, BALF3
A01.01 = 10





DWIETSFNSF
EBV, BALF3
A24.02 = 810





EILMKRINSV
EBV, BALF3
B08.01 = 120





ELARMRNHFL
EBV, BALF3
B08.01 = 350





ELYTRLNRA
EBV, BALF3
B08.01 = 550





ELYTRLNRAM
EBV, BALF3
B08.01 = 130





EVAELSELLY
EBV, BALF3
A01.01 = 220





FARERRLAL
EBV, BALF3
B07.02 = 8.5; B08.01 = 3.6





FARERRLALV
EBV, BALF3
B07.02 = 910; B08.01 = 20





FKDLYALLY
EBV, BALF3
A01.01 = 480





FLTSLIWPG
EBV, BALF3
A02.01 = 30





FLTSLIWPGI
EBV, BALF3
A02.01 = 6.6





FLYRRAFVS
EBV, BALF3
A02.01 = 230; B08.01 = 600





FLYRRAFVSG
EBV, BALF3
A02.01 = 610; B08.01 = 380





FPPPSNVAL
EBV, BALF3
B07.02 = 44





FYCMSHTMGL
EBV, BALF3
A24.02 = 660





GGHSRLSAL
EBV, BALF3
B08.01 = 800





GLAKLQSCL
EBV, BALF3
A02.01 = 350





GLAKLQSCLA
EBV, BALF3
A02.01 = 390





GLFENEVRQA
EBV, BALF3
A02.01 = 140





GLFPPPSNV
EBV, BALF3
A02.01 = 25





GLFPPPSNVA
EBV, BALF3
A02.01 = 430





GLLAAAYSQV
EBV, BALF3
A02.01 = 10





GLLAELEAI
EBV, BALF3
A02.01 = 25





GLLAELEAIL
EBV, BALF3
A02.01 = 100





GPSGALSDAL
EBV, BALF3
B07.02 = 47





HALTHSLYTF
EBV, BALF3
A24.02 = 630





HFLYRRAFV
EBV, BALF3
B08.01 = 350





HLFDAVAPG
EBV, BALF3
A02.01 = 420





HLFDAVAPGA
EBV, BALF3
A02.01 = 12





HLHALTHSL
EBV, BALF3
A02.01 = 78; B07.02 = 450; B08.01 = 160





HLHALTHSLY
EBV, BALF3
A03.01 = 120





HLLEAEEKA
EBV, BALF3
A02.01 = 810





HLLENSREA
EBV, BALF3
A02.01 = 350





HLQMRDDSA
EBV, BALF3
B08.01 = 870





HQKAFLTSL
EBV, BALF3
B08.01 = 450





HVSCNHLAV
EBV, BALF3
A02.01 = 960; B07.02 = 950





ILCRALREAV
EBV, BALF3
A02.01 = 390





ILLYKGRGWV
EBV, BALF3
A02.01 = 930





ILMKRINSV
EBV, BALF3
A02.01 = 6.7; B08.01 = 9.4





ILMKRINSVF
EBV, BALF3
A24.02 = 660; B08.01 = 75





IWPGIEPSDW
EBV, BALF3
A24.02 = 350





KTWGRSLIL
EBV, BALF3
A02.01 = 650





KTWGRSLILR
EBV, BALF3
A03.01 = 120





KVDVSLCLI
EBV, BALF3
A02.01 = 900





LAAAYSQVY
EBV, BALF3
A01.01 = 800





LAGARLTHL
EBV, BALF3
B08.01 = 320





LARMRNHFL
EBV, BALF3
B07.02 = 350; B08.01 = 22





LFARERRLAL
EBV, BALF3
B08.01 = 280





LGRFRGVSPI
EBV, BALF3
B08.01 = 300





LILRRADAV
EBV, BALF3
B08.01 = 350





LINDIEILM
EBV, BALF3
A02.01 = 880





LINDIEILMK
EBV, BALF3
A03.01 = 190





LLAAAYSQV
EBV, BALF3
A02.01 = 12





LLAAAYSQVY
EBV, BALF3
A03.01 = 850





LLAELEAIL
EBV, BALF3
A02.01 = 28





LLAELEAILL
EBV, BALF3
A02.01 = 17





LLYKGRGWV
EBV, BALF3
A02.01 = 460





LLYKGRGWVF
EBV, BALF3
B08.01 = 720





LLYLHLQMR
EBV, BALF3
A03.01 = 410





LLYWNSGGH
EBV, BALF3
A03.01 = 610





LLYWNSGGHA
EBV, BALF3
A02.01 = 750





LMKRINSVF
EBV, BALF3
B08.01 = 130





LMKRINSVFY
EBV, BALF3
A03.01 = 450





LPNHGNPST
EBV, BALF3
B07.02 = 400





LPNHGNPSTA
EBV, BALF3
B07.02 = 240





LSLELVHLL
EBV, BALF3
A02.01 = 630





LVRRACEEAL
EBV, BALF3
B07.02 = 370





LYALLYLHL
EBV, BALF3
A24.02 = 64





LYITSVPCW
EBV, BALF3
A24.02 = 60





LYKGRGWVF
EBV, BALF3
A24.02 = 68





LYNKTWGRSL
EBV, BALF3
A24.02 = 630





LYTFITGPL
EBV, BALF3
A24.02 = 350





LYWNSGGHAI
EBV, BALF3
A24.02 = 150





MSGLLAAAY
EBV, BALF3
A01.01 = 91





NSREASAAL
EBV, BALF3
B07.02 = 230





NVFSTVPPEV
EBV, BALF3
A02.01 = 46





PQYRAILQK
EBV, BALF3
A03.01 = 850





QVYALAVEL
EBV, BALF3
A02.01 = 520





RAILQKRNEL
EBV, BALF3
B07.02 = 260





RLDPRSLDV
EBV, BALF3
A02.01 = 500





RLFCGGVYS
EBV, BALF3
A02.01 = 390





RLFCGGVYSS
EBV, BALF3
A02.01 = 150





RLNRAMQRL
EBV, BALF3
A02.01 = 810





RLSALFARER
EBV, BALF3
A03.01 = 400





RLTHLFDAV
EBV, BALF3
A02.01 = 90





RMRNHFLYR
EBV, BALF3
A03.01 = 17





RMRNHFLYRR
EBV, BALF3
A03.01 = 61





RPAGAREPG
EBV, BALF3
B07.02 = 200





RPGGPEEGAV
EBV, BALF3
B07.02 = 180





RQAGLGHLL
EBV, BALF3
A02.01 = 460





RQVATEGLAK
EBV, BALF3
A03.01 = 780





RTPSVAYSV
EBV, BALF3
A02.01 = 430





SAALLAPGRK
EBV, BALF3
A03.01 = 1000





SLASSQQIL
EBV, BALF3
A02.01 = 600





SLCLINDIEI
EBV, BALF3
A02.01 = 690





SLILRRADA
EBV, BALF3
B08.01 = 330





SLILRRADAV
EBV, BALF3
A02.01 = 490; B08.01 = 190





SLYNKTWGR
EBV, BALF3
A03.01 = 170





SLYTFITGPL
EBV, BALF3
A02.01 = 31; B08.01 = 790





SPEVPRPAGA
EBV, BALF3
B07.02 = 630





SPIPDPRLYI
EBV, BALF3
B07.02 = 150





SQVYALAVEL
EBV, BALF3
A02.01 = 340





SVAYSVEFY
EBV, BALF3
A01.01 = 690





SVEFYGGHK
EBV, BALF3
A03.01 = 900





SVFYCMSHTM
EBV, BALF3
A02.01 = 900





TLSSEHLHAL
EBV, BALF3
A02.01 = 99





TLTETLCLRV
EBV, BALF3
A02.01 = 45





TPSVAYSVEF
EBV, BALF3
B07.02 = 180





TSFNSFYSV
EBV, BALF3
A02.01 = 210





TYDSDRPLI
EBV, BALF3
A24.02 = 990





VAELSELLY
EBV, BALF3
A01.01 = 74





VFKDLYALL
EBV, BALF3
A24.02 = 390





VFYCMSHTM
EBV, BALF3
A24.02 = 350





VLTVSLYNK
EBV, BALF3
A03.01 = 88





VPCWRCVGEL
EBV, BALF3
B07.02 = 170





VPGPGRPEA
EBV, BALF3
B07.02 = 260





VPPLRTPSV
EBV, BALF3
B07.02 = 250; B08.01 = 470





VPPLRTPSVA
EBV, BALF3
B07.02 = 850





VPRPAGARE
EBV, BALF3
B07.02 = 720





VPRPAGAREP
EBV, BALF3
B07.02 = 620





VVYWELARMR
EBV, BALF3
A03.01 = 660





VWGDVVYWEL
EBV, BALF3
A24.02 = 97





VYALAVELSV
EBV, BALF3
A24.02 = 340





WIETSFNSFY
EBV, BALF3
A01.01 = 120





WVFKDLYAL
EBV, BALF3
A02.01 = 46





WVFKDLYALL
EBV, BALF3
A02.01 = 130





YALAVELSV
EBV, BALF3
A02.01 = 110





YALLYLHLQM
EBV, BALF3
B08.01 = 690





YCMSHTMGL
EBV, BALF3
A02.01 = 940; B08.01 = 420





YLRQVATEGL
EBV, BALF3
A02.01 = 180; B07.02 = 860; B08.01 = 570





YLTYDSDRPL
EBV, BALF3
A02.01 = 98





YNKTWGRSL
EBV, BALF3
B08.01 = 240





YTFITGPLA
EBV, BALF3
A02.01 = 960





YWNSGGHAI
EBV, BALF3
A24.02 = 770





AARPRLLLSL
EBV, BARF0
B07.02 = 58; B08.01 = 70





AARVPIEEL
EBV, BARF0
B07.02 = 950





AGMSATLPL
EBV, BARF0
B07.02 = 550





ALLRQAGLQL
EBV, BARF0
A02.01 = 450





ALLWAARPR
EBV, BARF0
A03.01 = 590





ALLWAARPRL
EBV, BARF0
A02.01 = 130





ALPGRLLLA
EBV, BARF0
A02.01 = 620





APAGHRGDV
EBV, BARF0
B07.02 = 23





APAGHRGDVE
EBV, BARF0
B07.02 = 840





APGYAVEAV
EBV, BARF0
B07.02 = 130





AVEAVEGGLY
EBV, BARF0
A01.01 = 540





AVEGGLYPV
EBV, BARF0
A02.01 = 63





AVRLQRLHRV
EBV, BARF0
B08.01 = 730





AVRQRVQVL
EBV, BARF0
B07.02 = 37; B08.01 = 47





CPRQRLLAG
EBV, BARF0
B07.02 = 190; B08.01 = 180





CPSRPGHLRA
EBV, BARF0
B07.02 = 680





CTDSMAARV
EBV, BARF0
A01.01 = 220





EPRVELVPL
EBV, BARF0
B07.02 = 64; B08.01 = 410





EPRVELVPLL
EBV, BARF0
B07.02 = 430





FQALQPHGV
EBV, BARF0
A02.01 = 44





GLALLRQAGL
EBV, BARF0
A02.01 = 910





GLGKQVCFDV
EBV, BARF0
A02.01 = 99





GLGQGLALL
EBV, BARF0
A02.01 = 96





GMSATLPLPR
EBV, BARF0
A03.01 = 410





GPLCRGRVA
EBV, BARF0
B07.02 = 150





GPLCRGRVAV
EBV, BARF0
B07.02 = 23; B08.01 = 360





GPPHQGQATL
EBV, BARF0
B07.02 = 180





GPRDEGVQA
EBV, BARF0
B07.02 = 670





GPRDEGVQAV
EBV, BARF0
B07.02 = 35





GVQRSGRPL
EBV, BARF0
B07.02 = 330





GVRREGGGAV
EBV, BARF0
B07.02 = 860





HAIKHAIDSL
EBV, BARF0
B07.02 = 700





HLAQVLLLA
EBV, BARF0
A02.01 = 99





HLAQVLLLAL
EBV, BARF0
A02.01 = 42





HLRALPGRL
EBV, BARF0
B07.02 = 970; B08.01 = 700





HLRALPGRLL
EBV, BARF0
B07.02 = 370; B08.01 = 700





HLRGHCREDV
EBV, BARF0
B08.01 = 680





HQLAHTAPA
EBV, BARF0
A02.01 = 360; B08.01 = 880





ILKGGTLAGA
EBV, BARF0
A02.01 = 860





KQVCFDVLGI
EBV, BARF0
A02.01 = 140





LALLRQAGL
EBV, BARF0
B08.01 = 170





LCRGRVAVV
EBV, BARF0
B08.01 = 400





LLASAQPLH
EBV, BARF0
A03.01 = 950





LLLASAQPL
EBV, BARF0
A02.01 = 20; B08.01 = 790





LLQDGPVLGV
EBV, BARF0
A02.01 = 18





LLRQAGLQL
EBV, BARF0
B07.02 = 110; B08.01 = 280





LLVRQRTCGV
EBV, BARF0
A02.01 = 300





LLWAARPRL
EBV, BARF0
A02.01 = 39





LLWAARPRLL
EBV, BARF0
A02.01 = 73; B08.01 = 860





LPGKQGREA
EBV, BARF0
B07.02 = 280





LPGRLLLASA
EBV, BARF0
B07.02 = 560





LPLPRCTDSM
EBV, BARF0
B07.02 = 40; B08.01 = 600





LPRCTDSMA
EBV, BARF0
B07.02 = 140





LPRCTDSMAA
EBV, BARF0
B07.02 = 44; B08.01 = 860





LQDGPVLGV
EBV, BARF0
A02.01 = 200





LRIHRHRQV
EBV, BARF0
B08.01 = 840





LRIHRHRQVV
EBV, BARF0
B08.01 = 490





LVRQRTCGV
EBV, BARF0
B07.02 = 690; B08.01 = 72





LWAARPRLLL
EBV, BARF0
A24.02 = 770





LYPVARLDAW
EBV, BARF0
A24.02 = 85





MSATLPLPR
EBV, BARF0
A03.01 = 760





PPHQGQATL
EBV, BARF0
B07.02 = 320





PPRARDRAL
EBV, BARF0
B07.02 = 44; B08.01 = 760





PPRARDRALL
EBV, BARF0
B07.02 = 230





QPCPRQRLL
EBV, BARF0
B07.02 = 540





QPHGVRHAI
EBV, BARF0
B07.02 = 22





QTLGGHLAQV
EBV, BARF0
A02.01 = 600





QVLRAQGLGK
EBV, BARF0
A03.01 = 430





RALLWAARPR
EBV, BARF0
A03.01 = 960





RALPGRLLL
EBV, BARF0
B07.02 = 230





RARDRALLWA
EBV, BARF0
B07.02 = 1000





RIHRHRQVV
EBV, BARF0
B07.02 = 340; B08.01 = 130





RLLLASAQPL
EBV, BARF0
A02.01 = 54





RLLLSLQQV
EBV, BARF0
A02.01 = 22





RLRIHRHRQV
EBV, BARF0
B07.02 = 530; B08.01 = 58





RPGHLRALPG
EBV, BARF0
B07.02 = 410





RPLCLRPPRA
EBV, BARF0
B07.02 = 320





RPPRARDRA
EBV, BARF0
B07.02 = 640





RPPRARDRAL
EBV, BARF0
B07.02 = 9.5





RPRLLLSLQQ
EBV, BARF0
B07.02 = 630





RVQVLRAQGL
EBV, BARF0
B07.02 = 700





RVREGAGRA
EBV, BARF0
B07.02 = 990





RVREGAGRAG
EBV, BARF0
B07.02 = 970





RVWDGTYAPK
EBV, BARF0
A03.01 = 52





SLQQVPEPSL
EBV, BARF0
A02.01 = 230





SQGHVAGWGK
EBV, BARF0
A03.01 = 950





TLGGHLAQV
EBV, BARF0
A02.01 = 18





TLGGHLAQVL
EBV, BARF0
A02.01 = 760





TYAPKAAQQI
EBV, BARF0
A24.02 = 61





VLLLALERV
EBV, BARF0
A02.01 = 31





VLRAQGLGK
EBV, BARF0
A03.01 = 33





VPIEELREF
EBV, BARF0
B07.02 = 790





VPLLQDGPV
EBV, BARF0
B07.02 = 170





VPLLQDGPVL
EBV, BARF0
B07.02 = 68





VVAHAGQLPV
EBV, BARF0
A02.01 = 530





WAARPRLLL
EBV, BARF0
B07.02 = 260





WPYQGSQERL
EBV, BARF0
B07.02 = 35





YPVARLDAW
EBV, BARF0
B07.02 = 250









Example 2: HLA Class I and Class II Binding Assays

The following example of peptide binding to HLA molecules demonstrates quantification of binding affinities of HLA class I and class II peptides. Binding assays can be performed with peptides that are either motif-bearing or not motif-bearing.


Epstein-Barr virus (EBV)-transformed homozygous cell lines, fibroblasts, CIR, or 721.22 transfectants are used as sources of HLA class I molecules. Cell lysates are prepared and HLA molecules purified in accordance with disclosed protocols (Sidney et al., Current Protocols in Immunology 18.3.1 (1998); Sidney, et al., J. Immunol. 154:247 (1995); Sette, et al., Mol. Immunol. 31:813 (1994)). HLA molecules are purified from lysates by affinity chromatography. The lysates are passed over a column of Sepharose CL-4B beads coupled to an appropriate antibody. The anti-HLA column is then washed with 10 mM Tris-HCL, pH 8.0, in 1% NP-40, PBS, and PBS containing 0.4% n-octylglucoside and HLA molecules are eluted with 50 mM diethylamine in 0.15M NaCl containing 0.4% n-octylglucoside, pH 11.5. A 1/25 volume of 2.0M Tris, pH 6.8, is added to the eluate to reduce the pH to ˜8.0. Eluates are then concentrated by centrifugation in Centriprep 30 concentrators (Amicon, Beverly, Mass.). Protein content is evaluated by a BCA protein assay (Pierce Chemical Co., Rockford, Ill.) and confirmed by SDS-PAGE.


A detailed description of the protocol utilized to measure the binding of peptides to Class I and Class II MHC has been published (Sette et al., Mol. Immunol. 31:813, 1994; Sidney et al., in Current Protocols in Immunology, Margulies, Ed., John Wiley & Sons, New York, Section 18.3, 1998). Briefly, purified MHC molecules (5 to 500 nM) are incubated with various unlabeled peptide inhibitors and 1-10 nM 1251-radiolabeled probe peptides for 48h in PBS containing 0.05% Nonidet P-40 (NP40) (or 20% w/v digitonin for H-2 IA assays) in the presence of a protease inhibitor cocktail. All assays are at pH 7.0 with the exception of DRB1*0301, which was performed at pH 4.5, and DRB1*1601 (DR2w21β1) and DRB4*0101 (DRw53), which were performed at pH 5.0.


Following incubation, MHC-peptide complexes are separated from free peptide by gel filtration on 7.8 mm×15 cm TSK200 columns (TosoHaas 16215, Montgomeryville, Pa.). Because the large size of the radiolabeled peptide used for the DRB1*1501 (DR2w2β1) assay makes separation of bound from unbound peaks more difficult under these conditions, all DRB1*1501 (DR2w2β1) assays were performed using a 7.8 mm×30 cm TSK2000 column eluted at 0.6 mLs/min. The eluate from the TSK columns is passed through a Beckman 170 radioisotope detector, and radioactivity is plotted and integrated using a Hewlett-Packard 3396A integrator, and the fraction of peptide bound is determined.


Radiolabeled peptides are iodinated using the chloramine-T method. Typically, in preliminary experiments, each MHC preparation is titered in the presence of fixed amounts of radiolabeled peptides to determine the concentration of HLA molecules necessary to bind 10-20% of the total radioactivity. All subsequent inhibition and direct binding assays are performed using these HLA concentrations.


Since under these conditions flabell<[HLA] and IC50≥[HLA], the measured IC50 values are reasonable approximations of the true KD values. Peptide inhibitors are typically tested at concentrations ranging from 120 μg/ml to 1.2 ng/ml, and are tested in two to four completely independent experiments. To allow comparison of the data obtained in different experiments, a relative binding figure is calculated for each peptide by dividing the IC50 of a positive control for inhibition by the IC50 for each tested peptide (typically unlabeled versions of the radiolabeled probe peptide). For database purposes, and inter-experiment comparisons, relative binding values are compiled. These values can subsequently be converted back into IC50 nM values by dividing the IC50 nM of the positive controls for inhibition by the relative binding of the peptide of interest. This method of data compilation has proven to be the most accurate and consistent for comparing peptides that have been tested on different days, or with different lots of purified MHC.


Because the antibody used for HLA-DR purification (LB3.1) is α-chain specific, β1 molecules are not separated from β3 (and/or β4 and β5) molecules. The β1 specificity of the binding assay is obvious in the cases of DRB1*0101 (DR1), DRB1*0802 (DR8w2), and DRB1*0803 (DR8w3), where no β3 is expressed. It has also been demonstrated for DRB1*0301 (DR3) and DRB3*0101 (DR52a), DRB1*0401 (DR4w4), DRB1*0404 (DR4w14), DRB1*0405 (DR4w15), DRB1*1101 (DR5), DRB1*1201 (DR5w12), DRB1*1302 (DR6w19) and DRB1*0701 (DR7). The problem of β chain specificity for DRB1*1501 (DR2w2β1), DRB5*0101 (DR2w2β2), DRB1*1601 (DR2w21β1), DRB5*0201 (DR51Dw21), and DRB4*0101 (DRw53) assays is circumvented by the use of fibroblasts. Development and validation of assays with regard to DRβ molecule specificity have been described previously (see, e.g., Southwood et al., J. Immunol. 160:3363-3373, 1998).


The live cell/flow cytometry-based assays can also be used. This is a well-established assay utilizing the TAP-deficient hybridoma cell line T2 (American Type Culture Collection (ATCC Accession No. CRL-1992), Manassas, Va.). The TAP deficiency in this cell line leads to inefficient loading of MHCI in the ER and an excess of empty MHCIs. Salter and Cresswell, EMBO J. 5:943-49 (1986); Salter, Immunogenetics 21:235-46 (1985). Empty MHCIs are highly unstable, and are therefore short-lived. When T2 cells are cultured at reduced temperatures, empty MHCIs appear transiently on the cell surface, where they can be stabilized by the exogenous addition of MHCI-binding peptides. To perform this binding assay, peptide-receptive MHCIs were induced by culturing aliquots of 107 T2 cells overnight at 26° C. in serum free AIM-V medium alone, or in medium containing escalating concentrations (0.1 to 100 μM) of peptide. Cells were then washed twice with PBS, and subsequently incubated with a fluorescent tagged HLA-A0201-specific monoclonal antibody, BB7.2, to quantify cell surface expression. Samples were acquired on a FACS Calibur instrument (Becton Dickinson) and the mean fluorescence intensity (MFI) determined using the accompanying Cellquest software.


Example 3: Confirmation of Immunogenicity

In vitro education (IVE) assays are used to test the ability of each test peptide to expand CD8+ T-cells. Mature professional APCs are prepared for these assays in the following way. 80-90×106 PBMCs isolated from a healthy human donor are plated in 20 ml of RPMI media containing 2% human AB serum, and incubated at 37° C. for 2 hours to allow for plastic adherence by monocytes. Non-adherent cells are removed and the adherent cells are cultured in RPMI, 2% human AB serum, 800 IU/ml of GM-CSF and 500 IU/ml of IL-4. After 6 days, TNF-alpha is added to a final concentration of 10 ng/ml. On day 7, the dendritic cells (DC) are matured either by the addition of 12.5 μg/ml poly I:C or 0.3 μg/ml of CD40L. The mature dendritic cells (mDC) are harvested on day 8, washed, and either used directly or cryopreserved for future use.


For the IVE of CD8+ T-cells, aliquots of 2×105 mDCs are pulsed with each peptide at a final concentration of 100 μM, incubated for 4 hours at 37° C., and then irradiated (2500 rads). The peptide-pulsed mDCs are washed twice in RPMI containing 2% human AB serum. 2×105 mDCs and 2×106 autologous CD8+ cells are plated per well of a 24-well plate in 2 ml of RPMI containing 2% human AB, 20 ng/ml IL-7 and 100 μg/ml of IL-12, and incubated for 12 days. The CD8+ T-cells are then re-stimulated with peptide-pulsed, irradiated mDCs. Two to three days later, 20 IU/ml IL-2 and 20 ng/IL7 are added. Expanding CD8+ T-cells are re-stimulated every 8-10 days, and are maintained in media containing IL-2 and IL-7. Cultures are monitored for peptide-specific T-cells using a combination of functional assays and/or tetramer staining. Parallel IVEs with the modified and parent peptides allowed for comparisons of the relative efficiency with which the peptides expanded peptide-specific T-cells.


Quantitative and Functional Assessment of CD8+ T-Cells
Tetramer Staining

MHC tetramers are purchased or manufactured on-site, and are used to measure peptide-specific T-cell expansion in the IVE assays. For the assessment, tetramer is added to 1×105 cells in PBS containing 1% FCS and 0.1% sodium azide (FACS buffer) according to manufacturer's instructions. Cells are incubated in the dark for 20 minutes at room temperature. Antibodies specific for T-cell markers, such as CD8, are then added to a final concentration suggested by the manufacturer, and the cells are incubated in the dark at 4° C. for 20 minutes. Cells are washed with cold FACS buffer and resuspended in buffer containing 1% formaldehyde. Cells are acquired on a FACS Calibur (Becton Dickinson) instrument, and are analyzed by use of Cellquest software (Becton Dickinson). For analysis of tetramer positive cells, the lymphocyte gate is taken from the forward and side-scatter plots. Data are reported as the percentage of cells that were CD8+/Tetramer+.


ELISPOT

Peptide-specific T-cells are functionally enumerated using the ELISPOT assay (BD Biosciences), which measures the release of IFNgamma from T-cells on a single cell basis. Target cells (T2 or HLA-A0201 transfected C1Rs) were pulsed with 10 μM peptide for 1 hour at 37° C., and washed three times. 1×105 peptide-pulsed targets are co-cultured in the ELISPOT plate wells with varying concentrations of T-cells (5×102 to 2×103) taken from the IVE culture. Plates are developed according to the manufacturer's protocol, and analyzed on an ELISPOT reader (Cellular Technology Ltd.) with accompanying software. Spots corresponding to the number of IFNgamma-producing T-cells are reported as the absolute number of spots per number of T-cells plated. T-cells expanded on modified peptides are tested not only for their ability to recognize targets pulsed with the modified peptide, but also for their ability to recognize targets pulsed with the parent peptide.


CD107 Staining

CD107a and b are expressed on the cell surface of CD8+ T-cells following activation with cognate peptide. The lytic granules of T-cells have a lipid bilayer that contains lysosomal-associated membrane glycoproteins (“LAMPs”), which include the molecules CD107a and b. When cytotoxic T-cells are activated through the T-cell receptor, the membranes of these lytic granules mobilize and fuse with the plasma membrane of the T-cell. The granule contents are released, and this leads to the death of the target cell. As the granule membrane fuses with the plasma membrane, C107a and b are exposed on the cell surface, and therefore are markers of degranulation. Because degranulation as measured by CD107 a and b staining is reported on a single cell basis, the assay is used to functionally enumerate peptide-specific T-cells. To perform the assay, peptide is added to HLA-A0201-transfected cells C1R to a final concentration of 20 μM, the cells were incubated for 1 hour at 37° C., and washed three times. 1×105 of the peptide-pulsed C1R cells were aliquoted into tubes, and antibodies specific for CD107 a and b are added to a final concentration suggested by the manufacturer (Becton Dickinson). Antibodies are added prior to the addition of T-cells in order to “capture” the CD107 molecules as they transiently appear on the surface during the course of the assay. 1×105 T-cells from the IVE culture are added next, and the samples were incubated for 4 hours at 37° C. The T-cells are further stained for additional cell surface molecules such as CD8 and acquired on a FACS Calibur instrument (Becton Dickinson). Data is analyzed using the accompanying Cellquest software, and results were reported as the percentage of CD8+CD107 a and b+ cells.


CTL Lysis

Cytotoxic activity is measured using a chromium release assay. Target T2 cells are labeled for 1 hour at 37° C. with Na51Cr and washed 5×103 target T2 cells were then added to varying numbers of T-cells from the IVE culture. Chromium release is measured in supernatant harvested after 4 hours of incubation at 37° C. The percentage of specific lysis is calculated as: Experimental release-spontaneous release/Total release-spontaneous releasex100


Example 4: Selection of CTL and HTL Epitopes for Inclusion in an Tumor-Specific Vaccine

This example illustrates the procedure for the selection of peptide epitopes for vaccine compositions of the invention. The peptides in the composition can be in the form of a nucleic acid sequence, either single or one or more sequences (i.e., minigene) that encodes peptide(s), or may be single and/or polyepitopic peptides.


Epitopes are selected which, upon administration, mimic immune responses that have been observed to be correlated with tumor clearance. For example, vaccine can include 1-2 epitopes that come from at least one tumor antigen region. Epitopes from one region can be used in combination with epitopes from one or more additional tumor antigen regions.


Epitopes can be selected that have a binding affinity of an IC50 of 500 nM or less for an HLA class I molecule, or for class II, an IC50 of 1000 nM or less.


When creating a polyepitopic compositions, e.g. a minigene, it is typically desirable to generate the smallest peptide possible that encompasses the epitopes of interest. The principles employed are similar, if not the same, as those employed when selecting a peptide comprising nested epitopes. Additionally, however, upon determination of the nucleic acid sequence to be provided as a minigene, the peptide sequence encoded thereby is analyzed to determine whether any “junctional epitopes” have been created. A junctional epitope is a potential HLA binding epitope, as predicted, e.g., by motif analysis. Junctional epitopes are generally to be avoided because the recipient may bind to an HLA molecule and generate an immune response to that epitope, which is not present in a native protein sequence.


Peptide epitopes for inclusion in vaccine compositions are, for example, selected from those listed in the Tables. A vaccine composition comprised of selected peptides, when administered, is safe, efficacious, and elicits an immune response similar in magnitude of an immune response that inhibits tumor growth.


Example 5: Peptide Composition for Prophylactic or Therapeutic Uses

Immunogenic or vaccine compositions of the invention are used to inhibit tumor growth. For example, a polyepitopic composition (or a nucleic acid comprising the same) containing multiple CTL and HTL epitopes is administered to individuals having tumors. The composition is provided as a single lipidated polypeptide that encompasses multiple epitopes. The composition is administered in an aqueous carrier comprised of alum. The dose of peptide for the initial immunization is from about 1 to about 50,000 μg, generally 100-5,000 μg, for a 70 kg patient. The initial administration is followed by booster dosages at 4 weeks followed by evaluation of the magnitude of the immune response in the patient, by techniques that determine the presence of epitope-specific CTL populations in a PBMC sample. Additional booster doses are administered as required. The composition is found to be both safe and efficacious to inhibit tumor growth.


Alternatively, the polyepitopic composition can be administered as a nucleic acid, for example as RNA, in accordance with methodologies known in the art and disclosed herein.


Non-mutated protein epitope binding agents, such as TCR or CARs can be can be administered in accordance with methodologies known in the art and disclosed herein. The binding agents can be administered as polypeptides or polynucleotides, for example RNA, encoding the binding agents, or as a cellular therapy, by administering cells expressing the binding agents.


Non-mutated protein epitope peptides, polynucleotides, binding agents, or cells expressing these molecules can be delivered to the same patient via multiple methodologies known in the art, and can further be combined with other cancer therapies (e.g., chemotherapy, surgery, radiation, checkpoint inhibitors, etc.).


Example 6. Administration of Compositions Using Dendritic Cells

Vaccines comprising epitopes of the invention may be administered using dendritic cells. In this example, the peptide-pulsed dendritic cells can be administered to a patient to stimulate a CTL response in vivo. In this method dendritic cells are isolated, expanded, and pulsed with a vaccine comprising peptide CTL and HTL epitopes of the invention. The dendritic cells are infused back into the patient to elicit CTL and HTL responses in vivo. The induced CTL and HTL then destroy (CTL) or facilitate destruction (HTL) of the specific target tumor cells that bear the proteins from which the epitopes in the vaccine are derived.


Alternatively, ex vivo CTL or HTL responses to a particular tumor-associated antigen can be induced by incubating in tissue culture the patient's, or genetically compatible, CTL or HTL precursor cells together with a source of antigen-presenting cells, such as dendritic cells, and the appropriate immunogenic peptides.


After an appropriate incubation time (typically about 7-28 days), in which the precursor cells are activated and expanded into effector cells, the cells are infused back into the patient, where they will destroy (CTL) or facilitate destruction (HTL) of their specific target cells, i.e., tumor cells.


PARAGRAPHS OF THE EMBODIMENTS

An isolated antigenic peptide comprising an epitope from a sequence in Table 1 or 2.


An isolated antigenic peptide 100 amino acids or less in length which comprises an epitope from a sequence in Table 1 or 2.


An isolated antigenic peptide comprising an epitope from a sequence in Table 3 or 4.


An isolated antigenic peptide 100 amino acids or less in length which comprises an epitope from a sequence in Table 3 or 4.


An isolated antigenic peptide comprising an epitope from a sequence in Table 5 or 6.


An isolated antigenic peptide 100 amino acids or less in length which comprises an epitope from a sequence in Table 5 or 6.


The isolated antigenic peptide of paragraph [00291] or [00292], wherein the isolated antigenic peptide is a retroviral antigen.


The isolated antigenic peptide of paragraph [00293] or [00294], wherein the isolated antigenic peptide is a non-mutated overexpressed antigen.


The isolated antigenic peptide of paragraph [00295] or [00296], wherein the isolated antigenic peptide is a viral antigen.


The isolated antigenic peptide of any of paragraphs [00291]-[00299], which is between about 5 to about 50 amino acids in length.


The isolated antigenic peptide of any of paragraphs [00291]-[00300], which is between about 15 to about 35 amino acids in length.


The isolated antigenic peptide of paragraph [00301], which is about 15 amino acids or less in length.


The isolated antigenic peptide of paragraph [00302], which is between about 8 and about 11 amino acids in length.


The isolated antigenic peptide of paragraph [00303], which is 9 or 10 amino acids in length.


The isolated antigenic peptide of any of paragraphs [00291]-[00304], which binds major histocompatibility complex (MHC) class I.


The isolated antigenic peptide of paragraph [00305], which binds MHC class I with a binding affinity of less than about 500 nM.


The isolated antigenic peptide of any of paragraphs [00291]-[00296], which is about 30 amino acids or less in length.


The isolated antigenic peptide of paragraph [00307], which is between about 6 and about 25 amino acids in length.


The isolated antigenic peptide of paragraph [00308], which is between about 15 and about 24 amino acids in length.


The isolated antigenic peptide of paragraph [00308], which is between about 9 and about 15 amino acids in length.


The isolated antigenic peptide of any of paragraphs [00291]-[00296] and [00307]-[00310], which binds MHC class II.


The isolated antigenic peptide of paragraph [00311], which binds MHC class II with a binding affinity of less than about 1000 nM.


The isolated antigenic peptide of any of paragraphs [00291]-[00312], further comprising flanking amino acids.


The isolated antigenic peptide of paragraph [00313], wherein the flanking amino acids are not native flanking amino acids.


The isolated antigenic peptide of any of paragraphs [00291]-[00314], which is linked to at least a second antigenic peptide.


The isolated antigenic peptide of paragraph [00315], wherein peptides are linked using a poly-glycine or poly-serine linker.


The isolated antigenic peptide of paragraph [00315] or [00316], wherein the second antigenic peptide binds MHC class I or class II with a binding affinity of less than about 1000 nM.


The isolated antigenic peptide of paragraph [00317], wherein the second antigenic peptide binds MHC class I or class II with a binding affinity of less than about 500 nM.


The isolated antigenic peptide of paragraph [00317] or [00318], wherein both of the epitopes bind to human leukocyte antigen (HLA) -A, —B, —C, -DP, -DQ, or -DR.


The isolated antigenic peptide of any of paragraphs [00317]-[00319], wherein the isolated antigenic peptide binds a class I HLA and the second antigenic peptide binds a class II HLA.


The isolated antigenic peptide of any of paragraphs [00317]-[00319], wherein the isolated antigenic peptide binds a class II HLA and the second antigenic peptide binds a class I HLA.


The isolated antigenic peptide of any of paragraphs [00291]-[00321], further comprising modifications which increase in vivo half-life, cellular targeting, antigen uptake, antigen processing, MHC affinity, MHC stability, or antigen presentation.


The isolated antigenic peptide of paragraph [00322], wherein the modification is conjugation to a carrier protein, conjugation to a ligand, conjugation to an antibody, PEGylation, polysialylation HESylation, recombinant PEG mimetics, Fc fusion, albumin fusion, nanoparticle attachment, nanoparticulate encapsulation, cholesterol fusion, iron fusion, acylation, amidation, glycosylation, side chain oxidation, phosphorylation, biotinylation, the addition of a surface active material, the addition of amino acid mimetics, or the addition of unnatural amino acids.


The isolated antigenic peptide of paragraph [00322], wherein the cells that are targeted are antigen presenting cells.


The isolated antigenic peptide of paragraph [00324], wherein the antigen presenting cells are dendritic cells.


The isolated antigenic peptide of paragraph [00325], wherein the dendritic cells are targeted using DEC205, XCR1, CD197, CD80, CD86, CD123, CD209, CD273, CD283, CD289, CD184, CD85h, CD85j, CD85k, CD85d, CD85g, CD85a, CD141, CD11c, CD83, TSLP receptor, or CD1a marker.


The isolated antigenic peptide of paragraph [00326], wherein the dendritic cells are targeted using the CD141, DEC205, or XCR1 marker.


An in vivo delivery system comprising the isolated antigenic peptide of any of paragraphs [00291]-[00327].


The delivery system of paragraph [00328], wherein the delivery system includes cell-penetrating peptides, nanoparticulate encapsulation, virus like particles, or liposomes.


The delivery system of paragraph [00328], wherein the cell-penetrating peptide is TAT peptide, herpes simplex virus VP22, transportan, or Antp.


A cell comprising the isolated antigenic peptide of any of paragraphs [00291]-[00327].


The cell of paragraph [00331], which is an antigen presenting cell.


The cell of paragraph [00332], which is a dendritic cell.


A composition comprising the isolated antigenic peptide of any of paragraphs [00291]-[00327].


The composition of paragraph [00334], wherein the composition comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 of the isolated antigenic peptides comprising a tumor-specific epitope defined in Table 1 or 2.


The composition of paragraph [00334], wherein the composition comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 of the isolated antigenic peptides comprising a tumor-specific epitope defined in Table 3 or 4.


The composition of paragraph [00334], wherein the composition comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 of the isolated antigenic peptides comprising a tumor-specific epitope defined in Table 5 or 6.


The composition of any of paragraphs [00335]-[00337], wherein the composition comprises between 2 and 20 antigenic peptides.


The composition of any one of paragraphs [00334]-[00338], wherein the composition further comprises at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 additional antigenic peptides.


The composition of paragraph [00339], wherein the composition comprises between about 4 and about 20 additional antigenic peptides.


The composition of any of paragraphs [00334]-[00340], wherein the additional antigenic peptide is specific for an individual patient's tumor.


The composition of paragraph [00341], wherein the patient specific antigenic peptide is selected by identifying sequence differences between the genome, exome, and/or transcriptome of the patient's tumor sample and the genome, exome, and/or transcriptome of a non-tumor sample.


The composition of paragraph [00337], wherein the samples are fresh or formalin-fixed paraffin embedded tumor tissues, freshly isolated cells, or circulating tumor cells.


The composition of paragraph [00342] or [00343], wherein the sequence differences are determined by Next Generation Sequencing.


An isolated polynucleotide encoding the isolated antigenic peptide of any of paragraphs [00291]-[00300].


The isolated polynucleotide paragraph [00345], which is RNA, optionally a self-amplifying RNA.


The isolated polynucleotide of paragraph [00346], wherein the RNA is modified to increase stability, increase cellular targeting, increase translation efficiency, adjuvanticity, cytosol accessibility, and/or decrease cytotoxicity.


The isolated polynucleotide of paragraph [00347], wherein the modification is conjugation to a carrier protein, conjugation to a ligand, conjugation to an antibody, codon optimization, increased GC-content, incorporation of modified nucleosides, incorporation of 5′-cap or cap analog, and/or incorporation of an unmasked poly-A sequence.


A cell comprising the polynucleotide of any of paragraphs [00345]-[00348].


A vector comprising the polynucleotide of any one of paragraphs [00345]-[00348].


The vector of paragraph [00350], in which the polynucleotide is operably linked to a promoter.


The vector of paragraphs [00350] or [00351], which is a self-amplifying RNA replicon, plasmid, phage, transposon, cosmid, virus, or virion.


The vector of paragraph [00352], which is an adeno-associated virus, herpesvirus, lentivirus, or pseudotypes thereof.


An in vivo delivery system comprising the isolated polynucleotide of any of paragraphs [00345]-[00348].


The delivery system of paragraph [00350], wherein the delivery system includes spherical nucleic acids, viruses, virus-like particles, plasmids, bacterial plasmids, or nanoparticles.


A cell comprising the vector or delivery system of any of paragraphs [00350]-[00355].


The cell of paragraph [00356], which is an antigen presenting cell.


The cell of paragraph [00357], which is a dendritic cell.


The cell of paragraph [00358], which is an immature dendritic cell.


A composition comprising at least one polynucleotide of any of paragraphs [00345]-[00348].


The composition of paragraph [00360], wherein the composition comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 of the isolated polynucleotides.


The composition of paragraph [00361], wherein the composition comprises between about 2 and about 20 polynucleotides.


The composition of any one of paragraphs [00360]-[00362], wherein the composition further comprises at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 additional antigenic polynucleotides encoding for additional antigenic peptides.


The composition of paragraph [00363], wherein the composition comprises between about 4 and about 20 additional antigenic polynucleotides.


The composition of paragraph [00363], wherein the isolated polynucleotides and the additional antigenic polynucleotides are linked.


The composition of paragraph [00365], wherein the polynucleotides are linked using nucleic acids that encode a poly-glycine or poly-serine linker.


The composition of any of paragraphs [00360]-[00366], wherein at least one of the additional antigenic peptide is specific for an individual patient's tumor.


The composition of paragraph [00367], wherein the patient specific antigenic peptide is selected by identifying sequence differences between the genome, exome, and/or transcriptome of the patient's tumor sample and the genome, exome, and/or transcriptome of a non-tumor sample.


The composition of paragraph [00368], wherein the samples are fresh or formalin-fixed paraffin embedded tumor tissues, freshly isolated cells, or circulating tumor cells.


The composition of paragraphs [00368] or [00369], wherein the sequence differences are determined by Next Generation Sequencing.


A T cell receptor (TCR) capable of binding at least one antigenic peptide listed in any of paragraphs [00291]-[00324].


The TCR of paragraph [00371], which is capable of binding the isolated antigenic peptide in the context of MHC class I or class II.


A chimeric antigen receptor comprising: (i) a T cell activation molecule; (ii) a transmembrane region; and (iii) an antigen recognition moiety capable of binding an isolated antigenic peptide of any one of paragraphs [00291]-[00324].


The chimeric antigen receptor of paragraph [00373], wherein CD3-zeta is the T cell activation molecule.


The chimeric antigen receptor of paragraph [00373] or [00374], further comprising at least one costimulatory signaling domain.


The chimeric antigen receptor of any of paragraphs [00373]-[00375], wherein the signaling domain is CD28, 4-1BB, ICOS, OX40, ITAM, or Fc epsilon RI-gamma.


The chimeric antigen receptor of any of paragraphs [00373]-[00376], wherein the antigen recognition moiety is capable of binding the isolated antigenic peptide in the context of MHC class I or class II.


The chimeric antigen receptor of any of paragraphs [00373]-[00377], comprising the CD3-zeta, CD28, CTLA-4, ICOS, BTLA, KIR, LAG3, CD137, OX40, CD27, CD40L, Tim-3, A2aR, or PD-1 transmembrane region.


The chimeric antigen receptor of any of paragraphs [00373]-[00378], wherein the tumor-specific epitope is located in the extracellular domain of a tumor associated polypeptide.


A T cell comprising the T cell receptor or chimeric antigen receptor of any of paragraphs [00371]-[00379].


The T cell of paragraph [00380], which is a helper or cytotoxic T cell.


A nucleic acid comprising a promoter operably linked to a polynucleotide encoding the T cell receptor of paragraph [00371] or [00372].


The nucleic acid of paragraph [00382], wherein the TCR is capable of binding the at least one antigenic peptide in the context of major histocompatibility complex (MHC) class I or class II.


A nucleic acid comprising a promoter operably linked to a polynucleotide encoding the chimeric antigen receptor of any of paragraphs [00373]-[00379].


The nucleic acid of paragraph [00384], wherein the antigen recognition moiety is capable of binding the at least one antigenic peptide in the context of major histocompatibility complex (MHC) class I or class II.


The nucleic acid of paragraph [00384] or [00385], wherein the tumor-specific epitope is located in the extracellular domain of a tumor associated polypeptide.


The nucleic acid of any of paragraphs [00384]-[00386], comprising the CD3-zeta, CD28, CTLA-4, ICOS, BTLA, KIR, LAG3, CD137, OX40, CD27, CD40L, Tim-3, A2aR, or PD-1 transmembrane region.


An antibody capable of binding at least one antigenic peptide listed in Table 1 or 2.


An antibody capable of binding at least one antigenic peptide listed in Table 3 or 4.


An antibody capable of binding at least one antigenic peptide listed in Table 5 or 6.


An antibody of paragraph [00388], wherein the at least one antigenic peptide listed in Table 1 or 2 is a retroviral antigenic peptide.


An antibody of paragraph [00389], wherein the at least one antigenic peptide listed in Table 3 or 4 is a non-mutated overexpressed antigenic peptide.


An antibody of paragraph [00390], wherein the at least one antigenic peptide listed in Table 5 or 6 is a viral antigenic peptide.


A modified cell transfected or transduced with the nucleic acid of any one of paragraphs [00382]-[00387].


The modified cell of paragraph [00394], wherein the modified cell is a T cell, tumor infiltrating lymphocyte, NK-T cell, TCR-expressing cell, CD4+ T cell, CD8+ T cell, or NK cell.


A composition comprising the T cell receptor or chimeric antigen receptor of any of paragraphs [00371]-[00379].


A composition comprising autologous patient T cells containing the T cell receptor or chimeric antigen receptor of any of paragraphs [00371]-[00379].


The composition of paragraph [00395] or [00396], further comprising an immune checkpoint inhibitor.


The composition of paragraph [00396] or [00397], further comprising at least two immune checkpoint inhibitors.


The composition of paragraph [00398] or [00399], wherein each of the immune checkpoint inhibitors inhibits a checkpoint protein selected from the group consisting of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, and B-7 family ligands or a combination thereof.


The composition of paragraph [00398] or [00399], wherein each of the immune checkpoint inhibitors interacts with a ligand of a checkpoint protein selected from the group consisting of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, and B-7 family ligands or a combination thereof.


The composition of any of paragraphs [00334]-[00344], [00360]-[00369], and [00396]-[00401], further comprising an immune modulator or adjuvant.


The composition of paragraph [00402], wherein the immune modulator is a co-stimulatory ligand, a TNF ligand, an Ig superfamily ligand, CD28, CD80, CD86, ICOS, CD40L, OX40, CD27, GITR, CD30, DR3, CD69, or 4-1BB.


The composition of paragraph [00402], wherein the immune modulator is at least one cancer cell or cancer cell extract.


The composition of paragraph [00404], wherein the cancer cell is autologous to the subject in need of the composition.


The composition of paragraph [00405], wherein the cancer cell has undergone lysis or been exposed to UV radiation.


The composition of paragraph [00402], wherein the composition further comprises an adjuvant.


The composition of paragraph [00407], wherein the adjuvant is selected from the group consisting of: Poly(I:C), Poly-ICLC, STING agonist, 1018 ISS, aluminum salts, Amplivax, AS15, BCG, CP-870,893, CpG7909, CyaA, dSLIM, GM-CSF, IC30, IC31, Imiquimod, ImuFact IMP321, IS Patch, ISS, ISCOMATRIX, JuvImmune, LipoVac, MF59, monophosphoryl lipid A, Montanide IMS 1312 VG, Montanide ISA 206 VG, Montanide ISA 50 V2, Montanide ISA 51 VG, OK-432, OM-174, OM-197-MP-EC, ISA-TLR2 agonist, ONTAK, PepTel®. vector system, PLG microparticles, resiquimod, SRL172, virosomes and other virus-like particles, YF-17D, VEGF trap, R848, beta-glucan, Pam3Cys, Pam3CSK4, acrylic or methacrylic polymers, copolymers of maleic anhydride, and QS21 stimulon.


The composition of paragraph [00407] or [00408], wherein the adjuvant induces a humoral when administered to a subject.


The composition of paragraph [00409], wherein the adjuvant induces a T helper cell type 1 when administered to a subject.


A method of inhibiting growth of a tumor cell expressing a tumor-specific epitope defined in Table 1 or 2, comprising contacting the tumor cell with the peptide, polynucleotide, delivery system, vector, composition, antibody, or cells of any of paragraphs [00291]-[00410].


A method of inhibiting growth of a tumor cell expressing a tumor-specific epitope defined in Table 3 or 4, comprising contacting the tumor cell with the peptide, polynucleotide, delivery system, vector, composition, antibody, or cells of any of paragraphs [00291]-[00410].


A method of inhibiting growth of a tumor cell expressing a tumor-specific epitope defined in Table 5 or 6, comprising contacting the tumor cell with the peptide, polynucleotide, delivery system, vector, composition, antibody, or cells of any of paragraphs [00291]-[00410].


A method of treating cancer or initiating, enhancing, or prolonging an anti-tumor response in a subject in need thereof comprising administering to the subject the peptide, polynucleotide, vector, composition, antibody, or cells of any of paragraphs [00291]-[00410].


The method of any of paragraphs [00411]-[00414], wherein the subject is a human.


The method of paragraph [00415], wherein the subject has cancer.


The method of paragraph [00416], wherein the cancer is selected from the group consisting of urogenital, renal, gynecological, lung, gastrointestinal, head and neck cancer, malignant glioblastoma, malignant mesothelioma, non-metastatic or metastatic breast cancer, malignant melanoma, Merkel Cell Carcinoma or bone and soft tissue sarcomas, hematologic neoplasias, multiple myeloma, acute myelogenous leukemia, chronic myelogenous leukemia, myelodysplastic syndrome and acute lymphoblastic leukemia, non-small cell lung cancer (NSCLC), triple-negative breast cancer (TNBC), smoldering myeloma (SMM), breast cancer, metastatic colorectal cancers, hormone sensitive or hormone refractory prostate cancer, colorectal cancer, ovarian cancer, hepatocellular cancer, renal cell cancer, pancreatic cancer, gastric cancer, esophageal cancers, hepatocellular cancers, cholangiocellular cancers, head and neck squamous cell cancer soft tissue sarcoma, and small cell lung cancer.


The method of any of paragraphs [00411]-[00417], wherein the subject has undergone surgical removal of the tumor.


The method of any of paragraphs [00411]-[00418], wherein the peptide, polynucleotide, vector, composition, or cells is administered via intravenous, intraperitoneal, intratumoral, intradermal, or subcutaneous administration.


The method of paragraph [00419], wherein the peptide, polynucleotide, vector, composition, or cells is administered into an anatomic site that drains into a lymph node basin.


The method of paragraph [00420], wherein administration is into multiple lymph node basins.


The method of any one of paragraphs [00411]-[00421], wherein administration is by a subcutaneous or intradermal route.


The method of paragraph [00419], wherein peptide is administered.


The method of paragraph [00423], wherein administration is intratumorally.


The method of paragraph [00419], wherein polynucleotide, optionally RNA, is administered.


The method of paragraph [00419] or [00425], wherein the polynucleotide is administered intravenously.


The method of paragraph [00419], wherein the cell is a T cell or dendritic cell.


The method of paragraph [00419] or [00427], wherein the peptide or polynucleotide comprises an antigen presenting cell targeting moiety.


The method of any of paragraphs [00411]-[00428], further comprising administering at least one immune checkpoint inhibitor to the subject.


The method of paragraph [00429], wherein the checkpoint inhibitor is a biologic therapeutic or a small molecule.


The method of paragraph [00429] or [00430], wherein the checkpoint inhibitor is selected from the group consisting of a monoclonal antibody, a humanized antibody, a fully human antibody and a fusion protein or a combination thereof.


The method of any of paragraphs [00429]-[00431], wherein the checkpoint inhibitor inhibits a checkpoint protein selected from the group consisting of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, and B-7 family ligands or a combination thereof.


The method of any of paragraphs [00429]-[00432], wherein the checkpoint inhibitor interacts with a ligand of a checkpoint protein selected from the group consisting of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, and B-7 family ligands or a combination thereof.


The method of any of paragraphs [00429]-[00433], wherein two or more checkpoint inhibitors are administered.


The method of paragraph [00434], wherein the checkpoint inhibitors are: (i) ipilimumab or tremelimumab, and (ii) nivolumab.


The method of any of paragraphs [00429]-[00435], wherein the checkpoint inhibitor and the composition are administered simultaneously or sequentially in any order.


The method of paragraph [00436], wherein the peptide, polynucleotide, vector, composition, or cells is administered prior to the checkpoint inhibitor.


The method of paragraph [00436], wherein the peptide, polynucleotide, vector, composition, or cells is administered after the checkpoint inhibitor.


The method of paragraph [00436], wherein administration of the checkpoint inhibitor is continued throughout antigen peptide, polynucleotide, vector, composition, or cell therapy.


The method of any of paragraphs [00429]-[00439], wherein the antigen peptide, polynucleotide, vector, composition, or cell therapy is administered to subjects that only partially respond or do not respond to checkpoint inhibitor therapy.


The method of any one of paragraphs [00411]-[00428], wherein the composition is administered intravenously or subcutaneously.


The method of any one of paragraphs [00429]-[00440], wherein the checkpoint inhibitor is administered intravenously or subcutaneously.


The method of any one of paragraphs [00429]-[00441], wherein the checkpoint inhibitor is administered subcutaneously within about 2 cm of the site of administration of the composition.


The method of paragraph [00443], wherein the composition is administered into the same draining lymph node as the checkpoint inhibitor.


The method of any of paragraphs [00411]-[00444], further comprising administering an additional therapeutic agent to the subject either prior to, simultaneously with, or after treatment with the peptide, polynucleotide, vector, composition, or cells.


The method of paragraph [00445], wherein the additional agent is a chemotherapeutic agent, an immunomodulatory drug, an immune metabolism modifying drug, a targeted therapy, radiation an anti-angiogenesis agent, or an agent that reduces immune-suppression.


The method of paragraph [00446], wherein the chemotherapeutic agent is an alkylating agent, a topoisomerase inhibitor, an anti-metabolite, or an anti-mitotic agent.


The method of paragraph [00445], wherein the additional agent is an anti-glucocorticoid induced tumor necrosis factor family receptor (GITR) agonistic antibody or antibody fragment, ibrutinib, docetaxeol, cisplatin, or cyclophosphamide.


The method of any of paragraphs [00411]-[00448], which elicits a CD4+ T cell immune response.


The method of any of paragraphs [00411]-[00449], which elicits a CD4+ T cell immune response and a CD8+ T cell immune response.


A method for stimulating an immune response in a subject, comprising administering an effective amount of modified cells or composition of any of paragraphs [00394]-[00410].


The method of paragraph [00451], wherein the immune response is cytotoxic and/or humoral immune response.


The method of paragraph [00451], wherein the method stimulates a T cell-mediated immune response in a subject.


The method of paragraph [00453], wherein the T cell-mediated immune response is directed against a target cell.


The method of paragraph [00454], wherein the target cell is a tumor cell.


The method of any of paragraphs [00451]-[00455], wherein the modified cells are transfected or transduced in vivo.


The method of any of paragraphs [00451]-[00456], wherein the modified cells are transfected or transduced ex vivo.


The method of any of paragraphs [00451]-[00457], wherein the modified cells are autologous patient T cells.


The method of paragraph [00458], wherein the autologous patient T cells are obtained from a patient that has received an antigen peptide or nucleic acid vaccine.


The method of paragraph [00459], wherein the antigen peptide or nucleic acid vaccine comprises at least one personalized antigen.


The method of paragraph [00460], wherein the antigen peptide or nucleic acid vaccine comprises at least one additional antigenic peptide listed in Table 1 or 2.


The method of paragraph [00460], wherein the antigen peptide or nucleic acid vaccine comprises at least one additional antigenic peptide listed in Table 3 or 4.


The method of paragraph [00460], wherein the antigen peptide or nucleic acid vaccine comprises at least one additional antigenic peptide listed in Table 5 or 6.


The method of paragraph [00461], wherein the at least one additional antigenic peptide listed in Table 1 or 2 is a retroviral antigenic peptide.


The method of paragraph [00462], wherein the at least one additional antigenic peptide listed in Table 3 or 4 is a non-mutated overexpressed antigenic peptide.


The method of paragraph [00463], wherein the at least one additional antigenic peptide listed in Table 5 or 6 is a viral antigenic peptide.


The method of any of paragraphs [00461]-[00466], wherein the patient received a chemotherapeutic agent, an immunomodulatory drug, an immune metabolism modifying drug, targeted therapy or radiation prior to and/or during receipt of the antigen peptide or nucleic acid vaccine.


The method of any of paragraphs [00459]-[00467], wherein the patient receives treatment with at least one checkpoint inhibitor.


The method of any of paragraphs [00459]-[00468], wherein the autologous T cells are obtained from a patient that has already received at least one round of T cell therapy containing an antigen.


The method of any of paragraphs [00459]-[00469], wherein the method further comprises adoptive T cell therapy.


The method of paragraph [00470], wherein the adoptive T cell therapy comprises autologous T-cells.


The method of paragraph [00471], wherein the autologous T-cells are targeted against tumor antigens.


The method of paragraph [00470] or [00471], wherein the adoptive T cell therapy further comprises allogenic T-cells.


The method of paragraph [00473], wherein the allogenic T-cells are targeted against tumor antigens.


The method of any of paragraphs [00470]-[00474], wherein the adoptive T cell therapy is administered before the checkpoint inhibitor.


A method for evaluating the efficacy of any of paragraphs [00411]-[00475], comprising: (i) measuring the number or concentration of target cells in a first sample obtained from the subject before administering the modified cell, (ii) measuring the number concentration of target cells in a second sample obtained from the subject after administration of the modified cell, and (iii) determining an increase or decrease of the number or concentration of target cells in the second sample compared to the number or concentration of target cells in the first sample.


The method of paragraph [00476], wherein treatment efficacy is determined by monitoring a clinical outcome; an increase, enhancement or prolongation of anti-tumor activity by T cells; an increase in the number of anti-tumor T cells or activated T cells as compared with the number prior to treatment; B cell activity; CD4 T cell activity; or a combination thereof.


The method of paragraph [00477], wherein treatment efficacy is determined by monitoring a biomarker.


The method of paragraph [00478], wherein the biomarker is selected from the group consisting of CEA, Her-2/neu, bladder tumor antigen, thyroglobulin, alpha-fetoprotein, PSA, CA 125, CA19.9, CA 15.3, leptin, prolactin, osteopontin, IGF-II, CD98, fascin, sPIgR, 14-3-3 eta, troponin I, and b-type natriuretic peptide.


The method of paragraph [00477], wherein clinical outcome is selected from the group consisting of tumor regression; tumor shrinkage; tumor necrosis; anti-tumor response by the immune system; tumor expansion, recurrence or spread; or a combination thereof.


The method of paragraph [00477], wherein the treatment effect is predicted by presence of T cells or by presence of a gene signature indicating T cell inflammation or a combination thereof.


A method of treating cancer or initiating, enhancing, or prolonging an anti-tumor response in a subject in need thereof comprising administering to the subject:


the peptide, polynucleotide, vector, composition, antibody, or cells of any of paragraphs [00291]-[00410]; and


at least one checkpoint inhibitor.


The method of paragraph [00482], further comprising administration of an immunomodulator or adjuvant.


The method of paragraph [00483], wherein the immunomodulator or adjuvant is selected from the group consisting of Poly(I:C), Poly-ICLC, STING agonist, 1018 ISS, aluminum salts, Amplivax, AS15, BCG, CP-870,893, CpG7909, CyaA, dSLIM, GM-CSF, IC30, IC31, Imiquimod, ImuFact IMP321, IS Patch, ISS, ISCOMATRIX, JuvImmune, LipoVac, MF59, monophosphoryl lipid A, Montanide IMS 1312 VG, Montanide ISA 206 VG, Montanide ISA 50 V2, Montanide ISA 51 VG, OK-432, OM-174, OM-197-MP-EC, ISA-TLR2 agonist, ONTAK, PepTel® vector system, PLG microparticles, resiquimod, SRL172, virosomes and other virus-like particles, YF-17D, VEGF trap, R848, beta-glucan, Pam3Cys, Pam3CSK4, acrylic or methacrylic polymers, copolymers of maleic anhydride, and QS21 stimulon. a co-stimulatory ligand, a TNF ligand, an Ig superfamily ligand, CD28, CD80, CD86, ICOS, CD40L, OX40, CD27, GITR, CD30, DR3, CD69, or 4-1BB.


The method of paragraph [00484], wherein the immunomodulator or adjuvant is Poly-ICLC.


The method of any one of paragraphs [00482]-[00485], wherein the checkpoint inhibitor is an anti-PD1 antibody or antibody fragment.


The method of paragraph [00486], wherein the inhibitor of the PD-1 pathway is nivolumab.


The method of any one of paragraphs [00482]-[00485], wherein the checkpoint inhibitor is an anti-CTLA4 antibody or antibody fragment.


The method of paragraph [00488], wherein the anti-CTLA4 antibody is ipilimumab or tremelimumab.


The method of any one of paragraphs [00482]-[00489], wherein the method comprises administering both an anti-PD1 antibody and an anti-CTLA4 antibody.


The method of any one of paragraphs [00482]-[00489], wherein administration of the checkpoint inhibitor is initiated before initiation of administration of the peptide, polynucleotide, vector, composition, antibody, or cell.


The method of any one of paragraphs [00482]-[00489], wherein administration of the checkpoint inhibitor is initiated after initiation of administration of the peptide, polynucleotide, vector, composition, antibody, or cell.


The method of any one of paragraphs [00482]-[00489], wherein administration of the checkpoint inhibitor is initiated simultaneously with the initiation of administration of the peptide, polynucleotide, vector, composition, antibody, or cell.


The method of any one of paragraphs [00482]-[00493], wherein the peptide, polynucleotide, vector, composition, antibody, or cell is administered intravenously or subcutaneously.


The method of any one of paragraphs [00482]-[00493], wherein the checkpoint inhibitor is administered intravenously or subcutaneously.


The method of any one of paragraphs [00482]-[00495], wherein the checkpoint inhibitor is administered subcutaneously within about 2 cm of the site of administration of the peptide, polynucleotide, vector, composition, antibody, or cell.


The method of paragraph [00496], wherein the peptide, polynucleotide, vector, composition, antibody, or cell is administered into the same draining lymph node as the checkpoint inhibitor.


A kit comprising an antigen therapeutic of any of paragraphs [00291]-[00410].


The method of paragraph [00414], wherein the cancer is selected from the group consisting of: CRC, head and neck, stomach, lung squamous, lung adeno., prostate, bladder, stomach, renal cell carcinoma, and uterine.


The method of paragraph [00414], wherein the cancer is selected from the group consisting of: melanoma, lung squamous, DLBCL, uterine, head and neck, uterine, liver, and CRC.


The method of paragraph [00414], wherein the cancer is selected from the group consisting of: cervical, head and neck, anal, stomach, Burkitt's lymphoma, and nasopharyngeal carcinoma.


Provided herein is an immunogenic vaccine composition comprising a peptide comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6. In some embodiments, the peptide is a synthetic peptide. In some embodiments, the peptide is a recombinant peptide. In some embodiments, the peptide comprises a sequence from an endogenous retroviral protein. In some embodiments, the peptide comprises a sequence from an exogenous viral protein. In some embodiments, the peptide comprises a sequence of a protein expressed by a cancer cell of a subject with cancer, wherein the protein is expressed by the cancer cell at a level that is higher than a level expressed by a non-cancer cell of the subject. In some embodiments, the peptide is 100 amino acids or less in length. In some embodiments, the peptide is from about 5 to about 50 amino acids in length or from about 15 to about 35 amino acids in length. In some embodiments, the peptide is about 30 amino acids or less in length or about 15 amino acids or less in length. In some embodiments, the peptide comprises a sequence which binds a major histocompatibility complex (MHC) class I with a binding affinity of less than about 500 nM. In some embodiments, the peptide comprises a sequence which binds a major histocompatibility complex (MHC) class II with a binding affinity of less than about 1000 nM. In some embodiments, the peptide further comprises non-native amino acids flanking the at least 8 contiguous amino acids. In some embodiments, the composition further comprises a second peptide comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6, wherein the second antigenic peptide binds MHC class I or class II with a binding affinity of less than about 1000 nM. In some embodiments, the peptides are linked using a poly-glycine or poly-serine linker. In some embodiments, the second antigenic peptide binds MHC class I or class II with a binding affinity of less than about 1000 nM or less than about 500 nM. In some embodiments, the peptide further comprises a modification which increases in vivo half-life, cellular targeting, antigen uptake, antigen processing, MHC affinity, MHC stability, or antigen presentation. In some embodiments, the modification is conjugation to a carrier protein, conjugation to a ligand, conjugation to an antibody, PEGylation, polysialylation HESylation, recombinant PEG mimetics, Fc fusion, albumin fusion, nanoparticle attachment, nanoparticulate encapsulation, cholesterol fusion, iron fusion, acylation, amidation, glycosylation, side chain oxidation, phosphorylation, biotinylation, the addition of a surface active material, the addition of amino acid mimetics, or the addition of unnatural amino acids. In some embodiments, the peptide comprises a modification which increases targeting by antigen presenting cells. In some embodiments, the antigen presenting cells are dendritic cells. In some embodiments, the modification which increases targeting by the dendritic cells is a DEC205, XCR1, CD197, CD80, CD86, CD123, CD209, CD273, CD283, CD289, CD184, CD85h, CD85j, CD85k, CD85d, CD85g, CD85a, CD141, CD11c, CD83, TSLP receptor, or CD1a marker. In some embodiments, the composition comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 of peptides each comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6. In some embodiments, the composition comprises from 2 to 20 peptides each comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6. In some embodiments, the composition further comprises at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 additional antigenic peptides. In some embodiments, the additional antigenic peptides are specific for an individual patient's tumor. In some embodiments, the additional antigenic peptides are selected by identifying sequence differences between the genome, exome, and/or transcriptome of the patient's tumor sample and the genome, exome, and/or transcriptome of a non-tumor sample. In some embodiments, identifying sequence differences comprises performing Next Generation Sequencing. Provided herein is a composition comprising an antigen presenting cell comprising a peptide comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6. In some embodiments, the antigen presenting cell is a dendritic cell.


Provided herein is an in vivo delivery system comprising a composition described herein. In some embodiments, the delivery system includes a cell-penetrating peptide, nanoparticulate encapsulation, a virus like particle, or a liposome. In some embodiments, the cell-penetrating peptide is a TAT peptide, herpes simplex virus VP22, transportan, or Antp.


Provided herein is an immunogenic vaccine composition comprising a recombinant polynucleotide encoding a peptide comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6. In some embodiments, the recombinant polynucleotide is RNA, optionally a self-amplifying RNA. In some embodiments, the RNA is modified to increase stability, increase cellular targeting, increase translation efficiency, adjuvanticity, cytosol accessibility, and/or decrease cytotoxicity. In some embodiments, the modification is conjugation to a carrier protein, conjugation to a ligand, conjugation to an antibody, codon optimization, increased GC-content, incorporation of modified nucleosides, incorporation of 5′-cap or cap analog, and/or incorporation of an unmasked poly-A sequence.


Provided herein is a composition comprising a cell comprising a recombinant polynucleotide encoding a peptide comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6.


Provided herein is a composition comprising a vector comprising a polynucleotide comprising a sequence encoding a peptide comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6. In some embodiments, the polynucleotide is operably linked to a promoter. In some embodiments, the polynucleotide is a self-amplifying RNA replicon, plasmid, phage, transposon, cosmid, virus, or virion. In some embodiments, the virus is an adeno-associated virus, herpesvirus, lentivirus, or pseudotypes thereof.


Provided herein is an in vivo delivery system comprising a composition described herein. In some embodiments, the delivery system includes spherical nucleic acids, viruses, virus-like particles, plasmids, bacterial plasmids, or nanoparticle.


Provided herein is a T cell receptor (TCR) that specifically binds to a peptide:MHC complex, wherein the peptide of the peptide of the peptide:MHC complex is a peptide comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6.


Provided herein is a T cell comprising a T cell receptor (TCR) that specifically binds to a peptide:MHC complex, wherein the peptide of the peptide of the peptide:MHC complex is a peptide comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6. In some embodiments, the T cell is a helper or cytotoxic T cell. In some embodiments, the T cell is an autologous patient T cell.


Provided herein is a method of treating cancer in a subject in need thereof comprising administering to the subject a composition described herein; wherein the subject comprises cancer cells expressing a protein comprising at least 8 contiguous amino acids of a sequence in any one of Tables 1-6. In some embodiments, the subject is a human. In some embodiments, the cancer is selected from the group consisting of urogenital, gynecological, lung, gastrointestinal, head and neck cancer, malignant glioblastoma, malignant mesothelioma, non-metastatic or metastatic breast cancer, triple-negative breast cancer (TNBC), malignant melanoma, Merkel Cell Carcinoma or bone and soft tissue sarcomas, hematologic neoplasias, multiple myeloma, smoldering myeloma (SMM), acute myelogenous leukemia, chronic myelogenous leukemia, myelodysplastic syndrome and acute lymphoblastic leukemia, non-small cell lung cancer (NSCLC), breast cancer, metastatic colorectal cancers, hormone sensitive or hormone refractory prostate cancer, colorectal cancer, ovarian cancer, hepatocellular cancer, renal cell cancer, pancreatic cancer, gastric cancer, esophageal cancers, hepatocellular cancers, cholangiocellular cancers, head and neck squamous cell cancer soft tissue sarcoma, and small cell lung cancer. In some embodiments, the method further comprises administering at least one immune checkpoint inhibitor to the subject. In some embodiments, the checkpoint inhibitor inhibits a checkpoint protein selected from the group consisting of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GALS, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, and B-7 family ligands or a combination thereof.

Claims
  • 1.-51. (canceled)
  • 52. A method of treating a cancer in a subject in need thereof, the method comprising administering to the subject a peptide comprising a viral protein epitope sequence or a polynucleotide encoding the peptide, wherein the viral protein epitope sequence is encoded by a BALF5 gene of Epstein Barr virus (EBV) or an E6 gene of Human papillomavirus type 16 (HPV-16); and wherein: (a) the subject expresses a major histocompatibility complex (MHC) encoded by a human leukocyte antigen (HLA) A02:01 allele and the viral protein epitope sequence is SLYPSIIQA, VLDDGQQIRV, SLFVAPISL, TLMKGVELV, GLIPIDMYAV, or FAFRDLCIV;(b) the subject expresses an MHC encoded by an HLA A01:01 allele and the viral protein epitope sequence is ISEYRHYCY;(c) the subject expresses an MEW encoded by an HLA A03:01 allele and the viral protein epitope sequence is KISEYRHYCY;(d) the subject expresses an MHC encoded by an HLA B07:02 allele and the viral protein epitope sequence is NPYAVCDKCL; or(e) the subject expresses an MEW encoded by an HLA B08:01 allele and the viral protein epitope sequence is CVYCKQQLL.
  • 53. The method of claim 52, wherein the viral protein epitope sequence comprises SLYPSIIQA (SEQ ID NO: 398) and the subject expresses an MEW molecule encoded by an A02:01 HLA allele.
  • 54. The method of claim 52, wherein the viral protein epitope sequence comprises VLDDGQQIRV (SEQ ID NO: 420) and the subject expresses an MHC molecule encoded by an A02:01 HLA allele.
  • 55. The method of claim 52, wherein the viral protein epitope sequence comprises SLFVAPISL (SEQ ID NO: 394) and the subject expresses an MHC molecule encoded by an A02:01 HLA allele.
  • 56. The method of claim 52, wherein the viral protein epitope sequence comprises TLMKGVELV (SEQ ID NO: 409) and the subject expresses an MEW molecule encoded by an A02:01 HLA allele.
  • 57. The method of claim 52, wherein the viral protein epitope sequence comprises GLIPIDMYAV (SEQ ID NO: 273) and the subject expresses an MEW molecule encoded by an A02:01 HLA allele.
  • 58. The method of claim 52, wherein the viral protein epitope sequence comprises FAFRDLCIV (SEQ ID NO: 56) and the subject expresses an MHC molecule encoded by an A02:01 HLA allele.
  • 59. The method of claim 52, wherein the viral protein epitope sequence comprises ISEYRHYCY (SEQ ID NO: 58) and the subject expresses an MHC molecule encoded by an A01:01 HLA allele.
  • 60. The method of claim 52, wherein the viral protein epitope sequence comprises KISEYRHYCY (SEQ ID NO: 62) and the subject expresses an MEW molecule encoded by an A03:01 HLA allele.
  • 61. The method of claim 52, wherein the viral protein epitope sequence comprises NPYAVCDKCL (SEQ ID NO: 67) and the subject expresses an MEW molecule encoded by a B07:01 HLA allele.
  • 62. The method of claim 52, wherein the viral protein epitope sequence comprises CVYCKQQLL (SEQ ID NO: 51) and the subject expresses an MEW molecule encoded by a B08:01 HLA allele.
  • 63. The method of claim 52, wherein the peptide comprises at least two different viral protein epitope sequences, wherein the gene encoding each of the at least two different viral protein epitope sequences is BALF5 of EBV, E6 of EPV-16, or a combination thereof.
  • 64. The method of claim 52, wherein the viral protein epitope sequence binds an MHC class I with a binding affinity of 500 nM or less.
  • 65. The method of claim 52, wherein the peptide is from 8 to 100 amino acids in length.
  • 66. The method of claim 52, wherein the peptide further comprises a modification which increases in vivo half-life, cellular targeting, antigen uptake, antigen processing, MHC affinity, MHC stability, or antigen presentation.
  • 67. The method of claim 52, wherein the viral protein epitope sequence is encoded an E6 gene of HPV-16, and wherein the cancer is selected from the group consisting of cervical cancer, head and neck cancer, anal cancer, urogenital cancer, a gynecological cancer and uterine cancer.
  • 68. The method of claim 52, wherein the viral protein epitope sequence is encoded by a BALF5 gene of EBV, and wherein the cancer is selected from the group consisting of Burkitt's lymphoma, Hodgkin lymphoma, stomach cancer and nasopharyngeal carcinoma.
  • 69. The method of claim 52, further comprising administering an additional therapeutic agent to the subject for a combination therapy, wherein the additional therapeutic agent comprises a chemotherapeutic agent, an immunotherapeutic agent, or an immune checkpoint inhibitor.
  • 70. The method of claim 52, wherein the subject is a human.
  • 71. A composition comprising a peptide comprising a viral protein epitope sequence or a polynucleotide encoding the peptide, wherein the viral protein epitope sequence is encoded by a BALF5 gene of Epstein Barr virus (EBV) or an E6 gene of Human papillomavirus type 16 (HPV-16); wherein the viral protein epitope sequence is SLYPSIIQA, VLDDGQQIRV, SLFVAPISL, TLMKGVELV, GLIPIDMYAV, FAFRDLCIV, ISEYRHYCY, KISEYRHYCY, NPYAVCDKCL or CVYCKQQLL.
  • 72. A pharmaceutical composition comprising a therapeutically effective amount of the composition of claim 71 and pharmaceutically acceptable carrier.
CROSS REFERENCE

This application claims priority to U.S. Provisional Application No. 62/480,593, filed Apr. 3, 2017, U.S. Provisional Application No. 62/480,596, filed Apr. 3, 2017, and U.S. Provisional Application No. 62/480,597, filed Apr. 3, 2017, each of which is incorporated herein by reference in its entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2018/025933 4/3/2018 WO
Provisional Applications (3)
Number Date Country
62480593 Apr 2017 US
62480596 Apr 2017 US
62480597 Apr 2017 US