US20200024324A1 - New pdl2 compounds - Google Patents

New pdl2 compounds Download PDF

Info

Publication number
US20200024324A1
US20200024324A1 US16/344,445 US201716344445A US2020024324A1 US 20200024324 A1 US20200024324 A1 US 20200024324A1 US 201716344445 A US201716344445 A US 201716344445A US 2020024324 A1 US2020024324 A1 US 2020024324A1
Authority
US
United States
Prior art keywords
peptide fragment
seq
amino acids
peptide
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/344,445
Other languages
English (en)
Inventor
Mads Hald Andersen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
IO Biotech ApS
Original Assignee
IO Biotech ApS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by IO Biotech ApS filed Critical IO Biotech ApS
Assigned to IO BIOTECH APS reassignment IO BIOTECH APS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HERLEV HOSPITAL
Publication of US20200024324A1 publication Critical patent/US20200024324A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • the present invention relates to novel peptide compounds, such as fragments of PDL2, as well as compositions, uses, and kit-of-parts comprising these peptide compounds. Furthermore, the invention concerns nucleic acids, vectors, and host cells expressing said peptide compounds, for use in a method for treatment or prevention of a cancer, either alone or when administered simultaneously or sequentially with an additional cancer therapy.
  • Molecules of the B7-CD28 family play an important role in T-cell activation and tolerance. These pathways are not only responsible for providing positive costimulatory signals to sustain T-cell activity, but also contribute inhibitory signals that modulate the magnitude of T-cell responses. Inhibitory molecules of the B7/CD28 family play a key role in the induction of immune tolerance in the tumor microenvironment.
  • the programmed death-1 receptor (PD-1, CD279) with its ligands PD-L1 (CD274, B7-H1) and PD-L2 (CD273, B7-DC) constitutes one such inhibitory pathway.
  • the relevance of the PD-1/PD-L1 pathway in cancer has been extensively studied and therapeutic approaches targeting PD-1 (e.g.
  • nivolumab or pembrolizumab nivolumab or pembrolizumab
  • PD-L1 e.g. avelumab or atezolizumab
  • PD-L2 has not received as much attention and its role in modulating tumor immunity is less clear.
  • PD-L2 is an inhibitory molecule, expressed not only by antigen-presenting cells, but also by other immune cells and nonimmune cells in an inducible manner, mainly through Th2-associated cytokines.
  • the patterns of expression of PD-L1 and PD-L2 are quite distinct.
  • PD-L1 is constitutively expressed by a wide variety of immune cells and nonimmune cells and most normal tissue cells seem to be able to upregulate PD-L1.
  • PD-L2 expression was initially thought to be restricted to antigen-presenting cells such as macrophages and dendritic cells (DCs).
  • DCs dendritic cells
  • PD-L2 expression has been reported in patients with cancer.
  • PD-L2 may be expressed within human tumours in the absence of PD-L1. This could impact the understanding of the effectiveness of different targeted therapies to anti-PD-1 therapy even in the absence of PD-L1 expression.
  • PD-L2 expression has been described in several tumor types, including renal cell carcinoma, bladder carcinoma, melanoma, non-small-cell lung cancer (NSCLC), head and neck squamous carcinoma (HNSC), triple-negative breast cancer (TNBC), and gastric carcinoma.
  • PD-L2 may be expressed in individual tumor samples even in the absence of PD-L1 (http://www.esmo.org/Conferences/Past-Conferences/European-Cancer-Congress-2015/New s/Novel-Assay-Developed-to-Determine-PD-L2-Expression-in-Tumour-Samples).
  • Ohigashi et al. investigated the expression of PD-L1 and PD-L2 in human esophageal cancer to determine their clinical significance in patients' prognosis after surgery. Using RT-qPCR and immunohistochemistry, the authors showed that both PD-L1 and PD-L2 are expressed in frozen tissue samples of esophageal cancer patients and PD-L2-positive patients had a poorer prognosis than the negative patients, as was the case for PD-L1.
  • the present inventor has identified new immunogenic epitopes from extended PDL2 (human PDL2 including a signal sequence identified by SEQ ID NO 1 herein, reference NCBI accession Q9BQ51, version Q9BQ51.2).
  • extended PDL2 human PDL2 including a signal sequence identified by SEQ ID NO 1 herein, reference NCBI accession Q9BQ51, version Q9BQ51.2
  • Several PD-L2 derived peptides were selected based on binding affinity to the tissue type HLA-A2, of these a smaller selection was further analyzed by ELISPOT, since at least part of the sequences of these peptides are either in the signal peptide part of the PD-L2 sequence or in the transmembrane domain of the PD-L2 protein sequence.
  • PBMC peripheral blood mononuclear cells
  • the present invention concerns a peptide fragment of a human PDL2 protein of SEQ ID NO: 1, which fragment is up to 100 amino acids in length and wherein the peptide fragment comprises or consists of a consecutive sequence in a range from 8 to 100 amino acids of SEQ ID NO: 1; or a pharmaceutically acceptable salt thereof.
  • the present invention also concerns a peptide fragment of a human PDL2 protein of SEQ ID NO: 1, which fragment is up to 100 amino acids in length and wherein the peptide fragment consists of a consecutive sequence in a range from 8 to 100 amino acids of SEQ ID NO: 1; or a pharmaceutically acceptable salt thereof.
  • the peptide fragment is up to 60 amino acids in length, such as up to 50 amino acids, up to 40 amino acids, or up to 30 amino acids in length.
  • the corresponding upper range in the consecutive sequence is accordingly, 60, 50, 40 or 30 amino acids of SEQ ID NO: 1.
  • the peptide is up to 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acids in length.
  • the peptide fragment comprises or consists of a consecutive sequence in the range from 10 to 100 amino acids, such as from 10-17 amino acids, 20 to 30 amino acids, 30 to 40 amino acids, or from 40 to 50 amino acids.
  • the peptide fragment consists of a consecutive sequence in the range from 10 to 100 amino acids, such as from 10-17 amino acids, 20 to 30 amino acids, 30 to 40 amino acids, or from 40 to 50 amino acids. Typically from 10-17 amino acids or from 20 to 100 amino acids.
  • the peptide fragment does not comprise amino acid 1-3 of SEQ ID NO 1, that is MIF. Studies of PDL2 did not reveal any significant effect of these 3 amino acids, and they do not contribute to activation of T-cells.
  • the consecutive sequence comprises one or more sequences selected from any one of SEQ ID NO 2-12, such as any one of SEQ ID NO 2, 4, 11 and 12.
  • the consecutive sequence comprises both SEQ ID NO 2 and SEQ ID NO 4.
  • the peptide fragment SEQ ID NO: 12 comprises both SEQ ID NO 2 and SEQ ID NO 4.
  • the consecutive sequence comprises a sequence selected from SEQ ID NO 11, wherein the peptide fragment is up to 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acids in length.
  • the consecutive sequence consist of SEQ ID NO 11.
  • the consecutive sequence comprises a sequence selected from SEQ ID NO 12, wherein the peptide fragment is up to 25, 26, 27, 28, 29, or 30 amino acids in length.
  • the consecutive sequence comprises a sequence selected from SEQ ID NO 4, wherein the peptide fragment is up to 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acids in length.
  • the consecutive sequence comprises a sequence selected from SEQ ID NO 2, wherein the peptide fragment is up to 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acids in length.
  • the peptide fragment is capable of activating T-cells, such as CD4 and CD8 T-cells.
  • the activation is determined by an ELISPOT assay, such as the ELISPOT assay described herein.
  • the C terminal amino acid may be in the form of the acid or the amide, both are contemplated as individual embodiments of the peptide fragment of the present invention.
  • the peptide fragment is an isolated, immunogenic peptide fragment.
  • the present invention relates to a composition
  • a composition comprising the peptide fragment of the present invention, optionally together with a pharmaceutically acceptable additive, such as carrier or adjuvant.
  • such adjuvant is preferably selected from the group consisting of bacterial DNA based adjuvants, oil/surfactant based adjuvants, viral dsRNA based adjuvants, imidazochinilines, a Montanide ISA adjuvant.
  • the present invention relates to a peptide fragment of the present invention for use in the treatment of cancer, such as cancer characterized by expression of PDL2.
  • the present invention relates to a method of treating or preventing cancer in a patient, the method comprising administering to the cancer patient an effective amount of the peptide fragment of the present invention.
  • the method further comprises the simultaneous or sequential administration of an additional cancer therapy, such as a cytokine therapy, a T-cell therapy, an NK therapy, an immune system checkpoint inhibitor, chemotherapy, radiotherapy, immunostimulating substances, gene therapy, antibodies and dendritic cells.
  • an additional cancer therapy such as a cytokine therapy, a T-cell therapy, an NK therapy, an immune system checkpoint inhibitor, chemotherapy, radiotherapy, immunostimulating substances, gene therapy, antibodies and dendritic cells.
  • the additional cancer therapy is selected from one or more of Actimide, Azacitidine, Azathioprine, Bleomycin, Carboplatin, Capecitabine, Cisplatin, Chlorambucil, Cyclophosphamide, Cytarabine, Daunorubicin, Docetaxel, Doxifluridine, Doxorubicin, Epirubicin, Etoposide, Fludarabine, Fluorouracil, Gemcitabine, Hydroxyurea, Idarubicin, Irinotecan, Lenalidomide, Leucovorin, Mechlorethamine, Melphalan, Mercaptopurine, Methotrexate, Mitoxantrone, Nivolumab, Oxaliplatin, Paclitaxel, Pembrolizumab, Pemetrexed, Revlimid, Temozolomide, Teniposide, Thioguanine, Valrubicin, Vinblastine, Vincristine, Vindesine and Vinorelbine.
  • the present invention concerns a nucleic acid, such as DNA or RNA, encoding the peptide fragment of the present invention.
  • the present invention concerns a vector, such virus vector, comprising the nucleic acid of the present invention.
  • the present invention concerns a host cell, such as mammalian cell, comprising the vector of the present invention.
  • kit-of-parts comprising:
  • compositions are to be administered simultaneously or sequentially.
  • the present invention relates to a method of treating a clinical condition characterized by expression of PDL2 (SEQ ID NO 1), the method comprising administering to an individual suffering from said clinical condition an effective amount of the peptide compound or fragment of the present invention or the nucleic acid of the present invention or vector of the present invention or host cell of the present invention.
  • the present invention relates to a method of treating or preventing cancer in a patient, the method comprising administering to the cancer patient an effective amount of the peptide compound or fragment of the present invention or the nucleic acid of the present invention or vector of the present invention or host cell of the present invention.
  • the present invention relates to use of the peptide compound or fragment of the present invention or the nucleic acid of the present invention or vector of the present invention or host cell of the present invention for the manufacture of a medicament, such as an composition or vaccine, for the treatment or prevention of a cancer, such as cancer characterized by expression of PDL2.
  • the present invention relates to the peptide fragment of the present invention or the nucleic acid of the present invention or vector of the present invention or host cell of the present invention, for use in a method for treatment or prevention of a cancer, when administered simultaneously or sequentially with an additional cancer therapy, such as a cytokine therapy, a T-cell therapy, an NK therapy, an immune system checkpoint inhibitor, chemotherapy, radiotherapy, immunostimulating substances, gene therapy, antibodies and dendritic cells.
  • an additional cancer therapy such as a cytokine therapy, a T-cell therapy, an NK therapy, an immune system checkpoint inhibitor, chemotherapy, radiotherapy, immunostimulating substances, gene therapy, antibodies and dendritic cells.
  • the additional cancer therapy is selected from checkpoint blocking antibodies.
  • the additional cancer therapy is selected from one or more of Actimide, Azacitidine, Azathioprine, Bleomycin, Carboplatin, Capecitabine, Cisplatin, Chlorambucil, Cyclophosphamide, Cytarabine, Daunorubicin, Docetaxel, Doxifluridine, Doxorubicin, Epirubicin, Etoposide, Fludarabine, Fluorouracil, Gemcitabine, Hydroxyurea, Idarubicin, Irinotecan, Lenalidomide, Leucovorin, Mechlorethamine, Melphalan, Mercaptopurine, Methotrexate, Mitoxantrone, Nivolumab, Oxaliplatin, Paclitaxel, Pembrolizumab, Pemetrexed, Revlimid, Temozolomide, Teniposide, Thioguanine, Valrubicin, Vinblastine, Vincristine, Vindesine and Vinorelbine.
  • FIG. 1 Native T-cell responses to PD-L2.
  • A Examples of ELISPOT results for PBMCs isolated from patients with malignant melanoma (AA and MM), in response to PD-L201 (PD-L2(4-12); LLLMLSLEL) and PD-L205 (PD-L2(16-25); QIAALFTVTV).
  • B In-vitro IFN- ⁇ ELISPOT results. PBMCs from 9 patients with cancer were stimulated once in vitro with each peptide. Then, the PBMCs were exposed to the peptides, and IFN- ⁇ secretion was measured with ELISPOT. The response was calculated as the number of peptide-specific spots, minus the number of spots that reacted to an irrelevant peptide (HIV/HLA-A2; po1476-484; ILKEPVHGV), per 5 ⁇ 10 5 PBMCs.
  • FIG. 2 PD-L2 elicited responses in T cells from patients with cancer and T cells from healthy donors.
  • A Examples of IFN- ⁇ responses against PD-L201 (PD-L2(4-12)) and PD-L205 (PD-L2(16-25))(black bars) or irrelevant peptide (grey bars) in PBMCs from patients with malignant melanoma (AA and MM). All experiments were performed in triplicate, ** significant according to the DFR and DFRx2.
  • PBMCs were exposed to the peptides, and IFN- ⁇ secretion was measured with ELISPOT. The average number of peptide-specific spots (after subtracting the number of spots without added peptide) was calculated per 2-5 ⁇ 10 5 PBMCs.
  • D Ex vivo IFN- ⁇ ELISPOT results.
  • PD-L205 (PD-L2(16-25)) (black bars) or the irrelevant peptide (grey bars) elicited responses in PBMCs from two patients with malignant melanoma (AA) and in PBMCs from two healthy donors (HD).
  • FIG. 3 Reactivity towards long PD-L2 peptides spanning the signal peptide part of the PD-L2 sequence.
  • A In vitro IFN- ⁇ ELISPOT results. PBMCs from 11 patients with malignant melanoma and 11 healthy donors were stimulated with PD-L2long1 (PD-L2(9-29); SLELQLHQIAALFTVTVPKEL) or PD-L2long2 (PD-L2(1-25); MIFLLLMLSLELQLHQIAALFTVTV) and screened for IFN ⁇ responses, by measuring IFN ⁇ release in an in vitro ELISPOT assay.
  • B PBMCs from four non-hodgkin lymphoma patients (WM) screened for IFN ⁇ responses towards PD-L2long2 (PD-L2(1-25)) in an in vitro ELISPOT assay. All assays were made in triplicates with 3*10 ⁇ circumflex over ( ) ⁇ 6 cells per well, except one which were made in duplicates (WM-2). ** denotes as significant according to the DFR and DFRx2; * denotes significant according to only the DFR.
  • C Examples of ELISPOT well images for WM-5 patient in response to PD-L2long2.
  • TILs tumor infiltrating T-lymphocytes
  • FIG. 4 PD-L2-specific T cells are effector T cells.
  • A Intracellular cytokine staining showing CD4+ and CD8+ T cells that release TNF- ⁇ in response to either an irrelevant control peptide HIV peptide (HIV-1 po1476-484) or PD-L205 (PD-L2(16-25)) in cultures of PD-L2 T cells-A (left,) and PD-L2 T cells-B (right).
  • B Intracellular TNF- ⁇ and IFN- ⁇ cytokine staining of PD-L2 T-cells culture-A (left) and PD-L2 T-cells culture-B (right) in response to 5 hours stimulation with autologous DCs.
  • C IFN- ⁇ and TNF- ⁇ secretion by PD-L2 T-cell culture-A (top) and PD-L2 T-cell culture-B (bottom) towards PD-L205 (PD-L2(16-25)) peptide (black bars) and autologous DCs when cultured at ratio 1:5 (grey bars) as measured by ELISPOT assay.
  • D T2 cells pulsed either with PD-L205 (PD-L2(16-25)) or a control HIV peptide (HIV-1 po1476-484) as recognized by PD-L2 T-cell culture-A (left) and PD-L2 T-cell culture-B (right) in a standard 51Cr-release assay.
  • FIG. 5 PD-L2 dependent reactivity towards DCs.
  • FIG. 1 Flow cytometric analysis showing profile of PD-L2 surface expression on autologous DCs transfected with either PD-L2 siRNA or negative control siRNA, 48 hr after electroporation.
  • B PD-L2 T-cells culture-A (top) and PD-L2 T-cells culture-B (bottom) were stimulated with autologous DCs transfected PD-L2 siRNA or negative control siRNA for 5 hours at a ratio of 1:5 (DC:T-cell). Percentage of cytokine releasing CD4+ T cells (left) and CD8+ T cells (right) was measured using intracellular cytokine staining.
  • FIG. 6 No cross-reactivity between PD-L1-specific and PD-L2-specific T cells.
  • A The first 30 amino acid sequences of PD-L1 and PD-L2 and the location of the peptides PD-L101 (PDL1(15-23); LLNAFTVTV) and PD-L205 (PD-L2(16-25); QIAALFTVTV) in the signal peptide part of the proteins are marked in bold.
  • B In vitro IFN- ⁇ ELISPOT results show responses of T cells from five patients with cancer towards PD-L101 (PDL1(15-23)) and PD-L205 (PD-L2(16-25)) peptides.
  • (C) 51Cr-release assay results show percent lysis of T2 cells pulsed with PD-L101 (PDL1(15-23)), PD-L205 (PD-L2(16-25)), or an irrelevant HIV peptide (HIV-1 po1476-484) when exposed to PD-L101-specific T-cells (CTLs) at different effector-to-target ratios.
  • CTLs PD-L101-specific T-cells
  • Intracellular cytokine staining of cultured PD-L101-specific T-cells shows CD8+ T cell release of TNF- ⁇ , upon exposure to PD-L101 (PDL1(15-23)), PDL205 (PD-L2(16-25)), or an irrelevant HIV peptide (HIV-1 po1476-484).
  • D Percent lysis of T2-cells, pulsed with PDL205 (PD-L2(16-25)), PD-L101 peptide (PDL1(15-23)), or an irrelevant HIV peptide (HIV-1 po1476-484), after exposure to PD-L2 T-cell culture-A (left) or PD-L2 T-cell culture-B (right).
  • the present inventor investigated different PD-L2-derived epitopes that might elicit T cell reactivity, and tested spontaneous T-cell mediated reactivity against PD-L2 in samples from both healthy donors and patients with different cancers. Finally, it was determined whether PD-L2-specific T cells could recognize target cells expressing PD-L2.
  • Regulatory feedback mechanisms such as the upregulation of PD-L1 and PD-L2, are essential for limiting the strength and magnitude of immune responses that might otherwise harm the host.
  • immune evasion is detrimental in the framework of cancer immunotherapy.
  • the present invention concerns a peptide compound of PDL2 selected from:
  • “functional” means “capable of stimulating an immune response to the PDL2 having SEQ ID NO: 1”.
  • the present invention concerns a peptide fragment of a human PDL2 protein of SEQ ID NO: 1, which fragment is up to 100 amino acids in length and wherein the peptide fragment comprises or consists of a consecutive sequence in a range from 8 to 100 amino acids of SEQ ID NO: 1,
  • the peptide fragment comprises or consists of a consecutive sequence in the range from 10 to 100 amino acids, such as from 10-17 amino acids, 20 to 30 amino acids, 30 to 40 amino acids, or from 40 to 50 amino acids.
  • the peptide fragment consists of a consecutive sequence in the range from 10 to 100 amino acids, such as from 10 to 17 amino acids, 20 to 30 amino acids, 30 to 40 amino acids, or from 40 to 50 amino acids.
  • the peptide fragment is up to 60 amino acids in length, such as up to 50 amino acids, up to 40 amino acids, or up to 30 amino acids in length.
  • the corresponding upper range in the consecutive sequence is accordingly, 60, 50, 40 or 30 amino acids of SEQ ID NO: 1.
  • the peptide fragment is up to 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acids in length.
  • the consecutive sequence comprises one or more sequences selected from any one of SEQ ID NO 2, 4, 11 and 12. In a still further embodiment the consecutive sequence comprises the sequence SEQ ID NO 4. In a further embodiment the consecutive sequence consist of the sequence SEQ ID NO 4. In another embodiment the consecutive sequence comprises the sequence SEQ ID NO 11. In a further embodiment the consecutive sequence consist of the sequence SEQ ID NO 11. In another embodiment the consecutive sequence comprises the sequence SEQ ID NO 12. In a further embodiment the consecutive sequence consist of the sequence SEQ ID NO 12. In a further embodiment the consecutive sequence comprises both sequences SEQ ID NO 2 and SEQ ID NO 4.
  • the peptide fragment does not comprise amino acid 1-3 of SEQ ID NO 1, that is MIF.
  • the peptide fragment comprises or consist of LLLMLSLELQLHQIAALFTVTV (SEQ ID NO: 25)
  • the consecutive sequence comprises one or more sequences selected from any one of SEQ ID NO 2-12, such as any one of SEQ ID NO 2, 4, 11 and 12.
  • the consecutive sequence comprises both SEQ ID NO 2 and SEQ ID NO 4.
  • the peptide fragment SEQ ID NO: 12 comprises both SEQ ID NO 2 and SEQ ID NO 4.
  • the consecutive sequence comprises a sequence selected from SEQ ID NO 11, wherein the peptide fragment is up to 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acids in length. Typically, 21 amino acids in length.
  • the present invention concerns a peptide fragment of a human PDL2 protein of SEQ ID NO: 1, which peptide fragment is up to 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acids in length and wherein the peptide fragment comprises or consist of a sequence selected from SEQ ID NO 11; or a pharmaceutically acceptable salt thereof.
  • the consecutive sequence comprises a sequence selected from SEQ ID NO 12, wherein the peptide fragment is up to 25, 26, 27, 28, 29, or 30 amino acids in length.
  • the consecutive sequence comprises a sequence selected from SEQ ID NO 4, wherein the peptide fragment is up to 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acids in length.
  • the consecutive sequence comprises a sequence selected from SEQ ID NO 2, wherein the peptide fragment is up to 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acids in length.
  • the peptide compound is selected from b) a functional homologue having at least 70%, 80%, 90%, or 95% identity to SEQ ID NO 1 or the peptide fragment of a), wherein the C-terminal amino acid also comprises the amide; or a pharmaceutically acceptable salt thereof.
  • the functional homologue has at least 80% identity to SEQ ID NO 1.
  • the functional homologue has at least 90% identity to SEQ ID NO 1.
  • the functional homologue has at least 95% identity to SEQ ID NO 1.
  • the functional homologue has at least 70% identity to the peptide fragment of a).
  • the functional homologue has at least 80% identity to the peptide fragment of a).
  • the functional homologue has at least 90% identity to the peptide fragment of a).
  • the functional homologue has at least 95% identity to the peptide fragment of a).
  • the peptide compound is selected from c) a functional analogue wherein at least one amino acid has been deleted, inserted and/or substituted in SEQ ID NO 1 or the peptide fragment of a), wherein the C-terminal amino acid also comprises the amide; or a pharmaceutically acceptable salt thereof.
  • the peptide compound is selected from a) a peptide fragment of SEQ ID NO 1 consisting of a consecutive sequence of from 8 to 272 amino acids, wherein the C-terminal amino acid also comprises the amide; or a pharmaceutically acceptable salt thereof.
  • the peptide fragment consists of a consecutive sequence in the range of from 8 to 250 amino acids.
  • the peptide fragment consists of a consecutive sequence in the range of from 8 to 200 amino acids.
  • the peptide fragment consists of a consecutive sequence in the range of from 8 to 150 amino acids.
  • the peptide fragment consists of a consecutive sequence in the range of from 8 to 120 amino acids.
  • the peptide fragment consists of a consecutive sequence in the range of from 10 to 100 amino acids. In a still further embodiment, the peptide fragment consists of a consecutive sequence in the range of from 20 to 80 amino acids. In a further embodiment, the peptide fragment consists of a consecutive sequence in the range of from 30 to 60 amino acids. In a still further embodiment, the peptide fragment consists of a consecutive sequence in the range of from 40 to 50 amino acids. In a further embodiment, the peptide fragment consists of a consecutive sequence in the range of from 8 to 30 amino acids. In a still further embodiment, the peptide fragment consists of a consecutive sequence in the range of from 10 to 25 amino acids. In a further embodiment, the peptide fragment consists of a consecutive sequence in the range of from 8 to 25 amino acids.
  • the peptide fragment of SEQ ID NO 1 is selected from the group consisting of PDL2(1-25), PDL2(1-50), PDL2(1-150), PDL2(1-200), PDL2(50-100), PDL2(50-150), PDL2(50-200), PDL2(60-100), PDL2(60-150), PDL2(60-200), PDL2(70-100), PDL2(70-150), PDL2(70-200), PDL2(200-273), and PDL2(210-250).
  • the peptide fragment of SEQ ID NO 1 is selected from PDL2(1-25).
  • the peptide fragment of SEQ ID NO 1 is selected from PDL2(1-50). In a further preferred embodiment, the peptide fragment of SEQ ID NO 1 is selected from PDL2(200-273). In a preferred embodiment, the peptide fragment of SEQ ID NO 1 is selected from PDL2(210-250).
  • the consecutive sequence comprises one or more sequences selected from any one of SEQ ID NO 2-24, such as one sequence selected from SEQ ID NO 7 or two sequence selected from SEQ ID NO 2 and 4. In a preferred embodiment, the consecutive sequence is selected from SEQ ID NO 4. In a preferred embodiment, the consecutive sequence is selected from SEQ ID NO 2. In a preferred embodiment, the consecutive sequence is selected from SEQ ID NO 7.
  • the peptide fragment consists of a consecutive sequence in the range of from 8 to 120, it may at the same time be selected within the sequence of for instance PDL2(1-150), whereas a peptide fragment consisting of a consecutive sequence in the range of from 8 to 272, cannot be at the same time be selected within the sequence of for instance PDL2(1-150), this is known to the person skilled in the art. Otherwise all combinations are contemplated within the present invention.
  • PDL2(x-y), wherein x and y are integers selected from 1-273 as used herein means a peptide fragment of human extended PDL2 having the SEQ ID NO 1 as defined herein, wherein x is the N-terminal amino acid and y is the C-terminal amino acid, for instance PDL2(16-25) indicates the peptide fragment from amino acid 16 of SEQ ID NO 1 to amino acid 25 of SEQ ID NO 1 wherein amino acid 16 is Q and amino acid 25 is V.
  • the C terminal amino acid may optionally be replaced with the corresponding amide, to improve solubility and/or to aid with manufacture/isolation.
  • the polypeptide may have attached at the N and/or C terminus at least one additional moiety to improve solubility and/or to aid with manufacture/isolation.
  • Suitable moieties include hydrophilic amino acids.
  • the amino acids KR may be added at the N terminus and/or the amino acids RK may be added in order at the C terminus.
  • the present invention relates to a composition
  • a composition comprising a peptide fragment of a human PDL2 protein of SEQ ID NO: 1, which fragment is up to 100 amino acids in length and wherein the peptide fragment comprises or consists of a consecutive sequence in a range from 8 to 100 amino acids of SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof; optionally together with a pharmaceutically acceptable additive, such as a carrier or adjuvant.
  • identity refers to a relationship between the sequences of two or more peptides, such as polypeptides, as determined by comparing the sequences.
  • identity also means the degree of sequence relatedness between proteins or polypeptides, as determined by the number of matches between strings of two or more amino acid residues. “Identity” measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., “algorithms”). Identity of related proteins or peptides can be readily calculated by known methods. Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A.
  • Preferred methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity are described in publicly available computer programs. Preferred computer program methods to determine identity between two sequences include the GCG program package, including GAP (Devereux et al., Nucl. Acid. Res., 12, 387, (1984); Genetics Computer Group, University of Wisconsin, Madison, Wis.), BLASTP, BLASTN, and FASTA (Altschul et al., J. Mol. Biol., 215, 403-410, (1990)). The BLASTX program is publicly available from the National Center for Biotechnology Information (NCBI) and other sources (BLAST Manual, Altschul et al. NCB/NLM/NIH Bethesda, Md. 20894; Altschul et al., supra). The well-known Smith Waterman algorithm may also be used to determine identity.
  • NCBI National Center for Biotechnology Information
  • GAP Genetics Computer Group, University of Wisconsin, Madison, Wis.
  • two proteins for which the percent sequence identity is to be determined are aligned for optimal matching of their respective amino acids (the “matched span”, as determined by the algorithm).
  • a gap opening penalty (which is calculated as 3 times the average diagonal; the “average diagonal” is the average of the diagonal of the comparison matrix being used; the “diagonal” is the score or number assigned to each perfect amino acid match by the particular comparison matrix) and a gap extension penalty (which is usually 1/10 times the gap opening penalty), as well as a comparison matrix such as PAM 250 or BLOSUM 62 are used in conjunction with the algorithm.
  • a standard comparison matrix see Dayhoff et al., Atlas of Protein Sequence and Structure, vol.
  • the peptide fragment under a), the functional homologue under b), or the functional analogue under c) is capable of activating T-cells, such as CD4 and CD8 T-cells.
  • T-cells such as CD4 and CD8 T-cells.
  • the activation is determined by the ELISPOT assay described herein.
  • the peptide fragment under a) is capable of activating T-cells as determined by the ELISPOT assay described herein.
  • the present invention relates to a nucleic acid encoding the peptide compound of the present invention.
  • the peptide compound of the present invention is selected from any one of the above embodiments.
  • the nucleic acid is selected from the group consisting of DNA and RNA.
  • the present invention relates to a vector comprising the nucleic acid of the present invention.
  • the nucleic acid of the present invention is selected from any one of the above embodiments, and the peptide compound of the present invention is selected from any one of the above embodiments.
  • the vector is selected from a virus vector.
  • the present invention relates to a host cell comprising the vector of the present invention.
  • the vector of the present invention is selected from any one of the above embodiments
  • the nucleic acid of the present invention is selected from any one of the above embodiments
  • the peptide compound of the present invention is selected from any one of the above embodiments.
  • the host cell is selected from a mammalian cell.
  • the present invention relates to a composition
  • a composition comprising the peptide compound or fragment of the present invention or the nucleic acid of the present invention or the vector of the present invention or the host cell of the present invention, optionally together with a pharmaceutically acceptable additive, such as carrier or adjuvant.
  • the present invention relates to a composition
  • a composition comprising
  • the composition of the present invention is for use in a method for treatment or prevention of a disease, disorder or condition selected from cancer.
  • the cancer is a tumor forming cancer disease.
  • the cancer is selected from any one of melanoma, renal cell carcinoma, non-hodgkin lymphoma, and ovarian cancer.
  • the adjuvant is selected from the group consisting of bacterial DNA based adjuvants, oil/surfactant based adjuvants, viral dsRNA based adjuvants, imidazochinilines, and a Montanide ISA adjuvant.
  • kit-of-parts comprising;
  • composition of the present invention a) the composition of the present invention, and b) a composition comprising at least one second active ingredient, selected from an immunostimulating compound, such as an interleukin, e.g. IL-2 and or IL-21, an anti-cancer agent, such as a chemotherapeutic agent, e.g.
  • an immunostimulating compound such as an interleukin, e.g. IL-2 and or IL-21
  • an anti-cancer agent such as a chemotherapeutic agent, e.g.
  • Actimide Azacitidine, Azathio-prine, Bleomycin, Carboplatin, Capecitabine, Cisplatin, Chlorambucil, Cyclophospha-mide, Cytarabine, Daunorubicin, Docetaxel, Doxifluridine, Doxorubicin, Epirubicin, Etoposide, Fludarabine, Fluorouracil, Gemcitabine, Hydroxyurea, Idarubicin, Irinotecan, Lenalidomide, Leucovorin, Mechlorethamine, Melphalan, Mercaptopurine, Metho-trexate, Mitoxantrone, nivolumab, Oxaliplatin, Paclitaxel, pembrolizumab, Pemetrexed, Revlimid, Temozolomide, Teniposide, Thioguanine, Valrubicin, Vinblastine, Vincristine, Vindesine and Vinorelbine.
  • kits-of-parts the provided compositions are to be administered simultaneously or sequentially.
  • the present invention relates to a method of treating a clinical condition characterized by expression of PDL2 of SEQ ID NO 1, the method comprising administering to an individual suffering from said clinical condition an effective amount of the peptide compound of the present invention or the nucleic acid of the present invention or the vector of the present invention or the host cell of the present invention.
  • the present invention relates to use of the peptide compound of the present invention, or the nucleic acid of the present invention, or the vector of the present invention, or the host cell of the present invention, for the manufacture of a medicament, such as an composition or vaccine, for the treatment or prevention of a cancer characterized by expression of PDL2.
  • the present invention relates to the peptide compound of the present invention, or the nucleic acid of the present invention, or the vector of the present invention, or the host cell of the present invention, for use in a method for treatment or prevention of a cancer, when administered simultaneously or sequentially with an additional cancer therapy.
  • the additional cancer therapy is selected from the group consisting of a cytokine therapy, a T-cell therapy, an NK therapy, an immune system checkpoint inhibitor, chemotherapy, radiotherapy, immunostimulating substances, gene therapy, anti-bodies and dendritic cells.
  • the additional cancer therapy is selected from an immune system checkpoint inhibitor.
  • the immune system checkpoint inhibitor is a checkpoint blocking antibody.
  • the additional cancer therapy is selected from the group consisting of Actimide, Azacitidine, Azathioprine, Bleomycin, Carboplatin, Capecitabine, Cisplatin, Chlorambucil, Cyclophosphamide, Cytarabine, Daunorubicin, Docetaxel, Doxifluridine, Doxorubicin, Epirubicin, Etoposide, Fludarabine, Fluorouracil, Gemcitabine, Hydroxyurea, Idarubicin, Irinotecan, Lenalidomide, Leucovorin, Mechlorethamine, Melphalan, Mercaptopurine, Methotrexate, Mitoxantrone, Nivolumab, Oxaliplatin, Paclitaxel, Pembrolizumab, Pemetrexed, Revlimid, Temozolomide, Teniposide, Thioguanine, Valrubicin, Vinblastine, Vincristine, Vindesine and Vinorelbine
  • any amino acid sequence shown may be modified at the C-terminal amino acid to be on amide form (—CONH 2 ) or may be on acid form (—COOH), thus any one of these are preferred embodiments, and it is intended that any C-terminal amino acid, such as I, L, V, comprises both amide and acid form unless specified by —NH 2 or —OH.
  • the PDL2 peptide fragments disclosed herein are made by standard peptide synthesis, such as solid-phase peptide synthesis (SPPS).
  • SPPS is a standard method for synthesizing peptides in the lab. SPPS allows for the synthesis of natural peptides which are difficult to express in bacteria, the incorporation of unnatural amino acids, peptide/protein backbone modification, and the synthesis of D-proteins, which consist of D-amino acids.
  • Small porous beads are treated with functional units (‘linkers’) on which peptide chains can be built. The peptide will remain covalently attached to the bead until cleaved from it by a reagent such as anhydrous hydrogen fluoride or trifluoroacetic acid.
  • the peptide is thus ‘immobilized’ on the solid-phase and can be retained during a filtration process while liquid-phase reagents and by-products of synthesis are flushed away.
  • the general principle of SPPS is one of repeated cycles of deprotection-washcoupling-wash.
  • the free N-terminal amine of a solid-phase attached peptide is coupled to a single N-protected amino acid unit. This unit is then deprotected, revealing a new N-terminal amine to which a further amino acid may be attached.
  • the superiority of this technique partially lies in the ability to perform wash cycles after each reaction, removing excess reagent with all of the growing peptide of interest remaining covalently attached to the insoluble resin.
  • a therapeutically effective amount of at least one compound is administered to a mammal in need of said treatment.
  • amino acids are identified by the one or three letter code known to the person skilled in the art and shown in the table below for convenience:
  • Amino acids, one and three letter codes Amino acid Three letter code
  • alanine ala A arginine arg R asparagine asn N aspartic acid asp D asparagine or aspartic acid asx
  • cysteine cys C glutamic acid glu E glutamine gln Q glutamine or glutamic acid glx Z glycine gly G histidine
  • proline pro P serine ser S threonine thr T tryptophan trp W tyrosine tyr Y valine val
  • immunogenic means that a peptide fragment is capable of eliciting an immune response, preferably a T-cell response, in at least one individual after administration to said individual.
  • a polypeptide may be identified as immunogenic using any suitable method, including in vitro methods. For example, a peptide may be identified as immunogenic if it has at least one of the following characteristics:
  • treatment means the management and care of a patient for the purpose of combating a condition, such as a disease or a disorder.
  • the term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound to alleviate the symptoms or complications, to delay the progression of the disease, disorder or condition, to alleviate or relief the symptoms and complications, and/or to cure or eliminate the disease, disorder or condition as well as to prevent the condition, wherein prevention is to be understood as the management and care of a patient for the purpose of combating the disease, condition, or disorder and includes the administration of the active compounds to prevent the onset of the symptoms or complications.
  • the treatment may either be performed in an acute or in a chronic way.
  • the patient to be treated is preferably a mammal; in particular a human being, but it may also include animals, such as dogs, cats, cows, monkeys, apes, sheep and pigs.
  • a therapeutically effective amount of a peptide compound of the present invention or a peptide fragment disclosed herein, as used herein means an amount sufficient to cure, alleviate or partially arrest the clinical manifestations of a given disease and its complications. An amount adequate to accomplish this is defined as “therapeutically effective amount”. Effective amounts for each purpose will depend on the severity of the disease or injury as well as the weight and general state of the subject. It will be understood that determining an appropriate dosage may be achieved using routine experimentation, by constructing a matrix of values and testing different points in the matrix, which is all within the ordinary skills of a trained physician or veterinary.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the peptide compound, such as peptide fragment, of the present invention and optionally a pharmaceutically acceptable additive, such as a carrier or an excipient.
  • pharmaceutically acceptable additive is intended without limitation to include carriers, excipients, diluents, adjuvant, colorings, aroma, preservatives etc. that the skilled person would consider using when formulating a compound of the present invention in order to make a pharmaceutical composition.
  • the adjuvants, diluents, excipients and/or carriers that may be used in the composition of the invention must be pharmaceutically acceptable in the sense of being compatible with the peptide compound, peptide fragment, nucleic acid, vector, or host cell and the other ingredients of the pharmaceutical composition, and not deleterious to the recipient thereof. It is preferred that the compositions shall not contain any material that may cause an adverse reaction, such as an allergic reaction.
  • the adjuvants, diluents, excipients and carriers that may be used in the pharmaceutical composition of the invention are well known to a person within the art.
  • Adjuvants are any substance whose admixture into the composition increases or otherwise modifies the immune response elicited by the composition.
  • Adjuvants broadly defined, are substances which promote immune responses. Adjuvants may also preferably have a depot effect, in that they also result in a slow and sustained release of an active agent from the administration site. A general discussion of adjuvants is provided in Goding, Monoclonal Antibodies: Principles & Practice (2nd edition, 1986) at pages 61-63.
  • Adjuvants may be selected from the group consisting of: AlK(SO4)2, AlNa(SO4)2, AlNH4 (SO4), silica, alum, Al(OH)3, Ca3 (PO4)2, kaolin, carbon, aluminum hydroxide, muramyl dipeptides, N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-DMP), N-acetyl-nornuramyl-L-alanyl-D-isoglutamine (CGP 11687, also referred to as nor-MDP), N-acetylmuramyul-L-alanyl-D-isoglutaminyl-L-alanine-2-(1′2′-dipalmitoyl-sn-glycero-3-hydroxphosphoryloxy)-ethylamine (CGP 19835A, also referred to as MTP-PE), RIBI (MPL+TDM+CWS) in a
  • GM-CSF Granulocyte-macrophage colony stimulating factor
  • Preferred adjuvants to be used with the invention include oil/surfactant based adjuvants such as Montanide adjuvants (available from Seppic, Belgium), preferably Montanide ISA-51.
  • Other preferred adjuvants are bacterial DNA based adjuvants, such as adjuvants including CpG oligonucleotide sequences.
  • Yet other preferred adjuvants are viral dsRNA based adjuvants, such as poly I:C. GM-CSF and Imidazochinilines are also examples of preferred adjuvants.
  • the adjuvant is most preferably a Montanide ISA adjuvant.
  • the Montanide ISA adjuvant is preferably Montanide ISA 51 or Montanide ISA 720.
  • a peptide compound, peptide fragment, nucleic acid, vector, or host cell of an composition of the invention may be coupled to a carrier.
  • a carrier may be present independently of an adjuvant.
  • the function of a carrier can be, for example, to increase the molecular weight of the peptide compound, peptide fragment, nucleic acid, vector, or host cell in order to increase activity or immunogenicity, to confer stability, to increase the biological activity, or to increase serum half-life.
  • a carrier may aid in presenting the polypeptide or fragment thereof to T-cells.
  • the polypeptide or fragment thereof may be associated with a carrier, such as those set out below.
  • the carrier may be any suitable carrier known to a person skilled in the art, for example a protein or an antigen presenting cell, such as a dendritic cell (DC).
  • Carrier proteins include 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.
  • the carrier protein may be tetanus toxoid or diphtheria toxoid.
  • the carrier may be a dextran such as sepharose. The carrier must be physiologically acceptable to humans and safe.
  • the composition may optionally comprise a pharmaceutically acceptable excipient.
  • the excipient must be ‘acceptable’ in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof.
  • Auxiliary substances such as wetting or emulsifying agents, pH buffering substances and the like, may be present in the excipient.
  • These excipients and auxiliary substances are generally pharmaceutical agents that do not induce an immune response in the individual receiving the composition, and which may be administered without undue toxicity.
  • Pharmaceutically acceptable excipients include, but are not limited to, liquids such as water, saline, polyethyleneglycol, hyaluronic acid, glycerol and ethanol.
  • Pharmaceutically acceptable salts can also be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like
  • organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • compositions may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration.
  • injectable compositions may be prepared, packaged, or sold in unit dosage form, such as in ampoules or in multi-dose containers containing a preservative.
  • Compositions include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and implantable sustained-release or biodegradable formulations.
  • the active ingredient is provided in dry (for e.g., a powder or granules) form for reconstitution with a suitable vehicle (e. g., sterile pyrogen-free water) prior to administration of the reconstituted composition.
  • a suitable vehicle e. g., sterile pyrogen-free water
  • the composition may be prepared, packaged, or sold in the form of a sterile injectable aqueous or oily suspension or solution.
  • This suspension or solution may be formulated according to the known art, and may comprise, in addition to the active ingredient, additional ingredients such as the adjuvants, excipients and auxiliary substances described herein.
  • Such sterile injectable formulations may be prepared using a non-toxic parenterally-acceptable diluent or solvent, such as water or 1,3-butane diol, for example.
  • Other acceptable diluents and solvents include, but are not limited to, Ringer's solution, isotonic sodium chloride solution, and fixed oils such as synthetic mono- or di-glycerides.
  • compositions which are useful include those which comprise the active ingredient in microcrystalline form, in a liposomal preparation, or as a component of a biodegradable polymer systems.
  • Compositions for sustained release or implantation may comprise pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt.
  • the active ingredients of the composition may be encapsulated, adsorbed to, or associated with, particulate carriers. Suitable particulate carriers include those derived from polymethyl methacrylate polymers, as well as PLG microparticles derived from poly(lactides) and poly(lactide-co-glycolides).
  • particulate systems and polymers can also be used, for example, polymers such as polylysine, polyarginine, polyornithine, spermine, spermidine, as well as conjugates of these molecules.
  • compositions and particularly immunotherapetic compositions as herein disclosed may, in addition to the compounds herein disclosed, further comprise at least one pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier.
  • the pharmaceutical compositions comprise from 1 to 99 weight % of said at least one pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier and from 1 to 99 weight % of a compound as herein disclosed.
  • the combined amount of the active ingredient and of the pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier may not constitute more than 100% by weight of the composition, particularly the pharmaceutical composition.
  • only one compound or peptide as herein disclosed is used for the purposes discussed above.
  • two or more of the compound as herein disclosed are used in combination for the purposes discussed above.
  • composition particularly immunotherapeutic composition comprising a compound set forth herein may be adapted for oral, intravenous, topical, intraperitoneal, nasal, buccal, sublingual, or subcutaneous administration, or for administration via the respiratory tract in the form of, for example, an aerosol or an air-suspended fine powder. Therefore, the pharmaceutical composition may be in the form of, for example, tablets, capsules, powders, nanoparticles, crystals, amorphous substances, solutions, transdermal patches or suppositories.
  • PBMCs Peripheral blood mononuclear cells
  • LymphoprepTM Alere AS, cat. 1114547
  • HLA-typed HLA-typed
  • DMSO 10% DMSO
  • TIL Tumor Infiltrating lymphocytes
  • HLA-A2 restricted, 9-10 amino acid-long peptides in the human PD-L2 protein with an online epitope prediction database, SYFPEITHI (www.syfpeithi.de).
  • SYFPEITHI online epitope prediction database
  • peptides were: PD-L201 (PD-L2(4-12); LLLMLSLEL), PD-L204 (PD-L2(231-240); IIAFIFIATV), PD-L205 (PD-L2(16-25); QIAALFTVTV), PD-L208 (PD-L2(6-14); LMLSLELQL), PD-L209 (PD-L2(9-17); SLELQLHQI), PD-L215 (PD-L2(234-243); FIFIATVIAL), PD-L216 (PD-L2(11-20); ELQLHQIAAL), PD-L220 (PD-L2(1-10); MIFLLLMLSL), and PD-L222 (PD-L2(226-235); FIPFCIIAFI).
  • PD-L2long1 (PD-L2(9-29); SLELQLHQIAALFTVTVPKEL) and PD-L2long2 (PD-L2(1-25); MIFLLLMLSLELQLHQIAALFTVTV).
  • the HLA-A2 high affinity binding epitope, HIV-1 po1476-484 (ILKEPVHGV; SEQ ID NO 26) was used as an irrelevant control.
  • a previously described PD-L1 peptide named as PD-L101 (PDL1(15-23); LLNAFTVTV; SEQ ID NO 27) was used in cross reactivity experiments (1). All peptides were dissolved in either DMSO or sterile water before the experiments.
  • the IFN- ⁇ and TNF- ⁇ ELISPOT technique was performed as described previously (3). We performed the assays according to the guidelines provided by the cancer immunotherapy immunoguiding program (CIP; http://cimt.eu/cimt/files/dl/cip_guidelines.pdf). Unless stated otherwise, PBMCs were stimulated once in vitro with peptide prior to analysis to extend the sensitivity of the assay. To measure T-cell reactivity, nitrocellulose-bottomed 96-well plates (MultiScreen MSIPN4W; Millipore) were coated overnight at room temperature or two days at 4° C. with the relevant antibodies. The wells were washed and blocked with X-vivo medium for 2 h.
  • CIP cancer immunotherapy immunoguiding program
  • the PBMCs were added at different cell concentrations in triplicate wells, with PD-L2 peptide or with control peptide, and incubated overnight. The following day, the wells were washed, and the relevant biotinylated secondary antibody (Mabtech) was added, followed by the avidin-enzyme conjugate (AP-Avidin; Calbiochem/Invitrogen Life Technologies); finally, we added the enzyme substrate, NBT/BCIP (Invitrogen Life Technologies) for visualization. The spots on the developed ELISPOT plates were analyzed on a CTL ImmunoSpot S6 Ultimate-V analyzer with Immunospot software, v5.1.
  • PBMCs from a patient with melanoma were stimulated with irradiated (30 Gy) autolognus DCs, which had been pulsed with PD-L205 (PD-L2(16-25)) peptide (PBL:DC ratio 10:1) and IL-7 (40 U/ml) were added (PeproTech, London, UK). The next day, IL-12 (20 U/ml) was added (PeproTech, London, UK). At weekly intervals, we performed three identical stimulations, with irradiated autologous DCs loaded with PDL205 (PD-L2(16-25)), and IL-12 (20 U/ml) added the next day.
  • the culture was enriched for specific T cells, either by staining with an anti-CD137-PE antibody (BD-Biosciences) or by employing a TNF- ⁇ -enrichment and detection kit (according to the procedure by Miltenyi Biotec).
  • an anti-CD137-PE antibody BD-Biosciences
  • TNF- ⁇ -enrichment and detection kit accordinging to the procedure by Miltenyi Biotec.
  • the sorted cells were rapidly expanded by incubating with 0.6 ⁇ g anti-CD3 antibody (eBioscience, clone OKT3) and a high dose of IL-2 (Proleukin, Novartis).
  • Cells were washed, fixed, and permeabilized with Fixation/Permeabilization and Permeabilization Buffer (eBioscience), according to the manufacturer's instructions. Cells were subsequently stained with fluorochrome-conjugated antibodies to visualize intracellular cytokines. The following antibody-fluorochrome combinations were used: IFN ⁇ -PE-CY7/APC (eBioscience) and TNF ⁇ -APC/BV421 (eBioscience). Relevant isotype controls were used to enable correct compensation and confirm antibody specificity. Stained cells were analyzed with a BD FACSCanto II flow cytometer. Analysis was performed with BD FACSDiva Software.
  • Target cells were TAP deficient T2 cells, pulsed with HIV-1 po1476-484 (ILKEPVHGV), PD-L101 (PD-L1(15-23)), or PD-L205 (PD-L2(16-25)).
  • the PD-L2 siRNA duplexes consisted of three transcripts: 1. (sence) 5′-CAUCCUAAAGGUUCCAGAAtt-3′ (SEQ ID NO 28), (antisense) 5′-UU CUGGAACCUUUAGGAUGtg3′ (SEQ ID NO 29)—(siRNA ID # s37285), 2. (sense) 5′-CCUAAGGAACUGUACAUAAtt-3′ (SEQ ID NO 30), (antisense) 5′-UUAUGUACAGUUCCUUAGGga 3′ (SEQ ID NO 31)—(siRNA ID # s37286) and 3.
  • siRNA duplexes were dissolved in RNase-free water to a final concentration of 100 ⁇ M and subsequently stored at ⁇ 80° C.
  • Cells were kept on ice and added with 0.25 nmol of each PD-L2 siRNA duplexes. Subsequently, cells were transferred into a 2 mm kuvette and were electroporated with a single pulse at 250 Volts for 2 milli seconds using a BTX 830 square-wave electroporator (Harvard Apparatus, Holliston Mass., USA).
  • monocytes were transferred to prewarmed CellGro medium containing DC-maturation cocktail: IL-0(1,000 U/mL), IL-6 (1,000 U/mL), TNF- ⁇ (1,000 U/mL), and PGE2 (1 mg/mL) (all from PeproTech).
  • DC-maturation cocktail IL-0(1,000 U/mL)
  • IL-6 1,000 U/mL
  • TNF- ⁇ 1,000 U/mL
  • PGE2 (1 mg/mL) (all from PeproTech).
  • the transfected matured DCs were used for experimental analysis.
  • PD-L2 surface expression on the DCs transfected with PD-L2 siRNA and negative control siRNA was analyzed using anti-human PD-L2-PE (BD biosciences). Functionality of PD-L2-specific T-cell cultures towards transfected autologous DCs was analysed using ICS and ELISPOT assay as described above.
  • T-cell cultures were stimulated with either PD-L2 siRNA or negative control transfected DCs for 5 hours with ratio of 1:5.
  • EliSpot assay T-cell cultures were stimulated with the DCs for 24 hours with ratio of 1:5.
  • ELISPOT response was defined, based on the guidelines and recommendations provided by CIP and Moodie et al (5).
  • the non-parametric distribution-free resampling (DFR) and more conservative DFRx2 statistical test were used for a formal comparison between antigen-stimulated wells and negative-control wells.
  • the ELISPOT assays were performed at least in triplicate.
  • the amino acid sequence of the PD-L2 protein was screened with the “SYFPEITHI” database (15) to predict the best HLA-A2 peptide epitopes.
  • the algorithm identified 22 peptides that were top candidates, based on predictive scores in the range of 22-29.
  • These nine peptides were used with the IFN- ⁇ ELISPOT in vitro assay to test for the presence of specific T-cell responses in PBMCs from different HLA-A2+ patients with cancer.
  • FIGS. 2A and 2B we utilized both the IFN- ⁇ and TNF- ⁇ ELISPOT assays to examine 5 selected PBMCs for immune responses against PD-L201 (PD-L2(4-12)) and PD-L205 (PD-L2(16-25)) ( FIGS. 2A and 2B ). All IFN- ⁇ and TNF- ⁇ responses were statistically significant, according to the DFR test ( FIGS. 2A and 2B ). In addition, the IFN- ⁇ responses and one TNF- ⁇ response were statistically significant according to the DFRx2 rule ( FIGS. 2A and 2B ). Next, we tested PBMCs from healthy donors for immune responses against both PD-L2-derived epitopes with the IFN- ⁇ ELISPOT assay.
  • FIG. 2C We detected strong immune responses against PD-L205 (PD-L2(16-25)), and weaker responses against PD-L201 (PD-L2(4-12)) in healthy individuals ( FIG. 2C ). In general, PD-L205 (PD-L2(16-25)) appeared to be the dominant epitope for eliciting immune responses.
  • PBMCs from four donors, directly ex vivo (without prior in vitro peptide stimulation), for responses against PD-L205 (PD-L2(16-25)) with the IFN- ⁇ ELISPOT assay FIG. 2D ). The PBMCs from one of these donors showed an ex vivo IFN- ⁇ response that was statistically significant, according to the DFR rule ( FIG. 2D ).
  • PD-L2long1 (PD-L2(9-29); SLELQLHQIAALFTVTVPKEL), included the PD-L205 (PD-L2(16-25)) epitope; and the other, PD-L2long2 (PD-L2(1-25); MIFLLLMLSLELQLHQIAALFTVTV), included both the PD-L201 (PD-L2(4-12)) and PD-L205 (PD-L2(16-25)) epitopes.
  • Tumor infiltrating T-lymphocytes from two melanoma patients also elicited IFN- ⁇ CD4+ T-cell responses towards PD-L2long2 (PD-L2(1-25)) measured by using intracellular cytokine staining ( FIG. 3D ).
  • PD-L2-Specific T Cells were Effector Cells Releasing Pro-Inflammatory Cytokines
  • PD-L205-specific T cells To characterize the immune response elicited by PD-L2, we isolated PD-L205-specific T cells. Briefly, PBMCs isolated from a patient with melanoma (AA26) were expanded in vitro. Then, PBMCs were stimulated with irradiated autologous DCs that had been pulsed with PD-L205. Specific T cells were isolated either by staining with anti-CD137 antibody (PD-L2 T cell culture-A) or with the TNF- ⁇ enrichment method (PD-L2 T cell culture-B). These specific T cells were cultured and expanded with high dose IL-2, then analyzed for specificity against PD-L205 (PD-L2(16-25)) with intracellular cytokine staining ( FIGS.
  • FIG. 4A and 4B ELISPOT ( FIG. 4C ) and cytotoxicity assays ( FIG. 4D ).
  • TNF- ⁇ release in response to PD-L205(PD-L2(16-25)) in about 4.5% and 1% of CD4+ and CD8+ T cells, respectively, from PD-L2 T cell culture-A; and about 7% and 2% of CD4+ and CD8+ T cells, respectively, from PD-L2 T cell culture-B ( FIG. 4A ).
  • TNF- ⁇ release in response to autologous DCs in around 3% and 2.8% of CD4+ T cells from PD-L2 T cell culture-A and -B respectively FIG. 4B ).
  • IFN-Y and TNF- ⁇ T cell responses were observed towards PD-L205 (PD-L2(16-25)) peptide and autologous DCs in both PD-L2 specific cultures ( FIG. 4C ).
  • Both expanded cultures also recognized and lysed T2 cells that had been pulsed with PDL205 (PD-L2(16-25)) in conventional 51 chromium-release assays, but they did not recognize T2 cells pulsed with an irrelevant HIV peptide (HIV-1 po1476-484) ( FIG. 4D ).
  • PD-L2 can be induced in immune cells.
  • the dominant PD-L2 epitope in eliciting a T cell response was PD-L205 (PD-L2(16-25); QIAALFTVTV).
  • PD-L1 PD-L101 (PDL1(15-23); LLNAFTVTV) (1), (2), (6), which elicited a strong T cell response.
  • These two dominant T-cell epitopes were located in almost the same position in the PD-L1 and PD-L2 proteins ( FIG. 6A ).
  • these two dominant epitopes shared five amino acids (FTVTV; SEQ ID NO 34), due to sequence similarities between PD-L1 and PD-L2 ( FIG. 6A ).
  • FTVTV SEQ ID NO 34
  • PD-L101 (PDL1(15-23))-Specific T Cells.
  • 6C illustrates that the PD-L101-specific T cells only lysed T2-cells pulsed with PD-L101 (PDL1(15-23)), and no cytotoxicity was observed against T2-cells pulsed with either PD-L205 (PD-L2(16-25)), or the irrelevant HIV peptide.
  • PD-L101 PDL1(15-23)
  • PD-L205 PD-L2(16-25)
  • IFN- ⁇ and TNF- ⁇ intracellular cytokine staining assay to analyze cytokine release (IFN- ⁇ and TNF- ⁇ ) from PD-L101-specific CD8+ T cells, in response to PD-L101 (PDL1(15-23)), PD-L205 (PD-L2(16-25)), or the control HIV peptide ( FIG. 5C ).
  • PD-L101-specific T cells released cytokines only in response to PD-L101 (PDL1(15-23)) and not in response to PD-L205 (PD-L2(16-25)) or the irrelevant HIV peptide ( FIG. 6D ).
  • PD-L205-reactive T cells could specifically recognize PD-L205 (PD-L2(16-25)), but not PD-L101 (PDL1(15-23)).
  • PD-L205-reactive T-cell cultures with the standard 51Cr release assays, with TAP-deficient T2 cells as target cells.
  • the target cells were loaded with PD-L205 (PD-L2(16-25)), PD-L101 (PDL1(15-23)), or the control HIV peptide (HIV-1 po1476-484).
  • the PD-L205-specific T cells could indeed recognize T2 cells pulsed with PD-L205 (PD-L2(16-25)), but they did not kill T2 cells pulsed with PD-L101 (PDL1(15-23)), or with the control HIV peptide ( FIG. 6E ).
  • PD-L2 was examined as a target for specific T cells, and it was determined that PD-L2-specific T cells are spontaneously present in patients with different cancers including NHL.
  • the strongest responses were elicited by the peptide, PD-L205 (SEQ ID NO 4).
  • PD-L205 SEQ ID NO 4
  • CD8+ and CD4+ T cells that specifically recognized the minimal epitope in PD-L205(PD-L2(16-25)), selected for its HLA-A2 peptide binding motif.
  • the PD-L205(PD-L2(16-25)) epitope was similar to the main HLA-A2 restricted epitope that was identified previously in PD-L1.
  • PD-L1- and PD-L2-specific T cells did not cross react; therefore, they should be considered different T-cell antigens.
  • the results further showed that PD-L2-specific T cells specifically recognize PD-L2+ target cells.
  • PD-L2-specific T cells recognize target cells in response to their PD-L2 expression levels.
  • inducing/boosting T-cell responses against PD-L2 represents an attractive strategy for treating hematologic malignancies, including NHL.
  • the long PD-L2 epitope described in this study will be administered to high-risk patients with NHL that are in remission after second-line chemotherapy.
  • This approach represents a major difference from employing monoclonal antibodies to target the PD1/PDL pathway. Indeed, in addition to reducing the direct immunoregulatory effects of PD-L2, these PD-L2-specific T cells might also inhibit other routes of immune suppression that are mediated by PD-L2+ target cells (7).
  • PD-L2-specific T-cells released both IFN- ⁇ and TNF- ⁇ .
  • PD-L2-based vaccines should be viewed as complementary, rather than competitive, to other forms of immunotherapy.
  • combining a PD-L2 vaccination with a checkpoint blocker is believed to be an effective therapy.
  • a checkpoint blockade could boost vaccine-activated PD-L2-specific T cells by preventing their inhibition at the tumor site.
  • the vaccine-induced upregulation of checkpoint molecules due to the release of pro-inflammatory cytokines, would also be blocked by the checkpoint inhibitors.
  • T cells spontaneously reacted to PD-L2-derived epitopes located in the signal peptide region of PD-L2.
  • the PD-L2 epitopes recognized by T cells are therefore different than any epitope recognized by anti-PDL2 blocking antibodies.
  • cancer vaccines represent a way to eliminate minimal residual disease without inducing significant toxicity and secondary malignancies.
  • they have largely failed to demonstrate a significant improvement in patient outcome (9). This failure probably reflects the ability of malignant cells to suppress the function of the induced immune cells.
  • the addition of PD-L2 epitopes to current cancer vaccine strategies is likely to be highly beneficial, and it would be easy to implement.
  • stromal cell types in the tumor microenvironment are genetically stable, and thus, they represent attractive therapeutic targets with reduced risk of resistance and tumor recurrence.
  • PD-L2-specific T cells in patients with cancer.
  • PD-L2 may thus serve as a highly accessible target for immunotherapeutic strategies.
  • ATGYPLAEV PDL2(172-180) (SEQ ID NO. 19) GLYQVTSVL PDL2(168-176) (SEQ ID NO. 20) RTPEGLYQV PDL2(242-251) (SEQ ID NO. 21) ALRKQLCQKL 3PDL2(31-40) (SEQ ID NO. 22) IIEHGSNVTL PDL2(130-139) (SEQ ID NO. 23) ILKVPETDEV PDL2(149-158) (SEQ ID NO.
  • PLAEVSWPNV PDL2(4-25) (SEQ ID NO: 25) LLLMLSLELQLHQIAALFTVTV HIV-1 pol476-484 (SEQ ID NO: 26) ILKEPVHGV PDL1(15-23) (SEQ ID NO: 27) LLNAFTVTV PD-L2 siRNA duplexes (SEQ ID NO 28) 5′-CAUCCUAAAGGUUCCAGAAtt-3′ (SEQ ID NO 29) 5′-UUCUGGAACCUUUAGGAUGtg 3′ (SEQ ID NO 30) 5′-CCUAAGGAACUGUACAUAAtt-3′ (SEQ ID NO 31) 5′-UUAUGUACAGUUCCUUAGGga 3′ (SEQ ID NO 32) 5′-GAAAACAACUCUGUCAAAAtt-3′ (SEQ ID NO 33) 5′-UUUUGACAGAGUUGUUUUCtt 3′ PDL1 and PDL2 epitopes shared five amino acids (SEQ ID NO 34

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US16/344,445 2016-10-27 2017-10-13 New pdl2 compounds Abandoned US20200024324A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP16195949.9 2016-10-27
EP16195949 2016-10-27
PCT/EP2017/076179 WO2018077629A1 (en) 2016-10-27 2017-10-13 New pdl2 compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/076179 A-371-Of-International WO2018077629A1 (en) 2016-10-27 2017-10-13 New pdl2 compounds

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/911,996 Division US11447537B2 (en) 2016-10-27 2020-06-25 PDL2 compounds

Publications (1)

Publication Number Publication Date
US20200024324A1 true US20200024324A1 (en) 2020-01-23

Family

ID=57211335

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/344,445 Abandoned US20200024324A1 (en) 2016-10-27 2017-10-13 New pdl2 compounds
US16/911,996 Active US11447537B2 (en) 2016-10-27 2020-06-25 PDL2 compounds
US17/819,280 Pending US20230227529A1 (en) 2016-10-27 2022-08-11 Pdl2 compounds

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/911,996 Active US11447537B2 (en) 2016-10-27 2020-06-25 PDL2 compounds
US17/819,280 Pending US20230227529A1 (en) 2016-10-27 2022-08-11 Pdl2 compounds

Country Status (5)

Country Link
US (3) US20200024324A1 (ja)
EP (1) EP3532487A1 (ja)
JP (2) JP7312106B2 (ja)
CN (1) CN109890838A (ja)
WO (1) WO2018077629A1 (ja)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7211938B2 (ja) * 2016-10-07 2023-01-24 アイオー バイオテック エーピーエス 免疫原性アルギナーゼペプチド
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US58767A (en) 1866-10-16 John brougjbton
US5554372A (en) 1986-09-22 1996-09-10 Emory University Methods and vaccines comprising surface-active copolymers
AU2001271714A1 (en) 2000-06-30 2002-01-14 Human Genome Sciences, Inc. B7-like polynucleotides, polypeptides, and antibodies
AU2008293885A1 (en) * 2007-07-13 2009-03-05 The John Hopkins University B7-DC variants
US9243052B2 (en) * 2007-08-17 2016-01-26 Daniel Olive Method for treating and diagnosing hematologic malignancies
US20110223188A1 (en) * 2008-08-25 2011-09-15 Solomon Langermann Targeted costimulatory polypeptides and methods of use to treat cancer
ES2545609T3 (es) 2008-08-25 2015-09-14 Amplimmune, Inc. Composiciones de antagonistas de PD-1 y métodos de uso
US20130017199A1 (en) 2009-11-24 2013-01-17 AMPLIMMUNE ,Inc. a corporation Simultaneous inhibition of pd-l1/pd-l2
US20170044268A1 (en) * 2013-12-23 2017-02-16 OncoMed Pharmaceuticals Immunotherapy with Binding Agents
WO2016168771A2 (en) * 2015-04-17 2016-10-20 Alpine Immune Sciences, Inc. Immunomodulatory proteins with tunable affinities

Also Published As

Publication number Publication date
US20230227529A1 (en) 2023-07-20
US20200392202A1 (en) 2020-12-17
JP2020500171A (ja) 2020-01-09
JP7312106B2 (ja) 2023-07-20
JP2023109840A (ja) 2023-08-08
CN109890838A (zh) 2019-06-14
US11447537B2 (en) 2022-09-20
EP3532487A1 (en) 2019-09-04
WO2018077629A1 (en) 2018-05-03

Similar Documents

Publication Publication Date Title
JP5502823B2 (ja) Bcl−2ファミリ−に属するタンパク質およびそのフラグメント、ならびに癌患者におけるそれらの使用
US20230227529A1 (en) Pdl2 compounds
TWI318630B (en) Wt1 modified peptide
ES2510493T3 (es) Epítopos T CD4+ de la Survivina y sus aplicaciones
EP3112378B1 (en) Wt1 antigenic polypeptide, and anti-tumor agent containing said polypeptide
US20240167013A1 (en) Immunogenic arginase peptides
US20220372108A1 (en) Pdl1 peptides for use in cancer vaccines
WO2022070496A1 (ja) SARS-CoV-2蛋白由来ペプチドおよびそれを含むワクチン
CN113164545A (zh) 免疫原性精氨酸酶2多肽
FR2889959A1 (fr) Epitopes t cd4+ des antigenes mage-a restreints a hla-dp4 et leurs applications
TW201138803A (en) ECT2 peptides and vaccines including the same
KR102674521B1 (ko) 면역원성 아르기나제 펩타이드
WO2022149295A1 (ja) SARS-CoV-2蛋白由来ペプチドおよびそれを含むワクチン
TW201617361A (zh) 來自urlc10的胜肽及含其之疫苗
WO2020127996A2 (fr) Melanges d'epitopes t cd8+ immunogenes de la cycline b1

Legal Events

Date Code Title Description
AS Assignment

Owner name: IO BIOTECH APS, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HERLEV HOSPITAL;REEL/FRAME:048992/0222

Effective date: 20190128

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION