WO2023048518A1 - Dimère de protéine de fusion comprenant pd-1 et il-15 et utilisation associée - Google Patents

Dimère de protéine de fusion comprenant pd-1 et il-15 et utilisation associée Download PDF

Info

Publication number
WO2023048518A1
WO2023048518A1 PCT/KR2022/014304 KR2022014304W WO2023048518A1 WO 2023048518 A1 WO2023048518 A1 WO 2023048518A1 KR 2022014304 W KR2022014304 W KR 2022014304W WO 2023048518 A1 WO2023048518 A1 WO 2023048518A1
Authority
WO
WIPO (PCT)
Prior art keywords
fusion protein
protein
seq
cells
cancer
Prior art date
Application number
PCT/KR2022/014304
Other languages
English (en)
Korean (ko)
Inventor
서정근
Original Assignee
바이오엔시스템스 주식회사
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 바이오엔시스템스 주식회사 filed Critical 바이오엔시스템스 주식회사
Publication of WO2023048518A1 publication Critical patent/WO2023048518A1/fr

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5443IL-15
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the present invention relates to a fusion protein comprising a PD-1 protein and an IL-15 protein and uses thereof. Specifically, the present invention relates to a novel fusion protein dimer comprising PD-1, IL-15, and long-acting Fc in blood, which has an increased half-life in blood and has an immune-enhancing effect, and uses thereof for cancer treatment.
  • PD-1 Programmed death receptor-1
  • T cell activation mainly involved in the control of T cell activation and can regulate the strength and duration of an immune response.
  • PD-1 mediates and maintains body tissue autoimmune tolerance, prevents the immune system from over-activating and damaging autologous tissue during the inflammatory response, thus preventing autoimmune diseases from occurring. It works.
  • PD-1 is known to be involved in the development and development of tumor immunity and various autoimmune diseases (Sara Pilotto et.al. , Anticancer Agents Med Chem. , 15(3):307-313 , 2015).
  • PD-1 is mainly expressed on the surface of activated T cells, and is also expressed on B cells, NK cells, monocytes, and dendritic cells (DCs).
  • the ligands of PD-1, PD-L1 and PD-L2 are expressed on tumor cells, activated B and T cells, dendritic cells, and macrophages.
  • PD-1 binds to these ligands and induces T-cell apoptosis, thereby weakening the cellular immune response.
  • blocking the PD-1/PD-L1 or PD-L2 pathway is a promising therapeutic approach being explored in many types of cancer research (Miguel F. Sanmamed et. al. , Cancer J. , 20(4): 256-261, 2014).
  • IL-15 Interleukin-15
  • IL-15 is a cytokine structurally similar to IL-2 and is expressed in macrophages, monocytes, dendritic cells, fibroblasts, and the like.
  • IL-15 exhibits biological activity by binding to IL-15 receptors composed of IL-15R ⁇ , IL-2R ⁇ , and ⁇ c subunits.
  • IL-15 and IL-15R ⁇ are co-expressed in activated dendritic cells and function in the form of an IL-15/IL-15R ⁇ complex.
  • the IL-15/IL-15R ⁇ complex binds to IL-2R ⁇ / ⁇ c on NK cells and T cells and consequently induces differentiation and proliferation of T cells and NK cells.
  • it is possible to efficiently kill tumor cells by overcoming immune suppression through continuous cytolytic activity of T cells and NK cells (Ying Yang et.al. , Cancers , 12(12):3586, 2020). .
  • FcRn protection receptor binding site that lengthens the half-life of the antibody
  • FcRn is an MHC class I-related protein expressed in vascular endothelial cells and binds to IgG and albumin.
  • Antibody fragments without Fc, i.e., without an FcRn-binding site, have a half-life in the body of around 2 to 3 hours, but IgG1, IgG2, and IgG4 with an FcRn-binding site have an average half-life of 3 weeks in the body, which is longer than other proteins. .
  • the present inventors studied to develop an anti-cancer drug having an increased half-life in the body and excellent immune enhancing efficacy.
  • a novel fusion protein including PD-1, IL-15 and long-acting Fc in blood activates immune cells
  • the present invention was completed by confirming that the half-life was increased without affecting the anticancer activity.
  • one aspect of the present invention provides a fusion protein comprising a PD-1 protein and an IL-15 protein.
  • Another aspect of the present invention provides a fusion protein dimer in which the two fusion proteins are combined.
  • Another aspect of the present invention provides a polynucleotide encoding the fusion protein.
  • Another aspect of the present invention provides a vector containing the polynucleotide.
  • Another aspect of the present invention provides a host cell transformed with the vector.
  • Another aspect of the present invention provides a pharmaceutical composition for preventing or treating cancer disease comprising the fusion protein or fusion protein dimer as an active ingredient.
  • Another aspect of the present invention provides the use of the fusion protein for treating cancer disease.
  • a fusion protein containing PD-1, IL-15, and long-acting Fc in the blood can activate immune cells by PD-1 and IL-15, and maximize half-life in vivo by long-acting Fc in the blood. . Therefore, since the fusion protein can efficiently attack cancer cells, it can be usefully used in the treatment of cancer diseases.
  • FIG. 1 shows the obtained fusion proteins (BNS002; PD-1 D-Fc(29)-IL-15, PD-1 D-Fc(41)-IL-15, and PD-1 D-Fc(wt)-IL -15) was confirmed by SDS-PAGE.
  • Figure 2 shows the results of analyzing the purity of the fusion protein (BNS002) obtained by performing SEC-HPLC analysis.
  • Figure 3 confirms the concentration of the fusion protein (BNS002) obtained using a UV spectrophotometer.
  • Figure 4 confirms the thermodynamic stability of the fusion protein (BNS002) obtained using fluorescence and SLS (Static Light Scattering) detectors.
  • hPD-1 protein human programmed cell death protein-1
  • hPD-L1 human programmed cell death protein-1
  • BNS002 peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • Figure 25a confirms the effect of the obtained fusion protein (BNS002) and IL-15 protein on NK cell proliferation through FACS analysis.
  • Figure 25b is a graph of the results of confirming the effect of the obtained fusion protein (BNS002) and IL-15 protein on the proliferation of NK cells through FACS analysis.
  • FIG. 28 schematically shows a lentivirus vector map used to construct a cell line expressing Firefly luciferase and green fluorescent protein (GFP).
  • CMV is a promoter derived from Cytomegalovirus
  • RSV is a promoter derived from respiratory syncytial virus.
  • 29 shows the results of measuring the luminescence signal (left) and fluorescence signal (right) of the human melanoma cell line (A375-Luc-GFP) expressing firefly luciferase and GFP established in the present invention.
  • FIG. 30 shows PD-L1 and PD-L2 genes (left) specifically expressed in human melanoma cell line (A375-Luc-GFP) and PBMC containing immune cells according to an embodiment of the present invention.
  • FIG. 31 shows the IFN-secreted from the cells upon treatment with the obtained fusion protein (BNS002) and IL-15 in co-cultured human melanoma cell line (A375-Luc-GFP) and peripheral blood mononuclear cells (PBMC), respectively. This is the result of measuring the amount of ⁇ through ELISA analysis.
  • Figure 33 shows the cancer cell killing effect after treatment of the obtained fusion protein (BNS002) on co-cultured human melanoma cell line (A375-Luc-GFP) and peripheral blood mononuclear cells (PBMC) for 48 hours. It was confirmed by measuring the activity of tetrazolium reductase.
  • FIG. 36 schematically shows administration and experimental schedules for confirming the anticancer effect of the fusion protein (BNS002) in mice with mouse-derived colon cancer cells implanted therein.
  • Figure 39 confirms the degree of immune cell activity in cancer tissue by the obtained fusion protein (BNS002), and in detail, mice with mouse-derived colorectal cancer cells were treated with the fusion protein (BNS002) and excised on day 35 in the cancer tissue.
  • the results of FACS analysis of the activity levels of phagocytes, dendritic cells (DC), CD4+ cells, CD8+ cells and NK cells are shown in graphs.
  • FIG. 40 schematically shows administration and experimental schedules for confirming the anticancer effect of the fusion protein (BNS002) in humanized mice implanted with human-derived lung cancer cells (H460).
  • FIG. 43 schematically shows administration and experimental schedules for confirming the anticancer effect of the fusion protein (BNS002) in humanized mice implanted with human-derived colorectal cancer cells (HCT116).
  • BNS002 concentration-dependent tumor suppression effect by administration of the fusion protein (BNS002) in humanized mice implanted with human-derived colorectal cancer cells (HCT116).
  • FIG. 46 schematically shows administration and experimental schedules for confirming the anticancer effect between a fusion protein (BNS002 or BNS002S) and a control drug in humanized mice implanted with human-derived colorectal cancer cells (HCT116).
  • BNS002 or BNS002S fusion protein
  • HCT116 human-derived colorectal cancer cells
  • BNS002 PD-1 D-Fc-IL-15
  • HCT116 human-derived colorectal cancer cells
  • BNS002 PD-1 D-Fc-IL-15
  • HCT116 human-derived colorectal cancer cells
  • FIG. 49 shows the tumor suppression effect by administration of a fusion protein (BNS002; PD-1 D-Fc-IL-15) in humanized colorectal cancer cell (HCT116) implanted humanized mice in positive control groups atezolizumab, pembrolizumab, and Abel This is a result compared to Lumab administration.
  • BNS002 PD-1 D-Fc-IL-15
  • HCT116 humanized colorectal cancer cell
  • fusion protein BNS002; PD-1 D-Fc-IL-15
  • HCT116 human-derived colorectal cancer cells
  • BNS002S a fusion protein
  • HCT116 human-derived colorectal cancer cells
  • BNS001D PD-1 D -Fc
  • BNS002I Fc-IL-15
  • BNS002S fusion protein
  • PD-1D-Fc-IL-15/IL-15Ra human-derived colon cancer cells
  • HCT116 human-derived colon cancer cells
  • BNS001D PD-1 D -Fc
  • BNS002I Fc-IL-15
  • FIG. 53 shows the tumor suppression effect by administration of a fusion protein (BNS002S; PD-1D-Fc-IL-15/IL-15Ra) in humanized colon cancer cell (HCT116) implanted humanized mice as positive control groups, atezolizumab and pembroli. This is a result compared to administration of Zumab and Avelumab.
  • BNS002S a fusion protein
  • HCT116 humanized colon cancer cell
  • fusion proteins BNS002S; PD-1D-Fc-IL-15/IL-15Ra
  • HCT116 human-derived colorectal cancer cells
  • FIG. 55 schematically shows administration and experimental schedules for confirming the anticancer effect between a fusion protein (BNS002 or BNS002S) and a control drug in humanized mice implanted with human-derived lung cancer cells (A549).
  • BNS002 PD-1 D-Fc-IL-15
  • A549 human-derived lung cancer cells
  • 57 shows the degree of body weight change by administration of a fusion protein (BNS002; PD-1 D-Fc-IL-15) in humanized mice implanted with human-derived lung cancer cells (A549) as positive controls: atezolizumab, pembrolizumab, and avelumab. This is the result of comparison with administration.
  • BNS002 PD-1 D-Fc-IL-15
  • BNS002S a fusion protein
  • PD-1D-Fc-IL-15/IL-15Ra a fusion protein
  • 59 is a result of confirming the degree of body weight change according to administration of fusion protein (BNS002S; PD-1D-Fc-IL-15/IL-15Ra) at different concentrations in humanized mice implanted with human-derived lung cancer cells (A549).
  • 60 shows the tumor suppression effect by administration of a fusion protein (BNS002S; PD-1D-Fc-IL-15/IL-15Ra) in humanized mice implanted with human-derived lung cancer cells (A549) in positive control groups, atezolizumab and pembrolizumab. and the results compared with administration of Avelumab.
  • BNS002S a fusion protein
  • PD-1D-Fc-IL-15/IL-15Ra humanized lung cancer cells
  • 61 shows the degree of weight change by administration of fusion proteins (BNS002S; PD-1D-Fc-IL-15/IL-15Ra) in humanized mice implanted with human-derived lung cancer cells (A549) in positive control groups, atezolizumab and pembrolizumab. and the results compared with administration of Avelumab.
  • BNS002 or BNS002S fusion protein
  • MDA-MB-231 human-derived breast cancer cells
  • 63 shows the tumor suppression effect by administration of a fusion protein (BNS002; PD-1 D-Fc-IL-15) in humanized breast cancer cells (MDA-MB-231) implanted humanized mice in positive control groups, atezolizumab and pembroli. This is a result compared to administration of Zumab and Avelumab.
  • BNS002 PD-1 D-Fc-IL-15
  • MDA-MB-231 humanized breast cancer cells
  • BNS002 PD-1 D-Fc-IL-15
  • MDA-MB-2311 human-derived breast cancer cells
  • 65 shows the tumor suppression effect by administration of a fusion protein (BNS002S; PD-1D-Fc-IL-15/IL-15Ra) in humanized mice implanted with human-derived breast cancer cells (MDA-MB-231) as a positive control group, atezolizumab. , This is a result of comparison with administration of pembrolizumab and avelumab.
  • 66 shows the degree of body weight change by administration of a fusion protein (BNS002S; PD-1D-Fc-IL-15/IL-15Ra) in humanized mice implanted with human-derived breast cancer cells (MDA-MB-231) as a positive control group, atezolizumab. , This is a result of comparison with administration of pembrolizumab and avelumab.
  • BNS002S a fusion protein
  • MDA-MB-231 human-derived breast cancer cells
  • 67 is a schematic diagram of an embodiment of a fusion protein (BNS002) dimer.
  • 68 is a schematic diagram of an embodiment of a fusion protein (BNS002S) dimer.
  • Fusion protein comprising PD-1 protein and IL-15 protein
  • One aspect of the present invention provides a fusion protein comprising PD-1 protein and IL-15 protein.
  • the fusion protein may further include an IL-15 binding protein.
  • the fusion protein may further include an Fc region.
  • PD-1 programmed cell death protein 1 or programmed death receptor-1
  • CD279. immune checkpoint
  • PD-1 is widely expressed on the surface of activated T cells, natural killer T cells, B cells and macrophages.
  • PD-1 binds to its ligand, programmed cell death ligand-1 (PD-L1) or programmed cell death ligand-2 (PD-L2)
  • intracellular signaling inhibits CD3- and CD28-mediated T cell activation.
  • induce Downregulation of the T cell activation results in a decrease in T cell proliferation, IFN- ⁇ secretion, IL-2 secretion, and the like.
  • PD-1 and its ligand known to be expressed in various types of cancer cells e.g., melanoma, liver cancer, lung cancer, ovarian cancer, cervical cancer, bladder cancer, kidney cancer, pancreatic cancer, breast cancer, lymphoma, glioma, etc.
  • cancer cells e.g., melanoma, liver cancer, lung cancer, ovarian cancer, cervical cancer, bladder cancer, kidney cancer, pancreatic cancer, breast cancer, lymphoma, glioma, etc.
  • Interaction with PD-L1 or PD-L2 inactivates T cells and acts as a mechanism for cancer cells to evade the immune system's attack. Therefore, blocking the interaction between PD-1 and PD-L1 or PD-L2 activates T cells in the tumor microenvironment to eliminate tumor cells.
  • PD-1 programmed cell death receptor-1
  • programmed death protein-1 refers to a mammal, e.g., a primate (e.g., a human), unless otherwise indicated. ) and any wild-type PD-1 obtained from any vertebrate source, including rodents (eg, mice and rats).
  • the PD-1 may be obtained from animal cells, but also includes those obtained from recombinant cells capable of producing PD-1.
  • the PD-1 may be wild-type PD-1 or a fragment thereof.
  • PD-1 is a type I membrane protein consisting of 288 amino acids, and its structure consists of an extracellular IgV-like domain, a transmembrane domain, and a cytoplasmic domain.
  • the amino acid sequence of PD-1 is GenBank accession NO. NP_005009.2 or UniProtkB No. Q15116 (for the amino acid sequence of mouse PD-1, see Genbank accession No. NP_032824.1).
  • PDCD1 (programmed cell death 1) gene as a sequence encoding the PD-1 is GenBank accession NO. It may be a nucleotide sequence corresponding to a coding sequence (CDS) among the sequences described in NM_005018.3 (see GenBank accession No. NM_008798.3 for mouse sequence).
  • PD-1 protein refers to full-length PD-1 or PD-1 fragments. In the present specification, PD-1 or fragments thereof are collectively referred to as "PD-1 protein". PD-1, PD-1 proteins and PD-1 fragments specifically bind to, for example, PD-1's ligand, PD-L1 or PD-L2. This specific binding can be confirmed by methods known to those skilled in the art.
  • the ligands of PD-1, PD-L1 and PD-L2 are proteins expressed on the surface of cancer cells.
  • PD-L1 is also known as B7-H1 or CD274, and PD-L2 is also known as B7-DC or CD273.
  • PD-L1 or PD-L2 binds to PD-1 present in T cells, it suppresses the function of T cells by changing the immune checkpoint function. That is, T cell function is suppressed by the interaction of PD-1 and PD-L1 or PD-L2, and ultimately cancer cells evade the attack of immune cells.
  • the term "PD-1 fragment” means a truncated form of PD-1.
  • the PD-1 fragment may be an extracellular domain of PD-1.
  • One specific example of the PD-1 fragment may be one in which the 1st to 24th amino acids from the N-terminus, which is the signal sequence of PD-1, are excluded.
  • one specific example of the PD-1 fragment may be a protein composed of the 25th to 288th amino acids of SEQ ID NO: 18.
  • one specific example of the PD-1 fragment may be a protein composed of 25th to 170th amino acids of SEQ ID NO: 18.
  • one specific example of the PD-1 fragment may be a protein composed of the 25th to 150th amino acids of SEQ ID NO: 18.
  • one specific example of the PD-1 fragment may be a protein composed of 25th to 144th amino acids of SEQ ID NO: 18.
  • the PD-1 fragment may have the amino acid sequence of SEQ ID NO: 2 or 39.
  • the PD-1 fragment may be a protein composed of amino acids 25th to 169th of SEQ ID NO: 31.
  • the PD-1 fragment may have the amino acid sequence of SEQ ID NO: 23.
  • IL-15 or "interleukin-15” is a cytokine structurally similar to IL-2, and belongs to a family having a four ⁇ -helix structure. IL-15 is expressed in macrophages, monocytes, dendritic cells, fibroblasts, and the like. IL-15 binds to the receptor for IL-15, which consists of IL-15R ⁇ (IL-15 receptor alpha), IL-2R ⁇ (IL-2 receptor beta; also known as IL-15R ⁇ or CD122), and ⁇ c (also known as CD132) subunits. binding to exhibit biological activity.
  • IL-15R ⁇ IL-15 receptor alpha
  • IL-2R ⁇ IL-2 receptor beta
  • ⁇ c also known as CD132
  • IL-15 and IL-15R ⁇ are co-expressed in activated dendritic cells and function in the form of an IL-15/IL-15R ⁇ complex.
  • the IL-15/IL-15R ⁇ complex binds to IL-2R ⁇ / ⁇ c on NK cells and T cells and consequently induces differentiation and proliferation of T cells and NK cells. That is, it is possible to efficiently kill tumor cells by overcoming immunosuppression by continuous cytolytic activity of T cells and NK cells.
  • IL-15 promotes the induction of immunoglobulin synthesis by B cells and the regulation of lymphoid homeostasis.
  • IL-15 shares the IL-2R ⁇ and IL-2R ⁇ receptors with IL-2, but does not bind to the IL-2R ⁇ (CD25) receptor and thus does not activate regulatory T cells (Treg) that suppress immunity.
  • IL-15 or "interleukin-15”, unless stated otherwise, includes mammals such as primates (eg humans) and rodents (eg mice and rats). Any wild-type IL-15 obtained from any vertebrate source.
  • the IL-15 may be obtained from animal cells, but also includes those obtained from recombinant cells capable of producing IL-15.
  • the IL-15 may be wild-type IL-15, an isoform, or a variant thereof.
  • IL-15 has two IL-15 isoforms with different signal peptide lengths, but these two isoforms produce the same mature IL-15.
  • IL-15 isoform has IL-15 LSP (Isoform IL-15-S48AA; Interlekin-15 isoform 1 preproprotein, Homo sapiens , GenBank accession No. NP_000576.1) with a long signal peptide of 48 amino acids, 21
  • IL-15 SSP Isoform IL-15-S21AA; Interleukin-15 isofrom 2 preproprotein, Homo sapiens , GenBank accession No. NP_751915.1 in which a short signal peptide of four amino acids is present.
  • the two IL-15 isoforms share 11 amino acids (CFSAGLPKTEA) between the N-terminal signal sequences, and produce the same mature IL-15, although there are differences in intracellular trafficking.
  • the IL-15 LSP isoform is secreted from the Golgi apparatus, early endosomes, and endoplasmic reticulum (ER), whereas the IL-15 SSP isoform is not secreted and exists in the cytoplasm and nucleus.
  • IL-15 consists of a signal peptide and a mature polypeptide, the amino acid sequence of which is GenBank accession No. NP_000576.1, NP_751915.1, CAA71044.1 or UniProtkB No. P40933 (see Genbank accession No. NP_001241676.1 for the amino acid sequence of mouse IL-15).
  • the IL-15 gene is GenBank accession NO. It may be a nucleotide sequence corresponding to a coding sequence (CDS) among the sequences described in NM_000585.5, NM_172175.3 or Y09908.1 (for mouse sequences, see GenBank accession No. NM_001254747.1).
  • IL-15 consists of 136 amino acids, and may consist of a secretion signal sequence of amino acid residues 1 (Met) to 22 (Ala) and a mature polypeptide of amino acid residues 23 (Asn) to 136 (Ser).
  • IL-15 consists of 162 amino acids, and may consist of a secretion signal sequence of amino acid residues 1 (Met) to 48 (Ala) and a mature polypeptide of amino acid residues 49 (Asn) to 162 (Ser).
  • IL-15 consists of 135 amino acids, and may consist of a secretion signal sequence of amino acid residues 1 (Met) to 21 (Ala) and a mature polypeptide of amino acid residues 22 (Asn) to 135 (Ser).
  • IL-15 protein refers to full-length IL-15, IL-15 fragments, or IL-15 variants.
  • IL-15, IL-15 fragments, or variants thereof are collectively referred to as "IL-15 protein” or "IL-15 polypeptide”.
  • IL-15, IL-15 proteins, IL-15 polypeptides, and IL-15 variants specifically bind to, for example, IL-15R ⁇ or IL-2R ⁇ . This specific binding can be confirmed by methods known to those skilled in the art.
  • the IL-15 may have an amino acid sequence of SEQ ID NO: 19, 20, 21 or 32. Also, at this time, the IL-15 may be in a mature form. Specifically, the matured IL-15 may not contain a signal sequence and may have the amino acid sequence of SEQ ID NO: 6 or 24. In this case, the IL-15 may be used as a concept including a fragment in which a part of the N-terminus or C-terminus of wild-type IL-15 is truncated.
  • the fragment of IL-15 is 1, 2, 3, 4, 5, 6, 7, 8, 9 consecutively from the N-terminus of the protein having the amino acid sequence of SEQ ID NO: 19. , 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 or 22 amino acids may be deleted.
  • the fragments of IL-15 are 1, 2, 3, 4, 5, 6, 7, 8, 9 consecutively from the C-terminus of the protein having the amino acid sequence of SEQ ID NO: 19. , 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 or 22 amino acids may be deleted.
  • the fragments of IL-15 are 1, 2, 3, 4, 5, 6, 7, 8, 9 consecutively from the N-terminus of the protein having the amino acid sequence of SEQ ID NO: 20. , 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47 or 48 amino acids may be deleted.
  • the fragments of IL-15 are 1, 2, 3, 4, 5, 6, 7, 8, 9 consecutively from the C-terminus of the protein having the amino acid sequence of SEQ ID NO: 20. , 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47 or 48 amino acids may be deleted.
  • fragments of IL-15 are 1, 2, 3, 4, 5, 6, 7, 8, 9 consecutively from the N-terminus of the protein having the amino acid sequence of SEQ ID NO: 21. , 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 amino acids may be deleted.
  • fragment of IL-15 is continuously 1, 2, 3, 4, 5, 6, 7, 8, 9 from the C-terminus of the protein having the amino acid sequence of SEQ ID NO: 21. , 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 amino acids may be deleted.
  • the fragments of IL-15 are 1, 2, 3, 4, 5, 6, 7, 8, 9 consecutively from the N-terminus of the protein having the amino acid sequence of SEQ ID NO: 32. , 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47 or 48 amino acids may be deleted.
  • the fragments of IL-15 are 1, 2, 3, 4, 5, 6, 7, 8, 9 consecutively from the C-terminus of the protein having the amino acid sequence of SEQ ID NO: 32. , 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47 or 48 amino acids may be deleted.
  • the term "IL-15 variant” refers to a full-length IL-15 or a form in which a part of the amino acid of the aforementioned IL-15 fragment is substituted. That is, the IL-15 mutant may have an amino acid sequence different from wild-type IL-15 or a fragment thereof. However, the IL-15 variant may have an activity equivalent to or similar to that of wild-type IL-15.
  • “IL-15 activity” may mean, for example, binding specifically to IL-15R ⁇ or IL-2R ⁇ , and this specific binding can be measured by a method known to those skilled in the art.
  • the IL-15 mutant may be one in which some amino acids of wild-type IL-15 are deleted and/or substituted.
  • an IL-15 mutant by amino acid deletion and/or substitution is the secretion signal sequence of 1 (Met) to 22 (Ala) in the amino acid sequence of SEQ ID NO: 19, as disclosed in US Patent No. 10,450,359. deletion, and amino acid substitution of Asn(N) with Asp(D) or Asn(N) with Ala(A) at position 72 of the mature 114 amino acid peptide.
  • the secretion signal sequence of 1 (Met) to 48 (Ala) is deleted from the amino acid sequence of SEQ ID NO: 20, and Asn (N) is converted to Asp (D) or Asn (N) at position 72 of the mature 114 amino acid peptide. ) may be amino acid substitution with Ala (A).
  • the secretion signal sequence of 1 (Met) to 21 (Ala) was deleted from the amino acid sequence of SEQ ID NO: 21, and Asn (N) was converted to Asp (D) or Asn (N) at position 72 of the mature 114 amino acid peptide. ) may be amino acid substitution with Ala (A).
  • the IL-15 variant may be characterized in that the activity of IL-15 is maintained or improved compared to wild-type IL-15.
  • the PD-1 protein and the IL-15 protein may be linked by a linker or a carrier.
  • the PD-1 or a fragment thereof and the IL-15 or a variant thereof may be linked by a linker or a carrier.
  • a linker and a carrier are also used interchangeably.
  • the linker connects the two proteins.
  • One specific example of the linker may include 1 to 50 amino acids, albumin or a fragment thereof, or an Fc domain of an immunoglobulin.
  • the Fc domain of the immunoglobulin includes heavy chain constant region 2 (CH2) and heavy chain constant region 3 (CH3) of immunoglobulin, and includes variable regions and light chain constant region 1 (CH1) of heavy and light chains of immunoglobulin.
  • protein that does not The immunoglobulin may be IgG, IgA, IgE, IgD or IgM, preferably IgG1.
  • the Fc domain of wild-type immunoglobulin G1 may have the amino acid sequence of SEQ ID NO: 14.
  • the Fc domain of the immunoglobulin may be a wild-type Fc domain as well as an Fc domain variant.
  • Fc domain variant refers to a glycosylation pattern that is different from that of the wild-type Fc domain, increased sugar chains compared to the wild-type Fc domain, reduced sugar chains compared to the wild-type Fc domain, or sugar chains removed. (deglycosylated) form. Also included are aglycosylated Fc domains.
  • the Fc domain or variant may have sialic acid, fucosylation, and glycosylation of numbers adjusted through culture conditions or genetic manipulation of the host.
  • the sugar chain of the Fc domain of immunoglobulin can be modified by conventional methods such as chemical methods, enzymatic methods, and genetic engineering methods using microorganisms.
  • the Fc domain variant may be a mixed form of an immunoglobulin Fc region of IgG, IgA, IgE, IgD or IgM.
  • the Fc domain variant may be a form in which some amino acids of the Fc domain are substituted with other amino acids.
  • Fc domain variant is Q81R, M198L, L79G, T136S, L138A, Y177V, Q81R/M198L, L79G/M198L, T136S/L138A/Y177V or Q81R/T136S/L138A/Y177V in the amino acid sequence of SEQ ID NO: 14. Substitution of /M198L may have occurred.
  • Fc domain variant may have an amino acid sequence of SEQ ID NO: 4, 10, 55, 88 or 97.
  • Fc domain variant by amino acid substitution is an Fc domain variant comprising an amino acid substitution that regulates binding and dissociation to FcRn (neonatal Fc receptor).
  • Fc variants that exhibit increased binding affinity to FcRn at low pH and show no substantially altered binding at high pH, or functional variants thereof.
  • the Fc domain variant has a binding affinity to FcRn of 10% or more, 20% or more, 30% or more, 40% or more, 50% or more at pH 5.6 to 6.2 (preferably pH 5.8 to 6.0) compared to the wild-type Fc domain.
  • the degree of dissociation of the Fc domain variant from the neonatal Fc receptor (FcRn) at pH 7.0 to 7.8 may be the same as or substantially unchanged compared to the wild-type Fc domain.
  • the Fc domain variant may be characterized in that half-life is increased compared to the wild type.
  • the half-life of the Fc domain variant is 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, 90% or more compared to the wild-type Fc domain. Increased by at least 100% or more, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold or at least 10-fold more than the wild-type Fc domain there is.
  • neonatal Fc receptor (FcRn) or “neonatal Fc receptor” is an MHC class I related protein expressed in vascular endothelial cells and binds to IgG and albumin.
  • the characteristic point is that the binding between IgG and FcRn is strong when the pH is slightly acidic, and there is no binding force at neutral pH. Therefore, IgG entering cells by pinocytosis or endocytosis binds strongly to FcRn, a type of Fc gamma receptor (Fc ⁇ R), in endosomes under pH 6.0 conditions, and forms degradative lysosomes.
  • FcRn a type of Fc gamma receptor
  • Fc gamma receptor is an Fc receptor for IgG, and there are three types of Fc ⁇ RI (CD64), Fc ⁇ RII (CD32), and Fc ⁇ RIII (CD16).
  • An Fc receptor is a molecule that binds to immunoglobulin and allows the bound antibody to perform various biological functions independently of an antigen binding site, and is distributed on the surface of various cells and tissues. Fc receptors are involved in cytotoxicity, antibody-dependent cell-mediated cytotoxicity (ADCC), secretion of mediators such as cytokines, phagocytosis, initiation of oxidation, and regulation of antibody production.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • the fusion protein may have a structure in which PD-1 and IL-15 proteins are connected to the N-terminus and C-terminus of the Fc domain as a linker (or carrier), or IL-15 and PD-1 are connected, respectively. Connection between the N-terminus or C-terminus of the Fc domain and PD-1 or IL-15 may be optionally achieved by a linker peptide.
  • the fusion protein including the PD-1 protein or a fragment thereof and the IL-15 protein or a variant thereof may additionally include an IL-15 binding protein.
  • IL-15 binding protein may be a protein or peptide that specifically binds to IL-15.
  • one specific example of the IL-15 binding protein may be “the “sushi” domain of IL-15R ⁇ .
  • the "sushi” domain which corresponds to a portion of the extracellular domain of the IL-15R ⁇ , is required for binding to IL-15.
  • the sushi domain of IL-15R ⁇ is the sushi domain of mammalian IL-15R ⁇ , preferably the sushi domain of primate IL-15R ⁇ , more preferably the sushi domain of human IL-15R ⁇ .
  • One specific example of the “sushi” domain of the IL-15R ⁇ may have the amino acid sequence of SEQ ID NO: 95.
  • the “sushi” domain of IL-15R ⁇ may be a fragment of SEQ ID NO: 95 (ITCPPPMSVEHADIWVKSYSLYSRERYICNSGFKRKAGTSSLTECVLNKATNVAHWTTPSLKCIRDPALVHQRPAPP (SEQ ID NO: 95)).
  • the fusion protein may be any one of the groups consisting of structural formulas I to IV:
  • N' is the N-terminus of the fusion protein
  • the C' is the C-terminus of the fusion protein
  • A is PD-1 protein or a fragment thereof
  • B is an IL-15 protein or a variant thereof
  • S is an IL-15 binding protein
  • L 1 , L 2 and L 3 are each a peptide linker
  • n, m and l are each independently 0 or 1.
  • PD-1 protein or fragment thereof IL-15 protein or variant thereof, IL-15 binding protein, peptide linker, and Fc domain are as described above.
  • the form in which the IL-15 binding protein is removed from the structure of the fusion protein may have structural formula V or VI:
  • N' is the N-terminus of the fusion protein
  • the C' is the C-terminus of the fusion protein
  • A is PD-1 protein or a fragment thereof
  • B is an IL-15 protein or a variant thereof
  • L 1 and L 2 are each a peptide linker
  • n and m are each independently 0 or 1.
  • the PD-1 protein or a fragment thereof, the IL-15 protein or a variant thereof, the peptide linker, and the Fc domain are as described above.
  • the fusion protein may have structural formula V.
  • the PD-1 protein and the IL-15 protein are each as described above.
  • the PD-1 protein may be a truncated fragment of up to about 24 amino acid residues consecutively from the N-terminus or C-terminus of wild-type PD-1.
  • the IL-15 protein may be a fragment in which about 21 to 48 amino acid residues are consecutively deleted from the N-terminus or C-terminus of wild-type IL-15.
  • the IL-15 protein may be an IL-15 mutant in which some amino acids of an IL-15 fragment are deleted and/or substituted.
  • the fusion protein may have an amino acid sequence of SEQ ID NO: 8, 12, 16, 26, 29, 33, 37, 41, 46, 49, 57, 60, 77, 80, 83, 90 or 93 .
  • the fusion protein is for the amino acid sequence of SEQ ID NO: 8, 12, 16, 26, 29, 33, 37, 41, 46, 49, 57, 60, 77, 80, 83, 90 or 93 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity It includes a polypeptide having. At this time, the identity may be determined through, for example, percent homology, homology comparison software such as NCBI (National Center of Biotechnology Information) BlastN software.
  • a peptide linker L 1 may be included between the PD-1 protein and the Fc domain.
  • the peptide linker L 1 may consist of 5 to 80 consecutive amino acids, 20 to 60 consecutive amino acids, or 25 to 50 consecutive amino acids, or 30 to 40 amino acids. In one embodiment, the peptide linker L 1 may consist of 30 amino acids.
  • the peptide linker L 1 may include at least one cysteine. Specifically, it may contain 1, 2 or 3 cysteines.
  • the peptide linker L 1 may be derived from the hinge of an immunoglobulin. In one embodiment, the peptide linker L 1 may be a peptide linker consisting of the amino acid sequence of SEQ ID NO: 3 or 43.
  • the peptide linker L 2 or L 3 may consist of 1 to 50 consecutive amino acids, or 3 to 30 consecutive amino acids, or 5 to 15 amino acids.
  • the peptide linker L 2 or L 3 may be (G 4 S)n (where n is an integer from 1 to 10) or include this. In this case, in (G4S)n, n may be 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • the peptide linker L 2 may be a peptide linker consisting of the amino acid sequence of SEQ ID NO: 5, 44 or 62.
  • the peptide linker L 3 may be a peptide linker consisting of the amino acid sequence of SEQ ID NO: 96.
  • Another aspect of the present invention provides a fusion protein dimer in which two fusion proteins including the PD-1 protein or a fragment thereof and the IL-15 protein or a variant thereof are linked.
  • the fusion protein comprising the PD-1 or a fragment thereof and the IL-15 protein or a variant thereof is as described above.
  • Another aspect of the present invention provides a fusion protein dimer in which two fusion proteins including the PD-1 protein or a fragment thereof, the IL-15 protein or a variant thereof, and an IL-15 binding protein are linked.
  • the fusion protein comprising the PD-1 or a fragment thereof, the IL-15 protein or a variant thereof, and the IL-15 binding protein is as described above.
  • the binding between the fusion proteins constituting the dimer may be made by a disulfide bond by a cysteine present in the linker, but is not limited thereto.
  • the fusion proteins constituting the dimer may be the same or may be different fusion proteins.
  • the dimer may be a homodimer.
  • One embodiment of the fusion protein constituting the dimer is SEQ ID NO: 8, 12, 16, 26, 29, 33, 37, 41, 46, 49, 57, 60, 77, 80, 83, 90 or 93 amino acids It may be a protein having a sequence.
  • polynucleotide encoding a fusion protein comprising PD-1 protein and IL-15 protein.
  • the polynucleotide may include the nucleotide sequence of SEQ ID NO: 9, 13, 17, 27, 30, 35, 38, 42, 47, 50, 58, 61, 78, 81, 84, 91 or 94 there is.
  • the fusion protein comprising the PD-1 protein and the IL-15 protein is as described above.
  • the polynucleotide may be mutated by substitution, deletion, insertion, or a combination of one or more bases.
  • a synthesis method well known in the art for example, a method described in the literature (Engels and Uhlmann, Angew Chem IntEd Engl., 37:73-127, 1988) can be used , triester, phosphite, phosphoramidite and H-phosphate methods, PCR and other autoprimer methods, oligonucleotide synthesis methods on solid supports, and the like.
  • the polynucleotide is at least about 70% SEQ ID NO: 9, 13, 17, 27, 30, 35, 38, 42, 47, 50, 58, 61, 78, 81, 84, 91 or 94 , at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92% , at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, or at least about 100% identity.
  • the polynucleotide may additionally include a nucleic acid encoding a signal sequence or a leader sequence.
  • signal sequence refers to a signal peptide that directs the secretion of a target protein.
  • the signal peptide is cleaved after translation in the host cell.
  • the signal sequence is an amino acid sequence that initiates protein movement through an endoplasmic reticulum (ER) membrane.
  • the signal sequence may have the amino acid sequence of SEQ ID NO: 1 or 22.
  • the signal sequence is well known in the art and usually includes 16 to 30 amino acid residues, but may include more or less amino acid residues.
  • a typical signal peptide consists of three regions: a basic N-terminal region, a central hydrophobic region, and a more polar C-terminal region.
  • the central hydrophobic region contains 4 to 12 hydrophobic residues that anchor the signal sequence throughout the membrane lipid bilayer during migration of the immature polypeptide.
  • the signal sequence is cleaved within the lumen of the ER by cellular enzymes commonly known as signal peptidases.
  • the signal sequence may be tPa (tissue plasminogen activation), HSV gDs (signal sequence of herpes simplex virus glycoprotein D), or growth hormone secretion signal sequence.
  • secretion signal sequences used in higher eukaryotic cells including mammals and the like can be used.
  • a signal sequence included in wild-type PD-1 and/or IL-15 may be used, or a codon frequently expressed in the host cell may be substituted for use.
  • Another aspect of the present invention provides a vector containing the polynucleotide.
  • the vector can be introduced into a host cell and then recombinated and inserted into the genome of the host cell.
  • the vector is understood to be a nucleic acid vehicle comprising a polynucleotide sequence capable of autonomous replication as an episome.
  • Such vectors include linear nucleic acids, plasmids, phagemids, cosmids, RNA vectors, viral vectors and analogues thereof.
  • examples of viral vectors include, but are not limited to, retroviruses, adenoviruses, and adeno-associated viruses.
  • the vector may be plasmid DNA, phage DNA, etc., commercially developed plasmids (pUC18, pBAD, pIDTSAMRT-AMP, etc.), Escherichia coli-derived plasmids (pYG601BR322, pBR325, pUC118, pUC119, etc.), Bacillus subtilis plasmids derived from spp.
  • pUB110, pTP5, etc. yeast-derived plasmids (YEp13, YEp24, YCp50, etc.), phage DNA (Charon4A, Charon21A, EMBL3, EMBL4, ⁇ gt10, ⁇ gt11, ⁇ ZAP, etc.), animal viral vectors (retrovirus ), adenovirus, vaccinia virus, etc.), insect virus vectors (baculovirus, etc.). Since the expression level and modification of the protein of the vector appear differently depending on the host cell, it is preferable to select and use the host cell most suitable for the purpose.
  • the term "gene expression” or “expression” of a protein of interest is understood to mean transcription of DNA sequences, translation of mRNA transcripts and secretion of fusion protein products or fragments thereof.
  • a useful expression vector can be RcCMV (Invitrogen, Carlsbad) or variants thereof.
  • the expression vector includes a human cytomegalovirus (CMV) promoter for promoting continuous transcription of a target gene in mammalian cells, and a bovine growth hormone polyadenylation signal sequence for increasing the steady-state level of RNA after transcription. can do.
  • CMV human cytomegalovirus
  • Another aspect of the present invention provides a transformed host cell into which the vector is introduced.
  • Host cells of the transformed cells may include, but are not limited to, prokaryotic cells, eukaryotic cells, mammalian cells, plants, insects, fungi, or cells of cellular origin.
  • prokaryotic cell Escherichia coli may be used.
  • yeast may be used as an example of a eukaryotic cell.
  • CHO cells, F2N cells, CSO cells, BHK cells, Bowes melanoma cells, HeLa cells, 911 cells, AT1080 cells, A549 cells, HEK 293 cells, or HEK293T cells may be used as the mammalian cells.
  • any cell that can be used as a mammalian host cell known to those skilled in the art can be used.
  • the CaCl 2 precipitation method when the expression vector is introduced into the host cell, the CaCl 2 precipitation method, the Hanahan method with increased efficiency by using a reducing material called DMSO (dimethyl sulfoxide) for the CaCl 2 precipitation method, the electroporation method, and the calcium phosphate precipitation method , protoplast fusion method, agitation method using silicon carbide fibers, agrobacteria-mediated transformation method, transformation method using PEG, dextran sulfate, lipofectamine, and drying/inhibition-mediated transformation methods, etc. may be used.
  • DMSO dimethyl sulfoxide
  • the sugar chain pattern of the fusion protein e.g., sialic acid, fucosylation, glycosylation.
  • Another aspect of the present invention provides a method for producing a fusion protein comprising PD-1 protein and IL-15 protein, comprising culturing the transformed cell.
  • the production method includes i) culturing the transformed cells to obtain a culture product; and ii) recovering the fusion protein from the culture.
  • the method of culturing the transformed cells may be performed using a method widely known in the art. Specifically, the culture may be cultured continuously in a batch process or an injection batch or a repeated injection batch process (fed batch or repeated fed batch process).
  • Another aspect of the present invention is for the treatment or prevention of cancer, comprising as an active ingredient a fusion protein comprising PD-1 protein and IL-15 protein or a fusion protein dimer in which the two fusion proteins are combined, and/or It provides a pharmaceutical composition capable of increasing the therapeutic effect (efficacy).
  • the fusion protein including the PD-1 protein and the IL-15 protein or the fusion protein dimer in which the two fusion proteins are combined are as described above.
  • the cancer is colorectal cancer, melanoma, stomach cancer, liver cancer, lung cancer, breast cancer, prostate cancer, ovarian cancer, pancreatic cancer, cervical cancer, bladder cancer, kidney cancer, gallbladder cancer, thyroid cancer, laryngeal cancer, acute myelogenous leukemia, brain tumor, neuroblastoma, retinoblastoma , head and neck cancer, salivary gland cancer and lymphoma.
  • a preferred dosage of the pharmaceutical composition varies depending on the condition and body weight of the patient, the severity of the disease, the type of drug, the route and duration of administration, but can be appropriately selected by those skilled in the art.
  • the active ingredient may be included in any amount (effective amount) according to the purpose of use, formulation, combination, etc., as long as it can exhibit anticancer activity.
  • a typical effective amount is the entire composition. On a weight basis it will be determined within the range of 0.001% to 20.0% by weight.
  • "effective amount” refers to the amount of an active ingredient capable of inducing an anticancer effect. Such an effective amount can be determined empirically within the ordinary skill of the skilled artisan.
  • the term “treatment” may be used to include both therapeutic treatment and prophylactic treatment. At this time, prevention may be used in the sense of alleviating or reducing the pathological condition or disease of the subject.
  • the term “treatment” includes any form of administration or application to treat a disease in a mammal, including humans. The term also includes inhibiting or slowing the disease or progression of the disease; restoring or repairing a damaged or missing function, thereby partially or completely alleviating the disease; or stimulate inefficient processes; It includes the meaning of alleviating serious illness.
  • the term “efficacy” refers to survival or disease-free survival over a period of time, such as 1 year, 5 years, or 10 years, by one or more parameters, such as disease-free survival. can be determined Additionally, the parameter may include inhibition of the size of at least one tumor in the subject.
  • Pharmacokinetic parameters such as bioavailability and underlying parameters such as clearance rate may also affect efficacy.
  • "enhanced efficacy” eg, improvement in efficacy
  • terapéuticaally effective amount or “pharmaceutically effective amount” is an amount of a compound or composition effective for preventing or treating a target disease, sufficient to treat the disease with a reasonable benefit / risk ratio applicable to medical treatment It means an amount that does not cause side effects.
  • the level of the effective amount is the patient's state of health, type of disease, severity, activity of the drug, sensitivity to the drug, method of administration, time of administration, route of administration and excretion rate, duration of treatment, factors including drugs used in combination or concurrently, and It may be determined according to other factors well known in the medical field.
  • a therapeutically effective amount refers to an amount of a drug effective to treat cancer.
  • the pharmaceutical composition may further include a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier may be any carrier as long as it is a non-toxic material suitable for delivery to a patient. Distilled water, alcohol, fats, waxes and inert solids may be included as carriers. A pharmacologically acceptable adjuvant (buffer, dispersant) may also be included in the pharmaceutical composition.
  • the pharmaceutical composition may be prepared as a parenteral formulation according to an administration route by a conventional method known in the art, including a pharmaceutically acceptable carrier in addition to the active ingredient.
  • pharmaceutically acceptable means that it does not inhibit the activity of the active ingredient and does not have toxicity more than is adaptable to the subject of application (prescription).
  • the pharmaceutical composition When the pharmaceutical composition is prepared as a parenteral formulation, it may be formulated in the form of injection, transdermal administration, nasal inhalation, and suppository along with a suitable carrier according to a method known in the art.
  • a suitable carrier preferably Ringer's solution, PBS (phosphate buffered saline) containing triethanolamine or sterilization for injection water, isotonic solutions such as 5% dextrose, and the like can be used.
  • suitable carriers preferably Ringer's solution, PBS (phosphate buffered saline) containing triethanolamine or sterilization for injection water, isotonic solutions such as 5% dextrose, and the like can be used.
  • Formulation of pharmaceutical compositions is known in the art, and may be specifically referred to in Remington's Pharmaceutical Sciences (19th ed., 1995). These documents are considered as part of this specification.
  • the preferred dosage of the pharmaceutical composition is in the range of 0.01 ⁇ g/kg to 10 g/kg, or 0.01 mg/kg to 1 g/kg per day depending on the patient's condition, weight, sex, age, severity of the patient, and route of administration. can be Administration can be done once a day or divided into several times. These dosages should not be construed as limiting the scope of the present invention in any respect.
  • compositions of the present application are mammals and humans, particularly preferably humans.
  • the pharmaceutical composition of the present application may further include any compound or natural extract known to have a therapeutic effect on anticancer activity and whose safety has already been verified for enhancement or enhancement of anticancer activity.
  • Another aspect of the present invention provides the use of a fusion protein comprising PD-1 protein and IL-15 protein for treating cancer diseases.
  • Another aspect of the present invention provides the use of a fusion protein comprising a PD-1 protein and an IL-15 protein to enhance the therapeutic effect of cancer disease.
  • Another aspect of the present invention provides the use of a fusion protein comprising a PD-1 protein and an IL-15 protein for preparing a drug for treating cancer.
  • Another aspect of the present invention is a method for treating cancer disease comprising administering to a subject a fusion protein comprising PD-1 protein and IL-15 protein or a fusion protein dimer in which the two fusion proteins are combined; and/or a method for enhancing the therapeutic effect.
  • the subject may be an individual suffering from a cancer disease. Also, the subject may be a mammal, preferably a human.
  • the fusion protein including the PD-1 protein and the IL-15 protein or the fusion protein dimer in which the two fusion proteins are combined are as described above.
  • the administration route, dosage and frequency of administration of the fusion protein or fusion protein dimer can be administered to the subject in various ways and amounts depending on the condition of the patient and the presence or absence of side effects, and the optimal administration method, dosage and frequency of administration are A person skilled in the art can select an appropriate range.
  • the fusion protein or fusion protein dimer may be administered in combination with other drugs or physiologically active substances known to have therapeutic effects on the disease to be treated, or formulated in combination with other drugs.
  • Another aspect of the present invention provides a cell culture medium containing a fusion protein comprising PD-1 protein and IL-15 protein or a fusion protein dimer in which the two fusion proteins are linked.
  • the cells may be T cells or natural killer cells.
  • the cell culture medium may be a medium in which a fusion protein including PD-1 protein and IL-15 protein or a fusion protein dimer in which the two fusion proteins are combined is added to a cell culture medium.
  • the cell culture medium may include any one selected from the group consisting of amino acids, sugars, inorganic salts, and vitamins.
  • the medium for cell culture may contain all amino acids, sugars, inorganic salts and vitamins.
  • the term "cell culture medium” refers to a medium used to culture cells, specifically immune cells, more specifically CD4+ or CD8+ T cells; or a medium for culturing NK cells. Contains components required by cells for cell growth and survival in vitro, or contains components that help cell growth and survival. Specifically, the components may be vitamins, essential or non-essential amino acids, and trace elements.
  • the medium may be a medium used for culturing cells, preferably eukaryotic cells, more preferably T cells or NK cells.
  • Signal peptide SEQ ID NO: 1
  • PD-1 fragment SEQ ID NO: 2
  • Ig hinge to which a linker is attached to produce a fusion protein including human PD-1 fragment, long-acting Fc (29) domain and IL-15 (SEQ ID NO: 3), Fc (29) domain (SEQ ID NO: 4), linker (SEQ ID NO: 5) and IL-15 (SEQ ID NO: 6) in order from the N-terminus to a fusion protein (SEQ ID NO: 7)
  • a polynucleotide containing the coding nucleotide sequence SEQ ID NO: 9 was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfecting the CHO cells with the expression vector, cultured for 9 days according to the manufacturer's Max Protocol, the culture medium was recovered, and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 8) is referred to as "hPD-1 D-Fc(29)-hIL-15" or "BNS002(29)", and thus a fusion protein containing PD-1 and IL-15
  • the dimer was collectively referred to as "BNS002".
  • the fusion protein in the recovered culture medium was first purified using Affinity Chromatography with Protein A Resin (MabSelect Sure TM ; Cytiva). After binding the fusion protein with Buffer A (20 mM Sodium Phosphate, 150 mM NaCl, pH 7.1) and washing the resin, only the bound fusion protein was eluted with Buffer B (100 mM Citrate, pH 3.0). The eluted fusion protein was neutralized (pH 7.0) with 1 M Tris-HCl (pH 9.0) buffer, concentrated using a centrifugal filter (Amicon Ultra-15, Merck), and buffer exchanged with PBS.
  • Buffer A 20 mM Sodium Phosphate, 150 mM NaCl, pH 7.1
  • Buffer B 100 mM Citrate, pH 3.0
  • the eluted fusion protein was neutralized (pH 7.0) with 1 M Tris-HCl (pH 9.0) buffer, concentrated using a centrifugal filter
  • the buffer-exchanged fusion protein was secondarily purified by performing size exclusion chromatography using a HiLoad ® 26/600 Superdex ® 200 pg column (Cytiva).
  • the separated and purified fusion protein was quantified by IGGHB analysis of Cedex Bio HT Analyzer (Roche), and the purity was analyzed by performing SDS-PAGE and SEC-HPLC analysis (FIG. 1 and FIG. 2).
  • Signal peptide SEQ ID NO: 1
  • PD-1 fragment SEQ ID NO: 2
  • Ig hinge to which a linker is attached to produce a fusion protein including human PD-1 fragment, long-acting Fc (41) domain and IL-15 (SEQ ID NO: 3), Fc (41) domain (SEQ ID NO: 10), linker (SEQ ID NO: 5) and IL-15 (SEQ ID NO: 6) in order from the N-terminus to a fusion protein (SEQ ID NO: 11)
  • a polynucleotide containing the coding nucleotide sequence SEQ ID NO: 13
  • GenScript's Gene Synthesis service was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfecting the CHO cells with the expression vector, cultured for 9 days according to the manufacturer's Max Protocol, the culture medium was recovered, and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 12) is referred to as "hPD-1 D-Fc(41)-hIL-15" or "BNS002(41)", and thus a fusion protein containing PD-1 and IL-15
  • the dimer was collectively referred to as "BNS002".
  • Signal peptide SEQ ID NO: 1
  • PD-1 fragment SEQ ID NO: 2
  • linker-linked Ig hinge to produce a fusion protein including human PD-1 fragment, long-acting Fc (wt) domain and IL-15 (SEQ ID NO: 3), Fc (wt) domain (SEQ ID NO: 14), linker (SEQ ID NO: 5) and IL-15 (SEQ ID NO: 6) in order from the N-terminus to a fusion protein (SEQ ID NO: 15)
  • a polynucleotide containing the coding base sequence SEQ ID NO: 17
  • GenScript's Gene Synthesis service was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfecting the CHO cells with the expression vector, cultured for 9 days according to the manufacturer's Max Protocol, the culture medium was recovered, and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 16) is referred to as "hPD-1 D-Fc(wt)-hIL-15" or "BNS002(wt)", and thus a fusion protein containing PD-1 and IL-15
  • the dimer was collectively referred to as "BNS002".
  • Signal peptide (SEQ ID NO: 22), PD-1 fragment (SEQ ID NO: 23), linker-linked Ig hinge to produce a fusion protein including mouse PD-1 fragment, long-acting Fc (29) domain and IL-15 (SEQ ID NO: 3), Fc (29) domain (SEQ ID NO: 4), linker (SEQ ID NO: 5) and IL-15 (SEQ ID NO: 24) in order from the N-terminus to a fusion protein (SEQ ID NO: 25)
  • a polynucleotide containing the coding base sequence (SEQ ID NO: 27) was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 26) is referred to as "mPD-1 D-Fc (29) -mIL-15" or "mBNS002 (29)", and thus a fusion protein containing mPD-1 and mIL-15
  • the dimer was collectively referred to as "BNS002". Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • Signal peptide (SEQ ID NO: 22), PD-1 fragment (SEQ ID NO: 23), linker-linked Ig hinge to produce a fusion protein including mouse PD-1 fragment, persistent Fc (wt) domain and IL-15 (SEQ ID NO: 3), Fc (wt) domain (SEQ ID NO: 14), linker (SEQ ID NO: 5) and IL-15 (SEQ ID NO: 24) in order from the N-terminus to a fusion protein (SEQ ID NO: 28)
  • a polynucleotide containing the coding nucleotide sequence (SEQ ID NO: 30) was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 29) is referred to as "mPD-1 D-Fc (wt) -mIL-15" or "mBNS002 (wt)", and thus a fusion protein containing PD-1 and IL-15
  • the dimer was collectively referred to as "BNS002". Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • Signal peptide SEQ ID NO: 1
  • PD-1 fragment SEQ ID NO: 2
  • linker-linked Ig Fusion protein SEQ ID NO: 36
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 37) was called "hPD-1 D-Fc(29)-mIL-15", and the fusion protein dimer containing PD-1 and IL-15 was called "BNS002". collectively. Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • a fusion protein including human PD-1 fragment, long-acting Fc (29) domain and IL-15, signal peptide (SEQ ID NO: 1), PD-1 fragment (SEQ ID NO: 39), Ig hinge to which linker is attached (SEQ ID NO: 3), Fc (29) domain (SEQ ID NO: 4), linker (SEQ ID NO: 5) and IL-15 (SEQ ID NO: 6) in order from the N-terminus to a fusion protein (SEQ ID NO: 40)
  • a polynucleotide containing the coding nucleotide sequence (SEQ ID NO: 42) was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 41) was called "hPD-1 ECD-Fc(29)-hIL-15", and the fusion protein dimer containing PD-1 and IL-15 was called "BNS002". collectively. Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • Signal peptide (SEQ ID NO: 22), IL-15 (SEQ ID NO: 6), and an Ig hinge coupled with a linker to produce a fusion protein including a human IL-15 fragment, a persistent Fc (29) domain, and a PD-1 fragment (SEQ ID NO: 43), an Fc (29) domain (SEQ ID NO: 4), a linker (SEQ ID NO: 44), and a PD-1 fragment (SEQ ID NO: 2) in the above order from the N-terminus (SEQ ID NO: 45)
  • a polynucleotide containing the nucleotide sequence encoding (SEQ ID NO: 47) was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 46) was called "hIL-15-Fc(29)-hPD-1 D", and the fusion protein dimer containing PD-1 and IL-15 was called "BNS002". collectively. Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • Signal peptide (SEQ ID NO: 22), IL-15 (SEQ ID NO: 6), and an Ig hinge coupled with a linker to produce a fusion protein including a human IL-15 fragment, a persistent Fc (29) domain, and a PD-1 fragment (SEQ ID NO: 43), an Fc (29) domain (SEQ ID NO: 4), a linker (SEQ ID NO: 44), and a PD-1 fragment (SEQ ID NO: 39) in the above order from the N-terminus (SEQ ID NO: 48)
  • a polynucleotide containing the nucleotide sequence encoding (SEQ ID NO: 50) was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 49) was called "hIL-15-Fc(29)-hPD-1 ECD", and the fusion protein dimer containing PD-1 and IL-15 was called "BNS002". collectively. Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • Fusion protein including human PD-1 fragment, long-acting Fc (29, H) domain and IL-15, a signal peptide (SEQ ID NO: 1), PD-1 fragment (SEQ ID NO: 2), and a linker are combined.
  • Fusion protein comprising Ig hinge (SEQ ID NO: 3), Fc (29, H) domain (SEQ ID NO: 55), linker (SEQ ID NO: 5) and IL-15 (SEQ ID NO: 6) in the above order from the N-terminus (SEQ ID NO: 55) 56) was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 57) is called "hPD-1 D-Fc (29, H) -hIL-15", and the fusion protein dimer containing PD-1 and IL-15 is called "BNS002". referred to as ". Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • a fusion protein containing a human PD-1 fragment, a persistent Fc (29, H) domain, and a mouse IL-15 fragment a signal peptide (SEQ ID NO: 1), a PD-1 fragment (SEQ ID NO: 2), and a linker are required.
  • a fusion protein comprising a combined Ig hinge (SEQ ID NO: 3), an Fc (29, H) domain (SEQ ID NO: 55), a linker (SEQ ID NO: 5) and IL-15 (SEQ ID NO: 24) in the above order from the N-terminus.
  • a polynucleotide containing the nucleotide sequence (SEQ ID NO: 61) encoding (SEQ ID NO: 59) was synthesized through GenScript's Gene Synthesis service and introduced into pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 60) is called "hPD-1 D-Fc (29, H) -mIL-15", and the fusion protein dimer containing PD-1 and IL-15 is called "BNS002". referred to as ". Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • Signal peptide SEQ ID NO: 1
  • PD-1 fragment SEQ ID NO: 39
  • Ig hinge to which a linker is attached to produce a fusion protein including human PD-1 fragment, long-acting Fc (wt) domain and IL-15 (SEQ ID NO: 3), Fc (wt) domain (SEQ ID NO: 14), linker (SEQ ID NO: 5) and IL-15 (SEQ ID NO: 6) in order from the N-terminus to a fusion protein (SEQ ID NO: 76)
  • a polynucleotide containing the coding base sequence SEQ ID NO: 78
  • GenScript's Gene Synthesis service was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 77) was called "hPD-1 ECD-Fc(wt)-hIL-15", and the fusion protein dimer containing PD-1 and IL-15 was called "BNS002". collectively. Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • Signal peptide (SEQ ID NO: 22), IL-15 (SEQ ID NO: 6), Ig hinge coupled with a linker to produce a fusion protein including a human IL-15 fragment, a persistent Fc (wt) domain, and a PD-1 fragment (SEQ ID NO: 43), Fc (wt) domain (SEQ ID NO: 14), linker (SEQ ID NO: 44) and PD-1 fragment (SEQ ID NO: 2) in the above order from the N-terminus (SEQ ID NO: 79)
  • a polynucleotide containing the nucleotide sequence encoding (SEQ ID NO: 81) was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 80) was called "hIL-15-Fc(wt)-hPD-1 D", and the fusion protein dimer containing PD-1 and IL-15 was called "BNS002". collectively. Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • Signal peptide (SEQ ID NO: 22), IL-15 (SEQ ID NO: 6), Ig hinge coupled with a linker to produce a fusion protein including a human IL-15 fragment, a persistent Fc (wt) domain, and a PD-1 fragment (SEQ ID NO: 43), Fc (wt) domain (SEQ ID NO: 14), linker (SEQ ID NO: 44) and PD-1 fragment (SEQ ID NO: 39) in the above order from the N-terminus (SEQ ID NO: 82)
  • a polynucleotide containing the nucleotide sequence encoding (SEQ ID NO: 84) was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 83) was called "hIL-15-Fc(wt)-hPD-1 ECD", and the fusion protein dimer containing PD-1 and IL-15 was called "BNS002". collectively. Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • a fusion protein including human PD-1 fragment, long-acting Fc (wt, H) domain and IL-15 a signal peptide (SEQ ID NO: 1), PD-1 fragment (SEQ ID NO: 2), and a linker are combined.
  • a fusion protein comprising an Ig hinge (SEQ ID NO: 3), an Fc (wt, H) domain (SEQ ID NO: 88), a linker (SEQ ID NO: 5) and IL-15 (SEQ ID NO: 6) in the above order from the N-terminus (SEQ ID NO: 88) 89) was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 90) was referred to as "hPD-1 D-Fc (wt, H)-hIL-15", and the fusion protein dimer containing PD-1 and IL-15 was designated as "BNS002". referred to as ". Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • a fusion protein containing a human PD-1 fragment, a persistent Fc (wt, H) domain and a mouse IL-15 fragment, a signal peptide (SEQ ID NO: 1), a PD-1 fragment (SEQ ID NO: 2), and a linker are Containing the combined Ig hinge (SEQ ID NO: 3), Fc (wt, H) domain (SEQ ID NO: 88), linker (SEQ ID NO: 5) and mouse IL-15 fragment (SEQ ID NO: 24) in the above order from the N-terminus
  • a polynucleotide containing the nucleotide sequence (SEQ ID NO: 94) encoding the fusion protein (SEQ ID NO: 92) was synthesized through GenScript's Gene Synthesis service and introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 93) is called "hPD-1 D-Fc (wt, H) -mIL-15", and the fusion protein dimer containing PD-1 and IL-15 is called "BNS002". referred to as ". Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • a fusion protein including human PD-1 fragment, long-acting Fc (29) domain and IL-15R ⁇ /IL-15, signal peptide (SEQ ID NO: 1), PD-1 fragment (SEQ ID NO: 2), and linker Combined Ig hinge (SEQ ID NO: 3), Fc (29) domain (SEQ ID NO: 4), linker 1 (SEQ ID NO: 5), IL-15R ⁇ (SEQ ID NO: 95), linker 4 (SEQ ID NO: 96) and IL-15 (
  • a polynucleotide containing the nucleotide sequence (SEQ ID NO: 35) encoding the fusion protein (SEQ ID NO: 34) containing SEQ ID NO: 6) in the above order from the N-terminus was synthesized through Cosmogenetech's Gene Synthesis service, and pcDNA3. 4 TOPO vector (Thermo Fisher Scientific).
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfecting the CHO cells with the expression vector, cultured for 8 days according to the manufacturer's Max Protocol, the culture medium was recovered, and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 33) is referred to as "hPD-1 D-Fc-hIL-15R ⁇ /hIL-15", and thus a fusion protein dimer containing PD-1 and IL-15R ⁇ /IL-15 was collectively referred to as "BNS002S”.
  • the fusion protein in the recovered culture medium was first purified using Affinity Chromatography with Protein A Resin (KANEKA). After binding the fusion protein with DPBS buffer and washing the resin, only the bound fusion protein was eluted with 0.1 M glycine buffer (pH 3.3). The eluted fusion protein was subjected to buffer exchange by dialysis with DPBS buffer for one day, and secondary purification was performed by size exclusion chromatography using a HiLoad ® 16/600 Superdex ® 200 pg column (Cytiva). The separated and purified fusion protein was quantified using NanoDrop, and the purity was analyzed by performing SDS-PAGE and SEC-HPLC analysis.
  • KANEKA Affinity Chromatography with Protein A Resin
  • a fusion protein containing a human PD-1 fragment and a persistent Fc (29) domain a signal peptide (SEQ ID NO: 1), a PD-1 fragment (SEQ ID NO: 2), and an Ig hinge (SEQ ID NO: 3) to which a linker is coupled
  • a polynucleotide containing the nucleotide sequence SEQ ID NO: 100
  • encoding the fusion protein SEQ ID NO: 98
  • Fc (29) K domain SEQ ID NO: 97
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfection into CHO cells with the above expression vector, the cells were cultured for 11 days according to the manufacturer's Max Protocol, and then the culture medium was recovered and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 99) was referred to as "hPD-1 D-Fc", and the fusion protein dimer including PD-1 and Fc was collectively referred to as "BNS001D”. Fusion protein purification and recovery were performed in the same manner as in Preparation Example 17.
  • a signal peptide SEQ ID NO: 22
  • an Ig hinge SEQ ID NO: 57
  • an Fc (29) domain SEQ ID NO: 4
  • a linker SEQ ID NO: 5
  • IL-15 SEQ ID NO: 6
  • a polynucleotide comprising a nucleotide sequence (SEQ ID NO: 54) encoding a fusion protein (SEQ ID NO: 52) was prepared by CosmoGenetech. It was introduced into the pcDNA3.4 TOPO vector (Thermo Fisher Scientific) through Gene Cloning.
  • the expression vector was introduced into CHO cells (ExpiCHO-S TM , Thermo Fisher Scientific) to express the fusion protein. After transfecting the CHO cells with the expression vector, cultured for 10 days according to the manufacturer's Max Protocol, the culture medium was recovered, and the fusion protein was purified.
  • the purified fusion protein (SEQ ID NO: 53) was referred to as "Fc-IL-15", and the fusion protein dimer comprising Fc and IL-15 was collectively referred to as "BNS002I". Purification and recovery of the fusion protein were performed in the same manner as in Preparation Example 1 above.
  • the fusion protein was diluted to a concentration of 0.2, 0.4, 0.6, 0.8, or 1.0 mg/mL.
  • UV absorbance was measured in the wavelength range of 230 nm to 400 nm using a UV spectrophotometer (Little lunatic, Unchained Labs).
  • acid hydrolysis was performed.
  • the hydrolyzed amino acid was derivatized and UPLC (Ultra Performance Liquid Chromatography) was performed using a Waters ACQUITY UPLC system.
  • UPLC Ultra Performance Liquid Chromatography
  • Mobile phase A was started with 5% AccQ-tag A solvent and mobile phase B was AccQ-tag B solvent, starting with an A:B initial ratio of 99.9:0.1, and a gradient system with a ratio of 40.4:59.6 by 7.5 minutes. performed.
  • the sample injection amount was 1 ⁇ l, the flow rate was 0.7 mL/min, and the column temperature was 55°C.
  • the data system used Enpower 3 Software, and the data were fit with the standard solution (I.S.) added in the sample pretreatment step.
  • the extinction coefficient (Extinction coefficinet) was determined by checking the content of the separated and purified BNS002 fusion protein according to the absorbance. The concentration of the fusion protein was confirmed by the extinction coefficient, and it was confirmed that the fusion protein was included at a concentration of 1.99 mg/mL (FIG. 3).
  • the T m , T Agg & T onset analysis was performed.
  • the buffer was exchanged with ultrapure water for the fusion protein, and the sample was prepared to have 1.25 times the protein concentration required for analysis using UNCHAINED LABS Little Lunatic equipment. 1.25 ⁇ of the prepared sample and 5 ⁇ of the formulation buffer were diluted with ultrapure water.
  • the prepared sample was loaded into Uni (UNCHAINED LABS), and T m , T agg & T onset were measured with a Fluorescence & SLS detector at a wavelength of 266 nm from 15 to 95 ° C. Data were analyzed using UNCHAINED LABS software Uncle Analysis V4.01.
  • thermodynamic stability of the isolated and purified BNS002 fusion protein was measured as shown in FIG. 4 .
  • ATR correction is performed on the initially acquired FT-IR spectrum data, and 2300 cm in the subtracted spectrum obtained after correction The water vapor band of the -1 site was removed.
  • a final zero-order FT-IR spectrum was obtained by fitting with a baseline correction and an 11-point Savitzky-Golay function.
  • data in the 1700 to 1600 cm -1 interval were calculated with a linear-fit function to obtain a second derivative spectrum. Peaks were extracted from the second differential spectrum, and the extracted peaks were fitted with a Gaussian function. The size and position of the peak were adjusted with a tolerance of 3% to secure the final extracted spectrum.
  • the secondary structure was assigned according to the wavenumber for the peaks extracted from the second differential spectrum, and the secondary structure content in each sample was calculated by calculating the peak area for the assigned peak.
  • TSK G3000 SW XL , 5 ⁇ m SEC column (Tosoh Bioscience, LLC) and UFLC Shimadzu LC-20AD pump (Shimadzu co.) were used for 40 minutes at a flow rate of 0.5 mL/min.
  • 100 ⁇ l of each sample was separated.
  • 100 mM NaPi, 150 mM NaCl (pH 6.8) was used as a sample running buffer.
  • analysis was performed using a Dawn ® Heleos II MALS detector at 662 nm wavelength and an Optilab T-rEx ® refractive index detector (Wyatt) at 658 nm wavelength. Data were analyzed using Astra software (Wyatt) ver 6.1.5.
  • SEC-MALS Size Exclusion Chomatography - Multi-Angle Laser Light Scattering
  • the binding affinity was measured by surface plasmon resonance (SPR) analysis using a Biacore T200 (GE Healthcare).
  • PD-L1 ligand was diluted to 3-4 ⁇ g/mL in C 2 H 3 NaO 2 (pH 4.0, pH 5.5) and amine coupled on a CM5 sensor chip (GE Healthcare) pre-activated with HBS-EP, pH 7.4. A ring was used to secure it to about 1000 RU. Sensorgrams were recorded by flowing dilutions of BNS002 fusion protein prepared with HBS-EP, pH 7.4, in a range of concentrations from 0.78 nM to 400 nM. The binding between the BNS002 fusion protein and the PD-L1 ligand was measured at a flow rate of 30 ⁇ l/min with an association period of 4 minutes and a dissociation period of 10 minutes. The sensor chip surface was regenerated by injecting 10 mM NaOH between each run. Binding kinetics were analyzed using data analysis software (Ver3.2) and data were fit using a 1:1 binding model.
  • PD-L1 ligand was diluted to 3-4 ⁇ g/mL in C 2 H 3 NaO 2 (pH 4.0, pH 5.5) and amine coupled on a CM5 sensor chip (GE Healthcare) pre-activated with HBS-EP, pH 7.4. A ring was used to secure it to about 1000 RU. Sensorgrams were recorded by flowing dilutions of hPD-1 protein prepared with HBS-EP, pH 7.4, ranging in concentration from 0.78 nM to 400 nM.
  • the association between hPD-L1 ligand and hPD-1 protein was measured at a flow rate of 30 ⁇ l/min with an association period of 4 minutes and a dissociation period of 10 minutes.
  • the sensor chip surface was regenerated by injecting 10 mM NaOH between each run. Binding kinetics were analyzed using data analysis software (Ver3.2) and data were fit using a 1:1 binding model.
  • the binding affinity between the hPD-L1 ligand and the hPD-1 protein was measured as shown in FIG. 8 . It was confirmed that the binding affinity between the BNS002 fusion protein and the hPD-L1 ligand was similar to that between the hPD-1 protein and the hPD-L1 ligand.
  • PD-L2 ligand was diluted to 3-4 ⁇ g/mL in C 2 H 3 NaO 2 (pH 4.0, pH 5.5) and amine coupled on a CM5 sensor chip (GE Healthcare) pre-activated with HBS-EP, pH 7.4. A ring was used to secure it to about 1000 RU. Sensorgrams were recorded by flowing dilutions of BNS002 fusion protein prepared with HBS-EP, pH 7.4, in a range of concentrations from 0.78 nM to 400 nM. The binding between the BNS002 fusion protein and the PD-L2 ligand was measured at a flow rate of 30 ⁇ l/min with an association period of 4 minutes and a dissociation period of 10 minutes. The sensor chip surface was regenerated by injecting 10 mM NaOH between each run. Binding kinetics were analyzed using data analysis software (Ver3.2) and data were fit using a 1:1 binding model.
  • PD-L2 ligand was diluted to 3-4 ⁇ g/mL in C 2 H 3 NaO 2 (pH 4.0, pH 5.5) and amine coupled on a CM5 sensor chip (GE Healthcare) pre-activated with HBS-EP, pH 7.4. A ring was used to secure it to about 1000 RU. Sensorgrams were recorded by flowing dilutions of hPD-1 protein prepared with HBS-EP, pH 7.4, ranging in concentration from 0.78 nM to 400 nM.
  • the association between the hPD-L2 ligand and the hPD-1 protein was measured at a flow rate of 30 ⁇ l/min with an association period of 4 minutes and a dissociation period of 10 minutes.
  • the sensor chip surface was regenerated by injecting 10 mM NaOH between each run. Binding kinetics were analyzed using data analysis software (Ver3.2) and data were fit using a 1:1 binding model.
  • the binding affinity between the hPD-L2 ligand and the hPD-1 protein was measured as shown in FIG. 10 . It was confirmed that the binding affinity between the BNS002 fusion protein and the hPD-L2 ligand was similar to that between the hPD-1 protein and the hPD-L2 ligand.
  • hIL-15R ⁇ human Interleukin-15 receptor alpha
  • C 2 H 3 NaO 2 pH 4.0, pH 5.5
  • HBS-EP pH 7.4.
  • Sensorgrams were recorded by flowing dilutions of BNS002 fusion protein prepared with HBS-EP, pH 7.4 in a range of concentrations from 50 nM to 1600 nM.
  • the binding between the BNS002 fusion protein and hIL-15R ⁇ was measured at a flow rate of 30 ⁇ l/min with an association period of 4 minutes and a dissociation period of 10 minutes.
  • the sensor chip surface was regenerated by injecting 10 mM NaOH between each run. Binding kinetics were analyzed using data analysis software (Ver3.2) and data were fit using a 1:1 binding model.
  • hIL-15R ⁇ was diluted to 3-4 ⁇ g/mL in C 2 H 3 NaO 2 (pH 4.0, pH 5.5) and amine-coupled onto a CM5 sensor chip (GE Healthcare) pre-activated with HBS-EP, pH 7.4. was fixed up to about 1000 RU. Sensorgrams were recorded by flowing hIL-15 protein dilutions prepared with HBS-EP, pH 7.4 in a range of concentrations from 12.5 nM to 200 nM.
  • association between hIL-15R ⁇ and hIL-15 protein was measured at a flow rate of 30 ⁇ l/min with an association period of 4 minutes and a dissociation period of 10 minutes.
  • the sensor chip surface was regenerated by injecting 10 mM NaOH between each run. Binding kinetics were analyzed using data analysis software (Ver3.2) and data were fit using a 1:1 binding model.
  • the binding affinity between hIL-15R ⁇ and hIL-15 protein was measured as shown in FIG. 12 .
  • the binding affinity between the BNS002 fusion protein and hIL-15R ⁇ was confirmed to be 169 to 296 nM, but the binding affinity between the hIL-15 protein and hIL-15R ⁇ was not confirmed.
  • hIL-2R ⁇ human Interleukin-15 receptor beta
  • C 2 H 3 NaO 2 pH 4.0, pH 5.5
  • HBS-EP pH 7.4.
  • Sensorgrams were recorded by flowing dilutions of BNS002 fusion protein prepared with HBS-EP, pH 7.4 in a range of concentrations from 50 nM to 1600 nM.
  • the binding between the BNS002 fusion protein and hIL-2R ⁇ was measured at a flow rate of 30 ⁇ l/min with an association period of 4 minutes and a dissociation period of 10 minutes.
  • the sensor chip surface was regenerated by injecting 10 mM NaOH between each run. Binding kinetics were analyzed using data analysis software (Ver3.2) and data were fit using a 1:1 binding model.
  • hIL-2R ⁇ was diluted to 3-4 ⁇ g/mL in C 2 H 3 NaO 2 (pH 4.0, pH 5.5) and amine-coupled onto a CM5 sensor chip (GE Healthcare) pre-activated with HBS-EP, pH 7.4. was fixed up to about 1000 RU. Sensorgrams were recorded by flowing hIL-15 protein dilutions prepared with HBS-EP, pH 7.4, ranging in concentration from 0.39 nM to 200 nM.
  • association between hIL-2R ⁇ and hIL-15 protein was measured at a flow rate of 30 ⁇ l/min with an association period of 4 minutes and a dissociation period of 10 minutes.
  • the sensor chip surface was regenerated by injecting 10 mM NaOH between each run. Binding kinetics were analyzed using data analysis software (Ver3.2) and data were fit using a 1:1 binding model.
  • the binding affinity between hIL-2R ⁇ and hIL-15 proteins was measured as shown in FIG. 14 .
  • the binding affinity between the BNS002 fusion protein and hIL-2R ⁇ was confirmed to be about 200 times lower than that between the hIL-15 protein and hIL-2R ⁇ .
  • the hFcRn receptor was diluted to 5 ⁇ g/mL in C 2 H 3 NaO 2 (pH 5.0) and pre-activated with 20 mM phosphate, 150 mM NaCl, pH 6.0 and pH 7.4 on a CM5 sensor chip (GE Healthcare). was fixed up to about 1000 RU using amine coupling. Sensorgrams were recorded by flowing diluted fusion protein solutions prepared with 20 mM phosphate, 150 mM NaCl, 0.05% Tween20, pH 6.0 in various concentrations ranging from 3.125 nM to 400 nM. Fusion protein association was measured at a flow rate of 30 ⁇ l/min with an association period of 4 minutes and a dissociation phase of 10 minutes. The sensor chip surface was regenerated by injecting 50 mM NaOH between each run. Binding kinetics were analyzed using data analysis software (Ver3.2) and data were fit using a 1:1 binding model.
  • the binding affinity between the hFcRn receptor and the BNS002 fusion protein was measured as shown in FIG. 15 .
  • the BNS002 fusion proteins the PD-1 D-Fc(29)-IL-15 and PD-1 D-Fc(41)-IL-15 proteins have a higher affinity for the hFcRn receptor than the PD-1 D-Fc(wt)-IL-15 protein. The binding affinity for was confirmed to be strong.
  • the binding affinity was measured by immunological analysis.
  • 0.1 mL of PD-L1-Fc ligand at a concentration of 100 ng/mL was immobilized in a 96-well plate (High binding plate, corning, #CT9018) at 4°C for 16 hours, followed by 250 ⁇ l of blocking buffer. blocked for 2 hours at room temperature. After blocking, the washing process was performed 4 times with 300 ⁇ l of wash buffer. Then, after subdividing the serially diluted BNS002 fusion protein into 50 ⁇ l portions, 50 ⁇ l of the sample diluent was added thereto.
  • 0.1 mL of PD-L2-Fc ligand at a concentration of 100 ng/mL was immobilized in a 96-well plate (High binding plate, corning, #CT9018) at 4°C for 16 hours, and then incubated at room temperature with 250 ⁇ L of blocking buffer. Blocked for an hour. After blocking, the washing process was performed 4 times with 300 ⁇ l of washing buffer. Then, 50 ⁇ l of the serially diluted BNS002 fusion protein was subdivided, and then 50 ⁇ l of the sample dilution was added.
  • IL-15R ⁇ at a concentration of 100 ng/mL was immobilized at 0.1 mL each in a 96-well plate (High binding plate, corning, #CT9018) at 4°C for 16 hours, then blocked with 250 ⁇ l of blocking buffer at room temperature for 2 hours did After blocking, the washing process was performed 4 times with 300 ⁇ l of washing buffer. Then, 50 ⁇ l of the serially diluted BNS002 fusion protein was subdivided, and then 50 ⁇ l of the sample dilution was added.
  • IL-2R ⁇ at a concentration of 100 ng/mL was immobilized at 0.1 mL each in a 96-well plate (High binding plate, corning, #CT9018) at 4°C for 16 hours, then blocked with 250 ⁇ l of blocking buffer at room temperature for 2 hours did After blocking, the washing process was performed 4 times with 300 ⁇ l of washing buffer. Then, 50 ⁇ l of the serially diluted BNS002 fusion protein was subdivided, and then 50 ⁇ l of the sample dilution was added.
  • the binding affinity between the BNS002 fusion protein and the FcRn receptor was measured as shown in FIG. 20 .
  • PathHunter ® eXpress Dimerization Assay Kit Eurofins
  • PathHunter ® eXpress Dimerization Cells stored in liquid nitrogen were warmed with Cell Plating Reagent.
  • SPL 96-well plates
  • chemiluminescence was measured using Synergy Neo2 (BioTek). Based on the measured values, the EC 50 (Half maximal effective concentration; concentration of a drug that can show about half of the maximum effect that the drug can exhibit when the drug is administered) for the fusion protein was confirmed.
  • PBMC peripheral blood mononuclear cells isolated from humans
  • anti-CD3 antibody OKT3, 1 ⁇ g/ml, invitrogen
  • anti-CD28 antibody CD28 .2, 1 ⁇ g/ml, Invitrogen
  • culture medium RPMI1640 medium: FBS 10%, penicillin/streptomycin containing 200 ⁇ l
  • RPMI1640 culture medium FBS 10%, penicillin/streptomycin 200 ⁇ l, anti-CD3/anti-CD28 antibody (1 ⁇ g/each)
  • the BNS002 fusion protein and IL-15 were treated with or without anti-CD3/anti-CD28 antibody (each 1 ⁇ g/ml), and cultured in an incubator for 3 days.
  • PBMCs isolated from humans were thawed at 37°C and centrifuged at 1,500 rpm at 4°C for 5 minutes. After centrifugation, the suspension was removed, and resuspended in RPMI1640 culture medium (FBS 10%, penicillin/streptomycin 200 ⁇ l, BNS002 fusion protein, IL-15 protein (each 10 ⁇ g/ml)) at 37°C, 5% CO 2 cultured in an incubator.
  • the cultured PBMCs were washed twice with cold PBS (Sigma, pH 7.4 ), centrifuged, and then 1.5 cells per 100 ⁇ l (10 6 cells /100 ⁇ l) with PBS (containing 1% FBS). Resuspended in a microtube for ml.
  • the cells were stained with anti-CD8-PE-Texas Red (1:100, invitrogen, cat No. MHCD0817) for 30 minutes at room temperature. After dispensing 30 ⁇ l per well in a V-bottom 96-well plate, the degree of proliferation of these cells was confirmed by measuring the ratio of unlabeled cells among CD8+ T cells by flow cytometry.
  • the BNS002 fusion protein activates the proliferation of CD8+ T cells to a degree similar to that of the IL-15 protein (FIG. 23).
  • PBMCs isolated from humans were thawed at 37°C and centrifuged at 1,500 rpm at 4°C for 5 minutes. After centrifugation, the suspension was removed, and resuspended in RPMI1640 culture medium (FBS 10%, penicillin/streptomycin 200 ⁇ l, BNS002 fusion protein, IL-15 protein (each 10 ⁇ g/ml)) at 37°C, 5% CO 2 cultured in an incubator.
  • the cultured PBMCs were washed twice with cold PBS (Sigma, pH 7.4 ), centrifuged, and then 1.5 cells per 100 ⁇ l (10 6 cells /100 ⁇ l) with PBS (containing 1% FBS). Resuspended in a microtube for ml.
  • the cells were stained with anti-CD4-PE- Pacific Blue (2 ⁇ g/10 6 cells, BioLegend, cat No. 344620) for 30 minutes at room temperature. After dispensing 30 ⁇ l per well in a V-bottom 96-well plate, the degree of proliferation of these cells was confirmed by measuring the ratio of unlabeled cells among CD4+ T cells by flow cytometry.
  • the BNS002 fusion protein activated the proliferation of CD4+ T cells to a degree similar to that of the IL-15 protein, but did not increase the proliferation of CD4+/FoxP3+ Treg cells (FIG. 24).
  • PBMCs isolated from humans were slowly thawed at 37°C and centrifuged at 1,500 rpm at 4°C for 5 minutes. After centrifugation, the suspension was removed, and resuspended in RPMI1640 culture medium (FBS 10%, penicillin/streptomycin 200 ⁇ l, BNS002 fusion protein, IL-15 protein (each 10 ⁇ g/ml)) at 37°C, 5% CO 2 cultured in an incubator. The cultured PBMCs were washed twice with cold PBS (Sigma, pH 7.4 ), centrifuged, and then 1.5 cells per 100 ⁇ l (10 6 cells /100 ⁇ l) with PBS (containing 1% FBS).
  • RPMI1640 culture medium FBS 10%, penicillin/streptomycin 200 ⁇ l, BNS002 fusion protein, IL-15 protein (each 10 ⁇ g/ml)
  • the BNS002 fusion protein activates the proliferation of NK cells to a degree similar to that of the IL-15 protein (FIGS. 25a and 25b).
  • PD-L1 aAPC/CHO-K1 cells (target cells) stored in liquid nitrogen were thawed in a constant temperature water bath at 37° C. for 1 minute, and then suspended in a pre-warmed culture medium (Ham's F12: FBS 10%). 100 ⁇ l per well was dispensed into a 96-well white plate (SPL, cat No. 30196) and cultured in a 37°C, 5% CO 2 incubator.
  • the plate was covered and stored at room temperature. After thawing the effector cells stored in liquid nitrogen for 1 minute in a constant temperature water bath at 37 ° C and suspending them in pre-warmed culture medium (Ham's F12: FBS 10%), the target cells and samples were prepared. 40 ⁇ l was dispensed per each well of the plate and cultured for 6 hours in a 37° C., 5% CO 2 incubator.
  • Bio-Glo reagent was added, being careful not to create bubbles. Bio-Glo reagent was also added to the three edge wells and used as a blank to correct the background signal. After reacting at room temperature for 30 minutes, luminescence was measured with Synergy Neo2 (BioTek).
  • the BNS002 fusion protein binds to PD-1 expressed in effector T cells and activates the function of T cells rather than suppressing them (FIG. 26).
  • PD-L2 aAPC/CHO-K1 cells (target cells) stored in liquid nitrogen were thawed in a constant temperature water bath at 37° C. for 1 minute, and then suspended in a pre-warmed culture medium (Ham's F12: FBS 10%). 100 ⁇ l per well was dispensed into a 96-well white plate (SPL, cat No. 30196) and cultured in a 37°C, 5% CO 2 incubator. The next day, take out the plate, remove 95 ⁇ l of the culture medium, add 40 ⁇ l of BNS002 fusion protein and PD-L2-Fc serially diluted 2 times per well, and Jurkat NFAT-Luc/PD-1 cells (Effector cell).
  • the plate was covered and stored at room temperature. After thawing the effector cells stored in liquid nitrogen for 1 minute in a constant temperature water bath at 37 ° C and suspending them in pre-warmed culture medium (Ham's F12: FBS 10%), the target cells and samples were prepared. 40 ⁇ l was dispensed per each well of the plate and cultured for 6 hours in a 37° C., 5% CO 2 incubator.
  • Bio-Glo reagent was added, being careful not to create bubbles. Bio-Glo reagent was also added to the three edge wells and used as a blank to correct the background signal. After reacting at room temperature for 30 minutes, luminescence was measured with Synergy Neo2 (BioTek).
  • the BNS002 fusion protein binds to PD-1 expressed in effector T cells and activates the T cells instead of suppressing them (FIG. 27).
  • Human melanoma cell line A375 (ATCC, CRL-1619) cells were seeded in 1 ⁇ 10 5 cells in a 6-well plate and cultured in a 37° C., 5% CO 2 incubator. The next day, after removing the culture medium, lentivirus transfected with firefly luciferase (fLuc) and GFP genes (GenTarget, cat No. LVP914-G) (FIG. 28) 25 MOI (Multiplicity of infection, 25 ⁇ l) was added and incubated for 4 hours. After culturing for 4 hours, the cells were recovered with 0.05% Trypsin-EDTA (Gibco) solution, and then transferred to a 150 mm culture dish (TPP) and cultured. During cultivation, only GFP-expressing cells were selectively cultured while observing under a fluorescence microscope, and Firefly luciferase-expressing human melanoma cell line A375-Luc-GFP cell line was established.
  • fLuc firefly
  • PBMC isolated from humans were stained by reacting with membrane-dye (Red) (Sigma, cat No. PKH26) dye at a concentration of 1.25 ⁇ M at room temperature for 1 minute, and then the staining reaction was stopped by adding the same volume of FBS.
  • the dye not bound to the cells was centrifuged at 400 ⁇ g for 10 minutes to remove and wash the suspension, and then resuspended in RPMI1640 culture medium (FBS 10%, penicillin/streptomycin 200 ⁇ l). At this time, a total of three washing operations were performed. After resuspending in RPMI1640 culture medium (FBS 10%, penicillin/streptomycin 200 ⁇ l), fluorescence was observed.
  • A375-fLuc-GFP cells (1 ⁇ 10 5 ) were divided and fluorescently stained PBMC (1 ⁇ 10 6 ) were placed in 6 wells. Divided into plates and co-cultured.
  • the anti-CD3 antibody OKT3, 1 ⁇ g/ml, Invitrogen
  • the anti-CD28 antibody CD28.2, 1 ⁇ g/ml, Invitrogen
  • the BNS002 fusion protein (10 ⁇ g/ml) ml
  • PBMC Malignant melanoma cells A375 (ATCC, # CRL-1619) and PBMC (ATCC, # To confirm the transcription and expression of PD-L1, PD-L2 and PD1 genes in PCS-800-011), reverse transcription PCR was performed.
  • PBMCs were co-stimulated with an anti-CD3 antibody (1 ⁇ g/ml, Invitrogen) and an anti-CD28 antibody (1 ⁇ g/ml, Invitrogen), followed by active culture.
  • Total RNA was extracted from each cell line, cDNA was synthesized (TAKARA, RR036A), PCR was performed using a primer set (Table 1), and PD-L1/L2 and PD-1 specific replication was performed in A375 and PBMC.
  • the products 120 bp, 173 bp, 289 bp) were identified.
  • Primer name order sequence number PD-L1 F 5'-TGGCATTTGCTGAACGCATTT-3' 101 PD-L1 R: 5'-TGCAGCCAGGTCTAATTGTTTT-3' 102 PD-L2 F: 5'-CAGCAATGTGACCCTGGAAT-3' 103 PD-L2 R: 5'-GGACTTGAGGTATTGTGGAACG-3' 104 PD-1 F: 5'-TGCAGCTTCTCCAACACATC-3' 105 PD-1 R: 5'-CTGCCCTTCTCTCTGTCACC-3' 106 GAPDH-2 F: 5'-AGCCGCATCTTCTTTTGCGT-3' 107 GAPDH-2 R: 5'-TGACGAACATGGGGGCATCA-3' 108
  • PD-1-specific replication products 120 bp, 173 bp, 289 bp
  • PD-L1 and PD-L2 genes and immune cells were present in malignant melanoma cells (A375) (FIG. 30).
  • the cancer cell killing effect was statistically more significant in cells treated with BNS002 fusion protein than in untreated cells. It was confirmed that the increase was confirmed (FIG. 33).
  • the A375 cell line was suspended in a culture medium (RPMI1640: FBS 10%, Antibiotic-Antimycotic 1%), dispensed into a 96-well white plate at a cell concentration of 1 ⁇ 10 4 cells/100 ⁇ l, and cultured overnight in an incubator. The next day, after removing 95 ⁇ l of the culture medium from the plate in which the A375 cells were seeded, 25 ⁇ l of ADCC Assay Buffer (ADCC Assay Buffer: RPMI1640 culture medium, low IgG serum 0.25%) prepared in advance was added to effector cells ( NFAT-luc/Fc ⁇ RIIIa) was incubated at room temperature while preparing.
  • ADCC Assay Buffer RPMI1640 culture medium, low IgG serum 0.25%
  • the luminescent activity of the ADCC Assay is a test method that can obtain results only when target cells (A375) with surface antigens (PD-L1/L2), specific antibodies (BNS002 fusion protein), and effector cells expressing Fc ⁇ RIIIa are present.
  • ADCC activity for the fusion protein was measured as shown in FIG. 35 .
  • a frozen mouse tumor cell line specifically colorectal carcinoma cell line CT26 cell line 1 vial, was heat inactivated using RPMI1640 medium containing 10% FBS (Gibco, 10082-147). After thawing, the cells were placed in a flask for cell culture and cultured in an incubator at 37°C and 5% CO 2 . After culturing, the cells were washed with PBS, diluted 10-fold with 2.5% Trypsin-EDTA (Gibco, 15090), and added thereto to separate the cells. After cell separation, the supernatant was removed by centrifugation (1,000 rpm, 5 minutes), and a cell suspension was obtained with a fresh medium.
  • RPMI1640 medium containing 10% FBS (Gibco, 10082-147). After thawing, the cells were placed in a flask for cell culture and cultured in an incubator at 37°C and 5% CO 2 . After culturing, the cells were washed with PBS, diluted 10-fold with 2.
  • cell lines were prepared by diluting in a medium at a concentration of 5.0 ⁇ 10 6 cells/mL.
  • mice 5-week-old male BALB/c mice (Coretech) were used.
  • the cell line was transplanted, it was subcutaneously administered at a dose of 5.0 ⁇ 10 5 cells/0.1 mL/head. Cancer cells were transplanted, and after a certain period of time, the volume of the tumor was measured, and objects reaching about 60-90 mm 3 were isolated, and BNS002 fusion protein was intravenously administered at a dose of 5 mg/kg. A total of 4 administrations were performed once every 3 days after the first administration.
  • PBS was administered as a negative control group
  • atezolizumab an anti-PD-L1 antibody at 5 mg/kg
  • the size of the tumor was measured twice a week (FIG. 36).
  • mice of Experimental Example 29.1 the tumors extracted from only 3 mice per group were placed in RPMI medium containing 10% FBS and then used for FACS analysis.
  • cancer tissues are separated into single cells, and then analyzed for T cells, NK cells, DC (Dendritic Cell), and macrophages using the antibodies listed in Table 2 below was carried out.
  • CD3-efluor ® 450 (Thermo, 48-0031-82) CD45-PerCP-cyanine 5.5 (Thermo, 45-0451-82) CD4-PE-cy7 (Thermo, 25-0041-82) CD8-APC (Thermo, 17-0081-82) CD3-efluor ® 450 (Thermo, 48-0031-82) CD45-Super bright 600 (Thermo, 63-0451-82) CD335-PE (Thermo, 12-3351-82) CD11c-Percp-cy5 (Thermo, 45-0114-82) F4/80-PE-cy7 (Thermo, 25-4801-82) CD49b-APC (Thermo, 17-5971-82) CD11b-APC-eFluor 770 (Thermo, 47-0112-82)
  • a cell line was prepared by diluting in a medium at a concentration of 5.0 ⁇ 10 7 cells/mL.
  • peripheral blood mononuclear cells (PBMC) isolated from humans were actively cultured for one week as in Experimental Example 21.1.
  • PBMC peripheral blood mononuclear cells
  • a cell line was prepared by diluting in a medium at a concentration of 2.5 ⁇ 10 8 cells/mL.
  • a 6-week-old male NXG mouse (Janvier-labs) was used as the mouse, and after a one-week acclimatization process, body weight was measured to match the average weight of each group and regrouped.
  • a cell line was prepared in the same manner as in Experimental Example 30, except that the HCT116 cell line, a colon carcinoma cell line, was used as a frozen human tumor cell line.
  • peripheral blood mononuclear cells (PBMC) isolated from humans were actively cultured for one week as in Experimental Example 21.1. After confirming viability using a microscope, a cell line was prepared by diluting in a medium at a concentration of 2.5 ⁇ 10 8 cells/mL.
  • mice 6-week-old male NSG mice (NOD.Cg-B2mtm1UncPrkdcscidIl2rgtm1Wjl/SzJ) (Orient Bio) were used, and administration of human peripheral blood mononuclear cells 1 ⁇ 10 7 cells (hPBMC)/0.1 mL/head was used to prepare humanized mice. A liquid dose was administered subcutaneously into the tail vein. Humanized mice were identified by measuring hCD45+ expression in the blood by FACS analysis 14 days after administration of human peripheral blood mononuclear cells. After confirming the humanized mouse, when the cell line HCT116 was transplanted, 3 ⁇ 10 6 cells were administered subcutaneously.
  • hPBMC human peripheral blood mononuclear cells
  • BNS002 fusion protein (PD-1 D-Fc-IL-15) at doses of 5, 10, and 20 mg/kg. was administered intraperitoneally. A total of 4 doses were administered once every 3 to 4 days after the first dose. At this time, saline (JW Choongwa Pharmaceutical) was administered as a negative control group, and the size of the tumor was measured twice a week to confirm the anticancer effect (FIG. 43).
  • the size of the tumor was significantly reduced in a concentration-dependent manner, statistically significant, compared to the negative control group (Vehicle treatment) in the animal experiment of humanized mice transplanted with HCT116 cancer cell line treated with the BNS002 fusion protein.
  • the anticancer effect was the best when administered at a dose of 10 mg/kg (FIG. 44).
  • Humanized mice were prepared by preparing the HCT116 cell line and PBMC in the same manner as in Experimental Example 31.1. Two days after transplanting HCT116 cancer cells into humanized mice, they were anesthetized with an inhalational anesthetic (2% isoflurane, Hana Pharmaceutical). Then, BNS002 fusion protein (PD-1 D-Fc-IL-15) was intraperitoneally administered at a dose of 10 mg/kg. A total of 4 doses were administered once every 3 to 4 days after the first dose. At this time, saline was administered as a negative control group, and BNS001D (PD-1 D-Fc) and BNS002I (Fc-IL-15) fusion proteins were administered alone or in combination as a comparative control group.
  • BNS001D PD-1 D-Fc
  • BNS002I Fc-IL-15
  • the size of the tumor in the BNS002 fusion protein-administered group was smaller than that of the comparative control groups, BNS001D (PD-1 D-Fc) and BNS002I (Fc-IL-15)-administered groups.
  • BNS001D PD-1 D-Fc
  • BNS002I Fc-IL-15
  • the BNS002 fusion protein administration group had a smaller tumor size than the BNS001D (PD-1 D-Fc) + BNS002I (Fc-IL-15) combination administration group (FIG. 47).
  • the BNS002 fusion protein administration group was statistically significant compared to the commercial antibodies Pembrolizumab (Anti-PD-1 (Keytruda, MSD)) and Avelumab (Anti-PD-L1 (Bavencio, Merck)) administration groups used as positive control groups. It was confirmed that the size of the tumor was significantly reduced. Compared to the atezolizumab (Anti-PD-L1 (Tecentriq, Roche)) administration group, it was confirmed that the BNS002 fusion protein showed an equivalent level of tumor size reduction (FIG. 49).
  • Humanized mice were prepared by preparing the HCT116 cell line and PBMC in the same manner as in Experimental Example 31.1. Two days after transplanting HCT116 cancer cells into humanized mice, they were anesthetized with an inhalational anesthetic (2% isoflurane, Hana Pharmaceutical). Then, the BNS002S fusion protein (PD-1D-Fc-IL-15/IL-15Ra) was intraperitoneally administered at a dose of 5 mg/kg. A total of 4 doses were administered once every 3 to 4 days after the first dose. At this time, the negative control group, comparative control group, and positive control group were administered under the same conditions as Experimental Example 31.2. Thereafter, tumor size and weight were measured twice a week to confirm the anticancer effect of the administered material (FIG. 46).
  • BNS001D PD-1 D-Fc
  • BNS002I Fc-IL-15
  • the BNS002S fusion protein-administered group had a tumor size compared to the commercially available antibodies Pembrolizumab (Anti-PD-1 (Keytruda, MSD)) and Avelumab (Anti-PD-L1 (Bavencio, Merck))-administered groups used as positive control groups. It was confirmed that it was significantly reduced. Compared to the atezolizumab (Anti-PD-L1 (Tecentriq, Roche)) administration group, it was confirmed that the BNS002S fusion protein showed an equivalent level of tumor size reduction (FIG. 53).
  • a humanized mouse was prepared by preparing the A549 cell line and PBMC in the same manner as in Experimental Example 31.1, except that the A549 cell line, which is a lung cancer (NSCLC) cell line, was used as a frozen human tumor cell line.
  • NSCLC lung cancer
  • saline JW Pharmaceutical
  • saline JW Pharmaceutical
  • atezolizumab Anti-PD-L1 (Tecentriq, Roche)
  • avelumab Avelumab
  • Anti-PD-L1 Tecentriq, Roche
  • L1 Bovencio, Merck
  • pembrolizumab Anti-PD-1 (Keytruda, MSD)
  • tumor size and body weight were measured twice a week (FIG. 55).
  • the size of the tumor was significantly reduced statistically significantly compared to the negative control group in the animal experiment of humanized mice transplanted with the A549 cancer cell line treated with the BNS002 fusion protein (PD-1 D-Fc-IL-15).
  • the BNS002 fusion protein administration group was commercially available antibodies atezolizumab (Anti-PD-L1 (Tecentriq, Roche)), pembrolizumab (Anti-PD-1 (Keytruda, MSD)) and avelumab (Anti-PD-L1 (Keytruda, MSD)) used as a positive control group.
  • PD-L1 (Bavencio, Merck)) showed an equivalent level of tumor size reduction compared to the administration group (FIG. 56).
  • Humanized mice were prepared by preparing the A549 cell line and PBMC in the same manner as in Experimental Example 32.1. Two days after transplanting A549 cancer cells into humanized mice, after anesthesia with inhalation anesthetic (2% isoflurane, Hana Pharmaceutical), BNS002S (PD-1D-Fc-IL-15/IL) was administered at doses of 1 and 5 mg/kg. -15Ra) fusion protein was intraperitoneally administered. A total of 4 doses were administered once every 3 to 4 days after the first dose. At this time, saline was administered as a negative control group, and tumor size and body weight were measured twice a week to confirm the anticancer effect of the administered material.
  • BNS002S PD-1D-Fc-IL-15/IL
  • the tumor size was statistically significant compared to the negative control group (Vehicle treatment) in a concentration-dependent manner. It was confirmed that it was significantly reduced. In particular, the anticancer effect was the best when administered at a dose of 5 mg/kg (FIG. 58).
  • mice Humanized mice were prepared by preparing the A549 cell line and PBMC in the same manner as in Experimental Example 32.1. Two days after transplanting A549 cancer cells into humanized mice, they were anesthetized with an inhalational anesthetic (2% isoflurane, Hana Pharmaceutical). Then, the BNS002S fusion protein was intraperitoneally administered at a dose of 5 mg/kg. A total of 4 doses were administered once every 3 to 4 days after the first dose. At this time, the negative control group and the positive control group were administered under the same conditions as Experimental Example 32.1. Tumor size and body weight were measured twice a week to confirm the anticancer effect of the administered material.
  • an inhalational anesthetic 2% isoflurane, Hana Pharmaceutical
  • the size of the tumor was significantly reduced statistically compared to the negative control group in the animal experiment of humanized mice transplanted with the A549 cancer cell line treated with the BNS002S fusion protein (PD-1D-Fc-IL-15/IL-15Ra).
  • the BNS002S fusion protein administration group was commercially available antibodies atezolizumab (Anti-PD-L1 (Tecentriq, Roche)), pembrolizumab (Anti-PD-1 (Keytruda, MSD)) and avelumab (Anti-PD-L1 (Keytruda, MSD)) used as a positive control group.
  • PD-L1 (Bavencio, Merck) showed an equivalent level of tumor size reduction compared to the administration group (FIG. 60). On the other hand, it was confirmed that there was no effect on body weight change according to the BNS002S fusion protein treatment (FIG. 61).
  • the MDA-MB-231 cell line and PBMC were prepared in the same manner as in Experimental Example 31.1 to prepare a humanized mouse.
  • TNBC breast carcinoma
  • MDA-MB-231 cancer cells were anesthetized with an inhalational anesthetic (2% isoflurane, Hana Pharmaceutical).
  • BNS002 fusion protein (PD-1 D-Fc-IL-15) was intraperitoneally administered at a dose of 10 mg/kg. A total of 4 doses were administered once every 3 to 4 days after the first dose.
  • saline JW Pharmaceutical
  • saline JW Pharmaceutical
  • atezolizumab Anti-PD-L1 (Tecentriq, Roche)
  • avelumab Avelumab; Anti-PD-L1 (Tecentriq, Roche)
  • L1 Bovencio, Merck
  • pembrolizumab Anti-PD-1 (Keytruda, MSD)
  • tumor size and body weight were measured twice a week (FIG. 62).
  • mice Humanized mice were prepared by preparing the MDA-MB-231 cell line and PBMC in the same manner as in Experimental Example 33.1. Two days after transplanting MDA-MB-231 cancer cells into humanized mice, they were anesthetized with an inhalational anesthetic (2% isoflurane, Hana Pharmaceutical). Then, the BNS002S fusion protein was intraperitoneally administered at a dose of 5 mg/kg. A total of 4 doses were administered once every 3 to 4 days after the first dose. At this time, the negative control group and the positive control group were administered under the same conditions as Experimental Example 33.1. Tumor size and body weight were measured twice a week to confirm the anticancer effect of the administered material.
  • an inhalational anesthetic 2% isoflurane, Hana Pharmaceutical
  • the size of the tumor was statistically significant compared to the negative control group. confirmed a decrease.
  • the BNS002S fusion protein administration group was commercially available antibodies atezolizumab (Anti-PD-L1 (Tecentriq, Roche)), pembrolizumab (Anti-PD-1 (Keytruda, MSD)) and avelumab (Anti-PD-L1 (Keytruda, MSD)) used as positive control groups.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne une protéine de fusion comprenant la protéine PD-1 et la protéine IL-15, et une utilisation associée. La protéine de fusion comprenant PD-1, IL-15, et un Fc persistant dans le sérum prolongé selon un mode de réalisation spécifique peuvent activer des cellules immunitaires telles que des cellules NK et augmenter efficacement la demi-vie des cellules immunitaires dans le corps. Par conséquent, une composition pharmaceutique contenant la protéine de fusion en tant que principe actif augmente l'activité immunitaire dans le corps et peut donc être utilisée en tant que médicament anticancéreux pour des patients qui ne répondent pas à un traitement inhibiteur de point de contrôle immunitaire et des patients en récidive.
PCT/KR2022/014304 2021-09-24 2022-09-23 Dimère de protéine de fusion comprenant pd-1 et il-15 et utilisation associée WO2023048518A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20210126053 2021-09-24
KR10-2021-0126053 2021-09-24

Publications (1)

Publication Number Publication Date
WO2023048518A1 true WO2023048518A1 (fr) 2023-03-30

Family

ID=85720953

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2022/014304 WO2023048518A1 (fr) 2021-09-24 2022-09-23 Dimère de protéine de fusion comprenant pd-1 et il-15 et utilisation associée

Country Status (2)

Country Link
KR (1) KR20230044131A (fr)
WO (1) WO2023048518A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190060821A (ko) * 2016-10-14 2019-06-03 젠코어 인코포레이티드 IL-15/IL-15Rα FC-융합 단백질 및 PD-1 항체 단편을 포함하는 이중특이성 이종이량체 융합 단백질
KR20190123723A (ko) * 2017-01-05 2019-11-01 카 메디컬 리미티드 Pd1-41bbl 융합 단백질 및 이의 이용 방법
KR20210058902A (ko) * 2018-09-14 2021-05-24 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 sPD-1 변이체-FC 융합 단백질
WO2021119429A1 (fr) * 2019-12-13 2021-06-17 Cugene Inc. Nouvelles protéines de fusion d'interleukine 15 (il-15) et utilisations de celles-ci

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101957431B1 (ko) 2018-01-10 2019-03-12 국민대학교 산학협력단 혈중 반감기 연장을 위한 항체 Fc 변이체들

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190060821A (ko) * 2016-10-14 2019-06-03 젠코어 인코포레이티드 IL-15/IL-15Rα FC-융합 단백질 및 PD-1 항체 단편을 포함하는 이중특이성 이종이량체 융합 단백질
KR20190123723A (ko) * 2017-01-05 2019-11-01 카 메디컬 리미티드 Pd1-41bbl 융합 단백질 및 이의 이용 방법
KR20210058902A (ko) * 2018-09-14 2021-05-24 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 sPD-1 변이체-FC 융합 단백질
WO2021119429A1 (fr) * 2019-12-13 2021-06-17 Cugene Inc. Nouvelles protéines de fusion d'interleukine 15 (il-15) et utilisations de celles-ci

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
XU YUANMING, CARRASCOSA LUCIA CAMPOS, YEUNG YIK ANDY, CHU MATTHEW LING-HON, YANG WENJING, DJURETIC IVANA, PAPPAS DANIELLE C., ZEYT: "An Engineered IL15 Cytokine Mutein Fused to an Anti-PD1 Improves Intratumoral T-cell Function and Antitumor Immunity", CANCER IMMUNOLOGY RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 9, no. 10, 1 October 2021 (2021-10-01), US , pages 1141 - 1157, XP093054460, ISSN: 2326-6066, DOI: 10.1158/2326-6066.CIR-21-0058 *

Also Published As

Publication number Publication date
KR20230044131A (ko) 2023-04-03

Similar Documents

Publication Publication Date Title
WO2020060122A1 (fr) Protéine de fusion comprenant une protéine il-2 et une protéine cd80 et utilisation associée
WO2017116205A1 (fr) Conjugué persistant d'un triple activateur activant le récepteur du glucagon, du glp-1 et du gip
WO2017034335A1 (fr) Protéine recombinante (icp)-socs3 perméable dans les cellules améliorée et ses utilisations
WO2018194381A1 (fr) Molécule de liaison spécifique à protéine lrig-1 et son utilisation
WO2018030806A1 (fr) Cytokine fusionnée à un hétérodimère fc d'immunoglobuline et composition pharmaceutique la contenant
WO2015108398A1 (fr) Insuline à action prolongée et utilisation associée
WO2021107689A1 (fr) Composition pharmaceutique pour le traitement d'un cancer, comprenant un inhibiteur de point de contrôle immunitaire et une protéine de fusion constituée de la protéine il-2 et de la protéine cd80
WO2020050667A1 (fr) Récepteur antigénique chimérique pour un cancer solide et lymphocytes t exprimant ce récepteur chimérique de l'antigène
WO2019135668A1 (fr) Domaine extracellulaire de sous-unité alpha du récepteur fc ige, composition pharmaceutique le comprenant et son procédé de production
WO2019066586A1 (fr) Conjugué à action prolongée du dérivé de peptide-2 apparenté au glucagon (glp-2)
WO2011142514A1 (fr) Composition contenant du pias3 comme ingrédient actif pour la prévention ou le traitement d'un cancer ou d'une maladie immune
WO2017116207A1 (fr) Analogue de fgf21, conjugué de fgf21 et leur utilisation
WO2020080908A1 (fr) Anticorps anti-l1cam ou fragment de liaison à l'antigène de celui-ci, et récepteur antigénique chimérique le comprenant
WO2017030323A1 (fr) Protéine recombinante perméable à la cellule (cp)-δsocs3 et utilisations de ladite protéine
WO2019066570A1 (fr) Analogue d'insuline monocaténaire à action prolongée et conjugué de celui-ci
WO2021187922A1 (fr) Composition pharmaceutique pour le traitement du cancer comprenant une protéine de fusion comprenant une protéine il-2 et une protéine cd80 et un médicament anticancéreux
WO2021158073A1 (fr) Anticorps de fusion permettant la présentation d'un épitope d'antigène de lymphocyte t dérivé d'un antigène ou d'un peptide le contenant sur une surface cellulaire, et composition le comprenant
WO2020080854A1 (fr) Molécule de liaison spécifique de la protéine lrig-1 et son utilisation
WO2022005174A1 (fr) Protéine de fusion comprenant un anticorps anti-lag-3 et il-2 et son utilisation
WO2021153979A1 (fr) Protéine de fusion comprenant un anticorps anti-taa, un anticorps anti-pd-l1 et il-2 et les utilisations correspondantes
WO2022035200A1 (fr) Protéine de fusion comprenant il-12 et un anticorps anti-cd20 et utilisation associée
WO2023048518A1 (fr) Dimère de protéine de fusion comprenant pd-1 et il-15 et utilisation associée
WO2023048516A1 (fr) Dimère de protéine de fusion comprenant pd-1 et il-21, et son utilisation
WO2022139493A1 (fr) NOUVEAU PEPTIDE POUVANT INHIBER LA SIGNALISATION DU TGF-β ET SON UTILISATION
WO2021187904A1 (fr) Protéine de fusion comprenant une protéine il-2 et une protéine cd80 et utilisations associées

Legal Events

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

Ref document number: 22873221

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE