[go: up one dir, main page]

WO2024005422A1 - Variants de b7-h6 à affinité de liaison améliorée pour nkp30 - Google Patents

Variants de b7-h6 à affinité de liaison améliorée pour nkp30 Download PDF

Info

Publication number
WO2024005422A1
WO2024005422A1 PCT/KR2023/008393 KR2023008393W WO2024005422A1 WO 2024005422 A1 WO2024005422 A1 WO 2024005422A1 KR 2023008393 W KR2023008393 W KR 2023008393W WO 2024005422 A1 WO2024005422 A1 WO 2024005422A1
Authority
WO
WIPO (PCT)
Prior art keywords
variant
amino acid
nkp30
binding affinity
acid substitutions
Prior art date
Application number
PCT/KR2023/008393
Other languages
English (en)
Korean (ko)
Inventor
정상택
김보미
하지연
Original Assignee
고려대학교 산학협력단
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 고려대학교 산학협력단 filed Critical 고려대학교 산학협력단
Publication of WO2024005422A1 publication Critical patent/WO2024005422A1/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
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Definitions

  • the present invention relates to B7-H6 variants with improved binding to NKp30.
  • Cancer cells express immune checkpoint proteins on the cell surface, which are used by normal cells to suppress immune cell activation in order to avoid the killing mechanism of immune cells.
  • immunotherapy has been used as a method to treat cancer.
  • Research on checkpoint inhibitor proteins is actively underway. Patients who respond to immune checkpoint inhibitors have fewer side effects during the treatment process, and their use is increasing explosively as superior treatment effects are reported compared to existing anticancer drugs for various cancer types. However, more than half of patients still receive clinically approved immunizations.
  • Immune cells express immune checkpoint proteins on the cell surface that are used to suppress and activate themselves. Among various immune checkpoint receptors, it was confirmed that B7-H6 binds to NKp30, an activation receptor expressed on the surface of NK cells (Natural killer cells), thereby activating NK cells.
  • cell therapy is a medicine that induces regeneration using living cells to restore damaged or diseased cells/tissues, using physical, chemical, and biological methods such as culturing, proliferating, or selecting autologous, allogeneic, or xenogeneic cells in vitro. It refers to medicines manufactured by manipulating.
  • immunomodulatory cell therapy refers to medicines used to treat diseases by activating the body's immune response using immune cells such as dendritic cells, natural killer cells, and T cells. .
  • immunomodulatory cell therapies are mainly being developed for cancer treatment indications. Since they achieve therapeutic effects by activating immune function by administering immune cells directly to the patient, they are used in existing cancer treatments such as surgery, anticancer drugs, or radiation therapy.
  • Immunomodulatory cell therapy activates various immune cells, such as PBMC (pheripheral blood mononuclear cells), T cells, and natural killer cells (NK cells), isolated from patients through apheresis, with various antibodies and cytokines, and then administers them in vitro.
  • PBMC peripheral blood mononuclear cells
  • NK cells natural killer cells
  • TCR T-Cell Receptor
  • CAR Chimeric Antigen Receptor
  • PBMC peripheral blood mononuclear cells
  • CD3 - cells CD3-CD56 + cells
  • CD56 + cells are used as raw material cells
  • natural killer cell growth factors include IL-2, IL-12, IL-15, and IL.
  • Cytokines such as -21, LPS (Goodier et al., J. Immunol. 165(1):139-147, 2000), and OKT-3 antibody that stimulates CD3 (Condiotti et al., Experimental Hematol. 29( 1):104-113, 2001), but the proliferation rate is such that it is difficult to commercialize natural killer cells as a treatment.
  • the purpose of the present invention is to provide a B7-H6 variant with increased binding ability to natural killer cells.
  • an object of the present invention is to provide a natural killer cell activator.
  • an object of the present invention is to provide bispecific or multispecific antibodies.
  • an object of the present invention is to provide a pharmaceutical composition for treating or preventing cancer.
  • an object of the present invention is to provide a method for in vitro proliferation of activated natural killer cells.
  • an object of the present invention is to provide a use of the B7-H6 variant or a bispecific or multispecific antibody containing it for the prevention or treatment of cancer.
  • an object of the present invention is to provide a method for treating cancer.
  • the present invention provides a B7-H6 variant with increased binding affinity to NKp30 (Natural cytotoxicity triggering receptor 3).
  • the present invention provides a natural killer cell activator comprising the B7-H6 variant.
  • the present invention provides a bispecific or multispecific antibody comprising a portion that binds to the B7-H6 variant and a target antigen.
  • the present invention provides a pharmaceutical composition for the treatment or prevention of cancer comprising the B7-H6 variant or a bispecific or multispecific antibody.
  • the present invention provides a method for in vitro proliferation of activated natural killer cells.
  • the present invention provides a use of the B7-H6 variant, or bispecific or multispecific antibody, for preventing or treating cancer.
  • the present invention provides a method of treating cancer comprising administering the B7-H6 variant, or bispecific or multispecific antibody, in a pharmaceutically effective amount to an individual suffering from cancer.
  • the B7-H6 variant of the present invention has a significantly increased binding force to NKp30, an activation receptor for natural killer cells (NK cells), compared to the wild type, which can increase natural killer cell activation, and its size is significantly smaller than that of the antibody, so it can be used in the tumor microenvironment. Since it is easy to penetrate and produce, it can be usefully used alone or in combination with various immunotherapeutics to treat cancer or infectious diseases.
  • NK cells an activation receptor for natural killer cells
  • Figure 1 is a diagram showing an SDS-PAGE gel photograph of an expression vector for NKp30-streptavidin protein and purified protein.
  • Figure 2 is a diagram showing the results of flow cytometry for selecting the B7-H6 display method.
  • Figure 3 is a diagram showing the results of amino acid sequence analysis of the constructed initial library.
  • Figure 4 is a diagram showing the results of enrichment verification of clones with improved NKp30 binding ability according to the screening process using flow cytometry.
  • Figure 5 is a diagram showing the results of analysis of the binding affinity of B7-H6 variants with NKp30 obtained through flow cytometry.
  • Figure 6 is a diagram showing an SDS-PAGE gel photograph of purified B7-H6 variant-Fc fusion proteins.
  • Figure 7 is a diagram showing the results of ELISA analysis of the NKp30 binding capacity of B7-H6 variant-Fc fusion proteins.
  • Figure 8 is a diagram showing the results of amino acid sequence analysis of the constructed B7-H6 focused library.
  • Figure 9 is a diagram showing the results of verifying the amplification of B7-H6 variant clones with high NKp30 affinity according to the screening process using flow cytometry.
  • Figure 10 is a diagram showing the results of analysis of the binding affinity of B7-H6 variants to NKp30 using flow cytometry.
  • Figure 11 is a diagram showing an SDS-PAGE gel photograph of purified B7-H6 variant-Fc fusion proteins.
  • Figure 12 is a diagram showing the results of ELISA analysis of the NKp30 binding ability of the B7-H6 variant-Fc fusion proteins of the present invention.
  • amino acids referred to by abbreviations in the present invention are described according to the IUPAC-IUB nomenclature as follows:
  • the present invention relates to the 31st, 32nd, 37th, 40th, 51st, 53rd, 57th, 60th, One or more amino acids selected from the group consisting of the 67th, 86th, 101st, 102nd, 108th, 114th, 129th, 136th, 142nd and 143rd amino acids are substituted with a sequence different from the wild type amino acid. , relates to a B7-H6 variant or fragment thereof with increased binding to NKp30 (Natural cytotoxicity triggering receptor 3).
  • the amino acid of wild-type B7-H6 may include the amino acid sequence of SEQ ID NO: 1, and the amino acid position may be based on the amino acid sequence of SEQ ID NO: 1.
  • the B7-H6 variant of the invention is M31I, A32T, I37T, I37F, L40Q, F51I, F51S, F51L, F51Y, F51T, F51H, F51Q, F51K, F51R, S53G, N57D, S60I, S60Y, S60T , S60H, S60L, W67R, Q86L, K101E, S102C, S102R, R108M, L114M, L129M, Q136R, S142N and P143S. It may include any one or more amino acid substitutions selected from the group consisting of.
  • the B7-H6 variant of the present invention may be a variant comprising an amino acid mutation in the ectodomain region (SEQ ID NO: 2), which is a region exposed to the outside of the cell of all amino acids of wild-type B7-H6. .
  • the B7-H6 variant of the present invention may be B5 containing the amino acid substitutions F51S and S60I, and may include an extracellular domain region (SEQ ID NO: 3) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing the nucleotide sequence of 4.
  • the B7-H6 variant of the present invention may be B6 containing the amino acid substitution N57D, and may include an extracellular domain region (SEQ ID NO: 5) containing the amino acid substitution, which is of SEQ ID NO: 6. It can be encoded as a nucleic acid molecule containing a base sequence.
  • the B7-H6 variant of the present invention may be B7 containing the amino acid substitutions K101E and S102R, and may include an extracellular domain region (SEQ ID NO: 7) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing the nucleotide sequence of 8.
  • the B7-H6 variant of the present invention may be B8 containing the amino acid substitutions A32T and S60I, and may include an extracellular domain region (SEQ ID NO: 9) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 10 base sequences.
  • the B7-H6 variant of the present invention may be B9 containing amino acid substitutions A32T, L40Q and S60I, and may include an extracellular domain region (SEQ ID NO: 11) containing the amino acid substitutions, which It can be encoded as a nucleic acid molecule containing the base sequence of SEQ ID NO: 12.
  • the B7-H6 variant of the present invention may be B14 containing the amino acid substitutions F51L and S60I, and may include an extracellular domain region (SEQ ID NO: 13) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 14 base sequences.
  • the B7-H6 variant of the present invention may be B16 containing the amino acid substitution K101E, and may include an extracellular domain region (SEQ ID NO: 15) containing the amino acid substitution, which is of SEQ ID NO: 16 It can be encoded as a nucleic acid molecule containing a base sequence.
  • the B7-H6 variant of the present invention may be B19 comprising amino acid substitutions A32T, W67R, Q86L, K101E and L129M, and may include an extracellular domain region (SEQ ID NO: 17) comprising the amino acid substitutions. It can be encoded with a nucleic acid molecule containing the nucleotide sequence of SEQ ID NO: 18.
  • the B7-H6 variant of the present invention may be B23 containing amino acid substitutions S60I, L114M and S142N, and may include an extracellular domain region (SEQ ID NO: 19) containing the amino acid substitutions, which It can be encoded as a nucleic acid molecule containing the base sequence of SEQ ID NO: 20.
  • the B7-H6 variant of the present invention may be B29 containing amino acid substitutions M31I, A32T, F51I, S60I, S102C and R108M, and the extracellular domain region (SEQ ID NO: 21) containing the amino acid substitutions. It may be encoded by a nucleic acid molecule containing the nucleotide sequence of SEQ ID NO: 22.
  • the B7-H6 variant of the present invention may be B35 containing amino acid substitutions A32T, S60I, K101E and P143S, and may include an extracellular domain region (SEQ ID NO: 23) containing the amino acid substitutions, , which can be encoded as a nucleic acid molecule containing the base sequence of SEQ ID NO: 24.
  • the B7-H6 variant of the present invention may be B40 containing the amino acid substitutions I37T, Q86L and K101E, and may include an extracellular domain region (SEQ ID NO: 25) containing the amino acid substitutions, which It can be encoded as a nucleic acid molecule containing the base sequence of SEQ ID NO: 26.
  • the B7-H6 variant of the present invention may be B41 containing amino acid substitutions A32T, S53G, S60I and Q136R, and may include an extracellular domain region (SEQ ID NO: 27) containing the amino acid substitutions, , which can be encoded as a nucleic acid molecule containing the base sequence of SEQ ID NO: 28.
  • SEQ ID NO: 27 extracellular domain region
  • the B7-H6 variant of the present invention may be B47 containing the amino acid substitutions F51Y and S60I, and may include an extracellular domain region (SEQ ID NO: 29) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 30 base sequences.
  • the B7-H6 variant of the present invention may be B52 containing the amino acid substitutions S60I and K101E, and may include an extracellular domain region (SEQ ID NO: 31) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 32 base sequences.
  • the B7-H6 variant of the present invention may be B53 containing the amino acid substitutions A32T and K101E, and may include an extracellular domain region (SEQ ID NO: 33) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 34 base sequences.
  • the B7-H6 variant of the present invention may be B54 containing amino acid substitutions A32T, S60I and K101E, and may include an extracellular domain region (SEQ ID NO: 35) containing the amino acid substitutions, which It can be encoded as a nucleic acid molecule containing the base sequence of SEQ ID NO: 36.
  • the B7-H6 variant of the present invention may be BF2 containing the amino acid substitutions F51H and S60I, and may include an extracellular domain region (SEQ ID NO: 37) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 38 base sequences.
  • the B7-H6 variant of the present invention may be BF3 containing amino acid substitutions F51I and S60Y, and may include an extracellular domain region (SEQ ID NO: 39) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 40 base sequences.
  • the B7-H6 variant of the present invention may be BF5 containing amino acid substitutions I37F, F51L and S60T, and may include an extracellular domain region (SEQ ID NO: 41) containing the amino acid substitutions, which It can be encoded as a nucleic acid molecule containing the base sequence of SEQ ID NO: 42.
  • the B7-H6 variant of the present invention may be BF8 containing amino acid substitutions F51T and S60T, and may include an extracellular domain region (SEQ ID NO: 43) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 44 base sequences.
  • the B7-H6 variant of the present invention may be BF11 containing amino acid substitutions F51L and S60H, and may include an extracellular domain region (SEQ ID NO: 45) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 46 base sequences.
  • the B7-H6 variant of the present invention may be BF19 containing the amino acid substitutions F51T and S60Y, and may include an extracellular domain region (SEQ ID NO: 47) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 48 base sequences.
  • the B7-H6 variant of the present invention may be BF25 containing the amino acid substitutions F51Q and S60H, and may include an extracellular domain region (SEQ ID NO: 49) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 50 base sequences.
  • the B7-H6 variant of the present invention may be BF39 containing amino acid substitutions F51K and S60L, and may include an extracellular domain region (SEQ ID NO: 51) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 52 base sequences.
  • the B7-H6 variant of the present invention may be BF46 containing amino acid substitutions F51R and S60T, and may include an extracellular domain region (SEQ ID NO: 53) containing the amino acid substitutions, which is SEQ ID NO: It can be encoded as a nucleic acid molecule containing 54 base sequences.
  • the amino acid substitution position is described based on the total amino acids of wild-type B7-H6 (SEQ ID NO: 1), but the actual mutation position is located in the extracellular domain region (SEQ ID NO: 2), so it is based on the total amino acids of wild-type B7-H6.
  • the position described may be different from the position in the extracellular domain region containing the amino acid substitution.
  • the F51S amino acid substitution of the B5 variant is F27I based on the amino acid of SEQ ID NO: 3.
  • the B7-H6 variant of the present invention refers to a substitution of some amino acid sequences in the wild-type B7-H6 protein (or peptide), and as used in the present invention, the term “variant” refers to at least one amino acid difference ( refers to the corresponding amino acid sequence including substitutions, insertions or deletions). In certain embodiments, a “variant” has high amino acid sequence homology and/or conservative amino acid substitutions, deletions and/or insertions when compared to a reference sequence. Additionally, as used in the present invention, the term “B7-H6 variant” refers to a B7-H6 variant protein mutated in one or more amino acids to regulate its binding activity with NKp30.
  • the B7-H6 variant of the invention can be prepared by standard synthetic methods, recombinant expression systems, or any other art method. Accordingly, peptides according to the invention can be synthesized by a number of methods, including, for example, methods including:
  • a gene encoding the B7-H6 variant of the present invention can be prepared through genetic manipulation, transformed into a host cell, and then expressed to produce the B7-H6 variant of the present invention.
  • the present invention relates to a nucleic acid molecule encoding the B7-H6 variant of the present invention, a vector containing the same, and a host cell containing the vector.
  • nucleic acid molecule used in the present invention refers to deoxyribonucleotides or ribonucleotides that exist in single-stranded or double-stranded form, and includes natural nucleic acid analogues unless specifically stated otherwise (Scheit, Nucleotide Analogs, John Wiley , New York (1980); Uhlman and Peyman, Chemical Reviews, 90:543-584 (1990)).
  • vector refers to any nucleic acid containing a competent nucleotide sequence that is inserted into a host cell and recombines with and integrates into the host cell genome, or replicates spontaneously as an episome.
  • vectors include linear nucleic acids, plasmids, phagemids, cosmids, RNA vectors, viral vectors, etc.
  • host cell refers to a eukaryotic or prokaryotic cell into which one or more DNA or vectors are introduced, and should be understood to refer not only to a specific target cell but also to its descendants or potential descendants.
  • progeny are not identical to the parent cell since certain modifications may occur in subsequent generations due to mutations or environmental influences, but are still included within the scope of the term as used herein.
  • the present invention relates to a natural killer cell (NK cell) activator comprising the B7-H6 variant of the present invention or a fragment thereof.
  • NK cell natural killer cell
  • the fragment may be the ectodomain region of the B7-H6 variant of the present invention.
  • the present invention relates to a composition for detecting natural killer cells, comprising the B7-H6 variant of the present invention.
  • the composition can detect and quantify NKp30 protein expressed on the surface of natural killer cells.
  • the B7-H6 variant may be labeled with one selected from the group consisting of a chromogenic enzyme, a radioactive isotope, a chromophore, a luminescent substance, and a fluorescent substance
  • the fluorescent substance is a Cy (cyanine) series
  • It may be a Rhodamine series, Alexa series, BODIPY series or ROX series fluorescent substance, such as Nile Red, BODIPY, 4,4-difluoro-4-bora-3a,4a -diaza-s-indacene), cyanine, fluorescein, rhodamine, coumarine, or Alexa.
  • the present invention relates to a bispecific or multispecific antibody comprising the B7-H6 variant of the present invention and a portion that binds to a target antigen.
  • the bispecific antibody may be a bispecific NK cell engager.
  • the portion that binds to the target antigen may include an antibody or an immunologically active fragment thereof, and the immunologically active fragment may include Fab, Fd, Fab', dAb, F(ab'), F (ab') 2 , may be any one selected from the group consisting of scFv, Fv, single chain antibody, Fv dimer, complementarity determining region fragment, humanized antibody, chimeric antibody and diabody, and is scFv or Fab It is more desirable.
  • the target antigen is 17-1A antigen, GD3 ganglioside R24, EGFRvIII, PSMA, PSCA, HLA-DR, EpCAM, MUC1 core protein, aberrant glycosylation MUC1, a fibronectin isoform containing an ED-B domain, HER2/neu, carcinoembryonic antigen (CEA), gastrin-releasing peptide (GRP) receptor antigen, mucine antigen, epidermal growth factor receptor (EGF-R), HER3, HER4, MAGE antigen, SART antigen, MUC1 antigen.
  • c-erb-2 antigen TAG 72, carbonic anhydrase IX, alpha-fetoprotein, A3, antigen specific for A33 antibody, Ba 733, BrE3-antigen, CA125, CDl, CD1a, CD3, CD5, CDl5, CD16, CD19, CD20, CD21, CD22, CD23, CD25, CD30, CD33, CD38, CD40, CD45, CD52, CD74, CD79a, CD80, CD138, Colon-specific antigen- p(CSAp), CSAp, EGP-1, EGP-2, Ep-CAM, FIt-1, Flt-3, folate receptor, HLA-DR, human chorionic gonadotropin (HCG) and its Subunits, hypoxia-inducible factor (HIF-I), Ia, IL-2, IL-6, IL-8, insulin growth factor-1 (IGF-1), KC4-antigen, KS-1-antigen, KSl- 4, Le-Y, macrophage inhibitory factor
  • the apoptosis-related genes include ABL1, AKT1, AKT2, BARD1, BAX, BCL11B, BCL2, BCL2A1, BCL2L1, BCL2L12, BCL3, BCL6, BIRC2, BIRC3, BIRC5, BRAF, CARD11, CAV1, CBL, CDC25A, CDKN1A, CFLAR, CNR2, CTNNB1, CUL4A, DAXX, DDIT3, E2F1, E2F3, E2F5, ESPL1, FOXO1, HDAC1, HSPA5, IGF1R, IGF2, JUN, JUNB, JUND, MALT1, MAP3K7, MCL1, MDM2, MDM4, MYB, may be MYC, NFKB2, NPM1, NTRK1, PAK1, PAX3, PML, PRKCA, PRKCE, PTK2B, RAF1, RHOA, TGFB1, TNFRSF1B, TP73, TRAF6, YWHAG
  • the oncogene is SEPTIN9, ACOD1, ACTN4, ADAM28, ADAM9, ADGRF1, ADRBK2, AFF1, AFF3, AGAP2, AGFG1, AGRN, AHCYL1, AHI1, AIMP2, AKAP13, AKAP9, AKIRIN2, AKTIP, ALDH1A1, ALL1, ANIB1, ANP32C, ANP32D, AQP1, ARAF, ARHGEF1, ARHGEF2, ARHGEF5, ASPSCR1, AURKA, BAALC, BAIAP2L1, BANP, BCAR4, BCKDHB, BCL9, BCL9L, BCR, BMI1, BMP7, BOC, BRD4, BRF2, CABIN1, CAMK1D, CAPG, CBFB, CBLB, CBLL1, CBX7, CBX8, CCDC28A, CCDC6, CCNB1, CCNB2, CCND1, CCNE1, CCNL1, CD24, CDC25C, CDC6, CDH
  • the target antigen may be a cell surface antigen or an autoantigen
  • the cell surface antigen may be CEA, ED-B fibronectin, CD20, CD22, CDl9, EGFR, IGFlR, VEFGRl/Flt-1, VEGFR2/KDR, VEGRF3 /Flt-4, HER2/neu, CD30, CD33, CD3, CD16, CD64, CD89, CD2, adenovirus fiber knob, PfMSP-1, HN/NDV, EpCAM/17-lA, hTR, IL-2R/Tac, It may be any one or more selected from the group consisting of CAl9-9, MUCl, HLA class II, GD2, G250, TAG-72, PSMA, CEACAM6, HMWMAA, CD40, Ml3 coat protein, and GPIIb/IIIa.
  • the immunologically active fragment of the present invention is Fab, Fd, Fab', dAb, F(ab'), F(ab') 2 , scFv (single chain fragment variable), Fv, single chain antibody. , Fv dimer, complementarity determining region fragment, humanized antibody, chimeric antibody, and diabody.
  • the antibodies are in whole antibody form as well as functional fragments of the antibody molecule.
  • a full antibody has a structure of two full-length light chains and two full-length heavy chains, and each light chain is connected to the heavy chain by a disulfide bond.
  • a functional fragment of an antibody molecule refers to a fragment that possesses an antigen-binding function.
  • antibody fragments include (i) the variable region (VL) of the light chain, the variable region (VH) of the heavy chain, the constant region (CL) of the light chain, and Fab fragment consisting of the first constant region (CH1) of the heavy chain; (ii) Fd fragment consisting of VH and CH1 domains; (iii) an Fv fragment consisting of the VL and VH domains of a single antibody; (iv) a dAb fragment consisting of a VH domain (Ward ES et al., Nature 341:544-546 (1989)); (v) an isolated CDR region; (vi) a bivalent fragment comprising two linked Fab fragments.
  • F(ab')2 fragment (vii) single chain Fv molecule (scFv) joined by a peptide linker that joins the VH domain and VL domain to form an antigen binding site; (viii) bispecific single chain Fv dimer (PCT/US92/09965) and (ix) diabody WO94/13804, which is a multivalent or multispecific fragment produced by gene fusion.
  • scFv single chain Fv molecule
  • a peptide linker that joins the VH domain and VL domain to form an antigen binding site
  • bispecific single chain Fv dimer PCT/US92/09965
  • diabody WO94/13804 which is a multivalent or multispecific fragment produced by gene fusion.
  • the antibody or immunologically active fragment thereof of the present invention may be selected from the group consisting of animal-derived antibodies, chimeric antibodies, humanized antibodies, human antibodies, and immunologically active fragments thereof.
  • the antibody may be recombinantly or synthetically produced.
  • Animal-derived antibodies produced by immunizing an immunized animal with a desired antigen can generally cause immune rejection when administered to humans for therapeutic purposes, and chimeric antibodies have been developed to suppress such immune rejection.
  • a chimeric antibody is one in which the constant region of an animal-derived antibody, which causes an anti-isotype reaction, is replaced with the constant region of a human antibody using genetic engineering methods. Chimeric antibodies have significantly improved anti-isotype responses compared to animal-derived antibodies, but animal-derived amino acids still exist in the variable region, resulting in potential side effects on anti-idiotypic responses. I'm doing it. Humanized antibodies were developed to improve these side effects. It is produced by transplanting the CDR (complementary determining regions) region, which plays an important role in antigen binding, among the variable regions of a chimeric antibody, into the human antibody framework.
  • CDR complementary determining regions
  • CDR grafting technology to produce humanized antibodies.
  • use of an antibody database and crystal structure (crystal structure) Structure analysis, molecular modeling technology, etc. are used.
  • crystal structure crystal structure
  • the application of additional antibody engineering technology to restore antigen binding ability can be said to be essential.
  • the antibody or fragment thereof with immunological activity may be isolated from a living body (not present in the living body) or non-naturally occurring, for example, synthetically or recombinantly produced. You can.
  • antibody refers to a substance produced by stimulation of an antigen within the immune system, the type of which is not particularly limited, and can be obtained naturally or unnaturally (e.g., synthetically or recombinantly). You can. Antibodies are very stable not only in vitro but also in vivo and have a long half-life, making them advantageous for mass expression and production. In addition, antibodies inherently have a dimer structure, so their adhesion ability (avidity) is very high. A complete antibody has a structure of two full-length light chains and two full-length heavy chains, and each light chain is connected to the heavy chain by a disulfide bond.
  • the constant region of an antibody is divided into a heavy chain constant region and a light chain constant region, and the heavy chain constant region has gamma ( ⁇ ), mu ( ⁇ ), alpha ( ⁇ ), delta ( ⁇ ), and epsilon ( ⁇ ) types, and subclasses. It has gamma 1 ( ⁇ 1), gamma 2 ( ⁇ 2), gamma 3 ( ⁇ 3), gamma 4 ( ⁇ 4), alpha 1 ( ⁇ 1), and alpha 2 ( ⁇ 2).
  • the constant region of the light chain has kappa ( ⁇ ) and lambda ( ⁇ ) types.
  • the term “heavy chain” refers to a variable region domain V H and three constant region domains C H1 , C H2 and C H3 comprising an amino acid sequence with sufficient variable region sequence to confer specificity to an antigen. It is interpreted to include both the full-length heavy chain including the hinge and fragments thereof. Additionally, the term “light chain” refers to both a full-length light chain and fragments thereof comprising a variable region domain V L and a constant region domain C L comprising an amino acid sequence with sufficient variable region sequence to confer specificity to an antigen. It is interpreted to mean inclusive.
  • variable region or variable domain refers to a portion of an antibody molecule that performs the function of specifically binding to an antigen and exhibits many variations in sequence, and the variable region has complementary There are crystalline regions CDR1, CDR2 and CDR3. A framework region (FR) exists between the CDRs and serves to support the CDR ring.
  • FR framework region
  • the “complementarity determining region” is a ring-shaped region involved in antigen recognition, and as the sequence of this region changes, the specificity of the antibody to the antigen is determined.
  • scFv single chain fragment variable
  • scFv single chain fragment variable
  • CDR complementarity determining region
  • the term “specifically binds” or “specifically recognizes” has the same meaning commonly known to those skilled in the art, and means that an antigen and an antibody specifically interact to produce an immunological reaction. .
  • the term “antigen-binding fragment” refers to a fragment of the entire structure of an immunoglobulin and a portion of a polypeptide containing a portion to which an antigen can bind.
  • it may be scFv, (scFv) 2, scFv-Fc, Fab, Fab' or F(ab') 2, but is not limited thereto.
  • Fab has one antigen-binding site with a structure that includes the variable regions of the light and heavy chains, the constant region of the light chain, and the first constant region (C H1 ) of the heavy chain.
  • Fab' differs from Fab in that it has a hinge region containing one or more cysteine residues at the C-terminus of the heavy chain C H1 domain.
  • the F(ab') 2 antibody is produced when the cysteine residue in the hinge region of Fab' forms a disulfide bond.
  • Fv is a minimal antibody fragment containing only the heavy chain variable region and the light chain variable region, and recombinant techniques for producing Fv fragments are widely known in the art.
  • a two-chain Fv (two-chain Fv) is a non-covalent bond in which the heavy chain variable region and a light chain variable region are connected, while a single-chain Fv (single-chain Fv) is generally shared between the heavy chain variable region and the short chain variable region through a peptide linker. They can be connected by a bond or directly connected at the C-terminus to form a dimer-like structure, such as double-chain Fv.
  • the linker may be a peptide linker consisting of 1 to 100 or 2 to 50 amino acids, and suitable sequences are known in the art.
  • the antigen-binding fragment can be obtained using a proteolytic enzyme (for example, Fab can be obtained by restriction digestion of the entire antibody with papain, and F(ab') 2 fragment can be obtained by digestion with pepsin), It can be produced through genetic recombination technology.
  • a proteolytic enzyme for example, Fab can be obtained by restriction digestion of the entire antibody with papain, and F(ab') 2 fragment can be obtained by digestion with pepsin
  • the term "hinge region” refers to a region contained in the heavy chain of an antibody, which exists between the C H1 and C H2 regions and functions to provide flexibility of the antigen binding site in the antibody. It means area.
  • the hinge may be derived from a human antibody, specifically, IgA, IgE, or IgG, such as IgG1, IgG2, IgG 3, or IgG4.
  • the invention relates to an isolated nucleic acid molecule encoding the B7-H6 variant or fragment thereof, or bispecific or multispecific antibody of the invention, a vector containing the same, and a host cell transformed therewith. .
  • Nucleic acid molecules of the invention may be isolated or recombinant and include single- and double-stranded forms of DNA and RNA as well as corresponding complementary sequences.
  • An isolated nucleic acid in the case of a nucleic acid isolated from a naturally occurring source, is a nucleic acid that has been separated from the surrounding genetic sequence present in the genome of the individual from which the nucleic acid was isolated.
  • nucleic acids synthesized enzymatically or chemically from a template such as PCR products, cDNA molecules, or oligonucleotides
  • the nucleic acids resulting from these procedures may be understood as isolated nucleic acid molecules.
  • Isolated nucleic acid molecules refer to nucleic acid molecules either in the form of separate fragments or as components of larger nucleic acid constructs.
  • a nucleic acid is operably linked when placed in a functional relationship with another nucleic acid sequence.
  • the DNA of the presequence or secretion leader is operably linked to the DNA of the polypeptide when the polypeptide is expressed as a preprotein in a form before secretion
  • the promoter or enhancer is a polypeptide sequence. is operably linked to the coding sequence when it affects transcription
  • the ribosome binding site is operably linked to the coding sequence when configured to facilitate translation.
  • operably linked means that the DNA sequences to be linked are located adjacent to each other, and in the case of a secretory leader, it means that they are adjacent and exist within the same reading frame. However, enhancers do not need to be located adjacently. Linking is accomplished by ligation at convenient restriction enzyme sites. If such sites do not exist, synthetic oligonucleotide adapters or linkers are used according to conventional methods.
  • Isolated nucleic acid molecules encoding the antibodies of the present invention or immunologically active fragments thereof, or the bispecific or multispecific antibodies of the present invention may be used due to codon degeneracy or for the purpose of expressing the antibodies.
  • codons preferred in organisms various modifications can be made to the coding region within the range that does not change the amino acid sequence of the antibody expressed from the coding region, and does not affect gene expression in parts other than the coding region.
  • a person skilled in the art will understand that various modifications or modifications can be made within the gene, and that such modified genes are also included within the scope of the present invention.
  • nucleic acid molecule of the present invention encodes a protein with equivalent activity
  • one or more nucleic acid bases may be mutated by substitution, deletion, insertion, or a combination thereof, and these are also included within the scope of the present invention.
  • the sequence of these nucleic acid molecules may be single or double stranded, and may be DNA molecules or RNA (mRNA) molecules.
  • An isolated nucleic acid molecule encoding an antibody of the present invention or a fragment having immunological activity thereof, or a bispecific or multispecific antibody of the present invention may be inserted into an expression vector for protein expression.
  • Expression vectors typically contain proteins operably linked, i.e., placed in a functional relationship, with regulatory or control sequences, selectable markers, optional fusion partners, and/or additional elements.
  • a host cell transformed with a nucleic acid preferably containing an antibody of the invention or an immunologically active fragment thereof, or an isolated nucleic acid molecule encoding a bispecific or multispecific antibody of the invention, is expressed.
  • An antibody of the present invention, a fragment having immunological activity thereof, or a bispecific or multispecific antibody of the present invention can be produced by culturing a vector to induce protein expression.
  • a vector to induce protein expression A variety of suitable host cells can be used, including, but not limited to, mammalian cells, bacteria, insect cells, and yeast. Methods for introducing exogenous nucleic acids into host cells are known in the art and will vary depending on the host cell used. Preferably, E. coli, which has low production cost and thus has high industrial value, can be produced as a host cell.
  • Vectors of the present invention include, but are not limited to, plasmid vectors, cosmid vectors, bacteriophage vectors, viral vectors, etc.
  • Suitable vectors include expression control elements such as promoters, operators, start codons, stop codons, polyadenylation signals, and enhancers, as well as signal sequences or leader sequences for membrane targeting or secretion, and can be prepared in various ways depending on the purpose.
  • the promoter of the vector may be constitutive or inducible.
  • the signal sequence includes the PhoA signal sequence and OmpA signal sequence when the host is Escherichia sp., and the ⁇ -amylase signal sequence and subtilisin signal when the host is Bacillus sp.
  • the host is yeast, the MF ⁇ signal sequence, SUC2 signal sequence, etc. can be used, and if the host is an animal cell, the insulin signal sequence, ⁇ -interferon signal sequence, antibody molecule signal sequence, etc. can be used. It is not limited to this.
  • the vector may include a selection marker for selecting host cells containing the vector, and if it is a replicable expression vector, it will include an origin of replication.
  • vector refers to a carrier capable of inserting a nucleic acid sequence for introduction into a cell capable of replicating the nucleic acid sequence.
  • Nucleic acid sequences may be exogenous or heterologous.
  • Vectors include, but are not limited to, plasmids, cosmids, and viruses (eg, bacteriophages). Those skilled in the art can construct vectors by standard recombination techniques (Maniatis, et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y., 1988; and Ausubel et al., In: Current Protocols in Molecular Biology, John, Wiley & Sons, Inc, NY, 1994, etc.).
  • expression control sequences such as promoters, terminators, enhancers, etc., depending on the type of host cell for producing the antibody, membrane targeting or secretion Sequences, etc. can be appropriately selected and combined in various ways depending on the purpose.
  • expression vector refers to a vector containing a nucleic acid sequence encoding at least a portion of the gene product to be transcribed. In some cases, the RNA molecule is then translated into a protein, polypeptide, or peptide. Expression vectors may contain various control sequences. In addition to regulatory sequences that regulate transcription and translation, vectors and expression vectors may also contain nucleic acid sequences that also serve other functions.
  • the term “host cell” includes eukaryotes and prokaryotes and refers to any transformable organism capable of replicating the vector or expressing the gene encoded by the vector.
  • the host cell may be transfected or transformed by the vector, which refers to a process in which an exogenous nucleic acid molecule is transferred or introduced into the host cell.
  • the host cells may be bacteria or animal cells
  • the animal cell line may be CHO cells, HEK cells, or NSO cells
  • the bacteria may be Escherichia coli.
  • the present invention relates to a pharmaceutical composition for the treatment or prevention of cancer, comprising the B7-H6 variant or fragment thereof, or a bispecific or multispecific antibody of the present invention.
  • the cancer is brain tumor, melanoma, myeloma, non-small cell lung cancer, oral cancer, liver cancer, stomach cancer, colon cancer, breast cancer, lung cancer, bone cancer, pancreatic cancer, skin cancer, head or neck cancer, cervical cancer, ovarian cancer, and colon cancer.
  • soft tissue sarcoma urethral cancer
  • penile cancer prostate cancer
  • chronic or acute leukemia lymphocytic lymphoma
  • renal or ureteral cancer renal cell carcinoma, renal pelvic carcinoma
  • central nervous system tumor primary central nervous system lymphoma
  • spinal cord tumor brainstem glioma.
  • pituitary adenoma pituitary adenoma
  • prevention refers to all actions that inhibit or delay the occurrence, spread, and recurrence of a disease or condition by administering the composition according to the present invention.
  • treatment refers to any action that improves or beneficially changes the symptoms of a disease or condition and complications resulting therefrom by administering the composition according to the present invention.
  • anyone with ordinary knowledge in the technical field to which the present invention pertains can refer to the data presented by the Korean Medical Association, etc. to know the exact criteria for diseases for which our composition is effective and to determine the degree of improvement, improvement, and treatment. will be.
  • terapéuticaally effective amount used in combination with an active ingredient in the present invention refers to an amount effective in preventing or treating a disease or disorder, and the therapeutically effective amount of the composition of the present invention is determined by several factors, such as administration. It may vary depending on the method, target area, patient condition, etc. Therefore, when used in the human body, the dosage must be determined as appropriate by considering both safety and efficiency. It is also possible to estimate the amount used in humans from the effective amount determined through animal testing. These considerations in determining an effective amount include, for example, Hardman and Limbird, eds., Goodman and Gilman's The Pharmacological Basis of Therapeutics, 10th ed. (2001), Pergamon Press; and E.W. Martin ed., Remington's Pharmaceutical Sciences, 18th ed. (1990), Mack Publishing Co.
  • the pharmaceutical composition of the present invention is administered in a pharmaceutically effective amount.
  • pharmaceutically effective amount refers to an amount that is sufficient to treat a disease with a reasonable benefit/risk ratio applicable to medical treatment and does not cause side effects, and the effective dose level is determined by the patient's Factors including health status, type of disease or condition, severity of disease or condition, activity of drug, sensitivity to drug, method of administration, time of administration, route of administration and excretion rate, treatment period, drugs combined or used simultaneously, and other factors. It can be determined based on factors well known in the medical field.
  • composition of the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, may be administered sequentially or simultaneously with conventional therapeutic agents, and may be administered singly or multiple times. Considering all of the above factors, it is important to administer an amount that can achieve maximum effect with the minimum amount without side effects, and this can be easily determined by a person skilled in the art.
  • the pharmaceutical composition of the present invention may contain a carrier, diluent, excipient, or a combination of two or more commonly used in biological products.
  • a carrier diluent, excipient, or a combination of two or more commonly used in biological products.
  • pharmaceutically acceptable means that the composition exhibits non-toxic properties to cells or humans exposed to the composition.
  • the carrier is not particularly limited as long as it is suitable for in vivo delivery of the composition, for example, Merck Index, 13th ed., Merck & Co. Inc.
  • saline solution sterilized water, Ringer's solution, buffered saline solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and one or more of these ingredients can be mixed and used, and if necessary, other ingredients such as antioxidants, buffers, and bacteriostatic agents. Normal additives can be added.
  • diluents, dispersants, surfactants, binders, and lubricants can be additionally added to formulate dosage forms such as aqueous solutions, suspensions, emulsions, etc., into pills, capsules, granules, or tablets.
  • it can be preferably formulated according to each disease or ingredient using an appropriate method in the art or a method disclosed in Remington's Pharmaceutical Science (Mack Publishing Company, Easton PA, 18th, 1990).
  • the pharmaceutical composition may be one or more formulations selected from the group including oral formulations, topical formulations, suppositories, sterile injectable solutions, and sprays, with oral or injectable formulations being more preferable.
  • the term "administration” means providing a predetermined substance to an individual or patient by any appropriate method, and is administered parenterally (e.g., intravenously, subcutaneously, intraperitoneally) according to the desired method. Alternatively, it can be applied topically as an injection formulation) or orally administered, and the dosage range varies depending on the patient's weight, age, gender, health status, diet, administration time, administration method, excretion rate, and severity of the disease.
  • Liquid preparations for oral administration of the composition of the present invention include suspensions, oral solutions, emulsions, syrups, etc., and in addition to the commonly used simple diluents such as water and liquid paraffin, various excipients such as wetting agents, sweeteners, fragrances, and preservatives are used. etc. may be included together.
  • Preparations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, freeze-dried preparations, suppositories, etc.
  • the pharmaceutical composition of the present invention may be administered by any device capable of transporting the active agent to target cells.
  • Preferred administration methods and formulations include intravenous injection, subcutaneous injection, intradermal injection, intramuscular injection, and drip injection.
  • Injections include aqueous solvents such as physiological saline solution and Ringer's solution, non-aqueous solvents such as vegetable oil, higher fatty acid esters (e.g., ethyl oleate, etc.), and alcohols (e.g., ethanol, benzyl alcohol, propylene glycol, glycerin, etc.).
  • stabilizers to prevent deterioration
  • emulsifiers e.g., ascorbic acid, sodium bisulfite, sodium pyrosulphite, BHA, tocopherol, EDTA, etc.
  • buffers for pH adjustment e.g., buffers for pH adjustment
  • agents to prevent microbial growth e.g., ascorbic acid, sodium bisulfite, sodium pyrosulphite, BHA, tocopherol, EDTA, etc.
  • emulsifiers e.g., ascorbic acid, sodium bisulfite, sodium pyrosulphite, BHA, tocopherol, EDTA, etc.
  • emulsifiers e.g., buffers for pH adjustment
  • agents to prevent microbial growth e.g., buffers for pH adjustment, and
  • the term "individual” refers to monkeys, cows, horses, sheep, pigs, chickens, turkeys, quails, cats, dogs, mice, bats, including humans that have or may develop the disease or condition. It means any animal, including camel, rat, rabbit or guinea pig, and “specimen” may be droplets, sputum, whole blood, plasma, serum, urine or saliva isolated therefrom.
  • the pharmaceutical composition of the present invention may further include pharmaceutically acceptable additives, wherein the pharmaceutically acceptable additives include starch, gelatinized starch, microcrystalline cellulose, lactose, povidone, colloidal silicon dioxide, and calcium hydrogen phosphate. , lactose, mannitol, taffy, gum arabic, pregelatinized starch, corn starch, powdered cellulose, hydroxypropyl cellulose, Opadry, sodium starch glycolate, lead carnauba, synthetic aluminum silicate, stearic acid, magnesium stearate, aluminum stearate, Calcium stearate, white sugar, dextrose, sorbitol, and talc can be used.
  • the pharmaceutically acceptable additive according to the present invention is preferably contained in an amount of 0.1 to 90 parts by weight based on the composition, but is not limited thereto.
  • the present invention includes the steps of isolating natural killer cells; and culturing the isolated natural killer cells in the presence of the B7-H6 variant or fragment thereof of the present invention.
  • the present invention relates to the use of the B7-H6 variant of the present invention or a bispecific or multispecific antibody comprising the same for the prevention or treatment of cancer.
  • the present invention relates to a method of treating cancer comprising administering the B7-H6 variant of the present invention or a bispecific or multispecific antibody containing the same in a pharmaceutically effective amount to an individual suffering from cancer. will be.
  • the avidity effect was induced through tetramerization of the NKp30 protein.
  • streptavidin was expressed in the C-terminal part of NKp30 to induce tetramerization, and glycine and serine linkers were inserted between NKp30 and streptavidin to ensure the fluidity of each protein.
  • the NKp30 and streptavidin genes were each amplified using primers and Vent polymerase (New England Biolab), and then assembly PCR was performed using Vent polymerase. The created gene was subjected to restriction enzyme treatment using Bss HII and Xba I (New England Biolab).
  • NKp30-Streptavdin gene was ligated into pMAZ vector, a vector for animal cells, treated with the same restriction enzyme. After transforming the ligated plasmid into E. coli Jude1, a single clone was obtained and sequenced to confirm that NKp30-streptavidin was successfully inserted into the pMAZ vector ( Figure 1).
  • PEI Polyehylenimine, Polyscience, 23966
  • NKp30 prepared in Example 1 were added to 30 ml of Freestyle 293 expression culture medium (Gibco. 12338-018).
  • -Streptavidin-His tag expression vector was mixed at a ratio of 1:4, left at room temperature for 20 minutes, and then transfected into the Expi293F animal cells.
  • the cells were cultured in a CO 2 shaking incubator at 37°C, 125 rpm, and 8% CO 2 for 7 days, then centrifuged, and only the supernatant was collected.
  • the mixture was equilibrated using 25x PBS and filtered through a 0.2 ⁇ m filter (Merck Millipore) using a bottle top filter.
  • 1 ml of Ni-NTA resin was added to the filtered culture medium, stirred at 4°C for 16 hours, and then passed through a column to recover the resin and washed with 10 ml of PBS.
  • the washed resin was sequentially washed with 10 ml each of 10mM imidazole buffer and 20mM imidazole buffer, and then eluted with 4ml of 250mM imidazole buffer.
  • the purified NKp30-streptavidin tetramer protein was fluorescently labeled using an Alexa-488 labeling kit.
  • the amplified wild-type B7-H6 gene was subjected to restriction enzyme treatment using Sfi I (New England Biolab), and the restriction enzyme-treated gene was ligated into the pCTCON vector treated with the same restriction enzyme. After transforming the ligated plasmid into Jude1 E. coli, a single clone was obtained and sequenced, and two plasmids, pCTCON-Aga2-B7-H6_WT-FLAG and pCTCON-B7-H6_WT-Aga2-FLAG, were successfully cloned. was confirmed.
  • the display method was selected by verifying the binding affinity between B7-H6 expressed on the yeast surface and the probe NKp30-streptavidin using a flow cytometer. Specifically, the two plasmids prepared in Example 3-1 were transformed into the AWY101 (Trp-) strain, and 50 ⁇ g/ml of Kanamycin and 40 ⁇ g/ml of Chloramphenicol were added. 30 in 5 ml of medium containing SDCAA (20 g/L Glucose, 6.7 g/L Yeast nitrogen base without amino acids, 5 g/L casamino acids, 5.4 g/L Na 2 HPO 4 and 8.56 g/L NaH 2 PO 4 ).
  • 2x10 7 cells were centrifuged (14,000g, 30 seconds, 4°C) and recovered in an e-tube. Add 1 ml of PBSB (0.1% BSA in PBS) to each e-tube from which the cells were recovered, resuspend them, collect the cells again through centrifugation (14,000 g, 30 seconds, 4°C), and then add 0.5 ml of PBSB and resuspend. It was cloudy, making 4x10 7 cells/ml.
  • PBSB 0.1% BSA in PBS
  • pCTCON-B7-H6_WT-Aga2-FLAG contains Sfi I sites on both sides to allow random mutations to all regions of B7-H6.
  • a primer was designed. DNA was first amplified using the Error-Prone PCR technique using the designed primers, Taq Polymerase (TAKARA), dNTPs (Invitrogen), MgCl 2 and MnCl 2 (SIGMA). The amplified gene was prepared by secondary amplification using Vent polymerase (24 ⁇ g), and the vector was prepared by treatment with Sfi I restriction enzyme (8 ⁇ g). The two prepared genes were transformed into the AWY101 strain to construct a library through homologous recombination. The constructed library had a size of 5.1 This was confirmed ( Figure 3).
  • the cultured cells were recovered the next day, cultured in 100 ml of SGCAA at 20°C and 225 rpm for 2-3 days, and then inducted, before proceeding with the next round.
  • the above screening process was performed a total of four times while decreasing the concentration of the probe.
  • SDCAA (20 g/L Glucose, 6.7 g/L Yeast nitrogen base without amino acids, 5 g/L casamino acids, 5.4 g/L Na 2 HPO 4 ) with 50 ⁇ g/ml kanamycin and 40 ⁇ g/ml chloramphenicol , 8.56 g/L NaH 2 PO 4 )
  • the initials, 1st round, 2nd round, 3rd round, and 4th round libraries of Example 5 were separately inoculated in 100 ml of medium and cultured at 30°C and 225 rpm for 16 hours. .
  • Example 7 Amino acid sequence analysis of B7-H6 variants and securing of B7-H6 variants with increased binding affinity to NKp30
  • mutants were incubated with SDCAA (20 g/L Glucose, 6.7 g) with 50 ⁇ g/ml kanamycin and 40 ⁇ g/ml chloramphenicol, respectively.
  • SDCAA 20 g/L Glucose, 6.7 g
  • 5x10 7 cells were obtained by centrifugation (2,500g, 5 minutes, 4°C), and then incubated with SGCAA (20 g/L Galactose, 6.7 g/mL) to which 50 ⁇ g/ml kanamycin and 40 ⁇ g/ml chloramphenicol were added.
  • SGCAA 20 g/L Galactose, 6.7 g/mL
  • the wild-type B7-H6 the genes of three variants (B5, B7, and B14) selected from among the 17 B7-H6 variants selected in Example 7, the Fc domain, a designed primer, and Vent Polymerase ( After amplification using (New England Biolab), the amplified gene was subjected to Assembly PCR and then treated with Bss HII and Xba I restriction enzymes (New England Biolab). The genes of the restriction enzyme-treated B7-H6 mutants were ligated into the pMAZ vector, a vector for animal cells, treated with the same restriction enzyme. After transforming the ligated plasmid into E.
  • the constructed B7-H6 mutant-Fc fusion protein expression vector was transfected into Expi293F animal cells, cultured in a CO 2 shaking incubator at 37°C, 125 rpm, and 8% CO 2 for 7 days, and then centrifuged to collect only the supernatant. separated. Afterwards, it was equilibrated using 25x PBS and filtered using a 0.2 ⁇ m syringe filter (Sartorius, S6634).
  • ELISA was performed to analyze the NKp30 binding ability of the B7-H6 variant-Fc fusion proteins purified in Example 8. Specifically, the B7-H6 variant-Fc fusion proteins diluted to 4 ⁇ g/ml in 0.05 M Na 2 CO 3 pH 9.6 were dispensed in 50 ⁇ l portions into Flat Bottom Polystyrene High Bind 96-well microplates (costar) and incubated for 16 minutes at 4°C. After immobilization for an hour, the cells were blocked with 100 ⁇ l of 4% skim milk (Biopure) (in PBS, pH 7.4) at room temperature for 1 hour.
  • Biopure in PBS, pH 7.4
  • a library to further discover new B7-H6 variants with improved NKp30 binding by introducing mutations at two amino acid positions in the three variants (B5, B7, B14), excluding mutations matching wild-type B7-H6. was produced.
  • a yeast surface display library was constructed in the same manner as in Example 4 above. The constructed library was 1.5 ⁇ 10 7 in size, and through sequence analysis, it was confirmed that mutations were included at two locations (FIG. 8).
  • Yeasts with high binding affinity to NKp30 were recovered by measuring the fluorescence signal value of the library sample in the same manner as in Example 5, and the screening process was performed once.
  • the expression level of the variants and their binding ability to NKp30 were indirectly analyzed by measuring the fluorescence signal value in the same manner as in Example 7. Through this, a total of 9 variants (BF2, BF3, BF5, BF8, BF11, BF19, BF25, BF39, and BF46) with improved binding to NKp30 were selected (Table 2 and Figure 10).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne des variants de B7-H6 ayant une affinité de liaison améliorée pour NKp30. Les variants de B7-H6 selon la présente invention peuvent augmenter l'activation de cellules tueuses naturelles (NK) en raison d'une affinité de liaison significativement accrue pour NKp30, qui est un récepteur d'activation de cellules NK, par comparaison avec le type sauvage, sont faciles à pénétrer dans le micro-environnement tumoral en raison d'une taille beaucoup plus petite que les anticorps, et sont facilement produits, et peuvent ainsi être utilisés de manière efficace seuls ou en combinaison avec divers agents immunothérapeutiques pour traiter le cancer ou des maladies infectieuses.
PCT/KR2023/008393 2022-06-29 2023-06-16 Variants de b7-h6 à affinité de liaison améliorée pour nkp30 WO2024005422A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2022-0079826 2022-06-29
KR20220079826 2022-06-29

Publications (1)

Publication Number Publication Date
WO2024005422A1 true WO2024005422A1 (fr) 2024-01-04

Family

ID=89380778

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/008393 WO2024005422A1 (fr) 2022-06-29 2023-06-16 Variants de b7-h6 à affinité de liaison améliorée pour nkp30

Country Status (2)

Country Link
KR (1) KR20240003756A (fr)
WO (1) WO2024005422A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20100091950A (ko) * 2007-10-04 2010-08-19 지모제넥틱스, 인코포레이티드 B7 패밀리 구성원 zB7H6 및 관련된 조성물 및 방법
WO2011070443A1 (fr) * 2009-12-09 2011-06-16 Institut National De La Sante Et De La Recherche Medicale Anticorps monoclonaux qui se lient à b7h6 et leurs utilisations
US11291721B2 (en) * 2016-03-21 2022-04-05 Marengo Therapeutics, Inc. Multispecific and multifunctional molecules and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20100091950A (ko) * 2007-10-04 2010-08-19 지모제넥틱스, 인코포레이티드 B7 패밀리 구성원 zB7H6 및 관련된 조성물 및 방법
WO2011070443A1 (fr) * 2009-12-09 2011-06-16 Institut National De La Sante Et De La Recherche Medicale Anticorps monoclonaux qui se lient à b7h6 et leurs utilisations
US11291721B2 (en) * 2016-03-21 2022-04-05 Marengo Therapeutics, Inc. Multispecific and multifunctional molecules and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE Protein 19 March 2015 (2015-03-19), ANONYMOUS : "B7H6, partial [synthetic construct]", XP093121937, retrieved from NCBI Database accession no. AIC53717.1 *
M. G. JOYCE, TRAN P., ZHURAVLEVA M. A., JAW J., COLONNA M., SUN P. D.: "Crystal structure of human natural cytotoxicity receptor NKp30 and identification of its ligand binding site", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, vol. 108, no. 15, 12 April 2011 (2011-04-12), pages 6223 - 6228, XP055043601, ISSN: 00278424, DOI: 10.1073/pnas.1100622108 *
SKOŘEPA ONDŘEJ, PAZICKY SAMUEL, KALOUSKOVÁ BARBORA, BLÁHA JAN, ABREU CELESTE, JEČMEN TOMÁŠ, ROSŮLEK MICHAL, FISH ALEXANDER, SEDIVY: "Natural Killer Cell Activation Receptor NKp30 Oligomerization Depends on Its N-Glycosylation", CANCERS, CH, vol. 12, no. 7, 21 July 2020 (2020-07-21), CH , pages 1998, XP093121934, ISSN: 2072-6694, DOI: 10.3390/cancers12071998 *

Also Published As

Publication number Publication date
KR20240003756A (ko) 2024-01-09

Similar Documents

Publication Publication Date Title
JP7307919B2 (ja) インターロイキン-2のスーパーアンタゴニスト、パーシャルアゴニスト及びアンタゴニスト
Srivastava et al. Individually distinct transplantation antigens of chemically induced mouse tumors
Palladino Jr et al. Expression of a shared tumor-specific antigen by two chemically induced BALB/c sarcomas
JP2023542417A (ja) Kras変異に特異的t細胞受容体のスクリーニング及び抗腫瘍用途
KR20200064980A (ko) 종양 세포에서 면역 조절제 및 항-암 치료제를 생산하도록 프로그램된 미생물
JP4900884B2 (ja) 腫瘍抗原
WO2000058480A1 (fr) Nouvelle cytidine desaminase
JPH03501330A (ja) カケクチンと反応するモノクロナール抗体
JP5292550B2 (ja) T細胞レセプターβ鎖遺伝子及びα鎖遺伝子
CA2404233A1 (fr) Compositions et methodes pouvant traiter ou diagnostiquer le cancer du poumon
JP2000154153A (ja) 多段カスケ―ド型追加免疫ワクチン
EA006310B1 (ru) Ассоциированные с ганглиозидами рекомбинантные антитела и их применение в диагностике и лечении опухолей
CN112912391A (zh) T细胞受体及其用途
JPH06507384A (ja) 特定のt細胞群による病理的応答により生じる疾患に対するワクチン投与および方法
WO2018211245A1 (fr) Cellule
CN112442132A (zh) 靶向肿瘤的重组双功能融合蛋白及其应用
EP4242233A1 (fr) Anticorps de liaison au récepteur fc alpha
WO1998022510A2 (fr) Procedes de production d'anticorps monoclonaux de poulet
GB2603166A (en) Therapeutic and Diagnostic Agents and Uses Thereof
CA2130173A1 (fr) Anticorps produits par genie genetique
JP2004147649A (ja) 頭頚部癌の抗原
WO2022220603A1 (fr) Anticorps humain ciblant le virus de covid-19
KR102766045B1 (ko) Fc 알파 수용체 결합력이 향상된 항체
KR102663963B1 (ko) 코로나-19 바이러스 표적 인간 항체
WO2024005422A1 (fr) Variants de b7-h6 à affinité de liaison améliorée pour nkp30

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: 23831782

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE