WO2023234743A1 - Bispecific antibody including anti-tigit antibody, and use thereof - Google Patents

Bispecific antibody including anti-tigit antibody, and use thereof Download PDF

Info

Publication number
WO2023234743A1
WO2023234743A1 PCT/KR2023/007622 KR2023007622W WO2023234743A1 WO 2023234743 A1 WO2023234743 A1 WO 2023234743A1 KR 2023007622 W KR2023007622 W KR 2023007622W WO 2023234743 A1 WO2023234743 A1 WO 2023234743A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
cancer
bispecific antibody
tigit
Prior art date
Application number
PCT/KR2023/007622
Other languages
French (fr)
Korean (ko)
Inventor
장명호
고영준
Original Assignee
(주)지아이이노베이션
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 (주)지아이이노베이션 filed Critical (주)지아이이노베이션
Publication of WO2023234743A1 publication Critical patent/WO2023234743A1/en

Links

Images

Classifications

    • 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
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to an anti-TIGIT antibody or fragment thereof; and a bispecific antibody containing the IL-2 protein and a pharmaceutical composition for treating or preventing cancer using the same.
  • Cancer immunotherapy is a method of treating cancer using the body's immune system. Cancer immunotherapy can trigger the immune system to attack cancer cells by targeting antigens, such as cancer cell surface proteins. In particular, it has been reported that anti-cancer immunity can be activated through blocking the immune checkpoint pathway. Immune checkpoints are one of the main mechanisms by which tumor cells result in immune evasion. Therefore, inhibition or blockade of immune checkpoints can increase T cell activation, thereby enhancing anti-tumor immunity.
  • IL-2 is mainly synthesized by activated T cells, especially CD4+ helper T cells.
  • IL-2 stimulates the proliferation and differentiation of T cells and promotes the production of cytotoxic T lymphocytes (CTL).
  • CTL cytotoxic T lymphocytes
  • IL-2 has a dual function in the immune response in that it not only mediates the increase and activity of immune cells, but is also important in maintaining immune tolerance. Additionally, it has been reported that IL-2 may be suboptimal for inhibiting tumor growth. This is because, in the presence of IL-2, activation-induced cell death (AICD) may occur in the generated cytotoxic T lymphocytes, and the immune response may be suppressed by IL-2-dependent regulatory T cells (Treg). (Imai et al., Cancer Sci 98, 416-423, 2007).
  • TIGIT T cell immunoreceptor with Ig and ITIM domains
  • receptor an immune checkpoint protein (receptor) that binds to CD155 present in dendritic cells, macrophages, etc. to activate T cells and natural killer cells in vivo. is known to inhibit.
  • the present inventors conducted research to develop a new combination of dual-specific antibodies that enhance the activity of immune cells, and as a result confirmed that dual-specific antibodies including anti-TIGIT antibodies and IL-2 variants effectively regulate immune cells. did. Based on this, the present invention was completed by confirming that the bispecific antibody has an anticancer effect.
  • one aspect of the present invention is an antibody or fragment thereof that specifically binds to TIGIT; and IL-2 protein.
  • Another aspect of the present invention provides a polynucleotide encoding the bispecific antibody, an expression vector containing the polynucleotide, and a transformed cell into which the expression vector has been introduced.
  • Another aspect of the present invention provides a method for producing a bispecific antibody, comprising culturing the transformed cells and obtaining a bispecific antibody.
  • Another aspect of the present invention provides a pharmaceutical composition for preventing or treating cancer comprising the bispecific antibody as an active ingredient.
  • Another aspect of the present invention provides the use of the bispecific antibody for preparing a medicament for preventing or treating cancer.
  • Another aspect of the invention provides the use of the bispecific antibody for preventing or treating cancer.
  • Another aspect of the present invention provides a method for preventing or treating cancer comprising administering the bispecific antibody to a subject.
  • Bispecific antibodies including anti-TIGIT antibodies and IL-2 variants according to the present invention, can modulate TIGIT-related mechanisms and have the same or similar functions as IL-2.
  • the bispecific antibody can not only regulate the binding of TIGIT and CD155, but also activate immune cells. Additionally, tumor growth was significantly inhibited in mouse tumor models. Therefore, the bispecific antibody can be used as an anticancer agent.
  • Figure 1 is a diagram schematically illustrating the structure of the GI-106 bispecific antibody, which is an example.
  • Figure 2 is a diagram showing the results of confirming the purified bispecific antibody (GI-106) by SDS-PAGE.
  • Figure 3 is a diagram schematically illustrating the structure of the GI-106B7NH06Kv3 bispecific antibody, which is an example.
  • Figure 4 is a diagram showing the results of confirming the purified bispecific antibody (GI-106B7NH06Kv3) by SDS-PAGE.
  • Figure 5 is a diagram schematically illustrating the mechanism of action of the TIGIT/CD155 inhibition assay (Blockade Bioassay) for measuring the anti-hTIGIT antibody activity of GI-106.
  • Figure 6 is a graph showing the degree of inhibition of TIGIT/CD155 binding by concentration of GI-106 and anti-hTIGIT antibody in the TIGIT/CD155 inhibition assay.
  • Figure 7 is a graph showing the results of treating HEK-Blue TM IL-2 reporter cells with GI-106 or Proleukin® and measuring JAK-STAT signaling pathway activity.
  • Figure 8 is a graph showing the results of measuring the tumor volume of each group after administering PBS or GI-106 to mice implanted with human-derived breast cancer cells (MDA-MB-231).
  • Figure 9 is a graph showing the tumor volume of each individual in each group after administering PBS or GI-106 to mice implanted with human-derived breast cancer cells (MDA-MB-231).
  • Figure 10 is a graph showing the tumor volume for each individual after administration of PBS to mice implanted with human-derived breast cancer cells (MDA-MB-231).
  • Figure 11 is a graph showing the tumor volume for each individual after administering GI-106 to mice implanted with human-derived breast cancer cells (MDA-MB-231).
  • FIG 12 is a graph showing the tumor growth inhibition (TGI) of each group of mice after administering PBS or GI-106 to mice implanted with human-derived breast cancer cells (MDA-MB-231).
  • TGI tumor growth inhibition
  • Figure 13 is a graph showing the results of measuring the tumor volume of each group after administering hIgG4 or GI-106 B7NH06Kv3 to mice implanted with mouse-derived breast cancer cells (EMT-6).
  • Figure 14 is a graph showing the tumor growth inhibition rate of each group of mice after administering hIgG4 or GI-106 B7NH06Kv3 to mice implanted with mouse-derived breast cancer cells (EMT-6).
  • Figure 15 shows the results of each group after administering hIgG4, GI-106B7NH06Kv3, or GI-106B7NH06K-CN alone or in combination with GI-106B7NH06K-CN and Fc-IL-2v3 to mice implanted with mouse-derived colon cancer cells (MC38). This is a graph showing the results of measuring tumor volume.
  • Figure 16 shows the results of each group after single administration of hIgG4, GI-106B7NH06Kv3, or GI-106B7NH06K-CN or combined administration of GI-106B7NH06K-CN and Fc-IL-2v3 in mice implanted with mouse-derived colon cancer cells (MC38). This is a graph showing the tumor growth inhibition rate.
  • Figure 17 shows the results of each group after single administration of hIgG4, GI-106B7NH06Kv3, or GI-106B7NH06K-CN or combined administration of GI-106B7NH06K-CN and Fc-IL-2v3 in mice implanted with mouse-derived colon cancer cells (MC38). This is a graph showing the survival rate until the 25th day.
  • anti-TIGIT antibody or fragment thereof and a bispecific antibody containing the IL-2 protein.
  • One aspect of the present invention is an antibody or fragment thereof that specifically binds to TIGIT; and IL-2 protein.
  • antibody refers to an immunoglobulin molecule that reacts immunologically with a specific antigen, and refers to a protein molecule that specifically recognizes the antigen.
  • the heavy and light chains of immunoglobulins may each include a constant region and a variable region.
  • the light and heavy chain variable regions of immunoglobulins contain three variable regions called complementarity determining regions (CDRs) and four framework regions (FRs).
  • CDR complementarity determining regions
  • FRs framework regions
  • the term “dual specific antibody” refers to a substance that binds to at least one target or antigen.
  • the bispecific antibody may include an antigen binding site that specifically binds to TIGIT and an IL-2 protein or a variant thereof.
  • TIGIT is also known as WUCAM and Vstm2.
  • TIGIT is an immune receptor present on some T cells and natural killer cells (NK cells), and binds to CD155 present on dendritic cells and macrophages to inhibit the activity of T cells and natural killer cells in vivo. Inhibits and reduces immune response.
  • the antibody or fragment thereof that specifically binds to TIGIT may collectively refer to molecules capable of antigen-antibody binding specifically to TIGIT.
  • the antibody or fragment thereof that specifically binds to TIGIT may have any form as long as it contains an antigen-binding site capable of specifically binding to TIGIT.
  • the antibody or fragment thereof may contain other amino acids that are not directly involved in binding or amino acids whose effect is blocked by amino acid residues in the antigen binding site.
  • the antibody or fragment thereof that specifically binds to TIGIT is a heavy chain variable region consisting of HCDR1 containing the amino acid sequence of SEQ ID NO: 9, HCDR2 containing the amino acid sequence of SEQ ID NO: 10, and HCDR3 containing the amino acid sequence of SEQ ID NO: 11 and a light chain variable region consisting of LCDR1 including the amino acid sequence of SEQ ID NO: 12, LCDR2 including the amino acid sequence (GVK) of SEQ ID NO: 13, and LCDR3 including the amino acid sequence of SEQ ID NO: 14.
  • the antibody or fragment thereof that specifically binds to TIGIT may be composed of a heavy chain variable region including the amino acid sequence of SEQ ID NO: 2 and a light chain variable region including the amino acid sequence of SEQ ID NO: 7.
  • the anti-TIGIT antibody or fragment thereof includes a heavy chain constant region 1 (CH1) comprising the amino acid sequence of SEQ ID NO: 3 and a light chain constant region comprising the amino acid sequence of SEQ ID NO: 8 or SEQ ID NO: 24. (CL) may be included.
  • IL-2 or “interleukin-2” include mammals, such as primates (e.g., humans) and rodents (e.g., mice and rats), unless otherwise noted. refers to any wild type IL-2 obtained from any vertebrate source.
  • the IL-2 may be obtained from animal cells, but also includes those obtained from recombinant cells capable of producing IL-2. Additionally, the IL-2 may be wild-type IL-2 or a variant thereof.
  • IL-2 or its variants may be collectively used interchangeably with the terms “IL-2 protein,” “IL-2 polypeptide,” or “IL-2.”
  • IL-2, IL-2 protein, IL-2 polypeptide and IL-2 variants for example, bind specifically to the IL-2 receptor. The specific binding can be confirmed through methods known to those skilled in the art.
  • the IL-2 may be in a mature form. Specifically, the mature IL-2 may not contain a signal sequence and may contain a truncated fragment of the N-terminus or C-terminus of wild-type IL-2. At this time, the IL-2 may have the amino acid sequence of SEQ ID NO: 17.
  • IL-2 variant refers to a form in which some amino acids of the full-length IL-2 or fragment of IL-2 are substituted. That is, the IL-2 variant may have an amino acid sequence that differs from wild-type IL-2 or a fragment thereof. However, the IL-2 variant may have equivalent or similar activity to wild-type IL-2.
  • IL-2 activity may mean, for example, specific binding to the IL-2 receptor, and this specific binding can be measured through methods known to those skilled in the art.
  • the IL-2 variant may be one in which some amino acids of wild-type IL-2 have been substituted.
  • One specific example of an IL-2 variant resulting from amino acid substitution may be one in which at least one of the 38th, 42nd, 45th, 61st, and 72nd amino acids in the amino acid sequence of SEQ ID NO: 17 is substituted. According to one embodiment, three amino acids may be substituted as long as IL-2 activity is maintained.
  • the IL-2 variant may be one in which amino acids at the 38th, 42nd, and 61st positions in the amino acid sequence of SEQ ID NO: 17 are substituted.
  • the IL-2 variant may have at least one substitution selected from the group consisting of R38A, F42A, Y45A, E61R, and L72G in the amino acid sequence of SEQ ID NO: 17.
  • the IL-2 variant may have R38A, F42A, and E61R substitutions in the amino acid sequence of SEQ ID NO: 17.
  • the IL-2 variant may have the amino acid sequence of SEQ ID NO:6.
  • linker An antibody or fragment thereof that specifically binds to the TIGIT; And the IL-2 protein or its variant may be bound by a linker or carrier.
  • linker and carrier are also used interchangeably.
  • One specific example of the linker may include 1 to 50 amino acids, albumin or a fragment thereof, Fc (region), etc.
  • the Fc may be the Fc domain of an immunoglobulin.
  • the Fc region of the immunoglobulin includes the heavy chain constant region 2 (CH2) and the heavy chain constant region 3 (CH3) of the immunoglobulin, but does not include the variable regions of the heavy and light chains and the light chain constant region (CL) of the immunoglobulin. refers to proteins that do not
  • the immunoglobulin Fc region may be derived from IgG, IgA, IgE, IgD or IgM.
  • the immunoglobulin Fc region may be IgG1, IgG2, IgG3, or IgG4, which are subclasses of IgG, and preferably may be derived from IgG4.
  • the Fc domain of the immunoglobulin may be not only a wild-type Fc domain, but also an Fc domain variant.
  • the term "Fc domain variant" as used herein refers to a glycosylation pattern that is different from that of the wild-type Fc domain, has an increased sugar chain compared to the wild-type Fc domain, has a decreased sugar chain compared to the wild-type Fc domain, or has a sugar chain removed ( It may be in a deglycosylated form. Additionally, an aglycosylated Fc domain is also included.
  • the Fc domain or variant may have an adjusted number of sialic acids, fucosylation, and glycosylation through culture conditions or genetic manipulation of the host.
  • the sugar chain of the Fc domain of an 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 mixture of the Fc regions of immunoglobulins IgG, IgA, IgE, IgD, or IgM.
  • the Fc domain variant may be a form in which some amino acids of the Fc domain are replaced with other amino acids.
  • the Fc region may have the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 23.
  • the bispecific antibody of the present invention is a fusion protein comprising the light chain variable region (VL) and light chain constant region (CL) of an anti-TIGIT antibody, and the heavy chain variable region (VH) and heavy chain constant region 1 (CH1) of an anti-TIGIT antibody.
  • the bispecific antibody of the present invention is a fusion protein comprising the light chain variable region, light chain constant region, and IL-2 protein of an anti-TIGIT antibody, and the heavy chain variable region, heavy chain constant region 1 (CH1), and Fc of the anti-TIGIT antibody. It may contain a fusion protein containing a domain.
  • the bispecific antibody comprising the anti-TIGIT antibody and IL-2 protein or a variant thereof has the following structural formulas (I) and (II); Or it may include (III) and (IV).
  • N' is the N terminus
  • the X is the antigen binding site of the heavy chain of the anti-TIGIT antibody and includes a variable region (VH) and a CH1 region,
  • the X' is the light chain antigen binding site of the anti-TIGIT antibody and includes a variable region (VL) and a constant region (CL),
  • Y is IL-2 protein or a variant thereof
  • linker (1), linker (2), and linker (3) are each independently peptide linkers
  • the o, p and q are each independently O or 1.
  • anti-TIGIT antibody antigen binding site, variable region, constant region; IL-2 protein, IL-2 variant, and Fc region (or Fc domain) are as described above.
  • the IL-2 protein or a variant thereof may be bound to the C terminus of the Fc region or the C terminus of the light chain constant region of the anti-TIGIT antibody.
  • the IL-2 protein or its variant and the Fc region or light chain constant region may be combined through a peptide linker.
  • the peptide linker (1) may be composed of 1 to 50 consecutive amino acids, or 3 to 30 consecutive amino acids, or 5 to 15 amino acids. In one embodiment, the peptide linker (1) may be composed of 12 amino acids. Additionally, the peptide linker 1 may include at least one cysteine. Specifically, it may contain one, two or three cysteines. Additionally, the peptide linker (1) may be derived from the hinge of immunoglobulin. For example, the hinge can be selected from the hinge regions of various IgG subtype antibodies. Additionally, the hinge may be a form in which some amino acids in the hinge region derived from immunoglobulin are replaced with other amino acids, or may be a sequence in which some amino acid sequences are added. In one embodiment, the peptide linker (1) may be a peptide linker consisting of the amino acid sequence of SEQ ID NO: 18.
  • the peptide linker (2) or linker (3) may be composed of 1 to 30 consecutive amino acids, or 2 to 20 consecutive amino acids, or 2 to 10 amino acids.
  • the peptide linker (2) or linker (3) may be (G4S)n (where n is an integer of 1 to 10). At this time, n in (G4S)n may be 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • the peptide linker (2) or linker (3) may be a peptide linker consisting of the amino acid sequence of SEQ ID NO: 5.
  • Tables 1 and 2 The amino acid sequences of each region constituting the bispecific antibody are shown in Tables 1 and 2 below. Specifically, Table 1 describes the amino acid sequences of anti-hTIGIT HC-hIgG4 Fc-hIL2v3 and anti-hTIGIT LC (lambda). Additionally, Table 2 describes the amino acid sequences of anti-hTIGIT HC-hIgG4FcM1 and anti-hTIGIT LC(kappa)-hIL2v3.
  • Anti-hTIGIT HC-hIgG4Fc- hIL2v3 signal peptide (mIgG) MEWSWVFLFFLSVTTGVHS One Anti-hTIGIT VH QVQLQESGPGLVKPSGTLSLTCAVS GVSIRQGHW WSWVRQPPGKGLEWIGE IYQTGRT NYNPSLKSRVTISVGKSRNHISLKLSSVTAADTAVYYC TTGSGWYPIDY WGQGTLVTVSS 2 Anti-hTIGIT CH1 ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRV 3 first linker ESKYGPPCPPCP 18 F02(hIgG4 Fc) APEAAGGPSVFLFPPKPKDQLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNA
  • Anti-hTIGIT HC-hIgG4FcM1 signal peptide (mIgG) MEWSWVFLFFLSVTTGVHS One Anti-hTIGIT VH QVQLQESGPGLVKPSGTLSLTCAVS GVSIRQGH WWSWVRQPPGKGLEWIGE IYQTGRT NYNPSLKSRVTISVGKSRNHISLKLSSVTAADTAVYYC TTGSGWYPIDY WGQGTLVTVSS 2 Anti-hTIGIT CH1 ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRV 3 first linker ESKYGPPCPPCP 18 F02K(hIgG4 FcM1) APEAAGGPSVFLFPPKPKDQLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPRE
  • Another aspect of the invention is an anti-TIGIT antibody or fragment thereof; and a polynucleotide encoding a bispecific antibody comprising the IL-2 protein.
  • the anti-TIGIT antibody, fragment of anti-TIGIT antibody, IL-2 protein and bispecific antibody are the same as described above.
  • the polynucleotide may include the base sequence of SEQ ID NO: 32, which encodes the heavy chain variable region of an anti-TIGIT antibody. Additionally, the polynucleotide may include the base sequence of SEQ ID NO: 33, which encodes the light chain variable region of an anti-TIGIT antibody. In one embodiment of the present invention, the polynucleotide encoding the dual-specific antibody may include the base sequences of SEQ ID NO: 15 and SEQ ID NO: 16. In one embodiment of the present invention, the polynucleotide encoding the dual-specific antibody may include the base sequences of SEQ ID NO: 27 and SEQ ID NO: 28.
  • one or more bases may be mutated by substitution, deletion, insertion, or a combination thereof.
  • synthesis methods well known in the art, for example, methods described in the literature (Engels and Uhlmann, Angew Chem IntEd Engl., 37:73-127, 1988), can be used. and triester, phosphite, phosphoramidite and H-phosphate methods, PCR and other autoprimer methods, and oligonucleotide synthesis methods on solid supports.
  • the polynucleotide is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, at least about 88% of the base sequence of SEQ ID NO: 15. %, 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 is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, at least about 88% of the base sequence of SEQ ID NO: 16. %, 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 has the base sequence of SEQ ID NO: 27 and at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, and 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 has the base sequence of SEQ ID NO: 28 and at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, and 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 signal sequence or leader sequence.
  • signal sequence used herein refers to a nucleic acid encoding a signal peptide that directs secretion of a target protein.
  • the signal peptide is cleaved after translation in the host cell.
  • the signal sequence of the present invention is a polynucleotide encoding an amino acid sequence that initiates the movement of a protein across the ER (endoplasmic reticulum) membrane.
  • 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 while the immature polypeptide moves.
  • 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), IgG signal sequence, or growth hormone secretion signal sequence.
  • a secretion signal sequence used in higher eukaryotic cells, including mammals can be used.
  • Signal sequences useful in the present invention include antibody light chain signal sequences, such as antibody 14.18 (Gillies et al., J. Immunol. Meth 1989. 125:191-202), and antibody heavy chain signal sequences, such as MOPC141 antibody heavy chain signal.
  • the signal sequence may include the amino acid sequence of SEQ ID NO: 1.
  • the anti-TIGIT antibody or fragment thereof is the anti-TIGIT antibody or fragment thereof; and a vector loaded with a polynucleotide encoding a bispecific antibody containing the IL-2 protein.
  • the polynucleotide may include the base sequence of SEQ ID NO: 32, which encodes the heavy chain variable region of the anti-TIGIT antibody, and SEQ ID NO: 33, which encodes the light chain variable region of the anti-TIGIT antibody.
  • the bispecific antibody may be encoded by the base sequences of SEQ ID NO: 15 and SEQ ID NO: 16, or may be encoded by the base sequences of SEQ ID NO: 27 and SEQ ID NO: 28.
  • the term “vector” can be introduced into a host cell and recombined and inserted into the host cell genome.
  • the vector is understood as a nucleic acid vehicle containing a nucleotide sequence capable of spontaneous replication as an episome.
  • the vectors include linear nucleic acids, plasmids, phagemids, cosmids, RNA vectors, viral vectors, mini-chromosomes and analogs thereof.
  • 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.), E. coli-derived plasmids (pYG601BR322, pBR325, pUC118, pUC119, etc.), Bacillus subtilis.
  • plasmids pUB110, pTP5, etc.
  • yeast-derived plasmids yeast-derived plasmids
  • phage DNA Charon4A, Charon21A, EMBL3, EMBL4, ⁇ gt10, ⁇ gt11, ⁇ ZAP, etc.
  • animal virus vectors retroviruses
  • adenovirus vaccinia virus
  • insect virus vectors baculovirus, etc.
  • the plasmid may contain a selection marker, such as an antibiotic resistance gene, and host cells maintaining the plasmid may be cultured under selective conditions.
  • a selection marker such as an antibiotic resistance gene
  • the term “gene expression” or “expression” of a protein of interest is understood to mean transcription of a DNA sequence, translation of an mRNA transcript, and secretion of a bispecific antibody product or fragment thereof.
  • a useful expression vector may be RcCMV (Invitrogen, Carlsbad) or variants thereof.
  • the expression vector may include a human CMV (cytomegalovirus) promoter to promote continuous transcription of the target gene in mammalian cells and a bovine growth hormone polyadenylation signal sequence to increase the steady-state level of RNA after transcription. You can.
  • Another aspect of the invention is the anti-TIGIT antibody or fragment thereof; and an expression vector containing a polynucleotide encoding a bispecific antibody containing the IL-2 protein is provided.
  • the anti-TIGIT antibody, anti-TIGIT antibody fragment, IL-2 protein, and bispecific antibody are the same as described above.
  • transformed cell refers to prokaryotic cells and eukaryotic cells into which a recombinant expression vector can be introduced.
  • the transformed cells can be produced by introducing a vector into a host cell and transforming it.
  • the bispecific antibody of the present invention can be produced by expressing the polynucleotide contained in the vector.
  • the transformation can be performed by various methods. There is no particular limitation thereto, as long as the bispecific antibody of the present invention can be produced.
  • the transformation method is a CaCl 2 precipitation method, which increases efficiency by using a reducing substance called DMSO (dimethyl sulfoxide) in the CaCl 2 precipitation method.
  • DMSO dimethyl sulfoxide
  • Hanahan method, electroporation, calcium phosphate precipitation, protoplast fusion method, stirring method using silicon carbide fiber, Agrobacteria-mediated transformation method, transformation method using PEG, dextran sulfate, lipofectamine and Drying/inhibition-mediated transformation methods, etc. may be used.
  • the target can be delivered into cells using virus particles through infection. Additionally, vectors can be introduced into host cells by gene bombardment or the like.
  • the host cell used to produce the transformed cell is not particularly limited as long as it is capable of producing the antibody of the present invention.
  • the host cells may include, but are not limited to, cells of prokaryotic, eukaryotic, mammalian, plant, insect, fungal or cellular origin.
  • An example of the prokaryotic cell may be Escherichia coli.
  • yeast can be used as an example of a eukaryotic cell.
  • the mammalian cells include CHO cells, F2N cells, COS cells, BHK cells, Bowes melanoma cells, HeLa cells, 911 cells, AT1080 cells, A549 cells, SP2/0 cells, and human lymphoblastoids.
  • NSO cells HT-1080 cells, PERC.6 cells, HEK293 cells, or HEK293T cells can be used, but are not limited to these, and any cell that can be used as a mammalian host cell known to those skilled in the art can be used.
  • the glycosylation-related genes of the host cell are manipulated through methods known to those skilled in the art to manipulate the antibody's sugar chain pattern (e.g., sialic acid, fucose). misfire, saccharification) can be adjusted.
  • the antibody's sugar chain pattern e.g., sialic acid, fucose. misfire, saccharification
  • Another aspect of the invention is the anti-TIGIT antibody or fragment thereof; and a method for producing a bispecific antibody comprising the IL-2 protein.
  • the method for producing the bispecific antibody includes i) culturing the transformed cells; and ii) obtaining a bispecific antibody.
  • the anti-TIGIT antibody, anti-TIGIT antibody fragment, IL-2 protein, and bispecific antibody are the same as described above.
  • culture refers to a method of growing microorganisms under appropriately artificially controlled environmental conditions.
  • the method of culturing the transformed cells can be performed using methods widely known in the art.
  • the culture is not particularly limited as long as it can be produced by expressing the antibody of the present invention.
  • the culture may be continuously cultured in a batch process or fed batch or repeated fed batch process.
  • the step of obtaining the antibody from the culture may be performed by methods known in the art.
  • the obtaining method is not particularly limited as long as the produced bispecific antibody of the present invention can be obtained.
  • the obtaining method includes centrifugation, filtration, extraction, spraying, drying, evaporation, precipitation, crystallization, electrophoresis, fractional dissolution (e.g. ammonium sulfate precipitation), chromatography (e.g. ion exchange, It may be a method such as affinity, hydrophobicity, and size exclusion).
  • the anti-TIGIT antibody or fragment thereof; and a bispecific antibody containing IL-2 protein as an active ingredient provides a pharmaceutical composition for preventing or treating cancer.
  • the anti-TIGIT antibody, fragment of anti-TIGIT antibody, IL-2 protein and bispecific antibody are the same as described above.
  • cancer is classified as a disease in which normal tissue cells proliferate indefinitely for some reason and continue to grow rapidly regardless of the living phenomenon of the living body or the condition of surrounding tissues, etc.
  • Cancer in the present invention is Various cancers of the human body, such as stomach cancer, liver cancer, lung cancer, colon cancer, breast cancer, prostate cancer, ovarian cancer, pancreas cancer, cervical cancer, thyroid cancer, laryngeal cancer, acute myeloid leukemia, brain tumor, neuroblastoma, retinoblastoma, head and neck cancer, salivary gland cancer, and It may be any cancer selected from the group consisting of lymphoma, but is not limited to the above types. Additionally, for the purposes of the present invention, cancer may be resistant to radiation, but is not limited thereto.
  • the bispecific antibody may be included in an arbitrary amount (effective amount) depending on the use, formulation, formulation purpose, etc., as long as it exhibits anti-cancer activity or, in particular, can exhibit a cancer treatment effect.
  • a typical effective amount will be determined within the range of 0.001% by weight to 20.0% by weight based on the total weight of the composition.
  • “effective amount” refers to the amount of an active ingredient that can improve the condition of a disease or induce a treatment effect, especially an improvement in the condition of cancer or a treatment effect. Such effective amounts can be determined experimentally within the scope of the ordinary ability of those skilled in the art.
  • treatment can be used to include both therapeutic treatment and preventive treatment, and includes both application and any form of medication to treat diseases in mammals, including humans.
  • the term also includes inhibiting or slowing the progression of a disease; Restoring or repairing damaged or missing function, thereby partially or completely relieving a disease; or stimulating inefficient processes; It includes the meaning of alleviating serious diseases.
  • prevention may be used to mean alleviating or reducing the pathological condition or disease of an individual.
  • “enhanced efficacy” e.g., improvement in efficacy
  • improved efficacy measured by comparing parameters such as clearance rate and treatment or amelioration of cancer disease in test animals or human subjects. It can be.
  • the pharmaceutical composition of the present invention is administered in a “therapeutically effective amount.”
  • administration means introducing a predetermined substance into an individual by an appropriate method, and the composition may be administered through any general route as long as it can reach the target tissue. It may be administered intraperitoneally, intravenously, intramuscularly, subcutaneously, intradermally, orally, topically, intranasally, intrapulmonaryly, or rectally, but is not limited thereto.
  • the term "therapeutically effective amount” or “pharmaceutically effective amount” refers to the amount of a compound or composition effective in preventing or treating a target disease, which means treating the disease at a reasonable benefit/risk ratio applicable to medical treatment. It refers to an amount that is sufficient for treatment and does not cause side effects.
  • the level of the effective amount is determined by factors including the patient's health status, type and severity of the disease, activity of the drug, sensitivity to the drug, administration method, administration time, administration route and excretion rate, treatment period, combination or concurrent use of drugs, and It may be determined based on other factors well known in the medical field.
  • a therapeutically effective amount refers to an amount of drug that is effective in treating cancer.
  • the pharmaceutical composition may further include a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier may be any carrier that is a non-toxic material suitable for delivery to a patient. Distilled water, alcohol, fats, waxes and inert solids may be included as carriers. Pharmacologically acceptable adjuvants (buffers, dispersants) may also be included in the pharmaceutical composition.
  • the pharmaceutical composition may be prepared into a parenteral formulation according to the route of administration by a conventional method known in the art, including a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means that it does not inhibit the activity of the active ingredient and does not have toxicity beyond what is acceptable for the subject of application (prescription).
  • the pharmaceutical composition When the pharmaceutical composition is prepared as a parenteral formulation, it can be formulated in the form of injections, transdermal administration, nasal inhalation, and suppositories along with a suitable carrier according to methods known in the art.
  • suitable carriers include sterile water, ethanol, polyols such as glycerol or propylene glycol, or mixtures thereof, preferably Ringer's solution, phosphate buffered saline (PBS) containing triethanol amine, or sterile for injection. Isotonic solutions such as water or 5% dextrose can be used.
  • PBS phosphate buffered saline
  • Isotonic solutions such as water or 5% dextrose can be used.
  • the preferred dosage of the pharmaceutical composition varies depending on the patient's condition and weight, degree of disease, drug form, administration route and period, but can be appropriately selected by a person skilled in the art.
  • the preferred dosage of the pharmaceutical composition may range from 0.0001 ⁇ g/kg to 100 g/kg per day depending on the patient's condition, weight, gender, age, patient's severity, and administration route. Administration can be done once a day or divided into several times. These dosages should not be construed as limiting the scope of the invention in any respect.
  • the subjects to which the pharmaceutical composition can be applied are mammals and humans, and humans are particularly preferred.
  • the pharmaceutical composition of the present invention may further include any compounds or natural extracts known to have cancer treatment effects.
  • Another aspect of the present invention is the anti-TIGIT antibody or fragment thereof for preparing a medicament for preventing or treating cancer; and the use of a bispecific antibody comprising the IL-2 protein.
  • the cancer, prevention, treatment, anti-TIGIT antibody, fragment of anti-TIGIT antibody, IL-2 protein and bispecific antibody are the same as described above.
  • Another aspect of the present invention is the anti-TIGIT antibody or fragment thereof for preventing or treating cancer; and the use of a bispecific antibody comprising the IL-2 protein.
  • the cancer, prevention, treatment, anti-TIGIT antibody, fragment of anti-TIGIT antibody, IL-2 protein and bispecific antibody are the same as described above.
  • Another aspect of the invention is the anti-TIGIT antibody or fragment thereof; and administering to a subject a bispecific antibody containing the IL-2 protein.
  • a subject may be a mammal, preferably a human.
  • the individual may be a patient suffering from cancer or an individual with a high risk of suffering from cancer.
  • the bispecific antibody may be administered in combination with any compound or natural extract known to have a cancer treatment effect, or may be formulated in the form of a combination preparation with other drugs.
  • the vector was introduced into CHO cells (Expi-CHO TM , Thermo Fisher Scientific) to express bispecific antibodies. After introducing the vector, it was cultured at 37°C, 125 rpm, and 8% CO 2 conditions. Afterwards, when the cell survival rate was 50%, the culture medium was collected and the bispecific antibody was purified. The purified bispecific antibody was named “GI-106” ( Figure 1).
  • the bispecific antibody was collected with 50 mM glycine, pH 3.4.
  • the buffer containing the collected bispecific antibodies was changed to PBS buffer using dialysis, and the concentration was measured.
  • NanoDrop equipment Thermo Fisher Scientific
  • the molecular weight of the separated and purified bispecific antibody (GI-106) was confirmed through SDS-PAGE under reduced (R, reduced) or non-reduced (NR, non-reduced) conditions ( Figure 2), and size exclusion chromatography (SEC). ) Purity was confirmed through analysis. As a result, the purity of the separated and purified bispecific antibody was confirmed to be 87.42%.
  • a bispecific antibody comprising an anti-TIGIT antibody that specifically binds to TIGIT and an IL-2 variant, the signal peptide (SEQ ID NO: 1), the variable region (SEQ ID NO: 2) of the anti-TIGIT antibody heavy chain and the constant region (SEQ ID NO: 3), a polynucleotide (SEQ ID NO: 36) encoding the linker (1) (SEQ ID NO: 18) and an IgG4 FcM1 domain (SEQ ID NO: 23) and a signal peptide (SEQ ID NO: 1) of the anti-TIGIT antibody light chain.
  • the vector was introduced into CHO cells (Expi-CHO TM , Thermo Fisher Scientific) to express bispecific antibodies. After introducing the vector, it was cultured for 7 days at 37°C, 127 rpm, 5% CO 2 , and 80% humidity. Afterwards, the culture medium was collected and the bispecific antibody was purified. The purified bispecific antibody was named “GI-106B7NH06Kv3” ( Figure 3).
  • the bispecific antibody As a specific purification method for the bispecific antibody, it was purified using affinity chromatography containing Protein A resin. After removing impurities including cell debris generated during the cell culture process through filtration, the filtered culture medium was flowed through the column and combined. Afterwards, a buffer (pH 3.0) containing 100mM glycine and 100mM arginine was sequentially flowed to collect the bispecific antibody. The buffer containing the collected bispecific antibodies was changed to a buffer containing 4% sucrose, 50 mM histidine, and 50 mM arginine (pH 7.4) using dialysis, and the concentration was measured. When detected using Nanodrop equipment, it was confirmed that a bispecific antibody was included at a concentration of 1.87 mg/mL.
  • the molecular weight of the separated and purified bispecific antibody (GI-106B7NH06Kv3) was confirmed by SDS-PAGE under reducing (R, reduced) or non-reducing (NR, non-reduced) conditions ( Figure 4), and size exclusion chromatography (SEC). Purity was confirmed through analysis. As a result, the purity of the separated and purified bispecific antibody was confirmed to be 88.24%.
  • the polynucleotide (SEQ ID NO: 35) encoding was synthesized through GenScript's Gene Synthesis service and loaded into the pcDNA3.4 vector.
  • the vector was introduced into CHO cells (Expi-CHO TM , Thermo Fisher Scientific) to express the control anti-TIGIT antibody.
  • the culture medium was cultured at 37°C, 125 rpm, 8% CO 2 , and when the cell survival rate was 50%, the culture medium was collected and the control anti-TIGIT antibody was purified. Purification was performed in the same manner as Example 1.1.
  • Signal peptide (SEQ ID NO: 1), encoding the variable region (SEQ ID NO: 2) and constant region (SEQ ID NO: 3) of the anti-TIGIT antibody heavy chain, linker (1) (SEQ ID NO: 18), and IgG4 FcM1 domain (SEQ ID NO: 23)
  • a polynucleotide (SEQ ID NO: 36), a signal peptide (SEQ ID NO: 1), and a polynucleotide (SEQ ID NO: 38) encoding the variable region (SEQ ID NO: 7) and constant region (SEQ ID NO: 24) of the anti-TIGIT antibody light chain were used as BioXp. It was loaded into pCGS3 vector (Sigma-Aldrich®) using TM 3250 SYSTEM.
  • the vector was introduced into CHO cells (Expi-CHO TM , Thermo Fisher Scientific) to express the fusion protein. After introducing the vector, it was cultured for 7 days at 37°C, 127 rpm, 5% CO 2 , and 80% humidity. Afterwards, the culture medium was collected and the fusion protein was purified. The purified fusion protein was named “GI-106B7NH06K-CN”.
  • SEQ ID NO: 1 Signal peptide (SEQ ID NO: 1), linker (1) (SEQ ID NO: 18) and IgG4 FcM1 domain (SEQ ID NO: 23), linker (2) (SEQ ID NO: 5) and an IL-2 variant with three amino acids substituted (SEQ ID NO: 6)
  • the polynucleotide containing (SEQ ID NO: 39) was loaded into the pCGS3 vector (Sigma-Aldrich®) using BioXP TM 3250 SYSTEM.
  • the vector was introduced into CHO cells (Expi-CHO TM , Thermo Fisher Scientific) to express the fusion protein. After introducing the vector, it was cultured for 7 days at 37°C, 127 rpm, 5% CO 2 , and 80% humidity. Afterwards, the culture medium was collected and the Fc-IL2v3 fusion protein was purified.
  • Example 2 In vitro environment ( in vitro ) confirmed the ability of GI-106 to inhibit TIGIT/CD155 binding
  • This experiment evaluated the effect of TIGIT inhibition upon treatment with GI-106 and control anti-TIGIT antibody in an in vitro environment. Specifically, the experiment was conducted using the TIGIT/CD155 blockade bioassay kit (cat. J2205, Promega). TIGIT effector cells and CD155 aAPC/CHO-K1 cells were used in this experiment, and the blocking of TIGIT/CD155 interaction and activation of the CD226 signaling pathway due to anti-TIGIT antibody treatment was measured by quantifying the degree of luminescence. (Figure 5).
  • TIGIT effector cells stored in liquid nitrogen were dissolved in a constant temperature water bath at 37°C for 3 minutes and then suspended in 0.5 mL of preheated 12 mL assay buffer (90% RPMI 1640 (Promega) + 10% FBS (Promega)). Afterwards, 80 ⁇ L of the suspension was added to each well in a 96-well-white cell culture plate (cat. 3917, Corning), and then incubated at 37°C and 5% CO 2 conditions. It was cultured in an incubator for 20 hours.
  • GI-106 and the control anti-TIGIT antibody were diluted using assay buffer, and 20 ⁇ L of test substances at various concentrations were added to each well.
  • 20 ⁇ L of assay buffer was added for the negative control.
  • the 96-well plate was left at room temperature until aAPC/CHO-K1 cells expressing CD155 were dispensed.
  • CD155-expressing aAPC/CHO-K1 cells stored in liquid nitrogen were dissolved in a water bath at 37°C for 3 minutes, then suspended in 0.5 mL of preheated 3 mL assay buffer, followed by TIGIT effector cells and GI-106 or control antibody. 20 ⁇ L was dispensed into each well in a 96-well plate. For the negative control, 20 ⁇ L of assay buffer was added to make the final volume the same. Afterwards, it was cultured for 6 hours in an incubator under 37°C and 5% CO 2 conditions.
  • the 96-well plate was removed from the incubator and left at room temperature for 15 minutes. Afterwards, 120 ⁇ L of Bio-GloTM reagent (Promega) pre-dissolved at room temperature was added to each well, and the same amount was also added to the edge wells for background signal correction. After reacting at room temperature for 10 minutes, the degree of luminescence was measured using a Glomax Luminometer (cat. GM3000, Promega), and the results were expressed in a RLU (Relative Light Unit) graph.
  • Bio-GloTM reagent Promega
  • GI-106 and anti-TIGIT antibodies bind to TIGIT expressed in TIGIT effector cells in a concentration-dependent manner, increasing the luminescence signal.
  • the anti-TIGIT antibody of GI-106 binds to TIGIT expressed on the surface of the TIGIT effector cell, thereby inhibiting the binding to CD155, and as a result, CD155 binds to CD226 to activate signal transduction and increase the luminescent signal ( Figure 6).
  • This experiment is to confirm the activity of the IL-2 variant portion of GI-106. Specifically, the experiment was conducted using HEK-Blue TM IL-2 reporter cells (cat. hkb-il2, InvivoGen Inc.). HEK-Blue TM IL-2 reporter cells are induced to produce the reporter protein SEAP (secreted embryonic alkaline phosphatase) protein when the JAK-STAT pathway is activated by IL-2. In this experiment, the activation ability by IL-2 was confirmed by detecting SEAP protein.
  • SEAP secreted embryonic alkaline phosphatase
  • HEK-Blue TM IL-2 reporter cells were incubated with 10% FBS (Gibco), 100 U/mL penicillin (Welgene Inc.), 100 ⁇ g/mL streptomycin (Welgene Inc.), and 100 ⁇ g/mL Normocin TM (cat. Ant. -nr-1, InvivoGen Inc.) was cultured in DMEM medium (Gibco) containing.
  • HEK-Blue TM IL-2 reporter cells were subcultured to stabilize the cells, and then the cells were harvested using trypsin (Gibco). Afterwards, dead cells were removed by washing with PBS. The separated cells were created into a cell suspension in the culture medium so that the concentration was about 2.8 ⁇ 10 5 cells/mL.
  • GI-106 Aldesleukin (Product name: Proleukin®, Novartis, hereinafter referred to as Proleukin) as a positive control
  • Proleukin recombinant human TGF- ⁇ 1
  • a well-plate cat. 30096, SPL
  • 180 ⁇ L of the cell suspension prepared in the 96-well plate containing the test substance was added to each well to obtain a cell count of approximately 5 ⁇ 10 4 and cultured in an incubator at 37°C and 5% CO 2 for 24 hours.
  • the 96-well plate was removed from the incubator, centrifuged at 300 ⁇ g for 5 minutes, and 20 ⁇ L of the supernatant was transferred to a new 96-well plate.
  • 180 ⁇ L of QUANTI-Blue TM solution (cat. Rep-qbs, InvivoGen Inc,) dissolved at room temperature was dispensed into each well containing the supernatant, and then reacted in an incubator at 37°C and 5% CO 2 for 2 hours. . After the reaction, the absorbance was measured at a wavelength of 630 nm using a spectrophotometer (VersaMax TM Absorbance Microplate Reader).
  • This experiment evaluated the tumor growth inhibition effect after administering the test substance GI-106 in a tumor model in which MDA-MB-231 (human breast cancer cells) cells were transplanted into mice with a human immune system.
  • mice a suspension of human peripheral blood cells (StemExpress, LLC) was added to NSG-B2m female mice (7 weeks old, The jackson laboratory) at 1 ⁇ 107 cells/200 ⁇ L per mouse using a disposable syringe ( 31G, cat. 328820, BD medical diabetes care) and administered through the caudal vein. After cell transplantation, general symptoms were observed once daily.
  • stemExpress, LLC human peripheral blood cells
  • MDA-MB-231 cells were purchased from the Korea Cell Line Bank (KCLB No. 30026) and cultured in RPMI1640 medium (Gibco) containing 10% FBS (Gibco) and 1% antibiotic/antifungal agent (Gibco). Cultured MDA-MB-231 cells were harvested using trypsin (Gibco) and suspended in PBS. To establish a xenograft mouse tumor model, healthy mice 5 days after human peripheral blood cell transplantation were incubated with MDA-MB-231 cell suspension (5 ⁇ 10 6 cells/0.05 mL) and 0.05 mL BD Matrigel matrix phenol red-free (cat. 356237).
  • the tumor volume was measured on mice with no abnormalities in the animal's health, and 12 individuals were selected so that the average of each group reached 40 ⁇ 80 mm 3 .
  • Subjects were selected considering their physiological status (respiration, fur, behavior, tail, posture, body fluids, diet, deformation, metabolism, etc.), weight change, FACS analysis results, and tumor growth rate.
  • the selected animals were divided into 6 animals per group to be as equal as possible based on tumor volume and body weight. As shown in Table 3, the test group was formed and the test substance was administered.
  • Tumor volume was measured twice a week during the observation period using a caliper (digital caliper, mitutoyo) to measure the long axis (L, maximum length) and short axis (W, perpendicular width) of the tumor, and substitute them into Equation I below.
  • Tumor volume (TV) and tumor growth inhibition (TGI) were measured.
  • TGI (1-(Ti-T0)/(Vi-V0)) ⁇ 100
  • T0 Tumor volume of the test substance administered group at the time of group separation
  • V0 Tumor volume of the negative control group at the time of group separation
  • the tumor volume before administration of each subject was set to the value measured at the time of group separation, and the antitumor efficacy was evaluated by comparison with the control group (vehicle (PBS), G1).
  • tumor growth in the GI-106 administered group was inhibited compared to the control group (vehicle (PBS)) (FIGS. 8 to 11).
  • PBS vehicle
  • the tumor growth inhibition rate there was only one mouse in the PBS-treated group with a tumor growth inhibition rate of more than 30%, and there were no mice with tumor growth inhibition rates of more than 50% or more than 80%.
  • the GI-106 administration group there were 5 mice with a tumor growth inhibition rate of more than 30%, 3 mice with a tumor growth inhibition rate of 50% or more, and 2 mice with a tumor growth inhibition rate of 80% or more (Figure 12).
  • This experiment evaluated the tumor growth inhibition effect after administering the test substance GI-106B7NH06Kv3 in a BALB/c mouse tumor model transplanted with EMT-6 cells, a mouse breast cancer cell line derived from BALB/c mice. .
  • a suspension of EMT-6 cells, a mouse-derived breast cancer cell was administered to BALB/c female mice (7 weeks old, Orient Bio) at a rate of 2 ⁇ 104 cells/40 ⁇ L per mouse using a disposable syringe (31G, cat. 328820, BD) was used to open the left side of the animal's abdomen and administer it into the mammalian fat pad.
  • a disposable syringe 31G, cat. 328820, BD
  • EMT-6 cells were purchased from Orient Bio and cultured in RPMI1640 medium (A1049101, Thermo Fisher Scientific) containing 10% FBS (16000-044, Thermo Fisher Scientific) and 1% antibiotic/antimycotic (15140122, Thermo Fisher Scientific). It was cultured in . Cultured EMT-6 cells were harvested using trypsin-EDTA (Cat. 25200-072, Thermo Fisher Scientific) and then suspended in PBS (Cat. LB 001-04, Welgene Inc.). EMT-6 cell suspension (2 ⁇ 10 6 cells/0.4 mL) for transplantation of 10 animals was filled into a disposable syringe (31G, cat. 328820, BD medical diabetes care), opened on the left side of the animal's abdomen, and administered into the mammalian fat body. . After cell transplantation, general symptoms were observed once daily.
  • the tumor volume was measured in mice with no abnormalities in the animal's health, and 20 individuals were selected so that the average of each group reached 70 ⁇ 72 mm 3 . Individuals were selected considering their physiological state (respiration, fur, behavior, tail, posture, body fluids, diet, shape deformation, metabolism, etc.), weight change, and tumor growth rate. The selected animals were separated into groups of 10 as evenly as possible based on tumor volume and body weight. As shown in Table 4, the test group was formed and the test substance was administered.
  • tumor volume was measured three times a week during the observation period using calipers to measure the long and short axes of the tumor, and the tumor volume and tumor growth inhibition rate were measured by substituting equation I in Example 4.
  • the tumor volume before administration of each subject was set to the value measured at the time of group separation, and the antitumor efficacy was evaluated by comparison with the control group (vehicle (hIgG4), G1). All statistical calculations were performed using Prism 8.0 (Graph Pad Software Inc.). Comparison of tumor volume measurements was done using unpaired t-test. A p value of less than 0.05 was considered significant.
  • the tumor growth inhibition rate it was confirmed that the tumor growth of the GI-106B7NH06Kv3 administration group was inhibited compared to the control group (vehicle (hIgG4)).
  • the negative control group there were 4 mice with a tumor growth inhibition rate of more than 30%, of which 2 mice showed a tumor growth inhibition rate of more than 50%, and among the two mice, one mouse showed a tumor growth inhibition rate of more than 80%. There was 1 animal.
  • mice with a tumor growth inhibition rate of more than 30% there were 9 mice with a tumor growth inhibition rate of more than 30%, and among them, 7 mice showed a tumor growth inhibition rate of more than 50%, and among the 7 mice, one showed a tumor growth inhibition rate of more than 80%. There were 6 mice. ( Figure 14).
  • This experiment evaluated the tumor growth inhibition effect after administering the test substance GI-106B7NH06Kv3 in a tumor model transplanted with MC38 cells, a murine colon cancer cell line derived from C57BL/6 mice.
  • a suspension of MC38 cells, mouse-derived colon cancer cells was administered to C57BL/6 female mice (6 weeks old, Orient Bio) at 5 ⁇ 10 5 cells/50 ⁇ L using a disposable syringe (31G, cat. 328820, It was filled with BD medical diabetes care and administered subcutaneously to the right back of the animal. After cell transplantation, general symptoms were observed once daily.
  • MC38 cells were purchased from Applied Stem Cell Inc. and cultured in RPMI1640 medium (Gibco) containing 10% FBS (Gibco) and 1% antibiotic/antimycotic (Gibco). Cultured MC38 cells were harvested using trypsin (Gibco) and suspended in PBS.
  • a solution prepared by mixing MC38 cell suspension (5 ⁇ 10 5 cells/0.025 mL) and 0.025 mL BD Matrigel matrix phenol red-free (cat. 356231, BD biosciences) was injected into healthy mice using a disposable syringe ( 31G, cat. 328820, BD medical diabetes care) and implanted by administering 0.05 mL per animal subcutaneously on the right back of the animal. After MC38 cell transplantation, general symptoms were observed once daily during the engraftment and growth period.
  • the tumor volume was measured in mice with no abnormalities in the animal's health, and 40 individuals were selected so that the average of each group reached 60 ⁇ 120 mm 3 . Individuals were selected considering their physiological state (respiration, fur, behavior, tail, posture, body fluids, diet, shape deformation, metabolism, etc.), weight change, and tumor growth rate. The selected animals were separated into groups of 10 as evenly as possible based on tumor volume and body weight. As shown in Table 5, the test group was formed and the test substance was administered.
  • tumor volume was measured three times a week during the observation period, using calipers to measure the long and short axes of the tumor, and the tumor volume (TV) and tumor growth inhibition rate (TGI) were measured by substituting Equation I in Example 4. did.
  • the tumor volume before administration of each subject was set to the value measured at the time of group separation, and the antitumor efficacy was evaluated by comparison with the control group (vehicle (hIgG4), G1). All statistical calculations were performed using Prism 8.0 (Graph Pad Software Inc.). Comparison of tumor volume measurements was done using unpaired t-test.
  • a p value of less than 0.05 was considered significant.
  • the tumor volume of the GI-106B7NH06Kv3 administration group was significantly reduced by about 3 times compared to the control group (vehicle (hIgG4)).
  • the tumor volume of the GI-106B7NH06Kv3 administration group was significantly reduced by about 2.5 times compared to the GI-106NH06K-CN administration group and the GI-106NH06K-CN and Fc-IL2v3 combination administration group (FIG. 15).
  • mice in the control group (vehicle (hIgG4)) with a tumor growth inhibition rate of more than 30%.
  • the GI-106NH06K-CN administration group there were 3 mice with a tumor growth inhibition rate of more than 30%, and among them, 2 mice showed a tumor growth inhibition rate of more than 50%, and among the two mice, one showed a tumor growth inhibition rate of more than 80%. There was only one mouse.
  • the GI-106NH06K-CN and Fc-IL2v3 administration groups there were no mice with a tumor growth inhibition rate of more than 30%.
  • mice were checked for the survival rate of mice in each group, in the control group (vehicle), 7 mice died on the 15th day, and only 3 mice survived until the 25th day, showing a survival rate of 30%.
  • the GI-106NH06K-CN administration group 2 mice began to die on the 15th day, and 1 more mouse each died on the 18th, 20th, and 22nd days, and 5 mice survived until the 25th day, resulting in a 50% survival rate. indicated.
  • mice began to die from day 6 and all mice died on day 22.
  • the GI-106B7NH06Kv3 administration group all mice survived until day 25, showing a survival rate of 100% (Figure 17).

Abstract

The present invention relates to a bispecific antibody including: an anti-TIGIT antibody or a fragment thereof; and an IL-2 protein. The bispecific antibody including an anti-TIGIT antibody and an IL-2 variant according to the present invention can modulate mechanisms related to TIGIT and has the same or similar functions as IL-2. That is, the bispecific antibody can not only regulate the binding of TIGIT and CD155, but also activate immune cells. In addition, the bispecific antibody significantly inhibited tumor growth in xenograft mouse tumor models and allograft mouse tumor models. Thus, the bispecific antibody can be used as an anticancer agent.

Description

항-TIGIT 항체를 포함하는 이중 특이적 항체 및 이의 용도Bispecific antibodies, including anti-TIGIT antibodies, and uses thereof
본 발명은 항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체 및 이를 이용한 암 치료 또는 예방용 약학 조성물에 관한 것이다.The present invention relates to an anti-TIGIT antibody or fragment thereof; and a bispecific antibody containing the IL-2 protein and a pharmaceutical composition for treating or preventing cancer using the same.
암 면역요법(cancer immunotherapy)은 체내 면역 작용을 이용하여 암을 치료하는 방법이다. 암 면역요법은 면역계가 암 세포 표면 단백질과 같은 항원들을 표적으로 이용하여 암 세포를 공격하도록 유발할 수 있다. 특히, 면역관문(immune checkpoint) 경로의 차단을 통해 항암 면역을 활성화시킬 수 있다는 점이 보고되었다. 면역관문은 종양 세포가 면역 회피를 초래하는 주요 메커니즘 중 하나이다. 따라서, 면역 체크포인트의 저해 또는 차단은 T 세포 활성화를 증가시킬 수 있고, 이에 의해 항 종양 면역을 강화할 수 있다.Cancer immunotherapy is a method of treating cancer using the body's immune system. Cancer immunotherapy can trigger the immune system to attack cancer cells by targeting antigens, such as cancer cell surface proteins. In particular, it has been reported that anti-cancer immunity can be activated through blocking the immune checkpoint pathway. Immune checkpoints are one of the main mechanisms by which tumor cells result in immune evasion. Therefore, inhibition or blockade of immune checkpoints can increase T cell activation, thereby enhancing anti-tumor immunity.
한편, IL-2는 활성화된 T 세포 중 특히, CD4+ 헬퍼 T 세포(helper T cell)에 의해 주로 합성된다. IL-2는 T 세포의 증식 및 분화를 자극하고, 세포독성 T 림프구(cytotoxic T lymphocyte, CTL)의 생성을 촉진한다. 그러나, IL-2는 면역세포의 증가 및 활성을 매개할 뿐만 아니라, 면역 관용(immune tolerance)을 유지하는데 중요하다는 점에서 면역 반응에서 이중적 기능을 갖는다. 또한, IL-2는 종양의 성장을 억제하는데 최적이 아닐 수 있는 것으로 보고되었다. 그 이유는 IL-2의 존재 시, 생성된 세포독성 T 림프구에 AICD(activation-induced cell death)가 일어날 수 있고, 면역 반응이 IL-2 의존성 조절 T 세포(Treg)에 의해 억제될 수 있기 때문이다(Imai et al., Cancer Sci 98, 416-423, 2007).Meanwhile, IL-2 is mainly synthesized by activated T cells, especially CD4+ helper T cells. IL-2 stimulates the proliferation and differentiation of T cells and promotes the production of cytotoxic T lymphocytes (CTL). However, IL-2 has a dual function in the immune response in that it not only mediates the increase and activity of immune cells, but is also important in maintaining immune tolerance. Additionally, it has been reported that IL-2 may be suboptimal for inhibiting tumor growth. This is because, in the presence of IL-2, activation-induced cell death (AICD) may occur in the generated cytotoxic T lymphocytes, and the immune response may be suppressed by IL-2-dependent regulatory T cells (Treg). (Imai et al., Cancer Sci 98, 416-423, 2007).
한편, TIGIT(T cell immunoreceptor with Ig and ITIM domains)은 면역관문 단백질(수용체)로서 수지상 세포(dendritic cell), 대식 세포(macrophage) 등에 존재하는 CD155와 결합하여 생체 내 T 세포 및 자연살해세포의 활성을 억제하는 것으로 알려져 있다.Meanwhile, TIGIT (T cell immunoreceptor with Ig and ITIM domains) is an immune checkpoint protein (receptor) that binds to CD155 present in dendritic cells, macrophages, etc. to activate T cells and natural killer cells in vivo. is known to inhibit.
이에, 본 발명자들은 면역세포의 활성을 증진시키는 새로운 조합의 이중 특이 항체를 개발하기 위해 연구한 결과, 항-TIGIT 항체 및 IL-2 변이체를 포함하는 이중 특이 항체가 면역세포를 효과적으로 조절함을 확인하였다. 이를 기반으로, 상기 이중 특이 항체가 항암효과가 있다는 점을 확인하여 본 발명을 완성하였다.Accordingly, the present inventors conducted research to develop a new combination of dual-specific antibodies that enhance the activity of immune cells, and as a result confirmed that dual-specific antibodies including anti-TIGIT antibodies and IL-2 variants effectively regulate immune cells. did. Based on this, the present invention was completed by confirming that the bispecific antibody has an anticancer effect.
상기 목적 달성을 위해, 본 발명의 일 측면은, TIGIT에 특이적으로 결합하는 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체를 제공한다.To achieve the above object, one aspect of the present invention is an antibody or fragment thereof that specifically binds to TIGIT; and IL-2 protein.
본 발명이 다른 측면은, 상기 이중 특이 항체를 암호화하는 폴리뉴클레오티드, 상기 폴리뉴클레오티드를 포함하는 발현 벡터 및 상기 발현 벡터가 도입된 형질전환 세포를 제공한다.Another aspect of the present invention provides a polynucleotide encoding the bispecific antibody, an expression vector containing the polynucleotide, and a transformed cell into which the expression vector has been introduced.
본 발명의 또 다른 측면은, 상기 형질전환 세포를 배양하는 단계 및 이중 특이 항체를 수득하는 단계를 포함하는 이중 특이 항체의 제조 방법을 제공한다.Another aspect of the present invention provides a method for producing a bispecific antibody, comprising culturing the transformed cells and obtaining a bispecific antibody.
본 발명의 또 다른 측면은, 상기 이중 특이 항체를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물을 제공한다.Another aspect of the present invention provides a pharmaceutical composition for preventing or treating cancer comprising the bispecific antibody as an active ingredient.
본 발명의 또 다른 측면은 암의 예방 또는 치료용 약제를 제조하기 위한 상기 이중 특이 항체의 용도를 제공한다.Another aspect of the present invention provides the use of the bispecific antibody for preparing a medicament for preventing or treating cancer.
본 발명의 또 다른 측면은 암을 예방 또는 치료하기 위한 상기 이중 특이 항체의 용도를 제공한다.Another aspect of the invention provides the use of the bispecific antibody for preventing or treating cancer.
본 발명의 또 다른 측면은 상기 이중 특이 항체를 개체에 투여하는 단계를 포함하는 암을 예방 또는 치료하는 방법을 제공한다. Another aspect of the present invention provides a method for preventing or treating cancer comprising administering the bispecific antibody to a subject.
본 발명에 따른 항-TIGIT 항체 및 IL-2 변이체를 포함하는 이중 특이 항체는 TIGIT과 관련된 기작을 조절할 수 있고, IL-2와 동일 또는 유사한 기능을 할 수 있다. 즉, 상기 이중 특이 항체는 TIGIT과 CD155의 결합을 조절할 수 있을 뿐 아니라, 면역세포를 활성화시킬 수 있다. 또한, 마우스 종양 모델에서도 종양의 성장을 유의적으로 억제하였다. 따라서, 상기 이중 특이 항체는 항암제로 활용이 가능하다.Bispecific antibodies, including anti-TIGIT antibodies and IL-2 variants according to the present invention, can modulate TIGIT-related mechanisms and have the same or similar functions as IL-2. In other words, the bispecific antibody can not only regulate the binding of TIGIT and CD155, but also activate immune cells. Additionally, tumor growth was significantly inhibited in mouse tumor models. Therefore, the bispecific antibody can be used as an anticancer agent.
도 1은 일 실시예인 GI-106 이중 특이 항체의 구조를 도식화한 도면이다.Figure 1 is a diagram schematically illustrating the structure of the GI-106 bispecific antibody, which is an example.
도 2는 정제한 이중 특이 항체(GI-106)를 SDS-PAGE로 확인한 결과를 나타낸 도면이다. Figure 2 is a diagram showing the results of confirming the purified bispecific antibody (GI-106) by SDS-PAGE.
도 3은 일 실시예인 GI-106B7NH06Kv3 이중 특이 항체의 구조를 도식화한 도면이다.Figure 3 is a diagram schematically illustrating the structure of the GI-106B7NH06Kv3 bispecific antibody, which is an example.
도 4는 정제한 이중 특이 항체(GI-106B7NH06Kv3)를 SDS-PAGE로 확인한 결과를 나타낸 도면이다. Figure 4 is a diagram showing the results of confirming the purified bispecific antibody (GI-106B7NH06Kv3) by SDS-PAGE.
도 5는 GI-106의 항-hTIGIT 항체 활성을 측정하기 위한 TIGIT/CD155 저해 어세이(Blockade Bioassay)의 작용 기전을 도식화한 도면이다. Figure 5 is a diagram schematically illustrating the mechanism of action of the TIGIT/CD155 inhibition assay (Blockade Bioassay) for measuring the anti-hTIGIT antibody activity of GI-106.
도 6은 TIGIT/CD155 저해 어세이에서 GI-106과 항-hTIGIT 항체의 농도 별 TIGIT/CD155 결합 저해 정도를 나타낸 그래프이다. Figure 6 is a graph showing the degree of inhibition of TIGIT/CD155 binding by concentration of GI-106 and anti-hTIGIT antibody in the TIGIT/CD155 inhibition assay.
도 7은 GI-106 또는 프로류킨(Proleukin®)을 HEK-BlueTM IL-2 리포터 세포에 처리하고 JAK-STAT 신호 전달 경로 활성을 측정한 결과를 나타낸 그래프이다.Figure 7 is a graph showing the results of treating HEK-Blue TM IL-2 reporter cells with GI-106 or Proleukin® and measuring JAK-STAT signaling pathway activity.
도 8은 인간 유래 유방암 세포(MDA-MB-231) 식립 마우스에 PBS 또는 GI-106을 투여한 후, 각 군의 종양 부피를 측정한 결과를 나타낸 그래프이다.Figure 8 is a graph showing the results of measuring the tumor volume of each group after administering PBS or GI-106 to mice implanted with human-derived breast cancer cells (MDA-MB-231).
도 9는 인간 유래 유방암 세포(MDA-MB-231) 식립 마우스에 PBS 또는 GI-106을 투여한 후, 각 군의 개체별 종양 부피를 나타낸 그래프이다. Figure 9 is a graph showing the tumor volume of each individual in each group after administering PBS or GI-106 to mice implanted with human-derived breast cancer cells (MDA-MB-231).
도 10은 인간 유래 유방암 세포(MDA-MB-231) 식립 마우스에 PBS 투여한 후, 개체별 종양 부피를 나타낸 그래프이다. Figure 10 is a graph showing the tumor volume for each individual after administration of PBS to mice implanted with human-derived breast cancer cells (MDA-MB-231).
도 11은 인간 유래 유방암 세포(MDA-MB-231) 식립 마우스에 GI-106을 투여한 후, 개체별 종양 부피를 나타낸 그래프이다.Figure 11 is a graph showing the tumor volume for each individual after administering GI-106 to mice implanted with human-derived breast cancer cells (MDA-MB-231).
도 12는 인간 유래 유방암 세포(MDA-MB-231) 식립 마우스에 PBS 또는 GI-106을 투여한 후, 각 군의 마우스의 종양 성장 억제율(TGI, tumor growth inhibition)을 나타낸 그래프이다. Figure 12 is a graph showing the tumor growth inhibition (TGI) of each group of mice after administering PBS or GI-106 to mice implanted with human-derived breast cancer cells (MDA-MB-231).
도 13은 마우스 유래 유방암 세포(EMT-6) 식립 마우스에 hIgG4 또는 GI-106 B7NH06Kv3을 투여한 후, 각 군의 종양 부피를 측정한 결과를 나타낸 그래프이다.Figure 13 is a graph showing the results of measuring the tumor volume of each group after administering hIgG4 or GI-106 B7NH06Kv3 to mice implanted with mouse-derived breast cancer cells (EMT-6).
도 14는 마우스 유래 유방암 세포(EMT-6) 식립 마우스에 hIgG4 또는 GI-106 B7NH06Kv3을 투여한 후, 각 군의 마우스의 종양 성장 억제율을 나타낸 그래프이다. Figure 14 is a graph showing the tumor growth inhibition rate of each group of mice after administering hIgG4 or GI-106 B7NH06Kv3 to mice implanted with mouse-derived breast cancer cells (EMT-6).
도 15는 마우스 유래 대장암 세포(MC38) 식립 마우스에 hIgG4, GI-106B7NH06Kv3 또는 GI-106B7NH06K-CN을 단독투여 하거나, GI-106B7NH06K-CN 및 Fc-IL-2v3을 병용투여한 후, 각 군의 종양 부피를 측정한 결과를 나타낸 그래프이다.Figure 15 shows the results of each group after administering hIgG4, GI-106B7NH06Kv3, or GI-106B7NH06K-CN alone or in combination with GI-106B7NH06K-CN and Fc-IL-2v3 to mice implanted with mouse-derived colon cancer cells (MC38). This is a graph showing the results of measuring tumor volume.
도 16은 마우스 유래 대장암 세포(MC38) 식립 마우스에서 hIgG4, GI-106B7NH06Kv3 또는 GI-106B7NH06K-CN을 단독투여 하거나, GI-106B7NH06K-CN 및 Fc-IL-2v3을 병용투여한 후, 각 군의 종양 성장 억제율을 나타낸 그래프이다.Figure 16 shows the results of each group after single administration of hIgG4, GI-106B7NH06Kv3, or GI-106B7NH06K-CN or combined administration of GI-106B7NH06K-CN and Fc-IL-2v3 in mice implanted with mouse-derived colon cancer cells (MC38). This is a graph showing the tumor growth inhibition rate.
도 17은 마우스 유래 대장암 세포(MC38) 식립 마우스에서 hIgG4, GI-106B7NH06Kv3 또는 GI-106B7NH06K-CN을 단독투여 하거나, GI-106B7NH06K-CN 및 Fc-IL-2v3를 병용투여한 후, 각 군의 25일차까지의 생존율을 나타낸 그래프이다.Figure 17 shows the results of each group after single administration of hIgG4, GI-106B7NH06Kv3, or GI-106B7NH06K-CN or combined administration of GI-106B7NH06K-CN and Fc-IL-2v3 in mice implanted with mouse-derived colon cancer cells (MC38). This is a graph showing the survival rate until the 25th day.
항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체anti-TIGIT antibody or fragment thereof; and a bispecific antibody containing the IL-2 protein.
본 발명의 일 측면은, TIGIT에 특이적으로 결합하는 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체를 제공한다.One aspect of the present invention is an antibody or fragment thereof that specifically binds to TIGIT; and IL-2 protein.
본 명세서에서 사용하는 용어, "항체"는 특정 항원과 면역학적으로 반응하는 면역글로불린 분자로, 항원을 특이적으로 인식하는 단백질 분자를 의미한다. 면역글로불린의 중쇄 및 경쇄는 각각 불변 영역(constant region) 및 가변 영역(variable region)을 포함할 수 있다. 면역글로불린의 경쇄 및 중쇄 가변 영역은, 상보성 결정 영역(complementarity determining region, CDR)이라 불리는 3개의 다변가능한 영역 및 4개의 구조 영역(framework region, FR)을 포함한다. 상기 CDR은 주로 항원의 에피토프에 결합하는 항원 결합 부위이다.As used herein, the term “antibody” refers to an immunoglobulin molecule that reacts immunologically with a specific antigen, and refers to a protein molecule that specifically recognizes the antigen. The heavy and light chains of immunoglobulins may each include a constant region and a variable region. The light and heavy chain variable regions of immunoglobulins contain three variable regions called complementarity determining regions (CDRs) and four framework regions (FRs). The CDR is an antigen binding site that mainly binds to the epitope of the antigen.
본 명세서에서 사용하는 용어, "이중 특이 항체"는 적어도 하나 이상의 표적 또는 항원에 결합하는 물질을 의미한다. 본 발명에서 상기 이중 특이 항체는 TIGIT에 특이적으로 결합하는 항원 결합 부위 및 IL-2 단백질 또는 이의 변이체를 포함할 수 있다. As used herein, the term “dual specific antibody” refers to a substance that binds to at least one target or antigen. In the present invention, the bispecific antibody may include an antigen binding site that specifically binds to TIGIT and an IL-2 protein or a variant thereof.
본 발명의 명세서에서 사용한 용어, "TIGIT"은 WUCAM 및 Vstm2로도 알려져 있다. TIGIT은 일부 T 세포 및 자연살해세포(NK cell)에 존재하는 면역 수용체로, 수지상세포(dendritic cell), 대식세포(macrophage) 등에 존재하는 CD155와 결합하여 생체 내 T 세포 및 자연살해세포의 활성을 억제하여 면역 반응을 감소시킨다. The term “TIGIT” as used herein is also known as WUCAM and Vstm2. TIGIT is an immune receptor present on some T cells and natural killer cells (NK cells), and binds to CD155 present on dendritic cells and macrophages to inhibit the activity of T cells and natural killer cells in vivo. Inhibits and reduces immune response.
상기 TIGIT에 특이적으로 결합하는 항체 또는 이의 단편은 TIGIT에 특이적으로 항원-항체 결합할 수 있는 분자를 총칭할 수 있다. 상기 TIGIT에 특이적으로 결합하는 항체 또는 이의 단편은 TIGIT에 특이적으로 결합할 수 있는 항원 결합 부위를 포함하는 것이면, 어떠한 형태도 포함할 수 있다. 상기 항체 또는 이의 단편은 결합에 직접적으로 수반되지 않는 다른 아미노산 또는 항원 결합 부위의 아미노산 잔기에 의해 효과가 차단되는 아미노산을 포함할 수 있다.The antibody or fragment thereof that specifically binds to TIGIT may collectively refer to molecules capable of antigen-antibody binding specifically to TIGIT. The antibody or fragment thereof that specifically binds to TIGIT may have any form as long as it contains an antigen-binding site capable of specifically binding to TIGIT. The antibody or fragment thereof may contain other amino acids that are not directly involved in binding or amino acids whose effect is blocked by amino acid residues in the antigen binding site.
상기 TIGIT에 특이적으로 결합하는 항체 또는 이의 단편은 서열번호 9의 아미노산 서열을 포함하는 HCDR1, 서열번호 10의 아미노산 서열을 포함하는 HCDR2 및 서열번호 11의 아미노산 서열을 포함하는 HCDR3으로 이루어진 중쇄 가변 영역 및 서열번호 12의 아미노산 서열을 포함하는 LCDR1, 서열번호 13의 아미노산 서열(GVK)을 포함하는 LCDR2 및 서열번호 14의 아미노산 서열을 포함하는 LCDR3으로 이루어진 경쇄 가변 영역을 포함할 수 있다. The antibody or fragment thereof that specifically binds to TIGIT is a heavy chain variable region consisting of HCDR1 containing the amino acid sequence of SEQ ID NO: 9, HCDR2 containing the amino acid sequence of SEQ ID NO: 10, and HCDR3 containing the amino acid sequence of SEQ ID NO: 11 and a light chain variable region consisting of LCDR1 including the amino acid sequence of SEQ ID NO: 12, LCDR2 including the amino acid sequence (GVK) of SEQ ID NO: 13, and LCDR3 including the amino acid sequence of SEQ ID NO: 14.
일 실시예에서 상기 TIGIT에 특이적으로 결합하는 항체 또는 이의 단편은 서열번호 2의 아미노산 서열을 포함하는 중쇄 가변 영역 및 서열번호 7의 아미노산 서열을 포함하는 경쇄 가변 영역으로 이루어진 것일 수 있다. 또한, 본 발명의 일 실시예에서 항-TIGIT 항체 또는 이의 단편은 서열번호 3의 아미노산 서열을 포함하는 중쇄 불변 영역1(CH1) 및 서열번호 8 또는 서열번호 24의 아미노산 서열을 포함하는 경쇄 불변 영역(CL)을 포함할 수 있다.In one embodiment, the antibody or fragment thereof that specifically binds to TIGIT may be composed of a heavy chain variable region including the amino acid sequence of SEQ ID NO: 2 and a light chain variable region including the amino acid sequence of SEQ ID NO: 7. In addition, in one embodiment of the present invention, the anti-TIGIT antibody or fragment thereof includes a heavy chain constant region 1 (CH1) comprising the amino acid sequence of SEQ ID NO: 3 and a light chain constant region comprising the amino acid sequence of SEQ ID NO: 8 or SEQ ID NO: 24. (CL) may be included.
본 명세서에서 사용하는 용어, "IL-2" 또는 "인터루킨-2"는 달리 언급되지 않는 한, 포유동물, 예를 들어, 영장류(예, 인간) 및 설치류(예, 마우스 및 래트)를 포함하여 임의의 척추동물 공급원으로부터 수득한 임의의 야생형 IL-2를 의미한다. 상기 IL-2는 동물 세포에서 수득된 것일 수도 있으나, IL-2를 생산할 수 있는 재조합 세포로부터 수득된 것도 포함한다. 또한, 상기 IL-2는 야생형 IL-2 또는 이의 변이체일 수 있다. As used herein, the terms “IL-2” or “interleukin-2” include mammals, such as primates (e.g., humans) and rodents (e.g., mice and rats), unless otherwise noted. refers to any wild type IL-2 obtained from any vertebrate source. The IL-2 may be obtained from animal cells, but also includes those obtained from recombinant cells capable of producing IL-2. Additionally, the IL-2 may be wild-type IL-2 or a variant thereof.
본 명세서에서는 IL-2 혹은 이의 변이체를 총칭하여 "IL-2 단백질", "IL-2 폴리펩타이드" 또는 "IL-2"의 용어와 혼용될 수 있다. IL-2, IL-2 단백질, IL-2 폴리펩타이드 및 IL-2 변이체는 예를 들어 IL-2 수용체(receptor)에 특이적으로 결합한다. 상기 특이적인 결합은 당업자에게 알려진 방법을 통해 확인할 수 있다.In the present specification, IL-2 or its variants may be collectively used interchangeably with the terms “IL-2 protein,” “IL-2 polypeptide,” or “IL-2.” IL-2, IL-2 protein, IL-2 polypeptide and IL-2 variants, for example, bind specifically to the IL-2 receptor. The specific binding can be confirmed through methods known to those skilled in the art.
상기 IL-2는 성숙된 형태일 수 있다. 구체적으로, 상기 성숙된 IL-2는 신호서열을 포함하지 않는 것일 수 있으며, 야생형 IL-2의 N 말단 또는 C 말단의 일부가 결실된(truncated) 단편을 포함할 수 있다. 이때, 상기 IL-2는 서열번호 17의 아미노산 서열을 가질 수 있다. The IL-2 may be in a mature form. Specifically, the mature IL-2 may not contain a signal sequence and may contain a truncated fragment of the N-terminus or C-terminus of wild-type IL-2. At this time, the IL-2 may have the amino acid sequence of SEQ ID NO: 17.
본 명세서에서 사용하는 용어, "IL-2 변이체"는 전장(full-length) IL-2 또는 IL-2의 단편의 아미노산 일부가 치환된 형태를 의미한다. 즉, IL-2 변이체는 야생형 IL-2 또는 이의 단편과 다른 아미노산 서열을 가질 수 있다. 그러나, 상기 IL-2 변이체는 야생형 IL-2와 동등하거나 유사한 활성을 가질 수 있다. 여기에서, "IL-2 활성"은 예를 들어 IL-2 수용체에 특이적으로 결합하는 것을 의미할 수 있으며, 이 특이적 결합은 당업자에게 알려진 방법을 통해 측정할 수 있다.As used herein, the term “IL-2 variant” refers to a form in which some amino acids of the full-length IL-2 or fragment of IL-2 are substituted. That is, the IL-2 variant may have an amino acid sequence that differs from wild-type IL-2 or a fragment thereof. However, the IL-2 variant may have equivalent or similar activity to wild-type IL-2. Here, “IL-2 activity” may mean, for example, specific binding to the IL-2 receptor, and this specific binding can be measured through methods known to those skilled in the art.
구체적으로, 상기 IL-2 변이체는 야생형 IL-2의 아미노산 일부가 치환된 것일 수 있다. 아미노산 치환에 의한 IL-2 변이체의 일 구체예로는 서열번호 17의 아미노산 서열에서 38번째, 42번째, 45번째, 61번째 및 72번째 아미노산 중 적어도 하나가 치환된 것일 수 있다. 일 구체예에 따르면, IL-2 활성이 유지되는 한, 세 개의 아미노산이 치환될 수 있다. 상기 IL-2 변이체는 서열번호 17의 아미노산 서열에서 38번째, 42번째 및 61번째 위치의 아미노산이 치환된 것일 수 있다.Specifically, the IL-2 variant may be one in which some amino acids of wild-type IL-2 have been substituted. One specific example of an IL-2 variant resulting from amino acid substitution may be one in which at least one of the 38th, 42nd, 45th, 61st, and 72nd amino acids in the amino acid sequence of SEQ ID NO: 17 is substituted. According to one embodiment, three amino acids may be substituted as long as IL-2 activity is maintained. The IL-2 variant may be one in which amino acids at the 38th, 42nd, and 61st positions in the amino acid sequence of SEQ ID NO: 17 are substituted.
구체적으로, 상기 IL-2 변이체는 서열번호 17의 아미노산 서열에서 R38A, F42A, Y45A, E61R 및 L72G로 구성된 군으로부터 선택되는 적어도 어느 하나의 치환이 일어난 것일 수 있다. 상기 IL-2 변이체는 서열번호 17의 아미노산 서열에서 R38A, F42A 및 E61R 치환이 일어난 것일 수 있다. 일 실시예에서, 상기 IL-2 변이체는 서열번호 6의 아미노산 서열을 가질 수 있다.Specifically, the IL-2 variant may have at least one substitution selected from the group consisting of R38A, F42A, Y45A, E61R, and L72G in the amino acid sequence of SEQ ID NO: 17. The IL-2 variant may have R38A, F42A, and E61R substitutions in the amino acid sequence of SEQ ID NO: 17. In one embodiment, the IL-2 variant may have the amino acid sequence of SEQ ID NO:6.
상기 TIGIT에 특이적으로 결합하는 항체 또는 이의 단편; 및 IL-2 단백질 또는 이의 변이체는 링커 혹은 캐리어(carrier)에 의해 결합된 것일 수 있다. 본 명세서에서 링커와 캐리어는 호환적으로 사용되기도 한다. 링커의 일 구체예로는 1 내지 50개의 아미노산, 알부민 또는 이의 단편, Fc(영역) 등을 포함할 수 있다. 상기 Fc는 면역글로불린의 Fc 도메인일 수 있다. An antibody or fragment thereof that specifically binds to the TIGIT; And the IL-2 protein or its variant may be bound by a linker or carrier. In this specification, linker and carrier are also used interchangeably. One specific example of the linker may include 1 to 50 amino acids, albumin or a fragment thereof, Fc (region), etc. The Fc may be the Fc domain of an immunoglobulin.
이때, 상기 면역글로불린의 Fc 영역은 면역글로불린의 중쇄 불변 영역 2(CH2) 및 중쇄 불변 영역 3(CH3)을 포함하며, 면역글로불린의 중쇄 및 경쇄의 가변 영역 및 경쇄 불변 영역(CL)은 포함하지 않는 단백질을 의미한다. 상기 면역글로불린 Fc 영역은 IgG, IgA, IgE, IgD 또는 IgM 유래 일 수 있다. 구체적으로, 상기 면역글로불린 Fc 영역은 IgG의 하위 부류인 IgG1, IgG2, IgG3 또는 IgG4일 수 있으며, 바람직하게는 IgG4 유래일 수 있다. At this time, the Fc region of the immunoglobulin includes the heavy chain constant region 2 (CH2) and the heavy chain constant region 3 (CH3) of the immunoglobulin, but does not include the variable regions of the heavy and light chains and the light chain constant region (CL) of the immunoglobulin. refers to proteins that do not The immunoglobulin Fc region may be derived from IgG, IgA, IgE, IgD or IgM. Specifically, the immunoglobulin Fc region may be IgG1, IgG2, IgG3, or IgG4, which are subclasses of IgG, and preferably may be derived from IgG4.
또한, 상기 면역글로불린의 Fc 도메인은 야생형 Fc 도메인 뿐만 아니라, Fc 도메인 변이체일 수 있다. 또한, 본 명세서에서 사용하는 용어 "Fc 도메인 변이체"는 야생형 Fc 도메인의 당쇄 형태(glycosylation pattern)와 다르거나, 야생형 Fc 도메인에 비해 증가된 당쇄, 야생형 Fc 도메인에 비해 감소한 당쇄, 또는 당쇄가 제거(deglycosylated)된 형태일 수 있다. 또한, 무당쇄(aglycosylated) Fc 도메인도 포함된다. Fc 도메인 혹은 변이체는 배양조건 혹은 호스트의 유전자 조작을 통해 조정된 숫자의 시알산(sialic acid), 퓨코실화(fucosylation), 당화(glycosylation)를 갖도록 한 것일 수 있다.Additionally, the Fc domain of the immunoglobulin may be not only a wild-type Fc domain, but also an Fc domain variant. In addition, the term "Fc domain variant" as used herein refers to a glycosylation pattern that is different from that of the wild-type Fc domain, has an increased sugar chain compared to the wild-type Fc domain, has a decreased sugar chain compared to the wild-type Fc domain, or has a sugar chain removed ( It may be in a deglycosylated form. Additionally, an aglycosylated Fc domain is also included. The Fc domain or variant may have an adjusted number of sialic acids, fucosylation, and glycosylation through culture conditions or genetic manipulation of the host.
또한, 화학적 방법, 효소적 방법 및 미생물을 사용한 유전공학적 엔지니어링 방법 등과 같이 통상적인 방법으로 면역글로불린의 Fc 도메인의 당쇄를 변형시킬 수 있다. 또한, 상기 Fc 도메인 변이체는 면역글로불린 IgG, IgA, IgE, IgD 또는 IgM의 Fc 영역이 혼합된 형태일 수 있다. 또한, 상기 Fc 도메인 변이체는 상기 Fc 도메인의 일부 아미노산이 다른 아미노산으로 치환된 형태일 수 있다.Additionally, the sugar chain of the Fc domain of an immunoglobulin can be modified by conventional methods such as chemical methods, enzymatic methods, and genetic engineering methods using microorganisms. Additionally, the Fc domain variant may be a mixture of the Fc regions of immunoglobulins IgG, IgA, IgE, IgD, or IgM. Additionally, the Fc domain variant may be a form in which some amino acids of the Fc domain are replaced with other amino acids.
일 실시예에서 상기 Fc 영역은 서열번호 4 또는 서열번호 23의 아미노산 서열을 가질 수 있다.In one embodiment, the Fc region may have the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 23.
본 발명의 이중 특이 항체는 항-TIGIT 항체의 경쇄 가변 영역(VL) 및 경쇄 불변 영역(CL)을 포함하는 융합단백질과 항-TIGIT 항체의 중쇄 가변 영역(VH), 중쇄 불변 영역 1(CH1), Fc 도메인 및 IL-2 단백질을 포함하는 융합단백질을 포함할 수 있다. 또한, 본 발명이 이중 특이 항체는 항-TIGIT 항체의 경쇄 가변 영역, 경쇄 불변 영역 및 IL-2 단백질을 포함하는 융합단백질과 항-TIGIT 항체의 중쇄 가변 영역, 중쇄 불변 영역 1(CH1) 및 Fc 도메인을 포함하는 융합단백질을 포함할 수 있다. The bispecific antibody of the present invention is a fusion protein comprising the light chain variable region (VL) and light chain constant region (CL) of an anti-TIGIT antibody, and the heavy chain variable region (VH) and heavy chain constant region 1 (CH1) of an anti-TIGIT antibody. , may include a fusion protein containing an Fc domain and an IL-2 protein. In addition, the bispecific antibody of the present invention is a fusion protein comprising the light chain variable region, light chain constant region, and IL-2 protein of an anti-TIGIT antibody, and the heavy chain variable region, heavy chain constant region 1 (CH1), and Fc of the anti-TIGIT antibody. It may contain a fusion protein containing a domain.
구체적으로, 상기 항-TIGIT 항체 및 IL-2 단백질 또는 이의 변이체를 포함하는 이중 특이 항체는 하기 구조식 (I) 및 (II); 또는 (III) 및 (IV)를 포함하는 것일 수 있다.Specifically, the bispecific antibody comprising the anti-TIGIT antibody and IL-2 protein or a variant thereof has the following structural formulas (I) and (II); Or it may include (III) and (IV).
N'-X-[링커(1)]o-Fc 영역 단편 또는 이의 변이체-[링커(2)]p-Y-C' (I) 및N'-X-[Linker (1)]o-Fc region fragment or variant thereof-[Linker (2)]p-Y-C' (I) and
N'-X'-C' (II); N'-X'-C' (II);
또는or
N'-X-[링커(1)]o-Fc 영역 단편 또는 이의 변이체-C' (III) 및N'-X-[Linker (1)]o-Fc region fragment or variant thereof-C' (III) and
N'-X'-[링커(3)]q-Y-C' (IV)N'-X'-[Linker (3)]q-Y-C' (IV)
이때, 상기 구조식 (I), (II), (III) 및 (IV)에 있어서,At this time, in the structural formulas (I), (II), (III) and (IV),
상기 N'은 N 말단이고,Wherein N' is the N terminus,
상기 C'는 C 말단이며,Wherein C' is the C terminus,
상기 X는 항-TIGIT 항체의 중쇄의 항원 결합 부위로 가변 영역(VH) 및 CH1 영역을 포함하며,The X is the antigen binding site of the heavy chain of the anti-TIGIT antibody and includes a variable region (VH) and a CH1 region,
상기 X'는 항-TIGIT 항체의 경쇄 항원 결합 부위로 가변 영역(VL) 및 불변 영역(CL)을 포함하고, The X' is the light chain antigen binding site of the anti-TIGIT antibody and includes a variable region (VL) and a constant region (CL),
상기 Y는 IL-2 단백질 또는 이의 변이체이며,Y is IL-2 protein or a variant thereof,
상기 링커(1), 링커(2) 및 링커(3)은 각각 독립적으로 펩타이드 링커이고,The linker (1), linker (2), and linker (3) are each independently peptide linkers,
상기 o, p 및 q는 각각 독립적으로, O 또는 1이다.The o, p and q are each independently O or 1.
이때, 항-TIGIT 항체, 항원 결합 부위, 가변 영역, 불변 영역; IL-2 단백질, IL-2 변이체, Fc 영역(혹은 Fc 도메인)은 상술한 바와 같다. 또한, 상기 이중 특이 항체에서 IL-2 단백질 또는 이의 변이체는 Fc 영역의 C 말단 또는 항-TIGIT 항체의 경쇄 불변 영역의 C 말단에 결합될 수 있다. 이때, 상기 IL-2 단백질 또는 이의 변이체와 Fc 영역 또는 경쇄 불변 영역은 펩타이드 링커를 통해 결합할 수 있다. At this time, anti-TIGIT antibody, antigen binding site, variable region, constant region; IL-2 protein, IL-2 variant, and Fc region (or Fc domain) are as described above. Additionally, in the bispecific antibody, the IL-2 protein or a variant thereof may be bound to the C terminus of the Fc region or the C terminus of the light chain constant region of the anti-TIGIT antibody. At this time, the IL-2 protein or its variant and the Fc region or light chain constant region may be combined through a peptide linker.
상기 펩타이드 링커(1)은 1 내지 50개의 연속된 아미노산, 또는 3 내지 30개의 연속된 아미노산, 또는 5 내지 15개의 아미노산으로 이루어질 수 있다. 일 구체예로 상기 펩타이드 링커(1)은 12개의 아미노산으로 이루어질 수 있다. 또한, 펩타이드 링커(1)은 적어도 하나의 시스테인을 포함할 수 있다. 구체적으로, 하나, 두 개 또는 세 개의 시스테인을 포함할 수 있다. 또한, 상기 펩타이드 링커(1)은 면역글로불린의 힌지에서 유래된 것일 수 있다. 예를 들어, 상기 힌지는 다양한 IgG 하위 유형 항체의 힌지 영역으로부터 선택될 수 있다. 또한, 상기 힌지는 면역글로불린 유래의 힌지 영역의 일부 아미노산이 다른 아미노산으로 치환된 형태이거나, 일부 아미노산 서열이 추가된 서열일 수 있다. 일 구체예에서는, 상기 펩타이드 링커(1)이 서열번호 18의 아미노산 서열로 이루어진 펩타이드 링커일 수 있다. The peptide linker (1) may be composed of 1 to 50 consecutive amino acids, or 3 to 30 consecutive amino acids, or 5 to 15 amino acids. In one embodiment, the peptide linker (1) may be composed of 12 amino acids. Additionally, the peptide linker 1 may include at least one cysteine. Specifically, it may contain one, two or three cysteines. Additionally, the peptide linker (1) may be derived from the hinge of immunoglobulin. For example, the hinge can be selected from the hinge regions of various IgG subtype antibodies. Additionally, the hinge may be a form in which some amino acids in the hinge region derived from immunoglobulin are replaced with other amino acids, or may be a sequence in which some amino acid sequences are added. In one embodiment, the peptide linker (1) may be a peptide linker consisting of the amino acid sequence of SEQ ID NO: 18.
상기 펩타이드 링커(2) 또는 링커(3)은 1 내지 30개의 연속된 아미노산, 또는 2 내지 20개의 연속된 아미노산, 또는 2 내지 10개의 아미노산으로 이루어질 수 있다. 일 구체예로 상기 펩타이드 링커(2) 또는 링커(3)은 (G4S)n(이때, n은 1 내지 10의 정수) 일 수 있다. 이때, (G4S)n에서 n은 1, 2, 3, 4, 5, 6, 7, 8, 9 또는 10일 수 있다. 일 실시예로, 상기 펩타이드 링커(2) 또는 링커(3)은 서열번호 5의 아미노산 서열로 이루어진 펩타이드 링커일 수 있다. The peptide linker (2) or linker (3) may be composed of 1 to 30 consecutive amino acids, or 2 to 20 consecutive amino acids, or 2 to 10 amino acids. In one embodiment, the peptide linker (2) or linker (3) may be (G4S)n (where n is an integer of 1 to 10). At this time, n in (G4S)n may be 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10. In one embodiment, the peptide linker (2) or linker (3) may be a peptide linker consisting of the amino acid sequence of SEQ ID NO: 5.
상기 이중 특이 항체를 구성하는 영역의 각각의 아미노산 서열은 하기 표 1 및 표 2에 나타낸 바와 같다. 구체적으로, 표 1은 항-hTIGIT HC-hIgG4 Fc-hIL2v3 및 항-hTIGIT LC(lambda)의 아미노산 서열을 기술한 것이다. 또한, 표 2는 항-hTIGIT HC-hIgG4FcM1 및 항-hTIGIT LC(kappa)-hIL2v3의 아미노산 서열을 기술한 것이다.The amino acid sequences of each region constituting the bispecific antibody are shown in Tables 1 and 2 below. Specifically, Table 1 describes the amino acid sequences of anti-hTIGIT HC-hIgG4 Fc-hIL2v3 and anti-hTIGIT LC (lambda). Additionally, Table 2 describes the amino acid sequences of anti-hTIGIT HC-hIgG4FcM1 and anti-hTIGIT LC(kappa)-hIL2v3.
구분division 아미노산 서열amino acid sequence 서열번호sequence number
항-hTIGIT HC-hIgG4Fc-
hIL2v3
Anti-hTIGIT HC-hIgG4Fc-
hIL2v3
signal peptide(mIgG)signal peptide (mIgG) MEWSWVFLFFLSVTTGVHSMEWSWVFLFFLSVTTGVHS 1One
항-hTIGIT VHAnti-hTIGIT VH QVQLQESGPGLVKPSGTLSLTCAVSGVSIRQGHWWSWVRQPPGKGLEWIGEIYQTGRTNYNPSLKSRVTISVGKSRNHISLKLSSVTAADTAVYYCTTGSGWYPIDYWGQGTLVTVSS QVQLQESGPGLVKPSGTLSLTCAVS GVSIRQGHW WSWVRQPPGKGLEWIGE IYQTGRT NYNPSLKSRVTISVGKSRNHISLKLSSVTAADTAVYYC TTGSGWYPIDY WGQGTLVTVSS 22
항-hTIGIT CH1Anti-hTIGIT CH1 ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRV 33
제1링커 first linker ESKYGPPCPPCPESKYGPPCPPCP 1818
F02(hIgG4 Fc)F02(hIgG4 Fc) APEAAGGPSVFLFPPKPKDQLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVLHEALHNHYTQKSLSLSLG APEAAGGPSVFLFPPKPKDQLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSR WQEGNVFSCSVLHEALHNHYTQKSLLSLSLG 44
제2링커 2nd linker GGGGSGGGGS 55
hIL2v3 hIL2v3 APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTAMLTAKFYMPKKATELKHLQCLERELKPLEEVLNLAQSKNFHLRPRDLISNINVIVLELKGSETTF MCEYADETATIVEFLNRWITFCQSIISTLTAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTAMLTAKFYMPKKATELKHLQCLERELKPLEEVLNLAQSKNFHLRPRDLISNINVIVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLT 66
항-hTIGIT LC(lambda)Anti-hTIGIT LC(lambda) signal peptide(mIgG)signal peptide (mIgG) MEWSWVFLFFLSVTTGVHSMEWSWVFLFFLSVTTGVHS 1One
항-hTIGIT VLAnti-hTIGIT VL SYELTQDPAVSVALGQTVRITCQGDSLRGFYASWYQQKPGQAPLLVLYGVKDRPSGIPDRFSGSRSGTTASLIITGAQAEDEADYYCNSRDSNGAMMFGGGTKLTVLSYELTQDPAVSVALGQTVRITCQGD SLRGFY ASWYQQKPGQAPLLVLY GVK DRPSGIPDRFSGSRSGTTASLIITGAQAEDEADYYC NSRDSNGAMM FGGGTKLTVL 77
항-hTIGIT lambda CLanti-hTIGIT lambda CL QPKANPTVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADGSPVKAGVETTKPSKQSNNKYAASSYLSLTP EQWKSHRSYSCQVTHEGSTVEKTVAPTECSQPKANPTVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADGSPVKAGVETTKPSKQSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS 88
구분division 아미노산 서열amino acid sequence 서열번호sequence number
항-hTIGIT HC-hIgG4FcM1Anti-hTIGIT HC-hIgG4FcM1 signal peptide(mIgG)signal peptide (mIgG) MEWSWVFLFFLSVTTGVHSMEWSWVFLFFLSVTTGVHS 1One
항-hTIGIT VHAnti-hTIGIT VH QVQLQESGPGLVKPSGTLSLTCAVSGVSIRQGHWWSWVRQPPGKGLEWIGEIYQTGRTNYNPSLKSRVTISVGKSRNHISLKLSSVTAADTAVYYCTTGSGWYPIDYWGQGTLVTVSSQVQLQESGPGLVKPSGTLSLTCAVS GVSIRQGH WWSWVRQPPGKGLEWIGE IYQTGRT NYNPSLKSRVTISVGKSRNHISLKLSSVTAADTAVYYC TTGSGWYPIDY WGQGTLVTVSS 22
항-hTIGIT CH1Anti-hTIGIT CH1 ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRV 33
제1링커 first linker ESKYGPPCPPCPESKYGPPCPPCP 1818
F02K(hIgG4 FcM1)F02K(hIgG4 FcM1) APEAAGGPSVFLFPPKPKDQLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVLHEALHNHYTQKSLSLSLGKAPEAAGGPSVFLFPPKPKDQLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSR WQEGNVFSCSVLHEALHNHYTQKSLLSLSLGK 2323
항-hTIGIT LC(kappa)-hIL2v3Anti-hTIGIT LC(kappa)-hIL2v3 signal peptide(mIgG)signal peptide (mIgG) MEWSWVFLFFLSVTTGVHSMEWSWVFLFFLSVTTGVHS 1One
항-hTIGIT VLAnti-hTIGIT VL SYELTQDPAVSVALGQTVRITCQGDSLRGFYASWYQQKPGQAPLLVLYGVKDRPSGIPDRFSGSRSGTTASLIITGAQAEDEADYYCNSRDSNGAMMFGGGTKLTVLSYELTQDPAVSVALGQTVRITCQGD SLRGFY ASWYQQKPGQAPLLVLY GVK DRPSGIPDRFSGSRSGTTASLIITGAQAEDEADYYC NSRDSNGAMM FGGGTKLTVL 77
항-hTIGIT kappa CLAnti-hTIGIT kappa CL RSVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKAD YEKHKVYACEVTHQGLSSPVTKSFNRGECRSVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC 2424
제2링커 2nd linker GGGGSGGGGS 55
hIL2v3 hIL2v3 APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTAMLTAKFYMPKKATELKHLQCLERELKPLEEVLNLAQSKNFHLRPRDLISNINVIVLELKGSETTF MCEYADETATIVEFLNRWITFCQSIISTLTAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTAMLTAKFYMPKKATELKHLQCLERELKPLEEVLNLAQSKNFHLRPRDLISNINVIVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLT 66
이중 특이 항체를 암호화하는 폴리뉴클레오티드Polynucleotides encoding bispecific antibodies
본 발명의 다른 측면은 항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체를 암호화하는 폴리뉴클레오티드를 제공한다. 상기 항-TIGIT 항체, 항-TIGIT 항체의 단편, IL-2 단백질 및 이중 특이 항체는 상술한 바와 동일하다.Another aspect of the invention is an anti-TIGIT antibody or fragment thereof; and a polynucleotide encoding a bispecific antibody comprising the IL-2 protein. The anti-TIGIT antibody, fragment of anti-TIGIT antibody, IL-2 protein and bispecific antibody are the same as described above.
상기 폴리뉴클레오티드는 항-TIGIT 항체의 중쇄 가변 영역을 암호화하는 서열번호 32의 염기서열을 포함할 수 있다. 또한, 상기 폴리뉴클레오티드는 항-TIGIT 항체의 경쇄 가변 영역을 암호화하는 서열번호 33의 염기서열을 포함할 수 있다. 본 발명의 일 실시예에서 이중 특이 항체를 암호화하는 폴리뉴클레오티드는 서열번호 15 및 서열번호 16의 염기서열을 포함할 수 있다. 본 발명의 일 구체예에서 이중 특이 항체를 암호화하는 폴리뉴클레오티드는 서열번호 27 및 서열번호 28의 염기서열을 포함할 수 있다. The polynucleotide may include the base sequence of SEQ ID NO: 32, which encodes the heavy chain variable region of an anti-TIGIT antibody. Additionally, the polynucleotide may include the base sequence of SEQ ID NO: 33, which encodes the light chain variable region of an anti-TIGIT antibody. In one embodiment of the present invention, the polynucleotide encoding the dual-specific antibody may include the base sequences of SEQ ID NO: 15 and SEQ ID NO: 16. In one embodiment of the present invention, the polynucleotide encoding the dual-specific antibody may include the base sequences of SEQ ID NO: 27 and SEQ ID NO: 28.
또한, 상기 폴리뉴클레오티드는 동일한 폴리펩타이드를 암호화한다면, 하나 이상의 염기가 치환, 결실, 삽입 또는 이들의 조합에 의해 변이될 수 있다. 폴리뉴클레오티드 서열을 화학적으로 합성하여 제조하는 경우, 당업계에 널리 공지된 합성법, 예를 들어 문헌(Engels and Uhlmann, Angew Chem IntEd Engl., 37:73-127, 1988)에 기술된 방법을 이용할 수 있으며, 트리에스테르, 포스파이트, 포스포르아미다이트 및 H-포스페이트 방법, PCR 및 기타 오토프라이머 방법, 고체 지지체상의 올리고뉴클레오티드 합성법 등을 들 수 있다.Additionally, if the polynucleotide encodes the same polypeptide, one or more bases may be mutated by substitution, deletion, insertion, or a combination thereof. When preparing a polynucleotide sequence by chemical synthesis, synthesis methods well known in the art, for example, methods described in the literature (Engels and Uhlmann, Angew Chem IntEd Engl., 37:73-127, 1988), can be used. and triester, phosphite, phosphoramidite and H-phosphate methods, PCR and other autoprimer methods, and oligonucleotide synthesis methods on solid supports.
일 구체예에 따르면, 상기 폴리뉴클레오티드는 서열번호 15의 염기서열과 적어도 약 70%, 적어도 약 75%, 적어도 약 80%, 적어도 약 85%, 적어도 약 86%, 적어도 약 87%, 적어도 약 88%, 적어도 약 89%, 적어도 약 90%, 적어도 약 91%, 적어도 약 92%, 적어도 약 93%, 적어도 약 94%, 적어도 약 95%, 적어도 약 96%, 적어도 약 97%, 적어도 약 98%, 적어도 약 99%, 또는 적어도 약 100%의 동일성을 가지는 염기서열을 포함할 수 있다. According to one embodiment, the polynucleotide is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, at least about 88% of the base sequence of SEQ ID NO: 15. %, 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.
일 구체예에 따르면, 상기 폴리뉴클레오티드는 서열번호 16의 염기서열과 적어도 약 70%, 적어도 약 75%, 적어도 약 80%, 적어도 약 85%, 적어도 약 86%, 적어도 약 87%, 적어도 약 88%, 적어도 약 89%, 적어도 약 90%, 적어도 약 91%, 적어도 약 92%, 적어도 약 93%, 적어도 약 94%, 적어도 약 95%, 적어도 약 96%, 적어도 약 97%, 적어도 약 98%, 적어도 약 99%, 또는 적어도 약 100%의 동일성을 가지는 염기서열을 포함할 수 있다. According to one embodiment, the polynucleotide is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, at least about 88% of the base sequence of SEQ ID NO: 16. %, 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.
일 구체예에 따르면, 상기 폴리뉴클레오티드는 서열번호 27의 염기서열과 적어도 약 70%, 적어도 약 75%, 적어도 약 80%, 적어도 약 85%, 적어도 약 86%, 적어도 약 87%, 적어도 약 88%, 적어도 약 89%, 적어도 약 90%, 적어도 약 91%, 적어도 약 92%, 적어도 약 93%, 적어도 약 94%, 적어도 약 95%, 적어도 약 96%, 적어도 약 97%, 적어도 약 98%, 적어도 약 99%, 또는 적어도 약 100%의 동일성을 가지는 염기서열을 포함할 수 있다. According to one embodiment, the polynucleotide has the base sequence of SEQ ID NO: 27 and at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, and 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.
일 구체예에 따르면, 상기 폴리뉴클레오티드는 서열번호 28의 염기서열과 적어도 약 70%, 적어도 약 75%, 적어도 약 80%, 적어도 약 85%, 적어도 약 86%, 적어도 약 87%, 적어도 약 88%, 적어도 약 89%, 적어도 약 90%, 적어도 약 91%, 적어도 약 92%, 적어도 약 93%, 적어도 약 94%, 적어도 약 95%, 적어도 약 96%, 적어도 약 97%, 적어도 약 98%, 적어도 약 99%, 또는 적어도 약 100%의 동일성을 가지는 염기서열을 포함할 수 있다. According to one embodiment, the polynucleotide has the base sequence of SEQ ID NO: 28 and at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, and 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.
상기 폴리뉴클레오티드는 신호서열(signal sequence) 또는 리더 서열(leader sequence)을 추가적으로 포함할 수 있다. 여기에서 사용하는 용어 "신호서열"은 목적 단백질의 분비를 지시하는 신호 펩타이드를 암호화하는 핵산을 의미한다. 상기 신호 펩타이드는 숙주 세포에서 번역된 후에 절단된다. 구체적으로, 본원 발명의 신호서열은 ER(endoplasmic reticulum) 막을 관통하는 단백질의 이동을 개시하는 아미노산 서열을 암호화하는 폴리뉴클레오티드이다. The polynucleotide may additionally include a signal sequence or leader sequence. The term “signal sequence” used herein refers to a nucleic acid encoding a signal peptide that directs secretion of a target protein. The signal peptide is cleaved after translation in the host cell. Specifically, the signal sequence of the present invention is a polynucleotide encoding an amino acid sequence that initiates the movement of a protein across the ER (endoplasmic reticulum) membrane.
신호서열은 당업계에 그 특징이 잘 알려져 있으며, 통상 16 내지 30개의 아미노산 잔기를 포함하나, 그보다 더 많거나 적은 아미노산 잔기를 포함할 수 있다. 통상적인 신호 펩타이드는 기본 N 말단 영역, 중심의 소수성 영역 및 보다 극성인(polar) C 말단 영역의 세 영역으로 구성된다. 중심 소수성 영역은 미성숙 폴리펩타이드가 이동하는 동안 막지질 이중층을 통하여 신호서열을 고정시키는 4 내지 12개의 소수성 잔기를 포함한다.Signal sequences are well known in the art, and typically contain 16 to 30 amino acid residues, but may contain more or fewer 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 while the immature polypeptide moves.
개시 이후에, 신호서열은 흔히 신호 펩티다아제(signal peptidases)로 알려진 세포 효소에 의하여 ER의 루멘(lumen) 내에서 절단된다. 이때, 상기 신호서열은 tPa(tissue Plasminogen Activation), HSV gDs(signal sequence of Herpes simplex virus glycoprotein D), IgG 신호서열 또는 성장 호르몬(growth hormone)의 분비신호서열일 수 있다. 바람직하게, 포유동물 등을 포함하는 고등 진핵세포에서 사용되는 분비 신호서열을 사용할 수 있다. 본 발명에서 유용한 신호서열은 항체 경쇄 신호서열, 예를 들면 항체 14.18(Gillies et al., J. Immunol. Meth 1989. 125:191-202), 항체 중쇄 신호서열, 예를 들면, MOPC141 항체 중쇄 신호서열(Sakano et al., Nature, 1980. 286: 676-683) 및 당업계에 알려진 다른 신호서열(예, Watson et al., Nucleic Acid Research, 1984. 12:5145-5164를 참조)을 포함한다. 일 구체예로 상기 신호서열은 서열번호 1의 아미노산 서열을 포함할 수 있다. After initiation, the signal sequence is cleaved within the lumen of the ER by cellular enzymes commonly known as signal peptidases. At this time, the signal sequence may be tPa (tissue Plasminogen Activation), HSV gDs (signal sequence of Herpes simplex virus glycoprotein D), IgG signal sequence, or growth hormone secretion signal sequence. Preferably, a secretion signal sequence used in higher eukaryotic cells, including mammals, can be used. Signal sequences useful in the present invention include antibody light chain signal sequences, such as antibody 14.18 (Gillies et al., J. Immunol. Meth 1989. 125:191-202), and antibody heavy chain signal sequences, such as MOPC141 antibody heavy chain signal. sequence (Sakano et al., Nature, 1980. 286: 676-683) and other signal sequences known in the art (e.g., see Watson et al., Nucleic Acid Research, 1984. 12:5145-5164). . In one specific example, the signal sequence may include the amino acid sequence of SEQ ID NO: 1.
폴리뉴클레오티드가 적재된 벡터Vector loaded with polynucleotide
본 발명의 또 다른 측면은, 상기 항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체를 암호화하는 폴리뉴클레오티드가 적재된 벡터를 제공한다. 이때, 상기 폴리뉴클레오티드는 항-TIGIT 항체의 중쇄 가변 영역을 암호화하는 서열번호 32 및 항-TIGIT 항체의 경쇄 가변 영역을 암호화하는 서열번호 33의 염기서열을 포함할 수 있다. 구체적으로, 상기 이중 특이 항체는 서열번호 15 및 서열번호 16의 염기서열에 의해 암호화되거나, 서열번호 27 및 서열번호 28의 염기서열에 의해 암호화되는 것일 수 있다.Another aspect of the invention is the anti-TIGIT antibody or fragment thereof; and a vector loaded with a polynucleotide encoding a bispecific antibody containing the IL-2 protein. At this time, the polynucleotide may include the base sequence of SEQ ID NO: 32, which encodes the heavy chain variable region of the anti-TIGIT antibody, and SEQ ID NO: 33, which encodes the light chain variable region of the anti-TIGIT antibody. Specifically, the bispecific antibody may be encoded by the base sequences of SEQ ID NO: 15 and SEQ ID NO: 16, or may be encoded by the base sequences of SEQ ID NO: 27 and SEQ ID NO: 28.
본 명세서에서 사용하는 용어, "벡터"는 숙주 세포에 도입되어 숙주 세포 유전체 내로 재조합 및 삽입될 수 있다. 또는 상기 벡터는 에피좀으로서 자발적으로 복제될 수 있는 뉴클레오티드 서열을 포함하는 핵산 수단으로 이해된다. 상기 벡터는 선형 핵산, 플라스미드, 파지미드, 코스미드, RNA 벡터, 바이러스 벡터, 미니-염색체 및 이의 유사체들을 포함한다. 바이러스 벡터의 예로는 레트로바이러스, 아데노바이러스 및 아데노-관련 바이러스를 포함하나 이에 제한되지 않는다. As used herein, the term “vector” can be introduced into a host cell and recombined and inserted into the host cell genome. Alternatively, the vector is understood as a nucleic acid vehicle containing a nucleotide sequence capable of spontaneous replication as an episome. The vectors include linear nucleic acids, plasmids, phagemids, cosmids, RNA vectors, viral vectors, mini-chromosomes and analogs thereof. Examples of viral vectors include, but are not limited to, retroviruses, adenoviruses, and adeno-associated viruses.
구체적으로, 상기 벡터는 플라스미드 DNA, 파아지 DNA 등이 될 수 있고, 상업적으로 개발된 플라스미드(pUC18, pBAD, pIDTSAMRT-AMP 등), 대장균 유래 플라스미드(pYG601BR322, pBR325, pUC118, pUC119 등), 바실러스 서브틸리스 유래 플라스미드(pUB110, pTP5 등), 효모-유래 플라스미드(YEp13, YEp24, YCp50 등), 파아지 DNA(Charon4A, Charon21A, EMBL3, EMBL4, λgt10, λgt11, λZAP 등), 동물 바이러스 벡터(레트로바이러스(retrovirus), 아데노바이러스(adenovirus), 백시니아 바이러스(vaccinia virus) 등), 곤충 바이러스 벡터(배큘로바이러스(baculovirus) 등) 등이 될 수 있다. 상기 벡터는 숙주 세포에 따라서 단백질의 발현량과 수식 등이 다르게 나타나므로, 목적에 가장 적합한 숙주 세포를 선택하여 사용함이 바람직하다. Specifically, the vector may be plasmid DNA, phage DNA, etc., commercially developed plasmids (pUC18, pBAD, pIDTSAMRT-AMP, etc.), E. coli-derived plasmids (pYG601BR322, pBR325, pUC118, pUC119, etc.), Bacillus subtilis. plasmids (pUB110, pTP5, etc.), yeast-derived plasmids (YEp13, YEp24, YCp50, etc.), phage DNA (Charon4A, Charon21A, EMBL3, EMBL4, λgt10, λgt11, λZAP, etc.), animal virus vectors (retroviruses) ), adenovirus, vaccinia virus, etc.), insect virus vectors (baculovirus, etc.), etc. Since the expression level and modification of the protein of the vector varies depending on the host cell, it is preferable to select and use the host cell most suitable for the purpose.
또한, 상기 플라스미드는 항생제 내성 유전자와 같은 선별 마커를 포함할 수 있고, 플라스미드를 유지하는 숙주 세포는 선택적인 조건하에서 배양될 수 있다.Additionally, the plasmid may contain a selection marker, such as an antibiotic resistance gene, and host cells maintaining the plasmid may be cultured under selective conditions.
본 명세서에서 사용하는 용어, 목적 단백질의 "유전자 발현" 또는 "발현"은 DNA 서열의 전사, mRNA 전사체의 번역 및 이중 특이 항체 생산물 또는 이의 단편의 분비를 의미하는 것으로 이해된다. 유용한 발현 벡터는 RcCMV(Invitrogen, Carlsbad) 또는 이의 변이체일 수 있다. 상기 발현 벡터는 포유류 세포에서 목적 유전자의 연속적인 전사를 촉진하기 위한 인간 CMV(cytomegalovirus) 프로모터 및 전사 후 RNA의 안정상태 수준을 높이기 위한 우태 성장 인자(bovine growth hormone) 폴리아데닐레이션 신호서열을 포함할 수 있다.As used herein, the term “gene expression” or “expression” of a protein of interest is understood to mean transcription of a DNA sequence, translation of an mRNA transcript, and secretion of a bispecific antibody product or fragment thereof. A useful expression vector may be RcCMV (Invitrogen, Carlsbad) or variants thereof. The expression vector may include a human CMV (cytomegalovirus) promoter to promote continuous transcription of the target gene in mammalian cells and a bovine growth hormone polyadenylation signal sequence to increase the steady-state level of RNA after transcription. You can.
형질전환 세포transformed cells
본 발명의 또 다른 측면은, 상기 항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체를 암호화하는 폴리뉴클레오티드를 포함하는 발현 벡터가 도입된 형질전환 세포를 제공한다. 이때, 항-TIGIT 항체, 항-TIGIT 항체의 단편, IL-2 단백질 및 이중 특이 항체는 상술한 바와 동일하다.Another aspect of the invention is the anti-TIGIT antibody or fragment thereof; and an expression vector containing a polynucleotide encoding a bispecific antibody containing the IL-2 protein is provided. At this time, the anti-TIGIT antibody, anti-TIGIT antibody fragment, IL-2 protein, and bispecific antibody are the same as described above.
본 명세서에서 사용하는 용어, "형질전환 세포"는 재조합 발현 벡터가 도입될 수 있는 원핵세포 및 진핵세포를 나타낸다. 상기 형질전환 세포는 벡터를 숙주 세포에 도입하여 형질전환시킴으로써 제작할 수 있다. 또한, 상기 벡터에 포함된 폴리뉴클레오티드를 발현시켜 본 발명의 이중 특이 항체를 생산할 수 있다.As used herein, the term “transformed cell” refers to prokaryotic cells and eukaryotic cells into which a recombinant expression vector can be introduced. The transformed cells can be produced by introducing a vector into a host cell and transforming it. Additionally, the bispecific antibody of the present invention can be produced by expressing the polynucleotide contained in the vector.
상기 형질전환은 다양한 방법에 의하여 수행될 수 있다. 본 발명의 이중 특이 항체를 생산할 수 있는 한, 특별히 이에 제한되지 않는다, 구체적으로, 상기 형질전환 방법은 CaCl2 침전법, CaCl2 침전법에 DMSO(dimethyl sulfoxide)라는 환원물질을 사용함으로써 효율을 높인 Hanahan 방법, 전기천공법(electroporation), 인산칼슘 침전법, 원형질 융합법, 실리콘 카바이드 섬유를 이용한 교반법, 아그로박테리아 매개된 형질전환법, PEG를 이용한 형질전환법, 덱스트란 설페이트, 리포펙타민 및 건조/억제 매개된 형질전환 방법 등이 사용될 수 있다. 또한, 감염(infection)을 수단으로 하여 바이러스 입자를 사용하여 목적물을 세포 내로 전달시킬 수 있다. 또한, 유전자 밤바드먼트 등에 의해 벡터를 숙주 세포 내로 도입할 수 있다.The transformation can be performed by various methods. There is no particular limitation thereto, as long as the bispecific antibody of the present invention can be produced. Specifically, the transformation method is a CaCl 2 precipitation method, which increases efficiency by using a reducing substance called DMSO (dimethyl sulfoxide) in the CaCl 2 precipitation method. Hanahan method, electroporation, calcium phosphate precipitation, protoplast fusion method, stirring method using silicon carbide fiber, Agrobacteria-mediated transformation method, transformation method using PEG, dextran sulfate, lipofectamine and Drying/inhibition-mediated transformation methods, etc. may be used. Additionally, the target can be delivered into cells using virus particles through infection. Additionally, vectors can be introduced into host cells by gene bombardment or the like.
또한, 상기 형질전환 세포의 제작에 사용되는 숙주 세포 역시 본 발명의 항체를 생산할 수 있는 한, 특별히 이에 제한되지 않는다. 구체적으로 상기 숙주 세포는 원핵세포, 진핵세포, 포유동물, 식물, 곤충, 균류 또는 세포성 기원의 세포를 포함할 수 있지만 이에 한정되지 않는다. 상기 원핵세포의 일 예로는 대장균을 사용할 수 있다. 또한, 진핵세포의 일 예로는 효모를 사용할 수 있다. 또한, 상기 포유동물 세포로 CHO 세포, F2N 세포, COS 세포, BHK 세포, 바우스(Bowes) 흑색종 세포, HeLa 세포, 911 세포, AT1080 세포, A549 세포, SP2/0 세포, 인간 림프아세포(human lymphoblastoid), NSO 세포, HT-1080 세포, PERC.6 세포, HEK293 세포 또는 HEK293T 세포 등을 사용할 수 있으나, 이에 한정되지 않으며, 당업자에게 알려진 포유동물 숙주 세포로 사용 가능한 세포는 모두 이용 가능하다. Additionally, the host cell used to produce the transformed cell is not particularly limited as long as it is capable of producing the antibody of the present invention. Specifically, the host cells may include, but are not limited to, cells of prokaryotic, eukaryotic, mammalian, plant, insect, fungal or cellular origin. An example of the prokaryotic cell may be Escherichia coli. Additionally, yeast can be used as an example of a eukaryotic cell. In addition, the mammalian cells include CHO cells, F2N cells, COS cells, BHK cells, Bowes melanoma cells, HeLa cells, 911 cells, AT1080 cells, A549 cells, SP2/0 cells, and human lymphoblastoids. ), NSO cells, HT-1080 cells, PERC.6 cells, HEK293 cells, or HEK293T cells can be used, but are not limited to these, and any cell that can be used as a mammalian host cell known to those skilled in the art can be used.
또한, 항체의 치료제로서의 특성을 최적화하거나 기타 다른 목적을 위해, 호스트 세포가 갖고 있는 당화(glycosylation) 관련 유전자를 당업자에게 알려져 있는 방법을 통해 조작하여 항체의 당쇄 패턴(예를 들어, 시알산, 퓨코실화, 당화)을 조정할 수 있다.In addition, in order to optimize the characteristics of the antibody as a therapeutic agent or for other purposes, the glycosylation-related genes of the host cell are manipulated through methods known to those skilled in the art to manipulate the antibody's sugar chain pattern (e.g., sialic acid, fucose). misfire, saccharification) can be adjusted.
이중 특이 항체의 생산방법Method for producing bispecific antibodies
본 발명의 또 다른 측면은, 상기 항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체의 제조 방법을 제공한다.Another aspect of the invention is the anti-TIGIT antibody or fragment thereof; and a method for producing a bispecific antibody comprising the IL-2 protein.
상기 이중 특이 항체의 제조 방법은 i) 상기 형질전환 세포를 배양하는 단계; 및 ii) 이중 특이 항체를 수득하는 단계를 포함할 수 있다. 이때, 항-TIGIT 항체, 항-TIGIT 항체의 단편, IL-2 단백질 및 이중 특이 항체는 상술한 바와 동일하다.The method for producing the bispecific antibody includes i) culturing the transformed cells; and ii) obtaining a bispecific antibody. At this time, the anti-TIGIT antibody, anti-TIGIT antibody fragment, IL-2 protein, and bispecific antibody are the same as described above.
본 명세서에서 사용하는 용어, "배양"이란, 미생물을 적당히 인공적으로 조절한 환경조건에서 생육시키는 방법을 의미한다.The term “culture” used in this specification refers to a method of growing microorganisms under appropriately artificially controlled environmental conditions.
상기 형질전환 세포를 배양하는 방법은 당업계에 널리 알려져 있는 방법을 이용하여 수행할 수 있다. 구체적으로, 상기 배양은 본 발명의 항체를 발현시켜서 생산할 수 있는 한 특별히 이에 제한되지 않는다. 구체적으로, 상기 배양은 배치 공정 또는 주입 배치 또는 반복 주입 배치 공정(fed batch or repeated fed batch process)에서 연속식으로 배양할 수 있다.The method of culturing the transformed cells can be performed using methods widely known in the art. Specifically, the culture is not particularly limited as long as it can be produced by expressing the antibody of the present invention. Specifically, the culture may be continuously cultured in a batch process or fed batch or repeated fed batch process.
또한, 배양물로부터 상기 항체를 수득하는 단계는 당업계에 공지된 방법에 의해 수행될 수 있다. 구체적으로, 상기 수득 방법은 생산된 본 발명의 이중 특이 항체를 수득할 수 있는 한, 특별히 이에 제한되지 않는다. 바람직하게는, 상기 수득 방법은 원심분리, 여과, 추출, 분무, 건조, 증발, 침전, 결정화, 전기영동, 분별용해(예를 들면, 암모늄설페이트 침전), 크로마토그래피(예를 들면, 이온 교환, 친화성, 소수성 및 크기배제) 등의 방법일 수 있다.Additionally, the step of obtaining the antibody from the culture may be performed by methods known in the art. Specifically, the obtaining method is not particularly limited as long as the produced bispecific antibody of the present invention can be obtained. Preferably, the obtaining method includes centrifugation, filtration, extraction, spraying, drying, evaporation, precipitation, crystallization, electrophoresis, fractional dissolution (e.g. ammonium sulfate precipitation), chromatography (e.g. ion exchange, It may be a method such as affinity, hydrophobicity, and size exclusion).
이중 특이 항체의 용도Uses of Bispecific Antibodies
본 발명의 또 다른 측면으로, 상기 항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물을 제공한다. 여기서, 항-TIGIT 항체, 항-TIGIT 항체의 단편, IL-2 단백질 및 이중 특이 항체는 상술한 바와 동일하다.In another aspect of the invention, the anti-TIGIT antibody or fragment thereof; and a bispecific antibody containing IL-2 protein as an active ingredient. It provides a pharmaceutical composition for preventing or treating cancer. Here, the anti-TIGIT antibody, fragment of anti-TIGIT antibody, IL-2 protein and bispecific antibody are the same as described above.
본 명세서에서 사용하는 용어, "암"은 정상인 조직세포가 어떤 원인으로 무제한 증식하여 그 생체의 생활현상이나 주위의 조직상태 등에 관계없이 급속한 발육을 계속하는 질환으로 구분되며, 본 발명에서의 암은 인체의 각종 암, 예컨대 위암, 간암, 폐암, 대장암, 유방암, 전립선암, 난소암, 췌장암, 자궁경부암, 갑상선암, 후두암, 급성 골수성 백혈병, 뇌종양, 신경모세포종, 망막 모세포종, 두경부암, 침샘암 및 림프종으로 구성된 군으로부터 선택되는 어느 하나의 암일 수 있으나, 상기 종류에 한정되지는 않는다. 또한, 본 발명의 목적상 방사선에 저항성을 가지는 암일 수 있으나, 이에 제한되는 것은 아니다.As used herein, the term "cancer" is classified as a disease in which normal tissue cells proliferate indefinitely for some reason and continue to grow rapidly regardless of the living phenomenon of the living body or the condition of surrounding tissues, etc. Cancer in the present invention is Various cancers of the human body, such as stomach cancer, liver cancer, lung cancer, colon cancer, breast cancer, prostate cancer, ovarian cancer, pancreas cancer, cervical cancer, thyroid cancer, laryngeal cancer, acute myeloid leukemia, brain tumor, neuroblastoma, retinoblastoma, head and neck cancer, salivary gland cancer, and It may be any cancer selected from the group consisting of lymphoma, but is not limited to the above types. Additionally, for the purposes of the present invention, cancer may be resistant to radiation, but is not limited thereto.
본 발명의 암 예방 또는 치료용 약학 조성물에서 상기 이중 특이 항체는 항암 활성을 나타내거나, 특히, 암의 치료 효과를 나타낼 수 있는 한, 용도, 제형, 배합 목적 등에 따라 임의의 양(유효량)으로 포함될 수 있는데, 통상적인 유효량은 조성물 전체 중량을 기준으로 할 때 0.001 중량% 내지 20.0 중량% 범위 내에서 결정될 것이다. 여기서 "유효량"이란 질환의 상태 개선 또는 치료(treatment) 효과, 특히 암의 상태 개선 또는 치료 효과를 유도할 수 있는 유효성분의 양을 말한다. 이러한 유효량은 당업자의 통상의 능력 범위 내에서 실험적으로 결정될 수 있다.In the pharmaceutical composition for preventing or treating cancer of the present invention, the bispecific antibody may be included in an arbitrary amount (effective amount) depending on the use, formulation, formulation purpose, etc., as long as it exhibits anti-cancer activity or, in particular, can exhibit a cancer treatment effect. A typical effective amount will be determined within the range of 0.001% by weight to 20.0% by weight based on the total weight of the composition. Here, “effective amount” refers to the amount of an active ingredient that can improve the condition of a disease or induce a treatment effect, especially an improvement in the condition of cancer or a treatment effect. Such effective amounts can be determined experimentally within the scope of the ordinary ability of those skilled in the art.
본 명세서에서 사용하는 용어, "치료"는 치료학적 처리 및 예방적 처리를 모두 포함하는 의미로 사용될 수 있으며, 인간을 포함한 포유류에서 질환을 치료하기 위한 적용이나 어떠한 형태의 투약을 모두 포함한다. 또한, 상기 용어는 질환의 진행을 억제하거나 늦추는 것을 포함하며; 손상되거나, 결손된 기능을 회복시키거나, 수리하여, 질환을 부분적이거나 완전하게 완화시키거나; 또는 비효율적인 프로세스를 자극하거나; 심각한 질환을 완화하는 의미를 포함한다. 상기 "예방"은 개체의 병리학적 상태 또는 질환을 완화시키거나 감소시키는 의미로 사용될 수 있다. As used herein, the term “treatment” can be used to include both therapeutic treatment and preventive treatment, and includes both application and any form of medication to treat diseases in mammals, including humans. The term also includes inhibiting or slowing the progression of a disease; Restoring or repairing damaged or missing function, thereby partially or completely relieving a disease; or stimulating inefficient processes; It includes the meaning of alleviating serious diseases. The term “prevention” may be used to mean alleviating or reducing the pathological condition or disease of an individual.
생체이용률과 같은 약동학적 파라미터(pharmacokinetic parameters) 및 클리어런스율(clearance rate)과 같은 기본적인 파라미터(underlying parameters)도 효능에 영향을 줄 수 있다. 따라서, "향상된 효능"(예를 들어, 효능의 개선)은 향상된 약동학적 파라미터 및 향상된 효능에 기인할 수 있으며, 시험 동물 또는 인간 대상체에서 클리어런스율 및 암 질환 치료 또는 개선과 같은 파라미터를 비교하여 측정될 수 있다.Pharmacokinetic parameters such as bioavailability and underlying parameters such as clearance rate may also affect efficacy. Accordingly, “enhanced efficacy” (e.g., improvement in efficacy) may be due to improved pharmacokinetic parameters and improved efficacy, measured by comparing parameters such as clearance rate and treatment or amelioration of cancer disease in test animals or human subjects. It can be.
한편, 본 발명의 약학적 조성물은 "치료학적으로 유효한 양"으로 투여한다.Meanwhile, the pharmaceutical composition of the present invention is administered in a “therapeutically effective amount.”
본 명세서에서 사용하는 용어 "투여"란, 적절한 방법으로 개체에게 소정의 물질을 도입하는 것을 의미하며, 상기 조성물의 투여 경로는 목적 조직에 도달할 수 있는 한 어떠한 일반적인 경로를 통하여 투여될 수 있다. 복강내 투여, 정맥내 투여, 근육내 투여, 피하 투여, 피내 투여, 경구 투여, 국소 투여, 비내 투여, 폐내 투여, 직장내 투여될 수 있으나, 이에 한정되지는 않는다. The term “administration” used herein means introducing a predetermined substance into an individual by an appropriate method, and the composition may be administered through any general route as long as it can reach the target tissue. It may be administered intraperitoneally, intravenously, intramuscularly, subcutaneously, intradermally, orally, topically, intranasally, intrapulmonaryly, or rectally, but is not limited thereto.
본 명세서에서 사용하는 용어 "치료학적으로 유효한 양" 또는 "약학적으로 유효한 양"이란 대상 질환을 예방 또는 치료하는데 유효한 화합물 또는 조성물의 양으로서, 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분하며 부작용을 일으키지 않을 정도의 양을 의미한다. 상기 유효량의 수준은 환자의 건강상태, 질환의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 방법, 투여 시간, 투여 경로 및 배출 비율, 치료 기간, 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. 일 구현예에서 치료학적으로 유효한 양은 암을 치료하는데 효과적인 약물의 양을 의미한다.As used herein, the term "therapeutically effective amount" or "pharmaceutically effective amount" refers to the amount of a compound or composition effective in preventing or treating a target disease, which means treating the disease at a reasonable benefit/risk ratio applicable to medical treatment. It refers to an amount that is sufficient for treatment and does not cause side effects. The level of the effective amount is determined by factors including the patient's health status, type and severity of the disease, activity of the drug, sensitivity to the drug, administration method, administration time, administration route and excretion rate, treatment period, combination or concurrent use of drugs, and It may be determined based on other factors well known in the medical field. In one embodiment, a therapeutically effective amount refers to an amount of drug that is effective in treating cancer.
이때, 상기 약학 조성물은 약학적으로 허용 가능한 담체를 더 포함할 수 있다. 상기 약학적으로 허용 가능한 담체는 환자에게 전달하기에 적절한 비-독성 물질이면 어떠한 담체라도 가능하다. 증류수, 알코올, 지방, 왁스 및 비활성 고체가 담체로 포함될 수 있다. 약물학적으로 허용되는 애쥬번트(완충제, 분산제) 또한 약학 조성물에 포함될 수 있다.At this time, the pharmaceutical composition may further include a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier may be any carrier that is a non-toxic material suitable for delivery to a patient. Distilled water, alcohol, fats, waxes and inert solids may be included as carriers. Pharmacologically acceptable adjuvants (buffers, dispersants) may also be included in the pharmaceutical composition.
구체적으로, 상기 약학 조성물은 약제학적으로 허용되는 담체를 포함하여 당업계에 공지된 통상의 방법으로 투여 경로에 따라 비경구용 제형으로 제조될 수 있다. 여기서 "약제학적으로 허용되는" 의미는 유효성분의 활성을 억제하지 않으면서 적용(처방) 대상이 적응 가능한 이상의 독성을 지니지 않는다는 의미이다.Specifically, the pharmaceutical composition may be prepared into a parenteral formulation according to the route of administration by a conventional method known in the art, including a pharmaceutically acceptable carrier. Here, “pharmaceutically acceptable” means that it does not inhibit the activity of the active ingredient and does not have toxicity beyond what is acceptable for the subject of application (prescription).
상기 약학 조성물이 비경구용 제형으로 제조될 경우, 적합한 담체와 함께 당업계에 공지된 방법에 따라 주사제, 경피 투여제, 비강 흡입제 및 좌제의 형태로 제제화될 수 있다. 주사제로 제제화할 경우 적합한 담체로서는 멸균수, 에탄올, 글리세롤이나 프로필렌 글리콜 등의 폴리올 또는 이들의 혼합물을 사용할 수 있으며, 바람직하게는 링거 용액, 트리에탄올 아민이 함유된 PBS(phosphate buffered saline)나 주사용 멸균수, 5% 덱스트로스 같은 등장 용액 등을 사용할 수 있다. 약제학적 조성물의 제제화와 관련하여서는 당업계에 공지되어 있으며, 구체적으로 문헌[Remington's Pharmaceutical Sciences(19th ed., 1995)] 등을 참조할 수 있다. 상기 문헌은 본 명세서의 일부로서 간주된다.When the pharmaceutical composition is prepared as a parenteral formulation, it can be formulated in the form of injections, transdermal administration, nasal inhalation, and suppositories along with a suitable carrier according to methods known in the art. When formulated as an injection, suitable carriers include sterile water, ethanol, polyols such as glycerol or propylene glycol, or mixtures thereof, preferably Ringer's solution, phosphate buffered saline (PBS) containing triethanol amine, or sterile for injection. Isotonic solutions such as water or 5% dextrose can be used. Regarding the formulation of pharmaceutical compositions, it is known in the art, and specifically, references can be made to the literature [Remington's Pharmaceutical Sciences (19th ed., 1995)]. The above documents are considered part of this specification.
상기 약학 조성물의 바람직한 투여량은 환자의 상태 및 체중, 질병의 정도, 약물형태, 투여경로 및 기간에 따라 다르지만, 당업자에 의해 적절하게 선택될 수 있다. 상기 약학 조성물의 바람직한 투여량은 환자의 상태, 체중, 성별, 연령, 환자의 중증도, 투여 경로에 따라 1일 0.0001 ㎍/㎏ 내지 100 g/㎏ 범위일 수 있다. 투여는 1일 1회 또는 수회로 나누어 이루어질 수 있다. 이러한 투여량은 어떠한 측면으로든 본 발명의 범위를 제한하는 것으로 해석되어서는 아니 된다.The preferred dosage of the pharmaceutical composition varies depending on the patient's condition and weight, degree of disease, drug form, administration route and period, but can be appropriately selected by a person skilled in the art. The preferred dosage of the pharmaceutical composition may range from 0.0001 μg/kg to 100 g/kg per day depending on the patient's condition, weight, gender, age, patient's severity, and administration route. Administration can be done once a day or divided into several times. These dosages should not be construed as limiting the scope of the invention in any respect.
상기 약학 조성물이 적용(처방)될 수 있는 대상은 포유동물 및 사람이며, 특히 사람인 경우가 바람직하다. 본 발명의 약학 조성물은 암 치료 효과를 갖는 것으로 공지된 임의의 화합물이나 천연 추출물을 추가로 포함할 수 있다. The subjects to which the pharmaceutical composition can be applied (prescribed) are mammals and humans, and humans are particularly preferred. The pharmaceutical composition of the present invention may further include any compounds or natural extracts known to have cancer treatment effects.
본 발명의 또 다른 측면은, 암의 예방 또는 치료용 약제를 제조하기 위한 상기 항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체의 용도를 제공한다. 여기서, 암, 예방, 치료, 항-TIGIT 항체, 항-TIGIT 항체의 단편, IL-2 단백질 및 이중 특이 항체는 상술한 바와 동일하다.Another aspect of the present invention is the anti-TIGIT antibody or fragment thereof for preparing a medicament for preventing or treating cancer; and the use of a bispecific antibody comprising the IL-2 protein. Here, the cancer, prevention, treatment, anti-TIGIT antibody, fragment of anti-TIGIT antibody, IL-2 protein and bispecific antibody are the same as described above.
본 발명의 또 다른 측면은, 암을 예방 또는 치료하기 위한 상기 항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체의 용도를 제공한다. 여기서, 암, 예방, 치료, 항-TIGIT 항체, 항-TIGIT 항체의 단편, IL-2 단백질 및 이중 특이 항체는 상술한 바와 동일하다.Another aspect of the present invention is the anti-TIGIT antibody or fragment thereof for preventing or treating cancer; and the use of a bispecific antibody comprising the IL-2 protein. Here, the cancer, prevention, treatment, anti-TIGIT antibody, fragment of anti-TIGIT antibody, IL-2 protein and bispecific antibody are the same as described above.
본 발명의 또 다른 측면은, 상기 항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체를 개체에 투여하는 단계를 포함하는 암을 예방 또는 치료하는 방법을 제공한다. 여기서, 암, 예방, 치료, 항-TIGIT 항체, 항-TIGIT 항체의 단편, IL-2 단백질 및 이중 특이 항체 및 투여는 상술한 바와 동일하다. 상기 개체는 포유동물일 수 있으며, 바람직하게는 인간일 수 있다. 또한, 상기 개체는 암을 앓는 환자이거나 암을 앓을 가능성이 큰 개체일 수 있다. Another aspect of the invention is the anti-TIGIT antibody or fragment thereof; and administering to a subject a bispecific antibody containing the IL-2 protein. Here, cancer, prevention, treatment, anti-TIGIT antibody, fragment of anti-TIGIT antibody, IL-2 protein and bispecific antibody and administration are the same as described above. The subject may be a mammal, preferably a human. Additionally, the individual may be a patient suffering from cancer or an individual with a high risk of suffering from cancer.
또한, 상기 이중 특이 항체는 암 치료 효과를 갖는 것으로 공지된 임의의 화합물이나 천연 추출물과 병용하여 투여되거나, 다른 약물과의 조합 제제 형태로 제형화될 수 있다.Additionally, the bispecific antibody may be administered in combination with any compound or natural extract known to have a cancer treatment effect, or may be formulated in the form of a combination preparation with other drugs.
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 예시하기 위한 것으로, 본 발명의 범위가 이들 실시예에 의해 제한되는 것으로 해석되지 않는 것은 당업계에서 통상의 지식을 가진 자에게 있어서 자명할 것이다.Hereinafter, the present invention will be described in more detail through examples. These examples are only for illustrating the present invention, and it will be apparent to those skilled in the art that the scope of the present invention is not to be construed as limited by these examples.
실시예 1. 이중 특이 항체의 제조Example 1. Preparation of bispecific antibodies
실시예 1.1. 항-TIGIT-IgG4 Fc-IL-2 변이체의 제조: GI-106Example 1.1. Preparation of anti-TIGIT-IgG4 Fc-IL-2 variant: GI-106
TIGIT에 특이적으로 결합하는 항-TIGIT 항체 및 IL-2 변이체를 포함하는 이중 특이 항체를 생산하기 위해, 시그널 펩타이드(서열번호 1), 항-TIGIT 항체 중쇄의 가변 영역(서열번호 2)과 불변 영역(서열번호 3), 링커(1)(서열번호 18), IgG4 Fc 도메인(서열번호 4), 링커(2)(서열번호 5) 및 세 개의 아미노산이 치환된 IL-2 변이체(서열번호 6)를 암호화하는 폴리뉴클레오티드(서열번호 34)와 시그널 펩타이드(서열번호 1), 항-TIGIT 항체 경쇄의 가변 영역(서열번호 7)과 불변 영역(서열번호 8)을 암호화하는 폴리뉴클레오티드(서열번호 35)를 BioXpTM 3250 SYSTEM(Codex DNA)을 이용해 pCGS3 벡터(Sigma-Aldrich®)에 적재하였다.To produce a bispecific antibody comprising an anti-TIGIT antibody and an IL-2 variant that specifically binds to TIGIT, the signal peptide (SEQ ID NO: 1), the variable region (SEQ ID NO: 2) of the anti-TIGIT antibody heavy chain and the constant IL-2 variant in which region (SEQ ID NO: 3), linker (1) (SEQ ID NO: 18), IgG4 Fc domain (SEQ ID NO: 4), linker (2) (SEQ ID NO: 5) and three amino acids are substituted (SEQ ID NO: 6) ), a polynucleotide (SEQ ID NO: 34) encoding a signal peptide (SEQ ID NO: 1), and a polynucleotide (SEQ ID NO: 35) encoding the variable region (SEQ ID NO: 7) and constant region (SEQ ID NO: 8) of the anti-TIGIT antibody light chain. ) was loaded into the pCGS3 vector (Sigma-Aldrich®) using BioXp TM 3250 SYSTEM (Codex DNA).
또한 상기 벡터를 CHO 세포(Expi-CHOTM, Thermo Fisher Scientific)에 도입하여 이중 특이 항체를 발현시켰다. 벡터를 도입한 후 37℃, 125 rpm, 8% CO2 조건에서 배양하였다. 그 후, 세포의 생존율이 50%일 때 배양액을 수거하여 이중 특이 항체를 정제하였다. 상기 정제한 이중 특이 항체를 "GI-106"으로 명명하였다(도 1). Additionally, the vector was introduced into CHO cells (Expi-CHO , Thermo Fisher Scientific) to express bispecific antibodies. After introducing the vector, it was cultured at 37°C, 125 rpm, and 8% CO 2 conditions. Afterwards, when the cell survival rate was 50%, the culture medium was collected and the bispecific antibody was purified. The purified bispecific antibody was named “GI-106” (Figure 1).
구체적으로 Protein A resin이 포함된 크로마토그래피를 이용하여 정제하였다. 수거한 배양액을 여과시킨 후, 여과된 배양액을 컬럼(column)에 흘려주며 결합시켰다. 그 후, 50 mM 글리신(glycine), pH 3.4으로 이중 특이 항체를 수거하였다. 수거된 이중 특이 항체가 포함된 버퍼를 투석을 이용하여 PBS 버퍼로 바꾸어 주고, 농도를 측정하였다. NanoDrop 장비(Thermo Fisher Scientific)를 사용하여 검출 시 1.52 mg/mL의 농도로 이중 특이 항체가 포함된 것을 확인하였다.Specifically, it was purified using chromatography containing Protein A resin. After filtering the collected culture, the filtered culture was flowed through a column and combined. Afterwards, the bispecific antibody was collected with 50 mM glycine, pH 3.4. The buffer containing the collected bispecific antibodies was changed to PBS buffer using dialysis, and the concentration was measured. When detected using NanoDrop equipment (Thermo Fisher Scientific), it was confirmed that a bispecific antibody was included at a concentration of 1.52 mg/mL.
분리 정제된 이중 특이 항체(GI-106)는 환원(R, reduced) 또는 비환원(NR, non-reduced) 조건하에서 SDS-PAGE를 통해 분자량을 확인하고(도 2), 크기 배제 크로마토그래피(SEC) 분석을 통해 순도를 확인하였다. 그 결과, 분리 정제된 이중 특이 항체의 순도는 87.42%임을 확인하였다.The molecular weight of the separated and purified bispecific antibody (GI-106) was confirmed through SDS-PAGE under reduced (R, reduced) or non-reduced (NR, non-reduced) conditions (Figure 2), and size exclusion chromatography (SEC). ) Purity was confirmed through analysis. As a result, the purity of the separated and purified bispecific antibody was confirmed to be 87.42%.
실시예 1.2. 항-TIGIT-IgG4 Fc-IL-2 변이체의 제조: GI-106B7NH06kv3Example 1.2. Preparation of anti-TIGIT-IgG4 Fc-IL-2 variant: GI-106B7NH06kv3
TIGIT에 특이적으로 결합하는 항-TIGIT 항체에 IL-2 변이체를 포함하는 이중 특이 항체를 생산하기 위해, 시그널 펩타이드(서열번호 1), 항-TIGIT 항체 중쇄의 가변 영역(서열번호 2)과 불변 영역(서열번호 3), 링커(1)(서열번호 18) 및 IgG4 FcM1 도메인(서열번호 23)을 암호화하는 폴리뉴클레오티드(서열번호 36)와 시그널 펩타이드(서열번호 1), 항-TIGIT 항체 경쇄의 가변 영역(서열번호 7)과 불변 영역(서열번호 24), 링커(3)(서열번호 5) 및 세 개의 아미노산이 치환된 IL-2 변이체(서열번호 6)를 암호화하는 폴리뉴클레오티드(서열번호 37)를 BioXpTM 3250 SYSTEM을 이용해 pCGS3 벡터(Sigma-Aldrich®)에 적재하였다.To produce a bispecific antibody comprising an anti-TIGIT antibody that specifically binds to TIGIT and an IL-2 variant, the signal peptide (SEQ ID NO: 1), the variable region (SEQ ID NO: 2) of the anti-TIGIT antibody heavy chain and the constant region (SEQ ID NO: 3), a polynucleotide (SEQ ID NO: 36) encoding the linker (1) (SEQ ID NO: 18) and an IgG4 FcM1 domain (SEQ ID NO: 23) and a signal peptide (SEQ ID NO: 1) of the anti-TIGIT antibody light chain. A polynucleotide encoding an IL-2 variant (SEQ ID NO: 6) with a variable region (SEQ ID NO: 7), a constant region (SEQ ID NO: 24), a linker (3) (SEQ ID NO: 5), and three amino acids substituted (SEQ ID NO: 37) ) was loaded into the pCGS3 vector (Sigma-Aldrich®) using BioXp TM 3250 SYSTEM.
또한, 상기 벡터를 CHO 세포(Expi-CHOTM, Thermo Fisher Scientific)에 도입하여 이중 특이 항체를 발현시켰다. 벡터를 도입한 후 37℃, 127 rpm, 5% CO2, 80% 습도 조건에서 7일간 배양하였다. 그 후, 배양액을 수거하여 이중 특이 항체를 정제하였다. 상기 정제한 이중 특이 항체를 "GI-106B7NH06Kv3"으로 명명하였다(도 3).Additionally, the vector was introduced into CHO cells (Expi-CHO , Thermo Fisher Scientific) to express bispecific antibodies. After introducing the vector, it was cultured for 7 days at 37°C, 127 rpm, 5% CO 2 , and 80% humidity. Afterwards, the culture medium was collected and the bispecific antibody was purified. The purified bispecific antibody was named “GI-106B7NH06Kv3” (Figure 3).
이중 특이 항체의 구체적인 정제 방법으로 Protein A resin이 포함된 친화 크로마토그래피를 이용하여 정제하였다. 세포 배양과정에서 발생하는 세포 잔해물을 포함한 불순물을 여과를 통해 제거한 후, 여과된 배양액을 컬럼에 흘려주며 결합시켰다. 그 후, 100 mM 글리신 및 100 mM 아르기닌(arginine)이 포함된 버퍼(pH 3.0)를 순차적으로 흘려주어 이중 특이 항체를 수거하였다. 수거된 이중 특이 항체가 포함된 버퍼를 투석을 이용하여 4% 수크로스(sucrose), 50 mM 히스티딘(histidine) 및 50 mM 아르기닌(pH 7.4)이 포함된 버퍼로 바꾸어 주고, 농도를 측정하였다. Nanodrop 장비를 사용하여 검출 시 1.87 mg/mL의 농도로 이중 특이 항체가 포함된 것을 확인하였다.As a specific purification method for the bispecific antibody, it was purified using affinity chromatography containing Protein A resin. After removing impurities including cell debris generated during the cell culture process through filtration, the filtered culture medium was flowed through the column and combined. Afterwards, a buffer (pH 3.0) containing 100mM glycine and 100mM arginine was sequentially flowed to collect the bispecific antibody. The buffer containing the collected bispecific antibodies was changed to a buffer containing 4% sucrose, 50 mM histidine, and 50 mM arginine (pH 7.4) using dialysis, and the concentration was measured. When detected using Nanodrop equipment, it was confirmed that a bispecific antibody was included at a concentration of 1.87 mg/mL.
분리 정제된 이중 특이 항체(GI-106B7NH06Kv3)는 환원(R, reduced) 또는 비환원(NR, non-reduced) 조건하에서 SDS-PAGE로 분자량을 확인하고(도 4), 크기 배제 크로마토그래피(SEC) 분석을 통해 순도를 확인하였다. 그 결과, 분리 정제된 이중 특이 항체의 순도는 88.24%임을 확인하였다.The molecular weight of the separated and purified bispecific antibody (GI-106B7NH06Kv3) was confirmed by SDS-PAGE under reducing (R, reduced) or non-reducing (NR, non-reduced) conditions (Figure 4), and size exclusion chromatography (SEC). Purity was confirmed through analysis. As a result, the purity of the separated and purified bispecific antibody was confirmed to be 88.24%.
실시예 1.3. 항-TIGIT 항체의 제조Example 1.3. Preparation of anti-TIGIT antibodies
대조군인 항-TIGIT 항체를 생산하기 위하여, 시그널 펩타이드(서열번호 1), 항-TIGIT 항체 중쇄의 가변 영역(서열번호 2)과 불변 영역(서열번호 3), 링커(1)(서열번호 18), 및 IgG4 Fc 도메인(서열번호 4)을 암호화하는 폴리뉴클레오티드(서열번호 22)와 시그널 펩타이드(서열번호 1), 항-TIGIT 항체 경쇄의 가변 영역(서열번호 7)과 불변 영역(서열번호 8)을 암호화하는 폴리뉴클레오티드(서열번호 35)를 GenScript사 Gene Synthesis 서비스를 통해 합성하여 pcDNA3.4 벡터에 적재하였다.To produce a control anti-TIGIT antibody, signal peptide (SEQ ID NO: 1), variable region (SEQ ID NO: 2) and constant region (SEQ ID NO: 3) of the anti-TIGIT antibody heavy chain, and linker (1) (SEQ ID NO: 18) , and a polynucleotide (SEQ ID NO: 22) and signal peptide (SEQ ID NO: 1) encoding the IgG4 Fc domain (SEQ ID NO: 4), the variable region (SEQ ID NO: 7) and the constant region (SEQ ID NO: 8) of the anti-TIGIT antibody light chain. The polynucleotide (SEQ ID NO: 35) encoding was synthesized through GenScript's Gene Synthesis service and loaded into the pcDNA3.4 vector.
또한 상기 벡터를 CHO 세포(Expi-CHOTM, Thermo Fisher Scientific)에 도입하여 대조항체 항-TIGIT 항체를 발현시켰다. 벡터를 도입한 후 37℃, 125 rpm, 8% CO2 조건에서 배양 후, 세포의 생존율이 50%일 때 배양액을 수거하여 상기 대조항체 항-TIGIT 항체를 정제하였다. 정제는 실시예 1.1과 동일한 방법으로 수행하였다.Additionally, the vector was introduced into CHO cells (Expi-CHO , Thermo Fisher Scientific) to express the control anti-TIGIT antibody. After introducing the vector, the culture medium was cultured at 37°C, 125 rpm, 8% CO 2 , and when the cell survival rate was 50%, the culture medium was collected and the control anti-TIGIT antibody was purified. Purification was performed in the same manner as Example 1.1.
실시예 1.4. GI-106B7NH06-CN의 제조Example 1.4. Manufacturing of GI-106B7NH06-CN
시그널 펩타이드(서열번호 1), 항-TIGIT 항체 중쇄의 가변 영역(서열번호 2)과 불변 영역(서열번호 3), 링커(1)(서열번호 18) 및 IgG4 FcM1 도메인(서열번호 23)을 암호화하는 폴리뉴클레오티드(서열번호 36)와 시그널 펩타이드(서열번호 1), 항-TIGIT 항체 경쇄의 가변 영역(서열번호 7)과 불변 영역(서열번호 24)를 암호화하는 폴리뉴클레오티드(서열번호 38)를 BioXpTM 3250 SYSTEM을 이용해 pCGS3 벡터(Sigma-Aldrich®)에 적재하였다.Signal peptide (SEQ ID NO: 1), encoding the variable region (SEQ ID NO: 2) and constant region (SEQ ID NO: 3) of the anti-TIGIT antibody heavy chain, linker (1) (SEQ ID NO: 18), and IgG4 FcM1 domain (SEQ ID NO: 23) A polynucleotide (SEQ ID NO: 36), a signal peptide (SEQ ID NO: 1), and a polynucleotide (SEQ ID NO: 38) encoding the variable region (SEQ ID NO: 7) and constant region (SEQ ID NO: 24) of the anti-TIGIT antibody light chain were used as BioXp. It was loaded into pCGS3 vector (Sigma-Aldrich®) using TM 3250 SYSTEM.
또한, 상기 벡터를 CHO 세포(Expi-CHOTM, Thermo Fisher Scientific)에 도입하여 융합단백질을 발현시켰다. 벡터를 도입한 후 37℃, 127 rpm, 5% CO2, 80% 습도 조건에서 7일간 배양하였다. 그 후, 배양액을 수거하여 융합단백질을 정제하였다. 상기 정제한 융합단백질을 "GI-106B7NH06K-CN"으로 명명하였다.Additionally, the vector was introduced into CHO cells (Expi-CHO , Thermo Fisher Scientific) to express the fusion protein. After introducing the vector, it was cultured for 7 days at 37°C, 127 rpm, 5% CO 2 , and 80% humidity. Afterwards, the culture medium was collected and the fusion protein was purified. The purified fusion protein was named “GI-106B7NH06K-CN”.
실시예 1.5. Fc-IL2v3의 제조Example 1.5. Preparation of Fc-IL2v3
시그널 펩타이드(서열번호 1), 링커(1)(서열번호 18) 및 IgG4 FcM1 도메인(서열번호 23), 링커(2)(서열번호 5) 및 세 개의 아미노산이 치환된 IL-2 변이체(서열번호 6)를 포함하는 폴리뉴클레오티드(서열번호 39)를 BioXPTM 3250 SYSTEM을 이용해 pCGS3 벡터(Sigma-Aldrich®)에 적재하였다.Signal peptide (SEQ ID NO: 1), linker (1) (SEQ ID NO: 18) and IgG4 FcM1 domain (SEQ ID NO: 23), linker (2) (SEQ ID NO: 5) and an IL-2 variant with three amino acids substituted (SEQ ID NO: 6) The polynucleotide containing (SEQ ID NO: 39) was loaded into the pCGS3 vector (Sigma-Aldrich®) using BioXP TM 3250 SYSTEM.
또한, 상기 벡터를 CHO 세포(Expi-CHOTM, Thermo Fisher Scientific)에 도입하여 융합단백질을 발현시켰다. 벡터를 도입한 후 37℃, 127 rpm, 5% CO2, 80% 습도 조건에서 7일간 배양하였다. 그 후, 배양액을 수거하여 Fc-IL2v3 융합단백질을 정제하였다. Additionally, the vector was introduced into CHO cells (Expi-CHO , Thermo Fisher Scientific) to express the fusion protein. After introducing the vector, it was cultured for 7 days at 37°C, 127 rpm, 5% CO 2 , and 80% humidity. Afterwards, the culture medium was collected and the Fc-IL2v3 fusion protein was purified.
실시예 2. 시험관 내 환경(Example 2. In vitro environment ( in vitroin vitro )에서 GI-106에 의한 TIGIT/CD155 결합 저해 능력 확인) confirmed the ability of GI-106 to inhibit TIGIT/CD155 binding
본 실험은 시험관 내 환경에서 GI-106 및 대조항체 항-TIGIT 항체 처리 시 TIGIT 저해 효과를 평가한 실험이다. 구체적으로, TIGIT/CD155 blockade bioassay 키트(cat. J2205, Promega)를 사용하여 실험을 진행하였다. 본 실험에 TIGIT 효과기(effector) 세포 및 CD155 aAPC/CHO-K1 세포를 사용하였고, 항-TIGIT 항체 처리로 인해 TIGIT/CD155 상호 작용이 차단되고 CD226 신호 경로가 활성화되는 것을 발광 정도를 정량화하여 측정하였다(도 5).This experiment evaluated the effect of TIGIT inhibition upon treatment with GI-106 and control anti-TIGIT antibody in an in vitro environment. Specifically, the experiment was conducted using the TIGIT/CD155 blockade bioassay kit (cat. J2205, Promega). TIGIT effector cells and CD155 aAPC/CHO-K1 cells were used in this experiment, and the blocking of TIGIT/CD155 interaction and activation of the CD226 signaling pathway due to anti-TIGIT antibody treatment was measured by quantifying the degree of luminescence. (Figure 5).
액체질소에서 보관 중인 TIGIT 효과기 세포를 37℃ 항온수조에서 3분간 녹인 다음, 예열된 12 mL 어세이 버퍼(90% RPMI 1640(Promega)+10% FBS(Promega))에 0.5 mL 넣어 현탁하였다. 그 후, 96-웰-플레이트(96-well-white cell culture plate, cat. 3917, Corning)에 각 웰(well) 당 현탁물을 80 μL씩 첨가한 후, 37℃, 5% CO2 조건의 배양기에서 20시간 동안 배양하였다.TIGIT effector cells stored in liquid nitrogen were dissolved in a constant temperature water bath at 37°C for 3 minutes and then suspended in 0.5 mL of preheated 12 mL assay buffer (90% RPMI 1640 (Promega) + 10% FBS (Promega)). Afterwards, 80 μL of the suspension was added to each well in a 96-well-white cell culture plate (cat. 3917, Corning), and then incubated at 37°C and 5% CO 2 conditions. It was cultured in an incubator for 20 hours.
20시간 후, 어세이 버퍼를 이용하여 GI-106 및 대조항체인 항-TIGIT 항체를 희석하여, 다양한 농도의 시험물질을 각 웰 당 20 μL씩 첨가하였다. 음성 대조군의 경우 어세이 버퍼를 20 μL 첨가하였다. 이후 CD155 발현 aAPC/CHO-K1 세포를 분주하기 전까지 96-웰-플레이트를 상온에 두었다. After 20 hours, GI-106 and the control anti-TIGIT antibody were diluted using assay buffer, and 20 μL of test substances at various concentrations were added to each well. For the negative control, 20 μL of assay buffer was added. Afterwards, the 96-well plate was left at room temperature until aAPC/CHO-K1 cells expressing CD155 were dispensed.
액체질소에서 보관 중인 CD155 발현 aAPC/CHO-K1 세포를 37℃ 항온수조에서 3분간 녹인 다음, 예열된 3 mL 어세이 버퍼에 0.5 mL 넣어 현탁한 후, TIGIT 효과기 세포와 GI-106 또는 대조항체가 있는 96-웰-플레이트에 각 웰당 20 μL씩 분주하였다. 음성 대조군의 경우 어세이 버퍼를 20 μL를 첨가하여 최종 용량을 동일하게 하였다. 이후 37℃, 5% CO2 조건의 배양기에서 6시간 동안 배양하였다. CD155-expressing aAPC/CHO-K1 cells stored in liquid nitrogen were dissolved in a water bath at 37°C for 3 minutes, then suspended in 0.5 mL of preheated 3 mL assay buffer, followed by TIGIT effector cells and GI-106 or control antibody. 20 μL was dispensed into each well in a 96-well plate. For the negative control, 20 μL of assay buffer was added to make the final volume the same. Afterwards, it was cultured for 6 hours in an incubator under 37°C and 5% CO 2 conditions.
6시간 반응 후 배양기에서 96-웰-플레이트를 꺼내 상온에 15분 동안 놓아두었다. 이후 상온에서 미리 녹인 Bio-Glo™ reagent(Promega)를 각 웰당 120 μL씩 첨가하였고, 백그라운드(background) 신호 보정을 위해 가장자리 웰에도 동량을 첨가하였다. 10분 간 상온에서 반응시킨 후, Glomax Luminometer(cat. GM3000, Promega)를 이용해 발광 정도를 측정하였으며 결과는 RLU(Relative Light Unit) 그래프로 나타내었다.After 6 hours of reaction, the 96-well plate was removed from the incubator and left at room temperature for 15 minutes. Afterwards, 120 μL of Bio-Glo™ reagent (Promega) pre-dissolved at room temperature was added to each well, and the same amount was also added to the edge wells for background signal correction. After reacting at room temperature for 10 minutes, the degree of luminescence was measured using a Glomax Luminometer (cat. GM3000, Promega), and the results were expressed in a RLU (Relative Light Unit) graph.
그 결과, GI-106 및 항-TIGIT 항체는 TIGIT 효과기 세포에 발현된 TIGIT과 농도 의존적으로 결합하여, 발광 신호를 증가시킴을 확인하였다. 이는 GI-106의 항-TIGIT 항체가 TIGIT 효과기 세포 표면에 발현된 TIGIT에 결합함으로써 CD155와의 결합을 저해시키고, 결과적으로 CD155가 CD226에 결합하여 신호 전달을 활성화시켜 발광 신호를 증가시킨 것을 의미한다(도 6).As a result, it was confirmed that GI-106 and anti-TIGIT antibodies bind to TIGIT expressed in TIGIT effector cells in a concentration-dependent manner, increasing the luminescence signal. This means that the anti-TIGIT antibody of GI-106 binds to TIGIT expressed on the surface of the TIGIT effector cell, thereby inhibiting the binding to CD155, and as a result, CD155 binds to CD226 to activate signal transduction and increase the luminescent signal ( Figure 6).
실시예 3. GI-106의 IL-2 변이체에 의해 유도되는 JAK-STAT 경로 활성화 능력 확인Example 3. Confirmation of JAK-STAT pathway activation ability induced by IL-2 variant of GI-106
본 실험은 GI-106의 IL-2 변이체 부위의 활성을 확인하기 위한 실험이다. 구체적으로, HEK-BlueTM IL-2 리포터(reporter) 세포(cat. hkb-il2, InvivoGen Inc.)를 이용하여 실험을 진행하였다. HEK-BlueTM IL-2 리포터 세포는 IL-2에 의해 JAK-STAT 경로가 활성화될 경우 리포터 단백질인 SEAP(secreted embryonic alkaline phosphatase) 단백질 생산이 유도된다. 본 실험에서는 SEAP 단백질을 검출함으로써 IL-2에 의한 활성화 능력을 확인하였다. This experiment is to confirm the activity of the IL-2 variant portion of GI-106. Specifically, the experiment was conducted using HEK-Blue TM IL-2 reporter cells (cat. hkb-il2, InvivoGen Inc.). HEK-Blue TM IL-2 reporter cells are induced to produce the reporter protein SEAP (secreted embryonic alkaline phosphatase) protein when the JAK-STAT pathway is activated by IL-2. In this experiment, the activation ability by IL-2 was confirmed by detecting SEAP protein.
HEK-BlueTM IL-2 리포터 세포는 10% FBS(Gibco)와 100 U/mL 페니실린(Welgene Inc.), 100 μg/mL 스트렙토마이신(Welgene Inc.), 100 μg/mL NormocinTM(cat. Ant-nr-1, InvivoGen Inc.)을 포함하는 DMEM 배지(Gibco)에서 배양하였다. HEK-BlueTM IL-2 리포터 세포는 계대배양하여 세포를 안정화시킨 후, 트립신(Gibco)을 이용하여 세포를 수확하였다. 그 후, PBS로 세척하여 죽은 세포를 제거하였다. 분리된 세포는 배양 배지에 약 2.8Х105 세포/mL이 되도록 세포 현탁액을 만들었다.HEK-Blue TM IL-2 reporter cells were incubated with 10% FBS (Gibco), 100 U/mL penicillin (Welgene Inc.), 100 μg/mL streptomycin (Welgene Inc.), and 100 μg/mL Normocin TM (cat. Ant. -nr-1, InvivoGen Inc.) was cultured in DMEM medium (Gibco) containing. HEK-Blue TM IL-2 reporter cells were subcultured to stabilize the cells, and then the cells were harvested using trypsin (Gibco). Afterwards, dead cells were removed by washing with PBS. The separated cells were created into a cell suspension in the culture medium so that the concentration was about 2.8Х10 5 cells/mL.
GI-106 및 양성 대조군인 Aldesleukin(상품명: Proleukin®, Novartis, 이하, 프로류킨)과 음성 대조군인 재조합 인간 TGF-β1(cat. rcyc-htgfb1, InvivoGen Inc.)은 PBS를 이용하여 희석하여 96-웰-플레이트(cat. 30096, SPL)에 각 웰당 20 μL씩 분주하였다. 시험 물질이 들어간 96-웰-플레이트에 준비해둔 세포 현탁액을 약 5Х104 세포수가 되도록 각 웰당 180 μL씩 넣어주고 37℃, 5% CO2 조건의 배양기에서 24시간 동안 배양하였다.GI-106, Aldesleukin (Product name: Proleukin®, Novartis, hereinafter referred to as Proleukin) as a positive control, and recombinant human TGF-β1 (cat. rcyc-htgfb1, InvivoGen Inc.) as a negative control were diluted using PBS and incubated at 96- 20 μL was dispensed into each well of a well-plate (cat. 30096, SPL). 180 μL of the cell suspension prepared in the 96-well plate containing the test substance was added to each well to obtain a cell count of approximately 5Х10 4 and cultured in an incubator at 37°C and 5% CO 2 for 24 hours.
24시간 후, 배양기에서 96-웰-플레이트를 꺼내 300Хg에서 5분간 원심 분리하여 상층액 20 μL씩을 새로운 96-웰-플레이트로 옮겼다. 상층액이 담긴 각 웰에 상온에서 녹인 QUANTI-BlueTM solution(cat. Rep-qbs, InvivoGen Inc,)을 180 μL씩 분주하고, 이후 37℃, 5% CO2 조건의 배양기에서 2시간 동안 반응하였다. 반응 후, 분광광도계(VersaMaxTM Absorbance Microplate Reader)를 이용하여 630 nm 파장에서 흡광도를 측정하였다.After 24 hours, the 96-well plate was removed from the incubator, centrifuged at 300Хg for 5 minutes, and 20 μL of the supernatant was transferred to a new 96-well plate. 180 μL of QUANTI-Blue TM solution (cat. Rep-qbs, InvivoGen Inc,) dissolved at room temperature was dispensed into each well containing the supernatant, and then reacted in an incubator at 37°C and 5% CO 2 for 2 hours. . After the reaction, the absorbance was measured at a wavelength of 630 nm using a spectrophotometer (VersaMax TM Absorbance Microplate Reader).
그 결과, GI-106 및 프로류킨에 의해 농도 의존적으로 흡광도가 증가하는 것을 확인하였다. 이를 통해, GI-106의 IL-2 변이체 부위에 의해 JAK-STAT 신호 전달 경로가 활성화되는 것을 확인하였다(도 7). GI-106의 EC50 값은 13.13 ng/mL, 양성 대조군 프로류킨의 EC50 값은 0.766 ng/mL로 확인되었다.As a result, it was confirmed that the absorbance increased in a concentration-dependent manner due to GI-106 and proleukin. Through this, it was confirmed that the JAK-STAT signaling pathway was activated by the IL-2 variant portion of GI-106 (Figure 7). The EC 50 value of GI-106 was confirmed to be 13.13 ng/mL, and the EC 50 value of the positive control proleukin was confirmed to be 0.766 ng/mL.
실시예 4. 인간 유래 유방암세포 식립 마우스에서 GI-106 투여에 의한 항암 효과 확인Example 4. Confirmation of anticancer effect by GI-106 administration in mice implanted with human-derived breast cancer cells
본 실험은 인간 면역시스템이 도입된 마우스에 MDA-MB-231(human breast cancer cells) 세포를 이식한 종양 모델에서 시험물질 GI-106을 투여한 후 종양의 성장 억제효과를 평가한 것이다.This experiment evaluated the tumor growth inhibition effect after administering the test substance GI-106 in a tumor model in which MDA-MB-231 (human breast cancer cells) cells were transplanted into mice with a human immune system.
인간 면역시스템이 도입된 마우스 모델을 제작하기 위해, NSG-B2m 암컷 마우스(7주령, The jackson laboratory)에 사람 말초혈액 세포(StemExpress, LLC) 부유액을 마리 당 1Х107 세포/200 μL씩 일회용 주사기(31G, cat. 328820, BD medical diabetes care)에 충진하여 꼬리 미정맥을 통해 투여하였다. 세포 이식 후, 매일 1회 일반증상을 관찰하였다.To create a mouse model incorporating the human immune system, a suspension of human peripheral blood cells (StemExpress, LLC) was added to NSG-B2m female mice (7 weeks old, The jackson laboratory) at 1Х107 cells/200 μL per mouse using a disposable syringe ( 31G, cat. 328820, BD medical diabetes care) and administered through the caudal vein. After cell transplantation, general symptoms were observed once daily.
MDA-MB-231 세포는 한국세포주은행(KCLB No.30026)으로부터 분양 받아 10% FBS(Gibco) 및 1% 항생제/항진균제(Gibco)를 포함하는 RPMI1640 배지(Gibco)에서 배양하였다. 배양한 MDA-MB-231 세포는 트립신(Gibco)을 이용하여 수확한 후 PBS에 현탁하였다. 이종이식 마우스 종양 모델을 확립하기 위해 사람 말초혈액 세포 이식 후 5일차의 건강한 마우스에 MDA-MB-231 세포 부유액(5Х106 세포/0.05 mL) 및 0.05 mL BD Matrigel matrix phenol red-free(cat. 356237, BD Biosciences, USA)를 혼합하여 조제한 용액을 일회용 주사기(31G, cat. 328820, BD medical diabetes care)에 충진하여 동물의 우측 등 부위의 피하에 마리 당 0.1 mL씩 투여하여 이식하였다. MDA-MB-231 세포 이식 후, 생착 및 성장기간 동안 매일 1회 일반증상을 관찰하였다. MDA-MB-231 cells were purchased from the Korea Cell Line Bank (KCLB No. 30026) and cultured in RPMI1640 medium (Gibco) containing 10% FBS (Gibco) and 1% antibiotic/antifungal agent (Gibco). Cultured MDA-MB-231 cells were harvested using trypsin (Gibco) and suspended in PBS. To establish a xenograft mouse tumor model, healthy mice 5 days after human peripheral blood cell transplantation were incubated with MDA-MB-231 cell suspension (5Х10 6 cells/0.05 mL) and 0.05 mL BD Matrigel matrix phenol red-free (cat. 356237). , BD Biosciences, USA) was filled into a disposable syringe (31G, cat. 328820, BD medical diabetes care) and administered 0.1 mL per animal subcutaneously to the right back of the animal. After MDA-MB-231 cell transplantation, general symptoms were observed once daily during the engraftment and growth period.
MDA-MB-231 세포를 이식하고 약 20일 후에 동물의 건강상태에 이상이 없는 마우스에 대해 종양의 부피를 측정하여, 각 군의 평균이 40~80 mm3에 도달하도록 개체 12마리를 선별하였다. 개체 선정은 개체의 생리적 상태(호흡, 털, 행동, 꼬리, 자세, 체액, 식이, 형태변형, 대사 등), 체중 변화, FACS 분석 결과 및 종양 성장속도를 고려하여 선발하였다. 선별된 동물은 종양의 부피 및 체중을 기초로 하여 가능한 균등하도록 군당 6마리씩 분리하였다. 표 3과 같이 시험군을 구성하고 시험물질을 투여하였다.Approximately 20 days after transplanting MDA-MB-231 cells, the tumor volume was measured on mice with no abnormalities in the animal's health, and 12 individuals were selected so that the average of each group reached 40~80 mm 3 . Subjects were selected considering their physiological status (respiration, fur, behavior, tail, posture, body fluids, diet, deformation, metabolism, etc.), weight change, FACS analysis results, and tumor growth rate. The selected animals were divided into 6 animals per group to be as equal as possible based on tumor volume and body weight. As shown in Table 3, the test group was formed and the test substance was administered.
army 투여물질administered substance 투여경로Route of administration 투여주기Dosing cycle 투여용량Dosage 개체수population
G1G1 비이클 (PBS)Vehicle (PBS) i.v.i.v. BIW (2회/주)BIW (2 times/week) - mg/kg-mg/kg 66
G2G2 GI-106GI-106 i.v.i.v. BIW (2회/주)BIW (2 times/week) 6 mg/kg6mg/kg 66
시험물질 투여 후 25일의 시험기간 동안 매일 1회 외관, 행동 및 배설물 등의 일반증상을 관찰하여 개체별로 기록하였으며, 사망동물을 확인하였다. 종양의 부피 측정은 관찰기간 동안 주 2회, 캘리퍼스(digital caliper, mitutoyo)를 사용하여 종양의 장축(L, maximum length)과 단축(W, perpendicular width)을 측정하였고, 아래의 수학식 I에 대입하여 종양의 부피(TV, tumor volume)와 종양 성장 억제율(TGI, tumor growth inhibition)을 측정하였다.<수학식 I> During the 25-day test period after administration of the test substance, general symptoms such as appearance, behavior, and excrement were observed and recorded for each individual once a day, and dead animals were identified. Tumor volume was measured twice a week during the observation period using a caliper (digital caliper, mitutoyo) to measure the long axis (L, maximum length) and short axis (W, perpendicular width) of the tumor, and substitute them into Equation I below. Tumor volume (TV) and tumor growth inhibition (TGI) were measured. <Equation I>
TV(mm3)=(W2ΥL)/2TV(mm 3 )=(W 2 ΥL)/2
TGI=(1-(Ti-T0)/(Vi-V0))Х100TGI=(1-(Ti-T0)/(Vi-V0))Х100
Ti: 시험종료 시점에서 시험물질 투여군의 종양 볼륨Ti: Tumor volume of the test substance administered group at the end of the test
T0: 군분리 시점에서 시험물질 투여군의 종양 볼륨T0: Tumor volume of the test substance administered group at the time of group separation
Vi: 시험종료 시점에서 음성대조군의 종양 볼륨Vi: Tumor volume of the negative control group at the end of the test
V0: 군분리 시점에서 음성대조군의 종양 볼륨V0: Tumor volume of the negative control group at the time of group separation
각 개체의 투여 전 종양의 부피는 군분리 시 측정된 값으로 설정하였으며, 항종양 효능은 대조군(비이클(PBS), G1)과 비교하여 평가하였다. The tumor volume before administration of each subject was set to the value measured at the time of group separation, and the antitumor efficacy was evaluated by comparison with the control group (vehicle (PBS), G1).
모든 통계 계산은 Prism 8.0(Graph Pad Software Inc.)을 사용하여 수행하였다. 종양 부피 측정의 비교는 이원 분산 분석에 이어 Tukey의 다중 비교 테스트를 통해 이루어졌다. 0.05 미만의 p 값은 유의미한 것으로 간주되었다.All statistical calculations were performed using Prism 8.0 (Graph Pad Software Inc.). Comparison of tumor volume measurements was made using two-way analysis of variance followed by Tukey's multiple comparison test. A p value of less than 0.05 was considered significant.
그 결과, 대조군(비이클(PBS))에 비하여 GI-106 투여군의 종양 성장이 저해되었다(도 8 내지 도 11). 종양 성장 억제율을 살펴보면, PBS 처리군의 종양성장 억제율이 30% 이상인 마우스는 1마리였으며, 50% 이상 및 80% 이상인 마우스는 없었다. GI-106 투여군의 경우, 종양성장 억제율이 30% 이상인 마우스가 5마리, 50% 이상인 마우스가 3마리, 80% 이상 마우스가 2마리였다(도 12).As a result, tumor growth in the GI-106 administered group was inhibited compared to the control group (vehicle (PBS)) (FIGS. 8 to 11). Looking at the tumor growth inhibition rate, there was only one mouse in the PBS-treated group with a tumor growth inhibition rate of more than 30%, and there were no mice with tumor growth inhibition rates of more than 50% or more than 80%. In the GI-106 administration group, there were 5 mice with a tumor growth inhibition rate of more than 30%, 3 mice with a tumor growth inhibition rate of 50% or more, and 2 mice with a tumor growth inhibition rate of 80% or more (Figure 12).
실시예 5. 마우스 유래 유방암세포 식립 마우스에서 GI-106B7NH06Kv3 투여에 의한 항암 효과 확인Example 5. Confirmation of anti-cancer effect by GI-106B7NH06Kv3 administration in mouse-derived breast cancer cell implantation mice
본 실험은 BALB/c마우스 유래의 유방암세포주(mouse breast cancer cells)인 EMT-6 세포를 이식한 BALB/c 마우스 종양 모델에서 시험물질 GI-106B7NH06Kv3을 투여한 후 종양의 성장 억제효과를 평가한 것이다.This experiment evaluated the tumor growth inhibition effect after administering the test substance GI-106B7NH06Kv3 in a BALB/c mouse tumor model transplanted with EMT-6 cells, a mouse breast cancer cell line derived from BALB/c mice. .
암 세포주가 이식된 마우스 모델을 제작하기 위해, BALB/c 암컷 마우스(7주령, 오리엔트바이오)에 마우스 유래 유방암세포인 EMT-6 세포 부유액을 마리 당 2Х104 세포/40 μL씩 일회용 주사기(31G, cat. 328820, BD)를 이용하여 동물의 복부 좌측을 개복하여 포유류 지방체(mammalian fat pad) 내부에 투여하였다. To create a mouse model transplanted with a cancer cell line, a suspension of EMT-6 cells, a mouse-derived breast cancer cell, was administered to BALB/c female mice (7 weeks old, Orient Bio) at a rate of 2Х104 cells/40 μL per mouse using a disposable syringe (31G, cat. 328820, BD) was used to open the left side of the animal's abdomen and administer it into the mammalian fat pad.
구체적으로, EMT-6 세포는 오리엔트바이오로부터 구매하여 10% FBS(16000-044, Thermo Fisher Scientific) 및 1% 항생제/항진균제(15140122, Thermo Fisher Scientific)를 포함하는 RPMI1640 배지(A1049101, Thermo Fisher Scientific)에서 배양하였다. 배양한 EMT-6 세포는 트립신-EDTA(Cat. 25200-072, Thermo Fisher Scientific)을 이용하여 수확한 후 PBS(Cat. LB 001-04, Welgene Inc.)에 현탁하였다. 10마리 이식 분량으로 EMT-6 세포 부유액(2Х106 세포/0.4 mL)을 일회용 주사기(31G, cat. 328820, BD medical diabetes care)에 충진하여 동물의 복부 좌측을 개복하여 포유류 지방체 내부에 투여하였다. 세포 이식 후, 매일 1회 일반증상을 관찰하였다.Specifically, EMT-6 cells were purchased from Orient Bio and cultured in RPMI1640 medium (A1049101, Thermo Fisher Scientific) containing 10% FBS (16000-044, Thermo Fisher Scientific) and 1% antibiotic/antimycotic (15140122, Thermo Fisher Scientific). It was cultured in . Cultured EMT-6 cells were harvested using trypsin-EDTA (Cat. 25200-072, Thermo Fisher Scientific) and then suspended in PBS (Cat. LB 001-04, Welgene Inc.). EMT-6 cell suspension (2Х10 6 cells/0.4 mL) for transplantation of 10 animals was filled into a disposable syringe (31G, cat. 328820, BD medical diabetes care), opened on the left side of the animal's abdomen, and administered into the mammalian fat body. . After cell transplantation, general symptoms were observed once daily.
EMT-6 세포를 이식하고 약 15일 후에 동물의 건강상태에 이상이 없는 마우스에 대해 종양의 부피를 측정하여, 각 군의 평균이 70~72 mm3에 도달하도록 개체 20마리를 선별하였다. 개체 선정은 개체의 생리적 상태(호흡, 털, 행동, 꼬리, 자세, 체액, 식이, 형태변형, 대사 등), 체중 변화 및 종양 성장속도를 고려하여 선발하였다. 선별된 동물은 종양의 부피 및 체중을 기초로 하여 가능한 균등하도록 군당 10마리씩 분리하였다. 표 4와 같이 시험군을 구성하고 시험물질을 투여하였다.Approximately 15 days after transplantation of EMT-6 cells, the tumor volume was measured in mice with no abnormalities in the animal's health, and 20 individuals were selected so that the average of each group reached 70~72 mm 3 . Individuals were selected considering their physiological state (respiration, fur, behavior, tail, posture, body fluids, diet, shape deformation, metabolism, etc.), weight change, and tumor growth rate. The selected animals were separated into groups of 10 as evenly as possible based on tumor volume and body weight. As shown in Table 4, the test group was formed and the test substance was administered.
army 투여물질administered substance 투여경로Route of administration 투여 주기Dosing cycle 투여용량Dosage 개체수(n)Number of individuals (n)
G1G1 비이클(hIgG4)Vehicle (hIgG4) i.vi.v. QW(1회/1주)QW (1 time/1 week) 9 mg/kg9mg/kg 1010
G2G2 GI-106B7NH06Kv3GI-106B7NH06Kv3 i.v.i.v. QW(1회/1주)QW (1 time/1 week) 9 mg/kg9mg/kg 1010
시험물질 투여 후 15일의 시험기간 동안 매일 1회 외관, 행동 및 배설물 등의 일반증상을 관찰하여 개체별로 기록하였으며, 사망동물을 확인하였다. 종양의 부피 측정은 관찰기간 동안 주 3회, 캘리퍼스를 사용하여 종양의 장축과 단축을 측정하였고, 실시예 4의 수학식 I에 대입하여 종양의 부피와 종양 성장 저해율을 측정하였다. 각 개체의 투여 전 종양의 부피는 군 분리 시 측정된 값으로 설정하였으며, 항종양 효능은 대조군(비이클(hIgG4), G1)과 비교하여 평가하였다. 모든 통계 계산은 Prism 8.0(Graph Pad Software Inc.)을 사용하여 수행하였다. 종양 부피 측정의 비교는 비대응 t-검정(Unpaired t-test)를 통해 이루어졌다. 0.05 미만의 p 값은 유의미한 것으로 간주되었다.During the 15-day test period after administration of the test substance, general symptoms such as appearance, behavior, and excrement were observed and recorded for each individual once a day, and dead animals were identified. Tumor volume was measured three times a week during the observation period using calipers to measure the long and short axes of the tumor, and the tumor volume and tumor growth inhibition rate were measured by substituting equation I in Example 4. The tumor volume before administration of each subject was set to the value measured at the time of group separation, and the antitumor efficacy was evaluated by comparison with the control group (vehicle (hIgG4), G1). All statistical calculations were performed using Prism 8.0 (Graph Pad Software Inc.). Comparison of tumor volume measurements was done using unpaired t-test. A p value of less than 0.05 was considered significant.
그 결과, 대조군(비이클(hIgG4))에 비해 GI-106B7NH06Kv3 투여군의 종양 부피가 약 4배 정도 유의미하게 감소되는 것을 확인하였다(도 13).As a result, it was confirmed that the tumor volume of the GI-106B7NH06Kv3 administration group was significantly reduced by about 4 times compared to the control group (vehicle (hIgG4)) (FIG. 13).
또한, 종양 성장 억제율을 살펴보면, 대조군(비이클(hIgG4))에 비해 GI-106B7NH06Kv3 투여군의 종양성장이 억제된 것을 확인하였다. 구체적으로, 음성대조군에서 종양성장 억제율이 30% 이상인 마우스는 4마리였으며, 그 중에서 50% 이상의 종양성장 억제율을 보인 마우스는 2마리였고, 상기 2마리의 마우스 중 80% 이상의 종양성장 억제율을 보인 마우스는 1마리였다. In addition, looking at the tumor growth inhibition rate, it was confirmed that the tumor growth of the GI-106B7NH06Kv3 administration group was inhibited compared to the control group (vehicle (hIgG4)). Specifically, in the negative control group, there were 4 mice with a tumor growth inhibition rate of more than 30%, of which 2 mice showed a tumor growth inhibition rate of more than 50%, and among the two mice, one mouse showed a tumor growth inhibition rate of more than 80%. There was 1 animal.
반면, GI-106NH06Kv3 투여군에서 종양성장 억제율이 30% 이상인 마우스가 9마리였으며, 그 중에서 50% 이상의 종양성장 억제율을 보인 마우스는 7마리였고, 상기 7마리의 마우스 중 80% 이상의 종양성장 억제율을 보인 마우스는 6마리였다. (도 14).On the other hand, in the GI-106NH06Kv3 administration group, there were 9 mice with a tumor growth inhibition rate of more than 30%, and among them, 7 mice showed a tumor growth inhibition rate of more than 50%, and among the 7 mice, one showed a tumor growth inhibition rate of more than 80%. There were 6 mice. (Figure 14).
실시예 6. 마우스 유래 대장암세포 식립 마우스에서 GI-106B7NH06Kv3 투여에 의한 항암 효과 확인Example 6. Confirmation of anti-cancer effect by GI-106B7NH06Kv3 administration in mouse-derived colon cancer cell implantation mice
본 실험은 C57BL/6 마우스 유래의 대장암세포주(murine colon cancer cells)인 MC38 세포를 이식한 종양 모델에서 시험물질 GI-106B7NH06Kv3을 투여한 후 종양의 성장 억제효과를 평가한 것이다.This experiment evaluated the tumor growth inhibition effect after administering the test substance GI-106B7NH06Kv3 in a tumor model transplanted with MC38 cells, a murine colon cancer cell line derived from C57BL/6 mice.
암 세포주가 이식된 마우스 모델을 제작하기 위해, C57BL/6 암컷 마우스(6주령, 오리엔트바이오)에 마우스 유래 대장암세포인 MC38 세포 부유액을 5Х105 세포/50 μL씩 일회용 주사기(31G, cat. 328820, BD medical diabetes care)에 충진하여 동물의 우측 등 부위의 피하에 투여하였다. 세포 이식 후, 매일 1회 일반증상을 관찰하였다.To create a mouse model transplanted with a cancer cell line, a suspension of MC38 cells, mouse-derived colon cancer cells, was administered to C57BL/6 female mice (6 weeks old, Orient Bio) at 5Х10 5 cells/50 μL using a disposable syringe (31G, cat. 328820, It was filled with BD medical diabetes care and administered subcutaneously to the right back of the animal. After cell transplantation, general symptoms were observed once daily.
MC38 세포는 Applied Stem Cell Inc.로부터 구매하여 10% FBS(Gibco) 및 1% 항생제/항진균제(Gibco)를 포함하는 RPMI1640 배지(Gibco)에서 배양하였다. 배양한 MC38 세포는 트립신(Gibco)을 이용하여 수확한 후 PBS에 현탁하였다. 동종이식 마우스 종양 모델을 확립하기 위해 건강한 마우스에 MC38 세포 부유액(5Х105 세포/0.025 mL) 및 0.025 mL BD Matrigel matrix phenol red-free(cat. 356231, BD biosciences)를 혼합하여 조제한 용액을 일회용 주사기(31G, cat. 328820, BD medical diabetes care)에 충진하여 동물의 우측 등 부위의 피하에 마리 당 0.05 mL씩 투여하여 이식하였다. MC38 세포 이식 후, 생착 및 성장기간 동안 매일 1회 일반증상을 관찰하였다. MC38 cells were purchased from Applied Stem Cell Inc. and cultured in RPMI1640 medium (Gibco) containing 10% FBS (Gibco) and 1% antibiotic/antimycotic (Gibco). Cultured MC38 cells were harvested using trypsin (Gibco) and suspended in PBS. To establish an allograft mouse tumor model, a solution prepared by mixing MC38 cell suspension (5Х10 5 cells/0.025 mL) and 0.025 mL BD Matrigel matrix phenol red-free (cat. 356231, BD biosciences) was injected into healthy mice using a disposable syringe ( 31G, cat. 328820, BD medical diabetes care) and implanted by administering 0.05 mL per animal subcutaneously on the right back of the animal. After MC38 cell transplantation, general symptoms were observed once daily during the engraftment and growth period.
MC38 세포를 이식하고 약 8일 후에 동물의 건강상태에 이상이 없는 마우스에 대해 종양의 부피를 측정하여, 각 군의 평균이 60~120 mm3에 도달하도록 개체 40마리를 선별하였다. 개체 선정은 개체의 생리적 상태(호흡, 털, 행동, 꼬리, 자세, 체액, 식이, 형태변형, 대사 등), 체중 변화 및 종양 성장속도를 고려하여 선발하였다. 선별된 동물은 종양의 부피 및 체중을 기초로 하여 가능한 균등하도록 군당 10마리씩 분리하였다. 표 5와 같이 시험군을 구성하고 시험물질을 투여하였다.Approximately 8 days after transplantation of MC38 cells, the tumor volume was measured in mice with no abnormalities in the animal's health, and 40 individuals were selected so that the average of each group reached 60~120 mm 3 . Individuals were selected considering their physiological state (respiration, fur, behavior, tail, posture, body fluids, diet, shape deformation, metabolism, etc.), weight change, and tumor growth rate. The selected animals were separated into groups of 10 as evenly as possible based on tumor volume and body weight. As shown in Table 5, the test group was formed and the test substance was administered.
army 투여물질administered substance 투여경로Route of administration 투여 주기Dosing cycle 투여용량Dosage 개체수(n)Number of individuals (n)
G1G1 비이클(hIgG4)Vehicle (hIgG4) i.vi.v. Q2W(1회/2주)Q2W (1 time/2 weeks) 9 mg/kg9mg/kg 1010
G2G2 GI-106B7NH06Kv3GI-106B7NH06Kv3 i.v.i.v. Q2W(1회/2주)Q2W (1 time/2 weeks) 9 mg/kg9mg/kg 1010
G3G3 GI-106B7NH06K-CNGI-106B7NH06K-CN i.vi.v. Q2W(1회/2주)Q2W (1 time/2 weeks) 9 mg/kg9mg/kg 1010
G4G4 GI-106B7NH06Kv3-CN+Fc-IL2v3GI-106B7NH06Kv3-CN+Fc-IL2v3 i.vi.v. Q2W(1회/2주)Q2W (1 time/2 weeks) 9 mg/kg+3.9 mg/kg9 mg/kg+3.9 mg/kg 1010
시험물질 투여 후 24일의 시험기간 동안 매일 1회 외관, 행동 및 배설물 등의 일반증상을 관찰하여 개체별로 기록하였으며, 사망동물을 확인하였다. 종양의 부피 측정은 관찰기간 동안 주 3회, 캘리퍼스를 사용하여 종양의 장축과 단축을 측정하였고, 실시예 4의 수학식 I에 대입하여 종양의 부피(TV)와 종양 성장 억제율(TGI)을 측정하였다. 각 개체의 투여 전 종양의 부피는 군 분리 시 측정된 값으로 설정하였으며, 항종양 효능은 대조군(비이클(hIgG4), G1)과 비교하여 평가하였다. 모든 통계 계산은 Prism 8.0(Graph Pad Software Inc.)을 사용하여 수행하였다. 종양 부피 측정의 비교는 비대응 t-검정을 통해 이루어졌다. 0.05 미만의 p 값은 유의미한 것으로 간주되었다.그 결과, 대조군(비이클(hIgG4))에 비해 GI-106B7NH06Kv3 투여군의 종양 부피가 약 3배 정도 유의미하게 감소되는 것을 확인하였다. 또한, GI-106NH06K-CN 투여군과 GI-106NH06K-CN 및 Fc-IL2v3 병용 투여군에 비해서도 GI-106B7NH06Kv3 투여군의 종양 부피가 약 2.5배 정도 유의미하게 감소되는 것을 확인하였다(도 15).During the 24-day test period after administration of the test substance, general symptoms such as appearance, behavior, and excrement were observed and recorded for each individual once a day, and dead animals were identified. Tumor volume was measured three times a week during the observation period, using calipers to measure the long and short axes of the tumor, and the tumor volume (TV) and tumor growth inhibition rate (TGI) were measured by substituting Equation I in Example 4. did. The tumor volume before administration of each subject was set to the value measured at the time of group separation, and the antitumor efficacy was evaluated by comparison with the control group (vehicle (hIgG4), G1). All statistical calculations were performed using Prism 8.0 (Graph Pad Software Inc.). Comparison of tumor volume measurements was done using unpaired t-test. A p value of less than 0.05 was considered significant. As a result, it was confirmed that the tumor volume of the GI-106B7NH06Kv3 administration group was significantly reduced by about 3 times compared to the control group (vehicle (hIgG4)). In addition, it was confirmed that the tumor volume of the GI-106B7NH06Kv3 administration group was significantly reduced by about 2.5 times compared to the GI-106NH06K-CN administration group and the GI-106NH06K-CN and Fc-IL2v3 combination administration group (FIG. 15).
또한, 종양 성장 억제율을 살펴보면, 대조군(비이클(hIgG4))에서 종양성장 억제율이 30% 이상인 마우스는 없었다. GI-106NH06K-CN 투여군에서 종양성장 억제율이 30% 이상인 마우스가 3마리였으며, 그 중에서 50% 이상의 종양성장 억제율을 보인 마우스는 2마리였고, 상기 2마리의 마우스 중 80% 이상의 종양성장 억제율을 보인 마우스는 1마리뿐이었다. 또한, GI-106NH06K-CN 및 Fc-IL2v3 투여군에서는 종양성장 억제율이 30% 이상인 마우스는 없었다. 반면, GI-106B7NH06Kv3 투여군에서는 모두 30% 이상의 종양성장 억제율을 보였으며, 그 중 50% 이상의 종양성장 억제율을 보인 마우스가 6마리였고, 상기 6마리 마우스 중 80% 이상의 종양성장 억제율을 보인 마우스는 3마리였다(도 16).Additionally, looking at the tumor growth inhibition rate, there were no mice in the control group (vehicle (hIgG4)) with a tumor growth inhibition rate of more than 30%. In the GI-106NH06K-CN administration group, there were 3 mice with a tumor growth inhibition rate of more than 30%, and among them, 2 mice showed a tumor growth inhibition rate of more than 50%, and among the two mice, one showed a tumor growth inhibition rate of more than 80%. There was only one mouse. Additionally, in the GI-106NH06K-CN and Fc-IL2v3 administration groups, there were no mice with a tumor growth inhibition rate of more than 30%. On the other hand, all groups administered GI-106B7NH06Kv3 showed a tumor growth inhibition rate of more than 30%, of which 6 mice showed a tumor growth inhibition rate of more than 50%, and among the 6 mice, 3 mice showed a tumor growth inhibition rate of more than 80%. It was Marie (Figure 16).
나아가, 각 군의 마우스의 생존율을 확인한 결과, 대조군(비이클)의 경우 15일차에 7마리가 사망하였고, 25일차까지 3마리의 마우스만이 생존하여 30%의 생존율을 나타내었다. GI-106NH06K-CN 투여군의 경우에서도 15일차에 2마리가 사망하기 시작하여 18일, 20일 및 22일차에 각각 1마리씩 더 사망하였고, 25일차까지 5마리의 마우스가 생존하여 50%의 생존율을 나타내었다. GI-106NH06K-CN 및 Fc-IL2v3 투여군의 경우 6일차부터 마우스가 사망하기 시작하여 22일차에 마우스가 모두 사망하였다. 반면, GI-106B7NH06Kv3 투여군의 경우 25일차까지 마우스가 모두 생존하여 100%의 생존율을 나타내었다(도 17).Furthermore, as a result of checking the survival rate of mice in each group, in the control group (vehicle), 7 mice died on the 15th day, and only 3 mice survived until the 25th day, showing a survival rate of 30%. In the case of the GI-106NH06K-CN administration group, 2 mice began to die on the 15th day, and 1 more mouse each died on the 18th, 20th, and 22nd days, and 5 mice survived until the 25th day, resulting in a 50% survival rate. indicated. In the case of the GI-106NH06K-CN and Fc-IL2v3 administration group, mice began to die from day 6 and all mice died on day 22. On the other hand, in the GI-106B7NH06Kv3 administration group, all mice survived until day 25, showing a survival rate of 100% (Figure 17).

Claims (19)

  1. TIGIT에 특이적으로 결합하는 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체.An antibody or fragment thereof that specifically binds to TIGIT; and a bispecific antibody containing the IL-2 protein.
  2. 제1항에 있어서,According to paragraph 1,
    상기 TIGIT에 특이적으로 결합하는 항체 또는 이의 단편은Antibodies or fragments thereof that specifically bind to the TIGIT
    서열번호 9의 아미노산 서열을 포함하는 HCDR1, 서열번호 10의 아미노산 서열을 포함하는 HCDR2, 및 서열번호 11의 아미노산 서열을 포함하는 HCDR3으로 이루어진 중쇄 가변 영역; 및 A heavy chain variable region consisting of HCDR1 comprising the amino acid sequence of SEQ ID NO: 9, HCDR2 comprising the amino acid sequence of SEQ ID NO: 10, and HCDR3 comprising the amino acid sequence of SEQ ID NO: 11; and
    서열번호 12의 아미노산 서열을 포함하는 LCDR1, 서열번호 13의 아미노산 서열을 포함하는 LCDR2, 및 서열번호 14의 아미노산 서열을 포함하는 LCDR3으로 이루어진 경쇄 가변 영역을 포함하는 것인, 이중 특이 항체.A bispecific antibody comprising a light chain variable region consisting of LCDR1 containing the amino acid sequence of SEQ ID NO: 12, LCDR2 containing the amino acid sequence of SEQ ID NO: 13, and LCDR3 containing the amino acid sequence of SEQ ID NO: 14.
  3. 제1항에 있어서,According to paragraph 1,
    상기 TIGIT에 특이적으로 결합하는 항체 또는 이의 단편은 서열번호 2의 아미노산 서열을 포함하는 중쇄 가변 영역 및 서열번호 7의 아미노산 서열을 포함하는 경쇄 가변 영역으로 이루어진 것인, 이중 특이 항체.The antibody or fragment thereof that specifically binds to TIGIT is a bispecific antibody consisting of a heavy chain variable region including the amino acid sequence of SEQ ID NO: 2 and a light chain variable region including the amino acid sequence of SEQ ID NO: 7.
  4. 제1항에 있어서,According to paragraph 1,
    상기 IL-2 단백질은 IL-2 변이체인 것인, 이중 특이 항체.A bispecific antibody, wherein the IL-2 protein is an IL-2 variant.
  5. 제4항에 있어서,According to paragraph 4,
    상기 IL-2 변이체는 서열번호 17의 아미노산 서열에서 38번째, 42번째 및 61번째 위치의 아미노산이 치환된 것인, 이중 특이 항체.The IL-2 variant is a bispecific antibody in which amino acids at positions 38, 42, and 61 in the amino acid sequence of SEQ ID NO: 17 are substituted.
  6. 제5항에 있어서,According to clause 5,
    상기 IL-2 변이체는 서열번호 17의 아미노산 서열에서 R38A, F42A 및 E61R 치환이 일어난 것인, 이중 특이 항체.The IL-2 variant is a bispecific antibody in which R38A, F42A and E61R substitutions occurred in the amino acid sequence of SEQ ID NO: 17.
  7. 제1항에 있어서, According to paragraph 1,
    상기 TIGIT에 특이적으로 결합하는 항체 또는 이의 단편; 및 IL-2 단백질은 링커에 의해 결합된 것인, 이중 특이 항체. An antibody or fragment thereof that specifically binds to the TIGIT; and the IL-2 protein is linked by a linker.
  8. 제1항에 있어서,According to paragraph 1,
    상기 이중 특이 항체는 하기 구조식 (I) 및 (II)를 포함하는 것인, 이중 특이 항체:The bispecific antibody comprises the following structural formulas (I) and (II):
    N'-X-[링커(1)]o-Fc 영역 단편 또는 이의 변이체-[링커(2)]p-Y-C' (I) 및N'-X-[Linker (1)]o-Fc region fragment or variant thereof-[Linker (2)]p-Y-C' (I) and
    N'-X'-C' (II)N'-X'-C' (II)
    이때, 상기 구조식 (I) 및 (II)에 있어서,At this time, in the structural formulas (I) and (II),
    상기 N'은 N 말단이고,Wherein N' is the N terminus,
    상기 C'는 C 말단이며,Wherein C' is the C terminus,
    상기 X는 항-TIGIT 항체의 중쇄의 항원 결합 부위로 가변 영역(VH) 및 CH1 영역을 포함하며,The X is the antigen binding site of the heavy chain of the anti-TIGIT antibody and includes a variable region (VH) and a CH1 region,
    상기 X'는 항-TIGIT 항체의 경쇄 항원 결합 부위로 가변 영역(VL) 및 불변 영역(CL)을 포함하고, The X' is the light chain antigen binding site of the anti-TIGIT antibody and includes a variable region (VL) and a constant region (CL),
    상기 Y는 IL-2 단백질 또는 이의 변이체이며,Y is IL-2 protein or a variant thereof,
    상기 링커(1) 및 링커(2)는 펩타이드 링커이고,The linker (1) and linker (2) are peptide linkers,
    상기 o 및 p는 각각 독립적으로, O 또는 1이다.The o and p are each independently O or 1.
  9. 제1항에 있어서,According to paragraph 1,
    상기 이중 특이 항체는 하기 구조식 (III) 및 (IV)를 포함하는 것인, 이중 특이 항체:The bispecific antibody comprises the following structural formulas (III) and (IV):
    N'-X-[링커(1)]o-Fc 영역 단편 또는 이의 변이체-C' (III) 및N'-X-[Linker (1)]o-Fc region fragment or variant thereof-C' (III) and
    N'-X'-[링커(3)]q-Y-C' (IV)N'-X'-[Linker (3)]q-Y-C' (IV)
    이때, 상기 구조식 (III) 및 (IV)에 있어서,At this time, in the structural formulas (III) and (IV),
    상기 N'은 N 말단이고,Wherein N' is the N terminus,
    상기 C'는 C 말단이며,Wherein C' is the C terminus,
    상기 X는 항-TIGIT 항체의 중쇄의 항원 결합 부위로 가변 영역(VH) 및 CH1 영역을 포함하며,The X is the antigen binding site of the heavy chain of the anti-TIGIT antibody and includes a variable region (VH) and a CH1 region,
    상기 X'는 항-TIGIT 항체의 경쇄 항원 결합 부위로 가변 영역(VL) 및 불변 영역(CL)을 포함하고, The X' is the light chain antigen binding site of the anti-TIGIT antibody and includes a variable region (VL) and a constant region (CL),
    상기 Y는 IL-2 단백질 또는 이의 변이체이며,Y is IL-2 protein or a variant thereof,
    상기 링커(1) 및 링커(3)는 펩타이드 링커이고,The linker (1) and linker (3) are peptide linkers,
    상기 o 및 q는 각각 독립적으로, O 또는 1이다.The o and q are each independently O or 1.
  10. 제8항 또는 제9항에 있어서,According to clause 8 or 9,
    상기 Fc는 서열번호 4 또는 서열번호 23의 아미노산 서열을 포함하는 것인, 이중 특이 항체.The Fc is a dual specific antibody comprising the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 23.
  11. 제1항 내지 제10항 중 어느 한 항의 이중 특이 항체를 암호화하는 폴리뉴클레오티드.A polynucleotide encoding the bispecific antibody of any one of claims 1 to 10.
  12. 제11항의 폴리뉴클레오티드를 포함하는 발현 벡터.An expression vector comprising the polynucleotide of claim 11.
  13. 제12항의 발현 벡터가 도입된 형질전환 세포.A transformed cell into which the expression vector of claim 12 has been introduced.
  14. i) 제13항의 형질전환 세포를 배양하는 단계; 및i) culturing the transformed cells of claim 13; and
    ii) 이중 특이 항체를 수득하는 단계;를 포함하는 항-TIGIT 항체 또는 이의 단편; 및 IL-2 단백질을 포함하는 이중 특이 항체의 제조 방법.ii) obtaining a bispecific antibody; an anti-TIGIT antibody or fragment thereof comprising; and a method for producing a bispecific antibody comprising IL-2 protein.
  15. 제1항의 이중 특이 항체를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer comprising the bispecific antibody of claim 1 as an active ingredient.
  16. 제15항에 있어서,According to clause 15,
    상기 암은 위암, 간암, 폐암, 대장암, 유방암, 전립선암, 난소암, 췌장암, 자궁경부암, 갑상선암, 후두암, 급성 골수성 백혈병, 뇌종양, 신경모세포종, 망막 모세포종, 두경부암, 침샘암 및 림프종으로 구성된 군에서 선택되는 어느 하나인, 암 예방 또는 치료용 약학 조성물.The cancer consists of stomach cancer, liver cancer, lung cancer, colon cancer, breast cancer, prostate cancer, ovarian cancer, pancreas cancer, cervical cancer, thyroid cancer, laryngeal cancer, acute myeloid leukemia, brain tumor, neuroblastoma, retinoblastoma, head and neck cancer, salivary gland cancer, and lymphoma. A pharmaceutical composition for preventing or treating cancer, which is selected from the group.
  17. 암을 예방 또는 치료하기 위한 제1항의 이중 특이 항체의 용도.Use of the bispecific antibody of claim 1 to prevent or treat cancer.
  18. 암의 예방 또는 치료용 약제를 제조하기 위한 제1항의 이중 특이 항체의 용도.Use of the bispecific antibody of claim 1 for manufacturing a drug for preventing or treating cancer.
  19. 제1항의 이중 특이 항체를 개체에 투여하는 단계를 포함하는 암의 예방 또는 치료 방법.A method for preventing or treating cancer comprising administering the bispecific antibody of claim 1 to a subject.
PCT/KR2023/007622 2022-06-03 2023-06-02 Bispecific antibody including anti-tigit antibody, and use thereof WO2023234743A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2022-0068303 2022-06-03
KR20220068303 2022-06-03

Publications (1)

Publication Number Publication Date
WO2023234743A1 true WO2023234743A1 (en) 2023-12-07

Family

ID=89025237

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/007622 WO2023234743A1 (en) 2022-06-03 2023-06-02 Bispecific antibody including anti-tigit antibody, and use thereof

Country Status (2)

Country Link
KR (1) KR20230168273A (en)
WO (1) WO2023234743A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018184965A1 (en) * 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Immunoconjugates of il-2 with an anti-pd-1 and tim-3 bispecific antibody
KR20190088057A (en) * 2016-11-30 2019-07-25 온코메드 파마슈티칼스, 인크. A method of treating cancer comprising TIGIT-binding agents
KR20190140756A (en) * 2018-06-12 2019-12-20 주식회사 노보셀바이오 A bispecific antibody binding to natural killer cells and an use thereof
KR20200032009A (en) * 2018-09-17 2020-03-25 (주)지아이이노베이션 Fusion protein comprising il-2 protein and cd80 protein and use thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190088057A (en) * 2016-11-30 2019-07-25 온코메드 파마슈티칼스, 인크. A method of treating cancer comprising TIGIT-binding agents
WO2018184965A1 (en) * 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Immunoconjugates of il-2 with an anti-pd-1 and tim-3 bispecific antibody
KR20190140756A (en) * 2018-06-12 2019-12-20 주식회사 노보셀바이오 A bispecific antibody binding to natural killer cells and an use thereof
KR20200032009A (en) * 2018-09-17 2020-03-25 (주)지아이이노베이션 Fusion protein comprising il-2 protein and cd80 protein and use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MU, S. ET AL.: "PD-L1/TIGIT bispecific antibody showed survival advantage in animal model", CLIN. TRANSL. MED, vol. 12, 6 May 2022 (2022-05-06), pages 1 - 5, XP093097176, DOI: 10.1002/ctm2.754 *

Also Published As

Publication number Publication date
KR20230168273A (en) 2023-12-13

Similar Documents

Publication Publication Date Title
WO2017074117A1 (en) Long-acting fgf21 fusion proteins and pharmaceutical composition comprising same
WO2012011752A2 (en) Novel long-acting glucagon conjugate and pharmaceutical composition comprising the same for the prevention and treatment of obesity
WO2014133324A1 (en) Novel insulin analog and use thereof
WO2021096275A1 (en) Fusion protein including modified interleukin-7 and tgf beta receptor ii and use thereof
WO2018169282A2 (en) Pharmaceutical composition containing atpif1 for treatment of diabetes
KR20140041533A (en) Therapeutic canine immunoglobulins and methods of using the same
WO2016114633A1 (en) Long-acting fgf21 fusion proteins and pharmaceutical composition comprising the same
ES2277947T3 (en) PROCEDURES FOR USING A HUMAN IL-17 AFIN POLYPEPTIDE TO TREAT DISEASES.
WO2022005174A1 (en) Fusion protein comprising anti-lag-3 antibody and il-2, and use thereof
WO2021153979A1 (en) Fusion protein comprising anti-taa antibody, anti-pd-l1 antibody, and il-2, and uses thereof
WO2023136518A1 (en) Fusion protein comprising cd80 extracellular domain and anti-lag3 antibody fragment and use thereof
WO2023234743A1 (en) Bispecific antibody including anti-tigit antibody, and use thereof
TW202116803A (en) Gipr antibody and glp-1 fusion protein thereof, and their pharmaceutical compositions and therapeutic applications
WO2021107603A2 (en) Long-acting gdf15 fusion protein and pharmaceutical composition comprising same
WO2020159191A1 (en) Composition comprising mls-stat3 for prevention or treatment of immune disease
WO2017188653A1 (en) Fusion protein comprising ccl3 variant and use thereof cross-reference to related applications
WO2023249425A1 (en) Fusion protein comprising anti-cd73 antibody and il-2, and use thereof
WO2023229303A1 (en) Antibody binding to c-terminus of igsf1 and use thereof
WO2023244089A1 (en) Antibody specifically binding to human dr3 and use thereof
WO2019088658A1 (en) Dual-targeting antibody targeting scf and galectin-1 and use thereof
WO2022245183A1 (en) Composition for preventing or treating non-alcoholic fatty liver disease or non-alcoholic steatohepatitis comprising growth differentiation factor-15 variant
EP4122953A1 (en) Fusion protein comprising il-2 protein and cd80 protein fragment or variant thereof, and uses thereof
WO2022019652A1 (en) Fusion protein including ige fc receptor and use thereof for treatment of canine allergic disease
WO2023022271A1 (en) Anti-igsf1 antibody and use thereof
WO2024076121A1 (en) Chimeric antigen receptor targeting cd5 and immune cells expressing same

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: 23816403

Country of ref document: EP

Kind code of ref document: A1