WO2024085606A1 - Bispecific antibody including first antigen-binding site that specifically binds to human angiopoietin-2, and use thereof - Google Patents

Bispecific antibody including first antigen-binding site that specifically binds to human angiopoietin-2, and use thereof Download PDF

Info

Publication number
WO2024085606A1
WO2024085606A1 PCT/KR2023/016063 KR2023016063W WO2024085606A1 WO 2024085606 A1 WO2024085606 A1 WO 2024085606A1 KR 2023016063 W KR2023016063 W KR 2023016063W WO 2024085606 A1 WO2024085606 A1 WO 2024085606A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
vegf
binding site
specifically binds
Prior art date
Application number
PCT/KR2023/016063
Other languages
French (fr)
Korean (ko)
Inventor
김용인
오인재
김희곤
김예진
Original Assignee
(주)니오테스바이오
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
Priority claimed from KR1020230136843A external-priority patent/KR20240056422A/en
Application filed by (주)니오테스바이오 filed Critical (주)니오테스바이오
Publication of WO2024085606A1 publication Critical patent/WO2024085606A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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

Definitions

  • the present invention relates to a bispecific antibody comprising a first antigen binding site that specifically binds to human angiopoietin-2 and its applications.
  • Angiopoietin-2 (hereinafter also referred to as 'ANG-2') binds to the receptor Tie2 present on vascular endothelial cells, but acts as an antagonistic ligand, producing angiophore, an agonist of Tie2. It acts to inhibit signal transduction by Tie2 by competing with Angiopoietin-1 (Ang1) for binding to Tie2. Due to this mechanism of action, when vascular endothelial growth factor (Vascular Epidermal Growth Factor, hereinafter referred to as 'VEGF') is overexpressed or in a state of inflammation, vascular endothelial cells are activated and vascular permeability increases.
  • vascular endothelial growth factor Vascular Epidermal Growth Factor
  • Ang1 While it induces stabilization of endothelial cells and reduces vascular permeability, increased Ang2 in activated vascular endothelial cells competes with Ang1, thereby inhibiting stabilization of vascular endothelial cells by Ang1. Therefore, Ang2 inhibits Ang1-Tie2 binding and signaling through Ang1-Tie2, which maintains the stability of vascular endothelial cells in the presence of VEGF, and ultimately promotes angiogenesis in blood vessels, resulting in increased angiogenesis, destabilization of blood vessels, and vascular permeability.
  • Ang2 has been reported to have agonist properties that induce the activity of the Tie2 receptor in some specific situations, including the formation and maintenance of lymphatic vessels. Therefore, it functions as an antagonist and a weak agonist. It is believed to have all of the following functions and perform various functions depending on the situation.
  • neovascularization Since the process of neovascularization is an essential factor in the growth of cancer, there have been attempts to prevent further growth of cancer by inhibiting neovascularization by inhibiting the Tie2-dependent function of Ang2 as described above. However, in most cases, these are It is known to have the function of inhibiting binding and preventing its role as an antagonist. In addition to antibodies that interfere with the binding of Ang2 to Tie2, recombinant proteins or antibodies that directly bind to the Tie2 receptor and induce phosphorylation and activation have been reported.
  • Ranibizumab brand name Lucentis®
  • bevacizumab Avastin
  • this antibody has been affinity matured to provide stronger binding to VEGF-A (International Patent Application Publication No. WO 98/45331).
  • VEGF-A blockade may be associated with some systemic toxicity, it is known that ranibizumab is losing its Fc portion to reduce serum half-life and consequently systemic toxicity.
  • the object of the present invention is a bispecific antibody comprising a site that specifically binds to human ANG-2 and a site that specifically binds to human VEGF, TGF-beta, TIM-3 ligand or PEDF-R (PNPLA2, ATGL). It provides antibodies.
  • Another object of the present invention is to provide a pharmaceutical composition containing the bispecific antibody as an active ingredient.
  • the present invention provides a first antigen binding site that specifically binds to human angiopoietin (ANG)-2, human vascular endothelial growth factor (VEGF), transforming growth factor, Hereinafter referred to as 'TGF')-beta, the ligand of T-cell immunoglobulin and mucin-domain containing protein (T-cell immunoglobulin and mucin-domain containing, hereinafter referred to as 'TIM')-3, and pigment epithelium-derived factor receptor.
  • a bispecific antibody comprising a second antigen binding site that specifically binds to one antigen selected from the group consisting of (pigment epithelium-derived factor receptor, hereinafter referred to as 'PEDF-R'),
  • the first antigen binding site that specifically binds to the ANG-2 is the CDRH1 region of SEQ ID NO: 1 or 7 or 13, the CDRH2 region of SEQ ID NO: 2 or 8 or 14, and the CDRH3 of SEQ ID NO: 3 or 9 or 15 comprising a region within a heavy chain variable domain, a CDRL1 region of SEQ ID NO: 4 or 10 or 16, a CDRL2 region of SEQ ID NO: 5 or 11 or 17 and a CDRL3 region of SEQ ID NO: 6 or 12 or 18 within a light chain variable domain;
  • the second antigen binding site specifically binding to VEGF comprises a VEGFR1 D2 domain
  • the second antigen binding site specifically binding to TGF-beta comprises TGF-beta RII ECD
  • the second antigen binding site that specifically binds to the ligand of TIM-3 includes TIM-3 ECD
  • the second antigen binding site that specifically binds to the PEDF-R includes a 44-mer.
  • the antibody provides a bispecific antibody that induces Tie2 activation.
  • the VEGFR1 D2 domain is the VEGFR1 D2 domain of SEQ ID NO: 21
  • the TGF-beta RII ECD is the TGF-beta RII ECD of SEQ ID NO: 24
  • the TIM-3 ECD is the TIM of SEQ ID NO: 27 -3 ECD and the 44-mer PEDF preferably include the amino acid sequence of SEQ ID NO: 30, but are not limited thereto.
  • the VEGFR1 D2 domain, the TGF-beta RII ECD, the TIM-3 ECD, and the 44-mer PEDF are preferably connected to an immunoglobulin by a linker, and the linker It is more preferable that the linker is SEQ ID NO: 20, SEQ ID NO: 23, SEQ ID NO: 26, or SEQ ID NO: 29, respectively, but is not limited thereto.
  • the present invention provides a pharmaceutical composition for the treatment of cancer containing the antibody according to the present invention.
  • the present invention provides a pharmaceutical composition for the treatment of vascular diseases containing the antibody according to the present invention.
  • the present invention also provides a nucleic acid encoding the bispecific antibody according to the present invention.
  • the present invention also provides a method for producing a bispecific antibody according to the present invention, comprising expressing a nucleic acid encoding the bispecific antibody in a prokaryotic or eukaryotic host cell and recovering the bispecific antibody from the cell or cell culture supernatant.
  • a method for producing a bispecific antibody comprising expressing a nucleic acid encoding the bispecific antibody in a prokaryotic or eukaryotic host cell and recovering the bispecific antibody from the cell or cell culture supernatant.
  • a “bispecific antibody” according to the invention is an antibody with two different antigen-binding specificities. If an antibody has more than one specificity, the epitope recognized may be associated with a single antigen or more than one antigen.
  • the antibodies of the invention are specific for two different antigens, ANG-2 as the first antigen and VEGF, TGF-beta, the ligand of TIM-3 or PEDF-R as the second antigen.
  • Bispecific antibodies of the invention include, for example, multivalent single chain antibodies, diabodies and triabodies, as well as additional antigen binding sites (e.g., single chain Fv, It includes antibodies having the constant domain structure of a full-length antibody to which a VH domain and/or a VL domain, Fab, or (Fab)2) are linked.
  • Antibodies can be full length from a single species, chimeric or humanized. For antibodies with two or more antigen binding sites, some of the binding sites may be identical, as long as the protein has binding sites for two different antigens. That is, the first binding site is specific for ANG-2, while the second binding site is specific for VEGF, TGF-beta, TIM-3 ligand, and PEDF-R, and vice versa.
  • the antibodies of the invention further comprise immunoglobulin constant regions of one or more immunoglobulin classes.
  • Immunoglobulin classes include IgG, IgM, IgA, IgD and IgE isotypes and, in the case of IgG and IgA, their subtypes.
  • the antibodies of the invention have the constant domain structure of an IgG type antibody, but have four antigen binding sites.
  • chimeric antibody refers to an antibody comprising a variable region, i.e., a binding region, from one source or species and at least a portion of a constant region from a different source or species, typically produced by recombinant DNA technology. Chimeric antibodies comprising murine variable regions and human constant regions are preferred. Other preferred forms of “chimeric antibodies” encompassed by the present invention are those wherein the constant portions are altered or varied from the constant portions of the original antibody, thereby producing properties according to the present invention, particularly with regard to C1q binding and/or Fc receptor (FcR) binding. will be. Such chimeric antibodies are also referred to as “class-switched antibodies”.
  • Chimeric antibodies are the product of expressed immunoglobulin genes comprising a DNA segment encoding an immunoglobulin variable region and a DNA segment encoding an immunoglobulin constant region.
  • Methods for producing chimeric antibodies include conventional recombinant DNA and gene transfection techniques well known in the art. For example, Morrison, S.L., et al., Proc. Natl. Acad. Sci. USA 81 (1984) 6851-6855; See US 5,202,238 and US 5,204,244.
  • humanized antibody refers to an antibody whose framework or “complementarity determining regions” (CDRs) have been altered to include the CDRs of an immunoglobulin of different specificity compared to the parent immunoglobulin.
  • murine CDRs are grafted into the framework region of a human antibody to produce a “humanized antibody.”
  • CDRs complementarity determining regions
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • Human antibodies are well known in the art (van Dijk, M.A., and van de Winkel, J.G., Curr. Opin. Chem. Biol. 5 (2001) 368-374).
  • Human antibodies can also be produced in transgenic animals (e.g., mice) that can produce the full repertoire or selection of human antibodies upon immunization in the absence of endogenous immunoglobulin production. Transfer of a human germline immunoglobulin gene sequence into such germline mutant mice will result in the production of human antibodies upon challenge (e.g. Jakobovits, A., et al., Proc. Natl. Acad.
  • Boerner, P., et al.'s techniques are also available (Cole, A., et al., Monoclonal Antibodies and Cancer Therapy, Liss, A.L., p. 77 (1985); and Boerner, P., et al. al., J. Immunol. 147 (1991) 86-95).
  • the term “recombinant human antibody” refers to any human antibody that is manufactured, expressed, produced or isolated by recombinant methods, such as from a host cell such as NS0 or CHO cells or from an animal transgenic for human immunoglobulin genes (e.g. It is intended to include antibodies isolated from (e.g., mouse), or antibodies expressed using recombinant expression vectors transfected into host cells.
  • the recombinant human antibody has variable and constant regions in rearranged form.
  • CDR complementarity determining region
  • variable domain refers to each of the pairs of light and heavy chains that are directly involved in antibody binding to antigen.
  • the domains of the variable human light and heavy chains have the same general structure, with each domain consisting of four framework (FR) regions of broadly conserved sequence, linked by three "hypervariable regions” (or complementarity determining regions, CDRs). Includes.
  • the framework region has a ⁇ -sheet conformation, and the CDRs can form loops connecting the ⁇ -sheet structures.
  • the CDRs within each chain maintain their three-dimensional structure by framework regions and, together with CDRs from other chains, form an antigen-binding site.
  • hypervariable region or “antigen-binding site of an antibody” refers to the amino acid residues of an antibody that are responsible for antigen-binding. Hypervariable regions include amino acid residues from the “complementarity determining region” or “CDR”.
  • the “framework” or “FR” region is the variable domain region excluding the hypervariable region residues as defined herein. Therefore, the light and heavy chains of the antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • the CDRs on each chain are separated by the framework amino acids.
  • CDR3 of the heavy chain is the region that most contributes to antigen binding.
  • CDR and FR regions are determined according to the standard definitions of Kabat et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991).
  • Bispecific antibodies according to the invention also include said antibodies with “conservative sequence alterations” (this means “variants” of bispecific antibodies).
  • amino acids with uncharged polar side chains e.g. glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • amino acids with nonpolar side chains e.g. alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine
  • amino acids with beta-branched side chains e.g. threonine, valine, isoleucine
  • amino acids with aromatic side chains e.g. tyrosine, phenylalanine, tryptophan, histidine.
  • a predicted non-essential amino acid residue in a bispecific antibody may be replaced with another amino acid residue, preferably from the same side chain family.
  • a "variant" bispecific antibody is one whose amino acid sequence differs from the "parent" bispecific antibody amino acid sequence by at most 10, preferably about 2 to about 5, additions, deletions, and deletions within one or more variable or constant regions of the parent antibody. /or refer to a different molecule by substitution. Amino acid substitutions are as described in Riechmann, L., et al., Nature 332 (1988) 323-327 and Queen, C, et al., Proc. Natl. Acad. Sci. USA 86 (1989) 10029-10033 may be performed by mutagenesis based on molecular modeling. “Variant” bispecific antibodies according to the invention also include bispecific antibody forms in which the linker (if present) therein is modified or replaced by another linker.
  • binding refers to the binding of an antibody to an epitope of an antigen (human VEGF, ANG-2, TGF-beta, TIM-3 ligand or PEDF-R) in an in vitro assay. Mention.
  • epitope includes any polypeptide determinant capable of specifically binding to an antibody.
  • epitope determinants include amino acids, sugar side chains, chemically active surface groups of molecules such as phosphoryl or sulfonyl, and, in certain embodiments, may have specific three-dimensional structural properties and/or specific charge properties. there is.
  • An epitope is a region of an antigen that is bound by an antibody.
  • linker refers to a peptide having an amino acid sequence, preferably of synthetic origin. Typically, the peptides link a) VH-CH1 to VL-CL, b) VL-CL to VH-CH1, c) VH-CL to VL-CH1, or d) VL-CH1 to VH-CL.
  • the linker according to the present invention is used to connect human immunoglobulin G4 and human VEGFR1 domain 2, TGF-beta RII ECD, TIM-3 ECD, and the PEDF sequence of the 44-mer.
  • pharmaceutical carrier includes any and all physiologically compatible solvents, dispersion media, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like.
  • the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g. by injection or infusion).
  • composition of the present invention can be administered by various methods known in the art. As will be appreciated by those skilled in the art, the route and/or mode of administration will vary depending on the desired results. To administer an antibody of the invention by a particular route of administration, it may be necessary to coat the antibody with or co-administer the antibody with a material that will prevent its inactivation.
  • the antibody can be administered to a subject in a suitable carrier, such as liposomes or a diluent.
  • suitable carrier such as liposomes or a diluent.
  • Pharmaceutically acceptable diluents include saline and aqueous buffered solutions.
  • Pharmaceutical carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. The use of such media or agents for pharmaceutically active ingredients is known in the art.
  • parenteral administration and “administered parenterally” refer to any mode of administration other than enteral and topical administration, typically by injection, including intravenous, intramuscular, intraarterial, intrathecal, and intraarticular. Including, without limitation, intraorbital, intracardiac, intradermal, intraperitoneal, intratracheal, subcutaneous, intraepithelial, intraarticular, subcapsular, subarachnoid, intrathecal, epidural, and intrasternal injections and infusions.
  • cancer refers to proliferative diseases such as lymphoma, lymphocytic leukemia, lung cancer, non-small cell lung (NSCL) cancer, bronchioloalveolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, skin and intraocular melanoma, and uterine cancer.
  • NSCL non-small cell lung
  • bronchioloalveolar cell lung cancer bone cancer
  • pancreatic cancer skin cancer, head and neck cancer
  • skin and intraocular melanoma and uterine cancer.
  • ovarian cancer rectal cancer, anal area cancer, stomach cancer, gastrointestinal cancer, colon cancer, breast cancer, uterine cancer, fallopian tube carcinoma, endometrial carcinoma, cervical cancer, vaginal carcinoma, vulvar carcinoma, Hodgkin's disease, esophageal cancer, small intestine cancer, endocrine cancer, Thyroid cancer, parathyroid cancer, adrenal cancer, soft sarcoma, urethral cancer, penile cancer, prostate cancer, bladder cancer, kidney or urethral cancer, renal cell cancer, renal pelvis cancer, mesothelioma, hepatocellular cancer, biliary tract cancer, and tumors of the central nervous system (CNS) , spine tumor, brainstem tumor, glioblastoma multiforme, astrocytoma, schwannoma, ependymoma, medulloblastoma, meningioma, squamous cell carcinoma, pituitary adenoma and Ewing'
  • Another aspect of the invention is the bispecific antibody or said pharmaceutical composition according to the invention as an anti-angiogenic agent.
  • the anti-angiogenic substances can be used for the treatment of cancer, especially solid tumors, and other vascular diseases.
  • One embodiment of the invention is a bispecific antibody according to the invention for the treatment of vascular diseases.
  • Another aspect of the present invention is the pharmaceutical composition for treating vascular diseases.
  • Another aspect of the invention is the use of the antibody according to the invention for the manufacture of a medicament for the treatment of vascular diseases.
  • Another aspect of the invention is a method of treating a patient suffering from vascular disease by administering an antibody according to the invention to the patient in need of treatment.
  • vascular disease includes cancer, inflammatory disease, atherosclerosis, ischemia, trauma, sepsis, COPD, asthma, diabetes, AMD, retinopathy, stroke, adiposity, acute lung injury, hemorrhage, e.g. cytokine-induced Vascular effusion, allergy, Graves' disease, Hashimoto's autoimmune thyroiditis, idiopathic thrombocytopenic purpura, giant cell arteritis, rheumatoid arthritis, systemic lupus erythematosus (SLE), lupus nephritis, Crohn's disease, multiple sclerosis, ulcerative colitis, especially solid tumors.
  • cytokine-induced Vascular effusion allergy
  • Graves' disease Hashimoto's autoimmune thyroiditis, idiopathic thrombocytopenic purpura, giant cell arteritis, rheumatoid arthritis, systemic lupus erythematosus (S
  • intraocular neovascular syndromes such as proliferative retinopathy or age-related macular degeneration (AMD), rheumatoid arthritis and psoriasis
  • AMD age-related macular degeneration
  • rheumatoid arthritis rheumatoid arthritis
  • psoriasis psoriasis
  • the composition may also include adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the presence of microorganisms can be ensured by the above sterilization procedures and by the inclusion of various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid, etc. It may also be desirable to include isotonic substances such as sugars, sodium chloride, etc. into the composition. Additionally, sustained absorption of injectable pharmaceutical forms can be obtained by the inclusion of substances that delay absorption, such as aluminum monostearate and gelatin.
  • adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the presence of microorganisms can be ensured by the above sterilization procedures and by the inclusion of various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid, etc. It may also be desirable to include isotonic substances such as sugars
  • the antibodies of the invention which can be used in suitable hydrated forms and/or pharmaceutical compositions of the invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those skilled in the art.
  • the actual dosage levels of the active ingredient in the pharmaceutical compositions of the invention may vary to obtain an amount of active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, while not being toxic to the patient.
  • the dosage level selected will depend on the activity of the particular composition of the invention employed, the route of administration, the time of administration, the rate of excretion of the particular antibody to be employed, the duration of treatment, the other drugs, compounds and/or substances to be combined with the particular composition to be employed, the treatment. It will vary depending on various pharmacokinetic factors, including factors well known in the medical field, such as the patient's age, gender, weight, condition, general health, and past medical history.
  • compositions of the present invention must be fluid and sterile to the extent that the compositions can be delivered by syringe.
  • the carrier is preferably isotonic buffered saline. Adequate fluidity can be maintained, for example, by the use of coatings such as lecithin, by maintenance of the required particle size in case of dispersion, and by the use of surfactants.
  • isotonic substances such as sugars, polyhydric alcohols such as mannitol or sorbitol, and sodium chloride in the composition.
  • the present invention proposes an antibody that promotes downstream signaling by inhibiting Ang2, VEGF, TGF-beta, and TIM-3 ligands or activating PEDF-R while simultaneously activating the Tie2 receptor, thereby inhibiting neovascularization and improving vascular permeability. Suggests a new way to reduce it.
  • the antibody proposed in the present invention is expected to have applicability in the diagnosis and treatment of cancer, diseases related to abnormal angiogenesis, and/or diseases caused by increased vascular permeability.
  • Figure 1 is a schematic diagram of a bispecific antibody comprising a first antigen binding site that specifically binds to human angiopoietin-2 developed in the present invention.
  • Figure 2 shows the SDS-PAGE results of purified anti-Ang2/VEGF antibody.
  • Figures 3 and 4 show the results of SDS-PAGE purified anti-Ang2/TGF- ⁇ antibody ( Figure 3) and the results of Western blot confirmation of the TGF- ⁇ binding region, the second antigen binding site ( Figure 4), respectively.
  • Figures 5 and 6 show ELISA results analyzing the binding affinity of anti-Ang2/VEGF antibodies (Figure 5: 2C8-D2, Figure 6: 1D3-D2) to human Ang2, respectively.
  • Figures 7 and 8 are ELISA results showing the binding ability of anti-Ang2/VEGF antibody (Figure 7, 2C8-D2, Figure 8, 1D3-D2) to human VEGF or mouse VEGF, respectively.
  • Figure 9 shows ELISA results showing that anti-Ang2/VEGF antibody can bind to VEGF and Ang2 simultaneously.
  • Figure 10 is a Western blot result showing that anti-Ang2/VEGF antibody and anti-Ang2/TGF- ⁇ antibody phosphorylate AKT and ERK 42/44 in HUVEC along with Ang2.
  • Figure 11 is a result showing that anti-Ang2/VEGF antibody binds to VEGF competitively with soluble VEGFR2.
  • Figure 12 shows the results of anti-Ang2/VEGF antibody inhibiting HUVEC proliferation caused by VEGF.
  • Figures 13 to 15 show the inhibition of neovascularization by anti-Ang2/VEGF antibodies in the mouse laser-induced CNV (wet AMD) model (Figure 13. representative FFA images & CTF, Figure 14. representative OCT images & CNV lesion) and improvement in vision, respectively. This is a result showing the effect ( Figure 15. ERG).
  • Example 1 Construction of a bispecific antibody comprising a first antigen binding site that specifically binds to human angiopoietin-2
  • the heavy chain of the anti-Ang2/VEGF antibody is a heavy chain CDR sequence of 1A9 or 2C8 or 1D3 (SEQ ID NOs: 1 to 3 or 7 to 9 or 13 to 15) and C- The human immunoglobulin G4 heavy chain sequence and linker sequence (SEQ ID NO: 20) and VEGFR1 D2 sequence (SEQ ID NO: 21), in which terminal lysine was replaced with alanine, were composed into one and synthesized in GenScript.
  • the light chain of the anti-Ang2/VEGF antibody used was the light chain CDR sequence (SEQ ID NO: 4 to 6 or 10 to 12 or 16 to 18) of 1A9 or 2C8 or 1D3 described in Korean Patent Application No. 10-2022-0028751. .
  • the heavy chain of the anti-Ang2/TGF- ⁇ antibody is the heavy chain CDR sequence of 1A9 or 2C8 or 1D3 (SEQ ID NOs: 1 to 3 or 7 to 9 or 13 to 15) described in Korean Patent Application No. 10-2022-0028751.
  • the human immunoglobulin G4 heavy chain sequence and linker sequence (SEQ ID NO: 23), in which lysine at the C-terminal was replaced with alanine, and the TGF- ⁇ extracellular domain (SEQ ID NO: 24) were constructed as one, and then synthesized in GenScript.
  • the light chain of the anti-Ang2/TGF- ⁇ antibody has the light chain CDR sequence (SEQ ID NO: 4 to 6 or 10 to 12 or 16 to 18) of 1A9 or 2C8 or 1D3 described in Korean Patent Application No. 10-2022-0028751. used.
  • the heavy chain of the anti-Ang2/TIM-3 ligand antibody is the heavy chain CDR sequence of 1A9 or 2C8 or 1D3 (SEQ ID NOs: 1 to 3 or 7 to 9 or 13 to 15) described in Korean Patent Application No. 10-2022-0028751.
  • the human immunoglobulin G4 heavy chain sequence and linker sequence SEQ ID NO: 26
  • lysine at the C-terminal was replaced with alanine
  • the human TIM-3 extracellular domain SEQ ID NO: 27
  • the light chain of the anti-Ang2/TIM-3 ligand antibody is the light chain CDR sequence of 1A9 or 2C8 or 1D3 (SEQ ID NOs: 4 to 6 or 10 to 12 or 16 to 18) described in Korean Patent Application No. 10-2022-0028751. was used.
  • the heavy chain of the anti-Ang2/PEDF-R antibody is the heavy chain CDR sequence of 1A9 or 2C8 or 1D3 (SEQ ID NOs: 1 to 3 or 7 to 9 or 13 to 15) described in Korean Patent Application No. 10-2022-0028751, and The human immunoglobulin G4 heavy chain sequence and linker sequence (SEQ ID NO: 29), in which lysine at the C-terminal was replaced with alanine, and the human PEDF 44-mer peptide (SEQ ID NO: 30) were combined into one, and then synthesized in GenScript.
  • the light chain of the anti-Ang2/PEDF-R antibody has the light chain CDR sequence (SEQ ID NO: 4 to 6 or 10 to 12 or 16 to 18) of 1A9 or 2C8 or 1D3 described in Korean Patent Application No. 10-2022-0028751. used.
  • Table 1 is the CDR sequence of mouse anti-Ang2 antibody 1A9
  • Table 2 shows the CDR sequence of anti-Ang2 antibody 2C8
  • Table 3 shows the CDR sequence of anti-Ang2 antibody 1D3
  • Table 4 shows the linker connecting human immunoglobulin G4 and the (129-229) sequence containing human VEGFR1 domain 2, and the (129-229) sequence containing human VEGFR1 domain 2,
  • Table 5 shows the linker connecting human immunoglobulin G4 and human TGF- ⁇ RII extracellular domain sequence and human TGF- ⁇ RII extracellular domain sequence
  • Table 6 shows the linker connecting human immunoglobulin G4 and human TIM-3 extracellular domain sequence and human TIM-3 extracellular domain sequence
  • Table 7 shows the linker connecting human immunoglobulin G4 and human PEDF 44-mer peptide sequence and human PEDF 44-mer peptide sequence
  • the ExpiCHO expression system (Thermo Fisher) was used to produce a bispecific antibody containing a first antigen binding site that specifically binds to human angiopoietin-2.
  • ExpiCHO was cultured in a 125 mL flask (Corning) using 30 mL of ExpiCHO expression medium (Thermo Fisher) until the cell number reached 6
  • a total of 25 ⁇ g of heavy chain and light chain DNA prepared in Example 1 was prepared at a 1:1.5 ratio (w/w) using 1 mL of OptiPRO SFM (Thermo Fisher).
  • 80 ⁇ L of ExpiFectamine CHO reagent (Thermo Fisher) was prepared by adding it to 920 ⁇ L of OptiPRO SFM.
  • transfection was performed by adding it to 25 mL of the ExpiCHO culture medium prepared earlier within 5 minutes, and cultured at 37°C, 8% CO 2 , and 125 rpm.
  • 150 ⁇ L of ExpiFectamine CHO Enhancer (Thermo Fisher) and 4 mL of ExpiCHO Feed (Thermo Fisher) were added to the culture medium, and the conditions were changed to 32°C, 5% CO 2 , and 125 rpm.
  • an additional 4 mL of ExpiCHO Feed was added.
  • the culture was transferred to a conical tube (SPL) and centrifuged at 3,500 g for 30 minutes at 4°C. The supernatant was collected and filtered using a 0.22 ⁇ m CA syringe filter (Sartorius), and -70 Stored in a deep freezer at °C.
  • Amicon 30 kDa (Millipore) was used to change the buffer of the purified bispecific antibody to PBS. 5 mL of PBS was added to Amicon, centrifuged at 4°C, 5,000 rpm, and cleaned. After centrifugation, all of the solution was discarded, 10 mL of the sample eluted earlier was added, and then centrifuged at 4°C and 5,000 rpm to concentrate to about 3 mL. After adding 12 mL of PBS to the concentrated 3 mL, it was centrifuged again and concentrated to 3 mL. This process was repeated four additional times and the buffer was exchanged for PBS. Bispecific antibodies were prepared by performing sterile filtration on the sample after buffer exchange was completed using a 0.22 ⁇ m syringe filter (Millipore).
  • Example 3 SDS-PAGE analysis of purified anti-Ang2/VEGF antibody samples
  • Example 2 To confirm the purity and integrity of the anti-Ang2/VEGF antibody purified in Example 2, it was analyzed by SDS-PAGE. To 15 ⁇ L of a sample of appropriate protein concentration, add SDS-Sample Buffer 4X reducing (Gendepot) or Native SDS-Sample Buffer 4X non reducing (Gendepot) to 1X each, and heat the reducing sample at 98°C for 10 minutes using a heat block. Samples were prepared by heat treatment. NuPAGE 4-12%, Bis-Tris mini protein gel (Invitrogen) was installed in the XCell SureLock Mini-Cell (Invitrogen), and NuPage 1X MOPS SDS running buffer (Invitrogen) was added to prepare for electrophoresis.
  • SDS-Sample Buffer 4X reducing Gendepot
  • Native SDS-Sample Buffer 4X non reducing Gendepot
  • Example 4 SDS-PAGE and Western blot analysis of purified anti-Ang2/TGF- ⁇ antibody samples
  • Example 2 To confirm the purity and integrity of the anti-Ang2/TGF- ⁇ antibody purified in Example 2 in the same manner as in Example 3, it was analyzed by SDS-PAGE. As shown in Figure 3, a band of the desired size was confirmed under reducing (R) and non-reducing (NR) conditions, and it was confirmed that there was almost no impurity.
  • TGF- ⁇ binding region of the anti-Ang2/TGF- ⁇ antibody Western blot analysis was performed using anti-hTGF- ⁇ RII antibody. 60 ng and 600 ng of purified anti-Ang2/TGF- ⁇ antibody were subjected to SDS-PAGE electrophoresis in the same manner as in Example 3.
  • the gel was mounted on the After the transfer, the membrane was placed in 0.05% TBST (Tris Buffered Saline containing 0.05% (v/v) Tween-20) containing 5% (w/v) skim milk and blocked for 1 hour. After washing the blocked membrane three times with 0.1% TBST, it was added to primary antibody buffer (2 ⁇ g/mL Goat anti-hTGF- ⁇ RII antibody (R&D systems), 5% (w/v) skim milk, 0.05% TBST). A membrane was added and allowed to bind for 2 hours.
  • TBST Tris Buffered Saline containing 0.05% (v/v) Tween-20
  • primary antibody buffer 2 ⁇ g/mL Goat anti-hTGF- ⁇ RII antibody (R&D systems), 5% (w/v) skim milk, 0.05% TBST.
  • ELISA was performed to confirm the binding ability of the anti-Ang2/VEGF antibody with human Ang2, human VEGF, or mouse VEGF.
  • a 96-well MaxiSorpTM flat-bottom plate was coated with 1 ⁇ g/mL of human Ang2 (Sino Biological), human VEGF 165 (Sino Biological), or mouse VEGF 164 (Sino Biological). Then, the plate was washed five times with PBST (Phosphate Buffer Saline containing 0.05% (v/v) Tween-20) and then washed with PBST containing 1% (v/v) skim milk at room temperature for 2 hours. Blocked.
  • PBST Phosphate Buffer Saline containing 0.05% (v/v) Tween-20
  • ELISA was performed to confirm the ability of the anti-Ang2/VEGF antibody to simultaneously bind Ang2 and VEGF.
  • 1 ⁇ g/mL human VEGF 165 (Sino Biological) was coated on a 96-well MaxiSorpTM flat-bottom plate. The coated plate was washed five times with PBST and then blocked with PBST containing 1% (v/v) skim milk at room temperature for 2 hours. 60 nM of human Ang2 (Sino Biological) was mixed in advance with 11 different concentrations of anti-Ang2/VEGF antibody, serially diluted 3 times from 300 nM, and left at room temperature for 30 minutes.
  • the anti-Ang2/VEGF antibody developed in the present invention can bind to Ang2 and VEGF at the same time.
  • Bio-layer interferometry (BLI) analysis was performed to confirm the binding ability of the anti-Ang2/TGF- ⁇ antibody to Ang2 or TGF- ⁇ .
  • Octet Amine Reactive Second-Generation (AR2G) Biosensor (Sartorius) was placed in 1X kinetics buffer included in the AR2G kit and hydrated for 10 minutes. After completing hydration, the biosensor was placed in a 0.5 mL LightSafe micro centrifuge tube (Sigma) containing 1X kinetics buffer, and the initial baseline was measured for 60 seconds.
  • the biosensor was placed in a 0.5 mL LightSafe micro centrifuge tube containing 225 ⁇ L of distilled water and 12.5 ⁇ L each of 400 mM EDC and 200 mM S-NHS and activated for 300 seconds. After loading 4 ⁇ L of 10 mM acetate buffer (pH 5.0) containing 20 ⁇ g/mL TGF- ⁇ (Acrobiosystems) into the drop holder, the activated biosensor was added and immobilized for 300 seconds. The immobilized biosensor was placed in a 0.5ml LightSafe micro centrifuge tube containing 250 ⁇ L of 1 M ethanolamine (pH 8.5) and quenched for 300 seconds. After quenching was completed, the biosensor was placed back in 1 proceeded. After the association was completed, the biosensor was placed in 1X kinetics buffer and dissociation was measured for 600 seconds. After completing the analysis, ka, kd, and KD were calculated by global fitting.
  • Example 7 Western blot analysis to confirm activation of Tie2 signaling by anti-Ang2/VEGF antibody or anti-Ang2/TGF- ⁇ antibody
  • Ang2 binds to the Tie2 receptor expressed on vascular endothelial cells and acts as a weak agonist or antagonist.
  • the anti-Ang2/VEGF antibody or anti-Ang2/TGF- ⁇ antibody developed in the present invention can bind to Ang2 and form a complex with Ang2-Tie2, thereby activating the Tie2 receptor and promoting downstream signaling.
  • ERK and AKT phosphorylation experiments were performed in HUVEC. In order to compare the degree of activation of Tie2 downstream signaling, the same test was performed on the group treated only with Ang2 (Sino Biological).
  • HUVEC Longza cells
  • 60 nM of anti-Ang2/VEGF antibody or anti-Ang2/TGF- ⁇ antibody was mixed with 40 nM of Ang2 protein (Sino Biological) and left for 30 minutes. Then, the cultured cells were treated and incubated for another 10 minutes. For comparison, a group treated with only 40 nM Ang2 without antibody was also prepared. After washing the cells using PBS, lysis buffer (Ripa buffer (Biosesang), 0.15M Sodium chloride, 1% Triton , and 2 mM EDTA), centrifuged at 13,000 rpm for 15 minutes, and the supernatant was recovered to obtain a cell lysate.
  • anti-Ang2/VEGF antibody and anti-Ang2/TGF- ⁇ antibody showed AKT and ERK activation signals compared to the negative control group.
  • Example 8 Competitive VEGF binding of anti-Ang2/VEGF antibody to VEGFR2
  • VEGF is a representative pro-angiogenic cytokine that promotes blood vessel formation.
  • VEGF is known to regulate angiogenesis by binding to VEGFR2 and activating downstream signals.
  • the anti-Ang2/VEGF antibody designed in the present invention can bind to VEGF competitively with VEGFR2 and inhibit VEGF from activating the VEGFR2 downstream signal.
  • ELISA analysis was performed to confirm competitive VEGF binding of anti-Ang2/VEGF antibody and VEGFR2.
  • Greiner Bio-One 96-well Half Area ELISA Microplates were coated with 2 ⁇ g/mL human VEGFR2 ECD (Sino Biological). The plate was washed five times with PBST (Phosphate Buffer Saline containing 0.05% (v/v) Tween-20) and then blocked with PBST containing 1% (w/v) skim milk at room temperature for 2 hours. After washing the plate 5 times with PBST, pre-incubate 150 ⁇ g/mL of human VEGF (Sino Biological) with AVI tag and 11 different concentrations of anti-Ang2/VEGF antibody serially diluted 3 times from 1,500 nM for 10 minutes at room temperature.
  • PBST Phosphate Buffer Saline containing 0.05% (v/v) Tween-20
  • VEGF is a representative pro-angiogenic cytokine that promotes blood vessel formation, and VEGF is known to promote HUVEC proliferation.
  • Anti-Ang2/VEGF antibodies can inhibit HUVEC proliferation by binding to VEGF in the culture medium and inhibiting VEGF from binding to VEGFR2 of HUVEC. To confirm this, an experiment was performed to check the proliferation of HUVEC by adding VEGF or VEGF and anti-Ang2/VEGF antibody to HUVEC.
  • EBM-2 media (Lonza) containing 0.5% FBS
  • HUVEC HUVEC
  • SPL 96 well cell culture plate
  • EBM-2 media containing 0.5% FBS was prepared 30 minutes before treating the cells with VEGF 30 ng/mL, VEGF 30 ng/mL + anti-Ang2/VEGF antibody 2.5 nM.
  • 100 ⁇ L of the previously prepared sample was added to each well and cultured in an incubator (37°C, 5% CO 2 ) for 72 hours.
  • Example 10 Inhibition of neovascularization and improvement of visual acuity of anti-Ang2/VEGF antibody in Mouse Laser-induced CNV model
  • IVT intravitreal injection of CNV control group and positive control group, IgG and aflibercept
  • IgG and aflibercept positive control group
  • the test group, anti-Ang2/VEGF antibody was administered as a single IVT at a dose of 5.8 ⁇ g/ ⁇ L/eye (about 35 ⁇ M).
  • FFA fundus fluorescein angiography
  • OCT optical coherence tomography
  • Electroretinogram evaluation was performed on the scotopic ERG (electroretinogram) 12 days after CNV induction. Electroretinogram changes in response to mono-flash (0.9 log cds/m2) stimulation were evaluated using B-wave amplitude.
  • the fused cells were cultured in a medium (HAT mediaum) supplemented with hypoxantin, aminopterine, and thymidine, and cells (hybridoma) in which B lymphocytes and sp2/0 cells were fused were selectively selected.
  • the obtained hybridoma cells were separated into positive and negative cells using a serial dilution method, and the cloning process was repeated to prepare monoclonal cells that produce antibodies that react to the antigen.
  • the obtained antibody-producing hybridoma cells were cultured in DMEM (Dulbeco's Modified Eagle's Mediaum) containing 10% (v/v) FBS at 37°C and 5% CO 2 conditions, and then centrifuged to obtain antibody-producing cells.
  • the culture medium in which the antibodies were secreted was separated, and the antibodies were purified using an affinity column (Protein A/G agarose column, Protein A/G (GenDEPOT)).

Landscapes

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

Abstract

The present invention relates to a bispecific antibody including a first antigen-binding site that specifically binds to human angiopoietin-2 (ANG-2), and a second antigen-binding site that specifically binds to at least one antigen selected from the group consisting of human vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-beta, T-cell immunoglobulin and mucin domain-containing protein (TIM)-3 ligand, and pigment epithelium-derived factor receptor (PEDF-R), wherein the first antigen-binding site that specifically binds to the ANG-2 includes, in the heavy chain variable domain, a CDRH1 region of SEQ ID NO: 1 or 7 or 13, a CDRH2 region of SEQ ID NO: 2 or 8 or 14, and a CDRH3 region of SEQ ID NO: 3 or 9 or 15, and includes, in the light chain variable domain, a CDRL1 region of SEQ ID NO: 4 or 10 or 16, a CDRL2 region of SEQ ID NO: 5 or 11 or 17, and a CDRL3 region of SEQ ID NO: 6 or 12 or 18.

Description

인간 안지오포이에틴-2에 특이적으로 결합하는 제1 항원 결합 부위를 포함하는 이중특이적 항체 및 그 응용Bispecific antibody comprising a first antigen binding site that specifically binds to human angiopoietin-2 and applications thereof
본 발명은 인간 안지오포이에틴-2에 특이적으로 결합하는 제1 항원 결합 부위를 포함하는 이중특이적 항체 및 그 응용에 관한 것이다.The present invention relates to a bispecific antibody comprising a first antigen binding site that specifically binds to human angiopoietin-2 and its applications.
안지오포이에틴-2(이하, 'ANG-2'라고도 함)는 혈관내피세포에 존재하는 수용체 Tie2에 결합하지만 길항적인 리간드 (antagonistic ligand)로서 작용하여, Tie2의 작용물질 (agonist)인 안지오포이에틴-1(Angiopoietin-1; Ang1)과 Tie2 결합에 대해 경쟁함으로써 Tie2에 의한 신호전달을 억제하는 작용을 한다. 이러한 작용 기전으로 인하여, 혈관 내피 성장인자(Vascular Epidermal Growth Factor, 이하 'VEGF'라 함)의 과발현 또는 inflammation 상태에서는 혈관내피세포가 활성화되며, 혈관 투과성(vascular permeability)이 증가되는데, 이때 Ang1이 혈관내피세포의 안정화를 유도하고 혈관 투과성을 감소시키는 반면, 활성화된 혈관내피세포에서 증가된 Ang2는 Ang1과 경쟁함으로써 Ang1에 의한 혈관내피세포 안정화를 억제하는 역할을 한다. 따라서 Ang2는 VEGF가 존재할 때 혈관내피세포의 안정성을 유지하는 Ang1-Tie2 결합과 이를 통한 신호전달을 저해하여, 결과적으로 혈관의 신생혈관형성을 촉진함으로써 결과적으로 혈관 생성 증가, 혈관의 불안정화, 혈관 투과성 증가를 초래한다. 한편, Ang2는 Tie2 수용체의 비활성을 유도하는 antagonist로서의 역할 외에 림프관 형성 및 유지를 포함한 몇몇의 특정 상황에서는 Tie2 수용체의 활성을 유도하는 작용제(agonist) 특성이 보고된 바 있기 때문에 antagonist로서의 기능과 약한 agonist의 기능을 모두 갖고 상황에 따라서 다양한 기능을 수행하는 것으로 여겨진다. Angiopoietin-2 (hereinafter also referred to as 'ANG-2') binds to the receptor Tie2 present on vascular endothelial cells, but acts as an antagonistic ligand, producing angiophore, an agonist of Tie2. It acts to inhibit signal transduction by Tie2 by competing with Angiopoietin-1 (Ang1) for binding to Tie2. Due to this mechanism of action, when vascular endothelial growth factor (Vascular Epidermal Growth Factor, hereinafter referred to as 'VEGF') is overexpressed or in a state of inflammation, vascular endothelial cells are activated and vascular permeability increases. In this case, Ang1 While it induces stabilization of endothelial cells and reduces vascular permeability, increased Ang2 in activated vascular endothelial cells competes with Ang1, thereby inhibiting stabilization of vascular endothelial cells by Ang1. Therefore, Ang2 inhibits Ang1-Tie2 binding and signaling through Ang1-Tie2, which maintains the stability of vascular endothelial cells in the presence of VEGF, and ultimately promotes angiogenesis in blood vessels, resulting in increased angiogenesis, destabilization of blood vessels, and vascular permeability. causes an increase Meanwhile, in addition to its role as an antagonist that induces the inactivation of the Tie2 receptor, Ang2 has been reported to have agonist properties that induce the activity of the Tie2 receptor in some specific situations, including the formation and maintenance of lymphatic vessels. Therefore, it functions as an antagonist and a weak agonist. It is believed to have all of the following functions and perform various functions depending on the situation.
신생혈관 형성과정은 암의 성장에 필수적인 요소이므로, 위와 같은 Tie2 의존적인 Ang2의 기능을 저해하여 신생 혈관형성을 억제함으로써 암의 추가적인 성장을 막고자 하는 시도가 있어왔으나 이들은 대부분의 경우 Ang2와 Tie2의 결합을 저해하여 antagonist로서의 역할을 막는 기능을 갖는 것으로 알려져 있다. Ang2의 Tie2에 대한 결합을 방해하는 항체 외에도, Tie2 수용체에 직접 결합하여 인산화 및 활성화를 유도하는 재조합 단백질이나 항체들도 보고된 바 있다. Since the process of neovascularization is an essential factor in the growth of cancer, there have been attempts to prevent further growth of cancer by inhibiting neovascularization by inhibiting the Tie2-dependent function of Ang2 as described above. However, in most cases, these are It is known to have the function of inhibiting binding and preventing its role as an antagonist. In addition to antibodies that interfere with the binding of Ang2 to Tie2, recombinant proteins or antibodies that directly bind to the Tie2 receptor and induce phosphorylation and activation have been reported.
근래에는 Ang2에 결합하여 Tie2에 함께 결합함으로써 Tie2 clustering을 통하여 Tie2 인산화 및 활성화하는 항체도 보고되었던 바 있는데, 이는 단순히 Ang2의 antagonist 역할을 막는데 그치는 것이 아니라 오히려 agonist로 작용하게 함으로써 보다 효과적인 혈관 정상화의 가능성을 제시한 것이다.Recently, an antibody that binds to Ang2 and binds to Tie2 to phosphorylate and activate Tie2 through Tie2 clustering has been reported. This does not simply block Ang2's role as an antagonist, but rather acts as an agonist, leading to more effective vascular normalization. It suggests a possibility.
한편 라니비주맙(Ranibizumab)(상표명 루센티스(Lucentis)®)은 베바시주맙(bevacizumab)(아바스틴(Avastin))과 동일한 모 뮤린 항체로부터 유도된 단일클론 항체 단편이다. 그러나, 이 항체는 VEGF-A에 대한 보다 강한 결합을 제공하도록 친화 성숙되어 있다(국제 특허출원 공개 제WO 98/45331호). VEGF-A 차단이 일부 전신 독성과 관련되어 있을 수 있으므로, 라니비주맙은 혈청 반감기를 감소시켜 결과적으로 전신 독성을 감소시키도록 Fc 부분을 상실하고 있다고 공지되어 있다. Meanwhile, Ranibizumab (brand name Lucentis®) is a monoclonal antibody fragment derived from the same parent murine antibody as bevacizumab (Avastin). However, this antibody has been affinity matured to provide stronger binding to VEGF-A (International Patent Application Publication No. WO 98/45331). As VEGF-A blockade may be associated with some systemic toxicity, it is known that ranibizumab is losing its Fc portion to reduce serum half-life and consequently systemic toxicity.
국제 특허출원 공개 제WO 2010/040508호 및 제WO 2011/117329호는 이중특이적 항-VEGF/항-ANG-2 항체에 관한 것이다.International Patent Application Publication Nos. WO 2010/040508 and WO 2011/117329 relate to bispecific anti-VEGF/anti-ANG-2 antibodies.
본 발명의 목적은 인간 ANG-2에 특이적으로 결합하는 부위 및 인간 VEGF, TGF-베타, TIM-3 리간드 또는 PEDF-R (PNPLA2, ATGL)에 특이적으로 결합하는 부위를 포함하는 이중특이적 항체를 제공하는 것이다.The object of the present invention is a bispecific antibody comprising a site that specifically binds to human ANG-2 and a site that specifically binds to human VEGF, TGF-beta, TIM-3 ligand or PEDF-R (PNPLA2, ATGL). It provides antibodies.
본 발명의 다른 목적은 상기 이중특이적 항체를 유효성분으로 하는 약학조성물의 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition containing the bispecific antibody as an active ingredient.
상기의 목적을 달성하기 위하여 본 발명은 인간 안지오포이에틴(ANG)-2에 특이적으로 결합하는 제1 항원 결합 부위 및 인간 혈관내피 성장인자(VEGF), 형질전환 성장인자(Tansforming growth factor, 이하, 'TGF'라 함)-베타, T세포 면역글로불린 및 뮤신 도메인 함유 단백질(T-cell immunoglobulin and mucin-domain containing, 이하, 'TIM'이라 함)-3의 리간드, 및 색소상피유래인자 수용체(pigment epithelium-derived factor receptor, 이하, 'PEDF-R'라 함)로 구성된 군으로부터 선택된 하나의 항원에 특이적으로 결합하는 제2 항원 결합 부위를 포함하는 이중특이적 항체로서,In order to achieve the above object, the present invention provides a first antigen binding site that specifically binds to human angiopoietin (ANG)-2, human vascular endothelial growth factor (VEGF), transforming growth factor, Hereinafter referred to as 'TGF')-beta, the ligand of T-cell immunoglobulin and mucin-domain containing protein (T-cell immunoglobulin and mucin-domain containing, hereinafter referred to as 'TIM')-3, and pigment epithelium-derived factor receptor. A bispecific antibody comprising a second antigen binding site that specifically binds to one antigen selected from the group consisting of (pigment epithelium-derived factor receptor, hereinafter referred to as 'PEDF-R'),
i) 상기 ANG-2에 특이적으로 결합하는 상기 제1 항원 결합 부위가 서열번호 1 또는 7 또는 13의 CDRH1 영역, 서열번호 2 또는 8 또는 14의 CDRH2 영역 및 서열번호 3 또는 9 또는 15의 CDRH3 영역을 중쇄 가변 도메인 내에 포함하고, 서열번호 4 또는 10 또는 16의 CDRL1 영역, 서열번호 5 또는 11 또는 17의 CDRL2 영역 및 서열번호 6 또는 12 또는 18의 CDRL3 영역을 경쇄 가변 도메인 내에 포함하고;i) The first antigen binding site that specifically binds to the ANG-2 is the CDRH1 region of SEQ ID NO: 1 or 7 or 13, the CDRH2 region of SEQ ID NO: 2 or 8 or 14, and the CDRH3 of SEQ ID NO: 3 or 9 or 15 comprising a region within a heavy chain variable domain, a CDRL1 region of SEQ ID NO: 4 or 10 or 16, a CDRL2 region of SEQ ID NO: 5 or 11 or 17 and a CDRL3 region of SEQ ID NO: 6 or 12 or 18 within a light chain variable domain;
ii) 상기 VEGF에 특이적으로 결합하는 상기 제2 항원 결합 부위는 VEGFR1 D2 도메인을 포함하고, 상기 TGF-베타에 특이적으로 결합하는 상기 제2 항원 결합 부위는 TGF-베타 RII ECD을 포함하고, 상기 TIM-3의 리간드에 특이적으로 결합하는 상기 제2 항원 결합 부위는 TIM-3 ECD를 포함하고, 상기 PEDF-R에 특이적으로 결합하는 상기 제2 항원 결합 부위는 44-머(mer)의 PEDF를 포함하며,ii) the second antigen binding site specifically binding to VEGF comprises a VEGFR1 D2 domain, and the second antigen binding site specifically binding to TGF-beta comprises TGF-beta RII ECD, The second antigen binding site that specifically binds to the ligand of TIM-3 includes TIM-3 ECD, and the second antigen binding site that specifically binds to the PEDF-R includes a 44-mer. Contains a PEDF of
iii) 상기 항체는 Tie2 활성화를 유도하는, 이중특이적 항체를 제공한다.iii) The antibody provides a bispecific antibody that induces Tie2 activation.
본 발명의 일 구현예에 있어서, 상기 VEGFR1 D2도메인은 서열번호 21의 VEGFR1 D2 도메인, 상기 TGF-베타 RII ECD는 서열번호 24의 TGF-베타 RII ECD, 상기 TIM-3 ECD는 서열번호 27의 TIM-3 ECD, 및 상기 44-머(mer)의 PEDF는 서열번호 30의 아미노산 서열을 포함하는 것이 바람직하나 이에 한정되지 아니한다.In one embodiment of the present invention, the VEGFR1 D2 domain is the VEGFR1 D2 domain of SEQ ID NO: 21, the TGF-beta RII ECD is the TGF-beta RII ECD of SEQ ID NO: 24, and the TIM-3 ECD is the TIM of SEQ ID NO: 27 -3 ECD and the 44-mer PEDF preferably include the amino acid sequence of SEQ ID NO: 30, but are not limited thereto.
본 발명의 다른 구현예에 있어서, 상기 VEGFR1 D2 도메인, 상기 TGF-베타 RII ECD, 상기 TIM-3 ECD, 및 상기 44-머(mer)의 PEDF는 면역글로불린과 링커로 연결된 것이 바람직하며, 상기 링커는 각각 서열번호 20, 서열번호 23, 서열번호 26 또는 서열번호 29 링커인 것이 더욱 바람직하나 이에 한정되지 아니한다.In another embodiment of the present invention, the VEGFR1 D2 domain, the TGF-beta RII ECD, the TIM-3 ECD, and the 44-mer PEDF are preferably connected to an immunoglobulin by a linker, and the linker It is more preferable that the linker is SEQ ID NO: 20, SEQ ID NO: 23, SEQ ID NO: 26, or SEQ ID NO: 29, respectively, but is not limited thereto.
또 본 발명은 상기 본 발명에 따른 항체를 포함하는 암 치료를 위한 약학적 조성물을 제공한다.Additionally, the present invention provides a pharmaceutical composition for the treatment of cancer containing the antibody according to the present invention.
또 본 발명은 상기 본 발명에 따른 항체를 포함하는 혈관 질환 치료를 위한 약학적 조성물을 제공한다.Additionally, the present invention provides a pharmaceutical composition for the treatment of vascular diseases containing the antibody according to the present invention.
또한 본 발명은 상기 본 발명에 따른 이중특이성 항체를 인코딩하는 핵산을 제공한다.The present invention also provides a nucleic acid encoding the bispecific antibody according to the present invention.
또한 본 발명은 상기 본 발명에 따른 이중특이성 항체의 제조 방법으로서, 원핵 또는 진핵 숙주 세포에서 상기 이중특이성 항체를 인코딩하는 핵산을 발현하고, 세포 또는 세포 배양 상청액으로부터 이중특이성 항체를 회수하는 것을 특징으로 하는, 이중특이성 항체의 제조 방법을 제공한다.The present invention also provides a method for producing a bispecific antibody according to the present invention, comprising expressing a nucleic acid encoding the bispecific antibody in a prokaryotic or eukaryotic host cell and recovering the bispecific antibody from the cell or cell culture supernatant. Provides a method for producing a bispecific antibody.
본 발명에 따른 "이중특이성 항체"는 2 개의 상이한 항원-결합 특이성을 갖는 항체이다. 항체가 하나 이상의 특이성을 갖는 경우, 인식되는 에피토프는 단일 항원 또는 하나 이상의 항원과 관련될 수 있다. 본 발명의 항체는 2 개의 상이한 항원, 즉 제 1 항원으로서 ANG-2 및 제 2 항원으로서 VEGF, TGF-베타, TIM-3의 리간드 또는 PEDF-R에 대해 특이적이다.A “bispecific antibody” according to the invention is an antibody with two different antigen-binding specificities. If an antibody has more than one specificity, the epitope recognized may be associated with a single antigen or more than one antigen. The antibodies of the invention are specific for two different antigens, ANG-2 as the first antigen and VEGF, TGF-beta, the ligand of TIM-3 or PEDF-R as the second antigen.
본 발명의 이중특이성 항체는, 예를 들어, 다가 단쇄 항체, 디아바디(diabodies) 및 트리아바디(triabodies) 뿐만 아니라, 하나 이상의 펩티드-링커를 통해 추가의 항원 결합 부위 (예를 들어, 단쇄 Fv, VH 도메인 및/또는 VL 도메인, Fab, 또는 (Fab)2) 가 연결되는 전장 항체의 불변 도메인 구조를 갖는 항체를 포함한다. 항체는 단일 종으로부터의 전장이거나, 키메라화 또는 인간화될 수 있다. 2 이상의 항원 결합 부위를 갖는 항체의 경우, 단백질이 2 개의 상이한 항원에 대한 결합 부위를 갖는 한, 일부 결합 부위는 동일할 수 있다. 즉, 제 1 결합 부위는 ANG-2에 대해 특이적인 한편, 제 2 결합 부위는 VEGF, TGF-베타, TIM-3 리간드, 및 PEDF-R에 대해 특이적이고, 그 반대도 동일하다.Bispecific antibodies of the invention include, for example, multivalent single chain antibodies, diabodies and triabodies, as well as additional antigen binding sites (e.g., single chain Fv, It includes antibodies having the constant domain structure of a full-length antibody to which a VH domain and/or a VL domain, Fab, or (Fab)2) are linked. Antibodies can be full length from a single species, chimeric or humanized. For antibodies with two or more antigen binding sites, some of the binding sites may be identical, as long as the protein has binding sites for two different antigens. That is, the first binding site is specific for ANG-2, while the second binding site is specific for VEGF, TGF-beta, TIM-3 ligand, and PEDF-R, and vice versa.
특정 구현예에서, 본 발명의 항체는 하나 이상의 면역글로불린 클래스의 면역글로불린 불변부를 추가로 포함한다. 면역글로불린 클래스는 IgG, IgM, IgA, IgD 및 IgE 동종형 및, IgG 및 IgA의 경우, 그의 서브유형을 포함한다. 바람직한 구현예에서, 본 발명의 항체는 IgG 유형 항체의 불변 도메인 구조를 가지나, 4 개의 항원 결합 부위를 갖는다.In certain embodiments, the antibodies of the invention further comprise immunoglobulin constant regions of one or more immunoglobulin classes. Immunoglobulin classes include IgG, IgM, IgA, IgD and IgE isotypes and, in the case of IgG and IgA, their subtypes. In a preferred embodiment, the antibodies of the invention have the constant domain structure of an IgG type antibody, but have four antigen binding sites.
용어 "키메라 항체"는 통상적으로 재조합 DNA 기술에 의해 제조되는, 하나의 출처 또는 종으로부터의 가변부, 즉 결합부, 및 상이한 출처 또는 종으로부터 유래된 불변부의 적어도 일부를 포함하는 항체를 언급한다. 쥐 가변부 및 인간 불변부를 포함하는 키메라 항체가 바람직하다. 본 발명에 포함되는 "키메라 항체" 의 다른 바람직한 형태는 불변부가 원래의 항체의 불변부로부터 변경 또는 변화되어, 특히 C1q 결합 및/또는 Fc 수용체(FcR) 결합과 관련된 본 발명에 따른 특성을 생성하는 것이다. 상기 키메라 항체는 "클래스-전환된 항체"로서 또한 언급된다. 키메라 항체는 면역글로불린 가변부를 인코딩하는 DNA 분절 및 면역글로불린 불변부를 인코딩하는 DNA 분절을 포함하는 발현된 면역글로불린 유전자의 생성물이다. 키메라 항체의 제조 방법은 당업계에 잘 알려져 있는 종래의 재조합 DNA 및 유전자 트랜스펙션 기술을 포함한다. 예를 들어, Morrison,S.L., et al., Proc. Natl. Acad. Sci. USA 81 (1984) 6851-6855; US 5,202,238 및 US 5,204,244 참조.The term “chimeric antibody” refers to an antibody comprising a variable region, i.e., a binding region, from one source or species and at least a portion of a constant region from a different source or species, typically produced by recombinant DNA technology. Chimeric antibodies comprising murine variable regions and human constant regions are preferred. Other preferred forms of “chimeric antibodies” encompassed by the present invention are those wherein the constant portions are altered or varied from the constant portions of the original antibody, thereby producing properties according to the present invention, particularly with regard to C1q binding and/or Fc receptor (FcR) binding. will be. Such chimeric antibodies are also referred to as “class-switched antibodies”. Chimeric antibodies are the product of expressed immunoglobulin genes comprising a DNA segment encoding an immunoglobulin variable region and a DNA segment encoding an immunoglobulin constant region. Methods for producing chimeric antibodies include conventional recombinant DNA and gene transfection techniques well known in the art. For example, Morrison, S.L., et al., Proc. Natl. Acad. Sci. USA 81 (1984) 6851-6855; See US 5,202,238 and US 5,204,244.
용어 "인간화된 항체"는 골격 또는 "상보성 결정 부위" (CDR) 가 부모 면역글로불린과 비교하여 상이한 특이성의 면역글로불린의 CDR을 포함하도록 변경된 항체를 언급한다. 바람직한 구현예에서, 쥐 CDR 이 인간 항체의 골격 영역 내로 이식되어 "인간화된 항체" 를 제조한다. 예를 들어, Riechmann, L., et al., Nature 332 (1988) 323-327; 및 Neuberger, M.S., et al., Nature 314 (1985) 268-270 참조. 특히 바람직한 CDR은 키메라 항체에 대해 상기 언급된 항원을 인지하는 서열을 나타내는 것에 상응한다. The term “humanized antibody” refers to an antibody whose framework or “complementarity determining regions” (CDRs) have been altered to include the CDRs of an immunoglobulin of different specificity compared to the parent immunoglobulin. In a preferred embodiment, murine CDRs are grafted into the framework region of a human antibody to produce a “humanized antibody.” For example, Riechmann, L., et al., Nature 332 (1988) 323-327; and Neuberger, M.S., et al., Nature 314 (1985) 268-270. Particularly preferred CDRs correspond to those representing sequences recognizing the above-mentioned antigens for chimeric antibodies.
본원에서 사용된 용어 "인간 항체"는 인간 생식선 면역글로불린 서열로부터 유래된 가변 및 불변부를 갖는 항체를 포함하는 것으로 의도된다. 인간 항체는 당업계에 잘 알려져 있다 (van Dijk, M.A., 및 van de Winkel, J.G., Curr. Opin. Chem. Biol. 5 (2001) 368-374). 인간 항체는 면역화 시 내인성 면역글로불린 생산의 부재 하에 인간 항체의 전체 레퍼토리 또는 선택물을 생산할 수 있는 유전자 도입 동물 (예를 들어 마우스) 에서 또한 생산될 수 있다. 상기 생식선 돌연변이 마우스 내에 인간 생식선 면역글로불린 유전자 배열의 전달은 항원 접종시 인간 항체의 생산을 초래할 것이다 (예를 들어 Jakobovits, A., et al., Proc. Natl.Acad. Sci. USA 90 (1993) 2551-2555; Jakobovits, A., et al., Nature 362 (1993) 255-258; Bruggemann, M.,et al., Year Immunol. 7 (1993) 33-40 참조). 인간 항체는 파지 디스플레이 라이브러리에서 또한 생산될 수 있다 (Hoogenboom, H.R., and Winter, G., J. Mol. Biol. 227 (1992) 381-388; Marks, J.D., et al., J.Mol. Biol. 222 (1991) 581-597). 모노클로날 항체의 생산에 Cole, A., et al. and Boerner, P., et al.의 기술이 또한 이용가능하다 (Cole, A., et al., Monoclonal Antibodies and Cancer Therapy, Liss, A.L., p.77 (1985); 및 Boerner, P., et al., J. Immunol. 147 (1991) 86-95).As used herein, the term “human antibody” is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. Human antibodies are well known in the art (van Dijk, M.A., and van de Winkel, J.G., Curr. Opin. Chem. Biol. 5 (2001) 368-374). Human antibodies can also be produced in transgenic animals (e.g., mice) that can produce the full repertoire or selection of human antibodies upon immunization in the absence of endogenous immunoglobulin production. Transfer of a human germline immunoglobulin gene sequence into such germline mutant mice will result in the production of human antibodies upon challenge (e.g. Jakobovits, A., et al., Proc. Natl. Acad. Sci. USA 90 (1993) 2551-2555; Jakobovits, A., et al., Nature 362 (1993) 255-258; Bruggemann, M., et al., Year Immunol. Human antibodies can also be produced from phage display libraries (Hoogenboom, H.R., and Winter, G., J. Mol. Biol. 227 (1992) 381-388; Marks, J.D., et al., J. Mol. Biol 222 (1991) 581-597). For the production of monoclonal antibodies, Cole, A., et al. and Boerner, P., et al.'s techniques are also available (Cole, A., et al., Monoclonal Antibodies and Cancer Therapy, Liss, A.L., p. 77 (1985); and Boerner, P., et al. al., J. Immunol. 147 (1991) 86-95).
본원에서 사용된 용어 "재조합 인간 항체"는 재조합 방법에 의해 제조, 발현, 생성 또는 분리되는 모든 인간 항체, 예컨대 NS0 또는 CHO 세포와 같은 숙주 세포로부터 또는 인간 면역글로불린 유전자에 대해 유전자 도입된 동물 (예를 들어 마우스) 로부터 분리된 항체, 또는 숙주 세포 내로 트랜스펙션된 재조합 발현 벡터를 사용하여 발현된 항체를 포함하는 것으로 의도된다. 상기 재조합 인간 항체는 재배열된 형태의 가변 및 불변부를 갖는다. As used herein, the term “recombinant human antibody” refers to any human antibody that is manufactured, expressed, produced or isolated by recombinant methods, such as from a host cell such as NS0 or CHO cells or from an animal transgenic for human immunoglobulin genes (e.g. It is intended to include antibodies isolated from (e.g., mouse), or antibodies expressed using recombinant expression vectors transfected into host cells. The recombinant human antibody has variable and constant regions in rearranged form.
본원에서 사용된 용어, "CDR(complementarity determining region)"은 면역글로불린의 중쇄 및 경쇄의 고가변 영역(hypervariable region)의 아미노산 서열을 의미한다. 중쇄 및 경쇄는 각각 3개의 CDR을 포함할 수 있다(CDRH1, CDRH2, CDRH3 및 CDRL1, CDRL2, CDRL3). 상기 CDR은 항체가 항원 또는 에피토프에 결합하는 데 있어서 주요한 접촉 잔기를 제공할 수 있다. 한편, 본 명세서에 있어서, 용어, "특이적으로 결합" 또는 "특이적으로 인식"은 당업자에게 통상적으로 공지되어 있는 의미와 동일한 것으로서, 항원 및 항체가 특이적으로 상호작용하여 면역학적 반응을 하는 것을 의미한다.As used herein, the term “complementarity determining region (CDR)” refers to the amino acid sequence of the hypervariable region of the heavy and light chains of immunoglobulins. The heavy and light chains may each contain three CDRs (CDRH1, CDRH2, CDRH3 and CDRL1, CDRL2, CDRL3). The CDR may provide key contact residues for the antibody to bind to the antigen or epitope. Meanwhile, in the present specification, the term "specifically binds" or "specifically recognizes" has the same meaning commonly known to those skilled in the art, and refers to a condition in which an antigen and an antibody specifically interact to produce an immunological reaction. means that
본원에서 사용된 "가변 도메인" (경쇄 (VL)의 가변 도메인, 중쇄 (VH)의 가변부) 은 항원에 대한 항체 결합에 직접 관여하는 경 및 중쇄의 쌍의 각각을 의미한다. 가변 인간 경 및 중쇄의 도메인은 동일한 일반적 구조를 갖고, 각각의 도메인은 3 개의 "과가변부" (또는 상보성 결정 부위, CDR) 에 의해 연결된, 서열이 광범위하게 보존되는 4 개의 골격 (FR) 영역을 포함한다. 골격 영역은 β-시트 입체형태를 가지며, CDR 은 β-시트 구조를 연결하는 루프를 형성할 수 있다. 각각의 사슬 내의 CDR 은 골격 영역에 의해 3 차원적 구조를 유지하고, 다른 사슬로부터의 CDR 과 함께 항원 결합 부위를 형성한다.As used herein, “variable domain” (variable domain of the light chain (VL), variable region of the heavy chain (VH)) refers to each of the pairs of light and heavy chains that are directly involved in antibody binding to antigen. The domains of the variable human light and heavy chains have the same general structure, with each domain consisting of four framework (FR) regions of broadly conserved sequence, linked by three "hypervariable regions" (or complementarity determining regions, CDRs). Includes. The framework region has a β-sheet conformation, and the CDRs can form loops connecting the β-sheet structures. The CDRs within each chain maintain their three-dimensional structure by framework regions and, together with CDRs from other chains, form an antigen-binding site.
본원에서 사용된 용어 "과가변부" 또는 "항체의 항원-결합 부위"는 항원-결합을 책임지는 항체의 아미노산 잔기를 언급한다. 과가변부는 "상보성 결정 부위" 또는 "CDR" 로부터의 아미노산 잔기를 포함한다.As used herein, the term “hypervariable region” or “antigen-binding site of an antibody” refers to the amino acid residues of an antibody that are responsible for antigen-binding. Hypervariable regions include amino acid residues from the “complementarity determining region” or “CDR”.
"골격" 또는 "FR" 영역은 본원에서 정의된 과가변부 잔기를 제외한 가변 도메인 영역이다. 그러므로, 항체의 경 및 중쇄는 N- 으로부터 C-말단까지 도메인 FR1, CDR1, FR2, CDR2, FR3, CDR3 및 FR4 를 포함한다. 각각의 사슬 상의 CDR 은 상기 골격 아미노산에 의해 분리된다. 특히, 중쇄의 CDR3 은 항원 결합에 가장 기여하는 영역이다. CDR 및 FR 영역은 Kabat et al., Sequences of Proteins of Immunological Interest,5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991) 의 표준 정의에 따라 결정된다.The “framework” or “FR” region is the variable domain region excluding the hypervariable region residues as defined herein. Therefore, the light and heavy chains of the antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The CDRs on each chain are separated by the framework amino acids. In particular, CDR3 of the heavy chain is the region that most contributes to antigen binding. CDR and FR regions are determined according to the standard definitions of Kabat et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991).
본 발명에 따른 이중특이성 항체는 "보존적 서열 변경" 을 갖는 상기 항체 (이는 이중특이성 항체의 "변이체"를 의미함) 를 또한 포함한다. 이는 본 발명에 따른 항체의 상기 언급된 특성에 영향을 미치거나 이를 변경하지 않는 뉴클레오티드 및 아미노산 서열 변경을 의미한다. 변경은 부위-지정 돌연변이유발 및 PCR-매개 돌연변이유발과 같은 당업계에 알려진 표준 기술에 의해 도입될 수 있다. 보존적 아미노산 치환은 아미노산 잔기가 유사한 측쇄를 갖는 아미노산 잔기로 치환된 것을 포함한다. 유사한 측쇄를 갖는 아미노산 잔기의 패밀리가 당업계에서 규정되었다. 상기 패밀리는 염기성 측쇄를 갖는 아미노산 (예를 들어 라이신, 아르기닌, 히스티딘), 산성 측쇄를 갖는 아미노산 (예를 들어 아스파르트산, 글루탐산), 비하전된 극성 측쇄를 갖는 아미노산 (예를 들어 글라이신, 아스파라긴, 글루타민, 세린, 트레오닌, 타이로신, 시스테인, 트립토판), 무극성 측쇄를 갖는 아미노산 (예를 들어 알라닌, 발린, 류신, 이소류신, 프롤린, 페닐알라닌, 메티오닌), 베타-분지 측쇄를 갖는 아미노산 (예를 들어 트레오닌, 발린, 이소류신) 및 방향족 측쇄를 갖는 아미노산 (예를 들어 타이로신, 페닐알라닌, 트립토판, 히스티딘) 을 포함한다. 따라서 이중특이성 항체 내의 예측된 불필수 아미노산 잔기는 바람직하게는 동일한 측쇄 패밀리로부터의 또다른 아미노산 잔기로 치환될 수 있다.Bispecific antibodies according to the invention also include said antibodies with “conservative sequence alterations” (this means “variants” of bispecific antibodies). This means nucleotide and amino acid sequence changes that do not affect or alter the above-mentioned properties of the antibody according to the invention. Changes can be introduced by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions involve the replacement of an amino acid residue with an amino acid residue having a similar side chain. Families of amino acid residues with similar side chains have been defined in the art. This family includes amino acids with basic side chains (e.g. lysine, arginine, histidine), amino acids with acidic side chains (e.g. aspartic acid, glutamic acid), amino acids with uncharged polar side chains (e.g. glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), amino acids with nonpolar side chains (e.g. alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), amino acids with beta-branched side chains (e.g. threonine, valine, isoleucine) and amino acids with aromatic side chains (e.g. tyrosine, phenylalanine, tryptophan, histidine). Accordingly, a predicted non-essential amino acid residue in a bispecific antibody may be replaced with another amino acid residue, preferably from the same side chain family.
그러므로 본원에서 "변이체" 이중특이성 항체는 아미노산 서열이 "부모" 이중특이성 항체 아미노산 서열과 부모 항체의 하나 이상의 가변부 또는 불변부 내의 10 개 이하, 바람직하게는 약 2 내지 약 5 개의 부가, 결실 및/또는 치환에 의해 상이한 분자를 언급한다. 아미노산 치환은 Riechmann, L., et al., Nature 332 (1988) 323-327 및 Queen, C, et al., Proc. Natl. Acad. Sci. USA 86 (1989) 10029-10033 에 의해 기술된 분자 모델링에 기초한 돌연변이유발에 의해 수행될 수 있다. 본 발명에 따른 "변이체" 이중특이성 항체는 그 안의 링커 (존재하는 경우) 가 변형되거나, 또다른 링커에 의해 치환된 이중특이성 항체 형태를 또한 포함한다.Therefore, as used herein, a "variant" bispecific antibody is one whose amino acid sequence differs from the "parent" bispecific antibody amino acid sequence by at most 10, preferably about 2 to about 5, additions, deletions, and deletions within one or more variable or constant regions of the parent antibody. /or refer to a different molecule by substitution. Amino acid substitutions are as described in Riechmann, L., et al., Nature 332 (1988) 323-327 and Queen, C, et al., Proc. Natl. Acad. Sci. USA 86 (1989) 10029-10033 may be performed by mutagenesis based on molecular modeling. “Variant” bispecific antibodies according to the invention also include bispecific antibody forms in which the linker (if present) therein is modified or replaced by another linker.
본원에서 사용된 용어 "결합" 또는 "특이적 결합" 은 시험관내 검정에서, 항원 (인간 VEGF, ANG-2, TGF-베타, TIM-3 리간드 또는 PEDF-R)의 에피토프에 대한 항체의 결합을 언급한다. As used herein, the term “binding” or “specific binding” refers to the binding of an antibody to an epitope of an antigen (human VEGF, ANG-2, TGF-beta, TIM-3 ligand or PEDF-R) in an in vitro assay. Mention.
용어 "에피토프" 는 항체에 특이적으로 결합할 수 있는 임의의 폴리펩티드 결정인자를 포함한다. 특정 구현예에서, 에피토프 결정인자는 아미노산, 당 측쇄, 포스포릴 또는 술포닐과 같은 분자의 화학적 활성 표면기를 포함하고, 특정 구현예에서 특이적 3 차원적 구조 특성 및 또는 특이적 전하 특성을 가질 수 있다. 에피토프는 항체에 의해 결합되는 항원의 영역이다.The term “epitope” includes any polypeptide determinant capable of specifically binding to an antibody. In certain embodiments, epitope determinants include amino acids, sugar side chains, chemically active surface groups of molecules such as phosphoryl or sulfonyl, and, in certain embodiments, may have specific three-dimensional structural properties and/or specific charge properties. there is. An epitope is a region of an antigen that is bound by an antibody.
본원에 사용된 용어 "링커" 는, 바람직하게는 합성 기원인 아미노산 서열을 갖는 펩티드를 의미한다. 일반적으로 상기 펩티드는 a) VH-CH1를 VL-CL에, b) VL-CL를 VH-CH1에, c) VH-CL 를 VL-CH1에, 또는 d)VL-CH1 를 VH-CL에 연결하며, 본 발명에 따른 링커는 인간 immunoglobulin G4와 인간 VEGFR1 domain 2, TGF-베타 RII ECD, TIM-3 ECD, 및 상기 44-머(mer)의 PEDF 서열을 연결하는데 사용된다. As used herein, the term “linker” refers to a peptide having an amino acid sequence, preferably of synthetic origin. Typically, the peptides link a) VH-CH1 to VL-CL, b) VL-CL to VH-CH1, c) VH-CL to VL-CH1, or d) VL-CH1 to VH-CL. The linker according to the present invention is used to connect human immunoglobulin G4 and human VEGFR1 domain 2, TGF-beta RII ECD, TIM-3 ECD, and the PEDF sequence of the 44-mer.
본원에 사용된 "약학적 담체" 는 생리적으로 적합한 임의의 모든 용매, 분산 매질, 항박테리아 및 항진균제, 등장성 및 흡수 지연제 등을 포함한다. 바람직하게는, 담체는 (예를 들어 주사 또는 주입에 의한) 정맥내, 근육내, 피하, 비경구적, 척수 또는 표피 투여에 적합하다.As used herein, “pharmaceutical carrier” includes any and all physiologically compatible solvents, dispersion media, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. Preferably, the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g. by injection or infusion).
본 발명의 조성물은 당업계에 알려진 다양한 방법에 의해 투여될 수 있다. 당업자에 의해 이해될 바와 같이, 투여 경로 및/또는 방식은 원하는 결과에 따라 다를 것이다. 특정 투여 경로에 의해 본 발명의 항체를 투여하기 위해, 그의 불활성화를 방지할 물질로 항체를 코팅하거나 그와 함께 항체를 공동투여하는 것이 필요할 수 있다. 예를 들어, 항체는 적절한 담체, 예를 들어 리포좀 또는 희석제 내에서 대상에게 투여될 수 있다. 약학적으로 허용되는 희석제는 식염수 및 수성 완충 용액을 포함한다. 약학적 담체는 멸균 수용액 또는 분산물 및, 멸균 주사액 또는 분산물의 즉석 제조를 위한 멸균 분말을 포함한다. 약학적 활성 성분을 위한 상기 매질 또는 약제의 사용이 당업계에 알려져 있다.The composition of the present invention can be administered by various methods known in the art. As will be appreciated by those skilled in the art, the route and/or mode of administration will vary depending on the desired results. To administer an antibody of the invention by a particular route of administration, it may be necessary to coat the antibody with or co-administer the antibody with a material that will prevent its inactivation. For example, the antibody can be administered to a subject in a suitable carrier, such as liposomes or a diluent. Pharmaceutically acceptable diluents include saline and aqueous buffered solutions. Pharmaceutical carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. The use of such media or agents for pharmaceutically active ingredients is known in the art.
본원에 사용된 구절 "비경구적 투여" 및 "비경구적으로 투여되는" 은 통상적으로 주사에 의한, 장 및 국소 투여를 제외한 투여 방식을 의미하고, 정맥내, 근육내, 동맥내, 척추강내, 관절낭내, 안와내, 심장내, 진피내, 복강내, 기관내, 피하, 표피내, 관절강내, 피막하, 지주막하, 척수내, 경막외 및 흉골내 주사 및 주입을 제한 없이 포함한다.As used herein, the phrases “parenteral administration” and “administered parenterally” refer to any mode of administration other than enteral and topical administration, typically by injection, including intravenous, intramuscular, intraarterial, intrathecal, and intraarticular. Including, without limitation, intraorbital, intracardiac, intradermal, intraperitoneal, intratracheal, subcutaneous, intraepithelial, intraarticular, subcapsular, subarachnoid, intrathecal, epidural, and intrasternal injections and infusions.
본원에 사용된 용어 암은, 증식형 질환, 예컨대 림프종, 림프구성 백혈병, 폐암, 비 소세포 폐 (NSCL) 암, 세기관지 폐포 세포 폐암, 뼈암, 췌장암, 피부암, 두경부암, 피부 및 안내 흑색종, 자궁암, 난소암, 직장암, 항문영역의 암, 위암, 위장관 암, 결장암, 유방암, 자궁암, 난관 암종, 내막 암종, 자궁경부암, 질 암종, 외음부 암종, 호지킨병, 식도암, 소장암, 내분비계 암, 갑상선 암, 부갑상선 암, 부신 암, 연부육종, 요도암, 음경 암,전립선암, 방광 암, 신장 또는 요도 암, 신세포암, 신우암, 중피종, 간세포암, 담도암, 중추신경계 (CNS)의 종양, 척추 종양, 뇌간 종양, 다형성교아세포종, 성상세포종, 신경초종, 상의세포종, 수모세포종, 수막종, 편평세포암종, 뇌하수체 선종 및 유잉 육종을 의미하고, 상기 암 중 임의의 불응성 버젼 또는 상기 암 중 하나 이상의 조합을 포함한다.As used herein, the term cancer refers to proliferative diseases such as lymphoma, lymphocytic leukemia, lung cancer, non-small cell lung (NSCL) cancer, bronchioloalveolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, skin and intraocular melanoma, and uterine cancer. , ovarian cancer, rectal cancer, anal area cancer, stomach cancer, gastrointestinal cancer, colon cancer, breast cancer, uterine cancer, fallopian tube carcinoma, endometrial carcinoma, cervical cancer, vaginal carcinoma, vulvar carcinoma, Hodgkin's disease, esophageal cancer, small intestine cancer, endocrine cancer, Thyroid cancer, parathyroid cancer, adrenal cancer, soft sarcoma, urethral cancer, penile cancer, prostate cancer, bladder cancer, kidney or urethral cancer, renal cell cancer, renal pelvis cancer, mesothelioma, hepatocellular cancer, biliary tract cancer, and tumors of the central nervous system (CNS) , spine tumor, brainstem tumor, glioblastoma multiforme, astrocytoma, schwannoma, ependymoma, medulloblastoma, meningioma, squamous cell carcinoma, pituitary adenoma and Ewing's sarcoma, and a refractory version of any of the above cancers or one of the above cancers. Includes combinations of the above.
본 발명의 또다른 양상은 항-혈관생성 물질로서의 본 발명에 따른 이중특이성 항체 또는 상기 약학적 조성물이다. 상기 항-혈관생성 물질은 암, 특히 고형 종양, 및 다른 혈관 질환의 치료를 위해 사용될 수 있다.Another aspect of the invention is the bispecific antibody or said pharmaceutical composition according to the invention as an anti-angiogenic agent. The anti-angiogenic substances can be used for the treatment of cancer, especially solid tumors, and other vascular diseases.
본 발명의 하나의 구현예는 혈관 질환 치료를 위한 본 발명에 따른 이중특이성 항체이다.One embodiment of the invention is a bispecific antibody according to the invention for the treatment of vascular diseases.
본 발명의 또다른 양상은 혈관 질환 치료를 위한 상기 약학적 조성물이다.Another aspect of the present invention is the pharmaceutical composition for treating vascular diseases.
본 발명의 또다른 양상은 혈관 질환 치료용 약제의 제조를 위한 본 발명에 따른 항체의 용도이다.Another aspect of the invention is the use of the antibody according to the invention for the manufacture of a medicament for the treatment of vascular diseases.
본 발명의 또다른 양상은 치료를 필요로 하는 환자에게 본 발명에 따른 항체를 투여하는 것에 의한 혈관 질환을 겪는 환자의 치료 방법이다.Another aspect of the invention is a method of treating a patient suffering from vascular disease by administering an antibody according to the invention to the patient in need of treatment.
용어 "혈관 질환" 은 암, 염증성 질환, 죽상경화증, 국소성 빈혈, 트라우마, 패혈증, COPD, 천식, 당뇨병, AMD,망막병증, 뇌졸중, 지방과다증, 급성 폐 손상, 출혈, 예를 들어 사이토카인 유도된 혈관 유출, 알레르기, 그레이브병, 하시모토 자가면역 갑상샘염, 특발성 혈소판감소성 자반병, 거대 세포 동맥염, 류마티스 관절염, 전신홍반 루푸스 (SLE), 루푸스 신장염, 크론병, 다발성 경화증, 궤양성 결장염, 특히 고형 종양에 대한 것, 안구내 신생혈관 증후군 예컨대 증식성 망막병증 또는 연령관련 황반 변성 (AMD), 류마티스 관절염 및 건선을 포함한다(Folkman, J., et al., J. Biol. Chem. 267 (1992) 10931-10934; Klagsbrun, M., et al., Annu. Rev.Physiol. 53 (1991) 217-239; 및 Garner, A., Vascular diseases, In: Pathobiology of ocular disease, A dynamic approach, Garner, A., and Klintworth, G.K., (eds.), 2nd edition, Marcel Dekker, New York (1994), pp 1625-1710).The term “vascular disease” includes cancer, inflammatory disease, atherosclerosis, ischemia, trauma, sepsis, COPD, asthma, diabetes, AMD, retinopathy, stroke, adiposity, acute lung injury, hemorrhage, e.g. cytokine-induced Vascular effusion, allergy, Graves' disease, Hashimoto's autoimmune thyroiditis, idiopathic thrombocytopenic purpura, giant cell arteritis, rheumatoid arthritis, systemic lupus erythematosus (SLE), lupus nephritis, Crohn's disease, multiple sclerosis, ulcerative colitis, especially solid tumors. For, intraocular neovascular syndromes such as proliferative retinopathy or age-related macular degeneration (AMD), rheumatoid arthritis and psoriasis (Folkman, J., et al., J. Biol. Chem. 267 (1992) 10931-10934; Klagsbrun, M., et al., Rev. Physiol. 53 (1991) 217-239; and Garner, A., Pathobiology of ocular disease, A dynamic approach. ., and Klintworth, G.K., (eds.), 2nd edition, Marcel Dekker, New York (1994), pp 1625-1710).
상기 조성물은 보존제, 습윤제, 유화제 및 분산제와 같은 아쥬반트를 또한 포함할 수 있다. 미생물 존재의 방지는 상기 멸균 절차에 의해, 및 다양한 항박테리아 및 항진균제, 예를 들어 파라벤, 클로로부탄올, 페놀, 소르브산 등의 포함에 의해 보장될 수 있다. 당, 염화나트륨 등과 같은 등장성 물질을 조성물 내로 포함시키는 것이 또한 바람직할 수 있다. 또한, 주사가능한 약학적 형태의 지속된 흡수는 알루미늄 모노스테아레이트 및 젤라틴과 같이 흡수를 지연시키는 물질의 포함에 의해 수득될 수 있다.The composition may also include adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the presence of microorganisms can be ensured by the above sterilization procedures and by the inclusion of various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid, etc. It may also be desirable to include isotonic substances such as sugars, sodium chloride, etc. into the composition. Additionally, sustained absorption of injectable pharmaceutical forms can be obtained by the inclusion of substances that delay absorption, such as aluminum monostearate and gelatin.
선택된 투여 경로와 관계없이, 본 발명의 적합한 수화된 형태 및/또는 약학적 조성물로 사용될 수 있는 본 발명의 항체는 당업자에게 알려진 종래의 방법에 의해 약학적으로 허용가능한 투여 형태로 제형화된다.Regardless of the route of administration chosen, the antibodies of the invention, which can be used in suitable hydrated forms and/or pharmaceutical compositions of the invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those skilled in the art.
본 발명의 약학적 조성물 내 활성 성분의 실제 투여량 수준은, 환자에게 독성이지 않으면서, 특정 환자, 조성물 및 투여 방식에 대한 바람직한 치료 반응을 달성하는데 효과적인 활성 성분의 양을 수득하도록 다를 수 있다. 선택된 투여량 수준은 이용된 본 발명의 특정 조성물의 활성, 투여 경로, 투여 시간, 이용될 특정 항체의 배설 속도, 치료 지속시간, 이용될 특정 조성물과 병용될 다른 약물, 화합물 및/또는 물질, 치료될 환자의 연령, 성별, 체중, 상태, 전반적 건강 및 과거 병력 등, 의약 분야에서 잘 알려진 인자들을 포함하는 다양한 약동학적 인자들에 따라 다를 것이다.The actual dosage levels of the active ingredient in the pharmaceutical compositions of the invention may vary to obtain an amount of active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, while not being toxic to the patient. The dosage level selected will depend on the activity of the particular composition of the invention employed, the route of administration, the time of administration, the rate of excretion of the particular antibody to be employed, the duration of treatment, the other drugs, compounds and/or substances to be combined with the particular composition to be employed, the treatment. It will vary depending on various pharmacokinetic factors, including factors well known in the medical field, such as the patient's age, gender, weight, condition, general health, and past medical history.
본 발명의 조성물은 주사기에 의해 조성물이 전달가능할 정도로 유체이고 살균되어야 한다. 물에 더하여, 담체는 바람직하게는 등장성 완충 식염수이다. 적절한 유동성은 예를 들어 레시틴과 같은 코팅의 사용에 의해, 분산의 경우 필요한 입자 크기의 유지에 의해, 및 계면활성제의 사용에 의해 유지될 수 있다. 많은 경우, 등장성 물질, 예를 들어 당, 다가알콜, 예컨대 만니톨 또는 소르비톨, 및 염화나트륨을 조성물 내에 포함하는 것이 바람직하다.The compositions of the present invention must be fluid and sterile to the extent that the compositions can be delivered by syringe. In addition to water, the carrier is preferably isotonic buffered saline. Adequate fluidity can be maintained, for example, by the use of coatings such as lecithin, by maintenance of the required particle size in case of dispersion, and by the use of surfactants. In many cases, it is desirable to include isotonic substances such as sugars, polyhydric alcohols such as mannitol or sorbitol, and sodium chloride in the composition.
본 발명은 Ang2 및 VEGF, TGF-베타, TIM-3 리간드를 저해 또는 PEDF-R을 활성화하면서 동시에 Tie2 수용체를 활성화시켜 하류 신호 전달을 촉진하는 항체를 제안함으로써, 신생혈관형성을 저해하고 혈관 투과성을 감소시킬 수 있는 새로운 방법을 제시한다. 또한 본 발명에서 제안되는 항체는 암 및 비정상적 혈관 형성관련 질환 및/또는 혈관 투과성이 증가되어 유발되는 질환의 진단 및 치료에의 응용 가능성이 기대된다.The present invention proposes an antibody that promotes downstream signaling by inhibiting Ang2, VEGF, TGF-beta, and TIM-3 ligands or activating PEDF-R while simultaneously activating the Tie2 receptor, thereby inhibiting neovascularization and improving vascular permeability. Suggests a new way to reduce it. In addition, the antibody proposed in the present invention is expected to have applicability in the diagnosis and treatment of cancer, diseases related to abnormal angiogenesis, and/or diseases caused by increased vascular permeability.
도 1은 본 발명에서 개발한 인간 안지오포이에틴-2에 특이적으로 결합하는 제1 항원 결합 부위를 포함하는 이중특이적 항체의 모식도이다.Figure 1 is a schematic diagram of a bispecific antibody comprising a first antigen binding site that specifically binds to human angiopoietin-2 developed in the present invention.
도 2는 항 Ang2/VEGF 항체를 정제한 SDS-PAGE 결과이다.Figure 2 shows the SDS-PAGE results of purified anti-Ang2/VEGF antibody.
도 3 및 4는 각각 항 Ang2/TGF-β 항체를 정제한 SDS-PAGE 결과(도 3) 및 제 2 항원 결합 부위인 TGF-β binding region (도 4)을 Western blot으로 확인한 결과이다.Figures 3 and 4 show the results of SDS-PAGE purified anti-Ang2/TGF-β antibody (Figure 3) and the results of Western blot confirmation of the TGF-β binding region, the second antigen binding site (Figure 4), respectively.
도 5 및 6은 각각 항 Ang2/VEGF 항체(도 5. 2C8-D2, 도 6. 1D3-D2)의 인간 Ang2에 대한 결합력을 분석한 ELISA 결과이다.Figures 5 and 6 show ELISA results analyzing the binding affinity of anti-Ang2/VEGF antibodies (Figure 5: 2C8-D2, Figure 6: 1D3-D2) to human Ang2, respectively.
도 7 및 8은 각각 항 Ang2/VEGF 항체(도 7. 2C8-D2, 도 8. 1D3-D2)의 인간 VEGF 또는 마우스 VEGF에 대한 결합력을 보여주는 ELISA 결과이다.Figures 7 and 8 are ELISA results showing the binding ability of anti-Ang2/VEGF antibody (Figure 7, 2C8-D2, Figure 8, 1D3-D2) to human VEGF or mouse VEGF, respectively.
도 9는 항 Ang2/VEGF 항체가 VEGF와 Ang2에 동시에 결합할 수 있다는 것을 보여주는 ELISA 결과이다.Figure 9 shows ELISA results showing that anti-Ang2/VEGF antibody can bind to VEGF and Ang2 simultaneously.
도 10은 항 Ang2/VEGF 항체 및 항 Ang2/TGF-β 항체가 Ang2와 함께 HUVEC의 AKT와 ERK 42/44를 인산화 시키는 것을 보여주는 Western blot 결과이다.Figure 10 is a Western blot result showing that anti-Ang2/VEGF antibody and anti-Ang2/TGF-β antibody phosphorylate AKT and ERK 42/44 in HUVEC along with Ang2.
도 11은 항 Ang2/VEGF 항체가 soluble VEGFR2와 경쟁적으로 VEGF와 결합하는 것을 보여주는 결과이다.Figure 11 is a result showing that anti-Ang2/VEGF antibody binds to VEGF competitively with soluble VEGFR2.
도 12는 항 Ang2/VEGF 항체가 VEGF에 의한 HUVEC proliferation을 저해하는 결과이다.Figure 12 shows the results of anti-Ang2/VEGF antibody inhibiting HUVEC proliferation caused by VEGF.
도 13 내지 15는 각각 마우스 laser-induced CNV (wet AMD) model에서 항 Ang2/VEGF 항체의 신생혈관 형성 억제 (도 13. representative FFA images & CTF, 도 14. representative OCT images & CNV lesion) 및 시력 개선 효과 (도 15. ERG)를 나타내는 결과이다.Figures 13 to 15 show the inhibition of neovascularization by anti-Ang2/VEGF antibodies in the mouse laser-induced CNV (wet AMD) model (Figure 13. representative FFA images & CTF, Figure 14. representative OCT images & CNV lesion) and improvement in vision, respectively. This is a result showing the effect (Figure 15. ERG).
이하에서는 실시예를 들어 본 발명을 더욱 구체적으로 설명하고자 하나, 이는 예시적인 것에 불과할 뿐 본 발명의 범위를 제한하고자 함이 아니다. 아래 기재된 실시예들은 발명의 본질적인 요지를 벗어나지 않는 범위에서 변형될 수 있음은 당 업자들에게 있어 자명하다.Hereinafter, the present invention will be described in more detail through examples, but these are merely illustrative and are not intended to limit the scope of the present invention. It is obvious to those skilled in the art that the embodiments described below can be modified without departing from the essential gist of the invention.
실시예 1: 인간 안지오포이에틴-2에 특이적으로 결합하는 제1 항원 결합 부위를 포함하는 이중특이적 항체의 제작Example 1: Construction of a bispecific antibody comprising a first antigen binding site that specifically binds to human angiopoietin-2
인간 안지오포이에틴-2에 특이적으로 결합하는 제1 항원 결합 부위를 포함하는 이중특이적 항체를 만들기 위해 Ang2를 이용하여 Tie2 수용체를 활성화시켜 주는 것으로 알려진 항 Ang2 항체 (대한민국 특허 출원번호 10-2022-0028751)와 인간 VEGFR1 (서열번호 19, Accession: P17948)의 VEGF binding domain인 D2 또는 TGF-βextracellular domain (서열번호 24) 또는 인간 TIM-3 extracellular domain (서열번호 27) 또는 인간 PEDF 44-mer peptide (서열번호 30)를 활용하였다.An anti-Ang2 antibody known to activate the Tie2 receptor using Ang2 to create a bispecific antibody containing a first antigen binding site that specifically binds to human angiopoietin-2 (Korean Patent Application No. 10- 2022-0028751) and D2, the VEGF binding domain of human VEGFR1 (SEQ ID NO: 19, Accession: P17948) or TGF-βextracellular domain (SEQ ID NO: 24) or human TIM-3 extracellular domain (SEQ ID NO: 27) or human PEDF 44-mer Peptide (SEQ ID NO: 30) was used.
항 Ang2/VEGF 항체의 heavy chain은 대한민국 특허 출원번호 10-2022-0028751에 기술되어 있는 1A9 또는 2C8 또는 1D3의 heavy chain CDR sequence (서열번호 1 내지 3 또는 7 내지 9 또는 13 내지 15) 및 C-terminal의 lysine을 alanine으로 치환한 인간 immunoglobulin G4 heavy chain sequence 및 linker sequence (서열번호 20) 및 VEGFR1 D2 sequence (서열번호 21)를 하나로 구성한 후, GenScript에서 합성하였다. 항 Ang2/VEGF 항체의 light chain은 대한민국 특허 출원번호 10-2022-0028751에 기술되어 있는 1A9 또는 2C8 또는 1D3의 light chain CDR sequence (서열번호 4 내지 6 또는 10 내지 12 또는 16 내지 18)을 이용하였다.The heavy chain of the anti-Ang2/VEGF antibody is a heavy chain CDR sequence of 1A9 or 2C8 or 1D3 (SEQ ID NOs: 1 to 3 or 7 to 9 or 13 to 15) and C- The human immunoglobulin G4 heavy chain sequence and linker sequence (SEQ ID NO: 20) and VEGFR1 D2 sequence (SEQ ID NO: 21), in which terminal lysine was replaced with alanine, were composed into one and synthesized in GenScript. The light chain of the anti-Ang2/VEGF antibody used was the light chain CDR sequence (SEQ ID NO: 4 to 6 or 10 to 12 or 16 to 18) of 1A9 or 2C8 or 1D3 described in Korean Patent Application No. 10-2022-0028751. .
항 Ang2/TGF-β 항체의 heavy chain은 대한민국 특허 출원번호 10-2022-0028751에 기술되어 있는 1A9 또는 2C8 또는 1D3의 heavy chain CDR sequence (서열번호 1 내지 3 또는 7 내지 9 또는 13 내지 15) 및 C-terminal의 lysine을 alanine으로 치환한 인간 immunoglobulin G4 heavy chain sequence 및 linker sequence (서열번호 23) 및 TGF-βextracellular domain (서열번호 24)을 하나로 구성한 후, GenScript에서 합성하였다. 항 Ang2/TGF-β 항체의 light chain은 대한민국 특허 출원번호 10-2022-0028751에 기술되어 있는 1A9 또는 2C8 또는 1D3의 light chain CDR sequence (서열번호 4 내지 6 또는 10 내지 12 또는 16 내지 18)을 이용하였다.The heavy chain of the anti-Ang2/TGF-β antibody is the heavy chain CDR sequence of 1A9 or 2C8 or 1D3 (SEQ ID NOs: 1 to 3 or 7 to 9 or 13 to 15) described in Korean Patent Application No. 10-2022-0028751. The human immunoglobulin G4 heavy chain sequence and linker sequence (SEQ ID NO: 23), in which lysine at the C-terminal was replaced with alanine, and the TGF-βextracellular domain (SEQ ID NO: 24) were constructed as one, and then synthesized in GenScript. The light chain of the anti-Ang2/TGF-β antibody has the light chain CDR sequence (SEQ ID NO: 4 to 6 or 10 to 12 or 16 to 18) of 1A9 or 2C8 or 1D3 described in Korean Patent Application No. 10-2022-0028751. used.
항 Ang2/TIM-3 ligand 항체의 heavy chain은 대한민국 특허 출원번호 10-2022-0028751에 기술되어 있는 1A9 또는 2C8 또는 1D3의 heavy chain CDR sequence (서열번호 1 내지 3 또는 7 내지 9 또는 13 내지 15) 및 C-terminal의 lysine을 alanine으로 치환한 인간 immunoglobulin G4 heavy chain sequence 및 linker sequence (서열번호 26) 및 인간 TIM-3 extracellular domain (서열번호 27)을 하나로 구성한 후, GenScript에서 합성하였다. 항 Ang2/TIM-3 ligand 항체의 light chain은 대한민국 특허 출원번호 10-2022-0028751에 기술되어 있는 1A9 또는 2C8 또는 1D3의 light chain CDR sequence (서열번호 4 내지 6 또는 10 내지 12 또는 16 내지 18)을 이용하였다.The heavy chain of the anti-Ang2/TIM-3 ligand antibody is the heavy chain CDR sequence of 1A9 or 2C8 or 1D3 (SEQ ID NOs: 1 to 3 or 7 to 9 or 13 to 15) described in Korean Patent Application No. 10-2022-0028751. And the human immunoglobulin G4 heavy chain sequence and linker sequence (SEQ ID NO: 26), in which lysine at the C-terminal was replaced with alanine, and the human TIM-3 extracellular domain (SEQ ID NO: 27) were composed into one, and then synthesized in GenScript. The light chain of the anti-Ang2/TIM-3 ligand antibody is the light chain CDR sequence of 1A9 or 2C8 or 1D3 (SEQ ID NOs: 4 to 6 or 10 to 12 or 16 to 18) described in Korean Patent Application No. 10-2022-0028751. was used.
항 Ang2/PEDF-R 항체의 heavy chain은 대한민국 특허 출원번호 10-2022-0028751에 기술되어 있는 1A9 또는 2C8 또는 1D3의 heavy chain CDR sequence (서열번호 1 내지 3 또는 7 내지 9 또는 13 내지 15) 및 C-terminal의 lysine을 alanine으로 치환한 인간 immunoglobulin G4 heavy chain sequence 및 linker sequence (서열번호 29) 및 인간 PEDF 44-mer peptide (서열번호 30)을 하나로 구성한 후, GenScript에서 합성하였다. 항 Ang2/PEDF-R 항체의 light chain은 대한민국 특허 출원번호 10-2022-0028751에 기술되어 있는 1A9 또는 2C8 또는 1D3의 light chain CDR sequence (서열번호 4 내지 6 또는 10 내지 12 또는 16 내지 18)을 이용하였다.The heavy chain of the anti-Ang2/PEDF-R antibody is the heavy chain CDR sequence of 1A9 or 2C8 or 1D3 (SEQ ID NOs: 1 to 3 or 7 to 9 or 13 to 15) described in Korean Patent Application No. 10-2022-0028751, and The human immunoglobulin G4 heavy chain sequence and linker sequence (SEQ ID NO: 29), in which lysine at the C-terminal was replaced with alanine, and the human PEDF 44-mer peptide (SEQ ID NO: 30) were combined into one, and then synthesized in GenScript. The light chain of the anti-Ang2/PEDF-R antibody has the light chain CDR sequence (SEQ ID NO: 4 to 6 or 10 to 12 or 16 to 18) of 1A9 or 2C8 or 1D3 described in Korean Patent Application No. 10-2022-0028751. used.
DomainDomain CDRCDR CDR sequenceCDR sequence 서열번호sequence number
Heavy chainHeavy chain CDRH1-KabatCDRH1-Kabat DYWIGDYWIG 서열번호 1SEQ ID NO: 1
CDRH2-KabatCDRH2-Kabat DIYPGGGYTNCNEKFKGDIYPGGGYTNCNEKFKG 서열번호 2SEQ ID NO: 2
CDRH3-KabatCDRH3-Kabat SDYRNDEGFADSDYRNDEGFAD 서열번호 3SEQ ID NO: 3
Light chainLight chain CDRL1-KabatCDRL1-Kabat SASSSVSSSYLHSASSSVSSSYLH 서열번호 4SEQ ID NO: 4
CDRL2-KabatCDRL2-Kabat RTSNLASRTSNLAS 서열번호 5SEQ ID NO: 5
CDRL3-KabatCDRL3-Kabat QQWSGYPYTQQWSGYPYT 서열번호 6SEQ ID NO: 6
표 1은 마우스 항 Ang2 항체 1A9의 CDR 서열Table 1 is the CDR sequence of mouse anti-Ang2 antibody 1A9
ProteinProtein CDRCDR CDR sequenceCDR sequence 서열번호sequence number
Heavy chainHeavy chain CDRH1-KabatCDRH1-Kabat NYWIGNYWIG 서열번호 7SEQ ID NO: 7
CDRH2-KabatCDRH2-Kabat IYPGGGYTNYNEKFKIYPGGGYTNYNEKFK 서열번호 8SEQ ID NO: 8
CDRH3-KabatCDRH3-Kabat SDYRDDEGFAYSDYRDDEGFAY 서열번호 9SEQ ID NO: 9
Light chainLight chain CDRL1-KabatCDRL1-Kabat SASSSVSSSYLHSASSSVSSSYLH 서열번호 10SEQ ID NO: 10
CDRL2-KabatCDRL2-Kabat RTSNLASRTSNLAS 서열번호 11SEQ ID NO: 11
CDRL3-KabatCDRL3-Kabat QQWSGYPYTQQWSGYPYT 서열번호 12SEQ ID NO: 12
표 2는 항 Ang2 항체 2C8의 CDR 서열Table 2 shows the CDR sequence of anti-Ang2 antibody 2C8
ProteinProtein CDRCDR CDR sequenceCDR sequence 서열번호sequence number
Heavy chainHeavy chain CDRH1-KabatCDRH1-Kabat DYGWNDYGWN 서열번호 13SEQ ID NO: 13
CDRH2-KabatCDRH2-Kabat ISYSGTTSYNPSLKISYSGTTSYNPSLK 서열번호 14SEQ ID NO: 14
CDRH3-KabatCDRH3-Kabat SEGTGFYAMDYSEGTGFYAMDY 서열번호 15SEQ ID NO: 15
Light chainLight chain CDRL1-KabatCDRL1-Kabat KASQSVSNDVAKASQSVSNDVA 서열번호 16SEQ ID NO: 16
CDRL2-KabatCDRL2-Kabat YASNRYTYASNRYT 서열번호 17SEQ ID NO: 17
CDRL3-KabatCDRL3-Kabat QQDYSSPTQQDYSSPT 서열번호 18SEQ ID NO: 18
표 3은 항 Ang2 항체 1D3의 CDR 서열Table 3 shows the CDR sequence of anti-Ang2 antibody 1D3
ProteinProtein Amino acid sequenceAmino acid sequence 서열번호sequence number
Human VEGFR1Human VEGFR1 DLAARNILLSENNVVKICDFGLARDIYKNPDYVRKGDTRLPLKWMAPESIFDKIYSTKSDVWSYGVLLWEIFSLGGSPYPGVQMDEDFCSRLREGMRMRAPEYSTPEIYQIMLDCWHRDPKERPRFAELVEKLGDLLQANVQQDGKDYIPINAILTGNSGFTYSTPAFSEDFFKESISAPKFNSGSSDDVRYVNAFKFMSLERIKTFEELLPNATSMFDDYQGDSSTLLASPMLKRFTWTDSKPKASLKIDLRVTSKSKESGLSDVSRPSFCHSSCGHVSEGKRRFTYDHAELERKIACCSPPPDYNSVVLYSTPPIDLAARNILLSENNVVKICDFGLARDIYKNPDYVRKGDTRLPLKWMAPESIFDKIYSTKSDVWSYGVLLWEIFSLGGSPYPGVQMDEDFCSRLREGMRAPEYSTPEIYQIMLDCWHRDPKERPRFAELVEKLGDLLQANVQQDGKDYIPINAILTGNSGFTYSTPAFSEDFFKESISAPKFNSGSSDDVRYVNAFKFMSLERIKT FEELLPNATSMFDDYQGDSSTLLASPMLKRFTWTDSKPKASLKIDLRVTSKSKESGLSDVSRPSFCHSSCGHVSEGKRRFTYDHAELERKIACCSPPPDYNSVVLYSTPPI 서열번호 19SEQ ID NO: 19
Human IgG4와 VEGFR1 D2를 연결하는 linkerLinker connecting Human IgG4 and VEGFR1 D2 GGGGSGGGGGGGGSGGGG 서열번호 20SEQ ID NO: 20
Human VEGFR1 D2를 포함한 (129-229) 서열(129-229) sequence including Human VEGFR1 D2 SDTGRPFVEMYSEKPEIIHMTEGRELVIPCRVTSPNITVTLKKFPLDTLIPDGKRIIWDSRKGFIISNATYKEIGLLTCEATVNGHLYKTNYLTHRQTNTISDTGRPFVEMYSEKPEIIHMTEGRELVIPCRVTSPNITVTLKKFPLDTLIPDGKRIIWDSRKGFIISNATYKEIGLLTCEATVNGHLYKTNYLTHRQTNTI 서열번호 21SEQ ID NO: 21
표 4는 인간 immunoglobulin G4와 인간 VEGFR1 domain 2를 포함한 (129-229) 서열을 연결하는 linker 및 인간 VEGFR1 domain 2를 포함한 (129-229) 서열, Table 4 shows the linker connecting human immunoglobulin G4 and the (129-229) sequence containing human VEGFR1 domain 2, and the (129-229) sequence containing human VEGFR1 domain 2,
ProteinProtein Amino acid sequenceAmino acid sequence 서열번호sequence number
Human TGF-β RII isoform BHuman TGF-β RII isoform B MGRGLLRGLWPLHIVLWTRIASTIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPDLLLVIFQVTGISLLPPLGVAISVIIIFYCYRVNRQQKLSSTWETGKTRKLMEFSEHCAIILEDDRSDISSTCANNINHNTELLPIELDTLVGKGRFAEVYKAKLKQNTSEQFETVAVKIFPYEEYASWKTEKDIFSDINLKHENILQFLTAEERKTELGKQYWLITAFHAKGNLQEYLTRHVISWEDLRKLGSSLARGIAHLHSDHTPCGRPKMPIVHRDLKSSNILVKNDLTCCLCDFGLSLRLDPTLSVDDLANSGQVGTARYMAPEVLESRMNLENVESFKQTDVYSMALVLWEMTSRCNAVGEVKDYEPPFGSKVREHPCVESMKDNVLRDRGRPEIPSFWLNHQGIQMVCETLTECWDHDPEARLTAQCVAERFSELEHLDRLSGRSCSEEKIPEDGSLNTTKMGRGLLRGLWPLHIVLWTRIASTIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPDLLLVIFQVTGISLLPPLGVAISVIIIFYCYRVNRQQKLSSTWETGKTRKLMEF SEHCAIILEDDRSDISSTCANNINHNTELLPIELDTLVGKGRFAEVYKAKLKQNTSEQFETVAVKIFPYEEYASWKTEKDIFSDINLKHENILQFLTAEERKTELGKQYWLITAFHAKGNLQEYLTRHVISWEDLRKLGSSLARGIAHLHSDHTPCGRPKMPIVHRDLKSSNILVKNDLTCCLCDFGLSLRLDPTLSVDDLANSGQVGTARYMAP EVLESRMNLENVESFKQTDVYSMALVLWEMTSRCNAVGEVKDYEPPFGSKVREHPCVESMKDNVLRDRGRPEIPSFWLNHQGIQMVCETLTECWDHDPEARLTAQCVAERFSELEHLDRLSGRSCSEEKIPEDGSLNTTK 서열번호 22SEQ ID NO: 22
Human IgG4와 Human TGF-β RII extracellular domain을 연결하는 linkerLinker connecting human IgG4 and human TGF-β RII extracellular domain GGGGSGGGGSGGGGSGGGGSGGGGGSGGGGSGGGGSGGGGSG 서열번호 23SEQ ID NO: 23
Human TGF-β RII isoform B extracellular domain (23-159) 서열Human TGF-β RII isoform B extracellular domain (23-159) sequence TIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPDTIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPD 서열번호 24SEQ ID NO: 24
표 5는 인간 immunoglobulin G4와 인간 TGF-βRII extracellular domain 서열을 연결하는 linker 및 인간 TGF-βRII extracellular domain 서열, Table 5 shows the linker connecting human immunoglobulin G4 and human TGF-βRII extracellular domain sequence and human TGF-βRII extracellular domain sequence,
ProteinProtein Amino acid sequenceAmino acid sequence 서열번호sequence number
Human TIM-3Human TIM-3 MFSHLPFDCVLLLLLLLLTRSSEVEYRAEVGQNAYLPCFYTPAAPGNLVPVCWGKGACPVFECGNVVLRTDERDVNYWTSRYWLNGDFRKGDVSLTIENVTLADSGIYCCRIQIPGIMNDEKFNLKLVIKPAKVTPAPTRQRDFTAAFPRMLTTRGHGPAETQTLGSLPDINLTQISTLANELRDSRLANDLRDSGATIRIGIYIGAGICAGLALALIFGALIFKWYSHSKEKIQNLSLISLANLPPSGLANAVAEGIRSEENIYTIEENVYEVEEPNEYYCYVSSRQQPSQPLGCRFAMPMFSHLPFDCVLLLLLLLLTRSSEVEYRAEVGQNAYLPCFYTPAAPGNLVPVCWGKGACPVFECGNVVLRTDERDVNYWTSRYWLNGDFRKGDVSLTIENVTLADSGIYCCRIQIPGIMNDEKFNLKLVIKPAKVTPAPTRQRDFTAAFPRMLTTRGHGPAETQTLGSLPDINLTQISTLANELRDSRLANDLRDSGATIRIGIYIGAGICAGL ALALIFGALIFKWYSHSKEKIQNLSLISLANLPPSGLANAVAEGIRSEENIYTIEENVYEVEEPNEYYCYVSSRQQPSQPLGCRFAMP 서열번호 25SEQ ID NO: 25
Human IgG4와 Human TIM-3 extracellular domain을 연결하는 linkerLinker connecting human IgG4 and human TIM-3 extracellular domain GGGGSGGGGSGGGGSGGGGGGGGSGGGGSGGGGSGGGG 서열번호 26SEQ ID NO: 26
Human TIM-3 extracellular domain (22-200) 서열Human TIM-3 extracellular domain (22-200) sequence SEVEYRAEVGQNAYLPCFYTPAAPGNLVPVCWGKGACPVFECGNVVLRTDERDVNYWTSRYWLNGDFRKGDVSLTIENVTLADSGIYCCRIQIPGIMNDEKFNLKLVIKPAKVTPAPTRQRDFTAAFPRMLTTRGHGPAETQTLGSLPDINLTQISTLANELRDSRLANDLRDSGATIRSEVEYRAEVGQNAYLPCFYTPAAPGNLVPVCWGKGACPVFECGNVVLRTDERDVNYWTSRYWLNGDFRKGDVSLTIENVTLADSGIYCCRIQIPGIMNDEKFNLKLVIKPAKVTPAPTRQRDFTAAFPRMLTTRGHGPAETQTLGSLPDINLTQISTLANELRDSRLANDLRDSGATIR 서열번호 27SEQ ID NO: 27
표 6은 인간 immunoglobulin G4와 인간 TIM-3 extracellular domain 서열을 연결하는 linker 및 인간 TIM-3 extracellular domain 서열, Table 6 shows the linker connecting human immunoglobulin G4 and human TIM-3 extracellular domain sequence and human TIM-3 extracellular domain sequence,
ProteinProtein Amino acid sequenceAmino acid sequence 서열번호sequence number
Human PEDFHuman PEDF MQALVLLLCIGALLGHSSCQNPASPPEEGSPDPDSTGALVEEEDPFFKVPVNKLAAAVSNFGYDLYRVRSSTSPTTNVLLSPLSVATALSALSLGAEQRTESIIHRALYYDLISSPDIHGTYKELLDTVTAPQKNLKSASRIVFEKKLRIKSSFVAPLEKSYGTRPRVLTGNPRLDLQEINNWVQAQMKGKLARSTKEIPDEISILLLGVAHFKGQWVTKFDSRKTSLEDFYLDEERTVRVPMMSDPKAVLRYGLDSDLSCKIAQLPLTGSMSIIFFLPLKVTQNLTLIEESLTSEFIHDIDRELKTVQAVLTVPKLKLSYEGEVTKSLQEMKLQSLFDSPDFSKITGKPIKLTQVEHRAGFEWNEDGAGTTPSPGLQPAHLTFPLDYHLNQPFIFVLRDTDTGALLFIGKILDPRGPMQALVLLLCIGALLGHSSCQNPASPPEEGSPDPDSTGALVEEEDPFFKVPVNKLAAAVSNFGYDLYRVRSSTSPTTNVLLSPLSVATALSALSLGAEQRTESIIHRALYYDLISSPDIHGTYKELLDTVTAPQKNLKSASRIVFEKKLRIKSSFVAPLEKSYGTRPRVLTGNPRLDLQEINNWVQAQMKGKLARSTKEIPDEISILL LGVAHFKGQWVTKFDSRKTSLEDFYLDEERTVRVPMMSDPKAVLRYGLDSDLSCKIAQLPLTGSMSIIFFLPLKVTQNLTLIEESLTSEFIHDIDRELKTVQAVLTVPKLKLSYEGEVTKSLQEMKLQSLFDSPDFSKITGKPIKLTQVEHRAGFEWNEDGAGTTPSPGLQPAHLTFPLDYHLNQPFIFVLRDTDTGALLFIGKILDPRGP 서열번호 28SEQ ID NO: 28
Human IgG4와 Human PEDF 44-mer peptide를 연결하는 linkerLinker connecting human IgG4 and human PEDF 44-mer peptide GGGGSGGGGSGGGGSGGGGS 서열번호 29SEQ ID NO: 29
Human PEDF 44-mer peptide (78-121) 서열Human PEDF 44-mer peptide (78-121) sequence VLLSPLSVATALSALSLGAEQRTESIIHRALYYDLISSPDIHGTVLLSPLSVATALSALSLGAEQRTESIIHRALYYDLISSPDIHGT 서열번호 30SEQ ID NO: 30
표 7은 인간 immunoglobulin G4와 인간 PEDF 44-mer peptide 서열을 연결하는 linker 및 인간 PEDF 44-mer peptide 서열, Table 7 shows the linker connecting human immunoglobulin G4 and human PEDF 44-mer peptide sequence and human PEDF 44-mer peptide sequence,
실시예 2: 항체 발현 및 정제Example 2: Antibody expression and purification
인간 안지오포이에틴-2에 특이적으로 결합하는 제1 항원 결합 부위를 포함하는 이중특이적 항체를 생산하기 위해 ExpiCHO expression system (Thermo Fisher)을 이용하였다. 125 mL flask (Corning)에 ExpiCHO expression 배지 (Thermo Fisher) 30 mL을 이용하여 ExpiCHO를 세포수 6 X 106 cells/mL가 될 때까지 배양하였다. 상기 실시예 1에서 만든 heavy chain, light chain DNA를 1:1.5 비율 (w/w)로 총 25 μg을 OptiPRO SFM (Thermo Fisher) 1 mL을 이용하여 준비하였다. ExpiFectamine CHO reagent (Thermo Fisher) 80 μL을 OptiPRO SFM 920 μL에 넣어 준비하였다. 준비한 DNA와 ExpiFectamine CHO reagent를 섞어준 후, 5분 내에 앞에서 준비한 ExpiCHO 배양액 25 mL에 넣어주어 transfection을 진행하고, 37°C, 8% CO2, 125 rpm 조건에서 배양하였다. Transfection 후 1일차가 되는 날 ExpiFectamine CHO Enhancer (Thermo Fisher) 150 μL와 ExpiCHO Feed (Thermo Fisher) 4 mL을 배양액에 추가로 넣어주고, 32℃, 5% CO2, 125 rpm으로 조건을 바꾸어 배양하였다. Transfection 후 5일차가 되는 날 ExpiCHO Feed 4 mL을 추가로 넣어주었다. Transfection 후 7일차가 되는 날 배양액을 conical tube (SPL)로 옮겨서 3,500 g, 4℃에서 30분 동안 원심분리한 뒤, 상층액을 모아서 0.22 μm CA syringe filter (Sartorius)를 사용하여 filtering 하고, -70℃ deep freezer에 보관하였다.The ExpiCHO expression system (Thermo Fisher) was used to produce a bispecific antibody containing a first antigen binding site that specifically binds to human angiopoietin-2. ExpiCHO was cultured in a 125 mL flask (Corning) using 30 mL of ExpiCHO expression medium (Thermo Fisher) until the cell number reached 6 A total of 25 μg of heavy chain and light chain DNA prepared in Example 1 was prepared at a 1:1.5 ratio (w/w) using 1 mL of OptiPRO SFM (Thermo Fisher). 80 μL of ExpiFectamine CHO reagent (Thermo Fisher) was prepared by adding it to 920 μL of OptiPRO SFM. After mixing the prepared DNA and ExpiFectamine CHO reagent, transfection was performed by adding it to 25 mL of the ExpiCHO culture medium prepared earlier within 5 minutes, and cultured at 37°C, 8% CO 2 , and 125 rpm. On the first day after transfection, 150 μL of ExpiFectamine CHO Enhancer (Thermo Fisher) and 4 mL of ExpiCHO Feed (Thermo Fisher) were added to the culture medium, and the conditions were changed to 32°C, 5% CO 2 , and 125 rpm. On the fifth day after transfection, an additional 4 mL of ExpiCHO Feed was added. On the 7th day after transfection, the culture was transferred to a conical tube (SPL) and centrifuged at 3,500 g for 30 minutes at 4°C. The supernatant was collected and filtered using a 0.22 μm CA syringe filter (Sartorius), and -70 Stored in a deep freezer at ℃.
앞에서 준비한 배양액을 이용하여 정제를 진행하였다. Semi-Prep HPLC system (영인크로매스)에 Hitrap Mabselect Sure (Cytiva) 5mL column을 장착하였다. PBS를 2.5 mL/min 유속으로 5 CV 이상 흘러주어 평형을 잡아주었다. Harvest한 배양액을 0.2 μm filter로 filtration 한 후, 평형을 잡은 column에 2.5 mL/min 유속으로 loading 시켰다. Loading이 완료된 후, 다시 PBS를 UV 값이 평형이 될 때까지 흘러주고, elution buffer (0.1 M glycine, pH 2.7)를 흘러주어 항 Ang2/VEGF 항체를 용출하였다. 용출된 용액은 1 M Tris buffer를 이용하여 pH를 7.0으로 적정하였다.Purification was performed using the culture medium prepared previously. A Hitrap Mabselect Sure (Cytiva) 5mL column was mounted on a Semi-Prep HPLC system (Youngin Chromas). Equilibrium was achieved by flowing over 5 CV of PBS at a flow rate of 2.5 mL/min. The harvested culture was filtered through a 0.2 μm filter and loaded into an equilibrated column at a flow rate of 2.5 mL/min. After loading was completed, PBS was again flowed until the UV value reached equilibrium, and elution buffer (0.1 M glycine, pH 2.7) was flowed to elute the anti-Ang2/VEGF antibody. The pH of the eluted solution was adjusted to 7.0 using 1 M Tris buffer.
정제한 이중특이적 항체의 buffer를 PBS로 바꿔주기 위해 amicon 30 kDa (Millipore)을 이용하였다. Amicon에 PBS 5 mL을 넣어주고, 4℃, 5,000 rpm으로 원심 분리하여 닦아주었다. 원심 분리 후, 용액을 모두 버린 후, 앞에서 용출한 시료 10 mL을 넣어준 후, 4℃, 5,000 rpm으로 원심 분리하여 3 mL 정도로 농축하였다. 농축한 3 mL에 PBS 12 mL을 넣어준 후, 다시 원심 분리하여 3 mL로 농축하였다. 이와 같은 과정을 추가로 4회 반복하여 buffer를 PBS로 교환해 주었다. Buffer 교환이 완료된 시료를 0.22 μm syringe filter (Millipore)를 이용하여 무균 filtration을 수행하여 이중특이적 항체를 준비하였다.Amicon 30 kDa (Millipore) was used to change the buffer of the purified bispecific antibody to PBS. 5 mL of PBS was added to Amicon, centrifuged at 4°C, 5,000 rpm, and cleaned. After centrifugation, all of the solution was discarded, 10 mL of the sample eluted earlier was added, and then centrifuged at 4°C and 5,000 rpm to concentrate to about 3 mL. After adding 12 mL of PBS to the concentrated 3 mL, it was centrifuged again and concentrated to 3 mL. This process was repeated four additional times and the buffer was exchanged for PBS. Bispecific antibodies were prepared by performing sterile filtration on the sample after buffer exchange was completed using a 0.22 μm syringe filter (Millipore).
실시예 3: 항 Ang2/VEGF 항체 정제 시료의 SDS-PAGE 분석Example 3: SDS-PAGE analysis of purified anti-Ang2/VEGF antibody samples
실시예 2에서 정제한 항 Ang2/VEGF 항체의 purity 및 integrity를 확인하기 위해 SDS-PAGE로 분석하였다. 적절한 단백질 농도의 sample 15 μL에 SDS-Sample Buffer 4X reducing (Gendepot) 또는 Native SDS-Sample Buffer 4X non reducing (Gendepot)을 각각 1X가 되도록 넣어주고, reducing sample은 heat block를 사용하여 98℃에서 10분간 열처리하여 sample을 준비하였다. XCell SureLock Mini-Cell (Invitrogen)에 NuPAGE 4~12%, Bis-Tris mini protein gel (Invitrogen)을 설치하고, NuPage 1X MOPS SDS running buffer (Invitrogen)를 넣어 전기영동을 진행할 준비를 하였다. 준비한 gel의 well에 TOPview Prestained Protein Ladder Marker (Enzynomics) 및 상기에서 준비한 sample을 loading 한 후, PowerEase Touch 600W Power Supply (Invitrogen) 장비를 사용하여 200V, 42분간 전기영동 하였다. 전기영동이 끝난 gel을 sun gel staining solution (LPS solution)에 넣어 staining 했다. Staining이 완료된 gel을 증류수에 넣어 destaining 하였다.To confirm the purity and integrity of the anti-Ang2/VEGF antibody purified in Example 2, it was analyzed by SDS-PAGE. To 15 μL of a sample of appropriate protein concentration, add SDS-Sample Buffer 4X reducing (Gendepot) or Native SDS-Sample Buffer 4X non reducing (Gendepot) to 1X each, and heat the reducing sample at 98°C for 10 minutes using a heat block. Samples were prepared by heat treatment. NuPAGE 4-12%, Bis-Tris mini protein gel (Invitrogen) was installed in the XCell SureLock Mini-Cell (Invitrogen), and NuPage 1X MOPS SDS running buffer (Invitrogen) was added to prepare for electrophoresis. TOPview Prestained Protein Ladder Marker (Enzynomics) and the sample prepared above were loaded into the prepared gel well, and then electrophoresed at 200V for 42 minutes using PowerEase Touch 600W Power Supply (Invitrogen) equipment. The gel after electrophoresis was placed in sun gel staining solution (LPS solution) for staining. The gel for which staining was completed was placed in distilled water for destaining.
도 2와 같이 reducing (R), non-reducing (NR) 조건에서 원하는 size의 band를 확인하였으며, impurity도 거의 없는 것을 확인하였다.As shown in Figure 2, a band of the desired size was confirmed under reducing (R) and non-reducing (NR) conditions, and it was confirmed that there was almost no impurity.
실시예 4: 항 Ang2/TGF-β 항체 정제 시료의 SDS-PAGE 및 Western blot 분석Example 4: SDS-PAGE and Western blot analysis of purified anti-Ang2/TGF-β antibody samples
실시예 3와 같은 방법으로 실시예 2에서 정제한 항 Ang2/TGF-β 항체의 purity 및 integrity를 확인하기 위해 SDS-PAGE로 분석하였다. 도 3과 같이 reducing (R), non-reducing (NR) 조건에서 원하는 size의 band를 확인하였으며, impurity도 거의 없는 것을 확인하였다. 항 Ang2/TGF-β 항체의 TGF-β binding region을 확인하기 위해 anti-hTGF-β RII antibody를 이용하여 Western blot 분석을 수행하였다. 정제한 항 Ang2/TGF-β 항체 60 ng, 600 ng을 실시예 3과 같은 방법으로 SDS-PAGE 전기영동을 진행하였다. 전기영동이 끝난 gel을 XCell II Blot Module (Invitrogen)에 nitrocellulose membrane과 같이 장착하고, PowerEase Touch 600W Power Supply (Invitrogen) 장비를 사용하여 30V, 1 시간동안 transfer를 진행하였다. Transfer가 끝난 membrane을 5% (w/v) skim milk가 함유된 0.05% TBST (0.05% (v/v) Tween-20이 포함된 Tris Buffered Saline)에 넣어 1 시간 블로킹하였다. 0.1% TBST로 블로킹이 완료된 membrane을 3번 씻은 뒤, primary antibody buffer (2 μg/mL Goat anti-hTGF-β RII antibody (R&D systems), 5% (w/v) skim milk, 0.05% TBST)에 membrane을 넣어 2 시간 결합시켰다. 0.1% TBST로 primary antibody 결합이 완료된 membrane을 3번 씻은 뒤, secondary antibody buffer (1:5000 Rabbit anti-Goat IgG(H+L)-HRP (Gendepot), 5% (w/v) skim milk, 0.05% TBST)에 membrane을 넣어 1 시간 결합시켰다. Secondary antibody 결합이 완료된 membrane을 0.1% TBST로 3번 씻은 뒤, Clarity Western ECL Substrate peroxide solution (Bio-rad)와 Clarity Western ECL Substrate Luminol/enhancer (Bio-rad)를 1:1로 섞은 용액을 membrane에 뿌려준 후, ImageQuant LAS 500 (Cytiva) 장비로 band를 확인하였다.To confirm the purity and integrity of the anti-Ang2/TGF-β antibody purified in Example 2 in the same manner as in Example 3, it was analyzed by SDS-PAGE. As shown in Figure 3, a band of the desired size was confirmed under reducing (R) and non-reducing (NR) conditions, and it was confirmed that there was almost no impurity. To confirm the TGF-β binding region of the anti-Ang2/TGF-β antibody, Western blot analysis was performed using anti-hTGF-β RII antibody. 60 ng and 600 ng of purified anti-Ang2/TGF-β antibody were subjected to SDS-PAGE electrophoresis in the same manner as in Example 3. After electrophoresis, the gel was mounted on the After the transfer, the membrane was placed in 0.05% TBST (Tris Buffered Saline containing 0.05% (v/v) Tween-20) containing 5% (w/v) skim milk and blocked for 1 hour. After washing the blocked membrane three times with 0.1% TBST, it was added to primary antibody buffer (2 μg/mL Goat anti-hTGF-β RII antibody (R&D systems), 5% (w/v) skim milk, 0.05% TBST). A membrane was added and allowed to bind for 2 hours. After washing the membrane with complete primary antibody binding three times with 0.1% TBST, secondary antibody buffer (1:5000 Rabbit anti-Goat IgG(H+L)-HRP (Gendepot), 5% (w/v) skim milk, 0.05 % TBST) and allowed to bind for 1 hour. After washing the membrane with secondary antibody binding three times with 0.1% TBST, a 1:1 mixture of Clarity Western ECL Substrate peroxide solution (Bio-rad) and Clarity Western ECL Substrate Luminol/enhancer (Bio-rad) was applied to the membrane. After spraying, the band was confirmed using ImageQuant LAS 500 (Cytiva) equipment.
도 4와 같이 reducing (R), non-reducing (NR) 조건에서 anti-TGFβRII 항체를 이용하여 Western blot 분석을 수행한 결과 heavy chain size의 band를 확인함으로써 물질의 integrity를 확인하였다.As shown in Figure 4, Western blot analysis was performed using an anti-TGFβRII antibody under reducing (R) and non-reducing (NR) conditions, and the integrity of the material was confirmed by confirming a band of heavy chain size.
실시예 5: 항 Ang2/VEGF 항체의 binding propertyExample 5: Binding properties of anti-Ang2/VEGF antibody
항 Ang2/VEGF 항체와 인간 Ang2 또는 인간 VEGF 또는 마우스 VEGF 와의 결합능력을 확인하기 위하여 ELISA를 수행하였다. 96-well MaxiSorp™flat-bottom plate에 1 μg/mL의 인간 Ang2 (Sino Biological) 또는 인간 VEGF 165 (Sino Biological) 또는 마우스 VEGF 164 (Sino Biological)를 코팅하였다. 그런 다음, PBST (0.05% (v/v) Tween-20이 포함된 Phosphate Buffer Saline)로 상기 플레이트를 5회 씻은 후, 1% (v/v) skim milk가 함유된 PBST로 상온에서 2시간 동안 블로킹시켰다. 그 후, 0.05% Tween-20이 포함된 PBST로 상기 플레이트를 5회 씻은 후 300 nM 부터 3배 연속 희석한 11 가지 농도의 항 Ang2/VEGF 항체를 넣어 상온에서 2 시간 결합시켰다. PBST를 이용하여 플레이트를 5회 씻은 후 PBST에 1:10,000으로 희석한 Goat anti-Human Kappa/HRP (Novus)를 1시간 동안 결합시킨 후 PBST로 6회 씻어주었다. 마지막으로 상기 플레이트에 TMB 기질 (Kementec) 100 μL를 첨가하여 10분간 발색반응을 유도시킨 후, Stop 용액 (2 M H2SO4) 50 μL를 첨가하여 반응을 중지시키고, OD450 값을 plate reader (BioTek)로 측정하였다. 도 5 내지 8과 같이 본 발명에서 개발한 항 Ang2/VEGF 항체(2C8-D2 및1D3-D2)가 Ang2 및 VEGF에 결합하는 것을 확인하였다.ELISA was performed to confirm the binding ability of the anti-Ang2/VEGF antibody with human Ang2, human VEGF, or mouse VEGF. A 96-well MaxiSorp™ flat-bottom plate was coated with 1 μg/mL of human Ang2 (Sino Biological), human VEGF 165 (Sino Biological), or mouse VEGF 164 (Sino Biological). Then, the plate was washed five times with PBST (Phosphate Buffer Saline containing 0.05% (v/v) Tween-20) and then washed with PBST containing 1% (v/v) skim milk at room temperature for 2 hours. Blocked. Afterwards, the plate was washed five times with PBST containing 0.05% Tween-20, and 11 different concentrations of anti-Ang2/VEGF antibodies serially diluted 3 times from 300 nM were added and allowed to bind at room temperature for 2 hours. After washing the plate 5 times with PBST, Goat anti-Human Kappa/HRP (Novus) diluted 1:10,000 in PBST was added for 1 hour, and then washed 6 times with PBST. Finally, 100 μL of TMB substrate (Kementec) was added to the plate to induce a color reaction for 10 minutes, then 50 μL of Stop solution (2 MH 2 SO 4 ) was added to stop the reaction, and the OD450 value was measured using a plate reader (BioTek). ) was measured. As shown in Figures 5 to 8, it was confirmed that the anti-Ang2/VEGF antibodies (2C8-D2 and 1D3-D2) developed in the present invention bind to Ang2 and VEGF.
항 Ang2/VEGF 항체의 Ang2, VEGF 동시 결합능력을 확인하기 위하여 ELISA를 수행하였다. 96-well MaxiSorp™flat-bottom plate에 1 μg/mL의 인간 VEGF 165 (Sino Biological)를 코팅하였다. 코팅이 완료된 plate를 PBST로 5회 씻은 후, 1% (v/v) skim milk가 함유된 PBST로 상온에서 2시간 동안 블로킹시켰다. 인간 Ang2 (Sino Biological) 60 nM에 항 Ang2/VEGF 항체를 300 nM 부터 3배 연속 희석한 11 가지 농도로 미리 섞어 준 후, 상온에서 30분 정치하였다. 블로킹이 완료된 plate를 PBST로 5회 씻은 후, 미리 준비한 시료를 넣고, 상온에서 2 시간 결합시켰다. PBST를 이용하여 플레이트를 5회 씻은 후 PBST에 1:20,000으로 희석한 Mouse anti-His tag/HRP (SouthernBiotech)를 1시간 동안 결합시킨 후 PBST로 6회 씻어주었다. 마지막으로 상기 플레이트에 TMB 기질 (Kementec) 100 μL를 첨가하여 10분간 발색반응을 유도시킨 후, Stop 용액 (2 M H2SO4) 100 μL를 첨가하여 반응을 중지시키고, OD450 값을 plate reader (BioTek)로 측정하였다.ELISA was performed to confirm the ability of the anti-Ang2/VEGF antibody to simultaneously bind Ang2 and VEGF. 1 μg/mL human VEGF 165 (Sino Biological) was coated on a 96-well MaxiSorp™ flat-bottom plate. The coated plate was washed five times with PBST and then blocked with PBST containing 1% (v/v) skim milk at room temperature for 2 hours. 60 nM of human Ang2 (Sino Biological) was mixed in advance with 11 different concentrations of anti-Ang2/VEGF antibody, serially diluted 3 times from 300 nM, and left at room temperature for 30 minutes. After washing the plate on which blocking was completed 5 times with PBST, previously prepared samples were added and allowed to bind at room temperature for 2 hours. After washing the plate 5 times with PBST, Mouse anti-His tag/HRP (SouthernBiotech) diluted 1:20,000 in PBST was bound to the plate for 1 hour, and then washed 6 times with PBST. Finally, 100 μL of TMB substrate (Kementec) was added to the plate to induce a color reaction for 10 minutes, then 100 μL of Stop solution (2 MH 2 SO 4 ) was added to stop the reaction, and the OD450 value was measured using a plate reader (BioTek). ) was measured.
도 9와 같이 본 발명에서 개발한 항 Ang2/VEGF 항체는 Ang2와 VEGF에 동시에 결합할 수 있다는 것을 확인하였다.As shown in Figure 9, it was confirmed that the anti-Ang2/VEGF antibody developed in the present invention can bind to Ang2 and VEGF at the same time.
실시예 6: 항 Ang2/TGF-β 항체의 binding propertyExample 6: Binding properties of anti-Ang2/TGF-β antibody
항 Ang2/TGF-β 항체의 Ang2 또는 TGF-β와의 결합능력을 확인하기 위하여 BLI (Bio-layer interferometry) 분석을 수행하였다. Octet Amine Reactive Second-Generation (AR2G) Biosensor (Sartorius)를 AR2G kit에 포함된 1X kinetics buffer에 넣어 10분간 hydration 시켰다. Hydration이 완료된 biosensor를 1X kinetics buffer가 들어있는 0.5 mL LightSafe micro centrifuge tube (Sigma)에 넣어 60 초간 Initial baseline을 측정하였다. 증류수 225 μL와 400 mM EDC, 200 mM S-NHS를 각각 12.5 μL씩 넣은 0.5 mL LightSafe micro centrifuge tube에 biosensor를 넣어 300 초간 activation시켰다. 20 μg/mL TGF-β (Acrobiosystems)가 포함된 10 mM acetate buffer (pH 5.0)를 drop holder에 4 μL loading 한 후, activation이 완료된 biosensor를 넣어 300 초간 immobilization을 하였다. 1 M ethanolamine (pH 8.5) 250 μL가 들어있는 0.5ml LightSafe micro centrifuge tube에 immobilization이 완료된 biosensor를 넣고 300 초간 quenching 하였다. Quenching이 완료된 biosensor를 다시 1X kinetics buffer에 넣어 90 초간 baseline을 측정한 후, 0 - 500 nM 항 Ang2/TGF-β 항체가 포함된 1X kinetics buffer 4 μL를 loading 한 drop holder에 biosensor를 넣어 300 초간 association을 진행하였다. Association 종료 후, biosensor를 1X kinetics buffer에 넣어 600 초간 dissociation을 측정하였다. 분석을 완료한 후, global fitting으로 ka, kd, KD를 계산하였다.Bio-layer interferometry (BLI) analysis was performed to confirm the binding ability of the anti-Ang2/TGF-β antibody to Ang2 or TGF-β. Octet Amine Reactive Second-Generation (AR2G) Biosensor (Sartorius) was placed in 1X kinetics buffer included in the AR2G kit and hydrated for 10 minutes. After completing hydration, the biosensor was placed in a 0.5 mL LightSafe micro centrifuge tube (Sigma) containing 1X kinetics buffer, and the initial baseline was measured for 60 seconds. The biosensor was placed in a 0.5 mL LightSafe micro centrifuge tube containing 225 μL of distilled water and 12.5 μL each of 400 mM EDC and 200 mM S-NHS and activated for 300 seconds. After loading 4 μL of 10 mM acetate buffer (pH 5.0) containing 20 μg/mL TGF-β (Acrobiosystems) into the drop holder, the activated biosensor was added and immobilized for 300 seconds. The immobilized biosensor was placed in a 0.5ml LightSafe micro centrifuge tube containing 250 μL of 1 M ethanolamine (pH 8.5) and quenched for 300 seconds. After quenching was completed, the biosensor was placed back in 1 proceeded. After the association was completed, the biosensor was placed in 1X kinetics buffer and dissociation was measured for 600 seconds. After completing the analysis, ka, kd, and KD were calculated by global fitting.
ks [M-1s-1]ks [M-1s-1] kd [s-1]kd [s-1] KD [nM]K.D. [nM]
hAng2 RBDhAng2 RBD 5.31 x 104 5.31 x 10 4 6.64 x 10-4 6.64 x 10 -4 12.512.5
hTGF-β1hTGF-β1 4.82 x 104 4.82 x 10 4 9.65 x 10-4 9.65 x 10 -4 20.020.0
표 8과 같이 본 발명에서 개발한 항 Ang2/TGF-β 항체는 Ang2와 TGF-β에 결합할 수 있다는 것을 확인하였다.As shown in Table 8, it was confirmed that the anti-Ang2/TGF-β antibody developed in the present invention can bind to Ang2 and TGF-β.
실시예 7: 항 Ang2/VEGF 항체 또는 항 Ang2/TGF-β항체에 의한 Tie2 signaling의 활성화 유도 확인을 위한 Western blot 분석Example 7: Western blot analysis to confirm activation of Tie2 signaling by anti-Ang2/VEGF antibody or anti-Ang2/TGF-β antibody
Ang2는 혈관내피세포에 발현된 Tie2 수용체와 결합하여 약한 작용제 또는 길항제로서 작용한다. 본 발명에서 개발한 항 Ang2/VEGF 항체 또는 항 Ang2/TGF-β 항체는 Ang2에 결합하여 Ang2-Tie2와 복합체를 형성함으로써 Tie2 수용체를 활성화시켜 하부 신호전달을 촉진시키는 작용을 할 수 있다. 항 Ang2/VEGF 항체 또는 항 Ang2/TGF-β 항체의 Tie2 하부 신호전달에 미치는 영향을 분석하기 위하여 HUVEC에서 ERK와 AKT 인산화 실험을 수행하였다. Tie2 하부 신호전달 활성화 정도를 비교하기 위하여, Ang2 (Sino Biological)만 처리한 그룹에 대하여도 동일한 시험을 수행하였다. Ang2 binds to the Tie2 receptor expressed on vascular endothelial cells and acts as a weak agonist or antagonist. The anti-Ang2/VEGF antibody or anti-Ang2/TGF-β antibody developed in the present invention can bind to Ang2 and form a complex with Ang2-Tie2, thereby activating the Tie2 receptor and promoting downstream signaling. To analyze the effect of anti-Ang2/VEGF antibody or anti-Ang2/TGF-β antibody on Tie2 downstream signaling, ERK and AKT phosphorylation experiments were performed in HUVEC. In order to compare the degree of activation of Tie2 downstream signaling, the same test was performed on the group treated only with Ang2 (Sino Biological).
구체적으로, HUVEC (Lonza) 세포 (2 X 105개)를 EGM-2 (Lonza) 배지를 이용하여 37℃에서 배양 후, 80~90% confluency를 보이면, 0.5% FBS Basal media(Lonza)로 바꾸어 16시간 내지 24시간 동안 37℃에서 배양하였다. 항 Ang2/VEGF 항체 또는 항 Ang2/TGF-β 항체 60 nM을 Ang2 단백질 (Sino Biological) 40 nM과 혼합하여 30 분간 둔 뒤, 상기 배양된 세포에 처리하고 10 분간 더 배양하였다. 비교를 위하여, 항체 없이 Ang2 40 nM만 처리한 그룹도 준비하였다. PBS를 이용하여 상기 세포를 세척한 뒤, lysis buffer (Ripa buffer (바이오세상), 0.15M Sodium chloride, 1% Triton X-100, 1% Sodium deoxycholate, 0.1% SDS, 50 mM Tris-HCl, pH 7.5, and 2 mM EDTA)를 처리한 후, 13,000 rpm으로 15분간 원심 분리하여 상층액을 회수하여 세포용해물을 얻었다.Specifically, HUVEC (Lonza) cells ( 2 Cultured at 37°C for 16 to 24 hours. 60 nM of anti-Ang2/VEGF antibody or anti-Ang2/TGF-β antibody was mixed with 40 nM of Ang2 protein (Sino Biological) and left for 30 minutes. Then, the cultured cells were treated and incubated for another 10 minutes. For comparison, a group treated with only 40 nM Ang2 without antibody was also prepared. After washing the cells using PBS, lysis buffer (Ripa buffer (Biosesang), 0.15M Sodium chloride, 1% Triton , and 2 mM EDTA), centrifuged at 13,000 rpm for 15 minutes, and the supernatant was recovered to obtain a cell lysate.
세포용해물 20-30 μg에 reducing agent가 혼합된 sample buffer (GenDEPOT)를 넣고 95℃에서 5분간 끓인 뒤, 10% Tris-Glycine gel 또는 4-12% Bis-Tris gel (Invitrogen)에서 전기영동하고, NC 또는 PVDF 멤브레인 (Millipore)에 이동시켰다. Tie2 수용체의 downstream signaling에 관여하는 Akt, ERK 42/44의 인산화 여부를 확인하기 위하여, 위의 blot을 5%(v/v) skim milk (서울우유)가 혼합된 PBST로 1 시간 동안 blocking 한 뒤, 항 인산화 Akt 항체 (Cell signaling), 항 AKT 항체 (Cell signaling), 항 인산화 ERK 42/44 항체 (Cell signaling), 항 ERK 42/44 항체(Santa cruz)를 처리하였다.Add sample buffer (GenDEPOT) mixed with reducing agent to 20-30 μg of cell lysate, boil at 95°C for 5 minutes, and then electrophoresed on 10% Tris-Glycine gel or 4-12% Bis-Tris gel (Invitrogen). , transferred onto NC or PVDF membranes (Millipore). To check the phosphorylation of Akt and ERK 42/44, which are involved in downstream signaling of the Tie2 receptor, the above blot was blocked for 1 hour with PBST mixed with 5% (v/v) skim milk (Seoul Milk). , anti-phosphorylated Akt antibody (Cell signaling), anti-AKT antibody (Cell signaling), anti-phosphorylated ERK 42/44 antibody (Cell signaling), and anti-ERK 42/44 antibody (Santa Cruz).
도 10에서 보여지는 바와 같이, 항 Ang2/VEGF 항체 및 항 Ang2/TGF-β 항체는 음성대조군 대비 AKT 및 ERK 활성화 시그널을 보여주었다.As shown in Figure 10, anti-Ang2/VEGF antibody and anti-Ang2/TGF-β antibody showed AKT and ERK activation signals compared to the negative control group.
실시예 8: 항 Ang2/VEGF 항체와 VEGFR2의 경쟁적 VEGF 결합Example 8: Competitive VEGF binding of anti-Ang2/VEGF antibody to VEGFR2
VEGF는 혈관 형성을 촉진시켜주는 대표적인 pro-angiogenic cytokine으로, VEGF는 VEGFR2에 결합하여 하위 signal을 활성화시켜 줌으로써 angiogenesis를 조절하는 것으로 알려져 있다. 본 발명에서 고안한 항 Ang2/VEGF 항체는 VEGFR2와 경쟁적으로 VEGF에 결합하여 VEGF가 VEGFR2 하위 signal을 활성화시켜 주는 것을 저해할 수 있다. 항 Ang2/VEGF 항체와 VEGFR2의 경쟁적 VEGF 결합을 확인하기위해 ELISA 분석을 수행하였다.VEGF is a representative pro-angiogenic cytokine that promotes blood vessel formation. VEGF is known to regulate angiogenesis by binding to VEGFR2 and activating downstream signals. The anti-Ang2/VEGF antibody designed in the present invention can bind to VEGF competitively with VEGFR2 and inhibit VEGF from activating the VEGFR2 downstream signal. ELISA analysis was performed to confirm competitive VEGF binding of anti-Ang2/VEGF antibody and VEGFR2.
Greiner Bio-One 96-well Half Area ELISA Microplates에 2 μg/mL의 인간 VEGFR2 ECD (Sino Biological)를 코팅하였다. PBST (0.05% (v/v) Tween-20이 포함된 Phosphate Buffer Saline)로 상기 plate를 5회 씻은 후, 1% (w/v) skim milk가 함유된 PBST로 상온에서 2시간 동안 블로킹시켰다. PBST로 상기 plate를 5회 씻은 후, 1,500 nM 부터 3배 연속 희석한 11 가지 농도의 항 Ang2/VEGF 항체와 AVI tag이 있는 인간 VEGF (Sino Biological) 150 μg/mL을 상온에서 10분 pre-incubation시킨 sample을 상기에서 준비한 plate에 넣고, 상온에서 2 시간 결합시켰다. PBST로 plate를 5회 씻은 후, PBST에 1:15,000으로 희석한 Anti-Streptavidin-HRP (Abcam)를 plate에 넣어 1시간 동안 결합시키고, 다시 PBST로 6회 씻어주었다. 상기 plate에 TMB 기질 (Kementec) 50 μL를 첨가하여 10분간 발색반응을 유도시킨 후, Stop 용액 (2 M H2SO4) 25 μL를 첨가하여 반응을 중지시키고, OD450 값을 plate reader (BioTek)로 측정하였다. 도 11과 같이 항 Ang2/VEGF 항체의 농도를 높여줄수록 VEGFR2에 결합하는 VEGF의 양이 감소하는 것을 확인함으로써 항 Ang2/VEGF 항체는 VEGFR2와 경쟁적으로 VEGF에 결합하는 것을 확인하였다.Greiner Bio-One 96-well Half Area ELISA Microplates were coated with 2 μg/mL human VEGFR2 ECD (Sino Biological). The plate was washed five times with PBST (Phosphate Buffer Saline containing 0.05% (v/v) Tween-20) and then blocked with PBST containing 1% (w/v) skim milk at room temperature for 2 hours. After washing the plate 5 times with PBST, pre-incubate 150 μg/mL of human VEGF (Sino Biological) with AVI tag and 11 different concentrations of anti-Ang2/VEGF antibody serially diluted 3 times from 1,500 nM for 10 minutes at room temperature. The samples were placed in the plate prepared above and combined for 2 hours at room temperature. After washing the plate 5 times with PBST, Anti-Streptavidin-HRP (Abcam) diluted 1:15,000 in PBST was added to the plate, allowed to bind for 1 hour, and washed again 6 times with PBST. 50 μL of TMB substrate (Kementec) was added to the plate to induce a color reaction for 10 minutes, then 25 μL of Stop solution (2 MH 2 SO 4 ) was added to stop the reaction, and the OD450 value was recorded using a plate reader (BioTek). Measured. As shown in Figure 11, it was confirmed that the amount of VEGF bound to VEGFR2 decreased as the concentration of the anti-Ang2/VEGF antibody increased, thereby confirming that the anti-Ang2/VEGF antibody binds to VEGF competitively with VEGFR2.
실시예 9: 항 Ang2/VEGF 항체에 의한 HUVEC proliferation 저해Example 9: Inhibition of HUVEC proliferation by anti-Ang2/VEGF antibody
VEGF는 혈관 형성을 촉진시켜주는 대표적인 pro-angiogenic cytokine으로, VEGF는 HUVEC의 proliferation을 촉진시켜주는 것으로 알려져 있다. 항 Ang2/VEGF 항체는 배양액 내 VEGF와 결합하여 VEGF가 HUVEC의 VEGFR2에 결합하는 것을 저해함으로써 HUVEC proliferation을 저해할 수 있다. 이를 확인하기 위하여 HUVEC에 VEGF 또는 VEGF와 항 Ang2/VEGF 항체를 넣어주어 HUVEC의 proliferation을 확인하는 실험을 수행하였다.VEGF is a representative pro-angiogenic cytokine that promotes blood vessel formation, and VEGF is known to promote HUVEC proliferation. Anti-Ang2/VEGF antibodies can inhibit HUVEC proliferation by binding to VEGF in the culture medium and inhibiting VEGF from binding to VEGFR2 of HUVEC. To confirm this, an experiment was performed to check the proliferation of HUVEC by adding VEGF or VEGF and anti-Ang2/VEGF antibody to HUVEC.
구체적으로 0.5% FBS가 포함되어 있는 EBM-2 media (Lonza)를 이용하여 HUVEC (Lonza)을 0.1% gelatin이 coating된 96 well cell culture plate (SPL)에 50,000 - 0 cells/well이 되도록 넣어준 후, incubator (37℃, 5% CO2)에서 16시간 배양하였다. 0.5% FBS가 포함되어 있는 EBM-2 media에 VEGF 30 ng/mL, VEGF 30 ng/mL + 항 Ang2/VEGF 항체 2.5 nM이 되도록 세포에 처리하기 30분 전에 준비하였다. 16시간 배양이 끝난 plate의 상층액을 제거한 후, 미리 준비한 시료를 각 well에 100 μL씩 넣어준 후, incubator (37℃, 5% CO2)에서 72시간 배양하였다. 72시간 후, plate의 각 well에 MTS (Promega)를 20 μL씩 넣어주고, 다시 incubator에서 4시간 동안 반응시켰다. 반응 종료 후, plate reader (BioTek)를 이용하여 490 nm에서 OD를 측정하였다. 도 12의 결과와 같이 VEGF를 넣어주었을 때, 같은 세포수에서 OD 값이 VEGF를 넣지 않았을 때 보다 높은 것을 확인함으로써 VEGF에 의한 HUVEC proliferation을 확인할 수 있었다. VEGF와 항 Ang2/VEGF 항체를 함께 넣어준 경우, VEGF만 넣어줬을 때보다 같은 세포수에서 OD 값이 낮은 것을 확인함으로써 항 Ang2/VEGF 항체가 배지 내 VEGF와 결합하여 VEGF에 의한 HUVEC proliferation을 저해할 수 있다는 것을 확인하였다. Specifically, using EBM-2 media (Lonza) containing 0.5% FBS, HUVEC (Lonza) were placed in a 96 well cell culture plate (SPL) coated with 0.1% gelatin at 50,000 - 0 cells/well. , cultured in an incubator (37°C, 5% CO 2 ) for 16 hours. EBM-2 media containing 0.5% FBS was prepared 30 minutes before treating the cells with VEGF 30 ng/mL, VEGF 30 ng/mL + anti-Ang2/VEGF antibody 2.5 nM. After removing the supernatant of the plate after 16 hours of incubation, 100 μL of the previously prepared sample was added to each well and cultured in an incubator (37°C, 5% CO 2 ) for 72 hours. After 72 hours, 20 μL of MTS (Promega) was added to each well of the plate and reacted again in the incubator for 4 hours. After completion of the reaction, OD was measured at 490 nm using a plate reader (BioTek). As shown in the results in Figure 12, when VEGF was added, the OD value was higher at the same cell number than when VEGF was not added, confirming HUVEC proliferation caused by VEGF. When VEGF and anti-Ang2/VEGF antibody were added together, the OD value was lower for the same number of cells than when VEGF alone was added, suggesting that anti-Ang2/VEGF antibody binds to VEGF in the medium and inhibits HUVEC proliferation caused by VEGF. It was confirmed that it was possible.
실시예 10: Mouse Laser-induced CNV model에서 항 Ang2/VEGF 항체의 신생혈관 형성 억제 및 시력 개선 효과Example 10: Inhibition of neovascularization and improvement of visual acuity of anti-Ang2/VEGF antibody in Mouse Laser-induced CNV model
누수가 심한 비정상적인 신생혈관 생성에 의해 유발되는 습성 황반변성 (wet AMD) model에서 항 Ang2/VEGF 항체의 therapeutic potential을 확인하기 위한 연구를 수행하였다. 습성 황반변성 model로 많이 사용하는 mouse Laser-induced CNV (choroidal neovascularization) model을 이용하여 항 Ang2/VEGF 항체의 choroidal neovascularization 억제 및 망막기능 회복 효능을 positive control 약물인 aflibercept과 비교 평가하였다.A study was conducted to confirm the therapeutic potential of anti-Ang2/VEGF antibodies in a wet AMD model caused by abnormal neovascularization with severe leakage. Using the mouse Laser-induced CNV (choroidal neovascularization) model, which is widely used as a wet macular degeneration model, the efficacy of anti-Ang2/VEGF antibody in suppressing choroidal neovascularization and restoring retinal function was compared with aflibercept, a positive control drug.
C57BL/6 마우스에 레이저 CNV를 유도한 후, CNV 대조군 및 양성대조군인 IgG와 aflibercept를 각각 5 μg/μL/eye (약 35 μM)와 20 μg/μL/eye (약 177 μM) 단회 IVT (intravitreal injection) 투여하였다. 시험군인 항 Ang2/VEGF 항체를 5.8 μg/μL/eye (약 35 μM)의 용량으로 단회 IVT 투여하였다. 망막영상평가는 CNV 유도 후 11일째에 FFA (fundus fluorescein angiography) 및 OCT (optical coherence tomography)를 실시하여, CNV 병변의 크기와 leakage 정도, 그리고 병변의 부피를 각각 측정하였다. 망막 전위도 평가는 CNV 유도 후 12일째에 scotopic ERG (electroretinogram)에서 mono-flash (0.9 log cds/m2) 자극에 대한 망막 전위도의 변화를 B-wave amplitude로 평가하였다.After inducing laser CNV in C57BL/6 mice, a single IVT (intravitreal) injection of CNV control group and positive control group, IgG and aflibercept, was administered at 5 μg/μL/eye (about 35 μM) and 20 μg/μL/eye (about 177 μM), respectively. injection) was administered. The test group, anti-Ang2/VEGF antibody, was administered as a single IVT at a dose of 5.8 μg/μL/eye (about 35 μM). For retinal imaging evaluation, fundus fluorescein angiography (FFA) and optical coherence tomography (OCT) were performed 11 days after CNV induction to measure the size and degree of leakage of the CNV lesion, and the lesion volume, respectively. Electroretinogram evaluation was performed on the scotopic ERG (electroretinogram) 12 days after CNV induction. Electroretinogram changes in response to mono-flash (0.9 log cds/m2) stimulation were evaluated using B-wave amplitude.
항 Ang2/VEGF 항체를 5.8 μg/μL/eye의 용량으로 CNV 유도 직후에 단 회 IVT 투여하는 경우, CNV 병변의 크기 및 leakage를 감소시키고 (도 13, 도 14), scotopic ERG 상에서 B-wave amplitude를 유의하게 개선시키는 것 (도 15)을 확인하였다.When anti-Ang2/VEGF antibody is administered as a single IVT immediately after CNV induction at a dose of 5.8 μg/μL/eye, the size and leakage of CNV lesions are reduced (Figures 13 and 14), and the B-wave amplitude on scotopic ERG is reduced. It was confirmed that the was significantly improved (FIG. 15).
참고예 1: 항 Ang2 항체의 제조 및 스크리닝Reference Example 1: Preparation and screening of anti-Ang2 antibody
항원으로서 인간 Ang2에 특이적으로 결합하는 항체를 앱클론(대한민국)사에 주문 의뢰하였으며, 본 발명의 항체 제조는 Kohler and Milstein, Eur. J. Immunol. 6, 511 (1976)을 참고하였다. 요약하면, 마우스에 인간 Ang2을 항원으로 adjuvant(Sigma)와 혼합하여 2회 주입한 후 항체 생성여부를 ELISA법으로 확인하였다. 2회 면역 후 항체의 역가 (1:5000)가 증가하여 면역된 생쥐에서 비장을 떼어내어 B림프구를 분리한 다음 sp2/0 세포와 융합시켰다. 융합된 세포를 hypoxantin, aminopterine, thymidine이 첨가되어 있는 배지 (HAT mediaum)에서 배양하여 B림프구와 sp2/0세포가 융합된 세포 (hybridoma)를 선택적으로 선별하였다. 얻어진 hybridoma 세포를 단계희석법 (serial dilution method)를 이용하여 양성세포와 음성세포를 분리하는 과정(cloning)을 반복하여 항원에 반응하는 항체를 생산하는 단일클론세포를 제조하였다. 상기 얻어진 항체생산 하이브리도마 세포를 10% (v/v) FBS가 포함된 DMEM (Dulbeco's Modified Eagle's Mediaum)에서 37℃, 5% CO2 조건에서 배양한 후 원심분리를 통해 항체를 생산하는 세포를 제고하고 항체들이 분비된 배양액을 분리하여 친화성 컬럼(Protein A/G agarose column, Protein A/G (GenDEPOT))을 이용하여 항체를 순수 정제하였다.An antibody that specifically binds to human Ang2 as an antigen was ordered from AbClone (Korea), and the antibody of the present invention was manufactured by Kohler and Milstein, Eur. J Immunol. 6, 511 (1976). In summary, human Ang2 mixed with an adjuvant (Sigma) as an antigen was injected twice into mice, and antibody production was confirmed by ELISA. After two immunizations, the antibody titer (1:5000) increased, so the spleens were removed from the immunized mice, and B lymphocytes were isolated and fused with sp2/0 cells. The fused cells were cultured in a medium (HAT mediaum) supplemented with hypoxantin, aminopterine, and thymidine, and cells (hybridoma) in which B lymphocytes and sp2/0 cells were fused were selectively selected. The obtained hybridoma cells were separated into positive and negative cells using a serial dilution method, and the cloning process was repeated to prepare monoclonal cells that produce antibodies that react to the antigen. The obtained antibody-producing hybridoma cells were cultured in DMEM (Dulbeco's Modified Eagle's Mediaum) containing 10% (v/v) FBS at 37°C and 5% CO 2 conditions, and then centrifuged to obtain antibody-producing cells. The culture medium in which the antibodies were secreted was separated, and the antibodies were purified using an affinity column (Protein A/G agarose column, Protein A/G (GenDEPOT)).

Claims (8)

  1. 인간 안지오포이에틴(ANG)-2에 특이적으로 결합하는 제1 항원 결합 부위 및 A first antigen binding site that specifically binds to human angiopoietin (ANG)-2, and
    인간 혈관내피 성장인자(VEGF), 형질전환 성장인자(TGF)-베타, T세포 면역글로불린 및 뮤신 도메인 함유 단백질(TIM)-3 리간드, 및 색소 상피 유래인자 수용체(pigment epithelium-derived factor receptor, PEDF-R)로 구성된 군으로부터 선택된 하나의 항원에 특이적으로 결합하는 제2 항원 결합 부위를 포함하는 이중특이적 항체로서, Human vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-beta, T-cell immunoglobulin and mucin domain-containing protein (TIM)-3 ligand, and pigment epithelium-derived factor receptor (PEDF). -R), a bispecific antibody comprising a second antigen binding site that specifically binds to one antigen selected from the group consisting of,
    i) 상기 ANG-2에 특이적으로 결합하는 상기 제1 항원 결합 부위가 서열번호 1 또는 7 또는 13의 CDRH1 영역, 서열번호 2 또는 8 또는 14의 CDRH2 영역 및 서열번호 3 또는 9 또는 15의 CDRH3 영역을 중쇄 가변 도메인 내에 포함하고, 서열번호 4 또는 10 또는 16의 CDRL1 영역, 서열번호 5 또는 11 또는 17의 CDRL2 영역 및 서열번호 6 또는 12 또는 18의 CDRL3 영역을 경쇄 가변 도메인 내에 포함하고;i) The first antigen binding site that specifically binds to the ANG-2 is the CDRH1 region of SEQ ID NO: 1 or 7 or 13, the CDRH2 region of SEQ ID NO: 2 or 8 or 14, and the CDRH3 of SEQ ID NO: 3 or 9 or 15 comprising a region within a heavy chain variable domain, a CDRL1 region of SEQ ID NO: 4 or 10 or 16, a CDRL2 region of SEQ ID NO: 5 or 11 or 17 and a CDRL3 region of SEQ ID NO: 6 or 12 or 18 within a light chain variable domain;
    ii) 상기 VEGF에 특이적으로 결합하는 상기 제2 항원 결합 부위는 VEGFR1 D2 도메인을 포함하고, ii) the second antigen binding site that specifically binds to VEGF comprises a VEGFR1 D2 domain,
    상기 TGF-베타에 특이적으로 결합하는 상기 제2 항원 결합 부위는 TGF-베타 RII ECD을 포함하고,The second antigen binding site that specifically binds to TGF-beta includes TGF-beta RII ECD,
    상기 TIM-3의 리간드에 특이적으로 결합하는 상기 제2 항원 결합 부위는 TIM-3 ECD를 포함하고,The second antigen binding site that specifically binds to the ligand of TIM-3 includes the TIM-3 ECD,
    상기 PEDF-R에 특이적으로 결합하는 상기 제2 항원 결합 부위는 44-머(mer)의 PEDF를 포함하며,The second antigen binding site that specifically binds to the PEDF-R includes 44-mer PEDF,
    iii) 상기 항체는 Tie2 활성화를 유도하는, 이중특이적 항체.iii) the antibody is a bispecific antibody that induces Tie2 activation.
  2. 제1항에 있어서, 상기 VEGFR1 D2도메인은 서열번호 21의 VEGFR1 D2 도메인,The method of claim 1, wherein the VEGFR1 D2 domain is the VEGFR1 D2 domain of SEQ ID NO: 21,
    상기 TGF-베타 RII ECD는 서열번호 24의 TGF-베타 RII ECD,The TGF-beta RII ECD is the TGF-beta RII ECD of SEQ ID NO: 24,
    상기 TIM-3 ECD는 서열번호 27의 TIM-3 ECD,The TIM-3 ECD is TIM-3 ECD of SEQ ID NO: 27,
    상기 44-머(mer)의 PEDF는 서열번호 30의 아미노산 서열을 포함하는 것을 특징으로 하는 이중특이적 항체.The 44-mer PEDF is a bispecific antibody characterized in that it contains the amino acid sequence of SEQ ID NO: 30.
  3. 제1항 또는 제2항에 있어서, 상기 VEGFR1 D2 도메인, 상기 TGF-베타 RII ECD, 상기 TIM-3 ECD, 및 상기 44-머(mer)의 PEDF는 면역글로불린과 링커로 연결된 것을 특징으로 하는 이중특이적 항체.The method of claim 1 or 2, wherein the VEGFR1 D2 domain, the TGF-beta RII ECD, the TIM-3 ECD, and the 44-mer PEDF are linked to an immunoglobulin by a linker. Specific antibodies.
  4. 제3항에 있어서, 상기 링커는 각각 서열번호 20, 서열번호 23, 서열번호 26 또는 서열번호 29의 링커인 것을 특징으로 하는 이중특이적 항체.The bispecific antibody according to claim 3, wherein the linker is each of SEQ ID NO: 20, SEQ ID NO: 23, SEQ ID NO: 26, or SEQ ID NO: 29.
  5. 제 1 항 내지 제 4 항 중 어느 한 항에 따른 항체를 포함하는 암 치료를 위한 약학적 조성물.A pharmaceutical composition for the treatment of cancer comprising the antibody according to any one of claims 1 to 4.
  6. 제 1 항 내지 제 4 항 중 어느 한 항에 따른 항체를 포함하는 혈관 질환 치료를 위한 약학적 조성물.A pharmaceutical composition for the treatment of vascular diseases comprising the antibody according to any one of claims 1 to 4.
  7. 제 1 항 내지 제 4 항 중 어느 한 항에 따른 이중특이성 항체를 인코딩하는 핵산.A nucleic acid encoding a bispecific antibody according to any one of claims 1 to 4.
  8. 제 1 항 내지 제 4 항 중 어느 한 항에 따른 이중특이성 항체의 제조 방법으로서, 원핵 또는 진핵 숙주 세포에 서 상기 이중특이성 항체를 인코딩하는 핵산을 발현하고, 세포 또는 세포 배양 상청액으로부터 이중특이성 항체를 회수하는 것을 특징으로 하는, 이중특이성 항체의 제조 방법.A method for producing a bispecific antibody according to any one of claims 1 to 4, comprising expressing a nucleic acid encoding said bispecific antibody in a prokaryotic or eukaryotic host cell and producing the bispecific antibody from the cell or cell culture supernatant. A method for producing a bispecific antibody, characterized in that recovery.
PCT/KR2023/016063 2022-10-20 2023-10-17 Bispecific antibody including first antigen-binding site that specifically binds to human angiopoietin-2, and use thereof WO2024085606A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20220135696 2022-10-20
KR10-2022-0135696 2022-10-20
KR10-2023-0136843 2023-10-13
KR1020230136843A KR20240056422A (en) 2022-10-20 2023-10-13 Bispecific antibody comprising a first antigen binding site that specifically binds to human angiopoietin-2 and Use of the same

Publications (1)

Publication Number Publication Date
WO2024085606A1 true WO2024085606A1 (en) 2024-04-25

Family

ID=90738160

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/016063 WO2024085606A1 (en) 2022-10-20 2023-10-17 Bispecific antibody including first antigen-binding site that specifically binds to human angiopoietin-2, and use thereof

Country Status (1)

Country Link
WO (1) WO2024085606A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150079112A1 (en) * 2013-09-17 2015-03-19 Samsung Electronics Co., Ltd. Use of an anti-ang2 antibody
KR20190039577A (en) * 2016-08-23 2019-04-12 메디뮨 리미티드 Anti-VEGF-A and anti-ANG2 antibodies and uses thereof
US20210324062A1 (en) * 2018-10-29 2021-10-21 Hoffmann-La Roche Inc. Antibody formulation
US20220073599A1 (en) * 2020-09-04 2022-03-10 Hoffmann-La Roche Inc. Antibody that binds to vegf-a and ang2 and methods of use
WO2022212360A1 (en) * 2021-03-30 2022-10-06 Abpro Corporation Methods for treating choroidal neovascularization using anti-ang2 x vegf multi-specific antibodies

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150079112A1 (en) * 2013-09-17 2015-03-19 Samsung Electronics Co., Ltd. Use of an anti-ang2 antibody
KR20190039577A (en) * 2016-08-23 2019-04-12 메디뮨 리미티드 Anti-VEGF-A and anti-ANG2 antibodies and uses thereof
US20210324062A1 (en) * 2018-10-29 2021-10-21 Hoffmann-La Roche Inc. Antibody formulation
US20220073599A1 (en) * 2020-09-04 2022-03-10 Hoffmann-La Roche Inc. Antibody that binds to vegf-a and ang2 and methods of use
WO2022212360A1 (en) * 2021-03-30 2022-10-06 Abpro Corporation Methods for treating choroidal neovascularization using anti-ang2 x vegf multi-specific antibodies

Similar Documents

Publication Publication Date Title
US11248052B2 (en) Antigen binding proteins that bind to a complex comprising β-Klotho and an FGF receptor
JP7065822B2 (en) Human antigen-binding protein that binds to β-croto, FGF receptors, and their complexes
TWI816715B (en) Antibodies specific for immunoglobulin-like transcript 3 (ilt3) and uses thereof
JP2017504608A (en) Multifunctional antibody binding to EGFR and MET
US9109031B2 (en) Ligands that bind TGF-β receptor RII
JP7366068B2 (en) Anti-angiopoietin 2 antibody and its use
WO2019164219A1 (en) Anti-angiopoietin-2 antibodies and uses thereof
KR20200130350A (en) Anti-PHF-Tau antibodies and uses thereof
CN112739422B (en) CD200R agonist antibodies and uses thereof
JP2023113599A (en) Bispecific antibody against α-syn/IGF1R and use thereof
CN112480259B (en) anti-TNFR 2 antibodies and uses thereof
WO2024085606A1 (en) Bispecific antibody including first antigen-binding site that specifically binds to human angiopoietin-2, and use thereof
BRPI0414174B1 (en) Antigen-binding antibodies or fragments of the same use thereof, polynucleotide, vector, expression system, and pharmaceutical composition.
CA3233377A1 (en) Proteins binding nkg2d, cd16 and baff-r
US20230242658A1 (en) 4-1bb-binding protein and use thereof
KR20240056422A (en) Bispecific antibody comprising a first antigen binding site that specifically binds to human angiopoietin-2 and Use of the same
Finney et al. Function modifying NA v 1.7 antibodies
AU2021281828A1 (en) Anti-human NR1 antibody derivative
CN113164601A (en) Isolated antigen binding protein and uses thereof
CN113166264A (en) Isolated antigen binding protein and uses thereof
AU2012268396A1 (en) Human antigen binding proteins that bind to a complex comprising beta-Klotho and an FGF receptor