WO2022098080A1 - Nouvel anticorps anti-tie2 ou fragment de liaison à l'antigène de celui-ci et son utilisation - Google Patents

Nouvel anticorps anti-tie2 ou fragment de liaison à l'antigène de celui-ci et son utilisation Download PDF

Info

Publication number
WO2022098080A1
WO2022098080A1 PCT/KR2021/015786 KR2021015786W WO2022098080A1 WO 2022098080 A1 WO2022098080 A1 WO 2022098080A1 KR 2021015786 W KR2021015786 W KR 2021015786W WO 2022098080 A1 WO2022098080 A1 WO 2022098080A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
amino acid
antigen
seq
acid sequence
Prior art date
Application number
PCT/KR2021/015786
Other languages
English (en)
Korean (ko)
Inventor
민정기
이동광
이남경
박종길
황병태
김성식
이장욱
이재민
Original Assignee
한국생명공학연구원
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 KR1020210050134A external-priority patent/KR20220059888A/ko
Application filed by 한국생명공학연구원 filed Critical 한국생명공학연구원
Publication of WO2022098080A1 publication Critical patent/WO2022098080A1/fr

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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]

Definitions

  • the present invention relates to novel anti-Tie2 antibodies or antigen-binding fragments thereof and uses thereof.
  • Tie2 protein is a receptor-type tyrosine kinase specifically expressed in vascular endothelial cells, and is known to be phosphorylated and activated by a specific ligand called Ang1 (Angiopoietin-1), which is a representative multimeric glycoprotein. It is known to play a very important role in angiogenesis, structural and functional normalization and stabilization of blood vessels.
  • Activation of the Tie2 protein by Ang1 can induce structurally and functionally mature blood vessels by itself, and in particular is spotlighted as a therapeutic agent for ischemic diseases.
  • it has the ability to suppress vascular leakage, vascular inflammation, and blood vessel abnormality induced by VEGF, and recently, as a treatment strategy for diabetic retinopathy, sepsis and tumors, abnormal angiogenesis through VEGF inhibition is inhibited
  • Tie2 activation by Ang1 is receiving more attention as a strategy for inducing normalization and stabilization of pre-generated abnormal blood vessels through activation of the Tie2 receptor is suggested.
  • the Ang1 is synthesized in the form of a tetramer in vivo and exhibits physiological activity. Since the Ang1 tetramer is a recombinant protein, it is very difficult to produce, and it is difficult to use as a therapeutic agent because it has a short half-life in vivo. .
  • Ang1 In order to solve the problem of Ang1, research on various Tie2-specific activators that can replace Ang1 is being conducted, but there are problems such as low productivity and concerns about inducing an immune response, so there is a limit to its use as a therapeutic agent. Accordingly, research and development for a Tie2-specific activator capable of replacing Ang1 as described above is still required.
  • One object of the present invention is to provide a novel antibody or antigen-binding fragment thereof that specifically binds to the Tie2 protein.
  • Another object of the present invention is to provide a gene encoding the antibody or antigen-binding fragment thereof, and an expression vector and transformant comprising the gene.
  • Another object of the present invention is to provide a pharmaceutical composition for preventing or treating angiogenesis-related diseases comprising the antibody or antigen-binding fragment thereof.
  • one aspect of the present invention is an amino acid sequence of X 1 -X 2 -X 3 -X 4 -X 5 -X 6 -YX 7 -MHWX 8 (in the amino acid sequence, X 1 to X at least one heavy chain variable region comprising a CDR1-H having each 8 independently any amino acid), and X 17 -X 18 -X 19 -X 20 -X 21 -X 22 -X 23 -NX 24 - an amino acid sequence of X 25 -X 26 -Y (in the amino acid sequence, X 17 to X 20 and X 22 to X 26 are each independently any amino acid, and X 21 is selected from the group consisting of tyrosine, phenylalanine and tryptophan and at least one light chain variable region comprising a CDR1-L having an antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof may specifically bind to human or monkey Tie2 protein.
  • the antibody or antigen fragment thereof may be one that specifically phosphorylates human or monkey Tie2 protein.
  • another aspect of the present invention provides a gene encoding the antibody or antigen-binding fragment thereof, and an expression vector and transformant comprising the gene.
  • another aspect of the present invention provides a pharmaceutical composition for preventing or treating angiogenesis-related diseases comprising the antibody or antigen-binding fragment thereof.
  • novel antibody or antigen-binding fragment of the present invention binds to human or monkey Tie2 protein and phosphorylates it, while in terms of specificity or affinity for the antigen, it is not compatible with the existing ligands (Ang1, Ang2) or antibodies. shows a much better effect than
  • novel antibody or antigen-binding fragment thereof of the present invention is a fully human antibody, it can be applied as an active ingredient of a pharmaceutical composition for humans without fear of rejection, so that angiogenesis is insufficient or abnormal blood vessels are excessive. There is an advantage that can be effectively used for the treatment or prevention of induced angiogenesis-related diseases.
  • FIG. 2 shows the results of confirming the phosphorylation activity of the first and second primary antibodies (Ang1mab1, Ang1mab4 and Ang1mab2-1) on Tie2, AKT, and ERK proteins in HEK293 cells expressing human Tie2 protein.
  • FIG. 4 shows the results of confirming the phosphorylation activity of the first and second primary antibodies (Ang1mab1, Ang1mab4, and Ang1mab2-1) and mutant antibodies on Tie2, AKT, and ERK proteins in HUVEC.
  • FIG. 5 shows the results of confirming that the primary antibody (Ang1mab4) specifically phosphorylates only the Tie2 protein in HUVEC and HEK293 cells expressing human Tie2 protein.
  • 6 to 8 show the results of confirming cross-species cross-linking of the first and second primary antibodies (Ang1mab1, Ang1mab4, and Ang1mab2-1) and the variant antibodies.
  • FIG. 9 schematically shows a variant designed by removing major domains from human Tie2 protein one by one in order to identify antigenic determinants of the antibody of the present invention.
  • Figure 10a shows which domain of the basic antibody (Ang1mab1) binds to the human Tie2 protein
  • Figure 10b shows the results of confirming the specific amino acid sequence of the domain to which the basic antibody (Ang1mab1) binds, respectively.
  • FIG. 11 shows the results of confirming whether the first and second primary antibodies (Ang1mab1, Ang1mab4, and Ang1mab2-1) and mutant antibodies promote the angiogenesis of HUVECs.
  • FIG. 13 shows the results of confirming whether the human Tie2 protein is properly expressed in the blood vessels of the transgenic mouse prepared in a specific example of the present invention.
  • 15 and 16 show whether the primary antibody (Ang1mab4) promotes wound healing through the promotion of angiogenesis by activating the human Tie2 protein expressed in the transgenic mouse, and furthermore, the blood vessels by well surrounding the pericyte. It shows the results of confirming in vivo whether or not to stabilize.
  • FIG. 17 shows the results of confirming in vivo whether the primary antibody (Ang1mab4) can improve lower extremity ischemia through the promotion of angiogenesis by activating human Tie2 protein expressed in transgenic mice.
  • Ang1mab4 vascular endothelial growth factor (VEGF) in HUVECs, the so-called leakage phenomenon.
  • Ang1mab1 the primary antibody
  • Ang1mab1 can increase the influx of anti-tumor immune cells that inhibit tumor growth into tumors by activating human Tie2 protein expressed in transgenic mice to normalize abnormal tumor blood vessels. Shows the results confirmed in Vivo.
  • FIG. 23 shows whether or not tumor growth can be more effectively inhibited when the primary antibody (Ang1mab1) that activates human Tie2 protein and the immune checkpoint inhibitory antibody (Anti-PD-1) that inhibits immune evasion are administered in combination. Shows the results confirmed in vivo.
  • Ang1mab1 that activates human Tie2 protein
  • Anti-PD-1 immune checkpoint inhibitory antibody
  • the term 'antibody' as used herein refers to immunoglobulin molecules and multimers thereof having a structure in which one light chain is connected to each other by a disulfide bond and one light chain to each of two heavy chains connected to each other by a disulfide bond.
  • the light chain contains two regions, one variable region (light chain variable region, V L ) and one constant region (light chain constant region, CL ), and the heavy chain contains four regions, namely one variable region (heavy chain variable region). region, V H ) and three constant regions (heavy chain constant region, C H ; C H 1 , C H 2 and C H 3 ).
  • the constant regions of the light and heavy chains serve to confer biological properties such as binding between the light and/or heavy chains, secretion, complement binding, binding to Fc receptors (FcR), etc.
  • the variable regions of the light and heavy chains are antigen recognition and binding specificity.
  • the binding specificity of the antibody is due to the structural complementarity between the antibody binding site and the antigenic determinant (epitope).
  • Such an antibody binding site is mainly a complementarity determining region contained in the variable regions of the heavy and light chains. Since it consists of residues derived from three hypervariable regions called (complementarity determining region, CDR), such a complementarity determining region is referred to as amino acid sequences defining the binding specificity of an antibody.
  • CDRs are arranged between four relatively well-conserved regions called framework regions (FR), which are FR1, CDR1, FR2, CDR2, They are arranged in the order of FR3, CDR3 and FR4 and are linked.
  • FR framework regions
  • the three CDRs included in the heavy chain variable region are referred to as CDR1-H, CDR2-H and CDR3-H, respectively, and the three CDRs included in the light chain variable region are respectively CDR1-L, CDR2-L and CDR3-H. It is called L.
  • the term 'antigen-binding fragment' refers to a portion of an intact antibody, particularly any polypeptide or glycoprotein comprising an antigen-binding site or variable region of an intact antibody.
  • Antibody-binding fragments as described above may be produced by recombinant DNA techniques or by enzymatic or chemical digestion of intact antibodies, and examples of the antibody-binding fragments include Fv, Fab, F(ab')2, Fab', dsFv, (dsFv)2, scFv, sc(Fv)2, diabodies and the like, as well as bispecific and multispecific antibodies formed therefrom, are included, but are not limited thereto.
  • a 'monoclonal antibody' referred to in the present invention refers to an antibody molecule of a single amino acid sequence, also referred to as a “mAb”, which targets a specific antigen, and is not to be construed as requiring production of the antibody by any special method.
  • monoclonal antibodies may be produced by a monoclonal or hybridoma of B cells, but may also be recombinant.
  • a 'chimeric antibody' as used herein refers to an antibody produced by genetic engineering, which, in its broadest sense, contains one or more sites from one antibody and one or more sites from one or more other antibodies.
  • a chimeric antibody comprises the heavy and light chain variable regions of an antibody derived from a non-human animal such as mouse, rat, hamster, and rabbit together with the heavy and light chain constant regions of a human antibody.
  • a chimeric antibody may also refer to a multispecific antibody that exhibits specificity for at least two different antigens.
  • a 'humanized antibody' as referred to in the present invention is derived wholly or in part from a non-human animal, and to avoid or minimize an immune response in humans, for example, to replace certain amino acids in the framework regions of the variable regions of heavy and light chains. Refers to a modified antibody.
  • the constant regions of humanized antibodies are in most cases the constant regions of human heavy and light chains.
  • the term 'human antibody' is meant to include antibodies having variable and constant regions derived from human immunoglobulin sequences.
  • Human antibodies of the invention are encoded by human immunoglobulin sequences (eg, mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs, in particular in CDR3. It may contain non-amino acid residues.
  • the 'human antibody' referred to in the present invention does not include an antibody in which CDR sequences derived from germlines derived from other non-human animals such as mice are grafted onto human framework sequences.
  • the term 'epitope' refers to a site of an antigen that can specifically bind to an antigen binding site of an antibody known as a paratope.
  • a single antigen may have more than one antigenic determinant, and even if they bind to the same antigen, when the specific site of the antigen to which the antibodies bind is different, these antibodies have different antigenic determinants.
  • the antigenic determinant is usually composed of a group of chemically active surface molecules, such as amino acids or sugar side chains, and generally has specific three-dimensional structural characteristics as well as specific charge characteristics.
  • the antigenic determinant may have a three-dimensional or non-stereoscopic structure, and the three-dimensional antigenic determinant and the non-steric antigenic determinant lose binding to the stereogenic antigenic determinant in the presence of a denaturing solvent, but the binding to the non-steric antigenic determinant is not lost. distinguished in that they do not
  • conservative substitution means that an amino acid residue is substituted with an amino acid residue having a similar side chain. For example, substitution among amino acid residues having basic side chains such as lysine, arginine, and histidine corresponds to a conservative substitution.
  • amino acid residues having acidic side chains such as aspartic acid and glutamic acid
  • amino acid residues having uncharged polar side chains such as glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine
  • amino acid residues having nonpolar side chains such as alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • amino acid residues having ⁇ -branched side chains such as threonine, valine, isoleucine
  • Substitutions among amino acid residues with aromatic side chains such as tyrosine, phenylalanine, tryptophan, and histidine are also conservative substitutions.
  • One aspect of the present invention provides a novel antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof of the present invention comprises at least one heavy chain variable region and at least one light chain variable region.
  • the heavy chain variable region included in the antibody or antigen-binding fragment thereof of the present invention has an amino acid sequence of X 1 -X 2 -X 3 -X 4 -X 5 -X 6 -YX 7 -MHWX 8 (SEQ ID NO: 4) Including CDR1-H, in the amino acid sequence of SEQ ID NO: 4, X 1 to X 8 may each independently be any amino acid.
  • X 1 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith, and X 1 is glycine , may be any one selected from the group consisting of alanine and valine, wherein X 1 may be any one selected from the group consisting of glycine and alanine.
  • X 2 is any one selected from the group consisting of glycine, alanine, valine, isoleucine, leucine, tyrosine, phenylalanine and tryptophan, or any amino acid that can be conservatively substituted therewith , wherein X 2 may be any one selected from the group consisting of alanine, valine, isoleucine, leucine, tyrosine and phenylalanine, wherein X 2 is selected from the group consisting of alanine, valine, isoleucine, leucine, tyrosine and phenylalanine may be any one, wherein X 2 may be any one selected from the group consisting of alanine, isoleucine, leucine, tyrosine and phenylalanine, and X 2 is any one selected from the group consisting of alanine, leucine and phenylalanine can be one
  • X 3 is any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted and X 3 may be any one selected from the group consisting of serine, threonine, alanine, valine, isoleucine and leucine, and X 3 is any one selected from the group consisting of serine, threonine, alanine and valine may be one, and X 3 may be any one selected from the group consisting of threonine and alanine.
  • X 4 is any one selected from the group consisting of glycine, alanine, valine, isoleucine, leucine, tyrosine, phenylalanine and tryptophan, or any amino acid that can be conservatively substituted therewith , wherein X 4 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine, leucine, tyrosine and phenylalanine, and X 4 is selected from the group consisting of glycine, alanine, valine, tyrosine and phenylalanine and X 4 may be any one selected from the group consisting of glycine, alanine, tyrosine and phenylalanine, and X 4 may be any one selected from the group consisting of alanine and phenylalanine .
  • X 5 is any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted and X 5 may be any one selected from the group consisting of serine, threonine, alanine, valine, isoleucine and leucine, and X 5 is any one selected from the group consisting of serine, threonine, alanine and valine may be one, and X 5 may be any one selected from the group consisting of serine and alanine.
  • X 6 is any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted and X 6 may be any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine and valine, and X 6 is serine, threonine, asparagine, glutamine, alanine and valine It may be any one selected from the group consisting of, wherein X 6 may be any one selected from the group consisting of serine, asparagine and alanine.
  • X 7 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith, and X 7 is glycine , and may be any one selected from the group consisting of alanine and valine, wherein X 7 may be any one selected from the group consisting of glycine and alanine, and X 7 may be alanine.
  • X 8 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith, and X 8 is glycine , may be any one selected from the group consisting of alanine and valine, wherein X 8 may be any one selected from the group consisting of alanine and valine.
  • the amino acid sequence of SEQ ID NO: 4 is GLTFSNYAMHWV (SEQ ID NO: 10), ALTFSNYAMHWV (SEQ ID NO: 21), GATFSNYAMHWV (SEQ ID NO: 22), GLTFANYAMHWV (SEQ ID NO: 23), GLTFSNYAMHWA (SEQ ID NO: 24) and GFTFSSYAMHWV (SEQ ID NO: 16) It may be any one amino acid sequence selected from the group consisting of, or an amino acid sequence having 70% or more, 80% or more, 90% or more, 95% or more, or 99% or more homology with the amino acid sequence.
  • the heavy chain variable region included in the antibody or antigen-binding fragment thereof of the present invention may further include CDR2-H comprising the amino acid sequence of IX 9 -YX 10 -X 11 -X 12 -NK (SEQ ID NO: 5). And, in the amino acid sequence of SEQ ID NO: 5, X 9 to X 12 may each independently be any amino acid.
  • X 9 may be any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith and X 9 may be any one selected from the group consisting of serine, threonine, alanine, valine, isoleucine and leucine, and X 9 is any one selected from the group consisting of serine, threonine, alanine and valine and X 9 may be any one selected from the group consisting of serine and alanine.
  • X 10 is any one selected from the group consisting of aspartic acid, glutamic acid, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith, X 10 may be any one selected from the group consisting of aspartic acid, glutamic acid, glycine, alanine and valine, and X 10 may be any one selected from the group consisting of aspartic acid, glutamic acid, alanine and valine. and X 10 may be any one selected from the group consisting of aspartic acid and alanine.
  • X 11 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith, and X 11 is glycine , may be any one selected from the group consisting of alanine and valine, wherein X 11 may be any one selected from the group consisting of glycine and alanine.
  • X 12 is any one selected from the group consisting of aspartic acid, glutamic acid, serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or a conservative substitution thereof It may be any possible amino acid, wherein X 12 may be any one selected from the group consisting of aspartic acid, glutamic acid, serine, threonine, glycine, alanine, valine, isoleucine and leucine, wherein X 12 is aspartic acid, It may be any one selected from the group consisting of glutamic acid, serine, threonine, glycine, alanine and valine, and X 12 may be any one selected from the group consisting of glutamic acid, serine and alanine.
  • the amino acid sequence of SEQ ID NO: 5 is any one amino acid sequence selected from the group consisting of ISYDGENK (SEQ ID NO: 11) and ISYDGSNK (SEQ ID NO: 17), or 70% or more, 80% or more, 90% of the amino acid sequence It may be an amino acid sequence having at least 95% homology, or at least 99% homology.
  • the heavy chain variable region included in the antibody or antigen-binding fragment thereof of the present invention may further include CDR3-H.
  • the CDR3-H may include the amino acid sequence of X 13 -TPKGX 14 -X 15 -LEX 16 (SEQ ID NO: 6) or the amino acid sequence of ARQYSGVFEY (SEQ ID NO: 18), wherein X 13 in the amino acid sequence of SEQ ID NO: 6 to X 16 may each independently be any amino acid.
  • X 13 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith, and X 13 is glycine , and may be any one selected from the group consisting of alanine and valine, wherein X 13 may be any one selected from the group consisting of glycine and alanine, and X 13 may be alanine.
  • X 14 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith, and X 14 is glycine , may be any one selected from the group consisting of alanine and valine, wherein X 14 may be any one selected from the group consisting of glycine and alanine.
  • X 15 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith, and X 15 is glycine , and may be any one selected from the group consisting of alanine and valine, wherein X 15 may be any one selected from the group consisting of glycine and alanine, and X 15 may be alanine.
  • X 16 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith, and X 16 is alanine , may be any one selected from the group consisting of valine, isoleucine and leucine, wherein X 16 may be any one selected from the group consisting of alanine and isoleucine.
  • amino acid sequence of SEQ ID NO: 6 is the amino acid sequence of ATPKGGALEI (SEQ ID NO: 12), or an amino acid sequence having 70% or more, 80% or more, 90% or more, 95% or more, or 99% or more homology to the amino acid sequence can
  • the CDR3-H is the amino acid sequence of ARQYSGVFEY (SEQ ID NO: 18) or the amino acid sequence of SEQ ID NO: 18 and 70% or more, 80% or more, 90% or more, 95% or more, or an amino acid sequence having 99% or more homology may include
  • the heavy chain variable region included in the antibody or antigen-binding fragment thereof of the present invention is FR1-H, RQAPGKGLEWVAV (SEQ ID NO: 30) having an amino acid sequence of QVQLVQSGGGVVQPGRSLRLSCAAS (SEQ ID NO: 29) or an amino acid sequence of QVQLVESGGGVVQPGRSLRLSCAAS (SEQ ID NO: 37) ) FR2-H, YYADSVKGRFTISRDNSKNMLQLQMNRLRSEDTAVYYC (SEQ ID NO: 31) or YYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC (SEQ ID NO: 38), FR3-H, and WGQGTLITVVITVSS (SEQ ID NO: 32) 39) may include at least one framework selected from the group consisting of FR4-H having the amino acid sequence.
  • the heavy chain variable region included in the antibody or antigen-binding fragment thereof of the present invention may have
  • the light chain variable region included in the antibody or antigen-binding fragment thereof of the present invention is X 17 -X 18 -X 19 -X 20 -X 21 -X 22 -X 23 -NX 24 -X 25 -X 26 - a CDR1-L having the amino acid sequence of Y (SEQ ID NO: 7), wherein in the amino acid sequence of SEQ ID NO: 7, X 17 to X 20 and X 22 to X 26 are each independently any amino acid, and X 21 is tyrosine , may be any one selected from the group consisting of phenylalanine and tryptophan, or any amino acid that can be conservatively substituted therewith.
  • X 17 is any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted and X 17 may be any one selected from the group consisting of asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, and X 17 is selected from the group consisting of asparagine, glutamine, alanine and valine. and X 17 may be any one selected from the group consisting of glutamine and alanine.
  • X 18 is any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine, and leucine, or any amino acid that can be conservatively substituted and X 18 may be any one selected from the group consisting of serine, threonine, glycine, alanine, valine, isoleucine and leucine, and X 18 is selected from the group consisting of serine, threonine, alanine and valine and X 18 may be any one selected from the group consisting of serine and alanine.
  • X 19 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine or any amino acid that can be conservatively substituted therewith, and X 19 is glycine , may be any one selected from the group consisting of alanine and valine, wherein X 19 may be any one selected from the group consisting of alanine and valine.
  • X 20 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine or any amino acid that can be conservatively substituted therewith, and X 20 is alanine , may be any one selected from the group consisting of valine, isoleucine and leucine, wherein X 20 may be any one selected from the group consisting of alanine and leucine.
  • X 21 may be any one selected from the group consisting of tyrosine, phenylalanine and tryptophan, or any amino acid that can be conservatively substituted therewith, and X 21 is tyrosine and phenylalanine It may be any one selected from the group being
  • X 22 is any one selected from the group consisting of arginine, histidine, lysine, serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or a conservative substitution thereof may be any possible amino acid, wherein X 22 may be any one selected from the group consisting of arginine, lysine, serine, threonine, glycine, alanine, valine, isoleucine and leucine, and X 22 is arginine, lysine , may be any one selected from the group consisting of serine, threonine, glycine, alanine and valine, wherein X 22 may be any one selected from the group consisting of arginine, lysine, serine, threonine, alanine and valine, , wherein X 22 may be any one selected from the group consisting of arginine,
  • X 23 is any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted and X 23 may be any one selected from the group consisting of serine, threonine, glycine, alanine, valine, isoleucine and leucine, and X 23 is selected from the group consisting of serine, threonine, alanine and valine and X 23 may be any one selected from the group consisting of serine and alanine.
  • X 24 is any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted and X 24 may be any one selected from the group consisting of asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, and X 24 is asparagine, glutamine, glycine, alanine, and valine. may be any one selected from, and X 24 may be any one selected from the group consisting of asparagine and alanine.
  • X 25 is any one selected from the group consisting of arginine, histidine, lysine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith , wherein X 25 may be any one selected from the group consisting of arginine, lysine, glycine, alanine, valine, isoleucine and leucine, and X 25 is selected from the group consisting of arginine, lysine, glycine, alanine and valine and X 25 may be any one selected from the group consisting of arginine, lysine, alanine and valine, and X 25 is any one selected from the group consisting of arginine, lysine and alanine can
  • X 26 is any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted and X 26 may be any one selected from the group consisting of asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, and X 26 is asparagine, glutamine, glycine, alanine, and valine. may be any one selected from, and X 26 may be any one selected from the group consisting of asparagine and alanine.
  • the amino acid sequence of SEQ ID NO: 7 is QSVLYSSNNKNY (SEQ ID NO: 13), ASVLYSSNNKNY (SEQ ID NO: 25), QSALYSSNNKNY (SEQ ID NO: 26), QSVLYSANNKNY (SEQ ID NO: 27), QSVLFSSNNKNY (SEQ ID NO: 28) and QSVLYRSNNRNY (SEQ ID NO: 19) It may be any one amino acid sequence selected from the group consisting of, or an amino acid sequence having 70% or more, 80% or more, 90% or more, 95% or more, or 99% or more homology with the amino acid sequence.
  • the light chain variable region included in the antibody or antigen-binding fragment thereof of the present invention may further include a CDR-L2 comprising the amino acid sequence of WX 27 -X 28 (SEQ ID NO: 8), and the amino acid of SEQ ID NO: 8
  • X 27 and X 28 may each independently be any amino acid.
  • X 27 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted therewith, and X 27 is glycine , may be any one selected from the group consisting of alanine and valine, the X 27 may be any one selected from the group consisting of glycine and alanine, the X 27 may be alanine.
  • X 28 is any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted and X 28 may be any one selected from the group consisting of serine, threonine, glycine, alanine, valine, isoleucine and leucine, and X 28 is selected from the group consisting of serine, threonine, alanine and valine and X 28 may be any one selected from the group consisting of serine and alanine.
  • the amino acid sequence of SEQ ID NO: 8 is the amino acid sequence of WAS (SEQ ID NO: 14), or 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more or 90% of the amino acid sequence It may be an amino acid sequence having more than one homology.
  • the light chain variable region included in the antibody or antigen-binding fragment thereof of the present invention may further include a CDR3-L comprising the amino acids of QQYX 29 -SX 30 -PX 31 -X 32 (SEQ ID NO: 9), ,
  • X 30 and X 32 may each independently be any amino acid
  • X 29 is any one selected from the group consisting of tyrosine, phenylalanine and tryptophan, or any one in which conservative substitution is possible
  • It may be an amino acid
  • X 31 may be any one selected from the group consisting of isoleucine and tyrosine, or any amino acid that can be conservatively substituted therewith.
  • X 29 may be any one selected from the group consisting of tyrosine, phenylalanine and tryptophan or any amino acid that can be conservatively substituted therewith, and X 29 is composed of tyrosine and phenylalanine It may be any one selected from the group being
  • X 30 may be any one selected from the group consisting of glycine, alanine, valine, isoleucine and leucine or any amino acid that can be conservatively substituted therewith, and X 30 is alanine , may be any one selected from the group consisting of valine, isoleucine and leucine, wherein X 20 may be any one selected from the group consisting of alanine and isoleucine.
  • X 31 may be any one selected from the group consisting of isoleucine and tyrosine, or any amino acid capable of conservative substitution therewith.
  • X 32 is any one selected from the group consisting of serine, threonine, asparagine, glutamine, glycine, alanine, valine, isoleucine and leucine, or any amino acid that can be conservatively substituted and X 32 may be any one selected from the group consisting of serine, threonine, alanine, valine, isoleucine and leucine, and X 32 is any one selected from the group consisting of serine, threonine, alanine and valine may be one, and X 32 may be any one selected from the group consisting of threonine and alanine.
  • the amino acid sequence of SEQ ID NO: 9 is any one amino acid sequence selected from the group consisting of QQYYSAPIT (SEQ ID NO: 15) and QQYFSIPYT (SEQ ID NO: 20), or 70% or more, 80% or more, 90% of the amino acid sequence It may be an amino acid sequence having at least 95% homology, or at least 99% homology.
  • the light chain variable region included in the antibody or antigen-binding fragment thereof of the present invention is FR1-L, LAWYQQKPGQPPNLLMY (SEQ ID NO: 34) having the amino acid sequence of DIQMTQSPDSLAVSLGERATINCKSS (SEQ ID NO: 33), or LAWYQQKLGQPPQLLIY (SEQ ID NO: 40) ), FR2-L having the amino acid sequence of TRESGVPDRFSGSGTDFTLTISSLQAEDVAVYYC (SEQ ID NO: 35) or FR3-L having the amino acid sequence of TRESGVPDRFSGSGSGADFTLTISSLQAEDVAVYYC (SEQ ID NO: 41), and FGQGTRLEIK (SEQ ID NO: 36) 42) may include at least one framework selected from the group consisting of FR4-L having the amino acid sequence.
  • the light chain variable region included in the antibody or antigen-binding fragment thereof of the present invention may have the amino acid sequence of SEQ ID NO:
  • the antibody or antigen-binding fragment thereof of the present invention may be a humanized antibody or human antibody or antigen-binding fragment thereof, and in particular, a human antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof of the present invention is a heavy chain constant region comprising any one amino acid sequence selected from the group consisting of the amino acid sequence of SEQ ID NO: 47 and the amino acid sequence of SEQ ID NO: 49 and/or SEQ ID NO: 51 It may include a light chain constant region including an amino acid sequence.
  • the antibody or antigen-binding fragment thereof of the present invention may include a plurality of each of the heavy chain variable region and the light chain variable region.
  • the antibody or antigen-binding fragment thereof of the present invention is a bivalent antibody comprising two each of the heavy chain variable region and the light chain variable region, or 4 each comprising four of the heavy chain variable region and the light chain variable region may be an antibody.
  • the antibody or antigen-binding fragment thereof of the present invention may specifically bind to the amino acid sequence of SEQ ID NO: 3, particularly, the amino acid sequence of SEQ ID NO: 54.
  • the amino acid sequence of SEQ ID NO: 3, particularly the amino acid sequence of SEQ ID NO: 54 is an antigenic determinant of the antibody or antigen-binding fragment thereof of the present invention.
  • the antibody or antigen-binding fragment thereof of the present invention can specifically bind to a polypeptide or protein comprising the amino acid sequence of SEQ ID NO: 3, in particular, to a polypeptide or protein comprising the amino acid sequence of SEQ ID NO: 54.
  • the antibody or antigen-binding fragment thereof of the present invention can also specifically bind to the amino acid sequence of SEQ ID NO: 55.
  • the amino acid sequence of SEQ ID NO: 55 is also another antigenic determinant of the antibody or antigen-binding fragment thereof of the present invention.
  • the antibody or antigen-binding fragment thereof of the present invention can also specifically bind to a polypeptide or protein comprising the amino acid sequence of SEQ ID NO: 55.
  • the antibody or antigen-binding fragment thereof of the present invention comprises the amino acid sequence of SEQ ID NO: 3, in particular, the amino acid sequence of SEQ ID NO: 54; Alternatively, the amino acid sequence of SEQ ID NO: 55; may also specifically bind to a polypeptide or protein comprising the. the amino acid sequence of SEQ ID NO: 3, particularly the amino acid sequence of SEQ ID NO: 54; Alternatively, the polypeptide or protein comprising the amino acid sequence of SEQ ID NO: 55 is typically a Tie2 protein or an Ig2 domain thereof.
  • the Tie2 protein may be derived from mammals, such as humans, monkeys, rats, mice, dogs, cats, rabbits, pigs, horses, cattle, sheep, goats, and the like, and among them, may be derived from humans, monkeys, etc. .
  • the Tie2 protein means that it is derived from a human, and in particular, the human-derived Tie2 protein may have the amino acid sequence of SEQ ID NO: 1, and its Ig2 domain is the amino acid of SEQ ID NO: 2 It may have a sequence.
  • the amino acid sequence of SEQ ID NO: 3 is the 116th to 135th region in the amino acid sequence of SEQ ID NO: 1
  • the amino acid sequence of SEQ ID NO: 54 is the 131st to 135th region in the amino acid sequence of SEQ ID NO: 1
  • the amino acid sequence of SEQ ID NO: 55 is the region of 194th to 208th in the amino acid sequence of SEQ ID NO: 1.
  • the antibody or antigen-binding fragment thereof of the present invention comprises a Tie2 protein or an Ig2 domain thereof, in particular a human Tie2 protein or an Ig2 domain thereof, inter alia the amino acid sequence of SEQ ID NO: 3 or the amino acid sequence of SEQ ID NO: 55, inter alia SEQ ID NO: It may specifically bind to the amino acid sequence of 54 or the amino acid sequence of SEQ ID NO: 55.
  • the antibody or antigen-binding fragment thereof of the present invention may have a dissociation constant (K D ) value of 10 nM or less, 7 nM or less, 5 nM or less, 3 nM or less, 2 nM or less, 1.7 nM or less, or 1.5 nM or less.
  • K D dissociation constant
  • the antibody or antigen-binding fragment thereof of the present invention is 1.4nM or less, 1.3nM or less, 1.2nM or less, 1.1nM or less, 1.0nM or less, 0.97nM or less, 0.95nM or less, 0.93nM or less, 0.90nM or less, 0.87 or less nM or less, 0.85 nM or less, 0.83 nM or less, 0.80 nM or less, 0.77 nM or less, 0.75 nM or less, 0.73 nM or less, 0.70 nM or less, 0.67 nM or less, 0.65 nM or less, 0.63 nM or less, 0.60 nM or less, 0.57 nM or less , 0.55 nM or less, 0.53 nM or less, 0.50 nM or less, 0.47 nM or less, 0.45 nM or less, 0.43 nM or less, 0.40 nM or less, 0.37 nM or less, 0.35
  • the antibody or antigen-binding fragment thereof of the present invention is 1 ⁇ 10 4 M -1 s -1 or more, 2 ⁇ 10 4 M -1 s -1 or more, 3 ⁇ 10 4 M -1 s -1 or more, 4 A binding rate constant of at least ⁇ 10 4 M -1 s -1 , at least 5 ⁇ 10 4 M -1 s -1 , at least 6 ⁇ 10 4 M -1 s -1 , or at least 7 ⁇ 10 4 M -1 s -1 ( K on ) may have a value.
  • the antibody or antigen-binding fragment thereof of the present invention is 7.3 ⁇ 10 4 M -1 s -1 or more, 7.5 ⁇ 10 4 M -1 s -1 or more, 7.7 ⁇ 10 4 M -1 s -1 or more, 8.0 ⁇ 10 4 M -1 s -1 or more, 8.3 ⁇ 10 4 M -1 s -1 or more, 8.5 ⁇ 10 4 M -1 s -1 or more, 8.7 ⁇ 10 4 M -1 s -1 or more, 8.8 ⁇ 10 4 M -1 s -1 or more, 8.9 ⁇ 10 4 M -1 s -1 or more, 9.0 ⁇ 10 4 M -1 s -1 or more, 9.1 ⁇ 10 4 M -1 s -1 or more, 9.2 ⁇ 10 4 M -1 s -1 or more, 9.3 ⁇ 10 4 M -1 s -1 or more, 9.4 ⁇ 10 4 M -1 s -1 or more, 9.5 ⁇ 10 4 M -1 s -1 or more, 9.6 ⁇ 10 4 M -1 s
  • the antibody or antigen-binding fragment thereof of the present invention is 1 ⁇ 10 -3 s -1 or less, 7 ⁇ 10 -4 s -1 or less, 5 ⁇ 10 -4 s -1 or less, 3 ⁇ 10 -4 s - 1 or less or 2 ⁇ 10 ⁇ 4 s ⁇ 1 It may have a dissociation rate constant (K off ) value of ⁇ 1 or less.
  • the antibody or antigen-binding fragment thereof of the present invention is 1.7 ⁇ 10 -4 s -1 or less, 1.5 ⁇ 10 -4 s -1 or less, 1.0 ⁇ 10 -4 s -1 or less, 9.7 ⁇ 10 -5 s - 1 or less, 9.5 ⁇ 10 -5 s -1 or less, 9.3 ⁇ 10 -5 s -1 or less, 9.0 ⁇ 10 -5 s -1 or less, 8.7 ⁇ 10 -5 s -1 or less, 8.5 ⁇ 10 -5 s - 1 or less, 8.3 ⁇ 10 -5 s -1 or less, 8.0 ⁇ 10 -5 s -1 or less, 7.7 ⁇ 10 -5 s -1 or less, 7.5 ⁇ 10 -5 s -1 or less , 7.3 ⁇ 10 -5 s -1 or less 1 or less, 7.0 ⁇ 10 -5 s -1 or less, 6.7 ⁇ 10 -5 s -1 or less, 6.5 ⁇ 10 -5 s -1 or less, 6.3 ⁇ 10 -5 s -1 or
  • the antibody or antigen-binding fragment thereof of the present invention may have a melting temperature (T m ) of 70°C or higher, 73°C or higher, 75°C or higher, 77°C or higher, or 80°C or higher.
  • T m melting temperature
  • the antibody or antigen-binding fragment thereof of the present invention is 81 ° C. or higher, 82 ° C. or higher, 83 ° C. or higher, 84 ° C. or higher, 85 ° C. or higher, 86 ° C. or higher, 87 ° C. or higher, 88 ° C. or higher, 89 ° C.
  • 90 It may have a melting temperature (T m ) of °C or higher, 91 °C or higher, 92 °C or higher, 93 °C or higher, 94 °C or higher, or 95 °C or higher.
  • T m melting temperature
  • the antibody or antigen-binding fragment thereof of the present invention binds to the Tie2 protein comprising the amino acid sequence of SEQ ID NO: 3, the Tie2 protein is self-phosphorylated regardless of binding to its ligands Ang1, Ang2, etc. can be, thereby activating a signal transduction pathway in the cell. Accordingly, the antibody or binding fragment thereof of the present invention may serve to activate the Tie2 protein by replacing the ligand of the Tie2 protein.
  • Another aspect of the present invention provides a gene encoding all or a part of the antibody or antigen-binding fragment thereof of the present invention.
  • a gene encoding the heavy chain variable region of the antibody or antigen-binding fragment thereof of the present invention and a gene encoding the light chain variable region of the antibody or antigen-binding fragment thereof of the present invention may be provided, respectively.
  • the gene encoding the heavy chain variable region may, for example, encode the amino acid sequence of SEQ ID NO: 43 or the amino acid sequence of SEQ ID NO: 44, and the gene encoding the light chain variable region may include, for example, the amino acid sequence of SEQ ID NO: 45 or SEQ ID NO: 46 It may be one that encodes the amino acid sequence of In the gene encoding the variable region of the heavy or light chain, various modifications may be made to the coding region within a range that does not change the amino acid sequence of the variable regions expressed from the coding region, and the expression of the gene is also performed in parts other than the coding region.
  • nucleic acid bases may be mutated by substitution, deletion, insertion, or a combination thereof, and such mutated genes are also included within the scope of the present invention. do.
  • a gene encoding each of the CDRs included in the heavy chain variable region of the antibody or antigen-binding fragment thereof of the present invention Specifically, a gene encoding CDR1-H, a gene encoding CDR2-H, and a gene encoding CDR3-H are each provided individually.
  • the gene encoding the CDR1-H may encode the amino acid sequence of SEQ ID NO: 4, for example, the gene encoding the CDR1-H is the amino acid sequence of SEQ ID NO: 10, the amino acid sequence of SEQ ID NO: 21, SEQ ID NO: It may encode any one amino acid sequence selected from the group consisting of the amino acid sequence of 22, the amino acid sequence of SEQ ID NO: 23, the amino acid sequence of SEQ ID NO: 24 and SEQ ID NO: 16.
  • the gene encoding the CDR2-H may encode the amino acid sequence of SEQ ID NO: 5, for example, the amino acid sequence of SEQ ID NO: 11 or the amino acid sequence of SEQ ID NO: 17.
  • the gene encoding the CDR3-H may encode the amino acid sequence of SEQ ID NO: 6, particularly, the amino acid sequence of SEQ ID NO: 12, or encode the amino acid sequence of SEQ ID NO: 18.
  • a gene encoding each of the CDRs included in the light chain variable region of the antibody or antigen-binding fragment thereof of the present invention Specifically, a gene encoding CDR1-L, a gene encoding CDR2-L and a gene encoding CDR3-L are each provided individually.
  • the gene encoding the CDR1-L may encode the amino acid sequence of SEQ ID NO: 7, for example, the gene encoding the CDR1-L includes the amino acid sequence of SEQ ID NO: 13, the amino acid sequence of SEQ ID NO: 25, and SEQ ID NO: It may encode any one amino acid sequence selected from the group consisting of the amino acid sequence of 26, the amino acid sequence of SEQ ID NO: 27, the amino acid sequence of SEQ ID NO: 28, and the amino acid sequence of SEQ ID NO: 19.
  • the gene encoding the CDR2-L may be one encoding the amino acid sequence of SEQ ID NO: 8, particularly the amino acid sequence of SEQ ID NO: 14, and the gene encoding the CDR3-L is the amino acid sequence of SEQ ID NO: 9, particularly the sequence It may be one encoding the amino acid sequence of No. 15 or the amino acid sequence of SEQ ID NO: 20.
  • nucleic acid bases may be mutated by substitution, deletion, insertion, or a combination thereof as long as the gene of the CDRs encodes a protein having an equivalent activity, and such a mutated gene is also included in the scope of the present invention.
  • another aspect of the present invention provides a recombinant expression vector containing a gene encoding all or part of the antibody or antigen-binding fragment thereof of the present invention, and a transformant into which the expression vector is introduced.
  • the recombinant expression vector of the present invention includes a gene encoding all or part of the antibody or antigen-binding fragment thereof of the present invention, and the gene may be operably linked to a promoter.
  • the expression vector includes, but is not limited to, a plasmid vector, a cosmid vector, a bacteriophage vector, and a viral vector.
  • the recombinant expression vector of the present invention is a promoter, terminator, enhancer, etc. according to the type of host cell in which all or part of the antibody or antigen-binding fragment thereof of the present invention is to be expressed or produced.
  • the same expression control sequence, or a sequence for secretion, etc. can be appropriately combined depending on the purpose.
  • the expression vector may further include a selection marker for selecting a host cell into which the vector is introduced, and in the case of an expression vector capable of replication, it may include an origin of replication.
  • the recombinant expression vector may include a sequence for facilitating purification of the expression protein, specifically, a tag for separation and purification so as to be operable on a gene encoding all or part of the antibody or antigen-binding fragment thereof of the present invention Genes encoding for can be linked.
  • a tag for separation and purification GST, poly-Arg, FLAG, histidine-tag (His-tag), c-myc, etc. may be used alone or two or more of them may be sequentially connected.
  • the gene encoding all or part of the antibody or antigen-binding fragment thereof of the present invention may be cloned through a restriction enzyme cleavage site, and when a gene encoding a protease recognition site is used in the vector, the present invention It is linked in frame with the gene encoding all or part of the antibody or antigen-binding fragment thereof of the present invention, and after obtaining all or part of the antibody or antigen-binding fragment thereof of the present invention, cleavage with a proteolytic enzyme In this case, all or part of the antibody or antigen-binding fragment thereof of the present invention in its original form can be produced.
  • a recombinant expression vector containing a gene encoding all or part of the antibody or antigen-binding fragment thereof of the present invention is introduced into the transformant of the present invention.
  • Transformants can be prepared by transforming the recombinant expression vector according to the present invention into any suitable host cell selected from the group consisting of bacteria, yeast, E. coli, fungi, plant cells and animal cells depending on the purpose of expression.
  • the host cell may be Escherichia coli ( E. coli BL21(DE3), DH5 ⁇ , etc.) or yeast cells ( Saccharomyces genus, Pichia genus, etc.).
  • an appropriate culture method and medium conditions according to the type of host cell can be easily selected by those skilled in the art from known techniques in the art.
  • a known technique that is, a heat shock method, an electric shock method, or the like may be used.
  • the protein expressed from the transformant is the antibody or antigen-binding fragment thereof of the present invention
  • mass production of the antibody or antigen-binding fragment thereof of the present invention is facilitated by mass culturing the transformant to express the gene. can do.
  • Another aspect of the present invention provides a pharmaceutical composition for preventing or treating angiogenesis-related diseases comprising the antibody or antigen-binding fragment thereof of the present invention as an active ingredient.
  • the angiogenesis-related disease refers to the formation or growth of new blood vessels from existing blood vessels, and the angiogenesis-related disease refers to a disease related to the occurrence or progression of angiogenesis. As long as it can be prevented or treated by the antibody or antigen-binding fragment thereof of the present invention, the disease may be included in the scope of the angiogenesis-related disease without limitation.
  • the antibody or antigen-binding fragment thereof of the present invention can bind to the Tie2 protein comprising the amino acid sequence of SEQ ID NO: 3 to self-phosphorylate the Tie2 protein, thereby transducing signals involved in angiogenesis in cells path can be activated.
  • angiogenesis-related signal transduction pathways are activated due to phosphorylation of Tie2 protein, necessary angiogenesis is promoted (Fukuhara et al. , Histol. Histopathol. , 25 , 387-396 (2010)), abnormally The blood vessels that have been generated are matured and stabilized (Koh, Trends Mol. Med. , 19 (1), 31-39 (2013); Anisimov et al.
  • the antibody or antigen-binding fragment thereof of the present invention can be used as an active ingredient in the prevention or treatment of diseases caused by insufficient angiogenesis or diseases caused by abnormal blood vessels.
  • the angiogenesis-related disease may be a disease caused by insufficient angiogenesis.
  • diseases caused by insufficient angiogenesis include, for example, ischemia occurring in the brain, heart, and peripheral tissues, cerebral infarction, myocardial infarction, lower extremity ischemia, wound delay, foot ulcer, retinopathy of prematurity, and erectile dysfunction. , organ transplantation, etc., but is not limited thereto.
  • the angiogenesis-related disease may be a disease caused by abnormal blood vessels or excessive abnormal angiogenesis.
  • diseases caused by abnormal blood vessels or excessive abnormal angiogenesis may be caused by leakage of blood vessels, inflammation, or local ischemia, for example, cancer, metastatic cancer, diabetic retinopathy, macular degeneration, sepsis, Angiogenic glaucoma, psoriasis, rheumatoid arthritis, diabetic nephropathy, diabetic foot ulcers, vascular adhesion, atherosclerosis, It may be capillary formation of atherosclerotic plaques, acute liver failure, acute renal failure, acute lung failure, systemic inflammatory reaction syndrome, and the like, but is not limited thereto.
  • the prophylaxis refers to any measure that suppresses or delays the onset of a disease.
  • the treatment refers to all measures to improve or ameliorate the symptoms of the expressed disease, and desirable effects of the treatment include prevention of the occurrence or recurrence of the disease, alleviation of symptoms, any direct or indirect pathological pathology of the disease. reducing outcome, preventing metastasis, reducing the rate of disease progression, ameliorating or alleviating the disease state, and remission or improved prognosis.
  • the pharmaceutical composition may further include a pharmaceutically acceptable carrier or additive in addition to the antibody or antigen-binding fragment thereof of the present invention.
  • the meaning of 'pharmaceutically acceptable' means that it does not inhibit the activity of the active ingredient and does not have more toxicity than the application (prescription) target can adapt to.
  • the carrier means a compound that facilitates the addition of the antibody or antigen-binding fragment thereof of the present invention into a cell or tissue, and the additive is any component necessary to prepare a pharmaceutical composition into the required dosage form, such as an excipient , means a disintegrant, a binder, a flavoring agent, an antiseptic, a lubricant, a stabilizer, a viscous agent, and the like.
  • the pharmaceutical composition may be administered in combination with therapeutic agents for each disease.
  • the pharmaceutical composition may further include at least one therapeutic agent for each disease in addition to the antibody or antigen-binding fragment thereof of the present invention.
  • the other treatment for each disease may include an anti-angiogenic drug, an anti-inflammatory drug, and/or an anti-cancer drug.
  • another aspect of the present invention provides a method for preventing or treating angiogenesis-related diseases, comprising administering the antibody or antigen-binding fragment thereof of the present invention, or the pharmaceutical composition to a subject in need thereof.
  • the subject may be a mammal, for example a human, and in particular a patient.
  • the 'individual in need' refers to an individual suffering from, or likely to suffer from, an angiogenesis-related disease.
  • the antibody or antigen-binding fragment thereof of the present invention, or the pharmaceutical composition may be administered in a pharmaceutically effective amount.
  • the 'pharmaceutically effective amount' means an amount sufficiently exhibiting the effect of prevention or treatment with a reasonable benefit/risk ratio applicable to all medical treatments.
  • the 'pharmaceutically effective amount' refers to the severity of the disease to be treated, the age and sex of the patient, the type of disease, the activity of the drug, the sensitivity to the drug, the administration time, the administration route, the secretion rate, the treatment period, the drug depends on a variety of factors, including co-administration of the drug and other parameters known in the art.
  • the pharmaceutical composition may be administered alone or in combination with other therapies. In such cases, they may be administered sequentially or simultaneously with conventional therapy.
  • the composition may be administered in a single dose or divided into multiple doses. When these factors are fully considered, it is important to administer a minimum amount sufficient to obtain the maximum effect without side effects, and this dosage can be readily determined by a person skilled in the art.
  • the dosage of the pharmaceutical composition is not particularly limited, but varies according to various factors including the patient's health condition and weight, the severity of the disease, the type of drug, the administration route, and the administration time.
  • the pharmaceutical composition may be administered in single or multiple doses per day by the typically accepted route into mammals, including humans, such as orally, rectally, intravenously, subcutaneously, intraperitoneally. It can be administered (intraperitoneally), intrauterinely (intrauterinely) or intracerebrovascularly (intracerebrovascularly).
  • the antibody or antigen-binding fragment thereof of the present invention, or the pharmaceutical composition may be administered in combination with other therapeutic agents for angiogenesis-related diseases.
  • the antibody or antigen-binding fragment thereof of the present invention, or the pharmaceutical composition may be administered simultaneously or sequentially with other therapeutic agents such as anti-angiogenic drugs, anti-inflammatory drugs and/or anti-cancer drugs.
  • the antibody or antigen-binding fragment thereof, or the pharmaceutical composition of the present invention is added to the subject.
  • other therapeutic agents such as anti-angiogenic drugs, anti-inflammatory drugs and/or anti-cancer drugs are additionally administered to the subject can be administered.
  • the antibody or antigen-binding fragment thereof of the present invention, or the pharmaceutical composition and other therapeutic agents such as anti-angiogenic drugs, anti-inflammatory drugs and / or anti-cancer drugs may be administered to the subject at the same time.
  • the amino acid sequences of CDR1-H, CDR2-H and CDR3-H are designed as shown in Table 1 below, and each of them is FR1-H having the amino acid sequence of SEQ ID NO: 29, FR2-H having the amino acid sequence of SEQ ID NO: 30,
  • the first primary antibody, Ang1mab1 is interposed in the order of FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4 between FR3-H having the amino acid sequence of SEQ ID NO: 31 and FR4-H having the amino acid sequence of SEQ ID NO: 32 and the heavy chain variable region of Ang1mab4 were designed.
  • amino acid sequences of CDR1-L, CDR2-L and CDR3-L are designed as shown in Table 2 below, and each of them is FR1-L having the amino acid sequence of SEQ ID NO: 33, FR2- having the amino acid sequence of SEQ ID NO: 34 L, FR3-L having the amino acid sequence of SEQ ID NO: 35 and FR4-L having the amino acid sequence of SEQ ID NO: 36 Interposed in the order of FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4, the first primary antibody
  • the light chain variable regions of Ang1mab1 and Ang1mab4 were designed.
  • the light chain variable region designed as described above was arranged at the N-terminus of the light chain constant region (CL) having the amino acid sequence of SEQ ID NO: 51 to design the light chains of the first primary antibodies Ang1mab1 and Ang1mab4.
  • the amino acid sequences of CDR1-H, CDR2-H and CDR3-H are designed as shown in Table 3 below, and each of them is FR1-H having the amino acid sequence of SEQ ID NO: 37, FR2-H having the amino acid sequence of SEQ ID NO: 30;
  • the heavy chain of the Ang1mab2-1 antibody by interposing in the order of FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4 between FR3-H having the amino acid sequence of SEQ ID NO: 38 and FR4-H having the amino acid sequence of SEQ ID NO: 39 Variable regions were constructed.
  • amino acid sequences of CDR1-L, CDR2-L and CDR3-L are designed as shown in Table 4 below, and each of them is FR1-L having the amino acid sequence of SEQ ID NO: 33, FR2- having the amino acid sequence of SEQ ID NO: 40
  • the light chain variable region of Ang1mab2-1 was constructed.
  • the heavy chain variable region designed as described above the N-terminus of the heavy chain constant region having the amino acid sequence of SEQ ID NO: 47, in which the three constant regions are arranged in the order of C H 1-hinge-C H 2 -C H 3 to design the heavy chain of the second primary antibody Ang1mab2-1.
  • the light chain variable region designed as described above was arranged at the N-terminus of the light chain constant region (CL) having the amino acid sequence of SEQ ID NO: 51 to design the light chain of the second basic antibody Ang1mab2-1.
  • a total of 100 ug of each 50 ug each of the heavy and light chain expression vectors was diluted in 6 ml of Opti-MEM medium, and 320 ul of the ExpiFectamine TM 293 Transfection Kit reagent (ExpiFectamine TM 293 Reagent) was diluted in 6 ml of Opti-MEM medium and allowed to stand for 5 minutes each. .
  • the vector-added medium and the reagent-added medium were mixed and left still for 10 minutes, then added to 100 ml of Expi293F TM cells (2.0x10 6 cells/ml) and suspended in culture for 24 hours.
  • ExpiFectamine TM 293 Transfection Enhancer was added to the cultured cells as described above and further cultured for 4 days, then centrifuged at 10,000xg for 30 minutes and filtered through a 0.2um filter to recover the supernatant. Then, the protein expressed in the supernatant was bound to an affinity column (MabSelect prismA) using an AKTA purifier100 instrument, and then eluted with an acidic eluate (50 mM acetic acid buffer, pH 4.0). The protein eluted as described above was finally purified with PBS buffer using a desalting column (Hiprep 26/10).
  • the purified protein as described above is an antibody having a total molecular weight of about 150 kDa having a heavy chain (about 50 kDa) and a light chain (about 25 kDa) through SDS-PAGE analysis under non-reducing and reducing conditions. It was confirmed that
  • alanine scanning technique As a representative technique for confirming the contribution of a specific residue to the stability and function of a protein, there is an alanine scanning technique. Alanine is not large in size, is chemically inactive, and has a methyl functional group that mimics the secondary structure preference of other amino acids. It is a technique to confirm the contribution of a specific amino acid by checking how the function changes. For example, if there is no significant change in the stability or function of a protein despite the substitution of a specific amino acid with alanine, the specific amino acid is highly likely to be an amino acid that does not affect the stability or function of the protein.
  • each amino acid in the CDRs of Tables 1 and 2 constituting the heavy chain variable region and light chain variable region of Ang1mab1, which is one of the first basic antibodies actualized in Example [1-1-3] is as follows. As shown in Table 5, preliminary mutant antibodies substituted with alanine were designed. And, by point mutagenesis, the DNA codon encoding the amino acid at each position was converted into the DNA codon encoding the alanine to prepare expression vectors for the heavy and light chains of the preliminary mutated antibodies. The preliminary mutant antibodies prepared as described above were expressed and separated/purified in the same manner as in Example [1-1-3].
  • CDRs of heavy chain variable regions CDRs of light chain variable regions
  • One CDR1-H HC1-G01A One CDR1-L LC1-Q01A 2 CDR1-H HC1-L02A 2 CDR1-L LC1-S02A 3 CDR1-H HC1-T03A 3 CDR1-L LC1-V03A 4 CDR1-H HC1-F04A 4 CDR1-L LC1-L04A 5 CDR1-H HC1-S05A 5 CDR1-L LC1-Y05A 6 CDR1-H HC1-N06A 6 CDR1-L LC1-S06A 7 CDR1-H HC1-Y07A 7 CDR1-L LC1-S07A 8 CDR1-H HC1-M09A 8 CDR1-L LC1-N08A 9 CDR1-H HC1-H10A 9 CDR1-L LC1-N09A 10 C
  • each well was washed twice with 0.1% TBS-Tween20 (TBST), and then treated with goat anti-human IgG-HRP (HRP-conjugated goat anti-human IgG) and reacted for 1 hour. Again, each well was washed 3 times with TBST, and then treated with 50ul each of o-phenylenediamine, a substrate, and reacted for 5 minutes. And after the reaction was terminated by treating each well with 50ul of a reaction stop solution (2.5MH 2 SO 4 ), the absorbance was measured at 450 nm using an ELISA reader to compare the antigen binding power of the preliminary mutant antibodies.
  • a reaction stop solution 2.5MH 2 SO 4
  • Example [1-2] Based on the results of Example [1-2] above, a mutant antibody in which the 1st, 2nd, 5th, and 12th amino acids of the CDR1 of the heavy chain variable region of the basic antibody Ang1mab4 are substituted with alanine, and the CDR1 of the light chain variable region Mutant antibodies in which the 1st, 3rd and 7th amino acids were substituted with alanine were produced and separated/purified in the same manner as in Example [1-1-3], and additionally, CDR1 of the light chain variable region of the primary antibody Ang1mab4 A mutant antibody in which the 5th amino acid was substituted with phenylalanine was additionally designed and produced and separated/purified in the same manner as in Example [1-1-3], a total of 8 mutant antibodies having CDRs as shown in Table 6 below was prepared.
  • control antibody In the first basic antibodies Ang1mab1 and Ang1mab4, the heavy chain variable region CDR3-H was designed by changing the amino acid sequence of SEQ ID NO: 53, and the above Example [1-1- 3], two control antibodies (IgG1 control and IgG4 control) that do not bind to human Tie2 protein were prepared, respectively, by production and separation/purification in the same manner as in].
  • Example [1-1] For the three first and second primary antibodies (Ang1mab1, Ang1mab4 and Ang1mab2-1) prepared in Example [1-1] and the 8 mutant antibodies prepared in Example [1-3], antigens Binding ability to human Tie2 protein was confirmed.
  • the affinity of the antibodies was measured with an Octet K2 (ForteBio) device based on the BLI (Biolayer Interferometry) technique.
  • the AR2G biosensor tip was hydrated with tertiary distilled water for 10 minutes and then 20 mM EDC, 10 mM Sulfo-NHS It was activated using a buffer containing Then, 45 nM of human Tie2-Fc protein (antigen) was diluted in immobilization buffer (10 mM sodium acetate; pH 6.0) and reacted with the activated AR2G tip for 10 minutes to immobilize the antigen to the AR2G tip. .
  • the antigen immobilization reaction was terminated by treatment with a 1M ethanolamine (ethanolamine, pH 8.5) solution, and the AR2G tip was reacted in PBS buffer for 60 seconds to obtain a baseline of antigen-antibody binding.
  • the first and second primary antibodies or the eight variant antibodies were diluted in PBS to concentrations of 30, 15, 7.5, 3.75 and 1.63 nM, and the antigen was bound to the immobilized AR2G tip for 1,000 seconds. After (association), it was dissociated for 1,800 seconds. Based on the reaction curve confirmed through the association and dissociation reaction analysis, the affinity of each antibody was measured by calculating K on , K off , and K D values through global fitting analysis of the Data Analysis HT 12.0 program.
  • both the first and second primary antibodies and the variant antibodies had a K D value of 1.4 nM or less, mostly at a level of several hundreds of pM, indicating excellent affinity for the antigen human Tie2 protein. .
  • HEK293 cells (1x10 6 cells) expressing human Tie2 protein were cultured in a 60 mm culture dish at a temperature of 37 ° C. using DMEM medium.
  • the cells cultured as described above were maintained in serum-free DMEM medium for 6 hours to make them serum starved, and then the first and second primary antibodies (Ang1mab1, Ang1mab4 and Ang1mab2-1) and mutant antibodies (10ug/ml) ) and the positive control Ang1 (300 ng/ml) were treated hourly or for 30 minutes, and then cells were lysed with a lysis buffer to obtain a cell lysate.
  • peroxidase-conjugated secondary antibody was treated and reacted at room temperature for 1 hour, and then treated with ECL solution to reduce human Tie2 protein and lower signal transduction factors (AKT, ERK) on the signal transduction pathway involving the Tie2 protein. It was confirmed whether phosphorylation was induced.
  • the first and second primary antibodies (Ang1mab1, Ang1mab4 and Ang1mab2-1) and variant antibodies induce phosphorylation of Tie2, AKT, and ERK proteins.
  • all of the first and second primary antibodies (Ang1mab1, Ang1mab4 and Ang1mab2-1) exhibited an effect equal to or greater than that of the positive control, Ang1 (FIG. 2).
  • the mutant antibodies also induce phosphorylation of human Tie2 protein similarly to the first primary antibodies (Ang1mab1 and Ang1mab4) ( FIG. 3 ).
  • the first and second primary antibodies (Ang1mab1, Ang1mab4 and Ang1mab2) -1) and phosphorylation (P-Tie2, P-AKT, P-ERK) was checked.
  • Ang1mab4 one of the first primary antibodies obtained in Example [1-1], specifically phosphorylates only the Tie2 protein receptor in HEK293 cells expressing HUVEC and human Tie2 protein, or other protein receptors other than the Tie2 protein It was also confirmed that phosphorylation was carried out.
  • HUVECs (3x10 6 cells) and HEK293 cells (1x10 7 cells) were cultured in a 100 mm culture dish at a temperature of 37° C. using M199 and DMEM medium, respectively.
  • HUVEC and HEK293 cells cultured as described above were maintained in M199 and DMEM medium containing 1% serum for 6 hours, respectively, to make them into a serum starvation state, treated with Ang1mab4 hourly (10 minutes or 30 minutes), and then lysed Cells were lysed with a buffer solution to obtain a cell lysate.
  • the cell lysate obtained as described above was quantified by the BCA protein quantification method, and then reacted with a membrane containing specific phosphorylated antibodies to 49 human protein receptors. Then, a protein receptor induced by phosphorylation by Ang1mab4 was identified through western blotting.
  • first and second primary antibodies Ang1mab1, Ang1mab4 and Ang1mab2-1
  • mutant antibodies that bind to human Tie2 protein can cross-link to Tie2 protein from other species (cross-species binding) Confirmed.
  • Tie2 genes of mice, rats, dogs, pigs, and monkeys were synthesized and cloned into pcDNA3.4 vector to prepare Tie2 expression vectors for each species. 10 ug each of the Tie2 expression vector and 20 ul of Lipofectamine 2000 (invitrogen) were diluted in 1 ml of Opti-MEM medium, treated with HEK293T cells (2.0x10 6 cells), and cultured for 2 days.
  • the culture solution was centrifuged (300xg, 5 minutes) to separate and remove the supernatant, and the cells were washed with FACS buffer.
  • the first and second primary antibodies (Ang1mab1, Ang1mab4 and Ang1mab2-1) and mutant antibodies were treated with the Tie2 expression cells obtained as described above at a concentration of 5ug/ml, and after reaction at room temperature for 1 hour, FACS buffer was used twice. washed. Then, 100 ⁇ l of goat anti-human IgG-FITC (FITC-conjugated goat anti-human IgG; invitrogen) diluted 1:200 was added, reacted at room temperature for 30 minutes, and washed twice again with FACS buffer. Antibody binding was confirmed using a FACS calibur device (BD Bioscience).
  • the first and second primary antibodies (Ang1mab1, Ang1mab4 and Ang1mab2-1) and mutant antibodies were Tie2 protein derived from human ( FIG. 6 ) and monkey ( FIG. 7 ). It was confirmed that, although it did not bind to Tie2 protein derived from another species (FIG. 8).
  • the primary antibody binds, that is, the antigenic determinant of the antibody was analyzed.
  • mutants (Tie2 ⁇ Ig1; Tie2 ⁇ Ig1-2; Tie2 ⁇ Ig1-2,EGF; Tie2 ⁇ ) in which the full-length or extracellula domain of the human Tie2 protein has been partially removed as shown in FIG. Binding of the primary antibody to Ig1-2, EGF, Ig3) was confirmed. Specifically, the portion encoding the Ig1, Ig2, EGF or IgG3 domain was removed from the nucleotide sequence encoding the human Tie2 protein by recombinant-PCR, and then cloned into pcDNA3 vector.
  • the cells expressing the entire length or part of the human Tie2 protein obtained as described above were treated with the primary antibody Ang1mab1 at a concentration of 10 ug/ml, reacted at room temperature for 1 hour, and then washed twice with FACS buffer. Then, 100 ⁇ l of goat anti-human IgG-FITC (FITC-conjugated goat anti-human IgG; invitrogen) diluted 1:200 was added, reacted at room temperature for 30 minutes, and washed twice again with FACS buffer. Antibody binding was confirmed using Image FACS, FlowSight (Amnis).
  • the basic and mutant antibodies bind to the Ig2 domain of human Tie2 protein, and thus the Ig2 domain of human Tie2 protein is an antigenic determinant of the basic and mutant antibodies.
  • Example [6-1] In order to confirm the amino acid sequence of the antigenic determinant of the basic antibody more specifically among the domains identified in Example [6-1], a peptide array including the peptide sequence of the human Tie2 protein extracellular region was prepared and antigenic determinant mapping (epitope mapping) analysis was performed.
  • the basic antibody binds to the Ig2 domain of the human Tie2 protein in Example [6-1]
  • the 113th amino acid to the 342th amino acid in the amino acid sequence of the human Tie2 protein including the domain A number of peptides were synthesized so that 12 amino acids overlapped based on a length of 15 amino acids for the designated site as described above by requesting JPT Peptide Technologies. And the peptides synthesized as described above were fixed on a nitrocellulose membrane and an ELISA plate and used for antigenic determinant analysis.
  • the primary antibody Ang1mab1 at a concentration of 30ug/ml was treated and bound to the nitrocellulose membrane for 16 hours, and then HRP (horseradish peroxidase)-labeled anti-human secondary The antibody was treated and reacted for 2 hours. Then, the membrane was washed 3 times for 10 minutes each using PBST (Phosphate Buffered Saline with Tween), treated with Amersham TM ECL (GE Healthcare) for color development, reacted for 10 minutes or 20 minutes, and then reacted with light. area was confirmed.
  • PBST Phosphate Buffered Saline with Tween
  • the binding site of the primary antibody Ang1mab1 was reassayed using the ELISA plate coated with the peptides synthesized as described above.
  • the primary antibody Ang1mab1 at a concentration of 10ug/ml was treated on an ELISA plate for 1 hour, and then treated with an HRP-labeled anti-human secondary antibody and reacted for 1 hour. Then, the plate was washed 3 times for 10 minutes using PBST, and TMB (3,3',5,5'-Tetramethylbenzidine) substrate (BD Biosciences) was treated for 20 minutes for enzymatic reaction. Then, the enzyme reaction was terminated by treatment with 2N sulfuric acid, and the absorbance at OD450 was measured.
  • FIG. 10B (B) binding of the primary antibody Ang1mab1 was confirmed at peptide sites 1, 2, 7, 8, 9, 10, and 28 (FIG. 10B (B)).
  • the basic and mutant antibodies are the amino acid sequence of SEQ ID NO: 3, in particular, the amino acid sequence of SEQ ID NO: 54, present in the N-terminal region of the Ig2 domain of human Tie2 protein, or It can be seen that binding to the amino acid sequence of SEQ ID NO: 55 promotes the activity of human Tie2 protein.
  • HUVECs (1x10 5 cells), which became serum starved by maintaining for 6 hours in M199 medium containing 1% serum, were dispensed in a GFR-Matrigel-coated 24-well culture dish, and the HEVECs in the above example The degree of blood vessel formation was checked while incubating for 18 hours by treating the basic and mutant antibodies (20ug/ml) and control antibody (IgG4 control; 20ug/ml) prepared in 1 above.
  • both of the first and second primary antibodies (Ang1mab1, Ang1mab4 and Ang1mab2-1) and mutant antibodies increase blood vessel formation and maintain for a long time compared to the control antibody. was confirmed (FIG. 11).
  • HUVECs 3x10 6 cells
  • M199 medium at a temperature of 37 ° C. for 24 hours.
  • HUVECs were treated with the primary antibody Ang1mab4 (20ug/ml) and the positive control, Ang1 (300ng/ml), and cultured for 36 hours. Lysed to obtain a cell lysate.
  • the cell lysate obtained as described above was quantified by BCA protein quantitation, followed by SDS-PAGE, and transferred to a PVDF membrane and reacted with 0.1% Tween20 TBST buffer diluted with 3% BSA at room temperature for 2 hours. Then, the primary antibodies (caspase-3, caspase-8 or actin) diluted at 1:1000 were treated and reacted at room temperature for 2 hours, and peroxidase-conjugated (peroxidase-conjugated) diluted at 1:5000 was treated. ) After the secondary antibody was treated and reacted at room temperature for 1 hour, the expression of the caspase-3, caspase-8 and actin proteins was checked by treating the ECL solution.
  • FIG. 12 it was confirmed that the primary antibody suppressed the cell death of HUVECs induced in the starvation state at a level similar to that of the positive control Ang1 ( FIG. 12 (A) ). And, this inhibitory effect on cell death could be verified by confirming that the activities of cleaved Cas-3 and Cas-8, which are proteins inducing cell death, were inhibited (FIG. 12 (B)).
  • a linear nucleotide sequence consisting of a mouse Tie2 promoter gene, a human Tie2 gene, a poly A gene, and a mouse Tie2 enhancer gene in order was prepared.
  • Superovulation was induced by injecting pregnant mare serum gonadotropin and human chorionic gonadotropin into C57BL/6N female mice.
  • superovulation-induced female C57BL/6N mice were crossed with C57BL/6N male mice, and fertilized eggs were obtained from C57BL/6N female mice whose pregnancy was confirmed.
  • a linear nucleotide sequence consisting of a mouse Tie2 promoter gene, a human Tie2 gene, a poly A gene, and a mouse Tie2 enhancer gene was prepared in this order. injected.
  • a mouse having the human Tie2 gene (SEQ ID NO: 1) was obtained through genotyping. Then, in order to check whether the human Tie2 protein is normally expressed in the blood vessels of the mouse, the brain tissue was separated from the mouse obtained as above, fixed with formalin (Sigma) for 1 day, and then 30% sucrose was added. After processing the PBS for 3 days, it was made into a frozen block (frozen block).
  • a tissue slide was prepared by cryosectioning the frozen block, and an antibody of CD31 (Millipore, 1:200), which is a blood vessel-specific marker, and a Tie2 antibody that specifically binds to human Tie2 protein (Biolegend 1:200)
  • CD31 a blood vessel-specific marker
  • Tie2 antibody that specifically binds to human Tie2 protein
  • Biolegend 1:200 The mouse brain blood vessels were immunostained using Each antibody was treated at 4°C for 16 hours, and then further treated with a secondary antibody to which Cy3 or Alexa Fluor 488 blood vessels were attached at room temperature for 2 hours, and then the tissue slides were mounted using a Mounting Medium With DAPI (Vector). After immunostaining, it was observed through a fluorescence microscope.
  • Example [7-3] Using the transgenic mouse prepared in Example [7-3], the primary antibodies (Ang1mab1 and Ang1mab4) prepared in Example [1-1] induce vascular endothelial cell sprouting in arterial tissue. It was analyzed in ex vivo whether or not it exhibits the efficacy.
  • the aortic blood vessel was separated from the mouse prepared in Example [7-3], cut to a predetermined size (1 mm), and seeded on a matrigel-coated plate. Then, the primary antibodies Ang1mab1 (50 ⁇ g/ml) and Ang1mab4 (10, 50 ⁇ g/ml) and control antibodies (IgG4 control; 50 ⁇ g/ml), and positive controls VEGF (100 ng/ml) and Ang1 (200 ng/ml) ) was added and cultured for 6 days at an interval of 2 days, respectively, and the degree of germination of vascular endothelial cells from the arterial tissue was confirmed.
  • the primary antibodies Ang1mab1 (50 ⁇ g/ml) and Ang1mab4 (10, 50 ⁇ g/ml) and control antibodies (IgG4 control; 50 ⁇ g/ml)
  • IgG4 control 50 ⁇ g/ml
  • VEGF 100 ng/ml
  • both of the primary antibodies (Ang1mab1, Ang1mab4) were confirmed to significantly increase the germination of arteriovascular endothelial cells compared to the control antibody, and also compared to the positive control (VEGF, Ang1). It was confirmed to significantly increase the germination of arteriovascular endothelial cells (FIG. 14).
  • Example [1-1] In vivo analysis of how much the primary antibody prepared in Example [1-1] can promote wound healing through induction of angiogenesis using the transgenic mouse prepared in Example [7-3] did
  • Example [7-3] After subcutaneous hair removal of the transgenic mouse prepared in Example [7-3], a circular wound with a diameter of 4 mm was induced, and Ang1mab4, a basic antibody, and a control antibody (IgG4 control) were added to the wound site at 7.5 mg. After intraperitoneal injection 3 times at 2 day intervals at a dose of /kg, the degree of wound healing was comparatively analyzed for 6 days.
  • tissue slide was prepared and immunostained in the same manner as in Example [7-3] after separating the recovered wound tissue from the mouse.
  • an antibody to CD31, a blood vessel-specific marker (Millipore, 1:200) and an antibody to ⁇ SMC, a marker for perivascular cells that maintain blood vessel stabilization (Abcam, 1:100) were added at 4°C for 16 hours.
  • FIG. 16 it was confirmed that angiogenesis increased in the Ang1mab4-treated tissue, and that the blood vessel was well surrounded by cells around the blood vessel ( FIG. 16 ).
  • Example [1-1] In vivo analysis of how much the primary antibody prepared in Example [1-1] can improve lower extremity ischemia by promoting angiogenesis using the transgenic mouse prepared in Example [7-3] did
  • the transgenic mice prepared in Example [7-3] were anesthetized using a respiratory anesthesia system, and then the mouse legs were fixed with tape. After removing the hair on the surgical site with a hair removal cream, disinfect it with an alcohol swab, make an incision in the skin about 1 cm from the knee to the middle of the thigh using forceps and scissors, secure the view of the surgical site with a retractor, and then wash with PBS under a dissecting microscope. The subcutaneous fat around the femoral muscle was removed with a thin cotton swab, microscopic scissors, and fine scissors.
  • the femoral artery and vein in the groin area were separated from the nerve, and two 7-0 silks were passed through the lower part of the proximal end of the femoral artery/vein, and the femoral artery/vein was tied and occluded at regular intervals. Also, two 7-0 silks were passed under the distal part of the femoral artery/vein, tied at a certain distance and occluded. The tractor was removed and the mice were recovered on a heated mat by sutures with 5-0 vicryl sutures.
  • the primary antibody, Ang1mab4 was injected into the tissue surrounding the lower extremity ischemia of a mouse in which lower extremity ischemia was induced by cutting the arteries and veins in the lower extremity and blocking the flow of blood. Then, in order to compare blood vessel formation and blood flow, the blood flow in the lower extremities of the mouse was measured using a laser Doppler imaging system. Laser Doppler images were measured for mice before surgery, on days 1, 3, and 7 after surgery. After lower extremity ischemia surgery, the mice were divided into two groups of 7 mice each, and a dose of 10 mg/kg was administered on the 1st and 3rd days after the lower extremity ischemia surgery.
  • the primary antibody, Ang1mab4, and the control antibody (IgG4 control) were divided and injected into the tissues surrounding the surgery of each group of mice. And by taking Doppler images and comparing the blood flow of the non-operated leg and the operated leg, the effect of improving lower extremity ischemia was confirmed.
  • FIG. 17 in the group treated with the Ang1mab4, it was confirmed that angiogenesis in the lower extremity tissue was increased compared to the group treated with the control antibody (FIG. 17(A)), blood flow It was confirmed that the flow was also significantly improved (FIG. 17 (B)).
  • HUVECs (1X10 5 cells) were seeded in the upper chamber of a 12-well Transwell plate (0.4 ⁇ m pore) and cultured for 3 days, and then maintained in a serum starvation state for 6 hours. Then, in order to artificially increase vascular permeability, 50 ng/ml of VEGF was treated in all of the negative control, positive control, and experimental groups, while 100 ng/ml of Ang1 was administered to the positive control group, and Ang1mab4, the primary antibody, was administered to the experimental group by 10 each. , 25 and 50 ⁇ g/ml were treated for 5 hours.
  • fluorescence-labeled dextran (FITC-Dextran, 70 kDa) was added to the culture medium, and 30 minutes later, dextran transmitted through HUVECs was compared and analyzed with a fluorescence analyzer (SpectraMAx i3x).
  • HUVECs (1X10 5 cells) were seeded in 35 mm confocal dish (SPL) and cultured for 2 days, and then maintained in a serum starvation state for 6 hours. Then, while the negative control was treated with 50ng/ml of VEGF, the positive control was treated with 100ng/ml of Ang1, and the experimental group was treated with the primary antibody Ang1mab4 at a concentration of 25 ⁇ g/ml for 5 hours. Then, it was fixed with 4% paraformaldehyde for 10 minutes, and then permeated through the cell membrane for 10 minutes using PBS containing 0.2% Triton X-100, and then with PBS containing 1% BSA for 1 hour. Blocked at room temperature.
  • FIG. 18(B) As a result of confirming the change in the position of VE-cadherin involved in junction formation between HUVECs through cytoimmunochemical staining analysis, as shown in FIG. 18(B) , in the negative control treated with VEGF alone, cells Although junctions are formed remarkably weakly, in the cells of the test group treated with the primary antibody Ang1mab4, cell junctions between cells are well formed, and the interface between cells is strongly developed, equal to or greater than in the cells of the positive control group treated with Ang1. was confirmed (FIG. 18 (B)).
  • Example [7-3] Using the transgenic mouse prepared in Example [7-3], the primary antibody (Ang1mab4) prepared in Example [1-1] inhibits vascular leakage and enhances vascular endothelial integrity, resulting in sepsis was analyzed in vivo to see how much it can be inhibited.
  • Ang1mab4 the primary antibody prepared in Example [1-1] inhibits vascular leakage and enhances vascular endothelial integrity, resulting in sepsis was analyzed in vivo to see how much it can be inhibited.
  • the transgenic mouse prepared in Example [7-3] was anesthetized using a respiratory anesthesia system, the abdominal hairs were removed and wiped with a sterile alcohol swab, and the integument was applied to the central part of the abdomen in a size of 1 cm. incised. Then, the endothelium was further incised and the cecum of the mouse was removed, and the 75% position of the cecum was tied with 4-0 black silk suture.
  • a sepsis model was prepared by cecal ligation and puncture (CLP) by putting the incision in the ventilator and suturing the incision and then recovering the mouse on a heating mat.
  • CLP cecal ligation and puncture
  • Example [7-3] Using the transgenic mouse prepared in Example [7-3], it was examined how effectively the primary antibody prepared in Example [1-1] can inhibit tumor growth caused by abnormal angiogenesis. Analyzed in vivo.
  • GL261 cells (5x10 3 cells/ ⁇ L), which are mouse glioblastomas, were injected subcutaneously into the transgenic mice prepared in Example [7-3]. Changes in tumor size were measured at intervals of 2 days after tumor injection, using vernier calipers for the long axis and short axis of the tumor. By calculating the size of the measured tumor as (long axis x short axis x short axis)/2, when the size grew to an average of 30 mm 3 , a control antibody (IgG1 control; 7.5 mg/kg) was used in the control group, and the primary antibody Ang1mab1 in the experimental group. (7.5 mg/kg) was administered to the tail vein 5 times at 3-day intervals. On the 23rd day of tumor injection, all subjects were killed using CO 2 gas, and then laparotomy was performed to visually check for organ abnormalities and measure the weight of the tumor.
  • IgG1 control 7.5 mg/kg
  • FIG. 20 it was confirmed that the tumor size in mice treated with the primary antibody, Ang1mab1, was inhibited by about 50% or more compared to the control group (IgG1 control) (FIG. 20 (A)), It was confirmed that the weight of the tumor was also reduced by 70% or more (FIG. 20 (B)).
  • Example [7-3] Furthermore, in order to confirm whether the tumor growth inhibitory effect as described above was induced through normalization of abnormal tumor blood vessels, after separating the tumor tissue from the mouse, a tissue slide was performed in the same manner as in Example [7-3]. was prepared and immunostained. In the tumor tissue, an antibody of CD31, a blood vessel-specific marker (Millipore, 1:200), and an antibody of ⁇ SMC, a marker of perivascular cells that maintain blood vessel stabilization (Abcam, 1:100) were treated at 4°C for 16 hours.
  • an antibody of CD31 a blood vessel-specific marker (Millipore, 1:200)
  • ⁇ SMC a marker of perivascular cells that maintain blood vessel stabilization
  • FIG. 21 it was confirmed that the blood vessels in the tumor tissue treated with the primary antibody Ang1mab1 were well surrounded by perivascular cells and stabilized ( FIG. 21 ).
  • Example [8-3] suppressed tumor growth by increasing the influx of anti-tumor immune cells into the tumor due to the normalization and stabilization-inducing effect of abnormal tumor blood vessels.
  • CountBright Absolute count beads Invitrogen
  • NK cells FIG. 22(A)
  • CD8 CD4 T cells
  • macrophages FIG. 22(C)
  • Example [7-3] When using the transgenic mouse prepared in Example [7-3], the primary antibody prepared in Example [1-1] and the immune checkpoint inhibitory antibody (Anti-PD-1), which is one of the other anticancer agents, were concurrently administered , it was confirmed whether the tumor growth could be more effectively inhibited than when each was treated alone.
  • Anti-PD-1 immune checkpoint inhibitory antibody
  • LLC cells (5x10 3 cells/ ⁇ l), which are mouse lung tumor cells, were injected subcutaneously into the transgenic mice prepared in Example [7-3]. Changes in tumor size were measured at intervals of 2 days after tumor injection, using vernier calipers for the long axis and short axis of the tumor.
  • the control antibody IgG1 control; 7.5 mg / kg
  • the primary antibody Ang1mab1 7.5 mg / kg
  • Anti-PD-1 antibody 20mg/kg
  • Each antibody was administered to the tail vein a total of 5 times at 3-day intervals, and on the 23rd day of tumor injection, all subjects were killed using CO 2 gas, and then laparotomy was performed to visually check for organ abnormalities and the weight of the tumor. was measured.
  • FIG. 23 it was confirmed that the size of the tumor was inhibited by about 85% in the mouse group administered with the primary antibody Ang1mab1 and the Anti-PD1 antibody (FIG. 23 (A)), and the tumor It was confirmed that the weight was also reduced by more than 87% (FIG. 23 (B)).
  • a tetravalent antibody based on the first primary antibody Ang1mab1 was prepared.
  • tetravalent antibodies such as Ang1mab1 HCX2 and Ang1mab1 LCX2 shown in FIG. 24A were designed, and expression vectors for each of the heavy and light chains were constructed. Then, using the vector prepared as described above, the tetravalent antibody was produced and purified in the same manner as in Example 1, followed by SDS-PAGE analysis.
  • Example [9-1] The two types of tetravalent antibodies prepared in Example [9-1] were subjected to antigen affinity analysis in the same manner as in Example 2, and the control antibody (16A2X2) and the primary antibody Ang1mab1 were compared.
  • the affinity of the tetravalent antibody Ang1mab1 LCX2 was 41.3 pM, which was confirmed to be increased by about 7.5 times or more compared to the primary antibody Ang1mab1, which is a bivalent antibody, and the tetravalent antibody
  • the affinity of Ang1mab1 HCX2 was measured to be lower than the minimum measured value of 1 pM, and it was confirmed that the affinity was increased by at least 18 times compared to the primary antibody, Ang1mab1, which is a bivalent antibody (FIG. 25 (A)).
  • these antibodies are human Tie2 protein and downstream signals on the signal transduction pathway in which the Tie2 protein is involved. It was confirmed whether phosphorylation (P-Tie2, P-AKT, P-ERK) of the transfer factors (AKT, ERK) was induced.
  • FIG. 25B it was confirmed that the tetravalent antibodies (Ang1mab1 HCX2 and Ang1mab1 LCX2) induce phosphorylation of Tie2, AKT, and ERK proteins, and furthermore, the first and It was confirmed that the second primary antibodies (Ang1mab1 and Ang1mab4) or the positive control Ang1 phosphorylated the proteins at a much higher level (Fig. 25 (B)).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Ophthalmology & Optometry (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gynecology & Obstetrics (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

La présente invention concerne un nouvel anticorps anti-Tie2 ou un fragment de liaison à l'antigène de celui-ci et son utilisation. La présente invention concerne un anticorps ou un fragment de liaison à l'antigène de celui-ci, qui comprend : au moins une région variable de chaîne lourde comprenant CDR1-H ayant une séquence d'acides aminés de X1-X2-X3-X4-X5-X6-Y-X7-M-H-W-X8 (dans la séquence d'acides aminés, X1 à X8 représentent chacun indépendamment un acide aminé quelconque) ; et au moins une région variable de chaîne légère comprenant CDR1-L ayant une séquence d'acides aminés de X17-X18-X19-X20-X21-X22-X23-N-X24-X25-X26-Y (dans la séquence d'acides aminés, X17 à X20 et X22 à X26 représentent chacun indépendamment un acide aminé quelconque, et X21 est un quelconque choisi dans le groupe constitué par la tyrosine, la phénylalanine et le tryptophane. L'anticorps ou le fragment de liaison à l'antigène de celui-ci, selon la présente invention, se lie de manière spécifique à une protéine Tie2 humaine ou de singe, présentant ainsi l'activité de phosphorylation de celui-ci, tout en présentant un effet bien supérieur en termes de spécificité ou d'affinité d'antigène, par comparaison avec des ligands ou des anticorps existants, et est un anticorps entièrement humain, et peut donc être appliqué en tant que principe actif d'une composition pharmaceutique pour les êtres humains sans risque de rejet. Ainsi, l'anticorps ou le fragment de liaison à l'antigène de celui-ci a l'avantage d'être utilisé de manière efficace dans le traitement ou la prévention de maladies liées à l'angiogenèse.
PCT/KR2021/015786 2020-11-03 2021-11-03 Nouvel anticorps anti-tie2 ou fragment de liaison à l'antigène de celui-ci et son utilisation WO2022098080A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20200144961 2020-11-03
KR10-2020-0144961 2020-11-03
KR10-2021-0050134 2021-04-16
KR1020210050134A KR20220059888A (ko) 2020-11-03 2021-04-16 신규 항-Tie2 항체 또는 이의 항원 결합 단편, 및 이의 용도

Publications (1)

Publication Number Publication Date
WO2022098080A1 true WO2022098080A1 (fr) 2022-05-12

Family

ID=81458093

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/015786 WO2022098080A1 (fr) 2020-11-03 2021-11-03 Nouvel anticorps anti-tie2 ou fragment de liaison à l'antigène de celui-ci et son utilisation

Country Status (2)

Country Link
KR (1) KR20220059924A (fr)
WO (1) WO2022098080A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060057138A1 (en) * 2003-08-12 2006-03-16 Dyax Corporation Tie complex binding proteins
KR20170029508A (ko) * 2014-07-15 2017-03-15 아스텔라스세이야쿠 가부시키가이샤 신규 항인간 Tie2 항체
KR20190139148A (ko) * 2018-06-07 2019-12-17 기초과학연구원 Tie2에 결합하는 항체 및 이의 용도

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060057138A1 (en) * 2003-08-12 2006-03-16 Dyax Corporation Tie complex binding proteins
KR20170029508A (ko) * 2014-07-15 2017-03-15 아스텔라스세이야쿠 가부시키가이샤 신규 항인간 Tie2 항체
KR20190139148A (ko) * 2018-06-07 2019-12-17 기초과학연구원 Tie2에 결합하는 항체 및 이의 용도

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE PROTEIN 11 July 2018 (2018-07-11), ANONYMOUS : "immunoglobulin light chain variable region, partial [Homo sapiens]", XP055928955, retrieved from NCBI Database accession no. AXA12665 *
DATABASE PROTEIN 7 July 2013 (2013-07-07), ANONYMOUS : "immunoglobulin A heavy chain variable region. partial [Homo sapiens]", XP055928949, retrieved from NCBI Database accession no. AGP01910 *

Also Published As

Publication number Publication date
KR20220059924A (ko) 2022-05-10

Similar Documents

Publication Publication Date Title
WO2020076105A1 (fr) Nouvel anticorps anti-c-kit
WO2015005632A1 (fr) Nouvelles protéines à double cible se liant spécifiquement à dll4 et vegf et leur utilisation
WO2016153276A1 (fr) Peptide liant spécifiquement la neuropiline-1, protéine de fusion fusionnée à celui-ci et utilisation correspondante
WO2012165925A2 (fr) Anticorps anti-c-met présentant une activité hgf et son utilisation
WO2019004799A1 (fr) Conjugué de protéine de vegf-grab et de médicament, et son utilisation
WO2021010621A1 (fr) Agents thérapeutiques peptidiques pour maladies auto-immunes et maladies inflammatoires
WO2018088838A1 (fr) Composition pharmaceutique comprenant des protéines de fusion destinée à la prévention ou le traitement de l'hépatite, de la fibrose hépatique et de la cirrhose hépatique
WO2019164219A1 (fr) Anticorps anti-angiopoïétine-2 et leurs utilisations
AU2019224694A1 (en) Anti-angiopoietin-2 antibodies and uses thereof
CA3074437C (fr) Composition pour la prevention et le traitement des maladies de la peau comprenant une matiere se liant specifiquement a un peptide derive de lavimentine
WO2022098080A1 (fr) Nouvel anticorps anti-tie2 ou fragment de liaison à l'antigène de celui-ci et son utilisation
WO2020251316A1 (fr) ANTICORPS BISPÉCIFIQUE DIRIGÉ CONTRE α-SYN/IGF1R ET UTILISATION ASSOCIÉE
WO2020004937A1 (fr) Conjugué anticorps anti-bcma-médicament et utilisation associée
WO2023022271A1 (fr) Anticorps anti-igsf1 et son utilisation
WO2020184941A1 (fr) Protéines de fusion glp-1 et leurs utilisations
WO2014021693A2 (fr) Anticorps monoclonal inédit se liant spécifiquement à la protéine tm4sf5 et son utilisation
WO2020071881A1 (fr) Anticorps de récepteur pdgf et son utilisation
WO2018124851A1 (fr) Composition pharmaceutique pour la prévention ou le traitement du cancer, contenant un anticorps se liant spécifiquement à une protéine l1cam et un analogue de la pyrimidine et/ou un médicament anticancéreux à base de platine
WO2023229303A1 (fr) Anticorps se liant à l'extrémité c-terminale de igsf1 et son utilisation
WO2021029746A2 (fr) Anticorps anti-tie 2 et son utilisation
WO2023249425A1 (fr) Protéine de fusion comprenant un anticorps anti-cd73 et il-2, et son utilisation
WO2024054030A1 (fr) Anticorps anti-ptk7 et son utilisation
WO2019088658A1 (fr) Anticorps à double ciblage ciblant le scf et la galectine-1 et son utilisation
WO2022169269A1 (fr) Anticorps anti-ctla-4 et son utilisation
WO2023132547A1 (fr) Anticorps anti-c3b ou anticorps anti-c5 conjugué à un inhibiteur d'angiogenèse et son utilisation

Legal Events

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

Ref document number: 21889545

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21889545

Country of ref document: EP

Kind code of ref document: A1