WO2019235856A1 - Antibody binding to tie2 and use thereof - Google Patents

Antibody binding to tie2 and use thereof Download PDF

Info

Publication number
WO2019235856A1
WO2019235856A1 PCT/KR2019/006820 KR2019006820W WO2019235856A1 WO 2019235856 A1 WO2019235856 A1 WO 2019235856A1 KR 2019006820 W KR2019006820 W KR 2019006820W WO 2019235856 A1 WO2019235856 A1 WO 2019235856A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
seq
amino acid
antibody
variable region
Prior art date
Application number
PCT/KR2019/006820
Other languages
French (fr)
Korean (ko)
Inventor
고규영
배점일
김재령
Original Assignee
기초과학연구원
한국과학기술원
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 기초과학연구원, 한국과학기술원 filed Critical 기초과학연구원
Priority to EP19814472.7A priority Critical patent/EP3805265A4/en
Priority to CA3101506A priority patent/CA3101506A1/en
Priority to JP2020567832A priority patent/JP7390317B2/en
Priority to AU2019283520A priority patent/AU2019283520A1/en
Priority to CN201980038199.8A priority patent/CN112399975A/en
Priority claimed from KR1020190066622A external-priority patent/KR20190139148A/en
Publication of WO2019235856A1 publication Critical patent/WO2019235856A1/en
Priority to US17/111,413 priority patent/US20210179719A1/en
Priority to JP2023150164A priority patent/JP2023175830A/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants

Definitions

  • the present invention provides an antibody against Tie-2 or an antigen-binding fragment thereof, a nucleic acid encoding the same, a vector comprising the nucleic acid, a cell transformed with the vector, a method for producing the antibody or an antigen-binding fragment thereof, and a blood vessel comprising the same. It relates to a composition for preventing or treating neoplastic disease.
  • Angiogenesis occurs dynamically by a variety of regulatory factors during the development, growth, preservation and maintenance of homeostasis.
  • newly formed blood vessels serve as transport channels for various biomaterials such as nutrients, oxygen and hormones to surrounding cells.
  • Functional and structurally abnormal blood vessels are a direct and indirect cause of the onset and progression of various diseases.
  • Tumor blood vessels exacerbate hypoxia with functional and structural defects, leading to tumor progression and metastasis to other tissues, and to poor delivery of anticancer agents to the center of the tumor tissue.
  • Defective blood vessels can be identified in various diseases or disorders, in addition to cancer. Examples include acute inflammatory reactions such as various eye diseases (eg diabetic macular edema, senile macular degeneration), viral infections and sepsis.
  • Angiopoietin family plays an important role in the formation and maintenance of blood vessels and includes four Angiopoietins (Ang1, Ang2, Ang3 and Ang4).
  • Angiopoietin-1 binds to Tie2 receptors present on the surface of vascular endothelial cells to phosphorylate and activate Tie2 receptors, thereby stabilizing blood vessels.
  • Angiopoietin-2 binds to the Tie2 receptor but acts as an antagonist that induces inactivation of the Tie2 receptor, leading to blood vessel destabilization and vascular leakage.
  • Ang2 is known to act as an agent that induces activation of the Tie2 receptor in many processes, including lymphatic vessel formation and maintenance. Therefore, Ang2 seems to perform various functions depending on the environment and organization.
  • anti-Ang2 antibodies have been developed and clinical trials have been conducted by many biopharmaceuticals (eg, US Pat. No. 7,658,924 and US Pat. No. 8,987,420). These Ang2 antibodies have been shown to inhibit the binding of Ang2 to Tie2, and this Ang2 neutralizing effect ultimately appears to interfere with the formation of angiogenesis.
  • Angiogenesis inhibition and anticancer activity of anti-Ang2 antibodies has been demonstrated in many preclinical models, and several types of anti-Ang2 antibodies have been clinically tested in various cancer patients. However, it has been demonstrated that inhibition of angiogenesis through Ang2 neutralization is not sufficient for anticancer treatment.
  • direct activation of Tie2 is considered a new approach to inhibit abnormal angiogenesis and reduce vascular permeability.
  • Recombinant proteins that directly bind to the Tie2 receptor to induce Tie2 phosphorylation and activation have also been developed, and their therapeutic effects have been tested in many preclinical cancer and ocular models.
  • COMP-Angl and Vasculotide are examples. These agents exhibited anti-angiogenic and anti-invasive activity, but have the disadvantage of having very short half-lives and unstable physicochemical properties.
  • small molecule compounds (AKB-9778) have been developed as inhibitors of dephosphatase VE-PTP.
  • VE-PTP serves to inactivate Tie2 by removing phosphate groups from phosphorylated Tie2. These compounds have the disadvantage of nonspecifically activating other receptors, but they also indirectly increase Tie2 activity by inhibiting VE-PTP.
  • Tie2 activating antibodies have been developed (US6365154B1, US20170174789A1). These antibodies inhibited vascular leakage by increasing the survival rate of vascular endothelial cells.
  • one of the plant extracts induces Tie2 activity and can be used as a skin care cosmetic (eg JP2011102273A, JP2018043949A, JP2015168656A).
  • the inventors of the present application sought to develop an antibody that specifically binds Tie2.
  • the present inventors have developed a Tie2 antibody that binds Tie2 with high affinity, and confirms that the Tie2 antibody can play a role as a therapeutic agent for angiogenic diseases by inducing Tie2 phosphorylation and activation of the Tie2 receptor.
  • the present invention has been completed.
  • Another object of the present invention is to provide a nucleic acid encoding the antibody or antigen-binding fragment thereof.
  • Another object of the present invention is to provide a vector comprising the nucleic acid, a cell transformed with the vector, and a method of manufacturing the same.
  • Still another object of the present invention is to provide a composition for preventing or treating angiogenic diseases comprising the antibody or antigen-binding fragment thereof.
  • Still another object of the present invention is to provide a composition for co-administration with another therapeutic agent for angiogenic diseases, including the antibody or antigen-binding fragment thereof.
  • the present invention provides an anti-Tie2 antibody or antigen binding fragment thereof that binds to a Tie2 Ig3-FNIII (1-3) domain comprising the sequence of SEQ ID NO: 2.
  • the present invention also provides a heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NO: 3 to 5, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NO: 6 to 8; A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 13 to 15, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 16 to 18; A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 23 to 25, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 26 to 28; A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 33 to 35, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 36 to 38; Or an antibody binding to Tie
  • the present invention also provides a nucleic acid encoding the antibody or antigen-binding fragment thereof.
  • the present invention also provides a vector comprising the nucleic acid.
  • the present invention also provides a cell transformed with the vector.
  • the present invention also provides a method for producing the antibody or antigen-binding fragment thereof comprising the following steps: (a) culturing the cells; And (b) recovering the antibody or antigen-binding fragment thereof from the cultured cells.
  • the present invention also provides a composition for preventing or treating angiogenesis-related diseases comprising the antibody or antigen-binding fragment thereof as an active ingredient.
  • the present invention also provides a composition for co-administration with another therapeutic agent for angiogenic diseases, comprising the antibody or antigen-binding fragment thereof.
  • Figure 1 shows the analysis results for Akt phosphorylation induced by anti-Tie2 antibody.
  • HUVECs remained serum-starved for 6 hours and incubated with COMP-Ang1 (CA1, 0.5 ⁇ g / ml) or anti-Tie2 antibodies (11C4, 4A4, 3B2, 3E12 and 3H7) for 30 minutes.
  • Lysates of the cultured cells were subjected to SDS-PAGE / Western blotting and probed with p-Akt antibody (S473) or Akt antibody.
  • FIG. 2 shows the assay results for dose-dependent Tie2 phosphorylation (pTie2) induced by anti-Tie2 antibody 3H7.
  • the Tie2 phosphorylation capacity of 3H7 antibody was analyzed through immunoprecipitation and western blotting analysis. Serum starved HUVECs were incubated with various concentrations of 3H7 for 30 minutes. As a control, HUVECs were incubated at the same time in Ang2 / Control antibody (Control Ab) mixture. After immunoprecipitation of the Tie2 protein in the cell lysate using the Tie2 antibody, the degree of phosphorylation of Tie2 was measured by SDS-PAGE / Western blot analysis. Tie2 phosphorylation was probed by 4G10, a tyrosine phosphate (pY) antibody derived from mice.
  • pY tyrosine phosphate
  • FIG. 3 shows the results of translocation of FOXO1 and endocytosis of Tie2 receptor due to anti-Tie2 antibody 3H7.
  • HUVECs were placed in serum starvation for 6 hours and then incubated with negative control (Control), Ang2 / Control antibody mixture (A2 + Control Ab) or 3H7 for 30 minutes. After cell fixation, the cell nuclei were blue with DAPI staining, the Tie2 receptor was green with an anti-Tie2 antibody, and FOXO1 was stained red with an anti-FOXO1 antibody to examine the location of FOXO1 and clustered Tie2 receptors. Arrows in the figure labeled Tie2 receptors introduced into the cell.
  • FIG 4 is a result showing the inhibition of vascular permeability induced by VEGF or TNF-a through the Tie2 antibody 3H7.
  • HUVECs were inoculated into a trans well chamber and incubated for 3 days. At 100% confluence, HUVECs were pretreated with Ang2 (A2, 1 ⁇ g / ml), Ang2 / control antibody mixture (A2 + Control Ab, 1 ⁇ g / ml) or 3H7 (1 ⁇ g / ml) for 30 minutes each. Treatment with VEGF (500 ng / ml) for 45 minutes in the upper chamber (A) or TNF-a (100 ng / ml) for 22 hours (B).
  • Ang2 A2, 1 ⁇ g / ml
  • A2 + Control Ab 1 ⁇ g / ml
  • 3H7 1 ⁇ g / ml
  • the upper chamber was then treated with FITC-dextran for 20 minutes and the vascular permeability was analyzed by measuring the amount of FITC fluorescence transmitted through the lower chamber.
  • the measured value is mean ⁇ standard deviation. According to the one way ANOVA, * is p ⁇ 0.05, ** is p ⁇ 0.01, and *** is p ⁇ 0.001.
  • FIG. 5 shows a heat map showing a significant difference region in deuterium uptake as a result of analyzing Tie2 alone or Tie2 / 3H7 complex through hydrogen / deuterium (H / D) exchange-mass spectrometry.
  • H / D exchange-mass spectrometry data in the presence or absence of anti-Tie2 antibody 3H7 was used as heat maps for deuterium uptake per residue for hTie2 antigen at 0.333, 10, 60 and 240 minutes.
  • the color scale represents the percentage of H / D exchange per residue between Tie2 alone and Tie2 / 3H7 mixture at each time. Red color indicates increased deuterium exchange and blue color was not absorbed.
  • FIG. 6 is a schematic showing 3H7-binding epitope in Tie2. Binding-epitope (red) of anti-Tie2 antibody (3H7) in hTie2 antigen analyzed by H / D exchange-mass spectrometry, visualized with hTie2 FNIII (1-3) crystal structure (PDB: 5UTK) using PyMol software will be.
  • Figure 7 shows the results of phosphorylation (pAkt) of Akt induced by humanized anti-Tie2 antibodies.
  • Serum starved HUVECs were incubated with humanized anti-Tie2 antibodies for 30 minutes. Cell lysates were then blotted with SDS-PAGE / Western and probed with p-Akt (s473) antibody or anti-Akt antibody.
  • FIG. 8 shows the increase in Schlemm's canal (SC) area and the decrease in IOP by humanized Tie2 antibody (3H7H12G4) in primary open-angle glaucoma mouse model.
  • Tamoxifen was administered to 8-week-old A1: A2 i ⁇ / ⁇ mice for inducible deletion of Angioprotein-1 and Angioprotein-2.
  • A2 i ⁇ / ⁇ mice for inducible deletion of Angioprotein-1 and Angioprotein-2.
  • 3H7H12G4 one eye, 1 mg injection of 5 mg / ml solution
  • Fc opposite eye, 1 ⁇ injection of 5 mg / ml solution
  • the immunostaining intensity of Prox1 and Tie2 at CD144 + SC area and CD144 + SC was measured 2 weeks after administration of 3H7H12G4.
  • CNV choroidal neovascularization
  • CNV laser induced choroidal neovascularization
  • FIG. 10 is a fluorescence photograph showing co-localization of 3H7H12G4 and CD31 in endothelial cells in the choroidal neovascularization (CNV) region.
  • CNV choroidal neovascularization
  • Figure 11 shows inhibition of choroidal neovascularization (CNV) by subcutaneous injection of 3H7H12G4.
  • CNV choroidal neovascularization
  • Tie2 antibody as an agent that increases the activity of Tie2 by binding to a Tie2 Ig3-FNIII (1-3) domain comprising the sequence of SEQ ID NO: 2 of the human Tie2 full length sequence of SEQ ID NO: 1.
  • Tie2 is a receptor protein that promotes the differentiation and stabilization of blood vessels. It is highly expressed in blood vessels of diseases such as cancer, and activating the Tie2 receptor stabilizes cancer blood vessels and aggregates surrounding supporting cells.
  • activating Tie2 of cancer vessels normalizes the cancer vessels, relieves the increase of hypoxia within the tumors, increases blood flow into the tumors, and provides sufficient oxygen supply. May increase the penetration of immune cells.
  • the present invention relates to a Tie2 antibody or antigen binding fragment thereof that binds to a Tie2 Ig3-FNIII (1-3) domain comprising the sequence of SEQ ID NO: 2.
  • antibody refers to an antibody that specifically binds to Tie2.
  • the scope of the present invention includes not only complete antibody forms that specifically bind Tie2, but also antigen binding fragments of such antibody molecules.
  • a complete antibody is a structure having two full length light chains and two full length heavy chains, each of which is linked by heavy and disulfide bonds.
  • the heavy chain constant region has gamma ( ⁇ ), mu ( ⁇ ), alpha ( ⁇ ), delta ( ⁇ ) and epsilon ( ⁇ ) types and subclasses gamma 1 ( ⁇ 1), gamma 2 ( ⁇ 2), and gamma 3 ( ⁇ 3). ), Gamma 4 ( ⁇ 4), alpha 1 ( ⁇ 1) and alpha 2 ( ⁇ 2).
  • the constant regions of the light chains have kappa ( ⁇ ) and lambda ( ⁇ ) types.
  • An antigen binding fragment or antibody fragment of an antibody means a fragment having an antigen binding function and includes Fab, F (ab '), F (ab') 2, Fv and the like.
  • Fab in the antibody fragment has a structure having a variable region of the light and heavy chains, a constant region of the light chain and the first constant region (CH1) of the heavy chain has one antigen binding site.
  • Fab ′ is a hinge region comprising one or more cysteine residues at the C-terminus of the heavy chain CH1 domain
  • F (ab ') 2 antibodies are produced by disulfide bonds of cysteine residues in the hinge region of Fab'.
  • Fv is the minimum antibody fragment which has only a heavy chain light region and a light chain variable region.
  • Double-chain Fv is a non-covalent bond in which a heavy chain variable region and a light chain variable region are linked, and a single chain Fv (single-chain Fv, scFv) is generally a variable region of the heavy chain and the light chain through a peptide linker. This covalent linkage or the C-terminus is directly linked to form a dimer-like structure such as a double-chain Fv.
  • Such antibody fragments can be obtained using proteolytic enzymes (e.g., restriction digestion of the entire antibody with papain yields Fab and cleavage with pepsin yields F (ab ') 2 fragments). It can also be produced by recombinant technology.
  • proteolytic enzymes e.g., restriction digestion of the entire antibody with papain yields Fab and cleavage with pepsin yields F (ab ') 2 fragments. It can also be produced by recombinant technology.
  • the antibody according to the invention is in Fv form (eg scFv) or is in the form of a complete antibody.
  • the heavy chain constant region may be selected from any one isotype of gamma ( ⁇ ), mu ( ⁇ ), alpha ( ⁇ ), delta ( ⁇ ) or epsilon ( ⁇ ).
  • the constant region is gamma 1 (IgG1), gamma 3 (IgG3) or gamma 4 (IgG4).
  • the light chain constant region may be of kappa or lambda type.
  • the term “heavy chain” refers to a variable region domain VH comprising an amino acid sequence having sufficient variable region sequence to confer specificity to an antigen and a full length heavy chain comprising three constant region domains CH1, CH2 and CH3 And fragments thereof.
  • the term “light chain” as used herein refers to a full-length light chain and fragment thereof comprising a variable region domain VL and a constant region domain CL comprising an amino acid sequence having sufficient variable region sequence to confer specificity to the antigen. All means.
  • Antibodies of the invention include monoclonal antibodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, single chain Fvs (scFV), single chain antibodies, Fab fragments, F (ab ') fragments, disulfide-binding Fvs (sdFV) And anti-idiotype (anti-Id) antibodies, or epitope-binding fragments of the antibodies, and the like.
  • Said monoclonal antibody refers to the same except for possible naturally occurring mutations in which antibodies obtained from substantially homogeneous antibody populations, ie, individual antibodies in the population, may be present in trace amounts.
  • Monoclonal antibodies are highly specific and are directed against a single antigenic site. In contrast to conventional (polyclonal) antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • Epitope refers to a protein determinant to which an antibody can specifically bind.
  • Epitopes usually consist of a group of chemically active surface molecules, such as amino acids or sugar side chains, and generally have specific three dimensional structural characteristics as well as specific charge characteristics. Three-dimensional epitopes and non-stereo epitopes are distinguished in that the binding to the former is lost but not to the latter in the presence of a denatured solvent.
  • Tie2 antibody or antigen-binding fragment thereof when identifying epitopes through hydrogen / deuterium exchange, TLSDILPPQPEN and / or amino acids 633 to 644 of Tie2 comprising the sequence of SEQ ID NO: 1 Binding to amino acids 713-726 of FAENNIGSSNPAFS.
  • the non-human (eg murine) antibody of the “humanized” form may comprise one or more amino acid sequences derived from one or more non-human antibodies (donor or source antibody) containing a minimal sequence derived from a non-human immunoglobulin (eg Eg, a CDR sequence).
  • humanized antibodies are non-human species (donor antibodies) that retain the desired specificity, affinity, and capacity for residues from the hypervariable region of the recipient, for example mice, rats, rabbits, or non-humans.
  • Human immunoglobulins (receptor antibodies) replaced with residues from the hypervariable regions of primates.
  • residues in one or more framework domains (FR) of the variable region of the recipient human antibody may be replaced with corresponding residues from the non-human species donor antibody. This helps to maintain proper three-dimensional organization of the grafted CDR (s), thereby improving affinity and antibody stability.
  • Humanized antibodies may include new residues that do not appear in additional recipient antibodies or donor antibodies, for example to further refine antibody performance.
  • human antibody refers to a molecule derived from human immunoglobulin, in which all amino acid sequences constituting the antibody including complementarity determining regions and structural regions are composed of human immunoglobulins.
  • While the heavy and / or light chain portions are the same or homologous to the corresponding sequences in an antibody derived from a particular species or belonging to a particular antibody class or subclass, the remaining chain (s) are derived from another species or another antibody class or Included are "chimeric" antibodies (immunoglobulins) that are identical or homologous to the corresponding sequences in antibodies belonging to the subclass, as well as fragments of such antibodies that exhibit the desired biological activity.
  • 11C4, 4A4, 3B2, 3E12, and 3H7 were prepared using a mouse-derived Tie2 antibody, and 3H7H11G4, 3H7H12G4, 3H7H21G4 or 3H7H22G4 were CDR-grafted and humanized antibodies from a mouse-derived 3H7 antibody as a source antibody. Produced.
  • antibody variable domain refers to the light and heavy chain portions of an antibody molecule comprising the amino acid sequences of complementarity determining regions (CDRs; ie CDR1, CDR2, and CDR3), and framework regions (FR). .
  • CDRs complementarity determining regions
  • FR framework regions
  • VH refers to the variable domain of the heavy chain.
  • VL refers to the variable domain of the light chain.
  • CDRs Complementarity Determining Regions
  • ie CDR1, CDR2, and CDR3 refer to amino acid residues of antibody variable domains that are required for antigen binding, each variable domain typically identified as CDR1, CDR2 and CDR3. Three CDR regions.
  • the Tie2 antibody or antigen-binding fragment thereof comprises a heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 3 to 5, a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 6 to 8 Light chain variable region;
  • a heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 13 to 15, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 16 to 18;
  • a heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 23 to 25, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 26 to 28;
  • a heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 33 to 35, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 36 to 38; or
  • a heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NO: 43 to 45, and a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NO: 46 to 48.
  • FRs Framework regions
  • Each variable domain typically has four FRs identified as FR1, FR2, FR3 and FR4.
  • Tie2 antibodies are monovalent or bivalent and include single or double chains. Functionally, the binding affinity of Tie2 antibodies is in the range of 10-5M to 10-12M.
  • the binding affinity of a Tie2 antibody is 10 ⁇ 6 M to 10 ⁇ 12 M, 10 ⁇ 7 M to 10 ⁇ 12 M, 10 ⁇ 8 M to 10 ⁇ 12 M, 10 ⁇ 9 M to 10 ⁇ 12 M, 10 -5 M to 10 -11 M, 10 -6 M to 10 -11 M, 10 -7 M to 10 -11 M, 10 -8 M to 10 -11 M, 10 -9 M to 10 -11 M, 10 -10 M to 10 -11 M, 10 -5 M to 10 -10 M, 10 -6 M to 10 -10 M, 10 -7 M to 10 -10 M, 10 -8 M to 10 -10 M, 10 -9 M to 10 -10 M, 10 -5 M to 10 -9 M, 10 -6 M to 10 -9 M, 10 -7 M to 10 -9 M, 10 -8 M to 10 -9 M, 10
  • the antibody or antigen-binding fragment thereof that binds Tie2 may comprise a heavy chain variable region comprising an amino acid sequence of SEQ ID NO: 9 and a heavy chain variable region comprising an amino acid sequence of SEQ ID NO: 11;
  • a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 19 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 21;
  • a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 29 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 31;
  • a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 39 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 41;
  • a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 49 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 51;
  • a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 53 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 54;
  • a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 57 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 58;
  • a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 61 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 62; or
  • a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 65 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 66.
  • Antibodies or antibody fragments of the present invention may include not only the sequences of the anti-Tie2 antibodies of the present invention described herein, but also biological equivalents thereof, as long as they can specifically recognize Tie2.
  • further changes can be made to the amino acid sequence of the antibody to further improve the binding affinity and / or other biological properties of the antibody.
  • Such modifications include, for example, deletions, insertions and / or substitutions of amino acid sequence residues of the antibody.
  • Such amino acid variations are made based on the relative similarity of amino acid side chain substituents such as hydrophobicity, hydrophilicity, charge, size, and the like.
  • arginine, lysine and histidine are all positively charged residues; Alanine, glycine and serine have similar sizes; It can be seen that phenylalanine, tryptophan and tyrosine have a similar shape.
  • arginine, lysine and histidine; Alanine, glycine and serine; Phenylalanine, tryptophan and tyrosine are biologically equivalent functions.
  • the amino acid sequence possessed by the antibody of the present invention or the nucleic acid molecule encoding the same is interpreted to include a sequence showing substantial identity with the sequence described in SEQ ID NO.
  • the above substantial identity is at least 90% when the sequences of the present invention are aligned as closely as possible with any other sequences, and the aligned sequences are analyzed using algorithms commonly used in the art.
  • a homology most preferably at least 95% homology, at least 96%, at least 97%, at least 98%, at least 99% homology. Alignment methods for sequence comparison are known in the art.
  • BLAST The NCBI Basic Local Alignment Search Tool (BLAST) is accessible from NBCI and the like and can be used in conjunction with sequence analysis programs such as blastp, blasm, blastx, tblastn and tblastx on the Internet.
  • BLSAT is accessible at www.ncbi.nlm.nih.gov/BLAST/. Sequence homology comparisons using this program can be found at www.ncbi.nlm.nih.gov/BLAST/blast_help.html.
  • the antibody or antigen-binding fragment thereof of the present invention is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% compared to the specified sequence or all described in the specification. , 99%, or more homology.
  • homology can be determined by sequence comparison and / or alignment by methods known in the art. For example, sequence comparison algorithms (ie, BLAST or BLAST 2.0), manual alignment, visual inspection can be used to determine the percent sequence homology of nucleic acids or proteins of the invention.
  • the present invention relates to a nucleic acid encoding the antibody or antigen-binding fragment thereof.
  • the nucleic acid may comprise the sequence of SEQ ID NO: 10, 12, 20, 22, 30, 32, 40, 42, 50, 52, 55, 56, 59, 60, 63, 64, 67 or 68.
  • the nucleic acid encoding the antibody or antigen-binding fragment thereof of the present invention can be isolated to recombinantly produce the antibody or antigen-binding fragment thereof.
  • the nucleic acid is isolated and inserted into a replicable vector for further cloning (amplification of DNA) or for further expression. Based on this, the present invention relates to a vector comprising the nucleic acid in another aspect.
  • Nucleic acid is meant to encompass DNA (gDNA and cDNA) and RNA molecules inclusively, and the nucleotides that are the basic building blocks of nucleic acids include natural nucleotides as well as analogs with modified sugar or base sites. .
  • the sequences of nucleic acids encoding heavy and light chain variable regions of the invention can be modified. Such modifications include addition, deletion, or non-conservative or conservative substitutions of nucleotides.
  • the DNA encoding the antibody is readily isolated or synthesized using conventional procedures (e.g., by using oligonucleotide probes capable of specifically binding to the DNA encoding the heavy and light chains of the antibody).
  • Many vectors are available.
  • Vector components generally include, but are not limited to, one or more of the following: signal sequence, origin of replication, one or more marker genes, enhancer elements, promoters, and transcription termination sequences.
  • the term "vector” refers to a plasmid vector as a means for expressing a gene of interest in a host cell; Cosmid vector; Viral vectors such as bacteriophage vectors, adenovirus vectors, retrovirus vectors, and adeno-associated virus vectors, and the like.
  • the nucleic acid encoding the antibody in the vector is operably linked with a promoter.
  • “Operatively linked” means a functional binding between a nucleic acid expression control sequence (eg, an array of promoters, signal sequences, or transcriptional regulator binding sites) and another nucleic acid sequence, whereby the regulatory sequence is the other nucleic acid. To control transcription and / or translation of the sequence.
  • a nucleic acid expression control sequence eg, an array of promoters, signal sequences, or transcriptional regulator binding sites
  • promoters capable of promoting transcription e.g., tac promoter, lac promoter, lacUV5 promoter, lpp promoter, pL ⁇ promoter, pR ⁇ promoter, rac5 promoter, amp promoter, recA promoter, SP6 promoter, trp promoter and T7 promoter, etc.
  • ribosome binding sites for initiation of translation e.g., amp promoter, recA promoter, SP6 promoter, trp promoter and T7 promoter, etc.
  • a promoter derived from the genome of the mammalian cell e.g., a metallothionine promoter, a ⁇ -actin promoter, a human heroglobin promoter and a human muscle creatine promoter
  • a mammal Promoters derived from animal viruses e.g., adenovirus late promoter, vaccinia virus 7.5K promoter, SV40 promoter, cytomegalovirus (CMV) promoter, tk promoter of HSV, mouse breast tumor virus (MMTV) promoter, LTR promoter of HIV
  • a promoter derived from the genome of the mammalian cell e.g., a metallothionine promoter, a ⁇ -actin promoter, a human heroglobin promoter and a human muscle creatine promoter
  • a mammal Promoters derived from animal viruses e.g., adenovirus late promoter, vaccinia virus 7.5K promoter, SV40 promoter
  • the vector may be fused with other sequences to facilitate purification of the antibody expressed therefrom.
  • Sequences to be fused include, for example, glutathione S-transferase (Pharmacia, USA), maltose binding protein (NEB, USA), FLAG (IBI, USA) and 6x His (hexahistidine; Quiagen, USA).
  • Such vectors include antibiotic resistance genes commonly used in the art as selectable markers and include, for example, ampicillin, gentamicin, carbenicillin, chloramphenicol, streptomycin, kanamycin, geneticin, neomycin and tetracycline. There is a resistance gene.
  • the present invention relates to a cell transformed with the above-mentioned vector.
  • the cells used to produce the antibodies of the invention can be prokaryote, yeast or higher eukaryote cells, but are not limited thereto.
  • Bacillus strains such as Escherichia coli, Bacillus subtilis and Bacillus thuringiensis, Streptomyces, Pseudomonas (e.g. Pseudomonas putida), Proteus Prokaryotic host cells such as Proteus mirabilis and Staphylococcus (eg, Staphylocus carnosus) can be used.
  • Pseudomonas e.g. Pseudomonas putida
  • Proteus Prokaryotic host cells such as Proteus mirabilis and Staphylococcus (eg, Staphylocus carnosus) can be used.
  • examples of useful host cell lines are COS-7, BHK, CHO, CHOK1, DXB-11, DG-44, CHO / -DHFR, CV1, COS-7, HEK293, BHK, TM4, VERO, HELA, MDCK, BRL 3A, W138, Hep G2, SK-Hep, MMT, TRI, MRC 5, FS4, 3T3, RIN, A549, PC12, K562, PER.C6, SP2 / 0, NS-0 , U20S, or HT1080, but is not limited thereto.
  • the present invention (a) culturing the cells; And (b) recovering the antibody or antigen-binding fragment thereof from the cultured cells.
  • the cells can be cultured in various media. It can be used as a culture medium without limitation among commercial media. All other necessary supplements known to those skilled in the art may be included at appropriate concentrations. Culture conditions, such as temperature, pH, and the like, are already in use with host cells selected for expression, which will be apparent to those skilled in the art.
  • the recovery of the antibody or antigen-binding fragment thereof can be removed by, for example, centrifugation or ultrafiltration, and the resultant can be purified using, for example, affinity chromatography or the like. Further other purification techniques such as anion or cation exchange chromatography, hydrophobic interaction chromatography, hydroxylapatite chromatography and the like can be used.
  • the present invention relates to a composition for preventing or treating angiogenic diseases comprising the antibody or antigen-binding fragment thereof as an active ingredient.
  • Angiogenesis refers to the formation or growth of new blood vessels from previously existing blood vessels, and “angiogenesis-related disease” refers to a disease associated with the development or progression of angiogenesis. If the antibody can be treated, the disease can be included within the scope of angiogenesis-related diseases without limitation.
  • angiogenesis-related diseases include cancer, metastasis, diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, macular degeneration, angiogenesis Neovascular glaucoma, erythrosis, proliferative retinopathy, psoriasis, hemophilic arthritis, capillary formation of atherosclerotic plaques, keloid , Wound granulation, vascular adhesion, rheumatoid arthritis, osteoarthritis, autoimmune diseases, Crohn's disease, restenosis , Atherosclerosis, intestinal adhesions, cat scratch disease, ulcers, liver cirrhosis, nephritis, diabetic kidney One comprising a ring (diabetic nephropathy), diabetes mellitus (diabetes mellitus), inflammatory diseases (inflammatory diseases) and neurodegenerative disorders (neurodegenerative diseases), but is not limited to such.
  • the cancer is esophageal cancer (esophageal cancer), stomach cancer (stomach cancer), colon cancer (large intestine cancer), rectal cancer (rectal cancer), oral cancer (oral cancer), pharyngeal cancer (pharynx cancer), larynx cancer (larynx cancer), Lung cancer, colon cancer, breast cancer, breast cancer, uterine cervical cancer, endometrial cancer, ovarian cancer, prostate cancer, testicular cancer testis cancer, bladder cancer, renal cancer, liver cancer, liver cancer, pancreatic cancer, bone cancer, connective tissue cancer, skin cancer, Selected from the group consisting of brain cancer, thyroid cancer, leukemia, Hodgkin's lymphoma, lymphoma, and multiple myeloid blood cancer It is not.
  • prevention or prophylaxis refers to any measure which results in inhibiting or delaying the onset of the disease of interest by administering the antibody or composition of the invention.
  • treatment or therapy refers to any measure by which an antibody or composition of the invention is administered to improve or ameliorate the symptoms of a disease of interest.
  • compositions comprising the antibodies of the invention are preferably pharmaceutical compositions and may further comprise suitable vehicles, excipients or diluents typically used in the art.
  • compositions comprising pharmaceutically acceptable vehicles include tablets, pills, powders, granules, capsules, suspensions, internal solutions, emulsions, syrups, sterile aqueous solutions, non-aqueous solutions, suspensions, lyophilizates and suppositories. It can be in various oral or parenteral dosage forms.
  • the pharmaceutical compositions of the present invention may be formulated in combination with diluents or excipients such as fillers, thickeners, binders, wetting agents, disintegrants, surfactants and the like.
  • Solid preparations for oral administration may be in the form of tablets, pills, powders, granules, capsules and the like.
  • the compounds of the present invention may be formulated in combination with one or more excipients such as starch, calcium carbonate, sucrose, lactose, or gelatin.
  • Lubricants such as simple excipients, magnesium stearate, talc and the like can be further used.
  • Liquid preparations for oral administration may be suspensions, internal solutions, emulsions, syrups and the like.
  • excipients such as simple diluents such as water or liquid paraffin, wetting agents, sweeteners, aromatics, preservatives and the like can be included in the liquid formulation.
  • compositions of the present invention may be in parenteral dosage forms such as sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilisates, suppositories, and the like.
  • parenteral dosage forms such as sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilisates, suppositories, and the like.
  • Injectable propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and esters such as ethyl oleate may be suitable for non-aqueous solvents and suspensions.
  • Base materials for suppositories include Whitepsol, macrogol, Tween 61, cacao butter, laurin butter and glycerogelatin.
  • composition of the present invention is administered in a pharmaceutically effective amount.
  • pharmaceutically effective amount refers to the amount of a pharmaceutical composition for treating a disease sufficient to a reasonable benefit and risk ratio applicable to all medical treatments. Effective amounts include 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 time of administration, the route of administration, the rate of secretion, the duration of treatment, the coadministration of the drug, and other known in the art. It depends on various factors including the parameters.
  • the compositions of the present invention can be administered alone or in combination with other therapies. In such cases, they may be administered sequentially or simultaneously with conventional therapies.
  • the composition may be administered in one or multiple doses. Given these factors completely, it is important to administer the minimum amount sufficient to achieve the maximum effect without side effects, which dose can be readily determined by one of ordinary skill in the art.
  • the dosage of the pharmaceutical composition of the present invention is not particularly limited, but varies depending on various factors including the patient's health condition and weight, the severity of the disease, the type of drug, the route of administration, and the time of administration.
  • the compositions are typically accepted routes into mammals, including rats, mice, livestock, humans, etc., once or in multiple doses per day, for example, orally, rectally, intravenously, subcutaneously. It may be administered intrauterinely or intratracerebrovascularly.
  • the present invention relates to a method for inhibiting angiogenesis, a method for preventing or treating angiogenesis-related diseases, comprising administering the antibody or the composition to a subject in need.
  • the methods of the present invention comprise administering a pharmaceutically effective amount of the pharmaceutical composition to a subject in need thereof.
  • the subject may be a mammal such as a dog, cow, horse, rabbit, mouse, rat, chicken and human, but is not limited thereto.
  • the pharmaceutical composition includes parenterally, subcutaneously, intraperitoneally, intrapulmonarily or intranasally, if necessary intratralesional administration for topical treatment. Can be administered by any suitable method.
  • Preferred dosages of the pharmaceutical compositions of the present invention vary depending on various factors, including the health and weight of the individual, the severity of the disease, the type of medicament, the route of administration and the time of administration, and can be readily determined by one skilled in the art.
  • the present invention relates to a pharmaceutical composition for preventing or treating cancer, comprising the antibody, or a method for preventing or treating cancer, comprising administering the antibody or the composition to an individual in need thereof.
  • a pharmaceutical composition for preventing or treating cancer comprising the antibody, or a method for preventing or treating cancer, comprising administering the antibody or the composition to an individual in need thereof.
  • the terms 'antibody', 'prophylaxis' and 'treatment' are as mentioned above.
  • the cancer is not limited.
  • the antibody of the present invention can prevent the development or progression of cancer by inhibiting angiogenesis.
  • cancer include esophageal cancer, stomach cancer, large intestine cancer, rectal cancer, oral cancer, pharynx cancer, larynx cancer, lung cancer Lung cancer, colon cancer, breast cancer, breast cancer, uterine cervical cancer, endometrial cancer, ovarian cancer, prostate cancer, testicular cancer testis cancer, bladder cancer, renal cancer, liver cancer, liver cancer, pancreatic cancer, bone cancer, connective tissue cancer, skin cancer, Brain cancer, thyroid cancer, leukemia, Hodgkin's lymphoma, lymphoma and multiple myeloid blood cancer.
  • the antibodies of the invention can be used in combination with other antibodies or biologically active agents or materials for various purposes.
  • the present invention relates to a composition for co-administration with other therapeutic agents for angiogenic diseases comprising the antibody or antigen-binding fragment thereof in this respect.
  • Such other angiogenic disease therapeutic agents include anti-angiogenic drugs, anti-inflammatory drugs and / or anticancer drugs. This can overcome the resistance of each other and improve the efficacy.
  • the Tie2 antibody and the therapeutic agent for other angiogenic diseases may be administered sequentially or simultaneously.
  • a composition comprising an antibody against Tie2 or an antigen binding fragment thereof as an active ingredient is administered to the subject, or the composition is administered.
  • anti-angiogenic drugs, anti-inflammatory drugs, and / or anticancer drugs are administered to the subject.
  • the composition may be administered to the subject simultaneously with the anti-angiogenic drug, anti-inflammatory drug and / or anticancer drug.
  • the Ig3-FNIII (1-3) domain of human Tie2 (hTie2-Ig3-FNIII (1-3), SEQ ID NO: 2) was cloned into a vector containing a CMV promoter and transfected into a HEK293F cell line to express temporarily. . After incubation for 5 days, the expressed recombinant hTie2-Ig3-FNIII (1-3) protein was purified by an affinity column using Protein A. Twice a week for 6 weeks, 5 weeks old BALB / c mice were immunized with the injection of purified hTie2-Ig3-FNIII (1-3) mixed with adjuvant (100 ⁇ g / injection).
  • Tie2 antibody titers in the sera of immunized mice were examined with a human Tie2 (hTie2) ELISA kit (R & D).
  • hTie2 human Tie2
  • ELISA kit R & D
  • spleens were extracted from immunized mice
  • B lymphocytes were isolated from the spleen
  • myeloma cells SP2 / 0
  • the fused cells were cultured in HAT medium containing hypoxanthine, aminopterin and thymidine, and hybridoma cells containing only fusions of myeloma cells and B lymphocytes were selected and cultured.
  • hybridoma cells were cultured in 10% FBS containing-DMEM (Dulbesco Modified Eagle's Medium) in T75 (75 cm 2 area) flasks. When the concentration of cells reached about 90%, the cells were washed with PBS, incubated in 50 ml of serum-free medium (SFM, Gibco), and then incubated at 37 ° C for 3 days. Thereafter, the culture solution containing the antibody secreted from each monoclonal hybridoma was collected, centrifuged to remove cells, and the culture supernatant was collected and filtered.
  • FBS Dulbesco Modified Eagle's Medium
  • Antibodies were purified from the culture supernatant using an AKTA purification apparatus (GE Healthcare.) Equipped with a Protein G protein affinity column (GE Healthcare.). Thereafter, the buffer of the antibody solution was replaced with PBS using a centrifugal filter unit (Amicon), and the purified antibody was concentrated.
  • AKTA purification apparatus GE Healthcare.
  • Protein G protein affinity column GE Healthcare.
  • HUVEC (1 ⁇ 10 5 cells / ml) was incubated at 37 ° C. in EGM-2 medium (Lonza) in a 60 mm culture dish. Cells of 90% density were incubated for 6 hours in serum-free EBM-2 medium to bring serum starvation. Serum starved HUVECs were treated with anti-Tie2 antibody and further incubated for 30 minutes. Cells were washed with cold PBS, then treated with lysis buffer and lysed at 4 ° C. for 20 minutes. Subsequently, the precipitate was removed by centrifugation at 13000 rpm for 15 minutes to obtain cell lysate. 5x SDS sample buffer was added to the cell lysate, followed by protein electrophoresis (SDS-PAGE), followed by transfer to nitrocellulose membrane (NC membrane, GE).
  • SDS-PAGE protein electrophoresis
  • the affinity of the mouse monoclonal antibody for hTie2 was measured using an octet system (ForteBio) using Black 96-well plate (Greiner). The biosensors used for affinity measurements were hydrated for 10 minutes before being measured with an AR2G tip (ForteBio Octet). After hydration, hTie2 was diluted to 10 ⁇ g / ml concentration in 10 mM sodium acetate, pH6.0 buffer, fixed on AR2G biosensor and blocked with 1M ethanolamine. Mouse anti-Tie2 monoclonal antibodies were diluted to 50, 25, 12.5, 6.25, 3.125 and 0 nM with 1 ⁇ kinetic buffer, then bound for 300 seconds and dissociated for 900 seconds.
  • Anti-Tie2 antibodies according to the present invention appear to bind to Tie2 and induce Tie2 clustering, ultimately leading to Tie2 activation. Experiments were performed to analyze the effect of anti-Tie2 antibodies on Tie2 phosphorylation using HUVECs.
  • HUVEC was incubated in 37%, 5% CO 2 concentration conditions using EGM-2 (Lonza) medium in a 100mm culture dish.
  • EGM-2 Ligno-Benoyl-N-phenyl-N-phenyl-N-phenyl-N-phenyl-N-phenyl-N-phenyl-N-phenyl-N-phenyl-N-phenyl-N-phenyl-N-phenyl-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-N-phenyl
  • Tie2 antibody R & D systems, AF313
  • Dynabeads TM Protein G Life technologies
  • the beads were fixed on one side of the tube using a magnet and washed three times with lysis buffer and then incubated for 10 minutes at 70 ° C. with a 2 ⁇ SDS sample buffer containing reducing agent. Beads were removed from the sample and electrophoresed on 4-15% SDS protein gel (Bio-Rad) and then transferred to 0.45 ⁇ m PVDF membrane.
  • Membrane was blocked for 1 hour at room temperature using TBS-T mixed with 5% (v / v) BSA and incubated with anti-phosphotyrosine antibody (4G10, Millipore) at 4 ° C. for 8 hours before HRP conjugate
  • the anti-mouse antibody (HRP conjugated anti-mouse antibody) was incubated and analyzed by Western blot.
  • the membrane was reacted in stripping buffer (Thermo) for 15 minutes, then blocked again and probed again with anti-Tie2 antibodies (R & D systems, AF313). As shown in FIG.
  • anti-Tie2 antibody (3H7) induces activation of Tie2 receptor directly in human vascular endothelial cells.
  • HUVECs Intracellular influx of Tie2 by 3H7 antibody in HUVEC and FOXO1 translocation from nucleus to cytoplasm were examined by immunofluorescence. Specifically, HUVECs were inoculated in an 8-well slide chamber (Lab-TekII) and maintained in EGM-2 medium for 2-3 days. At 100% confluence, cells were serum starved for 4 hours using EBM-2 medium and then treated with 1 ⁇ g / ml Tie2 antibody (3H7) for 30 minutes.
  • Cells were then fixed with 4% formaldehyde / PBS solution for 10 minutes at room temperature (RT), permeabilized with 0.1% Triton X-100 / PBS solution, blocked with 1% BSA / PBS solution for 60 minutes at room temperature and , Incubated with primary antibody for 1 hour at RT.
  • primary antibodies antibodies against hTie2 and FOXO1 were used.
  • the cells were incubated with fluorescently labeled secondary antibody in dark conditions at room temperature for 1 hour, and mounted with a Vectashield mounting medium containing DAPI (Vector Labs) for nuclear staining. Cell images were then taken and analyzed with a laser scanning confocal microscope (LSM880, Carl Zeiss).
  • Vascular leak analysis in HUVEC was performed using the In Vitro Vascular Permeability Assay Kit (Millipore) according to the manufacturer's instructions. HUVECs were inoculated into transwell plate inserts and incubated for 3 days until 100% density. Ang2 (1 ⁇ g / ml) alone, a mixture of Ang2 (1 ⁇ g / ml) and control antibody (1 ⁇ g / ml) or 3H7 antibody (1 ⁇ g / ml) alone was preincubated in HUVECs for 30 minutes each, followed by VEGF (500 ng / ml). Or TNF- ⁇ (100 ng / ml) and cells were incubated separately at 37 ° C. for 45 minutes or 22 hours.
  • FITC-dextran was then added to the upper chamber and incubated for 20 minutes. The amount of FITC-dextran that penetrated the HUVEC monolayer and accumulated in the lower chamber was measured with a fluorescence reader at excitation and emission wavelengths of 485 and 535 nm, respectively. As shown in FIG. 4, pretreatment of anti-Tie2 antibody (3H7) inhibited vascular leakage caused by VEGF or TNF- ⁇ , a vascular leak promoter.
  • DNA sequences of the antibodies selected from Example 1.3 were analyzed. Specifically, hybridoma cells (2 ⁇ 10 6 cells / ml) were incubated in DMEM containing 10% FBS and total RNA was obtained using RNeasy mini kit (Qiagen). Next, RNA concentration was measured and cDNA was synthesized through reverse transcription (RT) reaction. To amplify heavy and light chain variable region gene sequences, PCR was performed using a mouse Ig-primer set (Novagen) under the following conditions with cDNA as a template: 94 ° C.
  • HDX-MS hydrogen / deuterium exchange-mass spectrometry
  • Binding-epitope of antibody (3H7) was analyzed using recombinant hTie2-Ig3-FNIII (1-3) protein.
  • the prepared hTie2-Ig3-FNIII (1-3) / antibody complex was diluted 15-fold with deuterium labeling buffer, the labeling proceeded several times, and quenched using the same volume of quenching buffer.
  • Labeling reaction times were 0 minutes (undeuterium), 0.33 minutes, 10 minutes, 60 minutes and 240 minutes, respectively. However, in deuterium conditions, deuterium labeling buffer was replaced with equilibrium buffer and the reaction was immediately stopped with quenching buffer. To perform mass spectrometry, deuterium-labeled hTie2-Ig3-FNIII (1-3) / antibody samples were loaded into a pepsin column and peptide digestion was performed. Mass spectrometry yielded 82.6% coverage data from a total of 50 peptides.
  • the differences in deuterium uptake between hTie2-Ig3-FNIII (1-3) and hTie2-Ig3-FNIII (1-3) / antibody complex conditions were analyzed. Corresponds to site or structurally altered site.
  • the deuterium uptake difference between hTie2-Ig3-FNIII (1-3) and hTie2-Ig3-FNIII (1-3) / antibody complex is considered significant when 0.5-1 Da or more, and is shown in bold in Table 13 .
  • H / D exchange-mass spectrometry data (FIG. 5) in the presence or absence of anti-Tie2 antibody (3H7) was used to create a heatmap for deuterium uptake per residue at 0.333, 10, 60 and 240 minutes for hTie2 antigen. It was.
  • the color scale represents the percentage of H / D exchange per residue between the antigen alone and the mixture of antigen / monoclonal antibodies at each individual time. Red color indicates increased deuterium exchange and blue color indicates no absorption.
  • the antibody was humanized in the following manner to remove the immunogenicity of the mouse anti-Tie2 antibody (3H7) when administered to humans.
  • the human antibody heavy chain variable gene IGHV1-46-01 showed 66% homology with the heavy chain sequence of antibody 3H7. Based on this analysis, three CDR regions of the 3H7 antibody were implanted into the human antibody heavy chain variable gene IGHV1-46-01. In this process, two humanized heavy chain antibody genes were designed (Table 14). Transplanted CDRs are underlined in the protein sequence. A back mutation to the mouse sequence was introduced into the heavy chain gene of humanized 3H7, shown in bold in the protein sequences of Table 14.
  • IGKV1-17-01 a human antibody light chain variable gene
  • IGKV1-17-01 a human antibody light chain variable gene
  • three CDR regions of the 3H7 antibody were implanted into the human antibody light chain variable gene IGKV1-17-01.
  • two humanized light chain antibody genes were designed (Table 14). Transplanted CDRs are underlined in the protein sequence. A back mutation into the mouse sequence was introduced into the light chain gene of humanized 3H7, shown in bold in the protein sequences of Table 14.
  • the humanized variable regions of the antibodies listed in Table 14 were cloned into the heavy and light chain vectors of the human IgG4 isotype backbone vector.
  • DNA fragments of the humanized heavy chain variable region (VH) of the antibody were synthesized into the sequence 'EcoRI-signal sequence-VH-NheI-CH-XhoI' (Bioneer, Inc.).
  • DNA fragments of the humanized light chain variable region (VL) of the antibody were also synthesized into the sequence 'EcoRI-signal sequence-VL-BsiWI-CL-XhoI'.
  • the DNA fragments encoding the heavy and light chains were cloned into pOptiVEC TM or pcDNA TM 3.3 vectors, respectively.
  • Expi293F (Gibco) was used, which can produce highly efficient recombinant proteins.
  • Expi293F cells (2x10 6 cells / ml) were cultured in Erlenmeyer flasks and plasmids encoding heavy and light chains were co-transfected into Expi293F cells using the ExpiFectamine 293 transformation kit. Cells were then incubated for 5 days at 37 ° C., 8% CO 2 conditions in a shaking incubator (orbital shaker, 125 rpm). The resulting cell culture medium was collected and centrifuged to remove the cells. Culture supernatants containing secreted antibodies were isolated and stored at 4 ° C. or immediately purified using an AKTA purification system (GE Healthcare) equipped with Protein A agarose column (GE Healthcare). Purified antibody was concentrated through a protein centrifuge filter (Amicon) to replace antibody buffer with PBS.
  • AKTA purification system GE Healthcare
  • Protein A agarose column
  • the affinity of the humanized Tie2 antibody for hTie2 was measured using Octet system (ForteBio) using black 96-well plates (96 well F-type black plates, Greiner). Biosensors used for affinity measurements were hydrated for 10 minutes before being measured with an AR2G tip (ForteBio Octet). After hydration the humanized anti-Tie2 antibody was diluted to 10 ⁇ g / ml concentration in 10 mM sodium acetate, pH6.0 buffer, fixed in AR2G biosensor and blocked with 1 M ethanolamine. Recombinant hTie2 was diluted to 50, 25, 12.5, 6.25, 3.125 and 0 nM using 1 ⁇ kinetic buffer, bound for 300 seconds and dissociated for 900 seconds.
  • HUVECs were treated with humanized Tie2 antibodies. Thereafter, the level of Akt phosphorylation, a major downstream signaling protein of the Tie2 receptor, was measured by immunoblotting.
  • COMP-Ang1 CA1
  • HUVEC cells (1 ⁇ 10 5 cells / ml) were incubated at 37 ° C. in EGM-2 medium (Lonza) in a 60 mm culture dish. 90% confluency cells were incubated with EBM-2 (Lonza) for 4 hours.
  • Serum-starved HUVECs were treated with anti-Tie2 antibodies and further incubated for 30 minutes.
  • the cells were washed with cold PBS, then treated with lysis buffer and lysed at 4 ° C. for 20 minutes.
  • Cell lysates were then prepared by centrifugation at 13000 rpm for 15 minutes.
  • 5 ⁇ SDS sample buffer was added to the cell lysate and the mixture was heated at 95 ° C. for 5 minutes. The mixture was then SDS-PAGE followed by Western blot.
  • the membrane was blocked for 1 hour at room temperature using TBST containing 5% skim milk and incubated with p-Akt antibody (S473) at 4 ° C. for about 8 hours. The amount of p-Akt was detected by ECL (Enhanced ChemiLuminescence). The membrane was then incubated for 15 minutes in stripping buffer (Thermo) and then probed again with anti-Akt antibody to determine the total amount of Akt.
  • p-Akt antibody S473
  • ECL Enhanced ChemiLuminescence
  • Akt phosphorylation was significantly increased by treatment of humanized 3H7 antibody.
  • a laser induced CNV model was used to test the ability of 3H7H12G4 to suppress choroidal neovascularization (CNV), which is characteristic of wet age-related macular degeneration (AMD).
  • CNV choroidal neovascularization
  • ALD wet age-related macular degeneration
  • the laser photocoagulator (Lumenis Inc.) and glass coverslips were used as contact lenses to visualize the retina.
  • a Nanoliter 2000 micro-injector (World Precision Instruments) equipped with a glass capillary pipette was used and each reagent containing 5 ⁇ g ( ⁇ 1 ⁇ l, 5 mg / ml) was added to the vitreous cavity. Injection.
  • CD31 + CNV volumes for the retinal pigment epithelium (RPE) -choroid-scleral planar mounts were calculated using the MATLAB image processing toolbox (MathWorks).
  • CD31 antibody (1: 200, Millipore) was used to detect endothelial cells of CNV.
  • VEGF-Trap effectively induced CNV degeneration by 64.4% compared to Fc, and 3H7H12G4 similarly induced CNV degeneration (65.7%) (FIG. 9B).
  • Fluorescein angiography (FA) and indocyanine green angiography (ICGA) were combined to measure vascular leakage in neovascularization around the area of laser injury.
  • Continuous-wave laser modules of 488nm and 755nm were used as stimulators of Fluorescein and ICG, respectively.
  • the raster scanning pattern of the stimulus laser consists of a scanner system consisting of a rotating polygonal mirror (MC-5; Lincoln Laser) and a galvanometer-based scanning mirror (6230H; Cambridge technology), which is transmitted to the rear aperture of the imaging lens.
  • An objective lens (PlanApo ⁇ , NA 0.75; Nikon) with a high numerical aperture (NA) was used as the imaging lens for wide-field fundus fluorescence images.
  • Fluorescence signals detected with a photoamplifier tube (R9110; Hamamatsu Photonics) were digitized with a frame grabber and reconstructed into an image of 512x512 pixels per frame.
  • a photoamplifier tube R9110; Hamamatsu Photonics
  • 10 mg of fluorescein sodium (Alcon) and 0.15 mg of Daiichi Pharmaceutical (ICG) were administered intraperitoneally and intravenously, respectively.
  • Imaging procedures were performed with general anesthesia and pupil dilation to improve the quality of the image.
  • VEGF-Trap 37.0%) and 3H7H12G4 (24.6%) similarly inhibited vascular leakage (FIG. 9C).
  • Fc treated group showed no significant difference in vascular leakage between 6 and 14 days after laser photocoagulation, but VEGF-Trap and 3H7H12G4 significantly reduced vascular leakage (45.6% and 42.5%, respectively). Therefore, there was no quantitative difference in the magnitude of inhibition of CNV and vascular leakage between VEGF-Trap and 3H7H12G4 in a mouse model of laser induced CNV.
  • 3H7H12G4 co-localization of 3H7H12G4 and CD31 in CNV endothelial cells was evaluated.
  • 3H7H12G4 25 mg / kg
  • Fc 25 mg / kg
  • co-localization of 3H7H12G4 and CD31 antibodies was directly detected in endothelial cells of CNV via anti-human IgG antibody (1: 1000, Jackson ImmunoResearch Laboratories) (FIG. 10A).
  • the administered 3H7H12G4 was detectable to a high extent in CD31 + endothelial cell CNV (FIGS. 10B-10D).
  • Example 10 CNV inhibitory effect by 3H7H12G4 antibody subcutaneous injection.
  • Subcutaneous administration of 3H7H12G4 was performed one day after laser photocoagulation to determine the effect of 3H7H12G4 subcutaneous injection on CNV inhibition.
  • Fc 25 mg / kg was administered to mice in the same manner as a control.
  • Anti-CD31 antibody (1: 200, Millipore) was used for detection in CNV endothelial cells and CD31 + CNV volume for retinal pigment epithelium (RPE) -choroid-scleral plane mounts on day 8 laser photocoagulation was calculated using the MATLAB image processing toolbox (MathWorks) (FIG. 11A).
  • 3H7H12G4 effectively inhibited CNV production by 69.9% compared to Fc (FIGS. 11B, 11C), indicating that not only intravitreal injection but also subcutaneous injection of 3H7H12G4 has an inhibitory effect on CNV.
  • the antibody or antigen-binding fragment thereof that binds Tie2 according to the present invention binds to Tie2 with high affinity while maintaining cross-reactivity with humans and mice, and exhibits the desired antigen reactivity.
  • the Tie2 antibody may be usefully used for the prevention or treatment of the desired angiogenic disease by inducing phosphorylation of Tie2 and activation of the Tie2 receptor.

Abstract

The present invention relates to an antibody against Tie-2 or an antigen binding fragment thereof, a nucleic acid for coding same, a vector comprising said nucleic acid, a cell transformed with said vector, a method for producing said antibody or the antigen binding fragment thereof, and a composition for preventing or treating angiogenic diseases, comprising said antibody or the antigen binding fragment thereof.

Description

TIE2에 결합하는 항체 및 이의 용도Antibodies that bind to TIE2 and uses thereof
본 발명은 Tie-2에 대한 항체 또는 이의 항원 결합 단편, 이를 코딩하는 핵산, 상기 핵산을 포함하는 벡터, 상기 벡터로 형질전환된 세포, 상기 항체 또는 그의 항원 결합 단편의 제조방법 및 이를 포함하는 혈관신생 질환의 예방 또는 치료용 조성물에 관한 것이다.The present invention provides an antibody against Tie-2 or an antigen-binding fragment thereof, a nucleic acid encoding the same, a vector comprising the nucleic acid, a cell transformed with the vector, a method for producing the antibody or an antigen-binding fragment thereof, and a blood vessel comprising the same. It relates to a composition for preventing or treating neoplastic disease.
혈관신생은 개체의 발생, 성장, 보존 및 항상성 유지 과정에서 다양한 조절 인자에 의해 역동적으로 발생한다. 이 과정에서 새롭게 형성된 혈관은 주위 세포에 영양소, 산소, 호르몬 등의 다양한 생체 재료를 위한 운송 채널의 역할을 한다. 기능 및 구조적으로 비정상적인 혈관은 다양한 질환의 발병 및 진행의 직간접적인 원인이 된다. 종양 혈관은 기능 및 구조 결함으로 저산소증을 악화시켜, 종양의 진행 및 다른 조직으로의 전이를 초래하고, 종양 조직의 중심에 항암제가 잘 전달되지 않도록 한다. 결함이 있는 혈관은 암 이외에도 다른 각종 질병이나 질환에서 확인될 수 있다. 그 예로는 다양한 안과 질환 (예를 들어, 당뇨병성 황반 부종, 노인성 황반변성), 바이러스 감염 및 패혈증 등의 급성 염증 반응을 들 수 있다. 따라서, 병리적 혈관을 정상화할 수 있는 치료제가 존재한다면, 혈관 이상을 가진 다양한 환자의 치료에 적용할 수 있다.Angiogenesis occurs dynamically by a variety of regulatory factors during the development, growth, preservation and maintenance of homeostasis. In this process, newly formed blood vessels serve as transport channels for various biomaterials such as nutrients, oxygen and hormones to surrounding cells. Functional and structurally abnormal blood vessels are a direct and indirect cause of the onset and progression of various diseases. Tumor blood vessels exacerbate hypoxia with functional and structural defects, leading to tumor progression and metastasis to other tissues, and to poor delivery of anticancer agents to the center of the tumor tissue. Defective blood vessels can be identified in various diseases or disorders, in addition to cancer. Examples include acute inflammatory reactions such as various eye diseases (eg diabetic macular edema, senile macular degeneration), viral infections and sepsis. Thus, if a therapeutic agent is available that can normalize pathological vessels, it can be applied to the treatment of various patients with vascular abnormalities.
Angiopoietin 패밀리는 혈관의 형성과 유지에 중요한 역할을 하고 있고, 4 개의 Angiopoietin (Ang1, Ang2, Ang3 및 Ang4)을 포함한다. Angiopoietin-1 (Ang1)은 혈관 내피 세포의 표면에 존재하는 Tie2 수용체에 결합하여 Tie2 수용체를 인산화 및 활성화하고, 그 결과 혈관을 안정화시킨다. 한편, Angiopoietin-2 (Ang2)는 Tie2 수용체에 결합하지만, Tie2 수용체의 불활성화를 유도하는 길항제로 작용하여, 혈관의 불안정화 및 혈관 누출을 초래한다. Ang2의 발현 수준은 암 환자의 혈액, 안과 질환, 바이러스 감염 및 세균 감염 및 염증성 질환에서 매우 증가하는 것으로 보고되었다 (Saharinen P et al., 2017, Nature Review Drug Discovery) . 그러나, Ang2는 림프관 형성 및 유지 등 여러 과정에서 Tie2 수용체의 활성화를 유도하는 작용제로 작용하는 것으로 알려져 있다. 따라서 Ang2는 주변환경이나 조직에 따라 다양한 기능을 수행하는 것으로 판단된다.The Angiopoietin family plays an important role in the formation and maintenance of blood vessels and includes four Angiopoietins (Ang1, Ang2, Ang3 and Ang4). Angiopoietin-1 (Ang1) binds to Tie2 receptors present on the surface of vascular endothelial cells to phosphorylate and activate Tie2 receptors, thereby stabilizing blood vessels. Angiopoietin-2 (Ang2), on the other hand, binds to the Tie2 receptor but acts as an antagonist that induces inactivation of the Tie2 receptor, leading to blood vessel destabilization and vascular leakage. Expression levels of Ang2 have been reported to be very increased in blood, eye disease, viral infections and bacterial infections and inflammatory diseases in cancer patients (Saharinen P et al., 2017, Nature Review Drug Discovery). However, Ang2 is known to act as an agent that induces activation of the Tie2 receptor in many processes, including lymphatic vessel formation and maintenance. Therefore, Ang2 seems to perform various functions depending on the environment and organization.
지금까지 많은 바이오제약사에 의해 다양한 항-Ang2 항체가 개발되고 임상 시험이 수행되어 왔다 (예를 들어, 미국특허 제7,658,924호 및 미국특허 제8,987,420호). 이러한 Ang2 항체는 Tie2에 Ang2의 결합을 저해하는 것으로 나타났고, 이 Ang2 중화 효과는 궁극적으로 혈관신생의 형성을 방해하는 것으로 나타났다. 항-Ang2 항체의 혈관신생 억제 및 항암 작용은 많은 전임상 모델에서 입증되고 있으며, 여러 종류의 항-Ang2 항체가 다양한 암 환자에서 임상 시험되고 있다. 그러나, Ang2 중화작용을 통한 혈관신생 억제는 항암 치료에 충분하지 않다는 사실이 증명되고 있다. 예를 들어, 암젠에 의해 수행된 3상 임상 시험에서 난소암 환자 중 Ang2 항체의 항암 효과가 유의하지 않은 것으로 나타났다 (Marth C et al., 2017, Eur. J. Cancer). 암 모델 뿐 아니라, 안과 환자에서 Ang2 중화 항체 Nesvacumab의 효능이 시험되었으나, 임상 2상 콤보 연구(combo-study)에서 Eylea (항-VEGF)의 효능을 개선시킬 수 없었다.To date, various anti-Ang2 antibodies have been developed and clinical trials have been conducted by many biopharmaceuticals (eg, US Pat. No. 7,658,924 and US Pat. No. 8,987,420). These Ang2 antibodies have been shown to inhibit the binding of Ang2 to Tie2, and this Ang2 neutralizing effect ultimately appears to interfere with the formation of angiogenesis. Angiogenesis inhibition and anticancer activity of anti-Ang2 antibodies has been demonstrated in many preclinical models, and several types of anti-Ang2 antibodies have been clinically tested in various cancer patients. However, it has been demonstrated that inhibition of angiogenesis through Ang2 neutralization is not sufficient for anticancer treatment. For example, in a phase 3 clinical trial conducted by Amgen, the anticancer effect of Ang2 antibodies in ovarian cancer patients was not significant (Marth C et al., 2017, Eur. J. Cancer). In addition to cancer models, the efficacy of the Ang2 neutralizing antibody Nesvacumab was tested in ophthalmic patients, but the efficacy of Eylea (anti-VEGF) could not be improved in the Phase 2 combo-study.
앞서 설명한 Ang2를 중화시키는 접근법과는 대조적으로, 직접 Tie2를 활성화시키는 것이 비정상적인 혈관신생을 억제하고 혈관 투과성을 감소시키기 위한 새로운 접근법으로 고려되고 있다. Tie2 수용체에 직접 결합하여 Tie2의 인산화 및 활성화를 유도하는 재조합 단백질도 개발되고 있고, 다수의 전임상 암 및 안구 모델에서 그 치료효과가 시험되고 있다. COMP-Angl 및 Vasculotide가 그 대표적인 예다. 이들 제제는 항-혈관신생 활성과 항-침투 활성을 나타냈지만, 매우 짧은 반감기 및 불안정한 물리화학적 성질을 가지는 단점이 있다. 또한, 소분자 화합물 (AKB - 9778)이 탈인산화효소 VE-PTP의 저해제로 개발되었다. VE-PTP는 인산화된 Tie2에서 인산기를 제거하여 Tie2을 불활성화시키는 역할을 한다. 이러한 화합물은 다른 수용체도 비특이적으로 활성화시킨다는 단점이 있지만, VE-PTP를 저해함으로써 Tie2 활성을 간접적으로 증가시키는 작용을 한다. 또한, Tie2 활성화 항체가 개발되어 있다 (US6365154B1, US20170174789A1). 이러한 항체는 혈관내피세포의 생존율을 높여 혈관 누출을 억제했다. 흥미롭게도, 식물 추출물중 하나가 Tie2 활성을 유도하는 것으로 나타나, 스킨 케어 화장품으로 사용될 수 있음이 청구되어 있다 (예를 들어, JP2011102273A, JP2018043949A, JP2015168656A).In contrast to the approach of neutralizing Ang2, direct activation of Tie2 is considered a new approach to inhibit abnormal angiogenesis and reduce vascular permeability. Recombinant proteins that directly bind to the Tie2 receptor to induce Tie2 phosphorylation and activation have also been developed, and their therapeutic effects have been tested in many preclinical cancer and ocular models. COMP-Angl and Vasculotide are examples. These agents exhibited anti-angiogenic and anti-invasive activity, but have the disadvantage of having very short half-lives and unstable physicochemical properties. In addition, small molecule compounds (AKB-9778) have been developed as inhibitors of dephosphatase VE-PTP. VE-PTP serves to inactivate Tie2 by removing phosphate groups from phosphorylated Tie2. These compounds have the disadvantage of nonspecifically activating other receptors, but they also indirectly increase Tie2 activity by inhibiting VE-PTP. In addition, Tie2 activating antibodies have been developed (US6365154B1, US20170174789A1). These antibodies inhibited vascular leakage by increasing the survival rate of vascular endothelial cells. Interestingly, it has been claimed that one of the plant extracts induces Tie2 activity and can be used as a skin care cosmetic (eg JP2011102273A, JP2018043949A, JP2015168656A).
이러한 기술적 배경하에서, 본 출원의 발명자들은 Tie2에 특이적으로 결합하는 항체를 개발하기 위하여 노력하였다. 그 결과, 본 발명자들은 Tie2에 높은 친화력으로 결합하는 Tie2 항체를 개발하고, 이러한 Tie2 항체가 Tie2의 인산화 및 Tie2 수용체의 활성화를 유도함으로써, 목적하는 혈관신생 질환 치료제의 역할을 할 수 있음을 확인하고, 본 발명을 완성하였다.Under this technical background, the inventors of the present application sought to develop an antibody that specifically binds Tie2. As a result, the present inventors have developed a Tie2 antibody that binds Tie2 with high affinity, and confirms that the Tie2 antibody can play a role as a therapeutic agent for angiogenic diseases by inducing Tie2 phosphorylation and activation of the Tie2 receptor. The present invention has been completed.
발명의 요약Summary of the Invention
본 발명의 목적은 Tie2에 대한 신규 항체 또는 그의 항원 결합 단편을 제공하는 데 있다.It is an object of the present invention to provide a novel antibody or antigen binding fragment thereof against Tie2.
본 발명의 다른 목적은 상기 항체 또는 그의 항원 결합 단편을 코딩하는 핵산을 제공하는 데 있다.Another object of the present invention is to provide a nucleic acid encoding the antibody or antigen-binding fragment thereof.
본 발명의 다른 목적은 상기 핵산을 포함하는 벡터, 상기 벡터로 형질전환된 세포 및 이의 제조방법을 제공하는 데 있다.Another object of the present invention is to provide a vector comprising the nucleic acid, a cell transformed with the vector, and a method of manufacturing the same.
본 발명의 또 다른 목적은 상기 항체 또는 그의 항원 결합 단편을 포함하는 혈관신생 질환의 예방 또는 치료용 조성물을 제공하는 데 있다.Still another object of the present invention is to provide a composition for preventing or treating angiogenic diseases comprising the antibody or antigen-binding fragment thereof.
본 발명의 또 다른 목적은 상기 항체 또는 그의 항원 결합 단편을 포함하는, 다른 혈관신생 질환 치료제와의 병용투여용 조성물을 제공하는 데 있다.Still another object of the present invention is to provide a composition for co-administration with another therapeutic agent for angiogenic diseases, including the antibody or antigen-binding fragment thereof.
상기 목적을 달성하기 위하여, 본 발명은 서열번호 2의 서열을 포함하는 Tie2 Ig3-FNIII (1-3) 도메인에 결합하는 항-Tie2 항체 또는 이의 항원 결합단편을 제공한다.In order to achieve the above object, the present invention provides an anti-Tie2 antibody or antigen binding fragment thereof that binds to a Tie2 Ig3-FNIII (1-3) domain comprising the sequence of SEQ ID NO: 2.
구체적으로, 본 발명은 또한 서열번호 3 내지 5의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 6 내지 8의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역; 서열번호 13 내지 15의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 16 내지 18의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역; 서열번호 23 내지 25의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 26 내지 28의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역; 서열번호 33 내지 35의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 36 내지 38의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역; 또는 서열번호 43 내지 45의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 46 내지 48의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역을 포함하는, Tie2에 결합하는 항체 또는 이의 항원 결합단편을 제공한다. Specifically, the present invention also provides a heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NO: 3 to 5, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NO: 6 to 8; A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 13 to 15, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 16 to 18; A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 23 to 25, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 26 to 28; A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 33 to 35, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 36 to 38; Or an antibody binding to Tie2 comprising a heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 43 to 45, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NO: 46 to 48, or Its antigen binding fragment is provided.
본 발명은 또한, 상기 항체 또는 그의 항원 결합 단편을 코딩하는 핵산을 제공한다.The present invention also provides a nucleic acid encoding the antibody or antigen-binding fragment thereof.
본 발명은 또한, 상기 핵산을 포함하는 벡터를 제공한다.The present invention also provides a vector comprising the nucleic acid.
본 발명은 또한, 상기 벡터로 형질전환된 세포를 제공한다.The present invention also provides a cell transformed with the vector.
본 발명은 또한, 다음 단계를 포함하는 상기 항체 또는 그의 항원 결합 단편의 제조방법을 제공한다: (a) 상기 세포를 배양하는 단계; 및 (b) 상기 배양된 세포에서 항체 또는 그의 항원 결합 단편을 회수하는 단계.The present invention also provides a method for producing the antibody or antigen-binding fragment thereof comprising the following steps: (a) culturing the cells; And (b) recovering the antibody or antigen-binding fragment thereof from the cultured cells.
본 발명은 또한, 상기 항체 또는 그의 항원 결합 단편을 유효성분으로 포함하는 혈관신생 관련 질환의 예방 또는 치료용 조성물을 제공한다.The present invention also provides a composition for preventing or treating angiogenesis-related diseases comprising the antibody or antigen-binding fragment thereof as an active ingredient.
본 발명은 또한, 상기 항체 또는 그의 항원 결합 단편을 포함하는, 다른 혈관신생 질환 치료제와의 병용투여용 조성물을 제공한다.The present invention also provides a composition for co-administration with another therapeutic agent for angiogenic diseases, comprising the antibody or antigen-binding fragment thereof.
도 1은 항-Tie2 항체에 의해 유도된 Akt 인산화에 대한 분석 결과이다. HUVEC는 6시간 동안 혈청 기아(serum-starved)상태로 유지되었으며 30분 동안 COMP-Ang1(CA1, 0.5 μg/ml) 또는 항-Tie2 항체(11C4, 4A4, 3B2, 3E12 및 3H7)와 함께 배양되었다. 상기 배양 세포의 용해액을 SDS-PAGE/웨스턴 블롯팅 하였고 p-Akt 항체(S473) 또는 Akt 항체로 프로빙한 결과이다.Figure 1 shows the analysis results for Akt phosphorylation induced by anti-Tie2 antibody. HUVECs remained serum-starved for 6 hours and incubated with COMP-Ang1 (CA1, 0.5 μg / ml) or anti-Tie2 antibodies (11C4, 4A4, 3B2, 3E12 and 3H7) for 30 minutes. . Lysates of the cultured cells were subjected to SDS-PAGE / Western blotting and probed with p-Akt antibody (S473) or Akt antibody.
도 2는 항-Tie2 항체 3H7에 의해 유도된 용량-의존적 Tie2 인산화(pTie2)에 대한 분석 결과이다. 3H7 항체의 Tie2 인산화 능력은 면역 침강법(immunoprecipitation) 및 웨스턴 블롯 기법(western blotting analysis)을 통해 분석되었다. 혈청 기아상태의 HUVEC는 여러가지 농도의 3H7과 함께 30분동안 배양되었다. 대조군으로, HUVEC는 Ang2/대조군 항체(Control Ab) 혼합물에서 같은 시간 배양되었다. Tie2 항체를 이용하여 세포 용해액에서 Tie2 단백질을 면역 침강 시킨 후, SDS-PAGE/웨스턴 블롯 분석을 통해 Tie2의 인산화정도를 측정하였다. Tie2 인산화는 마우스에서 유래한 인산 티로신(pY)항체인 4G10에 의해 프로빙되었다.FIG. 2 shows the assay results for dose-dependent Tie2 phosphorylation (pTie2) induced by anti-Tie2 antibody 3H7. The Tie2 phosphorylation capacity of 3H7 antibody was analyzed through immunoprecipitation and western blotting analysis. Serum starved HUVECs were incubated with various concentrations of 3H7 for 30 minutes. As a control, HUVECs were incubated at the same time in Ang2 / Control antibody (Control Ab) mixture. After immunoprecipitation of the Tie2 protein in the cell lysate using the Tie2 antibody, the degree of phosphorylation of Tie2 was measured by SDS-PAGE / Western blot analysis. Tie2 phosphorylation was probed by 4G10, a tyrosine phosphate (pY) antibody derived from mice.
도 3은 항-Tie2 항체 3H7으로 인한 Tie2 수용체의 세포 내 유입(endocytosis) 및 FOXO1의 전좌(translocation)를 나타낸 결과이다. HUVEC는 6시간 동안 혈청 기아 상태에 놓여진 이후 음성대조군(Control), Ang2/대조군 항체 혼합물(A2 + Control Ab) 또는 3H7과 함께 30분간 배양되었다. 세포 고정 후에, 세포 핵은 DAPI 염색으로 파란색, Tie2 수용체는 항-Tie2 항체를 이용하여 녹색, FOXO1은 항-FOXO1 항체를 이용하여 붉은색으로 염색하여 FOXO1 및 클러스터된 Tie2 수용체의 위치를 조사하였다. 도면의 화살표는 세포 내 유입된 Tie2 수용체를 표지한 것이다.FIG. 3 shows the results of translocation of FOXO1 and endocytosis of Tie2 receptor due to anti-Tie2 antibody 3H7. HUVECs were placed in serum starvation for 6 hours and then incubated with negative control (Control), Ang2 / Control antibody mixture (A2 + Control Ab) or 3H7 for 30 minutes. After cell fixation, the cell nuclei were blue with DAPI staining, the Tie2 receptor was green with an anti-Tie2 antibody, and FOXO1 was stained red with an anti-FOXO1 antibody to examine the location of FOXO1 and clustered Tie2 receptors. Arrows in the figure labeled Tie2 receptors introduced into the cell.
도 4는 Tie2 항체인 3H7를 통해 VEGF 또는 TNF-a에 의해 유도된 혈관투과성의 억제를 나타낸 결과이다. HUVEC를 트랜스 웰 챔버(trans well chamber)에 접종하고 3일 동안 배양하였다. 100% 밀집도(confluence) 상태에서, HUVEC를 Ang2(A2, 1μg/ml), Ang2/대조항체 혼합물(A2+Control Ab, 1μg/ml) 또는 3H7(1μg/ml)로 30분 동안 각각 전처리한 후, 상단 챔버에서 45분 동안 VEGF(500ng/ml)로 처리(A)하거나 22시간 동안 TNF-a(100ng/ml)로 처리하였다(B). 그 다음에 상단 챔버에 20분 동안 FITC-dextran을 처리하였고 하단 챔버에 투과된 FITC 형광량을 측정하여 혈관 투과성을 분석하였다. 측정값은 평균 ± 표준편차 이다. 일원배치 분산분석방식(one way ANOVA)에 의할 때 *는 p < 0.05, **는 p < 0.01, ***는 p < 0.001 이다.Figure 4 is a result showing the inhibition of vascular permeability induced by VEGF or TNF-a through the Tie2 antibody 3H7. HUVECs were inoculated into a trans well chamber and incubated for 3 days. At 100% confluence, HUVECs were pretreated with Ang2 (A2, 1 μg / ml), Ang2 / control antibody mixture (A2 + Control Ab, 1 μg / ml) or 3H7 (1 μg / ml) for 30 minutes each. Treatment with VEGF (500 ng / ml) for 45 minutes in the upper chamber (A) or TNF-a (100 ng / ml) for 22 hours (B). The upper chamber was then treated with FITC-dextran for 20 minutes and the vascular permeability was analyzed by measuring the amount of FITC fluorescence transmitted through the lower chamber. The measured value is mean ± standard deviation. According to the one way ANOVA, * is p <0.05, ** is p <0.01, and *** is p <0.001.
도 5는 Tie2 단독 또는 Tie2/3H7 복합체를 수소/중수소(H/D) 교환-질량 분석 실험을 통해 분석한 결과, 중수소 흡수량에서 유의미한 차이 영역을 보이는 히트 맵(Heat map)을 나타낸 것이다. 항-Tie2 항체 3H7의 존재 또는 부존재 하에서 H/D 교환-질량 분석 데이터를 사용하여 0.333, 10, 60 및 240분에서 hTie2 항원에 대한 잔기당 중수소 흡수를 히트 맵으로 나타냈다. 컬러 스케일은 각각의 시간에서 Tie2 단독 및 Tie2/3H7 혼합물 사이의 잔기당 H/D 교환 백분율을 나타낸다. 붉은색은 중수소 교환이 증가한 지역을 나타내고 푸른색은 흡수되지 않았음을 나타낸다.FIG. 5 shows a heat map showing a significant difference region in deuterium uptake as a result of analyzing Tie2 alone or Tie2 / 3H7 complex through hydrogen / deuterium (H / D) exchange-mass spectrometry. H / D exchange-mass spectrometry data in the presence or absence of anti-Tie2 antibody 3H7 was used as heat maps for deuterium uptake per residue for hTie2 antigen at 0.333, 10, 60 and 240 minutes. The color scale represents the percentage of H / D exchange per residue between Tie2 alone and Tie2 / 3H7 mixture at each time. Red color indicates increased deuterium exchange and blue color was not absorbed.
도 6은 Tie2에서 3H7-결합 에피토프를 나타내는 모식도이다. H/D 교환-질량 분석법에 의해 분석된 hTie2 항원에서 항-Tie2 항체(3H7)의 결합-에피토프(빨강)를 PyMol 소프트웨어를 이용해 hTie2 FNIII(1-3) 결정 구조(PDB: 5UTK)로 시각화한 것이다.6 is a schematic showing 3H7-binding epitope in Tie2. Binding-epitope (red) of anti-Tie2 antibody (3H7) in hTie2 antigen analyzed by H / D exchange-mass spectrometry, visualized with hTie2 FNIII (1-3) crystal structure (PDB: 5UTK) using PyMol software will be.
도 7은 인간화 항-Tie2 항체에 의해 유도된 Akt의 인산화(pAkt) 결과를 나타낸 것이다. 혈청 기아상태의 HUVEC를 인간화 항-Tie2 항체와 함께 30분동안 배양하였다. 그 이후 세포 용해액을 SDS-PAGE/웨스턴 블롯 하였고, p-Akt(s473) 항체 또는 항-Akt 항체로 프로빙하였다.Figure 7 shows the results of phosphorylation (pAkt) of Akt induced by humanized anti-Tie2 antibodies. Serum starved HUVECs were incubated with humanized anti-Tie2 antibodies for 30 minutes. Cell lysates were then blotted with SDS-PAGE / Western and probed with p-Akt (s473) antibody or anti-Akt antibody.
도 8은 원발 개방각 녹내장(primary open-angle glaucoma) 마우스 모델에서 인간화된 Tie2 항체(3H7H12G4)에 의한 쉴렘관(SC) 면적의 증가와 안압(IOP)의 감소를 나타낸 결과이다. Angioprotein-1 과 Angioprotein-2의 유도성 결실을 위해, 8주령의 A1:A2 iΔ/Δ 마우스에 Tamoxifen을 투여하였다. 12주령에서 3H7H12G4(한쪽 안구, 5mg/ml용액 1μ주사) 및 Fc(반대쪽 안구, 5mg/ml용액 1μ주사)가 안구 내에 투여되었다. 안압(intraocular pressure, IOP)은 12주령, 13주령, 14주령에 주기적으로 측정되었다. CD144+SC 면적과 CD144+SC에서의 Prox1 및 Tie2의 면역염색 강도는 3H7H12G4의 투여 후 2주뒤에 측정되었다. 기준척도는 100μm이며, 각 그룹당 n = 5이다. 본 도의 값은 평균 ± 표준편차이다. Kruskal-Wallis test에 이은 Tukey`s HSD test with ranks에 의할 때 *는 p < 0.05이다.FIG. 8 shows the increase in Schlemm's canal (SC) area and the decrease in IOP by humanized Tie2 antibody (3H7H12G4) in primary open-angle glaucoma mouse model. Tamoxifen was administered to 8-week-old A1: A2 iΔ / Δ mice for inducible deletion of Angioprotein-1 and Angioprotein-2. At 12 weeks of age, 3H7H12G4 (one eye, 1 mg injection of 5 mg / ml solution) and Fc (opposing eye, 1 μ injection of 5 mg / ml solution) were administered intraocularly. Intraocular pressure (IOP) was measured periodically at 12, 13, and 14 weeks of age. The immunostaining intensity of Prox1 and Tie2 at CD144 + SC area and CD144 + SC was measured 2 weeks after administration of 3H7H12G4. The reference scale is 100 μm, with n = 5 for each group. Values in this figure are mean ± standard deviation. According to the Kruskal-Wallis test followed by Tukey's HSD test with ranks, * is p <0.05.
도 9는 레이저 유도 맥락막 신생 혈관(CNV) 모델에서 3H7H12G4의 유리체강내 주입(intravitreally injected)에 의한 맥락막 혈관 신생(CNV) 및 혈관 누출 억제를 나타낸 결과이다. 레이저 광응고 후 7일차에 항체(5mg/ml 용액, 1μl 주사)가 유리체강내 투여되었다. CD31+ CNV 부피를 측정하였으며, 레이저 광응고 후 6일 및/또는 14일 되는 날에, FA 이미지에서 측정된 고-형광영역의 총합을 ICGA 이미지에서 측정된 CNV 영역의 총합으로 나눈 값으로 CNV 주변의 누출부위를 계산하였다. 각 그룹당 n = 11이며 값은 평균 ± 표준편차이다. one-way ANOVA 에 이은 Student-Newman-Keuls post-test에 의해 *는 p < 0.05, ***는p < 0.001이며; Paired Student’s t-test에 의해 ##는 p < 0.01, ###는 p < 0.001이다.9 shows the results of choroidal neovascularization (CNV) and vascular leakage inhibition by intravitreally injected of 3H7H12G4 in a laser induced choroidal neovascularization (CNV) model. Antibodies (5 mg / ml solution, 1 μl injection) were administered intravitreally on day 7 after laser photocoagulation. CD31 + CNV volume was measured, and on day 6 and / or 14 days after laser photocoagulation, the sum of the high-fluorescence regions measured in the FA image divided by the sum of the CNV regions measured in the ICGA image was around the CNV. The leaked area of was calculated. N = 11 for each group and the value is mean ± standard deviation. one-way ANOVA followed by Student-Newman-Keuls post-test, where * is p <0.05 and *** is p <0.001; According to the Paired Student's t-test, ## is p <0.01 and ### is p <0.001.
도 10은 맥락막 신생 혈관(CNV) 영역의 내피세포에서 3H7H12G4와 CD31의 공동 국소화(co-localization)를 나타내는 형광사진이다. 레이저 광응고 후 1일 뒤에 3H7H12G4가 피하투여 되었다. 레이저 광응고 후 2일, 4일, 8일차에 맥락막 신생 혈관(CNV) 주위의 내피세포에서 3H7H12G4와 CD31의 공동 국소화를 항-인간 IgG 2차 항체를 이용하여 염색하고 분석하였다.FIG. 10 is a fluorescence photograph showing co-localization of 3H7H12G4 and CD31 in endothelial cells in the choroidal neovascularization (CNV) region. One day after laser photocoagulation, 3H7H12G4 was subcutaneously administered. Co-localization of 3H7H12G4 and CD31 in endothelial cells around choroidal neovascularization (CNV) at 2, 4 and 8 days after laser photocoagulation were stained and analyzed using anti-human IgG secondary antibody.
도 11은 3H7H12G4의 피하주사에 의한 맥락막 혈관 신생(CNV)의 억제를 나타낸 것이다. 레이저 광응고 후 1일 뒤에 3H7H12G4를 피하투여하였다. 레이저 광응고 후 8일 뒤 CD31+ CNV량이 측정되었다. 기준척도는 100μm이며, 각 그룹당 n = 10이다. 값은 평균 ± 표준편차이며, unpaired Student’s t-test에 의해 ***는 p < 0.001이다.Figure 11 shows inhibition of choroidal neovascularization (CNV) by subcutaneous injection of 3H7H12G4. One day after laser photocoagulation, 3H7H12G4 was subcutaneously administered. Eight days after laser photocoagulation, the amount of CD31 + CNV was measured. The reference scale is 100 μm, with n = 10 in each group. Values are mean ± standard deviation and *** is p <0.001 by unpaired Student's t-test.
발명의 상세한 설명 및 바람직한 구현예Detailed Description of the Invention and Preferred Embodiments
다른 식으로 정의되지 않는 한, 본 명세서에서 사용된 모든 기술적 및 과학적 용어들은 본 발명이 속하는 기술분야에서 숙련된 전문가에 의해서 통상적으로 이해되는 것과 동일한 의미를 갖는다. 일반적으로, 본 명세서에서 사용된 명명법은 본 기술분야에서 잘 알려져 있고 통상적으로 사용되는 것이다.Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In general, the nomenclature used herein is well known and commonly used in the art.
본 출원의 발명자들은 서열번호 1의 인간 Tie2 전장 서열 중 서열번호 2의 서열을 포함하는 Tie2 Ig3-FNIII(1-3) 도메인에 결합하여, Tie2의 활성을 증가시키는 작용제로 Tie2 항체를 확인하였다. The inventors of the present application have identified a Tie2 antibody as an agent that increases the activity of Tie2 by binding to a Tie2 Ig3-FNIII (1-3) domain comprising the sequence of SEQ ID NO: 2 of the human Tie2 full length sequence of SEQ ID NO: 1.
Tie2는 혈관의 분화와 안정을 촉진하는 수용체 단백질로 암과 같은 질환의 혈관에서 발현이 높으며, Tie2 수용체를 활성화하면 암 혈관을 안정화하면서 주변지지세포들을 결집할 수 있게 된다. 본 발명에 따른 항체 또는 항원 결합단편에 의해, 암 혈관의 Tie2를 활성화하면 암 혈관을 정상화하여 종양 내부의 저산소증 증가를 해소하고 종양 내부로 혈류를 증가시켜 충분한 산소 공급이 이뤄지며 기타 다른 항암 약물 전달량과 면역세포의 침투가 증가하도록 할 수 있다. Tie2 is a receptor protein that promotes the differentiation and stabilization of blood vessels. It is highly expressed in blood vessels of diseases such as cancer, and activating the Tie2 receptor stabilizes cancer blood vessels and aggregates surrounding supporting cells. By the antibody or antigen-binding fragment according to the present invention, activating Tie2 of cancer vessels normalizes the cancer vessels, relieves the increase of hypoxia within the tumors, increases blood flow into the tumors, and provides sufficient oxygen supply. May increase the penetration of immune cells.
이러한 관점에서, 본 발명은 서열번호 2의 서열을 포함하는 Tie2 Ig3-FNIII (1-3) 도메인에 결합하는 Tie2 항체 또는 이의 항원 결합단편에 관한 것이다. In this respect, the present invention relates to a Tie2 antibody or antigen binding fragment thereof that binds to a Tie2 Ig3-FNIII (1-3) domain comprising the sequence of SEQ ID NO: 2.
본 명세서에서 사용된 용어, "항체(antibody)"는 Tie2에 특이적으로 결합하는 항체를 의미한다. 본 발명의 범위에는 Tie2에 특이적으로 결합하는 완전한 항체 형태 뿐 아니라, 상기 항체 분자의 항원 결합 단편도 포함된다.As used herein, the term "antibody" refers to an antibody that specifically binds to Tie2. The scope of the present invention includes not only complete antibody forms that specifically bind Tie2, but also antigen binding fragments of such antibody molecules.
완전한 항체는 2개의 전체 길이의 경쇄 및 2개의 전체 길이의 중쇄를 가지는 구조이며 각각의 경쇄는 중쇄와 다이설파이드 결합으로 연결되어 있다. 중쇄 불변영역은 감마(γ), 뮤(μ), 알파(α), 델타(δ) 및 엡실론(ε) 타입을 가지고 서브클래스로 감마1(γ1), 감마2(γ2), 감마3(γ3), 감마4(γ4), 알파1(α1) 및 알파2(α2)를 가진다. 경쇄의 불변영역은 카파(κ) 및 람다(λ) 타입을 가진다.A complete antibody is a structure having two full length light chains and two full length heavy chains, each of which is linked by heavy and disulfide bonds. The heavy chain constant region has gamma (γ), mu (μ), alpha (α), delta (δ) and epsilon (ε) types and subclasses gamma 1 (γ1), gamma 2 (γ2), and gamma 3 (γ3). ), Gamma 4 (γ4), alpha 1 (α1) and alpha 2 (α2). The constant regions of the light chains have kappa (κ) and lambda (λ) types.
항체의 항원 결합 단편 또는 항체 단편이란 항원 결합 기능을 보유하고 있는 단편을 의미하며, Fab, F(ab'), F(ab')2 및 Fv 등을 포함한다. 항체 단편 중 Fab는 경쇄 및 중쇄의 가변영역과 경쇄의 불변영역 및 중쇄의 첫 번째 불변영역(CH1)을 가지는 구조로 1개의 항원 결합 부위를 가진다. Fab'는 중쇄 CH1 도메인의 C-말단에 하나 이상의 시스테인 잔기를 포함하는 힌지 영역(hingeAn antigen binding fragment or antibody fragment of an antibody means a fragment having an antigen binding function and includes Fab, F (ab '), F (ab') 2, Fv and the like. Fab in the antibody fragment has a structure having a variable region of the light and heavy chains, a constant region of the light chain and the first constant region (CH1) of the heavy chain has one antigen binding site. Fab ′ is a hinge region comprising one or more cysteine residues at the C-terminus of the heavy chain CH1 domain
region)을 가진다는 점에서 Fab와 차이가 있다. F(ab')2 항체는 Fab'의 힌지 영역의 시스테인 잔기가 디설파이드 결합을 이루면서 생성된다. Fv는 중쇄 가변영역 및 경쇄 가변영역만을 가지고 있는 최소의 항체조각이다. 이중쇄 Fv(two-chain Fv)는 비공유 결합으로 중쇄 가변영역과 경쇄 가변영역이 연결되어 있고 단쇄 Fv(single-chain Fv, scFv)는 일반적으로 펩타이드 링커를 통하여 중쇄의 가변영역과 경쇄의 가변영역이 공유결합으로 연결되거나 또는 C-말단에서 바로 연결되어 있어서 이중쇄 Fv와 같이 다이머와 같은 구조를 이룰 수 있다. 이러한 항체 단편은 단백질 가수분해 효소를 이용해서 얻을 수 있고(예를 들어, 전체 항체를 파파인으로 제한 절단하면 Fab를 얻을 수 있고 펩신으로 절단하면 F(ab')2 단편을 얻을 수 있다), 유전자 재조합 기술을 통하여 제작할 수도 있다.It differs from Fab in that it has a region. F (ab ') 2 antibodies are produced by disulfide bonds of cysteine residues in the hinge region of Fab'. Fv is the minimum antibody fragment which has only a heavy chain light region and a light chain variable region. Double-chain Fv is a non-covalent bond in which a heavy chain variable region and a light chain variable region are linked, and a single chain Fv (single-chain Fv, scFv) is generally a variable region of the heavy chain and the light chain through a peptide linker. This covalent linkage or the C-terminus is directly linked to form a dimer-like structure such as a double-chain Fv. Such antibody fragments can be obtained using proteolytic enzymes (e.g., restriction digestion of the entire antibody with papain yields Fab and cleavage with pepsin yields F (ab ') 2 fragments). It can also be produced by recombinant technology.
하나의 실시예에서, 본 발명에 따른 항체는 Fv 형태(예컨대, scFv)이거나, 완전한 항체 형태이다. 또한, 중쇄 불변영역은 감마(γ), 뮤(μ), 알파(α), 델타(δ) 또는 엡실론(ε) 중의 어느 한 이소타입으로부터 선택될 수 있다. 예를 들어, 불변영역은 감마1(IgG1), 감마 3(IgG3) 또는 감마 4(IgG4)이다. 경쇄 불변영역은 카파 또는 람다 형일 수 있다.In one embodiment, the antibody according to the invention is in Fv form (eg scFv) or is in the form of a complete antibody. In addition, the heavy chain constant region may be selected from any one isotype of gamma (γ), mu (μ), alpha (α), delta (δ) or epsilon (ε). For example, the constant region is gamma 1 (IgG1), gamma 3 (IgG3) or gamma 4 (IgG4). The light chain constant region may be of kappa or lambda type.
본 명세서에서 사용되는 용어, "중쇄"는 항원에 특이성을 부여하기 위한 충분한 가변영역 서열을 갖는 아미노산 서열을 포함하는 가변영역 도메인 VH 및 3 개의 불변영역 도메인 CH1, CH2 및 CH3을 포함하는 전체길이 중쇄 및 이의 단편을 모두 의미한다. 또한, 본 명세서에서 사용되는 용어, "경쇄"는 항원에 특이성을 부여하기 위한 충분한 가변영역 서열을 갖는 아미노산 서열을 포함하는 가변영역 도메인 VL 및 불변영역 도메인 CL을 포함하는 전체길이 경쇄 및 이의 단편을 모두 의미한다.As used herein, the term “heavy chain” refers to a variable region domain VH comprising an amino acid sequence having sufficient variable region sequence to confer specificity to an antigen and a full length heavy chain comprising three constant region domains CH1, CH2 and CH3 And fragments thereof. In addition, the term "light chain" as used herein refers to a full-length light chain and fragment thereof comprising a variable region domain VL and a constant region domain CL comprising an amino acid sequence having sufficient variable region sequence to confer specificity to the antigen. All means.
본 발명의 항체는 단일클론 항체, 다특이적 항체, 인간 항체, 인간화 항체, 키메라 항체, 단쇄 Fvs(scFV), 단쇄 항체, Fab 단편, F(ab') 단편, 다이설파이드-결합 Fvs(sdFV) 및 항-이디오타입(항-Id) 항체, 또는 상기 항체들의 에피토프-결합 단편 등을 포함하나, 이에 한정되는 것은 아니다.Antibodies of the invention include monoclonal antibodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, single chain Fvs (scFV), single chain antibodies, Fab fragments, F (ab ') fragments, disulfide-binding Fvs (sdFV) And anti-idiotype (anti-Id) antibodies, or epitope-binding fragments of the antibodies, and the like.
상기 단일클론 항체는 실질적으로 동질적 항체 집단으로부터 수득한 항체, 즉 집단을 차지하고 있는 개개의 항체가 미량으로 존재할 수 있는 가능한 천연 발생적 돌연변이를 제외하고는 동일한 것을 지칭한다. 단일클론 항체는 고도로 특이적이어서, 단일 항원 부위에 대항하여 유도된다. 전형적으로 상이한 결정인자(에피토프)에 대해 지시된 상이한 항체를 포함하는 통상의 (폴리클로날) 항체 제제와는 대조적으로, 각각의 모노클로날 항체는 항원 상의 단일 결정인자에 대해 지시된다.Said monoclonal antibody refers to the same except for possible naturally occurring mutations in which antibodies obtained from substantially homogeneous antibody populations, ie, individual antibodies in the population, may be present in trace amounts. Monoclonal antibodies are highly specific and are directed against a single antigenic site. In contrast to conventional (polyclonal) antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
"에피토프"은 항체가 특이적으로 결합할 수 있는 단백질 결정부위 (determinant)를 의미한다. 에피토프는 통상 화학적으로 활성인 표면 분자군, 예를 들어 아미노산 또는 당 측쇄로 구성되며, 일반적으로 특정한 3차원의 구조적 특징뿐만 아니라 특정한 전하 특성을 갖는다. 입체적 에피토프 및 비입체적 에피토프는 변성 용매의 존재하에서 전자에 대한 결합은 소실되지만 후자에 대해서는 소실되지 않는다는 점에서 구별된다. "Epitope" refers to a protein determinant to which an antibody can specifically bind. Epitopes usually consist of a group of chemically active surface molecules, such as amino acids or sugar side chains, and generally have specific three dimensional structural characteristics as well as specific charge characteristics. Three-dimensional epitopes and non-stereo epitopes are distinguished in that the binding to the former is lost but not to the latter in the presence of a denatured solvent.
본 발명에 따른 Tie2 항체 또는 이의 항원 결합단편은 수소/중수소 교환반응(hydrogen/deuterium exchange)을 통해 에피토프를 확인하였을 때, 서열번호 1의 서열을 포함하는 Tie2의 633 내지 644번째 아미노산 TLSDILPPQPEN 및/또는 713 내지 726번째 아미노산 FAENNIGSSNPAFS에 결합함을 확인하였다. Tie2 antibody or antigen-binding fragment thereof according to the present invention, when identifying epitopes through hydrogen / deuterium exchange, TLSDILPPQPEN and / or amino acids 633 to 644 of Tie2 comprising the sequence of SEQ ID NO: 1 Binding to amino acids 713-726 of FAENNIGSSNPAFS.
상기 "인간화" 형태의 비-인간 (예: 뮤린) 항체는 비-인간 면역글로불린으로부터 유래된 최소 서열을 함유하는 하나 이상의 비-인간 항체(공여체 또는 공급원 항체)로부터 유래된 하나 이상의 아미노산 서열(예를 들어, CDR 서열)을 포함하는 키메라 항체이다. 대부분의 경우, 인간화 항체는, 수용자의 초가변 영역으로부터의 잔기를 목적하는 특이성, 친화성 및 능력을 보유하고 있는 비-인간 종 (공여자 항체), 예를 들어 마우스, 랫트, 토끼 또는 비-인간 영장류의 초가변 영역로부터의 잔기로 대체시킨 인간 면역글로불린 (수용자 항체)이다. 인간화를 위해 수용자 인간 항체의 가변 영역의 하나 이상의 프레임워크 도메인 (framework domain, FR) 내의 잔기가 비-인간 종 공여체 항체부터 상응하는 잔기로 대체될 수 있다. 이를 통해 그래프팅된 CDR(들)의 적절한 3차원 구성을 유지하도록 돕고, 이로써 친화성 및 항체 안정성을 개선시킬 수 있다. 인간화된 항체는, 예를 들어, 항체 성능을 추가로 세밀화하기 위해, 추가의 수용자 항체 또는 공여체 항체에서 나타나지 않는 새로운 잔기를 포함할 수 있다. The non-human (eg murine) antibody of the “humanized” form may comprise one or more amino acid sequences derived from one or more non-human antibodies (donor or source antibody) containing a minimal sequence derived from a non-human immunoglobulin (eg Eg, a CDR sequence). In most cases, humanized antibodies are non-human species (donor antibodies) that retain the desired specificity, affinity, and capacity for residues from the hypervariable region of the recipient, for example mice, rats, rabbits, or non-humans. Human immunoglobulins (receptor antibodies) replaced with residues from the hypervariable regions of primates. For humanization, residues in one or more framework domains (FR) of the variable region of the recipient human antibody may be replaced with corresponding residues from the non-human species donor antibody. This helps to maintain proper three-dimensional organization of the grafted CDR (s), thereby improving affinity and antibody stability. Humanized antibodies may include new residues that do not appear in additional recipient antibodies or donor antibodies, for example to further refine antibody performance.
상기 “인간 항체”는 인간 면역글로불린으로부터 유래하는 분자로서 상보성 결정영역, 구조 영역을 포함한 항체를 구성하는 모든 아미노산 서열 전체가 인간의 면역글로불린으로 구성되어 있는 것을 의미한다.The term “human antibody” refers to a molecule derived from human immunoglobulin, in which all amino acid sequences constituting the antibody including complementarity determining regions and structural regions are composed of human immunoglobulins.
중쇄 및/또는 경쇄 일부가 특별한 종으로부터 유래되거나 특별한 항체 부류 또는 아부류에 속하는 항체 내의 상응하는 서열과 동일하거나 이와 상동성인 반면, 나머지 쇄(들)는 또 다른 종으로부터 유래되거나 또 다른 항체 부류 또는 아부류에 속하는 항체 내의 상응하는 서열과 동일하거나 이와 상동성인 "키메라" 항체 (면역글로불린) 뿐 아니라 목적하는 생물학적 활성을 나타내는 상기 항체의 단편이 포함된다.While the heavy and / or light chain portions are the same or homologous to the corresponding sequences in an antibody derived from a particular species or belonging to a particular antibody class or subclass, the remaining chain (s) are derived from another species or another antibody class or Included are "chimeric" antibodies (immunoglobulins) that are identical or homologous to the corresponding sequences in antibodies belonging to the subclass, as well as fragments of such antibodies that exhibit the desired biological activity.
본 출원의 구체적 실시예에 따르면, 마우스 유래 Tie2 항체로 11C4, 4A4, 3B2, 3E12 및 3H7를 제작하였으며, 공급원 항체인 마우스 유래 3H7 항체에서 CDR 그래프트되고 인간화한 항체로 3H7H11G4, 3H7H12G4, 3H7H21G4 또는 3H7H22G4를 제작하였다. According to a specific example of the present application, 11C4, 4A4, 3B2, 3E12, and 3H7 were prepared using a mouse-derived Tie2 antibody, and 3H7H11G4, 3H7H12G4, 3H7H21G4 or 3H7H22G4 were CDR-grafted and humanized antibodies from a mouse-derived 3H7 antibody as a source antibody. Produced.
본원에 사용된 바와 같은 "항체 가변 도메인"은 상보성 결정 영역 (CDR; 즉, CDR1, CDR2, 및 CDR3), 및 골격 영역 (FR)의 아미노산 서열을 포함하는 항체 분자의 경쇄 및 중쇄 부분을 지칭한다. VH는 중쇄의 가변 도메인을 지칭한다. VL은 경쇄의 가변 도메인을 지칭한다.As used herein, “antibody variable domain” refers to the light and heavy chain portions of an antibody molecule comprising the amino acid sequences of complementarity determining regions (CDRs; ie CDR1, CDR2, and CDR3), and framework regions (FR). . VH refers to the variable domain of the heavy chain. VL refers to the variable domain of the light chain.
"상보성 결정 영역” (CDR; 즉, CDR1, CDR2, 및 CDR3)은 항원 결합을 위해 필요한 존재인, 항체 가변 도메인의 아미노산 잔기를 지칭한다. 각 가변 도메인은 전형적으로, CDR1, CDR2 및 CDR3으로서 확인된 3개의 CDR 영역을 갖는다. “Complementarity Determining Regions” (CDRs; ie CDR1, CDR2, and CDR3) refer to amino acid residues of antibody variable domains that are required for antigen binding, each variable domain typically identified as CDR1, CDR2 and CDR3. Three CDR regions.
하나의 실시예에서, 상기 Tie2 항체 또는 이의 항원 결합단편은 서열번호 3 내지 5의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 6 내지 8의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역; In one embodiment, the Tie2 antibody or antigen-binding fragment thereof comprises a heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 3 to 5, a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 6 to 8 Light chain variable region;
서열번호 13 내지 15의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 16 내지 18의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역;A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 13 to 15, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 16 to 18;
서열번호 23 내지 25의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 26 내지 28의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역;A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 23 to 25, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 26 to 28;
서열번호 33 내지 35의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 36 내지 38의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역; 또는A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 33 to 35, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 36 to 38; or
서열번호 43 내지 45의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 46 내지 48의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역을 포함할 수 있다. A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NO: 43 to 45, and a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NO: 46 to 48.
"골격 영역" (FR)은 CDR 잔기 이외의 가변 도메인 잔기이다. 각 가변 도메인은 전형적으로, FR1, FR2, FR3 및 FR4로서 확인된 4개의 FR을 가진다."Framework regions" (FRs) are variable domain residues other than CDR residues. Each variable domain typically has four FRs identified as FR1, FR2, FR3 and FR4.
Tie2 항체는 1가 또는 2가이고, 단쇄 또는 이중 쇄를 포함한다. 기능적으로, Tie2 항체의 결합 친화성은 10-5M 내지 10-12 M 범위 내에 있다. 예를 들어, Tie2 항체의 결합 친화성은 10-6 M 내지 10-12 M, 10-7 M 내지 10-12 M, 10-8 M 내지 10-12 M, 10-9 M 내지 10-12 M, 10-5 M 내지 10-11 M, 10-6 M 내지 10-11 M, 10-7 M 내지 10-11 M, 10-8 M 내지 10-11 M, 10-9 M 내지 10-11 M, 10-10 M 내지 10-11 M, 10-5 M 내지 10-10 M, 10-6 M 내지 10-10 M, 10-7 M 내지 10-10 M, 10-8 M 내지 10-10 M, 10-9 M 내지 10-10 M, 10-5 M 내지 10-9 M, 10-6 M 내지 10-9 M, 10-7 M 내지 10-9 M, 10-8 M 내지 10-9 M, 10-5 M 내지 10-8 M, 10-6 M 내지 10-8 M, 10-7 M 내지 10-8 M, 10-5 M 내지 10-7 M, 10-6 M 내지 10-7 M 또는 10-5 M 내지 10-6 M이다.Tie2 antibodies are monovalent or bivalent and include single or double chains. Functionally, the binding affinity of Tie2 antibodies is in the range of 10-5M to 10-12M. For example, the binding affinity of a Tie2 antibody is 10 −6 M to 10 −12 M, 10 −7 M to 10 −12 M, 10 −8 M to 10 −12 M, 10 −9 M to 10 −12 M, 10 -5 M to 10 -11 M, 10 -6 M to 10 -11 M, 10 -7 M to 10 -11 M, 10 -8 M to 10 -11 M, 10 -9 M to 10 -11 M, 10 -10 M to 10 -11 M, 10 -5 M to 10 -10 M, 10 -6 M to 10 -10 M, 10 -7 M to 10 -10 M, 10 -8 M to 10 -10 M, 10 -9 M to 10 -10 M, 10 -5 M to 10 -9 M, 10 -6 M to 10 -9 M, 10 -7 M to 10 -9 M, 10 -8 M to 10 -9 M, 10 -5 M to 10 -8 M, 10 -6 M to 10 -8 M, 10 -7 M to 10 -8 M, 10 -5 M to 10 -7 M, 10 -6 M to 10 -7 M or 10 -5 M to 10 -6 M.
상기 Tie2에 결합하는 항체 또는 이의 항원 결합단편은 서열번호 9의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 11의 아미노산 서열을 포함하는 중쇄 가변영역;The antibody or antigen-binding fragment thereof that binds Tie2 may comprise a heavy chain variable region comprising an amino acid sequence of SEQ ID NO: 9 and a heavy chain variable region comprising an amino acid sequence of SEQ ID NO: 11;
서열번호 19의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 21의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 19 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 21;
서열번호 29의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 31의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 29 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 31;
서열번호 39의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 41의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 39 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 41;
서열번호 49의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 51의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 49 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 51;
서열번호 53의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 54의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 53 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 54;
서열번호 57의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 58의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 57 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 58;
서열번호 61의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 62의 아미노산 서열을 포함하는 중쇄 가변영역; 또는A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 61 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 62; or
서열번호 65의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 66의 아미노산 서열을 포함하는 중쇄 가변영역을 포함할 수 있다. A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 65 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 66.
본 발명의 항체 또는 항체 단편은 Tie2을 특이적으로 인식할 수 있는 범위 내에서, 본 명세서에 기재된 본 발명의 항-Tie2 항체의 서열뿐만 아니라, 이의 생물학적 균등물도 포함할 수 있다. 예를 들면, 항체의 결합 친화도 및/또는 기타 생물학적 특성을 보다 더 개선시키기 위하여 항체의 아미노산 서열에 추가적인 변화를 줄 수 있다. 이러한 변형은 예를 들어, 항체의 아미노산 서열 잔기의 결실, 삽입 및/또는 치환을 포함한다. 이러한 아미노산 변이는 아미노산 곁사슬 치환체의 상대적 유사성, 예컨대, 소수성, 친수성, 전하, 크기 등에 기초하여 이루어진다. 아미노산 곁사슬 치환체의 크기, 모양 및 종류에 대한 분석에 의하여, 아르기닌, 라이신과 히스티딘은 모두 양전하를 띤 잔기이고; 알라닌, 글라이신과 세린은 유사한 크기를 가지며; 페닐알라닌, 트립토판과 타이로신은 유사한 모양을 갖는다는 것을 알 수 있다. 따라서, 이러한 고려 사항에 기초하여, 아르기닌, 라이신과 히스티딘; 알라닌, 글라이신과 세린; 그리고 페닐알라닌, 트립토판과 타이로신은 생물학적으로 기능 균등물이라 할 수 있다.Antibodies or antibody fragments of the present invention may include not only the sequences of the anti-Tie2 antibodies of the present invention described herein, but also biological equivalents thereof, as long as they can specifically recognize Tie2. For example, further changes can be made to the amino acid sequence of the antibody to further improve the binding affinity and / or other biological properties of the antibody. Such modifications include, for example, deletions, insertions and / or substitutions of amino acid sequence residues of the antibody. Such amino acid variations are made based on the relative similarity of amino acid side chain substituents such as hydrophobicity, hydrophilicity, charge, size, and the like. By analysis of the size, shape and type of amino acid side chain substituents, arginine, lysine and histidine are all positively charged residues; Alanine, glycine and serine have similar sizes; It can be seen that phenylalanine, tryptophan and tyrosine have a similar shape. Thus, based on these considerations, arginine, lysine and histidine; Alanine, glycine and serine; Phenylalanine, tryptophan and tyrosine are biologically equivalent functions.
상술한 생물학적 균등 활성을 갖는 변이를 고려한다면, 본 발명의 항체가 가지는 아미노산 서열 또는 이를 코딩하는 핵산 분자는 서열번호에 기재된 서열과 실질적인 동일성(substantial identity)을 나타내는 서열도 포함하는 것으로 해석된다. 상기의 실질적인 동일성은, 상기한 본 발명의 서열과 임의의 다른 서열을 최대한 대응되도록 얼라인하고, 당업계에서 통상적으로 이용되는 알고리즘을 이용하여 얼라인된 서열을 분석한 경우에, 최소 90%의 상동성, 가장 바람직하게는 최소 95%의 상동성, 96% 이상, 97% 이상, 98% 이상, 99% 이상의 상동성을 나타내는 서열을 의미한다. 서열비교를 위한 얼라인먼트 방법은 당업계에 공지되어 있다. NCBI Basic Local Alignment Search Tool(BLAST)은 NBCI 등에서 접근 가능하며, 인터넷 상에서 blastp, blasm, blastx, tblastn 및 tblastx와 같은 서열 분석 프로그램과 연동되어 이용할 수 있다. BLSAT는 www.ncbi.nlm.nih.gov/BLAST/에서 접속 가능하다. 이 프로그램을 이용한 서열 상동성 비교 방법은 www.ncbi.nlm.nih.gov/BLAST/blast_ help.html에서 확인할 수 있다.Considering the above-described variations having biologically equivalent activity, the amino acid sequence possessed by the antibody of the present invention or the nucleic acid molecule encoding the same is interpreted to include a sequence showing substantial identity with the sequence described in SEQ ID NO. The above substantial identity is at least 90% when the sequences of the present invention are aligned as closely as possible with any other sequences, and the aligned sequences are analyzed using algorithms commonly used in the art. By a homology, most preferably at least 95% homology, at least 96%, at least 97%, at least 98%, at least 99% homology. Alignment methods for sequence comparison are known in the art. The NCBI Basic Local Alignment Search Tool (BLAST) is accessible from NBCI and the like and can be used in conjunction with sequence analysis programs such as blastp, blasm, blastx, tblastn and tblastx on the Internet. BLSAT is accessible at www.ncbi.nlm.nih.gov/BLAST/. Sequence homology comparisons using this program can be found at www.ncbi.nlm.nih.gov/BLAST/blast_help.html.
이에 기초하여, 본 발명의 항체 또는 그의 항원 결합 단편은 명세서에 기재된 명시된 서열 또는 전체와 비교하여 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 또는 그 이상의 상동성을 가질 수 있다. 이러한 상동성은 당업계에 공지된 방법에 의한 서열 비교 및/또는 정렬에 의해 결정될 수 있다. 예를 들어, 서열 비교 알고리즘(즉, BLAST 또는 BLAST 2.0), 수동 정렬, 육안 검사를 이용하여 본 발명의 핵산 또는 단백질의 퍼센트 서열 상동성을 결정할 수 있다. Based on this, the antibody or antigen-binding fragment thereof of the present invention is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% compared to the specified sequence or all described in the specification. , 99%, or more homology. Such homology can be determined by sequence comparison and / or alignment by methods known in the art. For example, sequence comparison algorithms (ie, BLAST or BLAST 2.0), manual alignment, visual inspection can be used to determine the percent sequence homology of nucleic acids or proteins of the invention.
본 발명은 다른 관점에서, 상기 항체 또는 그의 항원 결합 단편을 코딩하는 핵산에 관한 것이다. In another aspect, the present invention relates to a nucleic acid encoding the antibody or antigen-binding fragment thereof.
상기 핵산은 서열번호 10, 12, 20, 22, 30, 32, 40, 42, 50, 52, 55, 56, 59, 60, 63, 64, 67 또는 68의 서열을 포함할 수 있다. The nucleic acid may comprise the sequence of SEQ ID NO: 10, 12, 20, 22, 30, 32, 40, 42, 50, 52, 55, 56, 59, 60, 63, 64, 67 or 68.
본 발명의 항체 또는 그의 항원 결합 단편을 코딩하는 핵산을 분리하여 항체 또는 그의 항원 결합 단편을 재조합적으로 생산할 수 있다. 핵산을 분리하고, 이를 복제 가능한 벡터 내로 삽입하여 추가로 클로닝하거나 (DNA의 증폭) 또는 추가로 발현시킨다. 이를 바탕으로, 본 발명은 또 다른 관점에서 상기 핵산을 포함하는 벡터에 관한 것이다. The nucleic acid encoding the antibody or antigen-binding fragment thereof of the present invention can be isolated to recombinantly produce the antibody or antigen-binding fragment thereof. The nucleic acid is isolated and inserted into a replicable vector for further cloning (amplification of DNA) or for further expression. Based on this, the present invention relates to a vector comprising the nucleic acid in another aspect.
"핵산"는 DNA(gDNA 및 cDNA) 및 RNA 분자를 포괄적으로 포함하는 의미를 가지며, 핵산에서 기본 구성단위인 뉴클레오타이드는 자연의 뉴클레오타이드 뿐만 아니라, 당 또는 염기 부위가 변형된 유사체(analogue)도 포함한다. 본 발명의 중쇄 및 경쇄 가변영역을 코딩하는 핵산의 서열은 변형될 수 있다. 상기 변형은 뉴클레오타이드의 추가, 결실, 또는 비보존적 치환 또는 보존적 치환을 포함한다."Nucleic acid" is meant to encompass DNA (gDNA and cDNA) and RNA molecules inclusively, and the nucleotides that are the basic building blocks of nucleic acids include natural nucleotides as well as analogs with modified sugar or base sites. . The sequences of nucleic acids encoding heavy and light chain variable regions of the invention can be modified. Such modifications include addition, deletion, or non-conservative or conservative substitutions of nucleotides.
상기 항체를 암호화하는 DNA는 통상적인 과정을 사용하여 (예를 들어, 항체의 중쇄와 경쇄를 암호화하는 DNA와 특이적으로 결합할 수 있는 올리고뉴클레오티드 프로브를 사용함으로써) 용이하게 분리 또는 합성한다. 많은 벡터가 입수 가능하다. 벡터 성분에는 일반적으로, 다음 중의 하나 이상이 포함되지만, 그에 제한되지 않는다: 신호 서열, 복제 기점, 하나 이상의 마커 유 전자, 증강인자 요소, 프로모터, 및 전사 종결 서열.The DNA encoding the antibody is readily isolated or synthesized using conventional procedures (e.g., by using oligonucleotide probes capable of specifically binding to the DNA encoding the heavy and light chains of the antibody). Many vectors are available. Vector components generally include, but are not limited to, one or more of the following: signal sequence, origin of replication, one or more marker genes, enhancer elements, promoters, and transcription termination sequences.
본 명세서에서 사용되는 용어, "벡터"는 숙주세포에서 목적 유전자를 발현시키기 위한 수단으로 플라스미드 벡터; 코즈미드 벡터; 박테리오파지 벡터, 아데노바이러스 벡터, 레트로바이러스 벡터 및 아데노-연관 바이러스벡터 같은 바이러스 벡터 등을 포함한다. 상기 벡터에서 항체를 코딩하는 핵산은 프로모터와 작동적으로 연결되어 있다. As used herein, the term "vector" refers to a plasmid vector as a means for expressing a gene of interest in a host cell; Cosmid vector; Viral vectors such as bacteriophage vectors, adenovirus vectors, retrovirus vectors, and adeno-associated virus vectors, and the like. The nucleic acid encoding the antibody in the vector is operably linked with a promoter.
“작동적으로 연결”은 핵산 발현조절서열(예: 프로모터, 시그널 서열, 또는 전사조절인자 결합 위치의 어레이)과 다른 핵산 서열사이의 기능적인 결합을 의미하며, 이에 의해 상기 조절서열은 상기 다른 핵산 서열의 전사 및/또는 해독을 조절하게 된다.“Operatively linked” means a functional binding between a nucleic acid expression control sequence (eg, an array of promoters, signal sequences, or transcriptional regulator binding sites) and another nucleic acid sequence, whereby the regulatory sequence is the other nucleic acid. To control transcription and / or translation of the sequence.
원핵세포를 숙주로 하는 경우에는, 전사를 진행시킬 수 있는 강력한 프로모터(예컨대, tac 프로모터, lac 프로모터, lacUV5 프로모터, lpp 프로모터, pLλ 프로모터, pRλ프로모터, rac5 프로모터, amp 프로모터, recA 프로모터, SP6 프로모터, trp 프로모터 및 T7 프로모터 등), 해독의 개시를 위한 라이보좀 결합 자리 및 전사/해독 종결 서열을 포함하는 것이 일반적이다. 또한, 예를 들어, 진핵 세포를 숙주로 하는 경우에는, 포유동물 세포의 지놈으로부터 유래된 프로모터(예: 메탈로티오닌 프로모터, β-액틴 프로모터, 사람 헤로글로빈 프로모터 및 사람 근육 크레아틴 프로모터) 또는 포유동물 바이러스로부터 유래된 프로모터(예: 아데노바이러스 후기 프로모터, 백시니아 바이러스 7.5K 프로모터, SV40프로모터, 사이토메갈로바이러스(CMV) 프로모터, HSV의 tk 프로모터, 마우스 유방종양 바이러스(MMTV) 프로모터, HIV의 LTR 프로모터, 몰로니 바이러스의 프로모터엡스타인바 바이러스(EBV)의 프로모터 및 로우스 사코마 바이러스(RSV)의 프로모터)가 이용될 수 있으며, 전사 종결 서열로서 폴리아데닐화 서열을 일반적으로 갖는다.In the case of a prokaryotic cell as a host, powerful promoters capable of promoting transcription (e.g., tac promoter, lac promoter, lacUV5 promoter, lpp promoter, pLλ promoter, pRλ promoter, rac5 promoter, amp promoter, recA promoter, SP6 promoter, trp promoter and T7 promoter, etc.), ribosome binding sites for initiation of translation, and transcription / detox termination sequences. Also, for example, when the eukaryotic cell is a host, a promoter derived from the genome of the mammalian cell (e.g., a metallothionine promoter, a β-actin promoter, a human heroglobin promoter and a human muscle creatine promoter) or a mammal Promoters derived from animal viruses (e.g., adenovirus late promoter, vaccinia virus 7.5K promoter, SV40 promoter, cytomegalovirus (CMV) promoter, tk promoter of HSV, mouse breast tumor virus (MMTV) promoter, LTR promoter of HIV) , Promoter of the Moroni virus Epsteinbar virus (EBV) and Loose Sacoma virus (RSV) promoter) can be used, and generally has a polyadenylation sequence as a transcription termination sequence.
경우에 따라서, 벡터는 그로부터 발현되는 항체의 정제를 용이하게 하기 위하여 다른 서열과 융합될 수도 있다. 융합되는 서열은, 예컨대 글루타티온 S-트랜스퍼라제(Pharmacia, USA), 말토스 결합 단백질(NEB, USA), FLAG(IBI, USA) 및 6x His(hexahistidine; Quiagen, USA) 등이 있다.In some cases, the vector may be fused with other sequences to facilitate purification of the antibody expressed therefrom. Sequences to be fused include, for example, glutathione S-transferase (Pharmacia, USA), maltose binding protein (NEB, USA), FLAG (IBI, USA) and 6x His (hexahistidine; Quiagen, USA).
상기 벡터는 선택표지로서 당업계에서 통상적으로 이용되는 항생제 내성 유전자를 포함하며, 예를 들어 암피실린, 겐타마이신, 카베니실린, 클로람페니콜, 스트렙토마이신, 카나마이신, 게네티신, 네오마이신 및 테트라사이클린에 대한 내성 유전자가 있다.Such vectors include antibiotic resistance genes commonly used in the art as selectable markers and include, for example, ampicillin, gentamicin, carbenicillin, chloramphenicol, streptomycin, kanamycin, geneticin, neomycin and tetracycline. There is a resistance gene.
본 발명은 또 다른 관점에서, 상기 언급된 벡터로 형질전환된 세포에 관한 것이다. 본 발명의 항체를 생성시키기 위해 사용된 세포는 원핵생물, 효모 또는 고등 진핵생물 세포일 수 있으며, 이에 제한되는 것은 아니다. In another aspect, the present invention relates to a cell transformed with the above-mentioned vector. The cells used to produce the antibodies of the invention can be prokaryote, yeast or higher eukaryote cells, but are not limited thereto.
에스케리치아 콜라이(Escherichia coli), 바실러스 서브틸리스 및 바실러스 츄린겐시스와 같은 바실러스 속 균주, 스트렙토마이세스(Streptomyces), 슈도모나스(Pseudomonas)(예를 들면, 슈도모나스 푸티다(Pseudomonas putida)), 프로테우스미라빌리스(Proteus mirabilis) 및 스타필로코쿠스(Staphylococcus)(예를 들면, 스타필로코쿠스 카르노수스(Staphylocus carnosus))와 같은 원핵 숙주세포를 이용할 수 있다.Bacillus strains such as Escherichia coli, Bacillus subtilis and Bacillus thuringiensis, Streptomyces, Pseudomonas (e.g. Pseudomonas putida), Proteus Prokaryotic host cells such as Proteus mirabilis and Staphylococcus (eg, Staphylocus carnosus) can be used.
다만, 동물 세포에 대한 관심이 가장 크며, 유용한 숙주 세포주의 예는 COS-7, BHK, CHO, CHOK1, DXB-11, DG-44, CHO/-DHFR, CV1, COS-7, HEK293, BHK, TM4, VERO, HELA, MDCK, BRL 3A, W138, Hep G2, SK-Hep, MMT, TRI, MRC 5, FS4, 3T3, RIN, A549, PC12, K562, PER.C6, SP2/0, NS-0, U20S, 또는 HT1080일 수 있으나, 이에 제한되는 것은 아니다.However, the greatest interest in animal cells, examples of useful host cell lines are COS-7, BHK, CHO, CHOK1, DXB-11, DG-44, CHO / -DHFR, CV1, COS-7, HEK293, BHK, TM4, VERO, HELA, MDCK, BRL 3A, W138, Hep G2, SK-Hep, MMT, TRI, MRC 5, FS4, 3T3, RIN, A549, PC12, K562, PER.C6, SP2 / 0, NS-0 , U20S, or HT1080, but is not limited thereto.
본 발명은 또 다른 관점에서, (a) 상기 세포를 배양하는 단계; 및 (b) 상기 배양된 세포에서 항체 또는 그의 항원 결합 단편을 회수하는 단계를 포함하는 상기 항체 또는 그의 항원 결합 단편의 제조방법에 관한 것이다. In another aspect, the present invention, (a) culturing the cells; And (b) recovering the antibody or antigen-binding fragment thereof from the cultured cells.
상기 세포는 각종 배지에서 배양할 수 있다. 시판용 배지 중 제한없이 배양 배지로서 사용할 수 있다. 당업자에게 공지되어 있는 기타 모든 필수 보충물이 적당한 농도로 포함될 수도 있다. 배양 조건, 예를 들어 온도, pH 등이 발현을 위해 선별된 숙주 세포와 함께 이미 사용되고 있고, 이는 당업자에게 명백할 것이다.The cells can be cultured in various media. It can be used as a culture medium without limitation among commercial media. All other necessary supplements known to those skilled in the art may be included at appropriate concentrations. Culture conditions, such as temperature, pH, and the like, are already in use with host cells selected for expression, which will be apparent to those skilled in the art.
상기 항체 또는 그의 항원 결합 단편의 회수는 예를 들어 원심분리 또는 한외여과에 의해 불순물을 제거하고, 그 결과물을 예를 들어 친화 크로마토그래피 등을 이용하여 정제할 수 있다. 추가의 기타 정제 기술 예를 들어 음이온 또는 양이온 교환 크로마토그래피, 소수성 상호 작용 크로마토그래피, 히드록실아파타이트 크로마토그래피 등이 사용될 수 있다. The recovery of the antibody or antigen-binding fragment thereof can be removed by, for example, centrifugation or ultrafiltration, and the resultant can be purified using, for example, affinity chromatography or the like. Further other purification techniques such as anion or cation exchange chromatography, hydrophobic interaction chromatography, hydroxylapatite chromatography and the like can be used.
본 발명은 또 다른 관점에서, 상기 항체 또는 그의 항원 결합 단편을 유효성분으로 포함하는 혈관신생 질환의 예방 또는 치료용 조성물에 관한 것이다. In another aspect, the present invention relates to a composition for preventing or treating angiogenic diseases comprising the antibody or antigen-binding fragment thereof as an active ingredient.
상기 혈관신생은 이전에 존재한 혈관으로부터 새로운 혈관의 형성 또는 성장을 의미하며, "혈관신생 관련 질환(angiogenesis-related disease)"은 혈관신생의 발생 또는 진행과 관련된 질환을 의미한다. 상기 항체로 치료될 수 있다면, 그 질병은 한정 없이 혈관신생 관련 질환의 범위에 포함될 수 있다. 혈관신생 관련 질환의 예로는 암, 전이(metastasis), 당뇨망막병증(diabetic retinopathy), 미숙아 망막병증(retinopathy of prematurity), 각막 이식 거부(corneal graft rejection), 황반변성(macular degeneration), 혈관신생성녹내장(neovascular glaucoma), 전신홍색증(erythrosis), 증식성망막증(proliferative retinopathy), 건선(psoriasis), 혈우병성 고관절염(hemophilic arthritis), 동맹경화성 플라크(atherosclerotic plaques)의 모세혈관 형성, 켈로이드(keloid), 상처 과립화(wound granulation), 혈관 유착(vascular adhesion), 류마티스 관절염(rheumatoid arthritis), 퇴행성 관절염(osteoarthritis), 자기면역질환(autoimmune diseases), 크론병(Crohn's disease), 레스테노시스(restenosis), 죽상동맥경화증(atherosclerosis), 장협착(intestinal adhesions), 고양이 찰과상 감염증(cat scratch disease), 궤양(ulcer), 간경변증(liver cirrhosis), 신장염(nephritis), 당뇨병성 신장질환(diabetic nephropathy), 진성 당뇨병(diabetes mellitus), 염증 질환(inflammatory diseases) 및 신경변성 질환(neurodegenerative diseases)을 포함하나, 이에 한정되는 것은 아니다. 또한, 상기 암은 식도암(esophageal cancer), 위암(stomach cancer), 대장암(large intestine cancer), 직장암(rectal cancer), 구강암(oral cancer), 인두암(pharynx cancer), 후두암(larynx cancer), 폐암(lung cancer), 결장암(colon cancer), 유방암(breast cancer), 자궁경부암(uterine cervical cancer), 자궁내막암(endometrial cancer), 난소암(ovarian cancer), 전립선암(prostate cancer), 고환암(testis cancer), 방광암(bladder cancer), 신장암(renal cancer), 간암(liver cancer), 췌장암(pancreatic cancer), 뼈암(bone cancer), 결합조직암(connective tissue cancer), 피부암(skin cancer), 뇌종양(brain cancer), 갑상선암(thyroid cancer), 백혈병(leukemia), 호지킨 림프종(Hodgkin's lymphoma), 림프종(lymphoma) 및 다발성 골수혈액암(multiple myeloid blood cancer)으로 구성된 군에서 선택되나, 이에 한정되는 것은 아니다.Angiogenesis refers to the formation or growth of new blood vessels from previously existing blood vessels, and “angiogenesis-related disease” refers to a disease associated with the development or progression of angiogenesis. If the antibody can be treated, the disease can be included within the scope of angiogenesis-related diseases without limitation. Examples of angiogenesis-related diseases include cancer, metastasis, diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, macular degeneration, angiogenesis Neovascular glaucoma, erythrosis, proliferative retinopathy, psoriasis, hemophilic arthritis, capillary formation of atherosclerotic plaques, keloid , Wound granulation, vascular adhesion, rheumatoid arthritis, osteoarthritis, autoimmune diseases, Crohn's disease, restenosis , Atherosclerosis, intestinal adhesions, cat scratch disease, ulcers, liver cirrhosis, nephritis, diabetic kidney One comprising a ring (diabetic nephropathy), diabetes mellitus (diabetes mellitus), inflammatory diseases (inflammatory diseases) and neurodegenerative disorders (neurodegenerative diseases), but is not limited to such. In addition, the cancer is esophageal cancer (esophageal cancer), stomach cancer (stomach cancer), colon cancer (large intestine cancer), rectal cancer (rectal cancer), oral cancer (oral cancer), pharyngeal cancer (pharynx cancer), larynx cancer (larynx cancer), Lung cancer, colon cancer, breast cancer, breast cancer, uterine cervical cancer, endometrial cancer, ovarian cancer, prostate cancer, testicular cancer testis cancer, bladder cancer, renal cancer, liver cancer, liver cancer, pancreatic cancer, bone cancer, connective tissue cancer, skin cancer, Selected from the group consisting of brain cancer, thyroid cancer, leukemia, Hodgkin's lymphoma, lymphoma, and multiple myeloid blood cancer It is not.
여기에 사용된 용어 "예방(prevention or prophylaxis)"은 본 발명의 항체 또는 조성물을 투여하여 관심 질환의 시작을 억제 또는 지연하게 하는 모든 조치를 가리킨다. 용어 "치료(treatment or therapy)"는 본 발명의 항체 또는 조성물을 투여하여 관심 질환의 증상이 개선되거나 증상이 호전되게 하는 모든 조치를 가리킨다.As used herein, the term "prevention or prophylaxis" refers to any measure which results in inhibiting or delaying the onset of the disease of interest by administering the antibody or composition of the invention. The term “treatment or therapy” refers to any measure by which an antibody or composition of the invention is administered to improve or ameliorate the symptoms of a disease of interest.
본 발명의 항체를 포함하는 조성물은 바람직하게는 약학적 조성물이고, 본 분야에서 전형적으로 사용되는 적절한 비히클, 부형제 또는 희석제를 추가로 포함할 수 있다.Compositions comprising the antibodies of the invention are preferably pharmaceutical compositions and may further comprise suitable vehicles, excipients or diluents typically used in the art.
약학적으로 허용가능한 비히클을 포함하는 약학적 조성물은 정제, 알약, 분말, 과립제, 캡슐제, 서스펜션, 내복용액, 에멀젼, 시럽, 멸균수용액, 비수용액, 서스펜션, 동결건조물(lyophilizates) 및 좌약과 같은 다양한 경구 또는 비경구 복용형태일 수 있다. 관련하여 본 발명의 약학적 조성물은 필러, 증점제, 바인더, 습윤제, 정제분해물질(disintegrant), 계면활성제 등과 같은 희석제 또는 부형제로 조합하여 제형화될 수 있다. 경구 투여를 위한 고상 조제는 정제, 알약, 분말, 과립제, 캡슐제 등과 같은 형태일 수 있다. 이들 고상제와 관련하여 본 발명의 화합물은 녹말, 칼슘 카보네이트, 수크로즈, 락토오즈, 또는 젤라틴과 같은 하나 이상의 부형제를 조합하여 제형화될 수 있다. 간단한 부형제, 마그네슘 스테아레이트, 탈크 등과 같은 윤활제를 추가로 사용할 수 있다. 경구 투여를 위한 액체 조제는 서스펜션, 내복용액, 에멀젼, 시럽 등일 수 있다. 물 또는 액체 파라핀과 같은 간단한 희석제, 습윤제, 감미제, 방향족, 방부제 등과 같은 다양한 부형제를 액체 조제 내에 포함시킬 수 있다. 또한, 본 발명의 약학적 조성물은 멸균수용액, 비수용성 용매, 서스펜션, 에멀젼, 동결건조물, 좌약 등과 같은 비경구 복용 형태일 수 있다. 주입가능한 프로필렌 글리콜, 폴리에틸렌 글리콜, 올리브유와 같은 식물성 기름 및 에틸 올레에이트와 같은 에스테르가 비수용성 용매 및 서스펜션에 적합할 수 있다. 좌약의 기본 물질은 위텝졸(Witepsol), 마크로골(macrogol), 트윈 61(Tween 61), 카카오버터(cacao butter), 라우린버터(laurin butter) 및 글리세로젤라틴(glycerogelatin)을 포함한다.Pharmaceutical compositions comprising pharmaceutically acceptable vehicles include tablets, pills, powders, granules, capsules, suspensions, internal solutions, emulsions, syrups, sterile aqueous solutions, non-aqueous solutions, suspensions, lyophilizates and suppositories. It can be in various oral or parenteral dosage forms. In this regard, the pharmaceutical compositions of the present invention may be formulated in combination with diluents or excipients such as fillers, thickeners, binders, wetting agents, disintegrants, surfactants and the like. Solid preparations for oral administration may be in the form of tablets, pills, powders, granules, capsules and the like. With respect to these solids, the compounds of the present invention may be formulated in combination with one or more excipients such as starch, calcium carbonate, sucrose, lactose, or gelatin. Lubricants such as simple excipients, magnesium stearate, talc and the like can be further used. Liquid preparations for oral administration may be suspensions, internal solutions, emulsions, syrups and the like. Various excipients such as simple diluents such as water or liquid paraffin, wetting agents, sweeteners, aromatics, preservatives and the like can be included in the liquid formulation. In addition, the pharmaceutical compositions of the present invention may be in parenteral dosage forms such as sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilisates, suppositories, and the like. Injectable propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and esters such as ethyl oleate may be suitable for non-aqueous solvents and suspensions. Base materials for suppositories include Whitepsol, macrogol, Tween 61, cacao butter, laurin butter and glycerogelatin.
본 발명의 조성물은 약학적으로 유효한 양으로 투여된다. 여기에 사용된 용어 "약학적으로 유효한 양(pharmaceutically effective amount)"은 모든 의학적 치료에 적용가능한 합당한 이익과 위험 비율(benefit/risk ratio)로 충분한 질환 치료용 약학적 조성물의 양을 가리킨다. 유효량은 치료해야 할 질환의 중증도, 환자의 나이 및 성별, 질환의 종류, 약제의 활성도, 약제에 대한 민감도, 투여시간, 투여경로, 분비속도, 치료기간, 약제의 공투여 및 본 분야에서 알려진 다른 파라미터를 포함한 다양한 요인에 따라서 달라진다. 본 발명의 조성물은 단독으로 또는 다른 치료법과 조합하여 투여될 수 있다. 이와 같은 경우에, 종래의 치료법과 함께 순서대로 또는 동시에 투여될 수 있다. 또한, 상기 조성물은 1회량 또는 다회 용량으로 나누어서 투여될 수 있다. 이들 요인들을 완전하게 고려할 때, 부작용 없이 최대의 효과를 얻기에 충분한 최소량을 투여하는 것이 중요하며, 이 복용량은 분야의 전문가에 의하여 용이하게 결정될 수 있다. 본 발명의 약학적 조성물의 복용량은 특별하게 한정되지는 않으나, 환자의 건강 상태 및 체중, 질환의 중증도, 약의 종류, 투여경로 및 투여시간을 포함한 다양한 요인에 따라 변경된다. 조성물은 하루에 1회량 또는 다회 용량으로 랫, 쥐, 가축, 인간 등을 포함하는 포유류 내로 전형적으로 허용된 경로, 예를 들면, 경구로, 직장으로, 정맥으로(intravenously), 피하로(subcutaneously), 자궁내로(intrauterinely) 또는 뇌혈관내로(intracerebrovascularly) 투여될 수 있다.The composition of the present invention is administered in a pharmaceutically effective amount. As used herein, the term “pharmaceutically effective amount” refers to the amount of a pharmaceutical composition for treating a disease sufficient to a reasonable benefit and risk ratio applicable to all medical treatments. Effective amounts include 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 time of administration, the route of administration, the rate of secretion, the duration of treatment, the coadministration of the drug, and other known in the art. It depends on various factors including the parameters. The compositions of the present invention can be administered alone or in combination with other therapies. In such cases, they may be administered sequentially or simultaneously with conventional therapies. In addition, the composition may be administered in one or multiple doses. Given these factors completely, it is important to administer the minimum amount sufficient to achieve the maximum effect without side effects, which dose can be readily determined by one of ordinary skill in the art. The dosage of the pharmaceutical composition of the present invention is not particularly limited, but varies depending on various factors including the patient's health condition and weight, the severity of the disease, the type of drug, the route of administration, and the time of administration. The compositions are typically accepted routes into mammals, including rats, mice, livestock, humans, etc., once or in multiple doses per day, for example, orally, rectally, intravenously, subcutaneously. It may be administered intrauterinely or intratracerebrovascularly.
본 발명은 또 다른 관점에서 상기 항체 또는 상기 조성물을 필요로 하는 개체에 투여하는 단계를 포함하는 혈관신생을 억제하는 방법, 혈관신생 관련 질환 예방 또는 치료 방법에 관한 것이다.In another aspect, the present invention relates to a method for inhibiting angiogenesis, a method for preventing or treating angiogenesis-related diseases, comprising administering the antibody or the composition to a subject in need.
본 발명의 방법은 혈관신생의 억제를 필요로 하는 개체에 약학적으로 유효한 양의 약학적 조성물을 투여하는 단계를 포함한다. 상기 개체는 개, 소, 말, 토끼, 마우스, 랫, 닭 및 인간과 같은 포유류일 수 있으나, 이에 한정되는 것은 아니다. 약학적 조성물은 비경구적으로(parenterally), 피하로(subcutaneously), 복강내로(intraperitoneally), 폐내로(intrapulmonarily) 또는 코내로(intranasally), 필요하다면, 국소치료를 위해 병변내(intralesional) 투여를 포함하는 적절한 방법에 의하여 투여될 수 있다. 본 발명의 약학적 조성물의 바람직한 복용량은 개체의 건강 상태 및 체중, 질환의 중증도, 약제의 종류, 투여경로 및 투여시간을 포함한 다양한 요인에 따라 변하며, 본 분야의 당업자에 의하여 용이하게 결정될 수 있다.The methods of the present invention comprise administering a pharmaceutically effective amount of the pharmaceutical composition to a subject in need thereof. The subject may be a mammal such as a dog, cow, horse, rabbit, mouse, rat, chicken and human, but is not limited thereto. The pharmaceutical composition includes parenterally, subcutaneously, intraperitoneally, intrapulmonarily or intranasally, if necessary intratralesional administration for topical treatment. Can be administered by any suitable method. Preferred dosages of the pharmaceutical compositions of the present invention vary depending on various factors, including the health and weight of the individual, the severity of the disease, the type of medicament, the route of administration and the time of administration, and can be readily determined by one skilled in the art.
본 발명은 또 다른 관점에서 상기 항체를 포함하는 암 예방 또는 치료용 약학적 조성물 또는 상기 항체 또는 상기 조성물을 필요로 하는 개체에 투여하는 단계를 포함하는 암의 예방 또는 치료 방법에 관한 것이다. 용어 '항체', '예방' 및 '치료'는 상기에 언급한 바와 같다.In another aspect, the present invention relates to a pharmaceutical composition for preventing or treating cancer, comprising the antibody, or a method for preventing or treating cancer, comprising administering the antibody or the composition to an individual in need thereof. The terms 'antibody', 'prophylaxis' and 'treatment' are as mentioned above.
본 발명의 항체로 치료 가능하다면, 암은 제한되지 않는다. 상세하게는 본 발명의 항체는 혈관신생을 억제함으로써 암의 발생 또는 진행을 예방할 수 있다. 암의 예로는 식도암(esophageal cancer), 위암(stomach cancer), 대장암(large intestine cancer), 직장암(rectal cancer), 구강암(oral cancer), 인두암(pharynx cancer), 후두암(larynx cancer), 폐암(lung cancer), 결장암(colon cancer), 유방암(breast cancer), 자궁경부암(uterine cervical cancer), 자궁내막암(endometrial cancer), 난소암(ovarian cancer), 전립선암(prostate cancer), `고환암(testis cancer), 방광암(bladder cancer), 신장암(renal cancer), 간암(liver cancer), 췌장암(pancreatic cancer), 뼈암(bone cancer), 결합조직암(connective tissue cancer), 피부암(skin cancer), 뇌종양(brain cancer), 갑상선암(thyroid cancer), 백혈병(leukemia), 호지킨 림프종(Hodgkin's lymphoma), 림프종(lymphoma) 및 다발성 골수혈액암(multiple myeloid blood cancer)을 포함하나, 이에 한정되는 것은 아니다.If treatable with the antibodies of the invention, the cancer is not limited. In detail, the antibody of the present invention can prevent the development or progression of cancer by inhibiting angiogenesis. Examples of cancer include esophageal cancer, stomach cancer, large intestine cancer, rectal cancer, oral cancer, pharynx cancer, larynx cancer, lung cancer Lung cancer, colon cancer, breast cancer, breast cancer, uterine cervical cancer, endometrial cancer, ovarian cancer, prostate cancer, testicular cancer testis cancer, bladder cancer, renal cancer, liver cancer, liver cancer, pancreatic cancer, bone cancer, connective tissue cancer, skin cancer, Brain cancer, thyroid cancer, leukemia, Hodgkin's lymphoma, lymphoma and multiple myeloid blood cancer.
또한, 본 발명의 항체는 다른 항체 또는 생물학적 활성제 또는 다양한 목적을 위한 물질과 조합하여 사용될 수 있다. 본 발명은 이러한 관점에서 상기 항체 또는 그의 항원 결합 단편을 포함하는, 다른 혈관신생 질환 치료제와의 병용투여용 조성물에 관한 것이다. In addition, the antibodies of the invention can be used in combination with other antibodies or biologically active agents or materials for various purposes. The present invention relates to a composition for co-administration with other therapeutic agents for angiogenic diseases comprising the antibody or antigen-binding fragment thereof in this respect.
상기 다른 혈관신생 질환 치료제는 항-혈관신생 약물, 소염 약물 및/또는 항암 약물을 들 수 있다. 이를 통해 서로의 저항성을 극복시키고 효능을 증진시킬 수 있다. Such other angiogenic disease therapeutic agents include anti-angiogenic drugs, anti-inflammatory drugs and / or anticancer drugs. This can overcome the resistance of each other and improve the efficacy.
본 발명에 따른 조성물에서 다른 혈관신생 질환 치료제와 병용 투여되는 경우 Tie2 항체와 다른 혈관신생 질환 치료제는 순차적 또는 동시에 투여될 수 있다. 예를 들면, 항-혈관신생 약물, 소염 약물 및/또는 항암 약물을 대상체에게 투여한 후, 활성 성분으로서 Tie2에 대한 항체 또는 이의 항원 결합단편을 포함하는 조성물을 상기 대상체에게 투여하거나, 상기 조성물을 대상체에게 투여한 후, 항-혈관신생 약물, 소염 약물 및/또는 항암 약물을 상기 대상체에게 투여한다. 경우에 따라서, 상기 조성물을 항-혈관신생 약물, 소염 약물 및/또는 항암 약물과 동시에 대상체에게 투여할 수 있다.When administered in combination with another therapeutic agent for angiogenic diseases in the composition according to the present invention, the Tie2 antibody and the therapeutic agent for other angiogenic diseases may be administered sequentially or simultaneously. For example, after administering an anti-angiogenic drug, an anti-inflammatory drug, and / or an anti-cancer drug to a subject, a composition comprising an antibody against Tie2 or an antigen binding fragment thereof as an active ingredient is administered to the subject, or the composition is administered. After administration to a subject, anti-angiogenic drugs, anti-inflammatory drugs, and / or anticancer drugs are administered to the subject. If desired, the composition may be administered to the subject simultaneously with the anti-angiogenic drug, anti-inflammatory drug and / or anticancer drug.
실시예Example
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 예시하기 위한 것으로서, 본 발명의 범위가 이들 실시예에 의해 제한되는 것으로 해석되지는 않는 것은 당업계에서 통상의 지식을 가진 자에게 있어서 자명할 것이다.Hereinafter, the present invention will be described in more detail with reference to Examples. These examples are only for illustrating the present invention, it will be apparent to those skilled in the art that the scope of the present invention is not to be construed as being limited by these examples.
실시예 1. 마우스 유래 Tie2 단일클론항체의 제작Example 1. Preparation of mouse-derived Tie2 monoclonal antibody
1.1. 인간 Tie2를 이용한 마우스 면역1.1. Mouse Immunization with Human Tie2
면역원으로 인간 Tie2의 Ig3-FNIII(1-3) 도메인(hTie2-Ig3-FNIII(1-3), 서열번호 2)을 CMV 프로모터를 함유하는 벡터 내로 클로닝 시키고, HEK293F 세포주에 transfection시켜 일시적으로 발현시켰다. 5일간 배양시킨 후, 발현된 재조합 hTie2-Ig3-FNIII(1-3) 단백질을 Protein A를 사용한 어피니티 컬럼(affinity column)으로 정제하였다. 6주간 매주 2회씩 생후 5주의 BALB/c 마우스에 정제된 hTie2-Ig3-FNIII(1-3)를 보조제와 혼합하여 주사하는 방법(100μg/주사)으로 면역화 시켰다. 면역을 획득한 마우스의 혈청 중 Tie2 항체 역가(titer)를 인간 Tie2(hTie2) ELISA 키트(R&D)로 검사하였다. 항체 역가(1:5,000 희석)가 적절하게 증가하면(OD>1.0), 면역을 획득한 생쥐에서 비장을 추출하고, 비장에서 B림프구를 분리하여 배양된 골수종 세포(myeloma cells, SP2/0)와 융합하였다. 융합된 세포는 hypoxanthine, aminopterin 및 thymidine을 함유한 HAT 배지에서 배양하고, 골수종 세포와 B림프구의 융합체만을 포함하는 하이브리도마 세포(hybridoma cells)를 선별하여 배양하였다. 생존한 하이브리도마 세포를 96-웰 플레이트(96-well plate)에 접종시키고 배양 상층액을 hTie2 ELISA를 통해 분석하였다. 한계 희석법(limiting dilution)을 통해 클론선택을 하기 위해 양성 신호를 나타내는 하이브리도마 풀(hybridoma pool)을 선별하였다. 최종적으로 37개의 단일 하이브리도마 세포주가 구축되었다. 그 중 몇 개의 Tie2 결합 항체가 Tie2 활성화 효과를 나타냈다. 후보 항체들은 Tie2 활성화 수준과 인간 Tie2에 대한 높은 친화력을 기준으로 선별되었으며, 후에 인간화(humanization)를 위해 가공되었다.As an immunogen, the Ig3-FNIII (1-3) domain of human Tie2 (hTie2-Ig3-FNIII (1-3), SEQ ID NO: 2) was cloned into a vector containing a CMV promoter and transfected into a HEK293F cell line to express temporarily. . After incubation for 5 days, the expressed recombinant hTie2-Ig3-FNIII (1-3) protein was purified by an affinity column using Protein A. Twice a week for 6 weeks, 5 weeks old BALB / c mice were immunized with the injection of purified hTie2-Ig3-FNIII (1-3) mixed with adjuvant (100 μg / injection). Tie2 antibody titers in the sera of immunized mice were examined with a human Tie2 (hTie2) ELISA kit (R & D). When antibody titers (1: 5,000 dilution) were properly increased (OD> 1.0), spleens were extracted from immunized mice, B lymphocytes were isolated from the spleen, and myeloma cells (SP2 / 0) were cultured. Fused. The fused cells were cultured in HAT medium containing hypoxanthine, aminopterin and thymidine, and hybridoma cells containing only fusions of myeloma cells and B lymphocytes were selected and cultured. Surviving hybridoma cells were seeded in 96-well plates and culture supernatants were analyzed by hTie2 ELISA. Hybridoma pools displaying positive signals were selected for clonal selection through limiting dilution. Finally 37 single hybridoma cell lines were constructed. Several of the Tie2 binding antibodies showed a Tie2 activation effect. Candidate antibodies were selected based on Tie2 activation level and high affinity for human Tie2 and later processed for humanization.
Figure PCTKR2019006820-appb-T000001
Figure PCTKR2019006820-appb-T000001
Figure PCTKR2019006820-appb-I000001
Figure PCTKR2019006820-appb-I000001
1.2. 마우스 항-Tie2 단일클론항체의 제조 및 정제1.2. Preparation and Purification of Mouse Anti-Tie2 Monoclonal Antibodies
ELISA 양성 신호에 기초하여 선별된 항-Tie2 항체를 생산하기 위해, T75(75cm2 면적) 플라스크 내 10% FBS함유-DMEM(Dulbesco Modified Eagle`s Medium)에서 하이브리도마 세포를 배양하였다. 세포의 밀집도(confluency of cell)가 약 90%에 달했을 때, PBS로 세포를 세척하고, 50ml의 serum-free medium (SFM, Gibco)에서 인큐베이션한 다음, 3일간 37℃에서 배양하였다. 이후, 각 단일클론 하이브리도마에서 분비되는 항체가 포함된 배양액을 수집하고, 원심분리하여 세포를 제거하였으며, 배양 상등액을 수집하여 여과하였다. Protein G 단백질 친화 컬럼(GE Healthcare.)이 장착된 AKTA 정제 장치(GE Healthcare.)를 이용하여 배양 상등액으로부터 항체를 정제하였다. 이후, 원심 여과 장치(centrifugal filter unit, Amicon)를 이용해 항체 용액의 버퍼를 PBS로 대체하고 정제된 항체를 농축하였다.To produce anti-Tie2 antibodies selected based on ELISA positive signals, hybridoma cells were cultured in 10% FBS containing-DMEM (Dulbesco Modified Eagle's Medium) in T75 (75 cm 2 area) flasks. When the concentration of cells reached about 90%, the cells were washed with PBS, incubated in 50 ml of serum-free medium (SFM, Gibco), and then incubated at 37 ° C for 3 days. Thereafter, the culture solution containing the antibody secreted from each monoclonal hybridoma was collected, centrifuged to remove cells, and the culture supernatant was collected and filtered. Antibodies were purified from the culture supernatant using an AKTA purification apparatus (GE Healthcare.) Equipped with a Protein G protein affinity column (GE Healthcare.). Thereafter, the buffer of the antibody solution was replaced with PBS using a centrifugal filter unit (Amicon), and the purified antibody was concentrated.
1.3. Tie2 활성화 항체(agonistic Tie2 antibody)의 스크리닝 및 선별.1.3. Screening and Screening of Aieistic Tie2 Antibodies.
마우스 항-Tie2 항체가 혈관내피세포(endothelial cell)에서 Tie2 수용체의 하위 신호 전달 과정을 유도하는지 여부를 조사하기 위해, HUVEC(Lonza)을 항-Tie2 항체로 처리한 후, Tie2 수용체의 주요 하위 신호 전달 단백질인 Akt 인산화 수준을 immunoblotting 기법으로 분석하였다. 양성 대조군으로, COMP-Ang1(CA1)을 세포에 처리하였다.To investigate whether the mouse anti-Tie2 antibody induces a downstream signaling process of the Tie2 receptor in endothelial cells, after treatment with HUVEC (Lonza) with an anti-Tie2 antibody, the major downstream signal of the Tie2 receptor The transfer protein Akt phosphorylation level was analyzed by immunoblotting technique. As a positive control, COMP-Angl (CA1) was treated with the cells.
구체적으로, 60mm 배양 접시 내 EGM-2 배지(Lonza)에서 HUVEC(1x105 cells/ml)를 37℃ 온도에서 배양하였다. 90% 밀집도의 세포를 혈청 기아 상태로 만들기 위해, 무혈청 EBM-2 배지에서 6시간 동안 인큐베이션하였다. 혈청 기아 상태의 HUVEC에 항-Tie2 항체를 처리하고 30분동안 추가로 배양하였다. 세포를 차가운 PBS로 세척한 뒤, 용해 버퍼를 처리하고 4℃에서 20분간 용해시켰다. 이어서, 13000rpm에서 15분간 원심분리하여 침전물을 제거하고 세포 용해물을 획득하였다. 세포용해물에 5x SDS 샘플 버퍼(5x SDS sample buffer)를 추가한 후 단백질 전기영동(SDS-PAGE) 하였고, 이어 니트로셀룰로오스 막(NC membrane, GE)에 옮겼다. Specifically, HUVEC (1 × 10 5 cells / ml) was incubated at 37 ° C. in EGM-2 medium (Lonza) in a 60 mm culture dish. Cells of 90% density were incubated for 6 hours in serum-free EBM-2 medium to bring serum starvation. Serum starved HUVECs were treated with anti-Tie2 antibody and further incubated for 30 minutes. Cells were washed with cold PBS, then treated with lysis buffer and lysed at 4 ° C. for 20 minutes. Subsequently, the precipitate was removed by centrifugation at 13000 rpm for 15 minutes to obtain cell lysate. 5x SDS sample buffer was added to the cell lysate, followed by protein electrophoresis (SDS-PAGE), followed by transfer to nitrocellulose membrane (NC membrane, GE).
Akt 인산화 여부를 확인하기 위해, 5% skim milk를 함유한 TBS-T로 실온(Room Temperature, RT) 에서 1시간 동안 블로킹하고. 4℃에서 8시간 동안 p-Akt 항체(S473)와 함께 인큐베이션하였다. p-Akt 신호는 ECL (Enhanced ChemiLuminescence)에 의해 검출 되었다. 이후, 막을 15분동안 스트리핑 버퍼(stripping buffer, Thermo)에서 15분간 인큐베이션한 다음, 항-Akt 항체로 다시 프로빙하여 총 Akt의 양을 확인하였다. 분석 결과, 11C4, 4A4, 3B2, 3E12 및 3H7과 같은 항-Tie2 항체로 처리된 몇몇 그룹에서 Akt의 S473 인산화가 강하게 유도되었다(도 1).To check for Akt phosphorylation, TBS-T containing 5% skim milk was blocked for 1 hour at room temperature (RT). Incubated with p-Akt antibody (S473) for 8 hours at 4 ° C. p-Akt signal was detected by ECL (Enhanced ChemiLuminescence). The membrane was then incubated for 15 minutes in a stripping buffer (thermo) for 15 minutes and then probed again with an anti-Akt antibody to confirm the total amount of Akt. As a result, several groups treated with anti-Tie2 antibodies such as 11C4, 4A4, 3B2, 3E12 and 3H7 strongly induced S473 phosphorylation of Akt (FIG. 1).
1.4. Octet 분석법에 의한 hTie2에 대한 Tie2항체의 친화도 측정.1.4. Determination of the affinity of Tie2 antibodies for hTie2 by Octet assay.
Black 96-웰 플레이트(96 well F-type black plates, Greiner)가 사용된 옥텟 시스템(octet system, ForteBio)을 이용하여 hTie2에 대한 마우스 단일클론항체의 친화도를 측정하였다. 친화도 측정에 사용된 바이오센서는 AR2G 팁(ForteBio Octet)으로 측정하기 전에 10분 동안 하이드레이션(hydration)되었다. 하이드레이션 이후, hTie2를 pH6.0 버퍼인 10mM 소듐 아세테이트에서 10μg/ml 농도로 희석하고, AR2G 바이오센서 상에 고정시킨 다음, 1M ethanolamine으로 블로킹하였다. 마우스 항-Tie2 단일클론항체를 1x kinetic 버퍼로 50, 25, 12.5, 6.25, 3.125 및 0nM으로 희석한 뒤, 300초 동안 결합시키고 900초 동안 해리시켰다. 친화도 측정(KD)을 위해, K-on(association rate) 및 K-off(dissociation rate)를 결합 곡선(global)으로 분석하고 Octet 데이터 분석 프로그램(v.9.0.0.10)을 사용하여 1:1 결합 모델에 맞추었다. 표 2는 마우스 항-Tie2 항체의 친화도를 나타낸 것이다.The affinity of the mouse monoclonal antibody for hTie2 was measured using an octet system (ForteBio) using Black 96-well plate (Greiner). The biosensors used for affinity measurements were hydrated for 10 minutes before being measured with an AR2G tip (ForteBio Octet). After hydration, hTie2 was diluted to 10 μg / ml concentration in 10 mM sodium acetate, pH6.0 buffer, fixed on AR2G biosensor and blocked with 1M ethanolamine. Mouse anti-Tie2 monoclonal antibodies were diluted to 50, 25, 12.5, 6.25, 3.125 and 0 nM with 1 × kinetic buffer, then bound for 300 seconds and dissociated for 900 seconds. For affinity measurements (KD), the association rate (K-on) and the dissociation rate (K-off) were analyzed globally and 1: 1 using Octet data analysis program (v.9.0.0.10). Fit to coupled model. Table 2 shows the affinity of mouse anti-Tie2 antibodies.
Figure PCTKR2019006820-appb-T000002
Figure PCTKR2019006820-appb-T000002
1.5. 마우스 Tie2 항체(3H7)에 의해 유도된 Tie2 인산화.1.5. Tie2 phosphorylation induced by mouse Tie2 antibody (3H7).
본 발명에 따른 항-Tie2 항체는 Tie2에 결합하고 Tie2 클러스터링을 유도하여 궁극적으로는 Tie2 활성화를 유도하는 것으로 보인다. HUVEC를 이용하여 Tie2 인산화에 대한 항-Tie2 항체의 효과를 분석하는 실험이 수행되었다.Anti-Tie2 antibodies according to the present invention appear to bind to Tie2 and induce Tie2 clustering, ultimately leading to Tie2 activation. Experiments were performed to analyze the effect of anti-Tie2 antibodies on Tie2 phosphorylation using HUVECs.
구체적으로, HUVEC는 100mm 배양 접시 내에서 EGM-2(Lonza) 배지를 이용해 37℃, 5% CO2농도 조건에서 배양되었다. 세포가 80-90%의 밀집도(confluency)가 되었을 때, 혈청 기아 상태로 만들기 위해 배지를 EBM-2(Lonza) 로 교체하고 세포를 6시간 동안 추가 배양하였다. 다양한 농도의 항-Tie2 항체(0.02μg/ml 내지 50μg/ml)를 배양된 세포에 처리하고 30분 동안 추가 인큐베이션하였다. 세포를 차가운 PBS로 2회 세척하고 1000μl의 용해 버퍼(10mM Tris-Cl pH 7.4, 150mM NaCl, 5mM EDTA, 10% glycerol, 1% Triton X-100, protease inhibitor, phosphatase inhibitor)를 추가한 후 4℃에서 60분간 인큐베이션하였다. 세포 용해물을 얻기 위해 12,000rpm에서 10분간 원심 분리하였고 상층액을 획득하였다. BCA 분석법으로 상층액의 단백질 농도를 정량하였다.Specifically, HUVEC was incubated in 37%, 5% CO 2 concentration conditions using EGM-2 (Lonza) medium in a 100mm culture dish. When the cells reached a confluency of 80-90%, the medium was replaced with EBM-2 (Lonza) to bring serum starvation and the cells were further incubated for 6 hours. Various concentrations of anti-Tie2 antibody (0.02 μg / ml to 50 μg / ml) were treated to the cultured cells and further incubated for 30 minutes. Wash cells twice with cold PBS and add 1000μl of lysis buffer (10mM Tris-Cl pH 7.4, 150mM NaCl, 5mM EDTA, 10% glycerol, 1% Triton X-100, protease inhibitor, phosphatase inhibitor) Incubate for 60 minutes at. The cells were centrifuged at 12,000 rpm for 10 minutes to obtain cell lysates and supernatants were obtained. Protein concentration of the supernatant was quantified by BCA assay.
Tie2를 면역 침전시키기 위해, Tie2 항체(R&D systems, AF313) 1μg을 0.5mg의 용해물에 첨가하고, 배양(4℃, overnight, shaking)하였다. 이후, Dynabeads ™ Protein G(Life technologies)를 첨가하여 2 시간 반응시켰다. 자석을 사용하여 튜브의 한 면에 비드를 고정시키고 용해 버퍼로 3 회 세척 한 다음, 환원제가 함유된 2x SDS 샘플 버퍼로 70℃에서 10 분 동안 인큐베이션하였다. 샘플에서 비드를 제거하고 4-15% SDS 단백질 겔(Bio-Rad)에서 전기영동한 다음, 0.45 μm PVDF 막으로 옮겼다.To immunoprecipitate Tie2, 1 μg of Tie2 antibody (R & D systems, AF313) was added to 0.5 mg of lysate and incubated (4 ° C., overnight, shaking). Thereafter, Dynabeads ™ Protein G (Life technologies) was added and reacted for 2 hours. The beads were fixed on one side of the tube using a magnet and washed three times with lysis buffer and then incubated for 10 minutes at 70 ° C. with a 2 × SDS sample buffer containing reducing agent. Beads were removed from the sample and electrophoresed on 4-15% SDS protein gel (Bio-Rad) and then transferred to 0.45 μm PVDF membrane.
막을 5%(v/v) BSA와 혼합한 TBS-T를 이용하여 실온에서 1시간 동안 블로킹시키고 항-인산 티로신 항체(4G10, Millipore)와 함께 4℃에서 8시간 동안 인큐베이션한 후, HRP 컨쥬게이트된 항-마우스 항체(HRP conjugated anti-mouse antibody)를 인큐베이션한 다음, 웨스턴 블롯으로 분석하였다. 면역침전된 Tie2의 양을 측정하기 위해, 막을 15분 동안 stripping 버퍼(Thermo)에서 반응시킨 다음, 다시 블로킹하고 항-Tie2 항체(R&D systems, AF313)로 다시 프로빙 하였다. 도 2에 도시된 바와 같이, 항-Tie2 항체(3H7)를 HUVEC 세포에 첨가하면 Tie2의 인산화가 용량 의존적으로 강하게 유도되었다. 이러한 데이터는 항-Tie2 항체(3H7)가 인간의 혈관내피세포에서 직접적으로 Tie2 수용체의 활성화를 유도함을 나타낸다.Membrane was blocked for 1 hour at room temperature using TBS-T mixed with 5% (v / v) BSA and incubated with anti-phosphotyrosine antibody (4G10, Millipore) at 4 ° C. for 8 hours before HRP conjugate The anti-mouse antibody (HRP conjugated anti-mouse antibody) was incubated and analyzed by Western blot. To determine the amount of immunoprecipitated Tie2, the membrane was reacted in stripping buffer (Thermo) for 15 minutes, then blocked again and probed again with anti-Tie2 antibodies (R & D systems, AF313). As shown in FIG. 2, the addition of anti-Tie2 antibody (3H7) to HUVEC cells induced a dose-dependently strong phosphorylation of Tie2. These data show that anti-Tie2 antibody (3H7) induces activation of Tie2 receptor directly in human vascular endothelial cells.
1.6 Tie2 항체(3H7)에 의해 HUVEC에서 클러스터링된 Tie2의 세포 내 유입(endocytosis) 및 FOXO1의 전좌(translocation)1.6 Endocytosis of Tie2 Clustered in HUVEC by Tie2 Antibody (3H7) and Translocation of FOXO1
HUVEC에서 3H7 항체에 의한 Tie2의 세포내 유입(endocytosis) 및 핵에서 세포질로의 FOXO1 전좌를 면역형광법으로 조사하였다. 구체적으로, HUVEC를 8-웰 슬라이드 챔버(Lab-TekII)에 접종하고, 2~3일 동안 EGM-2 배지에서 유지시켰다. 100% 밀집도(confluence)에서, 세포를 EBM-2 배지를 이용하여 4시간 동안 혈청 기아 상태로 만든 다음 1μg/ml의 Tie2 항체(3H7)로 30분 동안 처리하였다. 그 후, 세포를 실온(RT)에서 10분간 4% 포름알데히드/PBS 용액으로 고정하고, 0.1% Triton X-100/PBS 용액으로 투과시켰으며, 실온에서 60분간 1% BSA/PBS 용액으로 블로킹시키고, 1차 항체로 RT에서 1시간 동안 인큐베이션하였다. 1차 항체로는 hTie2 및 FOXO1에 대한 항체가 사용되었다. 이후, 세포를 1시간 동안 실온의 암조건에서 형광 표지된 2차 항체와 함께 배양하였으며, 핵 염색을 위해 DAPI(Vector Labs)가 포함된 Vectashield mounting medium으로 장착하였다. 이후 세포 이미지는 레이저 스캐닝 공초점 현미경(laser scanning confocal microscope; LSM880, Carl Zeiss)으로 촬영되어 분석되었다.Intracellular influx of Tie2 by 3H7 antibody in HUVEC and FOXO1 translocation from nucleus to cytoplasm were examined by immunofluorescence. Specifically, HUVECs were inoculated in an 8-well slide chamber (Lab-TekII) and maintained in EGM-2 medium for 2-3 days. At 100% confluence, cells were serum starved for 4 hours using EBM-2 medium and then treated with 1 μg / ml Tie2 antibody (3H7) for 30 minutes. Cells were then fixed with 4% formaldehyde / PBS solution for 10 minutes at room temperature (RT), permeabilized with 0.1% Triton X-100 / PBS solution, blocked with 1% BSA / PBS solution for 60 minutes at room temperature and , Incubated with primary antibody for 1 hour at RT. As primary antibodies, antibodies against hTie2 and FOXO1 were used. Thereafter, the cells were incubated with fluorescently labeled secondary antibody in dark conditions at room temperature for 1 hour, and mounted with a Vectashield mounting medium containing DAPI (Vector Labs) for nuclear staining. Cell images were then taken and analyzed with a laser scanning confocal microscope (LSM880, Carl Zeiss).
도 3에 도시된 바와 같이, 3H7 처리 시 Tie2 클러스터링 및 활성화를 유도하는 것으로 알려진 대조군 Ang2 항체(Han et al., 2016, Science Translation Medicine)와 마찬가지로 Tie2 세포 내 유입을 강하게 유도하였다. 인산화될 경우 FOXO1은 핵에서 세포질로 전좌(translocation)된다는 이전의 보고(Zhang et al, JBC 2002, 277, 45276-45284)와 같이, 혈청 기아 상태에서 음성 대조군(Control)의 FOXO1은 핵 안에 존재하는 것에 반하여(빨강), 3H7 처리 시 핵에서 FOXO1의 신호가 사라지고 세포질에서 대부분 검출되었다. As shown in FIG. 3, the influx into Tie2 cells was strongly induced as in the control Ang2 antibody (Han et al., 2016, Science Translation Medicine), which is known to induce Tie2 clustering and activation upon 3H7 treatment. As reported earlier (Zhang et al, JBC 2002, 277, 45276-45284) that FOXO1 translocates from the nucleus to the cytoplasm when phosphorylated, FOXO1 of the negative control in serum starvation is present in the nucleus. In contrast (red), 3H7 treatment disappeared the signal of FOXO1 in the nucleus and was mostly detected in the cytoplasm.
1.7. HUVEC에 항-Tie2 항체(3H7) 처리시 VEGF 또는 TNF-a에 의해 유도되는 혈관 투과성의 억제1.7. Inhibition of vascular permeability induced by VEGF or TNF-a upon HUVEC treatment with anti-Tie2 antibody (3H7)
HUVEC에서 혈관 누출 분석은 제조업자가 지시하는 방법에 따라 In Vitro Vascular Permeability Assay Kit (Millipore)를 사용하여 수행되었다. HUVEC를 트랜스웰 플레이트(transwell plate) 인서트에 접종하고 100% 밀집도가 될 때까지 3일 동안 배양하였다. Ang2(1μg/ml) 단독, Ang2(1μg/ml)와 대조군 항체(1μg/ml) 혼합물 또는 3H7 항체(1μg/ml) 단독을 각각 30분 동안 HUVEC에서 사전 인큐베이션한 다음, VEGF(500ng/ml) 또는 TNF-α(100ng/ml)를 첨가하고, 세포를 37 ℃에서 45분 또는 22시간 동안 별도로 인큐베이션하였다. 이어 FITC-dextran을 상부 챔버에 첨가하고 20분 동안 인큐베이션하였다. HUVEC 단일층을 투과하여 하부 챔버에 축적된 FITC-dextran의 양을 각각 485 및 535 nm의 여기(excitation) 파장 및 방출(emission) 파장에서 형광 판독기로 측정하였다. 도 4에 도시된 바와 같이, 항-Tie2 항체(3H7)의 사전처리는 혈관 누출 촉진인자인 VEGF 또는 TNF-α에 의해 유발되는 혈관 누출을 억제하였다.Vascular leak analysis in HUVEC was performed using the In Vitro Vascular Permeability Assay Kit (Millipore) according to the manufacturer's instructions. HUVECs were inoculated into transwell plate inserts and incubated for 3 days until 100% density. Ang2 (1 μg / ml) alone, a mixture of Ang2 (1 μg / ml) and control antibody (1 μg / ml) or 3H7 antibody (1 μg / ml) alone was preincubated in HUVECs for 30 minutes each, followed by VEGF (500 ng / ml). Or TNF-α (100 ng / ml) and cells were incubated separately at 37 ° C. for 45 minutes or 22 hours. FITC-dextran was then added to the upper chamber and incubated for 20 minutes. The amount of FITC-dextran that penetrated the HUVEC monolayer and accumulated in the lower chamber was measured with a fluorescence reader at excitation and emission wavelengths of 485 and 535 nm, respectively. As shown in FIG. 4, pretreatment of anti-Tie2 antibody (3H7) inhibited vascular leakage caused by VEGF or TNF-α, a vascular leak promoter.
실시예 2. 마우스 유래 Tie2 항체 유전자의 DNA 염기서열 분석.Example 2. DNA sequencing of mouse-derived Tie2 antibody genes.
실시예 1.3에서 선택된 항체(하이브리도마 세포 유래)의 DNA 염기서열을 분석하였다. 구체적으로, 하이브리도마 세포(2x106 cells/ml)를 10% FBS 함유 DMEM에서 배양한 뒤 RNeasy 미니 키트(Qiagen)를 사용하여 총 RNA를 수득하였다. 다음으로 RNA 농도를 측정하고, 역전사(RT) 반응을 통해 cDNA를 합성하였다. 중쇄 및 경쇄 가변영역 유전자 서열을 증폭하기 위해, cDNA를 주형으로 하여 하기 조건하에서 마우스 Ig-프라이머 세트(Novagen)를 사용하여 PCR을 수행하였다: 94℃ 5분; [94℃에서 1분, 50℃에서 1분, 72℃에서 2분]x35회; 72℃에서 6분; 4℃로 냉각. 각 반응에서 얻어진 PCR 산물을 TA벡터에 클로닝하여 DNA 시퀀싱하였으며 그 결과 각 항체의 CDR, 중쇄 가변 영역 및 경쇄 가변 영역을 코딩하는 염기 서열을 얻었다(표 3 내지 표 12). DNA sequences of the antibodies selected from Example 1.3 (derived from hybridoma cells) were analyzed. Specifically, hybridoma cells (2 × 10 6 cells / ml) were incubated in DMEM containing 10% FBS and total RNA was obtained using RNeasy mini kit (Qiagen). Next, RNA concentration was measured and cDNA was synthesized through reverse transcription (RT) reaction. To amplify heavy and light chain variable region gene sequences, PCR was performed using a mouse Ig-primer set (Novagen) under the following conditions with cDNA as a template: 94 ° C. 5 min; [35 min at 94 ° C., 1 min at 50 ° C., 2 min at 72 ° C.] × 35 times; 6 minutes at 72 ° C .; Cooled to 4 ° C. PCR products obtained in each reaction were cloned into TA vectors to sequence DNA, and as a result, base sequences encoding CDRs, heavy chain variable regions, and light chain variable regions of each antibody were obtained (Tables 3 to 12).
Figure PCTKR2019006820-appb-T000003
Figure PCTKR2019006820-appb-T000003
Figure PCTKR2019006820-appb-T000004
Figure PCTKR2019006820-appb-T000004
Figure PCTKR2019006820-appb-T000005
Figure PCTKR2019006820-appb-T000005
Figure PCTKR2019006820-appb-T000006
Figure PCTKR2019006820-appb-T000006
Figure PCTKR2019006820-appb-T000007
Figure PCTKR2019006820-appb-T000007
Figure PCTKR2019006820-appb-T000008
Figure PCTKR2019006820-appb-T000008
Figure PCTKR2019006820-appb-T000009
Figure PCTKR2019006820-appb-T000009
Figure PCTKR2019006820-appb-T000010
Figure PCTKR2019006820-appb-T000010
Figure PCTKR2019006820-appb-T000011
Figure PCTKR2019006820-appb-T000011
Figure PCTKR2019006820-appb-T000012
Figure PCTKR2019006820-appb-T000012
실시예 3. hTie2에 대한 항-Tie2 항체의 에피토프 매핑Example 3. Epitope Mapping of Anti-Tie2 Antibodies Against hTie2
마우스 단일클론항체(3H7)에 의해 인식되는 hTie2의 에피토프를 HDX-MS(수소/중수소 교환-질량 분석법) 기법으로 분석하였다. HDX-MS 분석 기법은 다음 Houde D, Engen JR (2013) Methods Mol. Biol. 988: 269-89와 Houde et al. (2011) J. Pharm. Sci. 100 (6), 2071.에 자세히 설명되어 있다.Epitopes of hTie2 recognized by mouse monoclonal antibody (3H7) were analyzed by HDX-MS (hydrogen / deuterium exchange-mass spectrometry) technique. HDX-MS analysis techniques are described in Houde D, Engen JR (2013) Methods Mol. Biol. 988: 269-89 and Houde et al. (2011) J. Pharm. Sci. 100 (6), 2071.
재조합 hTie2-Ig3-FNIII(1-3) 단백질을 사용하여 항체(3H7)의 결합-에피토프를 분석하였다. 중수소 표지 반응 전에 hTie2-Ig3-FNIII(1-3)/항체 혼합물을 3시간 이상 반응하여 15배로 희석한 중수소 표지 버퍼(KD=25nM)하에서 최대 결합상태(100%)가 유지되도록 하였다. 제조된 hTie2-Ig3-FNIII(1-3)/항체 복합체를 중수소 표지 버퍼로 15배 희석시켜, 여러 번 표지를 진행시킨 다음, 동일한 부피의 quenching buffer를 이용해 quenching시켰다. 표지 반응 시간은 각각 0분(무중수소, undeuterium), 0.33분, 10분, 60분 및 240분이었다. 그러나 무중수소 조건에서는, 중수소 표지 버퍼는 평형 버퍼(equilibrium buffer)로 대체되었고 반응은 quenching buffer로 즉시 중단시켰다. 질량 분석(mass spectrometry)을 하기 위해, 중수소가 표지된 hTie2-Ig3-FNIII(1-3)/항체 샘플을 펩신 컬럼(pepsin column)에 로딩하고, 펩타이드 digestion을 수행하였다. 질량 분석 결과 총 50개의 펩타이드(peptic peptide)로부터 82.6% 커버 데이터(coverage data)를 수득하였다.Binding-epitope of antibody (3H7) was analyzed using recombinant hTie2-Ig3-FNIII (1-3) protein. The hTie2-Ig3-FNIII (1-3) / antibody mixture was reacted for at least 3 hours prior to the deuterium labeling reaction to maintain the maximum binding state (100%) in 15-fold diluted deuterium labeling buffer (K D = 25 nM). The prepared hTie2-Ig3-FNIII (1-3) / antibody complex was diluted 15-fold with deuterium labeling buffer, the labeling proceeded several times, and quenched using the same volume of quenching buffer. Labeling reaction times were 0 minutes (undeuterium), 0.33 minutes, 10 minutes, 60 minutes and 240 minutes, respectively. However, in deuterium conditions, deuterium labeling buffer was replaced with equilibrium buffer and the reaction was immediately stopped with quenching buffer. To perform mass spectrometry, deuterium-labeled hTie2-Ig3-FNIII (1-3) / antibody samples were loaded into a pepsin column and peptide digestion was performed. Mass spectrometry yielded 82.6% coverage data from a total of 50 peptides.
hTie2-Ig3-FNIII(1-3) 와 hTie2-Ig3-FNIII(1-3)/항체 복합체 조건 사이의 중수소 흡수 차이를 비교하여 분석하였으며, 중수소의 흡수가 뚜렷하게 감소하는 영역은 항체가 직접 결합하는 부위 또는 구조적으로 변경된 부위에 해당한다. hTie2-Ig3-FNIII(1-3)와 hTie2-Ig3-FNIII(1-3)/항체 복합체 간의 중수소 흡수 차이가 0.5-1 Da 혹은 그 이상일 때에 유의미한 것으로 간주하여, 표 13에 굵은 글씨로 표시하였다.The differences in deuterium uptake between hTie2-Ig3-FNIII (1-3) and hTie2-Ig3-FNIII (1-3) / antibody complex conditions were analyzed. Corresponds to site or structurally altered site. The deuterium uptake difference between hTie2-Ig3-FNIII (1-3) and hTie2-Ig3-FNIII (1-3) / antibody complex is considered significant when 0.5-1 Da or more, and is shown in bold in Table 13 .
항-Tie2 항체(3H7)의 존재 또는 부재하에 H/D 교환-질량 분석 데이터(도 5)를 사용하여 hTie2 항원에 대해 0.333, 10, 60 및 240분에서 잔기당 중수소 흡수에 대한 히트맵을 작성하였다. 컬러 스케일은 각각의 개별적 시간에서 항원 단독과 항원/단일클론항체의 혼합물 사이의 잔기당 H/D 교환 백분율을 나타낸다. 붉은색은 중수소 교환이 증가한 영역을 나타내고 푸른색은 흡수가 없음을 나타낸다. 중수소 흡수 차이에 대한 히트맵 분석 결과, 항체(3H7)가 결합하는 에피토프가 hTie2의 633 내지 644 잔기 (서열번호 1, TLSDILPPQPEN) 및 713 내지 726 잔기 (서열번호 2, FAENNIGSSNPAFS)인 것을 나타내었다(도 13). 에피토프는 PyMol 소프트웨어를 사용하여 생성된 hTie2-FNIII 3D 구조에서 붉은색으로 나타내었다(도 6).H / D exchange-mass spectrometry data (FIG. 5) in the presence or absence of anti-Tie2 antibody (3H7) was used to create a heatmap for deuterium uptake per residue at 0.333, 10, 60 and 240 minutes for hTie2 antigen. It was. The color scale represents the percentage of H / D exchange per residue between the antigen alone and the mixture of antigen / monoclonal antibodies at each individual time. Red color indicates increased deuterium exchange and blue color indicates no absorption. Heatmap analysis of the difference in deuterium uptake indicated that the epitope to which the antibody (3H7) binds were 633 to 644 residues (SEQ ID NO: 1, TLSDILPPQPEN) and 713 to 726 residues (SEQ ID NO: 2, FAENNIGSSNPAFS) of hTie2 (Fig. 13). Epitopes are shown in red in the hTie2-FNIII 3D structure generated using PyMol software (FIG. 6).
Figure PCTKR2019006820-appb-T000013
Figure PCTKR2019006820-appb-T000013
Figure PCTKR2019006820-appb-I000002
Figure PCTKR2019006820-appb-I000002
실시예 4. 마우스 항-Tie2 항체의 인간화 및 전장(full-length) IgG 전환Example 4. Humanization and Full-length IgG Conversion of Mouse Anti-Tie2 Antibodies
인간에게 투여하는 경우 마우스 항-Tie2 항체(3H7)의 면역원성을 제거하기 위해 항체를 하기와 같은 방법으로 인간화시켰다.The antibody was humanized in the following manner to remove the immunogenicity of the mouse anti-Tie2 antibody (3H7) when administered to humans.
4.1. 중쇄 인간화4.1. Heavy chain humanization
인간 항체 중쇄가변 유전자 IGHV1-46-01은 항체 3H7의 중쇄 서열과 66%의 상동성을 보였다. 이러한 분석에 기초하여, 3H7 항체의 3개 CDR 영역을 인간 항체 중쇄 가변 유전자 IGHV1-46-01에 이식하였다. 이 과정에서, 2개의 인간화 중쇄 항체 유전자가 설계되었다(표 14). 이식된 CDR들은 단백질 서열에서 밑줄로 표시하였다. 인간화 3H7의 중쇄 유전자에 마우스 서열로의 회귀 돌연변이(back mutation)가 도입되었으며, 표 14의 단백질 서열에서 굵게 표시하였다.The human antibody heavy chain variable gene IGHV1-46-01 showed 66% homology with the heavy chain sequence of antibody 3H7. Based on this analysis, three CDR regions of the 3H7 antibody were implanted into the human antibody heavy chain variable gene IGHV1-46-01. In this process, two humanized heavy chain antibody genes were designed (Table 14). Transplanted CDRs are underlined in the protein sequence. A back mutation to the mouse sequence was introduced into the heavy chain gene of humanized 3H7, shown in bold in the protein sequences of Table 14.
4.2. 경쇄 인간화4.2. Light chain humanization
인간 항체 경쇄 가변 유전자인 IGKV1-17-01은 항체 3H7의 경쇄 서열과 68%의 상동성을 보였다. 이러한 분석에 기초하여, 3H7 항체의 3개의 CDR 영역을 인간 항체 경쇄 가변 유전자 IGKV1-17-01에 이식하였다. 이 과정에서, 2개의 인간화 경쇄 항체 유전자가 설계되었다(표14). 이식된 CDR들은 단백질 서열에서 밑줄로 표시하였다. 인간화 3H7의 경쇄 유전자에 마우스 서열로의 역 돌연변이(back mutation)가 도입되었으며, 표 14의 단백질 서열에서 굵게 표시하였다.IGKV1-17-01, a human antibody light chain variable gene, showed 68% homology with the light chain sequence of antibody 3H7. Based on this analysis, three CDR regions of the 3H7 antibody were implanted into the human antibody light chain variable gene IGKV1-17-01. In this process, two humanized light chain antibody genes were designed (Table 14). Transplanted CDRs are underlined in the protein sequence. A back mutation into the mouse sequence was introduced into the light chain gene of humanized 3H7, shown in bold in the protein sequences of Table 14.
4.3. 인간화 유전자의 합성 및 인간 전장(full-length) IgG 항체로의 클로닝4.3. Synthesis of humanized genes and cloning into human full-length IgG antibodies
표 14에 기재된 항체의 인간화 가변영역을 인간 IgG4 이소타입 백본 벡터(human IgG4 isotype backbone vector)의 중쇄 벡터 및 경쇄 벡터에 클로닝 하였다. 항체의 인간화 중쇄 가변영역(VH)의 DNA 단편을 ‘EcoRI-signal sequence-VH-NheI-CH-XhoI’서열로 합성하였다(Bioneer, Inc.). 항체의 인간화 경쇄 가변영역(VL)의 DNA 단편 또한 ‘EcoRI-signal sequence-VL-BsiWI-CL-XhoI’서열로 합성하였다. 중쇄와 경쇄를 코딩하고 있는 DNA 단편은 각각 pOptiVEC™ 또는 pcDNA™3.3 벡터에 클로닝 되었다.The humanized variable regions of the antibodies listed in Table 14 were cloned into the heavy and light chain vectors of the human IgG4 isotype backbone vector. DNA fragments of the humanized heavy chain variable region (VH) of the antibody were synthesized into the sequence 'EcoRI-signal sequence-VH-NheI-CH-XhoI' (Bioneer, Inc.). DNA fragments of the humanized light chain variable region (VL) of the antibody were also synthesized into the sequence 'EcoRI-signal sequence-VL-BsiWI-CL-XhoI'. The DNA fragments encoding the heavy and light chains were cloned into pOptiVEC ™ or pcDNA ™ 3.3 vectors, respectively.
Figure PCTKR2019006820-appb-T000014
Figure PCTKR2019006820-appb-T000014
Figure PCTKR2019006820-appb-I000003
Figure PCTKR2019006820-appb-I000003
Figure PCTKR2019006820-appb-I000004
Figure PCTKR2019006820-appb-I000004
4.4. 인간화 항-Tie2 항체의 제조 및 정제4.4. Preparation and Purification of Humanized Anti-Tie2 Antibodies
인간화된 Tie2항체를 생산하기 위해, 고효율의 재조합 단백질을 생산할 수 있는 Expi293F(Gibco)를 사용하였다. Expi293F 세포(2x106 cells/ml)를 Erlenmeyer 플라스크에서 배양하고 중쇄 및 경쇄를 코딩하는 플라스미드를 ExpiFectamine 293 형질 전환 키트를 이용하여 Expi293F 세포에 co-transfection 시켰다. 그 다음 쉐이킹 인큐베이터(orbital shaker, 125rpm)에서 37℃, 8% CO2 조건으로 5일 동안 세포를 배양하였다. 생성된 세포 배양 배지를 수집하고, 원심분리하여 세포를 제거하였다. 분비된 항체를 함유한 배양 상등액을 분리하고 4℃에서 보관하거나 어피니티 컬럼(Protein A agarose column, GE Healthcare)이 장착된 AKTA 정제 시스템(GE Healthcare)을 사용하여 즉시 정제하였다. 정제된 항체를 단백질 원심분리 필터(Amicon)를 통과시켜 농축하면서 항체 버퍼를 PBS로 대체하였다.To produce humanized Tie2 antibodies, Expi293F (Gibco) was used, which can produce highly efficient recombinant proteins. Expi293F cells (2x10 6 cells / ml) were cultured in Erlenmeyer flasks and plasmids encoding heavy and light chains were co-transfected into Expi293F cells using the ExpiFectamine 293 transformation kit. Cells were then incubated for 5 days at 37 ° C., 8% CO 2 conditions in a shaking incubator (orbital shaker, 125 rpm). The resulting cell culture medium was collected and centrifuged to remove the cells. Culture supernatants containing secreted antibodies were isolated and stored at 4 ° C. or immediately purified using an AKTA purification system (GE Healthcare) equipped with Protein A agarose column (GE Healthcare). Purified antibody was concentrated through a protein centrifuge filter (Amicon) to replace antibody buffer with PBS.
실시예 5. hTie2에 대한 인간화 항-Tie2항체의 친화도 측정Example 5. Determination of the Affinity of Humanized Anti-Tie2 Antibodies for hTie2
Black 96-well plates(96 well F-type black plates, Greiner)가 사용된 Octet 시스템(ForteBio)을 사용하여, hTie2에 대한 인간화 Tie2항체의 친화도를 측정하였다. 친화도 측정에 사용된 바이오센서는 AR2G tip(ForteBio Octet)으로 측정하기 전에 10분 동안 하이드레이션(hydration)하였다. 하이드레이션 이후에 인간화 항-Tie2 항체를 pH6.0 버퍼인 10mM 소듐 아세테이트에서 10μg/ml 농도로 희석시키고, AR2G 바이오센서에 고정시켰으며, 1M의 에탄올아민으로 블로킹시켰다. 재조합 hTie2를 1x키네틱 버퍼(1x kinetic buffer)를 이용하여 50, 25, 12.5, 6.25, 3.125 및 0nM로 희석하고, 300초 동안 결합시키고 900초 동안 해리시켰다. 친화력 측정(KD)을 위해 K-on(association rate) 및 K-off(dissociation rate)를 결합 곡선(global)으로 분석하고 옥텟 데이터 분석 프로그램(v9.0.0.10)을 사용하여 1:1 결합 모델에 맞추었다. KD 값은 하기 표 15에 나타내었다.The affinity of the humanized Tie2 antibody for hTie2 was measured using Octet system (ForteBio) using black 96-well plates (96 well F-type black plates, Greiner). Biosensors used for affinity measurements were hydrated for 10 minutes before being measured with an AR2G tip (ForteBio Octet). After hydration the humanized anti-Tie2 antibody was diluted to 10 μg / ml concentration in 10 mM sodium acetate, pH6.0 buffer, fixed in AR2G biosensor and blocked with 1 M ethanolamine. Recombinant hTie2 was diluted to 50, 25, 12.5, 6.25, 3.125 and 0 nM using 1 × kinetic buffer, bound for 300 seconds and dissociated for 900 seconds. Analyze K-on (association rate) and K-off (dissociation rate) globally for affinity measurements (KD) and use 1: 1 occlusion model using octet data analysis program (v9.0.0.10) To fit. KD values are shown in Table 15 below.
Figure PCTKR2019006820-appb-T000015
Figure PCTKR2019006820-appb-T000015
실시예 6. 선별된 인간화 항-Tie2 항체의 in vitro 생물학적 특성 분석Example 6 In vitro Biological Characterization of Selected Humanized Anti-Tie2 Antibodies
6.1. Akt 인산화6.1. Akt phosphorylation
인간화 항-Tie2 항체가 혈관내피세포 중 Tie2 수용체의 하위 신호 전달을 유도하는지 조사하기 위해, HUVEC를 인간화 Tie2 항체로 처리하였다. 이후, Tie2 수용체의 주요 하위 신호 전달 단백질인 Akt 인산화의 수준을 immunoblotting 기법으로 측정하였다. Akt 활성화 정도를 비교하기 위해, COMP-Ang1(CA1)을 양성 대조군으로 사용하였다. 구체적으로, 60mm 배양 접시 내 EGM-2 배지(Lonza)에서 HUVEC 세포(1x105 cells/ml)를 37℃에서 배양하였다. 90% 밀집도(confluency)의 세포를 EBM-2(Lonza)와 함께 4시간 동안 배양하였다. 혈청 기아 상태(serum-starved)의 HUVEC에 항-Tie2항체를 처리하고 30분 동안 추가 배양하였다. 세포를 차가운 PBS로 세척한 뒤, 용해 버퍼를 처리하고 4℃에서 20분 동안 용해시켰다. 이어서, 13000rpm에서 15분 동안 원심 분리하여 세포 용해물을 제조하였다. 5xSDS 샘플 버퍼를 세포 용해물에 첨가하고 혼합물을 95℃에서 5분 가열하였다. 그 다음 혼합물을 SDS-PAGE시킨 후 웨스턴블롯 하였다.To investigate whether the humanized anti-Tie2 antibody induces downstream signal transduction of the Tie2 receptor in vascular endothelial cells, HUVECs were treated with humanized Tie2 antibodies. Thereafter, the level of Akt phosphorylation, a major downstream signaling protein of the Tie2 receptor, was measured by immunoblotting. To compare the level of Akt activation, COMP-Ang1 (CA1) was used as a positive control. Specifically, HUVEC cells (1 × 10 5 cells / ml) were incubated at 37 ° C. in EGM-2 medium (Lonza) in a 60 mm culture dish. 90% confluency cells were incubated with EBM-2 (Lonza) for 4 hours. Serum-starved HUVECs were treated with anti-Tie2 antibodies and further incubated for 30 minutes. The cells were washed with cold PBS, then treated with lysis buffer and lysed at 4 ° C. for 20 minutes. Cell lysates were then prepared by centrifugation at 13000 rpm for 15 minutes. 5 × SDS sample buffer was added to the cell lysate and the mixture was heated at 95 ° C. for 5 minutes. The mixture was then SDS-PAGE followed by Western blot.
Akt의 인산화를 조사하기 위해, 막을 5% skim milk 함유 TBST를 사용하여 실온에서 1시간동안 블로킹시키고, p-Akt 항체 (S473)와 함께 4℃에서 약 8시간 동안 배양하였다. p-Akt의 양은 ECL (Enhanced ChemiLuminescence)에 의해 검출 되었다. 이 후 막을 15분동안 스트리핑 버퍼(Thermo)에서 15분간 인큐베이션한 다음, 항-Akt 항체로 다시 프로빙하여 총 Akt의 양을 결정하였다. To investigate the phosphorylation of Akt, the membrane was blocked for 1 hour at room temperature using TBST containing 5% skim milk and incubated with p-Akt antibody (S473) at 4 ° C. for about 8 hours. The amount of p-Akt was detected by ECL (Enhanced ChemiLuminescence). The membrane was then incubated for 15 minutes in stripping buffer (Thermo) and then probed again with anti-Akt antibody to determine the total amount of Akt.
도 7에 도시된 바와 같이, Akt 인산화는 인간화 3H7 항체의 처리에 의해 현저히 증가하였다. 이러한 데이터는 인간화 항-Tie2 항체가 혈관내피세포에서 Tie2 수용체의 하위 신호전달 분자인 Akt의 활성화를 강력하게 유도할 수 있음을 나타낸다.As shown in FIG. 7, Akt phosphorylation was significantly increased by treatment of humanized 3H7 antibody. These data indicate that humanized anti-Tie2 antibodies can strongly induce the activation of Akt, a downstream signaling molecule of the Tie2 receptor in vascular endothelial cells.
실시예 7. 원발성 개방각 녹내장(primary open-angle glaucoma) 마우스 모델에서 3H7H12G4의 효과Example 7 Effect of 3H7H12G4 in Primary Open-angle Glaucoma Mouse Model
3H7H12G4에 의한 Tie2 활성화가 퇴화된 쉴렘관(Schlemm`s Canal)을 회복하고 안압(IoP)을 낮출 수 있는지 여부를 테스트하기 위해, 원발 개방각 녹내장 마우스 모델을 사용하였다. Angiopoietin-1/-2 이중 유전자의 유도성 결실을 위해, 8주령의 Angiopoietin-1/Angiopoietin-2 결핍(A1:A2 iΔ/Δ) 마우스에 tamoxifen을 투여하여 모델을 제작하였다(도 8A). 12주령에 3H7H12G4(~5μg, 한쪽 눈) 및 FC(~5μg, 다른쪽 눈)를 안구 내 투여하였다(도 8A). 5 μg이 함유된 각 시약(~1 μl, 5mg/ml)를 유리 모세관 피펫(glass capillary pipette)이 장착된 Nanoliter 2000 micro-injector(World Precision Instruments)를 사용하여 유리체강에 주사되었다. IoP는 12, 13 및 14주령에 rebound tonometer(TonoLab)을 사용하여 측정되었다(도 8A). IoP는 마우스를 마취한 직후 각막 중심으로부터 약 1/8인치 정도 위치에 압력 센서의 끝을 위치시켜 측정하였다. 5번의 연속적 안압 측정의 디지털 판독값은 tonometer에서 수득하였다. 야생형 마우스에서는 3H7H12G4가 처리된 눈과 Fc가 처리된 눈 사이에 안압 차이가 없었다(도 8C). 반면, A1:A2 iΔ/Δ 마우스는 3H7H12G4를 처리한 눈에서 Fc를 처리한 눈과 비교하여 25.6%의 유의미한 IoP 감소를 보였다(도8D). CD144+SC 영역 및 CD144+SC에서 Prox1 및 Tie2 면역염색의 강도는 3H7H12G4투여 2주 뒤에 측정되었다. 항-CD144항체(1:200, BD Biosciences), 항-Prox1 항체(1:200, Relia Tech) 및 항-Tie2 항체(1:200, R&D systems)가 사용되었다. 등재된 각막 전체의 CD144+SC 영역은 CD144+SC 영역을 대조군 영역으로 나눈 백분율로 계산하였다. A1:A2 iΔ/Δ 마우스에서 3H7H12G4로 처리한 눈의 SC 영역이 FC로 처리한 눈의 SC 영역과 비교하여 114.8% 증가한 것을 나타내었다(도 8B, 8E). Prox1의 상대적인 발현을 정량하기 위해, CD144+SC의 핵 영역에서의 강도를 측정하였다. A1:A2 iΔ/Δ 마우스는 3H7H12G4로 처리한 눈에서 Prox1 강도가 FC로 처리한 눈에서의 Prox1 강도와 비교하여 88.4%의 유의미한 증가를 보였다(도 8B, 8F). Tie2의 발현을 정량하기 위해 CD144+SC 영역에서의 강도를 측정하였다. A1:A2 iΔ/Δ 마우스는 3H7H12G4로 처리한 눈의 Tie2 강도가 FC로 처리한 눈의 Tie2 강도와 비교하여 63.0%의 유의미한 증가를 보였다(도 8B, 8F). 전반적으로, 이러한 결과는 3H7H12G4에 의한 Tie2 활성화가 A1:A2 iΔ/Δ 마우스의 손상된 SC를 회복시킨다는 것을 나타낸다.To test whether Tie2 activation by 3H7H12G4 can restore the degraded Schlemm's Canal and lower the intraocular pressure (IoP), a primary open angle glaucoma mouse model was used. For inducible deletion of the Angiopoietin-1 / -2 double gene, a model was constructed by administering tamoxifen to 8-week-old Angiopoietin-1 / Angiopoietin-2 deficient ( A1: A2 iΔ / Δ ) mice (FIG. 8A). At 12 weeks of age, 3H7H12G4 (˜5 μg, one eye) and FC (˜5 μg, the other eye) were administered intraocularly (FIG. 8A). Each reagent containing 5 μg (˜1 μl, 5 mg / ml) was injected into the vitreous cavity using a Nanoliter 2000 micro-injector (World Precision Instruments) equipped with a glass capillary pipette. IoP was measured using a rebound tonometer (TonoLab) at 12, 13 and 14 weeks of age (FIG. 8A). IoP was measured by placing the tip of the pressure sensor about 1/8 inch from the center of the cornea immediately after anesthetizing the mouse. Digital readings of five consecutive intraocular pressure measurements were obtained on a tonometer. In wild-type mice, there was no intraocular pressure difference between eyes treated with 3H7H12G4 and eyes treated with Fc (FIG. 8C). On the other hand, A1: A2 iΔ / Δ mice showed a significant IoP reduction of 25.6% in eyes treated with 3H7H12G4 compared to eyes treated with Fc (FIG. 8D). Intensity of Prox1 and Tie2 immunostaining in the CD144 + SC region and CD144 + SC was measured 2 weeks after 3H7H12G4 administration. Anti-CD144 antibody (1: 200, BD Biosciences), anti-Prox1 antibody (1: 200, Relia Tech) and anti-Tie2 antibody (1: 200, R & D systems) were used. The CD144 + SC region of the entire cornea listed was calculated as a percentage of the CD144 + SC region divided by the control region. In the A1: A2 iΔ / Δ mice, the SC area of the eye treated with 3H7H12G4 was increased by 114.8% compared to the SC area of the eye treated with FC (FIGS. 8B and 8E). To quantify the relative expression of Prox1, the intensity in the nuclear region of CD144 + SC was measured. A1: A2 iΔ / Δ mice showed a significant increase of Prox1 intensity in eyes treated with 3H7H12G4 compared with Prox1 intensity in eyes treated with FC (88.4%) (FIGS. 8B, 8F). The intensity in the CD144 + SC region was measured to quantify the expression of Tie2. A1: A2 iΔ / Δ mice showed a significant increase of 63.0% in the Tie2 intensity of eyes treated with 3H7H12G4 compared to the Tie2 intensity of eyes treated with FC (FIGS. 8B, 8F). Overall, these results indicate that Tie2 activation by 3H7H12G4 restores impaired SC in A1: A2 iΔ / Δ mice.
실시예 8. 레이저 유도 맥락막 신생 혈관(CNV) 모델에서 유리체강내 3H7H12G4 주입에 의한 CNV 퇴화 및 혈관 누출 억제Example 8 Inhibition of CNV Degeneration and Vascular Leakage by Intravitreal 3H7H12G4 Injection in Laser-Induced Choroidal Neovascularization (CNV) Model
레이저 유도 CNV모델을 사용하여, 습식 노인성 황반 변성(wet age-related macular degeneration, AMD)의 특징인 맥락막 혈관 신생(CNV)에 대한 3H7H12G4의 억제 능력에 대해 시험하였다. 5mg/ml 페닐에프린(phenylephrine) 및 5mg/ml tropicamide eye drops(Santen pharmaceutical)로 동공을 확장하고 국소 마취를 위해 0.5% proparacaine hydrochloride eye drops(Alcon)을 적하하였으며, 슬릿 램프 전달 시스템(slit lamp delivery system)을 포함하는 Laser photocoagulator(Lumenis Inc.)와 contact lens로서 유리 커버슬립을 함께 사용하여 망막을 시각화 하였다. 각 눈의 4개 지역(posterior pole의 3, 6, 9, 12시 위치)에 충분한 레이저 에너지(532nm 파장, 250mW 전원, 100ms 지속, 50μm spot 크기)를 전달하였다. 이 연구에는 오직 레이저 광응고 당시 버블을 형성한 화상만이 포함되어 있으며, 이는 Bruch’s membrane이 파괴되었음을 나타낸다. 레이저 조사 부위에서 출혈을 포함하는 스팟은 분석에서 제외하였다. 임상적 상황을 요약하자면, 레이저 광응고 후 7일째에 3H7H12G4(5μg)를 마우스의 유리체강 내에 투여하였다(도 9A). 대조군으로서 또는 비교 분석을 위해, Fc 또는 VEGF-Trap(5μg)을 마우스에게 동일한 방식으로 투여하였다. 해당 시약을 투여하기 위해, 유리 모세관 피펫(glass capillary pipette)이 장착된 Nanoliter 2000 micro-injector(World Precision Instruments)를 사용하였고 5 μg이 함유된 각 시약(~1μl, 5mg/ml)이 유리체강에 주사되었다. 레이저 광응고 후 14일째에 망막 색소 상피(RPE)-맥락막-공막 평면 기저(mounts)에 대한 CD31+ CNV 부피는 MATLAB image processing toolbox(MathWorks)를 사용하여 계산되었다. CNV의 내피세포를 검출하는데 CD31 항체(1:200, Millipore)를 사용하였다. VEGF-Trap은 Fc와 비교하여 CNV 퇴화를 64.4%까지 효과적으로 유도하였고, 3H7H12G4도 비슷하게 CNV 퇴화를 유도하였다(65.7%)(도 9B). FA (Fluorescein angiography) 및 ICGA(indocyanine green angiography)을 결합하여 레이저 손상 부위 주변의 신생 혈관에서의 혈관 누출을 측정할 수 있었다. 488nm 및 755nm의 연속파(continuous-wave) 레이저 모듈은 각각 Fluorescein 과 ICG의 자극원으로 사용되었다. 자극 레이저의 래스터 스캐닝 패턴(raster scanning pattern)은 rotating polygonal mirror(MC-5; Lincoln Laser)와 galvanometer-based scanning mirror(6230H; Cambridge technology)로 구성된 스캐너 시스템으로 이루어졌고, 이미징 렌즈의 후방 조리개로 전달되었다. 높은 수치의 개구수(numerical aperture, NA)를 갖는 대물렌즈(PlanApo λ, NA 0.75; Nikon)가 광역 안저 형광 이미지(wide-field fundus fluorescence images)를 위해 이미징 렌즈로 사용되었다. 광전증폭관(R9110; Hamamatsu Photonics)으로 검출된 형광 신호를 프레임 그래버(frame grabber)로 디지털화 하고 프레임당 512x512픽셀 크기의 이미지로 재구성하였다. 혈관 조영술을 이용하여 후기 상(late-phase: 6분)의 FA 및 ICGA 이미지를 시각화하기 위해, 10mg의 fluorescein sodium(Alcon) 및 0.15mg의 ICG(Daiichi Pharmaceutical)를 각각 복강 내 및 정맥 내에 투여하였다. 이미징 절차는 전신 마취 및 동공 확장 상태로 수행되어 이미지의 품질을 향상시켰다. Java 기반 이미징 소프트웨어(ImajeJ; National Institutes of Health)를 사용하여 FA 이미지에서 측정된 하이퍼 형광영역의 총계를 ICGA이미지에서 측정된 CNV 지역으로 나누어 CNV의 누출 부위에 대해 계산하였다. Fc와 비교하여, VEGF-Trap(37.0%)과 3H7H12G4(24.6%)는 유사하게 혈관 누출을 억제하였다(도 9C). 특히, Fc 처리 그룹은 레이저 광응고 후 6일에서 14일 사이에 혈관 누출에 큰 차이를 보이지 않았으나, VEGF-Trap 및 3H7H12G4는 혈관 누출을 현저히 감소시켰다(각각 45.6% 및 42.5%). 따라서, 레이저 유도 CNV의 마우스 모델에서 VEGF-Trap 과 3H7H12G4 사이의 CNV 및 혈관 누출의 억제 규모는 정량적으로 차이가 없었다.A laser induced CNV model was used to test the ability of 3H7H12G4 to suppress choroidal neovascularization (CNV), which is characteristic of wet age-related macular degeneration (AMD). Dilated pupils with 5mg / ml phenylephrine and 5mg / ml tropicamide eye drops (Santen pharmaceutical) and drop 0.5% proparacaine hydrochloride eye drops (Alcon) for local anesthesia, and slit lamp delivery The laser photocoagulator (Lumenis Inc.) and glass coverslips were used as contact lenses to visualize the retina. Sufficient laser energy (532nm wavelength, 250mW power, 100ms duration, 50μm spot size) was delivered to four areas of each eye (positions 3, 6, 9, and 12 o'clock of the posterior pole). The study only included bubbles that formed during laser photocoagulation, indicating that the Bruch's membrane was destroyed. Spots containing bleeding at the laser irradiation site were excluded from the analysis. In summary, 3H7H12G4 (5 μg) was administered into the vitreous cavity of mice 7 days after laser photocoagulation (FIG. 9A). As a control or for comparative analysis, Fc or VEGF-Trap (5 μg) was administered to mice in the same manner. To administer the reagents, a Nanoliter 2000 micro-injector (World Precision Instruments) equipped with a glass capillary pipette was used and each reagent containing 5 μg (~ 1 μl, 5 mg / ml) was added to the vitreous cavity. Injection. At 14 days after laser photocoagulation, CD31 + CNV volumes for the retinal pigment epithelium (RPE) -choroid-scleral planar mounts were calculated using the MATLAB image processing toolbox (MathWorks). CD31 antibody (1: 200, Millipore) was used to detect endothelial cells of CNV. VEGF-Trap effectively induced CNV degeneration by 64.4% compared to Fc, and 3H7H12G4 similarly induced CNV degeneration (65.7%) (FIG. 9B). Fluorescein angiography (FA) and indocyanine green angiography (ICGA) were combined to measure vascular leakage in neovascularization around the area of laser injury. Continuous-wave laser modules of 488nm and 755nm were used as stimulators of Fluorescein and ICG, respectively. The raster scanning pattern of the stimulus laser consists of a scanner system consisting of a rotating polygonal mirror (MC-5; Lincoln Laser) and a galvanometer-based scanning mirror (6230H; Cambridge technology), which is transmitted to the rear aperture of the imaging lens. It became. An objective lens (PlanApo λ, NA 0.75; Nikon) with a high numerical aperture (NA) was used as the imaging lens for wide-field fundus fluorescence images. Fluorescence signals detected with a photoamplifier tube (R9110; Hamamatsu Photonics) were digitized with a frame grabber and reconstructed into an image of 512x512 pixels per frame. In order to visualize late-phase FA and ICGA images using angiography, 10 mg of fluorescein sodium (Alcon) and 0.15 mg of Daiichi Pharmaceutical (ICG) were administered intraperitoneally and intravenously, respectively. . Imaging procedures were performed with general anesthesia and pupil dilation to improve the quality of the image. Java-based imaging software (ImajeJ; National Institutes of Health) was used to calculate the leakage area of CNV by dividing the total amount of hyperfluorescence measured in FA images by the CNV region measured in ICGA images. Compared to Fc, VEGF-Trap (37.0%) and 3H7H12G4 (24.6%) similarly inhibited vascular leakage (FIG. 9C). In particular, the Fc treated group showed no significant difference in vascular leakage between 6 and 14 days after laser photocoagulation, but VEGF-Trap and 3H7H12G4 significantly reduced vascular leakage (45.6% and 42.5%, respectively). Therefore, there was no quantitative difference in the magnitude of inhibition of CNV and vascular leakage between VEGF-Trap and 3H7H12G4 in a mouse model of laser induced CNV.
실시예 9. CNV 내피세포에서 3H7H12G4 및 CD31의 공동 국소화(co-localization).Example 9 Co-localization of 3H7H12G4 and CD31 in CNV Endothelial Cells.
3H7H12G4의 피하주사 또한 CNV에 대한 치료효과를 나타낼 수 있는지 여부를 조사하기 위해, CNV의 내피세포에서 3H7H12G4 및 CD31의 공동 국소화를 평가하였다. 레이저 광응고 1일 뒤에 3H7H12G4(25mg/kg)를 피하투여 하였다. 대조군으로 Fc(25mg/kg)를 마우스에게 동일한 방식으로 투여하였다. 레이저 광응고 2일, 4일 및 8일 후에, 항-인간 IgG 항체(1:1000, Jackson ImmunoResearch Laboratories)를 통해 CNV의 내피세포에서 3H7H12G4 및 CD31항체의 공동 국소화를 직접 검출하였다(도 10A). 투여된 3H7H12G4는 CD31+ 내피세포 CNV에서 높은 정도로 검출 가능하였다(도 10B 내지 10D).To investigate whether subcutaneous injection of 3H7H12G4 could also have a therapeutic effect on CNV, co-localization of 3H7H12G4 and CD31 in CNV endothelial cells was evaluated. One day after laser photocoagulation, 3H7H12G4 (25 mg / kg) was subcutaneously administered. Fc (25 mg / kg) was administered to mice in the same manner as a control. Two, four and eight days after laser photocoagulation, co-localization of 3H7H12G4 and CD31 antibodies was directly detected in endothelial cells of CNV via anti-human IgG antibody (1: 1000, Jackson ImmunoResearch Laboratories) (FIG. 10A). The administered 3H7H12G4 was detectable to a high extent in CD31 + endothelial cell CNV (FIGS. 10B-10D).
실시예 10. 3H7H12G4항체 피하 주사에 의한 CNV 억제 효과.Example 10. CNV inhibitory effect by 3H7H12G4 antibody subcutaneous injection.
CNV 억제에서 3H7H12G4 피하주사의 효과를 측정하기 위해 레이저 광응고 1일 뒤에 3H7H12G4(25mg/kg)의 피하 투여를 수행하였다. 대조군으로 Fc(25mg/kg)를 마우스에게 동일한 방식으로 투여하였다. CNV의 내피세포에서 검출하기 위해 항-CD31 항체(1:200, Millipore)를 사용하였고, 레이저 광응고 8일째에 망막 색소 상피(RPE)-맥락막-공막 평면 기저(mounts)에 대한 CD31+ CNV 부피는 MATLAB image processing toolbox(MathWorks)를 사용하여 계산되었다(도 11A). 3H7H12G4는 Fc에 비해 CNV의 생성을 69.9% 로 효과적으로 억제하였으며(도 11B, 11C), 이는 유리체강내 주사뿐 아니라 3H7H12G4의 피하주사 또한 CNV의 억제효과가 있음을 나타낸다.Subcutaneous administration of 3H7H12G4 (25 mg / kg) was performed one day after laser photocoagulation to determine the effect of 3H7H12G4 subcutaneous injection on CNV inhibition. Fc (25 mg / kg) was administered to mice in the same manner as a control. Anti-CD31 antibody (1: 200, Millipore) was used for detection in CNV endothelial cells and CD31 + CNV volume for retinal pigment epithelium (RPE) -choroid-scleral plane mounts on day 8 laser photocoagulation Was calculated using the MATLAB image processing toolbox (MathWorks) (FIG. 11A). 3H7H12G4 effectively inhibited CNV production by 69.9% compared to Fc (FIGS. 11B, 11C), indicating that not only intravitreal injection but also subcutaneous injection of 3H7H12G4 has an inhibitory effect on CNV.
본 발명에 따른 Tie2에 결합하는 항체 또는 이의 항원 결합단편은 Tie2에 높은 친화력으로 결합하면서도, 인간과 마우스에 대한 교차반응성을 유지하며, 목적하는 항원 반응성을 나타낸다. 또한, Tie2 항체가 Tie2의 인산화 및 Tie2 수용체의 활성화를 유도함으로써, 목적하는 혈관신생 질환의 예방 또는 치료에 유용하게 사용될 수 있다.The antibody or antigen-binding fragment thereof that binds Tie2 according to the present invention binds to Tie2 with high affinity while maintaining cross-reactivity with humans and mice, and exhibits the desired antigen reactivity. In addition, the Tie2 antibody may be usefully used for the prevention or treatment of the desired angiogenic disease by inducing phosphorylation of Tie2 and activation of the Tie2 receptor.
이상으로 본 발명의 내용의 특정한 부분을 상세히 기술하였는바, 당업계의 통상의 지식을 가진 자에게 있어서, 이러한 구체적 기술은 단지 바람직한 실시양태일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백할 것이다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다고 할 것이다.As described above in detail a specific part of the content of the present invention, for those skilled in the art, such a specific description is only a preferred embodiment, which is not limited by the scope of the present invention Will be obvious. Thus, the substantial scope of the present invention will be defined by the appended claims and their equivalents.
전자파일 첨부하였음.Electronic file attached.

Claims (13)

  1. 서열번호 2의 서열을 포함하는 Tie2 Ig3-FNIII (1-3) 도메인에 결합하는 Tie2에 대한 항체 또는 이의 항원 결합단편.An antibody or antigen binding fragment thereof against Tie2 that binds to a Tie2 Ig3-FNIII (1-3) domain comprising the sequence of SEQ ID NO: 2.
  2. 제1항에 있어서, 서열번호 1의 서열을 포함하는 Tie2의 633 내지 644번째 아미노산 및/또는 713 내지 726번째 아미노산에 결합하는 항체 또는 이의 항원 결합단편.The antibody or antigen-binding fragment thereof of claim 1, wherein the antibody binds to amino acids 633 to 644 and / or 713 to 726 amino acids of Tie2 comprising the sequence of SEQ ID NO: 1.
  3. 제1항에 있어서, 서열번호 3 내지 5의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 6 내지 8의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역;According to claim 1, Heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NO: 3 to 5, Light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NO: 6 to 8;
    서열번호 13 내지 15의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 16 내지 18의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역;A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 13 to 15, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 16 to 18;
    서열번호 23 내지 25의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 26 내지 28의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역;A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 23 to 25, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 26 to 28;
    서열번호 33 내지 35의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 36 내지 38의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역; 또는A heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 33 to 35, a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NOs: 36 to 38; or
    서열번호 43 내지 45의 아미노산 서열을 포함하는 중쇄 CDR을 포함하는 중쇄 가변영역, 서열번호 46 내지 48의 아미노산 서열을 포함하는 경쇄 CDR을 포함하는 경쇄 가변영역을 포함하는, 항체 또는 이의 항원 결합단편.An antibody or antigen-binding fragment thereof, comprising a heavy chain variable region comprising a heavy chain CDR comprising the amino acid sequence of SEQ ID NOs: 43 to 45, and a light chain variable region comprising a light chain CDR comprising the amino acid sequence of SEQ ID NO: 46 to 48.
  4. 제1항에 있어서, The method of claim 1,
    서열번호 9의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 11의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 9 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 11;
    서열번호 19의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 21의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 19 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 21;
    서열번호 29의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 31의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 29 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 31;
    서열번호 39의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 41의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 39 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 41;
    서열번호 49의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 51의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 49 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 51;
    서열번호 53의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 54의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 53 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 54;
    서열번호 57의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 58의 아미노산 서열을 포함하는 중쇄 가변영역;A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 57 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 58;
    서열번호 61의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 62의 아미노산 서열을 포함하는 중쇄 가변영역; 또는A heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 61 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 62; or
    서열번호 65의 아미노산 서열을 포함하는 중쇄 가변영역 및 서열번호 66의 아미노산 서열을 포함하는 중쇄 가변영역을 포함하는, 항체 또는 이의 항원 결합단편.An antibody or antigen-binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 65 and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 66.
  5. 제 1 항 내지 제 4 항 중 어느 한 항의 항체 또는 이의 항원 결합단편을 코딩하는 핵산.A nucleic acid encoding the antibody of claim 1 or an antigen binding fragment thereof.
  6. 제5항에 있어서, 서열번호 10, 12, 20, 22, 30, 32, 40, 42, 50, 52, 55, 56, 59, 60, 63, 64, 67 또는 68의 서열을 포함하는 것을 특징으로 하는 핵산.The method of claim 5, characterized in that it comprises the sequence of SEQ ID NO: 10, 12, 20, 22, 30, 32, 40, 42, 50, 52, 55, 56, 59, 60, 63, 64, 67 or 68 Nucleic acid.
  7. 제5항의 핵산을 포함하는 발현벡터.An expression vector comprising the nucleic acid of claim 5.
  8. 제7항의 발현 벡터로 형질전환된 세포.A cell transformed with the expression vector of claim 7.
  9. 다음 단계를 포함하는 Tie2에 결합하는 항체 또는 그의 항원 결합 단편의 제조방법:A method for preparing an antibody or antigen-binding fragment thereof that binds Tie2, comprising the following steps:
    (a) 제8항의 세포를 배양하는 단계; 및(a) culturing the cells of claim 8; And
    (b) 상기 배양된 세포에서 항체 또는 그의 항원 결합 단편을 회수하는 단계.(b) recovering the antibody or antigen-binding fragment thereof from the cultured cells.
  10. 제 1 항 내지 제 4 항 중 어느 한 항의 항체 또는 그의 항원 결합 단편을 유효성분으로 포함하는 혈관신생 질환의 예방 또는 치료용 조성물.A composition for preventing or treating angiogenic diseases, comprising the antibody of claim 1 or an antigen-binding fragment thereof as an active ingredient.
  11. 제10항에 있어서, 상기 혈관신생 질환은 암, 전이(metastasis), 당뇨망막병증(diabetic retinopathy), 미숙아 망막병증(retinopathy of prematurity), 각막 이식 거부(corneal graft rejection), 황반변성 (macular degeneration), 혈관신생성녹내장(neovascular glaucoma), 전신홍색증(erythrosis), 증식성망막증(proliferative retinopathy), 건선(psoriasis), 혈우병성 고관절염(hemophilic arthritis), 동맹경화성 플라크(atherosclerotic plaques)의 모세혈관 형성, 켈로이드(keloid), 상처 과립화(wound granulation), 혈관 유착(vascular adhesion), 류마티스 관절염(rheumatoid arthritis), 퇴행성 관절염(osteoarthritis), 자기면역질환(autoimmune diseases), 크론병(Crohn's disease), 레스테노시스(restenosis), 죽상동맥경화증(atherosclerosis), 장협착(intestinal adhesions), 고양이 찰과상 감염증(cat scratch disease), 궤양(ulcer), 간경변증(liver cirrhosis), 신장염(nephritis), 당뇨병성 신장질환(diabetic nephropathy), 진성 당뇨병(diabetes mellitus), 염증 질환(inflammatory diseases) 및 신경변성 질환(neurodegenerative diseases)으로 구성된 군에서 선택된 것을 특징으로 하는 조성물.11. The method of claim 10, wherein the angiogenic disease is cancer, metastasis, diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, macular degeneration. , Neovascular glaucoma, neovascular glaucoma, erythrosis, proliferative retinopathy, psoriasis, hemophilic arthritis, capillary formation of atherosclerotic plaques, Keloid, wound granulation, vascular adhesion, rheumatoid arthritis, osteoarthritis, autoimmune diseases, Crohn's disease, lesteno Restenosis, atherosclerosis, intestinal adhesions, cat scratch disease, ulcers, liver cirrhosis, nephritis, diabetes Renal diseases (diabetic nephropathy), diabetes mellitus (diabetes mellitus), inflammatory diseases (inflammatory diseases) and neurodegenerative disorders (neurodegenerative diseases) a composition according to claim, selected from the group consisting of.
  12. 제11항에 있어서, 상기 암은 식도암(esophageal cancer), 위암(stomach cancer), 대장암(large intestine cancer), 직장암(rectal cancer), 구강암(oral cancer), 인두암(pharynx cancer), 후두암(larynx cancer), 폐암(lung cancer), 결장암(colon cancer), 유방암(breast cancer), 자궁경부암(uterine cervical cancer), 자궁내막암(endometrial cancer), 난소암(ovarian cancer), 전립선암(prostate cancer), 고환암(testis cancer), 방광암(bladder cancer), 신장암(renal cancer), 간암(liver cancer), 췌장암(pancreatic cancer), 뼈암(bone cancer), 결합조직암(connective tissue cancer), 피부암(skin cancer), 뇌종양(brain cancer), 갑상선암(thyroid cancer), 백혈병(leukemia), 호지킨 림프종(Hodgkin's lymphoma), 림프종(lymphoma) 및 다발성 골수혈액암(multiple myeloid blood cancer)으로 구성된 군에서 선택된 것을 특징으로 하는 조성물.The method of claim 11, wherein the cancer is esophageal cancer, stomach cancer, large intestine cancer, rectal cancer, oral cancer, pharynx cancer, laryngeal cancer larynx cancer, lung cancer, colon cancer, breast cancer, breast cancer, uterine cervical cancer, endometrial cancer, ovarian cancer, prostate cancer ), Testis cancer, bladder cancer, renal cancer, liver cancer, liver cancer, pancreatic cancer, bone cancer, connective tissue cancer, skin cancer ( skin cancer, brain cancer, thyroid cancer, leukemia, Hodgkin's lymphoma, lymphoma and multiple myeloid blood cancer Characterized in that the composition.
  13. 제 1 항 내지 제 4 항 중 어느 한 항의 항체 또는 그의 항원 결합 단편을 포함하는, 다른 혈관신생 질환 치료제와의 병용투여용 조성물.A composition for co-administration with another therapeutic agent for angiogenic diseases, comprising the antibody of claim 1 or an antigen-binding fragment thereof.
PCT/KR2019/006820 2018-06-07 2019-06-05 Antibody binding to tie2 and use thereof WO2019235856A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP19814472.7A EP3805265A4 (en) 2018-06-07 2019-06-05 Antibody binding to tie2 and use thereof
CA3101506A CA3101506A1 (en) 2018-06-07 2019-06-05 Anti-tie2 antibodies and use thereof
JP2020567832A JP7390317B2 (en) 2018-06-07 2019-06-05 Anti-Tie2 antibody and its use
AU2019283520A AU2019283520A1 (en) 2018-06-07 2019-06-05 Antibody binding to Tie2 and use thereof
CN201980038199.8A CN112399975A (en) 2018-06-07 2019-06-05 Antibodies that bind to Tie2 and uses thereof
US17/111,413 US20210179719A1 (en) 2018-06-07 2020-12-03 Antibody Binding To Tie2 And Use Thereof
JP2023150164A JP2023175830A (en) 2018-06-07 2023-09-15 Antibody against tie2 and use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201862682042P 2018-06-07 2018-06-07
US62/682,042 2018-06-07
KR1020190066622A KR20190139148A (en) 2018-06-07 2019-06-05 Antibodies Binding to Tie2 and Uses Thereof
KR10-2019-0066622 2019-06-05

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/111,413 Continuation US20210179719A1 (en) 2018-06-07 2020-12-03 Antibody Binding To Tie2 And Use Thereof

Publications (1)

Publication Number Publication Date
WO2019235856A1 true WO2019235856A1 (en) 2019-12-12

Family

ID=68769934

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2019/006820 WO2019235856A1 (en) 2018-06-07 2019-06-05 Antibody binding to tie2 and use thereof

Country Status (1)

Country Link
WO (1) WO2019235856A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021194913A1 (en) * 2020-03-24 2021-09-30 Genentech, Inc. Tie2-binding agents and methods of use
CN113993897A (en) * 2019-08-14 2022-01-28 药物抗体公司 anti-TIE 2 antibodies and uses thereof

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6365154B1 (en) 1998-09-28 2002-04-02 Smithkline Beecham Corporation Tie2 agonist antibodies
US7658924B2 (en) 2001-10-11 2010-02-09 Amgen Inc. Angiopoietin-2 specific binding agents
JP2011102273A (en) 2009-11-11 2011-05-26 Shiseido Co Ltd Tie2-activating agent, agent for maturing, normalizing or stabilizing blood vessel, lymphatic vessel-stabilizing agent, and wrinkle-preventing/ameliorating agent and dropsy-ameliorating/preventing agent
KR20140054303A (en) * 2011-08-19 2014-05-08 리제너론 파아마슈티컬스, 인크. Anti-tie2 antibodies and uses thereof
US8987420B2 (en) 2009-07-29 2015-03-24 Regeneron Pharmaceuticals, Inc. High affinity human antibodies to human angiopoietin-2
KR20150032075A (en) * 2013-09-17 2015-03-25 삼성전자주식회사 Anticancer composition containing an anti-Ang2 antibody inducing binding to Tie2 receptor
JP2015168656A (en) 2014-03-07 2015-09-28 丸善製薬株式会社 Tie2 activator, neovascularization inhibitor, agent for blood vessel maturation, agent for blood vessel normalization, and blood vessel stabilizer, and pharmaceutical composition
KR20170029508A (en) * 2014-07-15 2017-03-15 아스텔라스세이야쿠 가부시키가이샤 Novel anti-human tie-2 antibody
US20170174789A1 (en) 2011-08-19 2017-06-22 Regeneron Pharmaceuticals, Inc. Anti-TIE2 Antibodies and Uses Thereof
JP2018043949A (en) 2016-09-15 2018-03-22 サンスター株式会社 COMPOSITION FOR ACTIVATING Tie2

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6365154B1 (en) 1998-09-28 2002-04-02 Smithkline Beecham Corporation Tie2 agonist antibodies
US7658924B2 (en) 2001-10-11 2010-02-09 Amgen Inc. Angiopoietin-2 specific binding agents
US8987420B2 (en) 2009-07-29 2015-03-24 Regeneron Pharmaceuticals, Inc. High affinity human antibodies to human angiopoietin-2
JP2011102273A (en) 2009-11-11 2011-05-26 Shiseido Co Ltd Tie2-activating agent, agent for maturing, normalizing or stabilizing blood vessel, lymphatic vessel-stabilizing agent, and wrinkle-preventing/ameliorating agent and dropsy-ameliorating/preventing agent
KR20140054303A (en) * 2011-08-19 2014-05-08 리제너론 파아마슈티컬스, 인크. Anti-tie2 antibodies and uses thereof
US20170174789A1 (en) 2011-08-19 2017-06-22 Regeneron Pharmaceuticals, Inc. Anti-TIE2 Antibodies and Uses Thereof
KR20150032075A (en) * 2013-09-17 2015-03-25 삼성전자주식회사 Anticancer composition containing an anti-Ang2 antibody inducing binding to Tie2 receptor
JP2015168656A (en) 2014-03-07 2015-09-28 丸善製薬株式会社 Tie2 activator, neovascularization inhibitor, agent for blood vessel maturation, agent for blood vessel normalization, and blood vessel stabilizer, and pharmaceutical composition
KR20170029508A (en) * 2014-07-15 2017-03-15 아스텔라스세이야쿠 가부시키가이샤 Novel anti-human tie-2 antibody
JP2018043949A (en) 2016-09-15 2018-03-22 サンスター株式会社 COMPOSITION FOR ACTIVATING Tie2

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
CHO ET AL., PNAS, 2004
DATABASE GenBank 20 March 2018 (2018-03-20), "angiopoietin-1 receptor isoform X3 [Pan troglodytes", Database accession no. XP_016816188 .1 *
DAVID S ET AL., AM J PHYSIOL LUNG CELL MOL PHYSIOL, 2011
FRYE M, J EXP. MED, 2015
GOEL S, J NATL CANCER INST, 2013
HAN ET AL., SCIENCE, 2016
HAYASHI M, NATURE COMMUNICATION, 2013
HOUDE DENGEN JR, METHODS MOL. BIOL., vol. 988, 2013, pages 269 - 89
HOUDE ET AL., J. PHARM. SCI., vol. 100, no. 6, 2011, pages 2071
MARTH C ET AL., EUR. J. CANCER, 2017
MELLBERG S ET AL., FASEB J, 2009
MOORE, J. O. ET AL.: "Dimerization of Tie2 mediated by its membrane-proximal FNIII domains", PNAS, vol. 114, no. 17, 25 April 2017 (2017-04-25), pages 4382 - 4387, XP055661385 *
SAHARINEN P ET AL., NATURE REVIEW DRUG DISCOVERY, 2017
ZHANG ET AL., JBC, vol. 277, 2002, pages 45276 - 45284

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113993897A (en) * 2019-08-14 2022-01-28 药物抗体公司 anti-TIE 2 antibodies and uses thereof
WO2021194913A1 (en) * 2020-03-24 2021-09-30 Genentech, Inc. Tie2-binding agents and methods of use

Similar Documents

Publication Publication Date Title
WO2020076105A1 (en) Novel anti-c-kit antibody
JP7390317B2 (en) Anti-Tie2 antibody and its use
WO2019164219A1 (en) Anti-angiopoietin-2 antibodies and uses thereof
WO2015167293A1 (en) Antibody binding to fcrn for treating autoimmune diseases
EP3755714A1 (en) Anti-angiopoietin-2 antibodies and uses thereof
WO2019004799A1 (en) Conjugate of vegf-grab protein and drug, and use thereof
WO2019235856A1 (en) Antibody binding to tie2 and use thereof
WO2021107566A1 (en) Antibody against c-kit and use thereof
AU2021227413A1 (en) Anti-CD56 antibody-drug conjugates and their use in therapy
CN108290949B (en) Binding molecules specific for ASCT2 and uses thereof
US11208494B2 (en) Monoclonal antibodies specific to the plexin-semaphorin-integrin (PSI) domain of RON for drug delivery and its application in cancer therapy
WO2017074013A1 (en) Antibody to be cross-linked to human and mouse sema3a, and use thereof
WO2022015113A1 (en) Antibody specifically binding to ptk7 and use thereof
WO2019088658A1 (en) Dual-targeting antibody targeting scf and galectin-1 and use thereof
WO2020242200A1 (en) Antibody with enhanced binding affinity for endothelin receptor a
WO2019216675A1 (en) Epitope of regulatory t cell surface antigen and antibody specifically binding thereto
WO2022039507A1 (en) Modified antibody and method for manufacturing same
WO2023167560A1 (en) Composition for combi-therapy comprising vegf-grab and pd-1 or pd-l1 antagonist
WO2022235090A1 (en) Tie2 agonistic antibodies and uses thereof
WO2020022776A1 (en) Fusion protein comprising thyrotropin receptor variants and use thereof
WO2021029746A2 (en) Anti-tie2 antibody and use thereof
WO2024054030A1 (en) Anti-ptk7 antibody, and use thereof
WO2021261906A1 (en) Anti-pcsk9 antibody and use thereof
WO2023136648A1 (en) Fusion protein comprising modified anti-vegfr2 (kdr) antibody and use thereof
WO2022245183A1 (en) Composition for preventing or treating non-alcoholic fatty liver disease or non-alcoholic steatohepatitis comprising growth differentiation factor-15 variant

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: 19814472

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3101506

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2020567832

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019283520

Country of ref document: AU

Date of ref document: 20190605

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019814472

Country of ref document: EP

Effective date: 20210111