WO2023224390A1 - Antibody for regulating ccr7 activity - Google Patents

Antibody for regulating ccr7 activity Download PDF

Info

Publication number
WO2023224390A1
WO2023224390A1 PCT/KR2023/006705 KR2023006705W WO2023224390A1 WO 2023224390 A1 WO2023224390 A1 WO 2023224390A1 KR 2023006705 W KR2023006705 W KR 2023006705W WO 2023224390 A1 WO2023224390 A1 WO 2023224390A1
Authority
WO
WIPO (PCT)
Prior art keywords
ccr7
seq
antibody
antigen
binding fragment
Prior art date
Application number
PCT/KR2023/006705
Other languages
French (fr)
Korean (ko)
Inventor
유연규
장문성
Original Assignee
국민대학교 산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 국민대학교 산학협력단 filed Critical 국민대학교 산학협력단
Publication of WO2023224390A1 publication Critical patent/WO2023224390A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/715Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons
    • G01N2333/7158Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons for chemokines

Definitions

  • the present invention relates to an antibody having the activity of inhibiting the signaling function of human CCR7 and a specific antibody of CCR7. More specifically, the antibody of the present invention can be used for the treatment of cancer diseases by inhibiting the signaling function of CCR7. .
  • CCR7 (CC chemokine receptor 7) is one of the chemokine receptors belonging to the rhodopsin-type G protein-coupled receptor (GPCR), which regulates the chemotactic process of lymphocytes in lymph nodes and promotes immunity by antigen-presenting cells (APC). Contributes to inducing tolerance.
  • GPCR G protein-coupled receptor
  • CCR7 is overexpressed in various metastatic cancers, including non-small cell lung cancer (NSCLC), B-cell chronic lymphocytic leukemia (BLL), lung cancer, pancreatic cancer, breast cancer, and hepatocellular carcinoma.
  • CCR7 plays a key role in metastasis through activation of intracellular signaling pathways, including PI3/Akt, MAPK/ERK, and JAK/STAT pathways in cancer cells, activation of NF- ⁇ B, upregulation of MMP9, and upregulation of MMP9 in tumor-related leukocytes. Since it is known to induce endothelial mesenchymal transition, it is attracting attention as a major drug target.
  • antibody drugs targeting CCR7 have not been commercialized. Even when expanded to all GPCRs, the only antibody drug approved for sale is Poteligeo (Mogamulizumab) from Kyowa Hakko Kirin, which targets CCR4. Therefore, the need to discover new antibodies that specifically bind to CCR7 and regulate its activity has emerged.
  • the present inventors made diligent efforts to discover a new antibody that specifically binds to CCR7 and inhibits the signal transduction process.
  • the present invention was completed by confirming the cancer cell-dependent migration or invasion blocking activity by interfering with signal transduction.
  • the purpose of the present invention is to provide an antibody or fragment thereof that recognizes CCR7 (CC chemokine receptor 7) as an antigen and specifically binds to it.
  • CCR7 CC chemokine receptor 7
  • Another object of the present invention is to provide a nucleic acid molecule encoding the antibody or antigen-binding fragment thereof.
  • Another object of the present invention is to provide a vector containing the above nucleic acid molecule.
  • Another object of the present invention is to provide a host cell containing the vector.
  • Another object of the present invention is to provide a composition containing the monoclonal antibody, nucleic acid molecule, or vector.
  • the present invention provides an antibody comprising heavy chain CDR1, CDR2, CDR3 and light chain CDR1, CDR2, CDR3, which recognizes CCR7 (CC chemokine receptor 7) as an antigen and specifically binds to it, or an antigen thereof.
  • a binding fragment is provided.
  • CCR7 which stabilized the natural folded structure using the P9 protein of Pseudomonas phi6, as an antigen, and performed biopanning using this as an antigen.
  • a primary screening method antibodies showing competitive binding to CCR7 with CCL19 or CCL21 ligand were selected, and it was confirmed that this effectively inhibits the signaling function of CCR7.
  • CCR7 (CC chemokine receptor 7) is one of the chemokine receptors belonging to the rhodopsin-type G protein-coupled receptor (GPCR), which regulates the chemotactic process of lymphocytes in lymph nodes and promotes immunity by antigen-presenting cells (APC). Contributes to inducing tolerance.
  • GPCR G protein-coupled receptor
  • CCR7 is overexpressed in various metastatic cancers, including non-small cell lung cancer (NSCLC), B-cell chronic lymphocytic leukemia (BLL), lung cancer, pancreatic cancer, breast cancer, and hepatocellular carcinoma.
  • CCR7 plays a key role in metastasis through activation of intracellular signaling pathways, including PI3/Akt, MAPK/ERK, and JAK/STAT pathways in cancer cells, activation of NF- ⁇ B, upregulation of MMP9, and upregulation of MMP9 in tumor-related leukocytes. Since it is known to induce endothelial mesenchymal transition, it is attracting attention as a major drug target.
  • the antibody or antigen-binding fragment thereof of the present invention binds specifically to the extracellular region of CCR7, rather than the intracellular region, as an antigen (e.g., epitope).
  • the present invention provides an antibody or antigen-binding fragment thereof comprising heavy chain CDR1, CDR2, and CDR3 and light chain CDR1, CDR2, and CDR3, which recognizes CCR7 (CC chemokine receptor 7) as an antigen and specifically binds to it.
  • CCR7 CC chemokine receptor 7
  • the CCR7 (CC chemokine receptor 7) is responsible for the intracellular signaling process by binding to the ligand CCL19 or CCL21, and is attracting attention as an anti-cancer target material. However, antibodies that still recognize the extracellular portion of CCR7 as an antigen No drugs have been reported.
  • the CCR7 (CC chemokine receptor 7) may preferably include the sequence of NCBI: NM_001838.4.
  • the antibody or antigen-binding fragment thereof can be used as a blocking antibody or antagonist antibody for CCR7, which means an antibody that inhibits or reduces the biological activity of the antigen to which it binds.
  • a preferred blocking antibody or antagonist antibody may be one that substantially or completely inhibits the biological activity of the antigen.
  • epitope refers to a localized site on an antigen to which an antibody or fragment thereof can specifically bind.
  • a continuous amino acid in an antigenic polypeptide can become an epitope, and two or more non-contiguous regions of the polypeptide can together become an epitope due to folding in the tertiary structure.
  • Epitopes are at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 consecutive or discontinuous amino acids in the unique three-dimensional structure of the antigen. It can include at least 9 or more, at least 10 or more, at least 11 or more, at least 12 or more, at least 13 or more, at least 14 or more, or at least 15 or more.
  • the antibody or antigen-binding fragment thereof of the present invention recognizes the extracellular portion of CCR7 (e.g., CC chemokine receptor 7) as an antigen and specifically binds to it, and specifically binds to one or more extracellular portions as an epitope.
  • the epitope may include one or more amino acids.
  • Methods for determining the epitope to which an antibody or antigen-binding fragment thereof according to the present invention binds include various methods such as immunoblotting and immunoprecipitation tests through reactivity tests for antibodies. There is. Determination of the three-dimensional spatial structure of an epitope can be performed using various methods such as x-ray crystallography, 2-dimensional nuclear magnetic resonance, and HDX-MS. (Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G. E. Morris, Ed. (1996)).
  • the epitope to which the antibody or antigen-binding fragment thereof of the present invention can bind is determined by NMR spectroscopy, X-ray diffraction crystallography, ELISA analysis, and HDX-MS (hydrogen/deuterium exchange coupled with mass spectrometry). , array-based oligo-peptide scanning assays, and/or mutagenesis mapping (Giege R et al.
  • the term “antibody” may be any type of immunoglobulin molecule (e.g., IgG, IgE, IgM, IgD, IgA, or IgY), or any subtype of antibody (e.g., human In mice, it may be IgG1, IgG2, IgG3, and IgG4; and in mice, it may be IgG1, IgG2a, IgG2b, and IgG3).
  • Immunoglobulins eg, IgG1 may exist in various allotypes, and the term “antibody” herein includes generally known isotypes and allotypes. Additionally, the term “antibody” herein may be IgG1, IgG2, IgG3, or IgG4, or a hybrid type thereof (e.g., a hybrid of IgG2 and IgG4).
  • the term “monoglon antibody” or “monoclonal antibody” refers to an antibody that exhibits single binding specificity and affinity for a specific epitope.
  • the monoclonal antibody is used to include its fragment, and the fragment preferably refers to an antigen binding fragment.
  • the fragment can be prepared using various methods known in the art. For example, through proteolytic cleavage of immunoglobulin molecules using enzymes such as papain (production of Fab fragments) or pepsin (F(ab')2), Fab and F(ab')2 fragments. can be manufactured.
  • fragment may be Fab, Fab', F(ab')2, Fv, scFV (single chain Fv), or scFv containing the VH or VL domain of a monomer, and the fragment may be Well known in the industry.
  • the antibody or antigen-binding fragment thereof of the present invention preferably has a heavy chain variable region selected from the group consisting of CDR1 of SEQ ID NO: 1 or 7, CDR2 of SEQ ID NO: 2 or 8, and CDR3 of SEQ ID NO: 3 or 9. It may include.
  • the antibody has the activity of inhibiting at least one of CCR7-dependent intracellular signaling and CCR7 receptor internalization by any one CCR7 ligand selected from CCL19 and CCL21.
  • the antibody or antigen-binding fragment thereof of the present invention is an antigen-binding protein that specifically binds to CCR7, and may bind to primate CCR7, preferably human CCR7.
  • the reference amino acid sequence of human CCR7 may be that indicated by NCBI: NM_001838.4,
  • the antibody or antigen-binding fragment thereof of the present invention is an antibody that can bind to CCR7 with excellent affinity, making it sufficiently effective as a diagnostic or therapeutic agent targeting CCR7.
  • the antibody according to the present invention may have low binding to non-CCR7 proteins and high binding to CCR7 proteins.
  • RIA radioimmunoassay
  • ELISA ELISA
  • antibodies binding to CCR7 have a high dissociation constant (K D ) of 20 to 100 nM, preferably a dissociation constant (K D ) of 20 to 50 nM.
  • the antibody or antigen-binding fragment thereof of the present invention may affect at least one of killing, inducing apoptosis, blocking migration, blocking activation, blocking proliferation, and blocking proliferation of CCR7-expressing cells at the receptor. there is.
  • the antibody or antigen-binding fragment thereof of the present invention preferably has a light chain variable region selected from the group consisting of CDR1 of SEQ ID NO: 4 or 10, CDR2 of SEQ ID NO: 5 or 11, and CDR3 of SEQ ID NO: 6 or 12. It may include
  • the VH domain, or one or more CDRs can be linked to the constant domain to form a heavy chain. Additionally, the VL domain, or one or more CDRs, can be linked to the constant domain to form a light chain.
  • the full-length heavy chain and full-length light chain combine to make up a full-length antibody.
  • the antibody or antigen-binding fragment thereof may include any one CDR combination selected from the following group:
  • the heavy chain variable region includes CDR1 of SEQ ID NO: 1, CDR2 of SEQ ID NO: 2, and CDR3 of SEQ ID NO: 3, and the light chain variable region includes CDR1 of SEQ ID NO: 4, CDR2 of SEQ ID NO: 5, and CDR3 of SEQ ID NO: 6. include;
  • the heavy chain variable region includes CDR1 of SEQ ID NO: 1, CDR2 of SEQ ID NO: 2, and CDR3 of SEQ ID NO: 3, and the light chain variable region includes CDR1 of SEQ ID NO: 10, CDR2 of SEQ ID NO: 11, and CDR3 of SEQ ID NO: 12. include;
  • the heavy chain variable region includes CDR1 of SEQ ID NO: 7, CDR2 of SEQ ID NO: 8, and CDR3 of SEQ ID NO: 9, and the light chain variable region includes CDR1 of SEQ ID NO: 4, CDR2 of SEQ ID NO: 5, and CDR3 of SEQ ID NO: 6. include; and
  • the heavy chain variable region includes CDR1 of SEQ ID NO: 7, CDR2 of SEQ ID NO: 8, and CDR3 of SEQ ID NO: 9, and the light chain variable region includes CDR1 of SEQ ID NO: 10, CDR2 of SEQ ID NO: 11, and CDR3 of SEQ ID NO: 12.
  • the antibody or antigen-binding fragment thereof includes a heavy chain variable region of SEQ ID NO: 13 or 15 and a light chain variable region of SEQ ID NO: 14 or 16, and more preferably the heavy chain of SEQ ID NO: 13.
  • the antibody or antigen-binding fragment thereof may include the amino acid sequence of SEQ ID NO: 17 or SEQ ID NO: 19.
  • the antibody or antigen-binding fragment thereof according to the present invention blocks binding of CCR7 to its ligand, thereby inhibiting biological activity, for example, the activity of inhibiting at least one of CCR7-dependent intracellular signaling and CCR7 receptor internalization by the CCR7 ligand. It may be.
  • the antibody or antigen-binding fragment thereof according to the present invention was developed using phage display using a human scFv library, and has a clear difference in that it is designing a fully human IgG 4 antibody. .
  • the antibody or antigen-binding fragment thereof according to the present invention has a significantly superior affinity of about 10 to 50 times more than that of CAP-100 (about 0.8 nM), and its antagonistic effect on cAMP accumulation is also quite effective.
  • the antibody or antigen-binding fragment thereof according to the present invention binds to the extracellular region of CCR7, blocking ligand binding, metastasis and invasion of cancer cells, and blocking ⁇ -arrestin-mediated ERK1/2 phosphorylation. Because it has a significant effect, it has the activity of effectively blocking the movement of cancer cells overexpressing CCR7, so it can be used as a treatment for a wider range of metastatic cancers than CAP-100.
  • the present invention provides a nucleic acid molecule encoding the polypeptide, a vector containing the nucleic acid molecule, or a host cell containing the vector.
  • Nucleic acid molecules of the present invention may be isolated or recombinant and include single- and double-stranded forms of DNA and RNA as well as corresponding complementary sequences.
  • isolated nucleic acid means, in the case of a nucleic acid isolated from a naturally occurring source, a nucleic acid that has been separated from the surrounding genetic sequence present in the genome of the individual from which the nucleic acid was isolated.
  • nucleic acids synthesized enzymatically or chemically from a template such as PCR products, cDNA molecules, or oligonucleotides
  • the nucleic acids resulting from these procedures may be understood as isolated nucleic acid molecules.
  • Isolated nucleic acid molecules refer to nucleic acid molecules either in the form of separate fragments or as components of larger nucleic acid constructs.
  • a nucleic acid is “operably linked” when placed into a functional relationship with another nucleic acid sequence.
  • the DNA of the presequence or secretion leader is operably linked to the DNA of the polypeptide when the polypeptide is expressed as a preprotein in a form before secretion
  • the promoter or enhancer is a polypeptide sequence. is operably linked to the coding sequence when it affects transcription
  • the ribosome binding site is operably linked to the coding sequence when configured to facilitate translation.
  • operably linked means that the DNA sequences to be linked are located adjacent to each other, and in the case of a secretory leader, it means that they are adjacent and exist within the same reading frame. However, enhancers do not need to be located adjacently. Linking is accomplished by ligation at convenient restriction enzyme sites. If such sites do not exist, synthetic oligonucleotide adapters or linkers are used according to conventional methods.
  • vector refers to a carrier capable of inserting a nucleic acid sequence for introduction into cells capable of replicating the nucleic acid sequence.
  • Nucleic acid sequences may be exogenous or heterologous.
  • Vectors include, but are not limited to, plasmids, cosmids, and viruses (eg, bacteriophages). Those skilled in the art can construct vectors by standard recombination techniques (Maniatis, et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y., 1988; and Ausubel et al., In: Current Protocols in Molecular Biology, John, Wiley & Sons, Inc, NY, 1994, etc.).
  • expression vector refers to a vector containing a nucleic acid sequence encoding at least a portion of the gene product to be transcribed. In some cases, the RNA molecule is then translated into a protein, polypeptide, or peptide. Expression vectors may contain various control sequences. In addition to regulatory sequences that regulate transcription and translation, vectors and expression vectors may also contain nucleic acid sequences that also serve other functions.
  • the term “host cell” includes eukaryotes and prokaryotes, and refers to any transformable organism capable of replicating the vector or expressing the gene encoded by the vector.
  • the host cell may be transfected or transformed by the vector, which refers to a process in which an exogenous nucleic acid molecule is transferred or introduced into the host cell.
  • Host cells of the present invention preferably include bacterial cells, CHO cells, HeLa cells, HEK293 cells, BHK-21 cells, COS7 cells, COP5 cells, A549 cells, NIH3T3 cells, etc., but are not limited thereto. no.
  • the host cell is an isolated host cell.
  • the present invention provides a composition comprising the antibody or antigen-binding fragment thereof, the nucleic acid molecule, or the vector.
  • the composition of the present invention is a pharmaceutical composition for preventing or treating cancer, and a pharmaceutical composition for preventing or treating metastasis of cancer.
  • the pharmaceutical composition of the present invention includes (a) the antibody or fragment thereof, the nucleic acid molecule, or a vector containing the nucleic acid molecule; and (b) a pharmaceutically acceptable carrier.
  • the present invention provides a method for preventing or treating cancer, and a method for preventing or treating metastasis of cancer, including the step of administering the pharmaceutical composition to a subject in need thereof.
  • the type of cancer that the present invention aims to prevent or treat is not limited, and includes leukemias, acute lymphocytic leukemia, acute nonlymphocytic leukemias, chronic lymphocytic leukemia, chronic Lymphomas, such as chronic myelogenous leukemia, Hodgkin's disease, non-Hodgkin's lymphomas, and multiple myeloma, brain tumors, and glioblastoma , childhood solid tumors such as neuroblastoma, rhabdomyosarcoma, retinoblastoma, Wilms Tumor, bone tumors, and soft-tissue sarcomas.
  • leukemias acute lymphocytic leukemia, acute nonlymphocytic leukemias, chronic lymphocytic leukemia, chronic Lymphomas, such as chronic myelogenous leukemia, Hodgkin's disease, non-Hodgkin's lymphomas, and multiple myelo
  • tumors lung cancer, breast cancer, prostate cancer, urinary cancers, uterine cancers, oral cancers, pancreatic cancer, melanoma ( melanoma and other skin cancers, stomach cancer, ovarian cancer, brain tumors, liver cancer, laryngeal cancer, thyroid cancer, and esophageal cancer. It can be administered to treat a number of cancers, including common solid tumors in adults, such as cancer and testicular cancer.
  • the pharmaceutical composition of the present invention can provide a preventive or therapeutic effect on cancer metastasis by inhibiting the migration and invasion of cancer cells.
  • cancer metastasis refers to cancer cells leaving the primary organ, moving to another organ, and proliferating.
  • the spread of cancer to other parts of the body is largely divided into two types: cancer tissue grows from the primary cancer and directly invades surrounding organs, and distant metastasis occurs along blood vessels or lymphatic vessels to other distant organs.
  • the metastatic cancer is formed by spreading through blood circulation or lymph circulation, and usually moves to other organs through blood circulation and then grows into a new tumor. Cancer cells attached to it can also be formed by moving directly to neighboring tissues.
  • cancer metastasis includes both the spread of cancer cells by invasion where cancer cells directly move and penetrate neighboring tissues, and metastasis in which cancer cells move through the bloodstream and form a new tumor in an organ that is not physically adjacent to the primary cancer. do. Meanwhile, in cancer metastasis, cell movement is essential. Therefore, inhibiting the movement of these cancer cells is the primary way to prevent cancer metastasis.
  • cancer metastasis cell movement is involved in several stages, such as when cancer cells move from the initial primary cancer site through the extracellular matrix into blood vessels, when they move out of blood vessels from a second metastatic tissue, and into new blood vessels.
  • the process is very complex and has various functions, including involvement in the movement of vascular endothelial cells. Therefore, considering that general anticancer drugs have the effect of simply killing cancer cells or inhibiting the proliferation of cancer cells, the use for inhibiting cancer metastasis can be distinguished separately from the simple anticancer use.
  • Pharmaceutically acceptable carriers included in the pharmaceutical composition of the present invention are those commonly used in preparation, and include lactose, dextrose, sucrose, sorbitol, mannitol, starch, gum acacia, calcium phosphate, alginate, gelatin, Includes, but is limited to, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate, and mineral oil. It doesn't work.
  • the pharmaceutical composition of the present invention may further include lubricants, wetting agents, sweeteners, flavoring agents, emulsifiers, suspending agents, preservatives, etc.
  • lubricants wetting agents, sweeteners, flavoring agents, emulsifiers, suspending agents, preservatives, etc.
  • suitable pharmaceutically acceptable carriers and formulations are described in detail in Remington's Pharmaceutical Sciences (19th ed., 1995).
  • composition of the present invention can be administered orally or parenterally, preferably parenterally, for example, by intravenous injection, local injection, and intraperitoneal injection.
  • the appropriate dosage of the pharmaceutical composition of the present invention varies depending on factors such as formulation method, administration method, patient's age, weight, sex, pathological condition, food, administration time, administration route, excretion rate, and reaction sensitivity. Usually, a skilled physician can easily determine and prescribe an effective dosage for the desired treatment or prevention. According to a preferred embodiment of the present invention, the daily dosage of the pharmaceutical composition of the present invention is 0.0001-100 mg/kg.
  • the pharmaceutical composition of the present invention is prepared in unit dosage form by formulating it using a pharmaceutically acceptable carrier and/or excipient according to a method that can be easily performed by a person skilled in the art. Alternatively, it can be manufactured by placing it in a multi-capacity container. At this time, the formulation may be in the form of a solution, suspension, or emulsion in an oil or aqueous medium, or may be in the form of an extract, powder, granule, tablet, or capsule, and may additionally contain a dispersant or stabilizer.
  • the term "subject" refers to an individual who needs to administer the composition of the present invention, and may be a human, mammal, bird, reptile, or fish, and preferably may be a mammal including a human.
  • composition of the present invention can be used as a stand-alone therapy, but can also be used in combination with other conventional chemotherapy or radiotherapy, and when such a combination therapy is performed, cancer treatment can be performed more effectively.
  • Chemotherapeutic agents that can be used with the composition of the present invention include cisplatin, carboplatin, procarbazine, mechlorethamine, cyclophosphamide, and ipo.
  • Radiation therapy that can be used with the composition of the present invention includes X-ray irradiation and ⁇ -ray irradiation.
  • the present invention provides a method for quantifying CCR7 contained in a sample, comprising treating the sample with an antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof of the present invention specifically binds to CCR7, the amount of CCR7 contained in a sample can be accurately measured using it.
  • the sample is not limited, but is preferably a sample separated outside the body from a subject, and more preferably a sample of blood, plasma, saliva, hair, urine, stool, or tissue separated outside the body from a subject.
  • a method of providing information for diagnosis of a disease caused by overexpression of CCR7 comprising the following steps:
  • the CCR7 controls cell migration to chemokine expression sites such as lymph nodes and the central nervous system by binding to the ligands CCL19 and/or CCL21; It is overexpressed in various cancers such as bladder cancer, lung cancer, ovarian cancer, kidney cancer, colon cancer, prostate cancer, breast cancer, uterine cancer, rhabdomyosarcoma, and glioblastoma, and is directly involved in major cancer progression processes such as proliferation, migration, invasion, and metastasis of cancer cells. Because it is involved, by comparing the expression level of CCR7 with that of normal people, information can be provided for diagnosis of diseases caused by overexpression of CCR7.
  • the disease caused by overexpression of CCR7 may be cancer, preferably metastatic cancer.
  • the present invention provides a CCR7 quantitative kit comprising the antibody or antigen-binding fragment thereof.
  • the quantitative kit of the present invention can quantify the amount of CCR7 by analyzing the antigen for the antibody through an antigen-antibody binding reaction, and the antigen-antibody binding reaction can be performed using conventional ELISA (Enzyme-linked immunosorbent assay), RIA (Radioimmnoassay), It is preferably selected from the group consisting of Sandwich assay, Western Blot on polyacrylamide gel, Immunoblot assay, and Immunohistochemical staining, but is not limited thereto.
  • ELISA Enzyme-linked immunosorbent assay
  • RIA Radioimmnoassay
  • the fixture for the antigen-antibody binding reaction consists of a nitrocellulose membrane, a PVDF membrane, a well plate synthesized from polyvinyl resin or polystyrene resin, and a slide glass made of glass. Any selected from the group may be used, but are not limited thereto.
  • the secondary antibody is preferably labeled with a common coloring agent that produces a color reaction, such as HRP (Horseradish peroxidase), alkaline phosphatase, colloid gold, or FITC (Poly L-lysine-fluorescein isothiocyanate). ), RITC (Rhodamine-B-isothiocyanate), etc.
  • a common coloring agent such as HRP (Horseradish peroxidase), alkaline phosphatase, colloid gold, or FITC (Poly L-lysine-fluorescein isothiocyanate). ), RITC (Rhodamine-B-isothiocyanate), etc.
  • a fluorescent substance Fluorescein
  • Dye dye
  • the substrate that induces color development is preferably used according to the label that produces the color reaction, such as TMB (3,3',5,5'-tetramethyl bezidine), ABTS [2,2'-azino-bis (3-ethylbenzothiazoline), -6-sulfonic acid)] and OPD (ophenylenediamine), but it is not limited thereto.
  • the present invention provides an antibody or antigen-binding fragment thereof that recognizes the extracellular region of CCR7 as an antigen and specifically binds to it.
  • the present invention discovers an antibody that specifically binds to the extracellular region while maintaining the structural characteristics of CCR7, showing the function of inhibiting the signaling process of CCR7 even in practical applications, and using this function to suppress CCR7 It can be useful as a treatment for cancer or metastasis of cancer.
  • Figure 1a shows purified functional P9-CCR7, and shows the results of analyzing samples obtained at each purification step by SDS-PAGE using Coomassie staining.
  • M is the size marker
  • Load is the solubilized membrane fraction treated with 1% sarkosyl
  • FT is the flow-through through Ni-NTA chromatography
  • W1 and W2 are 0 and 5mM, respectively.
  • This is the wash fraction obtained by treatment with a washing buffer containing dazole
  • E is the elution fraction obtained from Ni-NTA chromatography each treated with an elution buffer containing 20mM imidazole.
  • Figure 1b is a graph showing P9-CCR7 purified using a Ni-NTA affinity column, stabilized by treatment with APG, and then further purified using size exclusion chromatography.
  • 1 is the monodisperse peak of P9-CCR7 stabilized with APG
  • 2 is the peak that appears when free APG is separated.
  • Figure 1c shows the results of ELISA analysis of the activity of P9-CCR7, plates treated with 5% skimmed milk blocking solution (brown diamond);
  • the amount of P9-CCR7 binding to the ligand was measured using an anti-P9 antibody and an anti-mouse Fc antibody bound to HRP.
  • Figure 1d shows the results of ELISA analysis of the specific binding activity of the ligand, showing plates treated with 3% BSA blocking solution (red triangle); Alternatively, plates immobilized with 5 ⁇ g/ml APG:P9 (purple square) or 5 ⁇ g/ml P9-CCR7 (blue or orange circle) were prepared, and various concentrations (15-1000 nM) of CCL19-His were added thereto. -FLAG or CCL21-His-FLAG was applied. The amount of ligand binding to P9-CCR7 was measured using an anti-FLAG antibody and an anti-mouse Fc antibody coupled to HRP.
  • Figure 2a shows the results of screening of scFv clones specific for CCR7. This is the result of analyzing the specificity of scFv for CCR7 for the 16 initially selected clones by ELISA.
  • Figure 2b is a graph showing the results of treating scFv at each concentration with 100 nM CCL19-His-FLAG and analyzing it by ELISA. Blue circles and green squares represent signals by scFv 6RG11 and scFv 72C7, respectively.
  • Figure 2c is a graph showing the results of treating P9-CCR7 with 0 to 500 nM scFv and analyzing it by ELISA. Blue circles and green squares represent signals by scFv 6RG11 and scFv 72C7, respectively.
  • Figure 3a shows the results of SDS-PAGE analysis of purified IgG 4 (6RG11) and IgG 4 (72C7).
  • M is a size marker
  • NR is 6 ⁇ g of IgG 4 loaded under non-reducing conditions
  • R is 6 ⁇ g of IgG 4 loaded under reducing conditions using 1mM DTT.
  • Figure 3b shows the results of ELISA analysis of the specific binding of human IgG 4 (6RG11) and human IgG 4 (72C7) to CCR7.
  • Figure 3c is a graph showing the results of flow cytometry analysis of the specific binding of human IgG 4 (6RG11) to CCR7.
  • Figure 3d is a graph showing the results of flow cytometry analysis of the specific binding of human IgG 4 (72C7) to CCR7.
  • Figures 4 and 5 are graphs showing the results of analyzing the antagonistic activity of IgG 4 on the ligand effect through cAMP assay
  • Figure 4a is a graph measuring the effect of CCL19 ligand on HEK293 cells expressing CCR7
  • Figure 4b is a graph This is a graph measuring the effect of CCL21 ligand on HEK293 cells expressing CCR7
  • Figure 4c is a graph measuring the effect of CCL19 ligand on MDA-MB-231 cells expressing CCR7
  • Figure 4d is a graph measuring the effect of CCL19 ligand on MDA-MB-231 cells expressing CCR7. This is a graph measuring the effect of CCL21 ligand on MDA-MB-231 cells.
  • Figure 5a is a graph measuring the effect of IgG4 (6RG11) clone on CCL19 ligand in HEK293 cells expressing CCR7
  • Figure 5b is a graph measuring the effect of IgG4 (6RG11) clone on CCL21 ligand in HEK293 cells expressing CCR7
  • It is a graph
  • Figure 5c is a graph measuring the effect of IgG4 (6RG11) clone on CCL19 ligand in MDA-MB-231 cells expressing CCR7
  • Figure 5d is a graph measuring the effect of CCL21 in MDA-MB-231 cells expressing CCR7. This is a graph measuring the effect of IgG4 (6RG11) clone on the ligand.
  • Figure 6 is a photograph showing the effect of IgG 4 (6RG11, 72C7) on CCR7-dependent invasion induced by CCL19 ligand in MDA-MB-231 cells expressing CCR7.
  • Figure 7 is a photograph showing the effect of IgG 4 (6RG11, 72C7) on CCR7-dependent invasion induced by CCL21 ligand in MDA-MB-231 cells expressing CCR7.
  • Figure 8 is a graph showing the number of migrated cells measured from Figures 6 and 7, suggesting migration inhibition by IgG4 (6RG11, 72C7).
  • Figure 8a shows the number of migrated cells in MDA-MB-231 cells expressing CCR7 to CCL19 ligand.
  • Figure 8b is a graph measuring the effect of the IgG4 (6RG11) clone on CCL19 ligand in MDA-MB-231 cells expressing CCR7
  • Figure 8c is a graph measuring the effect of the IgG4 (72C7) clone on CCR7.
  • the DNA sequence representing the CCR7 coding region is designated as NCBI: NM_001838.4, and was designed as the coding optimization sequence of E. coli .
  • the coding sequence of CCL19 or CCL21 is indicated as NCBI: NM_006274.3 and NCBI: NM_002989.4, and is a sequence with FLAG and 6X His-tag, and was designed as a mammalian codon-optimized sequence.
  • the designed DNA sequences were synthesized by LNC Bio (Korea).
  • the mammalian expression vector of CCR7 was purchased from Sino Biological (China). Restriction enzymes, DNA polymerase, and T4 DNA ligase were purchased from New England Biolabs (NEB) (UK).
  • Amphipathic polyglutamate was obtained from MPRAB (Korea).
  • the synthetic scFv-displayed M13 library was provided by Dr. Ewha University. It was used as provided by Shim.
  • CCL19 and CCL21 were purchased from Peprotech (USA).
  • HRP-conjugated anti-mouse IgG was purchased from Sigma (USA), and Ni-NTA resin was purchased from Qiagen (Germany).
  • the HiLoad superdex 200 16/600 PG column was obtained from GE Healthcare (USA).
  • CNBr-activated sepharose 4B was purchased from GE Healthcare (USA).
  • IPTG Isopropyl- ⁇ -D-thiogalactoside
  • PMSF phenylmethylsulfonyl fluoride
  • sarkosyl purchased from Sigma (USA).
  • EDTA-free protease inhibitor cocktail was purchased from GenDEPOT (USA).
  • all consumable reagents were of reagent grade, and GraphPad Prism 8 software was used for data analysis.
  • the DNA sequence representing the coding region of CCR7 was amplified through PCR reaction using forward primer (5'TATTTTCAGTCGACGATGGAATTCATGGATCTGGGTAAACCAATGAAG ?? 3'; SEQ ID NO. 21) and reverse primer (5' - GTGATGGTGAGAAGCTTCGAATTCTGGAGAAAAGGTGGTAGTAGTTTC ?? 3'; SEQ ID NO. 22). did.
  • the PCR amplification product was purified using the QIAquick® PCR Purification Kit (Qiagen, Germany) and digested using restriction enzymes Sal I and Hind III . This was purified using the QIAquick® PCR Purification Kit and then ligated to the Sal I and Hind III sites of the pP9 vector.
  • a DNA fragment representing the coding region of CCL19 or CCL21 with FLAG and 6 23) and reverse primer (5'-CCGGCCTTGCCGGCCTCGAGTCATTACTTGTCGTCATCGT - 3'; SEQ ID NO. 24), forward primer of CCL21 (5'-TCCAGCCTC CGGACTCTAGAGCCGCCACCATGGCTCAGTC-3'; SEQ ID NO. 25) and reverse primer (5'-ATCCGG CCTTGCCGGCCTCGAGTCATTACTTGTCGTCATC-3'; SEQ ID NO: 26)
  • the PCR amplification product was purified using the QIAquick® PCR Purification Kit and ligated into the Xba I and Xho I sites of pCDNA 3.4 vector (NEB, UK).
  • Sequences encoding the variable regions VL and VH were amplified through PCR reaction.
  • the signal sequence for each light chain and heavy chain was the light chain signal sequence (5'-GCCGCCACCATGGCCGGCTTCCCTCTCCTCCTCACCCTCCTCACTCACTGTGCAGGATCCTGGGCCCAGTCTGTGCTGACTCAGCCACCC-3'; SEQ ID NO. 27) and heavy chain signal sequence (5'- GCCGCCACCATGGAGTTTGGGCTGAGCTGGCTTTTTCTTGTGGCTATTTTAAAAGGTGTCCAGTGTGAGGTGCAGCTGTTGGAGTCTGGG-3'; SEQ ID NO. 28) through an overlap extension PCR reaction. This was commissioned by Cosmogenetech (Korea) to produce a synthetic oligomer.
  • Each product was purified using a gel extraction kit (Qiagen, Germany), and in the pTRIOZ-hIgG4(S228P) vector (Invivogen, USA), VL was linked to the Sgr AI and Kpn I sites, and VH was linked to Nhe I and Age I. connected to the area. At this time, NEBuilder® HiFi DNA Assembly (NEB, UK) was used.
  • E. coli BL21(DE3)-RIPL cells containing the pP9-CCR7 vector were cultured using a bioreactor (Marado-PDA, CNS, Korea). When the OD 600 of the culture was 60, the expression of P9-CCR7 was induced by adding 1 mM IPTG for 21 hours at 18°C. The cells were recovered through centrifugation and stored at -80°C. 20 g of the cell paste was redispersed in 100 ml of buffer A (25 mM HEPES, pH 7.0) containing 1 mM PMSF and 1 tablet of EDTA-free protease inhibitor cocktail, and incubated with microfluidics (model and company) was used to dissolve it. Cell debris was removed by centrifugation at 12,000 xg for 30 minutes, and the supernatant was further centrifuged at 100,000 xg for 1 hour to precipitate as a membrane fraction.
  • buffer A 25 mM HEPES, pH 7.0
  • microfluidics model
  • the membrane fraction was placed in buffer B (25mM HEPES pH 7.0, 1% sarkosyl), dissolved by gentle stirring for 2 hours at 4°C, and then centrifuged at 15,000 x g for 30 minutes to remove precipitates. .
  • the membrane fraction solution was loaded onto a Ni-NTA column equilibrated with buffer A, and bound P9-CCR7 was eluted with 20mM imidazole. 1% APG was added to the elution fraction and mixed for 16 hours at 4°C. P9-CCR7 was stabilized with APG, further purified by size exclusion chromatography (HiLoad superdex 200 16/600 PG) equilibrated with buffer A, and stored at -80°C.
  • buffer B 25mM HEPES pH 7.0, 1% sarkosyl
  • Expi 293 cells ( 3 ⁇ 10 6 cells/mL) (ATCC, USA) were transfected in 50 ml. The transfected cells were cultured until cell viability reached at least 60% and harvested by centrifugation at 2000 xg at 4°C for 20 minutes.
  • the supernatant was filtered using a syringe filter (PES, Pore 0.22 ⁇ m) (Sartorius, USA), and filtered using DPBS (137 mM NaCl, 2.7 mM KCl, Na 2 HPO 4 10 mM, KH 2 PO 4 1.8 mM, pH 7.4) was loaded onto a Ni-NTA column equilibrated.
  • PES syringe filter
  • the target protein was eluted using 100 and 200 mM imidazole in equilibration buffer.
  • the purified protein was desalted with DPBS and 10% glycerol using a Pd-10 column (GE Healthcare, USA), and the eluted protein was stored at -80°C.
  • the P9 expression system in E. coli was successfully applied for the expression of human CCR7.
  • the P9 sequence is in a transversal form and helps CCR7 expression in the plasma membrane of E. coli .
  • 0.8 mg of P9-CCR7 was expressed (Western blot data as supplementary data).
  • P9-CCR7 was efficiently solubilized with sarkosyl, and 8 mg of P9-CCR7 was purified using a Ni-NTA affinity column ( Figure 1a). As a result, it was confirmed that it was successfully purified.
  • APG-stabilized P9-CCR7 stabilized by treatment with APG was measured by gel filtration chromatography, and a monodisperse peak (Molecular Weight: 347 kDa) and separated free APG (Molecular Weight: 30 kDa) were confirmed ( Figure 1b). It was confirmed that 5 mg of P9-CCR7 stabilized by binding to the final APG was purified from the membrane fraction of E. coli . This means that P9-CCR7 expressed in 20 g of wet cells was 31%.
  • hCCR7 could be purified from E. coli and successfully produce APG:P9-CCR7 using P9 and APG polymer.
  • the binding affinity between the purified P9-CCR7 and the ligand was measured using ELISA.
  • the purified CCL19-His-FLAG and CCL21-His-FLAG were immobilized on a Maxi-binding 96-well plate (SPL, USA). Each well was blocked with DPBS containing 5% skimmed milk, and then purified P9-CCR7 was added to the plate at various concentrations. The plate was then sequentially treated with anti-P9 antibody, HRP-conjugated anti-mouse IgG, ultra TMB (Thermo fisher scientific), and 1 N HCl. The amount of P9-CCR7 bound to the ligand was measured using a Synergy H1 (BioTek, USA) microplate reader (Figure 1c).
  • the interaction between the purified P9-CCR9 and CCL19 or CCL21 was measured using a modified ELISA method.
  • 500 ng of purified P9-CCR7 was immobilized in a Maxi-bound 96-well plate.
  • the plates were then washed, blocked, and treated with various concentrations of CCL19-His-FLAG or CCL21-His-FLAG.
  • the plate was sequentially treated with anti-FLAG M2 antibody (Sigma-Aldrich, USA), HRP-conjugated anti-mouse IgG, ultra TMB (Thermo Fisher Scientific), and 1 N HCl.
  • the amount of P9-CCR7 bound to the ligand was measured using a microplate reader ( Figure 1d).
  • Figure 1c shows the results of ELISA analysis of the activity of P9-CCR7, plates treated with 5% skimmed milk blocking solution (brown diamond); Alternatively, plates immobilized with 10 ⁇ g/ml of CCL19-His-FLAG (blue circle) or 10 ⁇ g/ml of CCL21-His-FLAG (orange square) were prepared, and various concentrations (1 to 500 nM) of P9- CCR7 was applied. The amount of APG:P9-CCR7 binding to the ligand was measured using an anti-P9 antibody and an anti-mouse Fc antibody conjugated to HRP.
  • Figure 1d shows the results of ELISA analysis of the specific binding activity of the ligand, showing plates treated with 3% BSA blocking solution (red triangle);
  • FLAG or CCL21-His-FLAG was applied.
  • the amount of ligand binding to P9-CCR7 was measured using an anti-FLAG antibody and an anti-mouse Fc antibody coupled to HRP.
  • the dissociation constants (K D s) of CCL19 or CCL21 with respect to purified P9-CCR7 were measured to be 19 nM and 11 nM, respectively. This confirmed that it was similar to the EC 50 value measured by BRET (bioluminescence resonance energy transfer) analysis using CCR7+ cells. Therefore, this indicates that the CCR7 region of the P9-CCR7 fusion protein has a native form and is highly suitable for ligand binding.
  • the purified protein was used as an antigen for the biopanning process.
  • the resin on which P9-CCR7 was immobilized was treated with termination buffer (100 mM Tris-Cl pH 8.0) at room temperature for 1 hour, and washed with DPBS (Dulbecco's phosphate buffered saline) using a centrifuge tube filter (Corning, USA). did.
  • the first step of biopanning begins with incubating 20 ⁇ l of P9-CCR7 immobilized beads with 0.20 ml of scFv-displaying M13 phage library ( ⁇ 1 ⁇ 10 12 CFU/ml) for 14 hours at 4°C.
  • the eluted phage was infected with mid-log phase ER2738, and the infected cells were spread on LB/Agar plates containing carbenicillin.
  • a single colony was inoculated in 750 ⁇ l of SB medium containing carbenicillin in a deep 96-well plate, and then cultured at 37°C for about 4 hours until it became turbid.
  • IPTG at a final concentration of 1 mM was added and cultured with shaking at 180 rpm and 30°C overnight.
  • the deep well plate was centrifuged at 3,500 x g for 20 minutes, the supernatant was removed, and cooled on ice.
  • the cell pellet was resuspended in 160 ⁇ l of 1X TES (50 mM Tris-Cl pH 8.0, 1 mM EDTA, 20% sucrose) buffer and 0.2 240 ⁇ l was added. After incubation on ice for 30 minutes, the plate was centrifuged to obtain a periplasmic fraction. 100 ⁇ l of the periplasmic extract was applied to each target GPCR-immobilized 96 well Maxi binding plate and incubated at room temperature for 1 hour. Next, antigen-binding clones were selected by sequentially treating HRP-conjugated anti-HA antibody (1:3000 dilution), TMB solution, and 1N HCl solution. Among the 384 clones obtained from step 1), 50 hit clones binding to the antigen were isolated, and among these, 16 unique clones were confirmed through sequence analysis (Table 1).
  • scFv was purified from 16 secondary selected clones. Plasmid DNA extracted from individual cultured cells was transformed into TOP10F competent cells. 10 ml of seed culture was incubated overnight at 37°C in SB medium containing 0.05 mg/ml carbenicillin. The culture was transferred to 500 mL of SB medium containing carbenicillin and incubated at 37°C until the OD 600 reached 0.6-0.8. After adding a final 1mM of IPTG to the culture, it was further incubated at 30°C overnight. Cells were harvested by centrifugation at 3,500 xg for 20 minutes and the supernatant was discarded.
  • the reacted resin was packed in a 5ml disposable column (Thermo Scientific, USA) and washed with 10ml of DPBST (0.05% Tween-20) containing 5mM imidazole. ScFv was eluted with 5 ml of DPBS containing 200mM imidazole, and the eluate was analyzed by SDS-PAGE.
  • 16 types of scFv (500 nM) purified through the above-described process were treated with APG:P9-CCR7 or APG:P9-GLP1R, respectively, and the bound scFv was detected using an HRP-conjugated anti-HA antibody. More specifically, the specificity between CCR7 and 16 scFvs was analyzed through ELISA. Purified P9-CCR7 (500 ng) or P9-GLP1R (500 ng) was immobilized on a 96-well Maxi binding plate and treated with 3% BSA. Next, each of the 16 types of scFv (500 nM) was treated and incubated at room temperature for 1 hour. To measure the amount of scFv bound to CCR7 or GPCR, HRP-conjugated anti-HA antibody (1:3000 diluted) was used.
  • Figure 2a shows the results of screening of scFv clones specific for CCR7, and is the result of analyzing the specificity of scFv for CCR7 for 16 types of clones by ELISA.
  • the bar graph in Figure 2A represents the signal of APG:P9-CCR7 divided by the signal of APG:P9-GLP1R.
  • purified P9-CCR7 was immobilized on a 96-well Maxi binding plate and then treated with 3% BSA. After washing three times with DPBS, scFv at various concentrations (4 ⁇ 3000 nM) was treated and incubated at room temperature for 30 minutes. DPBS mixed with CCL19-His-FLAG was treated to a final concentration of 100 nM, and further incubated at room temperature for 30 minutes. After washing with DPBS, the cells were treated with monoclonal ANTI-FLAG® M2 antibody (Thermo Fisher Scientific, USA) (1:1000 diluted in DPBS) and incubated for 1 hour at room temperature.
  • monoclonal ANTI-FLAG® M2 antibody Thermo Fisher Scientific, USA
  • Figure 2b is a graph showing the results of treating scFv (6RG11, 72C7) at different concentrations with 100 nM CCL19-His-FLAG and analyzing it by ELISA. As shown in Figure 2b, among the seven types of scFvs, only two types of scFvs (6RG11, 72C7) formed CCL19-competitive binding to CCR7. Accordingly, two types of clones (6RG11, 72C7) were finally selected according to the ELISA graph for scFv (6RG11, 72C7).
  • 6RG11 scfv of SEQ ID NO: 17 and 72C7 scFv containing SEQ ID NO: 19 were confirmed (CDR1 of SEQ ID NO: 1 in the heavy chain variable region, respectively, sequence CDR2 of SEQ ID NO: 2 and CDR3 of SEQ ID NO: 3, and the light chain variable region includes CDR1 of SEQ ID NO: 4, CDR2 of SEQ ID NO: 5, and CDR3 of SEQ ID NO: 6; CDR1 of SEQ ID NO: 7 and CDR2 of SEQ ID NO: 8 in the heavy chain variable region and CDR3 of SEQ ID NO: 9 and CDR1 of SEQ ID NO: 10, CDR2 of SEQ ID NO: 11, and CDR3 of SEQ ID NO: 12 in the light chain variable region.
  • Figure 2c is a graph showing the results of treating APG-O:P9-CCR7 with 0 to 500 nM scFv (6RG11, 72C7) and analyzing it by ELISA. Blue circles and green squares represent signals by scFv 6RG11 and scFv 72C7, respectively.
  • scFvs with CCR7-specific activity were successfully screened through the scFv-display M13 library using P9-CCR7 purified as an antigen. Approximately 56% of the identified clones were non-specific for CCR7. Non-specific clones are believed to bind to the P9 sequence and not to the CCR7 domain. Non-specific clones are expected to inhibit the inclusion of P9 protein during the biopanning process.
  • CCR7-specific scFvs excluding 2 types of scFvs (6RG11, 72C7) were confirmed to be unable to compete with CCL19.
  • the five scFvs that were not finally selected are believed to interact with intracellular or extracellular regions far from the CCL19 binding site of CCR7.
  • the above two types of scFv (6RG11, 72C7) form competitive bonds with CCL19 and CCR7 and have excellent inhibitory activity against CCR7 ligand binding, so they were finally selected as therapeutic antibodies targeting CCR7.
  • variable region sequences of the two final selected clones are pTRIOZ-hIgG4 (pTRIOZ-hIgG4), which contains a human lambda Fc region sequence expressing IgG 4 type immunoglobulin. S228P) vector (Invitrogen, USA).
  • the IgG 4 expression vector was transfected into 50 ml of Expi293 cells (3 ⁇ 10 6 cells/ml) (ATCC, USA) maintained in Expi293TM medium using the ExpiFectamineTM 293 transfection kit (Thermo Fisher Scientific, USA). Transfected cells were cultured until cell viability reached less than 60% and harvested at 2,000 xg for 20 min at 4°C.
  • the supernatant was filtered using a syringe filter (PES, Pore 0.22 ⁇ m) (Sartorius, USA) and loaded into CaptivA® (Repligen) equilibrated with DPBS. After washing the column with DPBS, the target protein was eluted with 100 mM Glycine-HCl pH 2.7 and immediately neutralized with 1M Tris-HCl pH 8.0. Purified proteins were diluted with DPBS to prevent precipitation, and proteins eluted with 10% glycerol were stored at -80°C until needed. The binding activity of purified IgG 4 to P9-CCR7 was confirmed using ELISA, and bound IgG 4 was analyzed using Goat anti-Human IgG (H+L) HRP (Thermo Fisher Scientific, USA).
  • an expression vector for human IgG 4 containing the heavy and light chain variable regions of 6RG11 and 72C7 was constructed, and the two IgG 4 proteins were purified and analyzed by SDS-PAGE.
  • human IgG 4 was purified from the culture medium of Expi 293 cells containing an IgG 4 expression vector using CaptivA protein A resin, and IgG 4 purified through the above process was analyzed by SDS-PAGE using Coomassie staining. .
  • Figure 3a shows the results of SDS-PAGE analysis of purified IgG 4 (6RG11) and IgG 4 (72C7).
  • M is a size marker
  • NR is 6 ⁇ g of IgG 4 loaded under non-reducing conditions
  • R is 6 ⁇ g of IgG 4 loaded under reducing conditions using 1mM DTT.
  • the binding affinity between CCR7 and IgG 4 (6RG11, 72C7) was analyzed through ELISA.
  • Purified P9-CCR7 (500 ng) or P9 (500 ng) was immobilized on a 96-well Maxi binding plate and treated with 3% BSA.
  • various concentrations (1 to 1000 nM) of IgG 4 (6RG11, 72C7) were treated and incubated at room temperature for 1 hour.
  • HRP-conjugated anti-HA antibody 1:3000 diluted.
  • HEK293 CCR7+ cells were prepared using Lipofectamine® 3000 (Thermo Fisher Scientific, USA) with pCDNA3.1-FLAG-CCR7 DNA plasmid (1 ug/ ⁇ L).
  • HEK293 CCR7- cells were maintained in culture medium (DMEM, 10% FBS, 1% penicillin/streptomycin). The cells were treated with 1 ml of TrypLETM Express Enzyme (Thermo Fisher Scientific, USA) to obtain a single cell suspension, which was centrifuged at 300 xg for 3 minutes, the supernatant was removed, and the cell pellet was obtained. Washed twice with DPBS.
  • the washed cell pellet was resuspended in 5 ml of DPBS containing 3% BSA. At this time, the final density of cells in the solution was set to 1 ⁇ 10 6 cells/ml. The resuspension was incubated at 4°C for 1 hour, 1 ml of cell solution (1 ⁇ 10 6 cells) was added to each tube, and rinsed using 1 ml DPBS. DPBS or 20 nM of 6RG11 or 72C7 or Isotype control (Opdivo) was added thereto, incubated with rotation at 4°C for 1 hour, and then centrifuged.
  • DPBS or 20 nM of 6RG11 or 72C7 or Isotype control (Opdivo) was added thereto, incubated with rotation at 4°C for 1 hour, and then centrifuged.
  • DPBS is shown in red
  • 20 nM of Opdivo IgG 4 is shown in blue
  • 20 nM of purified human IgG 4 (6RG11) or human IgG 4 (72C7) is shown in orange.
  • Figure 3b shows the results of ELISA analysis of the specific binding of human IgG 4 (6RG11) and human IgG 4 (72C7) to CCR7
  • Figure 3c shows the results of flow cytometry analysis of the specific binding of human IgG 4 (6RG11) to CCR7.
  • This is a graph showing the results of the analysis
  • Figure 3d is a graph showing the results of analyzing the specific binding of human IgG 4 (72C7) to CCR7 using flow cytometry.
  • Example 10 Blocking the activity of human IgG 4 (6RG11) and human IgG 4 (72C7) on the CCR7 signaling axis
  • HEK293 CCR7 + cells or MDA-MB-231 CCR7 + cells were maintained in DMEM medium containing 10% FBS and 1% penicillin/streptomycin.
  • DMEM fetal calf serum
  • IBMX 3-isobutyl-1-methylxanthine
  • CCL19 or CCL21 was used in the form of a mixture prepared by mixing various concentrations of CCL19 (Peprotech, USA) or CCL21 (Peprotech, USA) in 2 ⁇ forskolin buffer (cAMP stimulation buffer containing 40 ⁇ M forskolin). .
  • the effect of CCL19 ligand or CCL21 ligand was analyzed using cAMP-GloTM assay kit (Promega, USA) and detected with Synergy H1 (BioTek, USA) microplate reader.
  • HEK293 CCR7 + cells or MDA-MB-231 CCR7 + cells were distributed in a 96-well culture plate at a concentration of 1 ⁇ 10 4 cells/well and cultured overnight at 5% carbon dioxide and 37°C.
  • Replace the medium with cAMP stimulation buffer (DMEM, 0.5mM 3-isobutyl-1-methylxanthine (IBMX)) or cAMP stimulation buffer containing various concentrations of IgG 4 , and incubate for 20 minutes at 5% carbon dioxide at 37°C. cultured for a while.
  • DMEM 0.5mM 3-isobutyl-1-methylxanthine
  • IBMX 3-isobutyl-1-methylxanthine
  • the cells were treated with 2 ⁇ forskolin buffer (cAMP stimulation buffer containing 40 ⁇ M forskolin) or only 2 ⁇ forskolin buffer containing 20 nM of CCL19 (Peprotech, USA) or CCL21 (Peprotech, USA) and treated for 5 days. % carbon dioxide, and incubated for 20 minutes at 37°C.
  • the effect of IgG 4 on CCL19 or CCL21 ligand binding was also analyzed using the cAMP-GloTM assay kit (Promega, USA) and detected with a Synergy H1 (BioTek, USA) microplate reader.
  • Figures 4 and 5 are graphs showing the results of analyzing the antagonistic activity of IgG 4 on the ligand effect through cAMP assay
  • Figure 4a is a graph measuring the effect of CCL19 ligand on HEK293 cells expressing CCR7
  • Figure 4b is a graph This is a graph measuring the effect of CCL21 ligand on HEK293 cells expressing CCR7
  • Figure 4c is a graph measuring the effect of CCL19 ligand on MDA-MB-231 cells expressing CCR7
  • Figure 4d is a graph measuring the effect of CCL19 ligand on MDA-MB-231 cells expressing CCR7. This is a graph measuring the effect of CCL21 ligand on MDA-MB-231 cells.
  • Figure 5a is a graph measuring the effect of IgG4 (6RG11) clone on CCL19 ligand in HEK293 cells expressing CCR7
  • Figure 5b is a graph measuring the effect of IgG4 (6RG11) clone on CCL21 ligand in HEK293 cells expressing CCR7
  • It is a graph
  • Figure 5c is a graph measuring the effect of IgG4 (6RG11) clone on CCL19 ligand in MDA-MB-231 cells expressing CCR7
  • Figure 5d is a graph measuring the effect of CCL21 in MDA-MB-231 cells expressing CCR7. This is a graph measuring the effect of IgG4 (6RG11) clone on the ligand.
  • cAMP accumulation was confirmed when MDA-MB-231 or HEK293 cells expressing CCR7 were treated with various concentrations of CCL19 and CCL21 ligands.
  • the minimum concentration of CCL19 and CCL21 ligands was 0.1 nM in HEK293 cells, and 1 nM in MDA-MB-231 cells.
  • the concentration of CCL19 and CCL21 at which maximum cAMP accumulation occurred was 10 nM in HEK293 cells and 50-100 nM in MDA-MB-231 cells.
  • the concentration of CCL19 or CCL21 was fixed at 20 nM and the human 6RG11 antibody was treated with HEK293 CCR7+ cells at various concentrations (1-200 nM), resulting in a significant EC 50 of 3-12 nM. A significant decrease in cAMP accumulation was confirmed.
  • IgG 4 (6RG11) according to the present invention has excellent antagonistic activity against the CCR7 signaling pathway, especially Gi-dependent cAMP accumulation.
  • CCR7 (CC-chemokine receptor 7) is a chemotactic receptor and is known to induce cell migration.
  • human antibodies against CCR7 IgG 4 (6RG11) and IgG 4 (72C7) obtained through experiments were shown to inhibit the migration and invasion of MDA-MB-231 cancer cells expressing CCR7 induced by CCL19 or CCL21. We wanted to check whether it was effective.
  • MDA-MB-231 cells were transfected with pCDNA3.1-FLAG-CCR7 DNA plasmid (1 ug/ ⁇ L) using Lipofectamine® 3000 (Thermo Fisher Scientific, USA), and the medium was changed after 20 h, followed by 5 % carbon dioxide, and cultured overnight at 37°C. The medium was exchanged for starvation medium (DMEM, 0.5% FBS, 1% ( ⁇ ) penicillin/streptomycin), and further cultured for 6 hours.
  • DMEM starvation medium
  • FBS 1% ( ⁇ ) penicillin/streptomycin
  • Matrigel matrix (growth factor reduction, Corning, USA) was diluted to a concentration of 200 ⁇ g/ml using cold DMEM medium, and then placed on a permeable support (8.0 ⁇ m PET Membrane, Falcon) in a 24 well clear TC-treated well plate (Costar, USA). , USA) were each treated with 200 ⁇ L of the diluted Matrigel. Matrigel was cultured on the treated plate for more than 2 hours until it solidified. Remove DMEM medium from the upper chamber and treat with 200 ⁇ L of starved 2 ⁇ 10 5 cells alone or 200 ⁇ L of starved 2 ⁇ 10 5 cells mixed with human IgG 4 (6RG11, 72C7). was dispensed into the upper chamber.
  • the lower chamber was filled with 800 ⁇ L of starvation medium alone or 800 ⁇ L of starvation medium containing 30 nM of CCL19 or CCL21.
  • the 24-well plate was cultured for 40 hours, the upper chamber was wiped with a cotton swab, and the migrated cells were stained using the Diff Quick staining kit and detected with an inverted microscope. did. The number of migrated cells was counted using Image J software and analyzed using GraphPad Prism 8 software.
  • Figure 6 is a photograph showing the effect of IgG 4 (6RG11, 72C7) on CCR7-dependent invasion induced by CCL19 ligand in MDA-MB-231 cells expressing CCR7.
  • 7 is a photograph showing the effect of IgG 4 (6RG11, 72C7) on CCR7-dependent invasion induced by CCL21 ligand in MDA-MB-231 cells expressing CCR7.
  • Figure 8 is a graph showing the number of migrated cells measured from Figures 6 and 7, suggesting migration inhibition by IgG4 (6RG11, 72C7).
  • Figure 8a shows the number of migrated cells in MDA-MB-231 cells expressing CCR7 to CCL19 ligand.
  • Figure 8b is a graph measuring the effect of the IgG4 (6RG11) clone on CCL19 ligand in MDA-MB-231 cells expressing CCR7
  • Figure 8c is a graph measuring the effect of the IgG4 (72C7) clone on CCR7.
  • IgG 4 (6RG11) or IgG 4 (72C7) prevent the migration of CCR7 + cells induced by CCL19 or CCL21. It was confirmed that IgG 4 (6RG11) had better efficacy than IgG 4 (72C7) under low concentration conditions of less than 50 nM.
  • IgG 4 (6RG11) and IgG 4 (72C7) exhibited inhibitory activity against CCR7-dependent invasion.
  • CCR7 and CCL19/CCL21 ligands are known to play different roles in G protein activation, ⁇ -arrestin recruitment, internalization, migration, and signaling pathways, respectively.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Microbiology (AREA)
  • Public Health (AREA)
  • Cell Biology (AREA)
  • Genetics & Genomics (AREA)
  • Oncology (AREA)
  • Biophysics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hospice & Palliative Care (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention relates to an antibody having an inhibitory activity against the signal transduction function of human CCR7. More specifically, the antibody exhibits the function of inhibiting the signal transduction process of CCR7, and thus can be effectively used as a therapeutic agent for cancer or cancer metastasis involving CCR7.

Description

CCR7의 활성 조절 항체Antibodies that regulate the activity of CCR7
본 발명은 인간 CCR7의 신호전달기능 억제 활성을 갖는 항체 및 CCR7의 특이적 항체에 관한 것으로서, 보다 상세하게는 본 발명의 항체는 CCR7의 신호전달기능을 억제시킴으로써 암질환의 치료용도로 사용될 수 있다.The present invention relates to an antibody having the activity of inhibiting the signaling function of human CCR7 and a specific antibody of CCR7. More specifically, the antibody of the present invention can be used for the treatment of cancer diseases by inhibiting the signaling function of CCR7. .
CCR7(CC chemokine receptor 7)은 로돕신 형태(rhodopsin-type) G 단백질-결합 수용체(GPCR)에 속하는 케모카인 수용체 중 하나로, 림프절에서 림프구의 화학주성 과정을 조절하고, APC(antigen-presenting cell)의 면역 관용 유도에 기여한다. CCR7은 비소세포폐암(non-small cell lung cancer, NSCLC), B세포 만성림프구백혈병(B-cell chronic lymphocytic leukemia, BLL), 폐암, 췌장암, 유방암 및 간세포암 등 다양한 전이성 암에서 과발현된다.CCR7 (CC chemokine receptor 7) is one of the chemokine receptors belonging to the rhodopsin-type G protein-coupled receptor (GPCR), which regulates the chemotactic process of lymphocytes in lymph nodes and promotes immunity by antigen-presenting cells (APC). Contributes to inducing tolerance. CCR7 is overexpressed in various metastatic cancers, including non-small cell lung cancer (NSCLC), B-cell chronic lymphocytic leukemia (BLL), lung cancer, pancreatic cancer, breast cancer, and hepatocellular carcinoma.
또한 CCR7은 암세포에서 PI3/Akt, MAPK/ERK 및 JAK/STAT 경로를 포함한 세포내 신호전달 경로의 활성화를 통해 전이에 핵심 역할을 하고, 종양 관련 백혈구에서 NF-κB의 활성화, MMP9의 상향조절 및 중간엽 전이(endothelial mesenchymal transition)를 유발하는 것으로 알려진 바, 주요 약물 표적물질로 각광받고 있다.Additionally, CCR7 plays a key role in metastasis through activation of intracellular signaling pathways, including PI3/Akt, MAPK/ERK, and JAK/STAT pathways in cancer cells, activation of NF-κB, upregulation of MMP9, and upregulation of MMP9 in tumor-related leukocytes. Since it is known to induce endothelial mesenchymal transition, it is attracting attention as a major drug target.
상기 CCR7의 내생 리간드(endogenous ligands)인 CCL19(C-C Motif Chemokine Ligand 19) 및 CCL21(C-C Motif Chemokine Ligand 21)은 사이토카인과 케모카인의 발현을 조절하며, Gi-신호전달 기전을 통해 cAMP 농도 변화를 유도하고, 세포증식, 세포사멸, 이동 및 침습과 같은 다양한 세포내 현상(phenomenon)을 조절하는 것으로 알려져 있다.The endogenous ligands of CCR7, CCL19 (C-C Motif Chemokine Ligand 19) and CCL21 (C-C Motif Chemokine Ligand 21), regulate the expression of cytokines and chemokines and induce changes in cAMP concentration through Gi-signaling mechanism. It is known to regulate various intracellular phenomena such as cell proliferation, apoptosis, migration, and invasion.
항체 약물은 저분자 합성 의약품에 비해 높은 특이성, 낮은 부작용, 높은 혈중 반감기, Fc 영역에 의한 결함세포 사멸 작용기작(ADCC, ADCP, CDC) 등의 장점에도 불구하고 기존에 CCR7을 표적하는 항체약물은 상용화된 바 없으며, GPCR 전체로 확대하여도 판매 허가된 항체 약물은 CCR4를 표적하는 Kyowa Hakko Kirin사의 Poteligeo(Mogamulizumab)이 유일하다. 따라서 CCR7에 특이적으로 결합하고 이의 활성을 조절하는 신규 항체의 발굴의 필요성이 대두되었다. 또한 CCR7 표적의 신규 항체 약물을 개발하기 위해서는 우선 항원의 세포외 부위(extracellular domain, ECD)에 특이적으로 결합해야하는데, CCR7의 전체 표면적 중에서 세포외부위가 차지하는 비율은 약 10% 내외로 매우 제한적이다. 게다가 CCR7은 CCL19/CCL21 리간드와의 결합에 구조적 변화가 발생하기 때문에 기존에 사용해오던 항체 발굴전략으로는 한계가 있다. Despite the advantages of antibody drugs compared to small molecule synthetic drugs, such as high specificity, low side effects, high blood half-life, and defective cell death mechanism (ADCC, ADCP, CDC) by the Fc region, antibody drugs targeting CCR7 have not been commercialized. Even when expanded to all GPCRs, the only antibody drug approved for sale is Poteligeo (Mogamulizumab) from Kyowa Hakko Kirin, which targets CCR4. Therefore, the need to discover new antibodies that specifically bind to CCR7 and regulate its activity has emerged. In addition, in order to develop a new antibody drug targeting CCR7, it must first bind specifically to the extracellular domain (ECD) of the antigen, but the proportion of the extracellular domain of the total surface area of CCR7 is very limited, approximately 10%. am. In addition, because structural changes occur in CCR7 when binding to the CCL19/CCL21 ligand, there are limitations to the previously used antibody discovery strategy.
상기한 배경기술로서 설명된 사항들은 본 발명의 배경에 대한 이해 증진을 위한 것일 뿐, 이 기술분야에서 통상의 지식을 가진 자에게 이미 알려진 종래기술에 해당함을 인정하는 것으로 받아들여져서는 안 될 것이다.The matters described as the above background art are only for the purpose of improving the understanding of the background of the present invention, and should not be taken as an acknowledgment that they correspond to prior art already known to those skilled in the art.
본 발명자들은 CCR7에 특이적으로 결합하여 신호전달과정을 저해하는 신규항체를 발굴하고자 예의 노력을 하였다. 그 결과, CCR7과 CCL19/CCL21의 경쟁적 결합을 활용한 특이적 항체 스크리닝 전략을 고안하였고, 이를 이용하여 CCR7이 안정 발현된 세포주에서의 결합활성, CCR7 의존적 cAMP 축적 억제활성 및 CCL19/CCL21의 리간드 매개 신호전달을 방해하여 암세포의 의존적 이동 또는 침습 차단활성을 확인함으로써 본 발명을 완성하였다.The present inventors made diligent efforts to discover a new antibody that specifically binds to CCR7 and inhibits the signal transduction process. As a result, we designed a specific antibody screening strategy utilizing competitive binding between CCR7 and CCL19/CCL21, and used this to demonstrate binding activity in cell lines stably expressing CCR7, CCR7-dependent cAMP accumulation inhibitory activity, and ligand-mediated CCL19/CCL21 activity. The present invention was completed by confirming the cancer cell-dependent migration or invasion blocking activity by interfering with signal transduction.
따라서, 본 발명의 목적은 CCR7(CC chemokine receptor 7)을 항원으로 인식하여 특이적으로 결합하는 항체 또는 이의 단편을 제공하는데 있다.Therefore, the purpose of the present invention is to provide an antibody or fragment thereof that recognizes CCR7 (CC chemokine receptor 7) as an antigen and specifically binds to it.
본 발명의 또 다른 목적은 상기 항체 또는 이의 항원 결합 단편을 코딩하는 핵산분자를 제공하는데 있다.Another object of the present invention is to provide a nucleic acid molecule encoding the antibody or antigen-binding fragment thereof.
본 발명의 또 다른 목적은 상기 핵산분자를 포함하는 벡터를 제공하는데 있다.Another object of the present invention is to provide a vector containing the above nucleic acid molecule.
본 발명의 또 다른 목적은 상기 벡터를 포함하는 숙주세포를 제공하는데 있다.Another object of the present invention is to provide a host cell containing the vector.
본 발명의 또 다른 목적은 상기 단일클론항체, 핵산분자 또는 벡터를 포함하는 조성물을 제공하는데 있다.Another object of the present invention is to provide a composition containing the monoclonal antibody, nucleic acid molecule, or vector.
본 발명의 다른 목적 및 이점은 하기의 발명의 상세한 설명, 청구범위 및 도면에 의해 보다 명확하게 된다.Other objects and advantages of the present invention will become clearer from the following detailed description, claims, and drawings.
본 발명의 일 양태에 따르면, 본 발명은 CCR7(CC chemokine receptor 7)을 항원으로 인식하여 이에 특이적으로 결합하는, 중쇄 CDR1, CDR2, CDR3 및 경쇄 CDR1, CDR2, CDR3을 포함하는 항체 또는 이의 항원 결합 단편을 제공한다.According to one aspect of the present invention, the present invention provides an antibody comprising heavy chain CDR1, CDR2, CDR3 and light chain CDR1, CDR2, CDR3, which recognizes CCR7 (CC chemokine receptor 7) as an antigen and specifically binds to it, or an antigen thereof. A binding fragment is provided.
본 발명자들은 CCR7에 특이적으로 결합하여 신호전달과정을 저해하는 신규항체를 발굴하고자 노력한 결과, Pseudomonas phi6의 P9 단백질을 이용해 천연 접힌 구조를 안정화한 CCR7을 항원으로 준비하고, 이를 항원으로 한 바이오패닝 방법을 통하여 1차 스크리닝하고, CCR7에 대해 CCL19 또는 CCL21 리간드와 경쟁적 결합을 나타내는 항체를 선별하였으며, 이는 CCR7의 신호전달 기능을 효과적으로 억제함을 확인하였다. As a result of efforts to discover a new antibody that specifically binds to CCR7 and inhibits the signal transduction process, the present inventors prepared CCR7, which stabilized the natural folded structure using the P9 protein of Pseudomonas phi6, as an antigen, and performed biopanning using this as an antigen. Through a primary screening method, antibodies showing competitive binding to CCR7 with CCL19 or CCL21 ligand were selected, and it was confirmed that this effectively inhibits the signaling function of CCR7.
CCR7(CC chemokine receptor 7)은 로돕신 형태(rhodopsin-type) G 단백질-결합 수용체(GPCR)에 속하는 케모카인 수용체 중 하나로, 림프절에서 림프구의 화학주성 과정을 조절하고, APC(antigen-presenting cell)의 면역 관용 유도에 기여한다. CCR7은 비소세포폐암(non-small cell lung cancer, NSCLC), B세포 만성림프구백혈병(B-cell chronic lymphocytic leukemia, BLL), 폐암, 췌장암, 유방암 및 간세포암 등 다양한 전이성 암에서 과발현된다.CCR7 (CC chemokine receptor 7) is one of the chemokine receptors belonging to the rhodopsin-type G protein-coupled receptor (GPCR), which regulates the chemotactic process of lymphocytes in lymph nodes and promotes immunity by antigen-presenting cells (APC). Contributes to inducing tolerance. CCR7 is overexpressed in various metastatic cancers, including non-small cell lung cancer (NSCLC), B-cell chronic lymphocytic leukemia (BLL), lung cancer, pancreatic cancer, breast cancer, and hepatocellular carcinoma.
또한 CCR7은 암세포에서 PI3/Akt, MAPK/ERK 및 JAK/STAT 경로를 포함한 세포내 신호전달 경로의 활성화를 통해 전이에 핵심 역할을 하고, 종양 관련 백혈구에서 NF-κB의 활성화, MMP9의 상향조절 및 중간엽 전이(endothelial mesenchymal transition)를 유발하는 것으로 알려진 바, 주요 약물 표적물질로 각광받고 있다.Additionally, CCR7 plays a key role in metastasis through activation of intracellular signaling pathways, including PI3/Akt, MAPK/ERK, and JAK/STAT pathways in cancer cells, activation of NF-κB, upregulation of MMP9, and upregulation of MMP9 in tumor-related leukocytes. Since it is known to induce endothelial mesenchymal transition, it is attracting attention as a major drug target.
상기 CCR7의 내생 리간드(endogenous ligands)인 CCL19(C-C Motif Chemokine Ligand 19) 및 CCL21(C-C Motif Chemokine Ligand 21)은 사이토카인과 케모카인의 발현을 조절하며, Gi-신호전달 기전을 통해 cAMP 농도 변화를 유도하고, 세포증식, 세포사멸, 이동 및 침습과 같은 다양한 세포내 현상(phenomenon)을 조절하는 것으로 알려져 있다.The endogenous ligands of CCR7, CCL19 (C-C Motif Chemokine Ligand 19) and CCL21 (C-C Motif Chemokine Ligand 21), regulate the expression of cytokines and chemokines and induce changes in cAMP concentration through Gi-signaling mechanism. It is known to regulate various intracellular phenomena such as cell proliferation, apoptosis, migration, and invasion.
본 발명 바람직한 구현예에 따르면, 본 발명의 항체 또는 이의 항원 결합 단편은 CCR7의 세포내 부위가 아닌 세포외 부위를 항원(예를 들어, 에피토프(epitope))으로 하여 이에 특이적으로 결합한다.According to a preferred embodiment of the present invention, the antibody or antigen-binding fragment thereof of the present invention binds specifically to the extracellular region of CCR7, rather than the intracellular region, as an antigen (e.g., epitope).
본 발명은 CCR7(CC chemokine receptor 7)을 항원으로 인식하여 이에 특이적으로 결합하는, 중쇄 CDR1, CDR2, CDR3 및 경쇄 CDR1, CDR2, CDR3을 포함하는 항체 또는 이의 항원 결합 단편을 제공한다.The present invention provides an antibody or antigen-binding fragment thereof comprising heavy chain CDR1, CDR2, and CDR3 and light chain CDR1, CDR2, and CDR3, which recognizes CCR7 (CC chemokine receptor 7) as an antigen and specifically binds to it.
상기 CCR7(CC chemokine receptor 7)은 리간드인 CCL19 또는 CCL21과의 결합에 의한 세포내 신호전달과정을 담당하며, 항암 표적물질로 각광을 받고 있으나, 아직까지 CCR7의 세포외 부위를 항원으로 인식하는 항체 약물은 보고된 바 없다. 상기 CCR7(CC chemokine receptor 7)는 바람직하게는 NCBI: NM_001838.4의 서열을 포함하는 것일 수 있다.The CCR7 (CC chemokine receptor 7) is responsible for the intracellular signaling process by binding to the ligand CCL19 or CCL21, and is attracting attention as an anti-cancer target material. However, antibodies that still recognize the extracellular portion of CCR7 as an antigen No drugs have been reported. The CCR7 (CC chemokine receptor 7) may preferably include the sequence of NCBI: NM_001838.4.
본 발명에서 상기 항체 또는 이의 항원 결합 단편은 CCR7에 대한 차단 항체 또는 길항제 항체로 사용될 수 있으며, 이는 결합하는 항원의 생물학적 활성을 억제하거나 감소시키는 항체를 의미한다. 바람직한 차단항체 또는 길항제 항체는 항원의 생물학적 활성을 실질적으로 또는 완전히 억제하는 것일 수 있다. In the present invention, the antibody or antigen-binding fragment thereof can be used as a blocking antibody or antagonist antibody for CCR7, which means an antibody that inhibits or reduces the biological activity of the antigen to which it binds. A preferred blocking antibody or antagonist antibody may be one that substantially or completely inhibits the biological activity of the antigen.
본 명세서에서, 용어 "에피토프"라 함은 항체 또는 이의 단편이 특이적으로 결합할 수 있는 항원의 국소화된(localized) 부위를 의미한다. 예를 들어, 항원인 폴리펩타이드 중 연속적인 아미노산이 에피토프가 될 수 있으며, 폴리펩타이드에서 3차 구조상 접힘(folding)에 의해 비연속적인 2 또는 그 이상의 부위가 함께 에피토프가 될 수 있다. 에피토프는 항원의 독특한 3차원 구조에서, 연속 또는 비연속의 아미노산을 최소 2개 이상, 최소 3개 이상, 최소 4개 이상, 최소 5개 이상, 최소 6개 이상, 최소 7개 이상, 최소 8개 이상, 최소 9개 이상, 최소 10개 이상, 최소 11개 이상, 최소 12개 이상, 최소 13개 이상, 최소 14개 이상 또는 최소 15개 이상 포함할 수 있다. 본 발명의 항체 또는 이의 항원 결합 단편은 CCR7(예를 들어, CC chemokine receptor 7)의 세포외 부위를 항원으로 인식하여 이에 특이적으로 결합하며, 1 이상의 세포외 부위를 에피토프로 하여 특이적으로 결합할 수 있으며, 상기 에피토프는 1 또는 그 이상의 아미노산을 포함할 수 있다.As used herein, the term “epitope” refers to a localized site on an antigen to which an antibody or fragment thereof can specifically bind. For example, a continuous amino acid in an antigenic polypeptide can become an epitope, and two or more non-contiguous regions of the polypeptide can together become an epitope due to folding in the tertiary structure. Epitopes are at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 consecutive or discontinuous amino acids in the unique three-dimensional structure of the antigen. It can include at least 9 or more, at least 10 or more, at least 11 or more, at least 12 or more, at least 13 or more, at least 14 or more, or at least 15 or more. The antibody or antigen-binding fragment thereof of the present invention recognizes the extracellular portion of CCR7 (e.g., CC chemokine receptor 7) as an antigen and specifically binds to it, and specifically binds to one or more extracellular portions as an epitope. The epitope may include one or more amino acids.
본 발명에 따른 항체 또는 이의 항원 결합 단편이 결합하는 에피토프를 결정하는 방법(예를 들어, 에피토프 맵핑)은 항체에 대한 반응성 테스트를 통한 면역블로팅(immunoblotting) 및 면역침강(immunoprecipitation) 시험 등 다양한 방법이 있다. 에피토프의 3차원 공간 구조를 결정하는 방법은 x-레이 결정학(x-ray crystallography), 2차원 핵자기 공명법(2-dimensional nuclear magnetic resonance) 및 HDX-MS 등 다양한 방법을 이용하여 수행할 수 있다(Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G. E. Morris, Ed. (1996)).Methods for determining the epitope to which an antibody or antigen-binding fragment thereof according to the present invention binds (e.g., epitope mapping) include various methods such as immunoblotting and immunoprecipitation tests through reactivity tests for antibodies. There is. Determination of the three-dimensional spatial structure of an epitope can be performed using various methods such as x-ray crystallography, 2-dimensional nuclear magnetic resonance, and HDX-MS. (Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G. E. Morris, Ed. (1996)).
본 발명의 바람직한 구현예에 따르면, 본 발명의 항체 또는 이의 항원 결합 단편이 결합할 수 있는 에피토프는 NMR 분광학, X-ray 회절 결정학, ELISA 분석, HDX-MS(hydrogen/deuterium exchange coupled with mass spectrometry), 어레이 기반 올리고-펩타이드 스캐닝(array-based oligo-peptide scanning assays), 및/또는 돌연변이 유발 맵핑(mutagenesis mapping)을 통해 결정될 수 있다(Giege R et al., (1994) Acta Crystallogr D Biol Crystallogr 50(Pt 4): 339-350; McPherson A (1990) Eur J Biochem 189: 1-23; Chayen NE (1997) Structure 5: 1269-1274; McPherson A (1976) J Biol Chem 251 : 6300-6303).According to a preferred embodiment of the present invention, the epitope to which the antibody or antigen-binding fragment thereof of the present invention can bind is determined by NMR spectroscopy, X-ray diffraction crystallography, ELISA analysis, and HDX-MS (hydrogen/deuterium exchange coupled with mass spectrometry). , array-based oligo-peptide scanning assays, and/or mutagenesis mapping (Giege R et al. , (1994) Acta Crystallogr D Biol Crystallogr 50 ( Pt 4): 339-350; McPherson A (1990) Eur J Biochem 189: 1-23; Chayen NE (1997) Structure 5: 1269-1274; McPherson A (1976) J Biol Chem 251: 6300-6303).
본 명세서에서, 용어 "항체"는 면역글로불린 분자 중 어느 유형의 항체(예를 들어, IgG, IgE, IgM, IgD, IgA, 또는 IgY)일 수 있으며, 어느 하위 유형의 항체(예를 들어, 인간에 있어서 IgG1, IgG2, IgG3, 및 IgG4; 및 마우스에 있어서 IgG1, IgG2a, IgG2b, 및 IgG3)일 수 있다. 면역글로불린(예를 들어, IgG1)은 다양한 알로타입(allotype)이 존재할 수 있으며, 본 명세서에서 용어 "항체"는 일반적으로 알려진 아이소타입(isotype) 및 알로타입(allotype)을 포함한다. 또한, 본 명세서에서 용어 "항체"는 IgG1, IgG2, IgG3, 또는 IgG4 이거나, 이의 하이브리드(hybrid) 유형일 수 있다(예를 들어, IgG2 및 IgG4의 하이브리드).As used herein, the term “antibody” may be any type of immunoglobulin molecule (e.g., IgG, IgE, IgM, IgD, IgA, or IgY), or any subtype of antibody (e.g., human In mice, it may be IgG1, IgG2, IgG3, and IgG4; and in mice, it may be IgG1, IgG2a, IgG2b, and IgG3). Immunoglobulins (eg, IgG1) may exist in various allotypes, and the term “antibody” herein includes generally known isotypes and allotypes. Additionally, the term “antibody” herein may be IgG1, IgG2, IgG3, or IgG4, or a hybrid type thereof (e.g., a hybrid of IgG2 and IgG4).
본 명세서에서 용어 "단일글론항체"또는 "monoclonal antibody"는 특정 에피토프에 대한 단일 결합 특이성(single binding specificity) 및 친화도(affinity)를 나타내는 항체를 의미한다.As used herein, the term “monoglon antibody” or “monoclonal antibody” refers to an antibody that exhibits single binding specificity and affinity for a specific epitope.
본 명세서에서 상기 단일클론항체는 이의 단편을 포함하는 의미로 사용되며, 상기 단편은 바람직하게는 항원 결합 단편(antigen binding fragment)을 의미한다. 상기 단편은 당업계에 알려진 다양한 방법을 이용하여 제조할 수 있다. 예를 들어, 파파인(Fab 단편의 생산) 또는 펩신(F(ab')2)과 같은 효소를 이용하여 면역글로불린 분자의 단백질분해성 절단(proteolytic cleavage)을 통하여, Fab 및 F(ab')2 단편을 제조할 수 있다.In this specification, the monoclonal antibody is used to include its fragment, and the fragment preferably refers to an antigen binding fragment. The fragment can be prepared using various methods known in the art. For example, through proteolytic cleavage of immunoglobulin molecules using enzymes such as papain (production of Fab fragments) or pepsin (F(ab')2), Fab and F(ab')2 fragments. can be manufactured.
본 명세서에서, 용어 "단편"은 Fab, Fab', F(ab')2, Fv, scFV(single chain Fv), 또는 모노머의 VH 또는 VL 도메인을 포함하는 scFv일 수 있으며, 상기 단편에 대해서는 당업계에 잘 알려져 있다. As used herein, the term "fragment" may be Fab, Fab', F(ab')2, Fv, scFV (single chain Fv), or scFv containing the VH or VL domain of a monomer, and the fragment may be Well known in the industry.
본 발명의 상기 항체 또는 이의 항원 결합 단편은 바람직하게는 서열번호 1 또는 7의 CDR1, 서열번호 2 또는 8의 CDR2 및 서열번호 3 또는 9의 CDR3로 구성된 군으로부터 선택되는 중쇄(heavy chain) 가변영역을 포함하는 것일 수 있다. 상기 항체는 CCL19 및 CCL21로부터 선택되는 어느 하나의 CCR7 리간드에 의해 CCR7 의존성 세포 내 신호전달 및 CCR7 수용체 내재화 중의 적어도 하나를 억제하는 활성을 갖는다.The antibody or antigen-binding fragment thereof of the present invention preferably has a heavy chain variable region selected from the group consisting of CDR1 of SEQ ID NO: 1 or 7, CDR2 of SEQ ID NO: 2 or 8, and CDR3 of SEQ ID NO: 3 or 9. It may include. The antibody has the activity of inhibiting at least one of CCR7-dependent intracellular signaling and CCR7 receptor internalization by any one CCR7 ligand selected from CCL19 and CCL21.
본 발명의 항체 또는 이의 항원 결합 단편은 CCR7에 특이적으로 결합하는 항원 결합 단백질로, 영장류 CCR7, 바람직하게는 인간 CCR7에 결합하는 것일 수 있다. 인간 CCR7의 참조 아미노산 서열은 NCBI: NM_001838.4로 표시되는 것일 수 있다, The antibody or antigen-binding fragment thereof of the present invention is an antigen-binding protein that specifically binds to CCR7, and may bind to primate CCR7, preferably human CCR7. The reference amino acid sequence of human CCR7 may be that indicated by NCBI: NM_001838.4,
본 발명의 항체 또는 이의 항원 결합 단편은 CCR7을 표적화하는 진단제 나 치료제로서 충분히 유효하도록, 우수한 친화성으로 CCR7에 결합할 수 있는 항체이다. 바람직하게 본 발명에 따른 항체는 비-CCR7 단백질에 대한 결합은 낮고, CCR7 단백질에 대해서는 높은 결합을 갖는 것일 수 있다. 일예로 방사성 면역분석(RIA), ELISA에 의해 측정되는 경우, CCR7에 결합하는 항체는 높은 20 내지 100 nM의 해리상수(KD), 바람직하게는 20 내지 50 nM의 해리상수(KD)를 갖는다.The antibody or antigen-binding fragment thereof of the present invention is an antibody that can bind to CCR7 with excellent affinity, making it sufficiently effective as a diagnostic or therapeutic agent targeting CCR7. Preferably, the antibody according to the present invention may have low binding to non-CCR7 proteins and high binding to CCR7 proteins. For example, when measured by radioimmunoassay (RIA) or ELISA, antibodies binding to CCR7 have a high dissociation constant (K D ) of 20 to 100 nM, preferably a dissociation constant (K D ) of 20 to 50 nM. have
본 발명의 항체 또는 이의 항원 결합 단편은 수용체에서 CCR7 발현 세포의 사멸, 세포자멸사(apoptosis)의 유도, 이동의 차단, 활성화의 차단, 증식의 차단 및 확산의 차단 중 적어도 하나에 영향을 미치는 것일 수 있다.The antibody or antigen-binding fragment thereof of the present invention may affect at least one of killing, inducing apoptosis, blocking migration, blocking activation, blocking proliferation, and blocking proliferation of CCR7-expressing cells at the receptor. there is.
본 발명의 항체 또는 이의 항원 결합 단편은 바람직하게는 서열번호 4 또는 10의 CDR1, 서열번호 5 또는 11의 CDR2 및 서열번호 6 또는 12의 CDR3로 구성된 군으로부터 선택되는 경쇄(light chain) 가변영역을 포함하는 것일 수 있다.The antibody or antigen-binding fragment thereof of the present invention preferably has a light chain variable region selected from the group consisting of CDR1 of SEQ ID NO: 4 or 10, CDR2 of SEQ ID NO: 5 or 11, and CDR3 of SEQ ID NO: 6 or 12. It may include
VH 도메인, 또는 하나 또는 그 이상의 CDR은 중쇄(heavy chain)를 형성하기 위하여 불변 도메인에 연결될 수 있다. 또한, VL 도메인, 또는 하나 또는 그 이상의 CDR은 경쇄(light chain)를 형성하기 위하여 불변 도메인에 연결될 수 있다. 전장(full length) 중쇄 및 전장 경쇄가 결합하여 전장 항체를 구성한다.The VH domain, or one or more CDRs, can be linked to the constant domain to form a heavy chain. Additionally, the VL domain, or one or more CDRs, can be linked to the constant domain to form a light chain. The full-length heavy chain and full-length light chain combine to make up a full-length antibody.
본 발명의 바람직한 구현예에 따르면, 상기 항체 또는 이의 항원 결합 단편은 하기 군에서 선택되는 어느 하나의 CDR 조합을 포함할 수 있다:According to a preferred embodiment of the present invention, the antibody or antigen-binding fragment thereof may include any one CDR combination selected from the following group:
(A) 중쇄 가변영역에 서열번호 1의 CDR1, 서열번호 2의 CDR2 및 서열번호 3의 CDR3을 포함하고, 경쇄 가변영역에 서열번호 4의 CDR1, 서열번호 5의 CDR2 및 서열번호 6의 CDR3를 포함;(A) The heavy chain variable region includes CDR1 of SEQ ID NO: 1, CDR2 of SEQ ID NO: 2, and CDR3 of SEQ ID NO: 3, and the light chain variable region includes CDR1 of SEQ ID NO: 4, CDR2 of SEQ ID NO: 5, and CDR3 of SEQ ID NO: 6. include;
(B) 중쇄 가변영역에 서열번호 1의 CDR1, 서열번호 2의 CDR2 및 서열번호 3의 CDR3을 포함하고, 경쇄 가변영역에 서열번호 10의 CDR1, 서열번호 11의 CDR2 및 서열번호 12의 CDR3를 포함;(B) The heavy chain variable region includes CDR1 of SEQ ID NO: 1, CDR2 of SEQ ID NO: 2, and CDR3 of SEQ ID NO: 3, and the light chain variable region includes CDR1 of SEQ ID NO: 10, CDR2 of SEQ ID NO: 11, and CDR3 of SEQ ID NO: 12. include;
(C) 중쇄 가변영역에 서열번호 7의 CDR1, 서열번호 8의 CDR2 및 서열번호 9의 CDR3을 포함하고, 경쇄 가변영역에 서열번호 4의 CDR1, 서열번호 5의 CDR2 및 서열번호 6의 CDR3를 포함; 및(C) The heavy chain variable region includes CDR1 of SEQ ID NO: 7, CDR2 of SEQ ID NO: 8, and CDR3 of SEQ ID NO: 9, and the light chain variable region includes CDR1 of SEQ ID NO: 4, CDR2 of SEQ ID NO: 5, and CDR3 of SEQ ID NO: 6. include; and
(D) 중쇄 가변영역에 서열번호 7의 CDR1, 서열번호 8의 CDR2 및 서열번호 9의 CDR3을 포함하고, 경쇄 가변영역에 서열번호 10의 CDR1, 서열번호 11의 CDR2 및 서열번호 12의 CDR3를 포함한다.(D) The heavy chain variable region includes CDR1 of SEQ ID NO: 7, CDR2 of SEQ ID NO: 8, and CDR3 of SEQ ID NO: 9, and the light chain variable region includes CDR1 of SEQ ID NO: 10, CDR2 of SEQ ID NO: 11, and CDR3 of SEQ ID NO: 12. Includes.
본 발명의 바람직한 구현예에 따르면, 상기 항체 또는 이의 항원 결합 단편은 서열번호 13 또는 15의 중쇄 가변영역 및 서열번호 14 또는 16의 경쇄 가변영역을 포함하며, 보다 더 바람직하게는 서열번호 13의 중쇄 가변영역 및 서열번호 14의 경쇄 가변영역; 또는 서열번호 15의 중쇄 가변영역 및 서열번호 16의 경쇄 가변영역;을 포함하는 것일 수 있다.According to a preferred embodiment of the present invention, the antibody or antigen-binding fragment thereof includes a heavy chain variable region of SEQ ID NO: 13 or 15 and a light chain variable region of SEQ ID NO: 14 or 16, and more preferably the heavy chain of SEQ ID NO: 13. Variable region and light chain variable region of SEQ ID NO: 14; Or it may include a heavy chain variable region of SEQ ID NO: 15 and a light chain variable region of SEQ ID NO: 16.
본 발명의 가장 바람직한 구현예에 따르면 상기 항체 또는 이의 항원 결합 단편은 서열번호 17 또는 서열번호 19의 아미노산 서열을 포함할 수 있다.According to the most preferred embodiment of the present invention, the antibody or antigen-binding fragment thereof may include the amino acid sequence of SEQ ID NO: 17 or SEQ ID NO: 19.
본 발명에 따른 항체 또는 이의 항원 결합 단편은 CCR7이 그의 리간드에 결합하는 것을 차단하여 생물학적 활성, 예를 들어 CCR7 리간드에 의해 CCR7 의존성 세포 내 신호전달 및 CCR7 수용체 내재화 중의 적어도 하나를 억제하는 활성을 저해하는 것일 수 있다.The antibody or antigen-binding fragment thereof according to the present invention blocks binding of CCR7 to its ligand, thereby inhibiting biological activity, for example, the activity of inhibiting at least one of CCR7-dependent intracellular signaling and CCR7 receptor internalization by the CCR7 ligand. It may be.
본 발명에 따른 항체 또는 이의 항원 결합 단편은 CCR7을 항원으로 하는 CAP-100과 달리 인간 scFv 라이브러리를 사용한 파지 디스플레이를 사용하여 개발된 것으로, 완전한 인간 IgG4 항체를 설계하고 있다는 점에서 분명한 차이가 있다. 또한 본 발명에 따른 항체 또는 이의 항원 결합 단편은 CAP-100(약 0.8 nM)에 비해 약 10~50배 이상 현저히 우수한 친화성을 가지며, cAMP 축적에 대한 길항 효과 역시 상당히 효과적이다. Unlike CAP-100, which uses CCR7 as an antigen, the antibody or antigen-binding fragment thereof according to the present invention was developed using phage display using a human scFv library, and has a clear difference in that it is designing a fully human IgG 4 antibody. . In addition, the antibody or antigen-binding fragment thereof according to the present invention has a significantly superior affinity of about 10 to 50 times more than that of CAP-100 (about 0.8 nM), and its antagonistic effect on cAMP accumulation is also quite effective.
나아가, 본 발명에 따른 항체 또는 이의 항원 결합 단편은 CAP-100과는 달리 CCR7의 세포외 영역에 결합하여 리간드 결합, 암세포의 전이 및 침습을 차단하고, β-arrestin 매개 ERK1/2 인산화를 차단하는 현저한 효과를 나타내므로, CCR7을 과발현하는 암세포의 이동을 효과적으로 차단하는 활성이 있어 CAP-100보다 광범위한 전이성 암의 치료제로 활용될 수 있다.Furthermore, unlike CAP-100, the antibody or antigen-binding fragment thereof according to the present invention binds to the extracellular region of CCR7, blocking ligand binding, metastasis and invasion of cancer cells, and blocking β-arrestin-mediated ERK1/2 phosphorylation. Because it has a significant effect, it has the activity of effectively blocking the movement of cancer cells overexpressing CCR7, so it can be used as a treatment for a wider range of metastatic cancers than CAP-100.
본 발명의 또 다른 양태에 따르면, 본 발명은 상기 폴리펩타이드를 코딩하는 핵산분자, 상기 핵산분자를 포함하는 벡터 또는 상기 벡터를 포함하는 숙주세포를 제공한다.According to another aspect of the present invention, the present invention provides a nucleic acid molecule encoding the polypeptide, a vector containing the nucleic acid molecule, or a host cell containing the vector.
본 발명의 핵산분자는 단리된 것이거나 재조합된 것일 수 있으며, 단일쇄 및 이중쇄 형태의 DNA 및 RNA뿐만 아니라 대응하는 상보성 서열이 포함된다. “단리된 핵산”은 천연 생성 원천에서 단리된 핵산의 경우, 핵산이 단리된 개체의 게놈에 존재하는 주변 유전 서열로부터 분리된 핵산이다. 주형으로부터 효소적으로 또는 화학적으로 합성된 핵산, 예컨대 PCR 산물, cDNA 분자, 또는 올리고뉴클레오타이드의 경우, 이러한 절차로부터 생성된 핵산이 단리된 핵산분자로 이해될 수 있다. 단리된 핵산분자는 별도 단편의 형태 또는 더 큰 핵산 구축물의 성분으로서의 핵산 분자를 나타낸다. 핵산은 다른 핵산 서열과 기능적 관계로 배치될 때 “작동가능하게 연결”된다. 예를 들면, 전서열 또는 분비 리더(leader)의 DNA는 폴리펩타이드가 분비되기 전의 형태인 전단백질(preprotein)로서 발현되는 경우 폴리펩타이드의 DNA에 작동가능하게 연결되고, 프로모터 또는 인핸서는 폴리펩타이드 서열의 전사에 영향을 주는 경우 코딩 서열에 작동가능하게 연결되며, 또는 리보솜 결합 부위는 번역을 촉진하도록 배치될 때 코딩 서열에 작동가능하게 연결된다. 일반적으로 “작동가능하게 연결된”은 연결될 DNA 서열들이 인접하여 위치함을 의미하며, 분비 리더의 경우 인접하여 동일한 리딩 프레임 내에 존재하는 것을 의미한다. 그러나 인핸서는 인접하여 위치할 필요는 없다. 연결은 편리한 제한 효소 부위에서 라이게이션에 의해 달성된다. 이러한 부위가 존재하지 않는 경우, 합성 올리고뉴클레오타이드 어댑터 또는 링커를 통상적인 방법에 따라 사용한다. Nucleic acid molecules of the present invention may be isolated or recombinant and include single- and double-stranded forms of DNA and RNA as well as corresponding complementary sequences. “Isolated nucleic acid” means, in the case of a nucleic acid isolated from a naturally occurring source, a nucleic acid that has been separated from the surrounding genetic sequence present in the genome of the individual from which the nucleic acid was isolated. In the case of nucleic acids synthesized enzymatically or chemically from a template, such as PCR products, cDNA molecules, or oligonucleotides, the nucleic acids resulting from these procedures may be understood as isolated nucleic acid molecules. Isolated nucleic acid molecules refer to nucleic acid molecules either in the form of separate fragments or as components of larger nucleic acid constructs. A nucleic acid is “operably linked” when placed into a functional relationship with another nucleic acid sequence. For example, the DNA of the presequence or secretion leader is operably linked to the DNA of the polypeptide when the polypeptide is expressed as a preprotein in a form before secretion, and the promoter or enhancer is a polypeptide sequence. is operably linked to the coding sequence when it affects transcription, or the ribosome binding site is operably linked to the coding sequence when configured to facilitate translation. Generally, “operably linked” means that the DNA sequences to be linked are located adjacent to each other, and in the case of a secretory leader, it means that they are adjacent and exist within the same reading frame. However, enhancers do not need to be located adjacently. Linking is accomplished by ligation at convenient restriction enzyme sites. If such sites do not exist, synthetic oligonucleotide adapters or linkers are used according to conventional methods.
본 명세서에서 용어 “벡터”는 핵산 서열을 복제할 수 있는 세포로의 도입을 위해서 핵산 서열을 삽입할 수 있는 전달체를 의미한다. 핵산 서열은 외생(exogenous) 또는 이종(heterologous)일 수 있다. 벡터로서는 플라스미드, 코스미드 및 바이러스(예를 들면 박테리오파지)를 들 수 있으나, 이에 제한되지 않는다. 당업자는 표준적인 재조합 기술에 의해 벡터를 구축할 수 있다(Maniatis, et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y., 1988; 및 Ausubel et al., In: Current Protocols in Molecular Biology, John, Wiley & Sons, Inc, NY, 1994 등).As used herein, the term “vector” refers to a carrier capable of inserting a nucleic acid sequence for introduction into cells capable of replicating the nucleic acid sequence. Nucleic acid sequences may be exogenous or heterologous. Vectors include, but are not limited to, plasmids, cosmids, and viruses (eg, bacteriophages). Those skilled in the art can construct vectors by standard recombination techniques (Maniatis, et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y., 1988; and Ausubel et al., In: Current Protocols in Molecular Biology, John, Wiley & Sons, Inc, NY, 1994, etc.).
본 명세서에서 용어 “발현 벡터”는 전사되는 유전자 산물 중 적어도 일부분을 코딩하는 핵산 서열을 포함한 벡터를 의미한다. 일부의 경우에는 그 후 RNA 분자가 단백질, 폴리펩타이드, 또는 펩타이드로 번역된다. 발현 벡터에는 다양한 조절서열을 포함할 수 있다. 전사 및 번역을 조절하는 조절서열과 함께 벡터 및 발현 벡터에는 또 다른 기능도 제공하는 핵산 서열도 포함될 수 있다.As used herein, the term “expression vector” refers to a vector containing a nucleic acid sequence encoding at least a portion of the gene product to be transcribed. In some cases, the RNA molecule is then translated into a protein, polypeptide, or peptide. Expression vectors may contain various control sequences. In addition to regulatory sequences that regulate transcription and translation, vectors and expression vectors may also contain nucleic acid sequences that also serve other functions.
본 명세서에서 용어 “숙주세포”는 진핵생물 및 원핵생물을 포함하며, 상기 벡터를 복제할 수 있거나 벡터에 의해 코딩되는 유전자를 발현할 수 있는 임의의 형질 전환 가능한 생물을 의미한다. 숙주세포는 상기 벡터에 의해 형질감염(transfected) 또는 형질전환(transformed) 될 수 있으며, 이는 외생의 핵산분자가 숙주세포 내에 전달되거나 도입되는 과정을 의미한다. As used herein, the term “host cell” includes eukaryotes and prokaryotes, and refers to any transformable organism capable of replicating the vector or expressing the gene encoded by the vector. The host cell may be transfected or transformed by the vector, which refers to a process in which an exogenous nucleic acid molecule is transferred or introduced into the host cell.
본 발명의 숙주세포는 바람직하게는 세균(bacteria)세포, CHO 세포, HeLa 세포, HEK293 세포, BHK-21 세포, COS7 세포, COP5 세포, A549 세포, NIH3T3 세포 등을 들 수 있으나, 이에 제한되는 것은 아니다. Host cells of the present invention preferably include bacterial cells, CHO cells, HeLa cells, HEK293 cells, BHK-21 cells, COS7 cells, COP5 cells, A549 cells, NIH3T3 cells, etc., but are not limited thereto. no.
본 발명의 바람직한 구현예에 따르면, 상기 숙주세포는 단리된 숙주세포이다.According to a preferred embodiment of the present invention, the host cell is an isolated host cell.
본 발명의 또 다른 양태에 따르면, 본 발명은 상기 항체 또는 이의 항원 결합 단편, 상기 핵산분자 또는 상기 벡터를 포함하는 조성물을 제공한다.According to another aspect of the present invention, the present invention provides a composition comprising the antibody or antigen-binding fragment thereof, the nucleic acid molecule, or the vector.
본 발명의 바람직한 구현예에 따르면, 본 발명의 조성물은 암의 예방 또는 치료용 약제학적 조성물, 암의 전이 예방 또는 치료용 약제학적 조성물이다.According to a preferred embodiment of the present invention, the composition of the present invention is a pharmaceutical composition for preventing or treating cancer, and a pharmaceutical composition for preventing or treating metastasis of cancer.
본 발명의 약제학적 조성물은 (a) 상기 항체 또는 이의 단편, 상기 핵산분자 또는 상기 핵산분자를 포함하는 벡터; 및 (b) 약제학적으로 허용되는 담체를 포함할 수 있다.The pharmaceutical composition of the present invention includes (a) the antibody or fragment thereof, the nucleic acid molecule, or a vector containing the nucleic acid molecule; and (b) a pharmaceutically acceptable carrier.
본 발명의 또 다른 양태에 따르면, 본 발명은 상기 약제학적 조성물을 이를 필요로 하는 대상(subject)에 투여하는 단계를 포함하는 암의 예방 또는 치료방법, 암의 전이 예방 또는 치료방법을 제공한다.According to another aspect of the present invention, the present invention provides a method for preventing or treating cancer, and a method for preventing or treating metastasis of cancer, including the step of administering the pharmaceutical composition to a subject in need thereof.
본 발명이 예방 또는 치료하고자 하는 암의 종류는 제한되지 않으며, 백혈병(leukemias) 및 급성 림프구 백혈병(acute lymphocytic leukemia), 급성 비림프구 백혈병(acute nonlymphocytic leukemias), 만성 림프구 백혈병(chronic lymphocytic leukemia), 만성 골수 백혈병(chronic myelogenous leukemia), 호지킨 병(Hodgkin's Disease), 비호지킨 림프종(non-Hodgkin's lymphomas) 및 다발 골수종(multiple myeloma) 등과 같은 림프종(lymphomas), 뇌종양(brain tumors), 교모세포종(glioblastoma), 신경모세포종(neuroblastoma), 횡문근육종 (Rhabdomyosarcoma), 망막모세포종(retinoblastoma), 윌름즈종양(Wilms Tumor), 골종양(bone tumors) 및 연부조직육종(soft-tissue sarcomas) 등과 같은 소아 고형 종양(childhood solid tumors), 폐암(lung cancer), 유방암(breast cancer), 전립선암(prostate cancer), 요로암(urinary cancers), 자궁암(uterine cancers), 구강암(oral cancers), 췌장암(pancreatic cancer), 흑색종(melanoma) 및 기타 피부암(skin cancers), 위암(stomach cancer), 난소암(ovarian cancer), 뇌종양(brain tumors), 간암(liver cancer), 후두암(laryngeal cancer), 갑상선암(thyroid cancer), 식도암(esophageal cancer) 및 고환암(testicular cancer) 등과 같은 성인들의 통상의 고형 종양(common solid tumors)들을 포함하여 다수의 암들을 치료하도록 투여될 수 있다.The type of cancer that the present invention aims to prevent or treat is not limited, and includes leukemias, acute lymphocytic leukemia, acute nonlymphocytic leukemias, chronic lymphocytic leukemia, chronic Lymphomas, such as chronic myelogenous leukemia, Hodgkin's disease, non-Hodgkin's lymphomas, and multiple myeloma, brain tumors, and glioblastoma , childhood solid tumors such as neuroblastoma, rhabdomyosarcoma, retinoblastoma, Wilms Tumor, bone tumors, and soft-tissue sarcomas. tumors, lung cancer, breast cancer, prostate cancer, urinary cancers, uterine cancers, oral cancers, pancreatic cancer, melanoma ( melanoma and other skin cancers, stomach cancer, ovarian cancer, brain tumors, liver cancer, laryngeal cancer, thyroid cancer, and esophageal cancer. It can be administered to treat a number of cancers, including common solid tumors in adults, such as cancer and testicular cancer.
특히, 본 발명의 약제학적 조성물은 암세포의 이동 및 침습을 억제하는 것을 확인하여 암전이 예방 또는 치료효과를 제공할 수 있다.In particular, the pharmaceutical composition of the present invention can provide a preventive or therapeutic effect on cancer metastasis by inhibiting the migration and invasion of cancer cells.
본 발명에서 용어 "암전이"는 암세포가 원발 장기를 떠나 다른 장기로 이동하여 증식하는 것을 말한다. 암이 신체의 다른 부분으로 퍼지는 것은 크게 원발암에서 암조직이 성장하여 직접적으로 주위 장기를 침습하는 것과 멀리 있는 다른 장기로 혈관이라 림프관을 따라 원격전이를 하는 것으로 구분된다. In the present invention, the term “cancer metastasis” refers to cancer cells leaving the primary organ, moving to another organ, and proliferating. The spread of cancer to other parts of the body is largely divided into two types: cancer tissue grows from the primary cancer and directly invades surrounding organs, and distant metastasis occurs along blood vessels or lymphatic vessels to other distant organs.
상기 전이암은 혈액순환이나 림프순환을 통해 퍼져나가서 형성되는 것으로, 대개는 혈액순환을 타고 다른 장기로 옮겨간 후 새로운 종양으로 자라난 것이다. 이와 달린 암세포가 이웃한 조직으로 직접 이동하여 형성되기도 한다. 본 발명에서 암 전이는 암세포가 이웃조직으로 직접 이동하고 침투하는 침윤에 의한 암세포의 확산 및 암세포가 혈류를 타고 이동하여 물리적으로 원발암과는 인접하지 않는 장기에서 새로운 종양을 형성하는 전이를 모두 포함한다. 한편 암 전이에 있어서, 세포의 이동은 필수적이다. 따라서 이러한 암세포의 이동을 저해하는 것이 암전이를 예방하는 일차적인 방법이다.The metastatic cancer is formed by spreading through blood circulation or lymph circulation, and usually moves to other organs through blood circulation and then grows into a new tumor. Cancer cells attached to it can also be formed by moving directly to neighboring tissues. In the present invention, cancer metastasis includes both the spread of cancer cells by invasion where cancer cells directly move and penetrate neighboring tissues, and metastasis in which cancer cells move through the bloodstream and form a new tumor in an organ that is not physically adjacent to the primary cancer. do. Meanwhile, in cancer metastasis, cell movement is essential. Therefore, inhibiting the movement of these cancer cells is the primary way to prevent cancer metastasis.
또한 암전이에 있어서, 세포의 이동은 여러 단계에 관여하는데, 암세포가 초기의 원발암 위치에서 세포간질(Extracellular matrix)을 지나 혈관으로 이동할 때, 제2의 전이 조직에서 혈관 밖으로 이동할 때 신생혈관에서 혈관 내피세포(Vascular endothelial cell)가 이동할 때에 관여하는 등 그 과정이 매우 복잡하고 다양한 기능을 갖고 있다. 따라서 일반적인 항암제가 단순히 암세포를 사멸시키거나 암세포의 증식을 억제세키는 효과를 갖는 것임을 고려할 때, 암전이 억제 용도는 단순 항암 용도와는 별도로 구분될 수 있다.In addition, in cancer metastasis, cell movement is involved in several stages, such as when cancer cells move from the initial primary cancer site through the extracellular matrix into blood vessels, when they move out of blood vessels from a second metastatic tissue, and into new blood vessels. The process is very complex and has various functions, including involvement in the movement of vascular endothelial cells. Therefore, considering that general anticancer drugs have the effect of simply killing cancer cells or inhibiting the proliferation of cancer cells, the use for inhibiting cancer metastasis can be distinguished separately from the simple anticancer use.
본 발명의 약제학적 조성물에 포함되는 약제학적으로 허용되는 담체는 제제시에 통상적으로 이용되는 것으로서, 락토스, 덱스트로스, 수크로스, 솔비톨, 만니톨, 전분, 아카시아 고무, 인산 칼슘, 알기네이트, 젤라틴, 규산 칼슘, 미세결정성 셀룰로스, 폴리비닐피롤리돈, 셀룰로스, 물, 시럽, 메틸 셀룰로스, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 활석, 스테아르산 마그네슘 및 미네랄 오일 등을 포함하나, 이에 한정되는 것은 아니다. 본 발명의 약제학적 조성물은 상기 성분들 이외에 윤활제, 습윤제, 감미제, 향미제, 유화제, 현탁제, 보존제 등을 추가로 포함할 수 있다. 적합한 약제학적으로 허용되는 담체 및 제제는 Remington's Pharmaceutical Sciences (19th ed., 1995)에 상세히 기재되어 있다. Pharmaceutically acceptable carriers included in the pharmaceutical composition of the present invention are those commonly used in preparation, and include lactose, dextrose, sucrose, sorbitol, mannitol, starch, gum acacia, calcium phosphate, alginate, gelatin, Includes, but is limited to, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate, and mineral oil. It doesn't work. In addition to the above ingredients, the pharmaceutical composition of the present invention may further include lubricants, wetting agents, sweeteners, flavoring agents, emulsifiers, suspending agents, preservatives, etc. Suitable pharmaceutically acceptable carriers and formulations are described in detail in Remington's Pharmaceutical Sciences (19th ed., 1995).
본 발명의 약제학적 조성물은 경구 또는 비경구로 투여할 수 있고, 바람직하게는 비경구 투여이며, 예컨대, 정맥 내 주입, 국소 주입 및 복강 주입 등으로 투여할 수 있다.The pharmaceutical composition of the present invention can be administered orally or parenterally, preferably parenterally, for example, by intravenous injection, local injection, and intraperitoneal injection.
본 발명의 약제학적 조성물의 적합한 투여량은 제제화 방법, 투여 방식, 환자의 연령, 체중, 성, 병적 상태, 음식, 투여 시간, 투여 경로, 배설 속도 및 반응 감응성과 같은 요인들에 의해 다양하며, 보통으로 숙련된 의사는 소망하는 치료 또는 예방에 효과적인 투여량을 용이하게 결정 및 처방할 수 있다. 본 발명의 바람직한 구현예에 따르면, 본 발명의 약제학적 조성물의 1일 투여량은 0.0001-100 ㎎/㎏이다.The appropriate dosage of the pharmaceutical composition of the present invention varies depending on factors such as formulation method, administration method, patient's age, weight, sex, pathological condition, food, administration time, administration route, excretion rate, and reaction sensitivity. Usually, a skilled physician can easily determine and prescribe an effective dosage for the desired treatment or prevention. According to a preferred embodiment of the present invention, the daily dosage of the pharmaceutical composition of the present invention is 0.0001-100 mg/kg.
본 발명의 약제학적 조성물은 당해 발명이 속하는 기술분야에서 통상의 지식을 가진 자가 용이하게 실시할 수 있는 방법에 따라, 약제학적으로 허용되는 담체 및/또는 부형제를 이용하여 제제화 함으로써 단위 용량 형태로 제조되거나 또는 다용량 용기 내에 내입시켜 제조될 수 있다. 이때 제형은 오일 또는 수성 매질중의 용액, 현탁액 또는 유화액 형태이거나 엑스제, 분말제, 과립제, 정제 또는 캅셀제 형태일 수도 있으며, 분산제 또는 안정화제를 추가적으로 포함할 수 있다.The pharmaceutical composition of the present invention is prepared in unit dosage form by formulating it using a pharmaceutically acceptable carrier and/or excipient according to a method that can be easily performed by a person skilled in the art. Alternatively, it can be manufactured by placing it in a multi-capacity container. At this time, the formulation may be in the form of a solution, suspension, or emulsion in an oil or aqueous medium, or may be in the form of an extract, powder, granule, tablet, or capsule, and may additionally contain a dispersant or stabilizer.
본 발명에서 용어 "대상"은 본 발명의 조성물을 투여할 필요가 있는 개체를 의미하며, 인간, 포유동물, 조류, 파충류, 어류일 수 있고, 바람직하게는 인간을 포함하는 포유동물일 수 있다.In the present invention, the term "subject" refers to an individual who needs to administer the composition of the present invention, and may be a human, mammal, bird, reptile, or fish, and preferably may be a mammal including a human.
본 발명의 약제학적 조성물은 단독의 요법으로 이용될 수 있으나, 다른 통상적인 화학 요법 또는 방사 요법과 함께 이용될 수도 있으며, 이러한 병행 요법을 실시하는 경우에는 보다 효과적으로 암 치료를 할 수 있다. 본 발명의 조성물과 함께 이용될 수 있는 화학 요법제는 시스플라틴(cisplatin), 카르보플라틴(carboplatin), 프로카르바진(procarbazine), 메클로레타민(mechlorethamine), 시클로포스파미드(cyclophosphamide), 이포스파미드(ifosfamide), 멜팔란(melphalan), 클로라부실(chlorambucil), 비술판(bisulfan), 니트로소우레아(nitrosourea), 디악티노마이신(dactinomycin), 다우노루비신(daunorubicin), 독소루비신(doxorubicin), 블레오마이신(bleomycin), 플리코마이신(plicomycin), 미토마이신(mitomycin), 에토포시드(etoposide), 탁목시펜(tamoxifen), 택솔(taxol), 트랜스플라티눔(transplatinum), 5-플루오로우라실(5-fluorouracil), 빈크리스틴(vincristin), 빈블라스틴(vinblastin) 및 메토트렉세이트(methotrexate) 등을 포함한다. 본 발명의 조성물과 함께 이용될 수 있는 방사 요법은 X-선 조사 및 γ-선 조사 등이다.The pharmaceutical composition of the present invention can be used as a stand-alone therapy, but can also be used in combination with other conventional chemotherapy or radiotherapy, and when such a combination therapy is performed, cancer treatment can be performed more effectively. Chemotherapeutic agents that can be used with the composition of the present invention include cisplatin, carboplatin, procarbazine, mechlorethamine, cyclophosphamide, and ipo. ifosfamide, melphalan, chlorambucil, bisulfan, nitrosourea, dactinomycin, daunorubicin, doxorubicin , bleomycin, plicomycin, mitomycin, etoposide, tamoxifen, taxol, transplatinum, 5-fluoro Includes 5-fluorouracil, vincristin, vinblastin, and methotrexate. Radiation therapy that can be used with the composition of the present invention includes X-ray irradiation and γ-ray irradiation.
본 발명의 또 다른 양태에 따르면, 본 발명은 항체 또는 이의 항원 결합 단편을 샘플에 처리하는 단계를 포함하는, 샘플 중에 포함된 CCR7의 정량 방법을 제공한다.According to another aspect of the present invention, the present invention provides a method for quantifying CCR7 contained in a sample, comprising treating the sample with an antibody or antigen-binding fragment thereof.
본 발명의 항체 또는 이의 항원 결합 단편은 CCR7에 특이적으로 결합하기 때문에 이를 이용하면 샘플 중에 포함된 CCR7의 양을 정확하게 측정 가능하다.Since the antibody or antigen-binding fragment thereof of the present invention specifically binds to CCR7, the amount of CCR7 contained in a sample can be accurately measured using it.
상기 샘플은 제한되지 않으나, 바람직하게는 피검자로부터 체외로 분리된 샘플이며, 보다 바람직하게는 피검자로부터 체외로 분리된 혈액, 혈장, 타액, 모발, 소변, 대변, 또는 조직 샘플이다.The sample is not limited, but is preferably a sample separated outside the body from a subject, and more preferably a sample of blood, plasma, saliva, hair, urine, stool, or tissue separated outside the body from a subject.
본 발명의 또 다른 양태에 따르면, 하기의 단계를 포함하는 CCR7의 과발현에 의한 질환의 진단을 위한 정보를 제공하는 방법을 제공한다:According to another aspect of the present invention, there is provided a method of providing information for diagnosis of a disease caused by overexpression of CCR7, comprising the following steps:
(a) 피검자로부터 체외로 분리된 샘플을 취득하는 단계;(a) obtaining a sample separated from the subject in vitro;
(b) 상기 항체 또는 이의 항원 결합 단편을 상기 샘플에 처리하는 단계; 및(b) treating the sample with the antibody or antigen-binding fragment thereof; and
(c) 상기 피검자의 샘플 중에 포함된 CCR7의 발현양이 정상군 샘플 중에 포함된 CCR7의 발현양 보다 높은지 여부를 확인하는 단계.(c) Confirming whether the expression level of CCR7 included in the subject's sample is higher than the expression level of CCR7 included in the normal group sample.
상기 CCR7은 리간드인 CCL19 및/또는 CCL21의 결합에 의해 림프절 및 중추신경계와 같은 케모카인 발현 위치로 세포 이동을 제어하며; 방광암, 폐암, 난소암, 신장암, 대장암, 전립선암, 유방암, 자궁암, 횡문근육종, 교모세포종 등의 다양한 암에서 과발현되어 암세포의 증식, 이동, 침투, 전이 등의 주요 암진행 과정에 직접적으로 관여하기 때문에, 상기 CCR7의 발현량을 정상인과 비교함으로써, CCR7의 과발현에 의한 질환의 진단을 위한 정보를 제공할 수 있다.The CCR7 controls cell migration to chemokine expression sites such as lymph nodes and the central nervous system by binding to the ligands CCL19 and/or CCL21; It is overexpressed in various cancers such as bladder cancer, lung cancer, ovarian cancer, kidney cancer, colon cancer, prostate cancer, breast cancer, uterine cancer, rhabdomyosarcoma, and glioblastoma, and is directly involved in major cancer progression processes such as proliferation, migration, invasion, and metastasis of cancer cells. Because it is involved, by comparing the expression level of CCR7 with that of normal people, information can be provided for diagnosis of diseases caused by overexpression of CCR7.
본 발명의 바람직한 구현예에 따르면, 상기 CCR7의 과발현에 의한 질환은 암일 수 있고, 바람직하게는 전이성 암일 수 있다.According to a preferred embodiment of the present invention, the disease caused by overexpression of CCR7 may be cancer, preferably metastatic cancer.
본 발명의 또 다른 양태에 따르면, 본 발명은 상기 항체 또는 이의 항원 결합 단편을 포함하는 CCR7 정량 키트를 제공한다. According to another aspect of the present invention, the present invention provides a CCR7 quantitative kit comprising the antibody or antigen-binding fragment thereof.
본 발명의 정량 키트는 항원항체 결합반응을 통하여 상기 항체에 대한 항원을 분석함으로써 CCR7 양을 정량할 수 있으며, 상기 항원항체 결합반응은 통상의 ELISA(Enzyme-linked immunosorbent assay), RIA(Radioimmnoassay), 샌드위치 측정법(Sandwich assay), 폴리아크릴아미드 겔 상의 웨스턴 블롯(Western Blot), 면역블롯 분석(Immunoblot assay) 및 면역조직화학염색 방법(Immnohistochemical staining)으로 이루어지는 군에서 선택되는 것이 바람직하나 이에 제한되지 않는다.The quantitative kit of the present invention can quantify the amount of CCR7 by analyzing the antigen for the antibody through an antigen-antibody binding reaction, and the antigen-antibody binding reaction can be performed using conventional ELISA (Enzyme-linked immunosorbent assay), RIA (Radioimmnoassay), It is preferably selected from the group consisting of Sandwich assay, Western Blot on polyacrylamide gel, Immunoblot assay, and Immunohistochemical staining, but is not limited thereto.
항원-항체 결합 반응을 위한 고정체로는 니트로셀룰로오스 막, PVDF막, 폴리비닐(Polyvinyl) 수지 또는 폴리스티렌(Polystyrene) 수지로 합성된 웰 플레이트(Well plate) 및 유리로 된 슬라이드 글라스(Slide glass)로 이루어지는 군으로부터 선택되는 것이 사용될 수 있으나, 이제 제한되는 것은 아니다.The fixture for the antigen-antibody binding reaction consists of a nitrocellulose membrane, a PVDF membrane, a well plate synthesized from polyvinyl resin or polystyrene resin, and a slide glass made of glass. Any selected from the group may be used, but are not limited thereto.
상기 2차 항체는 발색반응을 하는 통상의 발색제로 표지되는 것이 바람직하며, HRP(Horseradish peroxidase), 알칼리성 인산분해효소(Alkaline phosphatase), 콜로이드 골드(Coloid gold), FITC(Poly L-lysine-fluorescein isothiocyanate), RITC(Rhodamine-B-isothiocyanate) 등의 형광물질(Fluorescein) 및 색소(Dye)로 이루어지는 군으로부터 선택되는 어느 하나의 표지체가 사용될 수 있다. 발색을 유도하는 기질은 발색반응을 하는 표지체에 따라 사용하는 것이 바람직하며, TMB(3,3',5,5'-tetramethyl bezidine), ABTS[2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)] 및 OPD(ophenylenediamine)로 이루어지는 군으로부터 선택되는 어느 하나를 사용하는 것이 바람직하나 이에 제한되는 것은 아니다. The secondary antibody is preferably labeled with a common coloring agent that produces a color reaction, such as HRP (Horseradish peroxidase), alkaline phosphatase, colloid gold, or FITC (Poly L-lysine-fluorescein isothiocyanate). ), RITC (Rhodamine-B-isothiocyanate), etc. Any label selected from the group consisting of a fluorescent substance (Fluorescein) and a dye (Dye) may be used. The substrate that induces color development is preferably used according to the label that produces the color reaction, such as TMB (3,3',5,5'-tetramethyl bezidine), ABTS [2,2'-azino-bis (3-ethylbenzothiazoline), -6-sulfonic acid)] and OPD (ophenylenediamine), but it is not limited thereto.
본 발명의 특징 및 이점을 요약하면 다음과 같다: The features and advantages of the present invention are summarized as follows:
(ⅰ) 본 발명은 CCR7의 세포외 부위를 항원으로 인식하여 이에 특이적으로 결합하는 항체 또는 이의 항원 결합 단편을 제공한다.(i) The present invention provides an antibody or antigen-binding fragment thereof that recognizes the extracellular region of CCR7 as an antigen and specifically binds to it.
(ⅱ) 또한, 본 발명은 CCR7의 구조적 특징을 유지한 상태에서 세포외부위에 특이적으로 결합하는 항체를 발굴함으로써, 실질적 적용에서도 CCR7의 신호전달과정을 억제하는 기능을 보이며 이 기능을 이용하여 CCR7이 관여하는 암 또는 암의 전이에 대한 치료제로서 유용하게 활용될 수 있다.(ii) In addition, the present invention discovers an antibody that specifically binds to the extracellular region while maintaining the structural characteristics of CCR7, showing the function of inhibiting the signaling process of CCR7 even in practical applications, and using this function to suppress CCR7 It can be useful as a treatment for cancer or metastasis of cancer.
도 1a는 기능성 P9-CCR7를 정제한 것으로, 각 정제 단계에서 얻은 샘플들을 Coomassie 염색을 사용하여 SDS-PAGE로 분석한 결과이다. 도 1a에서 M은 size marker이고, Load는 1% sarkosyl로 처리한 가용화된 막 분획물이며, FT는 Ni-NTA 크로마토그래피를 통과한 액(flow-through)이며, W1 및 W2는 각 0, 5mM 이미다졸이 포함된 세척 버퍼로 처리하여 얻은 분획물(wash fraction)이며, E는 Ni-NTA 크로마토그래피로부터 각각 20mM 이미다졸을 포함하는 용리버퍼로 처리한 분획물(elution fraction)이다. Figure 1a shows purified functional P9-CCR7, and shows the results of analyzing samples obtained at each purification step by SDS-PAGE using Coomassie staining. In Figure 1a, M is the size marker, Load is the solubilized membrane fraction treated with 1% sarkosyl, FT is the flow-through through Ni-NTA chromatography, and W1 and W2 are 0 and 5mM, respectively. This is the wash fraction obtained by treatment with a washing buffer containing dazole, and E is the elution fraction obtained from Ni-NTA chromatography each treated with an elution buffer containing 20mM imidazole.
도 1b는 Ni-NTA affinity 컬럼으로 정제한 P9-CCR7에 APG를 처리하여 안정화한 다음, 크기 배제 크로마토그래피를 사용하여 추가로 정제한 그래프이다. 도 1b에서 ①은 APG로 안정화된 P9-CCR7의 단분산 피크이고, ②는 유리 APG가 분리되어 나타난 피크이다. Figure 1b is a graph showing P9-CCR7 purified using a Ni-NTA affinity column, stabilized by treatment with APG, and then further purified using size exclusion chromatography. In Figure 1b, ① is the monodisperse peak of P9-CCR7 stabilized with APG, and ② is the peak that appears when free APG is separated.
도 1c는 P9-CCR7의 활성을 ELISA로 분석한 결과로, 5%의 skimmed milk 차단 용액(갈색 다이아몬드)으로 처리한 플레이트; 또는 10㎍/ml의 CCL19-His-FLAG(파란색 원) 또는 10㎍/ml의 CCL21-His-FLAG(주황색 사각형)를 고정한 플레이트를 준비하고, 여기에 다양한 농도(1~500nM)의 P9-CCR7를 적용하였다. 리간드에 대한 P9-CCR7 결합량은 항-P9 항체와 HRP가 결합된 항-마우스 Fc 항체를 이용하여 측정하였다.Figure 1c shows the results of ELISA analysis of the activity of P9-CCR7, plates treated with 5% skimmed milk blocking solution (brown diamond); Alternatively, prepare a plate immobilized with 10 μg/ml of CCL19-His-FLAG (blue circle) or 10 μg/ml of CCL21-His-FLAG (orange square), and then add P9-CCR7 at various concentrations (1 to 500 nM). was applied. The amount of P9-CCR7 binding to the ligand was measured using an anti-P9 antibody and an anti-mouse Fc antibody bound to HRP.
도 1d는 리간드의 특이적 결합활성을 ELISA로 분석한 결과로, 3%의 BSA 차단 용액(빨간색 삼각형)으로 처리한 플레이트; 또는 5㎍/ml의 APG:P9(보락색 사각형) 또는 5㎍/ml의 P9-CCR7(파란색 또는 주황색 원)로 고정한 플레이트;를 준비하고, 여기에 다양한 농도(15~1000nM)의 CCL19-His-FLAG 또는 CCL21-His-FLAG를 적용하였다. P9-CCR7에 결합하는 리간드의 양은 항-FLAG 항체와 HRP가 결합된 항-마우스 Fc 항체를 이용하여 측정하였다.Figure 1d shows the results of ELISA analysis of the specific binding activity of the ligand, showing plates treated with 3% BSA blocking solution (red triangle); Alternatively, plates immobilized with 5 μg/ml APG:P9 (purple square) or 5 μg/ml P9-CCR7 (blue or orange circle) were prepared, and various concentrations (15-1000 nM) of CCL19-His were added thereto. -FLAG or CCL21-His-FLAG was applied. The amount of ligand binding to P9-CCR7 was measured using an anti-FLAG antibody and an anti-mouse Fc antibody coupled to HRP.
도 2a는 CCR7에 특이적인 scFv 클론의 스크리닝 결과를 도시한 것으로, 1차 선별된 16종의 클론들에 대하여 CCR7에 대한 scFv의 특이성을 ELISA로 분석한 결과이다. Figure 2a shows the results of screening of scFv clones specific for CCR7. This is the result of analyzing the specificity of scFv for CCR7 for the 16 initially selected clones by ELISA.
도 2b는 농도별 scFv에 100 nM CCL19-His-FLAG를 처리하고, ELISA로 분석한 결과 그래프이다. 파란색 원과 녹색 사각형은 각각 scFv 6RG11 및 scFv 72C7에 의한 신호를 나타낸다.Figure 2b is a graph showing the results of treating scFv at each concentration with 100 nM CCL19-His-FLAG and analyzing it by ELISA. Blue circles and green squares represent signals by scFv 6RG11 and scFv 72C7, respectively.
도 2c는 0 ~ 500 nM의 scFv를 P9-CCR7에 처리하고, ELISA로 분석한 결과 그래프이다. 파란색 원과 녹색 사각형은 각각 scFv 6RG11 및 scFv 72C7에 의한 신호를 나타낸다.Figure 2c is a graph showing the results of treating P9-CCR7 with 0 to 500 nM scFv and analyzing it by ELISA. Blue circles and green squares represent signals by scFv 6RG11 and scFv 72C7, respectively.
도 3a는 정제된 IgG4(6RG11), IgG4(72C7)에 대한 SDS-PAGE 분석 결과이다. 도 3a에서 M은 크기 마커이고, NR은 비환원 조건하에서 6μg의 IgG4를 로딩한 것이며, R은 1mM DTT를 사용한 환원 조건하에서 6μg의 IgG4를 로딩한 것이다. Figure 3a shows the results of SDS-PAGE analysis of purified IgG 4 (6RG11) and IgG 4 (72C7). In Figure 3a, M is a size marker, NR is 6 μg of IgG 4 loaded under non-reducing conditions, and R is 6 μg of IgG 4 loaded under reducing conditions using 1mM DTT.
도 3b는 CCR7에 대한 인간 IgG4(6RG11), 인간 IgG4(72C7)의 특이적 결합을 ELISA로 분석한 결과이다. Figure 3b shows the results of ELISA analysis of the specific binding of human IgG 4 (6RG11) and human IgG 4 (72C7) to CCR7.
도 3c는 CCR7에 대한 인간 IgG4(6RG11)의 특이적 결합을 유세포 분석으로 분석한 결과 그래프이다.Figure 3c is a graph showing the results of flow cytometry analysis of the specific binding of human IgG 4 (6RG11) to CCR7.
도 3d는 CCR7에 대한 인간 IgG4(72C7)의 특이적 결합을 유세포 분석으로 분석한 결과 그래프이다.Figure 3d is a graph showing the results of flow cytometry analysis of the specific binding of human IgG 4 (72C7) to CCR7.
도 4 및 도 5는 cAMP assay를 통한 리간드 효과에 대한 IgG4의 길항 활성을 분석한 결과 그래프로, 도 4a는 CCR7을 발현하는 HEK293 세포에 대한 CCL19 리간드의 영향을 측정한 그래프이고, 도 4b는 CCR7을 발현하는 HEK293 세포에 대한 CCL21 리간드의 영향을 측정한 그래프이며, 도 4c는 CCR7을 발현하는 MDA-MB-231 세포에 대한 CCL19 리간드의 영향을 측정한 그래프이고, 도 4d는 CCR7을 발현하는 MDA-MB-231 세포에 대한 CCL21 리간드의 영향을 측정한 그래프이다.Figures 4 and 5 are graphs showing the results of analyzing the antagonistic activity of IgG 4 on the ligand effect through cAMP assay, Figure 4a is a graph measuring the effect of CCL19 ligand on HEK293 cells expressing CCR7, and Figure 4b is a graph This is a graph measuring the effect of CCL21 ligand on HEK293 cells expressing CCR7, Figure 4c is a graph measuring the effect of CCL19 ligand on MDA-MB-231 cells expressing CCR7, and Figure 4d is a graph measuring the effect of CCL19 ligand on MDA-MB-231 cells expressing CCR7. This is a graph measuring the effect of CCL21 ligand on MDA-MB-231 cells.
도 5a는 CCR7을 발현하는 HEK293 세포에서 CCL19 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이고, 도 5b는 CCR7을 발현하는 HEK293 세포에서 CCL21 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이며, 도 5c는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL19 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이며, 도 5d는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL21 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이다.Figure 5a is a graph measuring the effect of IgG4 (6RG11) clone on CCL19 ligand in HEK293 cells expressing CCR7, and Figure 5b is a graph measuring the effect of IgG4 (6RG11) clone on CCL21 ligand in HEK293 cells expressing CCR7. It is a graph, and Figure 5c is a graph measuring the effect of IgG4 (6RG11) clone on CCL19 ligand in MDA-MB-231 cells expressing CCR7, and Figure 5d is a graph measuring the effect of CCL21 in MDA-MB-231 cells expressing CCR7. This is a graph measuring the effect of IgG4 (6RG11) clone on the ligand.
도 6은 CCR7을 발현하는 MDA-MB-231 세포에서, CCL19 리간드에 의해 유도된 CCR7-의존적 침입(CCR7-dependent invasion)에 대한 IgG4(6RG11, 72C7)의 효과를 측정하여 나타낸 사진이다.Figure 6 is a photograph showing the effect of IgG 4 (6RG11, 72C7) on CCR7-dependent invasion induced by CCL19 ligand in MDA-MB-231 cells expressing CCR7.
도 7은 CCR7을 발현하는 MDA-MB-231 세포에서, CCL21 리간드에 의해 유도된 CCR7-의존적 침입(CCR7-dependent invasion)에 대한 IgG4(6RG11, 72C7)의 효과를 측정하여 나타낸 사진이다.Figure 7 is a photograph showing the effect of IgG 4 (6RG11, 72C7) on CCR7-dependent invasion induced by CCL21 ligand in MDA-MB-231 cells expressing CCR7.
도 8은 도 6 및 7로부터 IgG4(6RG11, 72C7)에 따른 이동 억제를 시사하는 이동된 세포의 수를 측정하여 나타낸 그래프로, 도 8a는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL19 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이고, 도 8b는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL19 리간드에 대한 IgG4(72C7) 클론의 영향을 측정한 그래프이며, 도 8c는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL21 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이며, 도 8d는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL21 리간드에 대한 IgG4(72C7) 클론의 영향을 측정한 그래프이다.Figure 8 is a graph showing the number of migrated cells measured from Figures 6 and 7, suggesting migration inhibition by IgG4 (6RG11, 72C7). Figure 8a shows the number of migrated cells in MDA-MB-231 cells expressing CCR7 to CCL19 ligand. Figure 8b is a graph measuring the effect of the IgG4 (6RG11) clone on CCL19 ligand in MDA-MB-231 cells expressing CCR7, and Figure 8c is a graph measuring the effect of the IgG4 (72C7) clone on CCR7. This is a graph measuring the effect of the IgG4 (6RG11) clone on the CCL21 ligand in MDA-MB-231 cells expressing CCR7, and Figure 8d is a graph measuring the effect of the IgG4 (72C7) clone on the CCL21 ligand in MDA-MB-231 cells expressing CCR7. This is a graph measuring the impact.
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 보다 구체적으로 설명하기 위한 것으로, 본 발명의 요지에 따라 본 발명의 범위가 이들 실시 예에 의해 제한되지 않는다는 것은 당업계에서 통상의 지식을 가진 자에 있어서 자명할 것이다Hereinafter, the present invention will be described in more detail through examples. These examples are only for illustrating the present invention in more detail, and it will be apparent to those skilled in the art that the scope of the present invention is not limited by these examples according to the gist of the present invention.
실시예Example
실험재료experiment material
CCR7 암호화 영역을 표기하는 DNA 서열은 NCBI: NM_001838.4로 표시되고, E. coli의 암호화 최적화 서열로 설계하였다. 상기 CCL19 또는 CCL21의 암호화 서열은 NCBI: NM_006274.3와 NCBI: NM_002989.4로 표시되고, FLAG 및 6X His-tag가 있는 서열로, 포유류의 코돈 최적화된 서열로 설계하였다. 상기 설계된 DNA 서열들은 LNC Bio(Korea)에 의뢰하여 합성하였다. 상기 CCR7의 표유류 발현 벡터는 Sino Biological(China)로부터 구입하여 사용하였다. 제한 효소(Restriction enzymes), DNA 폴리머라제 및 T4 DNA 리가아제는 New England Biolabs(NEB)(UK)으로부터 구입하여 사용하였다. 양친매성 폴리글루타메이트(APG)는 MPRAB(Korea)로부터 입수하였다. 합성 scFv-표시 M13 라이브러리는 이화대학교 Dr. Shim로부터 제공받아 사용하였다. CCL19와 CCL21은 Peprotech(USA)으로부터 구입하였다. HRP-conjugated anti-mouse IgG는 Sigma(USA)로부터 구입하여 사용하였고, Ni-NTA 레진은 Qiagen(Germany)로부터 구입하여 사용하였다. The HiLoad superdex 200 16/600 PG 컬럼은 GE Healthcare(USA)로부터 얻었다. CNBr-activated sepharose 4B은 GE Healthcare(USA)로부터 구입하여 사용하였다. IPTG(Isopropyl-β-D-thiogalactoside), PMSF(phenylmethylsulfonyl fluoride) 및 sarkosyl은 Sigma(USA)로부터 구입하였다. EDTA-free protease inhibitor cocktail은 GenDEPOT(USA)로부터 구입하였다. 이외에 모든 소모성 시약은 시약 등급을 사용하였고, 데이터 분석에는 GraphPad Prism 8 소프트웨어를 사용하였다.The DNA sequence representing the CCR7 coding region is designated as NCBI: NM_001838.4, and was designed as the coding optimization sequence of E. coli . The coding sequence of CCL19 or CCL21 is indicated as NCBI: NM_006274.3 and NCBI: NM_002989.4, and is a sequence with FLAG and 6X His-tag, and was designed as a mammalian codon-optimized sequence. The designed DNA sequences were synthesized by LNC Bio (Korea). The mammalian expression vector of CCR7 was purchased from Sino Biological (China). Restriction enzymes, DNA polymerase, and T4 DNA ligase were purchased from New England Biolabs (NEB) (UK). Amphipathic polyglutamate (APG) was obtained from MPRAB (Korea). The synthetic scFv-displayed M13 library was provided by Dr. Ewha University. It was used as provided by Shim. CCL19 and CCL21 were purchased from Peprotech (USA). HRP-conjugated anti-mouse IgG was purchased from Sigma (USA), and Ni-NTA resin was purchased from Qiagen (Germany). The HiLoad superdex 200 16/600 PG column was obtained from GE Healthcare (USA). CNBr-activated sepharose 4B was purchased from GE Healthcare (USA). Isopropyl-β-D-thiogalactoside (IPTG), phenylmethylsulfonyl fluoride (PMSF), and sarkosyl were purchased from Sigma (USA). EDTA-free protease inhibitor cocktail was purchased from GenDEPOT (USA). In addition, all consumable reagents were of reagent grade, and GraphPad Prism 8 software was used for data analysis.
<실시예 1> P9-CCR7, CCL19, CCL12 및 IgG4 발현벡터 제조<Example 1> Preparation of P9-CCR7, CCL19, CCL12 and IgG4 expression vectors
1) P9-CCR7 발현벡터 제조1) Preparation of P9-CCR7 expression vector
CCR7의 코딩 영역을 표시하는 DNA 서열은 정방향 프라이머(5'TATTTTCAGTCGACGATGGAATTCATGGATCTGGGTAAACCAATGAAG ?? 3'; 서열번호 21) 및 역방향 프라이머(5' - GTGATGGTGAGAAGCTTCGAATTCTGGAGAAAAGGTGGTAGTAGTTTC ?? 3'; 서열번호 22)를 사용하여 PCR 반응을 통해 증폭하였다. 상기 PCR 증폭산물은 QIAquick® PCR Purification Kit(Qiagen, Germany)을 사용하여 정제하고, 제한효소 Sal I Hind III를 사용하여 절단하였다. 이를 QIAquick® PCR Purification Kit를 사용하여 정제한 후, pP9 벡터의 Sal I, Hind III 부위에 연결하였다. 이때, 항체 발굴에 필요한 pP9 벡터는 이광규 논문(Kwangkyu Lee, et al., Purification and characterization of recombinant human endothelin receptor type A, Protein expression and purification 2012; 84(1): 14-18)을 참고로 하였다.The DNA sequence representing the coding region of CCR7 was amplified through PCR reaction using forward primer (5'TATTTTCAGTCGACGATGGAATTCATGGATCTGGGTAAACCAATGAAG ?? 3'; SEQ ID NO. 21) and reverse primer (5' - GTGATGGTGAGAAGCTTCGAATTCTGGAGAAAAGGTGGTAGTAGTTTC ?? 3'; SEQ ID NO. 22). did. The PCR amplification product was purified using the QIAquick® PCR Purification Kit (Qiagen, Germany) and digested using restriction enzymes Sal I and Hind III . This was purified using the QIAquick® PCR Purification Kit and then ligated to the Sal I and Hind III sites of the pP9 vector. At this time, the pP9 vector required for antibody discovery was referenced in the paper by Kwangkyu Lee, et al. (Purification and characterization of recombinant human endothelin receptor type A, Protein expression and purification 2012; 84(1): 14-18).
2) CCL19 및 CCL12 발현벡터 제조2) Preparation of CCL19 and CCL12 expression vectors
FLAG 및 6X His-tag을 갖는 CCL19 또는 CCL21의 암호화 영역을 표시하는 DNA 단편을 하기 정방향 프라이머와 역방향 프라이머를 사용하여 PCR 반응으로 증폭하였다:CCL19의 정방향 프라이머(5'-TCCAGCCTCCGGACTCTAGAGCCGCCACCATGGCCCTGCT-3'; 서열번호 23) 및 역방향 프라이머(5'-CCGGCCTTGCCGGCCTCGAGTCATTACTTGTCGTCATCGT - 3'; 서열번호 24), CCL21의 정방향 프라이머(5'-TCCAGCCTC CGGACTCTAGAGCCGCCACCATGGCTCAGTC-3'; 서열번호 25) 및 역방향 프라이머(5' - ATCCGG CCTTGCCGGCCTCGAGTCATTACTTGTCGTCATC-3'; 서열번호 26)A DNA fragment representing the coding region of CCL19 or CCL21 with FLAG and 6 23) and reverse primer (5'-CCGGCCTTGCCGGCCTCGAGTCATTACTTGTCGTCATCGT - 3'; SEQ ID NO. 24), forward primer of CCL21 (5'-TCCAGCCTC CGGACTCTAGAGCCGCCACCATGGCTCAGTC-3'; SEQ ID NO. 25) and reverse primer (5'-ATCCGG CCTTGCCGGCCTCGAGTCATTACTTGTCGTCATC-3'; SEQ ID NO: 26)
상기 PCR 증폭산물은 QIAquick® PCR Purification Kit으로 정제하였고, pCDNA 3.4 vector(NEB, UK)의 Xba I Xho I 부위에 연결하였다.The PCR amplification product was purified using the QIAquick® PCR Purification Kit and ligated into the Xba I and Xho I sites of pCDNA 3.4 vector (NEB, UK).
3) IgG4 발현벡터 제조3) Production of IgG4 expression vector
가변 영역 VL 및 VH를 코딩하는 서열을 PCR 반응을 통해 증폭하였다, 각 경쇄(light chain)와 중쇄(heavy chain)를 위한 신호 서열은 경쇄 신호서열(5'-GCCGCCACCATGGCCGGCTTCCCTCTCCTCCTCACCCTCCTCACTCACTGTGCAGGATCCTGGGCCCAGTCTGTGCTGACTCAGCCACCC-3'; 서열번호 27)과 중쇄 신호서열(5'- GCCGCCACCATGGAGTTTGGGCTGAGCTGGCTTTTTCTTGTGGCTATTTTAAAAGGTGTCCAGTGTGAGGTGCAGCTGTTGGAGTCTGGG-3'; 서열번호 28)로 중첩 연장 PCR 반응(overlap extension PCR reaction)으로 연결시켰다. 이는 Cosmogenetech(Korea)에 의뢰하여 합성 올리고머를 제작하였다. 각각의 산물은 겔 추출키트(Qiagen, Germany)를 사용하여 정제하였고, pTRIOZ-hIgG4(S228P) 벡터(Invivogen, USA)에서 VL은 Sgr AI, Kpn I 부위에 연결하고, VH는 Nhe I, Age I 부위에 연결하였다. 이때 NEBuilder® HiFi DNA Assembly (NEB, UK)를 사용하였다.Sequences encoding the variable regions VL and VH were amplified through PCR reaction. The signal sequence for each light chain and heavy chain was the light chain signal sequence (5'-GCCGCCACCATGGCCGGCTTCCCTCTCCTCCTCACCCTCCTCACTCACTGTGCAGGATCCTGGGCCCAGTCTGTGCTGACTCAGCCACCC-3'; SEQ ID NO. 27) and heavy chain signal sequence (5'- GCCGCCACCATGGAGTTTGGGCTGAGCTGGCTTTTTCTTGTGGCTATTTTAAAAGGTGTCCAGTGTGAGGTGCAGCTGTTGGAGTCTGGG-3'; SEQ ID NO. 28) through an overlap extension PCR reaction. This was commissioned by Cosmogenetech (Korea) to produce a synthetic oligomer. Each product was purified using a gel extraction kit (Qiagen, Germany), and in the pTRIOZ-hIgG4(S228P) vector (Invivogen, USA), VL was linked to the Sgr AI and Kpn I sites, and VH was linked to Nhe I and Age I. connected to the area. At this time, NEBuilder® HiFi DNA Assembly (NEB, UK) was used.
<실시예 2> P9-CCR7 정제<Example 2> P9-CCR7 purification
pP9-CCR7 벡터를 포함하는 E. coli BL21(DE3)-RIPL 세포를 생물반응기(Marado-PDA, CNS, Korea)를 사용하여 배양하였다. 상기 배양물의 OD600가 60일 때, 18 ℃에서 21시간 동안 최종 1 mM IPTG를 첨가하여 P9-CCR7의 발현을 유도하였다. 상기 세포를 원심분리를 통해 회수하고, -80 ℃에서 보관하였다. 세포 페이스트(paste)의 20 g을 1 mM PMSF와 1정(tablet)의 EDTA-free protease inhibitor cocktail를 포함하는 버퍼 A(25 mM HEPES, pH 7.0) 100 ml에 재분산하였고, 미세유체(model and company)를 사용하여 용해하였다. 12,000 x g에서 30 분 동안 원심분리하여 세포 파편을 제거하고, 상층액을 100,000 x g에서 1 시간동안 추가로 원심분리하여 막 분획(membrane fraction)으로 침전하였다. E. coli BL21(DE3)-RIPL cells containing the pP9-CCR7 vector were cultured using a bioreactor (Marado-PDA, CNS, Korea). When the OD 600 of the culture was 60, the expression of P9-CCR7 was induced by adding 1 mM IPTG for 21 hours at 18°C. The cells were recovered through centrifugation and stored at -80°C. 20 g of the cell paste was redispersed in 100 ml of buffer A (25 mM HEPES, pH 7.0) containing 1 mM PMSF and 1 tablet of EDTA-free protease inhibitor cocktail, and incubated with microfluidics (model and company) was used to dissolve it. Cell debris was removed by centrifugation at 12,000 xg for 30 minutes, and the supernatant was further centrifuged at 100,000 xg for 1 hour to precipitate as a membrane fraction.
상기 막분획물(membrane fraction)을 버퍼 B(25 mM HEPES pH 7.0, 1% sarkosyl)에 넣고, 4 ℃에서 2 시간동안 부드럽게 교반하여 용해한 후, 15,000 x g에서 30분 동안 원심분리하여 침전물질을 제거하였다. 상기 막분획물 용해액을 버퍼 A로 평형화된 Ni-NTA 컬럼에 로딩한 후, 결합된 P9-CCR7을 20mM 이미다졸로 용리(elute)시켰다. 용리 분획물(elution fraction)에 1% APG를 첨가하고 4 ℃에서 16시간 동안 혼합하였다. P9-CCR7을 APG로 안정화하고, 버퍼 A로 평형화된 크기 배제 크로마토그래피(HiLoad superdex 200 16/600 PG)로 추가 정제한 다음, -80 ℃에서 보관하였다.The membrane fraction was placed in buffer B (25mM HEPES pH 7.0, 1% sarkosyl), dissolved by gentle stirring for 2 hours at 4°C, and then centrifuged at 15,000 x g for 30 minutes to remove precipitates. . The membrane fraction solution was loaded onto a Ni-NTA column equilibrated with buffer A, and bound P9-CCR7 was eluted with 20mM imidazole. 1% APG was added to the elution fraction and mixed for 16 hours at 4°C. P9-CCR7 was stabilized with APG, further purified by size exclusion chromatography (HiLoad superdex 200 16/600 PG) equilibrated with buffer A, and stored at -80°C.
<실시예 3> CCL19 및 CCL21 정제<Example 3> CCL19 and CCL21 purification
상기 CCL19-His-FLAG 및 CCL21-His-FLAG 발현 벡터를 Expi 293TM media using ExpiFectamineTM 293 transfection kit(Thermo Fisher Scientific, USA)를 사용하여 Expi 293TM 배지에 배양한 Expi 293 세포(3 × 106 cells/mL)(ATCC, USA) 50 ml에 형질감염하였다. 상기 형질감염된 세포는 세포 생존율 최소 60%에 도달할때까지 배양하고, 2000 x g로 4 ℃에서 20 분동안 원심분리하여 수확하였다.Expi 293 cells ( 3 × 10 6 cells/mL) (ATCC, USA) were transfected in 50 ml. The transfected cells were cultured until cell viability reached at least 60% and harvested by centrifugation at 2000 xg at 4°C for 20 minutes.
상기 상층액은 실린지 필터(PES, Pore 0.22 μm)(Sartorius, USA)을 사용하여 필터링하고, DPBS(137 mM NaCl, 2.7 mM KCl, Na2HPO4 10 mM, KH2PO4 1.8 mM, pH 7.4)로 평형화한 Ni-NTA 컬럼으로 로딩하였다. The supernatant was filtered using a syringe filter (PES, Pore 0.22 μm) (Sartorius, USA), and filtered using DPBS (137 mM NaCl, 2.7 mM KCl, Na 2 HPO 4 10 mM, KH 2 PO 4 1.8 mM, pH 7.4) was loaded onto a Ni-NTA column equilibrated.
이미다졸(imidazole) 농도구배를 사용하여 컬럼을 세척한 후, 표적 단백질을 평형화 버퍼에 100 mM, 200 mM 이미다졸을 사용하여 용리하였다. 상기 정제된 단백질을 DPBS, 10% glycerol으로 Pd-10 컬럼(GE Healthcare, USA)을 사용하여 탈염한 다음, 용리된 단백질은 -80 ℃에 보관하였다.After washing the column using an imidazole concentration gradient, the target protein was eluted using 100 and 200 mM imidazole in equilibration buffer. The purified protein was desalted with DPBS and 10% glycerol using a Pd-10 column (GE Healthcare, USA), and the eluted protein was stored at -80°C.
<실시예 4> APG에 의하여 능동적으로 접힌 구조를 갖는 P9-CCR7의 제조<Example 4> Preparation of P9-CCR7 with an actively folded structure by APG
대장균(E. coli) 에서의 P9 발현 시스템은 인간 CCR7의 발현을 위해 성공적으로 적용되었다. 상기 P9 서열은 가로지르는 형태로서 E. coli의 원형질막에서의 CCR7 발현을 돕는다. wet E. coli 세포 1 g의 막 분획물에서 P9-CCR7 0.8 mg이 발현되었다(Western blot data as supplementary data). wet 세포 20 g으로부터 얻은 막 분획물에서 P9-CCR7은 sarkosyl로 효율적으로 가용화되고, Ni-NTA affinity 컬럼을 사용하여 P9-CCR7 8 mg을 정제하였다(도 1a). 그 결과 성공적으로 정제되었음을 확인하였다.The P9 expression system in E. coli was successfully applied for the expression of human CCR7. The P9 sequence is in a transversal form and helps CCR7 expression in the plasma membrane of E. coli . In the membrane fraction of 1 g of wet E. coli cells, 0.8 mg of P9-CCR7 was expressed (Western blot data as supplementary data). In the membrane fraction obtained from 20 g of wet cells, P9-CCR7 was efficiently solubilized with sarkosyl, and 8 mg of P9-CCR7 was purified using a Ni-NTA affinity column (Figure 1a). As a result, it was confirmed that it was successfully purified.
다음으로 APG를 처리하여 안정화한 APG-stabilized P9-CCR7를 겔 여과 크로마토그래피로 측정한 결과, 단분산피크(Molecular Weight: 347 kDa)와 분리된 유리 APG(Molecular Weight: 30 kDa)를 확인하였다(도 1b). 최종 APG와 결합하여 안정화된 P9-CCR7 5 mg을 E. coli의 막분획물로부터 정제한다는 것을 확인하였다. 이는 습윤 세포 20 g에서 발현된 P9-CCR7가 31%인 것을 의미한다.Next, APG-stabilized P9-CCR7 stabilized by treatment with APG was measured by gel filtration chromatography, and a monodisperse peak (Molecular Weight: 347 kDa) and separated free APG (Molecular Weight: 30 kDa) were confirmed ( Figure 1b). It was confirmed that 5 mg of P9-CCR7 stabilized by binding to the final APG was purified from the membrane fraction of E. coli . This means that P9-CCR7 expressed in 20 g of wet cells was 31%.
즉, E. coli로부터 전장 hCCR7을 정제하고, 이를 P9와 APG 폴리머를 사용하여 APG:P9-CCR7을 성공적으로 제조할 수 있다는 것을 확인하였다.In other words, it was confirmed that full-length hCCR7 could be purified from E. coli and successfully produce APG:P9-CCR7 using P9 and APG polymer.
<실시예 5> P9-CCR7에 대한 CCL19 및 CCL21의 친화도 측정(Affinity determination)<Example 5> Affinity determination of CCL19 and CCL21 for P9-CCR7
정제된 P9-CCR7이 적절하게 접힌 형태를 형성하고 있는지를 확인하기 위하여, 상기 정제된 P9-CCR7와 리간드의 결합 친화도를 ELISA 방법으로 측정하였다. In order to confirm whether the purified P9-CCR7 was in an appropriately folded form, the binding affinity between the purified P9-CCR7 and the ligand was measured using ELISA.
구체적으로, 상기 정제된 CCL19-His-FLAG 및 CCL21-His-FLAG을 Maxi-결합 96-웰 플레이트(SPL, USA)에 고정하였다. 각 웰은 5% skimmed milk를 포함하는 DPBS로 차단한 다음, 정제된 P9-CCR7를 다양한 농도로 플레이트에 첨가하였다. 이어서 상기 플레이트는 항-P9 항체, HRP-conjugated anti-mouse IgG, ultra TMB(Thermo fisher scientific) 및 1 N HCl을 순차적으로 처리하였다. 상기 리간드에 결합한 P9-CCR7의 양은 Synergy H1(BioTek, USA) microplate reader를 사용하여 측정하였다(도 1c).Specifically, the purified CCL19-His-FLAG and CCL21-His-FLAG were immobilized on a Maxi-binding 96-well plate (SPL, USA). Each well was blocked with DPBS containing 5% skimmed milk, and then purified P9-CCR7 was added to the plate at various concentrations. The plate was then sequentially treated with anti-P9 antibody, HRP-conjugated anti-mouse IgG, ultra TMB (Thermo fisher scientific), and 1 N HCl. The amount of P9-CCR7 bound to the ligand was measured using a Synergy H1 (BioTek, USA) microplate reader (Figure 1c).
상기 정제된 P9-CCR9 및 CCL19 또는 CCL21의 상호작용은 modified ELISA 방법을 사용하여 측정하였다. 정제된 P9-CCR7의 500 ng을 Maxi-결합된 96-웰 플레이트에 고정하였다. 이후 플레이트를 세척하고, 블로킹하고, 상기 CCL19-His-FLAG 또는 CCL21-His-FLAG의 다양한 농도를 플레이트에 처리하였다. 상기 플레이트에 항-FLAG M2 항체(Sigma-Aldrich, USA), HRP-conjugated anti-mouse IgG, ultra TMB(Thermo Fisher Scientific) 및 1 N HCl을 순차적으로 처리하였다. 리간드에 결합된 P9-CCR7의 양은 마이크로플레이트 리더기를 사용하여 측정하였다(도 1d).The interaction between the purified P9-CCR9 and CCL19 or CCL21 was measured using a modified ELISA method. 500 ng of purified P9-CCR7 was immobilized in a Maxi-bound 96-well plate. The plates were then washed, blocked, and treated with various concentrations of CCL19-His-FLAG or CCL21-His-FLAG. The plate was sequentially treated with anti-FLAG M2 antibody (Sigma-Aldrich, USA), HRP-conjugated anti-mouse IgG, ultra TMB (Thermo Fisher Scientific), and 1 N HCl. The amount of P9-CCR7 bound to the ligand was measured using a microplate reader (Figure 1d).
도 1c는 P9-CCR7의 활성을 ELISA로 분석한 결과로, 5%의 skimmed milk 차단 용액(갈색 다이아몬드)으로 처리한 플레이트; 또는 10㎍/ml의 CCL19-His-FLAG(파란색 원) 또는 10㎍/ml의 CCL21-His-FLAG(주황색 사각형)를 고정한 플레이트;를 준비하고, 여기에 다양한 농도(1~500nM)의 P9-CCR7를 적용하였다. 리간드에 대한 APG:P9-CCR7 결합량은 항-P9 항체와 HRP가 결합된 항-마우스 Fc 항체를 이용하여 측정하였다.Figure 1c shows the results of ELISA analysis of the activity of P9-CCR7, plates treated with 5% skimmed milk blocking solution (brown diamond); Alternatively, plates immobilized with 10 μg/ml of CCL19-His-FLAG (blue circle) or 10 μg/ml of CCL21-His-FLAG (orange square) were prepared, and various concentrations (1 to 500 nM) of P9- CCR7 was applied. The amount of APG:P9-CCR7 binding to the ligand was measured using an anti-P9 antibody and an anti-mouse Fc antibody conjugated to HRP.
도 1d는 리간드의 특이적 결합활성을 ELISA로 분석한 결과로, 3%의 BSA 차단 용액(빨간색 삼각형)으로 처리한 플레이트; 또는 5㎍/ml의 APG:P9(보락색 사각형) 또는 5㎍/ml의 P9-CCR7(파란색 또는 주황색 원)로 고정한 플레이트를 준비하고, 여기에 다양한 농도(15~1000nM)의 CCL19-His-FLAG 또는 CCL21-His-FLAG를 적용하였다. P9-CCR7에 결합하는 리간드의 양은 항-FLAG 항체와 HRP가 결합된 항-마우스 Fc 항체를 이용하여 측정하였다.Figure 1d shows the results of ELISA analysis of the specific binding activity of the ligand, showing plates treated with 3% BSA blocking solution (red triangle); Alternatively, prepare plates immobilized with 5 μg/ml APG:P9 (purple squares) or 5 μg/ml P9-CCR7 (blue or orange circles), and then add CCL19-His- at various concentrations (15 to 1000 nM). FLAG or CCL21-His-FLAG was applied. The amount of ligand binding to P9-CCR7 was measured using an anti-FLAG antibody and an anti-mouse Fc antibody coupled to HRP.
도 1c 및 도 1d에 나타난 바와 같아, 정제된 P9-CCR7에 대한 CCL19 또는 CCL21의 해리상수(The dissociation constants)(KDs)는 각각 19 nM, 11 nM로 측정되었다. 이를 통해 CCR7+ 세포를 사용한 BRET(bioluminescence resonance energy transfer) 분석에 의해 측정된 EC50 수치와 유사한 것으로 확인되었다. 따라서 P9-CCR7 융합단백질의 CCR7 영역이 천연 형태를 가지며 리간드 결합에 매우 적합하다는 것을 나타낸다. 상기 정제된 단백질은 바이오패닝 과정을 위한 항원으로 사용되었다.As shown in Figures 1c and 1d, the dissociation constants (K D s) of CCL19 or CCL21 with respect to purified P9-CCR7 were measured to be 19 nM and 11 nM, respectively. This confirmed that it was similar to the EC 50 value measured by BRET (bioluminescence resonance energy transfer) analysis using CCR7+ cells. Therefore, this indicates that the CCR7 region of the P9-CCR7 fusion protein has a native form and is highly suitable for ligand binding. The purified protein was used as an antigen for the biopanning process.
<실시예 6> CCR7에 특이적 항체 발굴<Example 6> Discovery of antibodies specific to CCR7
1) 바이오패닝을 통한 항체 발굴(1차 선별)1) Antibody discovery through biopanning (primary screening)
상기 고정된 P9-CCR7을 사용한 합성 scFv 파지 라이브러리의 바이오패닝 기법을 이용하여 CCR7 특이적 항체를 발굴하고자 하였다. 이를 위해 우선, 상기 정제된 P9-CCR7을 제조사의 지시에 따라 CNBr-activated Sepharose resin(Cytiva, USA)을 사용하여 비드 표면에 고정하였다. 요약하자면, 상기 레진 파우더(40 μl)는 1 mM HCl(pH 3.0)로 헹궈낸 다음, 결합버퍼(100 mM sodium bicarbonate pH 8.2)로 세척하고, 정제된 P9-CCR7(0.2 mg)와 함께 4 ℃에서 밤새 배양하였다. 상기 P9-CCR7이 고정된 레진에 종결 버퍼(100 mM Tris-Cl pH 8.0)를 1 시간동안 실온에서 처리하고, 원심분리 튜브 필터(Corning, USA)를 사용하여 DPBS(Dulbecco's phosphate buffered saline)로 세척하였다. 상기 바이오패닝의 첫 번째 단계는 4 ℃에서 14 시간동안 scFv-displaying M13 파지 라이브러리(~1 × 1012 CFU/ml) 0.20 ml와 함께 P9-CCR7 고정된 비드 20 μl를 배양하는 것으로 시작한다. 다음 결합되지 않은 파지를 DPBS로 3회 세척하고, 상기 결합 파지를 용리 버퍼(100mM glycine pH 2.2, 1% BSA) 200 μl로 상온에서 10분 동안 배양하여 용리하고, 중화 버퍼(1 M Tris-Cl, pH 8.0) 40 μl를 처리하였다. ER2738 세포에 대한 용리된 파지의 감염과 M13K07 헬퍼 파지를 사용하여 용리된 파지의 레스큐 과정은 Yang, H.Y., et al[Yang, H.Y., et al., Construction of a large synthetic human scFv library with six diversified CDRs and high functional diversity. Mol Cells, 2009. 27(2): p. 225-35.]를 참고하여 수행하였다. 용리된 파지를 사용하여 바이오패닝 과정은 상기 과정을 반복하되, 세척 횟수를 증가시키면서 반복하였다. 4-7번째 패닝 단계에서, 파지용액과 함께 P9-GLP1R(0.1 mg/ml)을, P9-CCR7-고정된 비드에 첨가하여 P9 특이적 파지의 농축을 방지하였다. 상술한 과정을 통해 총 384종의 클론을 1차 선별하였다(표 1).We attempted to discover CCR7-specific antibodies using the biopanning technique of the synthetic scFv phage library using the immobilized P9-CCR7. For this purpose, first, the purified P9-CCR7 was immobilized on the bead surface using CNBr-activated Sepharose resin (Cytiva, USA) according to the manufacturer's instructions. Briefly, the resin powder (40 μl) was rinsed with 1 mM HCl (pH 3.0), washed with binding buffer (100 mM sodium bicarbonate pH 8.2), and incubated with purified P9-CCR7 (0.2 mg) at 4 °C. was cultured overnight. The resin on which P9-CCR7 was immobilized was treated with termination buffer (100 mM Tris-Cl pH 8.0) at room temperature for 1 hour, and washed with DPBS (Dulbecco's phosphate buffered saline) using a centrifuge tube filter (Corning, USA). did. The first step of biopanning begins with incubating 20 μl of P9-CCR7 immobilized beads with 0.20 ml of scFv-displaying M13 phage library (~1 × 10 12 CFU/ml) for 14 hours at 4°C. Next, the unbound phage was washed three times with DPBS, and the bound phage was eluted by incubating with 200 μl of elution buffer (100mM glycine pH 2.2, 1% BSA) at room temperature for 10 minutes, and neutralization buffer (1 M Tris-Cl). , pH 8.0) was treated with 40 μl. Infection of eluted phage into ER2738 cells and rescue process of eluted phage using M13K07 helper phage were performed as described in Yang, HY, et al. [Yang, HY, et al., Construction of a large synthetic human scFv library with six diversified CDRs. and high functional diversity. Mol Cells, 2009. 27(2): p. 225-35.]. The biopanning process using the eluted phage was repeated while increasing the number of washes. In the 4th to 7th panning steps, P9-GLP1R (0.1 mg/ml) along with the phage solution was added to the P9-CCR7-immobilized beads to prevent enrichment of P9-specific phage. A total of 384 clones were initially selected through the above-described process (Table 1).
2) CCR7에 특이적 scFv 클론 스크리닝(2차 선별)2) CCR7-specific scFv clone screening (secondary screening)
바이오패닝 후 용출된 파지를 mid-log phase ER2738에 감염시키고, 감염된 세포를 카르베니실린(carbenicillin)을 함유하는 LB/Agar 플레이트에 분주(spread)하였다. 단일 콜로니를 딥 96 웰 플레이트의 카르베니실린(carbenicillin)을 포함하는 SB 배지 750 ㎕에서 각각 접종한 후, 탁해질 때가지 약 4시간동안 37 ℃에서 배양하였다. 최종 1 mM 농도의 IPTG를 첨가하고 180 rpm, 30 ℃ 조건에서 밤새 진탕배양하였다. 상기 딥 웰 플레이트를 3,500 x g에서 20분 동안 원심분리한 후, 상층액을 제거하고, 얼음위에서 냉각시켰다. scFv를 함유하는 주변세포질 분획물(periplasmic fraction)을 회수하기 위하여, 세포 펠렛을 1X TES(50 mM Tris-Cl pH 8.0, 1 mM EDTA, 20% sucrose) 완충액 160 ㎕에 재현탁시키고, 0.2X TES 완충액 240 ㎕을 첨가하였다. 얼음 위에서 30분 동안 배양한 후, 상기 플레이트를 원심분리하여 주변세포질 분획물(periplasmic fraction)을 얻었다. 주변 세포질 추출물 100 ㎕을 각각 표적 GPCR-고정화된 96 웰 Maxi 결합 플레이트(target GPCR-immobilized 96 well Maxi binding plate)에 처리하고, 실온에서 1 시간동안 배양하였다. 다음으로 HRP 결합 항-HA 항체(1:3000 희석), TMB 용액 및 1N HCl 용액을 순차적으로 처리하여 항원 결합 클론을 선별하였다. 1) 단계로부터 얻은 384종의 클론 중에서 항원과 결합하는 50종의 히트 클론을 분리하였고, 이들 중에서 고유한 클론 16종을 서열 분석을 통해 확인하였다(표 1).After biopanning, the eluted phage was infected with mid-log phase ER2738, and the infected cells were spread on LB/Agar plates containing carbenicillin. A single colony was inoculated in 750 ㎕ of SB medium containing carbenicillin in a deep 96-well plate, and then cultured at 37°C for about 4 hours until it became turbid. IPTG at a final concentration of 1 mM was added and cultured with shaking at 180 rpm and 30°C overnight. The deep well plate was centrifuged at 3,500 x g for 20 minutes, the supernatant was removed, and cooled on ice. To recover the periplasmic fraction containing scFv, the cell pellet was resuspended in 160 μl of 1X TES (50 mM Tris-Cl pH 8.0, 1 mM EDTA, 20% sucrose) buffer and 0.2 240 μl was added. After incubation on ice for 30 minutes, the plate was centrifuged to obtain a periplasmic fraction. 100 ㎕ of the periplasmic extract was applied to each target GPCR-immobilized 96 well Maxi binding plate and incubated at room temperature for 1 hour. Next, antigen-binding clones were selected by sequentially treating HRP-conjugated anti-HA antibody (1:3000 dilution), TMB solution, and 1N HCl solution. Among the 384 clones obtained from step 1), 50 hit clones binding to the antigen were isolated, and among these, 16 unique clones were confirmed through sequence analysis (Table 1).
3) scFv 정제 및 CCR7에 대한 특이적 scFv 발굴(3차 선별)3) Purification of scFv and discovery of scFv specific for CCR7 (3rd screening)
2차 선별된 16종의 클론들 중에서 CCR7에 대한 특이적 scFv를 발굴하고자 하였다. 이를 위해 2차 선별된 16종의 클론으로부터 scFv를 정제하였다. 개별 배양 세포에서 추출한 플라스미드 DNA를 TOP10F 컴피턴트 세포로 형질전환시켰다. 0.05 mg/ml 카르베니실린을 함유하는 SB 배지에서 10 ml의 종자 배양물을 37℃에서 밤새 인큐베이션하였다. 배양물을 카르베니실린을 함유하는 500 mL의 SB 배지로 옮기고 OD600이 0.6-0.8에 도달할 때까지 37℃에서 배양하였다. 배양액에 최종 1mM의 IPTG를 첨가한 후, 30℃에서 밤새 추가로 인큐베이션하였다. 3,500 x g에서 20분 동안 원심분리하여 세포를 수확하고 상층액을 버렸다. 35ml의 차가운 1X TES 완충액을 세포 펠렛에 첨가하고 재현탁시켰다. 그런 다음, 0.25ml의 라이소자임(40 mg/ml)을 세포에 첨가하고 혼합물을 30분 동안 얼음 위에 두었다. 0.25ml의 1M MgCl2를 첨가하여 혼합물의 EDTA를 비활성화하고 얼음 위에서 10분 동안 인큐베이션하였다. 용액을 10,000 x g에서 20분 동안 원심분리하고 상청액을 DPBS로 미리 평형화된 Ni-NTA 수지 1 ml에 로딩하고 혼합물을 4℃에서 1시간 동안 인큐베이션하였다. 반응된 레진을 5ml의 일회용 컬럼(Thermo Scientific, USA)에 패킹하고 5mM 이미다졸이 함유된 DPBST(0.05% Tween-20) 10ml로 세척하였다. ScFv는 200mM 이미다졸을 함유하는 DPBS로 5ml 용출되었고 용출액은 SDS-PAGE로 분석하였다. Among the 16 clones selected secondarily, we attempted to discover a specific scFv for CCR7. For this purpose, scFv was purified from 16 secondary selected clones. Plasmid DNA extracted from individual cultured cells was transformed into TOP10F competent cells. 10 ml of seed culture was incubated overnight at 37°C in SB medium containing 0.05 mg/ml carbenicillin. The culture was transferred to 500 mL of SB medium containing carbenicillin and incubated at 37°C until the OD 600 reached 0.6-0.8. After adding a final 1mM of IPTG to the culture, it was further incubated at 30°C overnight. Cells were harvested by centrifugation at 3,500 xg for 20 minutes and the supernatant was discarded. 35 ml of cold 1X TES buffer was added to the cell pellet and resuspended. Then, 0.25 ml of lysozyme (40 mg/ml) was added to the cells and the mixture was placed on ice for 30 min. EDTA in the mixture was inactivated by adding 0.25 ml of 1M MgCl 2 and incubated on ice for 10 minutes. The solution was centrifuged at 10,000 xg for 20 min, the supernatant was loaded into 1 ml of Ni-NTA resin pre-equilibrated with DPBS, and the mixture was incubated at 4°C for 1 h. The reacted resin was packed in a 5ml disposable column (Thermo Scientific, USA) and washed with 10ml of DPBST (0.05% Tween-20) containing 5mM imidazole. ScFv was eluted with 5 ml of DPBS containing 200mM imidazole, and the eluate was analyzed by SDS-PAGE.
상술한 과정을 통해 정제된 16종의 scFv(500 nM)를 APG:P9-CCR7 또는 APG:P9-GLP1R에 각각 처리하고, HRP 접합된 항-HA 항체를 사용하여 결합된 scFv를 검출하였다. 보다 구체적으로 CCR7과 16종의 scFv 간의 특이성(specificity)를 ELISA을 통해 분석하였다. 정제된 P9-CCR7(500 ng) 또는 P9-GLP1R(500 ng)을 96웰 Maxi 결합 플레이트(Maxi binding plate)에 고정하고, 3% BSA를 처리하였다. 다음, 16종의 scFv(500 nM)를 각각 처리하고, 실온에서 1시간 동안 배양하였다. CCR7 또는 GPCR에 결합된 scFv의 양을 측정하기 위하여, HRP-접합 항-HA 항체(HRP conjugated anti-HA antibody)(1:3000 diluted)를 사용하였다. 16 types of scFv (500 nM) purified through the above-described process were treated with APG:P9-CCR7 or APG:P9-GLP1R, respectively, and the bound scFv was detected using an HRP-conjugated anti-HA antibody. More specifically, the specificity between CCR7 and 16 scFvs was analyzed through ELISA. Purified P9-CCR7 (500 ng) or P9-GLP1R (500 ng) was immobilized on a 96-well Maxi binding plate and treated with 3% BSA. Next, each of the 16 types of scFv (500 nM) was treated and incubated at room temperature for 1 hour. To measure the amount of scFv bound to CCR7 or GPCR, HRP-conjugated anti-HA antibody (1:3000 diluted) was used.
도 2a는 CCR7에 특이적인 scFv 클론의 스크리닝 결과를 도시한 것으로, 16종의 클론들에 대하여 CCR7에 대한 scFv의 특이성을 ELISA로 분석한 결과이다. 도 2a에서 막대 그래프는 APG:P9-CCR7의 신호를 APG:P9-GLP1R의 신호로 나눈 값을 나타낸다.Figure 2a shows the results of screening of scFv clones specific for CCR7, and is the result of analyzing the specificity of scFv for CCR7 for 16 types of clones by ELISA. The bar graph in Figure 2A represents the signal of APG:P9-CCR7 divided by the signal of APG:P9-GLP1R.
도 2a에 나타난 바와 같이, 상기 16종의 scFv는 P9-CCR7 및 P9-GLP1R에 특이적으로 결합하였으나, APG:P9-CCR7에 대한 scFv의 흡수치(absorption values)를 APG:P9-GLP1R에 대한 scFv의 흡수치(absorption values)로 나누었을 때, 2 이상인 것은 총 7종의 클론뿐임을 확인하였다(6RG8, 6RG11, 7RB11, 7RD10, 72C1, 72C7, 72G8). 7종의 클론만이 P9-GLP1R에 비해 P9-CCR7에 대해 우수한 특이적 결합을 형성하는 것으로 확인하여 선별하였다.As shown in Figure 2a, the 16 types of scFv specifically bound to P9-CCR7 and P9-GLP1R, but the absorption values of scFv for APG:P9-CCR7 were compared to those for APG:P9-GLP1R. When divided by the absorption values of scFv, it was confirmed that only 7 clones had a total of 2 or more (6RG8, 6RG11, 7RB11, 7RD10, 72C1, 72C7, 72G8). Only 7 clones were selected as they were found to form superior specific binding to P9-CCR7 compared to P9-GLP1R.
4) 최종 선별4) Final selection
상기 3차 선별된 7종의 scFv에 대해 CCL19 리간드와의 경쟁(competitor) 특성을 분석하고자, CCL19의 결합에 중요한 역할을 수행하는, CCR7의 세포외 영역(extracellular region)과 에피토프(epitope)가 겹치는 항체를 식별하였다. CCL19-His-FLAG의 존재 하에서 P9-CCR7와 정제 scFv를 사용하여 competitive ELISA를 수행하였다.In order to analyze the competitor characteristics of the seven scFvs selected for the third time with the CCL19 ligand, an epitope overlapping with the extracellular region of CCR7, which plays an important role in the binding of CCL19, was selected. Antibodies were identified. Competitive ELISA was performed using P9-CCR7 and purified scFv in the presence of CCL19-His-FLAG.
구체적으로, 정제 P9-CCR7을 96웰 Maxi 결합 플레이트에 고정시킨 후, 3% BSA를 처리하였다. DPBS로 3회 세척한 후, 다양한 농도(4~3000 nM)의 scFv를 처리하고, 실온에서 30분 동안 배양하였다. 최종 100 nM이 되도록 CCL19-His-FLAG를 혼합한 DPBS를 처리하고, 실온에서 30분 동안 추가 배양하였다. DPBS로 세척한 후, 모노클로날 ANTI-FLAG® M2 항체(Thermo Fisher Scientific, USA)(DPBS에 1:1000 희석)를 처리하고 실온에서 1시간 동안 배양하였다. DPBST(0.05% Tween-20)로 세척한 다음 HRP가 결합된 항-Mouse Fc 항체(DPBST에 1:5000 희석), TMB 용액, 1N HCl을 순차적으로 처리하고, P9-CCR7에 결합된 CCL19-His-FLAG를 검출하였다.Specifically, purified P9-CCR7 was immobilized on a 96-well Maxi binding plate and then treated with 3% BSA. After washing three times with DPBS, scFv at various concentrations (4~3000 nM) was treated and incubated at room temperature for 30 minutes. DPBS mixed with CCL19-His-FLAG was treated to a final concentration of 100 nM, and further incubated at room temperature for 30 minutes. After washing with DPBS, the cells were treated with monoclonal ANTI-FLAG® M2 antibody (Thermo Fisher Scientific, USA) (1:1000 diluted in DPBS) and incubated for 1 hour at room temperature. Washed with DPBST (0.05% Tween-20), sequentially treated with HRP-conjugated anti-Mouse Fc antibody (1:5000 dilution in DPBST), TMB solution, 1N HCl, and CCL19-His conjugated to P9-CCR7. -FLAG was detected.
도 2b는 농도별 scFv(6RG11, 72C7)에 100 nM CCL19-His-FLAG를 처리하고, ELISA로 분석한 결과 그래프이다. 도 2b에 나타난 바와 같이, 7종의 scFv 중에서 CCR7에 대한 CCL19-경쟁적 결합을 형성하는 항체는 오직 2종의 scFv(6RG11, 72C7)이였다. 이에 scFv(6RG11, 72C7)에 대한 ELISA 그래프를 따라서 최종적으로 2종의 클론(6RG11, 72C7)을 선별하였다.Figure 2b is a graph showing the results of treating scFv (6RG11, 72C7) at different concentrations with 100 nM CCL19-His-FLAG and analyzing it by ELISA. As shown in Figure 2b, among the seven types of scFvs, only two types of scFvs (6RG11, 72C7) formed CCL19-competitive binding to CCR7. Accordingly, two types of clones (6RG11, 72C7) were finally selected according to the ELISA graph for scFv (6RG11, 72C7).
상술한 과정을 통해 총 384종의 클론을 1차 스크리닝하고, 항원과의 결합 특성을 분석하여 히트 클론 50종을 2차 선별한 후, 서열 분석을 통해 유니크한 클론 16종을 선별하였다. 이후 CCR7-특이적 클론, 리간드 결합능력 등의 추가적인 특성분석을 통해 최종적으로 2종의 클론을 선별하였으며, 각 선별과정과 선정된 클론의 수를 표 1에 정리하여 나타내었다.Through the above-described process, a total of 384 clones were first screened, 50 hit clones were secondarily screened by analyzing their binding characteristics to antigens, and then 16 unique clones were selected through sequence analysis. Afterwards, two types of clones were finally selected through additional characterization of CCR7-specific clones and ligand binding ability, and each selection process and the number of selected clones are summarized in Table 1.
Number of clonesNumber of clones
Screened clonesScreened clones 384384
Primary hitsPrimary hits 5050
Unique clonesUnique clones 1616
CCR7-specific clonesCCR7-specific clones 77
Ligand competitors Ligand competitors 22
상술한 과정을 통해 최종 선정된 2종의 scFv에 대한 서열 분석한 결과, 서열번호 17의 6RG11 scfv과 서열번호 19을 포함하는 72C7 scFv을 확인하였다(각각 중쇄 가변영역에 서열번호 1의 CDR1, 서열번호 2의 CDR2 및 서열번호 3의 CDR3 및 경쇄 가변영역에 서열번호 4의 CDR1, 서열번호 5의 CDR2 및 서열번호 6의 CDR3을 포함; 중쇄 가변영역에 서열번호 7의 CDR1, 서열번호 8의 CDR2 및 서열번호 9의 CDR3 및 경쇄 가변영역에 서열번호 10의 CDR1, 서열번호 11의 CDR2 및 서열번호 12의 CDR3;을 포함).As a result of sequence analysis of the two types of scFv finally selected through the above-described process, 6RG11 scfv of SEQ ID NO: 17 and 72C7 scFv containing SEQ ID NO: 19 were confirmed (CDR1 of SEQ ID NO: 1 in the heavy chain variable region, respectively, sequence CDR2 of SEQ ID NO: 2 and CDR3 of SEQ ID NO: 3, and the light chain variable region includes CDR1 of SEQ ID NO: 4, CDR2 of SEQ ID NO: 5, and CDR3 of SEQ ID NO: 6; CDR1 of SEQ ID NO: 7 and CDR2 of SEQ ID NO: 8 in the heavy chain variable region and CDR3 of SEQ ID NO: 9 and CDR1 of SEQ ID NO: 10, CDR2 of SEQ ID NO: 11, and CDR3 of SEQ ID NO: 12 in the light chain variable region.
<실시예 7> CCR7에 대한 최종 선별된 2종 클론의 결합력<Example 7> Binding affinity of the two final selected clones to CCR7
상기 2종의 scFv(6RG11, 72C7)은 CCR7에 농도의존적으로 결합하는지를 ELISA를 통해 확인하고자 하였다. 구체적으로 다양한 농도(0 ~ 500 nM)의 scFv(6RG11, 72C7)를 APG-O:P9-CCR7에 처리하고, 실온에서 1시간 동안 배양한 후, 결합된 scFv(6RG11, 72C7)를 측정하기 위하여 HRP-접합 항-HA 항체(HRP conjugated anti-HA antibody)(1:3000 diluted)를 사용하였다.We sought to confirm through ELISA whether the two types of scFv (6RG11, 72C7) bind to CCR7 in a concentration-dependent manner. Specifically, various concentrations (0 ~ 500 nM) of scFv (6RG11, 72C7) were treated with APG-O:P9-CCR7, incubated at room temperature for 1 hour, and then bound scFv (6RG11, 72C7) was measured. HRP-conjugated anti-HA antibody (1:3000 diluted) was used.
도 2c는 0 ~ 500 nM의 scFv(6RG11, 72C7)를 APG-O:P9-CCR7에 처리하고, ELISA로 분석한 결과 그래프이다. 파란색 원과 녹색 사각형은 각각 scFv 6RG11 및 scFv 72C7에 의한 신호를 나타낸다.Figure 2c is a graph showing the results of treating APG-O:P9-CCR7 with 0 to 500 nM scFv (6RG11, 72C7) and analyzing it by ELISA. Blue circles and green squares represent signals by scFv 6RG11 and scFv 72C7, respectively.
도 2c에 나타난 바와 같이 상기 2종의 scFv(6RG11, 72C7)은 CCR7에 대한 농도 의존적으로 결합한다는 것을 확인하였고, 각각 90 nM(6RG11), 230 nM(72C7)의 KD 수치를 갖는 것으로 확인되었다. 즉, 항원으로 정제된 P9-CCR7을 사용한 scFv-display M13 라이브러리를 통해 CCR7-특이적 활성을 갖는 scFv를 성공적으로 스크리닝하였다. 확인된 클론의 약 56%가 CCR7에 대해 비특이적이였다. 비특이적 클론들은 CCR7 도메인이 아닌 P9 서열에 결합하는 것으로 여겨진다. 비특이적 클론들은 바이오패닝 과정동안 P9 단백질의 봉입을 억제할 것으로 예상된다. As shown in Figure 2c, the two types of scFv (6RG11, 72C7) were confirmed to bind to CCR7 in a concentration-dependent manner, and were confirmed to have K D values of 90 nM (6RG11) and 230 nM (72C7), respectively. . That is, scFvs with CCR7-specific activity were successfully screened through the scFv-display M13 library using P9-CCR7 purified as an antigen. Approximately 56% of the identified clones were non-specific for CCR7. Non-specific clones are believed to bind to the P9 sequence and not to the CCR7 domain. Non-specific clones are expected to inhibit the inclusion of P9 protein during the biopanning process.
7종의 항체들 중에서 2종의 scFv(6RG11, 72C7)을 제외한 5종의 CCR7-특이적 scFv는 CCL19와 경쟁하지 못하는 것으로 확인되었다. 최종 선별되지 못한 5종의 scFv는 CCR7의 CCL19 결합부위에서 멀리 떨어져 있는 세포 내 영역 또는 세포 외 영역과 상호작용하는 것으로 여겨진다. 상기 2종의 scFv(6RG11, 72C7)은, CCL19와 CCR7에 경쟁적 결합을 형성하며, 우수한 CCR7의 리간드 결합에 대한 억제활성을 가지므로, CCR7을 표적으로 하는 치료 항체로서 최종 선별되었다.Among the 7 types of antibodies, 5 types of CCR7-specific scFvs excluding 2 types of scFvs (6RG11, 72C7) were confirmed to be unable to compete with CCL19. The five scFvs that were not finally selected are believed to interact with intracellular or extracellular regions far from the CCL19 binding site of CCR7. The above two types of scFv (6RG11, 72C7) form competitive bonds with CCL19 and CCR7 and have excellent inhibitory activity against CCR7 ligand binding, so they were finally selected as therapeutic antibodies targeting CCR7.
<실시예 8> CCR7-특이적 IgG4의 제조<Example 8> Preparation of CCR7-specific IgG 4
상기 최종 선별된 2종의 클론(6RG11, 72C7)의 가변영역 서열(variable region sequences)은 IgG4형 면역글로불린을 발현하는 인간 람다 Fc 영역 서열(human lambda Fc region sequence)을 포함하는 pTRIOZ-hIgG4(S228P) 벡터(Invitrogen, USA)로 옮겼다. ExpiFectamine™ 293 형질전환 키트(Thermo Fisher Scientific, USA)를 사용하여 Expi293™ 배지에 유지된 Expi293 세포(3 × 106 cells/ml)(ATCC, USA) 50 ml에 IgG4 발현 벡터를 형질감염시켰다. 형질감염된 세포를 세포의 생존율이 60% 미만에 도달할 때까지 배양하고, 4 ℃에서 20분 동안 2,000 x g으로 수확하였다. 상층액(supernatant)을 시린지 필터(PES, Pore 0.22μm)(Sartorius, USA)를 사용하여 여과하고 DPBS로 평형화된 CaptivA®(Repligen)에 로딩하였다. DPBS로 컬럼을 세척한 후, 표적 단백질을 100 mM Glycine-HCl pH 2.7로 용출시키고, 즉시 1M Tris-HCl pH 8.0으로 중화시켰다. 정제된 단백질은 침전을 방지하기 위해 DPBS로 희석하고, 10% 글리세롤로 용출된 단백질은 필요할 때까지 -80℃에서 보관하였다. 정제된 IgG4의 P9-CCR7에 대한 결합 활성은 ELISA를 이용하여 확인하였고, 결합된 IgG4는 Goat anti-Human IgG(H+L) HRP(Thermo Fisher Scientific, USA)로 분석하였다.The variable region sequences of the two final selected clones (6RG11, 72C7) are pTRIOZ-hIgG4 (pTRIOZ-hIgG4), which contains a human lambda Fc region sequence expressing IgG 4 type immunoglobulin. S228P) vector (Invitrogen, USA). The IgG 4 expression vector was transfected into 50 ml of Expi293 cells (3 × 10 6 cells/ml) (ATCC, USA) maintained in Expi293™ medium using the ExpiFectamine™ 293 transfection kit (Thermo Fisher Scientific, USA). Transfected cells were cultured until cell viability reached less than 60% and harvested at 2,000 xg for 20 min at 4°C. The supernatant was filtered using a syringe filter (PES, Pore 0.22 μm) (Sartorius, USA) and loaded into CaptivA® (Repligen) equilibrated with DPBS. After washing the column with DPBS, the target protein was eluted with 100 mM Glycine-HCl pH 2.7 and immediately neutralized with 1M Tris-HCl pH 8.0. Purified proteins were diluted with DPBS to prevent precipitation, and proteins eluted with 10% glycerol were stored at -80°C until needed. The binding activity of purified IgG 4 to P9-CCR7 was confirmed using ELISA, and bound IgG 4 was analyzed using Goat anti-Human IgG (H+L) HRP (Thermo Fisher Scientific, USA).
상술한 바와 같이, 6RG11, 72C7의 중쇄 가변부위와 경쇄 가변부위를 포함하는 인간 IgG4의 발현 벡터를 구축하고, 두 IgG4 단백질을 정제한 후, SDS-PAGE로 분석하였다. 구체적으로 인간 IgG4는 IgG4 발현 벡터를 포함하는 Expi 293 세포의 배양 배지에서 CaptivA 단백질 A 수지를 사용하여 정제되었고, 상기 과정을 통해 정제된 IgG4는 Coomassie 염색을 사용하여 SDS-PAGE로 분석하였다.As described above, an expression vector for human IgG 4 containing the heavy and light chain variable regions of 6RG11 and 72C7 was constructed, and the two IgG 4 proteins were purified and analyzed by SDS-PAGE. Specifically, human IgG 4 was purified from the culture medium of Expi 293 cells containing an IgG 4 expression vector using CaptivA protein A resin, and IgG 4 purified through the above process was analyzed by SDS-PAGE using Coomassie staining. .
도 3a는 정제된 IgG4(6RG11), IgG4(72C7)에 대한 SDS-PAGE 분석 결과이다. 도 3a에서 M은 크기 마커이고, NR은 비환원 조건하에서 6μg의 IgG4를 로딩한 것이며, R은 1mM DTT를 사용한 환원 조건하에서 6μg의 IgG4를 로딩한 것이다. Figure 3a shows the results of SDS-PAGE analysis of purified IgG 4 (6RG11) and IgG 4 (72C7). In Figure 3a, M is a size marker, NR is 6 μg of IgG 4 loaded under non-reducing conditions, and R is 6 μg of IgG 4 loaded under reducing conditions using 1mM DTT.
도 3a에 나타난 바와 같이 상술한 과정을 통해 성공적으로 인간 IgG4(6RG11), 인간 IgG4(72C7)를 제조 및 정제하였음을 확인하였다.As shown in Figure 3a, it was confirmed that human IgG 4 (6RG11) and human IgG 4 (72C7) were successfully produced and purified through the above-described process.
<실시예 9> 인간 IgG4(6RG11), 인간 IgG4(72C7)의 CCR7에 대한 결합특성<Example 9> Binding characteristics of human IgG 4 (6RG11) and human IgG 4 (72C7) to CCR7
본 발명에 따른 인간 IgG4(6RG11), 인간 IgG4(72C7)의 결합 동역학(binding kinetic)을 분석하기 위하여 P9-CCR7을 사용하여 ELISA 및 유세포 분석을 수행하였다.To analyze the binding kinetics of human IgG 4 (6RG11) and human IgG 4 (72C7) according to the present invention, ELISA and flow cytometry were performed using P9-CCR7.
1) P9-CCR7에 대한 IgG4의 친화성(Affinity) 분석1) Affinity analysis of IgG 4 for P9-CCR7
CCR7과 IgG4(6RG11, 72C7) 간의 결합 친화도(binding affinity)를 ELISA을 통해 분석하였다. 정제된 P9-CCR7(500 ng) 또는 P9(500 ng)을 96웰 Maxi 결합 플레이트(Maxi binding plate)에 고정하고, 3% BSA를 처리하였다. 다음, 다양한 농도(1 ~ 1000 nM)의 IgG4(6RG11, 72C7)를 처리하고, 실온에서 1시간 동안 배양하였다. 결합한 IgG4(6RG11, 72C7)의 양을 측정하기 위하여, HRP-접합 항-HA 항체(HRP conjugated anti-HA antibody)(1:3000 diluted)를 사용하여 분석하였다.The binding affinity between CCR7 and IgG 4 (6RG11, 72C7) was analyzed through ELISA. Purified P9-CCR7 (500 ng) or P9 (500 ng) was immobilized on a 96-well Maxi binding plate and treated with 3% BSA. Next, various concentrations (1 to 1000 nM) of IgG 4 (6RG11, 72C7) were treated and incubated at room temperature for 1 hour. To measure the amount of bound IgG 4 (6RG11, 72C7), it was analyzed using HRP-conjugated anti-HA antibody (1:3000 diluted).
2) 세포 배양, 형질감염(transfection) 및 유세포 분석(flow cytometry)2) Cell culture, transfection and flow cytometry
HEK293 CCR7+ 세포는 pCDNA3.1-FLAG-CCR7 DNA 플라스미드(1 ug/μL)와 함께 Lipofectamine® 3000(Thermo Fisher Scientific, USA)을 사용하여 제조하였다. HEK293 CCR7- 세포는 배양 배지(DMEM, 10% FBS, 1% penicillin /streptomycin)에서 보관하였다. 상기 세포들에 TrypLE™ Express Enzyme(Thermo Fisher Scientific, USA) 1 ml를 처리하여, 단일 세포 현탁액을 얻었고, 이를 300 x g에서 3분 동안 원심분리한 후, 상층액을 제거한 다음 펠렛(cell pellet)을 DPBS로 2회 세척하였다. 상기 세척한 세포 펠렛을 3% BSA 함유 DPBS 5 ml로 재현탁하였다. 이때 용액 내 세포의 최종 밀도가 1 × 106 cells/ml가 되도록 하였다. 상기 재현탁액을 4 ℃에서 1시간 동안 배양하고, 세포 용액 1 ml(1 × 106 세포)를 각 튜브에 넣고, 1 ml DPBS를 사용하여 헹궜다. 여기에 DPBS 또는 20 nM의 6RG11 또는 72C7 또는 Isotype control(Opdivo)을 처리하고, 4 ℃에서 1시간 동안 회전 배양한 다음, 원심분리하였다. 상층액은 버리고, 펠렛을 DPBS 1 ml로 2회 세척한 후, 모든 세포에 Alexa 488 conjugated anti-Human Fc 항체(1:1000 희석)(Invitrogen, USA) 400 ㎕를 처리하고, 4 ℃에서 1시간 동안 배양하였다. 세포를 3회 세척하고 500 ㎕의 DPBS로 재현탁하였다. 결합된 IgG4의 형광 신호는 Guava® easyCyte™ Flow Cytometer(Luminex, USA)를 사용하여 측정하였다(도 3c, d). 도면 상에서 DPBS는 적색으로, 20 nM의 Opdivo IgG4(이소타입 대조군)는 청색으로, 20 nM의 정제된 인간 IgG4(6RG11) 또는 인간 IgG4(72C7)은 주황색으로 나타내었다.HEK293 CCR7+ cells were prepared using Lipofectamine® 3000 (Thermo Fisher Scientific, USA) with pCDNA3.1-FLAG-CCR7 DNA plasmid (1 ug/μL). HEK293 CCR7- cells were maintained in culture medium (DMEM, 10% FBS, 1% penicillin/streptomycin). The cells were treated with 1 ml of TrypLE™ Express Enzyme (Thermo Fisher Scientific, USA) to obtain a single cell suspension, which was centrifuged at 300 xg for 3 minutes, the supernatant was removed, and the cell pellet was obtained. Washed twice with DPBS. The washed cell pellet was resuspended in 5 ml of DPBS containing 3% BSA. At this time, the final density of cells in the solution was set to 1 × 10 6 cells/ml. The resuspension was incubated at 4°C for 1 hour, 1 ml of cell solution (1 × 10 6 cells) was added to each tube, and rinsed using 1 ml DPBS. DPBS or 20 nM of 6RG11 or 72C7 or Isotype control (Opdivo) was added thereto, incubated with rotation at 4°C for 1 hour, and then centrifuged. The supernatant was discarded, the pellet was washed twice with 1 ml of DPBS, and then all cells were treated with 400 ㎕ of Alexa 488 conjugated anti-Human Fc antibody (1:1000 dilution) (Invitrogen, USA) and incubated at 4°C for 1 hour. cultured for a while. Cells were washed three times and resuspended in 500 μl of DPBS. The fluorescence signal of bound IgG 4 was measured using a Guava® easyCyte™ Flow Cytometer (Luminex, USA) (FIG. 3c, d). In the figure, DPBS is shown in red, 20 nM of Opdivo IgG 4 (isotype control) is shown in blue, and 20 nM of purified human IgG 4 (6RG11) or human IgG 4 (72C7) is shown in orange.
도 3b는 CCR7에 대한 인간 IgG4(6RG11), 인간 IgG4(72C7)의 특이적 결합을 ELISA로 분석한 결과이고, 도 3c는 CCR7에 대한 인간 IgG4(6RG11)의 특이적 결합을 유세포 분석으로 분석한 결과 그래프이며, 도 3d는 CCR7에 대한 인간 IgG4(72C7)의 특이적 결합을 유세포 분석으로 분석한 결과 그래프이다.Figure 3b shows the results of ELISA analysis of the specific binding of human IgG 4 (6RG11) and human IgG 4 (72C7) to CCR7, and Figure 3c shows the results of flow cytometry analysis of the specific binding of human IgG 4 (6RG11) to CCR7. This is a graph showing the results of the analysis, and Figure 3d is a graph showing the results of analyzing the specific binding of human IgG 4 (72C7) to CCR7 using flow cytometry.
도 3b에 나타난 바와 같이, 인간 IgG4(6RG11), 인간 IgG4(72C7)는 모두 P9와 결합하지 않으면서, P9-CCR7에 높은 결합 친화도(각각 KD : 40 nM, KD : 37 nM)를 갖는 것을 확인하였다. 정제된 인간 IgG4(6RG11), 인간 IgG4(72C7)의 친화도는 scFv 형태일 때보다 2.2배(6RG11), 6.5 배(72C7) 더 높은 것을 확인할 수 있다. scFv 형태와 비교하여 인간 IgG4(6RG11), 인간 IgG4(72C7) 형태일 때, 친화도가 더 향상된다는 것을 확인하였다.As shown in Figure 3b, neither human IgG 4 (6RG11) nor human IgG 4 (72C7) binds to P9, but has high binding affinity to P9-CCR7 (K D : 40 nM, K D : 37 nM, respectively). ) was confirmed to have. It can be seen that the affinities of purified human IgG 4 (6RG11) and human IgG 4 (72C7) are 2.2 times (6RG11) and 6.5 times (72C7) higher than those in scFv form. It was confirmed that affinity was further improved in the form of human IgG 4 (6RG11) and human IgG 4 (72C7) compared to the scFv form.
본 발명에 따른 인간 IgG4(6RG11), 인간 IgG4(72C7)의 CCR7에 대한 특이적 결합능을 포유동물 세포에서 확인하고자 하였다. 도 3c 및 도 3d에 나타난 바와 같이, HEK293 세포에 인간 IgG4(6RG11), 인간 IgG4(72C7) 또는 아이소타입 대조군 항체(isotype control antibody)를 처리하였을 때, 인간 IgG4(6RG11), 인간 IgG4(72C7)의 어떠한 유의한 결합도 관찰되지 않았으나(도 3c, d에서 좌측 도면), CCR7을 발현하는 HEK293 세포는 인간 IgG4(6RG11), 인간 IgG4(72C7)와 강한 결합 신호를 나타내었다(도 3c, d에서 우측 도면). 이러한 결과를 통해 인간 IgG4(6RG11), 인간 IgG4(72C7)는 포유류세포에서 CCR7의 세포외 영역(extracellular region)에 결합할 수 있다는 것을 알 수 있다.We sought to confirm the specific binding ability of human IgG 4 (6RG11) and human IgG 4 (72C7) according to the present invention to CCR7 in mammalian cells. As shown in Figures 3c and 3d, when HEK293 cells were treated with human IgG 4 (6RG11), human IgG 4 (72C7), or isotype control antibody, human IgG 4 (6RG11), human IgG Although no significant binding of 4 (72C7) was observed (left view in Figure 3c, d), HEK293 cells expressing CCR7 showed strong binding signals with human IgG 4 (6RG11) and human IgG 4 (72C7). (right view in Fig. 3c,d). These results show that human IgG 4 (6RG11) and human IgG 4 (72C7) can bind to the extracellular region of CCR7 in mammalian cells.
<실시예 10> CCR7 신호축(signaling axis)에 대한 인간 IgG4(6RG11), 인간 IgG4(72C7)의 활성 차단<Example 10> Blocking the activity of human IgG 4 (6RG11) and human IgG 4 (72C7) on the CCR7 signaling axis
1) Cyclic AMP assay1) Cyclic AMP assay
HEK293 CCR7+세포 또는 MDA-MB-231 CCR7+세포를 10% FBS 및 1% 페니실린/스트렙토마이신을 함유하는 DMEM 배지에 보관하였다. 실험 하루 전, 1 × 104 cells/well의 농도로 96웰 배양 플레이트에 각각 접종하고, 5% 이산화탄소, 37 ℃ 조건에서 밤새 배양하였다. cAMP 자극 완충액(DMEM, 0.5mM 3-이소부틸-1-메틸크산틴(IBMX))으로 배지를 교체한 후, 5% 이산화탄소, 37 ℃ 조건에서 20분 동안 배양하고, CCL19 또는 CCL21를 처리한 다음 5% 이산화탄소, 37 ℃ 조건에서 20분 동안 배양하였다. 이때 CCL19 또는 CCL21은 2 × 포스콜린 완충액(40μM의 포스콜린이 포함된 cAMP 자극 완충액)에 다양한 농도의 CCL19(Peprotech, USA) 또는 CCL21(Peprotech, USA)을 혼합하여 제조된 혼합액의 형태를 사용하였다. CCL19 리간드 또는 CCL21 리간드의 효과는 cAMP-GloTM assay kit(Promega, USA)를 사용하여 분석하였고, Synergy H1(BioTek, USA) microplate reader로 검출하였다.HEK293 CCR7 + cells or MDA-MB-231 CCR7 + cells were maintained in DMEM medium containing 10% FBS and 1% penicillin/streptomycin. One day before the experiment, each was inoculated into a 96-well culture plate at a concentration of 1 × 10 4 cells/well and cultured overnight at 37°C in 5% carbon dioxide. After replacing the medium with cAMP stimulation buffer (DMEM, 0.5mM 3-isobutyl-1-methylxanthine (IBMX)), cultured for 20 minutes at 5% carbon dioxide at 37°C, treated with CCL19 or CCL21. It was incubated for 20 minutes at 5% carbon dioxide and 37°C. At this time, CCL19 or CCL21 was used in the form of a mixture prepared by mixing various concentrations of CCL19 (Peprotech, USA) or CCL21 (Peprotech, USA) in 2 × forskolin buffer (cAMP stimulation buffer containing 40 μM forskolin). . The effect of CCL19 ligand or CCL21 ligand was analyzed using cAMP-GloTM assay kit (Promega, USA) and detected with Synergy H1 (BioTek, USA) microplate reader.
HEK293 CCR7+세포 또는 MDA-MB-231 CCR7+세포를 1 × 104 cell/well의 농도로 96웰 배양 플레이트에 분주하고 5% 이산화탄소, 37 ℃ 조건에서 밤새 배양하였다. 다양한 농도의 IgG4가 포함된 cAMP 자극 완충액(DMEM, 0.5mM 3-이소부틸-1-메틸크산틴(IBMX)) 또는 cAMP 자극 완충액으로 배지를 교체하고, 5% 이산화탄소, 37 ℃ 조건에서 20분 동안 배양하였다. 여기에 20 nM의 CCL19(Peprotech, USA) 또는 CCL21(Peprotech, USA)을 포함하는 2×포스콜린 완충액(40μM의 포스콜린이 포함된 cAMP 자극 완충액) 또는 2×포스콜린 완충액만을 세포에 처리하고 5% 이산화탄소, 37 ℃ 조건에서 20분 동안 배양하였다. CCL19 또는 CCL21 리간드 결합에 대한 IgG4의 효과 역시 cAMP-GloTM assay kit(Promega, USA)를 사용하여 분석하였고, Synergy H1(BioTek, USA) microplate reader로 검출하였다.HEK293 CCR7 + cells or MDA-MB-231 CCR7 + cells were distributed in a 96-well culture plate at a concentration of 1 × 10 4 cells/well and cultured overnight at 5% carbon dioxide and 37°C. Replace the medium with cAMP stimulation buffer (DMEM, 0.5mM 3-isobutyl-1-methylxanthine (IBMX)) or cAMP stimulation buffer containing various concentrations of IgG 4 , and incubate for 20 minutes at 5% carbon dioxide at 37°C. cultured for a while. Here, the cells were treated with 2×forskolin buffer (cAMP stimulation buffer containing 40 μM forskolin) or only 2×forskolin buffer containing 20 nM of CCL19 (Peprotech, USA) or CCL21 (Peprotech, USA) and treated for 5 days. % carbon dioxide, and incubated for 20 minutes at 37°C. The effect of IgG 4 on CCL19 or CCL21 ligand binding was also analyzed using the cAMP-GloTM assay kit (Promega, USA) and detected with a Synergy H1 (BioTek, USA) microplate reader.
CCR7에 결합하기 위한 scFv의 리간드-경쟁(ligand-competition)과 포유류 세포에서 CCR7에 대한 IgG4(6RG11, 72C7)의 특이적 결합을 통해, 본 발명에서 스크리닝된 두 종의 인간 IgG4(6RG11, 72C7)를 확인하였고, 이들은 각각 CCR7에 의해 유도된 세포 전이를 방지하고, CCR7에 대한 길항활성을 갖는다는 것을 앞선 실험을 통해 확인하였다. 나아가 본 발명의 인간 IgG4(6RG11, 72C7)가 CCR7에 대해 길항활성을 갖는지 확인하고자 하였고, CCL19 또는 CCL21에 의해 유도된 CCR7 의존성 Gi 신호전달을 cAMP-GloTM 분석 키트(Promega, USA)를 사용하여 분석하였다.Through ligand-competition of scFv for binding to CCR7 and specific binding of IgG 4 (6RG11, 72C7) to CCR7 in mammalian cells, two types of human IgG 4 (6RG11, 72C7) screened in the present invention were obtained. 72C7), and it was confirmed through previous experiments that they each prevent cell metastasis induced by CCR7 and have antagonistic activity against CCR7. Furthermore, we sought to confirm whether human IgG 4 (6RG11, 72C7) of the present invention has antagonistic activity against CCR7, and CCR7-dependent Gi signaling induced by CCL19 or CCL21 was tested using a cAMP-GloTM assay kit (Promega, USA). analyzed.
도 4 및 도 5는 cAMP assay를 통한 리간드 효과에 대한 IgG4의 길항 활성을 분석한 결과 그래프로, 도 4a는 CCR7을 발현하는 HEK293 세포에 대한 CCL19 리간드의 영향을 측정한 그래프이고, 도 4b는 CCR7을 발현하는 HEK293 세포에 대한 CCL21 리간드의 영향을 측정한 그래프이며, 도 4c는 CCR7을 발현하는 MDA-MB-231 세포에 대한 CCL19 리간드의 영향을 측정한 그래프이고, 도 4d는 CCR7을 발현하는 MDA-MB-231 세포에 대한 CCL21 리간드의 영향을 측정한 그래프이다.Figures 4 and 5 are graphs showing the results of analyzing the antagonistic activity of IgG 4 on the ligand effect through cAMP assay, Figure 4a is a graph measuring the effect of CCL19 ligand on HEK293 cells expressing CCR7, and Figure 4b is a graph This is a graph measuring the effect of CCL21 ligand on HEK293 cells expressing CCR7, Figure 4c is a graph measuring the effect of CCL19 ligand on MDA-MB-231 cells expressing CCR7, and Figure 4d is a graph measuring the effect of CCL19 ligand on MDA-MB-231 cells expressing CCR7. This is a graph measuring the effect of CCL21 ligand on MDA-MB-231 cells.
도 5a는 CCR7을 발현하는 HEK293 세포에서 CCL19 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이고, 도 5b는 CCR7을 발현하는 HEK293 세포에서 CCL21 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이며, 도 5c는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL19 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이며, 도 5d는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL21 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이다.Figure 5a is a graph measuring the effect of IgG4 (6RG11) clone on CCL19 ligand in HEK293 cells expressing CCR7, and Figure 5b is a graph measuring the effect of IgG4 (6RG11) clone on CCL21 ligand in HEK293 cells expressing CCR7. It is a graph, and Figure 5c is a graph measuring the effect of IgG4 (6RG11) clone on CCL19 ligand in MDA-MB-231 cells expressing CCR7, and Figure 5d is a graph measuring the effect of CCL21 in MDA-MB-231 cells expressing CCR7. This is a graph measuring the effect of IgG4 (6RG11) clone on the ligand.
도 4에 나타난 바와 같이, CCR7을 발현하는 MDA-MB-231 또는 HEK293 세포에 다양한 농도의 CCL19, CCL21 리간드를 처리한 결과 cAMP 축적을 확인하였다. 이때, HEK293 세포에서는 CCL19와 CCL21 리간드의 최소농도가 0.1 nM이였고, MDA-MB-231 세포에서는 1 nM이였다. 최대 cAMP 축적이 발생한 CCL19와 CCL21 농도는 HEK293 세포에서는 10 nM이였고, MDA-MB-231 세포에서는 50-100 nM이였다.As shown in Figure 4, cAMP accumulation was confirmed when MDA-MB-231 or HEK293 cells expressing CCR7 were treated with various concentrations of CCL19 and CCL21 ligands. At this time, the minimum concentration of CCL19 and CCL21 ligands was 0.1 nM in HEK293 cells, and 1 nM in MDA-MB-231 cells. The concentration of CCL19 and CCL21 at which maximum cAMP accumulation occurred was 10 nM in HEK293 cells and 50-100 nM in MDA-MB-231 cells.
도 5에 나타난 바와 같이, CCL19 또는 CCL21의 농도를 20 nM로 고정하고, 인간 6RG11 항체를 다양한 농도(1-200 nM)로 HEK293 CCR7+ 세포에 처리한 결과, 3-12 nM의 EC50으로의 유의적인 cAMP 축적 감소가 확인되었다.As shown in Figure 5, the concentration of CCL19 or CCL21 was fixed at 20 nM and the human 6RG11 antibody was treated with HEK293 CCR7+ cells at various concentrations (1-200 nM), resulting in a significant EC 50 of 3-12 nM. A significant decrease in cAMP accumulation was confirmed.
상술한 실험을 통해 본 발명에 따른 IgG4(6RG11)는 CCR7 신호전달 경로, 특히 Gi 의존성 cAMP 축적에 대한 우수한 길항 활성을 갖는다는 것을 알 수 있다.Through the above-described experiment, it can be seen that IgG 4 (6RG11) according to the present invention has excellent antagonistic activity against the CCR7 signaling pathway, especially Gi-dependent cAMP accumulation.
<실시예 11> 암 전이 및 침습에 대한 인간 항체 효능 분석<Example 11> Analysis of human antibody efficacy against cancer metastasis and invasion
CCR7(CC-chemokine receptor 7)은 화학주성 수용체로, 세포 이동을 유도하는 것으로 알려져 있다. 이에 앞서 실험을 통해 얻은, CCR7에 대한 인간 항체(IgG4(6RG11) 및 IgG4(72C7))가 CCL19 또는 CCL21에 의해 유도된 CCR7을 발현하는 MDA-MB-231 암세포의 이동 및 침입을 억제하는 효과를 나타내는지 확인하고자 하였다.CCR7 (CC-chemokine receptor 7) is a chemotactic receptor and is known to induce cell migration. Prior to this, human antibodies against CCR7 (IgG 4 (6RG11) and IgG 4 (72C7)) obtained through experiments were shown to inhibit the migration and invasion of MDA-MB-231 cancer cells expressing CCR7 induced by CCL19 or CCL21. We wanted to check whether it was effective.
1) Migration and invasion assay 1) Migration and invasion assay
MDA-MB-231 세포를 Lipofectamine® 3000(Thermo Fisher Scientific, USA)을 사용하여 pCDNA3.1-FLAG-CCR7 DNA 플라스미드(1 ug/μL)로 형질감염시키고, 20 시간 후에 배지를 교환한 후, 5% 이산화탄소, 37 ℃ 조건에서 밤새 배양하였다. 배지를 기아배지(starvation medium)(DMEM, 0.5% FBS, 1%(α) penicillin/streptomycin)로 교환한 후, 6시간동안 추가 배양하였다. Matrigel 매트릭스(growth factor reduction, Corning, USA)를 차가운 DMEM 배지를 사용해 200μg/ml 농도로 희석한 후, 24 well clear TC로 처리된 웰 플레이트(Costar, USA)의 투과성 지지체(8.0μm PET Membrane, Falcon, USA)를 상기 희석된 Matrigel 200μL로 각각 처리하였다. 상기 처리된 플레이트에서 Matrigel이 응고될때까지 2 시간이상 배양하였다. 상부 챔버(upper chamver)로부터 DMEM 배지를 제거하고, 굶주린(starved) 2 × 105 세포 200 μL만 처리하거나 혹은 인간 IgG4(6RG11, 72C7)과 혼합한 굶주린(starved) 2 × 105 세포 200 μL를 상부 챔버에 분주하였다. 하부 챔버(lower chamber)는 기아배지 800 μL만을 채우거나 혹은 30 nM의 CCL19 또는 CCL21를 포함하는 기아배지 800 μL를 채웠다. 상기 24-웰 플레이트를 40 시간동안 배양하고, 상부 챔버를 면봉(cotton swab)으로 닦은 후, 이동한 세포(migrated cells)를 Diff Quick 염색 키트를 사용하여 염색하고, 도립 현미경(inverted microscopy)으로 검출하였다. 이동된 세포(migrated cells)의 수는 Image J 소프트웨어로 카운팅하였고, GraphPad Prism 8 소프트웨어을 이용하여 분석하였다.MDA-MB-231 cells were transfected with pCDNA3.1-FLAG-CCR7 DNA plasmid (1 ug/μL) using Lipofectamine® 3000 (Thermo Fisher Scientific, USA), and the medium was changed after 20 h, followed by 5 % carbon dioxide, and cultured overnight at 37°C. The medium was exchanged for starvation medium (DMEM, 0.5% FBS, 1% (α) penicillin/streptomycin), and further cultured for 6 hours. Matrigel matrix (growth factor reduction, Corning, USA) was diluted to a concentration of 200 μg/ml using cold DMEM medium, and then placed on a permeable support (8.0 μm PET Membrane, Falcon) in a 24 well clear TC-treated well plate (Costar, USA). , USA) were each treated with 200 μL of the diluted Matrigel. Matrigel was cultured on the treated plate for more than 2 hours until it solidified. Remove DMEM medium from the upper chamber and treat with 200 μL of starved 2 × 10 5 cells alone or 200 μL of starved 2 × 10 5 cells mixed with human IgG 4 (6RG11, 72C7). was dispensed into the upper chamber. The lower chamber was filled with 800 μL of starvation medium alone or 800 μL of starvation medium containing 30 nM of CCL19 or CCL21. The 24-well plate was cultured for 40 hours, the upper chamber was wiped with a cotton swab, and the migrated cells were stained using the Diff Quick staining kit and detected with an inverted microscope. did. The number of migrated cells was counted using Image J software and analyzed using GraphPad Prism 8 software.
모든 샘플은 DMEM, 0.5% FBS에서 테스트되었으며 이동된 세포의 수는 Image J 소프트웨어로 계산하였으며, p 값이 0.05 이하인 경우를 유의한 것으로 판정하였다(P > 0.05는 의미 없음, * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001).All samples were tested in DMEM, 0.5% FBS, and the number of migrated cells was calculated using Image J software, and a p value of 0.05 or less was considered significant (P > 0.05 is not significant, * P ≤ 0.05, * *P ≤ 0.01, ***P ≤ 0.001).
도 6은 CCR7을 발현하는 MDA-MB-231 세포에서, CCL19 리간드에 의해 유도된 CCR7-의존적 침입(CCR7-dependent invasion)에 대한 IgG4(6RG11, 72C7)의 효과를 측정하여 나타낸 사진이고, 도 7은 CCR7을 발현하는 MDA-MB-231 세포에서, CCL21 리간드에 의해 유도된 CCR7-의존적 침입(CCR7-dependent invasion)에 대한 IgG4(6RG11, 72C7)의 효과를 측정하여 나타낸 사진이다.Figure 6 is a photograph showing the effect of IgG 4 (6RG11, 72C7) on CCR7-dependent invasion induced by CCL19 ligand in MDA-MB-231 cells expressing CCR7. 7 is a photograph showing the effect of IgG 4 (6RG11, 72C7) on CCR7-dependent invasion induced by CCL21 ligand in MDA-MB-231 cells expressing CCR7.
도 8은 도 6 및 7로부터 IgG4(6RG11, 72C7)에 따른 이동 억제를 시사하는 이동된 세포의 수를 측정하여 나타낸 그래프로, 도 8a는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL19 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이고, 도 8b는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL19 리간드에 대한 IgG4(72C7) 클론의 영향을 측정한 그래프이며, 도 8c는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL21 리간드에 대한 IgG4(6RG11) 클론의 영향을 측정한 그래프이며, 도 8d는 CCR7을 발현하는 MDA-MB-231 세포에서 CCL21 리간드에 대한 IgG4(72C7) 클론의 영향을 측정한 그래프이다.Figure 8 is a graph showing the number of migrated cells measured from Figures 6 and 7, suggesting migration inhibition by IgG4 (6RG11, 72C7). Figure 8a shows the number of migrated cells in MDA-MB-231 cells expressing CCR7 to CCL19 ligand. Figure 8b is a graph measuring the effect of the IgG4 (6RG11) clone on CCL19 ligand in MDA-MB-231 cells expressing CCR7, and Figure 8c is a graph measuring the effect of the IgG4 (72C7) clone on CCR7. This is a graph measuring the effect of the IgG4 (6RG11) clone on the CCL21 ligand in MDA-MB-231 cells expressing CCR7, and Figure 8d is a graph measuring the effect of the IgG4 (72C7) clone on the CCL21 ligand in MDA-MB-231 cells expressing CCR7. This is a graph measuring the impact.
도 6 및 7에 나타난 바와 같이, IgG4(6RG11) 또는 IgG4(72C7)는 모두 CCL19 또는 CCL21에 의해 유도되는 CCR7+ 세포의 이동을 방지한다는 것을 알 수 있다. 50 nM 미만의 저농도 조건에서는 IgG4(6RG11)가 IgG4(72C7)보다 더 우수한 효능을 갖는 것을 확인하였다.As shown in Figures 6 and 7, it can be seen that both IgG 4 (6RG11) or IgG 4 (72C7) prevent the migration of CCR7 + cells induced by CCL19 or CCL21. It was confirmed that IgG 4 (6RG11) had better efficacy than IgG 4 (72C7) under low concentration conditions of less than 50 nM.
도 8에 나타난 바와 같이, IgG4(6RG11)와 IgG4(72C7)은 CCR7 의존적인 침습에 대해 억제 활성을 나타낸다는 것을 확인하였다.As shown in Figure 8, it was confirmed that IgG 4 (6RG11) and IgG 4 (72C7) exhibited inhibitory activity against CCR7-dependent invasion.
종합하면, CCR7과 CCL19/CCL21 리간드의 결합은 CCR7의 구조적 변화, G 단백질의 변위, β-아레스틴 모집, 동종이량체 또는 이종이량체화와 같은 여러 분자 현상을 유도하는 것으로 확인되었다. CCR7과 CCL19/CCL21 리간드는 각각 G 단백질 활성화, β-아레스틴 모집, 내재화, 이동, 신호 전달 경로에 서로 다른 역할로 관여한다고 알려져 있다. 본 발명에서는 상술한 CCR7과 CCL19/CCL21 리간드 결합을 차단하는 활성을 갖는 인간 항체(IgG4)를 스크리닝하고자 하였고, 그 결과 두 종의 항체(IgG4(6RG11)와 IgG4(72C7))를 발견하기에 이르렀다. 본 발명에 따른 IgG4(6RG11)와 IgG4(72C7)는 모두 리간드 결합 영역에 가까운 CCR7의 세포외 영역에 결합하여 리간드 결합, 암세포의 전이 및 침습을 차단하고, β-arrestin 매개 ERK1/2 인산화를 차단하는 효과를 나타내고 있다. 상기 결과를 통해 본원발명의 항체는 암세포에서 CCR7의 신호전달과정을 효과적으로 억제하는 활성이 있어 치료제로서 효능이 있음을 확인하였다.Taken together, the binding of CCR7 to CCL19/CCL21 ligands was found to induce several molecular events, such as structural changes in CCR7, displacement of G proteins, β-arrestin recruitment, and homodimerization or heterodimerization. CCR7 and CCL19/CCL21 ligands are known to play different roles in G protein activation, β-arrestin recruitment, internalization, migration, and signaling pathways, respectively. In the present invention, we attempted to screen human antibodies (IgG 4 ) with the activity of blocking the binding of the above-described CCR7 and CCL19/CCL21 ligands, and as a result, two types of antibodies (IgG 4 (6RG11) and IgG 4 (72C7)) were discovered. It came down to this. Both IgG 4 (6RG11) and IgG 4 (72C7) according to the present invention bind to the extracellular region of CCR7 close to the ligand binding region, blocking ligand binding, metastasis and invasion of cancer cells, and β-arrestin-mediated ERK1/2 phosphorylation. It has a blocking effect. Through the above results, it was confirmed that the antibody of the present invention has the activity of effectively inhibiting the signaling process of CCR7 in cancer cells and is therefore effective as a therapeutic agent.
이상으로 본 발명의 특정한 부분을 상세히 기술하였는바, 당업계의 통상의 지식을 가진 자에게 있어서 이러한 구체적인 기술은 단지 바람직한 구현 예일 뿐이며, 이에 본 발명의 범위가 제한되는 것이 아닌 점은 명백하다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항과 그의 등가물에 의하여 정의된다고 할 것이다.As the specific parts of the present invention have been described in detail above, it is clear to those skilled in the art that these specific techniques are merely preferred implementation examples and do not limit the scope of the present invention. Accordingly, the substantial scope of the present invention will be defined by the appended claims and their equivalents.

Claims (17)

  1. CCR7(CC chemokine receptor 7)을 항원으로 인식하여 이에 특이적으로 결합하는, 중쇄 CDR1, CDR2, CDR3 및 경쇄 CDR1, CDR2, CDR3을 포함하는 항체 또는 이의 항원 결합 단편으로서,An antibody or antigen-binding fragment thereof comprising heavy chain CDR1, CDR2, and CDR3 and light chain CDR1, CDR2, and CDR3 that recognizes CCR7 (CC chemokine receptor 7) as an antigen and specifically binds to it,
    상기 항체 또는 이의 항원 결합 단편은 하기 군에서 선택되는 어느 하나의 CDR 조합을 포함하는 것을 특징으로 하는 항체 또는 이의 항원 결합 단편:The antibody or antigen-binding fragment thereof is characterized in that it comprises any one CDR combination selected from the following group:
    (A) 중쇄 가변영역에 서열번호 1의 CDR1, 서열번호 2의 CDR2 및 서열번호 3의 CDR3을 포함하고, 경쇄 가변영역에 서열번호 4의 CDR1, 서열번호 5의 CDR2 및 서열번호 6의 CDR3을 포함; (A) The heavy chain variable region includes CDR1 of SEQ ID NO: 1, CDR2 of SEQ ID NO: 2, and CDR3 of SEQ ID NO: 3, and the light chain variable region includes CDR1 of SEQ ID NO: 4, CDR2 of SEQ ID NO: 5, and CDR3 of SEQ ID NO: 6. include;
    (B) 중쇄 가변영역에 서열번호 1의 CDR1, 서열번호 2의 CDR2 및 서열번호 3의 CDR3을 포함하고, 경쇄 가변영역에 서열번호 10의 CDR1, 서열번호 11의 CDR2 및 서열번호 12의 CDR3을 포함;(B) The heavy chain variable region includes CDR1 of SEQ ID NO: 1, CDR2 of SEQ ID NO: 2, and CDR3 of SEQ ID NO: 3, and the light chain variable region includes CDR1 of SEQ ID NO: 10, CDR2 of SEQ ID NO: 11, and CDR3 of SEQ ID NO: 12. include;
    (C) 중쇄 가변영역에 서열번호 7의 CDR1, 서열번호 8의 CDR2 및 서열번호 9의 CDR3을 포함하고, 경쇄 가변영역에 서열번호 4의 CDR1, 서열번호 5의 CDR2 및 서열번호 6의 CDR3을 포함; 및(C) The heavy chain variable region includes CDR1 of SEQ ID NO: 7, CDR2 of SEQ ID NO: 8, and CDR3 of SEQ ID NO: 9, and the light chain variable region includes CDR1 of SEQ ID NO: 4, CDR2 of SEQ ID NO: 5, and CDR3 of SEQ ID NO: 6. include; and
    (D) 중쇄 가변영역에 서열번호 7의 CDR1, 서열번호 8의 CDR2 및 서열번호 9의 CDR3을 포함하고, 경쇄 가변영역에 서열번호 10의 CDR1, 서열번호 11의 CDR2 및 서열번호 12의 CDR3을 포함.(D) The heavy chain variable region includes CDR1 of SEQ ID NO: 7, CDR2 of SEQ ID NO: 8, and CDR3 of SEQ ID NO: 9, and the light chain variable region includes CDR1 of SEQ ID NO: 10, CDR2 of SEQ ID NO: 11, and CDR3 of SEQ ID NO: 12. include.
  2. 제 1 항에 있어서, 상기 항체 또는 이의 항원 결합 단편은 서열번호 13 또는 15의 중쇄 가변영역 및 서열번호 14 또는 16의 경쇄 가변영역을 포함하는 것을 특징으로 하는 항체 또는 이의 항원 결합 단편.The antibody or antigen-binding fragment thereof according to claim 1, wherein the antibody or antigen-binding fragment thereof comprises a heavy chain variable region of SEQ ID NO: 13 or 15 and a light chain variable region of SEQ ID NO: 14 or 16.
  3. 제 1 항에 있어서, 상기 항체 또는 이의 항원 결합 단편은 서열번호 13의 중쇄 가변영역 및 서열번호 14의 경쇄 가변영역; 또는 서열번호 15의 중쇄 가변영역 및 서열번호 16의 경쇄 가변영역;을 포함하는 것을 특징으로 하는 항체 또는 이의 항원 결합 단편.The method of claim 1, wherein the antibody or antigen-binding fragment thereof comprises a heavy chain variable region of SEQ ID NO: 13 and a light chain variable region of SEQ ID NO: 14; Or an antibody or antigen-binding fragment thereof comprising a heavy chain variable region of SEQ ID NO: 15 and a light chain variable region of SEQ ID NO: 16.
  4. 제 1 항에 있어서, 상기 항체 또는 이의 항원 결합 단편은 서열번호 17 또는 서열번호 19의 아미노산 서열을 포함하는 것을 특징으로 하는 항체 또는 이의 항원 결합 단편.The antibody or antigen-binding fragment thereof according to claim 1, wherein the antibody or antigen-binding fragment thereof comprises the amino acid sequence of SEQ ID NO: 17 or SEQ ID NO: 19.
  5. 제1항에 있어서, 상기 항체는 IgG, IgA, IgM, IgE 및 IgD로 이루어진 군에서 선택되며, 상기 항원 결합 단편은 디아바디, Fab, F(ab'), F(ab')2, Fv, dsFv 및 scFv로 이루어진 군에서 선택되는 것을 특징으로 하는 항체 또는 이의 항원 결합 단편.The method of claim 1, wherein the antibody is selected from the group consisting of IgG, IgA, IgM, IgE and IgD, and the antigen-binding fragment is diabody, Fab, F(ab'), F(ab')2, Fv, An antibody or antigen-binding fragment thereof, characterized in that it is selected from the group consisting of dsFv and scFv.
  6. 제1항에 있어서, 상기 항체 또는 이의 항원 결합 단편은 CCL19 및 CCL21로부터 선택되는 어느 하나의 CCR7 리간드에 의해 CCR7 의존성 세포 내 신호전달 및 CCR7 수용체 내재화 중의 적어도 하나를 억제하는 활성을 저해할 수 있는 것을 특징으로 하는 항체 또는 이의 항원 결합 단편.The method of claim 1, wherein the antibody or antigen-binding fragment thereof is capable of inhibiting the activity of inhibiting at least one of CCR7-dependent intracellular signaling and CCR7 receptor internalization by any one CCR7 ligand selected from CCL19 and CCL21. Characterized antibody or antigen-binding fragment thereof.
  7. 제1항에 있어서, 상기 항체 또는 이의 항원 결합 단편은 CCR7에 대해 20 nM 내지 100 nM의 해리상수(KD)를 갖는 것을 특징으로 하는 항체 또는 이의 항원 결합 단편.The antibody or antigen-binding fragment thereof according to claim 1, wherein the antibody or antigen-binding fragment thereof has a dissociation constant (K D ) of 20 nM to 100 nM for CCR7.
  8. 제 1 항의 항체 또는 이의 항원 결합 단편을 코딩하는 핵산분자.A nucleic acid molecule encoding the antibody of claim 1 or an antigen-binding fragment thereof.
  9. 제 8 항의 핵산분자를 포함하는 벡터.A vector containing the nucleic acid molecule of claim 8.
  10. 제 9 항의 벡터를 포함하는 숙주세포.A host cell containing the vector of claim 9.
  11. 제 1 항의 항체 또는 이의 항원 결합 단편, 제 8 항의 핵산분자 또는 제 9 항의 벡터 및 약제학적으로 허용되는 담체를 포함하는 약제학적 조성물.A pharmaceutical composition comprising the antibody or antigen-binding fragment thereof of claim 1, the nucleic acid molecule of claim 8 or the vector of claim 9, and a pharmaceutically acceptable carrier.
  12. 제 11 항에 있어서, 상기 약제학적 조성물은 암 또는 암의 전이의 예방 또는 치료용 약제학적 조성물인 것을 특징으로 하는 약제학적 조성물.The pharmaceutical composition according to claim 11, wherein the pharmaceutical composition is a pharmaceutical composition for preventing or treating cancer or cancer metastasis.
  13. 제 1 항의 항체 또는 이의 항원 결합 단편을 샘플에 처리하는 단계를 포함하는, 샘플 중에 포함된 CCR7의 정량 방법.A method for quantifying CCR7 contained in a sample, comprising treating the sample with the antibody or antigen-binding fragment thereof of claim 1.
  14. 하기의 단계를 포함하는 CCR7의 과발현에 의한 질환의 진단을 위한 정보를 제공하는 방법:A method of providing information for diagnosis of a disease caused by overexpression of CCR7 comprising the following steps:
    (a) 피검자로부터 체외로 분리된 샘플을 취득하는 단계;(a) obtaining a sample separated from the subject in vitro;
    (b) 제 1 항의 항체 또는 이의 항원 결합 단편을 상기 샘플에 처리하는 단계; 및(b) treating the sample with the antibody or antigen-binding fragment thereof of claim 1; and
    (c) 상기 피검자의 샘플 중에 포함된 CCR7의 발현양이 정상군 샘플 중에 포함된 CCR7의 발현양 보다 높은지 여부를 확인하는 단계.(c) Confirming whether the expression level of CCR7 included in the subject's sample is higher than the expression level of CCR7 included in the normal group sample.
  15. 제 14 항에 있어서, 상기 CCR7의 과발현에 의한 질환은 암 또는 전이성 암인 것을 특징으로 하는 방법.The method of claim 14, wherein the disease caused by overexpression of CCR7 is cancer or metastatic cancer.
  16. 제 1 항의 항체 또는 이의 항원 결합 단편을 포함하는 CCR7 정량 키트.A CCR7 quantitative kit comprising the antibody or antigen-binding fragment thereof of claim 1.
  17. 제 11 항의 약제학적 조성물을 이를 필요로 하는 대상(subject)에 투여하는 단계를 포함하는 암 또는 암의 전이의 예방 또는 치료방법.A method for preventing or treating cancer or cancer metastasis, comprising administering the pharmaceutical composition of claim 11 to a subject in need thereof.
PCT/KR2023/006705 2022-05-19 2023-05-17 Antibody for regulating ccr7 activity WO2023224390A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020220061516A KR20230161759A (en) 2022-05-19 2022-05-19 Antibodies regulating functions of a CC chemokine receptor 7
KR10-2022-0061516 2022-05-19

Publications (1)

Publication Number Publication Date
WO2023224390A1 true WO2023224390A1 (en) 2023-11-23

Family

ID=88835792

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/006705 WO2023224390A1 (en) 2022-05-19 2023-05-17 Antibody for regulating ccr7 activity

Country Status (2)

Country Link
KR (1) KR20230161759A (en)
WO (1) WO2023224390A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20140120831A (en) * 2013-04-02 2014-10-14 삼성전자주식회사 Anti-idiotype antibody against anti-c-Met antibody
JP2016519066A (en) * 2013-03-15 2016-06-30 アムジエン・インコーポレーテツド Methods and compositions related to anti-CCR7 antigen binding proteins
KR20180030931A (en) * 2015-08-10 2018-03-26 펩맵 비.브이. Humanized anti-CCR7 receptor antibody
JP2020506703A (en) * 2017-02-03 2020-03-05 ノバルティス アーゲー Anti-CCR7 antibody drug conjugate
KR20210132644A (en) * 2018-12-18 2021-11-04 캐터펄트 테라퓨틱스 비.브이. Use of an anti-CCR7 mAb for the prevention or treatment of graft versus host disease (GvHD)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102014383B1 (en) 2017-06-14 2019-08-26 국민대학교 산학협력단 Human antibodies regulating functions of a G-protein coupled receptor

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016519066A (en) * 2013-03-15 2016-06-30 アムジエン・インコーポレーテツド Methods and compositions related to anti-CCR7 antigen binding proteins
KR20140120831A (en) * 2013-04-02 2014-10-14 삼성전자주식회사 Anti-idiotype antibody against anti-c-Met antibody
KR20180030931A (en) * 2015-08-10 2018-03-26 펩맵 비.브이. Humanized anti-CCR7 receptor antibody
JP2020506703A (en) * 2017-02-03 2020-03-05 ノバルティス アーゲー Anti-CCR7 antibody drug conjugate
KR20210132644A (en) * 2018-12-18 2021-11-04 캐터펄트 테라퓨틱스 비.브이. Use of an anti-CCR7 mAb for the prevention or treatment of graft versus host disease (GvHD)

Also Published As

Publication number Publication date
KR20230161759A (en) 2023-11-28

Similar Documents

Publication Publication Date Title
WO2020111913A1 (en) Anti-4-1bb antibody and use thereof
WO2019098682A1 (en) Anti-her2 antibody or antigen-binding fragment thereof, and chimeric antigen receptor comprising same
AU2019229076A1 (en) Anti-TIGIT antibodies and uses thereof
WO2015005632A1 (en) Novel dual-targeted protein specifically binding to dll4 and vegf, and use thereof
WO2021006619A1 (en) Antibody binding specifically to b7-h3 and use thereof
WO2014189303A1 (en) Trans-tumoral peptide specific to neuropilin and fusion protein having same peptide fused therein
WO2016153276A1 (en) Neuropilin-1 specific binding peptide, fusion protein fused with same, and use thereof
WO2016013870A1 (en) Method for positioning, in cytoplasm, antibody having complete immunoglobulin form by penetrating antibody through cell membrane, and use for same
WO2019112347A2 (en) Antibody or antigen binding fragment thereof for specifically recognizing b cell malignancy, chimeric antigen receptor comprising same, and use thereof
WO2020005003A1 (en) Monoclonal antibody specifically binding to lag-3 and use thereof
WO2021133036A1 (en) Anti-lilrb1 antibody and uses thereof
WO2017030370A1 (en) Chimeric antibody receptor to which anti-cotinine antibody is linked, and use thereof
WO2018222019A1 (en) Novel anti-cd40 antibodies and use thereof
WO2019132533A1 (en) Anti-pd-l1 antibody and use thereof
CN109415443B (en) HER-2 binding antibodies
WO2022216014A1 (en) Anti-cntn4 antibody and use thereof
WO2020116963A1 (en) Endothelin receptor type a activity regulating antibody
WO2020145669A1 (en) Anti-beta 1 integrin humanized antibody, and pharmaceutical composition for treating cancer, comprising same
WO2023224390A1 (en) Antibody for regulating ccr7 activity
WO2016084993A1 (en) Novel egfrviii antibody and composition comprising same
WO2022025585A1 (en) Anti-lilrb1 antibody and uses thereof
WO2020242200A1 (en) Antibody with enhanced binding affinity for endothelin receptor a
WO2022231032A1 (en) Anti-cntn4-specific antibodies and use thereof
WO2020071881A1 (en) Pdgf receptor antibody and use thereof
WO2023043123A1 (en) Glycosylated fc variants with improved selective binding affinity to fcγriiia

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: 23807909

Country of ref document: EP

Kind code of ref document: A1