WO2014069681A1 - Cancer-targeting antibody and composition for preventing or treating cancer comprising same - Google Patents

Cancer-targeting antibody and composition for preventing or treating cancer comprising same Download PDF

Info

Publication number
WO2014069681A1
WO2014069681A1 PCT/KR2012/008951 KR2012008951W WO2014069681A1 WO 2014069681 A1 WO2014069681 A1 WO 2014069681A1 KR 2012008951 W KR2012008951 W KR 2012008951W WO 2014069681 A1 WO2014069681 A1 WO 2014069681A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
cancer
amino acid
acid sequence
Prior art date
Application number
PCT/KR2012/008951
Other languages
French (fr)
Korean (ko)
Inventor
허대영
박가빈
김영석
송현근
김성한
이현경
박세광
양재욱
정하나
박솔지
Original Assignee
인제대학교 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 인제대학교 산학협력단 filed Critical 인제대학교 산학협력단
Priority to PCT/KR2012/008951 priority Critical patent/WO2014069681A1/en
Publication of WO2014069681A1 publication Critical patent/WO2014069681A1/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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Definitions

  • the present invention relates to monoclonal antibodies that bind to cancer cells and induce apoptosis and their medical use.
  • Cancer is one of the most deadly threats to human health. In the United States, nearly 1,300,000 new patients get cancer each year, causing about one in four deaths following heart disease. It is also predicted that cancer can surpass cardiovascular disease as the first cause of death within five years. Solid cancer accounts for most of these deaths. Although significant progress has been made in the medical treatment of certain cancers, the overall 5-year survival rate for all cancers has improved only about 10% over the past 20 years. Cancer or malignant tumors metastasize and grow rapidly in an uncontrolled manner, making it very difficult to detect and treat in a timely manner. In addition, cancer can occur from almost any tissue in the body through one or several normal cells in the tissue through malignant transformation, and differ from one type of cancer with a particular tissue origin.
  • Tumor cells differ in antigenicity from normal cells due to the presence of "tumor antigens". They may be specific to tumor cells, or are recognized as “tumor-associated antigens” (TAAs) by being expressed differently or in excess. Tumor cells not only differ in protein primary structure, i.e. from amino acid sequences, from normal cell changes, but also in secondary and tertiary due to changes in post-translational modifications, such as changes in glycosylation and phosphorylation. The structure may also differ from normal cells, which continually change the antigenicity of the protein and also specify it as a tumor-associated antigen.
  • TAAs tumor-associated antigens
  • protein mucin from the mammary gland which itself is not a change present in tumor cells, but autoantibodies to it are found in breast cancer patients (sometimes in ovarian cancer). The reason is presumably because the protein is highly glycosylated in normal cells and is not exposed at all due to the dense and thick coating of carbohydrate chains. Poor glycosylation in tumor cells results in exposure of protein fragments that act as antigenic determinants to the immune system.
  • TAA is currently thought to be a molecule that can be associated with certain tumors, such as lymphomas, carcinomas, sarcomas, or melanoma, and can elicit cellular and / or humoral immune responses against tumors and, rarely, against tumors. It also defends the host.
  • tumors such as lymphomas, carcinomas, sarcomas, or melanoma
  • Such TAAs are currently classified into three classes, which are highly specific for certain tumors that are present in only one or only a few individuals and are not found in normal cells, for example tumor-specific transplant antigens (class 1). ); Present in a large number of related tumors of different patients (second class); And present in normal cells and malignant cells and expressed in large quantities in malignant cells (third class). Since the second class of TAA is present in many tumors and rarely observed in normal subjects, it is considered to have the greatest potential for clinically useful analyses.
  • Korean Laid-Open Patent Publication 10-2010-0045903 discloses an antibody targeting anti DR5, which is an apoptosis receptor for cancer cells.
  • the present invention is to provide a novel antibody targeting a tumor cell specific antigen and a method for treating cancer inducing apoptosis of cancer cells through the antibody.
  • the present invention binds to cancer cells as a monoclonal antibody inducing apoptosis
  • a light chain variable region comprising a light chain CDR1 consisting of the amino acid sequence of SEQ ID NO: 1, a light chain CDR2 consisting of the amino acid sequence of SEQ ID NO: 2, and a light chain CDR3 consisting of the amino acid sequence of SEQ ID NO: 3;
  • an antibody comprising a heavy chain variable region comprising a heavy chain CDR1 consisting of the amino acid sequence of SEQ ID NO: 4, a heavy chain CDR2 consisting of the amino acid sequence of SEQ ID NO: 5, and a heavy chain CDR3 consisting of the amino acid sequence of SEQ ID NO: 6.
  • the antibody is a single-chain variable fragment (scFv, single-chain variable fragment) antibody, and may have an amino acid sequence of SEQ ID NO: 9, SEQ ID NO: 13, SEQ ID NO: 17 or SEQ ID NO: 21. .
  • the present invention also provides a gene encoding the single chain variable region antibody.
  • the present invention also provides a pharmaceutical composition for preventing or treating cancer containing the antibody as an active ingredient.
  • the present invention also provides a cancer cell targeting diagnostic or therapeutic conjugate comprising the antibody, wherein one or more diagnostic or anticancer agents are bound to the antibody.
  • the antibody of the present invention has an activity of specifically binding to a cancer cell line, and can be used as a cancer cell targeting diagnosis or therapeutic conjugate to effectively diagnose or treat cancer.
  • the antibody of the present invention shows a property of inducing apoptosis by binding to cancer cells, particularly lymphoma cells and ovarian cancer cells, through which the antibody of the present invention can be used as an anticancer agent.
  • Example 1 is a flow cytometry result for the IM9 cell line of Example 1.
  • Figure 2 shows the nucleotide and amino acid sequences of the scFv clone ITTab1-1 and CDRs and linker regions thereof according to the present invention.
  • Figure 3 shows the nucleotide and amino acid sequences of the scFv clone ITTab1-2 and its CDR and linker regions according to the present invention.
  • Figure 4 shows the nucleotide and amino acid sequence of the scFv clone ITTab1-3 and its CDR and linker regions according to the present invention.
  • Figure 5 shows the nucleotide and amino acid sequences of the scFv clone ITTab1-4 and its CDR and linker regions according to the present invention.
  • FIG. 6 is a photograph of confocal microscopy results of examining the binding of the antibody of the present invention to NK lymphoma cells.
  • Figure 7 is a flow cytometry test results of examining apoptosis of NK lymphoma cells by the antibody treatment of the present invention.
  • FIG. 9 is a flow cytometry test result of examining apoptosis of ovarian cancer cells by the antibody treatment of the present invention.
  • the present invention is a monoclonal antibody that binds to cancer cells and induces apoptosis
  • a light chain variable region comprising a light chain CDR1 consisting of the amino acid sequence of SEQ ID NO: 1, a light chain CDR2 consisting of the amino acid sequence of SEQ ID NO: 2, and a light chain CDR3 consisting of the amino acid sequence of SEQ ID NO: 3;
  • an antibody comprising a heavy chain variable region comprising a heavy chain CDR1 consisting of the amino acid sequence of SEQ ID NO: 4, a heavy chain CDR2 consisting of the amino acid sequence of SEQ ID NO: 5, and a heavy chain CDR3 consisting of the amino acid sequence of SEQ ID NO: 6.
  • the inventors of the present invention while studying a novel antibody targeting a tumor cell specific antigen, constructed a hybridoma cell that specifically binds to B cell tumor cells (IM9) using PBMC stimulated with ConA, The monoclonal antibody prepared by this specifically binds to cancer cells, in particular NK lymphoma cells and ovarian cancer cells, and confirmed that it also induces apoptosis, thereby completing the present invention.
  • IM9 B cell tumor cells
  • the light chain variable region may consist of the amino acid sequence of SEQ ID NO: 7, SEQ ID NO: 11, SEQ ID NO: 15 or SEQ ID NO: 19.
  • the heavy chain variable region may be composed of the amino acid sequence of SEQ ID NO: 8, SEQ ID NO: 12, SEQ ID NO: 16 or SEQ ID NO: 20.
  • an "antibody” may be an entire form of an antibody ("antibody") or a functional fragment thereof.
  • the whole antibody may be in the form of a monomer or a multimer in which two or more whole antibodies are bound.
  • the functional fragment of the antibody is an antibody having the heavy and light chain variable regions of the whole antibody, which means to recognize substantially the same epitope that the whole antibody recognizes.
  • Functional fragments of the antibody include, but are not limited to, single chain variable region fragments (scFv), (scFv) 2 , Fab, Fab 'and F (ab') 2, and the like. Antibodies in the form of single-chain variable fragments (scFv) are preferred.
  • the single chain variable region refers to an antibody fragment in which a heavy chain variable region and a light chain variable region are linked through a linker peptide to take the form of a single chain polypeptide.
  • the antibody can be generated using methods known in the art, such as phage display methods or yeast cell surface expression systems.
  • the antibody of the invention is a single chain variable region antibody, which may have an amino acid sequence of SEQ ID NO: 9, SEQ ID NO: 13, SEQ ID NO: 17 or SEQ ID NO: 21.
  • the antibodies of the present invention may be derived from any animal, including mammals, birds, and the like, including humans.
  • the antibody may be a human, mouse, donkey, sheep, rabbit, goat, guinea pig, camel, horse or chicken antibody.
  • the human antibody is an antibody having an amino acid sequence of human immunoglobulin, and includes an antibody isolated from a human immunoglobulin library or an antibody isolated from an animal transfected against one or more human immunoglobulins and not expressing an endogenous immunoglobulin.
  • the present invention also provides a gene encoding each of the single-chain variable region antibodies having the amino acid sequence of SEQ ID NO: 9, SEQ ID NO: 13, SEQ ID NO: 17 or SEQ ID NO: 21.
  • Gene sequences encoding the antibodies of the invention can be obtained by methods well known in the art. For example, based on DNA sequences or corresponding amino acid sequences encoding portions or all of the heavy and light chains of the antibody, oligonucleotide synthesis techniques well known in the art, for example site-directed mutagenesis And the polymerase chain reaction (PCR) method.
  • PCR polymerase chain reaction
  • the gene encoding the single-chain variable region antibody having the amino acid sequence of SEQ ID NO: 9 may be composed of the nucleotide sequence of SEQ ID NO: 10.
  • the gene encoding the single-chain variable region antibody having the amino acid sequence of SEQ ID NO: 13 may be composed of the nucleotide sequence of SEQ ID NO: 14.
  • the gene encoding the single-chain variable region antibody having the amino acid sequence of SEQ ID NO: 17 may be composed of the nucleotide sequence of SEQ ID NO: 18.
  • the gene encoding the single-chain variable region antibody having the amino acid sequence of SEQ ID NO: 21 may be composed of the nucleotide sequence of SEQ ID NO: 22.
  • the host cell is transformed with the gene and the expression vector including the same, and cultured under appropriate conditions to express and isolate the antibody, thereby producing an antibody molecule.
  • the antibody of the present invention exhibits a property of specifically binding to cancer cells, and in particular, binds to lymphoma cells and ovarian cancer cells and induces cell death, and thus can be used for the prevention or treatment of cancer.
  • the present invention provides a pharmaceutical composition for preventing or treating cancer containing the antibody as an active ingredient, the use of the antibody for the manufacture of an anticancer agent, and administering to the subject a therapeutically effective amount of the antibody to a subject. To provide.
  • the pharmaceutical composition for preventing or treating cancer containing the antibody as an active ingredient is a suitable carrier, excipient, disintegrant, sweetener, coating agent, swelling agent, It may further comprise one or more additives selected from the group consisting of lubricants, lubricants, flavors, antioxidants, buffers, bacteriostatics, diluents, dispersants, surfactants, binders and lubricants.
  • the carriers, excipients and diluents are lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline Cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil can be used, and solid preparations for oral administration include tablets, pills, powders, granules, capsules.
  • solid preparations may be prepared by mixing at least one excipient such as starch, calcium carbonate, sucrose or lactose, gelatin and the like in the composition.
  • excipients such as starch, calcium carbonate, sucrose or lactose, gelatin and the like
  • lubricants such as magnesium styrate and talc may also be used.
  • Oral liquid preparations include suspensions, solvents, emulsions, syrups, and the like, and may include various excipients such as wetting agents, sweeteners, fragrances, and preservatives, in addition to commonly used simple diluents such as water and liquid paraffin.
  • Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized preparations, suppositories, and the like.
  • non-aqueous solvent and suspending agent propylene glycol, polyethylene glycol, vegetable oil such as olive oil, injectable ester such as ethyl oleate and the like can be used.
  • Witsol, macrogol, tween 61, cacao butter, laurin butter, glycerogelatin and the like may be used as the base material of the suppository.
  • the pharmaceutical composition for preventing or treating cancer containing the antibody as an active ingredient is a granule, powder, coated tablet, tablet, pill, capsule, suppository, gel, syrup, juice It can be formulated as a suspension, emulsion, drop or liquid.
  • the pharmaceutical composition is intravenous, intraarterial, intraperitoneal, intramuscular, intraarterial, intraperitoneal, intrasternal, transdermal, nasal, inhaled, topical, rectal, oral, intraocular or intradermal Via the route can be administered to the subject in a conventional manner.
  • the preferred dosage of the antibody may vary depending on the condition and weight of the subject, the type and extent of the disease, the form of the drug, the route of administration, and the duration and may be appropriately selected by those skilled in the art. According to one embodiment of the present invention, but not limited thereto, the daily dosage may be 0.01 to 200 mg / kg, specifically 0.1 to 200 mg / kg, more specifically 0.1 to 100 mg / kg. Administration may be administered once a day or divided into several times, thereby not limiting the scope of the invention.
  • the 'subject' may be a mammal including a human, but is not limited thereto.
  • the cancer may be any one selected from liver cancer, lung cancer, colon cancer, ovarian cancer, breast cancer, prostate cancer, leukemia, lymphoma and pancreatic cancer.
  • the present invention also provides a cancer cell targeting diagnostic or therapeutic conjugate comprising the antibody and binding one or more diagnostic or anticancer agents to the antibody.
  • the diagnostic agent may be any one or two or more selected from the group consisting of radionuclides, paramagnetic metal ions, superparamagnetic nanoparticles, chromophores, chromophores, luminescent materials, and fluorescent materials
  • the contrast agent may be CT contrast agent, MRI. It may be any one selected from the group consisting of a contrast agent, an optical contrast agent, and an ultrasound contrast agent.
  • the anticancer agent may be used without limitation as long as it is conventionally used for the treatment of cancer, for example cisplatin, 5-fluorouracil, adriamycin, methotrexate, vinblastine, busulfan, chlorambucil, cyclophosphamide, Melphalan, nitrogen mustard, nitrosourea, taxol, paclitaxel, docetaxel, 6-mercaptopurine, 6-thioguanine, bleomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin-C And hydroxyurea can be used any one or two or more selected from the group consisting of.
  • the linkage between the anticancer agent and the antibody of the present invention can be carried out through methods known in the art, such as covalent bonds, crosslinking and the like.
  • the anticancer agent may be a therapeutic radionuclide, for example, 32 P, 47 Sc, 64 Ga, 64 Cu, 66 Cu, 67 Cu, 89 Sr, 90 Y, 91 Y, 103 Pd, 103 m Rh, 105 Rh, 111 Ag, 111 In, 117m Sn, 125 I, 131 I, 145 Sm, 149 Pm, 153 Sm, 165 Dy, 166 Ho, 169 Er, 176 Lu, 177 Lu, 182 Ta, 186 Re, 188 Re, 188 W , 192 Ir, 195 m Pt, 198 Au, 199 Au, 211 At, 212 Bi, 213 Bi, 223 Ra, 225 Ac, 230 U, 252 Cf, or a combination thereof.
  • a therapeutic radionuclide for example, 32 P, 47 Sc, 64 Ga, 64 Cu, 66 Cu, 67 Cu, 89 Sr, 90 Y, 91 Y, 103 Pd, 103 m Rh
  • the amount of the antibody of the present invention included in the cancer cell targeting therapeutic conjugate of the present invention may vary depending on the type and amount of the anticancer agent to be bound.
  • the amount of the anticancer agent administered for treatment may be an amount sufficient to deliver the target cancer disease site.
  • the drug is determined not only by the route of administration and the number of treatments, but also by various factors such as the age, weight, health condition, sex, severity of the disease, diet, and excretion rate, the effective dose to the patient is determined. In view of this, one of ordinary skill in the art will be able to determine an appropriate effective amount depending on the use of the antibody for treating cancer diseases by combining the antibody with an anticancer agent.
  • the cancer cell targeting therapeutic conjugate of the present invention may include a suitable buffer solution for maintaining / preserving the stability of the antibody.
  • the cancer cell targeting therapeutic conjugate of the present invention is not particularly limited to its formulation, route of administration, and method of administration so long as the desired effect is achieved in the present invention.
  • the conjugates of the present invention can be administered parenterally, such as subcutaneous injection, intramuscular injection, intravenous injection, urethral injection, intrabronchial inhalation, intrauterine dural or cerebrovascular injection, and the like.
  • cancer cell targeting therapeutic conjugates of the present invention include appropriate carriers, excipients, disintegrants, sweeteners, coating agents, swelling agents, lubricants, lubricants, flavoring agents, antioxidants, buffers, bactericides, diluents, It may further comprise one or more additives selected from the group consisting of dispersants, surfactants, binders and lubricants.
  • cancer cell targeting therapeutic conjugate of the present invention may be formulated into granules, powders, coated tablets, tablets, pills, capsules, suppositories, gels, syrups, juices, suspensions, emulsions, drops or solutions according to conventional methods.
  • injectables can be prepared in the form of unit dose ampoules or multiple dose inclusions.
  • mice were 5-6 week old female Balb / c mice purchased from Orient Bio. The purchased mouse was purified for 1 week by supplying sufficient solid feed and water in an indoor environment with a temperature of 20-25 ° C., a humidity of 60%, and an illuminance of 200-300 Lux .
  • PBMC 1X10 7 isolated from Ficoll-gradient method from healthy volunteers was stimulated with Con A (concanavalin A) 10 ug / ml and immunized with mouse. After 2 months, spleens of mice were isolated and 1X10 8 splenocytes were isolated. This was obtained and fused with 10 7 SP 2 / 0-Ag14 mouse myeloma cells using polyethylene glycol (PEG 4000; Sigma, St Louis, MO) method. The method is described in detail in Kohler G, Milstein C. Nature 256 (1975) 495-497. FACS staining was performed using the obtained culture supernatant of 100 hybridoma cell lines to obtain hybridoma cells reactive to B cell tumor cells (IM9, Raju, Ramos).
  • Con A canavalin A
  • ITT-ab1 Inje Tumor Targeting-antibody1 mouse antibody of the present invention
  • the library (library) production step (A) It was prepared, characterized in that it comprises an antibody function confirmation step (B).
  • scFv single-chain Fv
  • hybridoma F6 cells hybridoma cells obtained in the method 1 of production of antibodies that bind to B cell tumor cells (IM9).
  • IM9 B cell tumor cells
  • Separation and purification step (a1) for purifying RNA A polymerase chain reaction amplification step (a2) for amplifying the first strand cDNA by a polymerase chain reaction of a mouse antibody gene; A cell fabrication step (a3) of fabricating a competent cell; It is composed of a library completion step (a4) for completing a single chain Fv (scFv) library based on the production step.
  • Separation and purification step (a1) is to purify RNA, cultivate hybridoma F6 cells, add TRIZOL 2ml to F6 cell 1x10 7 using TRIZOL reagent for 5 minutes at room temperature, and then 200ul of chloroform per 1ml TRIZOL. I vortex for about 15 seconds. And centrifuged for 15 minutes at 13000rpm at 4 °C using a centrifuge. Then, the supernatant was carefully transferred to a microcentrifuge tube so as not to pick the middle layer, and isopropyl alcohol was mixed in the same ratio and reacted at room temperature for 10 minutes.
  • Polymerase chain reaction amplification step (a2) is a step of preparing the first strand cDNA and amplifying the polymerase chain reaction of the mouse antibody gene, and the purified RNA 2 ug in the purification step (a1) and 0.5 ug oligo (dT) and After mixing and adding distilled water to a final volume of 10 ul and reacted for 5 minutes at 75 °C. Thereafter, 15 ul of cDNA reaction solution (5 ul of 5 x M-MLV reaction solution, 100 units of M-MLV reverse transcriptase, 10 mM dNTP 1.25 ul, and RNase Inhibitor 4 units) were added and vortex mixed with an RNA sample. The reaction was carried out in a chain polymerization apparatus at 37 ° C. for 1 hour and at 95 ° C. for 5 minutes to amplify the polymerase chain reaction with an mGAPDH primer.
  • the antibody gene was amplified by polymerase chain reaction with a mouse antibody specific primer. Amplify the variable regions of the light and heavy chains, bind them with overlapping polymerase chain reaction using linker-specific primers, and clone them into the Sfi I restriction site of the pComb3X-TT expression vector. was produced.
  • the primer used at this time was determined by a manual (Barbas et al., Phage Display; A Laboratory Manual, Cold Spring Harbor Laboratory, (2001), New York), and was synthesized by making a request to Bioneer (Korea).
  • Polymerase chain reaction conditions were pre-denaturated at 94 ° C. for 5 minutes, followed by 25 cycles of reaction at 94 ° C. 30 sec, 57 ° C. 15 sec, and 72 ° C. 30 sec. The presence or absence of amplification was confirmed by 1% agarose gel electrophoresis, purified using Qiagen gel extraction kit, and the amplified light chain variable region and heavy chain variable region were subjected to the following overlapping polymerase chain reaction.
  • the polymerase chain reaction product was purified by DNA from agarose with Qiagen gel extraction kit, restriction enzyme digestion with Sfi I, and then gel extraction.
  • Sfi I was added to a mixture of the cut antibody gene 30 ng and Sfi I cleavage pComb3X-TT 30 ng after 10X buffer and a ligase (ligase, Gibco-BRL, USA ) 10 unit and the reaction at room temperature for 2 hours after TOP10F Transformed competent cells.
  • Cell production step (a3) is a step of producing a competent cell (Electrocompetent cell), incubation of E. coli ER2537 in a minimal plate (minimal plate) to induce the expression of the F 'cilia after the production of water-soluble cells (competent cell) Used.
  • ER2537 was inoculated in 2 ml of SB medium (10 g MOPS, 30 g, bacto-trypton, 20 g yeast extract in 1 L) and incubated at 37 ° C. for 18 hours, followed by OD 600 to 3.54.
  • 1 ml of the pre-culture was incubated in 100 ml of LB medium (10 g NaCl, 10 g, bacto-trypton, 5 g yeast extract in 1 L) so that the OD 600 was 0.4. Thereafter, centrifuge was performed for 10 minutes at 4,000 rpm at 4 °C. The cells were suspended with 20 ml of 50 mM CaCl 2 and then reacted for 30 minutes on ice, and then centrifuged at 4,000 rpm for 10 minutes at 4 ° C. The supernatant was removed and 3 ml of (10% glycerol + 50 mM CaCl 2 ) was added to the cells, followed by aliquoting of 50ul.
  • LB medium 10 g NaCl, 10 g, bacto-trypton, 5 g yeast extract in 1 L
  • the library completion step (a4) is a step of completing the scFv (single chain Fv) library based on the preparation step, and 100 ⁇ g / mL of ampicillin after infecting the recovered phage antibody with Escherichia coli ER2537. Plates were added to the added LB agar plates and incubated at 37 ° C for 18 hours. The colonies formed were inoculated into 5 mL of SB (SB / amp) liquid medium to which 100 ⁇ g / mL of ampicillin was added and shaken at 37 ° C. at 250 rpm for 4 hours.
  • SB SB / amp
  • phage formation 10 10 pfu of VCSM13 was added and allowed to stand at 37 ° C for 30 minutes for infection, followed by incubation at 250 rpm at 37 ° C for 1 hour 30 minutes, and kanamycin was added to 70 ⁇ g / mL for 18 hours at 37 ° C. Incubated. Thereafter, the culture solution was centrifuged at 2,500 rpm for 10 minutes to recover the supernatant containing phage.
  • the antibody function checking step (B) confirms the function of the antibodies completed in the library completion step (a4), and treats phage antibodies that recognize IM-9 cells with ⁇ -M13 and ⁇ -mIgG-PE. After FACS staining was confirmed whether the binding to IM9 B cell tumor cells.
  • ITTab1 obtained by phage display has the same characteristics as the antibody derived from the secured hybridoma
  • FACS staining was confirmed for binding to IM9 cell line, B cell tumor cells.
  • IM-9 cells (10 6 ) were treated with 1 ug of ITTab1 subclonal antibodies (ITTab 1-1 to ITTab 1-4), respectively, and washed with PBS after 30 min binding.
  • the washed cells were combined with FITC-conjugated goat anti-mouse IgG, (Sigma-Aldrich) for 30 minutes, washed with PBS three times, and observed with FACS Calibur (BD Pharmingen). The results are shown in FIG.
  • CDR Complimentarily-determining region
  • scFv short chain Fv
  • the underlined portion is a linker region, which is divided into a light chain and a heavy chain, and the heavy chain. From the sites marked in boldface at the top of the chain, CDR1, CDR2 and CDR3 of the heavy chain were determined. The sites shown in bold at the front of the light chain bordering the linker were determined by CDR1, CDR2 and CDR3 of the light chain.
  • Figure 2 shows the base and amino acid sequence of ITTab1-1 and its CDR and linker regions.
  • ITTab1-1 is a light chain CDR1 (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2), CDR3 (SEQ ID NO: 3), heavy chain CDR1 (SEQ ID NO: 4), CDR2 (SEQ ID NO: 1) No. 5), CDR3 (SEQ ID NO: 6).
  • the entire amino acid sequence of ITTab1-1 is shown in SEQ ID NO: 9, and the entire nucleotide sequence is shown in SEQ ID NO: 10.
  • FIG 3 shows the base and amino acid sequence of ITTab1-2 and its CDR and linker regions.
  • ITTab1-2 is a light chain CDR1 (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2), CDR3 (SEQ ID NO: 3), heavy chain CDR1 (SEQ ID NO: 4), CDR2 (SEQ ID NO: 1) No. 5), CDR3 (SEQ ID NO: 6).
  • the entire amino acid sequence of ITTab1-2 is shown in SEQ ID NO: 13, and the entire nucleotide sequence is shown in SEQ ID NO: 14.
  • ITTab1-3 is a light chain CDR1 (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2), CDR3 (SEQ ID NO: 3), heavy chain CDR1 (SEQ ID NO: 4), CDR2 (SEQ ID NO: 1) No. 5), CDR3 (SEQ ID NO: 6).
  • the entire amino acid sequence of ITTab1-3 is shown in SEQ ID NO: 17, and the entire nucleotide sequence is shown in SEQ ID NO: 18.
  • FIG. 5 shows the base and amino acid sequences of ITTab1-4 and their CDRs and linker regions.
  • ITTab1-4 is in the order of bold font CDR1 (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2), CDR3 (SEQ ID NO: 3), CDR1 (SEQ ID NO: 4), CDR2 (SEQ ID NO: 4) of the heavy chain No. 5), CDR3 (SEQ ID NO: 6).
  • the entire amino acid sequence of ITTab1-4 is shown in SEQ ID NO: 21, and the entire nucleotide sequence is shown in SEQ ID NO: 22.
  • the presence of the antigen of ITTab1 on NK lymphoma cells was analyzed by immunofluorescence and confocal microscopy.
  • NK lymphoma cells were incubated with 1 ug of ITTab1 for 30 minutes, the cells were recovered and washed with PBS.
  • the washed cell lines were incubated with FITC-conjugated goat anti-mouse IgG (Sigma-Aldrich) for 30 minutes and washed three times with PBS.
  • the washed cells were placed on a microscope slide under the cover-slip using fluorescent mounting medium (DakoCytomation, Glostrup, Denmark).
  • Fluorescent stained cells were observed with 400 ⁇ original magnification using a Confocal Laser-Scanning microscope (Carl ZEISS, 510 META, Jena, Germany) and analyzed by confocal Microscopy Software Release 3.0 (Carl ZEISS, 510 META).
  • FIG. 6 it can be seen from the confocal microscopy results that the cell surface and the cytoplasm are stained with green fluorescence by ITTab1. Through this, the target antigen for the ITTab1 antibody is present in NK lymphoma cells. there was.
  • ITTab1 antigen expressed on NK lymphoma cells on NK lymphoma cells To clarify the function of ITTab1 antigen expressed on NK lymphoma cells on NK lymphoma cells, the effect of ITTab1 on tumor cells after treatment with tumor cells was analyzed by an annexin-V assay.
  • NK lymphoma cells (10 6 cells) were incubated with each concentration of ITTab1 for 1 hour, and then the cells were recovered and washed twice with PBS. The washed cell lines were suspended with 100 ul of Annexin-V binding buffer (10 mM EPES / NaOH pH 7.4, 140 mM NaCl, 2.5 mM CaCl 2 ), followed by 2ul of FITC-conjugate Annexin-V (BD Pharmingen) and 7- ADD was processed. Treated NK lymphoma cells were incubated for 15 minutes at room temperature in the dark, followed by the addition of 400 ul of Annexin-V binding buffer and analyzed using FACS Calibur (BD Pharmingen).
  • Ovarian cancer cells were incubated with 1 ug of ITTab1 for 30 minutes and then cells were recovered and washed with PBS. The washed cell lines were incubated with FITC-conjugated goat anti-mouse IgG (Sigma-Aldrich) for 30 minutes and washed three times with PBS. Washed cells were analyzed using FACS calibur (BD Pharmingen).
  • ITTab1 antigen expressed in ovarian cancer cells on ovarian cancer cells was analyzed by cell cycle analysis by propidium iodide (PI) treatment.
  • Ovarian cancer cells (10 6 cells) were incubated with 5ug of ITTab1 for 1 hour and then recovered and washed twice with PBS. The washed cell lines were treated with propidium iodine (PI) and the cells were analyzed using FACS caliber (BD Pharmingen).
  • PI propidium iodine
  • ITTab1 of the present invention can be used as an apoptosis-inducing antibody in ovarian cancer cells.

Abstract

The present invention relates to a monoclonal antibody for inducing apoptosis by binding to a cancer cell and a medical use thereof, and the antibody according to the present invention exhibits activity for cancer cell line-specific binding and thus can be used in cancer cell-targeted diagnosis or as a drug conjugate to effectively diagnose or treat cancer. Also, the antibody according to the present invention exhibits the characteristic of inducing apoptosis by binding to cancer cells, particularly lymphoma cells and ovarian cancer cells, and thus the antibody according to the present invention can be used alone as an anticancer drug.

Description

암 표적 항체 및 이를 포함하는 암 예방 또는 치료용 조성물Cancer target antibody and composition for preventing or treating cancer comprising the same
본 발명은 암세포에 결합하여 세포사멸을 유도하는 단일클론 항체 및 이의 의학적 용도에 관한 것이다. The present invention relates to monoclonal antibodies that bind to cancer cells and induce apoptosis and their medical use.
암은 인간의 건강에 대한 가장 치명적인 위협 중의 하나이다. 미국에서는 매년 거의 1,300,000명의 새로운 환자가 암에 걸리며 심장병에 이어 제 2의 사망 원인으로 4명 중 약 1명에 이른다. 또한, 암은 5년 이내에 사망의 제 1 원인이 되어 심혈관 질병을 능가할 수 있다고 예측된다. 고형암이 이러한 사망의 대부분을 차지한다. 특정 암의 의학적 치료에 있어서는 상당한 진전이 있었지만, 모든 암에 대한 총 5-년 생존률은 지난 20년 동안 단지 약 10% 만이 개선되었다. 암 또는 악성 종양은 전이되며 조절되지 않는 방식으로 급속히 성장하므로, 적시에 발견하여 치료하기가 매우 어렵다. 또한, 암은 조직 내의 하나 또는 수개의 정상 세포가 악성 전환을 통해 신체 내의 거의 모든 조직으로부터 발생할 수 있으며, 특정한 조직 기원을 갖는 암의 유형 마다 서로 상이하다.Cancer is one of the most deadly threats to human health. In the United States, nearly 1,300,000 new patients get cancer each year, causing about one in four deaths following heart disease. It is also predicted that cancer can surpass cardiovascular disease as the first cause of death within five years. Solid cancer accounts for most of these deaths. Although significant progress has been made in the medical treatment of certain cancers, the overall 5-year survival rate for all cancers has improved only about 10% over the past 20 years. Cancer or malignant tumors metastasize and grow rapidly in an uncontrolled manner, making it very difficult to detect and treat in a timely manner. In addition, cancer can occur from almost any tissue in the body through one or several normal cells in the tissue through malignant transformation, and differ from one type of cancer with a particular tissue origin.
암을 치료하기 위한 현행 방법은 상대적으로 비-선택적이다. 수술로 질병을 갖는 조직을 제거하고; 방사선 치료로 고형 종양을 감소시키며; 화학치료로 빠르게 분열하는 세포를 죽인다. 특히, 화학치료는 다양한 부작용을 낳아, 일부의 경우엔 투여될 수 있는 용량을 제한하여 결국엔 잠재적으로 유효한 약물의 사용을 배제시킬 정도로 부작용이 매우 심각하다. 또한, 암은 종종 화학요법 약물에 대해 내성을 발달시킨다. 따라서, 특이적이고 보다 효과적인 암 치료가 시급하다.Current methods for treating cancer are relatively non-selective. Surgery to remove diseased tissue; Radiation treatment reduces solid tumors; Chemotherapy kills rapidly dividing cells. In particular, chemotherapy produces a variety of side effects, which in some cases are so severe that they limit the dose that can be administered and eventually rule out the use of potentially effective drugs. In addition, cancer often develops resistance to chemotherapy drugs. Therefore, specific and more effective cancer treatments are urgent.
종양세포는 "종양 항원"의 존재로 인해 정상 세포와 항원성에 있어서 상이하다. 이들은 종양세포에만 특유할 수 있거나, 또는 상이하게 발현되거나 과량으로 발현됨으로써 "종양-관련 항원"(TAA)으로서 인식된다. 종양세포는 단백질 1차 구조, 즉 아미노산 서열이 정상 세포와 상이할 뿐만 아니라(게놈 서열의 변화로부터 유래), 글리코실화, 포스포릴화의 변화와 같이 번역 후 변형의 변화로 인하여 2차 및 3차 구조도 정상 세포와 상이할 수 있으며, 이것은 계속해서 상기 단백질의 항원성을 변화시키고, 또한 그것을 종양-관련 항원으로서 특정한다. 한 전형적인 예는 유선에서 나오는 단백질 점액소인데, 이것 자체는 종양 세포에 존재하는 변화는 아니지만, 유방암 환자에서는 이것에 대한 자가항체가 발견된다(때로는 난소암에서도 그렇다). 그 이유는 아마도 정상 세포에서는 이 단백질이 고도로 글리코실화되고, 탄수화물 사슬의 치밀하고 두꺼운 코팅으로 인해서 전혀 노출되지 않기 때문인 것으로 추정된다. 종양세포에서 글리코실화가 불량하면, 항원성 결정소로서 작용하는 단백질 단편이 면역 시스템에 노출되게 된다.Tumor cells differ in antigenicity from normal cells due to the presence of "tumor antigens". They may be specific to tumor cells, or are recognized as "tumor-associated antigens" (TAAs) by being expressed differently or in excess. Tumor cells not only differ in protein primary structure, i.e. from amino acid sequences, from normal cell changes, but also in secondary and tertiary due to changes in post-translational modifications, such as changes in glycosylation and phosphorylation. The structure may also differ from normal cells, which continually change the antigenicity of the protein and also specify it as a tumor-associated antigen. One typical example is protein mucin from the mammary gland, which itself is not a change present in tumor cells, but autoantibodies to it are found in breast cancer patients (sometimes in ovarian cancer). The reason is presumably because the protein is highly glycosylated in normal cells and is not exposed at all due to the dense and thick coating of carbohydrate chains. Poor glycosylation in tumor cells results in exposure of protein fragments that act as antigenic determinants to the immune system.
TAA는 현재 특정 종양, 예를 들어 림프종, 암종, 육종 또는 흑색종과 결합될수 있는 분자인 것으로 생각되며, 종양에 대해 세포성 및/또는 체액성 면역반응을 도출할 수 있고, 드물게는 종양에 대해 숙주를 방어하기도 한다. 이와 같은 TAA는 현재 3가지 부류로 분류되는데, 1명의 또는 단지 소수의 개체에만 존재하고 정상 세포에서는 발견되지 않는 특정 종양에 대해 고도로 특이적인 것, 예를 들어 종양-특이적 이식 항원(제 1 부류); 상이한 환자들의 다수의 관련된 종양에 존재하는 것(제 2 부류); 및 정상세포와 악성 세포에 존재하며, 악성 세포에서 다량으로 발현되는 것(제 3 부류)이다. 제 2 부류의 TAA는 많은 종양에 존재하고 정상 피험자에서는 드물게 관찰되기 때문에 임상적으로 유용한 분석을 위한 잠재성이 가장 큰 것으로 생각된다.TAA is currently thought to be a molecule that can be associated with certain tumors, such as lymphomas, carcinomas, sarcomas, or melanoma, and can elicit cellular and / or humoral immune responses against tumors and, rarely, against tumors. It also defends the host. Such TAAs are currently classified into three classes, which are highly specific for certain tumors that are present in only one or only a few individuals and are not found in normal cells, for example tumor-specific transplant antigens (class 1). ); Present in a large number of related tumors of different patients (second class); And present in normal cells and malignant cells and expressed in large quantities in malignant cells (third class). Since the second class of TAA is present in many tumors and rarely observed in normal subjects, it is considered to have the greatest potential for clinically useful analyses.
따라서, 특이적이고 보다 효과적인 암 치료 방법으로 상기 종양세포 특이적 항원을 표적하는 항체를 사용하는 연구 및, 상기 항체를 통해 암세포의 세포사멸을 유도하는 암치료제 개발이 현재 관심을 받고 있다. 상기 기술과 관련하여 국내공개특허공보 10-2010-0045903은 암세포의 세포사멸 수용체인 항 DR5를 표적하는 항체를 개시하고 있다. Therefore, the use of antibodies targeting the tumor cell specific antigens as a specific and more effective cancer treatment method, and the development of cancer therapeutic agents that induce apoptosis of cancer cells through the antibody is currently of interest. In connection with the above technology, Korean Laid-Open Patent Publication 10-2010-0045903 discloses an antibody targeting anti DR5, which is an apoptosis receptor for cancer cells.
따라서 본 발명은 종양세포 특이적 항원을 표적하는 신규 항체 및 상기 항체를 통해 암세포의 세포사멸을 유도하는 암 치료 방법을 제공하고자 한다. Therefore, the present invention is to provide a novel antibody targeting a tumor cell specific antigen and a method for treating cancer inducing apoptosis of cancer cells through the antibody.
상기 과제의 해결을 위하여, 본 발명은 암세포에 결합하여 세포사멸을 유도하는 단일클론 항체로서, In order to solve the above problems, the present invention binds to cancer cells as a monoclonal antibody inducing apoptosis,
서열번호 1의 아미노산 서열로 이루어진 경쇄 CDR1, 서열번호 2의 아미노산 서열로 이루어진 경쇄 CDR2 및 서열번호 3의 아미노산 서열로 이루어진 경쇄 CDR3 를 포함하는 경쇄 가변 영역 및;A light chain variable region comprising a light chain CDR1 consisting of the amino acid sequence of SEQ ID NO: 1, a light chain CDR2 consisting of the amino acid sequence of SEQ ID NO: 2, and a light chain CDR3 consisting of the amino acid sequence of SEQ ID NO: 3;
서열번호 4의 아미노산 서열로 이루어진 중쇄 CDR1, 서열번호 5의 아미노산 서열로 이루어진 중쇄 CDR2 및 서열번호 6의 아미노산 서열로 이루어진 중쇄 CDR3 를 포함하는 중쇄 가변영역을 포함하는 항체를 제공한다. Provided is an antibody comprising a heavy chain variable region comprising a heavy chain CDR1 consisting of the amino acid sequence of SEQ ID NO: 4, a heavy chain CDR2 consisting of the amino acid sequence of SEQ ID NO: 5, and a heavy chain CDR3 consisting of the amino acid sequence of SEQ ID NO: 6.
본 발명의 한 구체예에서, 상기 항체는 단일쇄 가변영역 (scFv, Single-chain variable fragment) 항체 이며, 서열번호 9, 서열번호 13, 서열번호 17 또는 서열번호 21의 아미노산 서열을 갖는 것일 수 있다. In one embodiment of the invention, the antibody is a single-chain variable fragment (scFv, single-chain variable fragment) antibody, and may have an amino acid sequence of SEQ ID NO: 9, SEQ ID NO: 13, SEQ ID NO: 17 or SEQ ID NO: 21. .
또한 본 발명은 상기 단일쇄 가변영역 항체를 코딩하는 유전자를 제공한다. The present invention also provides a gene encoding the single chain variable region antibody.
또한 본 발명은 상기 항체를 유효성분으로 함유하는 암 예방 또는 치료용 약학 조성물을 제공한다. The present invention also provides a pharmaceutical composition for preventing or treating cancer containing the antibody as an active ingredient.
또한 본 발명은 상기 항체를 포함하고, 상기 항체에 하나 또는 둘 이상의 진단제 또는 항암제를 결합시킨 암세포 표적화 진단 또는 치료 컨쥬게이트를 제공한다. The present invention also provides a cancer cell targeting diagnostic or therapeutic conjugate comprising the antibody, wherein one or more diagnostic or anticancer agents are bound to the antibody.
본 발명의 항체는 암세포주에 특이적으로 결합하는 활성을 갖는 바, 이를 이용하여 암세포 표적화 진단 또는 치료 컨쥬게이트로서 활용하여 암을 효과적으로 진단하거나 치료할 수 있다. 또한, 본 발명의 항체는 암세포, 특히 림프종 세포 및 난소암 세포에 결합하여 세포사멸을 유도하는 특성을 보이는 바, 이를 통해 본 발명의 항체 자체를 항암제로 사용할 수도 있다.The antibody of the present invention has an activity of specifically binding to a cancer cell line, and can be used as a cancer cell targeting diagnosis or therapeutic conjugate to effectively diagnose or treat cancer. In addition, the antibody of the present invention shows a property of inducing apoptosis by binding to cancer cells, particularly lymphoma cells and ovarian cancer cells, through which the antibody of the present invention can be used as an anticancer agent.
도 1은 실시예 1의 IM9세포주에 대한 유세포분석 결과이다. 1 is a flow cytometry result for the IM9 cell line of Example 1.
도 2는 본 발명에 따른 scFv 클론 ITTab1-1의 뉴클레오타이드 및 아미노산 서열 및 이의 CDR 및 링커 영역을 나타낸 것이다. Figure 2 shows the nucleotide and amino acid sequences of the scFv clone ITTab1-1 and CDRs and linker regions thereof according to the present invention.
도 3은 본 발명에 따른 scFv 클론 ITTab1-2의 뉴클레오타이드 및 아미노산 서열 및 이의 CDR 및 링커 영역을 나타낸 것이다. Figure 3 shows the nucleotide and amino acid sequences of the scFv clone ITTab1-2 and its CDR and linker regions according to the present invention.
도 4는 본 발명에 따른 scFv 클론 ITTab1-3의 뉴클레오타이드 및 아미노산 서열 및 이의 CDR 및 링커 영역을 나타낸 것이다. Figure 4 shows the nucleotide and amino acid sequence of the scFv clone ITTab1-3 and its CDR and linker regions according to the present invention.
도 5는 본 발명에 따른 scFv 클론 ITTab1-4의 뉴클레오타이드 및 아미노산 서열 및 이의 CDR 및 링커 영역을 나타낸 것이다. Figure 5 shows the nucleotide and amino acid sequences of the scFv clone ITTab1-4 and its CDR and linker regions according to the present invention.
도 6은 본 발명의 항체의 NK 림프종세포에 대한 결합여부를 검토한 공초점현미경 결과 사진이다. 6 is a photograph of confocal microscopy results of examining the binding of the antibody of the present invention to NK lymphoma cells.
도 7은 본 발명의 항체처리로 인한 NK 림프종세포의 세포자멸사를 검토한 유세포분석 실험 결과이다. Figure 7 is a flow cytometry test results of examining apoptosis of NK lymphoma cells by the antibody treatment of the present invention.
도 8은 본 발명의 항체의 난소암 세포에 대한 결합여부를 검토한 유세포분석 실험 결과이다. 8 is a flow cytometry test result of examining the binding of the antibody of the present invention to ovarian cancer cells.
도 9는 본 발명의 항체처리로 인한 난소암세포의 세포자멸사를 검토한 유세포분석 실험 결과이다. 9 is a flow cytometry test result of examining apoptosis of ovarian cancer cells by the antibody treatment of the present invention.
본 발명은 암세포에 결합하여 세포사멸을 유도하는 단일클론 항체로서, The present invention is a monoclonal antibody that binds to cancer cells and induces apoptosis,
서열번호 1의 아미노산 서열로 이루어진 경쇄 CDR1, 서열번호 2의 아미노산 서열로 이루어진 경쇄 CDR2 및 서열번호 3의 아미노산 서열로 이루어진 경쇄 CDR3 를 포함하는 경쇄 가변 영역 및; 서열번호 4의 아미노산 서열로 이루어진 중쇄 CDR1, 서열번호 5의 아미노산 서열로 이루어진 중쇄 CDR2 및 서열번호 6의 아미노산 서열로 이루어진 중쇄 CDR3 를 포함하는 중쇄 가변영역을 포함하는 항체를 제공한다. A light chain variable region comprising a light chain CDR1 consisting of the amino acid sequence of SEQ ID NO: 1, a light chain CDR2 consisting of the amino acid sequence of SEQ ID NO: 2, and a light chain CDR3 consisting of the amino acid sequence of SEQ ID NO: 3; Provided is an antibody comprising a heavy chain variable region comprising a heavy chain CDR1 consisting of the amino acid sequence of SEQ ID NO: 4, a heavy chain CDR2 consisting of the amino acid sequence of SEQ ID NO: 5, and a heavy chain CDR3 consisting of the amino acid sequence of SEQ ID NO: 6.
본 발명의 발명자들은 종양세포 특이적 항원을 표적하는 신규 항체에 대해 연구하던 중, ConA 로 자극된 PBMC를 사용하여 B cell tumor cells (IM9)에 특이적으로 결합하는 하이브리도마 세포를 구축하였고, 이를 통해 제조된 단일클론항체가 암세포, 특히 NK 림프종 세포 및 난소암 세포와 특이적으로 결합하며, 이의 세포사멸 또한 유도한다는 점을 확인하고 본 발명을 완성하게 되었다. The inventors of the present invention, while studying a novel antibody targeting a tumor cell specific antigen, constructed a hybridoma cell that specifically binds to B cell tumor cells (IM9) using PBMC stimulated with ConA, The monoclonal antibody prepared by this specifically binds to cancer cells, in particular NK lymphoma cells and ovarian cancer cells, and confirmed that it also induces apoptosis, thereby completing the present invention.
본 발명의 한 구체예에서, 상기 경쇄 가변영역은 서열번호 7, 서열번호 11, 서열번호 15 또는 서열번호 19의 아미노산 서열로 이루어질 수 있다. In one embodiment of the invention, the light chain variable region may consist of the amino acid sequence of SEQ ID NO: 7, SEQ ID NO: 11, SEQ ID NO: 15 or SEQ ID NO: 19.
본 발명의 다른 구체예에서, 상기 중쇄 가변영역은 서열번호 8, 서열번호 12, 서열번호 16 또는 서열번호 20의 아미노산 서열로 이루어질 수 있다. In another embodiment of the present invention, the heavy chain variable region may be composed of the amino acid sequence of SEQ ID NO: 8, SEQ ID NO: 12, SEQ ID NO: 16 or SEQ ID NO: 20.
본 발명에서 "항체"는 전체 형태의 항체("전항체") 또는 그의 기능적인 단편일 수 있다. 상기 전항체는 단량체 또는 2 이상의 전항체가 결합되어 있는 다량체의 형태일 수 있다. 상기 항체의 기능적인 단편은 전항체의 중쇄 및 경쇄 가변영역을 갖는 항체로서, 실질적으로 전항체가 인식하는 것과 동일한 항원결합부위 (epitope)를 인식하는 것을 의미한다. 상기 항체의 기능적인 단편에는 단일쇄 가변영역 단편 (scFv), (scFv)2, Fab, Fab' 및 F(ab')2 등이 포함되나 이에 한정되지 않으며, 본 발명에서는 단일쇄 가변영역 단편(scFv, Single-chain variable fragment) 형태의 항체가 바람직하다. 상기 단일쇄 가변영역(scFv)은 중쇄 가변영역과 경쇄 가변영역이 링커 펩타이드를 통하여 연결되어 단일쇄 폴리펩티드 형태를 취하는 항체 단편을 의미한다. 상기 항체는 당업계에 알려져 있는 방법, 예를 들어, 파지 디스플레이 방법 또는 효모 세포 표면 발현 시스템을 사용하여 생성될 수 있다.In the present invention, an "antibody" may be an entire form of an antibody ("antibody") or a functional fragment thereof. The whole antibody may be in the form of a monomer or a multimer in which two or more whole antibodies are bound. The functional fragment of the antibody is an antibody having the heavy and light chain variable regions of the whole antibody, which means to recognize substantially the same epitope that the whole antibody recognizes. Functional fragments of the antibody include, but are not limited to, single chain variable region fragments (scFv), (scFv) 2 , Fab, Fab 'and F (ab') 2, and the like. Antibodies in the form of single-chain variable fragments (scFv) are preferred. The single chain variable region (scFv) refers to an antibody fragment in which a heavy chain variable region and a light chain variable region are linked through a linker peptide to take the form of a single chain polypeptide. The antibody can be generated using methods known in the art, such as phage display methods or yeast cell surface expression systems.
본 발명의 또 다른 구체예에서, 본 발명의 항체는 단일쇄 가변영역 항체이며, 이는 서열번호 9, 서열번호 13, 서열번호 17 또는 서열번호 21의 아미노산 서열을 갖는 것일 수 있다. In another embodiment of the invention, the antibody of the invention is a single chain variable region antibody, which may have an amino acid sequence of SEQ ID NO: 9, SEQ ID NO: 13, SEQ ID NO: 17 or SEQ ID NO: 21.
또한, 본 발명의 항체는 인간을 포함하는 포유동물, 조류 등을 포함한 임의의 동물로부터 유래한 것일 수 있다. 바람직하게는, 상기 항체는 인간, 생쥐, 당나귀, 양, 토끼, 염소, 기니피그, 낙타, 말 또는 닭의 항체일 수 있다. In addition, the antibodies of the present invention may be derived from any animal, including mammals, birds, and the like, including humans. Preferably, the antibody may be a human, mouse, donkey, sheep, rabbit, goat, guinea pig, camel, horse or chicken antibody.
여기서 인간 항체는 인간 면역글로불린의 아미노산 서열을 가진 항체로서, 인간 면역글로불린 라이브러리로부터 분리된 항체 또는 하나 이상의 인간 면역글로불린에 대하여 형질 이식되고 내재적 면역글로불린은 발현하지 않는 동물로부터 분리된 항체가 포함된다.Wherein the human antibody is an antibody having an amino acid sequence of human immunoglobulin, and includes an antibody isolated from a human immunoglobulin library or an antibody isolated from an animal transfected against one or more human immunoglobulins and not expressing an endogenous immunoglobulin.
또한 본 발명은 상기 서열번호 9, 서열번호 13, 서열번호 17 또는 서열번호 21의 아미노산 서열을 갖는 단일쇄 가변영역 항체 각각을 코딩하는 유전자를 제공한다. The present invention also provides a gene encoding each of the single-chain variable region antibodies having the amino acid sequence of SEQ ID NO: 9, SEQ ID NO: 13, SEQ ID NO: 17 or SEQ ID NO: 21.
본 발명의 항체를 코딩하는 유전자 서열은 당업계에 잘 알려진 방법에 의하여 얻어질 수 있다. 예를 들면, 상기 항체의 중쇄 및 경쇄의 일부분 또는 전부를 코딩하는 DNA 서열 또는 해당 아미노산 서열에 근거하여, 당분야에 잘 알려진 올리고뉴클레오티드 합성기법, 예를 들어 부위 지향적 돌연변이 발생법(site-directed mutagenesis) 및 중합효소 연쇄 반응(PCR)법 등을 사용하여 원하는 대로 합성할 수 있다.Gene sequences encoding the antibodies of the invention can be obtained by methods well known in the art. For example, based on DNA sequences or corresponding amino acid sequences encoding portions or all of the heavy and light chains of the antibody, oligonucleotide synthesis techniques well known in the art, for example site-directed mutagenesis And the polymerase chain reaction (PCR) method.
보다 구체적으로, 상기 서열번호 9의 아미노산 서열을 갖는 단일쇄 가변영역 항체를 코딩하는 유전자는 서열번호 10의 염기서열로 이루어진 것일 수 있다. More specifically, the gene encoding the single-chain variable region antibody having the amino acid sequence of SEQ ID NO: 9 may be composed of the nucleotide sequence of SEQ ID NO: 10.
또한, 상기 서열번호 13의 아미노산 서열을 갖는 단일쇄 가변영역 항체를 코딩하는 유전자는 서열번호 14의 염기서열로 이루어진 것일 수 있다. In addition, the gene encoding the single-chain variable region antibody having the amino acid sequence of SEQ ID NO: 13 may be composed of the nucleotide sequence of SEQ ID NO: 14.
또한, 상기 서열번호 17의 아미노산 서열을 갖는 단일쇄 가변영역 항체를 코딩하는 유전자는 서열번호 18의 염기서열로 이루어진 것일 수 있다. In addition, the gene encoding the single-chain variable region antibody having the amino acid sequence of SEQ ID NO: 17 may be composed of the nucleotide sequence of SEQ ID NO: 18.
또한, 상기 서열번호 21의 아미노산 서열을 갖는 단일쇄 가변영역 항체를 코딩하는 유전자는 서열번호 22의 염기서열로 이루어진 것일 수 있다. In addition, the gene encoding the single-chain variable region antibody having the amino acid sequence of SEQ ID NO: 21 may be composed of the nucleotide sequence of SEQ ID NO: 22.
본 발명의 일실시예에서, 상기 유전자 및 이를 포함하는 발현벡터로 숙주세포를 형질전환시켜 적절한 조건 하에서 배양하여 항체를 발현시키고 분리하여 항체 분자를 생산할 수 있다. In one embodiment of the present invention, the host cell is transformed with the gene and the expression vector including the same, and cultured under appropriate conditions to express and isolate the antibody, thereby producing an antibody molecule.
본 발명의 항체는 암세포에 특이적으로 결합하는 특성을 보이며, 특히 림프종 세포 및 난소암 세포에 결합하여 세포사멸을 유도하는 특성을 보이는 바, 이는 암의 예방 또는 치료에 사용될 수 있다. The antibody of the present invention exhibits a property of specifically binding to cancer cells, and in particular, binds to lymphoma cells and ovarian cancer cells and induces cell death, and thus can be used for the prevention or treatment of cancer.
따라서 본 발명은 상기 항체를 유효성분으로 함유하는 암 예방 또는 치료용 약학조성물, 항암제의 제조를 위한 상기 항체의 용도, 그리고 치료상 유효량의 상기 항체를 대상체에 투여하는 단계를 포함하는 암의 치료 방법을 제공한다. Accordingly, the present invention provides a pharmaceutical composition for preventing or treating cancer containing the antibody as an active ingredient, the use of the antibody for the manufacture of an anticancer agent, and administering to the subject a therapeutically effective amount of the antibody to a subject. To provide.
본 발명의 한 구체예에서, 상기 항체를 유효성분으로 함유하는 암 예방 또는 치료용 약학 조성물은 약학 조성물의 제조에 통상적으로 사용하는 사용하는 적절한 담체, 부형제, 붕해제, 감미제, 피복제, 팽창제, 윤활제, 활택제, 향미제, 항산화제, 완충액, 정균제, 희석제, 분산제, 계면활성제, 결합제 및 윤활제로 이루어진 군에서 선택되는 하나 이상의 첨가제를 추가로 포함할 수 있다.In one embodiment of the present invention, the pharmaceutical composition for preventing or treating cancer containing the antibody as an active ingredient is a suitable carrier, excipient, disintegrant, sweetener, coating agent, swelling agent, It may further comprise one or more additives selected from the group consisting of lubricants, lubricants, flavors, antioxidants, buffers, bacteriostatics, diluents, dispersants, surfactants, binders and lubricants.
구체적으로 담체, 부형제 및 희석제는 락토즈, 덱스트로즈, 수크로스, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로즈, 메틸 셀룰로즈, 미정질 셀룰로스, 폴리비닐 피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유를 사용할 수 있으며, 경구투여를 위한 고형제제에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 이러한 고형제제는 상기 조성물에 적어도 하나 이상의 부형제, 예를 들면, 전분, 칼슘카보네이트, 수크로스 또는 락토오스, 젤라틴 등을 섞어 조제할 수 있다. 또한 단순한 부형제 이외에 마그네슘 스티레이트, 탈크 같은 윤활제들도 사용할 수 있다. 경구를 위한 액상제제로는 현탁제, 내용액제, 유제, 시럽제 등이 있으며 흔히 사용되는 단순 희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조제제, 좌제 등이 포함된다. 비수성용제, 현탁제로는 프로필렌글리콜, 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기재로는 위텝솔(witepsol), 마크로골, 트윈(tween) 61, 카카오지, 라우린지, 글리세로제라틴 등이 사용될 수 있다.Specifically, the carriers, excipients and diluents are lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline Cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil can be used, and solid preparations for oral administration include tablets, pills, powders, granules, capsules. And the like, and such solid preparations may be prepared by mixing at least one excipient such as starch, calcium carbonate, sucrose or lactose, gelatin and the like in the composition. In addition to simple excipients, lubricants such as magnesium styrate and talc may also be used. Oral liquid preparations include suspensions, solvents, emulsions, syrups, and the like, and may include various excipients such as wetting agents, sweeteners, fragrances, and preservatives, in addition to commonly used simple diluents such as water and liquid paraffin. Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized preparations, suppositories, and the like. As the non-aqueous solvent and suspending agent, propylene glycol, polyethylene glycol, vegetable oil such as olive oil, injectable ester such as ethyl oleate and the like can be used. Witsol, macrogol, tween 61, cacao butter, laurin butter, glycerogelatin and the like may be used as the base material of the suppository.
본 발명의 다른 구체예에서, 상기 항체를 유효성분으로 함유하는 암 예방 또는 치료용 약학 조성물은 통상적인 방법에 따라 과립제, 산제, 피복정, 정제, 환제, 캡슐제, 좌제, 겔, 시럽, 즙, 현탁제, 유제, 점적제 또는 액제로 제형화하여 사용할 수 있다.In another embodiment of the present invention, the pharmaceutical composition for preventing or treating cancer containing the antibody as an active ingredient is a granule, powder, coated tablet, tablet, pill, capsule, suppository, gel, syrup, juice It can be formulated as a suspension, emulsion, drop or liquid.
본 발명의 일실시예에 따르면 상기 약학 조성물은 정맥내, 동맥내, 복강내, 근육내, 동맥내, 복강내, 흉골내, 경피, 비측내, 흡입, 국소, 직장, 경구, 안구내 또는 피내 경로를 통해 통상적인 방식으로 대상체로 투여할 수 있다.According to one embodiment of the invention the pharmaceutical composition is intravenous, intraarterial, intraperitoneal, intramuscular, intraarterial, intraperitoneal, intrasternal, transdermal, nasal, inhaled, topical, rectal, oral, intraocular or intradermal Via the route can be administered to the subject in a conventional manner.
상기 항체의 바람직한 투여량은 대상체의 상태 및 체중, 질환의 종류 및 정도, 약물 형태, 투여경로 및 기간에 따라 달라질 수 있으며 당업자에 의해 적절하게 선택될 수 있다. 본 발명의 일실시예에 따르면 이에 제한되는 것은 아니지만 1일 투여량이 0.01 내지 200 mg/kg, 구체적으로는 0.1 내지 200 mg/kg, 보다 구체적으로는 0.1 내지 100 mg/kg 일 수 있다. 투여는 하루에 한 번 투여할 수도 있고 수회로 나누어 투여할 수도 있으며, 이에 의해 본 발명의 범위가 제한되는 것은 아니다.The preferred dosage of the antibody may vary depending on the condition and weight of the subject, the type and extent of the disease, the form of the drug, the route of administration, and the duration and may be appropriately selected by those skilled in the art. According to one embodiment of the present invention, but not limited thereto, the daily dosage may be 0.01 to 200 mg / kg, specifically 0.1 to 200 mg / kg, more specifically 0.1 to 100 mg / kg. Administration may be administered once a day or divided into several times, thereby not limiting the scope of the invention.
본 발명에 있어서, 상기 '대상체'는 인간을 포함하는 포유동물일 수 있으나, 이들 예에 한정되는 것은 아니다.In the present invention, the 'subject' may be a mammal including a human, but is not limited thereto.
상기 암으로는 간암, 폐암, 대장암, 난소암, 유방암, 전립선암, 백혈병, 림프종 및 췌장암으로부터 선택되는 어느 하나일 수 있다.The cancer may be any one selected from liver cancer, lung cancer, colon cancer, ovarian cancer, breast cancer, prostate cancer, leukemia, lymphoma and pancreatic cancer.
또한, 본 발명은 상기 항체를 포함하고, 상기 항체에 하나 또는 둘 이상의 진단제 또는 항암제를 결합시킨 암세포 표적화 진단 또는 치료 컨쥬게이트를 제공한다.The present invention also provides a cancer cell targeting diagnostic or therapeutic conjugate comprising the antibody and binding one or more diagnostic or anticancer agents to the antibody.
상기 진단제로는 방사성핵종, 상자성 금속이온, 초상자성 나노입자, 발색효소, 크로모포어, 발광물질 및 형광물질로 이루어진 군에서 선택된 어느 하나 또는 둘 이상을 사용할 수 있으며, 상기 조영제는 CT 조영제, MRI 조영제, 및 광학 조영제, 및 초음파 조영제로 이루어진 군에서 선택된 어느 하나일 수 있다. The diagnostic agent may be any one or two or more selected from the group consisting of radionuclides, paramagnetic metal ions, superparamagnetic nanoparticles, chromophores, chromophores, luminescent materials, and fluorescent materials, and the contrast agent may be CT contrast agent, MRI. It may be any one selected from the group consisting of a contrast agent, an optical contrast agent, and an ultrasound contrast agent.
상기 항암제로는 종래 암의 치료에 사용되는 것이라면 제한없이 사용될 수 있으며, 예를들면 시스플라틴, 5-플로오우라실, 아드리아마이신, 메토트렉세이트, 빈블라스틴, 부설판, 클로람부실, 시클로포스파미드, 멜팔란, 니트로겐 무스타드, 니트로소우레아, 탁솔, 파클리탁셀, 도세탁셀, 6-머캅토퓨린, 6-티오구아닌, 블레오마이신, 다우노루비신, 독소루비신, 에피루비신, 이다루비신, 미토마이신-C 및 하이드록시우레아로 이루어진 군으로부터 선택되는 어느 하나 또는 둘 이상을 사용할 수 있다. 항암제와 본 발명의 항체와의 연결은 당업계에 공지된 방법, 예컨대, 공유 결합, 가교 등을 통해 수행될 수 있다.The anticancer agent may be used without limitation as long as it is conventionally used for the treatment of cancer, for example cisplatin, 5-fluorouracil, adriamycin, methotrexate, vinblastine, busulfan, chlorambucil, cyclophosphamide, Melphalan, nitrogen mustard, nitrosourea, taxol, paclitaxel, docetaxel, 6-mercaptopurine, 6-thioguanine, bleomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin-C And hydroxyurea can be used any one or two or more selected from the group consisting of. The linkage between the anticancer agent and the antibody of the present invention can be carried out through methods known in the art, such as covalent bonds, crosslinking and the like.
또한, 상기 항암제로는 치료용 방사성 핵종, 예를들어 32P, 47Sc, 64Ga, 64Cu, 66Cu, 67Cu, 89Sr, 90Y, 91Y, 103Pd, 103mRh, 105Rh, 111Ag, 111In, 117mSn, 125I, 131I, 145Sm, 149Pm, 153Sm, 165Dy, 166Ho, 169Er, 176Lu, 177Lu, 182Ta, 186Re, 188Re, 188W, 192Ir, 195mPt, 198Au, 199Au, 211At, 212Bi, 213Bi, 223Ra, 225Ac, 230U, 252Cf 또는 이들의 조합 등을 사용할 수 있다.In addition, the anticancer agent may be a therapeutic radionuclide, for example, 32 P, 47 Sc, 64 Ga, 64 Cu, 66 Cu, 67 Cu, 89 Sr, 90 Y, 91 Y, 103 Pd, 103 m Rh, 105 Rh, 111 Ag, 111 In, 117m Sn, 125 I, 131 I, 145 Sm, 149 Pm, 153 Sm, 165 Dy, 166 Ho, 169 Er, 176 Lu, 177 Lu, 182 Ta, 186 Re, 188 Re, 188 W , 192 Ir, 195 m Pt, 198 Au, 199 Au, 211 At, 212 Bi, 213 Bi, 223 Ra, 225 Ac, 230 U, 252 Cf, or a combination thereof.
본 발명의 암세포 표적화 치료 컨쥬게이트에 포함되는 본 발명의 항체의 양은 결합되는 항암제의 종류 및 양에 따라 달라질 수 있다. 바람직하게는 치료를 위해 투여되는 양의 항암제를 표적 암질환 부위에 충분히 전달할 수 있는 양일 수 있다. 그러나, 약물은 그 투여 경로 및 치료 횟수뿐 만 아니라 환자의 연령, 체중, 건강 상태, 성별, 질환의 중증도, 식이 및 배설율 등 다양한 요인들을 고려하여 환자에 대한 유효 투여량이 결정되는 것이므로, 이러한 점을 고려할 때 당 분야의 통상적인 지식을 가진 자라면 상기 항체를 항암제와 결합시켜 암질환을 치료하기 위한 용도에 따른 적절한 유효량을 결정할 수 있을 것이다.The amount of the antibody of the present invention included in the cancer cell targeting therapeutic conjugate of the present invention may vary depending on the type and amount of the anticancer agent to be bound. Preferably, the amount of the anticancer agent administered for treatment may be an amount sufficient to deliver the target cancer disease site. However, since the drug is determined not only by the route of administration and the number of treatments, but also by various factors such as the age, weight, health condition, sex, severity of the disease, diet, and excretion rate, the effective dose to the patient is determined. In view of this, one of ordinary skill in the art will be able to determine an appropriate effective amount depending on the use of the antibody for treating cancer diseases by combining the antibody with an anticancer agent.
본 발명의 암세포 표적화 치료 컨쥬게이트에는 상기 항체의 안정성을 유지/보존하기 위한 적당한 완충용액이 포함될 수 있다. 본 발명의 암세포 표적화 치료 컨쥬게이트는 본 발명에서 원하는 효과를 나타내는 한, 그 제형, 투여 경로 및 투여 방법에 특별히 제한되지 아니한다. 예컨대, 본 발명의 컨쥬게이트는 피하 주사, 근육내 주사, 정맥 주사, 요도 주입, 기관지내 흡입, 자궁내 경막 또는 뇌혈관내 주사 등과 같이 비경구로 투여될 수 있다.The cancer cell targeting therapeutic conjugate of the present invention may include a suitable buffer solution for maintaining / preserving the stability of the antibody. The cancer cell targeting therapeutic conjugate of the present invention is not particularly limited to its formulation, route of administration, and method of administration so long as the desired effect is achieved in the present invention. For example, the conjugates of the present invention can be administered parenterally, such as subcutaneous injection, intramuscular injection, intravenous injection, urethral injection, intrabronchial inhalation, intrauterine dural or cerebrovascular injection, and the like.
이외에도 본 발명의 암세포 표적화 치료 컨쥬게이트에는 일반적인 약학조성물에 통상적으로 첨가되는 적절한 담체, 부형제, 붕해제, 감미제, 피복제, 팽창제, 윤활제, 활택제, 향미제, 항산화제, 완충액, 정균제, 희석제, 분산제, 계면활성제, 결합제 및 윤활제로 이루어진 군에서 선택되는 하나 이상의 첨가제를 추가로 포함할 수 있다. In addition, the cancer cell targeting therapeutic conjugates of the present invention include appropriate carriers, excipients, disintegrants, sweeteners, coating agents, swelling agents, lubricants, lubricants, flavoring agents, antioxidants, buffers, bactericides, diluents, It may further comprise one or more additives selected from the group consisting of dispersants, surfactants, binders and lubricants.
또한 본 발명의 암세포 표적화 치료 컨쥬게이트는 통상적인 방법에 따라 과립제, 산제, 피복정, 정제, 환제, 캡슐제, 좌제, 겔, 시럽, 즙, 현탁제, 유제, 점적제 또는 액제로 제형화하여 사용할 수 있다. 예를 들어, 주사제의 경우에는 단위 투약 앰플 또는 다수회 투약 포함제 형태로 제조할 수 있다.In addition, the cancer cell targeting therapeutic conjugate of the present invention may be formulated into granules, powders, coated tablets, tablets, pills, capsules, suppositories, gels, syrups, juices, suspensions, emulsions, drops or solutions according to conventional methods. Can be used. For example, injectables can be prepared in the form of unit dose ampoules or multiple dose inclusions.
이하, 본 발명의 이해를 돕기 위하여 실시예를 들어 상세하게 설명하기로 한다. 다만 하기의 실시예는 본 발명의 내용을 예시하는 것일 뿐 본 발명의 범위가 하기 실시예에 한정되는 것은 아니다. 본 발명의 실시예는 당업계에서 평균적인 지식을 가진 자에게 본 발명을 보다 완전하게 설명하기 위해 제공되는 것이다.Hereinafter, examples will be described in detail to help understand the present invention. However, the following examples are merely to illustrate the content of the present invention is not limited to the scope of the present invention. The embodiments of the present invention are provided to more completely explain the present invention to those skilled in the art.
<제조예 1> scFv(single chain Fv)인 ITTab1 항체 제작Preparation Example 1 Preparation of ITTab1 Antibody as a scFv (single chain Fv)
1. 하이브리도마 세포주 구축1. Hybridoma Cell Line Construction
마우스는 5-6주령 암컷 Balb/c 마우스를 오리엔트바이오에서 구입하여 사용하였다. 구입한 마우스는 온도 20-25℃, 습도 60%, 조도 200-300 Lux 의 실내 환경에서 고형사료와 물을 충분히 공급하여 1주일간 순화를 진행하였다. Mice were 5-6 week old female Balb / c mice purchased from Orient Bio. The purchased mouse was purified for 1 week by supplying sufficient solid feed and water in an indoor environment with a temperature of 20-25 ° C., a humidity of 60%, and an illuminance of 200-300 Lux .
건강한 지원자로부터 Ficoll-gradient 방법으로 분리한 PBMC 1X107를 Con A (콘카나발린 A) 10ug/ml 로 자극한 후 마우스에 면역한 후, 2달 후에 마우스의 비장을 분리하여 1X108의 비장세포를 확보하고 이를 107의 SP 2/0-Ag14 마우스 골수종세포(myeloma cell)와 폴리에틸렌글리콜(PEG 4000; Sigma, St Louis, MO) 방법을 사용하여 융합하였다. 상기의 방법은 Kohler G, Milstein C. Nature 256 (1975) 495-497 에 자세히 기술되었다. 획득한 100개의 하이브리도마 세포주의 배양 상층액을 사용하여 FACS 염색을 수행, B cell tumor cells (IM9, Raju, Ramos)에 반응성이 있는 하이브리도마 세포를 획득하였다. PBMC 1X10 7 isolated from Ficoll-gradient method from healthy volunteers was stimulated with Con A (concanavalin A) 10 ug / ml and immunized with mouse. After 2 months, spleens of mice were isolated and 1X10 8 splenocytes were isolated. This was obtained and fused with 10 7 SP 2 / 0-Ag14 mouse myeloma cells using polyethylene glycol (PEG 4000; Sigma, St Louis, MO) method. The method is described in detail in Kohler G, Milstein C. Nature 256 (1975) 495-497. FACS staining was performed using the obtained culture supernatant of 100 hybridoma cell lines to obtain hybridoma cells reactive to B cell tumor cells (IM9, Raju, Ramos).
2. 하이브리도마 세포주로부터 scFv(single chain Fv)인 ITTab1 항체 제작 및 활성 검증 2. Fabrication and activity validation of scFv (single chain Fv) ITTab1 antibody from hybridoma cell line
본 발명의 ITT-ab1(Inje Tumor Targeting - antibody1) 마우스항체는, 라이브러리(library) 제작단계(A)와; 항체기능확인단계(B)를 포함하는 것을 특징으로 하여 제조되었다. ITT-ab1 (Inje Tumor Targeting-antibody1) mouse antibody of the present invention, the library (library) production step (A); It was prepared, characterized in that it comprises an antibody function confirmation step (B).
또한 라이브러리(library)제작단계(A)는, B cell tumor cells(IM9) 에 결합하는 항체를 생산의 방법 1에서 획득한 하이브리도마 세포(이하 하이브리도마 F6 세포)로부터 scFv(단쇄 Fv) 형태의 비면역 마우스 항체 분절 라이브러리(library)를 제작하는 단계로서, In the library production step (A), scFv (single-chain Fv) forms from hybridoma cells (hereinafter, hybridoma F6 cells) obtained in the method 1 of production of antibodies that bind to B cell tumor cells (IM9). To prepare a non-immune mouse antibody fragment library of
RNA를 정제하는 분리정제단계(a1)와; First strand cDNA 제작 및 마우스항체 유전자의 중합효소 연쇄반응으로 증폭시키는 중합효소 연쇄반응 증폭단계(a2)와; 컴피턴트 셀(competent cell)을 제작하는 셀 제작단계(a3)와; 상기 제작단계를 바탕으로 scFv(single chain Fv) 라이브러리를 완성하는 라이브러리(library) 완성단계(a4)로 구성된다. 각 단계별로 과정을 설명하면 아래와 같다. Separation and purification step (a1) for purifying RNA; A polymerase chain reaction amplification step (a2) for amplifying the first strand cDNA by a polymerase chain reaction of a mouse antibody gene; A cell fabrication step (a3) of fabricating a competent cell; It is composed of a library completion step (a4) for completing a single chain Fv (scFv) library based on the production step. The following describes the process for each step.
1) 분리정제단계(a1)1) Separation and purification step (a1)
분리정제단계(a1)는, RNA를 정제하는 것으로서, 하이브리도마 F6 세포를 배양하여 TRIZOL 시약을 사용하여 F6 cell 1x107에 TRIZOL 2ml을 넣어 상온에서 5분간 반응한 뒤, TRIZOL 1ml 당 클로로포름 200ul 씩 15초 가량 vortex해주었다. 그리고 원심분리기를 이용하여 4℃에서 13000rmp에 15분간 원심분리 하였다. 그리고 상층액을 중간층을 따지 않게 조심하여 마이크로센트리퓨트 튜브(microcentrifuge tube)에 옮겨 이소프로필 알콜을 같은 비율로 섞어 상온에서 10분간 반응시켰다. 이후 4℃에서 12000 x g 로 10분간 원심분리하여 상층액을 제거한 후, 75% 에탄올 1ml을 넣어 세척해주고 한번 더 4℃에서 12000 x g로 15분 동안 원심분리하고 상층액을 제거한 후 DEPC 처리된 증류수 30ul로 RNA를 회수하였다. RNA는 0.8% 아가로스 겔에서 전기영동 하여 RNA 순도를 확인하였고, 분광흡광계를 이용하여 정량한 후 실험에 사용하였다.Separation and purification step (a1) is to purify RNA, cultivate hybridoma F6 cells, add TRIZOL 2ml to F6 cell 1x10 7 using TRIZOL reagent for 5 minutes at room temperature, and then 200ul of chloroform per 1ml TRIZOL. I vortex for about 15 seconds. And centrifuged for 15 minutes at 13000rpm at 4 ℃ using a centrifuge. Then, the supernatant was carefully transferred to a microcentrifuge tube so as not to pick the middle layer, and isopropyl alcohol was mixed in the same ratio and reacted at room temperature for 10 minutes. After removing the supernatant by centrifuging at 12000 xg for 10 minutes at 4 ℃, washed with 1ml of 75% ethanol once more centrifuged at 12000 xg for 15 minutes at 4 ℃ and remove the supernatant 30 DEL treated distilled water 30ul RNA was recovered. RNA was electrophoresed on 0.8% agarose gel to confirm RNA purity, which was quantified using a spectrophotometer and used for experiments.
2) 중합효소 연쇄반응 증폭단계(a2)2) polymerase chain reaction amplification step (a2)
중합효소 연쇄반응증폭단계(a2)는, First strand cDNA 제작 및 마우스항체 유전자를 중합효소 연쇄반응 증폭시키는 단계로서, 분리정제단계(a1)에서 정제된 RNA 2 ug을 0.5 ug의 올리고(dT)과 혼합하고 증류수를 첨가하여 최종 부피가 10 ul가 되도록 한 다음 75℃에서 5분간 반응시켰다. 이후 cDNA 반응용액 15 ul (5 x M-MLV 반응용액 5 ul, M-MLV 역전사효소 100unit, 10 mM dNTP 1.25 ul, RNase Inhibitor 4unit)를 첨가하여 RNA 샘플과 섞어 vortex 하였다. 이를 연쇄중합반응 장치에서 37℃에서 1시간, 95℃에서 5분간 반응시킨 후 mGAPDH 프라이머로 항체유전자를 중합효소 연쇄반응 증폭하였다. Polymerase chain reaction amplification step (a2) is a step of preparing the first strand cDNA and amplifying the polymerase chain reaction of the mouse antibody gene, and the purified RNA 2 ug in the purification step (a1) and 0.5 ug oligo (dT) and After mixing and adding distilled water to a final volume of 10 ul and reacted for 5 minutes at 75 ℃. Thereafter, 15 ul of cDNA reaction solution (5 ul of 5 x M-MLV reaction solution, 100 units of M-MLV reverse transcriptase, 10 mM dNTP 1.25 ul, and RNase Inhibitor 4 units) were added and vortex mixed with an RNA sample. The reaction was carried out in a chain polymerization apparatus at 37 ° C. for 1 hour and at 95 ° C. for 5 minutes to amplify the polymerase chain reaction with an mGAPDH primer.
마우스 항체 특이 프라이머로 항체유전자를 중합효소 연쇄반응 증폭하였다. 경쇄(light chain)와 중쇄(heavy chain)의 가변영역(variable region)들을 각각 증폭하고 이를 링커 특이 프라이머를 이용한 오버래핑 중합효소 연쇄반응으로 결합한 뒤 pComb3X-TT 발현벡터의 SfiⅠ 제한효소 사이트에 클로닝하여 라이브러리를 제작하였다. 이때 사용되는 프라이머는 매뉴얼 (Barbas et al., Phage Display; A Laboratory Manual, Cold Spring Harbor Laboratory,(2001), New York )에 의해 결정하였고 Bioneer(Korea)에 제작 의뢰하여 합성하였다.The antibody gene was amplified by polymerase chain reaction with a mouse antibody specific primer. Amplify the variable regions of the light and heavy chains, bind them with overlapping polymerase chain reaction using linker-specific primers, and clone them into the Sfi I restriction site of the pComb3X-TT expression vector. Was produced. The primer used at this time was determined by a manual (Barbas et al., Phage Display; A Laboratory Manual, Cold Spring Harbor Laboratory, (2001), New York), and was synthesized by making a request to Bioneer (Korea).
중합효소 연쇄반응 조건은 94℃에서 5분간 pre-denaturation하고 94℃ 30초, 57℃ 15초, 72℃ 30초로 25 cycles 반응한 뒤 72℃에서 10분간 반응하였다. 증폭의 유무는 1% 아가로스 겔 전기영동으로 확인하고 Qiagen 겔 추출 킷트를 이용하여 정제하고 상기 증폭된 경쇄가변부위와 중쇄가변부위를 주형으로 하고 다음 오버래핑 중합효소 연쇄반응을 실시하였다. Polymerase chain reaction conditions were pre-denaturated at 94 ° C. for 5 minutes, followed by 25 cycles of reaction at 94 ° C. 30 sec, 57 ° C. 15 sec, and 72 ° C. 30 sec. The presence or absence of amplification was confirmed by 1% agarose gel electrophoresis, purified using Qiagen gel extraction kit, and the amplified light chain variable region and heavy chain variable region were subjected to the following overlapping polymerase chain reaction.
중합효소 연쇄반응 산물은 Qiagen 겔 추출 킷트로 아가로스에서 DNA를 정제하고 SfiI로 제한효소 절단을 한 다음 다시 겔 추출(gel extraction)을 하였다. SfiI 절단된 항체 유전자 30 ng과 SfiI 절단된 pComb3X-TT 30 ng을 혼합한 후 10X 버퍼와 리가아제(ligase, Gibco-BRL, USA) 10 unit를 첨가하고 실온에서 2시간동안 반응한 후 TOP10F’ competent cell에 형질변환하였다. The polymerase chain reaction product was purified by DNA from agarose with Qiagen gel extraction kit, restriction enzyme digestion with Sfi I, and then gel extraction. Sfi I was added to a mixture of the cut antibody gene 30 ng and Sfi I cleavage pComb3X-TT 30 ng after 10X buffer and a ligase (ligase, Gibco-BRL, USA ) 10 unit and the reaction at room temperature for 2 hours after TOP10F Transformed competent cells.
본 발명의 제조합 인간 단세포군 항체 개발에 사용된 마우스 IgG 특이 프라이머는 Phage Display 메뉴얼(Barbas et al., Phage Display; A Laboratory Manual, Cold Spring Harbor Laboratory,(2001), New York)에 상세하게 기술되어 있다.Mouse IgG specific primers used in the development of the synthetic human unicellular antibody of the present invention are described in detail in the Phage Display manual (Barbas et al., Phage Display; A Laboratory Manual, Cold Spring Harbor Laboratory, (2001), New York). It is.
3) 셀 제작단계(a3)3) Cell manufacturing step (a3)
셀제작단계(a3)는, 컴피턴트 셀(Electrocompetent cell)을 제작하는 단계로서, 대장균 ER2537을 미니멀 플레이트(minimal plate)에서 배양하여 F' 섬모의 발현을 유도한 후 수용성 세포 (competent cell) 제작에 사용하였다. SB 배지(10 g MOPS, 30 g, bacto-trypton, 20 g yeast extract in 1 L) 2 ㎖에 ER2537을 접종하여 37℃에서 18시간동안 배양한 후, O.D600가 3.54 되도록 배양하였다. 전 배양액 1 ㎖를 LB 배지(10 g NaCl, 10 g, bacto-trypton, 5 g yeast extract in 1 L) 100ml에 O.D600가 0.4가 되도록 배양하였다. 그후 원심분리기를 통해 4℃에서 4,000 rpm으로 10분간 원침하였다. 50mM CaCl2 20ml로 세포를 부유시킨 다음 아이스에서 30분간 반응 후 4℃ 에서 4,000 rpm으로 10분간 원침하였다. 상층액을 제거하고 (10% 글리세롤 + 50mM CaCl2) 3ml을 넣어 세포를 부유한 후 50ul씩 분주하여 사용하였다. Cell production step (a3) is a step of producing a competent cell (Electrocompetent cell), incubation of E. coli ER2537 in a minimal plate (minimal plate) to induce the expression of the F 'cilia after the production of water-soluble cells (competent cell) Used. ER2537 was inoculated in 2 ml of SB medium (10 g MOPS, 30 g, bacto-trypton, 20 g yeast extract in 1 L) and incubated at 37 ° C. for 18 hours, followed by OD 600 to 3.54. 1 ml of the pre-culture was incubated in 100 ml of LB medium (10 g NaCl, 10 g, bacto-trypton, 5 g yeast extract in 1 L) so that the OD 600 was 0.4. Thereafter, centrifuge was performed for 10 minutes at 4,000 rpm at 4 ℃. The cells were suspended with 20 ml of 50 mM CaCl 2 and then reacted for 30 minutes on ice, and then centrifuged at 4,000 rpm for 10 minutes at 4 ° C. The supernatant was removed and 3 ml of (10% glycerol + 50 mM CaCl 2 ) was added to the cells, followed by aliquoting of 50ul.
4) 라이브러리 완성단계(a4)4) Library Completion Step (a4)
라이브러리(library)완성단계(a4)는, 상기 제작단계를 바탕으로 scFv(단쇄 Fv) 라이브러리를 완성하는 단계로서, 회수된 파지 항체를 대수 증식기의 대장균 ER2537에 감염시킨 후 100 μg/mL의 암피실린이 첨가된 LB 아가 플레이트에 도말하여 37℃ 에서 18시간 배양하였다. 형성된 집락을 100 μg/mL의 암피실린이 첨가된 SB (SB/amp) 액체배지 5 mL에 접종하여 37℃에서 250 rpm으로 4시간 진탕배양 하였다. 파지 형성을 위하여 VCSM13을 1010 pfu 첨가하여 37℃ 에서 30분간 정치하여 감염시킨 후 37℃ 에서 250 rpm으로 1시간 30분 배양하고 카나마이신을 최종 70 μg/mL이 되게 첨가하여 37℃ 에서 18시간동안 배양하였다. 이후 배양액을 2,500 rpm 10분간 원침시켜 파지가 포함된 상층액을 회수하였다. The library completion step (a4) is a step of completing the scFv (single chain Fv) library based on the preparation step, and 100 μg / mL of ampicillin after infecting the recovered phage antibody with Escherichia coli ER2537. Plates were added to the added LB agar plates and incubated at 37 ° C for 18 hours. The colonies formed were inoculated into 5 mL of SB (SB / amp) liquid medium to which 100 μg / mL of ampicillin was added and shaken at 37 ° C. at 250 rpm for 4 hours. For phage formation, 10 10 pfu of VCSM13 was added and allowed to stand at 37 ° C for 30 minutes for infection, followed by incubation at 250 rpm at 37 ° C for 1 hour 30 minutes, and kanamycin was added to 70 μg / mL for 18 hours at 37 ° C. Incubated. Thereafter, the culture solution was centrifuged at 2,500 rpm for 10 minutes to recover the supernatant containing phage.
이후 항체기능확인단계(B)는, 라이브러리(library)완성단계(a4)에서 완성된 항체들의 기능을 확인하는 것으로 IM-9 세포를 인지하는 파지 항체를 α-M13과 α-mIgG-PE로 처리한후 FACS 염색을 수행하여, B cell tumor cells인 IM9에 결합하는지 여부를 확인하였다. Subsequently, the antibody function checking step (B) confirms the function of the antibodies completed in the library completion step (a4), and treats phage antibodies that recognize IM-9 cells with α-M13 and α-mIgG-PE. After FACS staining was confirmed whether the binding to IM9 B cell tumor cells.
<실시예 1> ITTab1의 B cell tumor cells (IM9) 결합능 검토 Example 1 Examination of the binding capacity of ITTab1 B cell tumor cells (IM9)
파지 디스플레이에 의해 확보된 ITTab1이 기 확보된 하이브리도마에서 유래된 항체와 같은 특징을 보유하는지 규명하기 위해 B cell tumor cells 인 IM9 세포주에 대한 결합능을 FACS staining으로 확인하였다. IM-9 세포 (106)에 1 ug의 각각 ITTab1 서브클론 항체(ITTab 1-1 내지 ITTab 1-4)를 처리하고 30분 결합 후 PBS로 세척하였다. 세척된 세포는 FITC-conjugated goat anti-mouse IgG, (Sigma-Aldrich)를 30분간 결합한후 3번 PBS로 세척하고 FACS Calibur (BD Pharmingen)로 관찰하였다. 결과를 도 1에 타내었다. In order to determine whether the ITTab1 obtained by phage display has the same characteristics as the antibody derived from the secured hybridoma, FACS staining was confirmed for binding to IM9 cell line, B cell tumor cells. IM-9 cells (10 6 ) were treated with 1 ug of ITTab1 subclonal antibodies (ITTab 1-1 to ITTab 1-4), respectively, and washed with PBS after 30 min binding. The washed cells were combined with FITC-conjugated goat anti-mouse IgG, (Sigma-Aldrich) for 30 minutes, washed with PBS three times, and observed with FACS Calibur (BD Pharmingen). The results are shown in FIG.
도 1을 참조하면, IM-9 세포의 표면에서 ITTab1의 서브클론 항체 모두가 결합하는 것이 관찰 되었다. 이러한 결과를 통해, 이 항체에 대한 타겟 항원이 IM-9 세포에 존재하며, 기 확보된 하이브리도마 세포에서 얻은 항체와 파지 디스플레이에 의해 확보된 본 발명의 ITTab1 항체가 동일한 항원을 인지하는 것을 확인할 수 있었다. Referring to FIG. 1, it was observed that all of the subclonal antibodies of ITTab1 bind to the surface of IM-9 cells. These results confirm that the target antigen for this antibody is present in IM-9 cells, and that the antibody obtained from previously obtained hybridoma cells and the ITTab1 antibody of the present invention obtained by phage display recognize the same antigen. Could.
<실시예 2> ITTab1의 항체 CDR 영역의 서열 규명 및 전체 핵산 및 아미노산 서열 규명Example 2 Sequence Identification and Total Nucleic Acid and Amino Acid Sequence of the Antibody CDR Region of ITTab1
파지 디스플레이에 의해 확보된 ITTab1의 서브클론인 ITTab1-1 내지 ITTab1-4의 CDR 영역의 서열을 규명하기 위해 시퀀싱을 진행하고 확보된 정보를 사용하여 특징을 관찰하였다. scFv (단쇄 Fv) 형의 ITTab1-1 내지 ITTab1-4의 가변영역 내 상보성 결정부위(Complimentarily-determining region, CDR)를 규명한 결과, 밑줄의 부위는 링커 부위 이고 이를 경계로 경쇄와 중쇄로 나뉘며 중쇄의 제일 선두에 나오는 굵은 글시체로 표시된 부위부터 중쇄의 CDR1, CDR2 그리고 CDR3로 결정되었다. 링커를 경계로 경쇄의 선두에 나오는 굵은 글씨체로 표시된 부위는 경쇄의 CDR1, CDR2 그리고 CDR3로 결정되었다.Sequencing was performed to identify sequences of CDR regions of ITTab1-1 to ITTab1-4, which are subclones of ITTab1 obtained by phage display, and the characteristics were observed using the obtained information. Complimentarily-determining region (CDR) in the variable region of ITTab1-1 to ITTab1-4 of scFv (short chain Fv) type was identified.The underlined portion is a linker region, which is divided into a light chain and a heavy chain, and the heavy chain. From the sites marked in boldface at the top of the chain, CDR1, CDR2 and CDR3 of the heavy chain were determined. The sites shown in bold at the front of the light chain bordering the linker were determined by CDR1, CDR2 and CDR3 of the light chain.
1) ITTab1-11) ITTab1-1
도 2에 ITTab1-1의 염기 및 아미노산 서열 및 이의 CDR 및 링커 영역을 나타내었다. Figure 2 shows the base and amino acid sequence of ITTab1-1 and its CDR and linker regions.
도 2를 참조하면, ITTab1-1는 굵은 글씨체의 순서대로 경쇄의 CDR1(서열번호 1), CDR2(서열번호 2), CDR3(서열번호 3), 중쇄의 CDR1(서열번호 4), CDR2(서열번호 5), CDR3(서열번호 6)을 포함한다. Referring to Figure 2, ITTab1-1 is a light chain CDR1 (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2), CDR3 (SEQ ID NO: 3), heavy chain CDR1 (SEQ ID NO: 4), CDR2 (SEQ ID NO: 1) No. 5), CDR3 (SEQ ID NO: 6).
또한, ITTab1-1의 경쇄 아미노산 서열을 서열번호 7에, 중쇄 아미노산서열을 서열번호 8에 나타내었고,  In addition, the light chain amino acid sequence of ITTab1-1 is shown in SEQ ID NO: 7, and the heavy chain amino acid sequence of SEQ ID NO: 8,
ITTab1-1의 전체 아미노산 서열을 서열번호 9에, 전체 염기서열을 서열번호 10 에 나타내었다. The entire amino acid sequence of ITTab1-1 is shown in SEQ ID NO: 9, and the entire nucleotide sequence is shown in SEQ ID NO: 10.
2) ITTab1-22) ITTab1-2
도 3에 ITTab1-2의 염기 및 아미노산 서열 및 이의 CDR 및 링커 영역을 나타내었다.3 shows the base and amino acid sequence of ITTab1-2 and its CDR and linker regions.
도 3를 참조하면, ITTab1-2는 굵은 글씨체의 순서대로 경쇄의 CDR1(서열번호 1), CDR2(서열번호 2), CDR3(서열번호 3), 중쇄의 CDR1(서열번호 4), CDR2(서열번호 5), CDR3(서열번호 6)을 포함한다. Referring to Figure 3, ITTab1-2 is a light chain CDR1 (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2), CDR3 (SEQ ID NO: 3), heavy chain CDR1 (SEQ ID NO: 4), CDR2 (SEQ ID NO: 1) No. 5), CDR3 (SEQ ID NO: 6).
또한, ITTab1-2의 경쇄 아미노산 서열을 서열번호 11에, 중쇄 아미노산서열을 서열번호 12에 나타내었고,  In addition, the light chain amino acid sequence of ITTab1-2 is shown in SEQ ID NO: 11, and the heavy chain amino acid sequence of SEQ ID NO: 12,
ITTab1-2의 전체 아미노산 서열을 서열번호 13에, 전체 염기서열을 서열번호 14 에 나타내었다. The entire amino acid sequence of ITTab1-2 is shown in SEQ ID NO: 13, and the entire nucleotide sequence is shown in SEQ ID NO: 14.
3) ITTab1-33) ITTab1-3
도 4에 ITTab1-3의 염기 및 아미노산 서열 및 이의 CDR 및 링커 영역을 나타내었다. 4 shows the base and amino acid sequence of ITTab1-3 and its CDR and linker regions.
도 4를 참조하면, ITTab1-3은 굵은 글씨체의 순서대로 경쇄의 CDR1(서열번호 1), CDR2(서열번호 2), CDR3(서열번호 3), 중쇄의 CDR1(서열번호 4), CDR2(서열번호 5), CDR3(서열번호 6)을 포함한다. Referring to Figure 4, ITTab1-3 is a light chain CDR1 (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2), CDR3 (SEQ ID NO: 3), heavy chain CDR1 (SEQ ID NO: 4), CDR2 (SEQ ID NO: 1) No. 5), CDR3 (SEQ ID NO: 6).
또한, ITTab1-3의 경쇄 아미노산 서열을 서열번호 15에, 중쇄 아미노산서열을 서열번호 16에 나타내었고,  In addition, the light chain amino acid sequence of ITTab1-3 is shown in SEQ ID NO: 15, and the heavy chain amino acid sequence of SEQ ID NO: 16,
ITTab1-3의 전체 아미노산 서열을 서열번호 17에, 전체 염기서열을 서열번호 18 에 나타내었다. The entire amino acid sequence of ITTab1-3 is shown in SEQ ID NO: 17, and the entire nucleotide sequence is shown in SEQ ID NO: 18.
4) ITTab1-44) ITTab1-4
도 5에 ITTab1-4의 염기 및 아미노산 서열 및 이의 CDR 및 링커 영역을 나타내었다.5 shows the base and amino acid sequences of ITTab1-4 and their CDRs and linker regions.
도 5를 참조하면, ITTab1-4는 굵은 글씨체의 순서대로 경쇄의 CDR1(서열번호 1), CDR2(서열번호 2), CDR3(서열번호 3), 중쇄의 CDR1(서열번호 4), CDR2(서열번호 5), CDR3(서열번호 6)을 포함한다. Referring to Figure 5, ITTab1-4 is in the order of bold font CDR1 (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2), CDR3 (SEQ ID NO: 3), CDR1 (SEQ ID NO: 4), CDR2 (SEQ ID NO: 4) of the heavy chain No. 5), CDR3 (SEQ ID NO: 6).
또한, ITTab1-4의 경쇄 아미노산 서열을 서열번호 19에, 중쇄 아미노산서열을 서열번호 20에 나타내었고,  In addition, the light chain amino acid sequence of ITTab1-4 is shown in SEQ ID NO: 19, and the heavy chain amino acid sequence of SEQ ID NO: 20,
ITTab1-4의 전체 아미노산 서열을 서열번호 21에, 전체 염기서열을 서열번호 22 에 나타내었다. The entire amino acid sequence of ITTab1-4 is shown in SEQ ID NO: 21, and the entire nucleotide sequence is shown in SEQ ID NO: 22.
<실시예 3> NK 림프종세포에 대한 ITTab1의 항원 발현 검출능 규명Example 3 Identification of Antigen Expression of ITTab1 on NK Lymphoma Cells
ITTab1의 항원에 대한 반응성과 종양세포에 대한 발현 패턴을 확인하기 위해, NK 림프종 세포에 대해 ITTab1의 항원이 존재하는지 여부를 면역형광법 및 공초점현미경으로 분석하였다.In order to confirm the reactivity with the antigen of ITTab1 and the expression pattern for tumor cells, the presence of the antigen of ITTab1 on NK lymphoma cells was analyzed by immunofluorescence and confocal microscopy.
NK 림프종 세포를 1ug의 ITTab1과 함께 30분간 배양 한 후 세포를 회수하여 PBS를 사용하여 세척하였다. 세척한 세포주를 FITC-conjugated goat anti-mouse IgG(Sigma-Aldrich)와 함께 30분간 배양한 후 PBS 로 3번 세척하였다. 세척된 세포는 형광 마운팅 배지 (DakoCytomation, Glostrup, Denmark)를 사용하여 커버-슬립 밑에 현미경 슬라이드 위에 놓고 마운팅하였다. 형광 염색된 세포는 Confocal Laser-Scanning microscope (Carl ZEISS, 510 META, Jena, Germany)를 사용하여 400× original magnification으로 관찰하고 confocal Microscopy Software Release 3.0 (Carl ZEISS, 510 META)로 분석하였다. After NK lymphoma cells were incubated with 1 ug of ITTab1 for 30 minutes, the cells were recovered and washed with PBS. The washed cell lines were incubated with FITC-conjugated goat anti-mouse IgG (Sigma-Aldrich) for 30 minutes and washed three times with PBS. The washed cells were placed on a microscope slide under the cover-slip using fluorescent mounting medium (DakoCytomation, Glostrup, Denmark). Fluorescent stained cells were observed with 400 × original magnification using a Confocal Laser-Scanning microscope (Carl ZEISS, 510 META, Jena, Germany) and analyzed by confocal Microscopy Software Release 3.0 (Carl ZEISS, 510 META).
결과를 도 6에 나타내었다. 도 6을 참조하면, 공초점현미경 결과에서 세포표면과 세포질 모두에서 ITTab1에 의해 초록색 형광으로 염색되는 결과를 확인할 수 있는 바, 이를 통해 ITTab1 항체에 대한 타겟 항원이 NK 림프종 세포에 존재함을 확인할 수 있었다. The results are shown in FIG. Referring to FIG. 6, it can be seen from the confocal microscopy results that the cell surface and the cytoplasm are stained with green fluorescence by ITTab1. Through this, the target antigen for the ITTab1 antibody is present in NK lymphoma cells. there was.
<실시예 4> ITTab1이 NK 림프종 세포의 세포자멸사에 미치는 영향 검토 Example 4 Examining the Effect of ITTab1 on Apoptosis of NK Lymphoma Cells
NK 림프종 세포에 발현되는 ITTab1 항원의 NK 림프종 세포에 대한 기능을 규명하기 위하여 먼저 ITTab1을 종양 세포에 처리 후 자멸사에 미치는 효과를 아넥신(annexin)-V 어세이로 분석하였다. To clarify the function of ITTab1 antigen expressed on NK lymphoma cells on NK lymphoma cells, the effect of ITTab1 on tumor cells after treatment with tumor cells was analyzed by an annexin-V assay.
NK 림프종 세포 (106 cells)를 각각의 농도의 ITTab1과 함께 1시간 배양 한 후 세포를 회수하여 PBS를 사용하여 두 번 세척하였다. 세척한 세포주를 100 ul의 아넥신-V 결합 버퍼 (10mM EPES/NaOH pH 7.4, 140mM NaCl, 2.5mM CaCl2)로 현탁한후 2ul의 FITC-컨쥬게이트 아넥신-V (BD Pharmingen)와 7-ADD를 처리하였다. 처리된 NK 림프종 세포를 어두운 상태의 실온에서 15분간 배양한후 400 ul의 아넥신-V 결합 버퍼를 더 첨가 해준 후 FACS Calibur (BD Pharmingen)를 사용하여 분석하였다. NK lymphoma cells (10 6 cells) were incubated with each concentration of ITTab1 for 1 hour, and then the cells were recovered and washed twice with PBS. The washed cell lines were suspended with 100 ul of Annexin-V binding buffer (10 mM EPES / NaOH pH 7.4, 140 mM NaCl, 2.5 mM CaCl 2 ), followed by 2ul of FITC-conjugate Annexin-V (BD Pharmingen) and 7- ADD was processed. Treated NK lymphoma cells were incubated for 15 minutes at room temperature in the dark, followed by the addition of 400 ul of Annexin-V binding buffer and analyzed using FACS Calibur (BD Pharmingen).
결과를 도 7에 나타내었다. 도 7을 참조하면, ITTab1항체가 증가되는 농도 의존적으로 세포의 자멸사가 유도되는 것을 관찰하였으며 이를 통해, 본 발명의 ITTab1가 NK 림프종 세포에서 세포자멸사 유도 항체로 발달될 수 있음을 시사한다. The results are shown in FIG. Referring to FIG. 7, it was observed that apoptosis was induced in a concentration-dependent manner in which the ITTab1 antibody was increased, thereby suggesting that ITTab1 of the present invention can be developed as an apoptosis-inducing antibody in NK lymphoma cells.
<실시예 5> 난소암 세포에 대한 ITTab1의 항원 발현 검출능 규명Example 5 Identification of Antigen Expression of ITTab1 on Ovarian Cancer Cells
ITTab1의 항원에 대한 반응성과 종양세포에 대한 발현 패턴을 확인하기 위해 난소암 세포에 대해 ITTab1의 항원이 존재하는지를 항체로 염색하고 FACS calibur(BD Pharmingen)로 관찰하였다.In order to confirm the reactivity with the antigen of ITTab1 and the expression pattern for tumor cells, the presence of ITTab1 antigen in ovarian cancer cells was stained with an antibody and observed with FACS calibur (BD Pharmingen).
난소암 세포는 1ug의 ITTab1과 함께 30분간 배양 한 후 세포를 회수하여 PBS를 사용하여 세척되었다. 세척한 세포주를 FITC-conjugated goat anti-mouse IgG(Sigma-Aldrich)와 함께 30분간 배양한 후 PBS 로 3번 세척하였다. 세척된 세포는 FACS calibur(BD Pharmingen)를 사용하여 분석하였다. Ovarian cancer cells were incubated with 1 ug of ITTab1 for 30 minutes and then cells were recovered and washed with PBS. The washed cell lines were incubated with FITC-conjugated goat anti-mouse IgG (Sigma-Aldrich) for 30 minutes and washed three times with PBS. Washed cells were analyzed using FACS calibur (BD Pharmingen).
결과를 도 8에 나타내었다. 도 8을 참조하면, 유세포분석 결과에서 세포표면과 세포질 모두에서 ITTab1에 의해 ITTab1의 항원이 발현되는 것을 확인할 수 있었으며, 상기 결과를 통해 본 발명의 항체에 대한 타겟 항원이 난소암 세포에 존재함을 알 수 있었다. The results are shown in FIG. Referring to FIG. 8, in the flow cytometry results, it was confirmed that the antigen of ITTab1 was expressed by ITTab1 on both the cell surface and the cytoplasm, and the target antigen for the antibody of the present invention was present in ovarian cancer cells. Could know.
<실시예 6> ITTab1이 난소암 세포의 세포자멸사에 미치는 영향 검토 Example 6 Examination of the Effect of ITTab1 on Apoptosis of Ovarian Cancer Cells
난소암 세포에 발현되는 ITTab1 항원의 난소암 세포에 대한 기능을 규명하기 위하여 먼저 ITTab1을 종양 세포에 처리 후 자멸사에 미치는 효과를 프로피듐 아이오드(PI)처리에 의한 세포 주기 분석으로 분석하였다. To investigate the function of the ITTab1 antigen expressed in ovarian cancer cells on ovarian cancer cells, the effect of ITTab1 on tumor cells after treatment with tumor cells was analyzed by cell cycle analysis by propidium iodide (PI) treatment.
난소암 세포(106 cells)는 5ug의 ITTab1과 함께 1시간 배양 한 후 세포를 회수하여 PBS를 사용하여 두 번 세척되었다. 세척한 세포주를 프로피듐 아이오드(PI)로 처리 한 후 세포를 FACS caliber (BD Pharmingen)를 사용하여 분석하였다. Ovarian cancer cells (10 6 cells) were incubated with 5ug of ITTab1 for 1 hour and then recovered and washed twice with PBS. The washed cell lines were treated with propidium iodine (PI) and the cells were analyzed using FACS caliber (BD Pharmingen).
결과를 도 9에 나타내었다. 난소암 세포에 ITTab1를 처리하게 되면 PI염색 결과 subG1 기에 해당하는 apoptosis 세포가 isotype control (MOPC) 처리한 세포에서 보다 항체의 농도가 증가할수록 증가되는 양상을 보여주었다. 이를 통해, 본 발명의 ITTab1가 난소암 세포에서 세포자멸사 유도 항체로 사용될 수 있음을 알 수 있었다. The results are shown in FIG. Treatment of ovarian cancer cells with ITTab1 showed that PI apoptosis cells in the subG1 phase increased with increasing antibody concentrations compared to those in isotype control (MOPC) cells. Through this, it can be seen that ITTab1 of the present invention can be used as an apoptosis-inducing antibody in ovarian cancer cells.
이상으로 본 발명의 특정한 부분을 상세히 기술하였는 바, 당업계의 통상의 지식을 가진 자에게 있어서, 이러한 구체적 기술은 단지 바람직한 실시예일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백할 것이다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다고 할 것이다. Having described the specific part of the present invention in detail, it will be apparent to those skilled in the art that such a specific description is only a preferred embodiment, thereby not limiting the scope of the present invention. will be. Thus, the substantial scope of the present invention will be defined by the appended claims and their equivalents.

Claims (17)

  1. 암세포에 결합하여 세포사멸을 유도하는 단일클론 항체로서, A monoclonal antibody that binds to cancer cells and induces apoptosis,
    서열번호 1의 아미노산 서열로 이루어진 경쇄 CDR1, 서열번호 2의 아미노산 서열로 이루어진 경쇄 CDR2 및 서열번호 3의 아미노산 서열로 이루어진 경쇄 CDR3 를 포함하는 경쇄 가변영역 및;A light chain variable region comprising a light chain CDR1 consisting of an amino acid sequence of SEQ ID NO: 1, a light chain CDR2 consisting of an amino acid sequence of SEQ ID NO: 2, and a light chain CDR3 consisting of an amino acid sequence of SEQ ID NO: 3;
    서열번호 4의 아미노산 서열로 이루어진 중쇄 CDR1, 서열번호 5의 아미노산 서열로 이루어진 중쇄 CDR2 및 서열번호 6의 아미노산 서열로 이루어진 중쇄 CDR3 를 포함하는 중쇄 가변영역을 포함하는 항체.An antibody comprising a heavy chain variable region comprising a heavy chain CDR1 consisting of an amino acid sequence of SEQ ID NO: 4, a heavy chain CDR2 consisting of an amino acid sequence of SEQ ID NO: 5, and a heavy chain CDR3 consisting of an amino acid sequence of SEQ ID NO: 6.
  2. 제1항에 있어서, The method of claim 1,
    상기 경쇄 가변영역은 The light chain variable region is
    서열번호 7, 서열번호 11, 서열번호 15 또는 서열번호 19의 아미노산 서열을 갖는 것인 항체.The antibody having the amino acid sequence of SEQ ID NO: 7, SEQ ID NO: 11, SEQ ID NO: 15, or SEQ ID NO: 19.
  3. 제1항에 있어서, The method of claim 1,
    상기 중쇄 가변영역은The heavy chain variable region is
    서열번호 8, 서열번호 12, 서열번호 16 또는 서열번호 20의 아미노산 서열을 갖는 것인 항체.The antibody having the amino acid sequence of SEQ ID NO: 8, SEQ ID NO: 12, SEQ ID NO: 16, or SEQ ID NO: 20.
  4. 제1항에 있어서, The method of claim 1,
    상기 항체는 단일쇄 가변영역 (scFv, Single-chain variable fragment) 항체 이며, The antibody is a single-chain variable fragment (scFv) antibody,
    서열번호 9, 서열번호 13, 서열번호 17 또는 서열번호 21의 아미노산 서열을 갖는 것인 항체.The antibody having the amino acid sequence of SEQ ID NO: 9, SEQ ID NO: 13, SEQ ID NO: 17, or SEQ ID NO: 21.
  5. 서열번호 9의 아미노산 서열을 갖는 단일쇄 가변영역 (scFv, Single-chain variable fragment) 항체를 코딩하는 유전자.A gene encoding a single-chain variable fragment (scFv) antibody having the amino acid sequence of SEQ ID NO.
  6. 제5항에 있어서, The method of claim 5,
    상기 유전자는 서열번호 10의 염기서열로 이루어진 것인 유전자. The gene is one consisting of the nucleotide sequence of SEQ ID NO: 10.
  7. 서열번호 13의 아미노산 서열을 갖는 단일쇄 가변영역 (scFv, Single-chain variable fragment) 항체를 코딩하는 유전자. A gene encoding a single-chain variable fragment (scFv) antibody having the amino acid sequence of SEQ ID NO: 13.
  8. 제7항에 있어서, The method of claim 7, wherein
    상기 유전자는 서열번호 14의 염기서열로 이루어진 것인 유전자. The gene is one consisting of the nucleotide sequence of SEQ ID NO: 14.
  9. 서열번호 17의 아미노산 서열을 갖는 단일쇄 가변영역 (scFv, Single-chain variable fragment) 항체를 코딩하는 유전자. A gene encoding a single-chain variable fragment (scFv) antibody having the amino acid sequence of SEQ ID NO: 17.
  10. 제9항에 있어서, The method of claim 9,
    상기 유전자는 서열번호 18의 염기서열로 이루어진 것인 유전자. The gene is one consisting of the nucleotide sequence of SEQ ID NO: 18.
  11. 서열번호 21의 아미노산 서열을 갖는 단일쇄 가변영역 (scFv, Single-chain variable fragment) 항체를 코딩하는 유전자. A gene encoding a single-chain variable fragment (scFv) antibody having the amino acid sequence of SEQ ID NO: 21.
  12. 제11항에 있어서, The method of claim 11,
    상기 유전자는 서열번호 22의 염기서열로 이루어진 것인 유전자. The gene is one consisting of the nucleotide sequence of SEQ ID NO: 22.
  13. 제1항 내지 제4항 중 어느 한 항에 따른 항체를 유효성분으로 함유하는 암 예방 또는 치료용 약학조성물.A pharmaceutical composition for preventing or treating cancer containing the antibody according to any one of claims 1 to 4 as an active ingredient.
  14. 제13항에 있어서, The method of claim 13,
    상기 암은 간암, 폐암, 대장암, 난소암, 유방암, 전립선암, 백혈병, 림프종 및 췌장암으로부터 선택되는 어느 하나인 것인 암 예방 또는 치료용 약학조성물.The cancer is any one selected from liver cancer, lung cancer, colon cancer, ovarian cancer, breast cancer, prostate cancer, leukemia, lymphoma and pancreatic cancer pharmaceutical composition for cancer prevention or treatment.
  15. 제1항 내지 제4항 중 어느 한 항에 따른 항체를 포함하고, 상기 항체에 하나 또는 둘 이상의 진단제 또는 항암제를 결합시킨 암세포 표적화 진단 또는 치료 컨쥬게이트.A cancer cell targeting diagnostic or therapeutic conjugate comprising an antibody according to any one of claims 1 to 4, wherein one or more diagnostic or anticancer agents are bound to the antibody.
  16. 제15항에 있어서, The method of claim 15,
    상기 진단제는 방사성핵종, 상자성 금속이온, 초상자성 나노입자, 발색효소, 크로모포어, 발광물질 및 형광물질로 이루어진 군에서 선택된 어느 하나 또는 둘 이상인 것인 암세포 표적화 진단 또는 치료 컨쥬게이트.The diagnostic agent is any one or two or more selected from the group consisting of radionuclides, paramagnetic metal ions, superparamagnetic nanoparticles, chromophores, chromophores, luminescent materials and fluorescent materials, cancer cell targeting diagnostic or therapeutic conjugate.
  17. 제15항에 있어서, The method of claim 15,
    상기 항암제는 시스플라틴, 5-플로오우라실, 아드리아마이신, 메토트렉세이트, 빈블라스틴, 부설판, 클로람부실, 시클로포스파미드, 멜팔란, 니트로겐 무스타드, 니트로소우레아, 탁솔, 파클리탁셀, 도세탁셀, 6-머캅토퓨린, 6-티오구아닌, 블레오마이신, 다우노루비신, 독소루비신, 에피루비신, 이다루비신, 미토마이신-C 및 하이드록시우레아로 이루어진 군으로부터 선택되는 어느 하나 또는 둘 이상인 것인 암세포 표적화 진단 또는 치료 컨쥬게이트.The anticancer agent is cisplatin, 5-fluorouracil, adriamycin, methotrexate, vinblastine, busulfan, chlorambucil, cyclophosphamide, melphalan, nitrogen mustard, nitrosourea, taxol, paclitaxel, docetaxel, Cancer cell which is any one or more selected from the group consisting of 6-mercaptopurine, 6-thioguanine, bleomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin-C and hydroxyurea Targeted diagnostic or therapeutic conjugates.
PCT/KR2012/008951 2012-10-29 2012-10-29 Cancer-targeting antibody and composition for preventing or treating cancer comprising same WO2014069681A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/KR2012/008951 WO2014069681A1 (en) 2012-10-29 2012-10-29 Cancer-targeting antibody and composition for preventing or treating cancer comprising same

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/KR2012/008951 WO2014069681A1 (en) 2012-10-29 2012-10-29 Cancer-targeting antibody and composition for preventing or treating cancer comprising same

Publications (1)

Publication Number Publication Date
WO2014069681A1 true WO2014069681A1 (en) 2014-05-08

Family

ID=50627572

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2012/008951 WO2014069681A1 (en) 2012-10-29 2012-10-29 Cancer-targeting antibody and composition for preventing or treating cancer comprising same

Country Status (1)

Country Link
WO (1) WO2014069681A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007515165A (en) * 2003-12-04 2007-06-14 バクシネックス インコーポレーティッド Method of killing tumor cells by targeting intracellular antigens exposed on apoptotic tumor cells
KR20090078339A (en) * 2006-10-06 2009-07-17 다케다 야쿠힌 고교 가부시키가이샤 Prophylactic or therapeutic agent for cancer
US7691376B2 (en) * 1999-08-13 2010-04-06 Immunocellular Therapeutics, Ltd. Ovarian cancer cell and myeloma cell surface glycoproteins, antibodies thereto, and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7691376B2 (en) * 1999-08-13 2010-04-06 Immunocellular Therapeutics, Ltd. Ovarian cancer cell and myeloma cell surface glycoproteins, antibodies thereto, and uses thereof
JP2007515165A (en) * 2003-12-04 2007-06-14 バクシネックス インコーポレーティッド Method of killing tumor cells by targeting intracellular antigens exposed on apoptotic tumor cells
KR20090078339A (en) * 2006-10-06 2009-07-17 다케다 야쿠힌 고교 가부시키가이샤 Prophylactic or therapeutic agent for cancer

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HEO, DAE YOUNG ET AL.: "Effect of CM1 on the Proliferation and Apoptosis of Germinal Center B Cells", INJE MEDICAL, vol. 21, no. 2, 2002, pages 349 - 356 *
KIM, DAEJIN ET AL.: "CMl Ligation Initiates Apoptosis in a Caspase 8 Dependent Manner in Ramos Cells and in a Mitochondria Controlled manner in Raji Cells", HUMAN IMMUNOLOGY, vol. 63, 2002, pages 576 - 587 *

Similar Documents

Publication Publication Date Title
TWI589590B (en) Anti-notch3 antibodies and antibody-drug conjugates
WO2020076105A1 (en) Novel anti-c-kit antibody
JP7458399B2 (en) Anti-claudin antibodies and their use
RU2515904C2 (en) Method for generating active antibodies against resistance antigen, antibodies produced by same method and using them
US20070112179A1 (en) Novel compounds
KR20100097660A (en) Anti-mesothelin antibodies and uses therefor
BR112021012608A2 (en) BINDING PORTION THAT SPECIFICALLY BINDS TO CLAUDIN18.2, CHIMERIC ANTIGEN RECEPTOR, NUCLEIC ACID, MANIPULATED IMMUNE CELL, PHARMACEUTICAL COMPOSITION, AND, METHOD OF TREATMENT OF A TUMOR OR CANCER EXPRESSING CLAUDIN18.2
JP2022023078A (en) Antibody binding specifically to muc1 and use thereof
WO2013081379A1 (en) Drug conjugate comprising drug linked to human c-met antibody, and use therefor
EP3495390A1 (en) Novel antibody against programmed cell death protein (pd-1), and use thereof
TW202031687A (en) A fusion protein and uses thereof
KR101828794B1 (en) A antibody able to neutralize middle east respiratory syndrome coronavirus
KR20150129649A (en) Anti-bag3 antibodies for therapeutic use
JP6087289B2 (en) Anti-tumor peptide derived from complementarity determining region of humanized monoclonal antibody to NAPI2B transporter
CA2515389A1 (en) Monoclonal antibody and gene encoding the same, hybridoma, pharmaceutical compostion, and diagnostic reagent
WO2014069681A1 (en) Cancer-targeting antibody and composition for preventing or treating cancer comprising same
WO2014069682A1 (en) Cancer-targeting antibody and composition for preventing or treating cancer comprising same
KR101471253B1 (en) Anti-cancer Antibody and Composition for Preventing or Treating Cancer Comprising the Same
KR101428867B1 (en) Anti-cancer Antibody and Composition for Preventing or Treating Cancer Comprising the Same
WO2022239766A1 (en) Anti-cadm1 antibody
WO2023211068A1 (en) Pharmaceutical composition for cancer treatment, comprising anti-igsf1 antibody and anti-pd-1 antibody
WO2024048978A1 (en) Novel tmem165-clock epitope and antibody thereto
US20240043568A1 (en) Development of new tumor engager therapeutic drug and use thereof
RU2815926C2 (en) Anti-claudin antibodies and use thereof
TWI793451B (en) Personalized cancer immunotherapy

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: 12887499

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12887499

Country of ref document: EP

Kind code of ref document: A1