WO2014069681A1 - Anticorps ciblant le cancer et composition permettant de prévenir ou de traiter le cancer en contenant - Google Patents

Anticorps ciblant le cancer et composition permettant de prévenir ou de traiter le cancer en contenant 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
English (en)
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/fr
Publication of WO2014069681A1 publication Critical patent/WO2014069681A1/fr

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

La présente invention concerne un anticorps monoclonal capable d'induire l'apoptose d'une cellule cancéreuse en se liant à elle, ainsi que son utilisation médicale. L'anticorps selon la présente invention présente une activité spécifique de liaison aux lignées cellulaires cancéreuses et peut donc être utilisé dans le cadre d'un diagnostic ciblant les cellules cancéreuses ou en tant que conjugué médicamenteux en vue d'un diagnostic ou d'un traitement efficace du cancer. L'anticorps selon la présente invention présente également la caractéristique d'induire l'apoptose des cellules cancéreuses en se liant à elles et, en particulier, l'apoptose des cellules de lymphome et de cancer de l'ovaire, si bien que l'anticorps selon la présente invention peut être utilisé seul en tant que médicament anticancéreux.
PCT/KR2012/008951 2012-10-29 2012-10-29 Anticorps ciblant le cancer et composition permettant de prévenir ou de traiter le cancer en contenant WO2014069681A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/KR2012/008951 WO2014069681A1 (fr) 2012-10-29 2012-10-29 Anticorps ciblant le cancer et composition permettant de prévenir ou de traiter le cancer en contenant

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/KR2012/008951 WO2014069681A1 (fr) 2012-10-29 2012-10-29 Anticorps ciblant le cancer et composition permettant de prévenir ou de traiter le cancer en contenant

Publications (1)

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

Family

ID=50627572

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2012/008951 WO2014069681A1 (fr) 2012-10-29 2012-10-29 Anticorps ciblant le cancer et composition permettant de prévenir ou de traiter le cancer en contenant

Country Status (1)

Country Link
WO (1) WO2014069681A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007515165A (ja) * 2003-12-04 2007-06-14 バクシネックス インコーポレーティッド アポトーシス腫瘍細胞上に露出した細胞内抗原をターゲッティングすることによって腫瘍細胞を死滅させる方法
KR20090078339A (ko) * 2006-10-06 2009-07-17 다케다 야쿠힌 고교 가부시키가이샤 암의 예방·치료제
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 (ja) * 2003-12-04 2007-06-14 バクシネックス インコーポレーティッド アポトーシス腫瘍細胞上に露出した細胞内抗原をターゲッティングすることによって腫瘍細胞を死滅させる方法
KR20090078339A (ko) * 2006-10-06 2009-07-17 다케다 야쿠힌 고교 가부시키가이샤 암의 예방·치료제

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 (zh) 抗切口3(anti-notch3)抗體及抗體-藥物共軛體
JP7458399B2 (ja) 抗クローディン抗体及びそれらの使用
RU2515904C2 (ru) Способ генерирования активных антител к антигену устойчивости, антитела, полученные этим способом, и их применения
US20070112179A1 (en) Novel compounds
TW201609151A (zh) 抗trop2抗體-藥物結合物
KR20100097660A (ko) 항-메소텔린 항체 및 이의 용도
BR112021012608A2 (pt) Porção de ligação que se liga especificamente à claudina18.2, receptor de antígeno quimérico, ácido nucleico, célula imune manipulada, composição farmacêutica, e, método de tratamento de um tumor ou câncer que expressa claudina18.2
WO2013081379A1 (fr) Conjugué médicamenteux comprenant un médicament lié à l'anticorps humain spécifique de c-met et son utilisation
BR112019019650A2 (pt) ligação de anticorpos especificamente à muc1 e utilização dos mesmos
EP3495390A1 (fr) Nouvel anticorps dirigé contre la protéine programmée de mort cellulaire (pd-1) et son utilisation
TW202031687A (zh) 一種融合蛋白及其用途
KR101828794B1 (ko) 중동호흡기증후군 코로나바이러스에 중화활성을 갖는 항체
KR20150129649A (ko) 치료용 안티-bag3 항체들
JP6087289B2 (ja) Napi2b輸送体に対するヒト化モノクローナル抗体の相補性決定領域に由来する抗腫瘍ペプチド
CA2515389A1 (fr) Anticorps monoclonal, gene codant pour un tel anticorps, hybridome, composition medicamenteuse et reactif de diagnostic
WO2014069681A1 (fr) Anticorps ciblant le cancer et composition permettant de prévenir ou de traiter le cancer en contenant
WO2014069682A1 (fr) Anticorps ciblant le cancer et composition permettant de prévenir ou de traiter le cancer en contenant
KR101471253B1 (ko) 암 표적 항체 및 이를 포함하는 암 예방 또는 치료용 조성물
KR101428867B1 (ko) 암 표적 항체 및 이를 포함하는 암 예방 또는 치료용 조성물
WO2022239766A1 (fr) Anticorps anti-cadm1
WO2023211068A1 (fr) Composition pharmaceutique pour le traitement du cancer, comprenant un anticorps anti-igsf1 et un anticorps anti-pd-1
WO2024048978A1 (fr) Nouvel épitope tmem165-clock et anticorps associé
US20240043568A1 (en) Development of new tumor engager therapeutic drug and use thereof
RU2815926C2 (ru) Антитела против клаудина и их применение
TWI793451B (zh) 個人化癌症免疫治療

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