WO2023211068A1 - Pharmaceutical composition for cancer treatment, comprising anti-igsf1 antibody and anti-pd-1 antibody - Google Patents

Pharmaceutical composition for cancer treatment, comprising anti-igsf1 antibody and anti-pd-1 antibody Download PDF

Info

Publication number
WO2023211068A1
WO2023211068A1 PCT/KR2023/005475 KR2023005475W WO2023211068A1 WO 2023211068 A1 WO2023211068 A1 WO 2023211068A1 KR 2023005475 W KR2023005475 W KR 2023005475W WO 2023211068 A1 WO2023211068 A1 WO 2023211068A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
antibody
igsf1
cells
seq
Prior art date
Application number
PCT/KR2023/005475
Other languages
French (fr)
Korean (ko)
Inventor
이민기
고지현
배승건
윤정미
정홍래
홍준기
Original Assignee
웰마커바이오 주식회사
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 웰마커바이오 주식회사 filed Critical 웰마커바이오 주식회사
Publication of WO2023211068A1 publication Critical patent/WO2023211068A1/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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/90Fusion polypeptide containing a motif for post-translational modification
    • C07K2319/92Fusion polypeptide containing a motif for post-translational modification containing an intein ("protein splicing")domain

Definitions

  • the present invention relates to a pharmaceutical composition for preventing or treating cancer comprising an anti-IGSF1 antibody or fragment thereof that specifically binds to IGSF1 and an anti-PD-1 antibody as active ingredients.
  • cancer treatment rates have improved in recent decades due to the development of diagnostic and treatment technologies, the 5-year survival rate for many advanced cancers remains at 5 to 50%. In addition, for some cancers, survival rates have not changed significantly over the past 20 years despite various research and treatments. In particular, cancer cannot be easily treated with conventional cancer treatment regimens, recurrence occurs, and metastasis occurs to other parts, so a more fundamental treatment method is required. Accordingly, there is increasing interest in developing substances to treat cancer by targeting biomarkers that characterize cancer cells, which are believed to be the cause of cancer malignancy, metastasis, and recurrence.
  • Republic of Korea Publication No. 2016-0014564 discloses that the IGSF1 (immunoglobulin superfamily member 1) gene can be used as a biomarker for predicting susceptibility to MET (mesenchymal-epithelial transition factor) inhibitors.
  • IGSF1 immunoglobulin superfamily member 1
  • MET mesenchymal-epithelial transition factor
  • Immune checkpoint inhibitors are anticancer drugs that help activate our body's immune system to attack cancer cells.
  • Anti-PD-1 antibodies such as Keytruda, bind to a specific receptor (PD-1) on T cells and block the pathway for cancer cells to avoid the surveillance system of activated T cells, allowing T cells in the body to attack cancer cells. By doing so, it exhibits an anti-cancer effect (Korean Publication No. 2018-0030580). Since cancer treatment to date has focused on killing rapidly dividing cells, which is a characteristic of cancer cells, it acts not only on cancer cells but also on rapidly dividing normal cells, causing side effects.
  • immunotherapies are known to have few side effects typical of existing anticancer drugs because they use the cancer patient's immune system to affect cancer cells.
  • the present inventors studied to develop a therapeutic agent to effectively treat cancer and found that when anti-IGSF1 antibody and anti-PD-1 antibody, which specifically binds to the C terminus of IGSF1, are used together, excellent anticancer effect is achieved.
  • the present invention was completed by confirming that it represents.
  • one aspect of the present invention is a pharmaceutical composition for preventing or treating cancer comprising an anti-IGSF1 antibody or fragment thereof that specifically binds to the C terminus of IGSF1 and an anti-PD-1 antibody as active ingredients. provides.
  • the anti-IGSF1 antibody according to the present invention showed high specificity and high binding affinity to IGSF1.
  • the anti-IGSF1 antibody according to the present invention increased the infiltration of immune cells within the spheroids when IGSF1-overexpressing lung cancer cell spheroids and human peripheral mononuclear cells were co-cultured.
  • the anti-IGSF1 antibody according to the present invention inhibited tumor growth in humanized mice transplanted with human lung cancer cells overexpressing IGSF1 and increased the expression of cytokines in tumor tissues.
  • the anti-IGSF1 antibody and anti-PD-1 antibody are administered in combination to tumor model mice transplanted with mouse colon cancer cell lines, the results are superior compared to the single administration of the anti-IGSF1 antibody or anti-PD-1 antibody. It showed tumor growth inhibitory activity. Therefore, a pharmaceutical composition containing anti-IGSF1 antibody and anti-PD-1 antibody as active ingredients can be usefully used for cancer prevention and treatment.
  • Figure 1 is a diagram confirming the level of IGSF1 expression in IGSF1 overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) and control group (NCI-H292 MOCK) through Western blot and RT-PCR.
  • FIG. 2 shows tumor infiltrating lymphocytes (TILs) present in the spheroids when IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) and control (NCI-H292 MOCK) spheroids were co-cultured with human peripheral mononuclear cells (PBMCs). ) is a drawing confirming this.
  • TILs tumor infiltrating lymphocytes
  • Figure 3 shows the distribution of hCD45+ cells using flow cytometry to confirm the presence of tumor-infiltrating lymphocytes in tumor tissues of mice transplanted with IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) or control group (NCI-H292 MOCK). This is a graph showing the results of the analysis.
  • Figure 4 shows the expression of IGSF1 and the presence of tumor-infiltrating lymphocytes in tumor tissues of mice transplanted with IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) or control group (NCI-H292 MOCK) using immunohistochemistry. This is a drawing showing the results confirmed.
  • Figure 5 is a graph showing the results of analyzing the binding affinity of the WM-A1-3389 antibody to the IGSF1 antigen using ELISA.
  • Figure 6 is a graph showing the results of analyzing the binding affinity of the WM-A1-3389 antibody to the intracellular IGSF1 antigen using FACS analysis.
  • Figure 7 shows the results of analyzing the binding affinity of the WM-A1-3389 antibody to IGSF1 expressed in IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) and control cells (NCI-H292 MOCK) using FACS analysis. This is a graph showing .
  • Figure 8 shows two types of IGSF1 overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E and HEK293E IGSF1 O/E) and a control (NCI-H292 MOCK and HEK293E MOCK) IGSF1 knockdown (K/D) cell line treated with shIGSF1.
  • NCI-H292 IGSF1 O/E and HEK293E IGSF1 O/E a control
  • IGSF1 knockdown (K/D) cell line treated with shIGSF1 NCI-H292 MOCK and HEK293E MOCK
  • Figure 9 shows tumor-infiltrating lymphocytes present in the spheroids after treatment with IgG or WM-A1-3389 antibody when co-culturing IGSF1-overexpressing human lung cancer cell (NCI-H292 IGSF1 O/E) spheroids and human peripheral mononuclear cells (PBMC). This is a diagram confirming (TIL) through a microscope image.
  • IGSF1-overexpressing human lung cancer cell NCI-H292 IGSF1 O/E
  • PBMC peripheral mononuclear cells
  • Figure 10 shows co-culture of IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) and control (NCI-H292 MOCK) spheroids with human peripheral mononuclear cells (PBMC), after treatment with IgG or WM-A1-3389 antibody.
  • PBMC peripheral mononuclear cells
  • Figure 11 is a graph showing the tumor size measured in the IgG or WM-A1-3389 antibody administered group in a mouse model transplanted with IGSF1 overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E).
  • Figure 12 is a graph showing the tumor size for each individual in the IgG or WM-A1-3389 antibody administration group in a mouse model transplanted with IGSF1 overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E).
  • Figure 13 is a diagram showing the results of analyzing the expression level of IGSF1 in tissues of Caucasian lung cancer patients.
  • Figure 14 shows tumors in the WM-A1-3389 antibody administration group, the anti-PD-1 antibody administration group, or the WM-A1-3389 antibody and anti-PD-1 antibody combination administration group in a mouse model transplanted with a mouse-derived colon cancer cell line (MC38). This is a graph showing the size measured.
  • Figure 15 shows tumors in the WM-A1-3389 antibody administration group, anti-PD-1 antibody administration group, or WM-A1-3389 antibody and anti-PD-1 antibody combination administration group in a mouse model transplanted with mouse-derived colon cancer cell line (CT26). This is a graph showing the size measured.
  • Figure 16 shows the tumor size of the WM-A1-3389 antibody administration group, the anti-PD-1 antibody administration group, or the WM-A1-3389 antibody and anti-PD-1 antibody combination administration group in a mouse model transplanted with a mouse-derived lung cancer cell line (LLC1). This is a graph shown by measuring .
  • One aspect of the present invention provides a pharmaceutical composition for preventing or treating cancer comprising an anti-IGSF1 antibody or fragment thereof that specifically binds to the C terminus of IGSF1 and an anti-PD-1 antibody as active ingredients.
  • PD-1 programmed cell death protein 1
  • CD279 CD279, and is a protein expressed on the surface of activated T cells. It reacts with PD-L1 (B7-H1) and PD-L2 (B7-DC), proteins on the surface of cancer cells, and inhibits T-cell activation, growth factors, and cytokine production mediated by TCR (T cell receptor) and CD28. This induces voice signal transmission.
  • PD-1 inhibitors include, for example, pembrolizumab (Keytruda ® ), MK-3475, nivolumab (Opdivo ® ), cemiplimab (Liptayo ® ), JTX-4014, spartalizumab, Camreli Zumab, sintilimab, thyslerizumab, toripalimab, dostalimab, INCMGA00012, AMP-224, and AMP-514.
  • IGSF1 is a membrane protein encoded by the IGSF1 gene found on the X chromosome of humans and other mammalian species. Although the function of IGSF1 in normal cells is not well known, IGSF1 mutations are known to cause diseases such as IGSF1 deficiency syndrome or central hypothyroidism.
  • the IGSF1 may be included without limitation as long as it is mammalian IGSF1, but preferably refers to human IGSF1. Additionally, in the present invention, the IGSF1 protein includes, but is not limited to, both native and mutant IGSF1 proteins.
  • the native IGSF1 protein generally refers to a polypeptide containing the amino acid sequence of the native IGSF1 protein, and the amino acid sequence of the native IGSF1 protein generally refers to the amino acid sequence found in naturally occurring IGSF1.
  • Information about IGSF1 can be obtained from known databases such as GenBank of the National Institutes of Health, and may have, for example, an amino acid sequence (SEQ ID NO: 19) with Genbank accession number NP_001164433.1, but is not limited thereto.
  • antibody refers to an immunoglobulin molecule that reacts immunologically with a specific antigen, and refers to a protein molecule that specifically recognizes the antigen.
  • the antibodies include whole antibodies, monoclonal antibodies, polyclonal antibodies, single domain antibodies, single chain antibodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, intrabodies, scFvs, Fab fragments, F (ab') fragment, disulfide bond-linked Fv (sdFv), and epitope-binding fragments of any of the above.
  • anti-IGSF1 antibody refers to an antibody capable of binding to IGSF1, and may be used interchangeably with “IGSF1-specific antibody” in the present invention.
  • anti-IGSF1 antibodies can specifically bind to the C terminus of IGSF1.
  • the form of the antibody may include both whole antibodies and antibody fragments.
  • the heavy and light chains of immunoglobulins may each include a constant region and a variable region.
  • the light and heavy chain variable regions of immunoglobulins include three variable regions called complementarity determining regions (CDRs) and four framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • the CDR mainly functions to bind to the epitope of the antigen.
  • the CDRs of each chain are typically called CDR1, CDR2, and CDR3 sequentially, starting from the N terminus, and are identified by the chain on which the specific CDR is located.
  • the anti-IGSF1 antibody or fragment thereof of the present invention may include a heavy chain variable region (VH) comprising H-CDR1 of SEQ ID NO: 1, H-CDR2 of SEQ ID NO: 2, and H-CDR3 of SEQ ID NO: 3.
  • the IGSF1-specific antibody and fragment thereof of the present invention may include a light chain variable region (VL) comprising L-CDR1 of SEQ ID NO: 4, L-CDR2 of SEQ ID NO: 5, and L-CDR3 of SEQ ID NO: 6. there is.
  • the heavy chain variable region may have the amino acid sequence of SEQ ID NO: 7
  • the light chain variable region may have the amino acid sequence of SEQ ID NO: 8.
  • the antibody may be referred to as WM-A1-3389.
  • the heavy chain variable region of the antibody is about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% or It may comprise or consist of an amino acid sequence having about 99% identity or 100% identity.
  • the light chain variable region of the antibody is about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% of the amino acid sequence of SEQ ID NO: 8. % or about 99% identity or 100% identity.
  • Immunoglobulin heavy chain constant regions exhibit different amino acid compositions and sequences and therefore possess different types of antigenicity. Therefore, immunoglobulins can be divided into five categories and referred to as immunoglobulin isotypes, namely IgM, IgD, IgG, IgA and IgE.
  • the corresponding heavy chains are ⁇ chain, ⁇ chain, ⁇ chain, ⁇ chain and ⁇ chain, respectively.
  • the same type of Ig can be classified into different subtypes. For example, IgG can be classified as IgG1, IgG2, IgG3, and IgG4.
  • Light chains can be classified as ⁇ or ⁇ chains depending on their different constant regions. Each of the five types of IgG can have either a ⁇ or ⁇ chain.
  • the anti-IGSF1 antibody or fragment thereof of the present invention may include a constant region derived from IgG, IgA, IgD, IgE, IgM, or a partial hybrid thereof.
  • hybrid means that sequences corresponding to immunoglobulin heavy chain constant regions of two or more different origins exist within the single-chain immunoglobulin heavy chain constant region.
  • a domain hybrid consisting of 1 to 4 domains selected from the group consisting of CH1, CH2, and CH3 of IgG, IgA, IgD, IgE, and IgM is possible.
  • the anti-IGSF1 antibody or fragment thereof of the present invention includes a light chain constant region (LC)
  • the light chain constant region may be derived from a ⁇ or ⁇ light chain.
  • antibody fragment refers to a scFv fragment, which is an Fv fragment that binds to IGSF1, as well as a Fab fragment, Fab' fragment, and F(ab')2 fragment with antigen-binding activity, and the present invention Contains the CDR regions of the antibodies described in.
  • the Fv fragment is the smallest antibody fragment that contains the heavy and light chain variable regions, without constant regions, and retains all antigen-binding sites.
  • composition containing the anti-IGSF1 antibody or fragment thereof and the anti-PD-1 antibody of the present invention as active ingredients exhibits preventive or therapeutic efficacy against cancer.
  • the cancer may be a cancer in which IGSF1 is overexpressed.
  • the above cancers include stomach cancer, liver cancer, lung cancer, non-small cell lung cancer, colon cancer, bladder cancer, bone cancer, blood cancer, breast cancer, melanoma, thyroid cancer, parathyroid cancer, bone marrow cancer, rectal cancer, throat cancer, larynx cancer, esophagus cancer, pancreas cancer, and tongue cancer.
  • it may be any one selected from the group consisting of skin cancer, sinus tumor, uterine cancer, head cancer, cervical cancer, gallbladder cancer, oral cancer, anal cancer, colon cancer, and central nervous system tumor.
  • prevention refers to all actions that inhibit the occurrence of cancer or delay its onset by administering the pharmaceutical composition.
  • treatment refers to any action that improves or beneficially changes cancer symptoms by administering the pharmaceutical composition.
  • the anti-IGSF1 antibody or fragment thereof and the anti-PD-1 antibody are used in any amount (effective amount) depending on the use, formulation, purpose of formulation, etc., as long as they can exhibit anticancer activity. may be included.
  • effective amount refers to the amount of an active ingredient that can induce an anticancer effect. Such effective amounts can be determined experimentally within the scope of the ordinary ability of those skilled in the art.
  • the pharmaceutical composition of the present invention contains the antibody as an active ingredient in an amount of about 0.1% to about 90% by weight, specifically about 0.5% by weight to about 75% by weight, and more specifically about 1% by weight to about 1% by weight, based on the total weight of the composition. It can be contained at 50% by weight.
  • “enhanced efficacy” e.g., improvement in efficacy
  • the term "efficacy” means survival over a period of time, such as 1 year, 5 years, or 10 years, or disease-free survival. can be decided. Additionally, the parameter may include that the size of at least one tumor in the subject is suppressed.
  • the pharmaceutical composition of the present invention may contain a conventional, non-toxic pharmaceutically acceptable carrier that is formulated into a preparation according to a conventional method.
  • the pharmaceutically acceptable carrier can be any carrier that is a non-toxic material suitable for delivery to a patient. Distilled water, alcohol, fats, waxes and inert solids may be included as carriers. Pharmacologically acceptable adjuvants (buffers, dispersants) may also be included in the pharmacological composition.
  • the term “pharmaceutically acceptable carrier” refers to a carrier or diluent that does not irritate living organisms and does not inhibit the biological activity and properties of the administered compound.
  • Acceptable pharmaceutical carriers in compositions formulated as liquid solutions include those that are sterile and biocompatible, such as saline solution, sterile water, Ringer's solution, buffered saline solution, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and One or more of these ingredients can be mixed and used, and other common additives such as sweeteners, solubilizers, wetting agents, emulsifiers, isotonic agents, absorbents, antioxidants, preservatives, lubricants, fillers, buffers, and bacteriostatic agents are added as needed. can do.
  • compositions of the present invention can be prepared in a variety of formulations for parenteral administration (e.g., intramuscular, intravenous, or subcutaneous injection).
  • parenteral administration e.g., intramuscular, intravenous, or subcutaneous injection.
  • the pharmaceutical composition of the present invention can be formulated in the form of injections, transdermal administration, nasal inhalation, and suppositories along with a suitable carrier according to methods known in the art.
  • injectable preparations include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized preparations, and suppositories.
  • Non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate.
  • injectables may contain conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifiers, stabilizers, preservatives, etc.
  • the antibody or composition of the present invention can be administered to a patient in a therapeutically effective amount or a pharmaceutically effective amount.
  • administration means introducing a predetermined substance into an individual by an appropriate method, and the composition may be administered through any general route as long as it can reach the target tissue. It may be administered intraperitoneally, intravenously, intramuscularly, subcutaneously, intradermally, locally, intranasally, or rectally, but is not limited thereto.
  • therapeutically effective amount refers to the amount of a compound or composition effective in preventing or treating the target disease, which is sufficient to treat the disease with a reasonable benefit/risk ratio applicable to medical treatment. It refers to an amount that does not cause side effects.
  • the level of the effective amount is determined by factors including the patient's health status, type and severity of the disease, activity of the drug, sensitivity to the drug, method of administration, time of administration, route of administration and excretion rate, treatment period, drugs combined or used simultaneously, and It may be determined based on other factors well known in the medical field.
  • a therapeutically effective amount refers to an amount of drug that is effective in treating cancer.
  • the effective amount of antibody in the composition of the present invention may vary depending on the patient's age, gender, and weight, and is generally about 0.1 mg to about 1,000 mg, or about 5 mg to about 200 mg per kg of body weight daily. It can be administered every other day or every 2 to 3 weeks, or it can be administered once to three times a day. However, since it may increase or decrease depending on the route of administration, severity of disease, gender, weight, age, etc., the scope of the present invention is not limited thereto.
  • the term “individual” refers to an object to which the composition of the present invention can be applied (prescribed), and may be a mammal such as a rat, mouse, or livestock, including humans. Preferably, it may be a human, but is not limited thereto.
  • the antibody of the present invention or a pharmaceutical composition containing the same may be administered as an individual therapeutic agent or in combination with other therapeutic agents, may be administered sequentially or simultaneously with conventional therapeutic agents, and may be administered singly or multiple times.
  • the other therapeutic agent may additionally include any compound or natural extract whose safety has already been verified and which is known to have anticancer activity in order to increase or reinforce anticancer activity.
  • Another aspect of the present invention provides the use of a pharmaceutical composition comprising the anti-IGSF1 antibody or fragment thereof and the anti-PD-1 antibody of the present invention as active ingredients for preparing a drug for preventing or treating cancer.
  • anti-IGSF1 antibody and fragment thereof, anti-PD-1 antibody, cancer, prevention and treatment are the same as described above.
  • Another aspect of the present invention provides a use for cancer prevention or treatment of a pharmaceutical composition
  • a pharmaceutical composition comprising the anti-IGSF1 antibody or fragment thereof and the anti-PD-1 antibody of the present invention as active ingredients.
  • anti-IGSF1 antibody and fragment thereof, anti-PD-1 antibody, cancer, prevention and treatment are the same as described above.
  • Another aspect of the present invention provides a method for preventing or treating cancer, comprising administering to a subject a pharmaceutical composition containing an anti-IGSF1 antibody or fragment thereof and an anti-PD-1 antibody as active ingredients.
  • anti-IGSF1 antibody and fragment thereof, anti-PD-1 antibody, cancer, administration, treatment and prevention are the same as described above.
  • the subject may be a mammal, preferably a human. Additionally, the individual may be a cancer patient or an individual at a high risk of suffering from cancer.
  • the administration route, dosage, and frequency of administration of the pharmaceutical composition may be administered to the subject in various ways and amounts depending on the patient's condition and the presence or absence of side effects, and the optimal administration method, dosage, and frequency of administration may be determined by a person skilled in the art. You can select by range. Additionally, the anti-IGSF1 antibody or fragment thereof may be administered in combination with other drugs or biochemically active substances known to have therapeutic effects on cancer, or may be formulated in the form of a combination preparation with other drugs.
  • a human Fc fragment crystallizable region
  • His-tag was fused to create an IGSF1 protein expression vector.
  • HEK293F cells were transfected with the prepared IGSF1 expression vector, and then cultured for 6 days in medium supplemented with 1 mM valporic acid (valproate). Afterwards, the IGSF1 extracellular domain was first purified using protein A agarose, and then the IGSF1 extracellular domain was secondarily purified using Superdex 200 gel filtration chromatography, and then used for antibody selection.
  • biopanning After coating and blocking the IGSF1 antigen, biopanning (Y Biologics Co., Ltd.) was performed using the prepared human antibody library phage (Y Biologics Co., Ltd.) to elute only phages that specifically bound to the antigen. did. The second and third rounds of biopanning were performed using the phage amplified in the first round of biopanning. ELISA was performed to confirm the antigen specificity of the positive phage antibody pool obtained through each round of biopanning. In addition, it was confirmed that anti-IGSF1 antibody was enriched in the phage pool obtained through the third round.
  • the antibodies obtained through this confirmation were converted from phage to whole IgG vectors. It was confirmed that the heavy chain and light chain sequences of the converted 95 clones matched the sequences of the phage antibodies.
  • the most optimized antibody was selected and named “WM-A1-3389”.
  • the CDR sequence of the WM-A1-3389 antibody is shown in Table 1 below.
  • a polynucleotide (SEQ ID NO: 23) encoding the heavy chain (SEQ ID NO: 21) was loaded into the N293F vector (Y Biologics Co., Ltd.) (hereinafter referred to as HC DNA).
  • a polynucleotide (SEQ ID NO: 24) encoding the light chain (SEQ ID NO: 22) was loaded into the N293F vector (Y Biologics Co., Ltd.) (hereinafter referred to as LC DNA).
  • LC DNA N293F vector
  • IGSF1 was overexpressed in NCI-H292, a human lung cancer cell line, or HEK293E, a human embryonic kidney cell line, to create a cell line in which IGSF1 was overexpressed ( Figure 1).
  • MOCK is a control group without IGSF1 expression.
  • an expression vector containing a polynucleotide encoding IGSF1 (OriGene Technologies, Inc., Cat No.RC209621) was transfected into human lung cancer cell line NCI-H292 cells or human embryonic kidney cell line HEK293F cells. Afterwards, the transfected cells were selected by culturing them in a medium containing G418 (neomycin). The IGSF1 expression level was confirmed for the selected clones, and the clone showing the highest IGSF1 expression was selected and used in the experiment.
  • MOCK refers to an empty vector in which the polynucleotide encoding IGSF1 is not loaded.
  • Example 2.2 Analysis of the association between IGSF1 expression and tumor-infiltrating lymphocytes in lung cancer cell line spheroids
  • tumor-infiltrating lymphocytes TIL
  • NCI-H292 IGSF1 O/E cells and NCI-H292 MOCK cells were each plated at 2 ⁇ 10 4 cells/well in a U-Bottom 96-well plate (Nunc, 174925). ) were seeded and cultured in a CO 2 incubator at 37°C for 72 hours.
  • Peripheral blood mononuclear cells (PBMC) were prepared by centrifuging at 1,200 rpm for 10 minutes to remove the supernatant and resuspending in PBS.
  • DMSO dimethyl methacrylate
  • CFSE carboxyfluorescein succinimidyl ester, Invitrogen, C34554
  • 1 ⁇ l of 1 mM CFSE solution was added per 1 ⁇ 10 6 cells/ml of prepared peripheral blood mononuclear cells, and then stained for 10 minutes in a CO 2 incubator at 37°C.
  • medium containing FBS fetal bovine serum
  • the reaction was performed at 37°C in a CO 2 incubator for 5 minutes. After removing the supernatant by centrifugation at 1,200 rpm for 10 minutes, the stained peripheral blood mononuclear cells were resuspended in medium to prepare peripheral blood mononuclear cells to be used in the experiment.
  • the formed spheroids were transferred to an ultra-low attachment 96-well plate (Corning, CLS3474), 2 wells at a time, and CFSE-stained peripheral blood mononuclear cells were inoculated at 1 ⁇ 10 5 cells/well and incubated at 37°C in a CO 2 incubator. were co-cultured for 24 hours. Tumor-infiltrating lymphocytes (TIL) were observed through fluorescence microscopy. At this time, NCI-H292 MOCK cells were used as a control for IGSF1 overexpressing cells.
  • TIL Tumor-infiltrating lymphocytes
  • TIL tumor infiltrating lymphocytes
  • Example 2.3 Analysis of the relationship between IGSF1 expression and tumor-infiltrating lymphocytes in tumor tissues of humanized mice transplanted with lung cancer cell lines.
  • tumor infiltrating lymphocytes TIL
  • NSG mice SID (NSGA) mice, F transplanted with human peripheral blood mononuclear cells (PBMC) were treated with NCI-H292 IGSF1 O/E cells, a human lung cancer cell line that overexpresses IGSF1, or NCI-H292 MOCK cells as a control group. Tumor parts of mice were collected 17 days after transplantation with 5 ⁇ 10 6 cells per mouse.
  • Human peripheral blood mononuclear cells were isolated from each collected tumor, analyzed by FACS, and the level of tumor infiltrating lymphocytes and IGSF1 expression within the tumor tissue was confirmed through immunohistochemistry.
  • the tumor tissue was first treated with collagenase B (Roche, cat. #11088815001) and reacted at 37°C for more than 2 hours to decompose the tumor tissue. When the tumor tissue was completely decomposed into cells, it was separated into single cells by pipetting with a 1 ml pipette.
  • the isolated single cells were transferred to a 50 ml tube (SPL, cat. #50050) and washed with 20 ml of PBS. Afterwards, it was centrifuged at 1,200 rpm for 3 minutes and the supernatant was removed. The remaining cells were treated with DNase I (Roche, cat. #11284932001) and incubated at 37°C for 20 minutes. After the reaction, 20 ml of PBS was added, and the supernatant was removed by centrifugation at 1,200 rpm for 3 minutes. The remaining cells were treated with 0.25% Trysin/EDTA (GIBCO, cat. #15400-054). After mixing the cells well, a cell strainer (SPL, cat.
  • NSG mice SID (NSGA) mice, F
  • PBMC peripheral blood mononuclear cells
  • the cells were immersed in target recovery buffer and heated in a microwave oven for 15 minutes. After heating, the cells were left in target recovery buffer for 30 minutes, washed three times with Tris-buffered saline-0.05% Tween 20 (TBS-T), and blocked with blocking solution for 60 minutes.
  • TBS-T Tris-buffered saline-0.05% Tween 20
  • anti-IGSF1 antibody As the primary antibody, anti-IGSF1 antibody (Santacruz, sc-393786) was diluted 1:100 and allowed to bind overnight at 4°C. The next day, it was washed three times with TBS-T and reacted with endogenous peroxidase blocking reagent (Cell Marque, 925B) at room temperature for 5 minutes.
  • the binding affinity of the WM-A1-3389 antibody to the IGSF1 antigen was confirmed at the in vitro level using ELISA analysis.
  • IGSF1 100 ng IGSF1 was treated to coat a 96-well plate, and then 200 ⁇ l of 4% skim milk (PBST) was added and blocked for 1 hour at room temperature.
  • the WM-A1-3389 antibody was serially diluted in 4% skim milk (PBST) at 12 concentrations of 1/3 each and then reacted at room temperature for 2 hours.
  • PBST 4% skim milk
  • the wells were washed with PBST, treated with human IgG Fc-HRP antibody, and reacted at room temperature for 1 hour.
  • TMB peroxidase substrate was added to check the degree of color development. At this time, for confirmation, the absorbance was measured at a wavelength of 450 nm and the results were compared and analyzed.
  • the Kd value of the WM-A1-3389 antibody was 2.2 ⁇ 10 -11 , showing high binding affinity to the IGSF1 antigen (FIG. 5).
  • the medium of NCI-H292 IGSF1 O/E cells and NCI-H292 MOCK cells was removed, washed once with PBS, and then treated with 2 ml of 0.25% trypsin-EDTA to separate the cells.
  • the separated cells were diluted with 8 ml of PBS (hereinafter referred to as FACS buffer) containing 2% FBS and 0.05% sodium azide, and then centrifuged at 1,200 rpm for 1 minute to remove the supernatant.
  • FACS buffer PBS
  • the cells were resuspended in FACS buffer to 1 ⁇ 10 5 cells/ml. After resuspension, cells were dispensed in 1 ml portions into FACS tubes and centrifuged at 1,200 rpm for 1 minute to remove the supernatant.
  • the cell pellet remaining in the FACS tube was vortexed, and WM-A1-3389 antibody was diluted by 1/4 from 20 ⁇ M to 0 ⁇ M per 200 ⁇ l of FACS buffer and added at a total of 12 concentrations. , and reacted at 4°C for 30 minutes. After the reaction was completed, 1 ml of FACS buffer was added to each tube and centrifuged at 1,200 rpm for 1 minute to remove the supernatant. This process was performed a total of two times.
  • the cell pellet remaining in the FACS tube was vortexed, and FITC-labeled goat anti-human IgG antibody (Invitrogen, 62-8411) was added at 5 ⁇ g/ml per 200 ⁇ l of FACS buffer and incubated at 4°C for 30 minutes. Reacted for minutes. After completion of the reaction, 1 ml of FACS buffer was added to each tube and centrifuged at 1,200 rpm for 1 minute to remove the supernatant. This process was performed a total of two times.
  • the remaining cell pellet was resuspended in 200 ⁇ l of FACS buffer and subjected to FACS analysis.
  • FACS analysis measured the fluorescence value of FITC labeled in each cell using a BD LSRFortessa TM Flow Cytometer. Afterwards, the results were analyzed using FlowJo software, and the EC50 value was calculated using the sigma plot program. At this time, NCI-H292 MOCK cells were used as a control for IGSF1 overexpressing cells.
  • WM-A1-3389 antibody In order to analyze the antigen-specific binding ability (target selectivity) of the WM-A1-3389 antibody to the IGSF1 antigen at the cellular level, human lung cancer cells overexpressing IGSF1 (NCI-H292 IGSF1 O/E) were used. The degree of binding of the WM-A1-3389 antibody to expressed IGSF1 was confirmed.
  • the medium of human lung cancer cells overexpressing IGSF1 (NCI-H292 IGSF1 O/E) and its control group (NCI-H292 MOCK) was removed, washed once with PBS, and then added with 2 ml of 0.25% trypsin-EDTA.
  • Cells were separated by treatment. The separated cells were diluted with 8 ml of PBS (hereinafter referred to as FACS buffer) containing 2% FBS and 0.05% sodium azide, and then centrifuged at 1,200 rpm for 1 minute to remove the supernatant. Afterwards, the cells were resuspended in FACS buffer to 1 ⁇ 10 5 cells/ml. After resuspension, cells were dispensed in 1 ml portions into FACS tubes and centrifuged at 1,200 rpm for 1 minute to remove the supernatant.
  • the cell pellet remaining in the FACS tube was vortexed, and 0.4 ⁇ g of human IgG isotype antibody (Bio After completion of the reaction, 1 ml of FACS buffer was added to each tube and centrifuged at 1,200 rpm for 1 minute to remove the supernatant. This process was performed a total of two times. The cell pellet remaining in the FACS tube was vortexed, and 0.4 ⁇ g of FITC-labeled goat anti-human IgG antibody (Invitrogen, 62-8411) was added per 200 ⁇ l of FACS buffer and reacted at 4°C for 30 minutes, protected from light. .
  • the WM-A1-3389 antibody treatment group showed a binding affinity of 2.6% in the control group (NCI-H292 MOCK) and 78.9% in IGSF1 overexpressing cells (NCI-H292 IGSF1 O/E) compared to the IgG isotype treatment group. Binding was shown ( Figure 7).
  • shRNA (hereinafter referred to as shIGSF1) that specifically binds to the mRNA encoding IGSF1 was transfected into NCI-H292 IGSF1 O/E cells and HEK293E IGSF1 O/E cells in which IGSF1 was overexpressed, thereby reducing the expression of IGSF1.
  • IGSF1 K/D cells the binding affinity of the WM-A1-3389 antibody to the intracellular IGSF1 antigen was measured.
  • scramble RNA (hereinafter referred to as sc cells) without shIGSF1 was used as a control for transfection (IGSF1 K/D), and human IgG isotype was used as a control for the WM-A1-3389 antibody.
  • the antigen specificity of the WM-A1-3389 antibody was compared with its binding affinity to IGSF1 K/D cells based on its binding affinity to sc cells.
  • NCI-H292 MOCK cells and HEK293E MOCK cells were used as controls for IGSF1 overexpressing cells, respectively.
  • NCI-H292 IGSF1 O/E and MOCK
  • HEK293E IGSF1 O/E and MOCK
  • the remaining cells were resuspended to a concentration of 1 ⁇ 10 5 cells/ml (NCI-H292) and 0.5 ⁇ 10 5 cells/ml (HEK293E), respectively, and then added at 3 ml each to a 60 mm culture plate and incubated at 37°C. It was cultured in an incubator for one day.
  • shIGSF1 transfection was performed the next day. 200 ⁇ l of jet PRIME buffer and 10 nM of shIGSF1 were added to a 1.5 ml tube and mixed. Afterwards, 4 ⁇ l of jet PRIME reagent was added, mixed, and reacted at room temperature for 10 minutes. Additionally, after replacing the cell medium prepared the day before, 200 ⁇ l of the transfection mixture was added to each cell and reacted in a cell incubator for 24 hours. After 24 hours, it was replaced with new culture medium and cultured for an additional 24 hours.
  • the medium was removed from the transfected cells, and FACS analysis was performed in the same manner as above.
  • Example 5 Analysis of immune anti-cancer efficacy of anti-IGSF1 antibody in lung cancer cell spheroids
  • lung cancer cell spheroids and peripheral blood mononuclear cells were co-cultured to determine tumor-infiltrating lymphocytes (TIL) and immunogenic cell death.
  • the co-cultured cells and supernatant were collected in a tube and centrifuged at 1,200 rpm for 2 minutes to remove the supernatant.
  • the cell pellet was treated with 500 ⁇ l of 0.25% trypsin-EDTA to form single cells, and then diluted with 2 ml of PBS (hereinafter referred to as FACS buffer) to which 2% FBS and 0.05% NaN 3 were added. Afterwards, the supernatant was removed by centrifugation at 1,200 rpm for 3 minutes. The remaining cell pellet was resuspended in 200 ⁇ l of FACS buffer, and anti-HMGB1 antibody (Biolegend, 651408) was added and stained at 4°C for 30 minutes.
  • TIL tumor infiltrating lymphocytes
  • NCI-H292 IGSF1 O/E IGSF1 overexpressing lung cancer cell
  • ICD immunogenic cell death
  • Example 6 By administering anti-IGSF1 antibody alone in a tumor mouse model transplanted with a lung cancer cell line Tumor growth inhibition efficacy analysis
  • mice 6-week-old female peripheral blood mononuclear cell humanized mice (Gem biosciences) were purchased and acclimatized for 1 week, and then IGSF1-overexpressing human lung cancer cells NCI-H292 IGSF1 O/E (5 ⁇ 10 6 cells/mouse) were added to PBS. and was diluted in Matrigel and injected subcutaneously (200 ⁇ l) into the right dorsal side of the mouse. When the tumor size reached approximately 120 mm 3 , IgG isotype (control group) or WM-A1-3389 antibody was administered intraperitoneally at a dose of 10 mg/kg, respectively. Administration was performed once every 3 days for 4 weeks, and the tumor size and body weight of the mice were measured twice a week.
  • the WM-A1-3389 antibody administration group showed higher tumor growth inhibition efficacy than the control group, and showed a tumor inhibition rate (TGI) of about 64.5% (FIG. 11). In addition, it was confirmed that tumor growth was suppressed in individual subjects (FIG. 12).
  • Example 7 Analysis of IGSF1 expression in lung cancer patient tissues of Caucasian ethnicity
  • IGSF1 The expression of IGSF1 in tissues of Caucasian lung cancer patients was confirmed by immunohistochemical staining.
  • human non-small cell lung cancer patient tissue sections were deparaffinized and rehydrated. Thereafter, for heat-induced epitope recovery, it was immersed in target recovery buffer and heated in a microwave oven for 15 minutes. After heating, it was further reacted in target recovery buffer for 30 minutes. Afterwards, it was washed three times with Tris-buffered saline 0.05% Tween 20 (TBS-T), and then blocked with blocking solution for 60 minutes.
  • TBS-T Tris-buffered saline 0.05% Tween 20
  • anti-IGSF1 antibody As the primary antibody, anti-IGSF1 antibody (Santacruz, sc-393786) was diluted 1:100 and allowed to bind overnight at 4°C.
  • tissue sections were washed three times with TBS-T and then reacted with endogenous peroxidase blocking reagent (Cell Marque, 925B) for 5 minutes. Afterwards, it was treated with secondary antibody (Vector, PK-6101 PK-6102) and allowed to bind at room temperature for 60 minutes. After binding, the mixture was washed three times with TBS-T. After washing, avidin-biotin was treated and reacted for 60 minutes. Afterwards, DAB staining (Vector, SK-4100) was performed, and the stained tissue sections were observed through a microscope (FIG. 13).
  • endogenous peroxidase blocking reagent Cell Marque, 925B
  • Example 8 Analysis of tumor growth inhibition efficacy by combined administration of anti-IGSF1 antibody and anticancer agent in tumor mouse model transplanted with various cancer cell lines
  • Example 8.1 Antitumor effect of combined administration of anti-IGSF1 antibody and anticancer agent on syngeneic mouse model transplanted with colon cancer cell line MC38
  • WM-A1-3389 antibody and anti-PD-1 antibody were administered to a mouse model transplanted with MC38, a colon cancer cell line. After co-administration of the antibodies, the tumor growth inhibition efficacy was evaluated.
  • mice 5-week-old female C57BL/6 mice (Orient Bio) were purchased and acclimatized for 1 week, and then the mouse colon cancer cell line MC38 (2.5 ⁇ 10 5 cells/mouse) was diluted in PBS and injected subcutaneously on the right ventral side of the mouse. (100 ⁇ l) was injected.
  • mIgG negative control
  • WM-A1-3389 antibody WM-A1-3389 antibody
  • anti-PD-1 antibody Bio
  • Administration was conducted once every three days for two weeks, and the tumor size and body weight of the mice were measured twice a week. After administration was completed, the experimental animals were euthanized, the tumors were extracted, and their weight was measured.
  • the group administered with the WM-A1-3389 antibody and anti-PD-1 antibody showed superior tumor growth inhibition efficacy compared to the group administered with the WM-A1-3389 antibody or anti-PD-1 antibody alone (FIG. 14 and Table 3).
  • WM-A1-3389 antibody anti-PD-1 antibody Combined use (WM-A1-3389 antibody + anti-PD-1 antibody) Dosage administered (mg/kg) 10 10 10 + 10 TGI (%) 39.1 ⁇ 7.2 43.3 ⁇ 5.2 68.2 ⁇ 8.1
  • Example 8.2 Anti-tumor effect following combined administration of anti-IGSF1 antibody and anticancer agent on syngeneic mouse model transplanted with colon cancer cell line CT26
  • Example 8.1 In order to confirm the anticancer efficacy of the combined administration of the WM-A1-3389 antibody and the anti-PD-1 antibody, the same method as Example 8.1 was performed, except that in this example, CT26, a colon cancer cell line, was used as the transplant cell line. And, 5-week-old female BALB/c mice were used as recipient mice.
  • the WM-A1-3389 antibody group was compared to the negative control group.
  • the tumor growth inhibition efficacy was confirmed in both the anti-PD-1 antibody administration group and the WM-A1-3389 antibody and anti-PD-1 antibody combination administration group.
  • the group administered with the WM-A1-3389 antibody and anti-PD-1 antibody showed superior tumor growth inhibition efficacy compared to the group administered with the WM-A1-3389 antibody or anti-PD-1 antibody alone (FIG. 15 and Table 4).
  • WM-A1-3389 antibody anti-PD-1 antibody Combined use (WM-A1-3389 antibody + anti-PD-1 antibody) Dosage administered (mg/kg) 10 10 10 + 10 TGI (%) 39.8 ⁇ 5.6 3.6 ⁇ 8.6 73.3 ⁇ 5.8
  • Example 8.1 In order to confirm the anticancer efficacy of the combined administration of the WM-A1-3389 antibody and the anti-PD-1 antibody, the same method as Example 8.1 was performed, except that in this example, the lung cancer cell line LLC1 was used as the transplant cell line. , Administration was conducted once every three days for 11 days.
  • WM-A1-3389 antibody and anti-PD-1 antibody were co-administered to a mouse model transplanted with LLC1, a lung cancer cell line, and the tumor growth inhibition efficacy was analyzed, compared to the negative control group, the WM-A1-3389 antibody administration group, Tumor growth inhibition efficacy was confirmed in both the anti-PD-1 antibody administration group and the WM-A1-3389 antibody and anti-PD-1 antibody combination administration group.
  • the group administered with the WM-A1-3389 antibody and anti-PD-1 antibody showed superior tumor growth inhibition efficacy compared to the group administered with the WM-A1-3389 antibody or anti-PD-1 antibody alone (FIG. 16 and Table 5).
  • WM-A1-3389 antibody anti-PD-1 antibody Combined use (WM-A1-3389 antibody + anti-PD-1 antibody) Dosage administered (mg/kg) 10 10 10 + 10 TGI (%) 55.4 ⁇ 10.5 22.2 ⁇ 12.0 84.9 ⁇ 2.6

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to a pharmaceutical composition for cancer prevention or treatment, comprising an anti-IGSF1 antibody and an anti-PD-1 antibody as active ingredients. The anti-IGSF1 antibody, according to the present invention, exhibited high specificity and high binding affinity to IGSF1. The anti-IGSF1 antibody inhibited tumor growth in a humanized mouse transplanted with IGSF1-overexpressing human lung cancer cells, and increased cytokine expression in tumor tissue. In addition, when the anti-IGSF1 antibody and the anti-PD-1 antibody were co-administered into a tumor model mouse transplanted with a mouse colorectal cancer cell line, more excellent tumor growth inhibition activity was exhibited compared to when the anti-IGSF1 antibody or the anti-PD-1 antibody is administered alone. Thus, the pharmaceutical composition comprising the anti-IGSF1 antibody and the anti-PD-1 antibody as active ingredients may be usefully employed for cancer prevention and treatment.

Description

항-IGSF1 항체 및 항-PD-1 항체를 포함하는 암 치료용 약학 조성물Pharmaceutical composition for cancer treatment comprising anti-IGSF1 antibody and anti-PD-1 antibody
본 발명은 IGSF1에 특이적으로 결합하는 항-IGSF1 항체 또는 이의 단편 및 항-PD-1 항체를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물에 관한 것이다.The present invention relates to a pharmaceutical composition for preventing or treating cancer comprising an anti-IGSF1 antibody or fragment thereof that specifically binds to IGSF1 and an anti-PD-1 antibody as active ingredients.
암의 치료에 있어 항암제와 같은 화학적 치료 용법은 어느 정도 효과를 거두고 있으나, 암의 다양한 발병 기전과 항암제 내성으로 인하여 많은 연구가 요구되고 있다. 최근 수십년간 진단과 치료 기술의 발달로 암 치료율이 향상되었으나, 많은 진행성 암에 있어서 5년 생존율은 5 내지 50%에 머무르고 있다. 또한, 일부 암에 있어서는 다양한 연구와 치료에도 불구하고 지난 20년간의 생존율이 크게 변하지 못한 상태이다. 특히, 암은 종래의 암 치료 요법에 의해 쉽게 치료되지 못하고 재발이 일어나고 있으며 다른 부위로의 전이가 일어나므로, 보다 본질적인 치료 방법이 요구되고 있는 실정이다. 이에, 암의 악성화, 전이 및 재발의 원인으로 판단되는 암 세포의 특징이 되는 바이오마커를 타겟으로 암을 치료하기 위한 물질 개발에 관심이 높아지고 있다.In the treatment of cancer, chemical treatment methods such as anticancer drugs are effective to some extent, but much research is required due to the various pathogenesis of cancer and resistance to anticancer drugs. Although cancer treatment rates have improved in recent decades due to the development of diagnostic and treatment technologies, the 5-year survival rate for many advanced cancers remains at 5 to 50%. In addition, for some cancers, survival rates have not changed significantly over the past 20 years despite various research and treatments. In particular, cancer cannot be easily treated with conventional cancer treatment regimens, recurrence occurs, and metastasis occurs to other parts, so a more fundamental treatment method is required. Accordingly, there is increasing interest in developing substances to treat cancer by targeting biomarkers that characterize cancer cells, which are believed to be the cause of cancer malignancy, metastasis, and recurrence.
한편, 대한민국 공개공보 제2016-0014564호에서는 IGSF1(immunoglobulin superfamily member 1) 유전자가 MET(mesenchymal-epithelial transition factor) 저해제에 대한 감수성 예측용 바이오마커로 사용할 수 있다는 점을 개시하고 있다. 상기 문헌에서는, 암 환자를 치료하기 전에 상기 바이오마커를 이용하여 환자 개개인의 감수성을 판정함으로써 치료 효과가 높은 항암제를 선택할 수 있음을 개시하고 있다.Meanwhile, Republic of Korea Publication No. 2016-0014564 discloses that the IGSF1 (immunoglobulin superfamily member 1) gene can be used as a biomarker for predicting susceptibility to MET (mesenchymal-epithelial transition factor) inhibitors. The above document discloses that an anticancer drug with high therapeutic effect can be selected by determining the sensitivity of each patient using the biomarker before treating the cancer patient.
최근 키트루다(Keytruda®)와 같은 면역관문 억제제가 각광을 받고 있다. 면역관문 억제제는 우리 몸의 면역계를 활성화시켜 암 세포를 공격하도록 도움을 주는 항암제이다. 키트루다와 같은 항-PD-1 항체는 T 세포의 특정 수용체(PD-1)에 결합하여 암세포가 활동성 T 세포의 감시 체계를 피하는 경로를 차단하여, 인체 내 T 세포가 암세포를 공격할 수 있게 함으로써 항암 효과를 나타내게 된다(대한민국 공개공보 제2018-0030580호). 현재까지 암 치료는 암세포의 특징인 빠르게 분열하는 세포를 죽이는데 초점을 맞추었기 때문에, 암세포뿐만 아니라 정상 세포 중 빠르게 분열하는 세포에도 작용하여 부작용이 나타났다. 그러나, 면역항암제는 암 환자의 면역체계를 활용해 암세포에 영향을 주므로 기존 항암제의 전형적인 부작용이 거의 없다고 알려져 있다.Recently, immune checkpoint inhibitors such as Keytruda ® have been in the spotlight. Immune checkpoint inhibitors are anticancer drugs that help activate our body's immune system to attack cancer cells. Anti-PD-1 antibodies, such as Keytruda, bind to a specific receptor (PD-1) on T cells and block the pathway for cancer cells to avoid the surveillance system of activated T cells, allowing T cells in the body to attack cancer cells. By doing so, it exhibits an anti-cancer effect (Korean Publication No. 2018-0030580). Since cancer treatment to date has focused on killing rapidly dividing cells, which is a characteristic of cancer cells, it acts not only on cancer cells but also on rapidly dividing normal cells, causing side effects. However, immunotherapies are known to have few side effects typical of existing anticancer drugs because they use the cancer patient's immune system to affect cancer cells.
이에, 본 발명자들은 암을 효과적으로 치료하기 위한 치료제를 개발하기 위해 연구한 결과, IGSF1의 C 말단에 특이적으로 결합하는 항-IGSF1 항체 및 항-PD-1 항체를 함께 사용하였을 시, 뛰어난 항암 효과를 나타낸다는 것을 확인함으로써 본 발명을 완성하였다.Accordingly, the present inventors studied to develop a therapeutic agent to effectively treat cancer and found that when anti-IGSF1 antibody and anti-PD-1 antibody, which specifically binds to the C terminus of IGSF1, are used together, excellent anticancer effect is achieved. The present invention was completed by confirming that it represents.
상기 목적 달성을 위해, 본 발명의 일 측면은, IGSF1의 C 말단에 특이적으로 결합하는 항-IGSF1 항체 또는 이의 단편 및 항-PD-1 항체를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물을 제공한다.To achieve the above object, one aspect of the present invention is a pharmaceutical composition for preventing or treating cancer comprising an anti-IGSF1 antibody or fragment thereof that specifically binds to the C terminus of IGSF1 and an anti-PD-1 antibody as active ingredients. provides.
본 발명에 따른 항-IGSF1 항체는 IGSF1에 높은 특이성과 높은 결합력을 나타냈다. 본 발명에 따른 항-IGSF1 항체는 IGSF1이 과발현된 폐암 세포 스페로이드와 인간 말초 단핵세포를 공동 배양 시, 스페로이드 내의 면역세포의 침윤을 증가시켰다. 또한, 본 발명에 따른 항-IGSF1 항체는 IGSF1이 과발현된 인간 폐암 세포를 이식한 인간화 마우스의 종양 성장을 억제시키고, 종양 조직 내 사이토카인의 발현을 증가시켰다. 상기 결과를 통해 항-IGSF1 항체는 IGSF1 발현이 증가된 폐암 조직 내로 면역세포의 침윤 및 면역반응을 증가시킴으로써 종양 성장을 억제시킬 수 있음을 확인하였다. 뿐만 아니라, 상기 항-IGSF1 항체 및 항-PD-1 항체를 마우스 대장암 세포주가 이식된 종양 모델 마우스에 병용 투여할 경우, 항-IGSF1 항체 또는 항-PD-1 항체의 단독 투여와 비교하여 월등한 종양 성장 억제 활성을 나타냈다. 따라서, 항-IGSF1 항체 및 항-PD-1 항체를 유효성분으로 포함하는 약학 조성물은 암 예방 및 치료에 유용하게 사용될 수 있다.The anti-IGSF1 antibody according to the present invention showed high specificity and high binding affinity to IGSF1. The anti-IGSF1 antibody according to the present invention increased the infiltration of immune cells within the spheroids when IGSF1-overexpressing lung cancer cell spheroids and human peripheral mononuclear cells were co-cultured. In addition, the anti-IGSF1 antibody according to the present invention inhibited tumor growth in humanized mice transplanted with human lung cancer cells overexpressing IGSF1 and increased the expression of cytokines in tumor tissues. Through the above results, it was confirmed that anti-IGSF1 antibody can inhibit tumor growth by increasing the infiltration of immune cells and immune response into lung cancer tissue with increased IGSF1 expression. In addition, when the anti-IGSF1 antibody and anti-PD-1 antibody are administered in combination to tumor model mice transplanted with mouse colon cancer cell lines, the results are superior compared to the single administration of the anti-IGSF1 antibody or anti-PD-1 antibody. It showed tumor growth inhibitory activity. Therefore, a pharmaceutical composition containing anti-IGSF1 antibody and anti-PD-1 antibody as active ingredients can be usefully used for cancer prevention and treatment.
도 1은 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E) 및 대조군(NCI-H292 MOCK)에서 IGSF1 발현량을 웨스턴 블롯 및 RT-PCR을 통해 확인한 도면이다.Figure 1 is a diagram confirming the level of IGSF1 expression in IGSF1 overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) and control group (NCI-H292 MOCK) through Western blot and RT-PCR.
도 2는 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E) 및 대조군(NCI-H292 MOCK) 스페로이드와 인간 말초 단핵세포(PBMC)를 공동 배양 시, 스페로이드 내에 존재하는 종양 침윤 림프구(TIL)를 확인한 도면이다.Figure 2 shows tumor infiltrating lymphocytes (TILs) present in the spheroids when IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) and control (NCI-H292 MOCK) spheroids were co-cultured with human peripheral mononuclear cells (PBMCs). ) is a drawing confirming this.
도 3은 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E) 또는 대조군(NCI-H292 MOCK)을 이식한 마우스의 종양 조직에서 종양 침윤 림프구의 존재를 확인하기 위하여, hCD45+ 세포의 분포 정도를 유세포 분석을 통해 분석한 결과를 나타낸 그래프이다.Figure 3 shows the distribution of hCD45+ cells using flow cytometry to confirm the presence of tumor-infiltrating lymphocytes in tumor tissues of mice transplanted with IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) or control group (NCI-H292 MOCK). This is a graph showing the results of the analysis.
도 4는 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E) 또는 대조군(NCI-H292 MOCK)을 이식한 마우스의 종양 조직에서 IGSF1의 발현 및 종양 침윤 림프구의 존재를 면역조직화학염색법(immunohistochemistry)으로 확인한 결과를 나타낸 도면이다.Figure 4 shows the expression of IGSF1 and the presence of tumor-infiltrating lymphocytes in tumor tissues of mice transplanted with IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) or control group (NCI-H292 MOCK) using immunohistochemistry. This is a drawing showing the results confirmed.
도 5는 IGSF1 항원에 대한 WM-A1-3389 항체의 결합 친화도를 ELISA를 이용하여 분석한 결과를 나타낸 그래프이다.Figure 5 is a graph showing the results of analyzing the binding affinity of the WM-A1-3389 antibody to the IGSF1 antigen using ELISA.
도 6은 세포 내 IGSF1 항원에 대한 WM-A1-3389 항체의 결합 친화도를 FACS 분석을 이용하여 분석한 결과를 나타낸 그래프이다.Figure 6 is a graph showing the results of analyzing the binding affinity of the WM-A1-3389 antibody to the intracellular IGSF1 antigen using FACS analysis.
도 7은 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E) 및 대조군(NCI-H292 MOCK)에서 세포에서 발현되는 IGSF1에 대한 WM-A1-3389 항체의 결합력을 FACS 분석을 이용하여 분석한 결과를 나타낸 그래프이다.Figure 7 shows the results of analyzing the binding affinity of the WM-A1-3389 antibody to IGSF1 expressed in IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) and control cells (NCI-H292 MOCK) using FACS analysis. This is a graph showing .
도 8은 2종의 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E 및 HEK293E IGSF1 O/E) 및 대조군(NCI-H292 MOCK 및 HEK293E MOCK)에 shIGSF1을 처리한 IGSF1 녹다운(K/D) 세포주에서 WM-A1-3389 항체의 결합 특이성을 분석한 결과를 나타낸 그래프이다.Figure 8 shows two types of IGSF1 overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E and HEK293E IGSF1 O/E) and a control (NCI-H292 MOCK and HEK293E MOCK) IGSF1 knockdown (K/D) cell line treated with shIGSF1. This is a graph showing the results of analyzing the binding specificity of the WM-A1-3389 antibody.
도 9는 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E) 스페로이드와 인간 말초 단핵세포(PBMC)를 공동 배양 시, IgG 또는 WM-A1-3389 항체 처리 후 스페로이드 내에 존재하는 종양 침윤 림프구(TIL)를 현미경 이미지를 통해 확인한 도면이다. Figure 9 shows tumor-infiltrating lymphocytes present in the spheroids after treatment with IgG or WM-A1-3389 antibody when co-culturing IGSF1-overexpressing human lung cancer cell (NCI-H292 IGSF1 O/E) spheroids and human peripheral mononuclear cells (PBMC). This is a diagram confirming (TIL) through a microscope image.
도 10은 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E) 및 대조군(NCI-H292 MOCK) 스페로이드와 인간 말초 단핵세포(PBMC)를 공동 배양 시, IgG 또는 WM-A1-3389 항체 처리 후 스페로이드 내의 HMGB1의 발현을 나타낸 그래프이다.Figure 10 shows co-culture of IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) and control (NCI-H292 MOCK) spheroids with human peripheral mononuclear cells (PBMC), after treatment with IgG or WM-A1-3389 antibody. This is a graph showing the expression of HMGB1 in spheroids.
도 11은 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E)를 이식한 마우스 모델에서 IgG 또는 WM-A1-3389 항체 투여군의 종양 크기를 측정하여 나타낸 그래프이다.Figure 11 is a graph showing the tumor size measured in the IgG or WM-A1-3389 antibody administered group in a mouse model transplanted with IGSF1 overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E).
도 12는 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E)를 이식한 마우스 모델에서 IgG 또는 WM-A1-3389 항체 투여군의 개체별 종양 크기를 나타낸 그래프이다.Figure 12 is a graph showing the tumor size for each individual in the IgG or WM-A1-3389 antibody administration group in a mouse model transplanted with IGSF1 overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E).
도 13은 코카서스 인종(caucasian) 폐암 환자 조직에서 IGSF1의 발현 정도를 분석한 결과를 나타낸 도면이다.Figure 13 is a diagram showing the results of analyzing the expression level of IGSF1 in tissues of Caucasian lung cancer patients.
도 14는 마우스 유래 대장암 세포주(MC38)를 이식한 마우스 모델에서 WM-A1-3389 항체 투여군, 항-PD-1 항체 투여군 또는 WM-A1-3389 항체 및 항-PD-1 항체 병용 투여군의 종양 크기를 측정하여 나타낸 그래프이다.Figure 14 shows tumors in the WM-A1-3389 antibody administration group, the anti-PD-1 antibody administration group, or the WM-A1-3389 antibody and anti-PD-1 antibody combination administration group in a mouse model transplanted with a mouse-derived colon cancer cell line (MC38). This is a graph showing the size measured.
도 15는 마우스 유래 대장암 세포주(CT26)를 이식한 마우스 모델에서 WM-A1-3389 항체 투여군, 항-PD-1 항체 투여군 또는 WM-A1-3389 항체 및 항-PD-1 항체 병용 투여군의 종양 크기를 측정하여 나타낸 그래프이다.Figure 15 shows tumors in the WM-A1-3389 antibody administration group, anti-PD-1 antibody administration group, or WM-A1-3389 antibody and anti-PD-1 antibody combination administration group in a mouse model transplanted with mouse-derived colon cancer cell line (CT26). This is a graph showing the size measured.
도 16은 마우스 유래 폐암 세포주(LLC1)를 이식한 마우스 모델에서 WM-A1-3389 항체 투여군, 항-PD-1 항체 투여군 또는 WM-A1-3389 항체 및 항-PD-1 항체 병용 투여군의 종양 크기를 측정하여 나타낸 그래프이다.Figure 16 shows the tumor size of the WM-A1-3389 antibody administration group, the anti-PD-1 antibody administration group, or the WM-A1-3389 antibody and anti-PD-1 antibody combination administration group in a mouse model transplanted with a mouse-derived lung cancer cell line (LLC1). This is a graph shown by measuring .
본 발명의 일 측면은, IGSF1의 C 말단에 특이적으로 결합하는 항-IGSF1 항체 또는 이의 단편 및 항-PD-1 항체를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물을 제공한다.One aspect of the present invention provides a pharmaceutical composition for preventing or treating cancer comprising an anti-IGSF1 antibody or fragment thereof that specifically binds to the C terminus of IGSF1 and an anti-PD-1 antibody as active ingredients.
항-PD-1 항체anti-PD-1 antibody
본 명세서에서 사용된 용어, "PD-1(programmed cell death protein 1)"은 CD279로 지칭되며, 활성화된 T 세포의 표면에 발현되는 단백질이다. 암세포 표면에 있는 단백질인 PD-L1(B7-H1) 및 PD-L2(B7-DC)와 반응하여 TCR(T cell receptor) 및 CD28로 매개된 T-세포 활성, 성장 인자 및 사이토카인 생성을 억제하여 음성 신호전달을 유도한다. PD-1 억제제는 예를 들어, 펨브롤리주맙(키트루다®), MK-3475, 니볼루맙(옵디보®), 세미플리맙(립타요®), JTX-4014, 스파르탈리주맙, 캄렐리주맙, 신틸리맙, 티슬레리주맙, 토리팔리맙, 도스탈리맙, INCMGA00012, AMP-224, 및 AMP-514이다.As used herein, the term “PD-1 (programmed cell death protein 1)” refers to CD279, and is a protein expressed on the surface of activated T cells. It reacts with PD-L1 (B7-H1) and PD-L2 (B7-DC), proteins on the surface of cancer cells, and inhibits T-cell activation, growth factors, and cytokine production mediated by TCR (T cell receptor) and CD28. This induces voice signal transmission. PD-1 inhibitors include, for example, pembrolizumab (Keytruda ® ), MK-3475, nivolumab (Opdivo ® ), cemiplimab (Liptayo ® ), JTX-4014, spartalizumab, Camreli Zumab, sintilimab, thyslerizumab, toripalimab, dostalimab, INCMGA00012, AMP-224, and AMP-514.
항-IGSF1 항체anti-IGSF1 antibody
본 명세서에서 사용된 용어, "IGSF1"은 인간 및 다른 포유류 종의 X 염색체에서 발견되는 IGSF1 유전자에 의해 암호화된 막단백질이다. 정상세포에서 IGSF1의 기능에 대해서는 잘 알려져 있지 않으나, IGSF1 돌연변이는 IGSF1 결핍 증후군(IGSF1 deficiency syndrome) 또는 중추성 갑상선 기능 저하증 등의 질병을 유발하는 것으로 알려져 있다.As used herein, the term “IGSF1” is a membrane protein encoded by the IGSF1 gene found on the X chromosome of humans and other mammalian species. Although the function of IGSF1 in normal cells is not well known, IGSF1 mutations are known to cause diseases such as IGSF1 deficiency syndrome or central hypothyroidism.
본 발명에서 상기 IGSF1은 포유류의 IGSF1이라면 제한없이 포함될 수 있으나, 바람직하게는 인간 IGSF1을 의미할 수 있다. 또한, 본 발명에서 상기 IGSF1 단백질은 천연형 또는 변이체 IGSF1 단백질을 모두 포함하나, 이에 제한되지 않는다. 상기 천연형 IGSF1 단백질이란 천연형 IGSF1 단백질의 아미노산 서열을 포함하는 폴리펩타이드를 일반적으로 지칭하며, 상기 천연형 IGSF1 단백질의 아미노산 서열이란 천연 발생 IGSF1에서 발견되는 아미노산 서열을 일반적으로 지칭한다. 상기 IGSF1에 대한 정보는 미국국립보건원의 GenBank 등 공지의 데이터베이스로부터 얻을 수 있으며, 예를 들어 Genbank accession number NP_001164433.1의 아미노산 서열(서열번호 19)을 가질 수 있으나, 이에 제한되지 않는다.In the present invention, the IGSF1 may be included without limitation as long as it is mammalian IGSF1, but preferably refers to human IGSF1. Additionally, in the present invention, the IGSF1 protein includes, but is not limited to, both native and mutant IGSF1 proteins. The native IGSF1 protein generally refers to a polypeptide containing the amino acid sequence of the native IGSF1 protein, and the amino acid sequence of the native IGSF1 protein generally refers to the amino acid sequence found in naturally occurring IGSF1. Information about IGSF1 can be obtained from known databases such as GenBank of the National Institutes of Health, and may have, for example, an amino acid sequence (SEQ ID NO: 19) with Genbank accession number NP_001164433.1, but is not limited thereto.
본 명세서에서 사용된 용어, "항체"는 특정 항원과 면역학적으로 반응하는 면역글로불린 분자로, 항원을 특이적으로 인식하는 단백질 분자를 의미한다. 상기 항체는 전체(whole) 항체, 단일클론항체, 다클론항체, 단일 도메인 항체, 단쇄 항체, 다중특이적 항체, 인간 항체, 인간화 항체, 키메라 항체, 인트라바디(intrabody), scFv, Fab 단편, F(ab') 단편, 이황화 결합으로 연결한 Fv(sdFv) 및 상기 중 임의의 에피토프(epitope) 결합 단편을 포함하지만, 이에 제한되지 않는다.As used herein, the term “antibody” refers to an immunoglobulin molecule that reacts immunologically with a specific antigen, and refers to a protein molecule that specifically recognizes the antigen. The antibodies include whole antibodies, monoclonal antibodies, polyclonal antibodies, single domain antibodies, single chain antibodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, intrabodies, scFvs, Fab fragments, F (ab') fragment, disulfide bond-linked Fv (sdFv), and epitope-binding fragments of any of the above.
본 명세서에서 사용된 용어, "항-IGSF1 항체"는 IGSF1에 결합할 수 있는 항체를 의미하며, 본 발명에서 "IGSF1에 특이적인 항체"와 혼용되어 사용될 수 있다. 특히, 항-IGSF1 항체는 IGSF1의 C 말단에 특이적으로 결합할 수 있다. 상기 항체의 형태는 전체(whole) 항체 및 항체 단편을 모두 포함할 수 있다.As used herein, the term “anti-IGSF1 antibody” refers to an antibody capable of binding to IGSF1, and may be used interchangeably with “IGSF1-specific antibody” in the present invention. In particular, anti-IGSF1 antibodies can specifically bind to the C terminus of IGSF1. The form of the antibody may include both whole antibodies and antibody fragments.
면역글로불린의 중쇄 및 경쇄는 각각 불변영역(constant region) 및 가변영역(variable region)을 포함할 수 있다. 면역글로불린의 경쇄 및 중쇄 가변영역은, 상보성 결정 영역(complementarity determining region, CDR)이라 불리는 3개의 다변가능한 영역 및 4개의 구조 영역(framework region, FR)을 포함한다. 상기 CDR은 주로 항원의 에피토프에 결합하는 역할을 한다. 각각의 사슬의 CDR은 전형적으로 N 말단으로부터 시작하여 순차적으로 CDR1, CDR2, CDR3로 불리고, 특정 CDR이 위치하고 있는 사슬에 의해서 식별된다.The heavy and light chains of immunoglobulins may each include a constant region and a variable region. The light and heavy chain variable regions of immunoglobulins include three variable regions called complementarity determining regions (CDRs) and four framework regions (FRs). The CDR mainly functions to bind to the epitope of the antigen. The CDRs of each chain are typically called CDR1, CDR2, and CDR3 sequentially, starting from the N terminus, and are identified by the chain on which the specific CDR is located.
본 발명의 항-IGSF1 항체 또는 이의 단편은 서열번호 1의 H-CDR1, 서열번호 2의 H-CDR2 및 서열번호 3의 H-CDR3을 포함하는 중쇄 가변영역(VH)을 포함할 수 있다. 또한, 본 발명의 IGSF1에 특이적인 항체 및 이의 단편은 서열번호 4의 L-CDR1, 서열번호 5의 L-CDR2 및 서열번호 6의 L-CDR3을 포함하는 경쇄 가변영역(VL)을 포함할 수 있다. 이때, 상기 중쇄 가변영역은 서열번호 7의 아미노산 서열을 가질 수 있으며, 상기 경쇄 가변영역은 서열번호 8의 아미노산 서열을 가질 수 있다. 본 명세서에서 상기 항체는 WM-A1-3389로 지칭될 수 있다.The anti-IGSF1 antibody or fragment thereof of the present invention may include a heavy chain variable region (VH) comprising H-CDR1 of SEQ ID NO: 1, H-CDR2 of SEQ ID NO: 2, and H-CDR3 of SEQ ID NO: 3. In addition, the IGSF1-specific antibody and fragment thereof of the present invention may include a light chain variable region (VL) comprising L-CDR1 of SEQ ID NO: 4, L-CDR2 of SEQ ID NO: 5, and L-CDR3 of SEQ ID NO: 6. there is. At this time, the heavy chain variable region may have the amino acid sequence of SEQ ID NO: 7, and the light chain variable region may have the amino acid sequence of SEQ ID NO: 8. In this specification, the antibody may be referred to as WM-A1-3389.
상기 항체의 중쇄 가변영역은 서열번호 7의 아미노산 서열과 약 90%, 약 91%, 약 92%, 약 93%, 약 94%, 약 95%, 약 96%, 약 97%, 약 98% 또는 약 99% 동일성 또는 100% 동일성을 갖는 아미노산 서열을 포함하거나 이들로 이루어질 수 있다. 또한, 상기 항체의 경쇄 가변영역은 서열번호 8의 아미노산 서열과 약 90%, 약 91%, 약 92%, 약 93%, 약 94%, 약 95%, 약 96%, 약 97%, 약 98% 또는 약 99% 동일성 또는 100% 동일성을 갖는 아미노산 서열을 포함하거나 이들로 이루어질 수 있다.The heavy chain variable region of the antibody is about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% or It may comprise or consist of an amino acid sequence having about 99% identity or 100% identity. In addition, the light chain variable region of the antibody is about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% of the amino acid sequence of SEQ ID NO: 8. % or about 99% identity or 100% identity.
면역글로불린 중쇄 불변영역(CH)은 상이한 아미노산 조성 및 순서를 나타내므로, 상이한 유형의 항원성을 보유한다. 따라서, 면역글로불린은 다섯 가지 카테고리로 분류될 수 있으며, 면역글로불린 이소형, 즉, IgM, IgD, IgG, IgA 및 IgE로 지칭될 수 있다. 이에 상응하는 중쇄는 각각 μ 사슬, δ 사슬, γ 사슬, α 사슬 및 ε 사슬이다. 또한, 힌지 영역(hinge region)의 아미노산 조성 및 중쇄 이황화 결합의 수와 위치에 따라, 동일한 유형의 Ig는 상이한 하위 유형으로 분류될 수 있다. 예를 들어, IgG는 IgG1, IgG2, IgG3, 및 IgG4로 분류될 수 있다. 경쇄는 상이한 불변영역에 따라 κ 또는 λ 사슬로 분류될 수 있다. 다섯 가지 유형의 IgG 각각은 κ 또는 λ 사슬을 가질 수 있다.Immunoglobulin heavy chain constant regions (CH) exhibit different amino acid compositions and sequences and therefore possess different types of antigenicity. Therefore, immunoglobulins can be divided into five categories and referred to as immunoglobulin isotypes, namely IgM, IgD, IgG, IgA and IgE. The corresponding heavy chains are μ chain, δ chain, γ chain, α chain and ε chain, respectively. Additionally, depending on the amino acid composition of the hinge region and the number and position of heavy chain disulfide bonds, the same type of Ig can be classified into different subtypes. For example, IgG can be classified as IgG1, IgG2, IgG3, and IgG4. Light chains can be classified as κ or λ chains depending on their different constant regions. Each of the five types of IgG can have either a κ or λ chain.
본 발명의 항-IGSF1 항체 또는 이의 단편이 불변영역을 포함하는 경우, IgG, IgA, IgD, IgE, IgM 유래, 또는 이들이 부분적으로 혼합(hybrid)된 불변영역을 포함할 수 있다.When the anti-IGSF1 antibody or fragment thereof of the present invention includes a constant region, it may include a constant region derived from IgG, IgA, IgD, IgE, IgM, or a partial hybrid thereof.
본 명세서에서 사용된 용어, "혼합된(hybrid)"은 단쇄 면역글로불린 중쇄 불변영역 내에 2개 이상의 상이한 기원의 면역글로불린 중쇄 불변영역에 해당하는 서열이 존재함을 의미한다. 예를 들어 IgG, IgA, IgD, IgE 및 IgM의 CH1, CH2 및 CH3으로 이루어진 그룹으로부터 선택되는 1개 내지 4개 도메인으로 이루어진 도메인의 하이브리드가 가능하다.As used herein, the term “hybrid” means that sequences corresponding to immunoglobulin heavy chain constant regions of two or more different origins exist within the single-chain immunoglobulin heavy chain constant region. For example, a domain hybrid consisting of 1 to 4 domains selected from the group consisting of CH1, CH2, and CH3 of IgG, IgA, IgD, IgE, and IgM is possible.
또한, 본 발명의 상기 항-IGSF1 항체 또는 이의 단편이 경쇄 불변영역(LC)을 포함하는 경우, 상기 경쇄 불변영역은 λ 또는 κ 경쇄 유래일 수 있다. Additionally, when the anti-IGSF1 antibody or fragment thereof of the present invention includes a light chain constant region (LC), the light chain constant region may be derived from a λ or κ light chain.
본 명세서에서 사용된 용어, "항체 단편"은 항원-결합 활성을 갖는 Fab 단편, Fab' 단편, F(ab')2 단편뿐만 아니라, IGSF1에 결합하는 Fv 단편인 scFv 단편을 지칭하며, 본 발명에 기술된 항체의 CDR 영역을 포함한다. Fv 단편은 불변 영역 없이, 중쇄 가변영역 및 경쇄 가변영역을 포함하며, 모든 항원-결합 자리를 보유하는 최소 항체 단편이다.As used herein, the term "antibody fragment" refers to a scFv fragment, which is an Fv fragment that binds to IGSF1, as well as a Fab fragment, Fab' fragment, and F(ab')2 fragment with antigen-binding activity, and the present invention Contains the CDR regions of the antibodies described in. The Fv fragment is the smallest antibody fragment that contains the heavy and light chain variable regions, without constant regions, and retains all antigen-binding sites.
약학 조성물pharmaceutical composition
본 발명의 항-IGSF1 항체 또는 이의 단편 및 항-PD-1 항체를 유효성분으로 포함하는 약학 조성물은 암에 대하여 예방 또는 치료 효능을 나타낸다.The pharmaceutical composition containing the anti-IGSF1 antibody or fragment thereof and the anti-PD-1 antibody of the present invention as active ingredients exhibits preventive or therapeutic efficacy against cancer.
이때, 상기 암은 IGSF1이 과발현되어 있는 암일 수 있다.At this time, the cancer may be a cancer in which IGSF1 is overexpressed.
또한, 상기 암은 위암, 간암, 폐암, 비소세포 폐암, 대장암, 방관암, 골암, 혈액암, 유방암, 흑색종양, 갑상선암, 부갑성선암, 골수암, 직장암, 인후암, 후두암, 식도암, 췌장암, 설암, 피부암, 죄종양, 자궁암, 두부암, 경부암, 담낭암, 구강암, 항문 부근암, 결장암 및 중추신경계 종양으로 구성된 군으로부터 선택되는 어느 하나인 것일 수 있다.In addition, the above cancers include stomach cancer, liver cancer, lung cancer, non-small cell lung cancer, colon cancer, bladder cancer, bone cancer, blood cancer, breast cancer, melanoma, thyroid cancer, parathyroid cancer, bone marrow cancer, rectal cancer, throat cancer, larynx cancer, esophagus cancer, pancreas cancer, and tongue cancer. , it may be any one selected from the group consisting of skin cancer, sinus tumor, uterine cancer, head cancer, cervical cancer, gallbladder cancer, oral cancer, anal cancer, colon cancer, and central nervous system tumor.
본 명세서에서 사용된 용어, "예방"은 상기 약학 조성물의 투여에 의해 암의 발생을 억제하거나 그의 발병을 지연시키는 모든 행위를 말한다. 상기 용어 "치료"는 상기 약학 조성물의 투여에 의해 암의 증세가 호전되거나 이롭게 변경하는 모든 행위를 말한다.As used herein, the term “prevention” refers to all actions that inhibit the occurrence of cancer or delay its onset by administering the pharmaceutical composition. The term “treatment” refers to any action that improves or beneficially changes cancer symptoms by administering the pharmaceutical composition.
본 발명의 암 예방 또는 치료용 약학 조성물에서 상기 항-IGSF1 항체 또는 이의 단편 및 항-PD-1 항체는 항암 활성을 나타낼 수 있는 한, 용도, 제형, 배합 목적 등에 따라 임의의 양(유효량)으로 포함될 수 있다. 여기서, "유효량"이란 항암 효과를 유도할 수 있는 유효성분의 양을 말한다. 이러한 유효량은 당업자의 통상의 능력 범위 내에서 실험적으로 결정될 수 있다. 본 발명의 약학 조성물은 유효성분으로서 상기 항체를 조성물의 총 중량을 기준으로 약 0.1 중량% 내지 약 90 중량%, 구체적으로 약 0.5 중량% 내지 약 75 중량%, 보다 구체적으로 약 1 중량% 내지 약 50 중량%로 함유할 수 있다.In the pharmaceutical composition for preventing or treating cancer of the present invention, the anti-IGSF1 antibody or fragment thereof and the anti-PD-1 antibody are used in any amount (effective amount) depending on the use, formulation, purpose of formulation, etc., as long as they can exhibit anticancer activity. may be included. Here, “effective amount” refers to the amount of an active ingredient that can induce an anticancer effect. Such effective amounts can be determined experimentally within the scope of the ordinary ability of those skilled in the art. The pharmaceutical composition of the present invention contains the antibody as an active ingredient in an amount of about 0.1% to about 90% by weight, specifically about 0.5% by weight to about 75% by weight, and more specifically about 1% by weight to about 1% by weight, based on the total weight of the composition. It can be contained at 50% by weight.
생체이용률과 같은 약동학적 파라미터(pharmacokinetic parameters) 및 클리어런스율(clearance rate)과 같은 기본적인 파라미터(underlying parameters)도 효능에 영향을 줄 수 있다. 따라서, "향상된 효능"(예를 들어, 효능의 개선)은 향상된 약동학적 파라미터 및 향상된 효능에 기인할 수 있으며, 시험 동물 또는 인간 대상체에서 클리어런스율 및 종양 성장을 비교하거나, 생존, 재발율 또는 질병이 없는 상태에서 생존과 같은 파라미터를 비교하여 측정될 수 있다.Pharmacokinetic parameters such as bioavailability and underlying parameters such as clearance rate may also affect efficacy. Accordingly, “enhanced efficacy” (e.g., improvement in efficacy) may be due to improved pharmacokinetic parameters and improved efficacy, compared to clearance rates and tumor growth in test animals or human subjects, or to survival, recurrence rates, or disease progression. It can be measured by comparing parameters such as survival in the absence of it.
본 명세서에서 사용된 용어, "효능(efficacy)"은 1년, 5년, 또는 10년과 같이 일정 기간에 걸쳐 생존 또는 질병이 없는 상태에서 생존(disease-free survival)과 같은 하나 이상의 파라미터에 의해 결정될 수 있다. 뿐만 아니라, 상기 파라미터는 개체에서 적어도 하나의 종양의 크기가 억제되는 것을 포함할 수 있다.As used herein, the term "efficacy" means survival over a period of time, such as 1 year, 5 years, or 10 years, or disease-free survival. can be decided. Additionally, the parameter may include that the size of at least one tumor in the subject is suppressed.
본 발명의 약학 조성물은, 통상적인 방법에 따라 제제로 배합되는 통상적이고 무독성인 약학적으로 허용가능한 담체를 포함할 수 있다.The pharmaceutical composition of the present invention may contain a conventional, non-toxic pharmaceutically acceptable carrier that is formulated into a preparation according to a conventional method.
상기 약학적으로 허용가능한 담체는 환자에게 전달하기에 적절한 비-독성 물질이면 어떠한 담체라도 가능하다. 증류수, 알코올, 지방, 왁스 및 비활성 고체가 담체로 포함될 수 있다. 약물학적으로 허용되는 애쥬번트(완충제, 분산제) 또한 약물학적 조성물에 포함될 수 있다.The pharmaceutically acceptable carrier can be any carrier that is a non-toxic material suitable for delivery to a patient. Distilled water, alcohol, fats, waxes and inert solids may be included as carriers. Pharmacologically acceptable adjuvants (buffers, dispersants) may also be included in the pharmacological composition.
본 명세서에서 사용된 용어, "약학적으로 허용가능한 담체"란 생물체를 자극하지 않고 투여 화합물의 생물학적 활성 및 특성을 저해하지 않는 담체 또는 희석제를 말한다. 액상 용액으로 제제화되는 조성물에 있어서 허용되는 약학적 담체로는, 멸균 및 생체에 적합한 것으로서, 식염수, 멸균수, 링거액, 완충 식염수, 알부민 주사용액, 덱스트로즈 용액, 말토덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 한 성분 이상을 혼합하여 사용할 수 있으며, 필요에 따라 감미제, 용해 보조제, 습윤제, 유화제, 등장화제, 흡수제, 항산화제, 보존제, 활택제, 충전제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다.As used herein, the term “pharmaceutically acceptable carrier” refers to a carrier or diluent that does not irritate living organisms and does not inhibit the biological activity and properties of the administered compound. Acceptable pharmaceutical carriers in compositions formulated as liquid solutions include those that are sterile and biocompatible, such as saline solution, sterile water, Ringer's solution, buffered saline solution, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and One or more of these ingredients can be mixed and used, and other common additives such as sweeteners, solubilizers, wetting agents, emulsifiers, isotonic agents, absorbents, antioxidants, preservatives, lubricants, fillers, buffers, and bacteriostatic agents are added as needed. can do.
본 발명의 조성물은 비경구 투여(예컨대, 근육내, 정맥내 또는 피하 주사)를 위한 다양한 제형으로 제조될 수 있다. 본 발명의 약학 조성물이 비경구용 제형으로 제조될 경우, 적합한 담체와 함께 당업계에 공지된 방법에 따라 주사제, 경피 투여제, 비강 흡입제 및 좌제의 형태로 제제화될 수 있다. 주사용 제제에는 멸균된 수용액제, 비수성용제, 현탁제, 유제, 동결건조 제제 및 좌제가 포함된다. 비수성용제, 현탁제로는 프로필렌글리콜, 폴리에틸렌글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔, 마크로골, 트윈61, 카카오지, 라우린지, 글리세로젤라틴 등이 사용될 수 있다. 한편, 주사제에는 용해제, 등장화제, 현탁화제, 유화제, 안정화제, 방부제 등과 같은 종래의 첨가제가 포함될 수 있다.The compositions of the present invention can be prepared in a variety of formulations for parenteral administration (e.g., intramuscular, intravenous, or subcutaneous injection). When the pharmaceutical composition of the present invention is prepared as a parenteral formulation, it can be formulated in the form of injections, transdermal administration, nasal inhalation, and suppositories along with a suitable carrier according to methods known in the art. Injectable preparations include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized preparations, and suppositories. Non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate. As a base for suppositories, Withepsol, Macrogol, Tween 61, cacao, laurin, glycerogelatin, etc. can be used. Meanwhile, injectables may contain conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifiers, stabilizers, preservatives, etc.
약제학적 조성물의 제제화와 관련하여서는 당업계에 공지되어 있으며, 구체적으로 문헌[Remington's Pharmaceutical Sciences(19th ed., 1995)] 등을 참조할 수 있다. 상기 문헌은 본 명세서의 일부로서 간주된다.Regarding the formulation of pharmaceutical compositions, it is known in the art, and specifically, references can be made to the literature [Remington's Pharmaceutical Sciences (19th ed., 1995)]. The above documents are considered part of this specification.
본 발명의 항체 또는 조성물은 치료학적으로 유효한 양 또는 약학적으로 유효한 양으로 환자에 투여될 수 있다.The antibody or composition of the present invention can be administered to a patient in a therapeutically effective amount or a pharmaceutically effective amount.
본 명세서에서 사용된 용어 "투여"란, 적절한 방법으로 개체에게 소정의 물질을 도입하는 것을 의미하며, 상기 조성물의 투여 경로는 목적 조직에 도달할 수 있는 한 어떠한 일반적인 경로를 통하여 투여될 수 있다. 복강내 투여, 정맥내 투여, 근육내 투여, 피하 투여, 피내 투여, 국소 투여, 비내 투여, 직장내 투여될 수 있으나, 이에 한정되지 않는다.The term "administration" used herein means introducing a predetermined substance into an individual by an appropriate method, and the composition may be administered through any general route as long as it can reach the target tissue. It may be administered intraperitoneally, intravenously, intramuscularly, subcutaneously, intradermally, locally, intranasally, or rectally, but is not limited thereto.
여기서, "치료학적으로 유효한 양" 또는 "약학적으로 유효한 양"이란 대상 질환을 예방 또는 치료하는데 유효한 화합물 또는 조성물의 양으로서, 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분하며 부작용을 일으키지 않을 정도의 양을 의미한다. 상기 유효량의 수준은 환자의 건강 상태, 질환의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 방법, 투여 시간, 투여 경로 및 배출 비율, 치료 기간, 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. 일 구체예에서, 치료학적으로 유효한 양은 암을 치료하는데 효과적인 약물의 양을 의미한다.Here, “therapeutically effective amount” or “pharmacologically effective amount” refers to the amount of a compound or composition effective in preventing or treating the target disease, which is sufficient to treat the disease with a reasonable benefit/risk ratio applicable to medical treatment. It refers to an amount that does not cause side effects. The level of the effective amount is determined by factors including the patient's health status, type and severity of the disease, activity of the drug, sensitivity to the drug, method of administration, time of administration, route of administration and excretion rate, treatment period, drugs combined or used simultaneously, and It may be determined based on other factors well known in the medical field. In one embodiment, a therapeutically effective amount refers to an amount of drug that is effective in treating cancer.
구체적으로, 본 발명의 조성물에서 항체의 유효량은 환자의 나이, 성별, 체중에 따라 달라질 수 있으며, 일반적으로는 체중 kg 당 약 0.1 mg 내지 약 1,000 mg, 또는 약 5 mg 내지 약 200 mg을 매일, 격일 또는 2주 내지 3주 간격으로 투여하거나, 1일 1회 내지 3회로 나누어 투여할 수 있다. 그러나, 투여 경로, 질병의 중증도, 성별, 체중, 연령 등에 따라서 증감될 수 있으므로, 본 발명의 범위는 이에 한정되지 않는다.Specifically, the effective amount of antibody in the composition of the present invention may vary depending on the patient's age, gender, and weight, and is generally about 0.1 mg to about 1,000 mg, or about 5 mg to about 200 mg per kg of body weight daily. It can be administered every other day or every 2 to 3 weeks, or it can be administered once to three times a day. However, since it may increase or decrease depending on the route of administration, severity of disease, gender, weight, age, etc., the scope of the present invention is not limited thereto.
상기 용어 "개체"란 본 발명의 조성물이 적용(처방)될 수 있는 대상을 의미하며, 인간을 포함한 쥐, 생쥐, 가축 등의 포유동물일 수 있다. 바람직하게는, 인간일 수 있으나 이에 제한되지 않는다.The term “individual” refers to an object to which the composition of the present invention can be applied (prescribed), and may be a mammal such as a rat, mouse, or livestock, including humans. Preferably, it may be a human, but is not limited thereto.
본 발명의 항체 또는 이를 포함하는 약학 조성물은 개별 치료제로 투여하거나 다른 치료제와 병용하여 투여될 수 있고, 종래의 치료제와 순차적으로 또는 동시에 투여될 수 있으며, 단일 또는 다중 투여될 수 있다. 이때, 상기 다른 치료제는 항암 활성의 상승, 보강을 위하여 이미 안전성이 검증되고 항암 활성을 갖는 것으로 공지된 임의의 화합물이나 천연 추출물을 추가로 포함할 수 있다.The antibody of the present invention or a pharmaceutical composition containing the same may be administered as an individual therapeutic agent or in combination with other therapeutic agents, may be administered sequentially or simultaneously with conventional therapeutic agents, and may be administered singly or multiple times. At this time, the other therapeutic agent may additionally include any compound or natural extract whose safety has already been verified and which is known to have anticancer activity in order to increase or reinforce anticancer activity.
상기한 요소들을 모두 고려하여 최소한의 부작용으로 또는 부작용 없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 이는 당업자에 의해 용이하게 결정될 수 있다.Taking all of the above factors into consideration, it is important to administer an amount that can achieve maximum effect with minimal or no side effects, and this can be easily determined by a person skilled in the art.
본 발명의 또 다른 측면은, 암 예방 또는 치료용 약제를 제조하기 위한 본 발명의 항-IGSF1 항체 또는 이의 단편 및 항-PD-1 항체를 유효성분으로 포함하는 약학 조성물의 용도를 제공한다. 여기서, 항-IGSF1 항체 및 이의 단편, 항-PD-1 항체, 암, 예방 및 치료는 상술한 바와 동일하다.Another aspect of the present invention provides the use of a pharmaceutical composition comprising the anti-IGSF1 antibody or fragment thereof and the anti-PD-1 antibody of the present invention as active ingredients for preparing a drug for preventing or treating cancer. Here, anti-IGSF1 antibody and fragment thereof, anti-PD-1 antibody, cancer, prevention and treatment are the same as described above.
본 발명의 또 다른 측면은, 본 발명의 항-IGSF1 항체 또는 이의 단편 및 항-PD-1 항체를 유효성분으로 포함하는 약학 조성물의 암 예방 또는 치료 용도를 제공한다. 여기서 항-IGSF1 항체 및 이의 단편, 항-PD-1 항체, 암, 예방 및 치료는 상술한 바와 동일하다.Another aspect of the present invention provides a use for cancer prevention or treatment of a pharmaceutical composition comprising the anti-IGSF1 antibody or fragment thereof and the anti-PD-1 antibody of the present invention as active ingredients. Here, anti-IGSF1 antibody and fragment thereof, anti-PD-1 antibody, cancer, prevention and treatment are the same as described above.
본 발명의 또 다른 측면은, 항-IGSF1 항체 또는 이의 단편 및 항-PD-1 항체를 유효성분으로 포함하는 약학 조성물을 개체에 투여하는 단계를 포함하는 암 예방 또는 치료 방법을 제공한다. 여기서 항-IGSF1 항체 및 이의 단편, 항-PD-1 항체, 암, 투여, 치료 및 예방은 상술한 바와 동일하다.Another aspect of the present invention provides a method for preventing or treating cancer, comprising administering to a subject a pharmaceutical composition containing an anti-IGSF1 antibody or fragment thereof and an anti-PD-1 antibody as active ingredients. Here, anti-IGSF1 antibody and fragment thereof, anti-PD-1 antibody, cancer, administration, treatment and prevention are the same as described above.
상기 개체는 포유동물일 수 있으며, 바람직하게는 인간일 수 있다. 또한, 상기 개체는 암 환자이거나 암을 앓을 가능성이 큰 개체일 수 있다.The subject may be a mammal, preferably a human. Additionally, the individual may be a cancer patient or an individual at a high risk of suffering from cancer.
상기 약학 조성물의 투여 경로, 투여량 및 투여 횟수는 환자의 상태 및 부작용의 유무에 따라 다양한 방법 및 양으로 대상에게 투여될 수 있고, 최적의 투여 방법, 투여량 및 투여 횟수는 통상의 기술자가 적절한 범위로 선택할 수 있다. 또한, 상기 항-IGSF1 항체 또는 이의 단편은 암에 대하여 치료 효과가 공지된 다른 약물 또는 생화학적 활성물질과 병용하여 투여되거나, 다른 약물과의 조합 제제 형태로 제형화될 수 있다.The administration route, dosage, and frequency of administration of the pharmaceutical composition may be administered to the subject in various ways and amounts depending on the patient's condition and the presence or absence of side effects, and the optimal administration method, dosage, and frequency of administration may be determined by a person skilled in the art. You can select by range. Additionally, the anti-IGSF1 antibody or fragment thereof may be administered in combination with other drugs or biochemically active substances known to have therapeutic effects on cancer, or may be formulated in the form of a combination preparation with other drugs.
이하, 본 발명을 하기 실시예에 의해 상세히 설명한다. 단, 하기 실시예는 본 발명을 예시하기 위한 것일 뿐, 본 발명이 이들에 의해 제한되는 것은 아니다.Hereinafter, the present invention will be explained in detail by the following examples. However, the following examples are only for illustrating the present invention, and the present invention is not limited thereto.
실시예 1. 항-IGSF1 항체 제작Example 1. Production of anti-IGSF1 antibody
실시예 1.1. IGSF1 항원 발현 및 정제Example 1.1. IGSF1 antigen expression and purification
Jurkat 세포 cDNA 라이브러리에서 PCR 방법을 통해 IGSF1의 세포외 도메인만을 증폭한 후, N293F 벡터(와이바이오로직스(주))를 이용하여 카복시 말단(C 말단)에 인간 Fc(fragment crystallizable region) 및 His-태그(His-tag)를 융합시켜 IGSF1 단백질 발현 벡터를 제작하였다. HEK293F 세포에 제작한 IGSF1 발현 벡터를 형질감염(transfection)시킨 후, 1 mM 발프로산(valporic acid, valproate)을 첨가한 배지에서 6일 동안 배양하였다. 이후, protein A 아가로오즈(agarose)를 사용하여 IGSF1 세포외 도메인을 1차 정제한 후, Superdex 200 겔 여과 크로마토그래피를 이용하여 IGSF1 세포외 도메인을 2차 정제한 후, 항체 선별에 사용하였다.After amplifying only the extracellular domain of IGSF1 through PCR from the Jurkat cell cDNA library, a human Fc (fragment crystallizable region) and a His-tag were attached to the carboxy terminus (C terminus) using the N293F vector (Y Biologics Co., Ltd.). (His-tag) was fused to create an IGSF1 protein expression vector. HEK293F cells were transfected with the prepared IGSF1 expression vector, and then cultured for 6 days in medium supplemented with 1 mM valporic acid (valproate). Afterwards, the IGSF1 extracellular domain was first purified using protein A agarose, and then the IGSF1 extracellular domain was secondarily purified using Superdex 200 gel filtration chromatography, and then used for antibody selection.
실시예 1.2. IGSF1 인간 항체의 선별Example 1.2. Selection of IGSF1 human antibodies
IGSF1 항원을 코팅하고 블로킹한 후, 준비된 인간 항체 라이브러리 파아지(와이바이오로직스(주))를 이용하여 바이오패닝(와이바이오로직스(주))을 진행하여 항원에 특이적으로 결합한 파아지들만 용출(elution)하였다. 첫 번째 라운드의 바이오패닝에서 증폭된 파아지를 이용하여 두 번째 및 세 번째 라운드의 바이오패닝을 수행하였다. 각 라운드의 바이오패닝을 통해 얻어진 양성 파아지 항체 풀(pool)에 대한 항원과의 특이성을 확인하고자 ELISA를 수행하였다. 또한, 세 번째 라운드를 통해 얻어진 파아지 풀에 항-IGSF1 항체가 인리치(enrich)된 것을 확인하였다. 각 폴리 파아지 ELISA에서 결합능이 큰 세 번째 라운드의 패닝으로부터 수 백 종의 모노클론들을 선별하였고, 이를 이용하여 ELISA 분석을 통해 IGSF1에 특이적으로 결합하는지 여부를 확인하여 예비 항체 클론을 확보하였다. 선별된 예비 항체 클론들에 대해 DNA 염기서열 분석을 통해 염기서열이 서로 다른 파아지를 99종 선별하였다. 선별된 99종의 양성 파아지 클론들은 항원인 IGSF1에는 강하게 결합하였으나, 다른 항원들에 대해서는 결합하지 않는 것을 확인하였다. 상기 방법을 통해 추가적으로 다양한 다른 항원을 이용하여 IGSF1 항원에 특이성을 나타내는 항체를 선별한 결과 총 95종을 선별할 수 있었다.After coating and blocking the IGSF1 antigen, biopanning (Y Biologics Co., Ltd.) was performed using the prepared human antibody library phage (Y Biologics Co., Ltd.) to elute only phages that specifically bound to the antigen. did. The second and third rounds of biopanning were performed using the phage amplified in the first round of biopanning. ELISA was performed to confirm the antigen specificity of the positive phage antibody pool obtained through each round of biopanning. In addition, it was confirmed that anti-IGSF1 antibody was enriched in the phage pool obtained through the third round. In each polyphage ELISA, hundreds of monoclones with high binding capacity were selected from the third round of panning, and preliminary antibody clones were secured by confirming whether they specifically bind to IGSF1 through ELISA analysis. For the selected preliminary antibody clones, 99 phages with different base sequences were selected through DNA sequence analysis. It was confirmed that the 99 selected positive phage clones strongly bound to the antigen, IGSF1, but did not bind to other antigens. Through the above method, a total of 95 types were selected as a result of selecting antibodies showing specificity for the IGSF1 antigen using various other antigens.
실시예 1.3. IGSF1 항원에 대한 특이성 확인Example 1.3. Confirmation of specificity for IGSF1 antigen
선별한 항체에 대해 IGSF1을 비롯한 다른 항원들에 대한 특이성을 ELISA 방법을 통해 비교 분석하였다. 대조군 항원들인 mFc, hRAGE-Fc, CD58-Fc, ITGA6-Fc 등 다양한 종류의 불특정 항원에 대해서 파아지 클론들의 결합 여부를 확인하였다. 이와 같은 확인을 통해 수득한 항체들에 대해 파아지에서 IgG whole 벡터로 전환하였다. 전환된 95개 클론의 중쇄 서열 및 경쇄 서열이 파아지 항체의 서열과 일치하는 것을 확인하였다. 수득된 항체 중 가장 최적화된 항체를 선별하고, 이를 "WM-A1-3389"라 명명하였다. WM-A1-3389 항체의 CDR 서열은 하기 표 1과 같다.The specificity of the selected antibodies to other antigens, including IGSF1, was compared and analyzed using ELISA. The binding of phage clones to various types of unspecified antigens, including control antigens mFc, hRAGE-Fc, CD58-Fc, and ITGA6-Fc, was confirmed. The antibodies obtained through this confirmation were converted from phage to whole IgG vectors. It was confirmed that the heavy chain and light chain sequences of the converted 95 clones matched the sequences of the phage antibodies. Among the obtained antibodies, the most optimized antibody was selected and named “WM-A1-3389”. The CDR sequence of the WM-A1-3389 antibody is shown in Table 1 below.
WM-A1-3389WM-A1-3389
CDRCDR 아미노산 서열amino acid sequence 서열번호sequence number
H-CDR1H-CDR1 GGTFSTYAGGTFSTYA 1One
H-CDR2H-CDR2 IIPFVGTVIIPFVGTV 22
H-CDR3H-CDR3 VRDGGRSYFDSVRDGRSYFDS 33
L-CDR1L-CDR1 TSNIGSNLTSNIGSNL 44
L-CDR2L-CDR2 DNHDNH 55
L-CDR3L-CDR3 VAWDDSLNGYVVAWDDSLNGYV 66
실시예 1.4. WM-A1-3389 항체 생산Example 1.4. WM-A1-3389 antibody production
WM-A1-3389 항체를 생산하기 위하여, 중쇄(서열번호 21)를 암호화하는 폴리뉴클레오티드(서열번호 23)를 N293F 벡터(와이바이오로직스(주))에 적재하였다(이하, HC DNA). 또한, 경쇄(서열번호 22)를 암호화하는 폴리뉴클레오티드(서열번호 24)를 N293F 벡터(와이바이오로직스(주))에 적재하였다(이하, LC DNA). 상기 벡터를 세포에 형질전환 시킨 후, WM-A1-3389 항체를 수득 및 정제하였다. 정제된 단백질은 SDS-PAGE를 통해 확인하였다.To produce the WM-A1-3389 antibody, a polynucleotide (SEQ ID NO: 23) encoding the heavy chain (SEQ ID NO: 21) was loaded into the N293F vector (Y Biologics Co., Ltd.) (hereinafter referred to as HC DNA). In addition, a polynucleotide (SEQ ID NO: 24) encoding the light chain (SEQ ID NO: 22) was loaded into the N293F vector (Y Biologics Co., Ltd.) (hereinafter referred to as LC DNA). After the vector was transformed into cells, WM-A1-3389 antibody was obtained and purified. Purified proteins were confirmed through SDS-PAGE.
아미노산 서열amino acid sequence 서열번호sequence number
중쇄
(heavy chain)
heavy chain
(heavy chain)
QVQLVQSGAEVKRPGSSVKVSCKASGGTFSTYAISWVRQAPGQGLEWMGRIIPFVGTVDYAQKFQDRVTITADKSTNTAYMELSSLRSEDTAVYYCVRDGGRSYFDSWGPGILVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKQVQLVQSGAEVKRPGSSVKVSCKASGGTFSTYAISWVRQAPGQGLEWMGRIIPFVGTVDYAQKFQDRVTITADKSTNTAYMELSSLRSEDTAVYYCVRDGGRSYFDSWGPGILVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNH KPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK 2121
경쇄
(light chain)
light chain
(light chain)
QFVLTQPPSVSAAPGQDVIISCSGNTSNIGSNLVSWFQQFPETAPKLLIYDNHKRPSGISDRFSGTKSGTSASLAISGLQSEDEADYYCVAWDDSLNGYVFGTGTKVTVLRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECQFVLTQPPSVSAAPGQDVIISCSGNTSNIGSNLVSWFQQFPETAPKLLIYDNHKRPSGISDRFSGTKSGTSASLAISGLQSEDEADYYCVAWDDSLNGYVFGTGTKVTVLRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKH KVYACEVTHQGLSSPVTKSFNRGEC 2222
실시예 2. IGSF1 발현과 종양 침윤 림프구의 관련성 분석Example 2. Analysis of the relationship between IGSF1 expression and tumor infiltrating lymphocytes
실시예 2.1. IGSF1 과발현 세포주 제작Example 2.1. Construction of IGSF1 overexpressing cell lines
IGSF1을 인간 폐암 세포주인 NCI-H292 또는 인간 배아 신장 세포주 HEK293E 세포에 과발현시켜, IGSF1이 과발현된 세포주를 제작하였다(도 1). 이때, MOCK는 IGSF1 발현이 없는 대조군이다.IGSF1 was overexpressed in NCI-H292, a human lung cancer cell line, or HEK293E, a human embryonic kidney cell line, to create a cell line in which IGSF1 was overexpressed (Figure 1). At this time, MOCK is a control group without IGSF1 expression.
구체적으로, IGSF1을 코딩하는 폴리뉴클레오티드가 포함된 발현 벡터(OriGene Technologies, Inc., Cat No.RC209621)를 인간 폐암 세포주 NCI-H292 세포 또는 인간 배아 신장 세포주 HEK293F 세포에 형질감염(transfection)시켰다. 이후, G418(neomycin)이 포함된 배지에서 배양하여 형질감염된 세포를 선별하였다. 선별된 클론들에 대해서 IGSF1 발현 수준을 확인하여, 가장 높은 IGSF1 발현을 보인 클론을 선택하여 실험에 사용하였다. MOCK는 IGSF1을 코딩하는 폴리뉴클레오티드가 적재되지 않은 공벡터를 의미한다.Specifically, an expression vector containing a polynucleotide encoding IGSF1 (OriGene Technologies, Inc., Cat No.RC209621) was transfected into human lung cancer cell line NCI-H292 cells or human embryonic kidney cell line HEK293F cells. Afterwards, the transfected cells were selected by culturing them in a medium containing G418 (neomycin). The IGSF1 expression level was confirmed for the selected clones, and the clone showing the highest IGSF1 expression was selected and used in the experiment. MOCK refers to an empty vector in which the polynucleotide encoding IGSF1 is not loaded.
실시예 2.2. 폐암 세포주 스페로이드에서 IGSF1 발현과 종양 침윤 림프구의 관련성 분석Example 2.2. Analysis of the association between IGSF1 expression and tumor-infiltrating lymphocytes in lung cancer cell line spheroids
폐암에서 IGSF1 발현과 종양 침윤 림프구(tumor-infiltrating lymphocytes, TIL)와의 상관관계를 세포 수준에서 확인하기 위하여, IGSF1을 과발현시킨 폐암 세포를 이용한 스페로이드에서 종양 침윤 림프구를 확인하였다.To confirm the correlation between IGSF1 expression and tumor-infiltrating lymphocytes (TIL) in lung cancer at the cellular level, tumor-infiltrating lymphocytes were identified in spheroids using lung cancer cells that overexpressed IGSF1.
먼저, 폐암 세포 스페로이드를 제작하기 위하여, NCI-H292 IGSF1 O/E 세포 및 NCI-H292 MOCK 세포를 각각 2×104 세포/웰(well)이 되도록 U-Bottom 96-웰 플레이트(Nunc, 174925)에 접종(seeding)하여 37℃, CO2 인큐베이터에서 72시간 동안 배양하였다. 말초혈액 단핵세포(peripheral blood mononuclear cell, PBMC)는 1,200 rpm에서 10분간 원심분리하여 상층액을 제거하고 PBS에 재현탁하여 준비하였다. CFSE(carboxyfluorescein succinimidyl ester, Invitrogen, C34554)에 DMSO 18 ㎕를 넣어 5 mM 농도로 만들고, PBS에 1 mM이 되도록 희석하였다. 준비한 말초혈액 단핵세포 1×106 세포/㎖ 당 1 mM CFSE 용액을 1 ㎕씩 첨가한 뒤, 37℃, CO2 인큐베이터에서 10분간 염색하였다. 이후, PBS 양의 5배 분량의 FBS(fetal bovine serum)가 포함된 배지를 염색한 말초혈액 단핵세포(PBMC)가 포함된 용액에 첨가하였다. 그 후, 37℃, CO2 인큐베이터에서 5분간 반응시켰다. 1,200 rpm에서 10분간 원심분리하여 상층액을 제거한 후, 염색된 말초혈액 단핵세포를 배지에 재현탁시켜 실험에 사용할 말초혈액 단핵세포를 준비하였다.First, to produce lung cancer cell spheroids, NCI-H292 IGSF1 O/E cells and NCI-H292 MOCK cells were each plated at 2×10 4 cells/well in a U-Bottom 96-well plate (Nunc, 174925). ) were seeded and cultured in a CO 2 incubator at 37°C for 72 hours. Peripheral blood mononuclear cells (PBMC) were prepared by centrifuging at 1,200 rpm for 10 minutes to remove the supernatant and resuspending in PBS. 18 ㎕ of DMSO was added to CFSE (carboxyfluorescein succinimidyl ester, Invitrogen, C34554) to make a concentration of 5 mM, and then diluted to 1 mM in PBS. 1 μl of 1 mM CFSE solution was added per 1 × 10 6 cells/ml of prepared peripheral blood mononuclear cells, and then stained for 10 minutes in a CO 2 incubator at 37°C. Afterwards, medium containing FBS (fetal bovine serum) in an amount five times the amount of PBS was added to the solution containing stained peripheral blood mononuclear cells (PBMC). Afterwards, the reaction was performed at 37°C in a CO 2 incubator for 5 minutes. After removing the supernatant by centrifugation at 1,200 rpm for 10 minutes, the stained peripheral blood mononuclear cells were resuspended in medium to prepare peripheral blood mononuclear cells to be used in the experiment.
그 후, 형성된 스페로이드를 2웰씩 초저부착 96-웰 플레이트(Corning, CLS3474)에 옮긴 후, CFSE로 염색된 말초혈액 단핵세포를 1×105 세포/웰이 되도록 접종하여 37℃, CO2 인큐베이터에서 24시간 동안 공동 배양하였다. 종양 침윤 림프구(TIL)는 형광 현미경을 통해 관찰하였다. 이때, NCI-H292 MOCK 세포는 IGSF1 과발현 세포에 대한 대조군으로 사용하였다.Afterwards, the formed spheroids were transferred to an ultra-low attachment 96-well plate (Corning, CLS3474), 2 wells at a time, and CFSE-stained peripheral blood mononuclear cells were inoculated at 1×10 5 cells/well and incubated at 37°C in a CO 2 incubator. were co-cultured for 24 hours. Tumor-infiltrating lymphocytes (TIL) were observed through fluorescence microscopy. At this time, NCI-H292 MOCK cells were used as a control for IGSF1 overexpressing cells.
그 결과, IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E) 스페로이드에서 대조군(NCI-H292 MOCK)에 비해 종양 침윤 림프구(TIL)가 감소하는 것을 확인하였다(도 2).As a result, it was confirmed that tumor infiltrating lymphocytes (TIL) were reduced in IGSF1-overexpressing human lung cancer cell (NCI-H292 IGSF1 O/E) spheroids compared to the control group (NCI-H292 MOCK) (Figure 2).
실시예 2.3. 폐암 세포주를 이식한 인간화 마우스의 종양 조직에서 IGSF1 발현과 종양 침윤 림프구의 관련성 분석Example 2.3. Analysis of the relationship between IGSF1 expression and tumor-infiltrating lymphocytes in tumor tissues of humanized mice transplanted with lung cancer cell lines.
폐암에서 IGSF1의 발현과 종양 침윤 림프구(TIL)와의 상관관계를 in vivo 수준에서 확인하기 위하여, IGSF1을 과발현시킨 인간 폐암 세포를 이식한 인간화 마우스의 종양 조직에서 종양 침윤 림프구를 확인하였다.To confirm the correlation between the expression of IGSF1 and tumor infiltrating lymphocytes (TIL) in lung cancer at the in vivo level, tumor infiltrating lymphocytes were identified in the tumor tissues of humanized mice transplanted with human lung cancer cells that overexpressed IGSF1.
구체적으로, 인간 말초혈액 단핵세포(PBMC)를 이식한 NSG 마우스(SID(NSGA) mice, F)에 IGSF1 과발현 인간 폐암 세포주인 NCI-H292 IGSF1 O/E 세포 또는 대조군인 NCI-H292 MOCK 세포를 한 마리당 5×106 세포로 이식 후 17일째에 마우스의 종양 부분을 채취하였다.Specifically, NSG mice (SID (NSGA) mice, F) transplanted with human peripheral blood mononuclear cells (PBMC) were treated with NCI-H292 IGSF1 O/E cells, a human lung cancer cell line that overexpresses IGSF1, or NCI-H292 MOCK cells as a control group. Tumor parts of mice were collected 17 days after transplantation with 5×10 6 cells per mouse.
채취된 종양에서 각각 인간 말초혈액 단핵세포를 분리하여 FACS 분석하고, 종양 조직의 면역조직화학염색(immunohistochemistry)을 통해 종양 조직 내의 종양 침윤 림프구 및 IGSF1 발현량을 확인하였다. 종양에 침윤된 인간 말초혈액 단핵세포를 확인하기 위하여, 먼저 종양 조직에 collagenase B(Roche, cat. #11088815001)를 처리하고 37℃에서 2시간 이상 반응시켜 종양 조직을 분해시켰다. 종양 조직이 완전히 세포로 분해되면 l ㎖ 파이펫으로 파이펫팅하여 단일 세포로 분리하였다.Human peripheral blood mononuclear cells were isolated from each collected tumor, analyzed by FACS, and the level of tumor infiltrating lymphocytes and IGSF1 expression within the tumor tissue was confirmed through immunohistochemistry. To identify human peripheral blood mononuclear cells infiltrating the tumor, the tumor tissue was first treated with collagenase B (Roche, cat. #11088815001) and reacted at 37°C for more than 2 hours to decompose the tumor tissue. When the tumor tissue was completely decomposed into cells, it was separated into single cells by pipetting with a 1 ml pipette.
분리된 단일 세포를 50 ㎖ 튜브(SPL, cat. #50050)로 옮긴 뒤, PBS 20 ㎖로 세척하였다. 이후, 1,200 rpm에서 3분간 원심분리하고 상층액을 제거하였다. 남아있는 세포에 DNase I(Roche, cat. #11284932001)을 처리하여 37℃에서 20분간 반응시켰다. 반응 후, PBS를 20 ㎖ 첨가하고, 1,200 rpm에서 3분간 원심분리하여 상층액을 제거하였다. 남아있는 세포에 0.25% Trysin/EDTA(GIBCO, cat. #15400-054)를 처리하였다. 세포를 잘 혼합한 후, 새로운 50 ㎖ 튜브 상에 셀 스트레이너(cell strainer; SPL, cat. #93070)를 배치하고 세포를 걸러주었다. 걸러진 세포가 있는 튜브에 20 ㎖ PBS를 첨가하고 잘 혼합하였다. 이후, 1,200 rpm에서 3분간 원심분리하고 상층액을 제거한 후, 남아있는 세포에 Stain Buffer(BD, cat. #554656) 1 ㎖을 넣고 세척하였다.The isolated single cells were transferred to a 50 ml tube (SPL, cat. #50050) and washed with 20 ml of PBS. Afterwards, it was centrifuged at 1,200 rpm for 3 minutes and the supernatant was removed. The remaining cells were treated with DNase I (Roche, cat. #11284932001) and incubated at 37°C for 20 minutes. After the reaction, 20 ml of PBS was added, and the supernatant was removed by centrifugation at 1,200 rpm for 3 minutes. The remaining cells were treated with 0.25% Trysin/EDTA (GIBCO, cat. #15400-054). After mixing the cells well, a cell strainer (SPL, cat. #93070) was placed on a new 50 ml tube and the cells were filtered. 20 mL of PBS was added to the tube containing the filtered cells and mixed well. Afterwards, centrifugation was performed at 1,200 rpm for 3 minutes and the supernatant was removed. 1 ml of Stain Buffer (BD, cat. #554656) was added to the remaining cells and washed.
분리한 세포의 비특이성 항체 반응을 차단하기 위해, Human Fc block(BD, cat. #564219)을 2 ㎍씩 첨가하고 10분간 상온에서 반응시켰다. 반응 후, 항-인간 CD45(BD, cat. #564357) 항체를 첨가하고, 4℃에서 30분간 암(dark) 상태로 반응시켰다. 반응 후, Stain Buffer 1 ㎖을 첨가하여 세척하였다. 이후, 1,200 rpm에서 3분간 원심분리하고 상층액을 제거하여 세포를 수집하였다. 수집된 세포에 Stain Buffer를 200 ㎕씩 첨가하여 BD LSRFortessaTM Flow Cytometer로 분석하였다(도 3).To block the non-specific antibody response of the isolated cells, 2 μg of Human Fc block (BD, cat. #564219) was added and reacted at room temperature for 10 minutes. After reaction, anti-human CD45 (BD, cat. #564357) antibody was added and reaction was performed in a dark state at 4°C for 30 minutes. After reaction, 1 ml of Stain Buffer was added and washed. Afterwards, the cells were collected by centrifuging at 1,200 rpm for 3 minutes and removing the supernatant. 200 ㎕ of Stain Buffer was added to the collected cells and analyzed with a BD LSRFortessa TM Flow Cytometer (Figure 3).
또한, 면역조직화학염색법을 통해 종양 내 발현된 종양 림프구 분포 및 IGSF1의 발현을 확인하였다. 구체적으로, 인간 말초혈액 단핵세포(PBMC)를 이식한 NSG 마우스(SID(NSGA) mice, F)에 인간 폐암 세포주인 NCI-H292 IGSF1 O/E 세포 또는 NCI-H292 MOCK 세포를 한 마리당 5×106 세포로 이식하였다. 이식 후 17일째에 마우스에서 채취한 종양 조직의 절편을 탈파라핀화(deparaffinization)하고 재수화하였다.In addition, the distribution of tumor lymphocytes and the expression of IGSF1 expressed within the tumor were confirmed through immunohistochemical staining. Specifically, NSG mice (SID (NSGA) mice, F) transplanted with human peripheral blood mononuclear cells (PBMC) were injected with 5 Transplanted with 6 cells. Sections of tumor tissue collected from mice 17 days after transplantation were deparaffinized and rehydrated.
이후, 열-유도된 에피토프 복구를 위해 표적 복구 완충액에 담궈 전자레인지에서 15분 동안 가열하였다. 가열 후, 표적 복구 완충액에서 30분 동안 놓아둔 후, 트리스 완충 식염수-0.05% 트윈 20(TBS-T)으로 3회 세척하고, 블로킹 용액으로 60분간 블로킹하였다. 1차 항체로 항-IGSF1 항체(Santacruz, sc-393786)를 1:100 희석하여 4℃에서 밤새(overnight) 결합시켰다. 다음날, TBS-T로 3회 세척하고, 내인성 퍼옥시다제 차단 시약(Cell Marque, 925B)으로 상온에서 5분간 반응시켰다. 이후, 2차 항체(Vector, PK-6101 PK-6102)를 상온에서 60분간 결합시켰다. TBS-T로 3회 세척 후, 아비딘-바이오틴과 60분 동안 반응시켰다. 최종 DAB 염색(Vector, SK-4100)을 실시한 후 탈수 과정을 거쳐 조직 염색을 마무리하고, 염색된 조직 절편을 현미경으로 관찰하였다.Afterwards, for heat-induced epitope recovery, the cells were immersed in target recovery buffer and heated in a microwave oven for 15 minutes. After heating, the cells were left in target recovery buffer for 30 minutes, washed three times with Tris-buffered saline-0.05% Tween 20 (TBS-T), and blocked with blocking solution for 60 minutes. As the primary antibody, anti-IGSF1 antibody (Santacruz, sc-393786) was diluted 1:100 and allowed to bind overnight at 4°C. The next day, it was washed three times with TBS-T and reacted with endogenous peroxidase blocking reagent (Cell Marque, 925B) at room temperature for 5 minutes. Afterwards, secondary antibodies (Vector, PK-6101 PK-6102) were bound at room temperature for 60 minutes. After washing three times with TBS-T, it was reacted with avidin-biotin for 60 minutes. After final DAB staining (Vector, SK-4100), tissue staining was completed through a dehydration process, and the stained tissue sections were observed under a microscope.
그 결과, 대조군(NCI-H292 MOCK)에 비해 IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E)가 이식된 인간화 마우스의 종양 조직에서 인간 면역세포인 hCD45+ 세포가 감소하는 것을 확인하였다(도 4).As a result, it was confirmed that hCD45+ cells, a human immune cell, decreased in the tumor tissues of humanized mice transplanted with IGSF1-overexpressing human lung cancer cells (NCI-H292 IGSF1 O/E) compared to the control group (NCI-H292 MOCK) (Figure 4 ).
실시예 3. 항-IGSF1 항체의 결합 친화도 분석Example 3. Binding affinity analysis of anti-IGSF1 antibody
실시예 3.1. Example 3.1. In vitro In vitro 수준에서 항-IGSF1 항체의 결합 친화도 분석Binding affinity analysis of anti-IGSF1 antibodies at the level
WM-A1-3389 항체의 IGSF1 항원에 대한 결합 친화도(binding affinity)를 ELISA 분석을 이용하여 in vitro 수준에서 확인하였다.The binding affinity of the WM-A1-3389 antibody to the IGSF1 antigen was confirmed at the in vitro level using ELISA analysis.
구체적으로, 100 ng IGSF1을 처리하여 96-웰 플레이트를 코팅(coating)한 후, 4% skim milk(PBST)를 200 ㎕씩 첨가하여 상온에서 1시간 동안 블로킹하였다. WM-A1-3389 항체를 4% skim milk(PBST)에 1/3씩 12개 농도로 연속 희석하여 처리한 후, 상온에서 2시간 동안 반응시켰다. 반응이 끝난 후, PBST로 웰을 세척하고 인간 IgG Fc-HRP 항체를 처리하여 상온에서 1시간 동안 반응시켰다. 그 다음, PBST로 웰을 세척한 후, TMB 과산화효소 기질(peroxidase substrate)을 첨가하여 발색 정도를 확인하였다. 이때, 확인은 450 nm 파장에서 흡광도를 측정하여 그 결과를 비교 분석하였다.Specifically, 100 ng IGSF1 was treated to coat a 96-well plate, and then 200 μl of 4% skim milk (PBST) was added and blocked for 1 hour at room temperature. The WM-A1-3389 antibody was serially diluted in 4% skim milk (PBST) at 12 concentrations of 1/3 each and then reacted at room temperature for 2 hours. After the reaction was completed, the wells were washed with PBST, treated with human IgG Fc-HRP antibody, and reacted at room temperature for 1 hour. Next, the wells were washed with PBST, and TMB peroxidase substrate was added to check the degree of color development. At this time, for confirmation, the absorbance was measured at a wavelength of 450 nm and the results were compared and analyzed.
분석 결과, WM-A1-3389 항체의 Kd 값이 2.2×10-11로, IGSF1 항원에 대해 높은 결합 친화도를 나타냈다(도 5).As a result of the analysis, the Kd value of the WM-A1-3389 antibody was 2.2×10 -11 , showing high binding affinity to the IGSF1 antigen (FIG. 5).
실시예 3.2. 세포 내 IGSF1에 대한 항-IGSF1 항체의 결합 친화능 분석Example 3.2. Binding affinity analysis of anti-IGSF1 antibodies to intracellular IGSF1
세포 수준에서 WM-A1-3389 항체의 IGSF1 항원에 대한 결합 친화 정도를 확인하기 위하여, IGSF1 과발현 인간 폐암 세포(NCI-H292 IGSF1 O/E) 및 대조군(NCI-H292 MOCK)을 이용하여 IGSF1에 대한 WM-A1-3389 항체의 결합력을 확인하였다.To confirm the binding affinity of the WM-A1-3389 antibody to the IGSF1 antigen at the cellular level, human lung cancer cells overexpressing IGSF1 (NCI-H292 IGSF1 O/E) and a control group (NCI-H292 MOCK) were used to determine the binding affinity of the WM-A1-3389 antibody to the IGSF1 antigen. The binding ability of the WM-A1-3389 antibody was confirmed.
구체적으로, NCI-H292 IGSF1 O/E 세포 및 NCI-H292 MOCK 세포의 배지를 제거하고, PBS로 1회 세척한 뒤, 0.25% trypsin-EDTA를 2 ㎖ 처리하여 세포를 분리하였다. 분리된 세포는 2% FBS 및 0.05% sodium azide가 포함된 PBS(이하, FACS 버퍼) 8 ㎖로 희석한 뒤, 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하였다. 이후, 세포를 1×105 세포/㎖이 되도록 FACS 버퍼로 재현탁시켰다. 재현탁 후, FACS 튜브에 1 ㎖씩 세포를 분주하고 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하였다.Specifically, the medium of NCI-H292 IGSF1 O/E cells and NCI-H292 MOCK cells was removed, washed once with PBS, and then treated with 2 ml of 0.25% trypsin-EDTA to separate the cells. The separated cells were diluted with 8 ml of PBS (hereinafter referred to as FACS buffer) containing 2% FBS and 0.05% sodium azide, and then centrifuged at 1,200 rpm for 1 minute to remove the supernatant. Afterwards, the cells were resuspended in FACS buffer to 1×10 5 cells/ml. After resuspension, cells were dispensed in 1 ml portions into FACS tubes and centrifuged at 1,200 rpm for 1 minute to remove the supernatant.
FACS 튜브에 남아있는 세포 펠렛(pellet)을 볼텍싱(vortexing)하고, WM-A1-3389 항체를 FACS 버퍼 200 ㎕당 20 μM에서 0 μM까지 1/4씩 희석하여 총 12개의 농도로 첨가한 후, 4℃에서 30분간 반응시켰다. 반응이 끝난 후, 각 튜브에 FACS 버퍼를 1 ㎖씩 첨가하고 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하였다. 이 과정을 총 2회 실시하였다. 또한, FACS 튜브에 남아있는 세포 펠렛을 볼텍싱하고, FITC가 표지된 염소 항-인간 IgG 항체(Invitrogen, 62-8411)를 FACS 버퍼 200 ㎕당 5 ㎍/㎖씩 첨가하고 4℃에서 차광하여 30분동안 반응시켰다. 반응 종료 후, 각 튜브에 FACS 버퍼를 1 ㎖씩 첨가하고 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하였다. 이 과정을 총 2회 실시하였다.The cell pellet remaining in the FACS tube was vortexed, and WM-A1-3389 antibody was diluted by 1/4 from 20 μM to 0 μM per 200 μl of FACS buffer and added at a total of 12 concentrations. , and reacted at 4°C for 30 minutes. After the reaction was completed, 1 ml of FACS buffer was added to each tube and centrifuged at 1,200 rpm for 1 minute to remove the supernatant. This process was performed a total of two times. Additionally, the cell pellet remaining in the FACS tube was vortexed, and FITC-labeled goat anti-human IgG antibody (Invitrogen, 62-8411) was added at 5 ㎍/㎖ per 200 ㎕ of FACS buffer and incubated at 4°C for 30 minutes. Reacted for minutes. After completion of the reaction, 1 ml of FACS buffer was added to each tube and centrifuged at 1,200 rpm for 1 minute to remove the supernatant. This process was performed a total of two times.
마지막으로, 상층액 제거 후 남아있는 세포 펠렛을 FACS 버퍼 200 ㎕에 재현탁하여 FACS 분석하였다. FACS 분석은 BD LSRFortessaTM Flow Cytometer를 이용하여 각 세포에 표지된 FITC 형광값을 측정하였다. 이후, FlowJo 소프트웨어를 사용하여 결과를 분석하였고, EC50 값은 sigma plot 프로그램을 사용하여 계산하였다. 이때, NCI-H292 MOCK 세포는 IGSF1 과발현 세포에 대한 대조군으로 사용하였다.Finally, after removal of the supernatant, the remaining cell pellet was resuspended in 200 μl of FACS buffer and subjected to FACS analysis. FACS analysis measured the fluorescence value of FITC labeled in each cell using a BD LSRFortessa TM Flow Cytometer. Afterwards, the results were analyzed using FlowJo software, and the EC50 value was calculated using the sigma plot program. At this time, NCI-H292 MOCK cells were used as a control for IGSF1 overexpressing cells.
분석 결과, 대조군(NCI-H292 MOCK)에서는 WA-A1-3389 항체의 농도와 무관하게 결합이 확인되지 않았다. 반면, IGSF1이 과발현된 인간 폐암 세포(NCI-H292 IGSF1 O/E)에서는 WM-A1-3389 항체의 EC50 값이 69 nM로 확인되었다(도 6).As a result of the analysis, no binding was confirmed in the control group (NCI-H292 MOCK) regardless of the concentration of WA-A1-3389 antibody. On the other hand, in human lung cancer cells overexpressing IGSF1 (NCI-H292 IGSF1 O/E), the EC50 value of the WM-A1-3389 antibody was confirmed to be 69 nM (FIG. 6).
실시예 4. 세포 내 IGSF1 항원에 대한 항-IGSF1 항체의 항원 특이성 분석Example 4. Analysis of antigen specificity of anti-IGSF1 antibody against intracellular IGSF1 antigen
세포 수준에서 IGSF1 항원에 대한 WM-A1-3389 항체의 항원 특이적 결합 능력(target selectivity)을 분석하기 위하여, IGSF1이 과발현된 인간 폐암 세포(NCI-H292 IGSF1 O/E)를 이용하여, 세포에서 발현되는 IGSF1에 대한 WM-A1-3389 항체의 결합 정도를 확인하였다.In order to analyze the antigen-specific binding ability (target selectivity) of the WM-A1-3389 antibody to the IGSF1 antigen at the cellular level, human lung cancer cells overexpressing IGSF1 (NCI-H292 IGSF1 O/E) were used. The degree of binding of the WM-A1-3389 antibody to expressed IGSF1 was confirmed.
구체적으로, IGSF1이 과발현된 인간 폐암 세포(NCI-H292 IGSF1 O/E) 및 이의 대조군(NCI-H292 MOCK)의 배지를 제거하고, PBS로 1회 세척한 뒤, 0.25% trypsin-EDTA를 2 ㎖ 처리하여 세포를 분리하였다. 분리된 세포는 2% FBS 및 0.05% sodium azide가 포함된 PBS(이하, FACS 버퍼) 8 ㎖로 희석한 뒤, 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하였다. 이후, 세포를 1×105 세포/㎖이 되도록 FACS 버퍼로 재현탁시켰다. 재현탁 후, FACS 튜브에 1 ㎖씩 세포를 분주하고 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하였다.Specifically, the medium of human lung cancer cells overexpressing IGSF1 (NCI-H292 IGSF1 O/E) and its control group (NCI-H292 MOCK) was removed, washed once with PBS, and then added with 2 ml of 0.25% trypsin-EDTA. Cells were separated by treatment. The separated cells were diluted with 8 ml of PBS (hereinafter referred to as FACS buffer) containing 2% FBS and 0.05% sodium azide, and then centrifuged at 1,200 rpm for 1 minute to remove the supernatant. Afterwards, the cells were resuspended in FACS buffer to 1×10 5 cells/ml. After resuspension, cells were dispensed in 1 ml portions into FACS tubes and centrifuged at 1,200 rpm for 1 minute to remove the supernatant.
FACS 튜브에 남아있는 세포 펠렛을 볼텍싱하고, 인간 IgG isotype 항체(Bio X cell, BE0297) 또는 WM-A1-3389 항체를 FACS 버퍼 200 ㎕ 당 0.4 ㎍씩 넣은 후 4℃에서 30분간 반응시켰다. 반응 종료 후, 각 튜브에 FACS 버퍼를 1 ㎖씩 첨가하고 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하였다. 이 과정을 총 2회 실시하였다. FACS 튜브에 남아있는 세포 펠렛을 볼텍싱하고, FITC가 표지된 염소 항-인간 IgG 항체(Invitrogen, 62-8411)를 FACS 버퍼 200 ㎕ 당 0.4 ㎍씩 첨가하고 4℃에서 차광하여 30분 동안 반응시켰다.The cell pellet remaining in the FACS tube was vortexed, and 0.4 μg of human IgG isotype antibody (Bio After completion of the reaction, 1 ml of FACS buffer was added to each tube and centrifuged at 1,200 rpm for 1 minute to remove the supernatant. This process was performed a total of two times. The cell pellet remaining in the FACS tube was vortexed, and 0.4 μg of FITC-labeled goat anti-human IgG antibody (Invitrogen, 62-8411) was added per 200 μl of FACS buffer and reacted at 4°C for 30 minutes, protected from light. .
반응이 끝나고 각 튜브에 FACS 버퍼를 1 ㎖씩 첨가하고 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하였다. 이 과정을 총 2회 실시하였다. 마지막으로, 상층액 제거 후 남아있는 세포 펠렛을 FACS 버퍼 200 ㎕에 재현탁하여 FACS 분석하였다. FACS 분석은 BD LSRFortessaTM Flow Cytometer를 이용하여 각 세포에 표지된 FITC 형광값을 측정하고, FlowJo 소프트웨어를 사용하여 결과를 분석하였다. 이때, NCI-H292 MOCK 세포는 IGSF1 과발현 세포에 대한 대조군으로 사용하였고, 인간 IgG isotype은 WM-A1-3389 항체에 대한 대조군으로 사용하였다.After the reaction was completed, 1 ml of FACS buffer was added to each tube and centrifuged at 1,200 rpm for 1 minute to remove the supernatant. This process was performed a total of two times. Finally, after removal of the supernatant, the remaining cell pellet was resuspended in 200 μl of FACS buffer and subjected to FACS analysis. For FACS analysis, the fluorescence value of FITC labeled in each cell was measured using a BD LSRFortessa TM Flow Cytometer, and the results were analyzed using FlowJo software. At this time, NCI-H292 MOCK cells were used as a control for IGSF1 overexpressing cells, and human IgG isotype was used as a control for WM-A1-3389 antibody.
그 결과, WM-A1-3389 항체 처리군은 IgG isotype 처리군과 비교하여 대조군(NCI-H292 MOCK)에서는 2.6%의 결합력을 보였고, IGSF1 과발현 세포(NCI-H292 IGSF1 O/E)에서는 78.9%의 결합을 나타냈다(도 7).As a result, the WM-A1-3389 antibody treatment group showed a binding affinity of 2.6% in the control group (NCI-H292 MOCK) and 78.9% in IGSF1 overexpressing cells (NCI-H292 IGSF1 O/E) compared to the IgG isotype treatment group. Binding was shown (Figure 7).
다음으로, IGSF1이 과발현된 NCI-H292 IGSF1 O/E 세포 및 HEK293E IGSF1 O/E 세포에 IGSF1을 코딩하는 mRNA에 특이적으로 결합하는 shRNA(이하, shIGSF1)를 형질주입하여 IGSF1의 발현을 감소시킨(이하, IGSF1 K/D 세포) 뒤, 세포 내 IGSF1 항원에 대한 WM-A1-3389 항체의 결합력을 측정하였다. 이때, 형질주입(IGSF1 K/D)의 대조군으로서 shIGSF1을 사용하지 않은 scramble RNA(이하, sc 세포)를 사용하였고, WM-A1-3389 항체에 대한 대조군으로 인간 IgG isotype을 사용하였다. WM-A1-3389 항체의 항원 특이성은 sc 세포에서의 결합력을 기준으로 IGSF1 K/D 세포에서의 결합력을 비교하였다. 또한, IGSF1 과발현 세포에 대한 대조군으로 각각 NCI-H292 MOCK 세포 및 HEK293E MOCK 세포를 사용하였다.Next, shRNA (hereinafter referred to as shIGSF1) that specifically binds to the mRNA encoding IGSF1 was transfected into NCI-H292 IGSF1 O/E cells and HEK293E IGSF1 O/E cells in which IGSF1 was overexpressed, thereby reducing the expression of IGSF1. (hereinafter referred to as IGSF1 K/D cells), the binding affinity of the WM-A1-3389 antibody to the intracellular IGSF1 antigen was measured. At this time, scramble RNA (hereinafter referred to as sc cells) without shIGSF1 was used as a control for transfection (IGSF1 K/D), and human IgG isotype was used as a control for the WM-A1-3389 antibody. The antigen specificity of the WM-A1-3389 antibody was compared with its binding affinity to IGSF1 K/D cells based on its binding affinity to sc cells. Additionally, NCI-H292 MOCK cells and HEK293E MOCK cells were used as controls for IGSF1 overexpressing cells, respectively.
구체적으로, NCI-H292(IGSF1 O/E 및 MOCK) 및 HEK293E(IGSF1 O/E 및 MOCK) 세포주의 배지를 제거하고, PBS로 1회 세척한 뒤, NCI-H292 세포는 0.25% trypsin-EDTA, HEK293E 세포는 0.05% trypsin-EDTA를 각각 2 ㎖씩 처리하여 세포를 분리하였다. 분리된 세포는 배양 배지 8 ㎖로 희석한 뒤, 800 rpm에서 3분간 원심분리하여 상층액을 제거하였다. 남아있는 세포를 각각 1×105 세포/㎖(NCI-H292), 0.5×105 세포/㎖(HEK293E) 농도가 되도록 재현탁시킨 후, 60 mm 배양 플레이트에 3 ㎖씩 첨가하고, 37℃ 세포 인큐베이터에서 하루 동안 배양하였다. 다음 날 shIGSF1 형질주입을 진행하였다. 1.5 ㎖ 튜브에 jet PRIME 버퍼 200 ㎕와 shIGSF1 10 nM을 첨가하고 혼합하였다. 이후, jet PRIME 시약 4 ㎕를 첨가하고 혼합하여 상온에서 10분간 반응시켰다. 또한, 전날 준비해 놓은 세포의 배지를 교체한 후, 형질주입 혼합물을 200 ㎕씩 각 세포에 첨가하여 세포 인큐베이터에서 24시간 동안 반응시켰다. 24시간 후 새로운 배양 배지로 교체하고, 24시간 동안 추가 배양하였다.Specifically, the medium of NCI-H292 (IGSF1 O/E and MOCK) and HEK293E (IGSF1 O/E and MOCK) cell lines was removed, washed once with PBS, and NCI-H292 cells were incubated with 0.25% trypsin-EDTA, HEK293E cells were separated by treating each cell with 2 ml of 0.05% trypsin-EDTA. The separated cells were diluted with 8 ml of culture medium and then centrifuged at 800 rpm for 3 minutes to remove the supernatant. The remaining cells were resuspended to a concentration of 1×10 5 cells/ml (NCI-H292) and 0.5×10 5 cells/ml (HEK293E), respectively, and then added at 3 ml each to a 60 mm culture plate and incubated at 37°C. It was cultured in an incubator for one day. shIGSF1 transfection was performed the next day. 200 ㎕ of jet PRIME buffer and 10 nM of shIGSF1 were added to a 1.5 ㎖ tube and mixed. Afterwards, 4 ㎕ of jet PRIME reagent was added, mixed, and reacted at room temperature for 10 minutes. Additionally, after replacing the cell medium prepared the day before, 200 ㎕ of the transfection mixture was added to each cell and reacted in a cell incubator for 24 hours. After 24 hours, it was replaced with new culture medium and cultured for an additional 24 hours.
형질주입이 끝난 세포는 배지를 제거하고, 상기와 같은 방법으로 FACS 분석을 수행하였다.The medium was removed from the transfected cells, and FACS analysis was performed in the same manner as above.
그 결과, sc 세포주에서 인간 IgG isotype 처리군 대비 WM-A1-3389 항체 결합을 확인하였다. 또한, 상기 결합력을 기준으로 IGSF1 발현을 감소시켰을 때(IGSF1 K/D 세포), WM-A1-3389 항체의 결합력이 함께 감소하는 것을 확인하였다(도 8).As a result, WM-A1-3389 antibody binding was confirmed in the sc cell line compared to the human IgG isotype treatment group. In addition, when IGSF1 expression was reduced based on the binding affinity (IGSF1 K/D cells), it was confirmed that the binding affinity of the WM-A1-3389 antibody also decreased (Figure 8).
실시예 5. 폐암 세포 스페로이드에서 항-IGSF1 항체의 면역 항암 효능 분석Example 5. Analysis of immune anti-cancer efficacy of anti-IGSF1 antibody in lung cancer cell spheroids
세포 수준에서 WM-A1-3389 항체의 면역 항암 효능을 분석하기 위하여, 폐암 세포 스페로이드와 말초혈액 단핵세포(PBMC)를 공동 배양하여 종양 침윤 림프구(TIL) 및 면역원성 세포 사멸을 확인하였다.To analyze the immuno-anticancer efficacy of the WM-A1-3389 antibody at the cellular level, lung cancer cell spheroids and peripheral blood mononuclear cells (PBMC) were co-cultured to determine tumor-infiltrating lymphocytes (TIL) and immunogenic cell death.
폐암 세포 스페로이드와 말초혈액 단핵세포의 공동 배양은 실시예 2.2와 동일한 방법으로 수행하였다.Co-culture of lung cancer cell spheroids and peripheral blood mononuclear cells was performed in the same manner as Example 2.2.
공동 배양한 세포와 상층액을 튜브에 모아 1,200 rpm에서 2분간 원심분리하여 상층액을 제거하였다. 세포 펠렛은 0.25% trypsin-EDTA 500 ㎕를 처리하여 단일 세포로 만든 후, 2% FBS와 0.05% NaN3를 첨가한 PBS(이하, FACS 버퍼) 2 ㎖로 희석하였다. 이후, 1,200 rpm에서 3분간 원심분리하여 상층액을 제거하였다. 남은 세포 펠렛을 FACS 버퍼 200 ㎕에 재현탁시킨 후, 항-HMGB1 항체(Biolegend, 651408)를 첨가하여 4℃에서 30분간 염색시켰다.The co-cultured cells and supernatant were collected in a tube and centrifuged at 1,200 rpm for 2 minutes to remove the supernatant. The cell pellet was treated with 500 ㎕ of 0.25% trypsin-EDTA to form single cells, and then diluted with 2 ㎖ of PBS (hereinafter referred to as FACS buffer) to which 2% FBS and 0.05% NaN 3 were added. Afterwards, the supernatant was removed by centrifugation at 1,200 rpm for 3 minutes. The remaining cell pellet was resuspended in 200 ㎕ of FACS buffer, and anti-HMGB1 antibody (Biolegend, 651408) was added and stained at 4°C for 30 minutes.
각 튜브에 FACS 버퍼 1 ㎖씩 넣고, 1,200 rpm에서 2분간 원심분리하여 상층액을 제거하였다. 이 과정을 총 2회 실시하였다. 이후, BD LSRFortessaTM Flow Cytometer를 이용하여 분석하였다. FACS 분석 결과는 FlowJo 소프트웨어를 사용하여 분석하였다. 또한, 종양 침윤 림프구(TIL)는 형광 현미경을 통해 관찰하였다. 이때, WM-A1-3389 항체에 대한 대조군으로 인간 IgG isotype을 사용하였다.1 ml of FACS buffer was added to each tube, centrifuged at 1,200 rpm for 2 minutes, and the supernatant was removed. This process was performed a total of two times. Afterwards, it was analyzed using a BD LSRFortessa TM Flow Cytometer. FACS analysis results were analyzed using FlowJo software. Additionally, tumor infiltrating lymphocytes (TIL) were observed through fluorescence microscopy. At this time, human IgG isotype was used as a control for the WM-A1-3389 antibody.
그 결과, IGSF1 과발현 폐암 세포(NCI-H292 IGSF1 O/E) 스페로이드에서 대조군에 비해 WM-A1-3389 항체 처리군에서 종양 침윤 림프구(TIL)가 증가하는 것을 확인하였다(도 9). 또한, 면역원성 세포 사멸(ICD)도 IGSF1 과발현 폐암 세포 스페로이드에서 대조군에 비해 WM-A1-3389 항체 처리군에서 증가하는 것을 확인하였다(도 10).As a result, it was confirmed that tumor infiltrating lymphocytes (TIL) increased in the WM-A1-3389 antibody treatment group in IGSF1 overexpressing lung cancer cell (NCI-H292 IGSF1 O/E) spheroids compared to the control group (FIG. 9). In addition, immunogenic cell death (ICD) was confirmed to increase in the WM-A1-3389 antibody treatment group compared to the control group in IGSF1-overexpressing lung cancer cell spheroids (FIG. 10).
실시예 6. 폐암 세포주를 이식한 종양 마우스 모델에서 항-IGSF1 항체 단독 투여에 의한Example 6. By administering anti-IGSF1 antibody alone in a tumor mouse model transplanted with a lung cancer cell line 종양 성장 억제 효능 분석Tumor growth inhibition efficacy analysis
동물 수준에서의 WM-A1-3389 항체의 항암 효능을 확인하기 위하여, 말초혈액 단핵세포 인간화 모델(PBMC humanized model) 마우스에 IGSF1이 과발현된 인간 폐암 세포(NCI-H292 IGSF1 O/E)를 이식한 후, WM-A1-3389 항체의 종양 성장 억제 효능을 평가하였다.To confirm the anticancer efficacy of the WM-A1-3389 antibody at the animal level, human lung cancer cells overexpressing IGSF1 (NCI-H292 IGSF1 O/E) were transplanted into peripheral blood mononuclear cell humanized model (PBMC humanized model) mice. Then, the tumor growth inhibition efficacy of the WM-A1-3389 antibody was evaluated.
구체적으로, 6주령 암컷 말초혈액 단핵세포 인간화 마우스(Gem biosciences)를 구입하여 1주 동안 순화시킨 후, IGSF1이 과발현 인간 폐암 세포 NCI-H292 IGSF1 O/E(5×106 세포/마리)를 PBS와 마트리겔(Matrigel)에 희석하여 마우스의 오른쪽 배측면에 피하(200 ㎕)로 주사하였다. 종양의 크기가 약 120 mm3이 되었을 때 IgG isotype(대조군) 또는 WM-A1-3389 항체를 각각 10 mg/kg 용량으로 복강 투여하였다. 투여는 3일에 한 번씩 4주 동안 실시하였으며, 주 2회 마우스의 종양 크기와 체중을 측정하였다. 투여 22일째 되는 날, 위성군(Satellite group) 마우스의 혈액과 종양을 채취하여 FACS 분석을 실시하였다. 투여가 종료된 후, 실험 동물을 안락사시켜 종양을 적출하여 무게를 측정하였다. WM-A1-3389 항체에 대한 대조군으로 인간 IgG isotype을 사용하였다.Specifically, 6-week-old female peripheral blood mononuclear cell humanized mice (Gem biosciences) were purchased and acclimatized for 1 week, and then IGSF1-overexpressing human lung cancer cells NCI-H292 IGSF1 O/E (5 × 10 6 cells/mouse) were added to PBS. and was diluted in Matrigel and injected subcutaneously (200 ㎕) into the right dorsal side of the mouse. When the tumor size reached approximately 120 mm 3 , IgG isotype (control group) or WM-A1-3389 antibody was administered intraperitoneally at a dose of 10 mg/kg, respectively. Administration was performed once every 3 days for 4 weeks, and the tumor size and body weight of the mice were measured twice a week. On the 22nd day of administration, blood and tumors from satellite group mice were collected and FACS analysis was performed. After administration was completed, the experimental animals were euthanized, the tumors were extracted, and their weight was measured. Human IgG isotype was used as a control for the WM-A1-3389 antibody.
그 결과, WM-A1-3389 항체 투여군은 대조군에 비해 종양 성장 억제 효능이 높게 나타났으며, 약 64.5%의 종양 억제율(TGI)을 보였다(도 11). 또한, 개별 개체들에 대해서도 종양 성장이 억제되는 것을 확인하였다(도 12).As a result, the WM-A1-3389 antibody administration group showed higher tumor growth inhibition efficacy than the control group, and showed a tumor inhibition rate (TGI) of about 64.5% (FIG. 11). In addition, it was confirmed that tumor growth was suppressed in individual subjects (FIG. 12).
실시예 7. 코카서스 인종 폐암 환자 조직에서 IGSF1 발현 분석 Example 7. Analysis of IGSF1 expression in lung cancer patient tissues of Caucasian ethnicity
코카서스 인종(caucasian) 폐암 환자 조직에서 IGSF1의 발현을 면역조직화학염색법으로 확인하였다.The expression of IGSF1 in tissues of Caucasian lung cancer patients was confirmed by immunohistochemical staining.
구체적으로, 인간 비소세포 폐암(non-small cell lung cancer) 환자 조직 절편을 탈파라핀화하고 재수화하였다. 이후, 열-유도된 에피토프 복구를 위해 표적 복구 완충액에 담근 후, 전자레인지에서 15분 동안 가열하였다. 가열 후, 표적 복구 완충액에 30분간 추가 반응시켰다. 이후, 트리스 완충 식염수 0.05% 트윈 20(TBS-T)으로 3회 세척한 다음, 블로킹 용액으로 60분 동안 블로킹하였다. 1차 항체로 항-IGSF1 항체(Santacruz, sc-393786)를 1:100으로 희석하여 4℃에서 밤새(overnight) 결합시켰다. 다음 날, 조직 절편을 TBS-T로 3회 세척한 후, 내인성 퍼옥시다제 차단 시약(Cell Marque, 925B)으로 5분 동안 반응시켰다. 이후, 2차 항체(Vector, PK-6101 PK-6102)를 처리하여 상온에서 60분 동안 결합시켰다. 결합시킨 다음 TBS-T로 3회 세척하였다. 세척 후 아비딘-바이오틴을 처리하고 60분 동안 반응시켰다. 이후, DAB 염색(Vector, SK-4100)을 수행하였으며, 염색된 조직 절편을 현미경을 통해 관찰하였다(도 13).Specifically, human non-small cell lung cancer patient tissue sections were deparaffinized and rehydrated. Thereafter, for heat-induced epitope recovery, it was immersed in target recovery buffer and heated in a microwave oven for 15 minutes. After heating, it was further reacted in target recovery buffer for 30 minutes. Afterwards, it was washed three times with Tris-buffered saline 0.05% Tween 20 (TBS-T), and then blocked with blocking solution for 60 minutes. As the primary antibody, anti-IGSF1 antibody (Santacruz, sc-393786) was diluted 1:100 and allowed to bind overnight at 4°C. The next day, the tissue sections were washed three times with TBS-T and then reacted with endogenous peroxidase blocking reagent (Cell Marque, 925B) for 5 minutes. Afterwards, it was treated with secondary antibody (Vector, PK-6101 PK-6102) and allowed to bind at room temperature for 60 minutes. After binding, the mixture was washed three times with TBS-T. After washing, avidin-biotin was treated and reacted for 60 minutes. Afterwards, DAB staining (Vector, SK-4100) was performed, and the stained tissue sections were observed through a microscope (FIG. 13).
실시예 8. 다양한 암 세포주를 이식한 종양 마우스 모델에서 항-IGSF1 항체 및 항암제 병용 투여에 의한 종양 성장 억제 효능 분석Example 8. Analysis of tumor growth inhibition efficacy by combined administration of anti-IGSF1 antibody and anticancer agent in tumor mouse model transplanted with various cancer cell lines
실시예 8.1. 대장암 세포주 MC38 이식 동계(syngeneic) 마우스 모델에 대한 항-IGSF1 항체 및 항암제 병용 투여에 따른 항종양 효과Example 8.1. Antitumor effect of combined administration of anti-IGSF1 antibody and anticancer agent on syngeneic mouse model transplanted with colon cancer cell line MC38
동물 수준에서 WM-A1-3389 항체와 항-PD-1 항체의 병용 투여 시 항암 효능을 확인하기 위하여, 대장암 세포주인 MC38을 이식한 마우스 모델에 WM-A1-3389 항체 및 항-PD-1 항체를 병용 투여 후 종양 성장 억제 효능을 평가하였다.To confirm the anticancer efficacy of combined administration of WM-A1-3389 antibody and anti-PD-1 antibody at the animal level, WM-A1-3389 antibody and anti-PD-1 were administered to a mouse model transplanted with MC38, a colon cancer cell line. After co-administration of the antibodies, the tumor growth inhibition efficacy was evaluated.
구체적으로, 5 주령 암컷 C57BL/6 마우스(Orient Bio)를 구입하여 1주 동안 순화시킨 후, 마우스 대장암 세포주 MC38(2.5×105 세포/마리)을 PBS에 희석하여 마우스의 오른쪽 배 측면에 피하(100 ㎕)로 주사하였다. 종양의 크기가 약 100 mm3이 되었을 때 mIgG(음성 대조군)(Sigma Aldrich, I5381), WM-A1-3389 항체, 또는 항-PD-1 항체(Bio X Cell, BE0146(RMP1-14))를 각각 10 mg/kg 용량으로, 또는 WM-A1-3389 항체 및 항-PD-1 항체를 각각 10 mg/kg씩 총 20 mg/kg 용량으로 복강 투여하였다. 투여는 3일에 한 번씩 2주 동안 투여 실시하였으며, 주 2회 마우스의 종양 크기와 체중을 측정하였다. 투여가 종료된 후, 실험 동물을 안락사시켜 종양을 적출하여 무게를 측정하였다.Specifically, 5-week-old female C57BL/6 mice (Orient Bio) were purchased and acclimatized for 1 week, and then the mouse colon cancer cell line MC38 (2.5 × 10 5 cells/mouse) was diluted in PBS and injected subcutaneously on the right ventral side of the mouse. (100 μl) was injected. When the tumor size reached approximately 100 mm 3 , mIgG (negative control) (Sigma Aldrich, I5381), WM-A1-3389 antibody, or anti-PD-1 antibody (Bio Each was administered intraperitoneally at a dose of 10 mg/kg, or WM-A1-3389 antibody and anti-PD-1 antibody were each administered at a dose of 10 mg/kg, for a total of 20 mg/kg. Administration was conducted once every three days for two weeks, and the tumor size and body weight of the mice were measured twice a week. After administration was completed, the experimental animals were euthanized, the tumors were extracted, and their weight was measured.
그 결과, 음성 대조군과 비교하여 WM-A1-3389 항체 투여군, 항-PD-1 항체 투여군, 및 WM-A1-3389 항체 및 항-PD-1 항체 병용 투여군 모두에서 종양 성장 억제 효능을 확인하였다. 특히, WM-A1-3389 항체 및 항-PD-1 항체 병용 투여군에서는 WM-A1-3389 항체 또는 항-PD-1 항체 단독 투여군에 비하여 뛰어난 종양 성장 억제 효능을 나타냈다(도 14 및 표 3).As a result, compared to the negative control group, tumor growth inhibition efficacy was confirmed in all the WM-A1-3389 antibody administration group, anti-PD-1 antibody administration group, and WM-A1-3389 antibody and anti-PD-1 antibody combination administration group. In particular, the group administered with the WM-A1-3389 antibody and anti-PD-1 antibody showed superior tumor growth inhibition efficacy compared to the group administered with the WM-A1-3389 antibody or anti-PD-1 antibody alone (FIG. 14 and Table 3).
WM-A1-3389 항체WM-A1-3389 antibody 항-PD-1 항체anti-PD-1 antibody 병용
(WM-A1-3389 항체+항-PD-1 항체)
Combined use
(WM-A1-3389 antibody + anti-PD-1 antibody)
투여 용량 (mg/kg)Dosage administered (mg/kg) 1010 1010 10 + 1010 + 10
TGI (%)TGI (%) 39.1 ± 7.239.1 ± 7.2 43.3 ± 5.243.3 ± 5.2 68.2 ± 8.168.2 ± 8.1
실시예 8.2. 대장암 세포주 CT26 이식 동계 마우스 모델에 대한 항-IGSF1 항체 및 항암제 병용 투여에 따른 항종양 효과Example 8.2. Anti-tumor effect following combined administration of anti-IGSF1 antibody and anticancer agent on syngeneic mouse model transplanted with colon cancer cell line CT26
WM-A1-3389 항체 및 항-PD-1 항체의 병용 투여에 따른 항암 효능을 확인하기 위하여, 상기 실시예 8.1.과 동일한 방법으로 수행하되 본 실시예에서는 이식 세포주로서 대장암 세포주인 CT26을 사용하고, 수여 마우스(recipient mouse)로는 5주령 암컷 BALB/c 마우스를 사용하였다.In order to confirm the anticancer efficacy of the combined administration of the WM-A1-3389 antibody and the anti-PD-1 antibody, the same method as Example 8.1 was performed, except that in this example, CT26, a colon cancer cell line, was used as the transplant cell line. And, 5-week-old female BALB/c mice were used as recipient mice.
대장암 세포주인 CT26을 이식한 마우스 모델에 WM-A1-3389 항체 및 항-PD-1 항체를 병용 투여한 후 종양 성장 억제 효능을 분석해 본 결과, 음성 대조군과 비교하여 WM-A1-3389 항체 투여군, 항-PD-1 항체 투여군, 및 WM-A1-3389 항체 및 항-PD-1 항체 병용 투여군 모두에서 종양 성장 억제 효능을 확인하였다. 특히, WM-A1-3389 항체 및 항-PD-1 항체 병용 투여군에서는 WM-A1-3389 항체 또는 항-PD-1 항체 단독 투여군에 비하여 뛰어난 종양 성장 억제 효능을 나타냈다(도 15 및 표 4).As a result of analyzing the efficacy of tumor growth inhibition after co-administration of WM-A1-3389 antibody and anti-PD-1 antibody to a mouse model transplanted with CT26, a colon cancer cell line, the WM-A1-3389 antibody group was compared to the negative control group. , the tumor growth inhibition efficacy was confirmed in both the anti-PD-1 antibody administration group and the WM-A1-3389 antibody and anti-PD-1 antibody combination administration group. In particular, the group administered with the WM-A1-3389 antibody and anti-PD-1 antibody showed superior tumor growth inhibition efficacy compared to the group administered with the WM-A1-3389 antibody or anti-PD-1 antibody alone (FIG. 15 and Table 4).
WM-A1-3389 항체WM-A1-3389 antibody 항-PD-1 항체anti-PD-1 antibody 병용
(WM-A1-3389 항체+항-PD-1 항체)
Combined use
(WM-A1-3389 antibody + anti-PD-1 antibody)
투여 용량 (mg/kg)Dosage administered (mg/kg) 1010 1010 10 + 1010 + 10
TGI (%)TGI (%) 39.8 ± 5.639.8 ± 5.6 3.6 ± 8.63.6 ± 8.6 73.3 ± 5.873.3 ± 5.8
실시예 8.3. 폐암 세포주 LLC1 이식 동계 마우스 모델에 대한 항-IGSF1 항체 및 항암제 병용 투여에 따른 항종양 효과Example 8.3. Antitumor effect of combined administration of anti-IGSF1 antibody and anticancer drug on syngeneic mouse model transplanted with lung cancer cell line LLC1
WM-A1-3389 항체 및 항-PD-1 항체의 병용 투여에 따른 항암 효능을 확인하기 위하여, 상기 실시예 8.1.과 동일한 방법으로 수행하되 본 실시예에서는 이식 세포주로서 폐암 세포주인 LLC1을 사용하였으며, 투여는 3일에 한 번씩 11일 동안 투여 진행하였다.In order to confirm the anticancer efficacy of the combined administration of the WM-A1-3389 antibody and the anti-PD-1 antibody, the same method as Example 8.1 was performed, except that in this example, the lung cancer cell line LLC1 was used as the transplant cell line. , Administration was conducted once every three days for 11 days.
폐암 세포주인 LLC1을 이식한 마우스 모델에 WM-A1-3389 항체 및 항-PD-1 항체를 병용 투여한 후 종양 성장 억제 효능을 분석해 본 결과, 음성 대조군과 비교하여 WM-A1-3389 항체 투여군, 항-PD-1 항체 투여군, 및 WM-A1-3389 항체 및 항-PD-1 항체 병용 투여군 모두에서 종양 성장 억제 효능을 확인하였다. 특히, WM-A1-3389 항체 및 항-PD-1 항체 병용 투여군에서는 WM-A1-3389 항체 또는 항-PD-1 항체 단독 투여군에 비하여 뛰어난 종양 성장 억제 효능을 나타냈다(도 16 및 표 5).When WM-A1-3389 antibody and anti-PD-1 antibody were co-administered to a mouse model transplanted with LLC1, a lung cancer cell line, and the tumor growth inhibition efficacy was analyzed, compared to the negative control group, the WM-A1-3389 antibody administration group, Tumor growth inhibition efficacy was confirmed in both the anti-PD-1 antibody administration group and the WM-A1-3389 antibody and anti-PD-1 antibody combination administration group. In particular, the group administered with the WM-A1-3389 antibody and anti-PD-1 antibody showed superior tumor growth inhibition efficacy compared to the group administered with the WM-A1-3389 antibody or anti-PD-1 antibody alone (FIG. 16 and Table 5).
WM-A1-3389 항체WM-A1-3389 antibody 항-PD-1 항체anti-PD-1 antibody 병용
(WM-A1-3389 항체+항-PD-1 항체)
Combined use
(WM-A1-3389 antibody + anti-PD-1 antibody)
투여 용량 (mg/kg)Dosage administered (mg/kg) 1010 1010 10 + 1010 + 10
TGI (%)TGI (%) 55.4 ± 10.555.4 ± 10.5 22.2 ± 12.022.2 ± 12.0 84.9 ± 2.684.9 ± 2.6

Claims (5)

  1. IGSF1의 C 말단에 특이적으로 결합하는 항-IGSF1 항체 또는 이의 단편 및 항-PD-1 항체를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer comprising an anti-IGSF1 antibody or fragment thereof that specifically binds to the C terminus of IGSF1 and an anti-PD-1 antibody as active ingredients.
  2. 제1항에 있어서,According to paragraph 1,
    상기 항-IGSF1 항체는 서열번호 1의 H-CDR1, 서열번호 2의 H-CDR2 및 서열번호 3의 H-CDR3을 포함하는 중쇄 가변영역; 및The anti-IGSF1 antibody includes a heavy chain variable region comprising H-CDR1 of SEQ ID NO: 1, H-CDR2 of SEQ ID NO: 2, and H-CDR3 of SEQ ID NO: 3; and
    서열번호 4의 L-CDR1, 서열번호 5의 L-CDR2 및 서열번호 6의 L-CDR3을 포함하는 경쇄 가변영역을 포함하는 것인, 암 예방 또는 치료용 약학 조성물. A pharmaceutical composition for preventing or treating cancer, comprising a light chain variable region including L-CDR1 of SEQ ID NO: 4, L-CDR2 of SEQ ID NO: 5, and L-CDR3 of SEQ ID NO: 6.
  3. 제2항에 있어서,According to paragraph 2,
    상기 중쇄 가변영역은 서열번호 7의 아미노산 서열을 가지며;The heavy chain variable region has the amino acid sequence of SEQ ID NO: 7;
    상기 경쇄 가변영역은 서열번호 8의 아미노산 서열을 가지는 것인, 암 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer, wherein the light chain variable region has the amino acid sequence of SEQ ID NO: 8.
  4. 제1항에 있어서,According to paragraph 1,
    상기 항-PD-1 항체는 펨브롤리주맙, 니볼루맙, 세미플리맙, JTX-4014, 스파르탈리주맙, 캄렐리주맙, 신틸리맙, 티슬레리주맙, 토리팔리맙, 도스탈리맙, INCMGA00012, AMP-224 및 AMP-514로 이루어진 군에서 선택되는 어느 하나인 것인, 암 예방 또는 치료용 약학 조성물.The anti-PD-1 antibodies include pembrolizumab, nivolumab, cemiplimab, JTX-4014, spartalizumab, camrelizumab, sintilimab, thyslerizumab, toripalimab, dostalimab, INCMGA00012 , a pharmaceutical composition for preventing or treating cancer, which is any one selected from the group consisting of AMP-224 and AMP-514.
  5. 제1항에 있어서,According to paragraph 1,
    상기 암은 위암, 간암, 폐암, 비소세포 폐암, 대장암, 방관암, 골암, 혈액암, 유방암, 흑색종양, 갑상선암, 부갑성선암, 골수암, 직장암, 인후암, 후두암, 식도암, 췌장암, 설암, 피부암, 죄종양, 자궁암, 두부암, 경부암, 담낭암, 구강암, 항문 부근암, 결장암 및 중추신경계 종양으로 구성된 군에서 선택되는 어느 하나인, 암 예방 또는 치료용 약학 조성물.The above cancers include stomach cancer, liver cancer, lung cancer, non-small cell lung cancer, colon cancer, ductal cancer, bone cancer, blood cancer, breast cancer, melanoma, thyroid cancer, parathyroid cancer, bone marrow cancer, rectal cancer, throat cancer, larynx cancer, esophagus cancer, pancreatic cancer, tongue cancer, and skin cancer. A pharmaceutical composition for preventing or treating cancer, which is any one selected from the group consisting of sinus cancer, uterine cancer, head cancer, cervical cancer, gallbladder cancer, oral cancer, anal cancer, colon cancer, and central nervous system tumor.
PCT/KR2023/005475 2022-04-25 2023-04-21 Pharmaceutical composition for cancer treatment, comprising anti-igsf1 antibody and anti-pd-1 antibody WO2023211068A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2022-0050763 2022-04-25
KR20220050763 2022-04-25

Publications (1)

Publication Number Publication Date
WO2023211068A1 true WO2023211068A1 (en) 2023-11-02

Family

ID=88519385

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/005475 WO2023211068A1 (en) 2022-04-25 2023-04-21 Pharmaceutical composition for cancer treatment, comprising anti-igsf1 antibody and anti-pd-1 antibody

Country Status (2)

Country Link
KR (1) KR20230151913A (en)
WO (1) WO2023211068A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20160014564A (en) * 2014-07-29 2016-02-11 재단법인 아산사회복지재단 Novel Biomarkers for Predicting Susceptibility to MET Inhibitor and Uses Thereof
KR20180081606A (en) * 2015-11-23 2018-07-16 파이브 프라임 테라퓨틱스, 인크. FGFR2 inhibitor alone or in combination with an immunostimulant in cancer therapy
KR20180132401A (en) * 2017-06-02 2018-12-12 주식회사 파멥신 Composition for treating or preventing cancer
KR20210122185A (en) * 2020-03-31 2021-10-08 웰마커바이오 주식회사 Pharmaceutical composition for preventing or treating cancer comprising antibodies against igsf1, and method for treating cancer using the same
US20210371529A1 (en) * 2017-12-27 2021-12-02 Innovent Biologics (Suzhou) Co., Ltd. Anti-lag-3 antibody and use thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017024465A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20160014564A (en) * 2014-07-29 2016-02-11 재단법인 아산사회복지재단 Novel Biomarkers for Predicting Susceptibility to MET Inhibitor and Uses Thereof
KR20180081606A (en) * 2015-11-23 2018-07-16 파이브 프라임 테라퓨틱스, 인크. FGFR2 inhibitor alone or in combination with an immunostimulant in cancer therapy
KR20180132401A (en) * 2017-06-02 2018-12-12 주식회사 파멥신 Composition for treating or preventing cancer
US20210371529A1 (en) * 2017-12-27 2021-12-02 Innovent Biologics (Suzhou) Co., Ltd. Anti-lag-3 antibody and use thereof
KR20210122185A (en) * 2020-03-31 2021-10-08 웰마커바이오 주식회사 Pharmaceutical composition for preventing or treating cancer comprising antibodies against igsf1, and method for treating cancer using the same

Also Published As

Publication number Publication date
KR20230151913A (en) 2023-11-02

Similar Documents

Publication Publication Date Title
KR101683884B1 (en) Anti-epcam antibody and uses thereof
JP7284557B2 (en) Pharmaceutical composition
EP3998287A1 (en) Anti-cd47/anti-pd-1 bispecific antibody, preparation method and use thereof
JP2018511586A (en) Combination of Taskinimod or a pharmaceutically acceptable salt thereof and a PD-1 and / or PD-L1 inhibitor for use as a medicament
KR20210142638A (en) CD3 antigen-binding fragment and applications thereof
US11351251B2 (en) Anti-PD-L1-anti-TIM-3 bispecific antibodies
US20220227871A1 (en) Anti-pd-l1 antibody
US20230414778A1 (en) COMBINATION OF ANTIBODY-DRUG CONJUGATE WITH ANTI-SIRPalpha ANTIBODY
JP2020500179A (en) Anti-BAG3 antibodies for therapeutic use in combination with immune checkpoint inhibitors
WO2023022271A1 (en) Anti-igsf1 antibody and use thereof
TW201841938A (en) Anti-ceacam1 antibody and use thereof
US11858984B2 (en) Anti-S100A8/A9 antibody and use thereof
WO2023211068A1 (en) Pharmaceutical composition for cancer treatment, comprising anti-igsf1 antibody and anti-pd-1 antibody
WO2022231032A1 (en) Anti-cntn4-specific antibodies and use thereof
IL298894A (en) Human anti-grp94 antibodies and uses
WO2024090916A1 (en) Pharmaceutical composition for cancer treatment comprising anti-igsf1 antibody
WO2023229303A1 (en) Antibody binding to c-terminus of igsf1 and use thereof
WO2024058289A1 (en) Pharmaceutical composition, for prevention or treatment of neuroendocrine neoplasm, containing antibody that specifically binds to pd-1 as active ingredient
WO2023219072A1 (en) COMBINATION OF ANTIBODY-DRUG CONJUGATE AND ANTI-SIRPα ANTIBODY
CN117510636B (en) GPRC5D antibody and application thereof
EP4349858A1 (en) Chimeric antigen receptor specifically binding to cd300c antigen or receptor thereof
WO2022005068A1 (en) Antibody specifically binding to lgals3bp, and use thereof
CN107648609B (en) Inhibition of effector T cell autophagy in sepsis by IL-35
KR101856904B1 (en) Antibody specifically binding to PAUF and use thereof
WO2012133914A1 (en) Potentiator of cancer immunity containing rankl antagonist

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: 23796715

Country of ref document: EP

Kind code of ref document: A1