WO2025249993A1 - Composition for treating cancerous diseases and screening method thereof - Google Patents
Composition for treating cancerous diseases and screening method thereofInfo
- Publication number
- WO2025249993A1 WO2025249993A1 PCT/KR2025/095352 KR2025095352W WO2025249993A1 WO 2025249993 A1 WO2025249993 A1 WO 2025249993A1 KR 2025095352 W KR2025095352 W KR 2025095352W WO 2025249993 A1 WO2025249993 A1 WO 2025249993A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cancer
- tam
- marker
- cells
- macrophage
- Prior art date
- Legal status (The legal status 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 status listed.)
- Pending
Links
Definitions
- the present invention relates to a composition for treating cancer and a method for screening the same.
- Cancer is one of the incurable diseases civilization faces, and massive amounts of capital are being invested globally in the development of treatments. In South Korea, it has been the leading cause of death since 1983, with over 100,000 people diagnosed annually and over 60,000 dying. Carcinogens, including smoking, ultraviolet rays, chemicals, food, and other environmental factors, are known to cause cancer. However, the diverse causes make the development of treatments challenging, and the effectiveness of treatments also varies depending on the site of the disease.
- anticancer drugs include biological agents such as enzyme preparations or vaccines, purely synthetic drugs, and drugs derived from natural products.
- anticancer drugs using genes, enzymes, and vaccines are not yet at the practical stage, and anticancer drugs developed through chemotherapy have significant toxicity and have side effects of destroying not only cancer cells but also normal cells because they cannot selectively eliminate only cancer cells.
- cancer cells have recently developed resistance to these drugs, making them ineffective in cancer treatment. Therefore, there is an urgent need to develop effective anticancer drugs that are less toxic and do not induce resistance in cancer cells for the treatment and prevention of cancer. Accordingly, the development of a simple method for screening new anticancer drugs is also required.
- the inventors of the present invention completed the present invention by confirming that the expression of tumor-associated macrophage (TAM) markers changed when a culture medium derived from cancer-associated fibroblasts exposed to killed cancer cells was treated.
- TAM tumor-associated macrophage
- the present invention provides a pharmaceutical composition for treating cancer, comprising a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured.
- CAFs cancer-associated fibroblasts
- the present invention provides a method for screening a cancer treatment agent, comprising the steps of: (a) administering a carcinogenic substance or cancer cells to a non-human experimental animal; (b) administering a test substance to the experimental animal; and (c) confirming the expression levels of tumor suppressive macrophage (M1 TAM) markers and tumor supportive macrophage (M2 TAM) markers in cells of the experimental animal.
- M1 TAM tumor suppressive macrophage
- M2 TAM tumor supportive macrophage
- the description of redundant content will be omitted below.
- the content of the invention is not limited to the content described below, and the content of the invention should be interpreted based on the overall content of the invention.
- the present invention provides a pharmaceutical composition for treating cancer, comprising a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured.
- CAFs cancer-associated fibroblasts
- cancer-associated fibroblast refers to ⁇ -SMA (alpha-smooth muscle actin) positive fibroblasts existing inside and/or around a cancer lesion, and the presence of CAF has been confirmed in various cancers such as colon cancer, lung cancer, prostate cancer, breast cancer, stomach cancer, cholangiocarcinoma, and basal cell carcinoma.
- CAF cancer-associated fibroblast
- the CAF may be associated with one or more cancers selected from the group consisting of fibrosarcoma, malignant fibrous histiocytoma, liposarcoma, rhabdomyosarcoma, leiomyosarcoma, hemangiosarcoma, malignant skin cancer, lymphangiosarcoma, synovial sarcoma, chondrosarcoma, osteosarcoma, lung cancer, gastric cancer, breast cancer, colon cancer, and prostate cancer.
- cancers selected from the group consisting of fibrosarcoma, malignant fibrous histiocytoma, liposarcoma, rhabdomyosarcoma, leiomyosarcoma, hemangiosarcoma, malignant skin cancer, lymphangiosarcoma, synovial sarcoma, chondrosarcoma, osteosarcoma, lung cancer, gastric cancer, breast cancer, colon cancer, and prostate cancer.
- apoptotic cancer cells may be cancer cells that have been induced to apoptotic by irradiating them with light of a specific wavelength.
- the irradiation of light of the specific wavelength may be ultraviolet ray (UV) irradiation.
- the wavelength may be irradiated for 5 to 30 minutes at a wavelength of 100 to 400 nm. More specifically, the UV irradiation may be irradiated for 20 minutes at a wavelength of 150 to 350 nm or for 10 to 15 minutes at a wavelength of 200 to 300 nm.
- co-culture may be achieved by co-culturing CAFs with apoptotic cancer cells.
- CAFs may be cultured with apoptotic cancer cells in X-VIVO or serum-free DMEM medium for 20 to 30 hours.
- culture medium refers to a culture product obtained through co-culturing CAFs and apoptotic cancer cells.
- the culture medium may be a liquid medium, a solid medium, or a semi-solid medium.
- the culture medium may be a conditioned medium (CM).
- cancer refers to a disease in which abnormally transformed cells proliferate rapidly and uncontrollably due to changes in the genes of cells for various reasons. Cancer cells can spread to various parts of the body through the bloodstream and lymphatic system.
- the cancer disease may be at least one selected from the group consisting of breast cancer, uterine cancer, esophageal cancer, stomach cancer, brain cancer, rectal cancer, colon cancer, lung cancer, skin cancer, ovarian cancer, cervical cancer, blood cancer, pancreatic cancer, prostate cancer, testicular cancer, laryngeal cancer, oral cancer, head and neck cancer, thyroid cancer, liver cancer, bladder cancer, osteosarcoma, lymphoma, and leukemia, and preferably may be lung cancer.
- the lung cancer may be lung adenocarcinoma or non-small cell lung cancer.
- a culture medium co-cultured with cancer-associated fibroblasts (CAFs) and apoptotic cancer cells can inhibit tumor growth by increasing the expression level of tumor suppressive macrophage (T1 TAM) markers and decreasing the expression level of tumor supportive macrophage (T2 TAM) markers.
- CAFs cancer-associated fibroblasts
- T1 TAM tumor suppressive macrophage
- T2 TAM tumor supporting macrophage
- a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured can increase the levels of pro-apoptotic markers including Bax, C-Cas3, and C-PARP, and can decrease the expression levels of anti-apoptotic markers including Mcl-1 and Bcl-xL.
- CAFs cancer-associated fibroblasts
- changes in the expression levels of the above markers may be due to the fact that the culture medium co-cultured with cancer-associated fibroblasts (CAFs) and apoptotic cancer cells not only inhibits the survival of M2 macrophages and promotes apoptosis through the WISP-1-STAT1 signaling pathway, but also induces reprogramming from M2 TAMs to TAMs expressing an M1-like phenotype.
- CAFs cancer-associated fibroblasts
- WISP-1 is a target protein of the WNT signaling pathway, and WNT signaling plays a role in lung development, regulating both epithelial and mesenchymal development through autocrine and paracrine signals.
- CD16 is an Fc-gamma receptor, also known as Fc ⁇ RIII, and is found on the surface of NK cells, neutrophils, monocytes, and macrophages.
- CD32 is an Fc-gamma receptor, also known as Fc ⁇ RII, and corresponds to a surface receptor glycoprotein belonging to the Ig gene family.
- CD80 is a B7, type I membrane protein belonging to the Ig gene family, and is known to be involved in regulating T cell activation and B cell activity.
- H2Ab1 corresponds to a variant of a histone protein encoded by the H2AFB1 gene, and activates various functions including peptide antigen binding activity.
- MHCII refers to the major histocompatibility complex found only in professional antigen-presenting cells such as dendritic cells, macrophages, and B cells.
- iNOS NOS2
- Ca 2+ -independent NOS Nitric oxide synthase
- IFN ⁇ is a dimerized soluble cytokine belonging to the type II interferon group, and is known to have a role in activating macrophages to increase phagocytosis, tumor sterilization, and intracellular apoptosis.
- IL12p40 is a subunit of the IL-12 cytokine family, functions as a chemoattractant for macrophages, and plays a role in promoting the movement of dendritic cells.
- TNF ⁇ is a member of the tumor necrosis factor (TNF) family, which is mainly secreted by activated macrophages and is known to play an important role in various immune-mediated inflammatory diseases.
- IL-1 ⁇ refers to a cytokine protein encoded by IL1B among the interleukin-1 genes, and is involved in various cell activities such as cell proliferation, differentiation, and apoptosis.
- CD163 is a high-affinity scavenger receptor for the hemoglobin-haptoglobin complex, and is also a marker for cells of the monocyte/macrophage lineage.
- CD206 is also called mannose receptor and corresponds to C-type lectin that mainly exists on the surface of macrophages, immature dendritic cells, and liver endothelial cells.
- Arginase 1 is a gene encoding arginase, which is known as a marker of M2a macrophages and myeloid-derived suppressor cells (MDSC), which are major mediators of T cell suppression.
- TGF ⁇ 1 is a polypeptide member of the transforming growth factor beta family of cytokines, and is a protein that performs various functions including regulation of cell growth, proliferation, differentiation, and apoptosis.
- IL-10 is an anti-inflammatory cytokine that plays a role in downregulating the expression of Th1 cytokines, MHCII antigens, and co-stimulatory molecules in macrophages.
- IL-4 is a cytokine that induces differentiation from naive Th0 cells into Th2 cells, and is an important regulator of humoral immunity and acquired immunity.
- IL-13 is a protein encoded by the IL13 gene, and is a cytokine secreted from Th2 cells, CD4 cells, mast cells, etc.
- Bax is also known as Bcl-2-like protein 4, which forms a heterodimer with Bcl-2 and functions as an apoptosis activator.
- C-Cas3 refers to Caspase 3, which is cleaved and activated during cell death, and transmits a cell death signal through enzymatic activity toward downstream targets including PARP and other substrates.
- C-PARP refers to Poly-ADP-ribose polymerase (PARP) that is cleaved by caspase during caspase-dependent apoptosis.
- Mcl-1 belongs to the Bcl-2 protein family and plays a role in regulating cell death, cell cycle progression, and mitochondrial homeostasis.
- Bcl-xL is a member of the Bcl-2 protein family and prevents the release of mitochondrial contents that induce caspase activation and cell death.
- treatment refers to any act of improving or beneficially changing the symptoms of a cancer disease by administering a composition according to the present invention.
- composition of the present invention may be formulated and provided in an appropriate form.
- the composition may be prepared by including one or more pharmaceutically acceptable carriers.
- the pharmaceutically acceptable carriers include, but are not limited to, those commonly used in the art, such as lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methyl hydroxybenzoate, propyl hydroxybenzoate, talc, magnesium stearate, and mineral oil.
- the pharmaceutical composition of the present invention may include, but is not limited to, diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrants, and surfactants, and other pharmaceutically acceptable additives.
- the pharmaceutical composition of the present invention when formulated as a solid oral preparation, it includes tablets, pills, powders, granules, capsules, etc., and such solid preparations may include at least one excipient, for example, starch, calcium carbonate, sucrose or lactose, gelatin, etc., and include, but are not limited to, lubricants such as magnesium stearate and talc.
- excipient for example, starch, calcium carbonate, sucrose or lactose, gelatin, etc.
- lubricants such as magnesium stearate and talc.
- the pharmaceutical composition of the present invention when formulated as an oral liquid, it includes a suspension, a solution, an emulsion, a syrup, etc., and includes, but is not limited to, a diluent such as water or liquid paraffin, a wetting agent, a sweetener, a fragrance, a preservative, etc.
- the pharmaceutical composition of the present invention when formulated for parenteral use, it includes a sterile aqueous solution, a non-aqueous solvent, a suspension, an emulsion, a lyophilized preparation, and a suppository.
- Non-aqueous solvents and suspensions include, but are not limited to, propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate.
- Bases for suppositories include, but are not limited to, witepsol, macrogol, tween 61, cacao butter, laurin butter, and glycerogelatin.
- composition may be administered in single or multiple doses in a pharmaceutically effective amount.
- pharmaceutically effective amount means an amount sufficient to prevent or treat a disease at a reasonable benefit/risk ratio applicable to medical prevention or treatment, and the effective dosage level may be determined according to factors including the severity of the disease, the activity of the drug, the patient's age, weight, health, sex, the patient's sensitivity to the drug, the time of administration of the composition of the present invention used, the route of administration and excretion rate, the duration of treatment, drugs combined with or used concurrently with the composition of the present invention used, and other factors well known in the medical field.
- the pharmaceutical composition of the present invention can be administered to mammals such as rats, mice, livestock, and humans by various routes, for example, but not limited to, oral administration, intrathecal, intra-auricular, intraperitoneal or intravenous, intramuscular, subcutaneous, intrauterine, sublingual, or intracerebrovascular injection.
- the pharmaceutical composition of the present invention can be formulated into a unit dosage form pharmaceutical preparation suitable for administration into the patient's body according to a conventional method in the pharmaceutical field and administered, and the preparation includes an effective dosage amount through one or more administrations.
- Preferred dosage forms for this purpose are parenteral administration preparations such as injections and infusions.
- the administration dosage of the pharmaceutical composition may vary depending on the patient's age, weight, sex, dosage form, health condition, and disease severity, and may be administered once or several times a day at regular intervals according to the judgment of a doctor or pharmacist.
- the route and method of administration of the pharmaceutical composition of the present invention may be independent of each other, and are not particularly limited in their method, and any route and method of administration may be followed as long as the pharmaceutical composition can reach the target area.
- administration refers to introducing a given substance into a human or animal by any suitable method.
- the therapeutic composition according to the present invention may be administered orally or parenterally via any common route, as long as it can reach the target tissue.
- the therapeutic composition according to the present invention may be administered by any device capable of transporting the active ingredient to target cells.
- the present invention provides a method for screening a cancer treatment agent.
- the screening method for a cancer treatment agent comprises the following steps:
- the present invention comprises a step of (a) administering a carcinogenic substance or cancer cells to an experimental animal other than a human.
- any substance known to be a carcinogen can be used to produce an animal model that induces cancer.
- arsenic, benzene, beryllium, cadmium, hexavalent chromium compounds, ethylene oxide, nickel, radon, or vinyl chloride can be used to induce cancer.
- any experimental animal commonly used in the art can be treated with arsenic, benzene, beryllium, cadmium, hexavalent chromium compounds, ethylene oxide, nickel, radon, and/or vinyl chloride, and the animal model itself or cells isolated therefrom in which the disease is induced can be used.
- the experimental animal model can be, for example, a mouse, hamster, rat, ferret, guinea pig, rabbit, dog, primate, or pig, and more preferably a mouse.
- any cancer cell for inducing the above cancer may be administered.
- the cancer cells may be one or more selected from the group consisting of fibrosarcoma, malignant fibrous histiocytoma, liposarcoma, rhabdomyosarcoma, leiomyosarcoma, hemangiosarcoma, malignant skin cancer, lymphangiosarcoma, synovial sarcoma, chondrosarcoma, osteosarcoma, lung cancer, gastric cancer, breast cancer, colon cancer, and prostate cancer.
- the administration of such cancer cells may be administered in an amount typically known to induce cancer in each experimental animal.
- the cancer-causing substance or cancer cells may be administered through a route appropriate to their characteristics, for example, through respiration, intramuscular injection, renal injection, intraperitoneal injection, or subcutaneous injection.
- the substances expressed or activated in vivo in non-cancer and cancer-induced states are different. Therefore, to screen for candidate substances for cancer prevention and treatment, cancer must first be induced.
- the method for screening a cancer treatment agent according to the present invention comprises the step of (b) administering a test substance to the experimental animal.
- test material refers to an unknown candidate substance used in screening to determine whether it affects the expression level of a gene or the expression or activity of a protein.
- the sample includes, but is not limited to, chemicals, nucleotides, antisense RNA, siRNA (small interference RNA), and natural product extracts.
- test substances can be administered via conventional routes of administration, including oral and parenteral administration.
- oral administration may be used, and parenteral administration may be selected from, but is not limited to, topical application to the skin, intraperitoneal injection, rectal injection, subcutaneous injection, intravenous injection, intramuscular injection, or intrathoracic injection.
- the dosage of the test substance can be adjusted to an appropriate level of dosage according to the type of drug and the drug efficacy, based on the general knowledge of those skilled in the art.
- the method for screening for cancer comprises the step of (c) confirming the expression level of a tumor suppressive macrophage (M1 TAM) marker and a tumor supporting macrophage (M2 TAM) marker in cells of the experimental animal.
- M1 TAM tumor suppressive macrophage
- M2 TAM tumor supporting macrophage
- the method for screening for cancer according to the present invention may include selecting a test substance that (i) increases the expression level of a tumor suppressive macrophage (M1 TAM) marker and (ii) decreases the expression level of a tumor supportive macrophage (T2 TAM) marker in step (c).
- M1 TAM tumor suppressive macrophage
- T2 TAM tumor supportive macrophage
- tumor-associated macrophages are macrophages that participate in the formation of a tumor microenvironment, and influence tumor angiogenesis, proliferation, metastasis, and immunosuppression by secreting various cytokines, chemokines, and proteolytic enzymes.
- the tumor-associated macrophages are divided into tumor-suppressive macrophages (M1 TAMs) and tumor-supportive macrophages (M2 TAMs).
- the above tumor suppressive macrophage (M1 TAM) marker may be any one or more selected from the group consisting of CD16/32, CD80, H2Ab1, MHCII, iNOS (NOS2), IFN ⁇ , IL12p40, TNF ⁇ , and IL-1 ⁇ , but is not limited thereto.
- the above tumor-supporting macrophage (M2 TAM) marker may be any one or more selected from the group consisting of, but is not limited to, CD163, CD206, Arginase 1 (Arg1), TGF ⁇ 1, IL-10, IL-4, and IL-13.
- the drug may be considered to have therapeutic efficacy against cancer.
- the tumor suppressive macrophage (M1 TAM) marker is expressed at a level comparable to that of the positive control, the drug may be considered to have therapeutic efficacy against cancer.
- the drug may be judged to have therapeutic efficacy against cancer.
- the tumor-supporting macrophage (M2 TAM) marker is expressed at a level equivalent to that of the positive control, the drug may be judged to have therapeutic efficacy against cancer.
- a control sample means all sample groups for which the expression levels of tumor suppressive macrophage (M1 TAM) markers and tumor supportive macrophage (M2 TAM) markers can be compared and determined according to test substance treatment, including a normal control group and a therapeutic substance control group known for treating cancer.
- M1 TAM tumor suppressive macrophage
- M2 TAM tumor supportive macrophage
- the expression levels of the above tumor suppressive macrophage (M1 TAM) marker and tumor supporting macrophage (M2 TAM) marker can be measured by any one selected from the group consisting of, but not limited to, enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qPCR), immunohistochemistry and Western blotting, and flow cytometry.
- ELISA enzyme-linked immunosorbent assay
- qPCR quantitative real-time polymerase chain reaction
- immunohistochemistry immunohistochemistry and Western blotting
- flow cytometry any one selected from the group consisting of, but not limited to, enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qPCR), immunohistochemistry and Western blotting, and flow cytometry.
- the method for screening for cancer according to the present invention may further include the step of (d) confirming (i) an increase in the expression level of a pro-apoptotic marker and (ii) a decrease in the expression level of an anti-apoptotic marker in cells of the experimental animal.
- the pro-apoptotic marker may be at least one selected from the group consisting of Bax, C-Cas3, and C-PARP, and the anti-apoptotic marker may be at least one selected from the group consisting of Mcl-1 and Bcl-xL.
- the test substance may be a pharmaceutical composition including a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured.
- CAFs cancer-associated fibroblasts
- the present invention comprises the steps of: (a) contacting a test substance with cancer cells;
- a method for screening a cancer treatment agent comprising the step of selecting a test substance having (i) an increased expression level of the tumor suppressive macrophage (M1 TAM) marker and (ii) a decreased expression level of the tumor supportive macrophage (M2 TAM) marker compared to a control sample;
- Any cancer cell can be used as the cancer cell, and any type of cancer cell mentioned above can be equally applied to the present screening method.
- the method for screening a cancer treatment agent of the present invention comprises a step of contacting a test substance with cancer cells.
- the cancer cells include cells from a patient with cancer, animal cells, or tissues thereof.
- cells induced with cancer-causing substances can be utilized, and any substance known to be a cancer-causing substance can be utilized.
- the cancer cells can be induced by treatment with arsenic, benzene, beryllium, cadmium, hexavalent chromium compounds, ethylene oxide, nickel, radon, or vinyl chloride.
- arsenic, benzene, beryllium, cadmium, hexavalent chromium compounds, ethylene oxide, nickel, radon, or vinyl chloride any laboratory animal commonly used in the art can be treated with arsenic, benzene, beryllium, cadmium, hexavalent chromium compounds, ethylene oxide, nickel, radon, or vinyl chloride, and the resulting animal model, or cells isolated therefrom, can be used.
- Contact with the above test substance means all contacts in vitro and in vivo , and includes all contacts in which the test substance is treated on cells in vitro or administered to animals, etc. in vivo .
- the method for screening a cancer treatment agent comprises the steps of: confirming the expression levels of tumor suppressive macrophage (M1 TAM) markers and tumor supportive macrophage (M2 TAM) markers in cancer cells that have come into contact with the test substance; and selecting a test substance in which (i) the expression level of the tumor suppressive macrophage (M1 TAM) marker increases and (ii) the expression level of the tumor supportive macrophage (M2 TAM) marker decreases compared to a control sample.
- M1 TAM tumor suppressive macrophage
- M2 TAM tumor supportive macrophage
- the expression levels of the tumor suppressive macrophage (M1 TAM) marker and the tumor supporting macrophage (M2 TAM) marker can be measured by any one selected from the group consisting of, but not limited to, enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qPCR), immunohistochemistry, Western blotting, and flow cytometry.
- ELISA enzyme-linked immunosorbent assay
- qPCR quantitative real-time polymerase chain reaction
- immunohistochemistry Western blotting
- flow cytometry any one selected from the group consisting of, but not limited to, enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qPCR), immunohistochemistry, Western blotting, and flow cytometry.
- the composition for treating cancer of the present invention can treat cancer by suppressing the survival of M2 macrophages and inducing reprogramming of M2 TAMs into M1 TAMs, thereby inhibiting tumor growth. Furthermore, the screening method of the present invention can be usefully utilized in the development and identification of novel cancer treatment drugs by easily identifying and obtaining substances effective in treating cancer among a large number of therapeutic candidates.
- Figure 1 is a diagram showing that the total TAM density of a primary tumor is reduced by administration of ApoSQ-CAF CM.
- Figure 2 is a diagram showing that the M2 TAM fraction of primary tumors is reduced by administration of ApoSQ-CAF CM.
- Figure 3 is a diagram showing that the M1 TAM fraction of primary tumors increases by administration of ApoSQ-CAF CM.
- Figure 4 is a diagram confirming that the cell death promotion effect is mediated by WISP-1.
- Figure 5 is a diagram confirming that reprogramming from M2 TAM to M1 TAM is induced by administration of ApoSQ-CAF CM.
- the upper part is a representative flow cytometry plot for CD11b + TAM, and the lower part shows the TAM ratio (CD163 + /MHCII + TAM ratio).
- Figure 6 is a diagram showing polarization into M1 and M2 macrophages.
- FIGS. 1 and (d) are diagrams showing the results of immunoblotting using M2 markers (CD163, CD206, and Arginase1 (Arg1)) and M1 markers (MHCII, iNOS, and IL12p40) on M1 and M2 macrophages (M2) derived from THP-1 cells and BMDM.
- M2 markers CD163, CD206, and Arginase1 (Arg1)
- M1 markers MHCII, iNOS, and IL12p40
- (e) and (f) are diagrams showing the results of immunofluorescence staining for the M1 marker CD86 and the M2 marker CD163.
- Figure 7 is a diagram confirming in vitro that the apoptosis of M2 macrophages increases by CM administration of CAFs exposed to killed cancer cells.
- Figure 8 is a diagram confirming in vitro that reprogramming from M2 TAM to M1 TAM is induced by CM administration of CAF exposed to killed cancer cells.
- (d) and (e) are diagrams showing the results of flow cytometry analysis on CD16 + and CD206 + cells among M2 macrophages derived from THP-1 and BMDM.
- Mouse rWISP-1 (1680-WS) and human rWISP-1 (1627-WS) were purchased from R&D Systems (Minneapolis, MN, USA).
- Neutralizing mouse WISP-1 antibody (MAB1680) and IgG (MAB0061) were purchased from R&D Systems (Minneapolis, MN, USA).
- CAFs were isolated from lung tumors of Kras-mutant (Kras LA1 ) mice by magnetic-activated cell sorting (MACS) using the fibroblast-specific marker Thy1. Isolated CAFs were cultured in ⁇ -MEM medium supplemented with 10% fetal bovine serum, penicillin/streptomycin (100 U/100 ⁇ g), 2 mM L-glutamine, and 1 mM sodium pyruvate. For immortalization, CAFs were stably transfected with the TERT plasmid (pCDH-3xFLAG-TERT; Addgene 51 plasmid #51631) using Lipofector-EXT (AptaBio).
- Cancer epithelial cell lines were irradiated with 254 nm of UV light for 15 min and then cultured for 2 h at 37°C under 5% CO2 conditions.
- Light microscopy of Wright-Giemsa-stained samples revealed that most irradiated cells were apoptotic.
- lysed (necrotic) cancer cells were obtained through multiple freeze-thaw cycles. Apoptosis and necrosis were confirmed by flow cytometric analysis using a FACSCalibur system (BD Biosciences) after staining with annexin V-FITC/propidium iodide (BD Biosciences).
- CAFs were plated at 3 ⁇ 10 5 cells/ml and incubated overnight at 37°C under 5% CO 2 conditions. After serum-starvation for 24 h, the cells were stimulated with X-VIVO 10 medium (04-380Q). For this, the culture medium was replaced with X-VIVO 10 medium containing killed or necrotic cancer cells (9 ⁇ 10 5 cells/ml). After 20 h, the supernatant was harvested by centrifugation and used as CM for stimulating target cancer epithelial cells (5 ⁇ 10 3 cells/ml). The CM was stored at -80°C for in vivo experiments.
- CAF CM were incubated with 10 ⁇ g/ml mouse anti-WISP-1 neutralizing antibody (R&D Systems) or 10 ⁇ g/ml IgG isotype control (R&D Systems) for 2 h.
- the anti-WISP-1 antibody neutralizing effect was tested by WISP-1 ELISA before use.
- mice were managed and handled in accordance with the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health (NIH).
- 344SQ cells a lung adenocarcinoma cell line (1 ⁇ 106 cells in 100 ⁇ l of PBS per mouse), were injected subcutaneously into the right posterior flank of syngeneic (129/Sv) mice. 2 days later, CM derived from CAFs (100 ⁇ l per mouse) was injected intratumorally three times a week. In addition, CM with or without neutralizing mouse anti-WISP-1 Ab (10 ⁇ g/ml) or isotype IgG was administered on the same schedule. Tumor growth in the mice was monitored daily, and the mice were sacrificed 6 weeks after injection. Autopsies were then performed to examine the diameter and weight of the subcutaneous tumor masses.
- formalin fixation was performed for 30 min at room temperature, the samples were washed three times for 5 min each with IF-Wash buffer (PBS containing 0.05% NaN 3 , 0.1% BSA, 0.2% Triton X-100, and 0.05% Tween-20) and permeabilized with 0.5% Triton X-100 in PBS (Sigma-Aldrich) for 5 min at room temperature.
- IF-Wash buffer PBS containing 0.05% NaN 3 , 0.1% BSA, 0.2% Triton X-100, and 0.05% Tween-20
- permeabilized with 0.5% Triton X-100 in PBS Sigma-Aldrich
- the target protein was captured with the primary antibody during an 18-hour incubation at 4°C, and the captured protein was visualized with fluorescence-conjugated IgG in a darkroom for 1 hour. After staining, the slides were mounted with VECTASHIELD mounting medium containing DAPI (Vector Laboratories) and imaged using a confocal microscope (LSM5 PASCAL). Information on the antibodies used is shown in Table 1 below.
- Equal amounts of protein were then dissolved in SDS-PAGE gels (#161-0158, Bio-Rad Laboratories) and transferred to nitrocellulose membranes (10600001, GE Healthcare Life Science) using a wet transfer system (Bio-Rad Laboratories).
- the membranes were blocked with 5% bovine serum albumin (BSA)-TBST or 5% milk-TBST for 1 h, incubated with labeled primary antibodies overnight, and then incubated with labeled secondary antibodies for 1 h at 37°C.
- BSA bovine serum albumin
- the Odyssey image analysis system was used for quantification.
- THP-1 cells were maintained in RPMI 1640 medium containing 10% FBS at 37°C under humidified conditions containing 5% CO2 .
- THP-1 cell-derived M1 or M2 macrophages were generated as a macrophage model. Specifically, THP-1 cells were primed with 150 ng/ml PMA for 6 h to produce nonpolarized macrophages. To generate M1 macrophages, nonpolarized macrophages were stimulated with 20 ng/ml IFN ⁇ and 100 ng/ml LPS for 48 h. To generate M2 macrophages, nonpolarized macrophages were further stimulated with 20 ng/ml IL-4 and 20 ng/ml IL-13 for 48 h.
- BMDMs isolated from the tibia and femur of C57BL/6 mice were cultured with L929 complement DMEM for 7 days and then polarized into M1 and M2 type macrophages.
- Macrophages (3.5 ⁇ 10 4 ) were plated in 96-well plates containing RPMI-1640 medium and cultured in X-VIVO 10 medium for 6 hours. CM or rWISP-1 was added to each group and incubated for 2–4 days under 5% CO 2 , 37°C conditions. Afterwards, cell counting kit-8 (CCK-8) solution was added to the wells and incubated for 30 minutes. The absorbance was measured at 450 nm using a microplate reader.
- CM or rWISP-1 was added to each group and incubated for 2–4 days under 5% CO 2 , 37°C conditions.
- cell counting kit-8 (CCK-8) solution was added to the wells and incubated for 30 minutes. The absorbance was measured at 450 nm using a microplate reader.
- CAFs or macrophages were transiently transfected with siRNAs specifically targeting WISP1 (Bioneer), STAT1 (Bioneer), or a control siRNA (SN-1003 AccuTarget TM Negative Control) at a final concentration of 50 nM using a transfection reagent (Lipofectamin RNAi MAX; Invitrogen, Carlsbad, CA). After overnight transfection, cells were cultured in appropriate media for 24 h and stimulated with ApoSQ cells.
- the siRNA sequences used were as follows (gene: sense, antisense):
- Apoptosis was detected using an annexin V-FITC/propidium iodide (PI) staining kit (BD Biosciences, San Jose, CA, USA) according to the manufacturer's instructions. After harvesting, macrophages were resuspended in 500 ⁇ l of binding buffer, and 100 ⁇ l of the suspension was stained with 5 ⁇ l of FITC-conjugated annexin V and 5 ⁇ l of PI for 15 min at room temperature in the dark. FITC-conjugated annexin V-positive cells were then detected by flow cytometry (ACEA NovoCyte, San Diego, CA, USA) using 400 ⁇ l of binding buffer, and data were analyzed using NovoExpress software 1.5.
- PI annexin V-FITC/propidium iodide
- NovoCyte (Agilent, Santa Clara, CA, USA) was used. All cell suspensions (1 ⁇ 10 6 CD11b + TAMs and M2 macrophages) were placed in 500 ⁇ l buffer (PBS + 2% FBS) and incubated for 60 min with fluorophore-conjugated anti-mouse antibodies at the manufacturer's recommended concentration. All antibodies used are listed in Table 3. Data acquisition was performed on NovoCyte (Agilent, Santa Clara, CA, USA), and NovoExpress Software 1.5 was used for analysis.
- TNF- ⁇ , IL-1 ⁇ , IL-4, and IL-13 in macrophage cultures were measured using ELISA kits (R&D Systems) according to the manufacturer's instructions.
- Pairwise comparisons were performed using the two-tailed Student's t-test, and multiple comparisons were performed using the Kruskal-Wallis test followed by Dunn's post hoc test. A P value less than 0.05 was considered statistically significant, and all data were analyzed using Prism 5 software (GraphPad Software Inc., San Diego, CA, USA).
- CM CM of CAFs
- ApoSQ-CAF CM CM of CAFs exposed to killed 344SQ cells
- the CMs were pre-incubated for 2 h with a neutralizing antibody against WISP-1 or an IgG isotype control antibody.
- Immunofluorescence staining using total TAM markers revealed that administration of ApoSQ-CAF CM significantly reduced total TAM density in both the central and marginal regions of primary tumors compared to administration of CAF CM (Fig. 1).
- administration of WISP-1 immunodepleted ApoSQ-CAF CM did not change total TAM density, but administration of CM pre-incubated with IgG isotype control antibody showed an effect similar to that of administration of ApoSQ-CAF CM.
- Example 2 Induction of reprogramming from M2 TAM to M1 TAM by ApoSQ-CM administration.
- M2 and M1 markers in CD11b + TAMs isolated from primary tumors were analyzed using RT-qPCR arrays, respectively.
- nine M2-related genes were downregulated more than twofold in the ApoSQ-CAF CM group compared to the CAF CM group (Fig. 5a).
- seven M1-related genes including Cd32, Ifng, Cd16, Tnf, Nos2, Socs3, and Cd80, were upregulated more than twofold in the ApoSQ-CAF CM group compared to the CAF CM group.
- qRT-PCR analysis of M2- and M1-specific markers and cytokines was also performed.
- THP-1 cells and primary mouse bone marrow-derived macrophages were polarized into M1 and M2 macrophages to mimic TAMs (Figs. 6a and 6b).
- Successful polarization into M1 and M2 macrophages was confirmed by immunoblotting using M2 markers (CD163, CD206, and Arg1) and M1 markers (MHCII, iNOS, and IL12p40) (Figs. 6c and 6d).
- confocal microscopy analysis also confirmed polarization using CD86 + M1 and CD163 + M2 markers (Figs. 6e and 6f).
- THP-1-derived M2 macrophages treatment of THP-1-derived M2 macrophages with ApoSQ-CAF CM enhanced the levels of M1 cytokines, including TNF ⁇ and IL-1 ⁇ , whereas suppressed the levels of M2 cytokines, including IL-4 and IL-13 (Fig. 8c).
- flow cytometry analysis showed that treatment of THP-1-derived M2 macrophages or BMDM-derived M2 macrophages with ApoSQ-CAF CM decreased the surface expression of the CD206 M2 marker and increased the surface expression of the CD16 M1 marker compared to treatment with CAF CM or NecSQ-CAF CM (Figs. 8d and 8e).
Landscapes
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Description
본 발명은 암 질환 치료용 조성물 및 이를 스크리닝하는 방법에 관한 것이다.The present invention relates to a composition for treating cancer and a method for screening the same.
암은 인류가 해결해야 할 난치병 중의 하나로, 전 세계적으로 이를 치유하기 위한 개발에 막대한 자본이 투자되고 있는 실정이며, 우리나라의 경우, 1983년 이후로 한국인의 사망원인 중 제1위의 질병으로서 연간 약 10만명 이상이 진단되고, 약 6만 명 이상이 사망하고 있다. 이러한 암의 유발 인자인 발암물질로는 흡연, 자외선, 화학물질, 음식물, 기타 환경인자들이 있으나, 그 유발 원인이 다양하여 치료제의 개발이 어려울 뿐만 아니라 발생하는 부위에 따라 치료제의 효과 또한 각기 다르다.Cancer is one of the incurable diseases humanity faces, and massive amounts of capital are being invested globally in the development of treatments. In South Korea, it has been the leading cause of death since 1983, with over 100,000 people diagnosed annually and over 60,000 dying. Carcinogens, including smoking, ultraviolet rays, chemicals, food, and other environmental factors, are known to cause cancer. However, the diverse causes make the development of treatments challenging, and the effectiveness of treatments also varies depending on the site of the disease.
현재 사용되는 항암제로는 효소제제 또는 백신 등의 생물학적 제제, 순수합성 의약품 및 천연물 유래의 의약품 등이 있으며, 이 중 유전자, 효소, 백신 등을 이용한 항암제는 실용단계에 있는 상태가 아니며 화학요법에 의해 개발된 항암제는 상당한 독성을 지니고 있고, 암세포만을 선택적으로 제거하지 못해 암세포뿐만 아니라 정상세포도 파괴시키는 부작용이 있으며, 최근에는 이에 대한 암세포의 내성이 발생되어 암 치료에 효과적이지 못한 상태이다. 따라서, 암의 치료 및 예방을 위하여 독성이 적고 암세포의 내성을 유발시키지 않는 효과적인 항암제의 개발이 절실히 필요하며, 이에 따라 신규 항암제를 간편하게 스크리닝하는 방법의 개발 역시 요구되는 실정이다.Currently used anticancer drugs include biological agents such as enzyme preparations or vaccines, purely synthetic drugs, and drugs derived from natural products. Among these, anticancer drugs using genes, enzymes, and vaccines are not yet at the practical stage, and anticancer drugs developed through chemotherapy have significant toxicity and have side effects of destroying not only cancer cells but also normal cells because they cannot selectively eliminate only cancer cells. In addition, cancer cells have recently developed resistance to these drugs, making them ineffective in cancer treatment. Therefore, there is an urgent need to develop effective anticancer drugs that are less toxic and do not induce resistance in cancer cells for the treatment and prevention of cancer. Accordingly, the development of a simple method for screening new anticancer drugs is also required.
이에, 본 발명자들은 사멸화된 암세포에 노출된 암 관련 섬유아세포 유래 배양액을 처리하였을 때 종양 관련 대식세포(TAM) 마커의 발현이 변화함을 확인하여 본 발명을 완성하였다.Accordingly, the inventors of the present invention completed the present invention by confirming that the expression of tumor-associated macrophage (TAM) markers changed when a culture medium derived from cancer-associated fibroblasts exposed to killed cancer cells was treated.
본 발명은 암 관련 섬유아세포(cancer-associated fibroblast, CAF) 및 사멸화된 암세포(apoptotic cancer cells)를 공동배양한 배양액을 포함하는 암 질환 치료용 약학적 조성물을 제공한다.The present invention provides a pharmaceutical composition for treating cancer, comprising a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured.
본 발명은 (a) 암 유발 물질 또는 암세포를 인간을 제외한 실험동물에 투여하는 단계; (b) 시험물질을 상기 실험동물에 투여하는 단계; 및 (c) 상기 실험동물의 세포에서 종양 억제성 대식세포(M1 TAM) 마커 및 종양 지지성 대식세포(M2 TAM) 마커의 발현 수준을 확인하는 단계;를 포함하는 암 질환 치료제의 스크리닝 방법을 제공한다.The present invention provides a method for screening a cancer treatment agent, comprising the steps of: (a) administering a carcinogenic substance or cancer cells to a non-human experimental animal; (b) administering a test substance to the experimental animal; and (c) confirming the expression levels of tumor suppressive macrophage (M1 TAM) markers and tumor supportive macrophage (M2 TAM) markers in cells of the experimental animal.
하기에서는 중복되는 내용의 혼잡을 방지하기 위하여, 중복되는 내용의 기재를 생략하고자 한다. 즉, 하기의 내용만으로 발명의 내용이 한정되는 것은 아니고, 전체적인 발명의 내용에 따라 발명의 내용이 해석되어야 할 것이다.To avoid confusion due to overlapping content, the description of redundant content will be omitted below. In other words, the content of the invention is not limited to the content described below, and the content of the invention should be interpreted based on the overall content of the invention.
이하, 본 발명을 상세하게 설명한다.Hereinafter, the present invention will be described in detail.
본 발명은 암 관련 섬유아세포(cancer-associated fibroblast, CAF) 및 사멸화된 암세포(apoptotic cancer cells)를 공동배양한 배양액을 포함하는 암 질환 치료용 약학적 조성물을 제공한다.The present invention provides a pharmaceutical composition for treating cancer, comprising a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured.
본 발명에서 "암 관련 섬유아세포(cancer-Associated Fibroblast, CAF)"란 암 병변의 내부 및/또는 주변에 존재하는 α-SMA(alpha-smooth muscle actin) 양성 섬유아세포를 의미하며, 상기 CAF는 대장암, 폐암, 전립선암, 유방암, 위암, 담관암, 기저세포암 등의 다양한 암에서 그 존재가 확인되고 있다.In the present invention, "cancer-associated fibroblast (CAF)" refers to α-SMA (alpha-smooth muscle actin) positive fibroblasts existing inside and/or around a cancer lesion, and the presence of CAF has been confirmed in various cancers such as colon cancer, lung cancer, prostate cancer, breast cancer, stomach cancer, cholangiocarcinoma, and basal cell carcinoma.
본 발명에서 상기 CAF는 섬유육종, 악성섬유성조직구종, 지방육종, 횡문 근육종, 평활근육종, 혈관육종, 악성피부암, 림프혈관육종, 활막육종, 연골육종, 골육종, 폐암, 위암, 유방암, 대장암 및 전립선암으로 이루어진 군에서 선택되는 하나 이상의 암과 관련된 것일 수 있다.In the present invention, the CAF may be associated with one or more cancers selected from the group consisting of fibrosarcoma, malignant fibrous histiocytoma, liposarcoma, rhabdomyosarcoma, leiomyosarcoma, hemangiosarcoma, malignant skin cancer, lymphangiosarcoma, synovial sarcoma, chondrosarcoma, osteosarcoma, lung cancer, gastric cancer, breast cancer, colon cancer, and prostate cancer.
본 발명에서 "사멸화된 암세포(apoptotic cancer cells)"는 암세포에 특정 파장의 빛을 조사하여 사멸화를 유도한 것일 수 있다. 상기 특정 파장의 빛의 조사는 자외선(Ultra-violet ray, UV) 조사일 수 있다. 구체적으로, 상기 파장은 100 내지 400 nm 파장으로 5 내지 30분 동안 조사될 수 있다. 보다 구체적으로, 상기 UV 조사는 150 내지 350 nm 파장으로 내지 20분 동안 또는 200 내지 300 nm 파장으로 10 내지 15분 동안 조사될 수 있다.In the present invention, "apoptotic cancer cells" may be cancer cells that have been induced to apoptotic by irradiating them with light of a specific wavelength. The irradiation of light of the specific wavelength may be ultraviolet ray (UV) irradiation. Specifically, the wavelength may be irradiated for 5 to 30 minutes at a wavelength of 100 to 400 nm. More specifically, the UV irradiation may be irradiated for 20 minutes at a wavelength of 150 to 350 nm or for 10 to 15 minutes at a wavelength of 200 to 300 nm.
본 발명에서 "공동 배양"은 CAF와 사멸화된 암세포를 함께 배양하여 이루어질 수 있다. 일 실시예에서, CAF를 20 내지 30 시간 동안 X-VIVO 또는 무혈청 DMEM 배지에서 사멸화된 암세포와 함께 배양할 수 있다.In the present invention, "co-culture" may be achieved by co-culturing CAFs with apoptotic cancer cells. In one embodiment, CAFs may be cultured with apoptotic cancer cells in X-VIVO or serum-free DMEM medium for 20 to 30 hours.
본 발명에서 "배양액"이란 CAFs와 사멸화된 암세포의 공동 배양을 통해 얻어진 배양 결과물을 의미한다. 본 발명의 일 실시예에서, 상기 배양액은 액체배지, 고체배지 또는 반고체배지일 수 있다. 일 실시예에서, 상기 배양액은 조정배지(conditioned medium, CM)일 수 있다.In the present invention, "culture medium" refers to a culture product obtained through co-culturing CAFs and apoptotic cancer cells. In one embodiment of the present invention, the culture medium may be a liquid medium, a solid medium, or a semi-solid medium. In one embodiment, the culture medium may be a conditioned medium (CM).
본 발명에서 "암(cancer)"은 여러 가지 이유로 인하여 세포의 유전자에 변화가 일어남으로써 비정상적으로 변한 세포가 신속하고 제어 불가능하게 증식하는 질환을 일컫는다. 암세포는 혈류 및 림프계를 통해 체내의 여러 부위로 퍼져 나갈 수 있다. 상기 암 질환은 유방암, 자궁암, 식도암, 위암, 뇌암, 직장암, 대장암, 폐암, 피부암, 난소암, 자궁경부암, 혈액암, 췌장암, 전립선암, 고환암, 후두암, 구강암, 두경부암, 갑상선암, 간암, 방광암, 골육종, 림프종 및 백혈병으로 이루어진 군에서 선택되는 하나 이상일 수 있으며, 바람직하게는 폐암일 수 있다. 상기 폐암은 폐선암 또는 비소세포폐암일 수 있다.In the present invention, "cancer" refers to a disease in which abnormally transformed cells proliferate rapidly and uncontrollably due to changes in the genes of cells for various reasons. Cancer cells can spread to various parts of the body through the bloodstream and lymphatic system. The cancer disease may be at least one selected from the group consisting of breast cancer, uterine cancer, esophageal cancer, stomach cancer, brain cancer, rectal cancer, colon cancer, lung cancer, skin cancer, ovarian cancer, cervical cancer, blood cancer, pancreatic cancer, prostate cancer, testicular cancer, laryngeal cancer, oral cancer, head and neck cancer, thyroid cancer, liver cancer, bladder cancer, osteosarcoma, lymphoma, and leukemia, and preferably may be lung cancer. The lung cancer may be lung adenocarcinoma or non-small cell lung cancer.
본 발명의 일 실시예에 따르면, 암 관련 섬유아세포(cancer-associated fibroblast, CAF) 및 사멸화된 암세포(apoptotic cancer cells)를 공동배양한 배양액은 종양 억제성 대식세포(T1 TAM) 마커의 발현 수준을 증가시키고, 종양 지지성 대식세포(T2 TAM) 마커의 발현 수준을 감소시킴으로써 종양의 성장을 억제할 수 있다.According to one embodiment of the present invention, a culture medium co-cultured with cancer-associated fibroblasts (CAFs) and apoptotic cancer cells can inhibit tumor growth by increasing the expression level of tumor suppressive macrophage (T1 TAM) markers and decreasing the expression level of tumor supportive macrophage (T2 TAM) markers.
상기 종양 억제성 대식세포(T1 TAM) 마커는 CD16/32, CD80, H2Ab1, MHCII, iNOS(NOS2), IFNγ, IL12p40, TNFα 및 IL-1β로 구성된 군으로부터 선택된 어느 하나 이상일 수 있으며, 종양 지지성 대식세포(T2 TAM) 마커는 CD163, CD206, Arginase 1(Arg1), TGFβ1, IL-10, IL-4 및 IL-13으로 구성된 군으로부터 선택된 어느 하나 이상일 수 있다.The above tumor suppressive macrophage (T1 TAM) marker may be at least one selected from the group consisting of CD16/32, CD80, H2Ab1, MHCII, iNOS (NOS2), IFNγ, IL12p40, TNFα, and IL-1β, and the tumor supporting macrophage (T2 TAM) marker may be at least one selected from the group consisting of CD163, CD206, Arginase 1 (Arg1), TGFβ1, IL-10, IL-4, and IL-13.
또한, 본 발명의 일 실시예에 따르면, 암 관련 섬유아세포(cancer-associated fibroblast, CAF) 및 사멸화된 암세포(apoptotic cancer cells)를 공동배양한 배양액은 Bax, C-Cas3, C-PARP를 포함한 친-사멸화(pro-apoptotic) 마커의 수준을 증가시킬 수 있으며, Mcl-1 및 Bcl-xL를 포함한 항-사멸화(anti-apoptotic) 마커의 발현 수준을 감소시킬 수 있다.In addition, according to one embodiment of the present invention, a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured can increase the levels of pro-apoptotic markers including Bax, C-Cas3, and C-PARP, and can decrease the expression levels of anti-apoptotic markers including Mcl-1 and Bcl-xL.
구체적으로, 상기 마커들의 발현 수준 변화는 암 관련 섬유아세포(cancer-associated fibroblast, CAF) 및 사멸화된 암세포(apoptotic cancer cells)를 공동배양한 배양액이 WISP-1-STAT1 신호 전달 경로를 통해 M2 대식세포의 생존을 억제하고 세포 사멸을 촉진할 뿐만 아니라, M2 TAM으로부터 M1 유사 표현형을 발현하는 TAM으로의 리프로그래밍을 유도하기 때문에 나타나는 것일 수 있다.Specifically, changes in the expression levels of the above markers may be due to the fact that the culture medium co-cultured with cancer-associated fibroblasts (CAFs) and apoptotic cancer cells not only inhibits the survival of M2 macrophages and promotes apoptosis through the WISP-1-STAT1 signaling pathway, but also induces reprogramming from M2 TAMs to TAMs expressing an M1-like phenotype.
상기 WISP-1은 WNT 시그널링 경로의 표적 단백질이며, WNT 시그널링은 자가분비(autocrine) 및 주변분비(paracrine) 신호를 통해 상피(epithelial) 및 중간엽(mesenchymal) 발달 모두를 조절하면서, 폐 발달에서 역할을 한다.The above WISP-1 is a target protein of the WNT signaling pathway, and WNT signaling plays a role in lung development, regulating both epithelial and mesenchymal development through autocrine and paracrine signals.
본 발명에서 "CD16"은 Fc-감마 수용체로서 FcγRⅢ로도 알려져 있으며, NK세포, 호중구, 단핵구, 대식세포의 표면에서 발견된다.In the present invention, "CD16" is an Fc-gamma receptor, also known as FcγRⅢ, and is found on the surface of NK cells, neutrophils, monocytes, and macrophages.
본 발명에서 "CD32"는 Fc-감마 수용체로서 FcγRⅡ로도 알려져 있으며, Ig 유전자 계통에 속하는 표면 수용체 당단백질에 해당한다.In the present invention, “CD32” is an Fc-gamma receptor, also known as FcγRⅡ, and corresponds to a surface receptor glycoprotein belonging to the Ig gene family.
본 발명에서 "CD80"는 Ig 유전자 계통에 속하는 B7, I형 막단백질이며, T세포 활성화 및 B세포 활성 조절에 관여하는 것으로 알려져 있다.In the present invention, "CD80" is a B7, type I membrane protein belonging to the Ig gene family, and is known to be involved in regulating T cell activation and B cell activity.
본 발명에서 "H2Ab1"은 H2AFB1 유전자에 의하여 암호화되는 히스톤 단백질의 변이체에 해당하며, 펩타이드 항원 결합 활성을 포함한 여러 기능을 활성화한다.In the present invention, "H2Ab1" corresponds to a variant of a histone protein encoded by the H2AFB1 gene, and activates various functions including peptide antigen binding activity.
본 발명에서 "MHCⅡ"는 수지상세포, 대식세포, B세포와 같은 전문적인 항원 제시 세포(antigen-presenting cell)에서만 발견되는 주조직 적합성 복합체(Major histocompatibility complex)를 의미한다.In the present invention, “MHCⅡ” refers to the major histocompatibility complex found only in professional antigen-presenting cells such as dendritic cells, macrophages, and B cells.
본 발명에서 "iNOS(NOS2)"는 사이토카인 및 기타 작용제에 의하여 광범위한 세포 및 조직에서 발현이 유도될 수 있는 고출력(high-output) Ca2+-독립적 NOS(Nitric oxide synthase)를 일컫는다.In the present invention, "iNOS (NOS2)" refers to a high-output Ca 2+ -independent NOS (Nitric oxide synthase) whose expression can be induced in a wide range of cells and tissues by cytokines and other agents.
본 발명에서 "IFNγ"는 Ⅱ형 인터페론 그룹에 속하는 이량체화된 가용성 사이토카인으로서, 대식세포를 활성화하여 식균작용, 종양살균작용, 세포내 사멸 등을 증가시키는 역할을 하는 것으로 알려져 있다.In the present invention, "IFNγ" is a dimerized soluble cytokine belonging to the type II interferon group, and is known to have a role in activating macrophages to increase phagocytosis, tumor sterilization, and intracellular apoptosis.
본 발명에서 "IL12p40"은 IL-12 사이토카인 계열의 소단위(subunit)로서, 대식세포에 대하여 화학유인물질(chemoattractant)로 기능하며, 수지상세포의 이동을 촉진하는 역할을 한다.In the present invention, "IL12p40" is a subunit of the IL-12 cytokine family, functions as a chemoattractant for macrophages, and plays a role in promoting the movement of dendritic cells.
본 발명에서 "TNFα"는 종양괴사인자(tumor necrosis factor, TNF) 계열의 구성원으로 주로 활성화된 대식세포에 의해 분비되며, 여러 면역 매개 염증성 질환에서 중요한 역할을 하는 것으로 알려져 있다.In the present invention, "TNFα" is a member of the tumor necrosis factor (TNF) family, which is mainly secreted by activated macrophages and is known to play an important role in various immune-mediated inflammatory diseases.
본 발명에서 "IL-1β"는 인터루킨(interleukine)-1 유전자 중 IL1B에 의하여 암호화되는 사이토카인 단백질을 의미하며, 세포 증식, 분화, 사멸 등 다양한 세포 활동에 관여한다.In the present invention, “IL-1β” refers to a cytokine protein encoded by IL1B among the interleukin-1 genes, and is involved in various cell activities such as cell proliferation, differentiation, and apoptosis.
본 발명에서 "CD163"은 헤모글로빈(hemoglobin)-합토글로빈(haptoglobin) 복합체에 대한 고친화성 청소제 수용체(scavenger receptor)이며, 단핵구/대식세포 계통의 세포에 대한 마커이기도 하다.In the present invention, "CD163" is a high-affinity scavenger receptor for the hemoglobin-haptoglobin complex, and is also a marker for cells of the monocyte/macrophage lineage.
본 발명에서 "CD206"은 만노스(Mannose) 수용체라고도 하며, 주로 대식세포, 미성숙 수지상 세포 및 간 내피세포의 표면에 존재하는 C형 렉틴(lectin)에 해당한다.In the present invention, "CD206" is also called mannose receptor and corresponds to C-type lectin that mainly exists on the surface of macrophages, immature dendritic cells, and liver endothelial cells.
본 발명에서 "Arginase 1(Arg1)"은 아르지네이스(arginase)를 암호화하는 유전자로, T세포 억제의 주요 매개체인 M2a 대식세포 및 골수 유래 억제 세포(MDSC)의 표지자로 알려져 있다.In the present invention, "Arginase 1 (Arg1)" is a gene encoding arginase, which is known as a marker of M2a macrophages and myeloid-derived suppressor cells (MDSC), which are major mediators of T cell suppression.
본 발명에서 "TGFβ1"은 사이토카인의 형질전환성장인자(transforming growth factor) 베타 계열의 폴리펩타이드 구성원으로서, 세포 성장, 증식, 분화 및 사멸의 조절을 포함한 다양한 기능을 수행하는 단백질이다.In the present invention, "TGFβ1" is a polypeptide member of the transforming growth factor beta family of cytokines, and is a protein that performs various functions including regulation of cell growth, proliferation, differentiation, and apoptosis.
본 발명에서 "IL-10"은 항염증 사이토카인으로, 대식세포에서 Th1 사이토카인, MHCⅡ 항원 및 공동자극 분자의 발현을 하향 조절하는 역할을 한다.In the present invention, "IL-10" is an anti-inflammatory cytokine that plays a role in downregulating the expression of Th1 cytokines, MHCⅡ antigens, and co-stimulatory molecules in macrophages.
본 발명에서 "IL-4"는 naive Th0 세포로부터 Th2 세포로의 분화를 유도하는 사이토카인으로서, 체액성 면역 및 후천성 면역의 중요한 조절자에 해당한다.In the present invention, “IL-4” is a cytokine that induces differentiation from naive Th0 cells into Th2 cells, and is an important regulator of humoral immunity and acquired immunity.
본 발명에서 "IL-13"은 IL13 유전자에 의하여 암호화되는 단백질로서, Th2 세포, CD4 세포, 비만세포 등에서 분비되는 사이토카인이다.In the present invention, “IL-13” is a protein encoded by the IL13 gene, and is a cytokine secreted from Th2 cells, CD4 cells, mast cells, etc.
본 발명에서 "Bax"는 Bcl-2 유사 단백질 4(Bcl-2-like protein 4)로도 알려져 있으며, Bcl-2와 이종이량체를 형성하고 세포사멸 활성화제로 기능한다.In the present invention, "Bax" is also known as Bcl-2-like protein 4, which forms a heterodimer with Bcl-2 and functions as an apoptosis activator.
본 발명에서 "C-Cas3"는 세포 사멸 시 절단되어 활성화되는 Caspase 3를 의미하며, PARP 및 기타 기질을 포함한 다운스트림 표적에 대한 효소 활성을 통해 세포 사멸 신호를 전파한다.In the present invention, “C-Cas3” refers to Caspase 3, which is cleaved and activated during cell death, and transmits a cell death signal through enzymatic activity toward downstream targets including PARP and other substrates.
본 발명에서 "C-PARP"는 Caspase-의존적 사멸화가 진행되는 동안 caspase에 의하여 절단되는 Poly-ADP-ribose polymerase(PARP)를 지칭한다.In the present invention, “C-PARP” refers to Poly-ADP-ribose polymerase (PARP) that is cleaved by caspase during caspase-dependent apoptosis.
본 발명에서 "Mcl-1"은 Bcl-2 단백질 계열에 속하며, 세포 사멸, 세포 주기 진행 및 미토콘드리아의 항상성을 조절하는 역할을 한다.In the present invention, "Mcl-1" belongs to the Bcl-2 protein family and plays a role in regulating cell death, cell cycle progression, and mitochondrial homeostasis.
본 발명에서 "Bcl-xL"는 Bcl-2 단백질 계열의 구성원이며, Caspase 활성화 및 세포 사멸을 유도하는 미토콘드리아의 내용물이 방출되는 것을 방지한다.In the present invention, "Bcl-xL" is a member of the Bcl-2 protein family and prevents the release of mitochondrial contents that induce caspase activation and cell death.
본 발명에서 용어 "치료"는 본 발명에 따른 조성물의 투여로 암 질환의 증세가 호전되거나 이롭게 변경하는 모든 행위를 말한다.In the present invention, the term "treatment" refers to any act of improving or beneficially changing the symptoms of a cancer disease by administering a composition according to the present invention.
본 발명의 약학적 조성물은 적절한 형태로 제제화되어 제공될 수 있다. 상기 유효성분인 배양액 이외에 추가로 약학적으로 허용 가능한 담체를 1종 이상 포함하여 제조할 수 있다.The pharmaceutical composition of the present invention may be formulated and provided in an appropriate form. In addition to the active ingredient, the culture medium, the composition may be prepared by including one or more pharmaceutically acceptable carriers.
상기 약학적으로 허용가능한 담체는 당업계에서 통상적으로 사용되는 것들, 예컨대 락토즈, 덱스트로즈, 수크로스, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로즈, 메틸 셀룰로즈, 미정질 셀룰로스, 폴리비닐 피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유 등을 포함하나 이에 국한되지 않는다. 또한, 본 발명의 약학적 조성물은 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제, 기타 약제학적으로 허용가능한 첨가제를 포함할 수 있으나, 이에 국한되지 않는다.The pharmaceutically acceptable carriers include, but are not limited to, those commonly used in the art, such as lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methyl hydroxybenzoate, propyl hydroxybenzoate, talc, magnesium stearate, and mineral oil. In addition, the pharmaceutical composition of the present invention may include, but is not limited to, diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrants, and surfactants, and other pharmaceutically acceptable additives.
본 발명의 약학적 조성물이 경구용 고형 제제로 제제화된 경우 정제, 환제, 산제, 과립제, 캡슐제 등을 포함하며, 이러한 고형제제는 적어도 하나 이상의 부형제 예를 들면, 전분, 칼슘카보네이트(calcium carbonate), 수크로스(sucrose) 또는 락토즈, 젤라틴 등을 포함할 수 있으며, 마그네슘 스테아레이트, 탈크 같은 윤활제 등을 포함하나 이에 국한되지 않는다. When the pharmaceutical composition of the present invention is formulated as a solid oral preparation, it includes tablets, pills, powders, granules, capsules, etc., and such solid preparations may include at least one excipient, for example, starch, calcium carbonate, sucrose or lactose, gelatin, etc., and include, but are not limited to, lubricants such as magnesium stearate and talc.
본 발명의 약학적 조성물이 경구용 액상 제제화된 경우 현탁제, 내용액제, 유제, 시럽제 등을 포함하며, 물, 리퀴드 파라핀 등의 희석제, 습윤제, 감미제, 방향제, 보존제 등을 포함하나 이에 국한되지 않는다. When the pharmaceutical composition of the present invention is formulated as an oral liquid, it includes a suspension, a solution, an emulsion, a syrup, etc., and includes, but is not limited to, a diluent such as water or liquid paraffin, a wetting agent, a sweetener, a fragrance, a preservative, etc.
본 발명의 약학적 조성물이 비경구용으로 제제화된 경우 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조 제제, 좌제를 포함하며, 비수성 용제, 현탁제로는 프로필렌글리콜(propylene glycol), 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르류 등을 포함하나 이에 국한되지 않는다. 좌제의 기제로는 위텝솔(witepsol), 마크로골, 트윈(tween) 61, 카카오지, 라우린지, 글리세로젤라틴 등이 사용될 수 있으나 이에 국한되지 않는다.When the pharmaceutical composition of the present invention is formulated for parenteral use, it includes a sterile aqueous solution, a non-aqueous solvent, a suspension, an emulsion, a lyophilized preparation, and a suppository. Non-aqueous solvents and suspensions include, but are not limited to, propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate. Bases for suppositories include, but are not limited to, witepsol, macrogol, tween 61, cacao butter, laurin butter, and glycerogelatin.
상기 조성물은 약학적으로 유효한 양으로 단일 또는 다중 투여될 수 있다. 본 발명의 용어 "약학적으로 유효한 양"이란 의학적 예방 또는 치료에 적용가능한 합리적인 수혜/위험 비율로 질환을 예방 또는 치료하기에 충분한 양을 의미하며, 유효 용량 수준은 질환의 중증도, 약물의 활성, 환자의 연령, 체중, 건강, 성별, 환자의 약물에 대한 민감도, 사용된 본 발명 조성물의 투여 시간, 투여 경로 및 배출 비율, 치료기간, 사용된 본 발명의 조성물과 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다.The composition may be administered in single or multiple doses in a pharmaceutically effective amount. The term "pharmaceutically effective amount" as used herein means an amount sufficient to prevent or treat a disease at a reasonable benefit/risk ratio applicable to medical prevention or treatment, and the effective dosage level may be determined according to factors including the severity of the disease, the activity of the drug, the patient's age, weight, health, sex, the patient's sensitivity to the drug, the time of administration of the composition of the present invention used, the route of administration and excretion rate, the duration of treatment, drugs combined with or used concurrently with the composition of the present invention used, and other factors well known in the medical field.
본 발명의 약학적 조성물은 랫트, 마우스, 가축, 인간 등의 포유동물에 다양한 경로로, 예를 들면, 경구 투여, 경막내, 내이, 복강 또는 정맥, 근육, 피하, 자궁 내 경막, 설하 또는 뇌혈관 내 주사에 의해 투여될 수 있으며, 이에 국한되지 않는다.The pharmaceutical composition of the present invention can be administered to mammals such as rats, mice, livestock, and humans by various routes, for example, but not limited to, oral administration, intrathecal, intra-auricular, intraperitoneal or intravenous, intramuscular, subcutaneous, intrauterine, sublingual, or intracerebrovascular injection.
또한, 본 발명의 약학적 조성물은 약학적 분야의 통상의 방법에 따라 환자의 신체 내 투여에 적합한 단위투여형의 약학적 제제로 제형화시켜 투여할 수 있으며, 상기 제제는 1회 또는 수회 투여에 의해 효과적인 투여량을 포함한다. 이러한 목적에 적합한 제형으로는 비경구투여 제제로서 주사제, 주입제 등이 바람직하다. 상기 약학적 조성물의 투여 용량은 환자의 나이, 몸무게, 성별, 투여형태, 건강상태 및 질환 정도에 따라 달라질 수 있으며, 의사 또는 약사의 판단에 따라 일정 시간간격으로 1일 1회 내지 수회로 분할 투여할 수 있다.In addition, the pharmaceutical composition of the present invention can be formulated into a unit dosage form pharmaceutical preparation suitable for administration into the patient's body according to a conventional method in the pharmaceutical field and administered, and the preparation includes an effective dosage amount through one or more administrations. Preferred dosage forms for this purpose are parenteral administration preparations such as injections and infusions. The administration dosage of the pharmaceutical composition may vary depending on the patient's age, weight, sex, dosage form, health condition, and disease severity, and may be administered once or several times a day at regular intervals according to the judgment of a doctor or pharmacist.
본 발명의 약학적 조성물의 투여 경로 및 투여 방식은 각각 독립적일 수 있으며, 그 방식에 있어 특별히 제한되지 아니하며, 목적하는 해당 부위에 상기 약학적 조성물이 도달할 수 있는 한 임의의 투여 경로 및 투여 방식에 따를 수 있다.The route and method of administration of the pharmaceutical composition of the present invention may be independent of each other, and are not particularly limited in their method, and any route and method of administration may be followed as long as the pharmaceutical composition can reach the target area.
본 발명의 용어 "투여"는 어떠한 적절한 방법으로 인간 또는 동물에게 소정의 물질을 도입하는 것을 의미하며, 본 발명에 따른 치료용 조성물의 투여 경로는 목적 조직에 도달할 수 있는 한 어떠한 일반적인 경로를 통하여 경구 또는 비경구 투여될 수 있다. 또한, 본 발명에 따른 치료용 조성물은 유효성분이 표적 세포로 이동할 수 있는 임의의 장치에 의해 투여될 수 있다.The term "administration" in the present invention refers to introducing a given substance into a human or animal by any suitable method. The therapeutic composition according to the present invention may be administered orally or parenterally via any common route, as long as it can reach the target tissue. Furthermore, the therapeutic composition according to the present invention may be administered by any device capable of transporting the active ingredient to target cells.
본 발명은 암 질환 치료제의 스크리닝 방법을 제공한다.The present invention provides a method for screening a cancer treatment agent.
구체적으로, 본 발명에 따른 암 질환 치료제의 스크리닝 방법은 하기의 단계를 포함한다:Specifically, the screening method for a cancer treatment agent according to the present invention comprises the following steps:
(a) 암 유발 물질 또는 암세포를 인간을 제외한 실험동물에 투여하는 단계;(a) a step of administering a carcinogenic substance or cancer cells to an experimental animal other than a human;
(b) 시험물질을 상기 실험동물에 투여하는 단계; 및(b) a step of administering a test substance to the experimental animal; and
(c) 상기 실험동물의 세포에서 종양 억제성 대식세포(M1 TAM) 마커 및 종양 지지성 대식세포(M2 TAM) 마커의 발현 수준을 확인하는 단계.(c) A step of confirming the expression level of tumor suppressive macrophage (M1 TAM) marker and tumor supporting macrophage (M2 TAM) marker in the cells of the above experimental animal.
본 발명은 (a) 암 유발 물질 또는 암세포를 인간을 제외한 실험동물에 투여하는 단계를 포함한다.The present invention comprises a step of (a) administering a carcinogenic substance or cancer cells to an experimental animal other than a human.
본 발명에서는 암 질환을 유발한 동물 모델을 제조하기 위하여, 암 유발 물질로 알려진 어떠한 물질이라도 사용할 수 있다. 구체적으로, 비소, 벤젠, 베릴륨, 카드뮴, 6가크롬화합물, 에틸렌옥사이드, 니켈, 라돈 또는 염화 비닐 등을 사용하여 암 질환을 유발할 수 있다. 예컨대, 비소, 벤젠, 베릴륨, 카드뮴, 6가크롬화합물, 에틸렌옥사이드, 니켈, 라돈 및/또는 염화 비닐을 당업계에서 통상적으로 이용되는 어떤 실험동물에 처리하고, 이로 인해 질환이 유도된 동물 모델 그 자체 또는 이로부터 분리된 세포를 사용할 수 있다. 본 실험 동물모델은 예를 들어 마우스, 햄스터, 랫트, 페렛, 기니피그, 토끼, 개, 영장류, 돼지 등을 사용할 수 있으며, 보다 바람직하게는 마우스일 수 있다. 본 실험동물은 인간을 제외한다. In the present invention, any substance known to be a carcinogen can be used to produce an animal model that induces cancer. Specifically, arsenic, benzene, beryllium, cadmium, hexavalent chromium compounds, ethylene oxide, nickel, radon, or vinyl chloride can be used to induce cancer. For example, any experimental animal commonly used in the art can be treated with arsenic, benzene, beryllium, cadmium, hexavalent chromium compounds, ethylene oxide, nickel, radon, and/or vinyl chloride, and the animal model itself or cells isolated therefrom in which the disease is induced can be used. The experimental animal model can be, for example, a mouse, hamster, rat, ferret, guinea pig, rabbit, dog, primate, or pig, and more preferably a mouse. The experimental animal excludes humans.
또한 상기 암 유발을 위한 임의의 암세포가 투여될 수 있다. 예를 들어, 섬유육종, 악성섬유성조직구종, 지방육종, 횡문 근육종, 평활근육종, 혈관육종, 악성피부암, 림프혈관육종, 활막육종, 연골육종, 골육종, 폐암, 위암, 유방암, 대장암 및 전립선암로 이루어진 군으로부터 선택되는 어느 하나 이상의 암세포일 수 있다. 이러한 암세포의 투여는 각각의 실험 동물에 따라 통상적으로 암을 유발할 수 있는 것으로 알려진 수준의 세포 수를 투여하는 것일 수 있다. Additionally, any cancer cell for inducing the above cancer may be administered. For example, the cancer cells may be one or more selected from the group consisting of fibrosarcoma, malignant fibrous histiocytoma, liposarcoma, rhabdomyosarcoma, leiomyosarcoma, hemangiosarcoma, malignant skin cancer, lymphangiosarcoma, synovial sarcoma, chondrosarcoma, osteosarcoma, lung cancer, gastric cancer, breast cancer, colon cancer, and prostate cancer. The administration of such cancer cells may be administered in an amount typically known to induce cancer in each experimental animal.
상기 (a)단계에서 암 질환 유발 물질 또는 암세포은 특성에 맞는 경로로 투여될 수 있으며, 예를 들어, 호흡, 근육주사, 신장주사, 복강내 주사 또는 피하 주사 등을 통해 투여될 수 있다. In the above step (a), the cancer-causing substance or cancer cells may be administered through a route appropriate to their characteristics, for example, through respiration, intramuscular injection, renal injection, intraperitoneal injection, or subcutaneous injection.
본 발명에 따른 생체 내에서 암 질환이 유발되지 않은 상태와 암 질환이 유발된 상태에서 발현 또는 활성화되는 물질들은 서로 다르다. 따라서 암 질환 예방 및 치료용 후보 물질을 스크리닝하기 위해서는 일단 암 질환을 유발할 필요가 있다.According to the present invention, the substances expressed or activated in vivo in non-cancer and cancer-induced states are different. Therefore, to screen for candidate substances for cancer prevention and treatment, cancer must first be induced.
본 발명에 따른 암 질환 치료제의 스크리닝 방법은 (b) 시험물질을 상기 실험동물에게 투여하는 단계를 포함한다.The method for screening a cancer treatment agent according to the present invention comprises the step of (b) administering a test substance to the experimental animal.
본 발명에 있어서 "시험물질"이란 유전자의 발현량에 영향을 미치거나, 단백질의 발현 또는 활성에 영향을 미치는지 여부를 검사하기 위하여 스크리닝에서 이용되는 미지의 후보 물질을 의미한다. 상기 시료는 화학물질, 뉴클레오타이드, 안티센스-RNA, siRNA(small interference RNA) 및 천연물 추출물을 포함하나, 이에 제한되는 것은 아니다.In the present invention, "test material" refers to an unknown candidate substance used in screening to determine whether it affects the expression level of a gene or the expression or activity of a protein. The sample includes, but is not limited to, chemicals, nucleotides, antisense RNA, siRNA (small interference RNA), and natural product extracts.
이러한 시험물질의 투여는 경구 및 비경구 투여를 포함하여, 통상의 투여 경로를 통해 투여될 수 있다. 예컨대, 경구 투여될 수 있으며, 비경구 투여 시 피부 외용 또는 복강 내 주사, 직장 내 주사, 피하주사, 정맥주사, 근육 내 주사 또는 흉부 내 주사 주입방식으로부터 선택될 수 있으나, 이에 제한되지 않는다.These test substances can be administered via conventional routes of administration, including oral and parenteral administration. For example, oral administration may be used, and parenteral administration may be selected from, but is not limited to, topical application to the skin, intraperitoneal injection, rectal injection, subcutaneous injection, intravenous injection, intramuscular injection, or intrathoracic injection.
시험물질의 투여량은 약물의 종류 및 약물 효능에 따라 당업자의 통상의 지식에 따라 적절한 수준의 투여량으로 조절될 수 있다.The dosage of the test substance can be adjusted to an appropriate level of dosage according to the type of drug and the drug efficacy, based on the general knowledge of those skilled in the art.
본 발명에 따른 암 질환의 스크리닝 방법은 (c) 상기 실험동물의 세포에서 종양 억제성 대식세포(M1 TAM) 마커 및 종양 지지성 대식세포(M2 TAM) 마커의 발현 수준을 확인하는 단계를 포함한다.The method for screening for cancer according to the present invention comprises the step of (c) confirming the expression level of a tumor suppressive macrophage (M1 TAM) marker and a tumor supporting macrophage (M2 TAM) marker in cells of the experimental animal.
본 발명에 따른 암 질환의 스크리닝 방법은 상기 (c) 단계에서 (i) 종양 억제성 대식세포(M1 TAM) 마커의 발현 수준이 증가하고, (ii) 종양 지지성 대식세포(T2 TAM) 마커의 발현 수준이 감소하는 시험물질을 선택하는 것을 포함할 수 있다.The method for screening for cancer according to the present invention may include selecting a test substance that (i) increases the expression level of a tumor suppressive macrophage (M1 TAM) marker and (ii) decreases the expression level of a tumor supportive macrophage (T2 TAM) marker in step (c).
본 발명에서 "종양 관련 대식세포(tumor-associated macrophages, TAM)"란, 종양 미세환경의 형성에 관여하는 대식세포로서, 다양한 사이토카인, 케모카인, 단백질 분해효소들을 분비함으로써 종양의 신생혈관형성, 증식, 전이 및 면역억제 등에 영향을 미친다. 상기 종양 관련 대식세포는 종양 억제성 대식세포(M1 TAM)와 종양 지지성 대식세포(M2 TAM)로 나뉜다.In the present invention, "tumor-associated macrophages (TAMs)" are macrophages that participate in the formation of a tumor microenvironment, and influence tumor angiogenesis, proliferation, metastasis, and immunosuppression by secreting various cytokines, chemokines, and proteolytic enzymes. The tumor-associated macrophages are divided into tumor-suppressive macrophages (M1 TAMs) and tumor-supportive macrophages (M2 TAMs).
상기 종양 억제성 대식세포(M1 TAM) 마커는 CD16/32, CD80, H2Ab1, MHCII, iNOS(NOS2), IFNγ, IL12p40, TNFα 및 IL-1β로 구성된 군으로부터 선택된 어느 하나 이상일 수 있지만, 이에 제한되는 것은 아니다.The above tumor suppressive macrophage (M1 TAM) marker may be any one or more selected from the group consisting of CD16/32, CD80, H2Ab1, MHCII, iNOS (NOS2), IFNγ, IL12p40, TNFα, and IL-1β, but is not limited thereto.
상기 종양 지지성 대식세포(M2 TAM) 마커는 CD163, CD206, Arginase 1(Arg1), TGFβ1, IL-10, IL-4 및 IL-13으로 구성된 군으로부터 선택된 어느 하나 이상일 수 있지만, 이에 제한되는 것은 아니다.The above tumor-supporting macrophage (M2 TAM) marker may be any one or more selected from the group consisting of, but is not limited to, CD163, CD206, Arginase 1 (Arg1), TGFβ1, IL-10, IL-4, and IL-13.
시험물질 처리에 따라 CD16/32, CD80, H2Ab1, MHCII, iNOS(NOS2), IFNγ, IL12p40, TNFα 및 IL-1β로 구성된 군으로부터 선택된 어느 하나 이상이 증가할 경우, 암 질환에 대한 치료 효능을 가지는 약물로 판단될 수 있다. 또한, 양성 대조군과 동등한 수준으로 종양 억제성 대식세포(M1 TAM) 마커가 발현되면 암 질환에 대한 치료 효능을 가지는 약물로 판단될 수 있다.If at least one of the following markers increases following treatment with the test substance: CD16/32, CD80, H2Ab1, MHCII, iNOS (NOS2), IFNγ, IL12p40, TNFα, and IL-1β, the drug may be considered to have therapeutic efficacy against cancer. Furthermore, if the tumor suppressive macrophage (M1 TAM) marker is expressed at a level comparable to that of the positive control, the drug may be considered to have therapeutic efficacy against cancer.
한편, 시험물질 처리에 따라 CD163, CD206, Arginase 1(Arg1), TGFβ1, IL-10, IL-4 및 IL-13로 구성된 군으로부터 선택된 어느 하나 이상이 감소할 경우, 암 질환에 대한 치료 효능을 가지는 약물로 판단될 수 있다. 또한, 양성 대조군과 동등한 수준으로 종양 지지성 대식세포(M2 TAM) 마커가 발현되면 암 질환에 대한 치료 효능을 가지는 약물로 판단될 수 있다.Meanwhile, if at least one selected from the group consisting of CD163, CD206, Arginase 1 (Arg1), TGFβ1, IL-10, IL-4, and IL-13 decreases following treatment with the test substance, the drug may be judged to have therapeutic efficacy against cancer. In addition, if the tumor-supporting macrophage (M2 TAM) marker is expressed at a level equivalent to that of the positive control, the drug may be judged to have therapeutic efficacy against cancer.
본 발명에 있어서, 대조군 시료는 정상 대조군, 암 질환 치료제에 대해 알려진 치료물질 대조군을 포함하여 시험물질 처리에 따라 종양 억제성 대식세포(M1 TAM) 마커 및 종양 지지성 대식세포(M2 TAM) 마커의 발현 수준을 비교 판단할 수 있는 모든 시료 그룹을 의미한다.In the present invention, a control sample means all sample groups for which the expression levels of tumor suppressive macrophage (M1 TAM) markers and tumor supportive macrophage (M2 TAM) markers can be compared and determined according to test substance treatment, including a normal control group and a therapeutic substance control group known for treating cancer.
상기 종양 억제성 대식세포(M1 TAM) 마커 및 종양 지지성 대식세포(M2 TAM) 마커의 발현 수준은 효소면역분석법(ELISA), 정략적 실시간 중합효소연쇄반응(qPCR), 면역조직화학 및 웨스턴 블랏(Western Blotting), 유세포 분석으로 구성된 군으로부터 선택된 어느 하나로 측정될 수 있지만, 이에 제한되는 것은 아니다.The expression levels of the above tumor suppressive macrophage (M1 TAM) marker and tumor supporting macrophage (M2 TAM) marker can be measured by any one selected from the group consisting of, but not limited to, enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qPCR), immunohistochemistry and Western blotting, and flow cytometry.
본 발명에 따른 암 질환의 스크리닝 방법은 (d) 상기 실험동물의 세포에서 (i) 친-사멸화(pro-apoptotic) 마커의 발현 수준 증가, 및 (ii) 항-사멸화(anti-apoptotic) 마커의 발현 수준 감소를 확인하는 단계;를 더 포함할 수 있다.The method for screening for cancer according to the present invention may further include the step of (d) confirming (i) an increase in the expression level of a pro-apoptotic marker and (ii) a decrease in the expression level of an anti-apoptotic marker in cells of the experimental animal.
상기 친-사멸화(pro-apoptotic) 마커는 Bax, C-Cas3 및 C-PARP로 구성된 군으로부터 선택된 어느 하나 이상일 수 있으며, 상기 항-사멸화(anti-apoptotic) 마커는 Mcl-1 및 Bcl-xL로 구성된 군으로부터 선택된 어느 하나 이상일 수 있다.The pro-apoptotic marker may be at least one selected from the group consisting of Bax, C-Cas3, and C-PARP, and the anti-apoptotic marker may be at least one selected from the group consisting of Mcl-1 and Bcl-xL.
본 발명에 따른 암 질환의 스크리닝 방법에서 상기 시험물질은 암 관련 섬유아세포(cancer-associated fibroblast, CAF) 및 사멸화된 암세포(apoptotic cancer cells)를 공동배양한 배양액을 포함하는 약학적 조성물일 수 있다.In the screening method for cancer according to the present invention, the test substance may be a pharmaceutical composition including a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured.
본 발명은 (a) 암세포에서 시험물질을 접촉시키는 단계;The present invention comprises the steps of: (a) contacting a test substance with cancer cells;
(b) 상기 시험물질을 접촉한 암세포에서 종양 억제성 대식세포(M1 TAM) 마커 및 종양 지지성 대식세포(M2 TAM) 마커의 발현 수준을 확인하는 단계; 및(b) a step of confirming the expression level of tumor suppressive macrophage (M1 TAM) marker and tumor supporting macrophage (M2 TAM) marker in cancer cells that have come into contact with the test substance; and
(c) 대조군 시료와 비교하여 (i) 상기 종양 억제성 대식세포(M1 TAM) 마커의 발현 수준이 증가하고, (ii) 상기 종양 지지성 대식세포(M2 TAM) 마커의 발현 수준이 감소한 시험물질을 선별하는 단계;를 포함하는 암 질환 치료제의 스크리닝 방법을 제공한다.(c) a method for screening a cancer treatment agent, comprising the step of selecting a test substance having (i) an increased expression level of the tumor suppressive macrophage (M1 TAM) marker and (ii) a decreased expression level of the tumor supportive macrophage (M2 TAM) marker compared to a control sample;
상기 암세포는 임의의 암세포가 사용될 수 있으며, 앞서 언급된 암세포의 일 종류라면 본 스크리닝 방법에 또한 동등하게 적용 가능하다. Any cancer cell can be used as the cancer cell, and any type of cancer cell mentioned above can be equally applied to the present screening method.
본 발명의 암 질환 치료제의 스크리닝 방법은 암세포에 시험물질을 접촉시키는 단계를 포함한다. 바람직하게는, 상기 암세포는 암 질환을 가진 환자의 세포, 동물 세포, 이들의 조직을 모두 포함하는 것이다. The method for screening a cancer treatment agent of the present invention comprises a step of contacting a test substance with cancer cells. Preferably, the cancer cells include cells from a patient with cancer, animal cells, or tissues thereof.
또한, 암 질환 유발 물질을 통해 질환이 유도된 세포도 이용 가능하며, 암 질환 유발 물질로 알려진 어떠한 물질도 사용 가능하다. 바람직하게는 비소, 벤젠, 베릴륨, 카드뮴, 6가크롬화합물, 에틸렌옥사이드, 니켈, 라돈 또는 염화 비닐 처리에 의해 유도된 암세포일 수 있다. 예컨대, 비소, 벤젠, 베릴륨, 카드뮴, 6가크롬화합물, 에틸렌옥사이드, 니켈, 라돈 또는 염화 비닐을 실험동물로 당업계에서 통상적으로 이용되는 어떤 실험동물에 처리하고, 이로 인해 질환이 유도된 동물 모델 그 자체 또는 이로부터 분리된 세포를 사용할 수 있다. Additionally, cells induced with cancer-causing substances can be utilized, and any substance known to be a cancer-causing substance can be utilized. Preferably, the cancer cells can be induced by treatment with arsenic, benzene, beryllium, cadmium, hexavalent chromium compounds, ethylene oxide, nickel, radon, or vinyl chloride. For example, any laboratory animal commonly used in the art can be treated with arsenic, benzene, beryllium, cadmium, hexavalent chromium compounds, ethylene oxide, nickel, radon, or vinyl chloride, and the resulting animal model, or cells isolated therefrom, can be used.
상기 시험물질의 접촉은 in vitro 및 in vivo 상의 모든 접촉을 의미하며, 시험물질이 in vitro 상에서 세포에 처리되거나 in vivo 상에서 동물 등에 투여되는 것을 모두 포함한다.Contact with the above test substance means all contacts in vitro and in vivo , and includes all contacts in which the test substance is treated on cells in vitro or administered to animals, etc. in vivo .
본 발명에 따른 암 질환 치료제의 스크리닝 방법은 상기 시험물질을 접촉한 암세포에서 종양 억제성 대식세포(M1 TAM) 마커 및 종양 지지성 대식세포(M2 TAM) 마커의 발현 수준을 확인하는 단계; 및 대조군 시료와 비교하여 (i) 상기 종양 억제성 대식세포(M1 TAM) 마커의 발현 수준이 증가하고, (ii) 상기 종양 지지성 대식세포(M2 TAM) 마커의 발현 수준이 감소한 시험물질을 선별하는 단계;를 포함한다.The method for screening a cancer treatment agent according to the present invention comprises the steps of: confirming the expression levels of tumor suppressive macrophage (M1 TAM) markers and tumor supportive macrophage (M2 TAM) markers in cancer cells that have come into contact with the test substance; and selecting a test substance in which (i) the expression level of the tumor suppressive macrophage (M1 TAM) marker increases and (ii) the expression level of the tumor supportive macrophage (M2 TAM) marker decreases compared to a control sample.
바람직하게는, 상기 종양 억제성 대식세포(M1 TAM) 마커 및 종양 지지성 대식세포(M2 TAM) 마커의 발현 수준은 효소면역분석법(ELISA), 정략적 실시간 중합효소연쇄반응(qPCR), 면역조직화학, 웨스턴 블랏(Western Blotting) 및 유세포 분석으로 구성된 군으로부터 선택된 어느 하나로 측정될 수 있지만, 이에 제한되는 것은 아니다.Preferably, the expression levels of the tumor suppressive macrophage (M1 TAM) marker and the tumor supporting macrophage (M2 TAM) marker can be measured by any one selected from the group consisting of, but not limited to, enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qPCR), immunohistochemistry, Western blotting, and flow cytometry.
본 발명의 암 질환 치료용 조성물은 M2 대식세포의 생존을 억제하고, M2 TAM으로부터 M1 TAM으로의 리프로그래밍을 유도하여 종양의 성장을 억제함으로써 암 질환을 치료할 수 있다. 또한, 본 발명의 스크리닝 방법은 다수의 치료 후보 물질 중 암 질환 치료에 효과적인 물질을 손쉽게 확인하고 이를 획득할 수 있게 함으로써 신규의 암 질환 치료용 약물 개발 및 확인에 유용하게 이용될 수 있다.The composition for treating cancer of the present invention can treat cancer by suppressing the survival of M2 macrophages and inducing reprogramming of M2 TAMs into M1 TAMs, thereby inhibiting tumor growth. Furthermore, the screening method of the present invention can be usefully utilized in the development and identification of novel cancer treatment drugs by easily identifying and obtaining substances effective in treating cancer among a large number of therapeutic candidates.
도 1은 ApoSQ-CAF CM의 투여에 의하여 원발성 종양의 총 TAM 밀도가 감소함을 나타낸 도이다.Figure 1 is a diagram showing that the total TAM density of a primary tumor is reduced by administration of ApoSQ-CAF CM.
(a) 및 (b)의 왼쪽: 원발성 종양의 중앙(central) 및 가장자리(marginal) 영역에서 범-대식세포(pan-macrophage) 마커인 CD11b(빨간색), F4/80(빨간색) 및 DAPI(파란색)을 이용한 면역형광염색 결과를 나타낸 도이다(scale bar: 100 μm).(a) and (b) Left: Immunofluorescence staining using pan-macrophage markers CD11b (red), F4/80 (red), and DAPI (blue) in the central and marginal areas of the primary tumor (scale bar: 100 μm).
(a) 및 (b)의 오른쪽: 원발성 종양의 중앙 및 가장자리 영역의 TAM 밀도를 정량화 하여 나타낸 도이다(**P < 0.01, ***P < 0.001, Kruskal-Wallis test with Dunn's post hoc test, 데이터는 각 그룹당 세 마리의 마우스에서 얻은 평균 ± 표준 오차로 표시됨).(a) and (b) Right: Quantification of TAM density in the central and marginal regions of primary tumors (**P < 0.01, ***P < 0.001, Kruskal-Wallis test with Dunn's post hoc test, data are presented as mean ± standard error from three mice per group).
도 2는 ApoSQ-CAF CM의 투여에 의하여 원발성 종양의 M2 TAM 분율이 감소함을 나타낸 도이다.Figure 2 is a diagram showing that the M2 TAM fraction of primary tumors is reduced by administration of ApoSQ-CAF CM.
(a) 내지 (c)의 위쪽: M2 마커인 Arg1 및 CD206, 범-대식세포(pan-macrophage) 마커인 CD11b(빨간색) 및 F4/80(빨간색)을 이용한 면역형광염색 결과를 나타낸 도이다(scale bar: 100 μm).(a) to (c) Top: Immunofluorescence staining results using M2 markers Arg1 and CD206, and pan-macrophage markers CD11b (red) and F4/80 (red) (scale bar: 100 μm).
(a) 내지 (c)의 아래쪽: Arg1+ 및 CD206+ TAM의 밀도 및 M2 TAM의 분율을 정량화 하여 나타낸 도이다. M2 TAM 분율은 CD11b+ 또는 F4/80+ TAM 내에서 M1 및 M2 TAM의 백분율로 결정되었다(**P < 0.05, ***P < 0.001, Kruskal-Wallis test with Dunn's post hoc test).(a) to (c) Bottom: Quantification of the density of Arg1 + and CD206 + TAMs and the fraction of M2 TAMs. The M2 TAM fraction was determined as the percentage of M1 and M2 TAMs within CD11b + or F4/80 + TAMs (**P < 0.05, ***P < 0.001, Kruskal-Wallis test with Dunn's post hoc test).
도 3은 ApoSQ-CAF CM의 투여에 의하여 원발성 종양의 M1 TAM 분율이 증가함을 나타낸 도이다.Figure 3 is a diagram showing that the M1 TAM fraction of primary tumors increases by administration of ApoSQ-CAF CM.
(a) 내지 (c)의 위쪽: M1 마커인 iNOS(녹색) 및 CD16/32(녹색), 범-대식세포(pan-macrophage) 마커인 CD11b(빨간색) 및 F4/80(빨간색)을 이용한 면역형광염색 결과를 나타낸 도이다(scale bar: 100 μm).(a) to (c) Top: Immunofluorescence staining results using M1 markers iNOS (green) and CD16/32 (green), and pan-macrophage markers CD11b (red) and F4/80 (red) (scale bar: 100 μm).
(a) 내지 (c)의 아래쪽: iNOS+ 및 CD16/32+ TAM의 밀도 및 M1 TAM의 분율을 정량화 하여 나타낸 도이다. M1 TAM 분율은 CD11b+ 또는 F4/80+ TAM 내에서 M1 및 M2 TAM의 백분율로 결정되었다(**P < 0.05, ***P < 0.001, Kruskal-Wallis test with Dunn's post hoc test).(a) to (c) Bottom: Quantification of the density of iNOS + and CD16/32 + TAMs and the fraction of M1 TAMs. The M1 TAM fraction was determined as the percentage of M1 and M2 TAMs within CD11b + or F4/80 + TAMs (**P < 0.05, ***P < 0.001, Kruskal-Wallis test with Dunn's post hoc test).
도 4는 WISP-1에 의하여 세포 사멸 촉진 효과가 매개됨을 확인한 도이다.Figure 4 is a diagram confirming that the cell death promotion effect is mediated by WISP-1.
(a) 내지 (c)의 왼쪽: 세포 사멸 마커인 C-Cas3(빨간색), M2 TAM 마커인 CD206(녹색), M1 TAM 마커인 CD16/32(녹색), 범-대식세포(pan-macrophage) 마커인 CD11b(빨간색), 및 DAPI(파란색)을 이용한 면역형광염색 결과를 나타낸 도이다(scale bar: 100 μm).Left of (a) to (c): Immunofluorescence staining results using C-Cas3 (red), an apoptosis marker, CD206 (green), an M2 TAM marker, CD16/32 (green), an M1 TAM marker, CD11b (red), a pan-macrophage marker, and DAPI (blue) (scale bar: 100 μm).
(a) 내지 (c)의 오른쪽: CD206+, CD16/32+ 또는 CD11b+ TAM에서 C-Cas3+ 세포의 분율을 정량화 하여 나타낸 도이다. (**P < 0.01, ***P < 0.001, Kruskal-Wallis test with Dunn's post hoc test).Right side of (a) to (c): Quantification of the fraction of C-Cas3 + cells in CD206 + , CD16/32 + , or CD11b + TAMs. (**P < 0.01, ***P < 0.001, Kruskal-Wallis test with Dunn's post hoc test).
도 5는 ApoSQ-CAF CM의 투여에 의하여 M2 TAM으로부터 M1 TAM으로의 리프로그래밍이 유도됨을 확인한 도이다.Figure 5 is a diagram confirming that reprogramming from M2 TAM to M1 TAM is induced by administration of ApoSQ-CAF CM.
(a) 원발성 종양에서 분리된 CD11b+ TAM에서 발현된 M1 및 M2 마커 관련 유전자의 발현 차이를 보여주는 히트맵(왼쪽, 빨간색은 고발현, 파란색은 저발현을 나타냄)과 대식세포 분극 마커에 대한 PCR 어레이 분석 결과 중 일부 유전자의 상대적 발현을 보여주는 그래프(오른쪽)이다. Log2 폴드 변화 값은 ApoSQ-CAF CM 대비 CAF CM을 기준으로 한다(NS, not significant; **P < 0.01, ***P < 0.001, two-tailed Student's t-test.).(a) Heatmap showing the differences in expression of genes related to M1 and M2 markers expressed in CD11b + TAMs isolated from primary tumors (left; red indicates high expression, blue indicates low expression) and graph showing the relative expression of some genes from PCR array analysis for macrophage polarization markers (right). Log2 fold change values are relative to CAF CM versus ApoSQ-CAF CM (NS, not significant; **P < 0.01, ***P < 0.001, two-tailed Student's t-test.).
(b) 원발성 종양에서 분리된 CD11b+ TAM에서 M2 마커(Arg1, CD206, CD163, IL-4, IL-10, TGF-β1) 및 M1 마커(TNFα, CD80, H2Ab1, NOS2, IFNγ 및 IL-12 p40)의 상대적 mRNA 수준을 qRT-PCR을 통해 확인한 도이다(NS, not significant; *P < 0.05, **P < 0.01, ***P < 0.001, Analysis of variance with Tukey's post hoc test.).(b) Relative mRNA levels of M2 markers (Arg1, CD206, CD163, IL-4, IL-10, TGF-β1) and M1 markers (TNFα, CD80, H2Ab1, NOS2, IFNγ, and IL-12 p40) in CD11b + TAMs isolated from primary tumors were confirmed by qRT-PCR (NS, not significant; *P < 0.05, **P < 0.01, ***P < 0.001, Analysis of variance with Tukey's post hoc test.).
(c) 원발성 종양에서 분리된 CD11b+ TAM에서 Arg1, CD206, iNOS 및 CD16/32의 면역블로팅을 실시한 결과를 나타낸 도이다.(c) A diagram showing the results of immunoblotting of Arg1, CD206, iNOS, and CD16/32 in CD11b + TAM isolated from primary tumors.
(d, e) 원발성 종양에서 분리된 CD11b+ TAM에서 M2 TAM(CD163+ 및 CD206+) 및 M1 TAM(CD16+ 및 CD80+)에 대한 유세포 분석을 실시한 결과를 나타낸 도이다(**P < 0.01, two-tailed Student's t-test) 우측은 평균 형광 강도(MFI, Mean Fluorescence Intensity) 값이다.(d, e) Flow cytometry analysis of M2 TAM (CD163 + and CD206 + ) and M1 TAM (CD16 + and CD80 + ) in CD11b + TAM isolated from primary tumors (**P < 0.01, two-tailed Student's t-test). The right side shows the mean fluorescence intensity (MFI) value.
(f) 위쪽은 CD11b+TAM에 대한 대표적인 유세포 분석 도표이고, 아래쪽은 TAM 비율(CD163+/MHCII+ TAM 비율)을 나타낸다.(f) The upper part is a representative flow cytometry plot for CD11b + TAM, and the lower part shows the TAM ratio (CD163 + /MHCII + TAM ratio).
(g-j) 각각 (g) M2 대식세포, (h) 조절 T 세포(Tregs), (i) M1 대식세포, (j) CD8 T+ 세포 집단에 대한 유세포 분석 결과이다. 림프단핵세포(lymphomononuclear cells)는 CD45, CD11b, CD3, CD4, CD8, FoxP3, MHCII, Ly6C 항체로 염색하였으며, 각 세포 유형의 절대 수치는 유세포 분석을 통해 계산되었다(NS, not significant; **P < 0.01, ***P < 0.001, two-tailed Student's t-test.).(g-j) Flow cytometric analysis results for (g) M2 macrophages, (h) regulatory T cells (Tregs), (i) M1 macrophages, and (j) CD8 T + cell populations, respectively. Lymphomononuclear cells were stained with CD45, CD11b, CD3, CD4, CD8, FoxP3, MHCII, and Ly6C antibodies, and the absolute numbers of each cell type were calculated by flow cytometry (NS, not significant; **P < 0.01, ***P < 0.001, two-tailed Student's t-test.).
(a-j) 모든 데이터는 조건당 3회 반복 수행하였고, 각 반복 실험마다 1마리 내지 3마리의 마우스에서 채취한 세포를 사용하였다.(a-j) All data were repeated three times per condition, and cells from one to three mice were used for each repeated experiment.
(k, l) 항-인산화 STAT1(빨간색), 항-CD206 항체(녹색), 항-CD16/32 항체(녹색) 및 DAPI(파란색)로 염색된 원발성 종양의 공초점 이미지를 나타낸 도(scale bar: 100 μm)와, CD206+ 또는 CD16/32+ 세포 중 인산화된 STAT1+ 세포를 정량화 하여 나타낸 도(각각 k의 오른쪽, l의 왼쪽)이다(NS, not significant; ***P < 0.001, Analysis of variance with Tukey's post hoc test.).(k, l) Confocal images of primary tumors stained with anti-phosphorylated STAT1 (red), anti-CD206 antibody (green), anti-CD16/32 antibody (green), and DAPI (blue) (scale bar: 100 μm), and quantification of phosphorylated STAT1 + cells among CD206 + or CD16/32 + cells (right in k, left in l, respectively) (NS, not significant; ***P < 0.001, Analysis of variance with Tukey's post hoc test.).
도 6은 M1 및 M2 대식세포로의 분극화(polarization)를 나타낸 도이다.Figure 6 is a diagram showing polarization into M1 and M2 macrophages.
(a) 및 (b) THP-1 세포 및 마우스 BMDM로부터 M1 및 M2 대식세포를 분극화 하는 것을 나타낸 모식도이다.(a) and (b) Schematic diagram showing polarization of M1 and M2 macrophages from THP-1 cells and mouse BMDM.
(c) 및 (d) THP-1 세포 및 BMDM에서 유래한 M1 및 M2 대식세포(M2)에 대하여 M2 마커(CD163, CD206 및 Arginase1(Arg1)) 및 M1 마커(MHCII, iNOS 및 IL12p40)를 이용하여 면역블로팅을 실시한 결과를 나타낸 도이다.(c) and (d) are diagrams showing the results of immunoblotting using M2 markers (CD163, CD206, and Arginase1 (Arg1)) and M1 markers (MHCII, iNOS, and IL12p40) on M1 and M2 macrophages (M2) derived from THP-1 cells and BMDM.
(e) 및 (f) M1 마커 CD86 및 M2 마커 CD163에 대한 면역형광염색을 실시한 결과를 나타낸 도이다.(e) and (f) are diagrams showing the results of immunofluorescence staining for the M1 marker CD86 and the M2 marker CD163.
도 7은 사멸화된 암세포에 노출된 CAF의 CM 투여에 의하여 M2 대식세포의 사멸이 증가함을 시험관 내(in vitro)에서 확인한 도이다.Figure 7 is a diagram confirming in vitro that the apoptosis of M2 macrophages increases by CM administration of CAFs exposed to killed cancer cells.
(a) 및 (b) THP-1 세포 및 BMDM에서 유래한 M1 및 M2 대식세포의 세포 생존율을 확인한 도이다.(a) and (b) Cell viability of M1 and M2 macrophages derived from THP-1 cells and BMDMs is shown.
(c) 및 (d)의 왼쪽: annexin V-FITC 및 PI 염색 후 유세포 분석을 실시한 결과를 나타낸 도이다.(c) and (d) Left: Diagram showing the results of flow cytometry analysis after annexin V-FITC and PI staining.
(c) 및 (d)의 오른쪽: 세포 사멸 초기 및 후기 단계의 백분율의 합으로 세포 사멸을 정량화 하여 나타낸 도이다(**P < 0.01, ***P < 0.001, two-tailed Student's t-test).Right side of (c) and (d): Cell death is quantified as the sum of the percentages of early and late stages of cell death (**P < 0.01, ***P < 0.001, two-tailed Student's t-test).
(e) THP-1 유래 M2 대식세포에서 Bax, Mcl-1, Bcl-xL, C-Cas3, C-PARP 및 β-actin을 이용하여 면역블로팅을 실시한 결과를 나타낸 도이다.(e) A diagram showing the results of immunoblotting using Bax, Mcl-1, Bcl-xL, C-Cas3, C-PARP, and β-actin in THP-1-derived M2 macrophages.
(f) 무혈청 배지, CFA CM, ApoA(사멸화된 A549 cells)-CAF CM 또는 NecA-CAF CM을 처리하였을 때의 THP-1 유래 M1 및 M2 대식세포의 세포 생존율을 나타낸 도이다.(f) A diagram showing the cell viability of THP-1-derived M1 and M2 macrophages when treated with serum-free medium, CFA CM, ApoA (killed A549 cells)-CAF CM, or NecA-CAF CM.
(g)의 왼쪽: annexin V-FITC 및 PI 염색 후 유세포 분석을 실시한 결과를 나타낸 도이다.(g) Left: A diagram showing the results of flow cytometry analysis after annexin V-FITC and PI staining.
(g)의 오른쪽: 세포 사멸 초기 및 후기 단계의 백분율의 합으로 세포 사멸을 정량화 하여 나타낸 도이다(**P < 0.01, ***P < 0.001, two-tailed Student's t-test).(g) Right: A graph showing the quantification of cell death as the sum of the percentages of early and late stages of cell death (**P < 0.01, ***P < 0.001, two-tailed Student's t-test).
도 8은 사멸화된 암세포에 노출된 CAF의 CM 투여에 의하여 M2 TAM으로부터 M1 TAM으로의 리프로그래밍이 유도됨을 시험관 내(in vitro)에서 확인한 도이다.Figure 8 is a diagram confirming in vitro that reprogramming from M2 TAM to M1 TAM is induced by CM administration of CAF exposed to killed cancer cells.
(a) 및 (b) THP-1 및 BMDM에서 유래한 M2 대식세포에서 M1 마커(NOS2, MHCII 및 IL12p40) 및 M2 마커(TGFβ1, IL10 및 IL4)의 상대적 mRNA 수준을 qRT-PCR을 통해 확인한 도이다.(a) and (b) Relative mRNA levels of M1 markers (NOS2, MHCII, and IL12p40) and M2 markers (TGFβ1, IL10, and IL4) in THP-1 and BMDM-derived M2 macrophages were determined by qRT-PCR.
(c) THP-1에서 유래한 M2 대식세포에서 TNF-α, IL-1β, IL-4 및 IL-13을 이용하여 ELISA를 실시한 결과를 나타낸 도이다.(c) This diagram shows the results of ELISA using TNF-α, IL-1β, IL-4, and IL-13 in M2 macrophages derived from THP-1.
(d) 및 (e) THP-1 및 BMDM에서 유래한 M2 대식세포 중 CD16+ 및 CD206+ 세포에 대한 유세포 분석을 실시한 결과를 나타낸 도이다.(d) and (e) are diagrams showing the results of flow cytometry analysis on CD16 + and CD206 + cells among M2 macrophages derived from THP-1 and BMDM.
(*P < 0.05, **P < 0.01, ***P < 0.001, two-tailed Student's t-test)(*P < 0.05, **P < 0.01, ***P < 0.001, two-tailed Student's t-test)
이하 본 발명을 실시예를 통하여 보다 상세하게 설명한다. 그러나 이들 실시예는 본 발명을 예시적으로 설명하기 위한 것으로 본 발명의 범위가 이들 실시예에 한정되는 것은 아니다.The present invention will be described in more detail below through examples. However, these examples are intended to exemplify the present invention and the scope of the present invention is not limited to these examples.
실험예 1. 시약(Reagents)Experimental Example 1. Reagents
마우스 rWISP-1(1680-WS) 및 인간 rWISP-1(1627-WS)는 R&D Systems(Minneapolis, MN, USA)에서 구입하였다. 중화 마우스 WISP-1 항체(MAB1680) 및 IgG(MAB0061)는 R&D Systems(Minneapolis, MN, USA)에서 구입하였다.Mouse rWISP-1 (1680-WS) and human rWISP-1 (1627-WS) were purchased from R&D Systems (Minneapolis, MN, USA). Neutralizing mouse WISP-1 antibody (MAB1680) and IgG (MAB0061) were purchased from R&D Systems (Minneapolis, MN, USA).
실험예 2. CAF 분리 및 세포 배양Experimental Example 2. CAF Isolation and Cell Culture
섬유아세포 특이적 마커인 Thy1을 사용한 자기 활성화 세포 분류(MACS)를 통해 Kras-돌연변이체(KrasLA1) 마우스의 폐 종양에서 CAF를 분리하였다. 분리한 CAF는 10% FBS, 페니실린/스트렙토마이신(100U/100μg), 2mM L-글루타민 및 1mM 피루브산 나트륨을 보충한 α-MEM 배지에서 배양하였다. 불멸화를 위해, CAF는 Lipofector-EXT(AptaBio)를 사용하여 TERT 플라스미드(pCDH-3xFLAG-TERT; Addgene 51 plasmid #51631)로 안정적으로 형질감염하였다. 실험에 사용된 1차 세포(primary cell)는 6회 미만으로 계대 배양하였으며, 인간 세포주는 ATCC(American Type Culture Collection)로부터 수득하였다. 344SQ 세포 및 다양한 인간 암세포주는 10% FBS 및 페니실린/스트렙토마이신(100U/100μg)을 보충한 RPMI 1640 배지(HyCloneTM)에서 배양하여 유지시켰다.CAFs were isolated from lung tumors of Kras-mutant (Kras LA1 ) mice by magnetic-activated cell sorting (MACS) using the fibroblast-specific marker Thy1. Isolated CAFs were cultured in α-MEM medium supplemented with 10% fetal bovine serum, penicillin/streptomycin (100 U/100 μg), 2 mM L-glutamine, and 1 mM sodium pyruvate. For immortalization, CAFs were stably transfected with the TERT plasmid (pCDH-3xFLAG-TERT; Addgene 51 plasmid #51631) using Lipofector-EXT (AptaBio). Primary cells used in the experiments were passaged less than six times, and human cell lines were obtained from the American Type Culture Collection (ATCC). 344SQ cells and various human cancer cell lines were maintained in RPMI 1640 medium (HyClone ™ ) supplemented with 10% FBS and penicillin/streptomycin (100 U/100 μg).
실험예 3. 세포 사멸 유도Experimental Example 3. Induction of Cell Death
암 상피 세포주는 15분 동안 254nmm의 자외선으로 조사(irradiation)한 후 37℃, 5% CO2 조건 하에 2시간 동안 배양하였다. Wright-Giemsa로 염색된 샘플에 대하여 광학 현미경을 이용한 핵 형태(morphology)를 평가한 결과, 조사된(irradiated) 세포들은 거의 사멸화된(apoptotic) 것으로 나타났다. 또한, 여러 번의 동결-해동 사이클을 통해 용해된(괴사성) 암세포를 얻었다. 사멸화(apoptosis) 및 괴사(necrosis)는 annexin V-FITC/propidium iodide(BD Biosciences)로 염색한 후 FACSCalibur 시스템(BD Biosciences)에서 유세포 분석(flow cytometric analysis)으로 확인하였다.Cancer epithelial cell lines were irradiated with 254 nm of UV light for 15 min and then cultured for 2 h at 37°C under 5% CO2 conditions. Light microscopy of Wright-Giemsa-stained samples revealed that most irradiated cells were apoptotic. Furthermore, lysed (necrotic) cancer cells were obtained through multiple freeze-thaw cycles. Apoptosis and necrosis were confirmed by flow cytometric analysis using a FACSCalibur system (BD Biosciences) after staining with annexin V-FITC/propidium iodide (BD Biosciences).
실험예 4. CAF 배양 및 CAF CM(Conditioned-Medium) 제조Experimental Example 4. CAF Culture and CAF Conditioned Medium (CM) Preparation
CAF를 3 X 105 cells/ml로 플레이팅하고 37℃, 5% CO2 조건 하에 밤새 인큐베이션한 후, X-VIVO 10 배지(04-380Q)로 24시간 동안 혈청 제거(serum-starvation)시키고 세포를 자극하였다. 이를 위해 배양 배지를 사멸화 또는 괴사된 암세포(9 X 105 cells/ml)를 포함하는 X-VIVO 10 배지로 교체하였다. 20시간 경과 후, 원심분리로 상층액을 수확하고 표적 암 상피 세포(5 X 103 cells/ml)의 자극을 위한 CM로 사용하였다. 상기 CM은 in vivo 실험을 위하여 -80℃에서 보관하였다.CAFs were plated at 3 × 10 5 cells/ml and incubated overnight at 37°C under 5% CO 2 conditions. After serum-starvation for 24 h, the cells were stimulated with X-VIVO 10 medium (04-380Q). For this, the culture medium was replaced with X-VIVO 10 medium containing killed or necrotic cancer cells (9 × 10 5 cells/ml). After 20 h, the supernatant was harvested by centrifugation and used as CM for stimulating target cancer epithelial cells (5 × 10 3 cells/ml). The CM was stored at -80°C for in vivo experiments.
실험예 5. CM에서의 WISP-1 중화Experimental Example 5. Neutralization of WISP-1 in CM
CAF CM을 10μg/ml의 마우스의 항-마우스 WISP-1 중화 항체(R&D Systems) 또는 10μg/ml IgG 이소타입 대조군(R&D Systems)으로 2시간 동안 인큐베이션하였다. 항-WISP-1 항체 중화 효과는 사용 전에 WISP-1 ELISA를 통해 검사하였다.CAF CM were incubated with 10 μg/ml mouse anti-WISP-1 neutralizing antibody (R&D Systems) or 10 μg/ml IgG isotype control (R&D Systems) for 2 h. The anti-WISP-1 antibody neutralizing effect was tested by WISP-1 ELISA before use.
실험예 6. 마우스 실험Experimental Example 6. Mouse Experiment
이화여자대학교 의과학연구소 동물보호위원회로부터 실험계획서(EWHA MEDIACUC 22-015-3)를 승인받아 실험을 진행하였으며, 마우스는 미국국립보건원(NIH)의 가이드(Guide for the Care and Use of Laboratory Animals)에 따라 관리 및 취급되었다.The experiment was conducted with approval of the experimental protocol (EWHA MEDIACUC 22-015-3) from the Animal Care Committee of the Ewha Womans University Medical Research Institute, and the mice were managed and handled in accordance with the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health (NIH).
폐 선암(lung adenocarcinoma) 세포주인 344SQ 세포(마우스당 PBS 100μl에 1 X 106개의 세포)를 동계(129/Sv) 마우스의 우측 후방 옆구리(posterior flank)에 피하 주사하였다. 2일 후부터 CAF에서 유래되는 CM(마우스당 100μl)을 주 3회 종양 내 주입하였다. 또한, 중화 마우스의 항-WISP-1 Ab(10μg/ml) 또는 이소타입 IgG가 있거나 없는 CM을 동일한 일정으로 투여하였다. 마우스의 종양 성장은 매일 모니터링하였으며, 마우스는 주입 후 6주가 경과하였을 때 희생시켰다. 이후, 피하 종양 덩어리의 직경 및 무게를 조사하기 위해 부검을 수행하였다.344SQ cells, a lung adenocarcinoma cell line (1 × 106 cells in 100 μl of PBS per mouse), were injected subcutaneously into the right posterior flank of syngeneic (129/Sv) mice. 2 days later, CM derived from CAFs (100 μl per mouse) was injected intratumorally three times a week. In addition, CM with or without neutralizing mouse anti-WISP-1 Ab (10 μg/ml) or isotype IgG was administered on the same schedule. Tumor growth in the mice was monitored daily, and the mice were sacrificed 6 weeks after injection. Autopsies were then performed to examine the diameter and weight of the subcutaneous tumor masses.
실험예 7. 원발성 종양에서 CD11bExperimental Example 7. CD11b in Primary Tumors ++ TAM의 분리Separation of TAM
부검된 종양은 4U/mL DNase I과 함께 1X collagenase/hyaluronidase를 포함하는 RPMI 1640 배지에서 분해되었다. 세포 현탁액을 70μm 및 40μm 멸균 나일론 메쉬(mesh)로 여과하고 적혈구 용해 완충액과 함께 인큐베이션하였다. 펄스-원심분리(pulse-centrifugation) 후, 대식세포의 분리를 위해 상층액을 수집하고 CD11b+ TAM을 CD11b MicroBeads(Miltenyi Biotec)로 분리하였다. 분리된 TAM을 20% FBS, 2mM L-글루타민, 2mM 피루브산 나트륨, 55μM 2-머캅토에탄올 및 1% 페니실린-스트렙토마이신이 포함된 DMEM 배지에서 배양하였다. 개별 세포는 무작위로 선택된 마우스 원발성 종양으로부터 분리하였으며, 분리된 개별 세포 그룹은 qRT-PCT을 통해 확인하였다.Autopsied tumors were lysed in RPMI 1640 medium containing 1X collagenase/hyaluronidase with 4 U/mL DNase I. The cell suspension was filtered through 70 μm and 40 μm sterile nylon mesh and incubated with red blood cell lysis buffer. After pulse centrifugation, the supernatant was collected for macrophage isolation, and CD11b + TAMs were isolated with CD11b MicroBeads (Miltenyi Biotec). Isolated TAMs were cultured in DMEM medium containing 20% FBS, 2 mM L-glutamine, 2 mM sodium pyruvate, 55 μM 2-mercaptoethanol, and 1% penicillin-streptomycin. Individual cells were isolated from randomly selected mouse primary tumors, and isolated individual cell groups were identified by qRT-PCR.
실험예 8. 면역형광염색(Immunofluorescence staining)Experimental Example 8. Immunofluorescence staining
컨플루언트(confluent)에 도달할 때까지 유리 커버슬립에서 성장한 대식세포(106/웰)를 실온에서 8분 동안 4% 파라포름알데히드에 고정하였다. 파라핀이 내장된 종양 샘플을 염색하기 위하여 실온에서 30분간 포르말린 고정을 실시하였으며, 상기 샘플을 IF-Wash 완충액(0.05% NaN3, 0.1% BSA, 0.2% Triton X-100 및 0.05% Tween-20를 포함하는 PBS)으로 5분씩 3회 세척하고 실온에서 5분 동안 PBS 내 0.5% Triton X-100(Sigma-Aldrich)으로 투과시켰다. 면역조직화학(immunohistochemistry) 및 면역세포화학(immunocytochemistry)을 위하여 IgG 차단 시약이 포함된 또는 포함되지 않은 PBS의 BSA(5%)을 사용하였다. 1시간 경과 후, 4℃에서 18시간 인큐베이션하는 동안 표적 단백질을 1차 항체로 포획하였으며, 포획된 단백질은 암실에서 1시간 동안 형광-접합된(fluorescence-conjugated) IgG로 시각화하였다. 염색 후 슬라이드에 DAPI(Vector Laboratories)가 포함된 VECTASHIELD mounting medium을 장착하고 공초점 현미경(LSM5 PASCAL)으로 이미지화하였다. 사용된 항체에 대한 정보는 하기 표 1에 나타내었다.Macrophages (10 6 /well) grown on glass coverslips until confluent were fixed in 4% paraformaldehyde for 8 min at room temperature. For staining paraffin-embedded tumor samples, formalin fixation was performed for 30 min at room temperature, the samples were washed three times for 5 min each with IF-Wash buffer (PBS containing 0.05% NaN 3 , 0.1% BSA, 0.2% Triton X-100, and 0.05% Tween-20) and permeabilized with 0.5% Triton X-100 in PBS (Sigma-Aldrich) for 5 min at room temperature. For immunohistochemistry and immunocytochemistry, BSA (5%) in PBS with or without IgG blocking reagent was used. After 1 hour, the target protein was captured with the primary antibody during an 18-hour incubation at 4°C, and the captured protein was visualized with fluorescence-conjugated IgG in a darkroom for 1 hour. After staining, the slides were mounted with VECTASHIELD mounting medium containing DAPI (Vector Laboratories) and imaged using a confocal microscope (LSM5 PASCAL). Information on the antibodies used is shown in Table 1 below.
(Alexa 488)Goat IgG
(Alexa 488)
scientificThermo fisher
scientific
(Alexa 568)Mouse IgG
(Alexa 568)
scientificThermo fisher
scientific
(HRP)Mouse IgG
(HRP)
(Alexa 488)Rabbit IgG
(Alexa 488)
scientificThermo fisher
scientific
(HRP)Rabbit IgG
(HRP)
실험예 9. 면역블로팅 분석(Immunoblotting analysis)Experimental Example 9. Immunoblotting Analysis
전체 세포 추출물을 사용하여 표준 웨스턴 블랏을 수행하였으며, 사용한 항체의 정보는 상기 표 1에 나타내었다. 전체 세포 추출물은 CD11b+ TAM, THP-1 또는 BMDM-유도된 M1 및 M2 대식세포로부터 제조되었다. 세포를 수확하고 차가운 PBS(phosphate-buffered saline)로 세척한 후, 프로테아제 억제제가 첨가된 방사면역침전(radioimmunoprecipitation) 분석 완충액(10 mM Tris(pH 7.2), 150 mM NaCl, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 1.0% Triton X-100 및 5 mM EDTA)로 30분간 얼음 위에서 용해시켰다. 이후, 동일한 양의 단백질을 SDS-PAGE 젤(#161-0158, Bio-Rad Laboratories)에 용해시킨 후, wet 이송 시스템(Bio-Rad Laboratories)을 사용하여 니트로셀룰로스 막(10600001, GE Healthcare Life Science)으로 전송하였다. 막을 5% 소 혈청 알부민(BSA)-TBST 또는 5% 우유-TBST로 1시간 동안 차단한 후, 표지된 1차 항체와 밤새 배양한 다음 표지된 2차 항체와 함께 37℃에서 1시간 동안 배양하였다. 정량화를 위해 Odyssey 이미지 분석 시스템(Licor Biosciences)을 사용하였다.Standard Western blotting was performed using whole cell extracts, and the information of the antibodies used is shown in Table 1. Whole cell extracts were prepared from CD11b + TAM, THP-1, or BMDM-derived M1 and M2 macrophages. Cells were harvested, washed with cold phosphate-buffered saline (PBS), and lysed in radioimmunoprecipitation assay buffer (10 mM Tris (pH 7.2), 150 mM NaCl, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 1.0% Triton X-100, and 5 mM EDTA) supplemented with protease inhibitors for 30 min on ice. Equal amounts of protein were then dissolved in SDS-PAGE gels (#161-0158, Bio-Rad Laboratories) and transferred to nitrocellulose membranes (10600001, GE Healthcare Life Science) using a wet transfer system (Bio-Rad Laboratories). The membranes were blocked with 5% bovine serum albumin (BSA)-TBST or 5% milk-TBST for 1 h, incubated with labeled primary antibodies overnight, and then incubated with labeled secondary antibodies for 1 h at 37°C. The Odyssey image analysis system (Licor Biosciences) was used for quantification.
실험예 10. 정량적 실시간 중합효소 연쇄 반응(qRT-PCR)Experimental Example 10. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR)
총 RNA는 암세포로부터 TRIzol 시약(RNAiso plus reagent, Takara Bio Inc.)를 이용해 추출하였고, cDNA는 AccuPower RT PreMix(Bioneer)를 이용해 제조자의 프로토콜에 따라 합성하였다. SYBR Green-기반 정량적 실시간 중합효소 연쇄반응(qRT-PCR)은 QuantStudioTM 3 Real-Time PCR System(Applied Biosystems, Foster City, CA, USA)을 사용하여 수행하였다. mRNA 수준은 hypoxanthine guanine phospho-ribosyl transferase HPRT mRNA으로 정규화하였으며, 대조군에 대한 발현의 배수 변화로 나타내었다. 표적 유전자의 증폭에 사용한 프라이머 서열은 하기 표 2에 나타내었다.Total RNA was extracted from cancer cells using TRIzol reagent (RNAiso plus reagent, Takara Bio Inc.), and cDNA was synthesized using AccuPower RT PreMix (Bioneer) according to the manufacturer's protocol. SYBR Green-based quantitative real-time polymerase chain reaction (qRT-PCR) was performed using the QuantStudio ™ 3 Real-Time PCR System (Applied Biosystems, Foster City, CA, USA). mRNA levels were normalized to hypoxanthine guanine phospho-ribosyl transferase HPRT mRNA and expressed as fold change in expression relative to the control group. The primer sequences used to amplify the target genes are shown in Table 2.
실험예 11.Experimental Example 11. THP-1 세포 및 BMDM에서의 분극Polarization in THP-1 cells and BMDM
THP-1 세포는 37℃, 5% CO2가 포함된 가습 조건 하에 10% FBS를 포함하는 RPMI 1640 배지에서 유지하였다. THP-1 세포 유래 M1 또는 M2 대식세포를 대식세포 모델로 생성하였다. 즉, THP-1 세포를 150ng/ml PMA로 6시간 동안 프라이밍하여 비편극(nonpolarized) 대식세포로 만들었다. M1 대식세포를 제조하기 위하여, 48시간 동안 20ng/ml의 IFNγ 및 100ng/ml의 LPS로 비편극 대식세포를 자극하였다. M2 대식세포를 제조하기 위하여, 추가로 48시간 동안 20ng/ml IL-4 및 20ng/ml IL-13으로 비편극 대식세포를 자극하였다. 이후, 면역블로팅 분석을 위해 세포를 수확하거나 표시된 마커의 면역형광염색을 위해 고정하였다. 또한, C57BL/6 마우스의 경골(tibia) 및 대퇴골(femur)에서 분리된 BMDM을 L929 complement DMEM과 함께 7일 동안 배양한 다음 M1 및 M2 유형 대식세포로 편광시켰다.THP-1 cells were maintained in RPMI 1640 medium containing 10% FBS at 37°C under humidified conditions containing 5% CO2 . THP-1 cell-derived M1 or M2 macrophages were generated as a macrophage model. Specifically, THP-1 cells were primed with 150 ng/ml PMA for 6 h to produce nonpolarized macrophages. To generate M1 macrophages, nonpolarized macrophages were stimulated with 20 ng/ml IFNγ and 100 ng/ml LPS for 48 h. To generate M2 macrophages, nonpolarized macrophages were further stimulated with 20 ng/ml IL-4 and 20 ng/ml IL-13 for 48 h. Cells were then harvested for immunoblotting analysis or fixed for immunofluorescence staining of the indicated markers. Additionally, BMDMs isolated from the tibia and femur of C57BL/6 mice were cultured with L929 complement DMEM for 7 days and then polarized into M1 and M2 type macrophages.
실험예 12.Experimental Example 12. 세포 생존 분석Cell survival assay
대식세포(3.5 X 104)를 RPMI-1640 배지가 있는 96-웰 플레이트에 플레이팅하고 X-VIVO 10 배지에서 6시간 동안 배양하였다. CM 또는 rWISP-1을 각각의 그룹에 첨가하고, 2-4일 동안 5% CO2, 37℃ 조건 하에 인큐베이션하였다. 이후, cell counting kit-8(CCK-8) 용액을 웰에 첨가하고 30분 동안 인큐베이션하였다. 흡광도는 마이크로 플레이트 리더를 사용하여 450nm에서 측정하였다.Macrophages (3.5 × 10 4 ) were plated in 96-well plates containing RPMI-1640 medium and cultured in X-VIVO 10 medium for 6 hours. CM or rWISP-1 was added to each group and incubated for 2–4 days under 5% CO 2 , 37°C conditions. Afterwards, cell counting kit-8 (CCK-8) solution was added to the wells and incubated for 30 minutes. The absorbance was measured at 450 nm using a microplate reader.
실험예 13. 일시적 형질감염(Transient transfection)Experimental Example 13. Transient transfection
형질감염 시약(Lipofectamin RNAi MAX; Invitrogen, Carlsbad, CA)을 사용하여 CAF 또는 대식세포를 최종 농도 50nM에서 WISP1(Bioneer), STAT1(Bioneer) 또는 대조군 siRNA(SN-1003 AccuTargetTM Negative Control)를 특이적 표적으로 하는 siRNA로 일시적 형질감염시켰다. 밤새 형질감염시킨 후, 세포를 24시간 동안 적절한 배지에서 배양하고 ApoSQ 세포로 자극하였다. 사용된 siRNA 서열은 다음과 같다(gene: sense, antisense):CAFs or macrophages were transiently transfected with siRNAs specifically targeting WISP1 (Bioneer), STAT1 (Bioneer), or a control siRNA (SN-1003 AccuTarget ™ Negative Control) at a final concentration of 50 nM using a transfection reagent (Lipofectamin RNAi MAX; Invitrogen, Carlsbad, CA). After overnight transfection, cells were cultured in appropriate media for 24 h and stimulated with ApoSQ cells. The siRNA sequences used were as follows (gene: sense, antisense):
- Stat1: 5'-CUGAAUCAAGACUGA-3'(서열번호 47), 5'-UCAGUUGAUCCAG-3'(서열번호 48)- Stat1: 5'-CUGAAUCAAGACUGA-3' (SEQ ID NO: 47), 5'-UCAGUUGAUCCAG-3' (SEQ ID NO: 48)
- WISP-1: 5'-GGAAUCCUACAGAUACU-3'(서열번호 49), 5'-AAAGAUCCUGAUCCU-3'(서열번호 50), 5'-AAAUCCUGAUCCU-3'(서열번호 51)- WISP-1: 5'-GGAAUCCUACAGAUACU-3' (SEQ ID NO: 49), 5'-AAAGAUCCUGAUCCU-3' (SEQ ID NO: 50), 5'-AAAUCCUGAUCCU-3' (SEQ ID NO: 51)
실험예 14. 세포 사멸 분석Experimental Example 14. Cell Death Analysis
세포 사멸은 annexin V-FITC/propidium iodide(PI) 염색 키트(BD Biosciences, San Jose, CA, USA)를 사용하여 제조업체의 지침에 따라 검출하였다. 대식세포는 수확 후 500μl의 결합 완충액에 재현탁하였으며, 이중 100μl의 현탁액은 암실에서 15분간 5μl FITC-conjugated annexin V 및 5μl PI로 실온에서 염색하였다. 이후, FITC-conjugated annexin V에 양성인 세포를 400μl의 결합 완충액을 사용하여 유세포 측정기(ACEA NovoCyte, San Diego, CA, USA)로 검출하였고, 데이터는 NovoExpress 소프트웨어 1.5를 사용하여 분석하였다. Apoptosis was detected using an annexin V-FITC/propidium iodide (PI) staining kit (BD Biosciences, San Jose, CA, USA) according to the manufacturer's instructions. After harvesting, macrophages were resuspended in 500 μl of binding buffer, and 100 μl of the suspension was stained with 5 μl of FITC-conjugated annexin V and 5 μl of PI for 15 min at room temperature in the dark. FITC-conjugated annexin V-positive cells were then detected by flow cytometry (ACEA NovoCyte, San Diego, CA, USA) using 400 μl of binding buffer, and data were analyzed using NovoExpress software 1.5.
실험예 15. 유세포 분석(Flow cytometric analysis)Experimental Example 15. Flow cytometric analysis
FACS 기반 유세포 분석을 위하여 NovoCyte(Agilent, Santa Clara, CA, USA)를 사용하였다. 모든 세포 현탁액(1x106 CD11b+ TAM 및 M2 대식세포)을 500μl 완충액(PBS + 2% FBS)에 넣고 제조업체의 권장 농도를 사용하여 형광단-접합된(fluorophore-conjugated) 항-마우스 항체와 함께 60분 동안 배양했다. 이때 사용된 모든 항체는 하기 표 3에 나타내었다. 데이터 수집은 NovoCyte(Agilent, Santa Clara, CA, USA)에서 수행하였으며, NovoExpress Software 1.5를 분석에 사용하였다.For FACS-based flow cytometry analysis, NovoCyte (Agilent, Santa Clara, CA, USA) was used. All cell suspensions (1 × 10 6 CD11b + TAMs and M2 macrophages) were placed in 500 μl buffer (PBS + 2% FBS) and incubated for 60 min with fluorophore-conjugated anti-mouse antibodies at the manufacturer's recommended concentration. All antibodies used are listed in Table 3. Data acquisition was performed on NovoCyte (Agilent, Santa Clara, CA, USA), and NovoExpress Software 1.5 was used for analysis.
실험예 16. ELISAExperimental Example 16. ELISA
대식세포 배양액에서 TNF-α, IL-1β, IL-4 및 IL-13은 제조업체의 지침에 따라 ELISA 키트(R&D Systems)를 사용하여 측정하였다.TNF-α, IL-1β, IL-4, and IL-13 in macrophage cultures were measured using ELISA kits (R&D Systems) according to the manufacturer's instructions.
실험예 17. 통계(Statistics)Experimental Example 17. Statistics
짝(pairwise) 비교는 two-tailed Student's t-테스트를 사용하여 수행하였고, 다중(multiple) 비교는 Kruskal-Wallis 테스트에 이어 Dunn's post hoc 테스트를 사용하여 수행하였다. 0.05보다 작은 P 값은 통계적으로 유의한 것으로 간주하였고, 모든 데이터는 Prism 5 소프트웨어(GraphPad Software Inc., San Diego, CA, USA)를 사용하여 분석하였다.Pairwise comparisons were performed using the two-tailed Student's t-test, and multiple comparisons were performed using the Kruskal-Wallis test followed by Dunn's post hoc test. A P value less than 0.05 was considered statistically significant, and all data were analyzed using Prism 5 software (GraphPad Software Inc., San Diego, CA, USA).
실시예 1. ApoSQ-CM 투여에 따른 TAM의 밀도 및 분율 변화Example 1. Changes in the density and fraction of TAMs following ApoSQ-CM administration
면역능력이 있는(immunocompetent) 동계(129/Sν) 마우스에 344SQ 투여 2일 후부터 CAF CM(CAF의 CM) 또는 ApoSQ-CAF CM(사멸화된 344SQ 세포에 노출된 CAF의 CM)을 매주 3회 종양 내 투여하였다. 또한, 상기 CM을 투여하기 전에, WISP-1에 대한 중화 항체 또는 IgG 동형(isotype) 대조 항체로 CM을 2시간 동안 사전 배양하였다.Immunocompetent syngeneic (129/Sν) mice were administered intratumorally three times weekly with CAF CM (CM of CAFs) or ApoSQ-CAF CM (CM of CAFs exposed to killed 344SQ cells) starting 2 days after 344SQ administration. In addition, prior to administration of the CM, the CMs were pre-incubated for 2 h with a neutralizing antibody against WISP-1 or an IgG isotype control antibody.
(1) 총 TAM 밀도 변화(1) Change in total TAM density
총 TAM 마커(CD11b 및 F4/80)를 사용하여 면역형광염색을 실시한 결과, ApoSQ-CAF CM을 투여하면 CAF CM을 투여했을 때와 비교하여 원발성 종양의 중앙 및 가장자리 영역 모두에서 총 TAM 밀도가 현저히 감소하는 것으로 나타났다(도 1). 한편, WISP-1 면역 결핍된(immunodepleted) ApoSQ-CAF CM을 투여한 경우에 총 TAM 밀도는 변하지 않았으나, IgG 동형 대조 항체와 함께 사전 배양된 CM을 투여한 경우에는 ApoSQ-CAF CM을 투여했을 때와 유사한 효과가 나타나는 것으로 확인되었다.Immunofluorescence staining using total TAM markers (CD11b and F4/80) revealed that administration of ApoSQ-CAF CM significantly reduced total TAM density in both the central and marginal regions of primary tumors compared to administration of CAF CM (Fig. 1). On the other hand, administration of WISP-1 immunodepleted ApoSQ-CAF CM did not change total TAM density, but administration of CM pre-incubated with IgG isotype control antibody showed an effect similar to that of administration of ApoSQ-CAF CM.
즉, ApoSQ-CAF CM의 투여에 의하여 원발성 종양의 총 TAM 밀도 감소가 WISP-1 작용을 통해 이루어짐을 의미한다. That is, the reduction in total TAM density in primary tumors by administration of ApoSQ-CAF CM is achieved through the action of WISP-1.
(2) TAM 분율(fraction) 변화(2) Change in TAM fraction
ApoSQ-CAF CM을 투여함으로써 원발성 종양에서 TAM의 하위 유형 분포가 어떻게 변화하는지 확인하기 위하여, M2 마커(Arg1 및 CD206) 및 M1 마커(iNOS 및 CD16/32)를 사용하여 면역형광염색을 실시하였다. 그 결과, ApoSQ-CAF CM의 투여 후 M2 TAM 분율은 현저히 감소하였으나(도 2), M1 TAM 분율은 증가함을 확인하였다(도 3). 한편, WISP-1 면역 결핍된(immunodepleted) ApoSQ-CAF CM을 투여한 경우에는 이러한 M1 및 M2 TAM 분율의 변화 양상이 역전되어 나타난 반면에, IgG 동형 대조 항체와 함께 사전 배양된 CM을 투여한 경우에는 ApoSQ-CAF CM을 투여했을 때와 동일한 변화 양상을 나타냈다.To determine how the subtype distribution of TAMs in primary tumors changes with the administration of ApoSQ-CAF CM, immunofluorescence staining was performed using M2 markers (Arg1 and CD206) and M1 markers (iNOS and CD16/32). As a result, it was confirmed that the M2 TAM fraction significantly decreased after the administration of ApoSQ-CAF CM (Fig. 2), whereas the M1 TAM fraction increased (Fig. 3). On the other hand, when WISP-1 immunodepleted ApoSQ-CAF CM was administered, the changes in the M1 and M2 TAM fractions were reversed, whereas when CM pre-incubated with an IgG isotype control antibody was administered, the same changes as when ApoSQ-CAF CM was administered.
또한, ApoSQ-CAF CM에 의하여 M2 TAM의 세포 사멸(Cleaved Caspase-3+(C-Cas3+)/CD11b+)은 증가하는 반면에, M1 TAM의 세포 사멸(C-Cas3+/CD16/32+)에는 거의 영향을 미치지 않는 것으로 확인되었다(도 4a 및 4b). 아울러, 원발성 종양에서 총 TAM의 세포사멸(Cleaved Caspase-3+(C-Cas3+)/CD11b+)이 ApoSQ-CAF CM의 투여에 의하여 유의미하게 증가하였다(도 4c). 한편, WISP-1 면역 결핍된(immunodepleted) ApoSQ-CAF CM을 투여한 경우에는 M2 TAM의 세포사멸 및 총 TAM의 세포사멸 중가 효과는 나타나지 않았다. 반면에, IgG 동형 대조 항체와 함께 사전 배양된 CM을 투여한 경우에는 ApoSQ-CAF CM을 투여했을 때와 동일한 세포사멸 증가 양상을 나타냈다. 즉, 상기 실험 결과들에 따르면 ApoSQ-CAF CM의 종양 성장 억제 효과는 WISP-1에 의해 매개된다.In addition, it was confirmed that ApoSQ-CAF CM increased the apoptosis of M2 TAM (Cleaved Caspase-3 + (C-Cas3 + )/CD11b + ), whereas it had little effect on the apoptosis of M1 TAM (C-Cas3 + /CD16/32 + ) (Figures 4a and 4b). In addition, the apoptosis of total TAM (Cleaved Caspase-3 + (C-Cas3 + )/CD11b + ) in primary tumors was significantly increased by administration of ApoSQ-CAF CM (Figure 4c). On the other hand, when WISP-1 immunodepleted ApoSQ-CAF CM was administered, the effect of increasing the apoptosis of M2 TAM and total TAM was not observed. In contrast, administration of CM pre-incubated with IgG isotype control antibody resulted in the same pattern of increased apoptosis as when ApoSQ-CAF CM was administered. In other words, according to the above experimental results, the tumor growth inhibitory effect of ApoSQ-CAF CM is mediated by WISP-1.
실시예 2. ApoSQ-CM 투여에 따른 M2 TAM으로부터 M1 TAM으로의 리프로그래밍 유도Example 2. Induction of reprogramming from M2 TAM to M1 TAM by ApoSQ-CM administration.
(1) M2 및 M1 마커의 발현 수준 변화(1) Changes in expression levels of M2 and M1 markers
원발성 종양으로부터 분리된 CD11b+ TAM에서 M2 및 M1 마커의 mRNA 수준을 RT-qPCR array 각각 분석하였다. 그 결과, ApoSQ-CAF CM을 투여하였을 때 M2 관련 유전자 9개가 CAF CM 그룹에 비해 2배 이상 하향 조절되었다(도 5a). 반대로, Cd32, Ifng, Cd16, Tnf, Nos2, Socs3, Cd80을 포함한 M1 관련 유전자 7개는 CAF CM 그룹에 비해 ApoSQ-CAF CM 그룹에서 2배 이상 상향 조절되었다. 이러한 mRNA 발현 변화를 확인하기위해, M2 및 M1 특이적 마커와 사이토카인에 대한 qRT-PCR 분석도 수행되었다. 그 결과, ApoSQ-CAF CM을 투여하였을 때 M2 마커(Arg1, CD206, CD163, IL-4, IL-10, TGFβ1)의 mRNA 수준은 유의하게 감소한 반면, M1 마커(TNFα, CD80, H2Ab1, NOS2, IFNγ 및 IL12p40)의 mRNA 수준은 증가하였다(도 5b). 또한, 면역블로팅을 실시한 결과, mRNA 수준의 변화와 마찬가지로 ApoSQ-CAF CM의 투여에 따라 M2 마커(Arg1 및 CD206)의 단백질 수준은 감소하였으나, M1 마커(iNOS 및 CD16/32)는 단백질 수준은 증가하였다(도 5c). 한편, WISP-1 면역 결핍된(immunodepleted) ApoSQ-CAF CM을 투여한 경우에 이러한 발현 수준의 변화는 역전되어 나타났으며, IgG 동형 대조 항체와 함께 사전 배양된 CM을 투여한 경우에는 ApoSQ-CAF CM을 투여했을 때와 동일한 모습을 보였다.The mRNA levels of M2 and M1 markers in CD11b + TAMs isolated from primary tumors were analyzed using RT-qPCR arrays, respectively. As a result, nine M2-related genes were downregulated more than twofold in the ApoSQ-CAF CM group compared to the CAF CM group (Fig. 5a). Conversely, seven M1-related genes, including Cd32, Ifng, Cd16, Tnf, Nos2, Socs3, and Cd80, were upregulated more than twofold in the ApoSQ-CAF CM group compared to the CAF CM group. To confirm these mRNA expression changes, qRT-PCR analysis of M2- and M1-specific markers and cytokines was also performed. As a result, when ApoSQ-CAF CM was administered, the mRNA levels of M2 markers (Arg1, CD206, CD163, IL-4, IL-10, and TGFβ1) significantly decreased, whereas the mRNA levels of M1 markers (TNFα, CD80, H2Ab1, NOS2, IFNγ, and IL12p40) increased (Fig. 5b). In addition, immunoblotting results showed that, similar to the change in mRNA levels, the protein levels of M2 markers (Arg1 and CD206) decreased with ApoSQ-CAF CM administration, whereas the protein levels of M1 markers (iNOS and CD16/32) increased (Fig. 5c). Meanwhile, when WISP-1 immunodepleted ApoSQ-CAF CM was administered, these changes in expression levels were reversed, and when CM pre-incubated with IgG isotype control antibody was administered, the same appearance as when ApoSQ-CAF CM was administered was observed.
또한, 원발성 종양에서 분리된 CD11b+ TAM에서 M2 및 M1 마커를 이용하여 유세포 분석을 실시하였다. 그 결과, ApoSQ-CAF CM의 투여에 의하여 M2 TAM(CD11b+ TAM에서 CD163+ 또는 CD206+)의 비율을 유의하게 감소시킨 반면에, M1 유사 표현형을 발현하는 TAM(CD11b+ TAM에서 CD16+ 또는 CD80+)의 비율을 현저히 증가시켰다(도 5d 및 5e).Additionally, flow cytometric analysis was performed using M2 and M1 markers in CD11b + TAMs isolated from primary tumors. As a result, administration of ApoSQ-CAF CM significantly reduced the proportion of M2 TAMs (CD163 + or CD206 + in CD11b + TAMs), whereas it markedly increased the proportion of TAMs expressing an M1-like phenotype (CD16 + or CD80 + in CD11b + TAMs) (Figures 5d and 5e).
아울러, 원발성 종양에서 분리된 CD11b+ TAM에 대해 M2 및 M1 마커를 이용한 유세포 분석을 수행한 결과, ApoSQ-CAF CM의 투여에 따라 CD163+/MHCII+ TAM 비율이 유의하게 감소하였다(도 5f). 또한, 림프단핵세포에 대해 CD45, CD11b, CD3, CD4, CD8, FoxP3, MHCII, Ly6C 항체로 염색하여 유세포 분석을 수행한 결과, (g) M2 대식세포, (h) 조절 T 세포(Tregs)의 수가 유의하게 감소한 반면, (i) M1 대식세포, (j) CD8+ T 세포의 수는 유의하게 증가하는 경향을 보였다(도 5g 내지 5j).In addition, flow cytometric analysis using M2 and M1 markers on CD11b + TAMs isolated from primary tumors showed that the ratio of CD163 + /MHCII + TAMs significantly decreased with the administration of ApoSQ-CAF CM (Fig. 5f). In addition, flow cytometric analysis of lymphomonocytes stained with CD45, CD11b, CD3, CD4, CD8, FoxP3, MHCII, and Ly6C antibodies showed that the numbers of (g) M2 macrophages and (h) regulatory T cells (Tregs) significantly decreased, whereas the numbers of (i) M1 macrophages and (j) CD8 + T cells tended to significantly increase (Figs. 5g to 5j).
따라서, 종양 성장을 억제하는 물질인 ApoSQ-CAF CM의 투여에 의하여 M2 마커의 발현은 감소하는 반면, M1 마커의 발현은 증가함을 확인하였다.Therefore, it was confirmed that the expression of M2 markers decreased while the expression of M1 markers increased by administration of ApoSQ-CAF CM, a substance that inhibits tumor growth.
(2) M2 TAM으로부터 M1 TAM으로의 리프로그래밍에 있어 STAT1의 역할(2) Role of STAT1 in reprogramming from M2 TAM to M1 TAM
원발성 종양 조직에서 면역형광염색을 실시하여 분석한 결과, ApoSQ-CAF 투여 후 M2 TAM(pSTAT1+/CD206+)에서는 인산화 STAT1(phosphorylated STAT1)이 증강된 것으로 나타났으나(도 5k), M1 TAM(pSTAT1+/CD16/32+)에서는 이러한 효과가 관찰되지 않았다(도 5l).Analysis of primary tumor tissues by immunofluorescence staining revealed that phosphorylated STAT1 was increased in M2 TAM (pSTAT1 + /CD206 + ) after ApoSQ-CAF administration (Fig. 5k), but this effect was not observed in M1 TAM (pSTAT1 + /CD16/32 + ) (Fig. 5l).
즉, 상기와 같은 실험 결과들은 ApoSQ-CAF CM이 WISP-1-STAT1 신호 전달 경로를 통해 M2 TAM으로부터 M1 유사 표현형을 발현하는 TAM으로의 리프로그래밍을 유도하기 때문에 나타남을 확인하였다.That is, the above experimental results confirmed that ApoSQ-CAF CM induced reprogramming from M2 TAM to TAM expressing an M1-like phenotype through the WISP-1-STAT1 signaling pathway.
실시예 3. ApoSQ-CM 투여에 따른 M2 TAM의 생존 억제 및 M2 TAM으로부터 M1 TAM으로의 변화 유도(in vitro)Example 3. Inhibition of M2 TAM survival and induction of transformation from M2 TAM to M1 TAM by ApoSQ-CM administration (in vitro)
(1) M1 및 M2 대식세포의 생존 및 사멸 변화(1) Changes in survival and death of M1 and M2 macrophages
시험관 내(in vitro) 연구를 위해 THP-1 세포 및 1차 마우스 골수 유래 대식세포(BMDM)를 M1 및 M2 대식세포로 분극화(polarized)하여 TAM을 모방하였다(도 6a 및 6b). 이후 M2 마커(CD163, CD206 및 Arg1) 및 M1 마커(MHCII, iNOS 및 IL12p40)를 이용하여 면역블로팅을 실시한 결과, M1 및 M2 대식세포로 성공적으로 분극화 되었음을 확인하였다(도 6c 및 6d). 또한, 공초점 현미경 분석에서도 CD86+ M1 및 CD163+ M2 마커를 이용하여 분극화 되었음을 확인할 수 있었다(도 6e 및 6f).For in vitro studies, THP-1 cells and primary mouse bone marrow-derived macrophages (BMDMs) were polarized into M1 and M2 macrophages to mimic TAMs (Figs. 6a and 6b). Successful polarization into M1 and M2 macrophages was confirmed by immunoblotting using M2 markers (CD163, CD206, and Arg1) and M1 markers (MHCII, iNOS, and IL12p40) (Figs. 6c and 6d). Furthermore, confocal microscopy analysis also confirmed polarization using CD86 + M1 and CD163 + M2 markers (Figs. 6e and 6f).
THP-1 세포 및 BMDM에서 분극된 M1 및 M2 대식세포를 혈청 없는 상태(serum free starvation)에서 4일간 CM으로 처리한 후 CCK-8 assay를 통해 세포 생존 분석을 실시하였다. 사멸화된 344SQ 세포(ApoSQ) 및 괴사된 344SQ 세포(NecSQ)에 노출되었는지 여부에 관계없이 CAF의 CM은 M1 대식세포의 2일차 및 4일차 세포 생존율(cell viability)에 영향을 미치지 않았다. 그러나, ApoSQ-CAF CM의 처리에 의하여 M2 대식세포의 2일차 및 4일차 세포 생존율은 감소하는 것으로 나타났다(도 7a 및 7b).Polarized M1 and M2 macrophages from THP-1 cells and BMDMs were treated with CM for 4 days under serum-free starvation, and cell viability was analyzed using the CCK-8 assay. Regardless of whether CAF CM was exposed to apoptotic 344SQ cells (ApoSQ) or necrotic 344SQ cells (NecSQ), M1 macrophage viability on days 2 and 4 was not affected. However, treatment with ApoSQ-CAF CM decreased M2 macrophage viability on days 2 and 4 (Figs. 7a and 7b).
또한, annexin V-FITC 및 PI 염색 후 유세포 분석을 실시한 결과, ApoSQ 또는 NecSQ에 노출되었는지 여부에 관계없이 CAF의 CM은 M1 대식세포의 세포 사멸에 영향을 미치지 않았다. 반면에 ApoSQ-CAF CM의 처리에 의하여 M2 대식세포의 세포 사멸은 증가하는 것으로 확인되었다(도 7c 및 7d).Furthermore, flow cytometric analysis after annexin V-FITC and PI staining revealed that CAF CM did not affect the apoptosis of M1 macrophages, regardless of whether they were exposed to ApoSQ or NecSQ. In contrast, treatment with ApoSQ-CAF CM increased the apoptosis of M2 macrophages (Figs. 7c and 7d).
한편, THP-1 유래 M2 대식세포를 ApoSQ-CAF CM으로 처리한 경우에 Bax, C-Cas3, C-PARP를 포함한 친-사멸화(pro-apoptotic) 바이오마커의 발현 수준은 증가하였으나, 항-사멸화(anti-apoptotic) 바이오마커인 Mcl-1 및 Bcl-xL의 발현 수준은 감소했다(도 7e).Meanwhile, when THP-1-derived M2 macrophages were treated with ApoSQ-CAF CM, the expression levels of pro-apoptotic biomarkers, including Bax, C-Cas3, and C-PARP, increased, but the expression levels of anti-apoptotic biomarkers, Mcl-1 and Bcl-xL, decreased (Fig. 7e).
마찬가지로, ApoA(사멸화된 A549 cells)-CAF CM의 처리에 의하여 THP-1 유래 M2 대식세포의 생존율이 감소하고 세포 사멸은 증가하였으나, M1 대식세포의 생존율 및 사멸에는 아무런 변화도 나타나지 않았다(도 7f 및 7g).Similarly, treatment with ApoA (dead A549 cells)-CAF CM decreased the viability and increased apoptosis of THP-1-derived M2 macrophages, but did not change the viability or apoptosis of M1 macrophages (Figs. 7f and 7g).
(2) M2 TAM으로부터 M1 TAM으로의 리프로그래밍(2) Reprogramming from M2 TAM to M1 TAM
THP-1 또는 BMDM 유래 M2 대식세포에 ApoSQ-CAF CM을 처리하였을 때 M1 마커(NOS2, MHCII 및 IL12p40)의 mRNA 수준이 증가하였으며, M2 마커(TGFβ1, IL10 및 IL4)의 mRNA 수준이 감소하였다. 그러나, 상기 M2 대식세포에 CAF CM 및 NecSQ-CAF CM을 처리하였을 때에는 마커들의 발현 변화가 나타나지 않았다(도 8a 및 8b).When THP-1 or BMDM-derived M2 macrophages were treated with ApoSQ-CAF CM, the mRNA levels of M1 markers (NOS2, MHCII, and IL12p40) increased, while the mRNA levels of M2 markers (TGFβ1, IL10, and IL4) decreased. However, when the M2 macrophages were treated with CAF CM and NecSQ-CAF CM, no changes in the expression of the markers were observed (Figs. 8a and 8b).
이와 마찬가지로, THP-1 유래 M2 대식세포에 ApoSQ-CAF CM을 처리함으로써 TNFα 및 IL-1β를 포함하는 M1 사이토카인의 수준은 향상되었으나, IL-4 및 IL-13을 포함하는 M2 사이토카인의 수준은 억제되었다(도 8c). 또한, 유세포 분석 결과, ApoSQ-CAF CM을 THP-1 유래 M2 대식세포 또는 BMDM-유래 M2 대식세포에 처리하면 CAF CM 또는 NecSQ-CAF CM을 처리하였을 때와 비교하여 CD206 M2 마커의 표면 발현이 감소하고 CD16 M1 마커의 표면 발현이 증가하였다(도 8d 및 8e).Similarly, treatment of THP-1-derived M2 macrophages with ApoSQ-CAF CM enhanced the levels of M1 cytokines, including TNFα and IL-1β, whereas suppressed the levels of M2 cytokines, including IL-4 and IL-13 (Fig. 8c). In addition, flow cytometry analysis showed that treatment of THP-1-derived M2 macrophages or BMDM-derived M2 macrophages with ApoSQ-CAF CM decreased the surface expression of the CD206 M2 marker and increased the surface expression of the CD16 M1 marker compared to treatment with CAF CM or NecSQ-CAF CM (Figs. 8d and 8e).
따라서, 생체 내(in vivo)에서의 결과와 동일하게, 시험관 내(in vitro)에서도 ApoSQ-CAF CM에 의하여 M2 대식세포의 생존이 억제되고, M2 대식세포가 M1 유사 표현형을 발현하는 대식세포로 리프로그래밍됨을 확인하였다.Therefore, similar to the in vivo results, we confirmed that the survival of M2 macrophages was inhibited by ApoSQ-CAF CM in vitro and that M2 macrophages were reprogrammed into macrophages expressing an M1-like phenotype.
이상의 설명으로부터, 본 발명이 속하는 기술분야의 당업자는 본 발명 그 기술적 사상이나 필수적 특징을 변경하지 않고서 다른 구체적인 형태로 실시될 수 있다는 것을 이해할 수 있을 것이다. 이와 관련하여, 이상에서 기술한 실시예들은 모든 면에서 예시적인 것이며 한정적인 것이 아닌 것으로 이해해야만 한다. 본 발명의 범위는 상기 상세한 설명보다는 후술하는 특허 청구범위의 의미 및 범위 그리고 그 등가 개념으로부터 도출되는 모든 변경 또는 변형된 형태가 본 발명의 범위에 포함되는 것으로 해석되어야 한다.From the above description, those skilled in the art will understand that the present invention can be implemented in other specific forms without altering the technical spirit or essential characteristics of the present invention. In this regard, it should be understood that the embodiments described above are illustrative in all respects and not restrictive. The scope of the present invention should be interpreted as encompassing all changes or modifications derived from the meaning and scope of the following claims and their equivalent concepts, rather than the detailed description above.
Claims (19)
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| KR10-2024-0070188 | 2024-05-29 | ||
| KR20240070188 | 2024-05-29 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025249993A1 true WO2025249993A1 (en) | 2025-12-04 |
Family
ID=97871114
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/KR2025/095352 Pending WO2025249993A1 (en) | 2024-05-29 | 2025-05-22 | Composition for treating cancerous diseases and screening method thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025249993A1 (en) |
-
2025
- 2025-05-22 WO PCT/KR2025/095352 patent/WO2025249993A1/en active Pending
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| WO2017078440A1 (en) | Peptide having neuronal loss prevention and regeneration effects, and composition containing same | |
| WO2015156649A1 (en) | Peptide having fibrosis inhibitory activity and composition containing same | |
| WO2015076621A1 (en) | Peptide having angiogenesis inhibitory activity and composition containing same | |
| WO2017022962A1 (en) | Composition for preventing or treating immune disease, comprising ripk inhibitor as active ingredient | |
| WO2011052883A9 (en) | Method for activating a natural killer cell by adjusting the expression of the socs2 gene | |
| WO2023096126A1 (en) | Method for screening mdsc inhibitor | |
| WO2020091463A1 (en) | Pharmaceutical composition comprising isolated mitochondria for preventing or treating tendinopathy | |
| WO2025249993A1 (en) | Composition for treating cancerous diseases and screening method thereof | |
| WO2018026212A2 (en) | Method for producing fibrosis disease model, and use of fibrosis disease model | |
| WO2022025455A1 (en) | Composition for preventing or treating inflammatory diseases, and use thereof | |
| WO2019245269A1 (en) | Composition comprising flt3 inhibitor as effective ingredient for inhibiting drug resistance in chronic myelogenous leukemia | |
| WO2016204545A1 (en) | Peptide for promoting osteogenesis or inhibiting osteolysis, and use thereof | |
| WO2020017788A1 (en) | Composition for preventing or treating diabetes by using cd9, and method for screening for antidiabetic agents | |
| KR20250172407A (en) | A composition for treating cancer diseases and screening method thereof | |
| WO2023200257A1 (en) | Extracellular vesicles isolated from stem cells, and uses thereof | |
| WO2022131742A1 (en) | Exosomes derived from cells treated with material inducing endoplasmic reticulum stress, and use thereof | |
| WO2011087343A2 (en) | Composition for treating cancer related to an hpv infection | |
| WO2022211473A1 (en) | Fas-associated factor 1 (faf1)-loaded exosomes and use thereof as anti-cancer agent | |
| WO2017061828A1 (en) | Pharmaceutical composition for preventing or treating cancer comprising inhibitor of plrg1 (pleiotropic regulator 1) as active ingredient | |
| WO2021033973A1 (en) | Composition for inhibiting myeloid-derived suppressor cell comprising mitf inhibitor as active ingredient | |
| WO2023055076A1 (en) | Composition for inhibiting cancer metastasis | |
| WO2017164486A1 (en) | Composition for inhibiting resistance to anticancer agents, containing tesk1 inhibitor, and method for screening for tesk1 inhibitor | |
| WO2023182792A1 (en) | Composition for inhibiting cancer growth | |
| WO2025053611A1 (en) | Pharmaceutical composition comprising pdk3 inhibitor for preventing or treating peripheral nerve tumor, and use thereof | |
| WO2023096351A1 (en) | Composition for treating cancer having epidermal growth factor receptor (egfr) mutation |