[go: up one dir, main page]

WO2023055076A1 - Composition for inhibiting cancer metastasis - Google Patents

Composition for inhibiting cancer metastasis Download PDF

Info

Publication number
WO2023055076A1
WO2023055076A1 PCT/KR2022/014563 KR2022014563W WO2023055076A1 WO 2023055076 A1 WO2023055076 A1 WO 2023055076A1 KR 2022014563 W KR2022014563 W KR 2022014563W WO 2023055076 A1 WO2023055076 A1 WO 2023055076A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
cafs
cells
aposq
wisp
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.)
Ceased
Application number
PCT/KR2022/014563
Other languages
French (fr)
Korean (ko)
Inventor
이지희
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ewha Womans University
Original Assignee
Ewha Womans University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR1020220058184A external-priority patent/KR20230046190A/en
Application filed by Ewha Womans University filed Critical Ewha Womans University
Publication of WO2023055076A1 publication Critical patent/WO2023055076A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES, NOT OTHERWISE PROVIDED FOR; PREPARATION OR TREATMENT THEREOF
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/13Tumour cells, irrespective of tissue of origin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/33Fibroblasts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Definitions

  • the present invention provides a pharmaceutical composition for inhibiting cancer metastasis containing a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured, a method for inhibiting cancer metastasis using the same, and It relates to the use of the pharmaceutical composition.
  • CAFs cancer-associated fibroblasts
  • Lung cancer both small cell and non-small cell cancer
  • Lung cancer is the most common cancer worldwide in both men and women, and is the leading cause of cancer-related death (18.0% of total cancer deaths).
  • About 75% of lung cancer patients are confirmed to have locally advanced or metastatic disease at the time of diagnosis.
  • metastasis is a multistep process including migration and invasion of cancer cells, and becomes a marker of malignant tumors.
  • CAFs Cancer-associated fibroblasts
  • Basal processes such as cancer cell migration and invasion that can promote progression to malignancy and metastatic spread.
  • CAFs physically remodel the matrix in the tumor stroma, allowing cancer cells to invade while still maintaining epithelial properties.
  • the underlying molecular mechanisms of CAFs-mediated regulation in tumor progression are still unclear.
  • Notch signaling in regulating fibroblast activation in the tumor microenvironment (TME) is well established. Activation of Notch signaling is generally tightly regulated by direct interaction with ligand-expressing cells, and problems in the regulation of Notch signaling result in developmental abnormalities or cancer. Interestingly, Notch activity is associated with both oncogenic and tumor-suppressive functions, which depend on the complex microenvironment of cellular responses that Notch induces. Stromal fibroblasts with an activated Notch pathway can attenuate melanoma growth and inhibit tumor angiogenesis, in part through upregulation of Wnt-induced signaling protein-1 (WISP-1). These findings will help elucidate the molecular mechanisms for the Notch1-dependent tumor-regulatory role in CAFs in other cancer types as well.
  • WISP-1 Wnt-induced signaling protein-1
  • TME tumor microenvironment
  • phagocyte-mediated clearance has been reported to suppress antitumor immune responses.
  • tumors can evade immune surveillance by inhibiting recognition for efferocytosis.
  • the anti-inflammatory and anti-inflammatory lipid autacoids specifically inhibit debris-stimulated cancer progression by promoting the clearance of cellular debris through macrophage phagocytosis in multiple tumor types.
  • macrophages exposed to UV-irradiated apoptotic lung cancer cells showed upregulation of exosome phosphatase and tensin homolog (PTEN) and peroxisome proliferator-activated receptor-gamma (PPARr).
  • PTEN exosome phosphatase and tensin homolog
  • PPARr peroxisome proliferator-activated receptor-gamma
  • the present inventors studied how the interaction between CAFs and apoptotic cancer cells regulates the migration and invasion of cancer cells and CAFs. It was confirmed that WISP-1 was induced to inhibit migration and invasion of cancer cells and CAFs themselves. In addition, after intratumoral administration of apoptotic lung cancer cells, inhibition of CAFs activation and cancer cell migration and invasion were confirmed. Furthermore, the present invention was completed by confirming the effect of inhibiting lung metastasis after intratumoral administration of the co-culture of apoptosis lung cancer cells and CAFs.
  • One aspect is to provide a pharmaceutical composition for inhibiting cancer metastasis containing a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured.
  • CAFs cancer-associated fibroblasts
  • Another aspect is to provide a pharmaceutical composition for inhibiting cancer metastasis, containing cancer-associated fibroblasts exposed to killed cancer cells.
  • Another aspect is to provide a health functional food for inhibiting cancer metastasis containing a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.
  • Another aspect is to provide a method for preparing a composition for inhibiting cancer metastasis, comprising co-cultivating cancer-related fibroblasts and apoptotic cancer cells.
  • Another aspect is to provide a method for inhibiting cancer metastasis, including the step of treating a subject with a culture medium in which cancer-related fibroblasts and apoptosis-killed cancer cells are co-cultured.
  • Another aspect is to provide a method for suppressing cancer metastasis, including treating a subject with Wnt-induced signaling protein-1 (WISP-1).
  • WISP-1 Wnt-induced signaling protein-1
  • Another aspect is to provide a method for preventing or treating cancer, which includes the step of treating a subject with a culture medium in which cancer-related fibroblasts and apoptosis cancer cells are co-cultured.
  • Another aspect is to provide a method for preventing or treating cancer, including treating a subject with Wnt-induced signaling protein-1 (WISP-1).
  • WISP-1 Wnt-induced signaling protein-1
  • Another aspect is to provide the use of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured for the manufacture of a drug for inhibiting cancer metastasis.
  • Another aspect is to provide use of WISP-1 (Wnt-induced signaling protein-1) for the manufacture of a drug for suppressing cancer metastasis.
  • WISP-1 Wi-induced signaling protein-1
  • Another aspect is to provide the use of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured for the manufacture of a drug for preventing or treating cancer.
  • WISP-1 Wnt-induced signaling protein-1
  • Another aspect is to provide a use for suppressing cancer metastasis of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.
  • Another aspect is to provide a use for inhibiting cancer metastasis of a pharmaceutical composition for inhibiting cancer metastasis containing a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.
  • Another aspect is to provide a use of Wnt-induced signaling protein-1 (WISP-1) to inhibit cancer metastasis.
  • WISP-1 Wnt-induced signaling protein-1
  • Another aspect is to provide a use for preventing or treating cancer of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.
  • Another aspect is to provide a pharmaceutical composition containing a culture medium in which cancer-related fibroblasts and killed cancer cells are co-cultured for use in preventing or treating cancer.
  • Another aspect is to provide a use for cancer prevention or treatment of Wnt-induced signaling protein-1 (WISP-1).
  • WISP-1 Wnt-induced signaling protein-1
  • the present invention provides a pharmaceutical composition for inhibiting cancer metastasis containing a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured.
  • CAFs cancer-associated fibroblasts
  • the culture medium in which the CAFs and the apoptotic cancer cells are co-cultured has an effect of inhibiting metastasis of cancer cells.
  • CAFs Cancer-Associated Fibroblasts
  • ⁇ -SMA smooth muscle actin
  • the CAFs are colorectal cancer.
  • its presence has been confirmed in various cancers such as lung cancer, prostate cancer, breast cancer, stomach cancer, cholangiocarcinoma, and basal cell carcinoma.
  • the cancers associated with the CAFs include brain tumor, head and neck cancer, breast cancer, lung cancer, esophageal cancer, stomach cancer, duodenal cancer, appendix cancer, colon cancer, rectal cancer, liver cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, anal cancer, renal cancer, ureter cancer, and bladder cancer.
  • prostate cancer penile cancer, testicular cancer, uterine cancer, ovarian cancer, vulvar cancer, vaginal cancer, and solid cancer such as skin cancer.
  • the CAFs are related to cancer and may be fibroblasts related to malignant solid tumors.
  • the CAFs are fibroblasts related to sarcomas such as fibrosarcoma, malignant fibrous histiocytoma, liposarcoma, rhabdomyosarcoma, leiomyosarcoma, angiosarcoma, malignant skin cancer, lymphangiosarcoma, synovial sarcoma, chondrosarcoma, osteosarcoma, etc. It may be, and may be fibroblasts related to lung cancer, stomach cancer, breast cancer, colon cancer or prostate cancer.
  • the CAFs can have enhanced phagocytic ability by the interaction of Notch1 signaling and BAI1/Rac1 signaling.
  • apoptotic cancer cells may be those in which apoptosis is induced by irradiating cancer cells with light of a specific wavelength.
  • the irradiation of light of the specific wavelength may be ultraviolet (Ultra-violet ray, UV) irradiation.
  • the wavelength may be irradiated for 5 to 30 minutes with a wavelength of 100 to 400 nm.
  • co-culture can be achieved by culturing CAFs and apoptotic cancer cells together.
  • CAFs may be cultured with apoptotic cancer cells in X-VIVO or serum-free DMEM medium for 20 to 30 hours.
  • culture medium means a culture product obtained through co-cultivation of 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.
  • the carcinoma of the killed cancer cells is 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, It may be at least one selected from the group consisting of testicular cancer, laryngeal cancer, oral cancer, head and neck cancer, thyroid cancer, liver cancer, bladder cancer, osteosarcoma, lymphoma, and leukemia.
  • the carcinoma may be at least one selected from the group consisting of lung cancer, breast cancer, stomach cancer, colon cancer, and prostate cancer
  • the lung cancer may be lung adenocarcinoma or non-small cell lung cancer.
  • the lung adenocarcinoma cells may be the 344SQ cell line
  • the non-small cell lung cancer cells may be the A549 cell line
  • the colon cancer cells may be the HCT116 cell line
  • the breast cancer cells may be the MCF-7 cell line.
  • the culture medium may contain WISP-1 (Wnt-induced signaling protein-1) as an active ingredient.
  • 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. do.
  • the WISP-1 may be generated by Notch1 signaling.
  • Notch1 signaling is cell contact-dependent through Notch receptors and plays an important role in development, regeneration, and maintenance of homeostasis, and the activity of Notch signaling is associated with both oncogenic and tumor-suppressive functions.
  • Notch1-WISP-1 signaling is known to determine the regulatory role of mesenchymal stem cell-derived stromal fibroblasts in melanoma invasion and metastasis.
  • the Notch1 signaling may be initiated by Dll1 (Delta-like ligand 1).
  • Dll1 is a type of Notch Delta ligand that plays a role in regulating cell fate decisions during blood formation and is known to be involved in cell-cell communication.
  • the Notch1 signaling may be enhanced by BAI1/Rac1 (Brain angiogenesis inhibitor 1/Ras-related C3 botulinum toxin substrate 1) signaling.
  • BAI1 is a type of aggregation GPCR that acts to inhibit angiogenesis
  • Rac1 is a type of GTPase that plays a role in regulating cell growth, cytoskeleton reorganization, cell cycle, cell-cell aggregation and migration, and the like.
  • the composition can enhance the phagocytic ability of cancer-related fibroblasts by the interaction of Notch1 signaling and BAI1/Rac1 signaling.
  • efferocytosis means phagocytosis to remove apoptotic cells.
  • the phagocytic ability refers to the ability of CAFs to remove apoptotic cancer cells through phagocytosis.
  • the "interaction” may mean crosstalk between the Notch1 signaling and the BAI1/Rac1 signaling, and more specifically, may mean positive crosstalk.
  • crosstalk means a phenomenon in which one or more components of a signal transduction pathway affect other components. Positive crosstalk refers to a phenomenon in which one signaling reinforces another signaling. Negative crosstalk refers to a phenomenon in which one signaling suppresses another signaling.
  • the BAI1/Rac1 signaling can promote phagocytosis of apoptotic cancer cells by the cancer-associated fibroblasts.
  • the composition can enhance WISP-1 production by the interaction of Notch1 signaling and BAI1/Rac1 signaling.
  • the composition can reduce activation markers in cancer-associated fibroblasts.
  • the activation marker may be one or more selected from the group consisting of Acta2, Col1 ⁇ 1, Fn, Itg beta 1, Spp1, Pdgfr ⁇ , Pdgfr beta, and Mmp1a, 2, 9, and 12.
  • the composition can reduce growth factors and chemokines in cancer-associated fibroblasts.
  • the growth factor may be one or more selected from the group consisting of Vegfa, Hgf, Cxcl12 and Cxcl14.
  • the composition can increase Notch1-related molecules in cancer-related fibroblasts.
  • the Notch1-related molecule may be one or more selected from the group consisting of Notch1, WISP-1 (Ccn4), Hey1, Hey2, Hes1, and Hes5.
  • metastasis refers to a state in which a malignant tumor has spread to other tissues away from an organ where it has occurred, and can be used as a marker of a malignant tumor.
  • the metastasis may include migration and invasion of cancer cells.
  • migration means that cells move to a specific location in response to a specific external signal
  • invasion means that cells expand and penetrate other nearby tissues.
  • the present invention provides a pharmaceutical composition for inhibiting cancer metastasis containing cancer-related fibroblasts exposed to apoptotic cancer cells.
  • CAFs exposed to the killed cancer cells contain WISP-1, and thus have an effect of suppressing metastasis of cancer cells.
  • exposure means a process of contacting or stimulating apoptosis cancer cells and CAFs to enable cell-to-cell interaction, and is distinguished from a conditioned medium in which apoptosis cancer cells and CAFs are co-cultured.
  • the pharmaceutical composition according to the present invention may include a "pharmaceutically acceptable carrier".
  • the pharmaceutically acceptable carrier is one commonly used in formulation and includes, but is not limited to, saline solution, sterile water, Ringer's solution, buffered saline solution, cyclodextrin, dextrose solution, maltodextrin solution, glycerol, ethanol, liposome, etc. It is not, and if necessary, other conventional additives such as antioxidants and buffers may be further included. In addition, diluents, dispersants, surfactants, binders, lubricants, etc.
  • compositions for injections such as aqueous solutions, suspensions, emulsions, etc., pills, capsules, granules, or tablets.
  • a suitable pharmaceutically acceptable carrier and formulation it can be preferably formulated according to each component using the method disclosed in Remington's literature.
  • the pharmaceutical composition of the present invention is not particularly limited in dosage form, but may be formulated as an injection, an inhalant, an external preparation for the skin, or an oral intake.
  • the pharmaceutical composition of the present invention may be administered orally or parenterally (for example, intravenously, subcutaneously, applied to the skin, nasal cavity, or respiratory tract) according to the desired method, and the dosage is determined according to the patient's condition and weight, disease Depending on the degree, drug form, administration route and time, it can be appropriately selected by those skilled in the art.
  • composition according to the present invention is administered in a pharmaceutically effective amount.
  • pharmaceutically effective amount means an amount sufficient to treat a disease with a reasonable benefit / risk ratio applicable to medical treatment, and the effective dose level is the type, severity, and activity of the drug of the patient's disease , sensitivity to the drug, time of administration, route of administration and excretion rate, duration of treatment, factors including drugs used concurrently, and other factors well known in the medical field.
  • the composition according to the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, may be administered sequentially or simultaneously with conventional therapeutic agents, and may be administered single or multiple times. Considering all of the above factors, it is important to administer an amount that can obtain the maximum effect with the minimum amount without side effects, which can be easily determined by those skilled in the art.
  • the effective amount of the composition according to the present invention may vary depending on the patient's age, sex, and weight, and is generally 0.001 to 150 mg per 1 kg of body weight, preferably 0.01 to 100 mg per day or every other day, or 1 It can be administered in 1 to 3 divided doses per day. However, since it may increase or decrease depending on the route of administration, severity of obesity, gender, weight, age, etc., the dosage is not limited to the scope of the present invention in any way.
  • the present invention provides a health functional food for inhibiting cancer metastasis, which contains a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.
  • the health functional food of the present invention may be formulated as one selected from the group consisting of tablets, pills, powders, granules, powders, capsules and liquid formulations by further including one or more of carriers, diluents, excipients and additives.
  • carriers diluents, excipients and additives.
  • examples of foods to which the extract of the present invention can be added include various foods, powders, granules, tablets, capsules, syrups, beverages, gum, tea, vitamin complexes, health functional foods, and the like.
  • Additives that may be further included in the present invention include natural carbohydrates, flavors, nutrients, vitamins, minerals (electrolytes), flavors (synthetic flavors, natural flavors, etc.), colorants, fillers (cheese, chocolate, etc.), One or more components selected from the group consisting of pectic acid and its salts, alginic acid and its salts, organic acids, protective colloidal thickeners, pH adjusters, stabilizers, preservatives, antioxidants, glycerin, alcohols, carbonating agents and fruit flesh may be used. .
  • natural carbohydrates examples include monosaccharides such as glucose, fructose, and the like; disaccharides such as maltose, sucrose and the like; and polysaccharides such as conventional sugars such as dextrins, cyclodextrins, and the like, and sugar alcohols such as xylitol, sorbitol, and erythritol.
  • natural flavoring agents thaumatin, stevia extract (eg, rebaudioside A, glycyrrhizin, etc.) and synthetic flavoring agents (saccharin, aspartame, etc.) can be advantageously used.
  • the health functional food of the present invention is various nutrients, vitamins, minerals (electrolytes), flavors such as synthetic flavors and natural flavors, colorants and enhancers (cheese, chocolate, etc.), pectic acid and its salts, alginic acid and salts thereof, organic acids, protective colloidal thickeners, pH adjusting agents, stabilizers, preservatives, glycerin, alcohol, carbonating agents used in carbonated beverages, and the like.
  • the composition according to the present invention may contain fruit flesh for preparing natural fruit juice and vegetable beverages. These components may be used independently or in combination.
  • carrier examples include, but are not limited to, lactose, dextrose, sucrose, sorbitol, mannitol, erythritol, starch, gum acacia, calcium phosphate, alginate, gelatin, calcium phosphate, calcium Silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, polyvinylpyrrolidone, methylcellulose, water, sugar syrup, methylcellulose, methyl hydroxy benzoate, propylhydroxy benzoate, talc, magnesium stearate And at least one selected from the group consisting of mineral oil is preferably used.
  • the health functional food of the present invention When formulating the health functional food of the present invention, it is prepared using diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants.
  • diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants.
  • the present invention provides a method for preparing a composition for inhibiting cancer metastasis, comprising co-cultivating cancer-related fibroblasts and apoptotic cancer cells.
  • the killing of cancer cells may be induced through UV irradiation, and the UV irradiation may be performed for 5 to 30 minutes with a wavelength of 100 to 400 nm. In one embodiment, the UV irradiation may be performed with a wavelength of 150 to 350 nm for 20 minutes or with a wavelength of 200 to 300 nm for 10 to 15 minutes.
  • the step of culturing may be to culture in X-VIVO medium for 24 hours to make it serum-starved.
  • the culture medium may be replaced with X-VIVO or serum-free DMEM medium containing killed cancer cells.
  • the co-cultivation may be performed for 10 to 30 hours, 15 to 25 hours, or 18 to 24 hours after medium replacement.
  • the present invention provides a method for suppressing cancer metastasis, comprising the step of treating a subject with a culture medium in which cancer-related fibroblasts and apoptosis-killed cancer cells are co-cultured.
  • the step of treating the subject with the culture medium is performed by treating the subject with a pharmaceutical composition for inhibiting cancer metastasis containing a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured. It could be
  • treatment may mean any act of adding or administering the culture medium to a subject according to experimental requirements.
  • subject means a subject in need of a method for preventing, controlling or treating a disease, and may include a mammal.
  • the mammal may include humans or primates, mice, cows, dogs, horses, pigs, and the like.
  • the present invention provides a method for inhibiting cancer metastasis, comprising treating a subject with Wnt-induced signaling protein-1 (WISP-1).
  • WISP-1 Wnt-induced signaling protein-1
  • the present invention provides a method for preventing or treating cancer, comprising the step of treating a subject with a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.
  • the culture medium in which the cancer-related fibroblasts and apoptosis cancer cells are co-cultured can be treated in the form of a pharmaceutical composition containing the same.
  • prevention may refer to any activity that inhibits or delays the onset of cancer
  • treatment may mean any activity that improves or beneficially changes cancer.
  • the present invention provides a method for preventing or treating cancer, including treating a subject with Wnt-induced signaling protein-1 (WISP-1).
  • WISP-1 Wnt-induced signaling protein-1
  • the present invention provides the use of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured for the manufacture of a drug for inhibiting cancer metastasis.
  • the present invention provides a use of WISP-1 (Wnt-induced signaling protein-1) for the preparation of a drug for suppressing cancer metastasis.
  • WISP-1 Wi-induced signaling protein-1
  • the present invention provides the use of a culture medium in which cancer-related fibroblasts and killed cancer cells are co-cultured for the manufacture of a drug for preventing or treating cancer.
  • the present invention provides a use of WISP-1 (Wnt-induced signaling protein-1) for the manufacture of a drug for preventing or treating cancer.
  • WISP-1 Wi-induced signaling protein-1
  • the present invention provides a use for inhibiting cancer metastasis of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.
  • the present invention provides a use for inhibiting cancer metastasis of a pharmaceutical composition for inhibiting cancer metastasis, which contains a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.
  • the present invention provides a use of WISP-1 (Wnt-induced signaling protein-1) to inhibit cancer metastasis.
  • the present invention provides a use for preventing or treating cancer of a culture medium in which cancer-related fibroblasts and killed cancer cells are co-cultured.
  • the present invention provides a use for preventing or treating cancer of a pharmaceutical composition containing a culture medium in which cancer-related fibroblasts and killed cancer cells are co-cultured.
  • the present invention provides a use of Wnt-induced signaling protein-1 (WISP-1) for preventing or treating cancer.
  • WISP-1 Wnt-induced signaling protein-1
  • CAFs exposed to the culture medium or killed cancer cells of the present invention can be usefully used as cancer metastasis suppressors.
  • FIG. 1 is a diagram showing that a conditioned medium (CM) containing a culture medium of cancer-associated fibroblasts (CAFs) co-cultured with apoptotic cancer cells inhibits migration and invasion of cancer cells.
  • Figure 1a shows a control, CAF conditioned medium (CAF CM), and 344SQ co-culture conditioned medium (ApoSQ-CAF CM) killed with CAFs in the presence or absence of TGF-beta 1 using 344SQ, a lung adenocarcinoma cell line.
  • It is a diagram measuring the migration and invasion of 344SQ cells in the 344SQ co-culture conditioned medium (NecSQ-CAF CM) group with CAFs and necrosis.
  • Figure 1b shows the control, CAF CM, CAFs and apoptosis A549 co-culture conditioned medium (ApoA-CAF CM), CAFs and A549 co-culture in the presence or absence of TGF-beta 1 using A549, a non-small cell lung cancer cell line. It is a diagram measuring migration and invasion of A549 cells in the culture conditioned medium (NecA-CAF CM) group.
  • FIG. 1c is a control, CAF CM, CAFs and apoptosis HCT116 co-culture conditioned medium (ApoH-CAF CM), and CAFs and necrosis HCT116 co-culture in the presence or absence of TGF-beta 1 using HCT116, a colorectal cancer cell line. It is a diagram measuring the migration and invasion of HCT116 cells in the conditioned medium (NecH-CAF CM) group.
  • Figure 1d is a control, CAF CM, CAFs and MCF-7 co-culture conditioned medium (ApoM-CAF CM), CAFs and necrosis in the presence or absence of TGF-beta 1 using MCF-7, a breast cancer cell line, respectively. It is a diagram measuring the migration and invasion of MCF-7 cells in the MCF-7 co-culture conditioned medium (NecM-CAF CM) group.
  • Figure 2 is a diagram confirming that there is no effect of inhibiting migration and invasion of cancer cells by direct treatment of the killed cancer cell culture medium alone and the killed cancer cells.
  • Figure 2a is a diagram showing the migration and invasion of 344SQ cells in the control, CAF CM, ApoSQ CM and NecSQ CM groups, respectively, in the presence of TGF-beta 1.
  • Figure 2b is a diagram measuring 344SQ cell migration and invasion in the control, ApoSQ, and NecSQ groups, respectively, in the presence or absence of TGF-beta 1.
  • FIG. 3 is a diagram showing that ApoSQ-CAF CM inhibits the signaling pathway activity related to TGF-beta 1 in 344SQ cells using immunoblot analysis.
  • Figure 3a is a diagram showing the results of immunoblot analysis of TGF-beta 1-related signaling pathway expressed after treatment of CAF CM and ApoSQ-CAF CM together with TGF-beta 1 in 344SQ cells.
  • Figure 3b is a diagram showing the total and phosphorylated amounts of Smad2, Smad3, FAK, AKT, SrC, ERK, and P38 expressed after treatment with CAF CM and ApoSQ-CAF CM in 344SQ cells.
  • FIG. 4 is a diagram showing that ApoSQ-CAF CM suppresses MMP2 and MMP12 mRNA expression and protein expression in 344SQ cells using qRT-PCR and immunoblot analysis.
  • A A diagram showing the levels of MMP2 and MMP12 mRNA expression after treatment of CAF CM and ApoSQ-CAF CM with TGF-beta in 344SQ cells.
  • B 344SQ cells treated with CAF CM and ApoSQ-CAF CM, and then immunoblot analysis of MMP2 and MMP12 proteins expressed and a diagram showing the protein amounts.
  • 5 is a diagram showing that only ApoSQ inhibits cell migration and invasion when CAFs are brought into contact with ApoSQ and NecSQ, respectively.
  • Figure 6 shows that when CAFs were exposed to ApoSQ-CAF CM, NecSQ-CAF CM, ApoA-CAF CM, NecA-CAF CM, ApoH-CAF CM and NecH-CAF CM in the presence or absence of TGF-beta 1, respectively, death It is a diagram showing that only the conditioned cancer cells and CAF-conditioned medium inhibit cell migration and invasion.
  • Figure 6a is a diagram showing migration and invasion of CAFs after treatment with CAF CM, ApoSQ-CAF CM, and NecSQ-CAF CM.
  • Figure 6b is a diagram showing migration and invasion of CAFs after treatment with CAF CM, ApoA-CAF CM, and NecA-CAF CM.
  • Figure 6c is a diagram showing migration and invasion of CAFs after treatment with CAF CM, ApoH-CAF CM, and NecH-CAF CM.
  • Figure 6d is a diagram showing migration and invasion of CAFs after treatment with CAF CM, ApoM-CAF CM, and NecM-CAF CM.
  • FIG. 7 is a diagram confirming that the culture medium of killed cancer cells alone has no effect of inhibiting migration and invasion of CAFs.
  • FIG. 8 is a diagram showing that ApoSQ inhibits the expression of CAF activation markers in CAFs using qRT-PCR, immunoblot analysis, and immunofluorescence staining.
  • Figure 8a is a diagram showing the results of analyzing the mRNA expression level of CAF activation markers when ApoSQ and NecSQ were treated with CAFs, and the results of immunoblot analysis of CAF activation markers and protein amounts when ApoSQ and NecSQ were treated with CAFs.
  • 8b is a diagram showing the fluorescence intensity of ⁇ -SMA when CAFs were treated with ApoSQ and NecSQ.
  • 9 is a diagram showing that ApoSQ inhibits the signaling pathway activity related to TGF-beta 1 in CAFs using immunoblot analysis.
  • 9a is a diagram showing the results of immunoblot analysis of TGF-beta 1-related signaling pathways after CAFs were treated with ApoSQ together with TGF-beta 1;
  • Figure 9b is a diagram showing the total and phosphorylated amounts of signaling pathways, that is, Smad2, Smad3, AKT, FAK, SrC, ERK, and P38, after CAFs were treated with ApoSQ along with TGF-beta 1.
  • FIG. 10 is a diagram showing that ApoSQ inhibits the expression of MMP mRNA and protein in CAFs using qRT-PCR and immunoblot analysis.
  • A A diagram showing the mRNA expression levels of MMP2 and MMP12 after ApoSQ was treated in CAFs.
  • B A diagram showing the results of immunoblot analysis and protein amounts of MMP2 and MMP12 proteins after CAFs were treated with ApoSQ.
  • FIG. 11 is a result of cytokine array analysis in CAF CM, ApoSQ-CAF CM and ApoSQ CM, and the expression of leukemia inhibitory factor (LIF) and WISP-1 among 111 types of cytokines was compared with CAF CM and ApoSQ-CAF CM groups. It is a diagram confirming that the cytokine increased the most in ApoSQ-CAF CM.
  • LIF leukemia inhibitory factor
  • FIG. 12 is a diagram showing that the expression level of WISP-1 decreases when WISP-1 is knocked down.
  • A A diagram showing the results of immunoblot analysis and the level of WISP-1 protein expression measured after transfecting CAFs with WISP-1-specific siRNA.
  • B It is a diagram showing the amount of WISP-1 measured by ELISA in the culture medium after CAFs were transfected with siRNA specific to WISP-1.
  • FIG. 13 is a diagram showing that the effect of inhibiting cell migration and invasion is reversed when CAFs are transfected with WISP-1 siRNA.
  • Figure 13a is a diagram showing migration and invasion of 344SQ cells upon treatment with CAF CM and ApoSQ-CAF CM in the presence or absence of WISP-1 siRNA.
  • Figure 13b is a diagram showing migration and invasion of CAFs upon ApoSQ treatment in the presence or absence of WISP-1 siRNA.
  • FIG. 14 is a diagram showing that the expression level of LIF decreases when LIF is knocked down.
  • A A diagram showing the result of immunoblot analysis and the amount of LIF protein measured after transfection of CAFs with LIF-specific siRNA.
  • B A diagram showing the amount of LIF measured by ELISA in CAF CM and ApoSQ-CAF CM after CAFs were transfected with LIF-specific siRNA.
  • C A diagram showing the amount of WISP-1 measured by ELISA in CAF CM and ApoSQ-CAF CM after CAFs were transfected with LIF-specific siRNA.
  • FIG. 15 is a diagram showing that the effect of inhibiting cell migration and invasion is maintained when CAFs are transfected with LIF siRNA.
  • Figure 15a is a diagram showing migration and invasion of 344SQ cells when treated with CAF CM and ApoSQ-CAF CM in the presence or absence of LIF siRNA.
  • Figure 15b is a diagram showing migration and invasion of CAFs upon ApoSQ treatment in the presence or absence of LIF siRNA.
  • FIG. 16 is a diagram showing that the expression level of WISP-1 increases when WISP-1 is overexpressed.
  • A A diagram showing the results of immunoblot analysis and protein expression level measured after transfection of CAFs with mock vector and WISP-1.
  • B A diagram showing the amount of WISP-1 measured by ELISA in CAF CM and ApoSQ-CAF CM after CAFs were transfected with mock vector and WISP-1.
  • 17 is a diagram showing that cell migration and invasion inhibitory effects are increased when CAFs are transfected with WISP-1 to overexpress WISP-1.
  • 17a is a diagram showing migration and invasion of 344SQ cells when treated with CAF CM and ApoSQ-CAF CM, depending on whether or not WISP-1 was transfected.
  • 17b is a diagram showing migration and invasion of CAFs upon ApoSQ treatment, depending on whether or not WISP-1 was transfected.
  • 18 is a diagram showing that the anti-WISP-1 antibody treatment of ApoSQ-CAF CM reverses the effect of inhibiting migration and invasion of 344SQ cells.
  • FIG. 19 is a diagram showing that migration and invasion of 344SQ cells or CAFs are inhibited in a concentration-dependent manner when recombinant WISP-1 is added.
  • Figure 19a is a diagram showing migration and invasion of 344SQ cells according to rWISP-1 concentration.
  • Figure 19b is a diagram showing migration and invasion of CAFs according to rWISP-1 concentration.
  • FIG. 20 is a diagram showing that rWISP-1 inhibits the activity of the signaling pathway related to TGF-beta 1 and MMP expression in 344SQ cells in a concentration-dependent manner using immunoblot analysis.
  • 20a is a diagram showing the results of immunoblot analysis of TGF-beta 1-related signaling molecules for each concentration of rWISP-1.
  • Figure 20b is a diagram showing the total amount and phosphorylation amount of TGF-beta 1-related signaling pathways, that is, Smad2, Smad3, FAK, AKT, SrC, ERK, and P38, when rWISP-1 was treated by concentration.
  • (A) is a diagram showing the amount of MMP2 and MMP12 mRNA expressed by rWISP-1 concentration.
  • (B) is a diagram showing the results of immunoblot analysis and protein amounts of MMP2 and MMP12 proteins expressed by rWISP-1 concentrations.
  • 21 is a diagram showing that when rWISP-1 is treated with TGF-beta 1, the activity of the signaling pathway related to TGF-beta 1 and the expression of MMP are inhibited in CAFs using immunoblot analysis and qRT-PCR.
  • 21a is a diagram showing the results of immunoblot analysis of the TGF-beta 1-related signaling pathway upon treatment with rWISP-1.
  • Figure 21b is a diagram showing the total and phosphorylated amounts of TGF-beta 1-related signaling pathways, that is, Smad2, Smad3, FAK, AKT, SrC, ERK and P38 upon rWISP-1 treatment.
  • (A) is a diagram showing the change in the mRNA expression amount of MMP2 and MMP12 when rWISP-1 and TGF-beta 1 are treated together
  • (B) is a diagram showing the change in the mRNA expression amount of rWISP-1 and TGF-beta 1 when treated together with MMP2 and MMP12 It is a diagram showing the results of protein immunoblot analysis and the amount of protein.
  • 22 is a diagram showing that the inhibition of migration and invasion according to the addition of WISP-1 is reversed in 344SQ cells or CAFs when a specific anti-integrin is added.
  • 22a is a diagram showing that the inhibition of migration and invasion according to the addition of WISP-1 is reversed in 344SQ cells when an anti-integrin ⁇ v or beta 3 antibody is added.
  • 22b is a diagram showing that the inhibition of migration and invasion according to the addition of WISP-1 is reversed in CAFs when an anti-integrin ⁇ v or beta 5 antibody is added.
  • 23 is a diagram showing that the activation of the signaling pathway related to TGF-beta 1 and the suppression of MMP expression are reversed in 344SQ cells when a specific anti-integrin is added using immunoblot analysis and qRT-PCR.
  • 23a is a diagram showing the results of immunoblot analysis of TGF-beta 1-related signaling molecules according to the addition of rWISP-1 and anti-integrin ⁇ v or beta 3 antibodies.
  • 23b is a diagram showing the mRNA amounts of MMP2 and MMP12 according to the addition of rWISP-1 and anti-integrin ⁇ v or beta 3 antibodies.
  • 23c is a diagram showing the results of immunoblot analysis of MMP2 and MMP12 proteins according to the addition of WISP-1 and anti-integrin ⁇ v or beta 3 antibodies.
  • FIG. 24 is a diagram showing that when a specific anti-integrin is added using immunoblot analysis and qRT-PCR, the TGF-beta 1-related signaling pathway activity and inhibition of MMP expression are reversed by the addition of WISP-1 in CAFs.
  • 24a is a diagram showing the results of immunoblot analysis of TGF-beta 1 related signaling molecules according to the addition of rWISP-1 and anti-integrin ⁇ v or beta 5 antibodies.
  • 24b is a diagram showing the mRNA amounts of MMP2 and MMP12 according to the addition of rWISP-1 and anti-integrin ⁇ v or beta 5 antibodies.
  • 24c is a diagram showing the results of immunoblot analysis of MMP2 and MMP12 proteins according to the addition of WISP-1 and anti-integrin ⁇ v or beta 5 antibodies.
  • 25 is a diagram showing that the amount of proteins of factors related to the Notch1 signaling pathway expressed in CAFs according to the presence or absence of ApoSQ is increased.
  • A A diagram showing the results of protein immunoblot analysis according to the presence or absence of ApoSQ in CAFs.
  • B A diagram showing the amount of WISP-1 expressed in CAF CM, ApoSQ-CAF CM and NecSQ-CAF CM.
  • 26 is a diagram showing that the mRNA expression levels of Hes1 and WISP-1, which are target genes of the Notch signaling pathway, and the activity of 4 ⁇ CSL luciferase are increased when ApoSQ or NecSQ is treated in CAFs.
  • A A diagram showing the mRNA expression levels of Hes1 and WISP-1 analyzed by qRT-PCR.
  • B A diagram showing the activity of 4 ⁇ CSL luciferase, an overall Notch signal transcriptional effector, upon ApoSQ treatment.
  • FIG. 27 is a diagram showing changes in immunofluorescence staining for NICD1 and WISP-1 according to ApoSQ or NecSQ treatment in CAFs.
  • 28 is a diagram showing that WISP-1 protein expression is suppressed when Notch1 is knocked down through siRNA.
  • 28a is a diagram showing the results of immunoblot analysis measured after CAFs were transfected with Notch1 siRNA.
  • 28B (A) is a diagram showing the immunoblot analysis results and protein amounts for NICD1, Hes1, and WISP-1 measured according to the presence or absence of ApoSQ or NecSQ after CAFs were transfected with Notch1 siRNA, (B) is a diagram showing the ELISA results of WISP-1 in CAF CM and ApoSQ-CAF CM after transfection of CAFs with Notch1 siRNA.
  • FIG. 29 is a diagram showing that when Notch1 is knocked down through siRNA, the anti-migration and anti-invasion effects of ApoSQ-CAF CM and ApoSQ are reversed in 344SQ cells and CAFs, respectively.
  • Figure 29a is a diagram showing migration and invasion of 344SQ cells when treated with CAF CM and ApoSQ-CAF CM in the presence or absence of Notch1 siRNA.
  • Figure 29b is a diagram showing migration and invasion of CAFs upon ApoSQ treatment in the presence or absence of Notch1 siRNA.
  • FIG. 30 is a diagram showing that when Notch1 activity is inhibited through DAPT, the expression of proteins related to the Notch1 signaling pathway is inhibited.
  • A A diagram showing the immunoblot analysis results and protein expression levels for NICD1, Hes1, and WISP-1 measured in the presence or absence of ApoSQ or NecSQ after CAFs were treated with 10 ⁇ M DAPT.
  • B A diagram showing the ELISA results of WISP-1 measured in CAF CM, ApoSQ-CAF CM, and NecSQ-CAF CM after CAFs were treated with 10 ⁇ M DAPT.
  • FIG. 31 is a diagram showing that when Notch1 is inhibited through DAPT, the anti-migration and anti-invasion effects of ApoSQ-CAF CM and ApoSQ are reversed in 344SQ cells and CAFs, respectively.
  • Figure 31a is a diagram showing migration and invasion of 344SQ cells upon treatment with CAF CM and ApoSQ-CAF CM in the presence or absence of DAPT.
  • Figure 31b is a diagram showing migration and invasion of CAFs upon ApoSQ treatment in the presence or absence of DAPT.
  • DLL Delta-like ligand
  • JAG Jagged-like
  • JAG2 Jagged-like
  • JAG2 Jagged-like
  • FIG. 33 is a diagram showing that only Dll1 expression is increased in killed cancer cells 344SQ, A549, and HCT116.
  • A A diagram showing the results of immunoblot analysis for Dll1, Dll3, Dll4, JAG1 and JAG2 in apoptotic cancer cells 344SQ, A549 and HCT116.
  • B A diagram showing the amount of Dll1, Dll3, Dll4, JAG1 and JAG2 proteins in the killed cancer cells 344SQ, A549 and HCT116.
  • FIG. 34 is a diagram showing that expression of proteins related to the Notch1 signaling pathway is suppressed in CAFs after ApoSQ treatment when Dll1 is neutralized with an anti-Dll1 antibody.
  • A A diagram showing the immunoblot analysis results and protein amounts for NICD1, Hes1, and WISP-1 measured in the presence or absence of anti-Dll1 antibody in CAFs contacted with ApoSQ.
  • B A diagram showing the ELISA results of WISP-1 measured in the presence or absence of anti-Dll1 antibody in CAFs contacted with ApoSQ.
  • 35 is a diagram showing that expression of proteins related to the Notch1 signaling pathway is suppressed in CAFs when Dll1 is knocked down through siRNA.
  • 35A is a diagram showing the results of immunoblot analysis and the amount of Dll1 protein measured after transfection of 344SQ cells with Dll1 siRNA.
  • FIG. 35b (A) is a diagram showing the immunoblot analysis results and protein amounts for NICD1, Hes1, and WISP-1 measured according to the presence or absence of ApoSQ in CAFs after transfection of 344SQ cells with Dll1 siRNA, (B ) is a diagram showing the ELISA results of WISP-1 measured according to the presence or absence of ApoSQ in CAFs after transfecting 344SQ cells with Dll1 siRNA.
  • FIG. 36 is a diagram showing that, when CAFs in contact with ApoSQ are treated with annexin V, brain-specific angiogenesis inhibitor 1 (BAI1) signals are inhibited, thereby inhibiting ApoSQ phagocytosis, Notch1 activation, and WISP-1 secretion in CAFs.
  • Figure 36a is a diagram showing the percentage of CAFs phagocytosis after exposure to ApoSQ cells in the presence or absence of annexin V by flow cytometry. In FIG.
  • (A) is a diagram showing the immunoblot analysis results for NICD1, Hes1, and WISP-1 in CAFs contacted with ApoSQ in the presence or absence of annexin V
  • (B) is a diagram showing the results of immunoblot analysis in the presence or absence of annexin V.
  • (C) shows the ELISA results of WISP-1 measured in CAFs in contact with ApoSQ in the presence or absence of annexin V. It is also
  • 37 is a diagram showing that when CAFs in contact with ApoSQ were transfected with BAI1 siRNA, phagocytosis of CAFs, Notch1 activation, and WISP-1 secretion were suppressed as BAI1 signal was suppressed.
  • 37a is a diagram showing the results of immunoblot analysis and the amount of BAI1 protein measured after CAFs were transfected with BAI1 siRNA.
  • Figure 37b is a diagram showing the phagocytosis of CAFs measured in % by flow cytometry after CAFs were transfected with BAI1 siRNA and exposed to ApoSQ cells. In FIG.
  • (A) is a diagram showing the immunoblot analysis results for NICD1, Hes1, and WISP-1 measured in the presence or absence of ApoSQ after CAFs were transfected with BAI1 siRNA
  • (B) is a diagram showing CAFs with BAI1 siRNA. After transfection, the amount of NICD1, Hes1, and WISP-1 proteins measured according to the presence or absence of ApoSQ is a diagram. It is a diagram showing the results of ELISA.
  • FIG. 38 is a diagram showing that when CAFs in contact with ApoSQ are treated with an anti-BAI1 antibody, BAI1 signals are suppressed, and phagocytosis, Notch1 activation, and WISP-1 secretion of CAFs are suppressed.
  • Figure 38a is a diagram showing the percentage of CAFs phagocytosis after exposure of ApoSQ cells in the presence or absence of anti-BAI1 antibody by flow cytometry. In FIG.
  • (A) is a diagram showing the results of immunoblot analysis for NICD1, Hes1, and WISP-1 in CAFs contacted with ApoSQ in the presence or absence of anti-BAI1 antibody
  • (B) is a diagram showing the results of anti-BAI1 antibody
  • (C) is WISP-1 measured in CAFs contacted with ApoSQ in the presence or absence of anti-BAI1 antibody. It is a diagram showing the ELISA result of 1.
  • 39 is a diagram showing that when CAFs contacted with ApoSQ were transfected with BAI1-Flag overexpressing BAI1, phagocytosis of CAFs, Notch1 activation, and WISP-1 secretion increased as BAI1 signal increased.
  • 39a is a diagram showing the results of immunoblot analysis and the amount of BAI1 protein measured after CAFs were transfected with BAI1-Flag.
  • Figure 39b is a diagram showing the phagocytosis of CAFs measured in % by flow cytometry after CAFs were transfected with BAI1-Flag and exposed to ApoSQ cells.
  • (A) is a diagram showing the results of immunoblot analysis for NICD1, Hes1, and WISP-1 measured according to the presence or absence of ApoSQ after CAFs were transfected with BAI1-Flag
  • (B) is a diagram showing CAFs transfected with BAI1-Flag.
  • 40 is a diagram showing that ApoSQ increases the activity of Rac1 in a time-dependent manner in CAFs.
  • 41 is a diagram showing that when CAFs contacted with ApoSQ are treated with NSC23766, a Rac1 inhibitor, phagocytosis of CAFs, Notch1 activation, and WISP-1 secretion are inhibited as Rac1 signaling is inhibited.
  • 41a and 41b are diagrams measuring the phagocytic activity of CAFs in contact with ApoSQ in the presence or absence of 100 ⁇ M NSC23766.
  • 41a is a diagram showing the number of CAFs phagocytosing ApoSQ cells in % by flow cytometry.
  • 41B is a diagram illustrating phagocytic activity as a phagocytic index using a confocal microscope. In FIG.
  • (A) is a diagram showing the immunoblot analysis results for NICD1, Hes1, and WISP-1 in CAFs contacted with ApoSQ in the presence or absence of NSC23766, and (B) is a diagram showing the results of immunoblot analysis for ApoSQ and ApoSQ in the presence or absence of NSC23766.
  • (C) is a diagram showing ELISA results of WISP-1 measured in CAFs contacted with ApoSQ in the presence or absence of NSC23766.
  • FIG. 42 is a diagram showing that when CAFs in contact with ApoSQ are transfected with Notch1 siRNA, the activity of Rac1 and the phagocytosis of CAFs are suppressed according to the inhibition of Notch1 signal.
  • A A diagram showing the activity of Rac1 measured after transfecting CAFs with Notch1 siRNA.
  • B After transfecting CAFs with Notch1 siRNA and exposing ApoSQ cells, the phagocytosis of CAFs was measured in % by flow cytometry.
  • FIG. 43 is a diagram showing that when CAFs contacted with ApoSQ were treated with DAPT, the activity of Rac1 and phagocytosis of CAFs were suppressed as the Notch1 signal was suppressed.
  • A A diagram showing the activity of Rac1 measured in the presence or absence of 20 ⁇ M DAPT.
  • B A diagram showing the phagocytosis of CAFs in contact with ApoSQ in the presence or absence of DAPT through flow cytometry.
  • FIG. 44 is a diagram showing an experimental timeline for injecting 344SQ cells subcutaneously on the flanks of syngeneic mice, subcutaneously administering ApoSQ 2 days later, sacrificing the mice 6 weeks later, and isolating Thy1 CAFs from primary tumor tissue. am.
  • Thy1 + CAFs of the control group CAF activation markers (including Acta2, Col1 ⁇ 1, Fn, Itg beta 1, Spp1, Pdgfr ⁇ , and Pdgfr beta), MMPs (including Mmp1a, 2, 9, and 12), growth factors and chemokines (Vegfa, Hgf, It is a diagram measured by qRT-PCR that the mRNA expression level of Notch1-related molecules (including Notch1, Wisp1 (Ccn4), Hey1, Hey2, and Hes1) is increased, while the mRNA expression level of Cxcl12 and Cxcl14 is decreased.
  • 45A is a diagram showing a heat map made based on qRT-PCR results.
  • 45B is a diagram showing qRT-PCR results for each
  • 46 is a diagram showing the experimental timeline of administering ApoSQ and LY3039478 to mice.
  • mice 47 is a diagram showing that there is no significant difference between each group as a result of measuring mouse weight, tumor weight and tumor volume by administering LY3039478 (8 mg/kg), ApoSQ or ApoSQ and LY3039478 (8 mg/kg) to mice .
  • A A diagram showing the mouse weight, tumor weight and tumor volume in each group.
  • B A diagram showing images of tumors in each group.
  • 51 shows a decrease in the expression of ⁇ -SMA and an increase in the expression of NICD1 and WISP-1 through immunocytochemical analysis in Thy1 + CAFs isolated from primary tumor tissues of mice administered with ApoSQ, but when LY3039478 is additionally administered. It is a diagram showing that the effect is reversed.
  • 51A is a diagram showing the expression level of ⁇ -SMA in each group through immunocytochemical analysis.
  • 51b is a diagram showing the expression levels of NICD1 and WISP-1 in each group through immunocytochemical analysis.
  • FIG. 53 is a diagram showing that cell migration and invasion are inhibited in CD326 + tumor cells and Thy1 + CAFs isolated from primary tumor tissues of mice administered with ApoSQ, but the above effects are reversed when LY3039478 is additionally administered.
  • Figure 53a is a diagram measuring migration and invasion of isolated CD326 + tumor cells.
  • Figure 53b is a diagram showing the migration and invasion of isolated Thy1 + CAFs.
  • FIG. 54 shows that phosphorylation of signaling molecules related to migration and invasion, such as Smad2/3, FAK, ERK, and Akt, and expression of MMP2/12 protein are inhibited in CD326 + tumor cells isolated from primary tumor tissues of mice administered with ApoSQ. , It is a diagram showing that the effect is reversed when LY3039478 is additionally administered.
  • A A diagram showing the results of immunoblot analysis for phosphorylation and MMP2/12 of signaling molecules related to cell migration and invasion in isolated CD326 + tumor cells.
  • (B) It is a diagram measuring the amount of phosphorylation and MMP2/12 of signaling molecules related to cell migration and invasion in isolated CD326 + tumor cells.
  • 55 is a diagram showing the experimental timeline of administering CAF CM, ApoSQ-CAF CM, ApoSQ-CAF CM and anti-WISP-1 antibody, or ApoSQ-CAF CM and IgG to mice after subcutaneous injection of 344SQ cells.
  • 57 is a result of measuring the number of tumor nodules and metastasis rate by administering CAF CM, ApoSQ-CAF CM, ApoSQ-CAF CM and anti-WISP-1 antibody or ApoSQ-CAF CM and IgG to mice, and ApoSQ-CAF CM administration
  • CAF CM ApoSQ-CAF CM
  • ApoSQ-CAF CM ApoSQ-CAF CM
  • anti-WISP-1 antibody ApoSQ-CAF CM
  • A A diagram showing the number and metastasis rate of tumor nodules that metastasized to the lungs in each group.
  • B It is a diagram showing images of tumor nodules that metastasized to the lungs in each group.
  • 58 shows that when ApoSQ-CAF CM was intratumorally administered to mice, compared to the CAF CM experimental group, cell migration and invasion were inhibited in CD326 + tumor cells and Thy1 + CAFs isolated from primary tumor tissue, but anti-WISP -1 It is a diagram showing that the effect is reversed when the antibody is additionally administered.
  • 58a is a diagram showing migration and invasion of CD326 + tumor cells isolated from primary tumor tissue.
  • 58b is a diagram showing the migration and invasion of Thy1 + CAFs isolated from primary tumor tissue.
  • CD326 + tumor cells isolated from primary tumor tissues were associated with migration and invasion, such as Smad2/3, FAK, ERK, and Akt. Phosphorylation of signal molecules and expression of MMP2/12 protein are suppressed, but the effect is reversed when anti-WISP-1 antibody is additionally administered.
  • A A diagram showing the results of immunoblot analysis for molecules involved in cell migration and invasion in CD326 + tumor cells.
  • B A diagram showing the amount of proteins involved in cell migration and invasion in isolated CD326 + tumor cells.
  • 60 shows CAF activity markers (Acta2, Col1 ⁇ 1, Itg beta 1 , Spp1 and Pdgfr ⁇ included ) and MMP (including Mmp1a, 2, and 12) mRNA expression levels are suppressed, but the effect is reversed when an anti-WISP-1 antibody is additionally administered.
  • 61 is a diagram showing an experimental timeline of administration of rWISP-1 at a concentration of 12.5 ⁇ g/kg or 25 ⁇ g/kg after subcutaneous injection of 344SQ cells into mice.
  • 62 is a diagram showing that when rWISP-1 was intratumorally administered to mice at a concentration of 12.5 ⁇ g/kg or 25 ⁇ g/kg, there was no significant difference in body weight between groups.
  • 63 shows the number of tumor nodules and the metastasis rate measured by administering rWISP-1 to mice at a concentration of 12.5 ⁇ g/kg or 25 ⁇ g/kg. It is a diagram showing a decrease in (A) A diagram showing the number and metastasis rate of tumor nodules that metastasized to the lungs in each group. (B) It is a diagram showing images of tumor nodules that metastasized to the lungs in each group.
  • FIG. 64 is a diagram showing that migration and invasion of CD326 + tumor cells and Thy1 + CAFs are inhibited in a concentration-dependent manner when rWISP-1 is intratumorally administered at a concentration of 12.5 ⁇ g/kg or 25 ⁇ g/kg to mice .
  • Figure 64a is a diagram showing the migration and invasion of CD326 + tumor cells.
  • Figure 64b is a diagram showing the migration and invasion of Thy1 + CAFs.
  • FIG. 65 shows signal molecules related to migration and invasion, such as Smad2/3, FAK, ERK, and Akt, in CD326 + tumor cells when rWISP-1 was intratumorally administered to mice at a concentration of 12.5 ⁇ g/kg or 25 ⁇ g/kg. It is a diagram showing that the phosphorylation of them and the expression of MMP2/12 protein are suppressed.
  • A A diagram showing the results of immunoblot analysis for molecules involved in cell migration and invasion in CD326 + tumor cells.
  • (B) A diagram showing the amount of proteins involved in cell migration and invasion in CD326 + tumor cells.
  • CAF activity markers including Acta2, Col1 ⁇ 1, Itg beta 1, Spp1 and Pdgfr ⁇
  • MMP including Mmp1a, 2, and 12
  • FIG. 68 is a diagram showing that, when ApoSQ-CAF CM was intratumorally administered to mice, gene expression related to cell adhesion and ECM remodeling was down-regulated in Thy1 + CAFs isolated from primary tumor tissues, compared to the CAF CM experimental group.
  • DAPT D5942 was purchased from Sigma-Aldrich (St. Louis, MO, USA), TGF-beta 1 (240-B-010) and mouse rWISP-1 (1680-WS), mouse neutralizing WISP-1 antibody (MAB1680 R&D Systems) and IgG (MAB0061) were purchased from R&D Systems (Minneapolis, MN, USA).
  • LY3039478 HY-12449 was purchased from MedChemExpress (Monmouth Junction, NJ, USA).
  • Antibodies used for Western blot, immunofluorescence staining, flow cytometry and cell sorting are as follows (see Table 1).
  • IHC 1:100 1:400 Goat IgG Alexa 488) Thermo Fisher Scientific A11055 Donkey Not applicable FACS 1:100 Goat IgG (Alexa 647) Thermo Fisher Scientific A21447 Donkey Not applicable FACS 1:100 Sheep IgG (Alexa 594) Thermo Fisher Scientific A11016 Donkey Not applicable IF, IHC 1:400 * Abbreviation: IB-Immunoblot, IHC-Immunohistochemistry, IHC-P-Immunohisrochemistry-paraffin, IF-Immunofluorescence, H-human, M-mouse, R-rat, Rb-rabbit, P-pig, Mi-mink, Hm-hamster , Mk-monkey, Dm-drosophila melanogaster, Z-zebrafish, B-Bovine, Pg-pig, Ce-caenorhabditis elegans, F-frog
  • CAFs Carcinoma-Associated Fibroblasts
  • CAFs were isolated from lung tumors of Kras-mutant (Kras LA1 ) mice using magnetic-activated cell sorting with the fibroblast-specific marker Thy1. CAFs were then supplemented with 10% fetal bovine serum (FBS), penicillin/streptomycin (100 U/100 ⁇ g, Welgene, Gyeongsan, Korea), 2 mM L-glutamine (Welgene), and 1 mM sodium pyruvate (Welgene). cultured in alpha-MEM (Welgene).
  • FBS fetal bovine serum
  • penicillin/streptomycin 100 U/100 ⁇ g, Welgene, Gyeongsan, Korea
  • Welgene 2 mM L-glutamine
  • Welgene 1 mM sodium pyruvate
  • CAFs were stably transfected with TERT plasmid (pCDH-3xFLAG-TERT, Addgene 51 plasmid # 51631) using Lipofector-EXT (AptaBio, Yongin, Korea). Primary cells used in the experiments were subcultured less than 6 times.
  • Human cancer cell lines were obtained from ATCC (American Type Culture Collection, Manassas, VA, USA). 344SQ cells (lung adenocarcinoma cell line, University of Texas MD Anderson Cancer Center, USA) and various human cancer cell lines [A549 (non-small cell lung cancer cell line), HCT116 (colorectal cancer cell line) and MCF-7 (breast cancer cell line)] were cultured in 10% FBS. and RPMI 1640 (Welgene) supplemented with penicillin/streptomycin (100 U/100 ⁇ g).
  • Cancer epithelial cell lines were exposed to 254 nm wavelength ultraviolet light for 15 minutes and then incubated with RPMI-1640 (with 10% FBS) at 37° C. and 5% CO 2 for 2 hours. As a result of evaluating nuclear morphology using a light microscope in the Wright-Giemsa-stained samples, it was confirmed that the irradiated cells were apoptotic. Lysed (necrotic) cancer cells were obtained by several freeze-thaw cycles.
  • Apoptosis and necrosis were confirmed by annexin V-FITC/propidium iodide (BD Biosciences, San Jose, CA, USA) staining followed by flow cytometry on a FACSCalibur system (ACEA Novocyte 3000, Agilent, Santa Clara, CA, USA).
  • CAFs were plated at 3 X 10 5 cells/ml and cultured in a suitable medium at 37°C and 5% CO 2 . After overnight incubation, serum was removed with X-VIVO 10 medium (04-380Q, Lonza, Basel, Switzerland) for 24 hours before cell stimulation. For stimulation, the culture medium was replaced with X-VIVO 10 containing killed or necrotic cancer cells (9 X 10 5 cells/ml). After 20 hours, the supernatant was harvested by centrifugation and used as a conditioned medium (CM) for stimulation of target cancer epithelial cells (5 X 10 5 cells/ml) or CAFs (3 X 10 5 cells/ml). For in vivo experiments, the conditioned medium was stored at -80°C until needed.
  • CM conditioned medium
  • CAFs migration and invasion in CAFs were performed 24 h after direct exposure with ApoSQ and stimulation with TGF-beta 1 (10 ng/ml).
  • TGF-beta 1 10 ng/ml
  • CAFs Prior to TGF-beta 1 stimulation, CAFs were directly exposed to apoptotic or necrotic cancer cells and then washed X-VIVO. After fixation in 4% paraformaldehyde, immobile or non-infiltrated cells on the upper surface of the membrane were scraped off with a cotton swab. Cells on the lower surface were stained with 0.1% crystal violet, washed with distilled water, and three random microscope fields were photographed (10X magnification) and counted.
  • a standardized Western blot was performed using whole cell extracts.
  • Whole cell extracts were prepared from killed cells or CAFs or target cancer cells co-cultured with conditioned media. Cells were harvested, washed with ice-cold phosphate-buffered saline (PBS), and washed in radioimmunoprecipitation assay (RIPA) buffer (10 mM Tris, pH 7.2, 150 mM NaCl, 1% Nonidet P-40, 0.5% sodium deoxygenate). cholate, 0.1% SDS, 1.0% Triton X-100, 5 mM EDTA) for 30 min on ice with protease inhibitors.
  • PBS ice-cold phosphate-buffered saline
  • cholate 0.1% SDS, 1.0% Triton X-100, 5 mM EDTA
  • Equal amounts of protein were run on SDS-PAGE gels (#161-0158, Bio-Rad Laboratories, Herc-Res, CA, USA) using a wet transfer system (Bio-Rad Laboratories) and transferred to nitrocellulose membranes (10600001, GE Healthcare Life Science). After blocking with 5% BSA-TBST or 5% milk-TBST for 1 hour, blots were incubated with primary antibody overnight, followed by incubation with secondary antibody for 1 hour at 37°C. Quantification was performed using the Odyssey image analysis system (Licor Biosciences, Lincoln, Iowa).
  • Cytokine arrays were performed using the Proteome Profiler Mouse XL Cytokine Array Kit (#ARY028, R&D Systems, USA) in CAF CM, ApoSQ-CAF CM and ApoSQ CM according to the manufacturer's instructions. Array membranes were incubated with conditioned medium overnight and membrane-bound cytokines were detected using a biotinylated detection antibody and streptavidin-horseradish peroxidase. Pixel density of cytokine spots was analyzed using Image J software (NIH, Bethesda, Maryland, USA; http://rsb). info.nih.gov/ij/).
  • formalin fixation was performed for 30 min at room temperature and IF-Wash buffer (0.05% NaN3, 0.1% BSA, 0.2% Triton X-100 and 0.05% Tween-20 in PBS) was used. .
  • IF-Wash buffer 0.05% NaN3, 0.1% BSA, 0.2% Triton X-100 and 0.05% Tween-20 in PBS
  • samples were washed three times with wash buffer for 5 minutes each and permeabilized with 0.5% Triton X-100 (Sigma-Aldrich) in PBS for 5 minutes at room temperature.
  • 5% BSA in PBS with or without mouse IgG blocking reagent was used for immunocytochemistry and immunohistochemistry, respectively.
  • target proteins were captured by each primary antibody for 18 hours at 4°C. Captured proteins were observed for 1 hour in the dark by fluorescence-coupled IgG. After staining, all slides were fixed with Vectashield Mounting medium (Vector Laboratories, Burlingame, CA, USA) containing DAPI (Vector Laboratories, Burlingame, CA, USA) and confocal microscopy (LSM5 PASCAL, Carl Zeiss, Jena, Germany). ) or observed through a fluorescence microscope (Eclipse Ti2-U, Nikon, Tokyo, Japan).
  • WISP-1 and LIF in conditioned media and serum were measured using ELISA kits (R&D Systems) according to the manufacturer's instructions.
  • serum cytokine quantification blood was collected from mice via puncture, and serum was isolated by centrifugation at 1600 ⁇ g for 5 minutes at 4° C.
  • CAFs were transfected with WISP-1 (Bioneer Inc, Daejeon, Korea), LIF (Bioneer), Notch1 (Bioneer) or BAI1 ( Dharmacon. CO, USA) was transiently transfected with either specifically targeting siRNA or control siRNA (Bioneer, Dharmacon) at a final concentration of 100 nM. After overnight transfection, cells were cultured in suitable media for 24 hours and stimulated with ApoSQ cells. 344SQ cells were transiently transfected with Dll1-specific siRNA (D-050912-01-0020, Dharmacon) or control siRNA before UV irradiation.
  • the siRNA sequences used to target the genes are as follows (see Table 3).
  • pEBB empty vector or pEBB-BAI-Flag plasmid For BAI1 overexpression, 2.0 ⁇ g of pEBB empty vector or pEBB-BAI-Flag plasmid (Gwangju Institute of Science and Technology, Gwangju, Korea) for 48 h using Lipofectamine 2000 reagent (Thermo Fisher Scientific) according to the manufacturer's instructions. CAFs were transfected. Cell lysates were measured for WISP-1 and BAI1 gene and protein expression to confirm the potency of the plasmids.
  • luciferase assay CAFs were transfected using Lipofectimin LTX Transfection Reagent and PLUS Reagent (Life Technologies, Darmstadt, Germany). Cells were transfected with 800 ng/well 4 ⁇ CSL luciferase plasmid (#41726 Addgene, Wattown, MA) to produce luciferase in response to Notch pathway activation and transfected with 200 ng/well Renilla luciferase plasmid . Luciferase activity was normalized to Renilla activity and values were reported as fold change relative to the PGL2-control vector. All conditions were performed in triplicate for each independent experiment, and luciferase experiments were performed using the Dual-Luciferase Assay System (Promega, Madison, WI, USA).
  • Conditioned medium from CAFs was incubated for 2 hours with 10 ⁇ g/ml mouse anti-mouse WISP-1 neutralizing antibody (R&D Systems) or 10 ⁇ g/ml IgG isotype control (R&D Systems).
  • the neutralizing effect of anti-WISP-1 antibodies was measured by WISP-1 ELISA.
  • UV-irradiated killed cancer cells or live cancer cells were cultured in FACS buffer (0.1% BSA and 0.1% sodium azide in PBS) with anti-Dll1, anti-Dll3, anti-Dll4, anti-Jag1 or anti-Jag2 (1 :100) for 30 minutes at room temperature. Cells were then incubated for 30 minutes at room temperature in FACS buffer with Alexa 488- or 594-labeled secondary antibodies. After incubation, the cells were washed three times with FACS buffer, and the expression level of Notch ligand was analyzed using a flow cytometer (ACEA Novocyte 3000, Agilent, Santa Clara, CA, USA). Data were analyzed using Novoexpress software (Agilent).
  • Phagocytosis analysis of killed cancer cells was performed through the flow cytometry and immunofluorescence method.
  • CAFs were stained with PKH26 (red), and then co-cultured with apoptotic 344SQ cells labeled with PKH67 (green) at a ratio of 1:3 for 24 hours. After washing, the percentage of phagocytosis by CAFs was measured by two-color flow cytometry.
  • killed 344SQ cells labeled with PKH67 were co-cultured with CAFs for 24 hours, washed, and CAFs were fixed with 3.7% w/v paraformaldehyde and 0.1% Triton X-100. Permeabilized for 15 minutes.
  • F-actin was stained with rhodamine phalloidin (Invitrogen) according to the manufacturer's instructions. Images were captured with a confocal microscope (LSM5 PASCAL; Carl Zeiss, Jena, Germany), and the phagocytic index was calculated by the formula (killed cells/200 total CAFs) x 100.
  • CAFs were pre-incubated for 2 hours with DAPT (10 or 20 ⁇ M) or transfected with Notch1 or control siRNA for 24 hours before co-culture with ApoSQ at a 1:3 ratio for 24 hours.
  • Rac1 activity was measured using the G-LISA Rac1 activity assay kit (Cytoskeleton, Denver, CO, USA) according to the manufacturer's instructions. That is, cell lysate containing the same amount of protein was added to the Rac1-GTP binding plate and incubated at 4° C. for 30 minutes. The plate was washed with PBS containing 0.05% of Tween 20 and reacted with an antigen presentation buffer. After washing, the plates were reacted with anti-Racl primary and secondary antibodies. The reaction was observed with horseradish peroxidase detection reagent and stopped with stop solution. The plate was measured for absorbance at 490 nm using a spectrophotometer.
  • the selective Notch1 inhibitor LY3039478 (8 mg/kg) was orally administered with 15% sugar gel vehicle three times a week for 6 weeks one day before ApoSQ injection.
  • conditioned media experiments 2 days after injection of 344SQ, conditioned media derived from CAFs (100 ⁇ l per mouse) with or without neutralizing anti-WISP-1 antibody (10 ⁇ g/ml) or isotype IgG was added for 1 week 3 It was administered via intratumor injection. Mice were monitored daily for tumor growth and sacrificed 6 weeks after injection.
  • a necropsy was performed to examine the diameter and weight of the subcutaneous tumor mass, the status of lung metastases (number or incidence of nodules), and histological evaluation of formalin-fixed, paraffin-embedded, immunofluorescence-stained primary tumors.
  • tumor tissue was dissociated using a tumor dissociation kit combined with a MACSTM Dissociator (all Miltenui Biotec Inc., San Diego, CA) according to the manufacturer's instructions. made it Briefly, tumor tissue from each mouse was transferred to a MACS C tube (Miltenui Biotec) containing the enzyme mixture from the kit for enzymatic digestion. After dissociation, each tissue homogenate was filtered through 70- and 40- ⁇ m sterile nylon mesh and red blood cells were lysed with red blood cell lysis buffer.
  • MACSTM Dissociator all Miltenui Biotec Inc., San Diego, CA
  • Thy1 + CAFs were sorted using CD90.2 MicroBeads and a MACS MS column (Miltenyi Biotec) according to the manufacturer's instructions.
  • CD326 + epithelial tumor cells and CD11b + tumor-associated macrophages were isolated using CD326 and CD11b MicroBeads (Miltenyi Biotec), respectively. Isolated cells were cultured in complete medium.
  • Thy1 + cells were cultured in ⁇ -MEM (Welgene) supplemented with 20% FBS and 1% penicillin/streptomycin, and CD326 + cells were cultured in RPMI 1640 (Welgene) supplemented with 10% FBS and 1% penicillin/streptomycin. and CD11b + cells were cultured in DMEM (Welgene) supplemented with 10% FBS and 1% penicillin/streptomycin. Isolated individual cell groups were validated by qRT-PCR, and individual cells were isolated within randomly selected mouse primary tumors within each group of two or three.
  • mouse tumor metastasis RT 2 Profiler TM PCR Array PAMM-028ZA-6, Qiagen, Hilden, Germany
  • mouse extracellular matrix and aggregation molecule RT 2 Profiler TM PCR Array PAMM-013ZA-6, Qiagen
  • RNA isolation, DNase treatment and RNA washing were performed according to the instructions of the manufacturer (Takara Bio). The isolated RNA was reverse transcribed into cDNA using the RT 2 First Strand kit (Qiagen).
  • PCR was performed using RT 2 SYBR Green qPCR Master Mix (Qiagen), QuantStudio 3 Real-Time PCR system and ABI PRISM7900 instrument (Applied Bio-Systems).
  • the expression level data was normalized using the average Ct value of Gapdh (glyceraldehyde 3-phosphate dehydrogenase), a housekeeping gene in the array. Each assay was performed in triplicate.
  • 344SQ cells were treated with CAFs and killed 344SQ co-culture conditioned medium (ApoSQ-CAF CM) or CAFs and necrotic 344SQ co-culture conditioned medium (NecSQ-CAF CM) in the presence or absence of TGF-beta 1 for 48 hours.
  • Transwell migration and invasion assays were performed to analyze cell migration and invasion capabilities against a chemo-attractant gradient.
  • TGF-beta 1 signal when activated, it induces migration and invasion of cancer cells by regulating Smad-dependent and -independent signaling. and suppression of TGF-beta 1 signals such as the p38 MAP kinase signaling system (FIGS. 3a and 3b). In addition, TGF-beta 1 signal induces upregulation of mRNA and protein expression of MMP-2 and MMP-12, which degrade and remodel the extracellular matrix, and it was confirmed that ApoSQ-CAF CM inhibited this (FIG. 4) .
  • the conditioned medium in which CAFs and apoptotic cancer cells were co-cultured not only suppressed the activation of TGF-beta 1 signal-induced migration and invasion-related signaling pathway molecules of cancer cells, but also related to ECM remodeling. It was verified that migration and invasion of cancer cells can be inhibited by inhibiting the expression of mRNA and protein of MMP2/12.
  • the migratory ability of CAFs is related to the activation state of CAFs, ie, activated fibroblasts migrate better than quiescent or resting fibroblasts.
  • CAFs were exposed to ApoSQ or NecSQ for 20 hours, followed by fresh medium in the presence or absence of TGF-beta 1. was replaced for 24 hours.
  • this inhibitory effect was not shown when CAFs were treated with NecSQ (FIG. 5).
  • direct exposure of CAFs to ApoSQ or NecSQ in the absence of TGF-beta 1 did not affect basal levels.
  • CAF CM ApoSQ alone medium
  • ApoSQ-CAF CM ApoSQ-CAF CM
  • WISP1-1 knockdown blocked the inhibitory effect of ApoSQ-CAF CM on TGF-beta 1-induced migration and invasion in 344SQ cells (Fig. 13a), and also blocked the direct inhibitory effect of ApoSQ on CAFs migration and invasion (Fig. 13b). In contrast, when LIF was knocked down, there was no effect of inhibiting migration and invasion in 344SQ cells or CAFs (FIGS. 14 to 15b).
  • the present inventors confirmed the effect of increasing ApoSQ-induced WISP-1 secretion by CAFs using WISP-1 overexpressing CAFs.
  • WIPS-1 expression was increased in CAFs transfected with the WISP-1 plasmid, and WISP-1 secretion was increased in CAFs transfected with or without ApoSQ (FIG. 16).
  • the TGF-beta 1-induced migration and invasion of 344SQ cells were further suppressed (Fig. 17a).
  • TGF-beta 1-induced migration and invasion of CAFs were reduced compared to those of control transfected CAFs in the absence of ApoSQ, but were more inhibited than ApoSQ treated control transfected CAFs.
  • the TGF-beta 1-induced migration and invasion inhibitory effects of WISP-1 overexpressed CAFs were reduced compared to control transfected CAFs (FIG. 17b).
  • WISP-1 acts in an autocrine/paracrine manner to induce anti-migration and anti-invasion effects
  • recombinant mouse WISP-1 (rWISP) was used at concentrations of 5 and 10 ng/ml, or 0.75-3 ng/ml. -1) was directly treated with 344SQ or CAFs, respectively, in the presence of TGF-beta 1.
  • treatment with this low dose of rWISP-1 inhibited TGF-beta 1-induced migration and invasion of 344SQ and CAFs in a concentration-dependent manner (FIGS. 19a and 19b).
  • rWISP-1 suppressed the TGF-beta 1-induced Smad or non-Smad signaling system and suppressed the expression of MMP2/MMP12 mRNA and protein in 344SQ cells or CAFs (FIGS. 20a to 21c).
  • WISP-1 is known to bind to integrin, a cell surface receptor, integrins ⁇ v, ⁇ 5, beta 1, beta Experiments were performed using blocking antibodies against 3 or beta 5.
  • WISP-1 inhibits the signaling pathway induced by TGF-beta 1 in an autocrine/paracrine manner through the integrin ⁇ v beta 3 receptor of 344SQ cells and the integrin ⁇ v beta 5 receptor of CAFs, thereby inhibiting cell migration and It was verified that the infiltration was controlled.
  • the Notch1-WISP-1 axis is known to determine the regulatory role of mesenchymal stem cell-derived stromal fibroblasts in melanoma invasion and metastasis. Therefore, the present inventors hypothesized that Notch1 signaling-dependent WISP-1 production in CAFs in response to killed lung cancer cells plays a critical role in the anti-migration and anti-invasion effects in cancer cells and CAFs.
  • Notch1 signaling pathway is activated in CAFs in response to ApoSQ.
  • Notch1 signaling proteins including Hes1 and WISP-1, known as sub-targets of the Notch pathway, as well as Notch intracellular domain 1 (NICD1), known to be secreted only when Notch1 signaling is activated, are induced by ApoSQ in CAFs It was confirmed (FIG. 25 (A)).
  • WISP-1 secretion increased in ApoSQ-CAF CM, but not in NecSQ-CAF CM (FIG. 25(B)).
  • Notch signaling is initiated by the interaction of cell-surface receptor Notch with cell-associated ligands, such as Delta-like ligands (DLL) 1,3 and 4 or Jagged-like (JAG) 1 and 2, so Notch-
  • DLL Delta-like ligands
  • JAG Jagged-like
  • BAI1 can contribute to the apoptotic cell removal process of apoptotic cervical cancer cells through primary fibroblasts
  • the thrombospondin type 1 repeats of BAI1 directly recognize phosphatidylserine (PtdSer) and target apoptotic cells
  • PtdSer phosphatidylserine
  • the percentage of PKH26-stained CAFs that phagocytosed PKH67-stained ApoSQ cells was determined using flow cytometry.
  • Annexin V was added, the phagocytosis of ApoSQ by CAFs was reduced (FIG. 36a), and ApoSQ-induced Notch1 activation and WISP-1 secretion were also downregulated (FIG. 36b).
  • BAI1 was knocked down, phagocytosis, Notch1 signaling, and WISP-1 secretion were downregulated in ApoSQ-treated CAFs (FIGS. 37a to 37c), and the same results were obtained when BAI1 was neutralized with an anti-BAI1 antibody. It was confirmed (FIG.
  • BAI1 a protein that binds to ApoSQ, promotes the removal of apoptotic cells in CAFs and regulates the production of WISP-1 in CAFs by being involved in the Notch1 signaling pathway.
  • the present inventors confirmed that a single injection of ApoSQ cells inhibited lung metastasis in syngeneic (129/Sv) immunocompetent mice. After that, the in vivo response of CAFs to ApoSQ injection was investigated. To this end, leukocytes, endothelial cells, and epithelial cells were first removed from mouse primary tumors, and then CAFs were isolated using magnetic-activated cell sorting using the fibroblast-specific marker Thy1 (FIG. 44), followed by qRT-PCR. used to analyze the amount of mRNA of CAF markers.
  • CAF activation markers such as Acta2, Col1 ⁇ 1, Fn, Itg beta 1, Spp1, Pdgfr ⁇ , Pdgfr beta, growth factors including Mmp1a, 2, 9 and 12, and Vegfa, Hgf, Cxcl12 and Cxcl14 after ApoSQ injection /
  • the mRNA expression levels of chemokines were significantly reduced after ApoSQ injection compared to the control group.
  • Notch downlink target genes including Notch1, WISP-1 (Ccn4), Hey1, Hey2, Hes1 and Hes5 were significantly improved compared to the control group, and notch ligands and other CCN gene families (Ccn1, Ccn2, The mRNA expression levels of Ccn5 and Ccn6) were almost unchanged after ApoSQ injection, but Ccn3 mRNA was approximately doubled in Thy1 + CAFs after ApoSQ injection compared to the control group (FIGS. 45a and 45b).
  • LY3039478 a Notch1 selective inhibitor, was orally administered at 8 mg/kg the day before ApoSQ injection, 3 times a week for 6 weeks (FIG. 46). . LY3039478 hardly changed body weight, tumor weight or tumor volume when compared with the control and ApoSQ groups (FIG. 47), but reversed the effect of ApoSQ administration on reducing the number of tumor nodules on the lung surface and the rate of metastasis (FIG. 48).
  • LY3039478 reversed the effect of ApoSQ administration on the decreased mRNA expression level of CAF markers, MMPs, growth factors, and chemokines in isolated Thy1 + CAFs, and also reversed the effect of increasing the mRNA expression level of Notch1 and Notch target genes. (FIG. 49).
  • LY3039478 also reversed the effect of decreasing ⁇ -SMA and increasing NICD1 and WISP-1 expression in Thy1 + CAFs (FIG. 50).
  • FIG. 51a and 51b Western e
  • CAF CM or ApoSQ-CAF CM was intratumorally injected 3 times a week for 6 weeks into syngeneic mice 2 days after 344SQ injection.
  • ApoSQ-CAF CM was pretreated with neutralizing anti-WISP-1 antibody or isotype IgG for 2 hours before injection (FIG. 55).
  • the mRNA expression levels of CAF activity markers namely Acta2, Col1 ⁇ 1, Itg beta 1, Spp1, Pdgfr ⁇ and Mmp1a, Mmp2, and Mmp12, in isolated Thy1 + CAFs were decreased by ApoSQ-CAF CM, but WISP-1 was immunoremoved.
  • the ApoSQ-CAF CM group showed no such inhibitory effect (FIG. 60).
  • rWISP1-1 was injected at a concentration of 12.5 ⁇ g/kg or 25 ⁇ g/kg 344SQ 2 days after injection to syngeneic mice 3 times a week , injected intratumorally for 6 weeks (FIG. 61)
  • CAF activity markers namely Acta2, Col1 ⁇ 1, Itg beta 1, Spp1, Pdgfr ⁇ and Mmp1a, Mmp2, and Mmp12 in Thy1 + CAFs
  • the mRNA expression level of was decreased (FIG. 66).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Virology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Developmental Biology & Embryology (AREA)
  • Epidemiology (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Food Science & Technology (AREA)
  • Nutrition Science (AREA)
  • Mycology (AREA)
  • Polymers & Plastics (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to a pharmaceutical composition for inhibiting cancer metastasis, a method for inhibiting cancer metastasis using same, and use of the pharmaceutical composition, the pharmaceutical composition containing a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured. In the present invention, exposing CAFs to apoptotic cancer cells was found to induce Notch1 signaling-dependent WISP-1 generation and thus inhibit the migration and invasion of the cancer cells and CAFs. Therefore, a culture medium, the main component of the culture medium (WISP-1), or CAF cells exposed to the apoptotic cancer cells according to the present invention can be effectively used as a cancer metastasis inhibitor.

Description

암 전이 억제용 조성물Composition for inhibiting cancer metastasis

본 발명은, 암 관련 섬유아세포(Cancer-Associated Fibroblasts, CAFs) 및 사멸화된 암 세포(Apoptotic cancer cells)를 공동 배양한 배양액을 함유하는 암 전이 억제용 약학적 조성물, 이를 이용한 암 전이 억제 방법 및 상기 약학적 조성물의 용도에 관한 것이다.The present invention provides a pharmaceutical composition for inhibiting cancer metastasis containing a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured, a method for inhibiting cancer metastasis using the same, and It relates to the use of the pharmaceutical composition.

폐암(소세포암 및 비소세포암 모두)은 남성 및 여성 모두에서 전세 계적으로 가장 흔한 암으로서, 암-관련 사망의 주요 원인이다(총 암 사망의 18.0%). 폐암 환자의 약 75%는 진단시에 국소 진행성 또는 전이성 질환이 존재하는 것으로 확인되며, 이때 전이란 암세포의 이동 및 침윤을 포함하는 다단계 과정으로, 악성 종양의 마커가 된다.Lung cancer (both small cell and non-small cell cancer) is the most common cancer worldwide in both men and women, and is the leading cause of cancer-related death (18.0% of total cancer deaths). About 75% of lung cancer patients are confirmed to have locally advanced or metastatic disease at the time of diagnosis. In this case, metastasis is a multistep process including migration and invasion of cancer cells, and becomes a marker of malignant tumors.

다양한 기원을 갖는 암 관련 섬유아세포(Cancer-associated fibroblasts, CAFs)는 종양-관련 기질 내에 존재하는 주된 세포 유형 중 하나이다. CAFs와 암 세포 사이의 측분비 교신(paracrine communication)은 악성 종양으로의 진행 및 전이 확산을 촉진할 수 있는 암 세포 이동 및 침윤과 같은 기저 과정을 촉진한다는 것이 알려져 있다. CAFs는 종양 기질에서 매트릭스를 물리적으로 리모델링하여, 암 세포가 여전히 상피 특성을 유지하면서 침윤하도록 한다. 그러나, 종양 진행에 있어서, CAFs-매개된 조절의 근본적인 분자 메카니즘은 여전히 불분명하다.Cancer-associated fibroblasts (CAFs) of various origins are one of the main cell types present in tumor-associated stroma. It is known that paracrine communication between CAFs and cancer cells promotes basal processes such as cancer cell migration and invasion that can promote progression to malignancy and metastatic spread. CAFs physically remodel the matrix in the tumor stroma, allowing cancer cells to invade while still maintaining epithelial properties. However, the underlying molecular mechanisms of CAFs-mediated regulation in tumor progression are still unclear.

종양미세환경(TME)에서 섬유아세포 활성화를 조절하는 데 있어서 Notch 신호전달(Notch signaling)의 중요성은 잘 확립되어 있다. Notch 신호전달의 활성화는 일반적으로 리간드-발현 세포와의 직접적인 상호작용에 의해 엄격하게 조절되고, Notch 신호전달의 조절에 문제가 생기면 발달 이상이나 암이 생긴다. 흥미롭게도, Notch 활성은 종양원성(oncogenic) 및 종양-억제성 기능 둘 모두와 관련되며, 이는 Notch가 유도하는 세포반응의 복잡한 미세환경에 달려있다. 활성화된 Notch 경로를 갖는 기질 섬유아세포는 부분적으로 WISP-1(Wnt-induced signaling protein-1)의 상향 조절을 통해 흑색종 성장을 약화시키고 종양 혈관신생을 억제할 수 있다. 이러한 발견은 다른 암 유형에 있어서도, CAFs에서 Notch1 의존적인 종양-조절 역할에 대한 분자 메커니즘을 밝히는데 도움을 줄 것이다.The importance of Notch signaling in regulating fibroblast activation in the tumor microenvironment (TME) is well established. Activation of Notch signaling is generally tightly regulated by direct interaction with ligand-expressing cells, and problems in the regulation of Notch signaling result in developmental abnormalities or cancer. Interestingly, Notch activity is associated with both oncogenic and tumor-suppressive functions, which depend on the complex microenvironment of cellular responses that Notch induces. Stromal fibroblasts with an activated Notch pathway can attenuate melanoma growth and inhibit tumor angiogenesis, in part through upregulation of Wnt-induced signaling protein-1 (WISP-1). These findings will help elucidate the molecular mechanisms for the Notch1-dependent tumor-regulatory role in CAFs in other cancer types as well.

종양 환경 내에서 높은 수준의 세포 사멸과 사멸화 암 세포를 제거하는 기전은 종양-특이적 면역에 크게 영향을 미칠 수 있다. 종양 미세환경(TME)에서, 식세포 매개 제거의 면역억제 효과는 항종양 면역 반응을 억제하는 것으로 보고된 바 있다. 반대로, 종양은 사멸세포 제거과정(efferocytosis)을 위한 인식을 억제함으로써 면역감시를 회피할 수 있다.High levels of apoptosis within the tumor environment and the mechanisms that clear apoptotic cancer cells can greatly influence tumor-specific immunity. In the tumor microenvironment (TME), the immunosuppressive effect of phagocyte-mediated clearance has been reported to suppress antitumor immune responses. Conversely, tumors can evade immune surveillance by inhibiting recognition for efferocytosis.

또한, 항염증 및 염증해소 촉진 지질 오타코이드(autacoid)는 다중 종양 유형에서 대식세포 탐식작용을 통해 세포 파편의 제거를 촉진시킴으로써 파편-자극된 암 진행을 특이적으로 억제한다. 더욱이, 본 발명자들의 이전 연구에서는, UV-조사된 사멸화(apoptotic) 폐암 세포에 노출된 대식세포가, 엑소좀 포스파타제 및 tensin homolog(PTEN) 및 퍼옥시좀 증식자-활성화 수용체-감마(PPARr) 리간드, 예컨대 15-하이드록시에이코사테트라엔산(HETE), 리폭신 A4 및 15d-프로스타글란딘 J2의 분비를 통해, 세포 극성 파괴, 암 세포의 EMT 및 침윤을 억제한다는 것을 입증한 바 있다(특허등록 10-1804852). 그러나, 아직까지 CAFs에 의한 암 세포의 efferocytosis 작용이 TME에서 CAFs 활성화를 조절하고 암의 진행과 전이를 차단하는지 여부는 연구된 바는 없다.In addition, the anti-inflammatory and anti-inflammatory lipid autacoids specifically inhibit debris-stimulated cancer progression by promoting the clearance of cellular debris through macrophage phagocytosis in multiple tumor types. Moreover, in previous studies by the present inventors, macrophages exposed to UV-irradiated apoptotic lung cancer cells showed upregulation of exosome phosphatase and tensin homolog (PTEN) and peroxisome proliferator-activated receptor-gamma (PPARr). It has been demonstrated that through the secretion of ligands such as 15-hydroxyeicosatetraenoic acid (HETE), lipoxin A4 and 15d-prostaglandin J2, inhibition of cell polarity disruption, EMT and invasion of cancer cells (patent registration) 10-1804852). However, it has not yet been studied whether CAFs-induced efferocytosis of cancer cells regulates CAFs activation in the TME and blocks cancer progression and metastasis.

이에, 본 발명자는 CAFs와 사멸화된(apoptotic) 암 세포의 상호작용 이 암 세포 및 CAFs의 이동 및 침윤을 어떻게 조절하는지 연구한 결과, CAFs에 사멸화된 암 세포를 노출시키면 Notch1 신호전달-의존적인 WISP-1 생성을 유도하여, 암 세포 및 CAFs 자체의 이동 및 침윤을 억제한다는 것을 확인하였다. 또한, 사멸화된 폐암 세포를 종양 내 투여한 후 CAFs 활성화 억제와 암 세포 이동 및 침윤 억제 작용을 확인하였다. 더 나아가, 사멸화된 폐암 세포와 CAFs의 공동 배양액을 종양 내 투여 후 폐 전이 억제 작용을 확인함으로써 본 발명을 완성하였다.Accordingly, the present inventors studied how the interaction between CAFs and apoptotic cancer cells regulates the migration and invasion of cancer cells and CAFs. It was confirmed that WISP-1 was induced to inhibit migration and invasion of cancer cells and CAFs themselves. In addition, after intratumoral administration of apoptotic lung cancer cells, inhibition of CAFs activation and cancer cell migration and invasion were confirmed. Furthermore, the present invention was completed by confirming the effect of inhibiting lung metastasis after intratumoral administration of the co-culture of apoptosis lung cancer cells and CAFs.

일 양상은 암 관련 섬유아세포(Cancer-Associated Fibroblasts, CAFs) 및 사멸화된 암 세포(apoptotic cancer cells)를 공동 배양한 배양액을 함유하는, 암 전이 억제용 약학적 조성물을 제공하는 것이다.One aspect is to provide a pharmaceutical composition for inhibiting cancer metastasis containing a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured.

다른 양상은 사멸화된 암 세포에 노출된 암 관련 섬유아세포를 함유하는, 암 전이 억제용 약학적 조성물을 제공하는 것이다.Another aspect is to provide a pharmaceutical composition for inhibiting cancer metastasis, containing cancer-associated fibroblasts exposed to killed cancer cells.

또 다른 양상은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 함유하는, 암 전이 억제용 건강기능식품을 제공하는 것이다.Another aspect is to provide a health functional food for inhibiting cancer metastasis containing a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.

또 다른 양상은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양하는 단계를 포함하는, 암 전이 억제용 조성물의 제조방법을 제공하는 것이다.Another aspect is to provide a method for preparing a composition for inhibiting cancer metastasis, comprising co-cultivating cancer-related fibroblasts and apoptotic cancer cells.

또 다른 양상은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 개체에 처리하는 단계를 포함하는, 암 전이 억제방법을 제공하는 것이다.Another aspect is to provide a method for inhibiting cancer metastasis, including the step of treating a subject with a culture medium in which cancer-related fibroblasts and apoptosis-killed cancer cells are co-cultured.

또 다른 양상은 WISP-1(Wnt-induced signaling protein-1)을 개체에 처리하는 단계를 포함하는, 암 전이 억제방법을 제공하는 것이다.Another aspect is to provide a method for suppressing cancer metastasis, including treating a subject with Wnt-induced signaling protein-1 (WISP-1).

또 다른 양상은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 개체에 처리하는 단계를 포함하는, 암 예방 또는 치료방법을 제공하는 것이다.Another aspect is to provide a method for preventing or treating cancer, which includes the step of treating a subject with a culture medium in which cancer-related fibroblasts and apoptosis cancer cells are co-cultured.

또 다른 양상은 WISP-1(Wnt-induced signaling protein-1)을 개체에 처리하는 단계를 포함하는, 암 예방 또는 치료방법을 제공하는 것이다.Another aspect is to provide a method for preventing or treating cancer, including treating a subject with Wnt-induced signaling protein-1 (WISP-1).

또 다른 양상은 암 전이 억제용 약제의 제조를 위한 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액의 용도를 제공하는 것이다.Another aspect is to provide the use of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured for the manufacture of a drug for inhibiting cancer metastasis.

또 다른 양상은 암 전이 억제용 약제의 제조를 위한 WISP-1(Wnt-induced signaling protein-1)의 용도를 제공하는 것이다.Another aspect is to provide use of WISP-1 (Wnt-induced signaling protein-1) for the manufacture of a drug for suppressing cancer metastasis.

또 다른 양상은 암 예방 또는 치료용 약제의 제조를 위한 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액의 용도를 제공하는 것이다.Another aspect is to provide the use of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured for the manufacture of a drug for preventing or treating cancer.

또 다른 양상은 암 예방 또는 치료용 약제의 제조를 위한 WISP-1(Wnt-induced signaling protein-1)의 용도를 제공하는 것이다.Another aspect is to provide use of Wnt-induced signaling protein-1 (WISP-1) for the manufacture of a drug for preventing or treating cancer.

또 다른 양상은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액의 암 전이 억제 용도를 제공하는 것이다.Another aspect is to provide a use for suppressing cancer metastasis of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.

또 다른 양상은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 함유하는, 암 전이 억제용 약학적 조성물의 암 전이 억제 용도를 제공하는 것이다.Another aspect is to provide a use for inhibiting cancer metastasis of a pharmaceutical composition for inhibiting cancer metastasis containing a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.

또 다른 양상은 WISP-1(Wnt-induced signaling protein-1)의 암 전이 억제 용도를 제공하는 것이다.Another aspect is to provide a use of Wnt-induced signaling protein-1 (WISP-1) to inhibit cancer metastasis.

또 다른 양상은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액의 암 예방 또는 치료 용도를 제공하는 것이다.Another aspect is to provide a use for preventing or treating cancer of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.

또 다른 양상은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 함유하는, 약학적 조성물의 암 예방 또는 치료용도를 제공하는 것이다.Another aspect is to provide a pharmaceutical composition containing a culture medium in which cancer-related fibroblasts and killed cancer cells are co-cultured for use in preventing or treating cancer.

또 다른 양상은 WISP-1(Wnt-induced signaling protein-1)의 암 예방 또는 치료 용도를 제공하는 것이다.Another aspect is to provide a use for cancer prevention or treatment of Wnt-induced signaling protein-1 (WISP-1).

본 발명은 암 관련 섬유아세포(Cancer-Associated Fibroblasts, CAFs) 및 사멸화된 암 세포(apoptotic cancer cells)를 공동 배양한 배양액을 함유하는, 암 전이 억제용 약학적 조성물을 제공한다.The present invention provides a pharmaceutical composition for inhibiting cancer metastasis containing a culture medium in which cancer-associated fibroblasts (CAFs) and apoptotic cancer cells are co-cultured.

본 발명에서 상기 CAFs와 사멸화된 암 세포를 공동 배양한 배양액은 암 세포의 전이를 억제하는 효과가 있다.In the present invention, the culture medium in which the CAFs and the apoptotic cancer cells are co-cultured has an effect of inhibiting metastasis of cancer cells.

본 발명에서 "암 관련 섬유아세포(Cancer-Associated Fibroblasts, CAFs)"란 암 병변의 내부 및/또는 주변에 존재하는 α-SMA(α-평활근엑틴) 양성 섬유아세포를 의미하며, 상기 CAFs는 대장암, 폐암, 전립선암, 유방암, 위암, 담관암, 기저세포암 등의 다양한 암에서 그 존재가 확인되고 있다.In the present invention, "Cancer-Associated Fibroblasts (CAFs)" refers to α-SMA (α-smooth muscle actin) positive fibroblasts present inside and/or around cancer lesions, and the CAFs are colorectal cancer. , its presence has been confirmed in various cancers such as lung cancer, prostate cancer, breast cancer, stomach cancer, cholangiocarcinoma, and basal cell carcinoma.

본 발명에서 상기 CAFs와 관련된 암은 뇌종양, 두경부암, 유방암, 폐암, 식도암, 위암, 십이지장암, 충수암, 대장암, 직장암, 간암, 췌장암, 담낭암, 담관암, 항문암, 신암, 수뇨관암, 방광암, 전립선 암, 음경암, 정소암, 자궁암, 난소암, 외음암, 질암, 피부암 등의 고형암일 수 있다.In the present invention, the cancers associated with the CAFs include brain tumor, head and neck cancer, breast cancer, lung cancer, esophageal cancer, stomach cancer, duodenal cancer, appendix cancer, colon cancer, rectal cancer, liver cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, anal cancer, renal cancer, ureter cancer, and bladder cancer. , prostate cancer, penile cancer, testicular cancer, uterine cancer, ovarian cancer, vulvar cancer, vaginal cancer, and solid cancer such as skin cancer.

본 발명의 일 실시예에서, 상기 CAFs는 암에 관련된 것으로서 악성고형종양과 관련된 섬유아세포일 수 있다. 일 구체예에서, 상기 CAFs는 섬유육종, 악성섬유성조직구종, 지방육종, 횡문 근육종, 평활근육종, 혈관육종, 악성피부암, 림프혈관육종, 활막육종, 연골육종, 골육종 등의 육종에 관련된 섬유아세포일 수 있으며, 폐암, 위암, 유방암, 대장암 또는 전립선암과 관련된 섬유아세포일 수 있다.In one embodiment of the present invention, the CAFs are related to cancer and may be fibroblasts related to malignant solid tumors. In one embodiment, the CAFs are fibroblasts related to sarcomas such as fibrosarcoma, malignant fibrous histiocytoma, liposarcoma, rhabdomyosarcoma, leiomyosarcoma, angiosarcoma, malignant skin cancer, lymphangiosarcoma, synovial sarcoma, chondrosarcoma, osteosarcoma, etc. It may be, and may be fibroblasts related to lung cancer, stomach cancer, breast cancer, colon cancer or prostate cancer.

본 발명의 일 실시예에서, 상기 CAFs는 Notch1 시그널링과 BAI1/Rac1 시그널링의 상호작용에 의해 탐식능이 강화될 수 있다.In one embodiment of the present invention, the CAFs can have enhanced phagocytic ability by the interaction of Notch1 signaling and BAI1/Rac1 signaling.

본 발명에서 "사멸화된 암 세포(apoptotic cancer cells)"는 암 세포에 특정 파장의 빛을 조사하여 사멸화를 유도한 것일 수 있다. 상기 특정 파장의 빛의 조사는 자외선(Ultra-violet ray, UV) 조사일 수 있다. 일 실시예에서, 상기 파장은 100 내지 400 nm 파장으로 5 내지 30분 동안 조사될 수 있다.In the present invention, “apoptotic cancer cells” may be those in which apoptosis is induced by irradiating cancer cells with light of a specific wavelength. The irradiation of light of the specific wavelength may be ultraviolet (Ultra-violet ray, UV) irradiation. In one embodiment, the wavelength may be irradiated for 5 to 30 minutes with a wavelength of 100 to 400 nm.

본 발명에서 "공동 배양"은 CAFs와 사멸화된 암 세포를 함께 배양하여 이루어질 수 있다. 일 실시예에서, CAFs를 20 내지 30 시간 동안 X-VIVO 또는 무혈청 DMEM 배지에서 사멸화된 암 세포와 함께 배양할 수 있다.In the present invention, "co-culture" can be achieved by culturing CAFs and apoptotic cancer cells together. In one example, 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 구체예에서, 상기 배양액은 조정배지(Conditioned medium)일 수 있다.In the present invention, "culture medium" means a culture product obtained through co-cultivation of 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.

본 발명의 일 실시예에서, 상기 사멸화된 암 세포의 암종은 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 선택된 하나 이상일 수 있다. 일 구체예에서 상기 암종은 폐암, 유방암, 위암, 대장암 및 전립선암으로 이루어진 군에서 선택된 하나 이상일 수 있으며, 상기 폐암은 폐선암 또는 비소세포폐암일 수 있다.In one embodiment of the present invention, the carcinoma of the killed cancer cells is 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, It may be at least one selected from the group consisting of testicular cancer, laryngeal cancer, oral cancer, head and neck cancer, thyroid cancer, liver cancer, bladder cancer, osteosarcoma, lymphoma, and leukemia. In one embodiment, the carcinoma may be at least one selected from the group consisting of lung cancer, breast cancer, stomach cancer, colon cancer, and prostate cancer, and the lung cancer may be lung adenocarcinoma or non-small cell lung cancer.

본 발명의 일 실시예에서, 상기 폐선암 세포는 344SQ 세포주, 비소세포폐암 세포는 A549 세포주, 대장암 세포는 HCT116 세포주, 유방암 세포는 MCF-7 세포주일 수 있다.In one embodiment of the present invention, the lung adenocarcinoma cells may be the 344SQ cell line, the non-small cell lung cancer cells may be the A549 cell line, the colon cancer cells may be the HCT116 cell line, and the breast cancer cells may be the MCF-7 cell line.

본 발명의 일 실시예에서, 상기 배양액은 유효성분으로서 WISP-1(Wnt-induced signaling protein-1)을 함유할 수 있다. 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.In one embodiment of the present invention, the culture medium may contain WISP-1 (Wnt-induced signaling protein-1) as an active ingredient. 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. do.

본 발명의 일 실시예에서, 상기 WISP-1은 Notch1 시그널링에 의해 생성될 수 있다. Notch1 시그널링은 Notch 수용체를 매개체로 하여 세포접촉의존적으로 이루어지고, 발생, 재생, 항상성 유지 등에 중요한 역할을 하며, 또한 Notch 시그널링의 활성은 종양원성(oncogenic) 및 종양-억제성 기능 둘 모두와 연관이 있다. Notch1-WISP-1 시그널링은 흑색종의 침윤 및 전이에서 중간엽 줄기 세포-유래 기질 섬유아세포의 조절 역할을 결정하는 것으로 알려져 있다.In one embodiment of the present invention, the WISP-1 may be generated by Notch1 signaling. Notch1 signaling is cell contact-dependent through Notch receptors and plays an important role in development, regeneration, and maintenance of homeostasis, and the activity of Notch signaling is associated with both oncogenic and tumor-suppressive functions. there is. Notch1-WISP-1 signaling is known to determine the regulatory role of mesenchymal stem cell-derived stromal fibroblasts in melanoma invasion and metastasis.

본 발명의 일 실시예에서, 상기 Notch1 시그널링은 Dll1(Delta-like ligand 1)에 의해 개시될 수 있다. Dll1은 Notch Delta ligand의 일종으로, 혈액 생성 시 세포의 운명 결정을 조절하는 역할을 하며, 세포-세포 간 커뮤니케이션에 관여하는 것으로 알려져 있다.In one embodiment of the present invention, the Notch1 signaling may be initiated by Dll1 (Delta-like ligand 1). Dll1 is a type of Notch Delta ligand that plays a role in regulating cell fate decisions during blood formation and is known to be involved in cell-cell communication.

본 발명의 일 실시예에서, 상기 Notch1 시그널링은 BAI1/Rac1(Brain angiogenesis inhibitor 1/Ras-related C3 botulinum toxin substrate 1) 시그널링에 의해 강화될 수 있다. BAI1은 응집 GPCR의 일종으로, 혈관신생을 억제하는 작용을 하며, Rac1은 GTPase의 일종으로, 세포 성장, 세포 골격 재구성, 세포 주기, 세포-세포 응집 및 이동 등을 조절하는 역할을 수행한다.In one embodiment of the present invention, the Notch1 signaling may be enhanced by BAI1/Rac1 (Brain angiogenesis inhibitor 1/Ras-related C3 botulinum toxin substrate 1) signaling. BAI1 is a type of aggregation GPCR that acts to inhibit angiogenesis, and Rac1 is a type of GTPase that plays a role in regulating cell growth, cytoskeleton reorganization, cell cycle, cell-cell aggregation and migration, and the like.

본 발명의 일 실시예에서, 상기 조성물은 Notch1 시그널링과 BAI1/Rac1 시그널링의 상호작용에 의해 암 관련 섬유아세포의 탐식능을 강화할 수 있다. In one embodiment of the present invention, the composition can enhance the phagocytic ability of cancer-related fibroblasts by the interaction of Notch1 signaling and BAI1/Rac1 signaling.

본 발명에서 "탐식(efferocytosis)"이란, 사멸화된 세포를 제거하기 위한 식세포 작용을 의미한다. 상기 탐식능은, CAFs가 탐식 작용을 통하여 사멸화된 암 세포를 제거할 수 있는 능력을 의미한다.In the present invention, "efferocytosis" means phagocytosis to remove apoptotic cells. The phagocytic ability refers to the ability of CAFs to remove apoptotic cancer cells through phagocytosis.

본 발명의 일 실시예에서, 상기 "상호작용"은 상기 Notch1 시그널링과 상기 BAI1/Rac1 시그널링과의 크로스톡(crosstalk)을 의미할 수 있고, 보다 구체적으로 긍정적 크로스톡(positive crosstalk)을 의미할 수 있다. 본 발명에서, "크로스톡(crosstalk)"이란, 신호 전달 경로의 하나 이상의 구성요소가 다른 구성요소에 영향을 미치는 현상을 의미한다. 긍정적 크로스톡은 한 시그널링이 다른 시그널링을 강화하는 현상을 의미한다. 부정적 크로스톡(negative crosstalk)은 한 시그널링이 다른 시그널링을 억제하는 현상을 의미한다.In one embodiment of the present invention, the "interaction" may mean crosstalk between the Notch1 signaling and the BAI1/Rac1 signaling, and more specifically, may mean positive crosstalk. there is. In the present invention, "crosstalk" means a phenomenon in which one or more components of a signal transduction pathway affect other components. Positive crosstalk refers to a phenomenon in which one signaling reinforces another signaling. Negative crosstalk refers to a phenomenon in which one signaling suppresses another signaling.

본 발명의 일 실시예에서, 상기 BAI1/Rac1 시그널링은 상기 암 관련 섬유아세포에 의한 사멸화된 암 세포의 탐식작용을 촉진할 수 있다.In one embodiment of the present invention, the BAI1/Rac1 signaling can promote phagocytosis of apoptotic cancer cells by the cancer-associated fibroblasts.

본 발명의 일 실시예에서, 상기 조성물은 Notch1 시그널링과 BAI1/Rac1 시그널링의 상호작용에 의해 WISP-1 생성을 강화할 수 있다.In one embodiment of the present invention, the composition can enhance WISP-1 production by the interaction of Notch1 signaling and BAI1/Rac1 signaling.

본 발명의 일 실시예에서, 상기 조성물은 암 관련 섬유아세포에서 활성화 마커를 감소시킬 수 있다. 일 구체예에서, 상기 활성화 마커는 Acta2, Col1α1, Fn, Itg beta 1, Spp1, Pdgfrα, Pdgfr beta 및 Mmp1a, 2, 9 및 12로 이루어진 군에서 선택된 하나 이상일 수 있다.In one embodiment of the present invention, the composition can reduce activation markers in cancer-associated fibroblasts. In one embodiment, the activation marker may be one or more selected from the group consisting of Acta2, Col1α1, Fn, Itg beta 1, Spp1, Pdgfrα, Pdgfr beta, and Mmp1a, 2, 9, and 12.

본 발명의 일 실시예에서, 상기 조성물은 암 관련 섬유아세포에서 성장 인자 및 케모카인을 감소시킬 수 있다. 일 구체예에서, 상기 성장 인자는 Vegfa, Hgf, Cxcl12 및 Cxcl14로 이루어진 군에서 선택되는 하나 이상일 수 있다.In one embodiment of the present invention, the composition can reduce growth factors and chemokines in cancer-associated fibroblasts. In one embodiment, the growth factor may be one or more selected from the group consisting of Vegfa, Hgf, Cxcl12 and Cxcl14.

본 발명의 일 실시예에서, 상기 조성물은 암 관련 섬유아세포에서 Notch1 관련 분자를 증가시킬 수 있다. 일 구체예에서, 상기 Notch1 관련 분자는 Notch1, WISP-1(Ccn4), Hey1, Hey2, Hes1 및 Hes5로 이루어진 군에서 선택되는 하나 이상일 수 있다.In one embodiment of the present invention, the composition can increase Notch1-related molecules in cancer-related fibroblasts. In one embodiment, the Notch1-related molecule may be one or more selected from the group consisting of Notch1, WISP-1 (Ccn4), Hey1, Hey2, Hes1, and Hes5.

본 발명에서 "전이"란 악성종양이 발병한 장기에서 떨어진 다른 조직으로 전파한 상태를 의미하며, 악성종양의 마커로 이용될 수 있다. 본 발명의 일 실시예에서, 상기 전이는 암 세포의 이동(migration) 및 침윤(invasion)을 포함할 수 있다.In the present invention, "metastasis" refers to a state in which a malignant tumor has spread to other tissues away from an organ where it has occurred, and can be used as a marker of a malignant tumor. In one embodiment of the present invention, the metastasis may include migration and invasion of cancer cells.

본 발명에서 "이동"은 세포가 특정 외부 신호에 반응하여 특정 위치로 옮겨가는 것을 의미하며, "침윤"은 세포가 근처 다른 조직으로 확장되어 관통하는 것을 의미한다.In the present invention, "migration" means that cells move to a specific location in response to a specific external signal, and "invasion" means that cells expand and penetrate other nearby tissues.

본 발명은 사멸화된 암 세포에 노출된 암 관련 섬유아세포를 함유하는, 암 전이 억제용 약학적 조성물을 제공한다.The present invention provides a pharmaceutical composition for inhibiting cancer metastasis containing cancer-related fibroblasts exposed to apoptotic cancer cells.

본 발명에서 상기 사멸화된 암 세포에 노출된 CAFs는 WISP-1을 함유하고 있어, 암 세포의 전이를 억제하는 효과가 있다.In the present invention, CAFs exposed to the killed cancer cells contain WISP-1, and thus have an effect of suppressing metastasis of cancer cells.

본 발명에서, "노출"은 사멸화된 암 세포와 CAFs를 세포 간 상호작용이 가능하도록 접촉 또는 자극하는 과정을 의미하며, 사멸화된 암 세포와 CAFs를 공동 배양한 조정 배지와는 구분된다.In the present invention, "exposure" means a process of contacting or stimulating apoptosis cancer cells and CAFs to enable cell-to-cell interaction, and is distinguished from a conditioned medium in which apoptosis cancer cells and CAFs are co-cultured.

본 발명에 따른 약학적 조성물은 "약학적으로 허용 가능한 담체"를 포함할 수 있다. 상기 약학적으로 허용 가능한 담체는 제제시에 통상적으로 이용되는 것으로서, 식염수, 멸균수, 링거액, 완충 식염수, 사이클로덱스트린, 덱스트로즈 용액, 말토덱스트린 용액, 글리세롤, 에탄올, 리포좀 등을 포함하지만 이에 한정되지 않으며, 필요에 따라 항산화제, 완충액 등 다른 통상의 첨가제를 더 포함할 수 있다. 또한 희석제, 분산제, 계면활성제, 결합제, 윤활제 등을 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형, 환약, 캡슐, 과립, 또는 정제로 제제화할 수 있다. 적합한 약학적으로 허용되는 담체 및 제제화에 관해서는 레밍턴의 문헌에 개시되어 있는 방법을 이용하여 각 성분에 따라 바람직하게 제제 화할 수 있다. 본 발명의 약학적 조성물은 제형에 특별한 제한은 없으나 주사제, 흡입제, 피부 외용제, 또는 경구 섭취제 등으로 제제화할 수 있다.The pharmaceutical composition according to the present invention may include a "pharmaceutically acceptable carrier". The pharmaceutically acceptable carrier is one commonly used in formulation and includes, but is not limited to, saline solution, sterile water, Ringer's solution, buffered saline solution, cyclodextrin, dextrose solution, maltodextrin solution, glycerol, ethanol, liposome, etc. It is not, and if necessary, other conventional additives such as antioxidants and buffers may be further included. In addition, diluents, dispersants, surfactants, binders, lubricants, etc. may be additionally added to formulate formulations for injections such as aqueous solutions, suspensions, emulsions, etc., pills, capsules, granules, or tablets. Regarding a suitable pharmaceutically acceptable carrier and formulation, it can be preferably formulated according to each component using the method disclosed in Remington's literature. The pharmaceutical composition of the present invention is not particularly limited in dosage form, but may be formulated as an injection, an inhalant, an external preparation for the skin, or an oral intake.

본 발명의 약학적 조성물은 목적하는 방법에 따라 경구 투여하거나 비경구투여(예를 들어, 정맥 내, 피하, 피부, 비강, 기도에 적용)할 수 있으며, 투여량은 환자의 상태 및 체중, 질병의 정도, 약물형태, 투여경로 및 시간에 따라 다르지만, 당업자에 의해 적절하게 선택될 수 있다.The pharmaceutical composition of the present invention may be administered orally or parenterally (for example, intravenously, subcutaneously, applied to the skin, nasal cavity, or respiratory tract) according to the desired method, and the dosage is determined according to the patient's condition and weight, disease Depending on the degree, drug form, administration route and time, it can be appropriately selected by those skilled in the art.

본 발명에 따른 조성물은 약학적으로 유효한 양으로 투여한다. 본 발명에 있어서, "약학적으로 유효한 양"은 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분한 양을 의미하며, 유효용량 수준은 환자의 질환의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 시간, 투여 경로 및 배출 비율, 치료기간, 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. 본 발명에 따른 조성물은 개별 치료제로 투여하거나 다른 치료제와 병용하여 투여될 수 있고 종래의 치료제와는 순차적 또는 동시에 투여될 수 있으며, 단일 또는 다중 투여될 수 있다. 상기한 요소들을 모두 고려하여 부작용 없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 이는 당업자에 의해 용이하게 결정될 수 있다.The composition according to the present invention is administered in a pharmaceutically effective amount. In the present invention, "pharmaceutically effective amount" means an amount sufficient to treat a disease with a reasonable benefit / risk ratio applicable to medical treatment, and the effective dose level is the type, severity, and activity of the drug of the patient's disease , sensitivity to the drug, time of administration, route of administration and excretion rate, duration of treatment, factors including drugs used concurrently, and other factors well known in the medical field. The composition according to the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, may be administered sequentially or simultaneously with conventional therapeutic agents, and may be administered single or multiple times. Considering all of the above factors, it is important to administer an amount that can obtain the maximum effect with the minimum amount without side effects, which can be easily determined by those skilled in the art.

구체적으로, 본 발명에 따른 조성물의 유효량은 환자의 나이, 성별, 체중에 따라 달라질 수 있으며, 일반적으로는 체중 1 kg 당 0.001 내지 150 mg, 바람직하게는 0.01 내지 100 mg을 매일 또는 격일 투여하거나 1일 1 내지 3회로 나누어 투여할 수 있다. 그러나 투여 경로, 비만의 중증도, 성별, 체중, 연령 등에 따라서 증감될 수 있으므로 상기 투여량이 어떠한 방법으로도 본 발명의 범위를 한정하는 것은 아니다.Specifically, the effective amount of the composition according to the present invention may vary depending on the patient's age, sex, and weight, and is generally 0.001 to 150 mg per 1 kg of body weight, preferably 0.01 to 100 mg per day or every other day, or 1 It can be administered in 1 to 3 divided doses per day. However, since it may increase or decrease depending on the route of administration, severity of obesity, gender, weight, age, etc., the dosage is not limited to the scope of the present invention in any way.

본 발명은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 함유하는, 암 전이 억제용 건강기능식품을 제공한다.The present invention provides a health functional food for inhibiting cancer metastasis, which contains a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.

본 발명의 건강기능식품은 담체, 희석제, 부형제 및 첨가제 중 하나 이상을 더 포함하여 정제, 환제, 산제, 과립제, 분말제, 캡슐제 및 액제 제형으로 이루어진 군에서 선택된 하나로 제형될 수 있다. 본 발명의 추출물을 첨가할 수 있는 식품으로는, 각종 식품류, 분말, 과립, 정제, 캡슐, 시럽제, 음료, 껌, 차, 비타민 복합제, 건강기능성 식품류 등이 있다.The health functional food of the present invention may be formulated as one selected from the group consisting of tablets, pills, powders, granules, powders, capsules and liquid formulations by further including one or more of carriers, diluents, excipients and additives. Examples of foods to which the extract of the present invention can be added include various foods, powders, granules, tablets, capsules, syrups, beverages, gum, tea, vitamin complexes, health functional foods, and the like.

상기 본 발명에 더 포함될 수 있는 첨가제로는, 천연 탄수화물, 향미제, 영양제, 비타민, 광물(전해질), 풍미제(합성 풍미제, 천연 풍미제 등), 착색제, 충진제(치즈, 초콜렛 등), 팩트산 및 그의 염, 알긴산 및 그의 염, 유기산, 보호성 콜로이드 증점제, pH조절제, 안정화제, 방부제, 산화 방지제, 글리세린, 알콜, 탄산화제 및 과육으로 이루어진 군으로부터 선택된 1종 이상의 성분을 사용할 수 있다.Additives that may be further included in the present invention include natural carbohydrates, flavors, nutrients, vitamins, minerals (electrolytes), flavors (synthetic flavors, natural flavors, etc.), colorants, fillers (cheese, chocolate, etc.), One or more components selected from the group consisting of pectic acid and its salts, alginic acid and its salts, organic acids, protective colloidal thickeners, pH adjusters, stabilizers, preservatives, antioxidants, glycerin, alcohols, carbonating agents and fruit flesh may be used. .

상술한 천연 탄수화물의 예는 모노사카라이드, 예를 들어, 포도당, 과당 등; 디사카라이드, 예를 들어 말토스, 슈크로스 등; 및 폴리사카라이드, 예를 들어 덱스트린, 시클로덱스트린 등과 같은 통상적인 당, 및 자일리톨, 소르비톨, 에리트리톨 등의 당알콜이다. 상기 향미제로서 천연 향미제(타우마틴, 스테비아 추출물(예를 들어 레바우디오시드 A, 글리시르히진등) 및 합성 향미제(사카린, 아스파르탐 등)를 유리하게 사용할 수 있다. Examples of the aforementioned natural carbohydrates include monosaccharides such as glucose, fructose, and the like; disaccharides such as maltose, sucrose and the like; and polysaccharides such as conventional sugars such as dextrins, cyclodextrins, and the like, and sugar alcohols such as xylitol, sorbitol, and erythritol. As the flavoring agent, natural flavoring agents (thaumatin, stevia extract (eg, rebaudioside A, glycyrrhizin, etc.) and synthetic flavoring agents (saccharin, aspartame, etc.) can be advantageously used.

상기 외에 본 발명의 건강기능식품은 여러 가지 영양제, 비타민, 광물(전해질), 합성 풍미제 및 천연 풍미제 등의 풍미제, 착색제 및 중진제(치즈, 초콜릿 등), 펙트산 및 그의 염, 알긴산 및 그의 염, 유기산, 보호성 콜로이드 증점제, pH 조절제, 안정화제, 방부제, 글리세린, 알콜, 탄산 음료에 사용되는 탄산화제 등을 함유할 수 있다. 그 밖에 본 발명에 따른 조성물은 천연 과일 쥬스 및 야채 음료의 제조를 위한 과육을 함유할 수 있다. 이러한 성분은 독립적으로 또는 조합하여 사용할 수 있다.In addition to the above, the health functional food of the present invention is various nutrients, vitamins, minerals (electrolytes), flavors such as synthetic flavors and natural flavors, colorants and enhancers (cheese, chocolate, etc.), pectic acid and its salts, alginic acid and salts thereof, organic acids, protective colloidal thickeners, pH adjusting agents, stabilizers, preservatives, glycerin, alcohol, carbonating agents used in carbonated beverages, and the like. In addition, the composition according to the present invention may contain fruit flesh for preparing natural fruit juice and vegetable beverages. These components may be used independently or in combination.

상기 담체, 부형제, 희석제 및 첨가제의 구체적인 예로는 이에 한정하는 것은 아니나, 락토즈, 덱스트로즈, 슈크로즈, 솔비톨, 만니톨, 에리스리톨, 전분, 아카시아 고무, 인산칼슘, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 미세결정성 셀룰로즈, 폴리비닐피롤리돈, 셀룰로즈, 폴리비닐피롤리돈, 메틸셀룰로즈, 물, 설탕시럽, 메틸셀룰로즈, 메틸 하이드록시 벤조에이트, 프로필하이드록시 벤조에이트, 활석, 스테아트산 마그네슘 및 미네랄 오일로 이루어진 그룹으로부터 선택된 1종 이상이 사용되는 것이 바람직하다.Specific examples of the carrier, excipient, diluent, and additive include, but are not limited to, lactose, dextrose, sucrose, sorbitol, mannitol, erythritol, starch, gum acacia, calcium phosphate, alginate, gelatin, calcium phosphate, calcium Silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, polyvinylpyrrolidone, methylcellulose, water, sugar syrup, methylcellulose, methyl hydroxy benzoate, propylhydroxy benzoate, talc, magnesium stearate And at least one selected from the group consisting of mineral oil is preferably used.

본 발명의 건강기능식품을 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제된다.When formulating the health functional food of the present invention, it is prepared using diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants.

본 발명은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양하는 단계를 포함하는, 암 전이 억제용 조성물의 제조방법을 제공한다.The present invention provides a method for preparing a composition for inhibiting cancer metastasis, comprising co-cultivating cancer-related fibroblasts and apoptotic cancer cells.

본 발명의 일 실시예에서, 상기 암 세포의 사멸화는 UV 조사를 통해 유도된 것일 수 있고, 상기 UV 조사는 100 내지 400 nm 파장으로 5 내지 30분 동안 수행될 수 있다. 일 구체예에서 상기 UV 조사는 150 내지 350 nm 파장으로 내지 20분 동안 또는 200 내지 300 nm 파장으로 10 내지 15분 동안 수행될 수 있다. In one embodiment of the present invention, the killing of cancer cells may be induced through UV irradiation, and the UV irradiation may be performed for 5 to 30 minutes with a wavelength of 100 to 400 nm. In one embodiment, the UV irradiation may be performed with a wavelength of 150 to 350 nm for 20 minutes or with a wavelength of 200 to 300 nm for 10 to 15 minutes.

본 발명의 일 실시예에서, 상기 배양하는 단계는 X-VIVO 배지에서 24시간 동안 배양하여 혈청결핍(serum-starved) 상태로 만드는 것 일 수 있다. 일 구체예에서, 배양 배지를 사멸화된 암 세포가 포함된 X-VIVO 또는 무혈청 DMEM 배지로 교체할 수 있다.In one embodiment of the present invention, the step of culturing may be to culture in X-VIVO medium for 24 hours to make it serum-starved. In one embodiment, the culture medium may be replaced with X-VIVO or serum-free DMEM medium containing killed cancer cells.

또한 일 구체예에서, 상기 공동 배양은 배지 교체 후 10 내지 30시간, 15 내지 25시간 또는 18내지 24시간 동안 이루어질 수 있다.Also, in one embodiment, the co-cultivation may be performed for 10 to 30 hours, 15 to 25 hours, or 18 to 24 hours after medium replacement.

본 발명은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 개체에 처리하는 단계를 포함하는, 암 전이 억제방법을 제공한다.The present invention provides a method for suppressing cancer metastasis, comprising the step of treating a subject with a culture medium in which cancer-related fibroblasts and apoptosis-killed cancer cells are co-cultured.

본 발명의 일 실시예에서, 상기 배양액을 개체에 처리하는 단계는 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 함유하는, 암 전이 억제용 약학적 조성물을 개체에 처리하여 수행되는 것일 수 있다.In one embodiment of the present invention, the step of treating the subject with the culture medium is performed by treating the subject with a pharmaceutical composition for inhibiting cancer metastasis containing a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured. it could be

본 발명에서, "처리"란 실험 요건에 따라 상기 배양액을 개체에 첨가 또는 투여하는 모든 행위를 의미할 수 있다.In the present invention, "treatment" may mean any act of adding or administering the culture medium to a subject according to experimental requirements.

본 발명에서, "개체"는 질병의 예방, 조절 또는 치료방법을 필요로 하는 대상을 의미하고, 포유동물을 포함할 수 있다. 일 구체예에서, 상기 포유동물은 인간 또는 영장류, 마우스, 소, 개, 말, 돼지 등을 포함할 수 있다.In the present invention, "subject" means a subject in need of a method for preventing, controlling or treating a disease, and may include a mammal. In one embodiment, the mammal may include humans or primates, mice, cows, dogs, horses, pigs, and the like.

본 발명은 WISP-1(Wnt-induced signaling protein-1)을 개체에 처리하는 단계를 포함하는, 암 전이 억제방법을 제공한다.The present invention provides a method for inhibiting cancer metastasis, comprising treating a subject with Wnt-induced signaling protein-1 (WISP-1).

본 발명은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 개체에 처리하는 단계를 포함하는, 암 예방 또는 치료방법을 제공한다.The present invention provides a method for preventing or treating cancer, comprising the step of treating a subject with a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.

본 발명의 일 실시예에서, 상기 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액은 이를 함유하는 약학적 조성물 형태로 개체에 처리될 수 있다.In one embodiment of the present invention, the culture medium in which the cancer-related fibroblasts and apoptosis cancer cells are co-cultured can be treated in the form of a pharmaceutical composition containing the same.

상기 용어 "예방"이란 암의 발병을 억제하거나 지연시키는 모든 행위를 의미할 수 있고, 상기 용어 "치료"란 암이 호전되거나 이롭게 변경되는 모든 행위를 의미할 수 있다.The term "prevention" may refer to any activity that inhibits or delays the onset of cancer, and the term "treatment" may mean any activity that improves or beneficially changes cancer.

본 발명은 WISP-1(Wnt-induced signaling protein-1)을 개체에 처리하는 단계를 포함하는, 암 예방 또는 치료방법을 제공한다.The present invention provides a method for preventing or treating cancer, including treating a subject with Wnt-induced signaling protein-1 (WISP-1).

본 발명은 암 전이 억제용 약제의 제조를 위한 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액의 용도를 제공한다.The present invention provides the use of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured for the manufacture of a drug for inhibiting cancer metastasis.

본 발명은 암 전이 억제용 약제의 제조를 위한 WISP-1(Wnt-induced signaling protein-1)의 용도를 제공한다.The present invention provides a use of WISP-1 (Wnt-induced signaling protein-1) for the preparation of a drug for suppressing cancer metastasis.

본 발명은 암 예방 또는 치료용 약제의 제조를 위한 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액의 용도를 제공한다.The present invention provides the use of a culture medium in which cancer-related fibroblasts and killed cancer cells are co-cultured for the manufacture of a drug for preventing or treating cancer.

본 발명은 암 예방 또는 치료용 약제의 제조를 위한 WISP-1(Wnt-induced signaling protein-1)의 용도를 제공한다.The present invention provides a use of WISP-1 (Wnt-induced signaling protein-1) for the manufacture of a drug for preventing or treating cancer.

본 발명은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액의 암 전이 억제 용도를 제공한다.The present invention provides a use for inhibiting cancer metastasis of a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.

또한, 본 발명은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 함유하는, 암 전이 억제용 약학적 조성물의 암 전이 억제 용도를 제공한다.In addition, the present invention provides a use for inhibiting cancer metastasis of a pharmaceutical composition for inhibiting cancer metastasis, which contains a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured.

본 발명은 WISP-1(Wnt-induced signaling protein-1)의 암 전이 억제 용도를 제공한다.The present invention provides a use of WISP-1 (Wnt-induced signaling protein-1) to inhibit cancer metastasis.

본 발명은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액의 암 예방 또는 치료 용도를 제공한다.The present invention provides a use for preventing or treating cancer of a culture medium in which cancer-related fibroblasts and killed cancer cells are co-cultured.

또한, 본 발명은 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 함유하는, 약학적 조성물의 암 예방 또는 치료용도를 제공한다.In addition, the present invention provides a use for preventing or treating cancer of a pharmaceutical composition containing a culture medium in which cancer-related fibroblasts and killed cancer cells are co-cultured.

본 발명은 WISP-1(Wnt-induced signaling protein-1)의 암 예방 또는 치료 용도를 제공한다.The present invention provides a use of Wnt-induced signaling protein-1 (WISP-1) for preventing or treating cancer.

본 발명에서는, CAFs와 사멸화된 암 세포의 상호작용이 암 세포 및 CAFs의 이동 및 침윤을 어떻게 조절하는지 연구한 결과, 사멸화된 암 세포와 CAF를 공동 배양한 조정 배지(ApoSQ-CAF CM)이 Notch1 신호전달-의존성 WISP-1 생성을 유도하여, 암 세포 및 CAFs 자체의 이동 및 침윤을 억제한다는 것을 확인하였다.In the present invention, as a result of studying how the interaction between CAFs and apoptotic cancer cells regulates the migration and invasion of cancer cells and CAFs, a conditioned medium in which apoptotic cancer cells and CAFs are co-cultured (ApoSQ-CAF CM) It was confirmed that this Notch1 signal transduction-dependent WISP-1 production was induced, thereby suppressing the migration and invasion of cancer cells and CAFs themselves.

또한, syngeneic 전이성 폐암 마우스 모델(in vivo)을 사용하여, 사멸화된 암 세포를 주입 시 1차 종양에서 분리된 Thy1+ CAFs의 활성화를 억제하고 Notch1 신호 활성화에 의한 WISP-1 생성의 증가 및 Thy1+ CAFs 배양액과 혈청에서의 WISP-1의 증가를 확인하였으며, 분리된 CD326+ 종양 세포와 Thy1+ CAFs의 이동 및 침윤 역시 억제됨을 확인하였다. ApoSQ-CAF CM을 주입할 경우, 폐로 전이된 노듈 수와 전이율이 억제되었으며, CD326+ 종양 세포에서 침윤 및 전이와 관련된 유전자 발현을 억제하고, Thy1+ CAFs에서 세포 부착과 세포외 기질 관련 유전자 발현을 억제한다는 것을 확인하였다.In addition, using a syngeneic metastatic lung cancer mouse model ( in vivo ), when apoptotic cancer cells are injected, activation of Thy1 + CAFs isolated from primary tumors is suppressed, and WISP-1 production is increased and Thy1 by Notch1 signal activation + It was confirmed that WISP-1 was increased in the culture medium and serum of CAFs, and the migration and invasion of isolated CD326 + tumor cells and Thy1 + CAFs were also inhibited. When injected with ApoSQ-CAF CM, the number and metastasis rate of nodules metastasized to the lung were suppressed, gene expression related to invasion and metastasis was suppressed in CD326 + tumor cells, and expression of genes related to cell adhesion and extracellular matrix in Thy1 + CAFs was confirmed to inhibit.

또한, ApoSQ-CAF CM을 주입할 경우에도 분리된 CD326+ 종양세포와 Thy1+ CAFs의 이동 및 침윤 역시 억제됨을 확인하였다.In addition, it was confirmed that the migration and invasion of isolated CD326 + tumor cells and Thy1 + CAFs were also inhibited when ApoSQ-CAF CM was injected.

따라서, 본 발명에 의할 경우 암 전이 억제 전략에 유용하게 이용될 수 있으며, 특히 본 발명의 배양액 또는 사멸화된 암 세포에 노출된 CAFs는 암 전이 억제제로서 유용하게 이용될 수 있다.Therefore, according to the present invention, it can be usefully used for cancer metastasis suppression strategies, and in particular, CAFs exposed to the culture medium or killed cancer cells of the present invention can be usefully used as cancer metastasis suppressors.

도 1은 사멸화된 암 세포와 공동 배양된 암 관련 섬유화세포(Cancer-Associated Fibroblasts, CAFs)의 배양액을 함유하는 조정배지(Conditioned medium, CM)가 암 세포의 이동 및 침윤을 억제함을 나타낸 도이다. 도 1a는 폐 선암종(adenocarcinoma) 세포주인 344SQ를 이용하여 TGF-beta 1의 존재 또는 부재하에 각각 대조군, CAF 조정배지(CAF CM), CAFs와 사멸화된 344SQ 공동 배양 조정배지(ApoSQ-CAF CM), CAFs와 괴사된 344SQ 공동 배양 조정배지(NecSQ-CAF CM) 그룹에서 344SQ 세포의 이동 및 침윤을 측정한 도이다. 도 1b는 비소세포폐암 세포주인 A549를 이용하여 TGF-beta 1의 존재 또는 부재하에 각각 대조군, CAF CM, CAFs와 사멸화된 A549 공동 배양 조정배지(ApoA-CAF CM), CAFs와 괴사된 A549 공동 배양 조정배지(NecA-CAF CM) 그룹에서 A549 세포의 이동 및 침윤을 측정한 도이다. 도 1c는 대장암 세포주인 HCT116를 이용하여 TGF-beta 1의 존재 또는 부재하에 각각 대조군, CAF CM, CAFs와 사멸화된 HCT116 공동 배양 조정배지(ApoH-CAF CM), CAFs와 괴사된 HCT116 공동 배양 조정배지(NecH-CAF CM) 그룹에서 HCT116 세포의 이동 및 침윤을 측정한 도이다. 도 1d는 유방암 세포주인 MCF-7을 이용하여 TGF-beta 1의 존재 또는 부재하에 각각 대조군, CAF CM, CAFs와 사멸화된 MCF-7 공동 배양 조정배지(ApoM-CAF CM), CAFs와 괴사된 MCF-7 공동 배양 조정배지(NecM-CAF CM) 그룹에서 MCF-7 세포의 이동 및 침윤을 측정한 도이다.1 is a diagram showing that a conditioned medium (CM) containing a culture medium of cancer-associated fibroblasts (CAFs) co-cultured with apoptotic cancer cells inhibits migration and invasion of cancer cells. am. Figure 1a shows a control, CAF conditioned medium (CAF CM), and 344SQ co-culture conditioned medium (ApoSQ-CAF CM) killed with CAFs in the presence or absence of TGF-beta 1 using 344SQ, a lung adenocarcinoma cell line. , It is a diagram measuring the migration and invasion of 344SQ cells in the 344SQ co-culture conditioned medium (NecSQ-CAF CM) group with CAFs and necrosis. Figure 1b shows the control, CAF CM, CAFs and apoptosis A549 co-culture conditioned medium (ApoA-CAF CM), CAFs and A549 co-culture in the presence or absence of TGF-beta 1 using A549, a non-small cell lung cancer cell line. It is a diagram measuring migration and invasion of A549 cells in the culture conditioned medium (NecA-CAF CM) group. 1c is a control, CAF CM, CAFs and apoptosis HCT116 co-culture conditioned medium (ApoH-CAF CM), and CAFs and necrosis HCT116 co-culture in the presence or absence of TGF-beta 1 using HCT116, a colorectal cancer cell line. It is a diagram measuring the migration and invasion of HCT116 cells in the conditioned medium (NecH-CAF CM) group. Figure 1d is a control, CAF CM, CAFs and MCF-7 co-culture conditioned medium (ApoM-CAF CM), CAFs and necrosis in the presence or absence of TGF-beta 1 using MCF-7, a breast cancer cell line, respectively. It is a diagram measuring the migration and invasion of MCF-7 cells in the MCF-7 co-culture conditioned medium (NecM-CAF CM) group.

도 2는 사멸화된 암 세포 단독 배양액과 사멸화된 암 세포의 직접 처리는 암 세포의 이동 및 침윤 억제 효과가 없음을 확인한 도이다. 도 2a는 TGF-beta 1의 존재 하에 각각 대조군, CAF CM, ApoSQ CM 및 NecSQ CM 그룹에서의 344SQ 세포 이동 및 침윤을 측정한 도이다. 도 2b는 TGF-beta 1의 존재 또는 부재하에 각각 대조군, ApoSQ 및 NecSQ 그룹에서의 344SQ 세포 이동 및 침윤을 측정한 도이다.Figure 2 is a diagram confirming that there is no effect of inhibiting migration and invasion of cancer cells by direct treatment of the killed cancer cell culture medium alone and the killed cancer cells. Figure 2a is a diagram showing the migration and invasion of 344SQ cells in the control, CAF CM, ApoSQ CM and NecSQ CM groups, respectively, in the presence of TGF-beta 1. Figure 2b is a diagram measuring 344SQ cell migration and invasion in the control, ApoSQ, and NecSQ groups, respectively, in the presence or absence of TGF-beta 1.

도 3은 면역 블랏 분석을 이용하여 ApoSQ-CAF CM이 344SQ 세포에서 TGF-beta 1과 관련된 신호전달 경로 활성을 억제함을 나타낸 도이다. 도 3a는 344SQ 세포에 TGF-beta 1와 함께 CAF CM 및 ApoSQ-CAF CM을 처리한 후 발현되는 TGF-beta 1 관련 신호전달 경로의 면역 블랏 분석 결과를 나타낸 도이다. 도 3b는 344SQ 세포에 CAF CM 및 ApoSQ-CAF CM을 처리한 후 발현되는 Smad2, Smad3, FAK, AKT, SrC, ERK 및 P38의 총양과 인산화 양을 나타낸 도이다.3 is a diagram showing that ApoSQ-CAF CM inhibits the signaling pathway activity related to TGF-beta 1 in 344SQ cells using immunoblot analysis. Figure 3a is a diagram showing the results of immunoblot analysis of TGF-beta 1-related signaling pathway expressed after treatment of CAF CM and ApoSQ-CAF CM together with TGF-beta 1 in 344SQ cells. Figure 3b is a diagram showing the total and phosphorylated amounts of Smad2, Smad3, FAK, AKT, SrC, ERK, and P38 expressed after treatment with CAF CM and ApoSQ-CAF CM in 344SQ cells.

도 4는 qRT-PCR 및 면역 블랏 분석을 이용하여 ApoSQ-CAF CM이 344SQ 세포에서 MMP2와 MMP12 mRNA 발현과 단백질 발현을 억제함을 나타낸 도이다. (A)344SQ 세포에 CAF CM 및 ApoSQ-CAF CM을 TGF-beta 와 함께 처리한 후 MMP2 및 MMP12 mRNA 발현 정도를 나타낸 도이다. (B)344SQ 세포에 CAF CM 및 ApoSQ-CAF CM을 처리한 후 발현되는 MMP2 및 MMP12의 단백질의 면역 블랏 분석 결과와 단백질 양을 나타낸 도이다.4 is a diagram showing that ApoSQ-CAF CM suppresses MMP2 and MMP12 mRNA expression and protein expression in 344SQ cells using qRT-PCR and immunoblot analysis. (A) A diagram showing the levels of MMP2 and MMP12 mRNA expression after treatment of CAF CM and ApoSQ-CAF CM with TGF-beta in 344SQ cells. (B) 344SQ cells treated with CAF CM and ApoSQ-CAF CM, and then immunoblot analysis of MMP2 and MMP12 proteins expressed and a diagram showing the protein amounts.

도 5는 CAFs를 ApoSQ 및 NecSQ에 각각 접촉시킬 경우, ApoSQ만 세포의 이동 및 침윤을 억제함을 나타낸 도이다.5 is a diagram showing that only ApoSQ inhibits cell migration and invasion when CAFs are brought into contact with ApoSQ and NecSQ, respectively.

도 6은 CAF를 TGF-beta 1의 존재 또는 부재하에 각각 ApoSQ-CAF CM, NecSQ-CAF CM, ApoA-CAF CM, NecA-CAF CM, ApoH-CAF CM 및 NecH-CAF CM에 노출한 경우, 사멸화된 암 세포와 CAF 조정배지만 세포의 이동 및 침윤을 억제함을 나타낸 도이다. 도 6a는 CAF CM, ApoSQ-CAF CM 및 NecSQ-CAF CM을 처리한 후 CAFs의 이동 및 침윤을 측정한 도이다. 도 6b는 CAF CM, ApoA-CAF CM 및 NecA-CAF CM을 처리한 후 CAFs의 이동 및 침윤을 측정한 도이다. 도 6c는 CAF CM, ApoH-CAF CM 및 NecH-CAF CM을 처리한 후 CAFs의 이동 및 침윤을 측정한 도이다. 도 6d는 CAF CM, ApoM-CAF CM 및 NecM-CAF CM을 처리한 후 CAFs의 이동 및 침윤을 측정한 도이다.Figure 6 shows that when CAFs were exposed to ApoSQ-CAF CM, NecSQ-CAF CM, ApoA-CAF CM, NecA-CAF CM, ApoH-CAF CM and NecH-CAF CM in the presence or absence of TGF-beta 1, respectively, death It is a diagram showing that only the conditioned cancer cells and CAF-conditioned medium inhibit cell migration and invasion. Figure 6a is a diagram showing migration and invasion of CAFs after treatment with CAF CM, ApoSQ-CAF CM, and NecSQ-CAF CM. Figure 6b is a diagram showing migration and invasion of CAFs after treatment with CAF CM, ApoA-CAF CM, and NecA-CAF CM. Figure 6c is a diagram showing migration and invasion of CAFs after treatment with CAF CM, ApoH-CAF CM, and NecH-CAF CM. Figure 6d is a diagram showing migration and invasion of CAFs after treatment with CAF CM, ApoM-CAF CM, and NecM-CAF CM.

도 7은 사멸화된 암 세포 단독 배양액은 CAFs의 이동 및 침윤 억제 효과가 없음을 확인한 도이다. 7 is a diagram confirming that the culture medium of killed cancer cells alone has no effect of inhibiting migration and invasion of CAFs.

도 8은 qRT-PCR, 면역 블랏 분석 및 면역형광염색을 이용하여 ApoSQ가 CAFs에서 CAF 활성화 마커의 발현을 억제함을 나타낸 도이다. 도 8a는 ApoSQ 및 NecSQ를 CAFs에 처리시 CAF 활성화 마커의 mRNA 발현 정도를 분석한 결과 및 ApoSQ 및 NecSQ를 CAFs에 처리시 CAF 활성화 마커의 면역 블랏 분석 결과 및 단백질 양을 나타낸 도이다. 도 8b는 ApoSQ 및 NecSQ를 CAFs에 처리시 α-SMA의 형광 세기를 측정한 도이다.8 is a diagram showing that ApoSQ inhibits the expression of CAF activation markers in CAFs using qRT-PCR, immunoblot analysis, and immunofluorescence staining. Figure 8a is a diagram showing the results of analyzing the mRNA expression level of CAF activation markers when ApoSQ and NecSQ were treated with CAFs, and the results of immunoblot analysis of CAF activation markers and protein amounts when ApoSQ and NecSQ were treated with CAFs. 8b is a diagram showing the fluorescence intensity of α-SMA when CAFs were treated with ApoSQ and NecSQ.

도 9는 면역 블랏 분석을 이용하여 ApoSQ가 CAFs에서 TGF-beta 1과 관련된 신호전달 경로 활성을 억제함을 나타낸 도이다. 도 9a는 CAFs에 TGF-beta 1와 함께 ApoSQ를 처리한 후 TGF-beta 1 관련 신호전달 경로의 면역 블랏 분석 결과를 나타낸 도이다. 도 9b는 CAFs에 TGF-beta 1와 함께 ApoSQ를 처리한 후 신호전달 경로, 즉 Smad2, Smad3, AKT, FAK, SrC, ERK 및 P38의 총양과 인산화 양을 나타낸 도이다. 9 is a diagram showing that ApoSQ inhibits the signaling pathway activity related to TGF-beta 1 in CAFs using immunoblot analysis. 9a is a diagram showing the results of immunoblot analysis of TGF-beta 1-related signaling pathways after CAFs were treated with ApoSQ together with TGF-beta 1; Figure 9b is a diagram showing the total and phosphorylated amounts of signaling pathways, that is, Smad2, Smad3, AKT, FAK, SrC, ERK, and P38, after CAFs were treated with ApoSQ along with TGF-beta 1.

도 10은 qRT-PCR 및 면역 블랏 분석을 이용하여 ApoSQ가 CAFs에서 MMP mRNA 및 단백질의 발현을 억제함을 나타낸 도이다. (A)CAFs에 ApoSQ를 처리한 후 MMP2 및 MMP12의 mRNA 발현 정도를 나타낸 도이다. (B)CAFs에 ApoSQ를 처리한 후 MMP2 및 MMP12의 단백질의 면역 블랏 분석 결과 및 단백질 양을 나타낸 도이다.10 is a diagram showing that ApoSQ inhibits the expression of MMP mRNA and protein in CAFs using qRT-PCR and immunoblot analysis. (A) A diagram showing the mRNA expression levels of MMP2 and MMP12 after ApoSQ was treated in CAFs. (B) A diagram showing the results of immunoblot analysis and protein amounts of MMP2 and MMP12 proteins after CAFs were treated with ApoSQ.

도 11은 CAF CM, ApoSQ-CAF CM 및 ApoSQ CM에서의 사이토카인 어레이 분석 결과 111 종류의 사이토카인 중 leukemia inhibitory factor (LIF) 와 WISP-1의 발현이 CAF CM과 ApoSQ-CAF CM 군과 비교 시 ApoSQ-CAF CM에서 가장 높게 증가한 사이토카인임을 확인한 도이다.11 is a result of cytokine array analysis in CAF CM, ApoSQ-CAF CM and ApoSQ CM, and the expression of leukemia inhibitory factor (LIF) and WISP-1 among 111 types of cytokines was compared with CAF CM and ApoSQ-CAF CM groups. It is a diagram confirming that the cytokine increased the most in ApoSQ-CAF CM.

도 12는 WISP-1을 넉다운시킬 경우, WISP-1의 발현양이 감소함을 나타낸 도이다. (A)CAFs를 WISP-1에 특이적인 siRNA로 형질감염 시킨 후 측정한 면역 블랏 분석 결과 및 WISP-1 단백질 발현 정도를 나타낸 도이다. (B)CAFs를 WISP-1에 특이적인 siRNA로 형질감염 시킨 후 배양액에서 ELISA를 이용하여 측정한 WISP-1의 양을 나타낸 도이다.12 is a diagram showing that the expression level of WISP-1 decreases when WISP-1 is knocked down. (A) A diagram showing the results of immunoblot analysis and the level of WISP-1 protein expression measured after transfecting CAFs with WISP-1-specific siRNA. (B) It is a diagram showing the amount of WISP-1 measured by ELISA in the culture medium after CAFs were transfected with siRNA specific to WISP-1.

도 13은 CAFs를 WISP-1 siRNA로 형질감염 시킬 경우 세포의 이동 및 침윤 억제 효과가 역전됨을 나타낸 도이다. 도 13a는 WISP-1 siRNA의 존재 또는 부재하에 CAF CM 및 ApoSQ-CAF CM 처리 시 344SQ 세포의 이동 및 침윤에 대해 나타낸 도이다. 도 13b는 WISP-1 siRNA의 존재 또는 부재하에 ApoSQ 처리 시 CAFs의 이동 및 침윤에 대해 나타낸 도이다.13 is a diagram showing that the effect of inhibiting cell migration and invasion is reversed when CAFs are transfected with WISP-1 siRNA. Figure 13a is a diagram showing migration and invasion of 344SQ cells upon treatment with CAF CM and ApoSQ-CAF CM in the presence or absence of WISP-1 siRNA. Figure 13b is a diagram showing migration and invasion of CAFs upon ApoSQ treatment in the presence or absence of WISP-1 siRNA.

도 14는 LIF를 넉다운시킬 경우, LIF의 발현양이 감소함을 나타낸 도이다. (A)CAFs를 LIF에 특이적인 siRNA로 형질감염 시킨 후 측정한 면역 블랏 분석 결과 및 LIF 단백질 양을 나타낸 도이다. (B)CAFs를 LIF에 특이적인 siRNA로 형질감염 시킨 후 CAF CM과 ApoSQ-CAF CM에서 ELISA를 이용하여 측정한 LIF의 양을 나타낸 도이다. (C)CAFs를 LIF에 특이적인 siRNA로 형질감염 시킨 후 CAF CM과 ApoSQ-CAF CM에서 ELISA를 이용하여 측정한 WISP-1의 양을 나타낸 도이다.14 is a diagram showing that the expression level of LIF decreases when LIF is knocked down. (A) A diagram showing the result of immunoblot analysis and the amount of LIF protein measured after transfection of CAFs with LIF-specific siRNA. (B) A diagram showing the amount of LIF measured by ELISA in CAF CM and ApoSQ-CAF CM after CAFs were transfected with LIF-specific siRNA. (C) A diagram showing the amount of WISP-1 measured by ELISA in CAF CM and ApoSQ-CAF CM after CAFs were transfected with LIF-specific siRNA.

도 15는 CAFs를 LIF siRNA로 형질감염 시킬 경우 세포의 이동 및 침윤 억제 효과가 유지됨을 나타낸 도이다. 도 15a는 LIF siRNA의 존재 또는 부재하에 CAF CM 및 ApoSQ-CAF CM 처리 시 344SQ 세포의 이동 및 침윤에 대해 나타낸 도이다. 도 15b는 LIF siRNA의 존재 또는 부재하에 ApoSQ 처리 시 CAFs의 이동 및 침윤에 대해 나타낸 도이다.15 is a diagram showing that the effect of inhibiting cell migration and invasion is maintained when CAFs are transfected with LIF siRNA. Figure 15a is a diagram showing migration and invasion of 344SQ cells when treated with CAF CM and ApoSQ-CAF CM in the presence or absence of LIF siRNA. Figure 15b is a diagram showing migration and invasion of CAFs upon ApoSQ treatment in the presence or absence of LIF siRNA.

도 16은 WISP-1을 과발현 시킬 경우, WISP-1의 발현양이 증가함을 나타낸 도이다. (A)CAFs를 모의 벡터와 WISP-1로 형질감염 시킨 후 측정한 면역 블랏 분석 결과 및 딘백질 발현 정도를 나타낸 도이다. (B)CAFs를 모의 벡터와 WISP-1로 형질감염 시킨 후 CAF CM과 ApoSQ-CAF CM에서 ELISA를 이용하여 측정한 WISP-1의 양을 나타낸 도이다.16 is a diagram showing that the expression level of WISP-1 increases when WISP-1 is overexpressed. (A) A diagram showing the results of immunoblot analysis and protein expression level measured after transfection of CAFs with mock vector and WISP-1. (B) A diagram showing the amount of WISP-1 measured by ELISA in CAF CM and ApoSQ-CAF CM after CAFs were transfected with mock vector and WISP-1.

도 17은 CAFs를 WISP-1을 과발현하도록 WISP-1로 형질감염 시킬 경우 세포의 이동 및 침윤 억제 효과가 증가함을 나타낸 도이다. 도 17a는 WISP-1 형질감염 여부에 따라 CAF CM 및 ApoSQ-CAF CM 처리 시 344SQ 세포의 이동 및 침윤에 대해 나타낸 도이다. 도 17b는 WISP-1 형질감염 여부에 따라 ApoSQ 처리 시 CAFs의 이동 및 침윤에 대해 나타낸 도이다.17 is a diagram showing that cell migration and invasion inhibitory effects are increased when CAFs are transfected with WISP-1 to overexpress WISP-1. 17a is a diagram showing migration and invasion of 344SQ cells when treated with CAF CM and ApoSQ-CAF CM, depending on whether or not WISP-1 was transfected. 17b is a diagram showing migration and invasion of CAFs upon ApoSQ treatment, depending on whether or not WISP-1 was transfected.

도 18은 ApoSQ-CAF CM에 항-WISP-1 항체를 처리할 경우 344SQ 세포의 이동 및 침윤 억제 효과가 역전됨을 나타낸 도이다.18 is a diagram showing that the anti-WISP-1 antibody treatment of ApoSQ-CAF CM reverses the effect of inhibiting migration and invasion of 344SQ cells.

도 19는 재조합 WISP-1을 첨가할 경우, 농도 의존적으로 344SQ 세포 또는 CAFs의 이동 및 침윤이 억제됨을 나타낸 도이다. 도 19a는 rWISP-1 농도에 따른 344SQ 세포의 이동 및 침윤에 대해 나타낸 도이다. 도 19b는 rWISP-1 농도에 따른 CAFs의 이동 및 침윤에 대해 나타낸 도이다.19 is a diagram showing that migration and invasion of 344SQ cells or CAFs are inhibited in a concentration-dependent manner when recombinant WISP-1 is added. Figure 19a is a diagram showing migration and invasion of 344SQ cells according to rWISP-1 concentration. Figure 19b is a diagram showing migration and invasion of CAFs according to rWISP-1 concentration.

도 20은 면역 블랏 분석을 이용하여 rWISP-1이 농도 의존적으로 344SQ 세포에서 TGF-beta 1과 관련된 신호전달경로의 활성과 MMP 발현을 억제함을 나타낸 도이다. 도 20a는 rWISP-1의 농도별로 TGF-beta 1 관련 신호 분자의 면역 블랏 분석 결과를 나타낸 도이다. 도 20b는 rWISP-1을 농도별로 처리시 TGF-beta 1 관련 신호전달경로 즉, Smad2, Smad3, FAK, AKT, SrC, ERK 및 P38 총양과 인산화 양을 나타낸 도이다. 도 20c에서 (A)는 rWISP-1의 농도별로 발현되는 MMP2 및 MMP12의 mRNA 양을 나타낸 도이다. (B)는 rWISP-1의 농도별로 발현되는 MMP2 및 MMP12 단백질의 면역 블랏 분석 결과 및 단백질양을 나타낸 도이다.20 is a diagram showing that rWISP-1 inhibits the activity of the signaling pathway related to TGF-beta 1 and MMP expression in 344SQ cells in a concentration-dependent manner using immunoblot analysis. 20a is a diagram showing the results of immunoblot analysis of TGF-beta 1-related signaling molecules for each concentration of rWISP-1. Figure 20b is a diagram showing the total amount and phosphorylation amount of TGF-beta 1-related signaling pathways, that is, Smad2, Smad3, FAK, AKT, SrC, ERK, and P38, when rWISP-1 was treated by concentration. In Figure 20c (A) is a diagram showing the amount of MMP2 and MMP12 mRNA expressed by rWISP-1 concentration. (B) is a diagram showing the results of immunoblot analysis and protein amounts of MMP2 and MMP12 proteins expressed by rWISP-1 concentrations.

도 21은 면역 블랏 분석 및 qRT-PCR을 이용하여 rWISP-1을 TGF-beta 1와 함께 처리시 CAFs에서 TGF-beta 1과 관련된 신호전달경로의 활성과 MMP 발현을 억제함을 나타낸 도이다. 도 21a는 rWISP-1 처리시 TGF-beta 1 관련 신호전달 경로의 면역 블랏 분석 결과를 나타낸 도이다. 도 21b는 rWISP-1 처리시 TGF-beta 1 관련된 신호전달경로, 즉, Smad2, Smad3, FAK, AKT, SrC, ERK 및 P38의 총양 및 인산화 양을 나타낸 도이다. 도 21c에서 (A)는 rWISP-1과 TGF-beta 1 함께 처리시 MMP2 및 MMP12의 mRNA 발현 양의 변화를 나타낸 도이고, (B)는 rWISP-1과 TGF-beta 1 함께 처리시MMP2 및 MMP12 단백질의 면역 블랏 분석 결과와 단백질양을 나타낸 도이다.21 is a diagram showing that when rWISP-1 is treated with TGF-beta 1, the activity of the signaling pathway related to TGF-beta 1 and the expression of MMP are inhibited in CAFs using immunoblot analysis and qRT-PCR. 21a is a diagram showing the results of immunoblot analysis of the TGF-beta 1-related signaling pathway upon treatment with rWISP-1. Figure 21b is a diagram showing the total and phosphorylated amounts of TGF-beta 1-related signaling pathways, that is, Smad2, Smad3, FAK, AKT, SrC, ERK and P38 upon rWISP-1 treatment. In Figure 21c, (A) is a diagram showing the change in the mRNA expression amount of MMP2 and MMP12 when rWISP-1 and TGF-beta 1 are treated together, (B) is a diagram showing the change in the mRNA expression amount of rWISP-1 and TGF-beta 1 when treated together with MMP2 and MMP12 It is a diagram showing the results of protein immunoblot analysis and the amount of protein.

도 22는 특정 항-인테그린 첨가 시 344SQ 세포 또는 CAFs에서 WISP-1 첨가에 따른 이동 및 침윤 억제 현상이 역전됨을 나타낸 도이다. 도 22a는 항-인테그린 αv 또는 beta 3 항체 첨가 시 344SQ 세포에서 WISP-1 첨가에 따른 이동 및 침윤 억제 현상이 역전됨을 나타낸 도이다. 도 22b는 항-인테그린 αv 또는 beta 5 항체 첨가 시 CAFs에서 WISP-1 첨가에 따른 이동 및 침윤 억제 현상이 역전됨을 나타낸 도이다.22 is a diagram showing that the inhibition of migration and invasion according to the addition of WISP-1 is reversed in 344SQ cells or CAFs when a specific anti-integrin is added. 22a is a diagram showing that the inhibition of migration and invasion according to the addition of WISP-1 is reversed in 344SQ cells when an anti-integrin αv or beta 3 antibody is added. 22b is a diagram showing that the inhibition of migration and invasion according to the addition of WISP-1 is reversed in CAFs when an anti-integrin αv or beta 5 antibody is added.

도 23은 면역 블랏 분석 및 qRT-PCR을 이용하여 특정 항-인테그린 첨가 시 344SQ 세포에서 WISP-1 첨가에 따른 TGF-beta 1과 관련된 신호전달경로 활성 및 MMP 발현 억제 현상이 역전됨을 나타낸 도이다. 도 23a는 rWISP-1 및 항-인테그린 αv 또는 beta 3 항체 첨가에 따른 TGF-beta 1 관련 신호 분자의 면역 블랏 분석 결과를 나타낸 도이다. 도 23b는 rWISP-1 및 항-인테그린 αv 또는 beta 3 항체 첨가에 따른 MMP2 및 MMP12의 mRNA 양을 나타낸 도이다. 도 23c는 WISP-1 및 항-인테그린 αv 또는 beta 3 항체 첨가에 따른 MMP2 및 MMP12 단백질의 면역 블랏 분석 결과를 나타낸 도이다.23 is a diagram showing that the activation of the signaling pathway related to TGF-beta 1 and the suppression of MMP expression are reversed in 344SQ cells when a specific anti-integrin is added using immunoblot analysis and qRT-PCR. 23a is a diagram showing the results of immunoblot analysis of TGF-beta 1-related signaling molecules according to the addition of rWISP-1 and anti-integrin αv or beta 3 antibodies. 23b is a diagram showing the mRNA amounts of MMP2 and MMP12 according to the addition of rWISP-1 and anti-integrin αv or beta 3 antibodies. 23c is a diagram showing the results of immunoblot analysis of MMP2 and MMP12 proteins according to the addition of WISP-1 and anti-integrin αv or beta 3 antibodies.

도 24는 면역 블랏 분석 및 qRT-PCR을 이용하여 특정 항-인테그린 첨가 시 CAFs에서 WISP-1 첨가에 따른 TGF-beta 1과 관련된 신호전달경로 활성 및 MMP 발현 억제 현상이 역전됨을 나타낸 도이다. 도 24a는 rWISP-1 및 항-인테그린 αv 또는 beta 5 항체 첨가에 따른 TGF-beta 1 관련 신호 분자의 면역 블랏 분석 결과를 나타낸 도이다. 도 24b는 rWISP-1 및 항-인테그린 αv 또는 beta 5 항체 첨가에 따른 MMP2 및 MMP12의 mRNA 양을 나타낸 도이다. 도 24c는 WISP-1 및 항-인테그린 αv 또는 beta 5 항체 첨가에 따른 MMP2 및 MMP12 단백질의 면역 블랏 분석 결과를 나타낸 도이다.FIG. 24 is a diagram showing that when a specific anti-integrin is added using immunoblot analysis and qRT-PCR, the TGF-beta 1-related signaling pathway activity and inhibition of MMP expression are reversed by the addition of WISP-1 in CAFs. 24a is a diagram showing the results of immunoblot analysis of TGF-beta 1 related signaling molecules according to the addition of rWISP-1 and anti-integrin αv or beta 5 antibodies. 24b is a diagram showing the mRNA amounts of MMP2 and MMP12 according to the addition of rWISP-1 and anti-integrin αv or beta 5 antibodies. 24c is a diagram showing the results of immunoblot analysis of MMP2 and MMP12 proteins according to the addition of WISP-1 and anti-integrin αv or beta 5 antibodies.

도 25는 CAFs에서 ApoSQ의 유무에 따라 발현되는 Notch1 신호전달 경로 관련 인자들의 단백질의 양이 증가됨을 나타낸 도이다. (A)CAFs에서 ApoSQ 유무에 따른 단백질 면역 블랏 분석 결과를 나타낸 도이다. (B)CAF CM, ApoSQ-CAF CM 및 NecSQ-CAF CM에서 발현되는 WISP-1의 양을 나타낸 도이다.25 is a diagram showing that the amount of proteins of factors related to the Notch1 signaling pathway expressed in CAFs according to the presence or absence of ApoSQ is increased. (A) A diagram showing the results of protein immunoblot analysis according to the presence or absence of ApoSQ in CAFs. (B) A diagram showing the amount of WISP-1 expressed in CAF CM, ApoSQ-CAF CM and NecSQ-CAF CM.

도 26은 CAFs에서 ApoSQ 또는 NecSQ 처리시 Notch 신호경로의 타겟 유전자인 Hes1 및 WISP-1의 mRNA 발현 양과 4×CSL luciferase의 활성이 증가됨을 나타낸 도이다. (A)qRT-PCR로 분석한 Hes1 및 WISP-1의 mRNA 발현 수준을 나타낸 도이다. (B)ApoSQ 처리시 전반적인 Notch 신호 전사 effector인 4×CSL luciferase의 활성을 나타낸 도이다.26 is a diagram showing that the mRNA expression levels of Hes1 and WISP-1, which are target genes of the Notch signaling pathway, and the activity of 4×CSL luciferase are increased when ApoSQ or NecSQ is treated in CAFs. (A) A diagram showing the mRNA expression levels of Hes1 and WISP-1 analyzed by qRT-PCR. (B) A diagram showing the activity of 4×CSL luciferase, an overall Notch signal transcriptional effector, upon ApoSQ treatment.

도 27은 CAFs에서 ApoSQ 또는 NecSQ 처리에 따라 NICD1 및 WISP-1에 대한 면역형광염색이 달라짐을 나타낸 도이다.27 is a diagram showing changes in immunofluorescence staining for NICD1 and WISP-1 according to ApoSQ or NecSQ treatment in CAFs.

도 28은 siRNA를 통해 Notch1을 넉다운시킬 경우, WISP-1 단백질 발현 등이 억제됨을 나타낸 도이다. 도 28a는 CAFs를 Notch1 siRNA로 형질감염 시킨 후 측정한 면역 블랏 분석 결과를 나타낸 도이다. 도 28b에서 (A)는 CAFs를 Notch1 siRNA로 형질감염 시킨 후 ApoSQ 또는 NecSQ의 유무에 따라 측정한 NICD1, Hes1 및 WISP-1에 대한 면역 블랏 분석 결과 및 단백질의 양을 나타낸 도이고, (B)는 CAFs를 Notch1 siRNA로 형질감염 시킨 후 CAF CM과 ApoSQ-CAF CM에서 WISP-1의 ELISA 결과를 나타낸 도이다.28 is a diagram showing that WISP-1 protein expression is suppressed when Notch1 is knocked down through siRNA. 28a is a diagram showing the results of immunoblot analysis measured after CAFs were transfected with Notch1 siRNA. 28B, (A) is a diagram showing the immunoblot analysis results and protein amounts for NICD1, Hes1, and WISP-1 measured according to the presence or absence of ApoSQ or NecSQ after CAFs were transfected with Notch1 siRNA, (B) is a diagram showing the ELISA results of WISP-1 in CAF CM and ApoSQ-CAF CM after transfection of CAFs with Notch1 siRNA.

도 29는 siRNA를 통해 Notch1을 넉다운시킬 경우, ApoSQ-CAF CM 및 ApoSQ의 항-이동 및 항-침윤 효과가 344SQ 세포 및 CAFs에서 각각 역전됨을 나타낸 도이다. 도 29a는 Notch1 siRNA의 존재 또는 부재하에 CAF CM 및 ApoSQ-CAF CM 처리 시 344SQ 세포의 이동 및 침윤에 대해 나타낸 도이다. 도 29b는 Notch1 siRNA의 존재 또는 부재하에 ApoSQ 처리 시 CAFs의 이동 및 침윤에 대해 나타낸 도이다.29 is a diagram showing that when Notch1 is knocked down through siRNA, the anti-migration and anti-invasion effects of ApoSQ-CAF CM and ApoSQ are reversed in 344SQ cells and CAFs, respectively. Figure 29a is a diagram showing migration and invasion of 344SQ cells when treated with CAF CM and ApoSQ-CAF CM in the presence or absence of Notch1 siRNA. Figure 29b is a diagram showing migration and invasion of CAFs upon ApoSQ treatment in the presence or absence of Notch1 siRNA.

도 30은 DAPT를 통해 Notch1 활성을 억제할 경우, Notch1 신호전달 경로 관련 단백질 발현 등이 억제됨을 나타낸 도이다. (A)CAFs를 10 μM DAPT로 처리한 후 ApoSQ 또는 NecSQ의 유무에 따라 측정한 NICD1, Hes1 및 WISP-1에 대한 면역 블랏 분석 결과 및 단백질의 발현 양을 나타낸 도이다. (B)CAFs를 10 μM DAPT로 처리한 후 CAF CM, ApoSQ-CAF CM 및 NecSQ-CAF CM에서 측정한 WISP-1의 ELISA 결과를 나타낸 도이다.30 is a diagram showing that when Notch1 activity is inhibited through DAPT, the expression of proteins related to the Notch1 signaling pathway is inhibited. (A) A diagram showing the immunoblot analysis results and protein expression levels for NICD1, Hes1, and WISP-1 measured in the presence or absence of ApoSQ or NecSQ after CAFs were treated with 10 μM DAPT. (B) A diagram showing the ELISA results of WISP-1 measured in CAF CM, ApoSQ-CAF CM, and NecSQ-CAF CM after CAFs were treated with 10 μM DAPT.

도 31은 DAPT를 통해 Notch1을 억제할 경우, ApoSQ-CAF CM 및 ApoSQ의 항-이동 및 항-침윤 효과가 344SQ 세포와 CAFs에서 각각 역전됨을 나타낸 도이다. 도 31a는 DAPT의 존재 또는 부재하에 CAF CM 및 ApoSQ-CAF CM 처리 시 344SQ 세포의 이동 및 침윤에 대해 나타낸 도이다. 도 31b는 DAPT의 존재 또는 부재하에 ApoSQ 처리 시 CAFs의 이동 및 침윤에 대해 나타낸 도이다.31 is a diagram showing that when Notch1 is inhibited through DAPT, the anti-migration and anti-invasion effects of ApoSQ-CAF CM and ApoSQ are reversed in 344SQ cells and CAFs, respectively. Figure 31a is a diagram showing migration and invasion of 344SQ cells upon treatment with CAF CM and ApoSQ-CAF CM in the presence or absence of DAPT. Figure 31b is a diagram showing migration and invasion of CAFs upon ApoSQ treatment in the presence or absence of DAPT.

도 32는 사멸화된 암 세포 344SQ, A549 및 HCT116에서 Notch 리간드인 Delta-like ligand(DLL) 1, Dll3, Dll4, Jagged-like(JAG) 1 및 JAG2의 발현양을 유세포 분석으로 확인한 결과, Dll1만 사멸화된 암 세포에서 발현이 증가됨을 나타낸 도이다.32 is a result of confirming the expression levels of Delta-like ligand (DLL) 1, Dll3, Dll4, Jagged-like (JAG) 1 and JAG2, which are Notch ligands, in apoptotic cancer cells 344SQ, A549 and HCT116 by flow cytometry, Dll1 It is a diagram showing that expression is increased only in cancer cells that have been killed.

도 33은 사멸화된 암 세포 344SQ, A549 및 HCT116에서 Dll1의 발현만이 증가함을 나타낸 도이다. (A)사멸화된 암 세포 344SQ, A549 및 HCT116에서 Dll1, Dll3, Dll4, JAG1 및 JAG2에 대한 면역 블랏 분석 결과를 나타낸 도이다. (B)사멸화된 암 세포 344SQ, A549 및 HCT116에서 Dll1, Dll3, Dll4, JAG1 및 JAG2 단백질의 양을 측정한 도이다.33 is a diagram showing that only Dll1 expression is increased in killed cancer cells 344SQ, A549, and HCT116. (A) A diagram showing the results of immunoblot analysis for Dll1, Dll3, Dll4, JAG1 and JAG2 in apoptotic cancer cells 344SQ, A549 and HCT116. (B) A diagram showing the amount of Dll1, Dll3, Dll4, JAG1 and JAG2 proteins in the killed cancer cells 344SQ, A549 and HCT116.

도 34는 항-Dll1 항체로 Dll1을 중화시킬 경우, ApoSQ 처리 후 CAFs에서 Notch1 신호전달 경로 관련 단백질 발현이 억제됨을 나타낸 도이다. (A)ApoSQ와 접촉시킨 CAFs에서 항-Dll1 항체의 존재 또는 부재하에 측정한 NICD1, Hes1 및 WISP-1에 대한 면역 블랏 분석 결과와 단백질의 양을 나타낸 도이다. (B)ApoSQ와 접촉시킨 CAFs에서 항-Dll1 항체의 존재 또는 부재하에 측정한 WISP-1의 ELISA 결과를 나타낸 도이다.34 is a diagram showing that expression of proteins related to the Notch1 signaling pathway is suppressed in CAFs after ApoSQ treatment when Dll1 is neutralized with an anti-Dll1 antibody. (A) A diagram showing the immunoblot analysis results and protein amounts for NICD1, Hes1, and WISP-1 measured in the presence or absence of anti-Dll1 antibody in CAFs contacted with ApoSQ. (B) A diagram showing the ELISA results of WISP-1 measured in the presence or absence of anti-Dll1 antibody in CAFs contacted with ApoSQ.

도 35는 siRNA를 통해 Dll1을 넉다운시킬 경우, CAFs에서 Notch1 신호전달 경로관련 단백질 발현이 억제됨을 나타낸 도이다. 도 35a는 344SQ 세포를 Dll1 siRNA로 형질감염 시킨 후 측정한 면역 블랏 분석 결과 및 Dll1 단백질의 양을 나타낸 도이다. 도 35b에서 (A)는 344SQ 세포를 Dll1 siRNA로 형질감염 시킨 후 CAFs에서 ApoSQ의 유무에 따라 측정한 NICD1, Hes1 및 WISP-1에 대한 면역 블랏 분석 결과와 단백질의 양을 나타낸 도이고, (B)는 344SQ 세포를 Dll1 siRNA로 형질감염 시킨 후 CAFs에서 ApoSQ의 유무에 따라 측정한 WISP-1의 ELISA 결과를 나타낸 도이다.35 is a diagram showing that expression of proteins related to the Notch1 signaling pathway is suppressed in CAFs when Dll1 is knocked down through siRNA. 35A is a diagram showing the results of immunoblot analysis and the amount of Dll1 protein measured after transfection of 344SQ cells with Dll1 siRNA. In Figure 35b (A) is a diagram showing the immunoblot analysis results and protein amounts for NICD1, Hes1, and WISP-1 measured according to the presence or absence of ApoSQ in CAFs after transfection of 344SQ cells with Dll1 siRNA, (B ) is a diagram showing the ELISA results of WISP-1 measured according to the presence or absence of ApoSQ in CAFs after transfecting 344SQ cells with Dll1 siRNA.

도 36은 ApoSQ와 접촉한 CAFs에 annexin V를 처리할 경우, Brain-specific angiogenesis inhibitor 1 (BAI1) 신호가 억제됨에 따라 CAFs의 ApoSQ 탐식 작용, Notch1 활성화 및 WISP-1 분비가 억제됨을 나타낸 도이다. 도 36a는 annexin V의 존재 또는 부재하에 ApoSQ 세포를 노출시킨 후 CAFs 탐식작용을 유세포 분석으로 %로 측정한 도이다. 도 36b에서 (A)는 annexin V의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs에서의 NICD1, Hes1 및 WISP-1에 대한 면역 블랏 분석 결과를 나타낸 도이고, (B)는 annexin V의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs에서의 NICD1, Hes1 및 WISP-1 단백질의 양을 측정한 도이고, (C)는 annexin V의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs에서 측정한 WISP-1의 ELISA 결과를 나타낸 도이다.36 is a diagram showing that, when CAFs in contact with ApoSQ are treated with annexin V, brain-specific angiogenesis inhibitor 1 (BAI1) signals are inhibited, thereby inhibiting ApoSQ phagocytosis, Notch1 activation, and WISP-1 secretion in CAFs. Figure 36a is a diagram showing the percentage of CAFs phagocytosis after exposure to ApoSQ cells in the presence or absence of annexin V by flow cytometry. In FIG. 36B, (A) is a diagram showing the immunoblot analysis results for NICD1, Hes1, and WISP-1 in CAFs contacted with ApoSQ in the presence or absence of annexin V, and (B) is a diagram showing the results of immunoblot analysis in the presence or absence of annexin V. A diagram measuring the amount of NICD1, Hes1 and WISP-1 proteins in CAFs in contact with ApoSQ, and (C) shows the ELISA results of WISP-1 measured in CAFs in contact with ApoSQ in the presence or absence of annexin V. It is also

도 37은 ApoSQ와 접촉한 CAFs를 BAI1 siRNA로 형질감염 시킬 경우, BAI1 신호가 억제됨에 따라 CAFs의 탐식 작용, Notch1 활성화 및 WISP-1 분비가 억제됨을 나타낸 도이다. 도 37a는 CAFs를 BAI1 siRNA로 형질감염 시킨 후 측정한 면역 블랏 분석 결과 및 BAI1 단백질의 양을 나타낸 도이다. 도 37b는 CAFs를 BAI1 siRNA로 형질감염 시킨 후 ApoSQ 세포를 노출시킨 후 CAFs 탐식작용을 유세포 분석으로 %로 측정한 도이다. 도 37c에서 (A)는 CAFs를 BAI1 siRNA로 형질감염 시킨 후 ApoSQ의 유무에 따라 측정한 NICD1, Hes1 및 WISP-1에 대한 면역 블랏 분석 결과를 나타낸 도이고, (B)는 CAFs를 BAI1 siRNA로 형질감염 시킨 후 ApoSQ의 유무에 따라 측정한 NICD1, Hes1 및 WISP-1 단백질의 양을 나타낸 도이고, (C)는 CAFs를 BAI1 siRNA로 형질감염 시킨 후 ApoSQ의 유무에 따라 측정한 WISP-1의 ELISA 결과를 나타낸 도이다.37 is a diagram showing that when CAFs in contact with ApoSQ were transfected with BAI1 siRNA, phagocytosis of CAFs, Notch1 activation, and WISP-1 secretion were suppressed as BAI1 signal was suppressed. 37a is a diagram showing the results of immunoblot analysis and the amount of BAI1 protein measured after CAFs were transfected with BAI1 siRNA. Figure 37b is a diagram showing the phagocytosis of CAFs measured in % by flow cytometry after CAFs were transfected with BAI1 siRNA and exposed to ApoSQ cells. In FIG. 37C, (A) is a diagram showing the immunoblot analysis results for NICD1, Hes1, and WISP-1 measured in the presence or absence of ApoSQ after CAFs were transfected with BAI1 siRNA, and (B) is a diagram showing CAFs with BAI1 siRNA. After transfection, the amount of NICD1, Hes1, and WISP-1 proteins measured according to the presence or absence of ApoSQ is a diagram. It is a diagram showing the results of ELISA.

도 38은 ApoSQ와 접촉한 CAFs에 항-BAI1 항체를 처리할 경우, BAI1 신호가 억제됨에 따라 CAFs의 탐식 작용, Notch1 활성화 및 WISP-1 분비가 억제됨을 나타낸 도이다. 도 38a는 항-BAI1 항체의 존재 또는 부재하에 ApoSQ 세포를 노출시킨 후 CAFs 탐식작용을 유세포 분석으로 %로 측정한 도이다. 도 38b에서 (A)는 항-BAI1 항체의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs에서의 NICD1, Hes1 및 WISP-1에 대한 면역 블랏 분석 결과를 나타낸 도이고, (B)는 항-BAI1 항체의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs에서의 NICD1, Hes1 및 WISP-1 단백질의 양을 측정한 도이고, (C)는 항-BAI1 항체의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs에서 측정한 WISP-1의 ELISA 결과를 나타낸 도이다.38 is a diagram showing that when CAFs in contact with ApoSQ are treated with an anti-BAI1 antibody, BAI1 signals are suppressed, and phagocytosis, Notch1 activation, and WISP-1 secretion of CAFs are suppressed. Figure 38a is a diagram showing the percentage of CAFs phagocytosis after exposure of ApoSQ cells in the presence or absence of anti-BAI1 antibody by flow cytometry. In FIG. 38B, (A) is a diagram showing the results of immunoblot analysis for NICD1, Hes1, and WISP-1 in CAFs contacted with ApoSQ in the presence or absence of anti-BAI1 antibody, and (B) is a diagram showing the results of anti-BAI1 antibody A diagram showing the amount of NICD1, Hes1 and WISP-1 proteins in CAFs contacted with ApoSQ in the presence or absence, and (C) is WISP-1 measured in CAFs contacted with ApoSQ in the presence or absence of anti-BAI1 antibody. It is a diagram showing the ELISA result of 1.

도 39는 ApoSQ와 접촉한 CAFs를 BAI1을 과발현하는 BAI1-Flag로 형질감염 시킬 경우, BAI1 신호가 증가됨에 따라 CAFs의 탐식 작용, Notch1 활성화 및 WISP-1 분비가 증가함을 나타낸 도이다. 도 39a는 CAFs를 BAI1-Flag로 형질감염 시킨 후 측정한 면역 블랏 분석 결과 및 BAI1 단백질의 양을 나타낸 도이다. 도 39b는 CAFs를 BAI1-Flag로 형질감염 시킨 후 ApoSQ 세포를 노출시킨 후 CAFs 탐식작용을 유세포 분석으로 %로 측정한 도이다. 도 39c에서 (A)는 CAFs를 BAI1-Flag로 형질감염 시킨 후 ApoSQ의 유무에 따라 측정한 NICD1, Hes1 및 WISP-1에 대한 면역 블랏 분석 결과를 나타낸 도이고, (B)는 CAFs를 BAI1-Flag로 형질감염 시킨 후 ApoSQ의 유무에 따라 측정한 NICD1, Hes1 및 WISP-1 단백질의 양을 나타낸 도이고, (C)는 CAFs를 BAI1-Flag로 형질감염 시킨 후 ApoSQ의 유무에 따라 측정한 WISP-1의 ELISA 결과를 나타낸 도이다.39 is a diagram showing that when CAFs contacted with ApoSQ were transfected with BAI1-Flag overexpressing BAI1, phagocytosis of CAFs, Notch1 activation, and WISP-1 secretion increased as BAI1 signal increased. 39a is a diagram showing the results of immunoblot analysis and the amount of BAI1 protein measured after CAFs were transfected with BAI1-Flag. Figure 39b is a diagram showing the phagocytosis of CAFs measured in % by flow cytometry after CAFs were transfected with BAI1-Flag and exposed to ApoSQ cells. In FIG. 39C, (A) is a diagram showing the results of immunoblot analysis for NICD1, Hes1, and WISP-1 measured according to the presence or absence of ApoSQ after CAFs were transfected with BAI1-Flag, and (B) is a diagram showing CAFs transfected with BAI1-Flag. A diagram showing the amounts of NICD1, Hes1, and WISP-1 proteins measured according to the presence or absence of ApoSQ after transfection with Flag. It is a diagram showing the ELISA result of -1.

도 40은 ApoSQ가 CAFs에서 Rac1의 활성을 시간 의존적으로 증가시킴을 나타낸 도이다.40 is a diagram showing that ApoSQ increases the activity of Rac1 in a time-dependent manner in CAFs.

도 41은 ApoSQ와 접촉한 CAFs에 Rac1 억제제인 NSC23766을 처리할 경우, Rac1 신호가 억제됨에 따라 CAFs의 탐식 작용, Notch1 활성화 및 WISP-1 분비가 억제됨을 나타낸 도이다. 도 41a와 41b는 100 μM NSC23766의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs의 탐식 작용을 측정한 도이다. 도 41a는 유세포 분석으로 ApoSQ 세포를 탐식한 CAFs 수를 %로 측정한 도이다. 도 41b는 공초점 현미경을 사용하여 탐식 작용 활동도를 탐식지수(phagocytic index)로 표현한 도이다. 도 41c에서 (A)는 NSC23766의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs에서의 NICD1, Hes1 및 WISP-1에 대한 면역 블랏 분석 결과를 나타낸 도이고, (B)는 NSC23766의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs에서의 NICD1, Hes1 및 WISP-1 단백질의 양을 측정한 도이고, (C)는 NSC23766의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs에서 측정한 WISP-1의 ELISA 결과를 나타낸 도이다.41 is a diagram showing that when CAFs contacted with ApoSQ are treated with NSC23766, a Rac1 inhibitor, phagocytosis of CAFs, Notch1 activation, and WISP-1 secretion are inhibited as Rac1 signaling is inhibited. 41a and 41b are diagrams measuring the phagocytic activity of CAFs in contact with ApoSQ in the presence or absence of 100 μM NSC23766. 41a is a diagram showing the number of CAFs phagocytosing ApoSQ cells in % by flow cytometry. 41B is a diagram illustrating phagocytic activity as a phagocytic index using a confocal microscope. In FIG. 41C, (A) is a diagram showing the immunoblot analysis results for NICD1, Hes1, and WISP-1 in CAFs contacted with ApoSQ in the presence or absence of NSC23766, and (B) is a diagram showing the results of immunoblot analysis for ApoSQ and ApoSQ in the presence or absence of NSC23766. A diagram showing the amounts of NICD1, Hes1, and WISP-1 proteins in CAFs contacted, and (C) is a diagram showing ELISA results of WISP-1 measured in CAFs contacted with ApoSQ in the presence or absence of NSC23766.

도 42는 ApoSQ와 접촉한 CAFs를 Notch1 siRNA로 형질감염 시킬 경우, Notch1 신호가 억제됨에 따라 Rac1의 활성 및 CAFs의 탐식 작용이 억제됨을 나타낸 도이다. (A)CAFs를 Notch1 siRNA로 형질감염 시킨 후 측정한 Rac1의 활성도를 나타낸 도이다. (B)CAFs를 Notch1 siRNA로 형질감염 시킨 후 ApoSQ 세포를 노출시킨 후 CAFs 탐식작용을 유세포 분석으로 %로 측정한 도이다. 42 is a diagram showing that when CAFs in contact with ApoSQ are transfected with Notch1 siRNA, the activity of Rac1 and the phagocytosis of CAFs are suppressed according to the inhibition of Notch1 signal. (A) A diagram showing the activity of Rac1 measured after transfecting CAFs with Notch1 siRNA. (B) After transfecting CAFs with Notch1 siRNA and exposing ApoSQ cells, the phagocytosis of CAFs was measured in % by flow cytometry.

도 43은 ApoSQ와 접촉한 CAFs에 DAPT를 처리할 경우, Notch1 신호가 억제됨에 따라 Rac1의 활성 및 CAFs의 탐식 작용이 억제됨을 나타낸 도이다. (A)20μM DAPT의 존재 또는 부재하에 측정한 Rac1의 활성도를 나타낸 도이다. (B)DAPT의 존재 또는 부재하에 ApoSQ와 접촉한 CAFs의 탐식 작용을 유세포 분석을 통하여 측정한 도이다.43 is a diagram showing that when CAFs contacted with ApoSQ were treated with DAPT, the activity of Rac1 and phagocytosis of CAFs were suppressed as the Notch1 signal was suppressed. (A) A diagram showing the activity of Rac1 measured in the presence or absence of 20 μM DAPT. (B) A diagram showing the phagocytosis of CAFs in contact with ApoSQ in the presence or absence of DAPT through flow cytometry.

도 44는 344SQ 세포를 동계(syngeneic) 마우스의 측면 피하에 주입하고 2일 후에 ApoSQ를 피하에 투여 후 6주 경과한 후 마우스를 희생시키고 원발성 종양 조직으로부터 Thy1 CAFs를 분리하는 실험 타임라인을 나타낸 도이다.44 is a diagram showing an experimental timeline for injecting 344SQ cells subcutaneously on the flanks of syngeneic mice, subcutaneously administering ApoSQ 2 days later, sacrificing the mice 6 weeks later, and isolating Thy1 CAFs from primary tumor tissue. am.

도 45는 344SQ 세포를 동계(syngeneic) 마우스의 측면 피하에 주입 후 2일 후에 ApoSQ를 피하에 투여하고 6주 경과 후 마우스를 희생시키고 원발성 종양 조직에서 분리한 Thy1+ CAFs에서, 대조군의 Thy1+ CAFs과 비교시, CAF 활성 마커(Acta2, Col1α1, Fn, Itg beta 1, Spp1, Pdgfrα, 및 Pdgfr beta 포함), MMP(Mmp1a, 2, 9, 및 12 포함), 성장 인자 및 케모카인 (Vegfa, Hgf, Cxcl12, 및 Cxcl14 포함)의 mRNA 발현양이 감소하며, Notch1 관련 분자(Notch1, Wisp1 (Ccn4), Hey1, Hey2, 및 Hes1 포함)의 mRNA 발현양이 증가함을 qRT-PCR을 통해 측정한 도이다. 도 45a는 qRT-PCR 결과를 토대로 만들어진 히트맵을 나타낸 도이다. 도 45b는 각 mRNA에 대한 qRT-PCR 결과를 나타낸 도이다.45 shows Thy1 + CAFs isolated from primary tumor tissue after 6 weeks of subcutaneous administration of ApoSQ 2 days after 344SQ cells were injected subcutaneously into the flanks of syngeneic mice, and mice sacrificed 6 weeks later. Thy1 + CAFs of the control group , CAF activation markers (including Acta2, Col1α1, Fn, Itg beta 1, Spp1, Pdgfrα, and Pdgfr beta), MMPs (including Mmp1a, 2, 9, and 12), growth factors and chemokines (Vegfa, Hgf, It is a diagram measured by qRT-PCR that the mRNA expression level of Notch1-related molecules (including Notch1, Wisp1 (Ccn4), Hey1, Hey2, and Hes1) is increased, while the mRNA expression level of Cxcl12 and Cxcl14 is decreased. . 45A is a diagram showing a heat map made based on qRT-PCR results. 45B is a diagram showing qRT-PCR results for each mRNA.

도 46은 마우스에 ApoSQ 및 LY3039478을 투여하는 실험 타임라인을 나타낸 도이다.46 is a diagram showing the experimental timeline of administering ApoSQ and LY3039478 to mice.

도 47은 마우스에 LY3039478(8 mg/kg), ApoSQ 또는 ApoSQ 및 LY3039478(8 mg/kg)을 투여하여 마우스 체중, 종양 중량 및 종양 부피를 측정한 결과 각 그룹 간 유의미한 차이가 없음을 나타낸 도이다. (A)각 그룹에서의 마우스 체중, 종양 중량 및 종양 부피를 측정한 도이다. (B)각 그룹에서의 종양의 이미지를 나타낸 도이다.47 is a diagram showing that there is no significant difference between each group as a result of measuring mouse weight, tumor weight and tumor volume by administering LY3039478 (8 mg/kg), ApoSQ or ApoSQ and LY3039478 (8 mg/kg) to mice . (A) A diagram showing the mouse weight, tumor weight and tumor volume in each group. (B) A diagram showing images of tumors in each group.

도 48은 마우스에 LY3039478, ApoSQ 또는 ApoSQ 및 LY3039478을 투여하여 폐로 전이된 종양 결절의 수 및 전이 비율을 측정한 결과 ApoSQ 투여 시 종양 결절의 수 및 전이 비율이 감소하고, LY3039478을 추가적으로 투여 시 상기 효과가 역전됨을 나타낸 도이다. (A)각 그룹에서의 종양 결절의 수 및 전이 비율을 나타낸 도이다. (B)각 그룹에서의 폐에 전이된 종양 결절의 이미지를 나타낸 도이다.48 is a result of measuring the number and metastasis rate of tumor nodules metastasized to the lung by administering LY3039478, ApoSQ or ApoSQ and LY3039478 to mice, and as a result, the number and metastasis rate of tumor nodules decreased when ApoSQ was administered, and the effect of additional administration of LY3039478 It is a diagram showing that is reversed. (A) A diagram showing the number of tumor nodules and metastasis rate in each group. (B) It is a diagram showing images of tumor nodules metastasized to the lungs in each group.

도 49는 ApoSQ를 투여한 경우 원발성 종양조직에서 분리된 Thy1+ CAFs에 CAF 활성 마커, MMP, 성장 인자, 케모카인의 mRNA 발현양이 감소하며, Notch1 관련 분자의 mRNA 발현양이 증가하나, LY3039478을 추가적으로 투여 시 상기 효과가 역전됨을 나타낸 도이다.49 shows that when ApoSQ was administered, the mRNA expression levels of CAF activity markers, MMPs, growth factors, and chemokines decreased in Thy1 + CAFs isolated from primary tumor tissue, and the mRNA expression levels of Notch1-related molecules increased, but LY3039478 was additionally administered. It is a diagram showing that the effect is reversed upon administration.

도 50은 면역형광염색을 통해 원발성 종양조직에서, ApoSQ를 피하로 투여한 경우, Thy-1+ CAFs에서α-SMA의 형광 세기가 감소하고 NICD1 및 WISP-1의 형광 세기가 증가하나, LY3039478을 추가적으로 투여 시 상기 효과가 역전됨을 나타낸 도이다.50 shows that in primary tumor tissue through immunofluorescence staining, when ApoSQ was subcutaneously administered, the fluorescence intensity of α-SMA decreased and the fluorescence intensity of NICD1 and WISP-1 increased in Thy-1 + CAFs, but LY3039478 It is a diagram showing that the effect is reversed when additionally administered.

도 51은 ApoSQ를 투여한 마우스의 원발성 종양조직에서 분리된 Thy1+ CAFs에서 면역세포화학 분석을 통해 α-SMA의 발현이 감소하고 NICD1 및 WISP-1의 발현이 증가하나, LY3039478을 추가적으로 투여 시 상기 효과가 역전됨을 나타낸 도이다. 도 51a는 각 그룹에서의 α-SMA 발현 정도를 면역세포화학 분석을 통해 나타낸 도이다. 도 51b는 각 그룹에서의 NICD1 및 WISP-1 발현 정도를 면역세포화학 분석을 통해 나타낸 도이다.51 shows a decrease in the expression of α-SMA and an increase in the expression of NICD1 and WISP-1 through immunocytochemical analysis in Thy1 + CAFs isolated from primary tumor tissues of mice administered with ApoSQ, but when LY3039478 is additionally administered. It is a diagram showing that the effect is reversed. 51A is a diagram showing the expression level of α-SMA in each group through immunocytochemical analysis. 51b is a diagram showing the expression levels of NICD1 and WISP-1 in each group through immunocytochemical analysis.

도 52는 ApoSQ를 투여한 마우스의 원발성 종양조직에서 분리된 Thy1+ CAFs의 배양액 및 혈청에 WISP-1의 발현이 증가하고, LY3039478을 추가적으로 투여 시 상기 효과가 역전되나, 분리된 CD326+ 종양 세포 및 CD11b+ 종양 관련 대식세포에는 상기 효과가 없음을 나타낸 도이다.52 shows that the expression of WISP-1 increases in the culture medium and serum of Thy1 + CAFs isolated from primary tumor tissues of mice administered with ApoSQ, and the effect is reversed when LY3039478 is additionally administered, but the isolated CD326 + tumor cells and It is a diagram showing that there is no such effect on CD11b + tumor-associated macrophages.

도 53은 ApoSQ를 투여한 마우스의 원발성 종양조직에서 분리된 CD326+ 종양 세포 및 Thy1+ CAFs에서 세포 이동 및 침윤이 억제되나, LY3039478을 추가적으로 투여 시 상기 효과가 역전됨을 나타낸 도이다. 도 53a는 분리된 CD326+ 종양 세포의 이동 및 침윤을 측정한 도이다. 도 53b는 분리된 Thy1+ CAFs의 이동 및 침윤을 측정한 도이다.53 is a diagram showing that cell migration and invasion are inhibited in CD326 + tumor cells and Thy1 + CAFs isolated from primary tumor tissues of mice administered with ApoSQ, but the above effects are reversed when LY3039478 is additionally administered. Figure 53a is a diagram measuring migration and invasion of isolated CD326 + tumor cells. Figure 53b is a diagram showing the migration and invasion of isolated Thy1 + CAFs.

도 54는 ApoSQ를 투여한 마우스의 원발성 종양조직에서 분리된 CD326+ 종양 세포에서 Smad2/3, FAK, ERK 및 Akt와 같은 이동 및 침윤에 관련된 신호분자들의 인산화와 MMP2/12 단백질의 발현이 억제되나, LY3039478을 추가적으로 투여 시 상기 효과가 역전됨을 나타낸 도이다. (A) 분리된 CD326+ 종양 세포에서의 세포 이동 및 침윤에 관련된 신호분자들의 인산화 및 MMP2/12에 대한 면역 블랏 분석 결과를 나타낸 도이다. (B)분리된 CD326+ 종양 세포에서의 세포 이동 및 침윤에 관련된 신호분자들의 인산화 및 MMP2/12 양을 측정한 도이다.54 shows that phosphorylation of signaling molecules related to migration and invasion, such as Smad2/3, FAK, ERK, and Akt, and expression of MMP2/12 protein are inhibited in CD326 + tumor cells isolated from primary tumor tissues of mice administered with ApoSQ. , It is a diagram showing that the effect is reversed when LY3039478 is additionally administered. (A) A diagram showing the results of immunoblot analysis for phosphorylation and MMP2/12 of signaling molecules related to cell migration and invasion in isolated CD326 + tumor cells. (B) It is a diagram measuring the amount of phosphorylation and MMP2/12 of signaling molecules related to cell migration and invasion in isolated CD326 + tumor cells.

도 55는 마우스에 344SQ 세포를 피하 주사 후 CAF CM, ApoSQ-CAF CM, ApoSQ-CAF CM 및 항-WISP-1 항체, 또는 ApoSQ-CAF CM 및 IgG를 투여하는 실험 타임라인을 나타낸 도이다.55 is a diagram showing the experimental timeline of administering CAF CM, ApoSQ-CAF CM, ApoSQ-CAF CM and anti-WISP-1 antibody, or ApoSQ-CAF CM and IgG to mice after subcutaneous injection of 344SQ cells.

도 56은 마우스에 CAF CM, ApoSQ-CAF CM, ApoSQ-CAF CM 및 항-WISP-1 항체 또는 ApoSQ-CAF CM 및 IgG를 종양내 투여할 경우, 마우스 체중은 각 그룹 간 유의미한 차이가 없음을 나타낸 도이다.56 shows that when CAF CM, ApoSQ-CAF CM, ApoSQ-CAF CM and anti-WISP-1 antibody or ApoSQ-CAF CM and IgG were intratumorally administered to mice, there was no significant difference in mouse weight between groups. It is also

도 57은 마우스에 CAF CM, ApoSQ-CAF CM, ApoSQ-CAF CM 및 항-WISP-1 항체 또는 ApoSQ-CAF CM 및 IgG를 투여하여 종양 결절의 수 및 전이 비율을 측정한 결과 ApoSQ-CAF CM 투여 시, CAF CM 실험군과 비교시, 폐로 전이된 종양 결절의 수 및 전이 비율이 감소하고, 항-WISP-1 항체를 추가적으로 투여 시 상기 효과가 역전됨을 나타낸 도이다. (A)각 그룹에서의 폐로 전이된 종양 결절의 수 및 전이 비율을 나타낸 도이다. (B)각 그룹에서의 폐로 전이된 종양 결절의 이미지를 나타낸 도이다.57 is a result of measuring the number of tumor nodules and metastasis rate by administering CAF CM, ApoSQ-CAF CM, ApoSQ-CAF CM and anti-WISP-1 antibody or ApoSQ-CAF CM and IgG to mice, and ApoSQ-CAF CM administration When compared with the CAF CM experimental group, the number and metastasis rate of tumor nodules metastasized to the lungs decreased, and the effect was reversed when an anti-WISP-1 antibody was additionally administered. (A) A diagram showing the number and metastasis rate of tumor nodules that metastasized to the lungs in each group. (B) It is a diagram showing images of tumor nodules that metastasized to the lungs in each group.

도 58은 마우스에 ApoSQ-CAF CM을 종양내 투여할 경우, CAF CM 실험군과 비교시, 원발성 종양조직에서 분리된 CD326+ 종양 세포 및 Thy1+ CAFs에 세포의 이동 및 침윤이 억제되나, 항-WISP-1 항체를 추가적으로 투여 시 상기 효과가 역전됨을 나타낸 도이다. 도 58a는 원발성 종양조직에서 분리된 CD326+ 종양 세포의 이동 및 침윤을 측정한 도이다. 도 58b는 원발성 종양조직에서 분리된 Thy1+ CAFs의 이동 및 침윤을 측정한 도이다.58 shows that when ApoSQ-CAF CM was intratumorally administered to mice, compared to the CAF CM experimental group, cell migration and invasion were inhibited in CD326 + tumor cells and Thy1 + CAFs isolated from primary tumor tissue, but anti-WISP -1 It is a diagram showing that the effect is reversed when the antibody is additionally administered. 58a is a diagram showing migration and invasion of CD326 + tumor cells isolated from primary tumor tissue. 58b is a diagram showing the migration and invasion of Thy1 + CAFs isolated from primary tumor tissue.

도 59는 마우스에 ApoSQ-CAF CM을 종양내 투여할 경우, CAF CM 실험군과 비교시, 원발성 종양조직에서 분리된 CD326+ 종양 세포에 Smad2/3, FAK, ERK 및 Akt와 같은 이동 및 침윤에 관련된 신호분자들의 인산화와 MMP2/12 단백질의 발현 양이 억제되나, 항-WISP-1 항체를 추가적으로 투여 시 상기 효과가 역전됨을 나타낸 도이다. (A)CD326+ 종양 세포에서의 세포 이동 및 침윤에 관련된 분자들에 대한 면역 블랏 분석 결과를 나타낸 도이다. (B)분리된 CD326+ 종양 세포에서의 세포 이동 및 침윤에 관련된 단백질들의 양을 측정한 도이다.59 shows that when ApoSQ-CAF CM was intratumorally administered to mice, compared to the CAF CM experimental group, CD326 + tumor cells isolated from primary tumor tissues were associated with migration and invasion, such as Smad2/3, FAK, ERK, and Akt. Phosphorylation of signal molecules and expression of MMP2/12 protein are suppressed, but the effect is reversed when anti-WISP-1 antibody is additionally administered. (A) A diagram showing the results of immunoblot analysis for molecules involved in cell migration and invasion in CD326 + tumor cells. (B) A diagram showing the amount of proteins involved in cell migration and invasion in isolated CD326 + tumor cells.

도 60은 마우스에 ApoSQ-CAF CM을 종양내 투여할 경우, CAF CM 실험군과 비교시, 원발성 종양조직에서 분리된 Thy1+ CAFs에서의 CAF 활성 마커(Acta2, Col1α1, Itg beta 1, Spp1 및 Pdgfrα 포함) 및 MMP(Mmp1a, 2, 및 12 포함)의 mRNA 발현 양이 억제되나, 항-WISP-1 항체를 추가적으로 투여 시 상기 효과가 역전됨을 나타낸 도이다.60 shows CAF activity markers (Acta2, Col1α1, Itg beta 1 , Spp1 and Pdgfrα included ) and MMP (including Mmp1a, 2, and 12) mRNA expression levels are suppressed, but the effect is reversed when an anti-WISP-1 antibody is additionally administered.

도 61은 마우스에 344SQ 세포를 피하 주사 후 rWISP-1을 12.5 μg/kg 또는 25 μg/kg의 농도로 투여하는 실험 타임라인을 나타낸 도이다.61 is a diagram showing an experimental timeline of administration of rWISP-1 at a concentration of 12.5 μg/kg or 25 μg/kg after subcutaneous injection of 344SQ cells into mice.

도 62는 마우스에 rWISP-1을 12.5 μg/kg 또는 25 μg/kg의 농도로 종양내 투여할 경우, 마우스 체중은 각 그룹 간 유의미한 차이가 없음을 나타낸 도이다.62 is a diagram showing that when rWISP-1 was intratumorally administered to mice at a concentration of 12.5 μg/kg or 25 μg/kg, there was no significant difference in body weight between groups.

도 63은 마우스에 rWISP-1을 12.5 μg/kg 또는 25 μg/kg의 농도로 투여하여 종양 결절의 수 및 전이 비율을 측정한 결과, 폐로 전이된 종양 결절의 수가 감소하였으며, 전이 비율은 농도 의존적으로 감소함을 나타낸 도이다. (A)각 그룹에서의 폐로 전이된 종양 결절의 수 및 전이 비율을 나타낸 도이다. (B)각 그룹에서의 폐로 전이된 종양 결절의 이미지를 나타낸 도이다.63 shows the number of tumor nodules and the metastasis rate measured by administering rWISP-1 to mice at a concentration of 12.5 μg/kg or 25 μg/kg. It is a diagram showing a decrease in (A) A diagram showing the number and metastasis rate of tumor nodules that metastasized to the lungs in each group. (B) It is a diagram showing images of tumor nodules that metastasized to the lungs in each group.

도 64는 마우스에 rWISP-1을 12.5 μg/kg 또는 25 μg/kg의 농도로 종양내 투여할 경우, CD326+ 종양 세포 및 Thy1+ CAFs에 세포의 이동 및 침윤이 농도 의존적으로 억제됨을 나타낸 도이다. 도 64a는 CD326+ 종양 세포의 이동 및 침윤을 측정한 도이다. 도 64b는 Thy1+ CAFs의 이동 및 침윤을 측정한 도이다.64 is a diagram showing that migration and invasion of CD326 + tumor cells and Thy1 + CAFs are inhibited in a concentration-dependent manner when rWISP-1 is intratumorally administered at a concentration of 12.5 μg/kg or 25 μg/kg to mice . Figure 64a is a diagram showing the migration and invasion of CD326 + tumor cells. Figure 64b is a diagram showing the migration and invasion of Thy1 + CAFs.

도 65는 마우스에 rWISP-1을 12.5 μg/kg 또는 25 μg/kg의 농도로 종양내 투여할 경우, CD326+ 종양 세포에 Smad2/3, FAK, ERK 및 Akt와 같은 이동 및 침윤에 관련된 신호분자들의 인산화와 MMP2/12 단백질의 발현 양이 억제됨을 나타낸 도이다. (A)CD326+ 종양 세포에서의 세포 이동 및 침윤에 관련된 분자들에 대한 면역 블랏 분석 결과를 나타낸 도이다. (B)CD326+ 종양 세포에서의 세포 이동 및 침윤에 관련된 단백질들의 양을 측정한 도이다.65 shows signal molecules related to migration and invasion, such as Smad2/3, FAK, ERK, and Akt, in CD326 + tumor cells when rWISP-1 was intratumorally administered to mice at a concentration of 12.5 μg/kg or 25 μg/kg. It is a diagram showing that the phosphorylation of them and the expression of MMP2/12 protein are suppressed. (A) A diagram showing the results of immunoblot analysis for molecules involved in cell migration and invasion in CD326 + tumor cells. (B) A diagram showing the amount of proteins involved in cell migration and invasion in CD326 + tumor cells.

도 66은 마우스에 rWISP-1을 12.5 μg/kg 또는 25 μg/kg의 농도로 종양내 투여할 경우, Thy1+ CAFs에서의 CAF 활성 마커(Acta2, Col1α1, Itg beta 1, Spp1 및 Pdgfrα 포함) 및 MMP(Mmp1a, 2, 및 12 포함)의 mRNA 발현 양이 억제됨을 나타낸 도이다.66 shows CAF activity markers (including Acta2, Col1α1, Itg beta 1, Spp1 and Pdgfrα) and It is a diagram showing that the mRNA expression level of MMP (including Mmp1a, 2, and 12) is suppressed.

도 67은 마우스에 ApoSQ-CAF CM을 종양내 투여할 경우, CAF CM 실험군과 비교시, 원발성 종양조직에서 분리된 CD326+ 종양 세포에 종양 진행 및 침윤에 관한 유전자 발현은 하향 조절되고, 종양 진행 및 침윤 억제에 관한 유전자 발현은 상향 조절됨을 나타낸 도이다.67 shows that when ApoSQ-CAF CM was intratumorally administered to mice, compared to the CAF CM experimental group, the expression of genes related to tumor progression and invasion was downregulated in CD326 + tumor cells isolated from primary tumor tissues, and tumor progression and It is a diagram showing that gene expression related to invasion inhibition is up-regulated.

도 68은 마우스에 ApoSQ-CAF CM을 종양내 투여할 경우, CAF CM 실험군과 비교시, 원발성 종양조직에서 분리된 Thy1+ CAFs에 세포 접착 및 ECM 리모델링에 관한 유전자 발현이 하향 조절됨을 나타낸 도이다.68 is a diagram showing that, when ApoSQ-CAF CM was intratumorally administered to mice, gene expression related to cell adhesion and ECM remodeling was down-regulated in Thy1 + CAFs isolated from primary tumor tissues, compared to the CAF CM experimental group.

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.Hereinafter, the present invention will be described in more detail through examples. However, these examples are intended to illustrate the present invention by way of example, and the scope of the present invention is not limited to these examples.

실험예 1: 재료 및 방법Experimental Example 1: Materials and Methods

1-1. 시약1-1. reagent

DAPT(D5942)는 Sigma-Aldrich(St. Louis, MO, USA)사로부터 구입하였고, TGF-beta 1(240-B-010) 및 마우스 rWISP-1(1680-WS), 마우스 중화 WISP-l 항체(MAB1680 R&D Systems) 및 IgG(MAB0061)는 R&D Systems(Minneapolis, MN, USA)사로부터 구입하였다. LY3039478(HY-12449)는 MedChemExpress(Monmouth Junction, NJ, USA)사로부터 구입하였다.DAPT (D5942) was purchased from Sigma-Aldrich (St. Louis, MO, USA), TGF-beta 1 (240-B-010) and mouse rWISP-1 (1680-WS), mouse neutralizing WISP-1 antibody (MAB1680 R&D Systems) and IgG (MAB0061) were purchased from R&D Systems (Minneapolis, MN, USA). LY3039478 (HY-12449) was purchased from MedChemExpress (Monmouth Junction, NJ, USA).

1-2. 항체1-2. antibody

웨스턴 블랏, 면역형광염색, 유세포 분석 및 세포 분류에 사용된 항체들은 다음과 같다(표 1 참조).Antibodies used for Western blot, immunofluorescence staining, flow cytometry and cell sorting are as follows (see Table 1).

AntigenAntigen VendorVendor Cat.NoCat. No SourceSource Species cross-reactivitySpecies cross-reactivity ApplicationApplication DilutionDilution p-Smad2p-Smad2 Cell SignalingCell Signaling 31083108 Rabbit monoclonalRabbit monoclonal H, M, R, MiH, M, R, Mi IBIB 1:10001:1000 p-Smad3p-Smad3 Cell SignalingCell Signaling 95209520 Rabbit monoclonalRabbit monoclonal H, M, RH, M, R IBIB 1:10001:1000 p-AKTp-AKT Cell SignalingCell Signaling 40604060 Rabbit monoclonalRabbit monoclonal H, M, R, Hm, Mk, Dm, Z, BH, M, R, Hm, Mk, Dm, Z, B IBIB 1:10001:1000 p-FAKp-FAK Cell SignalingCell Signaling 32833283 Rabbit polyclonalRabbit polyclonal H, M, R, Hm, PgH, M, R, Hm, Pg IBIB 1:10001:1000 p-Srcp-Src Cell SignalingCell Signaling 21012101 Rabbit polyclonalRabbit polyclonal H, M, RH, M, R IBIB 1:10001:1000 p-ERKp-ERK Cell SignalingCell Signaling 91019101 Rabbit polyclonalRabbit polyclonal B, Ce, Dm, Hm, H,
Mi, Mk, M
B, Ce, Dm, Hm, H,
Mi, Mk, M
IBIB 1:10001:1000
p-p38p-p38 Santa CruzSanta Cruz sc-17852-Rsc-17852-R Rabbit polyclonalRabbit polyclonal H, M, RH, M, R IBIB 1:10001:1000 Smad2/3Smad2/3 Cell SignalingCell Signaling 31023102 Rabbit polyclonalRabbit polyclonal H, M, R, MkH, M, R, MK IBIB 1:10001:1000 AktAkt Cell SignalingCell Signaling 46914691 Rabbit monoclonalRabbit monoclonal H, M, R, Mk, DmH, M, R, Mk, Dm IBIB 1:10001:1000 FAKFAK Cell SignalingCell Signaling 32853285 Rabbit polyclonalRabbit polyclonal H, M, R, Hm, B, PgH, M, R, Hm, B, Pg IBIB 1:10001:1000 SrcSrc Santa CruzSanta Cruz sc-19sc-19 Rabbit polyclonalRabbit polyclonal H, M, RH, M, R IBIB 1:10001:1000 ERKERK Santa CruzSanta Cruz sc-93-Gsc-93-G Goat polyclonalGoat polyclonal H, M, R, C, FH, M, R, C, F IBIB 1:10001:1000 p38p38 Santa CruzSanta Cruz sc-535-Gsc-535-G Goat polyclonalGoat polyclonal H, M, RH, M, R IBIB 1:10001:1000 MMP2MMP2 Cell SignalingCell Signaling #87809#87809 Rabbit monoclonalRabbit monoclonal H, MH, M IBIB 1:10001:1000 MMP12MMP12 Santa CruzSanta Cruz sc-390863sc-390863 Mouse monoclonalMouse monoclonal H, M, RH, M, R IBIB 1:10001:1000 α-SMAα-SMA AbcamAbcam ab7817ab7817 Mouse monoclonal Mouse monoclonal H, M, R, Rb, PgH, M, R, Rb, Pg IB, IF, IHCIB, IF, IHC 1:1000
1:200
1:1000
1:200
Col1αCol1α GeneTexGeneTex GRX82721GRX82721 Rabbit polyclonalRabbit polyclonal H, M, RH, M, R IBIB 1:10001:1000 FNFN AbcamAbcam ab2413ab2413 Rabbit polyclonalRabbit polyclonal H, MH, M IBIB 1:10001:1000 LIFLIF AbcamAbcam ab113262ab113262 Rabbit polyclonalRabbit polyclonal H, M, RH, M, R IBIB 1:10001:1000 WISP-1WISP-1 AbcamAbcam ab178547ab178547 Rabbit polyclonalRabbit polyclonal H, M, RH, M, R IB, IFIB, IF 1:10001:1000   R&D SystemsR&D Systems AF1680AF1680 Sheep polyclonalSheep polyclonal MM IF, IHCIF, IHC 1:2001:200 NICD1NICD1 Cell SignalingCell Signaling #4147#4147 Rabbit monoclonalRabbit monoclonal H, M, RH, M, R IB, IF, IHCIB, IF, IHC 1:1000
1:200
1:1000
1:200
  AbcamAbcam ab8925ab8925 RabbitRabbit M, H, RM, H, R IHC-P, IBIHC-P, IB 1:10001:1000 Notch-1Notch-1 Santa CruzSanta Cruz sc-373891sc-373891 Mouse monoclonal Mouse monoclonal H, M, RH, M, R IHCIHC 1: 1001:100 Hes1Hes1 AbcamAbcam ab71559ab71559 Rabbit polyclonalRabbit polyclonal H, M, RH, M, R IBIB 1:10001:1000 DLL1DLL1 InvitrogenInvitrogen PA5-19106PA5-19106 Goat polyclonalGoat polyclonal H, M, RH, M, R IB, FACS
Neutralizing
IB, FACS
Neutralizing
1:1000 1:100
20 μg/ml
1:1000 1:100
20 µg/ml
DLL3DLL3 InvitrogenInvitrogen PA5-26336PA5-26336 Rabbit polyclonalRabbit polyclonal H, M, RH, M, R IB, FACSIB, FACS 1:1000 1:1001:1000 1:100 DLL4DLL4 InvitrogenInvitrogen PA5-85931PA5-85931 Rabbit polyclonalRabbit polyclonal H, MH, M IB, FACSIB, FACS 1:1000 1:1001:1000 1:100 Jag1Jag1 Cell SignalingCell Signaling 26202620 Rabbit monoclonalRabbit monoclonal H, MH, M IB, FACSIB, FACS 1:1000 1:1001:1000 1:100 Jag2Jag2 Santa CruzSanta Cruz sc-515725sc-515725 Mouse monoclonal Mouse monoclonal H, M, RH, M, R IB, FACSIB, FACS 1:1000 1:1001:1000 1:100 beta-Actinbeta-Actin Santa CruzSanta Cruz sc-69879sc-69879 Mouse monoclonal Mouse monoclonal Broad speciesBroad species IBIB 1:10001:1000 α-Tubulinα-Tubulin SIGMASIGMA T5168T5168 Mouse monoclonal Mouse monoclonal M, RM,R IBIB 1:30001:3000 BAI1BAI1 R&D SystemsR&D Systems AF4969AF4969 Sheep polyclonalSheep polyclonal H, MH, M NeutralizingNeutralizing 8 μg/ml8 µg/ml WISP-1WISP-1 R&D SystemsR&D Systems MAB1680MAB1680 Rat monoclonalRat monoclonal MM NeutralizingNeutralizing 10 μg/ml10 µg/ml IgGIgG R&D SystemsR&D Systems MAB0061MAB0061 Rat monoclonalRat monoclonal MM NeutralizingNeutralizing 10 μg/ml10 µg/ml IgGIgG eBioscienceeBioscience 14-488814-4888 Armenian hamster monoclonalArmenian hamster monoclonal Broad speciesBroad species NeutralizingNeutralizing Integrin αvIntegrin αv eBioscienceeBioscience 14-051214-0512 Rat monoclonalRat monoclonal H, MH, M NeutralizingNeutralizing Integrin α5Integrin α5 eBioscienceeBioscience 14-049314-0493 Armenian hamster monoclonalArmenian hamster monoclonal M, RM,R NeutralizingNeutralizing 10 μg/ml(CAF)10 μg/ml (CAF) Integrin beta 1Integrin beta 1 eBioscienceeBioscience 16-029116-0291 Armenian hamster monoclonalArmenian hamster monoclonal M, RM,R NeutralizingNeutralizing 3 μg/ml(344SQ)3 µg/ml (344SQ) Integrin beta 3Integrin beta 3 eBioscienceeBioscience 11-061111-0611 Armenian hamster monoclonalArmenian hamster monoclonal M, RM,R NeutralizingNeutralizing Integrin beta 5Integrin beta 5 eBioscienceeBioscience 14-049714-0497 Mouse monoclonalMouse monoclonal Hm, H, MHm, H, M NeutralizingNeutralizing CD31CD31 AbcamAbcam ab7388ab7388 Rat monoclonalRat monoclonal MM Cell sortingCell sorting   CD45CD45 AbcamAbcam ab25386ab25386 Rat monoclonalRat monoclonal MM Cell sortingCell sorting   CD68CD68 AbcamAbcam ab53444ab53444 Rat monoclonalRat monoclonal MM Cell sortingCell sorting   CD326CD326 BD BioscienceBD Bioscience 552370552370 Rat monoclonalRat monoclonal MM Cell sortingCell sorting   Mouse IgG
(HRP)
Mouse IgG
(HRP)
GeneTexGeneTex GTX213111GTX213111 GoatGoat Not applicableNot applicable IBIB 1:50001:5000
Rabbit IgG (HRP)Rabbit IgG (HRP) GeneTexGeneTex GTX213110GTX213110 GoatGoat Not applicableNot applicable IBIB 1:50001:5000 Goat IgG
(HRP)
Goat IgG
(HRP)
Santa CruzSanta Cruz sc-2354sc-2354 MouseMouse Not applicableNot applicable IBIB 1:50001:5000
Mouse IgG
(Alexa 488)
Mouse IgG
(Alexa 488)
Thermo Fisher
Scientific
Thermo Fisher
Scientific
A11029A11029 GoatGoat Not applicableNot applicable FACS, IHCFACS, IHC 1:100
1:400
1:100
1:400
Mouse IgG
(Alexa 594)
Mouse IgG
(Alexa 594)
Thermo Fisher
Scientific
Thermo Fisher
Scientific
A11032A11032 GoatGoat Not applicableNot applicable IHCIHC 1:4001:400
Rabbit IgG
(Alexa 488)
Rabbit IgG
(Alexa 488)
Thermo Fisher
Scientific
Thermo Fisher
Scientific
A11034A11034 GoatGoat Not applicableNot applicable FACS, IF, IHCFACS, IF, IHC 1:100
1:400
1:100
1:400
Rabbit IgG
(Alexa 594)
Rabbit IgG
(Alexa 594)
Thermo Fisher
Scientific
Thermo Fisher
Scientific
A11012A11012 GoatGoat Not applicableNot applicable FACS. IF, IHCFACS. IF, IHC 1:100
1:400
1:100
1:400
Goat IgG
(Alexa 488)
Goat IgG
(Alexa 488)
Thermo Fisher
Scientific
Thermo Fisher
Scientific
A11055A11055 DonkeyDonkey Not applicableNot applicable FACSFACS 1:1001:100
Goat IgG
(Alexa 647)
Goat IgG
(Alexa 647)
Thermo Fisher
Scientific
Thermo Fisher
Scientific
A21447A21447 DonkeyDonkey Not applicableNot applicable FACSFACS 1:1001:100
Sheep IgG
(Alexa 594)
Sheep IgG
(Alexa 594)
Thermo Fisher
Scientific
Thermo Fisher
Scientific
A11016A11016 DonkeyDonkey Not applicableNot applicable IF, IHCIF, IHC 1:4001:400
* Abbreviation: IB-Immunoblot, IHC-Immunohistochemistry, IHC-P-Immunohisrochemistry-paraffin, IF-Immunofluorescence, H-human, M-mouse, R-rat, Rb-rabbit, P-pig, Mi-mink, Hm-hamster, Mk-monkey, Dm-drosophila melanogaster, Z-zebrafish, B-Bovine, Pg-pig, Ce-caenorhabditis elegans, F-frog* Abbreviation: IB-Immunoblot, IHC-Immunohistochemistry, IHC-P-Immunohisrochemistry-paraffin, IF-Immunofluorescence, H-human, M-mouse, R-rat, Rb-rabbit, P-pig, Mi-mink, Hm-hamster , Mk-monkey, Dm-drosophila melanogaster, Z-zebrafish, B-Bovine, Pg-pig, Ce-caenorhabditis elegans, F-frog

1-3. 암 관련 섬유화세포(Carcinoma-Associated Fibroblasts, CAFs)의 분리 및 세포 배양1-3. Isolation and cell culture of Carcinoma-Associated Fibroblasts (CAFs)

Kras-돌연변이체(KrasLA1) 마우스의 폐 종양으로부터, 섬유아세포-특이적 마커인 Thy1로 자기(magnetic)-활성화된 세포 분류를 사용하여, CAFs를 분리하였다. 이어서, CAFs를 10% 소 태아 혈청(FBS), 페니실린/스트렙토마이신(100 U/100 μg, Welgene, Gyeongsan, Korea), 2 mM L-글루타민(Welgene), 및 1 mM sodium pyruvate(Welgene)가 보충된 알파-MEM(Welgene)에서 배양하였다. 불멸화를 위해, CAFs를 리포펙터-EXT (AptaBio, Yongin, Korea)를 사용하여 TERT 플라스미드 (pCDH-3xFLAG-TERT, Addgene 51 플라스미드 # 51631)로 안정하게 형질감염시켰다. 실험에 사용된 1차 세포는 6회 미만으로 계대배양되었다. 인간 암 세포주는 ATCC(American Type Culture Collection, Manassas, VA, USA)로부터 얻었다. 344SQ 세포(폐선암 세포주, University of Texas MD Anderson Cancer Center, USA) 및 다양한 인간 암 세포주[A549(비소세포폐암 세포주), HCT116(대장암 세포주) 및 MCF-7(유방암 세포주)]를 10% FBS 및 페니실린/스트렙토마이신(100U/100 μg)이 보충된 RPMI 1640(Welgene)에서 유지시켰다.CAFs were isolated from lung tumors of Kras-mutant (Kras LA1 ) mice using magnetic-activated cell sorting with the fibroblast-specific marker Thy1. CAFs were then supplemented with 10% fetal bovine serum (FBS), penicillin/streptomycin (100 U/100 μg, Welgene, Gyeongsan, Korea), 2 mM L-glutamine (Welgene), and 1 mM sodium pyruvate (Welgene). cultured in alpha-MEM (Welgene). For immortalization, CAFs were stably transfected with TERT plasmid (pCDH-3xFLAG-TERT, Addgene 51 plasmid # 51631) using Lipofector-EXT (AptaBio, Yongin, Korea). Primary cells used in the experiments were subcultured less than 6 times. Human cancer cell lines were obtained from ATCC (American Type Culture Collection, Manassas, VA, USA). 344SQ cells (lung adenocarcinoma cell line, University of Texas MD Anderson Cancer Center, USA) and various human cancer cell lines [A549 (non-small cell lung cancer cell line), HCT116 (colorectal cancer cell line) and MCF-7 (breast cancer cell line)] were cultured in 10% FBS. and RPMI 1640 (Welgene) supplemented with penicillin/streptomycin (100 U/100 μg).

1-4. 세포 사멸 유도1-4. induction of apoptosis

암 상피 세포주를 15분 동안 254 nm 파장 자외선에 15분 간 노출시킨 후, RPMI-1640(10% FBS와 함께)로 37℃ 및 5% CO2에서 2시간 동안 배양하였다. Wright-Giemsa-염색된 샘플에 광 현미경을 사용하여 핵 형태를 평가한 결과, 조사된 세포가 세포사멸되었음을 확인하였다. 수회의 동결-해동 주기에 의해 용해(괴사) 암세포를 얻었다. 세포사멸 및 괴사는 annexin V-FITC/propidium iodide(BD Biosciences, San Jose, CA, USA) 염색에 이어서 FACSCalibur 시스템(ACEA Novocyte 3000, Agilent, Santa Clara, CA, USA)에서 유세포 분석에 의해 확인되었다.Cancer epithelial cell lines were exposed to 254 nm wavelength ultraviolet light for 15 minutes and then incubated with RPMI-1640 (with 10% FBS) at 37° C. and 5% CO 2 for 2 hours. As a result of evaluating nuclear morphology using a light microscope in the Wright-Giemsa-stained samples, it was confirmed that the irradiated cells were apoptotic. Lysed (necrotic) cancer cells were obtained by several freeze-thaw cycles. Apoptosis and necrosis were confirmed by annexin V-FITC/propidium iodide (BD Biosciences, San Jose, CA, USA) staining followed by flow cytometry on a FACSCalibur system (ACEA Novocyte 3000, Agilent, Santa Clara, CA, USA).

1-5. CAFs의 배양 및 CAF 조정배지의 제조1-5. Culture of CAFs and preparation of CAF conditioned medium

CAFs를 3 X 105 세포/ml로 플레이팅하고, 37℃ 및 5% CO2에서 적합한 배지로 배양시켰다. 밤새 인큐베이션한 후, 세포 자극 전에 24시간 동안 X-VIVO 10 배지(04-380Q, Lonza, Basel, Switzerland)로 혈청을 제거하였다. 자극을 위해, 배양 배지를 사멸화된 또는 괴사된 암 세포(9 X 105 세포/ml)를 함유하는 X-VIVO 10으로 대체하였다. 20시간 후, 상청액을 원심분리에 의해 수확하여, 표적 암 상피 세포(5 X 105 세포/ml) 또는 CAFs (3 X 105 세포/ml)의 자극을 위한 조정배지(CM)로 사용하였다. 생체내 실험을 위해, 조건배지를 필요시까지 -80℃에 보관하였다.CAFs were plated at 3 X 10 5 cells/ml and cultured in a suitable medium at 37°C and 5% CO 2 . After overnight incubation, serum was removed with X-VIVO 10 medium (04-380Q, Lonza, Basel, Switzerland) for 24 hours before cell stimulation. For stimulation, the culture medium was replaced with X-VIVO 10 containing killed or necrotic cancer cells (9 X 10 5 cells/ml). After 20 hours, the supernatant was harvested by centrifugation and used as a conditioned medium (CM) for stimulation of target cancer epithelial cells (5 X 10 5 cells/ml) or CAFs (3 X 10 5 cells/ml). For in vivo experiments, the conditioned medium was stored at -80°C until needed.

1-6. 이동 및 침윤 분석1-6. Migration and invasion assay

세포의 이동 및 침윤은 제조사의 지시에 따라 각각 30 μg/ml의 피브로넥틴과 300 μg/ml의 Matrigel 매트릭스로 코팅된 Transwell 챔버(Corning Inc, Corning, NY, USA)를 사용하여 시험하였다. 48시간 또는 24시간 동안 TGF-beta 1(10 ng/ml)의 부재 또는 존재 하에 CAFs로부터의 조정배지에서 사전 배양된 암 세포 또는 CAFs(이동 분석을 위해 1 X 105 세포/웰, 침윤 분석을 위해 1 X 105 세포/웰)를 상부 챔버에서 무혈청 RPMI와 함께 플레이팅하였고, 바닥 웰에서 10% FBS로 보충한 RPMI 1640로 16시간 이동시간 또는 24시간 침윤시간 동안 37℃에 두었다. 또한, CAFs에서 이동 및 침윤은 ApoSQ와 직접 노출 후 24시간 동안 TGF-beta 1(10 ng/ml) 자극 수행되었다. TGF-beta 1 자극 전에, CAFs는 사멸화된 또는 괴사된 암 세포에 직접 노출 후 X-VIVO로 세정하였다. 4% 파라포름알데히드에 고정시킨 후, 막의 상부 표면 상의 이동되지 않거나 침윤되지 않은 세포를 면봉으로 긁어내었다. 하면의 세포를 0.1% 크리스탈 바이올렛으로 염색하고 증류수로 세정하였고, 3개의 랜덤 현미경 시야를(10X 배율) 촬영하여 계수하였다.Cell migration and invasion were tested using Transwell chambers (Corning Inc, Corning, NY, USA) coated with 30 μg/ml of fibronectin and 300 μg/ml of Matrigel matrix, respectively, according to the manufacturer's instructions. Cancer cells or CAFs pre-cultured in conditioned medium from CAFs in the absence or presence of TGF-beta 1 (10 ng/ml) for 48 or 24 hours (1 X 10 5 cells/well for migration assay, invasion assay 1 X 10 5 cells/well) were plated with serum-free RPMI in the upper chamber, and placed in RPMI 1640 supplemented with 10% FBS in the bottom well at 37°C for 16 hour transfer time or 24 hour infiltration time. In addition, migration and invasion in CAFs were performed 24 h after direct exposure with ApoSQ and stimulation with TGF-beta 1 (10 ng/ml). Prior to TGF-beta 1 stimulation, CAFs were directly exposed to apoptotic or necrotic cancer cells and then washed X-VIVO. After fixation in 4% paraformaldehyde, immobile or non-infiltrated cells on the upper surface of the membrane were scraped off with a cotton swab. Cells on the lower surface were stained with 0.1% crystal violet, washed with distilled water, and three random microscope fields were photographed (10X magnification) and counted.

1-7. 웨스턴 블랏팅 분석1-7. Western blotting analysis

전체 세포 추출물을 사용하여 표준화된 웨스턴 블랏을 수행하였다. 전체 세포 추출물은 사멸화된 세포 또는 조정배지와 공동배양한 CAFs 또는 표적 암 세포로부터 제조하였다. 세포를 수거하고, 빙냉 포스페이트-완충 염 수(PBS)로 세척하고, 방사성 면역침전 분석(RIPA) 완충액(10 mM Tris, pH 7.2, 150 mM NaCl, 1% Nonidet P-40, 0.5% 나트륨 데옥시콜레이트, 0.1% SDS, 1.0% Triton X-100, 5 mM EDTA)에서 30분 동안 얼음 상에서 프로테아제 억제제로 용해시켰다. 동일한 양의 단백질을 습식 전사 시스템(Bio-Rad Laboratories)을 사용하여 SDS- PAGE 겔(#161-0158, Bio-Rad Laboratories, Herc-Res, CA, USA) 상에서 전개하고 니트로 셀룰로오스 막(10600001, GE Healthcare Life Science)으로 트랜스퍼하였다. 5% BSA-TBST 또는 5% milk-TBST로 1시간 동안 블락킹한 후, 블랏을 1차 항체와 하룻밤 동안 인큐베이션한 다음, 37℃에서 2차 항체와 1시간 동안 인큐베이션하였다. 오디세이 이미지 분석 시스템(Licor Biosciences, Lincoln, Nebraska)을 사용하여 정량화하였다.A standardized Western blot was performed using whole cell extracts. Whole cell extracts were prepared from killed cells or CAFs or target cancer cells co-cultured with conditioned media. Cells were harvested, washed with ice-cold phosphate-buffered saline (PBS), and washed in radioimmunoprecipitation assay (RIPA) buffer (10 mM Tris, pH 7.2, 150 mM NaCl, 1% Nonidet P-40, 0.5% sodium deoxygenate). cholate, 0.1% SDS, 1.0% Triton X-100, 5 mM EDTA) for 30 min on ice with protease inhibitors. Equal amounts of protein were run on SDS-PAGE gels (#161-0158, Bio-Rad Laboratories, Herc-Res, CA, USA) using a wet transfer system (Bio-Rad Laboratories) and transferred to nitrocellulose membranes (10600001, GE Healthcare Life Science). After blocking with 5% BSA-TBST or 5% milk-TBST for 1 hour, blots were incubated with primary antibody overnight, followed by incubation with secondary antibody for 1 hour at 37°C. Quantification was performed using the Odyssey image analysis system (Licor Biosciences, Lincoln, Nebraska).

1-8. 정량적 실시간 중합효소 연쇄 반응(qRT-PCR)1-8. Quantitative real-time polymerase chain reaction (qRT-PCR)

총 RNA는 분리된 세포로부터 TRIzol 시약(RNAiso plus, Takara Bio Inc., Kusatsu, Japan)을 이용해 추출하였다. ReverTra AceTM qPCR RT Kit와 Master Mix(Toyobo, Osaka, Japan)를 이용해 역전사를 시킨 뒤, SYBR Green-based qRT-PCR을 QuantStudioTM 3 Real-Time PCR 시스템(Applied Biosystems, Foster City, CA, USA)을 이용해 수행하였다. mRNA는 6-웰 플레이트 상에서 80% 컨플루언스로 성장된 세포로부터 추출하였고, 실시간 PCR 시스템(StepOnePlus 시스템, Applied Biosystems, Foster City, CA)을 사용하여 정량화되었다. mRNA 수준을 Hprt mRNA로 정규화하고, 대조군에 대한 발현의 배수 변화로 리포팅하였다. 타겟 유전자에 대해 사용한 프라이머 서열은 다음과 같다(표 2 참조).Total RNA was extracted from the isolated cells using TRIzol reagent (RNAiso plus, Takara Bio Inc., Kusatsu, Japan). After reverse transcription was performed using the ReverTra Ace TM qPCR RT Kit and Master Mix (Toyobo, Osaka, Japan), SYBR Green-based qRT-PCR was performed using the QuantStudio TM 3 Real-Time PCR System (Applied Biosystems, Foster City, CA, USA) was performed using mRNA was extracted from cells grown to 80% confluence on 6-well plates and quantified using a real-time PCR system (StepOnePlus system, Applied Biosystems, Foster City, Calif.). mRNA levels were normalized to Hprt mRNA and reported as fold change in expression relative to control. Primer sequences used for target genes are as follows (see Table 2).

murine genemurine genes forward (5' → 3')forward (5' → 3') reverse (5' → 3')reverse (5' → 3') Acta2Acta2 CCCAGATTATGTTTGAGACCTTCCCCAGATTATGTTTGAGACCTTC ATCTCCAGAGTCCAGCACAATACATCTCCAGAGTCCAGCACAATAC Cav-1Cav-1 CACACCAAGGAGATTGACCTGGCACACCAAGGAGATTGACCTGG CCTTCCAGATGCCGTCGAAACTCCTTCCAGATGCCGTCGAAACT Ccn1Ccn1 TTTACAGTTGGGCTGGAAGCTTTACAGTTGGGCTGGAAGC CACCGCTCTGAAAGGGATCTCACCGCTCTGAAAGGGATCT Ccn2Ccn2 GCTTGGCGATTTTAGGTGTCGCTTGGCGATTTTAGGTGTC CAGACTGGAGAAGCAGAGCCCAGACTGGAGAAGCAGAGCC Ccn3Ccn3 GTCACCAACAGGAATCGCCAGTGTCACCAACAGGAATCGCCAGT GTAGGTGGATGGCTTTCAGGGAGTAGGTGGATGGCTTTCAGGGA Ccn5Ccn5 TGTGTGACCAGGCAGTGATGCATGTGTGACCAGGCAGTGATGCA CAGGCTGTGCTCCAGTTTGGACCAGGCTGTGCTCCAGTTTGGAC Ccn6Ccn6 CACCTGTAACGAAGCCGAGATCCACCTGTAACGAAGCCGAGATC ACTCACATCCAACTGCCACAAGAACTCACATCCAACTGCCACAAGA Col1a1Col1a1 CAAGAAGACATCCCTGAAGTCCAAGAAGACATCCCTGAAGTC ACAGTCCAGTTCTTCATTGCACAGTCCAGTTCTTCATTGC Cxcl12Cxcl12 TGCATCAGTGACGGTAAACCATGCATCAGTGACGGTAAACCA CACAGTTTGGAGTGTTGAGGATCACAGTTTGGAGTGTTGAGGAT Cxcl14Cxcl14 TACCCACACTGCGAGGAGAAGATACCCACACTGCGAGGAGAAGA CGCTTCTCGTTCCAGGCATTGTCGCTTCTCGTTCCAGGCATTGT Dll1Dll1 CCGGCTGAAGCTACAGAAACCCGGCTGAAGCTACAGAAAC AGCCCCAATGATGCTAACAGAGCCCCAATGATGCTAACAG Dll3Dll3 CTGGTGTCTTCGAGCTACAAATCTGGTGTCTTCGAGCTACAAAT TGCTCCGTATAGACCGGGACTGCTCCGTATAGACCGGGAC Dll4Dll4 CCTCTCGAACTTGGACTTGCCCTCTCGAACTTGGACTTGC TGGAAATACAGATGCCCACATGGAAATACAGATGCCCACA FapFap GTCACCTGATCGGCAATTTGTGTCACCTGATCGGCAATTTGT CCCCATTCTGAAGGTCGTAGATCCCCATTCTGAAGGTCGTAGAT Fgf1Fgf1 CCAAGGAAACGTCCACAGTCAGCCAAGGAAACGTCCACAGTCAG ACGGCTGAAGACATCCTGTCTCACGGCTGAAGACATCCTGTCTC FnFn CACGATGCGGGTCACTTGCACGATGCGGGTCACTTG CTGCAACGTCCTCTTCATTCTTCCTGCAACGTCCTCTTCATTCTTC Hes1Hes1 CTACCCCAGCCAGTGTCAACCTACCCCAGCCAGTGTCAAC CCTTCGCCTCTTCTCCATGACCTTCGCCTCTTCTCCATGA Hes5Hes5 ATCAACAGCAGCATAGAGCAGATCAACAGCAGCATAGAGCAG CGAAGGCTTTGCTGTGTTTCACGAAGGCTTTGCTGTGTTTCA Hey1Hey1 ACATCGTCCCAGGTTTTGGCACATCGTCCCAGGTTTTGGC GCTTCTCAAAGGCACTGGGTGCTTCTCAAAGGCACTGGGT Hey2Hey2 TCCAGGCTACAGGGGGTAAATCCAGGCTACAGGGGGTAAA CAAGGCCTTCCACTGAGCTTCAAGGCCTTCCACTGAGCTT HgfHgf CCTGACACCACTTGGGAGTACCTGACACCACTTGGGAGTA CTTCTCCTTGGCCTTGAATGCTTCTCCTTGGCCTTGAATG HprtHprt CCAGTGTCAATTATATCTTCAACCCAGTGTCAATTATATCTTCAAC CAGACTGAAGAGCTACTGTAATGCAGACTGAAGAGCTACTGTAATG Itg beta 1Itg beta 1 CTCCAGAAGGTGGCTTTGATGCCTCCAGAAGGTGGCTTTGATGC GTGAAACCCAGCATCCGTGGAAGTGAAACCCAGCATCCGTGGAA Jag1Jag1 GGAAGTGGAGGAGGATGACAGGAAGTGGAGGAGGATGACA GTCCAGTTCGGGTGTTTTGTGTCCAGTTCGGGTGTTTTGT Jag2Jag2 TCCGAGTACGCTGTGATGAGTCCGAGTACGCTGTGATGAG GGCTTCTTTGCATTCTTTGCGGCTTTCTTTGCATTCTTTGC Mmp1aMmp1a AGGAAGGCGATATTGTGCTCTCCAGGAAGGCGATATTGTGCTCTCC TGGCTGGAAAGTGTGAGCAAGCTGGCTGGAAAGTGTGAGCAAGC Mmp2Mmp2 CAAGGATGGACTCCTGGCACATCAAGGATGGACTCCTGGCACAT TACTCGCCATCAGCGTTCCCATTACTCGCCATCAGCGTTCCCAT Mmp3Mmp3 CTCTGGAACCTGAGACATCACCCTCTGGAACCTGAGACATCACC AGGAGTCCTGAGAGATTTGCGCAGGAGTCCTGAGAGATTTGCGC Mmp9Mmp9 TGCCCAGCGACCACAACTCTGCCCAGCGACCACAACTC CGGACCCGAAGC GGACATTCGGACCCGAAGC GGACATT Mmp12Mmp12 CACACTTCCCAGGAATCAAGCCCACACTTCCCAGGAATCAAGCC TTTGGTGACACGACGGAACAGGTTTGGTGACACGACGGAACAGG Mmp14Mmp14 GTGAGCGTTGTGTGTGGGTAGTGAGCGTTGTGTGTGGGTA CCCAAGGCA GCAACTTCAGCCCAAGGCA GCAACTTCAG Notch1Notch1 GCCGCAAGAGGCTTGAGATGCCGCAAGAGGCTTGAGAT GGAGTCCTGGCATCGTTGGGGAGTCCTGGCATCGTTGG PdgfrαPdgfrα CTCTTGGAGATAGACTCCGTAGGCTCTTGGAGATAGACTCCGTAGG ACTTCTCTTCCTGCGAATGGACTTCTCTCCTGCGAATGG Pdgfr betaPdgfr beta TGGCCTCTGAGGACTAAAGCTGGCCTCTGAGGACTAAAGC AACAGAAGACAGCGAGGTGGAACAGAAGACAGCGAGGTGG Spp1Spp1 GCTTGGCTTATGGACTGAGGTCGCTTGGCTTATGGACTGAGGTC CCTTAGACTCACCGCTCTTCATGCCTTAGACTCACCGCTCTTCATG Tgf betaTgf beta TGCCGTACAACTCCAGTGACTGCCGTACAACTCCAGTGAC TGGAGCAACATGTGGAACTCTGGAGCAACATGTGGAACTC TncTnc CCATCAGTACCACGGCTACCCCATCAGTACCACGGCTACC CCCTTCATCAGCAGTCCAGGCCCTTCATCAGCAGTCCAGG VegfaVegfa GTACCTCCACCATGCCAAGTGTACCTCCACCATGCCAAGT TCACATCTGCAAGTACGTTCGTCACATCTGCAAGTACGTTCG Wisp-1(ccn4)Wisp-1 (ccn4) CAATAGGAGTGTGTGCACAGGTCAATAGGAGTGTGTGCACAGGT TACCTGCAGTTGGGTTGGAAGTACCTGCAGTTGGGTTGGAAG

1-9. 사이토카인 어레이1-9. cytokine array

사이토카인 어레이는 제조사의 지시에 따라 CAF CM, ApoSQ-CAF CM 및 ApoSQ CM에서 Proteome Profiler Mouse XL 사이토카인 어레이 키트(#ARY028, R&D Systems, USA)를 사용하여 수행되었다. 어레이 멤브레인을 조정배지와 함께 밤새 인큐베이션 하고, 비오티닐화된 검출 항체 및 스트렙타비딘-호스래디시 퍼옥시다제를 사용하여 막-결합된 사이토카인을 검출하였다. 사이토카인 스폿의 픽셀 밀도를 이미지 J 소프트웨어(NIH, Bethesda, Maryland, USA; http://rsb)를 사용하여 분석하였다. info.nih.gov/ij/).Cytokine arrays were performed using the Proteome Profiler Mouse XL Cytokine Array Kit (#ARY028, R&D Systems, USA) in CAF CM, ApoSQ-CAF CM and ApoSQ CM according to the manufacturer's instructions. Array membranes were incubated with conditioned medium overnight and membrane-bound cytokines were detected using a biotinylated detection antibody and streptavidin-horseradish peroxidase. Pixel density of cytokine spots was analyzed using Image J software (NIH, Bethesda, Maryland, USA; http://rsb). info.nih.gov/ij/).

1-10. 면역형광법1-10. immunofluorescence

컨플루언트될 때까지 유리 커버슬립 상에서 성장한 CAFs를 실온에서 8분 동안 4% 파라포름알데히드 용액으로 고정하였다. 파라핀 포매 종양의 염색을 위해, 포르말린 고정을 실온에서 30분 동안 수행하였고, IF-Wash 완충액(PBS 내 0.05% NaN3, 0.1% BSA, 0.2% Triton X-100 및 0.05% Tween-20)을 사용하였다. 고정 후, 샘플을 각각 5분 동안 세척 완충액으로 3회 세척하고, PBS 내 0.5% Triton X-100(Sigma-Aldrich)으로 실온에서 5분간 투과시켰다. 마우스 IgG 차단 시약의 존재 또는 부재 하에서 PBS 내 5% BSA를 각각 면역세포화학 및 면역조직화학에 사용하였다. 1시간 후에, 타겟 단백질이 각각의 1차 항체에 의해 18시간 동안 4℃ 하에서 포착되었다. 포착된 단백질은 형광이 결합된 IgG에 의해 암실에서 1시간 동안 관측되었다. 염색 후에, 모든 슬라이드는 DAPI를(Vector Laboratories, Burlingame, CA)를 함유하는 Vectashield Mounting 배지(Vector Laboratories, Burlingame, CA, USA)와 함께 고정되었고, 공초점 현미경(LSM5 PASCAL, Carl Zeiss, Jena, Germany) 또는 형광 현미경(Eclipse Ti2-U, Nikon, Tokyo, Japan)을 통해 관측되었다.CAFs grown on glass coverslips until confluent were fixed with a 4% paraformaldehyde solution for 8 min at room temperature. For staining of paraffin-embedded tumors, formalin fixation was performed for 30 min at room temperature and IF-Wash buffer (0.05% NaN3, 0.1% BSA, 0.2% Triton X-100 and 0.05% Tween-20 in PBS) was used. . After fixation, samples were washed three times with wash buffer for 5 minutes each and permeabilized with 0.5% Triton X-100 (Sigma-Aldrich) in PBS for 5 minutes at room temperature. 5% BSA in PBS with or without mouse IgG blocking reagent was used for immunocytochemistry and immunohistochemistry, respectively. After 1 hour, target proteins were captured by each primary antibody for 18 hours at 4°C. Captured proteins were observed for 1 hour in the dark by fluorescence-coupled IgG. After staining, all slides were fixed with Vectashield Mounting medium (Vector Laboratories, Burlingame, CA, USA) containing DAPI (Vector Laboratories, Burlingame, CA, USA) and confocal microscopy (LSM5 PASCAL, Carl Zeiss, Jena, Germany). ) or observed through a fluorescence microscope (Eclipse Ti2-U, Nikon, Tokyo, Japan).

1-11. ELISA1-11. ELISA

조정배지 및 혈청 중의 WISP-1 및 LIF를 제조사의 지시에 따라 ELISA 키트(R&D Systems)를 사용하여 측정하였다. 혈청 사이토카인 정량을 위해, 혈액을 천자를 통해 마우스로부터 수집하고, 혈청을 1600× g에서 5분 동안 4℃에서 원심분리하여 분리하였다.WISP-1 and LIF in conditioned media and serum were measured using ELISA kits (R&D Systems) according to the manufacturer's instructions. For serum cytokine quantification, blood was collected from mice via puncture, and serum was isolated by centrifugation at 1600×g for 5 minutes at 4° C.

1-12. 일시적 형질감염 및 루시퍼라제 활성 분석1-12. Transient transfection and luciferase activity assay

CAFs를 제조사의 지시에 따라 형질감염 시약(Lipofectamin RNAi MAX; Invitrogen, Carlsbad, CA, USA)을 사용하여 WISP-1(Bioneer Inc, Daejeon, Korea), LIF(Bioneer), Notch1(Bioneer) 또는 BAI1(Dharmacon. CO, USA)을 특이적으로 타겟하는 siRNA 또는 대조군 siRNA(Bioneer, Dharmacon)와 함께 100 nM의 최종 농도에서 일시적으로 형질감염시켰다. 밤새 형질감염 시킨 후에, 세포를 적합한 배지에서 24시간 동안 배양하고 ApoSQ 세포로 자극하였다. 344SQ 세포는 UV를 조사시키기 전에 Dll1에 특이적인 siRNA(D-050912-01-0020, Dharmacon) 또는 대조군 siRNA와 함께 일시적으로 형질감염시켰다. 유전자를 표적화하는데 사용된 siRNA 서열은 다음과 같다(표 3 참조).CAFs were transfected with WISP-1 (Bioneer Inc, Daejeon, Korea), LIF (Bioneer), Notch1 (Bioneer) or BAI1 ( Dharmacon. CO, USA) was transiently transfected with either specifically targeting siRNA or control siRNA (Bioneer, Dharmacon) at a final concentration of 100 nM. After overnight transfection, cells were cultured in suitable media for 24 hours and stimulated with ApoSQ cells. 344SQ cells were transiently transfected with Dll1-specific siRNA (D-050912-01-0020, Dharmacon) or control siRNA before UV irradiation. The siRNA sequences used to target the genes are as follows (see Table 3).

sensesense antisenseantisense WISP-1WISP-1 5'-GGAAUCCUAACGAUAUCUU-3' 5'-GGAAUCCUAACGAUAUCUU-3' 5'-AAGAUAUCGUUAGGAUUCC-3' 5′-AAGAUAUCGUUAGGAUUCC-3 LIFLIF 5'-CGACCACUCUGACAAAGAA-3'5′-CGACCACUCUGACAAAGAA-3′ 5'-UUCUUUGUCAGAGUGGUCG-3'5'-UUCUUUGUCAGAGUGGUCG-3' Notch1Notch1 5'-CCCUUUGAGUCUUCAUACA-3'5′-CCCUUUGAGUCUUCAUACA-3′ 5'-UGUAUGAAGACUCAAAGGG-3'5′-UGUAUGAAGACUCAAAGGG-3 Dll1Dll1 5'-GCACGGACCUUGAGGACAG-3'5′-GCACGGACCUUGAGGACAG-3′ 5'-CUGUCCUCAAGGUCCGUGC-3'5'-CUGUCCUCAAGGUCCGUGC-3' BAI1BAI1 5'-GUGUCAUCCUCGUACAGU-3'5'-GUGUCAUCCUCGUACAGU-3' 5'-ACUGUACGAGGAUGACAC-3'5'-ACUGUACGAGGAUGACAC-3'

유전자 과발현 실험은 CAFs 0.5 X 106 세포/ml를 60-mm 접시에 24시간 동안 플레이팅하여 수행하였다. 마우스 WISP-1 유전자 ORF cRNA 클론 발현 플라스미드(MG5A0255-CH, Sino Biological, North Wales, PA)를 이용하여, WISP-1 과발현을 분석하였다. CAFs를 제조사의 설명서에 따라 Lipofectamine 2000(#12566014; Thermo Fisher Scientific, Waltham, MA) 시약을 사용하여 2.5 μg/웰의 플라스미드 DNA로 형질감염시켰다. 세포는 형질감염 후 성장 배지에서 배양 24시간 후 추가 실험을 위해 준비되었다. 모의 형질감염된 세포를 WISP-1 형질감염된 세포와 동일한 방식으로 대조 군 벡터, pcDNA3으로 형질감염시켰다. BAI1 과발현을 위해, 제조사의 지시에 따라 Lipofectamine 2000 시약(Thermo Fisher Scientific)을 이용하여 48시간 동안 2.0 μg의 pEBB 공벡터 또는 pEBB-BAI-Flag 플라스미드(Gwangju Institute of Science and Technology, Gwangju, Korea)로 CAF를 형질감염시켰다. 플라스미드의 효능을 확인하기 위해 세포 용해물을 WISP-1 및 BAI1 유전자 및 단백질 발현에 대해 측정하였다.Gene overexpression experiments were performed by plating 0.5 X 10 6 cells/ml of CAFs in a 60-mm dish for 24 hours. WISP-1 overexpression was analyzed using a mouse WISP-1 gene ORF cRNA clone expression plasmid (MG5A0255-CH, Sino Biological, North Wales, PA). CAFs were transfected with 2.5 μg/well of plasmid DNA using Lipofectamine 2000 (#12566014; Thermo Fisher Scientific, Waltham, Mass.) reagent according to the manufacturer's instructions. Cells were ready for further experiments after 24 hours of culture in growth media after transfection. Mock-transfected cells were transfected with the control vector, pcDNA3, in the same way as WISP-1 transfected cells. For BAI1 overexpression, 2.0 μg of pEBB empty vector or pEBB-BAI-Flag plasmid (Gwangju Institute of Science and Technology, Gwangju, Korea) for 48 h using Lipofectamine 2000 reagent (Thermo Fisher Scientific) according to the manufacturer's instructions. CAFs were transfected. Cell lysates were measured for WISP-1 and BAI1 gene and protein expression to confirm the potency of the plasmids.

루시퍼라제 분석을 위해, CAFs를 리포펙티민 LTX 형질감염 시약 및 PLUS 시약(Life Technologies, Darmstadt, Germany)을 사용하여 형질감염시켰다. 세포를 800 ng/웰 4×CSL 루시퍼라제 플라스미드 (#41726 Addgene, Wattown, MA)로 형질감염시켜 Notch 경로 활성화에 대한 반응으로 루시퍼라제를 생성시키고, 200 ng/웰 Renilla 루시퍼라제 플라스미드로 형질감염시켰다. 루시퍼라제 활성은 Renilla 활성으로 정규화하고, 값을 PGL2-대조군 벡터에 대한 배수 변화로서 리포팅하였다. 모든 조건을 각각의 독립적인 실험에 대해 3회 수행하였고, 루시퍼라제 실험은 Dual-Luciferase 분석 시스템(Promega, Madison, WI, USA)을 통해 수행되었다.For the luciferase assay, CAFs were transfected using Lipofectimin LTX Transfection Reagent and PLUS Reagent (Life Technologies, Darmstadt, Germany). Cells were transfected with 800 ng/well 4×CSL luciferase plasmid (#41726 Addgene, Wattown, MA) to produce luciferase in response to Notch pathway activation and transfected with 200 ng/well Renilla luciferase plasmid . Luciferase activity was normalized to Renilla activity and values were reported as fold change relative to the PGL2-control vector. All conditions were performed in triplicate for each independent experiment, and luciferase experiments were performed using the Dual-Luciferase Assay System (Promega, Madison, WI, USA).

1-13. 조정배지 내 WISP-1의 중화1-13. Neutralization of WISP-1 in conditioned media

CAFs로부터의 조정배지를 10 μg/ml 마우스 항-마우스 WISP-1 중화 항체(R&D Systems) 또는 10 μg/ml IgG 이소타입 대조군(R&D Systems)과 함께 2시간 동안 인큐베이션하였다. 항-WISP-1 항체의 중화 효과는 WISP-1 ELISA를 통해 측정하였다.Conditioned medium from CAFs was incubated for 2 hours with 10 μg/ml mouse anti-mouse WISP-1 neutralizing antibody (R&D Systems) or 10 μg/ml IgG isotype control (R&D Systems). The neutralizing effect of anti-WISP-1 antibodies was measured by WISP-1 ELISA.

1-14. 유세포 분석1-14. flow cytometry

Notch 리간드 염색을 위해. UV 조사된 사멸화된 암 세포 또는 살아있는 암 세포를 FACS 완충액(PBS 내 0.1% BSA 및 0.1% sodium azide) 내에서 항-Dll1, 항-Dll3, 항-Dll4, 항-Jag1 또는 항-Jag2(1:100)와 함께 실온에서 30분 동안 인큐베이션하였다. 이어서, 세포를 Alexa 488- 또는 594- 로 표지된 2차 항체와 함께 FACS 완충액에서 실온에서 30분 간 인큐베이션하였다. 인큐베이션 후, 세포는 FACS 완충액으로 3번 세척되었고, Notch 리간드의 발현량은 유세포 분석기(ACEA Novocyte 3000, Agilent, Santa Clara, CA, USA)를 이용해 분석되었다. 데이터는 Novoexpress 소프트웨어(Agilent)를 사용하여 분석하였다.for Notch ligand staining. UV-irradiated killed cancer cells or live cancer cells were cultured in FACS buffer (0.1% BSA and 0.1% sodium azide in PBS) with anti-Dll1, anti-Dll3, anti-Dll4, anti-Jag1 or anti-Jag2 (1 :100) for 30 minutes at room temperature. Cells were then incubated for 30 minutes at room temperature in FACS buffer with Alexa 488- or 594-labeled secondary antibodies. After incubation, the cells were washed three times with FACS buffer, and the expression level of Notch ligand was analyzed using a flow cytometer (ACEA Novocyte 3000, Agilent, Santa Clara, CA, USA). Data were analyzed using Novoexpress software (Agilent).

1-15. 탐식 작용 분석1-15. phagocytosis assay

사멸화된 암세포 탐식 작용 분석은 상기 유세포 분석 및 면역형광법을 통해 수행되었다. 먼저 CAFs를 PKH26(red)으로 염색한 이후, PKH67(green)로 표지된 사멸화된 344SQ 세포와 1:3 비율로 24시간 동안 공동배양하였다. 세척 후 CAFs에 의한 탐식 비율은 두 색의 유세포 분석으로 측정되었다. 면역형광법을 위해, PKH67로 표지된 사멸화된 344SQ 세포는 CAFs와 24시간 동안 공동배양하고 세척한 후, CAFs를 3.7%의 w/v 파라포름알데히드로 고정시켰고, 0.1%의 Triton X-100으로 15분 간 투과시켰다. F-액틴은 제조사의 지시에 따라 rhodamine phalloidin(Invitrogen)으로 염색되었다. 이미지는 공초점 현미경(LSM5 PASCAL; Carl Zeiss, Jena, Germany)으로 캡쳐하였고, 탐식 지수는 (사멸화된 세포/200 총 CAFs) x 100의 수식으로 계산하였다.Phagocytosis analysis of killed cancer cells was performed through the flow cytometry and immunofluorescence method. First, CAFs were stained with PKH26 (red), and then co-cultured with apoptotic 344SQ cells labeled with PKH67 (green) at a ratio of 1:3 for 24 hours. After washing, the percentage of phagocytosis by CAFs was measured by two-color flow cytometry. For immunofluorescence, killed 344SQ cells labeled with PKH67 were co-cultured with CAFs for 24 hours, washed, and CAFs were fixed with 3.7% w/v paraformaldehyde and 0.1% Triton X-100. Permeabilized for 15 minutes. F-actin was stained with rhodamine phalloidin (Invitrogen) according to the manufacturer's instructions. Images were captured with a confocal microscope (LSM5 PASCAL; Carl Zeiss, Jena, Germany), and the phagocytic index was calculated by the formula (killed cells/200 total CAFs) x 100.

1-16. Rac1 활성 분석1-16. Rac1 activity assay

CAFs를 ApoSQ와 1:3 비율로 24시간 동안 공동배양 하기 전에, DAPT(10 또는 20 μM)로 2시간 동안 사전 인큐베이션시키거나, Notch1 또는 대조군 siRNA로 24시간 동안 형질감염시켰다. Rac1 활성은 제조사의 지시에 따라 G-LISA Rac1 활성 분석 키트(Cytoskeleton, Denver, CO, USA)를 통해 측정하였다. 즉, 동일한 양의 단백질을 포함한 세포 용해물을 Rac1-GTP 결합 플레이트에 첨가하였고 4℃에서 30분 간 인큐베이션시켰다. 플레이트를 0.05%의 Tween 20을 포함한 PBS로 세척하였고, 항원제시 완충액으로 반응시켰다. 세척 이후, 플레이트를 항-Rac1 1차 및 2차 항체로 반응시켰다. 반응은 horseradish peroxidase 검측 시약을 통해 관측되었고, 정지 용액을 통해 정지되었다. 플레이트를 분광 광도계를 이용하여 490 nm에서의 흡광도를 측정하였다.CAFs were pre-incubated for 2 hours with DAPT (10 or 20 μM) or transfected with Notch1 or control siRNA for 24 hours before co-culture with ApoSQ at a 1:3 ratio for 24 hours. Rac1 activity was measured using the G-LISA Rac1 activity assay kit (Cytoskeleton, Denver, CO, USA) according to the manufacturer's instructions. That is, cell lysate containing the same amount of protein was added to the Rac1-GTP binding plate and incubated at 4° C. for 30 minutes. The plate was washed with PBS containing 0.05% of Tween 20 and reacted with an antigen presentation buffer. After washing, the plates were reacted with anti-Racl primary and secondary antibodies. The reaction was observed with horseradish peroxidase detection reagent and stopped with stop solution. The plate was measured for absorbance at 490 nm using a spectrophotometer.

1-17. 마우스 실험1-17. mouse experiment

Ewha Medical Research Institute의 동물 케어 위원회로부터 하기 실험 프로토콜을 승인받았다(EUM19-0430). 마우스는 연구소 동물의 관리 및 사용을 위한 국립 보건원(National Institute of Health; NIH) 가이드에 따라 관리 및 취급되었다. 동계(syngeneic) 종양 실험을 이용한 폐암 전이 연구를 공지의 방법으로 수행하였다. 요약하면, 344SQ 세포(마우스 당 PBS 100 μl 중 1x106 세포)를 우측 후방 측면에서 동계(syngeneic)(129/Sv) 마우스 내로 피하 주사하였다. 제 1 주사 2일 후, 제 2 주사를 동일한 병변에서 1x107 개의 사멸화된 344SQ 세포의 존재 또는 부재하에서 100 μl의 PBS와 함께 주사하였다.The following experimental protocol was approved by the Animal Care Committee of Ewha Medical Research Institute (EUM19-0430). Mice were cared for and handled according to the National Institute of Health (NIH) Guide for the Care and Use of Laboratory Animals. Lung cancer metastasis studies using syngeneic tumor experiments were performed by a known method. Briefly, 344SQ cells (1×10 6 cells in 100 μl PBS per mouse) were injected subcutaneously into syngeneic (129/Sv) mice in the right posterior flank. Two days after the first injection, a second injection was given with 100 μl of PBS in the presence or absence of 1×10 7 killed 344SQ cells in the same lesion.

Notch1 억제 실험을 위해, 선택적 Notch1 억제제 LY3039478(8 mg/kg)을 ApoSQ 주사 1일 전에 6주간 주 3회로 15% 설탕 젤 vehicle과 함께 구강 투여하였다. 조정배지 실험을 위해, 344SQ 주사 2일 후에, 중화 항-WISP-1 항체(10 μg/ml) 또는 이소타입 IgG의 존재 또는 부재하에 CAFs로부터 유래된 조정배지(마우스당 100 μl)를 1주 3회 종양내 주사를 통해 투여하였다. 마우스를 종양 성장에 대해 매일 모니터링하고 주사 6주 후에 희생시켰다. 부검을 수행하여, 피하 종양 덩어리의 직경 및 중량, 폐 전이 상태(노듈의 수 또는 발생률) 및 포르말린 고정, 파라핀-포매, 면역형광-염색된 원발성 종양의 조직학적 평가를 조사하였다.For the Notch1 inhibition experiment, the selective Notch1 inhibitor LY3039478 (8 mg/kg) was orally administered with 15% sugar gel vehicle three times a week for 6 weeks one day before ApoSQ injection. For conditioned media experiments, 2 days after injection of 344SQ, conditioned media derived from CAFs (100 μl per mouse) with or without neutralizing anti-WISP-1 antibody (10 μg/ml) or isotype IgG was added for 1 week 3 It was administered via intratumor injection. Mice were monitored daily for tumor growth and sacrificed 6 weeks after injection. A necropsy was performed to examine the diameter and weight of the subcutaneous tumor mass, the status of lung metastases (number or incidence of nodules), and histological evaluation of formalin-fixed, paraffin-embedded, immunofluorescence-stained primary tumors.

1-18. 원발성 종양으로부터 Thy11-18. Thy1 from primary tumor ++ CAFs, CD326 CAFs, CD326 ++ 종양 세포 및 CD11b Tumor cells and CD11b ++ 종양 관련 대식세포의 분리 Isolation of tumor-associated macrophages

전이성 마우스 모델의 원발성 종양으로부터 단일 세포 현탁액을 획득하기 위해, 고체의 신선한 종양 조직을 제조자의 지시에 따라 MACSTM Dissociator(모두 Miltenui Biotec Inc., San Diego, CA)와 조합된 종양 해리 키트를 사용하여 해리시켰다. 간략하게, 각 마우스의 종양 조직을 효소 분해용 키트의 효소 혼합물을 함유하는 MACS C 튜브(Miltenui Biotec)로 옮겼다. 해리 후, 각 조직 균질액을 70- 및 40- μm의 살균한 나일론 그물로 여과하고 적혈구 세포를 적혈구 용해 완충액으로 용해시켰다. 300 g에서 10분 간 원심분리한 후, 잔여 세포는 비 CAF 세포를 선택하기 위해 항-CD45(Abcam, Cambridge, UK), 항-CD68(Abcam), 항-CD31(Abcam) 및 항-CD326(BD Bioscience, San Diego, CA, USA)으로 표지되었다. 표지된 세포를 총 세포 현탁액으로부터 사전 세척된 DynabeadsTM(Invitrogen)을 이용해 감소시켰고, PureProteomeTM 자성 스탠드(Millipore, Billerica, MA, USA)를 이용해 자성 구슬로부터 분리시켰다. 표지되지 않은 세포 상층액에서는, 제조사의 지시에 따라 CD90.2 MicroBeads와 MACS MS column(Miltenyi Biotec)을 이용해 Thy1+ CAF를 분류하였다. 음성 선택된 세포 상층액에서는, CD326+ 상피 종양 세포 및 CD11b+ 종양 관련 대식세포를 CD326 및 CD11b MicroBeads(Miltenyi Biotec)를 각각 이용해 분리하였다. 분리된 세포는 완전 배지에서 배양되었다. Thy1+ 세포는 20% FBS 및 1% 페니실린/스트렙토마이신이 보충된 α-MEM(Welgene)에서 배양되었고, CD326+ 세포는 10% FBS 및 1% 페니실린/스트렙토마이신이 보충된 RPMI 1640(Welgene)에서 배양되었고, CD11b+ 세포는 10% FBS 및 1% 페니실린/스트렙토마이신이 보충된 DMEM(Welgene)에서 배양되었다. 분리된 개별 세포 그룹은 qRT-PCR로 검증되었고, 개별 세포는 두개 또는 세개의 각 그룹내에서 무작위로 선택된 마우스 원발성 종양 내에서 분리되었다.To obtain a single cell suspension from the primary tumor of the metastatic mouse model, solid, fresh tumor tissue was dissociated using a tumor dissociation kit combined with a MACSTM Dissociator (all Miltenui Biotec Inc., San Diego, CA) according to the manufacturer's instructions. made it Briefly, tumor tissue from each mouse was transferred to a MACS C tube (Miltenui Biotec) containing the enzyme mixture from the kit for enzymatic digestion. After dissociation, each tissue homogenate was filtered through 70- and 40-μm sterile nylon mesh and red blood cells were lysed with red blood cell lysis buffer. After centrifugation at 300 g for 10 min, the remaining cells were collected with anti-CD45 (Abcam, Cambridge, UK), anti-CD68 (Abcam), anti-CD31 (Abcam) and anti-CD326 (Abcam) to select for non-CAF cells. BD Bioscience, San Diego, CA, USA). Labeled cells were reduced using pre-washed Dynabeads (Invitrogen) from the total cell suspension and separated from the magnetic beads using a PureProteome magnetic stand (Millipore, Billerica, MA, USA). In the unlabeled cell supernatant, Thy1 + CAFs were sorted using CD90.2 MicroBeads and a MACS MS column (Miltenyi Biotec) according to the manufacturer's instructions. In the negatively selected cell supernatant, CD326 + epithelial tumor cells and CD11b + tumor-associated macrophages were isolated using CD326 and CD11b MicroBeads (Miltenyi Biotec), respectively. Isolated cells were cultured in complete medium. Thy1 + cells were cultured in α-MEM (Welgene) supplemented with 20% FBS and 1% penicillin/streptomycin, and CD326 + cells were cultured in RPMI 1640 (Welgene) supplemented with 10% FBS and 1% penicillin/streptomycin. and CD11b + cells were cultured in DMEM (Welgene) supplemented with 10% FBS and 1% penicillin/streptomycin. Isolated individual cell groups were validated by qRT-PCR, and individual cells were isolated within randomly selected mouse primary tumors within each group of two or three.

1-19. 역전사-PCR 어레이1-19. Reverse transcription-PCR array

원발성 종양에서 분리된 CD326+ 종양 세포 내 종양 전이와 관련된 유전자 발현 분석 및 원발성 종양에서 분리된 Thy1+ CAFs 내 세포외 기질과 응집 분자 관련된 유전자 발현 분석을 위해서, 마우스 종양 전이 RT2 ProfilerTM PCR Array (PAMM-028ZA-6, Qiagen, Hilden, Germany) 및 마우스 세포외 기질 및 응집 분자 RT2 ProfilerTM PCR Array(PAMM-013ZA-6, Qiagen)를 이용하였다. RNA 분리, DNase 처리 및 RNA 세척은 제조사(Takara Bio)의 지시에 따라 수행하였다. 분리된 RNA를 RT2 First Strand 키트(Qiagen)를 이용해 cDNA로 역전사시켰다. PCR은 RT2 SYBR Green qPCR Master Mix(Qiagen), QuantStudio쪠3 Real-Time PCR 시스템 및 ABI PRISM7900 instrument(Applied Bio-Systems)를 이용해 수행하였다. 발현량 데이터를 어레이 내 세포유지유전자(housekeeping gene)인 Gapdh(glyceraldehyde 3-phosphate dehydrogenase)의 평균 Ct값을 이용해 정규화하였다. 각 분석을 3회씩 수행하였다.For gene expression analysis related to tumor metastasis in CD326 + tumor cells isolated from primary tumors and gene expression related to extracellular matrix and aggregation molecules in Thy1 + CAFs isolated from primary tumors, mouse tumor metastasis RT 2 Profiler TM PCR Array ( PAMM-028ZA-6, Qiagen, Hilden, Germany) and mouse extracellular matrix and aggregation molecule RT 2 Profiler TM PCR Array (PAMM-013ZA-6, Qiagen) were used. RNA isolation, DNase treatment and RNA washing were performed according to the instructions of the manufacturer (Takara Bio). The isolated RNA was reverse transcribed into cDNA using the RT 2 First Strand kit (Qiagen). PCR was performed using RT 2 SYBR Green qPCR Master Mix (Qiagen), QuantStudio 3 Real-Time PCR system and ABI PRISM7900 instrument (Applied Bio-Systems). The expression level data was normalized using the average Ct value of Gapdh (glyceraldehyde 3-phosphate dehydrogenase), a housekeeping gene in the array. Each assay was performed in triplicate.

1-20. 통계 분석1-20. statistical analysis

2개의 평균값인 ±SEM(대조군 대 실험군) 사이의 비교는 two-tailed Student's t-tests로 수행하였고, 다중 비교는 Kruskal-Wallis 테스트와 Dunn's post hoc 테스트로 수행하였다. 0.05 미만의 P 값은 통계적으로 유의한 것으로 간주하였다. 모든 데이터는 Graph Prism 5 소프트웨어(GraphPad Software Inc., San Diego, CA, USA)를 사용하여 분석하였다.Comparison between the two mean values, ±SEM (control group versus experimental group), was performed with two-tailed Student's t -tests, and multiple comparisons were performed with Kruskal-Wallis test and Dunn's post hoc test. A P value of less than 0.05 was considered statistically significant. All data were analyzed using Graph Prism 5 software (GraphPad Software Inc., San Diego, CA, USA).

실험예 2: 실험결과Experimental Example 2: Experimental results

2-1. CAFs와 사멸화된 암 세포 공동 배양 조정배지(ApoSQ-CAF CM)의 암 세포 이동 및 침윤 억제 확인2-1. Confirmation of cancer cell migration and invasion inhibition of CAFs and apoptosis cancer cell co-culture conditioned medium (ApoSQ-CAF CM)

CAFs와 암 세포 사이의 paracrine(주변분비) 작용이 암 세포 이동 및 침윤과 같은 기저 과정을 촉진한다고 알려져 있기 때문에, 본 실험에서는 UV-조사되어 사멸화된 암 세포와 CAFs를 공동 배양할 때, 생물활성 매개체 분비를 통하여 폐암 세포의 이동 및 침윤이 억제되는지 여부를 조사하였다.Since it is known that the paracrine (paracrine) action between CAFs and cancer cells promotes basic processes such as cancer cell migration and invasion, in this experiment, when co-culturing UV-irradiated and killed cancer cells and CAFs, Whether migration and invasion of lung cancer cells are inhibited through the secretion of active mediators was investigated.

섬유아세포-특이적 마커인 Thy1을 사용하여 Kras-돌연변이체(KrasLA1) 마우스의 폐 종양으로부터 CAFs를 분리한 후, 사멸화된 344SQ 세포주(ApoSQ)로 20시간 동안 처리하였으며, 이때 상기 세포주로 KrasLA1 및 53R172H 대립유전자(22)를 동시발현하는 마우스로부터 유래된 고침윤성 및 전이성 폐 선암종(adenocarcinoma) 세포주를 이용하였다.After isolating CAFs from lung tumors of Kras-mutant (Kras LA1 ) mice using the fibroblast-specific marker Thy1, they were treated with the apoptosis 344SQ cell line (ApoSQ) for 20 hours, at which time the cell line was used as Kras A highly invasive and metastatic lung adenocarcinoma cell line derived from mice co-expressing the LA1 and 53 R172H alleles (22) was used.

344SQ 세포는 CAFs와 사멸화된 344SQ 공동 배양 조정배지(ApoSQ-CAF CM) 또는 CAFs와 괴사된 344SQ 공동 배양 조정배지(NecSQ-CAF CM)로 TGF-beta 1의 존재 또는 부재하에 48시간 동안 처리하였다. 트랜스웰 이동 및 침윤 분석을 수행하여 화학적 유인 구배(chemo-attractant gradient)에 대하여 세포 이동능 및 침윤능을 분석하였다.344SQ cells were treated with CAFs and killed 344SQ co-culture conditioned medium (ApoSQ-CAF CM) or CAFs and necrotic 344SQ co-culture conditioned medium (NecSQ-CAF CM) in the presence or absence of TGF-beta 1 for 48 hours. . Transwell migration and invasion assays were performed to analyze cell migration and invasion capabilities against a chemo-attractant gradient.

그 결과, ApoSQ-CAF CM이 TGF-beta 1-유도 암 세포의 이동 및 침윤을 실질적으로 억제한 반면, 대조군 또는 NecSQ-CAF CM은 그렇지 않음을 확인하였다(도 1a). 또한, 비소세포 폐암 세포주 A549, 대장암 세포주 HCT116 및 유방암 세포주 MCF-7과 같은 다른 유형의 인간 암 세포주에서도 CAFs와 사멸화된 암 세포의 공동 배양 조정배지(ApoA-CAF CM, ApoH-CAF CM 및 ApoM-CAF CM)의 암 세포 이동 및 침윤 억제 효과를 확인할 수 있었다(도 1b 내지 1d).As a result, it was confirmed that ApoSQ-CAF CM substantially inhibited the migration and invasion of TGF-beta 1-induced cancer cells, whereas the control or NecSQ-CAF CM did not (FIG. 1a). In addition, other types of human cancer cell lines, such as non-small cell lung cancer cell line A549, colon cancer cell line HCT116, and breast cancer cell line MCF-7, were also co-cultured with CAFs and killed cancer cells in conditioned media (ApoA-CAF CM, ApoH-CAF CM and ApoM-CAF CM) was able to confirm the effect of inhibiting cancer cell migration and invasion (FIGS. 1b to 1d).

다음으로, 본 발명자들은 폐암 세포의 이동 및 침윤이 ApoSQ 단독 유래의 CM(ApoSQ CM) 또는 ApoSQ에 대한 직접적인 노출에 의해 영향을 받는지를 조사하였다. 그 결과, ApoSQ CM, NecSQ CM, ApoSQ 또는 NecSQ에 대한 직접적인 노출이 344SQ 세포의 TGF-beta 1이 유도하는 이동 및 침윤에 영향을 미치지 않음을 확인하였다(도 2a 및 2b). 이러한 결과들은, 항-이동 및 항-침윤 효과는 CAFs에 의해 분비되는 생물활성 매개체를 필요로 하며, 이는 사멸화된 암 세포 자극에 의해 CAFs가 기능적으로 변화된다는 것을 의미한다.Next, we investigated whether migration and invasion of lung cancer cells were affected by CM derived from ApoSQ alone (ApoSQ CM) or direct exposure to ApoSQ. As a result, it was confirmed that direct exposure to ApoSQ CM, NecSQ CM, ApoSQ or NecSQ did not affect the TGF-beta 1-induced migration and invasion of 344SQ cells (FIGS. 2a and 2b). These results suggest that anti-migration and anti-invasion effects require bioactive mediators secreted by CAFs, which are functionally altered by stimulation of apoptotic cancer cells.

한편, TGF-beta 1 신호가 활성화되면 Smad 의존성 및 비의존성 신호전달을 조절함으로써 암 세포의 이동 및 침윤을 유도하는데, ApoSQ-CAF CM이 344SQ 세포 내에서 Smad2/3, FAK, Akt, Src, ERK 및 p38 MAP kinase 신호체계와 같은 TGF-beta 1의 신호를 억제함을 확인하였다(도 3a 및 3b). 또한, TGF-beta 1 신호는 세포외 기질을 분해하고 리모델링하는 MMP-2 및 MMP-12의 mRNA 및 단백질 발현의 상향조절을 유도하는데, ApoSQ-CAF CM이 이를 억제함을 확인하였다(도 4).On the other hand, when TGF-beta 1 signal is activated, it induces migration and invasion of cancer cells by regulating Smad-dependent and -independent signaling. and suppression of TGF-beta 1 signals such as the p38 MAP kinase signaling system (FIGS. 3a and 3b). In addition, TGF-beta 1 signal induces upregulation of mRNA and protein expression of MMP-2 and MMP-12, which degrade and remodel the extracellular matrix, and it was confirmed that ApoSQ-CAF CM inhibited this (FIG. 4) .

상기 실험을 통하여, CAFs와 사멸화된 암 세포를 공동 배양한 조정배지가 TGF-beta 1 신호가 유도하는 암 세포의 이동 및 침윤 관련 신호전달 경로 분자들의 활성화를 억제할 뿐 아니라, ECM 리모델링과 관련된 MMP2/12의 mRNA 및 단백질의 발현을 억제함으로써, 암 세포의 이동 및 침윤을 억제할 수 있음을 검증하였다.Through the above experiments, the conditioned medium in which CAFs and apoptotic cancer cells were co-cultured not only suppressed the activation of TGF-beta 1 signal-induced migration and invasion-related signaling pathway molecules of cancer cells, but also related to ECM remodeling. It was verified that migration and invasion of cancer cells can be inhibited by inhibiting the expression of mRNA and protein of MMP2/12.

2-2. CAFs와 사멸화된 폐암 세포 간 상호작용에 의한 CAF의 이동 및 침윤 억제 확인2-2. Confirmation of inhibition of CAF migration and invasion by interaction between CAFs and killed lung cancer cells

CAFs의 이동 능력은 CAFs의 활성 상태와 관련되는데, 예를 들면 활 성화된 섬유아세포는 quiescent 또는 resting 섬유아세포보다 더 잘 이동한다. 사멸화된 폐암 세포에 대한 CAFs의 직접적인 노출이 CAFs 자체의 이동 능력에 영향을 미치는지를 확인하기 위해, CAFs를 20시간 동안 ApoSQ 또는 NecSQ에 노출시킨 다음, TGF-beta 1의 존재 또는 부재하에 새로운 배지로 24시간 동안 교체하였다. 그 결과, CAF와 ApoSQ의 직접적인 상호작용이 TGF-beta 1이 유도하는 CAFs 자체의 이동 및 침윤을 억제하는 반면, CAFs를 NecSQ로 처리한 경우에는 이러한 억제 효과가 나타나지 않았다(도 5). 더욱이, TGF-beta 1의 부재 하에 CAFs를 ApoSQ 또는 NecSQ에 직접 노출시킨 경우에는 기저 수준에 영향을 미치지 않았다.The migratory ability of CAFs is related to the activation state of CAFs, ie, activated fibroblasts migrate better than quiescent or resting fibroblasts. To confirm whether direct exposure of CAFs to apoptotic lung cancer cells affects the migration ability of CAFs themselves, CAFs were exposed to ApoSQ or NecSQ for 20 hours, followed by fresh medium in the presence or absence of TGF-beta 1. was replaced for 24 hours. As a result, while the direct interaction between CAFs and ApoSQ inhibited the migration and invasion of CAFs induced by TGF-beta 1, this inhibitory effect was not shown when CAFs were treated with NecSQ (FIG. 5). Moreover, direct exposure of CAFs to ApoSQ or NecSQ in the absence of TGF-beta 1 did not affect basal levels.

또한, 자가분비(autocrine secretion)를 통한 CAFs 자체에 미치는 항-이동 및 항-침윤 효과를 확인하기 위해, ApoSQ-CAF CM 또는 NecSQ-CAF CM을 TGF-beta 1의 존재 또는 부재하에 CAFs 단일층에 첨가하였다. ApoSQ의 직접적인 효과와 유사하게, ApoSQ-CAF CM 처리 시, TGF-beta 1이 유도하는 CAFs의 이동 및 침윤이 유의적으로 억제되었으나, NecSQ-CAF CM은 억제 효과가 거의 없었다(도 6a). 유사하게, CAF CM이 다른 유형의 사멸화된 암 세포, 예컨대 A549, HCT116 및 MCF-7과 공동 배양된 ApoA-CAF CM, ApoH-CAF CM 및 ApoM-CAF CM의 경우에도 CAFs의 이동 및 침윤이 억제되었다(도 6b 내지 6d). 그러나, ApoSQ CM 및 NecSQ CM 단독으로는 이러한 억제 효과가 없었다(도 7).In addition, to confirm the anti-migration and anti-invasion effects on CAFs themselves through autocrine secretion, ApoSQ-CAF CM or NecSQ-CAF CM was applied to CAFs monolayers in the presence or absence of TGF-beta 1. added. Similar to the direct effect of ApoSQ, treatment with ApoSQ-CAF CM significantly inhibited TGF-beta 1-induced migration and invasion of CAFs, but NecSQ-CAF CM had little inhibitory effect (Fig. 6a). Similarly, for ApoA-CAF CM, ApoH-CAF CM and ApoM-CAF CM in which CAF CMs were co-cultured with other types of killed cancer cells, such as A549, HCT116 and MCF-7, migration and invasion of CAFs were also observed. inhibited (Figs. 6b to 6d). However, ApoSQ CM and NecSQ CM alone did not have this inhibitory effect (FIG. 7).

추가적으로, CAFs는 TGF-beta 1 처리에 반응하여 더욱 공격적인 표현형을 나타낸다는 것이 알려져 있는 바, 본 발명자들은 ApoSQ에 대한 직접적인 노출이 CAF 활성화 마커를 억제하는지 여부를 조사하였다. 그 결과, ApoSQ 처리군은 TGF-beta 1이 유도하는 α-smooth muscle actin (SMA), collagen type 1 및 피브로넥틴과 같은 CAF 활성화 마커들의 유전자 및 단백질 발현 증가를 억제시킨 반면, NecSQ 처리군은 그렇지 않았다(도 8a). 또한, 면역세포화학 분석 결과, CAFs에서 TGF-beta 1이 유도하는 α-SMA 단백질의 발현이 ApoSQ와의 직접 노출에 의해서는 감소하였으나, NecSQ와의 노출에 의해서는 감소하지 않았다(도 8b).Additionally, as it is known that CAFs exhibit a more aggressive phenotype in response to TGF-beta 1 treatment, we investigated whether direct exposure to ApoSQ inhibits CAF activation markers. As a result, the ApoSQ-treated group suppressed TGF-beta 1-induced increases in gene and protein expression of CAF activation markers such as α-smooth muscle actin (SMA), collagen type 1, and fibronectin, whereas the NecSQ-treated group did not. (Fig. 8a). In addition, as a result of immunocytochemical analysis, the expression of TGF-beta 1-induced α-SMA protein in CAFs was decreased by direct exposure to ApoSQ, but not by exposure to NecSQ (Fig. 8b).

이와 유사하게, ApoSQ가 CAFs 내에서 TGF-beta 1이 유도하는 Smad 또는 비 Smad 신호의 활성화를 억제하였고(도 9a 및 9b), TGF-beta 1이 유도하는 MMP-2 및 MMP-12의 mRNA 및 단백질 발현 또한 억제함을 확인하였다(도 10).Similarly, ApoSQ inhibited the activation of TGF-beta 1-induced Smad or non-Smad signaling in CAFs (FIG. 9a and 9b), and TGF-beta 1-induced MMP-2 and MMP-12 mRNA and It was confirmed that protein expression was also inhibited (FIG. 10).

상기 실험을 통하여, CAFs와 사멸화된 암 세포를 공동 배양한 조정배지의 CAFs의 이동 및 침윤 억제 효과를 검증하는 한편, 암 관련 섬유아세포를 사멸화된 암 세포에 노출시켜 상호작용을 일으킬 경우, 상기 CAFs에서 TGF-beta 1이 유도하는 CAF 활성화 마커들의 발현이 억제되고, 세포 이동 및 침윤 관련 신호전달 경로의 활성화가 억제되고, MMP2/12 발현이 유전자 및 단백질 수준에서 억제됨으로써 암 세포의 이동 및 침윤을 억제할 수 있음을 검증하였다.Through the above experiments, while verifying the effect of inhibiting migration and invasion of CAFs in a conditioned medium in which CAFs and apoptotic cancer cells are co-cultured, when exposing cancer-related fibroblasts to apoptotic cancer cells to cause interaction, In the CAFs, the expression of TGF-beta 1-induced CAF activation markers is suppressed, the activation of cell migration and invasion-related signaling pathways is suppressed, and the expression of MMP2/12 is suppressed at the gene and protein level, resulting in cancer cell migration and It was verified that infiltration can be inhibited.

2-3. WISP-1이 암 세포 및 CAFs의 이동 및 침윤 억제에 필요함을 확인 2-3. Confirmation that WISP-1 is required for inhibition of migration and invasion of cancer cells and CAFs

CAFs로부터 분비된 autocrine/paracrine 인자들이 암 세포 및 CAFs의 이동 및 침윤을 억제하는데 중요한 역할을 하는지 확인하기 위해, CAF CM, ApoSQ 단독 배지, 또는 ApoSQ-CAF CM을 사용하여, 단일 샘플에서 111종까지의 사이토카인 측정이 가능한 사이토카인 어레이 분석을 실시하였다.To confirm that autocrine/paracrine factors secreted from CAFs play an important role in inhibiting the migration and invasion of cancer cells and CAFs, CAF CM, ApoSQ alone medium, or ApoSQ-CAF CM were used to examine up to 111 species in a single sample. A cytokine array analysis capable of measuring cytokines was performed.

그 결과, 이들 사이토카인 중에서 LIF(leukemia inhibitory factor) 및 WISP-1이, ApoSQ에 대한 반응으로 CAFs에 의해 가장 높게 유도되었고, 이들 사이토카인의 증가는 CAFs가 없는 ApoSQ 단독 CM에서는 나타나지 않았기 때문에(도 11), 본 발명자들은 특정 siRNA를 사용하여, 어떤 사이토카인이 ApoSQ-CAF CM의 항-이동 및 항-침윤 효과에 관여하는지를 조사하였다.As a result, among these cytokines, LIF (leukemia inhibitory factor) and WISP-1 were most highly induced by CAFs in response to ApoSQ, and the increase in these cytokines was not seen in ApoSQ alone CM without CAFs (Fig. 11), we used specific siRNAs to investigate which cytokines are involved in the anti-migration and anti-invasion effects of ApoSQ-CAF CM.

특이적 siRNA로 형질감염된 CAFs에서 넉다운된 WISP-1을 확인하였고, WISP-1 분비는 또한 ApoSQ의 존재 또는 부재하에 형질감염된 CAFs에서 감소되었다(도 12). WISP1-1 넉다운은 344SQ 세포에서 TGF-beta 1이 유도하는 이동 및 침윤에 대해 ApoSQ-CAF CM의 억제 효과를 차단하였으며(도 13a), 또한 CAFs 이동 및 침윤에 대한 ApoSQ의 직접적인 억제 효과를 차단하였다(도 13b). 이와 달리, LIF를 넉다운시킨 경우, 344SQ 세포 또는 CAFs 내에서의 이동 및 침윤 억제 효과가 나타나지 않았다(도 14 내지 15b).We confirmed knockdown of WISP-1 in CAFs transfected with specific siRNA, and WISP-1 secretion was also reduced in CAFs transfected with or without ApoSQ (FIG. 12). WISP1-1 knockdown blocked the inhibitory effect of ApoSQ-CAF CM on TGF-beta 1-induced migration and invasion in 344SQ cells (Fig. 13a), and also blocked the direct inhibitory effect of ApoSQ on CAFs migration and invasion (Fig. 13b). In contrast, when LIF was knocked down, there was no effect of inhibiting migration and invasion in 344SQ cells or CAFs (FIGS. 14 to 15b).

또한, 본 발명자들은 WISP-1 과발현 CAFs를 사용하여, CAFs에 의한 ApoSQ 유도성 WISP-1 분비 증가 효과를 확인하였다. 먼저, WISP-1 플라스미드로 형질감염된 CAFs에서 WIPS-1 발현이 증가되었음을 확인하였으며, WISP-1 분비는 ApoSQ의 존재 또는 부재하에 형질감염된 CAFs에서 증가되었다(도 16). ApoSQ 존재 시 WISP-1 과발현된 CAF CM으로 처리할 경우, 대조군(공벡터) 형질감염된 CAFs와 ApoSQ 공동 배양액과 비교하여, 344SQ 세포의 TGF-beta 1이 유도하는 이동 및 침윤을 더욱 억제하였다(도 17a). WISP-1 과발현된 CAFs에 ApoSQ를 처리시 TGF-beta 1이 유도하는 CAFs의 이동 및 침윤은, ApoSQ 부재 시 대조군 형질감염 CAFs의 것에 비해 감소되었지만, 대조군 형질감염 CAFs에 ApoSQ 처리한 것에 비해 더 억제되지는 않았고, ApoSQ 부재하에 WISP-1 과발현된 CAFs의 TGF-beta 1이 유도하는 이동 및 침윤 억제효과는, 대조군 형질감염 CAFs와 비교하여 감소되었다(도 17b). 또한, 항-WISP-1 항체로 중화 후 ApoSQ-CAF CM으로 처리 시, 344SQ 세포에 대한 항-이동 및 항-침윤 효과가 제거되는 반면, 대조군 IgG로 처리한 경우 이러한 효과가 나타나지 않았다(도 18).In addition, the present inventors confirmed the effect of increasing ApoSQ-induced WISP-1 secretion by CAFs using WISP-1 overexpressing CAFs. First, it was confirmed that WIPS-1 expression was increased in CAFs transfected with the WISP-1 plasmid, and WISP-1 secretion was increased in CAFs transfected with or without ApoSQ (FIG. 16). When treated with CAF CM overexpressing WISP-1 in the presence of ApoSQ, compared to control (empty vector) transfected CAFs and ApoSQ co-culture, the TGF-beta 1-induced migration and invasion of 344SQ cells were further suppressed (Fig. 17a). When CAFs overexpressing WISP-1 were treated with ApoSQ, TGF-beta 1-induced migration and invasion of CAFs were reduced compared to those of control transfected CAFs in the absence of ApoSQ, but were more inhibited than ApoSQ treated control transfected CAFs. In the absence of ApoSQ, the TGF-beta 1-induced migration and invasion inhibitory effects of WISP-1 overexpressed CAFs were reduced compared to control transfected CAFs (FIG. 17b). In addition, when treated with ApoSQ-CAF CM after neutralization with anti-WISP-1 antibody, the anti-migration and anti-invasion effects on 344SQ cells were eliminated, whereas these effects were not observed when treated with control IgG (FIG. 18 ).

WISP-1이 autocrine/paracrine 방식으로 작용하여 항-이동 및 항- 침윤 효과를 유도한다는 것을 입증하기 위해, 5 및 10 ng/ml, 또는 0.75-3 ng/ml 농도의 재조합 마우스 WISP-1(rWISP-1)을 TGF-beta 1의 존재 하에 각각 344SQ 또는 CAFs에 직접 처리하였다. 예상한대로, 이러한 저용량 rWISP-1로 처리시, TGF-beta 1 유도성 344SQ 및 CAFs의 이동 및 침윤이 농도 의존적으로 억제되었다(도 19a 및 19b). 또한, rWISP-1이 344SQ 세포 또는 CAFs 내에서 TGF-beta 1이 유도하는 Smad 또는 비 Smad 신호체계를 억제하고, MMP2/MMP12 mRNA 및 단백질의 발현을 억제함을 확인하였다(도 20a 내지 21c).To demonstrate that WISP-1 acts in an autocrine/paracrine manner to induce anti-migration and anti-invasion effects, recombinant mouse WISP-1 (rWISP) was used at concentrations of 5 and 10 ng/ml, or 0.75-3 ng/ml. -1) was directly treated with 344SQ or CAFs, respectively, in the presence of TGF-beta 1. As expected, treatment with this low dose of rWISP-1 inhibited TGF-beta 1-induced migration and invasion of 344SQ and CAFs in a concentration-dependent manner (FIGS. 19a and 19b). In addition, it was confirmed that rWISP-1 suppressed the TGF-beta 1-induced Smad or non-Smad signaling system and suppressed the expression of MMP2/MMP12 mRNA and protein in 344SQ cells or CAFs (FIGS. 20a to 21c).

상기 실험 결과들은, ApoSQ-CAF CM에서 특이적으로 높게 발현되는 WISP-1을 통해 암 세포 및 CAFs에서 항-이동 및 항-침윤 효과가 발휘되는 것임을 의미하는 한편, 상기 WISP-1이 CAFs가 ApoSQ와 접촉하여 상호작용함으로써 분비되는 것임을 시사한다.The above experimental results indicate that anti-migration and anti-invasion effects are exerted in cancer cells and CAFs through WISP-1, which is specifically highly expressed in ApoSQ-CAF CM, while the WISP-1 in CAFs exhibits ApoSQ It suggests that it is secreted by contacting and interacting with.

추가적으로, WISP-1은 세포 표면 수용체인 인테그린에 결합하는 것으로 알려져 있는 바, 344SQ 세포 및 CAFs에서 WISP-1의 autocrine/paracrine 기능을 매개하는 인테그린을 결정하기 위해, 인테그린 αv, α5, beta 1, beta 3 또는 beta 5에 대한 차단 항체를 이용하여 실험을 수행하였다.Additionally, since WISP-1 is known to bind to integrin, a cell surface receptor, integrins αv, α5, beta 1, beta Experiments were performed using blocking antibodies against 3 or beta 5.

실험 결과, 344SQ 세포의 경우, 항-인테그린 αv 또는 beta 3 항체를 처리하였을 때, 대조군 IgG 항체와 비교하여 rWISP-1의 항이동 및 항침윤 효과가 역전되었으며(도 22a), CAFs의 경우, 항-인테그린 αv 또는 beta 5 항체 처리에 의해 대조군 IgG 항체와 비교하여 rWISP-1의 항이동 및 항침윤 효과가 역전됨을 확인하였다(도 22b). 이와 달리, 항-인테그린 α5, beta 1, 또는 beta 5로 344SQ 세포를 처리하거나, 항-인테그린 α5, beta 1 또는 beta 3로 CAFs를 처리할 경우에는 세포의 이동 및 침윤에 유의한 변화가 나타나지 않았다.As a result of the experiment, in the case of 344SQ cells, when treated with anti-integrin αv or beta 3 antibodies, the anti-migration and anti-invasion effects of rWISP-1 were reversed compared to the control IgG antibody (FIG. 22a), and in the case of CAFs, the anti-migration and anti-invasion effects were reversed. - It was confirmed that the anti-migration and anti-invasion effects of rWISP-1 were reversed by treatment with integrin αv or beta 5 antibody compared to the control IgG antibody (FIG. 22b). In contrast, treatment of 344SQ cells with anti-integrin α5, beta 1, or beta 5, or treatment of CAFs with anti-integrin α5, beta 1, or beta 3 did not show significant changes in cell migration or invasion. .

또한, WISP-1이 344SQ 세포에서는 인테그린 αv 및 beta 3를 통해, CAFs에서는 인테그린 αv 및 beta 5를 통해 TGF-beta 1이 유도하는 신호전달 경로를 억제하는지 확인하기 위한 실험을 수행하였다.In addition, experiments were performed to confirm whether WISP-1 inhibits the signaling pathway induced by TGF-beta 1 through integrin αv and beta 3 in 344SQ cells and through integrin αv and beta 5 in CAFs.

그 결과, 항-인테그린 αv 또는 beta 3 항체로 344SQ 세포를 처리할 경우, TGF-beta 1이 유도하는 신호전달 경로와 MMP2/MMP12 mRNA 및 단백질 발현에 대한 rWISP-1의 억제 효과도 약화되었으며(도 23a 내지 23c), 항-인테그린 αv 또는 beta 3 항체로 CAFs를 처리할 경우에도, 상기와 같이 TGF-beta 1이 유도하는 신호전달 경로와 MMP2/MMP12 mRNA 및 단백질 발현에 대한 rWISP-1의 억제 효과가 약화됨을 확인하였다(도 24a 내지 24c).As a result, when 344SQ cells were treated with anti-integrin αv or beta 3 antibodies, the inhibitory effect of rWISP-1 on TGF-beta 1-induced signaling pathway and MMP2/MMP12 mRNA and protein expression was also attenuated (Fig. 23a to 23c), even when CAFs were treated with anti-integrin αv or beta 3 antibody, the inhibitory effect of rWISP-1 on TGF-beta 1-induced signaling pathway and MMP2/MMP12 mRNA and protein expression It was confirmed that was weakened (FIGS. 24a to 24c).

상기 실험을 통해, WISP-1이 344SQ 세포의 인테그린 αv beta 3 수용체 및 CAFs의 인테그린 αv beta 5 수용체를 통해 autocrine/paracrine 방식으로 TGF-beta 1이 유도하는 신호전달 경로를 억제함으로써, 세포의 이동 및 침윤을 조절함을 검증하였다.Through the above experiments, WISP-1 inhibits the signaling pathway induced by TGF-beta 1 in an autocrine/paracrine manner through the integrin αv beta 3 receptor of 344SQ cells and the integrin αv beta 5 receptor of CAFs, thereby inhibiting cell migration and It was verified that the infiltration was controlled.

2-4. 사멸화된 암 세포가 CAFs에서 Notch1 신호전달을 유도하여 WISP-1 생성을 초래함을 확인2-4. Confirmation that apoptotic cancer cells induce Notch1 signaling in CAFs resulting in WISP-1 production

Notch1-WISP-1 축은 흑색종 침윤 및 전이에서 중간엽 줄기 세포-유래 기질 섬유아세포의 조절 역할을 결정하는 것으로 알려져 있다. 따라서, 본 발명자는 사멸화된 폐암 세포에 대한 반응으로 CAFs에서 Notch1 신호전달-의존성 WISP-1 생성이 암 세포 및 CAFs에서의 항-이동 및 항-침윤 효과에 결정적인 역할을 한다고 가정하였다.The Notch1-WISP-1 axis is known to determine the regulatory role of mesenchymal stem cell-derived stromal fibroblasts in melanoma invasion and metastasis. Therefore, the present inventors hypothesized that Notch1 signaling-dependent WISP-1 production in CAFs in response to killed lung cancer cells plays a critical role in the anti-migration and anti-invasion effects in cancer cells and CAFs.

이러한 가정을 입증하기 위해, 우선 ApoSQ에 반응하여 CAFs에서 Notch1 신호전달 경로가 활성화되는지 여부를 조사하였다. 웨스턴 블랏 분석 결과, Notch 경로의 하위 표적으로 알려진 Hes1 및 WISP-1 뿐만 아니라, Notch1 신호의 활성화시에만 분비된다고 알려진 Notch intracellular domain 1(NICD1)을 포함하여, Notch1 신호 단백질들이 CAFs에서 ApoSQ에 의해 유도됨을 확인하였다(도 25(A)). 또한, ApoSQ-CAF CM에서 WISP-1 분비가 증가되었지만, NecSQ-CAF CM에서는 증가되지 않았다(도 25(B)).To verify this hypothesis, we first investigated whether the Notch1 signaling pathway is activated in CAFs in response to ApoSQ. As a result of Western blot analysis, Notch1 signaling proteins, including Hes1 and WISP-1, known as sub-targets of the Notch pathway, as well as Notch intracellular domain 1 (NICD1), known to be secreted only when Notch1 signaling is activated, are induced by ApoSQ in CAFs It was confirmed (FIG. 25 (A)). In addition, WISP-1 secretion increased in ApoSQ-CAF CM, but not in NecSQ-CAF CM (FIG. 25(B)).

더욱이, CAFs에서 Hes1 및 WISP-1 mRNA 발현이 증가되었고, 이들 증가는 ApoSQ 처리 후 각각 5시간 및 20시간 유지되었지만, NecSQ 자극시에는 효과를 나타내지 않았고, Notch 시그널링 활성의 일반적인 전사 이펙터인 Notch-반응성 리포터 4x-CSL-Luc 활성 실험결과, 20시간까지 ApoSQ에 대한 노출에 의해 CAFs에서 Notch 신호전달의 활성화가 증가된 반면, NecSQ 자극시에는 유의한 활성이 없었다(도 26). 추가적으로, 면역세포화학 분석 결과, ApoSQ 자극에 반응하여 CAFs에서 NICD1 및 WISP-1의 발현이 증가되었고, WISP-1 발현은 핵에 위치된 NICD1 염색-양성 CAFs에서 공동편재 (colocalized)되었음을 확인하였다(도 27).Moreover, Hes1 and WISP-1 mRNA expression was increased in CAFs, and these increases were maintained 5 and 20 h after ApoSQ treatment, respectively, but had no effect upon NecSQ stimulation, suggesting that Notch-responsiveness, a common transcriptional effector of Notch signaling activity, As a result of the reporter 4x-CSL-Luc activity test, the activation of Notch signaling in CAFs was increased by exposure to ApoSQ up to 20 hours, whereas there was no significant activity when stimulated with NecSQ (FIG. 26). Additionally, as a result of immunocytochemical analysis, it was confirmed that the expression of NICD1 and WISP-1 was increased in CAFs in response to ApoSQ stimulation, and that WISP-1 expression was colocalized in NICD1 stain-positive CAFs located in the nucleus ( Figure 27).

ApoSQ가 유도하는 Notch1 신호전달-의존성 WISP-1 분비가 결국 폐암 세포 및 CAFs의 이동 및 침윤의 억제를 매개한다는 가설을 확인하기 위해, Notch1 siRNA와 γ-secretase 억제제인 DAPT를 사용하여 추가 실험을 진행하였다. 그 결과, 특이적 siRNA를 이용하여 CAFs에서 Notch1을 넉다운시키면, 증가된 NICD1, Hes1 및 WISP-1 단백질 발현이 저해되었으며(도 28a 및 28b(A)), ApoSQ 처리에 의한 WIPS-1 분비도 저해되었고(도 28b(B)), 이는 결과적으로 344SQ(도 29a) 및 CAFs 자체에서(도 29b) 항-이동 및 항-침윤 효과를 저해하는 효과를 낳았다. Notch1 억제제인 DAPT를 10 μM로 전처리한 경우에도, ApoSQ 처리에 의해 CAFs에서 NICD1, Hes1 및 WISP-1의 발현이 증가되었던 것이 억제되었으며(도 30(A)), WISP-1 분비 또한 감소시켰다(도 30(B)). 결과적으로, CAFs에 DAPT를 처리할 경우, 344SQ에서 ApoSQ-CAF CM의 항-이동 및 항-침윤 효과를 상당히 저해시켰다(도 31a). 유사하게, ApoSQ의 항-이동 및 항-침윤 효과는 또한 CAFs 자체에서 DAPT에 의해 완전히 저해되었다(도 31b).To confirm the hypothesis that ApoSQ-induced Notch1 signaling-dependent WISP-1 secretion eventually mediates the inhibition of migration and invasion of lung cancer cells and CAFs, further experiments were conducted using Notch1 siRNA and the γ-secretase inhibitor DAPT. did As a result, when Notch1 was knocked down in CAFs using specific siRNA, increased NICD1, Hes1, and WISP-1 protein expression was inhibited (Fig. 28a and 28b (A)), and WIPS-1 secretion by ApoSQ treatment was also inhibited. (FIG. 28B (B)), which in turn had the effect of inhibiting the anti-migration and anti-invasion effects in 344SQ (FIG. 29A) and CAFs themselves (FIG. 29B). Even when pre-treatment with 10 μM of DAPT, a Notch1 inhibitor, increased expression of NICD1, Hes1, and WISP-1 in CAFs by ApoSQ treatment was suppressed (FIG. 30(A)), and WISP-1 secretion was also reduced ( Figure 30 (B)). As a result, when CAFs were treated with DAPT, the anti-migration and anti-invasion effects of ApoSQ-CAF CM were significantly inhibited in 344SQ (FIG. 31a). Similarly, the anti-migration and anti-invasion effects of ApoSQ were also completely inhibited by DAPT in CAFs themselves (FIG. 31B).

상기 실험을 통해 CAFs와 ApoSQ를 접촉시킬 경우, Notch1 신호가 활성화되어 WISP-1 발현이 증가하고 이로 인해 344SQ 세포 및 CAFs에서 세포의 이동 및 침윤이 억제됨을 확인함으로써, ApoSQ-CAF CM의 암 세포 이동 및 침윤 억제 효과 그리고 ApoSQ의 CAFs 이동 및 침윤 억제 효과가 Noth1-의존적인 WIPS-1 생성을 통하여 이루어짐을 검증하였다.Through the above experiment, when CAFs and ApoSQ are brought into contact, Notch1 signal is activated and WISP-1 expression is increased, which suppresses cell migration and invasion in 344SQ cells and CAFs, thereby confirming the cancer cell migration of ApoSQ-CAF CM and invasion inhibitory effect and ApoSQ's CAFs migration and invasion inhibitory effect were verified through Noth1-dependent WIPS-1 production.

2-5. UV 조사된 사멸화된 암 세포에서 Dll1의 발현이 증가함을 확인2-5. Confirmation of increased expression of Dll1 in UV-irradiated apoptotic cancer cells

대부분의 Notch 신호전달은 세포-표면 수용체 Notch와 세포 결합 리 간드, 예컨대 Delta-like ligand(DLL) 1,3 및 4 또는 Jagged-like(JAG) 1 및 2의 상호작용에 의해 개시되므로, Notch-1 신호전달의 활성화에 필요한 중요한 세포 표면 상호작용을 강조하기 위해, 본 발명자들은 먼저 유세포 분석을 사용하여 사멸화된 세포 표면상의 Notch 리간드의 발현을 조사하였다.Most Notch signaling is initiated by the interaction of cell-surface receptor Notch with cell-associated ligands, such as Delta-like ligands (DLL) 1,3 and 4 or Jagged-like (JAG) 1 and 2, so Notch- To highlight the important cell surface interactions required for activation of 1 signaling, we first examined the expression of Notch ligand on the surface of apoptotic cells using flow cytometry.

그 결과, 15분 동안 UV 조사 후 344SQ, A549 및 HCT116을 포함하는 암 세포에서 Dll1이 유의하게 증가되었음을 확인하였으나, 다른 리간드의 발현 수준은 변화되지 않았거나 오히려 감소되었고(도 32), 웨스턴 블랏팅 분석 결과는, 사멸화된 암 세포 용해물에서 Dll1이 유일하게 발현이 증가되었음을 보여준다(도 33).As a result, it was confirmed that Dll1 was significantly increased in cancer cells including 344SQ, A549 and HCT116 after UV irradiation for 15 minutes, but the expression levels of other ligands were not changed or rather decreased (FIG. 32), and Western blotting The analysis results show that the only expression of Dll1 increased in the killed cancer cell lysate (FIG. 33).

또한, Dll1을 항-Dll1 항체로 중화시킨 경우, ApoSQ가 유도하는 Notch1 신호가 감소하였고, CAFs의 WISP-1 분비 역시 감소하였으며(도 34), 이와 유사하게 344SQ 세포에서 UV 조사를 통한 사멸화 전에 특이적 siRNA를 이용해 Dll1을 넉다운시킨 경우, ApoSQ 처리 후 CAFs에서 Notch1 활성화와 WISP-1 분비가 억제되었다(도 35a 및 35b). In addition, when Dll1 was neutralized with an anti-Dll1 antibody, the Notch1 signal induced by ApoSQ was reduced, and the secretion of WISP-1 in CAFs was also reduced (FIG. 34). Similarly, before killing through UV irradiation in 344SQ cells, When Dll1 was knocked down using specific siRNA, Notch1 activation and WISP-1 secretion were suppressed in CAFs after ApoSQ treatment (FIGS. 35a and 35b).

상기 실험 결과들은 CAFs에서의 Notch1 신호전달 과정이 Dll1을 발현하는 인접한 사멸화된 암 세포와의 상호작용에 의해 개시됨을 의미한다.The above experimental results indicate that the Notch1 signaling process in CAFs is initiated by interaction with adjacent killed cancer cells expressing Dll1.

2-6. BAI1/Rac1 신호의 CAFs에 의한 사멸세포 탐식 작용(efferocytosis) 촉진 및 Notch1 신호와의 크로스톡(crosstalk) 확인2-6. Promote phagocytosis of apoptotic cells by CAFs of BAI1/Rac1 signals and confirm crosstalk with Notch1 signals

BAI1은 1차 섬유아세포를 통해 사멸화된 자궁경부암 세포의 사멸세포 제거과정에 기여할 수 있으므로, BAI1 신호가 CAFs에서 Notch1 신호에 영향을 줄 수 있는지 확인하고자 실험을 진행하였다. BAI1의 thrombospondin type 1 repeats이 직접 포스파티딜세린(phosphatidylserine, PtdSer)을 인식하여 사멸화된 세포를 표적하기 때문에, 사멸화된 세포의 PtdSer에 결합하는 annexin V를 첨가하여 CAFs에 BAI1과 ApoSQ 간의 상호작용을 저해함으로써 Notch1 신호를 억제하는지 여부를 조사하였다. 유세포 분석을 이용하여 PKH67로 염색된 ApoSQ 세포를 포식한 PKH26로 염색된 CAFs의 비율을 측정하였다. Annexin V를 첨가한 경우, CAFs에 의한 ApoSQ의 탐식 작용이 감소하였으며(도 36a), ApoSQ가 유도하는 Notch1 활성화 및 WISP-1의 분비 역시 하향 조절되었다(도 36b). 또한, BAI1을 넉다운시킨 경우, ApoSQ로 처리한 CAFs에서 탐식 작용, Notch1 신호 및 WISP-1의 분비가 하향 조절되었고(도 37a 내지 37c), BAI1을 항-BAI1 항체로 중화시킨 경우에도 같은 결과를 확인하였다(도 38a 및 38b). 이와 반대로, BAI1 과발현 CAFs의 경우, 모의 형질감염 세포에 비해 ApoSQ 탐식 작용 비율이 더 높았으며, Notch1 신호 및 WISP-1 분비 역시 더 증가하였다(도 39a 내지 39c).Since BAI1 can contribute to the apoptotic cell removal process of apoptotic cervical cancer cells through primary fibroblasts, an experiment was conducted to confirm whether BAI1 signal could affect Notch1 signal in CAFs. Since the thrombospondin type 1 repeats of BAI1 directly recognize phosphatidylserine (PtdSer) and target apoptotic cells, the interaction between BAI1 and ApoSQ in CAFs was reduced by adding annexin V that binds to PtdSer in apoptotic cells. Whether or not Notch1 signaling was suppressed by inhibition was investigated. The percentage of PKH26-stained CAFs that phagocytosed PKH67-stained ApoSQ cells was determined using flow cytometry. When Annexin V was added, the phagocytosis of ApoSQ by CAFs was reduced (FIG. 36a), and ApoSQ-induced Notch1 activation and WISP-1 secretion were also downregulated (FIG. 36b). In addition, when BAI1 was knocked down, phagocytosis, Notch1 signaling, and WISP-1 secretion were downregulated in ApoSQ-treated CAFs (FIGS. 37a to 37c), and the same results were obtained when BAI1 was neutralized with an anti-BAI1 antibody. It was confirmed (FIG. 38a and 38b). Conversely, in the case of BAI1 overexpressing CAFs, the ApoSQ phagocytosis rate was higher than that of mock-transfected cells, and Notch1 signaling and WISP-1 secretion were also increased (FIGS. 39a to 39c).

상기 실험 결과들은 ApoSQ에 결합하는 단백질인 BAI1는 CAFs의 사멸세포 제거작용을 촉진하며, 더불어 Notch1 신호경로에 관여하여 CAFs의 WISP-1 생산을 조절함을 의미한다.The above experimental results indicate that BAI1, a protein that binds to ApoSQ, promotes the removal of apoptotic cells in CAFs and regulates the production of WISP-1 in CAFs by being involved in the Notch1 signaling pathway.

추가적으로, CAFs의 사멸세포 제거과정과 Notch1 신호 사이의 관계를 자세히 분석하기 위하여, 사멸화된 세포의 탐식 작용을 하향 조절하는 Rac1 억제제 NSC23766을 사용하였다. ApoSQ가 CAFs에서 Rac1의 활성을 시간 의존적으로 증가시켰고, 24시간 후에 Rac1 활성이 가장 높았다(도 40). 유세포 분석 결과와 유사하게, 100 μM의 NSC23766 처리 시, CAFs의 탐식 작용이 감소하는 것을 공초점 현미경으로 확인하였다. 또한 NSC23766 처리시 Notch1 신호 및 WISP-1 분비가 감소하는 것을 확인하였다(도 41a 내지 41c).Additionally, in order to analyze the relationship between the apoptotic cell elimination process of CAFs and the Notch1 signal in detail, the Rac1 inhibitor NSC23766, which down-regulates the phagocytosis of apoptotic cells, was used. ApoSQ increased Rac1 activity in CAFs in a time-dependent manner, and Rac1 activity was highest after 24 hours (FIG. 40). Similar to the results of flow cytometry, it was confirmed by confocal microscopy that phagocytosis of CAFs was reduced when treated with 100 μM of NSC23766. In addition, it was confirmed that Notch1 signal and WISP-1 secretion were decreased when treated with NSC23766 (FIGS. 41a to 41c).

또한, Notch1을 넉다운시킨 경우 Rac1 활성의 감소 및 CAFs의 ApoSQ 제거작용이 감소하였으며(도 42), 20 mM의 DAPT 처리 시에도 Rac1의 활성과 CAFs의 ApoSQ 제거작용이 감소하였다(도 43).In addition, when Notch1 was knocked down, Rac1 activity and ApoSQ scavenging action of CAFs were reduced (FIG. 42), and Rac1 activity and ApoSQ scavenging action of CAFs were also reduced when 20 mM DAPT was treated (FIG. 43).

상기 실험 결과들은 Notch1 신호와 BAI1/Rac1 신호 경로가 사멸화된 암 세포의 자극시 긍정적인 크로스톡을 나타낼 수 있음을 시사한다.The above experimental results suggest that the Notch1 signal and the BAI1/Rac1 signal pathway can exhibit positive crosstalk when apoptotic cancer cells are stimulated.

2-7. 2-7. in vivoin vivo 에서 ApoSQ 처리 시 Notch1 신호에 의해 WISP-1 생산을 촉진하고, CAF 활성화 및 폐 전이를 억제함을 확인confirmed that WISP-1 production was promoted by Notch1 signaling during ApoSQ treatment, and CAF activation and lung metastasis were suppressed.

본 발명자는 이전 연구에서 동계(syngeneic)(129/Sv) 면역적격 마우스에서 ApoSQ 세포의 단일 주입이 폐 전이를 억제함을 확인하였는 바, 본 실험에서는 이 동계 마우스를 사용하여, 344SQ 세포를 피하 주사 후 ApoSQ 주사에 대한 CAFs의 생체내 반응을 조사하였다. 이를 위해, 먼저 마우스 원발성 종양으로부터 백혈구, 내피 세포 및 상피 세포를 제거한 후, 섬유아세포-특이적 마커인 Thy1을 사용한 자기-활성화 세포 분류를 이용하여 CAFs를 분리하고(도 44), qRT-PCR을 사용하여 CAF 마커의 mRNA 양을 분석하였다.In a previous study, the present inventors confirmed that a single injection of ApoSQ cells inhibited lung metastasis in syngeneic (129/Sv) immunocompetent mice. After that, the in vivo response of CAFs to ApoSQ injection was investigated. To this end, leukocytes, endothelial cells, and epithelial cells were first removed from mouse primary tumors, and then CAFs were isolated using magnetic-activated cell sorting using the fibroblast-specific marker Thy1 (FIG. 44), followed by qRT-PCR. used to analyze the amount of mRNA of CAF markers.

그 결과, ApoSQ 주입 후 Acta2, Col1α1, Fn, Itg beta 1, Spp1, Pdgfrα, Pdgfr beta와 같은 전형적인 CAF 활성화 마커, Mmp1a, 2, 9 및 12, 그리고 Vegfa, Hgf, Cxcl12 및 Cxcl14를 포함하는 성장 인자/케모카인의 mRNA 발현 수준이 ApoSQ 주입 후 대조군과 비교하여 유의하게 감소되었다. 그러나, Notch1과 WISP-1(Ccn4), Hey1, Hey2, Hes1 및 Hes5를 포함하는 Notch 하향 표적 유전자의 mRNA 발현 수준은 대조군과 비교하여 현저히 향상되었으며, Notch 리간드 및 다른 CCN 유전자 패밀리(Ccn1, Ccn2, Ccn5 및 Ccn6)의 mRNA 발현양은 ApoSQ 주입 후 거의 변하지 않았으나, Ccn3 mRNA는 대조군과 비교하여 ApoSQ 주입 후 Thy1+ CAFs에서 대략 2배 증가되었다(도 45a 및 도 45b).As a result, typical CAF activation markers such as Acta2, Col1α1, Fn, Itg beta 1, Spp1, Pdgfrα, Pdgfr beta, growth factors including Mmp1a, 2, 9 and 12, and Vegfa, Hgf, Cxcl12 and Cxcl14 after ApoSQ injection / The mRNA expression levels of chemokines were significantly reduced after ApoSQ injection compared to the control group. However, mRNA expression levels of Notch downlink target genes including Notch1, WISP-1 (Ccn4), Hey1, Hey2, Hes1 and Hes5 were significantly improved compared to the control group, and notch ligands and other CCN gene families (Ccn1, Ccn2, The mRNA expression levels of Ccn5 and Ccn6) were almost unchanged after ApoSQ injection, but Ccn3 mRNA was approximately doubled in Thy1 + CAFs after ApoSQ injection compared to the control group (FIGS. 45a and 45b).

생체 내에서 CAF 활성화 및 종양 발달에 있어 Notch1-WISP-1 신호의 억제 효과를 확인하기 위해, Notch1 선택적 억제제인 LY3039478을 ApoSQ 주사 전날 8 mg/kg으로 6주간 주 3회씩 경구 투여하였다(도 46). LY3039478은 대조군 및 ApoSQ 그룹과 대비하였을 때 체중, 종양 중량 또는 종양 부피를 거의 변화시키지 않았으나(도 47), ApoSQ 투여에 의한 폐 표면의 종양 결절 수 및 전이 비율 감소 효과를 역전시켰다(도 48).To confirm the inhibitory effect of Notch1-WISP-1 signaling on CAF activation and tumor development in vivo, LY3039478, a Notch1 selective inhibitor, was orally administered at 8 mg/kg the day before ApoSQ injection, 3 times a week for 6 weeks (FIG. 46). . LY3039478 hardly changed body weight, tumor weight or tumor volume when compared with the control and ApoSQ groups (FIG. 47), but reversed the effect of ApoSQ administration on reducing the number of tumor nodules on the lung surface and the rate of metastasis (FIG. 48).

또한, LY3039478은 분리된 Thy1+ CAFs에서 ApoSQ 투여에 의한 CAF 마커, MMP, 성장 인자 및 케모카인의 mRNA 발현양의 감소 효과를 역전시켰으며, Notch1 및 Notch 표적 유전자의 mRNA 발현양의 증가 효과 또한 역전시켰다(도 49). 원발성 종양 조직을 면역조직화학 분석한 결과, LY3039478은 Thy1+ CAFs에서 α-SMA의 감소 및 NICD1 및 WISP-1 발현의 증가 효과 역시 역전시켰다(도 50). 또한, 원발성 종양으로부터 분리된 Thy1+ CAFs를 면역세포화학 분석한 결과, 단백질 발현 양상이 역전되었음을 확인하였다(도 51a 및 51b).In addition, LY3039478 reversed the effect of ApoSQ administration on the decreased mRNA expression level of CAF markers, MMPs, growth factors, and chemokines in isolated Thy1 + CAFs, and also reversed the effect of increasing the mRNA expression level of Notch1 and Notch target genes. (FIG. 49). As a result of immunohistochemical analysis of primary tumor tissue, LY3039478 also reversed the effect of decreasing α-SMA and increasing NICD1 and WISP-1 expression in Thy1 + CAFs (FIG. 50). In addition, as a result of immunocytochemical analysis of Thy1 + CAFs isolated from the primary tumor, it was confirmed that the protein expression pattern was reversed (FIGS. 51a and 51b).

한편, ApoSQ가 투여된 마우스군의 원발성 종양조직에서 분리된 Thy1+ CAFs의 배양 배지와 혈청에서 증가한 WISP-1 단백질이 LY3039478을 처리할 경우 감소하였으나, ApoSQ 투여군과 ApoSQ + LY3039478 투여 군의 원발성 종양조직에서 분리된 CD326+ 종양 세포 및 CD11b+ 종양 관련 대식세포의 배양 배지에서, 대조군과 비교시, WISP-1 단백질 수준의 변화가 나타나지 않았다(도 52).On the other hand, the increased WISP-1 protein in the culture medium and serum of Thy1 + CAFs isolated from the primary tumor tissue of the ApoSQ-administered mouse group was decreased when LY3039478 was treated, but the primary tumor tissue of the ApoSQ-administered group and the ApoSQ + LY3039478-administered group In the culture media of CD326 + tumor cells and CD11b + tumor-associated macrophages isolated from WISP-1, no change in protein level was observed compared to the control group (FIG. 52).

또한, ApoSQ를 주사할 경우, 분리된 CD326+ 종양 세포 및 Thy1+ CAFs에 이동 및 침윤이 억제되었고, LY3039478을 함께 투여할 경우 이러한 효과가 역전되었다(도 53a 및 53b). 이와 유사하게, ApoSQ 주사 시 분리된 CD326+ 종양 세포에서 Smad2/3, FAK, ERK 및 Akt와 같은 이동 및 침윤에 관련된 신호분자의 활성화가 억제되고, MMP2 및 MMP12와 같은 단백질 발현이 억제되나, LY3039478을 투여할 경우 이러한 억제 효과가 역전되었다(도 54).In addition, injection of ApoSQ inhibited the migration and invasion of isolated CD326 + tumor cells and Thy1 + CAFs, and these effects were reversed when co-administered with LY3039478 (FIGS. 53A and 53B). Similarly, activation of signaling molecules related to migration and invasion, such as Smad2/3, FAK, ERK, and Akt, and protein expression, such as MMP2 and MMP12, were suppressed in isolated CD326 + tumor cells upon injection of ApoSQ, but LY3039478 Administration reversed this inhibitory effect (FIG. 54).

상기 실험을 통해 in vivo에서 ApoSQ 투여 시 CAFs에서 Notch1 신호가 활성화되어 WISP-1 생성이 촉진되고, 이에 따라 CAF 활성화 마커의 발현양이 감소하여 암 세포 및 CAFs의 이동 및 침윤이 억제됨을 검증하였다.Through the above experiment, it was verified that when ApoSQ was administered in vivo , Notch1 signal was activated in CAFs, WISP-1 production was promoted, and the expression level of CAF activation markers was reduced, thereby inhibiting migration and invasion of cancer cells and CAFs.

2-8. ApoSQ-CAF CM이 2-8. ApoSQ-CAF CM in vivoin vivo 에서 종양 진행 및 폐 전이를 억제함을 확인confirmed to inhibit tumor progression and lung metastasis in

ApoSQ-CAF CM의 종양 진행 및 폐 전이에 대한 in vivo 효과를 조사하기 위해, CAF CM 또는 ApoSQ-CAF CM을 344SQ 주사 2일 후에 동계 마우스에 1주 3회, 6주 동안 종양내로 주사하였다. 또한, 생체 내에서 CM 내 WISP-1의 역할을 확인하기 위해, ApoSQ-CAF CM을 주입하기 전에 2시간 동안 중화 항-WISP-1 항체 또는 이소타입 IgG로 전처리하였다(도 55).To investigate the in vivo effect of ApoSQ-CAF CM on tumor progression and lung metastasis, CAF CM or ApoSQ-CAF CM was intratumorally injected 3 times a week for 6 weeks into syngeneic mice 2 days after 344SQ injection. In addition, to confirm the role of WISP-1 in CM in vivo, ApoSQ-CAF CM was pretreated with neutralizing anti-WISP-1 antibody or isotype IgG for 2 hours before injection (FIG. 55).

그 결과, 체중은 그룹 간 유의미한 차이가 없었다(도 56). 또한, 단일 주사 ApoSQ의 효과와 마찬가지로, ApoSQ-CAF CM을 주사한 군은 대조군과 비교하여 344SQ 주사 6주 후에 폐 표면 상의 종양 결절의 수와 전이 비율이 감소하였고, 이소타입 IgG 투여 그룹에서도 같은 양상을 보였으나, WISP-1이 면역제거된 ApoSQ-CAF CM 그룹에서는 종양 결절 수 및 전이 비율에 변화가 없음을 확인하였다(도 57).As a result, there was no significant difference in body weight between groups (FIG. 56). In addition, similar to the effect of single injection ApoSQ, the group injected with ApoSQ-CAF CM decreased the number of tumor nodules and metastasis rate on the lung surface 6 weeks after 344SQ injection compared to the control group, and the same pattern was observed in the isotype IgG administered group. However, in the ApoSQ-CAF CM group in which WISP-1 was immunosuppressed, it was confirmed that there was no change in the number of tumor nodules and the rate of metastasis (FIG. 57).

in vitro 실험 결과와 유사하게, ApoSQ-CAF CM의 주입은, 분리된 CD326+ 종양 세포와 Thy1+ CAFs의 이동 및 침윤을 현저히 감소시킨 반면, WISP-1이 면역제거된 ApoSQ-CAF CM 그룹에서는 이러한 억제 효과가 나타나지 않았다(도 58a 및 58b). 또한, 분리된 CD326+ 종양 세포 내에서 이동 및 침윤에 관여하는 신호경로, 즉 Smad2/3, FAK, ERK, Akt의 활성화 및 MMP2 및 MMP12의 발현양이 ApoSQ-CAF CM에 의해 감소하였으나, WISP-1이 면역제거된 ApoSQ-CAF CM 그룹에서는 이러한 억제 효과를 보이지 않았다(도 59). 또한 분리된 Thy1+ CAFs에서 CAF 활성 마커, 즉 Acta2, Col1α1, Itg beta 1, Spp1, Pdgfrα 와 Mmp1a, Mmp2, 및 Mmp12의 mRNA 발현 양이 ApoSQ-CAF CM에 의해 감소하였으나, WISP-1이 면역제거된 ApoSQ-CAF CM 그룹에서는 이러한 억제 효과를 보이지 않았다(도 60).Similar to the results of in vitro experiments, injection of ApoSQ-CAF CM significantly reduced the migration and invasion of isolated CD326 + tumor cells and Thy1 + CAFs, whereas in the WISP-1 immunodepleted ApoSQ-CAF CM group, these No inhibitory effect was seen (FIGS. 58A and 58B). In addition, the activation of signaling pathways involved in migration and invasion, namely Smad2/3, FAK, ERK, and Akt, and the expression levels of MMP2 and MMP12 in isolated CD326 + tumor cells were decreased by ApoSQ-CAF CM, but WISP- The ApoSQ-CAF CM group in which 1 was immunodepleted did not show this inhibitory effect (FIG. 59). In addition, the mRNA expression levels of CAF activity markers, namely Acta2, Col1α1, Itg beta 1, Spp1, Pdgfrα and Mmp1a, Mmp2, and Mmp12, in isolated Thy1 + CAFs were decreased by ApoSQ-CAF CM, but WISP-1 was immunoremoved. The ApoSQ-CAF CM group showed no such inhibitory effect (FIG. 60).

또한, WISP-1의 항 종양 진행 및 항 전이에 대한 in vivo 효과를 확인하기 위해, rWISP1-1을 12.5 μg/kg 또는 25 μg/kg의 농도로 344SQ 주사 2일 후에 동계 마우스에 1주 3회, 6주 동안 종양내로 주사하였다(도 61)In addition, to confirm the in vivo effect of WISP-1 on anti-tumor progression and anti-metastasis, rWISP1-1 was injected at a concentration of 12.5 μg/kg or 25 μg/kg 344SQ 2 days after injection to syngeneic mice 3 times a week , injected intratumorally for 6 weeks (FIG. 61)

그 결과, ApoSQ-CAF CM을 주사한 경우와 유사하게, 체중은 그룹 간 유의미한 차이가 없었고(도 62), rWISP-1을 주사한 군은 대조군과 비교하여 폐 표면 상의 종양 결절의 수가 감소하였으며, 전이 비율은 농도 의존적으로 감소하였다(도 63). 또한, rWISP-1은 CD326+ 종양 세포와 Thy1+ CAFs의 이동 및 침윤을 농도 의존적으로 억제하였으며(도 64a 및 64b), CD326+ 종양 세포 내에서 이동 및 침윤에 관여하는 신호경로, 즉 Smad2/3, FAK, ERK, Akt의 활성화 및 MMP2 및 MMP12의 발현양을 감소시켰고(도 65), Thy1+ CAFs에서 CAF 활성 마커, 즉 Acta2, Col1α1, Itg beta 1, Spp1, Pdgfrα 와 Mmp1a, Mmp2, 및 Mmp12의 mRNA 발현양을 감소시켰다(도 66).As a result, similar to the case of ApoSQ-CAF CM injection, there was no significant difference in body weight between the groups (FIG. 62), and the number of tumor nodules on the lung surface was reduced in the group injected with rWISP-1 compared to the control group. The transition rate decreased in a concentration dependent manner (FIG. 63). In addition, rWISP-1 inhibited the migration and invasion of CD326 + tumor cells and Thy1 + CAFs in a concentration-dependent manner (Fig. 64a and 64b), and the signaling pathway involved in migration and invasion in CD326 + tumor cells, that is, Smad2/3 , FAK, ERK, Akt activation and MMP2 and MMP12 expression levels were reduced (FIG. 65), and CAF activity markers, namely Acta2, Col1α1, Itg beta 1, Spp1, Pdgfrα and Mmp1a, Mmp2, and Mmp12 in Thy1 + CAFs The mRNA expression level of was decreased (FIG. 66).

상기 실험을 통해, ApoSQ-CAF CM의 종양 억제 효과가 WISP-1에 의해 매개되는 것을 입증하였으며, rWISP-1이 in vivo에서 ApoSQ-CAF CM의 효과와 동일한 효과를 나타낼 수 있음을 확인하였다.Through the above experiments, it was demonstrated that the tumor suppression effect of ApoSQ-CAF CM was mediated by WISP-1, and it was confirmed that rWISP-1 could exhibit the same effect as that of ApoSQ-CAF CM in vivo .

추가적으로, ApoSQ-CAF CM의 생체 내 항 전이 효과를 유전자 수준에서 분석하기 위해, 분리된 CD326+ 종양 세포에서 마우스 종양 전이 RT2 Profiler PCR 어레이를 사용하여 침윤 및 전이에 관한 84개의 유전자 발현을 동시에 분석하였다. 그 결과, CAF CM 그룹과 비교하여, ApoSQ-CAF CM 주사 시 종양 진행 및 침윤에 관한 유전자 MMP2, Cdh6, Smad2, Kras, Igf1 등이 포함된 15종이 발현이 2배 이상 하향 조절되었고, 종양 진행 및 침윤 억제에 관한 유전자 (Cdkn2a, Ror beta, Ctnna1 등이 포함된 8종) 발현이 2배이상 상향 조절되었다(도 67).Additionally, to analyze the in vivo anti-metastatic effect of ApoSQ-CAF CM at the gene level, we simultaneously analyzed the expression of 84 genes related to invasion and metastasis using the mouse tumor metastasis RT 2 Profiler PCR array in isolated CD326 + tumor cells. did As a result, compared to the CAF CM group, the expression of 15 genes, including MMP2, Cdh6, Smad2, Kras, and Igf1, related to tumor progression and invasion, were down-regulated more than 2-fold when injected with ApoSQ-CAF CM, and tumor progression and The expression of genes related to inhibition of invasion (8 types including Cdkn2a, Ror beta, Ctnna1, etc.) was up-regulated more than twice (FIG. 67).

또한, ApoSQ-CAF CM이 CAFs의 침윤을 감소시키는 메커니즘을 검증하기 위하여, 분리된 Thy1+ CAF에서 세포 접착 및 ECM 리모델링에 관련된 84개의 유전자를 qRT-PCR 어레이를 통해 분석하였다. 그 결과, CAF CM 그룹과 비교하여, ApoSQ-CAF CM 그룹의 경우 Cdh2, Selp, Itg beta 1, Cdh3, Itga3, Ncam1, Vacm1, 및 Ncam2와 같은 8개의 세포 응집 관련 유전자 발현이 2배 이상 하향 조절되었고, ECM 리모델링 관련 유전자, 즉 ECM component (4종의 유전자) 및 MMPs(8종의 유전자)의 발현 또한 2배 이상 하향 조절되었다(도 68).In addition, to verify the mechanism by which ApoSQ-CAF CM reduces CAF invasion, 84 genes related to cell adhesion and ECM remodeling in isolated Thy1 + CAFs were analyzed by qRT-PCR array. As a result, compared to the CAF CM group, in the case of the ApoSQ-CAF CM group, the expression of 8 cell aggregation-related genes, such as Cdh2, Selp, Itg beta 1, Cdh3, Itga3, Ncam1, Vacm1, and Ncam2, was downregulated more than 2-fold. and the expression of ECM remodeling-related genes, that is, ECM components (4 genes) and MMPs (8 genes), was also down-regulated more than 2-fold (FIG. 68).

상기 실험을 통해 in vivo에서 ApoSQ-CAF CM을 투여할 경우 암 세포의 침윤 및 전이 그리고 CAFs의 침윤과 관련된 유전자 발현을 감소시킴으로써, 암 세포 및 CAFs의 이동 및 침윤을 억제할 수 있음을 검증하였다.Through the above experiments, it was verified that the in vivo administration of ApoSQ-CAF CM can inhibit the migration and invasion of cancer cells and CAFs by reducing the expression of genes related to cancer cell invasion and metastasis and CAFs invasion.

전술한 본 발명의 설명은 예시를 위한 것이며, 본 발명이 속하는 기술분야의 통상의 지식을 가진 자는 본 발명의 기술적 사상이나 필수적인 특징을 변경하지 않고서 다른 구체적인 형태로 쉽게 변형이 가능하다는 것을 이해할 수 있을 것이다. 그러므로 이상에서 기술한 실시예들은 모든 면에서 예시적인 것이며 한정적이 아닌 것으로 이해되어야 한다.The above description of the present invention is for illustrative purposes, and those skilled in the art can understand that it can be easily modified into other specific forms without changing the technical spirit or essential features of the present invention. will be. Therefore, it should be understood that the embodiments described above are illustrative in all respects and not restrictive.

Claims (26)

암 관련 섬유아세포(Cancer-Associated Fibroblasts, CAFs) 및 사멸화된 암 세포(apoptotic cancer cells)를 공동 배양한 배양액을 함유하는, 암 전이 억제용 약학적 조성물.Cancer-associated fibroblasts (CAFs) and apoptotic cancer cells (apoptotic cancer cells) containing the co-cultivated culture medium, a pharmaceutical composition for inhibiting cancer metastasis. 청구항 1에 있어서, 상기 암 관련 섬유아세포는 섬유육종, 악성섬유성조직구종, 지방육종, 횡문 근육종, 평활근육종, 혈관육종, 악성피부암, 림프혈관육종, 활막육종, 연골육종, 골육종, 폐암, 위암, 유방암, 대장암 및 전립선암으로 이루어진 군에서 선택된 하나 이상의 암과 관련된 섬유아세포인 것인, 약학적 조성물.The method according to claim 1, wherein the cancer-related fibroblasts are fibrosarcoma, malignant fibrous histiocytoma, liposarcoma, rhabdomyosarcoma, leiomyosarcoma, angiosarcoma, malignant skin cancer, lymphangiosarcoma, synovial sarcoma, chondrosarcoma, osteosarcoma, lung cancer, gastric cancer , Breast cancer, colorectal cancer and prostate cancer will be one or more cancer-related fibroblasts selected from the group consisting of, a pharmaceutical composition. 청구항 1에 있어서, 상기 사멸화된 암 세포는 자외선(Ultra-violet ray, UV) 조사에 의해 사멸된 것임을 특징으로 하는, 약학적 조성물.The pharmaceutical composition according to claim 1, wherein the killed cancer cells are killed by ultraviolet (Ultra-violet ray, UV) irradiation. 청구항 1에 있어서, 상기 사멸화된 암 세포의 암종은 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 선택된 하나 이상인 것인, 약학적 조성물.The method according to claim 1, wherein the carcinoma of the killed cancer cells is breast cancer, uterine cancer, esophageal cancer, stomach cancer, brain cancer, rectal cancer, colon cancer, lung cancer, skin cancer, ovarian cancer, cervical cancer, hematological cancer, pancreatic cancer, prostate cancer, testicular cancer, laryngeal cancer , Oral cancer, head and neck cancer, thyroid cancer, liver cancer, bladder cancer, osteosarcoma, at least one selected from the group consisting of lymphoma and leukemia, a pharmaceutical composition. 청구항 4에 있어서, 상기 암종은 폐암, 유방암, 위암, 대장암 및 전립선암으로 이루어진 군에서 선택된 하나 이상인 것인, 약학적 조성물.The pharmaceutical composition according to claim 4, wherein the carcinoma is at least one selected from the group consisting of lung cancer, breast cancer, stomach cancer, colon cancer and prostate cancer. 청구항 1에 있어서, 상기 배양액은 WISP-1(Wnt-induced signaling protein-1)을 함유하는 것을 특징으로 하는, 약학적 조성물.The pharmaceutical composition according to claim 1, wherein the culture medium contains WISP-1 (Wnt-induced signaling protein-1). 청구항 6에 있어서, 상기 WISP-1은 Notch1 시그널링에 의해 생성되는 것임을 특징으로 하는, 약학적 조성물.The pharmaceutical composition according to claim 6, wherein the WISP-1 is produced by Notch1 signaling. 청구항 7에 있어서, 상기 Notch1 시그널링은 Dll1(Delta-like ligand 1)에 의해 개시되는 것임을 특징으로 하는, 약학적 조성물.The pharmaceutical composition according to claim 7, wherein the Notch1 signaling is initiated by Dll1 (Delta-like ligand 1). 청구항 7에 있어서, 상기 Notch1 시그널링은 BAI1/Rac1(Brain angiogenesis inhibitor 1/Ras-related C3 botulinum toxin substrate 1) 시그널링에 의해 강화되는 것인, 약학적 조성물.The pharmaceutical composition of claim 7, wherein the Notch1 signaling is enhanced by BAI1/Rac1 (Brain angiogenesis inhibitor 1/Ras-related C3 botulinum toxin substrate 1) signaling. 청구항 9에 있어서, 상기 조성물은 Notch1 시그널링과 BAI1/Rac1 시그널링의 상호작용에 의해 암 관련 섬유아세포의 탐식능을 강화하는 것인, 약학적 조성물.The pharmaceutical composition according to claim 9, wherein the composition enhances the phagocytic ability of cancer-related fibroblasts by the interaction of Notch1 signaling and BAI1/Rac1 signaling. 청구항 9에 있어서, 상기 조성물은 Notch1 시그널링과 BAI1/Rac1 시그널링의 상호작용에 의해 WISP-1 생성을 강화하는 것인, 약학적 조성물.The pharmaceutical composition according to claim 9, wherein the composition enhances WISP-1 production by the interaction of Notch1 signaling and BAI1/Rac1 signaling. 청구항 1에 있어서, 상기 조성물은 암 관련 섬유아세포에서 활성화 마커를 감소시키는 것을 특징으로 하는, 약학적 조성물.The pharmaceutical composition according to claim 1, wherein the composition reduces activation markers in cancer-related fibroblasts. 청구항 12에 있어서, 상기 활성화 마커는 Acta2, Col1α1, Fn, Itg beta 1, Spp1, Pdgfrα, Pdgfr beta 및 Mmp1a, 2, 9 및 12로 이루어진 군에서 선택되는 하나 이상인 것인, 약학적 조성물.The pharmaceutical composition according to claim 12, wherein the activation marker is one or more selected from the group consisting of Acta2, Col1α1, Fn, Itg beta 1, Spp1, Pdgfrα, Pdgfr beta and Mmp1a, 2, 9 and 12. 청구항 1에 있어서, 상기 조성물은 암 관련 섬유아세포에서 성장 인자 및 케모카인을 감소시키는 것을 특징으로 하는, 약학적 조성물.The pharmaceutical composition according to claim 1, characterized in that the composition reduces growth factors and chemokines in cancer-associated fibroblasts. 청구항 14에 있어서, 상기 성장 인자는 Vegfa, Hgf, Cxcl12 및 Cxcl14로 이루어진 군에서 선택되는 하나 이상인 것인, 약학적 조성물.The pharmaceutical composition according to claim 14, wherein the growth factor is at least one selected from the group consisting of Vegfa, Hgf, Cxcl12 and Cxcl14. 청구항 1에 있어서, 상기 조성물은 암 관련 섬유아세포에서 Notch1 관련 분자를 증가시키는 것을 특징으로 하는, 약학적 조성물.The pharmaceutical composition according to claim 1, wherein the composition increases Notch1-related molecules in cancer-related fibroblasts. 청구항 16에 있어서, 상기 Notch1 관련 분자는 WISP-1(Ccn4), Hey1, Hey2, Hes1 및 Hes5로 이루어진 군에서 선택되는 하나 이상인 것인, 약학적 조성물.The pharmaceutical composition according to claim 16, wherein the Notch1-related molecule is one or more selected from the group consisting of WISP-1 (Ccn4), Hey1, Hey2, Hes1 and Hes5. 청구항 1에 있어서, 상기 암 전이는 암 세포의 이동 및 침윤을 포함하는 것을 특징으로 하는, 약학적 조성물.The pharmaceutical composition according to claim 1, wherein the cancer metastasis includes migration and invasion of cancer cells. 사멸화된 암 세포에 노출된 암 관련 섬유아세포를 함유하는, 암 전이 억제용 약학적 조성물.A pharmaceutical composition for inhibiting cancer metastasis, containing cancer-related fibroblasts exposed to apoptotic cancer cells. 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 함유하는, 암 전이 억제용 건강기능식품.A health functional food for inhibiting cancer metastasis, containing a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured. 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양하는 단계를 포함하는, 암 전이 억제용 조성물의 제조방법.A method for preparing a composition for inhibiting cancer metastasis, comprising co-cultivating cancer-related fibroblasts and apoptotic cancer cells. 청구항 21에 있어서, 상기 공동 배양을 통해 WISP-1을 분비시키는 단계를 더 포함하는, 암 전이 억제용 조성물의 제조방법.The method of claim 21, further comprising secreting WISP-1 through the co-culture. 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 개체에 처리하는 단계를 포함하는, 암 전이 억제방법.A method for suppressing cancer metastasis, comprising the step of treating a subject with a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured. 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액을 개체에 처리하는 단계를 포함하는, 암 예방 또는 치료방법.A method for preventing or treating cancer, comprising the step of treating a subject with a culture medium in which cancer-related fibroblasts and apoptotic cancer cells are co-cultured. 암 관련 섬유아세포 및 사멸화된 암 세포를 공동 배양한 배양액의 암 예방 또는 치료 용도.Use of a culture solution in which cancer-related fibroblasts and killed cancer cells are co-cultured for prevention or treatment of cancer. WISP-1(Wnt-induced signaling protein-1)의 암 예방 또는 치료 용도.Use of WISP-1 (Wnt-induced signaling protein-1) for cancer prevention or treatment.
PCT/KR2022/014563 2021-09-29 2022-09-28 Composition for inhibiting cancer metastasis Ceased WO2023055076A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2021-0128716 2021-09-29
KR20210128716 2021-09-29
KR10-2022-0058184 2022-05-12
KR1020220058184A KR20230046190A (en) 2021-09-29 2022-05-12 Composition for inhibiting migration and invasion of cancer

Publications (1)

Publication Number Publication Date
WO2023055076A1 true WO2023055076A1 (en) 2023-04-06

Family

ID=85783213

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2022/014563 Ceased WO2023055076A1 (en) 2021-09-29 2022-09-28 Composition for inhibiting cancer metastasis

Country Status (1)

Country Link
WO (1) WO2023055076A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030068678A1 (en) * 1997-10-29 2003-04-10 Genentech, Inc. WISP polypeptides and nucleic acids encoding same
US20190204319A1 (en) * 2017-12-29 2019-07-04 Chang Gung Memorial Hospital, Keelung Method of Screening Breast Cancer by Using Serum WISP1 Level as a Biomarker

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030068678A1 (en) * 1997-10-29 2003-04-10 Genentech, Inc. WISP polypeptides and nucleic acids encoding same
US20190204319A1 (en) * 2017-12-29 2019-07-04 Chang Gung Memorial Hospital, Keelung Method of Screening Breast Cancer by Using Serum WISP1 Level as a Biomarker

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DU YAN, SHAO HONGWEI, MOLLER MECKER, PROKUPETS ROCHELLE, TSE YEE TING, LIU ZHAO-JUN: "Intracellular Notch1 Signaling in Cancer-Associated Fibroblasts Dictates the Plasticity and Stemness of Melanoma Stem/Initiating Cells", STEM CELLS, vol. 37, no. 7, 1 July 2019 (2019-07-01), pages 865 - 875, XP093053379, ISSN: 1066-5099, DOI: 10.1002/stem.3013 *
KIM, H. J. ET AL.: "Interaction between cancer-associated fibroblasts and apoptotic lung cancer cells inhibits migration and invasion of cancer cells via downregulation of TGF-β1 signaling", (THE KOREAN PHYSIOLOGICAL SOCIETY. THE 73RD ANNUAL MEETING OF THE KOREAN PHYSIOLOGICAL SOCIETY. 27-28 OCTOBER 2021, October 2021 (2021-10-01), pages 1 - 9 *
KIM, H. J. ET AL: "Apoptotic lung cancer cells suppress migration and invasion of cancer-associated fibroblasts via inhibition of TGF-β1 signaling", THE KOREAN PHYSIOLOGICAL SOCIETY. THE 73RD ANNUAL MEETING OF THE KOREAN PHYSIOLOGICAL SOCIETY. 27-28 OCTOBER 2021, October 2021 (2021-10-01) *
LEE, J.: "Reprogramming of cancer-associated fibroblasts[CAFs] by apoptotic cancer cells", INFLAMMATION-CANCER MICROENVIRONMENT RESEARCH CENTER, 27 October 2021 (2021-10-27), pages 1 - 58 *
YONG-BAE KIM, YOUNG-HO AHN, JI-HAE JUNG, YE-JI LEE, JIN-HWA LEE, JIHEE LEE KANG: "Programming of macrophages by UV-irradiated apoptotic cancer cells inhibits cancer progression and lung metastasis", CELLULAR & MOLECULAR IMMUNOLOGY, NATURE PUBLISHING GROUP UK, LONDON, 6 March 2019 (2019-03-06), London, XP055620099, ISSN: 1672-7681, DOI: 10.1038/s41423-019-0209-1 *

Similar Documents

Publication Publication Date Title
WO2017022962A1 (en) Composition for preventing or treating immune disease, comprising ripk inhibitor as active ingredient
Wang et al. Kynurenic acid ameliorates lipopolysaccharide-induced endometritis by regulating the GRP35/NF-κB signaling pathway
WO2021167389A1 (en) Pharmaceutical composition for preventing or treating cancer, containing mtor-signaling inhibitor as active ingredient
WO2019194586A1 (en) Novel target for anti-cancer and immune-enhancing
WO2017078405A9 (en) Pharmaceutical composition for treatment of lung cancer comprising glucocorticoid-based compound
JP7527593B2 (en) Neuronal Oxidative Synthase Inhibitors for Immunotherapy
WO2023096126A1 (en) Method for screening mdsc inhibitor
WO2023055076A1 (en) Composition for inhibiting cancer metastasis
KR101547297B1 (en) Composition for prevention or treating recurrent or metastatic cancer having immunoresistance
Ju et al. Inhibition of pro-inflammatory cytokines in tumour associated macrophages is a potential anti-cancer mechanism of carboxyamidotriazole
Arranz et al. The impact of stress on tumor growth: peripheral CRF mediates tumor-promoting effects of stress
González-Ortega et al. Uveal melanoma expresses NK-1 receptors and cyclosporin A induces apoptosis in human melanoma cell lines overexpressing the NK-1 receptor
WO2018026212A2 (en) Method for producing fibrosis disease model, and use of fibrosis disease model
WO2018147612A1 (en) Composition for inhibiting cancer metastasis and treating cancer
WO2016167605A2 (en) Method for inhibiting lung cancer of smokers and non-smokers using hypertension medicine
WO2016204545A1 (en) Peptide for promoting osteogenesis or inhibiting osteolysis, and use thereof
WO2017034234A1 (en) Composite formulation for treating cancer having hdac inhibitor resistance
WO2014157965A1 (en) Composition for treatment or metastasis suppression of cancers which includes p34 expression inhibitor or activity inhibitor as active ingredient
WO2025249993A1 (en) Composition for treating cancerous diseases and screening method thereof
WO2023182792A1 (en) Composition for inhibiting cancer growth
KR20230046190A (en) Composition for inhibiting migration and invasion of cancer
WO2021033973A1 (en) Composition for inhibiting myeloid-derived suppressor cell comprising mitf inhibitor as active ingredient
WO2022211473A1 (en) Fas-associated factor 1 (faf1)-loaded exosomes and use thereof as anti-cancer agent
KR20250172407A (en) A composition for treating cancer diseases and screening method thereof
WO2024262844A1 (en) Nanoparticles, preparation method therefor, and pharmaceutical composition for preventing or treating cancer, comprising same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22876848

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22876848

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 22876848

Country of ref document: EP

Kind code of ref document: A1

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 02/09/2024)

122 Ep: pct application non-entry in european phase

Ref document number: 22876848

Country of ref document: EP

Kind code of ref document: A1